1
|
Becker L, Hausmann J, Wellpott R, Hartmann AM. Highly conserved ion binding sites are not all functionally relevant in mouse KCC4. Front Mol Biosci 2025; 12:1556250. [PMID: 40230454 PMCID: PMC11994965 DOI: 10.3389/fmolb.2025.1556250] [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: 01/06/2025] [Accepted: 03/03/2025] [Indexed: 04/16/2025] Open
Abstract
Introduction The potassium chloride cotransporter 4 (KCC4) is expressed in various tissues and plays an important role in distal renal acidification and hearing development. Although KCCs transport K+ and Cl- in a 1:1 stoichiometry, two Cl- coordination sites were indicated via cryo-electron microscopy (CryoEM). Methods In a comprehensive analysis, we analyzed here the consequences of point mutation of residues coordinating potassium, and chloride in the first (Cl1) and second (Cl2) coordinating site in KCC4 using Tl+ based flux measurements. Results Surprisingly, not all highly conserved coordination sites in KCC4 are essential. Three out of five residues (N131, Y216, and T432) are functionally relevant for potassium coordination. For chloride coordination in Cl1, all three residues (G134, V135, and I136) are important, whereas three out of four residues (G433, M435, and Y589) are relevant for chloride binding in Cl2. As all ion coordination sites are important in KCC2, this indicates that there is a certain flexibility in the stringency of ion coordination in KCC4. One possible reason for the different relevance of ion coordination sites could be the large extracellular loop (LEL). The LEL is structured differently within KCCs and is directly linked to the transmembrane domain (TM) 6, where most of the coordination sites reside. Substitution of ion coordination sites in the KCC22-4-2 chimera, in which the LEL from mouse KCC4 is exchanged with the LEL of rat KCC2, have the same effect as substitutions in rat KCC2. An exception is the substitution of the potassium coordination site I111 in TM1, which shows enhanced activity in the KCC22-4-2 chimera compared to the impaired activity in rat KCC2 and not affected activity in mouse KCC4. Conclusion Thus, the different relevance of the ion coordination sites between KCC2 and KCC4 cannot be attributed solely to the different structured LEL; other structural elements must also be involved here.
Collapse
Affiliation(s)
- Lisa Becker
- Division of Neurogenetics, School of Medicine and Health Science, Carl von Ossietzky University of Oldenburg, Oldenburg, Germany
| | - Jens Hausmann
- Division of Anatomy, School of Medicine and Health Science, Carl von Ossietzky University of Oldenburg, Oldenburg, Germany
| | - Rieke Wellpott
- Division of Neurogenetics, School of Medicine and Health Science, Carl von Ossietzky University of Oldenburg, Oldenburg, Germany
| | - Anna-Maria Hartmann
- Division of Neurogenetics, School of Medicine and Health Science, Carl von Ossietzky University of Oldenburg, Oldenburg, Germany
- Research Center Neurosensory Sciences, Carl von Ossietzky University of Oldenburg, Oldenburg, Germany
| |
Collapse
|
2
|
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
|
3
|
Huang 黄玉莹 Y, Chen 陈红 H, Shao 邵建英 JY, Zhou 周京京 JJ, Chen 陈少瑞 SR, Pan 潘惠麟 HL. Constitutive KCC2 Cell- and Synapse-Specifically Regulates NMDA Receptor Activity in the Spinal Cord. J Neurosci 2024; 44:e1943232023. [PMID: 38124193 PMCID: PMC10860486 DOI: 10.1523/jneurosci.1943-23.2023] [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: 10/13/2023] [Revised: 11/20/2023] [Accepted: 12/05/2023] [Indexed: 12/23/2023] Open
Abstract
K+-Cl- cotransporter-2 (KCC2) critically controls neuronal chloride homeostasis and maintains normal synaptic inhibition by GABA and glycine. Nerve injury diminishes synaptic inhibition in the spinal cord via KCC2 impairment. However, how KCC2 regulates nociceptive input to spinal excitatory and inhibitory neurons remains elusive. Here, we show that basal GABA reversal potentials were significantly more depolarized in vesicular GABA transporter (VGAT)-expressing inhibitory neurons than those in vesicular glutamate transporter-2 (VGluT2)-expressing excitatory neurons in spinal cords of male and female mice. Strikingly, inhibiting KCC2 with VU0463271 increased currents elicited by puff NMDA and the NMDAR-mediated frequency of mEPSCs in VGluT2, but not in VGAT, dorsal horn neurons. Notably, VU0463271 had no effect on EPSCs monosynaptically evoked from the dorsal root in VGluT2 neurons. Furthermore, VU0463271 augmented α2δ-1-NMDAR interactions and their protein levels in spinal cord synaptosomes. In Cacna2d1 KO mice, VU0463271 had no effect on puff NMDA currents or the mEPSC frequency in dorsal horn neurons. Disrupting α2δ-1-NMDAR interactions with α2δ-1 C-terminus mimicking peptide diminished VU0463271-induced potentiation in the mEPSC frequency and puff NMDA currents in VGluT2 neurons. Additionally, intrathecal injection of VU0463271 reduced mechanical and thermal thresholds in wild-type mice, but not in Cacna2d1 KO mice. VU0463271-induced pain hypersensitivity in mice was abrogated by co-treatment with the NMDAR antagonist, pregabalin (an α2δ-1 inhibitory ligand), or α2δ-1 C-terminus mimicking peptide. Our findings suggest that KCC2 controls presynaptic and postsynaptic NMDAR activity specifically in excitatory dorsal horn neurons. KCC2 impairment preferentially potentiates nociceptive transmission between spinal excitatory interneurons via α2δ-1-bound NMDARs.Significance statementImpaired function of potassium-chloride cotransporter-2 (KCC2), a key regulator of neuronal inhibition, in the spinal cord plays a major role in neuropathic pain. This study unveils that KCC2 controls spinal nociceptive synaptic strength via NMDA receptors in a cell type- and synapse type-specific manner. KCC2 inhibition preferentially augments presynaptic and postsynaptic NMDA receptor activity in spinal excitatory interneurons via α2δ-1 (previously known as a calcium channel subunit). Importantly, spinal KCC2 impairment triggers pain hypersensitivity through α2δ-1-coupled NMDA receptors. These findings pinpoint the cell and molecular substrates for the reciprocal relationship between spinal synaptic inhibition and excitation in chronic neuropathic pain. Targeting both KCC2 and α2δ-1–NMDA receptor complexes could be an effective strategy in managing neuropathic pain conditions.
Collapse
Affiliation(s)
- Yuying Huang 黄玉莹
- Department of Anesthesiology and Perioperative Medicine, Center for Neuroscience and Pain Research, The University of Texas MD Anderson Cancer Center, Houston 77030, Texas
| | - Hong Chen 陈红
- Department of Anesthesiology and Perioperative Medicine, Center for Neuroscience and Pain Research, The University of Texas MD Anderson Cancer Center, Houston 77030, Texas
| | - Jian-Ying Shao 邵建英
- Department of Anesthesiology and Perioperative Medicine, Center for Neuroscience and Pain Research, The University of Texas MD Anderson Cancer Center, Houston 77030, Texas
| | - Jing-Jing Zhou 周京京
- Department of Anesthesiology and Perioperative Medicine, Center for Neuroscience and Pain Research, The University of Texas MD Anderson Cancer Center, Houston 77030, Texas
| | - Shao-Rui Chen 陈少瑞
- Department of Anesthesiology and Perioperative Medicine, Center for Neuroscience and Pain Research, The University of Texas MD Anderson Cancer Center, Houston 77030, Texas
| | - Hui-Lin Pan 潘惠麟
- Department of Anesthesiology and Perioperative Medicine, Center for Neuroscience and Pain Research, The University of Texas MD Anderson Cancer Center, Houston 77030, Texas
| |
Collapse
|
4
|
Becker L, Hausmann J, Hartmann AM. Both chloride-binding sites are required for KCC2-mediated transport. J Biol Chem 2023; 299:105190. [PMID: 37625593 PMCID: PMC10518353 DOI: 10.1016/j.jbc.2023.105190] [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: 01/25/2023] [Revised: 07/30/2023] [Accepted: 08/01/2023] [Indexed: 08/27/2023] Open
Abstract
The K+-Cl- cotransporter 2 (KCC2) plays an important role in inhibitory neurotransmission, and its impairment is associated with neurological and psychiatric disorders, including epilepsy, schizophrenia, and autism. Although KCCs transport K+ and Cl- in a 1:1 stoichiometry, two Cl- coordination sites were indicated via cryo-EM. In a comprehensive analysis, we analyzed the consequences of point mutations of residues coordinating Cl- in Cl1 and Cl2. Individual mutations of residues in Cl1 and Cl2 reduce or abolish KCC2WT function, indicating a crucial role of both Cl- coordination sites for KCC2 function. Structural changes in the extracellular loop 2 by inserting a 3xHA tag switches the K+ coordination site to another position. To investigate, whether the extension of the extracellular loop 2 with the 3xHA tag also affects the coordination of the two Cl- coordination sites, we carried out the analogous experiments for both Cl- coordinating sites in the KCC2HA construct. These analyses showed that most of the individual mutation of residues in Cl1 and Cl2 in the KCC2HA construct reduces or abolishes KCC2 function, indicating that the coordination of Cl- remains at the same position. However, the coupling of K+ and Cl- in Cl1 is still apparent in the KCC2HA construct, indicating a mutual dependence of both ions. In addition, the coordination residue Tyr569 in Cl2 shifted in KCC2HA. Thus, conformational changes in the extracellular domain affect K+ and Cl--binding sites. However, the effect on the Cl--binding sites is subtler.
Collapse
Affiliation(s)
- Lisa Becker
- Division of Neurogenetics, School of Medicine and Health Sciences, Carl von Ossietzky University Oldenburg, Oldenburg, Germany
| | - Jens Hausmann
- Division of Anatomy, School of Medicine and Health Sciences, Carl von Ossietzky University 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
|
5
|
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
|
6
|
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
|
7
|
Lam P, Newland J, Faull RLM, Kwakowsky A. Cation-Chloride Cotransporters KCC2 and NKCC1 as Therapeutic Targets in Neurological and Neuropsychiatric Disorders. Molecules 2023; 28:1344. [PMID: 36771011 PMCID: PMC9920462 DOI: 10.3390/molecules28031344] [Citation(s) in RCA: 22] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2022] [Revised: 01/21/2023] [Accepted: 01/25/2023] [Indexed: 02/04/2023] Open
Abstract
Neurological diseases including Alzheimer's, Huntington's disease, Parkinson's disease, Down syndrome and epilepsy, and neuropsychiatric disorders such as schizophrenia, are conditions that affect not only individuals but societies on a global scale. Current therapies offer a means for small symptomatic relief, but recently there has been increasing demand for therapeutic alternatives. The γ-aminobutyric acid (GABA)ergic signaling system has been investigated for developing new therapies as it has been noted that any dysfunction or changes to this system can contribute to disease progression. Expression of the K-Cl-2 (KCC2) and N-K-C1-1 (NKCC1) cation-chloride cotransporters (CCCs) has recently been linked to the disruption of GABAergic activity by affecting the polarity of GABAA receptor signaling. KCC2 and NKCC1 play a part in multiple neurological and neuropsychiatric disorders, making them a target of interest for potential therapies. This review explores current research suggesting the pathophysiological role and therapeutic importance of KCC2 and NKCC1 in neuropsychiatric and neurological disorders.
Collapse
Affiliation(s)
- Patricia Lam
- Centre for Brain Research, Department of Anatomy and Medical Imaging, Faculty of Medical and Health Sciences, University of Auckland, Auckland 1023, New Zealand
| | - Julia Newland
- Centre for Brain Research, Department of Anatomy and Medical Imaging, Faculty of Medical and Health Sciences, University of Auckland, Auckland 1023, New Zealand
| | - Richard L. M. Faull
- Centre for Brain Research, Department of Anatomy and Medical Imaging, Faculty of Medical and Health Sciences, University of Auckland, Auckland 1023, New Zealand
| | - Andrea Kwakowsky
- Centre for Brain Research, Department of Anatomy and Medical Imaging, Faculty of Medical and Health Sciences, University of Auckland, Auckland 1023, New Zealand
- Pharmacology and Therapeutics, School of Medicine, Galway Neuroscience Centre, University of Galway, H91 W5P7 Galway, Ireland
| |
Collapse
|
8
|
Dedek A, Xu J, Lorenzo LÉ, Godin AG, Kandegedara CM, Glavina G, Landrigan JA, Lombroso PJ, De Koninck Y, Tsai EC, Hildebrand ME. Sexual dimorphism in a neuronal mechanism of spinal hyperexcitability across rodent and human models of pathological pain. Brain 2022; 145:1124-1138. [PMID: 35323848 PMCID: PMC9050559 DOI: 10.1093/brain/awab408] [Citation(s) in RCA: 32] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2021] [Revised: 10/03/2021] [Accepted: 10/15/2021] [Indexed: 12/23/2022] Open
Abstract
The prevalence and severity of many chronic pain syndromes differ across sex, and recent studies have identified differences in immune signalling within spinal nociceptive circuits as a potential mediator. Although it has been proposed that sex-specific pain mechanisms converge once they reach neurons within the superficial dorsal horn, direct investigations using rodent and human preclinical pain models have been lacking. Here, we discovered that in the Freund’s adjuvant in vivo model of inflammatory pain, where both male and female rats display tactile allodynia, a pathological coupling between KCC2-dependent disinhibition and N-methyl-D-aspartate receptor (NMDAR) potentiation within superficial dorsal horn neurons was observed in male but not female rats. Unlike males, the neuroimmune mediator brain-derived neurotrophic factor (BDNF) failed to downregulate inhibitory signalling elements (KCC2 and STEP61) and upregulate excitatory elements (pFyn, GluN2B and pGluN2B) in female rats, resulting in no effect of ex vivo brain-derived neurotrophic factor on synaptic NMDAR responses in female lamina I neurons. Importantly, this sex difference in spinal pain processing was conserved from rodents to humans. As in rodents, ex vivo spinal treatment with BDNF downregulated markers of disinhibition and upregulated markers of facilitated excitation in superficial dorsal horn neurons from male but not female human organ donors. Ovariectomy in female rats recapitulated the male pathological pain neuronal phenotype, with BDNF driving a coupling between disinhibition and NMDAR potentiation in adult lamina I neurons following the prepubescent elimination of sex hormones in females. This discovery of sexual dimorphism in a central neuronal mechanism of chronic pain across species provides a foundational step towards a better understanding and treatment for pain in both sexes.
