Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 54
Filter
Add more filters

Country/Region as subject
Publication year range
1.
J Biol Chem ; 292(18): 7578-7587, 2017 05 05.
Article in English | MEDLINE | ID: mdl-28302719

ABSTRACT

The expression of 11ß-hydroxysteroid dehydrogenase type 2 (11ß-HSD2), which acts as a placental glucocorticoid barrier, is silenced in cytotrophoblasts but substantially up-regulated during syncytialization. However, the repressive mechanism of 11ß-HSD2 expression before syncytialization and how this repression is lifted during syncytialization remain mostly unresolved. Here we found that enhancer of zeste homolog 2 (EZH2) accounts for the silence of 11ß-HSD2 expression via trimethylation of histone H3 lysine 27 at the promoter of the 11ß-HSD2 gene. Further studies revealed that, upon syncytialization, human chorionic gonadotropin reduced the phosphorylation of retinoblastoma protein (pRB) via activation of the cAMP/PKA pathway, which sequesters E2F transcription factor 1 (E2F1), the transcription factor for EZH2 expression. As a result of inactivation of the pRB-E2F1-EZH2 pathway, the repressive marker trimethylation of histone H3 lysine 27 at the 11ß-HSD2 promoter is removed, which leads to the robust expression of 11ß-HSD2 during syncytialization.


Subject(s)
11-beta-Hydroxysteroid Dehydrogenase Type 2/biosynthesis , Enhancer of Zeste Homolog 2 Protein/metabolism , Gene Expression Regulation, Enzymologic/physiology , Placenta/enzymology , Pregnancy Proteins/metabolism , Repressor Proteins/metabolism , Second Messenger Systems/physiology , 11-beta-Hydroxysteroid Dehydrogenase Type 2/genetics , Adult , Cyclic AMP/genetics , Cyclic AMP/metabolism , E2F1 Transcription Factor/genetics , E2F1 Transcription Factor/metabolism , Enhancer of Zeste Homolog 2 Protein/genetics , Female , Histones/genetics , Histones/metabolism , Humans , Methylation , Pregnancy , Pregnancy Proteins/genetics , Repressor Proteins/genetics , Retinoblastoma Protein/genetics , Retinoblastoma Protein/metabolism
2.
J Biol Chem ; 292(18): 7542-7553, 2017 05 05.
Article in English | MEDLINE | ID: mdl-28320863

ABSTRACT

The aim of the study was to characterize the role of Rac1 GTPase for the mineralocorticoid receptor (MR)-mediated pro-fibrotic remodeling. Transgenic mice with cardiac overexpression of constitutively active Rac1 (RacET) develop an age-dependent phenotype with atrial dilatation, fibrosis, and atrial fibrillation. Expression of MR was similar in RacET and WT mice. The expression of 11ß hydroxysteroid dehydrogenase type 2 (11ß-HSD2) was age-dependently up-regulated in the atria and the left ventricles of RacET mice on mRNA and protein levels. Statin treatment inhibiting Rac1 geranylgeranylation reduced 11ß-HSD2 up-regulation. Samples of human left atrial myocardium showed a positive correlation between Rac1 activity and 11ß-HSD2 expression (r = 0.7169). Immunoprecipitation showed enhanced Rac1-bound 11ß-HSD2 relative to Rac1 expression in RacET mice that was diminished with statin treatment. Both basal and phorbol 12-myristate 13-acetate (PMA)-induced NADPH oxidase activity were increased in RacET and correlated positively with 11ß-HSD2 expression (r = 0.788 and r = 0.843, respectively). In cultured H9c2 cardiomyocytes, Rac1 activation with l-buthionine sulfoximine increased; Rac1 inhibition with NSC23766 decreased 11ß-HSD2 mRNA and protein expression. Connective tissue growth factor (CTGF) up-regulation induced by aldosterone was prevented with NSC23766. Cardiomyocyte transfection with 11ß-HSD2 siRNA abolished the aldosterone-induced CTGF up-regulation. Aldosterone-stimulated MR nuclear translocation was blocked by the 11ß-HSD2 inhibitor carbenoxolone. In cardiac fibroblasts, nuclear MR translocation induced by aldosterone was inhibited with NSC23766 and spironolactone. NSC23766 prevented the aldosterone-induced proliferation and migration of cardiac fibroblasts and the up-regulation of CTGF and fibronectin. In conclusion, Rac1 GTPase regulates 11ß-HSD2 expression, MR activation, and MR-mediated pro-fibrotic signaling.


Subject(s)
11-beta-Hydroxysteroid Dehydrogenase Type 2/biosynthesis , Endomyocardial Fibrosis/enzymology , Fibroblasts/enzymology , Myocardium/enzymology , Myocytes, Cardiac/enzymology , Neuropeptides/biosynthesis , Signal Transduction , rac1 GTP-Binding Protein/biosynthesis , 11-beta-Hydroxysteroid Dehydrogenase Type 2/genetics , Aldosterone/pharmacology , Animals , Cell Line , Connective Tissue Growth Factor/biosynthesis , Connective Tissue Growth Factor/genetics , Endomyocardial Fibrosis/pathology , Fibroblasts/pathology , Fibronectins/biosynthesis , Fibronectins/genetics , Gene Expression Regulation/drug effects , Humans , Methionine/analogs & derivatives , Methionine/pharmacology , Mice , Mice, Mutant Strains , Myocardium/pathology , Myocytes, Cardiac/pathology , Neuropeptides/genetics , Rats , Rats, Sprague-Dawley , Sulfoxides/pharmacology , Tetradecanoylphorbol Acetate/pharmacology , rac1 GTP-Binding Protein/genetics
3.
Hum Mol Genet ; 25(23): 5167-5177, 2016 12 01.
Article in English | MEDLINE | ID: mdl-27798095

ABSTRACT

FDA-approved mineralocorticoid receptor (MR) antagonists are used to treat heart failure. We have recently demonstrated efficacy of MR antagonists for skeletal muscles in addition to heart in Duchenne muscular dystrophy mouse models and that mineralocorticoid receptors are present and functional in skeletal muscles. The goal of this study was to elucidate the underlying mechanisms of MR antagonist efficacy on dystrophic skeletal muscles. We demonstrate for the first time that infiltrating myeloid cells clustered in damaged areas of dystrophic skeletal muscles have the capacity to produce the natural ligand of MR, aldosterone, which in excess is known to exacerbate tissue damage. Aldosterone synthase protein levels are increased in leukocytes isolated from dystrophic muscles compared with controls and local aldosterone levels in dystrophic skeletal muscles are increased, despite normal circulating levels. All genes encoding enzymes in the pathway for aldosterone synthesis are expressed in muscle-derived leukocytes. 11ß-HSD2, the enzyme that inactivates glucocorticoids to increase MR selectivity for aldosterone, is also increased in dystrophic muscle tissues. These results, together with the demonstrated preclinical efficacy of antagonists, suggest MR activation is in excess of physiological need and likely contributes to the pathology of muscular dystrophy. This study provides new mechanistic insight into the known contribution of myeloid cells to muscular dystrophy pathology. This first report of myeloid cells having the capacity to produce aldosterone may have implications for a wide variety of acute injuries and chronic diseases with inflammation where MR antagonists may be therapeutic.


