Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 119
Filter
1.
Exp Clin Endocrinol Diabetes ; 117(9): 514-8, 2009 Oct.
Article in English | MEDLINE | ID: mdl-19235128

ABSTRACT

The enzyme 11beta-hydroxysteroid dehydrogenase type 1 (11beta-HSD1) catalyzes the interconversion between inactive 11-ketoglucocorticoids and their active 11beta-hydroxy derivatives, such as cortisol and corticosterone. We have investigated the expression of 11beta-HSD1 in freshly isolated human peripheral mononuclear leukocytes (MNL). The presence of 11beta-HSD1 mRNA was demonstrated in total RNA by RT-PCR using specific primers designed on the 4th and 5th exons of the human 11beta-HSD1 gene. Fragments of the expected size were consistently detected on agarose gels, and sequencing showed complete identity with the corresponding sequence deposited in GenBank. The occurrence of 11beta-HSD1 protein was established by Western immunoblot analysis with a specific polyclonal antibody. Enzyme oxo-reductase activity was investigated by incubating 12 samples of MNL isolated from from 8 subjects with [3H]cortisone and formation of cortisol was established only in 4 subjects (yield range: 0.15-1.3%) after acetylation and TLC, blank subtraction and correction for losses. 18beta-Glycyrrhetinic acid, an inhibitor of 11 beta-HSD1, reduced cortisol production below detection limit. Dehydrogenase activity could not be demonstrated. It is suggested that, although enzyme activity of 11beta-HSD1 in circulating MNL is low, it is apparently ready for enhancement after MNL migration to sites of inflammation.


Subject(s)
11-beta-Hydroxysteroid Dehydrogenase Type 1/metabolism , Leukocytes, Mononuclear/metabolism , 11-beta-Hydroxysteroid Dehydrogenase Type 1/genetics , Adult , Blotting, Western , Cortisone/metabolism , Female , Humans , Hydrocortisone/biosynthesis , Limit of Detection , Male , RNA, Messenger/genetics , RNA, Messenger/metabolism , Reverse Transcriptase Polymerase Chain Reaction
2.
Endocr Res ; 29(2): 211-6, 2003 May.
Article in English | MEDLINE | ID: mdl-12856808

ABSTRACT

The regulation of 11beta-hydroxysteroid dehydrogenase type II (11betaHSD2) expression at the level of specific mRNA and 11betaHSD2 protein was investigated in primary culture of renal epithelial cells of the rat. It has been shown that treatment of the SE cells with adenylyl cyclase activator, forskolin, known to stimulate the protein kinase A (PKA) pathway, resulted in an increase in 11betaHSD2 mRNA content in these cells. Semi-quantitative RT-PCR revealed that the effect of forskolin was attenuated by the addition of phorbol ester, tetradecanoyl phorbol acetate (TPA), an activator of the protein kinase C (PKC) pathway, whereas TPA on its own slightly reduced the basal level of 11betaHSD2 expression judging from the content of specific mRNA. Measurements of [35S]-methionine incorporation into immunoprecipitable 11betaHSD2 revealed an increased synthesis of this protein in renal epithelial cells treated with forskolin. Phorbol ester TPA markedly reduced the effect of forskolin on the synthesis of 11betaHSD2 and attenuated the basal level of synthesis of this protein. It is concluded that in renal epithelial cells in primary culture, stimulation of PKA pathway results in the induction of 11betaHSD2 both at a specific mRNA and at a protein level and that this effect is markedly reduced by activation of PKC pathway.


Subject(s)
11-beta-Hydroxysteroid Dehydrogenase Type 2/metabolism , Cyclic AMP-Dependent Protein Kinases/metabolism , Epithelial Cells/enzymology , Kidney/enzymology , Protein Kinase C/metabolism , Signal Transduction/physiology , 11-beta-Hydroxysteroid Dehydrogenase Type 2/drug effects , 11-beta-Hydroxysteroid Dehydrogenase Type 2/genetics , Animals , Colforsin/pharmacology , Cyclic AMP-Dependent Protein Kinase Type II , Enzyme Activators/pharmacology , Epithelial Cells/drug effects , Female , Gene Expression Regulation , Kidney/cytology , Kidney/drug effects , RNA, Messenger/analysis , Rats , Rats, Wistar , Tetradecanoylphorbol Acetate/pharmacology
3.
Mol Cell Endocrinol ; 183(1-2): 165-70, 2001 Oct 25.
Article in English | MEDLINE | ID: mdl-11604236

ABSTRACT

An important determinant of the potency of steroid hormones is the presence of activating and inactivating enzymes in target cells. The 11 beta-hydroxysteroid dehydrogenase type 1 and type 2 enzymes (11 beta HSD1 and 11 beta HSD2) modulate glucocorticoid action and may be important in regulating cellular growth. In the present study we examined 11 beta-hydroxysteroid dehydrogenase in Ishikawa endometrial cancer cells to see if modulation of enzyme activity could potentiate the antiproliferative effects of glucocorticoids. Ishikawa cells contain an NAD dependent enzyme migrating at 41 kDa on Western blots, consistent with the presence of the glucocorticoid-inactivating enzyme 11 beta HSD2, while the NADP dependent 11 beta HSD1 is barely detectable. Given that glucocorticoids decrease cellular proliferation we asked whether inhibition of 11 beta HSD2 could further enhance this effect. Cultivation of cells in the presence of 1 microM cortisol resulted in an elevation of 11 beta HSD2 and this was associated with a decrease in cell number. Enzyme activity and cell proliferation showed a biphasic response to the synthetic anti-progestin and anti-glucocorticoid RU38486, with < or =10 nM exerting agonistic effects and > or =100 nM producing antagonist effects in the presence of 1 microM cortisol. Inhibition of 11 beta HSD2 activity by glycyrrhetinic acid did not enhance the anti-proliferative effects of 1 microM cortisol, but the inhibitor showed significant antiproliferative activity in the absence of added glucocorticoid, consistent with protection of the low levels of glucocorticoids present in culture medium. Interestingly, the commonly used 11 beta HSD inhibitor, Carbenoxolone, did not block 11 beta HSD2 activity in whole Ishikawa cells, and there was no effect on cell proliferation, however, complete inhibition of 11 beta HSD2 was achieved in cellular homogenates suggesting that a barrier exists to entry of the inhibitor into intact cells. This study suggests that inhibition of 11 beta HSD2 activity can enhance the antiproliferative effects of low, but not high concentrations of glucocorticoids, and that beneficial effects may be attained in vivo at the nadir of diurnal glucocorticoid levels.


Subject(s)
Cell Division/physiology , Hydroxysteroid Dehydrogenases/metabolism , 11-beta-Hydroxysteroid Dehydrogenase Type 2 , Administration, Topical , Anti-Inflammatory Agents/pharmacology , Carbenoxolone/pharmacology , Cell Division/drug effects , Endometrium/cytology , Endometrium/metabolism , Enzyme Inhibitors/pharmacology , Female , Glycyrrhetinic Acid/pharmacology , Hormone Antagonists/pharmacology , Humans , Hydrocortisone/pharmacology , Hydroxysteroid Dehydrogenases/antagonists & inhibitors , Isoenzymes/metabolism , Mifepristone/pharmacology , Tumor Cells, Cultured
4.
J Steroid Biochem Mol Biol ; 76(1-5): 153-9, 2001.
Article in English | MEDLINE | ID: mdl-11384873

ABSTRACT

Manipulating the metabolism of glucocorticoids may serve as a useful adjunct in the treatment of breast cancer. The 11beta-hydroxysteroid dehydrogenase type 2 enzyme (11betaHSD2) potently inactivates glucocorticoids thereby protecting the non-selective mineralocorticoid receptor (MR) in fluid transporting tissues. In the present study, Western blot analysis showed the presence of 11betaHSD2 in 66% of the breast tumor samples. The 11betaHSD2 and MR are also present in the breast tumor cell line PMC42. Glycyrrhetinic acid abolished glucocorticoid metabolism and inhibited cell growth by 40%, the latter at concentrations consistent with glucocorticoid receptor (GR) and MR binding studies. Metabolism was increased by glucocorticoids, the anti-glucocorticoid RU 38486 and anti-mineralocorticoid spironolactone, while aldosterone had no effect. Neither cortisol nor aldosterone affected cell proliferation, but both RU 38486 and spironolactone caused a significant decrease in cell number. The effects of RU 38486 were only observed at micromolar concentrations and are inconsistent with an action via GR or progesterone receptor (PR). This study shows that 11betaHSD2 activity and cell proliferation of PMC42 cells can be modulated via steroid receptors.


Subject(s)
Breast Neoplasms/enzymology , Cell Division , Hydroxysteroid Dehydrogenases/metabolism , 11-beta-Hydroxysteroid Dehydrogenase Type 2 , Aldosterone/pharmacology , Blotting, Western , Breast Neoplasms/pathology , Cell Division/drug effects , Cell Line , Humans , Mifepristone/pharmacology , Mineralocorticoid Receptor Antagonists , Receptors, Glucocorticoid/agonists , Receptors, Glucocorticoid/antagonists & inhibitors , Receptors, Mineralocorticoid/agonists , Spironolactone/pharmacology
5.
Exp Lung Res ; 27(2): 143-55, 2001 Mar.
Article in English | MEDLINE | ID: mdl-11258802

ABSTRACT

We examined whether transalveolar fluid transport is modulated by aldosterone in adult rats. Because colocalization of mineralocorticoid receptors (MR) with 11beta-hydroxysteroid dehydrogenase type 2 (11betaHSD2) is important for aldosterone specific action, we first determined the immunohistochemical distribution of MR and 11betaHSD2 in the lung. We found that alveolar epithelial cells express both MR and 11betaHSD2. Reverse transcriptase polymerase chain reaction (RT-PCR) demonstrated that rat alveolar type II epithelial cells express both MR and 11betaHSD2. We then measured alveolar fluid clearance in rats treated with chronic low-sodium diet. A low-sodium diet (0.1% NaCl for 12 to 14 days) caused hyperaldosteronism accompanied by hypokalemia, whereas serum corticosterone and adrenaline levels remained normal. We found that hyperaldosteronism was associated with significantly higher alveolar fluid clearance and that this increase was related to the amiloride-sensitive component. In addition, the increase in alveolar fluid clearance was inhibited by spironolactone. Our results show that aldosterone is able to stimulate Na+ channels of alveolar epithelial cells. We conclude that alveolar epithelium is a physiological target tissue for aldosterone and transalveolar fluid absorption could in part be modulated by endogenous aldosterone acting via MR.


Subject(s)
Aldosterone/physiology , Extravascular Lung Water/metabolism , Pulmonary Alveoli/metabolism , 11-beta-Hydroxysteroid Dehydrogenase Type 2 , Absorption , Animals , Diet, Sodium-Restricted , Epithelial Cells/cytology , Epithelial Cells/drug effects , Epithelial Cells/metabolism , Hydroxysteroid Dehydrogenases/genetics , Hydroxysteroid Dehydrogenases/metabolism , Hyperaldosteronism/etiology , Hyperaldosteronism/metabolism , In Vitro Techniques , Male , Pulmonary Alveoli/cytology , RNA/isolation & purification , RNA, Messenger/metabolism , Rats , Rats, Sprague-Dawley , Receptors, Mineralocorticoid/genetics , Receptors, Mineralocorticoid/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Sodium Channels/metabolism , Specific Pathogen-Free Organisms
6.
Endocrinology ; 142(4): 1644-51, 2001 Apr.
Article in English | MEDLINE | ID: mdl-11250946

ABSTRACT

The 11beta-hydroxysteroid dehydrogenase type I enzyme (11betaHSD1) converts cortisone to cortisol in humans, and 11-dehydrocorticosterone to corticosterone in rodents. In the present study we used a new immunopurified polyclonal antibody, RAH113, to localize 11betaHSD1 at the light and electron microscopy levels in a wide range of rat tissues. 11betaHSD1 staining in the liver was of highest intensity around the central vein and decreased radially. In the lung, 11betaHSD1 was found at highest levels in the interstitial fibroblast, with levels in the type II pneumocyte an order of magnitude lower. RAH113 stained proximal tubules of the renal cortex and interstitial cells of the medulla and papilla. Adrenal 11betaHSD1 was confined to the glomerulosa and medulla, whereas the glucocorticoid-inactivating hydroxysteroid dehydrogenase isoform 11betaHSD2 was present in fascilulata/reticularis. 11betaHSD1 was found in parietal cells of the fundic region of the stomach, but not in the antrum. In the heart, 11betaHSD1 was detected in cells resembling interstitial fibroblasts of the endocardium and in the adventitial fibroblasts of blood vessels. Western blot analysis confirmed the presence of an antigen of the correct size (34 kDa) and intensity consistent with levels of enzyme activity previously reported in these tissues. Brain and testis also displayed the 34-kDa protein, confirming the expression of authentic 11betaHSD1 in these tissues. Electron microscopy of lung and kidney interstitial cells showed that 11betaHSD1 was localized both to the endoplasmic reticulum and the nuclear membrane. These results show that 11betaHSD1 is present in discrete cell populations where it may facilitate intracrine and paracrine glucocorticoid action in addition to its classical role of maintaining circulating glucocorticoids via activity in the liver.


Subject(s)
Hydroxysteroid Dehydrogenases/metabolism , 11-beta-Hydroxysteroid Dehydrogenase Type 2 , Animals , Blotting, Western , Immunohistochemistry , Male , Microscopy, Electron , Paracrine Communication/physiology , Rats , Rats, Sprague-Dawley , Subcellular Fractions/enzymology , Tissue Distribution
7.
Mol Cell Endocrinol ; 173(1-2): 121-5, 2001 Feb 28.
Article in English | MEDLINE | ID: mdl-11223183

ABSTRACT

11beta-hydroxysteroid dehydrogenase (11beta-HSD) regulates local actions of corticosteroids at glucocorticoid and mineralocorticoid receptors. Corticosteroids are thought to play important roles in ocular function. However, mechanisms of intraocular corticosteroid action are still unclear. Therefore, in this study, we examined the immunohistochemical localization of 11beta-HSD type 1 (11beta-HSD1), 11beta-HSD type 2 (11beta-HSD2), mineralocorticoid receptor (MR) and glucocorticoid receptor (GR) in human ocular tissues from patients (6 months to 78 years of age; n = 10) retrieved from surgical pathology files. Both 11beta-HSD2 and MR immunoreactivity was detected only in non-pigmented epithelium of the ciliary body, but was undetectable in cornea, lens, iris, retina, choroid and sclera, in all the cases examined. GR was detected in all cell types in the human eye. 11beta-HSD1 immunoreactivity was not detected in the human eye in this study. These results suggest that 11beta-HSD2 play an important role in human ocular mineralocorticoid action, such as the production of aqueous humor, in the ciliary body. The widespread expression of GR suggests that glucocorticoids may play an important role in the function and homeostasis of the human eye.


Subject(s)
Eye/enzymology , Hydroxysteroid Dehydrogenases/metabolism , 11-beta-Hydroxysteroid Dehydrogenases , Adolescent , Adult , Aged , Animals , Cell Nucleus/chemistry , Child , Child, Preschool , Ciliary Body/chemistry , Ciliary Body/cytology , Ciliary Body/enzymology , Cytoplasm/enzymology , Eye/chemistry , Eye/cytology , Humans , Hydroxysteroid Dehydrogenases/analysis , Immunohistochemistry , Infant , Middle Aged , Rabbits , Receptors, Glucocorticoid/analysis , Receptors, Mineralocorticoid/analysis
8.
Endocrinology ; 142(3): 1341-8, 2001 Mar.
Article in English | MEDLINE | ID: mdl-11181553

ABSTRACT

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.


Subject(s)
Cloning, Molecular , Collagen Type I , Collagen/genetics , Corticosterone/analogs & derivatives , Glucocorticoids/physiology , Hydroxysteroid Dehydrogenases/genetics , Osteoblasts/metabolism , Transgenes , 11-beta-Hydroxysteroid Dehydrogenases , Animals , Cell Division/drug effects , Cell Division/physiology , Cell Line , Chloramphenicol O-Acetyltransferase/genetics , Collagen Type I, alpha 1 Chain , Corticosterone/metabolism , Corticosterone/pharmacology , Dexamethasone/pharmacology , Gene Expression Regulation/physiology , Glucocorticoids/pharmacology , Hydrocortisone/pharmacology , Hydroxysteroid Dehydrogenases/metabolism , Mammary Tumor Virus, Mouse/genetics , Osteoblasts/cytology , Promoter Regions, Genetic/physiology , RNA, Messenger/metabolism , Rats , Transfection
9.
Mol Cell Endocrinol ; 171(1-2): 111-7, 2001 Jan 22.
Article in English | MEDLINE | ID: mdl-11165019

ABSTRACT

We describe a new member of the 17beta-hydroxysteroid dehydrogenase group of enzymes. Human Pan1b displays greatest activity with 5alpha-androstan-3alpha,17beta-diol (3alpha-Diol) as substrate, suggesting that it may be important in androgen metabolism. Enzymic activity was non-saturable with 3alpha-Diol but saturable with retinoids, although retinoids were not metabolized. Immunohistochemical studies on 10% formalin fixed and paraffin embedded sections of human tissues showed that Pan1b was present in acini and ciliated epithelia of the lung. In the fetus immuno reactivity was present in ciliated epithelia throughout gestation and staining appeared to be stronger in the second half of pregnancy. Pan1b was also expressed in the nonpigmented epithelium of the ciliary body, and in adrenocortical tumor cells. Although 3alpha-Diol is generally considered a degradation product of androgen metabolism it could have its own biological function. Pan1b may be an important modulator of the endocrine, or intracrine activity of this steroid.


Subject(s)
17-Hydroxysteroid Dehydrogenases/analysis , 17-Hydroxysteroid Dehydrogenases/metabolism , Alcohol Oxidoreductases , Aldehyde Oxidoreductases , Lung/enzymology , 17-Hydroxysteroid Dehydrogenases/genetics , Amino Acid Sequence , Androgens/metabolism , Androstane-3,17-diol/metabolism , Androstenedione/metabolism , Androsterone/metabolism , Carbenoxolone/metabolism , Cell Line , Cilia/enzymology , Epithelium/enzymology , Estradiol/metabolism , Estrone/metabolism , Humans , Immunohistochemistry , Infant, Newborn , Lung/embryology , Molecular Sequence Data , Retinoids/metabolism , Substrate Specificity , Testosterone/metabolism , Tissue Distribution , Trachea/enzymology , Transfection
10.
Am J Physiol Gastrointest Liver Physiol ; 279(3): G536-42, 2000 Sep.
Article in English | MEDLINE | ID: mdl-10960352

ABSTRACT

When small intestinal epithelial cells are incubated with [(3)H]corticosterone, nuclear binding is displaced neither by aldosterone nor RU-28362, suggesting that [(3)H]corticosterone is binding to a site distinct from mineralocorticoid receptor and glucocorticoid receptor. Saturation and Scatchard analysis of nuclear [(3)H]corticosterone binding demonstrate a single saturable binding site with a relatively low affinity (49 nM) and high capacity (5 fmol/microg DNA). Competitive binding assays indicate that this site has a unique steroid binding specificity, which distinguishes it from other steroid receptors. Steroid specificity of nuclear binding mirrors inhibition of the low 11beta-dehydrogenase activity, suggesting that binding may be to an 11beta-hydroxysteroid dehydrogenase (11betaHSD) isoform, although 11betaHSD1 is not present in small intestinal epithelia and 11betaHSD2 does not colocalize intracellularly with the binding site. In summary, a nuclear [(3)H]corticosterone binding site is present in small intestinal epithelia that is distinct from other steroid receptors and shares steroid specificity characteristics with 11betaHSD2 but is distinguishable from the latter by its distinct intracellular localization.


Subject(s)
Corticosterone/metabolism , Intestinal Mucosa/metabolism , Intestine, Small/metabolism , Receptors, Glucocorticoid/metabolism , 11-beta-Hydroxysteroid Dehydrogenase Type 1 , Animals , Binding, Competitive/physiology , Corticosterone/pharmacology , Dexamethasone/metabolism , Dexamethasone/pharmacology , Dose-Response Relationship, Drug , Epithelial Cells/chemistry , Epithelial Cells/enzymology , Glucocorticoids/metabolism , Glucocorticoids/pharmacology , Hydroxysteroid Dehydrogenases/metabolism , Intestinal Mucosa/chemistry , Intestine, Small/chemistry , Male , Radioligand Assay , Rats , Rats, Sprague-Dawley , Receptors, Mineralocorticoid/metabolism , Subcellular Fractions/chemistry , Subcellular Fractions/metabolism , Tritium
11.
Kidney Int ; 57(4): 1374-81, 2000 Apr.
Article in English | MEDLINE | ID: mdl-10760070