Collapse
Affiliation(s)
- Annemarie Dedek
- Department of Neuroscience, Carleton University, K1S 5B6 Ontario, Canada.,Neuroscience Program, Ottawa Hospital Research Institute, K1Y 4M9 Ontario, Canada
| | - Jian Xu
- Child Study Center, Yale University School of Medicine, New Haven, CT 06519, USA
| | | | - Antoine G Godin
- CERVO Brain Research Centre, Quebec Mental Health Institute, Quebec G1E 1T2, Canada.,Department of Psychiatry and Neuroscience, Université Laval, Quebec G1V 0A6, Canada
| | - Chaya M Kandegedara
- Department of Neuroscience, Carleton University, K1S 5B6 Ontario, Canada.,Neuroscience Program, Ottawa Hospital Research Institute, K1Y 4M9 Ontario, Canada
| | - Geneviève Glavina
- CERVO Brain Research Centre, Quebec Mental Health Institute, Quebec G1E 1T2, Canada
| | | | - Paul J Lombroso
- Child Study Center, Yale University School of Medicine, New Haven, CT 06519, USA
| | - Yves De Koninck
- CERVO Brain Research Centre, Quebec Mental Health Institute, Quebec G1E 1T2, Canada.,Department of Psychiatry and Neuroscience, Université Laval, Quebec G1V 0A6, Canada
| | - Eve C Tsai
- Neuroscience Program, Ottawa Hospital Research Institute, K1Y 4M9 Ontario, Canada.,Brain and Mind Research Institute, University of Ottawa, Ontario K1N 6N5, Canada.,Department of Surgery, Division of Neurosurgery, The Ottawa Hospital, Ontario K1Y 4E9, Canada
| | - Michael E Hildebrand
- Department of Neuroscience, Carleton University, K1S 5B6 Ontario, Canada.,Neuroscience Program, Ottawa Hospital Research Institute, K1Y 4M9 Ontario, Canada
| |
Collapse
|
9
|
Serranilla M, Woodin MA. Striatal Chloride Dysregulation and Impaired GABAergic Signaling Due to Cation-Chloride Cotransporter Dysfunction in Huntington’s Disease. Front Cell Neurosci 2022; 15:817013. [PMID: 35095429 PMCID: PMC8795088 DOI: 10.3389/fncel.2021.817013] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2021] [Accepted: 12/24/2021] [Indexed: 11/13/2022] Open
Abstract
Intracellular chloride (Cl–) levels in mature neurons must be tightly regulated for the maintenance of fast synaptic inhibition. In the mature central nervous system (CNS), synaptic inhibition is primarily mediated by gamma-amino butyric acid (GABA), which binds to Cl– permeable GABAA receptors (GABAARs). The intracellular Cl– concentration is primarily maintained by the antagonistic actions of two cation-chloride cotransporters (CCCs): Cl–-importing Na+-K+-Cl– co-transporter-1 (NKCC1) and Cl– -exporting K+-Cl– co-transporter-2 (KCC2). In mature neurons in the healthy brain, KCC2 expression is higher than NKCC1, leading to lower levels of intracellular Cl–, and Cl– influx upon GABAAR activation. However, in neurons of the immature brain or in neurological disorders such as epilepsy and traumatic brain injury, impaired KCC2 function and/or enhanced NKCC1 expression lead to intracellular Cl– accumulation and GABA-mediated excitation. In Huntington’s disease (HD), KCC2- and NKCC1-mediated Cl–-regulation are also altered, which leads to GABA-mediated excitation and contributes to the development of cognitive and motor impairments. This review summarizes the role of Cl– (dys)regulation in the healthy and HD brain, with a focus on the basal ganglia (BG) circuitry and CCCs as potential therapeutic targets in the treatment of HD.
Collapse
|
10
|
Noriega LG, Melo Z, Rajaram RD, Mercado A, Tovar AR, Velazquez‐Villegas LA, Castañeda‐Bueno M, Reyes‐López Y, Ryu D, Rojas‐Vega L, Magaña‐Avila G, López‐Barradas AM, Sánchez‐Hernández M, Debonneville A, Doucet A, Cheval L, Torres N, Auwerx J, Staub O, Gamba G. SIRT7 modulates the stability and activity of the renal K-Cl cotransporter KCC4 through deacetylation. EMBO Rep 2021; 22:e50766. [PMID: 33749979 PMCID: PMC8097349 DOI: 10.15252/embr.202050766] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2020] [Revised: 02/03/2021] [Accepted: 02/19/2021] [Indexed: 11/09/2022] Open
Abstract
SIRT7 is a NAD+ -dependent deacetylase that controls important aspects of metabolism, cancer, and bone formation. However, the molecular targets and functions of SIRT7 in the kidney are currently unknown. In silico analysis of kidney transcripts of the BXD murine genetic reference population revealed a positive correlation between Sirt7 and Slc12a7 mRNA expression, suggesting a link between the corresponding proteins that these transcripts encode, SIRT7, and the K-Cl cotransporter KCC4, respectively. Here, we find that protein levels and activity of heterologously expressed KCC4 are significantly modulated depending on its acetylation status in Xenopus laevis oocytes. Moreover, SIRT7 interacts with KCC4 in a NAD+ -dependent manner and increases its stability and activity in HEK293 cells. Interestingly, metabolic acidosis increases SIRT7 expression in kidney, as occurs with KCC4. In contrast, total SIRT7-deficient mice present lower KCC4 expression and an exacerbated metabolic acidosis than wild-type mice during an ammonium chloride challenge. Altogether, our data suggest that SIRT7 interacts with, stabilizes and modulates KCC4 activity through deacetylation, and reveals a novel role for SIRT7 in renal physiology.
Collapse
Affiliation(s)
- Lilia G Noriega
- Department of Nutrition PhysiologyInstituto Nacional de Ciencias Médicas y Nutrición Salvador ZubiránMexico CityMexico
| | - Zesergio Melo
- Department of Nephrology and Mineral MetabolismInstituto Nacional de Ciencias Médicas y Nutrición Salvador ZubiránMexico CityMexico
- CONACYT‐Centro de Investigación Biomédica de OccidenteInstituto Mexicano del Seguro SocialGuadalajaraJaliscoMexico
| | - Renuga D Rajaram
- Department of Pharmacology and ToxicologyUniversity of LausanneLausanneSwitzerland
- National Centre of Competence in Research, “Kidney.ch”ZurichSwitzerland
| | - Adriana Mercado
- Department of NephrologyInstituto Nacional de Cardiología Ignacio ChávezMexico CityMexico
| | - Armando R Tovar
- Department of Nutrition PhysiologyInstituto Nacional de Ciencias Médicas y Nutrición Salvador ZubiránMexico CityMexico
| | - Laura A Velazquez‐Villegas
- Department of Nutrition PhysiologyInstituto Nacional de Ciencias Médicas y Nutrición Salvador ZubiránMexico CityMexico
| | - María Castañeda‐Bueno
- Department of Nephrology and Mineral MetabolismInstituto Nacional de Ciencias Médicas y Nutrición Salvador ZubiránMexico CityMexico
| | - Yazmín Reyes‐López
- Department of Nutrition PhysiologyInstituto Nacional de Ciencias Médicas y Nutrición Salvador ZubiránMexico CityMexico
| | - Dongryeol Ryu
- Laboratory of Integrative and Systems Physiology (LISP)École Polytechnique Fédérale de LausanneLausanneSwitzerland
- Present address:
Department of Molecular Cell BiologySungkyunkwan University School of MedicineSuwonKorea
| | - Lorena Rojas‐Vega
- Department of Nephrology and Mineral MetabolismInstituto Nacional de Ciencias Médicas y Nutrición Salvador ZubiránMexico CityMexico
| | - German Magaña‐Avila
- Department of Nephrology and Mineral MetabolismInstituto Nacional de Ciencias Médicas y Nutrición Salvador ZubiránMexico CityMexico
| | - Adriana M López‐Barradas
- Department of Nutrition PhysiologyInstituto Nacional de Ciencias Médicas y Nutrición Salvador ZubiránMexico CityMexico
| | | | - Anne Debonneville
- Department of Pharmacology and ToxicologyUniversity of LausanneLausanneSwitzerland
- National Centre of Competence in Research, “Kidney.ch”ZurichSwitzerland
| | - Alain Doucet
- Centre de Recherche des CordeliersINSERM, Sorbonne Universités, USPC, Université Paris Descartes, Université Paris Diderot, Physiologie Rénale et TubulopathiesCNRS ERL 8228ParisFrance
| | - Lydie Cheval
- Centre de Recherche des CordeliersINSERM, Sorbonne Universités, USPC, Université Paris Descartes, Université Paris Diderot, Physiologie Rénale et TubulopathiesCNRS ERL 8228ParisFrance
| | - Nimbe Torres
- Department of Nutrition PhysiologyInstituto Nacional de Ciencias Médicas y Nutrición Salvador ZubiránMexico CityMexico
| | - Johan Auwerx
- Laboratory of Integrative and Systems Physiology (LISP)École Polytechnique Fédérale de LausanneLausanneSwitzerland
| | - Olivier Staub
- Department of Pharmacology and ToxicologyUniversity of LausanneLausanneSwitzerland
- National Centre of Competence in Research, “Kidney.ch”ZurichSwitzerland
| | - Gerardo Gamba
- Department of Nephrology and Mineral MetabolismInstituto Nacional de Ciencias Médicas y Nutrición Salvador ZubiránMexico CityMexico
- Molecular Physiology UnitInstituto de Investigaciones BiomédicasUniversidad Nacional Autónoma de MéxicoMexico CityMexico
| |
Collapse
|
11
|
Zhang S, Zhou J, Zhang Y, Liu T, Friedel P, Zhuo W, Somasekharan S, Roy K, Zhang L, Liu Y, Meng X, Deng H, Zeng W, Li G, Forbush B, Yang M. The structural basis of function and regulation of neuronal cotransporters NKCC1 and KCC2. Commun Biol 2021; 4:226. [PMID: 33597714 PMCID: PMC7889885 DOI: 10.1038/s42003-021-01750-w] [Citation(s) in RCA: 46] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2020] [Accepted: 01/22/2021] [Indexed: 11/08/2022] Open
Abstract
NKCC and KCC transporters mediate coupled transport of Na++K++Cl- and K++Cl- across the plasma membrane, thus regulating cell Cl- concentration and cell volume and playing critical roles in transepithelial salt and water transport and in neuronal excitability. The function of these transporters has been intensively studied, but a mechanistic understanding has awaited structural studies of the transporters. Here, we present the cryo-electron microscopy (cryo-EM) structures of the two neuronal cation-chloride cotransporters human NKCC1 (SLC12A2) and mouse KCC2 (SLC12A5), along with computational analysis and functional characterization. These structures highlight essential residues in ion transport and allow us to propose mechanisms by which phosphorylation regulates transport activity.
Collapse
Affiliation(s)
- Sensen Zhang
- Ministry of Education Key Laboratory of Protein Science, Tsinghua-Peking Center for Life Sciences, Beijing Advanced Innovation Center for Structural Biology, School of Life Sciences, Tsinghua University, Beijing, China
| | - Jun Zhou
- Ministry of Education Key Laboratory of Protein Science, Tsinghua-Peking Center for Life Sciences, Beijing Advanced Innovation Center for Structural Biology, School of Life Sciences, Tsinghua University, Beijing, China
| | - Yuebin Zhang
- State Key Laboratory of Molecular Reaction Dynamics, Dalian Institute of Chemical Physics, Chinese Academy of Sciences, Dalian, China
| | - Tianya Liu
- Ministry of Education Key Laboratory of Protein Science, Tsinghua-Peking Center for Life Sciences, Beijing Advanced Innovation Center for Structural Biology, School of Life Sciences, Tsinghua University, Beijing, China
| | - Perrine Friedel
- Department of Cellular and Molecular Physiology, Yale University School of Medicine, New Haven, CT, USA
| | - Wei Zhuo
- Ministry of Education Key Laboratory of Protein Science, Tsinghua-Peking Center for Life Sciences, Beijing Advanced Innovation Center for Structural Biology, School of Life Sciences, Tsinghua University, Beijing, China
| | - Suma Somasekharan
- Department of Cellular and Molecular Physiology, Yale University School of Medicine, New Haven, CT, USA
| | - Kasturi Roy
- Department of Cellular and Molecular Physiology, Yale University School of Medicine, New Haven, CT, USA
| | - Laixing Zhang
- Ministry of Education Key Laboratory of Protein Science, Tsinghua-Peking Center for Life Sciences, Beijing Advanced Innovation Center for Structural Biology, School of Life Sciences, Tsinghua University, Beijing, China
| | - Yang Liu
- Ministry of Education Key Laboratory of Protein Science, Tsinghua-Peking Center for Life Sciences, Beijing Advanced Innovation Center for Structural Biology, School of Life Sciences, Tsinghua University, Beijing, China
| | - Xianbin Meng
- MOE Key Laboratory of Bioinformatics, School of Life Sciences, Tsinghua University, Beijing, China
| | - Haiteng Deng
- MOE Key Laboratory of Bioinformatics, School of Life Sciences, Tsinghua University, Beijing, China
| | - Wenwen Zeng
- Center for Life Sciences, Institute for Immunology and School of Medicine, Tsinghua University, Beijing, China
| | - Guohui Li
- State Key Laboratory of Molecular Reaction Dynamics, Dalian Institute of Chemical Physics, Chinese Academy of Sciences, Dalian, China.
| | - Biff Forbush
- Department of Cellular and Molecular Physiology, Yale University School of Medicine, New Haven, CT, USA.
| | - Maojun Yang
- Ministry of Education Key Laboratory of Protein Science, Tsinghua-Peking Center for Life Sciences, Beijing Advanced Innovation Center for Structural Biology, School of Life Sciences, Tsinghua University, Beijing, China.
- School of Pharmacy, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.
| |
Collapse
|
12
|
Intricacies of GABA A Receptor Function: The Critical Role of the β3 Subunit in Norm and Pathology. Int J Mol Sci 2021; 22:ijms22031457. [PMID: 33535681 PMCID: PMC7867123 DOI: 10.3390/ijms22031457] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2020] [Revised: 01/27/2021] [Accepted: 01/28/2021] [Indexed: 12/23/2022] Open
Abstract
Neuronal intracellular chloride ([Cl−]i) is a key determinant in γ-aminobutyric acid type A (GABA)ergic signaling. γ-Aminobutyric acid type A receptors (GABAARs) mediate both inhibitory and excitatory neurotransmission, as the passive fluxes of Cl− and HCO3− via pores can be reversed by changes in the transmembrane concentration gradient of Cl−. The cation–chloride co-transporters (CCCs) are the primary systems for maintaining [Cl−]i homeostasis. However, despite extensive electrophysiological data obtained in vitro that are supported by a wide range of molecular biological studies on the expression patterns and properties of CCCs, the presence of ontogenetic changes in [Cl−]i—along with the consequent shift in GABA reversal potential—remain a subject of debate. Recent studies showed that the β3 subunit possesses properties of the P-type ATPase that participates in the ATP-consuming movement of Cl− via the receptor. Moreover, row studies have demonstrated that the β3 subunit is a key player in GABAAR performance and in the appearance of serious neurological disorders. In this review, we discuss the properties and driving forces of CCCs and Cl−, HCO3−ATPase in the maintenance of [Cl−]i homeostasis after changes in upcoming GABAAR function. Moreover, we discuss the contribution of the β3 subunit in the manifestation of epilepsy, autism, and other syndromes.