Subject(s)
Heart Failure/drug therapy , Mineralocorticoid Receptor Antagonists/administration & dosage , Muscle, Skeletal/drug effects , Muscular Dystrophy, Duchenne/drug therapy , 11-beta-Hydroxysteroid Dehydrogenase Type 2/biosynthesis , 11-beta-Hydroxysteroid Dehydrogenase Type 2/genetics , Aldosterone/metabolism , Animals , Cytochrome P-450 CYP11B2/biosynthesis , Cytochrome P-450 CYP11B2/genetics , Disease Models, Animal , Heart/drug effects , Heart/physiopathology , Heart Failure/metabolism , Heart Failure/pathology , Humans , Mice , Muscle, Skeletal/pathology , Muscular Dystrophy, Duchenne/genetics , Muscular Dystrophy, Duchenne/pathology , Myeloid Cells/drug effects , Myeloid Cells/pathology
4.
Biol Reprod ; 96(5): 1019-1030, 2017 May 01.
Article in English | MEDLINE | ID: mdl-28419184

ABSTRACT

Although in vitro exposure to physiological concentrations of glucorticoids did not affect maturation of mouse oocytes, it significantly inhibited nuclear maturation of pig oocytes. Studies on this species difference in oocyte sensitivity to glucocorticoids will contribute to our understanding of how stress/glucocorticoids affect oocytes. We showed that glucorticoid receptors (NR3C1) were expressed in both oocytes and cumulus cells (CCs) of both pigs and mice; however, while cortisol inhibition of oocyte maturation was overcome by NR3C1 inhibitor RU486 in pigs, it could not be relieved by RU486 in mice. The mRNA level of 11ß-hydroxysteroid dehydrogenase 1 (HSD11B1) was significantly higher than that of HSD11B2 in pig cumulus-oocyte complexes (COCs), whereas HSD11B2 was exclusively expressed in mouse COCs. Pig and mouse cumulus-denuded oocytes (DOs) expressed HSD11B2 predominantly and exclusively, respectively. In the presence of cortisol, although inhibiting HSD11B2 decreased maturation rates of COCs in both species, inhibiting HSD11B1 improved maturation of pig COCs while having no effect on mouse COCs. Cortisol-cortisone interconversion observation confirmed high HSD11B1 activities in pig oocytes but none in mouse oocytes, a higher HSD11B2 activity in mouse than in pig oocytes, and a rapid cortisol-cortisone interconversion in pig COCs catalyzed by HSD11B1 from CCs and HSD11B2 from DOs. In conclusion, the species difference in glucocorticoid sensitivity between pig and mouse oocytes is caused by their different contents/ratios of HSD11B1 and HSD11B2, which maintain different concentrations of active glucocorticoids. While cortisol inhibited pig oocytes by interacting with NR3C1, glucocorticoid suppression of mouse oocytes was apparently not mediated by NR3C1.


Subject(s)
Glucocorticoids/pharmacology , Oocytes/drug effects , 11-beta-Hydroxysteroid Dehydrogenase Type 1/antagonists & inhibitors , 11-beta-Hydroxysteroid Dehydrogenase Type 1/biosynthesis , 11-beta-Hydroxysteroid Dehydrogenase Type 2/antagonists & inhibitors , 11-beta-Hydroxysteroid Dehydrogenase Type 2/biosynthesis , Animals , Cortisone/metabolism , Cumulus Cells/drug effects , Cumulus Cells/metabolism , Female , Hydrocortisone/metabolism , Mice , Mifepristone/pharmacology , Oogenesis , Receptors, Glucocorticoid/antagonists & inhibitors , Receptors, Glucocorticoid/biosynthesis , Receptors, Glucocorticoid/genetics , Species Specificity , Sus scrofa
5.
Clin Exp Allergy ; 44(2): 197-211, 2014 Feb.
Article in English | MEDLINE | ID: mdl-24447082

ABSTRACT

BACKGROUND: Glucocorticoids are used to treat allergic rhinitis, but the mechanisms by which they induce disease remission are unclear. 11ß-hydroxysteroid dehydrogenase (11ß-HSD) is a tissue-specific regulator of glucocorticoid responses, inducing the interconversion of inactive and active glucocorticoids. OBJECTIVE: We analysed the expression and distribution patterns of 11ß-HSD1, 11ß-HSD2, and steroidogenic enzymes in normal and allergic nasal mucosa, and cytokine-driven regulation of their expression. The production levels of cortisol in normal, allergic nasal mucosa and in cultured epithelial cells stimulated with cytokines were also determined. METHODS: The expression levels of 11ß-HSD1, 11ß-HSD2, steroidogenic enzymes (CYP11B1, CYP11A1), and cortisol in normal, mild, and moderate/severe persistent allergic nasal mucosa were assessed by real-time PCR, Western blot, immunohistochemistry, and ELISA. The expression levels of 11ß-HSD1, 11ß-HSD2, CYP11B1, CYP11A1, and cortisol were also determined in cultured nasal epithelial cell treated with IL-4, IL-5, IL-13, IL-17A, and IFN-γ. Conversion ratio of cortisone to cortisol was evaluated using siRNA technique, 11ß-HSD1 inhibitor, and the measurement of 11ß-HSD1 activity. RESULTS: The expression levels of 11ß-HSD1, CYP11B1, and cortisol were up-regulated in mild and moderate/severe persistent allergic nasal mucosa. By contrast, 11ß-HSD2 expression was decreased in allergic nasal mucosa. In cultured epithelial cells treated with IL-4, IL-5, IL-13, and IL-17A, 11ß-HSD1 expression and activity increased in parallel with the expression levels of CYP11B1 and cortisol, but the production of 11ß-HSD2 decreased. CYP11A1 expression level was not changed in allergic nasal mucosa or in response to stimulation with cytokines. SiRNA technique or the measurement of 11ß-HSD1 activity showed that nasal epithelium activates cortisone to cortisol in a 11ß-HSD-dependent manner. CONCLUSIONS AND CLINICAL RELEVANCE: These results indicate that the localized anti-inflammatory effects of glucocorticoids are regulated by inflammatory cytokines, which can modulate the expression of 11ß-HSD1, 11ß-HSD2, and CYP11B1, and by the intracellular concentrations of bioactive glucocorticoids.


