1
|
Hoshi-Numahata M, Nakanishi-Kimura A, Watanabe H, Nishiura M, Nishimoto S, Ueno F, Koguchi R, Oka A, Sato Y, Kajii TS, Iimura T. Genome-wide analyses of susceptibility genes responsible for mandibular prognathism in the Japanese population. J Oral Biosci 2025:100670. [PMID: 40349918 DOI: 10.1016/j.job.2025.100670] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2025] [Revised: 05/02/2025] [Accepted: 05/07/2025] [Indexed: 05/14/2025]
Abstract
BACKGROUND Mandibular prognathism (MP) is a type of malocclusion characterized by an imbalance in the anteroposterior position of the upper and lower jaws. The prevalence of MP in Japan is relatively high, suggesting a unique genetic background in the population. HIGHLIGHT Genome-wide analyses identified susceptibility genes responsible for mandibular prognathism in the Japanese population. CONCLUSION Identification of the genes associated with malocclusion will pave the way for personalized and precise medicine and contribute to craniofacial biology.
Collapse
Affiliation(s)
- Marie Hoshi-Numahata
- Department of Pharmacology, Faculty and Graduate School of Dental Medicine, Hokkaido University, Sapporo 060-8686, Japan; Department of Orthodontics, Faculty and Graduate School of Dental Medicine, Hokkaido University, Sapporo 060-8686, Japan
| | - Atsuko Nakanishi-Kimura
- Department of Pharmacology, Faculty and Graduate School of Dental Medicine, Hokkaido University, Sapporo 060-8686, Japan; Department of Orthodontics, Faculty and Graduate School of Dental Medicine, Hokkaido University, Sapporo 060-8686, Japan
| | - Haruhisa Watanabe
- Department of Pharmacology, Faculty and Graduate School of Dental Medicine, Hokkaido University, Sapporo 060-8686, Japan; Department of Oral Medicine and Diagnostics, Faculty and Graduate School of Dental Medicine, Hokkaido University, Sapporo 060-8686, Japan
| | - Mai Nishiura
- Department of Pharmacology, Faculty and Graduate School of Dental Medicine, Hokkaido University, Sapporo 060-8686, Japan; Department of Dentistry for Children and Disabled Persons, Faculty and Graduate School of Dental Medicine, Hokkaido University, Sapporo 060-8686, Japan
| | - Shinnosuke Nishimoto
- Department of Pharmacology, Faculty and Graduate School of Dental Medicine, Hokkaido University, Sapporo 060-8686, Japan; Department of Orthodontics, Faculty and Graduate School of Dental Medicine, Hokkaido University, Sapporo 060-8686, Japan
| | - Fumi Ueno
- Department of Pharmacology, Faculty and Graduate School of Dental Medicine, Hokkaido University, Sapporo 060-8686, Japan; Department of Orthodontics, Faculty and Graduate School of Dental Medicine, Hokkaido University, Sapporo 060-8686, Japan
| | - Riyu Koguchi
- Department of Pharmacology, Faculty and Graduate School of Dental Medicine, Hokkaido University, Sapporo 060-8686, Japan; Department of Oral Medicine and Diagnostics, Faculty and Graduate School of Dental Medicine, Hokkaido University, Sapporo 060-8686, Japan
| | - Akira Oka
- Institute of Medical Sciences, Tokai University, Isehara, Kanagawa 259-1193, Japan
| | - Yoshiaki Sato
- Department of Orthodontics, Faculty and Graduate School of Dental Medicine, Hokkaido University, Sapporo 060-8686, Japan
| | - Takashi S Kajii
- Department of Orthodontics, Keiyu-kai Sapporo Hospital, Sapporo 060-0061, Japan
| | - Tadahiro Iimura
- Department of Pharmacology, Faculty and Graduate School of Dental Medicine, Hokkaido University, Sapporo 060-8686, Japan.
| |
Collapse
|
2
|
Yang M, Shi Y, Song Q, Wei Z, Dun X, Wang Z, Wang Z, Qiu CW, Zhang H, Cheng X. Optical sorting: past, present and future. LIGHT, SCIENCE & APPLICATIONS 2025; 14:103. [PMID: 40011460 DOI: 10.1038/s41377-024-01734-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/03/2024] [Revised: 12/02/2024] [Accepted: 12/24/2024] [Indexed: 02/28/2025]
Abstract
Optical sorting combines optical tweezers with diverse techniques, including optical spectrum, artificial intelligence (AI) and immunoassay, to endow unprecedented capabilities in particle sorting. In comparison to other methods such as microfluidics, acoustics and electrophoresis, optical sorting offers appreciable advantages in nanoscale precision, high resolution, non-invasiveness, and is becoming increasingly indispensable in fields of biophysics, chemistry, and materials science. This review aims to offer a comprehensive overview of the history, development, and perspectives of various optical sorting techniques, categorised as passive and active sorting methods. To begin, we elucidate the fundamental physics and attributes of both conventional and exotic optical forces. We then explore sorting capabilities of active optical sorting, which fuses optical tweezers with a diversity of techniques, including Raman spectroscopy and machine learning. Afterwards, we reveal the essential roles played by deterministic light fields, configured with lens systems or metasurfaces, in the passive sorting of particles based on their varying sizes and shapes, sorting resolutions and speeds. We conclude with our vision of the most promising and futuristic directions, including AI-facilitated ultrafast and bio-morphology-selective sorting. It can be envisioned that optical sorting will inevitably become a revolutionary tool in scientific research and practical biomedical applications.
Collapse
Affiliation(s)
- Meng Yang
- Institute of Precision Optical Engineering, School of Physics Science and Engineering, Tongji University, Shanghai, 200092, China
- MOE Key Laboratory of Advanced Micro-Structured Materials, Shanghai, 200092, China
- Shanghai Institute of Intelligent Science and Technology, Tongji University, Shanghai, 200092, China
- Shanghai Frontiers Science Center of Digital Optics, Shanghai, 200092, China
| | - Yuzhi Shi
- Institute of Precision Optical Engineering, School of Physics Science and Engineering, Tongji University, Shanghai, 200092, China.
- MOE Key Laboratory of Advanced Micro-Structured Materials, Shanghai, 200092, China.
- Shanghai Institute of Intelligent Science and Technology, Tongji University, Shanghai, 200092, China.
- Shanghai Frontiers Science Center of Digital Optics, Shanghai, 200092, China.
| | - Qinghua Song
- Tsinghua Shenzhen International Graduate School, Tsinghua University, Shenzhen, 518055, China
| | - Zeyong Wei
- Institute of Precision Optical Engineering, School of Physics Science and Engineering, Tongji University, Shanghai, 200092, China
- MOE Key Laboratory of Advanced Micro-Structured Materials, Shanghai, 200092, China
- Shanghai Institute of Intelligent Science and Technology, Tongji University, Shanghai, 200092, China
- Shanghai Frontiers Science Center of Digital Optics, Shanghai, 200092, China
| | - Xiong Dun
- Institute of Precision Optical Engineering, School of Physics Science and Engineering, Tongji University, Shanghai, 200092, China
- MOE Key Laboratory of Advanced Micro-Structured Materials, Shanghai, 200092, China
- Shanghai Institute of Intelligent Science and Technology, Tongji University, Shanghai, 200092, China
- Shanghai Frontiers Science Center of Digital Optics, Shanghai, 200092, China
| | - Zhiming Wang
- Institute of Fundamental and Frontier Sciences, University of Electronic Science and Technology of China, Chengdu, 610054, China
| | - Zhanshan Wang
- Institute of Precision Optical Engineering, School of Physics Science and Engineering, Tongji University, Shanghai, 200092, China
- MOE Key Laboratory of Advanced Micro-Structured Materials, Shanghai, 200092, China
- Shanghai Institute of Intelligent Science and Technology, Tongji University, Shanghai, 200092, China
- Shanghai Frontiers Science Center of Digital Optics, Shanghai, 200092, China
| | - Cheng-Wei Qiu
- Department of Electrical and Computer Engineering, National University of Singapore, Singapore, 117583, Singapore.
| | - Hui Zhang
- Institute of Precision Optical Engineering, School of Physics Science and Engineering, Tongji University, Shanghai, 200092, China.
- MOE Key Laboratory of Advanced Micro-Structured Materials, Shanghai, 200092, China.
- Shanghai Institute of Intelligent Science and Technology, Tongji University, Shanghai, 200092, China.
- Shanghai Frontiers Science Center of Digital Optics, Shanghai, 200092, China.
| | - Xinbin Cheng
- Institute of Precision Optical Engineering, School of Physics Science and Engineering, Tongji University, Shanghai, 200092, China.
- MOE Key Laboratory of Advanced Micro-Structured Materials, Shanghai, 200092, China.
- Shanghai Institute of Intelligent Science and Technology, Tongji University, Shanghai, 200092, China.
- Shanghai Frontiers Science Center of Digital Optics, Shanghai, 200092, China.
| |
Collapse
|
3
|
Stölting G, Hellmig N, Dinh HA, Butz F, Secener AK, Volkert M, Scholl UI. Expression and function of Connexin 43 and Connexin 37 in the murine zona glomerulosa. Physiol Rep 2025; 13:e70215. [PMID: 39877942 PMCID: PMC11775451 DOI: 10.14814/phy2.70215] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2024] [Revised: 01/13/2025] [Accepted: 01/13/2025] [Indexed: 01/31/2025] Open
Abstract
The zona glomerulosa (ZG) synthesizes the mineralocorticoid aldosterone. The primary role of aldosterone is the maintenance of volume and electrolyte homeostasis. Aldosterone synthesis is primarily regulated via tightly controlled oscillations in intracellular calcium levels in response to stimulation. It has previously been shown that calcium oscillations are synchronized through mechanical linkage between adjacent ZG cells. In many other cell types, similar synchronization is rather dependent on gap junctions (GJ). The recent discovery of mutations in CADM1 was linked to impaired GJ function in the ZG. Based on published transcriptomics data, we re-examined the presence and functional impact of GJ in the ZG. We found evidence for the expression of murine connexin 43 and 37 using microarray data, in-situ hybridization and immunohistology. Connexin 43 was also present in human samples. Calcium oscillations in ZG rosettes showed some degree of synchronization as reported previously. Unspecific GJ inhibition only had a small impact on this synchronicity. However, no signs of connections between cytosols could be observed as indicated by the lack of fluorescence recovery after photobleaching. We conclude that, while connexin proteins are expressed in the ZG, functional GJ in the physiological ZG are rare and of little consequence for calcium signaling.
Collapse
Affiliation(s)
- Gabriel Stölting
- Berlin Institute of Health at CharitéUniversitätsmedizin Berlin, Center of Functional GenomicsBerlinGermany
| | - Nicole Hellmig
- Berlin Institute of Health at CharitéUniversitätsmedizin Berlin, Center of Functional GenomicsBerlinGermany
| | - Hoang An Dinh
- Berlin Institute of Health at CharitéUniversitätsmedizin Berlin, Center of Functional GenomicsBerlinGermany
- Institute of Translational PhysiologyCharité—Universitätsmedizin BerlinBerlinGermany
| | - Frederike Butz
- Berlin Institute of Health at CharitéUniversitätsmedizin Berlin, Center of Functional GenomicsBerlinGermany
- Department of Surgery, Campus Charité Mitte, Campus Virchow‐KlinikumCharité—Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt‐Universität Zu BerlinBerlinGermany
| | - Ali Kerim Secener
- Berlin Institute of Health at CharitéUniversitätsmedizin Berlin, Center of Functional GenomicsBerlinGermany
- Max Delbrück Center for Molecular Medicine in the Helmholtz Association (MDC)BerlinGermany
- Institute of Chemistry and Biochemistry, Department of Biology, Chemistry and PharmacyFreie Universität BerlinBerlinGermany
| | - Marina Volkert
- Berlin Institute of Health at CharitéUniversitätsmedizin Berlin, Center of Functional GenomicsBerlinGermany
| | - Ute I. Scholl
- Berlin Institute of Health at CharitéUniversitätsmedizin Berlin, Center of Functional GenomicsBerlinGermany
- Department of Nephrology and Medical Intensive CareCharité—Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt‐Universität Zu BerlinBerlinGermany
| |
Collapse
|
4
|
Sun Z, Kemter E, Pang Y, Bidlingmaier M, Wolf E, Reincke M, Williams TA. ATP2A3 in Primary Aldosteronism: Machine Learning-Based Discovery and Functional Validation. Hypertension 2025; 82:319-332. [PMID: 39618394 DOI: 10.1161/hypertensionaha.124.23817] [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: 08/08/2024] [Accepted: 11/15/2024] [Indexed: 01/18/2025]
Abstract
BACKGROUND Aldosterone-producing adenomas (APAs) are a common cause of primary aldosteronism that can lead to cardiovascular complications if left untreated. Machine learning-based bioinformatics approaches have emerged as powerful tools for identifying potential disease markers, gaining widespread recognition in biomedical research. We aimed to use machine learning to discover novel biomarkers of APAs to identify new pathophysiological mechanisms. METHODS We applied 2 machine learning algorithms to published RNA sequencing data to identify APA feature genes. Validation was performed using APA tissue samples, spatial transcriptomics, pig adrenal glands, and in vitro assays in a human adrenocortical cell line. RESULTS Machine learning identified ATP2A3 as a key feature gene in APA, and its upregulation in APAs compared with the adjacent cortex was confirmed by spatial transcriptomics. In human adrenocortical cells, angiotensin II treatment increased ATP2A3 gene expression 9.15-fold. Silencing ATP2A3 decreased basal CYP11B2 expression and aldosterone secretion by 3.51-fold and 1.46-fold, respectively, and by 1.77-fold and 1.94-fold under angiotensin II stimulation. Dietary sodium restriction in pigs significantly increased ATP2A3 mRNA and protein levels. Spatial transcriptomics showed that APA cells exhibited higher ATP2A3 gene expression compared with all other adrenal cell types. The suppressive effect of ATP2A3 silencing on CYP11B2 expression was further enhanced by Ca2+ inhibitors. CONCLUSIONS The ATP2A3 gene is highly expressed in APA and is a key regulator of CYP11B2 expression and aldosterone production.
Collapse
Affiliation(s)
- Zhuolun Sun
- Medizinische Klinik und Poliklinik IV, Klinikum der Universität München, Ludwig-Maximilians-Universität München, Germany (Z.S., Y.P., M.B., M.R., T.A.W.)
| | - Elisabeth Kemter
- Chair for Molecular Animal Breeding and Biotechnology, Gene Center and Department of Veterinary Sciences, Ludwig-Maximilians-Universität München, Germany (E.K., E.W.)
| | - Yingxian Pang
- Medizinische Klinik und Poliklinik IV, Klinikum der Universität München, Ludwig-Maximilians-Universität München, Germany (Z.S., Y.P., M.B., M.R., T.A.W.)
| | - Martin Bidlingmaier
- Medizinische Klinik und Poliklinik IV, Klinikum der Universität München, Ludwig-Maximilians-Universität München, Germany (Z.S., Y.P., M.B., M.R., T.A.W.)
| | - Eckhard Wolf
- Chair for Molecular Animal Breeding and Biotechnology, Gene Center and Department of Veterinary Sciences, Ludwig-Maximilians-Universität München, Germany (E.K., E.W.)
| | - Martin Reincke
- Medizinische Klinik und Poliklinik IV, Klinikum der Universität München, Ludwig-Maximilians-Universität München, Germany (Z.S., Y.P., M.B., M.R., T.A.W.)
| | - Tracy Ann Williams
- Medizinische Klinik und Poliklinik IV, Klinikum der Universität München, Ludwig-Maximilians-Universität München, Germany (Z.S., Y.P., M.B., M.R., T.A.W.)
| |
Collapse
|
5
|
Altieri B, Secener AK, Sai S, Fischer C, Sbiera S, Arampatzi P, Kircher S, Herterich S, Landwehr L, Vitcetz SN, Braeuning C, Fassnacht M, Ronchi CL, Sauer S. Single-nucleus and spatial transcriptome reveal adrenal homeostasis in normal and tumoural adrenal glands. Clin Transl Med 2024; 14:e1798. [PMID: 39167619 PMCID: PMC11338279 DOI: 10.1002/ctm2.1798] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2023] [Revised: 07/11/2024] [Accepted: 07/26/2024] [Indexed: 08/23/2024] Open
Abstract
The human adrenal gland is a complex endocrine tissue. Studies on adrenal renewal have been limited to animal models or human foetuses. Enhancing our understanding of adult human adrenal homeostasis is crucial for gaining insights into the pathogenesis of adrenal diseases, such as adrenocortical tumours. Here, we present a comprehensive cellular genomics analysis of the adult human normal adrenal gland, combining single-nuclei RNA sequencing and spatial transcriptome data to reconstruct adrenal gland homeostasis. As expected, we identified primary cells of the various zones of the adrenal cortex and medulla, but we also uncovered additional cell types. They constitute the adrenal microenvironment, including immune cells, mostly composed of a large population of M2 macrophages, and new cell populations, including different subpopulations of vascular-endothelial cells and cortical-neuroendocrine cells. Utilizing spatial transcriptome and pseudotime trajectory analysis, we support evidence of the centripetal dynamics of adrenocortical cell maintenance and the essential role played by Wnt/β-catenin, sonic hedgehog, and fibroblast growth factor pathways in the adult adrenocortical homeostasis. Furthermore, we compared single-nuclei transcriptional profiles obtained from six healthy adrenal glands and twelve adrenocortical adenomas. This analysis unveiled a notable heterogeneity in cell populations within the adenoma samples. In addition, we identified six distinct adenoma-specific clusters, each with varying distributions based on steroid profiles and tumour mutational status. Overall, our results provide novel insights into adrenal homeostasis and molecular mechanisms potentially underlying early adrenocortical tumorigenesis and/or autonomous steroid secretion. Our cell atlas represents a powerful resource to investigate other adrenal-related pathologies.
Collapse
Affiliation(s)
- Barbara Altieri
- Division of Endocrinology and DiabetesDepartment of Internal Medicine IUniversity HospitalUniversity of WürzburgWürzburgGermany
| | - A. Kerim Secener
- Max Delbrück Center for Molecular MedicineBerlinGermany
- Berlin Institute of HealthBerlinGermany
- Department of BiologyChemistry and PharmacyInstitute of BiochemistryFree University BerlinBerlinGermany
| | - Somesh Sai
- Max Delbrück Center for Molecular MedicineBerlinGermany
- Berlin Institute of HealthBerlinGermany
- Department of BiologyChemistry and PharmacyInstitute of BiochemistryFree University BerlinBerlinGermany
| | - Cornelius Fischer
- Max Delbrück Center for Molecular MedicineBerlinGermany
- Berlin Institute of HealthBerlinGermany
| | - Silviu Sbiera
- Division of Endocrinology and DiabetesDepartment of Internal Medicine IUniversity HospitalUniversity of WürzburgWürzburgGermany
| | | | - Stefan Kircher
- Institute of PathologyUniversity of WürzburgWürzburgGermany
| | | | - Laura‐Sophie Landwehr
- Division of Endocrinology and DiabetesDepartment of Internal Medicine IUniversity HospitalUniversity of WürzburgWürzburgGermany
| | - Sarah N. Vitcetz
- Max Delbrück Center for Molecular MedicineBerlinGermany
- Berlin Institute of HealthBerlinGermany
| | | | - Martin Fassnacht
- Division of Endocrinology and DiabetesDepartment of Internal Medicine IUniversity HospitalUniversity of WürzburgWürzburgGermany
- Central Laboratory University Hospital WürzburgWürzburgGermany
| | - Cristina L. Ronchi
- Division of Endocrinology and DiabetesDepartment of Internal Medicine IUniversity HospitalUniversity of WürzburgWürzburgGermany
- Institute of Metabolism and System ResearchUniversity of BirminghamEdgabston, BirminghamUK
| | - Sascha Sauer
- Max Delbrück Center for Molecular MedicineBerlinGermany
- Berlin Institute of HealthBerlinGermany
- Core Unit SysMedUniversity of WürzburgWürzburgGermany
| |
Collapse
|
6
|
Azizan EAB, Drake WM, Brown MJ. Primary aldosteronism: molecular medicine meets public health. Nat Rev Nephrol 2023; 19:788-806. [PMID: 37612380 PMCID: PMC7615304 DOI: 10.1038/s41581-023-00753-6] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/21/2023] [Indexed: 08/25/2023]
Abstract
Primary aldosteronism is the most common single cause of hypertension and is potentially curable when only one adrenal gland is the culprit. The importance of primary aldosteronism to public health derives from its high prevalence but huge under-diagnosis (estimated to be <1% of all affected individuals), despite the consequences of poor blood pressure control by conventional therapy and enhanced cardiovascular risk. This state of affairs is attributable to the fact that the tools used for diagnosis or treatment are still those that originated in the 1970-1990s. Conversely, molecular discoveries have transformed our understanding of adrenal physiology and pathology. Many molecules and processes associated with constant adrenocortical renewal and interzonal metamorphosis also feature in aldosterone-producing adenomas and aldosterone-producing micronodules. The adrenal gland has one of the most significant rates of non-silent somatic mutations, with frequent selection of those driving autonomous aldosterone production, and distinct clinical presentations and outcomes for most genotypes. The disappearance of aldosterone synthesis and cells from most of the adult human zona glomerulosa is the likely driver of the mutational success that causes aldosterone-producing adenomas, but insights into the pathways that lead to constitutive aldosterone production and cell survival may open up opportunities for novel therapies.
Collapse
Affiliation(s)
- Elena A B Azizan
- Department of Medicine, Faculty of Medicine, The National University of Malaysia (UKM), Kuala Lumpur, Malaysia
- Endocrine Hypertension, Department of Clinical Pharmacology and Precision Medicine, William Harvey Research Institute, Queen Mary University of London, London, United Kingdom
| | - William M Drake
- St Bartholomew's Hospital, Barts Health NHS Trust, London, United Kingdom
- NIHR Barts Biomedical Research Centre, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London, United Kingdom
| | - Morris J Brown
- Endocrine Hypertension, Department of Clinical Pharmacology and Precision Medicine, William Harvey Research Institute, Queen Mary University of London, London, United Kingdom.
- NIHR Barts Biomedical Research Centre, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London, United Kingdom.
| |
Collapse
|
7
|
Wu X, Azizan EAB, Goodchild E, Garg S, Hagiyama M, Cabrera CP, Fernandes-Rosa FL, Boulkroun S, Kuan JL, Tiang Z, David A, Murakami M, Mein CA, Wozniak E, Zhao W, Marker A, Buss F, Saleeb RS, Salsbury J, Tezuka Y, Satoh F, Oki K, Udager AM, Cohen DL, Wachtel H, King PJ, Drake WM, Gurnell M, Ceral J, Ryska A, Mustangin M, Wong YP, Tan GC, Solar M, Reincke M, Rainey WE, Foo RS, Takaoka Y, Murray SA, Zennaro MC, Beuschlein F, Ito A, Brown MJ. Somatic mutations of CADM1 in aldosterone-producing adenomas and gap junction-dependent regulation of aldosterone production. Nat Genet 2023; 55:1009-1021. [PMID: 37291193 PMCID: PMC10260400 DOI: 10.1038/s41588-023-01403-0] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2022] [Accepted: 04/20/2023] [Indexed: 06/10/2023]
Abstract
Aldosterone-producing adenomas (APAs) are the commonest curable cause of hypertension. Most have gain-of-function somatic mutations of ion channels or transporters. Herein we report the discovery, replication and phenotype of mutations in the neuronal cell adhesion gene CADM1. Independent whole exome sequencing of 40 and 81 APAs found intramembranous p.Val380Asp or p.Gly379Asp variants in two patients whose hypertension and periodic primary aldosteronism were cured by adrenalectomy. Replication identified two more APAs with each variant (total, n = 6). The most upregulated gene (10- to 25-fold) in human adrenocortical H295R cells transduced with the mutations (compared to wildtype) was CYP11B2 (aldosterone synthase), and biological rhythms were the most differentially expressed process. CADM1 knockdown or mutation inhibited gap junction (GJ)-permeable dye transfer. GJ blockade by Gap27 increased CYP11B2 similarly to CADM1 mutation. Human adrenal zona glomerulosa (ZG) expression of GJA1 (the main GJ protein) was patchy, and annular GJs (sequelae of GJ communication) were less prominent in CYP11B2-positive micronodules than adjacent ZG. Somatic mutations of CADM1 cause reversible hypertension and reveal a role for GJ communication in suppressing physiological aldosterone production.
Collapse
Affiliation(s)
- Xilin Wu
- Endocrine Hypertension, Department of Clinical Pharmacology and Precision Medicine, William Harvey Research Institute, Queen Mary University of London, London, UK
- NIHR Barts Biomedical Research Centre, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London, UK
- St Bartholomew's Hospital, Barts Health NHS Trust, London, UK
| | - Elena A B Azizan
- Endocrine Hypertension, Department of Clinical Pharmacology and Precision Medicine, William Harvey Research Institute, Queen Mary University of London, London, UK.
- Department of Medicine, Faculty of Medicine, Universiti Kebangsaan Malaysia, Kuala Lumpur, Malaysia.
| | - Emily Goodchild
- Endocrine Hypertension, Department of Clinical Pharmacology and Precision Medicine, William Harvey Research Institute, Queen Mary University of London, London, UK
- NIHR Barts Biomedical Research Centre, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London, UK
- St Bartholomew's Hospital, Barts Health NHS Trust, London, UK
| | - Sumedha Garg
- Endocrine Hypertension, Department of Clinical Pharmacology and Precision Medicine, William Harvey Research Institute, Queen Mary University of London, London, UK
- Clinical Pharmacology Unit, University of Cambridge, Cambridge, UK
| | - Man Hagiyama
- Department of Pathology, Faculty of Medicine, Kindai University, Osakasayama, Japan
| | - Claudia P Cabrera
- NIHR Barts Biomedical Research Centre, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London, UK
- Centre for Translational Bioinformatics, William Harvey Research Institute, Queen Mary University of London, London, UK
| | | | | | - Jyn Ling Kuan
- Cardiovascular Disease Translational Research Programme, Department of Medicine, National University of Singapore, Singapore, Singapore
| | - Zenia Tiang
- Cardiovascular Disease Translational Research Programme, Department of Medicine, National University of Singapore, Singapore, Singapore
| | - Alessia David
- Centre for Bioinformatics, Department of Life Sciences, Imperial College London, London, UK
| | - Masanori Murakami
- Medizinische Klinik und Poliklinik IV, Klinikum der Universität, Ludwig-Maximilians-Universität München, Munich, Germany
| | - Charles A Mein
- Barts and London Genome Centre, School of Medicine and Dentistry, Blizard Institute, London, UK
| | - Eva Wozniak
- Barts and London Genome Centre, School of Medicine and Dentistry, Blizard Institute, London, UK
| | - Wanfeng Zhao
- Department of Histopathology, Addenbrooke's Hospital, Cambridge, UK
| | - Alison Marker
- Department of Histopathology, Addenbrooke's Hospital, Cambridge, UK
| | - Folma Buss
- Cambridge Institute for Medical Research, The Keith Peters Building, University of Cambridge, Cambridge, UK
| | - Rebecca S Saleeb
- Centre for Microvascular Research, William Harvey Research Institute, Queen Mary University of London, London, UK
| | - Jackie Salsbury
- Endocrine Hypertension, Department of Clinical Pharmacology and Precision Medicine, William Harvey Research Institute, Queen Mary University of London, London, UK
- NIHR Barts Biomedical Research Centre, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London, UK
- St Bartholomew's Hospital, Barts Health NHS Trust, London, UK
| | - Yuta Tezuka
- Division of Nephrology, Endocrinology, and Vascular Medicine, Tohoku University Hospital, Sendai, Japan
| | - Fumitoshi Satoh
- Division of Nephrology, Endocrinology, and Vascular Medicine, Tohoku University Hospital, Sendai, Japan
- Division of Clinical Hypertension, Endocrinology and Metabolism, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Kenji Oki
- Department of Molecular and Internal Medicine, Graduate School of Biomedical and Health Sciences, Hiroshima University, Hiroshima, Japan
| | - Aaron M Udager
- Department of Pathology, University of Michigan Medical School, Ann Arbor, MI, USA
| | - Debbie L Cohen
- Renal Division, Department of Medicine, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, USA
| | - Heather Wachtel
- Department of Surgery, Hospital of the University of Pennsylvania, Philadelphia, PA, USA
| | - Peter J King
- Department of Endocrinology, William Harvey Research Institute, Queen Mary University of London, London, UK
| | - William M Drake
- NIHR Barts Biomedical Research Centre, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London, UK
- St Bartholomew's Hospital, Barts Health NHS Trust, London, UK
| | - Mark Gurnell
- Metabolic Research Laboratories, Welcome Trust-MRC Institute of Metabolic Science, and NIHR Cambridge Biomedical Research Centre, Cambridge Biomedical Campus, Cambridge, UK
| | - Jiri Ceral
- 1st Department of Internal Medicine-Cardioangiology, Charles University Faculty of Medicine in Hradec Kralove and University Hospital Hradec Kralove, Hradec Kralove, Czech Republic
| | - Ales Ryska
- Department of Pathology, Charles University Faculty of Medicine in Hradec Kralove and University Hospital Hradec Kralove, Hradec Kralove, Czech Republic
| | - Muaatamarulain Mustangin
- Department of Pathology, Faculty of Medicine, Universiti Kebangsaan Malaysia, Kuala Lumpur, Malaysia
| | - Yin Ping Wong
- Department of Pathology, Faculty of Medicine, Universiti Kebangsaan Malaysia, Kuala Lumpur, Malaysia
| | - Geok Chin Tan
- Department of Pathology, Faculty of Medicine, Universiti Kebangsaan Malaysia, Kuala Lumpur, Malaysia
| | - Miroslav Solar
- 1st Department of Internal Medicine-Cardioangiology, Charles University Faculty of Medicine in Hradec Kralove and University Hospital Hradec Kralove, Hradec Kralove, Czech Republic
| | - Martin Reincke
- Medizinische Klinik und Poliklinik IV, Klinikum der Universität, Ludwig-Maximilians-Universität München, Munich, Germany
| | - William E Rainey
- Division of Metabolism, Endocrinology, and Diabetes, University of Michigan, Ann Arbor, MI, USA
| | - Roger S Foo
- Cardiovascular Disease Translational Research Programme, Department of Medicine, National University of Singapore, Singapore, Singapore
| | - Yutaka Takaoka
- Department of Computational Drug Design and Mathematical Medicine, Graduate School of Medicine and Pharmaceutical Sciences, University of Toyama, Toyoma, Japan
| | - Sandra A Murray
- Department of Cell Biology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Maria-Christina Zennaro
- Université Paris Cité, PARCC, Inserm, Paris, France
- Assistance Publique-Hôpitaux de Paris, Hôpital Européen Georges Pompidou, Service de Génétique, Paris, France
| | - Felix Beuschlein
- Klinik für Endokrinologie, Diabetologie und Klinische Ernährung, UniversitätsSpital Zürich (USZ) und Universität Zürich (UZH), Zurich, Switzerland
- Medizinische Klinik und Poliklinik IV, Klinikum der Universität, Ludwig-Maximilians-Universität München, Munich, Germany
| | - Akihiko Ito
- Department of Pathology, Faculty of Medicine, Kindai University, Osakasayama, Japan
| | - Morris J Brown
- Endocrine Hypertension, Department of Clinical Pharmacology and Precision Medicine, William Harvey Research Institute, Queen Mary University of London, London, UK.
- NIHR Barts Biomedical Research Centre, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London, UK.
| |
Collapse
|
8
|
Wu X, Senanayake R, Goodchild E, Bashari WA, Salsbury J, Cabrera CP, Argentesi G, O’Toole SM, Matson M, Koo B, Parvanta L, Hilliard N, Kosmoliaptsis V, Marker A, Berney DM, Tan W, Foo R, Mein CA, Wozniak E, Savage E, Sahdev A, Bird N, Laycock K, Boros I, Hader S, Warnes V, Gillett D, Dawnay A, Adeyeye E, Prete A, Taylor AE, Arlt W, Bhuva AN, Aigbirhio F, Manisty C, McIntosh A, McConnachie A, Cruickshank JK, Cheow H, Gurnell M, Drake WM, Brown MJ. [ 11C]metomidate PET-CT versus adrenal vein sampling for diagnosing surgically curable primary aldosteronism: a prospective, within-patient trial. Nat Med 2023; 29:190-202. [PMID: 36646800 PMCID: PMC9873572 DOI: 10.1038/s41591-022-02114-5] [Citation(s) in RCA: 42] [Impact Index Per Article: 21.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2021] [Accepted: 10/31/2022] [Indexed: 01/18/2023]
Abstract
Primary aldosteronism (PA) due to a unilateral aldosterone-producing adenoma is a common cause of hypertension. This can be cured, or greatly improved, by adrenal surgery. However, the invasive nature of the standard pre-surgical investigation contributes to fewer than 1% of patients with PA being offered the chance of a cure. The primary objective of our prospective study of 143 patients with PA ( NCT02945904 ) was to compare the accuracy of a non-invasive test, [11C]metomidate positron emission tomography computed tomography (MTO) scanning, with adrenal vein sampling (AVS) in predicting the biochemical remission of PA and the resolution of hypertension after surgery. A total of 128 patients reached 6- to 9-month follow-up, with 78 (61%) treated surgically and 50 (39%) managed medically. Of the 78 patients receiving surgery, 77 achieved one or more PA surgical outcome criterion for success. The accuracies of MTO at predicting biochemical and clinical success following adrenalectomy were, respectively, 72.7 and 65.4%. For AVS, the accuracies were 63.6 and 61.5%. MTO was not significantly superior, but the differences of 9.1% (95% confidence interval = -6.5 to 24.1%) and 3.8% (95% confidence interval = -11.9 to 9.4) lay within the pre-specified -17% margin for non-inferiority (P = 0.00055 and P = 0.0077, respectively). Of 24 serious adverse events, none was considered related to either investigation and 22 were fully resolved. MTO enables non-invasive diagnosis of unilateral PA.
Collapse
Affiliation(s)
- Xilin Wu
- grid.4868.20000 0001 2171 1133Endocrine Hypertension, Department of Clinical Pharmacology, William Harvey Research Institute, Queen Mary University of London, London, United Kingdom ,grid.4868.20000 0001 2171 1133NIHR Barts Cardiovascular Biomedical Research Centre, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London, United Kingdom ,grid.139534.90000 0001 0372 5777Department of Endocrinology, St Bartholomew’s Hospital, Barts Health NHS Trust, London, United Kingdom
| | - Russell Senanayake
- grid.5335.00000000121885934Metabolic Research Laboratories, Wellcome–MRC Institute of Metabolic Science, University of Cambridge, Cambridge, United Kingdom ,grid.24029.3d0000 0004 0383 8386NIHR Cambridge Biomedical Research Centre, Addenbrooke’s Hospital, Cambridge University Hospitals NHS Foundation Trust, Cambridge, United Kingdom ,grid.24029.3d0000 0004 0383 8386Department of Diabetes and Endocrinology, Addenbrooke’s Hospital, Cambridge University Hospitals NHS Foundation Trust, Cambridge, United Kingdom
| | - Emily Goodchild
- grid.4868.20000 0001 2171 1133Endocrine Hypertension, Department of Clinical Pharmacology, William Harvey Research Institute, Queen Mary University of London, London, United Kingdom ,grid.4868.20000 0001 2171 1133NIHR Barts Cardiovascular Biomedical Research Centre, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London, United Kingdom ,grid.139534.90000 0001 0372 5777Department of Endocrinology, St Bartholomew’s Hospital, Barts Health NHS Trust, London, United Kingdom
| | - Waiel A. Bashari
- grid.5335.00000000121885934Metabolic Research Laboratories, Wellcome–MRC Institute of Metabolic Science, University of Cambridge, Cambridge, United Kingdom ,grid.24029.3d0000 0004 0383 8386NIHR Cambridge Biomedical Research Centre, Addenbrooke’s Hospital, Cambridge University Hospitals NHS Foundation Trust, Cambridge, United Kingdom ,grid.24029.3d0000 0004 0383 8386Department of Diabetes and Endocrinology, Addenbrooke’s Hospital, Cambridge University Hospitals NHS Foundation Trust, Cambridge, United Kingdom
| | - Jackie Salsbury
- grid.4868.20000 0001 2171 1133Endocrine Hypertension, Department of Clinical Pharmacology, William Harvey Research Institute, Queen Mary University of London, London, United Kingdom ,grid.4868.20000 0001 2171 1133NIHR Barts Cardiovascular Biomedical Research Centre, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London, United Kingdom
| | - Claudia P. Cabrera
- grid.4868.20000 0001 2171 1133Centre for Translational Bioinformatics, William Harvey Research Institute, Queen Mary University of London, London, United Kingdom
| | - Giulia Argentesi
- grid.4868.20000 0001 2171 1133Endocrine Hypertension, Department of Clinical Pharmacology, William Harvey Research Institute, Queen Mary University of London, London, United Kingdom ,grid.4868.20000 0001 2171 1133NIHR Barts Cardiovascular Biomedical Research Centre, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London, United Kingdom ,grid.139534.90000 0001 0372 5777Department of Endocrinology, St Bartholomew’s Hospital, Barts Health NHS Trust, London, United Kingdom
| | - Samuel M. O’Toole
- grid.4868.20000 0001 2171 1133Endocrine Hypertension, Department of Clinical Pharmacology, William Harvey Research Institute, Queen Mary University of London, London, United Kingdom ,grid.4868.20000 0001 2171 1133NIHR Barts Cardiovascular Biomedical Research Centre, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London, United Kingdom ,grid.139534.90000 0001 0372 5777Department of Endocrinology, St Bartholomew’s Hospital, Barts Health NHS Trust, London, United Kingdom ,grid.416126.60000 0004 0641 6031Department of Endocrinology, Royal Hallamshire Hospital, Sheffield, United Kingdom
| | - Matthew Matson
- grid.139534.90000 0001 0372 5777Department of Radiology, St Bartholomew’s Hospital, Barts Health NHS Trust, London, United Kingdom
| | - Brendan Koo
- grid.24029.3d0000 0004 0383 8386Department of Radiology, Addenbrooke’s Hospital, Cambridge University Hospitals NHS Foundation Trust, Cambridge, United Kingdom
| | - Laila Parvanta
- grid.139534.90000 0001 0372 5777Department of Endocrinology, St Bartholomew’s Hospital, Barts Health NHS Trust, London, United Kingdom
| | - Nick Hilliard
- grid.24029.3d0000 0004 0383 8386Department of Radiology, Addenbrooke’s Hospital, Cambridge University Hospitals NHS Foundation Trust, Cambridge, United Kingdom
| | - Vasilis Kosmoliaptsis
- grid.24029.3d0000 0004 0383 8386Department of Surgery, Addenbrooke’s Hospital, Cambridge University Hospitals NHS Foundation Trust, Cambridge, United Kingdom
| | - Alison Marker
- grid.24029.3d0000 0004 0383 8386Department of Histopathology, Addenbrooke’s Hospital, Cambridge University Hospitals NHS Foundation Trust, Cambridge, United Kingdom
| | - Daniel M. Berney
- grid.139534.90000 0001 0372 5777Department of Histopathology, St Bartholomew’s Hospital, Barts Health NHS Trust, London, United Kingdom
| | - Wilson Tan
- grid.4280.e0000 0001 2180 6431Cardiovascular Research Institute, National University of Singapore, Singapore, Singapore
| | - Roger Foo
- grid.4280.e0000 0001 2180 6431Cardiovascular Research Institute, National University of Singapore, Singapore, Singapore
| | - Charles A. Mein
- grid.4868.20000 0001 2171 1133Barts and the London Genome Centre, School of Medicine and Dentistry, Blizard Institute, London, United Kingdom
| | - Eva Wozniak
- grid.4868.20000 0001 2171 1133Barts and the London Genome Centre, School of Medicine and Dentistry, Blizard Institute, London, United Kingdom
| | - Emmanuel Savage
- grid.4868.20000 0001 2171 1133Barts and the London Genome Centre, School of Medicine and Dentistry, Blizard Institute, London, United Kingdom
| | - Anju Sahdev
- grid.139534.90000 0001 0372 5777Department of Radiology, St Bartholomew’s Hospital, Barts Health NHS Trust, London, United Kingdom
| | - Nicholas Bird
- grid.24029.3d0000 0004 0383 8386Department of Radiology, Addenbrooke’s Hospital, Cambridge University Hospitals NHS Foundation Trust, Cambridge, United Kingdom
| | - Kate Laycock
- grid.4868.20000 0001 2171 1133Endocrine Hypertension, Department of Clinical Pharmacology, William Harvey Research Institute, Queen Mary University of London, London, United Kingdom ,grid.4868.20000 0001 2171 1133NIHR Barts Cardiovascular Biomedical Research Centre, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London, United Kingdom ,grid.139534.90000 0001 0372 5777Department of Endocrinology, St Bartholomew’s Hospital, Barts Health NHS Trust, London, United Kingdom
| | - Istvan Boros
- grid.5335.00000000121885934Wolfson Brain Imaging Centre, University of Cambridge, Cambridge, United Kingdom
| | - Stefan Hader
- grid.5335.00000000121885934Wolfson Brain Imaging Centre, University of Cambridge, Cambridge, United Kingdom
| | - Victoria Warnes
- grid.24029.3d0000 0004 0383 8386Department of Nuclear Medicine, Addenbrooke’s Hospital, Cambridge University Hospitals NHS Foundation Trust, Cambridge, United Kingdom
| | - Daniel Gillett
- grid.24029.3d0000 0004 0383 8386Department of Nuclear Medicine, Addenbrooke’s Hospital, Cambridge University Hospitals NHS Foundation Trust, Cambridge, United Kingdom
| | - Anne Dawnay
- grid.139534.90000 0001 0372 5777Department of Clinical Biochemistry, St Bartholomew’s Hospital, Barts Health NHS Trust, London, United Kingdom
| | - Elizabeth Adeyeye
- grid.420545.20000 0004 0489 3985Department of Cardiovascular Medicine/Diabetes, Guy’s and St Thomas’ NHS Foundation Trust, London, United Kingdom
| | - Alessandro Prete
- grid.6572.60000 0004 1936 7486Institute of Metabolism and Systems Research, University of Birmingham, Birmingham, United Kingdom
| | - Angela E. Taylor
- grid.6572.60000 0004 1936 7486Institute of Metabolism and Systems Research, University of Birmingham, Birmingham, United Kingdom
| | - Wiebke Arlt
- grid.6572.60000 0004 1936 7486Institute of Metabolism and Systems Research, University of Birmingham, Birmingham, United Kingdom ,grid.412563.70000 0004 0376 6589NIHR Birmingham Biomedical Research Centre, University Hospitals Birmingham NHS Foundation Trust and University of Birmingham, Birmingham, UK
| | - Anish N. Bhuva
- grid.139534.90000 0001 0372 5777Department of Cardiology, St Bartholomew’s Hospital, Barts Health NHS Trust, London, United Kingdom
| | - Franklin Aigbirhio
- grid.5335.00000000121885934Wolfson Brain Imaging Centre, University of Cambridge, Cambridge, United Kingdom
| | - Charlotte Manisty
- grid.139534.90000 0001 0372 5777Department of Cardiology, St Bartholomew’s Hospital, Barts Health NHS Trust, London, United Kingdom
| | - Alasdair McIntosh
- grid.8756.c0000 0001 2193 314XRobertson Centre for Biostatistics, University of Glasgow, Glasgow, United Kingdom
| | - Alexander McConnachie
- grid.8756.c0000 0001 2193 314XRobertson Centre for Biostatistics, University of Glasgow, Glasgow, United Kingdom
| | - J. Kennedy Cruickshank
- grid.420545.20000 0004 0489 3985Department of Cardiovascular Medicine/Diabetes, Guy’s and St Thomas’ NHS Foundation Trust, London, United Kingdom ,grid.13097.3c0000 0001 2322 6764School of Life Course/Nutritional Sciences, King’s College London, London, United Kingdom
| | - Heok Cheow
- grid.24029.3d0000 0004 0383 8386Department of Radiology, Addenbrooke’s Hospital, Cambridge University Hospitals NHS Foundation Trust, Cambridge, United Kingdom
| | - Mark Gurnell
- grid.5335.00000000121885934Metabolic Research Laboratories, Wellcome–MRC Institute of Metabolic Science, University of Cambridge, Cambridge, United Kingdom ,grid.24029.3d0000 0004 0383 8386NIHR Cambridge Biomedical Research Centre, Addenbrooke’s Hospital, Cambridge University Hospitals NHS Foundation Trust, Cambridge, United Kingdom ,grid.24029.3d0000 0004 0383 8386Department of Diabetes and Endocrinology, Addenbrooke’s Hospital, Cambridge University Hospitals NHS Foundation Trust, Cambridge, United Kingdom
| | - William M. Drake
- grid.4868.20000 0001 2171 1133NIHR Barts Cardiovascular Biomedical Research Centre, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London, United Kingdom ,grid.139534.90000 0001 0372 5777Department of Endocrinology, St Bartholomew’s Hospital, Barts Health NHS Trust, London, United Kingdom
| | - Morris J. Brown
- grid.4868.20000 0001 2171 1133Endocrine Hypertension, Department of Clinical Pharmacology, William Harvey Research Institute, Queen Mary University of London, London, United Kingdom ,grid.4868.20000 0001 2171 1133NIHR Barts Cardiovascular Biomedical Research Centre, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London, United Kingdom ,grid.139534.90000 0001 0372 5777Department of Endocrinology, St Bartholomew’s Hospital, Barts Health NHS Trust, London, United Kingdom
| |
Collapse
|
9
|
Bao M, Li H, Li J. Identification of potential
lncRNA‐miRNA‐mRNA
regulatory network contributing to aldosterone‐producing adenoma. J Cell Mol Med 2022; 26:5614-5623. [DOI: 10.1111/jcmm.17586] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2021] [Revised: 09/27/2022] [Accepted: 10/05/2022] [Indexed: 11/28/2022] Open
Affiliation(s)
- Minghui Bao
- Department of Cardiology, Peking University First Hospital Peking University Beijing China
| | - Haotong Li
- National Center for Cardiovascular Diseases, Fuwai Hospital Chinese Academy of Medical Sciences and Peking Union Medical College Beijing China
| | - Jianping Li
- Department of Cardiology, Peking University First Hospital Peking University Beijing China
| |
Collapse
|
10
|
Ji K, Jiao D, Yang G, Degen AA, Zhou J, Liu H, Wang W, Cong H. Transcriptome analysis revealed potential genes involved in thermogenesis in muscle tissue in cold-exposed lambs. Front Genet 2022; 13:1017458. [PMID: 36338953 PMCID: PMC9634817 DOI: 10.3389/fgene.2022.1017458] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2022] [Accepted: 10/05/2022] [Indexed: 11/28/2022] Open
Abstract
Cold tolerance is an important trait for sheep raised at high altitudes. Muscle tissue, comprising 30–40% of the total body mass, produces heat during cold exposure. However, little is known about the genetic mechanisms of this tissue and its role in thermogenesis in lambs. We examined genes in skeletal muscle tissue in a cold-adapted sheep breed, Altay, and a cold-intolerant sheep breed, Hu, when exposed to low air temperature. Three ewe-lambs of each breed were maintained at −5°C and three ewe-lambs of each breed were maintained at 20°C. After cold exposure for 25 days, the longissimus dorsi of each lamb was collected, and transcriptome profiles were sequenced and analyzed. The results of RNA-seq showed that the average reads among the four groups were 11.0 Gbase. The genome mapping rate averaged 88.1% and the gene mapping rate averaged 82.5%. The analysis of differentially expressed genes (DEGs) indicated that the peroxisome proliferator-activated receptors (PPAR), cAMP, and calcium signaling pathways and muscle contraction in muscle tissue were linked to thermogenesis in cold-exposed lambs. Furthermore, PCK1 (phosphoenolpyruvate carboxykinase1) increased glyceroneogenesis in cold-exposed Altay lambs, and APOC3 (apolipoprotein C3), LPL (lipoprotein lipase), and FABP4 (fatty acid binding protein 4, adipocyte) were involved in the intake and transport of free fatty acids. In Hu sheep, cAMP biosynthesis from ATP hydrolysis was regulated by ADCY10 (adenylate cyclase) and ADORA2a (adenosine A2a receptor). Skeletal muscle contraction was regulated by MYL2 (myosin light chain 2). In conclusion, cold exposure altered the expression level of genes involved in heat production in muscle tissue. Some potential mechanisms were revealed, including calcium ion transport in the calcium signaling pathway, fatty acid metabolism in the PPAR signaling pathway, and cAMP biosynthesis in the cAMP signaling pathway. This study implied that skeletal muscle plays an important role in thermoregulation in lambs.
Collapse
Affiliation(s)
- Kaixi Ji
- Key Laboratory of Stress Physiology and Ecology of Gansu Province, Northwest Institute of Eco-Environment and Resources, Chinese Academy of Sciences, Lanzhou, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Dan Jiao
- Key Laboratory of Stress Physiology and Ecology of Gansu Province, Northwest Institute of Eco-Environment and Resources, Chinese Academy of Sciences, Lanzhou, China
| | - Guo Yang
- Key Laboratory of Stress Physiology and Ecology of Gansu Province, Northwest Institute of Eco-Environment and Resources, Chinese Academy of Sciences, Lanzhou, China
- *Correspondence: Guo Yang,
| | - Abraham Allan Degen
- Desert Animal Adaptations and Husbandry, Wyler Department of Dryland Agriculture, Blaustein Institutes for Desert Research, Ben-Gurion University of Negev, Beer Sheva, Israel
| | - Jianwei Zhou
- State Key Laboratory of Grassland and Agro-Ecosystems, College of Pastoral Agriculture Science and Technology, Lanzhou University, Lanzhou, China
| | - Hu Liu
- College of Ecology, Lanzhou University, Lanzhou, China
| | - Wenqiang Wang
- College of Ecology, Lanzhou University, Lanzhou, China
| | - Haitao Cong
- Dongying Modern Animal Husbandry Development Service Center, Dongying, China
| |
Collapse
|
11
|
Tetti M, Gong S, Veglio F, Reincke M, Williams TA. Primary aldosteronism: Pathophysiological mechanisms of cell death and proliferation. Front Endocrinol (Lausanne) 2022; 13:934326. [PMID: 36004349 PMCID: PMC9393369 DOI: 10.3389/fendo.2022.934326] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/02/2022] [Accepted: 07/18/2022] [Indexed: 11/17/2022] Open
Abstract
Primary aldosteronism is the most common surgically curable form of hypertension. The sporadic forms of the disorder are usually caused by aldosterone overproduction from a unilateral adrenocortical aldosterone-producing adenoma or from bilateral adrenocortical hyperplasia. The main knowledge-advances in disease pathophysiology focus on pathogenic germline and somatic variants that drive the excess aldosterone production. Less clear are the molecular and cellular mechanisms that lead to an increased mass of the adrenal cortex. However, the combined application of transcriptomics, metabolomics, and epigenetics has achieved substantial insight into these processes and uncovered the evolving complexity of disrupted cell growth mechanisms in primary aldosteronism. In this review, we summarize and discuss recent progress in our understanding of mechanisms of cell death, and proliferation in the pathophysiology of primary aldosteronism.
Collapse
Affiliation(s)
- Martina Tetti
- Medizinische Klinik und Poliklinik IV, Klinikum der Universität München, Ludwig-Maximilians-Universität (LMU) München, München, Germany
- Division of Internal Medicine and Hypertension, Department of Medical Sciences, University of Turin, Turin, Italy
| | - Siyuan Gong
- Medizinische Klinik und Poliklinik IV, Klinikum der Universität München, Ludwig-Maximilians-Universität (LMU) München, München, Germany
| | - Franco Veglio
- Division of Internal Medicine and Hypertension, Department of Medical Sciences, University of Turin, Turin, Italy
| | - Martin Reincke
- Medizinische Klinik und Poliklinik IV, Klinikum der Universität München, Ludwig-Maximilians-Universität (LMU) München, München, Germany
| | - Tracy Ann Williams
- Medizinische Klinik und Poliklinik IV, Klinikum der Universität München, Ludwig-Maximilians-Universität (LMU) München, München, Germany
- Division of Internal Medicine and Hypertension, Department of Medical Sciences, University of Turin, Turin, Italy
| |
Collapse
|
12
|
Matsuoka H, Harada K, Sugawara A, Kim D, Inoue M. Expression of p11 and heteromeric TASK channels in mouse adrenal cortical cells and H295R cells. Acta Histochem 2022; 124:151898. [PMID: 35526370 DOI: 10.1016/j.acthis.2022.151898] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2022] [Revised: 04/21/2022] [Accepted: 04/21/2022] [Indexed: 11/30/2022]
Abstract
TWIK-related acid-sensitive K+ (TASK) channels are thought to contribute to the resting membrane potential in adrenal cortical (AC) cells. However, the molecular identity of TASK channels in AC cells have not yet been elucidated. Thus, immunocytochemical and molecular biological approaches were employed to investigate the expression and intracellular distribution of TASK1 and TASK3 in mouse AC cells and H295R cells derived from human adrenocortical carcinoma. Immunocytochemical study revealed that immunoreactive materials were mainly located in the cytoplasm for TASK1 and at the cell periphery for TASK3 in mouse AC cells. A similar pattern of localization was observed when GFP-TASK1 and GFP-TASK3 were exogenously expressed in H295R cells. In addition, p11 that is known to suppress the endoplasmic reticulum exit of TASK1 was localized in the cytoplasm in mouse AC and H295R cells, but not in adrenal medullary cells. Proximity ligation assay (PLA) suggested formation of heteromeric TASK1-3 channels that were found predominantly in the cytoplasm and weakly at the cell periphery. A similar distribution was observed following exogenous expression of tandem TASK1-3 channels in H295R cells. When stimulated by angiotensin II, however, tandem TASK1-3 channels were present mainly in the cytoplasm in all H295R cells. In contrast to that in H295R cells, tandem channels were exclusively located at the cell periphery in all non-stimulated and exclusively in the cytoplasm in stimulated PC12 cells, respectively. From these results, we conclude that TASK1 proteins are present mainly in the cytoplasm and minimally at the cell periphery as a heteromeric channel with TASK3, whereas the majority of TASK3 is at the cell periphery as homomeric and heteromeric channels.
Collapse
Affiliation(s)
- Hidetada Matsuoka
- Department of Cell and Systems Physiology, University of Occupational and Environmental Health School of Medicine, Kitakyushu 807-8555, Japan
| | - Keita Harada
- Department of Cell and Systems Physiology, University of Occupational and Environmental Health School of Medicine, Kitakyushu 807-8555, Japan
| | - Akira Sugawara
- Department of Molecular Endocrinology, Tohoku University Graduate School of Medical Science, Sendai 980-8575, Japan
| | - Donghee Kim
- Department of Physiology and Biophysics, Chicago Medical School, Rosalind Franklin University of Medicine and Science, North Chicago, IL 60064-3095, USA
| | - Masumi Inoue
- Department of Cell and Systems Physiology, University of Occupational and Environmental Health School of Medicine, Kitakyushu 807-8555, Japan.
| |
Collapse
|
13
|
Nanao Y, Oki K, Kobuke K, Itcho K, Baba R, Kodama T, Otagaki Y, Okada A, Yoshii Y, Nagano G, Ohno H, Arihiro K, Gomez-Sanchez CE, Hattori N, Yoneda M. Hypomethylation associated vitamin D receptor expression in ATP1A1 mutant aldosterone-producing adenoma. Mol Cell Endocrinol 2022; 548:111613. [PMID: 35257799 PMCID: PMC9082579 DOI: 10.1016/j.mce.2022.111613] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/06/2022] [Revised: 02/25/2022] [Accepted: 03/01/2022] [Indexed: 12/25/2022]
Abstract
DNA methylation alteration is tissue-specific and play a pivotal role in regulating gene transcription during cell proliferation and survival. We aimed to detect genes regulated by DNA methylation, and then investigated whether the gene influenced cell proliferation or survival in adrenal cells. DNA methylation and qPCR analyses were performed in nonfunctioning adrenocortical adenoma (NFA, n = 12) and aldosterone-producing adenoma (APA, n = 35) samples. The VDR gene promoter was markedly hypomethylated in APA with ATP1A1 mutation, and the promoter methylation levels showed a significant inverse association with the transcripts in APA. ATP1A1 mutation led to VDR transcription in HAC15 cells, and VDR suppression abrogated ATP1A1 mutation-mediated cell proliferation in HAC15 cells. We demonstrated that APA with ATP1A1 mutation showed entire hypomethylation in the VDR promoter and abundant VDR mRNA and protein expression. VDR suppression abrogated ATP1A1 mutation-mediated cell proliferation in HAC15 cells. Abundant VDR expression would be essential for ATP1A1 mutation-mediated cell proliferation.
Collapse
Affiliation(s)
- Yuta Nanao
- Department of Molecular and Internal Medicine, Graduate School of Biomedical and Health Sciences, Hiroshima University, Hiroshima, Japan
| | - Kenji Oki
- Department of Molecular and Internal Medicine, Graduate School of Biomedical and Health Sciences, Hiroshima University, Hiroshima, Japan.
| | - Kazuhiro Kobuke
- Department of Molecular and Internal Medicine, Graduate School of Biomedical and Health Sciences, Hiroshima University, Hiroshima, Japan
| | - Kiyotaka Itcho
- Department of Molecular and Internal Medicine, Graduate School of Biomedical and Health Sciences, Hiroshima University, Hiroshima, Japan
| | - Ryuta Baba
- Department of Molecular and Internal Medicine, Graduate School of Biomedical and Health Sciences, Hiroshima University, Hiroshima, Japan
| | - Takaya Kodama
- Department of Molecular and Internal Medicine, Graduate School of Biomedical and Health Sciences, Hiroshima University, Hiroshima, Japan
| | - Yu Otagaki
- Department of Molecular and Internal Medicine, Graduate School of Biomedical and Health Sciences, Hiroshima University, Hiroshima, Japan
| | - Akira Okada
- Department of Molecular and Internal Medicine, Graduate School of Biomedical and Health Sciences, Hiroshima University, Hiroshima, Japan
| | - Yoko Yoshii
- Department of Molecular and Internal Medicine, Graduate School of Biomedical and Health Sciences, Hiroshima University, Hiroshima, Japan
| | - Gaku Nagano
- Department of Molecular and Internal Medicine, Graduate School of Biomedical and Health Sciences, Hiroshima University, Hiroshima, Japan
| | - Haruya Ohno
- Department of Molecular and Internal Medicine, Graduate School of Biomedical and Health Sciences, Hiroshima University, Hiroshima, Japan
| | - Koji Arihiro
- Department of Anatomical Pathology, Hiroshima University Hospital, Hiroshima, Japan
| | - Celso E Gomez-Sanchez
- Division of Endocrinology, G.V. (Sonny) Montgomery VA Medical Center and University of Mississippi Medical Center, Jackson, MS, USA
| | - Noboru Hattori
- Department of Molecular and Internal Medicine, Graduate School of Biomedical and Health Sciences, Hiroshima University, Hiroshima, Japan
| | - Masayasu Yoneda
- Department of Molecular and Internal Medicine, Graduate School of Biomedical and Health Sciences, Hiroshima University, Hiroshima, Japan
| |
Collapse
|
14
|
Iwahashi N, Umakoshi H, Ogata M, Fukumoto T, Kaneko H, Terada E, Katsuhara S, Uchida N, Sasaki K, Yokomoto-Umakoshi M, Matsuda Y, Sakamoto R, Ogawa Y. Whole Transcriptome Profiling of Adrenocortical Tumors Using Formalin-Fixed Paraffin-Embedded Samples. Front Endocrinol (Lausanne) 2022; 13:808331. [PMID: 35185794 PMCID: PMC8850780 DOI: 10.3389/fendo.2022.808331] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/03/2021] [Accepted: 01/11/2022] [Indexed: 11/24/2022] Open
Abstract
Whole transcriptome profiling is a promising technique in adrenal studies; however, whole transcriptome profiling of adrenal disease using formalin-fixed paraffin-embedded (FFPE) samples has to be further explored. The aim of this study was to evaluate the utility of transcriptome data from FFPE samples of adrenocortical tumors. We performed whole transcriptome profiling of FFPE and fresh frozen samples of adrenocortical carcinoma (ACC, n = 3), aldosterone-producing adenoma (APA, n = 3), and cortisol-producing adenoma (CPA, n = 3), and examined the similarity between the transcriptome data. We further examined whether the transcriptome data of FFPE samples could be used to distinguish tumor types and detect marker genes. The number of read counts was smaller in FFPE samples than in fresh frozen samples (P < 0.01), while the number of genes detected was similar (P = 0.39). The gene expression profiles of FFPE and fresh frozen samples were highly correlated (r = 0.93, P < 0.01). Tumor types could be distinguished by consensus clustering and principal component analysis using transcriptome data from FFPE samples. In the differential expression analysis between ACC and APA-CPA, known marker genes of ACC (e.g., CCNB2, TOP2A, and MAD2L1) were detected in FFPE samples of ACC. In the differential expression analysis between APA and CPA, known marker genes of APA (e.g., CYP11B2, VSNL1, and KCNJ5) were detected in the APA of FFPE samples. The results suggest that FFPE samples may be a reliable alternative to fresh frozen samples for whole transcriptome profiling of adrenocortical tumors.