ABSTRACT

The renal 11beta-hydroxysteroid dehydrogenase type 2 (11betaHSD2) enzyme inactivates 11-hydroxy steroids in the kidney, thus protecting the nonselective mineralocorticoid receptor (MR) from occupation by glucocorticoids. The gene is highly expressed in all sodium-transporting epithelia, but also in human placenta, pancreas, and thyroid. Mutations in the HSD11B2 gene cause a rare monogenic juvenile hypertensive syndrome called apparent mineralocorticoid excess (AME). In AME, compromised 11betaHSD2 enzyme activity results in overstimulation of the MR by cortisol, causing sodium retention, hypokalemia, and salt-dependent hypertension. Recent evidence suggests a role of the 11betaHSD2 in essential hypertension. We found hypertension with no other characteristic signs of AME in the heterozygous father of a child with AME and in a girl with a homozygous gene mutation resulting in a mild deficiency of 11betaHSD2. Moreover, some studies in patients with essential hypertension showed a prolonged half-life of cortisol and an increased ratio of urinary cortisol to cortisone metabolites, suggesting a deficient 11betaHSD2 activity. These abnormalities may be genetically determined. A genetic association of a microsatellite flanking the HSD11B2 gene and hypertension in black patients with end-stage renal disease has been reported. We recently analyzed a CA-repeat allele polymorphism in unselected patients with essential hypertension, but did not find any correlation between this marker and blood pressure. However, we did find an association between this polymorphic CA microsatellite marker and salt sensitivity. Moreover, the activity of the 11betaHSD2, as shown by elevated mean ratios of urinary cortisol to cortisone metabolites, was decreased in salt-sensitive compared with salt-resistant subjects. These findings indicate that variants of the HSD11B2 gene contribute to the enhanced blood pressure response to salt in humans.


Subject(s)
Blood Pressure/physiology , Hydroxysteroid Dehydrogenases/physiology , 11-beta-Hydroxysteroid Dehydrogenases , Aldosterone/physiology , Humans , Hydroxysteroid Dehydrogenases/genetics , Hypertension/enzymology , Hypertension/etiology , Hypertension/genetics , Mineralocorticoids/metabolism , Receptors, Steroid/physiology
12.
Anticancer Res ; 20(1A): 323-8, 2000.
Article in English | MEDLINE | ID: mdl-10769675

ABSTRACT

We examined the immunohistochemical distribution of 11 beta-hydroxysteroid dehydrogenase type 2 (11 beta HSD2) and mineralocorticoid receptor (MR) in 63 primary lung carcinomas. Immunoreactivity of 11 beta HSD2 and MR was detected in 37 cases and in 32 cases of 42 adenocarcinomas, respectively. There was a significant correlation between 11 beta HSD2 and MR immunoreactivity. In three adenosquamous carcinomas, both 11 beta HSD2 and MR were detected only in adenocarcinomatous components. Neither 15 squamous cell carcinomas, 2 small cell carcinomas nor 1 large cell carcinoma expressed 11 beta HSD2 or MR. In papillary and acinar adenocarcinomas, both 11 beta HSD2 and MR immunoreactivity was significantly correlated with the grade of histological differentiation. The patterns of 11 beta HSD2 and MR expression in 10 lymph-node metastases were similar to those determined in the primary lesions. These data suggest that the patterns of 11 beta HSD2 and MR expression may reflect cellular origin and differentiation status of primary lung adenocarcinomas and serve as a new useful marker of differentiation.


Subject(s)
Carcinoma, Non-Small-Cell Lung/metabolism , Carcinoma, Small Cell/metabolism , Hydroxysteroid Dehydrogenases/biosynthesis , Isoenzymes/biosynthesis , Lung Neoplasms/metabolism , Receptors, Mineralocorticoid/biosynthesis , 11-beta-Hydroxysteroid Dehydrogenases , Adenocarcinoma/metabolism , Adenocarcinoma/pathology , Aged , Aged, 80 and over , Biomarkers , Carcinoma, Large Cell/metabolism , Carcinoma, Large Cell/pathology , Carcinoma, Non-Small-Cell Lung/pathology , Carcinoma, Small Cell/pathology , Carcinoma, Squamous Cell/metabolism , Carcinoma, Squamous Cell/pathology , Cell Differentiation , Female , Humans , Lung Neoplasms/pathology , Lymphatic Metastasis , Male , Middle Aged
13.
J Clin Endocrinol Metab ; 85(3): 1306-9, 2000 Mar.
Article in English | MEDLINE | ID: mdl-10720080

ABSTRACT

In mineralocorticoid target organs, 11beta-hydroxysteroid dehydrogenase type II (11beta-HSD2) confers specificity on the mineralocorticoid receptor (MR) by converting biologically active glucocorticoids to inactive metabolites. Placental 11beta-HSD2 is also thought to protect the fetus from high levels of circulating maternal glucocorticoid. In this study, we examined the immunoreactivity of 11beta-HSD2 and MR in human placenta from 5 weeks gestation to full term using immunohistochemistry, 11beta-HSD2 messenger RNA (mRNA) expression using Northern blot analysis, and MR mRNA expression using RT-PCR analysis. Marked 11beta-HSD2 immunoreactivity was detected in placental syncytiotrophoblasts at all gestational stages. MR immunoreactivity was moderately detected in syncytiotrophoblasts, some cytotrophoblasts, and interstitial cells of the villous core. Marked mRNA expression of 11beta-HSD2 was detected in placenta by Northern analysis. RT-PCR analysis of MR in placental tissues showed an amplified product consistent in length with the primers selected. These results suggest that placental 11beta-HSD2 is involved in not only regulating the passage of maternal active glucocorticoids into the fetal circulation but also in regulation of maternal-fetal electrolyte and water transport in the placenta, as in other mineralocorticoid target organs.


Subject(s)
Hydroxysteroid Dehydrogenases/metabolism , Placenta/metabolism , Receptors, Mineralocorticoid/metabolism , 11-beta-Hydroxysteroid Dehydrogenases , Blotting, Northern , Corticosterone/metabolism , Female , Humans , Hydroxysteroid Dehydrogenases/immunology , Immunohistochemistry , In Vitro Techniques , Placenta/enzymology , Pregnancy , Receptors, Mineralocorticoid/immunology , Reverse Transcriptase Polymerase Chain Reaction
14.
Mol Cell Endocrinol ; 154(1-2): 29-36, 1999 Aug 20.
Article in English | MEDLINE | ID: mdl-10509797

ABSTRACT

We have investigated hepatic expression and glucocorticoid regulation of the corticosteroid-binding globulin (CBG) gene in mice lacking a functional glucocorticoid receptor (GR). GR-/- mice show impaired negative feedback in the hypothalamic-pituitary-adrenal axis, resulting in elevated circulating levels of ACTH and corticosterone. This is seen in the neonatal period and continues into adulthood where ACTH and corticosterone levels are increased up to 4-5 fold. Despite high elevation of corticosterone we find no change in mean arterial blood pressure in GR-/- mice and no change in the renal activity of the glucocorticoid-metabolising enzymes 11beta-hydroxysteroid dehydrogenase type-1 (HSD1) and type-2 (HSD2). We do find markedly increased hepatic expression of CBG with a 50% increase in plasma CBG levels. Increased expression of CBG was detected in adult GR-/- mice and also at birth with a greater than 10-fold increase in CBG hepatic mRNA in day-18.5 embryonic GR-/- mice. Adult GR-/- mice were also resistant to dexamethasone-induced repression of CBG expression in the liver. These results indicate that in mice, GR is essential for maintaining the basal level of CBG gene expression in the liver, and is also required for dexamethasone-induced repression of the CBG gene in the adult.


Subject(s)
Dexamethasone/pharmacology , Gene Expression Regulation/drug effects , Receptors, Glucocorticoid/genetics , Transcortin/genetics , Adrenocorticotropic Hormone/blood , Animals , Corticosterone/blood , Kidney/metabolism , Mice , Mice, Knockout , Receptors, Glucocorticoid/deficiency , Receptors, Glucocorticoid/metabolism , Transcortin/drug effects
15.
Mol Cell Endocrinol ; 154(1-2): 71-7, 1999 Aug 20.
Article in English | MEDLINE | ID: mdl-10509802

ABSTRACT

The aim of this study was to investigate the ontogeny of localization of 11betaHSD-2 protein in the human adrenal gland. In addition, we have investigated the effects of abnormal adrenal function on 11betaHSD-2 by determining the pattern of localization of 11betaHSD-2 protein, and the amount and level of expression of 11betaHSD-2 mRNA and protein in human adrenal cortical carcinoma and adenoma. In the human foetal adrenal gland 11betaHSD-2 immunoreactivity (11betaHSD-2-ir) was detected in the foetal zone, whereas in normal adult adrenal glands 11betaHSD-2-ir was not detected by immunocytochemistry. In adrenal cortical carcinoma and adenoma, 11betaHSD-2-ir was detectable in specific regions, which have been identified as steroid synthesizing cells using 3betaHSD-ir as a marker. In adrenal cortical carcinoma and adenoma, 11betaHSD-2 mRNA and 11betaHSD-2 protein were detected by nuclease protection analysis and by western blot analysis, respectively. In summary, 11betaHSD-2-ir was detected in the foetal zone of the mid-gestation human foetal adrenal, whereas, 11betaHSD-2-ir was not detectable in the postnatal or normal adult adrenal gland. 11BetaHSD-2 protein and mRNA was induced in adult human adrenal cortical carcinoma and adenoma. The induction of expression of 11betaHSD-2 in the adrenal cortex suggests a possible role in regulating abnormal adrenal steroidogenic function in these patients.


Subject(s)
Adrenal Glands/embryology , Adrenal Glands/enzymology , Adrenocortical Adenoma/enzymology , Adrenocortical Carcinoma/enzymology , Hydroxysteroid Dehydrogenases/biosynthesis , 11-beta-Hydroxysteroid Dehydrogenases , Adrenal Glands/chemistry , Adrenal Glands/cytology , Adult , Blotting, Western , Fetus/chemistry , Fetus/cytology , Humans , Hydroxysteroid Dehydrogenases/genetics , Immunohistochemistry , Infant, Newborn , Kidney/enzymology , Methods , RNA, Messenger/analysis , Ribonucleases/analysis
16.
Ann Otol Rhinol Laryngol ; 108(10): 974-81, 1999 Oct.
Article in English | MEDLINE | ID: mdl-10526853

ABSTRACT

Mineralocorticoid hormones regulate secretion and absorption in a wide variety of epithelial tissues, although specific mechanisms in the olfactory mucosa are currently unknown. Utilizing reverse transcription-polymerase chain reaction (RT-PCR) analysis, we have demonstrated the expression of mineralocorticoid (type I) receptor messenger RNA in the rodent olfactory mucosa. Amplification products of predicted size were obtained with nucleotide sequences corresponding to respective mineralocorticoid receptor (MR) kidney transcripts. Immunocytochemistry, using an antibody with known specificity for MRs, was then utilized in order to localize the cellular site(s) of MR protein expression in the olfactory mucosa. The highest levels of MR immunoreactivity were localized to the supranuclear region of sustentacular cells, as well as the acinar cells of the Bowman's glands. The respiratory regions of the nasal cavity were devoid of appreciable MR immunoreactivity. This study demonstrates both MR transcript and protein expression in the olfactory mucosa. We hypothesize that the mineralocorticoid hormones may have a role in modulation of olfactory secretion and/or sensory transduction in the peripheral olfactory system.


Subject(s)
Olfactory Mucosa/cytology , Olfactory Mucosa/metabolism , Receptors, Mineralocorticoid/metabolism , Animals , Gene Expression/genetics , Guinea Pigs , Mice , Rats
17.
Dig Dis Sci ; 44(8): 1571-8, 1999 Aug.
Article in English | MEDLINE | ID: mdl-10492134

ABSTRACT

Mineralocorticoid action is essential for colonic sodium and water absorption and is mediated via mineralocorticoid receptors in the upper half of colonic crypts. On the other hand, it has been established that sodium butyrate induces differentiation-like phenomenon in vitro. The aim of this study is to investigate whether this bacterial product participates in the regulation of gene and protein expression of mineralocorticoid receptor in vitro. IEC6 and T84 cells were stimulated by sodium butyrate and RNAs extracted. Gene expression of mineralocorticoid receptor was evaluated by northern blotting or semiquantitative RT-PCR. Protein expression was determined in T84 cells using immunohistochemistry. To investigate whether MR induction was associated with cellular differentiation, we also measured alkaline phosphatase in situ. The mineralocorticoid receptor gene was induced by sodium butyrate in both IEC6 and T84 cells. Immunoreactivity increased in butyrate-treated T84 cells, but receptor-containing cells were not uniformly distributed and often formed clusters. Induction of alkaline phosphate activity was also demonstrated in both IEC6 and T84 cells. Double staining by immunoreactivity and alkaline phosphatase activity clearly demonstrated the colocalization of both after butyrate treatment. In conclusion, sodium butyrate up-regulates gene and protein expression of the functionally important mineralocorticoid receptor in epithelial cells, after induction by differentiation-like condition in vitro.


Subject(s)
Butyrates/pharmacology , Colon/metabolism , Intestinal Mucosa/metabolism , Intestine, Small/metabolism , Receptors, Mineralocorticoid/metabolism , Alkaline Phosphatase/metabolism , Animals , Cell Line, Transformed , Colon/cytology , Colon/drug effects , Enzyme Induction , Gene Expression/drug effects , Humans , Immunohistochemistry , Intestinal Mucosa/drug effects , Intestine, Small/cytology , Intestine, Small/drug effects , Rats , Receptors, Mineralocorticoid/genetics , Tissue Distribution
18.
J Steroid Biochem Mol Biol ; 69(1-6): 391-401, 1999.
Article in English | MEDLINE | ID: mdl-10419017

ABSTRACT

Local tissue concentrations of glucocorticoids are modulated by the enzyme 11beta-hydroxysteroid dehydrogenase which interconverts cortisol and the inactive glucocorticoid cortisone in man, and corticosterone and 11-dehydrocorticosterone in rodents. The type I isoform (11beta-HSD1) is a bidirectional enzyme but acts predominantly as a oxidoreductase to form the active glucocorticoids cortisol or corticosterone, while the type II enzyme (11beta-HSD2) acts unidirectionally producing inactive 11-keto metabolites. There are no known clinical conditions associated with 11beta-HSD1 deficiency, but gene deletion experiments in the mouse indicate that this enzyme is important both for the maintenance of normal serum glucocorticoid levels, and in the activation of key hepatic gluconeogenic enzymes. Other important sites of action include omental fat, the ovary, brain and vasculature. Congenital defects in the 11beta-HSD2 enzyme have been shown to account for the syndrome of apparent mineralocorticoid excess (AME), a low renin severe form of hypertension resulting from the overstimulation of the non-selective mineralocorticoid receptor by cortisol in the distal tubule of the kidney. Inactivation of the 11beta-HSD2 gene in mice results in a phenotype with similar features to AME. In addition, these mice show high neonatal mortality associated with marked colonic distention, and remarkable hypertrophy and hyperplasia of the distal tubule epithelia. 11Beta-HSD2 also plays an important role in decreasing the exposure of the fetus to the high levels of maternal glucocorticoids. Recent work suggests a role for 11beta-HSD2 in non-mineralocorticoid target tissues where it would modulate glucocorticoid access to the glucocorticoid receptor, in invasive breast cancer and as a mechanism providing ligand for the putative 11-dehydrocorticosterone receptor. While previous homologies between members of the SCAD superfamily have been of the order of 20-30% phylogenetic analysis of a new branch of retinol dehydrogenases indicates identities of > 60% and overlapping substrate specificities. The availability of crystal structures of family members has allowed the mapping of conserved 11beta-HSD domains A-D to a cleft in the protein structure (cofactor binding domain), two parallel beta-sheets, and an alpha-helix (active site), respectively.


