Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Resultados 1 - 20 de 34
Filtrar
1.
Osteoporos Int ; 24(10): 2659-70, 2013 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-23644877

RESUMEN

SUMMARY: The present study shows no adverse effects of the anti-diabetic drug metformin on bone mass and fracture healing in rodents but demonstrates that metformin is not osteogenic in vivo, as previously proposed. INTRODUCTION: In view of the increased incidence of fractures in patients with type 2 diabetes mellitus (T2DM), we investigated the effects of metformin, a widely used T2DM therapy, on bone mass and fracture healing in vivo using two different rodent models and modes of metformin administration. METHODS: We first subjected 12-week-old female C57BL/6 mice to ovariectomy (OVX). Four weeks after OVX, mice received either saline or metformin administered by gavage (100 mg/kg/daily). After 4 weeks of treatment, bone micro-architecture and cellular activity were determined in tibia by micro-CT and bone histomorphometry. In another experiment, female Wistar rats aged 3 months were given only water or metformin for 8 weeks via the drinking water (2 mg/ml). After 4 weeks of treatment, a mid-diaphyseal osteotomy was performed in the left femur. Rats were sacrificed 4 weeks after osteotomy and bone architecture analysed by micro-CT in the right tibia while fracture healing and callus volume were determined in the left femur by X-ray analysis and micro-CT, respectively. RESULTS: In both models, our results show no significant differences in cortical and trabecular bone architecture in metformin-treated rodents compared to saline. Metformin had no effect on bone resorption but reduced bone formation rate in trabecular bone. Mean X-ray scores assessed on control and metformin fractures showed no significant differences of healing between the groups. Fracture callus volume and mineral content after 4 weeks were similar in both groups. CONCLUSIONS: Our results indicate that metformin has no effect on bone mass in vivo or fracture healing in rodents.


Asunto(s)
Densidad Ósea/efectos de los fármacos , Curación de Fractura/efectos de los fármacos , Hipoglucemiantes/farmacología , Metformina/farmacología , Proteínas Quinasas Activadas por AMP/metabolismo , Animales , Densidad Ósea/fisiología , Remodelación Ósea/efectos de los fármacos , Callo Óseo/efectos de los fármacos , Callo Óseo/patología , Activación Enzimática/efectos de los fármacos , Femenino , Fracturas del Fémur/fisiopatología , Fémur/enzimología , Curación de Fractura/fisiología , Hipoglucemiantes/sangre , Metformina/sangre , Ratones , Ratones Endogámicos C57BL , Osteoporosis/fisiopatología , Ovariectomía , Ratas , Ratas Wistar , Tibia/diagnóstico por imagen , Tibia/efectos de los fármacos , Tibia/patología , Tibia/fisiopatología , Microtomografía por Rayos X/métodos
2.
Diabetologia ; 54(12): 3101-10, 2011 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-21947382

RESUMEN

AIM/HYPOTHESIS: The glucose-lowering drug metformin has been shown to activate hepatic AMP-activated protein kinase (AMPK), a master kinase regulating cellular energy homeostasis. However, the underlying mechanisms remain controversial and have never been investigated in primary human hepatocytes. METHODS: Hepatocytes isolated from rat, mouse and human livers were treated with various concentrations of metformin. Isoform-specific AMPKα abundance and activity, as well as intracellular adenine nucleotide levels and mitochondrial oxygen consumption rates were determined at different time points. RESULTS: Metformin dose- and time-dependently increased AMPK activity in rat and human hepatocytes, an effect associated with a significant rise in cellular AMP:ATP ratio. Surprisingly, we found that AMPKα2 activity was undetectable in human compared with rat hepatocytes, while AMPKα1 activities were comparable. Accordingly, metformin only increased AMPKα1 activity in human hepatocytes, although both AMPKα isoforms were activated in rat hepatocytes. Analysis of mRNA expression and protein levels confirmed that only AMPKα1 is present in human hepatocytes; it also showed that the distribution of ß and γ regulatory subunits differed between species. Finally, we demonstrated that the increase in AMP:ATP ratio in hepatocytes from liver-specific Ampkα1/2 (also known as Prkaa1/2) knockout mice and humans is due to a similar and specific inhibition of the mitochondrial respiratory-chain complex 1 by metformin. CONCLUSIONS/INTERPRETATION: Activation of hepatic AMPK by metformin results from a decrease in cellular energy status owing to metformin's AMPK-independent inhibition of the mitochondrial respiratory-chain complex 1. The unique profile of AMPK subunits found in human hepatocytes should be considered when developing new pharmacological agents to target the kinase.


