1
|
Xu Y, Li C, Yin H, Nowsheen S, Xu X, Kang W, Liu X, Chen L, Lou Z, Yi J, Deng M. STK39-mediated amplification of γ-H2A.X promotes homologous recombination and contributes to PARP inhibitor resistance. Nucleic Acids Res 2024; 52:13881-13895. [PMID: 39588777 DOI: 10.1093/nar/gkae1099] [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: 07/04/2024] [Revised: 09/17/2024] [Accepted: 10/28/2024] [Indexed: 11/27/2024] Open
Abstract
The phosphorylation of histone H2A.X into γH2A.X is a crucial early event in the DNA damage response, marking DNA damage sites and initiating repair processes. While ATM kinase is traditionally recognized as the primary mediator of H2A.X phosphorylation, our study identifies serine/threonine kinase 39 (STK39) as a novel enhancer of this critical signaling pathway. We demonstrate that after DNA damage, STK39 undergoes phosphorylation by the ATM kinase, facilitating its interaction with the Mre11-Rad50-Nbs1 complex and subsequent recruitment to chromatin. This recruitment enables STK39 to further phosphorylate H2A.X, thus amplifying γH2A.X production and promoting homologous recombination repair. Notably, we observe a significant upregulation of STK39 in pancreatic adenocarcinoma (PAAD) tissues, correlating with heightened resistance to PARPi therapy. Furthermore, we demonstrate the synergistic efficacy of combining STK39 inhibition with PARP inhibitors in suppressing and reversing PAAD growth. This study not only provides new insights into the molecular dynamics of H2A.X phosphorylation but also highlights the therapeutic potential of targeting STK39 to enhance PARPi sensitivity in PAAD (created with BioRender).
Collapse
Affiliation(s)
- Yi Xu
- State Key Laboratory of Molecular Oncology and Department of Radiation Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Panjiayuan Nanli No17, Chaoyang District, Beijing 100021, China
| | - Changying Li
- State Key Laboratory of Molecular Oncology and Department of Radiation Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Panjiayuan Nanli No17, Chaoyang District, Beijing 100021, China
| | - Huan Yin
- State Key Laboratory of Molecular Oncology and Department of Radiation Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Panjiayuan Nanli No17, Chaoyang District, Beijing 100021, China
| | - Somaira Nowsheen
- Department of Dermatology, University of California San Diego, 9500 Gilman Drive, La Jolla, San Diego, CA 92122, USA
| | - Xin Xu
- State Key Laboratory of Molecular Oncology and Department of Radiation Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Panjiayuan Nanli No17, Chaoyang District, Beijing 100021, China
| | - Wenjuan Kang
- State Key Laboratory of Molecular Oncology and Department of Radiation Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Panjiayuan Nanli No17, Chaoyang District, Beijing 100021, China
| | - Xin Liu
- State Key Laboratory of Molecular Oncology and Department of Radiation Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Panjiayuan Nanli No17, Chaoyang District, Beijing 100021, China
| | - Lifeng Chen
- Department of Gynecology, the First Affiliated Hospital, School of Medicine, Zhejiang University, No.79 Qingchun Road, Shangcheng District, Hangzhou 310003, China
| | - Zhenkun Lou
- Department of Oncology, Mayo Clinic, 200 First St SW, Rochester, MN 55905, USA
| | - Junlin Yi
- State Key Laboratory of Molecular Oncology and Department of Radiation Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Panjiayuan Nanli No17, Chaoyang District, Beijing 100021, China
| | - Min Deng
- State Key Laboratory of Molecular Oncology and Department of Radiation Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Panjiayuan Nanli No17, Chaoyang District, Beijing 100021, China
| |
Collapse
|
2
|
Maskey D, Granados Pineda J, Ortiz PA. Update on NKCC2 regulation in the thick ascending limb (TAL) by membrane trafficking, phosphorylation, and protein-protein interactions. Front Physiol 2024; 15:1508806. [PMID: 39717823 PMCID: PMC11663917 DOI: 10.3389/fphys.2024.1508806] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2024] [Accepted: 11/22/2024] [Indexed: 12/25/2024] Open
Abstract
Purpose of review The thick ascending limb (TAL) of loop of Henle is essential for NaCl, calcium and magnesium homeostasis, pH balance and for urine concentration. NKCC2 is the main transporter for NaCl reabsorption in the TAL and its regulation is very complex. There have been recent advancements toward understanding how NKCC2 is regulated by protein trafficking, protein-protein interaction, and phosphorylation/dephosphorylation. Here, we update the latest molecular mechanisms and players that control NKCC2 function, which gives an increasingly complex picture of NKKC2 regulation in the apical membrane of the TAL. Recent Findings Protein-protein interactions are required as a regulatory mechanism in many cellular processes. A handful of proteins have been recently identified as an interacting partner of NKCC2, which play major roles in regulating NKCC2 trafficking and activity. New players in NKCC2 internalization and trafficking have been identified. NKCC2 activity is also regulated by kinases and phosphatases, and there have been developments in that area as well. Summary Here we review the current understanding of apical trafficking of NKCC2 in the thick ascending limb (TAL) which is tightly controlled by protein-protein interactions, protein turnover and by phosphorylation and dephosphorylation. We discuss new proteins and processes that regulate NKCC2 that have physiological and pathological significance.
Collapse
Affiliation(s)
- Dipak Maskey
- Department of Internal Medicine, Hypertension and Vascular Research Division, Henry ford hospital, Detroit, MI, United States
- Department of Physiology, Integrative Bioscience Center, Wayne State University, Detroit, MI, United States
| | - Jessica Granados Pineda
- Department of Internal Medicine, Hypertension and Vascular Research Division, Henry ford hospital, Detroit, MI, United States
- Department of Physiology, Integrative Bioscience Center, Wayne State University, Detroit, MI, United States
| | - Pablo A. Ortiz
- Department of Internal Medicine, Hypertension and Vascular Research Division, Henry ford hospital, Detroit, MI, United States
- Department of Physiology, Integrative Bioscience Center, Wayne State University, Detroit, MI, United States
| |
Collapse
|
3
|
Polizio AH, Marino L, Duk-Min K, Yura Y, Rolauer L, Cochran JD, Evans MA, Park E, Doviak H, Miura-Yura E, Good ME, Wolpe AG, Grandoch M, Isakson B, Walsh K. Experimental TET2 Clonal Hematopoiesis Predisposes to Renal Hypertension Through an Inflammasome-Mediated Mechanism. Circ Res 2024; 135:933-950. [PMID: 39234670 PMCID: PMC11519839 DOI: 10.1161/circresaha.124.324492] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/22/2024] [Revised: 08/26/2024] [Accepted: 08/29/2024] [Indexed: 09/06/2024]
Abstract
BACKGROUND Hypertension incidence increases with age and represents one of the most prevalent risk factors for cardiovascular disease. Clonal events in the hematopoietic system resulting from somatic mutations in driver genes are prevalent in elderly individuals who lack overt hematologic disorders. This condition is referred to as age-related clonal hematopoiesis (CH), and it is a newly recognized risk factor for cardiovascular disease. It is not known whether CH and hypertension in the elderly are causally related and, if so, what are the mechanistic features. METHODS A murine model of adoptive bone marrow transplantation was employed to examine the interplay between Tet2 (ten-eleven translocation methylcytosine dioxygenase 2) clonal hematopoiesis and hypertension. RESULTS In this model, a subpressor dose of Ang II (angiotensin II) resulted in elevated systolic and diastolic blood pressure as early as 1 day after challenge. These conditions led to the expansion of Tet2-deficient proinflammatory monocytes and bone marrow progenitor populations. Tet2 deficiency promoted renal CCL5 (C-C motif ligand 5) chemokine expression and macrophage infiltration into the kidney. Consistent with macrophage involvement, Tet2 deficiency in myeloid cells promoted hypertension when mice were treated with a subpressor dose of Ang II. The hematopoietic Tet2-/- condition led to sodium retention, renal inflammasome activation, and elevated levels of IL (interleukin)-1β and IL-18. Analysis of the sodium transporters indicated NCC (sodium-chloride symporter) and NKCC2 (Na+-K+-Cl- cotransporter 2) activation at residues Thr53 and Ser105, respectively. Administration of the NLRP3 (NLR family pyrin domain containing 3) inflammasome inhibitor MCC950 reversed the hypertensive state, sodium retention, and renal transporter activation. CONCLUSIONS Tet2-mediated CH sensitizes mice to a hypertensive stimulus. Mechanistically, the expansion of hematopoietic Tet2-deficient cells promotes hypertension due to elevated renal immune cell infiltration and activation of the NLRP3 inflammasome, with consequences on sodium retention. These data indicate that carriers of TET2 CH could be at elevated risk for the development of hypertension and that immune modulators could be useful in treating hypertension in this patient population.
Collapse
Affiliation(s)
- Ariel H. Polizio
- Cardiovascular Medicine and the Robert M. Berne Cardiovascular Research Center, University of Virginia School of Medicine, Charlottesville, VA, USA
| | - Lucila Marino
- Cardiovascular Medicine and the Robert M. Berne Cardiovascular Research Center, University of Virginia School of Medicine, Charlottesville, VA, USA
| | - Kyung Duk-Min
- Cardiovascular Medicine and the Robert M. Berne Cardiovascular Research Center, University of Virginia School of Medicine, Charlottesville, VA, USA
| | - Yoshimitsu Yura
- Cardiovascular Medicine and the Robert M. Berne Cardiovascular Research Center, University of Virginia School of Medicine, Charlottesville, VA, USA
| | - Luca Rolauer
- Institute of Translational Pharmacology, University Hospital Düsseldorf, Heinrich-Heine-University Düsseldorf, Düsseldorf, Germany
| | - Jesse D. Cochran
- Cardiovascular Medicine and the Robert M. Berne Cardiovascular Research Center, University of Virginia School of Medicine, Charlottesville, VA, USA
- Medical Scientist Training Program, University of Virginia School of Medicine, Charlottesville, VA 22908, USA
| | - Megan A. Evans
- Cardiovascular Medicine and the Robert M. Berne Cardiovascular Research Center, University of Virginia School of Medicine, Charlottesville, VA, USA
| | - Eunbee Park
- Cardiovascular Medicine and the Robert M. Berne Cardiovascular Research Center, University of Virginia School of Medicine, Charlottesville, VA, USA
| | - Heather Doviak
- Cardiovascular Medicine and the Robert M. Berne Cardiovascular Research Center, University of Virginia School of Medicine, Charlottesville, VA, USA
| | - Emiri Miura-Yura
- Cardiovascular Medicine and the Robert M. Berne Cardiovascular Research Center, University of Virginia School of Medicine, Charlottesville, VA, USA
| | - Miranda E. Good
- Molecular Cardiology Research Institute, Tufts Medical Center, Boston MA, 02111, USA
| | | | - Maria Grandoch
- Institute of Translational Pharmacology, University Hospital Düsseldorf, Heinrich-Heine-University Düsseldorf, Düsseldorf, Germany
- CARID, Cardiovascular Research Institute Düsseldorf, Medical Faculty and University Düsseldorf, Heinrich Heine University Düsseldorf, Germany
| | - Brant Isakson
- Cardiovascular Medicine and the Robert M. Berne Cardiovascular Research Center, University of Virginia School of Medicine, Charlottesville, VA, USA
| | - Kenneth Walsh
- Cardiovascular Medicine and the Robert M. Berne Cardiovascular Research Center, University of Virginia School of Medicine, Charlottesville, VA, USA
| |
Collapse
|
4
|
Rodan AR. With No Lysine (K) Kinases and Sodium Transporter Function in Solute Exchange with Implications for BP Regulation as Elucidated through Drosophila. KIDNEY360 2024; 5:1553-1562. [PMID: 39186374 PMCID: PMC11556937 DOI: 10.34067/kid.0000000000000564] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/03/2024] [Accepted: 08/20/2024] [Indexed: 08/28/2024]
Abstract
Like other multicellular organisms, the fruit fly Drosophila melanogaster must maintain homeostasis of the internal milieu, including the maintenance of constant ion and water concentrations. In mammals, the with no lysine (K) (WNK)-Ste20-proline/alanine rich kinase/oxidative stress response 1 kinase cascade is an important regulator of epithelial ion transport in the kidney. This pathway regulates SLC12 family cotransporters, including sodium-potassium-2-chloride, sodium chloride, and potassium chloride cotransporters. The WNK-Ste20-proline/alanine rich kinase/oxidative stress response 1 kinase cascade also regulates epithelial ion transport via regulation of the Drosophila sodium-potassium-2-chloride cotransporter in the Malpighian tubule, the renal epithelium of the fly. Studies in Drosophila have contributed to the understanding of multiple regulators of WNK pathway signaling, including intracellular chloride and potassium, the scaffold protein Mo25, hypertonic stress, hydrostatic pressure, and macromolecular crowding. These will be discussed together, with implications for mammalian kidney function and BP control.
Collapse
Affiliation(s)
- Aylin R Rodan
- Molecular Medicine Program, University of Utah, Salt Lake City, Utah; Division of Nephrology and Hypertension, Department of Internal Medicine, University of Utah, Salt Lake City, Utah; Department of Human Genetics, University of Utah, Salt Lake City, Utah; and Medical Service, Veterans Affairs Salt Lake City Health Care System, Salt Lake City, Utah
| |
Collapse
|
5
|
Peng L, Zhang Z, Du W, Zhu J, Duan W. Proteomic and Phosphoproteomic analysis of thyroid papillary carcinoma: Identification of potential biomarkers for metastasis. J Proteomics 2024; 306:105260. [PMID: 39029786 DOI: 10.1016/j.jprot.2024.105260] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2023] [Revised: 07/14/2024] [Accepted: 07/15/2024] [Indexed: 07/21/2024]
Abstract
Thyroid cancer has emerged as the most rapidly proliferating solid neoplasm. In this study, we included a cohort of patients who underwent sonographic assessment and surgical intervention at the Sir Run Run Shaw Hospital, associated with the School of Medicine at Zhejiang University, spanning from January 2019 to June 2020. Stratification of cases was based on a combination of preoperative ultrasonographic evaluations and postoperative histopathological diagnoses, resulting in three distinct groups: high-risk papillary thyroid carcinoma (PTC) labeled as C1, low-risk PTC designated as C2, and a control group (N) composed of benign thyroid tissue adjacent to the carcinoma. Proteomic and phosphoproteomic analyses were conducted on PTC specimens. The comparative assessment revealed that proteins up-regulated in the C1/N and C2/N groups were predominantly involved in functions such as amino acid binding, binding of phosphorylated compounds, and serine protease activity. Notably, proteins like NADH dehydrogenase, ATP synthase, oxidoreductases, and iron ion channels were significantly elevated in the C1 versus C2 comparative group. Through meticulous analysis of differential expression multiples, statistical significance, and involvement in metabolic pathways, this study identified eight potential biomarkers pertinent to PTC metastasis diagnostics, encompassing phosphorylated myosin 10, phosphorylated proline-directed protein kinase, leucine tRNA synthetase, 2-oxo-isovalerate dehydrogenase, succinic semialdehyde dehydrogenase, ADP/ATPtranslocase, pyruvate carboxylase, and fibrinogen. Therapeutic assays employing metformin, an AMP-activated protein kinase (AMPK) activator, alongside the phosphorylation-specific inhibitor ML-7 targeting Myosin10, demonstrated attenuated cellular proliferation, migration, and invasion capabilities in thyroid cancer cells, accompanied by a reduction in amino acid pools. Cellular colocalization and interaction studies elucidated that AMPK activation imposes an inhibitory influence on Myosin10 levels. The findings of this research corroborate the utility of proteomic and phosphoproteomic platforms in the identification of metastatic markers for PTC and suggest that modulation of AMPK activity, coupled with the inhibition of Myosin10 phosphorylation, may forge novel therapeutic avenues in the management of thyroid carcinoma. SIGNIFICANCE: The significance of our research lies in its potential to transform the current understanding and management of thyroid papillary carcinoma (PTC), particularly in its metastatic form. By integrating both proteomic and phosphoproteomic analyses, our study not only sheds light on the molecular alterations associated with PTC but also identifies eight novel biomarkers that could serve as indicators of metastatic potential.
Collapse
Affiliation(s)
- Lingyao Peng
- Hangzhou TCM Hospital Affiliated to Zhejiang Chinese Medical University, Hangzhou 310007, China
| | - Zhenxian Zhang
- Hangzhou TCM Hospital Affiliated to Zhejiang Chinese Medical University, Hangzhou 310007, China
| | - Wei Du
- Hangzhou Institute of Standardization, Hangzhou 310000, China
| | - Jiang Zhu
- Women's Hospital School of Medicine Zhejiang University, 310006 Hangzhou, China.
| | - Wenkai Duan
- Hangzhou Vocational and Technical College, Hangzhou 310018, China.
| |
Collapse
|
6
|
Duan XP, Zhang CB, Wang WH, Lin DH. Role of calcineurin in regulating renal potassium (K +) excretion: Mechanisms of calcineurin inhibitor-induced hyperkalemia. Acta Physiol (Oxf) 2024; 240:e14189. [PMID: 38860527 PMCID: PMC11250626 DOI: 10.1111/apha.14189] [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: 03/21/2024] [Revised: 05/28/2024] [Accepted: 05/30/2024] [Indexed: 06/12/2024]
Abstract
Calcineurin, protein phosphatase 2B (PP2B) or protein phosphatase 3 (PP3), is a calcium-dependent serine/threonine protein phosphatase. Calcineurin is widely expressed in the kidney and regulates renal Na+ and K+ transport. In the thick ascending limb, calcineurin plays a role in inhibiting NKCC2 function by promoting the dephosphorylation of the cotransporter and an intracellular sorting receptor, called sorting-related-receptor-with-A-type repeats (SORLA), is involved in modulating the effect of calcineurin on NKCC2. Calcineurin also participates in regulating thiazide-sensitive NaCl-cotransporter (NCC) in the distal convoluted tubule. The mechanisms by which calcineurin regulates NCC include directly dephosphorylation of NCC, regulating Kelch-like-3/CUL3 E3 ubiquitin-ligase complex, which is responsible for WNK (with-no-lysin-kinases) ubiquitination, and inhibiting Kir4.1/Kir5.1, which determines NCC expression/activity. Finally, calcineurin is also involved in regulating ROMK (Kir1.1) channels in the cortical collecting duct and Cyp11 2 expression in adrenal zona glomerulosa. In summary, calcineurin is involved in the regulation of NKCC2, NCC, and inwardly rectifying K+ channels in the kidney, and it also plays a role in modulating aldosterone synthesis in adrenal gland, which regulates epithelial-Na+-channel expression/activity. Thus, application of calcineurin inhibitors (CNIs) is expected to abrupt calcineurin-mediated regulation of transepithelial Na+ and K+ transport in the kidney. Consequently, CNIs cause hypertension, compromise renal K+ excretion, and induce hyperkalemia.
