1
|
Kimera L, Nadimpalli S, Kurup S, Sessions Cole F, Huang R, Sisco K, Ranaivo HR, Shinawi M, Dickson P, Mian A, Reynolds M. Case report: ocular manifestations of a gain-of-function mutation in CLCN6, a newly diagnosed disease. Ophthalmic Genet 2024; 45:271-274. [PMID: 38095064 PMCID: PMC11176260 DOI: 10.1080/13816810.2023.2291683] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2023] [Revised: 10/31/2023] [Accepted: 11/30/2023] [Indexed: 02/23/2024]
Abstract
BACKGROUND In 2020, a new disease was reported by Polovitskaya et al., caused by a monoallelic, gain-of-function mutation in CLCN6, encoding the ClC-6 Cl-/H±exchanger. METHODS Here, we report the ophthalmic findings of one of the first three patients with this disease (the proband) and review the findings in the other two patients in the literature. RESULTS The CLCN6 gene is part of the voltage-dependent chloride channel protein family. It functions as either a chloride channel aiding in cell-volume regulation and acidification of intracellular organelles or as an antiporter, which are membrane proteins involved in the transport of molecules across a phospholipid membrane. This particular gene is found in late endosomes. Ion transport across endosome membranes is essential for endosomal function. The proband carried a de novo c.1658A>G (p.Tyr553Cys) mutation in CLCN6. The patient reported herein has a notable optic nerve appearance. The nerve initially appeared elevated. Over time, the optic nerve elevation appearance decreased, associated with progressive vision loss with a visual acuity of 20/470 at last follow-up. CONCLUSION While Clcn6-/- mice have been found to have a mild neuronal lysosomal storage phenotype, the three reported children with a de novo c.1658A>G (p.Tyr553Cys) variant displayed significant developmental delay and neurodegeneration.
Collapse
Affiliation(s)
- Lawrencia Kimera
- Department of Ophthalmology and Visual Sciences, Washington University in St. Louis School of Medicine, St. Louis, MO, USA
| | - Sameera Nadimpalli
- Division of Ophthalmology, Ann & Robert H. Lurie Children’s Hospital of Chicago, Chicago, IL, USA
| | - Sudhi Kurup
- Division of Ophthalmology, Ann & Robert H. Lurie Children’s Hospital of Chicago, Chicago, IL, USA
- Department of Ophthalmology, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - F. Sessions Cole
- Department of Pediatrics, Division of Neonatal Medicine, Washington University in St. Louis School of Medicine, St. Louis, MO, USA
- Department of Cell and Molecular Biology, Washington University in St. Louis School of Medicine, St. Louis, MO, USA
| | - Russell Huang
- Department of Ophthalmology, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Kathleen Sisco
- Department of Pediatrics, Division of Genetics and Genomic Medicine, Washington University in St. Louis School of Medicine, St. Louis, MO, USA
| | | | - Marwan Shinawi
- Department of Pediatrics, Division of Genetics and Genomic Medicine, Washington University in St. Louis School of Medicine, St. Louis, MO, USA
| | - Patricia Dickson
- Department of Pediatrics, Division of Genetics and Genomic Medicine, Washington University in St. Louis School of Medicine, St. Louis, MO, USA
| | - Ali Mian
- Department of Radiology, Section of Neuroradiology, Washington University in St. Louis School of Medicine, St. Louis, MO, USA
| | - Margaret Reynolds
- Department of Ophthalmology and Visual Sciences, Washington University in St. Louis School of Medicine, St. Louis, MO, USA
| | | |
Collapse
|
2
|
Raut S, Singh K, Sanghvi S, Loyo-Celis V, Varghese L, Singh E, Gururaja Rao S, Singh H. Chloride ions in health and disease. Biosci Rep 2024; 44:BSR20240029. [PMID: 38573803 PMCID: PMC11065649 DOI: 10.1042/bsr20240029] [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: 01/09/2024] [Revised: 03/26/2024] [Accepted: 04/04/2024] [Indexed: 04/06/2024] Open
Abstract
Chloride is a key anion involved in cellular physiology by regulating its homeostasis and rheostatic processes. Changes in cellular Cl- concentration result in differential regulation of cellular functions such as transcription and translation, post-translation modifications, cell cycle and proliferation, cell volume, and pH levels. In intracellular compartments, Cl- modulates the function of lysosomes, mitochondria, endosomes, phagosomes, the nucleus, and the endoplasmic reticulum. In extracellular fluid (ECF), Cl- is present in blood/plasma and interstitial fluid compartments. A reduction in Cl- levels in ECF can result in cell volume contraction. Cl- is the key physiological anion and is a principal compensatory ion for the movement of the major cations such as Na+, K+, and Ca2+. Over the past 25 years, we have increased our understanding of cellular signaling mediated by Cl-, which has helped in understanding the molecular and metabolic changes observed in pathologies with altered Cl- levels. Here, we review the concentration of Cl- in various organs and cellular compartments, ion channels responsible for its transportation, and recent information on its physiological roles.
Collapse
Affiliation(s)
- Satish K. Raut
- Department of Physiology and Cell Biology, College of Medicine, The Ohio State University, Columbus, OH, U.S.A
| | - Kulwinder Singh
- Department of Physiology and Cell Biology, College of Medicine, The Ohio State University, Columbus, OH, U.S.A
| | - Shridhar Sanghvi
- Department of Physiology and Cell Biology, College of Medicine, The Ohio State University, Columbus, OH, U.S.A
- Department of Molecular Cellular and Developmental Biology, The Ohio State University, Columbus, OH, U.S.A
| | - Veronica Loyo-Celis
- Department of Physiology and Cell Biology, College of Medicine, The Ohio State University, Columbus, OH, U.S.A
| | - Liyah Varghese
- Department of Physiology and Cell Biology, College of Medicine, The Ohio State University, Columbus, OH, U.S.A
| | - Ekam R. Singh
- Department of Physiology and Cell Biology, College of Medicine, The Ohio State University, Columbus, OH, U.S.A
| | | | - Harpreet Singh
- Department of Physiology and Cell Biology, College of Medicine, The Ohio State University, Columbus, OH, U.S.A
- Department of Molecular Cellular and Developmental Biology, The Ohio State University, Columbus, OH, U.S.A
| |
Collapse
|
3
|
Chávez JC, Carrasquel-Martínez G, Hernández-Garduño S, Matamoros Volante A, Treviño CL, Nishigaki T, Darszon A. Cytosolic and Acrosomal pH Regulation in Mammalian Sperm. Cells 2024; 13:865. [PMID: 38786087 PMCID: PMC11120249 DOI: 10.3390/cells13100865] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2024] [Revised: 05/07/2024] [Accepted: 05/09/2024] [Indexed: 05/25/2024] Open
Abstract
As in most cells, intracellular pH regulation is fundamental for sperm physiology. Key sperm functions like swimming, maturation, and a unique exocytotic process, the acrosome reaction, necessary for gamete fusion, are deeply influenced by pH. Sperm pH regulation, both intracellularly and within organelles such as the acrosome, requires a coordinated interplay of various transporters and channels, ensuring that this cell is primed for fertilization. Consistent with the pivotal importance of pH regulation in mammalian sperm physiology, several of its unique transporters are dependent on cytosolic pH. Examples include the Ca2+ channel CatSper and the K+ channel Slo3. The absence of these channels leads to male infertility. This review outlines the main transport elements involved in pH regulation, including cytosolic and acrosomal pH, that participate in these complex functions. We present a glimpse of how these transporters are regulated and how distinct sets of them are orchestrated to allow sperm to fertilize the egg. Much research is needed to begin to envision the complete set of players and the choreography of how cytosolic and organellar pH are regulated in each sperm function.
Collapse
Affiliation(s)
- Julio C. Chávez
- Departamento de Genética del Desarrollo y Fisiología Molecular, Instituto de Biotecnología (IBT), Universidad Nacional Autónoma de México (UNAM), Cuernavaca 62210, Morelos, Mexico; (J.C.C.); (G.C.-M.)
| | - Gabriela Carrasquel-Martínez
- Departamento de Genética del Desarrollo y Fisiología Molecular, Instituto de Biotecnología (IBT), Universidad Nacional Autónoma de México (UNAM), Cuernavaca 62210, Morelos, Mexico; (J.C.C.); (G.C.-M.)
- CITMER, Medicina Reproductiva, México City 11520, Mexico
| | - Sandra Hernández-Garduño
- Departamento de Morfología, Facultad de Medicina Veterinaria y Zootecnia, Universidad Nacional Autónoma de México (UNAM), México City 04510, Mexico;
| | - Arturo Matamoros Volante
- Department of Electrical and Computer Engineering and School of Biomedical Engineering, Colorado State University, Fort Collins, CO 80523, USA;
| | - Claudia L. Treviño
- Departamento de Genética del Desarrollo y Fisiología Molecular, Instituto de Biotecnología (IBT), Universidad Nacional Autónoma de México (UNAM), Cuernavaca 62210, Morelos, Mexico; (J.C.C.); (G.C.-M.)
| | - Takuya Nishigaki
- Departamento de Genética del Desarrollo y Fisiología Molecular, Instituto de Biotecnología (IBT), Universidad Nacional Autónoma de México (UNAM), Cuernavaca 62210, Morelos, Mexico; (J.C.C.); (G.C.-M.)
| | - Alberto Darszon
- Departamento de Genética del Desarrollo y Fisiología Molecular, Instituto de Biotecnología (IBT), Universidad Nacional Autónoma de México (UNAM), Cuernavaca 62210, Morelos, Mexico; (J.C.C.); (G.C.-M.)
| |
Collapse
|
4
|
Chon NL, Lin H. Fluoride Ion Binding and Translocation in the CLC F Fluoride/Proton Antiporter: Molecular Insights from Combined Quantum-Mechanical/Molecular-Mechanical Modeling. J Phys Chem B 2024; 128:2697-2706. [PMID: 38447081 PMCID: PMC10962343 DOI: 10.1021/acs.jpcb.4c00079] [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: 01/04/2024] [Revised: 02/15/2024] [Accepted: 02/20/2024] [Indexed: 03/08/2024]
Abstract
CLCF fluoride/proton antiporters move fluoride ions out of bacterial cells, leading to fluoride resistance in these bacteria. However, many details about their operating mechanisms remain unclear. Here, we report a combined quantum-mechanical/molecular-mechanical (QM/MM) study of a CLCF homologue from Enterococci casseliflavus (Eca), in accord with the previously proposed windmill mechanism. Our multiscale modeling sheds light on two critical steps in the transport cycle: (i) the external gating residue E118 pushing a fluoride in the external binding site into the extracellular vestibule and (ii) an incoming fluoride reconquering the external binding site by forcing out E118. Both steps feature competitions for the external binding site between the negatively charged carboxylate of E118 and the fluoride. Remarkably, the displaced E118 by fluoride accepts a proton from the nearby R117, initiating the next transport cycle. We also demonstrate the importance of accurate quantum descriptions of fluoride solvation. Our results provide clues to the mysterious E318 residue near the central binding site, suggesting that the transport activities are unlikely to be disrupted by the glutamate interacting with a well-solvated fluoride at the central binding site. This differs significantly from the structurally similar CLC chloride/proton antiporters, where a fluoride trapped deep in the hydrophobic pore causes the transporter to be locked down. A free-energy barrier of 10-15 kcal/mol was estimated via umbrella sampling for a fluoride ion traveling through the pore to repopulate the external binding site.
Collapse
Affiliation(s)
- Nara L. Chon
- Department of Chemistry, University of Colorado Denver, Denver, Colorado 80217, United States
| | - Hai Lin
- Department of Chemistry, University of Colorado Denver, Denver, Colorado 80217, United States
| |
Collapse
|
5
|
Huang WL, Wang XD, Ao YF, Wang QQ, Wang DX. Reversing the ion transport selectivity through arm modification of an artificial molecular hourglass. Chem Commun (Camb) 2023. [PMID: 37997041 DOI: 10.1039/d3cc04573k] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2023]
Abstract
An arm modification strategy, by replacing relatively rigid, electron-deficient side arms with flexible ether chain arms and linking them onto a tetraoxacalix[2]arene[2]triazine skeleton, was utilized to design an artificial molecular hourglass. The planar bilayer experiments confirmed the unimolecular channel mechanism and suggested reversed ion selectivity from the previously reported anion selectivity to weak cation selectivity.
Collapse
Affiliation(s)
- Wen-Long Huang
- Beijing National Laboratory for Molecular Sciences, CAS Key Laboratory of Molecular Recognition and Function, Institute of Chemistry, Chinese Academy of Sciences, Beijing 100190, China.
| | - Xu-Dong Wang
- Beijing National Laboratory for Molecular Sciences, CAS Key Laboratory of Molecular Recognition and Function, Institute of Chemistry, Chinese Academy of Sciences, Beijing 100190, China.
| | - Yu-Fei Ao
- Beijing National Laboratory for Molecular Sciences, CAS Key Laboratory of Molecular Recognition and Function, Institute of Chemistry, Chinese Academy of Sciences, Beijing 100190, China.
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Qi-Qiang Wang
- Beijing National Laboratory for Molecular Sciences, CAS Key Laboratory of Molecular Recognition and Function, Institute of Chemistry, Chinese Academy of Sciences, Beijing 100190, China.
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - De-Xian Wang
- Beijing National Laboratory for Molecular Sciences, CAS Key Laboratory of Molecular Recognition and Function, Institute of Chemistry, Chinese Academy of Sciences, Beijing 100190, China.
- University of Chinese Academy of Sciences, Beijing 100049, China
| |
Collapse
|
6
|
Zhang B, Zhang S, Polovitskaya MM, Yi J, Ye B, Li R, Huang X, Yin J, Neuens S, Balfroid T, Soblet J, Vens D, Aeby A, Li X, Cai J, Song Y, Li Y, Tartaglia M, Li Y, Jentsch TJ, Yang M, Liu Z. Molecular basis of ClC-6 function and its impairment in human disease. SCIENCE ADVANCES 2023; 9:eadg4479. [PMID: 37831762 PMCID: PMC10575590 DOI: 10.1126/sciadv.adg4479] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/25/2022] [Accepted: 09/08/2023] [Indexed: 10/15/2023]
Abstract
ClC-6 is a late endosomal voltage-gated chloride-proton exchanger that is predominantly expressed in the nervous system. Mutated forms of ClC-6 are associated with severe neurological disease. However, the mechanistic role of ClC-6 in normal and pathological states remains largely unknown. Here, we present cryo-EM structures of ClC-6 that guided subsequent functional studies. Previously unrecognized ATP binding to cytosolic ClC-6 domains enhanced ion transport activity. Guided by a disease-causing mutation (p.Y553C), we identified an interaction network formed by Y553/F317/T520 as potential hotspot for disease-causing mutations. This was validated by the identification of a patient with a de novo pathogenic variant p.T520A. Extending these findings, we found contacts between intramembrane helices and connecting loops that modulate the voltage dependence of ClC-6 gating and constitute additional candidate regions for disease-associated gain-of-function mutations. Besides providing insights into the structure, function, and regulation of ClC-6, our work correctly predicts hotspots for CLCN6 mutations in neurodegenerative disorders.
Collapse
Affiliation(s)
- Bing Zhang
- Shanghai Key Laboratory of Maternal Fetal Medicine, Shanghai Institute of Maternal-Fetal Medicine and Gynecologic Oncology, Department of Anesthesiology, Clinical and Translational Research Center, Shanghai First Maternity and Infant Hospital, School of Medicine, Tongji University, 201204 Shanghai, China
| | - Sensen Zhang
- Ministry of Education Key Laboratory of Protein Science, Beijing Advanced Innovation Center for Structural Biology, Beijing Frontier Research Center for Biological Structure, Tsinghua-Peking Center for Life Sciences, School of Life Sciences, Tsinghua University, 100084 Beijing, China
| | - Maya M. Polovitskaya
- Leibniz-Forschungsinstitut für Molekulare Pharmakologie (FMP), 13125 Berlin, Germany
- Max-Delbrück-Centrum für Molekulare Medizin (MDC), 13125 Berlin, Germany
| | - Jingbo Yi
- Ministry of Education Key Laboratory of Protein Science, Beijing Advanced Innovation Center for Structural Biology, Beijing Frontier Research Center for Biological Structure, Tsinghua-Peking Center for Life Sciences, School of Life Sciences, Tsinghua University, 100084 Beijing, China
| | - Binglu Ye
- Shanghai Key Laboratory of Maternal Fetal Medicine, Shanghai Institute of Maternal-Fetal Medicine and Gynecologic Oncology, Department of Anesthesiology, Clinical and Translational Research Center, Shanghai First Maternity and Infant Hospital, School of Medicine, Tongji University, 201204 Shanghai, China
| | - Ruochong Li
- Ministry of Education Key Laboratory of Protein Science, Beijing Advanced Innovation Center for Structural Biology, Beijing Frontier Research Center for Biological Structure, Tsinghua-Peking Center for Life Sciences, School of Life Sciences, Tsinghua University, 100084 Beijing, China
| | - Xueying Huang
- Shanghai Key Laboratory of Maternal Fetal Medicine, Shanghai Institute of Maternal-Fetal Medicine and Gynecologic Oncology, Department of Anesthesiology, Clinical and Translational Research Center, Shanghai First Maternity and Infant Hospital, School of Medicine, Tongji University, 201204 Shanghai, China
| | - Jian Yin
- Ministry of Education Key Laboratory of Protein Science, Beijing Advanced Innovation Center for Structural Biology, Beijing Frontier Research Center for Biological Structure, Tsinghua-Peking Center for Life Sciences, School of Life Sciences, Tsinghua University, 100084 Beijing, China
| | - Sebastian Neuens
- Department of Genetics, Hôpital Universitaire des Enfants Reine Fabiola, Université Libre de Bruxelles (ULB), Brussels, Belgium
| | - Tom Balfroid
- Department of Pediatric Neurology, Hôpital Universitaire des Enfants Reine Fabiola, Université Libre de Bruxelles (ULB), Brussels, Belgium
| | - Julie Soblet
- Department of Genetics, Hôpital Universitaire des Enfants Reine Fabiola, Université Libre de Bruxelles (ULB), Brussels, Belgium
- Department of Genetics, Hôpital Erasme, Université Libre de Bruxelles (ULB), Brussels, Belgium
- Interuniversity Institute of Bioinformatics in Brussels, Université Libre de Bruxelles (ULB), Brussels, Belgium
| | - Daphné Vens
- Pediatric Intensive Care Unit, Hôpital Universitaire des Enfants Reine Fabiola, Université Libre de Bruxelles (ULB), Brussels, Belgium
| | - Alec Aeby
- Department of Pediatric Neurology, Hôpital Universitaire des Enfants Reine Fabiola, Université Libre de Bruxelles (ULB), Brussels, Belgium
| | - Xiaoling Li
- Wuya College of Innovation, Shenyang Pharmaceutical University, 110016 Shenyang, China
| | - Jinjin Cai
- Key Laboratory of Receptor Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, 201203 Shanghai, China
| | - Yingcai Song
- Shanghai Key Laboratory of Maternal Fetal Medicine, Shanghai Institute of Maternal-Fetal Medicine and Gynecologic Oncology, Department of Anesthesiology, Clinical and Translational Research Center, Shanghai First Maternity and Infant Hospital, School of Medicine, Tongji University, 201204 Shanghai, China
| | - Yuanxi Li
- Institute for Cognitive Neurodynamics, School of Mathematics, East China University of Science and Technology, 200237 Shanghai, China
| | - Marco Tartaglia
- Molecular Genetics and Functional Genomics, Ospedale Pediatrico Bambino Gesù, IRCCS, 00146 Rome, Italy
| | - Yang Li
- Key Laboratory of Receptor Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, 201203 Shanghai, China
- National Clinical Research Center for Aging and Medicine, Huashan Hospital, Fudan University, Shanghai 200040, China
| | - Thomas J. Jentsch
- Leibniz-Forschungsinstitut für Molekulare Pharmakologie (FMP), 13125 Berlin, Germany
- Max-Delbrück-Centrum für Molekulare Medizin (MDC), 13125 Berlin, Germany
- NeuroCure Cluster of Excellence, Charité Universitätsmedizin Berlin, 10117 Berlin, Germany
| | - Maojun Yang
- Ministry of Education Key Laboratory of Protein Science, Beijing Advanced Innovation Center for Structural Biology, Beijing Frontier Research Center for Biological Structure, Tsinghua-Peking Center for Life Sciences, School of Life Sciences, Tsinghua University, 100084 Beijing, China
- Cryo-EM Facility Center, Southern University of Science & Technology, 518055 Shenzhen, Guangdong, China
| | - Zhiqiang Liu
- Shanghai Key Laboratory of Maternal Fetal Medicine, Shanghai Institute of Maternal-Fetal Medicine and Gynecologic Oncology, Department of Anesthesiology, Clinical and Translational Research Center, Shanghai First Maternity and Infant Hospital, School of Medicine, Tongji University, 201204 Shanghai, China
| |
Collapse
|
7
|
Yang Z, Zhang X, Ye S, Zheng J, Huang X, Yu F, Chen Z, Cai S, Zhang P. Molecular mechanism underlying regulation of Arabidopsis CLCa transporter by nucleotides and phospholipids. Nat Commun 2023; 14:4879. [PMID: 37573431 PMCID: PMC10423218 DOI: 10.1038/s41467-023-40624-z] [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: 05/12/2023] [Accepted: 08/03/2023] [Indexed: 08/14/2023] Open
Abstract
Chloride channels (CLCs) transport anion across membrane to regulate ion homeostasis and acidification of intracellular organelles, and are divided into anion channels and anion/proton antiporters. Arabidopsis thaliana CLCa (AtCLCa) transporter localizes to the tonoplast which imports NO3- and to a less extent Cl- from cytoplasm. The activity of AtCLCa and many other CLCs is regulated by nucleotides and phospholipids, however, the molecular mechanism remains unclear. Here we determine the cryo-EM structures of AtCLCa bound with NO3- and Cl-, respectively. Both structures are captured in ATP and PI(4,5)P2 bound conformation. Structural and electrophysiological analyses reveal a previously unidentified N-terminal β-hairpin that is stabilized by ATP binding to block the anion transport pathway, thereby inhibiting the AtCLCa activity. While AMP loses the inhibition capacity due to lack of the β/γ- phosphates required for β-hairpin stabilization. This well explains how AtCLCa senses the ATP/AMP status to regulate the physiological nitrogen-carbon balance. Our data further show that PI(4,5)P2 or PI(3,5)P2 binds to the AtCLCa dimer interface and occupies the proton-exit pathway, which may help to understand the inhibition of AtCLCa by phospholipids to facilitate guard cell vacuole acidification and stomatal closure. In a word, our work suggests the regulatory mechanism of AtCLCa by nucleotides and phospholipids under certain physiological scenarios and provides new insights for future study of CLCs.
Collapse
Affiliation(s)
- Zhao Yang
- National Key Laboratory of Plant Molecular Genetics, Center for Excellence in Molecular Plant Sciences, Institute of Plant Physiology and Ecology, Chinese Academy of Sciences, Shanghai, 200032, China
- University of Chinese Academy of Sciences, Beijing, 100039, China
| | - Xue Zhang
- National Key Laboratory of Plant Molecular Genetics, Center for Excellence in Molecular Plant Sciences, Institute of Plant Physiology and Ecology, Chinese Academy of Sciences, Shanghai, 200032, China
| | - Shiwei Ye
- University of Chinese Academy of Sciences, Beijing, 100039, China
- Center for Excellence in Brain Sciences and Intelligence Technology, Institute of Neuronscience, Chinese Academy of Sciences, Shanghai, 200031, China
| | - Jingtao Zheng
- Shanghai Key Laboratory of Plant Molecular Sciences, College of Life Sciences, Shanghai Normal University, Shanghai, 200234, China
| | - Xiaowei Huang
- National Key Laboratory of Plant Molecular Genetics, Center for Excellence in Molecular Plant Sciences, Institute of Plant Physiology and Ecology, Chinese Academy of Sciences, Shanghai, 200032, China
- University of Chinese Academy of Sciences, Beijing, 100039, China
| | - Fang Yu
- Shanghai Key Laboratory of Plant Molecular Sciences, College of Life Sciences, Shanghai Normal University, Shanghai, 200234, China
| | - Zhenguo Chen
- The Fifth People's Hospital of Shanghai, Institutes of Biomedical Sciences, School of Basic Medical Sciences, Fudan University, Shanghai, 200032, China.
| | - Shiqing Cai
- Center for Excellence in Brain Sciences and Intelligence Technology, Institute of Neuronscience, Chinese Academy of Sciences, Shanghai, 200031, China.
| | - Peng Zhang
- National Key Laboratory of Plant Molecular Genetics, Center for Excellence in Molecular Plant Sciences, Institute of Plant Physiology and Ecology, Chinese Academy of Sciences, Shanghai, 200032, China.
| |
Collapse
|
8
|
Holla VV, Phulpagar P, Saini J, Kamble N, Pal PK, Yadav R, Muthusamy B, Netravathi M. CLCN2-Related Leukoencephalopathy in Two Unrelated Patients Due to Novel Variants. Mov Disord Clin Pract 2023; 10:1155-1158. [PMID: 37476307 PMCID: PMC10354607 DOI: 10.1002/mdc3.13783] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2022] [Revised: 04/12/2023] [Accepted: 05/04/2023] [Indexed: 07/22/2023] Open
Affiliation(s)
- Vikram V. Holla
- Department of NeurologyNational Institute of Mental Health and NeurosciencesBengaluruIndia
| | - Prashant Phulpagar
- Institute of Bioinformatics, International Technology ParkBengaluruIndia
- Manipal Academy of Higher EducationManipalIndia
| | - Jitender Saini
- Department of Neuroimaging and Interventional RadiologyNational Institute of Mental Health and NeurosciencesBengaluruIndia
| | - Nitish Kamble
- Department of NeurologyNational Institute of Mental Health and NeurosciencesBengaluruIndia
| | - Pramod Kumar Pal
- Department of NeurologyNational Institute of Mental Health and NeurosciencesBengaluruIndia
| | - Ravi Yadav
- Department of NeurologyNational Institute of Mental Health and NeurosciencesBengaluruIndia
| | - Babylakshmi Muthusamy
- Institute of Bioinformatics, International Technology ParkBengaluruIndia
- Manipal Academy of Higher EducationManipalIndia
| | - Manjunath Netravathi
- Department of NeurologyNational Institute of Mental Health and NeurosciencesBengaluruIndia
| |
Collapse
|
9
|
Stevanović KS, Čepkenović B, Križak S, Živić MŽ, Todorović NV. Osmotically Activated Anion Current of Phycomyces Blakesleeanus-Filamentous Fungi Counterpart to Vertebrate Volume Regulated Anion Current. J Fungi (Basel) 2023; 9:637. [PMID: 37367573 DOI: 10.3390/jof9060637] [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: 05/08/2023] [Revised: 05/21/2023] [Accepted: 05/26/2023] [Indexed: 06/28/2023] Open
Abstract
Studies of ion currents in filamentous fungi are a prerequisite for forming a complete understanding of their physiology. Cytoplasmic droplets (CDs), obtained from sporangiophores of Phycomyces blakesleeanus, are a model system that enables the characterization of ion currents in the native membrane, including the currents mediated by the channels not yet molecularly identified. Osmotically activated anionic current with outward rectification (ORIC) is a dominant current in the membrane of cytoplasmic droplets under the conditions of hypoosmotic stimulation. We have previously reported remarkable functional similarities of ORIC with the vertebrate volume regulated anionic current (VRAC), such as dose-dependent activation by osmotic difference, ion selectivity sequence, and time and voltage dependent profile of the current. Using the patch clamp method on the CD membrane, we further resolve VRAC-like ORIC characteristics in this paper. We examine the inhibition by extracellular ATP and carbenoxolone, the permeation of glutamate in presence of chloride, selectivity for nitrates, and activation by GTP, and we show its single channel behavior in excised membrane. We propose that ORIC is a functional counterpart of vertebrate VRAC in filamentous fungi, possibly with a similar essential role in anion efflux during cell volume regulation.
Collapse
Affiliation(s)
- Katarina S Stevanović
- Faculty of Biology, Institute of Physiology and Biochemistry, University of Belgrade, Studentski Trg 16, 11158 Belgrade, Serbia
| | - Bogdana Čepkenović
- Faculty of Biology, Institute of Physiology and Biochemistry, University of Belgrade, Studentski Trg 16, 11158 Belgrade, Serbia
| | - Strahinja Križak
- Institute of Multidisciplinary Research, University of Belgrade, Kneza Višeslava 1, 11030 Belgrade, Serbia
| | - Miroslav Ž Živić
- Faculty of Biology, Institute of Physiology and Biochemistry, University of Belgrade, Studentski Trg 16, 11158 Belgrade, Serbia
| | - Nataša V Todorović
- Institute of Biological Research "Siniša Stanković", National Institute of the Republic of Serbia, University of Belgrade, Bulevar Despota Stefana 142, 11000 Belgrade, Serbia
| |
Collapse
|
10
|
The mechanisms of chromogranin B-regulated Cl- homeostasis. Biochem Soc Trans 2022; 50:1659-1672. [PMID: 36511243 DOI: 10.1042/bst20220435] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2022] [Revised: 11/25/2022] [Accepted: 12/01/2022] [Indexed: 12/15/2022]
Abstract
Chloride is the most abundant inorganic anions in almost all cells and in human circulation systems. Its homeostasis is therefore important for systems physiology and normal cellular activities. This topic has been extensively studied with chloride loaders and extruders expressed in both cell surfaces and intracellular membranes. With the newly discovered, large-conductance, highly selective Cl- channel formed by membrane-bound chromogranin B (CHGB), which differs from all other known anion channels of conventional transmembrane topology, and is distributed in plasma membranes, endomembrane systems, endosomal, and endolysosomal compartments in cells expressing it, we will discuss the potential physiological importance of the CHGB channels to Cl- homeostasis, cellular excitability and volume control, and cation uptake or release at the cellular and subcellular levels. These considerations and CHGB's association with human diseases make the CHGB channel a possible druggable target for future molecular therapeutics.
Collapse
|
11
|
Dukic E, Gollan PJ, Grebe S, Paakkarinen V, Herdean A, Aro EM, Spetea C. The Arabidopsis thylakoid chloride channel ClCe regulates ATP availability for light-harvesting complex II protein phosphorylation. FRONTIERS IN PLANT SCIENCE 2022; 13:1050355. [PMID: 36483957 PMCID: PMC9722747 DOI: 10.3389/fpls.2022.1050355] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 09/21/2022] [Accepted: 10/31/2022] [Indexed: 06/17/2023]
Abstract
Coping with changes in light intensity is challenging for plants, but well-designed mechanisms allow them to acclimate to most unpredicted situations. The thylakoid K+/H+ antiporter KEA3 and the voltage-dependent Cl- channel VCCN1 play important roles in light acclimation by fine-tuning electron transport and photoprotection. Good evidence exists that the thylakoid Cl- channel ClCe is involved in the regulation of photosynthesis and state transitions in conditions of low light. However, a detailed mechanistic understanding of this effect is lacking. Here we report that the ClCe loss-of-function in Arabidopsis thaliana results in lower levels of phosphorylated light-harvesting complex II (LHCII) proteins as well as lower levels of the photosystem I-LHCII complexes relative to wild type (WT) in low light conditions. The phosphorylation of the photosystem II core D1/D2 proteins was less affected either in low or high light conditions. In low light conditions, the steady-state levels of ATP synthase conductivity and of the total proton flux available for ATP synthesis were lower in ClCe loss-of-function mutants, but comparable to WT at standard and high light intensity. As a long-term acclimation strategy, expression of the ClCe gene was upregulated in WT plants grown in light-limiting conditions, but not in WT plants grown in standard light even when exposed for up to 8 h to low light. Taken together, these results suggest a role of ClCe in the regulation of the ATP synthase activity which under low light conditions impacts LHCII protein phosphorylation and state transitions.
Collapse
Affiliation(s)
- Emilija Dukic
- Department of Biological and Environmental Sciences, University of Gothenburg, Gothenburg, Sweden
| | - Peter J. Gollan
- Molecular Plant Biology Unit, Department of Life Technologies, University of Turku, Turku, Finland
| | - Steffen Grebe
- Molecular Plant Biology Unit, Department of Life Technologies, University of Turku, Turku, Finland
| | - Virpi Paakkarinen
- Molecular Plant Biology Unit, Department of Life Technologies, University of Turku, Turku, Finland
| | - Andrei Herdean
- Climate Change Cluster, University of Technology Sydney, Ultimo, NSW, Australia
| | - Eva-Mari Aro
- Molecular Plant Biology Unit, Department of Life Technologies, University of Turku, Turku, Finland
| | - Cornelia Spetea
- Department of Biological and Environmental Sciences, University of Gothenburg, Gothenburg, Sweden
| |
Collapse
|
12
|
Zhao R, Cao Y, Ge Y, Xu J, Li R, Yang M, Chen Y, Wu D, Xiao J, Li R. Single-Molecule and Vesicle Trafficking Analysis of Ubiquitination Involved in the Activity of Ammonium Transporter AMT1;3 in Arbidopsis under High Ammonium Stress. Cells 2022; 11:cells11223651. [PMID: 36429077 PMCID: PMC9688738 DOI: 10.3390/cells11223651] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2022] [Revised: 11/12/2022] [Accepted: 11/14/2022] [Indexed: 11/19/2022] Open
Abstract
Plants absorb nitrogen from the soil using ammonium transporters (AMTs). Plants can precisely regulate AMT1;3 levels using sophisticated regulatory systems, ensuring adequate nitrogen uptake without hazardous ammonium production. Here, we demonstrated that ubiquitylation can contribute to AMT1;3 degradation under high ammonium stress. Using the ubiquitin site mutant AMT1;3K75R,K233R-EGFP, we demonstrated that the loss of ubiquitination affects the dynamic characteristics of AMT1;3 proteins on the plasma membrane and markedly inhibits the endocytosis of AMT1;3 proteins under high ammonium stress. AMT1;3K75R,K233R-EGFP plants also showed inhibition of protein degradation that targets the vesicular pathway after being exposed to high levels of ammonium. Our findings showed that the dynamic properties, endocytosis, and vesicle trafficking pathways of AMT1;3 proteins are altered in AMT1;3K75R,K233R-EGFP under high ammonium conditions.
Collapse
Affiliation(s)
- Ran Zhao
- National Engineering Research Center of Tree Breeding and Ecological Restoration, College of Biological Sciences and Biotechnology, Beijing Forestry University, Beijing 100083, China
- Institute of Tree Development and Genome Editing, Beijing Forestry University, Beijing 100083, China
| | - Yangyang Cao
- National Engineering Research Center of Tree Breeding and Ecological Restoration, College of Biological Sciences and Biotechnology, Beijing Forestry University, Beijing 100083, China
- Institute of Tree Development and Genome Editing, Beijing Forestry University, Beijing 100083, China
- Shijiazhuang Zhonghua Avenue Primary School, Shijiazhuang 050000, China
| | - Yanrui Ge
- National Engineering Research Center of Tree Breeding and Ecological Restoration, College of Biological Sciences and Biotechnology, Beijing Forestry University, Beijing 100083, China
- Institute of Tree Development and Genome Editing, Beijing Forestry University, Beijing 100083, China
| | - Jing Xu
- National Engineering Research Center of Tree Breeding and Ecological Restoration, College of Biological Sciences and Biotechnology, Beijing Forestry University, Beijing 100083, China
- Institute of Tree Development and Genome Editing, Beijing Forestry University, Beijing 100083, China
| | - Ruofan Li
- National Engineering Research Center of Tree Breeding and Ecological Restoration, College of Biological Sciences and Biotechnology, Beijing Forestry University, Beijing 100083, China
- Institute of Tree Development and Genome Editing, Beijing Forestry University, Beijing 100083, China
| | - Mei Yang
- National Engineering Research Center of Tree Breeding and Ecological Restoration, College of Biological Sciences and Biotechnology, Beijing Forestry University, Beijing 100083, China
- Institute of Tree Development and Genome Editing, Beijing Forestry University, Beijing 100083, China
| | - Yingying Chen
- National Engineering Research Center of Tree Breeding and Ecological Restoration, College of Biological Sciences and Biotechnology, Beijing Forestry University, Beijing 100083, China
- Institute of Tree Development and Genome Editing, Beijing Forestry University, Beijing 100083, China
| | - Dingjie Wu
- National Engineering Research Center of Tree Breeding and Ecological Restoration, College of Biological Sciences and Biotechnology, Beijing Forestry University, Beijing 100083, China
- Institute of Tree Development and Genome Editing, Beijing Forestry University, Beijing 100083, China
| | - Jianwei Xiao
- National Engineering Research Center of Tree Breeding and Ecological Restoration, College of Biological Sciences and Biotechnology, Beijing Forestry University, Beijing 100083, China
- Institute of Tree Development and Genome Editing, Beijing Forestry University, Beijing 100083, China
| | - Ruili Li
- National Engineering Research Center of Tree Breeding and Ecological Restoration, College of Biological Sciences and Biotechnology, Beijing Forestry University, Beijing 100083, China
- Institute of Tree Development and Genome Editing, Beijing Forestry University, Beijing 100083, China
- Correspondence:
| |
Collapse
|
13
|
Elson A, Anuj A, Barnea-Zohar M, Reuven N. The origins and formation of bone-resorbing osteoclasts. Bone 2022; 164:116538. [PMID: 36028118 DOI: 10.1016/j.bone.2022.116538] [Citation(s) in RCA: 31] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/09/2022] [Revised: 08/17/2022] [Accepted: 08/18/2022] [Indexed: 02/07/2023]
Abstract
Osteoclasts (OCLs) are hematopoietic cells whose physiological function is to degrade bone. OCLs are key players in the processes that determine and maintain the mass, shape, and physical properties of bone. OCLs adhere to bone tightly and degrade its matrix by secreting protons and proteases onto the underlying surface. The combination of low pH and proteases degrades the mineral and protein components of the matrix and forms a resorption pit; the degraded material is internalized by the cell and then secreted into the circulation. Insufficient or excessive activity of OCLs can lead to significant changes in bone and either cause or exacerbate symptoms of diseases, as in osteoporosis, osteopetrosis, and cancer-induced bone lysis. OCLs are derived from monocyte-macrophage precursor cells whose origins are in two distinct embryonic cell lineages - erythromyeloid progenitor cells of the yolk sac, and hematopoietic stem cells. OCLs are formed in a multi-stage process that is induced by the cytokines M-CSF and RANKL, during which the cells differentiate, fuse to form multi-nucleated cells, and then differentiate further to become mature, bone-resorbing OCLs. Recent studies indicate that OCLs can undergo fission in vivo to generate smaller cells, called "osteomorphs", that can be "re-cycled" by fusing with other cells to form new OCLs. In this review we describe OCLs and discuss their cellular origins and the cellular and molecular events that drive osteoclastogenesis.
Collapse
Affiliation(s)
- Ari Elson
- Department of Molecular Genetics, The Weizmann Institute of Science, Rehovot 76100, Israel.
| | - Anuj Anuj
- Department of Molecular Genetics, The Weizmann Institute of Science, Rehovot 76100, Israel
| | - Maayan Barnea-Zohar
- Department of Molecular Genetics, The Weizmann Institute of Science, Rehovot 76100, Israel
| | - Nina Reuven
- Department of Molecular Genetics, The Weizmann Institute of Science, Rehovot 76100, Israel
| |
Collapse
|
14
|
Jingxuan L, Litian M, Yanyang T, Jianfang F. Knockdown of CLC-3 may improve cognitive impairment caused by diabetic encephalopathy. Diabetes Res Clin Pract 2022; 190:109970. [PMID: 35792204 DOI: 10.1016/j.diabres.2022.109970] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/01/2022] [Revised: 06/05/2022] [Accepted: 06/16/2022] [Indexed: 11/03/2022]
Abstract
BACKGROUND Diabetic encephalopathy(DE) is a neurological complication of diabetes, and its pathogenesis is unclear. Current studies indicate that insulin receptors and downstream signaling pathways play a key role in the occurrence and development of DE. Additionally, CLC-3, a member of the CLC family of anion channels and transporters, is closely related to the secretion and processing of insulin. Here, we investigated the changes and putative roles of CLC-3 in diabetic encephalopathy. RESULTS To this aim, we combined lentivirus and adeno-associated virus gene transfer to change the expression level of CLC-3 in the HT-22 hippocampal cell line and hippocampal CA1. We studied the role of CLC-3 in DE through the Morris water maze test.CLC-3 expression increased significantly in HT-22 cells cultured with high glucose and STZ-induced DE model hippocampus. Moreover, Insulin receptor(IR) and downstream PI3K/AKT/GSK3β signaling pathways were also dysfunctional. After knocking down CLC-3, impaired cell proliferation, apoptosis, IR and the downstream PI3K/AKT/GSK3β signaling pathways were significantly improved. However, when CLC-3 was overexpressed, the neurotoxicity induced by high glucose was further aggravated. Rescue experiments found that through the use of inhibitors such as GSK3β, the PI3K/AKT/GSK3β signaling pathways pathway changes with the use of inhibition, and the expression of related downstream signaling molecules such as Tau and p-Tau also changes accordingly. Using adeno-associated virus gene transfer to knock down CLC-3 in the hippocampal CA1 of the DE model, the IR caused by DE and the dysfunction of the downstream PI3K/AKT/GSK3β signaling pathway were significantly improved. In addition, the impaired spatial recognition of DE was partially restored. CONCLUSION Our study proposes that CLC-3, as a key molecule, may regulate insulin receptor signaling and downstream PI3K/AKT/GSK3β signaling pathways and affect the pathogenesis of diabetic encephalopathy.
Collapse
Affiliation(s)
- Lian Jingxuan
- Department of Endocrinology, Xijing Hospital, The Air Force Medical University, Xi'an 710032, China
| | - Ma Litian
- Department of Gastroenterology, Tangdu Hospital, The Air Force Medical University, Xi'an 710038, China
| | - Tu Yanyang
- The Air Force Medical University, Xi'an 710032, China.
| | - Fu Jianfang
- Department of Endocrinology, Xijing Hospital, The Air Force Medical University, Xi'an 710032, China.
| |
Collapse
|
15
|
Williamson G, Brito AS, Bizior A, Tamburrino G, Dias Mirandela G, Harris T, Hoskisson PA, Zachariae U, Marini AM, Boeckstaens M, Javelle A. Coexistence of Ammonium Transporter and Channel Mechanisms in Amt-Mep-Rh Twin-His Variants Impairs the Filamentation Signaling Capacity of Fungal Mep2 Transceptors. mBio 2022; 13:e0291321. [PMID: 35196127 PMCID: PMC9040831 DOI: 10.1128/mbio.02913-21] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2021] [Accepted: 01/26/2022] [Indexed: 12/01/2022] Open
Abstract
Ammonium translocation through biological membranes, by the ubiquitous Amt-Mep-Rh family of transporters, plays a key role in all domains of life. Two highly conserved histidine residues protrude into the lumen of the pore of these transporters, forming the family's characteristic Twin-His motif. It has been hypothesized that the motif is essential to confer the selectivity of the transport mechanism. Here, using a combination of in vitro electrophysiology on Escherichia coli AmtB, in silico molecular dynamics simulations, and in vivo yeast functional complementation assays, we demonstrate that variations in the Twin-His motif trigger a mechanistic switch between a specific transporter, depending on ammonium deprotonation, to an unspecific ion channel activity. We therefore propose that there is no selective filter that governs specificity in Amt-Mep-Rh transporters, but the inherent mechanism of translocation, dependent on the fragmentation of the substrate, ensures the high specificity of the translocation. We show that coexistence of both mechanisms in single Twin-His variants of yeast Mep2 transceptors disrupts the signaling function and so impairs fungal filamentation. These data support a signaling process driven by the transport mechanism of the fungal Mep2 transceptors. IMPORTANCE Fungal infections represent a significant threat to human health and cause huge damage to crop yields worldwide. The dimorphic switch between yeast and filamentous growth is associated with the virulence of pathogenic fungi. Of note, fungal Mep2 proteins of the conserved Amt-Mep-Rh family play a transceptor role in the induction of filamentation; however, the signaling mechanism remains largely unknown. Amt-Mep-Rh proteins ensure the specific scavenging of NH4+ through a mechanism relying on substrate deprotonation, thereby preventing competition and translocation of similar-sized K+. Our multidisciplinary approaches using E. coli AmtB, Saccharomyces cerevisiae, and Candida albicans Mep2 show that double variation of the family-defining Twin-His motif triggers a mechanistic switch from a specific transporter to an unspecific ion channel with both mechanisms still coexisting in single variants. Moreover, we show that this mechanistic alteration is associated with loss of signaling ability of Mep2, supporting a transport mechanism-driven process in filamentation induction.
Collapse
Affiliation(s)
- Gordon Williamson
- Strathclyde Institute of Pharmacy and Biomedical Sciences, University of Strathclyde, Glasgow, United Kingdom
| | - Ana Sofia Brito
- Biology of Membrane Transport Laboratory, Department of Molecular Biology, Université Libre de Bruxelles, Gosselies, Belgium
| | - Adriana Bizior
- Strathclyde Institute of Pharmacy and Biomedical Sciences, University of Strathclyde, Glasgow, United Kingdom
| | - Giulia Tamburrino
- Computational Biology, School of Life Sciences, University of Dundee, Dundee, United Kingdom
| | - Gaëtan Dias Mirandela
- Strathclyde Institute of Pharmacy and Biomedical Sciences, University of Strathclyde, Glasgow, United Kingdom
- Biology of Membrane Transport Laboratory, Department of Molecular Biology, Université Libre de Bruxelles, Gosselies, Belgium
| | - Thomas Harris
- Strathclyde Institute of Pharmacy and Biomedical Sciences, University of Strathclyde, Glasgow, United Kingdom
| | - Paul A. Hoskisson
- Strathclyde Institute of Pharmacy and Biomedical Sciences, University of Strathclyde, Glasgow, United Kingdom
| | - Ulrich Zachariae
- Computational Biology, School of Life Sciences, University of Dundee, Dundee, United Kingdom
| | - Anna Maria Marini
- Biology of Membrane Transport Laboratory, Department of Molecular Biology, Université Libre de Bruxelles, Gosselies, Belgium
| | - Mélanie Boeckstaens
- Biology of Membrane Transport Laboratory, Department of Molecular Biology, Université Libre de Bruxelles, Gosselies, Belgium
| | - Arnaud Javelle
- Strathclyde Institute of Pharmacy and Biomedical Sciences, University of Strathclyde, Glasgow, United Kingdom
| |
Collapse
|
16
|
Miszta P, Nazaruk E, Nieciecka D, Możajew M, Krysiński P, Bilewicz R, Filipek S. The EcCLC antiporter embedded in lipidic liquid crystalline films - molecular dynamics simulations and electrochemical methods. Phys Chem Chem Phys 2022; 24:3066-3077. [PMID: 35040466 DOI: 10.1039/d1cp03992j] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Lipidic-liquid crystalline nanostructures (lipidic cubic phases), which are biomimetic and stable in an excess of water, were used as a convenient environment to investigate the transport properties of the membrane antiporter E. coli CLC-1 (EcCLC). The chloride ion transfer by EcCLC was studied by all-atom molecular dynamics simulations combined with electrochemical methods at pH 7 and pH 5. The cubic phase film was used as the membrane between the chloride donor and receiving compartments and it was placed on the glassy carbon electrode and immersed in the chloride solution. Structural characterization of lipidic mesoscopic systems with and without the incorporation of EcCLC was performed using small-angle X-ray scattering. The EcCLC transported chloride ions more efficiently at more acidic pH, and the resistance of the film decreased at lower pH. 4,4-Diisothiocyanatostilbene-2,2'-disulfonic acid (DIDS) employed as an inhibitor of the protein was shown to decrease the transport efficiency upon hydrolysis to DADS at both pH 7 and pH 5. The molecular dynamics simulations, performed for the first time in lipidic cubic phases for EcCLC, allowed studying the collective movements of chloride ions which can help in elucidating the mechanism of transporting the ions by the EcCLC antiporter. The protein modified lipidic cubic phase film is a convenient and simple system for screening potential inhibitors of integral membrane proteins, as demonstrated by the example of the EcCLC antiporter. The use of lipidic cubic phases may also be important for the further development of new electrochemical sensors for membrane proteins and enzyme electrodes.
Collapse
Affiliation(s)
- Przemysław Miszta
- Biological and Chemical Research Centre, University of Warsaw, Zwirki i Wigury 101, 02-089 Warsaw, Poland. .,Faculty of Chemistry, University of Warsaw, Pasteura 1, 02-093 Warsaw, Poland.
| | - Ewa Nazaruk
- Faculty of Chemistry, University of Warsaw, Pasteura 1, 02-093 Warsaw, Poland.
| | - Dorota Nieciecka
- Faculty of Chemistry, University of Warsaw, Pasteura 1, 02-093 Warsaw, Poland.
| | - Mariusz Możajew
- Biological and Chemical Research Centre, University of Warsaw, Zwirki i Wigury 101, 02-089 Warsaw, Poland. .,Faculty of Chemistry, University of Warsaw, Pasteura 1, 02-093 Warsaw, Poland. .,Nencki Institute of Experimental Biology, Polish Academy of Sciences, Pasteura 3, 02-093 Warsaw, Poland
| | - Paweł Krysiński
- Faculty of Chemistry, University of Warsaw, Pasteura 1, 02-093 Warsaw, Poland.
| | - Renata Bilewicz
- Biological and Chemical Research Centre, University of Warsaw, Zwirki i Wigury 101, 02-089 Warsaw, Poland. .,Faculty of Chemistry, University of Warsaw, Pasteura 1, 02-093 Warsaw, Poland.
| | - Sławomir Filipek
- Biological and Chemical Research Centre, University of Warsaw, Zwirki i Wigury 101, 02-089 Warsaw, Poland. .,Faculty of Chemistry, University of Warsaw, Pasteura 1, 02-093 Warsaw, Poland.
| |
Collapse
|
17
|
Nedelyaeva OI, Popova LG, Volkov VS, Balnokin YV. Molecular Cloning and Characterization of SaCLCd, SaCLCf, and SaCLCg, Novel Proteins of the Chloride Channel Family (CLC) from the Halophyte Suaeda altissima (L.) Pall. PLANTS (BASEL, SWITZERLAND) 2022; 11:409. [PMID: 35161390 PMCID: PMC8839641 DOI: 10.3390/plants11030409] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 12/30/2021] [Revised: 01/28/2022] [Accepted: 01/29/2022] [Indexed: 06/14/2023]
Abstract
Coding sequences of the CLC family genes SaCLCd, SaCLCf, and SaCLCg, the putative orthologs of Arabidopsis thaliana AtCLCd, AtCLCf, and AtCLCg genes, were cloned from the euhalophyte Suaeda altissima (L.) Pall. The key conserved motifs and glutamates inherent in proteins of the CLC family were identified in SaCLCd, SaCLCf, and SaCLCg amino acid sequences. SaCLCd and SaCLCg were characterized by higher homology to eukaryotic (human) CLCs, while SaCLCf was closer to prokaryotic CLCs. Ion specificities of the SaCLC proteins were studied in complementation assays by heterologous expression of the SaCLC genes in the Saccharomyces cerevisiae GEF1 disrupted strain Δgef1. GEF1 encoded the only CLC family protein, the Cl- transporter Gef1p, in undisrupted strains of this organism. Expression of SaCLCd in Δgef1 cells restored their ability to grow on selective media. The complementation test and the presence of both the "gating" and "proton" conservative glutamates in SaCLCd amino acid sequence and serine specific for Cl- in its selectivity filter suggest that this protein operates as a Cl-/H+ antiporter. By contrast, expression of SaCLCf and SaCLCg did not complement the growth defect phenotype of Δgef1 cells. The selectivity filters of SaCLCf and SaCLCg also contained serine. However, SaCLCf included only the "gating" glutamate, while SaCLCg contained the "proton" glutamate, suggesting that SaCLCf and SaCLCg proteins act as Cl- channels. The SaCLCd, SaCLCf, and SaCLCg genes were shown to be expressed in the roots and leaves of S. altissima. In response to addition of NaCl to the growth medium, the relative transcript abundances of all three genes of S. altissima increased in the leaves but did not change significantly in the roots. The increase in expression of SaCLCd, SaCLCf, and SaCLCg in the leaves in response to increasing salinity was in line with Cl- accumulation in the leaf cells, indicating the possible participation of SaCLCd, SaCLCf, and SaCLCg proteins in Cl- sequestration in cell organelles. Generally, these results suggest the involvement of SaCLC proteins in the response of S. altissima plants to increasing salinity and possible participation in mechanisms underlying salt tolerance.
Collapse
|
18
|
Xu X, Lu F, Zhang L, Li H, Du S, Tang J. Novel CLCN4 variant associated with syndromic X-linked intellectual disability in a Chinese girl: a case report. BMC Pediatr 2021; 21:384. [PMID: 34479510 PMCID: PMC8414764 DOI: 10.1186/s12887-021-02860-4] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/10/2021] [Accepted: 08/26/2021] [Indexed: 12/17/2022] Open
Abstract
Background The Raynaud-Claes type of X-linked syndromic mental retardation (MRXSRC) is a very rare condition, by intellectual disability ranged from borderline to profound, impaired language development, brain abnormalities, facial dysmorphisms and seizures. MRXSRC is caused by variants in CLCN4 which encodes the 2Cl−/H+ exchanger ClC-4 prominently expressed in brain. Case presentation We present a 3-year-old Chinese girl with intellectual disability, dysmorphic features, brain abnormalities, significant language impairment and autistic features. Brain magnetic resonance imaging (MRI) showed a thin corpus callosum, a mega cisterna magna and ventriculomegaly. Whole exome sequencing (WES) was performed to detect the molecular basis of the disease. It was confirmed that this girl carried a novel heterozygous missense variant (c.1343C > T, p.Ala448Val) of CLCN4 gene, inherited from her mother. This variant has not been registered in public databases and was predicted to be pathogenic by multiple in silico prediction tools. Conclusion Our investigation expands the phenotype spectrum for CLCN4 variants with syndromic X-linked intellectual disability, which help to improve the understanding of CLCN4-related intellectual disability and will help in genetic counselling for this family. Supplementary Information The online version contains supplementary material available at 10.1186/s12887-021-02860-4.
Collapse
Affiliation(s)
- Xin Xu
- Department of Rehabilitation, Children's Hospital of Nanjing Medical University, No. 72 Guangzhou Road, Nanjing, 210008, Jiangsu Province, China
| | - Fen Lu
- Department of Rehabilitation, Children's Hospital of Nanjing Medical University, No. 72 Guangzhou Road, Nanjing, 210008, Jiangsu Province, China
| | - Li Zhang
- Department of Rehabilitation, Children's Hospital of Nanjing Medical University, No. 72 Guangzhou Road, Nanjing, 210008, Jiangsu Province, China.
| | - Hongying Li
- Department of Rehabilitation, Children's Hospital of Nanjing Medical University, No. 72 Guangzhou Road, Nanjing, 210008, Jiangsu Province, China
| | - Senjie Du
- Department of Rehabilitation, Children's Hospital of Nanjing Medical University, No. 72 Guangzhou Road, Nanjing, 210008, Jiangsu Province, China
| | | |
Collapse
|
19
|
Ion Channels, Transporters, and Sensors Interact with the Acidic Tumor Microenvironment to Modify Cancer Progression. Rev Physiol Biochem Pharmacol 2021; 182:39-84. [PMID: 34291319 DOI: 10.1007/112_2021_63] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/19/2022]
Abstract
Solid tumors, including breast carcinomas, are heterogeneous but typically characterized by elevated cellular turnover and metabolism, diffusion limitations based on the complex tumor architecture, and abnormal intra- and extracellular ion compositions particularly as regards acid-base equivalents. Carcinogenesis-related alterations in expression and function of ion channels and transporters, cellular energy levels, and organellar H+ sequestration further modify the acid-base composition within tumors and influence cancer cell functions, including cell proliferation, migration, and survival. Cancer cells defend their cytosolic pH and HCO3- concentrations better than normal cells when challenged with the marked deviations in extracellular H+, HCO3-, and lactate concentrations typical of the tumor microenvironment. Ionic gradients determine the driving forces for ion transporters and channels and influence the membrane potential. Cancer and stromal cells also sense abnormal ion concentrations via intra- and extracellular receptors that modify cancer progression and prognosis. With emphasis on breast cancer, the current review first addresses the altered ion composition and the changes in expression and functional activity of ion channels and transporters in solid cancer tissue. It then discusses how ion channels, transporters, and cellular sensors under influence of the acidic tumor microenvironment shape cancer development and progression and affect the potential of cancer therapies.
Collapse
|
20
|
Subba A, Tomar S, Pareek A, Singla-Pareek SL. The chloride channels: Silently serving the plants. PHYSIOLOGIA PLANTARUM 2021; 171:688-702. [PMID: 33034380 DOI: 10.1111/ppl.13240] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/07/2020] [Revised: 10/02/2020] [Accepted: 10/05/2020] [Indexed: 05/12/2023]
Abstract
Chloride channels (CLCs), member of anion transporting proteins, are present ubiquitously in all life forms. Diverging from its name, the CLC family includes both channel and exchanger (proton-coupled) proteins; nevertheless, they share conserved structural organization. They are engaged in diverse indispensable functions such as acid and fluoride tolerance in prokaryotes to muscle stabilization, transepithelial transport, and neuronal development in mammals. Mutations in genes encoding CLCs lead to several physiological disorders in different organisms, including severe diseases in humans. Even in plants, loss of CLC protein function severely impairs various cellular processes critical for normal growth and development. These proteins sequester Cl- into the vacuole, thus, making them an attractive target for improving salinity tolerance in plants caused by high abundance of salts, primarily NaCl. Besides, some CLCs are involved in NO3 - transport and storage function in plants, thus, influencing their nitrogen use efficiency. However, despite their high significance, not many studies have been carried out in plants. Here, we have attempted to concisely highlight the basic structure of CLC proteins and critical residues essential for their function and classification. We also present the diverse functions of CLCs in plants from their first cloning back in 1996 to the knowledge acquired as of now. We stress the need for carrying out more in-depth studies on CLCs in plants, for they may have future applications towards crop improvement.
Collapse
Affiliation(s)
- Ashish Subba
- Plant Stress Biology, International Centre for Genetic Engineering and Biotechnology, New Delhi, India
| | - Surabhi Tomar
- Plant Stress Biology, International Centre for Genetic Engineering and Biotechnology, New Delhi, India
| | - Ashwani Pareek
- Stress Physiology and Molecular Biology Laboratory, School of Life Sciences, Jawaharlal Nehru University, New Delhi, India
| | - Sneh L Singla-Pareek
- Plant Stress Biology, International Centre for Genetic Engineering and Biotechnology, New Delhi, India
| |
Collapse
|
21
|
Nowicka-Bauer K, Szymczak-Cendlak M. Structure and Function of Ion Channels Regulating Sperm Motility-An Overview. Int J Mol Sci 2021; 22:ijms22063259. [PMID: 33806823 PMCID: PMC8004680 DOI: 10.3390/ijms22063259] [Citation(s) in RCA: 40] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2021] [Revised: 03/18/2021] [Accepted: 03/19/2021] [Indexed: 12/18/2022] Open
Abstract
Sperm motility is linked to the activation of signaling pathways that trigger movement. These pathways are mainly dependent on Ca2+, which acts as a secondary messenger. The maintenance of adequate Ca2+ concentrations is possible thanks to proper concentrations of other ions, such as K+ and Na+, among others, that modulate plasma membrane potential and the intracellular pH. Like in every cell, ion homeostasis in spermatozoa is ensured by a vast spectrum of ion channels supported by the work of ion pumps and transporters. To achieve success in fertilization, sperm ion channels have to be sensitive to various external and internal factors. This sensitivity is provided by specific channel structures. In addition, novel sperm-specific channels or isoforms have been found with compositions that increase the chance of fertilization. Notably, the most significant sperm ion channel is the cation channel of sperm (CatSper), which is a sperm-specific Ca2+ channel required for the hyperactivation of sperm motility. The role of other ion channels in the spermatozoa, such as voltage-gated Ca2+ channels (VGCCs), Ca2+-activated Cl-channels (CaCCs), SLO K+ channels or voltage-gated H+ channels (VGHCs), is to ensure the activation and modulation of CatSper. As the activation of sperm motility differs among metazoa, different ion channels may participate; however, knowledge regarding these channels is still scarce. In the present review, the roles and structures of the most important known ion channels are described in regard to regulation of sperm motility in animals.
Collapse
Affiliation(s)
- Karolina Nowicka-Bauer
- Department of Chemical Physics, Faculty of Chemistry, Adam Mickiewicz University in Poznań, 61-614 Poznan, Poland
- Correspondence:
| | - Monika Szymczak-Cendlak
- Department of Animal Physiology and Development, Faculty of Biology, Adam Mickiewicz University in Poznań, 61-614 Poznan, Poland;
| |
Collapse
|
22
|
Mamaeva D, Jazouli Z, DiFrancesco ML, Erkilic N, Dubois G, Hilaire C, Meunier I, Boukhaddaoui H, Kalatzis V. Novel roles for voltage-gated T-type Ca 2+ and ClC-2 channels in phagocytosis and angiogenic factor balance identified in human iPSC-derived RPE. FASEB J 2021; 35:e21406. [PMID: 33724552 DOI: 10.1096/fj.202002754r] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2020] [Revised: 01/13/2021] [Accepted: 01/19/2021] [Indexed: 01/26/2023]
Abstract
Human-induced pluripotent stem cell (hiPSC)-derived retinal pigment epithelium (RPE) is a powerful tool for pathophysiological studies and preclinical therapeutic screening, as well as a source for clinical cell transplantation. Thus, it must be validated for maturity and functionality to ensure correct data readouts and clinical safety. Previous studies have validated hiPSC-derived RPE as morphologically characteristic of the tissue in the human eye. However, information concerning the expression and functionality of ion channels is still limited. We screened hiPSC-derived RPE for the polarized expression of a panel of L-type (CaV 1.1, CaV 1.3) and T-type (CaV 3.1, CaV 3.3) Ca2+ channels, K+ channels (Maxi-K, Kir4.1, Kir7.1), and the Cl- channel ClC-2 known to be expressed in native RPE. We also tested the roles of these channels in key RPE functions using specific inhibitors. In addition to confirming the native expression profiles and function of certain channels, such as L-type Ca2+ channels, we show for the first time that T-type Ca2+ channels play a role in both phagocytosis and vascular endothelial growth factor (VEGF) secretion. Moreover, we demonstrate that Maxi-K and Kir7.1 channels are involved in the polarized secretion of VEGF and pigment epithelium-derived factor (PEDF). Furthermore, we show a novel localization for ClC-2 channel on the apical side of hiPSC-derived RPE, with an overexpression at the level of fluid-filled domes, and demonstrate that it plays an important role in phagocytosis, as well as VEGF and PEDF secretion. Taken together, hiPSC-derived RPE is a powerful model for advancing fundamental knowledge of RPE functions.
Collapse
Affiliation(s)
- Daria Mamaeva
- Institute for Neurosciences of Montpellier, Inserm, Montpellier University, Montpellier, France
| | - Zhour Jazouli
- Institute for Neurosciences of Montpellier, Inserm, Montpellier University, Montpellier, France
| | - Mattia L DiFrancesco
- Institute for Neurosciences of Montpellier, Inserm, Montpellier University, Montpellier, France
| | - Nejla Erkilic
- Institute for Neurosciences of Montpellier, Inserm, Montpellier University, Montpellier, France.,National Reference Centre for Inherited Sensory Diseases, Montpellier University, CHU, Montpellier, France
| | - Gregor Dubois
- Institute for Neurosciences of Montpellier, Inserm, Montpellier University, Montpellier, France
| | - Cecile Hilaire
- Institute for Neurosciences of Montpellier, Inserm, Montpellier University, Montpellier, France
| | - Isabelle Meunier
- Institute for Neurosciences of Montpellier, Inserm, Montpellier University, Montpellier, France.,National Reference Centre for Inherited Sensory Diseases, Montpellier University, CHU, Montpellier, France
| | - Hassan Boukhaddaoui
- Institute for Neurosciences of Montpellier, Inserm, Montpellier University, Montpellier, France
| | - Vasiliki Kalatzis
- Institute for Neurosciences of Montpellier, Inserm, Montpellier University, Montpellier, France
| |
Collapse
|
23
|
|
24
|
Kwon HC, Yu Y, Fairclough RH, Chen TY. Proton-dependent inhibition, inverted voltage activation, and slow gating of CLC-0 Chloride Channel. PLoS One 2020; 15:e0240704. [PMID: 33362212 PMCID: PMC7757909 DOI: 10.1371/journal.pone.0240704] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2020] [Accepted: 12/03/2020] [Indexed: 11/21/2022] Open
Abstract
CLC-0, a prototype Cl- channel in the CLC family, employs two gating mechanisms that control its ion-permeation pore: fast gating and slow gating. The negatively-charged sidechain of a pore glutamate residue, E166, is known to be the fast gate, and the swinging of this sidechain opens or closes the pore of CLC-0 on the millisecond time scale. The other gating mechanism, slow gating, operates with much slower kinetics in the range of seconds to tens or even hundreds of seconds, and it is thought to involve still-unknown conformational rearrangements. Here, we find that low intracellular pH (pHi) facilitates the closure of the CLC-0’s slow gate, thus generating current inhibition. The rate of low pHi-induced current inhibition increases with intracellular H+ concentration ([H+]i)—the time constants of current inhibition by low pHi = 4.5, 5.5 and 6 are roughly 0.1, 1 and 10 sec, respectively, at room temperature. In comparison, the time constant of the slow gate closure at pHi = 7.4 at room temperature is hundreds of seconds. The inhibition by low pHi is significantly less prominent in mutants favoring the slow-gate open state (such as C212S and Y512A), further supporting the fact that intracellular H+ enhances the slow-gate closure in CLC-0. A fast inhibition by low pHi causes an apparent inverted voltage-dependent activation in the wild-type CLC-0, a behavior similar to those in some channel mutants such as V490W in which only membrane hyperpolarization can open the channel. Interestingly, when V490W mutation is constructed in the background of C212S or Y512A mutation, the inverted voltage-dependent activation disappears. We propose that the slow kinetics of CLC-0’s slow-gate closure may be due to low [H+]i rather than due to the proposed large conformational change of the channel protein. Our results also suggest that the inverted voltage-dependent opening observed in some mutant channels may result from fast closure of the slow gate by the mutations.
Collapse
Affiliation(s)
- Hwoi Chan Kwon
- Biophysics Graduate Program, University of California, Davis, California, United States of America
| | - Yawei Yu
- BMCDB Graduate Program, University of California, Davis, California, United States of America
| | - Robert H. Fairclough
- Biophysics Graduate Program, University of California, Davis, California, United States of America
- BMCDB Graduate Program, University of California, Davis, California, United States of America
- Center for Neuroscience, University of California, Davis, California, United States of America
| | - Tsung-Yu Chen
- Biophysics Graduate Program, University of California, Davis, California, United States of America
- BMCDB Graduate Program, University of California, Davis, California, United States of America
- Department of Neurology, University of California, Davis, California, United States of America
- Center for Neuroscience, University of California, Davis, California, United States of America
- * E-mail:
| |
Collapse
|
25
|
Polovitskaya MM, Barbini C, Martinelli D, Harms FL, Cole FS, Calligari P, Bocchinfuso G, Stella L, Ciolfi A, Niceta M, Rizza T, Shinawi M, Sisco K, Johannsen J, Denecke J, Carrozzo R, Wegner DJ, Kutsche K, Tartaglia M, Jentsch TJ. A Recurrent Gain-of-Function Mutation in CLCN6, Encoding the ClC-6 Cl -/H +-Exchanger, Causes Early-Onset Neurodegeneration. Am J Hum Genet 2020; 107:1062-1077. [PMID: 33217309 PMCID: PMC7820737 DOI: 10.1016/j.ajhg.2020.11.004] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2020] [Accepted: 11/02/2020] [Indexed: 12/16/2022] Open
Abstract
Dysfunction of the endolysosomal system is often associated with neurodegenerative disease because postmitotic neurons are particularly reliant on the elimination of intracellular aggregates. Adequate function of endosomes and lysosomes requires finely tuned luminal ion homeostasis and transmembrane ion fluxes. Endolysosomal CLC Cl-/H+ exchangers function as electric shunts for proton pumping and in luminal Cl- accumulation. We now report three unrelated children with severe neurodegenerative disease, who carry the same de novo c.1658A>G (p.Tyr553Cys) mutation in CLCN6, encoding the late endosomal Cl-/H+-exchanger ClC-6. Whereas Clcn6-/- mice have only mild neuronal lysosomal storage abnormalities, the affected individuals displayed severe developmental delay with pronounced generalized hypotonia, respiratory insufficiency, and variable neurodegeneration and diffusion restriction in cerebral peduncles, midbrain, and/or brainstem in MRI scans. The p.Tyr553Cys amino acid substitution strongly slowed ClC-6 gating and increased current amplitudes, particularly at the acidic pH of late endosomes. Transfection of ClC-6Tyr553Cys, but not ClC-6WT, generated giant LAMP1-positive vacuoles that were poorly acidified. Their generation strictly required ClC-6 ion transport, as shown by transport-deficient double mutants, and depended on Cl-/H+ exchange, as revealed by combination with the uncoupling p.Glu200Ala substitution. Transfection of either ClC-6Tyr553Cys/Glu200Ala or ClC-6Glu200Ala generated slightly enlarged vesicles, suggesting that p.Glu200Ala, previously associated with infantile spasms and microcephaly, is also pathogenic. Bafilomycin treatment abrogated vacuole generation, indicating that H+-driven Cl- accumulation osmotically drives vesicle enlargement. Our work establishes mutations in CLCN6 associated with neurological diseases, whose spectrum of clinical features depends on the differential impact of the allele on ClC-6 function.
Collapse
Affiliation(s)
- Maya M Polovitskaya
- Leibniz-Forschungsinstitut für Molekulare Pharmakologie (FMP), 13125 Berlin, Germany; Max-Delbrück-Centrum für Molekulare Medizin (MDC), 13125 Berlin, Germany
| | - Carlo Barbini
- Leibniz-Forschungsinstitut für Molekulare Pharmakologie (FMP), 13125 Berlin, Germany; Max-Delbrück-Centrum für Molekulare Medizin (MDC), 13125 Berlin, Germany
| | - Diego Martinelli
- Genetics and Rare Diseases Research Division, Ospedale Pediatrico Bambino Gesù, IRCCS, 00146 Rome, Italy
| | - Frederike L Harms
- Institute for Human Genetics, University Medical Center Hamburg-Eppendorf, 20246 Hamburg, Germany
| | - F Sessions Cole
- Division of Newborn Medicine, Edward Mallinckrodt Department of Pediatrics, Washington University School of Medicine, and St. Louis Children's Hospital, St. Louis, MO 63110, USA
| | - Paolo Calligari
- Department of Chemical Science and Technologies, University "Tor Vergata," 00133 Rome, Italy
| | - Gianfranco Bocchinfuso
- Department of Chemical Science and Technologies, University "Tor Vergata," 00133 Rome, Italy
| | - Lorenzo Stella
- Department of Chemical Science and Technologies, University "Tor Vergata," 00133 Rome, Italy
| | - Andrea Ciolfi
- Genetics and Rare Diseases Research Division, Ospedale Pediatrico Bambino Gesù, IRCCS, 00146 Rome, Italy
| | - Marcello Niceta
- Genetics and Rare Diseases Research Division, Ospedale Pediatrico Bambino Gesù, IRCCS, 00146 Rome, Italy
| | - Teresa Rizza
- Genetics and Rare Diseases Research Division, Ospedale Pediatrico Bambino Gesù, IRCCS, 00146 Rome, Italy
| | - Marwan Shinawi
- Division of Genetics and Genomic Medicine, Edward Mallinckrodt Department of Pediatrics, Washington University School of Medicine, and St. Louis Children's Hospital, St. Louis, MO 63110, USA
| | - Kathleen Sisco
- Division of Genetics and Genomic Medicine, Edward Mallinckrodt Department of Pediatrics, Washington University School of Medicine, and St. Louis Children's Hospital, St. Louis, MO 63110, USA
| | - Jessika Johannsen
- Children's Hospital, University Medical Center Hamburg-Eppendorf, 20246 Hamburg, Germany
| | - Jonas Denecke
- Children's Hospital, University Medical Center Hamburg-Eppendorf, 20246 Hamburg, Germany
| | - Rosalba Carrozzo
- Genetics and Rare Diseases Research Division, Ospedale Pediatrico Bambino Gesù, IRCCS, 00146 Rome, Italy
| | - Daniel J Wegner
- Division of Newborn Medicine, Edward Mallinckrodt Department of Pediatrics, Washington University School of Medicine, and St. Louis Children's Hospital, St. Louis, MO 63110, USA
| | - Kerstin Kutsche
- Institute for Human Genetics, University Medical Center Hamburg-Eppendorf, 20246 Hamburg, Germany
| | - Marco Tartaglia
- Genetics and Rare Diseases Research Division, Ospedale Pediatrico Bambino Gesù, IRCCS, 00146 Rome, Italy.
| | - Thomas J Jentsch
- Leibniz-Forschungsinstitut für Molekulare Pharmakologie (FMP), 13125 Berlin, Germany; Max-Delbrück-Centrum für Molekulare Medizin (MDC), 13125 Berlin, Germany; NeuroCure Cluster of Excellence, Charité Universitätsmedizin, 10117 Berlin, Germany.
| |
Collapse
|
26
|
Tchernookova BK, Gongwer MW, George A, Goeglein B, Powell AM, Caringal HL, Leuschner T, Phillips AG, Schantz AW, Kiedrowski L, Chappell R, Kreitzer MA, Malchow RP. ATP-mediated increase in H + flux from retinal Müller cells: a role for Na +/H + exchange. J Neurophysiol 2020; 125:184-198. [PMID: 33206577 DOI: 10.1152/jn.00546.2020] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Small alterations in extracellular H+ can profoundly alter neurotransmitter release by neurons. We examined mechanisms by which extracellular ATP induces an extracellular H+ flux from Müller glial cells, which surround synaptic connections throughout the vertebrate retina. Müller glia were isolated from tiger salamander retinae and H+ fluxes examined using self-referencing H+-selective microelectrodes. Experiments were performed in 1 mM HEPES with no bicarbonate present. Replacement of extracellular sodium by choline decreased H+ efflux induced by 10 µM ATP by 75%. ATP-induced H+ efflux was also reduced by Na+/H+ exchange inhibitors. Amiloride reduced H+ efflux initiated by 10 µM ATP by 60%, while 10 µM cariporide decreased H+ flux by 37%, and 25 µM zoniporide reduced H+ flux by 32%. ATP-induced H+ fluxes were not significantly altered by the K+/H+ pump blockers SCH28080 or TAK438, and replacement of all extracellular chloride with gluconate was without effect on H+ fluxes. Recordings of ATP-induced H+ efflux from cells that were simultaneously whole cell voltage clamped revealed no effect of membrane potential from -70 mV to 0 mV. Restoration of extracellular potassium after cells were bathed in 0 mM potassium produced a transient alteration in ATP-dependent H+ efflux. The transient response to extracellular potassium occurred only when extracellular sodium was present and was abolished by 1 mM ouabain, suggesting that alterations in sodium gradients were mediated by Na+/K+-ATPase activity. Our data indicate that the majority of H+ efflux elicited by extracellular ATP from isolated Müller cells is mediated by Na+/H+ exchange.NEW & NOTEWORTHY Glial cells are known to regulate neuronal activity, but the exact mechanism(s) whereby these "support" cells modulate synaptic transmission remains unclear. Small changes in extracellular levels of acidity are known to be particularly powerful regulators of neurotransmitter release. Here, we show that extracellular ATP, known to be a potent activator of glial cells, induces H+ efflux from retinal Müller (glial) cells and that the bulk of the H+ efflux is mediated by Na+/H+ exchange.
Collapse
Affiliation(s)
| | | | - Alexis George
- Department of Biology, Indiana Wesleyan University, Marion, Indiana
| | - Brock Goeglein
- Department of Biology, Indiana Wesleyan University, Marion, Indiana
| | - Alyssa M Powell
- Department of Biology, Indiana Wesleyan University, Marion, Indiana
| | | | - Thomas Leuschner
- Department of Biology, Indiana Wesleyan University, Marion, Indiana
| | - Anna G Phillips
- Department of Biology, Indiana Wesleyan University, Marion, Indiana
| | - Adam W Schantz
- Department of Biology, Indiana Wesleyan University, Marion, Indiana
| | - Lech Kiedrowski
- Department of Biological Sciences, University of Illinois at Chicago, Chicago, Illinois.,Spot Cells LLC, Chicago, Illinois
| | - Richard Chappell
- Department of Physiology and Biophysics, University at Buffalo, Buffalo, New York.,Eugene Bell Center, Marine Biological Laboratory, Woods Hole, Massachusetts
| | | | - Robert Paul Malchow
- Department of Biological Sciences, University of Illinois at Chicago, Chicago, Illinois.,Department of Ophthalmology and Visual Sciences, University of Illinois at Chicago, Chicago, Illinois
| |
Collapse
|
27
|
Lu C, Ma Z, Cheng X, Wu H, Tuo B, Liu X, Li T. Pathological role of ion channels and transporters in the development and progression of triple-negative breast cancer. Cancer Cell Int 2020; 20:377. [PMID: 32782435 PMCID: PMC7409684 DOI: 10.1186/s12935-020-01464-9] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2020] [Revised: 07/26/2020] [Accepted: 07/29/2020] [Indexed: 12/12/2022] Open
Abstract
Breast cancer is a common malignancy in women. Among breast cancer types, triple-negative breast cancer (TNBC) tends to affect younger women, is prone to axillary lymph node, lung, and bone metastases; and has a high recurrence rate. Due to a lack of classic biomarkers, the currently available treatments are surgery and chemotherapy; no targeted standard treatment options are available. Therefore, it is urgent to find a novel and effective therapeutic target. As alteration of ion channels and transporters in normal mammary cells may affect cell growth, resulting in the development and progression of TNBC, ion channels and transporters may be promising new therapeutic targets for TNBC. This review summarizes ion channels and transporters related to TNBC and may provide new tumor biomarkers and help in the development of novel targeted therapies.
Collapse
Affiliation(s)
- Chengli Lu
- Department of Thyroid and Breast Surgery, Affiliated Hospital of Zunyi Medical University, Zunyi, 563003 Guizhou Province China
| | - Zhiyuan Ma
- Department of Thyroid and Breast Surgery, Affiliated Hospital of Zunyi Medical University, Zunyi, 563003 Guizhou Province China
| | - Xiaoming Cheng
- Department of Thyroid and Breast Surgery, Affiliated Hospital of Zunyi Medical University, Zunyi, 563003 Guizhou Province China
| | - Huichao Wu
- Department of Gastroenterology, Affiliated Hospital of Zunyi Medical University, Zunyi, 563003, Guizhou Province China
| | - Biguang Tuo
- Department of Gastroenterology, Affiliated Hospital of Zunyi Medical University, Zunyi, 563003, Guizhou Province China.,Digestive Disease Institute of Guizhou Province, Zunyi, Guizhou Province China
| | - Xuemei Liu
- Department of Gastroenterology, Affiliated Hospital of Zunyi Medical University, Zunyi, 563003, Guizhou Province China.,Digestive Disease Institute of Guizhou Province, Zunyi, Guizhou Province China
| | - Taolang Li
- Department of Thyroid and Breast Surgery, Affiliated Hospital of Zunyi Medical University, Zunyi, 563003 Guizhou Province China
| |
Collapse
|
28
|
Zhang S, Liu Y, Zhang B, Zhou J, Li T, Liu Z, Li Y, Yang M. Molecular insights into the human CLC-7/Ostm1 transporter. SCIENCE ADVANCES 2020; 6:eabb4747. [PMID: 32851177 PMCID: PMC7423370 DOI: 10.1126/sciadv.abb4747] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/26/2020] [Accepted: 06/29/2020] [Indexed: 05/14/2023]
Abstract
CLC family proteins translocate chloride ions across cell membranes to maintain the membrane potential, regulate the transepithelial Cl- transport, and control the intravesicular pH among different organelles. CLC-7/Ostm1 is an electrogenic Cl-/H+ antiporter that mainly resides in lysosomes and osteoclast ruffled membranes. Mutations in human CLC-7/Ostm1 lead to lysosomal storage disorders and severe osteopetrosis. Here, we present the cryo-electron microscopy (cryo-EM) structure of the human CLC-7/Ostm1 complex and reveal that the highly glycosylated Ostm1 functions like a lid positioned above CLC-7 and interacts extensively with CLC-7 within the membrane. Our complex structure reveals a functionally crucial domain interface between the amino terminus, TMD, and CBS domains of CLC-7. Structural analyses and electrophysiology studies suggest that the domain interaction interfaces affect the slow gating kinetics of CLC-7/Ostm1. Thus, our study deepens understanding of CLC-7/Ostm1 transporter and provides insights into the molecular basis of the disease-related mutations.
Collapse
Affiliation(s)
- Sensen Zhang
- Ministry of Education Key Laboratory of Protein Science, Tsinghua-Peking Joint Center for Life Sciences, Beijing Advanced Innovation Center for Structural Biology, School of Life Sciences, Tsinghua University, Beijing 100084, China
| | - Yang Liu
- Ministry of Education Key Laboratory of Protein Science, Tsinghua-Peking Joint Center for Life Sciences, Beijing Advanced Innovation Center for Structural Biology, School of Life Sciences, Tsinghua University, Beijing 100084, China
| | - Bing Zhang
- Department of Anesthesiology, Shanghai First Maternity and Infant Hospital, Tongji University School of Medicine, Shanghai 201204, China
| | - Jun Zhou
- Ministry of Education Key Laboratory of Protein Science, Tsinghua-Peking Joint Center for Life Sciences, Beijing Advanced Innovation Center for Structural Biology, School of Life Sciences, Tsinghua University, Beijing 100084, China
| | - Tianyu Li
- CAS Key Laboratory of Receptor Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
| | - Zhiqiang Liu
- Department of Anesthesiology, Shanghai First Maternity and Infant Hospital, Tongji University School of Medicine, Shanghai 201204, China
- Corresponding author. (Z.L.); (Y.L.); (M.Y.)
| | - Yang Li
- CAS Key Laboratory of Receptor Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
- Corresponding author. (Z.L.); (Y.L.); (M.Y.)
| | - Maojun Yang
- Ministry of Education Key Laboratory of Protein Science, Tsinghua-Peking Joint Center for Life Sciences, Beijing Advanced Innovation Center for Structural Biology, School of Life Sciences, Tsinghua University, Beijing 100084, China
- School of Pharmacy, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
- Corresponding author. (Z.L.); (Y.L.); (M.Y.)
| |
Collapse
|
29
|
Chiariello MG, Bolnykh V, Ippoliti E, Meloni S, Olsen JMH, Beck T, Rothlisberger U, Fahlke C, Carloni P. Molecular Basis of CLC Antiporter Inhibition by Fluoride. J Am Chem Soc 2020; 142:7254-7258. [PMID: 32233472 DOI: 10.1021/jacs.9b13588] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
CLC channels and transporters conduct or transport various kinds of anions, with the exception of fluoride, which acts as an effective inhibitor. Here, we performed sub-nanosecond DFT-based QM/MM simulations of the E. coli anion/proton exchanger ClC-ec1 and observed that fluoride binds incoming protons within the selectivity filter, with excess protons shared with the gating glutamate E148. Depending on E148 conformation, the competition for the proton can involve either a direct F-/E148 interaction or the modulation of water molecules bridging the two anions. The direct interaction locks E148 in a conformation that does not allow for proton transport, and thus inhibits protein function.
Collapse
Affiliation(s)
- Maria Gabriella Chiariello
- Institute for Advanced Simulation (IAS-5) and Institute of Neuroscience and Medicine (INM-9), Computational Biomedicine, Forschungszentrum Jülich, 52425 Jülich, Germany.,JARA-HPC, Forschungszentrum Jülich, D-54245 Jülich, Germany
| | - Viacheslav Bolnykh
- Laboratory of Computational Chemistry and Biochemistry, Ecole Polytechnique Fédérale de Lausanne, CH-1015 Lausanne, Switzerland
| | - Emiliano Ippoliti
- Institute for Advanced Simulation (IAS-5) and Institute of Neuroscience and Medicine (INM-9), Computational Biomedicine, Forschungszentrum Jülich, 52425 Jülich, Germany.,JARA-HPC, Forschungszentrum Jülich, D-54245 Jülich, Germany
| | - Simone Meloni
- Dipartimento di Scienze Chimiche e Farmaceutiche, Università degli Studi di Ferrara, Via Luigi Borsari 46, I-44121 Ferrara, Italy
| | - Jógvan Magnus Haugaard Olsen
- Hylleraas Centre for Quantum Molecular Sciences, Department of Chemistry, UiT The Arctic University of Norway, N-9037 Tromsø, Norway
| | - Thomas Beck
- Department of Chemistry, University of Cincinnati, Cincinnati, Ohio 45221, United States
| | - Ursula Rothlisberger
- Laboratory of Computational Chemistry and Biochemistry, Ecole Polytechnique Fédérale de Lausanne, CH-1015 Lausanne, Switzerland
| | - Christoph Fahlke
- Institute of Biological Information Processing (IBI-1), Molekular- und Zellphysiologie, Forschungszentrum Jülich, D-54245 Jülich, Germany
| | - Paolo Carloni
- Institute for Advanced Simulation (IAS-5) and Institute of Neuroscience and Medicine (INM-9), Computational Biomedicine, Forschungszentrum Jülich, 52425 Jülich, Germany.,JARA-HPC, Forschungszentrum Jülich, D-54245 Jülich, Germany.,Department of Physics, RWTH Aachen University, 52056 Aachen, Germany.,Institute of Neuroscience and Medicine (INM-11), Molecular Neuroscience and Neuroimaging, Forschungszentrum Julich, 52425 Julich, Germany
| |
Collapse
|
30
|
McKiernan KA, Koster AK, Maduke M, Pande VS. Dynamical model of the CLC-2 ion channel reveals conformational changes associated with selectivity-filter gating. PLoS Comput Biol 2020; 16:e1007530. [PMID: 32226009 PMCID: PMC7145265 DOI: 10.1371/journal.pcbi.1007530] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2018] [Revised: 04/09/2020] [Accepted: 11/05/2019] [Indexed: 12/18/2022] Open
Abstract
This work reports a dynamical Markov state model of CLC-2 "fast" (pore) gating, based on 600 microseconds of molecular dynamics (MD) simulation. In the starting conformation of our CLC-2 model, both outer and inner channel gates are closed. The first conformational change in our dataset involves rotation of the inner-gate backbone along residues S168-G169-I170. This change is strikingly similar to that observed in the cryo-EM structure of the bovine CLC-K channel, though the volume of the intracellular (inner) region of the ion conduction pathway is further expanded in our model. From this state (inner gate open and outer gate closed), two additional states are observed, each involving a unique rotameric flip of the outer-gate residue GLUex. Both additional states involve conformational changes that orient GLUex away from the extracellular (outer) region of the ion conduction pathway. In the first additional state, the rotameric flip of GLUex results in an open, or near-open, channel pore. The equilibrium population of this state is low (∼1%), consistent with the low open probability of CLC-2 observed experimentally in the absence of a membrane potential stimulus (0 mV). In the second additional state, GLUex rotates to occlude the channel pore. This state, which has a low equilibrium population (∼1%), is only accessible when GLUex is protonated. Together, these pathways model the opening of both an inner and outer gate within the CLC-2 selectivity filter, as a function of GLUex protonation. Collectively, our findings are consistent with published experimental analyses of CLC-2 gating and provide a high-resolution structural model to guide future investigations.
Collapse
Affiliation(s)
- Keri A. McKiernan
- Department of Chemistry, Stanford University, Stanford, California, United States of America
| | - Anna K. Koster
- Department of Chemistry, Stanford University, Stanford, California, United States of America
- Department of Molecular & Cellular Physiology, Stanford University, Stanford, California, United States of America
| | - Merritt Maduke
- Department of Molecular & Cellular Physiology, Stanford University, Stanford, California, United States of America
| | - Vijay S. Pande
- Department of Bioengineering, Stanford University, Stanford, California, United States of America
| |
Collapse
|
31
|
Henríquez-Olguín C, Boronat S, Cabello-Verrugio C, Jaimovich E, Hidalgo E, Jensen TE. The Emerging Roles of Nicotinamide Adenine Dinucleotide Phosphate Oxidase 2 in Skeletal Muscle Redox Signaling and Metabolism. Antioxid Redox Signal 2019; 31:1371-1410. [PMID: 31588777 PMCID: PMC6859696 DOI: 10.1089/ars.2018.7678] [Citation(s) in RCA: 33] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Significance: Skeletal muscle is a crucial tissue to whole-body locomotion and metabolic health. Reactive oxygen species (ROS) have emerged as intracellular messengers participating in both physiological and pathological adaptations in skeletal muscle. A complex interplay between ROS-producing enzymes and antioxidant networks exists in different subcellular compartments of mature skeletal muscle. Recent evidence suggests that nicotinamide adenine dinucleotide phosphate (NADPH) oxidases (NOXs) are a major source of contraction- and insulin-stimulated oxidants production, but they may paradoxically also contribute to muscle insulin resistance and atrophy. Recent Advances: Pharmacological and molecular biological tools, including redox-sensitive probes and transgenic mouse models, have generated novel insights into compartmentalized redox signaling and suggested that NOX2 contributes to redox control of skeletal muscle metabolism. Critical Issues: Major outstanding questions in skeletal muscle include where NOX2 activation occurs under different conditions in health and disease, how NOX2 activation is regulated, how superoxide/hydrogen peroxide generated by NOX2 reaches the cytosol, what the signaling mediators are downstream of NOX2, and the role of NOX2 for different physiological and pathophysiological processes. Future Directions: Future research should utilize and expand the current redox-signaling toolbox to clarify the NOX2-dependent mechanisms in skeletal muscle and determine whether the proposed functions of NOX2 in cells and animal models are conserved into humans.
Collapse
Affiliation(s)
- Carlos Henríquez-Olguín
- Section of Molecular Physiology, Department of Nutrition, Exercise and Sports (NEXS), Faculty of Science, University of Copenhagen, Copenhagen, Denmark.,Muscle Cell Physiology Laboratory, Center for Exercise, Metabolism, and Cancer, Instituto de Ciencias Biomédicas, Universidad de Chile, Santiago, Chile
| | - Susanna Boronat
- Oxidative Stress and Cell Cycle Group, Universitat Pompeu Fabra, Barcelona, Spain
| | - Claudio Cabello-Verrugio
- Laboratory of Muscle Pathology, Fragility and Aging, Department of Biological Sciences, Faculty of Life Sciences, Universidad Andres Bello, Santiago, Chile.,Millennium Institute on Immunology and Immunotherapy, Santiago, Chile.,Center for the Development of Nanoscience and Nanotechnology (CEDENNA), Universidad de Santiago de Chile, Santiago, Chile
| | - Enrique Jaimovich
- Muscle Cell Physiology Laboratory, Center for Exercise, Metabolism, and Cancer, Instituto de Ciencias Biomédicas, Universidad de Chile, Santiago, Chile
| | - Elena Hidalgo
- Oxidative Stress and Cell Cycle Group, Universitat Pompeu Fabra, Barcelona, Spain
| | - Thomas E Jensen
- Section of Molecular Physiology, Department of Nutrition, Exercise and Sports (NEXS), Faculty of Science, University of Copenhagen, Copenhagen, Denmark
| |
Collapse
|
32
|
Göppner C, Orozco IJ, Hoegg-Beiler MB, Soria AH, Hübner CA, Fernandes-Rosa FL, Boulkroun S, Zennaro MC, Jentsch TJ. Pathogenesis of hypertension in a mouse model for human CLCN2 related hyperaldosteronism. Nat Commun 2019; 10:4678. [PMID: 31615979 PMCID: PMC6794291 DOI: 10.1038/s41467-019-12113-9] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2019] [Accepted: 08/21/2019] [Indexed: 12/31/2022] Open
Abstract
Human primary aldosteronism (PA) can be caused by mutations in several ion channel genes but mouse models replicating this condition are lacking. We now show that almost all known PA-associated CLCN2 mutations markedly increase ClC-2 chloride currents and generate knock-in mice expressing a constitutively open ClC-2 Cl− channel as mouse model for PA. The Clcn2op allele strongly increases the chloride conductance of zona glomerulosa cells, provoking a strong depolarization and increasing cytoplasmic Ca2+ concentration. Clcn2op mice display typical features of human PA, including high serum aldosterone in the presence of low renin activity, marked hypertension and hypokalemia. These symptoms are more pronounced in homozygous Clcn2op/op than in heterozygous Clcn2+/op mice. This difference is attributed to the unexpected finding that only ~50 % of Clcn2+/op zona glomerulosa cells are depolarized. By reproducing essential features of human PA, Clcn2op mice are a valuable model to study the pathological mechanisms underlying this disease. Mutations in the chloride channel ClC-2 have been found in primary aldosteronism (PA). Here, Göppner et al. generate transgenic mice expressing a mutant form of ClC-2 that displays increased chloride currents like patient mutations, and find it recapitulates the key pathological features of PA.
Collapse
Affiliation(s)
- Corinna Göppner
- Leibniz-Forschungsinstitut für Molekulare Pharmakologie (FMP), Berlin, Germany.,Max-Delbrück-Centrum für Molekulare Medizin (MDC), Berlin, Germany
| | - Ian J Orozco
- Leibniz-Forschungsinstitut für Molekulare Pharmakologie (FMP), Berlin, Germany.,Max-Delbrück-Centrum für Molekulare Medizin (MDC), Berlin, Germany
| | - Maja B Hoegg-Beiler
- Leibniz-Forschungsinstitut für Molekulare Pharmakologie (FMP), Berlin, Germany.,Max-Delbrück-Centrum für Molekulare Medizin (MDC), Berlin, Germany
| | - Audrey H Soria
- Leibniz-Forschungsinstitut für Molekulare Pharmakologie (FMP), Berlin, Germany.,Max-Delbrück-Centrum für Molekulare Medizin (MDC), Berlin, Germany
| | | | - Fabio L Fernandes-Rosa
- INSERM, UMRS_970, Paris Cardiovascular Research Center, Paris, France.,Université Paris Descartes, Sorbonne Paris Cité, Paris, France
| | - Sheerazed Boulkroun
- INSERM, UMRS_970, Paris Cardiovascular Research Center, Paris, France.,Université Paris Descartes, Sorbonne Paris Cité, Paris, France
| | - Maria-Christina Zennaro
- INSERM, UMRS_970, Paris Cardiovascular Research Center, Paris, France.,Université Paris Descartes, Sorbonne Paris Cité, Paris, France.,Assistance Publique-Hôpitaux de Paris, Hôpital Européen Georges Pompidou, Service de Génétique, Paris, France
| | - Thomas J Jentsch
- Leibniz-Forschungsinstitut für Molekulare Pharmakologie (FMP), Berlin, Germany. .,Max-Delbrück-Centrum für Molekulare Medizin (MDC), Berlin, Germany. .,NeuroCure Cluster of Excellence, Charité Universitätsmedizin Berlin, Berlin, Germany.
| |
Collapse
|
33
|
van der Wijst J, Belge H, Bindels RJM, Devuyst O. Learning Physiology From Inherited Kidney Disorders. Physiol Rev 2019; 99:1575-1653. [PMID: 31215303 DOI: 10.1152/physrev.00008.2018] [Citation(s) in RCA: 56] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
The identification of genes causing inherited kidney diseases yielded crucial insights in the molecular basis of disease and improved our understanding of physiological processes that operate in the kidney. Monogenic kidney disorders are caused by mutations in genes coding for a large variety of proteins including receptors, channels and transporters, enzymes, transcription factors, and structural components, operating in specialized cell types that perform highly regulated homeostatic functions. Common variants in some of these genes are also associated with complex traits, as evidenced by genome-wide association studies in the general population. In this review, we discuss how the molecular genetics of inherited disorders affecting different tubular segments of the nephron improved our understanding of various transport processes and of their involvement in homeostasis, while providing novel therapeutic targets. These include inherited disorders causing a dysfunction of the proximal tubule (renal Fanconi syndrome), with emphasis on epithelial differentiation and receptor-mediated endocytosis, or affecting the reabsorption of glucose, the handling of uric acid, and the reabsorption of sodium, calcium, and magnesium along the kidney tubule.
Collapse
Affiliation(s)
- Jenny van der Wijst
- Department of Physiology, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center , Nijmegen , The Netherlands ; Institute of Physiology, University of Zurich , Zurich , Switzerland ; and Division of Nephrology, Institute of Experimental and Clinical Research (IREC), Medical School, Université catholique de Louvain, Brussels, Belgium
| | - Hendrica Belge
- Department of Physiology, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center , Nijmegen , The Netherlands ; Institute of Physiology, University of Zurich , Zurich , Switzerland ; and Division of Nephrology, Institute of Experimental and Clinical Research (IREC), Medical School, Université catholique de Louvain, Brussels, Belgium
| | - René J M Bindels
- Department of Physiology, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center , Nijmegen , The Netherlands ; Institute of Physiology, University of Zurich , Zurich , Switzerland ; and Division of Nephrology, Institute of Experimental and Clinical Research (IREC), Medical School, Université catholique de Louvain, Brussels, Belgium
| | - Olivier Devuyst
- Department of Physiology, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center , Nijmegen , The Netherlands ; Institute of Physiology, University of Zurich , Zurich , Switzerland ; and Division of Nephrology, Institute of Experimental and Clinical Research (IREC), Medical School, Université catholique de Louvain, Brussels, Belgium
| |
Collapse
|
34
|
Nedelyaeva OI, Shuvalov AV, Karpichev IV, Beliaev DV, Myasoedov NA, Khalilova LA, Khramov DE, Popova LG, Balnokin YV. Molecular cloning and characterisation of SaCLCa1, a novel protein of the chloride channel (CLC) family from the halophyte Suaeda altissima (L.) Pall. JOURNAL OF PLANT PHYSIOLOGY 2019; 240:152995. [PMID: 31252320 DOI: 10.1016/j.jplph.2019.152995] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/29/2018] [Revised: 06/07/2019] [Accepted: 06/07/2019] [Indexed: 06/09/2023]
Abstract
The SaCLCa1 gene, a putative orthologue of AtCLCa, the Arabidopsis thaliana gene encoding a NO3-/H+ antiporter, was cloned from the halophyte Suaeda altissima. It belonged to the CLC family, comprising anionic channels and anion/H+ antiporters. SaCLCa1 ion specificity was studied by heterologous expression of this gene in Saccharomyces cerevisiae GEF1 disrupted strain, Δgef1, where GEF1 encoded the only CLC family protein, the Cl- transporter Gef1p, in undisrupted strains of this organism. For comparison, the function of another recently identified S. altissima CLC family gene, SaCLCc1, was also characterised. Expression of SaCLCc1 in Δgef1 cells restored their ability to grow on selective media. This supported the chloride specificity of this transporter. By contrast, expression of SaCLCa1 did not complement the growth defect phenotype of Δgef1 cells. However, growth of the Δgef1 mutant on the selective media was partially restored when it was transformed with SaCLCa1(C562 T), encoding the modified protein SaCLCa1(P188S), in which proline responsible for NO3- selectivity in selective filter was replaced by serine providing chloride selectivity. Quantitative real-time polymerase chain reactions (qRT-PCR) showed that significant induction of SaCLCa1 occurred in the roots of S. altissima when plants were grown on nitrate-deficient medium, while SaCLCc1 activation was observed in S. altissima leaves of plants grown in increasing Cl- concentrations of nutrient solution. These results suggested that SaCLCa1 and SaCLCc1 function as anionic transporters with nitrate and chloride specificities, respectively.
Collapse
Affiliation(s)
- O I Nedelyaeva
- K.A. Timiryazev Institute of Plant Physiology, Russian Academy of Sciences, 127276, Moscow, Botanicheskaya str., 35, Russia.
| | - A V Shuvalov
- K.A. Timiryazev Institute of Plant Physiology, Russian Academy of Sciences, 127276, Moscow, Botanicheskaya str., 35, Russia.
| | - I V Karpichev
- K.A. Timiryazev Institute of Plant Physiology, Russian Academy of Sciences, 127276, Moscow, Botanicheskaya str., 35, Russia.
| | - D V Beliaev
- K.A. Timiryazev Institute of Plant Physiology, Russian Academy of Sciences, 127276, Moscow, Botanicheskaya str., 35, Russia.
| | - N A Myasoedov
- K.A. Timiryazev Institute of Plant Physiology, Russian Academy of Sciences, 127276, Moscow, Botanicheskaya str., 35, Russia.
| | - L A Khalilova
- K.A. Timiryazev Institute of Plant Physiology, Russian Academy of Sciences, 127276, Moscow, Botanicheskaya str., 35, Russia.
| | - D E Khramov
- K.A. Timiryazev Institute of Plant Physiology, Russian Academy of Sciences, 127276, Moscow, Botanicheskaya str., 35, Russia.
| | - L G Popova
- K.A. Timiryazev Institute of Plant Physiology, Russian Academy of Sciences, 127276, Moscow, Botanicheskaya str., 35, Russia.
| | - Y V Balnokin
- K.A. Timiryazev Institute of Plant Physiology, Russian Academy of Sciences, 127276, Moscow, Botanicheskaya str., 35, Russia.
| |
Collapse
|
35
|
Foco L, Pattaro C. Genetics of Blood Pressure Regulation: Possible Paths in the Labyrinth. Am J Kidney Dis 2019; 74:421-424. [PMID: 31221530 DOI: 10.1053/j.ajkd.2019.05.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2019] [Accepted: 05/03/2019] [Indexed: 11/11/2022]
Affiliation(s)
- Luisa Foco
- Eurac Research, Institute for Biomedicine, Bolzano, Italy
| | | |
Collapse
|
36
|
Weston MR, Mindell JA. Characterizing chloride-dependent acidification in brain clathrin-coated vesicles 1. Biochem Cell Biol 2019; 97:315-324. [PMID: 30383978 PMCID: PMC8404411 DOI: 10.1139/bcb-2018-0142] [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] [Indexed: 11/22/2022] Open
Abstract
Endocytic organelles maintain their acidic pH using the V-type ATPase proton pump. However, proton accumulation across the membrane generates a voltage and requires the movement of an additional ion, known as a counterion, to dissipate charge buildup. The role of counterion movement in endosomes is not clear, but a subpopulation of early endosomes, clathrin-coated vesicles (CCVs), has previously been shown to use external chloride (Cl-) to allow V-ATPase-dependent acidification. We aimed to determine the identity and function of this presumed Cl- transporting protein. Our sample of highly enriched bovine brain CCVs exhibited V-type ATPase-facilitated acidification in the presence of external Cl-, independent of the monovalent cations present. While unsuccessful at identifying the mechanism of anion transport, we used glutamate-facilitated acidification, density gradients, and mass spectrometry to show that most brain CCVs are synaptic vesicles, complementing results from earlier studies that argued similarity only on the basis on protein content. The source of Cl--dependent acidification in brain CCVs may be vGLUT1, a synaptic vesicle glutamate transporter with known Cl- permeability, although CCVs in other tissues are likely to utilize different proteins to facilitate acidification.
Collapse
Affiliation(s)
- Mary R. Weston
- Membrane Transport Biophysics Section, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, Maryland, United States of America 20892
| | - Joseph A. Mindell
- Membrane Transport Biophysics Section, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, Maryland, United States of America 20892
| |
Collapse
|
37
|
Yarotskyy V, Malysz J, Petkov GV. Properties of single-channel and whole cell Cl - currents in guinea pig detrusor smooth muscle cells. Am J Physiol Cell Physiol 2019; 316:C698-C710. [PMID: 30566392 DOI: 10.1152/ajpcell.00327.2018] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Multiple types of Cl- channels regulate smooth muscle excitability and contractility in vascular, gastrointestinal, and airway smooth muscle cells. However, little is known about Cl- channels in detrusor smooth muscle (DSM) cells. Here, we used inside-out single channel and whole cell patch-clamp recordings for detailed biophysical and pharmacological characterizations of Cl- channels in freshly isolated guinea pig DSM cells. The recorded single Cl- channels displayed unique gating with multiple subconductive states, a fully opened single-channel conductance of 164 pS, and a reversal potential of -41.5 mV, which is close to the ECl of -65 mV, confirming preferential permeability to Cl-. The Cl- channel demonstrated strong voltage dependence of activation (half-maximum of mean open probability, V0.5, ~-20 mV) and robust prolonged openings at depolarizing voltages. The channel displayed similar gating when exposed intracellularly to solutions containing Ca2+-free or 1 mM Ca2+. In whole cell patch-clamp recordings, macroscopic current demonstrated outward rectification, inhibitions by 4,4'-diisothiocyano-2,2'-stilbenedisulfonic acid (DIDS) and niflumic acid, and insensitivity to chlorotoxin. The outward current was reversibly reduced by 94% replacement of extracellular Cl- with I-, Br-, or methanesulfonate (MsO-), resulting in anionic permeability sequence: Cl->Br->I->MsO-. While intracellular Ca2+ levels (0, 300 nM, and 1 mM) did not affect the amplitude of Cl- current and outward rectification, high Ca2+ slowed voltage-step current activation at depolarizing voltages. In conclusion, our data reveal for the first time the presence of a Ca2+-independent DIDS and niflumic acid-sensitive, voltage-dependent Cl- channel in the plasma membrane of DSM cells. This channel may be a key regulator of DSM excitability.
Collapse
Affiliation(s)
- Viktor Yarotskyy
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Tennessee Health Science Center , Memphis, Tennessee
| | - John Malysz
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Tennessee Health Science Center , Memphis, Tennessee
| | - Georgi V Petkov
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Tennessee Health Science Center , Memphis, Tennessee
| |
Collapse
|
38
|
Duster AW, Garza CM, Aydintug BO, Negussie MB, Lin H. Adaptive Partitioning QM/MM for Molecular Dynamics Simulations: 6. Proton Transport through a Biological Channel. J Chem Theory Comput 2019; 15:892-905. [DOI: 10.1021/acs.jctc.8b01128] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Affiliation(s)
- Adam W. Duster
- Chemistry Department, CB 194, University of Colorado, Denver, Colorado 80217, United States
| | - Christina M. Garza
- Chemistry Department, CB 194, University of Colorado, Denver, Colorado 80217, United States
| | - Baris O. Aydintug
- Chemistry Department, CB 194, University of Colorado, Denver, Colorado 80217, United States
| | - Mikias B. Negussie
- Chemistry Department, CB 194, University of Colorado, Denver, Colorado 80217, United States
| | - Hai Lin
- Chemistry Department, CB 194, University of Colorado, Denver, Colorado 80217, United States
| |
Collapse
|
39
|
Structural biology and structure–function relationships of membrane proteins. Biochem Soc Trans 2018; 47:47-61. [DOI: 10.1042/bst20180269] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2018] [Revised: 10/22/2018] [Accepted: 10/29/2018] [Indexed: 01/02/2023]
Abstract
Abstract
The study of structure–function relationships of membrane proteins (MPs) has been one of the major goals in the field of structural biology. Many Noble Prizes regarding remarkable accomplishments in MP structure determination and biochemistry have been awarded over the last few decades. Mutations or improper folding of these proteins are associated with numerous serious illnesses. Therefore, as important drug targets, the study of their primary sequence and three-dimensional fold, combined with cell-based assays, provides vital information about their structure–function relationships. Today, this information is vital to drug discovery and medicine. In the last two decades, many have been the technical advances and breakthroughs in the field of MP structural biology that have contributed to an exponential growth in the number of unique MP structures in the Protein Data Bank. Nevertheless, given the medical importance and many unanswered questions, it will never be an excess of MP structures, regardless of the method used. Owing to the extension of the field, in this brief review, we will only focus on structure–function relationships of the three most significant pharmaceutical classes: G protein-coupled receptors, ion channels and transporters.
Collapse
|
40
|
Teulon J, Planelles G, Sepúlveda FV, Andrini O, Lourdel S, Paulais M. Renal Chloride Channels in Relation to Sodium Chloride Transport. Compr Physiol 2018; 9:301-342. [DOI: 10.1002/cphy.c180024] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
|
41
|
Guzman RE, Fahlke C. ClC-3: biophysical properties clarify cellular functions. J Physiol 2018; 596:3823-3824. [PMID: 29974478 DOI: 10.1113/jp276670] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022] Open
Affiliation(s)
- Raul E Guzman
- Institute of Complex Systems - Zelluläre Biophysik (ICS-4), Forschungszentrum Jülich, Jülich, 52425, Germany
| | - Christoph Fahlke
- Institute of Complex Systems - Zelluläre Biophysik (ICS-4), Forschungszentrum Jülich, Jülich, 52425, Germany
| |
Collapse
|
42
|
Thomassen M, Hostrup M, Murphy RM, Cromer BA, Skovgaard C, Gunnarsson TP, Christensen PM, Bangsbo J. Abundance of ClC-1 chloride channel in human skeletal muscle: fiber type specific differences and effect of training. J Appl Physiol (1985) 2018; 125:470-478. [DOI: 10.1152/japplphysiol.01042.2017] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Cl− channel protein 1 (ClC-1) may be important for excitability and contractility in skeletal muscle, but ClC-1 abundance has not been examined in human muscle. The aim of the present study was to examine ClC-1 abundance in human skeletal muscle, including fiber type specific differences and the effect of exercise training. A commercially available antibody was tested with positive and negative control tissue, and it recognized specifically ClC-1 in the range from 100 to 150 kDa. Abundance of ClC-1 was 38% higher ( P < 0.01) in fast twitch Type IIa muscle fibers than in slow twitch Type I. Muscle ClC-1 abundance did not change with 4 wk of training consisting of 30 min cycling at 85% of maximal heart rate (HRmax) and 3 × 30-s all out sprints or during a 7-wk training period with 10–12 × 30 s uphill cycling and 4–5 × ~4 min cycling at 90%–95% of HRmax. ClC-1 abundance correlated negatively ( P < 0.01) with maximal oxygen consumption ( r = –0.552) and incremental exercise performance ( r = –0.546). In addition, trained cyclists had lower ( P < 0.01) ClC-1 abundance than lesser trained individuals. The present observations indicate that a low abundance of muscle ClC-1 may be beneficial for exercise performance, but the role of abundance and regulation of ClC-1 in skeletal muscle of humans with respect to exercise performance and trainability need to be elucidated. NEW & NOTEWORTHY Abundance of the Cl− channel protein 1 (ClC-1) chloride channel may be important for excitability and contractility in human skeletal muscle and may therefore have implications for fatigue development. In this study, we confirmed ClC-1 specificity for a commercially available antibody, and this study is first to our knowledge to determine ClC-1 protein abundance in human muscle by Western blotting. We observed that abundance of ClC-1 was higher in fast compared with slow twitch fibers and lower in trained individuals than in recreationally active.
Collapse
Affiliation(s)
- Martin Thomassen
- Section of Integrative Physiology, Department of Nutrition, Exercise and Sports, University of Copenhagen, Copenhagen, Denmark
| | - Morten Hostrup
- Section of Integrative Physiology, Department of Nutrition, Exercise and Sports, University of Copenhagen, Copenhagen, Denmark
| | - Robyn M. Murphy
- Department of Biochemistry & Genetics, La Trobe Institute for Molecular Science, La Trobe University, Melbourne, Victoria, Australia
| | - Brett A. Cromer
- Department of Chemistry and Biotechnology, Swinburne University, Melbourne, Victoria, Australia
| | - Casper Skovgaard
- Section of Integrative Physiology, Department of Nutrition, Exercise and Sports, University of Copenhagen, Copenhagen, Denmark
| | - Thomas P. Gunnarsson
- Section of Integrative Physiology, Department of Nutrition, Exercise and Sports, University of Copenhagen, Copenhagen, Denmark
| | - Peter M. Christensen
- Section of Integrative Physiology, Department of Nutrition, Exercise and Sports, University of Copenhagen, Copenhagen, Denmark
| | - Jens Bangsbo
- Section of Integrative Physiology, Department of Nutrition, Exercise and Sports, University of Copenhagen, Copenhagen, Denmark
| |
Collapse
|
43
|
Fujimoto M, Kito H, Kajikuri J, Ohya S. Transcriptional repression of human epidermal growth factor receptor 2 by ClC-3 Cl - /H + transporter inhibition in human breast cancer cells. Cancer Sci 2018; 109:2781-2791. [PMID: 29949674 PMCID: PMC6125433 DOI: 10.1111/cas.13715] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2018] [Accepted: 06/23/2018] [Indexed: 12/13/2022] Open
Abstract
Recent studies have indicated that the intracellular concentration of chloride ions (Cl−) regulates gene expression in several types of cells and that Cl− modulators positively or negatively regulate the PI3K/AKT/mammalian target of rapamycin (mTOR) and signal transducer and activator of transcription (STAT)3 signaling pathways. We previously reported that the Ca2+‐activated Cl− channel anoctamine (ANO)1 regulated human epidermal growth factor receptor 2 (HER2) transcription in breast cancer YMB‐1 cells. However, the mechanisms underlying ANO1‐regulated HER2 gene expression have not yet been elucidated. In the present study, we showed the involvement of intracellular organelle ClC‐3 Cl−/H+ transporter in HER2 transcription in breast cancer MDA‐MB‐453 cells. The siRNA‐mediated inhibition of ClC‐3, but not ANO1, markedly repressed HER2 transcription in MDA‐MB‐453 cells. Subsequently, treatments with the AKT inhibitor AZD 5363 and mTOR inhibitor everolimus significantly enhanced HER2 transcription in MDA‐MB‐453 cells, whereas that with the STAT3 inhibitor 5,15‐diphenylporphyrin (5,15‐DPP) inhibited it. AKT and mTOR inhibitors also significantly enhanced HER2 transcription in YMB‐1 cells. The siRNA‐mediated inhibition of ClC‐3 and ANO1 resulted in increased AKT phosphorylation and decreased STAT3 phosphorylation in MDA‐MB‐453 and YMB‐1 cells, respectively. The intracellular Cl− channel protein CLIC1 was expressed in both cells; however, its siRNA‐mediated inhibition did not elicit the transcriptional repression of HER2. Collectively, our results demonstrate that intracellular Cl− regulation by ANO1/ClC‐3 participates in HER2 transcription, mediating the PI3K/AKT/mTOR and/or STAT3 signaling pathway(s) in HER2‐positive breast cancer cells, and support the potential of ANO1/ClC‐3 blockers as therapeutic options for patients with resistance to anti‐HER2 therapies.
Collapse
Affiliation(s)
- Mayu Fujimoto
- Department of Pharmacology, Division of Pathological Sciences, Kyoto Pharmaceutical University, Kyoto, Japan
| | - Hiroaki Kito
- Department of Pharmacology, Graduate School of Medical Sciences, Nagoya City University, Nagoya, Japan
| | - Junko Kajikuri
- Department of Pharmacology, Graduate School of Medical Sciences, Nagoya City University, Nagoya, Japan
| | - Susumu Ohya
- Department of Pharmacology, Division of Pathological Sciences, Kyoto Pharmaceutical University, Kyoto, Japan.,Department of Pharmacology, Graduate School of Medical Sciences, Nagoya City University, Nagoya, Japan
| |
Collapse
|
44
|
Jentsch TJ, Pusch M. CLC Chloride Channels and Transporters: Structure, Function, Physiology, and Disease. Physiol Rev 2018; 98:1493-1590. [DOI: 10.1152/physrev.00047.2017] [Citation(s) in RCA: 214] [Impact Index Per Article: 30.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
CLC anion transporters are found in all phyla and form a gene family of eight members in mammals. Two CLC proteins, each of which completely contains an ion translocation parthway, assemble to homo- or heteromeric dimers that sometimes require accessory β-subunits for function. CLC proteins come in two flavors: anion channels and anion/proton exchangers. Structures of these two CLC protein classes are surprisingly similar. Extensive structure-function analysis identified residues involved in ion permeation, anion-proton coupling and gating and led to attractive biophysical models. In mammals, ClC-1, -2, -Ka/-Kb are plasma membrane Cl−channels, whereas ClC-3 through ClC-7 are 2Cl−/H+-exchangers in endolysosomal membranes. Biological roles of CLCs were mostly studied in mammals, but also in plants and model organisms like yeast and Caenorhabditis elegans. CLC Cl−channels have roles in the control of electrical excitability, extra- and intracellular ion homeostasis, and transepithelial transport, whereas anion/proton exchangers influence vesicular ion composition and impinge on endocytosis and lysosomal function. The surprisingly diverse roles of CLCs are highlighted by human and mouse disorders elicited by mutations in their genes. These pathologies include neurodegeneration, leukodystrophy, mental retardation, deafness, blindness, myotonia, hyperaldosteronism, renal salt loss, proteinuria, kidney stones, male infertility, and osteopetrosis. In this review, emphasis is laid on biophysical structure-function analysis and on the cell biological and organismal roles of mammalian CLCs and their role in disease.
Collapse
Affiliation(s)
- Thomas J. Jentsch
- Leibniz-Forschungsinstitut für Molekulare Pharmakologie (FMP) and Max-Delbrück-Centrum für Molekulare Medizin (MDC), Berlin, Germany; and Istituto di Biofisica, Consiglio Nazionale delle Ricerche, Genova, Italy
| | - Michael Pusch
- Leibniz-Forschungsinstitut für Molekulare Pharmakologie (FMP) and Max-Delbrück-Centrum für Molekulare Medizin (MDC), Berlin, Germany; and Istituto di Biofisica, Consiglio Nazionale delle Ricerche, Genova, Italy
| |
Collapse
|
45
|
Bignon Y, Alekov A, Frachon N, Lahuna O, Jean-Baptiste Doh-Egueli C, Deschênes G, Vargas-Poussou R, Lourdel S. A novel CLCN5 pathogenic mutation supports Dent disease with normal endosomal acidification. Hum Mutat 2018; 39:1139-1149. [PMID: 29791050 DOI: 10.1002/humu.23556] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2018] [Revised: 04/23/2018] [Accepted: 05/19/2018] [Indexed: 12/13/2022]
Abstract
Dent disease is an X-linked recessive renal tubular disorder characterized by low-molecular-weight proteinuria, hypercalciuria, nephrolithiasis, nephrocalcinosis, and progressive renal failure. Inactivating mutations of CLCN5, the gene encoding the 2Cl- /H+ exchanger ClC-5, have been reported in patients with Dent disease 1. In vivo studies in mice harboring an artificial mutation in the "gating glutamate" of ClC-5 (c.632A > C, p.Glu211Ala) and mathematical modeling suggest that endosomal chloride concentration could be an important parameter in endocytosis, rather than acidification as earlier hypothesized. Here, we described a novel pathogenic mutation affecting the "gating glutamate" of ClC-5 (c.632A>G, p.Glu211Gly) and investigated its molecular consequences. In HEK293T cells, the p.Glu211Gly ClC-5 mutant displayed unaltered N-glycosylation and normal plasma membrane and early endosomes localizations. In Xenopus laevis oocytes and HEK293T cells, we found that contrasting with wild-type ClC-5, the mutation abolished the outward rectification, the sensitivity to extracellular H+ and converted ClC-5 into a Cl- channel. Investigation of endosomal acidification in HEK293T cells using the pH-sensitive pHluorin2 probe showed that the luminal pH of cells expressing a wild-type or p.Glu211Gly ClC-5 was not significantly different. Our study further confirms that impaired acidification of endosomes is not the only parameter leading to defective endocytosis in Dent disease 1.
Collapse
Affiliation(s)
- Yohan Bignon
- Sorbonne Université, Université Paris-Descartes, INSERM, CNRS, Paris, France
| | - Alexi Alekov
- Institut für Neurophysiologie, Medizinische Hochschule Hannover, Hannover, Germany
| | - Nadia Frachon
- Sorbonne Université, Université Paris-Descartes, INSERM, CNRS, Paris, France
| | | | | | - Georges Deschênes
- Assistance Publique-Hôpitaux de Paris, Hôpital Robert Debré, Service de Néphrologie Pédiatrique, Paris, France.,Centre de Référence des Maladies Rénales Héréditaires de l'Enfant et de l'Adulte (MARHEA), Paris, France
| | - Rosa Vargas-Poussou
- Assistance Publique-Hôpitaux de Paris, Hôpital Européen Georges Pompidou, Département de génétique, Paris, France.,Université Paris-Descartes, Faculté de Médecine, Paris, France
| | - Stéphane Lourdel
- Sorbonne Université, Université Paris-Descartes, INSERM, CNRS, Paris, France
| |
Collapse
|
46
|
Zhang Y, Zhou L, Zhang J, Zhang L, Yan X, Su J. Suppression of chloride voltage-gated channel 3 expression increases sensitivity of human glioma U251 cells to cisplatin through lysosomal dysfunction. Oncol Lett 2018; 16:835-842. [PMID: 29963152 PMCID: PMC6019884 DOI: 10.3892/ol.2018.8736] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2017] [Accepted: 05/03/2018] [Indexed: 11/05/2022] Open
Abstract
The mechanism of cisplatin resistance is complex. Previous studies have indicated that chloride voltage-gated channel 3 (CLCN3) is associated with drug resistance; however, the mechanisms are not fully understood. Therefore, the present study explored the involvement of CLCN3 in cisplatin resistance in human glioma U251 cells. The effects of combined cisplatin treatment and CLCN3 suppression on cultured U251 cells were investigated. The decreased viability of cisplatin-treated U251 cells indicated the cytotoxic effects of CLCN3 silencing. Expression of the apoptosis-related gene TP53 and caspase 3 activation were enhanced in cisplatin-treated U251 cells. Furthermore, the ratio of BCL2/BAX expression was decreased. Notably, CLCN3 suppression promoted cisplatin-induced cell damage in U251 cells. Thus, the combined use of cisplatin and CLCN3 antisense had additive effects in U251 cells. In addition, the present results indicated that CLCN3 suppression decreased lysosome stabilization in U251 cells treated with cisplatin. To conclude, the present results indicated that CLCN3 suppression can sensitize glioma cells to cisplatin through lysosomal dysfunction.
Collapse
Affiliation(s)
- Yihe Zhang
- Department of Pathophysiology, Key Laboratory of Pathobiology, Ministry of Education, College of Basic Medical Sciences, Jilin University, Changchun, Jilin 130021, P.R. China.,Department of Neurology, The First Bethune Hospital of Jilin University, Changchun, Jilin 130021, P.R. China
| | - Lei Zhou
- Department of Pathology, Affiliated Hospital to Changchun University of Chinese Medicine, Changchun, Jilin 130021, P.R. China
| | - Juanjuan Zhang
- Department of Pathophysiology, Key Laboratory of Pathobiology, Ministry of Education, College of Basic Medical Sciences, Jilin University, Changchun, Jilin 130021, P.R. China
| | - Lichao Zhang
- Department of Pathophysiology, Key Laboratory of Pathobiology, Ministry of Education, College of Basic Medical Sciences, Jilin University, Changchun, Jilin 130021, P.R. China
| | - Xiaoyu Yan
- Department of Pathophysiology, Key Laboratory of Pathobiology, Ministry of Education, College of Basic Medical Sciences, Jilin University, Changchun, Jilin 130021, P.R. China
| | - Jing Su
- Department of Pathophysiology, Key Laboratory of Pathobiology, Ministry of Education, College of Basic Medical Sciences, Jilin University, Changchun, Jilin 130021, P.R. China
| |
Collapse
|
47
|
Rahmati N, Hoebeek FE, Peter S, De Zeeuw CI. Chloride Homeostasis in Neurons With Special Emphasis on the Olivocerebellar System: Differential Roles for Transporters and Channels. Front Cell Neurosci 2018; 12:101. [PMID: 29765304 PMCID: PMC5938380 DOI: 10.3389/fncel.2018.00101] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2017] [Accepted: 03/28/2018] [Indexed: 12/14/2022] Open
Abstract
The intraneuronal ionic composition is an important determinant of brain functioning. There is growing evidence that aberrant homeostasis of the intracellular concentration of Cl- ([Cl-]i) evokes, in addition to that of Na+ and Ca2+, robust impairments of neuronal excitability and neurotransmission and thereby neurological conditions. More specifically, understanding the mechanisms underlying regulation of [Cl-]i is crucial for deciphering the variability in GABAergic and glycinergic signaling of neurons, in both health and disease. The homeostatic level of [Cl-]i is determined by various regulatory mechanisms, including those mediated by plasma membrane Cl- channels and transporters. This review focuses on the latest advances in identification, regulation and characterization of Cl- channels and transporters that modulate neuronal excitability and cell volume. By putting special emphasis on neurons of the olivocerebellar system, we establish that Cl- channels and transporters play an indispensable role in determining their [Cl-]i and thereby their function in sensorimotor coordination.
Collapse
Affiliation(s)
- Negah Rahmati
- Department of Neuroscience, Erasmus Medical Center, Rotterdam, Netherlands
| | - Freek E. Hoebeek
- Department of Neuroscience, Erasmus Medical Center, Rotterdam, Netherlands
- NIDOD Institute, Wilhelmina Children's Hospital, University Medical Center Utrecht and Brain Center Rudolf Magnus, Utrecht, Netherlands
| | - Saša Peter
- Department of Neuroscience, Erasmus Medical Center, Rotterdam, Netherlands
| | - Chris I. De Zeeuw
- Department of Neuroscience, Erasmus Medical Center, Rotterdam, Netherlands
- Netherlands Institute for Neuroscience, Royal Dutch Academy for Arts and Sciences, Amsterdam, Netherlands
| |
Collapse
|
48
|
Abstract
CLC proteins are a ubiquitously expressed family of chloride-selective ion channels and transporters. A dearth of pharmacological tools for modulating CLC gating and ion conduction limits investigations aimed at understanding CLC structure/function and physiology. Herein, we describe the design, synthesis, and evaluation of a collection of N-arylated benzimidazole derivatives (BIMs), one of which (BIM1) shows unparalleled (>20-fold) selectivity for CLC-Ka over CLC-Kb, the two most closely related human CLC homologs. Computational docking to a CLC-Ka homology model has identified a BIM1 binding site on the extracellular face of the protein near the chloride permeation pathway in a region previously identified as a binding site for other less selective inhibitors. Results from site-directed mutagenesis experiments are consistent with predictions of this docking model. The residue at position 68 is 1 of only ∼20 extracellular residues that differ between CLC-Ka and CLC-Kb. Mutation of this residue in CLC-Ka and CLC-Kb (N68D and D68N, respectively) reverses the preference of BIM1 for CLC-Ka over CLC-Kb, thus showing the critical role of residue 68 in establishing BIM1 selectivity. Molecular docking studies together with results from structure-activity relationship studies with 19 BIM derivatives give insight into the increased selectivity of BIM1 compared with other inhibitors and identify strategies for further developing this class of compounds.
Collapse
|
49
|
Wang CH, Duster AW, Aydintug BO, Zarecki MG, Lin H. Chloride Ion Transport by the E. coli CLC Cl -/H + Antiporter: A Combined Quantum-Mechanical and Molecular-Mechanical Study. Front Chem 2018; 6:62. [PMID: 29594103 PMCID: PMC5859129 DOI: 10.3389/fchem.2018.00062] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2018] [Accepted: 02/26/2018] [Indexed: 01/05/2023] Open
Abstract
We performed steered molecular dynamics (SMD) and umbrella sampling simulations of Cl- ion migration through the transmembrane domain of a prototypical E. coli CLC Cl-/H+ antiporter by employing combined quantum-mechanical (QM) and molecular-mechanical (MM) calculations. The SMD simulations revealed interesting conformational changes of the protein. While no large-amplitude motions of the protein were observed during pore opening, the side chain rotation of the protonated external gating residue Glu148 was found to be critical for full access of the channel entrance by Cl-. Moving the anion into the external binding site (Sext) induced small-amplitude shifting of the protein backbone at the N-terminal end of helix F. As Cl- traveled through the pore, rigid-body swinging motions of helix R separated it from helix D. Helix R returned to its original position once Cl- exited the channel. Population analysis based on polarized wavefunction from QM/MM calculations discovered significant (up to 20%) charge loss for Cl- along the ion translocation pathway inside the pore. The delocalized charge was redistributed onto the pore residues, especially the functional groups containing π bonds (e.g., the Tyr445 side chain), while the charges of the H atoms coordinating Cl- changed almost negligibly. Potentials of mean force computed from umbrella sampling at the QM/MM and MM levels both displayed barriers at the same locations near the pore entrance and exit. However, the QM/MM PMF showed higher barriers (~10 kcal/mol) than the MM PMF (~2 kcal/mol). Binding energy calculations indicated that the interactions between Cl- and certain pore residues were overestimated by the semi-empirical PM3 Hamiltonian and underestimated by the CHARMM36 force fields, both of which were employed in the umbrella sampling simulations. In particular, CHARMM36 underestimated binding interactions for the functional groups containing π bonds, missing the stabilizations of the Cl- ion due to electron delocalization. The results suggested that it is important to explore these quantum effects for accurate descriptions of the Cl- transport.
Collapse
Affiliation(s)
- Chun-Hung Wang
- Department of Chemistry, University of Colorado Denver, Denver, CO, United States
| | - Adam W Duster
- Department of Chemistry, University of Colorado Denver, Denver, CO, United States
| | - Baris O Aydintug
- Department of Chemistry, University of Colorado Denver, Denver, CO, United States
| | - MacKenzie G Zarecki
- Department of Chemistry, University of Colorado Denver, Denver, CO, United States
| | - Hai Lin
- Department of Chemistry, University of Colorado Denver, Denver, CO, United States
| |
Collapse
|
50
|
Gaitán-Peñas H, Armand-Ugón M, Macaya A, Estévez R. CLCN1 Myotonia congenita mutation with a variable pattern of inheritance suggests a novel mechanism of dominant myotonia. Muscle Nerve 2018; 58:157-160. [PMID: 29424939 DOI: 10.1002/mus.26098] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/06/2018] [Indexed: 11/07/2022]
Abstract
INTRODUCTION Mutations in CLCN1 cause recessive or dominant forms of myotonia congenita (MC). Some mutations have been found to exhibit both patterns of inheritance but the mechanism explaining this behavior is unknown. METHODS A known recessive missense mutation, A493E, was identified in a family with dominant MC. The mutant p.A493E alone or in co-expression with wild-type (WT) ClC-1 was expressed in Xenopus oocytes. Currents were measured and biochemical assays were performed. RESULTS The mutant showed no significant activity and reduced total and plasma membrane (PM) protein levels. Co-expression with the mutant reduced the activity and PM levels of an engineered lower expression variant of ClC-1, whereas no effect was observed on a higher expression variant. DISCUSSION Our results suggest that the dominant effect of some CLCN1 mutations showing recessive or dominant inheritance patterns may be due to a dose-dependent defect in PM delivery of the WT channel. Muscle Nerve, 2018.
Collapse
Affiliation(s)
- Héctor Gaitán-Peñas
- Unitat de Fisiologia, Departament de Ciències Fisiològiques, IDIBELL-Universitat de Barcelona (Institut de Neurociències), L'Hospitalet de Llobregat, Spain
- U-750, CIBERER, ISCIII, Barcelona, Spain
| | - Mercedes Armand-Ugón
- Unitat de Fisiologia, Departament de Ciències Fisiològiques, IDIBELL-Universitat de Barcelona (Institut de Neurociències), L'Hospitalet de Llobregat, Spain
| | - Alfons Macaya
- Pediatric Neurology Research Group, Vall d'Hebron Research Institute, Hospital Universitari de Vall d'Hebron, Autonomous University of Barcelona, Barcelona, Spain
| | - Raúl Estévez
- Unitat de Fisiologia, Departament de Ciències Fisiològiques, IDIBELL-Universitat de Barcelona (Institut de Neurociències), L'Hospitalet de Llobregat, Spain
- U-750, CIBERER, ISCIII, Barcelona, Spain
| |
Collapse
|