Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 15 de 15
Filtrar
1.
Int J Mol Sci ; 25(11)2024 May 24.
Artículo en Inglés | MEDLINE | ID: mdl-38891908

RESUMEN

Chronic inflammation causes muscle wasting. Because most inflammatory cytokine signals are mediated via TGF-ß-activated kinase-1 (TAK1) activation, inflammatory cytokine-induced muscle wasting may be ameliorated by the inhibition of TAK1 activity. The present study was undertaken to clarify whether TAK1 inhibition can ameliorate inflammation-induced muscle wasting. SKG/Jcl mice as an autoimmune arthritis animal model were treated with a small amount of mannan as an adjuvant to enhance the production of TNF-α and IL-1ß. The increase in these inflammatory cytokines caused a reduction in muscle mass and strength along with an induction of arthritis in SKG/Jcl mice. Those changes in muscle fibers were mediated via the phosphorylation of TAK1, which activated the downstream signaling cascade via NF-κB, p38 MAPK, and ERK pathways, resulting in an increase in myostatin expression. Myostatin then reduced the expression of muscle proteins not only via a reduction in MyoD1 expression but also via an enhancement of Atrogin-1 and Murf1 expression. TAK1 inhibitor, LL-Z1640-2, prevented all the cytokine-induced changes in muscle wasting. Thus, TAK1 inhibition can be a new therapeutic target of not only joint destruction but also muscle wasting induced by inflammatory cytokines.


Asunto(s)
Citocinas , Quinasas Quinasa Quinasa PAM , Atrofia Muscular , Animales , Quinasas Quinasa Quinasa PAM/metabolismo , Quinasas Quinasa Quinasa PAM/antagonistas & inhibidores , Atrofia Muscular/metabolismo , Atrofia Muscular/patología , Atrofia Muscular/etiología , Atrofia Muscular/tratamiento farmacológico , Ratones , Citocinas/metabolismo , Debilidad Muscular/metabolismo , Debilidad Muscular/tratamiento farmacológico , Miostatina/metabolismo , Miostatina/antagonistas & inhibidores , Proteínas Musculares/metabolismo , Factor de Necrosis Tumoral alfa/metabolismo , FN-kappa B/metabolismo , Inflamación/metabolismo , Inflamación/patología , Inflamación/tratamiento farmacológico , Transducción de Señal/efectos de los fármacos , Proteínas de Motivos Tripartitos/metabolismo , Proteínas de Motivos Tripartitos/genética , Modelos Animales de Enfermedad , Interleucina-1beta/metabolismo , Fosforilación/efectos de los fármacos , Músculo Esquelético/metabolismo , Músculo Esquelético/patología , Músculo Esquelético/efectos de los fármacos , Zearalenona/farmacología , Zearalenona/análogos & derivados
2.
J Med Invest ; 69(3.4): 287-293, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-36244782

RESUMEN

Objective : To examine diagnostic performance of corticotropin-releasing hormone (CRH) test combined with baseline dehydroepiandrosterone sulfate (DHEA-S) in patients with a suspect of central adrenal insufficiency. Methods : Patients (n=215) requiring daily or intermittent hydrocortisone replacement, or no replacement were retrospectively checked with their peak cortisol after CRH test and baseline DHEA-S. Results :  None of 106 patients with the peak cortisol ≥ 17.5 µg / dL after CRH test required replacement, and all 64 patients with the peak cortisol < 10.0 µg / dL required daily replacement. Among 8 patients with 10.0 µg / dL ≤ the peak cortisol < 17.5 µg / dL and baseline DHEA-S below the reference range, 6 patients required daily replacement and 1 patient was under intermittent replacement. Among 37 patients with 10.0 µg / dL ≤ the peak cortisol < 17.5 µg / dL and baseline DHEA-S within the reference range, 10 and 6 patients were under intermittent and daily replacement, respectively. Conclusions : No patients with the peak cortisol ≥ 17.5 µg / dL required hydrocortisone replacement, and all patients with the peak cortisol below 10.0 µg / dL required daily replacement. Careful clinical evaluation was required to determine requirement for replacement in patients with 10.0 µg / dL ≤ the peak cortisol < 17.5 µg / dL even in combination with baseline DHEA-S. J. Med. Invest. 69 : 287-293, August, 2022.