Subject(s)
11-beta-Hydroxysteroid Dehydrogenase Type 1/biosynthesis , 11-beta-Hydroxysteroid Dehydrogenase Type 2/biosynthesis , Cytokines/biosynthesis , Nasal Mucosa/metabolism , Rhinitis, Allergic, Perennial/metabolism , Th2 Cells/metabolism , 11-beta-Hydroxysteroid Dehydrogenase Type 1/genetics , 11-beta-Hydroxysteroid Dehydrogenase Type 1/immunology , 11-beta-Hydroxysteroid Dehydrogenase Type 2/genetics , 11-beta-Hydroxysteroid Dehydrogenase Type 2/immunology , Adult , Cholesterol Side-Chain Cleavage Enzyme/biosynthesis , Cholesterol Side-Chain Cleavage Enzyme/immunology , Cytokines/immunology , Female , Glucocorticoids/administration & dosage , Glucocorticoids/pharmacokinetics , Humans , Hydrocortisone/immunology , Hydrocortisone/metabolism , Male , Middle Aged , Nasal Mucosa/immunology , Nasal Mucosa/pathology , Rhinitis, Allergic, Perennial/drug therapy , Rhinitis, Allergic, Perennial/immunology , Rhinitis, Allergic, Perennial/pathology , Steroid 11-beta-Hydroxylase/biosynthesis , Steroid 11-beta-Hydroxylase/immunology , Th2 Cells/immunology , Th2 Cells/pathology
6.
Reprod Biol Endocrinol ; 12: 77, 2014 Aug 09.
Article in English | MEDLINE | ID: mdl-25108313

ABSTRACT

BACKGROUND: Cadmium (Cd) is a major environmental pollutant that causes multiple adverse health effects in humans and animals. In this study, we investigated Cd-mediated toxic effects in rats during pregnancy and endocrine intervention in the placenta. METHODS: We exposed pregnant rats to intraperitoneal Cd (CdCl2) at various doses (0, 0.25, and 0.5 mg/kg BW/day) from days 5 to 19 of pregnancy and evaluated the maternal-placental-fetal parameters linked to preeclampsia. We measured the corticosterone level in rat serum and placental tissue by sensitive ELISA and also analyzed the expression of glucocorticoid synthesis enzymes in the placenta. RESULTS: Key features of preeclampsia (PE), including hypertension, proteinuria, glomerular endotheliosis, placental abnormalities and small fetal size, appeared in pregnant rats after injection with 0.5 mg/kg BW/day Cd. The placental corticosterone production and maternal and fetal plasma corticosterone levels were increased in rats treated with 0.5 mg/kg BW/day Cd (P <0.01). The expression of 21-hydroxylase (CYP21) and 11beta-hydroxylase (CYP11B1), enzymes essential for corticosteroid synthesis, were increased in Cd-exposed placenta (P <0.01). 11beta-hydroxysteroid dehydrogenase (11beta-HSD2), a dominant negative regulator of local glucocorticoid levels, was decreased in Cd-exposed placenta (P <0.01). CONCLUSIONS: Our study demonstrates for the first time that changes in placental glucocorticoid synthesis induced by Cd exposure during pregnancy could contribute to preeclamptic conditions in rats.


Subject(s)
Cadmium Poisoning/physiopathology , Glucocorticoids/metabolism , Placenta/drug effects , Pre-Eclampsia/etiology , Pregnancy Complications/physiopathology , Up-Regulation/drug effects , 11-beta-Hydroxysteroid Dehydrogenase Type 2/antagonists & inhibitors , 11-beta-Hydroxysteroid Dehydrogenase Type 2/biosynthesis , Animals , Cadmium Chloride/administration & dosage , Cadmium Chloride/metabolism , Cadmium Chloride/pharmacokinetics , Cadmium Chloride/toxicity , Cadmium Poisoning/blood , Cadmium Poisoning/metabolism , Cadmium Poisoning/pathology , Corticosterone/blood , Corticosterone/metabolism , Dose-Response Relationship, Drug , Environmental Pollutants/administration & dosage , Environmental Pollutants/metabolism , Environmental Pollutants/pharmacokinetics , Environmental Pollutants/toxicity , Enzyme Induction/drug effects , Enzyme Repression/drug effects , Female , Glucocorticoids/blood , Injections, Intraperitoneal , Placenta/enzymology , Placenta/metabolism , Placenta/pathology , Pregnancy , Pregnancy Complications/blood , Pregnancy Complications/metabolism , Pregnancy Complications/pathology , Random Allocation , Rats, Sprague-Dawley , Steroid 11-beta-Hydroxylase/biosynthesis , Steroid 21-Hydroxylase/biosynthesis , Tissue Distribution
7.
Mol Reprod Dev ; 80(3): 204-11, 2013 Mar.
Article in English | MEDLINE | ID: mdl-23325624

ABSTRACT

Luteinizing hormone (LH) is known as a key regulator of corpus luteum (CL) function, but the luteoprotective mechanisms of LH in the maintenance of bovine CL function are not well understood. The current study investigated if LH increases cell viability and induces cortisol conversion, and if the luteoprotective action of LH is mediated by stimulating the local production and action of progesterone (P4) and/or cortisol. Cultured bovine luteal cells obtained at the mid-luteal stage (Days 8-12 of the estrous cycle) were treated for 24 hr with LH (10 ng/ml) with/without onapristone (OP, a specific P4 receptor antagonist; 100 µM), cortisone (1 µM), and aminoglutethimide (AGT, a specific inhibitor of cytochrome P450 side-chain cleavage; 100 µM). LH with and without OP significantly increased the mRNA and protein expressions of 11ß-hydroxysteroid dehydrogenase (HSD11B) 1, but did not affect the mRNA or protein expression of HSD11B2. These treatments also significantly increased HSD11B1 activity. Cell viability was significantly increased by LH alone or by LH in combination with cortisone and OP. LH in combination with OP or AGT significantly decreased cell viability as compared to LH alone. The overall results suggest that LH stimulates not only P4 production but also HSD11B1 expression, thereby increasing the cortisol concentration in the bovine CL, and that LH prevents cell death through these survival pathways. LH may consequently support CL function during the luteal phase in cattle.


