1
|
Ruffenach G, Le Ribeuz H, Dutheil M, El Jekmek K, Dumont F, Willer AS, Humbert M, Capuano V, Medzikovic L, Eghbali M, Montani D, Antigny F. Transcriptome analyses reveal common immune system dysregulation in PAH patients and Kcnk3-deficient rats. Pulm Circ 2024; 14:e12434. [PMID: 39444497 PMCID: PMC11497494 DOI: 10.1002/pul2.12434] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/15/2024] [Revised: 07/26/2024] [Accepted: 08/15/2024] [Indexed: 10/25/2024] Open
Abstract
Pulmonary arterial hypertension (PAH) is a severe disease caused by progressive distal pulmonary artery obstruction. One cause of PAH are loss-of-function mutations in the potassium channel subfamily K member 3 (KCNK3). KCNK3 encodes a two-pore domain potassium channel, which is crucial for pulmonary circulation homeostasis. However, our understanding of the pathophysiological mechanisms underlying KCNK3 dysfunction in PAH is still incomplete. Taking advantage of unique Kcnk3-deficient rats, we analyzed the transcriptomic changes in the lungs from homozygous Kcnk3-deficient rats and wild-type (WT) littermates and compared them to PAH patient transcriptomic data. Transcriptome analysis of lung tissue obtained from WT and Kcnk3-deficient rats identified 1915 down- or upregulated genes. In addition, despite limited similarities at the gene level, we found a strong common signature at the pathway level in PAH patients and Kcnk3-deficient rat lungs, especially for immune response. Using the dysregulated genes involved in the immune response, we identified Spleen Associated Tyrosine Kinase (SYK), a significantly downregulated gene in human PAH patients and Kcnk3-deficient rats, as a hub gene. Our data suggests that the altered immune system response observed in PAH patients may be partly explained by KCNK3 dysfunction through the alteration of SYK expression.
Collapse
Affiliation(s)
- Grégoire Ruffenach
- Department of Anesthesiology and Perioperative Medicine, Division of Molecular Medicine, David Geffen School of MedicineUniversity of CaliforniaLos AngelesUSA
| | - Hélène Le Ribeuz
- Faculté de Médecine, Le Kremlin‐BicêtreUniversité Paris‐SaclayLe Kremlin‐BicêtreFrance
- INSERM UMR_S 999 “Hypertension Pulmonaire: Physiopathologie et Innovation Thérapeutique”, Hôpital Marie LannelongueLe Plessis‐RobinsonFrance
| | - Mary Dutheil
- Faculté de Médecine, Le Kremlin‐BicêtreUniversité Paris‐SaclayLe Kremlin‐BicêtreFrance
- INSERM UMR_S 999 “Hypertension Pulmonaire: Physiopathologie et Innovation Thérapeutique”, Hôpital Marie LannelongueLe Plessis‐RobinsonFrance
- Groupe Hospitalier Paris Saint‐Joseph, Hôpital Marie LannelongueLe Plessis‐RobinsonFrance
| | - Kristell El Jekmek
- Faculté de Médecine, Le Kremlin‐BicêtreUniversité Paris‐SaclayLe Kremlin‐BicêtreFrance
- INSERM UMR_S 999 “Hypertension Pulmonaire: Physiopathologie et Innovation Thérapeutique”, Hôpital Marie LannelongueLe Plessis‐RobinsonFrance
| | - Florent Dumont
- Faculté de Médecine, Le Kremlin‐BicêtreUniversité Paris‐SaclayLe Kremlin‐BicêtreFrance
- UMS Ingénierie et Plateformes au Service de l'Innovation ThérapeutiqueUniversité Paris‐SaclayOrsayFrance
| | - Anaïs Saint‐Martin Willer
- Faculté de Médecine, Le Kremlin‐BicêtreUniversité Paris‐SaclayLe Kremlin‐BicêtreFrance
- INSERM UMR_S 999 “Hypertension Pulmonaire: Physiopathologie et Innovation Thérapeutique”, Hôpital Marie LannelongueLe Plessis‐RobinsonFrance
| | - Marc Humbert
- Faculté de Médecine, Le Kremlin‐BicêtreUniversité Paris‐SaclayLe Kremlin‐BicêtreFrance
- INSERM UMR_S 999 “Hypertension Pulmonaire: Physiopathologie et Innovation Thérapeutique”, Hôpital Marie LannelongueLe Plessis‐RobinsonFrance
- Assistance Publique—Hôpitaux de Paris (AP‐HP), Service de Pneumologie et Soins Intensifs Respiratoires, Centre de Référence de l'Hypertension Pulmonaire, Hôpital Bicêtre, Le Kremlin‐BicêtreLe Kremlin‐BicêtreFrance
| | - Véronique Capuano
- Faculté de Médecine, Le Kremlin‐BicêtreUniversité Paris‐SaclayLe Kremlin‐BicêtreFrance
- INSERM UMR_S 999 “Hypertension Pulmonaire: Physiopathologie et Innovation Thérapeutique”, Hôpital Marie LannelongueLe Plessis‐RobinsonFrance
- Groupe Hospitalier Paris Saint‐Joseph, Hôpital Marie LannelongueLe Plessis‐RobinsonFrance
| | - Lejla Medzikovic
- Department of Anesthesiology and Perioperative Medicine, Division of Molecular Medicine, David Geffen School of MedicineUniversity of CaliforniaLos AngelesUSA
| | - Mansoureh Eghbali
- Department of Anesthesiology and Perioperative Medicine, Division of Molecular Medicine, David Geffen School of MedicineUniversity of CaliforniaLos AngelesUSA
| | - David Montani
- Faculté de Médecine, Le Kremlin‐BicêtreUniversité Paris‐SaclayLe Kremlin‐BicêtreFrance
- INSERM UMR_S 999 “Hypertension Pulmonaire: Physiopathologie et Innovation Thérapeutique”, Hôpital Marie LannelongueLe Plessis‐RobinsonFrance
- Groupe Hospitalier Paris Saint‐Joseph, Hôpital Marie LannelongueLe Plessis‐RobinsonFrance
- Assistance Publique—Hôpitaux de Paris (AP‐HP), Service de Pneumologie et Soins Intensifs Respiratoires, Centre de Référence de l'Hypertension Pulmonaire, Hôpital Bicêtre, Le Kremlin‐BicêtreLe Kremlin‐BicêtreFrance
| | - Fabrice Antigny
- Faculté de Médecine, Le Kremlin‐BicêtreUniversité Paris‐SaclayLe Kremlin‐BicêtreFrance
- INSERM UMR_S 999 “Hypertension Pulmonaire: Physiopathologie et Innovation Thérapeutique”, Hôpital Marie LannelongueLe Plessis‐RobinsonFrance
| |
Collapse
|
2
|
van Rooyen D, Lerario AM, Little DW, Ullenbruch MR, Taylor MJ, Gomez-Sanchez CE, Hammer GD, Rainey WE. Chronic activation of adrenal Gq signaling induces Cyp11b2 expression in the zona fasciculata and hyperaldosteronism. Mol Cell Endocrinol 2024; 585:112176. [PMID: 38341019 PMCID: PMC11907336 DOI: 10.1016/j.mce.2024.112176] [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: 09/02/2023] [Revised: 01/31/2024] [Accepted: 02/01/2024] [Indexed: 02/12/2024]
Abstract
Hyperaldosteronism is often associated with inappropriate aldosterone production and aldosterone synthase (Cyp11b2) expression. Normally, Cyp11b2 expression is limited to the adrenal zona glomerulosa (ZG) and regulated by angiotensin II which signals through Gq protein-coupled receptors. As cells migrate inwards, they differentiate into 11β-hydroxylase-expressing zona fasciculata (ZF) cells lacking Cyp11b2. The mechanism causing ZG-specific aldosterone biosynthesis is still unclear. We investigated the effect of chronic Gq signaling using transgenic mice with a clozapine N-oxide (CNO)-activated human M3 muscarinic receptor (DREADD) coupled to Gq (hM3Dq) that was expressed throughout the adrenal cortex. CNO raised circulating aldosterone in the presence of a high sodium diet with greater response seen in females compared to males. Immunohistochemistry and transcriptomics indicated disrupted zonal Cyp11b2 expression while Wnt signaling remained unchanged. Chronic Gq-DREADD signaling also induced an intra-adrenal RAAS in CNO-treated mice. Chronic Gq signaling disrupted adrenal cortex zonal aldosterone production associated with ZF expression of Cyp11b2.
Collapse
Affiliation(s)
- Desmaré van Rooyen
- Department of Molecular and Integrative Physiology, University of Michigan, Ann Arbor, MI, USA; Division of Metabolism, Endocrinology, and Diabetes, Department of Internal Medicine, University of Michigan, Ann Arbor, MI, USA
| | - Antonio M Lerario
- Division of Metabolism, Endocrinology, and Diabetes, Department of Internal Medicine, University of Michigan, Ann Arbor, MI, USA
| | - Donald W Little
- Division of Metabolism, Endocrinology, and Diabetes, Department of Internal Medicine, University of Michigan, Ann Arbor, MI, USA
| | - Matthew R Ullenbruch
- Department of Molecular and Integrative Physiology, University of Michigan, Ann Arbor, MI, USA
| | - Matthew J Taylor
- Department of Molecular and Integrative Physiology, University of Michigan, Ann Arbor, MI, USA
| | - Celso E Gomez-Sanchez
- Endocrine Section, G.V. (Sonny) Montgomery VA Medical Center and the Department of Pharmacology and Toxicology, University of Mississippi Medical Center, Jackson, MS, USA
| | - Gary D Hammer
- Department of Molecular and Integrative Physiology, University of Michigan, Ann Arbor, MI, USA; Division of Metabolism, Endocrinology, and Diabetes, Department of Internal Medicine, University of Michigan, Ann Arbor, MI, USA; Department of Cell and Development Biology, University of Michigan, Ann Arbor, MI, USA
| | - William E Rainey
- Department of Molecular and Integrative Physiology, University of Michigan, Ann Arbor, MI, USA; Division of Metabolism, Endocrinology, and Diabetes, Department of Internal Medicine, University of Michigan, Ann Arbor, MI, USA.
| |
Collapse
|
3
|
Dinh HA, Volkert M, Secener AK, Scholl UI, Stölting G. T- and L-Type Calcium Channels Maintain Calcium Oscillations in the Murine Zona Glomerulosa. Hypertension 2024; 81:811-822. [PMID: 38507511 PMCID: PMC10956685 DOI: 10.1161/hypertensionaha.123.21798] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2023] [Accepted: 01/31/2024] [Indexed: 03/22/2024]
Abstract
BACKGROUND The zona glomerulosa of the adrenal gland is responsible for the synthesis and release of the mineralocorticoid aldosterone. This steroid hormone regulates salt reabsorption in the kidney and blood pressure. The most important stimuli of aldosterone synthesis are the serum concentrations of angiotensin II and potassium. In response to these stimuli, voltage and intracellular calcium levels in the zona glomerulosa oscillate, providing the signal for aldosterone synthesis. It was proposed that the voltage-gated T-type calcium channel CaV3.2 is necessary for the generation of these oscillations. However, Cacna1h knock-out mice have normal plasma aldosterone levels, suggesting additional calcium entry pathways. METHODS We used a combination of calcium imaging, patch clamp, and RNA sequencing to investigate calcium influx pathways in the murine zona glomerulosa. RESULTS Cacna1h-/- glomerulosa cells still showed calcium oscillations with similar concentrations as wild-type mice. No calcium channels or transporters were upregulated to compensate for the loss of CaV3.2. The calcium oscillations observed were instead dependent on L-type voltage-gated calcium channels. Furthermore, we found that L-type channels can also partially compensate for an acute inhibition of CaV3.2 in wild-type mice. Only inhibition of both T- and L-type calcium channels abolished the increase of intracellular calcium caused by angiotensin II in wild-type. CONCLUSIONS Our study demonstrates that T-type calcium channels are not strictly required to maintain glomerulosa calcium oscillations and aldosterone production. Pharmacological inhibition of T-type channels alone will likely not significantly impact aldosterone production in the long term.
Collapse
Affiliation(s)
- Hoang An Dinh
- Berlin Institute of Health at Charité – Universitätsmedizin Berlin, Center of Functional Genomics, Germany (H.A.D., M.V., A.K.S., U.I.S., G.S.)
- Charité – Universitätsmedizin Berlin, Department of Translational Physiology, Germany (H.A.D.)
| | - Marina Volkert
- Berlin Institute of Health at Charité – Universitätsmedizin Berlin, Center of Functional Genomics, Germany (H.A.D., M.V., A.K.S., U.I.S., G.S.)
| | - Ali Kerim Secener
- Berlin Institute of Health at Charité – Universitätsmedizin Berlin, Center of Functional Genomics, Germany (H.A.D., M.V., A.K.S., U.I.S., G.S.)
- Genomics Technology Platform, Max Delbrück Center for Molecular Medicine in the Helmholtz Association (MDC), Berlin, Germany (A.K.S.)
- Institute of Chemistry and Biochemistry, Department of Biology, Chemistry and Pharmacy, Freie Universität Berlin, Germany (A.K.S.)
| | - Ute I. Scholl
- Berlin Institute of Health at Charité – Universitätsmedizin Berlin, Center of Functional Genomics, Germany (H.A.D., M.V., A.K.S., U.I.S., G.S.)
- Charité – Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Department of Nephrology and Medical Intensive Care, Berlin, Germany (U.I.S.)
| | - Gabriel Stölting
- Berlin Institute of Health at Charité – Universitätsmedizin Berlin, Center of Functional Genomics, Germany (H.A.D., M.V., A.K.S., U.I.S., G.S.)
| |
Collapse
|
4
|
Yegen CH, Lambert M, Beurnier A, Montani D, Humbert M, Planès C, Boncoeur E, Voituron N, Antigny F. KCNK3 channel is important for the ventilatory response to hypoxia in rats. Respir Physiol Neurobiol 2023; 318:104164. [PMID: 37739151 DOI: 10.1016/j.resp.2023.104164] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2023] [Revised: 08/30/2023] [Accepted: 09/17/2023] [Indexed: 09/24/2023]
Abstract
To clarify the contribution of KCNK3/TASK-1 channel chemoreflex in response to hypoxia and hypercapnia, we used a unique Kcnk3-deficient rat. We assessed ventilatory variables using plethysmography in Kcnk3-deficient and wild-type rats at rest in response to hypoxia (10% O2) and hypercapnia (4% CO2). Immunostaining for C-Fos, a marker of neuronal activity, was performed to identify the regions of the respiratory neuronal network involved in the observed response.Under basal conditions, we observed increased minute ventilation in Kcnk3-deficient rats, which was associated with increased c-Fos positive cells in the ventrolateral region of the medulla oblongata. Kcnk3-deficient rats show an increase in ventilatory response to hypoxia without changes in response to hypercapnia. In Kcnk3-deficient rats, linked to an increased hypoxia response, we observed a greater increase in c-Fos-positive cells in the first central relay of peripheral chemoreceptors and Raphe Obscurus. This study reports that KCNK3/TASK-1 deficiency in rats induces an inadequate peripheral chemoreflex, alternating respiratory rhythmogenesis, and hypoxic chemoreflex.
Collapse
Affiliation(s)
- Céline-Hivda Yegen
- Laboratoire Hypoxie & Poumon, UMR INSERM U1272, Université Sorbonne Paris Nord, Bobigny, France
| | - Mélanie Lambert
- Université Paris-Saclay, Faculté de Médecine, Le Kremlin-Bicêtre, France
| | - Antoine Beurnier
- Université Paris-Saclay, Faculté de Médecine, Le Kremlin-Bicêtre, France; INSERM UMR_S 999 " Hypertension pulmonaire: Physiopathologie et Innovation Thérapeutique ", Hôpital Marie Lannelongue, Le Plessis-Robinson, France; Service de Physiologie et d'explorations fonctionnelles, Hôpital Avicenne, APHP, Hôpitaux de Paris, France
| | - David Montani
- Université Paris-Saclay, Faculté de Médecine, Le Kremlin-Bicêtre, France; INSERM UMR_S 999 " Hypertension pulmonaire: Physiopathologie et Innovation Thérapeutique ", Hôpital Marie Lannelongue, Le Plessis-Robinson, France; Assistance Publique - Hôpitaux de Paris (AP-HP), Service de Pneumologie et Soins Intensifs Respiratoires, Centre de Référence de l'Hypertension Pulmonaire, Hôpital Bicêtre, Le Kremlin-Bicêtre, France
| | - Marc Humbert
- Université Paris-Saclay, Faculté de Médecine, Le Kremlin-Bicêtre, France; INSERM UMR_S 999 " Hypertension pulmonaire: Physiopathologie et Innovation Thérapeutique ", Hôpital Marie Lannelongue, Le Plessis-Robinson, France; Assistance Publique - Hôpitaux de Paris (AP-HP), Service de Pneumologie et Soins Intensifs Respiratoires, Centre de Référence de l'Hypertension Pulmonaire, Hôpital Bicêtre, Le Kremlin-Bicêtre, France
| | - Carole Planès
- Laboratoire Hypoxie & Poumon, UMR INSERM U1272, Université Sorbonne Paris Nord, Bobigny, France; AP-HP, Department of Physiology - Functional Explorations, DMU Thorinno, bi-site Hôpital Bicêtre (Le Kremlin Bicêtre) and Ambroise Paré (Boulogne-Billancourt), France
| | - Emilie Boncoeur
- Laboratoire Hypoxie & Poumon, UMR INSERM U1272, Université Sorbonne Paris Nord, Bobigny, France
| | - Nicolas Voituron
- Laboratoire Hypoxie & Poumon, UMR INSERM U1272, Université Sorbonne Paris Nord, Bobigny, France; Département STAPS, Université Sorbonne Paris Nord, Bobigny, France.
| | - Fabrice Antigny
- Université Paris-Saclay, Faculté de Médecine, Le Kremlin-Bicêtre, France.
| |
Collapse
|
5
|
Saint-Martin Willer A, Santos-Gomes J, Adão R, Brás-Silva C, Eyries M, Pérez-Vizcaino F, Capuano V, Montani D, Antigny F. Physiological and pathophysiological roles of the KCNK3 potassium channel in the pulmonary circulation and the heart. J Physiol 2023; 601:3717-3737. [PMID: 37477289 DOI: 10.1113/jp284936] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2023] [Accepted: 07/04/2023] [Indexed: 07/22/2023] Open
Abstract
Potassium channel subfamily K member 3 (KCNK3), encoded by the KCNK3 gene, is part of the two-pore domain potassium channel family, constitutively active at resting membrane potentials in excitable cells, including smooth muscle and cardiac cells. Several physiological and pharmacological mediators, such as intracellular signalling pathways, extracellular pH, hypoxia and anaesthetics, regulate KCNK3 channel function. Recent studies show that modulation of KCNK3 channel expression and function strongly influences pulmonary vascular cell and cardiomyocyte function. The altered activity of KCNK3 in pathological situations such as atrial fibrillation, pulmonary arterial hypertension and right ventricular dysfunction demonstrates the crucial role of KCNK3 in cardiovascular homeostasis. Furthermore, loss of function variants of KCNK3 have been identified in patients suffering from pulmonary arterial hypertension and atrial fibrillation. This review focuses on current knowledge of the role of the KCNK3 channel in pulmonary circulation and the heart, in healthy and pathological conditions.
Collapse
Affiliation(s)
- Anaïs Saint-Martin Willer
- Université Paris-Saclay, Faculté de Médecine, Le Kremlin-Bicêtre, France
- INSERM UMR_S 999 'Hypertension Pulmonaire: Physiopathologie et Innovation Thérapeutique', Hôpital Marie Lannelongue, Le Plessis-Robinson, France
| | - Joana Santos-Gomes
- Cardiovascular R&D Centre-UnIC@RISE, Department of Surgery and Physiology, Faculty of Medicine of the University of Porto, Porto, Portugal
| | - Rui Adão
- Cardiovascular R&D Centre-UnIC@RISE, Department of Surgery and Physiology, Faculty of Medicine of the University of Porto, Porto, Portugal
- Department of Pharmacology and Toxicology, School of Medicine, Universidad Complutense de Madrid, Madrid, Spain
- Instituto de Investigación Sanitaria Gregorio Marañón (IISGM), Madrid, Spain
- CIBER Enfermedades Respiratorias (Ciberes), Madrid, Spain
| | - Carmen Brás-Silva
- Cardiovascular R&D Centre-UnIC@RISE, Department of Surgery and Physiology, Faculty of Medicine of the University of Porto, Porto, Portugal
| | - Mélanie Eyries
- Département de génétique, Assistance Publique-Hôpitaux de Paris, Hôpital Pitié-Salpêtrière, Sorbonne Université, Paris, France
- INSERM UMRS1166, ICAN - Institute of CardioMetabolism and Nutrition, Sorbonne Université, Paris, France
| | - Francisco Pérez-Vizcaino
- Department of Pharmacology and Toxicology, School of Medicine, Universidad Complutense de Madrid, Madrid, Spain
- Instituto de Investigación Sanitaria Gregorio Marañón (IISGM), Madrid, Spain
- CIBER Enfermedades Respiratorias (Ciberes), Madrid, Spain
| | - Véronique Capuano
- Université Paris-Saclay, Faculté de Médecine, Le Kremlin-Bicêtre, France
- INSERM UMR_S 999 'Hypertension Pulmonaire: Physiopathologie et Innovation Thérapeutique', Hôpital Marie Lannelongue, Le Plessis-Robinson, France
| | - David Montani
- Université Paris-Saclay, Faculté de Médecine, Le Kremlin-Bicêtre, France
- INSERM UMR_S 999 'Hypertension Pulmonaire: Physiopathologie et Innovation Thérapeutique', Hôpital Marie Lannelongue, Le Plessis-Robinson, France
- Assistance Publique - Hôpitaux de Paris (AP-HP), Service de Pneumologie et Soins Intensifs Respiratoires, Centre de Référence de l'Hypertension Pulmonaire, Hôpital Bicêtre, Le Kremlin-Bicêtre, France
| | - Fabrice Antigny
- Université Paris-Saclay, Faculté de Médecine, Le Kremlin-Bicêtre, France
- INSERM UMR_S 999 'Hypertension Pulmonaire: Physiopathologie et Innovation Thérapeutique', Hôpital Marie Lannelongue, Le Plessis-Robinson, France
| |
Collapse
|
6
|
Kim D, Harada K, Inoue M. Expression and function of mitochondrial inhibitor factor-1 and TASK channels in adrenal cells. Biochem Biophys Res Commun 2023; 645:17-23. [PMID: 36657294 PMCID: PMC9900489 DOI: 10.1016/j.bbrc.2023.01.025] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2022] [Accepted: 01/10/2023] [Indexed: 01/13/2023]
Abstract
Adrenal medullary chromaffin (AMC) cells in the perinatal period and carotid body glomus cells after birth respond to hypoxia with catecholamine secretion. The hypoxia detection mechanism in such O2-sensitive cells is still not well defined. One hypothesis is that a decrease in cellular ATP may be involved in the hypoxia detection. This idea is based on ATP dependence of TASK channel activity that regulates the resting membrane potential and is suppressed by hypoxia in glomus cells. Mitochondrial ATPase inhibitor factor-1 (IF1), a physiological regulator of ATP synthase, helps prevent ATP hydrolysis under hypoxic conditions. In cells where IF1 expression is high, exposure to hypoxia is expected to have no effect on TASK channel activity. This possibility was electrophysiologically and immunocytochemically explored. Single channel recordings revealed that 36-pS TASK3-like channels contribute to the resting membrane potential in young rat adrenal cortical (AC) cells. TASK3-like channel activity in a cell-attached patch was not affected by bath application of mitochondrial inhibitors. Consistent with this finding, IF1-like immunoreactive material was well expressed in rat AC cells. In further support of our hypothesis, IF1-like immunoreactive material was well expressed in adult rat AMC cells that are known to be hypoxia-insensitive and minimally expressed in newborn AMC cells that are hypoxia-sensitive. These results provide evidence for the functional relevance of IF1 expression in excitability in O2-sensitive cells in response to mitochondrial inhibition.
Collapse
Affiliation(s)
- Donghee Kim
- Department of Physiology and Biophysics, Chicago Medical School, Rosalind Franklin University of Medicine and Science, North Chicago, IL, 60064-3095, USA
| | - Keita Harada
- Department of Cell and Systems Physiology, University of Occupational and Environmental Health, School of Medicine, Kitakyushu, 807-8555, Japan
| | - Masumi Inoue
- Department of Cell and Systems Physiology, University of Occupational and Environmental Health, School of Medicine, Kitakyushu, 807-8555, Japan.
| |
Collapse
|
7
|
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
|
8
|
Gancayco CA, Gerding MR, Breault DT, Beenhakker MP, Barrett PQ, Guagliardo NA. Intrinsic Adrenal TWIK-Related Acid-Sensitive TASK Channel Dysfunction Produces Spontaneous Calcium Oscillations Sufficient to Drive AngII (Angiotensin II)-Unresponsive Hyperaldosteronism. Hypertension 2022; 79:2552-2564. [PMID: 36129175 PMCID: PMC10167771 DOI: 10.1161/hypertensionaha.122.19557] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Background:
Ion channel mutations in calcium regulating genes strongly associate with AngII (angiotensin II)-independent aldosterone production. Here, we used an established mouse model of in vivo aldosterone autonomy,
Cyp11b2
-driven deletion of TWIK-related acid-sensitive potassium channels (TASK-1 and TASK-3, termed zona glomerulosa [zG]-TASK-loss-of-function), and selective pharmacological TASK channel inhibition to determine whether channel dysfunction in native, electrically excitable zG cell rosette-assemblies: (1) produces spontaneous calcium oscillatory activity and (2) is sufficient to drive substantial aldosterone autonomy.
Methods:
We imaged calcium activity in adrenal slices expressing a zG-specific calcium reporter (GCaMP3), an in vitro experimental approach that preserves the native rosette assembly and removes potentially confounding extra-adrenal contributions. In parallel experiments, we measured acute aldosterone production from adrenal slice cultures.
Results:
Absent from untreated WT slices, we find that either adrenal-specific genetic deletion or acute pharmacological TASK channel inhibition produces spontaneous oscillatory bursting behavior and steroidogenic activity (2.4-fold) that are robust, sustained, and equivalent to activities evoked by 3 nM AngII in WT slices. Moreover, spontaneous activity in zG-TASK-loss-of-function slices and inhibitor-evoked activity in WT slices are unresponsive to AngII regulation over a wide range of concentrations (50 pM to 3 µM).
Conclusions:
We provide proof of principle that spontaneous activity of zG cells within classic rosette assemblies evoked solely by a change in an intrinsic, dominant resting-state conductance can be a significant source of AngII-independent aldosterone production from native tissue.
Collapse
Affiliation(s)
| | - Molly R. Gerding
- Department of Pharmacology (M.R.G., M.P.B., P.Q.B., N.A.G.), University of Virginia, Charlottesville
| | - David T. Breault
- Division of Endocrinology, Boston Children’s Hospital, MA (D.T.B.)
- Harvard Stem Cell Institute, Cambridge, MA (D.T.B.)
| | - Mark P. Beenhakker
- Department of Pharmacology (M.R.G., M.P.B., P.Q.B., N.A.G.), University of Virginia, Charlottesville
| | - Paula Q. Barrett
- Department of Pharmacology (M.R.G., M.P.B., P.Q.B., N.A.G.), University of Virginia, Charlottesville
| | - Nick A. Guagliardo
- Department of Pharmacology (M.R.G., M.P.B., P.Q.B., N.A.G.), University of Virginia, Charlottesville
| |
Collapse
|
9
|
Lalli E, Figueiredo BC. Prolactin as an adrenocorticotropic hormone: Prolactin signalling is a conserved key regulator of sexually dimorphic adrenal gland function in health and disease. Bioessays 2022; 44:e2200109. [PMID: 36000778 DOI: 10.1002/bies.202200109] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2022] [Revised: 07/26/2022] [Accepted: 08/11/2022] [Indexed: 11/08/2022]
Abstract
A large number of previous reports described an effect of the pituitary hormone prolactin (PRL) on steroid hormone production by the adrenal cortex. However, those studies remained anecdotal and were never converted into a conceptual and mechanistic framework, let alone being translated into clinical care. In the light of our recently published landmark study where we described PRL signalling as a pivotal regulator of the sexually dimorphic adrenal phenotype in mouse and of adrenal androgen production in humans, we present here the overarching hypothesis that PRL signalling increases the activity of Steroidogenic Factor-1 (SF-1/NR5A1), a transcription factor that has an essential role in adrenal gland development and function, to regulate adrenal cortex growth and hormonal production in physiological and pathological conditions. PRL can then be considered as a bona fide adrenocorticotropic hormone synergizing with ACTH in the endocrine control of adrenal cortex function.
Collapse
Affiliation(s)
- Enzo Lalli
- EXPOGEN-CANCER CNRS International Research Project, 660 route des Lucioles, Sophia Antipolis, Valbonne, 06560, France
- Inserm, Institut de Pharmacologie Moléculaire et Cellulaire, Valbonne, France
- Université Côte d'Azur, CNRS, Institut de Pharmacologie Moléculaire et Cellulaire, Valbonne, France
- Pelé Pequeno Principe Research Institute, Curitiba, PR, Brazil
| | - Bonald C Figueiredo
- EXPOGEN-CANCER CNRS International Research Project, 660 route des Lucioles, Sophia Antipolis, Valbonne, 06560, France
- Pelé Pequeno Principe Research Institute, Curitiba, PR, Brazil
| |
Collapse
|
10
|
Le Floch E, Cosentino T, Larsen CK, Beuschlein F, Reincke M, Amar L, Rossi GP, De Sousa K, Baron S, Chantalat S, Saintpierre B, Lenzini L, Frouin A, Giscos-Douriez I, Ferey M, Abdellatif AB, Meatchi T, Empana JP, Jouven X, Gieger C, Waldenberger M, Peters A, Cusi D, Salvi E, Meneton P, Touvier M, Deschasaux M, Druesne-Pecollo N, Boulkroun S, Fernandes-Rosa FL, Deleuze JF, Jeunemaitre X, Zennaro MC. Identification of risk loci for primary aldosteronism in genome-wide association studies. Nat Commun 2022; 13:5198. [PMID: 36057693 PMCID: PMC9440917 DOI: 10.1038/s41467-022-32896-8] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2021] [Accepted: 08/23/2022] [Indexed: 11/23/2022] Open
Abstract
Primary aldosteronism affects up to 10% of hypertensive patients and is responsible for treatment resistance and increased cardiovascular risk. Here we perform a genome-wide association study in a discovery cohort of 562 cases and 950 controls and identify three main loci on chromosomes 1, 13 and X; associations on chromosome 1 and 13 are replicated in a second cohort and confirmed by a meta-analysis involving 1162 cases and 3296 controls. The association on chromosome 13 is specific to men and stronger in bilateral adrenal hyperplasia than aldosterone producing adenoma. Candidate genes located within the two loci, CASZ1 and RXFP2, are expressed in human and mouse adrenals in different cell clusters. Their overexpression in adrenocortical cells suppresses mineralocorticoid output under basal and stimulated conditions, without affecting cortisol biosynthesis. Our study identifies the first risk loci for primary aldosteronism and highlights new mechanisms for the development of aldosterone excess.
Collapse
Affiliation(s)
- Edith Le Floch
- Centre National de Recherche en Génomique Humaine, Institut de biologie François Jacob, CEA, Université Paris-Saclay, Evry, France
| | | | - Casper K Larsen
- Université Paris Cité, Inserm, PARCC, F-75015, Paris, France
| | - Felix Beuschlein
- Medizinische Klinik und Poliklinik IV, Ludwig-Maximilians-University, 80336, Munich, Germany
- Klinik für Endokrinologie, Diabetologie und Klinische Ernährung, Universitätsspital Zürich (USZ) und Universität Zürich (UZH), Zürich, Switzerland
| | - Martin Reincke
- Medizinische Klinik und Poliklinik IV, Ludwig-Maximilians-University, 80336, Munich, Germany
| | - Laurence Amar
- Université Paris Cité, Inserm, PARCC, F-75015, Paris, France
- Assistance Publique-Hôpitaux de Paris, Hôpital Européen Georges Pompidou, Unité Hypertension artérielle, Paris, France
| | - Gian-Paolo Rossi
- DMCS 'G. Patrassi' University of Padova Medical School, University Hospital, 35126, Padova, Italy
| | - Kelly De Sousa
- Université Paris Cité, Inserm, PARCC, F-75015, Paris, France
| | - Stéphanie Baron
- Université Paris Cité, F-75006, Paris, France
- Assistance Publique-Hôpitaux de Paris, Hôpital Européen Georges Pompidou, Service de Physiologie, Paris, France
| | - Sophie Chantalat
- Centre National de Recherche en Génomique Humaine, Institut de biologie François Jacob, CEA, Université Paris-Saclay, Evry, France
| | - Benjamin Saintpierre
- Université Paris Cité, Institut Cochin, Genom'IC platform, INSERM, CNRS, 75014, Paris, France
| | - Livia Lenzini
- DMCS 'G. Patrassi' University of Padova Medical School, University Hospital, 35126, Padova, Italy
| | - Arthur Frouin
- Centre National de Recherche en Génomique Humaine, Institut de biologie François Jacob, CEA, Université Paris-Saclay, Evry, France
| | | | - Matthis Ferey
- Université Paris Cité, Inserm, PARCC, F-75015, Paris, France
| | | | - Tchao Meatchi
- Université Paris Cité, Inserm, PARCC, F-75015, Paris, France
- Assistance Publique-Hôpitaux de Paris, Hôpital Européen Georges Pompidou, Service d'Anatomie Pathologique, Paris, France
| | | | - Xavier Jouven
- Université Paris Cité, Inserm, PARCC, F-75015, Paris, France
- Assistance Publique-Hôpitaux de Paris, Hôpital Européen Georges Pompidou, Service de Cardiologie, Paris, France
| | - Christian Gieger
- Research Unit of Molecular Epidemiology, Helmholtz Zentrum München, German Research Center for Environmental Health, Neuherberg, Germany
- Institute of Epidemiology, Helmholtz Zentrum München, German Research Center for Environmental Health, Neuherberg, Germany
- German Center for Diabetes Research (DZD), Neuherberg, Germany
| | - Melanie Waldenberger
- Research Unit of Molecular Epidemiology, Helmholtz Zentrum München, German Research Center for Environmental Health, Neuherberg, Germany
- Institute of Epidemiology, Helmholtz Zentrum München, German Research Center for Environmental Health, Neuherberg, Germany
- German Research Center for Cardiovascular Research (DZHK), Partner Site Munich Heart Alliance, Munich, Germany
| | - Annette Peters
- Institute of Epidemiology, Helmholtz Zentrum München, German Research Center for Environmental Health, Neuherberg, Germany
- German Center for Diabetes Research (DZD), Neuherberg, Germany
- German Research Center for Cardiovascular Research (DZHK), Partner Site Munich Heart Alliance, Munich, Germany
| | - Daniele Cusi
- Institute of Biomedical Technologies National Research Council of Italy, Milan, Italy
- Bio4Dreams-Business Nursery for Life Sciences, Milan, Italy
| | - Erika Salvi
- Neuroalgology Unit, Fondazione IRCCS Istituto Neurologico 'Carlo Besta', Milan, Italy
| | - Pierre Meneton
- UMR_1142, INSERM, Sorbonne Université, Université Paris 13, Paris, France
| | - Mathilde Touvier
- Sorbonne Paris Nord University, INSERM U1153, INRAe U1125, CNAM, Nutritional Epidemiology Research Team (EREN), Epidemiology and Statistics Research Center - Université Paris Cité (CRESS), 93017, Bobigny, France
| | - Mélanie Deschasaux
- Sorbonne Paris Nord University, INSERM U1153, INRAe U1125, CNAM, Nutritional Epidemiology Research Team (EREN), Epidemiology and Statistics Research Center - Université Paris Cité (CRESS), 93017, Bobigny, France
| | - Nathalie Druesne-Pecollo
- Sorbonne Paris Nord University, INSERM U1153, INRAe U1125, CNAM, Nutritional Epidemiology Research Team (EREN), Epidemiology and Statistics Research Center - Université Paris Cité (CRESS), 93017, Bobigny, France
| | | | | | - Jean-François Deleuze
- Centre National de Recherche en Génomique Humaine, Institut de biologie François Jacob, CEA, Université Paris-Saclay, Evry, France
| | - Xavier Jeunemaitre
- Université Paris Cité, Inserm, PARCC, F-75015, Paris, France
- Assistance Publique-Hôpitaux de Paris, Hôpital Européen Georges Pompidou, Service de Génétique, Paris, France
| | - Maria-Christina Zennaro
- Université Paris Cité, Inserm, PARCC, F-75015, Paris, France.
- Assistance Publique-Hôpitaux de Paris, Hôpital Européen Georges Pompidou, Service de Génétique, Paris, France.
| |
Collapse
|
11
|
McDonough AA, Fenton RA. Potassium homeostasis: sensors, mediators, and targets. Pflugers Arch 2022; 474:853-867. [PMID: 35727363 PMCID: PMC10163916 DOI: 10.1007/s00424-022-02718-3] [Citation(s) in RCA: 29] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2022] [Revised: 06/08/2022] [Accepted: 06/12/2022] [Indexed: 12/16/2022]
Abstract
Transmembrane potassium (K) gradients are key determinants of membrane potential that can modulate action potentials, control muscle contractility, and influence ion channel and transporter activity. Daily K intake is normally equal to the amount of K in the entire extracellular fluid (ECF) creating a critical challenge - how to maintain ECF [K] and membrane potential in a narrow range during feast and famine. Adaptations to maintain ECF [K] include sensing the K intake, sensing ECF [K] vs. desired set-point and activating mediators that regulate K distribution between ECF and ICF, and regulate renal K excretion. In this focused review, we discuss the basis of these adaptions, including (1) potential mechanisms for rapid feedforward signaling to kidney and muscle after a meal (before a rise in ECF [K]), (2) how skeletal muscles sense and respond to changes in ECF [K], (3) effects of K on aldosterone biosynthesis, and (4) how the kidney responds to changes in ECF [K] to modify K excretion. The concepts of sexual dimorphisms in renal K handling adaptation are introduced, and the molecular mechanisms that can account for the benefits of a K-rich diet to maintain cardiovascular health are discussed. Although the big picture of K homeostasis is becoming more clear, we also highlight significant pieces of the puzzle that remain to be solved, including knowledge gaps in our understanding of initiating signals, sensors and their connection to homeostatic adjustments of ECF [K].
Collapse
Affiliation(s)
- Alicia A McDonough
- Department of Physiology and Neuroscience, University of Southern California Keck School of Medicine, Los Angeles, CA, USA.
| | - Robert A Fenton
- Department of Biomedicine, Aarhus University, Aarhus, Denmark
| |
Collapse
|
12
|
Matsuoka H, Harada K, Sugawara A, Kim D, Inoue M. Expression of p11 and heteromeric TASK channels in mouse adrenal cortical cells and H295R cells. Acta Histochem 2022; 124:151898. [PMID: 35526370 DOI: 10.1016/j.acthis.2022.151898] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2022] [Revised: 04/21/2022] [Accepted: 04/21/2022] [Indexed: 11/30/2022]
Abstract
TWIK-related acid-sensitive K+ (TASK) channels are thought to contribute to the resting membrane potential in adrenal cortical (AC) cells. However, the molecular identity of TASK channels in AC cells have not yet been elucidated. Thus, immunocytochemical and molecular biological approaches were employed to investigate the expression and intracellular distribution of TASK1 and TASK3 in mouse AC cells and H295R cells derived from human adrenocortical carcinoma. Immunocytochemical study revealed that immunoreactive materials were mainly located in the cytoplasm for TASK1 and at the cell periphery for TASK3 in mouse AC cells. A similar pattern of localization was observed when GFP-TASK1 and GFP-TASK3 were exogenously expressed in H295R cells. In addition, p11 that is known to suppress the endoplasmic reticulum exit of TASK1 was localized in the cytoplasm in mouse AC and H295R cells, but not in adrenal medullary cells. Proximity ligation assay (PLA) suggested formation of heteromeric TASK1-3 channels that were found predominantly in the cytoplasm and weakly at the cell periphery. A similar distribution was observed following exogenous expression of tandem TASK1-3 channels in H295R cells. When stimulated by angiotensin II, however, tandem TASK1-3 channels were present mainly in the cytoplasm in all H295R cells. In contrast to that in H295R cells, tandem channels were exclusively located at the cell periphery in all non-stimulated and exclusively in the cytoplasm in stimulated PC12 cells, respectively. From these results, we conclude that TASK1 proteins are present mainly in the cytoplasm and minimally at the cell periphery as a heteromeric channel with TASK3, whereas the majority of TASK3 is at the cell periphery as homomeric and heteromeric channels.
Collapse
Affiliation(s)
- Hidetada Matsuoka
- Department of Cell and Systems Physiology, University of Occupational and Environmental Health School of Medicine, Kitakyushu 807-8555, Japan
| | - Keita Harada
- Department of Cell and Systems Physiology, University of Occupational and Environmental Health School of Medicine, Kitakyushu 807-8555, Japan
| | - Akira Sugawara
- Department of Molecular Endocrinology, Tohoku University Graduate School of Medical Science, Sendai 980-8575, Japan
| | - Donghee Kim
- Department of Physiology and Biophysics, Chicago Medical School, Rosalind Franklin University of Medicine and Science, North Chicago, IL 60064-3095, USA
| | - Masumi Inoue
- Department of Cell and Systems Physiology, University of Occupational and Environmental Health School of Medicine, Kitakyushu 807-8555, Japan.
| |
Collapse
|
13
|
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: 28] [Impact Index Per Article: 9.3] [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
|
14
|
Shimada H, Yamazaki Y, Sugawara A, Sasano H, Nakamura Y. Molecular Mechanisms of Functional Adrenocortical Adenoma and Carcinoma: Genetic Characterization and Intracellular Signaling Pathway. Biomedicines 2021; 9:biomedicines9080892. [PMID: 34440096 PMCID: PMC8389593 DOI: 10.3390/biomedicines9080892] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2021] [Revised: 07/20/2021] [Accepted: 07/22/2021] [Indexed: 02/06/2023] Open
Abstract
The adrenal cortex produces steroid hormones as adrenocortical hormones in the body, secreting mineralocorticoids, glucocorticoids, and adrenal androgens, which are all considered essential for life. Adrenocortical tumors harbor divergent hormonal activity, frequently with steroid excess, and disrupt homeostasis of the body. Aldosterone-producing adenomas (APAs) cause primary aldosteronism (PA), and cortisol-producing adenomas (CPAs) are the primary cause of Cushing’s syndrome. In addition, adrenocortical carcinoma (ACC) is a highly malignant cancer harboring poor prognosis. Various genetic abnormalities have been reported, which are associated with possible pathogenesis by the alteration of intracellular signaling and activation of transcription factors. In particular, somatic mutations in APAs have been detected in genes encoding membrane proteins, especially ion channels, resulting in hypersecretion of aldosterone due to activation of intracellular calcium signaling. In addition, somatic mutations have been detected in those encoding cAMP-PKA signaling-related factors, resulting in hypersecretion of cortisol due to its driven status in CPAs. In ACC, mutations in tumor suppressor genes and Wnt-β-catenin signaling-related factors have been implicated in its pathogenesis. In this article, we review recent findings on the genetic characteristics and regulation of intracellular signaling and transcription factors in individual tumors.
Collapse
Affiliation(s)
- Hiroki Shimada
- Division of Pathology, Faculty of Medicine, Tohoku Medical and Pharmaceutical University, 1-15-1 Fukumuro, Miyagino-ku, Sendai 983-8536, Miyagi, Japan;
| | - Yuto Yamazaki
- Department of Pathology, Tohoku University Graduate School of Medicine, 2-1 Seiryo-machi, Aoba-ku, Sendai 980-8575, Miyagi, Japan; (Y.Y.); (H.S.)
| | - Akira Sugawara
- Department of Molecular Endocrinology, Tohoku University Graduate School of Medicine, 2-1 Seiryo-machi, Aoba-ku, Sendai 980-8575, Miyagi, Japan;
| | - Hironobu Sasano
- Department of Pathology, Tohoku University Graduate School of Medicine, 2-1 Seiryo-machi, Aoba-ku, Sendai 980-8575, Miyagi, Japan; (Y.Y.); (H.S.)
| | - Yasuhiro Nakamura
- Division of Pathology, Faculty of Medicine, Tohoku Medical and Pharmaceutical University, 1-15-1 Fukumuro, Miyagino-ku, Sendai 983-8536, Miyagi, Japan;
- Correspondence: ; Tel.: +81-22-290-8731
| |
Collapse
|
15
|
The Sexually Dimorphic Adrenal Cortex: Implications for Adrenal Disease. Int J Mol Sci 2021; 22:ijms22094889. [PMID: 34063067 PMCID: PMC8124132 DOI: 10.3390/ijms22094889] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2021] [Revised: 04/26/2021] [Accepted: 04/28/2021] [Indexed: 02/06/2023] Open
Abstract
Many adrenocortical diseases are more prevalent in women than in men, but the reasons underlying this sex bias are still unknown. Recent studies involving gonadectomy and sex hormone replacement experiments in mice have shed some light onto the molecular basis of sexual dimorphism in the adrenal cortex. Indeed, it has been shown that gonadal hormones influence many aspects of adrenal physiology, ranging from stem cell-dependent tissue turnover to steroidogenesis and X-zone dynamics. This article reviews current knowledge on adrenal cortex sexual dimorphism and the potential mechanisms underlying sex hormone influence of adrenal homeostasis. Both topics are expected to contribute to personalized and novel therapeutic approaches in the future.
Collapse
|
16
|
Pignatti E, Flück CE. Adrenal cortex development and related disorders leading to adrenal insufficiency. Mol Cell Endocrinol 2021; 527:111206. [PMID: 33607267 DOI: 10.1016/j.mce.2021.111206] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/02/2020] [Revised: 02/02/2021] [Accepted: 02/03/2021] [Indexed: 02/07/2023]
Abstract
The adult human adrenal cortex produces steroid hormones that are crucial for life, supporting immune response, glucose homeostasis, salt balance and sexual maturation. It consists of three histologically distinct and functionally specialized zones. The fetal adrenal forms from mesodermal material and produces predominantly adrenal C19 steroids from its fetal zone, which involutes after birth. Transition to the adult cortex occurs immediately after birth for the formation of the zona glomerulosa and fasciculata for aldosterone and cortisol production and continues through infancy until the zona reticularis for adrenal androgen production is formed with adrenarche. The development of this indispensable organ is complex and not fully understood. This article gives an overview of recent knowledge gained of adrenal biology from two perspectives: one, from basic science studying adrenal development, zonation and homeostasis; and two, from adrenal disorders identified in persons manifesting with various isolated or syndromic forms of primary adrenal insufficiency.
Collapse
Affiliation(s)
- Emanuele Pignatti
- Pediatric Endocrinology, Diabetology and Metabolism, Department of Pediatrics, Bern and Department of BioMedical Research, University Hospital Inselspital, University of Bern, 3010, Bern, Switzerland.
| | - Christa E Flück
- Pediatric Endocrinology, Diabetology and Metabolism, Department of Pediatrics, Bern and Department of BioMedical Research, University Hospital Inselspital, University of Bern, 3010, Bern, Switzerland.
| |
Collapse
|
17
|
Le Ribeuz H, Montani D, Antigny F. The Experimental TASK-1 Potassium Channel Inhibitor A293 Can Be Employed for Rhythm Control of Persistent Atrial Fibrillation in a Translational Large Animal Model. Front Physiol 2021; 12:668267. [PMID: 33912077 PMCID: PMC8072364 DOI: 10.3389/fphys.2021.668267] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2021] [Accepted: 03/15/2021] [Indexed: 11/13/2022] Open
Affiliation(s)
- Hélène Le Ribeuz
- Université Paris-Saclay, Faculté de Médecine, Le Kremlin-Bicêtre, France.,INSERM UMR_S 999 ≪ Hypertension Pulmonaire: Physiopathologie et Innovation Thérapeutique ≫, Hôpital Marie Lannelongue, Le Plessis-Robinson, France.,Assistance Publique - Hôpitaux de Paris (AP-HP), Service de Pneumologie et Soins Intensifs Respiratoires, Centre de Référence de l'Hypertension Pulmonaire, Hôpital Bicêtre, Le Kremlin-Bicêtre, France
| | - David Montani
- Université Paris-Saclay, Faculté de Médecine, Le Kremlin-Bicêtre, France.,INSERM UMR_S 999 ≪ Hypertension Pulmonaire: Physiopathologie et Innovation Thérapeutique ≫, Hôpital Marie Lannelongue, Le Plessis-Robinson, France.,Assistance Publique - Hôpitaux de Paris (AP-HP), Service de Pneumologie et Soins Intensifs Respiratoires, Centre de Référence de l'Hypertension Pulmonaire, Hôpital Bicêtre, Le Kremlin-Bicêtre, France
| | - Fabrice Antigny
- Université Paris-Saclay, Faculté de Médecine, Le Kremlin-Bicêtre, France.,INSERM UMR_S 999 ≪ Hypertension Pulmonaire: Physiopathologie et Innovation Thérapeutique ≫, Hôpital Marie Lannelongue, Le Plessis-Robinson, France.,Assistance Publique - Hôpitaux de Paris (AP-HP), Service de Pneumologie et Soins Intensifs Respiratoires, Centre de Référence de l'Hypertension Pulmonaire, Hôpital Bicêtre, Le Kremlin-Bicêtre, France
| |
Collapse
|
18
|
Barrett PQ, Guagliardo NA, Bayliss DA. Ion Channel Function and Electrical Excitability in the Zona Glomerulosa: A Network Perspective on Aldosterone Regulation. Annu Rev Physiol 2020; 83:451-475. [PMID: 33176563 DOI: 10.1146/annurev-physiol-030220-113038] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Aldosterone excess is a pathogenic factor in many hypertensive disorders. The discovery of numerous somatic and germline mutations in ion channels in primary hyperaldosteronism underscores the importance of plasma membrane conductances in determining the activation state of zona glomerulosa (zG) cells. Electrophysiological recordings describe an electrically quiescent behavior for dispersed zG cells. Yet, emerging data indicate that in native rosette structures in situ, zG cells are electrically excitable, generating slow periodic voltage spikes and coordinated bursts of Ca2+ oscillations. We revisit data to understand how a multitude of conductances may underlie voltage/Ca2+ oscillations, recognizing that zG layer self-renewal and cell heterogeneity may complicate this task. We review recent data to understand rosette architecture and apply maxims derived from computational network modeling to understand rosette function. The challenge going forward is to uncover how the rosette orchestrates the behavior of a functional network of conditional oscillators to control zG layer performance and aldosterone secretion.
Collapse
Affiliation(s)
- Paula Q Barrett
- Department of Pharmacology, University of Virginia School of Medicine, Charlottesville, Virginia 22908, USA; , ,
| | - Nick A Guagliardo
- Department of Pharmacology, University of Virginia School of Medicine, Charlottesville, Virginia 22908, USA; , ,
| | - Douglas A Bayliss
- Department of Pharmacology, University of Virginia School of Medicine, Charlottesville, Virginia 22908, USA; , ,
| |
Collapse
|
19
|
Gürtler F, Jordan K, Tegtmeier I, Herold J, Stindl J, Warth R, Bandulik S. Cellular Pathophysiology of Mutant Voltage-Dependent Ca2+ Channel CACNA1H in Primary Aldosteronism. Endocrinology 2020; 161:5891807. [PMID: 32785697 DOI: 10.1210/endocr/bqaa135] [Citation(s) in RCA: 2] [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: 03/06/2020] [Accepted: 08/04/2020] [Indexed: 12/29/2022]
Abstract
The physiological stimulation of aldosterone production in adrenocortical glomerulosa cells by angiotensin II and high plasma K+ depends on the depolarization of the cell membrane potential and the subsequent Ca2+ influx via voltage-activated Ca2+ channels. Germline mutations of the low-voltage activated T-type Ca2+ channel CACNA1H (Cav3.2) have been found in patients with primary aldosteronism. Here, we investigated the electrophysiology and Ca2+ signaling of adrenal NCI-H295R cells overexpressing CACNA1H wildtype and mutant M1549V in order to understand how mutant CACNA1H alters adrenal cell function. Whole-cell patch-clamp measurements revealed a strong activation of mutant CACNA1H at the resting membrane potential of adrenal cells. Both the expression of wildtype and mutant CACNA1H led to a depolarized membrane potential. In addition, cells expressing mutant CACNA1H developed pronounced action potential-like membrane voltage oscillations. Ca2+ measurements showed an increased basal Ca2+ activity, an altered K+ sensitivity, and abnormal oscillating Ca2+ changes in cells with mutant CACNA1H. In addition, removal of extracellular Na+ reduced CACNA1H current, voltage oscillations, and Ca2+ levels in mutant cells, suggesting a role of the partial Na+ conductance of CACNA1H in cellular pathology. In conclusion, the pathogenesis of stimulus-independent aldosterone production in patients with CACNA1H mutations involves several factors: i) a loss of normal control of the membrane potential, ii) an increased Ca2+ influx at basal conditions, and iii) alterations in sensitivity to extracellular K+ and Na+. Finally, our findings underline the importance of CACNA1H in the control of aldosterone production and support the concept of the glomerulosa cell as an electrical oscillator.
Collapse
Affiliation(s)
- Florian Gürtler
- Medical Cell Biology, University of Regensburg, Regensburg, Germany
| | - Katrin Jordan
- Medical Cell Biology, University of Regensburg, Regensburg, Germany
| | - Ines Tegtmeier
- Medical Cell Biology, University of Regensburg, Regensburg, Germany
| | - Janina Herold
- Medical Cell Biology, University of Regensburg, Regensburg, Germany
| | - Julia Stindl
- Medical Cell Biology, University of Regensburg, Regensburg, Germany
| | - Richard Warth
- Medical Cell Biology, University of Regensburg, Regensburg, Germany
| | - Sascha Bandulik
- Medical Cell Biology, University of Regensburg, Regensburg, Germany
| |
Collapse
|
20
|
Taylor MJ, Ullenbruch MR, Frucci EC, Rege J, Ansorge MS, Gomez-Sanchez CE, Begum S, Laufer E, Breault DT, Rainey WE. Chemogenetic activation of adrenocortical Gq signaling causes hyperaldosteronism and disrupts functional zonation. J Clin Invest 2020; 130:83-93. [PMID: 31738186 DOI: 10.1172/jci127429] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2019] [Accepted: 09/18/2019] [Indexed: 02/04/2023] Open
Abstract
The mineralocorticoid aldosterone is produced in the adrenal zona glomerulosa (ZG) under the control of the renin-angiotensin II (AngII) system. Primary aldosteronism (PA) results from renin-independent production of aldosterone and is a common cause of hypertension. PA is caused by dysregulated localization of the enzyme aldosterone synthase (Cyp11b2), which is normally restricted to the ZG. Cyp11b2 transcription and aldosterone production are predominantly regulated by AngII activation of the Gq signaling pathway. Here, we report the generation of transgenic mice with Gq-coupled designer receptors exclusively activated by designer drugs (DREADDs) specifically in the adrenal cortex. We show that adrenal-wide ligand activation of Gq DREADD receptors triggered disorganization of adrenal functional zonation, with induction of Cyp11b2 in glucocorticoid-producing zona fasciculata cells. This result was consistent with increased renin-independent aldosterone production and hypertension. All parameters were reversible following termination of DREADD-mediated Gq signaling. These findings demonstrate that Gq signaling is sufficient for adrenocortical aldosterone production and implicate this pathway in the determination of zone-specific steroid production within the adrenal cortex. This transgenic mouse also provides an inducible and reversible model of hyperaldosteronism to investigate PA therapeutics and the mechanisms leading to the damaging effects of aldosterone on the cardiovascular system.
Collapse
Affiliation(s)
- Matthew J Taylor
- Department of Molecular and Integrative Physiology, University of Michigan, Ann Arbor, Michigan, USA
| | - Matthew R Ullenbruch
- Department of Molecular and Integrative Physiology, University of Michigan, Ann Arbor, Michigan, USA
| | - Emily C Frucci
- Department of Molecular and Integrative Physiology, University of Michigan, Ann Arbor, Michigan, USA
| | - Juilee Rege
- Department of Molecular and Integrative Physiology, University of Michigan, Ann Arbor, Michigan, USA
| | - Mark S Ansorge
- The Sackler Institute for Developmental Psychobiology, Columbia University, New York, New York, USA
| | - Celso E Gomez-Sanchez
- Endocrine Section, G.V. (Sonny) Montgomery VA Medical Center and the Department of Pharmacology and Toxicology, University of Mississippi Medical Center, Jackson, Mississippi, USA
| | - Salma Begum
- Department of Obstetrics, Gynecology and Women's Health, Rutgers, The State University of New Jersey, Newark, New Jersey, USA
| | - Edward Laufer
- Department of Human Genetics, University of Utah, Salt Lake City, Utah, USA
| | - David T Breault
- Department of Pediatrics, Division of Endocrinology, Boston Children's Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - William E Rainey
- Department of Molecular and Integrative Physiology, University of Michigan, Ann Arbor, Michigan, USA.,Division of Metabolism, Endocrinology, and Diabetes, Department of Internal Medicine, University of Michigan, Ann Arbor, Michigan, USA
| |
Collapse
|
21
|
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
|
22
|
TASK channels: channelopathies, trafficking, and receptor-mediated inhibition. Pflugers Arch 2020; 472:911-922. [DOI: 10.1007/s00424-020-02403-3] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2020] [Revised: 05/08/2020] [Accepted: 05/18/2020] [Indexed: 01/06/2023]
|
23
|
Duan W, Hicks J, Makara MA, Ilkayeva O, Abraham DM. TASK-1 and TASK-3 channels modulate pressure overload-induced cardiac remodeling and dysfunction. Am J Physiol Heart Circ Physiol 2020; 318:H566-H580. [PMID: 31977249 DOI: 10.1152/ajpheart.00739.2018] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Tandem pore domain acid-sensitive K+ (TASK) channels are present in cardiac tissue; however, their contribution to cardiac pathophysiology is not well understood. Here, we investigate the role of TASK-1 and TASK-3 in the pathogenesis of cardiac dysfunction using both human tissue and mouse models of genetic TASK channel loss of function. Compared with normal human cardiac tissue, TASK-1 gene expression is reduced in association with either cardiac hypertrophy alone or combined cardiac hypertrophy and heart failure. In a pressure overload cardiomyopathy model, TASK-1 global knockout (TASK-1 KO) mice have both reduced cardiac hypertrophy and preserved cardiac function compared with wild-type mice. In contrast to the TASK-1 KO mouse pressure overload response, TASK-3 global knockout (TASK-3 KO) mice develop cardiac hypertrophy and a delayed onset of cardiac dysfunction compared with wild-type mice. The cardioprotective effects observed in TASK-1 KO mice are associated with pressure overload-induced augmentation of AKT phosphorylation and peroxisome proliferator-activated receptor-γ coactivator-1α (PGC-1α) expression, with consequent augmentation of cardiac energetics and fatty acid oxidation. The protective effects of TASK-1 loss of function are associated with an enhancement of physiologic hypertrophic signaling and preserved metabolic functions. These findings may provide a rationale for TASK-1 channel inhibition in the treatment of cardiac dysfunction.NEW & NOTEWORTHY The role of tandem pore domain acid-sensitive K+ (TASK) channels in cardiac function is not well understood. This study demonstrates that TASK channel gene expression is associated with the onset of human cardiac hypertrophy and heart failure. TASK-1 and TASK-3 strongly affect the development of pressure overload cardiomyopathies in genetic models of TASK-1 and TASK-3 loss of function. The effects of TASK-1 loss of function were associated with enhanced AKT phosphorylation and expression of peroxisome proliferator-activated receptor-γ coactivator-1 (PGC-1) transcription factor. These data suggest that TASK channels influence the development of cardiac hypertrophy and dysfunction in response to injury.
Collapse
Affiliation(s)
- Wei Duan
- Department of Medicine, Duke University Medical Center, Durham, North Carolina
| | - Jonné Hicks
- Department of Medicine, Duke University Medical Center, Durham, North Carolina
| | | | - Olga Ilkayeva
- Duke Molecular Physiology Institute, Duke University, Durham, North Carolina
| | - Dennis M Abraham
- Department of Medicine, Duke University Medical Center, Durham, North Carolina
| |
Collapse
|
24
|
Wen ZY, Bian C, You X, Zhang X, Li J, Zhan Q, Peng Y, Li YY, Shi Q. Characterization of two kcnk3 genes in Nile tilapia (Oreochromis niloticus): Molecular cloning, tissue distribution, and transcriptional changes in various salinity of seawater. Genomics 2019; 112:2213-2222. [PMID: 31881264 DOI: 10.1016/j.ygeno.2019.12.017] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2019] [Revised: 12/23/2019] [Accepted: 12/23/2019] [Indexed: 02/07/2023]
Abstract
As one important member of the two-pore-domain potassium channel (K2P) family, potassium channel subfamily K member 3 (KCNK3) has been reported for thermogenesis regulation, energy homeostasis, membrane potential conduction, and pulmonary hypertension in mammals. However, its roles in fishes are far less examined and published. In the present study, we identified two kcnk3 genes (kcnk3a and kcnk3b) in an euryhaline fish, Nile tilapia (Oreochromis niloticus), by molecular cloning, genomic survey and laboratory experiments to investigate their potential roles for osmoregulation. We obtained full-length coding sequences of the kcnk3a and kcnk3b genes (1209 and 1173 bp), which encode 402 and 390 amino acids, respectively. Subsequent multiple sequence alignments, putative 3D-structure model prediction, genomic survey and phylogenetic analysis confirmed that two kcnk3 paralogs are widely presented in fish genomes. Interestingly, a DNA fragment inversion of a kcnk3a cluster was found in Cypriniforme in comparison with other fishes. Quantitative real-time PCRs demonstrated that both the tilapia kcnk3 genes were detected in all the examined tissues with a similar distribution pattern, and the highest transcriptions were observed in the heart. Meanwhile, both kcnk3 genes in the gill were proved to have a similar transcriptional change pattern in response to various salinity of seawater, implying that they might be involved in osmoregulation. Furthermore, three predicted transcription factors (arid3a, arid3b, and arid5a) of both kcnk3 genes also showed a similar pattern as their target genes in response to the various salinity, suggesting their potential positive regulatory roles. In summary, we for the first time characterized the two kcnk3 genes in Nile tilapia, and demonstrated their potential involvement in osmoregulation for this economically important fish.
Collapse
Affiliation(s)
- Zheng-Yong Wen
- BGI Education Center, University of Chinese Academy of Sciences, Shenzhen 518083, China; Shenzhen Key Lab of Marine Genomics, Guangdong Provincial Key Lab of Molecular Breeding in Marine Economic Animals, BGI Academy of Marine Sciences, BGI Marine, BGI, Shenzhen 518083, China
| | - Chao Bian
- Shenzhen Key Lab of Marine Genomics, Guangdong Provincial Key Lab of Molecular Breeding in Marine Economic Animals, BGI Academy of Marine Sciences, BGI Marine, BGI, Shenzhen 518083, China
| | - Xinxin You
- BGI Education Center, University of Chinese Academy of Sciences, Shenzhen 518083, China; Shenzhen Key Lab of Marine Genomics, Guangdong Provincial Key Lab of Molecular Breeding in Marine Economic Animals, BGI Academy of Marine Sciences, BGI Marine, BGI, Shenzhen 518083, China
| | - Xinhui Zhang
- Shenzhen Key Lab of Marine Genomics, Guangdong Provincial Key Lab of Molecular Breeding in Marine Economic Animals, BGI Academy of Marine Sciences, BGI Marine, BGI, Shenzhen 518083, China
| | - Jia Li
- Shenzhen Key Lab of Marine Genomics, Guangdong Provincial Key Lab of Molecular Breeding in Marine Economic Animals, BGI Academy of Marine Sciences, BGI Marine, BGI, Shenzhen 518083, China
| | - Qiuyao Zhan
- BGI Education Center, University of Chinese Academy of Sciences, Shenzhen 518083, China; Shenzhen Key Lab of Marine Genomics, Guangdong Provincial Key Lab of Molecular Breeding in Marine Economic Animals, BGI Academy of Marine Sciences, BGI Marine, BGI, Shenzhen 518083, China
| | - Yuxiang Peng
- BGI Education Center, University of Chinese Academy of Sciences, Shenzhen 518083, China; Shenzhen Key Lab of Marine Genomics, Guangdong Provincial Key Lab of Molecular Breeding in Marine Economic Animals, BGI Academy of Marine Sciences, BGI Marine, BGI, Shenzhen 518083, China
| | - Yuan-You Li
- School of Marine Sciences, South China Agricultural University, Guangzhou 510642, China.
| | - Qiong Shi
- BGI Education Center, University of Chinese Academy of Sciences, Shenzhen 518083, China; Shenzhen Key Lab of Marine Genomics, Guangdong Provincial Key Lab of Molecular Breeding in Marine Economic Animals, BGI Academy of Marine Sciences, BGI Marine, BGI, Shenzhen 518083, China.
| |
Collapse
|
25
|
Tevosian SG, Fox SC, Ghayee HK. Molecular Mechanisms of Primary Aldosteronism. Endocrinol Metab (Seoul) 2019; 34:355-366. [PMID: 31884735 PMCID: PMC6935778 DOI: 10.3803/enm.2019.34.4.355] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/29/2019] [Revised: 12/02/2019] [Accepted: 12/09/2019] [Indexed: 01/11/2023] Open
Abstract
Primary aldosteronism (PA) results from excess production of mineralocorticoid hormone aldosterone by the adrenal cortex. It is normally caused either by unilateral aldosterone-producing adenoma (APA) or by bilateral aldosterone excess as a result of bilateral adrenal hyperplasia. PA is the most common cause of secondary hypertension and associated morbidity and mortality. While most cases of PA are sporadic, an important insight into this debilitating disease has been derived through investigating the familial forms of the disease that affect only a minor fraction of PA patients. The advent of gene expression profiling has shed light on the genes and intracellular signaling pathways that may play a role in the pathogenesis of these tumors. The genetic basis for several forms of familial PA has been uncovered in recent years although the list is likely to expand. Recently, the work from several laboratories provided evidence for the involvement of mammalian target of rapamycin pathway and inflammatory cytokines in APAs; however, their mechanism of action in tumor development and pathophysiology remains to be understood.
Collapse
Affiliation(s)
- Sergei G Tevosian
- Department of Physiological Sciences, College of Veterinary Medicine, University of Florida, Gainesville, FL, USA
| | - Shawna C Fox
- Department of Pharmacotherapy and Translational Research, College of Pharmacy, University of Florida, Gainesville, FL, USA
| | - Hans K Ghayee
- Division of Endocrinology, Department of Medicine, Malcom Randall VA Medical Center, University of Florida, Gainesville, FL, USA.
| |
Collapse
|
26
|
Guagliardo NA, Yao J, Stipes EJ, Cechova S, Le TH, Bayliss DA, Breault DT, Barrett PQ. Adrenal Tissue-Specific Deletion of TASK Channels Causes Aldosterone-Driven Angiotensin II-Independent Hypertension. Hypertension 2019; 73:407-414. [PMID: 30580687 PMCID: PMC6326871 DOI: 10.1161/hypertensionaha.118.11962] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
The renin-angiotensin system tightly controls aldosterone synthesis. Dysregulation is evident in hypertension (primary aldosteronism), low renin, and resistant hypertension) but also can exist in normotension. Whether chronic, mild aldosterone autonomy can elicit hypertension remains untested. Previously, we reported that global genetic deletion of 2 pore-domain TWIK-relative acid-sensitive potassium channels, TASK-1 and TASK-3, from mice produces striking aldosterone excess, low renin, and hypertension. Here, we deleted TASK-1 and TASK-3 channels selectively from zona glomerulosa cells and generated a model of mild aldosterone autonomy with attendant hypertension that is aldosterone-driven and Ang II (angiotensin II)-independent. This study shows that a zona glomerulosa-specific channel defect can produce mild autonomous hyperaldosteronism sufficient to cause chronic blood pressure elevation.
Collapse
Affiliation(s)
- Nick A Guagliardo
- From the Department of Pharmacology (N.A.G., J.Y., E.J.S., D.A.B, P.Q.B.), University of Virginia School of Medicine, Charlottesville
| | - Junlan Yao
- From the Department of Pharmacology (N.A.G., J.Y., E.J.S., D.A.B, P.Q.B.), University of Virginia School of Medicine, Charlottesville
| | - Eric J Stipes
- From the Department of Pharmacology (N.A.G., J.Y., E.J.S., D.A.B, P.Q.B.), University of Virginia School of Medicine, Charlottesville
| | - Sylvia Cechova
- Division of Nephrology, Department of Medicine (S.C., T.H.L.), University of Virginia School of Medicine, Charlottesville
| | - Thu H Le
- Division of Nephrology, Department of Medicine (S.C., T.H.L.), University of Virginia School of Medicine, Charlottesville
| | - Douglas A Bayliss
- From the Department of Pharmacology (N.A.G., J.Y., E.J.S., D.A.B, P.Q.B.), University of Virginia School of Medicine, Charlottesville
| | - David T Breault
- Department of Pediatrics/Division of Endocrinology, Boston Children's Hospital, Harvard Medical School, MA (D.T.B.)
| | - Paula Q Barrett
- From the Department of Pharmacology (N.A.G., J.Y., E.J.S., D.A.B, P.Q.B.), University of Virginia School of Medicine, Charlottesville
| |
Collapse
|
27
|
Schewe J, Seidel E, Forslund S, Marko L, Peters J, Muller DN, Fahlke C, Stölting G, Scholl U. Elevated aldosterone and blood pressure in a mouse model of familial hyperaldosteronism with ClC-2 mutation. Nat Commun 2019; 10:5155. [PMID: 31727896 PMCID: PMC6856192 DOI: 10.1038/s41467-019-13033-4] [Citation(s) in RCA: 33] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2019] [Accepted: 10/17/2019] [Indexed: 12/11/2022] Open
Abstract
Gain-of-function mutations in the chloride channel ClC-2 were recently described as a cause of familial hyperaldosteronism type II (FH-II). Here, we report the generation of a mouse model carrying a missense mutation homologous to the most common FH-II-associated CLCN2 mutation. In these Clcn2R180Q/+ mice, adrenal morphology is normal, but Cyp11b2 expression and plasma aldosterone levels are elevated. Male Clcn2R180Q/+ mice have increased aldosterone:renin ratios as well as elevated blood pressure levels. The counterpart knockout model (Clcn2−/−), in contrast, requires elevated renin levels to maintain normal aldosterone levels. Adrenal slices of Clcn2R180Q/+ mice show increased calcium oscillatory activity. Together, our work provides a knockin mouse model with a mild form of primary aldosteronism, likely due to increased chloride efflux and depolarization. We demonstrate a role of ClC-2 in normal aldosterone production beyond the observed pathophysiology. Mutations in the chloride channel ClC-2 have been associated with familial forms of hyperaldosteronism. Here, Schewe et al. generated a mouse model carrying the most common mutation found in patients and find it recapitulates key features of the disease, providing a unique tool for future studies on its pathogenesis.
Collapse
Affiliation(s)
- Julia Schewe
- Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin Berlin Institute of Health, Department of Nephrology and Medical Intensive Care, Augustenburger Platz 1, Berlin, 13353, Germany.,Berlin Institute of Health (BIH), Anna-Louisa-Karsch-Str. 2, 10178, Berlin, Germany.,Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin and Berlin Institute of Health, BIH Center for Regenerative Therapies, Föhrer Str. 15, Berlin, 13353, Germany.,Department of Nephrology, School of Medicine, Heinrich-Heine-Universität Düsseldorf, Universitätsstraße 1, 40225, Düsseldorf, Germany
| | - Eric Seidel
- Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin Berlin Institute of Health, Department of Nephrology and Medical Intensive Care, Augustenburger Platz 1, Berlin, 13353, Germany.,Berlin Institute of Health (BIH), Anna-Louisa-Karsch-Str. 2, 10178, Berlin, Germany.,Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin and Berlin Institute of Health, BIH Center for Regenerative Therapies, Föhrer Str. 15, Berlin, 13353, Germany.,Department of Nephrology, School of Medicine, Heinrich-Heine-Universität Düsseldorf, Universitätsstraße 1, 40225, Düsseldorf, Germany
| | - Sofia Forslund
- Berlin Institute of Health (BIH), Anna-Louisa-Karsch-Str. 2, 10178, Berlin, Germany.,Max Delbruck Center for Molecular Medicine in the Helmholtz Association, Berlin, Germany.,Experimental and Clinical Research Center, a cooperation of Charité-Universitätsmedizin Berlin and Max Delbruck Center for Molecular Medicine, Lindenberger Weg 80, Berlin, 13125, Germany.,Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin and Berlin Institute of Health, Berlin, Germany
| | - Lajos Marko
- Berlin Institute of Health (BIH), Anna-Louisa-Karsch-Str. 2, 10178, Berlin, Germany.,Max Delbruck Center for Molecular Medicine in the Helmholtz Association, Berlin, Germany.,Experimental and Clinical Research Center, a cooperation of Charité-Universitätsmedizin Berlin and Max Delbruck Center for Molecular Medicine, Lindenberger Weg 80, Berlin, 13125, Germany.,Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin and Berlin Institute of Health, Berlin, Germany
| | - Jörg Peters
- Department of Physiology, Universitätsmedizin Greifswald, Friedrich-Ludwig-Jahn-Str. 15a, 17475, Greifswald, Germany
| | - Dominik N Muller
- Berlin Institute of Health (BIH), Anna-Louisa-Karsch-Str. 2, 10178, Berlin, Germany.,Max Delbruck Center for Molecular Medicine in the Helmholtz Association, Berlin, Germany.,Experimental and Clinical Research Center, a cooperation of Charité-Universitätsmedizin Berlin and Max Delbruck Center for Molecular Medicine, Lindenberger Weg 80, Berlin, 13125, Germany.,Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin and Berlin Institute of Health, Berlin, Germany
| | - Christoph Fahlke
- Institute of Complex Systems, Zelluläre Biophysik (ICS-4), Forschungszentrum Jülich, 52425, Jülich, Germany
| | - Gabriel Stölting
- Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin Berlin Institute of Health, Department of Nephrology and Medical Intensive Care, Augustenburger Platz 1, Berlin, 13353, Germany.,Berlin Institute of Health (BIH), Anna-Louisa-Karsch-Str. 2, 10178, Berlin, Germany.,Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin and Berlin Institute of Health, BIH Center for Regenerative Therapies, Föhrer Str. 15, Berlin, 13353, Germany.,Institute of Complex Systems, Zelluläre Biophysik (ICS-4), Forschungszentrum Jülich, 52425, Jülich, Germany
| | - Ute Scholl
- Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin Berlin Institute of Health, Department of Nephrology and Medical Intensive Care, Augustenburger Platz 1, Berlin, 13353, Germany. .,Berlin Institute of Health (BIH), Anna-Louisa-Karsch-Str. 2, 10178, Berlin, Germany. .,Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin and Berlin Institute of Health, BIH Center for Regenerative Therapies, Föhrer Str. 15, Berlin, 13353, Germany. .,Department of Nephrology, School of Medicine, Heinrich-Heine-Universität Düsseldorf, Universitätsstraße 1, 40225, Düsseldorf, Germany.
| |
Collapse
|
28
|
Genetic causes of primary aldosteronism. Exp Mol Med 2019; 51:1-12. [PMID: 31695023 PMCID: PMC6834635 DOI: 10.1038/s12276-019-0337-9] [Citation(s) in RCA: 33] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2019] [Revised: 08/21/2019] [Accepted: 09/09/2019] [Indexed: 11/09/2022] Open
Abstract
Primary aldosteronism is characterized by at least partially autonomous production of the adrenal steroid hormone aldosterone and is the most common cause of secondary hypertension. The most frequent subforms are idiopathic hyperaldosteronism and aldosterone-producing adenoma. Rare causes include unilateral hyperplasia, adrenocortical carcinoma and Mendelian forms (familial hyperaldosteronism). Studies conducted in the last eight years have identified somatic driver mutations in a substantial portion of aldosterone-producing adenomas, including the genes KCNJ5 (encoding inwardly rectifying potassium channel GIRK4), CACNA1D (encoding a subunit of L-type voltage-gated calcium channel CaV1.3), ATP1A1 (encoding a subunit of Na+/K+-ATPase), ATP2B3 (encoding a Ca2+-ATPase), and CTNNB1 (encoding ß-catenin). In addition, aldosterone-producing cells were recently reported to form small clusters (aldosterone-producing cell clusters) beneath the adrenal capsule. Such clusters accumulate with age and appear to be more frequent in individuals with idiopathic hyperaldosteronism. The fact that they are associated with somatic mutations implicated in aldosterone-producing adenomas also suggests a precursor function for adenomas. Rare germline variants of CYP11B2 (encoding aldosterone synthase), CLCN2 (encoding voltage-gated chloride channel ClC-2), KCNJ5, CACNA1H (encoding a subunit of T-type voltage-gated calcium channel CaV3.2), and CACNA1D have been reported in different subtypes of familial hyperaldosteronism. Collectively, these studies suggest that primary aldosteronism is largely due to genetic mutations in single genes, with potential implications for diagnosis and therapy.
Collapse
|
29
|
Göppner C, Orozco IJ, Hoegg-Beiler MB, Soria AH, Hübner CA, Fernandes-Rosa FL, Boulkroun S, Zennaro MC, Jentsch TJ. Pathogenesis of hypertension in a mouse model for human CLCN2 related hyperaldosteronism. Nat Commun 2019; 10:4678. [PMID: 31615979 PMCID: PMC6794291 DOI: 10.1038/s41467-019-12113-9] [Citation(s) in RCA: 32] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2019] [Accepted: 08/21/2019] [Indexed: 12/31/2022] Open
Abstract
Human primary aldosteronism (PA) can be caused by mutations in several ion channel genes but mouse models replicating this condition are lacking. We now show that almost all known PA-associated CLCN2 mutations markedly increase ClC-2 chloride currents and generate knock-in mice expressing a constitutively open ClC-2 Cl− channel as mouse model for PA. The Clcn2op allele strongly increases the chloride conductance of zona glomerulosa cells, provoking a strong depolarization and increasing cytoplasmic Ca2+ concentration. Clcn2op mice display typical features of human PA, including high serum aldosterone in the presence of low renin activity, marked hypertension and hypokalemia. These symptoms are more pronounced in homozygous Clcn2op/op than in heterozygous Clcn2+/op mice. This difference is attributed to the unexpected finding that only ~50 % of Clcn2+/op zona glomerulosa cells are depolarized. By reproducing essential features of human PA, Clcn2op mice are a valuable model to study the pathological mechanisms underlying this disease. Mutations in the chloride channel ClC-2 have been found in primary aldosteronism (PA). Here, Göppner et al. generate transgenic mice expressing a mutant form of ClC-2 that displays increased chloride currents like patient mutations, and find it recapitulates the key pathological features of PA.
Collapse
Affiliation(s)
- Corinna Göppner
- Leibniz-Forschungsinstitut für Molekulare Pharmakologie (FMP), Berlin, Germany.,Max-Delbrück-Centrum für Molekulare Medizin (MDC), Berlin, Germany
| | - Ian J Orozco
- Leibniz-Forschungsinstitut für Molekulare Pharmakologie (FMP), Berlin, Germany.,Max-Delbrück-Centrum für Molekulare Medizin (MDC), Berlin, Germany
| | - Maja B Hoegg-Beiler
- Leibniz-Forschungsinstitut für Molekulare Pharmakologie (FMP), Berlin, Germany.,Max-Delbrück-Centrum für Molekulare Medizin (MDC), Berlin, Germany
| | - Audrey H Soria
- Leibniz-Forschungsinstitut für Molekulare Pharmakologie (FMP), Berlin, Germany.,Max-Delbrück-Centrum für Molekulare Medizin (MDC), Berlin, Germany
| | | | - Fabio L Fernandes-Rosa
- INSERM, UMRS_970, Paris Cardiovascular Research Center, Paris, France.,Université Paris Descartes, Sorbonne Paris Cité, Paris, France
| | - Sheerazed Boulkroun
- INSERM, UMRS_970, Paris Cardiovascular Research Center, Paris, France.,Université Paris Descartes, Sorbonne Paris Cité, Paris, France
| | - Maria-Christina Zennaro
- INSERM, UMRS_970, Paris Cardiovascular Research Center, Paris, France.,Université Paris Descartes, Sorbonne Paris Cité, Paris, France.,Assistance Publique-Hôpitaux de Paris, Hôpital Européen Georges Pompidou, Service de Génétique, Paris, France
| | - Thomas J Jentsch
- Leibniz-Forschungsinstitut für Molekulare Pharmakologie (FMP), Berlin, Germany. .,Max-Delbrück-Centrum für Molekulare Medizin (MDC), Berlin, Germany. .,NeuroCure Cluster of Excellence, Charité Universitätsmedizin Berlin, Berlin, Germany.
| |
Collapse
|
30
|
Retinoic acid receptor α as a novel contributor to adrenal cortex structure and function through interactions with Wnt and Vegfa signalling. Sci Rep 2019; 9:14677. [PMID: 31605007 PMCID: PMC6789122 DOI: 10.1038/s41598-019-50988-2] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2019] [Accepted: 09/17/2019] [Indexed: 01/09/2023] Open
Abstract
Primary aldosteronism (PA) is the most frequent form of secondary arterial hypertension. Mutations in different genes increase aldosterone production in PA, but additional mechanisms may contribute to increased cell proliferation and aldosterone producing adenoma (APA) development. We performed transcriptome analysis in APA and identified retinoic acid receptor alpha (RARα) signaling as a central molecular network involved in nodule formation. To understand how RARα modulates adrenal structure and function, we explored the adrenal phenotype of male and female Rarα knockout mice. Inactivation of Rarα in mice led to significant structural disorganization of the adrenal cortex in both sexes, with increased adrenal cortex size in female mice and increased cell proliferation in males. Abnormalities of vessel architecture and extracellular matrix were due to decreased Vegfa expression and modifications in extracellular matrix components. On the molecular level, Rarα inactivation leads to inhibition of non-canonical Wnt signaling, without affecting the canonical Wnt pathway nor PKA signaling. Our study suggests that Rarα contributes to the maintenance of normal adrenal cortex structure and cell proliferation, by modulating Wnt signaling. Dysregulation of this interaction may contribute to abnormal cell proliferation, creating a propitious environment for the emergence of specific driver mutations in PA.