Asunto(s)
Insuficiencia Suprarrenal , Hidrocortisona , Insuficiencia Suprarrenal/diagnóstico , Insuficiencia Suprarrenal/tratamiento farmacológico , Hormona Adrenocorticotrópica , Algoritmos , Hormona Liberadora de Corticotropina , Sulfato de Deshidroepiandrosterona , Humanos , Estudios Retrospectivos
3.
JBMR Plus ; 3(7): e10182, 2019 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-31372589

RESUMEN

Activating mutations of calcium-sensing receptor (CaSR) cause autosomal dominant hypocalcemia type 1 (ADH1). Patients with ADH1 exhibit similar features to patients with hypoparathyroidism, including reduced serum parathyroid hormone (PTH) and Ca with low bone turnover. Although persistent suppression of bone turnover may increase bone fragility, bone strength in ADH1 patients has been unclear. We created knock-in mice harboring the A843E activating mutation of CaSR, mimicking severe features of ADH1 patients. The severe form of ADH1 model mice showed smaller body and bone size with lower bone mineral density (BMD) and cortical area of the femur compared with age-matched wild-type (WT) mice. Bone strength in the femur was lower in ADH1 mice even after correction by bone geometry and/or BMD. Microcracks were markedly increased in ADH1 mice, but were rarely detected in WT mice. There was a negative correlation between bone strength corrected by bone geometry and/or BMD and microcrack number or density in ADH1 and WT mice. Among ADH1 mice, negative correlation was still observed between bone strength and microcrack number or density. Microcracks increased with age in ADH1 mice, and were negatively correlated with bone strength. Treatment with PTH(1-34) or a calcilytic, JTT-305, increased bone turnover, reduced microcracks, and increased bone strength to similar levels to those in WT mice. The increase in microcracks was associated with a reduction in bone strength in ADH1 mice, and aging aggravates these changes. These results demonstrate that activating mutation of CaSR causes reduction in PTH secretion with suppressed bone turnover, that reduced bone turnover is associated with an age-dependent increase in microcracks with a reduction in bone strength, and that both PTH(1-34) and calcilytic ameliorate all these changes in bone turnover and strength. It is suggested that fracture susceptibility may be increased in severe types of ADH1 patients especially in the elderly. © 2019 The Authors. JBMR Plus published by Wiley Periodicals, Inc. on behalf of American Society for Bone and Mineral Research.

4.
Hepatol Commun ; 2(5): 571-581, 2018 May.
Artículo en Inglés | MEDLINE | ID: mdl-29761172

RESUMEN

Acetaminophen (APAP)-induced liver injury is closely associated with acute hepatic inflammation. Hypoxia-inducible factor-1 (HIF-1) is activated during immunological processes and regulates gene expressions in various types of immune cells. Although HIF-1 controls the differentiation and functions of conventional T cells in chronic inflammation, the pathological importance of HIF-1 in innate-like T cells during acute inflammation remains unknown. Here, we investigated the role of HIF-1 in innate-like γδ T cells during APAP-induced acute liver injury. In response to APAP administration, T-cell-specific Hif-1α gene knockout mice sustained severe liver damage compared to wild-type control mice but without any impacts on the initial hepatic insult. This severe liver damage was accompanied by excessive neutrophil infiltration into the liver, increased serum interleukin (IL)-17A levels, and increased hepatic expressions of C-X-C chemokine ligand (Cxcl) 1 and Cxcl2. Neutrophil depletion and IL-17A neutralization completely abolished the aggravated phenotypes in T-cell-specific Hif-1α gene knockout mice. Loss of the Hif-1α gene enhanced the aberrant accumulation of IL-17A-producing innate-like γδ T cells in the affected liver with no apparent effects on their IL-17A-producing ability. Adoptive transfer of Hif-1α-deficient splenic γδ T cells into recombination activating gene 2 (Rag2)-deficient mice aggravated APAP-induced liver injury with increased neutrophil accumulation in the liver compared to that of wild-type γδ T cells. Furthermore, Hif-1α-deficient γδ T cells selectively showed aberrantly enhanced migratory ability. This ability was totally abolished by treatment with the mitochondrial adenosine triphosphate synthase inhibitor oligomycin. Conclusion: Deletion of Hif-1α gene in T cells aggravates APAP-induced acute inflammatory responses by enhancing aberrant innate-like γδ T-cell recruitment, thereby increasing excessive neutrophil infiltration into the liver. (Hepatology Communications 2018;2:571-581).