Subject(s)
Corpus Luteum/drug effects , Corpus Luteum/metabolism , Glucocorticoids/metabolism , Luteinizing Hormone/pharmacology , Progesterone/biosynthesis , 11-beta-Hydroxysteroid Dehydrogenase Type 1/biosynthesis , 11-beta-Hydroxysteroid Dehydrogenase Type 1/genetics , 11-beta-Hydroxysteroid Dehydrogenase Type 1/metabolism , 11-beta-Hydroxysteroid Dehydrogenase Type 2/biosynthesis , 11-beta-Hydroxysteroid Dehydrogenase Type 2/genetics , 11-beta-Hydroxysteroid Dehydrogenase Type 2/metabolism , Animals , Caspase 3/genetics , Caspase 3/metabolism , Cattle , Cell Survival/drug effects , Cortisone/pharmacology , Female , Gene Expression Regulation/drug effects , Gonanes/pharmacology , Hormone Antagonists/pharmacology , Luteinizing Hormone/metabolism , Progesterone/metabolism
8.
Neurobiol Dis ; 43(2): 356-63, 2011 Aug.
Article in English | MEDLINE | ID: mdl-21530661

ABSTRACT

Glucocorticoids are used to treat respiratory dysfunction associated with premature birth but have been shown to cause neurodevelopmental deficits when used therapeutically. Recently, we established that acute glucocorticoid exposure at clinically relevant doses produces neural progenitor cell apoptosis in the external granule layer of the developing mouse cerebellum and permanent decreases in the number of cerebellar neurons. As the cerebellum naturally matures and neurogenesis is no longer needed, the external granule layer decreases proliferation and permanently disappears during the second week of life. At this same time, corticosterone (the endogenous rodent glucocorticoid) release increases and a glucocorticoid-metabolizing enzyme that protects the external granule layer against glucocorticoid receptor stimulation (11ß-Hydroxysteroid-Dehydrogenase-Type 2; HSD2) naturally disappears. Here we show that HSD2 inhibition and raising corticosterone to adult physiological levels both can independently increase neural progenitor cell apoptosis in the neonatal mouse. Conversely, glucocorticoid receptor antagonism decreases natural physiological apoptosis in this same progenitor cell population suggesting that endogenous glucocorticoid stimulation may regulate apoptosis in the external granule layer. We also found that glucocorticoids which HSD2 can effectively metabolize generate less external granule layer apoptosis than glucocorticoids this enzyme is ineffective at breaking down. This finding may explain why glucocorticoids that this enzyme can metabolize are clinically effective at treating respiratory dysfunction yet seem to produce no neurodevelopmental deficits. Finally, we demonstrate that both acute and chronic glucocorticoid exposures produce external granule layer apoptosis but without appropriate control groups this effect becomes masked. These results are discussed in terms of their implications for glucocorticoid therapy and neurodevelopment during the perinatal period.


Subject(s)
11-beta-Hydroxysteroid Dehydrogenase Type 2/metabolism , Apoptosis/physiology , Cerebellum/metabolism , Receptors, Glucocorticoid/metabolism , Stem Cells/metabolism , 11-beta-Hydroxysteroid Dehydrogenase Type 2/antagonists & inhibitors , 11-beta-Hydroxysteroid Dehydrogenase Type 2/biosynthesis , Animals , Animals, Newborn , Cell Differentiation/drug effects , Cell Differentiation/physiology , Cerebellum/cytology , Cerebellum/growth & development , Corticosterone/biosynthesis , Corticosterone/physiology , Mice , Mice, Inbred ICR , Neurogenesis/drug effects , Neurogenesis/physiology , Receptors, Glucocorticoid/antagonists & inhibitors , Receptors, Glucocorticoid/biosynthesis , Stem Cells/cytology
9.
Am J Physiol Heart Circ Physiol ; 301(1): H41-7, 2011 Jul.
Article in English | MEDLINE | ID: mdl-21536849

ABSTRACT

Experimental studies have suggested a role for the local renin-angiotensin-aldosterone system in the response to vascular injury. Clinical data support that aldosterone, via activation of the mineralocorticoid receptor (MR), is an important mediator of vascular damage in humans with cardiovascular disease. In mineralocorticoid-sensitive target tissue, aldosterone specificity for MR is conferred enzymatically by the cortisol-inactivating enzyme 11ß-hydroxysteroid-dehydrogenase-2 (11ßHSD2). However, the role of MR/aldosterone signaling in the venous system has not been explored. We hypothesized that MR expression and signaling in venous smooth muscle cells contributes to the arterialization of venous conduits and the injury response in vein bypass grafts. MR immunostaining was observed in all samples of excised human peripheral vein graft lesions and in explanted experimental rabbit carotid interposition vein grafts, with minimal staining in control greater saphenous vein. We also found upregulated transcriptional expression of both MR and 11ßHSD2 in human vein graft and rabbit vein graft, whereas control greater saphenous vein expressed minimal MR and no detectable 11ßHSD2. The expression of MR and 11ßHSD2 was confirmed in cultured human saphenous venous smooth muscle cells (hSVSMCs). Using an adenovirus containing a MR response element-driven reporter gene, we demonstrate that MR in hSVSMCs is capable of mediating aldosterone-induced gene activation. The functional significance for MR signaling in hSVSMCs is supported by the aldosterone-induced increase of angiotensin II type-1 receptor gene expression that was inhibited by the MR antagonist spironolactone. The upregulation of MR and 11ßHSD2 suggests that aldosterone-mediated tissue injury plays a role in vein graft arterialization.


Subject(s)
Aldosterone/physiology , Arteries/physiology , Myocytes, Smooth Muscle/physiology , Receptors, Mineralocorticoid/biosynthesis , Receptors, Mineralocorticoid/physiology , Signal Transduction/physiology , Veins/physiology , Veins/transplantation , 11-beta-Hydroxysteroid Dehydrogenase Type 2/biosynthesis , 11-beta-Hydroxysteroid Dehydrogenase Type 2/genetics , Aldosterone/pharmacology , Animals , Carotid Arteries/physiology , Gene Expression/physiology , HEK293 Cells , Humans , Immunohistochemistry , Myocytes, Smooth Muscle/metabolism , Rabbits , Receptor, Angiotensin, Type 1/biosynthesis , Receptors, Mineralocorticoid/genetics , Reverse Transcriptase Polymerase Chain Reaction , Saphenous Vein/cytology , Saphenous Vein/physiology , Signal Transduction/genetics , Up-Regulation/genetics , Up-Regulation/physiology
10.
J Neurosci Res ; 89(7): 1134-41, 2011 Jul.
Article in English | MEDLINE | ID: mdl-21416484

ABSTRACT

Negative early life experience may be associated with altered functioning of stress-related systems and may increase vulnerability to diseases later in life. Corticosteroids are important mediators of homeostasis and stress and exert their effects via two receptors, the mineralocorticoid receptor (MR) and the glucocorticoid receptor (GR), and through the glucocorticoid-metabolizing enzymes 11ß-hydroxysteroid dehydrogenase (11ß-HSD) types 1 and 2 in a brain-region-specific manner. However, relatively little is known about the postnatal ontogeny of these receptors and enzymes in the central nervous system. Here we describe, for the first time, the postnatal ontogeny of central GR, MR, 11ß-HSD1, and 11ß-HSD2 gene expression and monoamine levels in stress-related brain regions of domestic pigs at 7, 21, and 35 days of age. During the postnatal period, there was an increase in GR, MR, and 11ß-HSD1 mRNA expression in the pituitary and prefrontal cortex and an increase in MR mRNA expression in the hippocampus. We also demonstrated age-dependent changes in levels of noradrenaline and dopamine and their metabolites in the locus coeruleus, with the highest concentrations on day 7 compared with days 21 and 35. In conclusion, the dynamic changes in corticosteroid receptors and monoamines during neural development of postnatal pigs may represent periods of sensitivity to environmental stress that are comparable to some extent with those that are observed in primates and humans. Thus, these findings support the use of the domestic pig as an alternative animal model for humans in stress research.


Subject(s)
Aging/genetics , Biogenic Monoamines/metabolism , Brain/growth & development , Brain/metabolism , Gene Expression Regulation, Developmental/physiology , Receptors, Steroid/biosynthesis , 11-beta-Hydroxysteroid Dehydrogenase Type 1/biosynthesis , 11-beta-Hydroxysteroid Dehydrogenase Type 1/genetics , 11-beta-Hydroxysteroid Dehydrogenase Type 2/biosynthesis , 11-beta-Hydroxysteroid Dehydrogenase Type 2/genetics , Aging/physiology , Animals , Animals, Newborn , Brain/anatomy & histology , Female , Hippocampus/enzymology , Hippocampus/metabolism , Locus Coeruleus/enzymology , Locus Coeruleus/metabolism , Male , Pituitary Gland/enzymology , Pituitary Gland/metabolism , Prefrontal Cortex/enzymology , Prefrontal Cortex/metabolism , Receptors, Steroid/genetics , Species Specificity , Sus scrofa
11.
Biochem Biophys Res Commun ; 391(4): 1752-6, 2010 Jan 22.
Article in English | MEDLINE | ID: mdl-20045394

ABSTRACT

Two isoforms of 11beta-hydroxysteroid dehydrogenase (11beta-HSD1 and 11beta-HSD2) play an important role in regulation of glucocorticoid corticosterone (CORT, the active form in rodents) by the interconversion between CORT and 11-dehydrocorticosterone (11DHC, the biologically inert form). 11beta-HSD1 is an NADP+/NADPH-dependent oxidoreductase which is mainly expressed in liver and kidney, while 11beta-HSD2 is an NAD+-dependent oxidase which is predominantly expressed in kidney. The regulation of 11beta-HSD1 and 11beta-HSD2 mRNA (Hsd11b1 and Hsd11b2) levels and their activities by IGF-1 was performed in liver, kidney, and testis of IGF-1 knockout male mice. Real-time PCR showed that Hsd11b1 in liver was decreased while Hsd11b2 mRNA level was decreased in kidney of IGF-1 null mice. 11beta-HSD1 and 11beta-HSD2 activities fluctuated with the changes of their respective Hsd11b1 or Hsd11b2 mRNA levels. In conclusion, IGF-I tissue-specifically regulates Hsd11b1 and Hsd11b2 expression.


Subject(s)
11-beta-Hydroxysteroid Dehydrogenase Type 1/biosynthesis , 11-beta-Hydroxysteroid Dehydrogenase Type 2/biosynthesis , Insulin-Like Growth Factor I/physiology , Kidney/enzymology , Liver/enzymology , Testis/enzymology , 11-beta-Hydroxysteroid Dehydrogenase Type 1/genetics , 11-beta-Hydroxysteroid Dehydrogenase Type 2/genetics , Animals , Insulin-Like Growth Factor I/genetics , Male , Mice , Mice, Knockout , RNA, Messenger/biosynthesis , RNA, Messenger/genetics
12.
Brain Behav Immun ; 24(6): 975-84, 2010 Aug.
Article in English | MEDLINE | ID: mdl-20385225

ABSTRACT

Relapses during multiple sclerosis (MS) are treated by administration of exogenous corticosteroids. However, little is known about the bioavailability of endogenous steroids in the central nervous system (CNS) of MS patients. We thus determined cortisol and dehydroepiandrosterone (DHEA) levels in serum and cerebrospinal fluid (CSF) samples from 34 MS patients, 28 patients with non-inflammatory neurological diseases (NIND) and 16 patients with other inflammatory neurological diseases (OIND). This revealed that MS patients - in sharp contrast to patients with OIND - show normal cortisol concentrations in serum and lowered cortisol levels in the CSF during acute relapses. This local cortisol deficit may relate to poor local activation of cortisone via 11beta-hydroxysteroid dehydrogenase type 1 (11bHSD1) or to inactivation via 11bHSD2. Accordingly, 11bHSD2 was found to be expressed within active plaques, whereas 11bHSD1 was predominantly detected in surrounding "foamy" macrophages. Our study thus provides new insights into the impaired endogenous CNS cortisol regulation in MS patients and its possible relation to MS lesion pathogenesis. Moreover, an observed upregulation of 11bHSD1 in myelin-loaded macrophages in vitro suggests an intriguing hypothesis for the self-limiting nature of MS lesion development. Finally, our findings provide an attractive explanation for the effectivity of high- vs. low-dose exogenous corticosteroids in the therapy of acute relapses.


Subject(s)
11-beta-Hydroxysteroid Dehydrogenases/cerebrospinal fluid , Hydrocortisone/cerebrospinal fluid , Multiple Sclerosis/cerebrospinal fluid , Multiple Sclerosis/pathology , 11-beta-Hydroxysteroid Dehydrogenase Type 1/biosynthesis , 11-beta-Hydroxysteroid Dehydrogenase Type 1/cerebrospinal fluid , 11-beta-Hydroxysteroid Dehydrogenase Type 2/biosynthesis , 11-beta-Hydroxysteroid Dehydrogenase Type 2/cerebrospinal fluid , Adult , Brain/enzymology , Cell Count , Dehydroepiandrosterone/blood , Dehydroepiandrosterone/cerebrospinal fluid , Female , Foam Cells/physiology , Gene Expression/physiology , Humans , Hydrocortisone/blood , Immunohistochemistry , Macrophages/enzymology , Male , Multiple Sclerosis/enzymology , Myelin Proteins/physiology , Reverse Transcriptase Polymerase Chain Reaction
13.
Am J Physiol Renal Physiol ; 297(5): F1381-90, 2009 Nov.
Article in English | MEDLINE | ID: mdl-19710242

ABSTRACT

Previous studies have shown that mineralocorticoid receptor (MCR) blocker reduces proteinuria in diabetic nephropathy (DN), but the role of aldosterone in podocyte injury has never been explored in DN. This study was undertaken to elucidate whether a local aldosterone system existed in podocytes and to examine its role in podocyte apoptosis under diabetic conditions. In vitro, immortalized podocytes were exposed to 5.6 mM glucose (NG), NG + 24.4 mM mannitol, and 30 mM glucose (HG) with or without 10(-7) M spironolactone (SPR). In vivo, 32 Sprague-Dawley rats were injected with diluent (C, n = 16) or streptozotocin intraperitoneally [diabetes mellitus (DM), n = 16], and 8 rats from each group were treated with SPR for 3 mo. Aldosterone synthase (CYP11B2) and MCR mRNA and protein expression were determined by real-time PCR and Western blot, respectively, and aldosterone levels by radioimmunoassay. Western blot for apoptosis-related molecules, Hoechst 33342 staining, and terminal deoxynucleotidyl transferase-mediated dUTP nick end labeling (TUNEL) assay were performed to determine apoptosis. CYP11B2 and MCR expression were significantly higher in HG-stimulated podocytes and DM glomeruli compared with NG cells and C glomeruli, respectively, along with increased aldosterone levels. Western blot analysis revealed that cleaved caspase-3 and Bax expression was significantly increased, whereas Bcl-2 expression was significantly decreased in HG-stimulated podocytes and in DM glomeruli. Apoptosis determined by Hoechst 33342 staining and TUNEL assay were also significantly increased in podocytes under diabetic conditions. These changes in the expression of apoptosis-related proteins and the increase in apoptotic cells were inhibited by SPR treatment. These findings suggest that a local aldosterone system is activated and is involved in podocyte apoptosis under diabetic conditions.


Subject(s)
Aldosterone/physiology , Apoptosis/physiology , Diabetes Mellitus, Experimental/pathology , Podocytes/physiology , 11-beta-Hydroxysteroid Dehydrogenase Type 2/biosynthesis , 11-beta-Hydroxysteroid Dehydrogenase Type 2/genetics , Animals , Apoptosis Regulatory Proteins/biosynthesis , Apoptosis Regulatory Proteins/genetics , Benzimidazoles , Blotting, Western , Caspase 3/biosynthesis , Caspase 3/genetics , Cell Count , Cells, Cultured , Cytochrome P-450 CYP11B2/biosynthesis , Cytochrome P-450 CYP11B2/genetics , Fluorescent Antibody Technique , Fluorescent Dyes , In Situ Nick-End Labeling , Kidney/pathology , Kidney Glomerulus/metabolism , Male , Rats , Rats, Sprague-Dawley , Receptors, Mineralocorticoid/biosynthesis , Receptors, Mineralocorticoid/genetics , Reverse Transcriptase Polymerase Chain Reaction
14.
Physiol Genomics ; 32(3): 343-51, 2008 Feb 19.
Article in English | MEDLINE | ID: mdl-18073273

ABSTRACT

11beta-Hydroxysteroid dehydrogenase type 1 (11beta-HSD1), which regulates the local level of glucocorticoids, has been suggested to be involved in the development of obesity. A definitive functional role for 11beta-HSD1 in adipogenesis, however, remains to be established. We developed 3T3-L1 cell lines stably transfected with a small hairpin RNA (shRNA) targeting 11beta-HSD1. A shRNA containing two nucleotide substitutions was used as a control. Silencing of 11beta-HSD1 substantially attenuated the accumulation of lipid droplets and the expression of adipogenesis marker genes, which was induced by a mixture containing either corticosterone or dexamethasone. Silencing of 11beta-HSD1 increased the concentration of 11-dehydrocorticosterone in the culture supernatant but did not significantly affect the levels of corticosterone or dexamethasone. Translocation of glucocorticoid receptors to the nucleus in response to glucocorticoids was significantly attenuated by silencing 11beta-HSD1. The number of cells entering the S phase of the cell cycle following the induction of adipogenesis was significantly reduced by silencing 11beta-HSD1. 11beta-HSD1 shRNA delivered by lentiviral vectors after the induction of differentiation, however, did not affect the progression of adipogenesis. These results indicate that 11beta-HSD1 plays a significant functional role in the initiation of 3T3-L1 adipogenesis and provide new mechanistic insights into the role of 11beta-HSD1 in the development of obesity and related diseases.


Subject(s)
11-beta-Hydroxysteroid Dehydrogenase Type 1/antagonists & inhibitors , 3T3-L1 Cells/metabolism , Adipogenesis/genetics , RNA Interference , 11-beta-Hydroxysteroid Dehydrogenase Type 1/genetics , 11-beta-Hydroxysteroid Dehydrogenase Type 1/physiology , 11-beta-Hydroxysteroid Dehydrogenase Type 2/biosynthesis , 11-beta-Hydroxysteroid Dehydrogenase Type 2/genetics , 3T3-L1 Cells/drug effects , Adipogenesis/drug effects , Animals , Base Sequence , CCAAT-Enhancer-Binding Protein-alpha/biosynthesis , CCAAT-Enhancer-Binding Protein-alpha/genetics , Corticosterone/analogs & derivatives , Corticosterone/biosynthesis , Corticosterone/metabolism , Corticosterone/pharmacology , Dexamethasone/analogs & derivatives , Dexamethasone/metabolism , Dexamethasone/pharmacology , Enzyme Induction/drug effects , Gene Expression Regulation , Genetic Vectors/genetics , Lentivirus/genetics , Mice , Molecular Sequence Data , PPAR gamma/biosynthesis , PPAR gamma/genetics , RNA, Small Interfering/genetics , Signal Transduction/drug effects , Transduction, Genetic
15.
Mol Reprod Dev ; 75(5): 925-30, 2008 May.
Article in English | MEDLINE | ID: mdl-18033680

ABSTRACT

The objective of the present study was to determine whether glucocorticoid (GC) and its receptor (GC-R) are expressed in the porcine corpus luteum (CL), and whether GC influences porcine luteal hormone production. The gene expressions of 11beta-hydroxysteroid dehydrogenase type 1 (11-HSD1), type 2 (11-HSD2), GC-R, and the concentrations of GC were determined in the CL of Chinese Meishan pigs during the estrous cycle. Moreover, the effects of GC on progesterone (P(4)), estradiol-17beta (E(2)), and prostaglandin (PG) F2alpha secretion by cultured luteal cells were investigated. Messenger RNAs of the 11-HSD1, 11-HSD2, and GC-R were clearly expressed in the CL throughout the estrous cycle. The 11-HSD1 mRNA level in the CL was higher at the regressed stage than at the other stages (P < 0.05), whereas 11-HSD2 mRNA was lower at the regressed stage than at the other stages (P < 0.05). GC-R mRNA level was higher at the regressed stages than at the other stages (P < 0.01). Concentrations of GC were lower in the regressed CL than in the other stages (P < 0.01). When the cultured luteal cells obtained from mid-stage CL (Days 8-11) were exposed to GC (50-5,000 ng/ml), P(4) and PGF2alpha secretion by the cells were reduced (P < 0.05), whereas GC had no effect on E(2) secretion by the cells. The overall results suggest that GC is regulated locally by 11-HSD1 and 11-HSD2 in the porcine CL. GC inhibits P(4) and PGF2alpha production from luteal cells via their specific receptors, implying GC plays some roles in regulating porcine CL function throughout the estrous cycle.


Subject(s)
11-beta-Hydroxysteroid Dehydrogenase Type 1/biosynthesis , 11-beta-Hydroxysteroid Dehydrogenase Type 2/biosynthesis , Corpus Luteum Hormones/biosynthesis , Corpus Luteum/metabolism , Estrous Cycle/metabolism , Receptors, Glucocorticoid/biosynthesis , Animals , Corpus Luteum/cytology , Female , RNA, Messenger/biosynthesis , Swine
16.
J Antibiot (Tokyo) ; 61(3): 128-35, 2008 Mar.
Article in English | MEDLINE | ID: mdl-18503190

ABSTRACT

The novel 11beta-hydroxysteroid dehydrogenase type 1 (11beta-HSD1) inhibitors known as sterenin A, B, C and D were found in a solid-state culture of the producing basidiomycetes identified as Stereum sp. SANK 21205. Purification of the 50% aq Me(2)CO extract of the culture was performed by EtOAc extraction, reversed phase open-column chromatography and successive ODS HPLC preparation. These compounds, whose structures were determined by several spectroscopic methods, were found to be novel isoindolinone alkaloids which exhibited potent selective inhibitory activities against 11beta-HSD1.


Subject(s)
11-beta-Hydroxysteroid Dehydrogenase Type 1/antagonists & inhibitors , Basidiomycota/chemistry , Enzyme Inhibitors/pharmacology , Indoles/pharmacology , 11-beta-Hydroxysteroid Dehydrogenase Type 1/biosynthesis , 11-beta-Hydroxysteroid Dehydrogenase Type 2/antagonists & inhibitors , 11-beta-Hydroxysteroid Dehydrogenase Type 2/biosynthesis , Animals , Chemical Phenomena , Chemistry, Physical , Dose-Response Relationship, Drug , Fermentation , Humans , Indoles/chemistry , Kinetics , Magnetic Resonance Spectroscopy , Mice , Microsomes, Liver/drug effects , Microsomes, Liver/enzymology , Reverse Transcriptase Polymerase Chain Reaction , Spectrometry, Mass, Fast Atom Bombardment , Spores, Fungal/enzymology
17.
Immunol Lett ; 109(1): 72-5, 2007 Mar 15.
Article in English | MEDLINE | ID: mdl-17320974

ABSTRACT

Regulation of inflammation in leprosy may be influenced by local concentrations of active cortisol and inactive cortisone, whose concentrations are regulated by enzymes in the cortisol-cortisone shuttle. We investigated the cortisol-cortisone shuttle enzymes in the skin of leprosy patients with type 1 reactions (T1R), which are characterised by skin and nerve inflammation. Gene expression of the shuttle enzymes were quantified in skin biopsies from 15 leprosy patients with new T1R before and during prednisolone treatment and compared with levels in skin biopsies from 10 borderline leprosy patients without reactions. Gene expression of 11beta-hydroxysteroid dehydrogenase (11beta-HSD) type 2, which converts cortisol to cortisone, is down-regulated in skin from T1R lesions. However expression levels of 11beta-HSD type 1, which converts cortisone to cortisol, were similar in skin with and without reactions and did not change during anti-leprosy drug treatment. Prednisolone treatment of patients with reactions is associated with an upregulation of 11beta-HSD2 expression in skin. The down regulation of 11beta-HSD2 at the beginning of a reaction may be caused by pro-inflammatory cytokines in the leprosy reactional lesion and may be a local attempt to down-regulate inflammation. However in leprosy reactions this local response is insufficient and exogenous steroids are required to control inflammation.


Subject(s)
Cortisone/metabolism , Hydrocortisone/metabolism , 11-beta-Hydroxysteroid Dehydrogenase Type 1/biosynthesis , 11-beta-Hydroxysteroid Dehydrogenase Type 1/genetics , 11-beta-Hydroxysteroid Dehydrogenase Type 1/metabolism , 11-beta-Hydroxysteroid Dehydrogenase Type 2/biosynthesis , 11-beta-Hydroxysteroid Dehydrogenase Type 2/genetics , 11-beta-Hydroxysteroid Dehydrogenase Type 2/metabolism , Cortisone/immunology , Gene Expression , Humans , Hydrocortisone/immunology , India , Leprosy, Borderline/genetics , Leprosy, Borderline/immunology , Leprosy, Borderline/metabolism , Leprosy, Borderline/microbiology , Prednisolone/immunology
18.
Metabolism ; 56(10): 1326-33, 2007 Oct.
Article in English | MEDLINE | ID: mdl-17884440

ABSTRACT

In diabetes, dysregulation of the hypothalamic-pituitary-adrenocortical (HPA) axis causes effects such as elevation of corticotropin (ACTH) and glucocorticoids. Cholecystokinin and its receptors are involved in the HPA axis and influence the regulation of the HPA axis. We examined adrenocortical function in Otsuka Long-Evans Tokushima Fatty (OLETF) rats, a model of type 2 diabetes mellitus, that lack the cholecystokinin A receptor. We measured adrenal weight, plasma ACTH, serum and urinary corticosterone, and serum leptin in OLETF rats at 5 to 36 weeks of age. Messenger RNA (mRNA) expression of 11beta-hydroxysteroid dehydrogenase and 5alpha-reductase type 1 in adrenal glands of the rats were examined. Long-Evans Tokushima Otsuka (LETO) rats were used as controls. In OLETF rats at 32 to 36 weeks of age, plasma ACTH was significantly higher (P < .001); serum corticosterone and 24-hour urinary corticosterone were significantly lower (P < .005); and adrenal weight was significantly lower (P < .005) than those in LETO rats. At the same ages, serum leptin in OLETF rats was significantly higher (P < .001) than that in LETO rats. In the younger OLETF rats, these changes were not observed. Overall, there was an inverse correlation between serum corticosterone and serum leptin (r = -0.374, P < .0005), whereas there was a positive correlation between plasma ACTH and serum leptin (r = 0.654, P < .0001). At 5 and 36 weeks of age, mRNA expression of 5alpha-reductase type 1 in the adrenal gland of OLETF rats was significantly higher (P < .05) than that of LETO rats, whereas there was no significant difference in mRNA expressions of 11beta-hydroxysteroid dehydrogenase types 1 and 2. We showed that adrenocortical insufficiency and adrenal atrophy were acquired in OLETF rats, and the possibility of elevated serum leptin relates to this phenomenon.