Collapse
|
13
|
Hartmann AM, Fu L, Ziegler C, Winklhofer M, Nothwang HG. Structural changes in the extracellular loop 2 of the murine KCC2 potassium chloride cotransporter modulate ion transport. J Biol Chem 2021; 296:100793. [PMID: 34019872 PMCID: PMC8191313 DOI: 10.1016/j.jbc.2021.100793] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2020] [Revised: 05/11/2021] [Accepted: 05/14/2021] [Indexed: 01/22/2023] Open
Abstract
K+-Cl- cotransporters (KCCs) play important roles in physiological processes such as inhibitory neurotransmission and cell-volume regulation. KCCs exhibit significant variations in K+ affinities, yet recent atomic structures demonstrated that K+- and Cl--binding sites are highly conserved, raising the question of whether additional structural elements may contribute to ion coordination. The termini and the large extracellular domain (ECD) of KCCs exhibit only low sequence identity and were already discussed as modulators of transport activity. Here, we used the extracellular loop 2 (EL2) that links transmembrane helices (TMs) 3 and 4, as a mechanism to modulate ECD folding. We compared consequences of point mutations in the K+-binding site on the function of WT KCC2 and in a KCC2 variant, in which EL2 was structurally altered by insertion of a IFYPYDVPDYAGYPYDVPDYAGSYPYDVPDYAAHAAA (3xHA) tag (36 amino acids). In WT KCC2, individual mutations of five residues in the K+-binding site resulted in a 2- to 3-fold decreased transport rate. However, the same mutations in the KCC2 variant with EL2 structurally altered by insertion of a 3xHA tag had no effect on transport activity. Homology models of mouse KCC2 with the 3xHA tag inserted into EL2 using ab initio prediction were generated. The models suggest subtle conformational changes occur in the ECD upon EL2 modification. These data suggest that a conformational change in the ECD, for example, by interaction with EL2, might be an elegant way to modulate the K+ affinity of the different isoforms in the KCC subfamily.
Collapse
Affiliation(s)
- Anna-Maria Hartmann
- Division of Neurogenetics, School of Medicine and Health Sciences, Carl von Ossietzky University Oldenburg, Oldenburg, Germany; Research Center for Neurosensory Sciences, Carl von Ossietzky University Oldenburg, Oldenburg, Germany.
| | - Lifei Fu
- Biophysics II, Biophysics II-Structural Biology, Faculty of Biology and Pre-Clinical Medicine, University of Regensburg, Regensburg, Germany
| | - Christine Ziegler
- Biophysics II, Biophysics II-Structural Biology, Faculty of Biology and Pre-Clinical Medicine, University of Regensburg, Regensburg, Germany
| | - Michael Winklhofer
- Institute for Biology and Environmental Sciences IBU, Carl von Ossietzky University of Oldenburg, Oldenburg, Germany
| | - Hans Gerd Nothwang
- Division of Neurogenetics, School of Medicine and Health Sciences, Carl von Ossietzky University Oldenburg, Oldenburg, Germany; Research Center for Neurosensory Sciences, Carl von Ossietzky University Oldenburg, Oldenburg, Germany; Center of Excellence Hearing4all, Carl von Ossietzky University Oldenburg, Oldenburg, Germany
| |
Collapse
|
14
|
Smalley JL, Kontou G, Choi C, Ren Q, Albrecht D, Abiraman K, Santos MAR, Bope CE, Deeb TZ, Davies PA, Brandon NJ, Moss SJ. Isolation and Characterization of Multi-Protein Complexes Enriched in the K-Cl Co-transporter 2 From Brain Plasma Membranes. Front Mol Neurosci 2020; 13:563091. [PMID: 33192291 PMCID: PMC7643010 DOI: 10.3389/fnmol.2020.563091] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2020] [Accepted: 09/14/2020] [Indexed: 12/17/2022] Open
Abstract
Kcc2 plays a critical role in determining the efficacy of synaptic inhibition, however, the cellular mechanisms neurons use to regulate its membrane trafficking, stability and activity are ill-defined. To address these issues, we used affinity purification to isolate stable multi-protein complexes of K-Cl Co-transporter 2 (Kcc2) from the plasma membrane of murine forebrain. We resolved these using blue-native polyacrylamide gel electrophoresis (BN-PAGE) coupled to LC-MS/MS and label-free quantification. Data are available via ProteomeXchange with identifier PXD021368. Purified Kcc2 migrated as distinct molecular species of 300, 600, and 800 kDa following BN-PAGE. In excess of 90% coverage of the soluble N- and C-termini of Kcc2 was obtained. In total we identified 246 proteins significantly associated with Kcc2. The 300 kDa species largely contained Kcc2, which is consistent with a dimeric quaternary structure for this transporter. The 600 and 800 kDa species represented stable multi-protein complexes of Kcc2. We identified a set of novel structural, ion transporting, immune related and signaling protein interactors, that are present at both excitatory and inhibitory synapses, consistent with the proposed localization of Kcc2. These included spectrins, C1qa/b/c and the IP3 receptor. We also identified interactors more directly associated with phosphorylation; Akap5, Akap13, and Lmtk3. Finally, we used LC-MS/MS on the same purified endogenous plasma membrane Kcc2 to detect phosphorylation sites. We detected 11 sites with high confidence, including known and novel sites. Collectively our experiments demonstrate that Kcc2 is associated with components of the neuronal cytoskeleton and signaling molecules that may act to regulate transporter membrane trafficking, stability, and activity.
Collapse
Affiliation(s)
- Joshua L Smalley
- Department of Neuroscience, Tufts University School of Medicine, Boston, MA, United States
| | - Georgina Kontou
- Department of Neuroscience, Tufts University School of Medicine, Boston, MA, United States.,AstraZeneca Tufts Lab for Basic and Translational Neuroscience, Boston, MA, United States
| | - Catherine Choi
- Department of Neuroscience, Tufts University School of Medicine, Boston, MA, United States
| | - Qiu Ren
- Department of Neuroscience, Tufts University School of Medicine, Boston, MA, United States
| | - David Albrecht
- Department of Neuroscience, Tufts University School of Medicine, Boston, MA, United States.,AstraZeneca Tufts Lab for Basic and Translational Neuroscience, Boston, MA, United States
| | - Krithika Abiraman
- Department of Neuroscience, Tufts University School of Medicine, Boston, MA, United States.,AstraZeneca Tufts Lab for Basic and Translational Neuroscience, Boston, MA, United States
| | | | - Christopher E Bope
- Department of Neuroscience, Tufts University School of Medicine, Boston, MA, United States
| | - Tarek Z Deeb
- Department of Neuroscience, Tufts University School of Medicine, Boston, MA, United States.,AstraZeneca Tufts Lab for Basic and Translational Neuroscience, Boston, MA, United States
| | - Paul A Davies
- Department of Neuroscience, Tufts University School of Medicine, Boston, MA, United States
| | - Nicholas J Brandon
- AstraZeneca Tufts Lab for Basic and Translational Neuroscience, Boston, MA, United States.,Neuroscience, IMED Biotech Unit, AstraZeneca, Boston, MA, United States
| | - Stephen J Moss
- Department of Neuroscience, Tufts University School of Medicine, Boston, MA, United States.,Department of Neuroscience, Physiology, and Pharmacology, University College London, London, United Kingdom
| |
Collapse
|
15
|
Ferrini F, Perez-Sanchez J, Ferland S, Lorenzo LE, Godin AG, Plasencia-Fernandez I, Cottet M, Castonguay A, Wang F, Salio C, Doyon N, Merighi A, De Koninck Y. Differential chloride homeostasis in the spinal dorsal horn locally shapes synaptic metaplasticity and modality-specific sensitization. Nat Commun 2020; 11:3935. [PMID: 32769979 PMCID: PMC7414850 DOI: 10.1038/s41467-020-17824-y] [Citation(s) in RCA: 36] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2018] [Accepted: 07/22/2020] [Indexed: 02/06/2023] Open
Abstract
GABAA/glycine-mediated neuronal inhibition critically depends on intracellular chloride (Cl-) concentration which is mainly regulated by the K+-Cl- co-transporter 2 (KCC2) in the adult central nervous system (CNS). KCC2 heterogeneity thus affects information processing across CNS areas. Here, we uncover a gradient in Cl- extrusion capacity across the superficial dorsal horn (SDH) of the spinal cord (laminae I-II: LI-LII), which remains concealed under low Cl- load. Under high Cl- load or heightened synaptic drive, lower Cl- extrusion is unveiled in LI, as expected from the gradient in KCC2 expression found across the SDH. Blocking TrkB receptors increases KCC2 in LI, pointing to differential constitutive TrkB activation across laminae. Higher Cl- lability in LI results in rapidly collapsing inhibition, and a form of activity-dependent synaptic plasticity expressed as a continuous facilitation of excitatory responses. The higher metaplasticity in LI as compared to LII differentially affects sensitization to thermal and mechanical input. Thus, inconspicuous heterogeneity of Cl- extrusion across laminae critically shapes plasticity for selective nociceptive modalities.
Collapse
Affiliation(s)
- Francesco Ferrini
- Department of Veterinary Sciences, University of Turin, Turin, Italy.
- CERVO Brain Research Centre, Québec, QC, Canada.
- Department of Psychiatry and Neuroscience, Université Laval, Québec, QC, Canada.
- Graduate program in Neuroscience, Université Laval, Québec, QC, Canada.
| | - Jimena Perez-Sanchez
- CERVO Brain Research Centre, Québec, QC, Canada
- Graduate program in Neuroscience, Université Laval, Québec, QC, Canada
| | - Samuel Ferland
- CERVO Brain Research Centre, Québec, QC, Canada
- Graduate program in Neuroscience, Université Laval, Québec, QC, Canada
| | | | - Antoine G Godin
- CERVO Brain Research Centre, Québec, QC, Canada
- Department of Psychiatry and Neuroscience, Université Laval, Québec, QC, Canada
- Graduate program in Neuroscience, Université Laval, Québec, QC, Canada
| | - Isabel Plasencia-Fernandez
- CERVO Brain Research Centre, Québec, QC, Canada
- Graduate program in Neuroscience, Université Laval, Québec, QC, Canada
| | | | | | - Feng Wang
- CERVO Brain Research Centre, Québec, QC, Canada
| | - Chiara Salio
- Department of Veterinary Sciences, University of Turin, Turin, Italy
| | - Nicolas Doyon
- CERVO Brain Research Centre, Québec, QC, Canada
- Department of Mathematics and Statistics, Université Laval, Québec, QC, Canada
| | - Adalberto Merighi
- Department of Veterinary Sciences, University of Turin, Turin, Italy
| | - Yves De Koninck
- CERVO Brain Research Centre, Québec, QC, Canada
- Department of Psychiatry and Neuroscience, Université Laval, Québec, QC, Canada
- Graduate program in Neuroscience, Université Laval, Québec, QC, Canada
| |
Collapse
|
16
|
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
|
17
|
Dedek A, Xu J, Kandegedara CM, Lorenzo LÉ, Godin AG, De Koninck Y, Lombroso PJ, Tsai EC, Hildebrand ME. Loss of STEP61 couples disinhibition to N-methyl-d-aspartate receptor potentiation in rodent and human spinal pain processing. Brain 2020; 142:1535-1546. [PMID: 31135041 PMCID: PMC6536915 DOI: 10.1093/brain/awz105] [Citation(s) in RCA: 48] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2018] [Revised: 02/04/2019] [Accepted: 02/25/2019] [Indexed: 12/13/2022] Open
Abstract
Dysregulated excitability within the spinal dorsal horn is a critical mediator of chronic pain. In the rodent nerve injury model of neuropathic pain, BDNF-mediated loss of inhibition (disinhibition) gates the potentiation of excitatory GluN2B N-methyl-d-aspartate receptor (NMDAR) responses at lamina I dorsal horn synapses. However, the centrality of this mechanism across pain states and species, as well as the molecular linker involved, remain unknown. Here, we show that KCC2-dependent disinhibition is coupled to increased GluN2B-mediated synaptic NMDAR responses in a rodent model of inflammatory pain, with an associated downregulation of the tyrosine phosphatase STEP61. The decreased activity of STEP61 is both necessary and sufficient to prime subsequent phosphorylation and potentiation of GluN2B NMDAR by BDNF at lamina I synapses. Blocking disinhibition reversed the downregulation of STEP61 as well as inflammation-mediated behavioural hypersensitivity. For the first time, we characterize GluN2B-mediated NMDAR responses at human lamina I synapses and show that a human ex vivo BDNF model of pathological pain processing downregulates KCC2 and STEP61 and upregulates phosphorylated GluN2B at dorsal horn synapses. Our results demonstrate that STEP61 is the molecular brake that is lost following KCC2-dependent disinhibition and that the decrease in STEP61 activity drives the potentiation of excitatory GluN2B NMDAR responses in rodent and human models of pathological pain. The ex vivo human BDNF model may thus form a translational bridge between rodents and humans for identification and validation of novel molecular pain targets.
Collapse
Affiliation(s)
- Annemarie Dedek
- Department of Neuroscience, Carleton University, Ottawa, ON, Canada.,Neuroscience Program, Ottawa Hospital Research Institute, Ottawa, ON, Canada
| | - Jian Xu
- Child Study Center, Yale University School of Medicine, New Haven, CT, USA
| | - Chaya M Kandegedara
- Department of Neuroscience, Carleton University, Ottawa, ON, Canada.,Neuroscience Program, Ottawa Hospital Research Institute, Ottawa, ON, Canada
| | | | - Antoine G Godin
- CERVO Brain Research Centre, Quebec Mental Health Institute, Quebec, QC, Canada.,Department of Psychiatry and Neuroscience, Université Laval, Quebec, QC, Canada
| | - Yves De Koninck
- CERVO Brain Research Centre, Quebec Mental Health Institute, Quebec, QC, Canada.,Department of Psychiatry and Neuroscience, Université Laval, Quebec, QC, Canada.,Graduate Program in Neurobiology, Université Laval, Quebec, QC, Canada
| | - Paul J Lombroso
- Child Study Center, Yale University School of Medicine, New Haven, CT, USA
| | - Eve C Tsai
- Neuroscience Program, Ottawa Hospital Research Institute, Ottawa, ON, Canada.,Brain and Mind Research Institute, University of Ottawa, Ottawa, ON, Canada
| | - Michael E Hildebrand
- Department of Neuroscience, Carleton University, Ottawa, ON, Canada.,Neuroscience Program, Ottawa Hospital Research Institute, Ottawa, ON, Canada.,Brain and Mind Research Institute, University of Ottawa, Ottawa, ON, Canada
| |
Collapse
|
18
|
Lorenzo LE, Godin AG, Ferrini F, Bachand K, Plasencia-Fernandez I, Labrecque S, Girard AA, Boudreau D, Kianicka I, Gagnon M, Doyon N, Ribeiro-da-Silva A, De Koninck Y. Enhancing neuronal chloride extrusion rescues α2/α3 GABA A-mediated analgesia in neuropathic pain. Nat Commun 2020; 11:869. [PMID: 32054836 PMCID: PMC7018745 DOI: 10.1038/s41467-019-14154-6] [Citation(s) in RCA: 45] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2018] [Accepted: 12/16/2019] [Indexed: 02/06/2023] Open
Abstract
Spinal disinhibition has been hypothesized to underlie pain hypersensitivity in neuropathic pain. Apparently contradictory mechanisms have been reported, raising questions on the best target to produce analgesia. Here, we show that nerve injury is associated with a reduction in the number of inhibitory synapses in the spinal dorsal horn. Paradoxically, this is accompanied by a BDNF-TrkB-mediated upregulation of synaptic GABAARs and by an α1-to-α2GABAAR subunit switch, providing a mechanistic rationale for the analgesic action of the α2,3GABAAR benzodiazepine-site ligand L838,417 after nerve injury. Yet, we demonstrate that impaired Cl- extrusion underlies the failure of L838,417 to induce analgesia at high doses due to a resulting collapse in Cl- gradient, dramatically limiting the benzodiazepine therapeutic window. In turn, enhancing KCC2 activity not only potentiated L838,417-induced analgesia, it rescued its analgesic potential at high doses, revealing a novel strategy for analgesia in pathological pain, by combined targeting of the appropriate GABAAR-subtypes and restoring Cl- homeostasis.
Collapse
Affiliation(s)
- Louis-Etienne Lorenzo
- CERVO Brain Research Centre, Quebec Mental Health Institute, Québec, QC, Canada
- Department of Pharmacology & Therapeutics, McGill University, Montreal, QC, Canada
| | - Antoine G Godin
- CERVO Brain Research Centre, Quebec Mental Health Institute, Québec, QC, Canada
- Department of Psychiatry & Neuroscience, Université Laval, Québec, QC, Canada
- Graduate program in Neuroscience, Université Laval, Québec, QC, Canada
| | - Francesco Ferrini
- CERVO Brain Research Centre, Quebec Mental Health Institute, Québec, QC, Canada
- Department of Psychiatry & Neuroscience, Université Laval, Québec, QC, Canada
- Graduate program in Neuroscience, Université Laval, Québec, QC, Canada
- Department of Veterinary Sciences, University of Turin, Turin, Italy
| | - Karine Bachand
- CERVO Brain Research Centre, Quebec Mental Health Institute, Québec, QC, Canada
| | - Isabel Plasencia-Fernandez
- CERVO Brain Research Centre, Quebec Mental Health Institute, Québec, QC, Canada
- Graduate program in Neuroscience, Université Laval, Québec, QC, Canada
| | - Simon Labrecque
- CERVO Brain Research Centre, Quebec Mental Health Institute, Québec, QC, Canada
| | - Alexandre A Girard
- CERVO Brain Research Centre, Quebec Mental Health Institute, Québec, QC, Canada
- Ecole Polytechnique, IP Paris, Palaiseau, France
| | - Dominic Boudreau
- CERVO Brain Research Centre, Quebec Mental Health Institute, Québec, QC, Canada
- Graduate program in Neuroscience, Université Laval, Québec, QC, Canada
| | - Irenej Kianicka
- Chlorion Pharma, Laval, Québec, QC, Canada
- Laurent Pharmaceuticals Inc., Montreal, QC, Canada
| | - Martin Gagnon
- CERVO Brain Research Centre, Quebec Mental Health Institute, Québec, QC, Canada
- Centre for Innovation, University of Otago, Dunedin, New Zealand
| | - Nicolas Doyon
- CERVO Brain Research Centre, Quebec Mental Health Institute, Québec, QC, Canada
- Finite Element Interdisciplinary Research Group (GIREF), Université Laval, Québec, QC, Canada
| | - Alfredo Ribeiro-da-Silva
- Department of Pharmacology & Therapeutics, McGill University, Montreal, QC, Canada
- Department of Anatomy & Cell Biology, McGill University, Montreal, QC, Canada
- Alan Edwards Centre for Research on Pain, McGill University, Montreal, QC, Canada
| | - Yves De Koninck
- CERVO Brain Research Centre, Quebec Mental Health Institute, Québec, QC, Canada.
- Department of Pharmacology & Therapeutics, McGill University, Montreal, QC, Canada.
- Department of Psychiatry & Neuroscience, Université Laval, Québec, QC, Canada.
- Graduate program in Neuroscience, Université Laval, Québec, QC, Canada.
- Alan Edwards Centre for Research on Pain, McGill University, Montreal, QC, Canada.
| |
Collapse
|
19
|
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]
|
20
|
Plantier V, Sanchez-Brualla I, Dingu N, Brocard C, Liabeuf S, Gackière F, Brocard F. Calpain fosters the hyperexcitability of motoneurons after spinal cord injury and leads to spasticity. eLife 2019; 8:e51404. [PMID: 31815668 PMCID: PMC6927741 DOI: 10.7554/elife.51404] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2019] [Accepted: 12/08/2019] [Indexed: 12/12/2022] Open
Abstract
Up-regulation of the persistent sodium current (INaP) and down-regulation of the potassium/chloride extruder KCC2 lead to spasticity after spinal cord injury (SCI). We here identified calpain as the driver of the up- and down-regulation of INaP and KCC2, respectively, in neonatal rat lumbar motoneurons. Few days after SCI, neonatal rats developed behavioral signs of spasticity with the emergence of both hyperreflexia and abnormal involuntary muscle contractions on hindlimbs. At the same time, in vitro isolated lumbar spinal cords became hyperreflexive and displayed numerous spontaneous motor outputs. Calpain-I expression paralleled with a proteolysis of voltage-gated sodium (Nav) channels and KCC2. Acute inhibition of calpains reduced this proteolysis, restored the motoneuronal expression of Nav and KCC2, normalized INaP and KCC2 function, and curtailed spasticity. In sum, by up- and down-regulating INaP and KCC2, the calpain-mediated proteolysis of Nav and KCC2 drives the hyperexcitability of motoneurons which leads to spasticity after SCI.
Collapse
Affiliation(s)
- Vanessa Plantier
- Institut de Neurosciences de la Timone (UMR7289), Aix-Marseille Université and CNRSMarseilleFrance
| | - Irene Sanchez-Brualla
- Institut de Neurosciences de la Timone (UMR7289), Aix-Marseille Université and CNRSMarseilleFrance
| | - Nejada Dingu
- Institut de Neurosciences de la Timone (UMR7289), Aix-Marseille Université and CNRSMarseilleFrance
| | - Cécile Brocard
- Institut de Neurosciences de la Timone (UMR7289), Aix-Marseille Université and CNRSMarseilleFrance
| | - Sylvie Liabeuf
- Institut de Neurosciences de la Timone (UMR7289), Aix-Marseille Université and CNRSMarseilleFrance
| | - Florian Gackière
- Institut de Neurosciences de la Timone (UMR7289), Aix-Marseille Université and CNRSMarseilleFrance
| | - Frédéric Brocard
- Institut de Neurosciences de la Timone (UMR7289), Aix-Marseille Université and CNRSMarseilleFrance
| |
Collapse
|
21
|
Duy PQ, David WB, Kahle KT. Identification of KCC2 Mutations in Human Epilepsy Suggests Strategies for Therapeutic Transporter Modulation. Front Cell Neurosci 2019; 13:515. [PMID: 31803025 PMCID: PMC6873151 DOI: 10.3389/fncel.2019.00515] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2019] [Accepted: 11/01/2019] [Indexed: 11/28/2022] Open
Abstract
Epilepsy is a common neurological disorder characterized by recurrent and unprovoked seizures thought to arise from impaired balance between neuronal excitation and inhibition. Our understanding of the neurophysiological mechanisms that render the brain epileptogenic remains incomplete, reflected by the lack of satisfactory treatments that can effectively prevent epileptic seizures without significant drug-related adverse effects. Type 2 K+-Cl− cotransporter (KCC2), encoded by SLC12A5, is important for chloride homeostasis and neuronal excitability. KCC2 dysfunction attenuates Cl− extrusion and impairs GABAergic inhibition, and can lead to neuronal hyperexcitability. Converging lines of evidence from human genetics have secured the link between KCC2 dysfunction and the development of epilepsy. Here, we review KCC2 mutations in human epilepsy and discuss potential therapeutic strategies based on the functional impact of these mutations. We suggest that a strategy of augmenting KCC2 activity by antagonizing its critical inhibitory phosphorylation sites may be a particularly efficacious method of facilitating Cl− extrusion and restoring GABA inhibition to treat medication-refractory epilepsy and other seizure disorders.
Collapse
Affiliation(s)
- Phan Q Duy
- Department of Neurosurgery, Yale University School of Medicine, New Haven, CT, United States.,Medical Scientist Training Program, Yale University School of Medicine, New Haven, CT, United States
| | - Wyatt B David
- Department of Neurosurgery, Yale University School of Medicine, New Haven, CT, United States
| | - Kristopher T Kahle
- Department of Neurosurgery, Yale University School of Medicine, New Haven, CT, United States.,Department of Genetics, Yale University School of Medicine, New Haven, CT, United States.,Departments of Pediatrics and Cellular & Molecular Physiology, Yale University School of Medicine, New Haven, CT, United States.,Yale-Rockefeller NIH Centers for Mendelian Genomics, Yale University, New Haven, CT, United States.,Yale Stem Cell Center, Yale School of Medicine, New Haven, CT, United States
| |
Collapse
|
22
|
Huang H, Song S, Banerjee S, Jiang T, Zhang J, Kahle KT, Sun D, Zhang Z. The WNK-SPAK/OSR1 Kinases and the Cation-Chloride Cotransporters as Therapeutic Targets for Neurological Diseases. Aging Dis 2019; 10:626-636. [PMID: 31165006 PMCID: PMC6538211 DOI: 10.14336/ad.2018.0928] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2018] [Accepted: 09/28/2018] [Indexed: 02/05/2023] Open
Abstract
In recent years, cation-chloride cotransporters (CCCs) have drawn attention in the medical neuroscience research. CCCs include the family of Na+-coupled Cl- importers (NCC, NKCC1, and NKCC2), K+-coupled Cl- exporters (KCCs), and possibly polyamine transporters (CCC9) and CCC interacting protein (CIP1). For decades, CCCs have been the targets of several commonly used diuretic drugs, including hydrochlorothiazide, furosemide, and bumetanide. Genetic mutations of NCC and NKCC2 cause congenital renal tubular disorders and lead to renal salt-losing hypotension, secondary hyperreninemia, and hypokalemic metabolic alkalosis. New studies reveal that CCCs along with their regulatory WNK (Kinase with no lysine (K)), and SPAK (Ste20-related proline-alanine-rich kinase)/OSR1(oxidative stress-responsive kinase-1) are essential for regulating cell volume and maintaining ionic homeostasis in the nervous system, especially roles of the WNK-SPAK-NKCC1 signaling pathway in ischemic brain injury and hypersecretion of cerebrospinal fluid in post-hemorrhagic hydrocephalus. In addition, disruption of Cl- exporter KCC2 has an effect on synaptic inhibition, which may be involved in developing pain, epilepsy, and possibly some neuropsychiatric disorders. Interference with KCC3 leads to peripheral nervous system neuropathy as well as axon and nerve fiber swelling and psychosis. The WNK-SPAK/OSR1-CCCs complex emerges as therapeutic targets for multiple neurological diseases. This review will highlight these new findings.
Collapse
Affiliation(s)
- Huachen Huang
- Department of Neurology, The First Affiliate Hospital, Harbin Medical University, Harbin, Heilongjiang, China.
- Department of Neurology, University of Pittsburgh, Pittsburgh, PA, USA.
| | - Shanshan Song
- Department of Neurology, University of Pittsburgh, Pittsburgh, PA, USA.
| | - Suneel Banerjee
- Department of Neurology, University of Pittsburgh, Pittsburgh, PA, USA.
| | - Tong Jiang
- Department of Neurology, University of Pittsburgh, Pittsburgh, PA, USA.
| | - Jinwei Zhang
- Institute of Biomedical and Clinical Sciences, University of Exeter Medical School, Hatherly Laboratory, Exeter, EX4 4PS, UK.
| | - Kristopher T. Kahle
- Departments of Neurosurgery, Pediatrics, and Cellular & Molecular Physiology, Centers for Mendelian Genomics, Yale School of Medicine, New Haven, CT, USA.
| | - Dandan Sun
- Department of Neurology, University of Pittsburgh, Pittsburgh, PA, USA.
- Veterans Affairs Pittsburgh Health Care System, Geriatric Research, Education and Clinical Center, Pittsburgh, PA, USA.
| | - Zhongling Zhang
- Department of Neurology, The First Affiliate Hospital, Harbin Medical University, Harbin, Heilongjiang, China.
| |
Collapse
|
23
|
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
|
24
|
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
|
25
|
Delpire E, Gagnon KB. Water Homeostasis and Cell Volume Maintenance and Regulation. CURRENT TOPICS IN MEMBRANES 2018; 81:3-52. [PMID: 30243436 PMCID: PMC6457474 DOI: 10.1016/bs.ctm.2018.08.001] [Citation(s) in RCA: 52] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
From early unicellular organisms that formed in salty water environments to complex organisms that live on land away from water, cells have had to protect a homeostatic internal environment favorable to the biochemical reactions necessary for life. In this chapter, we will outline what steps were necessary to conserve the water within our cells and how mechanisms have evolved to maintain and regulate our cellular and organismal volume. We will first examine whole body water homeostasis and the relationship between kidney function, regulation of blood pressure, and blood filtration in the process of producing urine. We will then discuss how the composition of the lipid-rich bilayer affects its permeability to water and salts, and how the cell uses this differential to drive physiological and biochemical cellular functions. The capacity to maintain cell volume is vital to epithelial transport, neurotransmission, cell cycle, apoptosis, and cell migration. Finally, we will wrap up the chapter by discussing in some detail specific channels, cotransporters, and exchangers that have evolved to facilitate the movement of cations and anions otherwise unable to cross the lipid-rich bilayer and that are involved in maintaining or regulating cell volume.
Collapse
Affiliation(s)
- Eric Delpire
- Department of Anesthesiology, Vanderbilt University School of Medicine
| | | |
Collapse
|
26
|
Kelley MR, Cardarelli RA, Smalley JL, Ollerhead TA, Andrew PM, Brandon NJ, Deeb TZ, Moss SJ. Locally Reducing KCC2 Activity in the Hippocampus is Sufficient to Induce Temporal Lobe Epilepsy. EBioMedicine 2018; 32:62-71. [PMID: 29884458 PMCID: PMC6020795 DOI: 10.1016/j.ebiom.2018.05.029] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2018] [Revised: 05/22/2018] [Accepted: 05/23/2018] [Indexed: 12/03/2022] Open
Abstract
Mesial temporal lobe epilepsy (mTLE) is the most common form of epilepsy, believed to arise in part from compromised GABAergic inhibition. The neuronal specific K+/Cl- co-transporter 2 (KCC2) is a critical determinant of the efficacy of GABAergic inhibition and deficits in its activity are observed in mTLE patients and animal models of epilepsy. To test if reductions of KCC2 activity directly contribute to the pathophysiology of mTLE, we locally ablated KCC2 expression in a subset of principal neurons within the adult hippocampus. Deletion of KCC2 resulted in compromised GABAergic inhibition and the development of spontaneous, recurrent generalized seizures. Moreover, local ablation of KCC2 activity resulted in hippocampal sclerosis, a key pathological change seen in mTLE. Collectively, our results demonstrate that local deficits in KCC2 activity within the hippocampus are sufficient to precipitate mTLE.
Collapse
Affiliation(s)
- Matt R Kelley
- Department of Neuroscience, Tufts University School of Medicine, Boston, MA, USA
| | - Ross A Cardarelli
- AstraZeneca-Tufts University Laboratory for Basic and Translational Neuroscience Research, Boston, MA, USA
| | - Joshua L Smalley
- Department of Neuroscience, Tufts University School of Medicine, Boston, MA, USA
| | - Thomas A Ollerhead
- Department of Neuroscience, Tufts University School of Medicine, Boston, MA, USA
| | - Peter M Andrew
- Department of Neuroscience, Tufts University School of Medicine, Boston, MA, USA
| | - Nicholas J Brandon
- AstraZeneca-Tufts University Laboratory for Basic and Translational Neuroscience Research, Boston, MA, USA; Neuroscience, IMED Biotech Unit, AstraZeneca, Boston, MA, USA
| | - Tarek Z Deeb
- Department of Neuroscience, Tufts University School of Medicine, Boston, MA, USA; AstraZeneca-Tufts University Laboratory for Basic and Translational Neuroscience Research, Boston, MA, USA
| | - Stephen J Moss
- Department of Neuroscience, Tufts University School of Medicine, Boston, MA, USA; AstraZeneca-Tufts University Laboratory for Basic and Translational Neuroscience Research, Boston, MA, USA; Neuroscience, IMED Biotech Unit, AstraZeneca, Boston, MA, USA; Department of Neuroscience, Physiology and Pharmacology, University College, London, WC1E, 6BT, UK.
| |
Collapse
|
27
|
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
|
28
|
Markkanen M, Ludwig A, Khirug S, Pryazhnikov E, Soni S, Khiroug L, Delpire E, Rivera C, Airaksinen MS, Uvarov P. Implications of the N-terminal heterogeneity for the neuronal K-Cl cotransporter KCC2 function. Brain Res 2017; 1675:87-101. [PMID: 28888841 DOI: 10.1016/j.brainres.2017.08.034] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2017] [Revised: 08/25/2017] [Accepted: 08/30/2017] [Indexed: 12/16/2022]
Abstract
The neuron-specific K-Cl cotransporter KCC2 maintains the low intracellular chloride concentration required for the fast hyperpolarizing responses of the inhibitory neurotransmitters γ-aminobutyric acid (GABA) and glycine. The two KCC2 isoforms, KCC2a and KCC2b differ by their N-termini as a result of alternative promoter usage. Whereas the role of KCC2b in mediating the chloride transport is unequivocal, the physiological role of KCC2a in neurons has remained obscure. We show that KCC2a isoform can decrease the intracellular chloride concentration in cultured neurons and attenuate calcium responses evoked by application of the GABAA receptor agonist muscimol. While the biotinylation assay detected both KCC2 isoforms at the cell surface of cultured neurons, KCC2a was not detected at the plasma membrane in immunostainings, suggesting that the N-terminal KCC2a epitope is masked. Confirming this hypothesis, KCC2a surface expression was detected by the C-terminal KCC2 pan antibody but not by the N-terminal KCC2a antibody in KCC2b-deficient neurons. One possible cause for the epitope masking is the binding site of Ste20-related proline-alanine-rich kinase (SPAK) in the KCC2a N-terminus. SPAK, a known regulator of K-Cl cotransporters, was co-immunoprecipitated in a complex with KCC2a but not KCC2b isoform. Moreover, SPAK overexpression decreased the transport activity of KCC2a but not that of KCC2b, as revealed by rubidium flux assay in HEK293 cells. Thus, our data indicate that both KCC2 isoforms perform as chloride cotransporters in neuronal cells, while their N-terminal heterogeneity could play an important role in fine-tuning of the K-Cl transport activity.
Collapse
Affiliation(s)
- Marika Markkanen
- Department of Anatomy, Faculty of Medicine, University of Helsinki, Helsinki, Finland
| | | | | | | | - Shetal Soni
- Neuroscience Center, University of Helsinki, Helsinki, Finland
| | - Leonard Khiroug
- Neuroscience Center, University of Helsinki, Helsinki, Finland
| | - Eric Delpire
- Department of Anesthesiology, Vanderbilt University School of Medicine, Nashville, TN, USA
| | - Claudio Rivera
- Neuroscience Center, University of Helsinki, Helsinki, Finland; INSERM, Institut de Neurobiologie de la Méditerranée (INMED), Marseille, France; Aix-Marseille Université, UMR901 Marseille, France
| | - Matti S Airaksinen
- Department of Anatomy, Faculty of Medicine, University of Helsinki, Helsinki, Finland.
| | - Pavel Uvarov
- Department of Anatomy, Faculty of Medicine, University of Helsinki, Helsinki, Finland.
| |
Collapse
|
29
|
Klett NJ, Allen CN. Intracellular Chloride Regulation in AVP+ and VIP+ Neurons of the Suprachiasmatic Nucleus. Sci Rep 2017; 7:10226. [PMID: 28860458 PMCID: PMC5579040 DOI: 10.1038/s41598-017-09778-x] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2017] [Accepted: 07/28/2017] [Indexed: 12/15/2022] Open
Abstract
Several reports have described excitatory GABA transmission in the suprachiasmatic nucleus (SCN), the master pacemaker of circadian physiology. However, there is disagreement regarding the prevalence, timing, and neuronal location of excitatory GABA transmission in the SCN. Whether GABA is inhibitory or excitatory depends, in part, on the intracellular concentration of chloride ([Cl-]i). Here, using ratiometric Cl- imaging, we have investigated intracellular chloride regulation in AVP and VIP-expressing SCN neurons and found evidence suggesting that [Cl-]i is higher during the day than during the night in both AVP+ and VIP+ neurons. We then investigated the contribution of the cation chloride cotransporters to setting [Cl-]i in these SCN neurons and found that the chloride uptake transporter NKCC1 contributes to [Cl-]i regulation in SCN neurons, but that the KCCs are the primary regulators of [Cl-]i in SCN neurons. Interestingly, we observed that [Cl-]i is differentially regulated between AVP+ and VIP+ neurons-a low concentration of the loop diuretic bumetanide had differential effects on AVP+ and VIP+ neurons, while blocking the KCCs with VU0240551 had a larger effect on VIP+ neurons compared to AVP+ neurons.
Collapse
Affiliation(s)
- Nathan J Klett
- Neuroscience Graduate Program, School of Medicine, Oregon Health & Science University, Portland, OR, 97239, USA
- Oregon Institute for Occupational Health Sciences, School of Medicine, Oregon Health & Science University, Portland, OR, 97239, USA
| | - Charles N Allen
- Oregon Institute for Occupational Health Sciences, School of Medicine, Oregon Health & Science University, Portland, OR, 97239, USA.
- Department of Behavioral Neuroscience, School of Medicine, Oregon Health & Science University, Portland, OR, 97239, USA.
| |
Collapse
|
30
|
Plantier V, Brocard F. [Calpain as a new therapeutic target for treating spasticity after a spinal cord injury]. Med Sci (Paris) 2017; 33:629-636. [PMID: 28990565 DOI: 10.1051/medsci/20173306020] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/14/2022] Open
Abstract
After a spinal cord injury (SCI), patients develop spasticity, a motor disorder characterized by hyperreflexia and stiffness of muscles. Spasticity results from alterations in motoneurons with an upregulation of their persistent sodium current (I NaP), simultaneously with a disinhibition caused by a reduction of expression of chloride (Cl-) co-transporters KCC2. Until recently the origin of alterations was unknown. After reviewing pathophysiology of spasticity, the manuscript relates our recent work showing a tight relationship between the calpain-dependent proteolysis of voltage-gated sodium channels, the upregulation of I NaP and spasticity following SCI. We also discuss KCC2 as a substrate of calpains which may contribute to the disinhibition of motoneurons below the lesion. This led us to consider the proteolytic cleavage of both sodium channels and KCC2 as the upstream mechanism contributing to the development of spasticity after SCI.
Collapse
Affiliation(s)
- Vanessa Plantier
- Équipe P3M, Institut de Neurosciences de la Timone, UMR7289, Aix Marseille Université et Centre National de la Recherche Scientifique (CNRS), Marseille, France
| | - Frédéric Brocard
- Équipe P3M, Institut de Neurosciences de la Timone, UMR7289, Aix Marseille Université et Centre National de la Recherche Scientifique (CNRS), Marseille, France
| |
Collapse
|
31
|
Ferrini F, Lorenzo LE, Godin AG, Quang ML, De Koninck Y. Enhancing KCC2 function counteracts morphine-induced hyperalgesia. Sci Rep 2017. [PMID: 28634406 PMCID: PMC5478677 DOI: 10.1038/s41598-017-04209-3] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
Morphine-induced hyperalgesia (MIH) is a severe adverse effect accompanying repeated morphine treatment, causing a paradoxical decrease in nociceptive threshold. Previous reports associated MIH with a decreased expression of the Cl− extruder KCC2 in the superficial dorsal horn (SDH) of the spinal cord, weakening spinal GABAA/glycine-mediated postsynaptic inhibition. Here, we tested whether the administration of small molecules enhancing KCC2, CLP257 and its pro-drug CLP290, may counteract MIH. MIH was typically expressed within 6–8 days of morphine treatment. Morphine-treated rats exhibited decreased withdrawal threshold to mechanical stimulation and increased vocalizing behavior to subcutaneous injections. Chloride extrusion was impaired in SDH neurons measured as a depolarizing shift in EGABA under Cl− load. Delivering CLP257 to spinal cord slices obtained from morphine-treated rats was sufficient to restore Cl− extrusion capacity in SDH neurons. In vivo co-treatment with morphine and oral CLP290 prevented membrane KCC2 downregulation in SDH neurons. Concurrently, co-treatment with CLP290 significantly mitigated MIH and acute administration of CLP257 in established MIH restored normal nociceptive behavior. Our data indicate that enhancing KCC2 activity is a viable therapeutic approach for counteracting MIH. Chloride extrusion enhancers may represent an effective co-adjuvant therapy to improve morphine analgesia by preventing and reversing MIH.
Collapse
Affiliation(s)
- Francesco Ferrini
- Department of Veterinary Sciences, University of Turin, Turin, Italy.,CERVO Brain Research Centre, Institut universitaire en santé mentale de Québec, Québec, Canada
| | - Louis-Etienne Lorenzo
- CERVO Brain Research Centre, Institut universitaire en santé mentale de Québec, Québec, Canada
| | - Antoine G Godin
- CERVO Brain Research Centre, Institut universitaire en santé mentale de Québec, Québec, Canada
| | - Miorie Le Quang
- CERVO Brain Research Centre, Institut universitaire en santé mentale de Québec, Québec, Canada
| | - Yves De Koninck
- CERVO Brain Research Centre, Institut universitaire en santé mentale de Québec, Québec, Canada. .,Department of Psychiatry and Neuroscience, Université Laval, Québec, Canada.
| |
Collapse
|
32
|
Kursan S, McMillen TS, Beesetty P, Dias-Junior E, Almutairi MM, Sajib AA, Kozak JA, Aguilar-Bryan L, Di Fulvio M. The neuronal K +Cl - co-transporter 2 (Slc12a5) modulates insulin secretion. Sci Rep 2017; 7:1732. [PMID: 28496181 PMCID: PMC5431760 DOI: 10.1038/s41598-017-01814-0] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2016] [Accepted: 04/03/2017] [Indexed: 11/09/2022] Open
Abstract
Intracellular chloride concentration ([Cl-]i) in pancreatic β-cells is kept above electrochemical equilibrium due to the predominant functional presence of Cl- loaders such as the Na+K+2Cl- co-transporter 1 (Slc12a2) over Cl-extruders of unidentified nature. Using molecular cloning, RT-PCR, Western blotting, immunolocalization and in vitro functional assays, we establish that the "neuron-specific" K+Cl- co-transporter 2 (KCC2, Slc12a5) is expressed in several endocrine cells of the pancreatic islet, including glucagon secreting α-cells, but particularly in insulin-secreting β-cells, where we provide evidence for its role in the insulin secretory response. Three KCC2 splice variants were identified: the formerly described KCC2a and KCC2b along with a novel one lacking exon 25 (KCC2a-S25). This new variant is undetectable in brain or spinal cord, the only and most abundant known sources of KCC2. Inhibition of KCC2 activity in clonal MIN6 β-cells increases basal and glucose-stimulated insulin secretion and Ca2+ uptake in the presence of glibenclamide, an inhibitor of the ATP-dependent potassium (KATP)-channels, thus suggesting a possible mechanism underlying KCC2-dependent insulin release. We propose that the long-time considered "neuron-specific" KCC2 co-transporter is expressed in pancreatic islet β-cells where it modulates Ca2+-dependent insulin secretion.
