Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 51
Filtrar
Más filtros

Banco de datos
País/Región como asunto
Tipo del documento
País de afiliación
Intervalo de año de publicación
4.
Ren Fail ; 37(1): 160-4, 2015 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-25222109

RESUMEN

Recent studies have identified that high glucose-induced renal tubular cell damage. We previously demonstrated that high glucose treatment induced oxidative stress in human renal proximal tubular epithelial cells (RPTECs), and angiotensin II type 1 (AT1) receptor blockers reduce high glucose-induced oxidative stress in RPTEC possibly via blockade of intracellular as well as extracellular AT1 receptor. However, exact roles of tumor necrosis factor (TNF)-α and AT1 receptor on high glucose-induced renal tubular function remain unclear. N-acetyl-beta-glucosaminidase (NAG), concentrations of TNF-α/angiotensin II and p22(phox) protein levels after high glucose treatment with or without AT1 receptor blocker or thalidomide, an inhibitor of TNF-α protein synthesis, were measured in immortalized human renal proximal tubular epithelial cells (HK2 cells). AT1 receptor knockdown was performed with AT1 receptor small interfering RNA (siRNA). High glucose treatment (30 mM) significantly increased NAG release, TNF-α/angiotensin II concentrations in cell media and p22(phox) protein levels compared with those in regular glucose medium (5.6 mM). Candesartan, an AT1R blocker, showed a significant reduction on high glucose-induced NAG release, TNF-α concentrations and p22(phox) protein levels in HK2 cells. In addition, significant decreases of NAG release, TNF-α concentrations and p22(phox) protein levels in HK2 cells were observed in high glucose-treated group with thalidomide. AT1R knockdown with siRNA markedly reversed high glucose, angiotensin II or TNF-α-induced p22(phox) protein levels in HK2 cells. TNF-α may be involved in high glucose-induced renal tubular damage in HK2 cells possibly via AT1 receptor signaling.


Asunto(s)
Células Epiteliales , Glucosa , Túbulos Renales Proximales , Receptor de Angiotensina Tipo 1/metabolismo , Factor de Necrosis Tumoral alfa/metabolismo , Acetilglucosaminidasa/metabolismo , Angiotensina II/metabolismo , Bloqueadores del Receptor Tipo 1 de Angiotensina II/farmacología , Células Cultivadas , Relación Dosis-Respuesta a Droga , Células Epiteliales/efectos de los fármacos , Células Epiteliales/metabolismo , Células Epiteliales/patología , Glucosa/efectos adversos , Glucosa/metabolismo , Humanos , Túbulos Renales Proximales/efectos de los fármacos , Túbulos Renales Proximales/metabolismo , Túbulos Renales Proximales/patología , Estrés Oxidativo/efectos de los fármacos , Transducción de Señal/efectos de los fármacos
6.
Steroids ; 183: 109021, 2022 07.
Artículo en Inglés | MEDLINE | ID: mdl-35339573

RESUMEN

The 11ß hydroxysteroid dehydrogenase type-1 (11ßHSD-1) is a predominant 11ß-reductase regenerating bioactive glucocorticoids (cortisol, corticosterone) from inactive 11-keto forms (cortisone, dehydrocorticosterone), expressed mainly in the brain, liver and adipose tissue. Although the expression levels of 11ß HSD-1 mRNA are known to be influenced by glucocorticoids, its tissue-specific regulation is not completely elucidated. In this study, we examined the effect of persistent glucocorticoid excess on the expression of 11ß HSD-1 mRNA in the hippocampus, liver, and abdominal adipose tissue in vivo using quantitative real-time PCR. We found that, in C57BL/6J mice treated with corticosterone (CORT) pellet for 2 weeks, 11ß HSD-1 mRNA decreased in the hippocampus (HIPP) and liver, whereas it increased in the abdominal fat (FAT), compared with placebo treatment [HIPP: placebo 1.00 ± 0.14, CORT 0.63 ± 0.04; liver: placebo 1.00 ± 0.08, CORT 0.73 ± 0.06; FAT: placebo 1.00 ± 0.16, CORT 2.26 ± 0.39]. Moreover, in CRH transgenic mice, an animal model of Cushing's syndrome with high plasma CORT level, 11ß HSD-1 mRNA was also decreased in the hippocampus and liver, and increased in the abdominal adipose tissue compared to that in wild-type mice. These changes were reversed after adrenalectomy in CRH-Tg mice. Altogether, these results reveal the differential regulation of 11ß HSD-1 mRNA by glucocorticoid among the tissues examined.


Asunto(s)
11-beta-Hidroxiesteroide Deshidrogenasa de Tipo 1 , Síndrome de Cushing , 11-beta-Hidroxiesteroide Deshidrogenasa de Tipo 1/genética , Animales , Corticosterona/metabolismo , Síndrome de Cushing/genética , Modelos Animales de Enfermedad , Glucocorticoides/farmacología , Hidroxiesteroide Deshidrogenasas/genética , Hidroxiesteroide Deshidrogenasas/metabolismo , Ratones , Ratones Endogámicos C57BL , ARN Mensajero/genética , ARN Mensajero/metabolismo
7.
Endocr J ; 58(4): 279-86, 2011.
Artículo en Inglés | MEDLINE | ID: mdl-21389639

RESUMEN

Although glucocorticoid-induced hyperphagia is observed in the patients with glucocorticoid treatment or Cushing's syndrome, its molecular mechanism is not clear. We thus explored the expression of neuropeptide mRNAs in the hypothalamus related to appetite regulation in CRH over-expressing transgenic mice (CRH-Tg), a model of Cushing's syndrome. We measured food intake, body weight (including body fat weight) and plasma corticosterone levels in CRH-Tg and their wild-type littermates (WT) at 6 and 14 weeks old. We also examined neuropeptide Y (NPY), proopiomelanocortin (POMC) and Agouti-related protein (AgRP) mRNAs in the arcuate nucleus (ARC) using in situ hybridization. Circulating corticosterone levels in CRH-Tg were markedly elevated at both 6 and 14 weeks old. Body fat weight in CRH-Tg was significantly increased at 14 weeks old, which is considered as an effect of chronic glucocorticoid excess. At both 6 and 14 weeks old, CRH-Tg mice showed significant hyperphagia compared with WT (14w old: WT 3.9±0.1, CRH-Tg 5.1±0.7 g/day, p<0.05). Unexpectedly, NPY mRNA levels in CRH-Tg were significantly decreased at 14 weeks old (WT: 1571.5±111.2, CRH-Tg: 949.1±139.3 dpm/mg, p<0.05), and there were no differences in POMC mRNA levels between CRH-Tg and WT. On the other hand, AgRP mRNA levels in CRH-Tg were significantly increased compared with WT at both ages (14w old: WT 365.6±88.6, CRH-Tg 660.1±87.2 dpm/ mg, p<0.05). These results suggest that glucocorticoid-induced hyperphagia is associated with increased hypothalamic AgRP. Our results also indicate that hypothalamic NPY does not have an essential role in the increased food intake during glucocorticoid excess.


Asunto(s)
Proteína Relacionada con Agouti/genética , Núcleo Arqueado del Hipotálamo/metabolismo , Hormona Liberadora de Corticotropina/genética , Glucocorticoides/farmacología , Hiperfagia/inducido químicamente , Tejido Adiposo/metabolismo , Animales , Peso Corporal/genética , Corticosterona/sangre , Síndrome de Cushing/fisiopatología , Modelos Animales de Enfermedad , Ingestión de Alimentos/efectos de los fármacos , Ingestión de Alimentos/genética , Masculino , Ratones , Ratones Transgénicos , Neuropéptido Y/metabolismo , Proopiomelanocortina/metabolismo , ARN Mensajero/metabolismo
9.
Pituitary ; 13(1): 78-9, 2010.
Artículo en Inglés | MEDLINE | ID: mdl-19915981

RESUMEN

A 55-year-old woman with signs of acromegaly was referred to our hospital. Endocrinological examinations showed that she had high levels of growth hormone (GH; 5.54 ng ml(-1); normal range: 0.66-3.68 ng ml(-1)) and insulin-like growth factor-I (IGF-I; 508 ng ml(-1); normal range: 37-266 ng ml(-1)) levels, incomplete suppression of serum GH following a 75-gram oral glucose tolerance test (oGTT; trough GH 3.66 ng ml(-1)), and paradoxical GH responses to a TRH provocation test (peak GH 38.9 ng ml(-1)). Dynamic magnetic resonance imaging (MRI) suggested the presence of an intrasellar mass lesion (5.9 x 2.8 mm) in the left part of her pituitary gland (Fig. 1a, upper panel). F-18 fluorodeoxyglucose (FDG) positron emission tomographic (PET) imaging clearly showed focal but remarkable FDG uptake (Fig. 1a, lower panel), consistent with the localization of the tumor suspected on MRI. The tumor was removed by transsphenoidal surgery. Subsequent histological analysis confirmed the diagnosis of a GH-producing pituitary adenoma. After removal, serum IGF-I levels decreased to a normal range (178 ng ml(-1)), and serum GH was appropriately suppressed during oGTT (trough GH 0.30 ng ml(-1)), suggesting that complete resection was obtained [1]. While postsurgical changes made it difficult to detect any residual lesion on MRI (Fig. 1b, upper panel), abnormal FDG uptake was not seen on FDG-PET after surgery (Fig. 1b, lower panel). PET scans are reported to be a valuable tool for the detection of pituitary adenomas [2-4]. This case clearly showed that FDG-PET is also useful for re-evaluation of the disease after surgery. PET scans are recommended for patients with equivocal pituitary mass lesions on conventional MRI, and for follow-up examinations after surgery.


Asunto(s)
Dihidroxifenilalanina , Radioisótopos de Flúor , Adenoma Hipofisario Secretor de Hormona del Crecimiento/diagnóstico , Tomografía de Emisión de Positrones/métodos , Dihidroxifenilalanina/farmacocinética , Femenino , Adenoma Hipofisario Secretor de Hormona del Crecimiento/cirugía , Humanos , Imagen por Resonancia Magnética , Persona de Mediana Edad , Tomografía de Emisión de Positrones/normas
10.
Endocr J ; 57(4): 317-24, 2010.
Artículo en Inglés | MEDLINE | ID: mdl-20139635

RESUMEN

Both glucocorticoid and insulin are known to have an anabolic effect on lipogenesis. Acetyl-CoA, an intermediate product of glycolysis, is supplied for fatty acid synthesis when carbohydrate intake is sufficient. Acetyl-CoA carboxylase (ACC), consisting of two isoenzymes ACC1 and ACC2, mediates the conversion from acetyl-CoA to malonyl-CoA, and thus plays a key role for the regulation of lipogenesis. In this study, we surveyed the effects of glucocorticoid and insulin on the transcriptional activity of the alternative promoters of ACCs (PI-PIII for ACC1, and PI and PII for ACC2) using the HepG2 human hepatocyte cell line in vitro. We also examined the roles of the insulin and/or glucose-regulated transcriptional factor(s) such as SREBP1c, LXRalpha/beta, and ChREBP on each promoter of the ACC genes. We found that both insulin and glucocorticoid had potent positive effects on all the promoters examined, and additive effects of both hormones were recognized in ACC1 PI and ACC2 PI. Furthermore, a representative insulin-responsive transcription factor SREBP1c showed significant stimulatory effects on all the promoters of ACC genes, among which those on ACC1 PIII and ACC2 PI were most prominent. On the other hand, the effect of LXRalpha was rather selective; it showed a marked stimulatory effect only on ACC1 PII. LXRbeta and ChREBP had minimal, if any, effects on some of the promoters. Altogether, our data suggest that insulin and glucocorticoid have positive effects on both ACC1 and ACC2 gene transcription. SREBP1c might be a master regulator of the expression of both genes regardless of the promoter utilized, whereas LXRalpha seems to play a promoter-specific role. Since ACC1 facilitates lipogenesis by stimulating fatty acid synthesis and ACC2 inhibits lipolysis, both insulin and glucocorticoid seem to play an important role in the pathogenesis of obesity and/or hepatic steatosis.


Asunto(s)
Acetil-CoA Carboxilasa/genética , Regulación Enzimológica de la Expresión Génica/efectos de los fármacos , Glucocorticoides/farmacología , Insulina/farmacología , Factores de Transcripción Básicos con Cremalleras de Leucinas y Motivos Hélice-Asa-Hélice/genética , Factores de Transcripción Básicos con Cremalleras de Leucinas y Motivos Hélice-Asa-Hélice/fisiología , Expresión Génica , Regulación Enzimológica de la Expresión Génica/fisiología , Células Hep G2 , Hepatocitos/enzimología , Humanos , Isoenzimas/genética , Cinética , Receptores X del Hígado , Receptores Nucleares Huérfanos/genética , Receptores Nucleares Huérfanos/fisiología , Regiones Promotoras Genéticas/genética , ARN Mensajero/análisis , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Proteína 1 de Unión a los Elementos Reguladores de Esteroles/genética , Proteína 1 de Unión a los Elementos Reguladores de Esteroles/fisiología , Transcripción Genética/efectos de los fármacos , Transfección
11.
Endocr J ; 57(5): 403-13, 2010.
Artículo en Inglés | MEDLINE | ID: mdl-20160399

RESUMEN

NAD-dependent deacetylase SIRT1 is known to be activated by caloric restriction and is related to longevity. A natural polyphenolic compound resveratrol is also shown to increases SIRT1 activity and extends lifespan. However, the transcriptional regulation of SIRT1 gene has not completely examined in the context of metabolism. Thus, in this study, we characterized the 5' -flanking region of human SIRT1 gene. We first found that representative metabolic hormones and related factors (glucocorticoid, glucagon/cAMP, and insulin) did not show significant effect on SIRT1 gene transcription. PPARalpha and PPARgamma1 without/with their specific ligands did not have significant effect as well. In contrast, expression of PPARbeta/delta (PPARdelta markedly increased the 5' -promoter activity of SIRT1 gene, which was further amplified by the addition of GW501516, a selective PPARdelta agonist. Deletion/mutation mapping analyses failed to identify PPAR binding element but revealed the presence of canonical Sp1 binding site, which was conserved among species. The Sp1 site is functional, because Sp1 overexpresson significantly enhanced SIRT1 promoter activity, and the binding of Sp1 to the element was confirmed by EMSA and ChIP assays. Interestingly, specific Sp1 antagonist mithramycin completely abolished the PPARdelta-mediated induction of SIRT1 gene transcription. Altogether, our data suggest the predominant role of PPARdelta in the transcriptional regulation of SIRT1 gene. Furthermore, the effects of PPARdelta seem to be mediated by Sp1. We assume that, in vivo, starvation increases lipolysis-derived free fatty acid and activates PPARdelta and the resultant increase in SIRT1 expression, in addition to the activation by NAD and AMPK, facilitates the deacetylation of a variety of proteins involved in mitochondrial beta-oxidation pathway and cell survival.


Asunto(s)
PPAR-beta/fisiología , Sirtuina 1/genética , Factor de Transcripción Sp1/fisiología , Transcripción Genética , Secuencia de Bases , Sitios de Unión , Células Cultivadas , Regulación de la Expresión Génica/efectos de los fármacos , Hormonas/farmacología , Humanos , Modelos Biológicos , Datos de Secuencia Molecular , PPAR delta/agonistas , PPAR delta/metabolismo , PPAR delta/fisiología , PPAR-beta/agonistas , PPAR-beta/metabolismo , Regiones Promotoras Genéticas , Unión Proteica , Sirtuina 1/metabolismo , Factor de Transcripción Sp1/metabolismo , Especificidad por Sustrato , Tiazoles/farmacología , Transcripción Genética/efectos de los fármacos
12.
Mol Cell Endocrinol ; 298(1-2): 25-32, 2009 Jan 27.
Artículo en Inglés | MEDLINE | ID: mdl-18955107

RESUMEN

Although insulin is indispensable for maintaining glucose homeostasis, it is still controversial whether or not a high concentration of insulin is deleterious. We examined the effect of insulin on the transcriptional activity of NF-kappaB, which mediates the expression of a variety of inflammation/coagulation-related genes using hepatocyte cell lines in vitro. We found that insulin (1 nM) alone caused minimal increase in NF-kappaB-mediated transcription. On the other hand, when cells were simultaneously treated with proinflammatory cytokines such as TNFalpha, the following dual effect of insulin was observed: short-term (6h) suppressive, and long-term (36 h or later) stimulatory effects. The former effect was transient and appears to be mediated by the phosphatidylinositol 3 kinase (PI(3)K) signaling pathway. The latter effect, in contrast, was more pronounced, enhancing the TNFalpha-stimulated NF-kappaB-dependent transcription by more than sevenfold. This positive effect was NF-kappaB-specific, and was eliminated by mitogen-activated protein kinase (MAPK) inhibitors. Altogether, our data suggest that insulin has short-term anti-inflammatory but long-term proinflammatory effects. From a clinical standpoint, this implies that low basal and periodically high plasma insulin is beneficial, whereas a sustained rise in plasma insulin, as often seen in patients with obesity, may induce atherothrombotic disorders, because of the NF-kappaB-mediated overexpression of proinflammatory/procoagulant/antifibrinolytic proteins in the liver.


Asunto(s)
Inflamación/inducido químicamente , Inflamación/prevención & control , Insulina/farmacología , Antiinflamatorios/farmacología , Humanos , Hipoglucemiantes/farmacología , Inflamación/genética , Mediadores de Inflamación/farmacología , Hígado/efectos de los fármacos , Hígado/metabolismo , Modelos Biológicos , FN-kappa B/genética , FN-kappa B/metabolismo , Receptor de Insulina/genética , Receptor de Insulina/metabolismo , Transducción de Señal/efectos de los fármacos , Transducción de Señal/genética , Factores de Tiempo , Transcripción Genética , Células Tumorales Cultivadas
13.
Mol Cell Endocrinol ; 302(1): 18-25, 2009 Apr 10.
Artículo en Inglés | MEDLINE | ID: mdl-19146914

RESUMEN

The mineralocorticoid receptor (MR) plays an important functional role in the central nervous system; however, the molecular mechanism of MR-dependent gene expression is not entirely clear. In this study, we examined the MR-dependent transcriptional regulation using a human neuronal cell line BE(2)C and an MR/GR-dependent reporter gene (HRE-luciferase) in vitro. Western blot analysis revealed that the cell line expresses MR but not glucocorticoid receptor (GR). In this experimental condition, unexpectedly, the MR-specific ligand aldosterone did not induce HRE-dependent transcription in a native or MR-overexpressed condition, whereas significant transcriptional induction by aldosterone was observed when the GR was co-expressed. The effect of aldosterone was completely inhibited by the MR antagonist spironolactone, indicating an MR-dependent effect. We found similar results in T84 colonic cells expressing neither MR nor GR, such that the aldosterone effect was obtained only when both receptors were co-expressed. The co-operative effect of GR was not obvious with the dimer-deficient mutant GR. Finally, the above findings were reproducible with different promoters containing HRE such as ENaC and MMTV. These results suggest that GR plays an indispensable role in MR-dependent transcription, possibly by forming a MR/GR heterodimer or by acting as a co-activator of MR/MR homodimer.


Asunto(s)
Regulación de la Expresión Génica , Receptores de Glucocorticoides/metabolismo , Receptores de Mineralocorticoides/metabolismo , Aldosterona/farmacología , Animales , Western Blotting , Células COS , Línea Celular Tumoral , Células Cultivadas , Chlorocebus aethiops , Dexametasona/farmacología , Dimerización , Eliminación de Gen , Regulación de la Expresión Génica/efectos de los fármacos , Glucocorticoides/farmacología , Células HeLa , Humanos , Receptores de Glucocorticoides/deficiencia , Receptores de Mineralocorticoides/agonistas , Receptores de Mineralocorticoides/genética
14.
Am J Nephrol ; 29(2): 102-8, 2009.
Artículo en Inglés | MEDLINE | ID: mdl-18689996

RESUMEN

BACKGROUND/AIMS: Proteinuria is not merely a marker of chronic nephropathies, but may also be involved in the progression to end-stage renal failure. We investigated the effect of angiotensin II type 1 receptor blockers (ARBs) on albumin-induced cell damage in human renal proximal tubular epithelial cells (RPTEC). METHODS: The N-acetyl-beta-D-glucosaminidase (NAG) and 8-hydroxy-2'-deoxyguanosine (8-OHdG) levels in the medium after albumin treatment with ARBs were determined by commercially available kits. The levels of p22(phox) protein in RPTEC were measured using Western blotting after albumin treatment with ARBs. Angiotensin II concentrations in cell media and cell lysates were assayed with a commercially available kit. RESULTS: Human albumin (0.1-10 mg/ml) dose-dependently increased NAG release and olmesartan or valsartan (10(-9)-10(-7) mol/l) showed a significant reduction on albumin (1 mg/ml)-induced NAG release in RPTEC. Albumin treatment (1 mg/ml) showed significant increases in p22(phox) protein levels in RPTEC and ARBs significantly decreased albumin-induced p22(phox) protein levels. Significant increases in 8-OHdG levels were observed in the albumin (1 mg/ml)-treated group and ARBs markedly reduced albumin-induced 8-OHdG levels in RPTEC. Human albumin dose-dependently increased angiotensin II concentrations in both cell media and lysates. CONCLUSION: These observations suggest renal tubular cell-protective properties of ARBs related to decreased oxidative stress during proteinuria.


Asunto(s)
Albuminuria/tratamiento farmacológico , Bloqueadores del Receptor Tipo 1 de Angiotensina II/farmacología , Imidazoles/farmacología , Túbulos Renales Proximales/efectos de los fármacos , Tetrazoles/farmacología , Valina/análogos & derivados , 8-Hidroxi-2'-Desoxicoguanosina , Acetilglucosaminidasa/metabolismo , Albúminas/farmacología , Albuminuria/metabolismo , Albuminuria/patología , Angiotensina II/metabolismo , Células Cultivadas , Desoxiguanosina/análogos & derivados , Desoxiguanosina/metabolismo , Relación Dosis-Respuesta a Droga , Humanos , Túbulos Renales Proximales/metabolismo , Túbulos Renales Proximales/patología , NADPH Oxidasas/metabolismo , Estrés Oxidativo/efectos de los fármacos , Valina/farmacología , Valsartán
15.
Endocr J ; 56(8): 1019-30, 2009.
Artículo en Inglés | MEDLINE | ID: mdl-19706989

RESUMEN

Both glucocorticoid and insulin are known to have an anabolic effect on lipogenesis. The glycolytic pathway is a part of the lipogenic pathway in the liver, and glycolytic enzymes mediate the conversion from glucose to pyruvate, and pyruvate dehydrogenase complex (PDC) mediates the conversion from pyruvate to acetyl-CoA, the activity of which is regulated by pyruvate dehydrogenase kinases (PDKs) and phosphatases (PDPs). In this study, we surveyed the effects of glucocorticoid, insulin, and forskolin (used as a surrogate of glucagon) on the transcriptional activity of glucokinase (GK), phosphofructokinase-1 (PFK1), liver-type pyruvate kinase (LPK), and all the PDKs/PDPs isoform genes. We found that both glucocorticoid and insulin had positive effects on PFK1 and LPK, whereas on GK the two hormones showed the opposite effect. Regarding the PDKs/PDPs, glucocorticoid significantly stimulated the transcriptional activity of all PDKs, among which the effect on PDK4 was the most prominent. Insulin alone had minimal effects on PDKs, but dampened the positive effects of glucocorticoid. On PDPs, glucocorticoid and forskolin showed negative effects, whereas insulin had positive effects; insulin and glucocorticoid/forskolin antagonized each other. Altogether, our data suggest that both glucocorticoid and insulin have lipogenic effects through positive effects on PFK1 and LPK expression. However, glucocorticoid antagonizes the effect of insulin at the level of GK to maintain glucose homeostasis and that of PDKs/PDPs to facilitate gluconeogenesis. Glucagon may also enhance gluconeogenesis by inhibiting PDPs.


Asunto(s)
Glucólisis/efectos de los fármacos , Glucólisis/genética , Hormonas/farmacología , Proteínas Serina-Treonina Quinasas/genética , Piruvato Deshidrogenasa (Lipoamida)-Fosfatasa/genética , Células Cultivadas , Colforsina/farmacología , Dexametasona/administración & dosificación , Dexametasona/farmacología , Relación Dosis-Respuesta a Droga , Combinación de Medicamentos , Enzimas/genética , Enzimas/metabolismo , Regulación Enzimológica de la Expresión Génica/efectos de los fármacos , Glucocorticoides/administración & dosificación , Glucocorticoides/farmacología , Gluconeogénesis/efectos de los fármacos , Gluconeogénesis/genética , Humanos , Insulina/administración & dosificación , Insulina/farmacología , Hígado/efectos de los fármacos , Hígado/metabolismo , Proteínas Serina-Treonina Quinasas/metabolismo , Piruvato Deshidrogenasa (Lipoamida)-Fosfatasa/metabolismo , Piruvato Deshidrogenasa Quinasa Acetil-Transferidora , Factores de Tiempo , Transcripción Genética/efectos de los fármacos
16.
Mol Cell Endocrinol ; 285(1-2): 10-8, 2008 Mar 26.
Artículo en Inglés | MEDLINE | ID: mdl-18313835

RESUMEN

Although glucocorticoid, as "gluco-" literally implies, plays an important role in maintaining the blood glucose level, excess of glucocorticoid production/action is known to cause impaired glucose tolerance and diabetes. Since 11beta-hydroxysteroid dehydrogenase type 1 (11beta-HSD1), which converts inactive cortisone to active cortisol, is primarily expressed in the liver, an enhanced expression of the enzyme may increase the intracellular glucocorticoid level and thus increase the hepatic glucose production. In this study, we examined the effects of multiple humoral factors related to the metabolic syndrome on the transcriptional activity of 11beta-HSD1 gene in hepatocytes in vitro. We found that, among the factors examined, adipocyte-derived cytokines (adipokines), like TNFalpha and IL-1beta, potently stimulated the transcriptional activity of 11beta-HSD1 gene in human HuH7 cells. In contrast, only minimal effects of other humoral factors were observed when they were used alone. Interestingly, however, when applied in combination, they synergistically enhanced the transcriptional activity of 11beta-HSD1 gene. They also potentiated the effects of cytokines. Glucocorticoid receptor (GR)-dependent transcription was indeed increased even with an inactive glucocorticoid cortisone following TNFalpha pretreatment, indicating the enhanced intracellular conversion. Finally, PPARgamma/PPARalpha agonists, clinically used as anti-diabetic drugs, significantly inhibited the transcriptional activity of 11beta-HSD1. Altogether, our data strongly suggest that combination of the humoral factors related to the metabolic syndrome, including the adipokines, synergistically enhances the hepatic expression of 11beta-HSD1 gene and causes the intracellular Cushing state in the liver by increasing the intracellular glucocorticoid level. We assume that the observed synergistic effects of these factors on 11beta-HSD1 may, at least partly, explain the reason whereby accumulation of the multiple risk factors facilitates the derangement of glucose and lipid metabolism in the metabolic syndrome.


Asunto(s)
11-beta-Hidroxiesteroide Deshidrogenasa de Tipo 1 , Síndrome de Cushing/sangre , Síndrome de Cushing/enzimología , Regulación Enzimológica de la Expresión Génica , Síndrome Metabólico/sangre , Síndrome Metabólico/enzimología , 11-beta-Hidroxiesteroide Deshidrogenasa de Tipo 1/genética , 11-beta-Hidroxiesteroide Deshidrogenasa de Tipo 1/metabolismo , Animales , Anticolesterolemiantes/metabolismo , Secuencia de Bases , Línea Celular , Cromanos/metabolismo , Clofibrato/metabolismo , Cortisona/metabolismo , Síndrome de Cushing/fisiopatología , Dexametasona/metabolismo , Glucocorticoides/metabolismo , Humanos , Hidrocortisona/metabolismo , Hipoglucemiantes/metabolismo , Insulina/metabolismo , Interleucina-1beta/metabolismo , Hígado/metabolismo , Síndrome Metabólico/fisiopatología , Metformina/metabolismo , Datos de Secuencia Molecular , Receptores de Glucocorticoides/genética , Receptores de Glucocorticoides/metabolismo , Tiazolidinedionas/metabolismo , Factor de Transcripción AP-1/metabolismo , Troglitazona , Factor de Necrosis Tumoral alfa/metabolismo
17.
Life Sci ; 82(23-24): 1137-41, 2008 Jun 06.
Artículo en Inglés | MEDLINE | ID: mdl-18468644

RESUMEN

Although excess of glucocorticoid causes neuronal damage with cognitive disorders, the molecular mechanism for this remains unclear. In this study, we examined the effect of adrenal corticosteroids on the transcription of NMDA glutamate receptor subunit genes and Alzheimer disease-related genes such as amyloid precursor protein (APP), beta-site amyloid precursor protein-cleaving enzyme 1 (BACE1), and presenilin using neuronal cell lines in vitro. We found that synthetic glucocorticoid dexamethasone (dex) potently increased the promoter activity of NMDA1 and 2A subunit genes, but did not stimulate those of Alzheimer disease-related genes. The similar effect of dex was observed on intrinsic NMDA1 mRNA and protein expression. Furthermore, dex showed synergistic and additive effects with protein kinase A- and C-mediated signaling pathways, respectively. Finally, treatment of the Neuro2A cells, which express intrinsic glucocorticoid receptor, with dex significantly enhanced the glutamate-induced neurotoxicity. Our results suggest that glucocorticoid-induced neuronal damage may be, at least partly, attributable to enhanced expression of glutamate NMDA receptor with a resultant increase in the susceptibility of glutamate-induced excitotoxicity rather than to a direct effect of the hormone to the Alzheimer disease-related genes.


Asunto(s)
Dexametasona/efectos adversos , Regulación de la Expresión Génica/efectos de los fármacos , Glucocorticoides/efectos adversos , Receptores de N-Metil-D-Aspartato/genética , Transcripción Genética/efectos de los fármacos , Enfermedad de Alzheimer/genética , Enfermedad de Alzheimer/metabolismo , Animales , Western Blotting , Línea Celular Tumoral , Clonación Molecular , Proteínas Quinasas Dependientes de AMP Cíclico/metabolismo , Sinergismo Farmacológico , Eliminación de Gen , Humanos , Plásmidos , Regiones Promotoras Genéticas , Proteína Quinasa C/metabolismo , Ratas , Elementos de Respuesta , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Proteína Amiloide A Sérica/metabolismo , Transducción de Señal/efectos de los fármacos , Acetato de Tetradecanoilforbol/farmacología
18.
Life Sci ; 83(11-12): 426-32, 2008 Sep 12.
Artículo en Inglés | MEDLINE | ID: mdl-18692075

RESUMEN

Glucocorticoid hormone is activated by 11beta-hydroxysteroid dehydrogenase type 1 (11beta-HSD-1) mainly in glucocorticoid-target organs such as the liver and the anterior corticotroph cells, and inactivated by type 2 (11beta-HSD-2) in mineralocorticoid-target cells such as renal and colonic epithelial cells. In this study, we examined the expression and action of these glucocorticoid-metabolizing enzymes in the A10 rat aortic smooth muscle cells (VSMC) in vitro. We found that both 11beta-HSD-1 and -2 mRNAs as well as glucocorticoid receptor (GR) and mineralocorticoid receptor (MR) were expressed in the cells. Interestingly, the transcriptional activity of 11beta-HSD-1 was stimulated by a representative proinflammatory cytokine TNFalpha, and inflammation-related inducible transcription factors AP1 and C/EBPs might have been at least partly responsible for the effect. In contrast, the transcriptional activity of 11beta-HSD-2 was decreased during the same stimuli, and another inflammation-induced transcription factor Egr-1 might have mediated the effect by interfering with the effect of Sp1, which maintains the basal expression of 11beta-HSD-2. The increase and decrease in 11beta-HSD-1 and 11beta-HSD-2 expression during inflammatory stimuli, respectively, were expected to cause the enhancement in glucocorticoid action, which was confirmed by the fact that TNFalpha elicited the cortisone-to-cortisol conversion using our bioassay system which employs the glucocorticoid-responsive reporter gene. Altogether, our results strongly suggest that inflammatory stress facilitates the intracellular glucocorticoid activation, i.e. conversion from inactive cortisone to active cortisol, by modifying the expression of both 11beta-HSD-1 and 11beta-HSD-2.


