1
|
Charoensri S, Auchus RJ. Therapeutic management of congenital forms of endocrine hypertension. Eur J Endocrinol 2023; 189:R11-R22. [PMID: 37847213 DOI: 10.1093/ejendo/lvad140] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/02/2023] [Revised: 08/15/2023] [Accepted: 09/19/2023] [Indexed: 10/18/2023]
Abstract
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.
Collapse
Affiliation(s)
- Suranut Charoensri
- Division of Endocrinology and Metabolism, Department of Medicine, Faculty of Medicine, Khon Kaen University, Khon Kaen 40002, Thailand
- Division of Metabolism, Endocrinology, and Diabetes, Department of Internal Medicine, University of Michigan, Ann Arbor, MI 48109, United States
| | - Richard J Auchus
- Division of Metabolism, Endocrinology, and Diabetes, Department of Internal Medicine, University of Michigan, Ann Arbor, MI 48109, United States
- Department of Pharmacology, University of Michigan, Ann Arbor, MI 48109, United States
- Endocrinology & Metabolism Section, Medicine Service, LTC Charles S. Kettles VA Medical Center, Ann Arbor, MI 48104, United States
| |
Collapse
|
2
|
Ectopic localization of CYP11B1 and CYP11B2-expressing cells in the normal human adrenal gland. PLoS One 2022; 17:e0279682. [PMID: 36584094 PMCID: PMC9803228 DOI: 10.1371/journal.pone.0279682] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2022] [Accepted: 12/12/2022] [Indexed: 12/31/2022] Open
Abstract
The sharp line of demarcation between zona glomerulosa (ZG) and zona fasciculata (ZF) has been recently challenged suggesting that this interface is no longer a compartment boundary. We have used immunohistochemical analyses to study the steroid 11β-hydroxylase (CYP11B1) and aldosterone synthase (CYP11B2) pattern of expression and investigate the remodeling of the adrenal cortex in relation to aging. We analyzed human adrenal glands prepared from 47 kidney donors. No aldosterone-producing micronodules (APMs) were detectable in the younger donors aged between 22-39 but the functional ZG depicted by positive CYP11B2 staining demonstrated a lack of continuity. In contrast, the development of APMs was found in samples from individuals aged 40-70. Importantly, the progressive replacement of CYP11B2-expressing cells in the histological ZG by CYP11B1-expressing cells highlights the remodeling capacity of the adrenal cortex. In 70% of our samples, immunofluorescence studies revealed the presence of isolated or clusters of CYP11B2 positive cells in the ZF and zona reticularis. Our data emphasize that mineralocorticoid- and glucocorticoid-producing cells are distributed throughout the cortex and the medulla making the determination of the functional status of a cell or group of cells a unique tool in deciphering the changes occurring in adrenal gland particularly during aging. They also suggest that, in humans, steroidogenic cell phenotype defined by function is a stable feature and thus, the functional zonation might be not solely maintained by cell lineage conversion/migration.
Collapse
|
3
|
Lenzini L, Caroccia B, Seccia TM, Rossi GP. Peptidergic G Protein-Coupled Receptor Regulation of Adrenal Function: Bench to Bedside and Back. Endocr Rev 2022; 43:1038-1050. [PMID: 35436330 DOI: 10.1210/endrev/bnac011] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/21/2021] [Indexed: 11/19/2022]
Abstract
An altered secretion of adrenocortical and adrenomedullary hormones plays a role in the clinical syndromes of primary aldosteronism (PA), Cushing, and pheochromocytoma. Moreover, an altered production of adrenocortical hormones and/or an abnormal release of factors by the adrenal medulla are involved in several other diseases, including high blood pressure, congestive heart failure, liver cirrhosis, nephrotic syndrome, primary reninism, renovascular hypertension, Addison disease, Bartter, Gitelman, and virilization syndromes. Understanding the regulation of adrenal function and the interactions between adrenal cortex and medulla is, therefore, the prerequisite for mechanistic understanding of these disorders. Accumulating evidence indicates that the modulation of adrenal hormone biosynthesis is a process far more complex than originally thought, as it involves several factors, each cooperating with the other. Moreover, the tight vascular and neural interconnections between the adrenal cortex and medulla underlie physiologically relevant autocrine/paracrine interactions involving several peptides. Besides playing a pathophysiological role in common adrenal diseases, these complex mechanisms could intervene also in rare diseases, such as pheochromocytoma concomitant with adrenal Cushing or with PA, and PA co-occurring with Cushing, through mechanisms that remain to be fully understood at the molecular levels. Heterodimerization of G protein-coupled receptors (GPCRs) induced by peptide signaling is a further emerging new modulatory mechanism capable of finely tuning adrenal hormones synthesis and release. In this review we will examine current knowledge on the role of peptides that act via GPCRs in the regulation of adrenal hormone secretion with a particular focus on autocrine-paracrine signals.
Collapse
Affiliation(s)
- Livia Lenzini
- Emergency Medicine Unit, Center for blood pressure disorders -Regione Veneto and Specialized Center of Excellence for Hypertension of the European Society of Hypertension, Department of Medicine-DIMED, University of Padua, 35126 Padua, Italy
| | - Brasilina Caroccia
- Emergency Medicine Unit, Center for blood pressure disorders -Regione Veneto and Specialized Center of Excellence for Hypertension of the European Society of Hypertension, Department of Medicine-DIMED, University of Padua, 35126 Padua, Italy
| | - Teresa Maria Seccia
- Emergency Medicine Unit, Center for blood pressure disorders -Regione Veneto and Specialized Center of Excellence for Hypertension of the European Society of Hypertension, Department of Medicine-DIMED, University of Padua, 35126 Padua, Italy
| | - Gian Paolo Rossi
- Emergency Medicine Unit, Center for blood pressure disorders -Regione Veneto and Specialized Center of Excellence for Hypertension of the European Society of Hypertension, Department of Medicine-DIMED, University of Padua, 35126 Padua, Italy
| |
Collapse
|
4
|
Spaulding SC, Bollag WB. The role of lipid second messengers in aldosterone synthesis and secretion. J Lipid Res 2022; 63:100191. [PMID: 35278411 PMCID: PMC9020094 DOI: 10.1016/j.jlr.2022.100191] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2022] [Revised: 02/24/2022] [Accepted: 02/25/2022] [Indexed: 11/23/2022] Open
Abstract
Second messengers are small rapidly diffusing molecules or ions that relay signals between receptors and effector proteins to produce a physiological effect. Lipid messengers constitute one of the four major classes of second messengers. The hydrolysis of two main classes of lipids, glycerophospholipids and sphingolipids, generate parallel profiles of lipid second messengers: phosphatidic acid (PA), diacylglycerol (DAG), and lysophosphatidic acid versus ceramide, ceramide-1-phosphate, sphingosine, and sphingosine-1-phosphate, respectively. In this review, we examine the mechanisms by which these lipid second messengers modulate aldosterone production at multiple levels. Aldosterone is a mineralocorticoid hormone responsible for maintaining fluid volume, electrolyte balance, and blood pressure homeostasis. Primary aldosteronism is a frequent endocrine cause of secondary hypertension. A thorough understanding of the signaling events regulating aldosterone biosynthesis may lead to the identification of novel therapeutic targets. The cumulative evidence in this literature emphasizes the critical roles of PA, DAG, and sphingolipid metabolites in aldosterone synthesis and secretion. However, it also highlights the gaps in our knowledge, such as the preference for phospholipase D-generated PA or DAG, as well as the need for further investigation to elucidate the precise mechanisms by which these lipid second messengers regulate optimal aldosterone production.
Collapse
Affiliation(s)
- Shinjini C Spaulding
- Department of Physiology, Medical College of Georgia at Augusta University, Augusta, GA, USA
| | - Wendy B Bollag
- Department of Physiology, Medical College of Georgia at Augusta University, Augusta, GA, USA; Research Department, Charlie Norwood VA Medical Center, Augusta, GA, USA.
| |
Collapse
|
5
|
Williams TA, Reincke M. Pathophysiology and histopathology of primary aldosteronism. Trends Endocrinol Metab 2022; 33:36-49. [PMID: 34743804 DOI: 10.1016/j.tem.2021.10.002] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/22/2021] [Revised: 09/30/2021] [Accepted: 10/09/2021] [Indexed: 10/19/2022]
Abstract
Primary aldosteronism (PA) can be sporadic or familial and classified into unilateral and bilateral forms. Sporadic PA predominates with excessive aldosterone production usually arising from a unilateral aldosterone-producing adenoma (APA) or bilateral adrenocortical hyperplasia. Familial PA is rare and caused by germline variants, that partly correspond to somatic alterations in APAs. Classification into unilateral and bilateral PA determines the treatment approach but does not accurately mirror disease pathology. Some evidence indicates a disease continuum ranging from balanced aldosterone production from each adrenal to extreme asymmetrical bilateral aldosterone production. Nonetheless, surgical removal of the overactive adrenal in unilateral PA achieves highly successful outcomes and almost all patients are biochemically cured of their aldosteronism.
Collapse
Affiliation(s)
- Tracy Ann Williams
- Medizinische Klinik und Poliklinik IV, Klinikum der Universität München, LMU München, München, Germany; Division of Internal Medicine and Hypertension, Department of Medical Sciences, University of Turin, Turin, Italy.
| | - Martin Reincke
- Medizinische Klinik und Poliklinik IV, Klinikum der Universität München, LMU München, München, Germany
| |
Collapse
|
6
|
Tsilosani A, Gao C, Zhang W. Aldosterone-Regulated Sodium Transport and Blood Pressure. Front Physiol 2022; 13:770375. [PMID: 35197862 PMCID: PMC8859437 DOI: 10.3389/fphys.2022.770375] [Citation(s) in RCA: 32] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2021] [Accepted: 01/06/2022] [Indexed: 11/13/2022] Open
Abstract
Aldosterone is a major mineralocorticoid steroid hormone secreted by glomerulosa cells in the adrenal cortex. It regulates a variety of physiological responses including those to oxidative stress, inflammation, fluid disruption, and abnormal blood pressure through its actions on various tissues including the kidney, heart, and the central nervous system. Aldosterone synthesis is primarily regulated by angiotensin II, K+ concentration, and adrenocorticotrophic hormone. Elevated serum aldosterone levels increase blood pressure largely by increasing Na+ re-absorption in the kidney through regulating transcription and activity of the epithelial sodium channel (ENaC). This review focuses on the signaling pathways involved in aldosterone synthesis and its effects on Na+ reabsorption through ENaC.
Collapse
Affiliation(s)
- Akaki Tsilosani
- Department of Regenerative & Cancer Cell Biology, Albany Medical College, Albany, NY, United States
| | - Chao Gao
- Department of Regenerative & Cancer Cell Biology, Albany Medical College, Albany, NY, United States
| | - Wenzheng Zhang
- Department of Regenerative & Cancer Cell Biology, Albany Medical College, Albany, NY, United States
| |
Collapse
|
7
|
ATP1A1 Mutant in Aldosterone-Producing Adenoma Leads to Cell Proliferation. Int J Mol Sci 2021; 22:ijms222010981. [PMID: 34681640 PMCID: PMC8537586 DOI: 10.3390/ijms222010981] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2021] [Revised: 10/04/2021] [Accepted: 10/09/2021] [Indexed: 12/14/2022] Open
Abstract
The molecular mechanisms by which ATP1A1 mutation-mediated cell proliferation or tumorigenesis in aldosterone-producing adenomas (APAs) have not been elucidated. First, we investigated whether the APA-associated ATP1A1 L104R mutation stimulated cell proliferation. Second, we aimed to clarify the molecular mechanisms by which the ATP1A1 mutation-mediated cell proliferated. We performed transcriptome analysis in APAs with ATP1A1 mutation. ATP1A1 L104R mutation were modulated in human adrenocortical carcinoma (HAC15) cells (ATP1A1-mutant cells), and we evaluated cell proliferation and molecular signaling events. Transcriptome and immunohistochemical analysis showed that Na/K-ATPase (NKA) expressions in ATP1A1 mutated APA were more abundant than those in non-functioning adrenocortical adenoma or KCNJ5 mutated APAs. The significant increase of number of cells, amount of DNA and S-phase population were shown in ATP1A1-mutant cells. Fluo-4 in ATP1A1-mutant cells were significantly increased. Low concentration of ouabain stimulated cell proliferation in ATP1A1-mutant cells. ATP1A1-mutant cells induced Src phosphorylation, and low concentration of ouabain supplementation showed further Src phosphorylation. We demonstrated that NKAs were highly expressed in ATP1A1 mutant APA, and the mutant stimulated cell proliferation and Src phosphorylation in ATP1A1-mutant cells. NKA stimulations would be a risk factor for the progression and development to an ATP1A1 mutant APA.