5.
Exp Cell Res ; 359(1): 86-93, 2017 10 01.
Artículo en Inglés | MEDLINE | ID: mdl-28827061

RESUMEN

The mitochondria-associated ER membrane (MAM) is a specialized subdomain of ER that physically connects with mitochondria. Although disruption of inter-organellar crosstalk via the MAM impairs cellular homeostasis, its pathological significance in insulin resistance in type 2 diabetes mellitus remains unclear. Here, we reveal the importance of reduced MAM formation in the induction of fatty acid-evoked insulin resistance in hepatocytes. Palmitic acid (PA) repressed insulin-stimulated Akt phosphorylation in HepG2 cells within 12h. Treatment with an inhibitor of the ER stress response failed to restore PA-mediated suppression of Akt activation. Mitochondrial reactive oxygen species (ROS) production did not increase in PA-treated cells. Even short-term exposure (3h) to PA reduced the calcium flux from ER to mitochondria, followed by a significant decrease in MAM contact area, suggesting that PA suppressed the functional interaction between ER and mitochondria. Forced expression of mitofusin-2, a critical component of the MAM, partially restored MAM contact area and ameliorated the PA-elicited suppression of insulin sensitivity with Ser473 phosphorylation of Akt selectively improved. These results suggest that loss of proximity between ER and mitochondria, but not perturbation of homeostasis in the two organelles individually, plays crucial roles in PA-evoked Akt inactivation in hepatic insulin resistance.


Asunto(s)
Retículo Endoplásmico/metabolismo , Resistencia a la Insulina , Membranas Intracelulares/metabolismo , Mitocondrias/metabolismo , Ácido Palmítico/farmacología , Retículo Endoplásmico/efectos de los fármacos , Estrés del Retículo Endoplásmico/efectos de los fármacos , GTP Fosfohidrolasas , Células Hep G2 , Humanos , Insulina/farmacología , Membranas Intracelulares/efectos de los fármacos , Proteínas de la Membrana/metabolismo , Mitocondrias/efectos de los fármacos , Proteínas Mitocondriales/metabolismo , Fosforilación/efectos de los fármacos , Proteínas Proto-Oncogénicas c-akt/metabolismo , Especies Reactivas de Oxígeno/metabolismo , Transducción de Señal/efectos de los fármacos , eIF-2 Quinasa/metabolismo
6.
Sci Rep ; 7: 42959, 2017 02 20.
Artículo en Inglés | MEDLINE | ID: mdl-28218289

RESUMEN

Neuregulin1 is an epidermal growth factor (EGF)-like domain-containing protein that has multiple isoforms and functions as a local mediator in the control of various cellular functions. Here we show that type I isoform of neuregulin1 with an α-type EGF-like domain (Nrg1α) is the major isoform in mouse liver and regulates hepatic glucose production. Forced expression of Nrg1α in mouse liver enhanced systemic glucose disposal and decreased hepatic glucose production with reduced fasting blood glucose levels. Nuclear forkhead box protein O1 (FoxO1) and its downstream targets, PEPCK and G6Pase, were suppressed in liver and isolated hepatocytes by Nrg1α overexpression. In contrast, silencing of Nrg1α enhanced glucose production with increased PEPCK and G6Pase expressions in cAMP/dexamethasone-stimulated hepatocytes. Mechanistically, the recombinant α-type EGF-like domain of NRG1α (rNRG1α) stimulated the ERBB3 signalling pathway in hepatocytes, resulting in decreased nuclear FoxO1 accumulation via activation of both the AKT and ERK pathways. In addition, acute treatment with rNRG1α also suppressed elevation of blood glucose levels after both glucose and pyruvate challenge. Although a liver-specific deletion of Nrg1 gene in mice showed little effect on systemic glucose metabolism, these results suggest that NRG1α have a novel regulatory function in hepatic gluconeogenesis by regulating the ERBB3-AKT/ERK-FoxO1 cascade.


