Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 30
Filter
1.
Horm Metab Res ; 47(13): 947-52, 2015 Dec.
Article in English | MEDLINE | ID: mdl-26610199

ABSTRACT

Bilateral adrenal hyperplasia currently accounts for up to 2 thirds of cases of primary aldosteronism. As such, it represents a major opportunity for targeted medical management as opposed to unilateral surgically correctable forms of the disease. Although the majority of cases of primary aldosteronism are sporadic, bilateral adrenal hyperplasia may occur in the context of familial hyperaldosteronism where it is associated with specific germline mutations. Over the past 5 years, impressive progress has been made in our understanding of the genetic basis underlying primary aldosteronism, allowing us to identify and characterize new familial forms of the disease and to understand the mechanisms involved in the formation of aldosterone producing adenoma. In contrast, our knowledge of the genetic contribution to the development of bilateral adrenal hyperplasia, and in a larger context, to renin and aldosterone levels in the general population, is still poor. This review summarizes our current knowledge on the genetics of bilateral adrenal hyperplasia and addresses some open questions to be addressed by future research. In particular, genome-wide association studies in large populations may provide clues to understanding the genetic susceptibility underlying the development of primary aldosteronism.


Subject(s)
Adrenal Glands/pathology , Adrenogenital Syndrome/genetics , Genetic Predisposition to Disease , Germ-Line Mutation , Humans , Hyperaldosteronism/genetics , Hyperplasia
2.
Mol Cell Endocrinol ; 408: 198-204, 2015 Jun 15.
Article in English | MEDLINE | ID: mdl-25433205

ABSTRACT

Serotonin (5-hydroxytryptamine; 5-HT) is able to activate the hypothalamo-pituitary-adrenal axis via multiple actions at different levels. In the human adrenal gland, 5-HT, released by subcapsular mast cells, stimulates corticosteroid production through a paracrine mode of communication which involves 5-HT receptor type 4 (5-HT4) primarily located in zona glomerulosa. As a result, 5-HT is much more efficient to stimulate aldosterone secretion than cortisol release in vitro and administration of 5-HT4 receptor agonists to healthy individuals is followed by an increase in plasma aldosterone levels without any change in plasma cortisol concentrations. Interestingly, adrenocortical hyperplasias and tumors responsible for corticosteroid hypersecretion exhibit various cellular and molecular defects which tend to reinforce the intraadrenal serotonergic tone. These pathophysiological mechanisms, which are summarized in the present review, include an increase in adrenal 5-HT production and overexpression of 5-HT receptors in adrenal neoplastic tissues. Altogether, these data support the concept of adrenal serotonergic paracrinopathy and suggest that 5-HT and its receptors may constitute valuable targets for pharmacological treatments of primary adrenal diseases.


Subject(s)
Adrenal Cortex Neoplasms/drug therapy , Paracrine Communication/drug effects , Serotonin/pharmacology , Serotonin/therapeutic use , Steroids/biosynthesis , Animals , Humans , Hyperplasia , Models, Biological
3.
Ann Cardiol Angeiol (Paris) ; 63(3): 213-5, 2014 Jun.
Article in French | MEDLINE | ID: mdl-24952675

ABSTRACT

Itraconazole is a triazole agent used in the treatment of fungal infections and in some metastatic cancers. Its use has been associated with cardiovascular adverse events and particularly heart failure with preserved ejection fraction. We report the case of a 68-year-old male patient with a well-controlled hypertension treated with irbesartan 150mg/day since 2007. He developed a pulmonary aspergillosis on post-tuberculosis cavitary lesions treated in July 2011 with itraconazole 200mg/day. Early 2012, his antihypertensive treatment had to be gradually increased to a quadritherapy and his blood pressure was at 157/78mmHg at home. Hypokalemia was observed on several occasions as well as edema of the lower limbs. Plasma renin and plasma and urine aldosterone concentrations on treatment not interfering with the renin angiotensin system were low, associated with normal serum and urine cortisol, ACTH, SDHA and DOC, BNP and creatinine concentrations. Plasma itraconazole values were much above the therapeutic range. Left ventricular ejection fraction was preserved. There were no adrenal or renal artery abnormalities at the CT scan. Three months after stopping itraconazole, hypokalemia and edema disappeared and blood pressure was normalized with less treatment. Plasma renin and aldosterone concentrations were normalized. He had a pulmonary lobectomy for his pulmonary aspergillosis. Itraconazole may induce a resistant hypertension with low renin. The mechanisms of this adverse effect of itraconazole remain unknown.