Subject(s)
Adrenal Insufficiency/physiopathology , Diabetes Mellitus, Type 2/physiopathology , 11-beta-Hydroxysteroid Dehydrogenase Type 1/biosynthesis , 11-beta-Hydroxysteroid Dehydrogenase Type 1/genetics , 11-beta-Hydroxysteroid Dehydrogenase Type 2/biosynthesis , 11-beta-Hydroxysteroid Dehydrogenase Type 2/genetics , 3-Oxo-5-alpha-Steroid 4-Dehydrogenase/biosynthesis , 3-Oxo-5-alpha-Steroid 4-Dehydrogenase/genetics , Adrenal Cortex/pathology , Adrenal Cortex Function Tests , Adrenal Glands/metabolism , Adrenal Insufficiency/metabolism , Adrenal Insufficiency/pathology , Adrenocorticotropic Hormone/blood , Aging/physiology , Animals , Blood Glucose/metabolism , Body Weight/physiology , Corticosterone/blood , Corticosterone/urine , DNA Primers , Diabetes Mellitus, Type 2/metabolism , Insulin/blood , Leptin/blood , Organ Size/physiology , RNA, Messenger/biosynthesis , Rats , Rats, Inbred OLETF , Reverse Transcriptase Polymerase Chain Reaction
19.
Am J Obstet Gynecol ; 197(1): 43.e1-6, 2007 Jul.
Article in English | MEDLINE | ID: mdl-17618751

ABSTRACT

OBJECTIVE: The enzyme 11beta-hydroxysteroid dehydrogenase type 2 (11beta-HSD2) converts cortisol into cortisone. Reduced placental activity of 11beta-HSD2 in small-for-gestational-age (SGA) neonates results in fetal cortisol excess. In the present study, we examined the yet unknown gene expression of 11beta-HSD1, which primarily synthesizes cortisol in SGA placenta. STUDY DESIGN: In placentas taken from 24 women with normal-weight newborns and 16 women with SGA neonates, expression of 11beta-HSD1 and 11beta-HSD2 messenger ribonucleic acid (mRNA) was determined using reverse transcription-polymerase chain reaction. RESULTS: Placental mRNA expression of 11beta-HSD1 and 11beta-HSD2 was significantly reduced in the SGA group (P = .006 and P < .0001). Both enzymes showed a significant correlation to birthweight SD score and placental weight. Also, levels of both enzymes were significantly correlated. CONCLUSION: In placental tissue of SGA neonates 11beta-HSD2 and 11beta-HSD1 gene expression is reduced. Adapted levels of 11beta-HSD1 might result in a counterregulatory mechanism limiting transplacental passage of elevated cortisol levels.


Subject(s)
11-beta-Hydroxysteroid Dehydrogenase Type 1/genetics , 11-beta-Hydroxysteroid Dehydrogenase Type 2/genetics , Placenta/enzymology , 11-beta-Hydroxysteroid Dehydrogenase Type 1/biosynthesis , 11-beta-Hydroxysteroid Dehydrogenase Type 2/biosynthesis , Adult , Case-Control Studies , Female , Gene Expression , Gestational Age , Humans , Hydrocortisone/metabolism , Infant, Low Birth Weight/physiology , Infant, Newborn , Infant, Small for Gestational Age/physiology , Pregnancy
20.
Circ Res ; 96(6): 643-50, 2005 Apr 01.
Article in English | MEDLINE | ID: mdl-15718497

ABSTRACT

Inhibition or blockade of the angiotensin-aldosterone system consistently decreases ischemic cardiovascular events in clinical trials. The steroid hormone aldosterone acts by binding to the mineralocorticoid receptor (MR), a ligand activated transcription factor that is a member of the nuclear hormone receptor superfamily. MR binds and is activated by aldosterone and cortisol with equal affinity, but MR activation by cortisol is diminished in tissues that express the cortisol-inactivating enzyme 11-beta-hydroxysteroid-dehydrogenase-2 (11betaHSD2). Although previous studies support that the vasculature is a target tissue of aldosterone, MR-mediated gene expression in vascular cells has not been demonstrated or systematically explored. We investigated whether functional MR and 11betaHSD2 are expressed in human blood vessels. Human coronary and aortic vascular smooth muscle cells (VSMCs) express mRNA and protein for both MR and 11betaHSD2. The endogenous VSMC MR mediates aldosterone-dependent gene expression, which is blocked by the competitive MR antagonist spironolactone. Inhibition of 11betaHSD2 in coronary artery VSMCs enhances gene transactivation by cortisol, supporting that the VSMC 11betaHSD2 is functional. Angiotensin II also activates MR-mediated gene transcription in coronary artery VSMCs. Angiotensin II activation of MR-mediated gene expression is inhibited by both the AT1 receptor blocker losartan and by spironolactone, but not by aldosterone synthase inhibition. Microarray and quantitative RT-PCR experiments show that aldosterone activates expression of endogenous human coronary VSMC genes, including several involved in vascular fibrosis, inflammation, and calcification. These data support a new MR-dependent mechanism by which aldosterone and angiotensin II influence ischemic cardiovascular events, and suggest that ACE inhibitors and MR antagonists may decrease clinical ischemic events by inhibiting MR-dependent gene expression in vascular cells.


Subject(s)
Aldosterone/physiology , Angiotensin II/physiology , Gene Expression Regulation/physiology , Myocytes, Smooth Muscle/drug effects , Receptors, Mineralocorticoid/physiology , Transcription, Genetic/physiology , 11-beta-Hydroxysteroid Dehydrogenase Type 2/biosynthesis , 11-beta-Hydroxysteroid Dehydrogenase Type 2/genetics , Adenoviridae/genetics , Aldosterone/pharmacology , Angiotensin II/pharmacology , Angiotensin II Type 1 Receptor Blockers/pharmacology , Aorta/cytology , Cell Line, Transformed/drug effects , Cell Line, Transformed/metabolism , Coronary Vessels/cytology , Gene Expression Profiling , Gene Expression Regulation/drug effects , Genes, Reporter , Genetic Vectors/genetics , Humans , Hydrocortisone/pharmacology , Losartan/pharmacology , Mammary Tumor Virus, Mouse/genetics , Models, Biological , Muscle, Smooth, Vascular/drug effects , Muscle, Smooth, Vascular/metabolism , Myocardial Ischemia/drug therapy , Myocardial Ischemia/physiopathology , Myocytes, Smooth Muscle/metabolism , Oligonucleotide Array Sequence Analysis , Promoter Regions, Genetic/drug effects , Receptor, Angiotensin, Type 1/physiology , Receptors, Mineralocorticoid/biosynthesis , Receptors, Mineralocorticoid/drug effects , Receptors, Mineralocorticoid/genetics , Reverse Transcriptase Polymerase Chain Reaction , Signal Transduction/physiology , Spironolactone/pharmacology , Transcription, Genetic/drug effects , Transduction, Genetic
SELECTION OF CITATIONS
SEARCH DETAIL