Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 22
Filtrar
1.
Bioorg Med Chem Lett ; 27(12): 2721-2726, 2017 06 15.
Artículo en Inglés | MEDLINE | ID: mdl-28501511

RESUMEN

Interleukin-1 receptor associated kinase 4 (IRAK4) has been implicated in IL-1R and TLR based signaling. Therefore selective inhibition of the kinase activity of this protein represents an attractive target for the treatment of inflammatory diseases. Medicinal chemistry optimization of high throughput screening (HTS) hits with the help of structure based drug design led to the identification of orally-bioavailable quinazoline based IRAK4 inhibitors with excellent pharmacokinetic profile and kinase selectivity. These highly selective IRAK4 compounds show activity in vivo via oral dosing in a TLR7 driven model of inflammation.


Asunto(s)
Inflamación/tratamiento farmacológico , Quinasas Asociadas a Receptores de Interleucina-1/antagonistas & inhibidores , Inhibidores de Proteínas Quinasas/farmacología , Quinazolinas/farmacología , Administración Oral , Animales , Modelos Animales de Enfermedad , Relación Dosis-Respuesta a Droga , Ensayos Analíticos de Alto Rendimiento , Imidazoles/farmacología , Inflamación/enzimología , Quinasas Asociadas a Receptores de Interleucina-1/metabolismo , Interleucina-6/antagonistas & inhibidores , Interleucina-6/biosíntesis , Modelos Moleculares , Estructura Molecular , Inhibidores de Proteínas Quinasas/administración & dosificación , Inhibidores de Proteínas Quinasas/química , Quinazolinas/administración & dosificación , Quinazolinas/química , Ratas , Ratas Endogámicas Lew , Relación Estructura-Actividad
2.
Bioorg Med Chem Lett ; 25(22): 5384-8, 2015 Nov 15.
Artículo en Inglés | MEDLINE | ID: mdl-26403930

RESUMEN

IRAK4 plays a critical role in the IL-1R and TLR signalling, and selective inhibition of the kinase activity of the protein represents an attractive target for the treatment of inflammatory diseases. A series of permeable N-(1H-pyrazol-4-yl)carboxamides was developed by introducing lipophilic bicyclic cores in place of the polar pyrazolopyrimidine core of 5-amino-N-(1H-pyrazol-4-yl)pyrazolo[1,5-a]pyrimidine-3-carboxamides. Replacement of the pyrazolo[1,5-a]pyrimidine core with the pyrrolo[2,1-f][1,2,4]triazine, the pyrrolo[1,2-b]pyridazine, and thieno[2,3-b]pyrazine cores guided by cLogD led to the identification of highly permeable IRAK4 inhibitors with excellent potency and kinase selectivity.


Asunto(s)
Amidas/síntesis química , Amidas/farmacología , Ácidos Carboxílicos/química , Quinasas Asociadas a Receptores de Interleucina-1/antagonistas & inhibidores , Pirazoles/química , Amidas/química , Antiinflamatorios/síntesis química , Antiinflamatorios/química , Antiinflamatorios/farmacología , Ácidos Carboxílicos/síntesis química , Ácidos Carboxílicos/farmacología , Ciclización , Activación Enzimática/efectos de los fármacos , Humanos , Concentración 50 Inhibidora , Estructura Molecular , Pirazoles/síntesis química , Pirazoles/farmacología
3.
J Med Chem ; 2024 Jun 26.
Artículo en Inglés | MEDLINE | ID: mdl-38924388

RESUMEN

Oncogenic mutations in the RAS gene account for 30% of all human tumors; more than 60% of which present as KRAS mutations at the hotspot codon 12. After decades of intense pursuit, a covalent inhibition strategy has enabled selective targeting of this previously "undruggable" target. Herein, we disclose our journey toward the discovery of MK-1084, an orally bioavailable and low-dose KRASG12C covalent inhibitor currently in phase I clinical trials (NCT05067283). We leveraged structure-based drug design to identify a macrocyclic core structure, and hypothesis-driven optimization of biopharmaceutical properties to further improve metabolic stability and tolerability.

4.
Bioinformatics ; 27(20): 2775-81, 2011 Oct 15.
Artículo en Inglés | MEDLINE | ID: mdl-21846737

RESUMEN

MOTIVATION: Off-target activity commonly exists in RNA interference (RNAi) screens and often generates false positives. Existing analytic methods for addressing the off-target effects are demonstrably inadequate in RNAi confirmatory screens. RESULTS: Here, we present an analytic method assessing the collective activity of multiple short interfering RNAs (siRNAs) targeting a gene. Using this method, we can not only reduce the impact of off-target activities, but also evaluate the specific effect of an siRNA, thus providing information about potential off-target effects. Using in-house RNAi screens, we demonstrate that our method obtains more reasonable and sensible results than current methods such as the redundant siRNA activity (RSA) method, the RNAi gene enrichment ranking (RIGER) method, the frequency approach and the t-test. CONTACT: xiaohua_zhang@merck.com SUPPLEMENTARY INFORMATION: Supplementary data are available at Bioinformatics online.


Asunto(s)
Ensayos Analíticos de Alto Rendimiento , Interferencia de ARN , Enfermedad de Alzheimer/genética , Interpretación Estadística de Datos , Diabetes Mellitus/genética , Técnicas de Silenciamiento del Gen , Genómica/métodos , Herpesvirus Humano 3/genética , Humanos , ARN Interferente Pequeño
5.
Neoplasia ; 27: 100781, 2022 05.
Artículo en Inglés | MEDLINE | ID: mdl-35381456

RESUMEN

Cytotoxic CD8+ T cells are the primary effector cells mediating anti-tumor responses. In vivo monitoring of CD8+ T cells has broad implications for the development of novel cancer therapies. Here we describe the development of a genetically engineered mouse model (GEMM) in which CD8+ T cells are labeled with an optical reporter, enabling in vivo, longitudinal monitoring using bioluminescence imaging (BLI). Firefly luciferase (Luc2), human diphtheria toxin receptor (DTR), and enhanced green fluorescence protein (eGFP) cDNAs are engineered under the CD8α promoter to generate a transgenic mouse line. Luciferase mRNA and CD8α mRNA were generally correlated in various tissues from these mice. Sorted splenic CD8+ T cells, CD4+ T cells and CD3- non-T cells verified that the luciferase signal is specific to CD8+ T cells. In vivo imaging showed that luciferase signal was detected in various immune organs, such as lymph nodes, thymus, and spleen, and the detection was confirmed by ex vivo examination. Administration of diphtheria toxin markedly reduced luciferase signal systemically, confirming the function of the DTR. In the MC38 mouse syngeneic model, we observed significant increases in CD8+ T cells with mDX400 treatment, an anti PD-1 mouse monoclonal antibody that correlated with tumor growth inhibition. This novel reporter GEMM is a valuable drug discovery tool for profiling compounds and understanding mechanisms of action in immunotherapy of cancer.


Asunto(s)
Linfocitos T CD8-positivos , Luciferasas , Animales , Anticuerpos Monoclonales , Línea Celular Tumoral , Modelos Animales de Enfermedad , Genes Reporteros/genética , Luciferasas/genética , Luciferasas/metabolismo , Mediciones Luminiscentes , Ratones , Ratones Transgénicos , ARN Mensajero/metabolismo
6.
SLAS Discov ; 26(8): 1040-1054, 2021 09.
Artículo en Inglés | MEDLINE | ID: mdl-34130529

RESUMEN

One of the main reasons for the lack of drug efficacy in late-stage clinical trials is the lack of specific and selective target engagement. To increase the likelihood of success of new therapeutics, one approach is to conduct proximal target engagement testing during the early phases of preclinical drug discovery. To identify and optimize selective IRAK4 inhibitors, a kinase that has been implicated in multiple inflammatory and autoimmune diseases, we established an electrochemiluminescence (ECL)-based cellular endogenous IRAK1 activation assay as the most proximal functional evaluation of IRAK4 engagement to support structure-activity relationship (SAR) studies. Since IRAK1 activation is dependent on both the IRAK4 scaffolding function in Myddosome formation and IRAK4 kinase activity for signal transduction, this assay potentially captures inhibitors with different mechanisms of action. Data from this IRAK1 assay with compounds representing different structural classes showed statistically significant correlations when compared with results from both IRAK4 biochemical kinase activity and functional peripheral blood mononuclear cell (PBMC)-derived tumor necrosis factor α (TNFα) secretion assays, validating the biological relevancy of the IRAK1 target engagement as a biomarker of the IRAK4 activity. Plate uniformity and potency reproducibility evaluations demonstrated that this assay is amenable to high throughput. Using Bland-Altman assay agreement analysis, we demonstrated that incorporating such proximal pharmacological assessment of cellular target engagement to an in vitro screening funnel for SAR studies can prevent compound optimization toward off-target activity.


Asunto(s)
Descubrimiento de Drogas/métodos , Evaluación Preclínica de Medicamentos/métodos , Quinasas Asociadas a Receptores de Interleucina-1/antagonistas & inhibidores , Mediciones Luminiscentes/métodos , Inhibidores de Proteínas Quinasas/farmacología , Biomarcadores , Activación Enzimática/efectos de los fármacos , Humanos , Leucocitos Mononucleares/efectos de los fármacos , Leucocitos Mononucleares/metabolismo
7.
Dev Cell ; 5(1): 73-83, 2003 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-12852853

RESUMEN

beta cell dysfunction is an important component of type 2 diabetes, but the molecular basis for this defect is poorly understood. The transcriptional coactivator PGC-1alpha mRNA and protein levels are significantly elevated in islets from multiple animal models of diabetes; adenovirus-mediated expression of PGC-1alpha to levels similar to those present in diabetic rodents produces a marked inhibition of glucose-stimulated insulin secretion from islets in culture and in live mice. This inhibition coincides with changes in metabolic gene expression associated with impaired beta cell function, including the induction of glucose-6-phosphatase and suppression of GLUT2, glucokinase, and glycerol-3-phosphate dehydrogenase. These changes result in blunting of the glucose-induced rise in cellular ATP levels and membrane electrical activity responsible for Ca(2+) influx and insulin exocytosis. These results strongly suggest that PGC-1alpha plays a key functional role in the beta cell and is involved in the pathogenesis of the diabetic phenotype.


Asunto(s)
Metabolismo Energético , Insulina/metabolismo , Islotes Pancreáticos/metabolismo , Factores de Transcripción/farmacología , Células 3T3 , Potenciales de Acción/efectos de los fármacos , Adenosina Trifosfato/análisis , Animales , Células Cultivadas , Diabetes Mellitus Experimental/genética , Diabetes Mellitus Experimental/metabolismo , Glucoquinasa/metabolismo , Glucosa-6-Fosfatasa/metabolismo , Secreción de Insulina , Islotes Pancreáticos/efectos de los fármacos , Trasplante de Islotes Pancreáticos , Masculino , Ratones , Ratas , Ratas Mutantes , Ratas Zucker , Transfección
8.
Int J Biochem Cell Biol ; 40(12): 2702-6, 2008.
Artículo en Inglés | MEDLINE | ID: mdl-18678273

RESUMEN

Type 2 diabetes develops from insulin resistance and has become a worldwide epidemic. The c-Jun N-terminal kinases have been considered as signaling molecules linking inflammation and insulin resistance. Genetic disruption of c-Jun N-terminal kinase-1 gene prevents the development of insulin resistance in obese and diabetic mice. Inhibition of c-Jun N-terminal kinases by a small cell-permeable peptide improves insulin sensitivity in mice. Hepatic inhibition of c-Jun N-terminal kinases using a dominant-negative protein or knockdown of c-Jun N-terminal kinase-1 gene by RNA interference reduces blood glucose and insulin levels and enhances hepatic insulin signaling in mice. Recent evidence demonstrates that the hepatic c-Jun N-terminal kinase pathway plays an important role in lipid and lipoprotein homeostasis in mice. This review discusses recent advances in our understanding of the role of c-Jun N-terminal kinase pathway in metabolic control and its potential as a target for the treatment of type 2 diabetes.


Asunto(s)
Diabetes Mellitus Tipo 2/metabolismo , Hipoglucemiantes/metabolismo , Resistencia a la Insulina/fisiología , Insulina/metabolismo , Proteínas Quinasas JNK Activadas por Mitógenos/metabolismo , Animales , Proteínas Quinasas JNK Activadas por Mitógenos/fisiología , Ratones , Modelos Biológicos
9.
Mol Cell Endocrinol ; 473: 114-123, 2018 09 15.
Artículo en Inglés | MEDLINE | ID: mdl-29407196

RESUMEN

Min6 cells, a mouse ß cell line derived from transgenic mouse expressing the large T-antigen of SV40 in pancreatic beta cells, are commonly utilized as an in vitro cellular model for investigating targets involved in insulin secretion. Epac2, an exchange protein that can be directly activated by cyclic AMP (cAMP), is critical for pharmacologic stimuli-induced insulin secretion and has been hypothesized to be a direct target of sulfonylurea. Previous loss of function studies only specifically knocked out EPAC2 isoform A, leaving the other two isoforms intact. In this study, we investigated the function of EPAC2 in Min6 cells by generating EPAC2 knock-out sublines using CRISPR-Cas9 technology, by removing all three isoforms of EPAC2. Our results indicate that Min6 cells can be successfully cloned from a single cell after electroporation with plasmids expressing EPAC2 specific guide RNA, Cas9 and GFP, followed by sorting for GFP expressing single cells. Two clones were found to have a single nucleotide deletion in targeted site of EPAC2 gene by sequencing, therefore creating a frame shift in exon 13. The EPAC2 null clones have an unexpectedly increased secretion of insulin at basal level and an elevated total intracellular insulin content. However, EPAC2 deficiency impaires glucose and sulfonylurea induced insulin secretion without affecting sulfonylurea binding to cells. Potassium chloride induced insulin secretion remains intact. Interestingly, cAMP levels remained unchanged in EPAC2 null cells during these processes. To understand the global function of EPAC2, RNA Seq study was performed, which reveals that EPAC2 deficiency affects expression of multiple previously unrecognized genes, suggesting that EPAC2 can function through multiple pathways in addition to being a cAMP sensor.


Asunto(s)
Proteína 9 Asociada a CRISPR/metabolismo , Sistemas CRISPR-Cas/genética , Técnicas Genéticas , Factores de Intercambio de Guanina Nucleótido/genética , Animales , Línea Celular , Células Clonales , Regulación hacia Abajo/genética , Redes Reguladoras de Genes , Factores de Intercambio de Guanina Nucleótido/metabolismo , Insulina/metabolismo , Secreción de Insulina , Ratones Transgénicos , Plásmidos/metabolismo , Reproducibilidad de los Resultados , Transducción de Señal , Regulación hacia Arriba/genética
10.
JCI Insight ; 3(1)2018 01 11.
Artículo en Inglés | MEDLINE | ID: mdl-29321379

RESUMEN

Hypoglycemia is commonly associated with insulin therapy, limiting both its safety and efficacy. The concept of modifying insulin to render its glucose-responsive release from an injection depot (of an insulin complexed exogenously with a recombinant lectin) was proposed approximately 4 decades ago but has been challenging to achieve. Data presented here demonstrate that mannosylated insulin analogs can undergo an additional route of clearance as result of their interaction with endogenous mannose receptor (MR), and this can occur in a glucose-dependent fashion, with increased binding to MR at low glucose. Yet, these analogs retain capacity for binding to the insulin receptor (IR). When the blood glucose level is elevated, as in individuals with diabetes mellitus, MR binding diminishes due to glucose competition, leading to reduced MR-mediated clearance and increased partitioning for IR binding and consequent glucose lowering. These studies demonstrate that a glucose-dependent locus of insulin clearance and, hence, insulin action can be achieved by targeting MR and IR concurrently.


Asunto(s)
Glucosa/metabolismo , Hipoglucemia/tratamiento farmacológico , Insulina/farmacología , Animales , Antígenos CD , Glucemia , Línea Celular , Diabetes Mellitus Tipo 2 , Modelos Animales de Enfermedad , Hipoglucemiantes/farmacología , Lectinas Tipo C/efectos de los fármacos , Hígado/patología , Macrófagos , Masculino , Receptor de Manosa , Lectinas de Unión a Manosa/efectos de los fármacos , Ratones , Ratones Endogámicos C57BL , Ratas , Receptor de Insulina/efectos de los fármacos , Receptores de Superficie Celular/efectos de los fármacos
11.
J Med Chem ; 50(13): 3086-100, 2007 Jun 28.
Artículo en Inglés | MEDLINE | ID: mdl-17530838

RESUMEN

Stearoyl-CoA desaturase 1 (SCD1) catalyzes the committed step in the biosynthesis of monounsaturated fatty acids from saturated, long-chain fatty acids. Studies with SCD1 knockout mice have established that these animals are lean and protected from leptin deficiency-induced and diet-induced obesity, with greater whole body insulin sensitivity than wild-type animals. In this work, we have discovered a series of potent, selective, orally bioavailable SCD1 inhibitors based on a known pyridazine carboxamide template. The representative lead inhibitor 28c also demonstrates excellent cellular activity in blocking the conversion of saturated long-chain fatty acid-CoAs (LCFA-CoAs) to monounsaturated LCFA-CoAs in HepG2 cells.


