1
|
Uvarov P, Fudo S, Karakus C, Golubtsov A, Rotondo F, Sukhanova T, Soni S, Di Scala C, Kajander T, Rivera C, Ludwig A. Uncovering novel KCC2 regulatory motifs through a comprehensive transposon-based mutant library. Front Mol Neurosci 2025; 17:1505722. [PMID: 39881966 PMCID: PMC11774852 DOI: 10.3389/fnmol.2024.1505722] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2024] [Accepted: 12/10/2024] [Indexed: 01/31/2025] Open
Abstract
Introduction The neuron-specific K-Cl cotransporter KCC2 maintains low intracellular chloride levels, which are crucial for fast GABAergic and glycinergic neurotransmission. KCC2 also plays a pivotal role in the development of excitatory glutamatergic neurotransmission by promoting dendritic spine maturation. The cytoplasmic C-terminal domain (KCC2-CTD) plays a critical regulatory role in the molecular mechanisms controlling the cotransporter activity through dimerization, phosphorylation, and protein interaction. Methods To identify novel CTD regulatory motifs, we used the Mu transposon-based mutagenesis system to generate a library of KCC2 mutants with 5 amino acid insertions randomly distributed within the KCC2-CTD. We determined the insertion positions in 288 mutants by restriction analysis and selected clones with a single insertion site outside known KCC2 regulatory motifs. We analyzed the subcellular distribution of KCC2-CTD mutants in cultured cortical neurons using immunocytochemistry and selected ten mutants with ectopic expression patterns for detailed characterization. Results A fluorescent Cl--transport assay in HEK293 cells revealed mutants with both reduced and enhanced Cl--extrusion activity, which overall correlated with their glycosylation patterns. Live-cell immunostaining analysis of plasma membrane expression of KCC2-CTD mutants in cultured cortical neurons corroborated the glycosylation data. Furthermore, the somatodendritic chloride gradient in neurons transfected with the KCC2-CTD mutants correlated with their Cl--extrusion activity in HEK293 cells. Gain- and loss-of-function mutant positions were analyzed using available KCC2 cryo-EM structures. Discussion Two groups of mutants were identified based on 3D structural analysis. The first group, located near the interface of transmembrane and cytoplasmic domains, may affect interactions with the N-terminal inhibitory peptide regulating KCC2 activity. The second group, situated on the external surface of the cytoplasmic domain, may disrupt interactions with regulatory proteins. Analyzing CTD mutations that modulate KCC2 activity enhances our understanding of its function and is essential for developing novel anti-seizure therapies.
Collapse
Affiliation(s)
- Pavel Uvarov
- Neuroscience Center, HiLIFE, University of Helsinki, Helsinki, Finland
| | - Satoshi Fudo
- Institute of Biotechnology, HiLIFE, University of Helsinki, Helsinki, Finland
| | - Cem Karakus
- Neuroscience Center, HiLIFE, University of Helsinki, Helsinki, Finland
| | - Andrey Golubtsov
- Neuroscience Center, HiLIFE, University of Helsinki, Helsinki, Finland
| | - Federico Rotondo
- Neuroscience Center, HiLIFE, University of Helsinki, Helsinki, Finland
| | - Tatiana Sukhanova
- Neuroscience Center, HiLIFE, University of Helsinki, Helsinki, Finland
| | - Shetal Soni
- Neuroscience Center, HiLIFE, University of Helsinki, Helsinki, Finland
| | - Coralie Di Scala
- Neuroscience Center, HiLIFE, University of Helsinki, Helsinki, Finland
- INSERM, INMED, Aix Marseille University, Marseille, France
| | - Tommi Kajander
- Institute of Biotechnology, HiLIFE, University of Helsinki, Helsinki, Finland
| | - Claudio Rivera
- Neuroscience Center, HiLIFE, University of Helsinki, Helsinki, Finland
- INSERM, INMED, Aix Marseille University, Marseille, France
| | - Anastasia Ludwig
- Neuroscience Center, HiLIFE, University of Helsinki, Helsinki, Finland
| |
Collapse
|
2
|
Sagi R, Chakraborty M, Bogdanovic M, Asraf H, Sekler I, Kofman O, Cohen H, Hershfinkel M. Loss of the zinc receptor ZnR/GPR39 in mice enhances anxiety-related behavior and motor deficits, and modulates KCC2 expression in the amygdala. BEHAVIORAL AND BRAIN FUNCTIONS : BBF 2024; 20:31. [PMID: 39581978 PMCID: PMC11587656 DOI: 10.1186/s12993-024-00254-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/17/2023] [Accepted: 10/18/2024] [Indexed: 11/26/2024]
Abstract
BACKGROUND Mood disorders, particularly depression and anxiety, are associated with zinc dyshomeostasis and aberrant GABAergic signaling. Activation of ZnR/GPR39 by synaptic zinc in the hippocampus triggers phosphorylation of extracellular regulated kinase (ERK1/2), which regulates the K+/Cl- cotransporter (KCC2) and thereby GABAergic inhibitory neurotransmission and seizure activity. Therefore, we studied whether impaired ZnR/GPR39 signaling is linked to anxiety-related behavior in male or female mice. RESULTS Using the acoustic startle response, elevated plus maze, and open field test, we found increased anxiety-related behavior in ZnR/GPR39 knockout (KO) mice. Despite a well-established sex difference, where females are typically more prone to anxiety, both male and female ZnR/GPR39 KO mice exhibited increased anxiety-related behavior compared to wildtype (WT) mice. Additionally, ZnR/GPR39 KO mice displayed impaired motor coordination in the pole and rotarod tests but did not show reduced muscle strength, as indicated by a grip test. Finally, we found intrinsic alterations in the expression level of KCC2, a major Cl- transporter regulating GABAergic signaling, in the amygdala of naïve ZnR/GPR39 KO mice compared to controls. CONCLUSIONS Our findings indicate that loss of ZnR/GPR39 enhances anxiety-related behavior in both male and female mice. Moreover, ZnR/GPR39 KO mice exhibit impaired motor coordination, which may be associated with increased anxiety. Finally, we demonstrate that loss of ZnR/GPR39 modulates the expression of KCC2 in the amygdala. Thus, we propose that ZnR/GPR39 can serve as a target for regulating GABAergic signaling in anxiety treatment.
Collapse
Affiliation(s)
- Romi Sagi
- Department of Physiology and Cell Biology, School of Biomedical Research, Faculty of Health Sciences, Zelman Center for Neuroscience, Ben-Gurion University of the Negev, POB 653, Beer-Sheva, 84105, Israel.
- The Gonda Multidisciplinary Brain Research Center, Bar-Ilan University, Ramat-Gan, Israel.
| | - Moumita Chakraborty
- Department of Physiology and Cell Biology, School of Biomedical Research, Faculty of Health Sciences, Zelman Center for Neuroscience, Ben-Gurion University of the Negev, POB 653, Beer-Sheva, 84105, Israel
- Translational Hepatology Section, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, MD, 20892, USA
| | - Milos Bogdanovic
- Department of Physiology and Cell Biology, School of Biomedical Research, Faculty of Health Sciences, Zelman Center for Neuroscience, Ben-Gurion University of the Negev, POB 653, Beer-Sheva, 84105, Israel
| | - Hila Asraf
- Department of Physiology and Cell Biology, School of Biomedical Research, Faculty of Health Sciences, Zelman Center for Neuroscience, Ben-Gurion University of the Negev, POB 653, Beer-Sheva, 84105, Israel
| | - Israel Sekler
- Department of Physiology and Cell Biology, School of Biomedical Research, Faculty of Health Sciences, Zelman Center for Neuroscience, Ben-Gurion University of the Negev, POB 653, Beer-Sheva, 84105, Israel
| | - Ora Kofman
- Psychology Department, Zelman Center for Neuroscience, Ben-Gurion University of the Negev, POB 653, Beer-Sheva, 84105, Israel
| | - Hagit Cohen
- Department of Physiology and Cell Biology, School of Biomedical Research, Faculty of Health Sciences, Zelman Center for Neuroscience, Ben-Gurion University of the Negev, POB 653, Beer-Sheva, 84105, Israel
- Anxiety and Stress Research Unit, Beer-Sheva Mental Health Center, POB 653, Beer-Sheva, 84105, Israel
| | - Michal Hershfinkel
- Department of Physiology and Cell Biology, School of Biomedical Research, Faculty of Health Sciences, Zelman Center for Neuroscience, Ben-Gurion University of the Negev, POB 653, Beer-Sheva, 84105, Israel.
| |
Collapse
|
3
|
Vásquez E, Oresti GM, Paez MD, Callegari EA, Masone D, Muñoz EM. Impact of aging on the GABA B receptor-mediated connectome. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.07.31.606013. [PMID: 39131332 PMCID: PMC11312617 DOI: 10.1101/2024.07.31.606013] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 08/13/2024]
Abstract
GABA B receptors (GABABRs) are heterodimeric seven-transmembrane receptors that interact with a range of proteins and form large protein complexes on cholesterol-rich membrane microdomains. As the brain ages, membrane cholesterol levels exhibit alterations, although it remains unclear how these changes impact protein-protein interactions and downstream signaling. Herein, we studied the structural bases for the interaction between GABABR and the KCC2 transporter, including their protein expression and distribution, and we compared data between young and aged rat cerebella. Also, we analyzed lipid profiles for both groups, and we used molecular dynamics simulations on three plasma membrane systems with different cholesterol concentrations, to further explore the GABABR-transporter interaction. Based on our results, we report that a significant decrease in GABAB2 subunit expression occurs in the aged rat cerebella. After performing a comparative co-immunoprecipitation analysis, we confirm that GABABR and KCC2 form a protein complex in adult and aged rat cerebella, although their interaction levels are reduced substantially as the cerebellum ages. On the other hand, our lipid analyses reveal a significant increase in cholesterol and sphingomyelin levels of the aged cerebella. Finally, we used the Martini coarse-grained model to conduct molecular dynamics simulations, from which we observed that membrane cholesterol concentrations can dictate whether the GABABR tail domains physically establish G protein-independent contacts with a transporter, and the timing when those associations eventually occur. Taken together, our findings illustrate how age-related alterations in membrane cholesterol levels affect protein-protein interactions, and how they could play a crucial role in regulating GABABR's interactome-mediated signaling. Significance Statement This study elucidates age-related changes in cerebellar GABAB receptors (GABABRs), KCC2, and plasma membrane lipids, shedding light on mechanisms underlying neurological decline. Molecular dynamics simulations reveal how membrane lipids influence protein-protein interactions, offering insights into age-related neurodegeneration. The findings underscore the broader impact of cerebellar aging on motor functions, cognition, and emotional processing in the elderly. By elucidating plasma membrane regulation and GABAergic dynamics, this research lays the groundwork for understanding aging-related neurological disorders and inspires further investigation into therapeutic interventions.
Collapse
Affiliation(s)
- Elena Vásquez
- Instituto de Histología y Embriología de Mendoza (IHEM), Universidad Nacional de Cuyo, Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Mendoza, Argentina
| | - Gerardo M. Oresti
- Instituto de Investigaciones Bioquímicas de Bahía Blanca, Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET) y Departamento de Biología, Bioquímica y Farmacia, Universidad Nacional del Sur (UNS), Bahía Blanca, Argentina
| | - María D. Paez
- Division of Basic Biomedical Sciences, University of South Dakota Sanford School of Medicine, Vermillion, SD, USA
| | - Eduardo A. Callegari
- Division of Basic Biomedical Sciences, University of South Dakota Sanford School of Medicine, Vermillion, SD, USA
| | - Diego Masone
- Instituto de Histología y Embriología de Mendoza (IHEM), Universidad Nacional de Cuyo, Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Mendoza, Argentina
| | - Estela M. Muñoz
- Instituto de Histología y Embriología de Mendoza (IHEM), Universidad Nacional de Cuyo, Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Mendoza, Argentina
| |
Collapse
|
4
|
Kok M, Brodsky JL. The biogenesis of potassium transporters: implications of disease-associated mutations. Crit Rev Biochem Mol Biol 2024; 59:154-198. [PMID: 38946646 PMCID: PMC11444911 DOI: 10.1080/10409238.2024.2369986] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2024] [Revised: 06/02/2024] [Accepted: 06/16/2024] [Indexed: 07/02/2024]
Abstract
The concentration of intracellular and extracellular potassium is tightly regulated due to the action of various ion transporters, channels, and pumps, which reside primarily in the kidney. Yet, potassium transporters and cotransporters play vital roles in all organs and cell types. Perhaps not surprisingly, defects in the biogenesis, function, and/or regulation of these proteins are linked to range of catastrophic human diseases, but to date, few drugs have been approved to treat these maladies. In this review, we discuss the structure, function, and activity of a group of potassium-chloride cotransporters, the KCCs, as well as the related sodium-potassium-chloride cotransporters, the NKCCs. Diseases associated with each of the four KCCs and two NKCCs are also discussed. Particular emphasis is placed on how these complex membrane proteins fold and mature in the endoplasmic reticulum, how non-native forms of the cotransporters are destroyed in the cell, and which cellular factors oversee their maturation and transport to the cell surface. When known, we also outline how the levels and activities of each cotransporter are regulated. Open questions in the field and avenues for future investigations are further outlined.
Collapse
Affiliation(s)
- Morgan Kok
- Department of Biological Sciences, University of Pittsburgh, Pittsburgh, PA, USA
| | - Jeffrey L Brodsky
- Department of Biological Sciences, University of Pittsburgh, Pittsburgh, PA, USA
| |
Collapse
|
5
|
Kok M, Hartnett-Scott K, Happe CL, MacDonald ML, Aizenman E, Brodsky JL. The expression system influences stability, maturation efficiency, and oligomeric properties of the potassium-chloride co-transporter KCC2. Neurochem Int 2024; 174:105695. [PMID: 38373478 PMCID: PMC10923169 DOI: 10.1016/j.neuint.2024.105695] [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: 09/08/2023] [Revised: 02/02/2024] [Accepted: 02/06/2024] [Indexed: 02/21/2024]
Abstract
The neuron-specific K+/Cl- co-transporter 2, KCC2, which is critical for brain development, regulates γ-aminobutyric acid-dependent inhibitory neurotransmission. Consistent with its function, mutations in KCC2 are linked to neurodevelopmental disorders, including epilepsy, schizophrenia, and autism. KCC2 possesses 12 transmembrane spans and forms an intertwined dimer. Based on its complex architecture and function, reduced cell surface expression and/or activity have been reported when select disease-associated mutations are present in the gene encoding the protein, SLC12A5. These data suggest that KCC2 might be inherently unstable, as seen for other complex polytopic ion channels, thus making it susceptible to cellular quality control pathways that degrade misfolded proteins. To test these hypotheses, we examined KCC2 stability and/or maturation in five model systems: yeast, HEK293 cells, primary rat neurons, and rat and human brain synaptosomes. Although studies in yeast revealed that KCC2 is selected for endoplasmic reticulum-associated degradation (ERAD), experiments in HEK293 cells supported a more subtle role for ERAD in maintaining steady-state levels of KCC2. Nevertheless, this system allowed for an analysis of KCC2 glycosylation in the ER and Golgi, which serves as a read-out for transport through the secretory pathway. In turn, KCC2 was remarkably stable in primary rat neurons, suggesting that KCC2 folds efficiently in more native systems. Consistent with these data, the mature glycosylated form of KCC2 was abundant in primary rat neurons as well as in rat and human brain. Together, this work details the first insights into the influence that the cellular and membrane environments have on several fundamental KCC2 properties, acknowledges the advantages and disadvantages of each system, and helps set the stage for future experiments to assess KCC2 in a normal or disease setting.
Collapse
Affiliation(s)
- Morgan Kok
- Department of Biological Sciences, University of Pittsburgh, Pittsburgh, PA, USA
| | - Karen Hartnett-Scott
- Department of Neurobiology and the Pittsburgh Institute for Neurodegenerative Diseases, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Cassandra L Happe
- Department of Psychiatry, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Matthew L MacDonald
- Department of Psychiatry, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Elias Aizenman
- Department of Neurobiology and the Pittsburgh Institute for Neurodegenerative Diseases, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Jeffrey L Brodsky
- Department of Biological Sciences, University of Pittsburgh, Pittsburgh, PA, USA.
| |
Collapse
|
6
|
McMoneagle E, Zhou J, Zhang S, Huang W, Josiah SS, Ding K, Wang Y, Zhang J. Neuronal K +-Cl - cotransporter KCC2 as a promising drug target for epilepsy treatment. Acta Pharmacol Sin 2024; 45:1-22. [PMID: 37704745 PMCID: PMC10770335 DOI: 10.1038/s41401-023-01149-9] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2023] [Accepted: 08/02/2023] [Indexed: 09/14/2023] Open
Abstract
Epilepsy is a prevalent neurological disorder characterized by unprovoked seizures. γ-Aminobutyric acid (GABA) serves as the primary fast inhibitory neurotransmitter in the brain, and GABA binding to the GABAA receptor (GABAAR) regulates Cl- and bicarbonate (HCO3-) influx or efflux through the channel pore, leading to GABAergic inhibition or excitation, respectively. The neuron-specific K+-Cl- cotransporter 2 (KCC2) is essential for maintaining a low intracellular Cl- concentration, ensuring GABAAR-mediated inhibition. Impaired KCC2 function results in GABAergic excitation associated with epileptic activity. Loss-of-function mutations and altered expression of KCC2 lead to elevated [Cl-]i and compromised synaptic inhibition, contributing to epilepsy pathogenesis in human patients. KCC2 antagonism studies demonstrate the necessity of limiting neuronal hyperexcitability within the brain, as reduced KCC2 functioning leads to seizure activity. Strategies focusing on direct (enhancing KCC2 activation) and indirect KCC2 modulation (altering KCC2 phosphorylation and transcription) have proven effective in attenuating seizure severity and exhibiting anti-convulsant properties. These findings highlight KCC2 as a promising therapeutic target for treating epilepsy. Recent advances in understanding KCC2 regulatory mechanisms, particularly via signaling pathways such as WNK, PKC, BDNF, and its receptor TrkB, have led to the discovery of novel small molecules that modulate KCC2. Inhibiting WNK kinase or utilizing newly discovered KCC2 agonists has demonstrated KCC2 activation and seizure attenuation in animal models. This review discusses the role of KCC2 in epilepsy and evaluates its potential as a drug target for epilepsy treatment by exploring various strategies to regulate KCC2 activity.