Collapse
|
8
|
Rossi GP, Lenzini L, Caroccia B, Rossitto G, Seccia TM. Angiotensin peptides in the regulation of adrenal cortical function. EXPLORATION OF MEDICINE 2021. [DOI: 10.37349/emed.2021.00047] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022] Open
Abstract
The adrenal cortex plays a key role in the regulation of metabolism, salt and water homeostasis and sex differentiation by synthesizing glucocorticoid, mineralocorticoid and androgen hormones. Evidence exists that angiotensin II regulates adrenocortical function and it has been contended that angiotensin peptides of the non-canonical branch of the renin angiotensin system (RAS) might also modulate steroidogenesis in adrenals. Thus, the aim of this review is to examine the role of the RAS, and particularly of the angiotensin peptides and their receptors, in the regulation of adrenocortical hormones with particular focus on aldosterone production.
Collapse
Affiliation(s)
- Gian Paolo Rossi
- Emergency Medicine Unit and European Society of Hypertension Specialized Center of Excellence for Hypertension, Department of Medicine-DIMED, University of Padua, Italy
| | - Livia Lenzini
- Emergency Medicine Unit and European Society of Hypertension Specialized Center of Excellence for Hypertension, Department of Medicine-DIMED, University of Padua, Italy
| | - Brasilina Caroccia
- Emergency Medicine Unit and European Society of Hypertension Specialized Center of Excellence for Hypertension, Department of Medicine-DIMED, University of Padua, Italy
| | - Giacomo Rossitto
- Emergency Medicine Unit and European Society of Hypertension Specialized Center of Excellence for Hypertension, Department of Medicine-DIMED, University of Padua, Italy
| | - Teresa Maria Seccia
- Emergency Medicine Unit and European Society of Hypertension Specialized Center of Excellence for Hypertension, Department of Medicine-DIMED, University of Padua, Italy
| |
Collapse
|
9
|
Lim JS, Plaska SW, Rege J, Rainey WE, Turcu AF. Aldosterone-Regulating Receptors and Aldosterone-Driver Somatic Mutations. Front Endocrinol (Lausanne) 2021; 12:644382. [PMID: 33796077 PMCID: PMC8008747 DOI: 10.3389/fendo.2021.644382] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/21/2020] [Accepted: 02/08/2021] [Indexed: 11/13/2022] Open
Abstract
BACKGROUND Somatic gene mutations that facilitate inappropriate intracellular calcium entrance have been identified in most aldosterone-producing adenomas (APAs). Studies suggest that angiotensin II and adrenocorticotropic hormone (ACTH) augment aldosterone production from APAs. Little is known, however, regarding possible variations in response to hormonal stimuli between APAs with different aldosterone-driver mutations. OBJECTIVE To analyze the transcript expression of type 1 angiotensin II receptors (AGTR1), ACTH receptors (MC2R), and melanocortin 2 receptor accessory protein (MRAP) in APAs with known aldosterone-driver somatic mutations. METHODS RNA was isolated from APAs with mutations in: KCNJ5 (n = 14), ATP1A1 (n = 14), CACNA1D (n = 14), and ATP2B3 (n = 5), and from normal adjacent adrenal tissue (n = 45). Transcript expression of MC2R, MRAP, AGTR1, aldosterone synthase (CYP11B2), 17α-hydroxylase/17,20-lyase (CYP17A1), and 11β-hydroxylase (CYP11B1) were quantified using quantitative RT-PCR and normalized to β-actin. RESULTS Compared to adjacent normal adrenal tissue, APAs had higher transcript levels of CYP11B2 (2,216.4 [1,112.0, 2,813.5]-fold, p < 0.001), MC2R (2.88 [2.00, 4.52]-fold, p < 0.001), and AGTR1 (1.80 [1.02, 2.80]-fold, p < 0.001]), and lower transcript levels of MRAP, CYP17A1, and CYP11B1 (0.28-0.36, p < 0.001 for all). MC2R and CYP11B2 transcripts were lower in APAs with KCNJ5 vs. other mutations (p < 0.01 for both). MC2R expression correlated positively with that of AGTR1 in APAs harboring KCNJ5 and CACNA1D mutations, and with MRAP expression in APAs harboring ATPase mutations. CONCLUSIONS While MC2R and AGTR1 are expressed in all APAs, differences were observed based on the underlying aldosterone-driver somatic mutations. In tandem, our findings suggest that APAs with ATPase-mutations are more responsive to ACTH than KCNJ5-mutated APAs.
Collapse
Affiliation(s)
- Jung Soo Lim
- Department of Molecular and Integrative Physiology, University of Michigan, Ann Arbor, MI, United States
- Division of Endocrinology and Metabolism, Department of Internal Medicine, Yonsei University Wonju College of Medicine, Wonju Severance Christian Hospital, Wonju, South Korea
| | - Samuel W. Plaska
- Department of Molecular and Integrative Physiology, University of Michigan, Ann Arbor, MI, United States
| | - Juilee Rege
- Department of Molecular and Integrative Physiology, University of Michigan, Ann Arbor, MI, United States
| | - William E. Rainey
- Department of Molecular and Integrative Physiology, University of Michigan, Ann Arbor, MI, United States
- Division of Metabolism, Endocrine, and Diabetes, University of Michigan, Ann Arbor, MI, United States
| | - Adina F. Turcu
- Division of Metabolism, Endocrine, and Diabetes, University of Michigan, Ann Arbor, MI, United States
- *Correspondence: Adina F. Turcu,
| |
Collapse
|
10
|
Vohra T, Kemter E, Sun N, Dobenecker B, Hinrichs A, Burrello J, Gomez-Sanchez EP, Gomez-Sanchez CE, Wang J, Kinker IS, Teupser D, Fischer K, Schnieke A, Peitzsch M, Eisenhofer G, Walch A, Reincke M, Wolf E, Williams TA. Effect of Dietary Sodium Modulation on Pig Adrenal Steroidogenesis and Transcriptome Profiles. Hypertension 2020; 76:1769-1777. [PMID: 33070662 DOI: 10.1161/hypertensionaha.120.15998] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Primary aldosteronism is a frequent form of endocrine hypertension caused by aldosterone overproduction from the adrenal cortex. Regulation of aldosterone biosynthesis has been studied in rodents despite differences in adrenal physiology with humans. We, therefore, investigated pig adrenal steroidogenesis, morphology, and transcriptome profiles of the zona glomerulosa (zG) and zona fasciculata in response to activation of the renin-angiotensin-aldosterone system by dietary sodium restriction. Six-week-old pigs were fed a low- or high-sodium diet for 14 days (3 pigs per group, 0.4 g sodium/kg feed versus 6.8 g sodium/kg). Plasma aldosterone concentrations displayed a 43-fold increase (P=0.011) after 14 days of sodium restriction (day 14 versus day 0). Low dietary sodium caused a 2-fold increase in thickness of the zG (P<0.001) and an almost 3-fold upregulation of CYP11B (P<0.05) compared with high dietary sodium. Strong immunostaining of the KCNJ5 (G protein-activated inward rectifier potassium channel 4), which is frequently mutated in primary aldosteronism, was demonstrated in the zG. mRNA sequencing transcriptome analysis identified significantly altered expression of genes modulated by the renin-angiotensin-aldosterone system in the zG (n=1172) and zona fasciculata (n=280). These genes included many with a known role in the regulation of aldosterone synthesis and adrenal function. The most highly enriched biological pathways in the zG were related to cholesterol biosynthesis, steroid metabolism, cell cycle, and potassium channels. This study provides mechanistic insights into the physiology and pathophysiology of aldosterone production in a species closely related to humans and shows the suitability of pigs as a translational animal model for human adrenal steroidogenesis.
Collapse
Affiliation(s)
- Twinkle Vohra
- From the Medizinische Klinik und Poliklinik IV, Klinikum der Universität München (T.V., I.-S.K., M.R., T.A.W.), Ludwig-Maximilians-Universität München, Munich, Germany
| | - Elisabeth Kemter
- Chair for Molecular Animal Breeding and Biotechnology, Gene Center and Department of Veterinary Sciences (E.K., A.H., E.W.), Ludwig-Maximilians-Universität München, Munich, Germany
| | - Na Sun
- Research Unit Analytical Pathology, German Research Center for Environmental Health, Helmholtz Zentrum München, Neuherberg, Germany (N.S., J.W., A.W.)
| | - Britta Dobenecker
- Chair of Animal Nutrition and Dietetics, Department of Veterinary Sciences, Ludwig-Maximilians-Universität München, Oberschleißheim, Germany (B.D.)
| | - Arne Hinrichs
- Chair for Molecular Animal Breeding and Biotechnology, Gene Center and Department of Veterinary Sciences (E.K., A.H., E.W.), Ludwig-Maximilians-Universität München, Munich, Germany
| | - Jacopo Burrello
- Division of Internal Medicine and Hypertension, Department of Medical Sciences, University of Turin, Italy (J.B., T.A.W.)
| | - Elise P Gomez-Sanchez
- Department of Neurobiology and Anatomical Sciences, University of Mississippi Medical Center, Jackson (E.P.G.-S.)
| | - Celso E Gomez-Sanchez
- Endocrine Division, G.V. (Sonny) Montgomery VA Medical Center, Jackson, MS (C.E.G.-S.).,Department of Pharmacology and Toxicology and Medicine, University of Mississippi Medical Center, Jackson (C.E.G.-S.)
| | - Jun Wang
- Research Unit Analytical Pathology, German Research Center for Environmental Health, Helmholtz Zentrum München, Neuherberg, Germany (N.S., J.W., A.W.)
| | - Isabella-Sabrina Kinker
- From the Medizinische Klinik und Poliklinik IV, Klinikum der Universität München (T.V., I.-S.K., M.R., T.A.W.), Ludwig-Maximilians-Universität München, Munich, Germany
| | - Daniel Teupser
- Institute of Laboratory Medicine, University Hospital (D.T.), Ludwig-Maximilians-Universität München, Munich, Germany
| | - Konrad Fischer
- School of Life Sciences Weihenstephan, Technical University Munich, Freising, Germany (K.F., A.S.)
| | - Angelika Schnieke
- School of Life Sciences Weihenstephan, Technical University Munich, Freising, Germany (K.F., A.S.)
| | - Mirko Peitzsch
- Institute of Clinical Chemistry and Laboratory Medicine (M.P., G.E.), University Hospital Carl Gustav Carus, Technische Universität Dresden, Germany
| | - Graeme Eisenhofer
- Institute of Clinical Chemistry and Laboratory Medicine (M.P., G.E.), University Hospital Carl Gustav Carus, Technische Universität Dresden, Germany.,Department of Medicine III (G.E.), University Hospital Carl Gustav Carus, Technische Universität Dresden, Germany
| | - Axel Walch
- Research Unit Analytical Pathology, German Research Center for Environmental Health, Helmholtz Zentrum München, Neuherberg, Germany (N.S., J.W., A.W.)
| | - Martin Reincke
- From the Medizinische Klinik und Poliklinik IV, Klinikum der Universität München (T.V., I.-S.K., M.R., T.A.W.), Ludwig-Maximilians-Universität München, Munich, Germany
| | - Eckhard Wolf
- Chair for Molecular Animal Breeding and Biotechnology, Gene Center and Department of Veterinary Sciences (E.K., A.H., E.W.), Ludwig-Maximilians-Universität München, Munich, Germany
| | - Tracy Ann Williams
- From the Medizinische Klinik und Poliklinik IV, Klinikum der Universität München (T.V., I.-S.K., M.R., T.A.W.), Ludwig-Maximilians-Universität München, Munich, Germany.,Division of Internal Medicine and Hypertension, Department of Medical Sciences, University of Turin, Italy (J.B., T.A.W.)
| |
Collapse
|
11
|
Zennaro MC, Boulkroun S, Fernandes-Rosa FL. Pathogenesis and treatment of primary aldosteronism. Nat Rev Endocrinol 2020; 16:578-589. [PMID: 32724183 DOI: 10.1038/s41574-020-0382-4] [Citation(s) in RCA: 75] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 06/17/2020] [Indexed: 12/19/2022]
Abstract
Early diagnosis and appropriate treatment of primary aldosteronism, the most frequent cause of secondary hypertension, are crucial to prevent deleterious cardiovascular outcomes. In the past decade, the discovery of genetic abnormalities responsible for sporadic and familial forms of primary aldosteronism has improved the knowledge of the pathogenesis of this disorder. Mutations in genes encoding ion channels and pumps lead to increased cytosolic concentrations of calcium in zona glomerulosa cells, which triggers CYP11B2 expression and autonomous aldosterone production. Improved understanding of the mechanisms underlying the disease is key to improving diagnostics and to developing and implementing targeted treatments. This Review provides an update on the genetic abnormalities associated with sporadic and familial forms of primary aldosteronism, their frequency among different populations and the mechanisms explaining excessive aldosterone production and adrenal nodule development. The possible effects and uses of these findings for improving the diagnostics for primary aldosteronism are discussed. Furthermore, current treatment options of primary aldosteronism are reviewed, with particular attention to the latest studies on blood pressure and cardiovascular outcomes following medical or surgical treatment. The new perspectives regarding the use of targeted drug therapy for aldosterone-producing adenomas with specific somatic mutations are also addressed.