Asunto(s)
Gluconeogénesis , Neurregulina-1/metabolismo , Animales , Células Cultivadas , Dexametasona/farmacología , Proteína Forkhead Box O1/metabolismo , Gluconeogénesis/efectos de los fármacos , Glucosa/metabolismo , Glucosa-6-Fosfatasa/genética , Glucosa-6-Fosfatasa/metabolismo , Hepatocitos/citología , Hepatocitos/efectos de los fármacos , Hepatocitos/metabolismo , Hígado/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Neurregulina-1/antagonistas & inhibidores , Neurregulina-1/genética , Fosfoenolpiruvato Carboxiquinasa (GTP)/genética , Fosfoenolpiruvato Carboxiquinasa (GTP)/metabolismo , Isoformas de Proteínas/antagonistas & inhibidores , Isoformas de Proteínas/genética , Isoformas de Proteínas/metabolismo , Interferencia de ARN , ARN Interferente Pequeño/metabolismo , Receptor ErbB-3/metabolismo , Proteínas Recombinantes/biosíntesis , Proteínas Recombinantes/genética , Proteínas Recombinantes/farmacología , Transducción de Señal/efectos de los fármacos
7.
J Hepatol ; 2015 Feb 10.
Artículo en Inglés | MEDLINE | ID: mdl-25681160

RESUMEN

This article has been withdrawn at the request of the author(s) and/or editor. The Publisher apologizes for any inconvenience this may cause. The full Elsevier Policy on Article Withdrawal can be found at http://www.elsevier.com/locate/withdrawalpolicy.

8.
World J Gastroenterol ; 20(41): 15087-97, 2014 Nov 07.
Artículo en Inglés | MEDLINE | ID: mdl-25386057

RESUMEN

The liver is a central organ that metabolizes excessive nutrients for storage in the form of glycogen and lipids and supplies energy-producing substrates to the peripheral tissues to maintain their function, even under starved conditions. These processes require a considerable amount of oxygen, which causes a steep oxygen gradient throughout the hepatic lobules. Alcohol consumption and/or excessive food intake can alter the hepatic metabolic balance drastically, which can precipitate fatty liver disease, a major cause of chronic liver diseases worldwide, ranging from simple steatosis, through steatohepatitis and hepatic fibrosis, to liver cirrhosis. Altered hepatic metabolism and tissue remodeling in fatty liver disease further disrupt hepatic oxygen homeostasis, resulting in severe liver hypoxia. As master regulators of adaptive responses to hypoxic stress, hypoxia-inducible factors (HIFs) modulate various cellular and organ functions, including erythropoiesis, angiogenesis, metabolic demand, and cell survival, by activating their target genes during fetal development and also in many disease conditions such as cancer, heart failure, and diabetes. In the past decade, it has become clear that HIFs serve as key factors in the regulation of lipid metabolism and fatty liver formation. This review discusses the molecular mechanisms by which hypoxia and HIFs regulate lipid metabolism in the development and progression of fatty liver disease.


Asunto(s)
Hígado Graso/metabolismo , Hipoxia/metabolismo , Metabolismo de los Lípidos , Hígado/metabolismo , Oxígeno/metabolismo , Animales , Proteínas Reguladoras de la Apoptosis , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/metabolismo , Hígado Graso/diagnóstico , Hígado Graso/etiología , Humanos , Hipoxia/complicaciones , Hipoxia/diagnóstico , Subunidad alfa del Factor 1 Inducible por Hipoxia/metabolismo , Hígado/patología , Obesidad/complicaciones , Obesidad/metabolismo , Proteínas Represoras , Factores de Riesgo , Transducción de Señal , Apnea Obstructiva del Sueño/complicaciones , Apnea Obstructiva del Sueño/metabolismo
9.
Int J Hematol ; 95(5): 457-63, 2012 May.
Artículo en Inglés | MEDLINE | ID: mdl-22535382

