1
|
Fan Z, Karakone M, Nagarajan S, Nagy N, Mildenberger W, Petrova E, Hinte LC, Bijnen M, Häne P, Nelius E, Chen J, Ferapontova I, von Meyenn F, Trepiccione F, Berber M, Ribas DP, Eichmann A, Zennaro MC, Takeda N, Fischer JW, Spyroglou A, Reincke M, Beuschlein F, Loffing J, Greter M, Stockmann C. Macrophages preserve endothelial cell specialization in the adrenal gland to modulate aldosterone secretion and blood pressure. Cell Rep 2024; 43:114395. [PMID: 38941187 DOI: 10.1016/j.celrep.2024.114395] [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: 02/09/2023] [Revised: 04/22/2024] [Accepted: 06/07/2024] [Indexed: 06/30/2024] Open
Abstract
Macrophages play crucial roles in organ-specific functions and homeostasis. In the adrenal gland, macrophages closely associate with sinusoidal capillaries in the aldosterone-producing zona glomerulosa. We demonstrate that macrophages preserve capillary specialization and modulate aldosterone secretion. Using macrophage-specific deletion of VEGF-A, single-cell transcriptomics, and functional phenotyping, we found that the loss of VEGF-A depletes PLVAP+ fenestrated endothelial cells in the zona glomerulosa, leading to increased basement membrane collagen IV deposition and subendothelial fibrosis. This results in increased aldosterone secretion, called "haptosecretagogue" signaling. Human aldosterone-producing adenomas also show capillary rarefaction and basement membrane thickening. Mice with myeloid cell-specific VEGF-A deletion exhibit elevated serum aldosterone, hypokalemia, and hypertension, mimicking primary aldosteronism. These findings underscore macrophage-to-endothelial cell signaling as essential for endothelial cell specialization, adrenal gland function, and blood pressure regulation, with broader implications for other endocrine organs.
Collapse
Affiliation(s)
- Zheng Fan
- University of Zurich, Institute of Anatomy, 8057 Zurich, Switzerland.
| | - Mara Karakone
- University of Zurich, Institute of Anatomy, 8057 Zurich, Switzerland
| | | | - Nadine Nagy
- Division of Infectious Diseases and Geographic Medicine, Department of Medicine, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Wiebke Mildenberger
- University of Zurich, Institute for Experimental Immunology, 8057 Zurich, Switzerland
| | - Ekaterina Petrova
- University of Zurich, Institute for Experimental Immunology, 8057 Zurich, Switzerland
| | - Laura Catharina Hinte
- Laboratory of Nutrition and Metabolic Epigenetics, Institute for Food, Nutrition and Health, Department of Health Sciences and Technology, ETH Zurich, Zurich, Switzerland
| | - Mitchell Bijnen
- University of Zurich, Institute for Experimental Immunology, 8057 Zurich, Switzerland
| | - Philipp Häne
- University of Zurich, Institute for Experimental Immunology, 8057 Zurich, Switzerland
| | - Eric Nelius
- University of Zurich, Institute of Anatomy, 8057 Zurich, Switzerland
| | - Jing Chen
- University of Zurich, Institute of Anatomy, 8057 Zurich, Switzerland
| | - Irina Ferapontova
- University of Zurich, Institute of Anatomy, 8057 Zurich, Switzerland
| | - Ferdinand von Meyenn
- Laboratory of Nutrition and Metabolic Epigenetics, Institute for Food, Nutrition and Health, Department of Health Sciences and Technology, ETH Zurich, Zurich, Switzerland
| | - Francesco Trepiccione
- Department of Translational Medical Sciences, University of Campania Luigi Vanvitelli, Naples, Italy
| | - Mesut Berber
- University of Zurich, Institute of Anatomy, 8057 Zurich, Switzerland
| | - David Penton Ribas
- Electrophysiology Facility (e-phac), Department of Molecular Life Sciences, University of Zurich (UZH), 8057 Zürich, Switzerland
| | - Anne Eichmann
- Cardiovascular Research Center and Department of Cellular and Molecular Physiology, Yale University School of Medicine, New Haven, CT, USA
| | | | - Norihiko Takeda
- Department of Cardiovascular Medicine, Graduate School of Medicine, The University of Tokyo, 7-3-1, Hongo, Bunkyo-ku, Tokyo, Japan
| | - Jens W Fischer
- Institute of Pharmacology and Clinical Pharmacology, Medical Faculty, Heinrich-Heine-University Düsseldorf, Düsseldorf, Germany
| | - Ariadni Spyroglou
- Klinik für Endokrinologie, Diabetologie, und Klinische Ernährung, UniversitätsSpital Zürich (USZ) and UZH, Raemistrasse 100, 8091 Zurich, Switzerland; Medizinische Klinik und Poliklinik IV, Ludwig-Maximilians-University (LMU), Munich, Germany
| | - Martin Reincke
- Klinik für Endokrinologie, Diabetologie, und Klinische Ernährung, UniversitätsSpital Zürich (USZ) and UZH, Raemistrasse 100, 8091 Zurich, Switzerland
| | - Felix Beuschlein
- Klinik für Endokrinologie, Diabetologie, und Klinische Ernährung, UniversitätsSpital Zürich (USZ) and UZH, Raemistrasse 100, 8091 Zurich, Switzerland; Medizinische Klinik und Poliklinik IV, Ludwig-Maximilians-University (LMU), Munich, Germany
| | - Johannes Loffing
- University of Zurich, Institute of Anatomy, 8057 Zurich, Switzerland
| | - Melanie Greter
- University of Zurich, Institute for Experimental Immunology, 8057 Zurich, Switzerland
| | - Christian Stockmann
- University of Zurich, Institute of Anatomy, 8057 Zurich, Switzerland; INSERM U970, Paris Cardiovascular Research Center, Paris, France.
| |
Collapse
|
2
|
Harashima S, Yamazaki Y, Motomura N, Ono Y, Omata K, Tezuka Y, Morimoto R, Nakamura Y, Satoh F, Suzuki H, Kwon GE, Choi MH, Sasano H. Phenotype-genotype correlation in aldosterone-producing adenomas characterized by intracellular cholesterol metabolism. J Steroid Biochem Mol Biol 2022; 221:106116. [PMID: 35533918 DOI: 10.1016/j.jsbmb.2022.106116] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/26/2021] [Revised: 04/12/2022] [Accepted: 04/18/2022] [Indexed: 11/16/2022]
Abstract
Aldosterone-producing adenoma (APA) is histologically composed of clear and compact tumor cells. KCNJ5- mutated APAs were reported to be associated with higher plasma aldosterone concentration and more abundant clear tumor cells containing lipid droplets than non-KCNJ5- mutated APAs. However, the association among cholesterol uptake and/or synthesis, cellular morphology and genotypes has remained unknown. Therefore, in order to explore these differences, 52 APA cases (KCNJ5 mt: n = 33, non-KCNJ5 mt: n = 19; ATP1A1: n = 3, ATP2B3: n = 3, CACNA1D: n = 5, CTNNB1: n = 1, tumors without any mutation above: n = 7), zona glomerulosa (ZG) tissue adjacent to APA and 10 non-pathological adrenal glands (NAs) were examined for quantitative histopathological analysis of tumor morphology and immunohistochemical analysis of cholesterol receptors (SR-B1, LDL-R), cholesterol metabolic enzymes (ACAT1, ACAT2, HSL, DHCR24, StAR), and the enzymes required for steroid synthesis (CYP11A1, CYP17A, 3βHSD, CYP11B1, CYP11B2). Gas chromatography-mass spectrometry (GC-MS) analysis was further performed to profile cholesterol precursors and metabolites in 21 APA cases (KCNJ5 mt: n = 16, non-KCNJ5 mt: n = 5) and 14 adrenal cortex of adjacent adrenal tissues. Results demonstrated that both SR-B1 and DHCR24 were significantly lower in the ZG than in fasciculata or reticularis of NAs but LDL-R was not significantly different among them in immunohistochemical analysis. SR-B1 and DHCR24 were both significantly higher in APAs than in ZG tissue adjacent to APA. In GC-MS analysis, most cholesterol precursors and metabolites, except for lanosterol, and their metabolic ratios (= concentration of cholesterol/ precursor) were higher in APAs than in the adjacent adrenal cortex tissue. LDL-R, ACAT1/2, HSL, DHCR24 were all significantly lower in clear than in compact tumor cells of APA. LDL-R was significantly lower and cholesterol/lanosterol ratio was significantly higher in KCNJ5- mutated than non-KCNJ5- mutated APAs. We demonstrated SR-B1 mediated selective uptake of cholesterol ester and de novo cholesterol synthesis were both enhanced in APAs. In addition, cholesterol uptake and metabolism were different between clear and compact tumor cells. KCNJ5- mutated APAs were predominantly composed of clear tumor cells containing abundant cholesteryl ester but less activated LDL-R mediated uptake and increased de novo synthesis. Those findings above indicated their more pronounced functional deviation from the normal ZG cells in terms of their steroidogenic and intracellular cholesterol metabolism.
Collapse
Affiliation(s)
- Shogo Harashima
- Department of Pathology, National Hospital Organization Sendai medical center, Sendai, Japan; Department of Pathology, Tohoku Graduate School of Medicine, Tohoku University, Sendai, Japan
| | - Yuto Yamazaki
- Department of Pathology, Tohoku Graduate School of Medicine, Tohoku University, Sendai, Japan.
| | - Naoki Motomura
- Department of Pathology, Tohoku Graduate School of Medicine, Tohoku University, Sendai, Japan
| | - Yoshikiyo Ono
- Division of Clinical Hypertension, Endocrinology and Metabolism, Tohoku Graduate School of Medicine, Tohoku University, Sendai, Japan; Division of Nephrology, Endocrinology, and Vascular Medicine, Tohoku University Hospital, Sendai, Japan
| | - Kei Omata
- Division of Clinical Hypertension, Endocrinology and Metabolism, Tohoku Graduate School of Medicine, Tohoku University, Sendai, Japan; Division of Nephrology, Endocrinology, and Vascular Medicine, Tohoku University Hospital, Sendai, Japan
| | - Yuta Tezuka
- Division of Clinical Hypertension, Endocrinology and Metabolism, Tohoku Graduate School of Medicine, Tohoku University, Sendai, Japan; Division of Nephrology, Endocrinology, and Vascular Medicine, Tohoku University Hospital, Sendai, Japan
| | - Ryo Morimoto
- Division of Nephrology, Endocrinology, and Vascular Medicine, Tohoku University Hospital, Sendai, Japan
| | - Yasuhiro Nakamura
- Division of Pathology, Faculty of medicine, Tohoku Medical and Pharmaceutical University, Sendai, Japan
| | - Fumitoshi Satoh
- Division of Clinical Hypertension, Endocrinology and Metabolism, Tohoku Graduate School of Medicine, Tohoku University, Sendai, Japan; Division of Nephrology, Endocrinology, and Vascular Medicine, Tohoku University Hospital, Sendai, Japan
| | - Hiroyoshi Suzuki
- Department of Pathology, National Hospital Organization Sendai medical center, Sendai, Japan
| | - Go Eun Kwon
- Molecular Recognition Research Center, Korea Institute of Science and Technology, Seoul, Korea
| | - Man Ho Choi
- Molecular Recognition Research Center, Korea Institute of Science and Technology, Seoul, Korea
| | - Hironobu Sasano
- Department of Pathology, Tohoku Graduate School of Medicine, Tohoku University, Sendai, Japan
| |
Collapse
|
3
|
Progress on Genetic Basis of Primary Aldosteronism. Biomedicines 2021; 9:biomedicines9111708. [PMID: 34829937 PMCID: PMC8615950 DOI: 10.3390/biomedicines9111708] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2021] [Revised: 11/12/2021] [Accepted: 11/13/2021] [Indexed: 11/23/2022] Open
Abstract
Primary aldosteronism (PA) is a heterogeneous group of disorders caused by the autonomous overproduction of aldosterone with simultaneous suppression of plasma renin activity (PRA). It is considered to be the most common endocrine cause of secondary arterial hypertension (HT) and is associated with a high rate of cardiovascular complications. PA is most often caused by a bilateral adrenal hyperplasia (BAH) or aldosterone-producing adenoma (APA); rarer causes of PA include genetic disorders of steroidogenesis (familial hyperaldosteronism (FA) type I, II, III and IV), aldosterone-producing adrenocortical carcinoma, and ectopic aldosterone-producing tumors. Over the last few years, significant progress has been made towards understanding the genetic basis of PA, classifying it as a channelopathy. Recently, a growing body of clinical evidence suggests that mutations in ion channels appear to be the major cause of aldosterone-producing adenomas, and several mutations within the ion channel encoding genes have been identified. Somatic mutations in four genes (KCNJ5, ATP1A1, ATP2B3 and CACNA1D) have been identified in nearly 60% of the sporadic APAs, while germline mutations in KCNJ5 and CACNA1H have been reported in different subtypes of familial hyperaldosteronism. These new insights into the molecular mechanisms underlying PA may be associated with potential implications for diagnosis and therapy.
Collapse
|
4
|
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: 2.0] [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
|
5
|
Primary Aldosteronism: Metabolic Reprogramming and the Pathogenesis of Aldosterone-Producing Adenomas. Cancers (Basel) 2021; 13:cancers13153716. [PMID: 34359615 PMCID: PMC8345059 DOI: 10.3390/cancers13153716] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2021] [Revised: 06/29/2021] [Accepted: 07/21/2021] [Indexed: 12/19/2022] Open
Abstract
Simple Summary Primary aldosteronism is a common form of endocrine hypertension often caused by a hyper-secreting tumor of the adrenal cortex called an aldosterone-producing adenoma. Metabolic reprogramming plays a role in tumor progression and influences the tumor immune microenvironment by limiting immune-cell infiltration and suppressing its anti-tumor function. We hypothesized that the development of aldosterone-producing adenomas involves metabolic adaptations of its component tumor cells and intrinsically influences tumor pathogenesis. Herein, we use state-of-the-art computational tools for the comprehensive analysis of array-based gene expression profiles to demonstrate metabolic reprogramming and remodeling of the immune microenvironment in aldosterone-producing adenomas compared with paired adjacent adrenal cortical tissue. Our findings suggest metabolic alterations may function in the pathogenesis of aldosterone-producing adenomas by conferring survival advantages to their component tumor cells. Abstract Aldosterone-producing adenomas (APAs) are characterized by aldosterone hypersecretion and deregulated adrenocortical cell growth. Increased energy consumption required to maintain cellular tumorigenic properties triggers metabolic alterations that shape the tumor microenvironment to acquire necessary nutrients, yet our knowledge of this adaptation in APAs is limited. Here, we investigated adrenocortical cell-intrinsic metabolism and the tumor immune microenvironment of APAs and their potential roles in mediating aldosterone production and growth of adrenocortical cells. Using multiple advanced bioinformatics methods, we analyzed gene expression datasets to generate distinct metabolic and immune cell profiles of APAs versus paired adjacent cortex. APAs displayed activation of lipid metabolism, especially fatty acid β-oxidation regulated by PPARα, and glycolysis. We identified an immunosuppressive microenvironment in APAs, with reduced infiltration of CD45+ immune cells compared with adjacent cortex, validated by CD45 immunohistochemistry (3.45-fold, p < 0.001). APAs also displayed an association of lipid metabolism with ferroptosis and upregulation of antioxidant systems. In conclusion, APAs exhibit metabolic reprogramming towards fatty acid β-oxidation and glycolysis. Increased lipid metabolism via PPARα may serve as a key mechanism to modulate lipid peroxidation, a hallmark of regulated cell death by ferroptosis. These findings highlight survival advantages for APA tumor cells with metabolic reprogramming properties.
Collapse
|
6
|
Bennet D, Khorsandian Y, Pelusi J, Mirabella A, Pirrotte P, Zenhausern F. Molecular and physical technologies for monitoring fluid and electrolyte imbalance: A focus on cancer population. Clin Transl Med 2021; 11:e461. [PMID: 34185420 PMCID: PMC8214861 DOI: 10.1002/ctm2.461] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2021] [Revised: 05/11/2021] [Accepted: 05/29/2021] [Indexed: 12/23/2022] Open
Abstract
Several clinical examinations have shown the essential impact of monitoring (de)hydration (fluid and electrolyte imbalance) in cancer patients. There are multiple risk factors associated with (de)hydration, including aging, excessive or lack of fluid consumption in sports, alcohol consumption, hot weather, diabetes insipidus, vomiting, diarrhea, cancer, radiation, chemotherapy, and use of diuretics. Fluid and electrolyte imbalance mainly involves alterations in the levels of sodium, potassium, calcium, and magnesium in extracellular fluids. Hyponatremia is a common condition among individuals with cancer (62% of cases), along with hypokalemia (40%), hypophosphatemia (32%), hypomagnesemia (17%), hypocalcemia (12%), and hypernatremia (1-5%). Lack of hydration and monitoring of hydration status can lead to severe complications, such as nausea/vomiting, diarrhea, fatigue, seizures, cell swelling or shrinking, kidney failure, shock, coma, and even death. This article aims to review the current (de)hydration (fluid and electrolyte imbalance) monitoring technologies focusing on cancer. First, we discuss the physiological and pathophysiological implications of fluid and electrolyte imbalance in cancer patients. Second, we explore the different molecular and physical monitoring methods used to measure fluid and electrolyte imbalance and the measurement challenges in diverse populations. Hydration status is assessed in various indices; plasma, sweat, tear, saliva, urine, body mass, interstitial fluid, and skin-integration techniques have been extensively investigated. No unified (de)hydration (fluid and electrolyte imbalance) monitoring technology exists for different populations (including sports, elderly, children, and cancer). Establishing novel methods and technologies to facilitate and unify measurements of hydration status represents an excellent opportunity to develop impactful new approaches for patient care.
Collapse
Affiliation(s)
- Devasier Bennet
- Center for Applied NanoBioscience and MedicineThe University of ArizonaCollege of MedicinePhoenixUSA
| | - Yasaman Khorsandian
- Center for Applied NanoBioscience and MedicineThe University of ArizonaCollege of MedicinePhoenixUSA
| | | | | | - Patrick Pirrotte
- Collaborative Center for Translational Mass SpectrometryTranslational Genomics Research InstitutePhoenixUSA
| | - Frederic Zenhausern
- Center for Applied NanoBioscience and MedicineThe University of ArizonaCollege of MedicinePhoenixUSA
- HonorHealth Research InstituteScottsdaleUSA
- Collaborative Center for Translational Mass SpectrometryTranslational Genomics Research InstitutePhoenixUSA
| |
Collapse
|
7
|
Hypertension Editors' Picks: Hyperaldosteronism. Hypertension 2021; 77:e17-e28. [PMID: 33439730 DOI: 10.1161/hypertensionaha.120.15141] [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]
|
8
|
Knott EL, Leidenheimer NJ. A Targeted Bioinformatics Assessment of Adrenocortical Carcinoma Reveals Prognostic Implications of GABA System Gene Expression. Int J Mol Sci 2020; 21:ijms21228485. [PMID: 33187258 PMCID: PMC7697095 DOI: 10.3390/ijms21228485] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2020] [Revised: 11/05/2020] [Accepted: 11/09/2020] [Indexed: 12/12/2022] Open
Abstract
Adrenocortical carcinoma (ACC) is a rare but deadly cancer for which few treatments exist. Here, we have undertaken a targeted bioinformatics study of The Cancer Genome Atlas (TCGA) ACC dataset focusing on the 30 genes encoding the γ-aminobutyric acid (GABA) system—an under-studied, evolutionarily-conserved system that is an emerging potential player in cancer progression. Our analysis identified a subset of ACC patients whose tumors expressed a distinct GABA system transcriptome. Transcript levels of ABAT (encoding a key GABA shunt enzyme), were upregulated in over 40% of tumors, and this correlated with several favorable clinical outcomes including patient survival; while enrichment and ontology analysis implicated two cancer-related biological pathways involved in metastasis and immune response. The phenotype associated with ABAT upregulation revealed a potential metabolic heterogeneity among ACC tumors associated with enhanced mitochondrial metabolism. Furthermore, many GABAA receptor subunit-encoding transcripts were expressed, including two (GABRB2 and GABRD) prognostic for patient survival. Transcripts encoding GABAB receptor subunits and GABA transporters were also ubiquitously expressed. The GABA system transcriptome of ACC tumors is largely mirrored in the ACC NCI-H295R cell line, suggesting that this cell line may be appropriate for future functional studies investigating the role of the GABA system in ACC cell growth phenotypes and metabolism.
Collapse
|
9
|
Abstract
Primary aldosteronism is the most common form of secondary hypertension with a prevalence of 5-10% in hypertensive patients. Aldosterone-producing adenoma (APA) is a subtype of primary aldosteronism, and somatic mutations in KCNJ5, ATP1A1, ATP2B3, CACNA1D, CLCN2, or CTNNB1 were identified and recognized to drive aldosterone production and/or contribute to tumorigenesis in APA. Mutations of KCNJ5, ATP1A1, ATP2B3, CACNA1D, and CLCN2 are known to activate calcium signaling, and its activation potentiate CYP11B2 (aldosterone synthesis) transcription in adrenal cells. Transcriptome analyses combined with bioinformatics using APA samples were conductive for each gene mutation mediated pivotal pathway, gene ontology, and clustering. Several important intracellular molecules in increase aldosterone production were detected by transcriptome analysis, and additional functional analyses demonstrated intracellular molecular mechanisms of aldosterone production which focused on calcium signal, CYP11B2 transcription and translation. Furthermore, DNA methylation analysis revealed that promoter region of CYP11B2 was entirely hypomethylated, but that of other steroidogenic enzymes were not in APA. Integration of transcriptome and DNA methylome analysis clarified some DNA methylation associated gene expression, and the transcripts have a role for aldosterone production. In this article, we reviewed the intracellular molecular mechanisms of aldosterone production in APA, and discussed future challenges for basic studies leading to clinical practice.
Collapse
Affiliation(s)
- Kenji Oki
- Department of Molecular and Internal Medicine, Graduate School of Biomedical and Health Sciences, Hiroshima University, Hiroshima, Japan
| | - Celso E. Gomez-Sanchez
- Division of Endocrinology, G.V. (Sonny) Montgomery VA Medical Center and Department of Pharmacology and Toxicology, University of Mississippi Medical Center, Jackson, MS, USA
| |
Collapse
|
10
|
Vohra T, Kemter E, Sun N, Dobenecker B, Hinrichs A, Burrello J, Gomez-Sanchez EP, Gomez-Sanchez CE, Wang J, Kinker IS, Teupser D, Fischer K, Schnieke A, Peitzsch M, Eisenhofer G, Walch A, Reincke M, Wolf E, Williams TA. Effect of Dietary Sodium Modulation on Pig Adrenal Steroidogenesis and Transcriptome Profiles. Hypertension 2020; 76:1769-1777. [PMID: 33070662 DOI: 10.1161/hypertensionaha.120.15998] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Primary aldosteronism is a frequent form of endocrine hypertension caused by aldosterone overproduction from the adrenal cortex. Regulation of aldosterone biosynthesis has been studied in rodents despite differences in adrenal physiology with humans. We, therefore, investigated pig adrenal steroidogenesis, morphology, and transcriptome profiles of the zona glomerulosa (zG) and zona fasciculata in response to activation of the renin-angiotensin-aldosterone system by dietary sodium restriction. Six-week-old pigs were fed a low- or high-sodium diet for 14 days (3 pigs per group, 0.4 g sodium/kg feed versus 6.8 g sodium/kg). Plasma aldosterone concentrations displayed a 43-fold increase (P=0.011) after 14 days of sodium restriction (day 14 versus day 0). Low dietary sodium caused a 2-fold increase in thickness of the zG (P<0.001) and an almost 3-fold upregulation of CYP11B (P<0.05) compared with high dietary sodium. Strong immunostaining of the KCNJ5 (G protein-activated inward rectifier potassium channel 4), which is frequently mutated in primary aldosteronism, was demonstrated in the zG. mRNA sequencing transcriptome analysis identified significantly altered expression of genes modulated by the renin-angiotensin-aldosterone system in the zG (n=1172) and zona fasciculata (n=280). These genes included many with a known role in the regulation of aldosterone synthesis and adrenal function. The most highly enriched biological pathways in the zG were related to cholesterol biosynthesis, steroid metabolism, cell cycle, and potassium channels. This study provides mechanistic insights into the physiology and pathophysiology of aldosterone production in a species closely related to humans and shows the suitability of pigs as a translational animal model for human adrenal steroidogenesis.
Collapse
Affiliation(s)
- Twinkle Vohra
- From the Medizinische Klinik und Poliklinik IV, Klinikum der Universität München (T.V., I.-S.K., M.R., T.A.W.), Ludwig-Maximilians-Universität München, Munich, Germany
| | - Elisabeth Kemter
- Chair for Molecular Animal Breeding and Biotechnology, Gene Center and Department of Veterinary Sciences (E.K., A.H., E.W.), Ludwig-Maximilians-Universität München, Munich, Germany
| | - Na Sun
- Research Unit Analytical Pathology, German Research Center for Environmental Health, Helmholtz Zentrum München, Neuherberg, Germany (N.S., J.W., A.W.)
| | - Britta Dobenecker
- Chair of Animal Nutrition and Dietetics, Department of Veterinary Sciences, Ludwig-Maximilians-Universität München, Oberschleißheim, Germany (B.D.)
| | - Arne Hinrichs
- Chair for Molecular Animal Breeding and Biotechnology, Gene Center and Department of Veterinary Sciences (E.K., A.H., E.W.), Ludwig-Maximilians-Universität München, Munich, Germany
| | - Jacopo Burrello
- Division of Internal Medicine and Hypertension, Department of Medical Sciences, University of Turin, Italy (J.B., T.A.W.)
| | - Elise P Gomez-Sanchez
- Department of Neurobiology and Anatomical Sciences, University of Mississippi Medical Center, Jackson (E.P.G.-S.)
| | - Celso E Gomez-Sanchez
- Endocrine Division, G.V. (Sonny) Montgomery VA Medical Center, Jackson, MS (C.E.G.-S.).,Department of Pharmacology and Toxicology and Medicine, University of Mississippi Medical Center, Jackson (C.E.G.-S.)
| | - Jun Wang
- Research Unit Analytical Pathology, German Research Center for Environmental Health, Helmholtz Zentrum München, Neuherberg, Germany (N.S., J.W., A.W.)
| | - Isabella-Sabrina Kinker
- From the Medizinische Klinik und Poliklinik IV, Klinikum der Universität München (T.V., I.-S.K., M.R., T.A.W.), Ludwig-Maximilians-Universität München, Munich, Germany
| | - Daniel Teupser
- Institute of Laboratory Medicine, University Hospital (D.T.), Ludwig-Maximilians-Universität München, Munich, Germany
| | - Konrad Fischer
- School of Life Sciences Weihenstephan, Technical University Munich, Freising, Germany (K.F., A.S.)
| | - Angelika Schnieke
- School of Life Sciences Weihenstephan, Technical University Munich, Freising, Germany (K.F., A.S.)
| | - Mirko Peitzsch
- Institute of Clinical Chemistry and Laboratory Medicine (M.P., G.E.), University Hospital Carl Gustav Carus, Technische Universität Dresden, Germany
| | - Graeme Eisenhofer
- Institute of Clinical Chemistry and Laboratory Medicine (M.P., G.E.), University Hospital Carl Gustav Carus, Technische Universität Dresden, Germany.,Department of Medicine III (G.E.), University Hospital Carl Gustav Carus, Technische Universität Dresden, Germany
| | - Axel Walch
- Research Unit Analytical Pathology, German Research Center for Environmental Health, Helmholtz Zentrum München, Neuherberg, Germany (N.S., J.W., A.W.)
| | - Martin Reincke
- From the Medizinische Klinik und Poliklinik IV, Klinikum der Universität München (T.V., I.-S.K., M.R., T.A.W.), Ludwig-Maximilians-Universität München, Munich, Germany
| | - Eckhard Wolf
- Chair for Molecular Animal Breeding and Biotechnology, Gene Center and Department of Veterinary Sciences (E.K., A.H., E.W.), Ludwig-Maximilians-Universität München, Munich, Germany
| | - Tracy Ann Williams
- From the Medizinische Klinik und Poliklinik IV, Klinikum der Universität München (T.V., I.-S.K., M.R., T.A.W.), Ludwig-Maximilians-Universität München, Munich, Germany.,Division of Internal Medicine and Hypertension, Department of Medical Sciences, University of Turin, Italy (J.B., T.A.W.)
| |
Collapse
|
11
|
Vaduva P, Bonnet F, Bertherat J. Molecular Basis of Primary Aldosteronism and Adrenal Cushing Syndrome. J Endocr Soc 2020; 4:bvaa075. [PMID: 32783015 PMCID: PMC7412855 DOI: 10.1210/jendso/bvaa075] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/01/2020] [Accepted: 06/23/2020] [Indexed: 02/07/2023] Open
Abstract
This review reports the main molecular alterations leading to development of benign cortisol- and/or aldosterone-secreting adrenal tumors. Causes of adrenal Cushing syndrome can be divided in 2 groups: multiple bilateral tumors or adenomas secreting cortisol. Bilateral causes are mainly primary pigmented nodular adrenocortical disease, most of the time due to PRKAR1A germline-inactivating mutations, and primary bilateral macronodular adrenal hyperplasia that can be caused in some rare syndromic cases by germline-inactivating mutations of MEN1, APC, and FH and of ARMC5 in isolated forms. PRKACA somatic-activating mutations are the main alterations in unilateral cortisol-producing adenomas. In primary hyperaldosteronism (PA), familial forms were identified in 1% to 5% of cases: familial hyperaldosteronism type I (FH-I) due to a chimeric CYP11B1/CYP11B2 hybrid gene, FH-II due to CLCN-2 germline mutations, FH-III due to KCNJ5 germline mutations, FH-IV due to CACNA1H germline mutations and PA, and seizures and neurological abnormalities syndrome due to CACNA1D germline mutations. Several somatic mutations have been found in aldosterone-producing adenomas in KCNJ5, ATP1A1, ATP2B3, CACNA1D, and CTNNB1 genes. In addition to these genetic alterations, genome-wide approaches identified several new alterations in transcriptome, methylome, and miRnome studies, highlighting new pathways involved in steroid dysregulation.
Collapse
Affiliation(s)
- Patricia Vaduva
- Reference Center for Rare Adrenal Diseases, Department of Endocrinology, Assistance Publique Hôpitaux de Paris, Hôpital Cochin, Paris, France.,Institut Cochin, INSERM U1016, CNRS UMR8104, Paris University, Paris, France
| | - Fideline Bonnet
- Institut Cochin, INSERM U1016, CNRS UMR8104, Paris University, Paris, France.,Hormonal Biology Laboratory, Assistance Publique Hôpitaux de Paris, Hôpital Cochin, Paris, France
| | - Jérôme Bertherat
- Reference Center for Rare Adrenal Diseases, Department of Endocrinology, Assistance Publique Hôpitaux de Paris, Hôpital Cochin, Paris, France.,Institut Cochin, INSERM U1016, CNRS UMR8104, Paris University, Paris, France
| |
Collapse
|
12
|
Sun N, Meyer LS, Feuchtinger A, Kunzke T, Knösel T, Reincke M, Walch A, Williams TA. Mass Spectrometry Imaging Establishes 2 Distinct Metabolic Phenotypes of Aldosterone-Producing Cell Clusters in Primary Aldosteronism. Hypertension 2020; 75:634-644. [PMID: 31957522 DOI: 10.1161/hypertensionaha.119.14041] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Aldosterone-producing adenomas (APAs) are one of the main causes of primary aldosteronism and the most prevalent surgically correctable form of hypertension. Aldosterone-producing cell clusters (APCCs) comprise tight nests of zona glomerulosa cells, strongly positive for CYP11B2 (aldosterone synthase) in immunohistochemistry. APCCs have been suggested as possible precursors of APAs because they frequently carry driver mutations for constitutive aldosterone production, and a few adrenal lesions with histopathologic features of both APCCs and APAs have been identified. Our objective was to investigate the metabolic phenotypes of APCCs (n=27) compared with APAs (n=6) using in situ matrix-assisted laser desorption/ionization mass spectrometry imaging of formalin-fixed paraffin-embedded adrenals from patients with unilateral primary aldosteronism. Specific distribution patterns of metabolites were associated with APCCs and classified 2 separate APCC subgroups (subgroups 1 and 2) indistinguishable by CYP11B2 immunohistochemistry. Metabolic profiles of APCCs in subgroup 1 were tightly clustered and distinct from subgroup 2 and APAs. Multiple APCCs from the same adrenal displayed metabolic profiles of the same subgroup. Metabolites of APCC subgroup 2 were highly similar to the APA group and indicated enhanced metabolic pathways favoring cell proliferation compared with APCC subgroup 1. In conclusion, we demonstrate specific subgroups of APCCs with strikingly divergent distribution patterns of metabolites. One subgroup displays a metabolic phenotype convergent with APAs and may represent the progression of APCCs to APAs.
Collapse
Affiliation(s)
- Na Sun
- From the Research Unit Analytical Pathology, German Research Center for Environmental Health, Helmholtz Zentrum München (N.S., A.F., T. Kunzke, A.W.)
| | - Lucie S Meyer
- Medizinische Klinik und Poliklinik IV, Klinikum der Universität München, LMU München, Germany (L.S.M., M.R., T.A.W.)
| | - Annette Feuchtinger
- From the Research Unit Analytical Pathology, German Research Center for Environmental Health, Helmholtz Zentrum München (N.S., A.F., T. Kunzke, A.W.)
| | - Thomas Kunzke
- From the Research Unit Analytical Pathology, German Research Center for Environmental Health, Helmholtz Zentrum München (N.S., A.F., T. Kunzke, A.W.)
| | - Thomas Knösel
- Institute of Pathology, Ludwig-Maximilians-Universität München, Germany (T. Knösel)
| | - Martin Reincke
- Medizinische Klinik und Poliklinik IV, Klinikum der Universität München, LMU München, Germany (L.S.M., M.R., T.A.W.)
| | - Axel Walch
- From the Research Unit Analytical Pathology, German Research Center for Environmental Health, Helmholtz Zentrum München (N.S., A.F., T. Kunzke, A.W.)
| | - Tracy Ann Williams
- Medizinische Klinik und Poliklinik IV, Klinikum der Universität München, LMU München, Germany (L.S.M., M.R., T.A.W.).,Division of Internal Medicine and Hypertension, Department of Medical Sciences, University of Turin, Italy (T.A.W.)
| |
Collapse
|
13
|
Tevosian SG, Fox SC, Ghayee HK. Molecular Mechanisms of Primary Aldosteronism. Endocrinol Metab (Seoul) 2019; 34:355-366. [PMID: 31884735 PMCID: PMC6935778 DOI: 10.3803/enm.2019.34.4.355] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/29/2019] [Revised: 12/02/2019] [Accepted: 12/09/2019] [Indexed: 01/11/2023] Open
Abstract
Primary aldosteronism (PA) results from excess production of mineralocorticoid hormone aldosterone by the adrenal cortex. It is normally caused either by unilateral aldosterone-producing adenoma (APA) or by bilateral aldosterone excess as a result of bilateral adrenal hyperplasia. PA is the most common cause of secondary hypertension and associated morbidity and mortality. While most cases of PA are sporadic, an important insight into this debilitating disease has been derived through investigating the familial forms of the disease that affect only a minor fraction of PA patients. The advent of gene expression profiling has shed light on the genes and intracellular signaling pathways that may play a role in the pathogenesis of these tumors. The genetic basis for several forms of familial PA has been uncovered in recent years although the list is likely to expand. Recently, the work from several laboratories provided evidence for the involvement of mammalian target of rapamycin pathway and inflammatory cytokines in APAs; however, their mechanism of action in tumor development and pathophysiology remains to be understood.
Collapse
Affiliation(s)
- Sergei G Tevosian
- Department of Physiological Sciences, College of Veterinary Medicine, University of Florida, Gainesville, FL, USA
| | - Shawna C Fox
- Department of Pharmacotherapy and Translational Research, College of Pharmacy, University of Florida, Gainesville, FL, USA
| | - Hans K Ghayee
- Division of Endocrinology, Department of Medicine, Malcom Randall VA Medical Center, University of Florida, Gainesville, FL, USA.
| |
Collapse
|
14
|
Trinh B, Hepprich M, Betz MJ, Burkard T, Cavelti-Weder C, Seelig E, Meienberg F, Kratschmar DV, Beuschlein F, Reincke M, Odermatt A, Hall MN, Donath MY, Swierczynska MM. Treatment of Primary Aldosteronism With mTORC1 Inhibitors. J Clin Endocrinol Metab 2019; 104:4703-4714. [PMID: 31087053 DOI: 10.1210/jc.2019-00563] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/08/2019] [Accepted: 05/08/2019] [Indexed: 12/17/2022]
Abstract
CONTEXT Mammalian target of rapamycin complex 1 (mTORC1) activity is often increased in the adrenal cortex of patients with primary aldosteronism (PA), and mTORC1 inhibition decreases aldosterone production in adrenocortical cells, suggesting the mTORC1 pathway as a target for treatment of PA. OBJECTIVE To investigate the effect of mTORC1 inhibition on adrenal steroid hormones and hemodynamic parameters in mice and in patients with PA. DESIGN (i) Plasma aldosterone, corticosterone, and angiotensin II (Ang II) were measured in mice treated for 24 hours with vehicle or rapamycin. (ii) Plasma aldosterone levels after a saline infusion test, plasma renin, and 24-hour urine steroid hormone metabolome and hemodynamic parameters were measured during an open-label study in 12 patients with PA, before and after 2 weeks of treatment with everolimus and after a 2-week washout. MAIN OUTCOME MEASURES (i) Change in plasma aldosterone levels. (ii) Change in other steroid hormones, renin, Ang II, and hemodynamic parameters. RESULTS Treatment of mice with rapamycin significantly decreased plasma aldosterone levels (P = 0.007). Overall, treatment of PA patients with everolimus significantly decreased blood pressure (P < 0.05) and increased renin levels (P = 0.001) but did not decrease aldosterone levels significantly. However, prominent reduction of aldosterone levels upon everolimus treatment was observed in four patients. CONCLUSION In mice, mTORC1 inhibition was associated with reduced plasma aldosterone levels. In patients with PA, mTORC1 inhibition was associated with improved blood pressure and renin suppression. In addition, mTORC1 inhibition appeared to reduce plasma aldosterone in a subset of patients.
Collapse
Affiliation(s)
- Beckey Trinh
- Department of Endocrinology, Diabetes and Metabolism, University Hospital Basel, and Department of Biomedicine, University of Basel, Basel, Switzerland
| | - Matthias Hepprich
- Department of Endocrinology, Diabetes and Metabolism, University Hospital Basel, and Department of Biomedicine, University of Basel, Basel, Switzerland
| | - Matthias J Betz
- Department of Endocrinology, Diabetes and Metabolism, University Hospital Basel, and Department of Biomedicine, University of Basel, Basel, Switzerland
| | - Thilo Burkard
- European Society of Hypertension Centre of Excellence, Medical Outpatient Department, and Department of Cardiology, University Hospital Basel, Basel, Switzerland
| | - Claudia Cavelti-Weder
- Department of Endocrinology, Diabetes and Metabolism, University Hospital Basel, and Department of Biomedicine, University of Basel, Basel, Switzerland
| | - Eleonora Seelig
- Department of Endocrinology, Diabetes and Metabolism, University Hospital Basel, and Department of Biomedicine, University of Basel, Basel, Switzerland
| | - Fabian Meienberg
- Clinic of Endocrinology and Diabetology, Kantonsspital Baselland, Liestal, Switzerland
| | - Denise V Kratschmar
- Department of Pharmaceutical Sciences, University of Basel, Basel, Switzerland
| | - Felix Beuschlein
- Klinik für Endokrinologie, Diabetologie und Klinische Ernährung, Universitätsspital Zürich, Zürich, Switzerland
- Medizinische Klinik und Poliklinik IV, Klinikum der Universität München, Munich, Germany
| | - Martin Reincke
- Medizinische Klinik und Poliklinik IV, Klinikum der Universität München, Munich, Germany
| | - Alex Odermatt
- Department of Pharmaceutical Sciences, University of Basel, Basel, Switzerland
| | | | - Marc Y Donath
- Department of Endocrinology, Diabetes and Metabolism, University Hospital Basel, and Department of Biomedicine, University of Basel, Basel, Switzerland
| | | |
Collapse
|
15
|
Ma H, Li R, Di X, Jin X, Wang Y, Lai B, Shi C, Ji M, Zhu X, Wang K. ITRAQ-based proteomic analysis reveals possible target-related proteins in human adrenocortical adenomas. BMC Genomics 2019; 20:655. [PMID: 31419939 PMCID: PMC6697928 DOI: 10.1186/s12864-019-6030-5] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2018] [Accepted: 08/12/2019] [Indexed: 01/22/2023] Open
Abstract
Background Adrenocortical adenomas (ACAs) can lead to the autonomous secretion of aldosterone responsible for primary aldosteronism (PA), which is the most common form of secondary arterial hypertension. However, the authentic fundamental mechanisms underlying ACAs remain unclear. Objective Isobaric tags for relative and absolute quantitation (iTRAQ)-based proteomics and bioinformatics analyses from etiological studies of ACAs were performed to screen the differentially expressed proteins (DEPs) and investigate the relevant mechanisms of their occurrence and development. Results could help determine therapeutic targets of clinical significance. Methods In the present study, iTRAQ-based proteomics was applied to analyze ACA tissue samples from normal adrenal cortex tissues adjacent to the tumor. Using proteins extracted from a panel of four pairs of ACA samples, we identified some upregulated proteins and other downregulated proteins in all four pairs of ACA samples compared with adjacent normal tissue. Subsequently, we predicted protein–protein interaction networks of three DEPs to determine the authentic functional factors in ACA. Results A total of 753 DEPs were identified, including 347 upregulated and 406 downregulated proteins. The expression of three upregulated proteins (E2F3, KRT6A, and ALDH1A2) was validated by Western blot in 24 ACA samples. Our data suggested that some DEPs might be important hallmarks during the development of ACA. Conclusions This study is the first proteomic research to investigate alterations in protein levels and affected pathways in ACA using the iTRAQ technique. Thus, this study not only provides a comprehensive dataset on overall protein changes but also sheds light on its potential molecular mechanism in human ACAs. Electronic supplementary material The online version of this article (10.1186/s12864-019-6030-5) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- He Ma
- Department of Respiratory Medicine, the Second Hospital of Jilin University, Changchun, China.,Department of Anesthesiology, the Second Hospital of Jilin University, Changchun, China
| | - Ranwei Li
- Department of Urinary Surgery, the Second Hospital of Jilin University, Changchun, China
| | - Xin Di
- Department of Respiratory Medicine, the Second Hospital of Jilin University, Changchun, China
| | - Xin Jin
- Department of Hematology, the Second Hospital of Jilin University, Changchun, China
| | - Yan Wang
- Department of Respiratory Medicine, the Second Hospital of Jilin University, Changchun, China
| | - Bingjie Lai
- Department of Intensive Care Unit, the Second Hospital of Jilin University, Changchun, China
| | - Cailian Shi
- Department of Anesthesiology, the Second Hospital of Jilin University, Changchun, China
| | - Mingxin Ji
- Department of Anesthesiology, the Second Hospital of Jilin University, Changchun, China
| | - Xinran Zhu
- Department of Anesthesiology, the Second Hospital of Jilin University, Changchun, China
| | - Ke Wang
- Department of Respiratory Medicine, the Second Hospital of Jilin University, Changchun, China.
| |
Collapse
|
16
|
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
|