Asunto(s)
Oxadiazoles/síntesis química , Piridazinas/síntesis química , Estearoil-CoA Desaturasa/antagonistas & inhibidores , Acilcoenzima A/metabolismo , Animales , Disponibilidad Biológica , Línea Celular Tumoral , Humanos , Técnicas In Vitro , Ratones , Microsomas Hepáticos/metabolismo , Oxadiazoles/farmacocinética , Oxadiazoles/farmacología , Piridazinas/farmacocinética , Piridazinas/farmacología , Ratas , Ratas Sprague-Dawley , Relación Estructura-Actividad
12.
ACS Med Chem Lett ; 6(6): 683-8, 2015 Jun 11.
Artículo en Inglés | MEDLINE | ID: mdl-26101574

RESUMEN

Interleukin-1 receptor associated kinase 4 (IRAK4) is an essential signal transducer downstream of the IL-1R and TLR superfamily, and selective inhibition of the kinase activity of the protein represents an attractive target for the treatment of inflammatory diseases. A series of 5-amino-N-(1H-pyrazol-4-yl)pyrazolo[1,5-a]pyrimidine-3-carboxamides was developed via sequential modifications to the 5-position of the pyrazolopyrimidine ring and the 3-position of the pyrazole ring. Replacement of substituents responsible for poor permeability and improvement of physical properties guided by cLogD led to the identification of IRAK4 inhibitors with excellent potency, kinase selectivity, and pharmacokinetic properties suitable for oral dosing.

13.
PLoS One ; 7(5): e36384, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-22590537

RESUMEN

BACKGROUND: Hepatic insulin resistance impairs insulin's ability to suppress hepatic glucose production (HGP) and contributes to the development of type 2 diabetes (T2D). Although the interests to discover novel genes that modulate insulin sensitivity and HGP are high, it remains challenging to have a human cell based system to identify novel genes. METHODOLOGY/PRINCIPAL FINDINGS: To identify genes that modulate hepatic insulin signaling and HGP, we generated a human cell line stably expressing beta-lactamase under the control of the human glucose-6-phosphatase (G6PC) promoter (AH-G6PC cells). Both beta-lactamase activity and endogenous G6PC mRNA were increased in AH-G6PC cells by a combination of dexamethasone and pCPT-cAMP, and reduced by insulin. A 4-gene High-Throughput-Genomics assay was developed to concomitantly measure G6PC and pyruvate-dehydrogenase-kinase-4 (PDK4) mRNA levels. Using this assay, we screened an siRNA library containing pooled siRNA targeting 6650 druggable genes and identified 614 hits that lowered G6PC expression without increasing PDK4 mRNA levels. Pathway analysis indicated that siRNA-mediated knockdown (KD) of genes known to positively or negatively affect insulin signaling increased or decreased G6PC mRNA expression, respectively, thus validating our screening platform. A subset of 270 primary screen hits was selected and 149 hits were confirmed by target gene KD by pooled siRNA and 7 single siRNA for each gene to reduce G6PC expression in 4-gene HTG assay. Subsequently, pooled siRNA KD of 113 genes decreased PEPCK and/or PGC1alpha mRNA expression thereby demonstrating their role in regulating key gluconeogenic genes in addition to G6PC. Last, KD of 61 of the above 113 genes potentiated insulin-stimulated Akt phosphorylation, suggesting that they suppress gluconeogenic gene by enhancing insulin signaling. CONCLUSIONS/SIGNIFICANCE: These results support the proposition that the proteins encoded by the genes identified in our cell-based druggable genome siRNA screen hold the potential to serve as novel pharmacological targets for the treatment of T2D.


Asunto(s)
Diabetes Mellitus Tipo 2 , Genoma Humano , Gluconeogénesis/genética , Resistencia a la Insulina/genética , Hígado/metabolismo , ARN Interferente Pequeño , Línea Celular Tumoral , Diabetes Mellitus Tipo 2/genética , Diabetes Mellitus Tipo 2/metabolismo , Estudio de Asociación del Genoma Completo , Genómica/métodos , Humanos
14.
J Biomol Screen ; 15(9): 1123-31, 2010 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-20852024

RESUMEN

In genome-scale RNA interference (RNAi) screens, it is critical to control false positives and false negatives statistically. Traditional statistical methods for controlling false discovery and false nondiscovery rates are inappropriate for hit selection in RNAi screens because the major goal in RNAi screens is to control both the proportion of short interfering RNAs (siRNAs) with a small effect among selected hits and the proportion of siRNAs with a large effect among declared nonhits. An effective method based on strictly standardized mean difference (SSMD) has been proposed for statistically controlling false discovery rate (FDR) and false nondiscovery rate (FNDR) appropriate for RNAi screens. In this article, the authors explore the utility of the SSMD-based method for hit selection in RNAi screens. As demonstrated in 2 genome-scale RNAi screens, the SSMD-based method addresses the unmet need of controlling for the proportion of siRNAs with a small effect among selected hits, as well as controlling for the proportion of siRNAs with a large effect among declared nonhits. Furthermore, the SSMD-based method results in reasonably low FDR and FNDR for selecting inhibition or activation hits. This method works effectively and should have a broad utility for hit selection in RNAi screens with replicates.


