1
|
Albritton CF, Demirci M, Neikirk K, Ertuglu LA, Ishimwe JA, Mutchler AL, Sheng Q, Laffer CL, Wanjalla CN, Ahmed T, Haynes AP, Saleem M, Beasley HK, Marshall AG, Vue Z, Ikizler AT, Kleyman TR, Kon V, Hinton A, Kirabo A. Myeloid Cell Glucocorticoid, Not Mineralocorticoid Receptor Signaling, Contributes to Salt-Sensitive Hypertension in Humans via Cortisol. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.06.10.598374. [PMID: 38915603 PMCID: PMC11195113 DOI: 10.1101/2024.06.10.598374] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/26/2024]
Abstract
BACKGROUND Salt sensitivity of blood pressure (SSBP) is an independent risk factor for cardiovascular morbidity and mortality, yet the etiology is poorly understood. We previously found that serum/glucocorticoid-regulated kinase 1 (SGK1) and epoxyeicosatrienoic acids (EETs) regulate epithelial sodium channel (ENaC)-dependent sodium entry into monocyte-derived antigen-presenting cells (APCs) and activation of NADPH oxidase, leading to the formation of isolevuglandins (IsoLGs) in SSBP. Whereas aldosterone via the mineralocorticoid receptor (MR) activates SGK1 leading to hypertension, our past findings indicate that levels of plasma aldosterone do not correlate with SSBP, and there is little to no MR expression in APCs. Thus, we hypothesized that cortisol acting via the glucocorticoid receptor (GR), not the MR in APCs mediates SGK1 actions to induce SSBP. METHODS We performed cellular indexing of transcriptomes and epitopes by sequencing (CITE-Seq) analysis on peripheral blood mononuclear cells of humans rigorously phenotyped for SSBP using an inpatient salt loading/depletion protocol to determine expression of MR, GR, and SGK1 in immune cells. In additional experiments, we performed bulk transcriptomic analysis on isolated human monocytes following in vitro treatment with high salt from a separate cohort. We then measured urine and plasma cortisol, cortisone, renin, and aldosterone. Subsequently, we measured the association of these hormones with changes in systolic, diastolic, mean arterial pressure and pulse pressure as well as immune cell activation via IsoLG formation. RESULTS We found that myeloid APCs predominantly express the GR and SGK1 with no expression of the MR. Expression of the GR in APCs increased after salt loading and decreased with salt depletion in salt-sensitive but not salt-resistant people and was associated with increased expression of SGK1. Moreover, we found that plasma and urine cortisol/cortisone but not aldosterone/renin correlated with SSBP and APCs activation via IsoLGs. We also found that cortisol negatively correlates with EETs. CONCLUSION Our findings suggest that renal cortisol signaling via the GR but not the MR in APCs contributes to SSBP via cortisol. Urine and plasma cortisol may provide an important currently unavailable feasible diagnostic tool for SSBP. Moreover, cortisol-GR-SGK1-ENaC signaling pathway may provide treatment options for SSBP.
Collapse
Affiliation(s)
- Claude F. Albritton
- Department of Medicine, Division of Clinical Pharmacology, Vanderbilt University Medical Center, Nashville, TN 37212-8802, USA
- Department of Biomedical Sciences, School of Graduate Studies, Meharry Medical College, Nashville, TN 37208-3501, USA
| | - Mert Demirci
- Division of Nephrology and Hypertension, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, United States
| | - Kit Neikirk
- Department of Molecular Physiology & Biophysics, Vanderbilt University, Nashville, TN 37212-8802, USA
| | - Lale A. Ertuglu
- Division of Nephrology and Hypertension, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, United States
| | - Jeanne A Ishimwe
- Department of Medicine, Division of Clinical Pharmacology, Vanderbilt University Medical Center, Nashville, TN 37212-8802, USA
| | - Ashley L Mutchler
- Department of Medicine, Division of Clinical Pharmacology, Vanderbilt University Medical Center, Nashville, TN 37212-8802, USA
| | - Quanhu Sheng
- Department of Biostatistics, Vanderbilt University Medical Center, Nashville, TN 37212-8802, USA
| | - Cheryl L Laffer
- Department of Medicine, Division of Clinical Pharmacology, Vanderbilt University Medical Center, Nashville, TN 37212-8802, USA
| | - Celestine N. Wanjalla
- Department of Medicine, Division of Clinical Pharmacology, Vanderbilt University Medical Center, Nashville, TN 37212-8802, USA
| | - Taseer Ahmed
- Department of Medicine, Division of Clinical Pharmacology, Vanderbilt University Medical Center, Nashville, TN 37212-8802, USA
- Department of Pharmacology, College of Pharmacy, University of Sargodha, University Road, Sargodha, Punjab, 40100, Pakistan
| | - Alexandria Porcia Haynes
- Department of Medicine, Division of Clinical Pharmacology, Vanderbilt University Medical Center, Nashville, TN 37212-8802, USA
| | - Mohammad Saleem
- Department of Medicine, Division of Clinical Pharmacology, Vanderbilt University Medical Center, Nashville, TN 37212-8802, USA
| | - Heather K. Beasley
- Department of Molecular Physiology & Biophysics, Vanderbilt University, Nashville, TN 37212-8802, USA
| | - Andrea G. Marshall
- Department of Molecular Physiology & Biophysics, Vanderbilt University, Nashville, TN 37212-8802, USA
| | - Zer Vue
- Department of Molecular Physiology & Biophysics, Vanderbilt University, Nashville, TN 37212-8802, USA
| | - Alp T Ikizler
- Division of Nephrology and Hypertension, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, United States
| | - Thomas R. Kleyman
- Renal-Electrolyte Division, Department of Medicine, University of Pittsburgh, Pittsburgh, PA 15260, USA
| | - Valentina Kon
- Division of Nephrology, Department of Pediatrics, Vanderbilt University Medical Center, Nashville, TN
| | - Antentor Hinton
- Department of Molecular Physiology & Biophysics, Vanderbilt University, Nashville, TN 37212-8802, USA
| | - Annet Kirabo
- Department of Medicine, Division of Clinical Pharmacology, Vanderbilt University Medical Center, Nashville, TN 37212-8802, USA
- Department of Molecular Physiology & Biophysics, Vanderbilt University, Nashville, TN 37212-8802, USA
- Vanderbilt Center for Immunobiology
- Vanderbilt Institute for Infection, Immunology and Inflammation
- Vanderbilt Institute for Global Health
| |
Collapse
|
2
|
Gan J, Shi Y, Zhao R, Li D, Jin H, Wu M, Liu Z, Li X, Xu A, Li Y, Lin Z, Wu F. Adipose c-Jun NH2-terminal kinase promotes angiotensin II-induced and deoxycorticosterone acetate salt-induced hypertension and vascular dysfunction by inhibition of adiponectin production and activation of SGK1 in mice. J Hypertens 2024; 42:856-872. [PMID: 38164960 DOI: 10.1097/hjh.0000000000003649] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2024]
Abstract
BACKGROUND Adipose c-Jun NH2-terminal kinase 1/2 (JNK1/2) is a central mediator involved in the development of obesity and its complications. However, the roles of adipose JNK1/2 in hypertension remain elusive. Here we explored the role of adipose JNK1/2 in hypertension. METHODS AND RESULTS The roles of adipose JNK1/2 in hypertension were investigated by evaluating the impact of adipose JNK1/2 inactivation in both angiotensin II (Ang II)-induced and deoxycorticosterone acetate (DOCA) salt-induced hypertensive mice. Specific inactivation of JNK1/2 in adipocytes significantly alleviates Ang II-induced and DOCA salt-induced hypertension and target organ damage in mice. Interestingly, such beneficial effects are also observed in hypertensive mice after oral administration of JNK1/2 inhibitor SP600125. Mechanistically, adipose JNK1/2 acts on adipocytes to reduce the production of adiponectin (APN), then leads to promote serum and glucocorticoid-regulated kinase 1 (SGK1) phosphorylation and increases epithelial Na + channel α-subunit (ENaCα) expression in both renal cells and adipocytes, respectively, finally exacerbates Na + retention. In addition, chronic treatment of recombinant mouse APN significantly augments the beneficial effects of adipose JNK1/2 inactivation in DOCA salt-induced hypertension. By contrast, the blood pressure-lowering effects of adipose JNK1/2 inactivation are abrogated by adenovirus-mediated SGK1 overexpression in Ang II -treated adipose JNK1/2 inactivation mice. CONCLUSION Adipose JNK1/2 promotes hypertension and targets organ impairment via fine-tuning the multiorgan crosstalk among adipose tissue, kidney, and blood vessels.
Collapse
Affiliation(s)
- Jing Gan
- Department of Cardiology, the First Affiliated Hospital of Wenzhou Medical University
| | - Yaru Shi
- School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou
- Department of Pharmacy, the Sixth Affiliated Hospital of Wenzhou Medical University, Lishui
| | - Ruyi Zhao
- School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou
| | - Dan Li
- School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou
- Department of clinical pharmacy, the Forth People's Hospital of Liaocheng, Liaocheng
| | - Hua Jin
- School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou
| | - Maolan Wu
- School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou
| | - Zhen Liu
- School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou
| | - Xiaokun Li
- School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou
| | - Aimin Xu
- State Key Laboratory of Pharmaceutical Biotechnology, the University of Hong Kong, Hong Kong
| | - Yulin Li
- Beijing Institute of Heart, Lung, and Blood Vessel Diseases, Anzhen Hospital of Capital Medical University, Beijing
| | - Zhuofeng Lin
- Department of Cardiology, the First Affiliated Hospital of Wenzhou Medical University
- The laboratory of Animal Center, Wenzhou Medical University, Wenzhou, China
| | - Fan Wu
- School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou
| |
Collapse
|
3
|
Huang YM, Li TX, Li SY, Zhu XR, Li Y, Liu DY, Li WM, Yang LQ, Liu KS, Liu C. Glucocorticoids ameliorate cardiorenal syndrome through the NPR1/SGK1 pathway in natriuretic peptide receptor A‑heterozygous mice. Exp Ther Med 2023; 26:374. [PMID: 37415837 PMCID: PMC10320660 DOI: 10.3892/etm.2023.12073] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2022] [Accepted: 04/28/2023] [Indexed: 07/08/2023] Open
Abstract
Natriuretic peptides, which are produced by the heart, bind to natriuretic peptide receptor A (NPR1 encoded by natriuretic peptide receptor 1 gene) and cause vasodilation and natriuresis. Thus, they serve an important role in regulating blood pressure. In the present study, microinjection of CRISPR associated protein 9/single guide RNA into fertilized C57BL/6N mouse eggs was performed to generate filial generation zero (F0) Npr1 knockout homozygous mice (Npr1-/-). F0 mice mated with wild-type (WT) mice to obtain F1 Npr1 knockout heterozygous mice with stable heredity (Npr1+/-). F1 self-hybridization was used to expand the population of heterozygous mice (Npr1+/-). The present study performed echocardiography to investigate the impact of NPR1 gene knockdown on cardiac function. Compared with those in the WT group (C57BL/6N male mice), the left ventricular ejection fraction, myocardial contractility and renal sodium and potassium excretion and creatinine-clearance rates were decreased, indicating that Npr1 knockdown induced cardiac and renal dysfunction. In addition, expression of serum glucocorticoid-regulated kinase 1 (SGK1) increased significantly compared with that in WT mice. However, glucocorticoids (dexamethasone) upregulated NPR1 and inhibited SGK1 and alleviated cardiac and renal dysfunction caused by Npr1 gene heterozygosity. SGK1 inhibitor GSK650394 ameliorate cardiorenal syndrome by suppressing SGK1. Briefly, glucocorticoids inhibited SGK1 by upregulating NPR1, thereby ameliorating cardiorenal impairment caused by Npr1 gene heterozygosity. The present findings provided novel insight into the understanding of cardiorenal syndrome and suggested that glucocorticoids targeting the NPR1/SGK1 pathway may be a potential therapeutic target to treat cardiorenal syndrome.
Collapse
Affiliation(s)
- Yao-Meng Huang
- The First Cardiology Division, The First Hospital of Hebei Medical University, Shijiazhuang, Hebei 050030, P.R. China
| | - Tong-Xin Li
- The First Cardiology Division, The First Hospital of Hebei Medical University, Shijiazhuang, Hebei 050030, P.R. China
| | - Shu-Yu Li
- The First Cardiology Division, The First Hospital of Hebei Medical University, Shijiazhuang, Hebei 050030, P.R. China
- Department of Cardiology, Affiliated Hospital of North China University of Science and Technology, Tangshan, Hebei 063210, P.R. China
| | - Xiao-Ran Zhu
- The First Cardiology Division, The First Hospital of Hebei Medical University, Shijiazhuang, Hebei 050030, P.R. China
- Department of Pharmacy, Hebei General Hospital, Shijiazhuang, Hebei 050057, P.R. China
| | - Ying Li
- The First Cardiology Division, The First Hospital of Hebei Medical University, Shijiazhuang, Hebei 050030, P.R. China
| | - Dang-Yang Liu
- The First Cardiology Division, The First Hospital of Hebei Medical University, Shijiazhuang, Hebei 050030, P.R. China
| | - Wei-Min Li
- The First Cardiology Division, The First Hospital of Hebei Medical University, Shijiazhuang, Hebei 050030, P.R. China
| | - Lin-Quan Yang
- Department of Pharmacy, Hebei General Hospital, Shijiazhuang, Hebei 050057, P.R. China
| | - Kun-Shen Liu
- The First Cardiology Division, The First Hospital of Hebei Medical University, Shijiazhuang, Hebei 050030, P.R. China
| | - Chao Liu
- The First Cardiology Division, The First Hospital of Hebei Medical University, Shijiazhuang, Hebei 050030, P.R. China
| |
Collapse
|
4
|
Le QD, Duong VA, Lee SH, Nguyen TTL, Maeng HJ. Bioanalytical method validation, biopharmaceutical and pharmacokinetic evaluation of GSK-650394, a serum- and glucocorticoid-regulated kinase 1 inhibitor. ARAB J CHEM 2022. [DOI: 10.1016/j.arabjc.2022.104462] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022] Open
|
5
|
Abstract
The Epithelial Na+ Channel, ENaC, comprised of 3 subunits (αβγ, or sometimes δβγENaC), plays a critical role in regulating salt and fluid homeostasis in the body. It regulates fluid reabsorption into the blood stream from the kidney to control blood volume and pressure, fluid absorption in the lung to control alveolar fluid clearance at birth and maintenance of normal airway surface liquid throughout life, and fluid absorption in the distal colon and other epithelial tissues. Moreover, recent studies have also revealed a role for sodium movement via ENaC in nonepithelial cells/tissues, such as endothelial cells in blood vessels and neurons. Over the past 25 years, major advances have been made in our understanding of ENaC structure, function, regulation, and role in human disease. These include the recently solved three-dimensional structure of ENaC, ENaC function in various tissues, and mutations in ENaC that cause a hereditary form of hypertension (Liddle syndrome), salt-wasting hypotension (PHA1), or polymorphism in ENaC that contributes to other diseases (such as cystic fibrosis). Moreover, great strides have been made in deciphering the regulation of ENaC by hormones (e.g., the mineralocorticoid aldosterone, glucocorticoids, vasopressin), ions (e.g., Na+ ), proteins (e.g., the ubiquitin-protein ligase NEDD4-2, the kinases SGK1, AKT, AMPK, WNKs & mTORC2, and proteases), and posttranslational modifications [e.g., (de)ubiquitylation, glycosylation, phosphorylation, acetylation, palmitoylation]. Characterization of ENaC structure, function, regulation, and role in human disease, including using animal models, are described in this article, with a special emphasis on recent advances in the field. © 2021 American Physiological Society. Compr Physiol 11:1-29, 2021.
Collapse
Affiliation(s)
- Daniela Rotin
- The Hospital for Sick Children, and The University of Toronto, Toronto, Canada
| | - Olivier Staub
- Department of Biomedical Sciences, University of Lausanne, Lausanne, Switzerland
| |
Collapse
|
6
|
Hill MA, Yang Y, Zhang L, Sun Z, Jia G, Parrish AR, Sowers JR. Insulin resistance, cardiovascular stiffening and cardiovascular disease. Metabolism 2021; 119:154766. [PMID: 33766485 DOI: 10.1016/j.metabol.2021.154766] [Citation(s) in RCA: 280] [Impact Index Per Article: 93.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/22/2020] [Revised: 03/10/2021] [Accepted: 03/17/2021] [Indexed: 12/18/2022]
Abstract
The cardiometabolic syndrome (CMS) and obesity are typically characterized by a state of metabolic insulin resistance. As global and US rates of obesity increase there is an acceleration of the incidence and prevalence of insulin resistance along with associated cardiovascular disease (CVD). Under physiological conditions insulin regulates glucose homeostasis by enhancing glucose disposal in insulin sensitive tissues while also regulating delivery of nutrients through its vasodilation actions on small feed arteries. Specifically, insulin-mediated production of nitric oxide (NO) from the vascular endothelium leads to increased blood flow enhancing disposal of glucose. Typically, insulin resistance is considered as a decrease in sensitivity or responsiveness to the metabolic actions of insulin including insulin-mediated glucose disposal. However, a decreased sensitivity to the normal vascular actions of insulin, especially diminished nitric oxide production, plays an additional important role in the development of CVD in states of insulin resistance. One mechanism by which insulin resistance and attendant hyperinsulinemia promote CVD is via increases in vascular stiffness. Although obesity and insulin resistance are known to be associated with substantial increases in the prevalence of vascular fibrosis and stiffness the mechanisms and mediators that underlie vascular stiffening in insulin resistant states are complex and have only recently begun to be addressed. Current evidence supports the role of increased plasma levels of aldosterone and insulin and attendant reductions in bioavailable NO in the pathogenesis of impaired vascular relaxation and vascular stiffness in the CMS and obesity. Aldosterone and insulin both increase the activity of serum and glucocorticoid kinase 1 (SGK-1) which in turn is a major regulator of vascular and renal sodium (Na+) channel activity.The importance of SGK-1 in the pathogenesis of the CMS is highlighted by observations that gain of function mutations in SGK-1 in humans promotes hypertension, insulin resistance and obesity. In endothelial cells, an increase in Na+ flux contributes to remodeling of the cytoskeleton, reduced NO bioavailability and vascular stiffening. Thus, endothelial SGK-1 may represent a point of convergence for insulin and aldosterone signaling in arterial stiffness associated with obesity and the CMS. This review examines our contemporary understanding of the link between insulin resistance and increased vascular stiffness with emphasis placed on a role for enhanced SGK-1 signaling as a key node in this pathological process.
Collapse
Affiliation(s)
- Michael A Hill
- Dalton Cardiovascular Research Center, University of Missouri, Columbia, MO 65211, USA; Department of Medical Pharmacology and Physiology, University of Missouri School of Medicine, Columbia, MO 65212, USA.
| | - Yan Yang
- Dalton Cardiovascular Research Center, University of Missouri, Columbia, MO 65211, USA
| | - Liping Zhang
- Dalton Cardiovascular Research Center, University of Missouri, Columbia, MO 65211, USA; Department of Medical Pharmacology and Physiology, University of Missouri School of Medicine, Columbia, MO 65212, USA
| | - Zhe Sun
- Dalton Cardiovascular Research Center, University of Missouri, Columbia, MO 65211, USA; Department of Medical Pharmacology and Physiology, University of Missouri School of Medicine, Columbia, MO 65212, USA
| | - Guanghong Jia
- Dalton Cardiovascular Research Center, University of Missouri, Columbia, MO 65211, USA; Diabetes and Cardiovascular Center, University of Missouri School of Medicine, Columbia, MO 65212, USA; Department of Medicine, University of Missouri School of Medicine, Columbia, MO 65212, USA
| | - Alan R Parrish
- Department of Medical Pharmacology and Physiology, University of Missouri School of Medicine, Columbia, MO 65212, USA
| | - James R Sowers
- Dalton Cardiovascular Research Center, University of Missouri, Columbia, MO 65211, USA; Diabetes and Cardiovascular Center, University of Missouri School of Medicine, Columbia, MO 65212, USA; Department of Medical Pharmacology and Physiology, University of Missouri School of Medicine, Columbia, MO 65212, USA; Department of Medicine, University of Missouri School of Medicine, Columbia, MO 65212, USA.
| |
Collapse
|
7
|
Abstract
Cardiovascular diseases are the leading cause of death worldwide. Overweight and obesity are strongly associated with comorbidities such as hypertension and insulin resistance, which collectively contribute to the development of cardiovascular diseases and resultant morbidity and mortality. Forty-two percent of adults in the United States are obese, and a total of 1.9 billion adults worldwide are overweight or obese. These alarming numbers, which continue to climb, represent a major health and economic burden. Adipose tissue is a highly dynamic organ that can be classified based on the cellular composition of different depots and their distinct anatomical localization. Massive expansion and remodeling of adipose tissue during obesity differentially affects specific adipose tissue depots and significantly contributes to vascular dysfunction and cardiovascular diseases. Visceral adipose tissue accumulation results in increased immune cell infiltration and secretion of vasoconstrictor mediators, whereas expansion of subcutaneous adipose tissue is less harmful. Therefore, fat distribution more than overall body weight is a key determinant of the risk for cardiovascular diseases. Thermogenic brown and beige adipose tissue, in contrast to white adipose tissue, is associated with beneficial effects on the vasculature. The relationship between the type of adipose tissue and its influence on vascular function becomes particularly evident in the context of the heterogenous phenotype of perivascular adipose tissue that is strongly location dependent. In this review, we address the abnormal remodeling of specific adipose tissue depots during obesity and how this critically contributes to the development of hypertension, endothelial dysfunction, and vascular stiffness. We also discuss the local and systemic roles of adipose tissue derived secreted factors and increased systemic inflammation during obesity and highlight their detrimental impact on cardiovascular health.
Collapse
Affiliation(s)
- Mascha Koenen
- Laboratory of Molecular Metabolism, The Rockefeller University, New York (M.K., P.C.)
| | - Michael A Hill
- Dalton Cardiovascular Research Center, University of Missouri, Columbia (M.A.H., J.R.S.)
- Department of Medical Pharmacology and Physiology (M.A.H., J.R.S.), University of Missouri School of Medicine, Columbia
| | - Paul Cohen
- Laboratory of Molecular Metabolism, The Rockefeller University, New York (M.K., P.C.)
| | - James R Sowers
- Dalton Cardiovascular Research Center, University of Missouri, Columbia (M.A.H., J.R.S.)
- Department of Medical Pharmacology and Physiology (M.A.H., J.R.S.), University of Missouri School of Medicine, Columbia
- Diabetes and Cardiovascular Center (J.R.S.), University of Missouri School of Medicine, Columbia
- Department of Medicine (J.R.S.), University of Missouri School of Medicine, Columbia
| |
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
|
Gray Z, Tu W, Chertow GM, Bhalla V. Aldosterone sensitivity: an opportunity to explore the pathogenesis of hypertension. Am J Physiol Renal Physiol 2021; 320:F325-F335. [PMID: 33491565 DOI: 10.1152/ajprenal.00415.2020] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Aldosterone sensitivity is defined as an outcome variable for a given circulating level of aldosterone. In basic and translational studies, aldosterone sensitivity has been measured in differential tissue responses, e.g., lower urine sodium and higher urine potassium, as an index of the renal response; in clinical studies, aldosterone sensitivity has been measured in differential blood pressure responses. The concept of aldosterone sensitivity disrupts the conventional wisdom of the renin-angiotensin-aldosterone system and has the potential to uncover novel mechanisms of hypertension. Here, we review basic and translational science studies that uncovered differential renal responses to aldosterone and connect this earlier work to more recent observational studies and randomized trials that have demonstrated differential blood pressure responses for a given level of aldosterone in healthy and hypertensive persons. Black race and older age are associated with higher aldosterone sensitivity and blood pressure. We also discuss gaps in the field and how future basic and clinical studies might inform mechanisms of differential sensitivity.
Collapse
Affiliation(s)
- Zachary Gray
- Stanford Hypertension Center, Stanford University School of Medicine, Stanford, California
| | - Wanzhu Tu
- Department of Biostatistics, Indiana University School of Medicine, Indianapolis, Indiana
| | - Glenn M Chertow
- Stanford Hypertension Center, Stanford University School of Medicine, Stanford, California.,Division of Nephrology, Department of Medicine, Stanford University School of Medicine, Stanford, California
| | - Vivek Bhalla
- Stanford Hypertension Center, Stanford University School of Medicine, Stanford, California.,Division of Nephrology, Department of Medicine, Stanford University School of Medicine, Stanford, California
| |
Collapse
|
10
|
Guerriero I, Monaco G, Coppola V, Orlacchio A. Serum and Glucocorticoid-Inducible Kinase 1 (SGK1) in NSCLC Therapy. Pharmaceuticals (Basel) 2020; 13:ph13110413. [PMID: 33266470 PMCID: PMC7700219 DOI: 10.3390/ph13110413] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2020] [Revised: 11/19/2020] [Accepted: 11/20/2020] [Indexed: 12/21/2022] Open
Abstract
Non-small cell lung cancer (NSCLC) remains the most prevalent and one of the deadliest cancers worldwide. Despite recent success, there is still an urgent need for new therapeutic strategies. It is also becoming increasingly evident that combinatorial approaches are more effective than single modality treatments. This review proposes that the serum and glucocorticoid-inducible kinase 1 (SGK1) may represent an attractive target for therapy of NSCLC. Although ubiquitously expressed, SGK1 deletion in mice causes only mild defects of ion physiology. The frequent overexpression of SGK1 in tumors is likely stress-induced and provides a therapeutic window to spare normal tissues. SGK1 appears to promote oncogenic signaling aimed at preserving the survival and fitness of cancer cells. Most importantly, recent investigations have revealed the ability of SGK1 to skew immune-cell differentiation toward pro-tumorigenic phenotypes. Future studies are needed to fully evaluate the potential of SGK1 as a therapeutic target in combinatorial treatments of NSCLC. However, based on what is currently known, SGK1 inactivation can result in anti-oncogenic effects both on tumor cells and on the immune microenvironment. A first generation of small molecules to inactivate SGK1 has already been already produced.
Collapse
Affiliation(s)
- Ilaria Guerriero
- Biogem Institute for Genetic Research Gaetano Salvatore, Ariano Irpino, 83031 Avellino, Italy; (I.G.); (G.M.)
| | - Gianni Monaco
- Biogem Institute for Genetic Research Gaetano Salvatore, Ariano Irpino, 83031 Avellino, Italy; (I.G.); (G.M.)
| | - Vincenzo Coppola
- Department of Cancer Biology and Genetics, College of Medicine, The Ohio State University, Columbus, OH 43210, USA
- Arthur G. James Comprehensive Cancer Center, The Ohio State University, Columbus, OH 43210, USA
- Correspondence: (V.C.); (A.O.); Tel.: +1-614-688-8038 (V.C.); +1-646-552-0641 (A.O.)
| | - Arturo Orlacchio
- Department of Cancer Biology and Genetics, College of Medicine, The Ohio State University, Columbus, OH 43210, USA
- Arthur G. James Comprehensive Cancer Center, The Ohio State University, Columbus, OH 43210, USA
- Correspondence: (V.C.); (A.O.); Tel.: +1-614-688-8038 (V.C.); +1-646-552-0641 (A.O.)
| |
Collapse
|
11
|
Chen M, Chen Q, Li Y, Yang Z, Taylor EW, Zhao L. A G-quadruplex nanoswitch in the SGK1 promoter regulates isoform expression by K +/Na + balance and resveratrol binding. Biochim Biophys Acta Gen Subj 2020; 1865:129778. [PMID: 33144140 DOI: 10.1016/j.bbagen.2020.129778] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2020] [Revised: 10/05/2020] [Accepted: 10/28/2020] [Indexed: 10/23/2022]
Abstract
BACKGROUND High sodium intake can up-regulate the level of renal serum- and glucocorticoid-inducible kinase-1 (SGK1), which plays a pivotal role in controlling blood pressure via activation of the epithelial sodium channel (ENaC), which can lead to salt-sensitive hypertension. Increased potassium intake, or a vegetarian diet, counteracts salt-sensitive hypertension, but the underlying mechanisms are not fully understood. METHODS Bioinformatics and molecular modeling were used to identify G-quadruplex (G4) and their conformations in the SGK1 promoter. CD spectra and UV melting dynamics were measured to study the stability of G4 as influenced by potassium/sodium balance and resveratrol. RT-PCR and Western blot were employed to study the effects of potassium and resveratrol on the SGK1 isoform expression. RESULTS The SGK1 gene encodes a G4 structure in the proximal upstream of promoter-2; the G4 structure is stabilized by potassium or resveratrol, but destabilized by sodium. Super-physiological levels of sodium stimulate the transcription of all SGK1 isoforms, whereas resveratrol or potassium supplementation inhibits the transcription of iso-2 and iso-3, but not iso-1. CONCLUSIONS Stabilizing the G4 by potassium or resveratrol induces alternative promoter usage and/or pre-mRNA splicing in the transcription of SGK1. GENERAL SIGNIFICANCE Potassium/sodium ion balance or resveratrol binding can act to regulate G4 molecular switches for controlling SGK1 gene expression, thereby presenting a new avenue for drug development.
Collapse
Affiliation(s)
- Mengjie Chen
- Key Laboratory of Ministry of Education for Medicinal Plant Resource and Natural Pharmaceutical Chemistry, College of Life Sciences, Shaanxi Normal University, Xi'an 710119, China
| | - Qi Chen
- Key Laboratory of Ministry of Education for Medicinal Plant Resource and Natural Pharmaceutical Chemistry, College of Life Sciences, Shaanxi Normal University, Xi'an 710119, China
| | - Yirui Li
- Key Laboratory of Ministry of Education for Medicinal Plant Resource and Natural Pharmaceutical Chemistry, College of Life Sciences, Shaanxi Normal University, Xi'an 710119, China
| | - Zhenjun Yang
- School of Pharmaceutical Sciences, Peking University, Beijing 100083, China
| | - Ethan W Taylor
- Department of Chemistry and Biochemistry, University of North Carolina at Greensboro, Greensboro, NC 27402, USA.
| | - Lijun Zhao
- Key Laboratory of Ministry of Education for Medicinal Plant Resource and Natural Pharmaceutical Chemistry, College of Life Sciences, Shaanxi Normal University, Xi'an 710119, China.
| |
Collapse
|
12
|
Chen BY, Huang CC, Lv XF, Zheng HQ, Zhang YJ, Sun L, Wang GL, Ma MM, Guan YY. SGK1 mediates the hypotonic protective effect against H 2O 2-induced apoptosis of rat basilar artery smooth muscle cells by inhibiting the FOXO3a/Bim signaling pathway. Acta Pharmacol Sin 2020; 41:1073-1084. [PMID: 32139897 PMCID: PMC7470837 DOI: 10.1038/s41401-020-0357-y] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2019] [Accepted: 01/01/2020] [Indexed: 01/31/2023] Open
Abstract
Serum- and glucocorticoid-inducible kinease-1 (SGK1) is a serine/threonine kinase regulated by hypotonic stimuli, which is involved in regulation of cell cycle and apoptosis. Our previous study shows that activation of volume-regulated Cl- channels (VRCCs) protects rat basilar artery smooth muscle cells (BASMCs) against hydrogen peroxide (H2O2)-induced apoptosis. In the present study, we investigated whether SGK1 was involved in the protective effect of VRCCs in BASMCs. We showed that hypotonic challenge significantly reduced H2O2-induced apoptosis, and increased SGK1 phosphorylation, but did not affect SGK1 protein expression. The protective effect of hypotonic challenge against H2O2-induced apoptosis was mediated through inhibiting mitochondria-dependent apoptotic pathway, evidenced by increased Bcl-2/Bax ratio, stabilizing mitochondrial membrane potential (MMP), decreased cytochrome c release from the mitochondria to the cytoplasm, and inhibition of the activation of caspase-9 and caspase-3. These protective effects of hypotonic challenge against H2O2-induced apoptosis was diminished and enhanced, respectively, by SGK1 knockdown and overexpression. We further revealed that SGK1 activation significantly increased forkhead box O3a (FOXO3a) phosphorylation, and then inhibited the translocation of FOXO3a into nucleus and the subsequent expression of Bcl-2 interacting mediator of cell death (Bim). In conclusion, SGK1 mediates the protective effect of VRCCs against H2O2-induced apoptosis in BASMCs via inhibiting FOXO3a/Bim signaling pathway. Our results provide compelling evidences that SGK1 is a critical link between VRCCs and apoptosis, and shed a new light on the treatment of vascular apoptosis-associated diseases, such as vascular remodeling, angiogenesis, and atherosclerosis.
Collapse
|
13
|
Wu M, Liang C, Yu X, Song B, Yue Q, Zhai Y, Linck V, Cai Y, Niu N, Yang X, Zhang B, Wang Q, Zou L, Zhang S, Thai TL, Ma J, Sutliff RL, Zhang Z, Ma H. Lovastatin attenuates hypertension induced by renal tubule-specific knockout of ATP-binding cassette transporter A1, by inhibiting epithelial sodium channels. Br J Pharmacol 2019; 176:3695-3711. [PMID: 31222723 PMCID: PMC6715779 DOI: 10.1111/bph.14775] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2018] [Revised: 05/12/2019] [Accepted: 06/08/2019] [Indexed: 12/18/2022] Open
Abstract
BACKGROUND AND PURPOSE We have shown that cholesterol is synthesized in the principal cells of renal cortical collecting ducts (CCD) and stimulates the epithelial sodium channels (ENaC). Here we have determined whether lovastatin, a cholesterol synthesis inhibitor, can antagonize the hypertension induced by activated ENaC, following deletion of the cholesterol transporter (ATP-binding cassette transporter A1; ABCA1). EXPERIMENTAL APPROACH We selectively deleted ABCA1 in the principal cells of mouse CCD and used the cell-attached patch-clamp technique to record ENaC activity. Western blot and immunofluorescence staining were used to evaluate protein expression levels. Systolic BP was measured with the tail-cuff method. KEY RESULTS Specific deletion of ABCA1 elevated BP and ENaC single-channel activity in the principal cells of CCD in mice. These effects were antagonized by lovastatin. ABCA1 deletion elevated intracellular cholesterol levels, which was accompanied by elevated ROS, increased expression of serum/glucocorticoid regulated kinase 1 (Sgk1), phosphorylated neural precursor cell-expressed developmentally down-regulated protein 4-2 (Nedd4-2) and furin, along with shorten the primary cilium, and reduced ATP levels in urine. CONCLUSIONS AND IMPLICATIONS These data suggest that specific deletion of ABCA1 in principal cells increases BP by stimulating ENaC channels via a cholesterol-dependent pathway which induces several secondary responses associated with oxidative stress, activated Sgk1/Nedd4-2, increased furin expression, and reduced cilium-mediated release of ATP. As ABCA1 can be blocked by cyclosporine A, these results suggest further investigation of the possible use of statins to treat CsA-induced hypertension.
Collapse
Affiliation(s)
- Ming‐Ming Wu
- Departments of Cardiology and Clinic Pharmacy, Institute of Metabolic Disease, Heilongjiang Academy of Medical Science, Key Laboratories of Education Ministry for Myocardial Ischemia Mechanism and TreatmentHarbin Medical University Cancer HospitalHarbinChina
- Department of PhysiologyEmory University School of MedicineAtlantaGeorgia
| | - Chen Liang
- Departments of Cardiology and Clinic Pharmacy, Institute of Metabolic Disease, Heilongjiang Academy of Medical Science, Key Laboratories of Education Ministry for Myocardial Ischemia Mechanism and TreatmentHarbin Medical University Cancer HospitalHarbinChina
| | - Xiao‐Di Yu
- Departments of Cardiology and Clinic Pharmacy, Institute of Metabolic Disease, Heilongjiang Academy of Medical Science, Key Laboratories of Education Ministry for Myocardial Ischemia Mechanism and TreatmentHarbin Medical University Cancer HospitalHarbinChina
| | - Bin‐Lin Song
- Departments of Cardiology and Clinic Pharmacy, Institute of Metabolic Disease, Heilongjiang Academy of Medical Science, Key Laboratories of Education Ministry for Myocardial Ischemia Mechanism and TreatmentHarbin Medical University Cancer HospitalHarbinChina
- Department of PhysiologyEmory University School of MedicineAtlantaGeorgia
| | - Qiang Yue
- Department of PhysiologyEmory University School of MedicineAtlantaGeorgia
| | - Yu‐Jia Zhai
- Department of PhysiologyEmory University School of MedicineAtlantaGeorgia
| | - Valerie Linck
- Department of PhysiologyEmory University School of MedicineAtlantaGeorgia
| | - Yong‐Xu Cai
- Departments of Cardiology and Clinic Pharmacy, Institute of Metabolic Disease, Heilongjiang Academy of Medical Science, Key Laboratories of Education Ministry for Myocardial Ischemia Mechanism and TreatmentHarbin Medical University Cancer HospitalHarbinChina
| | - Na Niu
- Departments of Cardiology and Clinic Pharmacy, Institute of Metabolic Disease, Heilongjiang Academy of Medical Science, Key Laboratories of Education Ministry for Myocardial Ischemia Mechanism and TreatmentHarbin Medical University Cancer HospitalHarbinChina
| | - Xu Yang
- Departments of Cardiology and Clinic Pharmacy, Institute of Metabolic Disease, Heilongjiang Academy of Medical Science, Key Laboratories of Education Ministry for Myocardial Ischemia Mechanism and TreatmentHarbin Medical University Cancer HospitalHarbinChina
| | - Bao‐Long Zhang
- Departments of Cardiology and Clinic Pharmacy, Institute of Metabolic Disease, Heilongjiang Academy of Medical Science, Key Laboratories of Education Ministry for Myocardial Ischemia Mechanism and TreatmentHarbin Medical University Cancer HospitalHarbinChina
| | - Qiu‐Shi Wang
- Departments of Cardiology and Clinic Pharmacy, Institute of Metabolic Disease, Heilongjiang Academy of Medical Science, Key Laboratories of Education Ministry for Myocardial Ischemia Mechanism and TreatmentHarbin Medical University Cancer HospitalHarbinChina
| | - Li Zou
- Department of PhysiologyEmory University School of MedicineAtlantaGeorgia
| | - Shuai Zhang
- Department of PhysiologyEmory University School of MedicineAtlantaGeorgia
| | - Tiffany L. Thai
- Department of PhysiologyEmory University School of MedicineAtlantaGeorgia
| | - Jing Ma
- Division of Pulmonary, Allergy, Critical Care and Sleep Medicine, Department of MedicineAtlanta Veterans Affairs Medical CenterDecaturGeorgia
| | - Roy L. Sutliff
- Division of Pulmonary, Allergy, Critical Care and Sleep Medicine, Department of MedicineAtlanta Veterans Affairs Medical CenterDecaturGeorgia
| | - Zhi‐Ren Zhang
- Departments of Cardiology and Clinic Pharmacy, Institute of Metabolic Disease, Heilongjiang Academy of Medical Science, Key Laboratories of Education Ministry for Myocardial Ischemia Mechanism and TreatmentHarbin Medical University Cancer HospitalHarbinChina
| | - He‐Ping Ma
- Department of PhysiologyEmory University School of MedicineAtlantaGeorgia
| |
Collapse
|
14
|
Jeong JK, Horwath JA, Simonyan H, Blackmore KA, Butler SD, Young CN. Subfornical organ insulin receptors tonically modulate cardiovascular and metabolic function. Physiol Genomics 2019; 51:333-341. [PMID: 31172876 DOI: 10.1152/physiolgenomics.00021.2019] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023] Open
Abstract
Insulin acts within the central nervous system through the insulin receptor to influence both metabolic and cardiovascular physiology. While a major focus has been placed on hypothalamic regions, participation of extrahypothalamic insulin receptors in cardiometabolic regulation remains largely unknown. We hypothesized that insulin receptors in the subfornical organ (SFO), a forebrain circumventricular region devoid of a blood-brain barrier, are involved in metabolic and cardiovascular regulation. Immunohistochemistry in mice revealed widespread insulin receptor-positive cells throughout the rostral to caudal extent of the SFO. SFO-targeted adenoviral delivery of Cre-recombinase in insulin receptorlox/lox mice resulted in sufficient ablation of insulin receptors in the SFO. Interestingly, when mice were maintained on a normal chow diet, deletion of SFO insulin receptors resulted in greater weight gain and adiposity, relative to controls, independently of changes in food intake. In line with this, ablation of insulin receptors in the SFO was associated with marked hepatic steatosis and hypertriglyceridemia. Selective removal of SFO insulin receptors also resulted in a lower mean arterial blood pressure, which was primarily due to a reduction in diastolic blood pressure, whereas systolic blood pressure remained unchanged. Cre-mediated targeting of SFO insulin receptors did not influence heart rate. These data demonstrate multidirectional roles for insulin receptor signaling in the SFO, with ablation of SFO insulin receptors resulting in an overall deleterious metabolic state while at the same time maintaining blood pressure at low levels. These novel findings further suggest that alterations in insulin receptor signaling in the SFO could contribute to metabolic syndrome phenotypes.
Collapse
Affiliation(s)
- Jin Kwon Jeong
- Department of Pharmacology and Physiology, School of Medicine and Health Sciences, The George Washington University, Washington, District of Columbia
| | - Julie A Horwath
- Biomedical Sciences, College of Veterinary Medicine, Cornell University, Ithaca, New York
| | - Hayk Simonyan
- Department of Pharmacology and Physiology, School of Medicine and Health Sciences, The George Washington University, Washington, District of Columbia
| | - Katherine A Blackmore
- Department of Pharmacology and Physiology, School of Medicine and Health Sciences, The George Washington University, Washington, District of Columbia
| | - Scott D Butler
- Biomedical Sciences, College of Veterinary Medicine, Cornell University, Ithaca, New York
| | - Colin N Young
- Department of Pharmacology and Physiology, School of Medicine and Health Sciences, The George Washington University, Washington, District of Columbia
| |
Collapse
|
15
|
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
|
16
|
Peng LQ, Zhao H, Liu S, Yuan YP, Yuan CY, Mwamunyi MJ, Pearce D, Yao LJ. Lack of serum- and glucocorticoid-inducible kinase 3 leads to podocyte dysfunction. FASEB J 2018; 32:576-587. [PMID: 28935820 DOI: 10.1096/fj.201700393rr] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Serum- and glucocorticoid-inducible kinase 3 (SGK3) is a downstream mediator of PI3K, which is essential for maintaining the functional integrity of podocytes. However, little is known about the role of SGK3 in podocyte function. Herein, we demonstrated that SGK3 contributes to the maintenance of podocyte integrity. Conditionally immortalized mouse podocyte cells (MPCs) were treated with puromycin aminonucleoside (PAN). PAN treatment inhibited the activity of SGK3 and the expression of podocin. Short hairpin RNA (shRNA)-mediated knockdown of SGK3 also reduced podocin expression in the absence of PAN. Adriamycin (ADR)-treated mice developed proteinuria and had decreased renal glomerular SGK3 expression in comparison to control mice. Consistent with a role for SGK3 in the ADR effect, SGK3 knockout (KO) mice had markedly reduced kidney podocin expression and significantly elevated proteinuria compared with wild-type mice. Electron microscopy revealed that SGK3 KO mice displayed partial effacement of podocyte foot processes. Further, a SGK3 target protein, glycogen synthase kinase-3 (GSK3), was discovered to be dramatically activated in PAN and SGK3 shRNA-treated MPCs and in SGK3 KO mice. Taken together, these data strongly suggest that SGK3 plays a significant role in regulating podocyte function, likely by controlling the expression and activity of GSK3.-Peng, L.-Q., Zhao, H., Liu, S., Yuan, Y.-P., Yuan, C.-Y., Mwamunyi, M.-J., Pearce, D., Yao, L.-J. Lack of serum- and glucocorticoid-inducible kinase 3 leads to podocyte dysfunction.
Collapse
Affiliation(s)
- Li-Qin Peng
- Department of Nephrology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Hong Zhao
- Department of Trauma Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Song Liu
- Department of Nephrology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Ya-Pei Yuan
- Department of Nephrology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Cheng-Yan Yuan
- Department of Nephrology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Mercy-Julian Mwamunyi
- Department of Nephrology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - David Pearce
- Department of Medicine, University of California, San Francisco, San Francisco, California, USA.,Department of Molecular and Cellular Pharmacology, University of California, San Francisco, San Francisco, California, USA
| | - Li-Jun Yao
- Department of Nephrology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|
17
|
Abstract
PURPOSE High levels of NaCl in the diet are associated with both cardiac and renal fibrosis, but whether salt intake affects pulmonary fibrosis has not been examined. AIM OF THE STUDY To test the hypothesis that salt intake might affect pulmonary fibrosis. MATERIALS AND METHODS Mice were fed low, normal, or high salt diets for 2 weeks, and then treated with oropharyngeal bleomycin to induce pulmonary fibrosis, or oropharyngeal saline as a control. RESULTS As determined by collagen staining of lung sections, and protein levels and cell numbers in the bronchoalveolar lavage (BAL) fluid at 21 days after bleomycin, the high salt diet did not exacerbate bleomycin-induced fibrosis, while the low salt diet attenuated fibrosis. For the bleomycin-treated mice, staining of the post-BAL lung sections indicated that compared to the regular salt diet, high salt increased the number of Ly6c-positive macrophages and decreased the number of CD11c and CD206-positive macrophages and dendritic cells. The low salt diet caused bleomycin-induced leukocyte numbers to be similar to control saline-treated mice, but reduced numbers of CD45/collagen-VI positive fibrocytes. In the saline controls, low dietary salt decreased CD11b and CD11c positive cells in lung sections, and high dietary salt increased fibrocytes. CONCLUSIONS Together, these data suggest the possibility that a low salt diet might attenuate pulmonary fibrosis.
Collapse
Affiliation(s)
- Wensheng Chen
- a Department of Biology , Texas A&M University , College Station , TX , USA.,b Institute of Clinical Pharmacology, Anhui Medical University, Key Laboratory of Anti-inflammatory and Immune Medicine, Ministry of Education, Anhui Collaborative Innovation Center of Anti-inflammatory and Immune Medicine , Hefei , China
| | - Darrell Pilling
- a Department of Biology , Texas A&M University , College Station , TX , USA
| | - Richard H Gomer
- a Department of Biology , Texas A&M University , College Station , TX , USA
| |
Collapse
|
18
|
Sibiya N, Ngubane P, Mabandla M. Cardioprotective effects of pectin-insulin patch in streptozotocin-induced diabetic rats. J Diabetes 2017; 9:1073-1081. [PMID: 28220624 DOI: 10.1111/1753-0407.12538] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/03/2016] [Revised: 01/16/2017] [Accepted: 02/14/2017] [Indexed: 02/04/2023] Open
Abstract
BACKGROUND Cardiovascular complications are among the leading causes of morbidity and mortality in diabetes mellitus. Despite the beneficial effects of subcutaneous insulin, reports suggest that the therapy itself precipitates cardiovascular risks due to the high insulin concentration administered. It is therefore necessary to seek alternative routes of insulin administration that may bypass the undesirable effects associated with high plasma insulin concentrations. Accordingly, the present study investigated the effects of a novel transdermal pectin-insulin patch on selected markers of cardiovascular function in diabetes. METHODS Pectin-insulin matrix patches (20.0, 40.8, and 82.9 μg/kg) were prepared as described previously. The three formulations were applied to streptozotocin-induced diabetic rats thrice daily. Blood glucose concentrations and mean arterial pressure (MAP) were monitored weekly for 5 weeks. Rats were then killed and blood collected for analysis of the lipid profile, cardiotropin-1, tumor necrosis factor (TNF)-α, and high-sensitivity C-reactive protein (hsCRP). RESULTS The patches decreased blood glucose concentrations and diabetes-induced disturbances in lipid profile were attenuated by patch application (82.9 μg/kg). The diabetes-induced increase in MAP was also attenuated in patch (82.9 μg/kg)-treated rats. Patch treatment resulted in a decreased heart weight: body weight ratio, as well as reductions in cardiotropin-1, TNF-α, and hsCRP concentrations. CONCLUSIONS Application of the pectin-insulin patch protects against the debilitating cardiovascular effects associated with conventional diabetes treatment. This suggests that the pectin-insulin patch may provide an effective alternative therapeutic approach to the commonly used subcutaneous insulin injections in the management of diabetes.
Collapse
Affiliation(s)
- Ntethelelo Sibiya
- Department of Human Physiology, School of Laboratory Medicine and Medical Sciences, College of Health Sciences, University of KwaZulu-Natal, Durban, South Africa
| | - Phikelelani Ngubane
- Department of Human Physiology, School of Laboratory Medicine and Medical Sciences, College of Health Sciences, University of KwaZulu-Natal, Durban, South Africa
| | - Musa Mabandla
- Department of Human Physiology, School of Laboratory Medicine and Medical Sciences, College of Health Sciences, University of KwaZulu-Natal, Durban, South Africa
| |
Collapse
|
19
|
Molecular Mechanisms of Sodium-Sensitive Hypertension in the Metabolic Syndrome. Curr Hypertens Rep 2017; 19:60. [DOI: 10.1007/s11906-017-0759-5] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023]
|
20
|
Zhang D, Gu D, He J, Hixson JE, Rao DC, Li C, He H, Chen J, Huang J, Chen J, Rice TK, Chen S, Kelly TN. Associations of the Serum/Glucocorticoid Regulated Kinase Genes With BP Changes and Hypertension Incidence: The Gensalt Study. Am J Hypertens 2017; 30:95-101. [PMID: 27664953 DOI: 10.1093/ajh/hpw122] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2016] [Revised: 05/19/2016] [Accepted: 09/08/2016] [Indexed: 01/19/2023] Open
Abstract
BACKGROUND Single-marker and novel gene-based methods were employed to examine the associations of the serum/glucocorticoid regulated kinases (SGK) gene family with longitudinal blood pressure (BP) changes and hypertension incidence in a family-based cohort study. METHODS Totally, 1,768 Chinese participants from the Genetic Epidemiology Network of Salt Sensitivity (GenSalt) follow-up study were included in the current analyses. Nine BP measures were obtained at each of 3 visits during the GenSalt follow-up study. Mixed-model and Gene-based analyses were used to examine the associations of the SGK gene family with longitudinal BP phenotypes. Bonferroni correction was applied to account for multiple testing. RESULTS After an average 7.2-year follow-up, 32.2% (513) of participants free of hypertension at baseline developed hypertension. Four novel SNPs in the SGK1 gene were predictive of the longitudinal BP phenotypes. The major alleles of SGK1 rs1763498 and rs114414980 conferred 2.9- and 2.5-fold increased risks of hypertension development, respectively (P = 1.0×10-4 and 6.0×10-4, respectively). In addition, the major allele of SGK1 rs229133 was significantly associated with 0.4mm Hg larger annual increases in systolic BP (P = 4.2×10-4), while the major allele of rs6924468 was significantly associated with 0.2mm Hg smaller annual increases in diastolic BP (P = 4.2×10-4). Gene-based analyses revealed an association of the SGK1 gene with risk of hypertension development (P = 7.4×10-3). No evidence for the SGK2 and SGK3 genes was found. CONCLUSIONS The findings of the current study suggest that the SGK1 gene may play a role in long-term BP regulation and hypertension incidence.
Collapse
Affiliation(s)
- Dingding Zhang
- Department of Evidence Based Medicine, 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
- Department of Epidemiology, Tulane University School of Public Health and Tropical Medicine, New Orleans, Louisiana, USA
| | - Dongfeng Gu
- Department of Evidence Based Medicine, 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, USA
| | - James E Hixson
- Department of Epidemiology, Human Genetics and Environmental Sciences, University of Texas School of Public Health, Houston, Texas, USA
| | - Dabeeru C Rao
- Division of Biostatistics, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Changwei Li
- Department of Epidemiology, Tulane University School of Public Health and Tropical Medicine, New Orleans, Louisiana, USA
| | - Hua He
- Department of Epidemiology, Tulane University School of Public Health and Tropical Medicine, New Orleans, Louisiana, USA
| | - Jichun Chen
- Department of Evidence Based Medicine, 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
| | - Jianfeng Huang
- Department of Evidence Based Medicine, 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
| | - Jing Chen
- Department of Epidemiology, Tulane University School of Public Health and Tropical Medicine, New Orleans, Louisiana, USA
| | - Treva K Rice
- Division of Biostatistics, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Shufeng Chen
- Department of Evidence Based Medicine, 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, USA;
| |
Collapse
|
21
|
Pao AC. There and back again: insulin, ENaC, and the cortical collecting duct. Physiol Rep 2016; 4:4/10/e12809. [PMID: 27233302 PMCID: PMC4886174 DOI: 10.14814/phy2.12809] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2016] [Accepted: 05/03/2016] [Indexed: 01/02/2023] Open
Abstract
Cell culture models suggest mechanisms by which insulin stimulates ENaC in the cortical collecting duct. These mechanisms still need to be tested for physiological significance in animal models of insulin resistance.![]()
Collapse
Affiliation(s)
- Alan C Pao
- Department of Medicine Stanford University, Stanford, California
| |
Collapse
|
22
|
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
|
23
|
Elijovich F, Weinberger MH, Anderson CAM, Appel LJ, Bursztyn M, Cook NR, Dart RA, Newton-Cheh CH, Sacks FM, Laffer CL. Salt Sensitivity of Blood Pressure: A Scientific Statement From the American Heart Association. Hypertension 2016; 68:e7-e46. [PMID: 27443572 DOI: 10.1161/hyp.0000000000000047] [Citation(s) in RCA: 339] [Impact Index Per Article: 42.4] [Reference Citation Analysis] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
|
24
|
Yang L, Frindt G, Lang F, Kuhl D, Vallon V, Palmer LG. SGK1-dependent ENaC processing and trafficking in mice with high dietary K intake and elevated aldosterone. Am J Physiol Renal Physiol 2016; 312:F65-F76. [PMID: 27413200 DOI: 10.1152/ajprenal.00257.2016] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2016] [Accepted: 07/06/2016] [Indexed: 02/04/2023] Open
Abstract
We examined renal Na and K transporters in mice with deletions in the gene encoding the aldosterone-induced protein SGK1. The knockout mice were hyperkalemic, and had altered expression of the subunits of the epithelial Na channel (ENaC). The kidneys showed decreased expression of the cleaved forms of the γENaC subunit, and the fully glycosylated form of the βENaC subunits when animals were fed a high-K diet. Knockout animals treated with exogenous aldosterone also had reduced subunit processing and diminished surface expression of βENaC and γENaC. Expression of the three upstream Na transporters NHE3, NKCC2, and NCC was reduced in both wild-type and knockout mice in response to K loading. The activity of ENaC measured as whole cell amiloride-sensitive current (INa) in principal cells of the cortical collecting duct (CCD) was minimal under control conditions but was increased by a high-K diet to a similar extent in knockout and wild-type animals. INa in the connecting tubule also increased similarly in the two genotypes in response to exogenous aldosterone administration. The activities of both ROMK channels in principal cells and BK channels in intercalated cells of the CCD were unaffected by the deletion of SGK1. Acute treatment of animals with amiloride produced similar increases in Na excretion and decreases in K excretion in the two genotypes. The absence of changes in ENaC activity suggests compensation for decreased surface expression. Altered K balance in animals lacking SGK1 may reflect defects in ENaC-independent K excretion.
Collapse
Affiliation(s)
- Lei Yang
- Department of Physiology and Biophysics, Weill-Cornell Medical College, New York, New York.,Department of Physiology, Harbin Medical University, Harbin, China
| | - Gustavo Frindt
- Department of Physiology and Biophysics, Weill-Cornell Medical College, New York, New York
| | - Florian Lang
- Department of Cardiology, Vascular Medicine and Physiology, University of Tübingen, Tübingen, Germany
| | - Dietmar Kuhl
- Institute for Molecular and Cellular Cognition, Center for Molecular Neurobiology Hamburg, University Medical Center Hamburg-Eppendorf, Hamburg, Germany; and
| | - Volker Vallon
- Departments of Medicine and Pharmacology, University of California San Diego and Veterans Affairs San Diego Healthcare System, San Diego, California
| | - Lawrence G Palmer
- Department of Physiology and Biophysics, Weill-Cornell Medical College, New York, New York;
| |
Collapse
|
25
|
Nizar JM, Dong W, McClellan RB, Labarca M, Zhou Y, Wong J, Goens DG, Zhao M, Velarde N, Bernstein D, Pellizzon M, Satlin LM, Bhalla V. Na+-sensitive elevation in blood pressure is ENaC independent in diet-induced obesity and insulin resistance. Am J Physiol Renal Physiol 2016; 310:F812-20. [PMID: 26841823 PMCID: PMC4867314 DOI: 10.1152/ajprenal.00265.2015] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2015] [Accepted: 01/28/2016] [Indexed: 02/08/2023] Open
Abstract
The majority of patients with obesity, insulin resistance, and metabolic syndrome have hypertension, but the mechanisms of hypertension are poorly understood. In these patients, impaired sodium excretion is critical for the genesis of Na(+)-sensitive hypertension, and prior studies have proposed a role for the epithelial Na(+) channel (ENaC) in this syndrome. We characterized high fat-fed mice as a model in which to study the contribution of ENaC-mediated Na(+) reabsorption in obesity and insulin resistance. High fat-fed mice demonstrated impaired Na(+) excretion and elevated blood pressure, which was significantly higher on a high-Na(+) diet compared with low fat-fed control mice. However, high fat-fed mice had no increase in ENaC activity as measured by Na(+) transport across microperfused cortical collecting ducts, electrolyte excretion, or blood pressure. In addition, we found no difference in endogenous urinary aldosterone excretion between groups on a normal or high-Na(+) diet. High fat-fed mice provide a model of metabolic syndrome, recapitulating obesity, insulin resistance, impaired natriuresis, and a Na(+)-sensitive elevation in blood pressure. Surprisingly, in contrast to previous studies, our data demonstrate that high fat feeding of mice impairs natriuresis and produces elevated blood pressure that is independent of ENaC activity and likely caused by increased Na(+) reabsorption upstream of the aldosterone-sensitive distal nephron.
Collapse
Affiliation(s)
- Jonathan M Nizar
- Division of Nephrology, Department of Medicine, Stanford University, Palo Alto, California
| | - Wuxing Dong
- Division of Nephrology, Department of Medicine, Stanford University, Palo Alto, California
| | - Robert B McClellan
- Division of Nephrology, Department of Medicine, Stanford University, Palo Alto, California
| | - Mariana Labarca
- Division of Nephrology, Department of Medicine, Stanford University, Palo Alto, California
| | - Yuehan Zhou
- Division of Pediatric Nephrology, Department of Pediatrics, Icahn School of Medicine at Mount Sinai, New York, New York
| | - Jared Wong
- Division of Nephrology, Department of Medicine, Stanford University, Palo Alto, California
| | - Donald G Goens
- Division of Nephrology, Department of Medicine, Stanford University, Palo Alto, California
| | - Mingming Zhao
- Division of Pediatric Cardiology, Department of Pediatrics, Stanford University, Stanford, California; and
| | - Nona Velarde
- Division of Nephrology, Department of Medicine, Stanford University, Palo Alto, California
| | - Daniel Bernstein
- Division of Pediatric Cardiology, Department of Pediatrics, Stanford University, Stanford, California; and
| | | | - Lisa M Satlin
- Division of Pediatric Nephrology, Department of Pediatrics, Icahn School of Medicine at Mount Sinai, New York, New York
| | - Vivek Bhalla
- Division of Nephrology, Department of Medicine, Stanford University, Palo Alto, California;
| |
Collapse
|
26
|
Scott TA, Babayeva O, Banerjee S, Zhong W, Francis SC. SGK1 is modulated by resistin in vascular smooth muscle cells and in the aorta following diet-induced obesity. Obesity (Silver Spring) 2016; 24:678-86. [PMID: 26833885 PMCID: PMC4987962 DOI: 10.1002/oby.21425] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/25/2015] [Revised: 10/13/2015] [Accepted: 10/15/2015] [Indexed: 12/16/2022]
Abstract
OBJECTIVE Enhanced serum and glucocorticoid-inducible kinase 1 (SGK1) activity contributes to the pathogenesis of vascular disease. This study evaluated SGK1 modulation in vascular smooth muscle cells by the adipokine resistin and in aortic tissue in a murine model of diet-induced obesity (DIO). METHODS Modulation of SGK1 by resistin was assessed in human aortic smooth muscle cells (HAoSMC) in vitro by quantitative RT-PCR and Western blot analyses. To induce the lean or obese phenotype, mice were fed a 10 kcal% low-fat or 60 kcal% high-fat diet, respectively, for 8 weeks. Upon study completion, plasma resistin was assessed and aortic tissue was harvested to examine the effect of DIO on regulation of SGK1 in vivo. RESULTS Resistin increased SGK1 mRNA, total protein abundance, and its activation as determined by phosphorylation of its serine 422 residue (pSGK1) in HAoSMC. Resistin-mediated SGK1 phosphorylation was dependent upon phosphatidylinositol-3-kinase and Toll-like receptor 4. Furthermore, inhibition of SGK1 attenuated resistin-induced proliferation in HAoSMC. DIO led to up-regulation of total SGK1 protein levels and pSGK1 in association with increased plasma resistin. CONCLUSIONS These data suggest that high levels of resistin observed during obesity may activate SGK1 in the vasculature and contribute to the development of obesity-related vascular disease.
Collapse
Affiliation(s)
- Takara A. Scott
- Cardiovascular Research Institute, Morehouse School of Medicine
| | | | | | - Wei Zhong
- Cardiovascular Research Institute, Morehouse School of Medicine
| | - Sharon C. Francis
- Department of Physiology, Morehouse School of Medicine
- Cardiovascular Research Institute, Morehouse School of Medicine
| |
Collapse
|
27
|
Rossier BC, Baker ME, Studer RA. Epithelial sodium transport and its control by aldosterone: the story of our internal environment revisited. Physiol Rev 2015; 95:297-340. [PMID: 25540145 DOI: 10.1152/physrev.00011.2014] [Citation(s) in RCA: 155] [Impact Index Per Article: 17.2] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
Transcription and translation require a high concentration of potassium across the entire tree of life. The conservation of a high intracellular potassium was an absolute requirement for the evolution of life on Earth. This was achieved by the interplay of P- and V-ATPases that can set up electrochemical gradients across the cell membrane, an energetically costly process requiring the synthesis of ATP by F-ATPases. In animals, the control of an extracellular compartment was achieved by the emergence of multicellular organisms able to produce tight epithelial barriers creating a stable extracellular milieu. Finally, the adaptation to a terrestrian environment was achieved by the evolution of distinct regulatory pathways allowing salt and water conservation. In this review we emphasize the critical and dual role of Na(+)-K(+)-ATPase in the control of the ionic composition of the extracellular fluid and the renin-angiotensin-aldosterone system (RAAS) in salt and water conservation in vertebrates. The action of aldosterone on transepithelial sodium transport by activation of the epithelial sodium channel (ENaC) at the apical membrane and that of Na(+)-K(+)-ATPase at the basolateral membrane may have evolved in lungfish before the emergence of tetrapods. Finally, we discuss the implication of RAAS in the origin of the present pandemia of hypertension and its associated cardiovascular diseases.
Collapse
Affiliation(s)
- Bernard C Rossier
- Department of Pharmacology and Toxicology, University of Lausanne, Lausanne, Switzerland; Division of Nephrology-Hypertension, University of California San Diego, La Jolla, California; and Institute of Structural and Molecular Biology, Division of Biosciences, University College London, London, United Kingdom
| | - Michael E Baker
- Department of Pharmacology and Toxicology, University of Lausanne, Lausanne, Switzerland; Division of Nephrology-Hypertension, University of California San Diego, La Jolla, California; and Institute of Structural and Molecular Biology, Division of Biosciences, University College London, London, United Kingdom
| | - Romain A Studer
- Department of Pharmacology and Toxicology, University of Lausanne, Lausanne, Switzerland; Division of Nephrology-Hypertension, University of California San Diego, La Jolla, California; and Institute of Structural and Molecular Biology, Division of Biosciences, University College London, London, United Kingdom
| |
Collapse
|
28
|
Schreier B, Rabe S, Winter S, Ruhs S, Mildenberger S, Schneider B, Sibilia M, Gotthardt M, Kempe S, Mäder K, Grossmann C, Gekle M. Moderate inappropriately high aldosterone/NaCl constellation in mice: cardiovascular effects and the role of cardiovascular epidermal growth factor receptor. Sci Rep 2014; 4:7430. [PMID: 25503263 PMCID: PMC4262830 DOI: 10.1038/srep07430] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2014] [Accepted: 11/20/2014] [Indexed: 12/16/2022] Open
Abstract
Non-physiological activation of the mineralocorticoid receptor (MR), e.g. by aldosterone under conditions of high salt intake, contributes to the pathogenesis of cardiovascular diseases, although beneficial effects of aldosterone also have been described. The epidermal growth factor receptor (EGFR) contributes to cardiovascular alterations and mediates part of the MR effects. Recently, we showed that EGFR is required for physiological homeostasis and function of heart and arteries in adult animals. We hypothesize that moderate high aldosterone/NaCl, at normal blood pressure, affects the cardiovascular system depending on cardiovascular EGFR. Therefore we performed an experimental series in male and female animals each, using a recently established mouse model with EGFR knockout in vascular smooth muscle cells and cardiomyocytes and determined the effects of a mild-high aldosterone-to-NaCl constellation on a.o. marker gene expression, heart size, systolic blood pressure, impulse conduction and heart rate. Our data show that (i) cardiac tissue of male but not of female mice is sensitive to mild aldosterone/NaCl treatment, (ii) EGFR knockout induces stronger cardiac disturbances in male as compared to female animals and (iii) mild aldosterone/NaCl treatment requires the EGFR in order to disturb cardiac tissue homeostasis whereas beneficial effects of aldosterone seem to be independent of EGFR.
Collapse
Affiliation(s)
- Barbara Schreier
- Julius-Bernstein-Institute of Physiology, Medical Faculty, University of Halle-Wittenberg, Halle, Germany
| | - Sindy Rabe
- Julius-Bernstein-Institute of Physiology, Medical Faculty, University of Halle-Wittenberg, Halle, Germany
| | - Sabrina Winter
- Julius-Bernstein-Institute of Physiology, Medical Faculty, University of Halle-Wittenberg, Halle, Germany
| | - Stefanie Ruhs
- Julius-Bernstein-Institute of Physiology, Medical Faculty, University of Halle-Wittenberg, Halle, Germany
| | - Sigrid Mildenberger
- Julius-Bernstein-Institute of Physiology, Medical Faculty, University of Halle-Wittenberg, Halle, Germany
| | - Bettina Schneider
- Julius-Bernstein-Institute of Physiology, Medical Faculty, University of Halle-Wittenberg, Halle, Germany
| | - Maria Sibilia
- Institute of Cancer Research, Medical University of Vienna, Vienna, Austria
| | - Michael Gotthardt
- Max-Delbrück-Center for Molecular Medicine (MDC), Berlin-Buch, Germany
| | - Sabine Kempe
- Institute of Pharmacy, Faculty of Natural Sciences 1, University of Halle-Wittenberg, Halle, Germany
| | - Karsten Mäder
- Institute of Pharmacy, Faculty of Natural Sciences 1, University of Halle-Wittenberg, Halle, Germany
| | - Claudia Grossmann
- Julius-Bernstein-Institute of Physiology, Medical Faculty, University of Halle-Wittenberg, Halle, Germany
| | - Michael Gekle
- Julius-Bernstein-Institute of Physiology, Medical Faculty, University of Halle-Wittenberg, Halle, Germany
| |
Collapse
|
29
|
Kemp BA, Howell NL, Gildea JJ, Padia SH. Intrarenal ghrelin receptor antagonism prevents high-fat diet-induced hypertension in male rats. Endocrinology 2014; 155:2658-66. [PMID: 24797629 DOI: 10.1210/en.2013-2177] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
Abstract
Excess weight gain contributes up to 65% of the risk of primary hypertension, and the increase in blood pressure in response to high-fat diet (HFD) is preceded by significant increases in renal tubular sodium (Na(+)) reabsorption. In normal rats, intrarenal ghrelin infusion increases distal nephron-dependent Na(+) reabsorption via activation of the intrarenal ghrelin receptor (GHSR). This study focusses on the role of intrarenal GHSR-mediated Na(+) reabsorption in HFD-induced hypertension. Dahl salt-sensitive rats received standard diet or HFD for 6 weeks. Rats underwent uninephrectomy and osmotic minipump implantation for chronic intrarenal delivery of vehicle (0.25 μL/h × 28 d), selective GHSR antagonist [D-Lys-3]-growth hormone releasing peptide-6 (0.2μM/d), or GHSR inverse agonist [D-Arg(1), D-Phe(5), D-Trp(7,9), Leu(11)]-substance P (SUB-P) (3.6μM/d). HFD rats with vehicle pumps had significantly increased renal GHSR expression compared with standard diet (0.092 ± 0.005 vs 0.065 ± 0.004 arbitrary units; P < .05), whereas acyl ghrelin levels were similar (16.3±6.2 vs 15.7±8.7 pg/g tissue). HFD rats with vehicle pumps became hypertensive after 2 weeks (P < .05) and showed a significant reduction in 24-hour urine Na(+) before hypertension. At this time, these rats showed an increase in collecting duct α-epithelial Na(+) channel, thereby providing a potential mechanism for the excess Na(+) reabsorption. In contrast, HFD rats with [D-Lys-3]-growth hormone releasing peptide-6 or SUB-P pumps never became hypertensive and did not show the reduction in urine Na(+). Because SUB-P blocks the constitutive, but not ghrelin-dependent, activity of the GHSR, and HFD-induced α-epithelial Na(+) channel up-regulation was abolished during GHSR antagonism, these data suggest that HFD increases the constitutive activity of renal GHSR to increase Na(+) reabsorption and induce hypertension in rats.
Collapse
Affiliation(s)
- Brandon A Kemp
- Division of Endocrinology and Metabolism (B.A.K., N.L.H., S.H.P.), Department of Medicine, and Department of Pathology (J.J.G.), University of Virginia School of Medicine, Charlottesville, Virginia 22908-1414
| | | | | | | |
Collapse
|
30
|
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
|
31
|
Gopinath B, Harris DC, Flood VM, Burlutsky G, Mitchell P. A better diet quality is associated with a reduced likelihood of CKD in older adults. Nutr Metab Cardiovasc Dis 2013; 23:937-943. [PMID: 22902186 DOI: 10.1016/j.numecd.2012.07.003] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/28/2012] [Revised: 06/25/2012] [Accepted: 07/16/2012] [Indexed: 11/28/2022]
Abstract
BACKGROUND AND AIMS Studies of diet in relation to chronic kidney disease (CKD) have focused on individual nutrients. The relationship between overall patterns of food intake and renal function has not been well explored. We aimed to investigate the associations between diet quality with the prevalence, incidence and progression of CKD in a cohort of older adults. METHODS AND RESULTS 1952 participants aged ≥50 years at baseline were examined between 1992-1994 and 2002-2004. Dietary data were collected using a semi-quantitative food frequency questionnaire. A modified version of the Healthy Eating Index for Australians was developed to determine total diet scores (TDS). Baseline biochemistry including serum creatinine was measured. CKD was defined as MDRD estimated glomerular filtration rate (eGFR) <60 mL min⁻¹·1.73 m⁻². Participants in the highest quartile of mean TDS compared to those in the first quartile (reference), had a 41% reduced likelihood of having eGFR <60 mL min⁻¹·1.73 m⁻², [multivariable-adjusted odds ratio, OR, 0.59 (95% confidence intervals, CI, 0.41-0.85), P-trend = 0.005]. Each unit increase in TDS was associated with a 15% decrease in the odds of having prevalent CKD, multivariable-adjusted OR 0.85 (95% CI 0.74-0.97). A non-significant, inverse association between TDS and CKD incidence was observed (P-trend = 0.10). CONCLUSION Older adults with better diet quality had a reduced likelihood of having eGFR <60 mL min⁻¹·1.73 m⁻². Adherence to dietary guidelines were not prospectively associated with CKD incidence or progression. Further studies with adequate power are warranted to assess the longitudinal association between diet quality and CKD.
Collapse
Affiliation(s)
- B Gopinath
- Centre for Vision Research, Department of Ophthalmology and Westmead Millennium Institute, University of Sydney, Westmead Hospital, Hawkesbury Rd, Westmead, NSW 2145, Australia
| | | | | | | | | |
Collapse
|
32
|
Abstract
Hydroxy methyl glutaryl CoA inhibitors (statins) are the agents most frequently used to reduce elevated serum cholesterol. In addition to their cholesterol lowering effects, statins also have nonlipid lowering pleiotropic properties. These include reducing oxidative stress, renin-angiotensin and endothelin synthesis and activity, and improving nitric oxide (NO) synthesis and availability. Thus, one would predict that statins might be able to exert an antihypertensive effect. Experimental models bear out the blood pressure lowering effects but the data from clinical trials have been inconsistent perhaps due to inappropriate experimental designs, sample size, blood pressure measurement techniques etc. Moreover, although experimental models strongly suggest a role for salt intake in the potential antihypertensive responses to statins, available clinical trials fail to report salt intake in the studied populations. The statins' antihypertensive effects remain an unsettled hypothesis and calls for a large clinical trial at a wide range of doses and a controlled salt intake. Statins meanwhile remain as a excellent option to control high cholesterol and in tissue injury prevention.
Collapse
|
33
|
Serum- and glucocorticoid-inducible kinase 1 in the regulation of renal and extrarenal potassium transport. Clin Exp Nephrol 2011; 16:73-80. [DOI: 10.1007/s10157-011-0488-z] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2009] [Accepted: 04/08/2010] [Indexed: 01/24/2023]
|
34
|
Odermatt A. The Western-style diet: a major risk factor for impaired kidney function and chronic kidney disease. Am J Physiol Renal Physiol 2011; 301:F919-31. [PMID: 21880837 DOI: 10.1152/ajprenal.00068.2011] [Citation(s) in RCA: 164] [Impact Index Per Article: 12.6] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
The Western-style diet is characterized by its highly processed and refined foods and high contents of sugars, salt, and fat and protein from red meat. It has been recognized as the major contributor to metabolic disturbances and the development of obesity-related diseases including type 2 diabetes, hypertension, and cardiovascular disease. Also, the Western-style diet has been associated with an increased incidence of chronic kidney disease (CKD). A combination of dietary factors contributes to the impairment of renal vascularization, steatosis and inflammation, hypertension, and impaired renal hormonal regulation. This review addresses recent progress in the understanding of the association of the Western-style diet with the induction of dyslipidemia, oxidative stress, inflammation, and disturbances of corticosteroid regulation in the development of CKD. Future research needs to distinguish between acute and chronic effects of diets with high contents of sugars, salt, and fat and protein from red meat, and to uncover the contribution of each component. Improved therapeutic interventions should consider potentially altered drug metabolism and pharmacokinetics and be combined with lifestyle changes. A clinical assessment of the long-term risks of whole-body disturbances is strongly recommended to reduce metabolic complications and cardiovascular risk in kidney donors and patients with CKD.
Collapse
Affiliation(s)
- Alex Odermatt
- Div. of Molecular and Systems Toxicology, Dept. of Pharmaceutical Sciences, Univ. of Basel, Klingelbergstrasse 50, CH-4056 Basel, Switzerland.
| |
Collapse
|
35
|
Ackermann TF, Boini KM, Beier N, Scholz W, Fuchss T, Lang F. EMD638683, a novel SGK inhibitor with antihypertensive potency. Cell Physiol Biochem 2011; 28:137-46. [PMID: 21865856 DOI: 10.1159/000331722] [Citation(s) in RCA: 93] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/13/2011] [Indexed: 12/11/2022] Open
Abstract
UNLABELLED The serum- and glucocorticoid-inducible kinase 1 (SGK1) is transcriptionally upregulated by mineralocorticoids and activated by insulin. The kinase enhances renal tubular Na(+)-reabsorption and accounts for blood pressure increase following high salt diet in mice made hyperinsulinemic by dietary fructose or fat. The present study describes the in vitro and in vivo efficacy of a novel SGK1 inhibitor (EMD638683). EMD638683 was tested in vitro by determination of SGK1-dependent phosphorylation of NDRG1 (N-Myc downstream-regulated gene 1) in human cervical carcinoma HeLa-cells. In vivo EMD638683 (4460 ppm in chow, i.e. approx. 600 mg/kg/day) was administered to mice drinking tap water or isotonic saline containing 10% fructose. Blood pressure was determined by the tail cuff method, and urinary electrolyte (flame photometry) concentrations determined in metabolic cages. In vitro testing disclosed EMD638683 as a SGK1 inhibitor with an IC50 of 3 μM. Within 24 hours in vivo EMD638683 treatment significantly decreased blood pressure in fructose/saline-treated mice but not in control animals or in SGK1 knockout mice. EMD638683 failed to alter the blood pressure in SGK1 knockout mice. Following chronic (4 weeks) fructose/high salt treatment, additional EMD638683 treatment again decreased blood pressure. EMD638683 thus abrogates the salt sensitivity of blood pressure in hyperinsulinism without appreciably affecting blood pressure in the absence of hyperinsulinism. EMD638683 tended to increase fluid intake and urinary excretion of Na(+), significantly increased urinary flow rate and significantly decreased body weight. CONCLUSION EMD638683 could serve as a template for drugs counteracting hypertension in individuals with type II diabetes and metabolic syndrome.
Collapse
|
36
|
Umbach AT, Pathare G, Föller M, Brosens JJ, Artunc F, Lang F. SGK1-dependent salt appetite in pregnant mice. Acta Physiol (Oxf) 2011; 202:39-45. [PMID: 21251236 DOI: 10.1111/j.1748-1716.2011.02251.x] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
AIM Pregnancy is typically paralleled by substantial increase in maternal extracellular fluid volume, requiring net accumulation of water and NaCl. The positive water and salt balance is accomplished at least in part by increased uptake of salt secondary to enhanced salt appetite. Little is known about the underlying cellular mechanisms. Stimulation of salt appetite by mineralocorticoids, however, is known to be dependent on the serum- and glucocorticoid-inducible kinase SGK1. METHODS To test for a role of SGK1 in the stimulation of salt appetite during pregnancy, fluid intake was recorded in pregnant SGK1 knockout mice (sgk1(-/-) ) and their wild type littermates (sgk1(+/+) ). The mice were offered two bottles, one with plain water and the other with isotonic saline. RESULTS In early pregnancy, i.e. up to 10 days prior to parturition, the sgk1(+/+) mice displayed a significant preference for saline, whereas the sgk1(-/-) mice preferred water. Accordingly, the water intake was significantly smaller and saline intake was significantly larger in sgk1(+/+) mice than in sgk1(-/-) mice and the preference for water was significantly stronger in sgk1(-/-) mice than in sgk1(+/+) mice. Plasma aldosterone levels were higher in sgk1(-/-) mice than in sgk1(+/+) mice, a difference contrasting the enhanced salt appetite of sgk1(+/+) mice. CONCLUSIONS SGK1 participates in the stimulation of salt appetite during pregnancy.
Collapse
Affiliation(s)
- A T Umbach
- Department of Physiology, University of Tübingen, Germany
| | | | | | | | | | | |
Collapse
|
37
|
Boini KM, Zhang C, Xia M, Poklis JL, Li PL. Role of sphingolipid mediator ceramide in obesity and renal injury in mice fed a high-fat diet. J Pharmacol Exp Ther 2010; 334:839-46. [PMID: 20543095 DOI: 10.1124/jpet.110.168815] [Citation(s) in RCA: 80] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
The present study tested a hypothesis that excess accumulation of sphingolipid, ceramide, its metabolites, or a combination contributes to the development of obesity and associated kidney damage. Liquid chromatography/mass spectrometry analysis demonstrated that C57BL/6J mice on the high-fat diet (HFD) had significantly increased plasma total ceramide levels compared with animals fed a low-fat diet (LFD). Treatment of mice with the acid sphingomyelinase (ASMase) inhibitor amitriptyline significantly attenuated the HFD-induced plasma ceramide levels. Corresponding to increase in plasma ceramide, the HFD significantly increased the body weight gain, plasma leptin concentration, urinary total protein and albumin excretion, glomerular damage index, and adipose tissue ASMase activity compared with the LFD-fed mice. These HFD-induced changes were also significantly attenuated by treatment of mice with amitriptyline. In addition, the decline of plasma glucose concentration after an intraperitoneal injection of insulin (0.15 U/kg b.wt.) was more sustained in mice on the HFD with amitriptyline than on the HFD alone. Intraperitoneal injection of glucose (3 g/kg b.wt.) resulted in a slow increase followed by a rapid decrease in the plasma glucose concentration in LFD and HFD plus amitriptyline-treated mice, but such blood glucose response was not observed in HFD-fed mice. Immunofluorescence analysis demonstrated a decrease in the podocin and an increase in the desmin in the glomeruli of HFD-fed mice compared with the LFD and HFD plus amitriptyline-treated mice. In conclusion, our results reveal a pivotal role for ceramide biosynthesis in obesity, metabolic syndrome, and associated kidney damage.
Collapse
Affiliation(s)
- Krishna M Boini
- Department of Pharmacology and Toxicology, Medical College of Virginia Campus, Virginia Commonwealth University, 410 N, 12th St., Richmond, VA 23298, USA
| | | | | | | | | |
Collapse
|
38
|
The physiological impact of the serum and glucocorticoid-inducible kinase SGK1. Curr Opin Nephrol Hypertens 2009; 18:439-48. [PMID: 19584721 DOI: 10.1097/mnh.0b013e32832f125e] [Citation(s) in RCA: 113] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
PURPOSE OF REVIEW The role of serum and glucocorticoid-inducible kinase 1 (SGK1) in renal physiology and pathophysiology is reviewed with particular emphasis on recent advances. RECENT FINDINGS The mammalian target of rapamycin complex 2 has been shown to phosphorylate SGK1 at Ser422 (the so-called hydrophobic motif). Ser397 and Ser401 are two additional SGK1-phosphorylation sites required for maximal SGK1 activity. A 5' variant alternate transcript of human Sgk1 has been identified that is widely expressed and shows improved stability, enhanced membrane association, and greater stimulation of epithelial Na+ transport. SGK1 is essential for optimal processing of the epithelial sodium channel and also regulates the expression of the Na+-Cl- cotransporter. With regard to pathophysiology, SGK1 participates in the stimulation of renal tubular glucose transport in diabetes, the renal profibrotic effect of both angiotensin II and aldosterone, and in fetal programing of arterial hypertension. SUMMARY The outlined recent findings advanced our understanding of the molecular regulation of SGK1 as well as the role of the kinase in renal physiology and the pathophysiology of renal disease and hypertension. Future studies using pharmacological inhibitors of SGK1 will reveal the utility of the kinase as a new therapeutic target.
Collapse
|
39
|
Sgk1 activates MDM2-dependent p53 degradation and affects cell proliferation, survival, and differentiation. J Mol Med (Berl) 2009; 87:1221-39. [PMID: 19756449 DOI: 10.1007/s00109-009-0525-5] [Citation(s) in RCA: 75] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2009] [Revised: 08/05/2009] [Accepted: 08/20/2009] [Indexed: 12/11/2022]
Abstract
Serum and glucocorticoid regulated kinase 1 (Sgk1) is a serine-threonine kinase that is activated by serum, steroids, insulin, vasopressin, and interleukin 2 at the transcriptional and post-translational levels. Sgk1 is also important in transduction of growth factors and steroid-dependent survival signals and may have a role in the development of resistance to cancer chemotherapy. In the present paper, we demonstrate that Sgk1 activates MDM2-dependent p53 ubiquitylation. The results were obtained in RKO cells and other cell lines by Sgk1-specific RNA silencing and were corroborated in an original mouse model as well as in transiently and in stably transfected HeLa cells expressing wild-type or dominant negative Sgk1 mutant. Sgk1 contributes to cell survival, cell-cycle progression, and epithelial de-differentiation. We also show that the effects of Sgk1 on the clonogenic potential of different cancer cells depend on the expression of wild-type p53. Since transcription of Sgk1 is activated by p53, we propose a finely tuned feedback model where Sgk1 down-regulates the expression of p53 by enhancing its mono- and polyubiquitylation.
Collapse
|
40
|
Loffing J, Korbmacher C. Regulated sodium transport in the renal connecting tubule (CNT) via the epithelial sodium channel (ENaC). Pflugers Arch 2009; 458:111-35. [PMID: 19277701 DOI: 10.1007/s00424-009-0656-0] [Citation(s) in RCA: 126] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2009] [Revised: 02/18/2009] [Accepted: 02/22/2009] [Indexed: 12/29/2022]
Abstract
The aldosterone-sensitive distal nephron (ASDN) includes the late distal convoluted tubule 2, the connecting tubule (CNT) and the collecting duct. The appropriate regulation of sodium (Na(+)) absorption in the ASDN is essential to precisely match urinary Na(+) excretion to dietary Na(+) intake whilst taking extra-renal Na(+) losses into account. There is increasing evidence that Na(+) transport in the CNT is of particular importance for the maintenance of body Na(+) balance and for the long-term control of extra-cellular fluid volume and arterial blood pressure. Na(+) transport in the CNT critically depends on the activity and abundance of the amiloride-sensitive epithelial sodium channel (ENaC) in the luminal membrane of the CNT cells. As a rate-limiting step for transepithelial Na(+) transport, ENaC is the main target of hormones (e.g. aldosterone, angiotensin II, vasopressin and insulin/insulin-like growth factor 1) to adjust transepithelial Na(+) transport in this tubular segment. In this review, we highlight the structural and functional properties of the CNT that contribute to the high Na(+) transport capacity of this segment. Moreover, we discuss some aspects of the complex pathways and molecular mechanisms involved in ENaC regulation by hormones, kinases, proteases and associated proteins that control its function. Whilst cultured cells and heterologous expression systems have greatly advanced our knowledge about some of these regulatory mechanisms, future studies will have to determine the relative importance of the various pathways in the native tubule and in particular in the CNT.
Collapse
|
41
|
Hirata K, Kume S, Araki SI, Sakaguchi M, Chin-Kanasaki M, Isshiki K, Sugimoto T, Nishiyama A, Koya D, Haneda M, Kashiwagi A, Uzu T. Exendin-4 has an anti-hypertensive effect in salt-sensitive mice model. Biochem Biophys Res Commun 2009; 380:44-9. [PMID: 19150338 DOI: 10.1016/j.bbrc.2009.01.003] [Citation(s) in RCA: 114] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2008] [Accepted: 01/05/2009] [Indexed: 12/20/2022]
Abstract
The improvement of salt-sensitive hypertension is a therapeutic target for various vascular diseases. Glucagon-like peptide 1 (GLP-1), an incretin peptide, has been reported to have natriuretic effect as well as blood glucose lowering effect, although its exact mechanism and clinical usefulness remain unclear. Here, we examined anti-hypertensive effect of exendin-4, a GLP-1 analog, in salt-sensitive obese db/db mice and angiotensin II (angII)-infused C57BLK6/J mice. The treatment of exendin-4 for 12 weeks inhibited the development of hypertension in db/db mice. In db/db mice, the urinary sodium excretion was delayed and blood pressure was elevated in response to a high-salt load, whereas these were attenuated by exendin-4. In db/db mice, intra-renal angII concentration was increased. Furthermore, exendin-4 prevented angII-induced hypertension in non-diabetic mice and inhibited angII-induced phosphorylation of ERK1/2 in cultured renal cells. Considered together, our results indicate that exendin-4 has anti-hypertensive effects through the attenuation of angII-induced high-salt sensitivity.
Collapse
Affiliation(s)
- Kunio Hirata
- Department of Medicine, Shiga University of Medical Science, Tsukinowa-cho, Seta, Otsu, Shiga 520-2192, Japan
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
42
|
Yasuhara A, Wada J, Malakauskas SM, Zhang Y, Eguchi J, Nakatsuka A, Murakami K, Kanzaki M, Teshigawara S, Yamagata K, Le TH, Makino H. Collectrin is involved in the development of salt-sensitive hypertension by facilitating the membrane trafficking of apical membrane proteins via interaction with soluble N-ethylmaleiamide-sensitive factor attachment protein receptor complex. Circulation 2008; 118:2146-55. [PMID: 18981302 DOI: 10.1161/circulationaha.108.787259] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
BACKGROUND Collectrin, a homologue of angiotensin converting enzyme 2, is expressed in pancreatic beta cells and renal proximal tubular and collecting duct cells under the control of hepatocyte nuclear factors-1alpha and -1beta. Because collectrin interacts with the soluble N-ethylmaleiamide-sensitive factor attachment protein receptor (SNARE) complexes, we investigated whether collectrin is involved in sodium handling in hypertension by vesicle trafficking of apical membrane proteins. METHODS AND RESULTS Collectrin physically interacts with the SNARE complex: snapin, synaptosomal-associated protein 23 kDa, syntaxin-4, and vesicle-associated membrane protein-2 in mIMCD-3 cells. siRNA knockdown of collectrin resulted in a reduction in membrane-associated aquaporin-2, alpha-epithelial Na+ channel, and H+-ATPase. Collectrin and SNARE proteins were abundantly expressed in collecting ducts of Wistar-Kyoto rats. Wistar-Kyoto rats and spontaneously hypertensive rats 7 weeks of age were subjected to normal-salt (1% NaCl) and high-salt (8% NaCl) chow for 10 weeks. High-salt chow prominently elevated blood pressure, oral intake, and urinary excretion of NaCl and water in both groups. Although urinary excretion of aldosterone was significantly suppressed in both groups, collectrin expression was upregulated and associated with the maintenance of aquaporin-2, alpha-epithelial Na+ channel, and H+-ATPase in membrane fractions. Collectrin promoter activities and mRNA and protein expressions were upregulated and ubiquitinated collectrin was reduced by high NaCl (175 to 225 mmol/L) and not altered by 1 micromol/L aldosterone in mIMCD-3 cells. CONCLUSIONS Upregulation of collectrin by high NaCl independent of aldosterone functionally links to the trafficking of apical membrane proteins via the SNARE complex, and collectrin may be responsible for the sodium retention in salt-sensitive hypertension.
Collapse
Affiliation(s)
- Akihiro Yasuhara
- Department of Medicine and Clinical Science, Okayama University Graduate School of Medicine, 2-5-1 Shikata-cho, Okayama 700-8558, Japan
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
43
|
SGK1 activity in Na+ absorbing airway epithelial cells monitored by assaying NDRG1-Thr346/356/366 phosphorylation. Pflugers Arch 2008; 457:1287-301. [PMID: 18787837 DOI: 10.1007/s00424-008-0587-1] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2008] [Accepted: 08/29/2008] [Indexed: 01/02/2023]
Abstract
Studies of HeLa cells and serum- and glucocorticoid-regulated kinase 1 (SGK1) knockout mice identified threonine residues in the n-myc downstream-regulated gene 1 protein (NDRG1-Thr(346/356/366)) that are phosphorylated by SGK1 but not by related kinases (Murray et al., Biochem J 385:1-12, 2005). We have, therefore, monitored the phosphorylation of NDRG1-Thr(346/356/366) in order to explore the changes in SGK1 activity associated with the induction and regulation of the glucocorticoid-dependent Na(+) conductance (G (Na)) in human airway epithelial cells. Transient expression of active (SGK1-S422D) and inactive (SGK1-K127A) SGK1 mutants confirmed that activating SGK1 stimulates NDRG1-Thr(346/356/366) phosphorylation. Although G (Na) is negligible in hormone-deprived cells, these cells displayed basal SGK1 activity that was sensitive to LY294002, an inhibitor of 3-phosphatidylinositol phosphate kinase (PI3K). Dexamethasone (0.2 muM) acutely activated SGK1 and the peak of this response (2-3 h) coincided with the induction of G (Na), and both responses were PI3K-dependent. While these data suggest that SGK1 might mediate the rise in G (Na), transient expression of the inactive SGK1-K127A mutant did not affect the hormonal induction of G (Na) but did suppress the activation of SGK1. Dexamethasone-treated cells grown on permeable supports formed confluent epithelial sheets that generated short circuit current due to electrogenic Na(+) absorption. Forskolin and insulin both stimulated this current and the response to insulin, but not forskolin, was LY294002-sensitive and associated with the activation of SGK1. While these data suggest that SGK1 is involved in the control of G (Na), its role may be minor, which could explain why sgk1 knockout has different effects upon different tissues.
Collapse
|
44
|
Bhalla V, Hallows KR. Mechanisms of ENaC regulation and clinical implications. J Am Soc Nephrol 2008; 19:1845-54. [PMID: 18753254 DOI: 10.1681/asn.2008020225] [Citation(s) in RCA: 180] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022] Open
Abstract
The epithelial Na+ channel (ENaC) transports Na+ across tight epithelia, including the distal nephron. Different paradigms of ENaC regulation include extrinsic and intrinsic factors that affect the expression, single-channel properties, and intracellular trafficking of the channel. In particular, recent discoveries highlight new findings regarding proteolytic processing, ubiquitination, and recycling of the channel. Understanding the regulation of this channel is critical to the understanding of various clinical phenomena, including normal physiology and several diseases of kidney and lung epithelia, such as blood pressure (BP) control, edema, and airway fluid clearance. Significant progress has been achieved in this active field of research. Although ENaC is classically thought to be a mediator of BP and volume status through Na+ reabsorption in the distal nephron, several studies in animal models highlight important roles for ENaC in lung pathophysiology, including in cystic fibrosis. The purpose of this review is to highlight the various modes and mechanisms of ENaC regulation, with a focus on more recent studies and their clinical implications.
Collapse
Affiliation(s)
- Vivek Bhalla
- Division of Nephrology, Department of Medicine, Stanford University School of Medicine, Stanford, California, USA
| | | |
Collapse
|
45
|
SGK1 dependence of insulin induced hypokalemia. Pflugers Arch 2008; 457:955-61. [PMID: 18665390 DOI: 10.1007/s00424-008-0559-5] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2008] [Revised: 06/23/2008] [Accepted: 07/11/2008] [Indexed: 12/23/2022]
Abstract
Insulin stimulates cellular K+ uptake leading to hypokalemia. Cellular K+ uptake is accomplished by parallel stimulation of Na+/H+ exchange, Na+,K+,2Cl- co-transport, and Na+/K+ ATPase and leads to cell swelling, a prerequisite for several metabolic effects of the hormone. Little is known about underlying signaling. Insulin is known to activate the serum and glucocorticoid-inducible kinase SGK1, which in turn enhances the activity of all three transport proteins. The present study thus explored the contribution of SGK1 to insulin-induced hypokalemia. To this end, gene-targeted mice lacking SGK1 (sgk1-/-) and their wild-type littermates (sgk1+/+) have been infused with insulin (2 mU kg(-1) min(-1)) and glucose at rates leaving the plasma glucose concentration constant. Moreover, isolated liver perfusion experiments have been performed to determine stimulation of cellular K+ uptake by insulin (100 nM). As a result, combined glucose and insulin infusion significantly decreased plasma K+ concentration despite a significant decrease of urinary K+ excretion in sgk1+/+ but not in sgk1-/- mice. Accordingly, the plasma K+ concentration was within 60 min significantly lower in sgk1+/+ than in sgk1-/- mice. In isolated liver perfusion experiments, cellular K+ uptake was stimulated by insulin (100 nM), an effect blunted by 72% in sgk1-/- mice as compared to sgk1+/+ mice. Accordingly, insulin-induced cell hydration was 63% lower in sgk1-/- mice than in sgk1+/+ mice. Moreover, volume regulatory K+ release was 31% smaller in sgk1-/- mice than in sgk1+/+ mice. In conclusion, the serum and glucocorticoid-inducible kinase SGK1 participates in the signaling mediating the hypokalemic effect of insulin.
Collapse
|
46
|
Grenz A, Osswald H, Eckle T, Yang D, Zhang H, Tran ZV, Klingel K, Ravid K, Eltzschig HK. The reno-vascular A2B adenosine receptor protects the kidney from ischemia. PLoS Med 2008; 5:e137. [PMID: 18578565 PMCID: PMC2504049 DOI: 10.1371/journal.pmed.0050137] [Citation(s) in RCA: 173] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/05/2007] [Accepted: 05/09/2008] [Indexed: 11/25/2022] Open
Abstract
BACKGROUND Acute renal failure from ischemia significantly contributes to morbidity and mortality in clinical settings, and strategies to improve renal resistance to ischemia are urgently needed. Here, we identified a novel pathway of renal protection from ischemia using ischemic preconditioning (IP). METHODS AND FINDINGS For this purpose, we utilized a recently developed model of renal ischemia and IP via a hanging weight system that allows repeated and atraumatic occlusion of the renal artery in mice, followed by measurements of specific parameters or renal functions. Studies in gene-targeted mice for each individual adenosine receptor (AR) confirmed renal protection by IP in A1(-/-), A2A(-/-), or A3AR(-/-) mice. In contrast, protection from ischemia was abolished in A2BAR(-/-) mice. This protection was associated with corresponding changes in tissue inflammation and nitric oxide production. In accordance, the A2BAR-antagonist PSB1115 blocked renal protection by IP, while treatment with the selective A2BAR-agonist BAY 60-6583 dramatically improved renal function and histology following ischemia alone. Using an A2BAR-reporter model, we found exclusive expression of A2BARs within the reno-vasculature. Studies using A2BAR bone-marrow chimera conferred kidney protection selectively to renal A2BARs. CONCLUSIONS These results identify the A2BAR as a novel therapeutic target for providing potent protection from renal ischemia.
Collapse
Affiliation(s)
- Almut Grenz
- Department of Pharmacology and Toxicology, Tübingen University Hospital, Tübingen, Germany
- Mucosal Inflammation Program, Department of Anesthesiology and Perioperative Medicine, University of Colorado Health Sciences Center, Denver, Colorado, United States of America
| | - Hartmut Osswald
- Department of Pharmacology and Toxicology, Tübingen University Hospital, Tübingen, Germany
| | - Tobias Eckle
- Mucosal Inflammation Program, Department of Anesthesiology and Perioperative Medicine, University of Colorado Health Sciences Center, Denver, Colorado, United States of America
- Department of Anesthesiology and Intensive Care Medicine, Tübingen University Hospital, Tübingen, Germany
| | - Dan Yang
- Department of Biochemistry and Whitaker Cardiovascular Institute, Boston University School of Medicine, Boston, Massachusetts, United States of America
| | - Hua Zhang
- Department of Pharmacology and Toxicology, Tübingen University Hospital, Tübingen, Germany
| | - Zung Vu Tran
- Department of Biostatistics, University of Colorado, Denver, Colorado, United States of America
| | - Karin Klingel
- Department of Molecular Pathology, Tübingen University Hospital, Tübingen, Germany
| | - Katya Ravid
- Department of Biochemistry and Whitaker Cardiovascular Institute, Boston University School of Medicine, Boston, Massachusetts, United States of America
| | - Holger K Eltzschig
- Mucosal Inflammation Program, Department of Anesthesiology and Perioperative Medicine, University of Colorado Health Sciences Center, Denver, Colorado, United States of America
- Department of Anesthesiology and Intensive Care Medicine, Tübingen University Hospital, Tübingen, Germany
- * To whom correspondence should be addressed. E-mail:
| |
Collapse
|
47
|
Abstract
Aldosterone concentrations are inappropriately high in many patients with hypertension, as well as in an increasing number of individuals with metabolic syndrome and sleep apnoea. A growing body of evidence suggests that aldosterone and/or activation of the MR (mineralocorticoid receptor) contributes to cardiovascular remodelling and renal injury in these conditions. In addition to causing sodium retention and increased blood pressure, MR activation induces oxidative stress, endothelial dysfunction, inflammation and subsequent fibrosis. The MR may be activated by aldosterone and cortisol or via transactivation by the AT(1) (angiotenin II type 1) receptor through a mechanism involving the EGFR (epidermal growth factor receptor) and MAPK (mitogen-activated protein kinase) pathway. In addition, aldosterone can generate rapid non-genomic effects in the heart and vasculature. MR antagonism reduces mortality in patients with CHF (congestive heart failure) and following myocardial infarction. MR antagonism improves endothelial function in patients with CHF, reduces circulating biomarkers of cardiac fibrosis in CHF or following myocardial infarction, reduces blood pressure in resistant hypertension and decreases albuminuria in hypertensive and diabetic patients. In contrast, whereas adrenalectomy improves glucose homoeostasis in hyperaldosteronism, MR antagonism may worsen glucose homoeostasis and impairs endothelial function in diabetes, suggesting a possible detrimental effect of aldosterone via non-genomic pathways.
Collapse
Affiliation(s)
- Annis M Marney
- Division of Clinical Pharmacology, Departments of Medicine and Pharmacology, Vanderbilt University Medical Center, Nashville, TN 37232-6602, USA
| | | |
Collapse
|
48
|
Colussi G, Catena C, Lapenna R, Nadalini E, Chiuch A, Sechi LA. Insulin resistance and hyperinsulinemia are related to plasma aldosterone levels in hypertensive patients. Diabetes Care 2007; 30:2349-54. [PMID: 17575088 DOI: 10.2337/dc07-0525] [Citation(s) in RCA: 95] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
Abstract
OBJECTIVE An association between aldosterone and insulin resistance has been demonstrated in obesity and primary aldosteronism and in blacks with the metabolic syndrome. The aim of this study was to evaluate the relationship of plasma aldosterone with insulin sensitivity in white subjects. RESEARCH DESIGN AND METHODS In 356 patients with essential hypertension and 102 normotensive control subjects of comparable age and BMI, we measured, after discontinuation of treatment, plasma active renin, aldosterone, cortisol, glucose, insulin, and C-peptide levels and calculated markers of insulin sensitivity. Direct assessment of insulin sensitivity was obtained in a subset of 64 hypertensive patients by a hyperinsulinemic clamp. RESULTS Hypertensive patients had significantly greater fasting plasma insulin and C-peptide concentrations and homeostasis model assessment (HOMA) indexes than normotensive control subjects. A positive association with increasing plasma aldosterone concentrations was demonstrated for plasma glucose, insulin, C-peptides, and HOMA. Assessment of insulin sensitivity by clamp showed a significant decrease of the metabolic clearance rate of glucose with increasing aldosterone levels. Significant correlations were found between plasma aldosterone, plasma insulin, and C-peptide levels, HOMA, and glucose metabolic clearance rate. Blood pressure and plasma potassium, plasma cortisol, and renin levels, but not BMI, were also directly correlated with plasma aldosterone. Multiple regression analysis showed that HOMA, together with plasma potassium, cortisol, and renin levels, was independently correlated with plasma aldosterone. CONCLUSIONS This study demonstrates a direct relationship between aldosterone, insulin resistance, and hyperinsulinemia in white subjects. In patients with hypertension, this relationship might contribute to maintenance of high blood pressure and increased cardiovascular risk.
Collapse
Affiliation(s)
- Gianluca Colussi
- Hypertension and Diabetes Unit, Division of Internal Medicine, Department of Experimental and Clinical Pathology and Medicine, University of Udine, Udine, Italy
| | | | | | | | | | | |
Collapse
|
49
|
Tiwari S, Riazi S, Ecelbarger CA. Insulin's impact on renal sodium transport and blood pressure in health, obesity, and diabetes. Am J Physiol Renal Physiol 2007; 293:F974-84. [PMID: 17686957 DOI: 10.1152/ajprenal.00149.2007] [Citation(s) in RCA: 95] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023] Open
Abstract
Insulin has been shown to have antinatriuretic actions in humans and animal models. Moreover, endogenous hyperinsulinemia and insulin infusion have been correlated to increased blood pressure in some models. In this review, we present the current state of understanding with regard to the regulation of the major renal sodium transporters by insulin in the kidney. Several groups, using primarily cell culture, have demonstrated that insulin can directly increase activity of the epithelial sodium channel, the sodium-phosphate cotransporter, the sodium-hydrogen exchanger type III, and Na-K-ATPase. We and others have demonstrated alterations in the expression at the protein level of many of these same proteins with insulin infusion or in hyperinsulinemic models. We also discuss how this regulation is perturbed in type I and type II diabetes mellitus. Finally, we discuss a potential role for regulation of insulin receptor signaling in the kidney in contributing to sodium balance and blood pressure.
Collapse
Affiliation(s)
- Swasti Tiwari
- Division of Endocrinology and Metabolism, Department of Medicine, Georgetown University, Washington, District of Columbia 20007, USA
| | | | | |
Collapse
|
50
|
Abstract
Insulin has important vascular actions to stimulate production of nitric oxide from endothelium. This leads to capillary recruitment, vasodilation, increased blood flow, and subsequent augmentation of glucose disposal in classical insulin target tissues (e.g., skeletal muscle). Phosphatidylinositol 3-kinase-dependent insulin-signaling pathways regulating endothelial production of nitric oxide share striking parallels with metabolic insulin-signaling pathways. Distinct MAPK-dependent insulin-signaling pathways (largely unrelated to metabolic actions of insulin) regulate secretion of the vasoconstrictor endothelin-1 from endothelium. These and other cardiovascular actions of insulin contribute to coupling metabolic and hemodynamic homeostasis under healthy conditions. Cardiovascular diseases are the leading cause of morbidity and mortality in insulin-resistant individuals. Insulin resistance is typically defined as decreased sensitivity and/or responsiveness to metabolic actions of insulin. This cardinal feature of diabetes, obesity, and dyslipidemia is also a prominent component of hypertension, coronary heart disease, and atherosclerosis that are all characterized by endothelial dysfunction. Conversely, endothelial dysfunction is often present in metabolic diseases. Insulin resistance is characterized by pathway-specific impairment in phosphatidylinositol 3-kinase-dependent signaling that in vascular endothelium contributes to a reciprocal relationship between insulin resistance and endothelial dysfunction. The clinical relevance of this coupling is highlighted by the findings that specific therapeutic interventions targeting insulin resistance often also ameliorate endothelial dysfunction (and vice versa). In this review, we discuss molecular mechanisms underlying cardiovascular actions of insulin, the reciprocal relationships between insulin resistance and endothelial dysfunction, and implications for developing beneficial therapeutic strategies that simultaneously target metabolic and cardiovascular diseases.
Collapse
Affiliation(s)
- Ranganath Muniyappa
- Diabetes Unit, National Center for Complementary and Alternative Medicine, National Institutes of Health, Bethesda, Maryland 20892-1632, USA
| | | | | | | |
Collapse
|