Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 20 de 621
Filtrer
1.
BMC Endocr Disord ; 24(1): 116, 2024 Jul 16.
Article de Anglais | MEDLINE | ID: mdl-39010034

RÉSUMÉ

BACKGROUND: Somatic mutations have been observed to induce aldosterone-producing adenomas (APAs). These may be accelerated during pregnancy. Somatic PRKACA mutations are common in cortisol-producing adenomas (CPAs). However, their role in APAs, particularly aldosterone- and cortisol-producing adenomas (A/CPAs), is not well understood. This study aims to investigate the association between PRKACA mutations and the accelerated development of A/CPAs during pregnancy. CASE PRESENTATION: A patient with primary aldosteronism (PA) associated with severe Cushing's syndrome (CS) underwent surgical resection of an adrenal tumor one year after delivery. Pathologic examination revealed an adrenocortical adenoma characterized primarily by zona glomerulosa hyperplasia. Somatic mutation analysis revealed the presence of the somatic PRKACA mutation, which was validated as a deleterious mutation by various computational databases. Immunohistochemical results showed positive staining for cytochrome P450 family 11 subfamily B member 1 (CYP11B1), cytochrome P450 family 11 subfamily B member 2 (CYP11B2), and luteinizing hormone/chorionic gonadotropin receptor (LHCGR). Our study included a review of 20 previously documented cases of aldosterone- and cortisol-producing adenomas (A/CPAs), two of which were concurrently positive for both CYP11B1 and CYP11B2, consistent with our findings. CONCLUSION: Somatic mutations in PRKACA may correlate with the upregulation of LHCGR, which synergistically drives the accelerated growth of co-secretion tumors during pregnancy, thereby exacerbating disease progression.


Sujet(s)
Tumeurs corticosurrénaliennes , Adénome corticosurrénalien , Aldostérone , Cyclic AMP-Dependent Protein Kinase Catalytic Subunits , Hydrocortisone , Mutation , Complications tumorales de la grossesse , Humains , Femelle , Grossesse , Adulte , Hydrocortisone/métabolisme , Adénome corticosurrénalien/génétique , Adénome corticosurrénalien/anatomopathologie , Adénome corticosurrénalien/métabolisme , Adénome corticosurrénalien/chirurgie , Tumeurs corticosurrénaliennes/génétique , Tumeurs corticosurrénaliennes/anatomopathologie , Tumeurs corticosurrénaliennes/métabolisme , Aldostérone/métabolisme , Cyclic AMP-Dependent Protein Kinase Catalytic Subunits/génétique , Complications tumorales de la grossesse/génétique , Complications tumorales de la grossesse/anatomopathologie , Hyperaldostéronisme/génétique , Hyperaldostéronisme/anatomopathologie , Hyperaldostéronisme/chirurgie , Syndrome de Cushing/génétique , Syndrome de Cushing/anatomopathologie , Adénomes/génétique , Adénomes/anatomopathologie , Adénomes/métabolisme
3.
Eur J Endocrinol ; 190(4): G1-G14, 2024 Mar 30.
Article de Anglais | MEDLINE | ID: mdl-38571460

RÉSUMÉ

We describe herein the European Reference Network on Rare Endocrine Conditions clinical practice guideline on diagnosis and management of familial forms of hyperaldosteronism. The guideline panel consisted of 10 experts in primary aldosteronism, endocrine hypertension, paediatric endocrinology, and cardiology as well as a methodologist. A systematic literature search was conducted, and because of the rarity of the condition, most recommendations were based on expert opinion and small patient series. The guideline includes a brief description of the genetics and molecular pathophysiology associated with each condition, the patients to be screened, and how to screen. Diagnostic and treatment approaches for patients with genetically determined diagnosis are presented. The recommendations apply to patients with genetically proven familial hyperaldosteronism and not to families with more than one case of primary aldosteronism without demonstration of a responsible pathogenic variant.


Sujet(s)
Endocrinologie , Hyperaldostéronisme , Hypertension artérielle , Enfant , Humains , Hyperaldostéronisme/diagnostic , Hyperaldostéronisme/génétique , Hyperaldostéronisme/thérapie , Hypertension artérielle/diagnostic , Hypertension artérielle/génétique , Hypertension artérielle/thérapie
4.
Endocrinology ; 165(5)2024 Mar 29.
Article de Anglais | MEDLINE | ID: mdl-38573585

RÉSUMÉ

Klotho plays a critical role in the regulation of ion and fluid homeostasis. A previous study reported that haplo-insufficiency of Klotho in mice results in increased aldosterone synthase (CYP11B2) expression, elevated plasma aldosterone, and high blood pressure. This phenotype was presumed to be the result of diminished Klotho expression in zona glomerulosa (zG) cells of the adrenal cortex; however, systemic effects on adrenal aldosterone production could not be ruled out. To examine whether Klotho expressed in the zG is indeed a critical regulator of aldosterone synthesis, we generated a tamoxifen-inducible, zG-specific mouse model of Klotho deficiency by crossing Klotho-flox mice with Cyp11b2-CreERT mice (zG-Kl-KO). Tamoxifen-treated Cyp11b2-CreERT animals (zG-Cre) served as controls. Rosa26-mTmG reporter mice were used for Cre-dependent lineage-marking. Two weeks after tamoxifen induction, the specificity of the zG-Cre line was verified using immunofluorescence analysis to show that GFP expression was restricted to the zG. RNA in situ hybridization revealed a 65% downregulation of Klotho messenger RNA expression in the zG of zG-Kl-KO female mice at age 12 weeks compared to control mice. Despite this significant decrease, zG-Kl-KO mice exhibited no difference in plasma aldosterone levels. However, adrenal CYP11B2 expression and the CYP11B2 promotor regulatory transcription factors, NGFIB and Nurr1, were enhanced. Together with in vitro experiments, these results suggest that zG-derived Klotho modulates Cyp11b2 but does not evoke a systemic phenotype in young adult mice on a normal diet. Further studies are required to investigate the role of adrenal Klotho on aldosterone synthesis in aged animals.


Sujet(s)
Cortex surrénal , Hyperaldostéronisme , Femelle , Souris , Animaux , Zone glomérulée/métabolisme , Cytochrome P-450 CYP11B2/génétique , Cytochrome P-450 CYP11B2/métabolisme , Aldostérone/métabolisme , Cortex surrénal/métabolisme , Hyperaldostéronisme/génétique , Tamoxifène/pharmacologie
5.
Endocr Pract ; 30(7): 670-678, 2024 Jul.
Article de Anglais | MEDLINE | ID: mdl-38657793

RÉSUMÉ

BACKGROUND: While clinical features of KCNJ5-mutated aldosterone-producing adenoma (APA) have been reported, evidence of its clinical outcomes is lacking. We aimed to synthesize available literature about the associations between KCNJ5 mutation with cardiovascular and metabolic outcomes among patients with APA. METHODS: In this systematic review of observational studies, MEDLINE and Embase were searched through August 2022. Two independent authors screened the search results and extracted data from eligible observational studies investigating cardiovascular or metabolic outcomes between KCNJ5-mutated APAs and KCNJ5-non-mutated APAs. Risk of Bias In Non-randomized Studies of Interventions was used to assess the quality of the included studies. RESULTS: A total of 573 titles/abstracts were screened and after the expert opinion of the literature, full text was read in 20 titles/abstracts, of which 12 studies were included. Across 3 studies comparing the baseline or change in the cardiac function between KCNJ5-mutated APAs and KCNJ5-non-mutated APAs, all studies reported the association between impaired cardiac functions and KCNJ5 mutation status. Among 6 studies evaluating the cure of hypertension after surgery, all studies showed that KCNJ5 mutation was significantly associated with the cure of hypertension. In quality assessment, 7 studies were at serious risk of bias, while the remaining studies were at moderate risk of bias. CONCLUSIONS: This systematic review provided evidence of the significant association between KCNJ5 mutation and unfavorable cardiovascular outcomes in patients with primary aldosteronism. Further research is needed to improve the quality of evidence on this topic and elucidate the underlying mechanisms of the potential burden of KCNJ5 mutation.


Sujet(s)
Aldostérone , Canaux potassiques rectifiants entrants couplés aux protéines G , Mutation , Humains , Canaux potassiques rectifiants entrants couplés aux protéines G/génétique , Aldostérone/métabolisme , Aldostérone/biosynthèse , Maladies cardiovasculaires/génétique , Tumeurs corticosurrénaliennes/génétique , Hyperaldostéronisme/génétique , Adénome corticosurrénalien/génétique , Adénome corticosurrénalien/métabolisme , Adénomes/génétique , Adénomes/métabolisme
6.
Front Endocrinol (Lausanne) ; 15: 1286297, 2024.
Article de Anglais | MEDLINE | ID: mdl-38505749

RÉSUMÉ

Double somatic mutations in CTNNB1 and GNA11/Q have recently been identified in a small subset of aldosterone-producing adenomas (APAs). As a possible pathogenesis of APA due to these mutations, an association with pregnancy, menopause, or puberty has been proposed. However, because of its rarity, characteristics of APA with these mutations have not been well characterized. A 46-year-old Japanese woman presented with hypertension and hypokalemia. She had two pregnancies in the past but had no history of pregnancy-induced hypertension. She had regular menstrual cycle at presentation and was diagnosed as having primary aldosteronism after endocrinologic examinations. Computed tomography revealed a 2 cm right adrenal mass. Adrenal venous sampling demonstrated excess aldosterone production from the right adrenal gland. She underwent right laparoscopic adrenalectomy. The resected right adrenal tumor was histologically diagnosed as adrenocortical adenoma and subsequent immunohistochemistry (IHC) revealed diffuse immunoreactivity of aldosterone synthase (CYP11B2) and visinin like 1, a marker of the zona glomerulosa (ZG), whereas 11ß-hydroxylase, a steroidogenic enzyme for cortisol biosynthesis, was mostly negative. CYP11B2 IHC-guided targeted next-generation sequencing identified somatic CTNNB1 (p.D32Y) and GNA11 (p.Q209H) mutations. Immunofluorescence staining of the tumor also revealed the presence of activated ß-catenin, consistent with features of the normal ZG. The expression patterns of steroidogenic enzymes and related proteins indicated ZG features of the tumor cells. PA was clinically and biochemically cured after surgery. In conclusion, our study indicated that CTNNB1 and GNA11-mutated APA has characteristics of the ZG. The disease could occur in adults with no clear association with pregnancy or menopause.


Sujet(s)
Adénomes , Adénome corticosurrénalien , Hyperaldostéronisme , Hypertension artérielle , Adulte , Femelle , Grossesse , Humains , Adulte d'âge moyen , Adénome corticosurrénalien/complications , Adénome corticosurrénalien/génétique , Adénome corticosurrénalien/chirurgie , Aldostérone/métabolisme , Cytochrome P-450 CYP11B2/métabolisme , bêta-Caténine/génétique , bêta-Caténine/métabolisme , Hyperaldostéronisme/génétique , Hyperaldostéronisme/chirurgie , Adénomes/génétique , Adénomes/chirurgie , Adénomes/métabolisme , Hypertension artérielle/complications , Mutation , Sous-unités alpha des protéines G/génétique , Sous-unités alpha des protéines G/métabolisme
7.
Front Endocrinol (Lausanne) ; 15: 1336306, 2024.
Article de Anglais | MEDLINE | ID: mdl-38495792

RÉSUMÉ

Purpose: To compare the clinical and hormonal characteristics of patients with familial hyperaldosteronism (FH) and sporadic primary aldosteronism (PA). Methods: A systematic review of the literature was performed for the identification of FH patients. The SPAIN-ALDO registry cohort of patients with no suspicion of FH was chosen as the comparator group (sporadic group). Results: A total of 360 FH (246 FH type I, 73 type II, 29 type III, and 12 type IV) cases and 830 sporadic PA patients were included. Patients with FH-I were younger than sporadic cases, and women were more commonly affected (P = 0.003). In addition, the plasma aldosterone concentration (PAC) was lower, plasma renin activity (PRA) higher, and hypokalemia (P < 0.001) less frequent than in sporadic cases. Except for a younger age (P < 0.001) and higher diastolic blood pressure (P = 0.006), the clinical and hormonal profiles of FH-II and sporadic cases were similar. FH-III had a distinct phenotype, with higher PAC and higher frequency of hypokalemia (P < 0.001), and presented 45 years before sporadic cases. Nevertheless, the clinical and hormonal phenotypes of FH-IV and sporadic cases were similar, with the former being younger and having lower serum potassium levels. Conclusion: In addition to being younger and having a family history of PA, FH-I and III share other typical characteristics. In this regard, FH-I is characterized by a low prevalence of hypokalemia and FH-III by a severe aldosterone excess causing hypokalemia in more than 85% of patients. The clinical and hormonal phenotype of type II and IV is similar to the sporadic cases.


Sujet(s)
Hyperaldostéronisme , Hypokaliémie , Femelle , Humains , Aldostérone , Hyperaldostéronisme/complications , Hyperaldostéronisme/génétique , Hyperaldostéronisme/épidémiologie , Hypokaliémie/étiologie , Potassium
8.
Vitam Horm ; 124: 137-163, 2024.
Article de Anglais | MEDLINE | ID: mdl-38408798

RÉSUMÉ

Aldosterone is a cardiovascular hormone with a key role in blood pressure regulation, among other processes, mediated through its targeting of the mineralocorticoid receptor in the renal tubule and selected other tissues. Its secretion from the adrenal gland is a highly controlled process subject to regulatory influence from the renin-angiotensin system and the hypothalamic-pituitary-adrenal axis. MicroRNAs are small endogenous non-coding RNA molecules capable of regulating gene expression post-transcriptionally through stimulation of mRNA degradation or suppression of translation. Several studies have now identified that microRNA levels are changed in cases of aldosterone dysregulation and that microRNAs are capable of regulating the expression of various genes involved in aldosterone production and action. In this article we summarise the major studies concerning this topic. We also discuss the potential role for circulating microRNAs as diagnostic biomarkers for primary aldosteronism, a highly treatable form of secondary hypertension, which would be highly desirable given the current underdiagnosis of this condition.


Sujet(s)
Hyperaldostéronisme , Hypertension artérielle , microARN , Humains , Aldostérone/métabolisme , microARN/génétique , Hyperaldostéronisme/diagnostic , Hyperaldostéronisme/génétique , Axe hypothalamohypophysaire/métabolisme , Axe hypophyso-surrénalien/métabolisme , Hypertension artérielle/génétique
9.
Hypertension ; 81(4): 936-945, 2024 Apr.
Article de Anglais | MEDLINE | ID: mdl-38318706

RÉSUMÉ

BACKGROUND: Primary aldosteronism (PA) is frequently caused by a unilateral aldosterone-producing adenoma with a PA-driver mutation. Unilateral adrenalectomy has a high probability of short-term biochemical remission, but long-term postsurgical outcomes are relatively undefined. Our objective was to investigate the incidence of long-term recurrence of PA in individuals with postsurgical short-term biochemical remission. METHODS: Adrenalectomized patients for unilateral PA were included from a single referral center. Histopathology and outcomes were assessed according to international histopathology of unilateral primary aldosteronism and PASO (Primary Aldosteronism Surgical Outcome) consensuses. Genotyping was performed using CYP11B2 (aldosterone synthase)-guided sequencing. RESULTS: Classical adrenal histopathology, exemplified by a solitary aldosterone-producing adenoma, was observed in 78% of 90 adrenals, compared with 22% with nonclassical histopathology. The classical group displayed higher aldosterone-to-renin ratios (P=0.013) and lower contralateral ratios (P=0.008). Outcome assessments at both short (12 months [7; 12]) and long (89 months [48; 124]) terms were available for 57 patients. At short-term assessment, 53 (93%) displayed complete biochemical success (43 classical and 10 nonclassical), but long-term assessment demonstrated biochemical PA recurrence in 12 (23%) with an overrepresentation of the nonclassical histopathology (6 [60%] of 10 nonclassical histopathology versus 6 [14%] of 43 classical histopathology; P=0.005). PA-driver mutations were identified in 97% of 64 aldosterone-producing adenomas; there was no association of the aldosterone-producing adenoma genotype with PA recurrence. CONCLUSIONS: A substantial proportion of individuals display postsurgical biochemical recurrence of PA, which is related to the histopathology of the resected adrenal gland. These findings emphasize the role of histopathology and the requirement for continued outcome assessment in the management of surgically treated patients for PA.


Sujet(s)
Adénomes , Adénome corticosurrénalien , Hyperaldostéronisme , Humains , Surrénalectomie , Aldostérone , Récidive tumorale locale/chirurgie , Hyperaldostéronisme/diagnostic , Hyperaldostéronisme/génétique , Hyperaldostéronisme/chirurgie , Adénome corticosurrénalien/génétique , Adénome corticosurrénalien/chirurgie , Adénomes/chirurgie , Études rétrospectives
10.
Hypertension ; 81(3): 604-613, 2024 Mar.
Article de Anglais | MEDLINE | ID: mdl-38174562

RÉSUMÉ

BACKGROUND: Primary aldosteronism (PA) has been broadly dichotomized into unilateral and bilateral forms. Adrenal vein sampling (AVS) lateralization indices (LI) ≥2 to 4 are the standard-of-care to recommend unilateral adrenalectomy for presumed unilateral PA. We aimed to assess the rates and characteristics of residual PA after AVS-guided adrenalectomy. METHODS: We conducted an international, retrospective, cohort study of patients with PA from 7 referral centers who underwent unilateral adrenalectomy based on LI≥4 on baseline and/or cosyntropin-stimulated AVS. Aldosterone synthase (CYP11B2) immunohistochemistry and next generation sequencing were performed on available formalin-fixed paraffin-embedded adrenal tissue. RESULTS: The cohort included 283 patients who underwent AVS-guided adrenalectomy, followed for a median of 326 days postoperatively. Lack of PA cure was observed in 16% of consecutive patients, and in 22 patients with lateralized PA on both baseline and cosyntropin-stimulated AVS. Among patients with residual PA postoperatively, 73% had multiple CYP11B2 positive areas within the resected adrenal tissue (versus 23% in those cured), wherein CACNA1D mutations were most prevalent (63% versus 33% in those cured). In adjusted regression models, independent predictors of postoperative residual PA included Black versus White race (odds ratio, 5.10 [95% CI, 1.45-17.86]), AVS lateralization only at baseline (odds ratio, 8.93 [95% CI 3.00-26.32] versus both at baseline and after cosyntropin stimulation), and CT-AVS disagreement (odds ratio, 2.75 [95% CI, 1.20-6.31]). CONCLUSIONS: Multifocal, asymmetrical bilateral PA is relatively common, and it cannot be excluded by robust AVS lateralization. Long-term postoperative monitoring should be routinely pursued, to identify residual PA and afford timely initiation of targeted medical therapy.


Sujet(s)
Hyperaldostéronisme , Humains , Hyperaldostéronisme/diagnostic , Hyperaldostéronisme/génétique , Hyperaldostéronisme/chirurgie , Études rétrospectives , Aldostérone , Tétracosactide , Études de cohortes , Cytochrome P-450 CYP11B2 , Glandes surrénales/chirurgie , Glandes surrénales/vascularisation , Surrénalectomie
11.
Handb Exp Pharmacol ; 283: 59-79, 2024.
Article de Anglais | MEDLINE | ID: mdl-37495852

RÉSUMÉ

The mineralocorticoid aldosterone is produced in the zona glomerulosa of the adrenal cortex. Its synthesis is regulated by the serum concentrations of the peptide hormone angiotensin II and potassium. The primary role of aldosterone is to control blood volume and electrolytes. The autonomous production of aldosterone (primary aldosteronism, PA) is considered the most frequent cause of secondary hypertension. Aldosterone-producing adenomas and (micro-)nodules are frequent causes of PA and often carry somatic mutations in ion channels and transporters. Rare familial forms of PA are due to germline mutations. Both somatic and germline mutations in the chloride channel gene CLCN2, encoding ClC-2, have been identified in PA. Clinical findings and results from cell culture and animal models have advanced our knowledge about the role of anions in PA. The zona glomerulosa of the adrenal gland has now been firmly established as a tissue in which anions play a significant role for signaling. In this overview, we aim to summarize the current knowledge and highlight novel concepts as well as open questions.


Sujet(s)
Hyperaldostéronisme , Zone glomérulée , Animaux , Aldostérone , Hyperaldostéronisme/génétique , Canaux ioniques , Transduction du signal
12.
Hypertension ; 81(2): 361-371, 2024 Feb.
Article de Anglais | MEDLINE | ID: mdl-38095094

RÉSUMÉ

BACKGROUND: Recent advances in omics techniques have allowed detailed genetic characterization of aldosterone-producing adenoma (APA). The pathogenesis of APA is characterized by tumorigenesis-associated aldosterone synthesis. The pathophysiological intricacies of APAs have not yet been elucidated at the level of individual cells. Therefore, a single-cell level analysis is speculated to be valuable in studying the differentiation process of APA. METHODS: We conducted single-nucleus RNA sequencing of APAs with KCNJ5 mutation and nonfunctional adenomas obtained from 3 and 2 patients, respectively. RESULTS: The single-nucleus RNA sequencing revealed the intratumoral heterogeneity of APA and identified cell populations consisting of a shared cluster of nonfunctional adenoma and APA. In addition, we extracted 2 cell fates in APA and obtained a cell population specialized in aldosterone synthesis. Genes related to ribosomes and neurodegenerative diseases were upregulated in 1 of these fates, whereas those related to the regulation of glycolysis were upregulated in the other fate. Furthermore, the total RNA reads in the nucleus were higher in hormonally activated clusters, indicating a marked activation of transcription per cell. CONCLUSIONS: The single-nucleus RNA sequencing revealed intratumoral heterogeneity of APA with KCNJ5 mutation. The observation of 2 cell fates in KCNJ5-mutated APAs provides the postulation that a heterogeneous process of cellular differentiation was implicated in the pathophysiological mechanisms underlying APA tumors.