Collapse
Affiliation(s)
- Xin-Peng Duan
- Department of Physiology, Xuzhou Medical University, Xuzhou, China
| | - Cheng-Biao Zhang
- Department of Physiology, Xuzhou Medical University, Xuzhou, China
- Department of Pharmacology, New York Medical College, Valhalla, New York, USA
| | - Wen-Hui Wang
- Department of Pharmacology, New York Medical College, Valhalla, New York, USA
| | - Dao-Hong Lin
- Department of Pharmacology, New York Medical College, Valhalla, New York, USA
| |
Collapse
|
7
|
Chen C, Zhong W, Zheng H, Dai G, Zhao W, Wang Y, Dong Q, Shen B. The role of uromodulin in cardiovascular disease: a review. Front Cardiovasc Med 2024; 11:1417593. [PMID: 39049957 PMCID: PMC11267628 DOI: 10.3389/fcvm.2024.1417593] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2024] [Accepted: 06/25/2024] [Indexed: 07/27/2024] Open
Abstract
Uromodulin, also referred to as Tamm Horsfall protein (THP), is a renal protein exclusively synthesized by the kidneys and represents the predominant urinary protein under normal physiological conditions. It assumes a pivotal role within the renal system, contributing not only to ion transport and immune modulation but also serving as a critical factor in the prevention of urinary tract infections and kidney stone formation. Emerging evidence indicates that uromodulin may serve as a potential biomarker extending beyond renal function. Recent clinical investigations and Mendelian randomization studies have unveiled a discernible association between urinary regulatory protein levels and cardiovascular events and mortality. This review primarily delineates the intricate relationship between uromodulin and cardiovascular disease, elucidates its predictive utility as a novel biomarker for cardiovascular events, and delves into its involvement in various physiological and pathophysiological facets of the cardiovascular system, incorporating recent advancements in corresponding genetics.
Collapse
Affiliation(s)
- Chengqian Chen
- Department of Cardiology Center, The First Hospital of Jilin University, Changchun, China
| | - Wentao Zhong
- Department of Endocrinology and Metabolism, The First Hospital of Jilin University, Changchun, China
| | - Hao Zheng
- Department of Endocrinology and Metabolism, The First Hospital of Jilin University, Changchun, China
| | - Gaoying Dai
- Department of Cardiology Center, The First Hospital of Jilin University, Changchun, China
| | - Wei Zhao
- Department of Cardiology Center, The First Hospital of Jilin University, Changchun, China
| | - Yushi Wang
- Department of Cardiology Center, The First Hospital of Jilin University, Changchun, China
| | - Qi Dong
- Department of Cardiology Center, The First Hospital of Jilin University, Changchun, China
| | - Botao Shen
- Department of Cardiology Center, The First Hospital of Jilin University, Changchun, China
| |
Collapse
|
8
|
Taylor CA, Jung JU, Kankanamalage SG, Li J, Grzemska M, Jaykumar AB, Earnest S, Stippec S, Saha P, Sauceda E, Cobb MH. Predictive and Experimental Motif Interaction Analysis Identifies Functions of the WNK-OSR1/SPAK Pathway. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.06.26.600905. [PMID: 38979344 PMCID: PMC11230372 DOI: 10.1101/2024.06.26.600905] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/10/2024]
Abstract
The WNK-OSR1/SPAK protein kinase signaling pathway regulates ion homeostasis and cell volume, but its other functions are poorly understood. To uncover undefined signaling functions of the pathway we analyzed the binding specificity of the conserved C-terminal (CCT) domains of OSR1 and SPAK to find all possible interaction motifs in human proteins. These kinases bind the core consensus sequences R-F-x-V/I and R-x-F-x-V/I. Motifs were ranked based on sequence, conservation, cellular localization, and solvent accessibility. Out of nearly 3,700 motifs identified, 90% of previously published motifs were within the top 2% of those predicted. Selected candidates (TSC22D1, CAVIN1, ATG9A, NOS3, ARHGEF5) were tested. Upstream kinases WNKs 1-4 and their close relatives, the pseudokinases NRBP1/2, contain CCT-like domains as well. We identified additional distinct motif variants lacking the conserved arginine previously thought to be required, and found that the NRBP1 CCT-like domain binds TSC22D1 via the same motif as OSR1 and SPAK. Our results further highlight the rich and diverse functionality of CCT and CCT-like domains in connecting WNK signaling to cellular processes.
Collapse
|
9
|
Kok M, Brodsky JL. The biogenesis of potassium transporters: implications of disease-associated mutations. Crit Rev Biochem Mol Biol 2024; 59:154-198. [PMID: 38946646 PMCID: PMC11444911 DOI: 10.1080/10409238.2024.2369986] [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: 03/29/2024] [Revised: 06/02/2024] [Accepted: 06/16/2024] [Indexed: 07/02/2024]
Abstract
The concentration of intracellular and extracellular potassium is tightly regulated due to the action of various ion transporters, channels, and pumps, which reside primarily in the kidney. Yet, potassium transporters and cotransporters play vital roles in all organs and cell types. Perhaps not surprisingly, defects in the biogenesis, function, and/or regulation of these proteins are linked to range of catastrophic human diseases, but to date, few drugs have been approved to treat these maladies. In this review, we discuss the structure, function, and activity of a group of potassium-chloride cotransporters, the KCCs, as well as the related sodium-potassium-chloride cotransporters, the NKCCs. Diseases associated with each of the four KCCs and two NKCCs are also discussed. Particular emphasis is placed on how these complex membrane proteins fold and mature in the endoplasmic reticulum, how non-native forms of the cotransporters are destroyed in the cell, and which cellular factors oversee their maturation and transport to the cell surface. When known, we also outline how the levels and activities of each cotransporter are regulated. Open questions in the field and avenues for future investigations are further outlined.
Collapse
Affiliation(s)
- Morgan Kok
- Department of Biological Sciences, University of Pittsburgh, Pittsburgh, PA, USA
| | - Jeffrey L Brodsky
- Department of Biological Sciences, University of Pittsburgh, Pittsburgh, PA, USA
| |
Collapse
|
10
|
Higashihara E, Harada T, Fukuhara H. Juxtaglomerular apparatus-mediated homeostatic mechanisms: therapeutic implication for chronic kidney disease. Expert Opin Pharmacother 2024; 25:819-832. [PMID: 38773961 DOI: 10.1080/14656566.2024.2357188] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2024] [Accepted: 05/15/2024] [Indexed: 05/24/2024]
Abstract
INTRODUCTION Juxtaglomerular apparatus (JGA)-mediated homeostatic mechanism links to how sodium-glucose cotransporter 2 inhibitors (SGLT2is) slow progression of chronic kidney disease (CKD) and may link to how tolvaptan slows renal function decline in autosomal dominant polycystic kidney disease (ADPKD). AREA COVERED JGA-mediated homeostatic mechanism has been hypothesized based on investigations of tubuloglomerular feedback and renin-angiotensin system. We reviewed clinical trials of SGLT2is and tolvaptan to assess the relationship between this mechanism and these drugs. EXPERT OPINION When sodium load to macula densa (MD) increases, MD increases adenosine production, constricting afferent arteriole (Af-art) and protecting glomeruli. Concurrently, MD signaling suppresses renin secretion, increases urinary sodium excretion, and counterbalances reduced sodium filtration. However, when there is marked increase in sodium load per-nephron, as in advanced CKD, MD adenosine production increases, relaxing Af-art and maintaining sodium homeostasis at the expense of glomeruli. The beneficial effects of tolvaptan on renal function in ADPKD may also depend on the JGA-mediated homeostatic mechanisms since tolvaptan inhibits sodium reabsorption in the thick ascending limb.The JGA-mediated homeostatic mechanism regulates Af-arts, constricting to relaxing according to homeostatic needs. Understanding this mechanism may contribute to the development of pharmacotherapeutic compounds and better care for patients with CKD.
Collapse
Affiliation(s)
- Eiji Higashihara
- Department of Urology, Kyorin University School of Medicine, Mitaka, Japan
| | - Takeo Harada
- Department of Renal and Cardiovascular Research, Otsuka Pharmaceutical Co. Ltd, Tokushima, Japan
| | - Hiroshi Fukuhara
- Department of Urology, Kyorin University School of Medicine, Mitaka, Japan
| |
Collapse
|
11
|
AlShanableh Z, Ray EC. Magnesium in hypertension: mechanisms and clinical implications. Front Physiol 2024; 15:1363975. [PMID: 38665599 PMCID: PMC11044701 DOI: 10.3389/fphys.2024.1363975] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2023] [Accepted: 03/25/2024] [Indexed: 04/28/2024] Open
Abstract
Hypertension is associated with increased risk of cardiovascular disease and death. Evidence suggests that Mg2+ depletion contributes to hypertension. It is estimated that 25% or more of the United States population experiences chronic, latent Mg2+ depletion. This review explores mechanisms by which Mg2+ influences blood pressure, modifying risk of hypertension and complicating its treatment. Mechanisms addressed include effects upon i) sympathetic tone, via the modulation of N-methyl-D-aspartate (NMDA) receptor and N-type Ca2+ channel activity, influencing catecholamine release from sympathetic nerve endings; ii) vascular tone, via alteration of L-type Ca2+ and endothelial nitric oxide synthase (eNOS) activity and prostacyclin release; iii) renal K+ handling, influencing systemic K+ balance and potentially indirectly influencing blood pressure; iv) aldosterone secretion from the adrenal cortex; and v) modulation of pro-hypertensive inflammatory processes in dendritic cells and macrophages, including activation of the NLR family pyrin domain containing 3 (NLRP3) inflammasome and stimulation of isolevuglandin (IsoLG) production. Discovery of these mechanisms has furthered our understanding of the pathogenesis of hypertension, with implications for treatment and has highlighted the role of Mg2+ balance in hypertension and cardiovascular disease.
Collapse
Affiliation(s)
| | - Evan C. Ray
- Renal-Electrolyte Division, UPMC and University of Pittsburgh School of Medicine, Pittsburgh, PA, United States
| |
Collapse
|
12
|
Han X, Akinseye L, Sun Z. KDM6A Demethylase Regulates Renal Sodium Excretion and Blood Pressure. Hypertension 2024; 81:541-551. [PMID: 38164755 PMCID: PMC10922853 DOI: 10.1161/hypertensionaha.123.22026] [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: 09/07/2023] [Accepted: 12/13/2023] [Indexed: 01/03/2024]
Abstract
BACKGROUND KDM6A (Lysine-Specific Demethylase 6A) is a specific demethylase for histone 3 lysine (K) 27 trimethylation (H3K27me3). The purpose of this study is to investigate whether KDM6A in renal tubule cells plays a role in the regulation of kidney function and blood pressure. METHODS We first crossed Ksp-Cre+/- and KDM6Aflox/flox mice for generating inducible kidney-specific deletion of KDM6A gene. RESULTS Notably, conditional knockout of KDM6A gene in renal tubule cells (KDM6A-cKO) increased H3K27me3 levels which leads to a decrease in Na excretion and elevation of blood pressure. Further analysis showed that the expression of NKCC2 (Na-K-2Cl cotransporter 2) and NCC (Na-Cl cotransporters) was upregulated which contributes to impaired Na excretion in KDM6A-cKO mice. The expression of AQP2 (aquaporin 2) was also increased in KDM6A-cKO mice, which may facilitate water reabsorption in KDM6A-cKO mice. The expression of Klotho was downregulated while expression of aging markers including p53, p21, and p16 was upregulated in kidneys of KDM6A-cKO mice, indicating that deletion of KDM6A in the renal tubule cells promotes kidney aging. Interestingly, KDM6A-cKO mice developed salt-sensitive hypertension which can be rescued by treatment with Klotho. KDM6A deficiency induced salt-sensitive hypertension likely through downregulation of the Klotho/ERK (extracellular signal-regulated kinase) signaling and upregulation of the WNK (with-no-lysine kinase) signaling. CONCLUSIONS This study provides the first evidence that KDM6A plays an essential role in maintaining normal tubular function and blood pressure. Renal tubule cell specific KDM6A deficiency causes hypertension due to increased H3K27me3 levels and the resultant downregulation of Klotho gene expression which disrupts the Klotho/ERK/NCC/NKCC2 signaling.
Collapse
Affiliation(s)
- Xiaobin Han
- Department of Physiology, University of Tennessee Health Science Center, Memphis, TN 38163, USA
| | - Leah Akinseye
- Department of Physiology, University of Tennessee Health Science Center, Memphis, TN 38163, USA
| | - Zhongjie Sun
- Department of Physiology, University of Tennessee Health Science Center, Memphis, TN 38163, USA
| |
Collapse
|
13
|
Carbajal-Contreras H, Murillo-de-Ozores AR, Magaña-Avila G, Marquez-Salinas A, Bourqui L, Tellez-Sutterlin M, Bahena-Lopez JP, Cortes-Arroyo E, Behn-Eschenburg SG, Lopez-Saavedra A, Vazquez N, Ellison DH, Loffing J, Gamba G, Castañeda-Bueno M. Arginine vasopressin regulates the renal Na +-Cl - and Na +-K +-Cl - cotransporters through with-no-lysine kinase 4 and inhibitor 1 phosphorylation. Am J Physiol Renal Physiol 2024; 326:F285-F299. [PMID: 38096266 PMCID: PMC11207557 DOI: 10.1152/ajprenal.00343.2023] [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: 10/24/2023] [Revised: 11/15/2023] [Accepted: 12/03/2023] [Indexed: 01/25/2024] Open
Abstract
Vasopressin regulates water homeostasis via the V2 receptor in the kidney at least in part through protein kinase A (PKA) activation. Vasopressin, through an unknown pathway, upregulates the activity and phosphorylation of Na+-Cl- cotransporter (NCC) and Na+-K+-2Cl- cotransporter 2 (NKCC2) by Ste20-related proline/alanine-rich kinase (SPAK) and oxidative stress-responsive kinase 1 (OSR1), which are regulated by the with-no-lysine kinase (WNK) family. Phosphorylation of WNK4 at PKA consensus motifs may be involved. Inhibitor 1 (I1), a protein phosphatase 1 (PP1) inhibitor, may also play a role. In human embryonic kidney (HEK)-293 cells, we assessed the phosphorylation of WNK4, SPAK, NCC, or NKCC2 in response to forskolin or desmopressin. WNK4 and cotransporter phosphorylation were studied in desmopressin-infused WNK4-/- mice and in tubule suspensions. In HEK-293 cells, only wild-type WNK4 but not WNK1, WNK3, or a WNK4 mutant lacking PKA phosphorylation motifs could upregulate SPAK or cotransporter phosphorylation in response to forskolin or desmopressin. I1 transfection maximized SPAK phosphorylation in response to forskolin in the presence of WNK4 but not of mutant WNK4 lacking PP1 regulation. We observed direct PP1 regulation of NKCC2 dephosphorylation but not of NCC or SPAK in the absence of WNK4. WNK4-/- mice with desmopressin treatment did not increase SPAK/OSR1, NCC, or NKCC2 phosphorylation. In stimulated tubule suspensions from WNK4-/- mice, upregulation of pNKCC2 was reduced, whereas upregulation of SPAK phosphorylation was absent. These findings suggest that WNK4 is a central node in which kinase and phosphatase signaling converge to connect cAMP signaling to the SPAK/OSR1-NCC/NKCC2 pathway.NEW & NOTEWORTHY With-no-lysine kinases regulate the phosphorylation and activity of the Na+-Cl- and Na+-K+-2Cl- cotransporters. This pathway is modulated by arginine vasopressin (AVP). However, the link between AVP and WNK signaling remains unknown. Here, we show that AVP activates WNK4 through increased phosphorylation at putative protein kinase A-regulated sites and decreases its dephosphorylation by protein phosphatase 1. This work increases our understanding of the signaling pathways mediating AVP actions in the kidney.
Collapse
Affiliation(s)
- Hector Carbajal-Contreras
- Department of Nephrology and Mineral Metabolism, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Mexico City, Mexico
- PECEM, Facultad de Medicina, Universidad Nacional Autónoma de México, Mexico City, Mexico
| | - Adrian Rafael Murillo-de-Ozores
- Department of Nephrology and Mineral Metabolism, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Mexico City, Mexico
- Facultad de Medicina, Universidad Nacional Autónoma de México, Mexico City, Mexico
| | - German Magaña-Avila
- Department of Nephrology and Mineral Metabolism, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Mexico City, Mexico
- Facultad de Medicina, Universidad Nacional Autónoma de México, Mexico City, Mexico
| | - Alejandro Marquez-Salinas
- Department of Nephrology and Mineral Metabolism, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Mexico City, Mexico
- PECEM, Facultad de Medicina, Universidad Nacional Autónoma de México, Mexico City, Mexico
| | - Laurent Bourqui
- Institute of Anatomy, University of Zurich, Zurich, Switzerland
| | - Michelle Tellez-Sutterlin
- Department of Nephrology and Mineral Metabolism, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Mexico City, Mexico
- Molecular Physiology Unit, Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México, Mexico City, Mexico
| | - Jessica P Bahena-Lopez
- Division of Nephrology and Hypertension, Department of Medicine, Oregon Health and Science University, Portland, Oregon, United States
- Oregon Clinical and Translational Research Institute, Oregon Health and Science University, Portland, Oregon, United States
| | - Eduardo Cortes-Arroyo
- Department of Nephrology and Mineral Metabolism, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Mexico City, Mexico
| | - Sebastián González Behn-Eschenburg
- Department of Nephrology and Mineral Metabolism, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Mexico City, Mexico
- Molecular Physiology Unit, Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México, Mexico City, Mexico
| | - Alejandro Lopez-Saavedra
- Unidad de Aplicaciones Avanzadas en Microscopía del Instituto Nacional de Cancerología y la Red de Apoyo a la Investigación, Universidad Nacional Autónoma de México, Mexico City, Mexico
| | - Norma Vazquez
- Molecular Physiology Unit, Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México, Mexico City, Mexico
| | - David H Ellison
- Division of Nephrology and Hypertension, Department of Medicine, Oregon Health and Science University, Portland, Oregon, United States
- Oregon Clinical and Translational Research Institute, Oregon Health and Science University, Portland, Oregon, United States
- Veterans Affairs Portland Health Care System, Portland, Oregon, United States
| | | | - Gerardo Gamba
- Department of Nephrology and Mineral Metabolism, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Mexico City, Mexico
- PECEM, Facultad de Medicina, Universidad Nacional Autónoma de México, Mexico City, Mexico
- Molecular Physiology Unit, Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México, Mexico City, Mexico
| | - Maria Castañeda-Bueno
- Department of Nephrology and Mineral Metabolism, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Mexico City, Mexico
| |
Collapse
|
14
|
Maeoka Y, Nguyen LT, Sharma A, Cornelius RJ, Su XT, Gutierrez MR, Carbajal-Contreras H, Castañeda-Bueno M, Gamba G, McCormick JA. Dysregulation of the WNK4-SPAK/OSR1 pathway has a minor effect on baseline NKCC2 phosphorylation. Am J Physiol Renal Physiol 2024; 326:F39-F56. [PMID: 37881876 DOI: 10.1152/ajprenal.00100.2023] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2023] [Revised: 10/12/2023] [Accepted: 10/13/2023] [Indexed: 10/27/2023] Open
Abstract
The with-no-lysine kinase 4 (WNK4)-sterile 20/SPS-1-related proline/alanine-rich kinase (SPAK)/oxidative stress-responsive kinase 1 (OSR1) pathway mediates activating phosphorylation of the furosemide-sensitive Na+-K+-2Cl- cotransporter (NKCC2) and the thiazide-sensitive NaCl cotransporter (NCC). The commonly used pT96/pT101-pNKCC2 antibody cross-reacts with pT53-NCC in mice on the C57BL/6 background due to a five amino acid deletion. We generated a new C57BL/6-specific pNKCC2 antibody (anti-pT96-NKCC2) and tested the hypothesis that the WNK4-SPAK/OSR1 pathway strongly regulates the phosphorylation of NCC but not NKCC2. In C57BL/6 mice, anti-pT96-NKCC2 detected pNKCC2 and did not cross-react with NCC. Abundances of pT96-NKCC2 and pT53-NCC were evaluated in Wnk4-/-, Osr1-/-, Spak-/-, and Osr1-/-/Spak-/- mice and in several models of the disease familial hyperkalemic hypertension (FHHt) in which the CUL3-KLHL3 ubiquitin ligase complex that promotes WNK4 degradation is dysregulated (Cul3+/-/Δ9, Klhl3-/-, and Klhl3R528H/R528H). All mice were on the C57BL/6 background. In Wnk4-/- mice, pT53-NCC was almost absent but pT96-NKCC2 was only slightly lower. pT53-NCC was almost absent in Spak-/- and Osr1-/-/Spak-/- mice, but pT96-NKCC2 abundance did not differ from controls. pT96-NKCC2/total NKCC2 was slightly lower in Osr1-/- and Osr1-/-/Spak-/- mice. WNK4 expression colocalized not only with NCC but also with NKCC2 in Klhl3-/- mice, but pT96-NKCC2 abundance was unchanged. Consistent with this, furosemide-induced urinary Na+ excretion following thiazide treatment was similar between Klhl3-/- and controls. pT96-NKCC2 abundance was also unchanged in the other FHHt mouse models. Our data show that disruption of the WNK4-SPAK/OSR1 pathway only mildly affects NKCC2 phosphorylation, suggesting a role for other kinases in NKCC2 activation. In FHHt models NKCC2 phosphorylation is unchanged despite higher WNK4 abundance, explaining the thiazide sensitivity of FHHt.NEW & NOTEWORTHY The renal cation cotransporters NCC and NKCC2 are activated following phosphorylation mediated by the WNK4-SPAK/OSR1 pathway. While disruption of this pathway strongly affects NCC activity, effects on NKCC2 activity are unclear since the commonly used phospho-NKCC2 antibody was recently reported to cross-react with phospho-NCC in mice on the C57BL/6 background. Using a new phospho-NKCC2 antibody specific for C57BL/6, we show that inhibition or activation of the WNK4-SPAK/OSR1 pathway in mice only mildly affects NKCC2 phosphorylation.
Collapse
Affiliation(s)
- Yujiro Maeoka
- Division of Nephrology and Hypertension, Department of Medicine, Oregon Health and Science University, Portland, Oregon, United States
| | - Luan T Nguyen
- Division of Nephrology and Hypertension, Department of Medicine, Oregon Health and Science University, Portland, Oregon, United States
| | - Avika Sharma
- Division of Nephrology and Hypertension, Department of Medicine, Oregon Health and Science University, Portland, Oregon, United States
| | - Ryan J Cornelius
- Division of Nephrology and Hypertension, Department of Medicine, Oregon Health and Science University, Portland, Oregon, United States
| | - Xiao-Tong Su
- Division of Nephrology and Hypertension, Department of Medicine, Oregon Health and Science University, Portland, Oregon, United States
| | - Marissa R Gutierrez
- Division of Nephrology and Hypertension, Department of Medicine, Oregon Health and Science University, Portland, Oregon, United States
| | - Héctor Carbajal-Contreras
- Department of Nephrology and Mineral Metabolism, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Mexico City, Mexico
| | - María Castañeda-Bueno
- Department of Nephrology and Mineral Metabolism, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Mexico City, Mexico
| | - Gerardo Gamba
- Department of Nephrology and Mineral Metabolism, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Mexico City, Mexico
- Molecular Physiology Unit, Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México, Mexico City, Mexico
| | - James A McCormick
- Division of Nephrology and Hypertension, Department of Medicine, Oregon Health and Science University, Portland, Oregon, United States
| |
Collapse
|
15
|
Zhang S, Meor Azlan NF, Josiah SS, Zhou J, Zhou X, Jie L, Zhang Y, Dai C, Liang D, Li P, Li Z, Wang Z, Wang Y, Ding K, Wang Y, Zhang J. The role of SLC12A family of cation-chloride cotransporters and drug discovery methodologies. J Pharm Anal 2023; 13:1471-1495. [PMID: 38223443 PMCID: PMC10785268 DOI: 10.1016/j.jpha.2023.09.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2023] [Revised: 06/20/2023] [Accepted: 09/05/2023] [Indexed: 09/12/2023] Open
Abstract
The solute carrier family 12 (SLC12) of cation-chloride cotransporters (CCCs) comprises potassium chloride cotransporters (KCCs, e.g. KCC1, KCC2, KCC3, and KCC4)-mediated Cl- extrusion, and sodium potassium chloride cotransporters (N[K]CCs, NKCC1, NKCC2, and NCC)-mediated Cl- loading. The CCCs play vital roles in cell volume regulation and ion homeostasis. Gain-of-function or loss-of-function of these ion transporters can cause diseases in many tissues. In recent years, there have been considerable advances in our understanding of CCCs' control mechanisms in cell volume regulations, with many techniques developed in studying the functions and activities of CCCs. Classic approaches to directly measure CCC activity involve assays that measure the transport of potassium substitutes through the CCCs. These techniques include the ammonium pulse technique, radioactive or nonradioactive rubidium ion uptake-assay, and thallium ion-uptake assay. CCCs' activity can also be indirectly observed by measuring γ-aminobutyric acid (GABA) activity with patch-clamp electrophysiology and intracellular chloride concentration with sensitive microelectrodes, radiotracer 36Cl-, and fluorescent dyes. Other techniques include directly looking at kinase regulatory sites phosphorylation, flame photometry, 22Na+ uptake assay, structural biology, molecular modeling, and high-throughput drug screening. This review summarizes the role of CCCs in genetic disorders and cell volume regulation, current methods applied in studying CCCs biology, and compounds developed that directly or indirectly target the CCCs for disease treatments.
Collapse
Affiliation(s)
- Shiyao Zhang
- Xiamen Cardiovascular Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, Fujian, 363001, China
| | - Nur Farah Meor Azlan
- Institute of Biomedical and Clinical Sciences, Medical School, Faculty of Health and Life Sciences, University of Exeter, Exeter, EX4 4PS, UK
| | - Sunday Solomon Josiah
- Institute of Biomedical and Clinical Sciences, Medical School, Faculty of Health and Life Sciences, University of Exeter, Exeter, EX4 4PS, UK
| | - Jing Zhou
- Department of Neurology, Institutes of Brain Science, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institute of Biological Science, Zhongshan Hospital, Fudan University, Shanghai, 200032, China
| | - Xiaoxia Zhou
- Xiamen Cardiovascular Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, Fujian, 363001, China
| | - Lingjun Jie
- Xiamen Cardiovascular Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, Fujian, 363001, China
| | - Yanhui Zhang
- Xiamen Cardiovascular Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, Fujian, 363001, China
| | - Cuilian Dai
- Xiamen Cardiovascular Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, Fujian, 363001, China
| | - Dong Liang
- Aurora Discovery Inc., Foshan, Guangdong, 528300, China
| | - Peifeng Li
- Institute for Translational Medicine, Qingdao University, Qingdao, Shandong, 266021, China
| | - Zhengqiu Li
- School of Pharmacy, Jinan University, Guangzhou, 510632, China
| | - Zhen Wang
- State Key Laboratory of Chemical Biology, Research Center of Chemical Kinomics, Shanghai Institute of Organic Chemistry, Chinese Academy of Sciences, Shanghai, 200032, China
| | - Yun Wang
- Department of Neurology, Institutes of Brain Science, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institute of Biological Science, Zhongshan Hospital, Fudan University, Shanghai, 200032, China
| | - Ke Ding
- State Key Laboratory of Chemical Biology, Research Center of Chemical Kinomics, Shanghai Institute of Organic Chemistry, Chinese Academy of Sciences, Shanghai, 200032, China
| | - Yan Wang
- Xiamen Cardiovascular Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, Fujian, 363001, China
| | - Jinwei Zhang
- Xiamen Cardiovascular Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, Fujian, 363001, China
- Institute of Biomedical and Clinical Sciences, Medical School, Faculty of Health and Life Sciences, University of Exeter, Exeter, EX4 4PS, UK
- State Key Laboratory of Chemical Biology, Research Center of Chemical Kinomics, Shanghai Institute of Organic Chemistry, Chinese Academy of Sciences, Shanghai, 200032, China
| |
Collapse
|
16
|
Abstract
The with no lysine (K) (WNK) kinases are an evolutionarily ancient group of kinases with atypical placement of the catalytic lysine and diverse physiological roles. Recent studies have shown that WNKs are directly regulated by chloride, potassium, and osmotic pressure. Here, we review the discovery of WNKs as chloride-sensitive kinases and discuss physiological contexts in which chloride regulation of WNKs has been demonstrated. These include the kidney, pancreatic duct, neurons, and inflammatory cells. We discuss the interdependent relationship of osmotic pressure and intracellular chloride in cell volume regulation. We review the recent demonstration of potassium regulation of WNKs and speculate on possible physiological roles. Finally, structural and mechanistic aspects of intracellular ion and osmotic pressure regulation of WNKs are discussed.
Collapse
Affiliation(s)
- Elizabeth J Goldsmith
- Department of Biophysics, University of Texas Southwestern Medical Center, Dallas, Texas, USA
| | - Aylin R Rodan
- Molecular Medicine Program, University of Utah, Salt Lake City, Utah, USA; .,Division of Nephrology and Hypertension, Department of Internal Medicine, University of Utah, Salt Lake City, Utah, USA.,Department of Human Genetics, University of Utah, Salt Lake City, Utah, USA.,Medical Service, Veterans Affairs Salt Lake City Healthcare System, Salt Lake City, Utah, USA
| |
Collapse
|
17
|
The Post-Translational Modification Networking in WNK-Centric Hypertension Regulation and Electrolyte Homeostasis. Biomedicines 2022; 10:biomedicines10092169. [PMID: 36140271 PMCID: PMC9496095 DOI: 10.3390/biomedicines10092169] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2022] [Revised: 08/26/2022] [Accepted: 08/27/2022] [Indexed: 11/17/2022] Open
Abstract
The with-no-lysine (WNK) kinase family, comprising four serine-threonine protein kinases (WNK1-4), were first linked to hypertension due to their mutations in association with pseudohypoaldosteronism type II (PHAII). WNK kinases regulate crucial blood pressure regulators, SPAK/OSR1, to mediate the post-translational modifications (PTMs) of their downstream ion channel substrates, such as sodium chloride co-transporter (NCC), epithelial sodium chloride (ENaC), renal outer medullary potassium channel (ROMK), and Na/K/2Cl co-transporters (NKCCs). In this review, we summarize the molecular pathways dysregulating the WNKs and their downstream target renal ion transporters. We summarize each of the genetic variants of WNK kinases and the small molecule inhibitors that have been discovered to regulate blood pressure via WNK-triggered PTM cascades.
Collapse
|
18
|
WNK1 in the kidney. Curr Opin Nephrol Hypertens 2022; 31:471-478. [PMID: 35894282 DOI: 10.1097/mnh.0000000000000820] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
PURPOSE OF REVIEW The aim of this manuscript was to review recent evidence uncovering the roles of the With No lysine (K) kinase 1 (WNK1) in the kidney. RECENT FINDINGS Analyses of microdissected mouse nephron segments have revealed the abundance of long-WNK1 and kidney-specific-WNK1 transcripts in different segments. The low levels of L-WNK1 transcripts in the distal convoluted tubule (DCT) stand out and support functional evidence on the lack of L-WNK1 activity in this segment. The recent description of familial hyperkalaemic hypertension (FHHt)-causative mutations affecting the acidic domain of WNK1 supports the notion that KS-WNK1 activates the Na+:Cl- cotransporter NCC. The high sensitivity of KS-WNK1 to KLHL3-targeted degradation and the low levels of L-WNK1 in the DCT, led to propose that this type of FHHt is mainly due to increased KS-WNK1 protein in the DCT. The observation that KS-WNK1 renal protein expression is induced by low K+ diet and recent reassessment of the phenotype of KS-WNK1-/- mice suggested that KS-WNK1 may be necessary to achieve maximal NCC activation under this condition. Evidences on the regulation of other renal transport proteins by WNK1 are also summarized. SUMMARY The diversity of WNK1 transcripts in the kidney has complicated the interpretation of experimental data. Integration of experimental data with the knowledge of isoform abundance in renal cell types is necessary in future studies about WNK1 function in the kidney.
Collapse
|
19
|
Rodan AR. Regulation of Distal Nephron Transport by Intracellular Chloride and Potassium. Nephron Clin Pract 2022; 147:203-211. [PMID: 35977527 PMCID: PMC9935751 DOI: 10.1159/000526051] [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: 04/23/2022] [Accepted: 07/11/2022] [Indexed: 11/19/2022] Open
Abstract
BACKGROUND Low potassium increases the phosphorylation and activity of the sodium chloride cotransporter (NCC) in the distal convoluted tubule of the nephron, which contributes to the hypertensive effect of the modern low potassium/high sodium diet. A central mediator of potassium regulation of NCC is the chloride-sensitive With No Lysine [K] (WNK) kinase. SUMMARY Chloride directly inhibits WNKs by binding to the active site. The mechanisms underlying WNK regulation by extracellular potassium are reviewed, as well as the modulatory effect of kidney-specific-WNK1. WNK1, but not WNK1 kinase activity, is also required for the aldosterone-independent regulation of the epithelial sodium channel by potassium. Whether intracellular chloride could be involved in this process is discussed. Recent studies demonstrating direct regulation of WNKs by intracellular potassium are also reviewed, and the potential physiological relevance to renal epithelial ion transport is discussed. KEY MESSAGES WNKs are sensors of the intracellular ionic milieu. In the nephron, changes in extracellular ion concentrations, resulting in changes in intracellular ion concentration, regulate WNK activity and downstream transporters and channels to maintain total body ion homeostasis.
Collapse
Affiliation(s)
- Aylin R Rodan
- Molecular Medicine Program, University of Utah, Salt Lake City, Utah, USA
- Division of Nephrology and Hypertension, Department of Internal Medicine, University of Utah, Salt Lake City, Utah, USA
- Department of Human Genetics, University of Utah, Salt Lake City, Utah, USA
- Medical Service, Veterans Affairs Salt Lake City Health Care System, Salt Lake City, Utah, USA
| |
Collapse
|
20
|
STK39 enhances the progression of Cholangiocarcinoma via PI3K/AKT pathway. iScience 2021; 24:103223. [PMID: 34746696 PMCID: PMC8551078 DOI: 10.1016/j.isci.2021.103223] [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: 07/30/2021] [Revised: 09/10/2021] [Accepted: 09/30/2021] [Indexed: 02/07/2023] Open
Abstract
Serine/threonine kinase 39 (STK39) is overexpressed in various tumor tissues and plays an essential role in tumor progression. In this study, we investigated the clinical value, as well as the potential functions and mechanisms of STK39 in cholangiocarcinoma (CCA). The results showed that STK39 was overexpressed in CCA and negatively associated with the prognosis of patients with CCA. Functionally, STK39 knockdown suppressed cell proliferation, migration, and invasion, while STK39 overexpression facilitated tumor aggressiveness. The tumor-promoting effects of STK39 in CCA were also validated by in vivo experiments. Mechanistically, RNA-seq analysis identified that STK39 enhanced the progression of CCA by activating PI3K/AKT signaling pathway. Furthermore, overexpression of STK39 could induce gemcitabine resistance in CCA cells. Moreover, the increased expression of STK39 may be mediated by the dysregulation of miR-26a-5p. In summary, STK39 could be served as a valuable prognostic candidate and a potential therapeutic target of CCA. STK39 was overexpressed in CCA, negatively associated with the prognosis of patients with CCA STK39 knockdown suppressed cell proliferation and invasion. STK39 overexpression facilitated tumor aggressiveness STK39 mediates oncogenic effects on CCA cells by activating the PI3K/AKT signaling pathway STK39 reduces CCA sensitivity to gemcitabine. Increased expression of STK39 may be mediated by dysregulation of miR-26a-5p
Collapse
|
21
|
Saddhe AA, Karle SB, Aftab T, Kumar K. With no lysine kinases: the key regulatory networks and phytohormone cross talk in plant growth, development and stress response. PLANT CELL REPORTS 2021; 40:2097-2109. [PMID: 34110446 DOI: 10.1007/s00299-021-02728-y] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/26/2021] [Accepted: 06/03/2021] [Indexed: 05/25/2023]
Abstract
With No Lysine kinases (WNKs) are a distinct family of Serine/Threonine protein kinase with unique arrangement of catalytic residues in kinase domain. In WNK, an essential catalytic lysine requisite for attaching ATP and phosphorylation reaction is located in subdomain I, instead of subdomain II, which is essentially a typical feature of other Ser/Thr kinases. WNKs are identified in diverse organisms including multicellular and unicellular organisms. Mammalian WNKs are well characterized at structural and functional level, while plant WNKs are not explored much except few recent studies. Plant WNKs role in various physiological processes viz. ion maintenance, osmotic stress, pH homeostasis, circadian rhythms, regulation of flowering time, proliferation and organ development, and abiotic stresses are known, but the mechanisms involved are unclear. Plant WNKs are known to be involved in enhanced drought and salt stress response via ABA-signaling pathway, but the complete signaling cascade is yet to be elucidated. The current review will discuss the interplay between WNKs and growth regulators and their cross talks in plant growth and development. We have also highlighted the link between the stress phytohormones and WNK members in regulating abiotic stress responses in plants. The present review will provide an overall known mechanism on the involvement of WNKs in plant growth and development and abiotic stress response and highlight its role/applications in the development of stress-tolerant plants.
Collapse
Affiliation(s)
- Ankush Ashok Saddhe
- Department of Biological Sciences, Birla Institute of Technology and Science Pilani, K. K. Birla Goa Campus, Goa, 403 726, India
- Institute of Experimental Botany of the Czech Academy of Sciences, 16502, Prague 6, Czech Republic
| | - Suhas Balasaheb Karle
- Department of Biological Sciences, Birla Institute of Technology and Science Pilani, K. K. Birla Goa Campus, Goa, 403 726, India
| | - Tariq Aftab
- Department of Botany, Aligarh Muslim University, Uttar Pradesh, Aligarh, 202 002, India
| | - Kundan Kumar
- Department of Biological Sciences, Birla Institute of Technology and Science Pilani, K. K. Birla Goa Campus, Goa, 403 726, India.
| |
Collapse
|
22
|
Torres-Pinzon DL, Ralph DL, Veiras LC, McDonough AA. Sex-specific adaptations to high-salt diet preserve electrolyte homeostasis with distinct sodium transporter profiles. Am J Physiol Cell Physiol 2021; 321:C897-C909. [PMID: 34613843 PMCID: PMC8616593 DOI: 10.1152/ajpcell.00282.2021] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2021] [Revised: 10/04/2021] [Accepted: 10/04/2021] [Indexed: 02/04/2023]
Abstract
Kidneys continuously filter an enormous amount of sodium and adapt kidney Na+ reabsorption to match Na+ intake to maintain circulatory volume and electrolyte homeostasis. Males (M) respond to high-salt (HS) diet by translocating proximal tubule Na+/H+ exchanger isoform 3 (NHE3) to the base of the microvilli, reducing activated forms of the distal NaCl cotransporter (NCC) and epithelial Na+ channel (ENaC). Males (M) and females (F) on normal-salt (NS) diet present sex-specific profiles of "transporters" (cotransporters, channels, pumps, and claudins) along the nephron, e.g., F exhibit 40% lower NHE3 and 200% higher NCC abundance than M. We tested the hypothesis that adaptations to HS diet along the nephron will, likewise, exhibit sexual dimorphisms. C57BL/6J mice were fed for 15 days with 4% NaCl diet (HS) versus 0.26% NaCl diet (NS). On HS, M and F exhibited normal plasma [Na+] and [K+], similar urine volume, Na+, K+, and osmolal excretion rates normalized to body weight. In F, like M, HS lowered abundance of distal NCC, phosphorylated NCC, and cleaved (activated) forms of ENaC. The adaptations associated with achieving electrolyte homeostasis exhibit sex-dependent and independent mechanisms. Sex differences in baseline "transporters" abundance persist during HS diet, yet the fold changes during HS diet (normalized to NS) are similar along the distal nephron and collecting duct. Sex-dependent differences observed along the proximal tubule during HS show that female kidneys adapt differently from patterns reported in males, yet achieve and maintain fluid and electrolyte homeostasis.
Collapse
Affiliation(s)
- Diana L Torres-Pinzon
- Department of Physiology and Neuroscience, Keck School of Medicine of University of Southern California, Los Angeles, California
| | - Donna L Ralph
- Department of Physiology and Neuroscience, Keck School of Medicine of University of Southern California, Los Angeles, California
| | - Luciana C Veiras
- Department of Physiology and Neuroscience, Keck School of Medicine of University of Southern California, Los Angeles, California
| | - Alicia A McDonough
- Department of Physiology and Neuroscience, Keck School of Medicine of University of Southern California, Los Angeles, California
| |
Collapse
|
23
|
Portioli C, Ruiz Munevar MJ, De Vivo M, Cancedda L. Cation-coupled chloride cotransporters: chemical insights and disease implications. TRENDS IN CHEMISTRY 2021; 3:832-849. [PMID: 34604727 PMCID: PMC8461084 DOI: 10.1016/j.trechm.2021.05.004] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Cation-coupled chloride cotransporters (CCCs) modulate the transport of sodium and/or potassium cations coupled with chloride anions across the cell membrane. CCCs thus help regulate intracellular ionic concentration and consequent cell volume homeostasis. This has been largely exploited in the past to develop diuretic drugs that act on CCCs expressed in the kidney. However, a growing wealth of evidence has demonstrated that CCCs are also critically involved in a great variety of other pathologies, motivating most recent drug discovery programs targeting CCCs. Here, we examine the structure–function relationship of CCCs. By linking recent high-resolution cryogenic electron microscopy (cryo-EM) data with older biochemical/functional studies on CCCs, we discuss the mechanistic insights and opportunities to design selective CCC modulators to treat diverse pathologies. The structural topology and function of all cation-coupled chloride cotransporters (CCCs) have been continuously investigated over the past 40 years, with great progress also thanks to the recent cryogenic electron microscopy (cryo-EM) resolution of the structures of five CCCs. In particular, such studies have clarified the structure–function relationship for the Na-K-Cl cotransporter NKCC1 and K-Cl cotransporters KCC1–4. The constantly growing evidence of the crucial involvement of CCCs in physiological and various pathological conditions, as well as the evidence of their wide expression in diverse body tissues, has promoted CCCs as targets for the discovery and development of new, safer, and more selective/effective drugs for a plethora of pathologies. Post-translational modification anchor points on the structure of CCCs may offer alternative strategies for small molecule drug discovery.
Collapse
Affiliation(s)
- Corinne Portioli
- Brain Development and Disease Laboratory, Istituto Italiano di Tecnologia (IIT), Via Morego 30, 16163 Genoa, Italy.,Laboratory of Molecular Modeling and Drug Discovery, IIT, Via Morego, 30 16163 Genoa, Italy
| | | | - Marco De Vivo
- Laboratory of Molecular Modeling and Drug Discovery, IIT, Via Morego, 30 16163 Genoa, Italy
| | - Laura Cancedda
- Brain Development and Disease Laboratory, Istituto Italiano di Tecnologia (IIT), Via Morego 30, 16163 Genoa, Italy.,Dulbecco Telethon Institute, Via Varese 16b, 00185 Rome, Italy
| |
Collapse
|
24
|
Chen J, Zhou L, Yang J, Xie H, Liu L, Li Y. Knockdown of STK39 suppressed cell proliferation, migration, and invasion in hepatocellular carcinoma by repressing the phosphorylation of mitogen-activated protein kinase p38. Bioengineered 2021; 12:6529-6537. [PMID: 34519635 PMCID: PMC8806584 DOI: 10.1080/21655979.2021.1973876] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022] Open
Abstract
Hepatocellular carcinoma (HCC) is a serious malignant tumor of the liver. It has been reported that serine/threonine kinase 39 (STK39) participates in tumorigenesis. However, the role of STK39 in HCC remains unknown. In this study, the qRT-PCR and western blot assay demonstrated that STK39 expression was enhanced in HCC patients and tissues. Moreover, CCK-8 and colony formation assays confirmed that knockdown of STK39 suppressed SK-HEP-1 and Huh7 cells proliferation. Furthermore, wound healing assay and transwell assay revealed that knockdown of STK39 repressed SK-HEP-1 and Huh7 cells migration and invasion. Interestingly, knockdown of STK39 reduced p-p38/p38 ratio and levels of c-Myc. Consistently, knockdown of STK39 inhibited the HCC tumor growth in vivo. In summary, knockdown of STK39 suppressed the proliferation, migration, and invasion of HCC cells by inducing the lower levels of p-p38, which might provide a novel therapeutic target for HCC.
Collapse
Affiliation(s)
- Jian Chen
- Department of Hepatobiliary and Pancreatic Surgery, People's Hospital of DeYang City, Deyang City, Sichuan Province, China
| | - Luke Zhou
- Department of Hepatobiliary and Pancreatic Surgery, People's Hospital of DeYang City, Deyang City, Sichuan Province, China
| | - Jie Yang
- Department of Hepatobiliary and Pancreatic Surgery, People's Hospital of DeYang City, Deyang City, Sichuan Province, China
| | - Hui Xie
- Department of Hepatobiliary and Pancreatic Surgery, People's Hospital of DeYang City, Deyang City, Sichuan Province, China
| | - Lin Liu
- Department of Hepatobiliary and Pancreatic Surgery, People's Hospital of DeYang City, Deyang City, Sichuan Province, China
| | - Youwei Li
- Department of Hepatobiliary and Pancreatic Surgery, People's Hospital of DeYang City, Deyang City, Sichuan Province, China
| |
Collapse
|
25
|
Moser S, Sugano Y, Wengi A, Fisi V, Lindtoft Rosenbaek L, Mariniello M, Loffing‐Cueni D, McCormick JA, Fenton RA, Loffing J. A five amino acids deletion in NKCC2 of C57BL/6 mice affects analysis of NKCC2 phosphorylation but does not impact kidney function. Acta Physiol (Oxf) 2021; 233:e13705. [PMID: 34114742 PMCID: PMC8384713 DOI: 10.1111/apha.13705] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2020] [Revised: 05/04/2021] [Accepted: 06/07/2021] [Indexed: 12/24/2022]
Abstract
Aim The phosphorylation level of the furosemide‐sensitive Na+‐K+‐2Cl− cotransporter (NKCC2) in the thick ascending limb (TAL) is used as a surrogate marker for NKCC2 activation and TAL function. However, in mice, analyses of NKCC2 phosphorylation with antibodies against phosphorylated threonines 96 and 101 (anti‐pT96/pT101) give inconsistent results. We aimed (a) to elucidate these inconsistencies and (b) to develop a phosphoform‐specific antibody that ensures reliable detection of NKCC2 phosphorylation in mice. Methods Genetic information, molecular biology, biochemical techniques and mouse phenotyping was used to study NKCC2 and kidney function in two commonly used mouse strains (ie 129Sv and in C57BL/6 mice). Moreover, a new phosphoform‐specific mouse NKCC2 antibody was developed and characterized. Results Amino acids sequence alignment revealed that C57BL/6 mice have a strain‐specific five amino acids deletion (ΔF97‐T101) in NKCC2 that diminishes the detection of NKCC2 phosphorylation with previously developed pT96/pT101 NKCC2 antibodies. Instead, the antibodies cross‐react with the phosphorylated thiazide‐sensitive NaCl cotransporter (NCC), which can obscure interpretation of results. Interestingly, the deletion in NKCC2 does not impact on kidney function and/or expression of renal ion transport proteins as indicated by the analysis of the F2 generation of crossbred 129Sv and C57BL/6 mice. A newly developed pT96 NKCC2 antibody detects pNKCC2 in both mouse strains and shows no cross‐reactivity with phosphorylated NCC. Conclusion Our work reveals a hitherto unappreciated, but essential, strain difference in the amino acids sequence of mouse NKCC2 that needs to be considered when analysing NKCC2 phosphorylation in mice. The new pNKCC2 antibody circumvents this technical caveat.
Collapse
Affiliation(s)
- Sandra Moser
- Institute of Anatomy University of Zurich Zurich Switzerland
| | - Yuya Sugano
- Institute of Anatomy University of Zurich Zurich Switzerland
| | - Agnieszka Wengi
- Institute of Anatomy University of Zurich Zurich Switzerland
| | - Viktoria Fisi
- Institute of Anatomy University of Zurich Zurich Switzerland
| | | | | | | | - James A. McCormick
- Division of Nephrology and Hypertension Oregon Health & Science University Portland OR USA
| | | | - Johannes Loffing
- Institute of Anatomy University of Zurich Zurich Switzerland
- Swiss National Centre for Competence in Research “Kidney control of homeostasis” Zurich Switzerland
| |
Collapse
|
26
|
Jonniya NA, Zhang J, Kar P. Molecular Mechanism of Inhibiting WNK Binding to OSR1 by Targeting the Allosteric Pocket of the OSR1-CCT Domain with Potential Antihypertensive Inhibitors: An In Silico Study. J Phys Chem B 2021; 125:9115-9129. [PMID: 34369793 DOI: 10.1021/acs.jpcb.1c04672] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
The oxidative-stress-responsive kinase 1 (OSR1) and the STE20/SPS1-related proline-alanine-rich kinase (SPAK) are physiological substrates of the with-no-lysine (WNK) kinase. They are the master regulators of cation Cl- cotransporters that could be targeted for discovering novel antihypertensive agents. Both kinases have a conserved carboxy-terminal (CCT) domain that recognizes a unique peptide motif (Arg-Phe-Xaa-Val) present in their upstream kinases and downstream substrates. Here, we have combined molecular docking with molecular dynamics simulations and free-energy calculations to identify potential inhibitors that can bind to the allosteric pocket of the OSR1-CCT domain and impede its interaction with the WNK peptide. Our study revealed that STOCK1S-14279 and Closantel bound strongly to the allosteric pocket of OSR1 and displaced the WNK peptide from the primary pocket of OSR1. We showed that primarily Arg1004 and Gln1006 of the WNK4-peptide motif were involved in strong H-bond interactions with Glu453 and Arg451 of OSR1. Besides, our study revealed that atoms of Arg1004 were solvent-exposed in cases of STOCK1S-14279 and Closantel, implying that the WNK4 peptide was moved out of the pocket. Overall, the predicted potential inhibitors altogether abolish the OSR1-WNK4-peptide interaction, suggesting their potency as a prospective allosteric inhibitor against OSR1.
Collapse
Affiliation(s)
- Nisha Amarnath Jonniya
- Department of Biosciences and Biomedical Engineering, Indian Institute of Technology Indore, Khandwa Road, Indore, Madhya Pradesh 453552, India
| | - Jinwei Zhang
- Institute of Biomedical and Clinical Sciences, College of Medicine and Health, University of Exeter Medical School, Hatherly Laboratories, Prince of Wales Road, Exeter EX4 4PS, U.K
| | - Parimal Kar
- Department of Biosciences and Biomedical Engineering, Indian Institute of Technology Indore, Khandwa Road, Indore, Madhya Pradesh 453552, India
| |
Collapse
|
27
|
Taylor CA, Cobb MH. CCT and CCT-like Modular Protein Interaction Domains in WNK Signaling. Mol Pharmacol 2021; 101:201-212. [PMID: 34312216 DOI: 10.1124/molpharm.121.000307] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2021] [Accepted: 07/14/2021] [Indexed: 11/22/2022] Open
Abstract
The WNK (with-no lysine (K)) kinases and their downstream effector kinases, OSR1 (oxidative stress responsive 1) and SPAK (SPS/STE20-related proline-alanine rich kinase), have well-established functions in the maintenance of cell volume and ion homeostasis. Mutations in these kinases have been linked to an inherited form of hypertension, neurological defects, and other pathologies. A rapidly expanding body of evidence points to the involvement of WNKs in regulating multiple diverse cellular processes as well as the progression of some forms of cancer. How OSR1/SPAK contribute to these processes is well understood in some cases, but completely unknown in others. OSR1 and SPAK are targeted to both WNKs and substrates via their conserved C-terminal (CCT) protein interaction domains. Considerable effort has been put forth to understand the structure, function, and interaction specificity of the CCT domains in relation to WNK signaling, and multiple inhibitors of WNK signaling target these domains. The domains bind RFxV and RxFxV protein sequence motifs with the consensus sequence R-F-x-V/I or R-x-F-x-V/I, but residues outside the core motif also contribute to specificity. CCT interactions are required for OSR1 and SPAK activation and deactivation as well as cation-chloride cotransporter substrate phosphorylation. All four WNKs also contain CCT-like domains that have similar structures and conserved binding residues when compared to CCT domains, but their functions and interaction specificities are mostly unknown. A better understanding of the varied actions of these domains and their interactions will better define the known signaling mechanisms of the WNK pathway as well as uncover new ones. Significance Statement WNK kinases and downstream effector kinases, OSR1 and SPAK, have been shown to be involved in an array of diverse cellular processes. Here we review the function of modular protein interaction domains found in OSR1 and SPAK as well as related domains found in WNKs.
Collapse
Affiliation(s)
- Clinton A Taylor
- Pharmacology, University of Texas Southwestern Medical Center, United States
| | - Melanie H Cobb
- Pharmacology, University of Texas Southwestern Medical Center, United States
| |
Collapse
|
28
|
Marcoux A, Tremblay LE, Slimani S, Fiola M, Mac‐Way F, Garneau AP, Isenring P. Molecular characteristics and physiological roles of Na + -K + -Cl - cotransporter 2. J Cell Physiol 2021; 236:1712-1729. [PMID: 32776569 PMCID: PMC7818487 DOI: 10.1002/jcp.29997] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2020] [Revised: 06/28/2020] [Accepted: 07/24/2020] [Indexed: 12/23/2022]
Abstract
Na+ -K+ -Cl- cotransporter 2 (NKCC2; SLC12A1) is an integral membrane protein that comes as three splice variants and mediates the cotranslocation of Na+ , K+ , and Cl- ions through the apical membrane of the thick ascending loop of Henle (TALH). In doing so, and through the involvement of other ion transport systems, it allows this nephron segment to reclaim a large fraction of the ultrafiltered Na+ , Cl- , Ca2+ , Mg2+ , and HCO3- loads. The functional relevance of NKCC2 in human is illustrated by the many abnormalities that result from the inactivation of this transport system through the use of loop diuretics or in the setting of inherited disorders. The following presentation aims at discussing the physiological roles and molecular characteristics of Na+ -K+ -Cl- cotransport in the TALH and those of the individual NKCC2 splice variants more specifically. Many of the historical and recent data that have emerged from the experiments conducted will be outlined and their larger meaning will also be placed into perspective with the aid of various hypotheses.
Collapse
Affiliation(s)
- Andree‐Anne Marcoux
- Department of Medicine, Nephrology Research GroupLaval UniversityQuebec CityQuébecCanada
| | - Laurence E. Tremblay
- Department of Medicine, Nephrology Research GroupLaval UniversityQuebec CityQuébecCanada
| | - Samira Slimani
- Department of Medicine, Nephrology Research GroupLaval UniversityQuebec CityQuébecCanada
| | - Marie‐Jeanne Fiola
- Department of Medicine, Nephrology Research GroupLaval UniversityQuebec CityQuébecCanada
| | - Fabrice Mac‐Way
- Department of Medicine, Nephrology Research GroupLaval UniversityQuebec CityQuébecCanada
| | - Alexandre P. Garneau
- Department of Medicine, Nephrology Research GroupLaval UniversityQuebec CityQuébecCanada
- Cardiometabolic Axis, School of Kinesiology and Physical Activity SciencesUniversity of MontréalMontréalQuebecCanada
| | - Paul Isenring
- Department of Medicine, Nephrology Research GroupLaval UniversityQuebec CityQuébecCanada
| |
Collapse
|
29
|
Murillo-de-Ozores AR, Rodríguez-Gama A, Carbajal-Contreras H, Gamba G, Castañeda-Bueno M. WNK4 kinase: from structure to physiology. Am J Physiol Renal Physiol 2021; 320:F378-F403. [PMID: 33491560 DOI: 10.1152/ajprenal.00634.2020] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
With no lysine kinase-4 (WNK4) belongs to a serine-threonine kinase family characterized by the atypical positioning of its catalytic lysine. Despite the fact that WNK4 has been found in many tissues, the majority of its study has revolved around its function in the kidney, specifically as a positive regulator of the thiazide-sensitive NaCl cotransporter (NCC) in the distal convoluted tubule of the nephron. This is explained by the description of gain-of-function mutations in the gene encoding WNK4 that causes familial hyperkalemic hypertension. This disease is mainly driven by increased downstream activation of the Ste20/SPS1-related proline-alanine-rich kinase/oxidative stress responsive kinase-1-NCC pathway, which increases salt reabsorption in the distal convoluted tubule and indirectly impairs renal K+ secretion. Here, we review the large volume of information that has accumulated about different aspects of WNK4 function. We first review the knowledge on WNK4 structure and enumerate the functional domains and motifs that have been characterized. Then, we discuss WNK4 physiological functions based on the information obtained from in vitro studies and from a diverse set of genetically modified mouse models with altered WNK4 function. We then review in vitro and in vivo evidence on the different levels of regulation of WNK4. Finally, we go through the evidence that has suggested how different physiological conditions act through WNK4 to modulate NCC activity.
Collapse
Affiliation(s)
- Adrián Rafael Murillo-de-Ozores
- Department of Nephrology and Mineral Metabolism, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Tlalpan, Mexico City, Mexico.,Facultad de Medicina, Universidad Nacional Autónoma de México, Coyoacan, Mexico City, Mexico
| | | | - Héctor Carbajal-Contreras
- Department of Nephrology and Mineral Metabolism, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Tlalpan, Mexico City, Mexico.,Combined Studies Program in Medicine MD/PhD (PECEM), Facultad de Medicina, Universidad Nacional Autónoma de México, Coyoacan, Mexico City, Mexico, Mexico
| | - Gerardo Gamba
- Department of Nephrology and Mineral Metabolism, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Tlalpan, Mexico City, Mexico.,Molecular Physiology Unit, Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México, Tlalpan, Mexico City, Mexico.,Combined Studies Program in Medicine MD/PhD (PECEM), Facultad de Medicina, Universidad Nacional Autónoma de México, Coyoacan, Mexico City, Mexico, Mexico
| | - María Castañeda-Bueno
- Department of Nephrology and Mineral Metabolism, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Tlalpan, Mexico City, Mexico.,Combined Studies Program in Medicine MD/PhD (PECEM), Facultad de Medicina, Universidad Nacional Autónoma de México, Coyoacan, Mexico City, Mexico, Mexico
| |
Collapse
|
30
|
Chan CH, Wu SN, Bao BY, Li HW, Lu TL. MST3 Involvement in Na + and K + Homeostasis with Increasing Dietary Potassium Intake. Int J Mol Sci 2021; 22:ijms22030999. [PMID: 33498219 PMCID: PMC7863938 DOI: 10.3390/ijms22030999] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2020] [Revised: 01/14/2021] [Accepted: 01/15/2021] [Indexed: 02/07/2023] Open
Abstract
K+ loading inhibits NKCC2 (Na-K-Cl cotransporter) and NCC (Na-Cl cotransporter) in the early distal tubules, resulting in Na+ delivery to the late distal convoluted tubules (DCTs). In the DCTs, Na+ entry through ENaC (epithelial Na channel) drives K+ secretion through ROMK (renal outer medullary potassium channel). WNK4 (with-no-lysine 4) regulates the NCC/NKCC2 through SAPK (Ste20-related proline-alanine-rich kinase)/OSR1 (oxidative stress responsive). K+ loading increases intracellular Cl−, which binds to the WNK4, thereby inhibiting autophosphorylation and downstream signals. Acute K+ loading-deactivated NCC was not observed in Cl−-insensitive WNK4 mice, indicating that WNK4 was involved in K+ loading-inhibited NCC activity. However, chronic K+ loading deactivated NCC in Cl−-insensitive WNK4 mice, indicating that other mechanisms may be involved. We previously reported that mammalian Ste20-like protein kinase 3 (MST3/STK24) was expressed mainly in the medullary TAL (thick ascending tubule) and at lower levels in the DCTs. MST3−/− mice exhibited higher ENaC activity, causing hypernatremia and hypertension. To investigate MST3 function in maintaining Na+/K+ homeostasis in kidneys, mice were fed diets containing various concentrations of Na+ and K+. The 2% KCl diets induced less MST3 expression in MST3−/− mice than that in wild-type (WT) mice. The MST3−/− mice had higher WNK4, NKCC2-S130 phosphorylation, and ENaC expression, resulting in lower urinary Na+ and K+ excretion than those of WT mice. Lower urinary Na+ excretion was associated with elevated plasma [Na+] and hypertension. These results suggest that MST3 maintains Na+/K+ homeostasis in response to K+ loading by regulation of WNK4 expression and NKCC2 and ENaC activity.
Collapse
Affiliation(s)
- Chee-Hong Chan
- Department of Nephrology, Chang Bing Show Chwan Memorial Hospital, Lukang, Changhua 505, Taiwan;
| | - Sheng-Nan Wu
- Department of Physiology, National Cheng Kung University Medical College, Tainan 70101, Taiwan;
| | - Bo-Ying Bao
- School of Pharmacy, China Medical University, Taichung 406040, Taiwan;
- Department of Nursing, Asia University, Taichung 41354, Taiwan
| | - Houng-Wei Li
- Department of Medical Laboratory Science and Biotechnology, China Medical University, Taichung 406040, Taiwan;
| | - Te-Ling Lu
- School of Pharmacy, China Medical University, Taichung 406040, Taiwan;
- Correspondence:
| |
Collapse
|
31
|
Jonniya NA, Sk MF, Kar P. Characterizing an allosteric inhibitor-induced inactive state in with-no-lysine kinase 1 using Gaussian accelerated molecular dynamics simulations. Phys Chem Chem Phys 2021; 23:7343-7358. [DOI: 10.1039/d0cp05733a] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
The binding of an allosteric inhibitor in WNK1 leads to the inactive state.
Collapse
Affiliation(s)
- Nisha Amarnath Jonniya
- Department of Biosciences and Biomedical Engineering, Indian Institute of Technology Indore, Khandwa Road, MP
- India
| | - Md Fulbabu Sk
- Department of Biosciences and Biomedical Engineering, Indian Institute of Technology Indore, Khandwa Road, MP
- India
| | - Parimal Kar
- Department of Biosciences and Biomedical Engineering, Indian Institute of Technology Indore, Khandwa Road, MP
- India
| |
Collapse
|
32
|
Zhang C, Wang X, Fang D, Xu P, Mo X, Hu C, Abdelatty A, Wang M, Xu H, Sun Q, Zhou G, She J, Xia J, Hui KM, Xia H. STK39 is a novel kinase contributing to the progression of hepatocellular carcinoma by the PLK1/ERK signaling pathway. Theranostics 2021; 11:2108-2122. [PMID: 33500714 PMCID: PMC7797677 DOI: 10.7150/thno.48112] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2020] [Accepted: 10/26/2020] [Indexed: 02/06/2023] Open
Abstract
Rationale: Protein kinases are critical therapeutic targets for curing hepatocellular carcinoma (HCC). As a serine/threonine kinase, the potential roles of serine/threonine kinase 39 (STK39) in HCC remain to be explored. Methods: The expression of STK39 was examined by RT-qPCR, western blotting and immunohistochemistry. Cell proliferation and apoptosis were detected by CCK8 and TUNEL kit. Cell migration and invasion assays were performed using a transwell system with or without Matrigel. RNA-seq, mass spectrometry and luciferase reporter assays were used to identify STK39 binding proteins. Results: Here, we firstly report that STK39 was highly overexpressed in clinical HCC tissues compared with adjacent tissues, high expression of STK39 was induced by transcription factor SP1 and correlated with poor patient survival. Gain and loss of function assays revealed that overexpression of STK39 promoted HCC cell proliferation, migration and invasion. In contrast, the depletion of STK39 attenuated the growth and metastasis of HCC cells. Moreover, knockdown of STK39 induced the HCC cell cycle arrested in the G2/M phase and promoted apoptosis. In mechanistic studies, RNA-seq revealed that STK39 positively regulated the ERK signaling pathway. Mass spectrometry identified that STK39 bound to PLK1 and STK39 promoted HCC progression and activated ERK signaling pathway dependent on PLK1. Conclusions: Thus, our study uncovers a novel role of STK39/PLK1/ERK signaling axis in the progress of HCC and suggests STK39 as an indicator for prognosis and a potential drug target of HCC.
Collapse
Affiliation(s)
- Chengfei Zhang
- Department of Pathology, School of Basic Medical Sciences & Sir Run Run Hospital & State Key Laboratory of Reproductive Medicine & Key Laboratory of Antibody Technique of National Health Commission, Nanjing Medical University, Nanjing 211166, Jiangsu, China
- Jiangsu Cancer Hospital, The Affiliated Cancer Hospital of Nanjing Medical University, Jiangsu Institute of Cancer Research, Nanjing 210009, Jiangsu, China
| | - Xiaoming Wang
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Wannan Medical College, Wuhu 241001, Anhui, China
| | - Dan Fang
- Department of Pathology, School of Basic Medical Sciences & Sir Run Run Hospital & State Key Laboratory of Reproductive Medicine & Key Laboratory of Antibody Technique of National Health Commission, Nanjing Medical University, Nanjing 211166, Jiangsu, China
| | - Ping Xu
- Department of Pathology, School of Basic Medical Sciences & Sir Run Run Hospital & State Key Laboratory of Reproductive Medicine & Key Laboratory of Antibody Technique of National Health Commission, Nanjing Medical University, Nanjing 211166, Jiangsu, China
| | - Xiao Mo
- Department of Pathology, School of Basic Medical Sciences & Sir Run Run Hospital & State Key Laboratory of Reproductive Medicine & Key Laboratory of Antibody Technique of National Health Commission, Nanjing Medical University, Nanjing 211166, Jiangsu, China
| | - Chao Hu
- Department of Pathology, School of Basic Medical Sciences & Sir Run Run Hospital & State Key Laboratory of Reproductive Medicine & Key Laboratory of Antibody Technique of National Health Commission, Nanjing Medical University, Nanjing 211166, Jiangsu, China
| | - Alaa Abdelatty
- Department of Pathology, School of Basic Medical Sciences & Sir Run Run Hospital & State Key Laboratory of Reproductive Medicine & Key Laboratory of Antibody Technique of National Health Commission, Nanjing Medical University, Nanjing 211166, Jiangsu, China
| | - Mei Wang
- Department of Pathology, School of Basic Medical Sciences & Sir Run Run Hospital & State Key Laboratory of Reproductive Medicine & Key Laboratory of Antibody Technique of National Health Commission, Nanjing Medical University, Nanjing 211166, Jiangsu, China
| | - Haojun Xu
- Department of Pathology, School of Basic Medical Sciences & Sir Run Run Hospital & State Key Laboratory of Reproductive Medicine & Key Laboratory of Antibody Technique of National Health Commission, Nanjing Medical University, Nanjing 211166, Jiangsu, China
| | - Qi Sun
- Department of Pathology, School of Basic Medical Sciences & Sir Run Run Hospital & State Key Laboratory of Reproductive Medicine & Key Laboratory of Antibody Technique of National Health Commission, Nanjing Medical University, Nanjing 211166, Jiangsu, China
| | - Guoren Zhou
- Jiangsu Cancer Hospital, The Affiliated Cancer Hospital of Nanjing Medical University, Jiangsu Institute of Cancer Research, Nanjing 210009, Jiangsu, China
| | - Junjun She
- Department of High Talent & General Surgery & Med-X Institute, The First Affiliated Hospital of Xi'an Jiao Tong University, Xi'an, 710061, Shaanxi, China
| | - Jinglin Xia
- The First Affiliated Hospital of Wenzhou Medical University, Wenzhou 325000, Zhejiang, China
| | - Kam Man Hui
- Laboratory of Cancer Genomics, National Cancer Centre Singapore & Cancer and Stem Cell Biology Program, Duke-NUS Medical School, Singapore
| | - Hongping Xia
- Department of Pathology, School of Basic Medical Sciences & Sir Run Run Hospital & State Key Laboratory of Reproductive Medicine & Key Laboratory of Antibody Technique of National Health Commission, Nanjing Medical University, Nanjing 211166, Jiangsu, China
- Jiangsu Cancer Hospital, The Affiliated Cancer Hospital of Nanjing Medical University, Jiangsu Institute of Cancer Research, Nanjing 210009, Jiangsu, China
- Department of High Talent & General Surgery & Med-X Institute, The First Affiliated Hospital of Xi'an Jiao Tong University, Xi'an, 710061, Shaanxi, China
- Laboratory of Cancer Genomics, National Cancer Centre Singapore & Cancer and Stem Cell Biology Program, Duke-NUS Medical School, Singapore
| |
Collapse
|
33
|
Louis-Dit-Picard H, Kouranti I, Rafael C, Loisel-Ferreira I, Chavez-Canales M, Abdel-Khalek W, Argaiz ER, Baron S, Vacle S, Migeon T, Coleman R, Do Cruzeiro M, Hureaux M, Thurairajasingam N, Decramer S, Girerd X, O’Shaugnessy K, Mulatero P, Roussey G, Tack I, Unwin R, Vargas-Poussou R, Staub O, Grimm R, Welling PA, Gamba G, Clauser E, Hadchouel J, Jeunemaitre X. Mutation affecting the conserved acidic WNK1 motif causes inherited hyperkalemic hyperchloremic acidosis. J Clin Invest 2020; 130:6379-6394. [PMID: 32790646 PMCID: PMC7685730 DOI: 10.1172/jci94171] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2018] [Accepted: 08/11/2020] [Indexed: 01/01/2023] Open
Abstract
Gain-of-function mutations in with no lysine (K) 1 (WNK1) and WNK4 genes are responsible for familial hyperkalemic hypertension (FHHt), a rare, inherited disorder characterized by arterial hypertension and hyperkalemia with metabolic acidosis. More recently, FHHt-causing mutations in the Kelch-like 3-Cullin 3 (KLHL3-CUL3) E3 ubiquitin ligase complex have shed light on the importance of WNK's cellular degradation on renal ion transport. Using full exome sequencing for a 4-generation family and then targeted sequencing in other suspected cases, we have identified new missense variants in the WNK1 gene clustering in the short conserved acidic motif known to interact with the KLHL3-CUL3 ubiquitin complex. Affected subjects had an early onset of a hyperkalemic hyperchloremic phenotype, but normal blood pressure values"Functional experiments in Xenopus laevis oocytes and HEK293T cells demonstrated that these mutations strongly decrease the ubiquitination of the kidney-specific isoform KS-WNK1 by the KLHL3-CUL3 complex rather than the long ubiquitous catalytically active L-WNK1 isoform. A corresponding CRISPR/Cas9 engineered mouse model recapitulated both the clinical and biological phenotypes. Renal investigations showed increased activation of the Ste20 proline alanine-rich kinase-Na+-Cl- cotransporter (SPAK-NCC) phosphorylation cascade, associated with impaired ROMK apical expression in the distal part of the renal tubule. Together, these new WNK1 genetic variants highlight the importance of the KS-WNK1 isoform abundance on potassium homeostasis.
Collapse
Affiliation(s)
| | | | - Chloé Rafael
- Université de Paris, INSERM, PARCC, F-75006, Paris, France
- INSERM UMR_S1155, Tenon Hospital, Paris, France
- Université Paris-Diderot, Sorbonne Paris Cité, Paris, France
| | | | - Maria Chavez-Canales
- Université de Paris, INSERM, PARCC, F-75006, Paris, France
- Translational Medicine Unit, Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México and Instituto Nacional de Cardiología Ignacio Chávez, Tlalpan, Mexico City, Mexico
| | | | - Eduardo R. Argaiz
- Molecular Physiology Unit, Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México, and Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubiran, Mexico City, Mexico
| | - Stéphanie Baron
- Université de Paris, INSERM, PARCC, F-75006, Paris, France
- Service d’Explorations Fonctionnelles, Assistance Publique–Hôpitaux de Paris (AP-HP), F-75015, Paris, France
| | - Sarah Vacle
- Department of Pharmacology and Toxicology, University of Lausanne, Lausanne, Switzerland
| | | | - Richard Coleman
- Department of Physiology, University of Maryland School of Medicine, Baltimore, Maryland, USA
| | | | - Marguerite Hureaux
- Université de Paris, INSERM, PARCC, F-75006, Paris, France
- AP-HP, Département de Génétique, Hôpital Européen Georges Pompidou, Paris, France
| | | | - Stéphane Decramer
- Service de Néphrologie Pédiatrique, Hôpital des Enfants, Toulouse, France
| | - Xavier Girerd
- AP-HP, Institute of Cardiometabolism and Nutrition (ICAN), Unité de Prévention Cardiovasculaire, Hôpital de La Pitié-Salpêtrière, Paris, France
| | - Kevin O’Shaugnessy
- Department of Medicine, University of Cambridge, Cambridge, United Kingdom
| | - Paolo Mulatero
- Division of Internal Medicine and Hypertension Unit, Department of Medical Sciences, University of Torino, Torino, Italy
| | - Gwenaëlle Roussey
- Néphrologie Pédiatrique–Clinique Médicale Pédiatrique, Hôpital Mère Enfant, CHU de Nantes, Nantes, France
| | - Ivan Tack
- Service des Explorations Fonctionnelles Physiologiques, CHU de Toulouse et INSERM U1048-I2MC, Toulouse, France
| | - Robert Unwin
- UCL Department of Renal Medicine, University College London, Royal Free Campus and Hospital, London, United Kingdom
| | - Rosa Vargas-Poussou
- AP-HP, Département de Génétique, Hôpital Européen Georges Pompidou, Paris, France
| | - Olivier Staub
- Department of Pharmacology and Toxicology, University of Lausanne, Lausanne, Switzerland
| | - Richard Grimm
- Departments of Medicine, Nephrology, and Physiology, Johns Hopkins University Medical School, Baltimore, Maryland, USA
| | - Paul A. Welling
- Departments of Medicine, Nephrology, and Physiology, Johns Hopkins University Medical School, Baltimore, Maryland, USA
| | - Gerardo Gamba
- Molecular Physiology Unit, Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México, and Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubiran, Mexico City, Mexico
| | - Eric Clauser
- Université de Paris, INSERM, PARCC, F-75006, Paris, France
| | - Juliette Hadchouel
- Université de Paris, INSERM, PARCC, F-75006, Paris, France
- INSERM UMR_S1155, Tenon Hospital, Paris, France
- Université Paris-Diderot, Sorbonne Paris Cité, Paris, France
| | - Xavier Jeunemaitre
- Université de Paris, INSERM, PARCC, F-75006, Paris, France
- AP-HP, Département de Génétique, Hôpital Européen Georges Pompidou, Paris, France
| |
Collapse
|
34
|
Elzwawi A, Grafton G, Barnes NM, Mehellou Y. SPAK and OSR1 kinases bind and phosphorylate the β 2-Adrenergic receptor. Biochem Biophys Res Commun 2020; 532:88-93. [PMID: 32828531 DOI: 10.1016/j.bbrc.2020.07.143] [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] [Received: 07/27/2020] [Accepted: 07/31/2020] [Indexed: 12/20/2022]
Abstract
SPAK and OSR1 are two cytoplasmic serine/threonine protein kinases that regulate the function of a series of sodium, potassium and chloride co-transporters via phosphorylation. Over recent years, it has emerged that these two kinases may have diverse function beyond the regulation of ion co-transporters. Inspired by this, we explored whether SPAK and OSR1 kinases impact physically and phosphorylate the β2-adrenergic receptor (β2ADR). Herein, we report that the amino acid sequence of the human β2ADR displays a SPAK/OSR1 consensus binding motif and using a series of pulldown and in vitro kinase assays we show that SPAK and OSR1 bind the β2ADR and phosphorylate it in vitro. This work provides a notable example of SPAK and OSR1 kinases binding to a G-protein coupled receptor and taps into the potential of these protein kinases in regulating membrane receptors beyond ion co-transporters.
Collapse
Affiliation(s)
- Abdulrahman Elzwawi
- School of Pharmacy, College of Medical and Dental Sciences, University of Birmingham, Edgbaston, Birmingham, B15 2TT, UK
| | - Gillian Grafton
- School of Pharmacy, College of Medical and Dental Sciences, University of Birmingham, Edgbaston, Birmingham, B15 2TT, UK
| | - Nicholas M Barnes
- School of Pharmacy, College of Medical and Dental Sciences, University of Birmingham, Edgbaston, Birmingham, B15 2TT, UK
| | - Youcef Mehellou
- School of Pharmacy and Pharmaceutical Sciences, Cardiff University, King Edward VII Avenue, Cardiff, CF10 3NB, UK.
| |
Collapse
|
35
|
Fu Y, Yuan PP, Cao YG, Ke YY, Zhang Q, Hou Y, Zhang YL, Feng WS, Zheng XK. Geniposide in Gardenia jasminoides var. radicans Makino modulates blood pressure via inhibiting WNK pathway mediated by the estrogen receptors. J Pharm Pharmacol 2020; 72:1956-1969. [DOI: 10.1111/jphp.13361] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2020] [Accepted: 07/25/2020] [Indexed: 12/15/2022]
Abstract
Abstract
Objectives
To investigate the effects of geniposide in an iridoid found in Gardenia jasminoides var. radicans Makino (GJRM) in spontaneous hypertensive rat (SHR) and explore the possible mechanisms.
Methods
In this study, we detected the content of geniposide in GJRM by high-performance liquid chromatography (HPLC). Then, we used acute diuretic experiments to determine whether geniposide has diuretic effect. Moreover, we carried out experiments on SHR to further study the mechanism of hypertension, while real-time PCR, Western blot and immunohistochemistry were used for the experiments in vivo test. Hypotonic model was used for in vitro test.
Key findings
Our data showed that the content of geniposide in the extract of GJRM is 27.54%. Meanwhile, 50 mg/kg geniposide showed the strongest effect on promoting urine volume. Further study indicated that the extract of GJRM and geniposide could significantly reduce blood pressure and promote the excretion of urine and Na+ in SHR. In addition, geniposide significantly inhibited the activation of the with-no-lysine kinase (WNK) signalling pathway and significantly increases the protein expressions of estrogen receptor α (ERα), estrogen receptor β (ERβ) and G protein-coupled receptor 30 (GPR30) in SHR. In hypotonic model, geniposide significantly inhibits the phosphorylation of NKCC and NCC and could be antagonistic to estrogen receptor antagonists.
Conclusions
Collectively, we would suggest that geniposide may potentially be utilized as an adjunct to existing thiazide and thiazide-like diuretics to control hypertension, mainly through inhibiting the activation of the WNK signalling pathway mediated by the estrogen receptor.
Collapse
Affiliation(s)
- Yang Fu
- Henan University of Chinese Medicine, Zhengzhou, Henan, China
- The Engineering and Technology Center for Chinese Medicine Development of Henan Province, Zhengzhou, Henan, China
| | - Pei-pei Yuan
- Henan University of Chinese Medicine, Zhengzhou, Henan, China
- The Engineering and Technology Center for Chinese Medicine Development of Henan Province, Zhengzhou, Henan, China
| | - Yan-gang Cao
- Henan University of Chinese Medicine, Zhengzhou, Henan, China
- The Engineering and Technology Center for Chinese Medicine Development of Henan Province, Zhengzhou, Henan, China
| | - Ying-ying Ke
- Henan University of Chinese Medicine, Zhengzhou, Henan, China
- The Engineering and Technology Center for Chinese Medicine Development of Henan Province, Zhengzhou, Henan, China
| | - Qi Zhang
- Henan University of Chinese Medicine, Zhengzhou, Henan, China
- The Engineering and Technology Center for Chinese Medicine Development of Henan Province, Zhengzhou, Henan, China
| | - Ying Hou
- Henan University of Chinese Medicine, Zhengzhou, Henan, China
- The Engineering and Technology Center for Chinese Medicine Development of Henan Province, Zhengzhou, Henan, China
| | - Yan-li Zhang
- Henan University of Chinese Medicine, Zhengzhou, Henan, China
- The Engineering and Technology Center for Chinese Medicine Development of Henan Province, Zhengzhou, Henan, China
| | - Wei-sheng Feng
- Henan University of Chinese Medicine, Zhengzhou, Henan, China
- The Engineering and Technology Center for Chinese Medicine Development of Henan Province, Zhengzhou, Henan, China
| | - Xiao-ke Zheng
- Henan University of Chinese Medicine, Zhengzhou, Henan, China
- The Engineering and Technology Center for Chinese Medicine Development of Henan Province, Zhengzhou, Henan, China
| |
Collapse
|
36
|
Shin DH, Kim M, Kim Y, Jun I, Jung J, Nam JH, Cheng MH, Lee MG. Bicarbonate permeation through anion channels: its role in health and disease. Pflugers Arch 2020; 472:1003-1018. [PMID: 32621085 DOI: 10.1007/s00424-020-02425-x] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2020] [Revised: 06/19/2020] [Accepted: 06/26/2020] [Indexed: 12/31/2022]
Abstract
Many anion channels, frequently referred as Cl- channels, are permeable to different anions in addition to Cl-. As the second-most abundant anion in the human body, HCO3- permeation via anion channels has many important physiological roles. In addition to its classical role as an intracellular pH regulator, HCO3- also controls the activity and stability of dissolved proteins in bodily fluids such as saliva, pancreatic juice, intestinal fluid, and airway surface liquid. Moreover, HCO3- permeation through these channels affects membrane potentials that are the driving forces for transmembrane transport of solutes and water in epithelia and affect neuronal excitability in nervous tissue. Consequently, aberrant HCO3- transport via anion channels causes a number of human diseases in respiratory, gastrointestinal, genitourinary, and neuronal systems. Notably, recent studies have shown that the HCO3- permeabilities of several anion channels are not fixed and can be altered by cellular stimuli, findings which may have both physiological and pathophysiological significance. In this review, we summarize recent progress in understanding the molecular mechanisms and the physiological roles of HCO3- permeation through anion channels. We hope that the present discussions can stimulate further research into this very important topic, which will provide the basis for human disorders associated with aberrant HCO3- transport.
Collapse
Affiliation(s)
- Dong Hoon Shin
- Department of Pharmacology, Brain Korea 21 PLUS Project for Medical Sciences, Severance Biomedical Science Institute, Yonsei University College of Medicine, Seoul, 03722, South Korea
| | - Minjae Kim
- Department of Pharmacology, Brain Korea 21 PLUS Project for Medical Sciences, Severance Biomedical Science Institute, Yonsei University College of Medicine, Seoul, 03722, South Korea
| | - Yonjung Kim
- Department of Pharmacology, Brain Korea 21 PLUS Project for Medical Sciences, Severance Biomedical Science Institute, Yonsei University College of Medicine, Seoul, 03722, South Korea
| | - Ikhyun Jun
- Department of Pharmacology, Brain Korea 21 PLUS Project for Medical Sciences, Severance Biomedical Science Institute, Yonsei University College of Medicine, Seoul, 03722, South Korea
- The Institute of Vision Research, Department of Ophthalmology, Yonsei University College of Medicine, Seoul, 03722, South Korea
| | - Jinsei Jung
- Department of Pharmacology, Brain Korea 21 PLUS Project for Medical Sciences, Severance Biomedical Science Institute, Yonsei University College of Medicine, Seoul, 03722, South Korea
- Department of Otorhinolaryngology, Yonsei University College of Medicine, Seoul, 03722, South Korea
| | - Joo Hyun Nam
- Department of Physiology, Dongguk University College of Medicine, 123 Dongdae-ro, Kyungju, 780-714, Republic of Korea
| | - Mary Hongying Cheng
- Department of Computational & Systems Biology, University of Pittsburgh, Pittsburgh, PA, USA
| | - Min Goo Lee
- Department of Pharmacology, Brain Korea 21 PLUS Project for Medical Sciences, Severance Biomedical Science Institute, Yonsei University College of Medicine, Seoul, 03722, South Korea.
| |
Collapse
|
37
|
Abstract
PURPOSE OF REVIEW Studies of the genetic model organism, Drosophila melanogaster, have unraveled molecular pathways relevant to human physiology and disease. The Malpighian tubule, the Drosophila renal epithelium, is described here, including tools available to study transport; conserved transporters, channels, and the signaling pathways regulating them; and fly models of kidney stone disease. RECENT FINDINGS Tools to measure Malpighian tubule transport continue to advance, including use of a transgenic sensor to quantify intracellular pH and proton fluxes. A recent study generated an RNA-sequencing-based atlas of tissue-specific gene expression, with resulting insights into Malpighian tubule gene expression of transporters and channels. Advances have been made in understanding the molecular physiology of the With No Lysine kinase-Ste20-related proline/alanine rich kinase/oxidative stress response kinase cascade that regulates epithelial ion transport in flies and mammals. New studies in Drosophila kidney stone models have characterized zinc transporters and used Malpighian tubules to study the efficacy of a plant metabolite in decreasing stone burden. SUMMARY Study of the Drosophila Malpighian tubule affords opportunities to better characterize the molecular physiology of epithelial transport mechanisms relevant to mammalian renal physiology.
Collapse
|
38
|
Abstract
PURPOSE OF REVIEW The apical Na/K/2Cl cotransporter (NKCC2) mediates NaCl reabsorption by the thick ascending limb, contributing to maintenance of blood pressure (BP). Despite effective NKCC2 inhibition by loop diuretics, these agents are not viable for long-term management of BP due to side effects. Novel molecular mechanisms that control NKCC2 activity reveal an increasingly complex picture with interacting layers of NKCC2 regulation. Here, we review the latest developments that shine new light on NKCC2-mediated control of BP and potential new long-term therapies to treat hypertension. RECENT FINDINGS Emerging molecular NKCC2 regulators, often binding partners, reveal a complex overlay of interacting mechanisms aimed at fine tuning NKCC2 activity. Different factors achieve this by shifting the balance between trafficking steps like exocytosis, endocytosis, recycling and protein turnover, or by balancing phosphorylation vs. dephosphorylation. Further molecular details are also emerging on previously known pathways of NKCC2 regulation, and recent in-vivo data continues to place NKCC2 regulation at the center of BP control. SUMMARY Several layers of emerging molecular mechanisms that control NKCC2 activity may operate simultaneously, but they can also be controlled independently. This provides an opportunity to identify new pharmacological targets to fine-tune NKCC2 activity for BP management.
Collapse
|
39
|
Fu Y, Yuan P, Ke Y, Cao Y, Zhang Q, Hou Y, Wei Y, Gao L, Zheng X, feng W. Regulation of PI3k-WNK Pathway by Ethyl Acetate Partition Fraction of Gardenia jasminoides var. radicans Makino in SHR, NRK52e Cells, and IMCD3 Cells. Nat Prod Commun 2020. [DOI: 10.1177/1934578x20920989] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Abstract
The antihypertensive mechanism was studied of the ethyl acetate fraction of the ethanol extract (EAPF) of Gardenia jasminoides var. radicans Makino (GJRM). GJRM is a fake product of Gardenia jasminoides Ellis ( G. jasminoides), but in China’s Henan province, the production of GJRM is much more than G. jasminoides’s, but its traditional usage is as a dye. Gardenia jasminoides can be used to reduce blood pressure and blood glucose levels. The chemical compositions of GJRM and G. jasminoides are similar, and we previously confirmed that GJRM can also reduce blood pressure. Here, we report that the EAPF of GJRM could activate the phosphoinositide 3-kinases (PI3K) pathway in the kidneys of spontaneously hypertensive rats, thus increasing the content of nitric oxide and bradykinin in sera and decreasing endothelin-1 content. EAPF can also decrease the levels of with-no-lysine kinase 1 (WNK1) expression, WNK4 and oxidative stress-responsive kinase 1 messenger ribonucleic acid (mRNA), and Na-K-2Cl cotransporter and sodium chloride cotransporters mRNA and phosphorylation. To investigate the antihypertensive effects of the EAPF of GJRM, 5 monoterpenoids isolated from EAPF were studied for their effects on NRK52e and IMCD3 cells. These compounds inhibited the PI3K-WNK signaling pathway to varying degrees under hypotonic conditions; 4-methoxyrehmapicrogenin had the best effect.
Collapse
Affiliation(s)
- Yang Fu
- College of Pharmacy, Henan University of Chinese Medicine, Zhengzhou, China
| | - Peipei Yuan
- College of Pharmacy, Henan University of Chinese Medicine, Zhengzhou, China
| | - Yingying Ke
- College of Pharmacy, Henan University of Chinese Medicine, Zhengzhou, China
| | - Yangang Cao
- College of Pharmacy, Henan University of Chinese Medicine, Zhengzhou, China
| | - Qi Zhang
- College of Pharmacy, Henan University of Chinese Medicine, Zhengzhou, China
| | - Ying Hou
- College of Pharmacy, Henan University of Chinese Medicine, Zhengzhou, China
| | - Yaxin Wei
- College of Pharmacy, Henan University of Chinese Medicine, Zhengzhou, China
| | - Liyuan Gao
- College of Pharmacy, Henan University of Chinese Medicine, Zhengzhou, China
| | - Xiaoke Zheng
- College of Pharmacy, Henan University of Chinese Medicine, Zhengzhou, China
- Collaborative Innovation Center for Respiratory Disease Diagnosis and Treatment & Chinese Medicine Development of Henan Province, Zhengzhou, China
| | - Weisheng feng
- College of Pharmacy, Henan University of Chinese Medicine, Zhengzhou, China
- Collaborative Innovation Center for Respiratory Disease Diagnosis and Treatment & Chinese Medicine Development of Henan Province, Zhengzhou, China
| |
Collapse
|
40
|
Zhang J, Bhuiyan MIH, Zhang T, Karimy JK, Wu Z, Fiesler VM, Zhang J, Huang H, Hasan MN, Skrzypiec AE, Mucha M, Duran D, Huang W, Pawlak R, Foley LM, Hitchens TK, Minnigh MB, Poloyac SM, Alper SL, Molyneaux BJ, Trevelyan AJ, Kahle KT, Sun D, Deng X. Modulation of brain cation-Cl - cotransport via the SPAK kinase inhibitor ZT-1a. Nat Commun 2020; 11:78. [PMID: 31911626 PMCID: PMC6946680 DOI: 10.1038/s41467-019-13851-6] [Citation(s) in RCA: 68] [Impact Index Per Article: 13.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2018] [Accepted: 11/27/2019] [Indexed: 02/08/2023] Open
Abstract
The SLC12A cation-Cl- cotransporters (CCC), including NKCC1 and the KCCs, are important determinants of brain ionic homeostasis. SPAK kinase (STK39) is the CCC master regulator, which stimulates NKCC1 ionic influx and inhibits KCC-mediated efflux via phosphorylation at conserved, shared motifs. Upregulation of SPAK-dependent CCC phosphorylation has been implicated in several neurological diseases. Using a scaffold-hybrid strategy, we develop a novel potent and selective SPAK inhibitor, 5-chloro-N-(5-chloro-4-((4-chlorophenyl)(cyano)methyl)-2-methylphenyl)-2-hydroxybenzamide ("ZT-1a"). ZT-1a inhibits NKCC1 and stimulates KCCs by decreasing their SPAK-dependent phosphorylation. Intracerebroventricular delivery of ZT-1a decreases inflammation-induced CCC phosphorylation in the choroid plexus and reduces cerebrospinal fluid (CSF) hypersecretion in a model of post-hemorrhagic hydrocephalus. Systemically administered ZT-1a reduces ischemia-induced CCC phosphorylation, attenuates cerebral edema, protects against brain damage, and improves outcomes in a model of stroke. These results suggest ZT-1a or related compounds may be effective CCC modulators with therapeutic potential for brain disorders associated with impaired ionic homeostasis.
Collapse
Affiliation(s)
- Jinwei Zhang
- Institute of Biomedical and Clinical Sciences, Medical School, College of Medicine and Health, University of Exeter, Hatherly Laboratories, Exeter, EX4 4PS, UK.
- Xiamen Cardiovascular Hospital, School of Medicine, Xiamen University, Xiamen, Fujian, 361004, China.
| | - Mohammad Iqbal H Bhuiyan
- Department of Neurology and Pittsburgh Institute For Neurodegenerative Diseases, University of Pittsburgh, Pittsburgh, PA, 15213, USA
| | - Ting Zhang
- State Key Laboratory of Cellular Stress Biology, Innovation Center for Cell Signaling Network, School of Life Sciences, Xiamen University, Xiamen, Fujian, 361102, China
| | - Jason K Karimy
- Departments of Neurosurgery, Pediatrics, and Cellular & Molecular Physiology; Interdepartmental Neuroscience Program; and Centers for Mendelian Genomics, Yale School of Medicine, New Haven, CT, 06511, USA
| | - Zhijuan Wu
- Newcastle University Business School, Newcastle University, Newcastle upon Tyne, NE1 4SE, UK
| | - Victoria M Fiesler
- Department of Neurology and Pittsburgh Institute For Neurodegenerative Diseases, University of Pittsburgh, Pittsburgh, PA, 15213, USA
| | - Jingfang Zhang
- State Key Laboratory of Cellular Stress Biology, Innovation Center for Cell Signaling Network, School of Life Sciences, Xiamen University, Xiamen, Fujian, 361102, China
| | - Huachen Huang
- Department of Neurology and Pittsburgh Institute For Neurodegenerative Diseases, University of Pittsburgh, Pittsburgh, PA, 15213, USA
| | - Md Nabiul Hasan
- Department of Neurology and Pittsburgh Institute For Neurodegenerative Diseases, University of Pittsburgh, Pittsburgh, PA, 15213, USA
| | - Anna E Skrzypiec
- Institute of Biomedical and Clinical Sciences, Medical School, College of Medicine and Health, University of Exeter, Hatherly Laboratories, Exeter, EX4 4PS, UK
| | - Mariusz Mucha
- Institute of Biomedical and Clinical Sciences, Medical School, College of Medicine and Health, University of Exeter, Hatherly Laboratories, Exeter, EX4 4PS, UK
| | - Daniel Duran
- Departments of Neurosurgery, Pediatrics, and Cellular & Molecular Physiology; Interdepartmental Neuroscience Program; and Centers for Mendelian Genomics, Yale School of Medicine, New Haven, CT, 06511, USA
| | - Wei Huang
- State Key Laboratory of Cellular Stress Biology, Innovation Center for Cell Signaling Network, School of Life Sciences, Xiamen University, Xiamen, Fujian, 361102, China
| | - Robert Pawlak
- Institute of Biomedical and Clinical Sciences, Medical School, College of Medicine and Health, University of Exeter, Hatherly Laboratories, Exeter, EX4 4PS, UK
| | - Lesley M Foley
- Animal Imaging Center, University of Pittsburgh, Pittsburgh, PA, 15203, USA
| | - T Kevin Hitchens
- Animal Imaging Center, University of Pittsburgh, Pittsburgh, PA, 15203, USA
- Department of Neurobiology, University of Pittsburgh, Pittsburgh, PA, 15213, USA
| | - Margaret B Minnigh
- Department of Pharmaceutical Sciences, School of Pharmacy, University of Pittsburgh, Pittsburgh, PA, 15261, USA
| | - Samuel M Poloyac
- Department of Pharmaceutical Sciences, School of Pharmacy, University of Pittsburgh, Pittsburgh, PA, 15261, USA
| | - Seth L Alper
- Division of Nephrology, Department of Medicine, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, MA, 02215, USA
| | - Bradley J Molyneaux
- Department of Neurology and Pittsburgh Institute For Neurodegenerative Diseases, University of Pittsburgh, Pittsburgh, PA, 15213, USA
- Department of Critical Care Medicine, University of Pittsburgh, Pittsburgh, PA, 15213, USA
| | - Andrew J Trevelyan
- Institute of Neuroscience, Medical School, Newcastle University, Framlington Place, Newcastle upon Tyne, NE2 4HH, UK
| | - Kristopher T Kahle
- Departments of Neurosurgery, Pediatrics, and Cellular & Molecular Physiology; Interdepartmental Neuroscience Program; and Centers for Mendelian Genomics, Yale School of Medicine, New Haven, CT, 06511, USA.
| | - Dandan Sun
- Department of Neurology and Pittsburgh Institute For Neurodegenerative Diseases, University of Pittsburgh, Pittsburgh, PA, 15213, USA.
- Veterans Affairs Pittsburgh Health Care System, Geriatric Research, Educational and Clinical Center, Pittsburgh, PA, 15213, USA.
| | - Xianming Deng
- State Key Laboratory of Cellular Stress Biology, Innovation Center for Cell Signaling Network, School of Life Sciences, Xiamen University, Xiamen, Fujian, 361102, China.
| |
Collapse
|
41
|
Henriques AFA, Matos P, Carvalho AS, Azkargorta M, Elortza F, Matthiesen R, Jordan P. WNK1 phosphorylation sites in TBC1D1 and TBC1D4 modulate cell surface expression of GLUT1. Arch Biochem Biophys 2019; 679:108223. [PMID: 31816312 DOI: 10.1016/j.abb.2019.108223] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2019] [Revised: 11/13/2019] [Accepted: 12/05/2019] [Indexed: 02/06/2023]
Abstract
Glucose uptake by mammalian cells is a key mechanism to maintain cell and tissue homeostasis and relies mostly on plasma membrane-localized glucose transporter proteins (GLUTs). Two main cellular mechanisms regulate GLUT proteins in the cell: first, expression of GLUT genes is under dynamic transcriptional control and is used by cancer cells to increase glucose availability. Second, GLUT proteins are regulated by membrane traffic from storage vesicles to the plasma membrane (PM). This latter process is triggered by signaling mechanisms and well-studied in the case of insulin-responsive cells, which activate protein kinase AKT to phosphorylate TBC1D4, a RAB-GTPase activating protein involved in membrane traffic regulation. Previously, we identified protein kinase WNK1 as another kinase able to phosphorylate TBC1D4 and regulate the surface expression of the constitutive glucose transporter GLUT1. Here we describe that downregulation of WNK1 through RNA interference in HEK293 cells led to a 2-fold decrease in PM GLUT1 expression, concomitant with a 60% decrease in glucose uptake. By mass spectrometry, we identified serine (S) 704 in TBC1D4 as a WNK1-regulated phosphorylation site, and also S565 in the paralogue TBC1D1. Transfection of the respective phosphomimetic or unphosphorylatable TBC1D mutants into cells revealed that both affected the cell surface abundance of GLUT1. The results reinforce a regulatory role for WNK1 in cell metabolism and have potential impact for the understanding of cancer cell metabolism and therapeutic options in type 2 diabetes.
Collapse
Affiliation(s)
- Andreia F A Henriques
- Department of Human Genetics, National Health Institute 'Dr. Ricardo Jorge', Lisbon, Portugal; BioISI - Biosystems & Integrative Sciences Institute, Faculty of Sciences, University of Lisbon, Lisbon, Portugal
| | - Paulo Matos
- Department of Human Genetics, National Health Institute 'Dr. Ricardo Jorge', Lisbon, Portugal; BioISI - Biosystems & Integrative Sciences Institute, Faculty of Sciences, University of Lisbon, Lisbon, Portugal
| | - Ana Sofia Carvalho
- CEDOC-Chronic Diseases Research Centre, Nova Medical School, Faculdade de Ciências Médicas, Universidade Nova de Lisboa, Lisbon, Portugal
| | - Mikel Azkargorta
- Center for Cooperative Research in Biosciences (CIC bioGUNE), Building 800, Science and Technology Park of Bizkaia, 48160, Derio, Spain
| | - Felix Elortza
- Center for Cooperative Research in Biosciences (CIC bioGUNE), Building 800, Science and Technology Park of Bizkaia, 48160, Derio, Spain
| | - Rune Matthiesen
- CEDOC-Chronic Diseases Research Centre, Nova Medical School, Faculdade de Ciências Médicas, Universidade Nova de Lisboa, Lisbon, Portugal
| | - Peter Jordan
- Department of Human Genetics, National Health Institute 'Dr. Ricardo Jorge', Lisbon, Portugal; BioISI - Biosystems & Integrative Sciences Institute, Faculty of Sciences, University of Lisbon, Lisbon, Portugal.
| |
Collapse
|
42
|
Edwards A, McDonough AA. Impact of angiotensin II-mediated stimulation of sodium transporters in the nephron assessed by computational modeling. Am J Physiol Renal Physiol 2019; 317:F1656-F1668. [PMID: 31657247 DOI: 10.1152/ajprenal.00335.2019] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022] Open
Abstract
Angiotensin II (ANG II) raises blood pressure partly by stimulating tubular Na+ reabsorption. The effects of ANG II on tubular Na+ transporters (i.e., channels, pumps, cotransporters, and exchangers) vary between short-term and long-term exposure. To better understand the physiological impact, we used a computational model of transport along the rat nephron to predict the effects of short- and long-term ANG II-induced transporter activation on Na+ and K+ reabsorption/secretion, and to compare measured and calculated excretion rates. Three days of ANG II infusion at 200 ng·kg-1·min-1 is nonpressor, yet stimulates transporter accumulation. The increase in abundance of Na+/H+ exchanger 3 (NHE3) or activated Na+-K+-2Cl- cotransporter-2 (NKCC2-P) predicted significant reductions in urinary Na+ excretion, yet there was no observed change in urine Na+. The lack of antinatriuresis, despite Na+ transporter accumulation, was supported by Li+ and creatinine clearance measurements, leading to the conclusion that 3-day nonpressor ANG II increases transporter abundance without proportional activation. Fourteen days of ANG II infusion at 400 ng·kg-1·min-1 raises blood pressure and increases Na+ transporter abundance along the distal nephron; proximal tubule and medullary loop transporters are decreased and urine Na+ and volume output are increased, evidence for pressure natriuresis. Simulations indicate that decreases in NHE3 and NKCC2-P contribute significantly to reducing Na+ reabsorption along the nephron and to pressure natriuresis. Our results also suggest that differential regulation of medullary (decrease) and cortical (increase) NKCC2-P is important to preserve K+ while minimizing Na+ retention during ANG II infusion. Lastly, our model indicates that accumulation of active Na+-Cl- cotransporter counteracts epithelial Na+ channel-induced urinary K+ loss.
Collapse
Affiliation(s)
- Aurélie Edwards
- Department of Biomedical Engineering, Boston University, Boston, Massachusetts
| | - Alicia A McDonough
- Department of Physiology and Neuroscience, Keck School of Medicine of University of Southern California, Los Angeles, California
| |
Collapse
|
43
|
Kim Y, Jun I, Shin DH, Yoon JG, Piao H, Jung J, Park HW, Cheng MH, Bahar I, Whitcomb DC, Lee MG. Regulation of CFTR Bicarbonate Channel Activity by WNK1: Implications for Pancreatitis and CFTR-Related Disorders. Cell Mol Gastroenterol Hepatol 2019; 9:79-103. [PMID: 31561038 PMCID: PMC6889609 DOI: 10.1016/j.jcmgh.2019.09.003] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/11/2018] [Revised: 09/16/2019] [Accepted: 09/17/2019] [Indexed: 12/28/2022]
Abstract
BACKGRAOUD & AIMS Aberrant epithelial bicarbonate (HCO3-) secretion caused by mutations in the cystic fibrosis transmembrane conductance regulator (CFTR) gene is associated with several diseases including cystic fibrosis and pancreatitis. Dynamically regulated ion channel activity and anion selectivity of CFTR by kinases sensitive to intracellular chloride concentration ([Cl-]i) play an important role in epithelial HCO3- secretion. However, the molecular mechanisms of how [Cl-]i-dependent mechanisms regulate CFTR are unknown. METHODS We examined the mechanisms of the CFTR HCO3- channel regulation by [Cl-]i-sensitive kinases using an integrated electrophysiological, molecular, and computational approach including whole-cell, outside-out, and inside-out patch clamp recordings and molecular dissection of WNK1 and CFTR proteins. In addition, we analyzed the effects of pancreatitis-causing CFTR mutations on the WNK1-mediated regulation of CFTR. RESULTS Among the WNK1, SPAK, and OSR1 kinases that constitute a [Cl-]i-sensitive kinase cascade, the expression of WNK1 alone was sufficient to increase the CFTR bicarbonate permeability (PHCO3/PCl) and conductance (GHCO3) in patch clamp recordings. Molecular dissection of the WNK1 domains revealed that the WNK1 kinase domain is responsible for CFTR PHCO3/PCl regulation by direct association with CFTR, while the surrounding N-terminal regions mediate the [Cl-]i-sensitivity of WNK1. Furthermore, the pancreatitis-causing R74Q and R75Q mutations in the elbow helix 1 of CFTR hampered WNK1-CFTR physical associations and reduced WNK1-mediated CFTR PHCO3/PCl regulation. CONCLUSION The CFTR HCO3- channel activity is regulated by [Cl-]i and a WNK1-dependent mechanism. Our results provide new insights into the regulation of the ion selectivity of CFTR and the pathogenesis of CFTR-related disorders.
Collapse
Affiliation(s)
- Yonjung Kim
- Department of Pharmacology, Brain Korea 21 PLUS Project for Medical Sciences, Severance Biomedical Science Institute, Yonsei University College of Medicine, Seoul, Korea
| | - Ikhyun Jun
- Department of Pharmacology, Brain Korea 21 PLUS Project for Medical Sciences, Severance Biomedical Science Institute, Yonsei University College of Medicine, Seoul, Korea,Institute of Vision Research, Department of Ophthalmology, Yonsei University College of Medicine, Seoul, Korea
| | - Dong Hoon Shin
- Department of Pharmacology, Brain Korea 21 PLUS Project for Medical Sciences, Severance Biomedical Science Institute, Yonsei University College of Medicine, Seoul, Korea
| | - Jihoon G. Yoon
- Department of Pharmacology, Brain Korea 21 PLUS Project for Medical Sciences, Severance Biomedical Science Institute, Yonsei University College of Medicine, Seoul, Korea
| | - He Piao
- Department of Pharmacology, Brain Korea 21 PLUS Project for Medical Sciences, Severance Biomedical Science Institute, Yonsei University College of Medicine, Seoul, Korea
| | - Jinsei Jung
- Department of Pharmacology, Brain Korea 21 PLUS Project for Medical Sciences, Severance Biomedical Science Institute, Yonsei University College of Medicine, Seoul, Korea,Department of Otorhinolaryngology, Yonsei University College of Medicine, Seoul, Korea
| | - Hyun Woo Park
- Department of Biochemistry, College of Life Science and Biotechnology, Yonsei University, Seoul, Korea
| | - Mary Hongying Cheng
- Department of Computational & Systems Biology, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Ivet Bahar
- Department of Computational & Systems Biology, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - David C. Whitcomb
- Division of Gastroenterology, Hepatology and Nutrition, Department of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Min Goo Lee
- Department of Pharmacology, Brain Korea 21 PLUS Project for Medical Sciences, Severance Biomedical Science Institute, Yonsei University College of Medicine, Seoul, Korea,Correspondence Address correspondence to: Min Goo Lee, Department of Pharmacology, Yonsei University College of Medicine, 50-1 Yonsei-ro, Seoul 03722, Korea. fax: +82 2 313 1894.
| |
Collapse
|
44
|
Marcoux AA, Slimani S, Tremblay LE, Frenette-Cotton R, Garneau AP, Isenring P. Endocytic recycling of Na + -K + -Cl - cotransporter type 2: importance of exon 4. J Physiol 2019; 597:4263-4276. [PMID: 31216057 DOI: 10.1113/jp278024] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2019] [Accepted: 06/05/2019] [Indexed: 01/29/2023] Open
Abstract
KEY POINTS Na+ -K+ -Cl- cotransporter type 2 (NKCC2) is a 27-exon membrane protein that is expressed in the thick ascending limb (TAL) of Henle where it is involved in reabsorption of the ultrafiltered NaCl load. It comes as three splice variants that are identical to each other except for the residue composition of exon 4 and that differ in their transport characteristics, functional roles and distributions along the TAL. In this report, it is shown that the variants also differ in their trafficking properties and that two residues in exon 4 play a key role in this regard. One of these residues was also shown to sustain carrier internalization. Through these results, a novel function for the alternatively spliced exon of NKCC2 has been identified and a domain that is involved in carrier trafficking has been uncovered for the first time in a cation-Cl- cotransporter family member. ABSTRACT Na+ -K+ -Cl- cotransporter type 2 (NKCC2) is a 12-transmembrane (TM) domain cell surface glycoprotein that is expressed in the thick ascending limb (TAL) of Henle and stimulated during cell shrinkage. It comes as three splice variants (A, B and F) that are identical to each other except for TM2 and the following connecting segment (CS2). Yet, these variants do not share the same localization, transport characteristics and physiological roles along the TAL. We have recently found that while cell shrinkage could exert its activating effect by increasing NKCC2 expression at the cell surface, the variants also responded differentially to this stimulus. In the current work, a mutagenic approach was exploited to determine whether CS2 could play a role in carrier trafficking and identify the residues potentially involved. We found that when the residue of position 238 in NKCC2A (F) and NKCC2B (Y) was replaced by the corresponding residue in NKCC2F (V), carrier activity increased by over 3-fold and endocytosis decreased concomitantly. We also found that when the residue of position 230 in NKCC2F (M) was replaced by the one in NKCC2B (T), carrier activity and affinity for ions both increased substantially whereas expression at the membrane decreased. Taken together, these results suggest that CS2 is involved in carrier trafficking and that two of its residues, those of positions 238 and 230, are part of an internalization motif. They also indicate that the divergent residue of position 230 plays the dual role of specifying ion affinity and sustaining carrier internalization.
Collapse
Affiliation(s)
- Andrée-Anne Marcoux
- Nephrology Research Group, L'Hôtel-Dieu de Québec Research Center, Department of Medicine, Faculty of Medicine, Laval University, Québec, QC, Canada, G1R 2J6
| | - Samira Slimani
- Nephrology Research Group, L'Hôtel-Dieu de Québec Research Center, Department of Medicine, Faculty of Medicine, Laval University, Québec, QC, Canada, G1R 2J6
| | - Laurence E Tremblay
- Nephrology Research Group, L'Hôtel-Dieu de Québec Research Center, Department of Medicine, Faculty of Medicine, Laval University, Québec, QC, Canada, G1R 2J6
| | - Rachelle Frenette-Cotton
- Nephrology Research Group, L'Hôtel-Dieu de Québec Research Center, Department of Medicine, Faculty of Medicine, Laval University, Québec, QC, Canada, G1R 2J6
| | - Alexandre P Garneau
- Nephrology Research Group, L'Hôtel-Dieu de Québec Research Center, Department of Medicine, Faculty of Medicine, Laval University, Québec, QC, Canada, G1R 2J6.,Cardiometabolic Research Group, Department of Kinesiology, Faculty of Medicine, University of Montréal, Montréal, QC, Canada, H3T 1J4
| | - Paul Isenring
- Nephrology Research Group, L'Hôtel-Dieu de Québec Research Center, Department of Medicine, Faculty of Medicine, Laval University, Québec, QC, Canada, G1R 2J6
| |
Collapse
|
45
|
Haque MZ, Ortiz PA. Superoxide increases surface NKCC2 in the rat thick ascending limbs via PKC. Am J Physiol Renal Physiol 2019; 317:F99-F106. [PMID: 31091128 DOI: 10.1152/ajprenal.00232.2018] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
The apical Na+-K+-2Cl- cotransporter (NKCC2) mediates NaCl reabsorption by the thick ascending limb (TAL). The free radical superoxide ( O2- ) stimulates TAL NaCl absorption by enhancing NKCC2 activity. In contrast, nitric oxide (NO) scavenges O2- and inhibits NKCC2. NKCC2 activity depends on the number of NKCC2 transporters in the TAL apical membrane and its phosphorylation. We hypothesized that O2- stimulates NKCC2 activity by enhancing apical surface NKCC2 expression. We measured surface NKCC2 expression in rat TALs by surface biotinylation and Western blot analysis. Treatment of TALs with O2- produced by exogenous xanthine oxidase (1 mU/ml) and hypoxanthine (500 µM) stimulated surface NKCC2 expression by ~18 ± 5% (P < 0.05). O2- -stimulated surface NKCC2 expression was blocked by the O2- scavenger tempol (50 µM). Scavenging H2O2 with 100 U/ml catalase did not block the stimulatory effect of xanthine oxidase-hypoxanthine (22 ± 8% increase from control, P < 0.05). Inhibition of endogenous NO production with Nω-nitro-l-arginine methyl ester enhanced surface NKCC2 expression by 21 ± 6% and, when added together with xanthine oxidase-hypoxanthine, increased surface NKCC2 by 41 ± 10% (P < 0.05). Scavenging O2- with superoxide dismutase (300 U/ml) decreased this stimulatory effect by 60% (39 ± 4% to 15 ± 10%, P < 0.05). Protein kinase C inhibition with Gö-6976 (100 nM) blocked O2- -stimulated surface NKCC2 expression (P < 0.05). O2- did not affect NKCC2 phosphorylation at Thr96/101 or its upstream kinases STE20/SPS1-related proline/alanine-rich kinase-oxidative stress-responsive kinase 1. We conclude that O2- increases surface NKCC2 expression by stimulating protein kinase C and that this effect is blunted by endogenous NO. O2- -stimulated apical trafficking of NKCC2 may be involved in the enhanced surface NKCC2 expression observed in Dahl salt-sensitive rats.
Collapse
Affiliation(s)
- Mohammed Ziaul Haque
- Department of Internal Medicine, Hypertension and Vascular Research, Henry Ford Hospital , Detroit, Michigan
| | - Pablo A Ortiz
- Department of Internal Medicine, Hypertension and Vascular Research, Henry Ford Hospital , Detroit, Michigan
| |
Collapse
|
46
|
Rodan AR. Intracellular chloride: a regulator of transepithelial transport in the distal nephron. Curr Opin Nephrol Hypertens 2019; 28:360-367. [PMID: 30865168 PMCID: PMC6684285 DOI: 10.1097/mnh.0000000000000502] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
PURPOSE OF REVIEW This review focuses on the role of intracellular chloride in regulating transepithelial ion transport in the distal convoluted tubule (DCT) in response to perturbations in plasma potassium homeostasis. RECENT FINDINGS Low dietary potassium increases the phosphorylation and activity of the sodium chloride cotransporter (NCC) in the DCT, and vice versa, affecting sodium-dependent potassium secretion in the downstream aldosterone-sensitive distal nephron. In cells, NCC phosphorylation is increased by lowering of intracellular chloride, via activation of the chloride-sensitive with no lysine (WNK)-SPAK/OSR1 (Ste20-related proline/alanine-rich kinase/oxidative stress response) kinase cascade. In-vivo studies have demonstrated pathway activation in the kidney in response to low dietary potassium. A possible mechanism is lowering of DCT intracellular chloride in response to low potassium because of parallel basolateral potassium and chloride channels. Recent studies support a role for these channels in the response of NCC to varying potassium. Studies examining chloride-insensitive WNK mutants, in the Drosophila renal tubule and in the mouse, lend further support to a role for chloride in regulating WNK activity and transepithelial ion transport. Caveats, alternatives, and future directions are also discussed. SUMMARY Chloride sensing by WNK kinase provides a mechanism to allow coupling of extracellular potassium with NCC phosphorylation and activity to maintain potassium homeostasis.
Collapse
Affiliation(s)
- Aylin R. Rodan
- Department of Internal Medicine, Division of Nephrology and Hypertension and Molecular Medicine Program, University of Utah, and Medical Service, Veterans Affairs Salt Lake City Health Care System, Salt Lake City, UT
| |
Collapse
|
47
|
Tyrosine phosphorylation modulates cell surface expression of chloride cotransporters NKCC2 and KCC3. Arch Biochem Biophys 2019; 669:61-70. [PMID: 31145900 DOI: 10.1016/j.abb.2019.05.022] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2019] [Revised: 05/24/2019] [Accepted: 05/26/2019] [Indexed: 11/22/2022]
Abstract
Cellular chloride transport has a fundamental role in cell volume regulation and renal salt handling. Cellular chloride entry or exit are mediated at the plasma membrane by cotransporter proteins of the solute carrier 12 family. For example, NKCC2 resorbs chloride with sodium and potassium ions at the apical membrane of epithelial cells in the kidney, whereas KCC3 releases chloride with potassium ions at the basolateral membrane. Their ion transport activity is regulated by protein phosphorylation in response to signaling pathways. An additional regulatory mechanism concerns the amount of cotransporter molecules inserted into the plasma membrane. Here we describe that tyrosine phosphorylation of NKCC2 and KCC3 regulates their plasma membrane expression levels. We identified that spleen tyrosine kinase (SYK) phosphorylates a specific N-terminal tyrosine residue in each cotransporter. Experimental depletion of endogenous SYK or pharmacological inhibition of its kinase activity increased the abundance of NKCC2 at the plasma membrane of human embryonic kidney cells. In contrast, overexpression of a constitutively active SYK mutant decreased NKCC2 membrane abundance. Intriguingly, the same experimental approaches revealed the opposite effect on KCC3 abundance at the plasma membrane, compatible with the known antagonistic roles of NKCC and KCC cotransporters in cell volume regulation. Thus, we identified a novel pathway modulating the cell surface expression of NKCC2 and KCC3 and show that this same pathway has opposite functional outcomes for these two cotransporters. The findings have several biomedical implications considering the role of these cotransporters in regulating blood pressure and cell volume.
Collapse
|
48
|
Li J, Hu J, Xiang D, Ji B, Xu S, Shi L, Zhao S. KLHL3 single-nucleotide polymorphism is associated with essential hypertension in Chinese Han population. Medicine (Baltimore) 2019; 98:e15766. [PMID: 31096542 PMCID: PMC6531237 DOI: 10.1097/md.0000000000015766] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/27/2022] Open
Abstract
Hypertension, including secondary and essential hypertension (EH) variants, is a multifactorial disease, affecting more than one billion people worldwide. Secondary hypertension results from mutations in the putative gene KLHL3 (Kelch-like protein 3); however, it has not been reported whether the KLHL3 gene polymorphisms are associated with EH. Here, we investigated the association between KLHL3 (rs2301708 and rs7444370) polymorphisms and EH in the Chinese Han population.This case-control study included 522 subjects-260 patients with EH and 262 normotensive controls matched for age, gender, body mass index (BMI), hemoglobin A1c (HbA1c), total cholesterol (TC), triglyceride (TG), and levels of Na, K, and Cl. The distribution of functional rs2301708 and rs7444370 polymorphisms within the KLHL3 gene was assessed through polymerase chain reaction (PCR) and restriction-fragment length polymorphism (RFLP).There was no significant difference in allelic and genotypic frequencies of KLHL3 rs2301708 between the EH and normotensive groups; however, the rs7444370 T allele and CT genotype in females was significantly associated with a protective effect against EH (P = .001, P = .002; P = .019, P = .052), and the haplotype CT of rs2301708 and rs7444370 among females in the EH group was less than in the normotensive group (P = .000; P = .007).The KLHL3 rs7444370 variant could be a protective factor in the pathogenesis of females' EH.
Collapse
Affiliation(s)
- Jin Li
- School of Bioscience and Bioengineering, South China University of Technology
- Department of Pharmacy, Guangzhou United Family Hospital
| | | | | | - Bo Ji
- Department of Pharmacy, Guangzhou General Hospital of Guangzhou Military Command, Guangzhou, China
| | - Suowen Xu
- Department of Medicine, Aab Cardiovascular Research Institute, University of Rochester, Rochester, NY, USA
| | - Lei Shi
- Department of Pharmacy, Guangzhou General Hospital of Guangzhou Military Command, Guangzhou, China
| | - Shujin Zhao
- School of Bioscience and Bioengineering, South China University of Technology
- Department of Pharmacy, Guangzhou General Hospital of Guangzhou Military Command, Guangzhou, China
| |
Collapse
|
49
|
Marcoux AA, Slimani S, Tremblay LE, Frenette-Cotton R, Garneau AP, Isenring P. Regulation of Na +-K +-Cl - cotransporter type 2 by the with no lysine kinase-dependent signaling pathway. Am J Physiol Cell Physiol 2019; 317:C20-C30. [PMID: 30917032 DOI: 10.1152/ajpcell.00041.2019] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023]
Abstract
Na+-K+-Cl- cotransporter type 2 (NKCC2) is confined to the apical membrane of the thick ascending limb of Henle, where it reabsorbs a substantial fraction of the ultrafiltered NaCl load. It is expressed along this nephron segment as three main splice variants (called NKCC2A, NKCC2B, and NKCC2F) that differ in residue composition along their second transmembrane domain and first intracellular cytosolic connecting segment (CS2). NKCC2 is known to be activated by cell shrinkage and intracellular [Cl-] reduction. Although the with no lysine (WNK) kinases could play a role in this response, the mechanisms involved are ill defined, and the possibility of variant-specific responses has not been tested thus far. In this study, we have used the Xenopus laevis oocyte expression system to gain further insight in these regards. We have found for the first time that cell shrinkage could stimulate NKCC2A- and NKCC2B-mediated ion transport by increasing carrier abundance at the cell surface and that this response was achieved (at least in part) by the enzymatic function of a WNK kinase. Interestingly, we have also found that the activity and cell surface abundance of NKCC2F were less affected by cell shrinkage compared with the other variants and that ion transport by certain variants could be stimulated through WNK kinase expression in the absence of carrier redistribution. Taken together, these results suggest that the WNK kinase-dependent pathway can affect both the trafficking as well as intrinsic activity of NKCC2 and that CS2 plays an important role in carrier regulation.
Collapse
Affiliation(s)
- Andrée-Anne Marcoux
- Nephrology Research Group, L'Hôtel-Dieu de Québec Research Center, Department of Medicine, Faculty of Medicine, Laval University , Quebec City, Quebec , Canada
| | - Samira Slimani
- Nephrology Research Group, L'Hôtel-Dieu de Québec Research Center, Department of Medicine, Faculty of Medicine, Laval University , Quebec City, Quebec , Canada
| | - Laurence E Tremblay
- Nephrology Research Group, L'Hôtel-Dieu de Québec Research Center, Department of Medicine, Faculty of Medicine, Laval University , Quebec City, Quebec , Canada
| | - Rachelle Frenette-Cotton
- Nephrology Research Group, L'Hôtel-Dieu de Québec Research Center, Department of Medicine, Faculty of Medicine, Laval University , Quebec City, Quebec , Canada
| | - Alexandre P Garneau
- Nephrology Research Group, L'Hôtel-Dieu de Québec Research Center, Department of Medicine, Faculty of Medicine, Laval University , Quebec City, Quebec , Canada.,Cardiometabolic Research Group, Department of Kinesiology, Faculty of Medicine, University of Montreal , Montreal, Quebec , Canada
| | - Paul Isenring
- Nephrology Research Group, L'Hôtel-Dieu de Québec Research Center, Department of Medicine, Faculty of Medicine, Laval University , Quebec City, Quebec , Canada
| |
Collapse
|
50
|
Gonzalez-Vicente A, Saez F, Monzon CM, Asirwatham J, Garvin JL. Thick Ascending Limb Sodium Transport in the Pathogenesis of Hypertension. Physiol Rev 2019; 99:235-309. [PMID: 30354966 DOI: 10.1152/physrev.00055.2017] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
The thick ascending limb plays a key role in maintaining water and electrolyte balance. The importance of this segment in regulating blood pressure is evidenced by the effect of loop diuretics or local genetic defects on this parameter. Hormones and factors produced by thick ascending limbs have both autocrine and paracrine effects, which can extend prohypertensive signaling to other structures of the nephron. In this review, we discuss the role of the thick ascending limb in the development of hypertension, not as a sole participant, but one that works within the rich biological context of the renal medulla. We first provide an overview of the basic physiology of the segment and the anatomical considerations necessary to understand its relationship with other renal structures. We explore the physiopathological changes in thick ascending limbs occurring in both genetic and induced animal models of hypertension. We then discuss the racial differences and genetic defects that affect blood pressure in humans through changes in thick ascending limb transport rates. Throughout the text, we scrutinize methodologies and discuss the limitations of research techniques that, when overlooked, can lead investigators to make erroneous conclusions. Thus, in addition to advancing an understanding of the basic mechanisms of physiology, the ultimate goal of this work is to understand our research tools, to make better use of them, and to contextualize research data. Future advances in renal hypertension research will require not only collection of new experimental data, but also integration of our current knowledge.
Collapse
Affiliation(s)
| | - Fara Saez
- Department of Physiology and Biophysics, Case Western Reserve University , Cleveland, Ohio
| | - Casandra M Monzon
- Department of Physiology and Biophysics, Case Western Reserve University , Cleveland, Ohio
| | - Jessica Asirwatham
- Department of Physiology and Biophysics, Case Western Reserve University , Cleveland, Ohio
| | - Jeffrey L Garvin
- Department of Physiology and Biophysics, Case Western Reserve University , Cleveland, Ohio
| |
Collapse
|