Asunto(s)
Genoma/genética , Genómica/métodos , Interferencia de ARN , Línea Celular Tumoral , Diabetes Mellitus/genética , Reacciones Falso Negativas , Reacciones Falso Positivas , Humanos , Enfermedades del Sistema Nervioso/genética , ARN Interferente Pequeño/metabolismo , Reproducibilidad de los Resultados
15.
J Biol Chem ; 282(31): 22765-74, 2007 Aug 03.
Artículo en Inglés | MEDLINE | ID: mdl-17550900

RESUMEN

The c-Jun N-terminal kinases (JNKs) have been implicated in the development of insulin resistance, diabetes, and obesity. Genetic disruption of JNK1, but not JNK2, improves insulin sensitivity in diet-induced obese (DIO) mice. We applied RNA interference to investigate the specific role of hepatic JNK1 in contributing to insulin resistance in DIO mice. Adenovirus-mediated delivery of JNK1 short-hairpin RNA (Ad-shJNK1) resulted in almost complete knockdown of hepatic JNK1 protein without affecting JNK1 protein in other tissues. Liver-specific knockdown of JNK1 resulted in significant reductions in circulating insulin and glucose levels, by 57 and 16%, respectively. At the molecular level, JNK1 knockdown mice had sustained and significant increase of hepatic Akt phosphorylation. Furthermore, knockdown of JNK1 enhanced insulin signaling in vitro. Unexpectedly, plasma triglyceride levels were robustly elevated upon hepatic JNK1 knockdown. Concomitantly, expression of proliferator-activated receptor gamma coactivator 1 beta, glucokinase, and microsomal triacylglycerol transfer protein was increased. Further gene expression analysis demonstrated that knockdown of JNK1 up-regulates the hepatic expression of clusters of genes in glycolysis and several genes in triglyceride synthesis pathways. Our results demonstrate that liver-specific knockdown of JNK1 lowers circulating glucose and insulin levels but increases triglyceride levels in DIO mice.


Asunto(s)
Regulación Enzimológica de la Expresión Génica , Glucosa/metabolismo , Insulina/metabolismo , Hígado/metabolismo , Proteína Quinasa 8 Activada por Mitógenos/metabolismo , Transactivadores/biosíntesis , Triglicéridos/sangre , Adenoviridae/genética , Adenoviridae/metabolismo , Alimentación Animal , Animales , Cartilla de ADN/química , Ratones , Ratones Obesos , PPAR gamma/metabolismo , Coactivador 1-alfa del Receptor Activado por Proliferadores de Peroxisomas gamma , Fosforilación , Factores de Transcripción , Triglicéridos/metabolismo
16.
J RNAi Gene Silencing ; 3(1): 225-36, 2006 Nov 29.
Artículo en Inglés | MEDLINE | ID: mdl-19771218

RESUMEN

RNA interference (RNAi) is an exciting new tool to effect acute in vivo knockdown of genes for pharmacological target validation. Testing the application of this technology to metabolic disease targets, three RNAi delivery methods were compared in two frequently utilized preclinical models of obesity and diabetes, the diet-induced obese (DIO) and B6.V-Lep/J (ob/ob) mouse. Intraperitoneal (i.p.) and high pressure hydrodynamic intravenous (i.v.) administration of naked siRNA, and low pressure i.v. administration of shRNA-expressing adenovirus were assessed for both safety and gene knockdown efficacy using constructs targeting cJun N-terminal kinase 1 (JNK1). Hydrodynamic delivery of siRNA lowered liver JNK1 protein levels 40% in DIO mice, but was accompanied by iatrogenic liver damage. The ob/ob model proved even more intolerant of this technique, with hydrodynamic delivery resulting in severe liver damage and death of most animals. While well-tolerated, i.p. injections of siRNA in DIO mice did not result in any knockdown or phenotypic changes in the mice. On the other hand, i.v. injected adenovirus expressing shRNA potently reduced expression of JNK1 in vivo by 95% without liver toxicity. In conclusion, i.p. and hydrodynamic injections of siRNA were ineffective and/or inappropriate for in vivo gene targeting in DIO and ob/ob mice, while adenovirus-mediated delivery of shRNA provided a relatively benign and effective method for exploring liver target silencing.

17.
Cell ; 120(2): 261-73, 2005 Jan 28.
Artículo en Inglés | MEDLINE | ID: mdl-15680331

RESUMEN

The PGC-1 family of coactivators stimulates the activity of certain transcription factors and nuclear receptors. Transcription factors in the sterol responsive element binding protein (SREBP) family are key regulators of the lipogenic genes in the liver. We show here that high-fat feeding, which induces hyperlipidemia and atherogenesis, stimulates the expression of both PGC-1beta and SREBP1c and 1a in liver. PGC-1beta coactivates the SREBP transcription factor family and stimulates lipogenic gene expression. Further, PGC-1beta is required for SREBP-mediated lipogenic gene expression. However, unlike SREBP itself, PGC-1beta reduces fat accumulation in the liver while greatly increasing circulating triglycerides and cholesterol in VLDL particles. The stimulation of lipoprotein transport upon PGC-1beta expression is likely due to the simultaneous coactivation of the liver X receptor, LXRalpha, a nuclear hormone receptor with known roles in hepatic lipid transport. These data suggest a mechanism through which dietary saturated fats can stimulate hyperlipidemia and atherogenesis.


Asunto(s)
Proteínas Potenciadoras de Unión a CCAAT/biosíntesis , Proteínas de Unión al ADN/biosíntesis , Grasas de la Dieta/administración & dosificación , Regulación de la Expresión Génica/fisiología , Hiperlipidemias/metabolismo , Transactivadores/biosíntesis , Factores de Transcripción/biosíntesis , Animales , Colesterol/metabolismo , Grasas de la Dieta/metabolismo , Perfilación de la Expresión Génica , Hígado/metabolismo , Receptores X del Hígado , Masculino , Ratones , Receptores Nucleares Huérfanos , Coactivador 1-alfa del Receptor Activado por Proliferadores de Peroxisomas gamma , Receptores Citoplasmáticos y Nucleares/biosíntesis , Proteína 1 de Unión a los Elementos Reguladores de Esteroles
18.
J Biol Chem ; 278(26): 23418-25, 2003 Jun 27.
Artículo en Inglés | MEDLINE | ID: mdl-12697773

RESUMEN

Glycogen-targeting subunits of protein phosphatase-1 (PP-1) are scaffolding proteins that facilitate the regulation of key enzymes of glycogen metabolism by PP-1. In the current study, we have tested the effects of hepatic expression of GMDeltaC, a truncated version of the muscle-targeting subunit isoform, in rats rendered insulin-deficient via injection of a single moderate dose of streptozotocin (STZ). Three key findings emerged. First, GMDeltaC expression in liver was sufficient to fully normalize blood glucose levels (from 335 +/- 31 mg/dl prior to viral injection to 109 +/- 28 mg/dl 6 days after injection) and liver glycogen content in STZ-injected rats. Second, this normalization occurred despite very low levels of liver glucokinase expression in the insulin-deficient STZ-injected rats. Finally, the hyperphagia induced by STZ injection was completely reversed by GMDeltaC expression in liver. In contrast to these findings with GMDeltaC, overexpression of another targeting subunit, GL, in STZ-injected rats caused a large increase in liver glycogen stores but only a transient decrease in food intake and blood glucose levels. The surprising demonstration of a glucose-lowering effect of GMDeltaC in the background of depressed hepatic glucokinase expression suggests that controlled stimulation of liver glycogen storage may be an effective mechanism for improving glucose homeostasis, even when normal pathways of glucose disposal are impaired.


Asunto(s)
Diabetes Mellitus Experimental/terapia , Hiperglucemia/terapia , Hiperfagia/terapia , Hígado/metabolismo , Fosfoproteínas Fosfatasas/farmacología , Adenoviridae/genética , Animales , Glucemia/análisis , Glucemia/efectos de los fármacos , Terapia Genética/métodos , Glucoquinasa/sangre , Glucógeno/metabolismo , Hiperglucemia/inducido químicamente , Hiperfagia/inducido químicamente , Hígado/efectos de los fármacos , Masculino , Fosfoproteínas Fosfatasas/administración & dosificación , Fosfoproteínas Fosfatasas/genética , Proteína Fosfatasa 1 , Subunidades de Proteína/administración & dosificación , Subunidades de Proteína/genética , Subunidades de Proteína/farmacología , Ratas , Ratas Wistar , Estreptozocina
19.
J Biol Chem ; 277(2): 1524-30, 2002 Jan 11.
Artículo en Inglés | MEDLINE | ID: mdl-11707447

RESUMEN

Glycogen-targeting subunits of protein phosphatase-1 facilitate interaction of the phosphatase with enzymes of glycogen metabolism. Expression of one family member, PTG, in the liver of normal rats improves glucose tolerance without affecting other plasma variables but leaves animals unable to reduce hepatic glycogen stores in response to fasting. In the current study, we have tested whether expression of other targeting subunit isoforms, such as the liver isoform G(L), the muscle isoform G(M)/R(Gl), or a truncated version of G(M)/R(Gl) termed G(M)DeltaC in liver ameliorates glucose intolerance in rats fed on a high fat diet (HF). HF animals overexpressing G(M)DeltaC, but not G(L) or G(M)/R(Gl), exhibited a decline in blood glucose of 35-44 mg/dl relative to control HF animals during an oral glucose tolerance test (OGTT) such that levels were indistinguishable from those of normal rats fed on standard chow at all but one time point. Hepatic glycogen levels were 2.1-2.4-fold greater in G(L)- and G(M)DeltaC-overexpressing HF rats compared with control HF animals following OGTT. In a second set of studies on fed and 20-h fasted HF animals, G(M)DeltaC-overexpressing rats lowered their liver glycogen levels by 57% (from 402 +/- 54 to 173 +/- 27 microg of glycogen/mg of protein) in the fasted versus fed states compared with only 44% in G(L)-overexpressing animals (from 740 +/- 35 to 413 +/- 141 microg of glycogen/mg of protein). Since the OGTT studies were performed on 20-h fasted rats, this meant that G(M)DeltaC-overexpressing rats synthesized much more glycogen than G(L)-overexpressing HF rats during the OGTT (419 versus 117 microg of glycogen/mg of protein, respectively), helping to explain why G(M)DeltaC preferentially enhanced glucose clearance. We conclude that G(M)DeltaC has a unique combination of glycogenic potency and responsiveness to glycogenolytic signals that allows it to be used to lower blood glucose levels in diabetes.


Asunto(s)
Grasas de la Dieta/administración & dosificación , Intolerancia a la Glucosa/metabolismo , Glucosa/metabolismo , Glucógeno/metabolismo , Hígado/enzimología , Fosfoproteínas Fosfatasas/metabolismo , Adenoviridae/genética , Adenoviridae/metabolismo , Animales , Grasas de la Dieta/efectos adversos , Ayuno , Intolerancia a la Glucosa/enzimología , Prueba de Tolerancia a la Glucosa , Humanos , Resistencia a la Insulina/fisiología , Isoenzimas/metabolismo , Masculino , Músculo Esquelético/fisiología , Fosfoproteínas Fosfatasas/genética , Proteína Fosfatasa 1 , Subunidades de Proteína , Ratas , Ratas Wistar , Transgenes/genética
20.
J Biol Chem ; 278(33): 30843-8, 2003 Aug 15.
Artículo en Inglés | MEDLINE | ID: mdl-12807885

RESUMEN

Peroxisome proliferator-activated receptor gamma coactivator-1alpha (PGC-1alpha) is a transcriptional coactivator that regulates multiple aspects of cellular energy metabolism, including mitochondrial biogenesis, hepatic gluconeogenesis, and beta-oxidation of fatty acids. PGC-1alpha mRNA levels are increased in both type-1 and type-2 diabetes and may contribute to elevated hepatic glucose production in diabetic states. We have recently described PGC-1beta, a novel transcriptional coactivator that is a homolog of PGC-1alpha. Although PGC-1beta shares significant sequence similarity and tissue distribution with PGC-1alpha, the biological activities of PGC-1beta in the regulation of cellular metabolism is unknown. In this study, we used an adenoviral-mediated expression system to study the function of PGC-1beta both in cultured hepatocytes and in the liver of rats. PGC-1beta, like PGC-1alpha, potently induces the expression of an array of mitochondrial genes involved in oxidative metabolism. However, in contrast to PGC-1alpha, PGC-1beta poorly activates the expression of gluconeogenic genes in hepatocytes or liver in vivo, illustrating that these two coactivators play distinct roles in hepatic glucose metabolism. The reduced ability of PGC-1beta to induce gluconeogenic genes is due, at least in part, to its inability to physically associate with and coactivate hepatic nuclear receptor 4alpha (HNF4alpha) and forkhead transcription factor O1 (FOXO1), two critical transcription factors that mediate the activation of gluconeogenic gene expression by PGC-1alpha. These data illustrate that PGC-1beta and PGC-1alpha have distinct arrays of activities in hepatic energy metabolism.


Asunto(s)
Metabolismo Energético/fisiología , Glucosa/metabolismo , Hígado/metabolismo , Factores de Transcripción/metabolismo , Animales , Carcinoma Hepatocelular , Línea Celular Transformada , Diabetes Mellitus/metabolismo , Expresión Génica/fisiología , Gluconeogénesis/fisiología , Hígado/citología , Hígado/embriología , Neoplasias Hepáticas , Ratones , ARN Mensajero/análisis , Ratas , Factores de Transcripción/genética , Activación Transcripcional/fisiología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA