101
|
Nanba K, Blinder AR, Rege J, Hattangady NG, Else T, Liu CJ, Tomlins SA, Vats P, Kumar-Sinha C, Giordano TJ, Rainey WE. Somatic CACNA1H Mutation As a Cause of Aldosterone-Producing Adenoma. Hypertension 2020; 75:645-649. [PMID: 31983310 DOI: 10.1161/hypertensionaha.119.14349] [Citation(s) in RCA: 69] [Impact Index Per Article: 13.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Driver somatic mutations for aldosterone excess have been found in ≈90% of aldosterone-producing adenomas (APAs) using an aldosterone synthase (CYP11B2)-guided sequencing approach. In the present study, we identified a novel somatic CACNA1H mutation (c.T4289C, p.I1430T) in an APA without any currently known aldosterone-driver mutations using CYP11B2 immunohistochemistry-guided whole exome sequencing. The CACNA1H gene encodes a voltage-dependent T-type calcium channel alpha-1H subunit. Germline variants in this gene are known as a cause of familial hyperaldosteronism IV. Targeted next-generation sequencing detected identical CACNA1H variants in 2 additional APAs in a cohort of the University of Michigan, resulting in a prevalence of 4% (3/75) in APAs. We tested the functional effect of the variant on adrenal cell aldosterone production and CYP11B2 mRNA expression using the human adrenocortical HAC15 cell line with a doxycycline-inducible CACNA1HI1430T mutation. Doxycycline treatment increased CYP11B2 mRNA levels as well as aldosterone production, supporting a pathological role of the CACNA1H p.I1430T mutation on the development of primary aldosteronism. In conclusion, somatic CACNA1H mutation is a genetic cause of APAs. Although the prevalence of this mutation is low, this study will provide better understanding of molecular mechanism of inappropriate aldosterone production in APAs.
Collapse
Affiliation(s)
- Kazutaka Nanba
- From the Department of Molecular and Integrative Physiology, University of Michigan, Ann Arbor (K.N., A.R.B., J.R., W.E.R.).,Department of Endocrinology and Metabolism, National Hospital Organization Kyoto Medical Center, Kyoto, Japan (K.N.)
| | - Amy R Blinder
- From the Department of Molecular and Integrative Physiology, University of Michigan, Ann Arbor (K.N., A.R.B., J.R., W.E.R.)
| | - Juilee Rege
- From the Department of Molecular and Integrative Physiology, University of Michigan, Ann Arbor (K.N., A.R.B., J.R., W.E.R.)
| | - Namita G Hattangady
- Division of Metabolism, Endocrine, and Diabetes, Department of Internal Medicine, University of Michigan, Ann Arbor (N.G.H., T.E., T.J.G., W.E.R.)
| | - Tobias Else
- Division of Metabolism, Endocrine, and Diabetes, Department of Internal Medicine, University of Michigan, Ann Arbor (N.G.H., T.E., T.J.G., W.E.R.)
| | - Chia-Jen Liu
- Department of Pathology, University of Michigan, Ann Arbor (C.-J.L., S.A.T., P.V., C.K.-S., T.J.G.).,Rogel Cancer Center, University of Michigan, Ann Arbor (C.J.-L., S.A.T., T.J.G.).,Michigan Center for Translational Pathology, University of Michigan, Ann Arbor (C.-J.L., S.A.T., P.V., C.K.-S.)
| | - Scott A Tomlins
- Department of Pathology, University of Michigan, Ann Arbor (C.-J.L., S.A.T., P.V., C.K.-S., T.J.G.).,Rogel Cancer Center, University of Michigan, Ann Arbor (C.J.-L., S.A.T., T.J.G.).,Michigan Center for Translational Pathology, University of Michigan, Ann Arbor (C.-J.L., S.A.T., P.V., C.K.-S.)
| | - Pankaj Vats
- Department of Pathology, University of Michigan, Ann Arbor (C.-J.L., S.A.T., P.V., C.K.-S., T.J.G.).,Michigan Center for Translational Pathology, University of Michigan, Ann Arbor (C.-J.L., S.A.T., P.V., C.K.-S.)
| | - Chandan Kumar-Sinha
- Department of Pathology, University of Michigan, Ann Arbor (C.-J.L., S.A.T., P.V., C.K.-S., T.J.G.).,Michigan Center for Translational Pathology, University of Michigan, Ann Arbor (C.-J.L., S.A.T., P.V., C.K.-S.)
| | - Thomas J Giordano
- Department of Pathology, University of Michigan, Ann Arbor (C.-J.L., S.A.T., P.V., C.K.-S., T.J.G.).,Rogel Cancer Center, University of Michigan, Ann Arbor (C.J.-L., S.A.T., T.J.G.)
| | - William E Rainey
- From the Department of Molecular and Integrative Physiology, University of Michigan, Ann Arbor (K.N., A.R.B., J.R., W.E.R.).,Division of Metabolism, Endocrine, and Diabetes, Department of Internal Medicine, University of Michigan, Ann Arbor (N.G.H., T.E., T.J.G., W.E.R.)
| |
Collapse
|
102
|
Williams TA. Translational Control of Aldosterone Production in Aldosterone-Producing Adenomas. Hypertension 2019; 75:299-301. [PMID: 31865787 DOI: 10.1161/hypertensionaha.119.14196] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Affiliation(s)
- Tracy Ann Williams
- From the Medizinische Klinik und Poliklinik IV, Klinikum der Universität München, LMU München, Germany; and Division of Internal Medicine and Hypertension, Department of Medical Sciences, University of Turin, Italy (T.A.W.)
| |
Collapse
|
103
|
Turcu AF, Wannachalee T, Tsodikov A, Nanba AT, Ren J, Shields JJ, O'Day PJ, Giacherio D, Rainey WE, Auchus RJ. Comprehensive Analysis of Steroid Biomarkers for Guiding Primary Aldosteronism Subtyping. Hypertension 2019; 75:183-192. [PMID: 31786984 DOI: 10.1161/hypertensionaha.119.13866] [Citation(s) in RCA: 46] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
Adrenal vein sampling (AVS) is required to distinguish unilateral from bilateral aldosterone sources in primary aldosteronism (PA), and cortisol is used for AVS data interpretation, but cortisol has several pitfalls. In this study, we present the utility of several other steroids in PA subtyping, both during AVS, as well as in peripheral serum. We included patients with PA who underwent AVS at University of Michigan between 2012 and 2018. We used mass spectrometry to simultaneously quantify 17 steroids in adrenal veins (AV) and periphery, both at baseline and after cosyntropin administration. PA was classified as unilateral or bilateral based on a lateralization index ≥ or <4, respectively, separately for baseline and post-cosyntropin administration. Of 131 participants, AV catheterizations was deemed failed in 28 (21 %) patients (36 AVs) at baseline. Eight steroids demonstrated higher AV/periphery ratios than cortisol (P<0.01 for all); 11β-hydroxyandrostenedione, 11-deoxycortisol, and corticosterone rescued most failed baseline catheterizations. Lateralization was generally consistent when using these alternative steroids. Based on pre- and post-cosyntropin data, the remaining 103 patients were classified as: U/U, 37; B/B, 32; U/B, 20; B/U, 14. Discriminant analysis of multi-steroid panels from peripheral serum showed distinct profiles across the 4 groups, with highest aldosterone, 18-oxocortisol and 11-deoxycorticosterone in U/U patients. In conclusion, 11β-hydroxyandrostenedione and 11-deoxycortisol are superior to cortisol for AVS data interpretation. Single assay multi-steroid panels measured in peripheral serum are helpful in stratified PA subtyping and have the potential to circumvent AVS in a subset of patients with PA.
Collapse
Affiliation(s)
- Adina F Turcu
- From the Division of Metabolism, Endocrinology, and Diabetes (A.F.T., T.W., A.T.N., J.R., P.J.O., W.E.R., R.J.A.), University of Michigan, Ann Arbor
| | - Taweesak Wannachalee
- From the Division of Metabolism, Endocrinology, and Diabetes (A.F.T., T.W., A.T.N., J.R., P.J.O., W.E.R., R.J.A.), University of Michigan, Ann Arbor.,Division of Endocrinology and Metabolism, Siriraj Hospital, Mahidol University, Thailand (T.W.)
| | | | - Aya T Nanba
- From the Division of Metabolism, Endocrinology, and Diabetes (A.F.T., T.W., A.T.N., J.R., P.J.O., W.E.R., R.J.A.), University of Michigan, Ann Arbor
| | - Jianwei Ren
- From the Division of Metabolism, Endocrinology, and Diabetes (A.F.T., T.W., A.T.N., J.R., P.J.O., W.E.R., R.J.A.), University of Michigan, Ann Arbor
| | - James J Shields
- Department of Radiology (J.J.S.), University of Michigan, Ann Arbor
| | - Patrick J O'Day
- From the Division of Metabolism, Endocrinology, and Diabetes (A.F.T., T.W., A.T.N., J.R., P.J.O., W.E.R., R.J.A.), University of Michigan, Ann Arbor
| | - Donald Giacherio
- Department of Pathology (D.G.), University of Michigan, Ann Arbor
| | - William E Rainey
- From the Division of Metabolism, Endocrinology, and Diabetes (A.F.T., T.W., A.T.N., J.R., P.J.O., W.E.R., R.J.A.), University of Michigan, Ann Arbor.,Department of Molecular and Integrative Physiology (W.E.R.), University of Michigan, Ann Arbor
| | - Richard J Auchus
- From the Division of Metabolism, Endocrinology, and Diabetes (A.F.T., T.W., A.T.N., J.R., P.J.O., W.E.R., R.J.A.), University of Michigan, Ann Arbor.,Department of Pharmacology (R.J.A.), University of Michigan, Ann Arbor
| |
Collapse
|
104
|
Wannachalee T, Zhao L, Nanba K, Nanba AT, Shields JJ, Rainey WE, Auchus RJ, Turcu AF. Three Discrete Patterns of Primary Aldosteronism Lateralization in Response to Cosyntropin During Adrenal Vein Sampling. J Clin Endocrinol Metab 2019; 104:5867-5876. [PMID: 31408156 PMCID: PMC6800532 DOI: 10.1210/jc.2019-01182] [Citation(s) in RCA: 50] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/22/2019] [Accepted: 08/07/2019] [Indexed: 01/12/2023]
Abstract
CONTEXT Cosyntropin [ACTH (1-24)] stimulation during adrenal vein (AV) sampling (AVS) enhances the confidence in the success of AV cannulation and circumvents intraprocedure hormonal fluctuations. Cosyntropin's effect on primary aldosteronism (PA) lateralization, however, is controversial. OBJECTIVES To define the major patterns of time-dependent lateralization, and their determinants, after cosyntropin stimulation during AVS. METHODS We retrospectively studied patients with PA who underwent AVS before, 10, and 20 minutes after cosyntropin stimulation between 2009 and 2018. Unilateral (U) or bilateral (B) PA was determined on the basis of a lateralization index (LI) value ≥4 or <4, respectively. Available adrenal tissue underwent aldosterone synthase-guided next-generation sequencing. RESULTS PA lateralization was concordant between basal and cosyntropin-stimulated AVS in 169 of 222 patients (76%; U/U, n = 110; B/B, n = 59) and discordant in 53 patients (24%; U/B, n = 32; B/U, n = 21). Peripheral and dominant AV aldosterone concentrations and LI were highest in U/U patients and progressively lower across intermediate and B/B groups. LI response to cosyntropin increased in 27% of patients, decreased in 33%, and remained stable in 40%. Baseline aldosterone concentrations predicted the LI pattern across time (P < 0.001). Mutation status was defined in 61 patients. Most patients with KCNJ5 mutations had descending LI, whereas those with ATP1A1 and ATP2B3 mutations had ascending LI after cosyntropin stimulation. CONCLUSION Patients with severe PA lateralized robustly regardless of cosyntropin use. Cosyntropin stimulation reveals intermediate PA subtypes; its impact on LI varies with baseline aldosterone concentrations and aldosterone-driver mutations.
Collapse
Affiliation(s)
- Taweesak Wannachalee
- Division of Metabolism, Endocrinology, and Diabetes, University of Michigan, Ann Arbor, Michigan
- Division of Endocrinology and Metabolism, Siriraj Hospital, Mahidol University, Bangkok, Thailand
| | - Lili Zhao
- School of Public Health, University of Michigan, Ann Arbor, Michigan
| | - Kazutaka Nanba
- Department of Molecular and Integrative Physiology, University of Michigan, Ann Arbor, Michigan
| | - Aya T Nanba
- Division of Metabolism, Endocrinology, and Diabetes, University of Michigan, Ann Arbor, Michigan
| | - James J Shields
- Department of Radiology, University of Michigan, Ann Arbor, Michigan
| | - William E Rainey
- Division of Metabolism, Endocrinology, and Diabetes, University of Michigan, Ann Arbor, Michigan
- Department of Molecular and Integrative Physiology, University of Michigan, Ann Arbor, Michigan
| | - Richard J Auchus
- Division of Metabolism, Endocrinology, and Diabetes, University of Michigan, Ann Arbor, Michigan
- Department of Pharmacology, University of Michigan, Ann Arbor, Michigan
| | - Adina F Turcu
- Division of Metabolism, Endocrinology, and Diabetes, University of Michigan, Ann Arbor, Michigan
- Correspondence and Reprint Requests: Adina F. Turcu, MD, MS, Division of Metabolism, Endocrinology, and Diabetes, University of Michigan, 1150 West Medical Center Drive, MSRB II, 5570B, Ann Arbor, Michigan 48109. E-mail:
| |
Collapse
|
105
|
Hundemer GL, Vaidya A. Primary Aldosteronism Diagnosis and Management: A Clinical Approach. Endocrinol Metab Clin North Am 2019; 48:681-700. [PMID: 31655770 PMCID: PMC6824480 DOI: 10.1016/j.ecl.2019.08.002] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Primary aldosteronism used to be considered a rare cause of secondary hypertension. However, accruing evidence indicates that primary aldosteronism is more common than previously recognized. The implications of this increased prevalence are important to public health because autonomous aldosterone production contributes to cardiovascular disease and can be treated in a targeted manner. This article focuses on clinical approaches for diagnosing primary aldosteronism more frequently and earlier in its course, as well as practical treatment objectives to reduce the risk for incident cardiovascular disease.
Collapse
Affiliation(s)
- Gregory L Hundemer
- Division of Nephrology, Ottawa Hospital Research Institute, University of Ottawa, 501 Smyth Box 511, Ottawa, ON K1H 8L6, Canada
| | - Anand Vaidya
- Center for Adrenal Disorders, Brigham and Women's Hospital, Harvard Medical School, 221 Longwood Avenue, Boston, MA 02115, USA.
| |
Collapse
|
106
|
Nanba K, Omata K, Gomez-Sanchez CE, Stratakis CA, Demidowich AP, Suzuki M, Thompson LDR, Cohen DL, Luther JM, Gellert L, Vaidya A, Barletta JA, Else T, Giordano TJ, Tomlins SA, Rainey WE. Genetic Characteristics of Aldosterone-Producing Adenomas in Blacks. Hypertension 2019; 73:885-892. [PMID: 30739536 DOI: 10.1161/hypertensionaha.118.12070] [Citation(s) in RCA: 115] [Impact Index Per Article: 19.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
Somatic mutations have been identified in aldosterone-producing adenomas (APAs) in genes that include KCNJ5, ATP1A1, ATP2B3, and CACNA1D. Based on independent studies, there appears to be racial differences in the prevalence of somatic KCNJ5 mutations, particularly between East Asians and Europeans. Despite the high cardiovascular disease mortality of blacks, there have been no studies focusing on somatic mutations in APAs in this population. In the present study, we investigated genetic characteristics of APAs in blacks using a CYP11B2 (aldosterone synthase) immunohistochemistry-guided next-generation sequencing approach. The adrenal glands with adrenocortical adenomas from 79 black patients with primary aldosteronism were studied. Seventy-three tumors from 69 adrenal glands were confirmed to be APAs by CYP11B2 immunohistochemistry. Sixty-five of 73 APAs (89%) had somatic mutations in aldosterone-driver genes. Somatic CACNA1D mutations were the most prevalent genetic alteration (42%), followed by KCNJ5 (34%), ATP1A1 (8%), and ATP2B3 mutations (4%). CACNA1D mutations were more often observed in APAs from males than those from females (55% versus 29%, P=0.033), whereas KCNJ5 mutations were more prevalent in APAs from females compared with those from males (57% versus 13%, P<0.001). No somatic mutations in aldosterone-driver genes were identified in tumors without CYP11B2 expression. In conclusion, 89% of APAs in blacks harbor aldosterone-driving mutations, and unlike Europeans and East Asians, the most frequently mutated aldosterone-driver gene was CACNA1D. Determination of racial differences in the prevalence of aldosterone-driver gene mutations may facilitate the development of personalized medicines for patients with primary aldosteronism.
Collapse
Affiliation(s)
- Kazutaka Nanba
- From the Department of Molecular and Integrative Physiology (K.N., W.E.R.), University of Michigan, Ann Arbor
| | - Kei Omata
- Department of Pathology (K.O., T.J.G., S.A.T.), University of Michigan, Ann Arbor
| | - Celso E Gomez-Sanchez
- Endocrine and Research Service, G.V. (Sonny) Montgomery VA Medical Center, Jackson, MS (C.E.G.-S.).,Division of Endocrinology, University of Mississippi Medical Center, Jackson (C.E.G.-S.)
| | - Constantine A Stratakis
- Section of Endocrinology and Genetics, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD (C.A.S., A.P.D., M.S.)
| | - Andrew P Demidowich
- Section of Endocrinology and Genetics, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD (C.A.S., A.P.D., M.S.)
| | - Mari Suzuki
- Section of Endocrinology and Genetics, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD (C.A.S., A.P.D., M.S.)
| | - Lester D R Thompson
- Department of Pathology, Woodland Hills Medical Center, Southern California Permanente Medical Group (L.D.R.T.)
| | - Debbie L Cohen
- Renal, Electrolyte and Hypertension Division, Perelman School of Medicine, University of Pennsylvania, Philadelphia (D.L.C.)
| | - James M Luther
- Division of Clinical Pharmacology (J.M.L.), Vanderbilt University Medical Center, Nashville, TN
| | - Lan Gellert
- Department of Pathology, Microbiology and Immunology (L.G.), Vanderbilt University Medical Center, Nashville, TN
| | - Anand Vaidya
- Center for Adrenal Disorders, Division of Endocrinology, Diabetes, and Hypertension (A.V.), Brigham and Women's Hospital, Harvard Medical School, Boston, MA
| | - Justine A Barletta
- Department of Pathology (J.A.B.), Brigham and Women's Hospital, Harvard Medical School, Boston, MA
| | - Tobias Else
- Division of Metabolism, Endocrine, and Diabetes, Department of Internal Medicine (T.E., T.J.G., W.E.R.), University of Michigan, Ann Arbor
| | - Thomas J Giordano
- Department of Pathology (K.O., T.J.G., S.A.T.), University of Michigan, Ann Arbor.,Division of Metabolism, Endocrine, and Diabetes, Department of Internal Medicine (T.E., T.J.G., W.E.R.), University of Michigan, Ann Arbor.,Rogel Cancer Center (T.J.G., S.A.T.), University of Michigan, Ann Arbor
| | - Scott A Tomlins
- Department of Pathology (K.O., T.J.G., S.A.T.), University of Michigan, Ann Arbor.,Rogel Cancer Center (T.J.G., S.A.T.), University of Michigan, Ann Arbor.,Department of Urology (S.A.T.), University of Michigan, Ann Arbor.,Michigan Center for Translational Pathology (S.A.T.), University of Michigan, Ann Arbor
| | - William E Rainey
- From the Department of Molecular and Integrative Physiology (K.N., W.E.R.), University of Michigan, Ann Arbor.,Division of Metabolism, Endocrine, and Diabetes, Department of Internal Medicine (T.E., T.J.G., W.E.R.), University of Michigan, Ann Arbor
| |
Collapse
|
107
|
[Primary aldosteronism : Genetics and pathology]. DER PATHOLOGE 2019; 40:369-372. [PMID: 31705237 DOI: 10.1007/s00292-019-00682-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/25/2022]
Abstract
BACKGROUND Primary aldosteronism, the excessive production of the steroid hormone aldosterone, is the most common cause of secondary hypertension. Common subforms include bilateral adrenal hyperplasia and aldosterone-producing adenoma. OBJECTIVES The goal of this review is to summarize important publications on the genetic basis of primary aldosteronism. RESULTS Somatic mutations in the KCNJ5, CACNA1D, ATP1A1, and ATP2B3 genes have been described as causes of aldosterone-producing adenomas. They eventually all lead to increased cellular calcium influx and aldosterone production. The mechanisms of rare CTNNB1 mutations are less defined. Correlations between mutations and different histologic characteristics as well as gender and ethnicity remain unexplained. Recent publications suggest that bilateral hyperplasia is at least partially due to so-called aldosterone-producing cell clusters, often with mutations in CACNA1D. Rare familial forms show mutations in the CYP11B2, CLCN2, KCNJ5, CACNA1H, or CACNA1D genes. CONCLUSIONS These results suggest that a significant fraction of primary aldosteronism is due to somatic mutations in single genes.
Collapse
|
108
|
Genetic causes of primary aldosteronism. Exp Mol Med 2019; 51:1-12. [PMID: 31695023 PMCID: PMC6834635 DOI: 10.1038/s12276-019-0337-9] [Citation(s) in RCA: 33] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2019] [Revised: 08/21/2019] [Accepted: 09/09/2019] [Indexed: 11/09/2022] Open
Abstract
Primary aldosteronism is characterized by at least partially autonomous production of the adrenal steroid hormone aldosterone and is the most common cause of secondary hypertension. The most frequent subforms are idiopathic hyperaldosteronism and aldosterone-producing adenoma. Rare causes include unilateral hyperplasia, adrenocortical carcinoma and Mendelian forms (familial hyperaldosteronism). Studies conducted in the last eight years have identified somatic driver mutations in a substantial portion of aldosterone-producing adenomas, including the genes KCNJ5 (encoding inwardly rectifying potassium channel GIRK4), CACNA1D (encoding a subunit of L-type voltage-gated calcium channel CaV1.3), ATP1A1 (encoding a subunit of Na+/K+-ATPase), ATP2B3 (encoding a Ca2+-ATPase), and CTNNB1 (encoding ß-catenin). In addition, aldosterone-producing cells were recently reported to form small clusters (aldosterone-producing cell clusters) beneath the adrenal capsule. Such clusters accumulate with age and appear to be more frequent in individuals with idiopathic hyperaldosteronism. The fact that they are associated with somatic mutations implicated in aldosterone-producing adenomas also suggests a precursor function for adenomas. Rare germline variants of CYP11B2 (encoding aldosterone synthase), CLCN2 (encoding voltage-gated chloride channel ClC-2), KCNJ5, CACNA1H (encoding a subunit of T-type voltage-gated calcium channel CaV3.2), and CACNA1D have been reported in different subtypes of familial hyperaldosteronism. Collectively, these studies suggest that primary aldosteronism is largely due to genetic mutations in single genes, with potential implications for diagnosis and therapy.
Collapse
|
109
|
Retinoic acid receptor α as a novel contributor to adrenal cortex structure and function through interactions with Wnt and Vegfa signalling. Sci Rep 2019; 9:14677. [PMID: 31605007 PMCID: PMC6789122 DOI: 10.1038/s41598-019-50988-2] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2019] [Accepted: 09/17/2019] [Indexed: 01/09/2023] Open
Abstract
Primary aldosteronism (PA) is the most frequent form of secondary arterial hypertension. Mutations in different genes increase aldosterone production in PA, but additional mechanisms may contribute to increased cell proliferation and aldosterone producing adenoma (APA) development. We performed transcriptome analysis in APA and identified retinoic acid receptor alpha (RARα) signaling as a central molecular network involved in nodule formation. To understand how RARα modulates adrenal structure and function, we explored the adrenal phenotype of male and female Rarα knockout mice. Inactivation of Rarα in mice led to significant structural disorganization of the adrenal cortex in both sexes, with increased adrenal cortex size in female mice and increased cell proliferation in males. Abnormalities of vessel architecture and extracellular matrix were due to decreased Vegfa expression and modifications in extracellular matrix components. On the molecular level, Rarα inactivation leads to inhibition of non-canonical Wnt signaling, without affecting the canonical Wnt pathway nor PKA signaling. Our study suggests that Rarα contributes to the maintenance of normal adrenal cortex structure and cell proliferation, by modulating Wnt signaling. Dysregulation of this interaction may contribute to abnormal cell proliferation, creating a propitious environment for the emergence of specific driver mutations in PA.
Collapse
|
110
|
Vilela LAP, Rassi-Cruz M, Guimaraes AG, Moises CCS, Freitas TC, Alencar NP, Petenuci J, Goldbaum TS, Maciel AAW, Pereira MAA, Silva GV, Pio-Abreu A, Zerbini MCN, Cavalcante ACBS, Carnevale FC, Pilan B, Yamauchi F, Srougi V, Tanno FY, Chambo JL, Latronico AC, Mendonca BB, Fragoso MCBV, Bortolotto LA, Drager LF, Almeida MQ. KCNJ5 Somatic Mutation Is a Predictor of Hypertension Remission After Adrenalectomy for Unilateral Primary Aldosteronism. J Clin Endocrinol Metab 2019; 104:4695-4702. [PMID: 31216002 DOI: 10.1210/jc.2019-00531] [Citation(s) in RCA: 44] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/05/2019] [Accepted: 06/13/2019] [Indexed: 01/27/2023]
Abstract
CONTEXT Primary aldosteronism (PA) is the most common cause of endocrine hypertension (HT). HT remission (defined as blood pressure <140/90 mm Hg without antihypertensive drugs) has been reported in approximately 50% of patients with unilateral PA after adrenalectomy. HT duration and severity are predictors of blood pressure response, but the prognostic role of somatic KCNJ5 mutations is unclear. OBJECTIVE To determine clinical and molecular features associated with HT remission after adrenalectomy in patients with unilateral PA. METHODS We retrospectively evaluated 100 patients with PA (60 women; median age at diagnosis 48 years with a median follow-up of 26 months). Anatomopathological analysis revealed 90 aldosterone-producing adenomas, 1 carcinoma, and 9 unilateral adrenal hyperplasias. All patients had biochemical cure after unilateral adrenalectomy. KCNJ5 gene was sequenced in 76 cases. RESULTS KCNJ5 mutations were identified in 33 of 76 (43.4%) tumors: p.Gly151Arg (n = 17), p.Leu168Arg (n = 15), and p.Glu145Gln (n = 1). HT remission was reported in 37 of 100 (37%) patients. Among patients with HT remission, 73% were women (P = 0.04), 48.6% used more than three antihypertensive medications (P = 0.0001), and 64.9% had HT duration <10 years (P = 0.0015) compared with those without HT remission. Somatic KCNJ5 mutations were associated with female sex (P = 0.004), larger nodules (P = 0.001), and HT remission (P = 0.0001). In multivariate analysis, only a somatic KCNJ5 mutation was an independent predictor of HT remission after adrenalectomy (P = 0.004). CONCLUSION The presence of a KCNJ5 somatic mutation is an independent predictor of HT remission after unilateral adrenalectomy in patients with unilateral PA.
Collapse
Affiliation(s)
- Leticia A P Vilela
- Unidade de Suprarrenal, Laboratório de Hormônios e Genética Molecular LIM/42, Serviço de Endocrinologia e Metabologia, Hospital das Clínicas, Faculdade de Medicina da Universidade de São Paulo, São Paulo, Brazil
| | - Marcela Rassi-Cruz
- Unidade de Suprarrenal, Laboratório de Hormônios e Genética Molecular LIM/42, Serviço de Endocrinologia e Metabologia, Hospital das Clínicas, Faculdade de Medicina da Universidade de São Paulo, São Paulo, Brazil
| | - Augusto G Guimaraes
- Unidade de Suprarrenal, Laboratório de Hormônios e Genética Molecular LIM/42, Serviço de Endocrinologia e Metabologia, Hospital das Clínicas, Faculdade de Medicina da Universidade de São Paulo, São Paulo, Brazil
| | - Caio C S Moises
- Unidade de Suprarrenal, Laboratório de Hormônios e Genética Molecular LIM/42, Serviço de Endocrinologia e Metabologia, Hospital das Clínicas, Faculdade de Medicina da Universidade de São Paulo, São Paulo, Brazil
| | - Thais C Freitas
- Unidade de Suprarrenal, Laboratório de Hormônios e Genética Molecular LIM/42, Serviço de Endocrinologia e Metabologia, Hospital das Clínicas, Faculdade de Medicina da Universidade de São Paulo, São Paulo, Brazil
| | - Natalia P Alencar
- Unidade de Suprarrenal, Laboratório de Hormônios e Genética Molecular LIM/42, Serviço de Endocrinologia e Metabologia, Hospital das Clínicas, Faculdade de Medicina da Universidade de São Paulo, São Paulo, Brazil
| | - Janaina Petenuci
- Unidade de Suprarrenal, Laboratório de Hormônios e Genética Molecular LIM/42, Serviço de Endocrinologia e Metabologia, Hospital das Clínicas, Faculdade de Medicina da Universidade de São Paulo, São Paulo, Brazil
| | - Tatiana S Goldbaum
- Unidade de Suprarrenal, Laboratório de Hormônios e Genética Molecular LIM/42, Serviço de Endocrinologia e Metabologia, Hospital das Clínicas, Faculdade de Medicina da Universidade de São Paulo, São Paulo, Brazil
| | - Ana Alice W Maciel
- Unidade de Suprarrenal, Laboratório de Hormônios e Genética Molecular LIM/42, Serviço de Endocrinologia e Metabologia, Hospital das Clínicas, Faculdade de Medicina da Universidade de São Paulo, São Paulo, Brazil
| | - Maria Adelaide A Pereira
- Unidade de Suprarrenal, Laboratório de Hormônios e Genética Molecular LIM/42, Serviço de Endocrinologia e Metabologia, Hospital das Clínicas, Faculdade de Medicina da Universidade de São Paulo, São Paulo, Brazil
| | - Giovanio V Silva
- Unidade de Hipertensão, Disciplina de Nefrologia, Hospital das Clínicas, Faculdade de Medicina da Universidade de São Paulo, São Paulo, Brazil
| | - Andrea Pio-Abreu
- Unidade de Hipertensão, Disciplina de Nefrologia, Hospital das Clínicas, Faculdade de Medicina da Universidade de São Paulo, São Paulo, Brazil
| | - Maria Claudia N Zerbini
- Divisão de Anatomia Patológica, Faculdade de Medicina da Universidade de São Paulo, São Paulo, Brazil
| | - Aline C B S Cavalcante
- Instituto de Radiologia InRad, Hospital das Clínicas, Faculdade de Medicina da Universidade de São Paulo, São Paulo, Brazil
| | - Francisco C Carnevale
- Instituto de Radiologia InRad, Hospital das Clínicas, Faculdade de Medicina da Universidade de São Paulo, São Paulo, Brazil
| | - Bruna Pilan
- Instituto de Radiologia InRad, Hospital das Clínicas, Faculdade de Medicina da Universidade de São Paulo, São Paulo, Brazil
| | - Fernando Yamauchi
- Instituto de Radiologia InRad, Hospital das Clínicas, Faculdade de Medicina da Universidade de São Paulo, São Paulo, Brazil
| | - Vitor Srougi
- Serviço Urologia, Hospital das Clínicas, Faculdade de Medicina da Universidade de São Paulo, São Paulo, Brazil
| | - Fabio Y Tanno
- Serviço Urologia, Hospital das Clínicas, Faculdade de Medicina da Universidade de São Paulo, São Paulo, Brazil
| | - Jose L Chambo
- Serviço Urologia, Hospital das Clínicas, Faculdade de Medicina da Universidade de São Paulo, São Paulo, Brazil
| | - Ana Claudia Latronico
- Unidade de Suprarrenal, Laboratório de Hormônios e Genética Molecular LIM/42, Serviço de Endocrinologia e Metabologia, Hospital das Clínicas, Faculdade de Medicina da Universidade de São Paulo, São Paulo, Brazil
| | - Berenice B Mendonca
- Unidade de Suprarrenal, Laboratório de Hormônios e Genética Molecular LIM/42, Serviço de Endocrinologia e Metabologia, Hospital das Clínicas, Faculdade de Medicina da Universidade de São Paulo, São Paulo, Brazil
| | - Maria Candida B V Fragoso
- Unidade de Suprarrenal, Laboratório de Hormônios e Genética Molecular LIM/42, Serviço de Endocrinologia e Metabologia, Hospital das Clínicas, Faculdade de Medicina da Universidade de São Paulo, São Paulo, Brazil
- Servico de Endocrinologia, Instituto do Câncer do Estado de São Paulo (ICESP), Faculdade de Medicina da Universidade de São Paulo, São Paulo, Brazil
| | - Luiz A Bortolotto
- Unidade de Hipertensão, Instituto do Coração (InCor), Faculdade de Medicina da Universidade de São Paulo, São Paulo, Brazil
| | - Luciano F Drager
- Unidade de Hipertensão, Disciplina de Nefrologia, Hospital das Clínicas, Faculdade de Medicina da Universidade de São Paulo, São Paulo, Brazil
- Unidade de Hipertensão, Instituto do Coração (InCor), Faculdade de Medicina da Universidade de São Paulo, São Paulo, Brazil
| | - Madson Q Almeida
- Unidade de Suprarrenal, Laboratório de Hormônios e Genética Molecular LIM/42, Serviço de Endocrinologia e Metabologia, Hospital das Clínicas, Faculdade de Medicina da Universidade de São Paulo, São Paulo, Brazil
- Servico de Endocrinologia, Instituto do Câncer do Estado de São Paulo (ICESP), Faculdade de Medicina da Universidade de São Paulo, São Paulo, Brazil
| |
Collapse
|
111
|
Yang Y, Gomez-Sanchez CE, Jaquin D, Aristizabal Prada ET, Meyer LS, Knösel T, Schneider H, Beuschlein F, Reincke M, Williams TA. Primary Aldosteronism: KCNJ5 Mutations and Adrenocortical Cell Growth. Hypertension 2019; 74:809-816. [PMID: 31446799 DOI: 10.1161/hypertensionaha.119.13476] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Aldosterone-producing adenomas with somatic mutations in the KCNJ5 G-protein-coupled inwardly rectifying potassium channel are a cause of primary aldosteronism. These mutations drive aldosterone excess, but their role in cell growth is undefined. Our objective was to determine the role of KCNJ5 mutations in adrenal cell proliferation and apoptosis. The Ki67 proliferative index was positively correlated with adenoma diameter in aldosterone-producing adenomas with a KCNJ5 mutation (r=0.435, P=0.007), a negative correlation was noted in adenomas with no mutation detected (r=-0.548, P=0.023). Human adrenocortical cell lines were established with stable expression of cumate-inducible wild-type or mutated KCNJ5. Increased cell proliferation was induced by low-level induction of KCNJ5-T158A expression compared with control cells (P=0.009), but increased induction ablated this difference. KCNJ5-G151R displayed no apparent proliferative effect, but KCNJ5-G151E and L168R mutations each resulted in decreased cell proliferation (difference P<0.0001 from control cells, both comparisons). Under conditions tested, T158A had no effect on apoptosis, but apoptosis increased with expression of G151R (P<0.0001), G151E (P=0.008), and L168R (P<0.0001). We generated a specific KCNJ5 monoclonal antibody which was used in immunohistochemistry to demonstrate strong KCNJ5 expression in adenomas without a KCNJ5 mutation and in the zona glomerulosa adjacent to adenomas irrespective of genotype as well as in aldosterone-producing cell clusters. Double immunofluorescence staining for KCNJ5 and CYP11B2 (aldosterone synthase) showed markedly decreased KCNJ5 immunostaining in CYP11B2-positive cells compared with CYP11B2-negative cells in aldosterone-producing adenomas with a KCNJ5 mutation. Together, these findings support the concept that cell growth effects of KCNJ5 mutations are determined by the expression level of the mutated channel.
Collapse
Affiliation(s)
- Yuhong Yang
- From the Medizinische Klinik und Poliklinik IV, Klinikum der Universität München, LMU München, Germany (Y.Y., D.J., E.T.A.P., L.S.M., H.S., F.B., M.R., T.A.W.)
| | - Celso E Gomez-Sanchez
- Endocrine Division, G.V. (Sonny) Montgomery VA Medical Center, University of Mississippi Medical Center, Jackson (C.E.G.-S.)
| | - Diana Jaquin
- From the Medizinische Klinik und Poliklinik IV, Klinikum der Universität München, LMU München, Germany (Y.Y., D.J., E.T.A.P., L.S.M., H.S., F.B., M.R., T.A.W.)
| | - Elke Tatjana Aristizabal Prada
- From the Medizinische Klinik und Poliklinik IV, Klinikum der Universität München, LMU München, Germany (Y.Y., D.J., E.T.A.P., L.S.M., H.S., F.B., M.R., T.A.W.)
| | - Lucie S Meyer
- From the Medizinische Klinik und Poliklinik IV, Klinikum der Universität München, LMU München, Germany (Y.Y., D.J., E.T.A.P., L.S.M., H.S., F.B., M.R., T.A.W.)
| | - Thomas Knösel
- Institute of Pathology, Ludwig-Maximilians-Universität München, Germany (T.K.)
| | - Holger Schneider
- From the Medizinische Klinik und Poliklinik IV, Klinikum der Universität München, LMU München, Germany (Y.Y., D.J., E.T.A.P., L.S.M., H.S., F.B., M.R., T.A.W.)
| | - Felix Beuschlein
- From the Medizinische Klinik und Poliklinik IV, Klinikum der Universität München, LMU München, Germany (Y.Y., D.J., E.T.A.P., L.S.M., H.S., F.B., M.R., T.A.W.).,Klinik für Endokrinologie, Diabetologie und Klinische Ernährung, Universitätsspital Zürich, Switzerland (F.B.)
| | - Martin Reincke
- From the Medizinische Klinik und Poliklinik IV, Klinikum der Universität München, LMU München, Germany (Y.Y., D.J., E.T.A.P., L.S.M., H.S., F.B., M.R., T.A.W.)
| | - Tracy Ann Williams
- From the Medizinische Klinik und Poliklinik IV, Klinikum der Universität München, LMU München, Germany (Y.Y., D.J., E.T.A.P., L.S.M., H.S., F.B., M.R., T.A.W.).,Division of Internal Medicine and Hypertension, Department of Medical Sciences, University of Turin, Italy (T.A.W.)
| |
Collapse
|
112
|
Affiliation(s)
- John W Funder
- From the Hudson Institute of Medical Research and Monash University, Clayton, Victoria, Australia
| |
Collapse
|
113
|
Affiliation(s)
- Celso E Gomez-Sanchez
- From the Endocrine Division, G.V. (Sonny) Montgomery VA Medical Center, Jackson, MS (C.E.G.-S.).,University of Mississippi Medical Center, Jackson (C.E.G.-S.)
| | - Tracy A Williams
- Medizinische Klinik und Poliklinik IV, Klinikum der Ludwig-Maximilians-Universität München, Germany (T.A.W.).,Division of Internal Medicine and Hypertension, Department of Medical Sciences, University of Turin, Italy (T.A.W.)
| |
Collapse
|
114
|
Yang Y, Burrello J, Burrello A, Eisenhofer G, Peitzsch M, Tetti M, Knösel T, Beuschlein F, Lenders JWM, Mulatero P, Reincke M, Williams TA. Classification of microadenomas in patients with primary aldosteronism by steroid profiling. J Steroid Biochem Mol Biol 2019; 189:274-282. [PMID: 30654107 PMCID: PMC6876277 DOI: 10.1016/j.jsbmb.2019.01.008] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/13/2018] [Revised: 12/16/2018] [Accepted: 01/13/2019] [Indexed: 11/07/2022]
Abstract
In primary aldosteronism (PA) the differentiation of unilateral aldosterone-producing adenomas (APA) from bilateral adrenal hyperplasia (BAH) is usually performed by adrenal venous sampling (AVS) and/or computed tomography (CT). CT alone often lacks the sensitivity to identify micro-APAs. Our objectives were to establish if steroid profiling could be useful for the identification of patients with micro-APAs and for the development of an online tool to differentiate micro-APAs, macro-APAs and BAH. The study included patients with PA (n = 197) from Munich (n = 124) and Torino (n = 73) and comprised 33 patients with micro-APAs, 95 with macro-APAs, and 69 with BAH. Subtype differentiation was by AVS, and micro- and macro-APAs were selected according to pathology reports. Steroid concentrations in peripheral venous plasma were measured by liquid chromatography-tandem mass spectrometry. An online tool using a random forest model was built for the classification of micro-APA, macro-APA and BAH. Micro-APA were classified with low specificity (33%) but macro-APA and BAH were correctly classified with high specificity (93%). Improved classification of micro-APAs was achieved using a diagnostic algorithm integrating steroid profiling, CT scanning and AVS procedures limited to patients with discordant steroid and CT results. This would have increased the correct classification of micro-APAs to 68% and improved the overall classification to 92%. Such an approach could be useful to select patients with CT-undetectable micro-APAs in whom AVS should be considered mandatory.
Collapse
Affiliation(s)
- Yuhong Yang
- Medizinische Klinik und Poliklinik IV, Klinikum der Universität, Ludwig-Maximilians-Universität München, Munich, Germany
| | - Jacopo Burrello
- Division of Internal Medicine and Hypertension, Department of Medical Sciences, University of Turin, Turin, Italy
| | - Alessio Burrello
- Department of Electronics and Telecommunications, Polytechnic University of Turin, Turin, Italy
| | - Graeme Eisenhofer
- Institute of Clinical Chemistry and Laboratory Medicine, University Hospital Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany; Department of Medicine III, University Hospital Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany
| | - Mirko Peitzsch
- Institute of Clinical Chemistry and Laboratory Medicine, University Hospital Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany
| | - Martina Tetti
- Division of Internal Medicine and Hypertension, Department of Medical Sciences, University of Turin, Turin, Italy
| | - Thomas Knösel
- Institute of Pathology, Ludwig-Maximilians-Universität München, Munich, Germany
| | - Felix Beuschlein
- Medizinische Klinik und Poliklinik IV, Klinikum der Universität, Ludwig-Maximilians-Universität München, Munich, Germany; Klinik für Endokrinologie, Diabetologie und Klinische Ernährung, Universitätsspital Zürich, Zürich, Switzerland
| | - Jacques W M Lenders
- Department of Medicine III, University Hospital Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany; Department of Medicine, Radboud University Medical Center, Nijmegen, the Netherlands
| | - Paolo Mulatero
- 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, Ludwig-Maximilians-Universität München, Munich, Germany
| | - Tracy Ann Williams
- Medizinische Klinik und Poliklinik IV, Klinikum der Universität, Ludwig-Maximilians-Universität München, Munich, Germany; Division of Internal Medicine and Hypertension, Department of Medical Sciences, University of Turin, Turin, Italy.
| |
Collapse
|
115
|
Abstract
PURPOSE OF REVIEW The application of advanced genetic techniques has recently begun to unravel the genetic basis for familial primary aldosteronism type 2 (FH-II). RECENT FINDINGS Whole-exome sequencing in a large family with FH-II revealed a shared rare damaging heterozygous variant in CLCN2 (chr.3: g.184075850C>T, p.Arg172Gln) in three severely affected members. The gene encodes a chloride channel, ClC-2. A cohort of 80 unrelated individuals diagnosed with early-onset primary aldosteronism was also examined for CLCN2 mutations finding three further occurrences of p.Arg172Gln mutations and four single cases of other potentially damaging heterozygous mutations for an overall prevalence of 9.9%. A concurrent report also found a different CLCN2 mutation (p.Gly24Asp) in a single severely affected patient from a cohort of 12 with early-onset PA for a prevalence of 8.3%. Cases of primary aldosteronism associated with CLCN2 mutations appear to be bilateral and respond well to medical treatment. In the adrenal, ClC-2 has been demonstrated to localize predominantly to the zona glomerulosa (ZG), and functional analysis suggests that mutations in ClC-2 predispose ZG cells to depolarization, thus leading to calcium influx via activation of voltage-gated calcium channels and increased aldosterone production. Germline CLCN2 mutations appear to account for a substantial proportion of early-onset primary aldosteronism cases, and genetic testing for mutations in this gene should be considered in appropriate cases.
Collapse
|
116
|
Lana A, Alexander K, Castagna A, D'Alessandro A, Morandini F, Pizzolo F, Zorzi F, Mulatero P, Zolla L, Olivieri O. Urinary Metabolic Signature of Primary Aldosteronism: Gender and Subtype-Specific Alterations. Proteomics Clin Appl 2019; 13:e1800049. [PMID: 30580498 DOI: 10.1002/prca.201800049] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2018] [Revised: 12/03/2018] [Indexed: 12/19/2022]
Abstract
PURPOSE The current clinical investigation for primary aldosteronism (PA) diagnosis requires complex expensive tests from the initial suspicion to the final subtype classification, including invasive approaches; therefore, appropriate markers for subtype definition are greatly desirable. The present study performs a metabolomics analysis to further examine specific molecular signatures of PA urines EXPERIMENTAL DESIGN: The study considered PA subtype and gender-related differences using two orthogonal advanced UHPLC-MS metabolomics approaches. Patients with essential hypertension (n = 36) and PA (n = 50) who were referred to the outpatient hypertension clinic and matched healthy subjects (n = 10) are investigated. RESULTS Statistically significant changes (p < 0.05 ANOVA, Fc > 1.5) of metabolites involved in central carbon, energy, and nitrogen metabolism are identified, especially purine and pyrimidine nucleosides and precursors, and free amino acids. PLS-DA interpretation provides strong evidence of a disease-specific metabolic pattern with dAMP, diiodothyronine, and 5-methoxytryptophan as leading factors, and a sex-specific metabolic pattern associated with orotidine 5-phosphate, N-acetylalanine, hydroxyproline, and cysteine. The results are verified using an independent sample set, which confirms the identification of specific signatures. CONCLUSIONS AND CLINICAL RELEVANCE Metabolomics is used to identify low molecular weight molecular markers of PA, which paves the way for follow-up validation studies in larger cohorts.
Collapse
Affiliation(s)
- Alessandro Lana
- Department of Ecological and Biological Sciences, Tuscia University, 01100, Viterbo, Italy
| | - Keisha Alexander
- Departments of Biochemistry and Molecular Genetics, University of Colorado Denver, Anschutz Medical Campus, 80045, Aurora, CO, USA
| | - Annalisa Castagna
- Department of Medicine, Unit of Internal Medicine, University of Verona, 37134, Verona, Italy
| | - Angelo D'Alessandro
- Departments of Biochemistry and Molecular Genetics, University of Colorado Denver, Anschutz Medical Campus, 80045, Aurora, CO, USA
| | - Francesca Morandini
- Department of Medicine, Unit of Internal Medicine, University of Verona, 37134, Verona, Italy
| | - Francesca Pizzolo
- Department of Medicine, Unit of Internal Medicine, University of Verona, 37134, Verona, Italy
| | - Francesco Zorzi
- Department of Medicine, Unit of Internal Medicine, University of Verona, 37134, Verona, Italy
| | - Paolo Mulatero
- Department of Medicine and Experimental Oncology, Division of Internal Medicine and Hypertension Unit, University of Turin, 10124, Turin, Italy
| | - Lello Zolla
- Department of Ecological and Biological Sciences, Tuscia University, 01100, Viterbo, Italy
| | - Oliviero Olivieri
- Department of Medicine, Unit of Internal Medicine, University of Verona, 37134, Verona, Italy
| |
Collapse
|
117
|
Meyer LS, Reincke M, Williams TA. Timeline of Advances in Genetics of Primary Aldosteronism. EXPERIENTIA SUPPLEMENTUM (2012) 2019; 111:213-243. [PMID: 31588534 DOI: 10.1007/978-3-030-25905-1_11] [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: 06/10/2023]
Abstract
The overwhelming majority of cases of primary aldosteronism (PA) occur sporadically due to a unilateral aldosterone-producing adenoma (APA) or bilateral idiopathic adrenal hyperplasia. Familial forms of PA are rare with four subtypes defined to date (familial hyperaldosteronism types I-IV). The molecular basis of familial hyperaldosteronism type I (FH type I or glucocorticoid-remediable aldosteronism) was established in 1992; two decades later the genetic variant causing FH type III was identified and germline mutations causing FH type IV and FH type II were determined soon after. Effective diagnostic protocols and methods to detect the overactive gland in unilateral PA by adrenal venous sampling followed by laparoscopic adrenalectomy have made available APAs for scientific studies. In rapid succession, following the widespread use of next-generation sequencing, recurrent somatic driver mutations in APAs were identified in genes encoding ion channels and transporters. The development of highly specific monoclonal antibodies against key enzymes in adrenal steroidogenesis has unveiled the heterogeneous features of the diseased adrenal in PA and helped reveal the high proportion of APAs with driver mutations. We discuss what is known about the genetics of PA that has led to a clearer understanding of the disease pathophysiology.
Collapse
Affiliation(s)
- Lucie S Meyer
- Medizinische Klinik und Poliklinik IV, Klinikum der Universität München, Ludwig-Maximilians-Universität München, Munich, Germany
| | - Martin Reincke
- Medizinische Klinik und Poliklinik IV, Klinikum der Universität München, Ludwig-Maximilians-Universität München, Munich, Germany
| | - Tracy Ann Williams
- Medizinische Klinik und Poliklinik IV, Klinikum der Universität München, Ludwig-Maximilians-Universität München, Munich, Germany.
- Division of Internal Medicine and Hypertension, Department of Medical Sciences, University of Turin, Turin, Italy.
| |
Collapse
|
118
|
Decmann A, Nyírö G, Darvasi O, Turai P, Bancos I, Kaur RJ, Pezzani R, Iacobone M, Kraljevic I, Kastelan D, Parasiliti-Caprino M, Maccario M, Nirschl N, Heinrich D, Reincke M, Patócs A, Igaz P. Circulating miRNA Expression Profiling in Primary Aldosteronism. Front Endocrinol (Lausanne) 2019; 10:739. [PMID: 31736877 PMCID: PMC6828819 DOI: 10.3389/fendo.2019.00739] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/24/2019] [Accepted: 10/14/2019] [Indexed: 12/19/2022] Open
Abstract
Objective: Primary aldosteronism is a major cause of secondary hypertension. Its two principal forms are bilateral adrenal hyperplasia (BAH) and aldosterone-producing adenoma (APA) whose differentiation is clinically pivotal. There is a major clinical need for a reliable and easily accessible diagnostic biomarker for case identification and subtyping. Circulating microRNAs were shown to be useful as minimally invasive diagnostic markers. Our aim was to determine and compare the circulating microRNA expression profiles of adenoma and hyperplasia plasma samples, and to evaluate their applicability as minimally invasive markers. Methods: One hundred and twenty-three samples from primary aldosteronism patients were included. Next-generation sequencing was performed on 30 EDTA-anticoagulated plasma samples (discovery cohort). Significantly differently expressed miRNAs were validated by real-time reverse transcription-qPCR in an independent validation cohort (93 samples). Results: We have found relative overexpression of miR-30e-5p, miR-30d-5p, miR-223-3p, and miR-7-5p in hyperplasia compared to adenoma by next-generation sequencing. Validation by qRT-PCR confirmed significant overexpression of hsa-miR-30e-5p, hsa-miR-30d-5p, and hsa-miR-7-5p in hyperplasia samples. Regarding the microRNA expressional variations, adenoma is more heterogeneous at the miRNA level compared to hyperplasia. Conclusion: Three microRNAs were significantly overexpressed in hyperplasia samples compared to adenoma samples, but their sensitivity and specificity values are not good enough for introduction to clinical practice.
Collapse
Affiliation(s)
- Abel Decmann
- 2nd Department of Internal Medicine, Faculty of Medicine, Semmelweis University, Budapest, Hungary
| | - Gábor Nyírö
- MTA-SE Molecular Medicine Research Group, Hungarian Academy of Sciences and Semmelweis University, Budapest, Hungary
| | - Ottó Darvasi
- Hereditary Endocrine Tumors Research Group, Hungarian Academy of Sciences and Semmelweis University, Budapest, Hungary
| | - Péter Turai
- 2nd Department of Internal Medicine, Faculty of Medicine, Semmelweis University, Budapest, Hungary
| | - Irina Bancos
- Division of Endocrinology, Diabetes, Metabolism and Nutrition, Department of Internal Medicine, Mayo Clinic, Rochester, MN, United States
| | - Ravinder Jeet Kaur
- Division of Endocrinology, Diabetes, Metabolism and Nutrition, Department of Internal Medicine, Mayo Clinic, Rochester, MN, United States
| | - Raffaele Pezzani
- Endocrinology Unit, Department of Medicine, University of Padua, Padova, Italy
| | - Maurizio Iacobone
- Minimally Invasive Endocrine Surgery Unit, Department of Surgery, Oncology and Gastroenterology (DISCOG), University of Padova, Padova, Italy
| | - Ivana Kraljevic
- Department of Endocrinology, University Hospital Centre Zagreb, Zagreb, Croatia
| | - Darko Kastelan
- Department of Endocrinology, University Hospital Centre Zagreb, Zagreb, Croatia
| | - Mirko Parasiliti-Caprino
- Division of Endocrinology, Diabetology and Metabolism, Department of Medical Sciences, University of Turin, Turin, Italy
| | - Mauro Maccario
- Division of Endocrinology, Diabetology and Metabolism, Department of Medical Sciences, University of Turin, Turin, Italy
| | - Nina Nirschl
- Medizinische Klinik und Poliklinik IV, Ludwig Maximilian University Munich, Munich, Germany
| | - Daniel Heinrich
- Medizinische Klinik und Poliklinik IV, Ludwig Maximilian University Munich, Munich, Germany
| | - Martin Reincke
- Medizinische Klinik und Poliklinik IV, Ludwig Maximilian University Munich, Munich, Germany
| | - Attila Patócs
- Hereditary Endocrine Tumors Research Group, Hungarian Academy of Sciences and Semmelweis University, Budapest, Hungary
| | - Peter Igaz
- 2nd Department of Internal Medicine, Faculty of Medicine, Semmelweis University, Budapest, Hungary
- MTA-SE Molecular Medicine Research Group, Hungarian Academy of Sciences and Semmelweis University, Budapest, Hungary
- *Correspondence: Peter Igaz
| |
Collapse
|
119
|
Affiliation(s)
- Maria-Christina Zennaro
- From the INSERM, UMRS_970, Paris Cardiovascular Research Center, Paris, France; Université Paris Descartes, Sorbonne Paris Cité, Paris, France; and Assistance Publique-Hôpitaux de Paris, Hôpital Européen Georges Pompidou, Service de Génétique, Paris, France
| |
Collapse
|
120
|
Vaidya A, Mulatero P, Baudrand R, Adler GK. The Expanding Spectrum of Primary Aldosteronism: Implications for Diagnosis, Pathogenesis, and Treatment. Endocr Rev 2018; 39:1057-1088. [PMID: 30124805 PMCID: PMC6260247 DOI: 10.1210/er.2018-00139] [Citation(s) in RCA: 185] [Impact Index Per Article: 26.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/13/2018] [Accepted: 08/10/2018] [Indexed: 12/14/2022]
Abstract
Primary aldosteronism is characterized by aldosterone secretion that is independent of renin and angiotensin II and sodium status. The deleterious effects of primary aldosteronism are mediated by excessive activation of the mineralocorticoid receptor that results in the well-known consequences of volume expansion, hypertension, hypokalemia, and metabolic alkalosis, but it also increases the risk for cardiovascular and kidney disease, as well as death. For decades, the approaches to defining, diagnosing, and treating primary aldosteronism have been relatively constant and generally focused on detecting and treating the more severe presentations of the disease. However, emerging evidence suggests that the prevalence of primary aldosteronism is much greater than previously recognized, and that milder and nonclassical forms of renin-independent aldosterone secretion that impart heightened cardiovascular risk may be common. Public health efforts to prevent aldosterone-mediated end-organ disease will require improved capabilities to diagnose all forms of primary aldosteronism while optimizing the treatment approaches such that the excess risk for cardiovascular and kidney disease is adequately mitigated. In this review, we present a physiologic approach to considering the diagnosis, pathogenesis, and treatment of primary aldosteronism. We review evidence suggesting that primary aldosteronism manifests across a wide spectrum of severity, ranging from mild to overt, that correlates with cardiovascular risk. Furthermore, we review emerging evidence from genetic studies that begin to provide a theoretical explanation for the pathogenesis of primary aldosteronism and a link to its phenotypic severity spectrum and prevalence. Finally, we review human studies that provide insights into the optimal approach toward the treatment of primary aldosteronism.
Collapse
Affiliation(s)
- Anand Vaidya
- Center for Adrenal Disorders, Division of Endocrinology, Diabetes, and Hypertension, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts
| | - Paolo Mulatero
- Division of Internal Medicine and Hypertension, Department of Medical Sciences, University of Torino, Torino, Italy
| | - Rene Baudrand
- Program for Adrenal Disorders and Hypertension, Department of Endocrinology, Pontificia Universidad Catolica de Chile, Santiago, Chile
| | - Gail K Adler
- Center for Adrenal Disorders, Division of Endocrinology, Diabetes, and Hypertension, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts
| |
Collapse
|