RESUMEN

Over the course of evolution, aerobic organisms have developed sophisticated systems for responding to alterations in oxygen concentration, as oxygen acts as a final electron acceptor in oxidative phosphorylation for energy production. Hypoxia-inducible factor (HIF) plays a central role in the adaptive regulation of energy metabolism, by triggering a switch from mitochondrial oxidative phosphorylation to anaerobic glycolysis in hypoxic conditions. HIF also reduces oxygen consumption in mitochondria by inhibiting conversion of pyruvate to acetyl CoA, suppressing mitochondrial biogenesis and activating autophagy of mitochondria concomitantly with reduction in reactive oxygen species production. In addition, metabolic reprogramming in response to hypoxia through HIF activation is not limited to the regulation of carbohydrate metabolism; it occurs in lipid metabolism as well. Recent studies using in vivo gene-targeting technique have revealed unexpected, but novel functions of HIF in energy metabolism in a context- and cell type-specific manner, and shed light on the possibility of pharmaceutical targeting HIF as a new therapy against many diseases, including cancer, diabetes, and fatty liver.


Asunto(s)
Metabolismo Energético , Factor 1 Inducible por Hipoxia/metabolismo , Animales , Metabolismo de los Hidratos de Carbono , Humanos , Hipoxia/metabolismo , Ácido Láctico/metabolismo , Metabolismo de los Lípidos , Consumo de Oxígeno , Ácido Pirúvico/metabolismo
10.
J Hepatol ; 56(2): 441-7, 2012 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-21896344

RESUMEN

BACKGROUND & AIMS: Chronic alcohol intake stimulates hepatic oxygen consumption and subsequently causes liver hypoxia, leading to activation of hypoxia inducible factor-1 (HIF-1). Although HIF-1 plays a crucial role in the metabolic switch from aerobic to anaerobic metabolism in response to hypoxia, its roles in the regulation of lipid metabolism in alcoholic fatty liver remain unknown. METHODS: Wild-type and hepatocyte-specific HIF-1α-null mice were subjected to a 6% ethanol-containing liquid diet for 4 weeks, and functional effects of loss of the HIF-1α gene on lipid metabolism were examined in the liver. RESULTS: Hepatocyte-specific HIF-1α-null mice developed severe hypertriglyceridemia with enhanced accumulation of lipids in the liver of mice exposed to a 6% ethanol-containing liquid diet for 4 weeks. Sterol regulatory element-binding protein 1c (SREBP-1c) and its downstream target acetyl-CoA carboxylase were greatly activated as the hepatic steatosis progressed, and these alterations were inversely correlated with the expression of the HIF-1-regulated gene DEC1. Overexpression of DEC1 in the mutant liver abrogated the detrimental effects of loss of HIF-1α gene on ethanol-induced fatty liver with reduced SREBP-1c expression. Conversely, co-administration of the HIF hydroxylase inhibitor dimethyloxalylglycine for the last 2 weeks improved markedly the ethanol-induced fatty liver in mice. CONCLUSIONS: The current results provide direct evidence for protective roles of HIF-1 induction in the development of ethanol-induced fatty liver via activation of the HIF-1-regulated transcriptional repressor DEC1.


Asunto(s)
Hígado Graso Alcohólico/metabolismo , Hígado Graso Alcohólico/prevención & control , Subunidad alfa del Factor 1 Inducible por Hipoxia/biosíntesis , Metabolismo de los Lípidos , Acetil-CoA Carboxilasa/metabolismo , Aminoácidos Dicarboxílicos/farmacología , Animales , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/metabolismo , Susceptibilidad a Enfermedades , Etanol/administración & dosificación , Hígado Graso Alcohólico/genética , Expresión Génica/efectos de los fármacos , Proteínas de Homeodominio/metabolismo , Subunidad alfa del Factor 1 Inducible por Hipoxia/deficiencia , Subunidad alfa del Factor 1 Inducible por Hipoxia/genética , Hígado/efectos de los fármacos , Hígado/metabolismo , Masculino , Ratones , Ratones Noqueados , Proteína 1 de Unión a los Elementos Reguladores de Esteroles/genética , Proteína 1 de Unión a los Elementos Reguladores de Esteroles/metabolismo
11.
Biochem Biophys Res Commun ; 415(3): 445-9, 2011 Nov 25.
Artículo en Inglés | MEDLINE | ID: mdl-22051049