Subject(s)
Antifungal Agents/adverse effects , Hypertension/chemically induced , Itraconazole/adverse effects , Aged , Antifungal Agents/administration & dosage , Humans , Itraconazole/administration & dosage , Male , Pulmonary Aspergillosis/drug therapy , Withholding Treatment
5.
J Endocrinol Invest ; 33(7): 472-7, 2010.
Article in English | MEDLINE | ID: mdl-19955850

ABSTRACT

BACKGROUND/AIMS: Aldosterone and the mineralocorticoid receptor (MR) play a major role in sodium balance and blood pressure control. They are also involved in adipocyte metabolism. The aim of this study was to analyze the association between the MR p.I180V polymorphism with hypertension and markers of cardiovascular risk. DESIGN AND METHODS: Case-control study nested within a cohort of 2063 subjects followed since birth to date. All subjects (age 23-25 yr old) from the entire cohort with systolic and diastolic hypertension (no.=126) were paired with 398 normotensive controls. MR p.I180V genotype association with anthropometric and biochemical markers of cardiometabolic risk was tested. RESULTS: There was a significant association of the MR p.I180V genotype with body mass index (BMI) and LDL-cholesterol level (p<0.01). Hypertensive subjects carrying the polymorphic G allele (AG or GG genotypes) presented significantly higher BMI (30.0+/-6.0 vs 28.7+/-5.6 kg/m(2); p<0.01) and higher LDL-cholesterol (139.9+/-60.3 vs 109.9+/-35.5 mg/dl; p<0.01). The frequency of the polymorphism MR p.I180V was similar between hypertensive subjects and controls (p=0.15). CONCLUSIONS: The MR p.I180V polymorphism seems to be associated with cardiovascular risk factors including BMI and LDL-cholesterol levels. This original in vivo finding reinforces the role of MR in adipocyte biology and in cardiovascular disease.


Subject(s)
Body Mass Index , Cholesterol, LDL/blood , Hypertension/genetics , Receptors, Mineralocorticoid/genetics , Adult , Case-Control Studies , Cohort Studies , Female , Gene Frequency , Humans , Hypertension/blood , Male , Polymorphism, Genetic , Risk Factors
6.
Climacteric ; 11(3): 258-64, 2008 Jun.
Article in English | MEDLINE | ID: mdl-18568791

ABSTRACT

Beside their role in the control of water and electrolyte homeostasis, recent data clearly indicate that aldosterone and the mineralocorticoid receptor (MR) are involved in adipocyte biology. It has been recently shown that aldosterone promotes white and brown adipocyte differentiation in vitro through specific activation of the MR. In addition, a non-epithelial pro-inflammatory role for MR activation has been recently inferred from studies on mineralocorticoid/salt administration in experimental animal models and from clinical studies. The mineralocorticoid system could hence represent a potential target for new therapeutic strategies in obesity and the metabolic syndrome. Progesterone has high affinity for the MR and is a natural antagonist of aldosterone. Differently from classic synthetic progestins, which are devoid of antimineralocorticoid properties, progesterone and new progestogens show remarkable antimineralocorticoid effects. Here, we discuss the potential role of the antimineralocorticoid properties of progestogens in the control of body weight, adipose tissue proliferation and salt sensitivity; their therapeutic use in postmenopausal women, as well as in women affected by polycystic ovary syndrome, may open new and unexpected possibilities in the treatment of related metabolic disorders.


Subject(s)
Adipose Tissue/drug effects , Progestins/pharmacology , Receptors, Mineralocorticoid , Sodium Chloride, Dietary/adverse effects , Water-Electrolyte Balance/drug effects , Adipose Tissue/metabolism , Animals , Cell Differentiation/drug effects , Female , Humans , Metabolic Syndrome/drug therapy , Mineralocorticoid Receptor Antagonists/pharmacology , Obesity/drug therapy , Progestins/physiology , Receptors, Mineralocorticoid/drug effects , Receptors, Mineralocorticoid/metabolism , Water-Electrolyte Balance/physiology
7.
J Mol Endocrinol ; 33(3): 679-91, 2004 Dec.
Article in English | MEDLINE | ID: mdl-15591027

