1
|
Watanabe G, Horita S, Moriya RF, Masuishi Y, Misaka S, Taira S, Shimomura K, Shimabukuro M, Kazama JJ. Canagliflozin-induced renal glutathione distribution mapping in non-diabetic male rat kidneys. Physiol Rep 2025; 13:e70320. [PMID: 40223360 PMCID: PMC11994861 DOI: 10.14814/phy2.70320] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2024] [Revised: 03/17/2025] [Accepted: 04/01/2025] [Indexed: 04/15/2025] Open
Abstract
Canagliflozin, a sodium glucose cotransporter 2 (SGLT2) inhibitor, has direct renoprotective effects beyond lowering blood glucose levels. The inhibition of sodium reabsorption via SGLT2 reduces the overload on proximal tubules, thereby suppressing the generation of reactive oxygen species (ROS) and preventing a decline in renal function. To clarify the pharmacological mechanism of SGLT2 inhibitor, we investigated the effects of canagliflozin on oxidative stress in the kidneys of normal, non-diabetic Sprague-Dawley rats. Screening using mass spectrometry images revealed a significant elevation map of the reduced form of glutathione in the renal cortex of canagliflozin-treated non-diabetic rats. These results suggest that canagliflozin reduces oxidative stress through ROS scavenging mechanisms. Considering that ROS play major roles in renal dysfunction regardless of diabetes mellitus, these findings suggest that canagliflozin is applicable to a broader range of renal diseases beyond diabetes.
Collapse
Affiliation(s)
- Guy Watanabe
- Department of Nephrology and HypertensionFukushima Medical University School of MedicineFukushimaJapan
- Department of Bioregulation and Pharmacological MedicineFukushima Medical University School of MedicineFukushimaJapan
| | - Shoichiro Horita
- Department of Diabetes, Endocrinology, and MetabolismFukushima Medical University School of MedicineFukushimaJapan
| | - Reika Flora Moriya
- Department of Nephrology and HypertensionFukushima Medical University School of MedicineFukushimaJapan
| | - Yusuke Masuishi
- Department of Hygiene and Preventive MedicineFukushima Medical University School of MedicineFukushimaJapan
| | - Shingen Misaka
- Department of Bioregulation and Pharmacological MedicineFukushima Medical University School of MedicineFukushimaJapan
| | - Shu Taira
- Faculty of Food and Agricultural SciencesFukushima UniversityFukushimaJapan
| | - Kenju Shimomura
- Department of Bioregulation and Pharmacological MedicineFukushima Medical University School of MedicineFukushimaJapan
| | - Michio Shimabukuro
- Department of Diabetes, Endocrinology, and MetabolismFukushima Medical University School of MedicineFukushimaJapan
| | - Junichiro James Kazama
- Department of Nephrology and HypertensionFukushima Medical University School of MedicineFukushimaJapan
| |
Collapse
|
2
|
Amatya B, Polzin JQM, Villar VAM, Yang J, Konkalmatt P, Wang X, Cadme RC, Xu P, Gildea JJ, Cuevas S, Armando I, Felder RA, Jose PA, Lee H. SNX19 Interacts with Caveolin-1 and Flotillin-1 to Regulate D 1R Endocytosis and Signaling. Biomedicines 2025; 13:481. [PMID: 40002894 PMCID: PMC11853350 DOI: 10.3390/biomedicines13020481] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2025] [Revised: 02/04/2025] [Accepted: 02/07/2025] [Indexed: 02/27/2025] Open
Abstract
Background: Sorting nexin 19 (SNX19) is important in the localization and trafficking of the dopamine D1 receptor (D1R) to lipid raft microdomains. However, the interaction between SNX19 and the lipid raft components caveolin-1 or flotillin-1 and, in particular, their roles in the cellular endocytosis and cell membrane trafficking of the D1R have not been determined. Methods: Caveolin-1 and flotillin-1 motifs were analyzed by in silico analysis; colocalization was observed by confocal immunofluorescence microscopy; protein-protein interaction was determined by co-immunoprecipitation. Results: In silico analysis revealed the presence of putative caveolin-1 and flotillin-1 binding motifs within SNX19. In mouse and human renal proximal tubule cells (RPTCs), SNX19 was localized mainly in lipid rafts. In mouse RPTCs transfected with wild-type (WT) Snx19, fenoldopam (FEN), a D1-like receptor agonist, increased the colocalization of SNX19 with caveolin-1 and flotillin-1. FEN also increased the co-immunoprecipitation of SNX19 with caveolin-1 and flotillin-1, effects that were prevented by SCH39166, a D1-like receptor antagonist. The FEN-mediated increase in the residence of SNX19 in lipid rafts and the colocalization of the D1R with caveolin-1 and flotilin-1 were attenuated by the deletion of a caveolin-1 (YHTVNRRYREF) (ΔCav1) or a flotillin-1 (EEGPGTETETGLPVS) (ΔFlot1) binding motif. The FEN-mediated increase in intracellular cAMP production was also impaired by the deletion of either the flotillin-1 or caveolin-1 binding motif. Nocodazole, a microtubule depolymerization inhibitor, interfered with the FEN-mediated increase in the colocalization between SNX19 and D1R. Conclusion: SNX19 contains caveolin-1 and flotillin-1 binding motifs, which play an important role in D1R endocytosis and signaling.
Collapse
Affiliation(s)
- Bibhas Amatya
- Division of Kidney Diseases & Hypertension, Department of Medicine, The George Washington University School of Medicine & Health Sciences, Washington, DC 20052, USA; (B.A.); (J.Q.M.P.); (V.A.M.V.); (P.K.); (X.W.); (R.C.C.); (S.C.); (I.A.); (P.A.J.)
- Department of Medicine, University of Maryland School of Medicine, Baltimore, MD 21201, USA
| | - Jacob Q. M. Polzin
- Division of Kidney Diseases & Hypertension, Department of Medicine, The George Washington University School of Medicine & Health Sciences, Washington, DC 20052, USA; (B.A.); (J.Q.M.P.); (V.A.M.V.); (P.K.); (X.W.); (R.C.C.); (S.C.); (I.A.); (P.A.J.)
| | - Van A. M. Villar
- Division of Kidney Diseases & Hypertension, Department of Medicine, The George Washington University School of Medicine & Health Sciences, Washington, DC 20052, USA; (B.A.); (J.Q.M.P.); (V.A.M.V.); (P.K.); (X.W.); (R.C.C.); (S.C.); (I.A.); (P.A.J.)
- Division of Nephrology, Department of Medicine, University of Maryland School of Medicine, 20 Penn Street, HSF II, Baltimore, MD 21201, USA;
| | - Jiang Yang
- Division of Nephrology, Department of Medicine, University of Maryland School of Medicine, 20 Penn Street, HSF II, Baltimore, MD 21201, USA;
- Department of Clinical Nutrition, The Third Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China
| | - Prasad Konkalmatt
- Division of Kidney Diseases & Hypertension, Department of Medicine, The George Washington University School of Medicine & Health Sciences, Washington, DC 20052, USA; (B.A.); (J.Q.M.P.); (V.A.M.V.); (P.K.); (X.W.); (R.C.C.); (S.C.); (I.A.); (P.A.J.)
- Division of Nephrology, Department of Medicine, University of Maryland School of Medicine, 20 Penn Street, HSF II, Baltimore, MD 21201, USA;
| | - Xiaoyan Wang
- Division of Kidney Diseases & Hypertension, Department of Medicine, The George Washington University School of Medicine & Health Sciences, Washington, DC 20052, USA; (B.A.); (J.Q.M.P.); (V.A.M.V.); (P.K.); (X.W.); (R.C.C.); (S.C.); (I.A.); (P.A.J.)
- Division of Nephrology, Department of Medicine, University of Maryland School of Medicine, 20 Penn Street, HSF II, Baltimore, MD 21201, USA;
- Department of Nephrology, BenQ Medical Center, The Affiliated BenQ Hospital of Nanjing Medical University, Nanjing 210029, China
| | - Raisha C. Cadme
- Division of Kidney Diseases & Hypertension, Department of Medicine, The George Washington University School of Medicine & Health Sciences, Washington, DC 20052, USA; (B.A.); (J.Q.M.P.); (V.A.M.V.); (P.K.); (X.W.); (R.C.C.); (S.C.); (I.A.); (P.A.J.)
| | - Peng Xu
- Department of Pathology, University of Virginia Health Sciences Center, Charlottesville, VA 22908, USA; (P.X.); (J.J.G.); (R.A.F.)
| | - John J. Gildea
- Department of Pathology, University of Virginia Health Sciences Center, Charlottesville, VA 22908, USA; (P.X.); (J.J.G.); (R.A.F.)
| | - Santiago Cuevas
- Division of Kidney Diseases & Hypertension, Department of Medicine, The George Washington University School of Medicine & Health Sciences, Washington, DC 20052, USA; (B.A.); (J.Q.M.P.); (V.A.M.V.); (P.K.); (X.W.); (R.C.C.); (S.C.); (I.A.); (P.A.J.)
- Division of Nephrology, Department of Medicine, University of Maryland School of Medicine, 20 Penn Street, HSF II, Baltimore, MD 21201, USA;
- Physiopathology of the Inflammation and Oxidative Stress Laboratory, Molecular Inflammation Group, Biomedical Research Institute of Murcia Pascual Parrilla (IMIB), 30120 Palmar, Spain
| | - Ines Armando
- Division of Kidney Diseases & Hypertension, Department of Medicine, The George Washington University School of Medicine & Health Sciences, Washington, DC 20052, USA; (B.A.); (J.Q.M.P.); (V.A.M.V.); (P.K.); (X.W.); (R.C.C.); (S.C.); (I.A.); (P.A.J.)
- Division of Nephrology, Department of Medicine, University of Maryland School of Medicine, 20 Penn Street, HSF II, Baltimore, MD 21201, USA;
| | - Robin A. Felder
- Department of Pathology, University of Virginia Health Sciences Center, Charlottesville, VA 22908, USA; (P.X.); (J.J.G.); (R.A.F.)
| | - Pedro A. Jose
- Division of Kidney Diseases & Hypertension, Department of Medicine, The George Washington University School of Medicine & Health Sciences, Washington, DC 20052, USA; (B.A.); (J.Q.M.P.); (V.A.M.V.); (P.K.); (X.W.); (R.C.C.); (S.C.); (I.A.); (P.A.J.)
- Division of Nephrology, Department of Medicine, University of Maryland School of Medicine, 20 Penn Street, HSF II, Baltimore, MD 21201, USA;
- Department of Pharmacology & Physiology, The George Washington University School of Medicine & Health Sciences, Washington, DC 20052, USA
| | - Hewang Lee
- Division of Kidney Diseases & Hypertension, Department of Medicine, The George Washington University School of Medicine & Health Sciences, Washington, DC 20052, USA; (B.A.); (J.Q.M.P.); (V.A.M.V.); (P.K.); (X.W.); (R.C.C.); (S.C.); (I.A.); (P.A.J.)
- Division of Nephrology, Department of Medicine, University of Maryland School of Medicine, 20 Penn Street, HSF II, Baltimore, MD 21201, USA;
| |
Collapse
|
3
|
Cole ES, Maier W, Joachimiak E, Jiang YY, Lee C, Collet E, Chmelik C, Romero DP, Chalker D, Alli NK, Ruedlin TM, Ozzello C, Gaertig J. The Tetrahymena bcd1 mutant implicates endosome trafficking in ciliate, cortical pattern formation. Mol Biol Cell 2023; 34:ar82. [PMID: 37163326 PMCID: PMC10398878 DOI: 10.1091/mbc.e22-11-0501] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2022] [Revised: 03/15/2023] [Accepted: 05/01/2023] [Indexed: 05/11/2023] Open
Abstract
Ciliates, such as Tetrahymena thermophila, evolved complex mechanisms to determine both the location and dimensions of cortical organelles such as the oral apparatus (OA: involved in phagocytosis), cytoproct (Cyp: for eliminating wastes), and contractile vacuole pores (CVPs: involved in water expulsion). Mutations have been recovered in Tetrahymena that affect both the localization of such organelles along anterior-posterior and circumferential body axes and their dimensions. Here we describe BCD1, a ciliate pattern gene that encodes a conserved Beige-BEACH domain-containing protein a with possible protein kinase A (PKA)-anchoring activity. Similar proteins have been implicated in endosome trafficking and are linked to human Chediak-Higashi syndrome and autism. Mutations in the BCD1 gene broaden cortical organelle domains as they assemble during predivision development. The Bcd1 protein localizes to membrane pockets at the base of every cilium that are active in endocytosis. PKA activity has been shown to promote endocytosis in other organisms, so we blocked clathrin-mediated endocytosis (using "dynasore") and inhibited PKA (using H89). In both cases, treatment produced partial phenocopies of the bcd1 pattern mutant. This study supports a model in which the dimensions of diverse cortical organelle assembly-platforms may be determined by regulated balance between constitutive exocytic delivery and PKA-regulated endocytic retrieval of organelle materials and determinants.
Collapse
Affiliation(s)
- Eric S. Cole
- Biology Department, St. Olaf College, Northfield, MN 55057
| | - Wolfgang Maier
- Bioinformatics Group, Department of Computer Science, University of Freiburg, 79110 Freiburg, Germany
| | - Ewa Joachimiak
- Laboratory of Cytoskeleton and Cilia Biology, Nencki Institute of Experimental Biology of Polish Academy of Sciences, 02-093 Warsaw, Poland
| | - Yu-yang Jiang
- Molecular Genetics and Cell Biology, The University of Chicago, Chicago, IL 60637
| | - Chinkyu Lee
- Department of Cellular Biology, University of Georgia, Athens, GA 30605
| | - Erik Collet
- Department of Cell and Developmental Biology, University of Colorado Anschutz Medical Campus, Aurora, CO 80045
| | - Carl Chmelik
- Biology Department, St. Olaf College, Northfield, MN 55057
| | - Daniel P. Romero
- Department of Pharmacology, University of Minnesota, Minneapolis, MN 55455
| | - Douglas Chalker
- Department of Biology, Washington University in St. Louis, St. Louis, MO 63021
| | - Nurudeen K. Alli
- Department of Biology, Washington University in St. Louis, St. Louis, MO 63021
| | - Tina M. Ruedlin
- Department of Biology, Washington University in St. Louis, St. Louis, MO 63021
| | - Courtney Ozzello
- Molecular, Cellular, and Developmental Biology, University of Colorado Boulder, Boulder, CO 80309
| | - Jacek Gaertig
- Department of Cellular Biology, University of Georgia, Athens, GA 30605
| |
Collapse
|
4
|
Mukherji ST, Brambilla L, Stuart KB, Mayes I, Kutz LC, Chen Y, Barbosa LA, Elmadbouh I, McDermott JP, Haller ST, Romero MF, Soleimani M, Liu J, Shapiro JI, Blanco GV, Xie Z, Pierre SV. Na/K-ATPase signaling tonically inhibits sodium reabsorption in the renal proximal tubule. FASEB J 2023; 37:e22835. [PMID: 36856735 PMCID: PMC10028530 DOI: 10.1096/fj.202200785rr] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2022] [Revised: 02/08/2023] [Accepted: 02/10/2023] [Indexed: 03/02/2023]
Abstract
Through its classic ATP-dependent ion-pumping function, basolateral Na/K-ATPase (NKA) generates the Na+ gradient that drives apical Na+ reabsorption in the renal proximal tubule (RPT), primarily through the Na+ /H+ exchanger (NHE3). Accordingly, activation of NKA-mediated ion transport decreases natriuresis through activation of basolateral (NKA) and apical (NHE3) Na+ reabsorption. In contrast, activation of the more recently discovered NKA signaling function triggers cellular redistribution of RPT NKA and NHE3 and decreases Na+ reabsorption. We used gene targeting to test the respective contributions of NKA signaling and ion pumping to the overall regulation of RPT Na+ reabsorption. Knockdown of RPT NKA in cells and mice increased membrane NHE3 and Na+ /HCO3 - cotransporter (NBCe1A). Urine output and absolute Na+ excretion decreased by 65%, driven by increased RPT Na+ reabsorption (as indicated by decreased lithium clearance and unchanged glomerular filtration rate), and accompanied by elevated blood pressure. This hyper reabsorptive phenotype was rescued upon crossing with RPT NHE3-/- mice, confirming the importance of NKA/NHE3 coupling. Hence, NKA signaling exerts a tonic inhibition on Na+ reabsorption by regulating key apical and basolateral Na+ transporters. This action, lifted upon NKA genetic suppression, tonically counteracts NKA's ATP-driven function of basolateral Na+ reabsorption. Strikingly, NKA signaling is not only physiologically relevant but it also appears to be functionally dominant over NKA ion pumping in the control of RPT reabsorption.
Collapse
Affiliation(s)
- Shreya T. Mukherji
- Marshall Institute for Interdisciplinary Research, Marshall University, Huntington, WV
| | - Luca Brambilla
- Marshall Institute for Interdisciplinary Research, Marshall University, Huntington, WV
| | - Kailey B. Stuart
- Marshall Institute for Interdisciplinary Research, Marshall University, Huntington, WV
| | - Isabella Mayes
- Marshall Institute for Interdisciplinary Research, Marshall University, Huntington, WV
| | - Laura C. Kutz
- Marshall Institute for Interdisciplinary Research, Marshall University, Huntington, WV
| | - Yiliang Chen
- Department of Medicine, Medical College of Wisconsin, Milwaukee, Wisconsin
- Blood Research Institute, Versiti, WI
| | - Leandro A Barbosa
- Marshall Institute for Interdisciplinary Research, Marshall University, Huntington, WV
- Universidade Federal de São João del-Rei, Campus Centro-Oeste Dona Lindu, Divinópolis, MG, Brazil
| | - Ibrahim Elmadbouh
- Marshall Institute for Interdisciplinary Research, Marshall University, Huntington, WV
| | - Jeff P. McDermott
- Department of Molecular and Integrative Physiology, University of Kansas Medical Center, Kansas City, KS
| | - Steven T. Haller
- Department of Medicine, University of Toledo College of Medicine and Life Sciences, Toledo, OH
| | - Michael F. Romero
- Physiology & Biomedical Engineering and Nephrology & Hypertension, Mayo Clinic College of Medicine & Science, Rochester, MN
| | - Manoocher Soleimani
- Department of Medicine, The University of New Mexico Health Sciences Center, Albuquerque, NM
| | - Jiang Liu
- Joan C. Edwards School of Medicine, Marshall University, Huntington, WV
| | - Joseph I. Shapiro
- Joan C. Edwards School of Medicine, Marshall University, Huntington, WV
| | - Gustavo V. Blanco
- Department of Molecular and Integrative Physiology, University of Kansas Medical Center, Kansas City, KS
| | - Zijian Xie
- Marshall Institute for Interdisciplinary Research, Marshall University, Huntington, WV
| | - Sandrine V. Pierre
- Marshall Institute for Interdisciplinary Research, Marshall University, Huntington, WV
| |
Collapse
|
5
|
Nikolovska K, Seidler UE, Stock C. The Role of Plasma Membrane Sodium/Hydrogen Exchangers in Gastrointestinal Functions: Proliferation and Differentiation, Fluid/Electrolyte Transport and Barrier Integrity. Front Physiol 2022; 13:899286. [PMID: 35665228 PMCID: PMC9159811 DOI: 10.3389/fphys.2022.899286] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2022] [Accepted: 04/19/2022] [Indexed: 12/11/2022] Open
Abstract
The five plasma membrane Na+/H+ exchanger (NHE) isoforms in the gastrointestinal tract are characterized by distinct cellular localization, tissue distribution, inhibitor sensitivities, and physiological regulation. NHE1 (Slc9a1) is ubiquitously expressed along the gastrointestinal tract in the basolateral membrane of enterocytes, but so far, an exclusive role for NHE1 in enterocyte physiology has remained elusive. NHE2 (Slc9a2) and NHE8 (Slc9a8) are apically expressed isoforms with ubiquitous distribution along the colonic crypt axis. They are involved in pHi regulation of intestinal epithelial cells. Combined use of a knockout mouse model, intestinal organoid technology, and specific inhibitors revealed previously unrecognized actions of NHE2 and NHE8 in enterocyte proliferation and differentiation. NHE3 (Slc9a3), expressed in the apical membrane of differentiated intestinal epithelial cells, functions as the predominant nutrient-independent Na+ absorptive mechanism in the gut. The new selective NHE3 inhibitor (Tenapanor) allowed discovery of novel pathophysiological and drug-targetable NHE3 functions in cystic-fibrosis associated intestinal obstructions. NHE4, expressed in the basolateral membrane of parietal cells, is essential for parietal cell integrity and acid secretory function, through its role in cell volume regulation. This review focuses on the expression, regulation and activity of the five plasma membrane Na+/H+ exchangers in the gastrointestinal tract, emphasizing their role in maintaining intestinal homeostasis, or their impact on disease pathogenesis. We point to major open questions in identifying NHE interacting partners in central cellular pathways and processes and the necessity of determining their physiological role in a system where their endogenous expression/activity is maintained, such as organoids derived from different parts of the gastrointestinal tract.
Collapse
|
6
|
Nwia SM, Li XC, Leite APDO, Hassan R, Zhuo JL. The Na +/H + Exchanger 3 in the Intestines and the Proximal Tubule of the Kidney: Localization, Physiological Function, and Key Roles in Angiotensin II-Induced Hypertension. Front Physiol 2022; 13:861659. [PMID: 35514347 PMCID: PMC9062697 DOI: 10.3389/fphys.2022.861659] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2022] [Accepted: 02/25/2022] [Indexed: 01/29/2023] Open
Abstract
The sodium (Na+)/hydrogen (H+) exchanger 3 (NHE3) is one of the most important Na+/H+ antiporters in the small intestines of the gastrointestinal tract and the proximal tubules of the kidney. The roles of NHE3 in the regulation of intracellular pH and acid-base balance have been well established in cellular physiology using in vitro techniques. Localized primarily on the apical membranes in small intestines and proximal tubules, the key action of NHE3 is to facilitate the entry of luminal Na+ and the extrusion of intracellular H+ from intestinal and proximal tubule tubular epithelial cells. NHE3 is, directly and indirectly, responsible for absorbing the majority of ingested Na+ from small and large intestines and reabsorbing >50% of filtered Na+ in the proximal tubules of the kidney. However, the roles of NHE3 in the regulation of proximal tubular Na+ transport in the integrative physiological settings and its contributions to the basal blood pressure regulation and angiotensin II (Ang II)-induced hypertension have not been well studied previously due to the lack of suitable animal models. Recently, novel genetically modified mouse models with whole-body, kidney-specific, or proximal tubule-specific deletion of NHE3 have been generated by us and others to determine the critical roles and underlying mechanisms of NHE3 in maintaining basal body salt and fluid balance, blood pressure homeostasis, and the development of Ang II-induced hypertension at the whole-body, kidney, or proximal tubule levels. The objective of this invited article is to review, update, and discuss recent findings on the critical roles of intestinal and proximal tubule NHE3 in maintaining basal blood pressure homeostasis and their potential therapeutic implications in the development of angiotensin II (Ang II)-dependent hypertension.
Collapse
Affiliation(s)
- Sarah M. Nwia
- Tulane Hypertension and Renal Center of Excellence, Tulane University School of Medicine, New Orleans, LA, United States,Department of Physiology, Tulane University School of Medicine, New Orleans, LA, United States
| | - Xiao Chun Li
- Tulane Hypertension and Renal Center of Excellence, Tulane University School of Medicine, New Orleans, LA, United States,Department of Physiology, Tulane University School of Medicine, New Orleans, LA, United States
| | - Ana Paula de Oliveira Leite
- Tulane Hypertension and Renal Center of Excellence, Tulane University School of Medicine, New Orleans, LA, United States,Department of Physiology, Tulane University School of Medicine, New Orleans, LA, United States
| | - Rumana Hassan
- Tulane Hypertension and Renal Center of Excellence, Tulane University School of Medicine, New Orleans, LA, United States,Department of Physiology, Tulane University School of Medicine, New Orleans, LA, United States
| | - Jia Long Zhuo
- Tulane Hypertension and Renal Center of Excellence, Tulane University School of Medicine, New Orleans, LA, United States,Department of Physiology, Tulane University School of Medicine, New Orleans, LA, United States,*Correspondence: Jia Long Zhuo,
| |
Collapse
|
7
|
Han Y, Yun CC. Metformin Inhibits Na +/H + Exchanger NHE3 Resulting in Intestinal Water Loss. Front Physiol 2022; 13:867244. [PMID: 35444557 PMCID: PMC9014215 DOI: 10.3389/fphys.2022.867244] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2022] [Accepted: 03/07/2022] [Indexed: 11/14/2022] Open
Abstract
Glycemic control is the key to the management of type 2 diabetes. Metformin is an effective, widely used drug for controlling plasma glucose levels in diabetes, but it is often the culprit of gastrointestinal adverse effects such as abdominal pain, nausea, indigestion, vomiting, and diarrhea. Diarrhea is a complex disease and altered intestinal transport of electrolytes and fluid is a common cause of diarrhea. Na+/H+ exchanger 3 (NHE3, SLC9A3) is the major Na+ absorptive mechanism in the intestine and our previous study has demonstrated that decreased NHE3 contributes to diarrhea associated with type 1 diabetes. The goal of this study is to investigate whether metformin regulates NHE3 and inhibition of NHE3 contributes to metformin-induced diarrhea. We first determined whether metformin alters intestinal water loss, the hallmark of diarrhea, in type 2 diabetic db/db mice. We found that metformin decreased intestinal water absorption mediated by NHE3. Metformin increased fecal water content although mice did not develop watery diarrhea. To determine the mechanism of metformin-mediated regulation of NHE3, we used intestinal epithelial cells. Metformin inhibited NHE3 activity and the effect of metformin on NHE3 was mimicked by a 5'-AMP-activated protein kinase (AMPK) activator and blocked by pharmacological inhibition of AMPK. Metformin increased phosphorylation and ubiquitination of NHE3, resulting in retrieval of NHE3 from the plasma membrane. Previous studies have demonstrated the role of neural precursor cell expressed, developmentally down-regulated 4-2 (Nedd4-2) in regulation of human NHE3. Silencing of Nedd4-2 mitigated NHE3 inhibition and ubiquitination by metformin. Our findings suggest that metformin-induced diarrhea in type 2 diabetes is in part caused by reduced Na+ and water absorption that is associated with NHE3 inhibition, probably by AMPK.
Collapse
Affiliation(s)
- Yiran Han
- Gastroenterology Research, Atlanta Veterans Administration Medical Center, Decatur, GA, United States
- Division of Digestive Diseases, Department of Medicine, Emory University School of Medicine, Atlanta, GA, United States
| | - C. Chris Yun
- Gastroenterology Research, Atlanta Veterans Administration Medical Center, Decatur, GA, United States
- Division of Digestive Diseases, Department of Medicine, Emory University School of Medicine, Atlanta, GA, United States
- Winship Cancer Institute, Emory University School of Medicine, Atlanta, GA, United States
| |
Collapse
|
8
|
Leite APDO, Li XC, Nwia SM, Hassan R, Zhuo JL. Angiotensin II and AT 1a Receptors in the Proximal Tubules of the Kidney: New Roles in Blood Pressure Control and Hypertension. Int J Mol Sci 2022; 23:2402. [PMID: 35269547 PMCID: PMC8910592 DOI: 10.3390/ijms23052402] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2022] [Revised: 02/15/2022] [Accepted: 02/18/2022] [Indexed: 02/01/2023] Open
Abstract
Contrary to public perception, hypertension remains one of the most important public health problems in the United States, affecting 46% of adults with increased risk for heart attack, stroke, and kidney diseases. The mechanisms underlying poorly controlled hypertension remain incompletely understood. Recent development in the Cre/LoxP approach to study gain or loss of function of a particular gene has significantly helped advance our new insights into the role of proximal tubule angiotensin II (Ang II) and its AT1 (AT1a) receptors in basal blood pressure control and the development of Ang II-induced hypertension. This novel approach has provided us and others with an important tool to generate novel mouse models with proximal tubule-specific loss (deletion) or gain of the function (overexpression). The objective of this invited review article is to review and discuss recent findings using novel genetically modifying proximal tubule-specific mouse models. These new studies have consistently demonstrated that deletion of AT1 (AT1a) receptors or its direct downstream target Na+/H+ exchanger 3 (NHE3) selectively in the proximal tubules of the kidney lowers basal blood pressure, increases the pressure-natriuresis response, and induces natriuretic responses, whereas overexpression of an intracellular Ang II fusion protein or AT1 (AT1a) receptors selectively in the proximal tubules increases proximal tubule Na+ reabsorption, impairs the pressure-natriuresis response, and elevates blood pressure. Furthermore, the development of Ang II-induced hypertension by systemic Ang II infusion or by proximal tubule-specific overexpression of an intracellular Ang II fusion protein was attenuated in mutant mice with proximal tubule-specific deletion of AT1 (AT1a) receptors or NHE3. Thus, these recent studies provide evidence for and new insights into the important roles of intratubular Ang II via AT1 (AT1a) receptors and NHE3 in the proximal tubules in maintaining basal blood pressure homeostasis and the development of Ang II-induced hypertension.
Collapse
Affiliation(s)
- Ana Paula de Oliveira Leite
- Tulane Hypertension and Renal Center of Excellence, 1430 Tulane Avenue, New Orleans, LA 70112, USA; (A.P.d.O.L.); (X.C.L.); (S.M.N.); (R.H.)
- Department of Physiology, Tulane University School of Medicine, New Orleans, LA 70112, USA
| | - Xiao C. Li
- Tulane Hypertension and Renal Center of Excellence, 1430 Tulane Avenue, New Orleans, LA 70112, USA; (A.P.d.O.L.); (X.C.L.); (S.M.N.); (R.H.)
- Department of Physiology, Tulane University School of Medicine, New Orleans, LA 70112, USA
| | - Sarah M. Nwia
- Tulane Hypertension and Renal Center of Excellence, 1430 Tulane Avenue, New Orleans, LA 70112, USA; (A.P.d.O.L.); (X.C.L.); (S.M.N.); (R.H.)
- Department of Physiology, Tulane University School of Medicine, New Orleans, LA 70112, USA
| | - Rumana Hassan
- Tulane Hypertension and Renal Center of Excellence, 1430 Tulane Avenue, New Orleans, LA 70112, USA; (A.P.d.O.L.); (X.C.L.); (S.M.N.); (R.H.)
- Department of Physiology, Tulane University School of Medicine, New Orleans, LA 70112, USA
| | - Jia L. Zhuo
- Tulane Hypertension and Renal Center of Excellence, 1430 Tulane Avenue, New Orleans, LA 70112, USA; (A.P.d.O.L.); (X.C.L.); (S.M.N.); (R.H.)
- Department of Physiology, Tulane University School of Medicine, New Orleans, LA 70112, USA
| |
Collapse
|
9
|
Ran L, Yan T, Zhang Y, Niu Z, Kan Z, Song Z. The recycling regulation of sodium-hydrogen exchanger isoform 3(NHE3) in epithelial cells. Cell Cycle 2021; 20:2565-2582. [PMID: 34822321 DOI: 10.1080/15384101.2021.2005274] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/19/2022] Open
Abstract
As the main exchanger of electroneutral NaCl absorption, sodium-hydrogen exchanger isoform 3 (NHE3) circulates in the epithelial brush border (BB) and intracellular compartments in a multi-protein complex. The size of the NHE3 complex changes during rapid regulation events. Recycling regulation of NHE3 in epithelial cells can be roughly divided into three stages. First, when stimulated by Ca2+, cGMP, and cAMP-dependent signaling pathways, NHE3 is converted from an immobile complex found at the apical microvilli (MV) into an easily internalized and mobile form that relocates to a compartment near the base of the MV. Second, NHE3 is internalized by clathrin and albumin-dependent pathways into cytoplasmic endosomal compartments, where the complex is reprocessed and reassembled. Finally, NHE3 is translocated from the recycling endosomes (REs) to the apex of epithelial cells, a process that can be stimulated by an increase in sodium-glucose cotransporter 1 (SGLT1) activity, epidermal growth factor receptor (EGFR) signaling, Ca2+ signaling, and binding to βPix and SH3 and multiple ankyrin repeat domains 2 (Shank2) proteins. This review describes the molecular steps and protein interactions involved in the recycling movement of NHE3 from the apex of epithelial cells, into vesicles, where it is reprocessed and reassembled, and returned to its original location on the plasma membrane, where it exerts its physiological function.
Collapse
Affiliation(s)
- Ling Ran
- Department of Preventive Veterinary Medicine, College of Veterinary Medicine, Southwest University, Rongchang, China
| | - Tao Yan
- Department of Preventive Veterinary Medicine, College of Veterinary Medicine, Xinjiang Agricultural University, Urumqi, China
| | - Yiling Zhang
- Department of Preventive Veterinary Medicine, College of Veterinary Medicine, Southwest University, Rongchang, China
| | - Zheng Niu
- Department of Preventive Veterinary Medicine, College of Veterinary Medicine, Southwest University, Rongchang, China
| | - Zifei Kan
- Department of Preventive Veterinary Medicine, College of Veterinary Medicine, Southwest University, Rongchang, China
| | - Zhenhui Song
- Department of Preventive Veterinary Medicine, College of Veterinary Medicine, Southwest University, Rongchang, China
| |
Collapse
|
10
|
Jenkin KA, Han Y, Lin S, He P, Yun CC. Nedd4-2-dependent Ubiquitination Potentiates the Inhibition of Human NHE3 by Cholera Toxin and Enteropathogenic Escherichia coli. Cell Mol Gastroenterol Hepatol 2021; 13:695-716. [PMID: 34823064 PMCID: PMC8789535 DOI: 10.1016/j.jcmgh.2021.11.006] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/27/2021] [Revised: 11/12/2021] [Accepted: 11/15/2021] [Indexed: 12/20/2022]
Abstract
BACKGROUND & AIMS Diarrhea is one of the most common illnesses and is often caused by bacterial infection. Recently, we have shown that human Na+/H+ exchanger NHE3 (hNHE3), but not non-human NHE3s, interacts with the E3 ubiquitin ligase Nedd4-2. We hypothesize that this property of hNHE3 contributes to the increased severity of diarrhea in humans. METHODS We used humanized mice expressing hNHE3 in the intestine (hNHE3int) to compare the contribution of hNHE3 and mouse NHE3 to diarrhea induced by cholera toxin (CTX) and enteropathogenic Escherichia coli (EPEC). We measured Na+/H+ exchange activity and fluid absorption. The role of Nedd4-2 on hNHE3 activity and ubiquitination was determined by knockdown in Caco-2bbe cells. The effects of protein kinase A (PKA), the primary mediator of CTX-induced diarrhea, on Nedd4-2 and hNHE3 phosphorylation and their interaction were determined. RESULTS The effects of CTX and EPEC were greater in hNHE3int mice than in control wild-type (WT) mice, resulting in greater inhibition of NHE3 activity and increased fluid accumulation in the intestine, the hallmark of diarrhea. Activation of PKA increased ubiquitination of hNHE3 and enhanced interaction of Nedd4-2 with hNHE3 via phosphorylation of Nedd4-2 at S342. S342A mutation mitigated the Nedd4-2-hNHE3 interaction and blocked PKA-induced inhibition of hNHE3. Unlike non-human NHE3s, inhibition of hNHE3 by PKA is independent of NHE3 phosphorylation, suggesting a distinct mechanism of hNHE3 regulation. CONCLUSIONS The effects of CTX and EPEC on hNHE3 are amplified, and the unique properties of hNHE3 may contribute to diarrheal symptoms occurring in humans.
Collapse
Affiliation(s)
- Kayte A. Jenkin
- Division of Digestive Diseases, Department of Medicine, Emory University School of Medicine, Atlanta, Georgia,School of Science, Western Sydney University, Campbelltown, NSW 2560, Australia
| | - Yiran Han
- Division of Digestive Diseases, Department of Medicine, Emory University School of Medicine, Atlanta, Georgia,Atlanta VA Medical Center, Decatur, Georgia
| | - Songbai Lin
- Division of Digestive Diseases, Department of Medicine, Emory University School of Medicine, Atlanta, Georgia,Atlanta VA Medical Center, Decatur, Georgia
| | - Peijian He
- Division of Digestive Diseases, Department of Medicine, Emory University School of Medicine, Atlanta, Georgia
| | - C. Chris Yun
- Division of Digestive Diseases, Department of Medicine, Emory University School of Medicine, Atlanta, Georgia,Atlanta VA Medical Center, Decatur, Georgia,Winship Cancer Institute, Emory University School of Medicine, Atlanta, Georgia,Correspondence Address correspondence to: Chris Yun, PhD, Division of Digestive Diseases, Emory University School of Medicine, Atlanta, Georgia 30324. fax: (404) 727-5767.
| |
Collapse
|
11
|
New Insights into the Critical Importance of Intratubular Na +/H + Exchanger 3 and Its Potential Therapeutic Implications in Hypertension. Curr Hypertens Rep 2021; 23:34. [PMID: 34110521 DOI: 10.1007/s11906-021-01152-7] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/17/2021] [Indexed: 01/08/2023]
Abstract
PURPOSE OF REVIEW The sodium (Na+) and hydrogen (H+) exchanger 3 (NHE3), known as solute carrier family 9 member 3 (SLC9A3), mediates active transcellular Na+ and bicarbonate reabsorption in the small intestine of the gut and proximal tubules of the kidney. The purpose of this article is to review and discuss recent findings on the critical roles of intestinal and proximal tubule NHE3 in maintaining basal blood pressure (BP) homeostasis and their potential therapeutic implications in the development of angiotensin II (Ang II)-dependent hypertension. RECENT FINDINGS Recently, our and other laboratories have generated or used novel genetically modified mouse models with whole-body, kidney-specific, or proximal tubule-specific deletion of NHE3 to determine the critical roles and underlying mechanisms of NHE3 in maintaining basal BP homeostasis and the development of Ang II-induced hypertension at the whole-body, kidney, or proximal tubule levels. The new findings demonstrate that NHE3 contributes to about 10 to 15 mmHg to basal blood pressure levels, and that deletion of NHE3 at the whole-kidney or proximal tubule level, or pharmacological inhibition of NHE3 at the kidney level with an orally absorbable NHE3 inhibitor AVE-0657, attenuates ~ 50% of Ang II-induced hypertension in mice. The results support the proof-of-concept hypothesis that NHE3 plays critical roles in physiologically maintaining normal BP and in the development of Ang II-dependent hypertension. Our results also strongly suggest that NHE3 in the proximal tubules of the kidney may be therapeutically targeted to treat poorly controlled hypertension in humans.
Collapse
|
12
|
Du Z, Tian X, Ma M, Somlo S, Weinstein AM, Wang T. Restoration of proximal tubule flow-activated transport prevents cyst growth in polycystic kidney disease. JCI Insight 2021; 6:146041. [PMID: 33886508 PMCID: PMC8262298 DOI: 10.1172/jci.insight.146041] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2020] [Accepted: 04/21/2021] [Indexed: 01/11/2023] Open
Abstract
Flow-activated Na+ and HCO3– transport in kidney proximal tubules (PT) underlies relatively constant fractional reabsorption during changes in glomerular filtration rate (GFR) or glomerulotubular balance (GTB). In view of hypothesized connections of epithelial cilia to flow sensing, we examined flow-activated transport in 3 polycystic kidney disease–related mouse models based on inducible conditional KO of Pkd1, Pkd2, and Kif3a. PTs were harvested from mice after gene inactivation but prior to cyst formation, and flow-mediated PT transport was measured. We confirm that higher flow increased both Na+ and HCO3– absorption in control mice, and we observed that this flow effect was preserved in PTs of Pkd1–/– and Kif3a–/–mice. However, flow activation was absent in Pkd2+/– and Pkd2–/– PT. In heterozygous (Pkd2+/–) mice, a dopamine receptor 1 (DA1) antagonist (SCH23390) restored transport flow sensitivity. When given chronically, this same antagonist reduced renal cyst formation in Pkd2–/–, as evidenced by reduced kidney weight, BUN, and the cystic index, when compared with untreated mice. In contrast, SCH23390 did not prevent cyst formation in Pkd1–/– mice. These results indicate that Pkd2 is necessary for normal GTB and that restoration of flow-activated transport by DA1 antagonist can slow renal cyst formation in Pkd2–/– mice.
Collapse
Affiliation(s)
| | - Xin Tian
- Department of Internal Medicine (Nephrology), Yale University, New Haven, Connecticut, USA
| | - Ming Ma
- Department of Internal Medicine (Nephrology), Yale University, New Haven, Connecticut, USA
| | - Stefan Somlo
- Department of Internal Medicine (Nephrology), Yale University, New Haven, Connecticut, USA
| | - Alan M Weinstein
- Department of Physiology and Biophysics, Weill Medical College of Cornell University, New York, New York, USA
| | | |
Collapse
|
13
|
Lipid Rafts and Dopamine Receptor Signaling. Int J Mol Sci 2020; 21:ijms21238909. [PMID: 33255376 PMCID: PMC7727868 DOI: 10.3390/ijms21238909] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2020] [Revised: 11/18/2020] [Accepted: 11/20/2020] [Indexed: 02/07/2023] Open
Abstract
The renal dopaminergic system has been identified as a modulator of sodium balance and blood pressure. According to the Centers for Disease Control and Prevention, in 2018 in the United States, almost half a million deaths included hypertension as a primary or contributing cause. Renal dopamine receptors, members of the G protein-coupled receptor family, are divided in two groups: D1-like receptors that act to keep the blood pressure in the normal range, and D2-like receptors with a variable effect on blood pressure, depending on volume status. The renal dopamine receptor function is regulated, in part, by its expression in microdomains in the plasma membrane. Lipid rafts form platforms within the plasma membrane for the organization and dynamic contact of molecules involved in numerous cellular processes such as ligand binding, membrane sorting, effector specificity, and signal transduction. Understanding all the components of lipid rafts, their interaction with renal dopamine receptors, and their signaling process offers an opportunity to unravel potential treatment targets that could halt the progression of hypertension, chronic kidney disease (CKD), and their complications.
Collapse
|
14
|
Klinger S. Segment-specific effects of resveratrol on porcine small intestinal dipeptide absorption depend on the mucosal pH and are due to different mechanisms: potential roles of different transport proteins and protein kinases. J Nutr Biochem 2020; 85:108467. [PMID: 32738496 DOI: 10.1016/j.jnutbio.2020.108467] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2019] [Revised: 07/02/2020] [Accepted: 07/13/2020] [Indexed: 12/15/2022]
Abstract
Numerous beneficial features of the polyphenol resveratrol (RSV) have been demonstrated in several tissues and cell culture models. There is also evidence, that RSV impairs intestinal nutrient transport but the underlying mechanisms are not understood. The aim of the present study was to evaluate whether RSV has also an impact on the H+-coupled transport of peptides via the peptide transporter 1 (PepT1) and to characterize RSV mediated changes in the apical abundance of nutrients transport proteins and protein kinases that may be involved. RSV decreased the H+-coupled transport of peptides in the porcine small intestines in a pH and location specific manner (jejunum vs ileum) as measured in Ussing chamber experiments. The comparison of the effects of RSV with the effects of the cAMP/PKA-activating agent forskolin indicates that different mechanisms may be responsible in the intestinal segments. Additionally, it seems that the transport of peptides and glucose in the jejunum are inhibited via the same mechanism while there might be two mechanisms involved in the ileum. Functional data and protein expression data indicate, that, besides PepT1, the activity of the Na+/H+-exchanger 3 (NHE3) may be involved. Protein kinase A (PKA) and AMP-activated kinase (AMPK) are both activated by RSV while the extracellular regulated kinase (ERK) and the serum and glucocorticoid induced kinase (SGK) are widely unaffected. Although PKA and AMPK are activated, AMPK seems not to be related to the effects of RSV. Additionally, both the functional data and the protein expression data reveal some interesting pH- and segment-specific differences.
Collapse
Affiliation(s)
- Stefanie Klinger
- Institute for Physiology and Cell Biology, University of Veterinary Medicine Hannover, Foundation, Hannover, Germany.
| |
Collapse
|
15
|
Shi M, Maique J, Shaffer J, Davidson T, Sebti S, Fernández ÁF, Zou Z, Yan S, Levine B, Moe OW, Hu MC. The tripartite interaction of phosphate, autophagy, and αKlotho in health maintenance. FASEB J 2020; 34:3129-3150. [PMID: 31908069 PMCID: PMC7286356 DOI: 10.1096/fj.201902127r] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2019] [Revised: 12/19/2019] [Accepted: 12/19/2019] [Indexed: 12/11/2022]
Abstract
Aging-related organ degeneration is driven by multiple factors including the cell maintenance mechanisms of autophagy, the cytoprotective protein αKlotho, and the lesser known effects of excess phosphate (Pi), or phosphotoxicity. To examine the interplay between Pi, autophagy, and αKlotho, we used the BK/BK mouse (homozygous for mutant Becn1F121A ) with increased autophagic flux, and αKlotho-hypomorphic mouse (kl/kl) with impaired urinary Pi excretion, low autophagy, and premature organ dysfunction. BK/BK mice live longer than WT littermates, and have heightened phosphaturia from downregulation of two key NaPi cotransporters in the kidney. The multi-organ failure in kl/kl mice was rescued in the double-mutant BK/BK;kl/kl mice exhibiting lower plasma Pi, improved weight gain, restored plasma and renal αKlotho levels, decreased pathology of multiple organs, and improved fertility compared to kl/kl mice. The beneficial effects of heightened autophagy from Becn1F121A was abolished by chronic high-Pi diet which also shortened life span in the BK/BK;kl/kl mice. Pi promoted beclin 1 binding to its negative regulator BCL2, which impairs autophagy flux. Pi downregulated αKlotho, which also independently impaired autophagy. In conclusion, Pi, αKlotho, and autophagy interact intricately to affect each other. Both autophagy and αKlotho antagonizes phosphotoxicity. In concert, this tripartite system jointly determines longevity and life span.
Collapse
Affiliation(s)
- Mingjun Shi
- Charles and Jane Pak Center for Mineral Metabolism and Clinical Research, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Jenny Maique
- Charles and Jane Pak Center for Mineral Metabolism and Clinical Research, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Joy Shaffer
- Charles and Jane Pak Center for Mineral Metabolism and Clinical Research, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Taylor Davidson
- Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Salwa Sebti
- Center for Autophagy Research, University of Texas Southwestern Medical Center, Dallas, TX, USA
- Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Álvaro F. Fernández
- Center for Autophagy Research, University of Texas Southwestern Medical Center, Dallas, TX, USA
- Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Zhongju Zou
- Center for Autophagy Research, University of Texas Southwestern Medical Center, Dallas, TX, USA
- Department of Howard Hughes Medical Institute, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Shirley Yan
- Department of Pathology, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Beth Levine
- Center for Autophagy Research, University of Texas Southwestern Medical Center, Dallas, TX, USA
- Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, TX, USA
- Department of Microbiology, University of Texas Southwestern Medical Center, Dallas, TX, USA
- Department of Howard Hughes Medical Institute, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Orson W. Moe
- Charles and Jane Pak Center for Mineral Metabolism and Clinical Research, University of Texas Southwestern Medical Center, Dallas, TX, USA
- Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, TX, USA
- Department of Physiology, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Ming Chang Hu
- Charles and Jane Pak Center for Mineral Metabolism and Clinical Research, University of Texas Southwestern Medical Center, Dallas, TX, USA
- Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, TX, USA
| |
Collapse
|
16
|
Ranjan P, Awasthi M, Snell WJ. Transient Internalization and Microtubule-Dependent Trafficking of a Ciliary Signaling Receptor from the Plasma Membrane to the Cilium. Curr Biol 2019; 29:2942-2947.e2. [PMID: 31422889 DOI: 10.1016/j.cub.2019.07.022] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2019] [Revised: 07/03/2019] [Accepted: 07/09/2019] [Indexed: 12/24/2022]
Abstract
Cilia are ancient organelles used by unicellular and multicellular organisms not only for motility but also to receive and respond to multiple environmental cues, including light, odorants, morphogens, growth factors, and contact with cilia of other cells. Much is known about the cellular mechanisms that deliver membrane proteins to cilia during ciliogenesis. Execution of a ciliary signaling pathway, however, can critically depend on rapid alterations in the receptor composition of the cilium itself, and our understanding of the mechanisms that underlie these rapid, regulated alterations remains limited [1-6]. In the bi-ciliated, unicellular alga Chlamydomonas reinhardtii, interactions between cilia of mating type plus and mating type minus gametes mediated by adhesion receptors SAG1 and SAD1 activate a ciliary signaling pathway [7]. In response, a large, inactive pool of SAG1 on the plasma membrane of plus gametes rapidly becomes enriched in the peri-ciliary membrane and enters the cilia to become active and maintain and enhance ciliary adhesion and signaling [8-14]. Ciliary entry per se of SAG1 is independent of anterograde intraflagellar transport (IFT) [13], but the rapid apical enrichment requires cytoplasmic microtubules and the retrograde IFT motor, dynein 1b [14]. Whether the receptors move laterally within the plasma membrane or transit internally during redistribution is unknown. Here, in coupled immunolocalization/biochemical studies on SAG1, we show that, within minutes after gamete activation is initiated, cell-surface SAG1 is internalized, associates with an apico-basally polarized array of cytoplasmic microtubules, and returns to the cell surface at a peri-ciliary staging area for entry into cilia.
Collapse
Affiliation(s)
- Peeyush Ranjan
- Department of Cell Biology and Molecular Genetics, University of Maryland, College Park, MD 20742, USA
| | - Mayanka Awasthi
- Department of Cell Biology and Molecular Genetics, University of Maryland, College Park, MD 20742, USA
| | - William J Snell
- Department of Cell Biology and Molecular Genetics, University of Maryland, College Park, MD 20742, USA.
| |
Collapse
|
17
|
Crajoinas RO, Polidoro JZ, Girardi ACC. The potential role of myosin motor proteins in mediating the subcellular distribution of NHE3 in the renal proximal tubule. Am J Physiol Renal Physiol 2019; 316:F986-F992. [PMID: 30864843 DOI: 10.1152/ajprenal.00577.2018] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Isoform 3 of the Na+/H+ exchanger (NHE3) is responsible for the majority of the reabsorption of NaCl, NaHCO3, and, consequently, water in the renal proximal tubule. As such, this transporter plays an essential role in acid-base balance and extracellular fluid volume homeostasis and determining systemic arterial blood pressure levels. NHE3 activity is modulated by a number of mechanisms, including the redistribution of the transporter between the body of the microvilli (where NHE3 is active) and the base of the microvilli (where NHE3 is less active). Although the physiological, pathophysiological, and pharmacological importance of the subcellular distribution of NHE3 has been well established, the exact mechanism whereby NHE3 is translocated along microvilli microdomains of the proximal tubule apical membrane is unknown. Nonmuscle myosin IIA and unconventional myosin VI move cargoes in anterograde and retrograde directions, respectively, and are known to redistribute along with NHE3 in the proximal tubule in response to a variety of natriuretic and antinatriuretic stimuli, including stimulation or inhibition of the renin-angiotensin system, high dietary Na+ intake, and high blood pressure. Therefore, this review aims to discuss the current evidence that suggests a potential role of myosin IIA and myosin VI in mediating the subcellular distribution of NHE3 along the kidney proximal tubule microvilli and their possible contribution in modifying NHE3-mediated Na+ reabsorption under both physiological and pathophysiological conditions.
Collapse
Affiliation(s)
- Renato O Crajoinas
- Heart Institute (InCor), University of São Paulo Medical School , São Paulo , Brazil
| | - Juliano Z Polidoro
- Heart Institute (InCor), University of São Paulo Medical School , São Paulo , Brazil
| | - Adriana C C Girardi
- Heart Institute (InCor), University of São Paulo Medical School , São Paulo , Brazil
| |
Collapse
|
18
|
Li XC, Zheng X, Chen X, Zhao C, Zhu D, Zhang J, Zhuo JL. Genetic and genomic evidence for an important role of the Na +/H + exchanger 3 in blood pressure regulation and angiotensin II-induced hypertension. Physiol Genomics 2019; 51:97-108. [PMID: 30849009 PMCID: PMC6485378 DOI: 10.1152/physiolgenomics.00122.2018] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023] Open
Abstract
The sodium (Na+)/hydrogen (H+) exchanger 3 (NHE3) and sodium-potassium adenosine triphosphatase (Na+/K+-ATPase) are two of the most important Na+ transporters in the proximal tubules of the kidney. On the apical membrane side, NHE3 primarily mediates the entry of Na+ into and the exit of H+ from the proximal tubules, directly and indirectly being responsible for reabsorbing ~50% of filtered Na+ in the proximal tubules of the kidney. On the basolateral membrane side, Na+/K+-ATPase serves as a powerful engine driving Na+ out of, while pumping K+ into the proximal tubules against their concentration gradients. While the roles of NHE3 and Na+/K+-ATPase in proximal tubular Na+ transport under in vitro conditions are well recognized, their respective contributions to the basal blood pressure regulation and angiotensin II (ANG II)-induced hypertension remain poorly understood. Recently, we have been fortunate to be able to use genetically modified mouse models with global, kidney- or proximal tubule-specific deletion of NHE3 to directly determine the cause and effect relationship between NHE3, basal blood pressure homeostasis, and ANG II-induced hypertension at the whole body, kidney and/or proximal tubule levels. The purpose of this article is to review the genetic and genomic evidence for an important role of NHE3 with a focus in the regulation of basal blood pressure and ANG II-induced hypertension, as we learned from studies using global, kidney- or proximal tubule-specific NHE3 knockout mice. We hypothesize that NHE3 in the proximal tubules is necessary for maintaining basal blood pressure homeostasis and the development of ANG II-induced hypertension.
Collapse
Affiliation(s)
- Xiao C Li
- Laboratory of Receptor and Signal Transduction, Department of Pharmacology and Toxicology; Division of Nephrology, Internal Medicine; Cardiovascular and Renal Research Center; The University of Mississippi Medical Center , Jackson, Mississippi
| | - Xiaowen Zheng
- Laboratory of Receptor and Signal Transduction, Department of Pharmacology and Toxicology; Division of Nephrology, Internal Medicine; Cardiovascular and Renal Research Center; The University of Mississippi Medical Center , Jackson, Mississippi
| | - Xu Chen
- Laboratory of Receptor and Signal Transduction, Department of Pharmacology and Toxicology; Division of Nephrology, Internal Medicine; Cardiovascular and Renal Research Center; The University of Mississippi Medical Center , Jackson, Mississippi
| | - Chunling Zhao
- Laboratory of Receptor and Signal Transduction, Department of Pharmacology and Toxicology; Division of Nephrology, Internal Medicine; Cardiovascular and Renal Research Center; The University of Mississippi Medical Center , Jackson, Mississippi
| | - Dongmin Zhu
- Laboratory of Receptor and Signal Transduction, Department of Pharmacology and Toxicology; Division of Nephrology, Internal Medicine; Cardiovascular and Renal Research Center; The University of Mississippi Medical Center , Jackson, Mississippi
| | - Jianfeng Zhang
- Laboratory of Receptor and Signal Transduction, Department of Pharmacology and Toxicology; Division of Nephrology, Internal Medicine; Cardiovascular and Renal Research Center; The University of Mississippi Medical Center , Jackson, Mississippi
| | - Jia L Zhuo
- Laboratory of Receptor and Signal Transduction, Department of Pharmacology and Toxicology; Division of Nephrology, Internal Medicine; Cardiovascular and Renal Research Center; The University of Mississippi Medical Center , Jackson, Mississippi
| |
Collapse
|
19
|
The Na/K-ATPase Signaling: From Specific Ligands to General Reactive Oxygen Species. Int J Mol Sci 2018; 19:ijms19092600. [PMID: 30200500 PMCID: PMC6163532 DOI: 10.3390/ijms19092600] [Citation(s) in RCA: 37] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2018] [Revised: 08/10/2018] [Accepted: 08/28/2018] [Indexed: 12/16/2022] Open
Abstract
The signaling function of the Na/K-ATPase has been established for 20 years and is widely accepted in the field, with many excellent reports and reviews not cited here. Even though there is debate about the underlying mechanism, the signaling function is unquestioned. This short review looks back at the evolution of Na/K-ATPase signaling, from stimulation by cardiotonic steroids (also known as digitalis-like substances) as specific ligands to stimulation by reactive oxygen species (ROS) in general. The interplay of cardiotonic steroids and ROS in Na/K-ATPase signaling forms a positive-feedback oxidant amplification loop that has been implicated in some pathophysiological conditions.
Collapse
|
20
|
Engevik AC, Goldenring JR. Trafficking Ion Transporters to the Apical Membrane of Polarized Intestinal Enterocytes. Cold Spring Harb Perspect Biol 2018; 10:cshperspect.a027979. [PMID: 28264818 DOI: 10.1101/cshperspect.a027979] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
Abstract
Epithelial cells lining the gastrointestinal tract require distinct apical and basolateral domains to function properly. Trafficking and insertion of enzymes and transporters into the apical brush border of intestinal epithelial cells is essential for effective digestion and absorption of nutrients. Specific critical ion transporters are delivered to the apical brush border to facilitate fluid and electrolyte uptake. Maintenance of these apical transporters requires both targeted delivery and regulated membrane recycling. Examination of altered apical trafficking in patients with Microvillus Inclusion disease caused by inactivating mutations in MYO5B has led to insights into the regulation of apical trafficking by elements of the apical recycling system. Modeling of MYO5B loss in cell culture and animal models has led to recognition of Rab11a and Rab8a as critical regulators of apical brush border function. All of these studies show the importance of apical membrane trafficking dynamics in maintenance of polarized epithelial cell function.
Collapse
Affiliation(s)
- Amy Christine Engevik
- Section of Surgical Sciences, Vanderbilt University Medical Center, Nashville, Tennessee 37232.,Epithelial Biology Center, Vanderbilt University Medical Center, Nashville, Tennessee 37232
| | - James R Goldenring
- Section of Surgical Sciences, Vanderbilt University Medical Center, Nashville, Tennessee 37232.,Epithelial Biology Center, Vanderbilt University Medical Center, Nashville, Tennessee 37232.,Department of Cell and Developmental Biology, Vanderbilt University, Nashville, Tennessee 37232.,Nashville VA Medical Center, Nashville, Tennessee 37232
| |
Collapse
|
21
|
Okamoto CT. Regulation of Transporters and Channels by Membrane-Trafficking Complexes in Epithelial Cells. Cold Spring Harb Perspect Biol 2017; 9:a027839. [PMID: 28246186 PMCID: PMC5666629 DOI: 10.1101/cshperspect.a027839] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
Abstract
The vectorial secretion and absorption of fluid and solutes by epithelial cells is dependent on the polarized expression of membrane solute transporters and channels at the apical and basolateral membranes. The establishment and maintenance of this polarized expression of transporters and channels are affected by divers protein-trafficking complexes. Moreover, regulation of the magnitude of transport is often under control of physiological stimuli, again through the interaction of transporters and channels with protein-trafficking complexes. This review highlights the value in utilizing transporters and channels as cargo to characterize core trafficking machinery by which epithelial cells establish and maintain their polarized expression, and how this machinery regulates fluid and solute transport in response to physiological stimuli.
Collapse
Affiliation(s)
- Curtis T Okamoto
- Department of Pharmacology and Pharmaceutical Sciences, School of Pharmacy, University of Southern California, Los Angeles, California 90089-9121
| |
Collapse
|
22
|
Hu MC, Bobulescu IA, Quiñones H, Gisler SM, Moe OW. Dopamine reduces cell surface Na +/H + exchanger-3 protein by decreasing NHE3 exocytosis and cell membrane recycling. Am J Physiol Renal Physiol 2017; 313:F1018-F1025. [PMID: 28768665 DOI: 10.1152/ajprenal.00251.2017] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2017] [Revised: 07/14/2017] [Accepted: 07/24/2017] [Indexed: 01/06/2023] Open
Abstract
The intrarenal autocrine-paracrine dopamine (DA) system mediates a significant fraction of the natriuresis in response to a salt load. DA inhibits a number of Na+ transporters to effect sodium excretion, including the proximal tubule Na+/H+ exchanger-3 (NHE3). DA represent a single hormone that regulates NHE3 at multiple levels, including translation, degradation, endocytosis, and protein phosphorylation. Because cell surface NHE3 protein is determined by the balance between exocytotic insertion and endocytotic retrieval, we examined whether DA acutely affects the rate of NHE3 exocytosis in a cell culture model. DA inhibited NHE3 exocytosis at a dose-dependent manner with a half maximal around 10-6 M. The DA effect on NHE3 exocytosis was blocked by inhibition of protein kinase A and by brefeldin A, which inhibits endoplasmic reticulum-to-Golgi transport. NHE3 directly interacts with the ε-subunit of coatomer protein based on yeast-two-hybrid and coimmunoprecipitation. Because NHE3 has been shown to be recycled back to the cell membrane after endocytosis, we measured NHE3 recycling using a biochemical reinsertion assay and showed that reinsertion of NHE3 back to the membrane is also inhibited by DA. In conclusion, among the many mechanisms by which DA reduces apical membrane NHE3 and induces proximal tubule natriuresis, one additional mechanism is inhibition of exocytotic insertion and reinsertion of NHE3 in the apical cell surface.
Collapse
Affiliation(s)
- Ming Chang Hu
- Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, Texas; .,Charles and Jane Pak Center of Mineral Metabolism and Clinical Research, University of Texas Southwestern Medical Center, Dallas, Texas
| | - I Alexandru Bobulescu
- Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, Texas.,Charles and Jane Pak Center of Mineral Metabolism and Clinical Research, University of Texas Southwestern Medical Center, Dallas, Texas
| | - Henry Quiñones
- Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, Texas
| | - Serge M Gisler
- Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, Texas
| | - Orson W Moe
- Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, Texas.,Department of Physiology, University of Texas Southwestern Medical Center, Dallas, Texas; and.,Charles and Jane Pak Center of Mineral Metabolism and Clinical Research, University of Texas Southwestern Medical Center, Dallas, Texas
| |
Collapse
|
23
|
Wang T, Weinbaum S, Weinstein AM. Regulation of glomerulotubular balance: flow-activated proximal tubule function. Pflugers Arch 2017; 469:643-654. [PMID: 28271233 DOI: 10.1007/s00424-017-1960-8] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2016] [Revised: 02/15/2017] [Accepted: 02/17/2017] [Indexed: 12/23/2022]
Abstract
The purpose of this review is to summarize our knowledge and understanding of the physiological importance and the mechanisms underlying flow-activated proximal tubule transport. Since the earliest micropuncture studies of mammalian proximal tubule, it has been recognized that tubular flow is an important regulator of sodium, potassium, and acid-base transport in the kidney. Increased fluid flow stimulates Na+ and HCO3- absorption in the proximal tubule via stimulation of Na/H-exchanger isoform 3 (NHE3) and H+-ATPase. In the proximal tubule, brush border microvilli are the major flow sensors, which experience changes in hydrodynamic drag and bending moment as luminal flow velocity changes and which transmit the force of altered flow to cytoskeletal structures within the cell. The signal to NHE3 depends upon the integrity of the actin cytoskeleton; the signal to the H+-ATPase depends upon microtubules. We have demonstrated that alterations in fluid drag impact tubule function by modulating ion transporter availability within the brush border membrane of the proximal tubule. Beyond that, there is evidence that transporter activity within the peritubular membrane is also modulated by luminal flow. Secondary messengers that regulate the flow-mediated tubule function have also been delineated. Dopamine blunts the responsiveness of proximal tubule transporters to changes in luminal flow velocity, while a DA1 antagonist increases flow sensitivity of solute reabsorption. IP3 receptor-mediated intracellular Ca2+ signaling is critical to transduction of microvillus drag. In this review, we summarize our findings of the regulatory mechanism of flow-mediated Na+ and HCO3- transport in the proximal tubule and review available information about flow sensing and regulatory mechanism of glomerulotubular balance.
Collapse
Affiliation(s)
- Tong Wang
- Department of Cellular and Molecular Physiology, Yale University, 333 Cedar Street, New Haven, CT, 06520, USA.
| | - Sheldon Weinbaum
- Department of Biomedical Engineering, City College of New York, CUNY, New York, NY, USA
| | - Alan M Weinstein
- Department of Physiology and Biophysics, Weill Medical College of Cornell University, New York, NY, USA
| |
Collapse
|
24
|
Liu J, Yan Y, Nie Y, Shapiro JI. Na/K-ATPase Signaling and Salt Sensitivity: The Role of Oxidative Stress. Antioxidants (Basel) 2017; 6:E18. [PMID: 28257114 PMCID: PMC5384181 DOI: 10.3390/antiox6010018] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2017] [Revised: 02/10/2017] [Accepted: 02/22/2017] [Indexed: 02/07/2023] Open
Abstract
Other than genetic regulation of salt sensitivity of blood pressure, many factors have been shown to regulate renal sodium handling which contributes to long-term blood pressure regulation and have been extensively reviewed. Here we present our progress on the Na/K-ATPase signaling mediated sodium reabsorption in renal proximal tubules, from cardiotonic steroids-mediated to reactive oxygen species (ROS)-mediated Na/K-ATPase signaling that contributes to experimental salt sensitivity.
Collapse
Affiliation(s)
- Jiang Liu
- Department of Biomedical Sciences, Joan C. Edwards School of Medicine, Marshall University, Huntington, WV 25755, USA.
| | - Yanling Yan
- Department of Biomedical Sciences, Joan C. Edwards School of Medicine, Marshall University, Huntington, WV 25755, USA.
| | - Ying Nie
- Department of Biomedical Sciences, Joan C. Edwards School of Medicine, Marshall University, Huntington, WV 25755, USA.
| | - Joseph I Shapiro
- Department of Medicine, Joan C. Edwards School of Medicine, Marshall University, Huntington, WV 25701, USA
| |
Collapse
|
25
|
Crajoinas RO, Polidoro JZ, Carneiro de Morais CPA, Castelo-Branco RC, Girardi ACC. Angiotensin II counteracts the effects of cAMP/PKA on NHE3 activity and phosphorylation in proximal tubule cells. Am J Physiol Cell Physiol 2016; 311:C768-C776. [PMID: 27510906 DOI: 10.1152/ajpcell.00191.2016] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2016] [Accepted: 07/29/2016] [Indexed: 12/16/2022]
Abstract
Binding of angiotensin II (ANG II) to the AT1 receptor (AT1R) in the proximal tubule stimulates Na+/H+ exchanger isoform 3 (NHE3) activity through multiple signaling pathways. However, the effects of ANG II/AT1R-induced inihibitory G protein (Gi) activation and subsequent decrease in cAMP accumulation on NHE3 regulation are not well established. We therefore tested the hypothesis that ANG II reduces cAMP/PKA-mediated phosphorylation of NHE3 on serine 552 and, in doing so, stimulates NHE3 activity. Under basal conditions, ANG II stimulated NHE3 activity but did not affect PKA-mediated NHE3 phosphorylation at serine 552 in opossum kidney (OKP) cells. However, in the presence of the cAMP-elevating agent forskolin (FSK), ANG II blocked FSK-induced NHE3 inhibition, reduced intracellular cAMP concentrations, lowered PKA activity, and prevented the FSK-mediated increase in NHE3 serine 552 phosphorylation. All effects of ANG II were blocked by pretreating OKP cells with the AT1R antagonist losartan, highlighting the contribution of the AT1R/Gi pathway in ANG II-mediated NHE3 upregulation under cAMP-elevating conditions. Accordingly, Gi inhibition by pertussis toxin treatment decreased NHE3 activity both in vitro and in vivo and, more importantly, prevented the stimulatory effect of ANG II on NHE3 activity in rat proximal tubules. Collectively, our results suggest that ANG II counteracts the effects of cAMP/PKA on NHE3 phosphorylation and inhibition by activating the AT1R/Gi pathway. Moreover, these findings support the notion that NHE3 dephosphorylation at serine 552 may represent a key event in the regulation of renal proximal tubule sodium handling by ANG II in the presence of natriuretic hormones that promote cAMP accumulation and transporter phosphorylation.
Collapse
Affiliation(s)
- Renato O Crajoinas
- Laboratory of Genetics and Molecular Cardiology, Heart Institute (InCor), University of São Paulo Medical School, São Paulo, São Paulo, Brazil; and
| | - Juliano Z Polidoro
- Laboratory of Genetics and Molecular Cardiology, Heart Institute (InCor), University of São Paulo Medical School, São Paulo, São Paulo, Brazil; and
| | - Carla P A Carneiro de Morais
- Laboratory of Genetics and Molecular Cardiology, Heart Institute (InCor), University of São Paulo Medical School, São Paulo, São Paulo, Brazil; and
| | - Regiane C Castelo-Branco
- Department of Physiology and Biophysics, Institute of Biomedical Sciences, São Paulo, São Paulo, Brazil
| | - Adriana C C Girardi
- Laboratory of Genetics and Molecular Cardiology, Heart Institute (InCor), University of São Paulo Medical School, São Paulo, São Paulo, Brazil; and
| |
Collapse
|
26
|
Hu MC, Shi M, Zhang J, Addo T, Cho HJ, Barker SL, Ravikumar P, Gillings N, Bian A, Sidhu SS, Kuro-o M, Moe OW. Renal Production, Uptake, and Handling of Circulating αKlotho. J Am Soc Nephrol 2016; 27:79-90. [PMID: 25977312 PMCID: PMC4696570 DOI: 10.1681/asn.2014101030] [Citation(s) in RCA: 204] [Impact Index Per Article: 22.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2014] [Accepted: 03/14/2015] [Indexed: 01/13/2023] Open
Abstract
αKlotho is a multifunctional protein highly expressed in the kidney. Soluble αKlotho is released through cleavage of the extracellular domain from membrane αKlotho by secretases to function as an endocrine/paracrine substance. The role of the kidney in circulating αKlotho production and handling is incompletely understood, however. Here, we found higher αKlotho concentration in suprarenal compared with infrarenal inferior vena cava in both rats and humans. In rats, serum αKlotho concentration dropped precipitously after bilateral nephrectomy or upon treatment with inhibitors of αKlotho extracellular domain shedding. Furthermore, the serum half-life of exogenous αKlotho in anephric rats was four- to five-fold longer than that in normal rats, and exogenously injected labeled recombinant αKlotho was detected in the kidney and in urine of rats. Both in vivo (micropuncture) and in vitro (proximal tubule cell line) studies showed that αKlotho traffics from the basal to the apical side of the proximal tubule via transcytosis. Thus, we conclude that the kidney has dual roles in αKlotho homeostasis, producing and releasing αKlotho into the circulation and clearing αKlotho from the blood into the urinary lumen.
Collapse
Affiliation(s)
- Ming Chang Hu
- Departments of Internal Medicine, Charles and Jane Pak Center of Mineral Metabolism and Clinical Research, University of Texas Southwestern Medical Center, Dallas, Texas;
| | - Mingjun Shi
- Charles and Jane Pak Center of Mineral Metabolism and Clinical Research, University of Texas Southwestern Medical Center, Dallas, Texas
| | | | | | - Han Ju Cho
- Charles and Jane Pak Center of Mineral Metabolism and Clinical Research, University of Texas Southwestern Medical Center, Dallas, Texas
| | - Sarah L Barker
- Banting and Best Department of Medical Research and Department of Molecular Genetics, The Donnelly Centre, University of Toronto, Toronto, Ontario, Canada; and
| | - Priya Ravikumar
- Departments of Internal Medicine, Charles and Jane Pak Center of Mineral Metabolism and Clinical Research, University of Texas Southwestern Medical Center, Dallas, Texas
| | - Nancy Gillings
- Charles and Jane Pak Center of Mineral Metabolism and Clinical Research, University of Texas Southwestern Medical Center, Dallas, Texas
| | - Ao Bian
- Charles and Jane Pak Center of Mineral Metabolism and Clinical Research, University of Texas Southwestern Medical Center, Dallas, Texas
| | - Sachdev S Sidhu
- Banting and Best Department of Medical Research and Department of Molecular Genetics, The Donnelly Centre, University of Toronto, Toronto, Ontario, Canada; and
| | - Makoto Kuro-o
- Charles and Jane Pak Center of Mineral Metabolism and Clinical Research, University of Texas Southwestern Medical Center, Dallas, Texas; Pathology, Center for Molecular Medicine, Jichi Medical University, Tochigi, Japan
| | - Orson W Moe
- Departments of Internal Medicine, Charles and Jane Pak Center of Mineral Metabolism and Clinical Research, University of Texas Southwestern Medical Center, Dallas, Texas; Physiology, and
| |
Collapse
|
27
|
Soares-da-Silva P, Cabral JM, Magalhães D, Fraga S, Magro F. Amine neurotransmitters, inflammation and epithelial sodium transport. Exp Physiol 2015; 101:459-64. [PMID: 26548358 DOI: 10.1113/ep085284] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2015] [Accepted: 11/02/2015] [Indexed: 12/13/2022]
Abstract
NEW FINDINGS What is the topic of this review? The present work reviews the roles of renal and intestinal dopamine and 5-HT in the maintenance of fluid and electrolyte homeostasis. The role of inflammatory agents at the intestinal level that affect fluid and electrolyte homeostasis is also addressed. What advances does it highlight? General mechanisms of epithelial cell ion transport in the gastrointestinal tract and kidney share considerable similarities, particularly with regard to basolateral Na(+) ,K(+-) ATPase as a driving force for the movement of numerous substrates across the cell membrane. The physiological importance of the renal actions of monoamines (dopamine, noradrenaline and 5-HT) mainly depends on the sources of the amines in the kidney and on their availability to activate the amine-specific receptors. Dopamine and 5-HT are also relatively abundant in the mucosal cell layer of the intestine, and recent evidence suggests their physiological relevance in regulating electrolyte transport. The gastrointestinal tract can be an important site for the loss of water and electrolytes, in the presence of intestinal inflammation. General mechanisms of epithelial cell ion transport in the gastrointestinal tract and kidney share considerable similarities with regard to basolateral Na(+) ,K(+) -ATPase as a driving force for the movement of numerous substrates across the cell membrane. The present work reviews the roles of renal and intestinal dopamine and 5-HT in the maintenance of fluid and electrolyte homeostasis. The role of inflammatory agents at the intestinal level that affect fluid and electrolyte homeostasis is also addressed.
Collapse
Affiliation(s)
- Patrício Soares-da-Silva
- Department of Pharmacology and Therapeutics, Faculty of Medicine, University of Porto, Porto, Portugal.,MedInUP - Center for Drug Discovery and Innovative Medicines, University of Porto, Porto, Portugal
| | - José Miguel Cabral
- Department of Pharmacology and Therapeutics, Faculty of Medicine, University of Porto, Porto, Portugal
| | - Diogo Magalhães
- Department of Pharmacology and Therapeutics, Faculty of Medicine, University of Porto, Porto, Portugal
| | - Sónia Fraga
- Department of Pharmacology and Therapeutics, Faculty of Medicine, University of Porto, Porto, Portugal.,MedInUP - Center for Drug Discovery and Innovative Medicines, University of Porto, Porto, Portugal
| | - Fernando Magro
- Department of Pharmacology and Therapeutics, Faculty of Medicine, University of Porto, Porto, Portugal.,MedInUP - Center for Drug Discovery and Innovative Medicines, University of Porto, Porto, Portugal
| |
Collapse
|
28
|
Singh V, Yang J, Cha B, Chen TE, Sarker R, Yin J, Avula LR, Tse M, Donowitz M. Sorting nexin 27 regulates basal and stimulated brush border trafficking of NHE3. Mol Biol Cell 2015; 26:2030-43. [PMID: 25851603 PMCID: PMC4472014 DOI: 10.1091/mbc.e14-12-1597] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2014] [Accepted: 04/01/2015] [Indexed: 12/21/2022] Open
Abstract
In polarized epithelial cells, SNX27 regulates PDZ domain–directed trafficking of NHE3 from endosomes to the plasma membrane and increases the stability of brush border NHE3. This establishes SNX27 as an important regulator of polarized sorting in epithelial cells. Sorting nexin 27 (SNX27) contains a PDZ domain that is phylogenetically related to the PDZ domains of the NHERF proteins. Studies on nonepithelial cells have shown that this protein is located in endosomes, where it regulates trafficking of cargo proteins in a PDZ domain–dependent manner. However, the role of SNX27 in trafficking of cargo proteins in epithelial cells has not been adequately explored. Here we show that SNX27 directly interacts with NHE3 (C-terminus) primarily through the SNX27 PDZ domain. A combination of knockdown and reconstitution experiments with wild type and a PDZ domain mutant (GYGF → GAGA) of SNX27 demonstrate that the PDZ domain of SNX27 is required to maintain basal NHE3 activity and surface expression of NHE3 in polarized epithelial cells. Biotinylation-based recycling and degradation studies in intestinal epithelial cells show that SNX27 is required for the exocytosis (not endocytosis) of NHE3 from early endosome to plasma membrane. SNX27 is also required to regulate the retention of NHE3 on the plasma membrane. The findings of the present study extend our understanding of PDZ-mediated recycling of cargo proteins from endosome to plasma membrane in epithelial cells.
Collapse
Affiliation(s)
- Varsha Singh
- Gastroenterology Division, Departments of Physiology and Medicine, Johns Hopkins University School of Medicine, Baltimore, MD 21205
| | - Jianbo Yang
- Gastroenterology Division, Departments of Physiology and Medicine, Johns Hopkins University School of Medicine, Baltimore, MD 21205
| | - Boyoung Cha
- Gastroenterology Division, Departments of Physiology and Medicine, Johns Hopkins University School of Medicine, Baltimore, MD 21205
| | - Tiane-e Chen
- Gastroenterology Division, Departments of Physiology and Medicine, Johns Hopkins University School of Medicine, Baltimore, MD 21205
| | - Rafiquel Sarker
- Gastroenterology Division, Departments of Physiology and Medicine, Johns Hopkins University School of Medicine, Baltimore, MD 21205
| | - Jianyi Yin
- Gastroenterology Division, Departments of Physiology and Medicine, Johns Hopkins University School of Medicine, Baltimore, MD 21205
| | - Leela Rani Avula
- Gastroenterology Division, Departments of Physiology and Medicine, Johns Hopkins University School of Medicine, Baltimore, MD 21205
| | - Ming Tse
- Gastroenterology Division, Departments of Physiology and Medicine, Johns Hopkins University School of Medicine, Baltimore, MD 21205
| | - Mark Donowitz
- Gastroenterology Division, Departments of Physiology and Medicine, Johns Hopkins University School of Medicine, Baltimore, MD 21205
| |
Collapse
|
29
|
Chen T, Kocinsky HS, Cha B, Murtazina R, Yang J, Tse CM, Singh V, Cole R, Aronson PS, de Jonge H, Sarker R, Donowitz M. Cyclic GMP kinase II (cGKII) inhibits NHE3 by altering its trafficking and phosphorylating NHE3 at three required sites: identification of a multifunctional phosphorylation site. J Biol Chem 2014; 290:1952-65. [PMID: 25480791 DOI: 10.1074/jbc.m114.590174] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023] Open
Abstract
The epithelial brush-border Na(+)/H(+) exchanger NHE3 is acutely inhibited by cGKII/cGMP, but how cGKII inhibits NHE3 is unknown. This study tested the hypothesis that cGMP inhibits NHE3 by phosphorylating it and altering its membrane trafficking. Studies were carried out in PS120/NHERF2 and in Caco-2/Bbe cells overexpressing HA-NHE3 and cGKII, and in mouse ileum. NHE3 activity was measured with 2',7'-bis(carboxyethyl)-S-(and 6)carboxyfluorescein acetoxy methylester/fluorometry. Surface NHE3 was determined by cell surface biotinylation. Identification of NHE3 phosphorylation sites was by iTRAQ/LC-MS/MS with TiO2 enrichment and immunoblotting with specific anti-phospho-NHE3 antibodies. cGMP/cGKII rapidly inhibited NHE3, which was associated with reduced surface NHE3. cGMP/cGKII increased NHE3 phosphorylation at three sites (rabbit Ser(554), Ser(607), and Ser(663), equivalent to mouse Ser(552), Ser(605), and Ser(659)), all of which had to be present at the same time for cGMP to inhibit NHE3. NHE3-Ser(663) phosphorylation was not necessary for cAMP inhibition of NHE3. Dexamethasone (4 h) stimulated wild type NHE3 activity and increased surface expression but failed to stimulate NHE3 activity or increase surface expression when NHE3 was mutated to either S663A or S663D. We conclude that 1) cGMP inhibition of NHE3 is associated with phosphorylation of NHE3 at Ser(554), Ser(607), and Ser(663), all of which are necessary for cGMP/cGKII to inhibit NHE3. 2) Dexamethasone stimulates NHE3 by phosphorylation of a single site, Ser(663). The requirement for three phosphorylation sites in NHE3 for cGKII inhibition, and for phosphorylation of one of these sites for dexamethasone stimulation of NHE3, is a unique example of regulation by phosphorylation.
Collapse
Affiliation(s)
- Tiane Chen
- From the Departments of Physiology and Medicine, Gastroenterology Division, and
| | | | - Boyoung Cha
- From the Departments of Physiology and Medicine, Gastroenterology Division, and
| | - Rakhilya Murtazina
- From the Departments of Physiology and Medicine, Gastroenterology Division, and
| | - Jianbo Yang
- From the Departments of Physiology and Medicine, Gastroenterology Division, and
| | - C Ming Tse
- From the Departments of Physiology and Medicine, Gastroenterology Division, and
| | - Varsha Singh
- From the Departments of Physiology and Medicine, Gastroenterology Division, and
| | - Robert Cole
- the Biological Chemistry Department, Johns Hopkins University School of Medicine, Baltimore, Maryland 21205
| | - Peter S Aronson
- Internal Medicine, Yale University School of Medicine, New Haven, Connecticut 06520, and
| | - Hugo de Jonge
- the GI Division, Erasmus Medical Center, 3015CN Rotterdam, Netherlands
| | - Rafiquel Sarker
- From the Departments of Physiology and Medicine, Gastroenterology Division, and
| | - Mark Donowitz
- From the Departments of Physiology and Medicine, Gastroenterology Division, and
| |
Collapse
|
30
|
No YR, He P, Yoo BK, Yun CC. Unique regulation of human Na+/H+ exchanger 3 (NHE3) by Nedd4-2 ligase that differs from non-primate NHE3s. J Biol Chem 2014; 289:18360-72. [PMID: 24831004 DOI: 10.1074/jbc.m113.541706] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
Na(+)/H(+) exchanger NHE3 expressed in the intestine and kidney plays a major role in NaCl and HCO3 (-) absorption that is closely linked to fluid absorption and blood pressure regulation. The Nedd4 family of E3 ubiquitin ligases interacts with a number of transporters and channels via PY motifs. A comparison of NHE3 sequences revealed the presence of PY motifs in NHE3s from human and several non-human primates but not in non-primate NHE3s. In this study we evaluated the differences between human and non-primate NHE3s in ubiquitination and interaction with Nedd4-2. We found that Nedd4-2 ubiquitinated human NHE3 (hNHE3) and altered its expression and activity. Surprisingly, rat NHE3 co-immunoprecipitated Nedd4-2, but its expression and activity were not altered by silencing of Nedd4-2. Ubiquitination by Nedd4-2 rendered hNHE3 to undergo internalization at a significantly greater rate than non-primate NHE3s without altering protein stability. Insertion of a PY motif in rabbit NHE3 recapitulated the interaction with Nedd4-2 and enhanced internalization. Thus, we propose a new model where disruption of Nedd4-2 interaction elevates hNHE3 expression and activity.
Collapse
Affiliation(s)
- Yi Ran No
- From the Division of Digestive Diseases, Department of Medicine and
| | - Peijian He
- From the Division of Digestive Diseases, Department of Medicine and
| | - Byong Kwon Yoo
- From the Division of Digestive Diseases, Department of Medicine and
| | - C Chris Yun
- From the Division of Digestive Diseases, Department of Medicine and Winship Cancer Institute, Emory University, Atlanta, Georgia 30322
| |
Collapse
|
31
|
Zachos NC, Alamelumangpuram B, Lee LJ, Wang P, Kovbasnjuk O. Carbachol-mediated endocytosis of NHE3 involves a clathrin-independent mechanism requiring lipid rafts and Cdc42. Cell Physiol Biochem 2014; 33:869-81. [PMID: 24713550 PMCID: PMC4052452 DOI: 10.1159/000358659] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/10/2014] [Indexed: 12/17/2022] Open
Abstract
BACKGROUND In intestinal epithelial cells, acute regulation of the brush border Na(+)/H(+) exchanger, NHE3, usually occurs by changes in endocytosis and/or exocytosis. Constitutive NHE3 endocytosis involves clathrin. Carbachol (CCH), which elevates intracellular Ca(2+) ([Ca(2+)]i), decreases NHE3 activity and stimulates endocytosis; however, the mechanism involved in calcium-mediated endocytosis of NHE3 is unclear. A pool of NHE3 resides in lipid rafts, which contributes to basal, but not cAMP-mediated, NHE3 trafficking, suggesting that an alternative mechanism exists for NHE3 endocytosis. Cdc42 was demonstrated to play an integral role in some cases of cholesterol-sensitive, clathrin-independent endocytosis. Therefore, the current study was designed to test the hypotheses that (1) clathrin-mediated endocytosis (CME) is involved in constitutive, but not CCH-mediated, endocytosis of NHE3, and (2) CCH-mediated endocytosis of NHE3 occurs through a lipid raft, activated Cdc42-dependent pathway that does not involve clathrin. METHODS The role of Cdc42 and lipid rafts on NHE3 activity and endocytosis were investigated in polarized Caco-2/BBe cells using pharmacological and shRNA knockdown approaches. RESULTS Basal NHE3 activity was increased in the presence of CME blockers (chlorpromazine; K(+) depletion) supporting previous reports that constitutive NHE3 endocytosis is clathrin dependent. In contrast, CCH-inhibition of NHE3 activity was abolished in Caco-2/BBe cells treated with MβCD (to disrupt lipid rafts) as well as in Cdc42 knockdown cells but was unaffected by CME blockers. CONCLUSION CCH-mediated inhibition of NHE3 activity is not dependent on clathrin and involves lipid rafts and requires Cdc42.
Collapse
Affiliation(s)
- Nicholas C Zachos
- Department of Medicine/Gastroenterology and Hepatology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | | | | | | | | |
Collapse
|
32
|
Babich V, Vadnagara K, Di Sole F. The biophysical and molecular basis of intracellular pH sensing by Na+/H+ exchanger-3. FASEB J 2013; 27:4646-58. [PMID: 23934281 DOI: 10.1096/fj.12-225466] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
Epithelial Na(+)/H(+) exchanger-3 (NHE3) transport is fundamental for renal and intestinal sodium reabsorption. Cytoplasmic protons are thought to serve as allosteric modifiers of the exchanger and to trigger its transport through protein conformational change. This effect presupposes an intracellular pH (pHi) dependence of NHE3 activity, although the biophysical and molecular basis of NHE3 pHi sensitivity have not been defined. NHE3, when complexed with the calcineurin homologous protein-1 (CHP1), had a shift in pHi sensitivity (0.4 units) toward the acidic side in comparison with NHE3 alone, as measured by oscillating pH electrodes combined with whole-cell patch clamping. Indeed, CHP1 interaction with NHE3 inhibited NHE3 transport in a pHi -dependent manner. CHP1 binding to NHE3 also affected its acute regulation. Intracellular perfusion of peptide from the CHP1 binding region (or pHi modification to reduce the CHP1 amount bound to NHE3) was permissive and cooperative for dopamine inhibition of NHE3 but reversed that of adenosine. Thus, CHP1 interaction with NHE3 apparently establishes the exchanger set point for pHi, and modification in this set point is effective in the hormonal stimuli-mediated regulation of NHE3. CHP1 may serve as a regulatory cofactor for NHE3 conformational change, dependent on intracellular protonation.
Collapse
Affiliation(s)
- Victor Babich
- 1Department of Medicine, University of Maryland School of Medicine, 20 Penn Street, HSFII, Suite S005, Baltimore, MD 21201, USA.
| | | | | |
Collapse
|
33
|
Abstract
The kidney plays a fundamental role in maintaining body salt and fluid balance and blood pressure homeostasis through the actions of its proximal and distal tubular segments of nephrons. However, proximal tubules are well recognized to exert a more prominent role than distal counterparts. Proximal tubules are responsible for reabsorbing approximately 65% of filtered load and most, if not all, of filtered amino acids, glucose, solutes, and low molecular weight proteins. Proximal tubules also play a key role in regulating acid-base balance by reabsorbing approximately 80% of filtered bicarbonate. The purpose of this review article is to provide a comprehensive overview of new insights and perspectives into current understanding of proximal tubules of nephrons, with an emphasis on the ultrastructure, molecular biology, cellular and integrative physiology, and the underlying signaling transduction mechanisms. The review is divided into three closely related sections. The first section focuses on the classification of nephrons and recent perspectives on the potential role of nephron numbers in human health and diseases. The second section reviews recent research on the structural and biochemical basis of proximal tubular function. The final section provides a comprehensive overview of new insights and perspectives in the physiological regulation of proximal tubular transport by vasoactive hormones. In the latter section, attention is particularly paid to new insights and perspectives learnt from recent cloning of transporters, development of transgenic animals with knockout or knockin of a particular gene of interest, and mapping of signaling pathways using microarrays and/or physiological proteomic approaches.
Collapse
Affiliation(s)
- Jia L Zhuo
- Laboratory of Receptor and Signal Transduction, Department of Pharmacology and Toxicology, University of Mississippi Medical Center, Jackson, Mississippi, USA.
| | | |
Collapse
|
34
|
Zachos NC, Lee LJ, Kovbasnjuk O, Li X, Donowitz M. PLC-γ directly binds activated c-Src, which is necessary for carbachol-mediated inhibition of NHE3 activity in Caco-2/BBe cells. Am J Physiol Cell Physiol 2013; 305:C266-75. [PMID: 23703528 DOI: 10.1152/ajpcell.00277.2012] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
Elevated levels of intracellular Ca(2+) ([Ca(2+)]i) inhibit Na(+)/H(+) exchanger 3 (NHE3) activity in the intact intestine. We previously demonstrated that PLC-γ directly binds NHE3, an interaction that is necessary for [Ca(2+)]i inhibition of NHE3 activity, and that PLC-γ Src homology 2 (SH2) domains may scaffold Ca(2+) signaling proteins necessary for regulation of NHE3 activity. [Ca(2+)]i regulation of NHE3 activity is also c-Src dependent; however, the mechanism by which c-Src is involved is undetermined. We hypothesized that the SH2 domains of PLC-γ might link c-Src to NHE3-containing complexes to mediate [Ca(2+)]i inhibition of NHE3 activity. In Caco-2/BBe cells, carbachol (CCh) decreased NHE3 activity by ∼40%, an effect abolished with the c-Src inhibitor PP2. CCh treatment increased the amount of active c-Src as early as 1 min through increased Y(416) phosphorylation. Coimmunoprecipitation demonstrated that c-Src associated with PLC-γ, but not NHE3, under basal conditions, an interaction that increased rapidly after CCh treatment and occurred before the dissociation of PLC-γ and NHE3 that occurred 10 min after CCh treatment. Finally, direct binding to c-Src only occurred through the PLC-γ SH2 domains, an interaction that was prevented by blocking the PLC-γ SH2 domain. This study demonstrated that c-Src 1) activity is necessary for [Ca(2+)]i inhibition of NHE3 activity, 2) activation occurs rapidly (∼1 min) after CCh treatment, 3) directly binds PLC-γ SH2 domains and associates dynamically with PLC-γ under elevated [Ca(2+)]i conditions, and 4) does not directly bind NHE3. Under elevated [Ca(2+)]i conditions, PLC-γ scaffolds c-Src into NHE3-containing multiprotein complexes before dissociation of PLC-γ from NHE3 and subsequent endocytosis of NHE3.
Collapse
Affiliation(s)
- Nicholas C Zachos
- Department of Medicine/Gastroenterology and Hepatology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA.
| | | | | | | | | |
Collapse
|
35
|
Hu MC, Di Sole F, Zhang J, McLeroy P, Moe OW. Chronic regulation of the renal Na(+)/H(+) exchanger NHE3 by dopamine: translational and posttranslational mechanisms. Am J Physiol Renal Physiol 2013; 304:F1169-80. [PMID: 23427139 DOI: 10.1152/ajprenal.00630.2012] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023] Open
Abstract
The intrarenal autocrine/paracrine dopamine (DA) system contributes to natriuresis in response to both acute and chronic Na(+) loads. While the acute DA effect is well described, how DA induces natriuresis chronically is not known. We used an animal and a cell culture model to study the chronic effect of DA on a principal renal Na(+) transporter, Na(+)/H(+) exchanger-3 (NHE3). Intraperitoneal injection of Gludopa in rats for 2 days elevated DA excretion and decreased total renal cortical and apical brush-border NHE3 antigen. Chronic treatment of an opossum renal proximal cell line with DA decreased NHE3 activity, cell surface and total cellular NHE3 antigen, but not NHE3 transcript. The decrease in NHE3 antigen was dose and time dependent with maximal inhibition at 16-24 h and half maximal effect at 3 × 10(-7) M. This is in contradistinction to the acute effect of DA on NHE3 (half maximal at 2 × 10(-6) M), which was not associated with changes in total cellular NHE3 protein. The DA-induced decrease in total NHE3 protein was associated with decrease in NHE3 translation and mediated by cis-sequences in the NHE3 5'-untranslated region. DA also decreased cell surface and total cellular NHE3 protein half-life. The DA-induced decrease in total cellular NHE3 was partially blocked by proteasome inhibition but not by lysosome inhibition, and DA increased ubiquitylation of total and surface NHE3. In summary, chronic DA inhibits NHE3 with mechanisms distinct from its acute action and involves decreased NHE3 translation and increased NHE3 degradation, which are novel mechanisms for NHE3 regulation.
Collapse
Affiliation(s)
- Ming Chang Hu
- Dept. of Internal Medicine, Univ. of Texas Southwestern Medical Center, 5323 Harry Hines Blvd., Dallas, TX 75390-8885, USA
| | | | | | | | | |
Collapse
|
36
|
Du Z, Yan Q, Wan L, Weinbaum S, Weinstein AM, Wang T. Regulation of glomerulotubular balance. I. Impact of dopamine on flow-dependent transport. Am J Physiol Renal Physiol 2012; 303:F386-95. [PMID: 22552936 DOI: 10.1152/ajprenal.00531.2011] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023] Open
Abstract
In response to volume expansion, locally generated dopamine decreases proximal tubule reabsorption by reducing both Na/H-exchanger 3 (NHE3) and Na-K-ATPase activity. We have previously demonstrated that mouse proximal tubules in vitro respond to changes in luminal flow with proportional changes in Na(+) and HCO(3)(-) reabsorption and have suggested that this observation underlies glomerulotubular balance. In the present work, we investigate the impact of dopamine on the sensitivity of reabsorptive fluxes to changes in luminal flow. Mouse proximal tubules were microperfused in vitro at low and high flow rates, and volume and HCO(3)(-) reabsorption (J(v) and J(HCO3)) were measured, while Na(+) and Cl(-) reabsorption (J(Na) and J(Cl)) were estimated. Raising luminal flow increased J(v), J(Na), and J(HCO3) but did not change J(Cl). Luminal dopamine did not change J(v), J(Na), and J(HCO3) at low flow rates but completely abolished the increments of Na(+) absorption by flow and partially inhibited the flow-stimulated HCO(3)(-) absorption. The remaining flow-stimulated HCO(3)(-) absorption was completely abolished by bafilomycin. The DA1 receptor blocker SCH23390 and the PKA inhibitor H89 blocked the effect of exogenous dopamine and produced a two to threefold increase in the sensitivity of proximal Na(+) reabsorption to luminal flow rate. Under the variety of perfusion conditions, changes in cell volume were small and did not always parallel changes in Na(+) transport. We conclude that 1) dopamine inhibits flow-stimulated NHE3 activity by activation of the DA1 receptor via a PKA-mediated mechanism; 2) dopamine has no effect on flow-stimulated H-ATPase activity; 3) there is no evidence of flow stimulation of Cl(-) reabsorption; and 4) the impact of dopamine is a coordinated modulation of both luminal and peritubular Na(+) transporters.
Collapse
Affiliation(s)
- Zhaopeng Du
- Department of Cellular and Molecular Physiology, Yale University, New Haven, CT 06520-8026, USA
| | | | | | | | | | | |
Collapse
|
37
|
Huang H, Ren H, Chen C, Wang X, Yang J, Han Y, He D, Zhou L, Asico LD, Jose PA, Zeng C. D3 dopamine receptor regulation of D5 receptor expression and function in renal proximal tubule cells. Hypertens Res 2012; 35:639-47. [PMID: 22297482 DOI: 10.1038/hr.2012.11] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Dopamine receptor, via D(1)-like and D(2)-like receptors, increases sodium excretion in kidney. We have reported positive interactions between D(3) and D(1) receptors in renal proximal tubule (RPT) cells. These reports, however do not preclude that there may be also interaction between D(3) and D(5) receptors, because of the lack of selective D(1) and D(5) receptor agonists or antagonists. We hypothesize that D(3) receptors can regulate D(5) receptors, and that D(3) receptor regulation of D(5) receptors in RPTs is impaired in spontaneously hypertensive rats (SHRs). It showed that a D(3) receptor agonist, PD128907, by the activation of protein kinase C activity, increased the expression of D(5) receptors in a concentration- and time-dependent manner in RPT cells from Wistar-Kyoto (WKY) rats. The stimulatory effect of the D(3) receptor on D(5) receptor expression was impaired in RPT cells from SHRs. The effect of D(3) receptor on D(5) receptor is functionally relevant; stimulation of D(5) receptor decreases Na(+)-K(+) adenosine triphosphatase (ATPase) activity in WKY cells. Pretreatment with D(3) receptor agonist for 24 h enhances the D(5) receptor expression and D(5) receptor-mediated inhibitory effect on Na(+)-K(+) ATPase activity in WKY cells, but decreases them in SHR cells. The effect of D(3) receptor on D(5) receptor expression and function was also confirmed in the D(5) receptor-transfected HEK293 cells. It indicates that activation of D(3) receptor increases D(5) receptor expression and function. Altered regulation of D(3) receptor on D(5) receptors may have a role in the pathogenesis of hypertension.
Collapse
Affiliation(s)
- Hefei Huang
- Department of Cardiology, Daping Hospital, The Third Military Medical University, Chongqing, PR China
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
38
|
Carmosino M, Procino G, Tamma G, Mannucci R, Svelto M, Valenti G. Trafficking and phosphorylation dynamics of AQP4 in histamine-treated human gastric cells. Biol Cell 2012; 99:25-36. [PMID: 16895520 DOI: 10.1042/bc20060068] [Citation(s) in RCA: 45] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
BACKGROUND INFORMATION AQP4 (aquaporin 4) internalization and a concomitant decrease in the osmotic water permeability coefficient (Pf) after histamine exposure has been reported in AQP4-transfected gastric HGT1 cells. RESULTS In the present study we report that AQP4 internalization is followed by an increase in AQP4 phosphorylation. Histamine treatment for 30 min resulted in an approx. 10-fold increase in AQP4 phosphorylation that was inhibited by 1 microM H89, a specific PKA (protein kinase A) inhibitor, but not by PKC (protein kinase C) and CK2 inhibitors. Moreover, measurement of PKA activity after 30 min of histamine treatment showed that PKA activity was approx. 3-fold higher compared with basal conditions. AQP4 phosphorylation was prevented in cells treated with histamine for 30 min after pre-incubation with PAO (phenylarsine oxide), an inhibitor of protein endocytosis. Using an endo-exocytosis assay we showed that, after histamine washed out, internalized AQP4 recycled back to the cell surface, even in cells in which de novo protein synthesis was inhibited by cycloheximide. CONCLUSIONS Phosphorylation experiments, combined with immunolocalization studies, indicated that AQP4 phosphorylation is mediated by PKA and occurs subsequently to its internalization in late endosomes. We suggest that phosphorylation might be a mechanism involved in retaining AQP4 in a vesicle-recycling compartment.
Collapse
Affiliation(s)
- Monica Carmosino
- Department of General and Environmental Physiology, University of Bari, Via Amendola 165/A, 70126 Bari, Italy
| | | | | | | | | | | |
Collapse
|
39
|
Abstract
Dopamine is an important regulator of systemic blood pressure via multiple mechanisms. It affects fluid and electrolyte balance by its actions on renal hemodynamics and epithelial ion and water transport and by regulation of hormones and humoral agents. The kidney synthesizes dopamine from circulating or filtered L-DOPA independently from innervation. The major determinants of the renal tubular synthesis/release of dopamine are probably sodium intake and intracellular sodium. Dopamine exerts its actions via two families of cell surface receptors, D1-like receptors comprising D1R and D5R, and D2-like receptors comprising D2R, D3R, and D4R, and by interactions with other G protein-coupled receptors. D1-like receptors are linked to vasodilation, while the effect of D2-like receptors on the vasculature is variable and probably dependent upon the state of nerve activity. Dopamine secreted into the tubular lumen acts mainly via D1-like receptors in an autocrine/paracrine manner to regulate ion transport in the proximal and distal nephron. These effects are mediated mainly by tubular mechanisms and augmented by hemodynamic mechanisms. The natriuretic effect of D1-like receptors is caused by inhibition of ion transport in the apical and basolateral membranes. D2-like receptors participate in the inhibition of ion transport during conditions of euvolemia and moderate volume expansion. Dopamine also controls ion transport and blood pressure by regulating the production of reactive oxygen species and the inflammatory response. Essential hypertension is associated with abnormalities in dopamine production, receptor number, and/or posttranslational modification.
Collapse
Affiliation(s)
- Ines Armando
- Children’s National Medical Center—Center for Molecular Physiology Research, Washington, District of Columbia
| | - Van Anthony M. Villar
- Children’s National Medical Center—Center for Molecular Physiology Research, Washington, District of Columbia
| | - Pedro A. Jose
- Children’s National Medical Center—Center for Molecular Physiology Research, Washington, District of Columbia
| |
Collapse
|
40
|
Role of renalase in the regulation of blood pressure and the renal dopamine system. Curr Opin Nephrol Hypertens 2011; 20:31-6. [PMID: 21099685 DOI: 10.1097/mnh.0b013e3283412721] [Citation(s) in RCA: 56] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
PURPOSE OF REVIEW Renalase is a secreted amine oxidase that is synthesized in the kidney, and that metabolizes circulating catecholamines. Tissue and plasma renalase levels are decreased in models of chronic kidney disease. Recent data indicate that renalase deficiency is associated with increased blood pressure and elevated circulating catecholamines. The mechanisms of hypertension in renalase deficiency and the possibility that renalase regulates the renal dopamine system are discussed. RECENT FINDINGS Characterization of the renalase knockout mouse model revealed that renalase deficiency increases SBP and DBP. Renal and cardiac functions are unaffected, but there is evidence of sympathetic activation, with elevation of plasma and urine catecholamines. Renalase is continually excreted in urine, and is enzymatically active and could modulate catecholamines levels in tubular fluid. Renalase expression is modulated by salt intake, and recombinant renalase has a potent and prolonged hypotensive effect on blood pressure in Dahl salt-sensitive rats and rats with chronic kidney disease. Plasma renalase levels are inversely associated with SBP in patients with resistant hypertension. A functional mutation in renalase (Glu37Asp) associated with essential hypertension also predicts more severe cardiac hypertrophy, dysfunction, and ischemia in individuals with stable coronary artery disease, comparable blood pressure and normal renal function. SUMMARY Urinary renalase metabolizes urinary catecholamines, and perhaps regulates dopamine concentration in luminal fluid, and modulate proximal tubular sodium transport. Renalase deficiency is associated with increased sympathetic tone and resistant hypertension. Recombinant renalase is a potent antihypertensive agent in Dahl salt-sensitive rats and in rats with chronic kidney disease.
Collapse
|
41
|
Lin Z, Jin S, Duan X, Wang T, Martini S, Hulamm P, Cha B, Hubbard A, Donowitz M, Guggino SE. Chloride channel (Clc)-5 is necessary for exocytic trafficking of Na+/H+ exchanger 3 (NHE3). J Biol Chem 2011; 286:22833-45. [PMID: 21561868 DOI: 10.1074/jbc.m111.224998] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
ClC-5, a chloride/proton exchanger, is predominantly expressed and localized in subapical endosomes of the renal proximal tubule. Mutations of the CLCN5 gene cause Dent disease. The symptoms of Dent disease are replicated in Clcn5 knock-out mice. Absence of ClC-5 in mice is associated with reduced surface expression of NHE3 in proximal tubules. The molecular basis for this change is not fully understood. In this study, we investigated the mechanisms by which ClC-5 regulates trafficking of NHE3. Whether ClC-5-dependent endocytosis, exocytosis, or both contributed to the altered distribution of NHE3 was examined. First, NHE3 activity in proximal tubules of wild type (WT) and Clcn5 KO mice was determined by two-photon microscopy. Basal and dexamethasone-stimulated NHE3 activity of Clcn5 KO mice was decreased compared with that seen in WT mice, whereas the degree of inhibition of NHE3 activity by increasing cellular concentration of cAMP (forskolin) or Ca(2+) (A23187) was not different in WT and Clcn5 KO mice. Second, NHE3-dependent absorption of HCO(3)(-), measured by single tubule perfusion, was reduced in proximal tubules of Clcn5 KO mice. Third, by cell surface biotinylation, trafficking of NHE3 was examined in short hairpin RNA (shRNA) plasmid-transfected opossum kidney cells. Surface NHE3 was reduced in opossum kidney cells with reduced expression of ClC-5, whereas the total protein level of NHE3 did not change. Parathyroid hormone decreased NHE3 surface expression, but the extent of decrease and the rate of endocytosis observed in both scrambled and ClC-5 knockdown cells were not significantly different. However, the rates of basal and dexamethasone-stimulated exocytosis of NHE3 were attenuated in ClC-5 knockdown cells. These results show that ClC-5 plays an essential role in exocytosis of NHE3.
Collapse
Affiliation(s)
- Zhihong Lin
- Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland 21205, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
42
|
Zaarour N, Defontaine N, Demaretz S, Azroyan A, Cheval L, Laghmani K. Secretory carrier membrane protein 2 regulates exocytic insertion of NKCC2 into the cell membrane. J Biol Chem 2011; 286:9489-9502. [PMID: 21205824 PMCID: PMC3059028 DOI: 10.1074/jbc.m110.166546] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2010] [Revised: 12/23/2010] [Indexed: 12/20/2022] Open
Abstract
The renal-specific Na-K-2Cl co-transporter, NKCC2, plays a pivotal role in regulating body salt levels and blood pressure. NKCC2 mutations lead to type I Bartter syndrome, a life-threatening kidney disease. Regulation of NKCC2 trafficking behavior serves as a major mechanism in controlling NKCC2 activity across the plasma membrane. However, the identities of the protein partners involved in cell surface targeting of NKCC2 are largely unknown. To gain insight into these processes, we used a yeast two-hybrid system to screen a kidney cDNA library for proteins that interact with the NKCC2 C terminus. One binding partner we identified was SCAMP2 (secretory carrier membrane protein 2). Microscopic confocal imaging and co-immunoprecipitation assays confirmed NKCC2-SCAMP2 interaction in renal cells. SCAMP2 associated also with the structurally related co-transporter NCC, suggesting that the interaction with SCAMP2 is a common feature of sodium-dependent chloride co-transporters. Heterologous expression of SCAMP2 specifically decreased cell surface abundance as well as transport activity of NKCC2 across the plasma membrane. Co-immunolocalization experiments revealed that intracellularly retained NKCC2 co-localizes with SCAMP2 in recycling endosomes. The rate of NKCC2 endocytic retrieval, assessed by the sodium 2-mercaptoethane sulfonate cleavage assay, was not affected by SCAMP2. The surface-biotinylatable fraction of newly inserted NKCC2 in the plasma membrane was reduced by SCAMP2, demonstrating that SCAMP2-induced decrease in surface NKCC2 is due to decreased exocytotic trafficking. Finally, a single amino acid mutation, cysteine 201 to alanine, within the conserved cytoplasmic E peptide of SCAMP2, which is believed to regulate exocytosis, abolished SCAMP2-mediated down-regulation of the co-transporter. Taken together, these data are consistent with a model whereby SCAMP2 regulates NKCC2 transit through recycling endosomes and limits the cell surface targeting of the co-transporter by interfering with its exocytotic trafficking.
Collapse
Affiliation(s)
- Nancy Zaarour
- From INSERM, Centre de Recherche des Cordeliers, UMRS 872, CNRS, ERL7226, 75006 Paris, France
- Université Pierre et Marie Curie, 75006 Paris, France, and
- the Faculté de Médecine, Université Paris-Descartes, 75005 Paris, France
| | - Nadia Defontaine
- From INSERM, Centre de Recherche des Cordeliers, UMRS 872, CNRS, ERL7226, 75006 Paris, France
- Université Pierre et Marie Curie, 75006 Paris, France, and
- the Faculté de Médecine, Université Paris-Descartes, 75005 Paris, France
| | - Sylvie Demaretz
- From INSERM, Centre de Recherche des Cordeliers, UMRS 872, CNRS, ERL7226, 75006 Paris, France
- Université Pierre et Marie Curie, 75006 Paris, France, and
- the Faculté de Médecine, Université Paris-Descartes, 75005 Paris, France
| | - Anie Azroyan
- From INSERM, Centre de Recherche des Cordeliers, UMRS 872, CNRS, ERL7226, 75006 Paris, France
- Université Pierre et Marie Curie, 75006 Paris, France, and
- the Faculté de Médecine, Université Paris-Descartes, 75005 Paris, France
| | - Lydie Cheval
- From INSERM, Centre de Recherche des Cordeliers, UMRS 872, CNRS, ERL7226, 75006 Paris, France
- Université Pierre et Marie Curie, 75006 Paris, France, and
- the Faculté de Médecine, Université Paris-Descartes, 75005 Paris, France
| | - Kamel Laghmani
- From INSERM, Centre de Recherche des Cordeliers, UMRS 872, CNRS, ERL7226, 75006 Paris, France
- Université Pierre et Marie Curie, 75006 Paris, France, and
- the Faculté de Médecine, Université Paris-Descartes, 75005 Paris, France
| |
Collapse
|
43
|
Welling PA, Weisz OA. Sorting it out in endosomes: an emerging concept in renal epithelial cell transport regulation. Physiology (Bethesda) 2011; 25:280-92. [PMID: 20940433 DOI: 10.1152/physiol.00022.2010] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Ion and water transport by the kidney is continually adjusted in response to physiological cues. Selective endocytosis and endosomal trafficking of ion transporters are increasingly appreciated as mechanisms to acutely modulate renal function. Here, we discuss emerging paradigms in this new area of investigation.
Collapse
Affiliation(s)
- Paul A Welling
- Department of Physiology, University of Maryland School of Medicine, Baltimore, MD, USA
| | | |
Collapse
|
44
|
Pao AC, Bhargava A, Di Sole F, Quigley R, Shao X, Wang J, Thomas S, Zhang J, Shi M, Funder JW, Moe OW, Pearce D. Expression and role of serum and glucocorticoid-regulated kinase 2 in the regulation of Na+/H+ exchanger 3 in the mammalian kidney. Am J Physiol Renal Physiol 2010; 299:F1496-506. [PMID: 20926631 PMCID: PMC3006302 DOI: 10.1152/ajprenal.00075.2010] [Citation(s) in RCA: 37] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2010] [Accepted: 09/14/2010] [Indexed: 01/30/2023] Open
Abstract
Serum and glucocorticoid-regulated kinase 2 (sgk2) is 80% identical to the kinase domain of sgk1, an important mediator of mineralocorticoid-regulated sodium (Na(+)) transport in the distal nephron of the kidney. The expression pattern and role in renal function of sgk2 are virtually uncharacterized. In situ hybridization and immunohistochemistry of rodent kidney coupled with real-time RT-PCR of microdissected rat kidney tubules showed robust sgk2 expression in the proximal straight tubule and thick ascending limb of the loop of Henle. Sgk2 expression was minimal in distal tubule cells with aquaporin-2 immunostaining but significant in proximal tubule cells with Na(+)/H(+) exchanger 3 (NHE3) immunostaining. To ascertain whether mineralocorticoids regulate expression of sgk2 in a manner similar to sgk1, we examined sgk2 mRNA expression in the kidneys of adrenalectomized rats treated with physiological doses of aldosterone together with the glucocorticoid receptor antagonist RU486. Northern blot analysis and in situ hybridization showed that, unlike sgk1, sgk2 expression in the kidney was not altered by aldosterone treatment. Based on the observation that sgk2 is expressed in proximal tubule cells that also express NHE3, we asked whether sgk2 regulates NHE3 activity. We heterologously expressed sgk2 in opossum kidney (OKP) cells and measured Na(+)/H(+) exchange activity by Na(+)-dependent cell pH recovery. Constitutively active sgk2, but not sgk1, stimulated Na(+)/H(+) exchange activity by >30%. Moreover, the sgk2-mediated increase in Na(+)/H(+) exchange activity correlated with an increase in cell surface expression of NHE3. Together, these results suggest that the pattern of expression, regulation, and role of sgk2 within the mammalian kidney are distinct from sgk1 and that sgk2 may play a previously unrecognized role in the control of transtubular Na(+) transport through NHE3 in the proximal tubule.
Collapse
Affiliation(s)
- Alan C Pao
- Div. of Nephrology, Dept. of Medicine, Stanford Univ., 780 Welch Rd., Suite 106, Palo Alto, CA 94304, USA.
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
45
|
Shear stress-induced changes of membrane transporter localization and expression in mouse proximal tubule cells. Proc Natl Acad Sci U S A 2010; 107:21860-5. [PMID: 21106755 DOI: 10.1073/pnas.1015751107] [Citation(s) in RCA: 74] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Our previous studies of microperfused single proximal tubule showed that flow-dependent Na(+) and HCO(3)(-) reabsorption is due to a modulation of both NHE3 and vacuolar H(+)-ATPase (V-ATPase) activity. An intact actin cytoskeleton was indicated to provide a structural framework for proximal tubule cells to transmit mechanical forces and subsequently modulate cellular functions. In this study, we have used mouse proximal tubule (MPT) cells as a model to study the role of fluid shear stress (FSS) on apical NHE3 and V-ATPase and basolateral Na/K-ATPase trafficking and expression. Our hypothesis is that FSS stimulates both apical and basolateral transporter expression and trafficking, which subsequently mediates salt and volume reabsorption. We exposed MPT cells to 0.2 dynes/cm(2) FSS for 3 h and performed confocal microscopy and Western blot analysis to compare the localization and expression of both apical and basolateral transporters in control cells and cells subjected to FSS. Our findings show that FSS leads to an increment in the amount of protein expression, and a translocation of apical NHE3 and V-ATPase from the intracellular compartment to the apical plasma membrane and Na/K-ATPase to the basolateral membrane. Disrupting actin by cytochalasin D blocks the FSS-induced changes in NHE3 and Na/K-ATPase, but not V-ATPase. In contrast, FSS-induced V-ATPase redistribution and expression are largely inhibited by colchicine, an agent that blocks microtubule polymerization. Our findings suggest that the actin cytoskeleton plays an important role in FSS-induced NHE3 and Na/K-ATPase trafficking, and an intact microtubule network is critical in FSS-induced modulation of V-ATPase in proximal tubule cells.
Collapse
|
46
|
Weinbaum S, Duan Y, Satlin LM, Wang T, Weinstein AM. Mechanotransduction in the renal tubule. Am J Physiol Renal Physiol 2010; 299:F1220-36. [PMID: 20810611 DOI: 10.1152/ajprenal.00453.2010] [Citation(s) in RCA: 104] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022] Open
Abstract
The role of mechanical forces in the regulation of glomerulotubular balance in the proximal tubule (PT) and Ca(2+) signaling in the distal nephron was first recognized a decade ago, when it was proposed that the microvilli in the PT and the primary cilium in the cortical collecting duct (CCD) acted as sensors of local tubular flow. In this review, we present a summary of the theoretical models and experiments that have been conducted to elucidate the structure and function of these unique apical structures in the modulation of Na(+), HCO(3)(-), and water reabsorption in the PT and Ca(2+) signaling in the CCD. We also contrast the mechanotransduction mechanisms in renal epithelium with those in other cells in which fluid shear stresses have been recognized to play a key role in initiating intracellular signaling, most notably endothelial cells, hair cells in the inner ear, and bone cells. In each case, small hydrodynamic forces need to be greatly amplified before they can be sensed by the cell's intracellular cytoskeleton to enable the cell to regulate its membrane transporters or stretch-activated ion channels in maintaining homeostasis in response to changing flow conditions.
Collapse
Affiliation(s)
- Sheldon Weinbaum
- Dept. of Biomedical Engineering, The City College of New York, New York, NY 10031, USA.
| | | | | | | | | |
Collapse
|
47
|
Hu MC, Shi M, Zhang J, Pastor J, Nakatani T, Lanske B, Razzaque MS, Rosenblatt KP, Baum MG, Kuro-o M, Moe OW. Klotho: a novel phosphaturic substance acting as an autocrine enzyme in the renal proximal tubule. FASEB J 2010; 24:3438-50. [PMID: 20466874 DOI: 10.1096/fj.10-154765] [Citation(s) in RCA: 426] [Impact Index Per Article: 28.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Abstract
Klotho has profound effects on phosphate metabolism, but the mechanisms of how Klotho affects phosphate homeostasis is unknown. We detected Klotho in the proximal tubule cell, brush border, and urinary lumen, where phosphate homeostasis resides. Increasing Klotho in the kidney and urine chronically by transgenic overexpression or acutely by intravenous infusion caused hypophosphatemia, phosphaturia from decreased proximal phosphate reabsorption, and decreased activity and protein of the principal renal phosphate transporter NaPi-2a. The phosphaturic effect was present in FGF23-null mice, indicating a direct action distinct from Klotho's known role as a coreceptor for FGF23. Direct inhibition of NaPi-2a by Klotho was confirmed in cultured cells and in cell-free membrane vesicles characterized by acute inhibition of transport activity followed by decreased cell surface protein. Transport inhibition can be mimicked by recombinant beta-glucuronidase and is associated with proteolytic degradation and reduced surface NaPi-2a. The inhibitory effect of Klotho on NaPi-2a was blocked by beta-glucuronidase inhibitor but not by protease inhibitor. Klotho is a novel phosphaturic substance that acts as an enzyme in the proximal tubule urinary lumen by modifying glycans, which cause decreased transporter activity, followed by proteolytic degradation and possibly internalization of NaPi-2a from the apical membrane.
Collapse
Affiliation(s)
- Ming Chang Hu
- Department of Internal Medicine, University of Texas Southwestern Medical Center at Dallas, 5323 Harry Hines Blvd., Dallas, TX 75390-8885, USA
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
48
|
Bobulescu IA, Quiñones H, Gisler SM, Di Sole F, Hu MC, Shi M, Zhang J, Fuster DG, Wright N, Mumby M, Moe OW. Acute regulation of renal Na+/H+ exchanger NHE3 by dopamine: role of protein phosphatase 2A. Am J Physiol Renal Physiol 2010; 298:F1205-13. [PMID: 20181665 DOI: 10.1152/ajprenal.00708.2009] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023] Open
Abstract
Nephrogenic dopamine is a potent natriuretic paracrine/autocrine hormone that is central for mammalian sodium homeostasis. In the renal proximal tubule, dopamine induces natriuresis partly via inhibition of the sodium/proton exchanger NHE3. The signal transduction pathways and mechanisms by which dopamine inhibits NHE3 are complex and incompletely understood. This manuscript describes the role of the serine/threonine protein phosphatase 2A (PP2A) in the regulation of NHE3 by dopamine. The PP2A regulatory subunit B56δ (coded by the Ppp2r5d gene) directly associates with more than one region of the carboxy-terminal hydrophilic putative cytoplasmic domain of NHE3 (NHE3-cyto), as demonstrated by yeast-two-hybrid, coimmunoprecipitation, blot overlay, and in vitro pull-down assays. Phosphorylated NHE3-cyto is a substrate for purified PP2A in an in vitro dephosphorylation reaction. In cultured renal cells, inhibition of PP2A by either okadaic acid or by overexpression of the simian virus 40 (SV40) small T antigen blocks the ability of dopamine to inhibit NHE3 activity and to reduce surface NHE3 protein. Dopamine-induced NHE3 redistribution is also blocked by okadaic acid ex vivo in rat kidney cortical slices. These studies demonstrate that PP2A is an integral and critical participant in the signal transduction pathway between dopamine receptor activation and NHE3 inhibition.
Collapse
Affiliation(s)
- I Alexandru Bobulescu
- Dept. of Internal Medicine, Div. of Nephrology, Univ. of Texas Southwestern Medical Center, 5323 Harry Hines Blvd., Dallas, TX 75390-8856, USA
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
49
|
Liu J, Xie ZJ. The sodium pump and cardiotonic steroids-induced signal transduction protein kinases and calcium-signaling microdomain in regulation of transporter trafficking. Biochim Biophys Acta Mol Basis Dis 2010; 1802:1237-45. [PMID: 20144708 DOI: 10.1016/j.bbadis.2010.01.013] [Citation(s) in RCA: 96] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2009] [Revised: 01/28/2010] [Accepted: 01/30/2010] [Indexed: 12/12/2022]
Abstract
The Na/K-ATPase was discovered as an energy transducing ion pump. A major difference between the Na/K-ATPase and other P-type ATPases is its ability to bind a group of chemicals called cardiotonic steroids (CTS). The plant-derived CTS such as digoxin are valuable drugs for the management of cardiac diseases, whereas ouabain and marinobufagenin (MBG) have been identified as a new class of endogenous hormones. Recent studies have demonstrated that the endogenous CTS are important regulators of renal Na(+) excretion and blood pressure. The Na/K-ATPase is not only an ion pump, but also an important receptor that can transduce the ligand-like effect of CTS on intracellular protein kinases and Ca(2+) signaling. Significantly, these CTS-provoked signaling events are capable of reducing the surface expression of apical NHE3 (Na/H exchanger isoform 3) and basolateral Na/K-ATPase in renal proximal tubular cells. These findings suggest that endogenous CTS may play an important role in regulation of tubular Na(+) excretion under physiological conditions; conversely, a defect at either the receptor level (Na/K-ATPase) or receptor-effector coupling would reduce the ability of renal proximal tubular cells to excrete Na(+), thus culminating/resulting in salt-sensitive hypertension.
Collapse
Affiliation(s)
- Jiang Liu
- Department of Medicine, University of Toledo College of Medicine, Toledo, OH, USA
| | | |
Collapse
|
50
|
Musch MW, Arvans DL, Wang Y, Nakagawa Y, Solomaha E, Chang EB. Cyclic AMP-mediated endocytosis of intestinal epithelial NHE3 requires binding to synaptotagmin 1. Am J Physiol Gastrointest Liver Physiol 2010; 298:G203-11. [PMID: 19926819 PMCID: PMC2822502 DOI: 10.1152/ajpgi.00379.2009] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
The apical membrane Na(+)-H(+) exchanger (NHE)3 is regulated by cAMP-dependent phosphorylation, which inhibits its activity through membrane endocytosis. The clathrin complex adaptor protein synaptotagmin 1 (Syt 1) appears to be essential to this process, but little is known about its expression in intestinal epithelial cells or interaction with NHE3. The intestinal epithelial expression and apical location of Syt 1 were determined by Syt 1 mRNA profiling and immunolocalization. Tandem mass spectrometry was used for protein identification. Bis(sulfosuccinimidyl) suberate (BS(3)) cross linking suggested that NHE3 and Syt 1 were in a membrane complex following cAMP stimulation of Caco2BBE (Brush Border Expressions) cells. To investigate the regulation of NHE3 appearance in a Syt 1-containing membrane compartment, doxycycline-inducible hemaglutinin (HA)-tagged NHE3 was expressed in Caco2BBE cells. HA-NHE3 correctly targeted to the apical membrane, where, upon cAMP stimulation, it was internalized with a Syt 1-containing compartment. Site-directed mutagenesis of NHE3 showed that serine 605 (S605) was pivotal to NHE3 and Syt 1 association and internalization. Direct Syt 1 interaction with NHE3 was suggested by fluorescence resonance energy transfer (FRET) analysis. The physiological role of S552 was less clear. By FRET, this serine residue appeared to be involved in cAMP-induced Syt 1 binding of NHE3. However, when HA-tagged NHE3 S552A was expressed in Caco2 cells, the mutated construct was not inserted into the apical membrane. We conclude that intestinal epithelial Syt 1 plays an important role in cAMP-stimulated endocytosis of apical NHE3 through cAMP-dependent phosphorylation of S605 that is required for NHE3 and Syt 1 association.
Collapse
Affiliation(s)
| | | | - Yunwei Wang
- 1Martin Boyer Laboratories, Department of Medicine;
| | | | - Elena Solomaha
- 2Biophysical Research Core Facility, Divisions of Biological and Physical Sciences, University of Chicago, Chicago, Illinois
| | | |
Collapse
|