Asunto(s)
Proteínas Quinasas Activadas por AMP/efectos de los fármacos , Hepatocitos/efectos de los fármacos , Hipoglucemiantes/farmacología , Metformina/farmacología , Proteínas Quinasas Activadas por AMP/análisis , Nucleótidos de Adenina/análisis , Animales , Células Cultivadas , Hepatocitos/enzimología , Humanos , Isoenzimas/metabolismo , Masculino , Ratones , Ratones Noqueados , Mitocondrias Hepáticas/efectos de los fármacos , Mitocondrias Hepáticas/metabolismo , Consumo de Oxígeno/efectos de los fármacos , Ratas
3.
Nat Med ; 2(1): 80-6, 1996 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-8564847

RESUMEN

Fas is an apoptosis-signalling cell surface antigen that has been shown to trigger cell death upon specific ligand or antibody binding. Treatment of mice with an anti-Fas antibody causes fulminant hepatic failure due to massive apoptosis. To test a putative protective effect of the anti-apoptotic Bcl-2 protein, transgenic mice were generated to express the human bcl-2 gene product in hepatocytes. Early onset of massive hepatic apoptosis leading to death was observed in all nontransgenic mice treated with an anti-Fas antibody. By contrast, hepatic apoptosis was delayed and dramatically reduced in transgenic animals, yielding a 93% survival rate. These results demonstrate that Bcl-2 is able to protect from in vivo Fas-mediated cytotoxicity, and could be of significance for preventing fulminant hepatic failure due to viral hepatitis in humans.


Asunto(s)
Anticuerpos/toxicidad , Apoptosis/fisiología , Encefalopatía Hepática/prevención & control , Hígado/patología , Proteínas Proto-Oncogénicas/biosíntesis , Proto-Oncogenes , Receptor fas/fisiología , Animales , Northern Blotting , Western Blotting , Proteínas de Unión al GTP/biosíntesis , Encefalopatía Hepática/patología , Humanos , Hígado/metabolismo , Ratones , Ratones Endogámicos CBA , Ratones Transgénicos , Proteínas Proto-Oncogénicas/genética , Proteínas Proto-Oncogénicas c-bcl-2 , Receptor fas/inmunología
4.
Diabetologia ; 53(5): 924-36, 2010 May.
Artículo en Inglés | MEDLINE | ID: mdl-20221584

RESUMEN

AIMS/HYPOTHESIS: AMP-activated protein kinase (AMPK) is an evolutionarily conserved enzyme and a target of glucose-lowering agents, including metformin. However, the precise role or roles of the enzyme in controlling insulin secretion remain uncertain. METHODS: The catalytic alpha1 and alpha2 subunits of AMPK were ablated selectively in mouse pancreatic beta cells and hypothalamic neurons by breeding Ampkalpha1 [also known as Prkaa1]-knockout mice, bearing floxed Ampkalpha2 [also known as Prkaa2] alleles (Ampkalpha1 ( -/- ),alpha2( fl/fl ),), with mice expressing Cre recombinase under the rat insulin promoter (RIP2). RIP2 was used to express constitutively activated AMPK selectively in beta cells in transgenic mice. Food intake, body weight and urinary catecholamines were measured using metabolic cages. Glucose and insulin tolerance were determined after intraperitoneal injection. Beta cell mass and morphology were analysed by optical projection tomography and confocal immunofluorescence microscopy, respectively. Granule docking, insulin secretion, membrane potential and intracellular free Ca(2+) were measured with standard techniques. RESULTS: Trigenic Ampkalpha1 ( -/- ),alpha2( fl/fl ) expressing Cre recombinase and lacking both AMPKalpha subunits in the beta cell, displayed normal body weight and increased insulin sensitivity, but were profoundly insulin-deficient. Secreted catecholamine levels were unchanged. Total beta cell mass was unaltered, while mean islet and beta cell volume were reduced. AMPK-deficient beta cells displayed normal glucose-induced changes in membrane potential and intracellular free Ca(2+), while granule docking and insulin secretion were enhanced. Conversely, betaAMPK transgenic mice were glucose-intolerant and displayed defective insulin secretion. CONCLUSIONS/INTERPRETATION: Inhibition of AMPK activity within the beta cell is necessary, but not sufficient for stimulation of insulin secretion by glucose to occur. AMPK activation in extrapancreatic RIP2.Cre-expressing cells might also influence insulin secretion in vivo.


Asunto(s)
Proteínas Quinasas Activadas por AMP/metabolismo , Hipotálamo/metabolismo , Células Secretoras de Insulina/metabolismo , Insulina/metabolismo , Neuronas/metabolismo , Proteínas Quinasas Activadas por AMP/genética , Análisis de Varianza , Animales , Glucemia/metabolismo , Peso Corporal/genética , Grasas de la Dieta , Ingestión de Alimentos/genética , Electrofisiología , Técnica del Anticuerpo Fluorescente , Prueba de Tolerancia a la Glucosa , Hiperglucemia/genética , Hiperglucemia/metabolismo , Insulina/genética , Secreción de Insulina , Ratones , Ratones Noqueados , Regiones Promotoras Genéticas/genética , Ratas
5.
J Exp Med ; 194(11): 1683-9, 2001 Dec 03.
Artículo en Inglés | MEDLINE | ID: mdl-11733582

RESUMEN

The terminal components of the complement system contribute to host defense by forming the multiprotein membrane attack complex (MAC) which is responsible for cell lysis and several noncytotoxic effects. Most of the complement proteins are synthesized in the liver, but the mechanisms controlling their tissue-specific expression have not been elucidated. In this study we show that mice lacking the hepatic transcription factor hepatocyte nuclear factor 1alpha (HNF1alpha) fail to transcribe C5 and C8A complement genes. In addition, mRNAs encoding for several other terminal complement components or subunits are expressed at lower levels, including C8beta, C8gamma, and C9. We next used a reconstitution assay involving human sera with selective complement deficiencies to assess mouse complement activity. Sera from HNF1alpha-deficient mice showed negligible hemolytic activity of both C5 and C8alpha-gamma subunits. The activity of C8beta was severely affected despite only a 50% reduction in C8beta mRNA levels in the liver. This is reminiscent of C8alpha-gamma-deficient patients who accumulate extremely low levels of the C8beta subunit. Our results demonstrate that HNF1alpha plays a key role in the expression of C5 and C8A genes, two terminal complement component genes that are essential for the assembly of MAC as a result of complement activation.


Asunto(s)
Complemento C5/genética , Complemento C8/genética , Proteínas de Unión al ADN , Regulación de la Expresión Génica , Proteínas Nucleares/fisiología , Factores de Transcripción/fisiología , Animales , Secuencia de Bases , Cromatina , Complemento C5/inmunología , Complemento C8/inmunología , ADN Complementario , Pruebas Genéticas , Factor Nuclear 1 del Hepatocito , Factor Nuclear 1-alfa del Hepatocito , Factor Nuclear 1-beta del Hepatocito , Humanos , Hígado/inmunología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Datos de Secuencia Molecular , Proteínas Nucleares/genética , Factores de Transcripción/genética , Transcripción Genética
6.
Oncogene ; 37(7): 963-970, 2018 02 15.
Artículo en Inglés | MEDLINE | ID: mdl-29059169

RESUMEN

The anti-diabetic biguanide metformin may exert health-promoting effects via metabolic regulation of the epigenome. Here we show that metformin promotes global DNA methylation in non-cancerous, cancer-prone and metastatic cancer cells by decreasing S-adenosylhomocysteine (SAH), a strong feedback inhibitor of S-adenosylmethionine (SAM)-dependent DNA methyltransferases, while promoting the accumulation of SAM, the universal methyl donor for cellular methylation. Using metformin and a mitochondria/complex I (mCI)-targeted analog of metformin (norMitoMet) in experimental pairs of wild-type and AMP-activated protein kinase (AMPK)-, serine hydroxymethyltransferase 2 (SHMT2)- and mCI-null cells, we provide evidence that metformin increases the SAM:SAH ratio-related methylation capacity by targeting the coupling between serine mitochondrial one-carbon flux and CI activity. By increasing the contribution of one-carbon units to the SAM from folate stores while decreasing SAH in response to AMPK-sensed energetic crisis, metformin can operate as a metabolo-epigenetic regulator capable of reprogramming one of the key conduits linking cellular metabolism to the DNA methylation machinery.


Asunto(s)
Neoplasias de la Mama/tratamiento farmacológico , Carbono/metabolismo , Neoplasias del Colon/tratamiento farmacológico , Metilación de ADN/efectos de los fármacos , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Genoma Humano , Metformina/farmacología , Proteínas Quinasas Activadas por AMP/metabolismo , Animales , Biomarcadores de Tumor , Neoplasias de la Mama/enzimología , Neoplasias de la Mama/patología , Neoplasias del Colon/enzimología , Neoplasias del Colon/patología , Complejo I de Transporte de Electrón/metabolismo , Femenino , Estudios de Seguimiento , Humanos , Hipoglucemiantes/farmacología , Ratones , Mitocondrias/efectos de los fármacos , Mitocondrias/metabolismo , Mitocondrias/patología , S-Adenosilhomocisteína/metabolismo , S-Adenosilmetionina/metabolismo , Células Tumorales Cultivadas
7.
FEBS Lett ; 581(5): 1053-7, 2007 Mar 06.
Artículo en Inglés | MEDLINE | ID: mdl-17316628

RESUMEN

Diabetes Mellitus is found with increasing frequency in iron overload patients with hemochromatosis. In these conditions, the pancreas shows predominant iron overload in acini but also islet beta-cells. We assess glucose homeostasis status in iron-overloaded hepcidin-deficient mice. These mice presented with heavy pancreatic iron deposits but only in the acini. The beta-cell function was found unaffected with a normal production and secretion of insulin. The mutant mice were not diabetic, responded as the control group to glucose and insulin challenges, with no alteration of insulin signalling in the muscle and the liver. These results indicate that, beta-cells iron deposits-induced decreased insulin secretory capacity might be of primary importance to trigger diabetes in hemochromatosic patients.


Asunto(s)
Péptidos Catiónicos Antimicrobianos/deficiencia , Glucosa/metabolismo , Sobrecarga de Hierro/metabolismo , Animales , Péptidos Catiónicos Antimicrobianos/genética , Diabetes Mellitus/etiología , Hemocromatosis/complicaciones , Hemocromatosis/metabolismo , Hepcidinas , Homeostasis , Humanos , Insulina/biosíntesis , Insulina/metabolismo , Secreción de Insulina , Células Secretoras de Insulina/patología , Células Secretoras de Insulina/fisiología , Sobrecarga de Hierro/patología , Sobrecarga de Hierro/fisiopatología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Páncreas/metabolismo , Páncreas/patología , Transducción de Señal
8.
Mol Cell Biol ; 17(8): 4208-19, 1997 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-9234678

RESUMEN

Hepatocyte nuclear factor 4 (HNF4), a liver-enriched transcription factor of the nuclear receptor superfamily, is critical for development and liver-specific gene expression. Here, we demonstrate that its DNA-binding activity is modulated posttranslationally by phosphorylation in vivo, ex vivo, and in vitro. In vivo, HNF4 DNA-binding activity is reduced by fasting and by inducers of intracellular cyclic AMP (cAMP) accumulation. A consensus protein kinase A (PKA) phosphorylation site located within the A box of its DNA-binding domain has been identified, and its role in phosphorylation-dependent inhibition of HNF4 DNA-binding activity has been investigated. Mutants of HNF4 in which two potentially phosphorylatable serines have been replaced by either neutral or charged amino acids were able to bind DNA in vitro with affinity similar to that of the wild-type protein. However, phosphorylation by PKA strongly repressed the binding affinity of the wild-type factor but not that of HNF4 mutants. Accordingly, in transfection assays, expression vectors for the mutated HNF4 proteins activated transcription more efficiently than that for the wild-type protein-when cotransfected with the PKA catalytic subunit expression vector. Therefore, HNF4 is a direct target of PKA which might be involved in the transcriptional inhibition of liver genes by cAMP inducers.


Asunto(s)
Proteínas Quinasas Dependientes de AMP Cíclico/metabolismo , Proteínas de Unión al ADN/metabolismo , Fosfoproteínas/metabolismo , Factores de Transcripción/metabolismo , Activación Transcripcional/fisiología , Secuencia de Aminoácidos , Animales , Factores de Transcripción Básicos con Cremalleras de Leucinas y Motivos Hélice-Asa-Hélice , Células COS , Carcinoma Hepatocelular , Extractos Celulares , Núcleo Celular/metabolismo , AMP Cíclico/metabolismo , Proteínas de Unión al ADN/genética , Privación de Alimentos/fisiología , Factor Nuclear 4 del Hepatocito , Humanos , Hígado/enzimología , Hígado/metabolismo , Neoplasias Hepáticas , Datos de Secuencia Molecular , Mutación , Fosfoproteínas/genética , Fosforilación , Procesamiento Proteico-Postraduccional/fisiología , Ratas , Ratas Wistar , Serina/metabolismo , Factores de Transcripción/genética , Células Tumorales Cultivadas
9.
Mol Cell Biol ; 21(11): 3662-70, 2001 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-11340160

RESUMEN

The failure to transcribe the phenylalanine hydroxylase (PAH) gene in the liver of hepatocyte nuclear factor 1alpha (HNF1alpha)-deficient mice correlated with DNA hypermethylation and the presence of an inactive chromatin structure (M. Pontoglio, D. M. Faust, A. Doyen, M. Yaniv, and M. C. Weiss, Mol. Cell. Biol. 17:4948-4956, 1997). To evaluate the precise role played by HNF1alpha, DNA methylation, or histone acetylation in PAH gene silencing, we examined conditions that could restore PAH gene expression in HNF1alpha-deficient hepatocytes. We show that reactivation of PAH transcription can be achieved by reexpression of HNF1alpha in embryonic (i.e., embryonic day 12.5 [e12.5] to e13.5) hepatocytes but not in fetal (e17.5), newborn, and adult HNF1alpha-deficient hepatocytes. This defines a temporal competence window during which HNF1alpha can act to (re)program PAH gene transcription. We also show that PAH gene silencing can be partially relieved in HNF1alpha-deficient hepatocytes by treatment with the demethylating agent 5-azacytidine, even in the absence of HNF1alpha. Treatment using 5-azacytidine combined with trichostatin, a histone deacetylase inhibitor, resulted in a synergistic reactivation of the silenced PAH gene in adult hepatocytes, but this activity was not further increased by HNF1alpha reexpression. These results suggest that the HNF1alpha homeoprotein is involved in stage-specific developmental control of the methylation state and chromatin remodeling of the PAH gene.


Asunto(s)
Proteínas de Unión al ADN , Regulación Enzimológica de la Expresión Génica , Silenciador del Gen , Hígado/enzimología , Proteínas Nucleares , Fenilalanina Hidroxilasa/genética , Factores de Transcripción/fisiología , Activación Transcripcional , Acetilación , Animales , Animales Recién Nacidos , Azacitidina/farmacología , Células Cultivadas , Metilación de ADN , Desarrollo Embrionario y Fetal , Factor Nuclear 1 del Hepatocito , Factor Nuclear 1-alfa del Hepatocito , Factor Nuclear 1-beta del Hepatocito , Hepatocitos/citología , Hepatocitos/enzimología , Inhibidores de Histona Desacetilasas , Histonas , Ácidos Hidroxámicos/farmacología , Ratones , Factores de Transcripción/genética
10.
Mol Cell Biol ; 18(7): 3907-14, 1998 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-9632775

RESUMEN

DNA damage recognition by basal transcription factors follows different mechanisms. Using transcription-competition, nitrocellulose filter binding, and DNase I footprinting assays, we show that, although the general transcription factor TFIIH is able to target any kind of lesion which can be repaired by the nucleotide excision repair pathway, TATA binding protein (TBP)-TFIID is more selective in damage recognition. Only genotoxic agents which are able to induce kinked DNA structures similar to the one for the TATA box in its TBP complex are recognized. Indeed, DNase I footprinting patterns reveal that TBP protects equally 4 nucleotides upstream and 6 nucleotides downstream from the A-T (at position -29 of the noncoding strand) of the adenovirus major late promoter and from the G-G of a cisplatin-induced 1,2-d(GpG) cross-link. Together, our results may partially explain differences in transcription inhibition rates following DNA damage.


Asunto(s)
Daño del ADN , Proteínas de Unión al ADN/metabolismo , TATA Box , Factores de Transcripción TFII/metabolismo , Factores de Transcripción/metabolismo , Transcripción Genética , Adenoviridae/genética , Genes Virales , Humanos , Regiones Promotoras Genéticas , Proteína de Unión a TATA-Box , Factor de Transcripción TFIID
11.
Diabetes Metab ; 33(6): 395-402, 2007 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-17997341

RESUMEN

In the light of recent studies in humans and rodents, AMP-activated protein kinase (AMPK), a phylogenetically conserved serine/threonine protein kinase, has been described as an integrator of regulatory signals monitoring systemic and cellular energy status. AMP-activated protein kinase (AMPK) has been proposed to function as a 'fuel gauge' to monitor cellular energy status in response to nutritional environmental variations. Recently, it has been proposed that AMPK could provide a link in metabolic defects underlying progression to the metabolic syndrome. AMPK is a heterotrimeric enzyme complex consisting of a catalytic subunit alpha and two regulatory subunits beta and gamma. AMPK is activated by rising AMP and falling ATP. AMP activates the system by binding to the gamma subunit that triggers phosphorylation of the catalytic alpha subunit by the upstream kinases LKB1 and CaMKKbeta (calmodulin-dependent protein kinase kinase). AMPK system is a regulator of energy balance that, once activated by low energy status, switches on ATP-producing catabolic pathways (such as fatty acid oxidation and glycolysis), and switches off ATP-consuming anabolic pathways (such as lipogenesis), both by short-term effect on phosphorylation of regulatory proteins and by long-term effect on gene expression. As well as acting at the level of the individual cell, the system also regulates food intake and energy expenditure at the whole body level, in particular by mediating the effects of insulin sensitizing adipokines leptin and adiponectin. AMPK is robustly activated during skeletal muscle contraction and myocardial ischaemia playing a role in glucose transport and fatty acid oxidation. In liver, activation of AMPK results in enhanced fatty acid oxidation as well as decreased glucose production. Moreover, the AMPK system is one of the probable targets for the anti-diabetic drugs biguanides and thiazolidinediones. Thus, the relationship between AMPK activation and beneficial metabolic effects provide the rationale for the development of new therapeutic strategies in metabolic disorders.


Asunto(s)
Enfermedades Metabólicas/tratamiento farmacológico , Complejos Multienzimáticos/uso terapéutico , Proteínas Serina-Treonina Quinasas/uso terapéutico , Proteínas Quinasas Activadas por AMP , Animales , Apetito , Glucosa/metabolismo , Homeostasis , Humanos , Insulina/metabolismo , Secreción de Insulina , Células Secretoras de Insulina/metabolismo , Lípidos/fisiología , Hígado/metabolismo , Ratones , Ratones Noqueados , Complejos Multienzimáticos/genética , Complejos Multienzimáticos/metabolismo , Músculo Esquelético/fisiología , Isquemia Miocárdica/fisiopatología , Oxidación-Reducción , Proteínas Serina-Treonina Quinasas/genética , Proteínas Serina-Treonina Quinasas/metabolismo
12.
Nat Commun ; 7: 10856, 2016 Mar 08.
Artículo en Inglés | MEDLINE | ID: mdl-26952277

RESUMEN

Biguanides such as metformin have previously been shown to antagonize hepatic glucagon-stimulated cyclic AMP (cAMP) signalling independently of AMP-activated protein kinase (AMPK) via direct inhibition of adenylate cyclase by AMP. Here we show that incubation of hepatocytes with the small-molecule AMPK activator 991 decreases glucagon-stimulated cAMP accumulation, cAMP-dependent protein kinase (PKA) activity and downstream PKA target phosphorylation. Moreover, incubation of hepatocytes with 991 increases the Vmax of cyclic nucleotide phosphodiesterase 4B (PDE4B) without affecting intracellular adenine nucleotide concentrations. The effects of 991 to decrease glucagon-stimulated cAMP concentrations and activate PDE4B are lost in hepatocytes deleted for both catalytic subunits of AMPK. PDE4B is phosphorylated by AMPK at three sites, and by site-directed mutagenesis, Ser304 phosphorylation is important for activation. In conclusion, we provide a new mechanism by which AMPK antagonizes hepatic glucagon signalling via phosphorylation-induced PDE4B activation.


Asunto(s)
Proteínas Quinasas Activadas por AMP/metabolismo , AMP Cíclico/metabolismo , Fosfodiesterasas de Nucleótidos Cíclicos Tipo 4/metabolismo , Glucagón/metabolismo , Hepatocitos/enzimología , Proteínas Quinasas Activadas por AMP/genética , Secuencias de Aminoácidos , Animales , Células Cultivadas , Fosfodiesterasas de Nucleótidos Cíclicos Tipo 4/química , Fosfodiesterasas de Nucleótidos Cíclicos Tipo 4/genética , Activación Enzimática , Activadores de Enzimas/metabolismo , Hepatocitos/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Fosforilación , Transducción de Señal
13.
Oncogene ; 18(11): 1945-55, 1999 Mar 18.
Artículo en Inglés | MEDLINE | ID: mdl-10208416

RESUMEN

cdc2 gene expression is under the control of multiple factors. Although E2F/DP proteins have been reported to play a central role, they cannot account for all aspects of the fine modulation of cdc2 gene expression during cell cycle and embryonic development. To characterize the transcription factors that control cdc2 gene expression during nerve cell differentiation in avians, we have previously cloned the quail cdc2 gene promoter region. We had identified an octamer (CAGGTGGC) containing an E-box, which has important activity in regulating cdc2 transcription. Using in vivo genomic footprinting experiments, we show here that this motif, currently named IG, is the target of binding proteins at different stages of neuroretina development, confirming its importance as a regulatory response element for cdc2 gene expression. A subset of Helix-Loop-Helix family of transcription factors, known as Upstream Stimulatory Factors (USFs) specifically bind to this sequence as dimers. Moreover, our results indicate that USFs transactivate the promoter of cdc2 via the IG motif. These data may help to better understand the mechanisms that control cell division in differentiating nerve cells.


Asunto(s)
Proteína Quinasa CDC2/genética , Proteínas de Unión al ADN , Regulación de la Expresión Génica , Factores de Transcripción/metabolismo , Animales , Células COS , Huella de ADN , Humanos , Codorniz , Retina/metabolismo , Activación Transcripcional , Células Tumorales Cultivadas , Factores Estimuladores hacia 5'
14.
Biochim Biophys Acta ; 1574(2): 175-86, 2002 Mar 19.
Artículo en Inglés | MEDLINE | ID: mdl-11955627

RESUMEN

Glucose stimulates the transcription of the glucagon receptor gene in hepatocytes and in pancreatic beta cells. We recently identified a glucose response element in the immediate upstream non-coding region of the rat glucagon receptor gene. We previously showed that this DNA element is centered on a palindromic sequence of 19 nucleotides (termed as G box), containing two E boxes separated by three nucleotides. In the present study, we further characterized the DNA sequence requirements for the glucose induced expression of the rat glucagon receptor gene. Transfection study realized in the insulin-producing INS-1 cells revealed that a fragment of 49 nucleotides, centered on the G box, bears all the features required for the glucose activation. Mutations performed in the 5'-E box totally abolished the glucose responsiveness, whereas mutations or deletion of the 3'-E box only had a limited effect. Deletions performed upstream from the G box revealed that an accessory factor binding site, located in the region just upstream from the G box, is required for full stimulation by glucose. Finally, by using G box based probes in gel shift experiments, we demonstrated that USF1/USF2 transcription factors are part of the proteinic complex that binds to the glucose response element of the rat glucagon receptor gene promoter. In conclusion, in contrast to many other glucose regulated genes, only the 5'-E box appears to be a crucial DNA element for the glucose transcriptional effect. However, an accessory factor binding site located in the region just upstream from the G box is required for a complete stimulation by glucose.


Asunto(s)
Proteínas de Unión al ADN , Glucosa/genética , Insulina/metabolismo , Receptores de Glucagón/genética , Animales , Línea Celular , Elementos E-Box , Eliminación de Gen , Glucosa/farmacología , Ratas , Receptores de Glucagón/efectos de los fármacos , Elementos de Respuesta , Factores de Transcripción/metabolismo , Activación Transcripcional , Transfección , Factores Estimuladores hacia 5'
15.
Diabetes ; 50(7): 1515-21, 2001 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-11423471

RESUMEN

Mutations in the HNF4alpha gene are responsible for type 1 maturity-onset diabetes of the young (MODY1), which is characterized by a defect in insulin secretion. Hepatocyte nuclear factor (HNF)-4alpha is a transcription factor that plays a critical role in the transcriptional regulation of genes involved in glucose metabolism in both hepatocytes and pancreatic beta-cells. Recent evidence has implicated AMP-activated protein kinase (AMPK) in the modulation of both insulin secretion by pancreatic beta-cells and the control of glucose-dependent gene expression in both hepatocytes and beta-cells. Therefore, the question could be raised as to whether AMPK plays a role in these processes by modulating HNF-4alpha function. In this study, we show that activation of AMPK by 5-amino-4-imidazolecarboxamide riboside (AICAR) in hepatocytes greatly diminished HNF-4alpha protein levels and consequently downregulates the expression of HNF-4alpha target genes. Quantitative evaluation of HNF-4alpha target gene expression revealed diminished mRNA levels for HNF-1alpha, GLUT2, L-type pyruvate kinase, aldolase B, apolipoprotein (apo)-B, and apoCIII. Our data clearly demonstrate that the MODY1/HNF-4alpha transcription factor is a novel target of AMPK in hepatocytes. Accordingly, it can be suggested that in pancreatic beta-cells, AMPK also acts by decreasing HNF-4alpha protein level, and therefore insulin secretion. Hence, the possible role of AMPK in the physiopathology of type 2 diabetes should be considered.