Collapse
Affiliation(s)
- Maria-Christina Zennaro
- INSERM, PARCC, Université de Paris, Paris, France.
- Assistance Publique-Hôpitaux de Paris, Hôpital Européen Georges Pompidou, Service de Génétique, Paris, France.
| | | | | |
Collapse
|
12
|
Fernandes-Rosa FL, Boulkroun S, Zennaro MC. Genetic and Genomic Mechanisms of Primary Aldosteronism. Trends Mol Med 2020; 26:819-832. [PMID: 32563556 DOI: 10.1016/j.molmed.2020.05.005] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2020] [Revised: 05/13/2020] [Accepted: 05/20/2020] [Indexed: 02/07/2023]
Abstract
Aldosterone-producing adenoma (APA) and bilateral adrenal hyperplasia are the main cause of primary aldosteronism (PA), the most frequent form of secondary hypertension. Mutations in ion channels and ATPases have been identified in APA and inherited forms of PA, highlighting the central role of calcium signaling in PA development. Different somatic mutations are also found in aldosterone-producing cell clusters in adrenal glands from healthy individuals and from patients with unilateral and bilateral PA, suggesting additional pathogenic mechanisms. Recent mouse models have also contributed to a better understanding of PA. Application of genetic screening in familial PA, development of surrogate biomarkers for somatic mutations in APA, and use of targeted treatment directed at mutated proteins may allow improved management of patients.
Collapse
Affiliation(s)
| | | | - Maria-Christina Zennaro
- Inserm, PARCC, Université de Paris, F-75015 Paris, France; Assistance Publique-Hôpitaux de Paris, Hôpital Européen Georges Pompidou, Service de Génétique, Paris, France.
| |
Collapse
|
13
|
Sun N, Meyer LS, Feuchtinger A, Kunzke T, Knösel T, Reincke M, Walch A, Williams TA. Mass Spectrometry Imaging Establishes 2 Distinct Metabolic Phenotypes of Aldosterone-Producing Cell Clusters in Primary Aldosteronism. Hypertension 2020; 75:634-644. [PMID: 31957522 DOI: 10.1161/hypertensionaha.119.14041] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Aldosterone-producing adenomas (APAs) are one of the main causes of primary aldosteronism and the most prevalent surgically correctable form of hypertension. Aldosterone-producing cell clusters (APCCs) comprise tight nests of zona glomerulosa cells, strongly positive for CYP11B2 (aldosterone synthase) in immunohistochemistry. APCCs have been suggested as possible precursors of APAs because they frequently carry driver mutations for constitutive aldosterone production, and a few adrenal lesions with histopathologic features of both APCCs and APAs have been identified. Our objective was to investigate the metabolic phenotypes of APCCs (n=27) compared with APAs (n=6) using in situ matrix-assisted laser desorption/ionization mass spectrometry imaging of formalin-fixed paraffin-embedded adrenals from patients with unilateral primary aldosteronism. Specific distribution patterns of metabolites were associated with APCCs and classified 2 separate APCC subgroups (subgroups 1 and 2) indistinguishable by CYP11B2 immunohistochemistry. Metabolic profiles of APCCs in subgroup 1 were tightly clustered and distinct from subgroup 2 and APAs. Multiple APCCs from the same adrenal displayed metabolic profiles of the same subgroup. Metabolites of APCC subgroup 2 were highly similar to the APA group and indicated enhanced metabolic pathways favoring cell proliferation compared with APCC subgroup 1. In conclusion, we demonstrate specific subgroups of APCCs with strikingly divergent distribution patterns of metabolites. One subgroup displays a metabolic phenotype convergent with APAs and may represent the progression of APCCs to APAs.
Collapse
Affiliation(s)
- Na Sun
- From the Research Unit Analytical Pathology, German Research Center for Environmental Health, Helmholtz Zentrum München (N.S., A.F., T. Kunzke, A.W.)
| | - Lucie S Meyer
- Medizinische Klinik und Poliklinik IV, Klinikum der Universität München, LMU München, Germany (L.S.M., M.R., T.A.W.)
| | - Annette Feuchtinger
- From the Research Unit Analytical Pathology, German Research Center for Environmental Health, Helmholtz Zentrum München (N.S., A.F., T. Kunzke, A.W.)
| | - Thomas Kunzke
- From the Research Unit Analytical Pathology, German Research Center for Environmental Health, Helmholtz Zentrum München (N.S., A.F., T. Kunzke, A.W.)
| | - Thomas Knösel
- Institute of Pathology, Ludwig-Maximilians-Universität München, Germany (T. Knösel)
| | - Martin Reincke
- Medizinische Klinik und Poliklinik IV, Klinikum der Universität München, LMU München, Germany (L.S.M., M.R., T.A.W.)
| | - Axel Walch
- From the Research Unit Analytical Pathology, German Research Center for Environmental Health, Helmholtz Zentrum München (N.S., A.F., T. Kunzke, A.W.)
| | - Tracy Ann Williams
- Medizinische Klinik und Poliklinik IV, Klinikum der Universität München, LMU München, Germany (L.S.M., M.R., T.A.W.).,Division of Internal Medicine and Hypertension, Department of Medical Sciences, University of Turin, Italy (T.A.W.)
| |
Collapse
|
14
|
Kozek KA, Du Y, Sharma S, Prael FJ, Spitznagel BD, Kharade SV, Denton JS, Hopkins CR, Weaver CD. Discovery and Characterization of VU0529331, a Synthetic Small-Molecule Activator of Homomeric G Protein-Gated, Inwardly Rectifying, Potassium (GIRK) Channels. ACS Chem Neurosci 2019; 10:358-370. [PMID: 30136838 PMCID: PMC6528656 DOI: 10.1021/acschemneuro.8b00287] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
G protein-gated, inwardly rectifying, potassium (GIRK) channels are important regulators of cellular excitability throughout the body. GIRK channels are heterotetrameric and homotetrameric combinations of the Kir3.1-4 (GIRK1-4) subunits. Different subunit combinations are expressed throughout the central nervous system (CNS) and the periphery, and most of these combinations contain a GIRK1 subunit. For example, the predominance of GIRK channels in the CNS are composed of GIRK1 and GIRK2 subunits, while the GIRK channels in cardiac atrial myocytes are made up mostly of GIRK1 and GIRK4 subunits. Although the vast majority of GIRK channels contain a GIRK1 subunit, discrete populations of cells that express non-GIRK1-containing GIRK (non-GIRK1/X) channels do exist. For instance, dopaminergic neurons in the ventral tegmental area of the brain, associated with addiction and reward, do not express the GIRK1 subunit. Targeting these non-GIRK1/X channels with subunit-selective pharmacological probes could lead to important insights into how GIRK channels are involved in reward and addiction. Such insights may, in turn, reveal therapeutic opportunities for the treatment or prevention of addiction. Previously, our laboratory discovered small molecules that can specifically modulate the activity of GIRK1-containing GIRK channels. However, efforts to generate compounds active on non-GIRK1/X channels from these scaffolds have been unsuccessful. Recently, ivermectin was shown to modulate non-GIRK1/X channels, and historically, ivermectin is known to modulate a wide variety of neuronal channels and receptors. Further, ivermectin is a complex natural product, which makes it a challenging starting point for development of more selective, effective, and potent compounds. Thus, while ivermectin provides proof-of-concept as a non-GIRK1/X channel activator, it is of limited utility. Therefore, we sought to discover a synthetic small molecule that would serve as a starting point for the development of non-GIRK1/X channel modulators. To accomplish this, we used a high-throughput thallium flux assay to screen a 100 000-compound library in search of activators of homomeric GIRK2 channels. Using this approach, we discovered VU0529331, the first synthetic small molecule reported to activate non-GIRK1/X channels, to our knowledge. This discovery represents the first step toward developing potent and selective non-GIRK1/X channel probes. Such molecules will help elucidate the role of GIRK channels in addiction, potentially establishing a foundation for future development of therapies utilizing targeted GIRK channel modulation.
Collapse
Affiliation(s)
- Krystian A. Kozek
- Department of Pharmacology, Vanderbilt University, Nashville, Tennessee, USA
- vanderbilt Institute of Chemical Biology, Vanderbilt University, Nashville, Tennessee, USA
- vanderbilt Medical Scientist Training Program, Vanderbilt University, Nashville, Tennessee, USA
| | - Yu Du
- Department of Pharmacology, Vanderbilt University, Nashville, Tennessee, USA
- vanderbilt Institute of Chemical Biology, Vanderbilt University, Nashville, Tennessee, USA
| | - Swagat Sharma
- Department of Pharmaceutical Sciences, Center for Drug Discovery, College of Pharmacy, University of Nebraska Medical Center, Omaha, Nebraska, USA
| | - Francis J. Prael
- Department of Pharmacology, Vanderbilt University, Nashville, Tennessee, USA
- vanderbilt Institute of Chemical Biology, Vanderbilt University, Nashville, Tennessee, USA
| | - Brittany D. Spitznagel
- Department of Pharmacology, Vanderbilt University, Nashville, Tennessee, USA
- vanderbilt Institute of Chemical Biology, Vanderbilt University, Nashville, Tennessee, USA
| | - Sujay V. Kharade
- Department of Anesthesiology, Vanderbilt University, Nashville, Tennessee, USA
| | - Jerod S. Denton
- Department of Pharmacology, Vanderbilt University, Nashville, Tennessee, USA
- Department of Anesthesiology, Vanderbilt University, Nashville, Tennessee, USA
| | - Corey R. Hopkins
- Department of Pharmaceutical Sciences, Center for Drug Discovery, College of Pharmacy, University of Nebraska Medical Center, Omaha, Nebraska, USA
| | - C. David Weaver
- Department of Pharmacology, Vanderbilt University, Nashville, Tennessee, USA
- vanderbilt Institute of Chemical Biology, Vanderbilt University, Nashville, Tennessee, USA
| |
Collapse
|
15
|
Itcho K, Oki K, Kobuke K, Ohno H, Yoneda M, Hattori N. Angiotensin 1-7 suppresses angiotensin II mediated aldosterone production via JAK/STAT signaling inhibition. J Steroid Biochem Mol Biol 2019; 185:137-141. [PMID: 30125658 DOI: 10.1016/j.jsbmb.2018.08.007] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/26/2018] [Revised: 08/03/2018] [Accepted: 08/12/2018] [Indexed: 01/27/2023]
Abstract
Angiotensin 1-7 (Ang 1-7), which is a protein cleaved from angiotensin II (A-II), binds to the MAS receptor. Ang 1-7 has been demonstrated to exert protective effects against A-II-mediated cardiac, atherosclerotic, and renal damages. The aims of our study were to demonstrate the inhibitory role of Ang 1-7 in A-II-mediated aldosterone production by interacting with the MAS receptor in human adrenocortical carcinoma (HAC15) cells, and clarify the intracellular signaling mechanisms underlying the inhibition of aldosterone production by Ang 1-7. Ang 1-7 significantly suppressed A-II-stimulated aldosterone production, and partially abrogated A-II-induced upregulation of CYP11B2 expression. Treatment with a selective Ang 1-7 antagonist abrogated Ang 1-7-mediated inhibition of aldosterone production in HAC15 cells. Incubation of A-II-treated HAC15 cells with conditioned medium containing Ang 1-7 was demonstrated to suppress A-II-mediated aldosterone production and CYP11B2 expression. Proteomic analysis showed that Ang 1-7 predominantly inhibited the phosphorylation of JAK-STAT proteins in A-II stimulated HAC15 cells. Treatment of HAC15 cells with a STAT3 inhibitor partially but significantly repressed A-II-mediated aldosterone production by 63.2%. Similarly, treatment with a STAT5 inhibitor significantly abrogated A-II-stimulated aldosterone production in HAC15 cells by 60.7%. In conclusion, we demonstrated that Ang 1-7 negatively regulates A-II-mediated aldosterone production, and the observed inhibition of aldosterone production was associated with JAK/STAT signaling in human adrenal cells. Therefore, activation of Ang 1-7 or stimulation of the MAS receptor, which inhibits aldosterone production, is a promising therapeutic approach for the prevention of cardiovascular events that can directly affect the target organs.