RESUMEN

The liver plays a central role in glucose homeostasis in the whole-body by responding to environmental factors including nutrients, hormones, and oxygen. In conditions of metabolic overload such as diabetes mellitus and obesity, coordinated regulation between oxygen supply and consumption has been reported to be disrupted and subsequently cause tissue hypoxia, although pathological significance of the disease-related hypoxia remains elusive. To investigate the role of tissue hypoxia in the liver on systemic glucose homeostasis, mice lacking HIF-1α gene, a critical component of a master regulator of hypoxic response, in hepatocytes were exposed to high fat/sucrose diet (HFSD). Exposure to HFSD for 5 weeks elicited liver hypoxia with a transient increase in HIF-1α protein expression in the liver of control mice. Glucose disposal was marginally impaired in control mice when challenged oral glucose tolerance test, but such impairment was enhanced in the mutant mice. This alteration was accompanied by a complete inhibition of glucokinase induction with a significant reduction of hepatic glucose uptake. Mice fed HFSD for 20 weeks exhibited fasting hyperglycemia and glucose intolerance, whereas these metabolic phenotypes deteriorated considerably with severe insulin resistance in skeletal muscles and adipose tissues in the mutant mice. These findings suggest that HIF-1 in hepatocytes plays protective roles against the progression of diabetes mellitus.


Asunto(s)
Dieta Alta en Grasa/efectos adversos , Glucosa/metabolismo , Hepatocitos/metabolismo , Subunidad alfa del Factor 1 Inducible por Hipoxia/metabolismo , Obesidad/metabolismo , Animales , Diabetes Mellitus/metabolismo , Prueba de Tolerancia a la Glucosa , Subunidad alfa del Factor 1 Inducible por Hipoxia/genética , Resistencia a la Insulina , Ratones , Ratones Obesos , Obesidad/etiología , Sacarosa/administración & dosificación , Sacarosa/efectos adversos
12.
J Antibiot (Tokyo) ; 62(8): 425-9, 2009 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-19575044

RESUMEN

We searched for compounds that affect the cyclin D1/retinoblastoma protein pathway from the in-house natural product library using a recombinant adenovirus with the Cre/loxP-regulated cyclin D1 overexpression system, and we found that oligomycin inhibited cell growth more effectively in cyclin D1-overexpressing SW480 cells than in control SW480 cells. We also found that oligomycin reduced the expression levels of cyclin D1 protein and that this reduction is, at least in part, mediated by Thr-286 phosphorylation-dependent proteasomal degradation.


Asunto(s)
Ciclina D1/metabolismo , Genes bcl-1/efectos de los fármacos , Oligomicinas/farmacología , Complejo de la Endopetidasa Proteasomal/efectos de los fármacos , Adenoviridae/genética , Western Blotting , Línea Celular , Citometría de Flujo , Expresión Génica , Vectores Genéticos , Humanos , Indicadores y Reactivos , Plásmidos/genética , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Sales de Tetrazolio , Tiazoles , Transfección
13.
Gene ; 438(1-2): 49-56, 2009 Jun 01.
Artículo en Inglés | MEDLINE | ID: mdl-19303924

RESUMEN

Overexpression of fibroblast growth factor receptors (FGFRs) has been observed in many types of human tumors; however, the regulatory mechanism of human FGFR expression is still largely unknown. In the present study, we first identified the transcriptional initiation site in the human FGFR 1 gene by 5'-RACE. Furthermore, we show that the expression of human FGFR 1 is regulated by E2F-1. Characterization of the human FGFR 1 promoter demonstrated that two non-consensus E2F binding sequences at positions +4 to +22 and +25 to +43 relative to our identified transcriptional initiation site in the human FGFR 1 gene were critical for E2F-1-mediated transactivation of human FGFR 1 promoter. Mutations of these sites completely abolished the response of human FGFR 1 promoter to E2F-1 as well as E2F-1 binding in electrophoretic mobility-shift assays. Furthermore, chromatin immunoprecipitation assay showed that E2F-1 was able to bind in vivo to the human FGFR 1 promoter. Moreover, human FGFR 1 protein expression was up-regulated by the overexpression of E2F-1, but down-regulated by the overexpression of pRB in situ, suggesting that the expression of human FGFR 1 is regulated by the pRB/E2F pathway. Because disruption of the pRB/E2F pathway is frequently observed in tumor cells, our findings provide valuable information for studying the role of FGFR 1 in tumor progression.