Collapse
Affiliation(s)
- Shams Kursan
- Department of Pharmacology and Toxicology, Wright State University, School of Medicine, Dayton, OH, 45435, USA
| | | | - Pavani Beesetty
- Department of Neuroscience, Cell Biology and Physiology, Wright State University, School of Medicine, Dayton, OH, 45435, USA
| | - Eduardo Dias-Junior
- Department of Pharmacology and Toxicology, Wright State University, School of Medicine, Dayton, OH, 45435, USA
| | - Mohammed M Almutairi
- Department of Pharmacology and Toxicology, Wright State University, School of Medicine, Dayton, OH, 45435, USA
| | - Abu A Sajib
- Department of Genetic Engineering and Biotechnology, University of Dhaka, Dhaka, Bangladesh
| | - J Ashot Kozak
- Department of Neuroscience, Cell Biology and Physiology, Wright State University, School of Medicine, Dayton, OH, 45435, USA
| | | | - Mauricio Di Fulvio
- Department of Pharmacology and Toxicology, Wright State University, School of Medicine, Dayton, OH, 45435, USA.
| |
Collapse
|
33
|
Albers HE, Walton JC, Gamble KL, McNeill JK, Hummer DL. The dynamics of GABA signaling: Revelations from the circadian pacemaker in the suprachiasmatic nucleus. Front Neuroendocrinol 2017; 44:35-82. [PMID: 27894927 PMCID: PMC5225159 DOI: 10.1016/j.yfrne.2016.11.003] [Citation(s) in RCA: 72] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/30/2016] [Revised: 10/16/2016] [Accepted: 11/22/2016] [Indexed: 12/31/2022]
Abstract
Virtually every neuron within the suprachiasmatic nucleus (SCN) communicates via GABAergic signaling. The extracellular levels of GABA within the SCN are determined by a complex interaction of synthesis and transport, as well as synaptic and non-synaptic release. The response to GABA is mediated by GABAA receptors that respond to both phasic and tonic GABA release and that can produce excitatory as well as inhibitory cellular responses. GABA also influences circadian control through the exclusively inhibitory effects of GABAB receptors. Both GABA and neuropeptide signaling occur within the SCN, although the functional consequences of the interactions of these signals are not well understood. This review considers the role of GABA in the circadian pacemaker, in the mechanisms responsible for the generation of circadian rhythms, in the ability of non-photic stimuli to reset the phase of the pacemaker, and in the ability of the day-night cycle to entrain the pacemaker.
Collapse
Affiliation(s)
- H Elliott Albers
- Center for Behavioral Neuroscience, Atlanta, GA 30302, United States; Neuroscience Institute, Georgia State University, Atlanta, GA 30302, United States.
| | - James C Walton
- Center for Behavioral Neuroscience, Atlanta, GA 30302, United States; Neuroscience Institute, Georgia State University, Atlanta, GA 30302, United States
| | - Karen L Gamble
- Department of Psychiatry and Behavioral Neurobiology, University of Alabama at Birmingham, Birmingham, AL 35294, United States
| | - John K McNeill
- Center for Behavioral Neuroscience, Atlanta, GA 30302, United States; Neuroscience Institute, Georgia State University, Atlanta, GA 30302, United States
| | - Daniel L Hummer
- Center for Behavioral Neuroscience, Atlanta, GA 30302, United States; Department of Psychology, Morehouse College, Atlanta, GA 30314, United States
| |
Collapse
|
34
|
Mercado A, de Los Heros P, Melo Z, Chávez-Canales M, Murillo-de-Ozores AR, Moreno E, Bazúa-Valenti S, Vázquez N, Hadchouel J, Gamba G. With no lysine L-WNK1 isoforms are negative regulators of the K+-Cl- cotransporters. Am J Physiol Cell Physiol 2016; 311:C54-66. [PMID: 27170636 PMCID: PMC4967140 DOI: 10.1152/ajpcell.00193.2015] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2015] [Accepted: 05/02/2016] [Indexed: 12/11/2022]
Abstract
The K(+)-Cl(-) cotransporters (KCC1-KCC4) encompass a branch of the SLC12 family of electroneutral cation-coupled chloride cotransporters that translocate ions out of the cell to regulate various factors, including cell volume and intracellular chloride concentration, among others. L-WNK1 is an ubiquitously expressed kinase that is activated in response to osmotic stress and intracellular chloride depletion, and it is implicated in two distinct hereditary syndromes: the renal disease pseudohypoaldosteronism type II (PHAII) and the neurological disease hereditary sensory neuropathy 2 (HSN2). The effect of L-WNK1 on KCC activity is unknown. Using Xenopus laevis oocytes and HEK-293 cells, we show that the activation of KCCs by cell swelling was prevented by L-WNK1 coexpression. In contrast, the activity of the Na(+)-K(+)-2Cl(-) cotransporter NKCC1 was remarkably increased with L-WNK1 coexpression. The negative effect of L-WNK1 on the KCCs is kinase dependent. Elimination of the STE20 proline-alanine rich kinase (SPAK)/oxidative stress-responsive kinase (OSR1) binding site or the HQ motif required for the WNK-WNK interaction prevented the effect of L-WNK1 on KCCs, suggesting a required interaction between L-WNK1 molecules and SPAK. Together, our data support that NKCC1 and KCCs are coordinately regulated by L-WNK1 isoforms.
Collapse
Affiliation(s)
- Adriana Mercado
- Department of Nephrology, Instituto Nacional de Cardiología Ignacio Chávez, Tlalpan, Mexico City, Mexico
| | - Paola de Los Heros
- División de Investigación, Facultad de Medicina, Universidad Nacional Autónoma de México, Coyoacán, Mexico City, Mexico
| | - Zesergio Melo
- Molecular Physiology Unit, Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México, Tlalpan, Mexico City, Mexico
| | - María Chávez-Canales
- Molecular Physiology Unit, Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México, Tlalpan, Mexico City, Mexico; Department of Nephrology and Mineral Metabolism, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán. Tlalpan, Mexico City, Mexico; INSERM UMR970-Paris Cardiovascular Research Center, Paris, France; and University Paris-Descartes, Sorbonne Paris Cité, Faculty of Medicine, Paris, France
| | - Adrián R Murillo-de-Ozores
- Molecular Physiology Unit, Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México, Tlalpan, Mexico City, Mexico
| | - Erika Moreno
- Department of Nephrology and Mineral Metabolism, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán. Tlalpan, Mexico City, Mexico
| | - Silvana Bazúa-Valenti
- Molecular Physiology Unit, Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México, Tlalpan, Mexico City, Mexico; Department of Nephrology and Mineral Metabolism, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán. Tlalpan, Mexico City, Mexico
| | - Norma Vázquez
- Molecular Physiology Unit, Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México, Tlalpan, Mexico City, Mexico; Department of Nephrology and Mineral Metabolism, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán. Tlalpan, Mexico City, Mexico
| | - Juliette Hadchouel
- INSERM UMR970-Paris Cardiovascular Research Center, Paris, France; and University Paris-Descartes, Sorbonne Paris Cité, Faculty of Medicine, Paris, France
| | - Gerardo Gamba
- Molecular Physiology Unit, Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México, Tlalpan, Mexico City, Mexico; Department of Nephrology and Mineral Metabolism, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán. Tlalpan, Mexico City, Mexico;
| |
Collapse
|
35
|
Wu H, Che X, Tang J, Ma F, Pan K, Zhao M, Shao A, Wu Q, Zhang J, Hong Y. The K(+)-Cl(-) Cotransporter KCC2 and Chloride Homeostasis: Potential Therapeutic Target in Acute Central Nervous System Injury. Mol Neurobiol 2016; 53:2141-51. [PMID: 25941074 DOI: 10.1007/s12035-015-9162-x] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2014] [Accepted: 03/26/2015] [Indexed: 12/11/2022]
Abstract
The K(+)-Cl(-) cotransporter-2 (KCC2) is a well-known member of the electroneutral cation-chloride cotransporters with a restricted expression pattern to neurons. This transmembrane protein mediates the efflux of Cl(-) out of neurons and exerts a critical role in inhibitory γ-aminobutyric acidergic (GABAergic) and glycinergic neurotransmission. Moreover, KCC2 participates in the regulation of various physiological processes of neurons, including cell migration, dendritic outgrowth, spine morphology, and dendritic synaptogenesis. It is important to note that down-regulation of KCC2 is associated with the pathogenesis of multiple neurological diseases, which is of particular relevance to acute central nervous system (CNS) injury. In this review, we aim to survey the pathogenic significance of KCC2 down-regulation under the condition of acute CNS injuries. We propose that further elucidation of the molecular mechanisms regarding KCC2 down-regulation after acute CNS injuries is necessary because of potential promising avenues for prevention and treatment of acute CNS injury.
Collapse
Affiliation(s)
- Haijian Wu
- Department of Neurosurgery, Second Affiliated Hospital, School of Medicine, Zhejiang University, 88 Jiefang Road, Hangzhou, 310009, Zhejiang, China
| | - Xiaoru Che
- Department of Cardiology, Zhejiang Provincial People's Hospital, Hangzhou, China
| | - Junjia Tang
- Department of Neurosurgery, Second Affiliated Hospital, School of Medicine, Zhejiang University, 88 Jiefang Road, Hangzhou, 310009, Zhejiang, China
| | - Feiqiang Ma
- Department of Emergency Medicine, Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Kun Pan
- Department of Neurosurgery, New York-Presbyterian Hospital, New York, NY, USA
| | - Mingfei Zhao
- Department of Neurosurgery, Second Affiliated Hospital, School of Medicine, Zhejiang University, 88 Jiefang Road, Hangzhou, 310009, Zhejiang, China
| | - Anwen Shao
- Department of Neurosurgery, Second Affiliated Hospital, School of Medicine, Zhejiang University, 88 Jiefang Road, Hangzhou, 310009, Zhejiang, China
| | - Qun Wu
- Department of Neurosurgery, Second Affiliated Hospital, School of Medicine, Zhejiang University, 88 Jiefang Road, Hangzhou, 310009, Zhejiang, China
| | - Jianmin Zhang
- Department of Neurosurgery, Second Affiliated Hospital, School of Medicine, Zhejiang University, 88 Jiefang Road, Hangzhou, 310009, Zhejiang, China
| | - Yuan Hong
- Department of Neurosurgery, Second Affiliated Hospital, School of Medicine, Zhejiang University, 88 Jiefang Road, Hangzhou, 310009, Zhejiang, China.
| |
Collapse
|
36
|
Jentsch TJ. VRACs and other ion channels and transporters in the regulation of cell volume and beyond. Nat Rev Mol Cell Biol 2016; 17:293-307. [PMID: 27033257 DOI: 10.1038/nrm.2016.29] [Citation(s) in RCA: 229] [Impact Index Per Article: 25.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Cells need to regulate their volume to counteract osmotic swelling or shrinkage, as well as during cell division, growth, migration and cell death. Mammalian cells adjust their volume by transporting potassium, sodium, chloride and small organic osmolytes using plasma membrane channels and transporters. This generates osmotic gradients, which drive water in and out of cells. Key players in this process are volume-regulated anion channels (VRACs), the composition of which has recently been identified and shown to encompass LRRC8 heteromers. VRACs also transport metabolites and drugs and function in extracellular signal transduction, apoptosis and anticancer drug resistance.
Collapse
Affiliation(s)
- Thomas J Jentsch
- Leibniz-Institut für Molekulare Pharmakologie (FMP) and Max-Delbrück-Centrum für Molekulare Medizin (MDC), Robert-Rössle-Strasse 10, 13125 Berlin, Germany
| |
Collapse
|
37
|
Brown FC, Conway AJ, Cerruti L, Collinge JE, McLean C, Wiley JS, Kile BT, Jane SM, Curtis DJ. Activation of the erythroid K-Cl cotransporter Kcc1 enhances sickle cell disease pathology in a humanized mouse model. Blood 2015; 126:2863-70. [PMID: 26450986 DOI: 10.1182/blood-2014-10-609362] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2014] [Accepted: 09/24/2015] [Indexed: 12/18/2022] Open
Abstract
We used an N-ethyl-N-nitrosurea-based forward genetic screen in mice to identify new genes and alleles that regulate erythropoiesis. Here, we describe a mouse line expressing an activated form of the K-Cl cotransporter Slc12a4 (Kcc1), which results in a semi-dominant microcytosis of red cells. A missense mutation from methionine to lysine in the cytoplasmic tail of Kcc1 impairs phosphorylation of adjacent threonines required for inhibiting cotransporter activity. We bred Kcc1(M935K) mutant mice with a humanized mouse model of sickle cell disease to directly explore the relevance of the reported increase in KCC activity in disease pathogenesis. We show that a single mutant allele of Kcc1 induces widespread sickling and tissue damage, leading to premature death. This mouse model reveals important new insights into the regulation of K-Cl cotransporters and provides in vivo evidence that increased KCC activity worsened end-organ damage and diminished survival in sickle cell disease.
Collapse
Affiliation(s)
- Fiona C Brown
- Australian Centre for Blood Diseases, Central Clinical School, Monash University, Melbourne, Australia
| | - Ashlee J Conway
- Australian Centre for Blood Diseases, Central Clinical School, Monash University, Melbourne, Australia
| | - Loretta Cerruti
- Australian Centre for Blood Diseases, Central Clinical School, Monash University, Melbourne, Australia
| | - Janelle E Collinge
- Cancer and Haematology Division, The Walter and Eliza Hall Institute of Medical Research, Parkville, Australia
| | | | - James S Wiley
- The Florey Institute of Neuroscience and Mental Health, University of Melbourne, Parkville, Australia; and
| | - Ben T Kile
- Cancer and Haematology Division, The Walter and Eliza Hall Institute of Medical Research, Parkville, Australia
| | - Stephen M Jane
- Australian Centre for Blood Diseases, Central Clinical School, Monash University, Melbourne, Australia; The Alfred Hospital, Melbourne, Australia; Department of Medicine, Central Clinical School, Monash University, Melbourne, Australia
| | - David J Curtis
- Australian Centre for Blood Diseases, Central Clinical School, Monash University, Melbourne, Australia; The Alfred Hospital, Melbourne, Australia
| |
Collapse
|
38
|
Kahle KT, Khanna AR, Alper SL, Adragna NC, Lauf PK, Sun D, Delpire E. K-Cl cotransporters, cell volume homeostasis, and neurological disease. Trends Mol Med 2015; 21:513-23. [PMID: 26142773 PMCID: PMC4834970 DOI: 10.1016/j.molmed.2015.05.008] [Citation(s) in RCA: 96] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2015] [Revised: 05/10/2015] [Accepted: 05/29/2015] [Indexed: 11/24/2022]
Abstract
K(+)-Cl(-) cotransporters (KCCs) were originally characterized as regulators of red blood cell (RBC) volume. Since then, four distinct KCCs have been cloned, and their importance for volume regulation has been demonstrated in other cell types. Genetic models of certain KCCs, such as KCC3, and their inhibitory WNK-STE20/SPS1-related proline/alanine-rich kinase (SPAK) serine-threonine kinases, have demonstrated the evolutionary necessity of these molecules for nervous system cell volume regulation, structure, and function, and their involvement in neurological disease. The recent characterization of a swelling-activated dephosphorylation mechanism that potently stimulates the KCCs has pinpointed a potentially druggable switch of KCC activity. An improved understanding of WNK/SPAK-mediated KCC cell volume regulation in the nervous system might reveal novel avenues for the treatment of multiple neurological diseases.
Collapse
Affiliation(s)
- Kristopher T Kahle
- Department of Neurosurgery, Boston Children's Hospital and Harvard Medical School, Boston, MA 02114, USA; Manton Center for Orphan Disease Research, Children's Hospital Boston, 300 Longwood Avenue, Boston, MA 02114, USA.
| | - Arjun R Khanna
- Department of Neurosurgery, Boston Children's Hospital and Harvard Medical School, Boston, MA 02114, USA
| | - Seth L Alper
- Renal Division and Molecular and Vascular Medicine Division, Beth Israel Deaconess Medical Center; Department of Medicine, Harvard Medical School, Boston, MA 02215, USA
| | - Norma C Adragna
- Department of Pharmacology and Toxicology, Boonshoft School of Medicine, Wright State University, Dayton, OH 45435, USA
| | - Peter K Lauf
- Department of Pharmacology and Toxicology, Boonshoft School of Medicine, Wright State University, Dayton, OH 45435, USA; Department of Pathology, Boonshoft School of Medicine, Wright State University, Dayton, OH 45435, USA
| | - Dandan Sun
- Department of Neurology, University of Pittsburgh, Pittsburgh, PA 15217, USA; Veterans Affairs Pittsburgh Health Care System, Geriatric Research, Educational and Clinical Center, Pittsburgh, PA 15213, USA
| | - Eric Delpire
- Department of Anesthesiology, Vanderbilt University School of Medicine, Nashville, TN 37232, USA.
| |
Collapse
|
39
|
Titz S, Sammler EM, Hormuzdi SG. Could tuning of the inhibitory tone involve graded changes in neuronal chloride transport? Neuropharmacology 2015; 95:321-31. [PMID: 25843644 DOI: 10.1016/j.neuropharm.2015.03.026] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2014] [Revised: 03/17/2015] [Accepted: 03/24/2015] [Indexed: 11/15/2022]
Abstract
Hyperpolarizing synaptic inhibition through GABAA and glycine receptors depends on the presence of the neuronal cation-chloride-cotransporter protein, KCC2. Several transcriptional and post-transcriptional mechanisms have been shown to regulate KCC2 and thereby influence the polarity and efficacy of inhibitory synaptic transmission. It is unclear however whether regulation of KCC2 enables the transporter to attain different levels of activity thus allowing a neuron to modulate the strength of inhibitory synaptic transmission to its changing requirements. We therefore investigated whether phosphorylation can allow KCC2 to achieve distinct levels of [Cl(-)]i in neurons. We generated a variety of KCC2 alanine dephosphorylation mimics and used NH4(+)-induced pHi shifts in cultured hippocampal neurons to quantify the rate of KCC2 transport activity exhibited by these mutants. To explore the relationship between KCC2 transport and GABAA receptor-mediated current amplitudes we performed gramicidine perforated-patch recordings. The correlation between EGABA and NH4(+)-induced pHi shifts enabled an estimate of the range of chloride extrusion possible by kinase/phosphatase regulation of KCC2. Our results demonstrate that KCC2 transport can vary considerably in magnitude depending on the combination of alanine mutations present on the protein. Transport can be enhanced to sufficiently high levels that hyperpolarizing GABAA responses may be obtained even in neurons with an extremely negative resting membrane potential and at high extracellular K(+) concentrations. Our findings highlight the significant potential for regulating the inhibitory tone by KCC2-mediated chloride extrusion and suggest that cellular signaling pathways may act combinatorially to alter KCC2 phosphorylation/dephosphorylation and thereby tune the strength of synaptic inhibition.
Collapse
Affiliation(s)
- Stefan Titz
- Institute for Physiology und Pathophysiology, University of Heidelberg, Im Neuenheimer Feld 326, 69120 Heidelberg, Germany.
| | - Esther M Sammler
- Division of Neuroscience, Ninewells Hospital and Medical School, University of Dundee, Dundee DD1 9SY, UK
| | - Sheriar G Hormuzdi
- Division of Neuroscience, Ninewells Hospital and Medical School, University of Dundee, Dundee DD1 9SY, UK.
| |
Collapse
|
40
|
Hartmann AM, Nothwang HG. Molecular and evolutionary insights into the structural organization of cation chloride cotransporters. Front Cell Neurosci 2015; 8:470. [PMID: 25653592 PMCID: PMC4301019 DOI: 10.3389/fncel.2014.00470] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2014] [Accepted: 12/30/2014] [Indexed: 01/26/2023] Open
Abstract
Cation chloride cotransporters (CCC) play an essential role for neuronal chloride homeostasis. K(+)-Cl(-) cotransporter (KCC2), is the principal Cl(-)-extruder, whereas Na(+)-K(+)-Cl(-) cotransporter (NKCC1), is the major Cl(-)-uptake mechanism in many neurons. As a consequence, the action of the inhibitory neurotransmitters gamma-aminobutyric acid (GABA) and glycine strongly depend on the activity of these two transporters. Knowledge of the mechanisms involved in ion transport and regulation is thus of great importance to better understand normal and disturbed brain function. Although no overall 3-dimensional crystal structures are yet available, recent molecular and phylogenetic studies and modeling have provided new and exciting insights into structure-function relationships of CCC. Here, we will summarize our current knowledge of the gross structural organization of the proteins, their functional domains, ion binding and translocation sites, and the established role of individual amino acids (aa). A major focus will be laid on the delineation of shared and distinct organizational principles between KCC2 and NKCC1. Exploiting the richness of recently generated genome data across the tree of life, we will also explore the molecular evolution of these features.
Collapse
Affiliation(s)
- Anna-Maria Hartmann
- Systematics and Evolutionary Biology Group, Institute for Biology and Environmental Sciences, Carl von Ossietzky University Oldenburg Oldenburg, Germany
| | - Hans Gerd Nothwang
- Neurogenetics Group, Center of Excellence Hearing4All, 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
|
41
|
Kaila K, Price TJ, Payne JA, Puskarjov M, Voipio J. Cation-chloride cotransporters in neuronal development, plasticity and disease. Nat Rev Neurosci 2014; 15:637-54. [PMID: 25234263 DOI: 10.1038/nrn3819] [Citation(s) in RCA: 529] [Impact Index Per Article: 48.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Electrical activity in neurons requires a seamless functional coupling between plasmalemmal ion channels and ion transporters. Although ion channels have been studied intensively for several decades, research on ion transporters is in its infancy. In recent years, it has become evident that one family of ion transporters, cation-chloride cotransporters (CCCs), and in particular K(+)-Cl(-) cotransporter 2 (KCC2), have seminal roles in shaping GABAergic signalling and neuronal connectivity. Studying the functions of these transporters may lead to major paradigm shifts in our understanding of the mechanisms underlying brain development and plasticity in health and disease.
Collapse
Affiliation(s)
- Kai Kaila
- 1] Department of Biosciences, University of Helsinki, 00014 Helsinki, Finland. [2] Neuroscience Center, University of Helsinki, 00014 Helsinki, Finland
| | - Theodore J Price
- University of Texas at Dallas, School of Behavior and Brain Sciences, Dallas, Texas 75093, USA
| | - John A Payne
- Department of Physiology and Membrane Biology, School of Medicine, University of California, Davis, California 95616, USA
| | - Martin Puskarjov
- 1] Department of Biosciences, University of Helsinki, 00014 Helsinki, Finland. [2] Neuroscience Center, University of Helsinki, 00014 Helsinki, Finland
| | - Juha Voipio
- Department of Biosciences, University of Helsinki, 00014 Helsinki, Finland
| |
Collapse
|
42
|
Gephyrin clusters are absent from small diameter primary afferent terminals despite the presence of GABA(A) receptors. J Neurosci 2014; 34:8300-17. [PMID: 24920633 DOI: 10.1523/jneurosci.0159-14.2014] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
Whereas both GABA(A) receptors (GABA(A)Rs) and glycine receptors (GlyRs) play a role in control of dorsal horn neuron excitability, their relative contribution to inhibition of small diameter primary afferent terminals remains controversial. To address this, we designed an approach for quantitative analyses of the distribution of GABA(A)R-subunits, GlyR α1-subunit and their anchoring protein, gephyrin, on terminals of rat spinal sensory afferents identified by Calcitonin-Gene-Related-Peptide (CGRP) for peptidergic terminals, and by Isolectin-B4 (IB4) for nonpeptidergic terminals. The approach was designed for light microscopy, which is compatible with the mild fixation conditions necessary for immunodetection of several of these antigens. An algorithm was designed to recognize structures with dimensions similar to those of the microscope resolution. To avoid detecting false colocalization, the latter was considered significant only if the degree of pixel overlap exceeded that expected from randomly overlapping pixels given a hypergeometric distribution. We found that both CGRP(+) and IB4(+) terminals were devoid of GlyR α1-subunit and gephyrin. The α1 GABA(A)R was also absent from these terminals. In contrast, the GABA(A)R α2/α3/α5 and β3 subunits were significantly expressed in both terminal types, as were other GABA(A)R-associated-proteins (α-Dystroglycan/Neuroligin-2/Collybistin-2). Ultrastructural immunocytochemistry confirmed the presence of GABA(A)R β3 subunits in small afferent terminals. Real-time quantitative PCR (qRT-PCR) confirmed the results of light microscopy immunochemical analysis. These results indicate that dorsal horn inhibitory synapses follow different rules of organization at presynaptic versus postsynaptic sites (nociceptive afferent terminals vs inhibitory synapses on dorsal horn neurons). The absence of gephyrin clusters from primary afferent terminals suggests a more diffuse mode of GABA(A)-mediated transmission at presynaptic than at postsynaptic sites.
Collapse
|
43
|
Weber M, Hartmann AM, Beyer T, Ripperger A, Nothwang HG. A novel regulatory locus of phosphorylation in the C terminus of the potassium chloride cotransporter KCC2 that interferes with N-ethylmaleimide or staurosporine-mediated activation. J Biol Chem 2014; 289:18668-79. [PMID: 24849604 PMCID: PMC4081912 DOI: 10.1074/jbc.m114.567834] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2014] [Revised: 05/15/2014] [Indexed: 11/06/2022] Open
Abstract
The neuron-specific cation chloride cotransporter KCC2 plays a crucial role in hyperpolarizing synaptic inhibition. Transporter dysfunction is associated with various neurological disorders, raising interest in regulatory mechanisms. Phosphorylation has been identified as a key regulatory process. Here, we retrieved experimentally observed phosphorylation sites of KCC2 from public databases and report on the systematic analysis of six phosphorylated serines, Ser(25), Ser(26), Ser(937), Ser(1022), Ser(1025), and Ser(1026). Alanine or aspartate substitutions of these residues were analyzed in HEK-293 cells. All mutants were expressed in a pattern similar to wild-type KCC2 (KCC2(WT)). Tl(+) flux measurements demonstrated unchanged transport activity for Ser(25), Ser(26), Ser(1022), Ser(1025), and Ser(1026) mutants. In contrast, KCC2(S937D), mimicking phosphorylation, resulted in a significant up-regulation of transport activity. Aspartate substitution of Thr(934), a neighboring putative phosphorylation site, resulted in a comparable increase in KCC2 transport activity. Both KCC2(T934D) and KCC2(S937D) mutants were inhibited by the kinase inhibitor staurosporine and by N-ethylmaleimide, whereas KCC2(WT), KCC2(T934A), and KCC2(S937A) were activated. The inverse staurosporine effect on aspartate versus alanine substitutions reveals a cross-talk between different phosphorylation sites of KCC2. Immunoblot and cell surface labeling experiments detected no alterations in total abundance or surface expression of KCC2(T934D) and KCC2(S937D) compared with KCC2(WT). These data reveal kinetic regulation of transport activity by these residues. In summary, our data identify a novel key regulatory phosphorylation site of KCC2 and a functional interaction between different conformation-changing post-translational modifications. The action of pharmacological agents aimed to modulate KCC2 activity for therapeutic benefit might therefore be highly context-specific.
Collapse
Affiliation(s)
- Maren Weber
- From the Neurogenetics Group, Center of Excellence Hearing4All, School of Medicine and Health Sciences
| | - Anna-Maria Hartmann
- From the Neurogenetics Group, Center of Excellence Hearing4All, School of Medicine and Health Sciences, Systematics and Evolutionary Biology Group, Institute for Biology and Environmental Sciences, and
| | - Timo Beyer
- From the Neurogenetics Group, Center of Excellence Hearing4All, School of Medicine and Health Sciences
| | - Anne Ripperger
- From the Neurogenetics Group, Center of Excellence Hearing4All, School of Medicine and Health Sciences
| | - Hans Gerd Nothwang
- From the Neurogenetics Group, Center of Excellence Hearing4All, School of Medicine and Health Sciences, the Research Center for Neurosensory Sciences, Carl von Ossietzky University Oldenburg, 26111 Oldenburg, Germany
| |
Collapse
|
44
|
Puskarjov M, Seja P, Heron SE, Williams TC, Ahmad F, Iona X, Oliver KL, Grinton BE, Vutskits L, Scheffer IE, Petrou S, Blaesse P, Dibbens LM, Berkovic SF, Kaila K. A variant of KCC2 from patients with febrile seizures impairs neuronal Cl- extrusion and dendritic spine formation. EMBO Rep 2014; 15:723-9. [PMID: 24668262 PMCID: PMC4197883 DOI: 10.1002/embr.201438749] [Citation(s) in RCA: 129] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2014] [Accepted: 03/10/2014] [Indexed: 11/11/2022] Open
Abstract
Genetic variation in SLC12A5 which encodes KCC2, the neuron-specific cation-chloride cotransporter that is essential for hyperpolarizing GABAergic signaling and formation of cortical dendritic spines, has not been reported in human disease. Screening of SLC12A5 revealed a co-segregating variant (KCC2-R952H) in an Australian family with febrile seizures. We show that KCC2-R952H reduces neuronal Cl(-) extrusion and has a compromised ability to induce dendritic spines in vivo and in vitro. Biochemical analyses indicate a reduced surface expression of KCC2-R952H which likely contributes to the functional deficits. Our data suggest that KCC2-R952H is a bona fide susceptibility variant for febrile seizures.