ABSTRACT

The pituitary hormone prolactin (PRL) exerts pleiotropic effects, which are mediated by a membrane receptor (PRLR) present in numerous cell types including adipocytes. Brown adipose tissue (BAT) expresses uncoupling proteins (UCPs), involved in thermogenesis, but also secretes leptin, a key hormone involved in the control of body weight. To investigate PRL effects on BAT, we used the T37i brown adipose cell line, and demonstrated that PRLRs are expressed as a function of cell differentiation. Addition of PRL leads to activation of the JAK/STAT and MAP kinase signaling pathways, demonstrating that PRLRs are functional in these cells. Basal and catecholamine-induced UCP1 expression were not affected by PRL. However, PRL combined with insulin significantly increases leptin expression and release, indicating that PRL potentiates the stimulatory effect of insulin as revealed by the recruitment of insulin receptor substrates and the activation of phosphatidylinositol 3-kinase. To explore the in vivo physiological relevance of PRL action in BAT, we showed that leptin content was significantly increased in BAT of PRLR-null mice compared with wild-type mice, highlighting the involvement of PRL in the leptin secretion process. This study provides the first evidence for a functional link between PRL and energy balance via a cross-talk between insulin and PRL signaling pathways in brown adipocytes.


Subject(s)
Adipocytes/drug effects , Adipocytes/metabolism , Adipose Tissue, Brown/drug effects , Gene Expression Regulation/drug effects , Insulin/pharmacology , Leptin/metabolism , Prolactin/pharmacology , Adipocytes/cytology , Adipose Tissue, Brown/cytology , Adipose Tissue, Brown/metabolism , Animals , Carrier Proteins/metabolism , Cell Differentiation , Cell Line , DNA-Binding Proteins/metabolism , Ion Channels , Janus Kinase 2 , Membrane Proteins/metabolism , Mice , Mice, Knockout , Milk Proteins/metabolism , Mitochondrial Proteins , Mitogen-Activated Protein Kinases/metabolism , Protein Binding , Protein-Tyrosine Kinases/metabolism , Proto-Oncogene Proteins/metabolism , Receptors, Leptin , Receptors, Prolactin/deficiency , Receptors, Prolactin/genetics , Receptors, Prolactin/metabolism , STAT3 Transcription Factor , STAT5 Transcription Factor , Signal Transduction/drug effects , Trans-Activators/metabolism , Transcription, Genetic/genetics , Uncoupling Protein 1
8.
Mol Cell Endocrinol ; 217(1-2): 119-25, 2004 Mar 31.
Article in English | MEDLINE | ID: mdl-15134810

ABSTRACT

Type I pseudohypoaldosteronism (PHA1) is a rare form of mineralocorticoid resistance characterized by neonatal renal salt wasting and failure to thrive. Typical biochemical features include high levels of plasma aldosterone and renin, hyponatremia and hyperkalemia. Different mutations of the human mineralocorticoid receptor (hMR) gene have been identified in subjects affected by the autosomal dominant or sporadic form of the disease. Our laboratory has investigated a large number of subjects with familial and sporadic PHA1. Several different mutations have been detected, which are localized in different coding exons of the hMR gene. These mutations either create truncated proteins, either affect specific amino acids involved in receptor function. In this paper, we review hMR mutations described to date in PHA1 and their functional characterization. We discuss the absence of mutations in some kindreds and the role of precise phenotypic and biological examination of patients to allow for identification of other genes potentially involved in the disease.


Subject(s)
Exons/genetics , Mutation , Pseudohypoaldosteronism/genetics , Receptors, Mineralocorticoid/genetics , Aldosterone/blood , Genes, Dominant/genetics , Humans , Hypokalemia/genetics , Hypokalemia/physiopathology , Hyponatremia/genetics , Hyponatremia/physiopathology , Kidney/physiopathology , Mineralocorticoids/metabolism , Pedigree , Predictive Value of Tests , Pseudohypoaldosteronism/blood , Pseudohypoaldosteronism/congenital , Pseudohypoaldosteronism/physiopathology , Receptors, Mineralocorticoid/metabolism , Renin/blood , Salts/metabolism
9.
J Biol Chem ; 276(42): 38911-20, 2001 Oct 19.
Article in English | MEDLINE | ID: mdl-11495902

ABSTRACT

The mineralocorticoid receptor (MR), a ligand-dependent transcription factor, mediates aldosterone actions in a large variety of tissues. To explore the functional implication of MR in pathophysiology, transgenic mouse models were generated using the proximal human MR (hMR) promoter to drive expression of hMR in aldosterone target tissues. Tissue-specific analysis of transgene expression in two independent transgenic animal (TG) lines by ribonuclease protection assays revealed that hMR is expressed in all mineralocorticoid-sensitive tissues, most notably in the kidney and the heart. TG exhibit both renal and cardiac abnormalities. Enlarged kidneys were histologically associated with renal tubular dilation and cellular vacuolization whose prevalence increased with aging. Renal clearance studies also disclosed a significant decrease in urinary potassium excretion rate in TG. hMR-expressing animals had normal blood pressure but developed mild dilated cardiomyopathy (increased left ventricle diameters and decreased shortening fraction), which was accompanied by a significant increase in heart rate. Differential gene expression analysis revealed a 2- to 5-fold increase in cardiac expression of atrial natriuretic peptide, serum- and glucocorticoid-induced kinase, and early growth response gene 1 as detected by microarrays; renal serum- and glucocorticoid-induced kinase was also induced significantly. Altogether, TG exhibited specific alteration of renal and cardiac functions, thus providing useful pathophysiological models to gain new insights into the tissue-specific mineralocorticoid signaling pathways.


Subject(s)
Heart/physiology , Immediate-Early Proteins , Kidney/physiology , Mice, Transgenic , Nuclear Proteins , Receptors, Mineralocorticoid/biosynthesis , Animals , Blotting, Northern , DNA, Complementary/metabolism , DNA-Binding Proteins/biosynthesis , Early Growth Response Protein 1 , Humans , Kidney/metabolism , Male , Mice , Models, Genetic , Myocardium/metabolism , Oligonucleotide Array Sequence Analysis , Promoter Regions, Genetic , Protein Serine-Threonine Kinases/biosynthesis , Receptors, Mineralocorticoid/genetics , Recombinant Proteins/metabolism , Signal Transduction , Sodium-Potassium-Exchanging ATPase/biosynthesis , Time Factors , Tissue Distribution , Transcription Factors/biosynthesis
10.
Mol Endocrinol ; 15(9): 1586-98, 2001 Sep.
Article in English | MEDLINE | ID: mdl-11518808

ABSTRACT

Aldosterone effects are mediated by the MR, which possesses the same affinity for mineralocorticoids and glucocorticoids. In addition to the existence of mechanisms regulating intracellular hormone availability, we searched for human MR splice variants involved in tissue-specific corticosteroid function. We have identified a new human MR isoform, hMRDelta5,6, resulting from an alternative splicing event skipping exons 5 and 6 of the human MR gene. hMRDelta5,6 mRNAs are expressed in several human tissues at different levels compared with wild-type human MR, as shown by real time PCR. Introduction of a premature stop codon results in a 75-kDa protein lacking the entire hinge region and ligand binding domain. Interestingly, hMRDelta5,6 is still capable of binding to DNA and acts as a ligand-independent transactivator, with maximal transcriptional induction corresponding to approximately 30-40% of aldosterone-activated wild-type human MR. Coexpression of hMRDelta5,6 with human MR or human GR increases their transactivation potential at high doses of hormone. Finally, hMRDelta5,6 is able to recruit the coactivators, steroid receptor coactivator 1, receptor interacting protein 140, and transcription intermediary factor 1alpha, which enhance its transcriptional activity. Ligand-independent transactivation and enhancement of both wild-type MR and GR activities by hMRDelta5,6 suggests that this new variant might play a role in modulating corticosteroid effects in target tissues.


Subject(s)
Adrenal Cortex Hormones/metabolism , Alternative Splicing/genetics , Receptors, Mineralocorticoid/genetics , Trans-Activators/genetics , Amino Acid Sequence , Animals , Base Sequence , Cells, Cultured , Humans , Molecular Sequence Data , Protein Isoforms , Rabbits , Receptors, Mineralocorticoid/metabolism , Trans-Activators/metabolism , Transcription, Genetic/genetics
11.
Am J Physiol Endocrinol Metab ; 280(4): E640-9, 2001 Apr.
Article in English | MEDLINE | ID: mdl-11254472

ABSTRACT

Uncoupling proteins (UCP), specific mitochondrial proton transporters that function by uncoupling oxidative metabolism from ATP synthesis, are involved in thermoregulation and control of energy expenditure. The hibernoma-derived T37i cells, which possess functional endogenous mineralocorticoid receptors (MR), can undergo differentiation into brown adipocytes. In differentiated T37i cells, UCP1 mRNA levels increased 10- to 20-fold after retinoic acid or beta-adrenergic treatment. Interestingly, UCP2 and UCP3 mRNA was also detected. Aldosterone treatment induced a drastic decrease in isoproterenol- and retinoic acid-stimulated UCP1 mRNA levels in a time- and dose-dependent manner (IC(50) approximately 1 nM aldosterone). This inhibition was unaffected by cycloheximide and did not modify UCP1 mRNA stability (half-life time = 5 h), indicating that it occurs at the transcriptional level. It involves both the MR and/or the glucocorticoid receptor (GR), depending on the retinoic or catecholamine induction pathway. Basal UCP3 expression was also significantly reduced by aldosterone, whereas UCP2 mRNA levels were not modified. Finally, as demonstrated by JC1 aggregate formation in living cells, aldosterone restored mitochondrial membrane potential abolished by isoproterenol or retinoic acid. Our results demonstrate that MR and GR inhibit expression of UCP1 and UCP3, thus participating in the control of energy expenditure.


Subject(s)
Adipocytes/metabolism , Adipose Tissue, Brown/metabolism , Carrier Proteins/antagonists & inhibitors , Membrane Proteins/antagonists & inhibitors , Receptors, Glucocorticoid/physiology , Receptors, Mineralocorticoid/physiology , Adipose Tissue, Brown/cytology , Aldosterone/pharmacology , Animals , Carrier Proteins/metabolism , Carrier Proteins/physiology , Cell Line , Intracellular Membranes/physiology , Ion Channels , Membrane Potentials/drug effects , Membrane Proteins/metabolism , Membrane Proteins/physiology , Mice , Mitochondria/physiology , Mitochondrial Proteins , Tumor Cells, Cultured , Uncoupling Protein 1 , Uncoupling Protein 3
12.
Am J Physiol Endocrinol Metab ; 279(2): E386-94, 2000 Aug.
Article in English | MEDLINE | ID: mdl-10913039

ABSTRACT

By use of targeted oncogenesis, a brown adipocyte cell line was derived from a hibernoma of a transgenic mouse carrying the proximal promoter of the human mineralocorticoid receptor (MR) linked to the SV40 large T antigen. T37i cells remain capable of differentiating into brown adipocytes upon insulin and triiodothyronine treatment as judged by their ability to express uncoupling protein 1 and maintain MR expression. Aldosterone treatment of undifferentiated cells induced accumulation of intracytoplasmic lipid droplets and mitochondria. This effect was accompanied by a significant and dose-dependent increase in intracellular triglyceride content (half-maximally effective dose 10(-9) M) and involved MR, because it was unaffected by RU-38486 treatment but was totally abolished in the presence of aldosterone antagonists (spironolactone, RU-26752). The expression of early adipogenic gene markers, such as lipoprotein lipase, peroxisome proliferator-activated receptor-gamma, and adipocyte-specific fatty acid binding protein 2, was enhanced by aldosterone, confirming activation of the differentiation process. We demonstrate that, in the T37i cell line, aldosterone participates in the very early induction of brown adipocyte differentiation. Our findings may have a broader biological significance and suggest that MR is not only implicated in maintaining electrolyte homeostasis but could also play a role in metabolism and energy balance.


Subject(s)
Adipose Tissue, Brown/metabolism , Cell Differentiation/physiology , Neoplasm Proteins , Nerve Tissue Proteins , Receptors, Mineralocorticoid/metabolism , Spironolactone/analogs & derivatives , Adipose Tissue, Brown/pathology , Aldosterone/pharmacology , Animals , Carrier Proteins/genetics , Carrier Proteins/metabolism , Cell Differentiation/drug effects , Dose-Response Relationship, Drug , Fatty Acid-Binding Protein 7 , Fatty Acid-Binding Proteins , Hormone Antagonists/pharmacology , Lipoma/metabolism , Lipoma/pathology , Lipoma/ultrastructure , Lipoprotein Lipase/genetics , Lipoprotein Lipase/metabolism , Mice , Mifepristone/pharmacology , Mineralocorticoid Receptor Antagonists/pharmacology , Myelin P2 Protein/genetics , Myelin P2 Protein/metabolism , RNA, Messenger/biosynthesis , Receptors, Cytoplasmic and Nuclear/genetics , Receptors, Cytoplasmic and Nuclear/metabolism , Spironolactone/pharmacology , Transcription Factors/genetics , Transcription Factors/metabolism , Transcription, Genetic/drug effects , Triglycerides/metabolism , Tumor Cells, Cultured
13.
Ann Endocrinol (Paris) ; 61(1): 41-6, 2000 Feb.
Article in English | MEDLINE | ID: mdl-10790591

ABSTRACT

The cardiovascular system is now recognized as an important mineralocorticoid target. All -components required for specific and selective aldosterone effects are present in the cardiovascular system. Mineralocorticoid receptors (MR) are expressed in the heart and large blood vessels together with the 11 B-hydroxysteroid dehydrogenase type II, which ensures the enzymatic protection of MR against glucocorticoids. The recent description of local vascular and cardiac aldosterone biosynthesis strongly supports an autocrine/paracrine hormonal action. Establishment of transgenic mice models of targeted overexpression of the mineralocorticoid receptor should facilitate new insights into the molecular and cellular mechanisms of aldo-sterone actions in the cardiovascular system.


Subject(s)
Cardiovascular Physiological Phenomena , Receptors, Mineralocorticoid/analysis , Receptors, Mineralocorticoid/physiology , Aldosterone/biosynthesis , Aldosterone/pharmacology , Animals , Cardiovascular Diseases/etiology , Cardiovascular Diseases/physiopathology , Cardiovascular System/drug effects , Cardiovascular System/physiopathology , Gene Expression , Humans , Mice , Mice, Transgenic , Receptors, Mineralocorticoid/genetics
14.
Kidney Int ; 57(4): 1299-306, 2000 Apr.
Article in English | MEDLINE | ID: mdl-10760058

ABSTRACT

The mineralocorticoid receptor (MR) is a transcription factor that mediates aldosterone action. MR is expressed in a wide variety of tissues, most notably in sodium-transporting epithelia, but also in nonepithelial cells of the cardiovascular and central nervous systems. However, molecular mechanisms underlying mineralocorticoid signaling and the primary mineralocorticoid-regulated genes are not fully identified. We recently showed that the human MR (hMR) gene possesses two first 5'-untranslated exons 1alpha and 1beta, and demonstrated that the 5'-flanking regions of these exons, named P1 and P2, respectively, are functional promoters that differ by their basal and corticosteroid-regulated transcriptional activities. To gain insight into the tissue-specific expression and function of MR, we have established transgenic mouse models using both targeted oncogenesis and receptor overexpression strategies. P1 and P2 promoters were used to direct expression of the large T antigen (TAg) of SV40 in constitutively MR-expressing cells. P1.TAg mice developed lethal hibernomas, while P2.TAg animals died from cerebral neuroectodermal tumors and leiomyosarcomas. Quantification of TAg messenger RNA levels revealed that P1 and P2 were differentially utilized. P1 promoter was transcriptionally active in all MR-expressing tissues and importantly directed an appropriate transgene expression in the distal nephron. Conversely, P2 activity was weak and spatially restricted. Several immortalized cell lines were established, thus constituting valuable models to investigate on aldosterone-regulated proteins. We also used P1 and P2 to target overexpression of hMR cDNA in mice. Phenotypic characterization of these mice is currently under investigation. Some transgenic lines should represent useful systems to further explore multiple functions of MR in vivo.


Subject(s)
Mice, Transgenic/genetics , Receptors, Mineralocorticoid/genetics , Receptors, Mineralocorticoid/metabolism , Animals , Gene Targeting , Humans , Mice , Neoplasms, Experimental/genetics , Promoter Regions, Genetic/genetics
15.
J Biol Chem ; 275(11): 7878-86, 2000 Mar 17.
Article in English | MEDLINE | ID: mdl-10713103

ABSTRACT

The human mineralocorticoid receptor (hMR) is a nuclear receptor mediating aldosterone action, whose expression is driven by two alternative promoters, P1 and P2, flanking the two first 5'-untranslated exons. In vivo characterization of hMR regulatory regions was performed by targeted oncogenesis in mice using P1 or P2 directing expression of the large T antigen of SV40 (TAg). While transgenic P1.TAg founders rapidly developed lethal hibernomas from brown fat, cerebral primitive neuroectodermal tumors and facial leiomyosarcomas occurred in P2.TAg mice. Quantitative analyses of mouse MR (mMR) and transgene expression indicate that P1 promoter was transcriptionally active in all MR-expressing tissues, directing strong TAg expression in testis and salivary glands, moderate in lung, brain, uterus, liver, and heart but, unlike mMR, rather low in colon and kidney. Importantly, the renal transgene expression colocalized with mMR in the distal nephron. In contrast, P2 promoter was approximately 10 times less potent than P1, with no activity in the brain and colon. Several immortalized cell lines were established from both neoplastic and normal tissues of transgenic mice. These cells exhibited differentiated characteristics and maintained MR expression, thus providing useful models for further studies exploring the widespread expression and functions of MR. Our results demonstrate that hMR gene expression in vivo is controlled by complex regulatory mechanisms involving distinct tissue-specific utilization of alternative promoters.


Subject(s)
Promoter Regions, Genetic , Receptors, Mineralocorticoid/genetics , Adipose Tissue, Brown/pathology , Animals , Antigens, Viral, Tumor/genetics , Brain/pathology , Cell Transformation, Neoplastic/genetics , Gene Expression Regulation, Neoplastic , Humans , Leiomyosarcoma/genetics , Leiomyosarcoma/pathology , Lipoma/genetics , Lipoma/pathology , Mice , Mice, Transgenic , Mineralocorticoids/metabolism , Neuroectodermal Tumors/genetics , Neuroectodermal Tumors/pathology , Phenotype , RNA, Messenger/analysis , Receptors, Mineralocorticoid/metabolism , Signal Transduction , Simian virus 40 , Tissue Distribution
18.
J Clin Invest ; 101(6): 1254-60, 1998 Mar 15.
Article in English | MEDLINE | ID: mdl-9502766

ABSTRACT

Aldosterone is a major regulator of salt balance and blood pressure, exerting its effects via the mineralocorticoid receptor (MR). To analyze the regulatory mechanisms controlling tissue-specific expression of the human MR (hMR) in vivo, we have developed transgenic mouse models expressing the SV40 large T antigen (TAg) under the control of each of the two promoters of the hMR gene (P1 or P2). Unexpectedly, all five P1-TAg founder animals died prematurely from voluminous malignant liposarcomas originating from brown adipose tissue, as evidenced by the expression of the mitochondrial uncoupling protein ucp1, indicating that the proximal P1 promoter was transcriptionally active in brown adipocytes. No such hibernoma occurred in P2-TAg transgenic mice. Appropriate tissue-specific usage of P1 promoter sequences was confirmed by demonstrating the presence of endogenous MR in both neoplastic and normal brown adipose tissue. Several cell lines were derived from hibernomas; among them, the T37i cells can undergo terminal differentiation into brown adipocytes, which remain capable of expressing ucp1 upon adrenergic or retinoic acid stimulation. These cells possess endogenous functional MR, thus providing a new model to explore molecular mechanisms of mineralocorticoid action. Our data broaden the known functions of aldosterone and suggest a potential role for MR in adipocyte differentiation and regulation of thermogenesis.


Subject(s)
Adipose Tissue, Brown/metabolism , Aldosterone/metabolism , Antigens, Polyomavirus Transforming/genetics , Lipoma/genetics , Lipoma/metabolism , Receptors, Mineralocorticoid/genetics , Adipose Tissue, Brown/pathology , Animals , Blotting, Northern , Carrier Proteins/genetics , Carrier Proteins/metabolism , Cloning, Molecular , Gene Expression , Histocytochemistry , Humans , Immunohistochemistry , Ion Channels , Lipoma/pathology , Liposarcoma/genetics , Liposarcoma/metabolism , Liposarcoma/pathology , Membrane Proteins/genetics , Membrane Proteins/metabolism , Mice , Mice, Transgenic , Mitochondrial Proteins , Polymerase Chain Reaction , Promoter Regions, Genetic , Receptors, Mineralocorticoid/metabolism , Transcription, Genetic , Triglycerides/analysis , Triglycerides/metabolism , Tumor Cells, Cultured , Uncoupling Protein 1
19.
J Clin Endocrinol Metab ; 82(5): 1345-52, 1997 May.
Article in English | MEDLINE | ID: mdl-9141514

ABSTRACT

Expression of the mineralocorticoid receptor (MR) is restricted to some sodium-transporting epithelia and a few nonepithelial target tissues. Determination of the genomic structure of the human MR (hMR) revealed two different untranslated exons (1alpha and 1beta), which splice alternatively into the common exon 2, giving rise to two hMR mRNA isoforms (hMR alpha and hMR beta). We have investigated expression of hMR transcripts in renal, cardiac, skin, and colonic tissue samples by in situ hybridization with exon 1alpha and 1beta specific riboprobes, using an exon 2 probe as internal control. Specific signals for either exon 1alpha- and 1beta-containing mRNAs were detected in typically hMR-expressing cells in all tissues analyzed. hMR alpha and hMR beta were present in distal tubules of the kidney, in cardiomyocytes, in enterocytes of the colonic mucosa, and in keratinocytes and sweat glands. Interestingly, although both isoforms appear to be expressed at approximately the same level, the relative abundance of each message compared with that of exon 2-containing mRNA strikingly differs among aldosterone target tissues, suggesting the possibility of other tissue-specific transcripts originating from alternative splicing. Finally, functional hypermineralocorticism was associated with reduced expression of hMR beta in sweat glands of two patients affected by Conn's and Liddle's syndrome, whereas normal levels of hMR isoforms were found in one case of pseudohypoaldosteronism. Altogether, our results indicate a differential, tissue-specific expression of hMR mRNA isoforms, hMR beta being down-regulated in situations of positive sodium balance, independently of aldosterone levels.


Subject(s)
Gene Expression , RNA, Messenger/analysis , Receptors, Mineralocorticoid/genetics , Aldosterone/metabolism , Alternative Splicing , Colon/chemistry , Exons , Humans , In Situ Hybridization , Intestinal Mucosa/chemistry , Keratinocytes/chemistry , Kidney/chemistry , Mineralocorticoids/metabolism , Myocardium/chemistry , Pseudohypoaldosteronism/metabolism , Skin/chemistry , Sweat Glands/chemistry , Tissue Distribution
20.
Mol Endocrinol ; 10(12): 1549-60, 1996 Dec.
Article in English | MEDLINE | ID: mdl-8961265

ABSTRACT

The mineralocorticoid receptor (MR) is a ligand-dependent transcription factor involved in the regulation of sodium homeostasis. Two distinct mRNA isoforms of the human MR (hMR) differing in their untranslated 5'-ends have recently been identified, suggesting the existence of alternative promoters. To eludicate the regulatory mechanisms controlling hMR gene expression, we have isolated and characterized approximately 15 kb of hMR 5'-flanking region. Various deletion mutants of regions located immediately upstream of the untranslated exons 1 alpha and 1 beta (P1: 1 kb and P2: 1.7 kb, respectively) were inserted into a luciferase reporter gene and used in transient transfection experiments in CV-1 and human differentiated renal H5 cells. Both regions were shown to possess significant functional promoter activity, more pronounced in renal cells, although P1 directed higher levels of basal transcription. Cotransfection experiments with hMR or human glucocorticoid receptor (hGR) revealed that, while both promoters were glucocorticoid inducible, only the distal P2 promoter was stimulated by aldosterone in a dose- and hMR-dependent manner. Furthermore, we demonstrate that hMR and hGR are able to synergistically activate the P2 promoter, consistent with cooperativity between the two transduction pathways. Mineralocorticoid induction was localized to a region between -318 and +123 bp of P2. This region does not contain any consensus hormone responsive element, and direct binding of hMR to this DNA sequence was not observed, indicating that mineralocorticoid-induced transcriptional enhancement is mediated by nonclassical mechanisms. On the other hand, Sp1 and AP-2 bind to definite sequences on both promoters, suggesting that they represent important regulators of hMR promoter activity. Our results indicate that hMR gene expression is under the control of complex regulatory mechanisms involving alternative promoters and differential hormonal control, which might allow tissue-specific modulation of aldosterone action.


Subject(s)
Receptors, Mineralocorticoid/genetics , Receptors, Mineralocorticoid/metabolism , Regulatory Sequences, Nucleic Acid , Aldosterone/metabolism , Aldosterone/pharmacology , Alternative Splicing , Animals , Base Sequence , Binding Sites , Cloning, Molecular , DNA Footprinting , Deoxyribonuclease I/metabolism , Exons , Gene Expression Regulation , Humans , Molecular Sequence Data , Promoter Regions, Genetic , Receptors, Glucocorticoid/genetics , Receptors, Glucocorticoid/metabolism , Receptors, Mineralocorticoid/drug effects , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Sequence Analysis, DNA , Transcriptional Activation , Transfection
SELECTION OF CITATIONS
SEARCH DETAIL