Sujet(s)
Adénomes , Tumeurs corticosurrénaliennes , Adénome corticosurrénalien , Hyperaldostéronisme , Humains , Aldostérone , Adénome corticosurrénalien/génétique , Adénome corticosurrénalien/anatomopathologie , Adénomes/génétique , Adénomes/anatomopathologie , Canaux potassiques rectifiants entrants couplés aux protéines G/génétique , Mutation , Tumeurs corticosurrénaliennes/génétique , Hyperaldostéronisme/génétique
13.
Endocr J ; 71(3): 245-252, 2024 Mar 28.
Article de Anglais | MEDLINE | ID: mdl-38143086

RÉSUMÉ

11Beta-hydroxysteroid dehydrogenase 1 (11ß-HSD1) is a key enzyme involved in metabolic syndrome. Transcript-specific epigenetic regulation of the gene encoding 11ß-HSD1 (HSD11B1) has been reported. We examined the mRNA level and methylation status of the HSD11B1 promoter region in the adipose tissue of patients with primary aldosteronism (PA). We compared 10 tissue specimens from patients with PA caused by aldosterone-producing adenoma (APA) with 8 adipose tissue specimens from patients with subclinical Cushing's syndrome (SCS) caused by cortisol-producing adenomas, 4 tissue specimens from patients with Cushing's adenoma (Cu), or 7 tissue specimens from patients with non-functioning adrenal adenoma (NFA). PA, SCS, and Cu were diagnosed according to the guideline of the Japan Endocrine Society. The mRNA level of HSD11B1 was quantified using real-time PCR. Isolated DNA was treated with bisulfite and amplified using primers specific to the human HSD11B1 promoter region. The glycohemoglobin level was significantly higher in patients with APA, SCS, or Cu than in those with NFA (p < 0.05). Blood pressure was significantly higher in patients with APA than in those with SCS, Cu, or NFA (p < 0.01). The HSD11B1 mRNA level was significantly increased in the adipose tissues of APA or SCS patients compared with Cu or NFA patients (p < 0.05). The methylation ratio was significantly lower in SCS patients than in APA, Cu, or NFA patients (p < 0.05). HSD11B1 expression is partly controlled by an epigenetic mechanism in human tissues. The pathophysiological role of epigenetic regulation of HSD11B1 expression in adipose tissue requires further study.


Sujet(s)
Adénomes , Adénome corticosurrénalien , Hyperaldostéronisme , Humains , 11-beta-Hydroxysteroid dehydrogenase type 1/génétique , 11-beta-Hydroxysteroid dehydrogenase type 1/métabolisme , Épigenèse génétique , Tissu adipeux/métabolisme , Adénome corticosurrénalien/métabolisme , Hyperaldostéronisme/génétique , Hyperaldostéronisme/métabolisme , Adénomes/métabolisme , ARN messager/métabolisme
14.
High Blood Press Cardiovasc Prev ; 31(1): 15-21, 2024 Jan.
Article de Anglais | MEDLINE | ID: mdl-38123759

RÉSUMÉ

 INTRODUCTION: This study will test the hypothesis that primary aldosteronism (PA) involves alterations in Na+, K+, and water content in the skin that are corrected by adrenalectomy. AIM AND METHODS: In skin biopsies, we will measure the content of Na+, K+, water, by physical-chemical methods and the osmotic-stress-responsive transcription factor Tonicity-responsive Enhancer Binding Protein (TonEBP, NFAT5) mRNA copy number by droplet digital PCR, in sex-balanced cohorts of 18 -75-year-old consecutive consenting patients with unilateral and bilateral PA, primary (essential) hypertension, and normotension. Before surgery, the patients with unilateral PA will receive the mineralocorticoid receptor antagonist (MRA) canrenone at doses that correct hypokalemia and high blood pressure values. They will be reassessed in an identical way one month after surgical cure, while off MRA. PA patients not selected for adrenalectomy will similarly be assessed at diagnosis and follow-up while on stable MRA treatment. Since a pilot study showed a direct correlation of dry weight (DW) with skin electrolytes and water content and significant differences of biopsy DW between surgery and follow-up, meaningful comparison of the skin cations and water content and TonEBP mRNA copy number, between specimen obtained at different time points, will require DW- and total mRNA-adjustment, respectively. CONCLUSION: This study will provide novel information on the skin Na+, K+ and water content in PA, the paradigm of salt-dependent hypertension, and novel knowledge on the effect of surgical cure of hyperaldosteronism. The TonEBP-mediated regulation of Na+, K+ and water content in the skin will also be unveiled. TRAIL REGISTRY: Trial Registration number: NCT06090617. Date of Registration: 2023-10-19.