Asunto(s)
Aminoimidazol Carboxamida/análogos & derivados , Proteínas de Unión al ADN , Diabetes Mellitus Tipo 1/metabolismo , Complejos Multienzimáticos/metabolismo , Fosfoproteínas/metabolismo , Proteínas Serina-Treonina Quinasas/metabolismo , Factores de Transcripción/metabolismo , Proteínas Quinasas Activadas por AMP , Aminoimidazol Carboxamida/farmacología , Animales , Apolipoproteína C-III , Apolipoproteínas B/biosíntesis , Apolipoproteínas B/genética , Apolipoproteínas C/biosíntesis , Apolipoproteínas C/genética , Células Cultivadas , Diabetes Mellitus Tipo 1/genética , Regulación hacia Abajo , Activación Enzimática , Fructosa-Bifosfato Aldolasa/biosíntesis , Fructosa-Bifosfato Aldolasa/genética , Regulación de la Expresión Génica/efectos de los fármacos , Transportador de Glucosa de Tipo 2 , Factor Nuclear 4 del Hepatocito , Insulina/metabolismo , Secreción de Insulina , Islotes Pancreáticos/enzimología , Hígado/enzimología , Proteínas de Transporte de Monosacáridos/genética , Proteínas de Transporte de Monosacáridos/metabolismo , Piruvato Quinasa/biosíntesis , Piruvato Quinasa/genética , ARN Mensajero/metabolismo , Ratas , Ratas Sprague-Dawley , Ribonucleótidos/farmacología , Factores de Tiempo , Transcripción Genética
16.
Oncogene ; 34(28): 3627-39, 2015 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-25241895

RESUMEN

As a sensor of cellular energy status, the AMP-activated protein kinase (AMPK) is believed to act in opposition to the metabolic phenotypes favored by proliferating tumor cells. Consequently, compounds known to activate AMPK have been proposed as cancer therapeutics. However, the extent to which the anti-neoplastic properties of these agonists are mediated by AMPK is unclear. Here we examined the AMPK dependence of six commonly used AMPK agonists (metformin, phenformin, 5-aminoimidazole-4-carboxamide ribonucleotide (AICAR), 2-deoxy-D-glucose (2DG), salicylate and A-769662) and their influence on cellular processes often deregulated in tumor cells. We demonstrate that the majority of these agonists display AMPK-independent effects on cell proliferation and metabolism with only the synthetic activator, A-769662, exerting AMPK-dependent effects on these processes. We find that A-769662 promotes an AMPK-dependent increase in mitochondrial spare respiratory capacity. Finally, contrary to the view of AMPK activity being tumor suppressive, we find that A-769662 confers a selective proliferative advantage to tumor cells growing under nutrient deprivation. Our results indicate that many of the antigrowth properties of these agonists cannot be attributed to AMPK activity in cells, and thus any observed effects using these agonists should be confirmed using AMPK-deficient cells. Ultimately, our data urge caution not only regarding the type of AMPK agonist proposed for cancer treatment but also the context in which they are used.


Asunto(s)
Adenilato Quinasa/metabolismo , Glucosa/metabolismo , Neoplasias/metabolismo , Pironas/farmacología , Salicilato de Sodio/farmacología , Tiofenos/farmacología , Aminoimidazol Carboxamida/análogos & derivados , Aminoimidazol Carboxamida/farmacología , Animales , Compuestos de Bifenilo , Ciclo Celular/efectos de los fármacos , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Células HCT116 , Células HEK293 , Humanos , Hipoglucemiantes/farmacología , Ácido Láctico/metabolismo , Metformina/farmacología , Ratones , Neoplasias/patología , Fenformina/farmacología , Ribonucleótidos/farmacología
17.
Cell Death Discov ; 1: 15063, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-27551487

RESUMEN

Cancer cells exhibit unique metabolic response and adaptation to the fluctuating microenvironment, yet molecular and biochemical events imprinting this phenomenon are unclear. Here, we show that metabolic homeostasis and adaptation to metabolic stress in cancer cells are primarily achieved by an integrated response exerted by the activation of AMPK. We provide evidence that AMPK-p38-PGC-1α axis, by regulating energy homeostasis, maintains survival in cancer cells under glucose-limiting conditions. Functioning as a molecular switch, AMPK promotes glycolysis by activating PFK2, and facilitates mitochondrial metabolism of non-glucose carbon sources thereby maintaining cellular ATP level. Interestingly, we noted that AMPK can promote oxidative metabolism via increasing mitochondrial biogenesis and OXPHOS capacity via regulating expression of PGC-1α through p38MAPK activation. Taken together, our study signifies the fundamental role of AMPK in controlling cellular bioenergetics and mitochondrial biogenesis in cancer cells.

18.
Life Sci ; 100(1): 55-60, 2014 Mar 28.
Artículo en Inglés | MEDLINE | ID: mdl-24530742

RESUMEN

AIMS: SIRT1 and AMP-activated protein kinase (AMPK) share common activators, actions and target molecules. Previous studies have suggested that a putative SIRT1-AMPK regulatory network could act as the prime initial sensor for calorie restriction-induced adaptations in skeletal muscle-the major site of insulin-stimulated glucose disposal. Our study aimed to investigate whether a feedback loop exists between AMPK and SIRT1 in skeletal muscle and how this may be involved glucose tolerance. MAIN METHODS: To investigate this, we used skeletal muscle-specific AMPKα1/2 knockout mice (AMPKα1/2(-/-)) fed ad libitum (AL) or a 30% calorie restricted (CR) diet and L6 rat myoblasts incubated with SIRT1 inhibitor (EX527). KEY FINDINGS: CR-AMPKα1/2(-/-) displayed impaired glucose tolerance (*p<0.05), in association with down-regulated SIRT1 and PGC-1α expression (<300% vs. CR-WT, (±±)p<0.01). Moreover, AMPK activity was decreased following SIRT1 inhibition in L6 cells (~0.5-fold vs. control, *p<0.05). SIGNIFICANCE: This study demonstrates that skeletal muscle-specific AMPK deficiency impairs the beneficial effects of CR on glucose tolerance and that these effects may be dependent on reduced SIRT1 levels.


Asunto(s)
Proteínas Quinasas Activadas por AMP/genética , Intolerancia a la Glucosa , Sirtuinas/metabolismo , Proteínas Quinasas Activadas por AMP/deficiencia , Acetilación , Animales , Restricción Calórica , Células Cultivadas , Represión Enzimática , Femenino , Ratones , Ratones Noqueados , Músculo Esquelético/enzimología , Coactivador 1-alfa del Receptor Activado por Proliferadores de Peroxisomas gamma , Procesamiento Proteico-Postraduccional , Transducción de Señal , Factores de Transcripción/genética , Factores de Transcripción/metabolismo
19.
Eur J Cancer ; 50(18): 3187-97, 2014 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-25450947

RESUMEN

AIM OF THE STUDY: Metabolic adaptations are essential during tumour growth to maintain the high proliferation levels exhibited by cancer cells. In this study, we examined the transformations that occurred in the lipid metabolism in astrocytic tumours, and the possible role of the fuel-sensing enzyme AMPK. Metabolic targets might help design new and effective drugs for cancer. METHODS: To accomplish this objective, we studied both mice and human astrocytic tumours. We first used a mouse model of astrocytoma driven by oncogenic H-RasV12 and/or with PTEN deletion based on the common constitutive activation of the Raf/MEK/ERK and PI3K/AKT cascades in human astrocytomas. We then confirmed the results in human glioblastoma cell lines and in glioblastoma tissue samples from patients. RESULTS: We show that the high levels of activated AMPK, observed in astrocytic tumours, increase extracellular lipid internalisation and reduce energy expenditure by inhibiting 'de novo' fatty acid (FA) synthesis, which allows tumour cells to obtain building blocks and energy to be able to create new organelles and new cells. CONCLUSIONS: Our findings demonstrate that AMPK plays a crucial role in glioblastoma cell growth and suggest that blocking lipoprotein receptors could potentially be used as a plausible therapeutic approach for these and other type of tumours with high levels of AMPK.


Asunto(s)
Proteínas Quinasas Activadas por AMP/metabolismo , Neoplasias Encefálicas/enzimología , Glioblastoma/enzimología , Metabolismo de los Lípidos/fisiología , Animales , Astrocitos/enzimología , Astrocitos/patología , Neoplasias Encefálicas/patología , Proliferación Celular/fisiología , Ácidos Grasos/biosíntesis , Glioblastoma/patología , Humanos , Ratones Noqueados , Fosfohidrolasa PTEN/antagonistas & inhibidores , Receptores de Lipoproteína/antagonistas & inhibidores , Receptores de Lipoproteína/metabolismo , Transfección , Células Tumorales Cultivadas
20.
Acta Physiol (Oxf) ; 211(4): 585-96, 2014 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-24934978

RESUMEN

AIM: Interleukin-6 (IL-6) is a major cytokine controlling body weight and metabolism, but because many types of cells can synthesize and respond to IL-6 considerable uncertainty still exists about the mechanisms underlying IL-6 effects. Therefore, the aim of this study was to analyse the effects of tissue-specific deletion of IL-6 using a fatty acid binding protein (aP2) promoter-Cre inducible system (aP2-Cre-ERT2). METHODS: Tissue-specific IL-6 KO mice (aP2-IL-6 KO mice) were produced upon tamoxifen administration and were fed a high-fat diet (HFD, 58.4% kcal from fat) or a control diet (18%) for 14 weeks. RESULTS: aP2-IL-6 KO female mice on a HFD gained less weight and adiposity than littermate wild-type mice, but these effects were not observed in males. Hypothalamic factors such as NPY and AgRP showed a pattern of expression consistent with this sex-specific phenotype. PGC-1α expression was increased in several tissues in aP2-IL-6 KO female mice, which is compatible with increased energy expenditure. Serum leptin, insulin, glucose, cholesterol and triglycerides levels were increased by HFD, and in females IL-6 deficiency reversed this effect in the case of insulin and cholesterol. HFD induced impaired responses to insulin and glucose tolerance tests, but no significant differences between genotypes were observed. CONCLUSION: The present results demonstrate that deletion of IL-6 driven by aP2-Cre regulates body weight, body fat and metabolism in a sex-specific fashion.


Asunto(s)
Adiposidad/fisiología , Dieta Alta en Grasa/efectos adversos , Proteínas de Unión a Ácidos Grasos/metabolismo , Interleucina-6/metabolismo , Aumento de Peso/fisiología , Animales , Femenino , Hibridación in Situ , Interleucina-6/genética , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Reacción en Cadena en Tiempo Real de la Polimerasa
SELECCIÓN DE REFERENCIAS
Detalles de la búsqueda