Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 39
Filtrar
Más filtros

Medicinas Complementárias
Métodos Terapéuticos y Terapias MTCI
Bases de datos
Tipo del documento
Intervalo de año de publicación
1.
Appl Microbiol Biotechnol ; 103(6): 2665-2674, 2019 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-30734123

RESUMEN

7-Ketolithocholic acid (7-KLCA) is an important intermediate for the synthesis of ursodeoxycholic acid (UDCA). UDCA is the main effective component of bear bile powder that is used in traditional Chinese medicine for the treatment of human cholesterol gallstones. 7α-Hydroxysteroid dehydrogenase (7α-HSDH) is the key enzyme used in the industrial production of 7-KLCA. Unfortunately, the natural 7α-HSDHs reported have difficulty meeting the requirements of industrial application, due to their poor activities and strong substrate inhibition. In this study, a directed evolution strategy combined with high-throughput screening was applied to improve the catalytic efficiency and tolerance of high substrate concentrations of NADP+-dependent 7α-HSDH from Clostridium absonum. Compared with the wild type, the best mutant (7α-3) showed 5.5-fold higher specific activity and exhibited 10-fold higher and 14-fold higher catalytic efficiency toward chenodeoxycholic acid (CDCA) and NADP+, respectively. Moreover, 7α-3 also displayed significantly enhanced tolerance in the presence of high concentrations of substrate compared to the wild type. Owing to its improved catalytic efficiency and enhanced substrate tolerance, 7α-3 could efficiently biosynthesize 7-KLCA with a substrate loading of 100 mM, resulting in 99% yield of 7-KLCA at 2 h, in contrast to only 85% yield of 7-KLCA achieved for the wild type at 16 h.


Asunto(s)
Clostridium/enzimología , Evolución Molecular Dirigida , Hidroxiesteroide Deshidrogenasas/metabolismo , Ácido Litocólico/análogos & derivados , Clostridium/genética , Escherichia coli/genética , Ensayos Analíticos de Alto Rendimiento , Hidroxiesteroide Deshidrogenasas/genética , Cinética , Ácido Litocólico/biosíntesis , Simulación de Dinámica Molecular , Mutagénesis Sitio-Dirigida , Ácido Ursodesoxicólico/metabolismo
2.
Int J Biol Macromol ; 57: 22-5, 2013 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-23500436

RESUMEN

It was previously reported that an antitumor polysaccharide (PGPW1) was isolated from the root of Panax ginseng. To extend our study, we investigated here the anti-invasive and metastatic effects of PGPW1 on human gastric cancer cell line HGC-27 and tried to determine its possible mechanism of action. Both scratch wound-healing and Transwell assay identified that PGPW1 dose-dependently inhibited migration and invasiveness of HGC-27 cells. Furthermore, results of western blot showed that protein levels of Twist and AKR1C2 were inhibited by PGPW1, whereas an increase of NF1 was observed. Moreover, down-regulation of Twist expression by PGPW1 blocked epithelial-mesenchymal transition (EMT), characterized by a gain of epithelial cell markers, E-cadherin, and loss of the mesenchymal markers, vimentin and N-cadherin, at protein levels. Collectively, we confirmed that PGPW1 decreased migration and invasion of HGC-27 cells by regulation of Twist, AKR1C2, NF1, E-cadherin, vimentin and N-cadherin expression. In conclusion, PGPW1 may serve as a powerful chemopreventive agent against gastric cancer metastasis.


Asunto(s)
Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Proteínas Nucleares/biosíntesis , Panax/química , Polisacáridos/farmacología , Neoplasias Gástricas/metabolismo , Proteína 1 Relacionada con Twist/biosíntesis , Antígenos CD/biosíntesis , Antígenos CD/genética , Cadherinas/biosíntesis , Cadherinas/genética , Línea Celular Tumoral , Regulación hacia Abajo/efectos de los fármacos , Regulación hacia Abajo/genética , Transición Epitelial-Mesenquimal/efectos de los fármacos , Transición Epitelial-Mesenquimal/genética , Regulación Neoplásica de la Expresión Génica/genética , Humanos , Hidroxiesteroide Deshidrogenasas/biosíntesis , Hidroxiesteroide Deshidrogenasas/genética , Metástasis de la Neoplasia , Neurofibromina 1/biosíntesis , Neurofibromina 1/genética , Proteínas Nucleares/genética , Polisacáridos/química , Neoplasias Gástricas/genética , Neoplasias Gástricas/patología , Proteína 1 Relacionada con Twist/genética , Vimentina/biosíntesis , Vimentina/genética
3.
Zhonghua Nan Ke Xue ; 14(4): 371-6, 2008 Apr.
Artículo en Chino | MEDLINE | ID: mdl-18481435

RESUMEN

OBJECTIVE: To investigate the effects of Ginkgo biloba extract (EGB) on the testosterone synthesis in the Leydig cells of type 2 diabetic rats. METHODS: Thirty male SD rats were equally randomised into a normal control, a type 2 diabetic and an EGB group. Morphological changes of Leydig cells were observed by light microscopy (LM) and transmission electron microscopy (TEM), concentrations of serum luteinizing hormone (LH) and testosterone (T) were determined by enzyme linked immunosorbent assay (ELISA), and the mRNA levels in the steroidogenic acute regulatory protein (StAR), cytochrome P450 side chain cleavage (P450scc), cytochrome P450 17a-hydroxylase (P450c17), 17beta-hydroxysteroid dehydrogenase 3 (17beta-HSD3) and 3beta-hydroxysteroid dehydrogenase (3beta-HSD1) from the Leydig cells were examined by RT-PCR. RESULTS: Compared with the normal control, there was a significant decrease in the number and volume of Leydig cells, the levels of serum LH and T and the expression of mRNA in StAR, P450scc, 17beta-HSD3 and 3beta-HSD1 in the type 2 diabetes group. And the expression of the P450c17 gene showed a tendency of descending, but with no significance. Compared with the type 2 diabetes group, 12 weeks of EGB treatment caused very slight pathological changes in the Leydig cells, significantly increased the concentrations of blood LH and T, markedly elevated the levels of mRNA in StAR and P450scc and induced an ascending tendency of the expressions of P450c17, 17beta-HSD3 and 3beta-HSD1. CONCLUSION: EGB enhances testosterone synthesis and secretion of Leydig cells by reducing the impairment of the testis in type 2 diabetic rats.


Asunto(s)
Diabetes Mellitus Tipo 2/fisiopatología , Ginkgo biloba/química , Células Intersticiales del Testículo/efectos de los fármacos , Extractos Vegetales/farmacología , Testosterona/biosíntesis , 17-Hidroxiesteroide Deshidrogenasas/genética , Animales , Enzima de Desdoblamiento de la Cadena Lateral del Colesterol/genética , Diabetes Mellitus Tipo 2/sangre , Diabetes Mellitus Tipo 2/genética , Ensayo de Inmunoadsorción Enzimática , Expresión Génica/efectos de los fármacos , Hidroxiesteroide Deshidrogenasas/genética , Células Intersticiales del Testículo/metabolismo , Células Intersticiales del Testículo/ultraestructura , Hormona Luteinizante/sangre , Masculino , Microscopía Electrónica de Transmisión , Fosfoproteínas/genética , ARN Mensajero/genética , ARN Mensajero/metabolismo , Ratas , Ratas Sprague-Dawley , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Testosterona/sangre
4.
J Endocrinol ; 180(3): 457-67, 2004 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-15012600

RESUMEN

Estrogens are essential for bone mass accrual but their role before sexual maturation has remained elusive. Using in situ hybridization and immunohistochemistry, we investigated the expression of both estrogen receptor (ER) alpha and beta mRNA and protein as well as several mRNAs coding for enzymes involved in sex steroid metabolism (aromatase, type I and II 17 beta-hydroxysteroid dehydrogenase (17 beta-HSD), steroid sulfatase (STS) and type I 5 alpha-reductase) on sections of tibial metaphyses before (1- and 4-week-old), during (7-week-old) and after (16-week-old) sexual maturation in female and male rats. ER alpha and ER beta mRNA and protein were detected in metaphyseal bone in lining cells, osteoblasts, osteoclasts and some osteocytes with no apparent differences in expression during development or between the sexes. In contrast, aromatase, type I and II 17 beta-HSD and type I 5 alpha-reductase mRNAs were first detected in osteoblasts, osteoclasts and occasionally in osteocytes from sexual maturation (7-week-old rat) and onwards. Only STS was present before sexual maturation. To study the significance of ER alpha and beta expression in bone before sexual maturation when circulating sex steroid levels are low, 26-day-old female and male rats underwent gonadectomy or 17 beta-estradiol (E(2)) supplementation (0.5 mg/21 days) during 3 weeks. Following gonadectomy, trabecular bone volume (TBV) was lower in males (P=0.03) and there was a trend towards reduction in females (P=0.057). E(2) supplementation increased tibial TBV compared with controls in both genders as assessed by Masson-Goldner staining. These data suggest that the presence of ERs in bone cells before sex maturation might be of significance for bone mass accrual. Furthermore, based on the mRNA expression of the crucial enzymes aromatase and type I 17 beta-HSD, we suggest that bone cells in the tibial metaphysis acquire the intrinsic capacity to metabolize sex steroids from sexual maturation onwards. This process may contribute to the beneficial effects of estrogen on bone mass accrual, possibly by intracrinology.


Asunto(s)
Hormonas Esteroides Gonadales/metabolismo , Placa de Crecimiento/metabolismo , Receptores de Estrógenos/análisis , Maduración Sexual/fisiología , Tibia , 3-Oxo-5-alfa-Esteroide 4-Deshidrogenasa/análisis , Animales , Aromatasa/análisis , Aromatasa/genética , Estradiol/farmacología , Receptor alfa de Estrógeno , Receptor beta de Estrógeno , Femenino , Placa de Crecimiento/efectos de los fármacos , Hidroxiesteroide Deshidrogenasas/análisis , Hidroxiesteroide Deshidrogenasas/genética , Inmunohistoquímica/métodos , Hibridación in Situ/métodos , Masculino , Orquiectomía , ARN Mensajero/análisis , Ratas , Ratas Wistar , Receptores de Estrógenos/genética , Esteril-Sulfatasa/análisis , Esteril-Sulfatasa/genética
5.
Hypertension ; 42(2): 123-9, 2003 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-12860834

RESUMEN

Mutations in the gene encoding 11beta-hydroxysteroid dehydrogenase type 2, 11beta-HSD2 (HSD11B2), explain the molecular basis for the syndrome of apparent mineralocorticoid excess (AME), characterized by severe hypertension and hypokalemic alkalosis. Cortisol is the offending mineralocorticoid in AME, as the result of a lack of 11beta-HSD2-mediated cortisol to cortisone inactivation. In this study, we describe mutations in the HSD11B2 gene in 3 additional AME kindreds in which probands presented in adult life, with milder phenotypes including the original seminal case reported by Stewart and Edwards. Genetic analysis of the HSD11B2 gene revealed that all probands were compound heterozygotes, for a total of 7 novel coding and noncoding mutations. Of the 7 mutations detected, 6 were investigated for their effects on gene expression and enzyme activity by the use of mutant cDNA and minigene constructs transfected into HEK 293 cells. Four missense mutations resulted in enzymes with varying degrees of activity, all <10% of wild type. A further 2 mutations generated incorrectly spliced mRNA and predicted severely truncated, inactive enzyme. The mothers of 2 probands heterozygous for missense mutations have presented with a phenotype indistinguishable from "essential" hypertension. These genetic and biochemical data emphasize the heterogeneous nature of AME and the effects that heterozygosity at the HSD11B2 locus can have on blood pressure in later life.


Asunto(s)
Predisposición Genética a la Enfermedad , Hidroxiesteroide Deshidrogenasas/genética , Hipertensión/genética , Mutación , 11-beta-Hidroxiesteroide Deshidrogenasa de Tipo 2 , Adolescente , Adulto , Línea Celular , ADN Complementario/metabolismo , Femenino , Expresión Génica , Heterocigoto , Humanos , Hidroxiesteroide Deshidrogenasas/metabolismo , Hipertensión/diagnóstico , Hipopotasemia/diagnóstico , Masculino , Mineralocorticoides/metabolismo , Linaje , Fenotipo
6.
Am J Respir Crit Care Med ; 167(9): 1244-9, 2003 May 01.
Artículo en Inglés | MEDLINE | ID: mdl-12574078

RESUMEN

The actions of natural and synthetic glucocorticoids are in part determined by 11beta-hydroxysteroid dehydrogenase type 2 (11beta-HSD2). We examined whether carbenoxolone, a potent inhibitor of 11beta-HSD, would potentiate the inhibitory action of dexamethasone on interleukin-8 release from BEAS-2B cells, and whether prolonged treatment with dexamethasone at therapeutic doses would upregulate 11beta-HSD2 in the cells. We found that carbenoxolone increased the potency of dexamethasone almost 10-fold. Reverse transcription-polymerase chain reaction and Western blot revealed that BEAS-2B cells expressed 11beta-HSD2, but not 11beta-HSD1. An enzyme activity assay of the cell homogenate demonstrated only NAD+-dependent dehydrogenase activity. The Km value for cortisol in intact BEAS-2B cells was estimated to be 42 nM. When the cells were incubated with dexamethasone for up to 72 hours at increasing concentrations (10(-9) to 10(-5) M), there were considerable increases in mRNA and protein levels of 11beta-HSD2. Prolonged treatment with dexamethasone also increased the enzyme activity of 11beta-HSD in the cells in a dose- and time-dependent manner, with complete inhibition by RU38486. These results suggest that bronchial epithelial cells possess an autoregulatory system for glucocorticoids in the control of their own bioactive levels by inducing the expression of 11beta-HSD2, and that 11beta-HSD2 in the bronchial epithelium may play a role in the local regulation of inhaled glucocorticoid actions.


Asunto(s)
Antiinflamatorios/farmacología , Bronquios/efectos de los fármacos , Bronquios/enzimología , Dexametasona/farmacología , Hidroxiesteroide Deshidrogenasas/efectos de los fármacos , Mucosa Respiratoria/efectos de los fármacos , Mucosa Respiratoria/enzimología , Regulación hacia Arriba/efectos de los fármacos , 11-beta-Hidroxiesteroide Deshidrogenasa de Tipo 2 , Antiinflamatorios/farmacocinética , Disponibilidad Biológica , Western Blotting , Bronquios/citología , Carbenoxolona/farmacología , Línea Celular , Dexametasona/farmacocinética , Relación Dosis-Respuesta a Droga , Evaluación Preclínica de Medicamentos , Sinergismo Farmacológico , Humanos , Hidroxiesteroide Deshidrogenasas/antagonistas & inhibidores , Hidroxiesteroide Deshidrogenasas/genética , Inflamación , Mucosa Respiratoria/citología , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa
7.
Mol Pharmacol ; 63(3): 722-31, 2003 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-12606783

RESUMEN

Dehydroepiandrosterone (DHEA) is a C-19 adrenal steroid precursor to the gonadal steroids. In humans, circulating levels of DHEA, as its sulfated conjugate, are high at puberty and throughout early adulthood but decline with age. Dietary supplementation to maintain high levels of DHEA purportedly has beneficial effects on cognitive memory, the immune system, and fat and carbohydrate metabolism. In rodents, DHEA is a peroxisome proliferator that induces genes for the classical peroxisomal and microsomal enzymes associated with this response. These effects are mediated through activation of peroxisome proliferator-activated receptor alpha (PPAR alpha). However, DHEA can affect the expression of genes independently of PPAR alpha, including the gene for the major inducible drug and xenobiotic metabolizing enzyme, cytochrome P450 3A23. To elucidate the biochemistry associated with DHEA treatment, we employed a cDNA gene expression array using liver RNA from rats treated with DHEA or the classic peroxisome proliferator nafenopin. Principal components analysis identified 30 to 35 genes whose expression was affected by DHEA and/or nafenopin. Some were genes previously identified as PPAR-responsive genes. Changes in expression of several affected genes were verified by quantitative reverse transcriptase-polymerase chain reaction. These included aquaporin 3, which was induced by DHEA and to a lesser extent nafenopin, nuclear tyrosine phosphatase, which was induced by both agents, and 11 beta-hydroxysteroid dehydrogenase 1, which was decreased by treatment with DHEA in a dose-dependent fashion. Regulation of 11 beta-hydroxysteroid dehydrogenase 1 expression is important since the enzyme is believed to amplify local glucocorticoid signaling, and its repression may cause some of the metabolic effects associated with DHEA.


Asunto(s)
Deshidroepiandrosterona/farmacología , Expresión Génica/efectos de los fármacos , Hidroxiesteroide Deshidrogenasas/biosíntesis , Hígado/efectos de los fármacos , 11-beta-Hidroxiesteroide Deshidrogenasas , Animales , Acuaporina 3 , Acuaporinas/biosíntesis , Acuaporinas/genética , Perfilación de la Expresión Génica , Hidroxiesteroide Deshidrogenasas/genética , Hipolipemiantes/farmacología , Hígado/enzimología , Masculino , Nafenopina/farmacología , Análisis de Secuencia por Matrices de Oligonucleótidos , Ratas , Ratas Sprague-Dawley , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa
8.
Am J Physiol Endocrinol Metab ; 282(2): E466-73, 2002 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-11788380

RESUMEN

To evaluate the effects of altered corticosteroid metabolism on the hypothalamic-pituitary-adrenal axis, we examined rats treated with glycyrrhizic acid (G rats) or rifampicin (R rats) for 7 days. The half-life of exogenously administered hydrocortisone as a substitute for corticosterone was longer in G rats and shorter in R rats, with no differences in basal plasma levels of ACTH or corticosterone. The ACTH responses to human corticotropin-releasing factor (CRF) or insulin-induced hypoglycemia were greater in G rats and tended to be smaller in R rats compared with those in the control rats, whereas the corticosterone response was similar. No difference was observed in the content and mRNA level of hypothalamic CRF among the groups. The number and mRNA level of CRF receptor and type 1 11 beta-hydroxysteroid dehydrogenase (11-HSD1) mRNA level in the pituitary were increased in G rats but not changed in R rats, suggesting that chronically increased intrapituitary corticosterone upregulates pituitary CRF receptor expression. In contrast, CRF mRNA levels in the pituitary were increased in R rats. Our data indicate novel mechanisms of corticosteroid metabolic modulation and the involvement of pituitary 11-HSD1 and CRF in glucocorticoid feedback physiology.


Asunto(s)
Corticoesteroides/metabolismo , Hormona Adrenocorticotrópica/metabolismo , Hipófisis/metabolismo , 11-beta-Hidroxiesteroide Deshidrogenasas , Hormona Adrenocorticotrópica/sangre , Animales , Corticosterona/sangre , Hormona Liberadora de Corticotropina/genética , Hormona Liberadora de Corticotropina/farmacología , Ácido Glicirrínico/farmacología , Semivida , Humanos , Hidrocortisona/farmacocinética , Hidroxiesteroide Deshidrogenasas/genética , Hipoglucemia/sangre , Hipoglucemia/inducido químicamente , Hipotálamo/metabolismo , Insulina , Masculino , ARN Mensajero/metabolismo , Ratas , Ratas Wistar , Receptores de Hormona Liberadora de Corticotropina/genética , Rifampin/farmacología
9.
J Biol Chem ; 276(47): 44083-90, 2001 Nov 23.
Artículo en Inglés | MEDLINE | ID: mdl-11562362

RESUMEN

Metabolic activation of retinol (vitamin A) via sequential actions of retinol and retinal dehydrogenases produces the active metabolite all-trans-retinoic acid. This work reports cDNA cloning, enzymatic characterization, function in a reconstituted path of all-trans-retinoic acid biosynthesis in cell culture, and mRNA expression patterns in adult tissues and embryos of a mouse retinol dehydrogenase, RDH1. RDH1 represents a new member of the short chain dehydrogenase/reductase superfamily that differs from other mouse RDH in relative activity with all-trans and cis-retinols. RDH1 has a multifunctional catalytic nature, as do other short chain dehydrogenase/reductases. In addition to retinol dehydrogenase activity, RDH1 has strong 3alpha-hydroxy and weak 17beta-hydroxy steroid dehydrogenase activities. RDH1 has widespread and intense mRNA expression in tissues of embryonic and adult mice. The mouse embryo expresses RDH1 as early as 7.0 days post-coitus, and expression is especially intense within the neural tube, gut, and neural crest at embryo day 10.5. Cells cotransfected with RDH1 and any one of three retinal dehydrogenase isozymes synthesize all-trans-retinoic acid from retinol, demonstrating that RDH1contributes to a path of all-trans-retinoic acid biosynthesis in intact cells. These characteristics are consistent with RDH1 functioning in a path of all-trans-retinoic acid biosynthesis starting early during embryogenesis.


Asunto(s)
Oxidorreductasas de Alcohol/metabolismo , Hidroxiesteroide Deshidrogenasas/metabolismo , Vitamina A/metabolismo , Oxidorreductasas de Alcohol/química , Oxidorreductasas de Alcohol/genética , Secuencia de Aminoácidos , Animales , Secuencia de Bases , Línea Celular , Clonación Molecular , ADN Complementario , Embrión de Mamíferos/metabolismo , Hidroxiesteroide Deshidrogenasas/química , Hidroxiesteroide Deshidrogenasas/genética , Hibridación in Situ , Cinética , Ratones , Datos de Secuencia Molecular , ARN Mensajero/genética , ARN Mensajero/metabolismo , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa
10.
Endocrinology ; 142(3): 1341-8, 2001 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-11181553

RESUMEN

The NAD-dependent enzyme, 11beta-hydroxysteroid dehydrogenase type II (11 beta HSD2), catalyzes the unidirectional conversion of biologically active glucocorticoids to inactive metabolites. In vivo, 11 beta HSD2 protects the mineralocorticoid receptor from activation by glucocorticoids in mineralocorticoid target tissues such as kidney. The goal of the present study was to use targeted overexpression of 11 beta HSD2 as a novel means of disrupting glucocorticoid signaling in osteoblastic cells. Rat 11 beta HSD2 complementary DNA was cloned downstream of a 2.3- and 3.6-kb alpha 1(I)-collagen (Col1a1) promoter fragment to produce the expression plasmids Col2.3-HSD2 and Col3.6-HSD2, respectively, which were transiently and/or stably transfected in osteoblastic ROS 17/2.8 and MC3T3-E1 cells. Transgene messenger RNA and protein were detected in transfected cells by Northern blot analysis and immunostaining, respectively. Transfection of 11 beta HSD2 led to higher rates of conversion of [(3)H]corticosterone to [(3)H]dehydrocorticosterone and reduced glucocorticoid-dependent regulation of a mouse mammary tumor virus promoter-reporter construct, cell growth, and messenger RNA markers compared with transfection of a control vector. Expression of 11 beta HSD2 under the control of Col1a1 promoter fragments may provide a novel model to study the role of glucocorticoid signaling in osteoblastic cells.


Asunto(s)
Clonación Molecular , Colágeno Tipo I , Colágeno/genética , Corticosterona/análogos & derivados , Glucocorticoides/fisiología , Hidroxiesteroide Deshidrogenasas/genética , Osteoblastos/metabolismo , Transgenes , 11-beta-Hidroxiesteroide Deshidrogenasas , Animales , División Celular/efectos de los fármacos , División Celular/fisiología , Línea Celular , Cloranfenicol O-Acetiltransferasa/genética , Cadena alfa 1 del Colágeno Tipo I , Corticosterona/metabolismo , Corticosterona/farmacología , Dexametasona/farmacología , Regulación de la Expresión Génica/fisiología , Glucocorticoides/farmacología , Hidrocortisona/farmacología , Hidroxiesteroide Deshidrogenasas/metabolismo , Virus del Tumor Mamario del Ratón/genética , Osteoblastos/citología , Regiones Promotoras Genéticas/fisiología , ARN Mensajero/metabolismo , Ratas , Transfección
11.
Kidney Int ; 57(4): 1352-7, 2000 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-10760066

RESUMEN

Aldosterone selectivity in mineralocorticoid target tissues is mainly due to 11beta-hydroxysteroid dehydrogenase (11betaHSD), which converts cortisol to its inactive metabolite cortisone in humans. The defect of dehydrogenase activity would thus allow type 1 mineralocorticoid receptor (MR) to be occupied mostly by cortisol. It has been postulated that 11betaHSD type 2 (11betaHSD2) plays a significant role in conferring ligand specificity on the MR. We have demonstrated the diminished dehydrogenase activity in resistance vessels of genetically hypertensive rats. However, the mechanism that could link impaired vascular 11betaHSD activity and elevated blood pressure has been unclear. In this study, we showed the enzyme activity in human coronary artery smooth muscle cells. Glucocorticoids and mineralocorticoids increase vascular tone by up-regulating the receptors of pressor hormones such as angiotensin II (Ang II). Next, we found that physiological concentrations of a cortisol-induced increase in Ang II binding were significantly enhanced by the inhibition of dehydrogenase activity with an antisense DNA complementary to 11betaHSD2 mRNA, and the enhancement was partially but significantly abolished by a selective aldosterone receptor antagonist. This may indicate that impaired dehydrogenase activity in vascular wall results in increased vascular tone by the contribution of cortisol, which acts as a mineralocorticoid. In congenital 11betaHSD deficiency and after the administration of 11betaHSD inhibitors, suppression of dehydrogenase activity in the kidney has been believed to cause renal mineralocorticoid excess, resulting in sodium retention and hypertension. These results show that vascular 11betaHSD activity could influence blood pressure without invoking renal sodium retention.


Asunto(s)
Vasos Coronarios/enzimología , Hidroxiesteroide Deshidrogenasas/metabolismo , 11-beta-Hidroxiesteroide Deshidrogenasas , Angiotensina II/metabolismo , Células Cultivadas , Vasos Coronarios/citología , Vasos Coronarios/metabolismo , Humanos , Hidrocortisona/farmacología , Hidroxiesteroide Deshidrogenasas/antagonistas & inhibidores , Hidroxiesteroide Deshidrogenasas/genética , Músculo Liso Vascular/citología , Músculo Liso Vascular/enzimología , Músculo Liso Vascular/metabolismo , Oligonucleótidos Antisentido/farmacología
12.
J Steroid Biochem Mol Biol ; 69(1-6): 403-8, 1999.
Artículo en Inglés | MEDLINE | ID: mdl-10419018

RESUMEN

The type 2 isozyme of 11beta-hydroxysteroid dehydrogenase inactivates cortisol to cortisone and enables aldosterone to bind to the MR. Congenital deficiency of the enzyme results in cortisol-mediated mineralocorticoid excess and arises because of inactivating mutations in the HSD11B2 gene. Inhibition of the enzyme following licorice or carbenoxolone ingestion results in a similar, though milder phenotype and the enzyme is overwhelmed in ectopic ACTH syndrome. Loss of 11beta-HSD2 expression may be important in sodium balance and blood pressure control in some patients with renal disease. Finally, while some studies demonstrate impaired 11beta-HSD activity in broader populations of patients with hypertension, further studies are required to clarify the role of 11beta-HSD2 in 'essential' hypertension.


Asunto(s)
Hidrocortisona/fisiología , Mineralocorticoides/fisiología , 11-beta-Hidroxiesteroide Deshidrogenasas , Síndrome de Cushing/enzimología , Síndrome de Cushing/fisiopatología , Glycyrrhiza , Humanos , Hidroxiesteroide Deshidrogenasas/genética , Hidroxiesteroide Deshidrogenasas/metabolismo , Hipertensión/enzimología , Hipertensión/metabolismo , Hipertensión/fisiopatología , Enfermedades Renales/enzimología , Enfermedades Renales/metabolismo , Enfermedades Renales/fisiopatología , Plantas Medicinales
13.
Hypertension ; 33(5): 1179-84, 1999 May.
Artículo en Inglés | MEDLINE | ID: mdl-10334808

RESUMEN

11beta-Hydroxysteroid dehydrogenases (11beta-HSD) interconvert cortisol, the physiological glucocorticoid, and its inactive metabolite cortisone in humans. The diminished dehydrogenase activity (cortisol to cortisone) has been demonstrated in patients with essential hypertension and in resistance vessels of genetically hypertensive rats. 11beta-Hydroxysteroid dehydrogenase type 2 (11beta-HSD2) catalyzes only 11beta-dehydrogenation. However, a functional relationship between diminished vascular 11beta-HSD2 activity and elevated blood pressure has been unclear. In this study we showed the expression and enzyme activity of 11beta-HSD2 and 11beta-HSD type 1 (which is mainly oxoreductase, converting cortisone to cortisol) in human vascular smooth muscle cells. Glucocorticoids and mineralocorticoids increase vascular tone by upregulating the receptors of pressor hormones such as angiotensin II. We found that physiological concentrations of cortisol-induced increase in angiotensin II binding were significantly enhanced by the inhibition of 11beta-HSD2 activity with an antisense DNA complementary to 11beta-HSD2 mRNA, and the enhancement was partially but significantly abolished by a selective aldosterone receptor antagonist. This may indicate that impaired 11beta-HSD2 activity in vascular wall results in increased vascular tone by the contribution of cortisol, which acts as a mineralocorticoid. In congenital 11beta-HSD deficiency and after administration of 11beta-HSD inhibitors, suppression of 11beta-HSD2 activity in the kidney has been believed to cause renal mineralocorticoid excess, resulting in sodium retention and hypertension. In the present study we provide evidence for a mechanism that could link impaired vascular 11beta-HSD2 activity, increased vascular tone, and elevated blood pressure without invoking renal sodium retention.


Asunto(s)
Corticosterona , Hidroxiesteroide Deshidrogenasas/fisiología , Hipertensión/etiología , 11-beta-Hidroxiesteroide Deshidrogenasas , Angiotensina II/fisiología , Secuencia de Bases , Células Cultivadas , Cromatografía en Capa Delgada , Vasos Coronarios , Corticosterona/metabolismo , Cartilla de ADN , Expresión Génica , Humanos , Hidrocortisona/análisis , Hidrocortisona/fisiología , Hidroxiesteroide Deshidrogenasas/análisis , Hidroxiesteroide Deshidrogenasas/genética , Hipertensión/fisiopatología , Datos de Secuencia Molecular , Tono Muscular , Músculo Liso Vascular/citología , Músculo Liso Vascular/enzimología , ARN Mensajero/genética , Receptores de Angiotensina/fisiología
14.
J Clin Endocrinol Metab ; 83(12): 4490-7, 1998 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-9851798

RESUMEN

In adult life, the type 2 isozyme of 11beta-hydroxysteroid dehydrogenase (11betaHSD2) protects the mineralocorticoid receptor (MR) from glucocorticoid by inactivating cortisol to cortisone. 11betaHSD2 activity has been reported in human fetal tissues, where glucocorticoids may impair fetal growth yet are also required for normal fetal development. Using digoxigenin-labeled complementary ribonucleic acid (RNA) probes and an in-house 11betaHSD2 antiserum, we have analyzed the expression of 11betaHSD2, MR, and glucocorticoid receptor (GR) in human fetal tissues of gestational age 6-17 weeks (n=15). 11BetaHSD2 expression was absent at gestational age 6+ weeks, but was expressed in abundance in many fetal tissues between 8-12 weeks. At this time, 11betaHSD2 colocalized with GR messenger RNA (mRNA) expression in metanephros, gut, muscle, spinal cord and dorsal root ganglia, periderm, sex chords of testis, and adrenal. In particular within fetal kidney, intense expression of 11betaHSD2 and GR mRNA was observed over Bowman's capsule and the vascular tufts of developing glomeruli as they migrated from the surface of the kidney to the inner cortex. Only lung and adrenal medullary rests demonstrated high levels of GR mRNA but low levels of 11betaHSD2. 11BetaHSD2 mRNA and immunoreactivity staining patterns were similar, with the exception of the fetal adrenal, where mRNA was localized to the outer definitive zone but immunoreactivity was localized to the inner fetal zone. Colocalization of 11betaHSD2 (and GR mRNA) with MR mRNA was observed principally within epithelial cells of collecting ducts, particularly after 16 weeks gestation when the pattern of distribution of 11betaHSD2 became more adult in nature. High levels of MR mRNA were observed within developing bone. The data indicate that 11betaHSD2 in fetal life principally modulates ligand access to the GR in most fetal tissues, notably glomeruli and tubules in the developing kidney, testis, and periderm, and this may be have ramifications for fetal sodium homeostasis and differentiation. The development of tissues previously shown to have a critical requirement for glucocorticoids, such as lung and adrenal medulla, is facilitated by the expression of GR mRNA, but not 11betaHSD2. The expression of MR mRNA in high abundance in bone suggests a role for corticosteroids in human bone development, and the low/absent expression of 11betaHSD2 at this site suggests that it is functionally acting as a GR.


Asunto(s)
Feto/metabolismo , Hidroxiesteroide Deshidrogenasas/metabolismo , Isoenzimas/metabolismo , Receptores de Glucocorticoides/metabolismo , Receptores de Mineralocorticoides/metabolismo , 11-beta-Hidroxiesteroide Deshidrogenasas , Femenino , Edad Gestacional , Humanos , Hidroxiesteroide Deshidrogenasas/genética , Inmunohistoquímica , Hibridación in Situ , Isoenzimas/genética , Embarazo , Primer Trimestre del Embarazo , ARN Mensajero/metabolismo , Receptores de Mineralocorticoides/genética , Distribución Tisular
15.
Endocr Res ; 24(3-4): 521-9, 1998.
Artículo en Inglés | MEDLINE | ID: mdl-9888533

RESUMEN

Members of the NADPH-dependent short chain dehydrogenase/reductase (SDR) family control blood pressure, fertility, and natural and neoplastic growth. Despite the fact that only one amino acid residue is strictly conserved in the 100 known members of the family, all appear to have a dinucleotide-binding Rossmann fold and homologous catalytic residues including the conserved tyrosine. Variation in the binding pocket creates specificity for steroids, prostaglandins, sugars and alcohols. The critically important tyrosine appears to maintain a fixed position relative to the scaffolding of the Rossmann fold and the cofactor position, while the substrate-binding pocket alters in such a way that the dehydrogenation/reduction reaction site is brought into bonding distance of the tyrosine hydroxyl group. Licorice induces high blood pressure by inhibiting an SDR in the kidney. The crystal structure of the complex of 3alpha,20beta-hydroxysteroid dehydrogenase and carbenoxolone reveals the mechanism of enzyme inhibition by licorice. The most potent dehydrogenase enzyme inhibitors are those that displace substrate and cofactor and form strong hydrogen bonds to one or more amino acid residues involved in catalysis.


Asunto(s)
Hidroxiesteroide Deshidrogenasas/antagonistas & inhibidores , Hipertensión/enzimología , 11-beta-Hidroxiesteroide Deshidrogenasas , 20-Hidroxiesteroide Deshidrogenasas/genética , 20-Hidroxiesteroide Deshidrogenasas/metabolismo , Carbenoxolona/farmacología , Glycyrrhiza , Ácido Glicirrínico/farmacología , Humanos , Hidroxiesteroide Deshidrogenasas/química , Hidroxiesteroide Deshidrogenasas/genética , Conformación Molecular , Mapeo Peptídico , Plantas Medicinales
16.
Endocrinology ; 138(8): 3532-9, 1997 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-9231808

RESUMEN

Human 17beta-hydroxysteroid dehydrogenase (17-HSD) type 1 predominantly catalyzes the 17beta-reduction of estrone to estradiol. The present results, however, show that rat 17-HSD type 1 equally uses both estrone and androstenedione as substrates. Analyzing the activity of various rat/human chimeric enzymes indicated that the region between amino acids 148 and 268 is responsible for the difference in substrate specificity, which is in line with the structural data showing that the recognition end of the active site is primarily at residues 185-230. The enzymes are highly conserved between amino acids 148-191, and the data indicate that in this region Asn152HisAsp153Glu and Pro187Ala variations are most closely related to the differential steroid specificity. The structural analyses furthermore suggested that the presence of His instead of Asn at position 152 of the human enzyme might result in considerable rearrangement of the loop located close to the beta-face of the A- and B-rings of the bound substrate, and that the Pro187Ala variation could modify the flexible region involved in substrate recognition and access of the substrate to the active site. Altogether, our results indicate that the Asn152His and Pro187Ala variations, together with several amino acid variations at the recognition end of the catalytic cleft built by residues 190-230, alter the structure of the active site of rat 17-HSD type 1 to one more favorable to an androgenic substrate.


Asunto(s)
Hidroxiesteroide Deshidrogenasas/metabolismo , Proteínas Recombinantes de Fusión/metabolismo , Secuencia de Aminoácidos , Aminoácidos/análisis , Animales , Secuencia de Bases , Línea Celular , Sondas de ADN/análisis , Sondas de ADN/química , Sondas de ADN/genética , ADN Complementario/análisis , ADN Complementario/química , ADN Complementario/genética , Estradiol/metabolismo , Estrona/metabolismo , Humanos , Hidroxiesteroide Deshidrogenasas/química , Hidroxiesteroide Deshidrogenasas/genética , Datos de Secuencia Molecular , Mutagénesis Sitio-Dirigida , Reacción en Cadena de la Polimerasa , Ratas , Proteínas Recombinantes de Fusión/química , Proteínas Recombinantes de Fusión/genética , Spodoptera/citología , Especificidad por Sustrato , Factores de Tiempo
17.
Endocr Rev ; 18(1): 135-56, 1997 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-9034789

RESUMEN

Whereas aldosterone is normally a much stronger mineralocorticoid than cortisol in vivo, mineralocorticoid receptors have identical in vitro affinities for these hormones. The in vivo specificity of the receptors is, at least in part, the result of activity of 11-HSD, an enzyme located in most mineralocorticoid target tissues that converts cortisol to cortisone. Cortisone is not a ligand for the receptor, whereas aldosterone is not a substrate of the enzyme. The syndrome of AME is a rare form of juvenile hypertension in which 11-HSD is defective. This deficiency allows mineralocorticoid receptors to be occupied by cortisol, leading to hypertension, because plasma concentrations of cortisol are much higher than those of aldosterone. Licorice, which contains 11-HSD inhibitors, causes a similar syndrome. There are two known isozymes of 11-HSD. The liver or type I isozyme is expressed at high levels in the liver, has a relatively low affinity for steroids (micromolar Km), catalyzes both dehydrogenation and the reverse reductase reaction, and utilizes NADP+ or NADPH as cofactors. The kidney or type 2 isozyme is expressed at high levels in the kidney and placenta, has a high affinity (nanomolar Km) for steroids, catalyzes only dehydrogenation, and utilizes NAD+ as a cofactor. Mutations in the HSD11B2 (HSD11K) gene encoding the kidney isozyme of 11-HSD have been detected in all kindreds with AME studied thus far. This gene represents a candidate locus for the common, "essential" form of hypertension.


Asunto(s)
Hidroxiesteroide Deshidrogenasas/metabolismo , Hipertensión/enzimología , Isoenzimas/metabolismo , Mineralocorticoides/metabolismo , Receptores de Mineralocorticoides/fisiología , 11-beta-Hidroxiesteroide Deshidrogenasas , Síndrome de ACTH Ectópico/metabolismo , Aldosterona/metabolismo , Glycyrrhiza , Humanos , Hidroxiesteroide Deshidrogenasas/genética , Hipertensión/genética , Hipertensión/fisiopatología , Isoenzimas/genética , Mutación , Plantas Medicinales , Síndrome
18.
Steroids ; 62(1): 83-8, 1997 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-9029720

RESUMEN

The syndrome of apparent mineralocorticoid excess (AME) is an inherited form of hypertension in which 11 beta-hydroxysteroid dehydrogenase (11-HSD) is defective. This enzyme converts cortisol to its inactive metabolite, cortisone. The deficiency allows mineralocorticoid receptors to be occupied by cortisol, because these receptors themselves have similar affinities for cortisol and aldosterone. There are two isozymes of 11-HSD, a liver (L) or type 1 isozyme with a relatively low affinity for steroids, and a kidney (K) or type 2 isozyme with high steroid affinity. Mutations in the gene for the kidney isozyme of 11-HSD have been detected in all kindreds with AME. We expressed enzymes carrying all known missense mutations in cultured cells and determined their activity. For each patient with AME, we compared the enzymatic activity predicted by the genotype with the ratio of cortisol to cortisone metabolites in the urine, (THF + aTHF)/THE. These were strongly correlated, suggesting that the biochemical phenotype of AME is largely determined by genotype. The K isozyme of 11-HSD is also expressed in high levels in the placenta, where its function is unclear. AME patients often have low birth weight. By analogy with AME, low placental 11-HSD K activity in humans might be a risk factor for low birth weight and subsequent hypertension. However, we found that there was no significant correlation between 11-HSD activity, mRNA levels, and either fetal or placental weight.


