1
|
Suntornlohanakul O, Ronchi CL, Arlt W, Prete A. Sexual dimorphism in benign adrenocortical tumours. Eur J Endocrinol 2025; 192:R1-R12. [PMID: 40296186 PMCID: PMC12068951 DOI: 10.1093/ejendo/lvaf088] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/14/2025] [Revised: 03/26/2025] [Accepted: 04/24/2025] [Indexed: 04/30/2025]
Abstract
Benign adrenocortical tumours are the most common adrenal neoplasms. Evidence over the past few decades has highlighted sex differences in their prevalence, clinical characteristics, and treatment outcomes. Cortisol-producing adenomas causing either Cushing's syndrome, particularly those with PRKACA or GNAS somatic mutations associated with a more severe phenotype, or mild autonomous cortisol secretion (MACS) are more commonly observed in women. The mechanisms underpinning this sexual dimorphism remain to be fully elucidated. Studies in mice have revealed a protective role of androgens in males, leading to a decelerated growth rate of adrenocortical cells. Furthermore, evidence from human adrenal tumour tissue suggests that oestrogen, progesterone, and luteinising hormone/choriogonadotropin signalling in the adrenal cortex may play a role in adrenal tumourigenesis and steroid production. Clinically, this is supported by the increased incidence of cortisol-producing adrenocortical adenomas or nodular hyperplasia during puberty, pregnancy, and menopause. Notably, women with MACS seem to be more vulnerable to the harmful effects of cortisol excess and carry a higher mortality risk than men. Women with aldosterone-producing adenomas have a higher prevalence of somatic KCNJ5 mutations than men, and patients harbouring these mutations are likely to have more favourable clinical outcomes after adrenalectomy. In this review, we summarise the possible mechanisms behind the sexual dimorphism of benign adrenocortical tumours and provide an up-to-date overview of the sex-specific differences in their prevalence, clinical presentation, and outcomes, focusing on cortisol and aldosterone excess. Considering sexual dimorphism is crucial to guide diagnosis and management, and to counsel these patients for optimised care.
Collapse
Affiliation(s)
- Onnicha Suntornlohanakul
- Endocrinology and Metabolism Unit, Division of Internal Medicine, Faculty of Medicine, Prince of Songkla University, Songkhla 90110, Thailand
- Department of Metabolism and Systems Science, College of Medicine and Health, University of Birmingham, Birmingham B152TT, United Kingdom
| | - Cristina L Ronchi
- Department of Metabolism and Systems Science, College of Medicine and Health, University of Birmingham, Birmingham B152TT, United Kingdom
- Centre for Endocrinology, Diabetes and Metabolism, Birmingham Health Partners, Birmingham B152TT, United Kingdom
- Department of Endocrinology, Queen Elizabeth Hospital Birmingham, University Hospitals Birmingham NHS Foundation Trust, Birmingham B152GW, United Kingdom
| | - Wiebke Arlt
- Medical Research Council Laboratory of Medical Sciences, London W120NN, United Kingdom
- Institute of Clinical Sciences, Imperial College London, London SW72AZ, United Kingdom
| | - Alessandro Prete
- Department of Metabolism and Systems Science, College of Medicine and Health, University of Birmingham, Birmingham B152TT, United Kingdom
- Centre for Endocrinology, Diabetes and Metabolism, Birmingham Health Partners, Birmingham B152TT, United Kingdom
- Department of Endocrinology, Queen Elizabeth Hospital Birmingham, University Hospitals Birmingham NHS Foundation Trust, Birmingham B152GW, United Kingdom
- National Institute for Health and Care Research (NIHR) Birmingham Biomedical Research Centre, University of Birmingham, University Hospitals Birmingham NHS Foundation Trust, Birmingham B152GW, United Kingdom
| |
Collapse
|
2
|
Zhong J, Paljor T, Yu S, Qiu L. Unmasking primary aldosteronism: Transforming disease management with advanced steroid profiling. Clin Chim Acta 2025; 570:120186. [PMID: 39933688 DOI: 10.1016/j.cca.2025.120186] [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: 12/05/2024] [Revised: 02/07/2025] [Accepted: 02/07/2025] [Indexed: 02/13/2025]
Abstract
Primary aldosteronism (PA) is traditionally thought to result from the overproduction of aldosterone which is unregulated by the renin-angiotensin system. It leads to a significantly increased risk of cardiovascular and metabolic complications as compared to primary hypertension. However, approximately one-third of patients diagnosed with PA according to the traditional guidelines have been found to exhibit low aldosterone levels and other steroid hormones may also play a potentially critical role in PA development. Early diagnosis of PA remains challenging due to low screening rates and the complex diagnostic procedures. The need for invasive adrenal vein sampling for PA subtyping also leads to a dilemma in therapeutic strategy selection. With advances in techniques, an increasing number of steroid hormones have been discovered to be associated with PA, potentially optimizing the PA diagnostic procedures. Herein, we review the cutting-edge advances in steroid hormones, including aberrant hormone synthesis and metabolism related to the pathophysiological development of PA, quantitative assays, and potential clinical value. Mass spectrometry provides a robust technical foundation for the simultaneous profiling of a panel of steroid hormones. Steroid hormone profiling combined with machine learning algorithms holds great research promise for facilitating early diagnosis and minimally invasive subtyping of PA. Thus, current progress and future expectations in combining steroid hormones with advanced technologies for early disease diagnosis and management are also reviewed. Research Agenda. 1) Identify changes in steroid hormones and the underlying biochemical mechanisms associated with PA. 2) Investigate the role of advanced mass spectrometry techniques in steroid hormone profiling. 3) Discuss current advances and future expectations in combining machine learning algorithms with MS-based steroid hormone profiling for PA's systematic and practical management.
Collapse
Affiliation(s)
- Jian Zhong
- Department of Laboratory Medicine, Peking Union Medical College Hospital, Peking Union Medical College & Chinese Academy of Medical Sciences, Beijing 100730 China
| | - Tashi Paljor
- Department of Laboratory Medicine, Peking Union Medical College Hospital, Peking Union Medical College & Chinese Academy of Medical Sciences, Beijing 100730 China
| | - SongLin Yu
- Department of Laboratory Medicine, Peking Union Medical College Hospital, Peking Union Medical College & Chinese Academy of Medical Sciences, Beijing 100730 China.
| | - Ling Qiu
- Department of Laboratory Medicine, Peking Union Medical College Hospital, Peking Union Medical College & Chinese Academy of Medical Sciences, Beijing 100730 China; State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Peking Union Medical College & Chinese Academy of Medical Sciences, Beijing 100730 China.
| |
Collapse
|
3
|
Chu Y, Setayesh J, Dumontet T, Krumeich L, Werner J, Moretti IF, De Sousa K, Kennedy C, La Pensee C, Lerario AM, Hammer GD. Adrenocortical stem cells in health and disease. Nat Rev Endocrinol 2025:10.1038/s41574-025-01091-2. [PMID: 40065108 DOI: 10.1038/s41574-025-01091-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 02/10/2025] [Indexed: 04/13/2025]
Abstract
The adrenal cortex is the major site of production of steroid hormones, which are essential for life. The normal development and homeostatic renewal of the adrenal cortex depend on capsular stem cells and cortical progenitor cells. These cell populations are highly plastic and support adaptation to physiological demands, injury and disease, linking steroid production and adrenal (organ) homeostasis with systemic endocrine cues and organismal homeostasis. This Review integrates findings from the past decade, outlining the mechanisms that govern the establishment and maintenance of the adrenal stem cell niche under different physiological and pathological conditions. The sophisticated regulation of the stem cell niche by gene regulatory networks, coordinated through paracrine and endocrine signalling, is highlighted in a context-dependent and sex-specific manner. We discuss how dysregulation of this intricate regulatory network is implicated in a wide range of adrenal diseases, and how emerging knowledge from adrenal stem cell research is inspiring the future development of gene-based and cell-based therapeutic strategies.
Collapse
Affiliation(s)
- Yulan Chu
- Department of Cell and Developmental Biology, University of Michigan, Ann Arbor, MI, USA
| | - Jordan Setayesh
- Medical Scientist Training Program, University of Michigan, Ann Arbor, MI, USA
| | - Typhanie Dumontet
- Department of Internal Medicine, Division of Metabolism, Endocrinology, and Diabetes, University of Michigan, Ann Arbor, MI, USA
| | - Lauren Krumeich
- Department of Surgery, University of Michigan, Ann Arbor, MI, USA
| | - Johanna Werner
- Department of Internal Medicine, Division of Metabolism, Endocrinology, and Diabetes, University of Michigan, Ann Arbor, MI, USA
- Division of Endocrinology and Diabetology, Department of Internal Medicine I, University Hospital of Wuerzburg, Wuerzburg, Germany
| | - Isabele F Moretti
- Department of Internal Medicine, Division of Metabolism, Endocrinology, and Diabetes, University of Michigan, Ann Arbor, MI, USA
| | - Kelly De Sousa
- Department of Internal Medicine, Division of Metabolism, Endocrinology, and Diabetes, University of Michigan, Ann Arbor, MI, USA
| | - Christopher Kennedy
- Department of Molecular and Integrative Physiology, University of Michigan, Ann Arbor, MI, USA
| | - Christopher La Pensee
- Department of Internal Medicine, Division of Metabolism, Endocrinology, and Diabetes, University of Michigan, Ann Arbor, MI, USA
| | - Antonio M Lerario
- Department of Internal Medicine, Division of Metabolism, Endocrinology, and Diabetes, University of Michigan, Ann Arbor, MI, USA
| | - Gary D Hammer
- Department of Cell and Developmental Biology, University of Michigan, Ann Arbor, MI, USA.
- Department of Internal Medicine, Division of Metabolism, Endocrinology, and Diabetes, University of Michigan, Ann Arbor, MI, USA.
- Department of Molecular and Integrative Physiology, University of Michigan, Ann Arbor, MI, USA.
- Endocrine Oncology Program, Rogel Comprehensive Cancer Center, University of Michigan, Ann Arbor, MI, USA.
| |
Collapse
|
4
|
Goodchild E, Wu X, Senanayake R, MacFarlane J, Argentesi G, Laycock K, Bashari WA, Cabrera CP, O'Toole SM, Salsbury J, Benu D, Lee YN, Chua ACN, Matson M, Koo B, Parvanta L, Hilliard N, Kosmoliaptsis V, Marker A, Berney DM, Drew K, Tan W, Foo R, Mein CA, Wozniak E, Kearney J, Savage E, Sahdev A, Bird N, Smith G, Hird M, Warnes V, Gillett D, Dawnay A, Adeyeye E, Aigbirhio F, McIntosh A, McConnachie A, Cruickshank JK, Cheow H, Gurnell M, Drake WM, Brown MJ. Molecular Imaging Versus Adrenal Vein Sampling for the Detection of Surgically Curable Primary Aldosteronism : A Prospective Within-Patient Trial. Ann Intern Med 2025; 178:336-347. [PMID: 40030172 DOI: 10.7326/annals-24-00761] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 03/26/2025] Open
Abstract
BACKGROUND Primary aldosteronism (PA) causes hypertension and is potentially surgically curable when it is caused by a unilateral aldosterone-producing adrenal adenoma (APA). Adrenal vein sampling (AVS) is required to guide surgery, but it is invasive, is technically difficult, and has limited availability. OBJECTIVE To determine whether the accuracy of post-dexamethasone [11C]metomidate ([11C]MTO) positron emission tomography-computed tomography, a diagnostic test for APAs, is superior or noninferior to the accuracy of AVS in predicting outcomes from unilateral adrenalectomy, and whether [11C]MTO is interchangeable with its longer-half-life analogue, para-chloro-2-[18F]fluoroethyletomidate ([18F]CETO). DESIGN Prospective within-patient comparison of diagnostic interventions. (ClinicalTrials.gov: NCT02945904). SETTING Three referral centers. PARTICIPANTS 174 patients with PA desiring surgery if a unilateral source of PA was diagnosed. INTERVENTION [11C]MTO and AVS in 169 patients, plus [18F]CETO in the final 31. MEASUREMENTS International consensus criteria for biochemical and clinical success at 6 and 24 months after surgery; κ statistic and Bland-Altman analyses comparing predictions of surgical outcomes by [11C]MTO and [18F]CETO. RESULTS Eighty-nine of 169 (52.7%), 78 of 169 (46.2%), and 43 of 169 (25.4%) patients were predicted to have unilateral PA by [11C]MTO, AVS, or both, respectively. One hundred of 169 (59.1%) were assigned to adrenalectomy by the multidisciplinary team; primary outcome data were available for 156 of 169. Predictions were most accurate for complete or partial biochemical success ([11C]MTO, 71.3% [95% CI, 61.0% to 80.1%]; AVS, 62.8% [CI, 52.2% to 72.6%]) and least accurate for complete clinical success (home blood pressure <135/85 mm Hg off treatment). The 95% CIs around differences between accuracies crossed zero, excluding superiority for [11C]MTO, but not the prespecified lower bound of -17%, allowing [11C]MTO to be declared noninferior to AVS. [18F]CETO and [11C]MTO agreed in 29 of 31 patients (κ = 0.85 [CI, 0.68 to 1.00]). LIMITATION The accuracy of [11C]MTO could be assessed only in the surgical group. CONCLUSION Molecular imaging is an accurate, noninvasive alternative to AVS. PRIMARY FUNDING SOURCE National Institute for Health and Care Research.
Collapse
Affiliation(s)
- Emily Goodchild
- Clinical Pharmacology and Precision Medicine, William Harvey Research Institute, Queen Mary University of London; NIHR Barts Cardiovascular Biomedical Research Centre, Barts and The London School of Medicine and Dentistry, Queen Mary University of London; and Department of Endocrinology, St. Bartholomew's Hospital, Barts Health NHS Trust, London, United Kingdom (E.G., X.W., G.A., K.L., Y.-N.L., J.K., W.M.D., M.J.B.)
| | - Xilin Wu
- Clinical Pharmacology and Precision Medicine, William Harvey Research Institute, Queen Mary University of London; NIHR Barts Cardiovascular Biomedical Research Centre, Barts and The London School of Medicine and Dentistry, Queen Mary University of London; and Department of Endocrinology, St. Bartholomew's Hospital, Barts Health NHS Trust, London, United Kingdom (E.G., X.W., G.A., K.L., Y.-N.L., J.K., W.M.D., M.J.B.)
| | - Russell Senanayake
- Metabolic Research Laboratories, Institute of Metabolic Science, University of Cambridge; NIHR Cambridge Biomedical Research Centre, Cambridge Biomedical Campus; and Department of Diabetes and Endocrinology, Addenbrooke's Hospital, Cambridge University Hospitals NHS Foundation Trust, Cambridge, United Kingdom (R.S., J.M., W.A.B., M.G.)
| | - James MacFarlane
- Metabolic Research Laboratories, Institute of Metabolic Science, University of Cambridge; NIHR Cambridge Biomedical Research Centre, Cambridge Biomedical Campus; and Department of Diabetes and Endocrinology, Addenbrooke's Hospital, Cambridge University Hospitals NHS Foundation Trust, Cambridge, United Kingdom (R.S., J.M., W.A.B., M.G.)
| | - Giulia Argentesi
- Clinical Pharmacology and Precision Medicine, William Harvey Research Institute, Queen Mary University of London; NIHR Barts Cardiovascular Biomedical Research Centre, Barts and The London School of Medicine and Dentistry, Queen Mary University of London; and Department of Endocrinology, St. Bartholomew's Hospital, Barts Health NHS Trust, London, United Kingdom (E.G., X.W., G.A., K.L., Y.-N.L., J.K., W.M.D., M.J.B.)
| | - Kate Laycock
- Clinical Pharmacology and Precision Medicine, William Harvey Research Institute, Queen Mary University of London; NIHR Barts Cardiovascular Biomedical Research Centre, Barts and The London School of Medicine and Dentistry, Queen Mary University of London; and Department of Endocrinology, St. Bartholomew's Hospital, Barts Health NHS Trust, London, United Kingdom (E.G., X.W., G.A., K.L., Y.-N.L., J.K., W.M.D., M.J.B.)
| | - Waiel A Bashari
- Metabolic Research Laboratories, Institute of Metabolic Science, University of Cambridge; NIHR Cambridge Biomedical Research Centre, Cambridge Biomedical Campus; and Department of Diabetes and Endocrinology, Addenbrooke's Hospital, Cambridge University Hospitals NHS Foundation Trust, Cambridge, United Kingdom (R.S., J.M., W.A.B., M.G.)
| | - Claudia P Cabrera
- Clinical Pharmacology and Precision Medicine, William Harvey Research Institute, Queen Mary University of London; NIHR Barts Cardiovascular Biomedical Research Centre, Barts and The London School of Medicine and Dentistry, Queen Mary University of London; and Centre for Translational Bioinformatics, William Harvey Research Institute, Queen Mary University of London, London, United Kingdom (C.P.C.)
| | - Samuel M O'Toole
- Clinical Pharmacology and Precision Medicine, William Harvey Research Institute, Queen Mary University of London, London, United Kingdom; NIHR Barts Cardiovascular Biomedical Research Centre, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London, United Kingdom; Department of Endocrinology, St. Bartholomew's Hospital, Barts Health NHS Trust, London, United Kingdom; and Department of Endocrinology, The Royal Hallamshire Hospital, Sheffield, United Kingdom (S.M.O.)
| | - Jackie Salsbury
- Clinical Pharmacology and Precision Medicine, William Harvey Research Institute, Queen Mary University of London, and NIHR Barts Cardiovascular Biomedical Research Centre, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London, United Kingdom (J.S., D.B., K.D.)
| | - Daniela Benu
- Clinical Pharmacology and Precision Medicine, William Harvey Research Institute, Queen Mary University of London, and NIHR Barts Cardiovascular Biomedical Research Centre, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London, United Kingdom (J.S., D.B., K.D.)
| | - Yun-Ni Lee
- Clinical Pharmacology and Precision Medicine, William Harvey Research Institute, Queen Mary University of London; NIHR Barts Cardiovascular Biomedical Research Centre, Barts and The London School of Medicine and Dentistry, Queen Mary University of London; and Department of Endocrinology, St. Bartholomew's Hospital, Barts Health NHS Trust, London, United Kingdom (E.G., X.W., G.A., K.L., Y.-N.L., J.K., W.M.D., M.J.B.)
| | - Aldons C N Chua
- Department of Endocrinology, St. Bartholomew's Hospital, Barts Health NHS Trust, London, United Kingdom (A.C.N.C., L.P.)
| | - Matthew Matson
- Department of Radiology, St. Bartholomew's Hospital, Barts Health NHS Trust, London, United Kingdom (M.M., A.S.)
| | - Brendan Koo
- Department of Radiology, Addenbrooke's Hospital, Cambridge University Hospitals NHS Foundation Trust, Cambridge, United Kingdom (B.K., N.H., N.B.)
| | - Laila Parvanta
- Department of Endocrinology, St. Bartholomew's Hospital, Barts Health NHS Trust, London, United Kingdom (A.C.N.C., L.P.)
| | - Nicholas Hilliard
- Department of Radiology, Addenbrooke's Hospital, Cambridge University Hospitals NHS Foundation Trust, Cambridge, United Kingdom (B.K., N.H., N.B.)
| | - Vasilis Kosmoliaptsis
- Department of Surgery, Addenbrooke's Hospital, Cambridge University Hospitals NHS Foundation Trust, Cambridge, United Kingdom (V.K.)
| | - Alison Marker
- Department of Histopathology, Addenbrooke's Hospital, Cambridge University Hospitals NHS Foundation Trust, Cambridge, United Kingdom (A.Marker)
| | - Daniel M Berney
- Department of Cellular Pathology, Royal London Hospital, Barts Health NHS Trust, London, United Kingdom (D.M.B.)
| | - Kiera Drew
- Clinical Pharmacology and Precision Medicine, William Harvey Research Institute, Queen Mary University of London, and NIHR Barts Cardiovascular Biomedical Research Centre, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London, United Kingdom (J.S., D.B., K.D.)
| | - Wilson Tan
- Cardiovascular Research Institute, National University of Singapore, Singapore (W.T., R.F.)
| | - Roger Foo
- Cardiovascular Research Institute, National University of Singapore, Singapore (W.T., R.F.)