Asunto(s)
11-beta-Hidroxiesteroide Deshidrogenasa de Tipo 1/genética , 11-beta-Hidroxiesteroide Deshidrogenasa de Tipo 2/genética , Citocinas/farmacología , Regulación Enzimológica de la Expresión Génica/efectos de los fármacos , Regulación Enzimológica de la Expresión Génica/genética , Miocitos del Músculo Liso/enzimología , Secuencia de Bases , Western Blotting , Línea Celular , Genes Reporteros , Humanos , Luciferasas/genética , Datos de Secuencia Molecular , Miocitos del Músculo Liso/efectos de los fármacos , Plásmidos/genética , Regiones Promotoras Genéticas/genética , Receptores de Glucocorticoides/biosíntesis , Receptores de Glucocorticoides/genética , Receptores de Mineralocorticoides/biosíntesis , Receptores de Mineralocorticoides/genética , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Estimulación Química , Factor de Transcripción AP-1/genética , Transfección , Factor de Necrosis Tumoral alfa/farmacología
19.
Life Sci ; 82(13-14): 677-83, 2008 Mar 26.
Artículo en Inglés | MEDLINE | ID: mdl-18304585

RESUMEN

Although severe deficiency of bioactive vitamin D (1,25OH2D) causes rickets, mild insufficiency of the hormone, known as hypovitaminosis D, is responsible for the occurrence of secondary hyperparathyroidism and osteoporosis. To clarify the pathophysiology of the disease, we studied the negative feedback effect of 1,25OH2D and its precursor 25OHD on the transcriptional activity of parathyroid hormone (PTH) gene using the PT-r parathyroid cell line. We found that PT-r cells express endogenous 1alpha-hydroxylase as well as PTH mRNAs. We also found the potent suppressive effect of physiological concentration of 25OHD on the transcriptional activity of PTH gene. A similar effect was obtained with 1,25OH2D but only with pharmacological concentration. Interestingly, the effect of 25OHD was completely abolished when the cells were treated with 1alpha-hydroxylase inhibitor ketoconazole. These results suggest that the negative feedback regulation of vitamin D on PTH gene transcription occurs not by the end-product 1,25OH2D but by its prohormone 25OHD via intracellular activation by 1alpha-hydroxylase within the parathyroid cells.


Asunto(s)
Hormona Paratiroidea/genética , Transcripción Genética , Deficiencia de Vitamina D/genética , Vitamina D/análogos & derivados , Animales , Secuencia de Bases , Línea Celular , Clonación Molecular , Inhibidores Enzimáticos/farmacología , Retroalimentación Fisiológica , Vectores Genéticos , Humanos , Hiperparatiroidismo Secundario/genética , Cetoconazol/farmacología , Luciferasas/genética , Datos de Secuencia Molecular , Glándulas Paratiroides/citología , Glándulas Paratiroides/enzimología , Glándulas Paratiroides/metabolismo , Plásmidos , Regiones Promotoras Genéticas , Ratas , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Esteroide Hidroxilasas/antagonistas & inhibidores , Esteroide Hidroxilasas/fisiología , Transcripción Genética/efectos de los fármacos , Vitamina D/farmacología , Vitamina D/fisiología
20.
Endocr J ; 55(1): 217-23, 2008 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-18270430

RESUMEN

Erdheim-Chester disease (ECD) is a form of non-Langerhans histiocytosis. In this report, we show a case of ECD presenting diabetes insipidus and multiple xanthogranulomas received glucocorticoid treatment over a year. During this period, xanthogranulomas improved in response to the glucocorticoid therapy. Furthermore, the expression of osteopontin in xanthogranulomatous tissues significantly decreased following the treatment. Our data show the expression of osteopontin in xanthogranulomatous tissues of ECD. Furthermore, the osteopontin mRNA decreased following glucocorticoid therapy with xanthogranuloma regression, suggesting that the expression level of osteopontin could be a marker of the disease activity of ECD.


Asunto(s)
Enfermedad de Erdheim-Chester/tratamiento farmacológico , Enfermedad de Erdheim-Chester/genética , Granuloma/tratamiento farmacológico , Proteínas Asociadas a Microtúbulos/genética , Proteínas de Neoplasias/genética , Osteopontina/genética , Prednisolona/uso terapéutico , Xantomatosis/tratamiento farmacológico , Adulto , Femenino , Estudios de Seguimiento , Expresión Génica/efectos de los fármacos , Glucocorticoides/uso terapéutico , Granuloma/genética , Humanos , Proteínas Inhibidoras de la Apoptosis , Reacción en Cadena de la Polimerasa , Survivin , Xantomatosis/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA