Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 42
Filtrar
1.
J Pediatr ; 220: 249-253, 2020 05.
Artigo em Inglês | MEDLINE | ID: mdl-31987650

RESUMO

There are few biomarkers to predict efficacy of glucocorticoid treatment in childhood acute lymphoblastic leukemia (ALL) at diagnosis. Here, we demonstrate reciprocal regulation of 11beta-hydroxysteroid dehydrogenase (11ß-HSD), may predict the apoptotic response of ALL to glucocorticoid treatment. Our data may be useful to refine glucocorticoid treatment, to retain benefit while minimizing side effects.


Assuntos
11-beta-Hidroxiesteroide Desidrogenase Tipo 1/fisiologia , 11-beta-Hidroxiesteroide Desidrogenase Tipo 2/fisiologia , Dexametasona/uso terapêutico , Glucocorticoides/uso terapêutico , Leucemia-Linfoma Linfoblástico de Células Precursoras/tratamento farmacológico , Prednisolona/uso terapêutico , Adolescente , Células Cultivadas , Criança , Pré-Escolar , Feminino , Humanos , Lactente , Masculino , Resultado do Tratamento
2.
Proc Natl Acad Sci U S A ; 113(22): 6265-70, 2016 May 31.
Artigo em Inglês | MEDLINE | ID: mdl-27185937

RESUMO

Fetoplacental glucocorticoid overexposure is a significant mechanism underlying fetal growth restriction and the programming of adverse health outcomes in the adult. Placental glucocorticoid inactivation by 11ß-hydroxysteroid dehydrogenase type 2 (11ß-HSD2) plays a key role. We previously discovered that Hsd11b2(-/-) mice, lacking 11ß-HSD2, show marked underdevelopment of the placental vasculature. We now explore the consequences for fetal cardiovascular development and whether this is reversible. We studied Hsd11b2(+/+), Hsd11b2(+/-), and Hsd11b2(-/-) littermates from heterozygous (Hsd11b(+/-)) matings at embryonic day (E)14.5 and E17.5, where all three genotypes were present to control for maternal effects. Using high-resolution ultrasound, we found that umbilical vein blood velocity in Hsd11b2(-/-) fetuses did not undergo the normal gestational increase seen in Hsd11b2(+/+) littermates. Similarly, the resistance index in the umbilical artery did not show the normal gestational decline. Surprisingly, given that 11ß-HSD2 absence is predicted to initiate early maturation, the E/A wave ratio was reduced at E17.5 in Hsd11b2(-/-) fetuses, suggesting impaired cardiac function. Pravastatin administration from E6.5, which increases placental vascular endothelial growth factor A and, thus, vascularization, increased placental fetal capillary volume, ameliorated the aberrant umbilical cord velocity, normalized fetal weight, and improved the cardiac function of Hsd11b2(-/-) fetuses. This improved cardiac function occurred despite persisting indications of increased glucocorticoid exposure in the Hsd11b2(-/-) fetal heart. Thus, the pravastatin-induced enhancement of fetal capillaries within the placenta and the resultant hemodynamic changes correspond with restored fetal cardiac function. Statins may represent a useful therapeutic approach to intrauterine growth retardation due to placental vascular hypofunction.


Assuntos
11-beta-Hidroxiesteroide Desidrogenase Tipo 2/fisiologia , Retardo do Crescimento Fetal/prevenção & controle , Glucocorticoides/metabolismo , Cardiopatias/prevenção & controle , Doenças Placentárias/prevenção & controle , Pravastatina/farmacologia , Animais , Anticolesterolemiantes/farmacologia , Feminino , Retardo do Crescimento Fetal/metabolismo , Retardo do Crescimento Fetal/patologia , Cardiopatias/metabolismo , Cardiopatias/patologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Doenças Placentárias/metabolismo , Doenças Placentárias/patologia , Gravidez , RNA Mensageiro/genética , Reação em Cadeia da Polimerase em Tempo Real , Reação em Cadeia da Polimerase Via Transcriptase Reversa
3.
Circulation ; 133(14): 1360-70, 2016 Apr 05.
Artigo em Inglês | MEDLINE | ID: mdl-26951843

RESUMO

BACKGROUND: The hypertensive syndrome of Apparent Mineralocorticoid Excess is caused by loss-of-function mutations in the gene encoding 11ß-hydroxysteroid dehydrogenase type 2 (11ßHSD2), allowing inappropriate activation of the mineralocorticoid receptor by endogenous glucocorticoid. Hypertension is attributed to sodium retention in the distal nephron, but 11ßHSD2 is also expressed in the brain. However, the central contribution to Apparent Mineralocorticoid Excess and other hypertensive states is often overlooked and is unresolved. We therefore used a Cre-Lox strategy to generate 11ßHSD2 brain-specific knockout (Hsd11b2.BKO) mice, measuring blood pressure and salt appetite in adults. METHODS AND RESULTS: Basal blood pressure, electrolytes, and circulating corticosteroids were unaffected in Hsd11b2.BKO mice. When offered saline to drink, Hsd11b2.BKO mice consumed 3 times more sodium than controls and became hypertensive. Salt appetite was inhibited by spironolactone. Control mice fed the same daily sodium intake remained normotensive, showing the intrinsic salt resistance of the background strain. Dexamethasone suppressed endogenous glucocorticoid and abolished the salt-induced blood pressure differential between genotypes. Salt sensitivity in Hsd11b2.BKO mice was not caused by impaired renal sodium excretion or volume expansion; pressor responses to phenylephrine were enhanced and baroreflexes impaired in these animals. CONCLUSIONS: Reduced 11ßHSD2 activity in the brain does not intrinsically cause hypertension, but it promotes a hunger for salt and a transition from salt resistance to salt sensitivity. Our data suggest that 11ßHSD2-positive neurons integrate salt appetite and the blood pressure response to dietary sodium through a mineralocorticoid receptor-dependent pathway. Therefore, central mineralocorticoid receptor antagonism could increase compliance to low-sodium regimens and help blood pressure management in cardiovascular disease.


Assuntos
11-beta-Hidroxiesteroide Desidrogenase Tipo 2/metabolismo , Fissura/fisiologia , Hipertensão/genética , Síndrome de Excesso Aparente de Minerolocorticoides/fisiopatologia , Proteínas do Tecido Nervoso/deficiência , Receptores de Mineralocorticoides/fisiologia , Cloreto de Sódio na Dieta/toxicidade , Núcleo Solitário/enzimologia , 11-beta-Hidroxiesteroide Desidrogenase Tipo 2/genética , 11-beta-Hidroxiesteroide Desidrogenase Tipo 2/fisiologia , Animais , Barorreflexo/efeitos dos fármacos , Corticosterona/sangue , Dexametasona/farmacologia , Comportamento de Ingestão de Líquido , Genes Sintéticos , Hipertensão/fisiopatologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Síndrome de Excesso Aparente de Minerolocorticoides/tratamento farmacológico , Síndrome de Excesso Aparente de Minerolocorticoides/genética , Antagonistas de Receptores de Mineralocorticoides/uso terapêutico , Néfrons/fisiopatologia , Proteínas do Tecido Nervoso/genética , Proteínas do Tecido Nervoso/fisiologia , Nestina/genética , Neurônios/fisiologia , Potássio/urina , RNA Mensageiro/biossíntese , Reflexo Anormal , Núcleo Solitário/fisiopatologia , Espironolactona/farmacologia
4.
Rev. Hosp. Ital. B. Aires (2004) ; 36(1): 11-14, mar. 2016.
Artigo em Espanhol | LILACS | ID: biblio-1147657

RESUMO

Los síndromes endocrinológicos con hipofunción o hiperfunción con niveles paradójicos de dosajes hormonales han sido bien caracterizados en los últimos años del siglo XX, a partir del desarrollo de técnicas genéticas y moleculares. Presentamos dos pacientes con pseudohipoaldosteronismo y aparente exceso de mineralocorticoides como síndromes en espejo, con la intención de alertar al médico clínico respecto de su consideración como entidad diagnóstica en niños con alteraciones hidroelectrolíticas. (AU)


Endocrinological syndromes with underactive or overactive hormonal levels with paradoxical dosages have been well characterized over the years of the twentieth century, from the development of genetic and molecular techniques. We present two patients with pseudohypoaldosteronism and apparent mineralocorticoid excess as mirror syndromes, with the aim to alert the clinician regarding their consideration as a diagnostic entity in children with fluid and electrolyte disturbances. (AU)


Assuntos
Humanos , Masculino , Lactente , Pseudo-Hipoaldosteronismo/diagnóstico , Síndrome de Excesso Aparente de Minerolocorticoides/diagnóstico , Peso-Idade , Dexametasona/uso terapêutico , Hidrocortisona/fisiologia , Hidrocortisona/sangue , Hidrocortisona/uso terapêutico , Pseudo-Hipoaldosteronismo/fisiopatologia , Pseudo-Hipoaldosteronismo/genética , Cloreto de Sódio/administração & dosagem , Síndrome de Excesso Aparente de Minerolocorticoides/fisiopatologia , 11-beta-Hidroxiesteroide Desidrogenase Tipo 2/fisiologia , Diuréticos/uso terapêutico , Aldosterona/fisiologia , Aldosterona/sangue , Alcalose/sangue , Hiperpotassemia/sangue , Hipopotassemia/sangue , Hiponatremia/sangue , Hipotonia Muscular/etiologia
5.
Placenta ; 37: 79-84, 2016 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-26654513

RESUMO

INTRODUCTION: Glucocorticoid-induced fetal programming has been associated with negative metabolic and cardiovascular sequelae in the adult. The placental enzyme 11beta-hydroxysteroid dehydrogenase type 2 (11ß-HSD2) shields the fetus from maternal glucocorticoid excess by catalyzing the conversion of these hormones into biologically inactive derivatives. In vivo experiments addressing placental barrier function are mostly conducted in rodents. Therefore we set out to characterize species-specific differences of rat and human placental 11ß-HSD2 steroid turnover, introducing Liquid Chromatography Tandem Mass-Spectrometry (LC-MS/MS) as a tool for rat tissue analysis. MATERIALS AND METHODS: Using LC-MS/MS we determined corticotropin-releasing hormone (CRH), cortisol (F), cortisone (E), corticosterone (B) and 11-dehydrocorticosterone (A) in human and rat placenta at term and measured the enzymatic 11ß-HSD glucocorticoid conversion-rates in placental microsomes of both species. In parallel, further glucocorticoid derivatives and sex steroids were determined in the same placental samples. RESULTS: In contrast to the human placenta, we did not detect CRH in the rat placenta. While cortisol (F) and cortisone (E) were exclusively present in human term placenta (E/F-ratio >1), rat placenta showed significant levels of corticosterone (B) and 11-dehydrocorticosterone (A), with an A/B-ratio <1. In line with these species-specific findings, human placenta showed a prominent 11ß-HSD2 activity, while in rat placenta higher 11ß-HSD1 glucocorticoid turnover rates were determined. DISCUSSION: Placental steroid metabolism of human and rat shows relevant species-specific differences, especially regarding the barrier function of 11ß-HSD2 at term. The exclusive expression of CRH in the human placenta further points to relevant differences in the regulation of parturition in rats. Consideration of these findings is warranted when transferring results from rodent placental glucocorticoid metabolism into humans.


Assuntos
11-beta-Hidroxiesteroide Desidrogenase Tipo 2/fisiologia , Placenta/enzimologia , Nascimento a Termo/metabolismo , 11-beta-Hidroxiesteroide Desidrogenase Tipo 2/química , 11-beta-Hidroxiesteroide Desidrogenase Tipo 2/metabolismo , Adulto , Animais , Cromatografia Líquida , Corticosterona/análogos & derivados , Corticosterona/análise , Corticosterona/metabolismo , Hormônio Liberador da Corticotropina/análise , Hormônio Liberador da Corticotropina/metabolismo , Feminino , Glucocorticoides/análise , Glucocorticoides/metabolismo , Humanos , Placenta/química , Placenta/metabolismo , Gravidez , Ratos , Ratos Wistar , Especificidade da Espécie , Espectrometria de Massas em Tandem
6.
Biol Reprod ; 93(2): 38, 2015 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-26085523

RESUMO

In sheep, the elongating conceptus synthesizes and secretes interferon tau (IFNT) as well as prostaglandins (PGs) and cortisol. The enzymes, hydroxysteroid (11-beta) dehydrogenase 1 (HSD11B1) and HSD11B2 interconvert cortisone and cortisol. In sheep, HSD11B1 is expressed and active in the conceptus trophectoderm as well as in the endometrial luminal epithelia; in contrast, HSD11B2 expression is most abundant in conceptus trophectoderm. Cortisol is a biologically active glucocorticoid and ligand for the glucocorticoid receptor (NR3C1 or GR) and mineralocorticoid receptor (NR3C2 or MR). Expression of MR is not detectable in either the ovine endometrium or conceptus during early pregnancy. In tissues that do not express MR, HSD11B2 protects cells from the growth-inhibiting and/or proapoptotic effects of cortisol, particularly during embryonic development. In study one, an in utero loss-of-function analysis of HSD11B1 and HSD11B2 was conducted in the conceptus trophectoderm using morpholino antisense oligonucleotides (MAOs) that inhibit mRNA translation. Elongating, filamentous conceptuses were recovered on Day 14 from ewes infused with control morpholino or HSD11B2 MAO. In contrast, HSD11B1 MAO resulted in severely growth-retarded conceptuses or conceptus fragments with apoptotic trophectoderm. In study two, clustered regularly interspaced short palindromic repeat (CRISPR)/Cas9 genome editing was used to determine the role of GR in conceptus elongation and development. Elongating, filamentous-type conceptuses (12-14 cm in length) were recovered from ewes gestating control embryos (n = 7/7) and gestating GR-edited embryos (n = 6/7). These results support the idea that the effects of HSD11B1-derived cortisol on conceptus elongation are indirectly mediated by the endometrium and are not directly mediated through GR in the trophectoderm.


Assuntos
11-beta-Hidroxiesteroide Desidrogenase Tipo 1/genética , 11-beta-Hidroxiesteroide Desidrogenase Tipo 1/fisiologia , 11-beta-Hidroxiesteroide Desidrogenase Tipo 2/genética , 11-beta-Hidroxiesteroide Desidrogenase Tipo 2/fisiologia , Embrião de Mamíferos/metabolismo , Desenvolvimento Embrionário/fisiologia , Receptores de Glucocorticoides/genética , Receptores de Glucocorticoides/fisiologia , Sequência de Aminoácidos , Animais , Apoptose/efeitos dos fármacos , Proteínas Associadas a CRISPR/genética , Sistemas CRISPR-Cas , Transferência Embrionária , Desenvolvimento Embrionário/genética , Feminino , Hidrocortisona/farmacologia , Dados de Sequência Molecular , Oligonucleotídeos Antissenso/farmacologia , Gravidez , RNA Mensageiro/biossíntese , RNA Mensageiro/genética , Carneiro Doméstico
7.
Zhongguo Dang Dai Er Ke Za Zhi ; 16(9): 939-43, 2014 Sep.
Artigo em Chinês | MEDLINE | ID: mdl-25229964

RESUMO

OBJECTIVE: To study the roles of type II 11ß-hydroxysteroid dehydrogenase (11ß-HSD2) and it's signaling factors in the lung tissue in pathogenesis of persistent pulmonary hypertension (PPH) in neonatal rats. METHODS: Six Sprague-Dawley rats on the 19th day of pregnancy were randomly divided into PPH and control groups (n=3 each). The PPH group was intraperitoneally injected with indomethacin (0.5 mg/kg) twice daily and exposed in 12% oxygen for three days, in order to prepare a fetal rat model of PPH. The control group was intraperitoneally injected with an equal volume of normal saline and exposed to air. Neonatal rats were born by caesarean section from both groups on the 22nd day of pregnancy. In each group, 15 neonatal rats were randomly selected and sacrificed. 11ß-HSD2 expression in the lung tissue of neonatal rats were observed by Confocal laser technology, and serum cortisol levels and prostacyclin, renin, angiotensin and aldosterone in the lung tissue of both groups were measured using ELISA. RESULTS: 11ß-HSD2 protein was widely expressed in the lung tissue of the control and PPH groups. The levels of 11ß-HSD2 and prostacyclin in the lung tissue were lower in the PPH group than in the control group, while serum cortisol levels and renin, angiotensin and aldosterone in the lung tissue were higher in the PPH group than in the control group (P<0.05). CONCLUSIONS: 11ß-HSD2 and it's signaling factors play roles in pathogenesis of PPH in neonatal rats.


Assuntos
11-beta-Hidroxiesteroide Desidrogenase Tipo 2/fisiologia , Hipertensão Pulmonar/etiologia , 11-beta-Hidroxiesteroide Desidrogenase Tipo 2/análise , Animais , Animais Recém-Nascidos , Feminino , Hipertensão Pulmonar/enzimologia , Masculino , Ratos , Ratos Sprague-Dawley , Transdução de Sinais
8.
Clin Obstet Gynecol ; 56(3): 602-9, 2013 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-23722920

RESUMO

Fetal glucocorticoid overexposure is a key potential mechanism underlying the link between low birthweight and later life diseases. The fetus is protected from high maternal glucocorticoid levels by the placental enzyme 11ß-hydroxysteroid dehydrogenase type 2. Antenatal glucocorticoid administration to women at threat of preterm labor, and high endogenous maternal glucocorticoid levels during pregnancy associate with lower birthweight. Long-term consequences for offspring include hypothalamic-pituitary-adrenal axis activation, increased metabolic and cardiovascular disorders, and neurodevelopmental sequelae. Strategies are needed to limit antenatal glucocorticoid use to those most at risk of preterm labor and to identify those most at risk of future disease.


Assuntos
11-beta-Hidroxiesteroide Desidrogenase Tipo 2/fisiologia , Doenças Cardiovasculares/embriologia , Dislipidemias/embriologia , Glucocorticoides/efeitos adversos , Hiperglicemia/embriologia , Sistema Hipotálamo-Hipofisário/efeitos dos fármacos , Sistema Hipófise-Suprarrenal/efeitos dos fármacos , Peso ao Nascer/efeitos dos fármacos , Doenças Cardiovasculares/induzido quimicamente , Dislipidemias/induzido quimicamente , Feminino , Desenvolvimento Fetal/efeitos dos fármacos , Humanos , Hiperglicemia/induzido quimicamente , Recém-Nascido , Trabalho de Parto Prematuro , Gravidez , Efeitos Tardios da Exposição Pré-Natal
9.
Curr Atheroscler Rep ; 15(5): 320, 2013 May.
Artigo em Inglês | MEDLINE | ID: mdl-23512604

RESUMO

Atherosclerosis is a chronic inflammatory disease in which initial vascular damage leads to extensive macrophage and lymphocyte infiltration. Although acutely glucocorticoids suppress inflammation, chronic glucocorticoid excess worsens atherosclerosis, possibly by exacerbating systemic cardiovascular risk factors. However, glucocorticoid action within the lesion may reduce neointimal proliferation and inflammation. Glucocorticoid levels within cells do not necessarily reflect circulating levels due to pre-receptor metabolism by 11ß-hydroxysteroid dehydrogenases (11ß-HSDs). 11ß-HSD2 converts active glucocorticoids into inert 11-keto forms. 11ß-HSD1 catalyses the reverse reaction, regenerating active glucocorticoids. 11ß-HSD2-deficiency/inhibition causes hypertension, whereas deficiency/inhibition of 11ß-HSD1 generates a cardioprotective lipid profile and improves glycemic control. Importantly, 11ß-HSD1-deficiency/inhibition is atheroprotective, whereas 11ß-HSD2-deficiency accelerates atherosclerosis. These effects are largely independent of systemic risk factors, reflecting modulation of glucocorticoid action and inflammation within the vasculature. Here, we consider whether evidence linking the 11ß-HSDs to vascular inflammation suggests these isozymes are potential therapeutic targets in vascular injury and atherosclerosis.


Assuntos
11-beta-Hidroxiesteroide Desidrogenase Tipo 1/fisiologia , 11-beta-Hidroxiesteroide Desidrogenase Tipo 2/fisiologia , Aterosclerose/imunologia , Glucocorticoides/imunologia , Vasculite/imunologia , 11-beta-Hidroxiesteroide Desidrogenase Tipo 1/antagonistas & inibidores , 11-beta-Hidroxiesteroide Desidrogenase Tipo 1/imunologia , 11-beta-Hidroxiesteroide Desidrogenase Tipo 2/imunologia , Aterosclerose/complicações , Aterosclerose/enzimologia , Glucocorticoides/metabolismo , Humanos , Neointima/imunologia , Vasculite/complicações , Vasculite/enzimologia
10.
Psychoneuroendocrinology ; 38(1): 1-11, 2013 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-22998948

RESUMO

Low birthweight, a marker of an adverse in utero environment, is associated with cardiometabolic disease and brain disorders in adulthood. The adaptive changes made by the fetus in response to the intra-uterine environment result in permanent changes in physiology, structure and metabolism, a phenomenon termed early life programming. One of the key hypotheses to explain programming, namely over exposure of the developing fetus to glucocorticoids, was proposed nearly two decades ago, following the observation that the fetus was protected from high glucocorticoid levels in the mother by the actions of the placental barrier enzyme, 11ß-hydroxysteroid dehydrogenase, which converts active glucocorticoids into inactive products. Numerous mechanistic studies in animal models have been carried out to test this hypothesis using manipulations to increase maternal glucocorticoids. Overall, these have resulted in offspring of lower birthweight, with an activated hypothalamic-pituitary-adrenal (HPA) axis and an adverse metabolic profile and behavioural phenotype in adulthood. Altered glucocorticoid activity or action is a good candidate mechanism in humans to link low birthweight with cardiometabolic and brain disorders. We have carried out detailed studies in men and women showing that high levels of endogenous glucocorticoids, or treatment with exogenous glucocorticoids, is associated with an adverse metabolic profile, increased cardiovascular disease and altered mood and cognitive decline. Our laboratory carried out the first translational studies in humans to test the glucocorticoid hypothesis, firstly demonstrating in studies of adult men and women, that low birthweight was associated with high fasting cortisol levels. We went on to dissect the mechanisms underlying the high fasting cortisol, demonstrating activation of the HPA axis, with increased cortisol responses to stimulation with exogenous adrenocorticotrophin hormone, lack of habituation to the stress of venepuncture, and increased cortisol responses to psychosocial stress. We have developed new dynamic tests to dissect the mechanisms regulating HPA axis central negative feedback sensitivity in humans, and demonstrated that this may be altered in obesity, one component of the metabolic syndrome. There are now studies in humans demonstrating that high circulating levels of maternal cortisol during pregnancy correlate negatively with birthweight, suggesting that excess glucocorticoids can by-pass the placental barrier. Deficiencies in the barrier enzyme, potentially increasing fetal glucocorticoid exposure, can also arise in association with maternal stress, malnutrition and disease, and can be inhibited by consumption of liquorice, which contains glycyrrhizin, an HSD inhibitor. Importantly, studies in humans have now demonstrated that high maternal cortisol in pregnancy and/or inhibition of HSD2 are associated with programmed outcomes in childhood including higher blood pressure, behavioural disorders as well as altered brain structure. We are investigating this further, using novel magnetic resonance imaging techniques to study the developing fetal brain in utero. The translational studies in support of the glucocorticoid hypothesis, and demonstrating that glucocorticoids are both mediators and targets of programming, are exciting and raise the question of whether this information can be used to identify those individuals most at risk of later life disease. In a recent study we showed that alterations in DNA methylation at genes important in regulating cortisol levels, tissue glucocorticoid action, blood pressure and fetal growth, are present in adulthood in association with both early life parameters and cardiometabolic risk factors. These preliminary data add to the limited literature in humans indicating a persisting epigenetic link between early life events and subsequent disease risk. Such findings open novel avenues for further exploration of the contribution of glucocorticoids to later life disease.


Assuntos
Síndrome de Cushing/fisiopatologia , Glucocorticoides/fisiologia , Sistema Hipotálamo-Hipofisário/fisiopatologia , Recém-Nascido de Baixo Peso , Recém-Nascido Pequeno para a Idade Gestacional , Modelos Biológicos , Sistema Hipófise-Suprarrenal/fisiopatologia , Complicações na Gravidez/fisiopatologia , Efeitos Tardios da Exposição Pré-Natal , Estresse Fisiológico/fisiologia , Estresse Psicológico/fisiopatologia , 11-beta-Hidroxiesteroide Desidrogenase Tipo 2/antagonistas & inibidores , 11-beta-Hidroxiesteroide Desidrogenase Tipo 2/fisiologia , Adaptação Fisiológica , Adulto , Animais , Distinções e Prêmios , Encefalopatias Metabólicas/embriologia , Encefalopatias Metabólicas/etiologia , Encefalopatias Metabólicas/fisiopatologia , Doenças Cardiovasculares/embriologia , Doenças Cardiovasculares/etiologia , Doenças Cardiovasculares/fisiopatologia , Transtornos Cognitivos/etiologia , Transtornos Cognitivos/fisiopatologia , Síndrome de Cushing/embriologia , Síndrome de Cushing/psicologia , Deficiências do Desenvolvimento/etiologia , Deficiências do Desenvolvimento/fisiopatologia , Retroalimentação Fisiológica , Feminino , Feto/fisiologia , Glucocorticoides/efeitos adversos , Humanos , Recém-Nascido , Troca Materno-Fetal , Síndrome Metabólica/embriologia , Síndrome Metabólica/etiologia , Síndrome Metabólica/fisiopatologia , Neuroendocrinologia , Neuropsicologia , Placenta/enzimologia , Gravidez , Complicações na Gravidez/psicologia
11.
Best Pract Res Clin Endocrinol Metab ; 26(5): 689-700, 2012 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-22980050

RESUMO

Epidemiological evidence suggests that exposure to an adverse environment in early life is associated with an increased risk of cardio-metabolic and behavioral disorders in adulthood, a phenomenon termed 'early life programming'. One major hypothesis for early life programming is fetal glucocorticoid overexposure. In animal studies, prenatal glucocorticoid excess as a consequence of maternal stress or through exogenous administration to the mother or fetus is associated with programming effects on cardiovascular and metabolic systems and on the brain. These effects can be transmitted to subsequent generations. Studies in humans provide some evidence that prenatal glucocorticoid exposure may exert similar programming effects on glucose/insulin homeostasis, blood pressure and neurodevelopment. The mechanisms by which glucocorticoids mediate these effects are unclear but may include a role for epigenetic modifications. This review discusses the evidence for glucocorticoid programming in animal models and in humans.


Assuntos
Desenvolvimento Fetal/efeitos dos fármacos , Glucocorticoides/fisiologia , 11-beta-Hidroxiesteroide Desidrogenase Tipo 2/fisiologia , Animais , Pressão Sanguínea/efeitos dos fármacos , Encéfalo/efeitos dos fármacos , Encéfalo/embriologia , Epigênese Genética , Fígado Gorduroso/induzido quimicamente , Feminino , Feto/fisiologia , Glucocorticoides/administração & dosagem , Glucocorticoides/efeitos adversos , Glucose/metabolismo , Homeostase/efeitos dos fármacos , Humanos , Insulina/metabolismo , Hepatopatia Gordurosa não Alcoólica , Pâncreas/efeitos dos fármacos , Pâncreas/fisiologia , Gravidez , Efeitos Tardios da Exposição Pré-Natal
13.
Intern Med ; 49(20): 2235-9, 2010.
Artigo em Inglês | MEDLINE | ID: mdl-20962442

RESUMO

A 91-year-old depressed woman had resistant hypertension despite a triple combination of anti-hypertensives, including a Ca-antagonist, an angiotensin receptor blocker, and a thiazide diuretic at optimal doses. She had hypokalemic metabolic alkalosis, elevated plasma cortisol and ACTH, and elevated urinary cortisol. The high-dose dexamethasone did not suppress the elevated ACTH and cortisol. The addition of spironolactone to her previous medications controlled and normalized hypertension, hypokalemia, and hormonal abnormalities within 4 months. Her blood pressure, serum electrolytes, and the hormonal states remained normalized for more than a year thereafter. Her depressed mental state also improved after spironolactone.


Assuntos
11-beta-Hidroxiesteroide Desidrogenase Tipo 2/fisiologia , Síndrome de ACTH Ectópico/diagnóstico , Hormônio Adrenocorticotrópico/metabolismo , Síndrome de Cushing/etiologia , Hidrocortisona/metabolismo , Hipertensão/etiologia , Receptores de Glucocorticoides/fisiologia , Receptores de Mineralocorticoides/fisiologia , Espironolactona/uso terapêutico , Idoso de 80 Anos ou mais , Anti-Hipertensivos/uso terapêutico , Síndrome de Cushing/diagnóstico , Síndrome de Cushing/tratamento farmacológico , Síndrome de Cushing/fisiopatologia , Depressão/tratamento farmacológico , Depressão/etiologia , Depressão/fisiopatologia , Dexametasona , Diagnóstico Diferencial , Resistência a Medicamentos , Quimioterapia Combinada , Feminino , Humanos , Hipertensão/tratamento farmacológico , Hipopotassemia/etiologia , Hipopotassemia/fisiopatologia , Estresse Psicológico/complicações
15.
Physiol Genomics ; 42(3): 319-30, 2010 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-20571110

RESUMO

11beta-Hydroxysteroid dehydrogenase type 2 (11betaHSD2) is a short-chain dehydrogenase/reductase (SDR) responsible for inactivating cortisol and preventing its binding to the mineralocorticoid receptor (MR). Nonfunctional mutations in HSD11B2, the gene encoding 11betaHSD2, cause the hypertensive syndrome of apparent mineralocorticoid excess (AME). Like other such Mendelian disorders, AME is rare but has nevertheless helped to illuminate principles fundamental to the regulation of blood pressure. Furthermore, polymorphisms in HSD11B2 have been associated with salt sensitivity, a major risk factor for cardiovascular mortality. It is therefore highly likely that sequence variation in HSD11B2, having subtle functional ramifications, will affect blood pressure in the wider population. In this study, a three-dimensional homology model of 11betaHSD2 was created and used to hypothesize the functional consequences in terms of protein structure of published mutations in HSD11B2. This approach underscored the strong genotype-phenotype correlation of AME: severe forms of the disease, associated with little in vivo enzyme activity, arise from mutations occurring in invariant alignment positions. These were predicted to exert gross structural changes in the protein. In contrast, those mutations causing a mild clinical phenotype were in less conserved regions of the protein that were predicted to be relatively more tolerant to substitution. Finally, a number of pathogenic mutations are shown to be associated with regions predicted to participate in dimer formation, and in protein stabilization, which may therefore suggest molecular mechanisms of disease.


Assuntos
11-beta-Hidroxiesteroide Desidrogenase Tipo 2/genética , 11-beta-Hidroxiesteroide Desidrogenase Tipo 2/fisiologia , Biologia Computacional , 11-beta-Hidroxiesteroide Desidrogenase Tipo 2/química , 11-beta-Hidroxiesteroide Desidrogenase Tipo 2/metabolismo , Sequência de Aminoácidos , Animais , Biologia Computacional/métodos , Análise Mutacional de DNA/métodos , Bases de Dados Genéticas , Família , Predisposição Genética para Doença , Humanos , Hipertensão/genética , Síndrome de Excesso Aparente de Minerolocorticoides/genética , Modelos Biológicos , Modelos Moleculares , Dados de Sequência Molecular , Proteínas Mutantes/química , Proteínas Mutantes/genética , Proteínas Mutantes/metabolismo , Proteínas Mutantes/fisiologia , Polimorfismo Genético , Conformação Proteica , Homologia de Sequência de Aminoácidos , Relação Estrutura-Atividade
16.
Nephrol Dial Transplant ; 25(10): 3195-203, 2010 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-20566573

RESUMO

BACKGROUND: Intrauterine growth restriction (IUGR) is associated with systemic hypertension of the offspring later in life. The exact mechanisms are still incompletely understood. 11ß-Hydroxysteroid dehydrogenase 2 (11ß-HSD2) in the distal renal tubule protects the mineralocorticoid receptor from cortisol. As we did not find a suppression of 11ß-HSD2 in total kidney of IUGR animals, our objective was to investigate whether a suppression of 11ß-HSD2 could be detected on a more sophisticated level such as in situ protein and gene expression of 11ß-HSD2 in mildly hypertensive IUGR offspring. METHODS: IUGR rats after maternal low-protein diet (n = 17) were compared with controls (n = 18). At 70 and 120 days of age, in situ distribution of 11ß-HSD2 gene and protein expression was investigated by RT-PCR of microdissected tubules and immunohistochemistry. For in situ localization studies, double staining for 11ß-HSD2 and calbindin was used. Serum levels of corticosterone and dehydrocorticosterone were measured by tandem mass spectrometry. RESULTS: In IUGR rats, intra-arterial blood pressure significantly increased at Day 120 of life. Serum corticosterone/dehydrocorticosterone ratios and 11ß-HSD2 mRNA in total kidney were not altered in IUGR animals. However, 11ß-HSD2 mRNA concentration was significantly lower in microdissected tubuli of IUGR animals (Day 120: 0.18 ± 0.14 vs 1.00 ± 0.32 rel. units in controls; P < 0.05). In IUGR animals, immunostaining scores for 11ß-HSD2 were significantly lower than in controls (P < 0.05). Double staining with calbindin showed lower expression of 11ß-HSD2 in distal segments of the distal tubule. CONCLUSIONS: Our data indicate lower gene and protein expression of the pre-receptor enzyme 11ß-HSD2 in IUGR animals when looking at specific renal compartments, but not in total kidney extracts. Thus, lower 11ß-HSD2 as a mechanism for hypertension later in life might be missed without methods for in situ detection.


Assuntos
11-beta-Hidroxiesteroide Desidrogenase Tipo 2/fisiologia , Dieta com Restrição de Proteínas , Retardo do Crescimento Fetal/enzimologia , 11-beta-Hidroxiesteroide Desidrogenase Tipo 2/análise , 11-beta-Hidroxiesteroide Desidrogenase Tipo 2/genética , Animais , Corticosterona/análogos & derivados , Corticosterona/sangue , Feminino , Hipertensão/etiologia , RNA Mensageiro/análise , Ratos , Ratos Wistar , Receptores de Glucocorticoides/genética , Receptores de Mineralocorticoides/genética
17.
Clin Sci (Lond) ; 119(10): 437-42, 2010 Aug 05.
Artigo em Inglês | MEDLINE | ID: mdl-20515440

RESUMO

Abnormal glucocorticoid metabolism contributes to vascular dysfunction and cardiovascular disease. Cortisol activation of vascular mineralocorticoid and glucocorticoid receptors is regulated by two types of 11beta-HSD (11-beta hydroxysteroid dehydrogenase), namely 11beta-HSD2 and 11beta-HSD1 (type 2 and type 1 11beta-HSD respectively). We hypothesized that inhibition of 11beta-HSD would attenuate vascular function in healthy humans. A total of 15 healthy subjects were treated with the selective 11beta-HSD inhibitor GA (glycyrrhetinic acid) or matching placebo in a randomized double-blinded cross-over trial. 11beta-HSD activity was assessed by the urinary cortisol/cortisone ratio, and vascular function was measured using strain-gauge plethysmography. Endothelial function was measured through incremental brachial artery administration of methacholine (0.3-10 microg/min) and vascular smooth muscle function with incremental verapamil (10-300 microg/min). GA increased the 24-h urinary cortisol/cortisone ratio compared with placebo (P=0.008). GA tended to reduce the FBF (forearm blood flow) response to methacholine (P=0.09) and significantly reduced the FBF response to verapamil compared with placebo (P=0.04). MAP (mean arterial pressure) did not differ between the study conditions. 11beta-HSD inhibition attenuated vascular smooth muscle vasodilatory function in healthy humans. Disturbances in cortisol activity resulting from 11beta-HSD inactivation is therefore a second plausible mechanism for mineralocorticoid-mediated hypertension in humans.


Assuntos
Inibidores Enzimáticos/farmacologia , Ácido Glicirretínico/farmacologia , Músculo Liso Vascular/efeitos dos fármacos , Vasodilatação/efeitos dos fármacos , 11-beta-Hidroxiesteroide Desidrogenase Tipo 2/antagonistas & inibidores , 11-beta-Hidroxiesteroide Desidrogenase Tipo 2/fisiologia , Adulto , Pressão Sanguínea/efeitos dos fármacos , Cortisona/urina , Estudos Cross-Over , Método Duplo-Cego , Endotélio Vascular/efeitos dos fármacos , Endotélio Vascular/fisiologia , Feminino , Humanos , Hidrocortisona/urina , Masculino , Músculo Liso Vascular/fisiologia , Fluxo Sanguíneo Regional/efeitos dos fármacos , Fluxo Sanguíneo Regional/fisiologia , Vasodilatação/fisiologia , Adulto Jovem
18.
Blood Purif ; 29(2): 111-3, 2010.
Artigo em Inglês | MEDLINE | ID: mdl-20093814

RESUMO

Aldosterone was in the past considered only as a prohypertensinogenic agent. It has recently become clear that apart from the classical endocrine action, i.e. causing blood pressure elevation as a result of salt retention, aldosterone has numerous blood-pressure-independent actions on nonepithelial tissue. Under conditions of high salt concentration, aldosterone is injurious to the kidney, heart and vasculature. Of particular interest are recent observations that aldosterone is a permissive factor for the effect of minor increases in plasma sodium concentration on endothelial cell dysfunction. Despite surprising effects of aldosterone blockade on blood pressure of anuric dialysis patients, the potential role of mineralocorticoid receptor blockade in dialysis patients is currently unclear and requires controlled investigation to define the risk of potential hazards, specifically hyperkalemia.


Assuntos
Aldosterona/fisiologia , Hiperaldosteronismo/complicações , Hipertensão/fisiopatologia , Antagonistas de Receptores de Mineralocorticoides , Antagonistas de Receptores de Mineralocorticoides/uso terapêutico , Sódio na Dieta/efeitos adversos , Uremia/fisiopatologia , 11-beta-Hidroxiesteroide Desidrogenase Tipo 2/fisiologia , Células Endoteliais/efeitos dos fármacos , Endotélio Vascular/fisiopatologia , Humanos , Hidrocortisona/efeitos adversos , Hidrocortisona/fisiologia , Hiperaldosteronismo/tratamento farmacológico , Hiperpotassemia/induzido quimicamente , Hipertensão/sangue , Hipertensão/tratamento farmacológico , Hipertensão/etiologia , Hipertensão/prevenção & controle , Síndrome Metabólica/fisiopatologia , Antagonistas de Receptores de Mineralocorticoides/efeitos adversos , Receptores de Mineralocorticoides/química , Receptores de Mineralocorticoides/fisiologia , Sistema Renina-Angiotensina/efeitos dos fármacos , Sistema Renina-Angiotensina/fisiologia , Sódio/sangue , Sódio na Dieta/farmacocinética , Espironolactona/efeitos adversos , Espironolactona/uso terapêutico , Uremia/sangue , Resistência Vascular/efeitos dos fármacos , Resistência Vascular/fisiologia
19.
Braz J Med Biol Res ; 43(1): 52-6, 2010 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-20027486

RESUMO

Aldosterone concentrations vary in advanced chronic renal failure (CRF). The isozyme 11beta-hydroxysteroid dehydrogenase 2 (11beta-HSD2), which confers aldosterone specificity for mineralocorticoid receptors in distal tubules and collecting ducts, has been reported to be decreased or normal in patients with renal diseases. Our objective was to determine the role of aldosterone and 11beta-HSD2 renal microsome activity, normalized for glomerular filtration rate (GFR), in maintaining K+ homeostasis in 5/6 nephrectomized rats. Male Wistar rats weighing 180-220 g at the beginning of the study were used. Rats with experimental CRF obtained by 5/6 nephrectomy (N = 9) and sham rats (N = 10) were maintained for 4 months. Systolic blood pressure and plasma creatinine (Pcr) concentration were measured at the end of the experiment. Sodium and potassium excretion and GFR were evaluated before and after spironolactone administration (10 mg.kg-1.day-1 for 7 days) and 11beta-HSD2 activity on renal microsomes was determined. Systolic blood pressure (means +/- SEM; Sham = 105 +/- 8 and CRF = 149 +/- 10 mmHg) and Pcr (Sham = 0.42 +/- 0.03 and CRF = 2.53 +/- 0.26 mg/dL) were higher (P < 0.05) while GFR (Sham = 1.46 +/- 0.26 and CRF = 0.61 +/- 0.06 mL/min) was lower (P < 0.05) in CRF, and plasma aldosterone (Pald) was the same in the two groups. Urinary sodium and potassium excretion was similar in the two groups under basal conditions but, after spironolactone treatment, only potassium excretion was decreased in CRF rats (sham = 0.95 +/- 0.090 (before) vs 0.89 +/- 0.09 microEq/min (after) and CRF = 1.05 +/- 0.05 (before) vs 0.37 +/- 0.07 microEq/min (after); P < 0.05). 11beta-HSD2 activity on renal microsomes was lower in CRF rats (sham = 0.807 +/- 0.09 and CRF = 0.217 +/- 0.07 nmol.min-1.mg protein-1; P < 0.05), although when normalized for mL GFR it was similar in both groups. We conclude that K+ homeostasis is maintained during CRF development despite normal Pald levels. This adaptation may be mediated by renal 11beta-HSD2 activity, which, when normalized for GFR, became similar to that of control rats, suggesting that mineralocorticoid receptors maintain their aldosterone selectivity.


Assuntos
11-beta-Hidroxiesteroide Desidrogenase Tipo 2/fisiologia , Homeostase/fisiologia , Falência Renal Crônica/metabolismo , Microssomos/enzimologia , Potássio/metabolismo , 11-beta-Hidroxiesteroide Desidrogenase Tipo 2/metabolismo , Aldosterona/sangue , Animais , Pressão Sanguínea/fisiologia , Falência Renal Crônica/enzimologia , Masculino , Nefrectomia , Ratos , Ratos Wistar
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...