Collapse
|
31
|
Bedoya M, Rinné S, Kiper AK, Decher N, González W, Ramírez D. TASK Channels Pharmacology: New Challenges in Drug Design. J Med Chem 2019; 62:10044-10058. [PMID: 31260312 DOI: 10.1021/acs.jmedchem.9b00248] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
Abstract
Rational drug design targeting ion channels is an exciting and always evolving research field. New medicinal chemistry strategies are being implemented to explore the wild chemical space and unravel the molecular basis of the ion channels modulators binding mechanisms. TASK channels belong to the two-pore domain potassium channel family and are modulated by extracellular acidosis. They are extensively distributed along the cardiovascular and central nervous systems, and their expression is up- and downregulated in different cancer types, which makes them an attractive therapeutic target. However, TASK channels remain unexplored, and drugs designed to target these channels are poorly selective. Here, we review TASK channels properties and their known blockers and activators, considering the new challenges in ion channels drug design and focusing on the implementation of computational methodologies in the drug discovery process.
Collapse
Affiliation(s)
- Mauricio Bedoya
- Centro de Bioinformática y Simulación Molecular (CBSM) , Universidad de Talca , 1 Poniente No. 1141 , 3460000 Talca , Chile
| | - Susanne Rinné
- Institute for Physiology and Pathophysiology, Vegetative Physiology and Marburg Center for Mind, Brain and Behavior, MCMBB , Philipps-University of Marburg , Deutschhausstraße 2 , Marburg 35037 , Germany
| | - Aytug K Kiper
- Institute for Physiology and Pathophysiology, Vegetative Physiology and Marburg Center for Mind, Brain and Behavior, MCMBB , Philipps-University of Marburg , Deutschhausstraße 2 , Marburg 35037 , Germany
| | - Niels Decher
- Institute for Physiology and Pathophysiology, Vegetative Physiology and Marburg Center for Mind, Brain and Behavior, MCMBB , Philipps-University of Marburg , Deutschhausstraße 2 , Marburg 35037 , Germany
| | - Wendy González
- Centro de Bioinformática y Simulación Molecular (CBSM) , Universidad de Talca , 1 Poniente No. 1141 , 3460000 Talca , Chile.,Millennium Nucleus of Ion Channels-Associated Diseases (MiNICAD) , Universidad de Talca , 1 Poniente No. 1141 , 3460000 Talca , Chile
| | - David Ramírez
- Instituto de Ciencias Biomédicas, Facultad de Ciencias de la Salud , Universidad Autónoma de Chile , El Llano Subercaseaux 2801, Piso 6 , 8900000 Santiago , Chile
| |
Collapse
|
32
|
Iqbal F, Thompson AJ, Riaz S, Pehar M, Rice T, Syed NI. Anesthetics: from modes of action to unconsciousness and neurotoxicity. J Neurophysiol 2019; 122:760-787. [PMID: 31242059 DOI: 10.1152/jn.00210.2019] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Modern anesthetic compounds and advanced monitoring tools have revolutionized the field of medicine, allowing for complex surgical procedures to occur safely and effectively. Faster induction times and quicker recovery periods of current anesthetic agents have also helped reduce health care costs significantly. Moreover, extensive research has allowed for a better understanding of anesthetic modes of action, thus facilitating the development of more effective and safer compounds. Notwithstanding the realization that anesthetics are a prerequisite to all surgical procedures, evidence is emerging to support the notion that exposure of the developing brain to certain anesthetics may impact future brain development and function. Whereas the data in support of this postulate from human studies is equivocal, the vast majority of animal research strongly suggests that anesthetics are indeed cytotoxic at multiple brain structure and function levels. In this review, we first highlight various modes of anesthetic action and then debate the evidence of harm from both basic science and clinical studies perspectives. We present evidence from animal and human studies vis-à-vis the possible detrimental effects of anesthetic agents on both the young developing and the elderly aging brain while discussing potential ways to mitigate these effects. We hope that this review will, on the one hand, invoke debate vis-à-vis the evidence of anesthetic harm in young children and the elderly, and on the other hand, incentivize the search for better and less toxic anesthetic compounds.
Collapse
Affiliation(s)
- Fahad Iqbal
- Vi Riddell Pain Program, Alberta Children's Hospital Research Institute, Hotchkiss Brain Institute, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
| | - Andrew J Thompson
- Vi Riddell Pain Program, Alberta Children's Hospital Research Institute, Hotchkiss Brain Institute, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada.,Department of Neuroscience, Faculty of Science, University of Calgary, Calgary, Alberta, Canada
| | - Saba Riaz
- Vi Riddell Pain Program, Alberta Children's Hospital Research Institute, Hotchkiss Brain Institute, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
| | - Marcus Pehar
- Vi Riddell Pain Program, Alberta Children's Hospital Research Institute, Hotchkiss Brain Institute, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
| | - Tiffany Rice
- Department of Anesthesiology, Perioperative and Pain Medicine, Alberta Children's Hospital, University of Calgary, Calgary, Alberta, Canada
| | - Naweed I Syed
- Vi Riddell Pain Program, Alberta Children's Hospital Research Institute, Hotchkiss Brain Institute, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
| |
Collapse
|
33
|
Seccia TM, Caroccia B, Gomez-Sanchez EP, Gomez-Sanchez CE, Rossi GP. The Biology of Normal Zona Glomerulosa and Aldosterone-Producing Adenoma: Pathological Implications. Endocr Rev 2018; 39:1029-1056. [PMID: 30007283 PMCID: PMC6236434 DOI: 10.1210/er.2018-00060] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/18/2018] [Accepted: 07/03/2018] [Indexed: 01/09/2023]
Abstract
The identification of several germline and somatic ion channel mutations in aldosterone-producing adenomas (APAs) and detection of cell clusters that can be responsible for excess aldosterone production, as well as the isolation of autoantibodies activating the angiotensin II type 1 receptor, have rapidly advanced the understanding of the biology of primary aldosteronism (PA), particularly that of APA. Hence, the main purpose of this review is to discuss how discoveries of the last decade could affect histopathology analysis and clinical practice. The structural remodeling through development and aging of the human adrenal cortex, particularly of the zona glomerulosa, and the complex regulation of aldosterone, with emphasis on the concepts of zonation and channelopathies, will be addressed. Finally, the diagnostic workup for PA and its subtyping to optimize treatment are reviewed.
Collapse
Affiliation(s)
- Teresa M Seccia
- Department of Medicine-DIMED, University of Padua, Padua PD, Italy
| | | | - Elise P Gomez-Sanchez
- Department of Pharmacology and Toxicology, G.V. (Sonny) Montgomery VA Medical Center, Jackson, Mississippi
| | - Celso E Gomez-Sanchez
- Division of Endocrinology, G.V. (Sonny) Montgomery VA Medical Center, Jackson, Mississippi.,University of Mississippi Medical Center, Jackson, Mississippi
| | - Gian Paolo Rossi
- Department of Medicine-DIMED, University of Padua, Padua PD, Italy
| |
Collapse
|
34
|
Inoue M, Matsuoka H, Lesage F, Harada K. Lack of p11 expression facilitates acidity‐sensing function of TASK1 channels in mouse adrenal medullary cells. FASEB J 2018; 33:455-468. [DOI: 10.1096/fj.201800407rr] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Affiliation(s)
- Masumi Inoue
- Department of Cell and Systems PhysiologySchool of MedicineUniversity of Occupational and Environmental Health Kitakyushu Japan
| | - Hidetada Matsuoka
- Department of Cell and Systems PhysiologySchool of MedicineUniversity of Occupational and Environmental Health Kitakyushu Japan
| | - Florian Lesage
- Université Côte d'AzurINSERMCentre National de la Recherche Scientifique (CNRS)Institut de Pharmacologie Moléculaire et CellulaireLaboratory of Excellence in Ion Channel Science and Therapeutics (LabEx ICST) Valbonne France
| | - Keita Harada
- Department of Cell and Systems PhysiologySchool of MedicineUniversity of Occupational and Environmental Health Kitakyushu Japan
| |
Collapse
|
35
|
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]
|
36
|
Lenzini L, Prisco S, Gallina M, Kuppusamy M, Rossi GP. Mutations of the Twik-Related Acid-Sensitive K+ Channel 2 Promoter in Human Primary Aldosteronism. Endocrinology 2018; 159:1352-1359. [PMID: 29293917 DOI: 10.1210/en.2017-03119] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/07/2017] [Accepted: 12/20/2017] [Indexed: 12/16/2022]
Abstract
Because blunted expression of the twik-related acid-sensitive K+ channel 2 (TASK-2) is a common feature of aldosterone-producing adenoma (APA) causing primary aldosteronism (PA), we sequenced the promoter region of the TASK-2 gene (KCNK5) in APAs (n = 76), primary hypertensive patients (n = 98), and 20-year-old healthy volunteers (n = 71), searching for variants that could affect expression of this channel. We found TASK-2 promoter mutations in 25% of the APAs: C999T in 6.6%, G595A in 5.3%, G36A in 5.3%, and C562T, Gins468, G265C, C1247T, G1140T, and C1399T in 1.3% each. The C999T mutation was found in only one of the 98 primary hypertensive patients, but mutations were detected also in 12% of volunteers: 4 carried the C999T, 3 G1288C, 1 the G1140T mutation, and 1 the 468ins mutation. After a 16-year follow-up, none of these patients developed hypertension or PA. The effect of C999T mutation was investigated in H295R cells using reporter vectors with the mutated or the wild-type (WT) TASK-2 promoters. TASK-2 gene expression was decreased by 31% ± 18% (P = 0.01) in mutated compared with WT APA. Likewise, in transfected H295R cells, the C999T mutation decreased TASK-2 transcriptional activity by 35% (normalized luciferase signal fold change: 0.65 ± 0.25, P < 0.001). Thus, mutations in the promoter region of the TASK-2 gene can account for the low expression in ∼25% of APAs. As they did not result in hypertension or PA during long-term follow-up in healthy participants, these mutations do not seem to be a factor in causing PA by themselves.
Collapse
Affiliation(s)
- Livia Lenzini
- Department of Medicine, Universita degli Studi di Padova, Padua, Italy
| | - Selene Prisco
- Department of Medicine, Universita degli Studi di Padova, Padua, Italy
| | - Michele Gallina
- Department of Military Legal Medicine, Centro Ospedaliero Militare di Taranto, Taranto, Italy
| | | | - Gian Paolo Rossi
- Department of Medicine, Universita degli Studi di Padova, Padua, Italy
| |
Collapse
|
37
|
Targeted disruption of the Kcnj5 gene in the female mouse lowers aldosterone levels. Clin Sci (Lond) 2018; 132:145-156. [PMID: 29222092 PMCID: PMC6365593 DOI: 10.1042/cs20171285] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2017] [Revised: 11/24/2017] [Accepted: 12/04/2017] [Indexed: 01/21/2023]
Abstract
Aldosterone is released from adrenal zona glomerulosa (ZG) cells and plays an important role in Na and K homoeostasis. Mutations in the human inwardly rectifying K channel CNJ type (KCNJ) 5 (KCNJ5) gene encoding the G-coupled inwardly rectifying K channel 4 (GIRK4) cause abnormal aldosterone secretion and hypertension. To better understand the role of wild-type (WT) GIRK4 in regulating aldosterone release, we have looked at aldosterone secretion in a Kcnj5 knockout (KO) mouse. We found that female but not male KO mice have reduced aldosterone levels compared with WT female controls, but higher levels of aldosterone after angiotensin II (Ang-II) stimulation. These differences could not be explained by sex differences in aldosterone synthase (Cyp11B2) gene expression in the mouse adrenal. Using RNAseq analysis to compare WT and KO adrenals, we showed that females also have a much larger set of differentially expressed adrenal genes than males (395 compared with 7). Ingenuity Pathway Analysis (IPA) of this gene set suggested that peroxisome proliferator activated receptor (PPAR) nuclear receptors regulated aldosterone production and altered signalling in the female KO mouse, which could explain the reduced aldosterone secretion. We tested this hypothesis in H295R adrenal cells and showed that the selective PPARα agonist fenofibrate can stimulate aldosterone production and induce Cyp11b2. Dosing mice in vivo produced similar results. Together our data show that Kcnj5 is important for baseline aldosterone secretion, but its importance is sex-limited at least in the mouse. It also highlights a novel regulatory pathway for aldosterone secretion through PPARα that may have translational potential in human hyperaldosteronism.
Collapse
|
38
|
Aragao-Santiago L, Gomez-Sanchez CE, Mulatero P, Spyroglou A, Reincke M, Williams TA. Mouse Models of Primary Aldosteronism: From Physiology to Pathophysiology. Endocrinology 2017; 158:4129-4138. [PMID: 29069360 PMCID: PMC5711388 DOI: 10.1210/en.2017-00637] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/14/2017] [Accepted: 10/16/2017] [Indexed: 01/08/2023]
Abstract
Primary aldosteronism (PA) is a common form of endocrine hypertension that is characterized by the excessive production of aldosterone relative to suppressed plasma renin levels. PA is usually caused by either a unilateral aldosterone-producing adenoma or bilateral adrenal hyperplasia. Somatic mutations have been identified in several genes that encode ion pumps and channels that may explain the aldosterone excess in over half of aldosterone-producing adenomas, whereas the pathophysiology of bilateral adrenal hyperplasia is largely unknown. A number of mouse models of hyperaldosteronism have been described that recreate some features of the human disorder, although none replicate the genetic basis of human PA. Animal models that reproduce the genotype-phenotype associations of human PA are required to establish the functional mechanisms that underlie the endocrine autonomy and deregulated cell growth of the affected adrenal and for preclinical studies of novel therapeutics. Herein, we discuss the differences in adrenal physiology across species and describe the genetically modified mouse models of PA that have been developed to date.
Collapse
Affiliation(s)
- Leticia Aragao-Santiago
- Medizinische Klinik und Poliklinik IV, Klinikum der Ludwig-Maximilians-Universität München, Germany
| | - Celso E Gomez-Sanchez
- Endocrinology Division, G.V. (Sonny) Montgomery Veterans Affairs Medical Center and University of Mississippi Medical Center
| | - Paolo Mulatero
- Division of Internal Medicine and Hypertension, Department of Medical Sciences, University of Turin, Italy
| | - Ariadni Spyroglou
- Medizinische Klinik und Poliklinik IV, Klinikum der Ludwig-Maximilians-Universität München, Germany
| | - Martin Reincke
- Medizinische Klinik und Poliklinik IV, Klinikum der Ludwig-Maximilians-Universität München, Germany
| | - Tracy Ann Williams
- Medizinische Klinik und Poliklinik IV, Klinikum der Ludwig-Maximilians-Universität München, Germany
- Division of Internal Medicine and Hypertension, Department of Medical Sciences, University of Turin, Italy
| |
Collapse
|
39
|
Boyer HG, Wils J, Renouf S, Arabo A, Duparc C, Boutelet I, Lefebvre H, Louiset E. Dysregulation of Aldosterone Secretion in Mast Cell–Deficient Mice. Hypertension 2017; 70:1256-1263. [DOI: 10.1161/hypertensionaha.117.09746] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2017] [Revised: 06/08/2017] [Accepted: 09/20/2017] [Indexed: 02/06/2023]
Affiliation(s)
- Hadrien-Gaël Boyer
- From the Normandie Université, UNIROUEN, INSERM U1239, Rouen, France (H.-G.B., J.W., S.R., C.D., I.B., H.L., E.L.); Department of Pharmacology, Institute for Biomedical Research, Rouen University Hospital, France (J.W.); Normandie Université, UNIROUEN, Rouen, France (A.A.); and Department of Endocrinology, Diabetes, and Metabolic Diseases, Institute for Research and Innovation in Biomedicine, University Hospital of Rouen, France (H.L.)
| | - Julien Wils
- From the Normandie Université, UNIROUEN, INSERM U1239, Rouen, France (H.-G.B., J.W., S.R., C.D., I.B., H.L., E.L.); Department of Pharmacology, Institute for Biomedical Research, Rouen University Hospital, France (J.W.); Normandie Université, UNIROUEN, Rouen, France (A.A.); and Department of Endocrinology, Diabetes, and Metabolic Diseases, Institute for Research and Innovation in Biomedicine, University Hospital of Rouen, France (H.L.)
| | - Sylvie Renouf
- From the Normandie Université, UNIROUEN, INSERM U1239, Rouen, France (H.-G.B., J.W., S.R., C.D., I.B., H.L., E.L.); Department of Pharmacology, Institute for Biomedical Research, Rouen University Hospital, France (J.W.); Normandie Université, UNIROUEN, Rouen, France (A.A.); and Department of Endocrinology, Diabetes, and Metabolic Diseases, Institute for Research and Innovation in Biomedicine, University Hospital of Rouen, France (H.L.)