Subject(s)
Hydroxysteroid Dehydrogenases/metabolism , Isoenzymes/metabolism , 11-beta-Hydroxysteroid Dehydrogenases , Animals , Arterioles/enzymology , Brain/enzymology , Corticosterone/analogs & derivatives , Corticosterone/metabolism , Humans , Hydroxysteroid Dehydrogenases/chemistry , Isoenzymes/chemistry , Kidney/enzymology , Neoplasms/enzymology , Placenta/enzymology , Protein Conformation , Receptors, Glucocorticoid/metabolism
19.
Mol Cell Endocrinol ; 151(1-2): 121-7, 1999 May 25.
Article in English | MEDLINE | ID: mdl-10411326

ABSTRACT

The 11beta-hydroxysteroid dehydrogenase enzymes (11beta-HSD) interconvert cortisol and cortisone in man, and corticosterone and 11-dehydrocorticosterone in rodents. Two distantly related congeners have been isolated and conserved domains identified by multiple alignment and hydrophobic cluster analysis. 11Beta-HSD1 in the liver acts mainly as an oxoreductase maintaining circulating glucocorticoid levels. Gene deletion studies suggest it plays an important role in providing elevated local concentrations of hormone. In contrast, 11beta-HSD2 inactivates glucocorticoids and is pivotal in the distal tubule where it protects the mineralocorticoid receptor from occupation, thus endowing specificity on a non-selective receptor. Mutations in 11beta-HSD2 result in sodium retention and severe hypertension, account for the syndrome of apparent mineralocorticoid excess and may be responsible for other forms of hypertension. 11Beta-HSD2 is also present in the placenta where it protects the fetus from high circulating levels of maternal glucocorticoids. Attenuated placental 11beta-HSD2 activity has recently been shown to be associated with intrauterine growth retardation. 11Beta-HSD2 may also play important roles in pulmonary physiology and breast cancer. This review focuses on recent developments.


Subject(s)
Hydroxysteroid Dehydrogenases/physiology , Steroids/physiology , 11-beta-Hydroxysteroid Dehydrogenases , Animals , Humans
20.
J Clin Endocrinol Metab ; 84(7): 2568-73, 1999 Jul.
Article in English | MEDLINE | ID: mdl-10404838

ABSTRACT

The role of mineralocorticoids in human gastrointestinal tract is well established. In the stomach, aldosterone is thought to regulate electrolyte transport associated with gastric acid secretion. In mineralocorticoid target organs, the action of the glucocorticoid inactivating enzyme 11beta-hydroxysteroid dehydrogenase type 2 (11beta-HSD2) facilitates aldosterone binding to a nonselective mineralocorticoid receptor (MR) in the face of high levels of circulating glucocorticoids. In the present study, we examined 25 specimens of human stomach for the presence of MR and 11beta-HSD2 using a [3H]aldosterone binding assay, Northern blot analysis, RT-PCR, and immunohistochemistry. Specific [3H]aldosterone binding sites were detected in gastric fundic mucosa, but not in the antrum. In fundic mucosa the Kd was 0.72+/-0.05 nmol/L (mean +/- SE), and Bmax was 6.0+/-1.4 fmol per milligram of protein. Northern blot analysis demonstrated a faint band for MR mRNA at 6.0 kb, although message for 11beta-HSD2 was undetectable. However, RT-PCR demonstrated specific PCR products for both MR and 11beta-HSD2. Immunohistochemistry demonstrated the colocalization of MR and 11beta-HSD2 only in parietal cells. MR-positive cells were further characterized by electron microscopy, confirming the identity of parietal cells. This study shows that parietal cells contain both MR and 11beta-HSD2, suggesting that the human stomach is a novel target organ for mineralocorticoids. Aldosterone may, therefore, regulate biological functions of parietal cells including gastric acid secretion.


Subject(s)
Gastric Mucosa/metabolism , Gene Expression , Hydroxysteroid Dehydrogenases/genetics , Receptors, Mineralocorticoid/genetics , 11-beta-Hydroxysteroid Dehydrogenases , Aldosterone/metabolism , Blotting, Northern , Gastric Fundus/chemistry , Gastric Fundus/metabolism , Gastric Mucosa/chemistry , Humans , Hydroxysteroid Dehydrogenases/analysis , Hydroxysteroid Dehydrogenases/metabolism , Immunohistochemistry , Microscopy, Electron , RNA, Messenger/analysis , Receptors, Mineralocorticoid/analysis , Receptors, Mineralocorticoid/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Stomach Neoplasms/metabolism , Tritium
SELECTION OF CITATIONS
SEARCH DETAIL