Collapse
Affiliation(s)
- Erin McMoneagle
- Institute of Biomedical and Clinical Sciences, Medical School, Faculty of Health and Life Sciences, University of Exeter, Hatherly Laboratories, Streatham Campus, Exeter, EX4 4PS, UK
| | - Jin Zhou
- Department of Neurology, Institutes of Brain Science, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institute of Biological Science, Zhongshan Hospital, Fudan University, Shanghai, 200032, China
| | - Shiyao Zhang
- Institute of Cardiovascular Diseases, Xiamen Cardiovascular Hospital Xiamen University, School of Medicine, Xiamen University, Xiang'an Nan Lu, Xiamen, 361102, China
| | - Weixue Huang
- State Key Laboratory of Chemical Biology, Research Center of Chemical Kinomics, Shanghai Institute of Organic Chemistry, Chinese Academy of Sciences, Shanghai, 200032, China
| | - Sunday Solomon Josiah
- Institute of Biomedical and Clinical Sciences, Medical School, Faculty of Health and Life Sciences, University of Exeter, Hatherly Laboratories, Streatham Campus, Exeter, EX4 4PS, UK
| | - Ke Ding
- State Key Laboratory of Chemical Biology, Research Center of Chemical Kinomics, Shanghai Institute of Organic Chemistry, Chinese Academy of Sciences, Shanghai, 200032, China.
| | - Yun Wang
- Department of Neurology, Institutes of Brain Science, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institute of Biological Science, Zhongshan Hospital, Fudan University, Shanghai, 200032, China.
| | - Jinwei Zhang
- Institute of Biomedical and Clinical Sciences, Medical School, Faculty of Health and Life Sciences, University of Exeter, Hatherly Laboratories, Streatham Campus, Exeter, EX4 4PS, UK.
- Institute of Cardiovascular Diseases, Xiamen Cardiovascular Hospital Xiamen University, School of Medicine, Xiamen University, Xiang'an Nan Lu, Xiamen, 361102, China.
- State Key Laboratory of Chemical Biology, Research Center of Chemical Kinomics, Shanghai Institute of Organic Chemistry, Chinese Academy of Sciences, Shanghai, 200032, China.
| |
Collapse
|
7
|
Radulovic T, Rajaram E, Ebbers L, Pagella S, Winklhofer M, Kopp-Scheinpflug C, Nothwang HG, Milenkovic I, Hartmann AM. Serine 937 phosphorylation enhances KCC2 activity and strengthens synaptic inhibition. Sci Rep 2023; 13:21660. [PMID: 38066086 PMCID: PMC10709408 DOI: 10.1038/s41598-023-48884-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2023] [Accepted: 11/30/2023] [Indexed: 12/18/2023] Open
Abstract
The potassium chloride cotransporter KCC2 is crucial for Cl- extrusion from mature neurons and thus key to hyperpolarizing inhibition. Auditory brainstem circuits contain well-understood inhibitory projections and provide a potent model to study the regulation of synaptic inhibition. Two peculiarities of the auditory brainstem are (i) posttranslational activation of KCC2 during development and (ii) extremely negative reversal potentials in specific circuits. To investigate the role of the potent phospho-site serine 937 therein, we generated a KCC2 Thr934Ala/Ser937Asp double mutation, in which Ser937 is replaced by aspartate mimicking the phosphorylated state, and the neighbouring Thr934 arrested in the dephosphorylated state. This double mutant showed a twofold increased transport activity in HEK293 cells, raising the hypothesis that auditory brainstem neurons show lower [Cl-]i. and increased glycinergic inhibition. This was tested in a mouse model carrying the same KCC2 Thr934Ala/Ser937Asp mutation by the use of the CRISPR/Cas9 technology. Homozygous KCC2 Thr934Ala/Ser937Asp mice showed an earlier developmental onset of hyperpolarisation in the auditory brainstem. Mature neurons displayed stronger glycinergic inhibition due to hyperpolarized ECl-. These data demonstrate that phospho-regulation of KCC2 Ser937 is a potent way to interfere with the excitation-inhibition balance in neural circuits.
Collapse
Affiliation(s)
- Tamara Radulovic
- Division of Physiology School of Medicine and Health Sciences, Carl Von Ossietzky University Oldenburg, 26111, Oldenburg, Germany
- Research Center Neurosensory Science, Carl Von Ossietzky University Oldenburg, 26111, Oldenburg, Germany
| | - Ezhilarasan Rajaram
- Division of Neurobiology, Faculty of Biology, Ludwig-Maximilians-University Munich, 82152, Planegg-Martinsried, Germany
| | - Lena Ebbers
- Division of Neurogenetics, School of Medicine and Health Sciences, Carl Von Ossietzky University Oldenburg, 26111, Oldenburg, Germany
| | - Sara Pagella
- Division of Neurobiology, Faculty of Biology, Ludwig-Maximilians-University Munich, 82152, Planegg-Martinsried, Germany
| | - Michael Winklhofer
- Research Center Neurosensory Science, Carl Von Ossietzky University Oldenburg, 26111, Oldenburg, Germany
- Institute for Biology and Environmental Sciences IBU, Carl Von Ossietzky University of Oldenburg, 26111, Oldenburg, Germany
| | - Conny Kopp-Scheinpflug
- Division of Neurobiology, Faculty of Biology, Ludwig-Maximilians-University Munich, 82152, Planegg-Martinsried, Germany
| | - Hans Gerd Nothwang
- Division of Neurogenetics, School of Medicine and Health Sciences, Carl Von Ossietzky University Oldenburg, 26111, Oldenburg, Germany
- Research Center Neurosensory Science, Carl Von Ossietzky University Oldenburg, 26111, Oldenburg, Germany
- Center of Excellence Hearing4all, Carl Von Ossietzky University Oldenburg, 26111, Oldenburg, Germany
| | - Ivan Milenkovic
- Division of Physiology School of Medicine and Health Sciences, Carl Von Ossietzky University Oldenburg, 26111, Oldenburg, Germany
- Research Center Neurosensory Science, Carl Von Ossietzky University Oldenburg, 26111, Oldenburg, Germany
| | - Anna-Maria Hartmann
- Division of Neurogenetics, School of Medicine and Health Sciences, Carl Von Ossietzky University Oldenburg, 26111, Oldenburg, Germany.
- Research Center Neurosensory Science, Carl Von Ossietzky University Oldenburg, 26111, Oldenburg, Germany.
| |
Collapse
|
8
|
Zhang S, Meor Azlan NF, Josiah SS, Zhou J, Zhou X, Jie L, Zhang Y, Dai C, Liang D, Li P, Li Z, Wang Z, Wang Y, Ding K, Wang Y, Zhang J. The role of SLC12A family of cation-chloride cotransporters and drug discovery methodologies. J Pharm Anal 2023; 13:1471-1495. [PMID: 38223443 PMCID: PMC10785268 DOI: 10.1016/j.jpha.2023.09.002] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2023] [Revised: 06/20/2023] [Accepted: 09/05/2023] [Indexed: 09/12/2023] Open
Abstract
The solute carrier family 12 (SLC12) of cation-chloride cotransporters (CCCs) comprises potassium chloride cotransporters (KCCs, e.g. KCC1, KCC2, KCC3, and KCC4)-mediated Cl- extrusion, and sodium potassium chloride cotransporters (N[K]CCs, NKCC1, NKCC2, and NCC)-mediated Cl- loading. The CCCs play vital roles in cell volume regulation and ion homeostasis. Gain-of-function or loss-of-function of these ion transporters can cause diseases in many tissues. In recent years, there have been considerable advances in our understanding of CCCs' control mechanisms in cell volume regulations, with many techniques developed in studying the functions and activities of CCCs. Classic approaches to directly measure CCC activity involve assays that measure the transport of potassium substitutes through the CCCs. These techniques include the ammonium pulse technique, radioactive or nonradioactive rubidium ion uptake-assay, and thallium ion-uptake assay. CCCs' activity can also be indirectly observed by measuring γ-aminobutyric acid (GABA) activity with patch-clamp electrophysiology and intracellular chloride concentration with sensitive microelectrodes, radiotracer 36Cl-, and fluorescent dyes. Other techniques include directly looking at kinase regulatory sites phosphorylation, flame photometry, 22Na+ uptake assay, structural biology, molecular modeling, and high-throughput drug screening. This review summarizes the role of CCCs in genetic disorders and cell volume regulation, current methods applied in studying CCCs biology, and compounds developed that directly or indirectly target the CCCs for disease treatments.
Collapse
Affiliation(s)
- Shiyao Zhang
- Xiamen Cardiovascular Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, Fujian, 363001, China
| | - Nur Farah Meor Azlan
- Institute of Biomedical and Clinical Sciences, Medical School, Faculty of Health and Life Sciences, University of Exeter, Exeter, EX4 4PS, UK
| | - Sunday Solomon Josiah
- Institute of Biomedical and Clinical Sciences, Medical School, Faculty of Health and Life Sciences, University of Exeter, Exeter, EX4 4PS, UK
| | - Jing Zhou
- Department of Neurology, Institutes of Brain Science, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institute of Biological Science, Zhongshan Hospital, Fudan University, Shanghai, 200032, China
| | - Xiaoxia Zhou
- Xiamen Cardiovascular Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, Fujian, 363001, China
| | - Lingjun Jie
- Xiamen Cardiovascular Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, Fujian, 363001, China
| | - Yanhui Zhang
- Xiamen Cardiovascular Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, Fujian, 363001, China
| | - Cuilian Dai
- Xiamen Cardiovascular Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, Fujian, 363001, China
| | - Dong Liang
- Aurora Discovery Inc., Foshan, Guangdong, 528300, China
| | - Peifeng Li
- Institute for Translational Medicine, Qingdao University, Qingdao, Shandong, 266021, China
| | - Zhengqiu Li
- School of Pharmacy, Jinan University, Guangzhou, 510632, China
| | - Zhen Wang
- State Key Laboratory of Chemical Biology, Research Center of Chemical Kinomics, Shanghai Institute of Organic Chemistry, Chinese Academy of Sciences, Shanghai, 200032, China
| | - Yun Wang
- Department of Neurology, Institutes of Brain Science, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institute of Biological Science, Zhongshan Hospital, Fudan University, Shanghai, 200032, China
| | - Ke Ding
- State Key Laboratory of Chemical Biology, Research Center of Chemical Kinomics, Shanghai Institute of Organic Chemistry, Chinese Academy of Sciences, Shanghai, 200032, China
| | - Yan Wang
- Xiamen Cardiovascular Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, Fujian, 363001, China
| | - Jinwei Zhang
- Xiamen Cardiovascular Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, Fujian, 363001, China
- Institute of Biomedical and Clinical Sciences, Medical School, Faculty of Health and Life Sciences, University of Exeter, Exeter, EX4 4PS, UK
- State Key Laboratory of Chemical Biology, Research Center of Chemical Kinomics, Shanghai Institute of Organic Chemistry, Chinese Academy of Sciences, Shanghai, 200032, China
| |
Collapse
|
9
|
Sun C, Deng J, Ma Y, Meng F, Cui X, Li M, Li J, Li J, Yin P, Kong L, Zhang L, Tang P. The dual role of microglia in neuropathic pain after spinal cord injury: Detrimental and protective effects. Exp Neurol 2023; 370:114570. [PMID: 37852469 DOI: 10.1016/j.expneurol.2023.114570] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2023] [Revised: 09/21/2023] [Accepted: 10/11/2023] [Indexed: 10/20/2023]
Abstract
Spinal cord injury (SCI) is a debilitating condition that is frequently accompanied by neuropathic pain, resulting in significant physical and psychological harm to a vast number of individuals globally. Despite the high prevalence of neuropathic pain following SCI, the precise underlying mechanism remains incompletely understood. Microglia are a type of innate immune cell that are present in the central nervous system (CNS). They have been observed to have a significant impact on neuropathic pain following SCI. This article presents a comprehensive overview of recent advances in understanding the role of microglia in the development of neuropathic pain following SCI. Specifically, the article delves into the detrimental and protective effects of microglia on neuropathic pain following SCI, as well as the mechanisms underlying their interconversion. Furthermore, the article provides a thorough overview of potential avenues for future research in this area.
Collapse
Affiliation(s)
- Chang Sun
- Department of Orthopedics, The Fourth Medical Center of Chinese PLA General Hospital, Beijing, China; National Clinical Research Center for Orthopedics, Sports Medicine and Rehabilitation, Beijing, China; Department of Orthopedics, Air Force Medical Center, PLA, Beijing, China
| | - Junhao Deng
- Department of Orthopedics, The Fourth Medical Center of Chinese PLA General Hospital, Beijing, China; National Clinical Research Center for Orthopedics, Sports Medicine and Rehabilitation, Beijing, China; School of Life Sciences, Tsinghua University, Beijing, China; State Key Laboratory of Precision Measurement Technology and Instruments, Department of Precision Instruments, Tsinghua University, Beijing, China; IDG/McGovern Institute for Brain Research, Tsinghua University, Beijing, China
| | - Yifei Ma
- School of Medicine, Nankai University, Tianjin, China
| | - Fanqi Meng
- Department of Anesthesiology, Xuanwu Hospital, Capital Medical University, Beijing, China
| | - Xiang Cui
- Department of Orthopedics, The Fourth Medical Center of Chinese PLA General Hospital, Beijing, China; National Clinical Research Center for Orthopedics, Sports Medicine and Rehabilitation, Beijing, China
| | - Ming Li
- Department of Orthopedics, The Fourth Medical Center of Chinese PLA General Hospital, Beijing, China; National Clinical Research Center for Orthopedics, Sports Medicine and Rehabilitation, Beijing, China
| | - Jiantao Li
- Department of Orthopedics, The Fourth Medical Center of Chinese PLA General Hospital, Beijing, China; National Clinical Research Center for Orthopedics, Sports Medicine and Rehabilitation, Beijing, China
| | - Jia Li
- Department of Orthopedics, The Fourth Medical Center of Chinese PLA General Hospital, Beijing, China; National Clinical Research Center for Orthopedics, Sports Medicine and Rehabilitation, Beijing, China
| | - Pengbin Yin
- Department of Orthopedics, The Fourth Medical Center of Chinese PLA General Hospital, Beijing, China; National Clinical Research Center for Orthopedics, Sports Medicine and Rehabilitation, Beijing, China
| | - Lingjie Kong
- State Key Laboratory of Precision Measurement Technology and Instruments, Department of Precision Instruments, Tsinghua University, Beijing, China; IDG/McGovern Institute for Brain Research, Tsinghua University, Beijing, China.
| | - Licheng Zhang
- Department of Orthopedics, The Fourth Medical Center of Chinese PLA General Hospital, Beijing, China; National Clinical Research Center for Orthopedics, Sports Medicine and Rehabilitation, Beijing, China.
| | - Peifu Tang
- Department of Orthopedics, The Fourth Medical Center of Chinese PLA General Hospital, Beijing, China; National Clinical Research Center for Orthopedics, Sports Medicine and Rehabilitation, Beijing, China.
| |
Collapse
|
10
|
Lee J, Clause A, Kandler K. Structural and Functional Development of Inhibitory Connections from the Medial Nucleus of the Trapezoid Body to the Superior Paraolivary Nucleus. J Neurosci 2023; 43:7766-7779. [PMID: 37734946 PMCID: PMC10648534 DOI: 10.1523/jneurosci.0920-23.2023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2023] [Revised: 08/21/2023] [Accepted: 09/13/2023] [Indexed: 09/23/2023] Open
Abstract
The medial nucleus of the trapezoid body (MNTB) in the auditory brainstem is the principal source of synaptic inhibition to several functionally distinct auditory nuclei. Prominent projections of individual MNTB neurons comprise the major binaural nuclei that are involved in the early processing stages of sound localization as well as the superior paraolivary nucleus (SPON), which contains monaural neurons that extract rapid changes in sound intensity to detect sound gaps and rhythmic oscillations that commonly occur in animal calls and human speech. While the processes that guide the development and refinement of MNTB axon collaterals to the binaural nuclei have become increasingly understood, little is known about the development of MNTB collaterals to the monaural SPON. In this study, we investigated the development of MNTB-SPON connections in mice of both sexes from shortly after birth to three weeks of age, which encompasses the time before and after hearing onset. Individual axon reconstructions and electrophysiological analysis of MNTB-SPON connectivity demonstrate a dramatic increase in the number of MNTB axonal boutons in the SPON before hearing onset. However, this proliferation was not accompanied by changes in the strength of MNTB-SPON connections or by changes in the structural or functional topographic precision. However, following hearing onset, the spread of single-axon boutons along the tonotopic axis increased, indicating an unexpected decrease in the tonotopic precision of the MNTB-SPON pathway. These results provide new insight into the development and organization of inhibition to SPON neurons and the regulation of developmental plasticity in diverging inhibitory pathways.SIGNIFICANCE STATEMENT The superior paraolivary nucleus (SPON) is a prominent auditory brainstem nucleus involved in the early detection of sound gaps and rhythmic oscillations. The ability of SPON neurons to fire at the offset of sound depends on strong and precise synaptic inhibition provided by glycinergic neurons in the medial nucleus of the trapezoid body (MNTB). Here, we investigated the anatomic and physiological maturation of MNTB-LSO connectivity in mice before and after the onset of hearing. We observed a period of bouton proliferation without accompanying changes in topographic precision before hearing onset. This was followed by bouton elimination and an unexpected decrease in the tonotopic precision after hearing onset. These results provide new insight into the development of inhibition to the SPON.
Collapse
Affiliation(s)
- Jongwon Lee
- Department of Neurobiology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania 15261
| | - Amanda Clause
- Department of Neurobiology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania 15261
| | - Karl Kandler
- Department of Neurobiology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania 15261
- Department of Bioengineering, University of Pittsburgh, Pittsburgh, Pennsylvania 15261
| |
Collapse
|
11
|
Trejo F, Elizalde S, Mercado A, Gamba G, de losHeros P. SLC12A cryo-EM: analysis of relevant ion binding sites, structural domains, and amino acids. Am J Physiol Cell Physiol 2023; 325:C921-C939. [PMID: 37545407 DOI: 10.1152/ajpcell.00089.2023] [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: 03/13/2023] [Revised: 07/24/2023] [Accepted: 07/24/2023] [Indexed: 08/08/2023]
Abstract
The solute carrier family 12A (SLC12A) superfamily of membrane transporters modulates the movement of cations coupled with chloride across the membrane. In doing so, these cotransporters are involved in numerous aspects of human physiology: cell volume regulation, ion homeostasis, blood pressure regulation, and neurological action potential via intracellular chloride concentration modulation. Their physiological characterization has been largely studied; however, understanding the mechanics of their function and the relevance of structural domains or specific amino acids has been a pending task. In recent years, single-particle cryogenic electron microscopy (cryo-EM) has been successfully applied to members of the SLC12A family including all K+:Cl- cotransporters (KCCs), Na+:K+:2Cl- cotransporter NKCC1, and recently Na+:Cl- cotransporter (NCC); revealing structural elements that play key roles in their function. The present review analyzes the data provided by these cryo-EM reports focusing on structural domains and specific amino acids involved in ion binding, domain interactions, and other important SCL12A structural elements. A comparison of cryo-EM data from NKCC1 and KCCs is presented in the light of the two recent NCC cryo-EM studies, to propose insight into structural elements that might also be found in NCC and are necessary for its proper function. In the final sections, the importance of key coordination residues for substrate specificity and their implication on various pathophysiological conditions and genetic disorders is reviewed, as this could provide the basis to correlate structural elements with the development of novel and selective treatments, as well as mechanistic insight into the function and regulation of cation-coupled chloride cotransporters (CCCs).
Collapse
Affiliation(s)
- Fátima Trejo
- Unidad de Investigación UNAM-INC, Facultad de Medicina, Universidad Nacional Autónoma de México, Mexico City, Mexico
| | - Stephanie Elizalde
- Departamento de Nefrología y Metabolismo Mineral, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Mexico City, Mexico
| | - Adriana Mercado
- Departamento de Bioquímica, Instituto Nacional de Cardiología Ignacio Chávez, Mexico City, Mexico
| | - Gerardo Gamba
- Departamento de Nefrología y Metabolismo Mineral, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Mexico City, Mexico
- Molecular Physiology Unit, Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México, Mexico City, Mexico
| | - Paola de losHeros
- Unidad de Investigación UNAM-INC, Facultad de Medicina, Universidad Nacional Autónoma de México, Mexico City, Mexico
| |
Collapse
|
12
|
Furukawa T, Fukuda A. Maternal taurine as a modulator of Cl - homeostasis as well as of glycine/GABA A receptors for neocortical development. Front Cell Neurosci 2023; 17:1221441. [PMID: 37601283 PMCID: PMC10435090 DOI: 10.3389/fncel.2023.1221441] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2023] [Accepted: 07/20/2023] [Indexed: 08/22/2023] Open
Abstract
During brain and spinal cord development, GABA and glycine, the inhibitory neurotransmitters, cause depolarization instead of hyperpolarization in adults. Since glycine and GABAA receptors (GABAARs) are chloride (Cl-) ion channel receptor, the conversion of GABA/glycine actions during development is influenced by changes in the transmembrane Cl- gradient, which is regulated by Cl- transporters, NKCC1 (absorption) and KCC2 (expulsion). In immature neurons, inhibitory neurotransmitters are released in a non-vesicular/non-synaptic manner, transitioning to vesicular/synaptic release as the neuron matures. In other word, in immature neurons, neurotransmitters generally act tonically. Thus, the glycine/GABA system is a developmentally multimodal system that is required for neurogenesis, differentiation, migration, and synaptogenesis. The endogenous agonists for these receptors are not fully understood, we address taurine. In this review, we will discuss about the properties and function of taurine during development of neocortex. Taurine cannot be synthesized by fetuses or neonates, and is transferred from maternal blood through the placenta or maternal milk ingestion. In developing neocortex, taurine level is higher than GABA level, and taurine tonically activates GABAARs to control radial migration as a stop signal. In the marginal zone (MZ) of the developing neocortex, endogenous taurine modulates the spread of excitatory synaptic transmission, activating glycine receptors (GlyRs) as an endogenous agonist. Thus, taurine affects information processing and crucial developmental processes such as axonal growth, cell migration, and lamination in the developing cerebral cortex. Additionally, we also refer to the possible mechanism of taurine-regulating Cl- homeostasis. External taurine is uptake by taurine transporter (TauT) and regulates NKCC1 and KCC2 mediated by intracellular signaling pathway, with-no-lysine kinase 1 (WNK1) and its subsequent kinases STE20/SPS1-related proline-alanine-rich protein kinase (SPAK) and oxidative stress response kinase-1 (OSR1). Through the regulation of NKCC1 and KCC2, mediated by the WNK-SPAK/OSR1 signaling pathway, taurine plays a role in maintaining Cl- homeostasis during normal brain development.
Collapse
Affiliation(s)
- Tomonori Furukawa
- Department of Neurophysiology, Hirosaki University Graduate School of Medicine, Hirosaki, Japan
| | - Atsuo Fukuda
- Department of Neurophysiology, Hamamatsu University School of Medicine, Hamamatsu, Japan
| |
Collapse
|
13
|
Pressey JC, de Saint-Rome M, Raveendran VA, Woodin MA. Chloride transporters controlling neuronal excitability. Physiol Rev 2023; 103:1095-1135. [PMID: 36302178 DOI: 10.1152/physrev.00025.2021] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023] Open
Abstract
Synaptic inhibition plays a crucial role in regulating neuronal excitability, which is the foundation of nervous system function. This inhibition is largely mediated by the neurotransmitters GABA and glycine that activate Cl--permeable ion channels, which means that the strength of inhibition depends on the Cl- gradient across the membrane. In neurons, the Cl- gradient is primarily mediated by two secondarily active cation-chloride cotransporters (CCCs), NKCC1 and KCC2. CCC-mediated regulation of the neuronal Cl- gradient is critical for healthy brain function, as dysregulation of CCCs has emerged as a key mechanism underlying neurological disorders including epilepsy, neuropathic pain, and autism spectrum disorder. This review begins with an overview of neuronal chloride transporters before explaining the dependent relationship between these CCCs, Cl- regulation, and inhibitory synaptic transmission. We then discuss the evidence for how CCCs can be regulated, including by activity and their protein interactions, which underlie inhibitory synaptic plasticity. For readers who may be interested in conducting experiments on CCCs and neuronal excitability, we have included a section on techniques for estimating and recording intracellular Cl-, including their advantages and limitations. Although the focus of this review is on neurons, we also examine how Cl- is regulated in glial cells, which in turn regulate neuronal excitability through the tight relationship between this nonneuronal cell type and synapses. Finally, we discuss the relatively extensive and growing literature on how CCC-mediated neuronal excitability contributes to neurological disorders.
Collapse
Affiliation(s)
- Jessica C Pressey
- Department of Cell and Systems Biology, University of Toronto, Toronto, Ontario, Canada
| | - Miranda de Saint-Rome
- Department of Cell and Systems Biology, University of Toronto, Toronto, Ontario, Canada
| | - Vineeth A Raveendran
- Department of Cell and Systems Biology, University of Toronto, Toronto, Ontario, Canada
| | - Melanie A Woodin
- Department of Cell and Systems Biology, University of Toronto, Toronto, Ontario, Canada
| |
Collapse
|
14
|
Hu JJ, Liu Y, Yao H, Cao B, Liao H, Yang R, Chen P, Song XJ. Emergence of consciousness from anesthesia through ubiquitin degradation of KCC2 in the ventral posteromedial nucleus of the thalamus. Nat Neurosci 2023; 26:751-764. [PMID: 36973513 DOI: 10.1038/s41593-023-01290-y] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2021] [Accepted: 02/23/2023] [Indexed: 03/29/2023]
Abstract
The emergence of consciousness from anesthesia, once assumed to be a passive process, is now considered as an active and controllable process. In the present study, we show in mice that, when the brain is forced into a minimum responsive state by diverse anesthetics, a rapid downregulation of K+/Cl- cotransporter 2 (KCC2) in the ventral posteromedial nucleus (VPM) serves as a common mechanism by which the brain regains consciousness. Ubiquitin-proteasomal degradation is responsible for KCC2 downregulation, which is driven by ubiquitin ligase Fbxl4. Phosphorylation of KCC2 at Thr1007 promotes interaction between KCC2 and Fbxl4. KCC2 downregulation leads to γ-aminobutyric acid type A receptor-mediated disinhibition, enabling accelerated recovery of VPM neuron excitability and emergence of consciousness from anesthetic inhibition. This pathway to recovery is an active process and occurs independent of anesthetic choice. The present study demonstrates that ubiquitin degradation of KCC2 in the VPM is an important intermediate step en route to emergence of consciousness from anesthesia.
Collapse
|
15
|
Tsuji M, Mukai T, Sato Y, Azuma Y, Yamamoto S, Cayetanot F, Bodineau L, Onoda A, Nagamura-Inoue T, Coq JO. Umbilical cord-derived mesenchymal stromal cell therapy to prevent the development of neurodevelopmental disorders related to low birth weight. Sci Rep 2023; 13:3841. [PMID: 36882440 PMCID: PMC9992354 DOI: 10.1038/s41598-023-30817-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2022] [Accepted: 03/01/2023] [Indexed: 03/09/2023] Open
Abstract
Low birth weight (LBW) increases the risk of neurodevelopmental disorders (NDDs) such as attention-deficit/hyperactive disorder and autism spectrum disorder, as well as cerebral palsy, for which no prophylactic measure exists. Neuroinflammation in fetuses and neonates plays a major pathogenic role in NDDs. Meanwhile, umbilical cord-derived mesenchymal stromal cells (UC-MSCs) exhibit immunomodulatory properties. Therefore, we hypothesized that systemic administration of UC-MSCs in the early postnatal period may attenuate neuroinflammation and thereby prevent the emergence of NDDs. The LBW pups born to dams subjected to mild intrauterine hypoperfusion exhibited a significantly lesser decrease in the monosynaptic response with increased frequency of stimulation to the spinal cord preparation from postnatal day 4 (P4) to P6, suggesting hyperexcitability, which was improved by intravenous administration of human UC-MSCs (1 × 105 cells) on P1. Three-chamber sociability tests at adolescence revealed that only LBW males exhibited disturbed sociability, which tended to be ameliorated by UC-MSC treatment. Other parameters, including those determined via open-field tests, were not significantly improved by UC-MSC treatment. Serum or cerebrospinal fluid levels of pro-inflammatory cytokines were not elevated in the LBW pups, and UC-MSC treatment did not decrease these levels. In conclusion, although UC-MSC treatment prevents hyperexcitability in LBW pups, beneficial effects for NDDs are marginal.
Collapse
Affiliation(s)
- Masahiro Tsuji
- Department of Food and Nutrition, Kyoto Women's University, 35 Kitahiyoshi-cho, Imakumano, Higashiyama-ku, Kyoto, 605-8501, Japan.
| | - Takeo Mukai
- Department of Cell Processing and Transfusion, Institute of Medical Science, The University of Tokyo, Tokyo, Japan
| | - Yoshiaki Sato
- Division of Neonatology, Center for Maternal-Neonatal Care, Nagoya University Hospital, Nagoya, Japan
| | - Yasue Azuma
- Department of Food and Nutrition, Kyoto Women's University, 35 Kitahiyoshi-cho, Imakumano, Higashiyama-ku, Kyoto, 605-8501, Japan
| | - Saki Yamamoto
- Department of Food and Nutrition, Kyoto Women's University, 35 Kitahiyoshi-cho, Imakumano, Higashiyama-ku, Kyoto, 605-8501, Japan
| | - Florence Cayetanot
- Institut National de la Santé et de la Recherche Médicale (Inserm), UMR_S1158 Neurophysiologie Respiratoire Expérimentale et Clinique, Sorbonne Université, Paris, France
| | - Laurence Bodineau
- Institut National de la Santé et de la Recherche Médicale (Inserm), UMR_S1158 Neurophysiologie Respiratoire Expérimentale et Clinique, Sorbonne Université, Paris, France
| | - Atsuto Onoda
- Division of Neonatology, Center for Maternal-Neonatal Care, Nagoya University Hospital, Nagoya, Japan
| | - Tokiko Nagamura-Inoue
- Department of Cell Processing and Transfusion, Institute of Medical Science, The University of Tokyo, Tokyo, Japan
| | - Jacques-Olivier Coq
- Centre National de la Recherche Scientifique (CNRS), Institut des Sciences du Mouvement (ISM) UMR7287, Aix Marseille Université, 163 avenue de Luminy, CC 910, 13288, Marseille Cedex 09, France.
| |
Collapse
|
16
|
Kurki SN, Uvarov P, Pospelov AS, Trontti K, Hübner AK, Srinivasan R, Watanabe M, Hovatta I, Hübner CA, Kaila K, Virtanen MA. Expression patterns of NKCC1 in neurons and non-neuronal cells during cortico-hippocampal development. Cereb Cortex 2022; 33:5906-5923. [PMID: 36573432 PMCID: PMC10183754 DOI: 10.1093/cercor/bhac470] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2022] [Revised: 11/05/2022] [Accepted: 11/06/2022] [Indexed: 12/28/2022] Open
Abstract
Abstract
The Na-K-2Cl cotransporter NKCC1 is widely expressed in cells within and outside the brain. However, our understanding of its roles in brain functions throughout development, as well as in neuropsychiatric and neurological disorders, has been severely hindered by the lack of reliable data on its developmental and (sub)cellular expression patterns. We provide here the first properly controlled analysis of NKCC1 protein expression in various cell types of the mouse brain using custom-made antibodies and an NKCC1 knock-out validated immunohistochemical procedure, with parallel data based on advanced mRNA approaches. NKCC1 protein and mRNA are expressed at remarkably high levels in oligodendrocytes. In immature neurons, NKCC1 protein was located in the somata, whereas in adult neurons, only NKCC1 mRNA could be clearly detected. NKCC1 immunoreactivity is also seen in microglia, astrocytes, developing pericytes, and in progenitor cells of the dentate gyrus. Finally, a differential expression of NKCC1 splice variants was observed, with NKCC1a predominating in non-neuronal cells and NKCC1b in neurons. Taken together, our data provide a cellular basis for understanding NKCC1 functions in the brain and enable the identification of major limitations and promises in the development of neuron-targeting NKCC1-blockers.
Collapse
Affiliation(s)
- Samu N Kurki
- University of Helsinki Molecular and Integrative Biosciences, , 00014 Helsinki , Finland
- Helsinki Institute of Life Science, University of Helsinki Neuroscience Center, , 00014 Helsinki , Finland
| | - Pavel Uvarov
- University of Helsinki Molecular and Integrative Biosciences, , 00014 Helsinki , Finland
- Helsinki Institute of Life Science, University of Helsinki Neuroscience Center, , 00014 Helsinki , Finland
| | - Alexey S Pospelov
- University of Helsinki Molecular and Integrative Biosciences, , 00014 Helsinki , Finland
- Helsinki Institute of Life Science, University of Helsinki Neuroscience Center, , 00014 Helsinki , Finland
| | - Kalevi Trontti
- Helsinki Institute of Life Science, University of Helsinki Neuroscience Center, , 00014 Helsinki , Finland
- University of Helsinki SleepWell Research Program, Faculty of Medicine, , 00014 Helsinki , Finland
- University of Helsinki Department of Psychology and Logopedics, , 00014 Helsinki , Finland
| | - Antje K Hübner
- Jena University Hospital, Friedrich Schiller Universität Institute of Human Genetics, , 07747 Jena , Germany
| | - Rakenduvadhana Srinivasan
- University of Helsinki Molecular and Integrative Biosciences, , 00014 Helsinki , Finland
- Helsinki Institute of Life Science, University of Helsinki Neuroscience Center, , 00014 Helsinki , Finland
| | - Masahiko Watanabe
- Hokkaido University Department of Anatomy, Faculty of Medicine, , Sapporo 060–8638 , Japan
| | - Iiris Hovatta
- Helsinki Institute of Life Science, University of Helsinki Neuroscience Center, , 00014 Helsinki , Finland
- University of Helsinki SleepWell Research Program, Faculty of Medicine, , 00014 Helsinki , Finland
- University of Helsinki Department of Psychology and Logopedics, , 00014 Helsinki , Finland
| | - Christian A Hübner
- Jena University Hospital, Friedrich Schiller Universität Institute of Human Genetics, , 07747 Jena , Germany
| | - Kai Kaila
- University of Helsinki Molecular and Integrative Biosciences, , 00014 Helsinki , Finland
- Helsinki Institute of Life Science, University of Helsinki Neuroscience Center, , 00014 Helsinki , Finland
| | - Mari A Virtanen
- University of Helsinki Molecular and Integrative Biosciences, , 00014 Helsinki , Finland
- Helsinki Institute of Life Science, University of Helsinki Neuroscience Center, , 00014 Helsinki , Finland
| |
Collapse
|
17
|
Salceda R. Glycine neurotransmission: Its role in development. Front Neurosci 2022; 16:947563. [PMID: 36188468 PMCID: PMC9525178 DOI: 10.3389/fnins.2022.947563] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2022] [Accepted: 08/30/2022] [Indexed: 11/13/2022] Open
Abstract
The accurate function of the central nervous system (CNS) depends of the consonance of multiple genetic programs and external signals during the ontogenesis. A variety of molecules including neurotransmitters, have been implied in the regulation of proliferation, survival, and cell-fate of neurons and glial cells. Among these, neurotransmitters may play a central role since functional ligand-gated ionic channel receptors have been described before the establishment of synapses. This review argues on the function of glycine during development, and show evidence indicating it regulates morphogenetic events by means of their transporters and receptors, emphasizing the role of glycinergic activity in the balance of excitatory and inhibitory signals during development. Understanding the mechanisms involved in these processes would help us to know the etiology of cognitive dysfunctions and lead to improve brain repair strategies.
Collapse
|
18
|
Hui KK, Chater TE, Goda Y, Tanaka M. How Staying Negative Is Good for the (Adult) Brain: Maintaining Chloride Homeostasis and the GABA-Shift in Neurological Disorders. Front Mol Neurosci 2022; 15:893111. [PMID: 35875665 PMCID: PMC9305173 DOI: 10.3389/fnmol.2022.893111] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2022] [Accepted: 06/10/2022] [Indexed: 01/27/2023] Open
Abstract
Excitatory-inhibitory (E-I) imbalance has been shown to contribute to the pathogenesis of a wide range of neurodevelopmental disorders including autism spectrum disorders, epilepsy, and schizophrenia. GABA neurotransmission, the principal inhibitory signal in the mature brain, is critically coupled to proper regulation of chloride homeostasis. During brain maturation, changes in the transport of chloride ions across neuronal cell membranes act to gradually change the majority of GABA signaling from excitatory to inhibitory for neuronal activation, and dysregulation of this GABA-shift likely contributes to multiple neurodevelopmental abnormalities that are associated with circuit dysfunction. Whilst traditionally viewed as a phenomenon which occurs during brain development, recent evidence suggests that this GABA-shift may also be involved in neuropsychiatric disorders due to the "dematuration" of affected neurons. In this review, we will discuss the cell signaling and regulatory mechanisms underlying the GABA-shift phenomenon in the context of the latest findings in the field, in particular the role of chloride cotransporters NKCC1 and KCC2, and furthermore how these regulatory processes are altered in neurodevelopmental and neuropsychiatric disorders. We will also explore the interactions between GABAergic interneurons and other cell types in the developing brain that may influence the GABA-shift. Finally, with a greater understanding of how the GABA-shift is altered in pathological conditions, we will briefly outline recent progress on targeting NKCC1 and KCC2 as a therapeutic strategy against neurodevelopmental and neuropsychiatric disorders associated with improper chloride homeostasis and GABA-shift abnormalities.
Collapse
Affiliation(s)
- Kelvin K. Hui
- Department of Developmental and Molecular Biology, Albert Einstein College of Medicine, Bronx, NY, United States
- Institute for Aging Research, Albert Einstein College of Medicine, Bronx, NY, United States
| | - Thomas E. Chater
- Laboratory for Synaptic Plasticity and Connectivity, RIKEN Center for Brain Science, Wako, Japan
| | - Yukiko Goda
- Laboratory for Synaptic Plasticity and Connectivity, RIKEN Center for Brain Science, Wako, Japan
- Synapse Biology Unit, Okinawa Institute for Science and Technology Graduate University, Onna, Japan
| | - Motomasa Tanaka
- Laboratory for Protein Conformation Diseases, RIKEN Center for Brain Science, Wako, Japan
| |
Collapse
|
19
|
Structural basis for inhibition of the Cation-chloride cotransporter NKCC1 by the diuretic drug bumetanide. Nat Commun 2022; 13:2747. [PMID: 35585053 PMCID: PMC9117670 DOI: 10.1038/s41467-022-30407-3] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2021] [Accepted: 04/28/2022] [Indexed: 11/08/2022] Open
Abstract
Cation-chloride cotransporters (CCCs) NKCC1 and NKCC2 catalyze electroneutral symport of 1 Na+, 1 K+, and 2 Cl− across cell membranes. NKCC1 mediates trans-epithelial Cl− secretion and regulates excitability of some neurons and NKCC2 is critical to renal salt reabsorption. Both transporters are inhibited by the so-called loop diuretics including bumetanide, and these drugs are a mainstay for treating edema and hypertension. Here, our single-particle electron cryo-microscopy structures supported by functional studies reveal an outward-facing conformation of NKCC1, showing bumetanide wedged into a pocket in the extracellular ion translocation pathway. Based on these and the previously published inward-facing structures, we define the translocation pathway and the conformational changes necessary for ion translocation. We also identify an NKCC1 dimer with separated transmembrane domains and extensive transmembrane and C-terminal domain interactions. We further define an N-terminal phosphoregulatory domain that interacts with the C-terminal domain, suggesting a mechanism whereby (de)phosphorylation regulates NKCC1 by tuning the strength of this domain association. Loop diuretics including bumetanide inhibit Na+-K+-Cl−-cotransporters (NKCCs) and are used for the treatment of edema and hypertension. Here, Zhao et. al. report structures of NKCC1 with bumetanide bound, revealing its mechanism of action that would facilitate design of novel diuretics.
Collapse
|
20
|
Branchereau P, Cattaert D. Chloride Homeostasis in Developing Motoneurons. ADVANCES IN NEUROBIOLOGY 2022; 28:45-61. [PMID: 36066820 DOI: 10.1007/978-3-031-07167-6_2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/15/2023]
Abstract
Maturation of GABA/Glycine chloride-mediated synaptic inhibitions is crucial for the establishment of a balance between excitation and inhibition. GABA and glycine are excitatory neurotransmitters on immature neurons that exhibit elevated [Cl-]i. Later in development [Cl-]i drops leading to the occurrence of inhibitory synaptic activity. This ontogenic change is closely correlated to a differential expression of two cation-chloride cotransporters that are the Cl- channel K+/Cl- co-transporter type 2 (KCC2) that extrudes Cl- ions and the Na+-K+-2Cl- cotransporter NKCC1 that accumulates Cl- ions. The classical scheme built from studies performed on cortical and hippocampal networks proposes that immature neurons display high [Cl-]i because NKCC1 is overexpressed compared to KCC2 and that the co-transporters ratio reverses in mature neurons, lowering [Cl-]i. In this chapter, we will see that this classical scheme is not true in motoneurons (MNs) and that an early alteration of the chloride homeostasis may be involved in pathological conditions.
Collapse
Affiliation(s)
- Pascal Branchereau
- Institut de Neurosciences Cognitives et Intégratives d'Aquitaine (INCIA), Univ. Bordeaux, UMR 5287, CNRS, Bordeaux, France.
| | - Daniel Cattaert
- Institut de Neurosciences Cognitives et Intégratives d'Aquitaine (INCIA), Univ. Bordeaux, UMR 5287, CNRS, Bordeaux, France
| |
Collapse
|
21
|
Verma V, Kumar MJV, Sharma K, Rajaram S, Muddashetty R, Manjithaya R, Behnisch T, Clement JP. Pharmacological intervention in young adolescents rescues synaptic physiology and behavioural deficits in Syngap1 +/- mice. Exp Brain Res 2021; 240:289-309. [PMID: 34739555 DOI: 10.1007/s00221-021-06254-x] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2021] [Accepted: 10/21/2021] [Indexed: 01/04/2023]
Abstract
Haploinsufficiency in SYNGAP1 is implicated in intellectual disability (ID) and autism spectrum disorder (ASD) and affects the maturation of dendritic spines. The abnormal spine development has been suggested to cause a disbalance of excitatory and inhibitory (E/I) neurotransmission at distinct developmental periods. In addition, E/I imbalances in Syngap1+/- mice might be due to abnormalities in K+-Cl- co-transporter function (NKCC1, KCC2), in a maner similar to the murine models of Fragile-X and Rett syndromes. To study whether an altered intracellular chloride ion concentration represents an underlying mechanism of modified function of GABAergic synapses in Dentate Gyrus Granule Cells of Syngap1+/- recordings were performed at different developmental stages of the mice. We observed depolarised neurons at P14-15 as illustrated by decreased Cl- reversal potential in Syngap1+/- mice. The KCC2 expression was decreased compared to Wild-type (WT) mice at P14-15. The GSK-3β inhibitor, 6-bromoindirubin-3'-oxime (6BIO) that crosses the blood-brain barrier, was tested to restore the function of GABAergic synapses. We discovered that the intraperitoneal administration of 6BIO during the critical period or young adolescents [P30 to P80 (4-week to 10-week)] normalised an altered E/I balance, the deficits of synaptic plasticity, and behavioural performance like social novelty, anxiety, and memory of the Syngap1+/- mice. In summary, altered GABAergic function in Syngap1+/- mice is due to reduced KCC2 expression leading to an increase in the intracellular chloride concentration that can be counteracted by the 6BIO, which restored cognitive, emotional, and social symptoms by pharmacological intervention, particularly in adulthood.
Collapse
Affiliation(s)
- Vijaya Verma
- Neuroscience Unit, Jawaharlal Nehru Centre for Advanced Scientific Research, Bangalore, 560064, India
| | - M J Vijay Kumar
- Neuroscience Unit, Jawaharlal Nehru Centre for Advanced Scientific Research, Bangalore, 560064, India
| | - Kavita Sharma
- International Centre for Material Sciences, Jawaharlal Nehru Centre for Advanced Scientific Research, Bangalore, 560064, India
| | - Sridhar Rajaram
- International Centre for Material Sciences, Jawaharlal Nehru Centre for Advanced Scientific Research, Bangalore, 560064, India
| | - Ravi Muddashetty
- Institute for Stem Cell Biology and Regenerative Medicine, Bangalore, 560065, India
| | - Ravi Manjithaya
- Neuroscience Unit, Jawaharlal Nehru Centre for Advanced Scientific Research, Bangalore, 560064, India.,Molecular Biology and Genetics Unit, Jawaharlal Nehru Centre for Advanced Scientific Research, Bangalore, 560064, India
| | - Thomas Behnisch
- Institutes of Brain Sciences, Fudan University, Shanghai, 200032, China
| | - James P Clement
- Neuroscience Unit, Jawaharlal Nehru Centre for Advanced Scientific Research, Bangalore, 560064, India.
| |
Collapse
|
22
|
Tonotopic Specializations in Number, Size, and Reversal Potential of GABAergic Inputs Fine-Tune Temporal Coding at Avian Cochlear Nucleus. J Neurosci 2021; 41:8904-8916. [PMID: 34518306 DOI: 10.1523/jneurosci.0884-21.2021] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2021] [Revised: 08/31/2021] [Accepted: 09/04/2021] [Indexed: 11/21/2022] Open
Abstract
GABAergic inhibition in neurons plays a critical role in determining the output of neural circuits. Neurons in avian nucleus magnocellularis (NM) use several tonotopic-region-dependent specializations to relay the timing information of sound in the auditory nerve to higher auditory nuclei. Previously, we showed that feedforward GABAergic inhibition in NM has a different dependence on the level of auditory nerve activity, with the low-frequency region having a low-threshold and linear relationship, while the high-frequency region has a high-threshold and step-like relationship. However, it remains unclear how the GABAergic synapses are tonotopically regulated and interact with other specializations of NM neurons. In this study, we examined GABAergic transmission in the NM of chickens of both sexes and explored its contributions to the temporal coding of sound at each tonotopic region. We found that the number and size of unitary GABAergic currents and their reversal potential were finely tuned at each tonotopic region in the NM. At the lower-frequency region, unitary GABAergic currents were larger in number but smaller in size. In addition, their reversal potential was close to the resting potential of neurons, which enabled reliable inhibition despite the smaller potassium conductance. At the higher-frequency region, on the other hand, unitary GABAergic currents were fewer, larger, and highly depolarizing, which enabled powerful inhibition via activating the large potassium conductance. Thus, we propose that GABAergic synapses are coordinated with the characteristics of excitatory synapses and postsynaptic neurons, ensuring the temporal coding for wide frequency and intensity ranges.SIGNIFICANCE STATEMENT We found in avian cochlear nucleus that the number and size of unitary GABAergic inputs differed among tonotopic regions and correlated to respective excitatory inputs; it was larger in number but smaller in size for neurons tuned to lower-frequency sound. Furthermore, GABAergic reversal potential also differed among the regions in accordance with the size of Kv1 current; it was less depolarized in the lower-frequency neurons with smaller Kv1 current. These differentiations of GABAergic transmission maximized the effects of inhibition at each tonotopic region, ensuring precise and reliable temporal coding across frequencies and intensities. Our results emphasize the importance of optimizing characteristics of GABAergic transmission within individual neurons for proper neural circuit function.
Collapse
|
23
|
Jin X, Kim WB, Kim MN, Jung WW, Kang HK, Hong EH, Kim YS, Shim WJ, Han HC, Colwell CS, Kim YB, Kim YI. Oestrogen inhibits salt-dependent hypertension by suppressing GABAergic excitation in magnocellular AVP neurons. Cardiovasc Res 2021; 117:2263-2274. [PMID: 32960965 PMCID: PMC10616626 DOI: 10.1093/cvr/cvaa271] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/10/2020] [Revised: 06/24/2020] [Accepted: 09/08/2020] [Indexed: 12/25/2022] Open
Abstract
AIMS Abundant evidence indicates that oestrogen (E2) plays a protective role against hypertension. Yet, the mechanism underlying the antihypertensive effect of E2 is poorly understood. In this study, we sought to determine the mechanism through which E2 inhibits salt-dependent hypertension. METHODS AND RESULTS To this end, we performed a series of in vivo and in vitro experiments employing a rat model of hypertension that is produced by deoxycorticosterone acetate (DOCA)-salt treatment after uninephrectomy. We found that E2 prevented DOCA-salt treatment from inducing hypertension, raising plasma arginine-vasopressin (AVP) level, enhancing the depressor effect of the V1a receptor antagonist (Phenylac1,D-Tyr(Et)2,Lys6,Arg8,des-Gly9)-vasopressin, and converting GABAergic inhibition to excitation in hypothalamic magnocellular AVP neurons. Moreover, we obtained results indicating that the E2 modulation of the activity and/or expression of NKCC1 (Cl- importer) and KCC2 (Cl- extruder) underpins the effect of E2 on the transition of GABAergic transmission in AVP neurons. Lastly, we discovered that, in DOCA-salt-treated hypertensive ovariectomized rats, CLP290 (prodrug of the KCC2 activator CLP257, intraperitoneal injections) lowered blood pressure, and plasma AVP level and hyperpolarized GABA equilibrium potential to prevent GABAergic excitation from emerging in the AVP neurons of these animals. CONCLUSION Based on these results, we conclude that E2 inhibits salt-dependent hypertension by suppressing GABAergic excitation to decrease the hormonal output of AVP neurons.
Collapse
Affiliation(s)
- Xiangyan Jin
- Department of Physiology and Neuroscience Research Institute, Korea University College of Medicine, 126-1 Anam-dong 5-ga, Seoul 136-705, Republic of Korea
| | - Woong Bin Kim
- Department of Physiology and Neuroscience Research Institute, Korea University College of Medicine, 126-1 Anam-dong 5-ga, Seoul 136-705, Republic of Korea
| | - Mi-Na Kim
- Department of Internal Medicine, Cardiovascular Section, Korea University Anam Hospital, Seoul 136-705, Republic of Korea
| | - Won Woo Jung
- Department of Physiology and Neuroscience Research Institute, Korea University College of Medicine, 126-1 Anam-dong 5-ga, Seoul 136-705, Republic of Korea
| | - Hyung Kyung Kang
- Department of Physiology and Neuroscience Research Institute, Korea University College of Medicine, 126-1 Anam-dong 5-ga, Seoul 136-705, Republic of Korea
| | - Eun-Hwa Hong
- Department of Physiology and Neuroscience Research Institute, Korea University College of Medicine, 126-1 Anam-dong 5-ga, Seoul 136-705, Republic of Korea
| | - Yoon Sik Kim
- Department of Physiology and Neuroscience Research Institute, Korea University College of Medicine, 126-1 Anam-dong 5-ga, Seoul 136-705, Republic of Korea
| | - Wan Joo Shim
- Department of Internal Medicine, Cardiovascular Section, Korea University Anam Hospital, Seoul 136-705, Republic of Korea
| | - Hee Chul Han
- Department of Physiology and Neuroscience Research Institute, Korea University College of Medicine, 126-1 Anam-dong 5-ga, Seoul 136-705, Republic of Korea
| | - Christopher S Colwell
- Department of Psychiatry and Biobehavioral Sciences, University of California, Los Angeles, CA, USA
| | - Young-Beom Kim
- Department of Physiology and Neuroscience Research Institute, Korea University College of Medicine, 126-1 Anam-dong 5-ga, Seoul 136-705, Republic of Korea
| | - Yang In Kim
- Department of Physiology and Neuroscience Research Institute, Korea University College of Medicine, 126-1 Anam-dong 5-ga, Seoul 136-705, Republic of Korea
| |
Collapse
|
24
|
Parrini M, Naskar S, Alberti M, Colombi I, Morelli G, Rocchi A, Nanni M, Piccardi F, Charles S, Ronzitti G, Mingozzi F, Contestabile A, Cancedda L. Restoring neuronal chloride homeostasis with anti-NKCC1 gene therapy rescues cognitive deficits in a mouse model of Down syndrome. Mol Ther 2021; 29:3072-3092. [PMID: 34058387 PMCID: PMC8531145 DOI: 10.1016/j.ymthe.2021.05.023] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2020] [Revised: 04/27/2021] [Accepted: 05/18/2021] [Indexed: 01/24/2023] Open
Abstract
A common feature of diverse brain disorders is the alteration of GABA-mediated inhibition because of aberrant, intracellular chloride homeostasis induced by changes in the expression and/or function of chloride transporters. Notably, pharmacological inhibition of the chloride importer NKCC1 is able to rescue brain-related core deficits in animal models of these pathologies and in some human clinical studies. Here, we show that reducing NKCC1 expression by RNA interference in the Ts65Dn mouse model of Down syndrome (DS) restores intracellular chloride concentration, efficacy of gamma-aminobutyric acid (GABA)-mediated inhibition, and neuronal network dynamics in vitro and ex vivo. Importantly, adeno-associated virus (AAV)-mediated, neuron-specific NKCC1 knockdown in vivo rescues cognitive deficits in diverse behavioral tasks in Ts65Dn animals. Our results highlight a mechanistic link between NKCC1 expression and behavioral abnormalities in DS mice and establish a molecular target for new therapeutic approaches, including gene therapy, to treat brain disorders characterized by neuronal chloride imbalance.
Collapse
Affiliation(s)
- Martina Parrini
- Brain Development and Disease Laboratory, Istituto Italiano di Tecnologia, 16163 Genoa, Italy
| | - Shovan Naskar
- Brain Development and Disease Laboratory, Istituto Italiano di Tecnologia, 16163 Genoa, Italy
| | - Micol Alberti
- Brain Development and Disease Laboratory, Istituto Italiano di Tecnologia, 16163 Genoa, Italy
| | - Ilaria Colombi
- Brain Development and Disease Laboratory, Istituto Italiano di Tecnologia, 16163 Genoa, Italy
| | - Giovanni Morelli
- Brain Development and Disease Laboratory, Istituto Italiano di Tecnologia, 16163 Genoa, Italy
| | - Anna Rocchi
- Center for Synaptic Neuroscience and Technology, Istituto Italiano di Tecnologia, 16132 Genoa, Italy; IRCSS Ospedale Policlinico San Martino, 16132 Genoa, Italy
| | - Marina Nanni
- Brain Development and Disease Laboratory, Istituto Italiano di Tecnologia, 16163 Genoa, Italy
| | - Federica Piccardi
- Animal Facility, Istituto Italiano di Tecnologia, 16163 Genoa, Italy
| | - Severine Charles
- Genethon, 91000 Evry, France; Paris-Saclay University, University Evry, Inserm, Integrare research unit UMR_S951, 91000 Evry, France
| | - Giuseppe Ronzitti
- Genethon, 91000 Evry, France; Paris-Saclay University, University Evry, Inserm, Integrare research unit UMR_S951, 91000 Evry, France
| | - Federico Mingozzi
- Genethon, 91000 Evry, France; Paris-Saclay University, University Evry, Inserm, Integrare research unit UMR_S951, 91000 Evry, France
| | - Andrea Contestabile
- Brain Development and Disease Laboratory, Istituto Italiano di Tecnologia, 16163 Genoa, Italy.
| | - Laura Cancedda
- Brain Development and Disease Laboratory, Istituto Italiano di Tecnologia, 16163 Genoa, Italy; Dulbecco Telethon Institute, 00185 Rome, Italy.
| |
Collapse
|
25
|
Kladisios N, Fischer L, Felmy F. Minimal Number of Required Inputs for Temporally Precise Action Potential Generation in Auditory Brainstem Nuclei. Front Cell Neurosci 2020; 14:592213. [PMID: 33250717 PMCID: PMC7674839 DOI: 10.3389/fncel.2020.592213] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2020] [Accepted: 10/07/2020] [Indexed: 11/18/2022] Open
Abstract
The auditory system relies on temporal precise information transfer, requiring an interplay of synchronously activated inputs and rapid postsynaptic integration. During late postnatal development synaptic, biophysical, and morphological features change to enable mature auditory neurons to perform their appropriate function. How the number of minimal required input fibers and the relevant EPSC time course integrated for action potential generation changes during late postnatal development is unclear. To answer these questions, we used in vitro electrophysiology in auditory brainstem structures from pre-hearing onset and mature Mongolian gerbils of either sex. Synaptic and biophysical parameters changed distinctively during development in the medial nucleus of the trapezoid body (MNTB), the medial superior olive (MSO), and the ventral and dorsal nucleus of the lateral lemniscus (VNLL and DNLL). Despite a reduction in input resistance in most cell types, all required fewer inputs in the mature stage to drive action potentials. Moreover, the EPSC decay time constant is a good predictor of the EPSC time used for action potential generation in all nuclei but the VNLL. Only in MSO neurons, the full EPSC time course is integrated by the neuron’s resistive element, while otherwise, the relevant EPSC time matches only 5–10% of the membrane time constant, indicating membrane charging as a dominant role for output generation. We conclude, that distinct developmental programs lead to a general increase in temporal precision and integration accuracy matched to the information relaying properties of the investigated nuclei.
Collapse
Affiliation(s)
- Nikolaos Kladisios
- Institute of Zoology, University of Veterinary Medicine, Hannover, Germany
| | - Linda Fischer
- Institute of Zoology, University of Veterinary Medicine, Hannover, Germany
| | - Felix Felmy
- Institute of Zoology, University of Veterinary Medicine, Hannover, Germany
| |
Collapse
|
26
|
Wirth MJ, Ackels T, Kriebel A, Kriebel K, Mey J, Kuenzel T, Wagner H. Expression patterns of chloride transporters in the auditory brainstem of developing chicken. Hear Res 2020; 393:108013. [PMID: 32554128 DOI: 10.1016/j.heares.2020.108013] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/05/2019] [Revised: 04/29/2020] [Accepted: 05/29/2020] [Indexed: 01/23/2023]
Abstract
GABAergic transmission changes from depolarization to hyperpolarization in most vertebrate brain regions during development. By contrast, in the auditory brainstem of chicken a depolarizing effect of GABA persists after hatching. Since auditory brainstem neurons that receive GABAergic input have a Cl- reversal potential above resting membrane potential, a specifically tuned activity of Cl- transporters is likely. We here present a developmental study of the expression patterns of several members of the SLC12 family (NKCC1, NKCC2, KCC1, KCC2, KCC4, CCC6, CCC9) and of AE3 at developmental ages E7, E10, E12, E15, E17, and P1 with quantitative RT-PCR. NKCC2 and CCC9 were not detected in auditory brainstem (positive control: kidney). KCC1, CCC6 and AE3 were expressed, but not regulated, while NKCC1, KCC2 and KCC4 were regulated. The expression of the latter transporters increased, with KCC2 exhibiting the strongest expression at all time points. Biochemical analysis of the protein expression of NKCC1, KCC2 and KCC4 corroborated the findings on the mRNA level. All three transporters showed a localization at the outer rim of the cells, with NKCC1 and KCC2 expressed in neurons, and KCC4 predominantly in glia. The comparison of the published chloride reversal potential and expression of transporter proteins suggest strong differences in the efficiency of the three transporters. Further, the strong KCC2 expression could reflect a role in the structural development of auditory brainstem synapses that might lead to changes in the physiological properties.
Collapse
Affiliation(s)
- Marcus J Wirth
- Department for Chemosensation, RWTH Aachen University, Aachen, Germany.
| | - Tobias Ackels
- Neurophysiology of Behaviour Lab, The Francis Crick Institute, London, United Kingdom; Department of Neuroscience, Physiology and Pharmacology, University College London, London, United Kingdom
| | - Andreas Kriebel
- Department for Zoology and Animal Physiology, RWTH Aachen University, Aachen, Germany
| | - Katharina Kriebel
- Department for Zoology and Animal Physiology, RWTH Aachen University, Aachen, Germany
| | - Jörg Mey
- Department for Chemosensation, RWTH Aachen University, Aachen, Germany; Hospital Nacional de Parapléjicos, Toledo, Spain
| | - Thomas Kuenzel
- Department for Chemosensation, RWTH Aachen University, Aachen, Germany
| | - Hermann Wagner
- Department for Zoology and Animal Physiology, RWTH Aachen University, Aachen, Germany
| |
Collapse
|
27
|
Zhang J, Cordshagen A, Medina I, Nothwang HG, Wisniewski JR, Winklhofer M, Hartmann AM. Staurosporine and NEM mainly impair WNK-SPAK/OSR1 mediated phosphorylation of KCC2 and NKCC1. PLoS One 2020; 15:e0232967. [PMID: 32413057 PMCID: PMC7228128 DOI: 10.1371/journal.pone.0232967] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2019] [Accepted: 04/24/2020] [Indexed: 02/05/2023] Open
Abstract
The pivotal role of KCC2 and NKCC1 in development and maintenance of fast inhibitory neurotransmission and their implication in severe human diseases arouse interest in posttranscriptional regulatory mechanisms such as (de)phosphorylation. Staurosporine (broad kinase inhibitor) and N-ethylmalemide (NEM) that modulate kinase and phosphatase activities enhance KCC2 and decrease NKCC1 activity. Here, we investigated the regulatory mechanism for this reciprocal regulation by mass spectrometry and immunoblot analyses using phospho-specific antibodies. Our analyses revealed that application of staurosporine or NEM dephosphorylates Thr1007 of KCC2, and Thr203, Thr207 and Thr212 of NKCC1. Dephosphorylation of Thr1007 of KCC2, and Thr207 and Thr212 of NKCC1 were previously demonstrated to activate KCC2 and to inactivate NKCC1. In addition, application of the two agents resulted in dephosphorylation of the T-loop and S-loop phosphorylation sites Thr233 and Ser373 of SPAK, a critical kinase in the WNK-SPAK/OSR1 signaling module mediating phosphorylation of KCC2 and NKCC1. Taken together, these results suggest that reciprocal regulation of KCC2 and NKCC1 via staurosporine and NEM is based on WNK-SPAK/OSR1 signaling. The key regulatory phospho-site Ser940 of KCC2 is not critically involved in the enhanced activation of KCC2 upon staurosporine and NEM treatment, as both agents have opposite effects on its phosphorylation status. Finally, NEM acts in a tissue-specific manner on Ser940, as shown by comparative analysis in HEK293 cells and immature cultured hippocampal neurons. In summary, our analyses identified phospho-sites that are responsive to staurosporine or NEM application. This provides important information towards a better understanding of the cooperative interactions of different phospho-sites.
Collapse
Affiliation(s)
- Jinwei Zhang
- Hatherly Laboratories, Medical School, College of Medicine and Health, Institute of Biomedical and Clinical Sciences, University of Exeter, Exeter, United Kingdom
- Xiamen Cardiovascular Hospital, School of Medicine, Xiamen University, Xiamen, China
| | - Antje Cordshagen
- Division of Neurogenetics, School of Medicine and Health Sciences, Carl von Ossietzky University Oldenburg, Oldenburg, Germany
| | - Igor Medina
- INSERM (Institut National de la Santé et de la Recherche Médicale) Unité 1249, INMED (Institut de Neurobiologie de la Méditerranée), Aix-Marseille University UMR 1249, Marseille, France
| | - Hans Gerd Nothwang
- Division of Neurogenetics, School of Medicine and Health Sciences, Carl von Ossietzky University Oldenburg, Oldenburg, Germany
- Research Center for Neurosensory Sciences, Carl von Ossietzky University Oldenburg, Oldenburg, Germany
- Center of Excellence Hearing4all, Carl von Ossietzky University Oldenburg, Oldenburg, Germany
| | - Jacek R. Wisniewski
- Department of Proteomics and Signal Transduction, Biochemical Proteomics Group, Max-Planck-Institute of Biochemistry, Martinsried, Germany
| | - Michael Winklhofer
- Research Center for Neurosensory Sciences, Carl von Ossietzky University Oldenburg, Oldenburg, Germany
- Institute for Biology and Environmental Sciences IBU, Carl von Ossietzky University of Oldenburg, Oldenburg, Germany
| | - Anna-Maria Hartmann
- Division of Neurogenetics, School of Medicine and Health Sciences, Carl von Ossietzky University Oldenburg, Oldenburg, Germany
- Research Center for Neurosensory Sciences, Carl von Ossietzky University Oldenburg, Oldenburg, Germany
| |
Collapse
|
28
|
Pressey JC, Woodin MA. Kainate receptor regulation of synaptic inhibition in the hippocampus. J Physiol 2020; 599:485-492. [PMID: 32162694 DOI: 10.1113/jp279645] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2020] [Accepted: 03/02/2020] [Indexed: 12/13/2022] Open
Abstract
Kainate receptors (KARs) are glutamate-type receptors that mediate both canonical ionotropic currents and non-canonical metabotropic signalling. While KARs are expressed widely throughout the brain, synaptic KAR currents have only been recorded at a limited set of synapses, and the KAR currents that have been recorded are relatively small and slow, which has led to the question, what is the functional significance of KARs? While the KAR current itself is relatively modest, its impact on inhibition in the hippocampus can be profound. In the CA1 region of the hippocampus, presynaptic KAR activation bidirectionally regulates γ-aminobutyric acid (GABA) release in a manner that depends on the glutamate concentration; lower levels of glutamate facilitate GABA release via an ionotropic pathway, while higher levels of glutamate depress GABA release via a metabotropic pathway. Postsynaptic interneuron KAR activation increases spike frequency through an ionotropic current, which in turn can strengthen inhibition. In the CA3 region, postsynaptic KAR activation in pyramidal neurons also strengthens inhibition, but in this case through a metabotropic pathway which regulates the neuronal chloride gradient and hyperpolarizes the reversal potential for GABA (EGABA ). Taken together, the evidence for KAR-mediated regulation of the strength of inhibition via pre- and postsynaptic mechanisms provides compelling evidence that KARs are ideally positioned to regulate excitation-inhibition balance - through sensing the excitatory tone and concomitantly tuning the strength of inhibition.
Collapse
Affiliation(s)
- Jessica C Pressey
- Department of Cell and Systems Biology, University of Toronto, Toronto, Ontario, Canada
| | - Melanie A Woodin
- Department of Cell and Systems Biology, University of Toronto, Toronto, Ontario, Canada
| |
Collapse
|
29
|
Reid MS, Kern DM, Brohawn SG. Cryo-EM structure of the potassium-chloride cotransporter KCC4 in lipid nanodiscs. eLife 2020; 9:e52505. [PMID: 32286222 PMCID: PMC7200160 DOI: 10.7554/elife.52505] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2019] [Accepted: 04/12/2020] [Indexed: 01/24/2023] Open
Abstract
Cation-chloride-cotransporters (CCCs) catalyze transport of Cl- with K+ and/or Na+across cellular membranes. CCCs play roles in cellular volume regulation, neural development and function, audition, regulation of blood pressure, and renal function. CCCs are targets of clinically important drugs including loop diuretics and their disruption has been implicated in pathophysiology including epilepsy, hearing loss, and the genetic disorders Andermann, Gitelman, and Bartter syndromes. Here we present the structure of a CCC, the Mus musculus K+-Cl- cotransporter (KCC) KCC4, in lipid nanodiscs determined by cryo-EM. The structure, captured in an inside-open conformation, reveals the architecture of KCCs including an extracellular domain poised to regulate transport activity through an outer gate. We identify binding sites for substrate K+ and Cl- ions, demonstrate the importance of key coordinating residues for transporter activity, and provide a structural explanation for varied substrate specificity and ion transport ratio among CCCs. These results provide mechanistic insight into the function and regulation of a physiologically important transporter family.
Collapse
Affiliation(s)
- Michelle S Reid
- Department of Molecular and Cell Biology, University of California BerkeleyBerkeleyUnited States
- Helen Wills Neuroscience Institute, University of California BerkeleyBerkeleyUnited States
| | - David M Kern
- Department of Molecular and Cell Biology, University of California BerkeleyBerkeleyUnited States
- Helen Wills Neuroscience Institute, University of California BerkeleyBerkeleyUnited States
| | - Stephen Graf Brohawn
- Department of Molecular and Cell Biology, University of California BerkeleyBerkeleyUnited States
- Helen Wills Neuroscience Institute, University of California BerkeleyBerkeleyUnited States
| |
Collapse
|
30
|
Venugopalan P, Cameron EG, Zhang X, Nahmou M, Muller KJ, Goldberg JL. Physiologic maturation is both extrinsically and intrinsically regulated in progenitor-derived neurons. Sci Rep 2020; 10:2337. [PMID: 32047174 PMCID: PMC7012889 DOI: 10.1038/s41598-020-58120-5] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2016] [Accepted: 01/03/2020] [Indexed: 12/15/2022] Open
Abstract
During development, newly-differentiated neurons undergo several morphological and physiological changes to become functional, mature neurons. Physiologic maturation of neuronal cells derived from isolated stem or progenitor cells may provide insight into maturation in vivo but is not well studied. As a step towards understanding how neuronal maturation is regulated, we studied the developmental switch of response to the neurotransmitter GABA, from excitatory depolarization to inhibitory hyperpolarization. We compared acutely isolated retinal ganglion cells (RGCs) at various developmental stages and RGCs differentiated in vitro from embryonic retinal progenitors for the effects of aging and, independently, of retinal environment age on their GABAA receptor (GABAAR) responses, elicited by muscimol. We found that neurons generated in vitro from progenitors exhibited depolarizing, immature GABA responses, like those of early postnatal RGCs. As progenitor-derived neurons aged from 1 to 3 weeks, their GABA responses matured. Interestingly, signals secreted by the early postnatal retina suppressed acquisition of mature GABA responses. This suppression was not associated with changes in expression of GABAAR or of the chloride co-transporter KCC2, but rather with inhibition of KCC2 dimerization in differentiating neurons. Taken together, these data indicate GABA response maturation depends on release of inhibition by developmentally regulated diffusible signals from the retina.
Collapse
Affiliation(s)
- Praseeda Venugopalan
- Neuroscience Program, University of Miami, Miami, FL, 33136, USA
- Shiley Eye Institute, University of California, San Diego, CA, 92093, USA
| | - Evan G Cameron
- Shiley Eye Institute, University of California, San Diego, CA, 92093, USA
- Byers Eye Institute, Stanford University, Stanford, CA, 94303, USA
| | - Xiong Zhang
- Shiley Eye Institute, University of California, San Diego, CA, 92093, USA
| | - Michael Nahmou
- Byers Eye Institute, Stanford University, Stanford, CA, 94303, USA
| | - Kenneth J Muller
- Neuroscience Program, University of Miami, Miami, FL, 33136, USA.
- Department of Physiology & Biophysics, University of Miami Miller School of Medicine, Miami, FL, 33136, USA.
| | - Jeffrey L Goldberg
- Neuroscience Program, University of Miami, Miami, FL, 33136, USA.
- Shiley Eye Institute, University of California, San Diego, CA, 92093, USA.
- Byers Eye Institute, Stanford University, Stanford, CA, 94303, USA.
| |
Collapse
|
31
|
Delpire E, Guo J. Cryo-EM structures of DrNKCC1 and hKCC1: a new milestone in the physiology of cation-chloride cotransporters. Am J Physiol Cell Physiol 2020; 318:C225-C237. [PMID: 31747317 PMCID: PMC7052613 DOI: 10.1152/ajpcell.00465.2019] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2019] [Revised: 11/20/2019] [Accepted: 11/20/2019] [Indexed: 01/29/2023]
Abstract
New milestones have been reached in the field of cation-Cl- cotransporters with the recently released cryo-electron microscopy (EM) structures of the Danio rerio (zebrafish) Na+-K+-2Cl- cotransporter (DrNKCC1) and the human K+-Cl- cotransporter (hKCC1). In this review we provide a brief timeline that identifies the multiple breakthroughs in the field of solute carrier 12 transporters that led to the structure resolution of two of its key members. While cation-Cl- cotransporters share the overall architecture of carriers belonging to the amino acid-polyamine-organocation (APC) superfamily and some of their substrate binding sites, several new insights are gained from the two individual structures. A first major feature relates to the largest extracellular domain between transmembrane domain (TMD) 5 and TMD6 of KCC1, which stabilizes the dimer and forms a cap that likely participates in extracellular gating. A second feature is the conservation of the K+ and Cl- binding sites in both structures and evidence of an unexpected second Cl- coordination site in the KCC1 structure. Structural data are discussed in the context of previously published studies that examined the basic and kinetics properties of these cotransport mechanisms. A third characteristic is the evidence of an extracellular gate formed by conserved salt bridges between charged residues located toward the end of TMD3 and TMD4 in both transporters and the existence of an additional neighboring bridge in the hKCC1 structure. A fourth feature of these newly solved structures relates to the multiple points of contacts between the monomer forming the cotransporter homodimer units. These involve the TMDs, the COOH-terminal domains, and the large extracellular loop for hKCC1.
Collapse
Affiliation(s)
- Eric Delpire
- Department of Anesthesiology, Vanderbilt University School of Medicine, Nashville, Tennessee
| | - Jiangtao Guo
- Department of Biophysics, Department of Pathology of Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China
| |
Collapse
|
32
|
Sullivan BJ, Kadam SD. The involvement of neuronal chloride transporter deficiencies in epilepsy. NEURONAL CHLORIDE TRANSPORTERS IN HEALTH AND DISEASE 2020:329-366. [DOI: 10.1016/b978-0-12-815318-5.00014-5] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/05/2025]
|
33
|
Halbhuber L, Achtner C, Luhmann HJ, Sinning A, Kilb W. Coincident Activation of Glutamate Receptors Enhances GABA A Receptor-Induced Ionic Plasticity of the Intracellular Cl --Concentration in Dissociated Neuronal Cultures. Front Cell Neurosci 2019; 13:497. [PMID: 31787883 PMCID: PMC6856009 DOI: 10.3389/fncel.2019.00497] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2019] [Accepted: 10/21/2019] [Indexed: 01/09/2023] Open
Abstract
Massive activation of γ-amino butyric acid A (GABAA) receptors during pathophysiological activity induces an increase in the intracellular Cl--concentration ([Cl-]i), which is sufficient to render GABAergic responses excitatory. However, to what extent physiological levels of GABAergic activity can influence [Cl-]i is not known. Aim of the present study is to reveal whether moderate activation of GABAA receptors mediates functionally relevant [Cl-]i changes and whether these changes can be augmented by coincident glutamatergic activity. To address these questions, we used whole-cell patch-clamp recordings from cultured cortical neurons [at days in vitro (DIV) 6-22] to determine changes in the GABA reversal potential (EGABA) induced by short bursts of GABAergic and/or synchronized glutamatergic stimulation. These experiments revealed that pressure-application of 10 short muscimol pulses at 10 Hz induced voltage-dependent [Cl-]i changes. Under current-clamp conditions this muscimol burst induced a [Cl-]i increase of 3.1 ± 0.4 mM (n = 27), which was significantly enhanced to 4.6 ± 0.5 mM (n = 27) when glutamate was applied synchronously with the muscimol pulses. The muscimol-induced [Cl-]i increase significantly attenuated the inhibitory effect of GABA, as determined by the GABAergic rheobase shift. The synchronous coapplication of glutamate pulses had no additional effect on the attenuation of GABAergic inhibition, despite the larger [Cl-]i transients under these conditions. In summary, these results indicate that moderate GABAergic activity can induce functionally relevant [Cl-]i transients, which were enhanced by coincident glutamate pulses. This ionic plasticity of [Cl-]i may contribute to short-term plasticity of the GABAergic system.
Collapse
Affiliation(s)
- Lisa Halbhuber
- Institute of Physiology, University Medical Center Mainz, Johannes Gutenberg University, Mainz, Germany
| | - Cécilia Achtner
- Institute of Physiology, University Medical Center Mainz, Johannes Gutenberg University, Mainz, Germany
| | - Heiko J Luhmann
- Institute of Physiology, University Medical Center Mainz, Johannes Gutenberg University, Mainz, Germany
| | - Anne Sinning
- Institute of Physiology, University Medical Center Mainz, Johannes Gutenberg University, Mainz, Germany
| | - Werner Kilb
- Institute of Physiology, University Medical Center Mainz, Johannes Gutenberg University, Mainz, Germany
| |
Collapse
|
34
|
Liu S, Chang S, Han B, Xu L, Zhang M, Zhao C, Yang W, Wang F, Li J, Delpire E, Ye S, Bai XC, Guo J. Cryo-EM structures of the human cation-chloride cotransporter KCC1. Science 2019; 366:505-508. [PMID: 31649201 DOI: 10.1126/science.aay3129] [Citation(s) in RCA: 52] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2019] [Accepted: 09/26/2019] [Indexed: 11/02/2022]
Abstract
Cation-chloride cotransporters (CCCs) mediate the coupled, electroneutral symport of cations with chloride across the plasma membrane and are vital for cell volume regulation, salt reabsorption in the kidney, and γ-aminobutyric acid (GABA)-mediated modulation in neurons. Here we present cryo-electron microscopy (cryo-EM) structures of human potassium-chloride cotransporter KCC1 in potassium chloride or sodium chloride at 2.9- to 3.5-angstrom resolution. KCC1 exists as a dimer, with both extracellular and transmembrane domains involved in dimerization. The structural and functional analyses, along with computational studies, reveal one potassium site and two chloride sites in KCC1, which are all required for the ion transport activity. KCC1 adopts an inward-facing conformation, with the extracellular gate occluded. The KCC1 structures allow us to model a potential ion transport mechanism in KCCs and provide a blueprint for drug design.
Collapse
Affiliation(s)
- Si Liu
- Department of Biophysics, Department of Pathology of Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou 310058, China
- Tianjin Key Laboratory of Function and Application of Biological Macromolecular Structures, School of Life Sciences, Tianjin University, 92 Weijin Road, Nankai District, Tianjin 300072, China
| | - Shenghai Chang
- Department of Biophysics, Department of Pathology of Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou 310058, China
- Center of Cryo-Electron Microscopy, Zhejiang University School of Medicine, Hangzhou 310058, China
| | - Binming Han
- Zhejiang Province Key Laboratory of Quantum Technology and Device, Institute of Quantitative Biology, Department of Physics, Zhejiang University, Hangzhou, Zhejiang 310027, China
| | - Lingyi Xu
- Department of Biophysics, Department of Pathology of Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou 310058, China
- Life Sciences Institute and Innovation Center for Cell Signaling Network, Zhejiang University, Hangzhou, Zhejiang 310058, China
| | - Mingfeng Zhang
- Department of Biophysics, Institute of Neuroscience, NHC and CAMS Key Laboratory of Medical Neurobiology, Zhejiang University School of Medicine, Hangzhou 310058, China
| | - Cheng Zhao
- Department of Biophysics, Department of Pathology of Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou 310058, China
- Department of Biophysics, Institute of Neuroscience, NHC and CAMS Key Laboratory of Medical Neurobiology, Zhejiang University School of Medicine, Hangzhou 310058, China
| | - Wei Yang
- Department of Biophysics, Institute of Neuroscience, NHC and CAMS Key Laboratory of Medical Neurobiology, Zhejiang University School of Medicine, Hangzhou 310058, China
| | - Feng Wang
- Wuxi Biortus Biosciences Co. Ltd., 6 Dongsheng West Road, Jiangyin 214437, China
| | - Jingyuan Li
- Zhejiang Province Key Laboratory of Quantum Technology and Device, Institute of Quantitative Biology, Department of Physics, Zhejiang University, Hangzhou, Zhejiang 310027, China.
| | - Eric Delpire
- Department of Anesthesiology, Vanderbilt University School of Medicine, Nashville, TN 37232, USA.
| | - Sheng Ye
- Tianjin Key Laboratory of Function and Application of Biological Macromolecular Structures, School of Life Sciences, Tianjin University, 92 Weijin Road, Nankai District, Tianjin 300072, China.
- Life Sciences Institute and Innovation Center for Cell Signaling Network, Zhejiang University, Hangzhou, Zhejiang 310058, China
| | - Xiao-Chen Bai
- Departments of Biophysics and Cell Biology, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA.
| | - Jiangtao Guo
- Department of Biophysics, Department of Pathology of Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou 310058, China.
- Department of Biophysics, Institute of Neuroscience, NHC and CAMS Key Laboratory of Medical Neurobiology, Zhejiang University School of Medicine, Hangzhou 310058, China
| |
Collapse
|
35
|
Early Life Inflammation Increases CA1 Pyramidal Neuron Excitability in a Sex and Age Dependent Manner through a Chloride Homeostasis Disruption. J Neurosci 2019; 39:7244-7259. [PMID: 31308096 DOI: 10.1523/jneurosci.2973-18.2019] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2018] [Revised: 05/31/2019] [Accepted: 07/04/2019] [Indexed: 02/07/2023] Open
Abstract
Early life, systemic inflammation causes long-lasting changes in behavior. To unmask possible mechanisms associated with this phenomenon, we asked whether the intrinsic membrane properties in hippocampal neurons were altered as a consequence of early life inflammation. C57BL/6 mice were bred in-house and both male and female pups from multiple litters were injected with lipopolysaccharide (LPS; 100 μg/kg, i.p.) or vehicle at postnatal day (P)14, and kept until adolescence (P35-P45) or adulthood (P60-P70), when brain slices were prepared for whole-cell and perforated-patch recordings from CA1 hippocampal pyramidal neurons. In neurons of adult male mice pretreated with LPS, the number of action potentials elicited by depolarizing current pulses was significantly increased compared with control neurons, concomitant with increased input resistance, and a lower action potential threshold. Although these changes were not associated with changes in relevant sodium channel expression or differences in capacitance or dendritic architecture, they were linked to a mechanism involving intracellular chloride overload, revealed through a depolarized GABA reversal potential and increased expression of the chloride transporter, NKCC1. In contrast, no significant changes were observed in neurons of adult female mice pretreated with LPS, nor in adolescent mice of either sex. These data uncover a potential mechanism involving neonatal inflammation-induced plasticity in chloride homeostasis, which may contribute to early life inflammation-induced behavioral alterations.SIGNIFICANCE STATEMENT Early life inflammation results in long-lasting changes in many aspects of adult physiology. In this paper we reveal that a brief exposure to early life peripheral inflammation with LPS increases excitability in hippocampal neurons in a sex- and age-dependent manner through a chloride homeostasis disruption. As this hyperexcitability was only seen in adult males, and not in adult females or adolescent animals of either sex, it raises the possibility of a hormonal interaction with early life inflammation. Furthermore, as neonatal inflammation is a normal feature of early life in most animals, as well as humans, these findings may be very important for the development of animal models of disease that more appropriately resemble the human condition.
Collapse
|
36
|
Tillman L, Zhang J. Crossing the Chloride Channel: The Current and Potential Therapeutic Value of the Neuronal K +-Cl - Cotransporter KCC2. BIOMED RESEARCH INTERNATIONAL 2019; 2019:8941046. [PMID: 31240228 PMCID: PMC6556333 DOI: 10.1155/2019/8941046] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/08/2019] [Revised: 04/15/2019] [Accepted: 05/06/2019] [Indexed: 02/05/2023]
Abstract
Chloride (Cl-) homeostasis is an essential process involved in neuronal signalling and cell survival. Inadequate regulation of intracellular Cl- interferes with synaptic signalling and is implicated in several neurological diseases. The main inhibitory neurotransmitter of the central nervous system is γ-aminobutyric acid (GABA). GABA hyperpolarises the membrane potential by activating Cl- permeable GABAA receptor channels (GABAAR). This process is reliant on Cl- extruder K+-Cl- cotransporter 2 (KCC2), which generates the neuron's inward, hyperpolarising Cl- gradient. KCC2 is encoded by the fifth member of the solute carrier 12 family (SLC12A5) and has remained a poorly understood component in the development and severity of many neurological diseases for many years. Recent advancements in next-generation sequencing and specific gene targeting, however, have indicated that loss of KCC2 activity is involved in a number of diseases including epilepsy and schizophrenia. It has also been implicated in neuropathic pain following spinal cord injury. Any variant of SLC12A5 that negatively regulates the transporter's expression may, therefore, be implicated in neurological disease. A recent whole exome study has discovered several causative mutations in patients with epilepsy. Here, we discuss the implications of KCC2 in neurological disease and consider the evolving evidence for KCC2's potential as a therapeutic target.
Collapse
Affiliation(s)
- Luke Tillman
- Institute of Biomedical and Clinical Sciences, Medical School, College of Medicine and Health, University of Exeter, Hatherly Laboratories, Exeter EX4 4PS, UK
| | - Jinwei Zhang
- Institute of Biomedical and Clinical Sciences, Medical School, College of Medicine and Health, University of Exeter, Hatherly Laboratories, Exeter EX4 4PS, UK
| |
Collapse
|
37
|
Pathogenic potential of human SLC12A5 variants causing KCC2 dysfunction. Brain Res 2019; 1710:1-7. [DOI: 10.1016/j.brainres.2018.12.025] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2018] [Revised: 11/21/2018] [Accepted: 12/17/2018] [Indexed: 12/29/2022]
|
38
|
Côme E, Marques X, Poncer JC, Lévi S. KCC2 membrane diffusion tunes neuronal chloride homeostasis. Neuropharmacology 2019; 169:107571. [PMID: 30871970 DOI: 10.1016/j.neuropharm.2019.03.014] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2018] [Revised: 02/26/2019] [Accepted: 03/10/2019] [Indexed: 02/05/2023]
Abstract
Neuronal Cl- homeostasis is regulated by the activity of two cation chloride co-transporters (CCCs), the K+-Cl- cotransporter KCC2 and the Na+-K+-Cl- cotransporter NKCC1, which are primarily extruding and importing chloride in neurons, respectively. Several neurological and psychiatric disorders including epilepsy, neuropathic pain, schizophrenia and autism are associated with altered neuronal chloride (Cl-) homeostasis. A current view is that the accumulation of intracellular Cl- in neurons as a result of KCC2 down-regulation and/or NKCC1 up-regulation may weaken inhibitory GABA signaling and thereby promote the development of pathological activities. CCC activity is determined mainly by their level of expression in the plasma membrane. Furthermore, CCCs undergo "diffusion-trapping" in the membrane, a mechanism that is rapidly adjusted by activity-dependent post-translational modifications i.e. phosphorylation/dephosphorylation of key serine and threonine residues. This represents probably the most rapid cellular mechanism for adapting CCC function to changes in neuronal activity. Therefore, interfering with these mechanisms may help restoring Cl- homeostasis and inhibition under pathological conditions. This article is part of the special issue entitled 'Mobility and trafficking of neuronal membrane proteins'.
Collapse
Affiliation(s)
- Etienne Côme
- INSERM UMR-S 1270, 75005, Paris, France; Sorbonne Université, 75005, Paris, France; Institut du Fer à Moulin, 75005, Paris, France
| | - Xavier Marques
- INSERM UMR-S 1270, 75005, Paris, France; Sorbonne Université, 75005, Paris, France; Institut du Fer à Moulin, 75005, Paris, France
| | - Jean Christophe Poncer
- INSERM UMR-S 1270, 75005, Paris, France; Sorbonne Université, 75005, Paris, France; Institut du Fer à Moulin, 75005, Paris, France
| | - Sabine Lévi
- INSERM UMR-S 1270, 75005, Paris, France; Sorbonne Université, 75005, Paris, France; Institut du Fer à Moulin, 75005, Paris, France.
| |
Collapse
|
39
|
Allen LL, Seven YB, Baker TL, Mitchell GS. Cervical spinal contusion alters Na +-K +-2Cl- and K +-Cl- cation-chloride cotransporter expression in phrenic motor neurons. Respir Physiol Neurobiol 2019; 261:15-23. [PMID: 30590202 PMCID: PMC6939623 DOI: 10.1016/j.resp.2018.12.009] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2018] [Revised: 12/21/2018] [Accepted: 12/23/2018] [Indexed: 12/11/2022]
Abstract
Spinal chloride-dependent synaptic inhibition is critical in regulating breathing and requires neuronal chloride gradients established by cation-chloride cotransporters Na+-K+-2Cl- (NKCC1) and K+-Cl- (KCC2). Spinal transection disrupts NKCC1/KCC2 balance, diminishing chloride gradients in neurons below injury, contributing to spasticity and chronic pain. It is not known if similar disruptions in NKCC1/KCC2 balance occur in respiratory motor neurons after incomplete cervical contusion (C2SC). We hypothesized that C2SC disrupts NKCC1/KCC2 balance in phrenic motor neurons. NKCC1 and KCC2 immunoreactivity was assessed in CtB-positive phrenic motor neurons. Five weeks post-C2SC: 1) neither membrane-bound nor cytosolic NKCC1 expression were significantly changed, although the membrane/cytosolic ratio increased, consistent with net chloride influx; and 2) both membrane and cytosolic KCC2 expression increased, although the membrane/cytosolic ratio decreased, consistent with net chloride efflux. Thus, contrary to our original hypothesis, complex shifts in NKCC1/KCC2 balance occur post-C2SC. The functional significance of these changes remains unclear.
Collapse
Affiliation(s)
- Latoya L Allen
- Department of Physical Therapy, University of Florida, Gainesville, FL 32611 USA; Department of Neuroscience, University of Florida, Gainesville, FL 32610 USA; Center for Respiratory Research and Rehabilitation, University of Florida, Gainesville, FL 32611 USA
| | - Yasin B Seven
- Department of Physical Therapy, University of Florida, Gainesville, FL 32611 USA; Center for Respiratory Research and Rehabilitation, University of Florida, Gainesville, FL 32611 USA
| | - Tracy L Baker
- Department of Comparative Biosciences, University of Wisconsin-Madison, Madison, WI 53706 USA
| | - Gordon S Mitchell
- Department of Physical Therapy, University of Florida, Gainesville, FL 32611 USA; Center for Respiratory Research and Rehabilitation, University of Florida, Gainesville, FL 32611 USA.
| |
Collapse
|
40
|
Côme E, Heubl M, Schwartz EJ, Poncer JC, Lévi S. Reciprocal Regulation of KCC2 Trafficking and Synaptic Activity. Front Cell Neurosci 2019; 13:48. [PMID: 30842727 PMCID: PMC6391895 DOI: 10.3389/fncel.2019.00048] [Citation(s) in RCA: 31] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2018] [Accepted: 02/01/2019] [Indexed: 01/05/2023] Open
Abstract
The main inhibitory neurotransmitter receptors in the adult central nervous system (CNS) are type A γ-aminobutyric acid receptors (GABAARs) and glycine receptors (GlyRs). Synaptic responses mediated by GlyR and GABAAR display a hyperpolarizing shift during development. This shift relies mainly on the developmental up-regulation of the K+-Cl- co-transporter KCC2 responsible for the extrusion of Cl-. In mature neurons, altered KCC2 function-mainly through increased endocytosis-leads to the re-emergence of depolarizing GABAergic and glycinergic signaling, which promotes hyperexcitability and pathological activities. Identifying signaling pathways and molecular partners that control KCC2 surface stability thus represents a key step in the development of novel therapeutic strategies. Here, we present our current knowledge on the cellular and molecular mechanisms governing the plasma membrane turnover rate of the transporter under resting conditions and in response to synaptic activity. We also discuss the notion that KCC2 lateral diffusion is one of the first parameters modulating the transporter membrane stability, allowing for rapid adaptation of Cl- transport to changes in neuronal activity.
Collapse
Affiliation(s)
- Etienne Côme
- INSERM UMR-S 1270, Paris, France.,Sorbonne Université, Paris, France.,Institut du Fer à Moulin, Paris, France
| | - Martin Heubl
- INSERM UMR-S 1270, Paris, France.,Sorbonne Université, Paris, France.,Institut du Fer à Moulin, Paris, France
| | - Eric J Schwartz
- INSERM UMR-S 1270, Paris, France.,Sorbonne Université, Paris, France.,Institut du Fer à Moulin, Paris, France
| | - Jean Christophe Poncer
- INSERM UMR-S 1270, Paris, France.,Sorbonne Université, Paris, France.,Institut du Fer à Moulin, Paris, France
| | - Sabine Lévi
- INSERM UMR-S 1270, Paris, France.,Sorbonne Université, Paris, France.,Institut du Fer à Moulin, Paris, France
| |
Collapse
|
41
|
Awad PN, Amegandjin CA, Szczurkowska J, Carriço JN, Fernandes do Nascimento AS, Baho E, Chattopadhyaya B, Cancedda L, Carmant L, Di Cristo G. KCC2 Regulates Dendritic Spine Formation in a Brain-Region Specific and BDNF Dependent Manner. Cereb Cortex 2018; 28:4049-4062. [PMID: 30169756 PMCID: PMC6188549 DOI: 10.1093/cercor/bhy198] [Citation(s) in RCA: 31] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2017] [Revised: 07/17/2018] [Accepted: 07/27/2018] [Indexed: 01/17/2023] Open
Abstract
KCC2 is the major chloride extruder in neurons. The spatiotemporal regulation of KCC2 expression orchestrates the developmental shift towards inhibitory GABAergic drive and the formation of glutamatergic synapses. Whether KCC2's role in synapse formation is similar in different brain regions is unknown. First, we found that KCC2 subcellular localization, but not overall KCC2 expression levels, differed between cortex and hippocampus during the first postnatal week. We performed site-specific in utero electroporation of KCC2 cDNA to target either hippocampal CA1 or somatosensory cortical pyramidal neurons. We found that a premature expression of KCC2 significantly decreased spine density in CA1 neurons, while it had the opposite effect in cortical neurons. These effects were cell autonomous, because single-cell biolistic overexpression of KCC2 in hippocampal and cortical organotypic cultures also induced a reduction and an increase of dendritic spine density, respectively. In addition, we found that the effects of its premature expression on spine density were dependent on BDNF levels. Finally, we showed that the effects of KCC2 on dendritic spine were dependent on its chloride transporter function in the hippocampus, contrary to what was observed in cortex. Altogether, these results demonstrate that KCC2 regulation of dendritic spine development, and its underlying mechanisms, are brain-region specific.
Collapse
Affiliation(s)
- Patricia Nora Awad
- Department of Neurosciences, Université de Montréal, Montréal, Québec, Canada
- CHU Sainte-Justine Research Center, Montréal, Québec, Canada
| | - Clara Akofa Amegandjin
- Department of Neurosciences, Université de Montréal, Montréal, Québec, Canada
- CHU Sainte-Justine Research Center, Montréal, Québec, Canada
| | - Joanna Szczurkowska
- Neuroscience and Brain Technologies, Instituto Italiano di Tecnologia, Genova, Italy
| | | | | | - Elie Baho
- Department of Neurosciences, Université de Montréal, Montréal, Québec, Canada
- CHU Sainte-Justine Research Center, Montréal, Québec, Canada
| | - Bidisha Chattopadhyaya
- Department of Neurosciences, Université de Montréal, Montréal, Québec, Canada
- CHU Sainte-Justine Research Center, Montréal, Québec, Canada
| | - Laura Cancedda
- Neuroscience and Brain Technologies, Instituto Italiano di Tecnologia, Genova, Italy
- Telethon Dulbecco Institute, Italy
| | - Lionel Carmant
- Department of Neurosciences, Université de Montréal, Montréal, Québec, Canada
- CHU Sainte-Justine Research Center, Montréal, Québec, Canada
| | - Graziella Di Cristo
- Department of Neurosciences, Université de Montréal, Montréal, Québec, Canada
- CHU Sainte-Justine Research Center, Montréal, Québec, Canada
| |
Collapse
|
42
|
Schmidt T, Ghaffarian N, Philippot C, Seifert G, Steinhäuser C, Pape HC, Blaesse P. Differential regulation of chloride homeostasis and GABAergic transmission in the thalamus. Sci Rep 2018; 8:13929. [PMID: 30224811 PMCID: PMC6141474 DOI: 10.1038/s41598-018-31762-2] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2018] [Accepted: 08/23/2018] [Indexed: 01/09/2023] Open
Abstract
The thalamus is important for sensory integration with the ventrobasal thalamus (VB) as relay controlled by GABAergic projections from the nucleus reticularis thalami (NRT). Depending on the [Cl-]i primarily set by cation-chloride-cotransporters, GABA is inhibitory or excitatory. There is evidence that VB and NRT differ in terms of GABA action, with classical hyperpolarization in VB due to the expression of the Cl- extruder KCC2 and depolarizing/excitatory GABA action in the NRT, where KCC2 expression is low and Cl- accumulation by the Cl- inward transporter NKCC1 has been postulated. However, data on NKCC1 expression and functional analysis of both transporters are missing. We show that KCC2-mediated Cl- extrusion set the [Cl-]i in VB, while NKCC1 did not contribute substantially to Cl- accumulation and depolarizing GABA action in the NRT. The finding that NKCC1 did not play a major role in NRT neurons is of high relevance for ongoing studies on the therapeutic use of NKCC1 inhibitors trying to compensate for a disease-induced up-regulation of NKCC1 that has been described for various brain regions and disease states like epilepsy and chronic pain. These data suggest that NKCC1 inhibitors might have no major effect on healthy NRT neurons due to limited NKCC1 function.
Collapse
Affiliation(s)
- Tobias Schmidt
- Institute of Physiology I, University Hospital Münster, Münster, Germany
- Department of Anesthesiology, Intensive Care and Pain Medicine, University Hospital Münster, Münster, Germany
| | - Nikoo Ghaffarian
- Institute of Physiology I, University Hospital Münster, Münster, Germany
| | - Camille Philippot
- Institute of Cellular Neurosciences, Medical Faculty, University of Bonn, Bonn, Germany
| | - Gerald Seifert
- Institute of Cellular Neurosciences, Medical Faculty, University of Bonn, Bonn, Germany
| | - Christian Steinhäuser
- Institute of Cellular Neurosciences, Medical Faculty, University of Bonn, Bonn, Germany
| | | | - Peter Blaesse
- Institute of Physiology I, University Hospital Münster, Münster, Germany.
| |
Collapse
|
43
|
Cordshagen A, Busch W, Winklhofer M, Nothwang HG, Hartmann AM. Phosphoregulation of the intracellular termini of K +-Cl - cotransporter 2 (KCC2) enables flexible control of its activity. J Biol Chem 2018; 293:16984-16993. [PMID: 30201606 DOI: 10.1074/jbc.ra118.004349] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2018] [Revised: 09/01/2018] [Indexed: 12/22/2022] Open
Abstract
The pivotal role of K+-Cl- cotransporter 2 (KCC2) in inhibitory neurotransmission and severe human diseases fosters interest in understanding posttranslational regulatory mechanisms such as (de)phosphorylation. Here, the regulatory role of the five bona fide phosphosites Ser31, Thr34, Ser932, Thr999, and Thr1008 was investigated by the use of alanine and aspartate mutants. Tl+-based flux analyses in HEK-293 cells demonstrated increased transport activity for S932D (mimicking phosphorylation) and T1008A (mimicking dephosphorylation), albeit to a different extent. Increased activity was due to changes in intrinsic activity, as it was not caused by increased cell-surface abundance. Substitutions of Ser31, Thr34, or Thr999 had no effect. Additionally, we show that the indirect actions of the known KCC2 activators staurosporine and N-ethylmaleimide (NEM) involved multiple phosphosites. S31D, T34A, S932A/D, T999A, or T1008A/D abrogated staurosporine mediated stimulation, and S31A, T34D, or S932D abolished NEM-mediated stimulation. This demonstrates for the first time differential effects of staurosporine and NEM on KCC2. In addition, the staurosporine-mediated effects involved both KCC2 phosphorylation and dephosphorylation with Ser932 and Thr1008 being bona fide target sites. In summary, our data reveal a complex phosphoregulation of KCC2 that provides the transporter with a toolbox for graded activity and integration of different signaling pathways.
Collapse
Affiliation(s)
- Antje Cordshagen
- From the Neurogenetics group, Center of Excellence Hearing4all, School of Medicine and Health Sciences
| | - Wiebke Busch
- From the Neurogenetics group, Center of Excellence Hearing4all, School of Medicine and Health Sciences
| | - Michael Winklhofer
- Institute for Biology and Environmental Sciences IBU, and.,Research Center for Neurosensory Sciences, Carl von Ossietzky University Oldenburg, 26111 Oldenburg, Germany
| | - Hans Gerd Nothwang
- From the Neurogenetics group, Center of Excellence Hearing4all, School of Medicine and Health Sciences.,Research Center for Neurosensory Sciences, Carl von Ossietzky University Oldenburg, 26111 Oldenburg, Germany
| | - Anna-Maria Hartmann
- From the Neurogenetics group, Center of Excellence Hearing4all, School of Medicine and Health Sciences, .,Research Center for Neurosensory Sciences, Carl von Ossietzky University Oldenburg, 26111 Oldenburg, Germany
| |
Collapse
|
44
|
Coq JO, Delcour M, Ogawa Y, Peyronnet J, Castets F, Turle-Lorenzo N, Montel V, Bodineau L, Cardot P, Brocard C, Liabeuf S, Bastide B, Canu MH, Tsuji M, Cayetanot F. Mild Intrauterine Hypoperfusion Leads to Lumbar and Cortical Hyperexcitability, Spasticity, and Muscle Dysfunctions in Rats: Implications for Prematurity. Front Neurol 2018; 9:423. [PMID: 29973904 PMCID: PMC6020763 DOI: 10.3389/fneur.2018.00423] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2018] [Accepted: 05/22/2018] [Indexed: 12/22/2022] Open
Abstract
Intrauterine ischemia-hypoxia is detrimental to the developing brain and leads to white matter injury (WMI), encephalopathy of prematurity (EP), and often to cerebral palsy (CP), but the related pathophysiological mechanisms remain unclear. In prior studies, we used mild intrauterine hypoperfusion (MIUH) in rats to successfully reproduce the diversity of clinical signs of EP, and some CP symptoms. Briefly, MIUH led to inflammatory processes, diffuse gray and WMI, minor locomotor deficits, musculoskeletal pathologies, neuroanatomical and functional disorganization of the primary somatosensory and motor cortices, delayed sensorimotor reflexes, spontaneous hyperactivity, deficits in sensory information processing, memory and learning impairments. In the present study, we investigated the early and long-lasting mechanisms of pathophysiology that may be responsible for the various symptoms induced by MIUH. We found early hyperreflexia, spasticity and reduced expression of KCC2 (a chloride cotransporter that regulates chloride homeostasis and cell excitability). Adult MIUH rats exhibited changes in muscle contractile properties and phenotype, enduring hyperreflexia and spasticity, as well as hyperexcitability in the sensorimotor cortex. Taken together, these results show that reduced expression of KCC2, lumbar hyperreflexia, spasticity, altered properties of the soleus muscle, as well as cortical hyperexcitability may likely interplay into a self-perpetuating cycle, leading to the emergence, and persistence of neurodevelopmental disorders (NDD) in EP and CP, such as sensorimotor impairments, and probably hyperactivity, attention, and learning disorders.
Collapse
Affiliation(s)
- Jacques-Olivier Coq
- Centre National de la Recherche Scientifique, Institut de Neurosciences de la Timone, UMR 7289, Aix Marseille Université, Marseille, France.,Centre National de la Recherche Scientifique, Neurosciences Intégratives et Adaptatives, UMR 7260, Aix Marseille Université, Marseille, France
| | - Maxime Delcour
- Centre National de la Recherche Scientifique, Neurosciences Intégratives et Adaptatives, UMR 7260, Aix Marseille Université, Marseille, France
| | - Yuko Ogawa
- Department of Regenerative Medicine and Tissue Engineering, National Cerebral and Cardiovascular Center, Suita, Japan
| | - Julie Peyronnet
- Centre National de la Recherche Scientifique, Institut de Neurosciences de la Timone, UMR 7289, Aix Marseille Université, Marseille, France
| | - Francis Castets
- Centre National de la Recherche Scientifique, Institut de Biologie du Développement de Marseille, UMR 7288, Aix-Marseille Université, Marseille, France
| | - Nathalie Turle-Lorenzo
- FR 3512 Fédération 3C, Aix Marseille Université - Centre National de la Recherche Scientifique, Marseille, France
| | - Valérie Montel
- EA 7369 ≪Activité Physique, Muscle et Santé≫ - URePSSS - Unité de Recherche Pluridisciplinaire Sport Santé Société, Université de Lille, Lille, France
| | - Laurence Bodineau
- Institut National de la Santé et de la Recherche Médicale, UMR_S1158 Neurophysiologie Respiratoire Expérimentale et Clinique, Sorbonne Université, Paris, France
| | - Phillipe Cardot
- Institut National de la Santé et de la Recherche Médicale, UMR_S1158 Neurophysiologie Respiratoire Expérimentale et Clinique, Sorbonne Université, Paris, France
| | - Cécile Brocard
- Centre National de la Recherche Scientifique, Institut de Neurosciences de la Timone, UMR 7289, Aix Marseille Université, Marseille, France
| | - Sylvie Liabeuf
- Centre National de la Recherche Scientifique, Institut de Neurosciences de la Timone, UMR 7289, Aix Marseille Université, Marseille, France
| | - Bruno Bastide
- EA 7369 ≪Activité Physique, Muscle et Santé≫ - URePSSS - Unité de Recherche Pluridisciplinaire Sport Santé Société, Université de Lille, Lille, France
| | - Marie-Hélène Canu
- EA 7369 ≪Activité Physique, Muscle et Santé≫ - URePSSS - Unité de Recherche Pluridisciplinaire Sport Santé Société, Université de Lille, Lille, France
| | - Masahiro Tsuji
- Department of Regenerative Medicine and Tissue Engineering, National Cerebral and Cardiovascular Center, Suita, Japan
| | - Florence Cayetanot
- Centre National de la Recherche Scientifique, Institut de Neurosciences de la Timone, UMR 7289, Aix Marseille Université, Marseille, France.,Institut National de la Santé et de la Recherche Médicale, UMR_S1158 Neurophysiologie Respiratoire Expérimentale et Clinique, Sorbonne Université, Paris, France
| |
Collapse
|
45
|
Dai S, Qi Y, Fu J, Li N, Zhang X, Zhang J, Zhang W, Xu H, Zhou H, Ma Z. Dexmedetomidine attenuates persistent postsurgical pain by upregulating K +-Cl - cotransporter-2 in the spinal dorsal horn in rats. J Pain Res 2018; 11:993-1004. [PMID: 29872336 PMCID: PMC5973459 DOI: 10.2147/jpr.s158737] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022] Open
Abstract
Background Dexmedetomidine (DEX) could have an analgesic effect on pain transmission through the modulation of brain-derived neurotrophic factor (BDNF). In addition, KCC2-induced shift in neuronal Cl− homeostasis is crucial for postsynaptic inhibition mediated by GABAA receptors. Accumulating evidence shows that nerve injury, peripheral inflammation and stress activate the spinal BDNF/TrkB signal, which results in the downregulation of KCC2 transport and expression, eventually leads to GAGAergic disinhibition and hyperalgesia. The aim of this experiment was to explore the interaction between DEX and KCC2 at a molecular level in rats in the persistent postsurgical pain (PPSP). Methods PPSP in rats was evoked by the skin/muscle incision and retraction (SMIR). Mechanical hypersensitivity was assessed with the Dynamic Plantar Aesthesiometer. Western blot and immunofluorescence assay were used to assess the expressions of related proteins. Results In the first part of our experiment, the results revealed that the BDNF/TrkB-KCC2 signal plays a critical role in the development of SMIR-evoked PPSP; the second part showed that intraperitoneal administrations of 40 µg/kg DEX at 15 min presurgery and 1 to 3 days post-surgery significantly attenuated SMIR-evoked PPSP. Simultaneously, SMIR-induced KCC2 downregulation was partly reversed, which coincided with the inhibition of the BDNF/TrkB signal in the spinal dorsal horn. Moreover, intrathecal administrations of KCC2 inhibitor VU0240551 significantly reduced the analgesic effect of DEX on SMIR-evoked PPSP. Conclusion The results of our study indicated that DEX attenuated PPSP by restoring KCC2 function through reducing BDNF/TrkB signal in the spinal dorsal horn in rats, which provides a new insight into the treatment of chronic pain in clinical postsurgical pain management.
Collapse
Affiliation(s)
- Shuhong Dai
- Department of Anesthesiology, XuZhou Central Hospital, Xuzhou, China
| | - Yu Qi
- Department of Anesthesiology, XuZhou Central Hospital, Xuzhou, China
| | - Jie Fu
- Department of Anesthesiology, XuZhou Central Hospital, Xuzhou, China
| | - Na Li
- Department of Anesthesiology, XuZhou Central Hospital, Xuzhou, China
| | - Xu Zhang
- Department of Anesthesiology, XuZhou Central Hospital, Xuzhou, China
| | - Juan Zhang
- The Affiliated Nanjing Drum Tower Hospital, Medical School, Nanjing University, Nanjing, China
| | - Wei Zhang
- The Affiliated Nanjing Drum Tower Hospital, Medical School, Nanjing University, Nanjing, China
| | - Haijun Xu
- Department of Anesthesiology, XuZhou Central Hospital, Xuzhou, China
| | - Hai Zhou
- Department of Anesthesiology, XuZhou Central Hospital, Xuzhou, China
| | - Zhengliang Ma
- The Affiliated Nanjing Drum Tower Hospital, Medical School, Nanjing University, Nanjing, China
| |
Collapse
|
46
|
Di Cristo G, Awad PN, Hamidi S, Avoli M. KCC2, epileptiform synchronization, and epileptic disorders. Prog Neurobiol 2018; 162:1-16. [DOI: 10.1016/j.pneurobio.2017.11.002] [Citation(s) in RCA: 42] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2017] [Revised: 11/09/2017] [Accepted: 11/28/2017] [Indexed: 12/31/2022]
|
47
|
Dargaei Z, Bang JY, Mahadevan V, Khademullah CS, Bedard S, Parfitt GM, Kim JC, Woodin MA. Restoring GABAergic inhibition rescues memory deficits in a Huntington's disease mouse model. Proc Natl Acad Sci U S A 2018; 115:E1618-E1626. [PMID: 29382760 PMCID: PMC5816181 DOI: 10.1073/pnas.1716871115] [Citation(s) in RCA: 62] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/01/2022] Open
Abstract
Huntington's disease (HD) is classically characterized as a movement disorder, however cognitive impairments precede the motor symptoms by ∼15 y. Based on proteomic and bioinformatic data linking the Huntingtin protein (Htt) and KCC2, which is required for hyperpolarizing GABAergic inhibition, and the important role of inhibition in learning and memory, we hypothesized that aberrant KCC2 function contributes to the hippocampal-associated learning and memory deficits in HD. We discovered that Htt and KCC2 interact in the hippocampi of wild-type and R6/2-HD mice, with a decrease in KCC2 expression in the hippocampus of R6/2 and YAC128 mice. The reduced expression of the Cl--extruding cotransporter KCC2 is accompanied by an increase in the Cl--importing cotransporter NKCC1, which together result in excitatory GABA in the hippocampi of HD mice. NKCC1 inhibition by the FDA-approved NKCC1 inhibitor bumetanide abolished the excitatory action of GABA and rescued the performance of R6/2 mice on hippocampal-associated behavioral tests.
Collapse
Affiliation(s)
- Zahra Dargaei
- Department of Cell and Systems Biology, University of Toronto, Toronto, ON M5S 3G5, Canada
| | - Jee Yoon Bang
- Department of Psychology, University of Toronto, Toronto, ON M5S 3G3, Canada
| | - Vivek Mahadevan
- Department of Cell and Systems Biology, University of Toronto, Toronto, ON M5S 3G5, Canada
| | - C Sahara Khademullah
- Department of Cell and Systems Biology, University of Toronto, Toronto, ON M5S 3G5, Canada
| | - Simon Bedard
- Department of Cell and Systems Biology, University of Toronto, Toronto, ON M5S 3G5, Canada
| | - Gustavo Morrone Parfitt
- Department of Cell and Systems Biology, University of Toronto, Toronto, ON M5S 3G5, Canada
- Department of Psychology, University of Toronto, Toronto, ON M5S 3G3, Canada
| | - Jun Chul Kim
- Department of Psychology, University of Toronto, Toronto, ON M5S 3G3, Canada
| | - Melanie A Woodin
- Department of Cell and Systems Biology, University of Toronto, Toronto, ON M5S 3G5, Canada;
| |
Collapse
|
48
|
Frenette-Cotton R, Marcoux AA, Garneau AP, Noel M, Isenring P. Phosphoregulation of K + -Cl - cotransporters during cell swelling: Novel insights. J Cell Physiol 2018; 233:396-408. [PMID: 28276587 DOI: 10.1002/jcp.25899] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2016] [Accepted: 03/06/2017] [Indexed: 01/21/2023]
Abstract
The K+ -Cl- cotransporters (KCCs) belong to the cation-Cl- cotransporter family and consist of four isoforms and many splice variants. Their main role is to promote electroneutral efflux of K+ and Cl- ions across the surface of many cell types and, thereby, to regulate intracellular ion concentration, cell volume, and epithelial salt movement. These transport systems are induced by an increase in cell volume and are less active at lower intracellular [Cl- ] (Cli ), but the mechanisms at play are still ill-defined. In this work, we have exploited the Xenopus laevis expression system to study the role of lysine-deficient protein kinases (WNKs), protein phosphatases 1 (PP1s), and SPS1-related proline/alanine-rich kinase (SPAK) in KCC4 regulation during cell swelling. We have found that WNK4 and PP1 regulate KCC4 activity as part of a common signaling module, but that they do not exert their effects through SPAK or carrier dephosphorylation. We have also found that the phosphatases at play include PP1α and PP1γ1, but that WNK4 acts directly on the PP1s instead of the opposite. Unexpectedly, however, both cell swelling and a T926A substitution in the C-terminus of full-length KCC4 led to higher levels of heterologous K+ -Cl- cotransport and overall carrier phosphorylation. These results imply that the response to cell swelling must also involve allosteric-sensitive kinase-dependent phosphoacceptor sites in KCC4. They are thus partially inconsistent with previous models of KCC regulation.
Collapse
Affiliation(s)
| | - Andrée-Anne Marcoux
- Nephrology Research Group, Department of Medicine, Laval University, Québec, Québec, Canada
| | - Alexandre P Garneau
- Nephrology Research Group, Department of Medicine, Laval University, Québec, Québec, Canada
| | - Micheline Noel
- Nephrology Research Group, Department of Medicine, Laval University, Québec, Québec, Canada
| | - Paul Isenring
- Nephrology Research Group, Department of Medicine, Laval University, Québec, Québec, Canada
| |
Collapse
|
49
|
Agez M, Schultz P, Medina I, Baker DJ, Burnham MP, Cardarelli RA, Conway LC, Garnier K, Geschwindner S, Gunnarsson A, McCall EJ, Frechard A, Audebert S, Deeb TZ, Moss SJ, Brandon NJ, Wang Q, Dekker N, Jawhari A. Molecular architecture of potassium chloride co-transporter KCC2. Sci Rep 2017; 7:16452. [PMID: 29184062 PMCID: PMC5705597 DOI: 10.1038/s41598-017-15739-1] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2017] [Accepted: 10/27/2017] [Indexed: 01/15/2023] Open
Abstract
KCC2 is a neuron specific K+-Cl− co-transporter that controls neuronal chloride homeostasis, and is critically involved in many neurological diseases including brain trauma, epilepsies, autism and schizophrenia. Despite significant accumulating data on the biology and electrophysiological properties of KCC2, structure-function relationships remain poorly understood. Here we used calixarene detergent to solubilize and purify wild-type non-aggregated and homogenous KCC2. Specific binding of inhibitor compound VU0463271 was demonstrated using surface plasmon resonance (SPR). Mass spectrometry revealed glycosylations and phosphorylations as expected from functional KCC2. We show by electron microscopy (EM) that KCC2 exists as monomers and dimers in solution. Monomers are organized into “head” and “core” domains connected by a flexible “linker”. Dimers are asymmetrical and display a bent “S-shape” architecture made of four distinct domains and a flexible dimerization interface. Chemical crosslinking in reducing conditions shows that disulfide bridges are involved in KCC2 dimerization. Moreover, we show that adding a tag to the C-terminus is detrimental to KCC2 function. We postulate that the conserved KCC2 C-ter may be at the interface of dimerization. Taken together, our findings highlight the flexible multi-domain structure of KCC2 with variable anchoring points at the dimerization interface and an important C-ter extremity providing the first in-depth functional architecture of KCC2.
Collapse
Affiliation(s)
- Morgane Agez
- CALIXAR, 60 avenue Rockefeller, 69008, Lyon, France
| | - Patrick Schultz
- Department of Integrated Structural Biology, IGBMC (Institut de Génétique et de Biologie Moléculaire et Cellulaire) INSERM, U964; CNRS/Strasbourg University, UMR7104 1, rue Laurent Fries, BP10142, 67404, Illkirch, France
| | | | - David J Baker
- Discovery Sciences, IMED Biotech Unit, AstraZeneca, Cambridge, UK
| | - Matthew P Burnham
- Discovery Sciences, IMED Biotech Unit, AstraZeneca, Alderley Park, UK
| | - Ross A Cardarelli
- AstraZeneca Tufts Laboratory for Basic and Translational Neuroscience, Boston, Massachusetts, 02111, USA
| | - Leslie C Conway
- AstraZeneca Tufts Laboratory for Basic and Translational Neuroscience, Boston, Massachusetts, 02111, USA
| | | | | | - Anders Gunnarsson
- Discovery Sciences, IMED Biotech Unit, AstraZeneca, Gothenburg, Sweden
| | - Eileen J McCall
- Discovery Sciences, IMED Biotech Unit, AstraZeneca, Cambridge, UK
| | - Alexandre Frechard
- Department of Integrated Structural Biology, IGBMC (Institut de Génétique et de Biologie Moléculaire et Cellulaire) INSERM, U964; CNRS/Strasbourg University, UMR7104 1, rue Laurent Fries, BP10142, 67404, Illkirch, France
| | - Stéphane Audebert
- Aix Marseille Univ, CNRS, INSERM, Institut Paoli-Calmettes, CRCM, Marseille Protéomique, Marseille, France
| | - Tarek Z Deeb
- AstraZeneca Tufts Laboratory for Basic and Translational Neuroscience, Boston, Massachusetts, 02111, USA
| | - Stephen J Moss
- Department of Neuroscience, Tufts University School of Medicine, Boston, MA, 02111, USA.,Department of Neuroscience, Physiology and Pharmacology, University College, London, WC1E, 6BT, UK
| | - Nicholas J Brandon
- AstraZeneca Tufts Laboratory for Basic and Translational Neuroscience, Boston, Massachusetts, 02111, USA.,Neuroscience, IMED Biotech Unit, AstraZeneca, Boston, MA, USA
| | - Qi Wang
- AstraZeneca Tufts Laboratory for Basic and Translational Neuroscience, Boston, Massachusetts, 02111, USA.,Neuroscience, IMED Biotech Unit, AstraZeneca, Boston, MA, USA
| | - Niek Dekker
- Discovery Sciences, IMED Biotech Unit, AstraZeneca, Gothenburg, Sweden.
| | - Anass Jawhari
- CALIXAR, 60 avenue Rockefeller, 69008, Lyon, France.
| |
Collapse
|
50
|
Abstract
K+-Cl- co-transporter 2 (KCC2/SLC12A5) is a neuronal specific cation chloride co-transporter which is active under isotonic conditions, and thus a key regulator of intracellular Cl- levels. It also has an ion transporter-independent structural role in modulating the maturation and regulation of excitatory glutamatergic synapses. KCC2 levels are developmentally regulated, and a postnatal upregulation of KCC2 generates a low intracellular chloride concentration that allows the neurotransmitters γ-aminobutyric acid (GABA) and glycine to exert inhibitory neurotransmission through its Cl- permeating channel. Functional expression of KCC2 at the neuronal cell surface is necessary for its activity, and impairment in KCC2 cell surface transport and/or internalization may underlie a range of neuropathological conditions. Although recent advances have shed light on a range of cellular mechanisms regulating KCC2 activity, little is known about its membrane trafficking itinerary and regulatory proteins. In this review, known membrane trafficking signals, pathways and mechanisms pertaining to KCC2's functional surface expression are discussed.
Collapse
Affiliation(s)
- Bor Luen Tang
- a Department of Biochemistry, Yong Loo Lin School of Medicine , National University Health System , Singapore.,b NUS Graduate School for Integrative Sciences and Engineering , National University of Singapore , Singapore
| |
Collapse
|