Sujet(s)
Hyperaldostéronisme , Hypertension artérielle , Humains , Adolescent , Jeune adulte , Adulte , Adulte d'âge moyen , Sujet âgé , Projets pilotes , Hyperaldostéronisme/diagnostic , Hyperaldostéronisme/génétique , Hyperaldostéronisme/chirurgie , Hypertension artérielle/diagnostic , Hypertension artérielle/traitement médicamenteux , Hypertension artérielle/génétique , Chlorure de sodium alimentaire , Électrolytes/usage thérapeutique , ARN messager/usage thérapeutique
15.
J Formos Med Assoc ; 123 Suppl 2: S135-S140, 2024 Mar.
Article de Anglais | MEDLINE | ID: mdl-38097431

RÉSUMÉ

Unilateral primary aldosteronism is thought to be a surgically curable disease, and unilateral adrenalectomy is the mainstay treatment. The Primary Aldosteronism Surgical Outcome (PASO) consensus was developed to assess clinical and biochemical outcomes to standardize the classification of surgical outcomes. However, fewer than half of patients are cured of hypertension after adrenalectomy; therefore, preoperative patient counseling and evaluation might be necessary. Moreover, current studies show that genetic mutations and histopathology classification are associated with the treatment outcome. The Task Force of Taiwan PA recommends using a specific scoring system, including the PASO score and nomogram-based preoperative score, to predict the clinical outcome before adrenalectomy. Herein, we discuss the associations of current histopathological classification and specific somatic gene mutations with clinical outcomes after surgery.


Sujet(s)
Hyperaldostéronisme , Hypertension artérielle , Humains , Études rétrospectives , Hyperaldostéronisme/génétique , Hyperaldostéronisme/chirurgie , Résultat thérapeutique , Surrénalectomie , Hypertension artérielle/complications
16.
J Steroid Biochem Mol Biol ; 237: 106445, 2024 03.
Article de Anglais | MEDLINE | ID: mdl-38104729

RÉSUMÉ

Primary aldosteronism (PA) causes 5-10% of hypertension cases, but only a minority of patients are currently diagnosed and treated because of a complex, stepwise, and partly invasive workup. We tested the performance of urine steroid metabolomics, the computational analysis of 24-hour urine steroid metabolome data by machine learning, for the identification and subtyping of PA. Mass spectrometry-based multi-steroid profiling was used to quantify the excretion of 34 steroid metabolites in 24-hour urine samples from 158 adults with PA (88 with unilateral PA [UPA] due to aldosterone-producing adenomas [APAs]; 70 with bilateral PA [BPA]) and 65 sex- and age-matched healthy controls. All APAs were resected and underwent targeted gene sequencing to detect somatic mutations associated with UPA. Patients with PA had increased urinary metabolite excretion of mineralocorticoids, glucocorticoids, and glucocorticoid precursors. Urine steroid metabolomics identified patients with PA with high accuracy, both when applied to all 34 or only the three most discriminative steroid metabolites (average areas under the receiver-operating characteristics curve [AUCs-ROC] 0.95-0.97). Whilst machine learning was suboptimal in differentiating UPA from BPA (average AUCs-ROC 0.65-0.73), it readily identified APA cases harbouring somatic KCNJ5 mutations (average AUCs-ROC 0.79-85). These patients showed a distinctly increased urine excretion of the hybrid steroid 18-hydroxycortisol and its metabolite 18-oxo-tetrahydrocortisol, the latter identified by machine learning as by far the most discriminative steroid. In conclusion, urine steroid metabolomics is a non-invasive candidate test for the accurate identification of PA cases and KCNJ5-mutated APAs.


