1
|
Aljaibeji H, Heydarpour M, Stanton AM, Williams JS, Pojoga LH, Romero JR, Williams GH. Role of Raptor Gene Variants in Hypertension: Influence on Blood Pressure Independent of Salt Intake in White Population. Hypertension 2024; 81:1167-1177. [PMID: 38497230 PMCID: PMC11023780 DOI: 10.1161/hypertensionaha.123.22273] [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: 10/20/2023] [Accepted: 01/29/2024] [Indexed: 03/19/2024]
Abstract
BACKGROUND The mTOR (mechanistic target of rapamycin) is an essential regulator of fundamental biological processes. mTOR forms 2 distinct complexes, mTORC1 (mTOR complex 1) when it binds with RAPTOR (Regulatory-associated Protein of mTOR) and mTORC2 (mTOR complex 2) when it associates with RICTOR (Rapamycin-insesitive companion of mTOR). Due to the previous link between the mTOR pathway, aldosterone, and blood pressure (BP), we anticipated that variants in the mTOR complex might be associated with salt-sensitive BP. METHODS BP and other parameters were assessed after a one-week liberal Na+ (200 mmol/d) and a one-week restricted Na+ (10 mmol/d) diet in 608 White subjects from the Hypertensive Pathotype cohort, single-nucleotide variants in MTOR, RPTOR, and RICTOR genes were obtained for candidate genes analyses. RESULTS The analysis revealed a significant association between a single nucleotide variants within the RPTOR gene and BP. Individuals carrying the RPTOR rs9901846 homozygous risk allele (AA) and heterozygous risk allele (GA) exhibited a 5 mm Hg increase in systolic BP on a liberal diet compared with nonrisk allele individuals (GG), but only in women. This single nucleotide variants effect was more pronounced on the restricted diet and present in both sexes, with AA carriers having a 9 mm Hg increase and GA carriers having a 5 mm Hg increase in systolic BP compared with GG. Interestingly, there were no significant associations between MTOR or RICTOR gene variants and BP. CONCLUSIONS The RPTOR gene variation is associated with elevated BP in White participants, regardless of salt intake, specifically in females.
Collapse
Affiliation(s)
- Hayat Aljaibeji
- Division of Endocrinology, Diabetes and Hypertension, Department of Medicine, Brigham and Women's Hospital/Harvard Medical School, Boston, MA (H.A., M.H., A.M.S., J.S.W., L.H.P., J.R.R., G.H.W.)
| | - Mahyar Heydarpour
- Division of Endocrinology, Diabetes and Hypertension, Department of Medicine, Brigham and Women's Hospital/Harvard Medical School, Boston, MA (H.A., M.H., A.M.S., J.S.W., L.H.P., J.R.R., G.H.W.)
| | - Ana Maria Stanton
- Division of Endocrinology, Diabetes and Hypertension, Department of Medicine, Brigham and Women's Hospital/Harvard Medical School, Boston, MA (H.A., M.H., A.M.S., J.S.W., L.H.P., J.R.R., G.H.W.)
| | - Jonathan S Williams
- Division of Endocrinology, Diabetes and Hypertension, Department of Medicine, Brigham and Women's Hospital/Harvard Medical School, Boston, MA (H.A., M.H., A.M.S., J.S.W., L.H.P., J.R.R., G.H.W.)
| | - Luminita H Pojoga
- Division of Endocrinology, Diabetes and Hypertension, Department of Medicine, Brigham and Women's Hospital/Harvard Medical School, Boston, MA (H.A., M.H., A.M.S., J.S.W., L.H.P., J.R.R., G.H.W.)
| | - Jose R Romero
- Division of Endocrinology, Diabetes and Hypertension, Department of Medicine, Brigham and Women's Hospital/Harvard Medical School, Boston, MA (H.A., M.H., A.M.S., J.S.W., L.H.P., J.R.R., G.H.W.)
| | - Gordon H Williams
- Division of Endocrinology, Diabetes and Hypertension, Department of Medicine, Brigham and Women's Hospital/Harvard Medical School, Boston, MA (H.A., M.H., A.M.S., J.S.W., L.H.P., J.R.R., G.H.W.)
| |
Collapse
|
2
|
Heydarpour M, Parksook WW, Hopkins PN, Pojoga LH, Williams GH, Williams JS. A candidate locus in the renalase gene and susceptibility to blood pressure responses to the dietary salt. J Hypertens 2023; 41:723-732. [PMID: 36789764 PMCID: PMC10079562 DOI: 10.1097/hjh.0000000000003391] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2022] [Revised: 01/05/2023] [Accepted: 01/20/2023] [Indexed: 02/16/2023]
Abstract
BACKGROUND High dietary salt confers a risk of elevating blood pressure (BP) and the development of hypertension. BP to salt intake may be determined in part by individual genetic predisposition. Identifying these genetic underpinnings will enhance our understanding of the biological mechanisms of BP regulation. This study aims to assess the genetic association with salt sensitivity of BP (SSBP) within two well-phenotyped multinational cohorts. METHODS A total of 720 white participants from the HyperPATH consortium program were selected and genotyped using a multiethnic genotyping array. Individuals consumed two study diets containing high (>200 mEq/day) and low (<10 mEq/day) sodium content, after which SSBP, aldosterone, and plasma renin activity (PRA) were assessed in a controlled inpatient research setting. RESULTS A top signal (rs10887801; beta = 4.57, P = 5.03E - 07) at the renalase gene ( RNLS ) region was significantly associated with SSBP. We also identified seven single nucleotide variants with linkage disequilibrium to the top signal at this region that comprised a significant haplotype (TCTTAGTT, P = 0.00081). Homozygous carriers of the T-risk allele of the key single nucleotide variant had higher SSBP ( P ≤ 0.00001) and lower PRA ( P = 0.0076) compared with the nonrisk allele. CONCLUSION We identified significant associations between genetic variants of the RNLS gene and BP responses to dietary salt intervention and PRA that suggest susceptibility to volume-driven hypertension. These findings may contribute to a better understanding of the genetic mechanisms underlying BP regulation, support the role of RNLS in the pathogenesis of SSBP, and identify individuals who may be at risk from excess dietary salt intake.
Collapse
Affiliation(s)
- Mahyar Heydarpour
- Division of Endocrinology, Diabetes and Hypertension, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Wasita W. Parksook
- Division of Endocrinology, Diabetes and Hypertension, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts, USA
- Division of Endocrinology and Metabolism, and Division of General Internal Medicine, Department of Medicine, Faculty of Medicine, Chulalongkorn University and King Chulalongkorn Memorial Hospital, Thai Red Cross Society, Bangkok, Thailand
| | - Paul N. Hopkins
- Cardiovascular Genetics Research Unit, University of Utah School of Medicine, Salt Lake City, Utah, USA
| | - Luminita H. Pojoga
- Division of Endocrinology, Diabetes and Hypertension, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Gordon H. Williams
- Division of Endocrinology, Diabetes and Hypertension, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Jonathan S. Williams
- Division of Endocrinology, Diabetes and Hypertension, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts, USA
| |
Collapse
|
3
|
KIANI AYSHAKARIM, BONETTI GABRIELE, DONATO KEVIN, KAFTALLI JURGEN, HERBST KARENL, STUPPIA LIBORIO, FIORETTI FRANCESCO, NODARI SAVINA, PERRONE MARCO, CHIURAZZI PIETRO, BELLINATO FRANCESCO, GISONDI PAOLO, BERTELLI MATTEO. Polymorphisms, diet and nutrigenomics. JOURNAL OF PREVENTIVE MEDICINE AND HYGIENE 2022; 63:E125-E141. [PMID: 36479483 PMCID: PMC9710387 DOI: 10.15167/2421-4248/jpmh2022.63.2s3.2754] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Subscribe] [Scholar Register] [Indexed: 01/25/2023]
Abstract
Every human being possesses an exclusive nutritional blueprint inside their genes. Bioactive food components and nutrients affect the expression of such genes. Nutrigenomics is the science that analyzes gene-nutrient interactions (nutrigenetics), which can lead to the development of personalized nutritional recommendations to maintain optimal health and prevent disease. Genomic diversity among various ethnic groups might affect nutrients bioavailability as well as their metabolism. Nutrigenomics combines different branches of science including nutrition, bioinformatics, genomics, molecular biology, molecular medicine, and epidemiology. Genes regulate intake and metabolism of different nutrients, while nutrients positively or negatively influence the expression of a number of genes; testing of specific genetic polymorphisms may therefore become a useful tool to manage weight loss and to fully understand gene-nutrient interactions. Indeed, several approaches are used to study gene-nutrient interactions: epigenetics, the study of genome modification not related to changes in nucleotide sequence; transcriptomics, the study of tissue-specific and time-specific RNA transcripts; proteomics, the study of proteins involved in biological processes; and metabolomics, the study of changes of primary and secondary metabolites in body fluids and tissues. Hence, the use of nutrigenomics to improve and optimize a healthy, balanced diet in clinical settings could be an effective approach for long-term lifestyle changes that might lead to consistent weight loss and improve quality of life.
Collapse
Affiliation(s)
| | - GABRIELE BONETTI
- MAGI’S LAB, Rovereto (TN), Italy
- Correspondence: Gabriele Bonetti, MAGI’S LAB, Rovereto (TN), 38068, Italy. E-mail:
| | | | | | - KAREN L. HERBST
- Total Lipedema Care, Beverly Hills California and Tucson Arizona, USA
| | - LIBORIO STUPPIA
- Department of Psychological, Health and Territorial Sciences, School of Medicine and Health Sciences, “G. d’Annunzio” University, Chieti, Italy
| | - FRANCESCO FIORETTI
- Department of Cardiology, University of Brescia and ASST “Spedali Civili” Hospital, Brescia, Italy
| | - SAVINA NODARI
- Department of Cardiology, University of Brescia and ASST “Spedali Civili” Hospital, Brescia, Italy
| | - MARCO PERRONE
- Department of Cardiology and CardioLab, University of Rome Tor Vergata, Rome, Italy
| | - PIETRO CHIURAZZI
- Istituto di Medicina Genomica, Università Cattolica del Sacro Cuore, Rome, Italy
- UOC Genetica Medica, Fondazione Policlinico Universitario “A. Gemelli” IRCCS, Rome, Italy
| | - FRANCESCO BELLINATO
- Section of Dermatology and Venereology, Department of Medicine, University of Verona, Verona, Italy
| | - PAOLO GISONDI
- Section of Dermatology and Venereology, Department of Medicine, University of Verona, Verona, Italy
| | - MATTEO BERTELLI
- MAGI EUREGIO, Bolzano, Italy
- MAGI’S LAB, Rovereto (TN), Italy
- MAGISNAT, Peachtree Corners (GA), USA
| |
Collapse
|
4
|
Maaliki D, Itani MM, Itani HA. Pathophysiology and genetics of salt-sensitive hypertension. Front Physiol 2022; 13:1001434. [PMID: 36176775 PMCID: PMC9513236 DOI: 10.3389/fphys.2022.1001434] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2022] [Accepted: 08/29/2022] [Indexed: 11/13/2022] Open
Abstract
Most hypertensive cases are primary and heavily associated with modifiable risk factors like salt intake. Evidence suggests that even small reductions in salt consumption reduce blood pressure in all age groups. In that regard, the ACC/AHA described a distinct set of individuals who exhibit salt-sensitivity, regardless of their hypertensive status. Data has shown that salt-sensitivity is an independent risk factor for cardiovascular events and mortality. However, despite extensive research, the pathogenesis of salt-sensitive hypertension is still unclear and tremendously challenged by its multifactorial etiology, complicated genetic influences, and the unavailability of a diagnostic tool. So far, the important roles of the renin-angiotensin-aldosterone system, sympathetic nervous system, and immune system in the pathogenesis of salt-sensitive hypertension have been studied. In the first part of this review, we focus on how the systems mentioned above are aberrantly regulated in salt-sensitive hypertension. We follow this with an emphasis on genetic variants in those systems that are associated with and/or increase predisposition to salt-sensitivity in humans.