Collapse
Affiliation(s)
- Kiyotaka Itcho
- Department of Molecular and Internal Medicine, Graduate School of Biomedical and Health Sciences, Hiroshima University, 1-2-3 Kasumi, Minami-ku, Hiroshima, 734-8551, Japan
| | - Kenji Oki
- Department of Molecular and Internal Medicine, Graduate School of Biomedical and Health Sciences, Hiroshima University, 1-2-3 Kasumi, Minami-ku, Hiroshima, 734-8551, Japan.
| | - Kazuhiro Kobuke
- Department of Molecular and Internal Medicine, Graduate School of Biomedical and Health Sciences, Hiroshima University, 1-2-3 Kasumi, Minami-ku, Hiroshima, 734-8551, Japan
| | - Haruya Ohno
- Department of Molecular and Internal Medicine, Graduate School of Biomedical and Health Sciences, Hiroshima University, 1-2-3 Kasumi, Minami-ku, Hiroshima, 734-8551, Japan
| | - Masayasu Yoneda
- Department of Molecular and Internal Medicine, Graduate School of Biomedical and Health Sciences, Hiroshima University, 1-2-3 Kasumi, Minami-ku, Hiroshima, 734-8551, Japan
| | - Noboru Hattori
- Department of Molecular and Internal Medicine, Graduate School of Biomedical and Health Sciences, Hiroshima University, 1-2-3 Kasumi, Minami-ku, Hiroshima, 734-8551, Japan
| |
Collapse
|
16
|
Abstract
Primary aldosteronism (PA) is now considered as one of leading causes of secondary hypertension, accounting for 5-10% of all hypertensive patients and more strikingly 20% of those with resistant hypertension. Importantly, those with the unilateral disease could be surgically cured when diagnosed appropriately. On the other hand, only a very limited portion of those suspected to have PA has been screened, diagnosed, or treated to date. With current advancement in medical technologies and genetic research, expanding knowledge of PA has been accumulated and recent achievements have also been documented in the care of those with PA. This review is aimed to have focused description on updated topics of the following; importance of PA screening both in the general and specialized settings and careful interpretation of screening data, recent achievements in hormone assays and sampling methods and their clinical relevance, and expanding knowledge on PA genetics. Improvement in workup processes and novel treatment options, as well as better understanding of the PA pathogenesis based on genetic research, might be expected to result in increased cure and better care of the patients.
Collapse
Affiliation(s)
- Ryo Morimoto
- Division of Nephrology, Endocrinology and Vascular Medicine, Department of Medicine, Tohoku University Hospital, Sendai, Miyagi, Japan
| | - Kei Omata
- Division of Nephrology, Endocrinology and Vascular Medicine, Department of Medicine, Tohoku University Hospital, Sendai, Miyagi, Japan
- Division of Clinical Hypertension, Endocrinology and Metabolism, Tohoku University Graduate School of Medicine, Sendai, Miyagi, Japan
- Department of Pathology, University of Michigan, Ann Arbor, Michigan, USA
| | - Sadayoshi Ito
- Division of Nephrology, Endocrinology and Vascular Medicine, Department of Medicine, Tohoku University Hospital, Sendai, Miyagi, Japan
| | - Fumitoshi Satoh
- Division of Nephrology, Endocrinology and Vascular Medicine, Department of Medicine, Tohoku University Hospital, Sendai, Miyagi, Japan
- Division of Clinical Hypertension, Endocrinology and Metabolism, Tohoku University Graduate School of Medicine, Sendai, Miyagi, Japan
| |
Collapse
|
17
|
Regulation of aldosterone production by ion channels: From basal secretion to primary aldosteronism. Biochim Biophys Acta Mol Basis Dis 2018; 1864:871-881. [DOI: 10.1016/j.bbadis.2017.12.034] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2017] [Revised: 12/05/2017] [Accepted: 12/23/2017] [Indexed: 01/07/2023]
|
18
|
Abstract
Primary aldosteronism (PA) significantly increases the risk of cardiovascular complications, and early diagnosis and targeted treatment based on its pathophysiology is warranted. Next-generation sequencing (NGS) has revealed recurrent somatic mutations in aldosterone-driving genes in aldosterone-producing adenoma (APA). By applying CYP11B2 (aldosterone synthase) immunohistochemistry and NGS to adrenal glands from normal subjects and PA patients, we and others have shown that CYP11B2-positive cells make small clusters, termed aldosterone-producing cell clusters (APCC), beneath the adrenal capsule, and that APCC harbor somatic mutations in genes mutated in APA. We have shown that APCC are increased in CT-negative PA adrenals, while others showed potential progression from APCC to micro APA through mutations. These results suggest that APCC are a key factor for understanding the origin of PA, and further investigation on the relation between APCC and PA is highly needed.
Collapse
Affiliation(s)
- Kei Omata
- Department of Pathology, University of Michigan, 1500 E Medical Center Drive, 48109 Ann Arbor, MI, USA
- Division of Nephrology, Endocrinology and Vascular Medicine, Tohoku University, 2-1 Seiryo-machi, Aoba-ku, 980-0872 Sendai, Miyagi, Japan
- Division of Clinical Hypertension, Endocrinology & Metabolism, Tohoku University, 2-1 Seiryo-machi, Aoba-ku, 980-0872 Sendai, Miyagi, Japan
| | - Scott A. Tomlins
- Department of Pathology, University of Michigan, 1500 E Medical Center Drive, 48109 Ann Arbor, MI, USA
- Michigan Center for Translational Pathology, University of Michigan, 1500 E Medical Center Drive, 48109 Ann Arbor, MI, USA
- Department of Urology, University of Michigan, 1500 E Medical Center Drive, 48109 Ann Arbor, MI, USA
- Comprehensive Cancer Center, University of Michigan, 1500 E Medical Center Drive, 48109 Ann Arbor, MI, USA
| | - William E. Rainey
- Department of Molecular and Integrative Physiology, University of Michigan, 1500 E Medical Center Drive, 48109 Ann Arbor, MI, USA
- Department of Medicine, University of Michigan, 1500 E Medical Center Drive, 48109 Ann Arbor, MI, USA
| |
Collapse
|
19
|
Kobuke K, Oki K, Gomez-Sanchez CE, Gomez-Sanchez EP, Ohno H, Itcho K, Yoshii Y, Yoneda M, Hattori N. Calneuron 1 Increased Ca 2+ in the Endoplasmic Reticulum and Aldosterone Production in Aldosterone-Producing Adenoma. Hypertension 2017; 71:125-133. [PMID: 29109191 DOI: 10.1161/hypertensionaha.117.10205] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2017] [Revised: 09/01/2017] [Accepted: 10/10/2017] [Indexed: 11/16/2022]
Abstract
Aldosterone production is initiated by angiotensin II stimulation and activation of intracellular Ca2+ signaling. In aldosterone-producing adenoma (APA) cells, the activation of intracellular Ca2+ signaling is independent of the renin-angiotensin-aldosterone systems. The purpose of our study was to clarify molecular mechanisms of aldosterone production related to Ca2+ signaling. Transcriptome analysis revealed that the CALN1 gene encoding calneuron 1 had the strongest correlation with CYP11B2 (aldosterone synthase) among genes encoding Ca2+-binding proteins in APA. CALN1 modulation and synthetic or fluorescent compounds were used for functional studies in human adrenocortical carcinoma (HAC15) cells. CALN1 expression was 4.4-fold higher in APAs than nonfunctioning adrenocortical adenomas. CALN1 expression colocalized with CYP11B2 expression as investigated using immunohistochemistry in APA and zona glomerulosa of male rats fed by a low-salt diet. CALN1 expression was detected in the endoplasmic reticulum (ER) by using GFP-fused CALN1, CellLight ER-RFP, and the corresponding antibodies. CALN1-overexpressing HAC15 cells showed increased Ca2+ in the ER and cytosol fluorescence-based studies. Aldosterone production was potentiated in HAC15 cells by CALN1 expression, and dose-responsive inhibition with TMB-8 showed that CALN1-mediated Ca2+ storage in ER involved sarcoendoplasmic reticulum calcium transport ATPase. The silencing of CALN1 decreased Ca2+ in ER, and abrogated angiotensin II- or KCNJ5 T158A-mediated aldosterone production in HAC15 cells. Increased CALN1 expression in APA was associated with elevated Ca2+ storage in ER and aldosterone overproduction. Suppression of CALN1 expression prevented angiotensin II- or KCNJ5 T158A-mediated aldosterone production in HAC15 cells, suggesting that CALN1 is a potential therapeutic target for excess aldosterone production.
Collapse
Affiliation(s)
- Kazuhiro Kobuke
- From the Department of Molecular and Internal Medicine, Graduate School of Biomedical and Health Sciences, Hiroshima University, Japan (K.K., K.O., H.O., K.I., Y.Y., M.Y., N.H.); Division of Endocrinology, G.V. (Sonny) Montgomery VA Medical Center, Jackson, MS (C.E.G.-S., E.P.G.-S.); and University of Mississippi Medical Center, Jackson (C.E.G.-S., E.P.G.-S.)
| | - Kenji Oki
- From the Department of Molecular and Internal Medicine, Graduate School of Biomedical and Health Sciences, Hiroshima University, Japan (K.K., K.O., H.O., K.I., Y.Y., M.Y., N.H.); Division of Endocrinology, G.V. (Sonny) Montgomery VA Medical Center, Jackson, MS (C.E.G.-S., E.P.G.-S.); and University of Mississippi Medical Center, Jackson (C.E.G.-S., E.P.G.-S.).
| | - Celso E Gomez-Sanchez
- From the Department of Molecular and Internal Medicine, Graduate School of Biomedical and Health Sciences, Hiroshima University, Japan (K.K., K.O., H.O., K.I., Y.Y., M.Y., N.H.); Division of Endocrinology, G.V. (Sonny) Montgomery VA Medical Center, Jackson, MS (C.E.G.-S., E.P.G.-S.); and University of Mississippi Medical Center, Jackson (C.E.G.-S., E.P.G.-S.)
| | - Elise P Gomez-Sanchez
- From the Department of Molecular and Internal Medicine, Graduate School of Biomedical and Health Sciences, Hiroshima University, Japan (K.K., K.O., H.O., K.I., Y.Y., M.Y., N.H.); Division of Endocrinology, G.V. (Sonny) Montgomery VA Medical Center, Jackson, MS (C.E.G.-S., E.P.G.-S.); and University of Mississippi Medical Center, Jackson (C.E.G.-S., E.P.G.-S.)
| | - Haruya Ohno
- From the Department of Molecular and Internal Medicine, Graduate School of Biomedical and Health Sciences, Hiroshima University, Japan (K.K., K.O., H.O., K.I., Y.Y., M.Y., N.H.); Division of Endocrinology, G.V. (Sonny) Montgomery VA Medical Center, Jackson, MS (C.E.G.-S., E.P.G.-S.); and University of Mississippi Medical Center, Jackson (C.E.G.-S., E.P.G.-S.)
| | - Kiyotaka Itcho
- From the Department of Molecular and Internal Medicine, Graduate School of Biomedical and Health Sciences, Hiroshima University, Japan (K.K., K.O., H.O., K.I., Y.Y., M.Y., N.H.); Division of Endocrinology, G.V. (Sonny) Montgomery VA Medical Center, Jackson, MS (C.E.G.-S., E.P.G.-S.); and University of Mississippi Medical Center, Jackson (C.E.G.-S., E.P.G.-S.)
| | - Yoko Yoshii
- From the Department of Molecular and Internal Medicine, Graduate School of Biomedical and Health Sciences, Hiroshima University, Japan (K.K., K.O., H.O., K.I., Y.Y., M.Y., N.H.); Division of Endocrinology, G.V. (Sonny) Montgomery VA Medical Center, Jackson, MS (C.E.G.-S., E.P.G.-S.); and University of Mississippi Medical Center, Jackson (C.E.G.-S., E.P.G.-S.)
