Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 30
Filter
1.
Steroids ; 163: 108711, 2020 11.
Article in English | MEDLINE | ID: mdl-32739291

ABSTRACT

AIM: The present study investigated the effects of anabolic steroid (AS) excess on blood pressure regulation. METHODS: Male Wistar rats were treated with nandrolone decanoate (AS) or vehicle (CTL) for 8 or 10 weeks. Saline (1.8%) and water intake were measured in metabolic cages. Urinary volume, osmolarity, Na+ and K+ concentrations, and plasma osmolarity were measured. The autonomic balance was estimated by heart rate variability at baseline or after icv injection of losartan. Cardiac function was assessed by echocardiography and ex vivo recordings. Myocardial collagen deposition was evaluated by Picrosirius-Red staining. Vascular reactivity and wall thickness were investigated in aortic sections. Blood pressure (BP) was assessed by tail-cuff plethysmography. Angiotensin II type I receptor (AT1R), renin, and mineralocorticoid receptor (MR) mRNA expression was measured in the kidneys and whole hypothalamus. RESULTS: AS group exhibited decreased urinary volume and Na+ concentration, while urinary K+ concentration, plasma osmolarity, and renal AT1R and renin mRNA levels were increased compared to CTL (p < 0.05). Water intake was increased, and saline intake was decreased in the AS group (p < 0.01). AS group exhibited increased low-frequency/high-frequency-ratio, while it was decreased by icv injection of losartan (p < 0.05) compared to baseline. Neither cardiac function nor vascular reactivity/morphology was affected by AS excess (p > 0.05). Ultimately, BP levels were not altered by AS excess (p > 0.05). CONCLUSION: AS excess promoted hydroelectrolytic and autonomic imbalance but did not alter vascular or cardiac function/morphology.


Subject(s)
Anabolic Agents/pharmacology , Autonomic Nervous System/drug effects , Autonomic Nervous System/physiology , Blood Pressure/drug effects , Nandrolone Decanoate/pharmacology , Animals , Gene Expression Regulation/drug effects , Hypothalamus/drug effects , Hypothalamus/metabolism , Kidney/drug effects , Kidney/metabolism , Male , Mineralocorticoids/genetics , RNA, Messenger/genetics , Rats , Rats, Wistar , Receptor, Angiotensin, Type 1/genetics , Renin/genetics
2.
Horm Res Paediatr ; 89(5): 292-310, 2018.
Article in English | MEDLINE | ID: mdl-29874650

ABSTRACT

Disorders of adrenal steroidogenesis comprise autosomal recessive conditions affecting steroidogenic enzymes of the adrenal cortex. Those are located within the 3 major branches of the steroidogenic machinery involved in the production of mineralocorticoids, glucocorticoids, and androgens. This mini review describes the principles of adrenal steroidogenesis, including the newly appreciated 11-oxygenated androgen pathway. This is followed by a description of pathophysiology, biochemistry, and clinical implications of steroidogenic disorders, including mutations affecting cholesterol import and steroid synthesis, the latter comprising both mutations affecting steroidogenic enzymes and co-factors required for efficient catalysis. A good understanding of adrenal steroidogenic pathways and their regulation is crucial as the basis for sound management of these disorders, which in the majority present in early childhood.


Subject(s)
Adrenal Gland Diseases/enzymology , Adrenal Gland Diseases/genetics , Genetic Diseases, Inborn/enzymology , Genetic Diseases, Inborn/genetics , Mutation , Adrenal Gland Diseases/therapy , Androgens/genetics , Androgens/metabolism , Cholesterol/genetics , Cholesterol/metabolism , Genetic Diseases, Inborn/therapy , Glucocorticoids/genetics , Glucocorticoids/metabolism , Humans , Mineralocorticoids/genetics , Mineralocorticoids/metabolism
3.
Ann Endocrinol (Paris) ; 79(3): 174-181, 2018 Jun.
Article in English | MEDLINE | ID: mdl-29661472