Collapse
Affiliation(s)
- Norifusa Iwahashi
- Department of Medicine and Bioregulatory Science, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Hironobu Umakoshi
- Department of Medicine and Bioregulatory Science, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
- *Correspondence: Hironobu Umakoshi, ; Yoshihiro Ogawa,
| | - Masatoshi Ogata
- Department of Medicine and Bioregulatory Science, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Tazuru Fukumoto
- Department of Medicine and Bioregulatory Science, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Hiroki Kaneko
- Department of Medicine and Bioregulatory Science, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Eriko Terada
- Department of Medicine and Bioregulatory Science, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Shunsuke Katsuhara
- Department of Medicine and Bioregulatory Science, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Naohiro Uchida
- Department of Medicine and Bioregulatory Science, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Katsuhiko Sasaki
- Clinical Laboratory Department, Kyushu Pro Search Limited Liability Partnership, Fukuoka, Japan
| | - Maki Yokomoto-Umakoshi
- Department of Medicine and Bioregulatory Science, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Yayoi Matsuda
- Department of Medicine and Bioregulatory Science, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Ryuichi Sakamoto
- Department of Medicine and Bioregulatory Science, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Yoshihiro Ogawa
- Department of Medicine and Bioregulatory Science, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
- *Correspondence: Hironobu Umakoshi, ; Yoshihiro Ogawa,
| |
Collapse
|
15
|
Baba R, Oki K, Gomez-Sanchez CE, Otagaki Y, Itcho K, Kobuke K, Kodama T, Nagano G, Ohno H, Yoneda M, Hattori N. Genotype-specific cortisol production associated with Cushing's syndrome adenoma with PRKACA mutations. Mol Cell Endocrinol 2021; 538:111456. [PMID: 34520814 PMCID: PMC8551059 DOI: 10.1016/j.mce.2021.111456] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/21/2021] [Revised: 09/07/2021] [Accepted: 09/08/2021] [Indexed: 12/24/2022]
Abstract
The intracellular molecular mechanisms underlying the genotype of cortisol-producing adenoma (CPA) have not been fully determined. We analyzed gene expressions in CPA and the human adrenocortical cell line (HAC15 cells) with PRKACA mutation. Clustering analysis using a gene set associated with responses to cAMP revealed the possible differences between PRKACA mutant CPAs and GNAS and CTNNB1 mutant CPAs. The levels of STAR, CYP11A1, CYP17A1, CYP21A2, and FDX1 transcripts and cortisol levels per unit area in PRKACA mutant CPAs were significantly higher than those in GNAS mutant CPAs. PRKACA mutations led to an increase in steroidogenic enzyme expression and cortisol production in HAC15 cells. Transcriptome analysis revealed differences between PRKACA mutant CPAs and GNAS and CTNNB1 mutant CPAs. Cortisol production in PRKACA mutant CPAs is increased by the cAMP-PKA signaling pathway-mediated upregulation of steroidogenic enzymes transcription. The intracellular molecular mechanisms underlying these processes would be notably important in PRKACA mutant CPAs.
Collapse
Affiliation(s)
- Ryuta Baba
- Department of Molecular and Internal Medicine, Graduate School of Biomedical and Health Sciences, Hiroshima University, Hiroshima, Japan
| | - 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 University of Mississippi Medical Center, Jackson, MS, USA; Department of Pharmacology & Toxicology, University of Mississippi Medical Center, Jackson, MS, USA
| | - Yu Otagaki
- Department of Molecular and Internal Medicine, Graduate School of Biomedical and Health Sciences, Hiroshima University, Hiroshima, Japan
| | - Kiyotaka Itcho
- Department of Molecular and Internal Medicine, Graduate School of Biomedical and Health Sciences, Hiroshima University, Hiroshima, Japan
| | - Kazuhiro Kobuke
- Department of Molecular and Internal Medicine, Graduate School of Biomedical and Health Sciences, Hiroshima University, Hiroshima, Japan
| | - Takaya Kodama
- Department of Molecular and Internal Medicine, Graduate School of Biomedical and Health Sciences, Hiroshima University, Hiroshima, Japan
| | - Gaku Nagano
- Department of Molecular and Internal Medicine, Graduate School of Biomedical and Health Sciences, Hiroshima University, Hiroshima, Japan
| | - Haruya Ohno
- Department of Molecular and Internal Medicine, Graduate School of Biomedical and Health Sciences, Hiroshima University, Hiroshima, Japan
| | - Masayasu Yoneda
- Department of Molecular and Internal Medicine, Graduate School of Biomedical and Health Sciences, Hiroshima University, Hiroshima, Japan
| | - Noboru Hattori
- Department of Molecular and Internal Medicine, Graduate School of Biomedical and Health Sciences, Hiroshima University, Hiroshima, Japan
| |
Collapse
|
16
|
Transcriptomics, Epigenetics, and Metabolomics of Primary Aldosteronism. Cancers (Basel) 2021; 13:cancers13215582. [PMID: 34771744 PMCID: PMC8583505 DOI: 10.3390/cancers13215582] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2021] [Revised: 10/17/2021] [Accepted: 11/05/2021] [Indexed: 12/30/2022] Open
Abstract
INTRODUCTION Primary aldosteronism (PA) is the most common cause of endocrine hypertension, mainly caused by aldosterone-producing adenomas or hyperplasia; understanding its pathophysiological background is important in order to provide ameliorative treatment strategies. Over the past several years, significant progress has been documented in this field, in particular in the clarification of the genetic and molecular mechanisms responsible for the pathogenesis of aldosterone-producing adenomas (APAs). METHODS Systematic searches of the PubMed and Cochrane databases were performed for all human studies applying transcriptomic, epigenetic or metabolomic analyses to PA subjects. Studies involving serial analysis of gene expression and microarray, epigenetic studies with methylome analyses and micro-RNA expression profiles, and metabolomic studies focused on improving understanding of the regulation of autonomous aldosterone production in PA were all included. RESULTS In this review we summarize the main findings in this area and analyze the interplay between primary aldosteronism and several signaling pathways with differential regulation of the RNA and protein expression of several factors involved in, among others, steroidogenesis, calcium signaling, and nuclear, membrane and G-coupled protein receptors. Distinct transcriptomic and metabolomic patterns are also presented herein, depending on the mutational status of APAs. In particular, two partially opposite transcriptional and steroidogenic profiles appear to distinguish APAs carrying a KCNJ5 mutation from all other APAs, which carry different mutations. CONCLUSIONS These findings can substantially contribute to the development of personalized treatment in patients with PA.
Collapse
|
17
|
ATP1A1 Mutant in Aldosterone-Producing Adenoma Leads to Cell Proliferation. Int J Mol Sci 2021; 22:ijms222010981. [PMID: 34681640 PMCID: PMC8537586 DOI: 10.3390/ijms222010981] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2021] [Revised: 10/04/2021] [Accepted: 10/09/2021] [Indexed: 12/14/2022] Open
Abstract
The molecular mechanisms by which ATP1A1 mutation-mediated cell proliferation or tumorigenesis in aldosterone-producing adenomas (APAs) have not been elucidated. First, we investigated whether the APA-associated ATP1A1 L104R mutation stimulated cell proliferation. Second, we aimed to clarify the molecular mechanisms by which the ATP1A1 mutation-mediated cell proliferated. We performed transcriptome analysis in APAs with ATP1A1 mutation. ATP1A1 L104R mutation were modulated in human adrenocortical carcinoma (HAC15) cells (ATP1A1-mutant cells), and we evaluated cell proliferation and molecular signaling events. Transcriptome and immunohistochemical analysis showed that Na/K-ATPase (NKA) expressions in ATP1A1 mutated APA were more abundant than those in non-functioning adrenocortical adenoma or KCNJ5 mutated APAs. The significant increase of number of cells, amount of DNA and S-phase population were shown in ATP1A1-mutant cells. Fluo-4 in ATP1A1-mutant cells were significantly increased. Low concentration of ouabain stimulated cell proliferation in ATP1A1-mutant cells. ATP1A1-mutant cells induced Src phosphorylation, and low concentration of ouabain supplementation showed further Src phosphorylation. We demonstrated that NKAs were highly expressed in ATP1A1 mutant APA, and the mutant stimulated cell proliferation and Src phosphorylation in ATP1A1-mutant cells. NKA stimulations would be a risk factor for the progression and development to an ATP1A1 mutant APA.
Collapse
|
18
|
Shimada H, Yamazaki Y, Sugawara A, Sasano H, Nakamura Y. Molecular Mechanisms of Functional Adrenocortical Adenoma and Carcinoma: Genetic Characterization and Intracellular Signaling Pathway. Biomedicines 2021; 9:biomedicines9080892. [PMID: 34440096 PMCID: PMC8389593 DOI: 10.3390/biomedicines9080892] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2021] [Revised: 07/20/2021] [Accepted: 07/22/2021] [Indexed: 02/06/2023] Open
Abstract
The adrenal cortex produces steroid hormones as adrenocortical hormones in the body, secreting mineralocorticoids, glucocorticoids, and adrenal androgens, which are all considered essential for life. Adrenocortical tumors harbor divergent hormonal activity, frequently with steroid excess, and disrupt homeostasis of the body. Aldosterone-producing adenomas (APAs) cause primary aldosteronism (PA), and cortisol-producing adenomas (CPAs) are the primary cause of Cushing’s syndrome. In addition, adrenocortical carcinoma (ACC) is a highly malignant cancer harboring poor prognosis. Various genetic abnormalities have been reported, which are associated with possible pathogenesis by the alteration of intracellular signaling and activation of transcription factors. In particular, somatic mutations in APAs have been detected in genes encoding membrane proteins, especially ion channels, resulting in hypersecretion of aldosterone due to activation of intracellular calcium signaling. In addition, somatic mutations have been detected in those encoding cAMP-PKA signaling-related factors, resulting in hypersecretion of cortisol due to its driven status in CPAs. In ACC, mutations in tumor suppressor genes and Wnt-β-catenin signaling-related factors have been implicated in its pathogenesis. In this article, we review recent findings on the genetic characteristics and regulation of intracellular signaling and transcription factors in individual tumors.
Collapse
Affiliation(s)
- Hiroki Shimada
- Division of Pathology, Faculty of Medicine, Tohoku Medical and Pharmaceutical University, 1-15-1 Fukumuro, Miyagino-ku, Sendai 983-8536, Miyagi, Japan;
| | - Yuto Yamazaki
- Department of Pathology, Tohoku University Graduate School of Medicine, 2-1 Seiryo-machi, Aoba-ku, Sendai 980-8575, Miyagi, Japan; (Y.Y.); (H.S.)
| | - Akira Sugawara
- Department of Molecular Endocrinology, Tohoku University Graduate School of Medicine, 2-1 Seiryo-machi, Aoba-ku, Sendai 980-8575, Miyagi, Japan;
| | - Hironobu Sasano
- Department of Pathology, Tohoku University Graduate School of Medicine, 2-1 Seiryo-machi, Aoba-ku, Sendai 980-8575, Miyagi, Japan; (Y.Y.); (H.S.)
| | - Yasuhiro Nakamura
- Division of Pathology, Faculty of Medicine, Tohoku Medical and Pharmaceutical University, 1-15-1 Fukumuro, Miyagino-ku, Sendai 983-8536, Miyagi, Japan;
- Correspondence: ; Tel.: +81-22-290-8731
| |
Collapse
|
19
|
Update on Genetics of Primary Aldosteronism. Biomedicines 2021; 9:biomedicines9040409. [PMID: 33920271 PMCID: PMC8069207 DOI: 10.3390/biomedicines9040409] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2021] [Revised: 04/06/2021] [Accepted: 04/07/2021] [Indexed: 12/13/2022] Open
Abstract
Primary aldosteronism (PA) is the most common form of secondary hypertension, with a prevalence of 5–10% among patients with hypertension. PA is mainly classified into two subtypes: aldosterone-producing adenoma (APA) and bilateral idiopathic hyperaldosteronism. Recent developments in genetic analysis have facilitated the discovery of mutations in KCNJ5, ATP1A1, ATP2B3, CACNA1D, CACNA1H, CLCN2, and CTNNB1 in sporadic or familial forms of PA in the last decade. These findings have greatly advanced our understanding of the mechanism of excess aldosterone synthesis, particularly in APA. Most of the causative genes encode ion channels or pumps, and their mutations lead to depolarization of the cell membrane due to impairment of ion transport. Depolarization activates voltage-gated Ca2+ channels and intracellular calcium signaling and promotes the transcription of aldosterone synthase, resulting in overproduction of aldosterone. In this article, we review recent findings on the genetic and molecular mechanisms of PA.
Collapse
|
20
|
Pays E. The function of apolipoproteins L (APOLs): relevance for kidney disease, neurotransmission disorders, cancer and viral infection. FEBS J 2021; 288:360-381. [PMID: 32530132 PMCID: PMC7891394 DOI: 10.1111/febs.15444] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2020] [Revised: 05/24/2020] [Accepted: 06/03/2020] [Indexed: 12/17/2022]
Abstract
The discovery that apolipoprotein L1 (APOL1) is the trypanolytic factor of human serum raised interest about the function of APOLs, especially following the unexpected finding that in addition to their protective action against sleeping sickness, APOL1 C-terminal variants also cause kidney disease. Based on the analysis of the structure and trypanolytic activity of APOL1, it was proposed that APOLs could function as ion channels of intracellular membranes and be involved in mechanisms triggering programmed cell death. In this review, the recent finding that APOL1 and APOL3 inversely control the synthesis of phosphatidylinositol-4-phosphate (PI(4)P) by the Golgi PI(4)-kinase IIIB (PI4KB) is commented. APOL3 promotes Ca2+ -dependent activation of PI4KB, but due to their increased interaction with APOL3, APOL1 C-terminal variants can inactivate APOL3, leading to reduction of Golgi PI(4)P synthesis. The impact of APOLs on several pathological processes that depend on Golgi PI(4)P levels is discussed. I propose that through their effect on PI4KB activity, APOLs control not only actomyosin activities related to vesicular trafficking, but also the generation and elongation of autophagosomes induced by inflammation.
Collapse
Affiliation(s)
- Etienne Pays
- Laboratory of Molecular ParasitologyIBMMUniversité Libre de BruxellesGosseliesBelgium
| |
Collapse
|
21
|
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
|
22
|
Vohra T, Kemter E, Sun N, Dobenecker B, Hinrichs A, Burrello J, Gomez-Sanchez EP, Gomez-Sanchez CE, Wang J, Kinker IS, Teupser D, Fischer K, Schnieke A, Peitzsch M, Eisenhofer G, Walch A, Reincke M, Wolf E, Williams TA. Effect of Dietary Sodium Modulation on Pig Adrenal Steroidogenesis and Transcriptome Profiles. Hypertension 2020; 76:1769-1777. [PMID: 33070662 DOI: 10.1161/hypertensionaha.120.15998] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Primary aldosteronism is a frequent form of endocrine hypertension caused by aldosterone overproduction from the adrenal cortex. Regulation of aldosterone biosynthesis has been studied in rodents despite differences in adrenal physiology with humans. We, therefore, investigated pig adrenal steroidogenesis, morphology, and transcriptome profiles of the zona glomerulosa (zG) and zona fasciculata in response to activation of the renin-angiotensin-aldosterone system by dietary sodium restriction. Six-week-old pigs were fed a low- or high-sodium diet for 14 days (3 pigs per group, 0.4 g sodium/kg feed versus 6.8 g sodium/kg). Plasma aldosterone concentrations displayed a 43-fold increase (P=0.011) after 14 days of sodium restriction (day 14 versus day 0). Low dietary sodium caused a 2-fold increase in thickness of the zG (P<0.001) and an almost 3-fold upregulation of CYP11B (P<0.05) compared with high dietary sodium. Strong immunostaining of the KCNJ5 (G protein-activated inward rectifier potassium channel 4), which is frequently mutated in primary aldosteronism, was demonstrated in the zG. mRNA sequencing transcriptome analysis identified significantly altered expression of genes modulated by the renin-angiotensin-aldosterone system in the zG (n=1172) and zona fasciculata (n=280). These genes included many with a known role in the regulation of aldosterone synthesis and adrenal function. The most highly enriched biological pathways in the zG were related to cholesterol biosynthesis, steroid metabolism, cell cycle, and potassium channels. This study provides mechanistic insights into the physiology and pathophysiology of aldosterone production in a species closely related to humans and shows the suitability of pigs as a translational animal model for human adrenal steroidogenesis.