| | - Charles A Mein
- Barts and London Genome Centre, School of Medicine and Dentistry, Blizard Institute, London, United Kingdom (C.A.M., E.W., E.S.)
| | - Eva Wozniak
- Barts and London Genome Centre, School of Medicine and Dentistry, Blizard Institute, London, United Kingdom (C.A.M., E.W., E.S.)
| | - Jessica Kearney
- Clinical Pharmacology and Precision Medicine, William Harvey Research Institute, Queen Mary University of London; NIHR Barts Cardiovascular Biomedical Research Centre, Barts and The London School of Medicine and Dentistry, Queen Mary University of London; and Department of Endocrinology, St. Bartholomew's Hospital, Barts Health NHS Trust, London, United Kingdom (E.G., X.W., G.A., K.L., Y.-N.L., J.K., W.M.D., M.J.B.)
| | - Emanuel Savage
- Barts and London Genome Centre, School of Medicine and Dentistry, Blizard Institute, London, United Kingdom (C.A.M., E.W., E.S.)
| | - Anju Sahdev
- Department of Radiology, St. Bartholomew's Hospital, Barts Health NHS Trust, London, United Kingdom (M.M., A.S.)
| | - Nicholas Bird
- Department of Radiology, Addenbrooke's Hospital, Cambridge University Hospitals NHS Foundation Trust, Cambridge, United Kingdom (B.K., N.H., N.B.)
| | - Graham Smith
- Wolfson Brain Imaging Centre, University of Cambridge, Cambridge, United Kingdom (G.S., M.H., F.A.)
| | - Matthew Hird
- Wolfson Brain Imaging Centre, University of Cambridge, Cambridge, United Kingdom (G.S., M.H., F.A.)
| | - Victoria Warnes
- Department of Nuclear Medicine, Addenbrooke's Hospital, Cambridge University Hospitals NHS Foundation Trust, Cambridge, United Kingdom (V.W., D.G.)
| | - Daniel Gillett
- Department of Nuclear Medicine, Addenbrooke's Hospital, Cambridge University Hospitals NHS Foundation Trust, Cambridge, United Kingdom (V.W., D.G.)
| | - Anne Dawnay
- Department of Clinical Biochemistry, St. Bartholomew's Hospital, Barts Health NHS Trust, London, United Kingdom (A.D.)
| | - Elizabeth Adeyeye
- Department of Clinical Pharmacology, Guy's and St. Thomas' NHS Foundation Trust, London, United Kingdom (E.A.)
| | - Franklin Aigbirhio
- Wolfson Brain Imaging Centre, University of Cambridge, Cambridge, United Kingdom (G.S., M.H., F.A.)
| | - Alasdair McIntosh
- Robertson Centre for Biostatistics, School of Health and Wellbeing, University of Glasgow, Glasgow, United Kingdom (A.McIntosh, A.McConnachie)
| | - Alex McConnachie
- Robertson Centre for Biostatistics, School of Health and Wellbeing, University of Glasgow, Glasgow, United Kingdom (A.McIntosh, A.McConnachie)
| | - J Kennedy Cruickshank
- Department of Clinical Pharmacology, Guy's and St. Thomas' NHS Foundation Trust, and School of Life-Course/Nutritional Sciences, King's College London, London, United Kingdom (J.K.J.C.)
| | - Heok Cheow
- Department of Radiology, Addenbrooke's Hospital, Cambridge University Hospitals NHS Foundation Trust, and Department of Nuclear Medicine, Addenbrooke's Hospital, Cambridge University Hospitals NHS Foundation Trust, Cambridge, United Kingdom (H.C.)
| | - Mark Gurnell
- Metabolic Research Laboratories, Institute of Metabolic Science, University of Cambridge; NIHR Cambridge Biomedical Research Centre, Cambridge Biomedical Campus; and Department of Diabetes and Endocrinology, Addenbrooke's Hospital, Cambridge University Hospitals NHS Foundation Trust, Cambridge, United Kingdom (R.S., J.M., W.A.B., M.G.)
| | - William M Drake
- Department of Radiology, Addenbrooke's Hospital, Cambridge University Hospitals NHS Foundation Trust, and Department of Nuclear Medicine, Addenbrooke's Hospital, Cambridge University Hospitals NHS Foundation Trust, Cambridge, United Kingdom (H.C.)
| | - Morris J Brown
- Clinical Pharmacology and Precision Medicine, William Harvey Research Institute, Queen Mary University of London; NIHR Barts Cardiovascular Biomedical Research Centre, Barts and The London School of Medicine and Dentistry, Queen Mary University of London; and Department of Endocrinology, St. Bartholomew's Hospital, Barts Health NHS Trust, London, United Kingdom (E.G., X.W., G.A., K.L., Y.-N.L., J.K., W.M.D., M.J.B.)
| |
Collapse
|
5
|
Gao Y, Wang Y, Zhou Y, Ding J, Chang X, Qiu L, Huo L, Wen J, Zhang Y, Tong A. Two-hit model for the development of aldosterone-producing adenoma: supporting from two new cases. J Hypertens 2025; 43:544-548. [PMID: 39445622 DOI: 10.1097/hjh.0000000000003888] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2024] [Accepted: 09/10/2024] [Indexed: 10/25/2024]
Abstract
Recently, a two-hit model for the development of aldosterone-producing adenoma (APA) was proposed but until now, only two cases supporting the model have been reported. Here, we present two new cases of primary aldosteronism (PA), both of which had large functional adenomas with somatic mutations in aldosterone-driving genes. Furthermore, the first patient, who had a history of colorectal cancer, was found to have a germline and an additional somatic mutation in APC , and APC inactivation was confirmed by immunohistochemistry. The other patient had pathogenic somatic mutation in CTNNB1 . These pro-proliferation mutations resulted in abnormal activation of the Wnt/β-catenin pathway. Two consecutive events apparent in these patients, namely, the first event leading to cell proliferation and the second driving hormonal hypersecretion, supported the two-hit model of APA development. The two-hit model usually occurs in the larger adenomas, and the driving factors of the first hit that promote cell proliferation still require further research and exploration.
Collapse
Affiliation(s)
- Yinjie Gao
- NHC Key Laboratory of Endocrinology, Department of Endocrinology, Peking Union Medical College Hospital, Peking Union Medical College, Chinese Academy of Medical Sciences
- Beijing Tsinghua Changgung Hospital, School of Clinical Medicine, Tsinghua University, Beijing, China
| | - Yu Wang
- NHC Key Laboratory of Endocrinology, Department of Endocrinology, Peking Union Medical College Hospital, Peking Union Medical College, Chinese Academy of Medical Sciences
| | - Yue Zhou
- NHC Key Laboratory of Endocrinology, Department of Endocrinology, Peking Union Medical College Hospital, Peking Union Medical College, Chinese Academy of Medical Sciences
| | - Jie Ding
- Department of Nuclear Medicine, Beijing Key Laboratory of Molecular Targeted Diagnosis and Therapy in Nuclear Medicine and State Key Laboratory of Complex Severe and Rare Diseases
| | | | - Ling Qiu
- State Key Laboratory of Complex Severe and Rare Diseases, Department of Clinical Laboratory
| | - Li Huo
- Department of Nuclear Medicine, Beijing Key Laboratory of Molecular Targeted Diagnosis and Therapy in Nuclear Medicine and State Key Laboratory of Complex Severe and Rare Diseases
| | - Jin Wen
- Department of Urology, Peking Union Medical College Hospital, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, China
| | - Yushi Zhang
- Department of Urology, Peking Union Medical College Hospital, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, China
| | - Anli Tong
- NHC Key Laboratory of Endocrinology, Department of Endocrinology, Peking Union Medical College Hospital, Peking Union Medical College, Chinese Academy of Medical Sciences
| |
Collapse
|
6
|
Glinicki P, Sawicka-Gutaj N, Araujo-Castro M. Editorial: Advances in diagnostics and management of adrenal tumors. Front Endocrinol (Lausanne) 2025; 16:1543773. [PMID: 39950027 PMCID: PMC11821480 DOI: 10.3389/fendo.2025.1543773] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/11/2024] [Accepted: 01/06/2025] [Indexed: 02/16/2025] Open
Affiliation(s)
- Piotr Glinicki
- EndoLab Laboratory, Centre of Postgraduate Medical Education, Warsaw, Poland
- Department of Endocrinology, Centre of Postgraduate Medical Education, Warsaw, Poland
| | - Nadia Sawicka-Gutaj
- Department of Endocrinology, Metabolism and Internal Medicine, Poznań University of Medical Sciences, Poznań, Poland
| | - Marta Araujo-Castro
- Endocrinology & Nutrition Department, Hospital Universitario Ramón y Cajal, Madrid, Spain
- Instituto de Investigación Biomédica Ramón y Cajal (IRYCIS), Madrid, Spain
| |
Collapse
|
7
|
Watson A, Syme H, Brown M. Somatic GNAQ, CTNNB1, and CACNA1C Mutations in Cat Aldosterone-Secreting Tumors. Hypertension 2024; 81:2489-2500. [PMID: 39429164 DOI: 10.1161/hypertensionaha.124.23501] [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: 06/18/2024] [Accepted: 10/04/2024] [Indexed: 10/22/2024]
Abstract
BACKGROUND Primary aldosteronism (PA) is a common cause of human hypertension. Somatic mutations in KCNJ5, CACNA1D, ATP1A1, and ATP2B3 are found in at least 80% of aldosterone-producing adenomas, which cause unilateral PA in humans. Somatic mutations have been identified infrequently in 7 other genes; few of these were known to play a role in aldosterone secretion before the discovery of their mutations. Interrogating somatic mutations in the domestic cat, in which spontaneous PA is also known to occur, might improve the understanding of normal adrenal gland physiology and the pathophysiology of PA. METHODS DNA and RNA extracted from tissue from 13 cats with unilateral aldosterone-secreting tumors, including 8 carcinomas and 5 adenomas, underwent whole genome sequencing, targeted Sanger sequencing, and RNA sequencing. Single-nucleotide substitution variants were filtered to select those with a predicted deleterious effect on protein function and a suspected role in aldosterone secretion. RESULTS Probable functional somatic single-nucleotide polymorphisms (n=8) were found in 3 adenomas and 2 carcinomas. Mutations with predicted significant effects were identified in 2 genes also mutated in human PA; GNAQ and CTNNB1, and in a residue of CACNA1C analogous to a common CACNA1D mutation. In contrast to humans, CACNA1C expression was much greater than CACNA1D in both feline tumor and nontumor adrenal tissue. No mutations were identified in KCNJ5, CACNA1D, ATP1A1, or ATP2B3. CONCLUSIONS Similar mutations were identified in cats to those found in humans. It is, therefore, likely that both species have shared underlying selection pressures for mutations that increase aldosterone secretion.
Collapse
Affiliation(s)
- Alice Watson
- Clinical Science and Services, Royal Veterinary College, London, United Kingdom (A.W., H.S.)
- Clinical Pharmacology and Precision Medicine, Queen Mary University of London, United Kingdom (A.W., M.B.)
| | - Harriet Syme
- Clinical Science and Services, Royal Veterinary College, London, United Kingdom (A.W., H.S.)
| | - Morris Brown
- Clinical Pharmacology and Precision Medicine, Queen Mary University of London, United Kingdom (A.W., M.B.)
| |
Collapse
|
8
|
Lacroix A, Bourdeau I, Chasseloup F, Kamenický P, Lopez AG, Louiset E, Lefebvre H. Aberrant hormone receptors regulate a wide spectrum of endocrine tumors. Lancet Diabetes Endocrinol 2024; 12:837-855. [PMID: 39326429 DOI: 10.1016/s2213-8587(24)00200-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/27/2024] [Revised: 06/27/2024] [Accepted: 07/11/2024] [Indexed: 09/28/2024]
Abstract
Aberrant G-protein coupled receptor (GPCR) expression is highly prevalent in cortisol-secreting primary bilateral macronodular adrenal hyperplasia (PBMAH) and unilateral adenomas. The aberrant expression of diverse GPCRs and their ligands play an important role in the over-function of various endocrine tumours. Examples include aberrant expression of MC2R, 5-HT4R, AVPR1A, LHCGR, and GnRHR in primary aldosteronism; GCGR, LHCGR, and 5-HT4R in phaeochromocytomas and paragangliomas; TRHR, GnRHR, GIPR, and GRP101 in pituitary somatotroph tumours; AVPR2, D2DR, and SSTR5 in pituitary corticotroph tumours; GLP1R, GIPR, and somatostatin receptors in medullary thyroid carcinoma; and SSTRs, GLP1R, and GIPR in other neuroendocrine tumours. The genetic mechanisms causing the ectopic expression of GIPR in cortisol-secreting PBMAHs and unilateral adenomas have been identified, but distinct mechanisms are implicated in other endocrine tumours. Development of functional imaging targeting aberrant GPCRs should be useful for identification and for specific therapies of this wide spectrum of tumours. The aim of this review is to show that the regulation of endocrine tumours by aberrant GPCR is not restricted to cortisol-secreting adrenal lesions, but also occurs in tumours of several other organs.
Collapse
Affiliation(s)
- André Lacroix
- Division of Endocrinology, Department of Medicine and Research Center, Centre hospitalier de l' Université de Montréal (CHUM), Montréal, QC, Canada.
| | - Isabelle Bourdeau
- Division of Endocrinology, Department of Medicine and Research Center, Centre hospitalier de l' Université de Montréal (CHUM), Montréal, QC, Canada
| | - Fanny Chasseloup
- Université Paris-Saclay, Inserm, Physiologie et Physiopathologie Endocriniennes, Assistance Publique-Hôpitaux de Paris, Hôpital Bicêtre, Service d'Endocrinologie et des Maladies de la Reproduction, Le Kremlin-Bicêtre, France
| | - Peter Kamenický
- Université Paris-Saclay, Inserm, Physiologie et Physiopathologie Endocriniennes, Assistance Publique-Hôpitaux de Paris, Hôpital Bicêtre, Service d'Endocrinologie et des Maladies de la Reproduction, Le Kremlin-Bicêtre, France
| | - Antoine-Guy Lopez
- Univ Rouen Normandie, Inserm, NorDiC UMR 1239, Rouen, France; Department of Endocrinology, Diabetes and Metabolic Diseases, Rouen, France
| | - Estelle Louiset
- Univ Rouen Normandie, Inserm, NorDiC UMR 1239, Rouen, France; Department of Endocrinology, Diabetes and Metabolic Diseases, Rouen, France
| | - Hervé Lefebvre
- Univ Rouen Normandie, Inserm, NorDiC UMR 1239, Rouen, France; Department of Endocrinology, Diabetes and Metabolic Diseases, Rouen, France
| |
Collapse
|
9
|
Pilch J, Mizera J, Tota M, Donizy P. GNAQ/GNA11-Related Benign and Malignant Entities-A Common Histoembriologic Origin or a Tissue-Dependent Coincidence. Cancers (Basel) 2024; 16:3672. [PMID: 39518110 PMCID: PMC11544895 DOI: 10.3390/cancers16213672] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2024] [Revised: 10/24/2024] [Accepted: 10/29/2024] [Indexed: 11/16/2024] Open
Abstract
Uveal melanoma (UM), recognized as the most prevalent primary intraocular malignancy in adults, is primarily driven by mutations in the GNAQ and GNA11 genes. These genetic alterations are also implicated in other conditions, which exhibit distinct morphological characteristics. In this article, we investigate the role of GNAQ and GNA11 mutations across varied disorders (e.g., UM, skin blue nevi, and hemangiomas), emphasizing the shared pathogenic mechanisms that connect them despite their differing clinical manifestations. By investigating the molecular pathways affected by these mutations, we provide insights into the potential for targeted therapies that could address not only UM but also other disorders associated with GNAQ/GNA11 mutations. Moreover, we discuss the role of SOX10-positive perivascular cells that may be implicated in the complex pathophysiology of GNAQ/GNA11-related entities. Understanding the common molecular foundation of these conditions opens new ways for research and treatment opportunities, potentially leading to more effective, personalized therapeutic strategies.
Collapse
Affiliation(s)
- Justyna Pilch
- Department of Clinical and Experimental Pathology, Wroclaw Medical University, 50-556 Wroclaw, Poland
| | - Jakub Mizera
- Department of Clinical and Experimental Pathology, Wroclaw Medical University, 50-556 Wroclaw, Poland
| | - Maciej Tota
- Department of Clinical and Experimental Pathology, Wroclaw Medical University, 50-556 Wroclaw, Poland
| | - Piotr Donizy
- Department of Clinical and Experimental Pathology, Wroclaw Medical University, 50-556 Wroclaw, Poland
- Department of Pathology and Clinical Cytology, Jan Mikulicz-Radecki University Hospital, 50-556 Wroclaw, Poland
| |
Collapse
|
10
|
van Rooyen D, Bandulik S, Coon G, Laukemper M, Kumar-Sinha C, Udager AM, Lee C, Wachtel H, Cohen DL, Luther JM, Giordano T, Turcu A, Warth R, Rainey WE, Rege J. Somatic Mutations in MCOLN3 in Aldosterone-Producing Adenomas cause Primary Aldosteronism. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.10.20.619295. [PMID: 39484451 PMCID: PMC11526969 DOI: 10.1101/2024.10.20.619295] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/03/2024]
Abstract
Primary aldosteronism is characterized by renin-independent hyperaldosteronism that originates from aldosterone-producing lesions in the adrenal glands. Under physiological conditions, aldosterone synthase ( CYP11B2 ) expression is confined to the adrenal zona glomerulosa where it catalyzes the final reaction yielding aldosterone. The regulation of CYP11B2 transcription depends on the control of cellular membrane potential and cytosolic calcium activity. In primary aldosteronism, aldosterone-producing adenomas (APAs) are characterized by disrupted regulation of CYP11B2 expression resulting in autonomous biosynthesis of aldosterone. These lesions often harbor aldosterone-driver somatic mutations in genes encoding ion transporters/channels/pumps that increase cytosolic calcium activity causing increased CYP11B2 expression and aldosterone biosynthesis. We investigated APAs devoid of known somatic mutations and detected a missense mutation and a deletion-insertion variant in MCOLN3 which encodes for mucolipin-3 (TRPML3) - a highly conserved inwardly-rectifying, cation-permeable channel. These MCOLN3 mutations were identified in three APAs derived from male patients with primary aldosteronism: p. Y391D and p.N411_V412delinsI. Both mutations are located near the ion pore and selectivity filter of TRPML3. This is the first report of disease-causing MCOLN3 mutations in humans. Functional studies suggest MCOLN3 Y391D might directly or indirectly via membrane depolarization alter calcium influx of transfected adrenocortical cells, resulting in increased CYP11B2 transcription and aldosterone production. This study implicates mutated MCOLN3 as a driver of aldosterone excess in primary aldosteronism. Significance Statement Primary aldosteronism is a common but under-diagnosed endocrine disease that contributes to global hypertension burden and cardiovascular mortality and morbidity. Hyperaldosteronism in primary aldosteronism is mainly caused by adrenal lesions harboring somatic mutations that disrupt intracellular calcium levels and consequently aldosterone synthase expression and aldosterone production. Majority of these mutations have been identified in genes encoding ion transporters/channels/pumps. Herein, we report the first disease-causing somatic mutations in human MCOLN3 in aldosterone-producing adenomas (APAs) devoid of known mutations. In vitro investigations showed the MCOLN3 variant (p.Y391D) caused an influx of cytosolic calcium in adrenocortical cells and the subsequent increase in aldosterone synthase and aldosterone biosynthesis.
Collapse
|
11
|
Kim S, Chaudhary PK, Kim S. Molecular and Genetics Perspectives on Primary Adrenocortical Hyperfunction Disorders. Int J Mol Sci 2024; 25:11341. [PMID: 39518893 PMCID: PMC11545009 DOI: 10.3390/ijms252111341] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2024] [Revised: 10/15/2024] [Accepted: 10/20/2024] [Indexed: 11/16/2024] Open
Abstract
Adrenocortical disorders encompass a broad spectrum of conditions ranging from benign hyperplasia to malignant tumors, significantly disrupting hormone balance and causing a variety of clinical manifestations. By leveraging next-generation sequencing and in silico analyses, recent studies have uncovered the genetic and molecular pathways implicated in these transitions. In this review, we explored the molecular and genetic alterations in adrenocortical disorders, with a particular focus on the transitions from normal adrenal function to hyperfunction. The insights gained are intended to enhance diagnostic and therapeutic strategies, offering up-to-date knowledge for managing these complex conditions effectively.
Collapse
Affiliation(s)
| | | | - Soochong Kim
- College of Veterinary Medicine, Chungbuk National University, Cheongju 28644, Republic of Korea; (S.K.); (P.K.C.)
| |
Collapse
|
12
|
McEvoy JW, McCarthy CP, Bruno RM, Brouwers S, Canavan MD, Ceconi C, Christodorescu RM, Daskalopoulou SS, Ferro CJ, Gerdts E, Hanssen H, Harris J, Lauder L, McManus RJ, Molloy GJ, Rahimi K, Regitz-Zagrosek V, Rossi GP, Sandset EC, Scheenaerts B, Staessen JA, Uchmanowicz I, Volterrani M, Touyz RM. 2024 ESC Guidelines for the management of elevated blood pressure and hypertension. Eur Heart J 2024; 45:3912-4018. [PMID: 39210715 DOI: 10.1093/eurheartj/ehae178] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 09/04/2024] Open
|
13
|
Borges KS, Little DW, Magalhães TDA, Ribeiro C, Dumontet T, Lapensee C, Basham KJ, Seth A, Azova S, Guagliardo NA, Barrett PQ, Berber M, O'Connell AE, Turcu AF, Lerario AM, Mohan DR, Rainey W, Carlone DL, Hirschhorn JN, Salic A, Breault DT, Hammer GD. Non-canonical Wnt signaling triggered by WNT2B drives adrenal aldosterone production. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.08.23.609423. [PMID: 39229119 PMCID: PMC11370552 DOI: 10.1101/2024.08.23.609423] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 09/05/2024]
Abstract
The steroid hormone aldosterone, produced by the zona glomerulosa (zG) of the adrenal gland, is a master regulator of plasma electrolytes and blood pressure. While aldosterone control by the renin-angiotensin system is well understood, other key regulatory factors have remained elusive. Here, we replicated a prior association between a non-coding variant in WNT2B and an increased risk of primary aldosteronism, a prevalent and debilitating disease caused by excessive aldosterone production. We further show that in both mice and humans, WNT2B is expressed in the mesenchymal capsule surrounding the adrenal cortex, in close proximity to the zG. Global loss of Wnt2b in the mouse results in a dysmorphic and hypocellular zG, with impaired aldosterone production. Similarly, humans harboring WNT2B loss-of-function mutations develop a novel form of Familial Hyperreninemic Hypoaldosteronism, designated here as Type 4. Additionally, we demonstrate that WNT2B signals by activating the non-canonical Wnt/planar cell polarity pathway. Our findings identify WNT2B as a key regulator of zG function and aldosterone production with important clinical implications.
Collapse
Affiliation(s)
- Kleiton S Borges
- Division of Endocrinology, Boston Children's Hospital, Boston, MA, 02115, USA
- Department of Pediatrics, Harvard Medical School, Boston, MA, 02115, USA
| | - Donald W Little
- Doctoral Program in Cancer Biology, University of Michigan, Ann Arbor, MI, 48109, USA
- Department of Internal Medicine, Division of Metabolism, Endocrinology, and Diabetes, University of Michigan, Ann Arbor, MI, 48109, USA
| | | | - Claudio Ribeiro
- Division of Endocrinology, Boston Children's Hospital, Boston, MA, 02115, USA
| | - Typhanie Dumontet
- Department of Internal Medicine, Division of Metabolism, Endocrinology, and Diabetes, University of Michigan, Ann Arbor, MI, 48109, USA
| | - Chris Lapensee
- Department of Internal Medicine, Division of Metabolism, Endocrinology, and Diabetes, University of Michigan, Ann Arbor, MI, 48109, USA
| | - Kaitlin J Basham
- Department of Oncological Sciences, Huntsman Cancer Institute, University of Utah, Salt Lake City, UT, 84112, USA
| | - Aishwarya Seth
- Division of Endocrinology, Boston Children's Hospital, Boston, MA, 02115, USA
- Broad Institute of MIT and Harvard, Cambridge MA, 02142
| | - Svetlana Azova
- Division of Endocrinology, Boston Children's Hospital, Boston, MA, 02115, USA
- Department of Pediatrics, Harvard Medical School, Boston, MA, 02115, USA
| | - Nick A Guagliardo
- Department of Pharmacology, University of Virginia, Charlottesville, VA, 22908-0735, USA
| | - Paula Q Barrett
- Department of Pharmacology, University of Virginia, Charlottesville, VA, 22908-0735, USA
| | - Mesut Berber
- Division of Endocrinology, Boston Children's Hospital, Boston, MA, 02115, USA
- Department of Pediatrics, Harvard Medical School, Boston, MA, 02115, USA
| | - Amy E O'Connell
- Department of Pediatrics, Harvard Medical School, Boston, MA, 02115, USA
- Division of Newborn Medicine, Boston Children's Hospital, Boston, MA, 02115, USA
| | - Adina F Turcu
- Department of Internal Medicine, Division of Metabolism, Endocrinology, and Diabetes, University of Michigan, Ann Arbor, MI, 48109, USA
| | - Antonio Marcondes Lerario
- Department of Internal Medicine, Division of Metabolism, Endocrinology, and Diabetes, University of Michigan, Ann Arbor, MI, 48109, USA
| | - Dipika R Mohan
- Doctoral Program in Cancer Biology, University of Michigan, Ann Arbor, MI, 48109, USA
- Department of Internal Medicine, Division of Metabolism, Endocrinology, and Diabetes, University of Michigan, Ann Arbor, MI, 48109, USA
- Medical Scientist Training Program, University of Michigan, Ann Arbor, MI 48109, USA
| | - William Rainey
- Department of Internal Medicine, Division of Metabolism, Endocrinology, and Diabetes, University of Michigan, Ann Arbor, MI, 48109, USA
- Department of Molecular and Integrative Physiology, University of Michigan, Ann Arbor, MI, 48109, USA
| | - Diana L Carlone
- Division of Endocrinology, Boston Children's Hospital, Boston, MA, 02115, USA
- Department of Pediatrics, Harvard Medical School, Boston, MA, 02115, USA
- Harvard Stem Cell Institute, Cambridge, MA, 02138, USA
| | - Joel N Hirschhorn
- Division of Endocrinology, Boston Children's Hospital, Boston, MA, 02115, USA
- Department of Pediatrics, Harvard Medical School, Boston, MA, 02115, USA
- Broad Institute of MIT and Harvard, Cambridge MA, 02142
| | - Adrian Salic
- Department of Cell Biology, Harvard Medical School, Boston, MA, 02115, USA
| | - David T Breault
- Division of Endocrinology, Boston Children's Hospital, Boston, MA, 02115, USA
- Department of Pediatrics, Harvard Medical School, Boston, MA, 02115, USA
- Broad Institute of MIT and Harvard, Cambridge MA, 02142
- Harvard Stem Cell Institute, Cambridge, MA, 02138, USA
| | - Gary D Hammer
- Department of Internal Medicine, Division of Metabolism, Endocrinology, and Diabetes, University of Michigan, Ann Arbor, MI, 48109, USA
- Endocrine Oncology Program, Rogel Cancer Center, University of Michigan, Ann Arbor, MI, 48109, USA
| |
Collapse
|
14
|
Aminuddin A, Brown MJ, Azizan EA. Evaluating the role of aldosterone synthesis on adrenal cell fate. Front Endocrinol (Lausanne) 2024; 15:1423027. [PMID: 39170743 PMCID: PMC11335638 DOI: 10.3389/fendo.2024.1423027] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/25/2024] [Accepted: 07/23/2024] [Indexed: 08/23/2024] Open
Abstract
Hypertension affects one-third of the adult population worldwide, with primary aldosteronism (PA) accounting for at least 5-10% of these cases. The aldosterone synthase enzyme (CYP11B2) plays a pivotal role in PA manifestation, as increased expression of CYP11B2 leads to excess aldosterone synthesis. Physiological expression of CYP11B2 in humans is normally limited to cells of the adrenal zona glomerulosa under tight homeostatic regulation. In PA, however, there are CYP11B2-positive lesions in the adrenal cortex that autonomously secrete aldosterone, highlighting the dysregulation of adrenal cortex zonation and function as a key aspect of PA pathogenesis. Thus, this review aims to summarize the development of the adrenal glands, the key regulators of adrenal cortex homeostasis, and the dysregulation of this homeostasis. It also discusses the development of CYP11B2 inhibitors for therapeutic use in patients with hypertension, as well as the current knowledge of the effects of CYP11B2 inhibition on adrenal cortex homeostasis and cell fate. Understanding the control of adrenal cell fate may offer valuable insights into both the pathogenesis of PA and the development of alternative treatment approaches for PA.
Collapse
Affiliation(s)
- Amnani Aminuddin
- Department of Medicine, Faculty of Medicine, Universiti Kebangsaan Malaysia, Kuala Lumpur, Malaysia
| | - Morris J. Brown
- Endocrine Hypertension, Department of Clinical Pharmacology and Precision Medicine, William Harvey Research Institute, Queen Mary University of London, London, United Kingdom
- National Institute for Health Research (NIHR) Barts Biomedical Research Centre, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London, United Kingdom
| | - Elena Aisha Azizan
- Department of Medicine, Faculty of Medicine, Universiti Kebangsaan Malaysia, Kuala Lumpur, Malaysia
- Endocrine Hypertension, Department of Clinical Pharmacology and Precision Medicine, William Harvey Research Institute, Queen Mary University of London, London, United Kingdom
- Research Center, Hospital Tunku Ampuan Besar Tuanku Aishah Rohani, Universiti Kebangsaan Malaysia Specialist Children’s Hospital, Kuala Lumpur, Malaysia
| |
Collapse
|
15
|
Lin J, Li Y, Huang Z, Zhu Y, Li L, Yang H, Liang X, Qin Y, Zhou J, Xian J, Liu D, Lu D, Luo Z. Rare correlation of somatic PRKACA mutations with pregnancy-associated aldosterone- and cortisol-producing adenomas: a case report and literature review. BMC Endocr Disord 2024; 24:116. [PMID: 39010034 PMCID: PMC11251286 DOI: 10.1186/s12902-024-01645-x] [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: 01/29/2024] [Accepted: 07/03/2024] [Indexed: 07/17/2024] Open
Abstract
BACKGROUND Somatic mutations have been observed to induce aldosterone-producing adenomas (APAs). These may be accelerated during pregnancy. Somatic PRKACA mutations are common in cortisol-producing adenomas (CPAs). However, their role in APAs, particularly aldosterone- and cortisol-producing adenomas (A/CPAs), is not well understood. This study aims to investigate the association between PRKACA mutations and the accelerated development of A/CPAs during pregnancy. CASE PRESENTATION A patient with primary aldosteronism (PA) associated with severe Cushing's syndrome (CS) underwent surgical resection of an adrenal tumor one year after delivery. Pathologic examination revealed an adrenocortical adenoma characterized primarily by zona glomerulosa hyperplasia. Somatic mutation analysis revealed the presence of the somatic PRKACA mutation, which was validated as a deleterious mutation by various computational databases. Immunohistochemical results showed positive staining for cytochrome P450 family 11 subfamily B member 1 (CYP11B1), cytochrome P450 family 11 subfamily B member 2 (CYP11B2), and luteinizing hormone/chorionic gonadotropin receptor (LHCGR). Our study included a review of 20 previously documented cases of aldosterone- and cortisol-producing adenomas (A/CPAs), two of which were concurrently positive for both CYP11B1 and CYP11B2, consistent with our findings. CONCLUSION Somatic mutations in PRKACA may correlate with the upregulation of LHCGR, which synergistically drives the accelerated growth of co-secretion tumors during pregnancy, thereby exacerbating disease progression.
Collapse
Affiliation(s)
- Jianfan Lin
- Department of Endocrinology, The First Affiliated Hospital of Guangxi Medical University, No.6, Shuangyong Road, Nanning, 530021, P.R. China
| | - Yufei Li
- Department of Endocrinology, The First Affiliated Hospital of Guangxi Medical University, No.6, Shuangyong Road, Nanning, 530021, P.R. China
| | - Zhenxing Huang
- Department of Endocrinology, The First Affiliated Hospital of Guangxi Medical University, No.6, Shuangyong Road, Nanning, 530021, P.R. China
| | - Yingli Zhu
- Department of Endocrinology, The First Affiliated Hospital of Guangxi Medical University, No.6, Shuangyong Road, Nanning, 530021, P.R. China
| | - Li Li
- Department of Endocrinology, The First Affiliated Hospital of Guangxi Medical University, No.6, Shuangyong Road, Nanning, 530021, P.R. China
| | - Haiyan Yang
- Department of Endocrinology, The First Affiliated Hospital of Guangxi Medical University, No.6, Shuangyong Road, Nanning, 530021, P.R. China
| | - Xinghuan Liang
- Department of Endocrinology, The First Affiliated Hospital of Guangxi Medical University, No.6, Shuangyong Road, Nanning, 530021, P.R. China
| | - Yingfen Qin
- Department of Endocrinology, The First Affiliated Hospital of Guangxi Medical University, No.6, Shuangyong Road, Nanning, 530021, P.R. China
| | - Jia Zhou
- Department of Endocrinology, The First Affiliated Hospital of Guangxi Medical University, No.6, Shuangyong Road, Nanning, 530021, P.R. China
| | - Jing Xian
- Department of Endocrinology, The First Affiliated Hospital of Guangxi Medical University, No.6, Shuangyong Road, Nanning, 530021, P.R. China
| | - Deyun Liu
- Department of Urology, The First Affiliated Hospital of Guangxi Medical University, Nanning, 530021, P. R. China
| | - Decheng Lu
- Department of Endocrinology, The First Affiliated Hospital of Guangxi Medical University, No.6, Shuangyong Road, Nanning, 530021, P.R. China
| | - Zuojie Luo
- Department of Endocrinology, The First Affiliated Hospital of Guangxi Medical University, No.6, Shuangyong Road, Nanning, 530021, P.R. China.
| |
Collapse
|
16
|
Trogdon M, Abbott K, Arang N, Lande K, Kaur N, Tong M, Bakhoum M, Gutkind JS, Stites EC. Systems modeling of oncogenic G-protein and GPCR signaling reveals unexpected differences in downstream pathway activation. NPJ Syst Biol Appl 2024; 10:75. [PMID: 39013872 PMCID: PMC11252164 DOI: 10.1038/s41540-024-00400-1] [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: 07/12/2023] [Accepted: 06/27/2024] [Indexed: 07/18/2024] Open
Abstract
Mathematical models of biochemical reaction networks are an important and emerging tool for the study of cell signaling networks involved in disease processes. One promising potential application of such mathematical models is the study of how disease-causing mutations promote the signaling phenotype that contributes to the disease. It is commonly assumed that one must have a thorough characterization of the network readily available for mathematical modeling to be useful, but we hypothesized that mathematical modeling could be useful when there is incomplete knowledge and that it could be a tool for discovery that opens new areas for further exploration. In the present study, we first develop a mechanistic mathematical model of a G-protein coupled receptor signaling network that is mutated in almost all cases of uveal melanoma and use model-driven explorations to uncover and explore multiple new areas for investigating this disease. Modeling the two major, mutually-exclusive, oncogenic mutations (Gαq/11 and CysLT2R) revealed the potential for previously unknown qualitative differences between seemingly interchangeable disease-promoting mutations, and our experiments confirmed oncogenic CysLT2R was impaired at activating the FAK/YAP/TAZ pathway relative to Gαq/11. This led us to hypothesize that CYSLTR2 mutations in UM must co-occur with other mutations to activate FAK/YAP/TAZ signaling, and our bioinformatic analysis uncovers a role for co-occurring mutations involving the plexin/semaphorin pathway, which has been shown capable of activating this pathway. Overall, this work highlights the power of mechanism-based computational systems biology as a discovery tool that can leverage available information to open new research areas.
Collapse
Affiliation(s)
- Michael Trogdon
- Integrative Biology Laboratory, Salk Institute for Biological Studies, La Jolla, CA, 92037, USA
- Pfizer, La Jolla, CA, 92037, USA
| | - Kodye Abbott
- Department of Laboratory Medicine, Yale School of Medicine, New Haven, CT, 06520, USA
| | - Nadia Arang
- Moores Cancer Center, University of California, San Diego, La Jolla, CA, 92093, USA
- Biomedical Sciences Graduate Program, University of California, San Diego, La Jolla, CA, 92093, USA
| | - Kathryn Lande
- Razavi Newman Integrative Genomics and Bioinformatics Core, Salk Institute for Biological Studies, La Jolla, CA, 92037, USA
| | - Navneet Kaur
- Department of Laboratory Medicine, Yale School of Medicine, New Haven, CT, 06520, USA
| | - Melinda Tong
- Integrative Biology Laboratory, Salk Institute for Biological Studies, La Jolla, CA, 92037, USA
| | - Mathieu Bakhoum
- Department of Ophthalmology and Visual Science, Yale School of Medicine, New Haven, CT, 06520, USA
- Yale Cancer Center, Yale School of Medicine, New Haven, CT, 06520, USA
| | - J Silvio Gutkind
- Moores Cancer Center, University of California, San Diego, La Jolla, CA, 92093, USA
- Department of Pharmacology, University of California, San Diego, La Jolla, CA, 92093, USA
| | - Edward C Stites
- Department of Laboratory Medicine, Yale School of Medicine, New Haven, CT, 06520, USA.
- Yale Cancer Center, Yale School of Medicine, New Haven, CT, 06520, USA.
| |
Collapse
|
17
|
Culver SA, Suleman N, Kavuru V, Siragy HM. Renal Hypokalemia: An Endocrine Perspective. J Clin Endocrinol Metab 2024; 109:1694-1706. [PMID: 38546505 PMCID: PMC12102726 DOI: 10.1210/clinem/dgae201] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/20/2023] [Indexed: 06/18/2024]
Abstract
The majority of disorders that cause renal potassium wasting present with abnormalities in adrenal hormone secretion. While these findings frequently lead patients to seek endocrine evaluation, clinicians often struggle to accurately diagnose these conditions, delaying treatment and adversely impacting patient care. At the same time, growing insight into the genetic and molecular basis of these disorders continues to improve their diagnosis and management. In this review, we outline a practical integrated approach to the evaluation of renal hypokalemia syndromes that are seen in endocrine practice while highlighting recent advances in understanding of the genetics and pathophysiology behind them.
Collapse
Affiliation(s)
- Silas A Culver
- Division of Endocrinology, Department of Medicine, University of Virginia Health System, Charlottesville, VA 22908, USA
| | - Nawar Suleman
- Division of Endocrinology, Department of Medicine, University of Virginia Health System, Charlottesville, VA 22908, USA
| | - Varun Kavuru
- Department of Medicine, University of Virginia Health System, Charlottesville, VA 22908, USA
| | - Helmy M Siragy
- Division of Endocrinology, Department of Medicine, University of Virginia Health System, Charlottesville, VA 22908, USA
| |
Collapse
|
18
|
Tetti M, Brüdgam D, Jacopo Burrello, Udager AM, Riester A, Knösel T, Beuschlein F, Rainey WE, Reincke M, Williams TA. Unilateral Primary Aldosteronism: Long-Term Disease Recurrence After Adrenalectomy. Hypertension 2024; 81:936-945. [PMID: 38318706 PMCID: PMC10954406 DOI: 10.1161/hypertensionaha.123.22281] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2023] [Accepted: 01/17/2024] [Indexed: 02/07/2024]
Abstract
BACKGROUND Primary aldosteronism (PA) is frequently caused by a unilateral aldosterone-producing adenoma with a PA-driver mutation. Unilateral adrenalectomy has a high probability of short-term biochemical remission, but long-term postsurgical outcomes are relatively undefined. Our objective was to investigate the incidence of long-term recurrence of PA in individuals with postsurgical short-term biochemical remission. METHODS Adrenalectomized patients for unilateral PA were included from a single referral center. Histopathology and outcomes were assessed according to international histopathology of unilateral primary aldosteronism and PASO (Primary Aldosteronism Surgical Outcome) consensuses. Genotyping was performed using CYP11B2 (aldosterone synthase)-guided sequencing. RESULTS Classical adrenal histopathology, exemplified by a solitary aldosterone-producing adenoma, was observed in 78% of 90 adrenals, compared with 22% with nonclassical histopathology. The classical group displayed higher aldosterone-to-renin ratios (P=0.013) and lower contralateral ratios (P=0.008). Outcome assessments at both short (12 months [7; 12]) and long (89 months [48; 124]) terms were available for 57 patients. At short-term assessment, 53 (93%) displayed complete biochemical success (43 classical and 10 nonclassical), but long-term assessment demonstrated biochemical PA recurrence in 12 (23%) with an overrepresentation of the nonclassical histopathology (6 [60%] of 10 nonclassical histopathology versus 6 [14%] of 43 classical histopathology; P=0.005). PA-driver mutations were identified in 97% of 64 aldosterone-producing adenomas; there was no association of the aldosterone-producing adenoma genotype with PA recurrence. CONCLUSIONS A substantial proportion of individuals display postsurgical biochemical recurrence of PA, which is related to the histopathology of the resected adrenal gland. These findings emphasize the role of histopathology and the requirement for continued outcome assessment in the management of surgically treated patients for PA.
Collapse
Affiliation(s)
- Martina Tetti
- Medizinische Klinik und Poliklinik IV,
LMU Klinikum, LMU München, Munich, Germany
| | - Denise Brüdgam
- Medizinische Klinik und Poliklinik IV,
LMU Klinikum, LMU München, Munich, Germany
| | - Jacopo Burrello
- Division of Internal Medicine and Hypertension, Department of Medical Sciences, University of Turin, Turin, Italy
| | - Aaron M Udager
- Department of Pathology, University of Michigan, Ann Arbor, MI, 48109, USA
- Rogel Cancer Center, University of Michigan, Ann Arbor, MI, 48109, USA
| | - Anna Riester
- Medizinische Klinik und Poliklinik IV,
LMU Klinikum, LMU München, Munich, Germany
| | - Thomas Knösel
- Institute of Pathology, Ludwig-Maximilians-Universität München, Munich, Germany
| | - Felix Beuschlein
- Medizinische Klinik und Poliklinik IV,
LMU Klinikum, LMU München, Munich, Germany
- Klinik für Endokrinologie, Diabetologie und Klinische Ernährung, Universitätsspital Zürich (USZ) and Universität Zürich (UZH), Zürich, Switzerland
- The LOOP Zurich - Medical Research Center, Zurich, Switzerland
| | - William E Rainey
- Department of Molecular and Integrative Physiology, University of Michigan, Ann Arbor, MI, 48109, USA
- Division of Metabolism, Endocrine, and Diabetes, Department of Internal Medicine, University of Michigan, Ann Arbor, MI, 48109, USA
| | - Martin Reincke
- Medizinische Klinik und Poliklinik IV,
LMU Klinikum, LMU München, Munich, Germany
| | - Tracy Ann Williams
- Medizinische Klinik und Poliklinik IV,
LMU Klinikum, LMU München, Munich, Germany
| |
Collapse
|
19
|
Nanba K, Blinder AR, Udager AM, Hirokawa Y, Miura T, Okuno H, Moriyoshi K, Yamazaki Y, Sasano H, Yasoda A, Satoh-Asahara N, Rainey WE, Tagami T. Double somatic mutations in CTNNB1 and GNA11 in an aldosterone-producing adenoma. Front Endocrinol (Lausanne) 2024; 15:1286297. [PMID: 38505749 PMCID: PMC10948454 DOI: 10.3389/fendo.2024.1286297] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/31/2023] [Accepted: 02/21/2024] [Indexed: 03/21/2024] Open
Abstract
Double somatic mutations in CTNNB1 and GNA11/Q have recently been identified in a small subset of aldosterone-producing adenomas (APAs). As a possible pathogenesis of APA due to these mutations, an association with pregnancy, menopause, or puberty has been proposed. However, because of its rarity, characteristics of APA with these mutations have not been well characterized. A 46-year-old Japanese woman presented with hypertension and hypokalemia. She had two pregnancies in the past but had no history of pregnancy-induced hypertension. She had regular menstrual cycle at presentation and was diagnosed as having primary aldosteronism after endocrinologic examinations. Computed tomography revealed a 2 cm right adrenal mass. Adrenal venous sampling demonstrated excess aldosterone production from the right adrenal gland. She underwent right laparoscopic adrenalectomy. The resected right adrenal tumor was histologically diagnosed as adrenocortical adenoma and subsequent immunohistochemistry (IHC) revealed diffuse immunoreactivity of aldosterone synthase (CYP11B2) and visinin like 1, a marker of the zona glomerulosa (ZG), whereas 11β-hydroxylase, a steroidogenic enzyme for cortisol biosynthesis, was mostly negative. CYP11B2 IHC-guided targeted next-generation sequencing identified somatic CTNNB1 (p.D32Y) and GNA11 (p.Q209H) mutations. Immunofluorescence staining of the tumor also revealed the presence of activated β-catenin, consistent with features of the normal ZG. The expression patterns of steroidogenic enzymes and related proteins indicated ZG features of the tumor cells. PA was clinically and biochemically cured after surgery. In conclusion, our study indicated that CTNNB1 and GNA11-mutated APA has characteristics of the ZG. The disease could occur in adults with no clear association with pregnancy or menopause.
Collapse
Affiliation(s)
- Kazutaka Nanba
- Department of Endocrinology and Metabolism, National Hospital Organization Kyoto Medical Center, Kyoto, Japan
- Department of Endocrinology, Metabolism, and Hypertension Research, Clinical Research Institute, National Hospital Organization Kyoto Medical Center, Kyoto, Japan
- Department of Molecular and Integrative Physiology, University of Michigan, Ann Arbor, MI, United States
| | - Amy R. Blinder
- Department of Molecular and Integrative Physiology, University of Michigan, Ann Arbor, MI, United States
| | - Aaron M. Udager
- Department of Pathology, University of Michigan, Ann Arbor, MI, United States
- Michigan Center for Translational Pathology, University of Michigan, Ann Arbor, MI, United States
- Rogel Cancer Center, University of Michigan, Ann Arbor, MI, United States
| | - Yuusuke Hirokawa
- Department of Radiology, National Hospital Organization Kyoto Medical Center, Kyoto, Japan
| | - Takayoshi Miura
- Department of Urology, National Hospital Organization Kyoto Medical Center, Kyoto, Japan
| | - Hiroshi Okuno
- Department of Urology, National Hospital Organization Kyoto Medical Center, Kyoto, Japan
| | - Koki Moriyoshi
- Department of Diagnostic Pathology, National Hospital Organization Kyoto Medical Center, Kyoto, Japan
| | - Yuto Yamazaki
- Department of Pathology, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Hironobu Sasano
- Department of Pathology, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Akihiro Yasoda
- Clinical Research Institute, National Hospital Organization Kyoto Medical Center, Kyoto, Japan
| | - Noriko Satoh-Asahara
- Department of Endocrinology, Metabolism, and Hypertension Research, Clinical Research Institute, National Hospital Organization Kyoto Medical Center, Kyoto, Japan
| | - William E. Rainey
- Department of Molecular and Integrative Physiology, University of Michigan, Ann Arbor, MI, United States
- Division of Metabolism, Endocrinology, and Diabetes, University of Michigan, Ann Arbor, MI, United States
| | - Tetsuya Tagami
- Department of Endocrinology and Metabolism, National Hospital Organization Kyoto Medical Center, Kyoto, Japan
- Department of Endocrinology, Metabolism, and Hypertension Research, Clinical Research Institute, National Hospital Organization Kyoto Medical Center, Kyoto, Japan
| |
Collapse
|
20
|
Murakami M, Hara K, Ikeda K, Horino M, Okazaki R, Niitsu Y, Takeuchi A, Aoki J, Shiba K, Tsujimoto K, Komiya C, Nakamura Y, Kurata M, Akashi T, Fujii Y, Yamada T. Single-Nucleus Analysis Reveals Tumor Heterogeneity of Aldosterone-Producing Adenoma. Hypertension 2024; 81:361-371. [PMID: 38095094 DOI: 10.1161/hypertensionaha.123.21446] [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: 04/28/2023] [Accepted: 12/03/2023] [Indexed: 01/19/2024]
Abstract
BACKGROUND Recent advances in omics techniques have allowed detailed genetic characterization of aldosterone-producing adenoma (APA). The pathogenesis of APA is characterized by tumorigenesis-associated aldosterone synthesis. The pathophysiological intricacies of APAs have not yet been elucidated at the level of individual cells. Therefore, a single-cell level analysis is speculated to be valuable in studying the differentiation process of APA. METHODS We conducted single-nucleus RNA sequencing of APAs with KCNJ5 mutation and nonfunctional adenomas obtained from 3 and 2 patients, respectively. RESULTS The single-nucleus RNA sequencing revealed the intratumoral heterogeneity of APA and identified cell populations consisting of a shared cluster of nonfunctional adenoma and APA. In addition, we extracted 2 cell fates in APA and obtained a cell population specialized in aldosterone synthesis. Genes related to ribosomes and neurodegenerative diseases were upregulated in 1 of these fates, whereas those related to the regulation of glycolysis were upregulated in the other fate. Furthermore, the total RNA reads in the nucleus were higher in hormonally activated clusters, indicating a marked activation of transcription per cell. CONCLUSIONS The single-nucleus RNA sequencing revealed intratumoral heterogeneity of APA with KCNJ5 mutation. The observation of 2 cell fates in KCNJ5-mutated APAs provides the postulation that a heterogeneous process of cellular differentiation was implicated in the pathophysiological mechanisms underlying APA tumors.
Collapse
Affiliation(s)
- Masanori Murakami
- Department of Molecular Endocrinology and Metabolism, Graduate School of Medical and Dental Sciences (M.M., K.H., K.I., M.H., R.O., Y.N., A.T., J.A., K.S., K.T., C.K., T.Y.), Tokyo Medical and Dental University, Japan
| | - Kazunari Hara
- Department of Molecular Endocrinology and Metabolism, Graduate School of Medical and Dental Sciences (M.M., K.H., K.I., M.H., R.O., Y.N., A.T., J.A., K.S., K.T., C.K., T.Y.), Tokyo Medical and Dental University, Japan
| | - Kenji Ikeda
- Department of Molecular Endocrinology and Metabolism, Graduate School of Medical and Dental Sciences (M.M., K.H., K.I., M.H., R.O., Y.N., A.T., J.A., K.S., K.T., C.K., T.Y.), Tokyo Medical and Dental University, Japan
| | - Masato Horino
- Department of Molecular Endocrinology and Metabolism, Graduate School of Medical and Dental Sciences (M.M., K.H., K.I., M.H., R.O., Y.N., A.T., J.A., K.S., K.T., C.K., T.Y.), Tokyo Medical and Dental University, Japan
| | - Rei Okazaki
- Department of Molecular Endocrinology and Metabolism, Graduate School of Medical and Dental Sciences (M.M., K.H., K.I., M.H., R.O., Y.N., A.T., J.A., K.S., K.T., C.K., T.Y.), Tokyo Medical and Dental University, Japan
| | - Yoshihiro Niitsu
- Department of Molecular Endocrinology and Metabolism, Graduate School of Medical and Dental Sciences (M.M., K.H., K.I., M.H., R.O., Y.N., A.T., J.A., K.S., K.T., C.K., T.Y.), Tokyo Medical and Dental University, Japan
| | - Akira Takeuchi
- Department of Molecular Endocrinology and Metabolism, Graduate School of Medical and Dental Sciences (M.M., K.H., K.I., M.H., R.O., Y.N., A.T., J.A., K.S., K.T., C.K., T.Y.), Tokyo Medical and Dental University, Japan
| | - Jun Aoki
- Department of Molecular Endocrinology and Metabolism, Graduate School of Medical and Dental Sciences (M.M., K.H., K.I., M.H., R.O., Y.N., A.T., J.A., K.S., K.T., C.K., T.Y.), Tokyo Medical and Dental University, Japan
| | - Kumiko Shiba
- Department of Molecular Endocrinology and Metabolism, Graduate School of Medical and Dental Sciences (M.M., K.H., K.I., M.H., R.O., Y.N., A.T., J.A., K.S., K.T., C.K., T.Y.), Tokyo Medical and Dental University, Japan
- Center for Personalized Medicine for Healthy Aging (K.S.), Tokyo Medical and Dental University, Japan
| | - Kazutaka Tsujimoto
- Department of Molecular Endocrinology and Metabolism, Graduate School of Medical and Dental Sciences (M.M., K.H., K.I., M.H., R.O., Y.N., A.T., J.A., K.S., K.T., C.K., T.Y.), Tokyo Medical and Dental University, Japan
| | - Chikara Komiya
- Department of Molecular Endocrinology and Metabolism, Graduate School of Medical and Dental Sciences (M.M., K.H., K.I., M.H., R.O., Y.N., A.T., J.A., K.S., K.T., C.K., T.Y.), Tokyo Medical and Dental University, Japan
| | - Yuki Nakamura
- Department of Urology, Graduate School of Medical and Dental Sciences (Y.N., Y.F.), Tokyo Medical and Dental University, Japan
| | - Morito Kurata
- Department of Comprehensive Pathology, Graduate School of Medical and Dental Sciences (M.K.), Tokyo Medical and Dental University, Japan
| | - Takumi Akashi
- Department of Diagnostic Pathology, Graduate School of Medical and Dental Sciences (T.A.), Tokyo Medical and Dental University, Japan
- Division of Surgical Pathology, Tokyo Medical and Dental University Hospital, Japan (T.A.)
| | - Yasuhisa Fujii
- Department of Urology, Graduate School of Medical and Dental Sciences (Y.N., Y.F.), Tokyo Medical and Dental University, Japan
| | - Tetsuya Yamada
- Department of Molecular Endocrinology and Metabolism, Graduate School of Medical and Dental Sciences (M.M., K.H., K.I., M.H., R.O., Y.N., A.T., J.A., K.S., K.T., C.K., T.Y.), Tokyo Medical and Dental University, Japan
| |
Collapse
|
21
|
Li Y, Lin J, Fu S, Li L, Huang Z, Yang H, Liang X, Qin Y, Zhou J, Liu D, Luo Z. The mystery of transient pregnancy-induced cushing's syndrome: a case report and literature review highlighting GNAS somatic mutations and LHCGR overexpression. Endocrine 2024; 83:473-482. [PMID: 37828397 DOI: 10.1007/s12020-023-03549-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/10/2023] [Accepted: 09/23/2023] [Indexed: 10/14/2023]
Abstract
PURPOSE Transient pregnancy-induced Cushing's syndrome is a rare condition characterized by the manifestation of symptoms solely during pregnancy, which typically resolve spontaneously following delivery or miscarriage. While it has been established that GNAS is associated with adrenal tumors, its specific role in the pathogenesis of pregnancy-induced Cushing's syndrome remains uncertain.This work aims to examine the association between GNAS mutation and pregnancy-induced Cushing's syndrome. METHODS DNA was extracted from patients' peripheral blood and tumor tissues for whole-exome sequencing (WES) and Sanger sequencing. We used AlphaFold to predict the protein structure of wild-type and mutant GNAS and to make functional predictions, and immunohistochemistry was used to detect disease-associated protein expression. A review and summary of reported cases of transient pregnancy-induced Cushing's syndrome induced by pregnancy was conducted. RESULTS Using WES, we identified a somatic mutation in GNAS (NM_000516, c.C601T, p.R201C) that was predicted to have a deleterious effect using computational methods, such as AlphaFold. Human chorionic gonadotropin (hCG) stimulation tests had weakly positive results, and immunohistochemical staining of adrenal adenoma tissue also revealed positivity for luteinizing hormone/chorionic gonadotropin receptor (LHCGR) and cytochrome P450 family 11 subfamily B member 1 (CYP11B1). We reviewed 15 published cases of transient Cushing's syndrome induced by pregnancy. Among these cases, immunohistochemical staining of the adrenal gland showed positive LHCGR expression in 3 case reports, similar to our findings. CONCLUSION Transient pregnancy-induced Cushing's syndrome may be associated with somatic GNAS mutations and altered adrenal pathology due to abnormal activation of LHCGR.
Collapse
Affiliation(s)
- Yufei Li
- Department of Endocrinology, The First Affiliated Hospital of Guangxi Medical University, Nanning, 530021, P. R. China
| | - Jianfan Lin
- Department of Endocrinology, The First Affiliated Hospital of Guangxi Medical University, Nanning, 530021, P. R. China
| | - Shien Fu
- Department of Endocrinology, The First Affiliated Hospital of Guangxi Medical University, Nanning, 530021, P. R. China
| | - Li Li
- Department of Endocrinology, The First Affiliated Hospital of Guangxi Medical University, Nanning, 530021, P. R. China
| | - Zhenxing Huang
- Department of Endocrinology, The First Affiliated Hospital of Guangxi Medical University, Nanning, 530021, P. R. China
| | - Haiyan Yang
- Department of Endocrinology, The First Affiliated Hospital of Guangxi Medical University, Nanning, 530021, P. R. China
| | - Xinghuan Liang
- Department of Endocrinology, The First Affiliated Hospital of Guangxi Medical University, Nanning, 530021, P. R. China
| | - Yingfen Qin
- Department of Endocrinology, The First Affiliated Hospital of Guangxi Medical University, Nanning, 530021, P. R. China
| | - Jia Zhou
- Department of Endocrinology, The First Affiliated Hospital of Guangxi Medical University, Nanning, 530021, P. R. China
| | - Deyun Liu
- Department of Urology, The First Affiliated Hospital of Guangxi Medical University, Nanning, 530021, P. R. China
| | - Zuojie Luo
- Department of Endocrinology, The First Affiliated Hospital of Guangxi Medical University, Nanning, 530021, P. R. China.
| |
Collapse
|
22
|
Stölting G, Scholl UI. Adrenal Anion Channels: New Roles in Zona Glomerulosa Physiology and in the Pathophysiology of Primary Aldosteronism. Handb Exp Pharmacol 2024; 283:59-79. [PMID: 37495852 DOI: 10.1007/164_2023_680] [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] [Indexed: 07/28/2023]
Abstract
The mineralocorticoid aldosterone is produced in the zona glomerulosa of the adrenal cortex. Its synthesis is regulated by the serum concentrations of the peptide hormone angiotensin II and potassium. The primary role of aldosterone is to control blood volume and electrolytes. The autonomous production of aldosterone (primary aldosteronism, PA) is considered the most frequent cause of secondary hypertension. Aldosterone-producing adenomas and (micro-)nodules are frequent causes of PA and often carry somatic mutations in ion channels and transporters. Rare familial forms of PA are due to germline mutations. Both somatic and germline mutations in the chloride channel gene CLCN2, encoding ClC-2, have been identified in PA. Clinical findings and results from cell culture and animal models have advanced our knowledge about the role of anions in PA. The zona glomerulosa of the adrenal gland has now been firmly established as a tissue in which anions play a significant role for signaling. In this overview, we aim to summarize the current knowledge and highlight novel concepts as well as open questions.
Collapse
Affiliation(s)
- Gabriel Stölting
- Center of Functional Genomics, Berlin Institute of Health at Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - Ute I Scholl
- Center of Functional Genomics, Berlin Institute of Health at Charité - Universitätsmedizin Berlin, Berlin, Germany.
| |
Collapse
|
23
|
Azizan EAB, Drake WM, Brown MJ. Primary aldosteronism: molecular medicine meets public health. Nat Rev Nephrol 2023; 19:788-806. [PMID: 37612380 PMCID: PMC7615304 DOI: 10.1038/s41581-023-00753-6] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/21/2023] [Indexed: 08/25/2023]
Abstract
Primary aldosteronism is the most common single cause of hypertension and is potentially curable when only one adrenal gland is the culprit. The importance of primary aldosteronism to public health derives from its high prevalence but huge under-diagnosis (estimated to be <1% of all affected individuals), despite the consequences of poor blood pressure control by conventional therapy and enhanced cardiovascular risk. This state of affairs is attributable to the fact that the tools used for diagnosis or treatment are still those that originated in the 1970-1990s. Conversely, molecular discoveries have transformed our understanding of adrenal physiology and pathology. Many molecules and processes associated with constant adrenocortical renewal and interzonal metamorphosis also feature in aldosterone-producing adenomas and aldosterone-producing micronodules. The adrenal gland has one of the most significant rates of non-silent somatic mutations, with frequent selection of those driving autonomous aldosterone production, and distinct clinical presentations and outcomes for most genotypes. The disappearance of aldosterone synthesis and cells from most of the adult human zona glomerulosa is the likely driver of the mutational success that causes aldosterone-producing adenomas, but insights into the pathways that lead to constitutive aldosterone production and cell survival may open up opportunities for novel therapies.
Collapse
Affiliation(s)
- Elena A B Azizan
- Department of Medicine, Faculty of Medicine, The National University of Malaysia (UKM), Kuala Lumpur, Malaysia
- Endocrine Hypertension, Department of Clinical Pharmacology and Precision Medicine, William Harvey Research Institute, Queen Mary University of London, London, United Kingdom
| | - William M Drake
- St Bartholomew's Hospital, Barts Health NHS Trust, London, United Kingdom
- NIHR Barts Biomedical Research Centre, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London, United Kingdom
| | - Morris J Brown
- Endocrine Hypertension, Department of Clinical Pharmacology and Precision Medicine, William Harvey Research Institute, Queen Mary University of London, London, United Kingdom.
- NIHR Barts Biomedical Research Centre, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London, United Kingdom.
| |
Collapse
|
24
|
Rege J, Bandulik S, Nanba K, Kosmann C, Blinder AR, Plain A, Vats P, Kumar-Sinha C, Lerario AM, Else T, Yamazaki Y, Satoh F, Sasano H, Giordano TJ, Williams TA, Reincke M, Turcu AF, Udager AM, Warth R, Rainey WE. Somatic SLC30A1 mutations altering zinc transporter ZnT1 cause aldosterone-producing adenomas and primary aldosteronism. Nat Genet 2023; 55:1623-1631. [PMID: 37709865 PMCID: PMC12051258 DOI: 10.1038/s41588-023-01498-5] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2022] [Accepted: 08/08/2023] [Indexed: 09/16/2023]
Abstract
Primary aldosteronism (PA) is the most common form of endocrine hypertension and is characterized by inappropriately elevated aldosterone production via a renin-independent mechanism. Driver somatic mutations for aldosterone excess have been found in approximately 90% of aldosterone-producing adenomas (APAs). Other causes of lateralized adrenal PA include aldosterone-producing nodules (APNs). Using next-generation sequencing, we identified recurrent in-frame deletions in SLC30A1 in four APAs and one APN (p.L51_A57del, n = 3; p.L49_L55del, n = 2). SLC30A1 encodes the ubiquitous zinc efflux transporter ZnT1 (zinc transporter 1). The identified SLC30A1 variants are situated close to the zinc-binding site (His43 and Asp47) in transmembrane domain II and probably cause abnormal ion transport. Cases of PA with SLC30A1 mutations showed male dominance and demonstrated increased aldosterone and 18-oxocortisol concentrations. Functional studies of the SLC30A151_57del variant in a doxycycline-inducible adrenal cell system revealed pathological Na+ influx. An aberrant Na+ current led to depolarization of the resting membrane potential and, thus, to the opening of voltage-gated calcium (Ca2+) channels. This resulted in an increase in cytosolic Ca2+ activity, which stimulated CYP11B2 mRNA expression and aldosterone production. Collectively, these data implicate zinc transporter alterations as a dominant driver of aldosterone excess in PA.
Collapse
Affiliation(s)
- Juilee Rege
- Department of Molecular & Integrative Physiology, University of Michigan, Ann Arbor, MI, USA
| | - Sascha Bandulik
- Medical Cell Biology, University of Regensburg, Regensburg, Germany
| | - Kazutaka Nanba
- Department of Molecular & Integrative Physiology, University of Michigan, Ann Arbor, MI, USA
- Department of Endocrinology and Metabolism, National Hospital Organization Kyoto Medical Center, Kyoto, Japan
| | - Carla Kosmann
- Medical Cell Biology, University of Regensburg, Regensburg, Germany
| | - Amy R Blinder
- Department of Molecular & Integrative Physiology, University of Michigan, Ann Arbor, MI, USA
| | - Allein Plain
- Medical Cell Biology, University of Regensburg, Regensburg, Germany
| | - Pankaj Vats
- Rogel Cancer Center, University of Michigan, Ann Arbor, MI, USA
- Michigan Center for Translational Pathology, University of Michigan, Ann Arbor, MI, USA
| | - Chandan Kumar-Sinha
- Rogel Cancer Center, University of Michigan, Ann Arbor, MI, USA
- Michigan Center for Translational Pathology, University of Michigan, Ann Arbor, MI, USA
| | - Antonio M Lerario
- Division of Metabolism, Endocrine, and Diabetes, Department of Internal Medicine, University of Michigan, Ann Arbor, MI, USA
| | - Tobias Else
- Division of Metabolism, Endocrine, and Diabetes, Department of Internal Medicine, University of Michigan, Ann Arbor, MI, USA
| | - Yuto Yamazaki
- Department of Pathology, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Fumitoshi Satoh
- Division of Clinical Hypertension, Endocrinology and Metabolism, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Hironobu Sasano
- Department of Pathology, Tohoku University Graduate School of Medicine, Sendai, Japan
| | | | - Tracy Ann Williams
- Medizinische Klinik und Poliklinik IV, Klinikum der Universität München, Ludwig Maximilian University of Munich, Munich, Germany
- Division of Internal Medicine and Hypertension, Department of Medical Sciences, University of Turin, Turin, Italy
| | - Martin Reincke
- Medizinische Klinik und Poliklinik IV, Klinikum der Universität München, Ludwig Maximilian University of Munich, Munich, Germany
| | - Adina F Turcu
- Division of Metabolism, Endocrine, and Diabetes, Department of Internal Medicine, University of Michigan, Ann Arbor, MI, USA
| | - Aaron M Udager
- Rogel Cancer Center, University of Michigan, Ann Arbor, MI, USA
- Michigan Center for Translational Pathology, University of Michigan, Ann Arbor, MI, USA
- Department of Pathology, University of Michigan, Ann Arbor, MI, USA
| | - Richard Warth
- Medical Cell Biology, University of Regensburg, Regensburg, Germany
| | - William E Rainey
- Department of Molecular & Integrative Physiology, University of Michigan, Ann Arbor, MI, USA.
- Division of Metabolism, Endocrine, and Diabetes, Department of Internal Medicine, University of Michigan, Ann Arbor, MI, USA.
| |
Collapse
|
25
|
MacKenzie SM, Birch LA, Lamprou S, Rezvanisanijouybari P, Fayad M, Zennaro MC, Davies E. MicroRNAs in aldosterone production and action. VITAMINS AND HORMONES 2023; 124:137-163. [PMID: 38408798 DOI: 10.1016/bs.vh.2023.07.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/28/2024]
Abstract
Aldosterone is a cardiovascular hormone with a key role in blood pressure regulation, among other processes, mediated through its targeting of the mineralocorticoid receptor in the renal tubule and selected other tissues. Its secretion from the adrenal gland is a highly controlled process subject to regulatory influence from the renin-angiotensin system and the hypothalamic-pituitary-adrenal axis. MicroRNAs are small endogenous non-coding RNA molecules capable of regulating gene expression post-transcriptionally through stimulation of mRNA degradation or suppression of translation. Several studies have now identified that microRNA levels are changed in cases of aldosterone dysregulation and that microRNAs are capable of regulating the expression of various genes involved in aldosterone production and action. In this article we summarise the major studies concerning this topic. We also discuss the potential role for circulating microRNAs as diagnostic biomarkers for primary aldosteronism, a highly treatable form of secondary hypertension, which would be highly desirable given the current underdiagnosis of this condition.
Collapse
Affiliation(s)
- Scott M MacKenzie
- School of Cardiovascular and Metabolic Health, BHF Glasgow Cardiovascular Research Centre, University of Glasgow, Glasgow, United Kingdom.
| | - Lara A Birch
- School of Cardiovascular and Metabolic Health, BHF Glasgow Cardiovascular Research Centre, University of Glasgow, Glasgow, United Kingdom
| | - Stelios Lamprou
- School of Cardiovascular and Metabolic Health, BHF Glasgow Cardiovascular Research Centre, University of Glasgow, Glasgow, United Kingdom
| | - Parisa Rezvanisanijouybari
- School of Cardiovascular and Metabolic Health, BHF Glasgow Cardiovascular Research Centre, University of Glasgow, Glasgow, United Kingdom
| | - May Fayad
- School of Cardiovascular and Metabolic Health, BHF Glasgow Cardiovascular Research Centre, University of Glasgow, Glasgow, United Kingdom; Université Paris Cité, PARCC, INSERM, Paris, France
| | - Maria-Christina Zennaro
- Université Paris Cité, PARCC, INSERM, Paris, France; Assistance Publique-Hôpitaux de Paris, Hôpital Européen Georges Pompidou, Service de Génétique, Paris, France
| | - Eleanor Davies
- School of Cardiovascular and Metabolic Health, BHF Glasgow Cardiovascular Research Centre, University of Glasgow, Glasgow, United Kingdom
| |
Collapse
|
26
|
Antoniotti V, Amore M, Caputo M, Fania C, Mancioppi V, Casoli G, Tini S, Antonioli A, Aimaretti G, Rabbone I, Bellone S, Prodam F. Glucose Alterations, Insulin Resistance, Arterial Hypertension, and Renin are Strictly Associated in Pediatric Obesity. J Endocr Soc 2023; 7:bvad088. [PMID: 37424701 PMCID: PMC10326241 DOI: 10.1210/jendso/bvad088] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/02/2023] [Indexed: 07/11/2023] Open
Abstract
Context Insulin resistance, glucose alterations, arterial hypertension (HTN), and the renin-angiotensin-aldosterone system (RAAS) are related in adult obesity. This crosstalk is still unexplored in childhood. Objective Characterize the relationships of fasting and postload glucose and insulin levels with new American Academy of Pediatrics classification of HTN and RAAS in pediatric obesity. Methods This was a retrospective observational study; 799 pediatric outpatients (11.4 ± 3.1 years) at a tertiary center who were overweight or obese and not yet on diet were included. The main outcome measures were mean and correlations among parameters of a complete clinical and metabolic screening (body mass index, blood pressure, and glucose and insulin levels during an oral glucose tolerance test, and renin and aldosterone levels and their ratio). Results 774 subjects had all the parameters, of whom 87.6% had HTN (5% elevated blood pressure, 29.2% stage I HTN, and 53.4% stage II HTN). Eighty subjects had 1 or more glucose alterations, and more frequently presented HTN. Blood pressure levels were higher in subjects with glucose alterations than in those with normal glucose levels. Fasting and stimulated glucose and insulin levels were directly related to the HTN stages, and insulin sensitivity was lower in HTN than in normal blood pressure. Aldosterone, renin, and aldosterone-renin ratio (ARR) were similar in sexes, whereas aldosterone was higher in prepubertal individuals. Subjects with impaired glucose tolerance (IGT) had higher renin and lower ARR. Renin was positively correlated with postload glucose, and ARR was negatively correlated with the Homeostatic Model Assessment for Insulin Resistance index. Conclusion A close relationship exists among insulin resistance, glucose alterations, HTN, and renin in childhood obesity. Specific categories of risk could provide indicators for strict clinical surveillance.
Collapse
Affiliation(s)
- Valentina Antoniotti
- SCDU of Pediatrics, Department of Health Sciences, University of Piemonte Orientale, 28100 Novara, Italy
| | - Martina Amore
- SCDU of Pediatrics, Department of Health Sciences, University of Piemonte Orientale, 28100 Novara, Italy
| | - Marina Caputo
- Endocrinology, Department of Translational Medicine, University of Piemonte Orientale, 28100 Novara, Italy
- Department of Health Sciences, University of Piemonte Orientale, 28100 Novara, Italy
| | - Chiara Fania
- SCDU Clinical Chemistry Laboratory, Maggiore della Carità University Hospital, 28100 Novara, Italy
| | - Valentina Mancioppi
- SCDU of Pediatrics, Department of Health Sciences, University of Piemonte Orientale, 28100 Novara, Italy
| | - Gloria Casoli
- SCDU Clinical Chemistry Laboratory, Maggiore della Carità University Hospital, 28100 Novara, Italy
| | - Sabrina Tini
- Department of Health Sciences, University of Piemonte Orientale, 28100 Novara, Italy
| | - Alessandro Antonioli
- Department of Health Sciences, University of Piemonte Orientale, 28100 Novara, Italy
| | - Gianluca Aimaretti
- Endocrinology, Department of Translational Medicine, University of Piemonte Orientale, 28100 Novara, Italy
- Department of Health Sciences, University of Piemonte Orientale, 28100 Novara, Italy
| | - Ivana Rabbone
- SCDU of Pediatrics, Department of Health Sciences, University of Piemonte Orientale, 28100 Novara, Italy
| | - Simonetta Bellone
- SCDU of Pediatrics, Department of Health Sciences, University of Piemonte Orientale, 28100 Novara, Italy
| | - Flavia Prodam
- Correspondence: Flavia Prodam, MD, PhD, Department of Health Sciences, SCDU Endocrinology, University of Piemonte Orientale, Via Solaroli 17, 28100, Novara, Italy.
| |
Collapse
|
27
|
Dinh HA, Stölting G, Scholl UI. Ca V3.2 (CACNA1H) in Primary Aldosteronism. Handb Exp Pharmacol 2023. [PMID: 37311830 DOI: 10.1007/164_2023_660] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/15/2023]
Abstract
Aldosterone is a steroid hormone produced in the zona glomerulosa (ZG) of the adrenal cortex. The most prominent function of aldosterone is the control of electrolyte homeostasis and blood pressure via the kidneys. The primary factors regulating aldosterone synthesis are the serum concentrations of angiotensin II and potassium. The T-type voltage-gated calcium channel CaV3.2 (encoded by CACNA1H) is an important component of electrical as well as intracellular calcium oscillations, which govern aldosterone production in the ZG. Excessive aldosterone production that is (partially) uncoupled from physiological stimuli leads to primary aldosteronism, the most common cause of secondary hypertension. Germline gain-of-function mutations in CACNA1H were identified in familial hyperaldosteronism, whereas somatic mutations are a rare cause of aldosterone-producing adenomas. In this review, we summarize these findings, put them in perspective, and highlight missing knowledge.
Collapse
Affiliation(s)
- Hoang An Dinh
- Berlin Institute of Health at Charité - Universitätsmedizin Berlin, Center of Functional Genomics, Berlin, Germany
| | - Gabriel Stölting
- Berlin Institute of Health at Charité - Universitätsmedizin Berlin, Center of Functional Genomics, Berlin, Germany
| | - Ute I Scholl
- Berlin Institute of Health at Charité - Universitätsmedizin Berlin, Center of Functional Genomics, Berlin, Germany.
- Department of Nephrology and Medical Intensive Care, Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany.
| |
Collapse
|
28
|
Wu X, Azizan EAB, Goodchild E, Garg S, Hagiyama M, Cabrera CP, Fernandes-Rosa FL, Boulkroun S, Kuan JL, Tiang Z, David A, Murakami M, Mein CA, Wozniak E, Zhao W, Marker A, Buss F, Saleeb RS, Salsbury J, Tezuka Y, Satoh F, Oki K, Udager AM, Cohen DL, Wachtel H, King PJ, Drake WM, Gurnell M, Ceral J, Ryska A, Mustangin M, Wong YP, Tan GC, Solar M, Reincke M, Rainey WE, Foo RS, Takaoka Y, Murray SA, Zennaro MC, Beuschlein F, Ito A, Brown MJ. Somatic mutations of CADM1 in aldosterone-producing adenomas and gap junction-dependent regulation of aldosterone production. Nat Genet 2023; 55:1009-1021. [PMID: 37291193 PMCID: PMC10260400 DOI: 10.1038/s41588-023-01403-0] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2022] [Accepted: 04/20/2023] [Indexed: 06/10/2023]
Abstract
Aldosterone-producing adenomas (APAs) are the commonest curable cause of hypertension. Most have gain-of-function somatic mutations of ion channels or transporters. Herein we report the discovery, replication and phenotype of mutations in the neuronal cell adhesion gene CADM1. Independent whole exome sequencing of 40 and 81 APAs found intramembranous p.Val380Asp or p.Gly379Asp variants in two patients whose hypertension and periodic primary aldosteronism were cured by adrenalectomy. Replication identified two more APAs with each variant (total, n = 6). The most upregulated gene (10- to 25-fold) in human adrenocortical H295R cells transduced with the mutations (compared to wildtype) was CYP11B2 (aldosterone synthase), and biological rhythms were the most differentially expressed process. CADM1 knockdown or mutation inhibited gap junction (GJ)-permeable dye transfer. GJ blockade by Gap27 increased CYP11B2 similarly to CADM1 mutation. Human adrenal zona glomerulosa (ZG) expression of GJA1 (the main GJ protein) was patchy, and annular GJs (sequelae of GJ communication) were less prominent in CYP11B2-positive micronodules than adjacent ZG. Somatic mutations of CADM1 cause reversible hypertension and reveal a role for GJ communication in suppressing physiological aldosterone production.
Collapse
Affiliation(s)
- Xilin Wu
- Endocrine Hypertension, Department of Clinical Pharmacology and Precision Medicine, William Harvey Research Institute, Queen Mary University of London, London, UK
- NIHR Barts Biomedical Research Centre, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London, UK
- St Bartholomew's Hospital, Barts Health NHS Trust, London, UK
| | - Elena A B Azizan
- Endocrine Hypertension, Department of Clinical Pharmacology and Precision Medicine, William Harvey Research Institute, Queen Mary University of London, London, UK.
- Department of Medicine, Faculty of Medicine, Universiti Kebangsaan Malaysia, Kuala Lumpur, Malaysia.
| | - Emily Goodchild
- Endocrine Hypertension, Department of Clinical Pharmacology and Precision Medicine, William Harvey Research Institute, Queen Mary University of London, London, UK
- NIHR Barts Biomedical Research Centre, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London, UK
- St Bartholomew's Hospital, Barts Health NHS Trust, London, UK
| | - Sumedha Garg
- Endocrine Hypertension, Department of Clinical Pharmacology and Precision Medicine, William Harvey Research Institute, Queen Mary University of London, London, UK
- Clinical Pharmacology Unit, University of Cambridge, Cambridge, UK
| | - Man Hagiyama
- Department of Pathology, Faculty of Medicine, Kindai University, Osakasayama, Japan
| | - Claudia P Cabrera
- NIHR Barts Biomedical Research Centre, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London, UK
- Centre for Translational Bioinformatics, William Harvey Research Institute, Queen Mary University of London, London, UK
| | | | | | - Jyn Ling Kuan
- Cardiovascular Disease Translational Research Programme, Department of Medicine, National University of Singapore, Singapore, Singapore
| | - Zenia Tiang
- Cardiovascular Disease Translational Research Programme, Department of Medicine, National University of Singapore, Singapore, Singapore
| | - Alessia David
- Centre for Bioinformatics, Department of Life Sciences, Imperial College London, London, UK
| | - Masanori Murakami
- Medizinische Klinik und Poliklinik IV, Klinikum der Universität, Ludwig-Maximilians-Universität München, Munich, Germany
| | - Charles A Mein
- Barts and London Genome Centre, School of Medicine and Dentistry, Blizard Institute, London, UK
| | - Eva Wozniak
- Barts and London Genome Centre, School of Medicine and Dentistry, Blizard Institute, London, UK
| | - Wanfeng Zhao
- Department of Histopathology, Addenbrooke's Hospital, Cambridge, UK
| | - Alison Marker
- Department of Histopathology, Addenbrooke's Hospital, Cambridge, UK
| | - Folma Buss
- Cambridge Institute for Medical Research, The Keith Peters Building, University of Cambridge, Cambridge, UK
| | - Rebecca S Saleeb
- Centre for Microvascular Research, William Harvey Research Institute, Queen Mary University of London, London, UK
| | - Jackie Salsbury
- Endocrine Hypertension, Department of Clinical Pharmacology and Precision Medicine, William Harvey Research Institute, Queen Mary University of London, London, UK
- NIHR Barts Biomedical Research Centre, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London, UK
- St Bartholomew's Hospital, Barts Health NHS Trust, London, UK
| | - Yuta Tezuka
- Division of Nephrology, Endocrinology, and Vascular Medicine, Tohoku University Hospital, Sendai, Japan
| | - Fumitoshi Satoh
- Division of Nephrology, Endocrinology, and Vascular Medicine, Tohoku University Hospital, Sendai, Japan
- Division of Clinical Hypertension, Endocrinology and Metabolism, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Kenji Oki
- Department of Molecular and Internal Medicine, Graduate School of Biomedical and Health Sciences, Hiroshima University, Hiroshima, Japan
| | - Aaron M Udager
- Department of Pathology, University of Michigan Medical School, Ann Arbor, MI, USA
| | - Debbie L Cohen
- Renal Division, Department of Medicine, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, USA
| | - Heather Wachtel
- Department of Surgery, Hospital of the University of Pennsylvania, Philadelphia, PA, USA
| | - Peter J King
- Department of Endocrinology, William Harvey Research Institute, Queen Mary University of London, London, UK
| | - William M Drake
- NIHR Barts Biomedical Research Centre, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London, UK
- St Bartholomew's Hospital, Barts Health NHS Trust, London, UK
| | - Mark Gurnell
- Metabolic Research Laboratories, Welcome Trust-MRC Institute of Metabolic Science, and NIHR Cambridge Biomedical Research Centre, Cambridge Biomedical Campus, Cambridge, UK
| | - Jiri Ceral
- 1st Department of Internal Medicine-Cardioangiology, Charles University Faculty of Medicine in Hradec Kralove and University Hospital Hradec Kralove, Hradec Kralove, Czech Republic
| | - Ales Ryska
- Department of Pathology, Charles University Faculty of Medicine in Hradec Kralove and University Hospital Hradec Kralove, Hradec Kralove, Czech Republic
| | - Muaatamarulain Mustangin
- Department of Pathology, Faculty of Medicine, Universiti Kebangsaan Malaysia, Kuala Lumpur, Malaysia
| | - Yin Ping Wong
- Department of Pathology, Faculty of Medicine, Universiti Kebangsaan Malaysia, Kuala Lumpur, Malaysia
| | - Geok Chin Tan
- Department of Pathology, Faculty of Medicine, Universiti Kebangsaan Malaysia, Kuala Lumpur, Malaysia
| | - Miroslav Solar
- 1st Department of Internal Medicine-Cardioangiology, Charles University Faculty of Medicine in Hradec Kralove and University Hospital Hradec Kralove, Hradec Kralove, Czech Republic
| | - Martin Reincke
- Medizinische Klinik und Poliklinik IV, Klinikum der Universität, Ludwig-Maximilians-Universität München, Munich, Germany
| | - William E Rainey
- Division of Metabolism, Endocrinology, and Diabetes, University of Michigan, Ann Arbor, MI, USA
| | - Roger S Foo
- Cardiovascular Disease Translational Research Programme, Department of Medicine, National University of Singapore, Singapore, Singapore
| | - Yutaka Takaoka
- Department of Computational Drug Design and Mathematical Medicine, Graduate School of Medicine and Pharmaceutical Sciences, University of Toyama, Toyoma, Japan
| | - Sandra A Murray
- Department of Cell Biology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Maria-Christina Zennaro
- Université Paris Cité, PARCC, Inserm, Paris, France
- Assistance Publique-Hôpitaux de Paris, Hôpital Européen Georges Pompidou, Service de Génétique, Paris, France
| | - Felix Beuschlein
- Klinik für Endokrinologie, Diabetologie und Klinische Ernährung, UniversitätsSpital Zürich (USZ) und Universität Zürich (UZH), Zurich, Switzerland
- Medizinische Klinik und Poliklinik IV, Klinikum der Universität, Ludwig-Maximilians-Universität München, Munich, Germany
| | - Akihiko Ito
- Department of Pathology, Faculty of Medicine, Kindai University, Osakasayama, Japan
| | - Morris J Brown
- Endocrine Hypertension, Department of Clinical Pharmacology and Precision Medicine, William Harvey Research Institute, Queen Mary University of London, London, UK.
- NIHR Barts Biomedical Research Centre, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London, UK.
| |
Collapse
|
29
|
Fujii W, Shibata S. Mineralocorticoid Receptor Antagonists for Preventing Chronic Kidney Disease Progression: Current Evidence and Future Challenges. Int J Mol Sci 2023; 24:ijms24097719. [PMID: 37175424 PMCID: PMC10178637 DOI: 10.3390/ijms24097719] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2023] [Revised: 04/05/2023] [Accepted: 04/20/2023] [Indexed: 05/15/2023] Open
Abstract
Regulation and action of the mineralocorticoid receptor (MR) have been the focus of intensive research over the past 80 years. Genetic and physiological/biochemical analysis revealed how MR and the steroid hormone aldosterone integrate the responses of distinct tubular cells in the face of environmental perturbations and how their dysregulation compromises fluid homeostasis. In addition to these roles, the accumulation of data also provided unequivocal evidence that MR is involved in the pathophysiology of kidney diseases. Experimental studies delineated the diverse pathological consequences of MR overactivity and uncovered the multiple mechanisms that result in enhanced MR signaling. In parallel, clinical studies consistently demonstrated that MR blockade reduces albuminuria in patients with chronic kidney disease. Moreover, recent large-scale clinical studies using finerenone have provided evidence that the non-steroidal MR antagonist can retard the kidney disease progression in diabetic patients. In this article, we review experimental data demonstrating the critical importance of MR in mediating renal injury as well as clinical studies providing evidence on the renoprotective effects of MR blockade. We also discuss areas of future investigation, which include the benefit of non-steroidal MR antagonists in non-diabetic kidney disease patients, the identification of surrogate markers for MR signaling in the kidney, and the search for key downstream mediators whereby MR blockade confers renoprotection. Insights into these questions would help maximize the benefit of MR blockade in subjects with kidney diseases.
Collapse
Affiliation(s)
- Wataru Fujii
- Division of Nephrology, Department of Internal Medicine, Graduate School of Medicine, Teikyo University, Tokyo 173-8605, Japan
| | - Shigeru Shibata
- Division of Nephrology, Department of Internal Medicine, Graduate School of Medicine, Teikyo University, Tokyo 173-8605, Japan
| |
Collapse
|
30
|
Fernandes-Rosa FL, Boulkroun S, Fedlaoui B, Hureaux M, Travers-Allard S, Drossart T, Favier J, Zennaro MC. New advances in endocrine hypertension: from genes to biomarkers. Kidney Int 2023; 103:485-500. [PMID: 36646167 DOI: 10.1016/j.kint.2022.12.021] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2022] [Revised: 12/16/2022] [Accepted: 12/19/2022] [Indexed: 01/15/2023]
Abstract
Hypertension (HT) is a major cardiovascular risk factor that affects 10% to 40% of the general population in an age-dependent manner. Detection of secondary forms of HT is particularly important because it allows the targeted management of the underlying disease. Among hypertensive patients, the prevalence of endocrine HT reaches up to 10%. Adrenal diseases are the most frequent cause of endocrine HT and are associated with excess production of mineralocorticoids (mainly primary aldosteronism), glucocorticoids (Cushing syndrome), and catecholamines (pheochromocytoma). In addition, a few rare diseases directly affecting the action of mineralocorticoids and glucocorticoids in the kidney also lead to endocrine HT. Over the past years, genomic and genetic studies have allowed improving our knowledge on the molecular mechanisms of endocrine HT. Those discoveries have opened new opportunities to transfer knowledge to clinical practice for better diagnosis and specific treatment of affected subjects. In this review, we describe the physiology of adrenal hormone biosynthesis and action, the clinical and biochemical characteristics of different forms of endocrine HT, and their underlying genetic defects. We discuss the impact of these discoveries on diagnosis and management of patients, as well as new perspectives related to the use of new biomarkers for improved patient care.
Collapse
Affiliation(s)
| | | | | | - Marguerite Hureaux
- Université Paris Cité, PARCC, Inserm, Paris, France; Assistance Publique-Hôpitaux de Paris, Hôpital Européen Georges Pompidou, Service de Génétique, Paris, France
| | - Simon Travers-Allard
- Université Paris Cité, PARCC, Inserm, Paris, France; Assistance Publique-Hôpitaux de Paris, Hôpital Européen Georges Pompidou, Service de Physiologie, Paris, France
| | - Tom Drossart
- Université Paris Cité, PARCC, Inserm, Paris, France; Université de Paris Cité, PARCC, Inserm, Equipe Labellisée par la Ligue contre le Cancer, Paris, France
| | - Judith Favier
- Université Paris Cité, PARCC, Inserm, Paris, France; Université de Paris Cité, PARCC, Inserm, Equipe Labellisée par la Ligue contre le Cancer, Paris, France
| | - Maria-Christina Zennaro
- Université Paris Cité, PARCC, Inserm, Paris, France; Assistance Publique-Hôpitaux de Paris, Hôpital Européen Georges Pompidou, Service de Génétique, Paris, France.
| |
Collapse
|
31
|
Autoantibodies to PAX5, PTCH1, and GNA11 as Serological Biomarkers in the Detection of Hepatocellular Carcinoma in Hispanic Americans. Int J Mol Sci 2023; 24:ijms24043721. [PMID: 36835134 PMCID: PMC9959316 DOI: 10.3390/ijms24043721] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2023] [Revised: 02/07/2023] [Accepted: 02/10/2023] [Indexed: 02/16/2023] Open
Abstract
Studies have demonstrated that autoantibodies to tumor-associated antigens (TAAs) may be used as efficient biomarkers with low-cost and highly sensitive characteristics. In this study, an enzyme-linked immunosorbent assay (ELISA) was conducted to analyze autoantibodies to paired box protein Pax-5 (PAX5), protein patched homolog 1 (PTCH1), and guanine nucleotide-binding protein subunit alpha-11 (GNA11) in sera from Hispanic Americans including hepatocellular carcinoma (HCC) patients, patients with liver cirrhosis (LC), patients with chronic hepatitis (CH), as well as normal controls. Meanwhile, 33 serial sera from eight HCC patients before and after diagnosis were used to explore the potential of these three autoantibodies as early biomarkers. In addition, an independent non-Hispanic cohort was used to evaluate the specificity of these three autoantibodies. In the Hispanic cohort, at the 95.0% specificity for healthy controls, 52.0%, 44.0%, and 44.0% of HCC patients showed significantly elevated levels of autoantibodies to PAX5, PTCH1, and GNA11, respectively. Among patients with LC, the frequencies for autoantibodies to PAX5, PTCH1, and GNA11 were 32.1%, 35.7%, and 25.0%, respectively. The area under the ROC curves (AUCs) of autoantibodies to PAX5, PTCH1, and GNA11 for identifying HCC from healthy controls were 0.908, 0.924, and 0.913, respectively. When these three autoantibodies were combined as a panel, the sensitivity could be improved to 68%. The prevalence of PAX5, PTCH1, and GNA11 autoantibodies has already occurred in 62.5%, 62.5%, or 75.0% of patients before clinical diagnosis, respectively. In the non-Hispanic cohort, autoantibodies to PTCH1 showed no significant difference; however, autoantibodies to PAX5, PTCH1, and GNA11 showed potential value as biomarkers for early detection of HCC in the Hispanic population and they may monitor the transition of patients with high-risk (LC, CH) to HCC. Using a panel of the three anti-TAA autoantibodies may enhance the detection of HCC.
Collapse
|
32
|
Clifton-Bligh RJ. The diagnosis and management of pheochromocytoma and paraganglioma during pregnancy. Rev Endocr Metab Disord 2023; 24:49-56. [PMID: 36637675 PMCID: PMC9884650 DOI: 10.1007/s11154-022-09773-2] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 11/24/2022] [Indexed: 01/14/2023]
Abstract
Diagnosis of pheochromocytoma or paraganglioma (PPGL) in pregnancy has been associated historically with high rates of materno-fetal morbidity and mortality. Recent evidence suggests outcomes are improved by recognition of PPGL before or during pregnancy and appropriate medical management with alpha-blockade. Whether antepartum surgery (before the third trimester) is required remains controversial and open to case-based merits. Women with PPGL in pregnancy are more commonly delivered by Caesarean section, although vaginal delivery appears to be safe in selected cases. At least some PPGLs express the luteinizing hormone/chorionic gonadotropin receptor (LHCGR) which may explain their dramatic manifestation in pregnancy. PPGLs in pregnancy are often associated with heritable syndromes, and genetic counselling and testing should be offered routinely in this setting. Since optimal outcomes are only achieved by early recognition of PPGL in (or ideally before) pregnancy, it is incumbent for clinicians to be aware of this diagnosis in a pregnant woman with hypertension occurring before 20 weeks' gestation, and acute and/or refractory hypertension particularly if paroxysmal and accompanied by sweating, palpitations and/or headaches. All women with a past history of PPGL and/or heritable PPGL syndrome should be carefully assessed for the presence of residual or recurrent disease before considering pregnancy.
Collapse
Affiliation(s)
- Roderick J Clifton-Bligh
- University of Sydney, Sydney, NSW, Australia.
- Department of Endocrinology, Royal North Shore Hospital, St Leonards, NSW, 2065, Australia.
| |
Collapse
|
33
|
Abstract
Tightly controlled Ca2+ influx through voltage-gated Ca2+ channels (Cavs) is indispensable for proper physiological function. Thus, it is not surprising that Cav loss and/or gain of function have been implicated in human pathology. Deficiency of Cav1.3 L-type Ca2+ channels (LTCCs) causes deafness and bradycardia, whereas several genetic variants of CACNA1D, the gene encoding the pore-forming α1 subunit of Cav1.3, have been linked to various disease phenotypes, such as hypertension, congenital hypoglycemia, or autism. These variants include not only common polymorphisms associated with an increased disease risk, but also rare de novo missense variants conferring high risk. This review provides a concise summary of disease-associated CACNA1D variants, whereas the main focus lies on de novo germline variants found in individuals with a neurodevelopmental disorder of variable severity. Electrophysiological recordings revealed activity-enhancing gating changes induced by these de novo variants, and tools to predict their pathogenicity and to study the resulting pathophysiological consequences will be discussed. Despite the low number of affected patients, potential phenotype-genotype correlations and factors that could impact the severity of symptoms will be covered. Since increased channel activity is assumed as the disease-underlying mechanism, pharmacological inhibition could be a treatment option. In the absence of Cav1.3-selective blockers, dihydropyridine LTCC inhibitors clinically approved for the treatment of hypertension may be used for personalized off-label trials. Findings from in vitro studies and treatment attempts in some of the patients seem promising as outlined. Taken together, due to advances in diagnostic sequencing techniques the number of reported CACNA1D variants in human diseases is constantly rising. Evidence from in silico, in vitro, and in vivo disease models can help to predict the pathogenic potential of such variants and to guide diagnosis and treatment in the clinical practice when confronted with patients harboring CACNA1D variants.
Collapse
Affiliation(s)
- Nadine J Ortner
- Department of Pharmacology and Toxicology, Institute of Pharmacy, Center for Molecular Biosciences Innsbruck, University of Innsbruck, Innsbruck, Austria.
| |
Collapse
|
34
|
Wu X, Senanayake R, Goodchild E, Bashari WA, Salsbury J, Cabrera CP, Argentesi G, O’Toole SM, Matson M, Koo B, Parvanta L, Hilliard N, Kosmoliaptsis V, Marker A, Berney DM, Tan W, Foo R, Mein CA, Wozniak E, Savage E, Sahdev A, Bird N, Laycock K, Boros I, Hader S, Warnes V, Gillett D, Dawnay A, Adeyeye E, Prete A, Taylor AE, Arlt W, Bhuva AN, Aigbirhio F, Manisty C, McIntosh A, McConnachie A, Cruickshank JK, Cheow H, Gurnell M, Drake WM, Brown MJ. [ 11C]metomidate PET-CT versus adrenal vein sampling for diagnosing surgically curable primary aldosteronism: a prospective, within-patient trial. Nat Med 2023; 29:190-202. [PMID: 36646800 PMCID: PMC9873572 DOI: 10.1038/s41591-022-02114-5] [Citation(s) in RCA: 42] [Impact Index Per Article: 21.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2021] [Accepted: 10/31/2022] [Indexed: 01/18/2023]
Abstract
Primary aldosteronism (PA) due to a unilateral aldosterone-producing adenoma is a common cause of hypertension. This can be cured, or greatly improved, by adrenal surgery. However, the invasive nature of the standard pre-surgical investigation contributes to fewer than 1% of patients with PA being offered the chance of a cure. The primary objective of our prospective study of 143 patients with PA ( NCT02945904 ) was to compare the accuracy of a non-invasive test, [11C]metomidate positron emission tomography computed tomography (MTO) scanning, with adrenal vein sampling (AVS) in predicting the biochemical remission of PA and the resolution of hypertension after surgery. A total of 128 patients reached 6- to 9-month follow-up, with 78 (61%) treated surgically and 50 (39%) managed medically. Of the 78 patients receiving surgery, 77 achieved one or more PA surgical outcome criterion for success. The accuracies of MTO at predicting biochemical and clinical success following adrenalectomy were, respectively, 72.7 and 65.4%. For AVS, the accuracies were 63.6 and 61.5%. MTO was not significantly superior, but the differences of 9.1% (95% confidence interval = -6.5 to 24.1%) and 3.8% (95% confidence interval = -11.9 to 9.4) lay within the pre-specified -17% margin for non-inferiority (P = 0.00055 and P = 0.0077, respectively). Of 24 serious adverse events, none was considered related to either investigation and 22 were fully resolved. MTO enables non-invasive diagnosis of unilateral PA.