ABSTRACT

Primary adrenal insufficiency (PAI) is characterized by impaired production of steroid hormones due to an adrenal cortex defect. This condition incurs a risk of acute insufficiency which may be life-threatening. Today, 80% of pediatric forms of PAI have a genetic origin but 5% have no clear genetic support. Recently discovered mutations in genes relating to oxidative stress have opened the way to research on genes unrelated to the adrenal gland. Identification of causal mutations in a gene responsible for PAI allows genetic counseling, guidance of follow-up and prevention of complications. This is particularly true for stress oxidative anomalies, as extra-adrenal manifestations may occur due to the sensitivity to oxidative stress of other organs such as the heart, thyroid, liver, kidney and pancreas.


Subject(s)
Adrenal Insufficiency/congenital , Addison Disease/genetics , Adrenal Glands/abnormalities , Adrenal Glands/embryology , Adrenal Insufficiency/classification , Adrenal Insufficiency/genetics , Glucocorticoids/deficiency , Glucocorticoids/genetics , Humans , Mineralocorticoids/deficiency , Mineralocorticoids/genetics , Mutation , Receptors, Glucocorticoid/genetics , Receptors, Mineralocorticoid/genetics , Syndrome
4.
J Mol Endocrinol ; 55(1): 55-68, 2015 Aug.
Article in English | MEDLINE | ID: mdl-26108487

ABSTRACT

The salmonid corticosteroid receptors (CRs), glucocorticoid receptors 1 and 2 (GR1 and GR2) and the mineralocorticoid receptor (MR) share a high degree of homology with regard to structure, ligand- and DNA response element-binding, and cellular co-localization. Typically, these nuclear hormone receptors homodimerize to confer transcriptional activation of target genes, but a few studies using mammalian receptors suggest some degree of heterodimerization. We observed that the trout MR confers a several fold lower transcriptional activity compared to the trout GRs. This made us question the functional relevance of the MR when this receptor is located in the same cells as the GRs and activated by cortisol. A series of co-transfection experiments using different glucocorticoid response elements (GREs) containing promoter-reporter constructs were carried out to investigate any possible interaction between the piscine CRs. Co-transfection of the GRs with the MR significantly reduced the total transcriptional activity even at low MR levels, suggesting interaction between these receptors. Co-transfection of GR1 or GR2 with the MR did not affect the subcellular localization of the GRs, and the MR-mediated inhibition seemed to be independent of specific activation or inhibition of the MR. Site-directed mutagenesis of the DNA-binding domain and dimerization interface of the MR showed that the inhibition was dependent on DNA binding but not necessarily on dimerization ability. Thus, we suggest that the interaction between MR and the GRs may regulate the cortisol response in cell types where the receptors co-localize and propose a dominant-negative role for the MR in cortisol-mediated transcriptional activity.


Subject(s)
Mineralocorticoids/metabolism , Receptors, Glucocorticoid/metabolism , Trout/metabolism , Animals , COS Cells , Cell Line , Chlorocebus aethiops , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Glucocorticoids/genetics , Glucocorticoids/metabolism , Hydrocortisone/genetics , Hydrocortisone/metabolism , Mineralocorticoids/genetics , Mutagenesis, Site-Directed/methods , Promoter Regions, Genetic/genetics , Receptors, Glucocorticoid/genetics , Receptors, Mineralocorticoid/genetics , Receptors, Mineralocorticoid/metabolism , Receptors, Steroid/genetics , Receptors, Steroid/metabolism , Response Elements/genetics , Transcription, Genetic/genetics , Transcriptional Activation/genetics , Transfection/methods , Trout/genetics
5.
Ugeskr Laeger ; 176(25A)2014 Dec 15.
Article in Danish | MEDLINE | ID: mdl-25497610

ABSTRACT

Ventricular fibrillation is an unknown complication to the syndrome of apparent mineralocorticoid excess (SAME). This case report describes a young woman admitted with hypo-kalaemia and hypertension. Concentrations of both P-renin and P-aldosterone were low and urinary steroid metabolites revealed an abnormal excretion pattern pointing to the diagnosis of SAME. Three years later the woman suffered from ventricular fibrillation due to the hypokalaemia caused by her disease. This case report demonstrates the need for increased attention on the potassium concentration in patients with SAME.


Subject(s)
Mineralocorticoid Excess Syndrome, Apparent/complications , Ventricular Fibrillation/etiology , Adult , Female , Humans , Hypertension/drug therapy , Hypertension/etiology , Hypokalemia/drug therapy , Hypokalemia/etiology , Mineralocorticoid Excess Syndrome, Apparent/drug therapy , Mineralocorticoids/genetics , Mineralocorticoids/pharmacokinetics , Ventricular Fibrillation/therapy
6.
Proc Natl Acad Sci U S A ; 110(42): 17035-40, 2013 Oct 15.
Article in English | MEDLINE | ID: mdl-24082121

ABSTRACT

Heart failure is a leading cause of death in humans, and stress is increasingly associated with adverse cardiac outcomes. Glucocorticoids are primary stress hormones, but their direct role in cardiovascular health and disease is poorly understood. To determine the in vivo function of glucocorticoid signaling in the heart, we generated mice with cardiomyocyte-specific deletion of the glucocorticoid receptor (GR). These mice are born at the expected Mendelian ratio, but die prematurely from spontaneous cardiovascular disease. By 3 mo of age, mice deficient in cardiomyocyte GR display a marked reduction in left ventricular systolic function, as evidenced by decreases in ejection fraction and fractional shortening. Heart weight and left ventricular mass are elevated, and histology revealed cardiac hypertrophy without fibrosis. Removal of endogenous glucocorticoids and mineralocorticoids neither augmented nor lessened the hypertrophic response. Global gene expression analysis of knockout hearts before pathology onset revealed aberrant regulation of a large cohort of genes associated with cardiovascular disease as well as unique disease genes associated with inflammatory processes. Genes important for maintaining cardiac contractility, repressing cardiac hypertrophy, promoting cardiomyocyte survival, and inhibiting inflammation had decreased expression in the GR-deficient hearts. These findings demonstrate that a deficiency in cardiomyocyte glucocorticoid signaling leads to spontaneous cardiac hypertrophy, heart failure, and death, revealing an obligate role for GR in maintaining normal cardiovascular function. Moreover, our findings suggest that selective activation of cardiomyocyte GR may represent an approach for the prevention of heart disease.


Subject(s)
Cardiomegaly/metabolism , Cardiomegaly/prevention & control , Glucocorticoids/metabolism , Mineralocorticoids/metabolism , Myocardium/metabolism , Receptors, Glucocorticoid/metabolism , Receptors, Glucocorticoid/physiology , Signal Transduction , Aging/genetics , Aging/metabolism , Aging/pathology , Animals , Cardiomegaly/genetics , Cardiomegaly/pathology , Cell Survival , Glucocorticoids/genetics , Mice , Mice, Knockout , Mineralocorticoids/genetics , Myocardium/pathology , Myocytes, Cardiac/metabolism , Myocytes, Cardiac/pathology , Organ Specificity/genetics , Receptors, Glucocorticoid/genetics
7.
Eur J Endocrinol ; 169(1): R15-25, 2013 Jul.
Article in English | MEDLINE | ID: mdl-23610123

ABSTRACT

Aldosterone plays a major role in the regulation of sodium and potassium homeostasis and blood pressure. More recently, aldosterone has emerged as a key hormone mediating end organ damage. In extreme cases, dysregulated aldosterone production leads to primary aldosteronism (PA), the most common form of secondary hypertension. However, even within the physiological range, high levels of aldosterone are associated with an increased risk of developing hypertension over time. PA represents the most common and curable form of hypertension, with a prevalence that increases with the severity of hypertension. Although genetic causes underlying glucocorticoid-remediable aldosteronism, one of the three Mendelian forms of PA, were established some time ago, somatic and inherited mutations in the potassium channel GIRK4 have only recently been implicated in the formation of aldosterone-producing adenoma (APA) and in familial hyperaldosteronism type 3. Moreover, recent findings have shown somatic mutations in two additional genes, involved in maintaining intracellular ionic homeostasis and cell membrane potential, in a subset of APAs. This review summarizes our current knowledge on the genetic determinants that contribute to variations in plasma aldosterone and renin levels in the general population and the genetics of familial and sporadic PA. Various animal models that have significantly improved our understanding of the pathophysiology of excess aldosterone production are also discussed. Finally, we outline the cardiovascular, renal, and metabolic consequences of mineralocorticoid excess beyond blood pressure regulation.


Subject(s)
Aldosterone/metabolism , G Protein-Coupled Inwardly-Rectifying Potassium Channels/genetics , Hyperaldosteronism/complications , Hyperaldosteronism/genetics , Hypertension/etiology , Renin/metabolism , Adrenal Cortex Neoplasms/metabolism , Adrenocortical Adenoma/metabolism , Aldosterone/biosynthesis , Aldosterone/blood , Aldosterone/genetics , Animals , Disease Models, Animal , Humans , Hyperaldosteronism/blood , Hyperaldosteronism/metabolism , Hypertension/blood , Hypertension/genetics , Mineralocorticoids/genetics , Mutation , Plasma Membrane Calcium-Transporting ATPases/genetics , Renin/blood , Renin/genetics , Sodium-Potassium-Exchanging ATPase/genetics
11.
Med Klin (Munich) ; 98(4): 208-17, 2003 Apr 15.
Article in German | MEDLINE | ID: mdl-12715144

ABSTRACT

Four types of monogenic hypertension belong to the group of mineralocorticoid hypertension, which are characterized by high renal water and sodium retention and resulting suppression of plasma renin activity (PRA), high urinary potassium secretion and consecutive low plasma potassium:1. increased production of the hormone aldosterone: glucocorticoid-remediable aldosteronism (GRH), 2. prereceptor disorder with loss of selectivity of the mineralocorticoid receptor: apparent mineralocorticoid excess (AME), 3. receptor disorder with constitutive activation of the mineralocorticoid receptor: "Geller syndrome", 4. postreceptor disorder with enhanced function of the epithelial sodium channel: Liddle's syndrome. While in GRH high synthesis of aldosterone results in high plasma aldosterone and low PRA, in the primary renal malfunctions of the AME, constitutive activation of the mineralocorticoid receptor and the Liddle's syndrome both plasma aldosterone and PRA are low. These forms of hypertension are rather rare in their complete expression, but they point to candidate genes whose mutations may predispose to hypertension. A point mutation of the ENaC beta-subunit (T594M) occurs rather frequent in people of African origin, with 5%. Therefore it is suggested to analyze the genotype of black hypertensive patients as a prerequisite for a rational amiloride therapy. Contrarily, the rather frequent (A[2139]G) polymorphism of the promoter of the alpha-subunit is supposed to mark a lower risk of hypertension. Mutations in the serine-threonine kinases WNK1 or WNK4 cause pseudohypoaldosteronism type II. WNK1 and WNK4 are expressed in the distal part of the nephron. Stimulation of sodium reabsorption by aldosterone is normal but without influence on hyperkalemia. An extrarenal disorder is suggested to be the cause of autosomal-dominant hypertension with brachydactyly: the patients react with a severely impaired baroreflex und show neurovascular contact. The mutation causing this syndrome is not known.


Subject(s)
Hypertension/genetics , Adolescent , Adult , Aldosterone/blood , Child , Female , Genes, Dominant , Genotype , Humans , Hyperaldosteronism/complications , Hyperaldosteronism/genetics , Hyperaldosteronism/physiopathology , Hypertension/blood , Hypertension/drug therapy , Hypertension/etiology , Hypertension/physiopathology , Male , Mineralocorticoids/genetics , Phenotype , Point Mutation , Polymorphism, Genetic , Pseudohypoaldosteronism/genetics , Pseudohypoaldosteronism/physiopathology , Receptors, Mineralocorticoid/genetics , Receptors, Mineralocorticoid/physiology , Renin/blood , Risk Factors , Sodium Channels/physiology , Syndrome
13.
Curr Hypertens Rep ; 4(1): 18-24, 2002 Feb.
Article in English | MEDLINE | ID: mdl-11790287

ABSTRACT

Abnormalities in steroid biosynthesis have been known for years to cause hypertension in some cases of congenital adrenal hyperplasia. In these patients hypertension usually accompanies a characteristic phenotype with abnormal sexual differentiation. Recently, the molecular basis of four forms of severe hypertension transmitted on an autosomal basis but without additional phenotypic features has been elucidated. All these conditions are characterized primarily by low plasma renin, normal or low serum potassium, and salt-sensitive hypertension, indicating an increased mineralocorticoid effect. These four disorders, the glucocorticoid remediable aldosteronism, the syndrome of apparent mineralocorticoid excess, the activating mutation of the mineralocorticoid receptor, and the Liddle syndrome are a consequence of either abnormal biosynthesis, metabolism, or action of steroid hormones, and are ultimately characterized by an overactivation of the epithelial sodium channel in distal renal tubules. Hyperactivity of this channel results in increased sodium reabsorption and volume expansion leading to an increase in blood pressure as well as potassium loss. With the advent of molecular biology in clinical practice, it has become evident that some genetic defect may present with a more discrete phenotype, with only moderate hypertension with or without hypokalemia as the sole feature. A search for genetic disorders of the mineralocorticoid axis should be an integral part of the diagnostic work-up, particularly in young adults with hypertension.


Subject(s)
Adrenal Glands/physiology , Cytochrome P-450 CYP11B2/genetics , Hydroxysteroid Dehydrogenases/genetics , Hypertension/genetics , Mineralocorticoids/genetics , Steroid 11-beta-Hydroxylase/genetics , Aldosterone/metabolism , Humans , Hydrocortisone/metabolism , Hydroxysteroid Dehydrogenases/metabolism , Hypertension/metabolism , Mineralocorticoids/metabolism , Mutation , Potassium/blood , Receptors, Mineralocorticoid/genetics , Renin/blood , Sodium Channels/genetics
16.
Curr Hypertens Rep ; 2(3): 295-301, 2000 Jun.
Article in English | MEDLINE | ID: mdl-10981163

ABSTRACT

Our basic understanding of Na(+) transport mechanisms provides unique insights into epithelial transport processes. Unusual clinical syndromes can arise from mutations of these ion transporters. These genetic disorders affect Na(+) balance, resulting in both N(a+) retaining and Na(+) wasting conditions. A major focus has been the epithelial sodium channel (ENaC), which can be activated by mutations (eg, Liddle's syndrome), changes in the response to mineralocorticoids (apparent mineralocorticoid excess syndrome), or production of mineralocorticoids (glucocorticoid-remediable aldosteronism). As a result, we now have clearly defined Mendelian syndromes in which ENaC activity is "dysregulated." This dysregulation leads to systemic hypertension associated with suppressed plasma renin activity, which can be attributed to a primary renal mechanism. Applying these insights to the far more common disorder of low-renin hypertension may shed new light on the underlying pathophysiology of this common form of human hypertension.


Subject(s)
Aldosterone/genetics , Hypertension/genetics , Cytochrome P-450 CYP11B2/genetics , Epithelium/metabolism , Humans , Hyperaldosteronism/genetics , Hypertension/blood , Hypertension/metabolism , Hypertension/physiopathology , Ion Transport/genetics , Kidney/metabolism , Mineralocorticoids/genetics , Mutation/genetics , Renin/blood , Renin/physiology , Sodium/metabolism , Sodium Channels/genetics , Syndrome
17.
Rev Med Chil ; 128(1): 17-26, 2000 Jan.
Article in Spanish | MEDLINE | ID: mdl-10883518

ABSTRACT

An 11-year old girl was seen in 1981 with hypokalemia, low renin, low aldosterone, and severe hypertension. A medical adrenalectomy with dexamethasone and aminoglutethimide, and the blockade of mineralocorticoid receptors with spironolactone improved her condition, but the blockade of glucocorticoid receptors with RU-486 worsened it. An aldosterone infusion induced no changes. A sister was born in 1982 with similar findings. Both patients had an impaired ability to convert cortisol to cortisone after an oral load of 200 mg cortisol. In urine, an elevated ratio for metabolites of cortisol to metabolites of cortisone was found. These data suggested a defect in the activity of renal 11 beta-hydroxysteroid dehydrogenase. Both parents were asymptomatic, phenotypically normal and non-consanguineous. Their urinary metabolites of cortisol and cortisone were normal before and after stimulation with ACTH. However, the mother reached a peak plasma cortisone concentration 3 SD below the mean reached by normal subjects after an oral 200-mg cortisol load, a fact that suggests that this test could be used to detect heterozygotes. The genetic studies revealed a homozygous mutation on exon 3 of the HSD11K gene, which by substituting TGC for CGC changes Arg 213 for Cys and induces a loss of 84% of the enzymatic activity in transfected cells. Both unrelated parents had the same heterozygous mutation. Both patients have been treated with dexamethasone but have also required spironolactone. The older sister has also required high doses of nifedipine to lower her blood pressure. After 19 years of follow-up, the older sister has become normotensive and normokalemic under therapy, and reached a final height of 140 cm at age 17. The younger sister has increased her mean blood pressure at a rate of 1 mm Hg per year, in spite of treatment. Her final height is 143.5 cm.


Subject(s)
Cortisone/metabolism , Hydrocortisone/metabolism , Hydroxysteroid Dehydrogenases/deficiency , Hypertension/genetics , Hypokalemia/genetics , Mineralocorticoids/metabolism , 11-beta-Hydroxysteroid Dehydrogenase Type 1 , Adolescent , Aminoglutethimide/therapeutic use , Blood Pressure , Body Height , Child , Child, Preschool , Dexamethasone/therapeutic use , Enzyme Inhibitors/therapeutic use , Female , Follow-Up Studies , Humans , Hypertension/drug therapy , Hypertension/metabolism , Hypokalemia/drug therapy , Hypokalemia/metabolism , Infant , Infant, Newborn , Male , Mineralocorticoids/genetics , Mutation/genetics , Syndrome
18.
Hypertension ; 34(4 Pt 1): 638-42, 1999 Oct.
Article in English | MEDLINE | ID: mdl-10523339