Asunto(s)
Factor de Transcripción E2F1/genética , Regulación de la Expresión Génica , Receptor Tipo 1 de Factor de Crecimiento de Fibroblastos/genética , Factores de Transcripción/fisiología , Adenoviridae/genética , Secuencia de Bases , Sitios de Unión/genética , Línea Celular Tumoral , Inmunoprecipitación de Cromatina , Ensayo de Cambio de Movilidad Electroforética , Genes Reporteros , Vectores Genéticos , Humanos , Luciferasas/metabolismo , Datos de Secuencia Molecular , Plásmidos , Regiones Promotoras Genéticas , Unión Proteica/genética , Receptor Tipo 1 de Factor de Crecimiento de Fibroblastos/metabolismo , Proteína de Retinoblastoma/fisiología , Sitio de Iniciación de la Transcripción , Activación Transcripcional
14.
Biosci Biotechnol Biochem ; 72(3): 759-66, 2008 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-18323664

RESUMEN

Recently, we identified a novel human platelet-derived growth factor (PDGF) receptor alpha mRNA (type II) that differs from the known human PDGF receptor alpha mRNA (type I) only in its 5'-untranslated region and is regulated by E2F-1. In this study, we determined the nucleotide sequence of the mouse type II PDGF receptor alpha mRNA and found that the expression of this transcript was regulated by E2F-1 through an E2F-1-responsive sequence located +711/+718 downstream of the transcription start site of the type I transcript. Furthermore, we observed embryo-predominant expression of the type II transcript.


Asunto(s)
Factor de Transcripción E2F1/fisiología , ARN Mensajero/genética , Receptor alfa de Factor de Crecimiento Derivado de Plaquetas/genética , Animales , Secuencia de Bases , Embrión de Mamíferos , Regulación de la Expresión Génica , Ratones , Regiones Promotoras Genéticas , ARN Mensajero/aislamiento & purificación , Elementos de Respuesta , Transcripción Genética
15.
Gene ; 403(1-2): 89-97, 2007 Nov 15.
Artículo en Inglés | MEDLINE | ID: mdl-17881156

RESUMEN

Platelet-derived growth factors (PDGFs) and their receptors play an important role in cell proliferation, angiogenesis, and differentiation during normal development, and have also been implicated in tumorigenesis. In this study, we identified a novel variant of human PDGF receptor alpha mRNA (type II), which contains the same open reading frame as the known PDGF receptor alpha mRNA (type I) but a different 5'-untranslated region (5'-UTR). The 5'-UTR of the type II transcript was identified as a 363-bp exon located in intron 1 at position +1,210 to +1,572 relative to the transcriptional initiation site of the type I transcript. This type II transcript was expressed in a subset of human cell lines, such as MG-63 and MNNG/HOS cells. Moreover, transcription of the type II, but not the type I, was regulated by E2F-1 through an E2F-1-responsive site located at position +1,086/+1,093 downstream of the transcriptional initiation site of the type I transcript. Furthermore, epigenetic modulation might be involved in the expression of the type II transcript. Our findings provide new insights into the regulatory mechanism of PDGF receptor alpha transcription in normal and tumor cells.


Asunto(s)
Factor de Transcripción E2F1/metabolismo , Variación Genética , Receptor alfa de Factor de Crecimiento Derivado de Plaquetas/genética , Transcripción Genética , Regiones no Traducidas 5' , Emparejamiento Base , Línea Celular , Línea Celular Tumoral , Cartilla de ADN , Factor de Transcripción E2F1/genética , Epigénesis Genética , Exones , Regulación de la Expresión Génica , Genes Reporteros , Humanos , Intrones , Riñón/citología , Luciferasas/metabolismo , Modelos Genéticos , Sistemas de Lectura Abierta , Osteosarcoma/patología , Plásmidos , ARN Mensajero/genética , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Sitio de Iniciación de la Transcripción
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...