Collapse
Affiliation(s)
- Dina Maaliki
- Department of Pharmacology and Toxicology, Faculty of Medicine, American University of Beirut, Beirut, Lebanon
| | - Maha M. Itani
- Department of Pharmacology and Toxicology, Faculty of Medicine, American University of Beirut, Beirut, Lebanon
| | - Hana A. Itani
- Department of Pharmacology and Toxicology, Faculty of Medicine, American University of Beirut, Beirut, Lebanon
- Division of Clinical Pharmacology, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, United States
| |
Collapse
|
5
|
TRPM7 deficiency exacerbates cardiovascular and renal damage induced by aldosterone-salt. Commun Biol 2022; 5:746. [PMID: 35882956 PMCID: PMC9325869 DOI: 10.1038/s42003-022-03715-z] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2020] [Accepted: 07/14/2022] [Indexed: 12/04/2022] Open
Abstract
Hyperaldosteronism causes cardiovascular disease as well as hypomagnesemia. Mechanisms are ill-defined but dysregulation of TRPM7, a Mg2+-permeable channel/α-kinase, may be important. We examined the role of TRPM7 in aldosterone-dependent cardiovascular and renal injury by studying aldosterone-salt treated TRPM7-deficient (TRPM7+/Δkinase) mice. Plasma/tissue [Mg2+] and TRPM7 phosphorylation were reduced in vehicle-treated TRPM7+/Δkinase mice, effects recapitulated in aldosterone-salt-treated wild-type mice. Aldosterone-salt treatment exaggerated vascular dysfunction and amplified cardiovascular and renal fibrosis, with associated increased blood pressure in TRPM7+/Δkinase mice. Tissue expression of Mg2+-regulated phosphatases (PPM1A, PTEN) was downregulated and phosphorylation of Smad3, ERK1/2, and Stat1 was upregulated in aldosterone-salt TRPM7-deficient mice. Aldosterone-induced phosphorylation of pro-fibrotic signaling was increased in TRPM7+/Δkinase fibroblasts, effects ameliorated by Mg2+ supplementation. TRPM7 deficiency amplifies aldosterone-salt-induced cardiovascular remodeling and damage. We identify TRPM7 downregulation and associated hypomagnesemia as putative molecular mechanisms underlying deleterious cardiovascular and renal effects of hyperaldosteronism. Deficiency of the Mg2+-permeable channel/α-kinase TRPM7 in mice increases susceptibility to cardiovascular and renal fibrosis induced by aldosterone and salt.
Collapse
|
6
|
Parksook WW, Heydarpour M, Gholami SK, Luther JM, Hopkins PN, Pojoga LH, Williams JS. Salt Sensitivity of Blood Pressure and Aldosterone: Interaction Between the Lysine-specific Demethylase 1 Gene, Sex, and Age. J Clin Endocrinol Metab 2022; 107:1294-1302. [PMID: 35022775 PMCID: PMC9016472 DOI: 10.1210/clinem/dgac011] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/08/2021] [Indexed: 01/13/2023]
Abstract
CONTEXT Salt sensitivity of blood pressure (SSBP) is associated with increased cardiovascular risk, especially in individuals of African descent, although the underlying mechanisms remain obscure. Lysine-specific demethylase 1 (LSD1) is a salt-sensitive epigenetic regulator associated with SSBP and aldosterone dysfunction. An LSD1 risk allele in humans is associated with SSBP and lower aldosterone levels in hypertensive individuals of African but not European descent. Heterozygous knockout LSD1 mice display SSBP and aldosterone dysregulation, but this effect is modified by age and biological sex. This might explain differences in cardiovascular risk with aging and biological sex in humans. OBJECTIVE This work aims to determine if LSD1 risk allele (rs587618) carriers of African descent display a sex-by-age interaction with SSBP and aldosterone regulation. METHODS We analyzed 297 individuals of African and European descent from the HyperPATH cohort. We performed multiple regression analyses for outcome variables related to SSBP and aldosterone. RESULTS LSD1 risk allele carriers of African (but not European) descent had greater SSBP than nonrisk homozygotes. Female LSD1 risk allele carriers of African descent had greater SSBP, mainly relationship-driven by women with low estrogen (postmenopausal). There was a statistically significant LSD1 genotype-sex interaction in aldosterone response to angiotensin II stimulation in individuals aged 50 years or younger, with female carriers displaying decreased aldosterone responsiveness. CONCLUSION SSBP associated with LSD1 risk allele status is driven by women with a depleted estrogen state. Mechanisms related to a resistance to develop SSBP in females are uncertain but may relate to an estrogen-modulating effect on mineralocorticoid receptor (MR) activation and/or LSD1 epigenetic regulation of the MR.
Collapse
Affiliation(s)
- Wasita W Parksook
- Division of Endocrinology, Diabetes and Hypertension, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts 02115, USA
- Department of Medicine (Division of Endocrinology and Metabolism, and Division of General Internal Medicine), Faculty of Medicine, Chulalongkorn University, and King Chulalongkorn Memorial Hospital, Thai Red Cross Society, Bangkok 10330, Thailand
| | - Mahyar Heydarpour
- Division of Endocrinology, Diabetes and Hypertension, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts 02115, USA
| | - Shadi K Gholami
- Division of Endocrinology, Diabetes and Hypertension, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts 02115, USA
| | - James M Luther
- Division of Clinical Pharmacology, Department of Medicine, Vanderbilt University Medical Center, Vanderbilt Hypertension Center, Nashville, Tennessee 37232, USA
| | - Paul N Hopkins
- Cardiovascular Genetics Research Unit, University of Utah School of Medicine, Salt Lake City, Utah 84132, USA
| | - Luminita H Pojoga
- Division of Endocrinology, Diabetes and Hypertension, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts 02115, USA
| | - Jonathan S Williams
- Division of Endocrinology, Diabetes and Hypertension, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts 02115, USA
| |
Collapse
|
7
|
Haas AV, En Yee L, Yuan YE, Wong YH, Hopkins PN, Jeunemaitre X, Lasky-Su J, Williams JS, Adler GK, Williams GH. Genetic Predictors of Salt Sensitivity of Blood Pressure: The Additive Impact of 2 Hits in the Same Biological Pathway. Hypertension 2021; 78:1809-1817. [PMID: 34757767 DOI: 10.1161/hypertensionaha.121.18033] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
[Figure: see text].
Collapse
Affiliation(s)
- Andrea V Haas
- Division of Endocrinology, Diabetes and Hypertension, Brigham and Women's Hospital, Harvard Medical School, Boston, MA (A.V.H., Y.E.Y., J.S.W., G.K.A., G.H.W.)
| | - Li En Yee
- Cell and Molecular Biology Laboratory, Department of Cellular Biology and Pharmacology, Faculty of Medicine and Health Sciences, UCSI University, Cheras, Kuala Lumpur, Malaysia (L.E.Y., Y.H.W.)
| | - Yan E Yuan
- Division of Endocrinology, Diabetes and Hypertension, Brigham and Women's Hospital, Harvard Medical School, Boston, MA (A.V.H., Y.E.Y., J.S.W., G.K.A., G.H.W.)
| | - Yin H Wong
- Cell and Molecular Biology Laboratory, Department of Cellular Biology and Pharmacology, Faculty of Medicine and Health Sciences, UCSI University, Cheras, Kuala Lumpur, Malaysia (L.E.Y., Y.H.W.)
| | - Paul N Hopkins
- Professor Emeritus, Department of Internal Medicine, University of Utah School of Medicine, Salt Lake City (P.N.H.)
| | - Xavier Jeunemaitre
- Université de Paris, Inserm U970, Paris Centre de Recherche Cardiovasculaire (X.J.).,AP-HP, Hôpital Européen Georges Pompidou, Paris (X.J.)
| | - Jessica Lasky-Su
- Channing Division of Network Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA (J.L.-S.)
| | - Jonathan S Williams
- Division of Endocrinology, Diabetes and Hypertension, Brigham and Women's Hospital, Harvard Medical School, Boston, MA (A.V.H., Y.E.Y., J.S.W., G.K.A., G.H.W.)
| | - Gail K Adler
- Division of Endocrinology, Diabetes and Hypertension, Brigham and Women's Hospital, Harvard Medical School, Boston, MA (A.V.H., Y.E.Y., J.S.W., G.K.A., G.H.W.)
| | - Gordon H Williams
- Division of Endocrinology, Diabetes and Hypertension, Brigham and Women's Hospital, Harvard Medical School, Boston, MA (A.V.H., Y.E.Y., J.S.W., G.K.A., G.H.W.)
| |
Collapse
|
8
|
Sierra-Ramos C, Velazquez-Garcia S, Keskus AG, Vastola-Mascolo A, Rodríguez-Rodríguez AE, Luis-Lima S, Hernández G, Navarro-González JF, Porrini E, Konu O, Alvarez de la Rosa D. Increased SGK1 activity potentiates mineralocorticoid/NaCl-induced kidney injury. Am J Physiol Renal Physiol 2021; 320:F628-F643. [PMID: 33586495 DOI: 10.1152/ajprenal.00505.2020] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
Serum and glucocorticoid-regulated kinase 1 (SGK1) stimulates aldosterone-dependent renal Na+ reabsorption and modulates blood pressure. In addition, genetic ablation or pharmacological inhibition of SGK1 limits the development of kidney inflammation and fibrosis in response to excess mineralocorticoid signaling. In this work, we tested the hypothesis that a systemic increase in SGK1 activity would potentiate mineralocorticoid/salt-induced hypertension and kidney injury. To that end, we used a transgenic mouse model with increased SGK1 activity. Mineralocorticoid/salt-induced hypertension and kidney damage was induced by unilateral nephrectomy and treatment with deoxycorticosterone acetate and NaCl in the drinking water for 6 wk. Our results show that although SGK1 activation did not induce significantly higher blood pressure, it produced a mild increase in glomerular filtration rate, increased albuminuria, and exacerbated glomerular hypertrophy and fibrosis. Transcriptomic analysis showed that extracellular matrix- and immune response-related terms were enriched in the downregulated and upregulated genes, respectively, in transgenic mice. In conclusion, we propose that systemically increased SGK1 activity is a risk factor for the development of mineralocorticoid-dependent kidney injury in the context of low renal mass and independently of blood pressure.NEW & NOTEWORTHY Increased activity of the protein kinase serum and glucocorticoid-regulated kinase 1 may be a risk factor for accelerated renal damage. Serum and glucocorticoid-regulated kinase 1 expression could be a marker for the rapid progression toward chronic kidney disease and a potential therapeutic target to slow down the process.
Collapse
Affiliation(s)
- Catalina Sierra-Ramos
- Departamento de Ciencias Médicas Básicas, Universidad de La Laguna, La Laguna, Tenerife, Spain
| | - Silvia Velazquez-Garcia
- Departamento de Ciencias Médicas Básicas, Universidad de La Laguna, La Laguna, Tenerife, Spain.,Instituto de Tecnologías Biomédicas, Universidad de La Laguna, La Laguna, Tenerife, Spain
| | - Ayse G Keskus
- Interdisciplinary Neuroscience Program, Bilkent University, Ankara, Turkey
| | - Arianna Vastola-Mascolo
- Departamento de Ciencias Médicas Básicas, Universidad de La Laguna, La Laguna, Tenerife, Spain
| | | | - Sergio Luis-Lima
- Instituto de Tecnologías Biomédicas, Universidad de La Laguna, La Laguna, Tenerife, Spain.,Departamento de Medicina Interna, Universidad de La Laguna, La Laguna, Tenerife, Spain
| | - Guadalberto Hernández
- Departamento de Ciencias Médicas Básicas, Universidad de La Laguna, La Laguna, Tenerife, Spain.,Instituto de Tecnologías Biomédicas, Universidad de La Laguna, La Laguna, Tenerife, Spain
| | - Juan F Navarro-González
- Instituto de Tecnologías Biomédicas, Universidad de La Laguna, La Laguna, Tenerife, Spain.,Unidad de Investigación y Servicio de Nefrología, Hospital Universitario Nuestra Señora de Candelaria, Santa Cruz de Tenerife, Spain
| | - Esteban Porrini
- Instituto de Tecnologías Biomédicas, Universidad de La Laguna, La Laguna, Tenerife, Spain.,Departamento de Medicina Interna, Universidad de La Laguna, La Laguna, Tenerife, Spain
| | - Ozlen Konu
- Interdisciplinary Neuroscience Program, Bilkent University, Ankara, Turkey.,Department of Molecular Biology and Genetics, Faculty of Science, Bilkent University, Ankara, Turkey.,UNAM-Institute of Materials Science and Nanotechnology, Ankara, Turkey
| | - Diego Alvarez de la Rosa
- Departamento de Ciencias Médicas Básicas, Universidad de La Laguna, La Laguna, Tenerife, Spain.,Instituto de Tecnologías Biomédicas, Universidad de La Laguna, La Laguna, Tenerife, Spain
| |
Collapse
|
9
|
Chamoun E, Liu AS, Duizer LM, Feng Z, Darlington G, Duncan AM, Haines J, Ma DWL. Single nucleotide polymorphisms in sweet, fat, umami, salt, bitter and sour taste receptor genes are associated with gustatory function and taste preferences in young adults. Nutr Res 2021; 85:40-46. [PMID: 33444969 DOI: 10.1016/j.nutres.2020.12.007] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2020] [Revised: 11/18/2020] [Accepted: 12/01/2020] [Indexed: 01/13/2023]
Abstract
Taste is a fundamental mechanism whereby compounds are detected orally, yet it is highly variable among individuals. The variability in taste that is attributable to genetics is not well-characterized despite its potential role in food selection, and therefore, eating habits that contribute to risk of overweight and obesity. In order to implicate measures of taste function and preference as potentially deterministic factors in adverse eating behaviors that lead to obesity, it must be shown that a relationship exists between genetic variation in taste receptor genes and psychophysical measures of taste in the absence high body mass index. The primary objective of this pilot study was to investigate the relationship between single nucleotide polymorphisms (SNPs) in taste receptor genes and 3 different psychophysical measures of taste in healthy young adults. Sweet, salt, umami, fat, sour, and bitter taste receptor gene SNPs were genotyped in 49 participants (ages 24.6 ± 0.6 years) who completed testing to determine oral detection threshold (DT), suprathreshold sensitivity (ST) and taste preference (PR). A simultaneous association test was conducted between each SNP and the 3 taste outcomes (DT, ST, and PR). Twelve SNPs were associated with at least one of the 3 taste outcomes. Associations were observed between SNPs in taste receptor genes and psychophysical measures of sweet, fat, umami, and salt taste. These results suggest that differences in interindividual psychophysical measures of tastes, namely DT, ST, and PR, may be partially attributed to genetic variation in taste receptor genes. Future studies are warranted to investigate if these findings have consequences for habitual dietary intake of foods that elicit these tastes.
Collapse
Affiliation(s)
- Elie Chamoun
- Department of Human Health and Nutritional Sciences, University of Guelph, Guelph, ON, Canada, N1G2W1
| | - Angel S Liu
- Department of Human Health and Nutritional Sciences, University of Guelph, Guelph, ON, Canada, N1G2W1
| | - Lisa M Duizer
- Department of Food Science, University of Guelph, Guelph, ON, Canada, N1G2W1
| | - Zeny Feng
- Department of Mathematics and Statistics, University of Guelph, Guelph, ON, Canada, N1G2W1
| | - Gerarda Darlington
- Department of Mathematics and Statistics, University of Guelph, Guelph, ON, Canada, N1G2W1
| | - Alison M Duncan
- Department of Human Health and Nutritional Sciences, University of Guelph, Guelph, ON, Canada, N1G2W1
| | - Jess Haines
- Department of Family Relations and Applied Nutrition, University of Guelph, Guelph, ON, Canada, N1G2W1
| | - David W L Ma
- Department of Human Health and Nutritional Sciences, University of Guelph, Guelph, ON, Canada, N1G2W1.
| |
Collapse
|
10
|
Naureen Z, Miggiano GAD, Aquilanti B, Velluti V, Matera G, Gagliardi L, Zulian A, Romanelli R, Bertelli M. Genetic test for the prescription of diets in support of physical activity. ACTA BIO-MEDICA : ATENEI PARMENSIS 2020; 91:e2020011. [PMID: 33170161 PMCID: PMC8023120 DOI: 10.23750/abm.v91i13-s.10584] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Subscribe] [Scholar Register] [Received: 09/03/2020] [Accepted: 09/17/2020] [Indexed: 01/03/2023]
Abstract
Owing to the fields of nutrigenetics and nutrigenomics today we can think of devising approaches to optimize health, delay onset of diseases and reduce its severity according to our genetic blue print. However this requires a deep understanding of nutritional impact on expression of genes that may result in a specific phenotype. The extensive research and observational studies during last two decades reporting interactions between genes, diet and physical activity suggest a cross talk between various genetic and environmental factors and lifestyle interventions. Although considerable efforts have been made in unraveling the mechanisms of gene-diet interactions the scientific evidences behind developing commercial genetic tests for providing personalized nutrition recommendations are still scarce. In this scenario the current mini-review aims to provide useful insights into salient feature of nutrition based genetic research and its commercial application and the ethical issue and concerns related to its outcome.
Collapse
Affiliation(s)
- Zakira Naureen
- Department of Biological Sciences and Chemistry, College of Arts and Sciences, University of Nizwa, Nizwa, Oman.
| | | | - Barbara Aquilanti
- UOC Nutrizione Clinica, Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, Italy.
| | - Valeria Velluti
- UOC Nutrizione Clinica, Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, Italy.
| | - Giuseppina Matera
- UOC Nutrizione Clinica, Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, Italy.
| | - Lucilla Gagliardi
- UOC Nutrizione Clinica, Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, Italy.
| | | | | | - Matteo Bertelli
- MAGI'S LAB, Rovereto (TN), Italy; MAGI EUREGIO, Bolzano, Italy; EBTNA-LAB, Rovereto (TN), Italy.
| |
Collapse
|
11
|
Yang C, Li J, Sun F, Zhou H, Yang J, Yang C. The functional duality of SGK1 in the regulation of hyperglycemia. Endocr Connect 2020; 9:R187-R194. [PMID: 32621586 PMCID: PMC7424354 DOI: 10.1530/ec-20-0225] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/23/2020] [Accepted: 07/02/2020] [Indexed: 12/11/2022]
Abstract
Hyperglycemia is the consequence of blood glucose dysregulation and a driving force of diabetic complications including retinopathy, nephropathy and cardiovascular diseases. The serum and glucocorticoid inducible kinase-1 (SGK1) has been suggested in the modulation of various pathophysiological activities. However, the role of SGK1 in blood glucose homeostasis remains less appreciated. In this review, we intend to summarize the function of SGK1 in glucose level regulation and to examine the evidence supporting the therapeutic potential of SGK1 inhibitors in hyperglycemia. Ample evidence points to the controversial roles of SGK1 in pancreatic insulin secretion and peripheral insulin sensitivity, which reflects the complex interplay between SGK1 activation and blood glucose fluctuation. Furthermore, SGK1 is engaged in glucose absorption and excretion in intestine and kidney and participates in the progression of hyperglycemia-induced secondary organ damage. As a net effect, blockage of SGK1 activation via either pharmacological inhibition or genetic manipulation seems to be helpful in glucose control at varying diabetic stages.
Collapse
Affiliation(s)
- Chunliang Yang
- Department of Integrated Traditional Chinese and Western Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- The Center for Biomedical Research, Tongji Hospital Research Building, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Junyi Li
- Department of Integrated Traditional Chinese and Western Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Fei Sun
- The Center for Biomedical Research, Tongji Hospital Research Building, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Haifeng Zhou
- Department of Integrated Traditional Chinese and Western Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Jia Yang
- Department of Integrated Traditional Chinese and Western Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Correspondence should be addressed to C Yang or J Yang: or
| | - Chao Yang
- Department of Gerontology, Hubei Provincial Hospital of Integrated Chinese and Western Medicine, Wuhan, China
- Correspondence should be addressed to C Yang or J Yang: or
| |
Collapse
|
12
|
Han W, Zhang H, Gong X, Guo Y, Yang M, Zhang H, Zhou X, Li G, Liu Y, Jiang P, Yan G. Association of SGK1 Polymorphisms With Susceptibility to Coronary Heart Disease in Chinese Han Patients With Comorbid Depression. Front Genet 2019; 10:921. [PMID: 31632443 PMCID: PMC6779850 DOI: 10.3389/fgene.2019.00921] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2018] [Accepted: 08/30/2019] [Indexed: 11/26/2022] Open
Abstract
There is a strong link between heart disease and depression, both of which are closely related to lifetime stress exposure. Serum/glucocorticoid-regulated kinase 1 (SGK1) is a stress-responsive gene with a pivotal role in both the heart and brain. To determine the role of SGK1 polymorphisms (rs2758151, rs1743963, rs9493857, rs1763509, rs9376026, and rs9389154) in susceptibility to comorbid coronary heart disease (CHD) and depression, we conducted a hospital-based case–control study involving 257 CHD cases (including 69 cases with depression and 188 cases without depression) and 107 controls in a Chinese Han population. Six single-nucleotide polymorphisms (SNPs) in the SGK1 gene were successfully genotyped by polymerase chain reaction–ligase detection reaction (PCR-LDR) assay. Our results showed no significant differences in SGK1 genetic polymorphisms between CHD patients and controls, whereas significant associations were observed between SGK1 SNPs (rs1743963 and rs1763509) and the development of depression in CHD patients (P = 0.018 by genotype, P = 0.032 by allele; P = 0.017 by genotype, P = 0.003 by allele, respectively). However, none of these associations remained significant after Bonferroni correction (P = 0.054 for rs1743963; P = 0.051 for rs1763509). Interestingly, both the GG genotype of SGK1 rs1743963 and AA genotype of SGK1 rs1763509 were associated with a higher risk of depression in CHD patients; for rs1763509, the Patient Health Questionnaire-9 (PHQ-9) scores in the carriers of the risk genotype for comorbid depression, AA, were significantly higher than in GG and AG carriers (P = 0.008). Notably, haplotype analysis indicated that haplotype GGA significantly increased the risk of depression in CHD patients (P = 0.011, odds ratio (OR) = 1.717, 95% confidence interval (CI) = 1.132–2.605), whereas haplotype AAG may be a protective factor for CHD patients with comorbid depression (P = 0.038, OR = 0.546, 95% CI = 0.307–0.972). It should be noted that only the significance of haplotype GGA survived after Bonferroni adjustment (P = 0.044) and that no significant differences were found for other SGK1 SNPs (rs2758151, rs9493857, rs9376026, and rs9389154) between CHD patients with and without depression. These findings, for the first time, elucidate the important role of SGK1 variants in the comorbidity of CHD and depression.