| | - Masayasu Yoneda
- From the Department of Molecular and Internal Medicine, Graduate School of Biomedical and Health Sciences, Hiroshima University, Japan (K.K., K.O., H.O., K.I., Y.Y., M.Y., N.H.); Division of Endocrinology, G.V. (Sonny) Montgomery VA Medical Center, Jackson, MS (C.E.G.-S., E.P.G.-S.); and University of Mississippi Medical Center, Jackson (C.E.G.-S., E.P.G.-S.)
| | - Noboru Hattori
- From the Department of Molecular and Internal Medicine, Graduate School of Biomedical and Health Sciences, Hiroshima University, Japan (K.K., K.O., H.O., K.I., Y.Y., M.Y., N.H.); Division of Endocrinology, G.V. (Sonny) Montgomery VA Medical Center, Jackson, MS (C.E.G.-S., E.P.G.-S.); and University of Mississippi Medical Center, Jackson (C.E.G.-S., E.P.G.-S.)
| |
Collapse
|
20
|
Bandulik S. Of channels and pumps: different ways to boost the aldosterone? Acta Physiol (Oxf) 2017; 220:332-360. [PMID: 27862984 DOI: 10.1111/apha.12832] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2016] [Revised: 08/10/2016] [Accepted: 11/11/2016] [Indexed: 01/19/2023]
Abstract
The mineralocorticoid aldosterone is a major factor controlling the salt and water balance and thereby also the arterial blood pressure. Accordingly, primary aldosteronism (PA) characterized by an inappropriately high aldosterone secretion is the most common form of secondary hypertension. The physiological stimulation of aldosterone synthesis in adrenocortical glomerulosa cells by angiotensin II and an increased plasma K+ concentration depends on a membrane depolarization and an increase in the cytosolic Ca2+ activity. Recurrent gain-of-function mutations of ion channels and transporters have been identified in a majority of cases of aldosterone-producing adenomas and in familial forms of PA. In this review, the physiological role of these genes in the regulation of aldosterone synthesis and the altered function of the mutant proteins as well are described. The specific changes of the membrane potential and the cellular ion homoeostasis in adrenal cells expressing the different mutants are compared, and their impact on autonomous aldosterone production and proliferation is discussed.
Collapse
Affiliation(s)
- S. Bandulik
- Medical Cell Biology; University of Regensburg; Regensburg Germany
| |
Collapse
|
21
|
Scholl UI, Abriola L, Zhang C, Reimer EN, Plummer M, Kazmierczak BI, Zhang J, Hoyer D, Merkel JS, Wang W, Lifton RP. Macrolides selectively inhibit mutant KCNJ5 potassium channels that cause aldosterone-producing adenoma. J Clin Invest 2017; 127:2739-2750. [PMID: 28604387 PMCID: PMC5490757 DOI: 10.1172/jci91733] [Citation(s) in RCA: 51] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2016] [Accepted: 04/20/2017] [Indexed: 11/17/2022] Open
Abstract
Aldosterone-producing adenomas (APAs) are benign tumors of the adrenal gland that constitutively produce the salt-retaining steroid hormone aldosterone and cause millions of cases of severe hypertension worldwide. Either of 2 somatic mutations in the potassium channel KCNJ5 (G151R and L168R, hereafter referred to as KCNJ5MUT) in adrenocortical cells account for half of APAs worldwide. These mutations alter channel selectivity to allow abnormal Na+ conductance, resulting in membrane depolarization, calcium influx, aldosterone production, and cell proliferation. Because APA diagnosis requires a difficult invasive procedure, patients often remain undiagnosed and inadequately treated. Inhibitors of KCNJ5MUT could allow noninvasive diagnosis and therapy of APAs carrying KCNJ5 mutations. Here, we developed a high-throughput screen for rescue of KCNJ5MUT-induced lethality and identified a series of macrolide antibiotics, including roxithromycin, that potently inhibit KCNJ5MUT, but not KCNJ5WT. Electrophysiology demonstrated direct KCNJ5MUT inhibition. In human aldosterone-producing adrenocortical cancer cell lines, roxithromycin inhibited KCNJ5MUT-induced induction of CYP11B2 (encoding aldosterone synthase) expression and aldosterone production. Further exploration of macrolides showed that KCNJ5MUT was similarly selectively inhibited by idremcinal, a macrolide motilin receptor agonist, and by synthesized macrolide derivatives lacking antibiotic or motilide activity. Macrolide-derived selective KCNJ5MUT inhibitors thus have the potential to advance the diagnosis and treatment of APAs harboring KCNJ5MUT.
Collapse
Affiliation(s)
- Ute I Scholl
- Department of Genetics and Howard Hughes Medical Institute, Yale University School of Medicine, New Haven, Connecticut, USA.,Department of Nephrology, Medical School, Heinrich Heine University and University Hospital Düsseldorf, Düsseldorf, Germany
| | - Laura Abriola
- Yale Center for Molecular Discovery, Yale University, West Haven, Connecticut, USA
| | - Chengbiao Zhang
- Department of Pharmacology, New York Medical College, Valhalla, New York, USA
| | - Esther N Reimer
- Department of Nephrology, Medical School, Heinrich Heine University and University Hospital Düsseldorf, Düsseldorf, Germany
| | - Mark Plummer
- Yale Center for Molecular Discovery, Yale University, West Haven, Connecticut, USA
| | - Barbara I Kazmierczak
- Department of Medicine (Infectious Diseases), Yale University School of Medicine, New Haven, Connecticut, USA
| | - Junhui Zhang
- Department of Genetics and Howard Hughes Medical Institute, Yale University School of Medicine, New Haven, Connecticut, USA
| | - Denton Hoyer
- Yale Center for Molecular Discovery, Yale University, West Haven, Connecticut, USA
| | - Jane S Merkel
- Yale Center for Molecular Discovery, Yale University, West Haven, Connecticut, USA
| | - Wenhui Wang
- Department of Pharmacology, New York Medical College, Valhalla, New York, USA
| | - Richard P Lifton
- Department of Genetics and Howard Hughes Medical Institute, Yale University School of Medicine, New Haven, Connecticut, USA
| |
Collapse
|
22
|
Familial hyperaldosteronism type III. J Hum Hypertens 2017; 31:776-781. [PMID: 28447626 DOI: 10.1038/jhh.2017.34] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2016] [Revised: 01/15/2017] [Accepted: 02/03/2017] [Indexed: 11/08/2022]
Abstract
Primary aldosteronism is the most common form of endocrine hypertension. This disorder comprises both sporadic and familial forms. Four familial forms of primary aldosteronism (FH-I to FH-IV) have been described. FH-III is caused by germline mutations in KCNJ5, encoding the potassium channel Kir3.4 (also called GIRK4). These mutations alter the selectivity filter of the channel and lead to abnormal ion currents with loss of potassium selectivity, sodium influx and consequent increased intracellular calcium that causes excessive aldosterone biosynthesis. To date, eleven families have been reported, carrying six different mutations. Although the clinical features are variable, FH-III patients often display severe hyperaldosteronism with an early onset, associated with hypokalemia and diabetes insipidus-like symptoms. In most cases FH-III patients are resistant to pharmacological therapy and require bilateral adrenalectomy to control symptoms. In the present manuscript, we review the genetics and pathological basis of FH-III, the diagnostic work-up, clinical features and therapeutic management. Finally, we will describe a new case of FH-III of an Italian patient carrying a Gly151Arg mutation.
Collapse
|
23
|
Gomez-Sanchez CE, Qi X, Gomez-Sanchez EP, Sasano H, Bohlen MO, Wisgerhof M. Disordered zonal and cellular CYP11B2 enzyme expression in familial hyperaldosteronism type 3. Mol Cell Endocrinol 2017; 439:74-80. [PMID: 27793677 PMCID: PMC5123946 DOI: 10.1016/j.mce.2016.10.025] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/15/2016] [Revised: 10/23/2016] [Accepted: 10/24/2016] [Indexed: 11/27/2022]
Abstract
Three forms of familial primary aldosteronism have been recognized. Familial Hyperaldosteronism type 1 (FH1) or dexamethasone suppressible hyperaldosteronism, FH2, the most common form of as yet unknown cause(s), and FH3. FH3 is due to activating mutations of the potassium channel gene KCNJ5 that increase constitutive and angiotensin II-induced aldosterone synthesis. In this study we examined the cellular distribution of CYP11B2, CYP11B1, CYP17A1 and KCNJ5 in adrenals from two FH3 siblings using immunohistochemistry and immunofluorescence and obtained unexpected results. The adrenals were markedly enlarged with loss of zonation. CYP11B2 was expressed sporadically throughout the adrenal cortex. CYP11B2 was most often expressed by itself, relatively frequently with CYP17A1, and less frequently with CYP11B1. KCNJ5 was co-expressed with CYP11B2 and in some cells with CYP11B1. This aberrant co-expression of enzymes likely explains the abnormally high secretion rate of the hybrid steroid, 18-oxocortisol.
Collapse
Affiliation(s)
- Celso E Gomez-Sanchez
- Endocrinology Division, G.V. (Sonny) Montgomery VA Medical Center, Jackson, MS, United States; University of Mississippi Medical Center, Jackson, MS, United States.
| | - Xin Qi
- University of Mississippi Medical Center, Jackson, MS, United States
| | - Elise P Gomez-Sanchez
- Department of Pharmacology and Toxicology and Medicine, University of Mississippi Medical Center, Jackson, MS, United States
| | | | - Martin O Bohlen
- Department of Anatomical Sciences, University of Mississippi Medical Center, Jackson, MS, United States
| | - Max Wisgerhof
- Division of Endocrinology, Henry Ford Health System, Detroit, MI, United States
| |
Collapse
|
24
|
Abstract
The recently available genomic sequencing techniques have led to breakthroughs in understanding of the underlying genetic mechanisms in adrenocortical tumours. Disease-causing mutations have been described for aldosterone-producing adenomas, cortisol-producing adenomas and adrenocortical carcinomas. Further, knowledge gained from transcriptome analyses and methylation arrays has provided new insights into the development of these tumours. Elucidation of the genomic landscape of adrenocortical tumours and improved techniques may in the future be useful for early diagnosis through the detection of mutated DNA in the circulation. Moreover, compounds that bind specifically to altered proteins may be used as screening targets or therapeutic agents. Regulation of cortisol release by interaction with an altered subunit in adenylate cyclase may be more complex, but may provide a new option for regulating steroid release. Information about derangements in adrenocortical carcinoma is already helpful for determining patient prognosis. With further knowledge, we may be able to identify novel biomarkers that effectively and noninvasively help in differentiating between benign and malignant disease. It is clear that the next few years will provide much novel information that hopefully will aid in the treatment of patients with adrenocortical tumours.
Collapse
Affiliation(s)
- T Åkerström
- Department of Surgical Sciences, Uppsala University, Uppsala, Sweden
| | - T Carling
- Endocrine Research Unit, Yale University, New Haven, CT, USA
| | - F Beuschlein
- Medizinische Klinik und Poliklinik IV, Klinikum der Universität München, Munich, Germany
| | - P Hellman
- Department of Surgical Sciences, Uppsala University, Uppsala, Sweden
| |
Collapse
|
25
|
Stowasser M, Gordon RD. Primary Aldosteronism: Changing Definitions and New Concepts of Physiology and Pathophysiology Both Inside and Outside the Kidney. Physiol Rev 2016; 96:1327-84. [DOI: 10.1152/physrev.00026.2015] [Citation(s) in RCA: 86] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
In the 60 years that have passed since the discovery of the mineralocorticoid hormone aldosterone, much has been learned about its synthesis (both adrenal and extra-adrenal), regulation (by renin-angiotensin II, potassium, adrenocorticotrophin, and other factors), and effects (on both epithelial and nonepithelial tissues). Once thought to be rare, primary aldosteronism (PA, in which aldosterone secretion by the adrenal is excessive and autonomous of its principal regulator, angiotensin II) is now known to be the most common specifically treatable and potentially curable form of hypertension, with most patients lacking the clinical feature of hypokalemia, the presence of which was previously considered to be necessary to warrant further efforts towards confirming a diagnosis of PA. This, and the appreciation that aldosterone excess leads to adverse cardiovascular, renal, central nervous, and psychological effects, that are at least partly independent of its effects on blood pressure, have had a profound influence on raising clinical and research interest in PA. Such research on patients with PA has, in turn, furthered knowledge regarding aldosterone synthesis, regulation, and effects. This review summarizes current progress in our understanding of the physiology of aldosterone, and towards defining the causes (including genetic bases), epidemiology, outcomes, and clinical approaches to diagnostic workup (including screening, diagnostic confirmation, and subtype differentiation) and treatment of PA.
Collapse
Affiliation(s)
- Michael Stowasser
- Endocrine Hypertension Research Centre, University of Queensland School of Medicine, Greenslopes and Princess Alexandra Hospitals, Brisbane, Queensland, Australia
| | - Richard D. Gordon
- Endocrine Hypertension Research Centre, University of Queensland School of Medicine, Greenslopes and Princess Alexandra Hospitals, Brisbane, Queensland, Australia
| |
Collapse
|
26
|
Seidel E, Scholl UI. Intracellular Molecular Differences in Aldosterone- Compared to Cortisol-Secreting Adrenal Cortical Adenomas. Front Endocrinol (Lausanne) 2016; 7:75. [PMID: 27445978 PMCID: PMC4921773 DOI: 10.3389/fendo.2016.00075] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/30/2016] [Accepted: 06/14/2016] [Indexed: 12/18/2022] Open
Abstract
The adrenal cortex is a major site of steroid hormone production. Two hormones are of particular importance: aldosterone, which is produced in the zona glomerulosa in response to volume depletion and hyperkalemia, and cortisol, which is produced in the zona fasciculata in response to stress. In both cases, acute stimulation leads to increased hormone production, and chronic stimulation causes hyperplasia of the respective zone. Aldosterone- and cortisol-producing adenomas (APAs and CPAs) are benign tumors of the adrenal cortex that cause excess hormone production, leading to primary aldosteronism and Cushing's syndrome, respectively. About 40% of the APAs carry somatic heterozygous gain-of-function mutations in the K(+) channel KCNJ5. These mutations lead to sodium permeability, depolarization, activation of voltage-gated Ca(2+) channels, and Ca(2+) influx. Mutations in the Na(+)/K(+)-ATPase subunit ATP1A1 and the plasma membrane Ca(2+)-ATPase ATP2B3 similarly cause Na(+) or H(+) permeability and depolarization, whereas mutations in the Ca(2+) channel CACNA1D directly lead to increased calcium influx. One in three CPAs carries a recurrent gain-of-function mutation (L206R) in the PRKACA gene, encoding the catalytic subunit of PKA. This mutation causes constitutive PKA activity by abolishing the binding of the inhibitory regulatory subunit to the catalytic subunit. These mutations activate pathways that are relatively specific to the respective cell type (glomerulosa versus fasciculata), and there is little overlap in mutation spectrum between APAs and CPAs, but co-secretion of both hormones can occur. Mutations in CTNNB1 (beta-catenin) and GNAS (Gsα) are exceptions, as they can cause both APAs and CPAs through pathways that are incompletely understood.
Collapse
Affiliation(s)
- Eric Seidel
- Department of Nephrology, University Hospital Düsseldorf, Heinrich Heine University, Düsseldorf, Germany
| | - Ute I. Scholl
- Department of Nephrology, University Hospital Düsseldorf, Heinrich Heine University, Düsseldorf, Germany
- *Correspondence: Ute I. Scholl,
| |
Collapse
|
27
|
Chen AX, Nishimoto K, Nanba K, Rainey WE. Potassium channels related to primary aldosteronism: Expression similarities and differences between human and rat adrenals. Mol Cell Endocrinol 2015; 417:141-8. [PMID: 26375812 PMCID: PMC4646165 DOI: 10.1016/j.mce.2015.09.011] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/07/2014] [Revised: 08/18/2015] [Accepted: 09/10/2015] [Indexed: 11/26/2022]
Abstract
Three potassium channels have been associated with primary aldosteronism (PA) in rodents and humans: KCNK3 (TASK-1), KCNK9 (TASK-3), and KCNJ5 (Kir3.4). Mice with deficiency in Kcnk3 and Kcnk9 have elevated aldosterone production and blood pressure. In humans, adrenal tumors with somatic mutations in KCNJ5 cause PA. However, there are very few reports on the expression patterns of these genes in humans versus rodents. Herein, we compared human and rat mRNA expression (by quantitative real-time polymerase chain reaction (qPCR) and protein levels (by immunohistochemistry) across three tissues (adrenal, brain, heart) and two laser-captured adrenal zones (zona glomerulosa, zona fasciculata). Our findings show that expression patterns of KCNK3, KCNK9, and KCNJ5 are inconsistent between rats and humans across both tissues and adrenal zones. Thus, species variation in the expression of PA-related potassium channels indicates an evolutionary divergence in their role in regulating adrenal aldosterone production.
Collapse
Affiliation(s)
- Andrew X Chen
- Department of Molecular and Integrative Physiology, University of Michigan, 1150 W. Medical Center Dr, Ann Arbor, MI 48109, USA; Department of Internal Medicine, University of Michigan, 1150 W. Medical Center Dr, Ann Arbor, MI 48109, USA
| | - Koshiro Nishimoto
- Department of Molecular and Integrative Physiology, University of Michigan, 1150 W. Medical Center Dr, Ann Arbor, MI 48109, USA; Department of Internal Medicine, University of Michigan, 1150 W. Medical Center Dr, Ann Arbor, MI 48109, USA
| | - Kazutaka Nanba
- Department of Molecular and Integrative Physiology, University of Michigan, 1150 W. Medical Center Dr, Ann Arbor, MI 48109, USA; Department of Internal Medicine, University of Michigan, 1150 W. Medical Center Dr, Ann Arbor, MI 48109, USA
| | - William E Rainey
- Department of Molecular and Integrative Physiology, University of Michigan, 1150 W. Medical Center Dr, Ann Arbor, MI 48109, USA; Department of Internal Medicine, University of Michigan, 1150 W. Medical Center Dr, Ann Arbor, MI 48109, USA.
| |
Collapse
|
28
|
Stindl J, Tauber P, Sterner C, Tegtmeier I, Warth R, Bandulik S. Pathogenesis of Adrenal Aldosterone-Producing Adenomas Carrying Mutations of the Na(+)/K(+)-ATPase. Endocrinology 2015; 156:4582-91. [PMID: 26418325 DOI: 10.1210/en.2015-1466] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Aldosterone-producing adenoma (APA) is a major cause of primary aldosteronism, leading to secondary hypertension. Somatic mutations in the gene for the α1 subunit of the Na(+)/K(+)-ATPase were found in about 6% of APAs. APA-related α1 subunit of the Na(+)/K(+)-ATPase mutations lead to a loss of the pump function of the Na(+)/K(+)-ATPase, which is believed to result in membrane depolarization and Ca(2+)-dependent stimulation of aldosterone synthesis in adrenal cells. In addition, H(+) and Na(+) leak currents via the mutant Na(+)/K(+)-ATPase were suggested to contribute to the phenotype. The aim of this study was to investigate the cellular pathophysiology of adenoma-associated Na(+)/K(+)-ATPase mutants (L104R, V332G, G99R) in adrenocortical NCI-H295R cells. The expression of these Na(+)/K(+)-ATPase mutants depolarized adrenal cells and stimulated aldosterone secretion. However, an increase of basal cytosolic Ca(2+) levels in Na(+)/K(+)-ATPase mutant cells was not detectable, and stimulation with high extracellular K(+) hardly increased Ca(2+) levels in cells expressing L104R and V332G mutant Na(+)/K(+)-ATPase. Cytosolic pH measurements revealed an acidification of L104R and V332G mutant cells, despite an increased activity of the Na(+)/H(+) exchanger. The possible contribution of cellular acidification to the hypersecretion of aldosterone was supported by the observation that aldosterone secretion of normal adrenocortical cells was stimulated by acetate-induced acidification. Taken together, mutations of the Na(+)/K(+)-ATPase depolarize adrenocortical cells, disturb the K(+) sensitivity, and lower intracellular pH but, surprisingly, do not induce an overt increase of intracellular Ca(2+). Probably, the autonomous aldosterone secretion is caused by the concerted action of several pathological signaling pathways and incomplete cellular compensation.
Collapse
Affiliation(s)
- J Stindl
- Medical Cell Biology, University of Regensburg, 93053 Regensburg, Germany
| | - P Tauber
- Medical Cell Biology, University of Regensburg, 93053 Regensburg, Germany
| | - C Sterner
- Medical Cell Biology, University of Regensburg, 93053 Regensburg, Germany
| | - I Tegtmeier
- Medical Cell Biology, University of Regensburg, 93053 Regensburg, Germany
| | - R Warth
- Medical Cell Biology, University of Regensburg, 93053 Regensburg, Germany
| | - S Bandulik
- Medical Cell Biology, University of Regensburg, 93053 Regensburg, Germany
| |
Collapse
|
29
|
Zennaro MC, Fernandes-Rosa F, Boulkroun S, Jeunemaitre X. Bilateral Idiopathic Adrenal Hyperplasia: Genetics and Beyond. Horm Metab Res 2015; 47:947-52. [PMID: 26610199 DOI: 10.1055/s-0035-1565198] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/22/2022]
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.
Collapse
Affiliation(s)
- M-C Zennaro
- INSERM, UMRS 970, Paris Cardiovascular Research Center, Paris, France
| | - F Fernandes-Rosa
- INSERM, UMRS 970, Paris Cardiovascular Research Center, Paris, France
| | - S Boulkroun
- INSERM, UMRS 970, Paris Cardiovascular Research Center, Paris, France
| | - X Jeunemaitre
- INSERM, UMRS 970, Paris Cardiovascular Research Center, Paris, France
| |
Collapse
|
30
|
Kienitz MC, Mergia E, Pott L. NCI-H295R cell line as in vitro model of hyperaldosteronism lacks functional KCNJ5 (GIRK4; Kir3.4) channels. Mol Cell Endocrinol 2015; 412:272-80. [PMID: 25998841 DOI: 10.1016/j.mce.2015.05.013] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/10/2015] [Revised: 04/22/2015] [Accepted: 05/11/2015] [Indexed: 11/22/2022]
Abstract
As a major cause of aldosterone producing adenomas, numerous gain-of-function mutations in the KCNJ5 gene (encoding the K(+) channel subunit GIRK4) have been identified. The human adrenocortical carcinoma cell line NCI-H295R is the most frequently used cellular model for in vitro studies related to regulation of aldosterone-synthesis. Because of the undefined role of KCNJ5 (GIRK4) in regulating synthesis of aldosterone, we aimed at identifying basal and G protein-activated GIRK4 currents in this paradigmatic cell line. The GIRK-specific blocker Tertiapin-Q did not affect basal current. Neither loading of the cells with GTP-γ-S via the patch-clamp pipette nor agonist stimulation of an infected A1-adenosine receptor resulted in activation of GIRK current. In cells co-infected with KCNJ5, robust activation of basal and adenosine-activated inward-rectifying current was observed. Although GIRK4 protein can be detected in Western blots of H295R homogenates, we suggest that GIRK4 in aldosterone-producing cells does not form functional G(βγ)-activated channels.
Collapse
Affiliation(s)
| | - Evanthia Mergia
- Department of Pharmacology and Toxicology, Ruhr-University Bochum, D-44780 Bochum, Germany
| | - Lutz Pott
- Institute of Physiology, Ruhr-University Bochum, D-44780 Bochum, Germany
| |
Collapse
|
31
|
Abstract
Aldosterone plays an essential role in the maintenance of fluid and electrolyte homeostasis in the distal nephron. Monogenic forms of mineralocorticoid hypertension result from genetic defects leading to excessive production of aldosterone (or other mineralocorticoids) from the adrenal cortex or to illegitimate mineralocorticoid effects in the kidney. They are characterized in the majority of cases by early onset, severe or resistant hypertension and associated with suppressed renin levels. Depending on their causes, these diseases are distinguished at the clinical and biochemical level and differently affect aldosterone levels and kalemia. The diagnosis is confirmed by genetic testing, which allows in many cases targeted treatment to prevent severe cardiovascular consequences of high blood pressure or aldosterone excess. In this review we describe the different forms of inherited mineralocorticoid hypertension, providing an overview of their clinical and biochemical features, their underlying genetic defects and specific therapeutic options.
Collapse
Affiliation(s)
- Maria-Christina Zennaro
- INSERM, UMRS_970, Paris Cardiovascular Research Center, Paris, France; University Paris Descartes, Sorbonne Paris Cité, Paris, France; Assistance Publique-Hôpitaux de Paris, Hôpital Européen Georges Pompidou, Service de Génétique, Paris, France.
| | - Sheerazed Boulkroun
- INSERM, UMRS_970, Paris Cardiovascular Research Center, Paris, France; University Paris Descartes, Sorbonne Paris Cité, Paris, France
| | - Fabio Fernandes-Rosa
- INSERM, UMRS_970, Paris Cardiovascular Research Center, Paris, France; University Paris Descartes, Sorbonne Paris Cité, Paris, France; Assistance Publique-Hôpitaux de Paris, Hôpital Européen Georges Pompidou, Service de Génétique, Paris, France
| |
Collapse
|
32
|
Bandulik S, Tauber P, Lalli E, Barhanin J, Warth R. Two-pore domain potassium channels in the adrenal cortex. Pflugers Arch 2015; 467:1027-1042. [PMID: 25339223 PMCID: PMC4428839 DOI: 10.1007/s00424-014-1628-6] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2014] [Revised: 10/02/2014] [Accepted: 10/03/2014] [Indexed: 12/31/2022]
Abstract
The physiological control of steroid hormone secretion from the adrenal cortex depends on the function of potassium channels. The "two-pore domain K(+) channels" (K2P) TWIK-related acid sensitive K(+) channel 1 (TASK1), TASK3, and TWIK-related K(+) channel 1 (TREK1) are strongly expressed in adrenocortical cells. They confer a background K(+) conductance to these cells which is important for the K(+) sensitivity as well as for angiotensin II and adrenocorticotropic hormone-dependent stimulation of aldosterone and cortisol synthesis. Mice with single deletions of the Task1 or Task3 gene as well as Task1/Task3 double knockout mice display partially autonomous aldosterone synthesis. It appears that TASK1 and TASK3 serve different functions: TASK1 affects cell differentiation and prevents expression of aldosterone synthase in the zona fasciculata, while TASK3 controls aldosterone secretion in glomerulosa cells. TREK1 is involved in the regulation of cortisol secretion in fasciculata cells. These data suggest that a disturbed function of K2P channels could contribute to adrenocortical pathologies in humans.
Collapse
Affiliation(s)
- Sascha Bandulik
- Medical Cell Biology, University of Regensburg, Universitaetsstrasse 31, 93053, Regensburg, Germany,
| | | | | | | | | |
Collapse
|
33
|
Cheng CJ, Sung CC, Huang CL, Lin SH. Inward-rectifying potassium channelopathies: new insights into disorders of sodium and potassium homeostasis. Pediatr Nephrol 2015; 30:373-83. [PMID: 24899236 DOI: 10.1007/s00467-014-2764-0] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/29/2013] [Revised: 12/11/2013] [Accepted: 01/10/2014] [Indexed: 11/30/2022]
Abstract
Inward-rectifying potassium (Kir) channels allow more inward than outward potassium flux when channels are open in mammalian cells. At physiological resting membrane potentials, however, they predominantly mediate outward potassium flux and play important roles in regulating the resting membrane potential in diverse cell types and potassium secretion in the kidneys. Mutations of Kir channels cause human hereditary diseases collectively called Kir channelopathies, many of which are characterized by disorders of sodium and potassium homeostasis. Studies on these genetic Kir channelopathies have shed light on novel pathophysiological mechanisms, including renal sodium and potassium handling, potassium shifting in skeletal muscles, and aldosterone production in the adrenal glands. Here, we review several recent advances in Kir channels and their clinical implications in sodium and potassium homeostasis.
Collapse
Affiliation(s)
- Chih-Jen Cheng
- Department of Medicine, Division of Nephrology, Tri-Service General Hospital, National Defense Medical Center, No. 325, Section 2, Cheng-Kung Road, Neihu 114, Taipei, Taiwan
| | | | | | | |
Collapse
|
34
|
Abstract
Aldosterone is a steroid hormone synthesized in and secreted from the outer layer of the adrenal cortex, the zona glomerulosa. Aldosterone is responsible for regulating sodium homeostasis, thereby helping to control blood volume and blood pressure. Insufficient aldosterone secretion can lead to hypotension and circulatory shock, particularly in infancy. On the other hand, excessive aldosterone levels, or those too high for sodium status, can cause hypertension and exacerbate the effects of high blood pressure on multiple organs, contributing to renal disease, stroke, visual loss, and congestive heart failure. Aldosterone is also thought to directly induce end-organ damage, including in the kidneys and heart. Because of the significance of aldosterone to the physiology and pathophysiology of the cardiovascular system, it is important to understand the regulation of its biosynthesis and secretion from the adrenal cortex. Herein, the mechanisms regulating aldosterone production in zona glomerulosa cells are discussed, with a particular emphasis on signaling pathways involved in the secretory response to the main controllers of aldosterone production, the renin-angiotensin II system, serum potassium levels and adrenocorticotrophic hormone. The signaling pathways involved include phospholipase C-mediated phosphoinositide hydrolysis, inositol 1,4,5-trisphosphate, cytosolic calcium levels, calcium influx pathways, calcium/calmodulin-dependent protein kinases, diacylglycerol, protein kinases C and D, 12-hydroxyeicostetraenoic acid, phospholipase D, mitogen-activated protein kinase pathways, tyrosine kinases, adenylate cyclase, and cAMP-dependent protein kinase. A complete understanding of the signaling events regulating aldosterone biosynthesis may allow the identification of novel targets for therapeutic interventions in hypertension, primary aldosteronism, congestive heart failure, renal disease, and other cardiovascular disorders.
Collapse
Affiliation(s)
- Wendy B Bollag
- Charlie Norwood VA Medical Center, Augusta, Georgia; Department of Physiology, Medical College of Georgia at Georgia Regents University, Augusta, Georgia
| |
Collapse
|
35
|
Abstract
Primary aldosteronism (PA) is the most common and curable form of secondary hypertension. It is caused in the majority of cases by either unilateral aldosterone overproduction due to an aldosterone-producing adenoma (APA) or by bilateral adrenal hyperplasia. Recent advances in genome technology have allowed researchers to unravel part of the genetic abnormalities underlying the development of APA and familial hyperaldosteronism. Recurrent somatic mutations in genes coding for ion channels (KCNJ5 and CACNA1D) and ATPases (ATP1A1 and ATP2B3) regulating intracellular ionic homeostasis and cell membrane potential have been identified in APA. Similar germline mutations of KCNJ5 were identified in a severe familial form of PA, familial hyperaldosteronism type 3 (FH3), whereas de novo germline CACNA1D mutations were found in two cases of hyperaldosteronism associated with a complex neurological disorder. These results have allowed a pathophysiological model of APA development to be established. This model involves modifications in intracellular ionic homeostasis and membrane potential, accounting for ∼50% of all tumors, associated with specific gender differences and severity of PA. In this review, we describe the different genetic abnormalities associated with PA and discuss the mechanisms whereby they lead to increased aldosterone production and cell proliferation. We also address some of the foreseeable consequences that genetic knowledge may contribute to improve diagnosis and patient care.
Collapse
Affiliation(s)
- Maria-Christina Zennaro
- INSERMUMRS_970, Paris Cardiovascular Research Center - PARCC, 56, rue Leblanc, 75015 Paris, FranceUniversity Paris DescartesSorbonne Paris Cité, Paris, FranceAssistance Publique-Hôpitaux de ParisHôpital Européen Georges Pompidou, Service de Génétique, Paris, France INSERMUMRS_970, Paris Cardiovascular Research Center - PARCC, 56, rue Leblanc, 75015 Paris, FranceUniversity Paris DescartesSorbonne Paris Cité, Paris, FranceAssistance Publique-Hôpitaux de ParisHôpital Européen Georges Pompidou, Service de Génétique, Paris, France INSERMUMRS_970, Paris Cardiovascular Research Center - PARCC, 56, rue Leblanc, 75015 Paris, FranceUniversity Paris DescartesSorbonne Paris Cité, Paris, FranceAssistance Publique-Hôpitaux de ParisHôpital Européen Georges Pompidou, Service de Génétique, Paris, France
| | - Sheerazed Boulkroun
- INSERMUMRS_970, Paris Cardiovascular Research Center - PARCC, 56, rue Leblanc, 75015 Paris, FranceUniversity Paris DescartesSorbonne Paris Cité, Paris, FranceAssistance Publique-Hôpitaux de ParisHôpital Européen Georges Pompidou, Service de Génétique, Paris, France INSERMUMRS_970, Paris Cardiovascular Research Center - PARCC, 56, rue Leblanc, 75015 Paris, FranceUniversity Paris DescartesSorbonne Paris Cité, Paris, FranceAssistance Publique-Hôpitaux de ParisHôpital Européen Georges Pompidou, Service de Génétique, Paris, France
| | - Fabio Fernandes-Rosa
- INSERMUMRS_970, Paris Cardiovascular Research Center - PARCC, 56, rue Leblanc, 75015 Paris, FranceUniversity Paris DescartesSorbonne Paris Cité, Paris, FranceAssistance Publique-Hôpitaux de ParisHôpital Européen Georges Pompidou, Service de Génétique, Paris, France INSERMUMRS_970, Paris Cardiovascular Research Center - PARCC, 56, rue Leblanc, 75015 Paris, FranceUniversity Paris DescartesSorbonne Paris Cité, Paris, FranceAssistance Publique-Hôpitaux de ParisHôpital Européen Georges Pompidou, Service de Génétique, Paris, France INSERMUMRS_970, Paris Cardiovascular Research Center - PARCC, 56, rue Leblanc, 75015 Paris, FranceUniversity Paris DescartesSorbonne Paris Cité, Paris, FranceAssistance Publique-Hôpitaux de ParisHôpital Européen Georges Pompidou, Service de Génétique, Paris, France
| |
Collapse
|
36
|
Monticone S, Else T, Mulatero P, Williams TA, Rainey WE. Understanding primary aldosteronism: impact of next generation sequencing and expression profiling. Mol Cell Endocrinol 2015; 399:311-20. [PMID: 25240470 PMCID: PMC4285708 DOI: 10.1016/j.mce.2014.09.015] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/02/2014] [Revised: 09/11/2014] [Accepted: 09/15/2014] [Indexed: 10/24/2022]
Abstract
Primary aldosteronism (PA) encompasses a broad, heterogeneous group of disorders including both sporadic and familial forms (familial hyperaldosteronism type I, II and III). PA is the most common form of secondary hypertension and associated with a higher rate of cardiovascular complications, compared with essential hypertension. Despite significant progress in the diagnosis and management of PA, until recently the molecular mechanisms leading to inappropriate aldosterone production were largely unknown. The introduction of next-generation sequencing has had a profound impact on the field of human genetics and has given new insight in the molecular determinants that lead to both sporadic and familial forms of PA. Here we review the recent progress toward understanding of the genetic and molecular mechanisms leading to autonomous aldosterone production in PA.
Collapse
Affiliation(s)
- Silvia Monticone
- Department of Medical Sciences, Division of Internal Medicine and Hypertension, University of Torino, Torino, Italy.
| | - Tobias Else
- Department of Internal Medicine, Division of Metabolism, Endocrinology and Diabetes, University of Michigan Medical School, Ann Arbor, MI, USA
| | - Paolo Mulatero
- Department of Medical Sciences, Division of Internal Medicine and Hypertension, University of Torino, Torino, Italy
| | - Tracy A Williams
- Department of Medical Sciences, Division of Internal Medicine and Hypertension, University of Torino, Torino, Italy
| | - William E Rainey
- Department of Molecular and Integrative Physiology, University of Michigan Medical School, Ann Arbor, MI, USA
| |
Collapse
|
37
|
Velarde-Miranda C, Gomez-Sanchez EP, Gomez-Sanchez CE. Regulation of aldosterone biosynthesis by the Kir3.4 (KCNJ5) potassium channel. Clin Exp Pharmacol Physiol 2014; 40:895-901. [PMID: 23829355 DOI: 10.1111/1440-1681.12151] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2013] [Revised: 06/30/2013] [Accepted: 07/01/2013] [Indexed: 11/30/2022]
Abstract
The G-protein-activated inwardly rectifying potassium channel Kir3.4 is expressed in the zona glomerulosa cell membrane and transports potassium out of the cell. Angiotensin II stimulation of aldosterone secretion is mediated, in part, by suppression of the transcription of KCNJ5, the gene coding for Kir3.4, and blocking channel activity. This results in membrane depolarization, mobilization of intracellular calcium, activation of the calcium-calmodulin pathway and increasing gene transcription of steroidogenic enzymes required for aldosterone secretion. In 40-60% of aldosterone-producing adenomas there is a somatic mutation in the region of the KCNJ5 gene that codes for the selectivity filter that decreases potassium selectivity, allowing sodium to leak into the cells, thus depolarizing the membrane and initiating events that result in increased aldosterone synthesis. The mechanism by which mutated KCNJ5 induces cell proliferation and adenoma formation remains unclear.
Collapse
Affiliation(s)
- Carolina Velarde-Miranda
- Research and Endocrine Service, GV (Sonny) Montgomery VA Medical Center, University of Mississippi Medical Center, Jackson, MS, USA; Division of Endocrinology, University of Mississippi Medical Center, Jackson, MS, USA
| | | | | |
Collapse
|
38
|
Lenzini L, Caroccia B, Campos AG, Fassina A, Belloni AS, Seccia TM, Kuppusamy M, Ferraro S, Skander G, Bader M, Rainey WE, Rossi GP. Lower expression of the TWIK-related acid-sensitive K+ channel 2 (TASK-2) gene is a hallmark of aldosterone-producing adenoma causing human primary aldosteronism. J Clin Endocrinol Metab 2014; 99:E674-82. [PMID: 24285684 DOI: 10.1210/jc.2013-2900] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/13/2023]
Abstract
CONTEXT The molecular mechanisms of primary aldosteronism, a common cause of human hypertension, are unknown, but alterations of K(+) channels can play a key role. OBJECTIVE The objective of the study was to investigate the following: 1) the expression of the Twik-related acid-sensitive K(+) channels (TASK) in aldosterone producing adenomas (APAs); 2) the role of TASK-2 in aldosterone synthesis; and 3) the determinants of TASK-2-blunted expression in APAs. DESIGN We analyzed the transcriptome and the microRNA profiles of 32 consecutive APAs and investigated the protein expression and localization of TASK-2 in APA and adrenocortical cell lines (H295R and HAC15) using immunoblotting and confocal microscopy. The functional effect of TASK-2 blunted activity caused by a dominant-negative mutation on steroidogenic enzymes, and aldosterone production was also assessed. TASK-2 regulation by selected microRNA was studied by a luciferase assay. RESULTS TASK-2 was consistently less expressed at the transcript and protein levels in APAs than in the normal human adrenal cortex. H295R cell transfection with a TASK-2 dominant-negative mutant construct significantly increased the aldosterone production by 153% and the gene expression of aldosterone synthase (CYP11B2, gene expression fold change 3.1 vs control, P < .05) and the steroidogenic acute regulatory protein (gene expression fold change 1.8 vs control, P < .05). Two microRNAs, hsa-miR-23 and hsa-miR-34, were found to decrease the TASK-2 expression by binding to the 3' untranslated region of the TASK-2 gene. CONCLUSIONS The TASK-2 channel lower expression represents a hallmark of APA and is associated with a higher expression of hsa-miR-23 and hsa-miR-34. The ensuing blunted TASK-2 activity increased the production of aldosterone in vitro and the expression of steroidogenic acute regulatory protein and CYP11B2. Hence, the lower expression of TASK-2 channel in APA cells can explain high aldosterone secretion in human primary aldosteronism despite the suppression of angiotensin II, hypertension, and hypokalemia.
Collapse
Affiliation(s)
- Livia Lenzini
- Internal Medicine 4 (L.L., B.C., A.G.C., T.M.S., M.K., S.F., G.S., G.P.R.) and Surgical Pathology and Cytopathology Unit (A.F.), Department of Medicine, Department of Medicine, Section of Human Anatomy and Physiology (A.S.B.), Department of Molecular Medicine, University of Padua, 35128 Padua, Italy; Max-Delbrück-Center for Molecular Medicine (M.B.), 13092 Berlin, Germany; and Department of Molecular and Integrative Physiology (W.E.R.), University of Michigan, Ann Arbor, Michigan 48109
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
39
|
Tauber P, Penton D, Stindl J, Humberg E, Tegtmeier I, Sterner C, Beuschlein F, Reincke M, Barhanin J, Bandulik S, Warth R. Pharmacology and pathophysiology of mutated KCNJ5 found in adrenal aldosterone-producing adenomas. Endocrinology 2014; 155:1353-62. [PMID: 24506072 DOI: 10.1210/en.2013-1944] [Citation(s) in RCA: 49] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Somatic mutations of the potassium channel KCNJ5 are found in 40% of aldosterone producing adenomas (APAs). APA-related mutations of KCNJ5 lead to a pathological Na(+) permeability and a rise in cytosolic Ca(2+), the latter presumably by depolarizing the membrane and activating voltage-gated Ca(2+) channels. The aim of this study was to further investigate the effects of mutated KCNJ5 channels on intracellular Na(+) and Ca(2+) homeostasis in human adrenocortical NCI-H295R cells. Expression of mutant KCNJ5 led to a 2-fold increase in intracellular Na(+) and, in parallel, to a substantial rise in intracellular Ca(2+). The increase in Ca(2+) appeared to be caused by activation of voltage-gated Ca(2+) channels and by an impairment of Ca(2+) extrusion by Na(+)/Ca(2+) exchangers. The mutated KCNJ5 exhibited a pharmacological profile that differed from the one of wild-type channels. Mutated KCNJ5 was less Ba(2+) and tertiapin-Q sensitive but was inhibited by blockers of Na(+) and Ca(2+)-transporting proteins, such as verapamil and amiloride. The clinical use of these drugs might influence aldosterone levels in APA patients with KCNJ5 mutations. This might implicate diagnostic testing of APAs and could offer new therapeutic strategies.
Collapse
Affiliation(s)
- P Tauber
- Medical Cell Biology (P.T., J.S., E.H., I.T., C.S., S.B., R.W.), University of Regensburg, 93053 Regensburg, Germany; Laboratoire de PhysioMédecine Moléculaire (D.P., J.B.), Centre National de la Recherche Scientifique, and Université de Nice Sophia Antipolis, FRE3472-Laboratoire de PhysioMédecine Moléculaire, 06108 Nice Cedex, France; Laboratories of Excellence, Ion Channel Science and Therapeutics (D.P., J.B.), France; and Medizinische Klinik und Poliklinik IV (F.B., M.R.), Ludwig-Maximilians-Universität, 80336 Munich, Germany
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
40
|
Affiliation(s)
- Celso E Gomez-Sanchez
- Division of Endocrinology, University of Mississippi Medical Center, 2500 N State St, Jackson, MS 39216.
| |
Collapse
|
41
|
Gomez-Sanchez CE, Oki K. Minireview: potassium channels and aldosterone dysregulation: is primary aldosteronism a potassium channelopathy? Endocrinology 2014; 155:47-55. [PMID: 24248457 PMCID: PMC5398635 DOI: 10.1210/en.2013-1733] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/18/2023]
Abstract
Primary aldosteronism is the most common form of secondary hypertension and has significant cardiovascular consequences. Aldosterone-producing adenomas (APAs) are responsible for half the cases of primary aldosteronism, and about half have mutations of the G protein-activated inward rectifying potassium channel Kir3.4. Under basal conditions, the adrenal zona glomerulosa cells are hyperpolarized with negative resting potentials determined by membrane permeability to K(+) mediated through various K(+) channels, including the leak K(+) channels TASK-1, TASK-3, and Twik-Related Potassium Channel 1, and G protein inward rectifying potassium channel Kir3.4. Angiotensin II decreases the activity of the leak K(+) channels and Kir3.4 channel and decreases the expression of the Kir3.4 channel, resulting in membrane depolarization, increased intracellular calcium, calcium-calmodulin pathway activation, and increased expression of cytochrome P450 aldosterone synthase (CYP11B2), the last enzyme for aldosterone production. Somatic mutations of the selectivity filter of the Kir3.4 channel in APA results in loss of selectivity for K(+) and entry of sodium, resulting in membrane depolarization, calcium mobilization, increased CYP11B2 expression, and hyperaldosteronism. Germ cell mutations cause familial hyperaldosteronism type 3, which is associated with adrenal zona glomerulosa hyperplasia, rather than adenoma. Less commonly, somatic mutations of the sodium-potassium ATPase, calcium ATPase, or the calcium channel calcium channel voltage-dependent L type alpha 1D have been found in some APAs. The regulation of aldosterone secretion is exerted to a significant degree by activation of membrane K(+) and calcium channels or pumps, so it is not surprising that the known causes of disorders of aldosterone secretion in APA have been channelopathies, which activate mechanisms that increase aldosterone synthesis.
Collapse
Affiliation(s)
- Celso E Gomez-Sanchez
- Endocrinology Division (C.E.G.-S.), G. V. (Sonny) Montgomery Veterans Affairs Medical Center and University of Mississippi Medical Center, Jackson, Mississippi 39216; and Department of Endocrinology and Diabetes (K.O.), Hiroshima University Hospital, Hiroshima 734-8551, Japan
| | | |
Collapse
|
42
|
Berthon A, Drelon C, Ragazzon B, Boulkroun S, Tissier F, Amar L, Samson-Couterie B, Zennaro MC, Plouin PF, Skah S, Plateroti M, Lefèbvre H, Sahut-Barnola I, Batisse-Lignier M, Assié G, Lefrançois-Martinez AM, Bertherat J, Martinez A, Val P. WNT/β-catenin signalling is activated in aldosterone-producing adenomas and controls aldosterone production. Hum Mol Genet 2013; 23:889-905. [PMID: 24087794 DOI: 10.1093/hmg/ddt484] [Citation(s) in RCA: 142] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Primary aldosteronism (PA) is the main cause of secondary hypertension, resulting from adrenal aldosterone-producing adenomas (APA) or bilateral hyperplasia. Here, we show that constitutive activation of WNT/β-catenin signalling is the most frequent molecular alteration found in 70% of APA. We provide evidence that decreased expression of the WNT inhibitor SFRP2 may be contributing to deregulated WNT signalling and APA development in patients. This is supported by the demonstration that mice with genetic ablation of Sfrp2 have increased aldosterone production and ectopic differentiation of zona glomerulosa cells. We further show that β-catenin plays an essential role in the control of basal and Angiotensin II-induced aldosterone secretion, by activating AT1R, CYP21 and CYP11B2 transcription. This relies on both LEF/TCF-dependent activation of AT1R and CYP21 regulatory regions and indirect activation of CYP21 and CYP11B2 promoters, through increased expression of the nuclear receptors NURR1 and NUR77. Altogether, these data show that aberrant WNT/β-catenin activation is associated with APA development and suggest that WNT pathway may be a good therapeutic target in PA.
Collapse
Affiliation(s)
- Annabel Berthon
- Clermont Université, Université Blaise Pascal, GReD, BP 10448, F-63000 Clermont-Ferrand, France
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
43
|
New insights into aldosterone-producing adenomas and hereditary aldosteronism. Curr Opin Nephrol Hypertens 2013; 22:141-7. [DOI: 10.1097/mnh.0b013e32835cecf8] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
|
44
|
Mulatero P, Monticone S, Rainey WE, Veglio F, Williams TA. Role of KCNJ5 in familial and sporadic primary aldosteronism. Nat Rev Endocrinol 2013; 9:104-12. [PMID: 23229280 DOI: 10.1038/nrendo.2012.230] [Citation(s) in RCA: 80] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Primary aldosteronism is characterised by the dysregulation of aldosterone production and comprises both sporadic forms, caused by an aldosterone-producing adenoma or bilateral adrenal hyperplasia, and familial forms (familial hyperaldosteronism types I, II and III). The two principal physiological regulators of aldosterone synthesis are angiotensin II and serum K(+), which reverse the high resting K(+) conductance and hyperpolarized membrane potential of adrenal glomerulosa cells. The resulting membrane depolarization causes the opening of voltage-gated Ca(2+) channels and an increase in intracellular Ca(2+) that stimulates aldosterone biosynthesis. Point mutations in the KCNJ5 gene, which encodes the G-protein-activated inward rectifier K(+) channel 4 (GIRK4), have been implicated in the pathogenesis of both sporadic and familial forms of primary aldosteronism. These mutations interfere with the selectivity filter of GIRK4 causing Na(+) entry, cell depolarization and Ca(2+) channel opening, resulting in constitutive aldosterone production. Seven families with familial hyperaldosteronism caused by KCNJ5 germline mutations have so far been described, and multicentre studies have reported KCNJ5 mutations in approximately 40% of sporadic aldosterone-producing adenomas. Herein, we review the role of GIRK4 in adrenal pathophysiology and provide an overview of the clinical and biochemical phenotypes resulting from KCNJ5 mutations in patients with sporadic and familial primary aldosteronism.
Collapse
Affiliation(s)
- Paolo Mulatero
- University of Torino, Department of Medical Sciences, Division of Internal Medicine and Hypertension, Italy. paolo.mulatero@ unito.it
| | | | | | | | | |
Collapse
|