1
|
Duan S, Agger K, Messling JE, Nishimura K, Han X, Peña-Rømer I, Shliaha P, Damhofer H, Douglas M, Kohli M, Pal A, Asad Y, Van Dyke A, Reilly R, Köchl R, Tybulewicz VLJ, Hendrickson RC, Raynaud FI, Gallipoli P, Poulogiannis G, Helin K. WNK1 signalling regulates amino acid transport and mTORC1 activity to sustain acute myeloid leukaemia growth. Nat Commun 2025; 16:4920. [PMID: 40425534 PMCID: PMC12116911 DOI: 10.1038/s41467-025-59969-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2024] [Accepted: 05/08/2025] [Indexed: 05/29/2025] Open
Abstract
The lack of curative therapies for acute myeloid leukaemia (AML) remains an ongoing challenge despite recent advances in the understanding of the molecular basis of the disease. Here we identify the WNK1-OXSR1/STK39 pathway as a previously uncharacterised dependency in AML. We show that genetic depletion and pharmacological inhibition of WNK1 or its downstream phosphorylation targets OXSR1 and STK39 strongly reduce cell proliferation and induce apoptosis in leukaemia cells in vitro and in vivo. Furthermore, we show that the WNK1-OXSR1/STK39 pathway controls mTORC1 signalling via regulating amino acid uptake through a mechanism involving the phosphorylation of amino acid transporters, such as SLC38A2. Our findings underscore an important role of the WNK1-OXSR1/STK39 pathway in regulating amino acid uptake and driving AML progression.
Collapse
Affiliation(s)
- Shunlei Duan
- Division of Cell and Molecular Biology, The Institute of Cancer Research, Londo, UK
- Biotech Research and Innovation Centre, University of Copenhagen, Copenhagen, Denmark
| | - Karl Agger
- Biotech Research and Innovation Centre, University of Copenhagen, Copenhagen, Denmark
| | - Jan-Erik Messling
- Biotech Research and Innovation Centre, University of Copenhagen, Copenhagen, Denmark
| | - Koutarou Nishimura
- Biotech Research and Innovation Centre, University of Copenhagen, Copenhagen, Denmark
- Cell Biology Program and Center for Epigenetics Research, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Xuerui Han
- Division of Cell and Molecular Biology, The Institute of Cancer Research, Londo, UK
- Biotech Research and Innovation Centre, University of Copenhagen, Copenhagen, Denmark
| | - Isabel Peña-Rømer
- Division of Cell and Molecular Biology, The Institute of Cancer Research, Londo, UK
| | - Pavel Shliaha
- Microchemistry and Proteomics Core Facility, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Helene Damhofer
- Division of Cell and Molecular Biology, The Institute of Cancer Research, Londo, UK
- Biotech Research and Innovation Centre, University of Copenhagen, Copenhagen, Denmark
- Cell Biology Program and Center for Epigenetics Research, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Max Douglas
- Division of Cell and Molecular Biology, The Institute of Cancer Research, Londo, UK
| | - Manas Kohli
- Division of Cell and Molecular Biology, The Institute of Cancer Research, Londo, UK
| | - Akos Pal
- Division of Cancer Therapeutics, The Institute of Cancer Research, London, UK
| | - Yasmin Asad
- Division of Cancer Therapeutics, The Institute of Cancer Research, London, UK
| | - Aaron Van Dyke
- Department of Chemistry & Biochemistry, Fairfield University, Fairfield, CT, USA
| | - Raquel Reilly
- Department of Chemistry & Biochemistry, Fairfield University, Fairfield, CT, USA
| | | | | | - Ronald C Hendrickson
- Microchemistry and Proteomics Core Facility, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Florence I Raynaud
- Division of Cancer Therapeutics, The Institute of Cancer Research, London, UK
| | - Paolo Gallipoli
- Centre for Haemato-Oncology, Barts Cancer Institute, Queen Mary University of London, London, UK
| | - George Poulogiannis
- Division of Cell and Molecular Biology, The Institute of Cancer Research, Londo, UK
| | - Kristian Helin
- Division of Cell and Molecular Biology, The Institute of Cancer Research, Londo, UK.
- Biotech Research and Innovation Centre, University of Copenhagen, Copenhagen, Denmark.
- Cell Biology Program and Center for Epigenetics Research, Memorial Sloan Kettering Cancer Center, New York, NY, USA.
| |
Collapse
|
2
|
Ferdaus MZ, Delpire E. Calcium-binding protein 39 in with-no-lysine kinase signaling and the modulation of renal tubular transport. Curr Opin Nephrol Hypertens 2025:00041552-990000000-00235. [PMID: 40357626 DOI: 10.1097/mnh.0000000000001083] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/15/2025]
Abstract
PURPOSE OF REVIEW The regulation of renal tubular transport is essential for maintaining electrolyte balance and blood pressure. Calcium-binding protein 39 (Cab39), also known as mouse protein-25 (MO25), plays a pivotal role in modulating this process through its interaction with WNK (with no lysine) kinases and Ste20-like kinases, including STE20/SPS1-related proline-alanine-rich kinase (SPAK) and oxidative stress response 1 (OSR1). By stabilizing and facilitating the activation of these kinases, Cab39 plays a crucial role in the regulation of key ion transporters, such as the sodium-chloride cotransporter (NCC) and the sodium-potassium-chloride cotransporters (NKCC1 and NKCC2). This review provides a comprehensive analysis of Cab39 structural properties, molecular interactions, and functional roles in renal physiology, emphasizing its significance in ion homeostasis. RECENT FINDINGS Studies reveal that Cab39 enhances SPAK activity up to 100-fold. Importantly, the role of Cab39 extends beyond simple kinase activation, as it supports kinase complex assembly and localization, enabling precise control over transporter regulation. Evidence also suggests that Cab39 may influence the regulation of NCC and NKCC2 through similar mechanisms, making it a promising target for therapeutic interventions in disorders such as hypertension and salt-wasting syndromes. SUMMARY The discovery of a small-molecule Cab39 inhibitor highlights its potential as a pharmacological target. Understanding the multifaceted functions of Cab39 may unlock novel strategies for managing renal and cardiovascular disorders.
Collapse
Affiliation(s)
- Mohammed Z Ferdaus
- Department of Anesthesiology, Vanderbilt University Medical School, Nashville, Tennessee, USA
| | | |
Collapse
|
3
|
Wade Wei TY, Hsia JY, Yang TY, Huang YX, Wang GZ, Liu YC, Wang YW, He F, Maureen Chen JM, Zhao GJ, Lyu PC, Su LJ, Chiu SC, Ricky Yu CT. Mechanistic Insights into CLNS1A-Mediated Chemoresistance and Tumor Progression in Non-small Cell Lung Cancer. Cancer Lett 2025:217783. [PMID: 40345428 DOI: 10.1016/j.canlet.2025.217783] [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: 03/19/2025] [Revised: 05/05/2025] [Accepted: 05/06/2025] [Indexed: 05/11/2025]
Abstract
CLNS1A is a chloride channel protein and an essential component of the methylosome complex, which additionally comprises PRMT5 and MEP50. In this study, we investigated its contribution to lung cancer and its potential as a therapeutic target. Analysis of transcriptomic datasets and western blotting revealed that CLNS1A, PRMT5, and MEP50 were overexpressed in lung cancer tissues, with elevated CLNS1A expression correlating with poor patient survival. CLNS1A overexpression enhanced platinum clearance from cells, increased the IC50 values for chemotherapy, and improved cell survival. Conversely, the knockdown of CLNS1A increased drug accumulation, reduced survival, and increased sensitivity to chemotherapy. The 3W mutant, a chloride channel-defective variant with steric hindrance at key bottleneck residues, impaired chloride ion transport, thereby reducing drug resistance, migration, and anchorage-independent growth. Mechanistically, CLNS1A promotes drug efflux through its chloride channel activity and activates the FAK-SRC-RAC1 pathway to enhance motility and clonogenicity. It also facilitates PRMT5-mediated RUVBL1 methylation to support anti-apoptotic DNA damage response signaling. In vivo, CLNS1A overexpression accelerated tumor growth and reduced survival, whereas CLNS1A knockdown sensitized tumors to cisplatin, enhancing therapeutic efficacy. These findings suggest that CLNS1A is a potential biomarker and therapeutic target, and its inhibition offers a strategy to overcome drug resistance and limit the metastatic progression of lung cancer.
Collapse
Affiliation(s)
- Tong-You Wade Wei
- Department of Applied Chemistry, National Chi Nan University, Nantou, Taiwan; Division of Cardiology, Department of Medicine, University of California, San Diego, La Jolla, California, USA.
| | - Jiun-Yi Hsia
- Division of Thoracic Surgery, Chung Shan Medical University Hospital, Taichung, Taiwan; School of Medicine, Chung Shan Medical University, Taichung, Taiwan
| | - Tsung-Ying Yang
- Department of Chest Medicine, Taichung Veterans General Hospital, Taichung, Taiwan; Doctoral Program in Translational Medicine, National Chung Hsing University, Taichung, Taiwan
| | - Yu-Xu Huang
- Department of Medicine, JingGangShan University, Ji'an, China
| | - Guo-Zhi Wang
- Division of Thoracic Surgery, Chung Shan Medical University Hospital, Taichung, Taiwan
| | - Yi-Chung Liu
- Department of Life Sciences, National Tsing Hua University, Hsinchu, Taiwan
| | - Yi-Wun Wang
- Department of Life Sciences, National Tsing Hua University, Hsinchu, Taiwan
| | - Fangzhou He
- Cardiovascular Research Center, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, Xi'an, China
| | - Jo-Mei Maureen Chen
- Department of Applied Chemistry, National Chi Nan University, Nantou, Taiwan
| | - Guo-Jun Zhao
- Department of Cardiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Ping-Chiang Lyu
- Institute of Bioinformatics and Structural Biology, Department of Life Science, National Tsing Hua University, Hsinchu, Taiwan
| | - Li-Jen Su
- Department of Biomedical Sciences and Engineering, Education and Research Center for Technology Assisted Substance Abuse Prevention and Management, and Core Facilities for High Throughput Experimental Analysis, National Central University, Taoyuan, Taiwan
| | - Shao-Chih Chiu
- Graduate Institute of Biomedical Sciences, China Medical University, Taichung, Taiwan; Translational Cell Therapy Center, Department of Medical Research, China Medical University Hospital, Taichung, Taiwan.
| | - Chang-Tze Ricky Yu
- Department of Applied Chemistry, National Chi Nan University, Nantou, Taiwan.
| |
Collapse
|
4
|
Ranganathan R, Sari F, Wang SX, Thiery A, Buzzi AL, Guerra R, Moody SA, Streit A. Targets of the transcription factor Six1 identify previously unreported candidate deafness genes. Development 2025; 152:dev204533. [PMID: 40213817 PMCID: PMC12045605 DOI: 10.1242/dev.204533] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2024] [Accepted: 02/12/2025] [Indexed: 05/03/2025]
Abstract
Branchio-otic (BOS) and branchio-oto-renal (BOR) syndromes are autosomal dominant disorders featuring multiple birth defects including ear, renal and branchial malformations. Mutations in the homeodomain transcription factor SIX1 and its co-factor EYA1 have been identified in about 50% of individuals with BOS or BOR, while causative mutations are unknown in the other half. We hypothesise that SIX1 target genes represent new BOS and BOR candidates. Using published transcriptomic and epigenomic data from chick ear progenitors, we first identify putative Six1 targets. Next, we provide evidence that Six1 directly regulates some of these candidates: Six1 binds to their enhancers, and functional experiments in Xenopus and chick confirm that Six1 controls their expression. Finally, we show that most putative chick Six1 targets are also expressed in the human developing ear and are associated with known deafness loci. Together, our results not only characterise the molecular mechanisms that mediate Six1 function in the developing ear, but also provide new candidates for human congenital deafness.
Collapse
Affiliation(s)
- Ramya Ranganathan
- Centre for Craniofacial and Regenerative Biology, King's College London, London SE1 9RT, UK
| | - Fereshteh Sari
- Centre for Craniofacial and Regenerative Biology, King's College London, London SE1 9RT, UK
| | - Scarlet Xiaoyan Wang
- Centre for Craniofacial and Regenerative Biology, King's College London, London SE1 9RT, UK
| | - Alexandre Thiery
- Centre for Craniofacial and Regenerative Biology, King's College London, London SE1 9RT, UK
| | - Ailin Leticia Buzzi
- Centre for Craniofacial and Regenerative Biology, King's College London, London SE1 9RT, UK
| | - Rosalinda Guerra
- Centre for Craniofacial and Regenerative Biology, King's College London, London SE1 9RT, UK
| | - Sally A. Moody
- Department of Anatomy & Cell Biology, George Washington University School of Medicine and Health Sciences, Washington, DC 20052, USA
| | - Andrea Streit
- Centre for Craniofacial and Regenerative Biology, King's College London, London SE1 9RT, UK
| |
Collapse
|
5
|
Lu X, Xue J, Zhou Q, Huang J. A "Knob Switch" Model for the Phosphoregulatory Mechanism of KCC3 at the Carboxy-Terminal Domain. Biochemistry 2025; 64:1405-1414. [PMID: 40009635 DOI: 10.1021/acs.biochem.4c00523] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/28/2025]
Abstract
Phosphorylation is a reversible post-translational modification that can modulate protein function. For example, phosphorylation modifications of solute carrier family 12 (SLC12) proteins function as molecular switches that precisely regulate cation-chloride ion transport. Elucidating the phosphoregulatory mechanism of SLC12 at the carboxy-terminal domain (CTD) through structural determination approaches remains challenging due to the domain's disordered and flexible nature. In this study, molecular dynamics (MD) simulations and enhanced sampling techniques were employed to investigate the CTD phosphoregulatory mechanism of SLC12A6 (also known as KCC3). Atomistic MD and metadynamics simulations revealed that the dephosphorylation of residues T940 and T997 stabilizes CTD to a favorable state that "switches on" the solvent accessibility of the inward-facing pocket. Meanwhile, phosphorylation induces distinct orientations of the CTD, transitioning the dimer into another favorable state that "switches off" the solvent accessibility. The alteration of solvent accessibility in the inward-facing pocket influences the water and ion dynamics. Based on these findings, we propose a "knob switch" model to illustrate how CTD phosphorylation regulates ion transport in KCC3.
Collapse
Affiliation(s)
- Xiaoli Lu
- Institute of Biology, Westlake Institute for Advanced Study, Hangzhou 310024, China
- Key Laboratory of Structural Biology of Zhejiang Province, School of Life Sciences, Westlake University, Hangzhou 310024, China
- Westlake AI Therapeutics Lab, Westlake Laboratory of Life Sciences and Biomedicine, Hangzhou 310024, China
| | - Jing Xue
- Institute of Biology, Westlake Institute for Advanced Study, Hangzhou 310024, China
- Key Laboratory of Structural Biology of Zhejiang Province, School of Life Sciences, Westlake University, Hangzhou 310024, China
- Westlake AI Therapeutics Lab, Westlake Laboratory of Life Sciences and Biomedicine, Hangzhou 310024, China
| | - Qiang Zhou
- Institute of Biology, Westlake Institute for Advanced Study, Hangzhou 310024, China
- Key Laboratory of Structural Biology of Zhejiang Province, School of Life Sciences, Westlake University, Hangzhou 310024, China
| | - Jing Huang
- Institute of Biology, Westlake Institute for Advanced Study, Hangzhou 310024, China
- Key Laboratory of Structural Biology of Zhejiang Province, School of Life Sciences, Westlake University, Hangzhou 310024, China
- Westlake AI Therapeutics Lab, Westlake Laboratory of Life Sciences and Biomedicine, Hangzhou 310024, China
| |
Collapse
|
6
|
Fdel AM, Waters L, Sharma I, Jones S, Gee J, Atack JR, Banerjee S, Mehellou Y. Oxidative Stress-Responsive 1 Kinase Catalytic Activity Promotes Triple Negative Breast Cancer Oncogenic Potential. ACS Pharmacol Transl Sci 2025; 8:726-735. [PMID: 40109757 PMCID: PMC11915029 DOI: 10.1021/acsptsci.4c00603] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2024] [Revised: 02/04/2025] [Accepted: 02/24/2025] [Indexed: 03/22/2025]
Abstract
The protein kinase OSR1 has been highlighted as a biomarker for a poor prognosis in breast cancer (BC) patients. To further decipher the mechanism underpinning this, we studied the expression, phosphorylation status, and catalytic activity of OSR1 across a series of BC cell lines. OSR1 was found to be expressed across the various luminal and triple negative BC (TNBC) cell lines studied, although it was only constitutively active in the highly migratory TNBC cell line MDA-MB-231. Although this cell line carries p53 mutations, our data indicated that OSR1 constitutive kinase activity of the OSR1 in MDA-MB-231 was independent of p53. Interestingly, the inhibition of OSR1 had no significant impact on MDA-MB-231 cell viability, but it was found to contribute to its substantial cell migration and invasion, as this was significantly attenuated by the WNK/OSR1 inhibitor WNK463. Analogously, the overexpression of constitutively active OSR1 in the poorly migrating BC cell line MCF7 enhanced its cell mobility. Collectively, our results indicate that the pharmacological inhibition of OSR1 could be a promising novel strategy for preventing the oncogenic potential of TNBC.
Collapse
Affiliation(s)
- Azeza M Fdel
- Cardiff School of Pharmacy and Pharmaceutical Sciences, Cardiff University, Cardiff CF10 3NB, U.K
| | - Loren Waters
- Medicines Discovery Institute, Cardiff University, Cardiff CF10 3AT, U.K
| | - Ira Sharma
- Division of Cancer Research, School of Medicine, University of Dundee, Dundee DD1 9SY, U.K
| | - Samuel Jones
- Cardiff School of Pharmacy and Pharmaceutical Sciences, Cardiff University, Cardiff CF10 3NB, U.K
| | - Julia Gee
- Cardiff School of Pharmacy and Pharmaceutical Sciences, Cardiff University, Cardiff CF10 3NB, U.K
| | - John R Atack
- Medicines Discovery Institute, Cardiff University, Cardiff CF10 3AT, U.K
| | - Sourav Banerjee
- Division of Cancer Research, School of Medicine, University of Dundee, Dundee DD1 9SY, U.K
| | - Youcef Mehellou
- Cardiff School of Pharmacy and Pharmaceutical Sciences, Cardiff University, Cardiff CF10 3NB, U.K
- Medicines Discovery Institute, Cardiff University, Cardiff CF10 3AT, U.K
| |
Collapse
|
7
|
Deng Z, Wang H, Zhong K, Li Y, Deng H, Gao B, Huang K, Tong A, Zhou L. The Role of Choroid Plexus in Hydrocephalus from the Perspective of Structure and Function: a Therapeutic Target. Mol Neurobiol 2025:10.1007/s12035-025-04823-7. [PMID: 40085357 DOI: 10.1007/s12035-025-04823-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2024] [Accepted: 03/06/2025] [Indexed: 03/16/2025]
Abstract
Hydrocephalus is one of the most common neurological diseases, characterized by abnormal excessive accumulation of cerebrospinal fluid (CSF) in the ventricular system. Its pathophysiological mechanism is believed to be related to the imbalance of CSF circulation and homeostasis. As the main source of CSF secretion, the choroid plexus is closely related to hydrocephalus. The choroid plexus is a specialized vascularized tissue located within the cerebral ventricles. It has multiple physiological functions including regulating CSF, immune response, endocrine metabolism, etc. Strategies that reduce choroid plexus CSF secretion have been shown to be effective in the treatment of hydrocephalus. However, the role of other physiological functions of the choroid plexus in hydrocephalus is still unclear. Recent studies on the choroid plexus and the blood-CSF barrier have deepened our understanding of the structure and function of the choroid plexus. The idea of targeting the choroid plexus to treat hydrocephalus has spawned many branches: choroid plexus epithelial cells, choroid plexus immune cells, choroid plexus peptides, and choroid plexus cilia, etc. This review introduces the basic structure and function of the choroid plexus, summarizes their changes in hydrocephalus, and analyzes the possibility of the choroid plexus as a therapeutic target for hydrocephalus.
Collapse
Affiliation(s)
- Ziang Deng
- Department of Neurosurgery, West China Hospital, Sichuan University, Chengdu, China
| | - Haoxiang Wang
- Department of Neurosurgery, West China Hospital, Sichuan University, Chengdu, China
| | - Kunhong Zhong
- State Key Laboratory of Biotherapy and Cancer Center, Research Unit of Gene and Immunotherapy, Chinese Academy of Medical Sciences, Collaborative Innovation Center of Biotherapy, West China Hospital, Sichuan University, Chengdu, China
| | - Yuanyou Li
- Department of Pediatric Neurosurgery, West China Second University Hospital, Sichuan University, Chengdu, China
| | - Huajiang Deng
- Department of Neurosurgery, Affiliated Hospital of Southwest Medical University, Sichuan Province, Luzhou City, China
| | - Baocheng Gao
- Department of Neurosurgery, The First People'S Hospital of Yunnan Province, the Affiliated Hospital of Kunming University of Science and Technology) Kunming, Yunan, China
| | - Keru Huang
- Department of Neurosurgery, West China Hospital, Sichuan University, Chengdu, China
| | - Aiping Tong
- State Key Laboratory of Biotherapy and Cancer Center, Research Unit of Gene and Immunotherapy, Chinese Academy of Medical Sciences, Collaborative Innovation Center of Biotherapy, West China Hospital, Sichuan University, Chengdu, China.
- Frontiers Medical Center, Tianfu Jincheng Laboratory, Chengdu, China.
| | - Liangxue Zhou
- Department of Neurosurgery, West China Hospital, Sichuan University, Chengdu, China.
- Department of Neurosurgery, Mianyang Central Hospital, School of Medicine, University of Electronic Science and Technology of China, Mianyang, China.
- Department of Neurosurgery, The Fifth People's Hospital of Ningxia, Shizuishan, China.
| |
Collapse
|
8
|
Sun J, Sun Y, Ma B, Qi R, Hao X, Lv J, Shi J, Wu W, Fu X, Shi R. Mechanisms of polygalasaponin F against brain ischemia-reperfusion injury by targeting NKCC1. Exp Neurol 2025; 385:115076. [PMID: 39608559 DOI: 10.1016/j.expneurol.2024.115076] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2024] [Revised: 11/01/2024] [Accepted: 11/24/2024] [Indexed: 11/30/2024]
Abstract
Stroke is a serious threat to human health and current clinical therapies remain unsatisfactory. Elevated expression of Na+-K+-2Cl- cotransporter 1 (NKCC1) following stroke can disrupt the blood-brain barrier (BBB) and result in brain edema, indicating that NKCC1 may be a potential therapeutic target for improving stroke outcomes. Polygalasaponin F (PGSF) is a triterpenoid saponin isolated from Polygala japonica Houtt, which has showed neuroprotective effects in previous studies. The present study aimed to assess the protective effects of PGSF on cerebral ischemia-reperfusion injury (CIRI) in vivo and elucidate its underlying mechanism by targeting NKCC1. Experimental results revealed that following CIRI, rats displayed neurological deficits, cerebral infarction and brain edema, concurrent with increased NKCC1 mRNA and protein expression in the cerebral tissue. Notably, the administration of PGSF at both 10 mg/kg and 20 mg/kg effectively mitigated these adverse outcomes. To explore the mechanism of PGSF, pyrosequencing was used to find that CIRI reduces the methylation of the NKCC1 promoter, while PGSF enhances it. It was thereby demonstrated that PGSF could reduce NKCC1 expression in this manner. Simultaneously, we also observed that the protein expression of DNA methyltransferase 1 (DNMT1) in the ischemic penumbra was augmented after CIRI, whereas PGSF reduced the expression of DNMT1, which was contrary to the trend of NKCC1 methylation under the treatment of PGSF. These results imply that the enhancement of NKCC1 methylation by PGSF may not be catalyzed by DNMT1 and that the reduction of NKCC1 methylation level after CIRI may not be related to DNMT1. Finally, we discovered that PGSF can decrease the leakage of the BBB and enhance the expression of the BBB structural proteins occludin and ZO-1. In conclusion, PGSF can target NKCC1 as an epigenetic target and downregulate its expression following CIRI by enhancing DNA methylation of NKCC1, thereby safeguarding the structure and function of brain tissue.
Collapse
Affiliation(s)
- Jianqi Sun
- Department of Physiology, Baotou Medical College, Baotou, Inner Mongolia 014040, China; The Second Affiliated Hospital of Baotou Medical College, Baotou, Inner Mongolia 014000, China
| | - Yao Sun
- Department of Physiology, Baotou Medical College, Baotou, Inner Mongolia 014040, China
| | - Baohui Ma
- Department of Physiology, Baotou Medical College, Baotou, Inner Mongolia 014040, China
| | - Ruifang Qi
- Department of Physiology, Baotou Medical College, Baotou, Inner Mongolia 014040, China
| | - Xiaoqiong Hao
- Department of Physiology, Baotou Medical College, Baotou, Inner Mongolia 014040, China
| | - Jun Lv
- Department of Physiology, Baotou Medical College, Baotou, Inner Mongolia 014040, China
| | - Jinghua Shi
- Department of Physiology, Baotou Medical College, Baotou, Inner Mongolia 014040, China
| | - Wei Wu
- Department of Physiology, Baotou Medical College, Baotou, Inner Mongolia 014040, China
| | - Xuyang Fu
- Department of Physiology, Baotou Medical College, Baotou, Inner Mongolia 014040, China
| | - Ruili Shi
- Department of Physiology, Baotou Medical College, Baotou, Inner Mongolia 014040, China; Institute of Neuroscience, Baotou Medical College, Baotou, Inner Mongolia 014040, China.
| |
Collapse
|
9
|
Zhao Y, Vidossich P, Forbush B, Ma J, Rinehart J, De Vivo M, Cao E. Structural basis for human NKCC1 inhibition by loop diuretic drugs. EMBO J 2025; 44:1540-1562. [PMID: 39875725 PMCID: PMC11876703 DOI: 10.1038/s44318-025-00368-6] [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: 06/14/2024] [Revised: 12/11/2024] [Accepted: 01/02/2025] [Indexed: 01/30/2025] Open
Abstract
Na+-K+-Cl- cotransporters functions as an anion importers, regulating trans-epithelial chloride secretion, cell volume, and renal salt reabsorption. Loop diuretics, including furosemide, bumetanide, and torsemide, antagonize both NKCC1 and NKCC2, and are first-line medicines for the treatment of edema and hypertension. NKCC1 activation by the molecular crowding sensing WNK kinases is critical if cells are to combat shrinkage during hypertonic stress; however, how phosphorylation accelerates NKCC1 ion transport remains unclear. Here, we present co-structures of phospho-activated NKCC1 bound with furosemide, bumetanide, or torsemide showing that furosemide and bumetanide utilize a carboxyl group to coordinate and co-occlude a K+, whereas torsemide encroaches and expels the K+ from the site. We also found that an amino-terminal segment of NKCC1, once phosphorylated, interacts with the carboxyl-terminal domain, and together, they engage with intracellular ion exit and appear to be poised to facilitate rapid ion translocation. Together, these findings enhance our understanding of NKCC-mediated epithelial ion transport and the molecular mechanisms of its inhibition by loop diuretics.
Collapse
Affiliation(s)
- Yongxiang Zhao
- Department of Biochemistry, University of Utah School of Medicine, Salt Lake City, UT, 84112-5650, USA
- Key Laboratory of Magnetic Resonance in Biological Systems, State Key Laboratory of Magnetic Resonance and Atomic and Molecular Physics, National Center for Magnetic Resonance in Wuhan, Wuhan Institute of Physics and Mathematics, Innovation Academy for Precision Measurement Science and Technology, Chinese Academy of Sciences, 430071, Wuhan, P. R. China
| | - Pietro Vidossich
- Laboratory of Molecular Modelling & Drug Discovery, Istituto Italiano di Tecnologia, Genoa, Via Morego 30, 16163, Italy
| | - Biff Forbush
- Department of Cellular and Molecular Physiology, Yale University School of Medicine, New Haven, CT, USA
| | - Junfeng Ma
- Lombardi Comprehensive Cancer Center, Georgetown University Medical Center, Washington DC, 20057, USA
| | - Jesse Rinehart
- Department of Cellular and Molecular Physiology, Yale University School of Medicine, New Haven, CT, USA
- Systems Biology Institute, Yale University, West Haven, CT, USA
| | - Marco De Vivo
- Laboratory of Molecular Modelling & Drug Discovery, Istituto Italiano di Tecnologia, Genoa, Via Morego 30, 16163, Italy
| | - Erhu Cao
- Department of Biochemistry, University of Utah School of Medicine, Salt Lake City, UT, 84112-5650, USA.
| |
Collapse
|
10
|
Biggs O'May J, Vanes L, de Boer LL, Lewis DA, Hartweger H, Kunzelmann S, Hayward D, Llorian M, Köchl R, Tybulewicz VLJ. WNK1-dependent water influx is required for CD4 + T cell activation and T cell-dependent antibody responses. Nat Commun 2025; 16:1857. [PMID: 39984435 PMCID: PMC11845700 DOI: 10.1038/s41467-025-56778-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2022] [Accepted: 01/29/2025] [Indexed: 02/23/2025] Open
Abstract
Signaling from the T cell antigen receptor (TCR) on CD4+ T cells plays a critical role in adaptive immune responses by inducing T cell activation, proliferation, and differentiation. Here we demonstrate that WNK1, a kinase implicated in osmoregulation in the kidney, is required in T cells to support T-dependent antibody responses. We show that the canonical WNK1-OXSR1-STK39 kinase signaling pathway is required for TCR signaling in CD4+ T cells, their subsequent entry into the cell cycle, and suppression of the ATR-mediated G2/M cell cycle checkpoint. We show that the WNK1 pathway regulates ion influx leading to water influx, potentially through AQP3, and that water influx is required for TCR-induced signaling and cell cycle entry. Thus, TCR signaling via WNK1, OXSR1, STK39 and AQP3 leads to water entry that is essential for CD4+ T cell proliferation and hence T cell-dependent antibody responses.
Collapse
Affiliation(s)
| | - Lesley Vanes
- The Francis Crick Institute, London, NW1 1AT, UK
| | - Leonard L de Boer
- The Francis Crick Institute, London, NW1 1AT, UK
- Imperial College, London, W12 0NN, UK
- Science for Life Laboratory, Department of Women's and Children's Health, Karolinska Institute, Box 1031, SE-171 21, Solna, Sweden
| | | | - Harald Hartweger
- The Francis Crick Institute, London, NW1 1AT, UK
- Laboratory of Molecular Immunology, The Rockefeller University, 10065, New York, NY, USA
| | | | - Darryl Hayward
- The Francis Crick Institute, London, NW1 1AT, UK
- GSK, Stevenage, SG1 2NY, UK
| | | | - Robert Köchl
- The Francis Crick Institute, London, NW1 1AT, UK
- Kings College London, London, SE1 9RT, UK
| | | |
Collapse
|
11
|
Bakouh N, Castaño-Martín R, Metais A, Dan EL, Balducci E, Chhuon C, Lepicka J, Barcia G, Losito E, Lourdel S, Planelles G, Muresan RC, Moca VV, Kaminska A, Bourgeois M, Chemaly N, Rguez Y, Auvin S, Huberfeld G, Varlet P, Asnafi V, Guerrera IC, Kabashi E, Nabbout R, Ciura S, Blauwblomme T. Chloride deregulation and GABA depolarization in MTOR-related malformations of cortical development. Brain 2025; 148:549-563. [PMID: 39106285 PMCID: PMC11788215 DOI: 10.1093/brain/awae262] [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: 03/11/2024] [Revised: 06/15/2024] [Accepted: 07/19/2024] [Indexed: 08/09/2024] Open
Abstract
Focal cortical dysplasia, hemimegalencephaly and cortical tubers are paediatric epileptogenic malformations of cortical development (MCDs) frequently pharmacoresistant and mostly treated surgically by the resection of epileptic cortex. Availability of cortical resection samples has allowed significant mechanistic discoveries directly from human material. Causal brain somatic or germline mutations in the AKT/PI3K/DEPDC5/MTOR genes have been identified. GABAA-mediated paradoxical depolarization, related to altered chloride (Cl-) homeostasis, has been shown to participate to ictogenesis in human paediatric MCDs. However, the link between genomic alterations and neuronal hyperexcitability is unclear. Here, we studied the post-translational interactions between the mTOR pathway and the regulation of cation-chloride cotransporters (CCCs), KCC2 and NKCC1, that are largely responsible for controlling intracellular Cl- and, ultimately, GABAergic transmission. For this study, 35 children (25 MTORopathies and 10 pseudo-controls, diagnosed by histology plus genetic profiling) were operated for drug-resistant epilepsy. Postoperative cortical tissues were recorded on a multi-electrode array to map epileptic activities. CCC expression level and phosphorylation status of the WNK1/SPAK-OSR1 pathway was measured during basal conditions and after pharmacological modulation. Direct interactions between mTOR and WNK1 pathway components were investigated by immunoprecipitation. Membranous incorporation of MCD samples in Xenopus laevis oocytes enabled measurement of the Cl- conductance and equilibrium potential for GABA. Of the 25 clinical cases, half harboured a somatic mutation in the mTOR pathway, and pS6 expression was increased in all MCD samples. Spontaneous interictal discharges were recorded in 65% of the slices. CCC expression was altered in MCDs, with a reduced KCC2/NKCC1 ratio and decreased KCC2 membranous expression. CCC expression was regulated by the WNK1/SPAK-OSR1 kinases through direct phosphorylation of Thr906 on KCC2, which was reversed by WNK1 and SPAK antagonists (N-ethylmaleimide and staurosporine). The mSIN1 subunit of MTORC2 was found to interact with SPAK-OSR1 and WNK1. Interactions between these key epileptogenic pathways could be reversed by the mTOR-specific antagonist rapamycin, leading to a dephosphorylation of CCCs and recovery of the KCC2/NKCC1 ratio. The functional effect of such recovery was validated by the restoration of the depolarizing shift in the equilibrium potential for GABA by rapamycin, measured after incorporation of MCD membranes into X. laevis oocytes, in line with a re-establishment of normal Cl- reversal potential. Our study deciphers a protein interaction network through a phosphorylation cascade between MTOR and WNK1/SPAK-OSR1 leading to deregulation of chloride cotransporters, increased neuronal Cl- levels and GABAA dysfunction in malformations of cortical development, linking genomic defects and functional effects and paving the way to target epilepsy therapy.
Collapse
Affiliation(s)
- Naziha Bakouh
- Translational Research in Neuroscience Lab, Institut Imagine, Université Paris Cité, INSERM U1163, 75015 Paris, France
| | - Reyes Castaño-Martín
- Translational Research in Neuroscience Lab, Institut Imagine, Université Paris Cité, INSERM U1163, 75015 Paris, France
| | - Alice Metais
- Institute of Psychiatry and Neuroscience of Paris (IPNP), Université Paris Cité, INSERM U1266, 75014 Paris, France
- Service de Neuropathologie, GHU-Paris Psychiatrie et Neurosciences, Hôpital Sainte Anne, F-75014 Paris, France
| | | | - Estelle Balducci
- Department of Pediatric Neurosurgery Hôpital Necker, Assistance Publique Hôpitaux de Paris, 75015 Paris, France
- Department of Pediatric Neurology, Hôpital Necker, Assistance Publique Hôpitaux de Paris, 75015 Paris, France
| | - Cerina Chhuon
- INSERM US24, Proteomic platform, SFR Necker, 75015 Paris, France
| | - Joanna Lepicka
- INSERM US24, Proteomic platform, SFR Necker, 75015 Paris, France
| | - Giulia Barcia
- Translational Research in Neuroscience Lab, Institut Imagine, Université Paris Cité, INSERM U1163, 75015 Paris, France
- Department of Pediatric Neurosurgery Hôpital Necker, Assistance Publique Hôpitaux de Paris, 75015 Paris, France
- Department of Pediatric Neurology, Hôpital Necker, Assistance Publique Hôpitaux de Paris, 75015 Paris, France
| | - Emma Losito
- Department of Pediatric Neurosurgery Hôpital Necker, Assistance Publique Hôpitaux de Paris, 75015 Paris, France
- Department of Pediatric Neurology, Hôpital Necker, Assistance Publique Hôpitaux de Paris, 75015 Paris, France
| | - Stéphane Lourdel
- Cordeliers Research Center, INSERM, Sorbonne University, Paris Cité University, 75006 Paris, France
- CNRS EMR 8228—Laboratory of Renal Physiology and Tubulopathies, Université de Paris Cité, Centre de Recherche des Cordeliers, 75006 Paris, France
| | - Gabrielle Planelles
- Cordeliers Research Center, INSERM, Sorbonne University, Paris Cité University, 75006 Paris, France
- CNRS EMR 8228—Laboratory of Renal Physiology and Tubulopathies, Université de Paris Cité, Centre de Recherche des Cordeliers, 75006 Paris, France
| | - Raul C Muresan
- STAR-UBB Institute, Babeş-Bolyai University, 400084 Cluj-Napoca, Romania
| | - Vasile Vlad Moca
- STAR-UBB Institute, Babeş-Bolyai University, 400084 Cluj-Napoca, Romania
| | - Anna Kaminska
- Department of Pediatric Neurosurgery Hôpital Necker, Assistance Publique Hôpitaux de Paris, 75015 Paris, France
- Department of Pediatric Neurology, Hôpital Necker, Assistance Publique Hôpitaux de Paris, 75015 Paris, France
| | - Marie Bourgeois
- Department of Pediatric Neurosurgery Hôpital Necker, Assistance Publique Hôpitaux de Paris, 75015 Paris, France
- Department of Pediatric Neurology, Hôpital Necker, Assistance Publique Hôpitaux de Paris, 75015 Paris, France
| | - Nicole Chemaly
- Department of Pediatric Neurosurgery Hôpital Necker, Assistance Publique Hôpitaux de Paris, 75015 Paris, France
- Department of Pediatric Neurology, Hôpital Necker, Assistance Publique Hôpitaux de Paris, 75015 Paris, France
| | - Yasmine Rguez
- Institute of Psychiatry and Neuroscience of Paris (IPNP), Université Paris Cité, INSERM U1266, 75014 Paris, France
| | - Stéphane Auvin
- Hôpital Robert Debré, Assistance Publique Hôpitaux de Paris, 75019 Paris, France
| | - Gilles Huberfeld
- Institute of Psychiatry and Neuroscience of Paris (IPNP), Université Paris Cité, INSERM U1266, 75014 Paris, France
| | - Pascale Varlet
- Institute of Psychiatry and Neuroscience of Paris (IPNP), Université Paris Cité, INSERM U1266, 75014 Paris, France
- Service de Neuropathologie, GHU-Paris Psychiatrie et Neurosciences, Hôpital Sainte Anne, F-75014 Paris, France
| | - Vahid Asnafi
- Department of Pediatric Neurosurgery Hôpital Necker, Assistance Publique Hôpitaux de Paris, 75015 Paris, France
- Department of Pediatric Neurology, Hôpital Necker, Assistance Publique Hôpitaux de Paris, 75015 Paris, France
| | | | - Edor Kabashi
- Translational Research in Neuroscience Lab, Institut Imagine, Université Paris Cité, INSERM U1163, 75015 Paris, France
| | - Rima Nabbout
- Translational Research in Neuroscience Lab, Institut Imagine, Université Paris Cité, INSERM U1163, 75015 Paris, France
- Department of Pediatric Neurosurgery Hôpital Necker, Assistance Publique Hôpitaux de Paris, 75015 Paris, France
- Department of Pediatric Neurology, Hôpital Necker, Assistance Publique Hôpitaux de Paris, 75015 Paris, France
| | - Sorana Ciura
- Translational Research in Neuroscience Lab, Institut Imagine, Université Paris Cité, INSERM U1163, 75015 Paris, France
| | - Thomas Blauwblomme
- Translational Research in Neuroscience Lab, Institut Imagine, Université Paris Cité, INSERM U1163, 75015 Paris, France
- Department of Pediatric Neurosurgery Hôpital Necker, Assistance Publique Hôpitaux de Paris, 75015 Paris, France
- Department of Pediatric Neurology, Hôpital Necker, Assistance Publique Hôpitaux de Paris, 75015 Paris, France
| |
Collapse
|
12
|
Castagna A, Mango G, Martinelli N, Marzano L, Moruzzi S, Friso S, Pizzolo F. Sodium Chloride Cotransporter in Hypertension. Biomedicines 2024; 12:2580. [PMID: 39595146 PMCID: PMC11591633 DOI: 10.3390/biomedicines12112580] [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: 10/07/2024] [Revised: 10/29/2024] [Accepted: 11/08/2024] [Indexed: 11/28/2024] Open
Abstract
The sodium chloride cotransporter (NCC) is essential for electrolyte balance, blood pressure regulation, and pathophysiology of hypertension as it mediates the reabsorption of ultrafiltered sodium in the renal distal convoluted tubule. Given its pivotal role in the maintenance of extracellular fluid volume, the NCC is regulated by a complex network of cellular pathways, which eventually results in either its phosphorylation, enhancing sodium and chloride ion absorption from urines, or dephosphorylation and ubiquitination, which conversely decrease NCC activity. Several factors could influence NCC function, including genetic alterations, hormonal stimuli, and pharmacological treatments. The NCC's central role is also highlighted by several abnormalities resulting from genetic mutations in its gene and consequently in its structure, leading to dysregulation of blood pressure control. In the last decade, among other improvements, the acquisition of knowledge on the NCC and other renal ion channels has been favored by studies on extracellular vesicles (EVs). Dietary sodium and potassium intake are also implicated in the tuning of NCC activity. In this narrative review, we present the main cornerstones and recent evidence related to NCC control, focusing on the context of blood pressure pathophysiology, and promising new therapeutical approaches.
Collapse
Affiliation(s)
- Annalisa Castagna
- Department of Medicine, University of Verona, 37134 Verona, Italy; (A.C.); (G.M.); (N.M.); (S.F.)
| | - Gabriele Mango
- Department of Medicine, University of Verona, 37134 Verona, Italy; (A.C.); (G.M.); (N.M.); (S.F.)
| | - Nicola Martinelli
- Department of Medicine, University of Verona, 37134 Verona, Italy; (A.C.); (G.M.); (N.M.); (S.F.)
| | - Luigi Marzano
- Unit of Internal Medicine B, Department of Medicine, University of Verona School of Medicine, Azienda Ospedaliera Universitaria Integrata Verona, Policlinico “G.B. Rossi”, 37134 Verona, Italy; (L.M.); (S.M.)
| | - Sara Moruzzi
- Unit of Internal Medicine B, Department of Medicine, University of Verona School of Medicine, Azienda Ospedaliera Universitaria Integrata Verona, Policlinico “G.B. Rossi”, 37134 Verona, Italy; (L.M.); (S.M.)
| | - Simonetta Friso
- Department of Medicine, University of Verona, 37134 Verona, Italy; (A.C.); (G.M.); (N.M.); (S.F.)
| | - Francesca Pizzolo
- Department of Medicine, University of Verona, 37134 Verona, Italy; (A.C.); (G.M.); (N.M.); (S.F.)
| |
Collapse
|
13
|
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
|
14
|
Thorne CA, Grey AC, Lim JC, Donaldson PJ. The Synergistic Effects of Polyol Pathway-Induced Oxidative and Osmotic Stress in the Aetiology of Diabetic Cataracts. Int J Mol Sci 2024; 25:9042. [PMID: 39201727 PMCID: PMC11354722 DOI: 10.3390/ijms25169042] [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: 07/11/2024] [Revised: 08/13/2024] [Accepted: 08/16/2024] [Indexed: 09/03/2024] Open
Abstract
Cataracts are the world's leading cause of blindness, and diabetes is the second leading risk factor for cataracts after old age. Despite this, no preventative treatment exists for cataracts. The altered metabolism of excess glucose during hyperglycaemia is known to be the underlying cause of diabetic cataractogenesis, resulting in localised disruptions to fibre cell morphology and cell swelling in the outer cortex of the lens. In rat models of diabetic cataracts, this damage has been shown to result from osmotic stress and oxidative stress due to the accumulation of intracellular sorbitol, the depletion of NADPH which is used to regenerate glutathione, and the generation of fructose metabolites via the polyol pathway. However, differences in lens physiology and the metabolism of glucose in the lenses of different species have prevented the translation of successful treatments in animal models into effective treatments in humans. Here, we review the stresses that arise from hyperglycaemic glucose metabolism and link these to the regionally distinct metabolic and physiological adaptations in the lens that are vulnerable to these stressors, highlighting the evidence that chronic oxidative stress together with osmotic stress underlies the aetiology of human diabetic cortical cataracts. With this information, we also highlight fundamental gaps in the knowledge that could help to inform new avenues of research if effective anti-diabetic cataract therapies are to be developed in the future.
Collapse
Affiliation(s)
- Courtney A. Thorne
- Department of Physiology, School of Medical Sciences, University of Auckland, Auckland 1023, New Zealand; (C.A.T.); (A.C.G.); (P.J.D.)
- New Zealand National Eye Centre, University of Auckland, Auckland 1023, New Zealand
| | - Angus C. Grey
- Department of Physiology, School of Medical Sciences, University of Auckland, Auckland 1023, New Zealand; (C.A.T.); (A.C.G.); (P.J.D.)
- New Zealand National Eye Centre, University of Auckland, Auckland 1023, New Zealand
| | - Julie C. Lim
- Department of Physiology, School of Medical Sciences, University of Auckland, Auckland 1023, New Zealand; (C.A.T.); (A.C.G.); (P.J.D.)
- New Zealand National Eye Centre, University of Auckland, Auckland 1023, New Zealand
| | - Paul J. Donaldson
- Department of Physiology, School of Medical Sciences, University of Auckland, Auckland 1023, New Zealand; (C.A.T.); (A.C.G.); (P.J.D.)
- New Zealand National Eye Centre, University of Auckland, Auckland 1023, New Zealand
| |
Collapse
|
15
|
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
|
16
|
Rodan AR. Circadian Rhythm Regulation by Pacemaker Neuron Chloride Oscillation in Flies. Physiology (Bethesda) 2024; 39:0. [PMID: 38411570 PMCID: PMC11368518 DOI: 10.1152/physiol.00006.2024] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2024] [Revised: 02/14/2024] [Accepted: 02/15/2024] [Indexed: 02/28/2024] Open
Abstract
Circadian rhythms in physiology and behavior sync organisms to external environmental cycles. Here, circadian oscillation in intracellular chloride in central pacemaker neurons of the fly, Drosophila melanogaster, is reviewed. Intracellular chloride links SLC12 cation-coupled chloride transporter function with kinase signaling and the regulation of inwardly rectifying potassium channels.
Collapse
Affiliation(s)
- Aylin R Rodan
- Molecular Medicine Program, University of Utah, Salt Lake City, Utah, United States
- Department of Internal Medicine, Division of Nephrology and Hypertension, University of Utah, Salt Lake City, Utah, United States
- Department of Human Genetics, University of Utah, Salt Lake City, Utah, United States
- Medical Service, Veterans Affairs Salt Lake City Health Care System, Salt Lake City, Utah, United States
| |
Collapse
|
17
|
Ray S, Gurung P, Manning RS, Kravchuk AA, Singhvi A. Neuron cilia restrain glial KCC-3 to a microdomain to regulate multisensory processing. Cell Rep 2024; 43:113844. [PMID: 38421867 PMCID: PMC11296322 DOI: 10.1016/j.celrep.2024.113844] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2023] [Revised: 12/15/2023] [Accepted: 02/06/2024] [Indexed: 03/02/2024] Open
Abstract
Glia interact with multiple neurons, but it is unclear whether their interactions with each neuron are different. Our interrogation at single-cell resolution reveals that a single glial cell exhibits specificity in its interactions with different contacting neurons. Briefly, C. elegans amphid sheath (AMsh) glia apical-like domains contact 12 neuron-endings. At these ad-neuronal membranes, AMsh glia localize the K/Cl transporter KCC-3 to a microdomain exclusively around the thermosensory AFD neuron to regulate its properties. Glial KCC-3 is transported to ad-neuronal regions, where distal cilia of non-AFD glia-associated chemosensory neurons constrain it to a microdomain at AFD-contacting glial membranes. Aberrant KCC-3 localization impacts both thermosensory (AFD) and chemosensory (non-AFD) neuron properties. Thus, neurons can interact non-synaptically through a shared glial cell by regulating microdomain localization of its cues. As AMsh and glia across species compartmentalize multiple cues like KCC-3, we posit that this may be a broadly conserved glial mechanism that modulates information processing across multimodal circuits.
Collapse
Affiliation(s)
- Sneha Ray
- Division of Basic Sciences, Fred Hutchinson Cancer Center, Seattle, WA 98109, USA; Neuroscience Graduate Program, University of Washington, Seattle, WA 98195, USA
| | - Pralaksha Gurung
- Division of Basic Sciences, Fred Hutchinson Cancer Center, Seattle, WA 98109, USA; Neuroscience Graduate Program, University of Washington, Seattle, WA 98195, USA
| | - R Sean Manning
- Division of Basic Sciences, Fred Hutchinson Cancer Center, Seattle, WA 98109, USA
| | - Alexandra A Kravchuk
- Division of Basic Sciences, Fred Hutchinson Cancer Center, Seattle, WA 98109, USA; Department of Biology, University of Washington, Seattle, WA 98195, USA
| | - Aakanksha Singhvi
- Division of Basic Sciences, Fred Hutchinson Cancer Center, Seattle, WA 98109, USA; Department of Biological Structure, University of Washington School of Medicine, Seattle, WA 98195, USA.
| |
Collapse
|
18
|
Wardman JH, Andreassen SN, Toft-Bertelsen TL, Jensen MN, Wilhjelm JE, Styrishave B, Hamann S, Heegaard S, Sinclair AJ, MacAulay N. CSF hyperdynamics in rats mimicking the obesity and androgen excess characteristic of patients with idiopathic intracranial hypertension. Fluids Barriers CNS 2024; 21:10. [PMID: 38273331 PMCID: PMC10810013 DOI: 10.1186/s12987-024-00511-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2023] [Accepted: 01/05/2024] [Indexed: 01/27/2024] Open
Abstract
BACKGROUND Idiopathic intracranial hypertension (IIH) is a syndrome exhibiting elevated intracranial pressure (ICP), visual disturbances, and severe headache. IIH primarily affects young obese women, though it can occur in individuals of any age, BMI, and sex. IIH is characterized by systemic metabolic dysregulation with a profile of increased androgen hormones. However, the contribution of obesity/hormonal perturbations to cerebrospinal fluid (CSF) dynamics remains unresolved. METHODS We employed obese female Zucker rats and adjuvant testosterone to reveal IIH causal drivers. ICP and CSF dynamics were determined with in vivo experimentation and magnetic resonance imaging, testosterone levels assessed with mass spectrometry, and choroid plexus function revealed with transcriptomics. RESULTS Obese rats had undisturbed CSF testosterone levels and no changes in ICP or CSF dynamics. Adjuvant testosterone treatment of obese rats elevated the CSF secretion rate, although with no effect on the ICP, due to elevated CSF drainage capacity of these rats. CONCLUSIONS Obesity in itself therefore does not suffice to recapitulate the IIH symptoms in rats, but modulation of CSF dynamics appears with adjuvant testosterone treatment, which mimics the androgen excess observed in female IIH patients. Obesity-induced androgen dysregulation may thus contribute to the disease mechanism of IIH and could potentially serve as a future therapeutic target.
Collapse
Affiliation(s)
- Jonathan H Wardman
- Department of Neuroscience, University of Copenhagen, Blegdamsvej 3, Copenhagen, DK-2200, Denmark
| | - Søren Norge Andreassen
- Department of Neuroscience, University of Copenhagen, Blegdamsvej 3, Copenhagen, DK-2200, Denmark
| | - Trine L Toft-Bertelsen
- Department of Neuroscience, University of Copenhagen, Blegdamsvej 3, Copenhagen, DK-2200, Denmark
| | - Mette Nyholm Jensen
- Department of Neuroscience, University of Copenhagen, Blegdamsvej 3, Copenhagen, DK-2200, Denmark
| | - Jens E Wilhjelm
- Department of Neurophysiology, Rigshospitalet, Copenhagen, Denmark
- Department of Health Technology, Technical University of Denmark, Copenhagen, Denmark
| | - Bjarne Styrishave
- Department of Pharmacy, University of Copenhagen, Copenhagen, Denmark
| | - Steffen Hamann
- Department of Ophthalmology, Rigshospitalet, Copenhagen, Denmark
- Department of Clinical Medicine, University of Copenhagen, Copenhagen, Denmark
| | - Steffen Heegaard
- Department of Ophthalmology, Rigshospitalet, Copenhagen, Denmark
- Department of Clinical Medicine, University of Copenhagen, Copenhagen, Denmark
| | - Alexandra J Sinclair
- Institute of Metabolism and Systems Research, College of Medical and Dental Sciences, University of Birmingham, Birmingham, UK
| | - Nanna MacAulay
- Department of Neuroscience, University of Copenhagen, Blegdamsvej 3, Copenhagen, DK-2200, Denmark.
| |
Collapse
|
19
|
Sriperumbuduri S, Welling P, Ruzicka M, Hundemer GL, Hiremath S. Potassium and Hypertension: A State-of-the-Art Review. Am J Hypertens 2024; 37:91-100. [PMID: 37772757 DOI: 10.1093/ajh/hpad094] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2023] [Revised: 09/25/2023] [Accepted: 09/27/2023] [Indexed: 09/30/2023] Open
Abstract
Hypertension is the single most important and modifiable risk factor for cardiovascular morbidity and mortality worldwide. Non pharmacologic interventions, in particular dietary modifications have been established to decrease blood pressure (BP) and hypertension related adverse cardiovascular events. Among those dietary modifications, sodium intake restriction dominates guidelines from professional organizations and has garnered the greatest attention from the mainstream media. Despite guidelines and media exhortations, dietary sodium intake globally has not noticeably changed over recent decades. Meanwhile, increasing dietary potassium intake has remained on the sidelines, despite similar BP-lowering effects. New research reveals a potential mechanism of action, with the elucidation of its effect on natriuresis via the potassium switch effect. Additionally, potassium-substituted salt has been shown to not only reduce BP, but also reduce the risk for stroke and cardiovascular mortality. With these data, we argue that the focus on dietary modification should shift from a sodium-focused to a sodium- and potassium-focused approach with an emphasis on intervention strategies which can easily be implemented into clinical practice.
Collapse
Affiliation(s)
- Sriram Sriperumbuduri
- Division of Nephrology, University of Mississippi Medical Center, Jackson, Mississippi, USA
| | - Paul Welling
- Division of Nephrology, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
- Department of Physiology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Marcel Ruzicka
- Division of Nephrology, Department of Medicine, University of Ottawa and the Ottawa Hospital Research Institute, Ottawa, Canada
| | - Gregory L Hundemer
- Division of Nephrology, Department of Medicine, University of Ottawa and the Ottawa Hospital Research Institute, Ottawa, Canada
| | - Swapnil Hiremath
- Division of Nephrology, Department of Medicine, University of Ottawa and the Ottawa Hospital Research Institute, Ottawa, Canada
| |
Collapse
|
20
|
de Boer LL, Vanes L, Melgrati S, Biggs O'May J, Hayward D, Driscoll PC, Day J, Griffiths A, Magueta R, Morrell A, MacRae JI, Köchl R, Tybulewicz VLJ. T cell migration requires ion and water influx to regulate actin polymerization. Nat Commun 2023; 14:7844. [PMID: 38057317 PMCID: PMC10700356 DOI: 10.1038/s41467-023-43423-8] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2022] [Accepted: 11/08/2023] [Indexed: 12/08/2023] Open
Abstract
Migration of T cells is essential for their ability to mount immune responses. Chemokine-induced T cell migration requires WNK1, a kinase that regulates ion influx into the cell. However, it is not known why ion entry is necessary for T cell movement. Here we show that signaling from the chemokine receptor CCR7 leads to activation of WNK1 and its downstream pathway at the leading edge of migrating CD4+ T cells, resulting in ion influx and water entry by osmosis. We propose that WNK1-induced water entry is required to swell the membrane at the leading edge, generating space into which actin filaments can polymerize, thereby facilitating forward movement of the cell. Given the broad expression of WNK1 pathway proteins, our study suggests that ion and water influx are likely to be essential for migration in many cell types, including leukocytes and metastatic tumor cells.
Collapse
Affiliation(s)
- Leonard L de Boer
- The Francis Crick Institute, London, NW1 1AT, UK
- Department of Immunology and Inflammation, Imperial College London, London, W12 0NN, UK
- Science for Life Laboratory, Department of Women's and Children's Health, Karolinska Institutet, 171 65, Stockholm, Sweden
| | - Lesley Vanes
- The Francis Crick Institute, London, NW1 1AT, UK
| | - Serena Melgrati
- The Francis Crick Institute, London, NW1 1AT, UK
- Department of Immunology and Inflammation, Imperial College London, London, W12 0NN, UK
- Institute for Research in Biomedicine, Università della Svizzera Italiana, Bellinzona, Switzerland
| | | | - Darryl Hayward
- The Francis Crick Institute, London, NW1 1AT, UK
- GSK, Stevenage, SG1 2NY, UK
| | | | - Jason Day
- Department of Earth Sciences, University of Cambridge, Cambridge, CB2 3EQ, UK
| | - Alexander Griffiths
- London Metallomics Facility, Research Management & Innovation Directorate, King's College London, London, SE1 1UL, UK
| | - Renata Magueta
- London Metallomics Facility, Research Management & Innovation Directorate, King's College London, London, SE1 1UL, UK
| | - Alexander Morrell
- London Metallomics Facility, Research Management & Innovation Directorate, King's College London, London, SE1 1UL, UK
| | | | - Robert Köchl
- The Francis Crick Institute, London, NW1 1AT, UK
- Kings College London, London, SE1 9RT, UK
| | | |
Collapse
|
21
|
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: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [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
|
22
|
Sapio MR, King DM, Staedtler ES, Maric D, Jahanipour J, Kurochkina NA, Manalo AP, Ghetti A, Mannes AJ, Iadarola MJ. Expression pattern analysis and characterization of the hereditary sensory and autonomic neuropathy 2 A (HSAN2A) gene with no lysine kinase (WNK1) in human dorsal root ganglion. Exp Neurol 2023; 370:114552. [PMID: 37793538 DOI: 10.1016/j.expneurol.2023.114552] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2023] [Revised: 08/20/2023] [Accepted: 09/27/2023] [Indexed: 10/06/2023]
Abstract
Inherited painless neuropathies arise due to genetic insults that either block the normal signaling of or destroy the sensory afferent neurons in the dorsal root ganglion (DRG) responsible for transducing noxious stimuli. Complete loss of these neurons leads to profound insensitivity to all sensory modalities including pain. Hereditary sensory and autonomic neuropathy type 2 (HSNAII) is a rare genetic neuropathy characterized by a progressive distal early onset sensory loss. This syndrome is caused by autosomal recessive mutations in the with-no-lysine protein kinase 1 (WNK1) serine-threonine kinase gene. Of interest, disease-associated mutations are found in the large exon, termed "HSN2," which encodes a 498 amino acid domain C-terminal to the kinase domain. These mutations lead to truncation of the HSN2-containing proteins through the addition of an early stop codon (nonsense mutation) leading to loss of the C-terminal domains of this large protein. The present study evaluates the transcripts, gene structure, and protein structure of HSN2-containing WNK1 splice variants in DRG and spinal cord in order to establish the basal expression patterns of WNK1 and HSN2-containing WNK1 splice variants using multiplex fluorescent situ hybridization. We hypothesized that these transcripts would be enriched in pain-sensing DRG neurons, and, potentially, that enrichment in nociceptive neurons was responsible for the painless phenotypes observed. However, our in-depth analyses revealed that the HSN2-WNK1 splice variants were ubiquitously expressed but were not enriched in tachykinin 1-expressing C-fiber neurons, a class of neurons with a highly nociceptive character. We subsequently identified other subpopulations of DRG neurons with higher levels of HSN2-WNK1 expression, including mechanosensory large fibers. These data are inconsistent with the hypothesis that this transcript is enriched in nociceptive fibers, and instead suggest it may be related to general axon maintenance, or that nociceptive fibers are more sensitive to the genetic insult. These findings clarify the molecular and cellular expression pattern of this painless neuropathy gene in human tissue.
Collapse
Affiliation(s)
- Matthew R Sapio
- Department of Perioperative Medicine, Clinical Center, National Institutes of Health, Bethesda, MD 20892, USA
| | - Diana M King
- Department of Perioperative Medicine, Clinical Center, National Institutes of Health, Bethesda, MD 20892, USA
| | - Ellen S Staedtler
- Department of Perioperative Medicine, Clinical Center, National Institutes of Health, Bethesda, MD 20892, USA
| | - Dragan Maric
- National Institute of Neurological Disorders and Stroke, Flow and Imaging Cytometry Core Facility, Bethesda, MD 20892, USA
| | - Jahandar Jahanipour
- National Institute of Neurological Disorders and Stroke, Flow and Imaging Cytometry Core Facility, Bethesda, MD 20892, USA
| | | | - Allison P Manalo
- Department of Perioperative Medicine, Clinical Center, National Institutes of Health, Bethesda, MD 20892, USA
| | | | - Andrew J Mannes
- Department of Perioperative Medicine, Clinical Center, National Institutes of Health, Bethesda, MD 20892, USA
| | - Michael J Iadarola
- Department of Perioperative Medicine, Clinical Center, National Institutes of Health, Bethesda, MD 20892, USA.
| |
Collapse
|
23
|
Lashhab R, Essuman G, Chavez-Canales M, Alexander RT, Cordat E. Expression of the kidney anion exchanger 1 affects WNK4 and SPAK phosphorylation and results in claudin-4 phosphorylation. Heliyon 2023; 9:e22280. [PMID: 38034706 PMCID: PMC10687047 DOI: 10.1016/j.heliyon.2023.e22280] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2023] [Revised: 11/07/2023] [Accepted: 11/08/2023] [Indexed: 12/02/2023] Open
Abstract
In the renal collecting ducts, chloride reabsorption occurs through both transcellular and paracellular pathways. Recent literature highlights a functional interplay between both pathways. We recently showed that in polarized inner medullary collecting duct cells, expression of the basolateral kidney anion exchanger 1 (kAE1) results in a decreased transepithelial electrical resistance (TEER), in a claudin-4 dependent pathway. Claudin-4 is a paracellular sodium blocker and chloride pore. Here, we show that kAE1 expression in mouse inner medullary collecting duct cells triggers WNK4, SPAK and claudin-4 phosphorylation. Expression of a functionally dead kAE1 E681Q mutant has no effect on phosphorylation of these proteins. Expression of a catalytically inactive WNK4 D321A or chloride-insensitive WNK4 L319F mutant abolishes kAE1 effect on TEER, supporting a contribution of WNK4 to the process. We propose that variations of the cytosolic pH and chloride concentration upon kAE1 expression alter WNK4 kinase activity and tight junction properties.
Collapse
Affiliation(s)
- Rawad Lashhab
- Department of Physiology and Membrane Protein Disease Research Group, University of Alberta, Edmonton, Alberta, Canada
| | - Grace Essuman
- Department of Physiology and Membrane Protein Disease Research Group, University of Alberta, Edmonton, Alberta, Canada
| | - Maria Chavez-Canales
- Unidad de Investigación UNAM-INCICh, Instituto Nacional de Cardiología Ignacio Chávez and Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México, Tlalpan, Mexico City, 14080, Mexico
| | - R. Todd Alexander
- Department of Physiology and Membrane Protein Disease Research Group, University of Alberta, Edmonton, Alberta, Canada
- Department of Pediatrics, University of Alberta, Edmonton, Alberta, Canada
| | - Emmanuelle Cordat
- Department of Physiology and Membrane Protein Disease Research Group, University of Alberta, Edmonton, Alberta, Canada
| |
Collapse
|
24
|
Yarikipati P, Jonusaite S, Pleinis JM, Dominicci Cotto C, Sanchez-Hernandez D, Morrison DE, Goyal S, Schellinger J, Pénalva C, Curtiss J, Rodan AR, Jenny A. Unanticipated domain requirements for Drosophila Wnk kinase in vivo. PLoS Genet 2023; 19:e1010975. [PMID: 37819975 PMCID: PMC10593226 DOI: 10.1371/journal.pgen.1010975] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2022] [Revised: 10/23/2023] [Accepted: 09/14/2023] [Indexed: 10/13/2023] Open
Abstract
WNK (With no Lysine [K]) kinases have critical roles in the maintenance of ion homeostasis and the regulation of cell volume. Their overactivation leads to pseudohypoaldosteronism type II (Gordon syndrome) characterized by hyperkalemia and high blood pressure. More recently, WNK family members have been shown to be required for the development of the nervous system in mice, zebrafish, and flies, and the cardiovascular system of mice and fish. Furthermore, human WNK2 and Drosophila Wnk modulate canonical Wnt signaling. In addition to a well-conserved kinase domain, animal WNKs have a large, poorly conserved C-terminal domain whose function has been largely mysterious. In most but not all cases, WNKs bind and activate downstream kinases OSR1/SPAK, which in turn regulate the activity of various ion transporters and channels. Here, we show that Drosophila Wnk regulates Wnt signaling and cell size during the development of the wing in a manner dependent on Fray, the fly homolog of OSR1/SPAK. We show that the only canonical RF(X)V/I motif of Wnk, thought to be essential for WNK interactions with OSR1/SPAK, is required to interact with Fray in vitro. However, this motif is unexpectedly dispensable for Fray-dependent Wnk functions in vivo during fly development and fluid secretion in the Malpighian (renal) tubules. In contrast, a structure function analysis of Wnk revealed that the less-conserved C-terminus of Wnk, that recently has been shown to promote phase transitions in cell culture, is required for viability in vivo. Our data thus provide novel insights into unexpected in vivo roles of specific WNK domains.
Collapse
Affiliation(s)
- Prathibha Yarikipati
- Department of Developmental and Molecular Biology, Albert Einstein College of Medicine, New York, United States of America
| | - Sima Jonusaite
- Molecular Medicine Program, University of Utah, Salt Lake City, Utah, United States of America
| | - John M. Pleinis
- Molecular Medicine Program, University of Utah, Salt Lake City, Utah, United States of America
| | - Carihann Dominicci Cotto
- Department of Developmental and Molecular Biology, Albert Einstein College of Medicine, New York, United States of America
| | - David Sanchez-Hernandez
- Department of Developmental and Molecular Biology, Albert Einstein College of Medicine, New York, United States of America
| | - Daryl E. Morrison
- Molecular Medicine Program, University of Utah, Salt Lake City, Utah, United States of America
| | - Suhani Goyal
- Department of Internal Medicine, Division of Nephrology, University of Texas Southwestern, Dallas, Texas, United States of America
| | - Jeffrey Schellinger
- Department of Internal Medicine, Division of Nephrology, University of Texas Southwestern, Dallas, Texas, United States of America
| | - Clothilde Pénalva
- Molecular Medicine Program, University of Utah, Salt Lake City, Utah, United States of America
| | - Jennifer Curtiss
- Department of Cell & Developmental Biology, New Mexico State University, Las Cruces, New Mexico, United States of America
| | - Aylin R. Rodan
- Molecular Medicine Program, University of Utah, Salt Lake City, Utah, United States of America
- Department of Internal Medicine, Division of Nephrology and Hypertension, University of Utah, Salt Lake City, Utah, United States of America
- Department of Human Genetics, University of Utah, Salt Lake City, Utah, United States of America
- Medical Service, Veterans Affairs Salt Lake City Health Care System, Salt Lake City, Utah, United States of America
| | - Andreas Jenny
- Department of Developmental and Molecular Biology, Albert Einstein College of Medicine, New York, United States of America
- Department of Genetics, Albert Einstein College of Medicine, New York, New York, United States of America
| |
Collapse
|
25
|
Wardman JH, Jensen MN, Andreassen SN, Styrishave B, Wilhjelm JE, Sinclair AJ, MacAulay N. Modelling idiopathic intracranial hypertension in rats: contributions of high fat diet and testosterone to intracranial pressure and cerebrospinal fluid production. Fluids Barriers CNS 2023; 20:44. [PMID: 37328884 DOI: 10.1186/s12987-023-00436-1] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2023] [Accepted: 04/29/2023] [Indexed: 06/18/2023] Open
Abstract
BACKGROUND Idiopathic intracranial hypertension (IIH) is a condition characterized by increased intracranial pressure (ICP), impaired vision, and headache. Most cases of IIH occur in obese women of childbearing age, though age, BMI, and female sex do not encompass all aspects of IIH pathophysiology. Systemic metabolic dysregulation has been identified in IIH with a profile of androgen excess. However, the mechanistic coupling between obesity/hormonal perturbations and cerebrospinal fluid dynamics remains unresolved. METHODS Female Wistar rats were either fed a high fat diet (HFD) for 21 weeks or exposed to adjuvant testosterone treatment for 28 days to recapitulate IIH causal drivers. Cerebrospinal fluid (CSF) and blood testosterone levels were determined with mass spectrometry, ICP and CSF dynamics with in vivo experimentation, and the choroid plexus function revealed with transcriptomics and ex vivo isotope-based flux assays. RESULTS HFD-fed rats presented with increased ICP (65%), which was accompanied by increased CSF outflow resistance (50%) without altered CSF secretion rate or choroid plexus gene expression. Chronic adjuvant testosterone treatment of lean rats caused elevated ICP (55%) and CSF secretion rate (85%), in association with increased activity of the choroid plexus Na+,K+,2Cl- cotransporter, NKCC1. CONCLUSIONS HFD-induced ICP elevation in experimental rats occurred with decreased CSF drainage capacity. Adjuvant testosterone, mimicking the androgen excess observed in female IIH patients, elevated the CSF secretion rate and thus ICP. Obesity-induced androgen dysregulation may thus contribute to the disease mechanism of IIH.
Collapse
Affiliation(s)
- Jonathan H Wardman
- Department of Neuroscience, Faculty of Health and Medical Sciences, University of Copenhagen, Blegdamsvej 3, 2200, Copenhagen N, Denmark
| | - Mette N Jensen
- Department of Neuroscience, Faculty of Health and Medical Sciences, University of Copenhagen, Blegdamsvej 3, 2200, Copenhagen N, Denmark
| | - Søren N Andreassen
- Department of Neuroscience, Faculty of Health and Medical Sciences, University of Copenhagen, Blegdamsvej 3, 2200, Copenhagen N, Denmark
| | - Bjarne Styrishave
- Department of Pharmacy, University of Copenhagen, Copenhagen, Denmark
| | - Jens E Wilhjelm
- Department of Health Technology, The Technical University of Denmark, Kgs. Lyngby, Denmark
| | - Alexandra J Sinclair
- Institute of Metabolism and Systems Research, University of Birmingham, Birmingham, UK
| | - Nanna MacAulay
- Department of Neuroscience, Faculty of Health and Medical Sciences, University of Copenhagen, Blegdamsvej 3, 2200, Copenhagen N, Denmark.
| |
Collapse
|
26
|
Yan L, Yun-Lin L, Yong-Ling L, Wei-Wei Z, Yue-Shan P. Alteration of GABAergic neurons and abnormality of NKCC1/KCC2 in focal cortical dysplasia (FCD) type Ⅱ lesions. Epilepsy Res 2023; 194:107180. [PMID: 37352729 DOI: 10.1016/j.eplepsyres.2023.107180] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2022] [Revised: 05/09/2023] [Accepted: 06/13/2023] [Indexed: 06/25/2023]
Abstract
BACKGROUND The current conclusions of molecular genetics still cannot satisfactorily explain the pathogenesis of focal cortical dysplasia (FCD) and the reason for drug resistance. The interneurons of GABA deserve attention. To observe the distribution and changes of GABAergic neurons and to explore the expression of cation chloride cotransporter NKCC1/KCC2 in focal cortical dysplasia (FCD) type II lesions is a highly significant job. METHODS The expressions of GAD67(a marker of active GABAergic neuron), NKCC1 and KCC2 were detected by immunohistochemistry and immunohistochemistry double staining in 10 cases of FCD Ⅱa and 10 cases of FCD Ⅱb. The number of GAD67 positive neurons was counted, and the average absorbance (IA) of NKCC1 positive expression was measured, using Image Pro-Plus7.0 software. The data were statistically analyzed. RESULTS The density of GABAergic neuron in focal dysplastic regions was significantly lower than that in the histologically "normal" cerebral cortex, regions from the same specimen (p < 0.0001, t-test). Compared to the NKCC1 staining intensity of neurons in the control group (measuring 1000 cells each), the IA value of dysmorphic neurons was significantly increased (p < 0.05, t'-test Cochran & Cox method). Intracytoplasmic concentration of KCC2 was evident in dysmorphic neurons but not in the other mature neurons. Most of the balloon cells were negative for NKCC1, except for few balloon cells showing sparse colored particles. The expression of KCC2 was negative in all balloon cells. CONCLUSIONS The changes in the expression of NKCC1 and KCC2 may indicate that dysmorphic neurons were in a state similar to that of immature neurons. This state may be related to the abnormal electrophysiology of epilepsy. The difference between the number of GAD67 positive cells in the lesion site and the remote site of the same case may be an evaluation index of the effectiveness of surgery.
Collapse
Affiliation(s)
- Li Yan
- Department of Pathology, Haidian Hospital, Haidian District of Peking University Third Hospital, Beijing, China
| | - Li Yun-Lin
- Department of Neurosurgery, Children's Hospital, Capital Institute of Pediatrics, Beijing, China
| | - Liu Yong-Ling
- Department of Pathology, Haidian Hospital, Haidian District of Peking University Third Hospital, Beijing, China
| | - Zhang Wei-Wei
- Department of Pathology, Beijing Tiantan Hospital Affiliated to Capital Medical University, Beijing, China
| | - Piao Yue-Shan
- Department of Pathology, Xuanwu Hospital, Capital Medical University, Beijing 100053, China; Clinical Research Center for Epilepsy, Capital Medical University, Beijing 100053, China; National Center for Neurological Disorders, Beijing 100053, China.
| |
Collapse
|
27
|
Zigo M, Kerns K, Sutovsky P. The Ubiquitin-Proteasome System Participates in Sperm Surface Subproteome Remodeling during Boar Sperm Capacitation. Biomolecules 2023; 13:996. [PMID: 37371576 PMCID: PMC10296210 DOI: 10.3390/biom13060996] [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: 05/01/2023] [Revised: 06/08/2023] [Accepted: 06/12/2023] [Indexed: 06/29/2023] Open
Abstract
Sperm capacitation is a complex process endowing biological and biochemical changes to a spermatozoon for a successful encounter with an oocyte. The present study focused on the role of the ubiquitin-proteasome system (UPS) in the remodeling of the sperm surface subproteome. The sperm surface subproteome from non-capacitated and in vitro capacitated (IVC) porcine spermatozoa, with and without proteasomal inhibition, was selectively isolated. The purified sperm surface subproteome was analyzed using high-resolution, quantitative liquid chromatography-mass spectrometry (LC-MS) in four replicates. We identified 1680 HUGO annotated proteins, out of which we found 91 to be at least 1.5× less abundant (p < 0.05) and 141 to be at least 1.5× more abundant (p < 0.05) on the surface of IVC spermatozoa. These proteins were associated with sperm capacitation, hyperactivation, metabolism, acrosomal exocytosis, and fertilization. Abundances of 14 proteins were found to be significantly different (p < 0.05), exceeding a 1.5-fold abundance between the proteasomally inhibited (100 µM MG132) and vehicle control (0.2% ethanol) groups. The proteins NIF3L1, CSE1L, NDUFB7, PGLS, PPP4C, STK39, and TPRG1L were found to be more abundant; while BPHL, GSN, GSPT1, PFDN4, STYXL1, TIMM10, and UBXN4 were found to be less abundant in proteasomally inhibited IVC spermatozoa. Despite the UPS having a narrow range of targets, it modulated sperm metabolism and binding by regulating susceptible surface proteins. Changes in CSE1L, PFDN4, and STK39 during in vitro capacitation were confirmed using immunocytochemistry, image-based flow cytometry, and Western blotting. The results confirmed the active participation of the UPS in the extensive sperm surface proteome remodeling that occurs during boar sperm capacitation. This work will help us to identify new pharmacological mechanisms to positively or negatively modulate sperm fertilizing ability in food animals and humans.
Collapse
Affiliation(s)
- Michal Zigo
- Division of Animal Science, University of Missouri, Columbia, MO 65211, USA;
| | - Karl Kerns
- Division of Animal Science, University of Missouri, Columbia, MO 65211, USA;
- Department of Animal Science, Iowa State University, Ames, IA 50011, USA;
| | - Peter Sutovsky
- Department of Animal Science, Iowa State University, Ames, IA 50011, USA;
- Department of Obstetrics, Gynecology and Women’s Health, University of Missouri, Columbia, MO 65211, USA
| |
Collapse
|
28
|
Paez-Gonzalez P, Lopez-de-San-Sebastian J, Ceron-Funez R, Jimenez AJ, Rodríguez-Perez LM. Therapeutic strategies to recover ependymal barrier after inflammatory damage: relevance for recovering neurogenesis during development. Front Neurosci 2023; 17:1204197. [PMID: 37397456 PMCID: PMC10308384 DOI: 10.3389/fnins.2023.1204197] [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: 04/11/2023] [Accepted: 05/22/2023] [Indexed: 07/04/2023] Open
Abstract
The epithelium covering the surfaces of the cerebral ventricular system is known as the ependyma, and is essential for maintaining the physical and functional integrity of the central nervous system. Additionally, the ependyma plays an essential role in neurogenesis, neuroinflammatory modulation and neurodegenerative diseases. Ependyma barrier is severely affected by perinatal hemorrhages and infections that cross the blood brain barrier. The recovery and regeneration of ependyma after damage are key to stabilizing neuroinflammatory and neurodegenerative processes that are critical during early postnatal ages. Unfortunately, there are no effective therapies to regenerate this tissue in human patients. Here, the roles of the ependymal barrier in the context of neurogenesis and homeostasis are reviewed, and future research lines for development of actual therapeutic strategies are discussed.
Collapse
Affiliation(s)
- Patricia Paez-Gonzalez
- Department of Cell Biology, Genetics and Physiology, University of Malaga, Málaga, Spain
- Instituto de Investigación Biomédica de Málaga y Plataforma en Nanomedicina-IBIMA Plataforma BIONAND, Málaga, Spain
| | | | - Raquel Ceron-Funez
- Department of Cell Biology, Genetics and Physiology, University of Malaga, Málaga, Spain
| | - Antonio J. Jimenez
- Instituto de Investigación Biomédica de Málaga y Plataforma en Nanomedicina-IBIMA Plataforma BIONAND, Málaga, Spain
| | - Luis Manuel Rodríguez-Perez
- Department of Cell Biology, Genetics and Physiology, University of Malaga, Málaga, Spain
- Instituto de Investigación Biomédica de Málaga y Plataforma en Nanomedicina-IBIMA Plataforma BIONAND, Málaga, Spain
- Department of Human Physiology, Human Histology, Pathological Anatomy and Sports, University of Malaga, Málaga, Spain
| |
Collapse
|
29
|
Pressey JC, de Saint-Rome M, Raveendran VA, Woodin MA. Chloride transporters controlling neuronal excitability. Physiol Rev 2023; 103:1095-1135. [PMID: 36302178 DOI: 10.1152/physrev.00025.2021] [Citation(s) in RCA: 32] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023] Open
Abstract
Synaptic inhibition plays a crucial role in regulating neuronal excitability, which is the foundation of nervous system function. This inhibition is largely mediated by the neurotransmitters GABA and glycine that activate Cl--permeable ion channels, which means that the strength of inhibition depends on the Cl- gradient across the membrane. In neurons, the Cl- gradient is primarily mediated by two secondarily active cation-chloride cotransporters (CCCs), NKCC1 and KCC2. CCC-mediated regulation of the neuronal Cl- gradient is critical for healthy brain function, as dysregulation of CCCs has emerged as a key mechanism underlying neurological disorders including epilepsy, neuropathic pain, and autism spectrum disorder. This review begins with an overview of neuronal chloride transporters before explaining the dependent relationship between these CCCs, Cl- regulation, and inhibitory synaptic transmission. We then discuss the evidence for how CCCs can be regulated, including by activity and their protein interactions, which underlie inhibitory synaptic plasticity. For readers who may be interested in conducting experiments on CCCs and neuronal excitability, we have included a section on techniques for estimating and recording intracellular Cl-, including their advantages and limitations. Although the focus of this review is on neurons, we also examine how Cl- is regulated in glial cells, which in turn regulate neuronal excitability through the tight relationship between this nonneuronal cell type and synapses. Finally, we discuss the relatively extensive and growing literature on how CCC-mediated neuronal excitability contributes to neurological disorders.
Collapse
Affiliation(s)
- Jessica C Pressey
- Department of Cell and Systems Biology, University of Toronto, Toronto, Ontario, Canada
| | - Miranda de Saint-Rome
- Department of Cell and Systems Biology, University of Toronto, Toronto, Ontario, Canada
| | - Vineeth A Raveendran
- Department of Cell and Systems Biology, University of Toronto, Toronto, Ontario, Canada
| | - Melanie A Woodin
- Department of Cell and Systems Biology, University of Toronto, Toronto, Ontario, Canada
| |
Collapse
|
30
|
Ray S, Gurung P, Manning RS, Kravchuk A, Singhvi A. Neuron cilia constrain glial regulators to microdomains around distal neurons. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.03.18.533255. [PMID: 36993507 PMCID: PMC10055228 DOI: 10.1101/2023.03.18.533255] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 06/19/2023]
Abstract
Each glia interacts with multiple neurons, but the fundamental logic of whether it interacts with all equally remains unclear. We find that a single sense-organ glia modulates different contacting neurons distinctly. To do so, it partitions regulatory cues into molecular microdomains at specific neuron contact-sites, at its delimited apical membrane. For one glial cue, K/Cl transporter KCC-3, microdomain-localization occurs through a two-step, neuron-dependent process. First, KCC-3 shuttles to glial apical membranes. Second, some contacting neuron cilia repel it, rendering it microdomain-localized around one distal neuron-ending. KCC-3 localization tracks animal aging, and while apical localization is sufficient for contacting neuron function, microdomain-restriction is required for distal neuron properties. Finally, we find the glia regulates its microdomains largely independently. Together, this uncovers that glia modulate cross-modal sensor processing by compartmentalizing regulatory cues into microdomains. Glia across species contact multiple neurons and localize disease-relevant cues like KCC-3. Thus, analogous compartmentalization may broadly drive how glia regulate information processing across neural circuits.
Collapse
Affiliation(s)
- Sneha Ray
- Division of Basic Sciences, Fred Hutchinson Cancer Research Center, Seattle, WA 98109
- Neuroscience Graduate Program, University of Washington, Seattle, WA
| | - Pralaksha Gurung
- Neuroscience Graduate Program, University of Washington, Seattle, WA
| | - R. Sean Manning
- Division of Basic Sciences, Fred Hutchinson Cancer Research Center, Seattle, WA 98109
| | - Alexandra Kravchuk
- Division of Basic Sciences, Fred Hutchinson Cancer Research Center, Seattle, WA 98109
- University of Washington School of Medicine, WA 98195
| | - Aakanksha Singhvi
- Division of Basic Sciences, Fred Hutchinson Cancer Research Center, Seattle, WA 98109
- Department of Biological Structure, University of Washington School of Medicine, WA 98195
| |
Collapse
|
31
|
Moreno E, Pacheco-Alvarez D, Chávez-Canales M, Elizalde S, Leyva-Ríos K, Gamba G. Structure-function relationships in the sodium chloride cotransporter. Front Physiol 2023; 14:1118706. [PMID: 36998989 PMCID: PMC10043231 DOI: 10.3389/fphys.2023.1118706] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2022] [Accepted: 02/27/2023] [Indexed: 03/16/2023] Open
Abstract
The thiazide sensitive Na+:Cl− cotransporter (NCC) is the principal via for salt reabsorption in the apical membrane of the distal convoluted tubule (DCT) in mammals and plays a fundamental role in managing blood pressure. The cotransporter is targeted by thiazide diuretics, a highly prescribed medication that is effective in treating arterial hypertension and edema. NCC was the first member of the electroneutral cation-coupled chloride cotransporter family to be identified at a molecular level. It was cloned from the urinary bladder of the Pseudopleuronectes americanus (winter flounder) 30 years ago. The structural topology, kinetic and pharmacology properties of NCC have been extensively studied, determining that the transmembrane domain (TM) coordinates ion and thiazide binding. Functional and mutational studies have discovered residues involved in the phosphorylation and glycosylation of NCC, particularly on the N-terminal domain, as well as the extracellular loop connected to TM7-8 (EL7-8). In the last decade, single-particle cryogenic electron microscopy (cryo-EM) has permitted the visualization of structures at high atomic resolution for six members of the SLC12 family (NCC, NKCC1, KCC1-KCC4). Cryo-EM insights of NCC confirm an inverted conformation of the TM1-5 and TM6-10 regions, a characteristic also found in the amino acid-polyamine-organocation (APC) superfamily, in which TM1 and TM6 clearly coordinate ion binding. The high-resolution structure also displays two glycosylation sites (N-406 and N-426) in EL7-8 that are essential for NCC expression and function. In this review, we briefly describe the studies related to the structure-function relationship of NCC, beginning with the first biochemical/functional studies up to the recent cryo-EM structure obtained, to acquire an overall view enriched with the structural and functional aspects of the cotransporter.
Collapse
Affiliation(s)
- Erika Moreno
- Department of Nephrology and Mineral Metabolism, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Mexico City, Mexico
| | | | - María Chávez-Canales
- Unidad de Investigación UNAM-INC, Instituto Nacional de Cardiología Ignacio Chávez and Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México, Mexico City, Mexico
| | - Stephanie Elizalde
- Department of Nephrology and Mineral Metabolism, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Mexico City, Mexico
| | - Karla Leyva-Ríos
- Escuela de Medicina, Universidad Panamericana, Mexico City, Mexico
| | - Gerardo Gamba
- Department of Nephrology and Mineral Metabolism, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Mexico City, Mexico
- Molecular Phisiology Unit, Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México, Mexico City, Mexico
- *Correspondence: Gerardo Gamba,
| |
Collapse
|
32
|
Hayward DA, Vanes L, Wissmann S, Sivapatham S, Hartweger H, Biggs O’May J, de Boer LL, Mitter R, Köchl R, Stein JV, Tybulewicz VL. B cell-intrinsic requirement for WNK1 kinase in antibody responses in mice. J Exp Med 2023; 220:e20211827. [PMID: 36662229 PMCID: PMC9872328 DOI: 10.1084/jem.20211827] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2021] [Revised: 11/20/2022] [Accepted: 12/23/2022] [Indexed: 01/21/2023] Open
Abstract
Migration and adhesion play critical roles in B cells, regulating recirculation between lymphoid organs, migration within lymphoid tissue, and interaction with CD4+ T cells. However, there is limited knowledge of how B cells integrate chemokine receptor and integrin signaling with B cell activation to generate efficient humoral responses. Here, we show that the WNK1 kinase, a regulator of migration and adhesion, is essential in B cells for T-dependent and -independent antibody responses. We demonstrate that WNK1 transduces signals from the BCR, CXCR5, and CD40, and using intravital imaging, we show that WNK1 regulates migration of naive and activated B cells, and their interactions with T cells. Unexpectedly, we show that WNK1 is required for BCR- and CD40-induced proliferation, acting through the OXSR1 and STK39 kinases, and for efficient B cell-T cell collaboration in vivo. Thus, WNK1 is critical for humoral immune responses, by regulating B cell migration, adhesion, and T cell-dependent activation.
Collapse
Affiliation(s)
| | | | - Stefanie Wissmann
- Department of Oncology, Microbiology and Immunology, University of Fribourg, Fribourg, Switzerland
| | - Sujana Sivapatham
- Department of Oncology, Microbiology and Immunology, University of Fribourg, Fribourg, Switzerland
| | | | | | | | | | | | - Jens V. Stein
- Department of Oncology, Microbiology and Immunology, University of Fribourg, Fribourg, Switzerland
| | | |
Collapse
|
33
|
Stangherlin A. Ion dynamics and the regulation of circadian cellular physiology. Am J Physiol Cell Physiol 2023; 324:C632-C643. [PMID: 36689675 DOI: 10.1152/ajpcell.00378.2022] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023]
Abstract
Circadian rhythms in physiology and behavior allow organisms to anticipate the daily environmental changes imposed by the rotation of our planet around its axis. Although these rhythms eventually manifest at the organismal level, a cellular basis for circadian rhythms has been demonstrated. Significant contributors to these cell-autonomous rhythms are daily cycles in gene expression and protein translation. However, recent data revealed cellular rhythms in other biological processes, including ionic currents, ion transport, and cytosolic ion abundance. Circadian rhythms in ion currents sustain circadian variation in action potential firing rate, which coordinates neuronal behavior and activity. Circadian regulation of metal ions abundance and dynamics is implicated in distinct cellular processes, from protein translation to membrane activity and osmotic homeostasis. In turn, studies showed that manipulating ion abundance affects the expression of core clock genes and proteins, suggestive of a close interplay. However, the relationship between gene expression cycles, ion dynamics, and cellular function is still poorly characterized. In this review, I will discuss the mechanisms that generate ion rhythms, the cellular functions they govern, and how they feed back to regulate the core clock machinery.
Collapse
Affiliation(s)
- Alessandra Stangherlin
- Faculty of Medicine and University Hospital Cologne, Cluster of Excellence Cellular Stress Responses in Aging-associated Diseases (CECAD), Institute for Mitochondrial Diseases and Ageing, University of Cologne, Cologne, Germany
| |
Collapse
|
34
|
Robert SM, Reeves BC, Kiziltug E, Duy PQ, Karimy JK, Mansuri MS, Marlier A, Allington G, Greenberg ABW, DeSpenza T, Singh AK, Zeng X, Mekbib KY, Kundishora AJ, Nelson-Williams C, Hao LT, Zhang J, Lam TT, Wilson R, Butler WE, Diluna ML, Feinberg P, Schafer DP, Movahedi K, Tannenbaum A, Koundal S, Chen X, Benveniste H, Limbrick DD, Schiff SJ, Carter BS, Gunel M, Simard JM, Lifton RP, Alper SL, Delpire E, Kahle KT. The choroid plexus links innate immunity to CSF dysregulation in hydrocephalus. Cell 2023; 186:764-785.e21. [PMID: 36803604 PMCID: PMC10069664 DOI: 10.1016/j.cell.2023.01.017] [Citation(s) in RCA: 67] [Impact Index Per Article: 33.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/01/2022] [Revised: 09/26/2022] [Accepted: 01/12/2023] [Indexed: 02/18/2023]
Abstract
The choroid plexus (ChP) is the blood-cerebrospinal fluid (CSF) barrier and the primary source of CSF. Acquired hydrocephalus, caused by brain infection or hemorrhage, lacks drug treatments due to obscure pathobiology. Our integrated, multi-omic investigation of post-infectious hydrocephalus (PIH) and post-hemorrhagic hydrocephalus (PHH) models revealed that lipopolysaccharide and blood breakdown products trigger highly similar TLR4-dependent immune responses at the ChP-CSF interface. The resulting CSF "cytokine storm", elicited from peripherally derived and border-associated ChP macrophages, causes increased CSF production from ChP epithelial cells via phospho-activation of the TNF-receptor-associated kinase SPAK, which serves as a regulatory scaffold of a multi-ion transporter protein complex. Genetic or pharmacological immunomodulation prevents PIH and PHH by antagonizing SPAK-dependent CSF hypersecretion. These results reveal the ChP as a dynamic, cellularly heterogeneous tissue with highly regulated immune-secretory capacity, expand our understanding of ChP immune-epithelial cell cross talk, and reframe PIH and PHH as related neuroimmune disorders vulnerable to small molecule pharmacotherapy.
Collapse
Affiliation(s)
- Stephanie M Robert
- Department of Neurosurgery, Yale School of Medicine, New Haven, CT 06520, USA
| | - Benjamin C Reeves
- Department of Neurosurgery, Yale School of Medicine, New Haven, CT 06520, USA
| | - Emre Kiziltug
- Department of Neurosurgery, Yale School of Medicine, New Haven, CT 06520, USA
| | - Phan Q Duy
- Department of Neurosurgery, Yale School of Medicine, New Haven, CT 06520, USA
| | - Jason K Karimy
- Department of Neurosurgery, Yale School of Medicine, New Haven, CT 06520, USA
| | - M Shahid Mansuri
- Department of Neurosurgery, Yale School of Medicine, New Haven, CT 06520, USA; Department of Molecular Biophysics and Biochemistry, Yale University School of Medicine, New Haven, CT 06520, USA
| | - Arnaud Marlier
- Department of Neurosurgery, Yale School of Medicine, New Haven, CT 06520, USA
| | - Garrett Allington
- Department of Neurosurgery, Yale School of Medicine, New Haven, CT 06520, USA; Department of Pathology, Yale School of Medicine, New Haven, CT 06520, USA
| | - Ana B W Greenberg
- Department of Neurosurgery, Yale School of Medicine, New Haven, CT 06520, USA
| | - Tyrone DeSpenza
- Department of Neurosurgery, Yale School of Medicine, New Haven, CT 06520, USA
| | - Amrita K Singh
- Department of Neurosurgery, Yale School of Medicine, New Haven, CT 06520, USA
| | - Xue Zeng
- Department of Neurosurgery, Yale School of Medicine, New Haven, CT 06520, USA
| | - Kedous Y Mekbib
- Department of Neurosurgery, Yale School of Medicine, New Haven, CT 06520, USA
| | - Adam J Kundishora
- Department of Neurosurgery, Yale School of Medicine, New Haven, CT 06520, USA
| | | | - Le Thi Hao
- Department of Neurosurgery, Yale School of Medicine, New Haven, CT 06520, USA
| | - Jinwei Zhang
- Institute of Biomedical and Clinical Sciences, University of Exeter Medical School, Hatherly Laboratory, Exeter EX1 2LU, UK
| | - TuKiet T Lam
- Department of Molecular Biophysics and Biochemistry, Yale University School of Medicine, New Haven, CT 06520, USA; Keck MS & Proteomics Resource, Yale University School of Medicine, New Haven, CT 06520, USA
| | - Rashaun Wilson
- Department of Molecular Biophysics and Biochemistry, Yale University School of Medicine, New Haven, CT 06520, USA; Keck MS & Proteomics Resource, Yale University School of Medicine, New Haven, CT 06520, USA
| | - William E Butler
- Department of Neurosurgery, Massachusetts General Hospital and Harvard Medical School, Boston, MA 02114, USA
| | - Michael L Diluna
- Department of Neurosurgery, Yale School of Medicine, New Haven, CT 06520, USA
| | - Philip Feinberg
- Department of Neurobiology, Brudnick Neuropsychiatric Research Institute University of Massachusetts Chan Medical School, Worcester, MA 01655, USA; Medical Scientist Training Program, UMass Chan Medical School, Worcester, MA 01655, USA
| | - Dorothy P Schafer
- Department of Neurobiology, Brudnick Neuropsychiatric Research Institute University of Massachusetts Chan Medical School, Worcester, MA 01655, USA
| | - Kiavash Movahedi
- Laboratory of Cellular and Molecular Immunology, Vrije Universiteit Brussel, 1050 Brussels, Belgium; Myeloid Cell Immunology Laboratory, VIB Center for Inflammation Research, 1050 Brussels, Belgium
| | - Allen Tannenbaum
- Department of Applied Mathematics and Statistics, Stony Brook University, Stony Brook, NY 11794, USA; Department of Medical Physics, Memorial Sloan Kettering Cancer Center, New York City, NY 11794, USA
| | - Sunil Koundal
- Department of Anesthesiology, Yale School of Medicine, New Haven, CT 06520, USA
| | - Xinan Chen
- Department of Applied Mathematics and Statistics, Stony Brook University, Stony Brook, NY 11794, USA
| | - Helene Benveniste
- Department of Anesthesiology, Yale School of Medicine, New Haven, CT 06520, USA
| | - David D Limbrick
- Department of Neurosurgery, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Steven J Schiff
- Department of Neurosurgery, Yale School of Medicine, New Haven, CT 06520, USA
| | - Bob S Carter
- Department of Neurosurgery, Massachusetts General Hospital and Harvard Medical School, Boston, MA 02114, USA
| | - Murat Gunel
- Department of Neurosurgery, Yale School of Medicine, New Haven, CT 06520, USA
| | - J Marc Simard
- Department of Neurosurgery, University of Maryland, School of Medicine, Baltimore, MD 21201, USA; Department of Pathology, University of Maryland, School of Medicine, Baltimore, MD 21201, USA; Department of Physiology, University of Maryland, School of Medicine, Baltimore, MD 21201, USA
| | - Richard P Lifton
- Laboratory of Human Genetics and Genomics, the Rockefeller University, New York, NY 10065, USA
| | - Seth L Alper
- Department of Medicine, Harvard Medical School, Boston, MA 02115, USA
| | - Eric Delpire
- Department of Anesthesiology, Vanderbilt University School of Medicine, Nashville, TN 37232, USA
| | - Kristopher T Kahle
- Department of Neurosurgery, Massachusetts General Hospital and Harvard Medical School, Boston, MA 02114, USA; Broad Institute of Harvard and MIT, Cambridge, MA, 02142, USA; Department of Neurosurgery and Division of Genetics and Genomics, Boston Children's Hospital, Boston, MA, USA.
| |
Collapse
|
35
|
Lam P, Newland J, Faull RLM, Kwakowsky A. Cation-Chloride Cotransporters KCC2 and NKCC1 as Therapeutic Targets in Neurological and Neuropsychiatric Disorders. Molecules 2023; 28:1344. [PMID: 36771011 PMCID: PMC9920462 DOI: 10.3390/molecules28031344] [Citation(s) in RCA: 22] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2022] [Revised: 01/21/2023] [Accepted: 01/25/2023] [Indexed: 02/04/2023] Open
Abstract
Neurological diseases including Alzheimer's, Huntington's disease, Parkinson's disease, Down syndrome and epilepsy, and neuropsychiatric disorders such as schizophrenia, are conditions that affect not only individuals but societies on a global scale. Current therapies offer a means for small symptomatic relief, but recently there has been increasing demand for therapeutic alternatives. The γ-aminobutyric acid (GABA)ergic signaling system has been investigated for developing new therapies as it has been noted that any dysfunction or changes to this system can contribute to disease progression. Expression of the K-Cl-2 (KCC2) and N-K-C1-1 (NKCC1) cation-chloride cotransporters (CCCs) has recently been linked to the disruption of GABAergic activity by affecting the polarity of GABAA receptor signaling. KCC2 and NKCC1 play a part in multiple neurological and neuropsychiatric disorders, making them a target of interest for potential therapies. This review explores current research suggesting the pathophysiological role and therapeutic importance of KCC2 and NKCC1 in neuropsychiatric and neurological disorders.
Collapse
Affiliation(s)
- Patricia Lam
- Centre for Brain Research, Department of Anatomy and Medical Imaging, Faculty of Medical and Health Sciences, University of Auckland, Auckland 1023, New Zealand
| | - Julia Newland
- Centre for Brain Research, Department of Anatomy and Medical Imaging, Faculty of Medical and Health Sciences, University of Auckland, Auckland 1023, New Zealand
| | - Richard L. M. Faull
- Centre for Brain Research, Department of Anatomy and Medical Imaging, Faculty of Medical and Health Sciences, University of Auckland, Auckland 1023, New Zealand
| | - Andrea Kwakowsky
- Centre for Brain Research, Department of Anatomy and Medical Imaging, Faculty of Medical and Health Sciences, University of Auckland, Auckland 1023, New Zealand
- Pharmacology and Therapeutics, School of Medicine, Galway Neuroscience Centre, University of Galway, H91 W5P7 Galway, Ireland
| |
Collapse
|
36
|
Chen B, Xie C, Shi T, Yue S, Li W, Huang G, Zhang Y, Liu W. Activation of Swell1 in microglia suppresses neuroinflammation and reduces brain damage in ischemic stroke. Neurobiol Dis 2023; 176:105936. [PMID: 36511337 DOI: 10.1016/j.nbd.2022.105936] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2022] [Revised: 10/17/2022] [Accepted: 11/25/2022] [Indexed: 11/29/2022] Open
Abstract
Cl- movement and Cl--sensitive signal pathways contributes to the survival and switch of inflammatory phenotype of microglia and are believed to play a key role in the inflammatory brain injury after ischemic stroke. Here, we demonstrated an important role of Cl- transmembrane transporter Swell1, in the survival and M2-like polarization of microglia in ischemic stroke. Knockdown or overexpression of Swell1 in cultured microglia inhibited or increased hypotonic-activated Cl- currents, respectively, and these changes were completely blocked by the volume-regulated anion channels (VRACs) inhibitor DCPIB. Swell1 conditional knock-in mice promoted microglia survival in ischemic brain region and resulted in significant reductions in neural cell death, infarction volume and neurological deficits following transient middle cerebral artery occlusion (tMCAO). Using gene manipulating technique and pharmacological inhibitors, we further revealed that Swell1 opening led to SGK1 (a Cl--sensitive kinase)-mediated activation of FOXO3a/CREB as well as WNK1 (another Cl--sensitive kinase)-mediated SPAK/OSR1-CCCs activation, which promoted microglia survival and M2-like polarization, thereby attenuating neuroinflammation and ischemic brain injury. Taken together, our results demonstrated that Swell1 is an essential component of microglia VRACs and its activation protects against ischemic brain injury through promoting microglia survival and M2-like polarization.
Collapse
Affiliation(s)
- Baoyi Chen
- Department of Neurosurgery, Shenzhen Key Laboratory of Neurosurgery, The First Affiliated Hospital of Shenzhen University, Shenzhen Second People's Hospital, Shenzhen 518035, China
| | - Cong Xie
- Department of Neurosurgery, Shenzhen Key Laboratory of Neurosurgery, The First Affiliated Hospital of Shenzhen University, Shenzhen Second People's Hospital, Shenzhen 518035, China; Department of Neurosurgery, The First Affiliated Hospital of Shenzhen University, Shenzhen Second People's Hospital, Graduate School of Guangzhou Medical University, Shenzhen 518035, China
| | - Tengrui Shi
- Department of Neurosurgery, Shenzhen Key Laboratory of Neurosurgery, The First Affiliated Hospital of Shenzhen University, Shenzhen Second People's Hospital, Shenzhen 518035, China
| | - Shiqin Yue
- Department of Neurosurgery, Shenzhen Key Laboratory of Neurosurgery, The First Affiliated Hospital of Shenzhen University, Shenzhen Second People's Hospital, Shenzhen 518035, China; School of Pharmaceutical Sciences, Health Science Center, Shenzhen University, Shenzhen 518035, China
| | - Weiping Li
- Department of Neurosurgery, Shenzhen Key Laboratory of Neurosurgery, The First Affiliated Hospital of Shenzhen University, Shenzhen Second People's Hospital, Shenzhen 518035, China
| | - Guodong Huang
- Department of Neurosurgery, Shenzhen Key Laboratory of Neurosurgery, The First Affiliated Hospital of Shenzhen University, Shenzhen Second People's Hospital, Shenzhen 518035, China
| | - Yuan Zhang
- Department of Neurosurgery, Shenzhen Key Laboratory of Neurosurgery, The First Affiliated Hospital of Shenzhen University, Shenzhen Second People's Hospital, Shenzhen 518035, China.
| | - Wenlan Liu
- Department of Neurosurgery, Shenzhen Key Laboratory of Neurosurgery, The First Affiliated Hospital of Shenzhen University, Shenzhen Second People's Hospital, Shenzhen 518035, China.
| |
Collapse
|
37
|
Lones L, DiAntonio A. SIK3 and Wnk converge on Fray to regulate glial K+ buffering and seizure susceptibility. PLoS Genet 2023; 19:e1010581. [PMID: 36626385 PMCID: PMC9870106 DOI: 10.1371/journal.pgen.1010581] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2022] [Revised: 01/23/2023] [Accepted: 12/19/2022] [Indexed: 01/11/2023] Open
Abstract
Glial cells play a critical role in maintaining homeostatic ion concentration gradients. Salt-inducible kinase 3 (SIK3) regulates a gene expression program that controls K+ buffering in glia, and upregulation of this pathway suppresses seizure behavior in the eag, Shaker hyperexcitability mutant. Here we show that boosting the glial SIK3 K+ buffering pathway suppresses seizures in three additional molecularly diverse hyperexcitable mutants, highlighting the therapeutic potential of upregulating glial K+ buffering. We then explore additional mechanisms regulating glial K+ buffering. Fray, a transcriptional target of the SIK3 K+ buffering program, is a kinase that promotes K+ uptake by activating the Na+/K+/Cl- co-transporter, Ncc69. We show that the Wnk kinase phosphorylates Fray in Drosophila glia and that this activity is required to promote K+ buffering. This identifies Fray as a convergence point between the SIK3-dependent transcriptional program and Wnk-dependent post-translational regulation. Bypassing both regulatory mechanisms via overexpression of a constitutively active Fray in glia is sufficient to robustly suppress seizure behavior in multiple Drosophila models of hyperexcitability. Finally, we identify cortex glia as a critical cell type for regulation of seizure susceptibility, as boosting K+ buffering via expression of activated Fray exclusively in these cells is sufficient to suppress seizure behavior. These findings highlight Fray as a key convergence point for distinct K+ buffering regulatory mechanisms and cortex glia as an important locus for control of neuronal excitability.
Collapse
Affiliation(s)
- Lorenzo Lones
- Department of Developmental Biology, Washington University School of Medicine, St. Louis, Missouri, United States of America
| | - Aaron DiAntonio
- Department of Developmental Biology, Washington University School of Medicine, St. Louis, Missouri, United States of America
- Needleman Center for Neurometabolism and Axonal Therapeutics, Washington University School of Medicine, St. Louis, Missouri, United States of America
| |
Collapse
|
38
|
Regulation of lens water content: Effects on the physiological optics of the lens. Prog Retin Eye Res 2022:101152. [DOI: 10.1016/j.preteyeres.2022.101152] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2022] [Revised: 11/21/2022] [Accepted: 11/22/2022] [Indexed: 12/09/2022]
|
39
|
Zhao Y, Cao E. Structural Pharmacology of Cation-Chloride Cotransporters. MEMBRANES 2022; 12:1206. [PMID: 36557113 PMCID: PMC9784483 DOI: 10.3390/membranes12121206] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 09/09/2022] [Revised: 11/20/2022] [Accepted: 11/25/2022] [Indexed: 06/17/2023]
Abstract
Loop and thiazide diuretics have been cornerstones of clinical management of hypertension and fluid overload conditions for more than five decades. The hunt for their molecular targets led to the discovery of cation-chloride cotransporters (CCCs) that catalyze electroneutral movement of Cl- together with Na+ and/or K+. CCCs consist of two 1 Na+-1 K+-2 Cl- (NKCC1-2), one 1 Na+-1 Cl- (NCC), and four 1 K+-1 Cl- (KCC1-4) transporters in human. CCCs are fundamental in trans-epithelia ion secretion and absorption, homeostasis of intracellular Cl- concentration and cell volume, and regulation of neuronal excitability. Malfunction of NKCC2 and NCC leads to abnormal salt and water retention in the kidney and, consequently, imbalance in electrolytes and blood pressure. Mutations in KCC2 and KCC3 are associated with brain disorders due to impairments in regulation of excitability and possibly cell volume of neurons. A recent surge of structures of CCCs have defined their dimeric architecture, their ion binding sites, their conformational changes associated with ion translocation, and the mechanisms of action of loop diuretics and small molecule inhibitors. These breakthroughs now set the stage to expand CCC pharmacology beyond loop and thiazide diuretics, developing the next generation of diuretics with improved potency and specificity. Beyond drugging renal-specific CCCs, brain-penetrable therapeutics are sorely needed to target CCCs in the nervous system for the treatment of neurological disorders and psychiatric conditions.
Collapse
|
40
|
Gan Y, Wei Z, Liu C, Li G, Feng Y, Deng Y. Solute carrier transporter disease and developmental and epileptic encephalopathy. Front Neurol 2022; 13:1013903. [PMID: 36419532 PMCID: PMC9676364 DOI: 10.3389/fneur.2022.1013903] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2022] [Accepted: 09/07/2022] [Indexed: 09/14/2023] Open
Abstract
The International League Against Epilepsy officially revised its classification in 2017, which amended "epileptic encephalopathy" to "developmental and epileptic encephalopathy". With the development of genetic testing technology, an increasing number of genes that cause developmental and epileptic encephalopathies are being identified. Among these, solute transporter dysfunction is part of the etiology of developmental and epileptic encephalopathies. Solute carrier transporters play an essential physiological function in the human body, and their dysfunction is associated with various human diseases. Therefore, in-depth studies of developmental and epileptic encephalopathies caused by solute carrier transporter dysfunction can help develop new therapeutic modalities to facilitate the treatment of refractory epilepsy and improve patient prognosis. In this article, the concept of transporter protein disorders is first proposed, and nine developmental and epileptic encephalopathies caused by solute carrier transporter dysfunction are described in detail in terms of pathogenesis, clinical manifestations, ancillary tests, and precise treatment to provide ideas for the precise treatment of epilepsy.
Collapse
Affiliation(s)
- Yajing Gan
- Department of Neurology, Epilepsy Center of Xijing Hospital, Fourth Military Medical University, Xi'an, China
| | - Zihan Wei
- Department of Neurology, Epilepsy Center of Xijing Hospital, Fourth Military Medical University, Xi'an, China
| | - Chao Liu
- Department of Neurology, Epilepsy Center of Xijing Hospital, Fourth Military Medical University, Xi'an, China
| | - Guoyan Li
- Department of Neurology, Epilepsy Center of Xijing Hospital, Fourth Military Medical University, Xi'an, China
| | - Yan Feng
- Department of Neurology, Epilepsy Center of Xijing Hospital, Fourth Military Medical University, Xi'an, China
| | - Yanchun Deng
- Department of Neurology, Epilepsy Center of Xijing Hospital, Fourth Military Medical University, Xi'an, China
- Xijing Institute of Epilepsy and Encephalopathy, Xi'an, China
| |
Collapse
|
41
|
Murillo-de-Ozores AR, Carbajal-Contreras H, Magaña-Ávila GR, Valdés R, Grajeda-Medina LI, Vázquez N, Zariñán T, López-Saavedra A, Sharma A, Lin DH, Wang WH, Delpire E, Ellison DH, Gamba G, Castañeda-Bueno M. Multiple molecular mechanisms are involved in the activation of the kidney sodium-chloride cotransporter by hypokalemia. Kidney Int 2022; 102:1030-1041. [PMID: 35870644 PMCID: PMC10411384 DOI: 10.1016/j.kint.2022.06.027] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2022] [Revised: 05/30/2022] [Accepted: 06/23/2022] [Indexed: 12/14/2022]
Abstract
Low potassium intake activates the kidney sodium-chloride cotransporter (NCC) whose phosphorylation and activity depend on the With-No-Lysine kinase 4 (WNK4) that is inhibited by chloride binding to its kinase domain. Low extracellular potassium activates NCC by decreasing intracellular chloride thereby promoting chloride dissociation from WNK4 where residue L319 of WNK4 participates in chloride coordination. Since the WNK4-L319F mutant is constitutively active and chloride-insensitive in vitro, we generated mice harboring this mutation that displayed slightly increased phosphorylated NCC and mild hyperkalemia when on a 129/sv genetic background. On a low potassium diet, upregulation of phosphorylated NCC was observed, suggesting that in addition to chloride sensing by WNK4, other mechanisms participate which may include modulation of WNK4 activity and degradation by phosphorylation of the RRxS motif in regulatory domains present in WNK4 and KLHL3, respectively. Increased levels of WNK4 and kidney-specific WNK1 and phospho-WNK4-RRxS were observed in wild-type and WNK4L319F/L319F mice on a low potassium diet. Decreased extracellular potassium promoted WNK4-RRxS phosphorylation in vitro and ex vivo as well. These effects might be secondary to intracellular chloride depletion, as reduction of intracellular chloride in HEK293 cells increased phospho-WNK4-RRxS. Phospho-WNK4-RRxS levels were increased in mice lacking the Kir5.1 potassium channel, which presumably have decreased distal convoluted tubule intracellular chloride. Similarly, phospho-KLHL3 was modulated by changes in intracellular chloride in HEK293 cells. Thus, our data suggest that multiple chloride-regulated mechanisms are responsible for NCC upregulation by low extracellular potassium.
Collapse
Affiliation(s)
- Adrián R Murillo-de-Ozores
- Department of Nephrology and Mineral Metabolism, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Tlalpan, Mexico City; Faculty of Medicine, Universidad Nacional Autónoma de México, Coyoacan, Mexico City
| | - Héctor Carbajal-Contreras
- Department of Nephrology and Mineral Metabolism, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Tlalpan, Mexico City; PECEM, Faculty of Medicine, Universidad Nacional Autónoma de México, Coyoacan, Mexico City, Mexico
| | - Germán R Magaña-Ávila
- Department of Nephrology and Mineral Metabolism, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Tlalpan, Mexico City; Faculty of Medicine, Universidad Nacional Autónoma de México, Coyoacan, Mexico City
| | - Raquel Valdés
- Department of Nephrology and Mineral Metabolism, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Tlalpan, Mexico City; Molecular Physiology Unit, Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México, Tlalpan, Mexico City
| | - Leoneli I Grajeda-Medina
- Department of Nephrology and Mineral Metabolism, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Tlalpan, Mexico City
| | - Norma Vázquez
- Molecular Physiology Unit, Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México, Tlalpan, Mexico City
| | - Teresa Zariñán
- Red de Apoyo a la Investigación (RAI), Universidad Nacional Autónoma de México (UNAM), Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán (INCMNSZ), Mexico City, Mexico
| | - Alejandro López-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
| | - Avika Sharma
- Division of Nephrology and Hypertension, Department of Medicine, Oregon Health and Science University, Portland, Oregon, USA
| | - Dao-Hong Lin
- 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
| | - Eric Delpire
- Department of Anesthesiology, Vanderbilt University School of Medicine, Nashville, Tennessee, USA
| | - David H Ellison
- Division of Nephrology and Hypertension, Department of Medicine, Oregon Health and Science University, Portland, Oregon, USA; Oregon Clinical & Translational Research Institute, Oregon Health & Science University, Portland, Oregon, USA; Renal Section, VA Portland Health Care System, Portland, Oregon, USA
| | - Gerardo Gamba
- Department of Nephrology and Mineral Metabolism, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Tlalpan, Mexico City; PECEM, Faculty of Medicine, Universidad Nacional Autónoma de México, Coyoacan, Mexico City, Mexico; Molecular Physiology Unit, Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México, Tlalpan, Mexico City
| | - María Castañeda-Bueno
- Department of Nephrology and Mineral Metabolism, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Tlalpan, Mexico City.
| |
Collapse
|
42
|
Sinha AS, Wang T, Watanabe M, Hosoi Y, Sohara E, Akita T, Uchida S, Fukuda A. WNK3 kinase maintains neuronal excitability by reducing inwardly rectifying K+ conductance in layer V pyramidal neurons of mouse medial prefrontal cortex. Front Mol Neurosci 2022; 15:856262. [PMID: 36311015 PMCID: PMC9613442 DOI: 10.3389/fnmol.2022.856262] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2022] [Accepted: 09/22/2022] [Indexed: 11/13/2022] Open
Abstract
The with-no-lysine (WNK) family of serine-threonine kinases and its downstream kinases of STE20/SPS1-related proline/alanine-rich kinase (SPAK) and oxidative stress-responsive kinase-1 (OSR1) may regulate intracellular Cl− homeostasis through phosphorylation of cation-Cl− co-transporters. WNK3 is expressed in fetal and postnatal brains, and its expression level increases during development. Its roles in neurons, however, remain uncertain. Using WNK3 knockout (KO) mice, we investigated the role of WNK3 in the regulation of the intracellular Cl− concentration ([Cl−]i) and the excitability of layer V pyramidal neurons in the medial prefrontal cortex (mPFC). Gramicidin-perforated patch-clamp recordings in neurons from acute slice preparation at the postnatal day 21 indicated a significantly depolarized reversal potential for GABAA receptor-mediated currents by 6 mV, corresponding to the higher [Cl−]i level by ~4 mM in KO mice than in wild-type littermates. However, phosphorylation levels of SPAK and OSR1 and those of neuronal Na+-K+-2Cl− co-transporter NKCC1 and K+-Cl− co-transporter KCC2 did not significantly differ between KO and wild-type mice. Meanwhile, the resting membrane potential of neurons was more hyperpolarized by 7 mV, and the minimum stimulus current necessary for firing induction was increased in KO mice. These were due to an increased inwardly rectifying K+ (IRK) conductance, mediated by classical inwardly rectifying (Kir) channels, in KO neurons. The introduction of an active form of WNK3 into the recording neurons reversed these changes. The potential role of KCC2 function in the observed changes of KO neurons was investigated by applying a selective KCC2 activator, CLP290. This reversed the enhanced IRK conductance in KO neurons, indicating that both WNK3 and KCC2 are intimately linked in the regulation of resting K+ conductance. Evaluation of synaptic properties revealed that the frequency of miniature excitatory postsynaptic currents (mEPSCs) was reduced, whereas that of inhibitory currents (mIPSCs) was slightly increased in KO neurons. Together, the impact of these developmental changes on the membrane and synaptic properties was manifested as behavioral deficits in pre-pulse inhibition, a measure of sensorimotor gating involving multiple brain regions including the mPFC, in KO mice. Thus, the basal function of WNK3 would be the maintenance and/or development of both intrinsic and synaptic excitabilities.
Collapse
Affiliation(s)
- Adya Saran Sinha
- Department of Neurophysiology, Hamamatsu University School of Medicine, Hamamatsu, Japan
| | - Tianying Wang
- Department of Neurophysiology, Hamamatsu University School of Medicine, Hamamatsu, Japan
| | - Miho Watanabe
- Department of Neurophysiology, Hamamatsu University School of Medicine, Hamamatsu, Japan
| | - Yasushi Hosoi
- Department of Neurophysiology, Hamamatsu University School of Medicine, Hamamatsu, Japan
| | - Eisei Sohara
- Department of Nephrology, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, Tokyo, Japan
| | - Tenpei Akita
- Department of Neurophysiology, Hamamatsu University School of Medicine, Hamamatsu, Japan
| | - Shinichi Uchida
- Department of Nephrology, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, Tokyo, Japan
| | - Atsuo Fukuda
- Department of Neurophysiology, Hamamatsu University School of Medicine, Hamamatsu, Japan
- *Correspondence: Atsuo Fukuda
| |
Collapse
|
43
|
Rehman T, Karp PH, Thurman AL, Mather SE, Jain A, Cooney AL, Sinn PL, Pezzulo AA, Duffey ME, Welsh MJ. WNK Inhibition Increases Surface Liquid pH and Host Defense in Cystic Fibrosis Airway Epithelia. Am J Respir Cell Mol Biol 2022; 67:491-502. [PMID: 35849656 PMCID: PMC9564924 DOI: 10.1165/rcmb.2022-0172oc] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2022] [Accepted: 07/18/2022] [Indexed: 02/05/2023] Open
Abstract
In cystic fibrosis (CF), reduced HCO3- secretion acidifies the airway surface liquid (ASL), and the acidic pH disrupts host defenses. Thus, understanding the control of ASL pH (pHASL) in CF may help identify novel targets and facilitate therapeutic development. In diverse epithelia, the WNK (with-no-lysine [K]) kinases coordinate HCO3- and Cl- transport, but their functions in airway epithelia are poorly understood. Here, we tested the hypothesis that WNK kinases regulate CF pHASL. In primary cultures of differentiated human airway epithelia, inhibiting WNK kinases acutely increased both CF and non-CF pHASL. This response was HCO3- dependent and involved downstream SPAK/OSR1 (Ste20/SPS1-related proline-alanine-rich protein kinase/oxidative stress responsive 1 kinase). Importantly, WNK inhibition enhanced key host defenses otherwise impaired in CF. Human airway epithelia expressed two WNK isoforms in secretory cells and ionocytes, and knockdown of either WNK1 or WNK2 increased CF pHASL. WNK inhibition decreased Cl- secretion and the response to bumetanide, an NKCC1 (sodium-potassium-chloride cotransporter 1) inhibitor. Surprisingly, bumetanide alone or basolateral Cl- substitution also alkalinized CF pHASL. These data suggest that WNK kinases influence the balance between transepithelial Cl- versus HCO3- secretion. Moreover, reducing basolateral Cl- entry may increase HCO3- secretion and raise pHASL, thereby improving CF host defenses.
Collapse
Affiliation(s)
| | - Philip H. Karp
- Department of Internal Medicine and
- Howard Hughes Medical Institute, University of Iowa, Iowa City, Iowa; and
| | | | | | | | | | | | | | - Michael E. Duffey
- Department of Physiology and Biophysics, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, Buffalo, New York
| | - Michael J. Welsh
- Department of Internal Medicine and
- Department of Molecular Physiology and Biophysics, Roy J. and Lucille A. Carver College of Medicine, and
- Howard Hughes Medical Institute, University of Iowa, Iowa City, Iowa; and
| |
Collapse
|
44
|
KCC2 drives chloride microdomain formation in dendritic blebbing. Cell Rep 2022; 41:111556. [DOI: 10.1016/j.celrep.2022.111556] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2021] [Revised: 06/23/2022] [Accepted: 09/30/2022] [Indexed: 11/22/2022] Open
|
45
|
Gamba G. Arterial Blood Pressure, Neuronal Excitability, Mineral Metabolism and Cell Volume Regulation Mechanisms Revealed by Xenopus laevis oocytes. MEMBRANES 2022; 12:911. [PMID: 36295670 PMCID: PMC9612257 DOI: 10.3390/membranes12100911] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 06/28/2022] [Revised: 08/16/2022] [Accepted: 09/16/2022] [Indexed: 06/16/2023]
Abstract
Xenopus laevis oocytes have been an invaluable tool to discover and explore the molecular mechanisms and characteristics of many proteins, in particular integral membrane proteins. The oocytes were fundamental in many projects designed to identify the cDNA encoding a diversity of membrane proteins including receptors, transporters, channels and pores. In addition to being a powerful tool for cloning, oocytes were later used to experiment with the functional characterization of many of the identified proteins. In this review I present an overview of my personal 30-year experience using Xenopus laevis oocytes and the impact this had on a variety of fields such as arterial blood pressure, neuronal excitability, mineral metabolism and cell volume regulation.
Collapse
Affiliation(s)
- Gerardo Gamba
- Molecular Physiology Unit, Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México and Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Mexico City 04510, Mexico
| |
Collapse
|
46
|
Xiu M, Li L, Li Y, Gao Y. An update regarding the role of WNK kinases in cancer. Cell Death Dis 2022; 13:795. [PMID: 36123332 PMCID: PMC9485243 DOI: 10.1038/s41419-022-05249-y] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2022] [Revised: 09/07/2022] [Accepted: 09/08/2022] [Indexed: 01/23/2023]
Abstract
Mammalian WNK kinases (WNKs) are serine/threonine kinases that contain four members, WNK1-4. They function to maintain ion homeostasis and regulate blood pressure in mammals. Recent studies have revealed that the dysregulation of WNKs contributes to tumor growth, metastasis, and angiogenesis through complex mechanisms, especially through phosphorylating kinase substrates SPS1-related proline/alanine-rich kinase (SPAK) and oxidative stress-responsive kinase 1 (OSR1). Here, we review and discuss the relationships between WNKs and several key factors/biological processes in cancer, including ion channels, cation chloride cotransporters, sodium bicarbonate cotransporters, signaling pathways, angiogenesis, autophagy, and non-coding RNAs. In addition, the potential drugs for targeting WNK-SPAK/OSR1 signaling have also been discussed. This review summarizes and discusses knowledge of the roles of WNKs in cancer, which provides a comprehensive reference for future studies.
Collapse
Affiliation(s)
- Mengxi Xiu
- grid.24516.340000000123704535Department of Oncology, Shanghai East Hospital, Tongji University School of Medicine, 200120 Shanghai, China
| | - Li Li
- grid.24516.340000000123704535Department of Oncology, Shanghai East Hospital, Tongji University School of Medicine, 200120 Shanghai, China
| | - Yandong Li
- grid.24516.340000000123704535Department of Oncology, Shanghai East Hospital, Tongji University School of Medicine, 200120 Shanghai, China
| | - Yong Gao
- grid.24516.340000000123704535Department of Oncology, Shanghai East Hospital, Tongji University School of Medicine, 200120 Shanghai, China
| |
Collapse
|
47
|
Küry S, Zhang J, Besnard T, Caro-Llopis A, Zeng X, Robert SM, Josiah SS, Kiziltug E, Denommé-Pichon AS, Cogné B, Kundishora AJ, Hao LT, Li H, Stevenson RE, Louie RJ, Deb W, Torti E, Vignard V, McWalter K, Raymond FL, Rajabi F, Ranza E, Grozeva D, Coury SA, Blanc X, Brischoux-Boucher E, Keren B, Õunap K, Reinson K, Ilves P, Wentzensen IM, Barr EE, Guihard SH, Charles P, Seaby EG, Monaghan KG, Rio M, van Bever Y, van Slegtenhorst M, Chung WK, Wilson A, Quinquis D, Bréhéret F, Retterer K, Lindenbaum P, Scalais E, Rhodes L, Stouffs K, Pereira EM, Berger SM, Milla SS, Jaykumar AB, Cobb MH, Panchagnula S, Duy PQ, Vincent M, Mercier S, Gilbert-Dussardier B, Le Guillou X, Audebert-Bellanger S, Odent S, Schmitt S, Boisseau P, Bonneau D, Toutain A, Colin E, Pasquier L, Redon R, Bouman A, Rosenfeld JA, Friez MJ, Pérez-Peña H, Akhtar Rizvi SR, Haider S, Antonarakis SE, Schwartz CE, Martínez F, Bézieau S, Kahle KT, Isidor B. Rare pathogenic variants in WNK3 cause X-linked intellectual disability. Genet Med 2022; 24:1941-1951. [PMID: 35678782 DOI: 10.1016/j.gim.2022.05.009] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2021] [Revised: 05/10/2022] [Accepted: 05/11/2022] [Indexed: 02/08/2023] Open
Abstract
PURPOSE WNK3 kinase (PRKWNK3) has been implicated in the development and function of the brain via its regulation of the cation-chloride cotransporters, but the role of WNK3 in human development is unknown. METHOD We ascertained exome or genome sequences of individuals with rare familial or sporadic forms of intellectual disability (ID). RESULTS We identified a total of 6 different maternally-inherited, hemizygous, 3 loss-of-function or 3 pathogenic missense variants (p.Pro204Arg, p.Leu300Ser, p.Glu607Val) in WNK3 in 14 male individuals from 6 unrelated families. Affected individuals had ID with variable presence of epilepsy and structural brain defects. WNK3 variants cosegregated with the disease in 3 different families with multiple affected individuals. This included 1 large family previously diagnosed with X-linked Prieto syndrome. WNK3 pathogenic missense variants localize to the catalytic domain and impede the inhibitory phosphorylation of the neuronal-specific chloride cotransporter KCC2 at threonine 1007, a site critically regulated during the development of synaptic inhibition. CONCLUSION Pathogenic WNK3 variants cause a rare form of human X-linked ID with variable epilepsy and structural brain abnormalities and implicate impaired phospho-regulation of KCC2 as a pathogenic mechanism.
Collapse
Affiliation(s)
- Sébastien Küry
- Nantes Université, CHU Nantes, Service de Génétique Médicale, Nantes, France; Nantes Université, CHU Nantes, CNRS, INSERM, l'institut du thorax, Nantes, France.
| | - Jinwei Zhang
- Hatherly Laboratories, The Institute of Biomedical and Clinical Sciences, College of Medicine and Health, University of Exeter, Exeter, United Kingdom; Department of Neurosurgery, Yale School of Medicine, Yale University, New Haven, CT; State Key Laboratory of Bio-Organic and Natural Products Chemistry, Research Center of Chemical Kinomics, Shanghai Institute of Organic Chemistry, Chinese Academy of Sciences, Shanghai, China
| | - Thomas Besnard
- Nantes Université, CHU Nantes, Service de Génétique Médicale, Nantes, France; Nantes Université, CHU Nantes, CNRS, INSERM, l'institut du thorax, Nantes, France
| | - Alfonso Caro-Llopis
- Unidad de Genética, Hospital Universitario y Politécnico La Fe, Valencia, Spain
| | - Xue Zeng
- Department of Genetics, Yale School of Medicine, Yale University, New Haven, CT
| | - Stephanie M Robert
- Department of Genetics, Yale School of Medicine, Yale University, New Haven, CT
| | - Sunday S Josiah
- Hatherly Laboratories, The Institute of Biomedical and Clinical Sciences, College of Medicine and Health, University of Exeter, Exeter, United Kingdom
| | - Emre Kiziltug
- Department of Genetics, Yale School of Medicine, Yale University, New Haven, CT
| | - Anne-Sophie Denommé-Pichon
- Département de Biochimie et Génétique, Centre Hospitalier Universitaire Angers, Angers, France; UMR CNRS 6214, INSERM 1083, Université d'Angers, Angers, France
| | - Benjamin Cogné
- Nantes Université, CHU Nantes, Service de Génétique Médicale, Nantes, France; Nantes Université, CHU Nantes, CNRS, INSERM, l'institut du thorax, Nantes, France
| | - Adam J Kundishora
- Department of Neurosurgery, Yale School of Medicine, Yale University, New Haven, CT
| | - Le T Hao
- Department of Neurosurgery, Yale School of Medicine, Yale University, New Haven, CT
| | - Hong Li
- Departments of Human Genetics and Pediatrics, School of Medicine, Emory University, Atlanta, GA
| | | | | | - Wallid Deb
- Nantes Université, CHU Nantes, Service de Génétique Médicale, Nantes, France; Nantes Université, CHU Nantes, CNRS, INSERM, l'institut du thorax, Nantes, France
| | | | - Virginie Vignard
- Nantes Université, CHU Nantes, CNRS, INSERM, l'institut du thorax, Nantes, France
| | | | - F Lucy Raymond
- Department of Medical Genetics, Cambridge Institute for Medical Research, University of Cambridge, Biomedical Campus Cambridge, Cambridge, United Kingdom
| | - Farrah Rajabi
- Division of Genetics and Genomics, Boston Children's Hospital, Boston, MA
| | - Emmanuelle Ranza
- Medigenome, Swiss Institute of Genomic Medicine, Geneva, Switzerland
| | - Detelina Grozeva
- Department of Medical Genetics, Cambridge Institute for Medical Research, University of Cambridge, Biomedical Campus Cambridge, Cambridge, United Kingdom; Centre for Trials Research, Cardiff University, Cardiff, United Kingdom
| | - Stephanie A Coury
- Division of Genetics and Genomics, Boston Children's Hospital, Boston, MA
| | - Xavier Blanc
- Medigenome, Swiss Institute of Genomic Medicine, Geneva, Switzerland
| | - Elise Brischoux-Boucher
- Centre de Génétique Humaine, CHU de Besançon, Université de Bourgogne Franche-Comté, Besançon, France
| | - Boris Keren
- Department of Genetics, Centre de Référence Déficiences Intellectuelles de Causes Rares, Pitié-Salpêtrière Hospital, Assistance Publique-Hôpitaux de Paris, Paris, France
| | - Katrin Õunap
- Department of Clinical Genetics, Genetics and Personalized Medicine Clinic, Tartu University Hospital, Tartu, Estonia; Department of Clinical Genetics, Institute of Clinical Medicine, Faculty of Medicine, University of Tartu, Tartu, Estonia
| | - Karit Reinson
- Department of Clinical Genetics, Genetics and Personalized Medicine Clinic, Tartu University Hospital, Tartu, Estonia; Department of Clinical Genetics, Institute of Clinical Medicine, Faculty of Medicine, University of Tartu, Tartu, Estonia
| | - Pilvi Ilves
- Department of Clinical Genetics, Institute of Clinical Medicine, Faculty of Medicine, University of Tartu, Tartu, Estonia; Department of Radiology, Tartu University Hospital, Tartu, Estonia
| | | | - Eileen E Barr
- Departments of Human Genetics and Pediatrics, School of Medicine, Emory University, Atlanta, GA
| | - Solveig Heide Guihard
- Department of Genetics, Centre de Référence Déficiences Intellectuelles de Causes Rares, Pitié-Salpêtrière Hospital, Assistance Publique-Hôpitaux de Paris, Paris, France; Groupe de Recherche Clinique, Déficience Intellectuelle et Autisme, Sorbonne University, Paris, France
| | - Perrine Charles
- Department of Genetics, Centre de Référence Déficiences Intellectuelles de Causes Rares, Pitié-Salpêtrière Hospital, Assistance Publique-Hôpitaux de Paris, Paris, France
| | - Eleanor G Seaby
- Program in Medical and Population Genetics, Broad Institute of MIT and Harvard, Cambridge, MA; Genomic Informatics Group, University of Southampton, Southampton, United Kingdom
| | | | - Marlène Rio
- Developmental Brain Disorders laboratory, INSERM UMR 1163, Imagine Institute, University of Paris, Paris, France; Department of Genetics, Centre de Référence Déficiences Intellectuelles de Causes Rares, Necker Enfants Malades Hospital, APHP, Paris, France
| | - Yolande van Bever
- Department of Clinical Genetics, Erasmus MC University Medical Center, Rotterdam, the Netherlands
| | - Marjon van Slegtenhorst
- Department of Clinical Genetics, Erasmus MC University Medical Center, Rotterdam, the Netherlands
| | - Wendy K Chung
- Departments of Pediatrics and Medicine, Columbia University Irving Medical Center, Columbia University New York, NY
| | - Ashley Wilson
- Division of Clinical Genetics, Department of Pediatrics, Columbia University Vagelos College of Physicians and Surgeons and New York-Presbyterian Morgan Stanley Children's Hospital, New York, NY
| | - Delphine Quinquis
- Nantes Université, CHU Nantes, Service de Génétique Médicale, Nantes, France
| | - Flora Bréhéret
- Nantes Université, CHU Nantes, Service de Génétique Médicale, Nantes, France
| | | | - Pierre Lindenbaum
- Nantes Université, CHU Nantes, CNRS, INSERM, l'institut du thorax, Nantes, France
| | - Emmanuel Scalais
- Division of Pediatric Neurology, Department of Pediatrics, Centre Hospitalier de Luxembourg, Luxembourg City, Luxembourg
| | | | - Katrien Stouffs
- Neurogenetics Research Group, Reproduction and Genetics, Vrije Universiteit Brussel (VUB), Universitair Ziekenhuis Brussel (UZ Brussel), Laarbeeklaan, Brussels, Belgium
| | - Elaine M Pereira
- Division of Clinical Genetics, Department of Pediatrics, Columbia University Vagelos College of Physicians and Surgeons and New York-Presbyterian Morgan Stanley Children's Hospital, New York, NY
| | - Sara M Berger
- Division of Clinical Genetics, Department of Pediatrics, Columbia University Vagelos College of Physicians and Surgeons and New York-Presbyterian Morgan Stanley Children's Hospital, New York, NY
| | - Sarah S Milla
- Department of Radiology and Imaging Sciences, School of Medicine, Emory University, Atlanta, GA
| | - Ankita B Jaykumar
- Department of Pharmacology, UT Southwestern Medical Center, Dallas, TX
| | - Melanie H Cobb
- Department of Pharmacology, UT Southwestern Medical Center, Dallas, TX
| | - Shreyas Panchagnula
- Unidad de Genética, Hospital Universitario y Politécnico La Fe, Valencia, Spain
| | - Phan Q Duy
- Department of Neurosurgery, Yale School of Medicine, Yale University, New Haven, CT
| | - Marie Vincent
- Nantes Université, CHU Nantes, Service de Génétique Médicale, Nantes, France; Nantes Université, CHU Nantes, CNRS, INSERM, l'institut du thorax, Nantes, France
| | - Sandra Mercier
- Nantes Université, CHU Nantes, Service de Génétique Médicale, Nantes, France; Nantes Université, CHU Nantes, CNRS, INSERM, l'institut du thorax, Nantes, France
| | | | | | | | - Sylvie Odent
- Service de Génétique Clinique, ERN ITHACA, CHU Rennes, Rennes, France; Institut de Génétique et Développement de Rennes, IGDR UMR 6290 CNRS, INSERM, IGDR Univ Rennes, Rennes, France
| | - Sébastien Schmitt
- Nantes Université, CHU Nantes, Service de Génétique Médicale, Nantes, France
| | - Pierre Boisseau
- Nantes Université, CHU Nantes, Service de Génétique Médicale, Nantes, France
| | - Dominique Bonneau
- Département de Biochimie et Génétique, Centre Hospitalier Universitaire Angers, Angers, France; UMR CNRS 6214, INSERM 1083, Université d'Angers, Angers, France
| | - Annick Toutain
- Unité de Génétique Médicale, Centre Hospitalier Régional Universitaire de Tours, France; Unité Mixte de Recherche 1253, iBrain, Université de Tours, Institut National de la Santé et de la Recherche Médicale, Tours, France
| | - Estelle Colin
- Département de Biochimie et Génétique, Centre Hospitalier Universitaire Angers, Angers, France; UMR CNRS 6214, INSERM 1083, Université d'Angers, Angers, France
| | - Laurent Pasquier
- Service de Génétique Clinique, ERN ITHACA, CHU Rennes, Rennes, France; Institut de Génétique et Développement de Rennes, IGDR UMR 6290 CNRS, INSERM, IGDR Univ Rennes, Rennes, France
| | - Richard Redon
- Nantes Université, CHU Nantes, CNRS, INSERM, l'institut du thorax, Nantes, France
| | - Arjan Bouman
- Department of Clinical Genetics, Erasmus MC University Medical Center, Rotterdam, the Netherlands
| | - Jill A Rosenfeld
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX
| | | | - Helena Pérez-Peña
- Department of Pharmaceutical and Biological Chemistry, UCL School of Pharmacy, University College London, London, United Kingdom
| | - Syed Raza Akhtar Rizvi
- Department of Pharmaceutical and Biological Chemistry, UCL School of Pharmacy, University College London, London, United Kingdom
| | - Shozeb Haider
- Department of Pharmaceutical and Biological Chemistry, UCL School of Pharmacy, University College London, London, United Kingdom; Centre for Advanced Research Computing, University College London, London, United Kingdom
| | - Stylianos E Antonarakis
- Medigenome, Swiss Institute of Genomic Medicine, Geneva, Switzerland; Department of Genetic Medicine and Development, University of Geneva, Geneva, Switzerland; iGE3, Institute of Genetics and Genomics of Geneva, University of Geneva, Geneva, Switzerland
| | | | - Francisco Martínez
- Unidad de Genética, Hospital Universitario y Politécnico La Fe, Valencia, Spain
| | - Stéphane Bézieau
- Nantes Université, CHU Nantes, Service de Génétique Médicale, Nantes, France; Nantes Université, CHU Nantes, CNRS, INSERM, l'institut du thorax, Nantes, France
| | - Kristopher T Kahle
- Department of Neurosurgery, Yale School of Medicine, Yale University, New Haven, CT; Department of Cellular and Molecular Physiology, Yale School of Medicine, Yale University, New Haven, CT; NIH-Yale Centers for Mendelian Genomics, Yale School of Medicine, Yale University, New Haven, CT; Yale Stem Cell Center, Yale School of Medicine, Yale University, New Haven, CT.
| | - Bertrand Isidor
- Nantes Université, CHU Nantes, Service de Génétique Médicale, Nantes, France; Nantes Université, CHU Nantes, CNRS, INSERM, l'institut du thorax, Nantes, France
| |
Collapse
|
48
|
Hartmann AM, Nothwang HG. NKCC1 and KCC2: Structural insights into phospho-regulation. Front Mol Neurosci 2022; 15:964488. [PMID: 35935337 PMCID: PMC9355526 DOI: 10.3389/fnmol.2022.964488] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2022] [Accepted: 07/06/2022] [Indexed: 11/13/2022] Open
Abstract
Inhibitory neurotransmission plays a fundamental role in the central nervous system, with about 30–50% of synaptic connections being inhibitory. The action of both inhibitory neurotransmitter, gamma-aminobutyric-acid (GABA) and glycine, mainly relies on the intracellular Cl– concentration in neurons. This is set by the interplay of the cation chloride cotransporters NKCC1 (Na+, K+, Cl– cotransporter), a main Cl– uptake transporter, and KCC2 (K+, Cl– cotransporter), the principle Cl– extruder in neurons. Accordingly, their dysfunction is associated with severe neurological, psychiatric, and neurodegenerative disorders. This has triggered great interest in understanding their regulation, with a strong focus on phosphorylation. Recent structural data by cryogenic electron microscopy provide the unique possibility to gain insight into the action of these phosphorylations. Interestingly, in KCC2, six out of ten (60%) known regulatory phospho-sites reside within a region of 134 amino acid residues (12% of the total residues) between helices α8 and α9 that lacks fixed or ordered three-dimensional structures. It thus represents a so-called intrinsically disordered region. Two further phospho-sites, Tyr903 and Thr906, are also located in a disordered region between the ß8 strand and the α8 helix. We make the case that especially the disordered region between helices α8 and α9 acts as a platform to integrate different signaling pathways and simultaneously constitute a flexible, highly dynamic linker that can survey a wide variety of distinct conformations. As each conformation can have distinct binding affinities and specificity properties, this enables regulation of [Cl–]i and thus the ionic driving force in a history-dependent way. This region might thus act as a molecular processor underlying the well described phenomenon of ionic plasticity that has been ascribed to inhibitory neurotransmission. Finally, it might explain the stunning long-range effects of mutations on phospho-sites in KCC2.
Collapse
Affiliation(s)
- Anna-Maria Hartmann
- Division of Neurogenetics, School of Medicine and Health Sciences, Carl von Ossietzky University Oldenburg, Oldenburg, Germany
- Research Center for Neurosensory Sciences, Carl von Ossietzky University Oldenburg, Oldenburg, Germany
- *Correspondence: Anna-Maria Hartmann,
| | - Hans Gerd Nothwang
- Division of Neurogenetics, School of Medicine and Health Sciences, Carl von Ossietzky University Oldenburg, Oldenburg, Germany
- Research Center for Neurosensory Sciences, Carl von Ossietzky University Oldenburg, Oldenburg, Germany
- Center of Excellence Hearing4all, Carl von Ossietzky University Oldenburg, Oldenburg, Germany
| |
Collapse
|
49
|
Hui KK, Chater TE, Goda Y, Tanaka M. How Staying Negative Is Good for the (Adult) Brain: Maintaining Chloride Homeostasis and the GABA-Shift in Neurological Disorders. Front Mol Neurosci 2022; 15:893111. [PMID: 35875665 PMCID: PMC9305173 DOI: 10.3389/fnmol.2022.893111] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2022] [Accepted: 06/10/2022] [Indexed: 01/27/2023] Open
Abstract
Excitatory-inhibitory (E-I) imbalance has been shown to contribute to the pathogenesis of a wide range of neurodevelopmental disorders including autism spectrum disorders, epilepsy, and schizophrenia. GABA neurotransmission, the principal inhibitory signal in the mature brain, is critically coupled to proper regulation of chloride homeostasis. During brain maturation, changes in the transport of chloride ions across neuronal cell membranes act to gradually change the majority of GABA signaling from excitatory to inhibitory for neuronal activation, and dysregulation of this GABA-shift likely contributes to multiple neurodevelopmental abnormalities that are associated with circuit dysfunction. Whilst traditionally viewed as a phenomenon which occurs during brain development, recent evidence suggests that this GABA-shift may also be involved in neuropsychiatric disorders due to the "dematuration" of affected neurons. In this review, we will discuss the cell signaling and regulatory mechanisms underlying the GABA-shift phenomenon in the context of the latest findings in the field, in particular the role of chloride cotransporters NKCC1 and KCC2, and furthermore how these regulatory processes are altered in neurodevelopmental and neuropsychiatric disorders. We will also explore the interactions between GABAergic interneurons and other cell types in the developing brain that may influence the GABA-shift. Finally, with a greater understanding of how the GABA-shift is altered in pathological conditions, we will briefly outline recent progress on targeting NKCC1 and KCC2 as a therapeutic strategy against neurodevelopmental and neuropsychiatric disorders associated with improper chloride homeostasis and GABA-shift abnormalities.
Collapse
Affiliation(s)
- Kelvin K. Hui
- Department of Developmental and Molecular Biology, Albert Einstein College of Medicine, Bronx, NY, United States
- Institute for Aging Research, Albert Einstein College of Medicine, Bronx, NY, United States
| | - Thomas E. Chater
- Laboratory for Synaptic Plasticity and Connectivity, RIKEN Center for Brain Science, Wako, Japan
| | - Yukiko Goda
- Laboratory for Synaptic Plasticity and Connectivity, RIKEN Center for Brain Science, Wako, Japan
- Synapse Biology Unit, Okinawa Institute for Science and Technology Graduate University, Onna, Japan
| | - Motomasa Tanaka
- Laboratory for Protein Conformation Diseases, RIKEN Center for Brain Science, Wako, Japan
| |
Collapse
|
50
|
Structural basis for inhibition of the Cation-chloride cotransporter NKCC1 by the diuretic drug bumetanide. Nat Commun 2022; 13:2747. [PMID: 35585053 PMCID: PMC9117670 DOI: 10.1038/s41467-022-30407-3] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2021] [Accepted: 04/28/2022] [Indexed: 11/08/2022] Open
Abstract
Cation-chloride cotransporters (CCCs) NKCC1 and NKCC2 catalyze electroneutral symport of 1 Na+, 1 K+, and 2 Cl− across cell membranes. NKCC1 mediates trans-epithelial Cl− secretion and regulates excitability of some neurons and NKCC2 is critical to renal salt reabsorption. Both transporters are inhibited by the so-called loop diuretics including bumetanide, and these drugs are a mainstay for treating edema and hypertension. Here, our single-particle electron cryo-microscopy structures supported by functional studies reveal an outward-facing conformation of NKCC1, showing bumetanide wedged into a pocket in the extracellular ion translocation pathway. Based on these and the previously published inward-facing structures, we define the translocation pathway and the conformational changes necessary for ion translocation. We also identify an NKCC1 dimer with separated transmembrane domains and extensive transmembrane and C-terminal domain interactions. We further define an N-terminal phosphoregulatory domain that interacts with the C-terminal domain, suggesting a mechanism whereby (de)phosphorylation regulates NKCC1 by tuning the strength of this domain association. Loop diuretics including bumetanide inhibit Na+-K+-Cl−-cotransporters (NKCCs) and are used for the treatment of edema and hypertension. Here, Zhao et. al. report structures of NKCC1 with bumetanide bound, revealing its mechanism of action that would facilitate design of novel diuretics.
Collapse
|