Collapse
Affiliation(s)
- Martin Puskarjov
- Department of Biosciences, University of Helsinki, Helsinki, Finland Neuroscience Center, University of Helsinki, Helsinki, Finland
| | - Patricia Seja
- Department of Biosciences, University of Helsinki, Helsinki, Finland Neuroscience Center, University of Helsinki, Helsinki, Finland
| | - Sarah E Heron
- Epilepsy Research Program, School of Pharmacy and Medical Sciences, University of South Australia, Adelaide, SA, Australia Sansom Institute for Health Research, University of South Australia, Adelaide SA, Australia
| | - Tristiana C Williams
- Epilepsy Research Program, School of Pharmacy and Medical Sciences, University of South Australia, Adelaide, SA, Australia
| | - Faraz Ahmad
- Department of Biosciences, University of Helsinki, Helsinki, Finland Neuroscience Center, University of Helsinki, Helsinki, Finland
| | - Xenia Iona
- Epilepsy Research Program, School of Pharmacy and Medical Sciences, University of South Australia, Adelaide, SA, Australia
| | - Karen L Oliver
- Department of Medicine, Epilepsy Research Center, The University of Melbourne Austin Health, Melbourne Vic., Australia
| | - Bronwyn E Grinton
- Department of Paediatrics, Florey Institute, Royal Children's Hospital The University of Melbourne, Melbourne, Vic., Australia
| | - Laszlo Vutskits
- Department of Anesthesiology, Pharmacology and Intensive Care, University Hospital of Geneva, Geneva, Switzerland
| | - Ingrid E Scheffer
- Department of Medicine, Epilepsy Research Center, The University of Melbourne Austin Health, Melbourne Vic., Australia Department of Paediatrics, Florey Institute, Royal Children's Hospital The University of Melbourne, Melbourne, Vic., Australia
| | - Steven Petrou
- Florey Institute of Neuroscience and Mental Health and the Center for Neural Engineering, The University of Melbourne, Parkville, Vic., Australia
| | - Peter Blaesse
- Department of Biosciences, University of Helsinki, Helsinki, Finland Institute of Physiology I, Westfälische Wilhelms-University Münster, Münster, Germany
| | - Leanne M Dibbens
- Epilepsy Research Program, School of Pharmacy and Medical Sciences, University of South Australia, Adelaide, SA, Australia Sansom Institute for Health Research, University of South Australia, Adelaide SA, Australia
| | - Samuel F Berkovic
- Department of Medicine, Epilepsy Research Center, The University of Melbourne Austin Health, Melbourne Vic., Australia
| | - Kai Kaila
- Department of Biosciences, University of Helsinki, Helsinki, Finland Neuroscience Center, University of Helsinki, Helsinki, Finland
| |
Collapse
|
45
|
Medina I, Friedel P, Rivera C, Kahle KT, Kourdougli N, Uvarov P, Pellegrino C. Current view on the functional regulation of the neuronal K(+)-Cl(-) cotransporter KCC2. Front Cell Neurosci 2014; 8:27. [PMID: 24567703 PMCID: PMC3915100 DOI: 10.3389/fncel.2014.00027] [Citation(s) in RCA: 141] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2013] [Accepted: 01/18/2014] [Indexed: 12/22/2022] Open
Abstract
In the mammalian central nervous system (CNS), the inhibitory strength of chloride (Cl(-))-permeable GABAA and glycine receptors (GABAAR and GlyR) depends on the intracellular Cl(-) concentration ([Cl(-)]i). Lowering [Cl(-)]i enhances inhibition, whereas raising [Cl(-)]i facilitates neuronal activity. A neuron's basal level of [Cl(-)]i, as well as its Cl(-) extrusion capacity, is critically dependent on the activity of the electroneutral K(+)-Cl(-) cotransporter KCC2, a member of the SLC12 cation-Cl(-) cotransporter (CCC) family. KCC2 deficiency compromises neuronal migration, formation and the maturation of GABAergic and glutamatergic synaptic connections, and results in network hyperexcitability and seizure activity. Several neurological disorders including multiple epilepsy subtypes, neuropathic pain, and schizophrenia, as well as various insults such as trauma and ischemia, are associated with significant decreases in the Cl(-) extrusion capacity of KCC2 that result in increases of [Cl(-)]i and the subsequent hyperexcitability of neuronal networks. Accordingly, identifying the key upstream molecular mediators governing the functional regulation of KCC2, and modifying these signaling pathways with small molecules, might constitute a novel neurotherapeutic strategy for multiple diseases. Here, we discuss recent advances in the understanding of the mechanisms regulating KCC2 activity, and of the role these mechanisms play in neuronal Cl(-) homeostasis and GABAergic neurotransmission. As KCC2 mediates electroneutral transport, the experimental recording of its activity constitutes an important research challenge; we therefore also, provide an overview of the different methodological approaches utilized to monitor function of KCC2 in both physiological and pathological conditions.
Collapse
Affiliation(s)
- Igor Medina
- INSERM, Institut de Neurobiologie de la Méditerranée (INMED)Marseille, France
- Aix-Marseille Université, UMR901Marseille, France
| | - Perrine Friedel
- INSERM, Institut de Neurobiologie de la Méditerranée (INMED)Marseille, France
- Aix-Marseille Université, UMR901Marseille, France
| | - Claudio Rivera
- INSERM, Institut de Neurobiologie de la Méditerranée (INMED)Marseille, France
- Aix-Marseille Université, UMR901Marseille, France
- Neuroscience Center, University of HelsinkiHelsinki, Finland
| | - Kristopher T. Kahle
- Department of Cardiology, Manton Center for Orphan Disease Research, Howard Hughes Medical Institute, Boston Children's HospitalBoston, MA, USA
- Department of Neurosurgery, Massachusetts General Hospital and Harvard Medical SchoolBoston, MA, USA
| | - Nazim Kourdougli
- INSERM, Institut de Neurobiologie de la Méditerranée (INMED)Marseille, France
- Aix-Marseille Université, UMR901Marseille, France
| | - Pavel Uvarov
- Institute of Biomedicine, Anatomy, University of HelsinkiHelsinki, Finland
| | - Christophe Pellegrino
- INSERM, Institut de Neurobiologie de la Méditerranée (INMED)Marseille, France
- Aix-Marseille Université, UMR901Marseille, France
| |
Collapse
|
46
|
Kahle KT, Deeb TZ, Puskarjov M, Silayeva L, Liang B, Kaila K, Moss SJ. Modulation of neuronal activity by phosphorylation of the K-Cl cotransporter KCC2. Trends Neurosci 2013; 36:726-737. [PMID: 24139641 PMCID: PMC4381966 DOI: 10.1016/j.tins.2013.08.006] [Citation(s) in RCA: 154] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2013] [Revised: 08/19/2013] [Accepted: 08/26/2013] [Indexed: 12/29/2022]
Abstract
The K-Cl cotransporter KCC2 establishes the low intraneuronal Cl- levels required for the hyperpolarizing inhibitory postsynaptic potentials mediated by ionotropic γ-aminobutyric acid receptors (GABAARs) and glycine receptors (GlyRs). Decreased KCC2-mediated Cl- extrusion and impaired hyperpolarizing GABAAR- and/or GlyR-mediated currents have been implicated in epilepsy, neuropathic pain, and spasticity. Recent evidence suggests that the intrinsic ion transport rate, cell surface stability, and plasmalemmal trafficking of KCC2 are rapidly and reversibly modulated by the (de)phosphorylation of critical serine, threonine, and tyrosine residues in the C terminus of this protein. Alterations in KCC2 phosphorylation have been associated with impaired KCC2 function in several neurological diseases. Targeting KCC2 phosphorylation directly or indirectly via upstream regulatory kinases might be a novel strategy to modulate GABA- and/or glycinergic signaling for therapeutic benefit.
Collapse
Affiliation(s)
- Kristopher T Kahle
- Department of Neurosurgery, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114, USA
- Howard Hughes Medical Institute, Department of Cardiology, Manton Center for Orphan Disease Research, Boston Children's Hospital, Boston, MA 02115, USA
| | - Tarek Z Deeb
- Department of Neuroscience, Tufts University School of Medicine, Boston, MA 02111, USA
| | - Martin Puskarjov
- Department of Biosciences and Neuroscience Center, University of Helsinki, FI-00014 Helsinki, Finland
| | - Liliya Silayeva
- Department of Neuroscience, Tufts University School of Medicine, Boston, MA 02111, USA
| | - Bo Liang
- Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, Boston, MA 02115, USA
| | - Kai Kaila
- Department of Biosciences and Neuroscience Center, University of Helsinki, FI-00014 Helsinki, Finland
| | - 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
| |
Collapse
|
47
|
Hartmann AM, Tesch D, Nothwang HG, Bininda-Emonds OR. Evolution of the Cation Chloride Cotransporter Family: Ancient Origins, Gene Losses, and Subfunctionalization through Duplication. Mol Biol Evol 2013; 31:434-47. [DOI: 10.1093/molbev/mst225] [Citation(s) in RCA: 46] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023] Open
|
48
|
Arroyo JP, Kahle KT, Gamba G. The SLC12 family of electroneutral cation-coupled chloride cotransporters. Mol Aspects Med 2013; 34:288-98. [PMID: 23506871 DOI: 10.1016/j.mam.2012.05.002] [Citation(s) in RCA: 113] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2012] [Accepted: 04/09/2012] [Indexed: 11/21/2022]
Abstract
The SLC12 family encodes electroneutral cation-coupled chloride cotransporters that are critical for several physiological processes including cell volume regulation, modulation of intraneuronal chloride concentration, transepithelial ion movement, and blood pressure regulation. Members of this family are the targets of the most commonly used diuretic drugs, have been shown to be the causative genes for inherited disease such as Gitelman, Bartter and Andermann syndromes, and potentially play a role in polygenic complex diseases like arterial hypertension, epilepsy, osteoporosis, and cancer.
Collapse
Affiliation(s)
- Juan Pablo Arroyo
- Molecular Physiology Unit, Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México, and Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Mexico
| | | | | |
Collapse
|
49
|
Gagnon KB, Delpire E. Physiology of SLC12 transporters: lessons from inherited human genetic mutations and genetically engineered mouse knockouts. Am J Physiol Cell Physiol 2013; 304:C693-714. [PMID: 23325410 DOI: 10.1152/ajpcell.00350.2012] [Citation(s) in RCA: 116] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Among the over 300 members of the solute carrier (SLC) group of integral plasma membrane transport proteins are the nine electroneutral cation-chloride cotransporters belonging to the SLC12 gene family. Seven of these transporters have been functionally described as coupling the electrically silent movement of chloride with sodium and/or potassium. Although in silico analysis has identified two additional SLC12 family members, no physiological role has been ascribed to the proteins encoded by either the SLC12A8 or the SLC12A9 genes. Evolutionary conservation of this gene family from protists to humans confirms their importance. A wealth of physiological, immunohistochemical, and biochemical studies have revealed a great deal of information regarding the importance of this gene family to human health and disease. The sequencing of the human genome has provided investigators with the capability to link several human diseases with mutations in the genes encoding these plasma membrane proteins. The availability of bacterial artificial chromosomes, recombination engineering techniques, and the mouse genome sequence has simplified the creation of targeting constructs to manipulate the expression/function of these cation-chloride cotransporters in the mouse in an attempt to recapitulate some of these human pathologies. This review will summarize the three human disorders that have been linked to the mutation/dysfunction of the Na-Cl, Na-K-2Cl, and K-Cl cotransporters (i.e., Bartter's, Gitleman's, and Andermann's syndromes), examine some additional pathologies arising from genetically modified mouse models of these cotransporters including deafness, blood pressure, hyperexcitability, and epithelial transport deficit phenotypes.
Collapse
Affiliation(s)
- Kenneth B Gagnon
- Department of Anatomy and Cell Biology, University of Saskatchewan, Saskatoon, Saskatchewan, Canada
| | | |
Collapse
|
50
|
Antrobus SP, Lytle C, Payne JA. K+-Cl- cotransporter-2 KCC2 in chicken cardiomyocytes. Am J Physiol Cell Physiol 2012; 303:C1180-91. [PMID: 23034386 PMCID: PMC3530769 DOI: 10.1152/ajpcell.00274.2012] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2012] [Accepted: 09/25/2012] [Indexed: 12/15/2022]
Abstract
Using antibodies prepared against a unique region (exon 22-24) of rat K(+)-Cl(-) cotransporter-2 (KCC2), we confirmed that the ~140-kDa KCC2 protein is exclusively expressed in rat brain, but in chicken, we observed strong reactivity not only with the ~140-kDa KCC2 protein in brain but also a slightly larger ~145-kDa protein in heart. In silico analysis showed that while exon 22 of KCC2 is unique to this isoform in therian mammals, it is retained in KCC2's closest paralog, KCC4, of lower vertebrates, including chicken. To eliminate potential cross-reactivity with chicken KCC4, the antibodies were preadsorbed with blocking peptides prepared over the only two regions showing significant sequence identity to chicken KCC4. This completely eliminated antibody recognition of exogenously expressed chicken KCC4 but not of the ~145-kDa protein in chicken heart, indicating that chicken heart expresses KCC2. Real-time PCR confirmed robust KCC2 transcript expression in both chicken brain and heart. Chicken heart expressed predominantly the longer KCC2a splice variant consistent with the larger ~145-kDa protein in chicken heart. Immunofluorescence microscopy revealed prominent plasma membrane KCC2 labeling in chicken ventricular cardiomyocytes. We hypothesize that KCC2 is an important Cl(-) extrusion pathway in avian cardiomyocytes that counters channel-mediated Cl(-) loading during high heart rates with β-adrenergic stimulation. While KCC2 is absent from mammalian cardiomyocytes, understanding the role that the other KCC isoforms play in Cl(-) homeostasis of these cells represents a nascent area of research.
Collapse
Affiliation(s)
- Shane P Antrobus
- Dept. of Physiology and Membrane Biology, School of Medicine, Univ. of California, Davis, CA 95616, USA
| | | | | |
Collapse
|