ABSTRACT

Mutations in the kidney isozyme of human 11-hydroxysteroid dehydrogenase (11-HSD2) cause apparent mineralocorticoid excess, an autosomal recessive form of familial hypertension. We studied 4 patients with AME, identifying 4 novel and 3 previously reported mutations in the HSD11B2 (HSD11K) gene. Point mutations causing amino acid substitutions were introduced into a pCMV5/11HSD2 expression construct and expressed in mammalian CHOP cells. Mutations L179R and R208H abolished activity in whole cells. Mutants S180F, A237V, and A328V had 19%, 72%, and 25%, respectively, of the activity of the wild-type enzyme in whole cells when cortisol was used as the substrate and 80%, 140%, and 55%, respectively, of wild-type activity when corticosterone was used as the substrate. However, these mutant proteins were only 0.6% to 5.7% as active as the wild-type enzyme in cell lysates, suggesting that these mutations alter stability of the enzyme. In regression analyses of all AME patients with published genotypes, several biochemical and clinical parameters were highly correlated with mutant enzymatic activity, demonstrated in whole cells, when cortisol was used as the substrate. These included the ratio of urinary cortisone to cortisol metabolites (R(2)=0.648, P<0.0001), age at presentation (R(2)=0.614, P<0.0001), and birth weight (R(2)=0.576, P=0.0004). Approximately 5% conversion of cortisol to cortisone is predicted in subjects with mutations that completely inactivate HSD11B2, suggesting that a low level of enzymatic activity is mediated by another enzyme, possibly 11-HSD1.


Subject(s)
Hydroxysteroid Dehydrogenases/genetics , Mineralocorticoids/metabolism , Mutation , 11-beta-Hydroxysteroid Dehydrogenases , Adolescent , Blood Pressure , Child, Preschool , Chromosomes, Human, Pair 16 , Exons , Female , Genotype , Humans , Hydrocortisone/metabolism , Hydroxysteroid Dehydrogenases/metabolism , Hypertension/genetics , Hypertension/metabolism , Infant , Isoenzymes/genetics , Isoenzymes/metabolism , Male , Mineralocorticoids/genetics , Phenotype , Polymerase Chain Reaction
19.
Rev Med Chil ; 127(5): 511-3, 1999 May.
Article in Spanish | MEDLINE | ID: mdl-10451619

ABSTRACT

There is a group of genetic alterations that are phenotypically related to mineralocorticoid hypertension. They include, among others, some forms of primary hyperaldosteronism and of hyporeninemic aldosteronism that can be specifically treated, thus becoming secondary forms of hypertension. These could account for 10 to 15% of cases of essential hypertension, but more studies are required to accept these figures. The screening for these forms of hypertension should be done measuring aldosterone levels and plasma renin activity. An aldosterone/plasma renin activity ratio over 25 should lead to the suspicion of the disease. However, the cost effectiveness of the widespread measurement of these parameters would be very low. Therefore it is mandatory to determine the epidemiological features of these diseases to perform a selective screening among subjects with essential hypertension.


Subject(s)
Hyperaldosteronism/complications , Hypertension/etiology , Mineralocorticoids , Renin-Angiotensin System/genetics , Aldosterone/blood , Humans , Hyperaldosteronism/genetics , Mineralocorticoids/genetics , Renin/blood
20.
Rev Med Chil ; 127(5): 604-10, 1999 May.
Article in Spanish | MEDLINE | ID: mdl-10451632

ABSTRACT

Recently, some genetic forms of hypertension have been well characterized. These forms can be globally called mineralocorticoid hypertension and are due to different alterations of the renin-angiotensin-aldosterone system (SRAA). Among these, classic primary hyperaldosteronism and its glucocorticoid remediable variety, in which hypertension is secondary to aldosterone production, must be considered. There are also conditions in which mineralocorticoid activity does not depend on aldosterone production. These conditions generate a hyporeninemic hyperaldosteronism, observed in Liddle syndrome, apparent mineralocorticoid hypertension, 11- and 17-hydroxylase deficiency, among others. The detection of these forms of hypertension is only feasible if the renin-angiotensin-aldosterone system is assessed, measuring renin and aldosterone levels. This article reviews these forms of hypertension, their clinical workup and their relevance in the usual hypertensive patients.


Subject(s)
Hyperaldosteronism/complications , Hypertension/etiology , Mineralocorticoids , Aldosterone/blood , Electrolytes/blood , Humans , Hypoaldosteronism/physiopathology , Mineralocorticoids/genetics , Mineralocorticoids/physiology , Renin/blood , Renin-Angiotensin System/physiology
SELECTION OF CITATIONS
SEARCH DETAIL