Collapse
Affiliation(s)
- Twinkle Vohra
- From the Medizinische Klinik und Poliklinik IV, Klinikum der Universität München (T.V., I.-S.K., M.R., T.A.W.), Ludwig-Maximilians-Universität München, Munich, Germany
| | - Elisabeth Kemter
- Chair for Molecular Animal Breeding and Biotechnology, Gene Center and Department of Veterinary Sciences (E.K., A.H., E.W.), Ludwig-Maximilians-Universität München, Munich, Germany
| | - Na Sun
- Research Unit Analytical Pathology, German Research Center for Environmental Health, Helmholtz Zentrum München, Neuherberg, Germany (N.S., J.W., A.W.)
| | - Britta Dobenecker
- Chair of Animal Nutrition and Dietetics, Department of Veterinary Sciences, Ludwig-Maximilians-Universität München, Oberschleißheim, Germany (B.D.)
| | - Arne Hinrichs
- Chair for Molecular Animal Breeding and Biotechnology, Gene Center and Department of Veterinary Sciences (E.K., A.H., E.W.), Ludwig-Maximilians-Universität München, Munich, Germany
| | - Jacopo Burrello
- Division of Internal Medicine and Hypertension, Department of Medical Sciences, University of Turin, Italy (J.B., T.A.W.)
| | - Elise P Gomez-Sanchez
- Department of Neurobiology and Anatomical Sciences, University of Mississippi Medical Center, Jackson (E.P.G.-S.)
| | - Celso E Gomez-Sanchez
- Endocrine Division, G.V. (Sonny) Montgomery VA Medical Center, Jackson, MS (C.E.G.-S.).,Department of Pharmacology and Toxicology and Medicine, University of Mississippi Medical Center, Jackson (C.E.G.-S.)
| | - Jun Wang
- Research Unit Analytical Pathology, German Research Center for Environmental Health, Helmholtz Zentrum München, Neuherberg, Germany (N.S., J.W., A.W.)
| | - Isabella-Sabrina Kinker
- From the Medizinische Klinik und Poliklinik IV, Klinikum der Universität München (T.V., I.-S.K., M.R., T.A.W.), Ludwig-Maximilians-Universität München, Munich, Germany
| | - Daniel Teupser
- Institute of Laboratory Medicine, University Hospital (D.T.), Ludwig-Maximilians-Universität München, Munich, Germany
| | - Konrad Fischer
- School of Life Sciences Weihenstephan, Technical University Munich, Freising, Germany (K.F., A.S.)
| | - Angelika Schnieke
- School of Life Sciences Weihenstephan, Technical University Munich, Freising, Germany (K.F., A.S.)
| | - Mirko Peitzsch
- Institute of Clinical Chemistry and Laboratory Medicine (M.P., G.E.), University Hospital Carl Gustav Carus, Technische Universität Dresden, Germany
| | - Graeme Eisenhofer
- Institute of Clinical Chemistry and Laboratory Medicine (M.P., G.E.), University Hospital Carl Gustav Carus, Technische Universität Dresden, Germany.,Department of Medicine III (G.E.), University Hospital Carl Gustav Carus, Technische Universität Dresden, Germany
| | - Axel Walch
- Research Unit Analytical Pathology, German Research Center for Environmental Health, Helmholtz Zentrum München, Neuherberg, Germany (N.S., J.W., A.W.)
| | - Martin Reincke
- From the Medizinische Klinik und Poliklinik IV, Klinikum der Universität München (T.V., I.-S.K., M.R., T.A.W.), Ludwig-Maximilians-Universität München, Munich, Germany
| | - Eckhard Wolf
- Chair for Molecular Animal Breeding and Biotechnology, Gene Center and Department of Veterinary Sciences (E.K., A.H., E.W.), Ludwig-Maximilians-Universität München, Munich, Germany
| | - Tracy Ann Williams
- From the Medizinische Klinik und Poliklinik IV, Klinikum der Universität München (T.V., I.-S.K., M.R., T.A.W.), Ludwig-Maximilians-Universität München, Munich, Germany.,Division of Internal Medicine and Hypertension, Department of Medical Sciences, University of Turin, Italy (J.B., T.A.W.)
| |
Collapse
|
23
|
Alipoor B, Parvar SN, Sabati Z, Ghaedi H, Ghasemi H. An updated review of the H19 lncRNA in human cancer: molecular mechanism and diagnostic and therapeutic importance. Mol Biol Rep 2020; 47:6357-6374. [PMID: 32743775 DOI: 10.1007/s11033-020-05695-x] [Citation(s) in RCA: 43] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2020] [Accepted: 07/26/2020] [Indexed: 12/24/2022]
Abstract
Accumulating evidence has reported that H19 long non-coding RNA (lncRNA) expression level is deregulated in human cancer. It has been also demonstrated that de-regulated levels of H19 could affect cancer biology by various mechanisms including microRNA (miRNA) production (like miR-675), miRNA sponging and epigenetic modifications. Furthermore, lncRNA could act as a potential diagnosis and prognosis biomarkers and also a candidate therapeutic approach for different human cancers. In this narrative review, we shed light on the molecular mechanism of H19 in cancer development and pathogenesis. Moreover, we discussed the expression pattern and diagnostic and therapeutic importance of H19 as a potential biomarker in a range of human malignancies from breast to osteosarcoma cancer.
Collapse
Affiliation(s)
- Behnam Alipoor
- Department of Laboratory Sciences, Faculty of Paramedicine, Yasuj University of Medical Sciences, Yasuj, Iran
| | - Seyedeh Nasrin Parvar
- Department of Biochemistry, Faculty of Medicine, Yasuj University of Medical Sciences, Yasuj, Iran
| | - Zolfaghar Sabati
- Student Research Committee, Abadan Faculty of Medical Sciences, Abadan, Iran
| | - Hamid Ghaedi
- Department of Medical Genetics, Faculty of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Hassan Ghasemi
- Department of Clinical Biochemistry, Abadan Faculty of Medical Sciences, Abadan, Iran.
| |
Collapse
|
24
|
Hearn JI, Green TN, Chopra M, Nursalim YNS, Ladvanszky L, Knowlton N, Blenkiron C, Poulsen RC, Singleton DC, Bohlander SK, Kalev-Zylinska ML. N-Methyl-D-Aspartate Receptor Hypofunction in Meg-01 Cells Reveals a Role for Intracellular Calcium Homeostasis in Balancing Megakaryocytic-Erythroid Differentiation. Thromb Haemost 2020; 120:671-686. [PMID: 32289863 PMCID: PMC7286128 DOI: 10.1055/s-0040-1708483] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023]
Abstract
The release of calcium ions (Ca
2+
) from the endoplasmic reticulum (ER) and related store-operated calcium entry (SOCE) regulate maturation of normal megakaryocytes. The
N
-methyl-D-aspartate (NMDA) receptor (NMDAR) provides an additional mechanism for Ca
2+
influx in megakaryocytic cells, but its role remains unclear. We created a model of NMDAR hypofunction in Meg-01 cells using CRISPR-Cas9 mediated knockout of the
GRIN1
gene, which encodes an obligate, GluN1 subunit of the NMDAR. We found that compared with unmodified Meg-01 cells, Meg-01-
GRIN1−/−
cells underwent atypical differentiation biased toward erythropoiesis, associated with increased basal ER stress and cell death. Resting cytoplasmic Ca
2+
levels were higher in Meg-01-
GRIN1−/−
cells, but ER Ca
2+
release and SOCE were lower after activation. Lysosome-related organelles accumulated including immature dense granules that may have contributed an alternative source of intracellular Ca
2+
. Microarray analysis revealed that Meg-01-
GRIN1−/−
cells had deregulated expression of transcripts involved in Ca
2+
metabolism, together with a shift in the pattern of hematopoietic transcription factors toward erythropoiesis. In keeping with the observed pro-cell death phenotype induced by
GRIN1
deletion, memantine (NMDAR inhibitor) increased cytotoxic effects of cytarabine in unmodified Meg-01 cells. In conclusion, NMDARs comprise an integral component of the Ca
2+
regulatory network in Meg-01 cells that help balance ER stress and megakaryocytic-erythroid differentiation. We also provide the first evidence that megakaryocytic NMDARs regulate biogenesis of lysosome-related organelles, including dense granules. Our results argue that intracellular Ca
2+
homeostasis may be more important for normal megakaryocytic and erythroid differentiation than currently recognized; thus, modulation may offer therapeutic opportunities.
Collapse
Affiliation(s)
- James I Hearn
- Department of Molecular Medicine and Pathology, School of Medical Sciences, University of Auckland, Auckland, New Zealand
| | - Taryn N Green
- Department of Molecular Medicine and Pathology, School of Medical Sciences, University of Auckland, Auckland, New Zealand
| | - Martin Chopra
- Department of Molecular Medicine and Pathology, School of Medical Sciences, University of Auckland, Auckland, New Zealand
| | - Yohanes N S Nursalim
- Department of Molecular Medicine and Pathology, School of Medical Sciences, University of Auckland, Auckland, New Zealand
| | - Leandro Ladvanszky
- Department of Molecular Medicine and Pathology, School of Medical Sciences, University of Auckland, Auckland, New Zealand
| | - Nicholas Knowlton
- Department of Molecular Medicine and Pathology, School of Medical Sciences, University of Auckland, Auckland, New Zealand
| | - Cherie Blenkiron
- Department of Molecular Medicine and Pathology, School of Medical Sciences, University of Auckland, Auckland, New Zealand
| | - Raewyn C Poulsen
- Department of Medicine, School of Medicine, University of Auckland, Auckland, New Zealand.,Department of Pharmacology and Clinical Pharmacology, School of Medical Sciences, University of Auckland, Auckland, New Zealand
| | - Dean C Singleton
- Auckland Cancer Society Research Centre, University of Auckland, Auckland, New Zealand
| | - Stefan K Bohlander
- Department of Molecular Medicine and Pathology, School of Medical Sciences, University of Auckland, Auckland, New Zealand
| | - Maggie L Kalev-Zylinska
- Department of Molecular Medicine and Pathology, School of Medical Sciences, University of Auckland, Auckland, New Zealand.,LabPlus Haematology, Auckland City Hospital, Auckland, New Zealand
| |
Collapse
|
25
|
Vesnina A, Prosekov A, Kozlova O, Atuchin V. Genes and Eating Preferences, Their Roles in Personalized Nutrition. Genes (Basel) 2020; 11:genes11040357. [PMID: 32230794 PMCID: PMC7230842 DOI: 10.3390/genes11040357] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2020] [Revised: 03/20/2020] [Accepted: 03/26/2020] [Indexed: 12/20/2022] Open
Abstract
At present, personalized diets, which take into account consumer genetic characteristics, are growing popular. Nutrigenetics studies the effect of gene variations on metabolism and nutrigenomics, which branches off further and investigates how nutrients and food compounds affect genes. This work deals with the mutations affecting the assimilation of metabolites, contributing to nutrigenetic studies. We searched for the genes responsible for eating preferences which allow for the tailoring of personalized diets. Presently, genetic nutrition is growing in demand, as it contributes to the prevention and/or rehabilitation of non-communicable diseases, both monogenic and polygenic. In this work, we showed single-nucleotide polymorphisms in genes-missense mutations that change the functions of coded proteins, resulting in a particular eating preferences or a disease. We studied the genes influencing food preferences-particularly those responsible for fats and carbohydrates absorption, food intolerance, metabolism of vitamins, taste sensations, oxidation of xenobiotics, eating preferences and food addiction. As a result, 34 genes were identified that affect eating preferences. Significant shortcomings were found in the methods/programs for developing personalized diets that are used today, and the weaknesses were revealed in the development of nutrigenetics (inconsistency of data on SNP genes, ignoring population genetics data, difficult information to understand consumer, etc.). Taking into account all the shortcomings, an approximate model was proposed in the review for selecting an appropriate personalized diet. In the future, it is planned to develop the proposed model for the compilation of individual diets.
Collapse
Affiliation(s)
- Anna Vesnina
- Department of Bionanotechnology, Kemerovo State University, 650043 Kemerovo, Russia; (A.V.); (O.K.)
| | - Alexander Prosekov
- Laboratory of Biocatalysis, Kemerovo State University, 650043 Kemerovo, Russia;
| | - Oksana Kozlova
- Department of Bionanotechnology, Kemerovo State University, 650043 Kemerovo, Russia; (A.V.); (O.K.)
| | - Victor Atuchin
- Laboratory of Optical Materials and Structures, Institute of Semiconductor Physics, 630090 Novosibirsk, Russia
- Laboratory of Semiconductor and Dielectric Materials, Novosibirsk State University, 630090 Novosibirsk, Russia
- Research and Development Department, Kemerovo State University, 650000 Kemerovo, Russia
- Correspondence: ; Tel.: +7-(383)-3308889
| |
Collapse
|
26
|
Uzureau S, Lecordier L, Uzureau P, Hennig D, Graversen JH, Homblé F, Mfutu PE, Oliveira Arcolino F, Ramos AR, La Rovere RM, Luyten T, Vermeersch M, Tebabi P, Dieu M, Cuypers B, Deborggraeve S, Rabant M, Legendre C, Moestrup SK, Levtchenko E, Bultynck G, Erneux C, Pérez-Morga D, Pays E. APOL1 C-Terminal Variants May Trigger Kidney Disease through Interference with APOL3 Control of Actomyosin. Cell Rep 2020; 30:3821-3836.e13. [PMID: 32187552 PMCID: PMC7090385 DOI: 10.1016/j.celrep.2020.02.064] [Citation(s) in RCA: 49] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2019] [Revised: 01/17/2020] [Accepted: 02/14/2020] [Indexed: 11/18/2022] Open
Abstract
The C-terminal variants G1 and G2 of apolipoprotein L1 (APOL1) confer human resistance to the sleeping sickness parasite Trypanosoma rhodesiense, but they also increase the risk of kidney disease. APOL1 and APOL3 are death-promoting proteins that are partially associated with the endoplasmic reticulum and Golgi membranes. We report that in podocytes, either APOL1 C-terminal helix truncation (APOL1Δ) or APOL3 deletion (APOL3KO) induces similar actomyosin reorganization linked to the inhibition of phosphatidylinositol-4-phosphate [PI(4)P] synthesis by the Golgi PI(4)-kinase IIIB (PI4KB). Both APOL1 and APOL3 can form K+ channels, but only APOL3 exhibits Ca2+-dependent binding of high affinity to neuronal calcium sensor-1 (NCS-1), promoting NCS-1-PI4KB interaction and stimulating PI4KB activity. Alteration of the APOL1 C-terminal helix triggers APOL1 unfolding and increased binding to APOL3, affecting APOL3-NCS-1 interaction. Since the podocytes of G1 and G2 patients exhibit an APOL1Δ or APOL3KO-like phenotype, APOL1 C-terminal variants may induce kidney disease by preventing APOL3 from activating PI4KB, with consecutive actomyosin reorganization of podocytes.
Collapse
Affiliation(s)
- Sophie Uzureau
- Laboratory of Molecular Parasitology, IBMM, Université Libre de Bruxelles, 6041 Gosselies, Belgium
| | - Laurence Lecordier
- Laboratory of Molecular Parasitology, IBMM, Université Libre de Bruxelles, 6041 Gosselies, Belgium
| | - Pierrick Uzureau
- Laboratory of Experimental Medicine (ULB222), CHU Charleroi, Université Libre de Bruxelles, Montigny le Tilleul, Belgium
| | - Dorle Hennig
- Department of Molecular Medicine, Cancer and Inflammation Research, University of Southern Denmark, 5000 Odense C, Denmark
| | - Jonas H Graversen
- Department of Molecular Medicine, Cancer and Inflammation Research, University of Southern Denmark, 5000 Odense C, Denmark
| | - Fabrice Homblé
- Laboratory of Structure and Function of Biological Membranes, Université Libre de Bruxelles, 1050 Brussels, Belgium
| | - Pepe Ekulu Mfutu
- Pediatric Nephrology, University Hospital Leuven, 3000 Leuven, Belgium
| | | | - Ana Raquel Ramos
- Institute of Interdisciplinary Research in Human and Molecular Biology, Campus Erasme, Université Libre de Bruxelles, 1070 Brussels, Belgium
| | - Rita M La Rovere
- Laboratory of Molecular and Cellular Signalling, KU Leuven, Herestraat 49, 3000 Leuven, Belgium
| | - Tomas Luyten
- Laboratory of Molecular and Cellular Signalling, KU Leuven, Herestraat 49, 3000 Leuven, Belgium
| | - Marjorie Vermeersch
- Center for Microscopy and Molecular Imaging (CMMI), Université Libre de Bruxelles, 6041 Gosselies, Belgium
| | - Patricia Tebabi
- Laboratory of Molecular Parasitology, IBMM, Université Libre de Bruxelles, 6041 Gosselies, Belgium
| | - Marc Dieu
- URBC-Narilis, University of Namur, 5000 Namur, Belgium
| | - Bart Cuypers
- Biomedical Sciences Department, Institute of Tropical Medicine, 2000 Antwerpen, Belgium; Adrem Data Lab, Department of Mathematics and Computer Science, University of Antwerp, 2000 Antwerpen, Belgium
| | - Stijn Deborggraeve
- Biomedical Sciences Department, Institute of Tropical Medicine, 2000 Antwerpen, Belgium
| | - Marion Rabant
- Adult Nephrology-Transplantation Department, Paris Hospitals and Paris Descartes University, 75006 Paris, France
| | - Christophe Legendre
- Pathology Department, Paris Hospitals and Paris Descartes University, 75006 Paris, France
| | - Søren K Moestrup
- Department of Molecular Medicine, Cancer and Inflammation Research, University of Southern Denmark, 5000 Odense C, Denmark; Department of Biomedicine, University of Aarhus, 8000 Aarhus, Denmark
| | - Elena Levtchenko
- Pediatric Nephrology, University Hospital Leuven, 3000 Leuven, Belgium
| | - Geert Bultynck
- Laboratory of Molecular and Cellular Signalling, KU Leuven, Herestraat 49, 3000 Leuven, Belgium
| | - Christophe Erneux
- Institute of Interdisciplinary Research in Human and Molecular Biology, Campus Erasme, Université Libre de Bruxelles, 1070 Brussels, Belgium
| | - David Pérez-Morga
- Laboratory of Molecular Parasitology, IBMM, Université Libre de Bruxelles, 6041 Gosselies, Belgium; Center for Microscopy and Molecular Imaging (CMMI), Université Libre de Bruxelles, 6041 Gosselies, Belgium
| | - Etienne Pays
- Laboratory of Molecular Parasitology, IBMM, Université Libre de Bruxelles, 6041 Gosselies, Belgium.
| |
Collapse
|
27
|
Itcho K, Oki K, Gomez-Sanchez CE, Gomez-Sanchez EP, Ohno H, Kobuke K, Nagano G, Yoshii Y, Baba R, Hattori N, Yoneda M. Endoplasmic Reticulum Chaperone Calmegin Is Upregulated in Aldosterone-Producing Adenoma and Associates With Aldosterone Production. Hypertension 2019; 75:492-499. [PMID: 31865789 DOI: 10.1161/hypertensionaha.119.14062] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
The endoplasmic reticulum (ER) plays a pivotal role in syntheses of proteins and steroid hormones and regulation of intracellular Ca2+ level. We aimed to investigate ER-associated genes in aldosterone-producing adenomas (APAs) and clarify their effect on aldosterone production. Microarray analysis targeting 288 ER-associated genes was conducted using nonfunctioning adrenocortical adenomas (n=5) and APAs (n=19). Immunohistochemistry and quantitative polymerase chain reaction analyses were performed with 13 nonfunctioning adrenocortical adenoma and 48 APA samples. Functional studies were performed with human adrenocortical carcinoma (HAC15) cells, some of which were genetically modified using lentiviruses. The ER chaperone calmegin (CLGN) was the most highly expressed ER-associated gene in APAs relative to nonfunctioning adrenocortical adenomas. Analysis with quantitative polymerase chain reaction revealed CLGN to be 9.5-fold upregulated in APAs relative to nonfunctioning adrenocortical adenomas. There were no differences among different APA genotypes affecting aldosterone production. Immunohistochemistry analysis revealed that CLGN was strongly expressed in APAs and aldosterone-producing cell clusters. Angiotensin II stimulation or KCNJ5 T158A overexpression in HAC15 cells did not affect CLGN mRNA levels. CLGN overexpression in HAC15 cells increased aldosterone levels but did not stimulate CYP11B2 mRNA levels. Pathway and gene ontology analyses using RNA sequencing results showed that tRNA aminoacyl metabolism was the most enriched pathway in CLGN-overexpressing cells. CYP11B2 (aldosterone synthase) and HSD3B2 (3 beta-hydroxysteroid dehydrogenase/delta 5->4-isomerase type 2) protein expression were more abundant in CLGN-overexpressing cells. CLGN knockdown using CRISPR/Cas9 (clustered regularly interspaced short palindromic repeats/clustered regularly interspaced short palindromic repeat-associated 9) method in HAC15 cells that carry the KCNJ5 mutation did not affect aldosterone production. To summarize, CLGN was upregulated and associated with aldosterone production via translational regulation of CYP11B2 in APAs.
Collapse
Affiliation(s)
- Kiyotaka Itcho
- From the Department of Molecular and Internal Medicine, Graduate School of Biomedical and Health Sciences, Hiroshima University, Japan (K.I., K.O., H.O., K.K., G.N., Y.Y., R.B., N.H., M.Y.)
| | - Kenji Oki
- From the Department of Molecular and Internal Medicine, Graduate School of Biomedical and Health Sciences, Hiroshima University, Japan (K.I., K.O., H.O., K.K., G.N., Y.Y., R.B., N.H., M.Y.)
| | - 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 (C.E.G.-S., E.P.G.-S.)
| | - Elise P Gomez-Sanchez
- Division of Endocrinology, G.V. (Sonny) Montgomery VA Medical Center and Department of Pharmacology and Toxicology, University of Mississippi Medical Center, Jackson (C.E.G.-S., E.P.G.-S.)
| | - Haruya Ohno
- From the Department of Molecular and Internal Medicine, Graduate School of Biomedical and Health Sciences, Hiroshima University, Japan (K.I., K.O., H.O., K.K., G.N., Y.Y., R.B., N.H., M.Y.)
| | - Kazuhiro Kobuke
- From the Department of Molecular and Internal Medicine, Graduate School of Biomedical and Health Sciences, Hiroshima University, Japan (K.I., K.O., H.O., K.K., G.N., Y.Y., R.B., N.H., M.Y.)
| | - Gaku Nagano
- From the Department of Molecular and Internal Medicine, Graduate School of Biomedical and Health Sciences, Hiroshima University, Japan (K.I., K.O., H.O., K.K., G.N., Y.Y., R.B., N.H., M.Y.)
| | - Yoko Yoshii
- From the Department of Molecular and Internal Medicine, Graduate School of Biomedical and Health Sciences, Hiroshima University, Japan (K.I., K.O., H.O., K.K., G.N., Y.Y., R.B., N.H., M.Y.)
| | - Ryuta Baba
- From the Department of Molecular and Internal Medicine, Graduate School of Biomedical and Health Sciences, Hiroshima University, Japan (K.I., K.O., H.O., K.K., G.N., Y.Y., R.B., N.H., M.Y.)
| | - Noboru Hattori
- From the Department of Molecular and Internal Medicine, Graduate School of Biomedical and Health Sciences, Hiroshima University, Japan (K.I., K.O., H.O., K.K., G.N., Y.Y., R.B., N.H., M.Y.)
| | - Masayasu Yoneda
- From the Department of Molecular and Internal Medicine, Graduate School of Biomedical and Health Sciences, Hiroshima University, Japan (K.I., K.O., H.O., K.K., G.N., Y.Y., R.B., N.H., M.Y.)
| |
Collapse
|
28
|
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
|
29
|
Jouinot A, Armignacco R, Assié G. Genomics of benign adrenocortical tumors. J Steroid Biochem Mol Biol 2019; 193:105414. [PMID: 31207362 DOI: 10.1016/j.jsbmb.2019.105414] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/01/2018] [Revised: 04/25/2019] [Accepted: 06/13/2019] [Indexed: 12/14/2022]
Abstract
Benign adrenocortical adenomas and hyperplasia are relatively common and include a spectrum of distinct entities, which diagnosis depends on the macroscopic aspect and the secretion profile. Recent advances in genomics have proposed high-throughput molecular characterization of adrenal tumors, thereby improving our knowledge on the pathophysiology and tumorigenesis of these tumors. Genomic (exome and chromosome alteration profiles), epigenomic (micro-RNAs expression and methylation profiles) and transcriptomic (gene expression profiles) studies highlighted the major roles of intracellular calcium signaling in aldosterone-producing adenomas (APA), of protein kinase A (PKA)/cAMP pathway in cortisol-producing tumors, and of Wnt/beta-catenin pathway in non-secreting tumors. Exome sequencing revealed new major drivers in all tumor types, including KCNJ5, ATP1A1, ATP2B3, CACNA1D and CACNA1H mutations in APA, PRKACA mutations in cortisol-producing adenomas (CPA) and ARMC5 mutations in primary macronodular adrenocortical hyperplasia (PMAH). The clinical impact of these findings is just starting to evolve. The identification of genetic syndromes, such as germline ARMC5 mutations in PMAH, has allowed genetic counseling. Key molecular alterations could serve as a basis for the development of targeted medical treatments for benign adrenal tumors. The recent developments in genomics, including single-cell technologies, and in proteomics and metabolomics will probably offer new perspectives for characterizing benign adrenal tumorigenesis.
Collapse
Affiliation(s)
- Anne Jouinot
- Institut Cochin, INSERM U1016, CNRS UMR 8104, Paris Descartes University, Paris, France; Department of Endocrinology, Referral Center for Rare Adrenal Diseases, Hôpital Cochin, Paris, France
| | - Roberta Armignacco
- Institut Cochin, INSERM U1016, CNRS UMR 8104, Paris Descartes University, Paris, France
| | - Guillaume Assié
- Institut Cochin, INSERM U1016, CNRS UMR 8104, Paris Descartes University, Paris, France; Department of Endocrinology, Referral Center for Rare Adrenal Diseases, Hôpital Cochin, Paris, France.
| |
Collapse
|
30
|
Yu Q, Tian X, Sun C, Shao L, Li X, Dai R. Comparative transcriptomics to reveal muscle-specific molecular differences in the early postmortem of Chinese Jinjiang yellow cattle. Food Chem 2019; 301:125262. [PMID: 31377625 DOI: 10.1016/j.foodchem.2019.125262] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2019] [Revised: 07/05/2019] [Accepted: 07/25/2019] [Indexed: 01/09/2023]
Abstract
Differences in the expression of functional genes between beef Longissimus Lumborum (LL) and Psoas Major (PM) are not well understood. The aim of present study is to reveal transcriptome changes of beef LL and PM during early postmortem by high-throughput Illumina Hiseq4000 Sequencing. Hierarchical clustering analysis indicated significant differences in transcriptome profiles between LL and PM as well as 1 h and 12 h postmortem. A total of 65 genes differentially expressed between LL and PM (fold change ≥3, and p < 0.05; 34 were up-regulated in LL and 31 in PM), and the majority of them (53 genes) occurred at 12 h postmortem. These differentially expressed genes mainly involved in energy production and conversion, nucleotide metabolic, posttranslational modification, and transcription. KEGG analysis revealed that oxidative phosphorylation was one of the important pathways. This study gave new perspectives to understand the underlying mechanisms associated with muscle-specific beef quality.
Collapse
Affiliation(s)
- Qianqian Yu
- College of Food Science and Nutritional Engineering, China Agricultural University, No. 17 Qinghua East Road, Haidian District, Beijing 100083, PR China; Beijing Higher Institution Engineering Research Center of Animal Product, China Agricultural University, No. 17 Qinghua East Road, Haidian District, Beijing 100083, PR China
| | - Xiaojing Tian
- College of Food Science and Nutritional Engineering, China Agricultural University, No. 17 Qinghua East Road, Haidian District, Beijing 100083, PR China; Beijing Higher Institution Engineering Research Center of Animal Product, China Agricultural University, No. 17 Qinghua East Road, Haidian District, Beijing 100083, PR China
| | - Chengfeng Sun
- College of Life Science, Yantai University, No. 30 Qingquan Road, Laishan District, Yantai, Shandong Province, PR China
| | - Lele Shao
- College of Food Science and Nutritional Engineering, China Agricultural University, No. 17 Qinghua East Road, Haidian District, Beijing 100083, PR China; Beijing Higher Institution Engineering Research Center of Animal Product, China Agricultural University, No. 17 Qinghua East Road, Haidian District, Beijing 100083, PR China
| | - Xingmin Li
- College of Food Science and Nutritional Engineering, China Agricultural University, No. 17 Qinghua East Road, Haidian District, Beijing 100083, PR China; Beijing Higher Institution Engineering Research Center of Animal Product, China Agricultural University, No. 17 Qinghua East Road, Haidian District, Beijing 100083, PR China
| | - Ruitong Dai
- College of Food Science and Nutritional Engineering, China Agricultural University, No. 17 Qinghua East Road, Haidian District, Beijing 100083, PR China; Beijing Higher Institution Engineering Research Center of Animal Product, China Agricultural University, No. 17 Qinghua East Road, Haidian District, Beijing 100083, PR China.
| |
Collapse
|
31
|
Rendic SP, Peter Guengerich F. Human cytochrome P450 enzymes 5-51 as targets of drugs and natural and environmental compounds: mechanisms, induction, and inhibition - toxic effects and benefits. Drug Metab Rev 2019; 50:256-342. [PMID: 30717606 DOI: 10.1080/03602532.2018.1483401] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Cytochrome P450 (P450, CYP) enzymes have long been of interest due to their roles in the metabolism of drugs, pesticides, pro-carcinogens, and other xenobiotic chemicals. They have also been of interest due to their very critical roles in the biosynthesis and metabolism of steroids, vitamins, and certain eicosanoids. This review covers the 22 (of the total of 57) human P450s in Families 5-51 and their substrate selectivity. Furthermore, included is information and references regarding inducibility, inhibition, and (in some cases) stimulation by chemicals. We update and discuss important aspects of each of these 22 P450s and questions that remain open.
Collapse
Affiliation(s)
| | - F Peter Guengerich
- b Department of Biochemistry , Vanderbilt University School of Medicine , Nashville , TN , USA
| |
Collapse
|
32
|
Burgoyne RD, Helassa N, McCue HV, Haynes LP. Calcium Sensors in Neuronal Function and Dysfunction. Cold Spring Harb Perspect Biol 2019; 11:cshperspect.a035154. [PMID: 30833454 DOI: 10.1101/cshperspect.a035154] [Citation(s) in RCA: 55] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Calcium signaling in neurons as in other cell types can lead to varied changes in cellular function. Neuronal Ca2+ signaling processes have also become adapted to modulate the function of specific pathways over a wide variety of time domains and these can have effects on, for example, axon outgrowth, neuronal survival, and changes in synaptic strength. Ca2+ also plays a key role in synapses as the trigger for fast neurotransmitter release. Given its physiological importance, abnormalities in neuronal Ca2+ signaling potentially underlie many different neurological and neurodegenerative diseases. The mechanisms by which changes in intracellular Ca2+ concentration in neurons can bring about diverse responses is underpinned by the roles of ubiquitous or specialized neuronal Ca2+ sensors. It has been established that synaptotagmins have key functions in neurotransmitter release, and, in addition to calmodulin, other families of EF-hand-containing neuronal Ca2+ sensors, including the neuronal calcium sensor (NCS) and the calcium-binding protein (CaBP) families, play important physiological roles in neuronal Ca2+ signaling. It has become increasingly apparent that these various Ca2+ sensors may also be crucial for aspects of neuronal dysfunction and disease either indirectly or directly as a direct consequence of genetic variation or mutations. An understanding of the molecular basis for the regulation of the targets of the Ca2+ sensors and the physiological roles of each protein in identified neurons may contribute to future approaches to the development of treatments for a variety of human neuronal disorders.
Collapse
Affiliation(s)
- Robert D Burgoyne
- Department of Cellular and Molecular Physiology, Institute of Translational Medicine, University of Liverpool, Liverpool, United Kingdom
| | - Nordine Helassa
- Department of Cellular and Molecular Physiology, Institute of Translational Medicine, University of Liverpool, Liverpool, United Kingdom
| | - Hannah V McCue
- Centre for Genomic Research, University of Liverpool, Liverpool, United Kingdom
| | - Lee P Haynes
- Department of Cellular and Molecular Physiology, Institute of Translational Medicine, University of Liverpool, Liverpool, United Kingdom
| |
Collapse
|
33
|
Mundhenk J, Fusi C, Kreutz MR. Caldendrin and Calneurons-EF-Hand CaM-Like Calcium Sensors With Unique Features and Specialized Neuronal Functions. Front Mol Neurosci 2019; 12:16. [PMID: 30787867 PMCID: PMC6372560 DOI: 10.3389/fnmol.2019.00016] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2018] [Accepted: 01/17/2019] [Indexed: 01/02/2023] Open
Abstract
The calmodulin (CaM)-like Ca2+-sensor proteins caldendrin, calneuron-1 and -2 are members of the neuronal calcium-binding protein (nCaBP)-family, a family that evolved relatively late during vertebrate evolution. All three proteins are abundant in brain but show a strikingly different subcellular localization. Whereas caldendrin is enriched in the postsynaptic density (PSD), calneuron-1 and -2 accumulate at the trans-Golgi-network (TGN). Caldendrin exhibit a unique bipartite structure with a basic and proline-rich N-terminus while calneurons are the only EF-Hand CaM-like transmembrane proteins. These uncommon structural features come along with highly specialized functions of calneurons in Golgi-to-plasma-membrane trafficking and for caldendrin in actin-remodeling in dendritic spine synapses. In this review article, we will provide a synthesis of available data on the structure and biophysical properties of all three proteins. We will then discuss their cellular function with special emphasis on synaptic neurotransmission. Finally, we will summarize the evidence for a role of these proteins in neuropsychiatric disorders.
Collapse
Affiliation(s)
- Jennifer Mundhenk
- RG Neuroplasticity, Leibniz Institute for Neurobiology, Magdeburg, Germany
| | - Camilla Fusi
- RG Neuroplasticity, Leibniz Institute for Neurobiology, Magdeburg, Germany
| | - Michael R Kreutz
- RG Neuroplasticity, Leibniz Institute for Neurobiology, Magdeburg, Germany.,Leibniz Group "Dendritic Organelles and Synaptic Function", Center for Molecular Neurobiology, ZMNH, Hamburg, Germany
| |
Collapse
|
34
|
Itcho K, Oki K, Kobuke K, Ohno H, Yoneda M, Hattori N. Angiotensin 1-7 suppresses angiotensin II mediated aldosterone production via JAK/STAT signaling inhibition. J Steroid Biochem Mol Biol 2019; 185:137-141. [PMID: 30125658 DOI: 10.1016/j.jsbmb.2018.08.007] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/26/2018] [Revised: 08/03/2018] [Accepted: 08/12/2018] [Indexed: 01/27/2023]
Abstract
Angiotensin 1-7 (Ang 1-7), which is a protein cleaved from angiotensin II (A-II), binds to the MAS receptor. Ang 1-7 has been demonstrated to exert protective effects against A-II-mediated cardiac, atherosclerotic, and renal damages. The aims of our study were to demonstrate the inhibitory role of Ang 1-7 in A-II-mediated aldosterone production by interacting with the MAS receptor in human adrenocortical carcinoma (HAC15) cells, and clarify the intracellular signaling mechanisms underlying the inhibition of aldosterone production by Ang 1-7. Ang 1-7 significantly suppressed A-II-stimulated aldosterone production, and partially abrogated A-II-induced upregulation of CYP11B2 expression. Treatment with a selective Ang 1-7 antagonist abrogated Ang 1-7-mediated inhibition of aldosterone production in HAC15 cells. Incubation of A-II-treated HAC15 cells with conditioned medium containing Ang 1-7 was demonstrated to suppress A-II-mediated aldosterone production and CYP11B2 expression. Proteomic analysis showed that Ang 1-7 predominantly inhibited the phosphorylation of JAK-STAT proteins in A-II stimulated HAC15 cells. Treatment of HAC15 cells with a STAT3 inhibitor partially but significantly repressed A-II-mediated aldosterone production by 63.2%. Similarly, treatment with a STAT5 inhibitor significantly abrogated A-II-stimulated aldosterone production in HAC15 cells by 60.7%. In conclusion, we demonstrated that Ang 1-7 negatively regulates A-II-mediated aldosterone production, and the observed inhibition of aldosterone production was associated with JAK/STAT signaling in human adrenal cells. Therefore, activation of Ang 1-7 or stimulation of the MAS receptor, which inhibits aldosterone production, is a promising therapeutic approach for the prevention of cardiovascular events that can directly affect the target organs.
Collapse
Affiliation(s)
- Kiyotaka Itcho
- Department of Molecular and Internal Medicine, Graduate School of Biomedical and Health Sciences, Hiroshima University, 1-2-3 Kasumi, Minami-ku, Hiroshima, 734-8551, Japan
| | - Kenji Oki
- Department of Molecular and Internal Medicine, Graduate School of Biomedical and Health Sciences, Hiroshima University, 1-2-3 Kasumi, Minami-ku, Hiroshima, 734-8551, Japan.
| | - Kazuhiro Kobuke
- Department of Molecular and Internal Medicine, Graduate School of Biomedical and Health Sciences, Hiroshima University, 1-2-3 Kasumi, Minami-ku, Hiroshima, 734-8551, Japan
| | - Haruya Ohno
- Department of Molecular and Internal Medicine, Graduate School of Biomedical and Health Sciences, Hiroshima University, 1-2-3 Kasumi, Minami-ku, Hiroshima, 734-8551, Japan
| | - Masayasu Yoneda
- Department of Molecular and Internal Medicine, Graduate School of Biomedical and Health Sciences, Hiroshima University, 1-2-3 Kasumi, Minami-ku, Hiroshima, 734-8551, Japan
| | - Noboru Hattori
- Department of Molecular and Internal Medicine, Graduate School of Biomedical and Health Sciences, Hiroshima University, 1-2-3 Kasumi, Minami-ku, Hiroshima, 734-8551, Japan
| |
Collapse
|
35
|
Seccia TM, Caroccia B, Gomez-Sanchez EP, Gomez-Sanchez CE, Rossi GP. The Biology of Normal Zona Glomerulosa and Aldosterone-Producing Adenoma: Pathological Implications. Endocr Rev 2018; 39:1029-1056. [PMID: 30007283 PMCID: PMC6236434 DOI: 10.1210/er.2018-00060] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/18/2018] [Accepted: 07/03/2018] [Indexed: 01/09/2023]
Abstract
The identification of several germline and somatic ion channel mutations in aldosterone-producing adenomas (APAs) and detection of cell clusters that can be responsible for excess aldosterone production, as well as the isolation of autoantibodies activating the angiotensin II type 1 receptor, have rapidly advanced the understanding of the biology of primary aldosteronism (PA), particularly that of APA. Hence, the main purpose of this review is to discuss how discoveries of the last decade could affect histopathology analysis and clinical practice. The structural remodeling through development and aging of the human adrenal cortex, particularly of the zona glomerulosa, and the complex regulation of aldosterone, with emphasis on the concepts of zonation and channelopathies, will be addressed. Finally, the diagnostic workup for PA and its subtyping to optimize treatment are reviewed.
Collapse
Affiliation(s)
- Teresa M Seccia
- Department of Medicine-DIMED, University of Padua, Padua PD, Italy
| | | | - Elise P Gomez-Sanchez
- Department of Pharmacology and Toxicology, G.V. (Sonny) Montgomery VA Medical Center, Jackson, Mississippi
| | - Celso E Gomez-Sanchez
- Division of Endocrinology, G.V. (Sonny) Montgomery VA Medical Center, Jackson, Mississippi.,University of Mississippi Medical Center, Jackson, Mississippi
| | - Gian Paolo Rossi
- Department of Medicine-DIMED, University of Padua, Padua PD, Italy
| |
Collapse
|
36
|
Crona J, Beuschlein F, Pacak K, Skogseid B. Advances in adrenal tumors 2018. Endocr Relat Cancer 2018; 25:R405-R420. [PMID: 29794126 PMCID: PMC5976083 DOI: 10.1530/erc-18-0138] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/19/2018] [Accepted: 04/24/2018] [Indexed: 12/14/2022]
Abstract
This review aims to provide clinicians and researchers with a condensed update on the most important studies in the field during 2017. We present the academic output measured by active clinical trials and peer-reviewed published manuscripts. The most important and contributory manuscripts were summarized for each diagnostic entity, with a particular focus on manuscripts that describe translational research that have the potential to improve clinical care. Finally, we highlight the importance of collaborations in adrenal tumor research, which allowed for these recent advances and provide structures for future success in this scientific field.
Collapse
Affiliation(s)
- J Crona
- Department of Medical SciencesUppsala University, Uppsala, Sweden
| | - F Beuschlein
- Medizinische Klinik und Poliklinik IVKlinikum der Universität München, Munich, Germany
- Klinik für EndokrinologieDiabetologie und Klinische Ernährung, UniversitätsSpital Zürich, Zürich, Switzerland
| | - K Pacak
- Section on Medical NeuroendocrinologyEunice Kennedy Shriver National Institute of Child Health & Human Development, National Institutes of Health, Bethesda, Maryland, USA
| | - B Skogseid
- Department of Medical SciencesUppsala University, Uppsala, Sweden
| |
Collapse
|
37
|
Aristizabal Prada ET, Castellano I, Sušnik E, Yang Y, Meyer LS, Tetti M, Beuschlein F, Reincke M, Williams TA. Comparative Genomics and Transcriptome Profiling in Primary Aldosteronism. Int J Mol Sci 2018; 19:ijms19041124. [PMID: 29642543 PMCID: PMC5979346 DOI: 10.3390/ijms19041124] [Citation(s) in RCA: 9] [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: 02/15/2018] [Revised: 04/04/2018] [Accepted: 04/06/2018] [Indexed: 12/19/2022] Open
Abstract
Primary aldosteronism is the most common form of endocrine hypertension with a prevalence of 6% in the general population with hypertension. The genetic basis of the four familial forms of primary aldosteronism (familial hyperaldosteronism FH types I–IV) and the majority of sporadic unilateral aldosterone-producing adenomas has now been resolved. Familial forms of hyperaldosteronism are, however, rare. The sporadic forms of the disease prevail and these are usually caused by either a unilateral aldosterone-producing adenoma or bilateral adrenal hyperplasia. Aldosterone-producing adenomas frequently carry a causative somatic mutation in either of a number of genes with the KCNJ5 gene, encoding an inwardly rectifying potassium channel, a recurrent target harboring mutations at a prevalence of more than 40% worldwide. Other than genetic variations, gene expression profiling of aldosterone-producing adenomas has shed light on the genes and intracellular signalling pathways that may play a role in the pathogenesis and pathophysiology of these tumors.
Collapse
Affiliation(s)
- Elke Tatjana Aristizabal Prada
- Medizinische Klinik und Poliklinik IV, Klinikum der Universität, Ludwig-Maximilians-Universität München, 80336 Munich, Germany.
| | - Isabella Castellano
- Division of Pathology, Department of Medical Sciences, University of Torino, 10124 Torino, Italy.
| | - Eva Sušnik
- Medizinische Klinik und Poliklinik IV, Klinikum der Universität, Ludwig-Maximilians-Universität München, 80336 Munich, Germany.
| | - Yuhong Yang
- Medizinische Klinik und Poliklinik IV, Klinikum der Universität, Ludwig-Maximilians-Universität München, 80336 Munich, Germany.
| | - Lucie S Meyer
- Medizinische Klinik und Poliklinik IV, Klinikum der Universität, Ludwig-Maximilians-Universität München, 80336 Munich, Germany.
| | - Martina Tetti
- Division of Internal Medicine and Hypertension, Department of Medical Sciences, University of Torino, 10126 Torino, Italy.
| | - Felix Beuschlein
- Medizinische Klinik und Poliklinik IV, Klinikum der Universität, Ludwig-Maximilians-Universität München, 80336 Munich, Germany.
- Klinik für Endokrinologie, Diabetologie und Klinische Ernährung, UniversitätsSpital Zürich, CH-8091 Zurich, Switzerland.
| | - Martin Reincke
- Medizinische Klinik und Poliklinik IV, Klinikum der Universität, Ludwig-Maximilians-Universität München, 80336 Munich, Germany.
| | - Tracy A Williams
- Medizinische Klinik und Poliklinik IV, Klinikum der Universität, Ludwig-Maximilians-Universität München, 80336 Munich, Germany.
- Division of Internal Medicine and Hypertension, Department of Medical Sciences, University of Torino, 10126 Torino, Italy.
| |
Collapse
|