Sujet(s)
Adénomes , Tumeurs corticosurrénaliennes , Adénome corticosurrénalien , Hyperaldostéronisme , Adulte , Humains , Hyperaldostéronisme/diagnostic , Hyperaldostéronisme/génétique , Hyperaldostéronisme/métabolisme , Adénome corticosurrénalien/génétique , Adénomes/diagnostic , Stéroïdes , Spectrométrie de masse , Aldostérone/métabolisme , Mutation , Canaux potassiques rectifiants entrants couplés aux protéines G/génétique , Canaux potassiques rectifiants entrants couplés aux protéines G/métabolisme , Tumeurs corticosurrénaliennes/génétique
17.
Cardiovasc Diabetol ; 22(1): 352, 2023 12 20.
Article de Anglais | MEDLINE | ID: mdl-38124109

RÉSUMÉ

BACKGROUND AND AIMS: Primary aldosteronism (PA) is an adrenal disorder of autonomous aldosterone secretion which promotes arterial injury. We aimed to explore whether PA is causally associated with lower-extremity arterial disease (LEAD). METHODS: We included 39,713 patients with diabetes and 419,312 participants without diabetes from UK Biobank. We derived a polygenic risk score (PRS) for PA based on previous genome-wide association studies (GWAS). Outcomes included LEAD and LEAD related gangrene or amputation. We conducted a two-sample Mendelian randomization analysis for PA and outcomes to explore their potential causal relationship. RESULTS: In whole population, individuals with a higher PA PRS had an increased risk of LEAD. Among patients with diabetes, compared to the subjects in the first tertile of PA PRS, subjects in the third tertile showed a 1.24-fold higher risk of LEAD (OR 1.24, 95% CI 1.03-1.49) and a 2.09-fold higher risk of gangrene (OR 2.09, 95% CI 1.27-3.44), and 1.72-fold higher risk of amputation (OR 1.72, 95% CI 1.10-2.67). Among subjects without diabetes, there was no significant association between PA PRS and LEAD, gangrene or amputation. Two-sample Mendelian randomization analysis indicated that genetically predictors of PA was significantly associated with higher risks of LEAD and gangrene (inverse variance weighted OR 1.20 [95% CI 1.08-1.34]) for LEAD, 1.48 [95% CI 1.28-1.70] for gangrene), with no evidence of significant heterogeneity or directional pleiotropy. CONCLUSIONS: Primary aldosteronism is genetically and causally associated with higher risks of LEAD and gangrene, especially among patients with diabetes. Targeting on the autonomous aldosterone secretion may prevent LEAD progression.


Sujet(s)
Diabète , Hyperaldostéronisme , Maladies vasculaires , Humains , Gangrène , Aldostérone , Étude d'association pangénomique , Analyse de randomisation mendélienne , , Membre inférieur , Hyperaldostéronisme/diagnostic , Hyperaldostéronisme/épidémiologie , Hyperaldostéronisme/génétique , Polymorphisme de nucléotide simple
19.
Eur J Endocrinol ; 189(5): R11-R22, 2023 Nov 08.
Article de Anglais | MEDLINE | ID: mdl-37847213

RÉSUMÉ

Congenital forms of endocrine hypertension are rare and potentially life-threatening disorders, primarily caused by genetic defects affecting adrenal steroid synthesis and activation pathways. These conditions exhibit diverse clinical manifestations, which can be distinguished by their unique molecular mechanisms and steroid profiles. Timely diagnosis and customized management approach are crucial to mitigate unfavorable outcomes associated with uncontrolled hypertension and other related conditions. Treatment options for these disorders depend on the distinct underlying pathophysiology, which involves specific pharmacological therapies or surgical adrenalectomy in some instances. This review article summarizes the current state of knowledge on the therapeutic management of congenital forms of endocrine hypertension, focusing on familial hyperaldosteronism (FH), congenital adrenal hyperplasia, apparent mineralocorticoid excess, and Liddle syndrome. We provide an overview of the genetic and molecular pathogenesis underlying each disorder, describe the clinical features, and discuss the various therapeutic approaches available and their risk of adverse effects, aiming to improve outcomes in patients with these rare and complex conditions.


Sujet(s)
Hyperplasie congénitale des surrénales , Hyperaldostéronisme , Hypertension artérielle , Syndrome d'excès apparent en minéralocorticoïdes , Humains , Hypertension artérielle/génétique , Hypertension artérielle/thérapie , Hyperaldostéronisme/diagnostic , Hyperaldostéronisme/génétique , Hyperaldostéronisme/thérapie , Syndrome d'excès apparent en minéralocorticoïdes/diagnostic , Syndrome d'excès apparent en minéralocorticoïdes/génétique , Hyperplasie congénitale des surrénales/diagnostic , Hyperplasie congénitale des surrénales/génétique , Hyperplasie congénitale des surrénales/thérapie , Stéroïdes , Aldostérone
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE
...