Collapse
Affiliation(s)
- Wenxiu Han
- Institute of Clinical Pharmacy & Pharmacology, Jining First People's Hospital, Jining Medical University, Jining, China
| | - Haixia Zhang
- Department of Cardiology, Jining First People's Hospital, Jining Medical University, Jining, China
| | - Xiaoxue Gong
- Institute of Clinical Pharmacy & Pharmacology, Jining First People's Hospital, Jining Medical University, Jining, China
| | - Yujin Guo
- Institute of Clinical Pharmacy & Pharmacology, Jining First People's Hospital, Jining Medical University, Jining, China
| | - Mengqi Yang
- Institute of Clinical Pharmacy & Pharmacology, Jining First People's Hospital, Jining Medical University, Jining, China
| | - Hailiang Zhang
- Institute of Clinical Pharmacy & Pharmacology, Jining First People's Hospital, Jining Medical University, Jining, China
| | - Xueyuan Zhou
- Department of Cardiology, Jining First People's Hospital, Jining Medical University, Jining, China
| | - Gongying Li
- Department of Mental Health, Jining Medical University, Jining, China
| | - Yuanyuan Liu
- Research Center of Basic Medical Sciences, Tianjin Medical University, Tianjin, China
| | - Pei Jiang
- Institute of Clinical Pharmacy & Pharmacology, Jining First People's Hospital, Jining Medical University, Jining, China
| | - Genquan Yan
- Department of Pharmacy, Shandong Provincial Hospital affiliated to Shandong University, Jinan, China
| |
Collapse
|
13
|
Haas AV, Hopkins PN, Brown NJ, Pojoga LH, Williams JS, Adler GK, Williams GH. Higher urinary cortisol levels associate with increased cardiovascular risk. Endocr Connect 2019; 8:634-640. [PMID: 31018177 PMCID: PMC6528405 DOI: 10.1530/ec-19-0182] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/10/2019] [Accepted: 04/23/2019] [Indexed: 12/31/2022]
Abstract
There are conflicting data on whether variations of physiologic cortisol levels associated with cardiovascular risk. We hypothesize that prior discordant findings are related to problems associated with varying sample size, techniques for assessing cardiovascular risk and failure to adequately account for environmental factors. To address these issues, we utilized a large sample size, selected the Framingham risk score to compute cardiovascular risk and performed the study in a highly controlled setting. We had two main objectives: determine whether higher, yet physiologic, cortisol levels associated with increased cardiovascular risk and determine whether caveolin-1 (rs926198) risk allele carriers associated with increased cardiovascular risk. This was a cross-sectional study of 574 non-diabetic individuals who completed a common protocol. Data collection included fasting blood samples, blood pressure measurements and a 24-h urine-free cortisol collection. Five hundred seventeen of these participants also completed caveolin-1 genotyping. Subjects were classified as belonging to either the low-mode or high-mode urine-free cortisol groups, based on the bimodal distribution of urine-free cortisol. In multivariate analysis, Framingham risk score was statistically higher in the high-mode cortisol group (10.22 (mean) ± 0.43 (s.e.m.)) compared to the low-mode cortisol group (7.73 ± 0.34), P < 0.001. Framingham risk score was also statistically higher in the caveolin-1 risk allele carriers (8.91 ± 0.37) compared to caveolin-1 non-risk allele carriers (7.59 ± 0.48), P = 0.034. Overall, the estimated effect on Framingham risk score of carrying the caveolin-1 risk allele was 1.33 ± 0.61, P = 0.029. Both urinary cortisol and caveolin-1 risk allele status are independent predictors of Framingham risk score.
Collapse
Affiliation(s)
- Andrea V Haas
- Division of Endocrinology, Diabetes and Hypertension, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, Massachusetts, USA
- Correspondence should be addressed to A V Haas:
| | - Paul N Hopkins
- Cardiovascular Genetics, Department of Internal Medicine, University of Utah School of Medicine, Salt Lake City, Utah, USA
| | - Nancy J Brown
- Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - Luminita H Pojoga
- Division of Endocrinology, Diabetes and Hypertension, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Jonathan S Williams
- Division of Endocrinology, Diabetes and Hypertension, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Gail K Adler
- Division of Endocrinology, Diabetes and Hypertension, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Gordon H Williams
- Division of Endocrinology, Diabetes and Hypertension, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, Massachusetts, USA
| |
Collapse
|
14
|
Manosroi W, Williams GH. Genetics of Human Primary Hypertension: Focus on Hormonal Mechanisms. Endocr Rev 2019; 40:825-856. [PMID: 30590482 PMCID: PMC6936319 DOI: 10.1210/er.2018-00071] [Citation(s) in RCA: 63] [Impact Index Per Article: 12.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/01/2018] [Accepted: 09/07/2018] [Indexed: 02/06/2023]
Abstract
Increasingly, primary hypertension is being considered a syndrome and not a disease, with the individual causes (diseases) having a common sign-an elevated blood pressure. To determine these causes, genetic tools are increasingly employed. This review identified 62 proposed genes. However, only 21 of them met our inclusion criteria: (i) primary hypertension, (ii) two or more supporting cohorts from different publications or within a single publication or one supporting cohort with a confirmatory genetically modified animal study, and (iii) 600 or more subjects in the primary cohort; when including our exclusion criteria: (i) meta-analyses or reviews, (ii) secondary and monogenic hypertension, (iii) only hypertensive complications, (iv) genes related to blood pressure but not hypertension per se, (v) nonsupporting studies more common than supporting ones, and (vi) studies that did not perform a Bonferroni or similar multiassessment correction. These 21 genes were organized in a four-tiered structure: distant phenotype (hypertension); intermediate phenotype [salt-sensitive (18) or salt-resistant (0)]; subintermediate phenotypes under salt-sensitive hypertension [normal renin (4), low renin (8), and unclassified renin (6)]; and proximate phenotypes (specific genetically driven hypertensive subgroup). Many proximate hypertensive phenotypes had a substantial endocrine component. In conclusion, primary hypertension is a syndrome; many proposed genes are likely to be false positives; and deep phenotyping will be required to determine the utility of genetics in the treatment of hypertension. However, to date, the positive genes are associated with nearly 50% of primary hypertensives, suggesting that in the near term precise, mechanistically driven treatment and prevention strategies for the specific primary hypertension subgroups are feasible.
Collapse
Affiliation(s)
- Worapaka Manosroi
- Division of Endocrinology, Diabetes, and Hypertension, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts.,Division of Endocrinology and Metabolism, Faculty of Medicine, Chiang Mai University, Chiang Mai, Thailand
| | - Gordon H Williams
- Division of Endocrinology, Diabetes, and Hypertension, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts
| |
Collapse
|
15
|
Sodium sensitivity of blood pressure in Chinese populations. J Hum Hypertens 2019; 34:94-107. [PMID: 30631129 DOI: 10.1038/s41371-018-0152-0] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2018] [Revised: 11/15/2018] [Accepted: 12/06/2018] [Indexed: 12/12/2022]
Abstract
Hypertension is an enormous public-health challenge in the world due to its high prevalence and consequent increased cardiovascular disease morbidity and mortality. Observational epidemiologic studies and clinical trials have demonstrated a causal relationship between sodium intake and elevated blood pressure (BP). However, BP changes in response to sodium intervention vary among individuals-a trait called sodium sensitivity. This paper aims to review the recent advances in sodium-sensitivity research in Chinese and other populations. Older age, female gender, and black race are associated with high sodium sensitivity. Both genetic and environmental factors influence BP sodium sensitivity. Physical activity and dietary potassium intake are associated with reduced sodium sensitivity while obesity, metabolic syndrome, and elevated BP are associated with increased sodium sensitivity. Familial studies have documented a moderate heritability of sodium sensitivity. Candidate gene association studies, genome-wide association studies, whole-exome, and whole-genome sequencing studies have been conducted to elucidate the genomic mechanisms of sodium sensitivity. The Genetic Epidemiology Network of Salt Sensitivity (GenSalt) study, the largest family-based feeding study to date, was conducted among 1906 Han Chinese in rural northern China. This study showed that ~32.4% of Chinese adults were sodium sensitive. Additionally, several genetic variants were found to be associated with sodium sensitivity. Findings from the GenSalt Study and others indicate that sodium sensitivity is a reproducible trait and both lifestyle factors and genetic variants play a role in this complex trait. Discovering biomarkers and underlying mechanisms for sodium sensitivity will help to develop individualized intervention strategies for hypertension.
Collapse
|
16
|
The Relationship between Single Nucleotide Polymorphisms in Taste Receptor Genes, Taste Function and Dietary Intake in Preschool-Aged Children and Adults in the Guelph Family Health Study. Nutrients 2018; 10:nu10080990. [PMID: 30060620 PMCID: PMC6115723 DOI: 10.3390/nu10080990] [Citation(s) in RCA: 33] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2018] [Revised: 07/19/2018] [Accepted: 07/27/2018] [Indexed: 12/14/2022] Open
Abstract
Taste is a fundamental determinant of food selection, and inter-individual variations in taste perception may be important risk factors for poor eating habits and obesity. Characterizing differences in taste perception and their influences on dietary intake may lead to an improved understanding of obesity risk and a potential to develop personalized nutrition recommendations. This study explored associations between 93 single nucleotide polymorphisms (SNPs) in sweet, fat, bitter, salt, sour, and umami taste receptors and psychophysical measures of taste. Forty-four families from the Guelph Family Health Study participated, including 60 children and 65 adults. Saliva was collected for genetic analysis and parents completed a three-day food record for their children. Parents underwent a test for suprathreshold sensitivity (ST) and taste preference (PR) for sweet, fat, salt, umami, and sour as well as a phenylthiocarbamide (PTC) taste status test. Children underwent PR tests and a PTC taste status test. Analysis of SNPs and psychophysical measures of taste yielded 23 significant associations in parents and 11 in children. After adjusting for multiple hypothesis testing, the rs713598 in the TAS2R38 bitter taste receptor gene and rs236514 in the KCNJ2 sour taste-associated gene remained significantly associated with PTC ST and sour PR in parents, respectively. In children, rs173135 in KCNJ2 and rs4790522 in the TRPV1 salt taste-associated gene remained significantly associated with sour and salt taste PRs, respectively. A multiple trait analysis of PR and nutrient composition of diet in the children revealed that rs9701796 in the TAS1R2 sweet taste receptor gene was associated with both sweet PR and percent energy from added sugar in the diet. These findings provide evidence that for bitter, sour, salt, and sweet taste, certain genetic variants are associated with taste function and may be implicated in eating patterns. (Support was provided by the Ontario Ministry of Agriculture, Food, and Rural Affairs).
Collapse
|
17
|
Harvey BJ, Thomas W. Aldosterone-induced protein kinase signalling and the control of electrolyte balance. Steroids 2018; 133:67-74. [PMID: 29079406 DOI: 10.1016/j.steroids.2017.10.009] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/27/2017] [Revised: 10/18/2017] [Accepted: 10/21/2017] [Indexed: 01/20/2023]
Abstract
Aldosterone acts through the mineralocorticoid receptor (MR) to modulate gene expression in target tissues. In the kidney, the principal action of aldosterone is to promote sodium conservation in the distal nephron and so indirectly enhance water conservation under conditions of hypotension. Over the last twenty years the rapid activation of protein kinase signalling cascades by aldosterone has been described in various tissues. This review describes the integration of rapid protein kinase D signalling responses with the non-genomic actions of aldosterone and transcriptional effects of MR activation.
Collapse
Affiliation(s)
- Brian J Harvey
- Molecular Medicine Laboratories, Royal College of Surgeons in Ireland, Education Centre, Beaumont Hospital, Dublin, Ireland
| | - Warren Thomas
- Molecular Medicine Laboratories, Royal College of Surgeons in Ireland, Education Centre, Beaumont Hospital, Dublin, Ireland; Perdana University - Royal College of Surgeons in Ireland School of Medicine, Serdang, Selangor, Malaysia.
| |
Collapse
|
18
|
Manosroi W, Tan JW, Rariy CM, Sun B, Goodarzi MO, Saxena AR, Williams JS, Pojoga LH, Lasky-Su J, Cui J, Guo X, Taylor KD, Chen YDI, Xiang AH, Hsueh WA, Raffel LJ, Buchanan TA, Rotter JI, Williams GH, Seely EW. The Association of Estrogen Receptor-β Gene Variation With Salt-Sensitive Blood Pressure. J Clin Endocrinol Metab 2017; 102:4124-4135. [PMID: 28938457 PMCID: PMC5673274 DOI: 10.1210/jc.2017-00957] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/22/2017] [Accepted: 08/29/2017] [Indexed: 11/19/2022]
Abstract
CONTEXT Hypertension in young women is uncommon compared with young men and older women. Estrogen appears to protect most women against hypertension, with incidence increasing after menopause. Because some premenopausal women develop hypertension, estrogen may play a different role in these women. Genetic variations in the estrogen receptor (ER) are associated with cardiovascular disease. ER-β, encoded by ESR2, is the ER predominantly expressed in vascular smooth muscle. OBJECTIVE To determine an association of single nucleotide polymorphisms in ESR2 with salt sensitivity of blood pressure (SSBP) and estrogen status in women. METHODS Candidate gene association study with ESR2 and SSBP conducted in normotensive and hypertensive women and men in two cohorts: International Hypertensive Pathotype (HyperPATH) (n = 584) (discovery) and Mexican American Hypertension-Insulin Resistance Study (n = 662) (validation). Single nucleotide polymorphisms in ESR1 (ER-α) were also analyzed. Analysis conducted in younger (<51 years, premenopausal, "estrogen-replete") and older women (≥51 years, postmenopausal, "estrogen-deplete"). Men were analyzed to control for aging. RESULTS Multivariate analyses of HyperPATH data between variants of ESR2 and SSBP documented that ESR2 rs10144225 minor (risk) allele carriers had a significantly positive association with SSBP driven by estrogen-replete women (β = +4.4 mm Hg per risk allele, P = 0.004). Findings were confirmed in Hypertension Insulin-Resistance Study premenopausal women. HyperPATH cohort analyses revealed risk allele carriers vs noncarriers had increased aldosterone/renin ratios. No associations were detected with ESR1. CONCLUSIONS The variation at rs10144225 in ESR2 was associated with SSBP in premenopausal women (estrogen-replete) and not in men or postmenopausal women (estrogen-deplete). Inappropriate aldosterone levels on a liberal salt diet may mediate the SSBP.
Collapse
Affiliation(s)
- Worapaka Manosroi
- Division of Endocrinology, Diabetes, and Hypertension, Brigham and Women’s Hospital, Harvard Medical School, Boston, Massachusetts 02115
- Division of Endocrinology and Metabolism, Faculty of Medicine, Chiang Mai University, Chiang Mai 50200, Thailand
| | - Jia Wei Tan
- Division of Endocrinology, Diabetes, and Hypertension, Brigham and Women’s Hospital, Harvard Medical School, Boston, Massachusetts 02115
- Cell and Molecular Biology Laboratory, Department of Cellular Biology and Pharmacology, Faculty of Medicine and Health Sciences, UCSI University, Cheras 56000, Kuala Lumpur, Malaysia
| | - Chevon M. Rariy
- Division of Endocrinology, Diabetes, and Hypertension, Brigham and Women’s Hospital, Harvard Medical School, Boston, Massachusetts 02115
| | - Bei Sun
- Division of Endocrinology, Diabetes, and Hypertension, Brigham and Women’s Hospital, Harvard Medical School, Boston, Massachusetts 02115
| | - Mark O. Goodarzi
- Division of Endocrinology, Diabetes and Metabolism, Cedars-Sinai Medical Center, Los Angeles, California 90048
| | - Aditi R. Saxena
- Division of Endocrinology, Diabetes, and Hypertension, Brigham and Women’s Hospital, Harvard Medical School, Boston, Massachusetts 02115
| | - Jonathan S. Williams
- Division of Endocrinology, Diabetes, and Hypertension, Brigham and Women’s Hospital, Harvard Medical School, Boston, Massachusetts 02115
| | - Luminita H. Pojoga
- Division of Endocrinology, Diabetes, and Hypertension, Brigham and Women’s Hospital, Harvard Medical School, Boston, Massachusetts 02115
| | - Jessica Lasky-Su
- Channing Laboratory, Department of Medicine, Brigham and Women’s Hospital and Harvard Medical School, Boston, Massachusetts 02115
| | - Jinrui Cui
- Division of Endocrinology, Diabetes and Metabolism, Cedars-Sinai Medical Center, Los Angeles, California 90048
| | - Xiuqing Guo
- Institute for Translational Genomics and Population Sciences, Harbor-UCLA Medical Center, Torrance, California 90502
- Department of Pediatrics, Los Angeles Biomedical Research Institute, Harbor-UCLA Medical Center, Torrance, California 90502
| | - Kent D. Taylor
- Institute for Translational Genomics and Population Sciences, Harbor-UCLA Medical Center, Torrance, California 90502
- Department of Pediatrics, Los Angeles Biomedical Research Institute, Harbor-UCLA Medical Center, Torrance, California 90502
| | - Yii-Der I. Chen
- Institute for Translational Genomics and Population Sciences, Harbor-UCLA Medical Center, Torrance, California 90502
- Department of Pediatrics, Los Angeles Biomedical Research Institute, Harbor-UCLA Medical Center, Torrance, California 90502
| | - Anny H. Xiang
- Department of Research and Evaluation, Kaiser Permanente Southern California, Pasadena, California 91101
| | - Willa A. Hsueh
- Department of Research and Evaluation, Kaiser Permanente Southern California, Pasadena, California 91101
- Division of Endocrinology, Diabetes and Metabolism and Diabetes and Metabolism Research Center, The Ohio State University, Columbus, Ohio 43210
| | - Leslie J. Raffel
- Division of Genetic and Genomic Medicine, Department of Pediatrics, University of California, Irvine, California 92868
| | - Thomas A. Buchanan
- Department of Medicine, Keck School of Medicine, University of Southern California, Los Angeles, Los Angeles, California 90089
| | - Jerome I. Rotter
- Institute for Translational Genomics and Population Sciences, Harbor-UCLA Medical Center, Torrance, California 90502
- Department of Pediatrics, Los Angeles Biomedical Research Institute, Harbor-UCLA Medical Center, Torrance, California 90502
| | - Gordon H. Williams
- Division of Endocrinology, Diabetes, and Hypertension, Brigham and Women’s Hospital, Harvard Medical School, Boston, Massachusetts 02115
| | - Ellen W. Seely
- Division of Endocrinology, Diabetes, and Hypertension, Brigham and Women’s Hospital, Harvard Medical School, Boston, Massachusetts 02115
| |
Collapse
|
19
|
Lang F, Guelinckx I, Lemetais G, Melander O. Two Liters a Day Keep the Doctor Away? Considerations on the Pathophysiology of Suboptimal Fluid Intake in the Common Population. Kidney Blood Press Res 2017; 42:483-494. [PMID: 28787716 DOI: 10.1159/000479640] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2017] [Accepted: 04/19/2017] [Indexed: 11/19/2022] Open
Abstract
Suboptimal fluid intake may require enhanced release of antidiuretic hormone (ADH) or vasopressin for the maintenance of adequate hydration. Enhanced copeptin levels (reflecting enhanced vasopressin levels) in 25% of the common population are associated with enhanced risk of metabolic syndrome with abdominal obesity, type 2 diabetes, hypertension, coronary artery disease, heart failure, vascular dementia, cognitive impairment, microalbuminuria, chronic kidney disease, inflammatory bowel disease, cancer, and premature mortality. Vasopressin stimulates the release of glucocorticoids which in turn up-regulate the serum- and glucocorticoid-inducible kinase 1 (SGK1). Moreover, dehydration upregulates the transcription factor NFAT5, which in turn stimulates SGK1 expression. SGK1 is activated by insulin, growth factors and oxidative stress via phosphatidylinositide-3-kinase, 3-phosphoinositide-dependent kinase PDK1 and mTOR. SGK1 is a powerful stimulator of Na+/K+-ATPase, carriers (e.g. the Na+,K+,2Cl- cotransporter NKCC, the NaCl cotransporter NCC, the Na+/H+ exchanger NHE3, and the Na+ coupled glucose transporter SGLT1), and ion channels (e.g. the epithelial Na+ channel ENaC, the Ca2+ release activated Ca2+ channel Orai1 with its stimulator STIM1, and diverse K+ channels). SGK1 further participates in the regulation of the transcription factors nuclear factor kappa-B NFκB, p53, cAMP responsive element binding protein (CREB), activator protein-1, and forkhead transcription factor FKHR-L1 (FOXO3a). Enhanced SGK1 activity fosters the development of hypertension, obesity, diabetes, thrombosis, stroke, inflammation including inflammatory bowel disease and autoimmune disease, cardiac fibrosis, proteinuria, renal failure as well as tumor growth. The present brief review makes the case that suboptimal fluid intake in the common population may enhance vasopressin and glucocorticoid levels thus up-regulating SGK1 expression and favouring the development of SGK1 related pathologies.
Collapse
Affiliation(s)
- Florian Lang
- Department of Physiology I, University of Tuebingen, Tuebingen, Germany
| | | | | | - Olle Melander
- Department of Clinical Sciences, Lund University, Malmö, Sweden
| |
Collapse
|
20
|
Iatrino R, Manunta P, Zagato L. Salt Sensitivity: Challenging and Controversial Phenotype of Primary Hypertension. Curr Hypertens Rep 2016; 18:70. [DOI: 10.1007/s11906-016-0677-y] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
|
21
|
Lou Y, Zhang F, Luo Y, Wang L, Huang S, Jin F. Serum and Glucocorticoid Regulated Kinase 1 in Sodium Homeostasis. Int J Mol Sci 2016; 17:ijms17081307. [PMID: 27517916 PMCID: PMC5000704 DOI: 10.3390/ijms17081307] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2016] [Revised: 08/02/2016] [Accepted: 08/03/2016] [Indexed: 12/13/2022] Open
Abstract
The ubiquitously expressed serum and glucocorticoid regulated kinase 1 (SGK1) is tightly regulated by osmotic and hormonal signals, including glucocorticoids and mineralocorticoids. Recently, SGK1 has been implicated as a signal hub for the regulation of sodium transport. SGK1 modulates the activities of multiple ion channels and carriers, such as epithelial sodium channel (ENaC), voltage-gated sodium channel (Nav1.5), sodium hydrogen exchangers 1 and 3 (NHE1 and NHE3), sodium-chloride symporter (NCC), and sodium-potassium-chloride cotransporter 2 (NKCC2); as well as the sodium-potassium adenosine triphosphatase (Na+/K+-ATPase) and type A natriuretic peptide receptor (NPR-A). Accordingly, SGK1 is implicated in the physiology and pathophysiology of Na+ homeostasis. Here, we focus particularly on recent findings of SGK1’s involvement in Na+ transport in renal sodium reabsorption, hormone-stimulated salt appetite and fluid balance and discuss the abnormal SGK1-mediated Na+ reabsorption in hypertension, heart disease, edema with diabetes, and embryo implantation failure.
Collapse
Affiliation(s)
- Yiyun Lou
- Department of Reproductive Endocrinology, Women's Hospital, School of Medicine, Zhejiang University, Hangzhou 310006, Zhejiang, China.
- Department of Gynaecology, Hangzhou Hospital of Traditional Chinese Medicine, Hangzhou 310007, Zhejiang, China.
| | - Fan Zhang
- Department of Reproductive Endocrinology, Women's Hospital, School of Medicine, Zhejiang University, Hangzhou 310006, Zhejiang, China.
| | - Yuqin Luo
- Department of Reproductive Endocrinology, Women's Hospital, School of Medicine, Zhejiang University, Hangzhou 310006, Zhejiang, China.
| | - Liya Wang
- Department of Reproductive Endocrinology, Women's Hospital, School of Medicine, Zhejiang University, Hangzhou 310006, Zhejiang, China.
| | - Shisi Huang
- Department of Reproductive Endocrinology, Women's Hospital, School of Medicine, Zhejiang University, Hangzhou 310006, Zhejiang, China.
| | - Fan Jin
- Department of Reproductive Endocrinology, Women's Hospital, School of Medicine, Zhejiang University, Hangzhou 310006, Zhejiang, China.
- Key Laboratory of Reproductive Genetics, National Ministry of Education (Zhejiang University), Women's Reproductive Healthy Laboratory of Zhejiang Province, Hangzhou 310058, Zhejiang, China.
| |
Collapse
|
22
|
Kohlmeier M, De Caterina R, Ferguson LR, Görman U, Allayee H, Prasad C, Kang JX, Nicoletti CF, Martinez JA. Guide and Position of the International Society of Nutrigenetics/Nutrigenomics on Personalized Nutrition: Part 2 - Ethics, Challenges and Endeavors of Precision Nutrition. JOURNAL OF NUTRIGENETICS AND NUTRIGENOMICS 2016; 9:28-46. [PMID: 27286972 DOI: 10.1159/000446347] [Citation(s) in RCA: 51] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Nutrigenetics considers the influence of individual genetic variation on differences in response to dietary components, nutrient requirements and predisposition to disease. Nutrigenomics involves the study of interactions between the genome and diet, including how nutrients affect the transcription and translation process plus subsequent proteomic and metabolomic changes, and also differences in response to dietary factors based on the individual genetic makeup. Personalized characteristics such as age, gender, physical activity, physiological state and social status, and special conditions such as pregnancy and risk of disease can inform dietary advice that more closely meets individual needs. Precision nutrition has a promising future in treating the individual according to their phenotype and genetic characteristics, aimed at both the treatment and prevention of disease. However, many aspects are still in progress and remain as challenges for the future of nutrition. The integration of the human genotype and microbiome needs to be better understood. Further advances in data interpretation tools are also necessary, so that information obtained through newer tests and technologies can be properly transferred to consumers. Indeed, precision nutrition will integrate genetic data with phenotypical, social, cultural and personal preferences and lifestyles matters to provide a more individual nutrition, but considering public health perspectives, where ethical, legal and policy aspects need to be defined and implemented.
Collapse
Affiliation(s)
- Martin Kohlmeier
- Department of Nutrition, School of Public Health, School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, N.C., USA
| | | | | | | | | | | | | | | | | |
Collapse
|
23
|
Beltrán-Debón R, Rodríguez-Gallego E, Fernández-Arroyo S, Senan-Campos O, Massucci FA, Hernández-Aguilera A, Sales-Pardo M, Guimerà R, Camps J, Menendez JA, Joven J. The acute impact of polyphenols from Hibiscus sabdariffa in metabolic homeostasis: an approach combining metabolomics and gene-expression analyses. Food Funct 2016; 6:2957-66. [PMID: 26234931 DOI: 10.1039/c5fo00696a] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023]
Abstract
We explored the acute multifunctional effects of polyphenols from Hibiscus sabdariffa in humans to assess possible consequences on the host's health. The expected dynamic response was studied using a combination of transcriptomics and metabolomics to integrate specific functional pathways through network-based methods and to generate hypotheses established by acute metabolic effects and/or modifications in the expression of relevant genes. Data were obtained from healthy male volunteers after 3 hours of ingestion of an aqueous Hibiscus sabdariffa extract. The data were compared with data obtained prior to the ingestion, and the overall findings suggest that these particular polyphenols had a simultaneous role in mitochondrial function, energy homeostasis and protection of the cardiovascular system. These findings suggest beneficial actions in inflammation, endothelial dysfunction, and oxidation, which are interrelated mechanisms. Among other effects, the activation of the heme oxygenase-biliverdin reductase axis, the systemic inhibition of the renin-angiotensin system, the inhibition of the angiotensin-converting enzyme, and several actions mirroring those of the peroxisome proliferator-activated receptor agonists further support this notion. We also found concordant findings in the serum of the participants, which include a decrease in cortisol levels and a significant increase in the active vasodilator metabolite of bradykinin (des-Arg(9)-bradykinin). Therefore, our data support the view that polyphenols from Hibiscus sabdariffa play a regulatory role in metabolic health and in the maintenance of blood pressure, thus implying a multi-faceted impact in metabolic and cardiovascular diseases.
Collapse
Affiliation(s)
- Raúl Beltrán-Debón
- Unitat de Recerca Biomèdica, Hospital Universitari Sant Joan, Institut d'Investigació Sanitària Pere Virgili, Universitat Rovira i Virgili, Campus of International excellence Southern Catalonia, Carrer Sant Llorenç 21, 43201-Reus, Spain.
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
24
|
SGK-1 protects kidney cells against apoptosis induced by ceramide and TNF-α. Cell Death Dis 2015; 6:e1890. [PMID: 26379195 PMCID: PMC4650437 DOI: 10.1038/cddis.2015.232] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2015] [Revised: 06/25/2015] [Accepted: 07/06/2015] [Indexed: 12/21/2022]
Abstract
Ceramide regulates several different cellular responses including mechanisms leading to apoptosis. Serum- and glucocorticoid-inducible protein kinase (SGK)-1 is a serine threonine kinase, which activates survival pathways in response to stress stimuli. Recently, we demonstrated an anti-apoptotic role of SGK-1 in human umbilical endothelial cells treated with high glucose. In the present study, since ceramide induces apoptosis by multiple mechanisms in diabetes and its complication such as nephropathy, we aimed to investigate whether SGK-1 may protect even against apoptosis induced by ceramide in kidney cells. Human embryonic kidney (HEK)-293 cells stable transfected with SGK-1 wild type (SGK-1wt) and its dominant negative gene (SGK-1dn) have been used in this study. Apoptotic stimuli were induced by C2-ceramide and TNF-α to increase endogenous synthesis of ceramide. Upon activation with these stimuli, SGK-1wt transfected cells have a statistically significant reduction of apoptosis compared with SGK-1dn cells (P<0.001). This protection was dependent on activation of caspase-3 and Poly-ADP-ribose-polymerase-1 (PARP-1) cleavage. SGK-1 and AKT-1 two highly homologous kinases differently reacted to ceramide treatment, since SGK-1 increases in response to apoptotic stimulus while AKT-1 decreases. This enhancement of SGK-1 was dependent on p38-mitogen-activated-protein kinases (p38MAPK), cyclic-adenosine-monophosphate/protein kinase A (cAMP/PKA) and phosphoinositide-3-kinase (PI3K) pathways. Especially, by using selective LY294002 inhibitor, we demonstrated that the most involved pathway in the SGK-1 mediated process of protection was PI3K. Treatment with inhibitor of SGK-1 (GSK650394) significantly enhanced TNF-α-dependent apoptosis in HEK-293 cells overexpressing SGK-1wt. Caspase-3, -8 and -9 selective inhibitors confirmed that SGK-1 reduced the activation of caspase-dependent apoptosis, probably by both intrinsic and extrinsic pathways. In conclusion, we demonstrated that in kidney cells, overexpression of SGK-1 is protective against ceramide-induced apoptosis and the role of SGK-1 can be potentially explored as a therapeutic target in conditions like diabetes, where ceramide levels are increased.
Collapse
|
25
|
Lang F, Pearce D. Regulation of the epithelial Na+ channel by the mTORC2/SGK1 pathway. Nephrol Dial Transplant 2015; 31:200-5. [PMID: 26163195 DOI: 10.1093/ndt/gfv270] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2015] [Accepted: 06/05/2015] [Indexed: 12/25/2022] Open
Abstract
The epithelial Na(+) channel (ENaC) is decisive for sodium reabsorption by the aldosterone-sensitive distal nephron (ASDN) of the kidney. ENaC is regulated by the serum- and glucocorticoid-inducible kinase 1 (SGK1), a kinase genomically upregulated by several hormones including glucocorticoids and mineralocorticoids. SGK1 is activated by the serine/threonine kinase mammalian target of rapamycin (mTOR) isoform mTORC2. SGK1 knockout (sgk1(-/-) mice) impairs renal Na(+) retention during salt depletion. The mTOR catalytic site inhibitor, PP242, but not mTORC1 inhibitor rapamycin, inhibits ENaC, decreases Na(+) flux in isolated perfused tubules and induces natriuresis in wild-type mice. PP242 does not lead to further impairment of Na(+) reabsorption in sgk1(-/-) mice. The mTORC2/SGK1 sensitive renal Na(+) retention leads to extracellular volume expansion with increase of blood pressure. A SGK1 gene variant (prevalence ∼ 3-5% in Caucasians, ∼ 10% in Africans) predisposes to hypertension, stroke, obesity and type 2 diabetes. Future studies will be required to define the role of mTORC2 in the regulation of further SGK1 sensitive transport proteins, such as further ion channels, carriers and the Na(+)/K(+)-ATPase. Moreover, studies are required disclosing the impact of mTORC2 on SGK1 sensitive disorders, such as hypertension, obesity, diabetes, thrombosis, stroke, inflammation, autoimmune disease, fibrosis and tumour growth.
Collapse
Affiliation(s)
- Florian Lang
- Department of Physiology, University of Tübingen, Tübingen, Germany
| | - David Pearce
- Division of Nephrology, Department of Medicine, University of California at San Francisco, San Francisco, CA, USA
| |
Collapse
|
26
|
Duan P, Wang ZM, Liu J, Wang LN, Yang Z, Tu P. Association of gene polymorphisms in RANKL/RANK/OPG system with hypertension and blood pressure in Chinese women. J Hum Hypertens 2015; 29:749-53. [PMID: 25810067 DOI: 10.1038/jhh.2015.13] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2014] [Revised: 12/09/2014] [Accepted: 01/20/2015] [Indexed: 02/05/2023]
Abstract
Recent studies have revealed that the receptor activator of nuclear factor-kappa B ligand/RANK/osteoprotegerin (RANKL/RANK/OPG) system has an important role in vascular calcification, which is contributory to various cardiovascular diseases and intimately linked to the regulation of blood pressure. Therefore, we performed a case-control study to investigate the associations of 21 single-nucleotide polymorphisms (SNPs) in the TNFSF11, TNFRSF11A and TNFRSF11B genes in the RANKL/RANK/OPG system with hypertension and blood pressure in post-menopausal Chinese women. In this study, 503 hypertensive patients and 509 normal controls were recruited. Genotyping was performed using the high-throughput Sequenom genotyping platform. The results showed that two SNPs (rs6567270 and rs4603673) in the TNFRSF11A were associated with hypertension (P=0.010 and P=0.013, respectively) and systolic blood pressure (P=0.024 and P=0.023, respectively). One SNP (rs9646629) in the TNFRSF11A showed significant association with diastolic blood pressure (P=0.031). The results of this study suggest that TNFRSF11A but not TNFSF11 and TNFRSF11B genetic variation is associated with hypertension and blood pressure in Chinese women. The findings provide additional support for the genetic role of RANKL/RANK/OPG system in hypertension and blood pressure regulation.
Collapse
Affiliation(s)
- P Duan
- Department of Endocrinology and Metabolism, Nanchang Key Laboratory of Diabetes, The Third Hospital of Nanchang, Nanchang city, People's Republic of China
| | - Z-M Wang
- Department of Endocrinology and Metabolism, Nanchang Key Laboratory of Diabetes, The Third Hospital of Nanchang, Nanchang city, People's Republic of China
| | - J Liu
- Department of Endocrinology and Metabolism, Nanchang Key Laboratory of Diabetes, The Third Hospital of Nanchang, Nanchang city, People's Republic of China
| | - L-N Wang
- Department of Endocrinology and Metabolism, Nanchang Key Laboratory of Diabetes, The Third Hospital of Nanchang, Nanchang city, People's Republic of China
| | - Z Yang
- Department of Endocrinology and Metabolism, Nanchang Key Laboratory of Diabetes, The Third Hospital of Nanchang, Nanchang city, People's Republic of China
| | - P Tu
- Department of Endocrinology and Metabolism, Nanchang Key Laboratory of Diabetes, The Third Hospital of Nanchang, Nanchang city, People's Republic of China
| |
Collapse
|
27
|
Akyol M, Erol MK, Ozdemir O, Coban DT, Bilgin AB, Sari ES, Turkoglu EB. A novel mutation of sgk-1 gene in central serous chorioretinopathy. Int J Ophthalmol 2015; 8:23-8. [PMID: 25709902 DOI: 10.3980/j.issn.2222-3959.2015.01.04] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2014] [Accepted: 09/12/2014] [Indexed: 11/02/2022] Open
Abstract
AIM To investigate the association of serum glucocorticoid kinase gene-1 (SGK-1) DNA variants with chronic central serous chorioretinopathy (CSC). METHODS We enrolled 32 eyes of 32 patients who were diagnosed with chronic CSC and composed 32 normal eyes as a control group. Peripheral blood was used for DNA extraction and polymerase chain reaction (PCR) amplification. SGK1 gene was sequenced by using BigDye(®) Terminator v3.1 cycle sequencing KIT (Applied Biosystems, Foster City, CA, USA). The SGK1 gene and its variants were investigated in CSC patient group and control group. RESULTS We identified a new polymorphism M32V in two person in the patient group (Minor allele frequency (MAF)=0.009) on the region of 1-60 amino acids. The rs1057293 was located in the encoder region of the SGK 1 gene but not associated with CSC (P=0.68). An intrinsic rs1743966 is also not associated (P=0.28). CONCLUSIONS The new polymorphism M32V is located on the region of 1-60 amino acids which is necessary for localization to the mitochondria in CSC patient. This mutation is probably important for the energy metabolism and plays an important role in the cellular response to hyperosmotic stress and other stress stimuli. Both rs1057293 and rs1743966 are not associated with CSC.
Collapse
Affiliation(s)
- Mahmut Akyol
- Human Gene and Cell Therapy Centre, Akdeniz University Faculty of Medicine, 07070, Antalya, Turkey
| | - Muhammet Kazım Erol
- Department of Ophthalmology, Antalya Education and Research Hospital, Antalya 07125, Turkey
| | - Ozdemir Ozdemir
- Department of Ophthalmology, Zekai Tahir Burak Women's Health Education and Research Hospital, Ankara 06100, Turkey
| | - Deniz Turgut Coban
- Department of Ophthalmology, Antalya Education and Research Hospital, Antalya 07125, Turkey
| | - Ahmet Burak Bilgin
- Department of Ophthalmology, Akdeniz University Faculty of Medicine, Antalya 07070, Turkey
| | - Esin Sogutlu Sari
- Department of Ophthalmology, Balıkesir University Faculty of Medicine, Balıkesir 10145, Turkey
| | - Elif Betul Turkoglu
- Department of Ophthalmology, Akdeniz University Faculty of Medicine, Antalya 07070, Turkey
| |
Collapse
|
28
|
Ferrelli F, Pastore D, Capuani B, Lombardo MF, Blot-Chabaud M, Coppola A, Basello K, Galli A, Donadel G, Romano M, Caratelli S, Pacifici F, Arriga R, Di Daniele N, Sbraccia P, Sconocchia G, Bellia A, Tesauro M, Federici M, Della-Morte D, Lauro D. Serum glucocorticoid inducible kinase (SGK)-1 protects endothelial cells against oxidative stress and apoptosis induced by hyperglycaemia. Acta Diabetol 2015; 52:55-64. [PMID: 24961472 DOI: 10.1007/s00592-014-0600-4] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/07/2014] [Accepted: 05/17/2014] [Indexed: 02/07/2023]
Abstract
Diabetic hyperglycaemia causes endothelial dysfunction mainly by impairing endothelial nitric oxide (NO) production. Moreover, hyperglycaemia activates several noxious cellular pathways including apoptosis, increase in reactive oxygen species (ROS) levels and diminishing Na(+)-K(+) ATPase activity which exacerbate vascular damage. Serum glucocorticoid kinase (SGK)-1, a member of the serine/threonine kinases, plays a pivotal role in regulating NO production through inducible NO synthase activation and other cellular mechanisms. Therefore, in this study, we aimed to investigate the protective role of SGK-1 against hyperglycaemia in human umbilical endothelial cells (HUVECs). We used retrovirus to infect HUVECs with either SGK-1, SGK-1Δ60 (lacking of the N-60 amino acids-increase SGK-1 activity) or SGK-1Δ60KD (kinase-dead constructs). We tested our hypothesis in vitro after high glucose and glucosamine incubation. Increase in SGK-1 expression and activity (SGK-1Δ60) resulted in higher production of NO, inhibition of ROS synthesis and lower apoptosis in endothelial cell after either hyperglycaemia or glucosamine treatments. Moreover, in this study, we showed increased GLUT-1 membrane translocation and Na(+)-K(+) ATPase activity in cell infected with SGK-1Δ60 construct. These results suggest that as in endothelial cells, an increased SGK-1 activity and expression reduces oxidative stress, improves cell survival and restores insulin-mediated NO production after different noxae stimuli. Therefore, SGK-1 may represent a specific target to further develop novel therapeutic options against diabetic vascular disease.
Collapse
Affiliation(s)
- Francesca Ferrelli
- Laboratory of Molecular Medicine, Department of Systems Medicine, University of Rome Tor Vergata, Montpellier, 1 Street, 00133, Rome, Italy
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
29
|
Lu X, Li M, Zhou L, Jiang H, Wang H, Chen J. Urinary serum- and glucocorticoid-inducible kinase SGK1 reflects renal injury in patients with immunoglobulin A nephropathy. Nephrology (Carlton) 2015; 19:307-17. [PMID: 24602173 DOI: 10.1111/nep.12225] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
AIM Serum- and glucocorticoid-inducible kinase SGK1 functions as an important regulator of transepithelial sodium transport by activating epithelial sodium channel in renal tubules. Considerable evidence demonstrated that SGK1 was associated with hypertension and fibrosing diseases, such as diabetic nephropathy and glomerulonephritis. The present study was performed to evaluate the role of SGK1 played in immunoglobulin A (IgA) nephropathy. METHODS Seventy-six patients of biopsy-proven IgA nephropathy and 33 healthy volunteers were enrolled in this study. All patients and healthy volunteers' urinary and serum samples were tested for SGK1 expression by indirect enzyme-linked immunosorbent assay. Meanwhile all patients' renal tissues were semi-quantified for SGK1 expression by immunohistochemistry assay. The relationships between SGK1 expressions and clinical or pathological parameters were also assessed. RESULTS SGK1 expression was upregulated in urine and renal tubules in patients of Oxford classification T1 and T2, whereas its expression in serum did not increase significantly. Relationship analysis indicated that urinary and tissue SGK1 expressions were associated with heavy proteinuria and renal insufficiency in patients with IgA nephropathy. On the other hand, RAS blockades would reduce the SGK1 levels both in urine and renal tissues. CONCLUSION These results suggested that urinary SGK1 should be a good indicator of tubulointerstitial damage in patients of IgA nephropathy. SGK1 expressions in urine and renal tissues were associated with the activity of renin-angiotensin-aldosterone system.
Collapse
Affiliation(s)
- Xiaoqian Lu
- Kidney Disease Center, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, China; Kidney Disease Immunology Laboratory, The Third Grade Laboratory, State Administration of Traditional Chinese Medicine of China, Hangzhou, China; Key Laboratory of Multiple Organ Transplantation, Ministry of Health of China, Hangzhou, China; Key Laboratory of Nephropathy of Zhejiang Province, Hangzhou, China
| | | | | | | | | | | |
Collapse
|
30
|
Armando I, Villar VAM, Jose PA. Genomics and Pharmacogenomics of Salt-sensitive Hypertension. Curr Hypertens Rev 2015; 11:49-56. [PMID: 26028245] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2015] [Revised: 04/08/2015] [Accepted: 04/24/2015] [Indexed: 06/04/2023]
Abstract
Salt sensitivity is estimated to be present in 51% of the hypertensive and 26% of the normotensive populations. The individual blood pressure response to salt is heterogeneous and possibly related to inherited susceptibility. Although the mechanisms underlying salt sensitivity are complex and not well understood, genetics can help to determine the blood response to salt intake. So far only a few genes have been found to be associated with salt-sensitive hypertension using candidate gene association studies. The kidney is critical to overall fluid and electrolyte balance and long-term regulation of blood pressure. Thus, the pathogenesis of salt sensitivity must involve a derangement in renal NaCl handling: an inability to decrease renal sodium transport and increase sodium excretion in the face of an increase in NaCl load that could be caused by aberrant counter-regulatory natriuretic/antinatriuretic pathways. We review here the literature regarding the gene variants associated with salt-sensitive hypertension and how the presence of these gene variants influences the response to antihypertensive therapy.
Collapse
Affiliation(s)
- Ines Armando
- Division of Nephrology, Department of Medicine, and Department of Physiology, University of Maryland School of Medicine, Baltimore, MD 21201, USA.
| | | | | |
Collapse
|
31
|
Armando I, Villar VAM, Jose PA. Genomics and pharmacogenomics of salt-sensitive hypertension Minireview. Curr Hypertens Rev 2015; 11:49-56. [PMID: 28392754 PMCID: PMC4875776] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/07/2023]
Abstract
Salt sensitivity is estimated to be present in 51% of the hypertensive and 26% of the normotensive populations. The individual blood pressure response to salt is heterogeneous and possibly related to inherited susceptibility. Although the mechanisms underlying salt sensitivity are complex and not well understood, genetics can help to determine the blood response to salt intake. So far only a few genes have been found to be associated with salt-sensitive hypertension using candidate gene association studies. The kidney is critical to overall fluid and electrolyte balance and long-term regulation of blood pressure. Thus, the pathogenesis of salt sensitivity must involve a derangement in renal NaCl handling: an inability to decrease renal sodium transport and increase sodium excretion in the face of an increase in NaCl load that could be caused by aberrant counter-regulatory natriuretic/antinatriuretic pathways. We review here the literature regarding the gene variants associated with salt-sensitive hypertension and how the presence of these gene variants influences the response to antihypertensive therapy.
Collapse
Affiliation(s)
- Ines Armando
- Division of Nephrology, Department of Medicine, and Department of Physiology, University of Maryland School of Medicine, Baltimore, MD 21201, USA
| | - Van Anthony M Villar
- Division of Nephrology, Department of Medicine, and Department of Physiology, University of Maryland School of Medicine, Baltimore, MD 21201, USA
| | - Pedro A Jose
- Division of Nephrology, Department of Medicine, and Department of Physiology, University of Maryland School of Medicine, Baltimore, MD 21201, USA
| |
Collapse
|
32
|
Kusche-Vihrog K, Schmitz B, Brand E. Salt controls endothelial and vascular phenotype. Pflugers Arch 2014; 467:499-512. [DOI: 10.1007/s00424-014-1657-1] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2014] [Revised: 11/11/2014] [Accepted: 11/14/2014] [Indexed: 01/11/2023]
|
33
|
Sundstrom B, Johansson I, Rantapaa-Dahlqvist S. Interaction between dietary sodium and smoking increases the risk for rheumatoid arthritis: results from a nested case-control study. Rheumatology (Oxford) 2014; 54:487-93. [DOI: 10.1093/rheumatology/keu330] [Citation(s) in RCA: 90] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
|
34
|
Moes AD, Hesselink DA, Zietse R, van Schaik RHN, van Gelder T, Hoorn EJ. Calcineurin inhibitors and hypertension: a role for pharmacogenetics? Pharmacogenomics 2014; 15:1243-51. [DOI: 10.2217/pgs.14.87] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023] Open
Abstract
Hypertension is a common side effect of calcineurin inhibitors (CNIs), which are drugs used to prevent rejection after transplantation. Hypertension after kidney transplantation has been associated with earlier graft failure and higher cardiovascular mortality in the recipient. Recent data indicate that enzymes and transporters involved in CNI pharmacokinetics and pharmacodynamics, including CYP3A5, ABCB1, WNK4 and SPAK, are also associated with salt-sensitive hypertension. These insights raise the question whether polymorphisms in the genes encoding these proteins increase the risk of CNI-induced hypertension. Predicting who is at risk for CNI-induced hypertension may be useful for when selecting specific interventions, including dietary salt restriction, thiazide diuretics or a CNI-free immunosuppressive regimen. This review aims to explore the pharmacogenetics of CNI-induced hypertension, highlighting the knowns and unknowns.
Collapse
Affiliation(s)
- Arthur D Moes
- Department of Internal Medicine, Nephrology & Transplantation, Erasmus Medical Center, PO Box 2040 – Room H-438, 3000 CA Rotterdam, The Netherlands
| | - Dennis A Hesselink
- Department of Internal Medicine, Nephrology & Transplantation, Erasmus Medical Center, PO Box 2040 – Room H-438, 3000 CA Rotterdam, The Netherlands
| | - Robert Zietse
- Department of Internal Medicine, Nephrology & Transplantation, Erasmus Medical Center, PO Box 2040 – Room H-438, 3000 CA Rotterdam, The Netherlands
| | - Ron HN van Schaik
- Department of Clinical Chemistry, Erasmus Medical Center, Rotterdam, The Netherlands
| | - Teun van Gelder
- Department of Internal Medicine, Nephrology & Transplantation, Erasmus Medical Center, PO Box 2040 – Room H-438, 3000 CA Rotterdam, The Netherlands
- Department of Hospital Pharmacy, Clinical Pharmacology Unit, Erasmus Medical Center, Rotterdam, The Netherlands
| | - Ewout J Hoorn
- Department of Internal Medicine, Nephrology & Transplantation, Erasmus Medical Center, PO Box 2040 – Room H-438, 3000 CA Rotterdam, The Netherlands
| |
Collapse
|
35
|
Li C, Yang X, He J, Hixson JE, Gu D, Rao DC, Shimmin LC, Huang J, Gu CC, Chen J, Li J, Kelly TN. A gene-based analysis of variants in the serum/glucocorticoid regulated kinase (SGK) genes with blood pressure responses to sodium intake: the GenSalt Study. PLoS One 2014; 9:e98432. [PMID: 24878720 PMCID: PMC4039502 DOI: 10.1371/journal.pone.0098432] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2014] [Accepted: 04/22/2014] [Indexed: 11/18/2022] Open
Abstract
BACKGROUND Serum and glucocorticoid regulated kinase (SGK) plays a critical role in the regulation of renal sodium transport. We examined the association between SGK genes and salt sensitivity of blood pressure (BP) using single-marker and gene-based association analysis. METHODS A 7-day low-sodium (51.3 mmol sodium/day) followed by a 7-day high-sodium intervention (307.8 mmol sodium/day) was conducted among 1,906 Chinese participants. BP measurements were obtained at baseline and each intervention using a random-zero sphygmomanometer. Additive associations between each SNP and salt-sensitivity phenotypes were assessed using a mixed linear regression model to account for family dependencies. Gene-based analyses were conducted using the truncated p-value method. The Bonferroni-method was used to adjust for multiple testing in all analyses. RESULTS In single-marker association analyses, SGK1 marker rs2758151 was significantly associated with diastolic BP (DBP) response to high-sodium intervention (P = 0.0010). DBP responses (95% confidence interval) to high-sodium intervention for genotypes C/C, C/T, and T/T were 2.04 (1.57 to 2.52), 1.79 (1.42 to 2.16), and 0.85 (0.30 to 1.41) mmHg, respectively. Similar trends were observed for SBP and MAP responses although not significant (P = 0.15 and 0.0026, respectively). In addition, gene-based analyses demonstrated significant associations between SGK1 and SBP, DBP and MAP responses to high sodium intervention (P = 0.0002, 0.0076, and 0.00001, respectively). Neither SGK2 nor SGK3 were associated with the salt-sensitivity phenotypes in single-maker or gene-based analyses. CONCLUSIONS The current study identified association of the SGK1 gene and BP salt-sensitivity in the Han Chinese population. Further studies are warranted to identify causal SGK1 gene variants.
Collapse
Affiliation(s)
- Changwei Li
- Department of Epidemiology, Tulane University School of Public Health and Tropical Medicine, New Orleans, Louisiana, United States of America
| | - Xueli Yang
- State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center of Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Jiang He
- Department of Epidemiology, Tulane University School of Public Health and Tropical Medicine, New Orleans, Louisiana, United States of America
- Department of Medicine, Tulane University School of Medicine, New Orleans, Louisiana, United States of America
| | - James E. Hixson
- Department of Epidemiology, Human Genetics and Environmental Sciences, University of Texas School of Public Health, Houston, Texas, United States of America
| | - Dongfeng Gu
- State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center of Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Dabeeru C. Rao
- Division of Biostatistics, Washington University School of Medicine, St. Louis, Missouri, United States of America
| | - Lawrence C. Shimmin
- Department of Epidemiology, Human Genetics and Environmental Sciences, University of Texas School of Public Health, Houston, Texas, United States of America
| | - Jianfeng Huang
- State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center of Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Charles C. Gu
- Division of Biostatistics, Washington University School of Medicine, St. Louis, Missouri, United States of America
| | - Jichun Chen
- State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center of Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Jianxin Li
- State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center of Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Tanika N. Kelly
- Department of Epidemiology, Tulane University School of Public Health and Tropical Medicine, New Orleans, Louisiana, United States of America
| |
Collapse
|
36
|
Lang F, Stournaras C, Alesutan I. Regulation of transport across cell membranes by the serum- and glucocorticoid-inducible kinase SGK1. Mol Membr Biol 2014; 31:29-36. [PMID: 24417516 DOI: 10.3109/09687688.2013.874598] [Citation(s) in RCA: 52] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
The serum- and glucocorticoid-inducible kinase 1 (SGK1) is genomically upregulated by cell stress including energy depletion and hyperosmotic shock as well as a variety of hormones including glucocorticoids, mineralocorticoids and TGFβ. SGK1 is activated by insulin, growth factors and oxidative stress via phosphatidylinositide-3-kinase, 3-phosphoinositide-dependent kinase PDK1 and mTOR. SGK1 is a powerful stimulator of Na(+)/K(+)-ATPase, carriers (e.g., NCC, NKCC, NHE1, NHE3, SGLT1, several amino acid transporters) and ion channels (e.g., ENaC, SCN5A, TRPV4-6, ORAI1/STIM1, ROMK, KCNE1/KCNQ1, GluR6, CFTR). Mechanisms employed by SGK1 in transport regulation include direct phosphorylation of target transport proteins, phosphorylation and thus activation of other transport regulating kinases, stabilization of membrane proteins by phosphorylation and thus inactivation of the ubiquitin ligase NEDD4-2, as well as stimulation of transport protein expression by upregulation transcription factors (e.g., nuclear factor kappa-B [NFκB]) and by fostering of protein translation. SGK1 sensitivity of pump, carrier and channel activities participate in the regulation of epithelial transport, cardiac and neuronal excitability, degranulation, platelet function, migration, cell proliferation and apoptosis. SGK1-sensitive functions do not require the presence of SGK1 but are markedly upregulated by SGK1. Accordingly, the phenotype of SGK1 knockout mice is mild. The mice are, however, less sensitive to excessive activation of transport by glucocorticoids, mineralocorticoids, insulin and inflammation. Moreover, excessive SGK1 activity contributes to the pathophysiology of hypertension, obesity, diabetes, thrombosis, stroke, inflammation, autoimmune disease, fibrosis and tumor growth.
Collapse
Affiliation(s)
- Florian Lang
- Department of Physiology, University of Tübingen , Germany and
| | | | | |
Collapse
|
37
|
Warnock DG, Kusche-Vihrog K, Tarjus A, Sheng S, Oberleithner H, Kleyman TR, Jaisser F. Blood pressure and amiloride-sensitive sodium channels in vascular and renal cells. Nat Rev Nephrol 2014; 10:146-57. [PMID: 24419567 DOI: 10.1038/nrneph.2013.275] [Citation(s) in RCA: 84] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Sodium transport in the distal nephron is mediated by epithelial sodium channel activity. Proteolytic processing of external domains and inhibition with increased sodium concentrations are important regulatory features of epithelial sodium channel complexes expressed in the distal nephron. By contrast, sodium channels expressed in the vascular system are activated by increased external sodium concentrations, which results in changes in the mechanical properties and function of endothelial cells. Mechanosensitivity and shear stress affect both epithelial and vascular sodium channel activity. Guyton's hypothesis stated that blood pressure control is critically dependent on vascular tone and fluid handling by the kidney. The synergistic effects, and complementary regulation, of the epithelial and vascular systems are consistent with the Guytonian model of volume and blood pressure regulation, and probably reflect sequential evolution of the two systems. The integration of vascular tone, renal perfusion and regulation of renal sodium reabsorption is the central underpinning of the Guytonian model. In this Review, we focus on the expression and regulation of sodium channels, and we outline the emerging evidence that describes the central role of amiloride-sensitive sodium channels in the efferent (vascular) and afferent (epithelial) arms of this homeostatic system.
Collapse
Affiliation(s)
- David G Warnock
- Division of Nephrology, Department of Medicine, University of Alabama at Birmingham, 1720 2nd Avenue South, Birmingham, AL 34294-0007, USA
| | - Kristina Kusche-Vihrog
- Institut für Physiologie II, Westfälische Wilhelms Universität, Robert-Koch-Straße 27, 48149 Münster, Germany
| | - Antoine Tarjus
- INSERM U872 Team 1, Centre de Recherche des Cordeliers, Université René Descartes, Université Pierre et Marie Curie, 15 rue de l'Ecole de Médecine, 75006 Paris, France
| | - Shaohu Sheng
- Renal and Electrolyte Division, Department of Medicine, University of Pittsburgh, 3550 Terrace Street, Pittsburgh, PA 15261, USA
| | - Hans Oberleithner
- Institut für Physiologie II, Westfälische Wilhelms Universität, Robert-Koch-Straße 27, 48149 Münster, Germany
| | - Thomas R Kleyman
- Renal and Electrolyte Division, Department of Medicine, University of Pittsburgh, 3550 Terrace Street, Pittsburgh, PA 15261, USA
| | - Frederic Jaisser
- INSERM U872 Team 1, Centre de Recherche des Cordeliers, Université René Descartes, Université Pierre et Marie Curie, 15 rue de l'Ecole de Médecine, 75006 Paris, France
| |
Collapse
|
38
|
|
39
|
Abstract
PURPOSE OF REVIEW One-third of the world's population has hypertension and it is responsible for almost 50% of deaths from stroke or coronary heart disease. These statistics do not distinguish salt-sensitive from salt-resistant hypertension or include normotensives who are salt-sensitive even though salt sensitivity, independent of blood pressure, is a risk factor for cardiovascular and other diseases, including cancer. This review describes new personalized diagnostic tools for salt sensitivity. RECENT FINDINGS The relationship between salt intake and cardiovascular risk is not linear, but rather fits a J-shaped curve relationship. Thus, a low-salt diet may not be beneficial to everyone and may paradoxically increase blood pressure in some individuals. Current surrogate markers of salt sensitivity are not adequately sensitive or specific. Tests in the urine that could be surrogate markers of salt sensitivity with a quick turn-around time include renal proximal tubule cells, exosomes, and microRNA shed in the urine. SUMMARY Accurate testing of salt sensitivity is not only laborious but also expensive, and with low patient compliance. Patients who have normal blood pressure but are salt-sensitive cannot be diagnosed in an office setting and there are no laboratory tests for salt sensitivity. Urinary surrogate markers for salt sensitivity are being developed.
Collapse
|
40
|
Preference for salt contributes to sympathovagal imbalance in the genesis of prehypertension. Eur J Clin Nutr 2013; 67:586-91. [DOI: 10.1038/ejcn.2013.64] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
|
41
|
Abstract
The assessment of salt sensitivity of blood pressure is difficult because of the lack of universal consensus on definition. Regardless of the variability in the definition of salt sensitivity, increased salt intake, independent of the actual level of blood pressure, is also a risk factor for cardiovascular morbidity and mortality and kidney disease. A modest reduction in salt intake results in an immediate decrease in blood pressure, with long-term beneficial consequences. However, some have suggested that dietary sodium restriction may not be beneficial to everyone. Thus, there is a need to distinguish salt-sensitive from salt-resistant individuals, but it has been difficult to do so with phenotypic studies. Therefore, there is a need to determine the genes that are involved in salt sensitivity. This review focuses on genes associated with salt sensitivity, with emphasis on the variants associated with salt sensitivity in humans that are not due to monogenic causes. Special emphasis is given to gene variants associated with salt sensitivity whose protein products interfere with cell function and increase blood pressure in transgenic mice.
Collapse
Affiliation(s)
- Hironobu Sanada
- Division of Health Science Research, Fukushima Welfare Federation of Agricultural Cooperatives, Fukushima, Japan.
| | | | | |
Collapse
|