| | - Arnaud Arabo
- From the Normandie Université, UNIROUEN, INSERM U1239, Rouen, France (H.-G.B., J.W., S.R., C.D., I.B., H.L., E.L.); Department of Pharmacology, Institute for Biomedical Research, Rouen University Hospital, France (J.W.); Normandie Université, UNIROUEN, Rouen, France (A.A.); and Department of Endocrinology, Diabetes, and Metabolic Diseases, Institute for Research and Innovation in Biomedicine, University Hospital of Rouen, France (H.L.)
| | - Céline Duparc
- From the Normandie Université, UNIROUEN, INSERM U1239, Rouen, France (H.-G.B., J.W., S.R., C.D., I.B., H.L., E.L.); Department of Pharmacology, Institute for Biomedical Research, Rouen University Hospital, France (J.W.); Normandie Université, UNIROUEN, Rouen, France (A.A.); and Department of Endocrinology, Diabetes, and Metabolic Diseases, Institute for Research and Innovation in Biomedicine, University Hospital of Rouen, France (H.L.)
| | - Isabelle Boutelet
- From the Normandie Université, UNIROUEN, INSERM U1239, Rouen, France (H.-G.B., J.W., S.R., C.D., I.B., H.L., E.L.); Department of Pharmacology, Institute for Biomedical Research, Rouen University Hospital, France (J.W.); Normandie Université, UNIROUEN, Rouen, France (A.A.); and Department of Endocrinology, Diabetes, and Metabolic Diseases, Institute for Research and Innovation in Biomedicine, University Hospital of Rouen, France (H.L.)
| | - Hervé Lefebvre
- From the Normandie Université, UNIROUEN, INSERM U1239, Rouen, France (H.-G.B., J.W., S.R., C.D., I.B., H.L., E.L.); Department of Pharmacology, Institute for Biomedical Research, Rouen University Hospital, France (J.W.); Normandie Université, UNIROUEN, Rouen, France (A.A.); and Department of Endocrinology, Diabetes, and Metabolic Diseases, Institute for Research and Innovation in Biomedicine, University Hospital of Rouen, France (H.L.)
| | - Estelle Louiset
- From the Normandie Université, UNIROUEN, INSERM U1239, Rouen, France (H.-G.B., J.W., S.R., C.D., I.B., H.L., E.L.); Department of Pharmacology, Institute for Biomedical Research, Rouen University Hospital, France (J.W.); Normandie Université, UNIROUEN, Rouen, France (A.A.); and Department of Endocrinology, Diabetes, and Metabolic Diseases, Institute for Research and Innovation in Biomedicine, University Hospital of Rouen, France (H.L.)
| |
Collapse
|
40
|
Olschewski A, Veale EL, Nagy BM, Nagaraj C, Kwapiszewska G, Antigny F, Lambert M, Humbert M, Czirják G, Enyedi P, Mathie A. TASK-1 (KCNK3) channels in the lung: from cell biology to clinical implications. Eur Respir J 2017; 50:50/5/1700754. [PMID: 29122916 DOI: 10.1183/13993003.00754-2017] [Citation(s) in RCA: 54] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2017] [Accepted: 08/05/2017] [Indexed: 12/18/2022]
Abstract
TWIK-related acid-sensitive potassium channel 1 (TASK-1 encoded by KCNK3) belongs to the family of two-pore domain potassium channels. This gene subfamily is constitutively active at physiological resting membrane potentials in excitable cells, including smooth muscle cells, and has been particularly linked to the human pulmonary circulation. TASK-1 channels are sensitive to a wide array of physiological and pharmacological mediators that affect their activity such as unsaturated fatty acids, extracellular pH, hypoxia, anaesthetics and intracellular signalling pathways. Recent studies show that modulation of TASK-1 channels, either directly or indirectly by targeting their regulatory mechanisms, has the potential to control pulmonary arterial tone in humans. Furthermore, mutations in KCNK3 have been identified as a rare cause of both familial and idiopathic pulmonary arterial hypertension. This review summarises our current state of knowledge of the functional role of TASK-1 channels in the pulmonary circulation in health and disease, with special emphasis on current advancements in the field.
Collapse
Affiliation(s)
- Andrea Olschewski
- Ludwig Boltzmann Institute for Lung Vascular Research Graz, Graz, Austria .,Institute of Physiology, Medical University of Graz, Graz, Austria
| | - Emma L Veale
- Medway School of Pharmacy, University of Kent, Central Avenue, Chatham Maritime, UK
| | - Bence M Nagy
- Institute of Physiology, Medical University of Graz, Graz, Austria
| | - Chandran Nagaraj
- Ludwig Boltzmann Institute for Lung Vascular Research Graz, Graz, Austria.,Institute of Physiology, Medical University of Graz, Graz, Austria
| | - Grazyna Kwapiszewska
- Ludwig Boltzmann Institute for Lung Vascular Research Graz, Graz, Austria.,Institute of Physiology, Medical University of Graz, Graz, Austria
| | - Fabrice Antigny
- Univ. Paris-Sud, Faculté de Médecine, Kremlin-Bicêtre, France.,AP-HP, Centre de Référence de l'Hypertension Pulmonaire Sévère, Département Hospitalo-Universitaire (DHU) Thorax Innovation, Service de Pneumologie et Réanimation Respiratoire, Hôpital de Bicêtre, Le Kremlin-Bicêtre, France.,UMRS 999, INSERM and Univ. Paris-Sud, Laboratoire d'Excellence (LabEx) en Recherche sur le Médicament et l'Innovation Thérapeutique (LERMIT), Hôpital-Marie-Lannelongue, Le Plessis Robinson, France
| | - Mélanie Lambert
- Univ. Paris-Sud, Faculté de Médecine, Kremlin-Bicêtre, France.,AP-HP, Centre de Référence de l'Hypertension Pulmonaire Sévère, Département Hospitalo-Universitaire (DHU) Thorax Innovation, Service de Pneumologie et Réanimation Respiratoire, Hôpital de Bicêtre, Le Kremlin-Bicêtre, France.,UMRS 999, INSERM and Univ. Paris-Sud, Laboratoire d'Excellence (LabEx) en Recherche sur le Médicament et l'Innovation Thérapeutique (LERMIT), Hôpital-Marie-Lannelongue, Le Plessis Robinson, France
| | - Marc Humbert
- Univ. Paris-Sud, Faculté de Médecine, Kremlin-Bicêtre, France.,AP-HP, Centre de Référence de l'Hypertension Pulmonaire Sévère, Département Hospitalo-Universitaire (DHU) Thorax Innovation, Service de Pneumologie et Réanimation Respiratoire, Hôpital de Bicêtre, Le Kremlin-Bicêtre, France.,UMRS 999, INSERM and Univ. Paris-Sud, Laboratoire d'Excellence (LabEx) en Recherche sur le Médicament et l'Innovation Thérapeutique (LERMIT), Hôpital-Marie-Lannelongue, Le Plessis Robinson, France
| | - Gábor Czirják
- Dept of Physiology, Semmelweis University, Budapest, Hungary
| | - Péter Enyedi
- Dept of Physiology, Semmelweis University, Budapest, Hungary
| | - Alistair Mathie
- Medway School of Pharmacy, University of Kent, Central Avenue, Chatham Maritime, UK
| |
Collapse
|
41
|
Sex-dependent differences in the in vivo respiratory phenotype of the TASK-1 potassium channel knockout mouse. Respir Physiol Neurobiol 2017; 245:13-28. [DOI: 10.1016/j.resp.2016.11.005] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2016] [Revised: 11/04/2016] [Accepted: 11/07/2016] [Indexed: 11/19/2022]
|
42
|
Infante M, Armani A, Mammi C, Fabbri A, Caprio M. Impact of Adrenal Steroids on Regulation of Adipose Tissue. Compr Physiol 2017; 7:1425-1447. [PMID: 28915330 DOI: 10.1002/cphy.c160037] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Corticosteroids are secreted by the adrenal glands and control the functions of adipose tissue via the activation of mineralocorticoid receptor (MR) and glucocorticoid receptor (GR). In turn, adipocytes release a large variety of adipokines into the bloodstream, regulating the function of several organs and tissues, including the adrenal glands, hereby controlling corticosteroid production. In adipose tissue, the activation of the MR by glucocorticoids (GC) and aldosterone affects important processes such as adipocyte differentiation, oxidative stress, autophagic flux, adipokine expression as well as local production of GC through upregulation of the enzyme 11β-hydroxysteroid dehydrogenase type 1 (11β-HSD1). Notably, the proinflammatory responses induced by the MR are counteracted by activation of the GR, whose activity inhibits the expression of inflammatory adipokines. Both GR and MR are deeply involved in adipogenesis and adipose expansion; hence pharmacological blockade of these two receptors has proven effective against adipose tissue dysfunction in experimental models of obesity and metabolic syndrome (MetS), suggesting a potential use for MR and GR antagonists in these clinical settings. Importantly, obesity and Cushing's syndrome (CS) share metabolic similarities and are characterized by high levels of circulating corticosteroids, which in turn are able to deeply affect adipose tissue. In addition, pharmacological approaches aimed at reducing aldosterone and GC levels, by means of the inhibition of CYP11B2 (aldosterone synthase) or 11β-HSD1, represent alternative strategies to counter the detrimental effects of excessive levels of corticosteroids, which are often observed in obesity and, more general, in MetS. © 2017 American Physiological Society. Compr Physiol 7:1425-1447, 2017.
Collapse
Affiliation(s)
- Marco Infante
- Unit of Endocrinology and Metabolic Diseases, Department of Systems Medicine, CTO A. Alesini Hospital, ASL Roma 2, University Tor Vergata, Rome, Italy
| | - Andrea Armani
- Laboratory of Cardiovascular Endocrinology, IRCCS San Raffaele Pisana, Rome, Italy
| | - Caterina Mammi
- Laboratory of Cardiovascular Endocrinology, IRCCS San Raffaele Pisana, Rome, Italy
| | - Andrea Fabbri
- Unit of Endocrinology and Metabolic Diseases, Department of Systems Medicine, CTO A. Alesini Hospital, ASL Roma 2, University Tor Vergata, Rome, Italy
| | - Massimiliano Caprio
- Laboratory of Cardiovascular Endocrinology, IRCCS San Raffaele Pisana, Rome, Italy
- Department of Human Sciences and Promotion of the Quality of Life, San Raffaele Roma Open University, Rome, Italy
| |
Collapse
|
43
|
Buehler PK, Bleiler D, Tegtmeier I, Heitzmann D, Both C, Georgieff M, Lesage F, Warth R, Thomas J. Abnormal respiration under hyperoxia in TASK-1/3 potassium channel double knockout mice. Respir Physiol Neurobiol 2017; 244:17-25. [PMID: 28673876 DOI: 10.1016/j.resp.2017.06.009] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2017] [Revised: 06/23/2017] [Accepted: 06/24/2017] [Indexed: 01/29/2023]
Abstract
Despite intensive research, the exact function of TASK potassium channels in central and peripheral chemoreception is still under debate. In this study, we investigated the respiration of unrestrained TASK-3 (TASK-3-/-) and TASK-1/TASK-3 double knockout (TASK-1/3-/-) adult male mice in vivo using a plethysmographic device. Ventilation parameters of TASK-3-/- mice were normal under control condition (21% O2) and upon hypoxia and hypercapnia they displayed the physiological increase of ventilation. TASK-1/3-/- mice showed increased ventilation under control conditions. This increase of ventilation was caused by increased tidal volumes (VT), a phenomenon similarly observed in TASK-1-/- mice. Under acute hypoxia, TASK-1/3-/- mice displayed the physiological increase of the minute volume. Interestingly, this increase was not related to an increase of the respiratory frequency (fR), as observed in wild-type mice, but was caused by a strong increase of VT. This particular respiratory phenotype is reminiscent of the respiratory phenotype of carotid body-denervated rodents in the compensated state. Acute hypercapnia (5% CO2) stimulated ventilation in TASK-1/3-/- and wild-type mice to a similar extent; however, at higher CO2 concentrations (>5% CO2) the stimulation of ventilation was more pronounced in TASK-1/3-/- mice. At hyperoxia (100% O2), TASK-1-/-, TASK-3-/- and wild-type mice showed the physiological small decrease of ventilation. In sharp contrast, TASK-1/3-/- mice exhibited an abnormal increase of ventilation under hyperoxia. In summary, these measurements showed a grossly normal respiration of TASK-3-/- mice and a respiratory phenotype of TASK-1/3-/- mice that was characterized by a markedly enhanced tidal volume, similar to the one observed in TASK-1-/- mice. The abnormal hyperoxia response, exclusively found in TASK-1/3-/- double mutant mice, indicates that both TASK-1 and TASK-3 are essential for the hyperoxia-induced hypoventilation. The peculiar respiratory phenotype of TASK-1/3 knockout mice is reminiscent of the respiration of animals with long-term carotid body dysfunction. Taken together, TASK-1 and TASK-3 appear to serve specific and distinct roles in the complex processes underlying chemoreception and respiratory control.
Collapse
Affiliation(s)
- Philipp K Buehler
- University Children's Hospital, Steinwiesstr. 75, CH-8032 Zürich, Switzerland
| | - Doris Bleiler
- Institute of Physiology, University of Regensburg, D-93053 Regensburg, Germany; Department of Anaesthesia, University Hospital Regensburg, 93042 Regensburg, Germany
| | - Ines Tegtmeier
- Institute of Physiology, University of Regensburg, D-93053 Regensburg, Germany
| | - Dirk Heitzmann
- Institute of Physiology, University of Regensburg, D-93053 Regensburg, Germany; University Medical Centre Mannheim, V. Medical Clinic, Theodor-Kutzer-Ufer 1-3, 68167 Mannheim, Germany
| | - Christian Both
- University Children's Hospital, Steinwiesstr. 75, CH-8032 Zürich, Switzerland
| | - Michael Georgieff
- Institute of Anesthesiology, University of Ulm, D-89081 Ulm, Germany
| | - Florian Lesage
- Université Côte d'Azur, Inserm, CNRS, Institut de Pharmacologie Moléculaire et Cellulaire, LabEx ICST, 660 Route des Lucioles, 06560, Valbonne, France
| | - Richard Warth
- Institute of Physiology, University of Regensburg, D-93053 Regensburg, Germany
| | - Jörg Thomas
- University Children's Hospital, Steinwiesstr. 75, CH-8032 Zürich, Switzerland.
| |
Collapse
|
44
|
Fernandes-Rosa FL, Boulkroun S, Zennaro MC. Somatic and inherited mutations in primary aldosteronism. J Mol Endocrinol 2017; 59:R47-R63. [PMID: 28400483 DOI: 10.1530/jme-17-0035] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/05/2017] [Accepted: 04/11/2017] [Indexed: 01/22/2023]
Abstract
Primary aldosteronism (PA), the most common form of secondary hypertension, is caused in the majority of cases by unilateral aldosterone-producing adenoma (APA) or bilateral adrenal hyperplasia. Over the past few years, somatic mutations in KCNJ5, CACNA1D, ATP1A1 and ATP2B3 have been proven to be associated with APA development, representing more than 50% of sporadic APA. The identification of these mutations has allowed the development of a model for APA involving modification on the intracellular ionic equilibrium and regulation of cell membrane potential, leading to autonomous aldosterone overproduction. Furthermore, somatic CTNNB1 mutations have also been identified in APA, but the link between these mutations and APA development remains unknown. The sequence of events responsible for APA formation is not completely understood, in particular, whether a single hit or a double hit is responsible for both aldosterone overproduction and cell proliferation. Germline mutations identified in patients with early-onset PA have expanded the classification of familial forms (FH) of PA. The description of germline KCNJ5 and CACNA1H mutations has identified FH-III and FH-IV based on genetic findings; germline CACNA1D mutations have been identified in patients with very early-onset PA and severe neurological abnormalities. This review summarizes current knowledge on the genetic basis of PA, the association of driver gene mutations and clinical findings and in the contribution to patient care, plus the current understanding on the mechanisms of APA development.
Collapse
Affiliation(s)
- Fabio Luiz Fernandes-Rosa
- INSERMUMRS_970, Paris Cardiovascular Research Center, Paris, France
- University Paris DescartesSorbonne Paris Cité, Paris, France
- Assistance Publique-Hôpitaux de ParisHôpital Européen Georges Pompidou, Service de Génétique, Paris, France
| | - Sheerazed Boulkroun
- INSERMUMRS_970, Paris Cardiovascular Research Center, Paris, France
- University Paris DescartesSorbonne Paris Cité, Paris, France
| | - Maria-Christina Zennaro
- INSERMUMRS_970, Paris Cardiovascular Research Center, Paris, France
- University Paris DescartesSorbonne Paris Cité, Paris, France
- Assistance Publique-Hôpitaux de ParisHôpital Européen Georges Pompidou, Service de Génétique, Paris, France
| |
Collapse
|
45
|
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
|
46
|
Yao J, McHedlishvili D, McIntire WE, Guagliardo NA, Erisir A, Coburn CA, Santarelli VP, Bayliss DA, Barrett PQ. Functional TASK-3-Like Channels in Mitochondria of Aldosterone-Producing Zona Glomerulosa Cells. Hypertension 2017. [PMID: 28630209 DOI: 10.1161/hypertensionaha.116.08871] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Ca2+ drives aldosterone synthesis in the cytosolic and mitochondrial compartments of the adrenal zona glomerulosa cell. Membrane potential across each of these compartments regulates the amplitude of the Ca2+ signal; yet, only plasma membrane ion channels and their role in regulating cell membrane potential have garnered investigative attention as pathological causes of human hyperaldosteronism. Previously, we reported that genetic deletion of TASK-3 channels (tandem pore domain acid-sensitive K+ channels) from mice produces aldosterone excess in the absence of a change in the cell membrane potential of zona glomerulosa cells. Here, we report using yeast 2-hybrid, immunoprecipitation, and electron microscopic analyses that TASK-3 channels are resident in mitochondria, where they regulate mitochondrial morphology, mitochondrial membrane potential, and aldosterone production. This study provides proof of principle that mitochondrial K+ channels, by modulating inner mitochondrial membrane morphology and mitochondrial membrane potential, have the ability to play a pathological role in aldosterone dysregulation in steroidogenic cells.
Collapse
Affiliation(s)
- Junlan Yao
- From the Departments of Pharmacology (J.Y., D.M., W.E.M., N.A.G., D.A.B., P.Q.B.) and Psychology (A.E.), University of Virginia School of Medicine, Charlottesville; Silverback Therapeutics, Inc, Seattle, WA (C.A.C.); and Department of Neuroscience, Merck & Co, Inc, West point, PA (V.P.S.)
| | - David McHedlishvili
- From the Departments of Pharmacology (J.Y., D.M., W.E.M., N.A.G., D.A.B., P.Q.B.) and Psychology (A.E.), University of Virginia School of Medicine, Charlottesville; Silverback Therapeutics, Inc, Seattle, WA (C.A.C.); and Department of Neuroscience, Merck & Co, Inc, West point, PA (V.P.S.)
| | - William E McIntire
- From the Departments of Pharmacology (J.Y., D.M., W.E.M., N.A.G., D.A.B., P.Q.B.) and Psychology (A.E.), University of Virginia School of Medicine, Charlottesville; Silverback Therapeutics, Inc, Seattle, WA (C.A.C.); and Department of Neuroscience, Merck & Co, Inc, West point, PA (V.P.S.)
| | - Nick A Guagliardo
- From the Departments of Pharmacology (J.Y., D.M., W.E.M., N.A.G., D.A.B., P.Q.B.) and Psychology (A.E.), University of Virginia School of Medicine, Charlottesville; Silverback Therapeutics, Inc, Seattle, WA (C.A.C.); and Department of Neuroscience, Merck & Co, Inc, West point, PA (V.P.S.)
| | - Alev Erisir
- From the Departments of Pharmacology (J.Y., D.M., W.E.M., N.A.G., D.A.B., P.Q.B.) and Psychology (A.E.), University of Virginia School of Medicine, Charlottesville; Silverback Therapeutics, Inc, Seattle, WA (C.A.C.); and Department of Neuroscience, Merck & Co, Inc, West point, PA (V.P.S.)
| | - Craig A Coburn
- From the Departments of Pharmacology (J.Y., D.M., W.E.M., N.A.G., D.A.B., P.Q.B.) and Psychology (A.E.), University of Virginia School of Medicine, Charlottesville; Silverback Therapeutics, Inc, Seattle, WA (C.A.C.); and Department of Neuroscience, Merck & Co, Inc, West point, PA (V.P.S.)
| | - Vincent P Santarelli
- From the Departments of Pharmacology (J.Y., D.M., W.E.M., N.A.G., D.A.B., P.Q.B.) and Psychology (A.E.), University of Virginia School of Medicine, Charlottesville; Silverback Therapeutics, Inc, Seattle, WA (C.A.C.); and Department of Neuroscience, Merck & Co, Inc, West point, PA (V.P.S.)
| | - Douglas A Bayliss
- From the Departments of Pharmacology (J.Y., D.M., W.E.M., N.A.G., D.A.B., P.Q.B.) and Psychology (A.E.), University of Virginia School of Medicine, Charlottesville; Silverback Therapeutics, Inc, Seattle, WA (C.A.C.); and Department of Neuroscience, Merck & Co, Inc, West point, PA (V.P.S.)
| | - Paula Q Barrett
- From the Departments of Pharmacology (J.Y., D.M., W.E.M., N.A.G., D.A.B., P.Q.B.) and Psychology (A.E.), University of Virginia School of Medicine, Charlottesville; Silverback Therapeutics, Inc, Seattle, WA (C.A.C.); and Department of Neuroscience, Merck & Co, Inc, West point, PA (V.P.S.).
| |
Collapse
|
47
|
Gomez-Sanchez CE, Kuppusamy M, Gomez-Sanchez EP. Of Mice and Man and the Regulation of Aldosterone Secretion. Hypertension 2017. [PMID: 28630207 DOI: 10.1161/hypertensionaha.117.09013] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Affiliation(s)
- Celso E Gomez-Sanchez
- From the Endocrine Section, G.V. (Sonny) Montgomery VA Medical Center (C.E.G.-S.), Division of Endocrinology (C.E.G.-S., M.K.), and Department of Pharmacology and Toxicology (E.P.G.-S.), University of Mississippi Medical Center, Jackson.
| | - Maniselvan Kuppusamy
- From the Endocrine Section, G.V. (Sonny) Montgomery VA Medical Center (C.E.G.-S.), Division of Endocrinology (C.E.G.-S., M.K.), and Department of Pharmacology and Toxicology (E.P.G.-S.), University of Mississippi Medical Center, Jackson
| | - Elise P Gomez-Sanchez
- From the Endocrine Section, G.V. (Sonny) Montgomery VA Medical Center (C.E.G.-S.), Division of Endocrinology (C.E.G.-S., M.K.), and Department of Pharmacology and Toxicology (E.P.G.-S.), University of Mississippi Medical Center, Jackson
| |
Collapse
|
48
|
Perez-Rivas LG, Rhayem Y, Sabrautzki S, Hantel C, Rathkolb B, Hrabě de Angelis M, Reincke M, Beuschlein F, Spyroglou A. Genetic characterization of a mouse line with primary aldosteronism. J Mol Endocrinol 2017; 58:67-78. [PMID: 27965370 DOI: 10.1530/jme-16-0200] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/05/2016] [Accepted: 12/13/2016] [Indexed: 12/17/2022]
Abstract
In an attempt to define novel genetic loci involved in the pathophysiology of primary aldosteronism, a mutagenesis screen after treatment with the alkylating agent N-ethyl-N-nitrosourea was established for the parameter aldosterone. One of the generated mouse lines with hyperaldosteronism was phenotypically and genetically characterized. This mouse line had high aldosterone levels but normal creatinine and urea values. The steroidogenic enzyme expression levels in the adrenal gland did not differ significantly among phenotypically affected and unaffected mice. Upon exome sequencing, point mutations were identified in seven candidate genes (Sspo, Dguok, Hoxaas2, Clstn3, Atm, Tipin and Mapk6). Subsequently, animals were stratified into wild-type and mutated groups according to their genotype for each of these candidate genes. A correlation of their genotypes with the respective aldosterone, aldosterone-to-renin ratio (ARR), urea and creatinine values as well as steroidogenic enzyme expression levels was performed. Aldosterone values were significantly higher in animals carrying mutations in four different genes (Sspo, Dguok, Hoxaas2 and Clstn3) and associated statistically significant adrenal Cyp11b2 overexpression as well as increased ARR was present only in mice with Sspo mutation. In contrast, mutations of the remaining candidate genes (Atm, Tipin and Mapk6) were associated with lower aldosterone values and lower Hsd3b6 expression levels. In summary, these data demonstrate association between the genes Sspo, Dguok, Hoxaas2 and Clstn3 and hyperaldosteronism. Final proofs for the causative nature of the mutations have to come from knock-out and knock-in experiments.
Collapse
Affiliation(s)
- L G Perez-Rivas
- Medizinische Klinik und Poliklinik IVEndocrine Research Unit, Klinikum der Universität München, LMU, Munich, Germany
| | - Y Rhayem
- Medizinische Klinik und Poliklinik IVEndocrine Research Unit, Klinikum der Universität München, LMU, Munich, Germany
| | - S Sabrautzki
- Helmholtz Zentrum MünchenGerman Research Center for Environmental Health (GmbH), Institute of Experimental Genetics and German Mouse Clinic, Neuherberg, Germany
- Helmholtz Zentrum MünchenGerman Research Center for Environmental Health (GmbH), Research Unit Comparative Medicine, Neuherberg, Germany
| | - C Hantel
- Medizinische Klinik und Poliklinik IVEndocrine Research Unit, Klinikum der Universität München, LMU, Munich, Germany
| | - B Rathkolb
- Helmholtz Zentrum MünchenGerman Research Center for Environmental Health (GmbH), Institute of Experimental Genetics and German Mouse Clinic, Neuherberg, Germany
- Member of German Center for Diabetes Research (DZD)Neuherberg, Germany
- Ludwig-Maximilians-Universität MünchenChair for Molecular Animal Breeding and Biotechnology, Gene Center of the München, Germany
| | - M Hrabě de Angelis
- Helmholtz Zentrum MünchenGerman Research Center for Environmental Health (GmbH), Institute of Experimental Genetics and German Mouse Clinic, Neuherberg, Germany
- Member of German Center for Diabetes Research (DZD)Neuherberg, Germany
- Lehrstuhl für Experimentelle GenetikTechnische Universität München, Freising-Weihenstephan, Germany
| | - M Reincke
- Medizinische Klinik und Poliklinik IVEndocrine Research Unit, Klinikum der Universität München, LMU, Munich, Germany
| | - F Beuschlein
- Medizinische Klinik und Poliklinik IVEndocrine Research Unit, Klinikum der Universität München, LMU, Munich, Germany
| | - A Spyroglou
- Medizinische Klinik und Poliklinik IVEndocrine Research Unit, Klinikum der Universität München, LMU, Munich, Germany
| |
Collapse
|
49
|
Franceschini N, Carty CL, Lu Y, Tao R, Sung YJ, Manichaikul A, Haessler J, Fornage M, Schwander K, Zubair N, Bien S, Hindorff LA, Guo X, Bielinski SJ, Ehret G, Kaufman JD, Rich SS, Carlson CS, Bottinger EP, North KE, Rao DC, Chakravarti A, Barrett PQ, Loos RJF, Buyske S, Kooperberg C. Variant Discovery and Fine Mapping of Genetic Loci Associated with Blood Pressure Traits in Hispanics and African Americans. PLoS One 2016; 11:e0164132. [PMID: 27736895 PMCID: PMC5063457 DOI: 10.1371/journal.pone.0164132] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2016] [Accepted: 09/12/2016] [Indexed: 01/11/2023] Open
Abstract
Despite the substantial burden of hypertension in US minority populations, few genetic studies of blood pressure have been conducted in Hispanics and African Americans, and it is unclear whether many of the established loci identified in European-descent populations contribute to blood pressure variation in non-European descent populations. Using the Metabochip array, we sought to characterize the genetic architecture of previously identified blood pressure loci, and identify novel cardiometabolic variants related to systolic and diastolic blood pressure in a multi-ethnic US population including Hispanics (n = 19,706) and African Americans (n = 18,744). Several known blood pressure loci replicated in African Americans and Hispanics. Fourteen variants in three loci (KCNK3, FGF5, ATXN2-SH2B3) were significantly associated with blood pressure in Hispanics. The most significant diastolic blood pressure variant identified in our analysis, rs2586886/KCNK3 (P = 5.2 x 10−9), also replicated in independent Hispanic and European-descent samples. African American and trans-ethnic meta-analysis data identified novel variants in the FGF5, ULK4 and HOXA-EVX1 loci, which have not been previously associated with blood pressure traits. Our identification and independent replication of variants in KCNK3, a gene implicated in primary hyperaldosteronism, as well as a variant in HOTTIP (HOXA-EVX1) suggest that further work to clarify the roles of these genes may be warranted. Overall, our findings suggest that loci identified in European descent populations also contribute to blood pressure variation in diverse populations including Hispanics and African Americans—populations that are understudied for hypertension genetic risk factors.
Collapse
Affiliation(s)
- Nora Franceschini
- Department of Epidemiology, University of North Carolina, Chapel Hill, North Carolina, United States of America
- * E-mail:
| | - Cara L. Carty
- Center for Translational Science, George Washington University and Children’s National Medical Center, Washington, District of Columbia, United States of America
| | - Yingchang Lu
- Genetics of Obesity and Related Metabolic Traits Program, The Charles Bronfman Institute for Personalized Medicine, Icahn School of Medicine at Mount Sinai, New York, New York, United States of America
| | - Ran Tao
- Department of Biostatistics, University of North Carolina, Chapel Hill, North Carolina, United States of America
| | - Yun Ju Sung
- Division of Biostatistics, Washington University, St. Louis, Missouri, United States of America
| | - Ani Manichaikul
- Center for Public Health Genomics, University of Virginia, Charlottesville, Virginia, 22908, United States of America
| | - Jeff Haessler
- Division of Public Health Sciences, Fred Hutchinson Cancer Research Center, Seattle, Washington, United States of America
| | - Myriam Fornage
- Brown Foundation Institute of Molecular Medicine & Human Genetics Center, University of Texas Health Science Center, Houston, Texas, United States of America
| | - Karen Schwander
- Division of Biostatistics, Washington University, St. Louis, Missouri, United States of America
| | - Niha Zubair
- Division of Public Health Sciences, Fred Hutchinson Cancer Research Center, Seattle, Washington, United States of America
| | - Stephanie Bien
- Division of Public Health Sciences, Fred Hutchinson Cancer Research Center, Seattle, Washington, United States of America
| | - Lucia A. Hindorff
- Division of Genomic Medicine, National Human Genome Research Institute, National Institutes of Health, Bethesda, Maryland, United States of America
| | - Xiuqing Guo
- Institute for Translational Genomics and Population Sciences and Department of Pediatrics, LABiomed at Harbor-University of California at Los Angeles Medical Center, Torrance, California, United States of America
| | - Suzette J. Bielinski
- Division of Epidemiology, Department of Health Sciences Research, Mayo Clinic, Rochester, Minnesota, United States of America
| | - Georg Ehret
- Center for Complex Disease Genomics, Johns Hopkins University School of Medicine, Baltimore, Maryland, United States of America
- Department of Specialties of Internal Medicine, Geneva University Hospital, Geneva, Switzerland
| | - Joel D. Kaufman
- Department of Environmental and Occupational Health Sciences, Epidemiology, and Medicine, University of Washington, Seattle, Washington, United States of America
| | - Stephen S. Rich
- Center for Public Health Genomics, University of Virginia, Charlottesville, Virginia, 22908, United States of America
| | - Christopher S. Carlson
- Division of Public Health Sciences, Fred Hutchinson Cancer Research Center, Seattle, Washington, United States of America
| | - Erwin P. Bottinger
- Genetics of Obesity and Related Metabolic Traits Program, The Charles Bronfman Institute for Personalized Medicine, Icahn School of Medicine at Mount Sinai, New York, New York, United States of America
| | - Kari E. North
- Department of Epidemiology, University of North Carolina, Chapel Hill, North Carolina, United States of America
| | - D. C. Rao
- Division of Biostatistics, Washington University, St. Louis, Missouri, United States of America
| | - Aravinda Chakravarti
- Center for Complex Disease Genomics, Johns Hopkins University School of Medicine, Baltimore, Maryland, United States of America
| | - Paula Q. Barrett
- Department of Pharmacology, University of Virginia, Charlottesville, Virginia, United States of America
| | - Ruth J. F. Loos
- Genetics of Obesity and Related Metabolic Traits Program, The Charles Bronfman Institute for Personalized Medicine, Icahn School of Medicine at Mount Sinai, New York, New York, United States of America
| | - Steven Buyske
- Department of Statistics and Biostatistics, Rutgers University, Piscataway, New Jersey, United States of America
| | - Charles Kooperberg
- Division of Public Health Sciences, Fred Hutchinson Cancer Research Center, Seattle, Washington, United States of America
| |
Collapse
|
50
|
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
|