Collapse
Affiliation(s)
- Xilin Wu
- grid.4868.20000 0001 2171 1133Endocrine Hypertension, Department of Clinical Pharmacology, William Harvey Research Institute, Queen Mary University of London, London, United Kingdom ,grid.4868.20000 0001 2171 1133NIHR Barts Cardiovascular Biomedical Research Centre, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London, United Kingdom ,grid.139534.90000 0001 0372 5777Department of Endocrinology, St Bartholomew’s Hospital, Barts Health NHS Trust, London, United Kingdom
| | - Russell Senanayake
- grid.5335.00000000121885934Metabolic Research Laboratories, Wellcome–MRC Institute of Metabolic Science, University of Cambridge, Cambridge, United Kingdom ,grid.24029.3d0000 0004 0383 8386NIHR Cambridge Biomedical Research Centre, Addenbrooke’s Hospital, Cambridge University Hospitals NHS Foundation Trust, Cambridge, United Kingdom ,grid.24029.3d0000 0004 0383 8386Department of Diabetes and Endocrinology, Addenbrooke’s Hospital, Cambridge University Hospitals NHS Foundation Trust, Cambridge, United Kingdom
| | - Emily Goodchild
- grid.4868.20000 0001 2171 1133Endocrine Hypertension, Department of Clinical Pharmacology, William Harvey Research Institute, Queen Mary University of London, London, United Kingdom ,grid.4868.20000 0001 2171 1133NIHR Barts Cardiovascular Biomedical Research Centre, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London, United Kingdom ,grid.139534.90000 0001 0372 5777Department of Endocrinology, St Bartholomew’s Hospital, Barts Health NHS Trust, London, United Kingdom
| | - Waiel A. Bashari
- grid.5335.00000000121885934Metabolic Research Laboratories, Wellcome–MRC Institute of Metabolic Science, University of Cambridge, Cambridge, United Kingdom ,grid.24029.3d0000 0004 0383 8386NIHR Cambridge Biomedical Research Centre, Addenbrooke’s Hospital, Cambridge University Hospitals NHS Foundation Trust, Cambridge, United Kingdom ,grid.24029.3d0000 0004 0383 8386Department of Diabetes and Endocrinology, Addenbrooke’s Hospital, Cambridge University Hospitals NHS Foundation Trust, Cambridge, United Kingdom
| | - Jackie Salsbury
- grid.4868.20000 0001 2171 1133Endocrine Hypertension, Department of Clinical Pharmacology, William Harvey Research Institute, Queen Mary University of London, London, United Kingdom ,grid.4868.20000 0001 2171 1133NIHR Barts Cardiovascular Biomedical Research Centre, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London, United Kingdom
| | - Claudia P. Cabrera
- grid.4868.20000 0001 2171 1133Centre for Translational Bioinformatics, William Harvey Research Institute, Queen Mary University of London, London, United Kingdom
| | - Giulia Argentesi
- grid.4868.20000 0001 2171 1133Endocrine Hypertension, Department of Clinical Pharmacology, William Harvey Research Institute, Queen Mary University of London, London, United Kingdom ,grid.4868.20000 0001 2171 1133NIHR Barts Cardiovascular Biomedical Research Centre, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London, United Kingdom ,grid.139534.90000 0001 0372 5777Department of Endocrinology, St Bartholomew’s Hospital, Barts Health NHS Trust, London, United Kingdom
| | - Samuel M. O’Toole
- grid.4868.20000 0001 2171 1133Endocrine Hypertension, Department of Clinical Pharmacology, William Harvey Research Institute, Queen Mary University of London, London, United Kingdom ,grid.4868.20000 0001 2171 1133NIHR Barts Cardiovascular Biomedical Research Centre, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London, United Kingdom ,grid.139534.90000 0001 0372 5777Department of Endocrinology, St Bartholomew’s Hospital, Barts Health NHS Trust, London, United Kingdom ,grid.416126.60000 0004 0641 6031Department of Endocrinology, Royal Hallamshire Hospital, Sheffield, United Kingdom
| | - Matthew Matson
- grid.139534.90000 0001 0372 5777Department of Radiology, St Bartholomew’s Hospital, Barts Health NHS Trust, London, United Kingdom
| | - Brendan Koo
- grid.24029.3d0000 0004 0383 8386Department of Radiology, Addenbrooke’s Hospital, Cambridge University Hospitals NHS Foundation Trust, Cambridge, United Kingdom
| | - Laila Parvanta
- grid.139534.90000 0001 0372 5777Department of Endocrinology, St Bartholomew’s Hospital, Barts Health NHS Trust, London, United Kingdom
| | - Nick Hilliard
- grid.24029.3d0000 0004 0383 8386Department of Radiology, Addenbrooke’s Hospital, Cambridge University Hospitals NHS Foundation Trust, Cambridge, United Kingdom
| | - Vasilis Kosmoliaptsis
- grid.24029.3d0000 0004 0383 8386Department of Surgery, Addenbrooke’s Hospital, Cambridge University Hospitals NHS Foundation Trust, Cambridge, United Kingdom
| | - Alison Marker
- grid.24029.3d0000 0004 0383 8386Department of Histopathology, Addenbrooke’s Hospital, Cambridge University Hospitals NHS Foundation Trust, Cambridge, United Kingdom
| | - Daniel M. Berney
- grid.139534.90000 0001 0372 5777Department of Histopathology, St Bartholomew’s Hospital, Barts Health NHS Trust, London, United Kingdom
| | - Wilson Tan
- grid.4280.e0000 0001 2180 6431Cardiovascular Research Institute, National University of Singapore, Singapore, Singapore
| | - Roger Foo
- grid.4280.e0000 0001 2180 6431Cardiovascular Research Institute, National University of Singapore, Singapore, Singapore
| | - Charles A. Mein
- grid.4868.20000 0001 2171 1133Barts and the London Genome Centre, School of Medicine and Dentistry, Blizard Institute, London, United Kingdom
| | - Eva Wozniak
- grid.4868.20000 0001 2171 1133Barts and the London Genome Centre, School of Medicine and Dentistry, Blizard Institute, London, United Kingdom
| | - Emmanuel Savage
- grid.4868.20000 0001 2171 1133Barts and the London Genome Centre, School of Medicine and Dentistry, Blizard Institute, London, United Kingdom
| | - Anju Sahdev
- grid.139534.90000 0001 0372 5777Department of Radiology, St Bartholomew’s Hospital, Barts Health NHS Trust, London, United Kingdom
| | - Nicholas Bird
- grid.24029.3d0000 0004 0383 8386Department of Radiology, Addenbrooke’s Hospital, Cambridge University Hospitals NHS Foundation Trust, Cambridge, United Kingdom
| | - Kate Laycock
- grid.4868.20000 0001 2171 1133Endocrine Hypertension, Department of Clinical Pharmacology, William Harvey Research Institute, Queen Mary University of London, London, United Kingdom ,grid.4868.20000 0001 2171 1133NIHR Barts Cardiovascular Biomedical Research Centre, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London, United Kingdom ,grid.139534.90000 0001 0372 5777Department of Endocrinology, St Bartholomew’s Hospital, Barts Health NHS Trust, London, United Kingdom
| | - Istvan Boros
- grid.5335.00000000121885934Wolfson Brain Imaging Centre, University of Cambridge, Cambridge, United Kingdom
| | - Stefan Hader
- grid.5335.00000000121885934Wolfson Brain Imaging Centre, University of Cambridge, Cambridge, United Kingdom
| | - Victoria Warnes
- grid.24029.3d0000 0004 0383 8386Department of Nuclear Medicine, Addenbrooke’s Hospital, Cambridge University Hospitals NHS Foundation Trust, Cambridge, United Kingdom
| | - Daniel Gillett
- grid.24029.3d0000 0004 0383 8386Department of Nuclear Medicine, Addenbrooke’s Hospital, Cambridge University Hospitals NHS Foundation Trust, Cambridge, United Kingdom
| | - Anne Dawnay
- grid.139534.90000 0001 0372 5777Department of Clinical Biochemistry, St Bartholomew’s Hospital, Barts Health NHS Trust, London, United Kingdom
| | - Elizabeth Adeyeye
- grid.420545.20000 0004 0489 3985Department of Cardiovascular Medicine/Diabetes, Guy’s and St Thomas’ NHS Foundation Trust, London, United Kingdom
| | - Alessandro Prete
- grid.6572.60000 0004 1936 7486Institute of Metabolism and Systems Research, University of Birmingham, Birmingham, United Kingdom
| | - Angela E. Taylor
- grid.6572.60000 0004 1936 7486Institute of Metabolism and Systems Research, University of Birmingham, Birmingham, United Kingdom
| | - Wiebke Arlt
- grid.6572.60000 0004 1936 7486Institute of Metabolism and Systems Research, University of Birmingham, Birmingham, United Kingdom ,grid.412563.70000 0004 0376 6589NIHR Birmingham Biomedical Research Centre, University Hospitals Birmingham NHS Foundation Trust and University of Birmingham, Birmingham, UK
| | - Anish N. Bhuva
- grid.139534.90000 0001 0372 5777Department of Cardiology, St Bartholomew’s Hospital, Barts Health NHS Trust, London, United Kingdom
| | - Franklin Aigbirhio
- grid.5335.00000000121885934Wolfson Brain Imaging Centre, University of Cambridge, Cambridge, United Kingdom
| | - Charlotte Manisty
- grid.139534.90000 0001 0372 5777Department of Cardiology, St Bartholomew’s Hospital, Barts Health NHS Trust, London, United Kingdom
| | - Alasdair McIntosh
- grid.8756.c0000 0001 2193 314XRobertson Centre for Biostatistics, University of Glasgow, Glasgow, United Kingdom
| | - Alexander McConnachie
- grid.8756.c0000 0001 2193 314XRobertson Centre for Biostatistics, University of Glasgow, Glasgow, United Kingdom
| | - J. Kennedy Cruickshank
- grid.420545.20000 0004 0489 3985Department of Cardiovascular Medicine/Diabetes, Guy’s and St Thomas’ NHS Foundation Trust, London, United Kingdom ,grid.13097.3c0000 0001 2322 6764School of Life Course/Nutritional Sciences, King’s College London, London, United Kingdom
| | - Heok Cheow
- grid.24029.3d0000 0004 0383 8386Department of Radiology, Addenbrooke’s Hospital, Cambridge University Hospitals NHS Foundation Trust, Cambridge, United Kingdom
| | - Mark Gurnell
- grid.5335.00000000121885934Metabolic Research Laboratories, Wellcome–MRC Institute of Metabolic Science, University of Cambridge, Cambridge, United Kingdom ,grid.24029.3d0000 0004 0383 8386NIHR Cambridge Biomedical Research Centre, Addenbrooke’s Hospital, Cambridge University Hospitals NHS Foundation Trust, Cambridge, United Kingdom ,grid.24029.3d0000 0004 0383 8386Department of Diabetes and Endocrinology, Addenbrooke’s Hospital, Cambridge University Hospitals NHS Foundation Trust, Cambridge, United Kingdom
| | - William M. Drake
- grid.4868.20000 0001 2171 1133NIHR Barts Cardiovascular Biomedical Research Centre, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London, United Kingdom ,grid.139534.90000 0001 0372 5777Department of Endocrinology, St Bartholomew’s Hospital, Barts Health NHS Trust, London, United Kingdom
| | - Morris J. Brown
- grid.4868.20000 0001 2171 1133Endocrine Hypertension, Department of Clinical Pharmacology, William Harvey Research Institute, Queen Mary University of London, London, United Kingdom ,grid.4868.20000 0001 2171 1133NIHR Barts Cardiovascular Biomedical Research Centre, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London, United Kingdom ,grid.139534.90000 0001 0372 5777Department of Endocrinology, St Bartholomew’s Hospital, Barts Health NHS Trust, London, United Kingdom
| |
Collapse
|
35
|
Castinetti F, Guerin C, Louiset E, Lacroix A. HCG-responsive aldosteronoma with transient secretion during pregnancy confirmed through HCG-stimulated adrenal venous sampling. Front Endocrinol (Lausanne) 2023; 14:1153374. [PMID: 36926028 PMCID: PMC10011616 DOI: 10.3389/fendo.2023.1153374] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/29/2023] [Accepted: 02/16/2023] [Indexed: 03/08/2023] Open
Abstract
Primary aldosteronism can be regulated by the ectopic expression of G-protein coupled receptors in aldosteronomas or bilateral hyperplasias. We report a rare case of a young woman in whom 2 pregnancies were complicated by pre-eclampsia and 1 miscarriage. The transient primary aldosteronism during pregnancies suggested the possibility of HCG stimulated aberrant adrenal expression of LHCG receptor in her adrenal tissues. This was supported by increased aldosterone and renin suppression during 5-day HCG stimulation test outside of pregnancy. Following a second 5-day HCG stimulation test, bilateral simultaneous adrenal vein sampling identified a lateralized source of aldosterone from an 8 mm right adrenal nodule. A right laparoscopic adrenalectomy resulted in clinical and biochemical cure and allowed a further uneventful pregnancy a few years later. This case illustrates the indication to investigate for potential primary aldosteronism in woman with transient hypertension during pregnancy.
Collapse
Affiliation(s)
- Frederic Castinetti
- Department of Endocrinology, Aix Marseille University, Assistance Publique-Hopitaux de Marseille, INSERM, Marseille Medical Genetics, Marmara Institute, La Conception Hospital, Marseille, France
- *Correspondence: Frederic Castinetti,
| | - Carole Guerin
- Aix Marseille University, Assistance Publique Hopitaux de Marseille, Department of Endocrine Surgery, La Conception Hospital, Marseille, France
| | - Estelle Louiset
- Univ Rouen Normandie, INSERM, NORDIC UMR 1239, Rouen, France
| | - André Lacroix
- Division of Endocrinology, Department of Medicine, Research Center, Centre hospitalier de l’Université de Montréal (CHUM), Montréal, Québec, QC, Canada
| |
Collapse
|
36
|
Chang YY, Lee BC, Chen ZW, Tsai CH, Chang CC, Liao CW, Pan CT, Peng KY, Chou CH, Lu CC, Wu VC, Hung CS, Lin YH, TAIPAI study group. Cardiovascular and metabolic characters of KCNJ5 somatic mutations in primary aldosteronism. Front Endocrinol (Lausanne) 2023; 14:1061704. [PMID: 36950676 PMCID: PMC10025475 DOI: 10.3389/fendo.2023.1061704] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/04/2022] [Accepted: 02/22/2023] [Indexed: 03/08/2023] Open
Abstract
BACKGROUND Primary aldosteronism (PA) is the leading cause of curable endocrine hypertension, which is associated with a higher risk of cardiovascular and metabolic insults compared to essential hypertension. Aldosterone-producing adenoma (APA) is a major cause of PA, which can be treated with adrenalectomy. Somatic mutations are the main pathogenesis of aldosterone overproduction in APA, of which KCNJ5 somatic mutations are most common, especially in Asian countries. This article aimed to review the literature on the impacts of KCNJ5 somatic mutations on systemic organ damage. EVIDENCE ACQUISITION PubMed literature research using keywords combination, including "aldosterone-producing adenoma," "somatic mutations," "KCNJ5," "organ damage," "cardiovascular," "diastolic function," "metabolic syndrome," "autonomous cortisol secretion," etc. RESULTS APA patients with KCNJ5 somatic mutations are generally younger, female, have higher aldosterone levels, lower potassium levels, larger tumor size, and higher hypertension cure rate after adrenalectomy. This review focuses on the cardiovascular and metabolic aspects of KCNJ5 somatic mutations in APA patients, including left ventricular remodeling and diastolic function, abdominal aortic thickness and calcification, arterial stiffness, metabolic syndrome, abdominal adipose tissue, and correlation with autonomous cortisol secretion. Furthermore, we discuss modalities to differentiate the types of mutations before surgery. CONCLUSION KCNJ5 somatic mutations in patients with APA had higher left ventricular mass (LVM), more impaired diastolic function, thicker aortic wall, lower incidence of metabolic syndrome, and possibly a lower incidence of concurrent autonomous cortisol secretion, but better improvement in LVM, diastolic function, arterial stiffness, and aortic wall thickness after adrenalectomy compared to patients without KCNJ5 mutations.
Collapse
Affiliation(s)
- Yi-Yao Chang
- Cardiology Division of Cardiovascular Medical Center, Far Eastern Memorial Hospital, New Taipei City, Taiwan
- Graduate Institute of Medicine, Yuan Ze University, Taoyuan, Taiwan
| | - Bo-Ching Lee
- Department of Medical Imaging, National Taiwan University Hospital and National Taiwan University College of Medicine, Taipei, Taiwan
| | - Zheng-Wei Chen
- Department of Internal Medicine, National Taiwan University Hospital Yun-Lin Branch, Yun-Lin, Taiwan
| | - Cheng-Hsuan Tsai
- Division of Cardiology, Department of Internal Medicine, National Taiwan University Hospital and National Taiwan University College of Medicine, Taipei, Taiwan
| | - Chin-Chen Chang
- Department of Medical Imaging, National Taiwan University Hospital and National Taiwan University College of Medicine, Taipei, Taiwan
| | - Che-Wei Liao
- Department of Medicine, National Taiwan University Cancer Center, Taipei, Taiwan
| | - Chien-Ting Pan
- Department of Internal Medicine, National Taiwan University Hospital Yun-Lin Branch, Yun-Lin, Taiwan
| | - Kang-Yung Peng
- Division of Cardiology, Department of Internal Medicine, National Taiwan University Hospital and National Taiwan University College of Medicine, Taipei, Taiwan
| | - Chia-Hung Chou
- Department of Obstetrics and Gynecology, National Taiwan University Hospital and National Taiwan University College of Medicine, Taipei, Taiwan
| | - Ching-Chu Lu
- Department of Nuclear Medicine, National Taiwan University Hospital, Taipei, Taiwan
| | - Vin-Cent Wu
- Division of Nephrology, Department of Internal Medicine, National Taiwan University Hospital and National Taiwan University College of Medicine, Taipei, Taiwan
| | - Chi-Sheng Hung
- Division of Cardiology, Department of Internal Medicine, National Taiwan University Hospital and National Taiwan University College of Medicine, Taipei, Taiwan
- Cardiovascular Center, National Taiwan University Hospital, Taipei, Taiwan
| | - Yen-Hung Lin
- Division of Cardiology, Department of Internal Medicine, National Taiwan University Hospital and National Taiwan University College of Medicine, Taipei, Taiwan
- Cardiovascular Center, National Taiwan University Hospital, Taipei, Taiwan
- *Correspondence: Yen-Hung Lin,
| | | |
Collapse
|
37
|
Vaidya A, Hundemer GL, Nanba K, Parksook WW, Brown JM. Primary Aldosteronism: State-of-the-Art Review. Am J Hypertens 2022; 35:967-988. [PMID: 35767459 PMCID: PMC9729786 DOI: 10.1093/ajh/hpac079] [Citation(s) in RCA: 48] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2022] [Revised: 06/15/2022] [Accepted: 06/27/2022] [Indexed: 12/15/2022] Open
Abstract
We are witnessing a revolution in our understanding of primary aldosteronism (PA). In the past 2 decades, we have learned that PA is a highly prevalent syndrome that is largely attributable to pathogenic somatic mutations, that contributes to cardiovascular, metabolic, and kidney disease, and that when recognized, can be adequately treated with widely available mineralocorticoid receptor antagonists and/or surgical adrenalectomy. Unfortunately, PA is rarely diagnosed, or adequately treated, mainly because of a lack of awareness and education. Most clinicians still possess an outdated understanding of PA; from primary care physicians to hypertension specialists, there is an urgent need to redefine and reintroduce PA to clinicians with a modern and practical approach. In this state-of-the-art review, we provide readers with the most updated knowledge on the pathogenesis, prevalence, diagnosis, and treatment of PA. In particular, we underscore the public health importance of promptly recognizing and treating PA and provide pragmatic solutions to modify clinical practices to achieve this.
Collapse
Affiliation(s)
- Anand Vaidya
- Department of Medicine, Center for Adrenal Disorders, Brigham and Women’s Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Gregory L Hundemer
- Department of Medicine (Division of Nephrology) and the Ottawa Hospital Research Institute, University of Ottawa, Ottawa, Ontario, Canada
| | - Kazutaka Nanba
- Department of Endocrinology and Metabolism, National Hospital Organization Kyoto Medical Center, Kyoto, Japan
- Department of Molecular and Integrative Physiology, University of Michigan, Ann Arbor, Michigan, USA
| | - Wasita W Parksook
- Department of Medicine, Division of Endocrinology and Metabolism, and Division of General Internal Medicine, Faculty of Medicine, Chulalongkorn University, and King Chulalongkorn Memorial Hospital, Thai Red Cross Society, Bangkok, Thailand
| | - Jenifer M Brown
- Division of Cardiovascular Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, Massachusetts, USA
| |
Collapse
|
38
|
McGlacken-Byrne SM, Abdelmaksoud A, Haini M, Palm L, Ashworth M, Li J, Wang W, Wang X, Wang J, Callaghan B, Kinsler VA, Faravelli F, Dattani MT. Mosaic PRKACA duplication causing a novel and distinct phenotype of early-onset Cushing's syndrome and acral cutaneous mucinosis. Eur J Endocrinol 2022; 187:K55-K61. [PMID: 36691942 DOI: 10.1530/eje-22-0287] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/04/2022] [Revised: 09/21/2022] [Accepted: 10/17/2022] [Indexed: 02/01/2023]
Abstract
SIGNIFICANCE STATEMENT We describe a mosaic PRKACA duplication in a young infant who presented with a Carney-like complex: bilateral non-pigmented micronodular adrenal hyperplasia, severe early-onset Cushing's syndrome, and distinct acral soft tissue overgrowth due to cutaneous mucinosis. This represents a novel manifestation of PRKACA disruption and broadens the extra-adrenal phenotype of PRKACA-associated Cushing's syndrome. Our data suggest that Cushing's syndrome phenotypes arising from somatic and germline PRKACA abnormalities can exist on a spectrum. We emphasise the value of ascertaining a genetic diagnosis for PRKACA-mediated adrenal and extra-adrenal disease to guide individualised and targeted care.
Collapse
Affiliation(s)
- Sinéad M McGlacken-Byrne
- Department of Paediatric Endocrinology, Great Ormond Street Hospital for Children, London, UK
- Genetics and Genomic Medicine Programme, UCL GOS Institute of Child Health, London, UK
| | - Ashraf Abdelmaksoud
- International and Private Patient Department, Great Ormond Street Hospital NHS Foundation Trust, London, UK
| | - Mohammad Haini
- Department of Histopathology, Great Ormond Street Hospital for Children, London, UK
| | - Liina Palm
- Department of Histopathology, Great Ormond Street Hospital for Children, London, UK
| | - Michael Ashworth
- Department of Histopathology, Great Ormond Street Hospital for Children, London, UK
| | - Juan Li
- Department of Endocrinology and Metabolism, Shanghai Children's Medical Center, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Wei Wang
- Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Xiumin Wang
- Department of Endocrinology and Metabolism, Shanghai Children's Medical Center, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Jian Wang
- Department of Medical Genetics and Molecular Diagnostic Laboratory, Shanghai Children's Medical Center, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Bridget Callaghan
- International and Private Patient Department, Great Ormond Street Hospital NHS Foundation Trust, London, UK
| | - Veronica A Kinsler
- Genetics and Genomic Medicine Programme, UCL GOS Institute of Child Health, London, UK
- Department of Dermatology, Great Ormond Street Hospital for Children, London, UK
- Mosaicism and Precision Medicine Laboratory, Francis Crick Institute, London, UK
| | - Francesca Faravelli
- North East Thames Regional Genetic Service, Great Ormond Street Hospital, London, UK
| | - Mehul T Dattani
- Department of Paediatric Endocrinology, Great Ormond Street Hospital for Children, London, UK
- Genetics and Genomic Medicine Programme, UCL GOS Institute of Child Health, London, UK
| |
Collapse
|
39
|
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
|
40
|
Tetti M, Gong S, Veglio F, Reincke M, Williams TA. Primary aldosteronism: Pathophysiological mechanisms of cell death and proliferation. Front Endocrinol (Lausanne) 2022; 13:934326. [PMID: 36004349 PMCID: PMC9393369 DOI: 10.3389/fendo.2022.934326] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/02/2022] [Accepted: 07/18/2022] [Indexed: 11/17/2022] Open
Abstract
Primary aldosteronism is the most common surgically curable form of hypertension. The sporadic forms of the disorder are usually caused by aldosterone overproduction from a unilateral adrenocortical aldosterone-producing adenoma or from bilateral adrenocortical hyperplasia. The main knowledge-advances in disease pathophysiology focus on pathogenic germline and somatic variants that drive the excess aldosterone production. Less clear are the molecular and cellular mechanisms that lead to an increased mass of the adrenal cortex. However, the combined application of transcriptomics, metabolomics, and epigenetics has achieved substantial insight into these processes and uncovered the evolving complexity of disrupted cell growth mechanisms in primary aldosteronism. In this review, we summarize and discuss recent progress in our understanding of mechanisms of cell death, and proliferation in the pathophysiology of primary aldosteronism.
Collapse
Affiliation(s)
- Martina Tetti
- Medizinische Klinik und Poliklinik IV, Klinikum der Universität München, Ludwig-Maximilians-Universität (LMU) München, München, Germany
- Division of Internal Medicine and Hypertension, Department of Medical Sciences, University of Turin, Turin, Italy
| | - Siyuan Gong
- Medizinische Klinik und Poliklinik IV, Klinikum der Universität München, Ludwig-Maximilians-Universität (LMU) München, München, Germany
| | - Franco Veglio
- Division of Internal Medicine and Hypertension, Department of Medical Sciences, University of Turin, Turin, Italy
| | - Martin Reincke
- Medizinische Klinik und Poliklinik IV, Klinikum der Universität München, Ludwig-Maximilians-Universität (LMU) München, München, Germany
| | - Tracy Ann Williams
- Medizinische Klinik und Poliklinik IV, Klinikum der Universität München, Ludwig-Maximilians-Universität (LMU) München, München, Germany
- Division of Internal Medicine and Hypertension, Department of Medical Sciences, University of Turin, Turin, Italy
| |
Collapse
|
41
|
Hu M, Zhang X, Hu C, Teng T, Tang QZ. A brief overview about the adipokine: Isthmin-1. Front Cardiovasc Med 2022; 9:939757. [PMID: 35958402 PMCID: PMC9360543 DOI: 10.3389/fcvm.2022.939757] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2022] [Accepted: 06/30/2022] [Indexed: 11/24/2022] Open
Abstract
Isthmin-1 is a secreted protein with multiple capability; however, it truly attracts our attention since the definition as an adipokine in 2021, which exerts indispensable roles in various pathophysiological processes through the endocrine or autocrine manners. In this review, we summarize recent knowledge of isthmin-1, including its distribution, structure, receptor and potential function.
Collapse
Affiliation(s)
- Min Hu
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, China
- Hubei Key Laboratory of Metabolic and Chronic Diseases, Wuhan, China
| | - Xin Zhang
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, China
- Hubei Key Laboratory of Metabolic and Chronic Diseases, Wuhan, China
| | - Can Hu
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, China
- Hubei Key Laboratory of Metabolic and Chronic Diseases, Wuhan, China
| | - Teng Teng
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, China
- Hubei Key Laboratory of Metabolic and Chronic Diseases, Wuhan, China
| | - Qi-Zhu Tang
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, China
- Hubei Key Laboratory of Metabolic and Chronic Diseases, Wuhan, China
- *Correspondence: Qi-Zhu Tang
| |
Collapse
|
42
|
Abstract
Primary aldosteronism is considered the commonest cause of secondary hypertension. In affected individuals, aldosterone is produced in an at least partially autonomous fashion in adrenal lesions (adenomas, [micro]nodules or diffuse hyperplasia). Over the past decade, next-generation sequencing studies have led to the insight that primary aldosteronism is largely a genetic disorder. Sporadic cases are due to somatic mutations, mostly in ion channels and pumps, and rare cases of familial hyperaldosteronism are caused by germline mutations in an overlapping set of genes. More than 90% of aldosterone-producing adenomas carry somatic mutations in K+ channel Kir3.4 (KCNJ5), Ca2+ channel CaV1.3 (CACNA1D), alpha-1 subunit of the Na+/K+ ATPase (ATP1A1), plasma membrane Ca2+ transporting ATPase 3 (ATP2B3), Ca2+ channel CaV3.2 (CACNA1H), Cl− channel ClC-2 (CLCN2), β-catenin (CTNNB1), and/or G-protein subunits alpha q/11 (GNAQ/11). Mutations in some of these genes have also been identified in aldosterone-producing (micro)nodules, suggesting a disease continuum from a single cell, acquiring a somatic mutation, via a nodule to adenoma formation, and from a healthy state to subclinical to overt primary aldosteronism. Individual glands can have multiple such lesions, and they can occur on both glands in bilateral disease. Familial hyperaldosteronism, typically with early onset, is caused by germline mutations in steroid 11-beta hydroxylase/ aldosterone synthase (CYP11B1/2), CLCN2, KCNJ5, CACNA1H, and CACNA1D.
Collapse
Affiliation(s)
- Ute I Scholl
- Berlin Institute of Health at Charité, Universitätsmedizin Berlin, Center of Functional Genomics, Germany
| |
Collapse
|
43
|
De Sousa K, Abdellatif AB, Giscos-Douriez I, Meatchi T, Amar L, Fernandes-Rosa FL, Boulkroun S, Zennaro MC. Colocalization of Wnt/β-Catenin and ACTH Signaling Pathways and Paracrine Regulation in Aldosterone-producing Adenoma. J Clin Endocrinol Metab 2022; 107:419-434. [PMID: 34570225 DOI: 10.1210/clinem/dgab707] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/11/2021] [Indexed: 11/19/2022]
Abstract
CONTEXT Aldosterone-producing adenomas (APAs) are a common cause of primary aldosteronism (PA). Despite the discovery of somatic mutations in APA and the characterization of multiple factors regulating adrenal differentiation and function, the sequence of events leading to APA formation remains to be determined. OBJECTIVE We investigated the role of Wnt/β-catenin and adrenocorticotropin signaling, as well as elements of paracrine regulation of aldosterone biosynthesis in adrenals with APA and their relationship to intratumoral heterogeneity and mutational status. METHODS We analyzed the expression of aldosterone-synthase (CYP11B2), CYP17A1, β-catenin, melanocortin type 2 receptor (MC2R), phosphorlyated cAMP response element-binding protein (pCREB), tryptase, S100, CD34 by multiplex immunofluorescence, and immunohistochemistry-guided reverse transcription-quantitative polymerase chain reaction. Eleven adrenals with APA and 1 with micronodular hyperplasia from patients with PA were analyzed. Main outcome measures included localization of CYP11B2, CYP17A1, β-catenin, MC2R, pCREB, tryptase, S100, CD34 in APA and aldosterone-producing cell clusters (APCCs). RESULTS Immunofluorescence revealed abundant mast cells and a dense vascular network in APA, independent of mutational status. Within APA, mast cells were localized in areas expressing CYP11B2 and were rarely colocalized with nerve fibers, suggesting that their degranulation is not controlled by innervation. In these same areas, ß-catenin was activated, suggesting a zona glomerulosa cell identity. In heterogeneous APA with KCNJ5 mutations, MC2R and vascular endothelial growth factor A expression was higher in areas expressing CYP11B2. A similar pattern was observed in APCC, with high expression of CYP11B2, activated β-catenin, and numerous mast cells. CONCLUSION Our results suggest that aldosterone-producing structures in adrenals with APA share common molecular characteristics and cellular environment, despite different mutation status, suggesting common developmental mechanisms.
Collapse
Affiliation(s)
| | | | | | - Tchao Meatchi
- Université de Paris, PARCC, Inserm, 75015 Paris, France
- Assistance Publique-Hôpitaux de Paris, Hôpital Européen Georges Pompidou, Service d'Anatomie Pathologique, 75015 Paris, France
| | - Laurence Amar
- Université de Paris, PARCC, Inserm, 75015 Paris, France
- Assistance Publique-Hôpitaux de Paris, Hôpital Européen Georges Pompidou, Unité Hypertension artérielle, 75015 Paris, France
| | | | | | - Maria-Christina Zennaro
- Université de Paris, PARCC, Inserm, 75015 Paris, France
- Assistance Publique-Hôpitaux de Paris, Hôpital Européen Georges Pompidou, Service de Génétique, 75015 Paris, France
| |
Collapse
|
44
|
Rege J, Hoxie J, Liu CJ, Cash MN, Luther JM, Gellert L, Turcu AF, Else T, Giordano TJ, Udager AM, Rainey WE, Nanba K. Targeted Mutational Analysis of Cortisol-Producing Adenomas. J Clin Endocrinol Metab 2022; 107:e594-e603. [PMID: 34534321 PMCID: PMC8764218 DOI: 10.1210/clinem/dgab682] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/21/2021] [Indexed: 11/19/2022]
Abstract
CONTEXT Somatic gene mutations have been identified in only about half of cortisol-producing adenomas (CPAs). Affected genes include PRKACA, GNAS, PRKAR1A, and CTNNB1. OBJECTIVE This work aims to expand our understanding of the prevalence of somatic mutations in CPAs from patients with overt Cushing syndrome (OCS) and "subclinical" mild autonomous cortisol excess (MACE), with an immunohistochemistry (IHC)‒guided targeted amplicon sequencing approach using formalin-fixed paraffin-embedded (FFPE) tissue. METHODS We analyzed FFPE adrenal tissue from 77 patients (n = 12 men, 65 women) with either OCS (n = 32) or MACE (n = 45). Using IHC for 17α-hydroxylase/17,20-lyase (CYP17A1) and 3β-hydroxysteroid dehydrogenase (HSD3B2), we identified 78 CPAs (32 OCS CPAs and 46 MACE CPAs). Genomic DNA was isolated from the FFPE CPAs and subjected to targeted amplicon sequencing for identification of somatic mutations. RESULTS Somatic mutations were identified in 71.8% (56/78) of the CPAs. While PRKACA was the most frequently mutated gene in OCS CPAs (14/32, 43.8%), somatic genetic aberrations in CTNNB1 occurred in 56.5% (26/46) of the MACE CPAs. Most GNAS mutations were observed in MACE CPAs (5/7, 71.4%). No mutations were observed in PRKAR1A. In addition to the known mutations, we identified one previously unreported mutation in PRKACA. Two patients with MACE harbored 2 adjacent tumors within the same adrenal gland - one patient had 2 CPAs, and the other patient had a CPA and an aldosterone-producing adenoma (identified by IHC for aldosterone synthase). CONCLUSION A comprehensive FFPE IHC-guided gene-targeted sequencing approach identified somatic mutations in 71.8% of the CPAs. OCS CPAs demonstrated a distinct mutation profile compared to MACE CPAs.
Collapse
Affiliation(s)
- Juilee Rege
- Department of Molecular and Integrative Physiology, University of Michigan, Ann Arbor, Michigan 48109, USA
| | - Jessie Hoxie
- Department of Molecular and Integrative Physiology, University of Michigan, Ann Arbor, Michigan 48109, USA
| | - Chia-Jen Liu
- Department of Pathology, University of Michigan, Ann Arbor, Michigan 48109, USA
| | - Morgan N Cash
- University of Kentucky College of Medicine, Lexington, Kentucky, USA
| | - James M Luther
- Division of Clinical Pharmacology, Vanderbilt University Medical Center, Nashville, Tennessee 37232, USA
| | - Lan Gellert
- Department of Pathology, Microbiology and Immunology, Vanderbilt University Medical Center, Nashville, Tennessee 37232, USA
| | - Adina F Turcu
- Division of Metabolism, Endocrinology, and Diabetes, Department of Internal Medicine, University of Michigan, Ann Arbor, Michigan 48109, USA
| | - Tobias Else
- Division of Metabolism, Endocrinology, and Diabetes, Department of Internal Medicine, University of Michigan, Ann Arbor, Michigan 48109, USA
| | - Thomas J Giordano
- Department of Pathology, University of Michigan, Ann Arbor, Michigan 48109, USA
- Rogel Cancer Center, University of Michigan, Ann Arbor, Michigan 48109, USA
- Michigan Center for Translational Pathology, University of Michigan, Ann Arbor, Michigan 48109, USA
| | - Aaron M Udager
- Department of Pathology, University of Michigan, Ann Arbor, Michigan 48109, USA
- Rogel Cancer Center, University of Michigan, Ann Arbor, Michigan 48109, USA
- Michigan Center for Translational Pathology, University of Michigan, Ann Arbor, Michigan 48109, USA
| | - William E Rainey
- Department of Molecular and Integrative Physiology, University of Michigan, Ann Arbor, Michigan 48109, USA
- Division of Metabolism, Endocrinology, and Diabetes, Department of Internal Medicine, University of Michigan, Ann Arbor, Michigan 48109, USA
- Correspondence: William E. Rainey, PhD, Department of Molecular and Integrative Physiology, University of Michigan, Room 2560C, MSRB II, 1150 W Medical Center Dr, Ann Arbor, MI 48109-5622, USA.
| | - Kazutaka Nanba
- Department of Molecular and Integrative Physiology, University of Michigan, Ann Arbor, Michigan 48109, USA
- Department of Endocrinology and Metabolism, National Hospital Organization Kyoto Medical Center, Kyoto 612-8555, Japan
- Kazutaka Nanba, MD, Department of Molecular and Integrative Physiology, University of Michigan, 1150 W Medical Center Dr, Ann Arbor, MI, 48109, USA; Department of Endocrinology and Metabolism, National Hospital Organization Kyoto Medical Center, 1-1 Mukaihata-cho, Fukakusa, Fushimi-ku, Kyoto, 612-8555, Japan.
| |
Collapse
|
45
|
Delles C. Maternally Inherited Essential Hypertension: Adding Further Complexity to an Already Complex Condition. Am J Hypertens 2022; 35:16-18. [PMID: 34427578 DOI: 10.1093/ajh/hpab133] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2021] [Accepted: 08/24/2021] [Indexed: 12/11/2022] Open
Affiliation(s)
- Christian Delles
- Institute of Cardiovascular and Medical Sciences, University of Glasgow, Glasgow, UK
| |
Collapse
|
46
|
Williams TA, Reincke M. Pathophysiology and histopathology of primary aldosteronism. Trends Endocrinol Metab 2022; 33:36-49. [PMID: 34743804 DOI: 10.1016/j.tem.2021.10.002] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/22/2021] [Revised: 09/30/2021] [Accepted: 10/09/2021] [Indexed: 10/19/2022]
Abstract
Primary aldosteronism (PA) can be sporadic or familial and classified into unilateral and bilateral forms. Sporadic PA predominates with excessive aldosterone production usually arising from a unilateral aldosterone-producing adenoma (APA) or bilateral adrenocortical hyperplasia. Familial PA is rare and caused by germline variants, that partly correspond to somatic alterations in APAs. Classification into unilateral and bilateral PA determines the treatment approach but does not accurately mirror disease pathology. Some evidence indicates a disease continuum ranging from balanced aldosterone production from each adrenal to extreme asymmetrical bilateral aldosterone production. Nonetheless, surgical removal of the overactive adrenal in unilateral PA achieves highly successful outcomes and almost all patients are biochemically cured of their aldosteronism.
Collapse
Affiliation(s)
- Tracy Ann Williams
- Medizinische Klinik und Poliklinik IV, Klinikum der Universität München, LMU München, München, Germany; Division of Internal Medicine and Hypertension, Department of Medical Sciences, University of Turin, Turin, Italy.
| | - Martin Reincke
- Medizinische Klinik und Poliklinik IV, Klinikum der Universität München, LMU München, München, Germany
| |
Collapse
|
47
|
Abdellatif AB, Fernandes-Rosa FL, Boulkroun S, Zennaro MC. Vascular and hormonal interactions in the adrenal gland. Front Endocrinol (Lausanne) 2022; 13:995228. [PMID: 36506065 PMCID: PMC9731668 DOI: 10.3389/fendo.2022.995228] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/15/2022] [Accepted: 11/07/2022] [Indexed: 11/25/2022] Open
Abstract
Primary aldosteronism is the most common form of secondary arterial hypertension, due to excessive aldosterone production from the adrenal gland. Although somatic mutations have been identified in aldosterone producing adenoma, the exact mechanisms leading to increased cell proliferation and nodule formation remain to be established. One hypothesis is that changes in vascular supply to the adrenal cortex, due to phenomena of atherosclerosis or high blood pressure, may influence the morphology of the adrenal cortex, resulting in a compensatory growth and nodule formation in response to local hypoxia. In this review, we will summarize our knowledge on the mechanisms regulating adrenal cortex development and function, describe adrenal vascularization in normal and pathological conditions and address the mechanisms allowing the cross-talk between the hormonal and vascular components to allow the extreme tissue plasticity of the adrenal cortex in response to endogenous and exogenous stimuli. We will then address recent evidence suggesting a role for alterations in the vascular compartment that could eventually be involved in nodule formation and the development of primary aldosteronism.
Collapse
Affiliation(s)
| | | | - Sheerazed Boulkroun
- Université Paris Cité, PARCC, INSERM, Paris, France
- *Correspondence: Maria-Christina Zennaro, ; Sheerazed Boulkroun,
| | - Maria-Christina Zennaro
- Université Paris Cité, PARCC, INSERM, Paris, France
- Assistance Publique-Hôpitaux de Paris, Hôpital Européen Georges Pompidou, Service de Génétique, Paris, France
- *Correspondence: Maria-Christina Zennaro, ; Sheerazed Boulkroun,
| |
Collapse
|
48
|
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
|
49
|
Transcriptomics, Epigenetics, and Metabolomics of Primary Aldosteronism. Cancers (Basel) 2021; 13:cancers13215582. [PMID: 34771744 PMCID: PMC8583505 DOI: 10.3390/cancers13215582] [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: 08/23/2021] [Revised: 10/17/2021] [Accepted: 11/05/2021] [Indexed: 12/30/2022] Open
Abstract
INTRODUCTION Primary aldosteronism (PA) is the most common cause of endocrine hypertension, mainly caused by aldosterone-producing adenomas or hyperplasia; understanding its pathophysiological background is important in order to provide ameliorative treatment strategies. Over the past several years, significant progress has been documented in this field, in particular in the clarification of the genetic and molecular mechanisms responsible for the pathogenesis of aldosterone-producing adenomas (APAs). METHODS Systematic searches of the PubMed and Cochrane databases were performed for all human studies applying transcriptomic, epigenetic or metabolomic analyses to PA subjects. Studies involving serial analysis of gene expression and microarray, epigenetic studies with methylome analyses and micro-RNA expression profiles, and metabolomic studies focused on improving understanding of the regulation of autonomous aldosterone production in PA were all included. RESULTS In this review we summarize the main findings in this area and analyze the interplay between primary aldosteronism and several signaling pathways with differential regulation of the RNA and protein expression of several factors involved in, among others, steroidogenesis, calcium signaling, and nuclear, membrane and G-coupled protein receptors. Distinct transcriptomic and metabolomic patterns are also presented herein, depending on the mutational status of APAs. In particular, two partially opposite transcriptional and steroidogenic profiles appear to distinguish APAs carrying a KCNJ5 mutation from all other APAs, which carry different mutations. CONCLUSIONS These findings can substantially contribute to the development of personalized treatment in patients with PA.
Collapse
|