Asunto(s)
Hidroxiesteroide Deshidrogenasas/genética , Hidroxiesteroide Deshidrogenasas/metabolismo , Hipertensión/metabolismo , Mineralocorticoides/metabolismo , 11-beta-Hidroxiesteroide Deshidrogenasas , Alelos , Animales , Clonación Molecular , ADN Complementario/genética , Femenino , Humanos , Hidroxiesteroide Deshidrogenasas/deficiencia , Hipertensión/genética , Mutación , Placenta/enzimología , Embarazo , Ratas , Síndrome , Transcripción Genética
19.
Pediatr Res ; 41(1): 25-9, 1997 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-8979285

RESUMEN

Aldosterone, the most important mineralocorticoid, regulates electrolyte excretion and intravascular volume mainly through its effects on renal distal tubules and cortical collecting ducts, where it acts to increase sodium resorption from and potassium excretion into the urine. Excess secretion of aldosterone or other mineralocorticoids, or abnormal sensitivity to mineralocorticoids, may results in hypokalemia, suppressed plasma renin activity, and hypertension. The syndrome of apparent mineralocorticoid excess (AME) is an inherited form of hypertension in which 11 beta-hydroxysteroid dehydrogenase (11 beta-HSD) is defective. This enzyme converts cortisol to its inactive metabolite, cortisone. Because mineralocorticoid receptors themselves have similar affinities for cortisol and aldosterone, it is hypothesized that the deficiency allows these receptors to be occupied by cortisol, which normally circulates at levels far higher than those of aldosterone. We cloned cDNA and genes encoding two isozymes of 11 beta-HSD. The liver (L) or type 1 isozyme has relatively low affinity for steroids, is expressed at high levels in the liver but poorly in the kidney, and is not defective in AME. The kidney (K) or type 2 isozyme has high steroid affinity and is expressed at high levels in the kidney and placenta. Mutations in the gene for the latter isozyme have been detected in all kindreds with AME. Moreover, the in vitro enzymatic activity conferred by each mutation is strongly correlated with the ratio of cortisol to cortisone metabolites in the urine [tetrahydrocortisone (THF) +allo-THF]/THE. This suggests that the biochemical phenotype of AME is largely determined by genotype.


Asunto(s)
Hidroxiesteroide Deshidrogenasas/metabolismo , Mineralocorticoides/metabolismo , 11-beta-Hidroxiesteroide Deshidrogenasas , Clonación Molecular , ADN Complementario/metabolismo , Humanos , Hidroxiesteroide Deshidrogenasas/deficiencia , Hidroxiesteroide Deshidrogenasas/genética , Hipertensión/etiología , Hipertensión/fisiopatología , Isoenzimas/genética , Mutación , Síndrome
20.
J Hypertens Suppl ; 14(5): S19-24, 1996 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-9120678

RESUMEN

CONVERSION OF CORTISOL TO CORTISONE: 11 beta-Hydroxysteroid dehydrogenase (11 beta-HSD) is a microsomal enzyme complex which, in humans, catalyses the interconversion between biologically active cortisol and inactive cortisone. This prereceptor signalling mechanism is essential for maintaining the aldosterone selectivity of the intrinsically non-specific mineralocorticoid receptor and for modulating glucocorticoid access to the glucocorticoid receptor. Apparent mineralocorticoid excess (AME) is a syndrome of severe low-renin mineralocorticoid hypertension associated with marked hypokalaemia which arises from a congenital deficiency of 11 beta-HSD. In AME patients, therefore, it is cortisol and not aldosterone which behaves as a potent mineralocorticoid. ISOFORMS OF 11 BETA-HSD: Two isoforms of human 11 beta-HSD have now been characterized and cloned. The type 1 isoform (11 beta-HSD1) is a low-affinity reduced nicotinamide adenine dinucleotide phosphate (NADP) dependent dehydrogenase-oxoreductase which is expressed in predominantly glucocorticoid target tissues and the encoding sequence of which is normal in patients with AME. In contrast, the type 2 isoform (11 beta-HSD2) is a high-affinity NADP-dependent unidirectional dehydrogenase which is expressed in placenta and mineralocorticoid target tissues such as renal collecting ducts and distal colonic epithelia. Exon- and intron-specific polymerase chain reaction amplification of the 11 beta-HSD2 gene from genomic DNA from members of a consanguinous kindred with AME consistently revealed a single missense mutation (C1228T) in two affected sibs and twin stillbirths. This mutation in codon 374 of exon 5 of the 11 beta-HSD2 gene creates an inframe premature stop (TGA) and, as such, results in a truncated 11 beta-HSD2 protein lacking the carboxyl-terminal proline-rich 32 amino acids. In keeping with an autosomal recessive mode of inheritance, both parents were phenotypically and biochemically normal but were heterozygous for this mutation. Unique to this kindred were expression analyses of the native mutant 11 beta-HSD2 enzyme in the stillbirth-affected placenta, which was almost completely devoid of NADP-dependent 11 beta-dehydrogenase activity. Immunohistochemical and Western blot analyses revealed the absence of 11 beta-HSD2 protein using antisera raised against synthetic peptide sequences corresponding either to the carboxyl terminus or other domains of the enzyme. MISSENSE MUTATION: In this kindred with AME, congenital deficiency of 11 beta-HSD activity is due to a single missense mutation in exon 5 of the 11 beta-HSD2 gene. Simultaneous studies by two other groups have similarly revealed no gross deletions or rearrangements of the 11 beta-HSD2 gene, but have described a number of single point mutations and oligonucleotide deletions in exons 3, 4 and 5, and adjacent to a splice site in intron 3. Recombinant expression analysis of site-directed mutant 11 beta-HSD2 complementary DNA constructs suggests a correlation between the predicted severity of these mutations and the biochemical and clinical phenotype. AME AS A CAUSE OF HYPERTENSION: The mutations in the 11 beta-HSD2 gene, together with those currently being sought by us for other kindreds with AME, establishes AME as a monogenic cause of human hypertension and will provide insight into the structure-function relationships of this important enzyme.


Asunto(s)
Hidroxiesteroide Deshidrogenasas/genética , Hipertensión/genética , Hipertensión/metabolismo , Mineralocorticoides/metabolismo , Mutación , 11-beta-Hidroxiesteroide Deshidrogenasas , Secuencia de Aminoácidos , Secuencia de Bases , Preescolar , Predisposición Genética a la Enfermedad , Humanos , Hidroxiesteroide Deshidrogenasas/deficiencia , Lactante , Masculino , Datos de Secuencia Molecular
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA