1
|
Kok M, Singh I, Aizenman E, Brodsky JL. Inefficient maturation of disease-linked mutant forms of the KCC2 potassium-chloride cotransporter correlates with predicted pathogenicity. J Biol Chem 2025; 301:108399. [PMID: 40074080 PMCID: PMC12001125 DOI: 10.1016/j.jbc.2025.108399] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2024] [Revised: 02/20/2025] [Accepted: 02/26/2025] [Indexed: 03/14/2025] Open
Abstract
The potassium-chloride cotransporter 2 (KCC2) is required for neuronal development, and KCC2 dysregulation is implicated in several neurodevelopmental disorders, including schizophrenia, autism, and epilepsy. A dozen mutations in the KCC2-encoding gene, SLC12A5, are associated with these disorders, but few are fully characterized. To this end, we examined KCC2 biogenesis in a HEK293 cell model. While most of the examined disease-associated mutants matured efficiently, the L403P mutant was unable to traffic to the Golgi. Two other mutants, A191V and R857L, exhibited more subtle defects in maturation. Cell surface biotinylation assays showed that these mutants were also depleted from the cell surface. Another disease-associated variant, R952H, acquired Golgi-associated glycans yet was significantly depleted from the plasma membrane, consistent with loss of a plasma membrane-stabilizing phosphorylation site. To determine whether the ability of KCC2 to mature to the Golgi could be predicted, we employed a computational pathogenicity program, Rhapsody, which was shown in past work to predict endoplasmic reticulum-associated degradation-targeting of an unrelated ion channel. We discovered that the Rhapsody pathogenicity score correlated with relative defects in KCC2 maturation, and the algorithm outperformed two other commonly used programs. These data demonstrate the efficacy of a bioinformatic tool to predict the efficiency of KCC2 biogenesis. We also propose that Rhapsody can be used to develop hypotheses on defects associated with other disease-associated SLC12A5 alleles as they are identified.
Collapse
Affiliation(s)
- Morgan Kok
- Department of Biological Sciences, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Ishika Singh
- Department of Biological Sciences, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Elias Aizenman
- Department of Neurobiology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
| | - Jeffrey L Brodsky
- Department of Biological Sciences, University of Pittsburgh, Pittsburgh, Pennsylvania, USA.
| |
Collapse
|
2
|
Magro G. Early Polytherapy for Probably Benzodiazepine Refractory Naïve Status Epilepticus (Stage 1 Plus). Neurol Int 2025; 17:11. [PMID: 39852775 PMCID: PMC11767287 DOI: 10.3390/neurolint17010011] [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: 12/16/2024] [Revised: 01/10/2025] [Accepted: 01/16/2025] [Indexed: 01/26/2025] Open
Abstract
Stage 1 Plus is defined here as a naïve, previously untreated, status epilepticus (SE) that is probably refractory to Benzodiazepines (BDZ). These cases include not only prolonged SE as previously proposed by the author (SE lasting > 10 min) but also other cases notoriously associated with BDZ refractoriness such as the absence of prominent motor phenomena and acute etiology (especially primary central nervous system etiology). Interestingly, the absence of prominent motor phenomena as is the case of non convulsive SE might implicitly fall in the category of prolonged SE due to the delay in recognition and treatment. Future studies should help identify other factors associated with BDZ refractoriness, therefore widening the definition of Stage 1 Plus. The appropriate timing for defining prolonged SE may also differ depending on different etiology. Consequently, in future tailored models of SE, the definition of prolonged SE could be enhanced by defining it for a longer duration than Tx, a time point that changes based on different etiologies (x), Tx being much shorter than 10 min in acute etiologies. These cases of naïve probably BDZ refractory SE (Stage 1 Plus) might require a different approach: combined polytherapy from the start. The objective of this review is to provide pathophysiological and pre-clinical evidence, mostly from animal studies, for the different approach of combined polytherapy from the start for those cases of SE falling in the definition of Stage 1 Plus.
Collapse
Affiliation(s)
- Giuseppe Magro
- Department of Neuroscience, "Giovanni Paolo II" Hospital, Lamezia Terme, 88046 Catanzaro, Italy
| |
Collapse
|
3
|
Henderson SW, Nourmohammadi S, Hrmova M. Protein Structural Modeling and Transport Thermodynamics Reveal That Plant Cation-Chloride Cotransporters Mediate Potassium-Chloride Symport. Int J Mol Sci 2024; 25:12955. [PMID: 39684666 DOI: 10.3390/ijms252312955] [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: 10/25/2024] [Revised: 11/17/2024] [Accepted: 12/01/2024] [Indexed: 12/18/2024] Open
Abstract
Plant cation-chloride cotransporters (CCCs) are proposed to be Na+-K+-2Cl- transporting membrane proteins, although evolutionarily, they associate more closely with K+-Cl- cotransporters (KCCs). Here, we investigated grapevine (Vitis vinifera L.) VvCCC using 3D protein modeling, bioinformatics, and electrophysiology with a heterologously expressed protein. The 3D protein modeling revealed that the signatures of ion binding sites in plant CCCs resembled those of animal KCCs, which was supported by phylogenomic analyses and ancestral sequence reconstruction. The conserved features of plant CCCs and animal KCCs included predicted K+ and Cl--binding sites and the absence of a Na+-binding site. Measurements with VvCCC-injected Xenopus laevis oocytes with VvCCC localizing to plasma membranes indicated that the oocytes had depleted intracellular Cl- and net 86Rb fluxes, which agreed with thermodynamic predictions for KCC cotransport. The 86Rb uptake by VvCCC-injected oocytes was Cl--dependent, did not require external Na+, and was partially inhibited by the non-specific CCC-blocker bumetanide, implying that these properties are typical of KCC transporters. A loop diuretic-insensitive Na+ conductance in VvCCC-injected oocytes may account for earlier observations of Na+ uptake by plant CCC proteins expressed in oocytes. Our data suggest plant CCC membrane proteins are likely to function as K+-Cl- cotransporters, which opens the avenues to define their biophysical properties and roles in plant physiology.
Collapse
Affiliation(s)
- Sam W Henderson
- School of Agriculture, Food and Wine, Waite Research Institute, Faculty of Sciences, Engineering and Technology, University of Adelaide, Waite Campus Precinct, Glen Osmond, Adelaide, SA 5064, Australia
| | - Saeed Nourmohammadi
- School of Agriculture, Food and Wine, Waite Research Institute, Faculty of Sciences, Engineering and Technology, University of Adelaide, Waite Campus Precinct, Glen Osmond, Adelaide, SA 5064, Australia
| | - Maria Hrmova
- School of Agriculture, Food and Wine, Waite Research Institute, Faculty of Sciences, Engineering and Technology, University of Adelaide, Waite Campus Precinct, Glen Osmond, Adelaide, SA 5064, Australia
| |
Collapse
|
4
|
Raveendran VA, Serranilla M, Asgarihafshejani A, de Saint-Rome M, Cherednychenko M, Mullany S, Mitchell JA, Pressey JC, Woodin MA. SNARE protein SNAP25 regulates the chloride-transporter KCC2 in neurons. iScience 2024; 27:111156. [PMID: 39507243 PMCID: PMC11539599 DOI: 10.1016/j.isci.2024.111156] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2024] [Revised: 09/03/2024] [Accepted: 10/09/2024] [Indexed: 11/08/2024] Open
Abstract
Inhibitory synaptic neurotransmission mediated by GABA requires a low concentration of chloride ions (Cl-) in neurons, which is established and maintained by the potassium-chloride co-transporter 2 (KCC2). While KCC2-interacting proteins are known to regulate KCC2 protein level and function, specific KCC2-interacting partners are still being identified and characterized. We asked whether SNAP25, an integral component of the SNARE-complex and a novel KCC2 interactor, regulates KCC2 protein and function in mice. We demonstrated that SNAP25 interacts with KCC2, and that this interaction is regulated by protein kinase C (PKC)-mediated phosphorylation. We also discovered that SNAP25 knockdown decreases total KCC2 in cortical neurons, and reduces the strength of synaptic inhibition, as demonstrated through a depolarization of the reversal potential for GABA (EGABA), indicating reduced KCC2 function. Our biochemical and electrophysiological data combined demonstrate that SNAP25 regulates KCC2 membrane expression and function, and in doing so, regulates inhibitory synaptic transmission.
Collapse
Affiliation(s)
| | - Melissa Serranilla
- Department of Cell and Systems Biology, University of Toronto, Toronto, ON M5S 3G5, Canada
| | - Azam Asgarihafshejani
- Department of Cell and Systems Biology, University of Toronto, Toronto, ON M5S 3G5, Canada
| | - Miranda de Saint-Rome
- Department of Cell and Systems Biology, University of Toronto, Toronto, ON M5S 3G5, Canada
| | - Mariia Cherednychenko
- Department of Cell and Systems Biology, University of Toronto, Toronto, ON M5S 3G5, Canada
| | - Shanelle Mullany
- Department of Cell and Systems Biology, University of Toronto, Toronto, ON M5S 3G5, Canada
| | - Jennifer A. Mitchell
- Department of Cell and Systems Biology, University of Toronto, Toronto, ON M5S 3G5, Canada
| | - Jessica C. Pressey
- Department of Cell and Systems Biology, University of Toronto, Toronto, ON M5S 3G5, Canada
| | - Melanie A. Woodin
- Department of Cell and Systems Biology, University of Toronto, Toronto, ON M5S 3G5, Canada
| |
Collapse
|
5
|
Takano H, Hsu FC, Coulter DA. Prolonged Hyperactivity Elicits Massive and Persistent Chloride Ion Redistribution in Subsets of Cultured Hippocampal Dentate Granule Cells. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.10.16.618704. [PMID: 39464011 PMCID: PMC11507851 DOI: 10.1101/2024.10.16.618704] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 10/29/2024]
Abstract
Chloride ions play a critical role in neuronal inhibition through the activity of chloride-permeable GABAA receptor channels. Ion transporters, chloride channels, and immobile ion species tightly regulate intracellular chloride concentrations. Several studies related to epilepsy suggest that chloride extrusion function may decrease in an activity-dependent manner. Consequently, it is crucial to investigate whether intense neuronal activity, as observed during status epilepticus, could lead to sustained increases in intracellular chloride levels in neurons, which in turn could contribute to epilepsy-associated hyperexcitability. This study utilized the chloride sensitive indicator (6-Methoxyquinolinio) acetic acid ethyl ester bromide (MQAE) combined with fluorescence lifetime imaging (FLIM) to examine whether application of the convulsant, pilocarpine, a muscarinic acetylcholine receptor agonist, could induce synchronous epileptiform activity and elevate intracellular chloride concentrations in hippocampal slice cultures. Using a Gaussian mixture model, we identified a multimodal distribution of intracellular chloride levels among neurons, with a significant subset of these cells exhibiting massive and prolonged (days) chloride accumulation. The combination of multicellular imaging and statistical analysis served as a powerful tool for studying the emergence of multiple, distinct populations of neurons in pathological conditions, in contrast to homogeneous populations evident under control conditions.
Collapse
Affiliation(s)
- Hajime Takano
- Division of Neurology, The Children’s Hospital of Philadelphia, Philadelphia, PA, 19104
- Department of Neurology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104
| | - Fu-Chun Hsu
- Division of Neurology, The Children’s Hospital of Philadelphia, Philadelphia, PA, 19104
| | - Douglas A. Coulter
- Division of Neurology, The Children’s Hospital of Philadelphia, Philadelphia, PA, 19104
- Department of Neurology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104
- Department of Neuroscience, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104
| |
Collapse
|
6
|
Cai J, Wu Z, Wang G, Zhao X, Wang X, Wang BH, Yu J, Liu X, Wang Y. The suppressive effect of the specific KCC2 modulator CLP290 on seizure in mice. Epilepsy Res 2024; 203:107365. [PMID: 38677001 DOI: 10.1016/j.eplepsyres.2024.107365] [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: 12/18/2023] [Revised: 04/03/2024] [Accepted: 04/15/2024] [Indexed: 04/29/2024]
Abstract
Epilepsy is a chronic neurological disorder characterized by episodic dysfunction of central nervous system. The most basic mechanism of epilepsy falls to the imbalance between excitation and inhibition. In adults, GABAA receptor (GABAAR) is the main inhibitory receptor to prevent neurons from developing hyperexcitability, while its inhibition relies on the low intracellular chloride anion concentration ([Cl-]i). Neuronal-specific electroneutral K+-Cl- cotransporter (KCC2) can mediate chloride efflux to lower [Cl-]i for GABAAR mediated inhibition. Our previous study has revealed that the coordinated downregulation of KCC2 and GABAAR participates in epilepsy. According to a high-throughout screen for compounds that reduce [Cl-]i, CLP290 turns out to be a specific KCC2 functional modulator. In current study, we first confirmed that CLP290 could dose-dependently suppress convulsant-induced seizures in mice in vivo as well as the epileptiform burst activities in cultured hippocampal neurons in vitro. Then, we discovered that CLP290 functioned through preventing the downregulation of the KCC2 phosphorylation at Ser940 and hence the KCC2 membrane expression during convulsant stimulation, and consequently restored the GABA inhibition. In addition, while CLP290 was given in early epileptogenesis period, it also effectively decreased the spontaneous recurrent seizures. Generally, our current results demonstrated that CLP290, as a specific KCC2 modulator by enhancing KCC2 function, not only inhibits the occurrence of the ictal seizures, but also suppresses the epileptogenic process. Therefore, we believe KCC2 may be a suitable target for future anti-epileptic drug development.
Collapse
Affiliation(s)
- Jingyi Cai
- Department of Neurology, Institutes of Brain Science, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Zhongshan Hospital, Fudan University, Shanghai 200032, China
| | - Zhuoyi Wu
- Department of Neurology, Institutes of Brain Science, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Zhongshan Hospital, Fudan University, Shanghai 200032, China
| | - Guoxiang Wang
- Department of Neurology, Institutes of Brain Science, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Zhongshan Hospital, Fudan University, Shanghai 200032, China
| | - Xiran Zhao
- Department of Neurology, Institutes of Brain Science, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Zhongshan Hospital, Fudan University, Shanghai 200032, China
| | - Xiaohan Wang
- Department of Neurology, Institutes of Brain Science, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Zhongshan Hospital, Fudan University, Shanghai 200032, China
| | - Benjamin H Wang
- Department of Neurology, Institutes of Brain Science, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Zhongshan Hospital, Fudan University, Shanghai 200032, China
| | - Jiangning Yu
- Department of Neurology, Institutes of Brain Science, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Zhongshan Hospital, Fudan University, Shanghai 200032, China
| | - Xu Liu
- Department of Neurology, Institutes of Brain Science, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Zhongshan Hospital, Fudan University, Shanghai 200032, China.
| | - Yun Wang
- Department of Neurology, Institutes of Brain Science, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Zhongshan Hospital, Fudan University, Shanghai 200032, China.
| |
Collapse
|
7
|
Kok M, Brodsky JL. The biogenesis of potassium transporters: implications of disease-associated mutations. Crit Rev Biochem Mol Biol 2024; 59:154-198. [PMID: 38946646 PMCID: PMC11444911 DOI: 10.1080/10409238.2024.2369986] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2024] [Revised: 06/02/2024] [Accepted: 06/16/2024] [Indexed: 07/02/2024]
Abstract
The concentration of intracellular and extracellular potassium is tightly regulated due to the action of various ion transporters, channels, and pumps, which reside primarily in the kidney. Yet, potassium transporters and cotransporters play vital roles in all organs and cell types. Perhaps not surprisingly, defects in the biogenesis, function, and/or regulation of these proteins are linked to range of catastrophic human diseases, but to date, few drugs have been approved to treat these maladies. In this review, we discuss the structure, function, and activity of a group of potassium-chloride cotransporters, the KCCs, as well as the related sodium-potassium-chloride cotransporters, the NKCCs. Diseases associated with each of the four KCCs and two NKCCs are also discussed. Particular emphasis is placed on how these complex membrane proteins fold and mature in the endoplasmic reticulum, how non-native forms of the cotransporters are destroyed in the cell, and which cellular factors oversee their maturation and transport to the cell surface. When known, we also outline how the levels and activities of each cotransporter are regulated. Open questions in the field and avenues for future investigations are further outlined.
Collapse
Affiliation(s)
- Morgan Kok
- Department of Biological Sciences, University of Pittsburgh, Pittsburgh, PA, USA
| | - Jeffrey L Brodsky
- Department of Biological Sciences, University of Pittsburgh, Pittsburgh, PA, USA
| |
Collapse
|
8
|
McMoneagle E, Zhou J, Zhang S, Huang W, Josiah SS, Ding K, Wang Y, Zhang J. Neuronal K +-Cl - cotransporter KCC2 as a promising drug target for epilepsy treatment. Acta Pharmacol Sin 2024; 45:1-22. [PMID: 37704745 PMCID: PMC10770335 DOI: 10.1038/s41401-023-01149-9] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/12/2023] [Accepted: 08/02/2023] [Indexed: 09/14/2023]
Abstract
Epilepsy is a prevalent neurological disorder characterized by unprovoked seizures. γ-Aminobutyric acid (GABA) serves as the primary fast inhibitory neurotransmitter in the brain, and GABA binding to the GABAA receptor (GABAAR) regulates Cl- and bicarbonate (HCO3-) influx or efflux through the channel pore, leading to GABAergic inhibition or excitation, respectively. The neuron-specific K+-Cl- cotransporter 2 (KCC2) is essential for maintaining a low intracellular Cl- concentration, ensuring GABAAR-mediated inhibition. Impaired KCC2 function results in GABAergic excitation associated with epileptic activity. Loss-of-function mutations and altered expression of KCC2 lead to elevated [Cl-]i and compromised synaptic inhibition, contributing to epilepsy pathogenesis in human patients. KCC2 antagonism studies demonstrate the necessity of limiting neuronal hyperexcitability within the brain, as reduced KCC2 functioning leads to seizure activity. Strategies focusing on direct (enhancing KCC2 activation) and indirect KCC2 modulation (altering KCC2 phosphorylation and transcription) have proven effective in attenuating seizure severity and exhibiting anti-convulsant properties. These findings highlight KCC2 as a promising therapeutic target for treating epilepsy. Recent advances in understanding KCC2 regulatory mechanisms, particularly via signaling pathways such as WNK, PKC, BDNF, and its receptor TrkB, have led to the discovery of novel small molecules that modulate KCC2. Inhibiting WNK kinase or utilizing newly discovered KCC2 agonists has demonstrated KCC2 activation and seizure attenuation in animal models. This review discusses the role of KCC2 in epilepsy and evaluates its potential as a drug target for epilepsy treatment by exploring various strategies to regulate KCC2 activity.
Collapse
Affiliation(s)
- Erin McMoneagle
- Institute of Biomedical and Clinical Sciences, Medical School, Faculty of Health and Life Sciences, University of Exeter, Hatherly Laboratories, Streatham Campus, Exeter, EX4 4PS, UK
| | - Jin Zhou
- Department of Neurology, Institutes of Brain Science, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institute of Biological Science, Zhongshan Hospital, Fudan University, Shanghai, 200032, China
| | - Shiyao Zhang
- Institute of Cardiovascular Diseases, Xiamen Cardiovascular Hospital Xiamen University, School of Medicine, Xiamen University, Xiang'an Nan Lu, Xiamen, 361102, China
| | - Weixue Huang
- State Key Laboratory of Chemical Biology, Research Center of Chemical Kinomics, Shanghai Institute of Organic Chemistry, Chinese Academy of Sciences, Shanghai, 200032, China
| | - Sunday Solomon Josiah
- Institute of Biomedical and Clinical Sciences, Medical School, Faculty of Health and Life Sciences, University of Exeter, Hatherly Laboratories, Streatham Campus, Exeter, EX4 4PS, UK
| | - Ke Ding
- State Key Laboratory of Chemical Biology, Research Center of Chemical Kinomics, Shanghai Institute of Organic Chemistry, Chinese Academy of Sciences, Shanghai, 200032, China.
| | - Yun Wang
- Department of Neurology, Institutes of Brain Science, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institute of Biological Science, Zhongshan Hospital, Fudan University, Shanghai, 200032, China.
| | - Jinwei Zhang
- Institute of Biomedical and Clinical Sciences, Medical School, Faculty of Health and Life Sciences, University of Exeter, Hatherly Laboratories, Streatham Campus, Exeter, EX4 4PS, UK.
- Institute of Cardiovascular Diseases, Xiamen Cardiovascular Hospital Xiamen University, School of Medicine, Xiamen University, Xiang'an Nan Lu, Xiamen, 361102, China.
- State Key Laboratory of Chemical Biology, Research Center of Chemical Kinomics, Shanghai Institute of Organic Chemistry, Chinese Academy of Sciences, Shanghai, 200032, China.
| |
Collapse
|
9
|
van van Hugte EJH, Schubert D, Nadif Kasri N. Excitatory/inhibitory balance in epilepsies and neurodevelopmental disorders: Depolarizing γ-aminobutyric acid as a common mechanism. Epilepsia 2023; 64:1975-1990. [PMID: 37195166 DOI: 10.1111/epi.17651] [Citation(s) in RCA: 30] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2022] [Revised: 05/13/2023] [Accepted: 05/15/2023] [Indexed: 05/18/2023]
Abstract
Epilepsy is one of the most common neurological disorders. Although many factors contribute to epileptogenesis, seizure generation is mostly linked to hyperexcitability due to alterations in excitatory/inhibitory (E/I) balance. The common hypothesis is that reduced inhibition, increased excitation, or both contribute to the etiology of epilepsy. Increasing evidence shows that this view is oversimplistic, and that increased inhibition through depolarizing γ-aminobutyric acid (GABA) similarly contributes to epileptogenisis. In early development, GABA signaling is depolarizing, inducing outward Cl- currents due to high intracellular Cl- concentrations. During maturation, the mechanisms of GABA action shift from depolarizing to hyperpolarizing, a critical event during brain development. Altered timing of this shift is associated with both neurodevelopmental disorders and epilepsy. Here, we consider the different ways that depolarizing GABA contributes to altered E/I balance and epileptogenesis, and discuss that alterations in depolarizing GABA could be a common denominator underlying seizure generation in neurodevelopmental disorders and epilepsies.
Collapse
Affiliation(s)
- Eline J H van van Hugte
- Department of Human Genetics, Radboud University Medical Center, Donders Institute for Brain, Cognition, and Behavior, Nijmegen, the Netherlands
- Department of Epileptology, Academic Centre for Epileptology (ACE) Kempenhaeghe, Heeze, the Netherlands
| | - Dirk Schubert
- Department of Cognitive Neuroscience, Radboud University Medical Center, Donders Institute for Brain, Cognition, and Behavior, Nijmegen, the Netherlands
| | - Nael Nadif Kasri
- Department of Human Genetics, Radboud University Medical Center, Donders Institute for Brain, Cognition, and Behavior, Nijmegen, the Netherlands
- Department of Epileptology, Academic Centre for Epileptology (ACE) Kempenhaeghe, Heeze, the Netherlands
| |
Collapse
|
10
|
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
|
11
|
Talifu Z, Pan Y, Gong H, Xu X, Zhang C, Yang D, Gao F, Yu Y, Du L, Li J. The role of KCC2 and NKCC1 in spinal cord injury: From physiology to pathology. Front Physiol 2022; 13:1045520. [PMID: 36589461 PMCID: PMC9799334 DOI: 10.3389/fphys.2022.1045520] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2022] [Accepted: 12/05/2022] [Indexed: 12/23/2022] Open
Abstract
The balance of ion concentrations inside and outside the cell is an essential homeostatic mechanism in neurons and serves as the basis for a variety of physiological activities. In the central nervous system, NKCC1 and KCC2, members of the SLC12 cation-chloride co-transporter (CCC) family, participate in physiological and pathophysiological processes by regulating intracellular and extracellular chloride ion concentrations, which can further regulate the GABAergic system. Over recent years, studies have shown that NKCC1 and KCC2 are essential for the maintenance of Cl- homeostasis in neural cells. NKCC1 transports Cl- into cells while KCC2 transports Cl- out of cells, thereby regulating chloride balance and neuronal excitability. An imbalance of NKCC1 and KCC2 after spinal cord injury will disrupt CI- homeostasis, resulting in the transformation of GABA neurons from an inhibitory state into an excitatory state, which subsequently alters the spinal cord neural network and leads to conditions such as spasticity and neuropathic pain, among others. Meanwhile, studies have shown that KCC2 is also an essential target for motor function reconstruction after spinal cord injury. This review mainly introduces the physiological structure and function of NKCC1 and KCC2 and discusses their pathophysiological roles after spinal cord injury.
Collapse
Affiliation(s)
- Zuliyaer Talifu
- School of Rehabilitation, Capital Medical University, Beijing, China,Department of Spinal and Neural Functional Reconstruction, China Rehabilitation Research Center, Beijing, China,Chinese Institute of Rehabilitation Science, Beijing, China,Center of Neural Injury and Repair, Beijing Institute for Brain Disorders, Beijing, China,Beijing Key Laboratory of Neural Injury and Rehabilitation, Beijing, China,School of Rehabilitation Sciences and Engineering, University of Health and Rehabilitation Sciences, Qingdao, China
| | - Yunzhu Pan
- School of Rehabilitation, Capital Medical University, Beijing, China,Department of Spinal and Neural Functional Reconstruction, China Rehabilitation Research Center, Beijing, China,Chinese Institute of Rehabilitation Science, Beijing, China,Center of Neural Injury and Repair, Beijing Institute for Brain Disorders, Beijing, China,Beijing Key Laboratory of Neural Injury and Rehabilitation, Beijing, China,School of Rehabilitation Sciences and Engineering, University of Health and Rehabilitation Sciences, Qingdao, China
| | - Han Gong
- School of Rehabilitation, Capital Medical University, Beijing, China,Department of Spinal and Neural Functional Reconstruction, China Rehabilitation Research Center, Beijing, China,Chinese Institute of Rehabilitation Science, Beijing, China,Center of Neural Injury and Repair, Beijing Institute for Brain Disorders, Beijing, China,Beijing Key Laboratory of Neural Injury and Rehabilitation, Beijing, China
| | - Xin Xu
- School of Rehabilitation, Capital Medical University, Beijing, China,Department of Spinal and Neural Functional Reconstruction, China Rehabilitation Research Center, Beijing, China,Chinese Institute of Rehabilitation Science, Beijing, China,Center of Neural Injury and Repair, Beijing Institute for Brain Disorders, Beijing, China,Beijing Key Laboratory of Neural Injury and Rehabilitation, Beijing, China
| | - Chunjia Zhang
- School of Rehabilitation, Capital Medical University, Beijing, China,Department of Spinal and Neural Functional Reconstruction, China Rehabilitation Research Center, Beijing, China,Chinese Institute of Rehabilitation Science, Beijing, China,Center of Neural Injury and Repair, Beijing Institute for Brain Disorders, Beijing, China,Beijing Key Laboratory of Neural Injury and Rehabilitation, Beijing, China
| | - Degang Yang
- School of Rehabilitation, Capital Medical University, Beijing, China,Department of Spinal and Neural Functional Reconstruction, China Rehabilitation Research Center, Beijing, China,Center of Neural Injury and Repair, Beijing Institute for Brain Disorders, Beijing, China,Beijing Key Laboratory of Neural Injury and Rehabilitation, Beijing, China
| | - Feng Gao
- School of Rehabilitation, Capital Medical University, Beijing, China,Department of Spinal and Neural Functional Reconstruction, China Rehabilitation Research Center, Beijing, China,Center of Neural Injury and Repair, Beijing Institute for Brain Disorders, Beijing, China,Beijing Key Laboratory of Neural Injury and Rehabilitation, Beijing, China
| | - Yan Yu
- School of Rehabilitation, Capital Medical University, Beijing, China,Chinese Institute of Rehabilitation Science, Beijing, China,Center of Neural Injury and Repair, Beijing Institute for Brain Disorders, Beijing, China,Beijing Key Laboratory of Neural Injury and Rehabilitation, Beijing, China
| | - Liangjie Du
- School of Rehabilitation, Capital Medical University, Beijing, China,Department of Spinal and Neural Functional Reconstruction, China Rehabilitation Research Center, Beijing, China,Center of Neural Injury and Repair, Beijing Institute for Brain Disorders, Beijing, China,Beijing Key Laboratory of Neural Injury and Rehabilitation, Beijing, China,*Correspondence: Liangjie Du, ; Jianjun Li,
| | - Jianjun Li
- School of Rehabilitation, Capital Medical University, Beijing, China,Department of Spinal and Neural Functional Reconstruction, China Rehabilitation Research Center, Beijing, China,Chinese Institute of Rehabilitation Science, Beijing, China,Center of Neural Injury and Repair, Beijing Institute for Brain Disorders, Beijing, China,Beijing Key Laboratory of Neural Injury and Rehabilitation, Beijing, China,School of Rehabilitation Sciences and Engineering, University of Health and Rehabilitation Sciences, Qingdao, China,*Correspondence: Liangjie Du, ; Jianjun Li,
| |
Collapse
|
12
|
Proskurina EY, Zaitsev AV. Regulation of Potassium and Chloride Concentrations in Nervous Tissue as a Method of Anticonvulsant Therapy. J EVOL BIOCHEM PHYS+ 2022. [DOI: 10.1134/s0022093022050015] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
Abstract
Under some pathological conditions, such as pharmacoresistant
epilepsy, status epilepticus or certain forms of genetic abnormalities,
spiking activity of GABAergic interneurons may enhance excitation
processes in neuronal circuits and provoke the generation of ictal
discharges. As a result, anticonvulsants acting on the GABAergic
system may be ineffective or even increase seizure activity. This
paradoxical effect of the inhibitory system is due to ionic imbalances
in nervous tissue. This review addresses the mechanisms of ictal
discharge initiation in neuronal networks due to the imbalance of
chloride and potassium ions, as well as possible ways to regulate
ionic concentrations. Both the enhancement (or attenuation) of the
activity of certain neuronal ion transporters and ion pumps and
their additional expression via gene therapy can be effective in
suppressing seizure activity caused by ionic imbalances. The Na+–K+-pump,
NKCC1 and KCC2 cotransporters are important for maintaining proper
K+ and Cl– concentrations
in nervous tissue, having been repeatedly considered as pharmacological
targets for antiepileptic exposures. Further progress in this direction
is hampered by the lack of sufficiently selective pharmacological
tools and methods for providing effective drug delivery to the epileptic
focus. The use of the gene therapy techniques, such as overexpressing
of the KCC2 transporter in the epileptic focus, seems to be a more promising
approach. Another possible direction could be the use of optogenetic
tools, namely specially designed light-activated ion pumps or ion
channels. In this case, photon energy can be used to create the
required gradients of chloride and potassium ions, although these
methods also have significant limitations which complicate their
rapid introduction into medicine.
Collapse
|
13
|
Hartmann AM, Nothwang HG. NKCC1 and KCC2: Structural insights into phospho-regulation. Front Mol Neurosci 2022; 15:964488. [PMID: 35935337 PMCID: PMC9355526 DOI: 10.3389/fnmol.2022.964488] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2022] [Accepted: 07/06/2022] [Indexed: 11/13/2022] Open
Abstract
Inhibitory neurotransmission plays a fundamental role in the central nervous system, with about 30–50% of synaptic connections being inhibitory. The action of both inhibitory neurotransmitter, gamma-aminobutyric-acid (GABA) and glycine, mainly relies on the intracellular Cl– concentration in neurons. This is set by the interplay of the cation chloride cotransporters NKCC1 (Na+, K+, Cl– cotransporter), a main Cl– uptake transporter, and KCC2 (K+, Cl– cotransporter), the principle Cl– extruder in neurons. Accordingly, their dysfunction is associated with severe neurological, psychiatric, and neurodegenerative disorders. This has triggered great interest in understanding their regulation, with a strong focus on phosphorylation. Recent structural data by cryogenic electron microscopy provide the unique possibility to gain insight into the action of these phosphorylations. Interestingly, in KCC2, six out of ten (60%) known regulatory phospho-sites reside within a region of 134 amino acid residues (12% of the total residues) between helices α8 and α9 that lacks fixed or ordered three-dimensional structures. It thus represents a so-called intrinsically disordered region. Two further phospho-sites, Tyr903 and Thr906, are also located in a disordered region between the ß8 strand and the α8 helix. We make the case that especially the disordered region between helices α8 and α9 acts as a platform to integrate different signaling pathways and simultaneously constitute a flexible, highly dynamic linker that can survey a wide variety of distinct conformations. As each conformation can have distinct binding affinities and specificity properties, this enables regulation of [Cl–]i and thus the ionic driving force in a history-dependent way. This region might thus act as a molecular processor underlying the well described phenomenon of ionic plasticity that has been ascribed to inhibitory neurotransmission. Finally, it might explain the stunning long-range effects of mutations on phospho-sites in KCC2.
Collapse
Affiliation(s)
- Anna-Maria Hartmann
- Division of Neurogenetics, School of Medicine and Health Sciences, Carl von Ossietzky University Oldenburg, Oldenburg, Germany
- Research Center for Neurosensory Sciences, Carl von Ossietzky University Oldenburg, Oldenburg, Germany
- *Correspondence: Anna-Maria Hartmann,
| | - Hans Gerd Nothwang
- Division of Neurogenetics, School of Medicine and Health Sciences, Carl von Ossietzky University Oldenburg, Oldenburg, Germany
- Research Center for Neurosensory Sciences, Carl von Ossietzky University Oldenburg, Oldenburg, Germany
- Center of Excellence Hearing4all, Carl von Ossietzky University Oldenburg, Oldenburg, Germany
| |
Collapse
|
14
|
Hui KK, Chater TE, Goda Y, Tanaka M. How Staying Negative Is Good for the (Adult) Brain: Maintaining Chloride Homeostasis and the GABA-Shift in Neurological Disorders. Front Mol Neurosci 2022; 15:893111. [PMID: 35875665 PMCID: PMC9305173 DOI: 10.3389/fnmol.2022.893111] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2022] [Accepted: 06/10/2022] [Indexed: 01/27/2023] Open
Abstract
Excitatory-inhibitory (E-I) imbalance has been shown to contribute to the pathogenesis of a wide range of neurodevelopmental disorders including autism spectrum disorders, epilepsy, and schizophrenia. GABA neurotransmission, the principal inhibitory signal in the mature brain, is critically coupled to proper regulation of chloride homeostasis. During brain maturation, changes in the transport of chloride ions across neuronal cell membranes act to gradually change the majority of GABA signaling from excitatory to inhibitory for neuronal activation, and dysregulation of this GABA-shift likely contributes to multiple neurodevelopmental abnormalities that are associated with circuit dysfunction. Whilst traditionally viewed as a phenomenon which occurs during brain development, recent evidence suggests that this GABA-shift may also be involved in neuropsychiatric disorders due to the "dematuration" of affected neurons. In this review, we will discuss the cell signaling and regulatory mechanisms underlying the GABA-shift phenomenon in the context of the latest findings in the field, in particular the role of chloride cotransporters NKCC1 and KCC2, and furthermore how these regulatory processes are altered in neurodevelopmental and neuropsychiatric disorders. We will also explore the interactions between GABAergic interneurons and other cell types in the developing brain that may influence the GABA-shift. Finally, with a greater understanding of how the GABA-shift is altered in pathological conditions, we will briefly outline recent progress on targeting NKCC1 and KCC2 as a therapeutic strategy against neurodevelopmental and neuropsychiatric disorders associated with improper chloride homeostasis and GABA-shift abnormalities.
Collapse
Affiliation(s)
- Kelvin K. Hui
- Department of Developmental and Molecular Biology, Albert Einstein College of Medicine, Bronx, NY, United States
- Institute for Aging Research, Albert Einstein College of Medicine, Bronx, NY, United States
| | - Thomas E. Chater
- Laboratory for Synaptic Plasticity and Connectivity, RIKEN Center for Brain Science, Wako, Japan
| | - Yukiko Goda
- Laboratory for Synaptic Plasticity and Connectivity, RIKEN Center for Brain Science, Wako, Japan
- Synapse Biology Unit, Okinawa Institute for Science and Technology Graduate University, Onna, Japan
| | - Motomasa Tanaka
- Laboratory for Protein Conformation Diseases, RIKEN Center for Brain Science, Wako, Japan
| |
Collapse
|
15
|
Hills KE, Kostarelos K, Wykes RC. Converging Mechanisms of Epileptogenesis and Their Insight in Glioblastoma. Front Mol Neurosci 2022; 15:903115. [PMID: 35832394 PMCID: PMC9271928 DOI: 10.3389/fnmol.2022.903115] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2022] [Accepted: 05/25/2022] [Indexed: 12/15/2022] Open
Abstract
Glioblastoma (GBM) is the most common and advanced form of primary malignant tumor occurring in the adult central nervous system, and it is frequently associated with epilepsy, a debilitating comorbidity. Seizures are observed both pre- and post-surgical resection, indicating that several pathophysiological mechanisms are shared but also prompting questions about how the process of epileptogenesis evolves throughout GBM progression. Molecular mutations commonly seen in primary GBM, i.e., in PTEN and p53, and their associated downstream effects are known to influence seizure likelihood. Similarly, various intratumoral mechanisms, such as GBM-induced blood-brain barrier breakdown and glioma-immune cell interactions within the tumor microenvironment are also cited as contributing to network hyperexcitability. Substantial alterations to peri-tumoral glutamate and chloride transporter expressions, as well as widespread dysregulation of GABAergic signaling are known to confer increased epileptogenicity and excitotoxicity. The abnormal characteristics of GBM alter neuronal network function to result in metabolically vulnerable and hyperexcitable peri-tumoral tissue, properties the tumor then exploits to favor its own growth even post-resection. It is evident that there is a complex, dynamic interplay between GBM and epilepsy that promotes the progression of both pathologies. This interaction is only more complicated by the concomitant presence of spreading depolarization (SD). The spontaneous, high-frequency nature of GBM-associated epileptiform activity and SD-associated direct current (DC) shifts require technologies capable of recording brain signals over a wide bandwidth, presenting major challenges for comprehensive electrophysiological investigations. This review will initially provide a detailed examination of the underlying mechanisms that promote network hyperexcitability in GBM. We will then discuss how an investigation of these pathologies from a network level, and utilization of novel electrophysiological tools, will yield a more-effective, clinically-relevant understanding of GBM-related epileptogenesis. Further to this, we will evaluate the clinical relevance of current preclinical research and consider how future therapeutic advancements may impact the bidirectional relationship between GBM, SDs, and seizures.
Collapse
Affiliation(s)
- Kate E. Hills
- Nanomedicine Lab, Faculty of Biology, Medicine and Health, University of Manchester, Manchester, United Kingdom
| | - Kostas Kostarelos
- Nanomedicine Lab, Faculty of Biology, Medicine and Health, University of Manchester, Manchester, United Kingdom
- Catalan Institute for Nanoscience and Nanotechnology (ICN2), Edifici ICN2, Campus UAB, Barcelona, Spain
| | - Robert C. Wykes
- Nanomedicine Lab, Faculty of Biology, Medicine and Health, University of Manchester, Manchester, United Kingdom
- Department of Clinical and Experimental Epilepsy, UCL Queen Square Institute of Neurology, London, United Kingdom
- *Correspondence: Robert C. Wykes
| |
Collapse
|
16
|
Chen L, Yu J, Wan L, Wu Z, Wang G, Hu Z, Ren L, Zhou J, Qian B, Zhao X, Zhang J, Liu X, Wang Y. Furosemide prevents membrane KCC2 downregulation during convulsant stimulation in the hippocampus. IBRO Neurosci Rep 2022; 12:355-365. [PMID: 35746976 PMCID: PMC9210493 DOI: 10.1016/j.ibneur.2022.04.010] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2022] [Revised: 04/15/2022] [Accepted: 04/25/2022] [Indexed: 02/08/2023] Open
Abstract
In adults, γ-aminobutyric acid (GABA) type A receptor (GABAAR)-mediated inhibition depends on the maintenance of low intracellular chloride anion concentration through neuron-specific potassium-chloride cotransporter-2 (KCC2). KCC2 has been widely reported to have a plasticity change during the course of epilepsy development, with an early downregulation and late recovery in neuronal cell membranes after epileptic stimulation, which facilitates epileptiform burst activity. Furosemide is a clinical loop diuretic that inhibits KCC2. Here, we first confirmed that furosemide pretreatment could effectively prevented convulsant stimulation-induced neuronal membrane KCC2 downregulation in the hippocampus in both in vivo and in vitro cyclothiazide-induced seizure model. Second, we verified that furosemide pretreatment rescued KCC2 function deficits, as indicated by E GABA depolarizing shift and GABAAR inhibitory function impairment induced via cyclothiazide treatment. Further, we demonstrated that furosemide also suppressed cyclothiazide-induced epileptiform burst activity in cultured hippocampal neurons and lowered the mortality rate during acute seizure induction. Overall, furosemide prevents membrane KCC2 downregulation during acute seizure induction, restores KCC2-mediated GABA inhibition, and interrupts the progression from acute seizure to epileptogenesis.
Collapse
Affiliation(s)
- Lulan Chen
- Department of Neurology, Institutes of Brain Science, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institute of Biological Science, Zhongshan Hospital, Fudan University, Shanghai 200032, China
| | - Jiangning Yu
- Department of Neurology, Institutes of Brain Science, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institute of Biological Science, Zhongshan Hospital, Fudan University, Shanghai 200032, China
| | - Li Wan
- Department of Neurology, Institutes of Brain Science, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institute of Biological Science, Zhongshan Hospital, Fudan University, Shanghai 200032, China
- Rehabilitation Center, Shenzhen Second People's Hospital/ the First Affiliated Hospital of Shenzhen University Health Science Center, Shenzhen 518035, China Institute of
| | - Zheng Wu
- Department of Neurology, Institutes of Brain Science, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institute of Biological Science, Zhongshan Hospital, Fudan University, Shanghai 200032, China
| | - Guoxiang Wang
- Department of Neurology, Institutes of Brain Science, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institute of Biological Science, Zhongshan Hospital, Fudan University, Shanghai 200032, China
| | - Zihan Hu
- Department of Anesthesiology, Shanghai Tenth People’s Hospital, Tongji University School of Medicine, Shanghai, China
| | - Liang Ren
- Department of Neurology, Institutes of Brain Science, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institute of Biological Science, Zhongshan Hospital, Fudan University, Shanghai 200032, China
| | - Jing Zhou
- Department of Neurology, Institutes of Brain Science, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institute of Biological Science, Zhongshan Hospital, Fudan University, Shanghai 200032, China
- Rehabilitation Center, Shenzhen Second People's Hospital/ the First Affiliated Hospital of Shenzhen University Health Science Center, Shenzhen 518035, China Institute of
| | - Binbin Qian
- Department of Neurology, Institutes of Brain Science, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institute of Biological Science, Zhongshan Hospital, Fudan University, Shanghai 200032, China
| | - Xuan Zhao
- Department of Anesthesiology, Shanghai Tenth People’s Hospital, Tongji University School of Medicine, Shanghai, China
| | - Jinwei Zhang
- Biomedical and Clinical Sciences, Medical School, College of Medicine and Health, University of Exeter, Hatherly Laboratories, Exeter EX4 4PS, UK
| | - Xu Liu
- Department of Neurology, Institutes of Brain Science, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institute of Biological Science, Zhongshan Hospital, Fudan University, Shanghai 200032, China
| | - Yun Wang
- Department of Neurology, Institutes of Brain Science, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institute of Biological Science, Zhongshan Hospital, Fudan University, Shanghai 200032, China
| |
Collapse
|
17
|
Liedtke W. Long March Toward Safe and Effective Analgesia by Enhancing Gene Expression of Kcc2: First Steps Taken. Front Mol Neurosci 2022; 15:865600. [PMID: 35645734 PMCID: PMC9137411 DOI: 10.3389/fnmol.2022.865600] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2022] [Accepted: 03/08/2022] [Indexed: 11/15/2022] Open
Abstract
Low intraneuronal chloride in spinal cord dorsal horn pain relay neurons is critical for physiologic transmission of primary pain afferents because low intraneuronal chloride dictates whether GABA-ergic and glycin-ergic neurotransmission is inhibitory. If the neuronal chloride elevates to pathologic levels, then spinal cord primary pain relay becomes leaky and exhibits the behavioral hallmarks of pathologic pain, namely hypersensitivity and allodynia. Low chloride in spinal cord dorsal horn neurons is maintained by proper gene expression of Kcc2 and sustained physiologic function of the KCC2 chloride extruding electroneutral transporter. Peripheral nerve injury and other forms of neural injury evoke greatly diminished Kcc2 gene expression and subsequent corruption of inhibitory neurotransmission in the spinal cord dorsal horn, thus causing derailment of the gate function for pain. Here I review key discoveries that have helped us understand these fundamentals, and focus on recent insights relating to the discovery of Kcc2 gene expression enhancing compounds via compound screens in neurons. One such study characterized the kinase inhibitor, kenpaullone, more in-depth, revealing its function as a robust and long-lasting analgesic in preclinical models of nerve injury and cancer bone pain, also elucidating its mechanism of action via GSK3β inhibition, diminishing delta-catenin phosphorylation, and facilitating its nuclear transfer and subsequent enhancement of Kcc2 gene expression by de-repressing Kaiso epigenetic transcriptional regulator. Future directions re Kcc2 gene expression enhancement are discussed, namely combination with other analgesics and analgesic methods, such as spinal cord stimulation and electroacupuncture, gene therapy, and leveraging Kcc2 gene expression-enhancing nanomaterials.
Collapse
|
18
|
Why won't it stop? The dynamics of benzodiazepine resistance in status epilepticus. Nat Rev Neurol 2022; 18:428-441. [PMID: 35538233 DOI: 10.1038/s41582-022-00664-3] [Citation(s) in RCA: 31] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/19/2022] [Indexed: 11/08/2022]
Abstract
Status epilepticus is a life-threatening neurological emergency that affects both adults and children. Approximately 36% of episodes of status epilepticus do not respond to the current preferred first-line treatment, benzodiazepines. The proportion of episodes that are refractory to benzodiazepines is higher in low-income and middle-income countries (LMICs) than in high-income countries (HICs). Evidence suggests that longer episodes of status epilepticus alter brain physiology, thereby contributing to the emergence of benzodiazepine resistance. Such changes include alterations in GABAA receptor function and in the transmembrane gradient for chloride, both of which erode the ability of benzodiazepines to enhance inhibitory synaptic signalling. Often, current management guidelines for status epilepticus do not account for these duration-related changes in pathophysiology, which might differentially impact individuals in LMICs, where the average time taken to reach medical attention is longer than in HICs. In this Perspective article, we aim to combine clinical insights and the latest evidence from basic science to inspire a new, context-specific approach to efficiently managing status epilepticus.
Collapse
|
19
|
Lam P, Vinnakota C, Guzmán BCF, Newland J, Peppercorn K, Tate WP, Waldvogel HJ, Faull RLM, Kwakowsky A. Beta-Amyloid (Aβ 1-42) Increases the Expression of NKCC1 in the Mouse Hippocampus. Molecules 2022; 27:2440. [PMID: 35458638 PMCID: PMC9027496 DOI: 10.3390/molecules27082440] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2022] [Revised: 03/22/2022] [Accepted: 04/06/2022] [Indexed: 11/16/2022] Open
Abstract
Alzheimer's disease (AD) is a neurodegenerative disorder with an increasing need for developing disease-modifying treatments as current therapies only provide marginal symptomatic relief. Recent evidence suggests the γ-aminobutyric acid (GABA) neurotransmitter system undergoes remodeling in AD, disrupting the excitatory/inhibitory (E/I) balance in the brain. Altered expression levels of K-Cl-2 (KCC2) and N-K-Cl-1 (NKCC1), which are cation-chloride cotransporters (CCCs), have been implicated in disrupting GABAergic activity by regulating GABAA receptor signaling polarity in several neurological disorders, but these have not yet been explored in AD. NKCC1 and KCC2 regulate intracellular chloride [Cl-]i by accumulating and extruding Cl-, respectively. Increased NKCC1 expression in mature neurons has been reported in these disease conditions, and bumetanide, an NKCC1 inhibitor, is suggested to show potential therapeutic benefits. This study used primary mouse hippocampal neurons to explore if KCC2 and NKCC1 expression levels are altered following beta-amyloid (Aβ1-42) treatment and the potential neuroprotective effects of bumetanide. KCC2 and NKCC1 expression levels were also examined in 18-months-old male C57BL/6 mice following bilateral hippocampal Aβ1-42 stereotaxic injection. No change in KCC2 and NKCC1 expression levels were observed in mouse hippocampal neurons treated with 1 nM Aβ1-42, but NKCC1 expression increased 30-days post-Aβ1-42-injection in the CA1 region of the mouse hippocampus. Primary mouse hippocampal cultures were treated with 1 nM Aβ1-42 alone or with various concentrations of bumetanide (1 µM, 10 µM, 100 µM, 1 mM) to investigate the effect of the drug on cell viability. Aβ1-42 produced 53.1 ± 1.4% cell death after 5 days, and the addition of bumetanide did not reduce this. However, the drug at all concentrations significantly reduced cell viability, suggesting bumetanide is highly neurotoxic. In summary, these results suggest that chronic exposure to Aβ1-42 alters the balance of KCC2 and NKCC1 expression in a region-and layer-specific manner in mouse hippocampal tissue; therefore, this process most likely contributes to altered hippocampal E/I balance in this model. Furthermore, bumetanide induces hippocampal neurotoxicity, thus questioning its suitability for AD therapy. Further investigations are required to examine the effects of Aβ1-42 on KCC2 and NKCC1 expression and whether targeting CCCs might offer a therapeutic approach for AD.
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; (P.L.); (C.V.); (B.C.-F.G.); (J.N.); (H.J.W.); (R.L.M.F.)
| | - Chitra Vinnakota
- Centre for Brain Research, Department of Anatomy and Medical Imaging, Faculty of Medical and Health Sciences, University of Auckland, Auckland 1023, New Zealand; (P.L.); (C.V.); (B.C.-F.G.); (J.N.); (H.J.W.); (R.L.M.F.)
| | - Beatriz Calvo-Flores Guzmán
- Centre for Brain Research, Department of Anatomy and Medical Imaging, Faculty of Medical and Health Sciences, University of Auckland, Auckland 1023, New Zealand; (P.L.); (C.V.); (B.C.-F.G.); (J.N.); (H.J.W.); (R.L.M.F.)
| | - 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; (P.L.); (C.V.); (B.C.-F.G.); (J.N.); (H.J.W.); (R.L.M.F.)
| | - Katie Peppercorn
- Department of Biochemistry, University of Otago, Dunedin 9054, New Zealand; (K.P.); (W.P.T.)
| | - Warren P. Tate
- Department of Biochemistry, University of Otago, Dunedin 9054, New Zealand; (K.P.); (W.P.T.)
| | - Henry J. Waldvogel
- Centre for Brain Research, Department of Anatomy and Medical Imaging, Faculty of Medical and Health Sciences, University of Auckland, Auckland 1023, New Zealand; (P.L.); (C.V.); (B.C.-F.G.); (J.N.); (H.J.W.); (R.L.M.F.)
| | - 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; (P.L.); (C.V.); (B.C.-F.G.); (J.N.); (H.J.W.); (R.L.M.F.)
| | - 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; (P.L.); (C.V.); (B.C.-F.G.); (J.N.); (H.J.W.); (R.L.M.F.)
- Pharmacology and Therapeutics, Galway Neuroscience Centre, School of Medicine, National University of Ireland Galway, H91 W5P7 Galway, Ireland
| |
Collapse
|
20
|
Shen Q, Wu X, Zhang Z, Zhang D, Yang S, Xing D. Gamma frequency light flicker regulates amyloid precursor protein trafficking for reducing β-amyloid load in Alzheimer's disease model. Aging Cell 2022; 21:e13573. [PMID: 35199454 PMCID: PMC8920449 DOI: 10.1111/acel.13573] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2021] [Revised: 01/05/2022] [Accepted: 02/02/2022] [Indexed: 11/28/2022] Open
Abstract
Inducing gamma oscillations with non‐invasive light flicker has been reported to impact Alzheimer's disease‐related pathology. However, it is unclear which signaling pathways are involved in reducing amyloid load. Here, we found that gamma frequency light flicker increased anchoring of amyloid precursor protein (APP) to the plasma membrane for non‐amyloidogenic processing, and then physically interacted with KCC2, a neuron‐specific K+‐Cl− cotransporter, suggesting that it is essential to maintain surface GABAA receptor α1 levels and reduce β‐amyloid (Aβ) production. Stimulation with such light flicker limited KCC2 internalization and subsequent degradation via both tyrosine phosphorylation and ubiquitination, leading to an increase in surface‐KCC2 levels. Specifically, PKC‐dependent phosphorylation of APP on a serine residue was induced by gamma frequency light flicker, which was responsible for maintaining plasma membrane levels of full‐length APP, leading to its reduced trafficking to endosomes and inhibiting the β‐secretase cleavage pathway. The activated PKC from the gamma frequency light flicker subsequently phosphorylated serine of KCC2 and stabilized it onto the cell surface, which contributed to the upregulation of surface GABAA receptor α1 levels. Together, these data indicate that enhancement of APP trafficking to the plasma membrane via light flicker plays a critical modulatory role in reduction of Aβ load in Alzheimer's disease.
Collapse
Affiliation(s)
- Qi Shen
- MOE Key Laboratory of Laser Life Science & Institute of Laser Life Science South China Normal University Guangzhou China
- College of Biophotonics South China Normal University Guangzhou China
| | - Xiaolei Wu
- MOE Key Laboratory of Laser Life Science & Institute of Laser Life Science South China Normal University Guangzhou China
- College of Biophotonics South China Normal University Guangzhou China
| | - Zhan Zhang
- MOE Key Laboratory of Laser Life Science & Institute of Laser Life Science South China Normal University Guangzhou China
- College of Biophotonics South China Normal University Guangzhou China
| | - Di Zhang
- MOE Key Laboratory of Laser Life Science & Institute of Laser Life Science South China Normal University Guangzhou China
- College of Biophotonics South China Normal University Guangzhou China
| | - Sihua Yang
- MOE Key Laboratory of Laser Life Science & Institute of Laser Life Science South China Normal University Guangzhou China
- College of Biophotonics South China Normal University Guangzhou China
| | - Da Xing
- MOE Key Laboratory of Laser Life Science & Institute of Laser Life Science South China Normal University Guangzhou China
- College of Biophotonics South China Normal University Guangzhou China
| |
Collapse
|
21
|
Shimizu-Okabe C, Kobayashi S, Kim J, Kosaka Y, Sunagawa M, Okabe A, Takayama C. Developmental Formation of the GABAergic and Glycinergic Networks in the Mouse Spinal Cord. Int J Mol Sci 2022; 23:ijms23020834. [PMID: 35055019 PMCID: PMC8776010 DOI: 10.3390/ijms23020834] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2021] [Revised: 01/07/2022] [Accepted: 01/11/2022] [Indexed: 12/15/2022] Open
Abstract
Gamma-aminobutyric acid (GABA) and glycine act as inhibitory neurotransmitters. Three types of inhibitory neurons and terminals, GABAergic, GABA/glycine coreleasing, and glycinergic, are orchestrated in the spinal cord neural circuits and play critical roles in regulating pain, locomotive movement, and respiratory rhythms. In this study, we first describe GABAergic and glycinergic transmission and inhibitory networks, consisting of three types of terminals in the mature mouse spinal cord. Second, we describe the developmental formation of GABAergic and glycinergic networks, with a specific focus on the differentiation of neurons, formation of synapses, maturation of removal systems, and changes in their action. GABAergic and glycinergic neurons are derived from the same domains of the ventricular zone. Initially, GABAergic neurons are differentiated, and their axons form synapses. Some of these neurons remain GABAergic in lamina I and II. Many GABAergic neurons convert to a coreleasing state. The coreleasing neurons and terminals remain in the dorsal horn, whereas many ultimately become glycinergic in the ventral horn. During the development of terminals and the transformation from radial glia to astrocytes, GABA and glycine receptor subunit compositions markedly change, removal systems mature, and GABAergic and glycinergic action shifts from excitatory to inhibitory.
Collapse
Affiliation(s)
- Chigusa Shimizu-Okabe
- Department of Molecular Anatomy, Graduate School of Medicine, University of the Ryukyus, 207 Uehara, Nishihara 903-0215, Japan; (C.S.-O.); (S.K.); (Y.K.); (M.S.)
| | - Shiori Kobayashi
- Department of Molecular Anatomy, Graduate School of Medicine, University of the Ryukyus, 207 Uehara, Nishihara 903-0215, Japan; (C.S.-O.); (S.K.); (Y.K.); (M.S.)
| | - Jeongtae Kim
- Department of Anatomy, Kosin University College of Medicine, Busan 49267, Korea;
| | - Yoshinori Kosaka
- Department of Molecular Anatomy, Graduate School of Medicine, University of the Ryukyus, 207 Uehara, Nishihara 903-0215, Japan; (C.S.-O.); (S.K.); (Y.K.); (M.S.)
| | - Masanobu Sunagawa
- Department of Molecular Anatomy, Graduate School of Medicine, University of the Ryukyus, 207 Uehara, Nishihara 903-0215, Japan; (C.S.-O.); (S.K.); (Y.K.); (M.S.)
| | - Akihito Okabe
- Department of Nutritional Science, Faculty of Health and Welfare, Seinan Jo Gakuin University, Fukuoka 803-0835, Japan;
| | - Chitoshi Takayama
- Department of Molecular Anatomy, Graduate School of Medicine, University of the Ryukyus, 207 Uehara, Nishihara 903-0215, Japan; (C.S.-O.); (S.K.); (Y.K.); (M.S.)
- Correspondence: ; Tel.: +81-98-895-1103 or +81-895-1405
| |
Collapse
|
22
|
Lim WM, Chin EWM, Tang BL, Chen T, Goh ELK. WNK3 Maintains the GABAergic Inhibitory Tone, Synaptic Excitation and Neuronal Excitability via Regulation of KCC2 Cotransporter in Mature Neurons. Front Mol Neurosci 2021; 14:762142. [PMID: 34858138 PMCID: PMC8631424 DOI: 10.3389/fnmol.2021.762142] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2021] [Accepted: 10/14/2021] [Indexed: 11/13/2022] Open
Abstract
The activation of chloride (Cl−)permeable gamma (γ)-aminobutyric acid type A(GABAA) receptors induces synaptic inhibition in mature and excitation in immature neurons. This developmental “switch” in GABA function controlled by its polarity depends on the postnatal decrease in intraneuronal Cl− concentration mediated by KCC2, a member of cation-chloride cotransporters (CCCs). The serine-threonine kinase WNK3 (With No Lysine [K]), is a potent regulator of all CCCs and is expressed in neurons. Here, we characterized the functions of WNK3 and its role in GABAergic signaling in cultured embryonic day 18 (E18) hippocampal neurons. We observed a decrease in WNK3 expression as neurons mature. Knocking down of WNK3 significantly hyperpolarized EGABA in mature neurons (DIV13–15) but had no effect on immature neurons (DIV6–8). This hyperpolarized EGABA in WNK3-deficient neurons was not due to the total expression of NKCC1 and KCC2, that remained unchanged. However, there was a reduction in phosphorylated KCC2 at the membrane, suggesting an increase in KCC2 chloride export activity. Furthermore, hyperpolarized EGABA observed in WNK3-deficient neurons can be reversed by the KCC2 inhibitor, VU024055, thus indicating that WNK3 acts through KCC2 to influence EGABA. Notably, WNK3 knockdown resulted in morphological changes in mature but not immature neurons. Electrophysiological characterization of WNK3-deficient mature neurons revealed reduced capacitances but increased intrinsic excitability and synaptic excitation. Hence, our study demonstrates that WNK3 maintains the “adult” GABAergic inhibitory tone in neurons and plays a role in the morphological development of neurons and excitability.
Collapse
Affiliation(s)
- Wee Meng Lim
- Neuroscience Academic Clinical Programme, Duke-NUS Medical School, Singapore, Singapore.,NUS Graduate School for Integrative Sciences and Engineering, Singapore, Singapore
| | - Eunice W M Chin
- Neuroscience Academic Clinical Programme, Duke-NUS Medical School, Singapore, Singapore.,NUS Graduate School for Integrative Sciences and Engineering, Singapore, Singapore.,Neuroscience and Mental Health Faculty, Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore, Singapore
| | - Bor Luen Tang
- NUS Graduate School for Integrative Sciences and Engineering, Singapore, Singapore.,Department of Biochemistry, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Tingting Chen
- School of Pharmacy, Nantong University, Nantong, China
| | - Eyleen L K Goh
- Neuroscience Academic Clinical Programme, Duke-NUS Medical School, Singapore, Singapore.,Neuroscience and Mental Health Faculty, Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore, Singapore
| |
Collapse
|
23
|
Portioli C, Ruiz Munevar MJ, De Vivo M, Cancedda L. Cation-coupled chloride cotransporters: chemical insights and disease implications. TRENDS IN CHEMISTRY 2021; 3:832-849. [PMID: 34604727 PMCID: PMC8461084 DOI: 10.1016/j.trechm.2021.05.004] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Cation-coupled chloride cotransporters (CCCs) modulate the transport of sodium and/or potassium cations coupled with chloride anions across the cell membrane. CCCs thus help regulate intracellular ionic concentration and consequent cell volume homeostasis. This has been largely exploited in the past to develop diuretic drugs that act on CCCs expressed in the kidney. However, a growing wealth of evidence has demonstrated that CCCs are also critically involved in a great variety of other pathologies, motivating most recent drug discovery programs targeting CCCs. Here, we examine the structure–function relationship of CCCs. By linking recent high-resolution cryogenic electron microscopy (cryo-EM) data with older biochemical/functional studies on CCCs, we discuss the mechanistic insights and opportunities to design selective CCC modulators to treat diverse pathologies. The structural topology and function of all cation-coupled chloride cotransporters (CCCs) have been continuously investigated over the past 40 years, with great progress also thanks to the recent cryogenic electron microscopy (cryo-EM) resolution of the structures of five CCCs. In particular, such studies have clarified the structure–function relationship for the Na-K-Cl cotransporter NKCC1 and K-Cl cotransporters KCC1–4. The constantly growing evidence of the crucial involvement of CCCs in physiological and various pathological conditions, as well as the evidence of their wide expression in diverse body tissues, has promoted CCCs as targets for the discovery and development of new, safer, and more selective/effective drugs for a plethora of pathologies. Post-translational modification anchor points on the structure of CCCs may offer alternative strategies for small molecule drug discovery.
Collapse
Affiliation(s)
- Corinne Portioli
- Brain Development and Disease Laboratory, Istituto Italiano di Tecnologia (IIT), Via Morego 30, 16163 Genoa, Italy.,Laboratory of Molecular Modeling and Drug Discovery, IIT, Via Morego, 30 16163 Genoa, Italy
| | | | - Marco De Vivo
- Laboratory of Molecular Modeling and Drug Discovery, IIT, Via Morego, 30 16163 Genoa, Italy
| | - Laura Cancedda
- Brain Development and Disease Laboratory, Istituto Italiano di Tecnologia (IIT), Via Morego 30, 16163 Genoa, Italy.,Dulbecco Telethon Institute, Via Varese 16b, 00185 Rome, Italy
| |
Collapse
|
24
|
Ma JJ, Zhang TY, Diao XT, Yao L, Li YX, Suo ZW, Yang X, Hu XD, Liu YN. BDNF modulated KCC2 ubiquitylation in spinal cord dorsal horn of mice. Eur J Pharmacol 2021; 906:174205. [PMID: 34048740 DOI: 10.1016/j.ejphar.2021.174205] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2020] [Revised: 05/21/2021] [Accepted: 05/21/2021] [Indexed: 10/21/2022]
Abstract
The K+-Cl- co-transporter 2 (KCC2) is a neuron-specific Cl- extruder in the dorsal horn of spinal cord. The low intracellular Cl- concentration established by KCC2 is critical for GABAergic and glycinergic systems to generate synaptic inhibition. Peripheral nerve lesions have been shown to cause KCC2 dysfunction in adult spinal cord through brain-derived neurotrophic factor (BDNF) signaling, which switches the hyperpolarizing inhibitory transmission to be depolarizing and excitatory. However, the mechanisms by which BDNF impairs KCC2 function remain to be elucidated. Here we found that BDNF treatment enhanced KCC2 ubiquitination in the dorsal horn of adult mice, a post-translational modification that leads to KCC2 degradation. Our data showed that spinal BDNF application promoted KCC2 interaction with Casitas B-lineage lymphoma b (Cbl-b), one of the E3 ubiquitin ligases that are involved in the spinal processing of nociceptive information. Knockdown of Cbl-b expression decreased KCC2 ubiquitination level and attenuated the pain hypersensitivity induced by BDNF. Spared nerve injury significantly increased KCC2 ubiquitination, which could be reversed by inhibition of TrkB receptor. Our data implicated that KCC2 was one of the important pain-related substrates of Cbl-b and that ubiquitin modification contributed to BDNF-induced KCC2 hypofunction in the spinal cord.
Collapse
Affiliation(s)
- Juan-Juan Ma
- Department of Molecular Pharmacology, School of Pharmacy, Lanzhou University, Lanzhou, Gansu, 730000, PR China
| | - Tian-Yu Zhang
- Department of Molecular Pharmacology, School of Pharmacy, Lanzhou University, Lanzhou, Gansu, 730000, PR China
| | - Xin-Tong Diao
- Department of Molecular Pharmacology, School of Pharmacy, Lanzhou University, Lanzhou, Gansu, 730000, PR China
| | - Lin Yao
- Department of Molecular Pharmacology, School of Pharmacy, Lanzhou University, Lanzhou, Gansu, 730000, PR China
| | - Yin-Xia Li
- Department of Molecular Pharmacology, School of Pharmacy, Lanzhou University, Lanzhou, Gansu, 730000, PR China
| | - Zhan-Wei Suo
- Department of Molecular Pharmacology, School of Pharmacy, Lanzhou University, Lanzhou, Gansu, 730000, PR China
| | - Xian Yang
- Department of Molecular Pharmacology, School of Pharmacy, Lanzhou University, Lanzhou, Gansu, 730000, PR China
| | - Xiao-Dong Hu
- Department of Molecular Pharmacology, School of Pharmacy, Lanzhou University, Lanzhou, Gansu, 730000, PR China.
| | - Yan-Ni Liu
- Department of Molecular Pharmacology, School of Pharmacy, Lanzhou University, Lanzhou, Gansu, 730000, PR China.
| |
Collapse
|
25
|
Rahmati N, Normoyle KP, Glykys J, Dzhala VI, Lillis KP, Kahle KT, Raiyyani R, Jacob T, Staley KJ. Unique Actions of GABA Arising from Cytoplasmic Chloride Microdomains. J Neurosci 2021; 41:4957-4975. [PMID: 33903223 PMCID: PMC8197632 DOI: 10.1523/jneurosci.3175-20.2021] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2020] [Revised: 03/10/2021] [Accepted: 04/10/2021] [Indexed: 12/21/2022] Open
Abstract
Developmental, cellular, and subcellular variations in the direction of neuronal Cl- currents elicited by GABAA receptor activation have been frequently reported. We found a corresponding variance in the GABAA receptor reversal potential (EGABA) for synapses originating from individual interneurons onto a single pyramidal cell. These findings suggest a similar heterogeneity in the cytoplasmic intracellular concentration of chloride ([Cl-]i) in individual dendrites. We determined [Cl-]i in the murine hippocampus and cerebral cortex of both sexes by (1) two-photon imaging of the Cl--sensitive, ratiometric fluorescent protein SuperClomeleon; (2) Fluorescence Lifetime IMaging (FLIM) of the Cl--sensitive fluorophore MEQ (6-methoxy-N-ethylquinolinium); and (3) electrophysiological measurements of EGABA by pressure application of GABA and RuBi-GABA uncaging. Fluorometric and electrophysiological estimates of local [Cl-]i were highly correlated. [Cl-]i microdomains persisted after pharmacological inhibition of cation-chloride cotransporters, but were progressively modified after inhibiting the polymerization of the anionic biopolymer actin. These methods collectively demonstrated stable [Cl-]i microdomains in individual neurons in vitro and in vivo and the role of immobile anions in its stability. Our results highlight the existence of functionally significant neuronal Cl- microdomains that modify the impact of GABAergic inputs.SIGNIFICANCE STATEMENT Microdomains of varying chloride concentrations in the neuronal cytoplasm are a predictable consequence of the inhomogeneous distribution of anionic polymers such as actin, tubulin, and nucleic acids. Here, we demonstrate the existence and stability of these microdomains, as well as the consequence for GABAergic synaptic signaling: each interneuron produces a postsynaptic GABAA response with a unique reversal potential. In individual hippocampal pyramidal cells, the range of GABAA reversal potentials evoked by stimulating different interneurons was >20 mV. Some interneurons generated postsynaptic responses in pyramidal cells that reversed at potentials beyond what would be considered purely inhibitory. Cytoplasmic chloride microdomains enable each pyramidal cell to maintain a compendium of unique postsynaptic responses to the activity of individual interneurons.
Collapse
Affiliation(s)
- Negah Rahmati
- Department of Neurology, Harvard Medical School and Massachusetts General Hospital, Boston, Massachusetts 02114
| | - Kieran P Normoyle
- Department of Neurology, Harvard Medical School and Massachusetts General Hospital, Boston, Massachusetts 02114
| | - Joseph Glykys
- Department of Pediatrics and Neurology, Iowa Neuroscience Institute, Carver College of Medicine, University of Iowa, Iowa City, Iowa 52242
| | - Volodymyr I Dzhala
- Department of Neurology, Harvard Medical School and Massachusetts General Hospital, Boston, Massachusetts 02114
| | - Kyle P Lillis
- Department of Neurology, Harvard Medical School and Massachusetts General Hospital, Boston, Massachusetts 02114
| | - Kristopher T Kahle
- Departments of Neurosurgery, Pediatrics, and Cellular & Molecular Physiology, Yale School of Medicine, New Haven, Connecticut 06510
| | - Rehan Raiyyani
- Department of Neurology, Harvard Medical School and Massachusetts General Hospital, Boston, Massachusetts 02114
| | - Theju Jacob
- Department of Neurology, Harvard Medical School and Massachusetts General Hospital, Boston, Massachusetts 02114
| | - Kevin J Staley
- Department of Neurology, Harvard Medical School and Massachusetts General Hospital, Boston, Massachusetts 02114
| |
Collapse
|
26
|
Bilchak JN, Yeakle K, Caron G, Malloy D, Côté MP. Enhancing KCC2 activity decreases hyperreflexia and spasticity after chronic spinal cord injury. Exp Neurol 2021; 338:113605. [PMID: 33453210 PMCID: PMC7904648 DOI: 10.1016/j.expneurol.2021.113605] [Citation(s) in RCA: 33] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2020] [Revised: 12/21/2020] [Accepted: 01/09/2021] [Indexed: 02/03/2023]
Abstract
After spinal cord injury (SCI), the majority of individuals develop spasticity, a debilitating condition involving involuntary movements, co-contraction of antagonistic muscles, and hyperreflexia. By acting on GABAergic and Ca2+-dependent signaling, current anti-spastic medications lead to serious side effects, including a drastic decrease in motoneuronal excitability which impairs motor function and rehabilitation efforts. Exercise, in contrast, decreases spastic symptoms without decreasing motoneuron excitability. These functional improvements coincide with an increase in expression of the chloride co-transporter KCC2 in lumbar motoneurons. Thus, we hypothesized that spastic symptoms can be alleviated directly through restoration of chloride homeostasis and endogenous inhibition by increasing KCC2 activity. Here, we used the recently developed KCC2 enhancer, CLP257, to evaluate the effects of acutely increasing KCC2 extrusion capability on spastic symptoms after chronic SCI. Sprague Dawley rats received a spinal cord transection at T12 and were either bike-trained or remained sedentary for 5 weeks. Increasing KCC2 activity in the lumbar enlargement improved the rate-dependent depression of the H-reflex and reduced both phasic and tonic EMG responses to muscle stretch in sedentary animals after chronic SCI. Furthermore, the improvements due to this pharmacological treatment mirror those of exercise. Together, our results suggest that pharmacologically increasing KCC2 activity is a promising approach to decrease spastic symptoms in individuals with SCI. By acting to directly restore endogenous inhibition, this strategy has potential to avoid severe side effects and improve the quality of life of affected individuals.
Collapse
Affiliation(s)
- Jadwiga N Bilchak
- Marion Murray Spinal Cord Injury Research Center, Department of Neurobiology and Anatomy, Drexel University College of Medicine, Philadelphia, PA 19129, United States of America
| | - Kyle Yeakle
- Marion Murray Spinal Cord Injury Research Center, Department of Neurobiology and Anatomy, Drexel University College of Medicine, Philadelphia, PA 19129, United States of America
| | - Guillaume Caron
- Marion Murray Spinal Cord Injury Research Center, Department of Neurobiology and Anatomy, Drexel University College of Medicine, Philadelphia, PA 19129, United States of America
| | - Dillon Malloy
- Marion Murray Spinal Cord Injury Research Center, Department of Neurobiology and Anatomy, Drexel University College of Medicine, Philadelphia, PA 19129, United States of America
| | - Marie-Pascale Côté
- Marion Murray Spinal Cord Injury Research Center, Department of Neurobiology and Anatomy, Drexel University College of Medicine, Philadelphia, PA 19129, United States of America.
| |
Collapse
|
27
|
Basu T, Maguire J, Salpekar JA. Hypothalamic-pituitary-adrenal axis targets for the treatment of epilepsy. Neurosci Lett 2021; 746:135618. [PMID: 33429002 DOI: 10.1016/j.neulet.2020.135618] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2020] [Revised: 12/25/2020] [Accepted: 12/29/2020] [Indexed: 12/20/2022]
Abstract
Stress is a common seizure trigger in persons with epilepsy. The body's physiological response to stress is mediated by the hypothalamic-pituitary-adrenal (HPA) axis and involves a hormonal cascade that includes corticotropin releasing hormone (CRH), adrenocorticotropin releasing hormone (ACTH) and the release of cortisol (in humans and primates) or corticosterone (in rodents). The prolonged exposure to stress hormones may not only exacerbate pre-existing medical conditions including epilepsy, but may also increase the predisposition to psychiatric comorbidities. Hyperactivity of the HPA axis negatively impacts the structure and function of the temporal lobe of the brain, a region that is heavily involved in epilepsy and mood disorders like anxiety and depression. Seizures themselves damage temporal lobe structures, further disinhibiting the HPA axis, setting off a vicious cycle of neuronal damage and increasing susceptibility for subsequent seizures and psychiatric comorbidity. Treatments targeting the HPA axis may be beneficial both for epilepsy and for associated stress-related comorbidities such as anxiety or depression. This paper will highlight the evidence demonstrating dysfunction in the HPA axis associated with epilepsy which may contribute to the comorbidity of psychiatric disorders and epilepsy, and propose treatment strategies that may dually improve seizure control as well as alleviate stress related psychiatric comorbidities.
Collapse
Affiliation(s)
- Trina Basu
- Department of Neuroscience, Tufts University School of Medicine, Boston, MA 02111, United States
| | - Jamie Maguire
- Department of Neuroscience, Tufts University School of Medicine, Boston, MA 02111, United States
| | - Jay A Salpekar
- Kennedy Krieger Institute, Johns Hopkins University Medical School, Baltimore, MD 21205, United States.
| |
Collapse
|
28
|
Xie Y, Chang S, Zhao C, Wang F, Liu S, Wang J, Delpire E, Ye S, Guo J. Structures and an activation mechanism of human potassium-chloride cotransporters. SCIENCE ADVANCES 2020; 6:eabc5883. [PMID: 33310850 PMCID: PMC7732191 DOI: 10.1126/sciadv.abc5883] [Citation(s) in RCA: 39] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/02/2020] [Accepted: 10/21/2020] [Indexed: 05/07/2023]
Abstract
Potassium-chloride cotransporters KCC1 to KCC4 mediate the coupled export of potassium and chloride across the plasma membrane and play important roles in cell volume regulation, auditory system function, and γ-aminobutyric acid (GABA) and glycine-mediated inhibitory neurotransmission. Here, we present 2.9- to 3.6-Å resolution structures of full-length human KCC2, KCC3, and KCC4. All three KCCs adopt a similar overall architecture, a domain-swap dimeric assembly, and an inward-facing conformation. The structural and functional studies reveal that one unexpected N-terminal peptide binds at the cytosolic facing cavity and locks KCC2 and KCC4 at an autoinhibition state. The C-terminal domain (CTD) directly interacts with the N-terminal inhibitory peptide, and the relative motions between the CTD and the transmembrane domain (TMD) suggest that CTD regulates KCCs' activities by adjusting the autoinhibitory effect. These structures provide the first glimpse of full-length structures of KCCs and an autoinhibition mechanism among the amino acid-polyamine-organocation transporter superfamily.
Collapse
Affiliation(s)
- Yuan Xie
- Department of Biophysics, and Department of Pathology of Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou 310058, China
| | - Shenghai Chang
- Department of Biophysics, and Department of Pathology of Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou 310058, China
- Center of Cryo Electron Microscopy, Zhejiang University School of Medicine, Hangzhou 310058, China
| | - Cheng Zhao
- Department of Biophysics, and Department of Pathology of Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou 310058, China
| | - Feng Wang
- Wuxi Biortus Biosciences Co. Ltd., 6 Dongsheng West Road, Jiangyin, 214437, China
| | - Si Liu
- Tianjin Key Laboratory of Function and Application of Biological Macromolecular Structures, School of Life Sciences, Tianjin University, 92 Weijin Road, Nankai District, Tianjin 300072, China
| | - Jin Wang
- Department of Pathology of Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou 310058, China
| | - Eric Delpire
- Department of Anesthesiology, Vanderbilt University School of Medicine, Nashville, TN 37232, USA.
| | - Sheng Ye
- Tianjin Key Laboratory of Function and Application of Biological Macromolecular Structures, School of Life Sciences, Tianjin University, 92 Weijin Road, Nankai District, Tianjin 300072, China.
- Life Sciences Institute and Innovation Center for Cell Signaling Network, Zhejiang University, Hangzhou, Zhejiang 310058, China
| | - Jiangtao Guo
- Department of Biophysics, and Department of Pathology of Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou 310058, China.
- Department of Cardiology, Key Laboratory of Cardiovascular Intervention and Regenerative Medicine of Zhejiang Province, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, 310016, China
| |
Collapse
|
29
|
Murillo-de-Ozores AR, Chávez-Canales M, de los Heros P, Gamba G, Castañeda-Bueno M. Physiological Processes Modulated by the Chloride-Sensitive WNK-SPAK/OSR1 Kinase Signaling Pathway and the Cation-Coupled Chloride Cotransporters. Front Physiol 2020; 11:585907. [PMID: 33192599 PMCID: PMC7606576 DOI: 10.3389/fphys.2020.585907] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2020] [Accepted: 09/29/2020] [Indexed: 12/15/2022] Open
Abstract
The role of Cl- as an intracellular signaling ion has been increasingly recognized in recent years. One of the currently best described roles of Cl- in signaling is the modulation of the With-No-Lysine (K) (WNK) - STE20-Proline Alanine rich Kinase (SPAK)/Oxidative Stress Responsive Kinase 1 (OSR1) - Cation-Coupled Cl- Cotransporters (CCCs) cascade. Binding of a Cl- anion to the active site of WNK kinases directly modulates their activity, promoting their inhibition. WNK activation due to Cl- release from the binding site leads to phosphorylation and activation of SPAK/OSR1, which in turn phosphorylate the CCCs. Phosphorylation by WNKs-SPAK/OSR1 of the Na+-driven CCCs (mediating ions influx) promote their activation, whereas that of the K+-driven CCCs (mediating ions efflux) promote their inhibition. This results in net Cl- influx and feedback inhibition of WNK kinases. A wide variety of alterations to this pathway have been recognized as the cause of several human diseases, with manifestations in different systems. The understanding of WNK kinases as Cl- sensitive proteins has allowed us to better understand the mechanistic details of regulatory processes involved in diverse physiological phenomena that are reviewed here. These include cell volume regulation, potassium sensing and intracellular signaling in the renal distal convoluted tubule, and regulation of the neuronal response to the neurotransmitter GABA.
Collapse
Affiliation(s)
- Adrián Rafael Murillo-de-Ozores
- Department of Nephrology and Mineral Metabolism, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Mexico City, Mexico
- Facultad de Medicina, Universidad Nacional Autónoma de México, Mexico City, Mexico
| | - María Chávez-Canales
- Unidad de Investigación UNAM-INC, Instituto Nacional de Cardiología Ignacio Chávez and Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México, Mexico City, Mexico
| | - Paola de los Heros
- Unidad de Investigación UNAM-INC, Research Division, Facultad de Medicina, Universidad Nacional Autónoma de México, Mexico City, Mexico
| | - Gerardo Gamba
- Department of Nephrology and Mineral Metabolism, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Mexico City, Mexico
- Molecular Physiology Unit, Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México, Mexico City, Mexico
| | - María Castañeda-Bueno
- Department of Nephrology and Mineral Metabolism, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Mexico City, Mexico
| |
Collapse
|
30
|
Zimanyi CM, Guo M, Mahmood A, Hendrickson WA, Hirsh D, Cheung J. Structure of the Regulatory Cytosolic Domain of a Eukaryotic Potassium-Chloride Cotransporter. Structure 2020; 28:1051-1060.e4. [PMID: 32679039 PMCID: PMC8408865 DOI: 10.1016/j.str.2020.06.009] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2020] [Revised: 05/18/2020] [Accepted: 06/25/2020] [Indexed: 01/01/2023]
Abstract
Cation-chloride cotransporters (CCCs) regulate the movement of chloride across membranes, controlling physiological processes from cell volume maintenance to neuronal signaling. Human CCCs are clinical targets for existing diuretics and potentially additional indications. Here, we report the X-ray crystal structure of the soluble C-terminal regulatory domain of a eukaryotic potassium-chloride cotransporter, Caenorhabditis elegans KCC-1. We observe a core α/β fold conserved among CCCs. Using structure-based sequence alignment, we analyze similarities and differences to the C-terminal domains of other CCC family members. We find that important regulatory motifs are in less-structured regions and residues important for dimerization are not widely conserved, suggesting that oligomerization and its effects may vary within the larger family. This snapshot of a eukaryotic KCC is a valuable starting point for the rational design of studies of cellular chloride regulation.
Collapse
Affiliation(s)
- Christina M Zimanyi
- New York Structural Biology Center, 89 Convent Avenue, New York, NY 10027, USA.
| | - Mo Guo
- New York Structural Biology Center, 89 Convent Avenue, New York, NY 10027, USA
| | - Arshad Mahmood
- New York Structural Biology Center, 89 Convent Avenue, New York, NY 10027, USA
| | - Wayne A Hendrickson
- New York Structural Biology Center, 89 Convent Avenue, New York, NY 10027, USA; Department of Biochemistry and Molecular Biophysics, Columbia University, New York, NY 10032, USA
| | - David Hirsh
- New York Structural Biology Center, 89 Convent Avenue, New York, NY 10027, USA; Department of Biochemistry and Molecular Biophysics, Columbia University, New York, NY 10032, USA
| | - Jonah Cheung
- New York Structural Biology Center, 89 Convent Avenue, New York, NY 10027, USA
| |
Collapse
|
31
|
Fukuda A. Chloride homeodynamics underlying modal shifts in cellular and network oscillations. Neurosci Res 2020; 156:14-23. [PMID: 32105770 DOI: 10.1016/j.neures.2020.02.010] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/25/2019] [Revised: 11/22/2019] [Accepted: 02/21/2020] [Indexed: 11/16/2022]
Abstract
γ-Aminobutyric acid (GABA) generally induces hyperpolarization and inhibition in the adult brain, but causes depolarization (and can be excitatory) in the immature brain. Depolarizing GABA actions are necessary for neurogenesis, differentiation, migration, and synaptogenesis. Additionally, the conversion of GABA responses from inhibition to excitation can be induced in adults by pathological conditions. Because GABAA receptors are Cl- channels, alternating GABA actions between hyperpolarization (Cl- influx) and depolarization (Cl- efflux) are induced by changes in the Cl- gradient, which is regulated by C- transporters. Thus, the dynamics of neural functions are modulated by active Cl- homeostasis (Cl- homeodynamics), alternating inhibition and excitation, and could underlie the modal shifts in cellular and network oscillations. Such a modal shift in GABA actions is required for normal development. Thus disturbances in this developmental GABA modal shift and/or the induction of excitatory GABA action in adult could underlie the pathogenesis of diverse neurological diseases (so-called network diseases).
Collapse
Affiliation(s)
- Atsuo Fukuda
- Department of Neurophysiology, Hamamatsu University School of Medicine, Hamamatsu, Japan; Advanced Research Facilities and Services, Preeminent Medical Photonics Education and Research Center, Hamamatsu University School of Medicine, Hamamatsu, Japan.
| |
Collapse
|
32
|
Venugopalan P, Cameron EG, Zhang X, Nahmou M, Muller KJ, Goldberg JL. Physiologic maturation is both extrinsically and intrinsically regulated in progenitor-derived neurons. Sci Rep 2020; 10:2337. [PMID: 32047174 PMCID: PMC7012889 DOI: 10.1038/s41598-020-58120-5] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2016] [Accepted: 01/03/2020] [Indexed: 12/15/2022] Open
Abstract
During development, newly-differentiated neurons undergo several morphological and physiological changes to become functional, mature neurons. Physiologic maturation of neuronal cells derived from isolated stem or progenitor cells may provide insight into maturation in vivo but is not well studied. As a step towards understanding how neuronal maturation is regulated, we studied the developmental switch of response to the neurotransmitter GABA, from excitatory depolarization to inhibitory hyperpolarization. We compared acutely isolated retinal ganglion cells (RGCs) at various developmental stages and RGCs differentiated in vitro from embryonic retinal progenitors for the effects of aging and, independently, of retinal environment age on their GABAA receptor (GABAAR) responses, elicited by muscimol. We found that neurons generated in vitro from progenitors exhibited depolarizing, immature GABA responses, like those of early postnatal RGCs. As progenitor-derived neurons aged from 1 to 3 weeks, their GABA responses matured. Interestingly, signals secreted by the early postnatal retina suppressed acquisition of mature GABA responses. This suppression was not associated with changes in expression of GABAAR or of the chloride co-transporter KCC2, but rather with inhibition of KCC2 dimerization in differentiating neurons. Taken together, these data indicate GABA response maturation depends on release of inhibition by developmentally regulated diffusible signals from the retina.
Collapse
Affiliation(s)
- Praseeda Venugopalan
- Neuroscience Program, University of Miami, Miami, FL, 33136, USA
- Shiley Eye Institute, University of California, San Diego, CA, 92093, USA
| | - Evan G Cameron
- Shiley Eye Institute, University of California, San Diego, CA, 92093, USA
- Byers Eye Institute, Stanford University, Stanford, CA, 94303, USA
| | - Xiong Zhang
- Shiley Eye Institute, University of California, San Diego, CA, 92093, USA
| | - Michael Nahmou
- Byers Eye Institute, Stanford University, Stanford, CA, 94303, USA
| | - Kenneth J Muller
- Neuroscience Program, University of Miami, Miami, FL, 33136, USA.
- Department of Physiology & Biophysics, University of Miami Miller School of Medicine, Miami, FL, 33136, USA.
| | - Jeffrey L Goldberg
- Neuroscience Program, University of Miami, Miami, FL, 33136, USA.
- Shiley Eye Institute, University of California, San Diego, CA, 92093, USA.
- Byers Eye Institute, Stanford University, Stanford, CA, 94303, USA.
| |
Collapse
|
33
|
Liu R, Wang J, Liang S, Zhang G, Yang X. Role of NKCC1 and KCC2 in Epilepsy: From Expression to Function. Front Neurol 2020; 10:1407. [PMID: 32010056 PMCID: PMC6978738 DOI: 10.3389/fneur.2019.01407] [Citation(s) in RCA: 66] [Impact Index Per Article: 13.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2019] [Accepted: 12/23/2019] [Indexed: 01/21/2023] Open
Abstract
As a main inhibitory neurotransmitter in the central nervous system, γ-aminobutyric acid (GABA) activates chloride-permeable GABAa receptors (GABAa Rs) and induces chloride ion (Cl−) flow, which relies on the intracellular chloride concentration ([Cl−]i) of the postsynaptic neuron. The Na-K-2Cl cotransporter isoform 1 (NKCC1) and the K-Cl cotransporter isoform 2 (KCC2) are two main cation-chloride cotransporters (CCCs) that have been implicated in human epilepsy. NKCC1 and KCC2 reset [Cl−]i by accumulating and extruding Cl−, respectively. Previous studies have shown that the profile of NKCC1 and KCC2 in neonatal neurons may reappear in mature neurons under some pathophysiological conditions, such as epilepsy. Although increasing studies focusing on the expression of NKCC1 and KCC2 have suggested that impaired chloride plasticity may be closely related to epilepsy, additional neuroelectrophysiological research aimed at studying the functions of NKCC1 and KCC2 are needed to understand the exact mechanism by which they induce epileptogenesis. In this review, we aim to briefly summarize the current researches surrounding the expression and function of NKCC1 and KCC2 in epileptogenesis and its implications on the treatment of epilepsy. We will also explore the potential for NKCC1 and KCC2 to be therapeutic targets for the development of novel antiepileptic drugs.
Collapse
Affiliation(s)
- Ru Liu
- Neuroelectrophysiological Laboratory, Xuanwu Hospital, Capital Medical University, Beijing, China.,Center of Epilepsy, Center for Brain Disorders Research, Capital Medical University, Beijing, China.,Center of Epilepsy, Beijing Institute of Brain Disorders, Beijing, China.,Guangzhou Regenerative Medicine and Health Guangdong Laboratory, Guangzhou, China
| | - Junling Wang
- Neuroelectrophysiological Laboratory, Xuanwu Hospital, Capital Medical University, Beijing, China.,Center of Epilepsy, Center for Brain Disorders Research, Capital Medical University, Beijing, China.,Center of Epilepsy, Beijing Institute of Brain Disorders, Beijing, China.,Guangzhou Regenerative Medicine and Health Guangdong Laboratory, Guangzhou, China
| | - Shuli Liang
- Department of Functional Neurosurgery, Beijing Children's Hospital, Capital Medical University, Beijing, China
| | - Guojun Zhang
- Department of Functional Neurosurgery, Xuanwu Hospital, Capital Medical University, Beijing, China
| | - Xiaofeng Yang
- Neuroelectrophysiological Laboratory, Xuanwu Hospital, Capital Medical University, Beijing, China.,Center of Epilepsy, Center for Brain Disorders Research, Capital Medical University, Beijing, China.,Center of Epilepsy, Beijing Institute of Brain Disorders, Beijing, China.,Guangzhou Regenerative Medicine and Health Guangdong Laboratory, Guangzhou, China
| |
Collapse
|
34
|
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]
|
35
|
Watanabe M, Zhang J, Mansuri MS, Duan J, Karimy JK, Delpire E, Alper SL, Lifton RP, Fukuda A, Kahle KT. Developmentally regulated KCC2 phosphorylation is essential for dynamic GABA-mediated inhibition and survival. Sci Signal 2019; 12:eaaw9315. [PMID: 31615901 DOI: 10.1126/scisignal.aaw9315.(#,] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/23/2024]
Abstract
Despite its importance for γ-aminobutyric acid (GABA) inhibition and involvement in neurodevelopmental disease, the regulatory mechanisms of the K+/Cl- cotransporter KCC2 (encoded by SLC12A5) during maturation of the central nervous system (CNS) are not entirely understood. Here, we applied quantitative phosphoproteomics to systematically map sites of KCC2 phosphorylation during CNS development in the mouse. KCC2 phosphorylation at Thr906 and Thr1007, which inhibits KCC2 activity, underwent dephosphorylation in parallel with the GABA excitatory-inhibitory sequence in vivo. Knockin mice expressing the homozygous phosphomimetic KCC2 mutations T906E/T1007E (Kcc2E/E ), which prevented the normal developmentally regulated dephosphorylation of these sites, exhibited early postnatal death from respiratory arrest and a marked absence of cervical spinal neuron respiratory discharges. Kcc2E/E mice also displayed disrupted lumbar spinal neuron locomotor rhythmogenesis and touch-evoked status epilepticus associated with markedly impaired KCC2-dependent Cl- extrusion. These data identify a previously unknown phosphorylation-dependent KCC2 regulatory mechanism during CNS development that is essential for dynamic GABA-mediated inhibition and survival.
Collapse
Affiliation(s)
- Miho Watanabe
- Department of Neurophysiology, Hamamatsu University School of Medicine, Hamamatsu, Shizuoka 431-3192, Japan
| | - Jinwei Zhang
- Institute of Biomedical and Clinical Sciences, Medical School, College of Medicine and Health, University of Exeter, Hatherly Laboratories, Exeter EX4 4PS, UK
| | - M Shahid Mansuri
- Department of Neurosurgery, Yale School of Medicine, New Haven, CT 06510, USA
| | - Jingjing Duan
- Human Aging Research Institute, School of Life Sciences, Nanchang University, Nanchang, Jiangxi 330031, China
| | - Jason K Karimy
- Department of Neurosurgery, Yale School of Medicine, New Haven, CT 06510, USA
| | - Eric Delpire
- Department of Anesthesiology, Vanderbilt University School of Medicine, Nashville, TN 37232, USA
| | - Seth L Alper
- Division of Nephrology, Department of Medicine, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, MA 02215, USA
- The Broad Institute of Harvard and the Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Richard P Lifton
- Laboratory of Human Genetics and Genomics, The Rockefeller University, New York, NY, USA
- Department of Genetics, Yale University School of Medicine, New Haven, CT 06511, USA
| | - Atsuo Fukuda
- Department of Neurophysiology, Hamamatsu University School of Medicine, Hamamatsu, Shizuoka 431-3192, Japan.
- Advanced Research Facilities and Services, Preeminent Medical Photonics Education and Research Center, Hamamatsu University School of Medicine, Hamamatsu, Shizuoka 431-3192, Japan
| | - Kristopher T Kahle
- Departments of Neurosurgery, Pediatrics, and Cellular and Molecular Physiology, Centers for Mendelian Genomics, Yale School of Medicine, New Haven, CT 06510, USA.
| |
Collapse
|
36
|
Watanabe M, Zhang J, Mansuri MS, Duan J, Karimy JK, Delpire E, Alper SL, Lifton RP, Fukuda A, Kahle KT. Developmentally regulated KCC2 phosphorylation is essential for dynamic GABA-mediated inhibition and survival. Sci Signal 2019; 12:eaaw9315. [PMID: 31615901 PMCID: PMC7219477 DOI: 10.1126/scisignal.aaw9315] [Citation(s) in RCA: 48] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
Despite its importance for γ-aminobutyric acid (GABA) inhibition and involvement in neurodevelopmental disease, the regulatory mechanisms of the K+/Cl- cotransporter KCC2 (encoded by SLC12A5) during maturation of the central nervous system (CNS) are not entirely understood. Here, we applied quantitative phosphoproteomics to systematically map sites of KCC2 phosphorylation during CNS development in the mouse. KCC2 phosphorylation at Thr906 and Thr1007, which inhibits KCC2 activity, underwent dephosphorylation in parallel with the GABA excitatory-inhibitory sequence in vivo. Knockin mice expressing the homozygous phosphomimetic KCC2 mutations T906E/T1007E (Kcc2E/E ), which prevented the normal developmentally regulated dephosphorylation of these sites, exhibited early postnatal death from respiratory arrest and a marked absence of cervical spinal neuron respiratory discharges. Kcc2E/E mice also displayed disrupted lumbar spinal neuron locomotor rhythmogenesis and touch-evoked status epilepticus associated with markedly impaired KCC2-dependent Cl- extrusion. These data identify a previously unknown phosphorylation-dependent KCC2 regulatory mechanism during CNS development that is essential for dynamic GABA-mediated inhibition and survival.
Collapse
Affiliation(s)
- Miho Watanabe
- Department of Neurophysiology, Hamamatsu University School of Medicine, Hamamatsu, Shizuoka 431-3192, Japan
| | - Jinwei Zhang
- Institute of Biomedical and Clinical Sciences, Medical School, College of Medicine and Health, University of Exeter, Hatherly Laboratories, Exeter EX4 4PS, UK
| | - M Shahid Mansuri
- Department of Neurosurgery, Yale School of Medicine, New Haven, CT 06510, USA
| | - Jingjing Duan
- Human Aging Research Institute, School of Life Sciences, Nanchang University, Nanchang, Jiangxi 330031, China
| | - Jason K Karimy
- Department of Neurosurgery, Yale School of Medicine, New Haven, CT 06510, USA
| | - Eric Delpire
- Department of Anesthesiology, Vanderbilt University School of Medicine, Nashville, TN 37232, USA
| | - Seth L Alper
- Division of Nephrology, Department of Medicine, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, MA 02215, USA
- The Broad Institute of Harvard and the Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Richard P Lifton
- Laboratory of Human Genetics and Genomics, The Rockefeller University, New York, NY, USA
- Department of Genetics, Yale University School of Medicine, New Haven, CT 06511, USA
| | - Atsuo Fukuda
- Department of Neurophysiology, Hamamatsu University School of Medicine, Hamamatsu, Shizuoka 431-3192, Japan.
- Advanced Research Facilities and Services, Preeminent Medical Photonics Education and Research Center, Hamamatsu University School of Medicine, Hamamatsu, Shizuoka 431-3192, Japan
| | - Kristopher T Kahle
- Departments of Neurosurgery, Pediatrics, and Cellular and Molecular Physiology, Centers for Mendelian Genomics, Yale School of Medicine, New Haven, CT 06510, USA.
| |
Collapse
|
37
|
Pisella LI, Gaiarsa JL, Diabira D, Zhang J, Khalilov I, Duan J, Kahle KT, Medina I. Impaired regulation of KCC2 phosphorylation leads to neuronal network dysfunction and neurodevelopmental pathology. Sci Signal 2019; 12:eaay0300. [PMID: 31615899 PMCID: PMC7192243 DOI: 10.1126/scisignal.aay0300] [Citation(s) in RCA: 38] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
KCC2 is a vital neuronal K+/Cl- cotransporter that is implicated in the etiology of numerous neurological diseases. In normal cells, KCC2 undergoes developmental dephosphorylation at Thr906 and Thr1007 We engineered mice with heterozygous phosphomimetic mutations T906E and T1007E (KCC2E/+ ) to prevent the normal developmental dephosphorylation of these sites. Immature (postnatal day 15) but not juvenile (postnatal day 30) KCC2E/+ mice exhibited altered GABAergic inhibition, an increased glutamate/GABA synaptic ratio, and greater susceptibility to seizure. KCC2E/+ mice also had abnormal ultrasonic vocalizations at postnatal days 10 to 12 and impaired social behavior at postnatal day 60. Postnatal bumetanide treatment restored network activity by postnatal day 15 but failed to restore social behavior by postnatal day 60. Our data indicate that posttranslational KCC2 regulation controls the GABAergic developmental sequence in vivo, indicating that deregulation of KCC2 could be a risk factor for the emergence of neurological pathology.
Collapse
Affiliation(s)
- Lucie I Pisella
- Aix-Marseille University, UMR 1249, INSERM (Institut National de la Santé et de la Recherche Médicale) Unité 1249, INMED (Institut de Neurobiologie de la Méditerranée), Marseille, France
| | - Jean-Luc Gaiarsa
- Aix-Marseille University, UMR 1249, INSERM (Institut National de la Santé et de la Recherche Médicale) Unité 1249, INMED (Institut de Neurobiologie de la Méditerranée), Marseille, France
| | - Diabé Diabira
- Aix-Marseille University, UMR 1249, INSERM (Institut National de la Santé et de la Recherche Médicale) Unité 1249, INMED (Institut de Neurobiologie de la Méditerranée), Marseille, France
| | - Jinwei Zhang
- Institute of Biomedical and Clinical Sciences, College of Medicine and Health, University of Exeter Medical School, Hatherly Laboratories, Exeter EX4 4PS, UK
| | - Ilgam Khalilov
- Aix-Marseille University, UMR 1249, INSERM (Institut National de la Santé et de la Recherche Médicale) Unité 1249, INMED (Institut de Neurobiologie de la Méditerranée), Marseille, France
- Laboratory of Neurobiology, Kazan Federal University, Kazan 420008, Russia
| | - JingJing Duan
- Department of Neurobiology, Howard Hughes Medical Institute, Boston Children's Hospital, Boston, MA 02115, USA
- Departments of Neurosurgery, Pediatrics, and Cellular and Molecular Physiology and Centers for Mendelian Genomics, Yale School of Medicine, New Haven, CT 06510, USA
| | - Kristopher T Kahle
- Departments of Neurosurgery, Pediatrics, and Cellular and Molecular Physiology and Centers for Mendelian Genomics, Yale School of Medicine, New Haven, CT 06510, USA.
| | - Igor Medina
- Aix-Marseille University, UMR 1249, INSERM (Institut National de la Santé et de la Recherche Médicale) Unité 1249, INMED (Institut de Neurobiologie de la Méditerranée), Marseille, France.
| |
Collapse
|
38
|
Goutierre M, Al Awabdh S, Donneger F, François E, Gomez-Dominguez D, Irinopoulou T, Menendez de la Prida L, Poncer JC. KCC2 Regulates Neuronal Excitability and Hippocampal Activity via Interaction with Task-3 Channels. Cell Rep 2019; 28:91-103.e7. [PMID: 31269453 DOI: 10.1016/j.celrep.2019.06.001] [Citation(s) in RCA: 40] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2018] [Revised: 04/18/2019] [Accepted: 05/30/2019] [Indexed: 10/26/2022] Open
Abstract
KCC2 regulates neuronal transmembrane chloride gradients and thereby controls GABA signaling in the brain. KCC2 downregulation is observed in numerous neurological and psychiatric disorders. Paradoxical, excitatory GABA signaling is usually assumed to contribute to abnormal network activity underlying the pathology. We tested this hypothesis and explored the functional impact of chronic KCC2 downregulation in the rat dentate gyrus. Although the reversal potential of GABAA receptor currents is depolarized in KCC2 knockdown neurons, this shift is compensated by depolarization of the resting membrane potential. This reflects downregulation of leak potassium currents. We show KCC2 interacts with Task-3 (KCNK9) channels and is required for their membrane expression. Increased neuronal excitability upon KCC2 suppression altered dentate gyrus rhythmogenesis, which could be normalized by chemogenetic hyperpolarization. Our data reveal KCC2 downregulation engages complex synaptic and cellular alterations beyond GABA signaling that perturb network activity thus offering additional targets for therapeutic intervention.
Collapse
Affiliation(s)
- Marie Goutierre
- INSERM UMR-S 1270, 75005 Paris, France; Sorbonne Université, 75005 Paris, France; Institut du Fer à Moulin, 75005 Paris, France
| | - Sana Al Awabdh
- INSERM UMR-S 1270, 75005 Paris, France; Sorbonne Université, 75005 Paris, France; Institut du Fer à Moulin, 75005 Paris, France
| | - Florian Donneger
- INSERM UMR-S 1270, 75005 Paris, France; Sorbonne Université, 75005 Paris, France; Institut du Fer à Moulin, 75005 Paris, France
| | - Emeline François
- INSERM UMR-S 1270, 75005 Paris, France; Sorbonne Université, 75005 Paris, France; Institut du Fer à Moulin, 75005 Paris, France
| | - Daniel Gomez-Dominguez
- Instituto Cajal, Consejo Superior de Investigaciones Científicas (CSIC), Madrid 28002, Spain
| | - Theano Irinopoulou
- 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.
| |
Collapse
|
39
|
Tang BL. Amyloid Precursor Protein (APP) and GABAergic Neurotransmission. Cells 2019; 8:E550. [PMID: 31174368 PMCID: PMC6627941 DOI: 10.3390/cells8060550] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2019] [Revised: 05/25/2019] [Accepted: 06/06/2019] [Indexed: 12/16/2022] Open
Abstract
The amyloid precursor protein (APP) is the parent polypeptide from which amyloid-beta (Aβ) peptides, key etiological agents of Alzheimer's disease (AD), are generated by sequential proteolytic processing involving β- and γ-secretases. APP mutations underlie familial, early-onset AD, and the involvement of APP in AD pathology has been extensively studied. However, APP has important physiological roles in the mammalian brain, particularly its modulation of synaptic functions and neuronal survival. Recent works have now shown that APP could directly modulate γ-aminobutyric acid (GABA) neurotransmission in two broad ways. Firstly, APP is shown to interact with and modulate the levels and activity of the neuron-specific Potassium-Chloride (K+-Cl-) cotransporter KCC2/SLC12A5. The latter is key to the maintenance of neuronal chloride (Cl-) levels and the GABA reversal potential (EGABA), and is therefore important for postsynaptic GABAergic inhibition through the ionotropic GABAA receptors. Secondly, APP binds to the sushi domain of metabotropic GABAB receptor 1a (GABABR1a). In this regard, APP complexes and is co-transported with GABAB receptor dimers bearing GABABR1a to the axonal presynaptic plasma membrane. On the other hand, secreted (s)APP generated by secretase cleavages could act as a GABABR1a-binding ligand that modulates presynaptic vesicle release. The discovery of these novel roles and activities of APP in GABAergic neurotransmission underlies the physiological importance of APP in postnatal brain function.
Collapse
Affiliation(s)
- Bor Luen Tang
- Department of Biochemistry, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117597, Singapore.
- NUS Graduate School for Integrative Sciences and Engineering, National University of Singapore, Singapore 117597, Singapore.
| |
Collapse
|
40
|
Tillman L, Zhang J. Crossing the Chloride Channel: The Current and Potential Therapeutic Value of the Neuronal K +-Cl - Cotransporter KCC2. BIOMED RESEARCH INTERNATIONAL 2019; 2019:8941046. [PMID: 31240228 PMCID: PMC6556333 DOI: 10.1155/2019/8941046] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/08/2019] [Revised: 04/15/2019] [Accepted: 05/06/2019] [Indexed: 02/05/2023]
Abstract
Chloride (Cl-) homeostasis is an essential process involved in neuronal signalling and cell survival. Inadequate regulation of intracellular Cl- interferes with synaptic signalling and is implicated in several neurological diseases. The main inhibitory neurotransmitter of the central nervous system is γ-aminobutyric acid (GABA). GABA hyperpolarises the membrane potential by activating Cl- permeable GABAA receptor channels (GABAAR). This process is reliant on Cl- extruder K+-Cl- cotransporter 2 (KCC2), which generates the neuron's inward, hyperpolarising Cl- gradient. KCC2 is encoded by the fifth member of the solute carrier 12 family (SLC12A5) and has remained a poorly understood component in the development and severity of many neurological diseases for many years. Recent advancements in next-generation sequencing and specific gene targeting, however, have indicated that loss of KCC2 activity is involved in a number of diseases including epilepsy and schizophrenia. It has also been implicated in neuropathic pain following spinal cord injury. Any variant of SLC12A5 that negatively regulates the transporter's expression may, therefore, be implicated in neurological disease. A recent whole exome study has discovered several causative mutations in patients with epilepsy. Here, we discuss the implications of KCC2 in neurological disease and consider the evolving evidence for KCC2's potential as a therapeutic target.
Collapse
Affiliation(s)
- Luke Tillman
- Institute of Biomedical and Clinical Sciences, Medical School, College of Medicine and Health, University of Exeter, Hatherly Laboratories, Exeter EX4 4PS, UK
| | - Jinwei Zhang
- Institute of Biomedical and Clinical Sciences, Medical School, College of Medicine and Health, University of Exeter, Hatherly Laboratories, Exeter EX4 4PS, UK
| |
Collapse
|
41
|
Pathogenic potential of human SLC12A5 variants causing KCC2 dysfunction. Brain Res 2019; 1710:1-7. [DOI: 10.1016/j.brainres.2018.12.025] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2018] [Revised: 11/21/2018] [Accepted: 12/17/2018] [Indexed: 12/29/2022]
|
42
|
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
|
43
|
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
|
44
|
Rahmanzadeh R, Mehrabi S, Barati M, Ahmadi M, Golab F, Kazmi S, Joghataei MT, Seifi M, Gholipourmalekabadi M. Effect of Co-administration of Bumetanide and Phenobarbital on Seizure Attacks in Temporal Lobe Epilepsy. Basic Clin Neurosci 2018; 9:408-416. [PMID: 30719255 PMCID: PMC6359685 DOI: 10.32598/bcn.9.6.408] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2016] [Revised: 10/20/2016] [Accepted: 01/20/2018] [Indexed: 01/14/2023] Open
Abstract
Introduction: The resistance of temporal lobe epilepsy to classic drugs is thought to be due to disruption in the excitation/inhibition of this pathway. Two chloride transporters, NKCC1 and KCC2, are expressed differently for the excitatory state of Gamma-Amino Butyric Acid (GABA). The present study explored the effect of bumetanide as a selective NKCC1 inhibitor either alone or in combination with the phenobarbital in the pilocarpine model of epilepsy. Methods: An animal model of Status Epilepticus (SE) was induced with pilocarpine in Wistar male rats followed by phenobarbital and or bumetanide or saline administration for 45 days after the induction of SE by Intraperitoneal (IP) injection. The rats were monitored, their behavior was recorded, and after 24 hours they were sacrificed to study the expression of NKCC1 and KCC2 using real time PCR. Results: The data showed that the effects of a combination of bumetanide with phenobarbital on frequency rate and duration of seizure attack were more than those of the phenobarbital alone. In addition, in the bumetanide and combined treatment groups, NKCC1 expression decreased significantly, compared with untreated epileptic animals. A delayed decrement in NKCC1/KCC2 expression ratio after bumetanide application was also observed. Conclusion: The combination of bumetanide with phenobarbital increases the inhibition of SE and maximizes the potential of GABA signaling pathway, and can be considered as an effective therapeutic strategy in patients with epilepsy.
Collapse
Affiliation(s)
- Reza Rahmanzadeh
- Cellular and Molecular Research Center, Iran University of Medical Sciences, Tehran, Iran
| | - Soraya Mehrabi
- Cellular and Molecular Research Center, Iran University of Medical Sciences, Tehran, Iran
| | - Mahmood Barati
- Department of Biotechnology, School of Allied Medicine, Iran University of Medical Science, Tehran, Iran
| | - Milad Ahmadi
- Shefa Neuroscience Research Center, Tehran, Iran
| | - Fereshteh Golab
- Cellular and Molecular Research Center, Iran University of Medical Sciences, Tehran, Iran
| | - Sareh Kazmi
- Cellular and Molecular Research Center, Iran University of Medical Sciences, Tehran, Iran
| | - Mohammad Taghi Joghataei
- Cellular and Molecular Research Center, Iran University of Medical Sciences, Tehran, Iran.,Department of Anatomy, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Morteza Seifi
- Department of Medical Genetics, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, Alberta, Canada
| | - Mazaher Gholipourmalekabadi
- Department of Tissue Engineering & Regenerative Medicine, School of Advanced Technologies in Medicine, Iran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
45
|
Cordshagen A, Busch W, Winklhofer M, Nothwang HG, Hartmann AM. Phosphoregulation of the intracellular termini of K +-Cl - cotransporter 2 (KCC2) enables flexible control of its activity. J Biol Chem 2018; 293:16984-16993. [PMID: 30201606 DOI: 10.1074/jbc.ra118.004349] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2018] [Revised: 09/01/2018] [Indexed: 12/22/2022] Open
Abstract
The pivotal role of K+-Cl- cotransporter 2 (KCC2) in inhibitory neurotransmission and severe human diseases fosters interest in understanding posttranslational regulatory mechanisms such as (de)phosphorylation. Here, the regulatory role of the five bona fide phosphosites Ser31, Thr34, Ser932, Thr999, and Thr1008 was investigated by the use of alanine and aspartate mutants. Tl+-based flux analyses in HEK-293 cells demonstrated increased transport activity for S932D (mimicking phosphorylation) and T1008A (mimicking dephosphorylation), albeit to a different extent. Increased activity was due to changes in intrinsic activity, as it was not caused by increased cell-surface abundance. Substitutions of Ser31, Thr34, or Thr999 had no effect. Additionally, we show that the indirect actions of the known KCC2 activators staurosporine and N-ethylmaleimide (NEM) involved multiple phosphosites. S31D, T34A, S932A/D, T999A, or T1008A/D abrogated staurosporine mediated stimulation, and S31A, T34D, or S932D abolished NEM-mediated stimulation. This demonstrates for the first time differential effects of staurosporine and NEM on KCC2. In addition, the staurosporine-mediated effects involved both KCC2 phosphorylation and dephosphorylation with Ser932 and Thr1008 being bona fide target sites. In summary, our data reveal a complex phosphoregulation of KCC2 that provides the transporter with a toolbox for graded activity and integration of different signaling pathways.
Collapse
Affiliation(s)
- Antje Cordshagen
- From the Neurogenetics group, Center of Excellence Hearing4all, School of Medicine and Health Sciences
| | - Wiebke Busch
- From the Neurogenetics group, Center of Excellence Hearing4all, School of Medicine and Health Sciences
| | - Michael Winklhofer
- Institute for Biology and Environmental Sciences IBU, and.,Research Center for Neurosensory Sciences, Carl von Ossietzky University Oldenburg, 26111 Oldenburg, Germany
| | - Hans Gerd Nothwang
- From the Neurogenetics group, Center of Excellence Hearing4all, School of Medicine and Health Sciences.,Research Center for Neurosensory Sciences, Carl von Ossietzky University Oldenburg, 26111 Oldenburg, Germany
| | - Anna-Maria Hartmann
- From the Neurogenetics group, Center of Excellence Hearing4all, School of Medicine and Health Sciences, .,Research Center for Neurosensory Sciences, Carl von Ossietzky University Oldenburg, 26111 Oldenburg, Germany
| |
Collapse
|
46
|
Carter BM, Sullivan BJ, Landers JR, Kadam SD. Dose-dependent reversal of KCC2 hypofunction and phenobarbital-resistant neonatal seizures by ANA12. Sci Rep 2018; 8:11987. [PMID: 30097625 PMCID: PMC6086916 DOI: 10.1038/s41598-018-30486-7] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2018] [Accepted: 07/31/2018] [Indexed: 01/22/2023] Open
Abstract
Neonatal seizures have an incidence of 3.5 per 1000 newborns; while hypoxic-ischemic encephalopathy (HIE) accounts for 50-60% of cases, half are resistant to 1st-line anti-seizure drugs such as phenobarbital (PB). Tyrosine receptor kinase B (TrkB) activation following ischemic injury is known to increase neuronal excitability by downregulation of K-Cl co-transporter 2 (KCC2); a neuronal chloride (Cl-) co-transporter. In this study, three graded doses of ANA12, a small-molecule selective TrkB antagonist, were tested in CD1 mice at P7 and P10 following induction of neonatal ischemia by a unilateral carotid ligation. The PB loading dose remained the same in all treatment groups at both ages. Evaluation criteria for the anti-seizure efficacy of ANA12 were: (1) quantitative electroencephalographic (EEG) seizure burden and power, (2) rescue of post-ischemic KCC2 and pKCC2-S940 downregulation and (3) reversal of TrkB pathway activation following ischemia. ANA12 significantly rescued PB resistant seizures in a dose-dependent manner at P7 and improved PB efficacy at P10. Additionally, female pups responded better to lower doses of ANA12 compared to males. ANA12 significantly reversed post-ischemic KCC2 downregulation and TrkB pathway activation at P7 when PB alone was inefficacious. Rescuing KCC2 hypofunction may be critical for preventing emergence of refractory seizures.
Collapse
Affiliation(s)
- B M Carter
- Neuroscience Laboratory, Hugo Moser Research Institute at Kennedy Krieger, Baltimore, MD, USA
| | - B J Sullivan
- Neuroscience Laboratory, Hugo Moser Research Institute at Kennedy Krieger, Baltimore, MD, USA
| | - J R Landers
- Neuroscience Laboratory, Hugo Moser Research Institute at Kennedy Krieger, Baltimore, MD, USA
| | - S D Kadam
- Neuroscience Laboratory, Hugo Moser Research Institute at Kennedy Krieger, Baltimore, MD, USA.
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD, 21205, USA.
| |
Collapse
|
47
|
Schulte JT, Wierenga CJ, Bruining H. Chloride transporters and GABA polarity in developmental, neurological and psychiatric conditions. Neurosci Biobehav Rev 2018; 90:260-271. [PMID: 29729285 DOI: 10.1016/j.neubiorev.2018.05.001] [Citation(s) in RCA: 90] [Impact Index Per Article: 12.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2018] [Revised: 03/20/2018] [Accepted: 05/01/2018] [Indexed: 12/22/2022]
Abstract
Neuronal chloride regulation is a determinant factor for the dynamic tuning of GABAergic inhibition during and beyond brain development. This regulation is mainly dependent on the two co-transporters K+/Cl- co-transporter KCC2 and Na+/K+/Cl- co-transporter NKCC1, whose activity can decrease or increase neuronal chloride concentrations respectively. Altered expression and/or activity of either of these co-transporters has been associated with a wide variety of brain disorders including developmental disorders, epilepsy, schizophrenia and stroke. Here, we review current knowledge on chloride transporter expression and activity regulation and highlight the intriguing potential for existing and future interventions to support chloride homeostasis across a wide range of mental disorders and neurological conditions.
Collapse
Affiliation(s)
- Joran T Schulte
- Department of Psychiatry, Brain Center Rudolf Magnus, University Medical Center, Heidelberglaan 100, 3508 GA Utrecht The Netherlands
| | - Corette J Wierenga
- Division of Biology, Faculty of Science, Utrecht University, Padualaan 8, 3584 CH, Utrecht, The Netherlands
| | - Hilgo Bruining
- Department of Psychiatry, Brain Center Rudolf Magnus, University Medical Center, Heidelberglaan 100, 3508 GA Utrecht The Netherlands.
| |
Collapse
|
48
|
Di Cristo G, Awad PN, Hamidi S, Avoli M. KCC2, epileptiform synchronization, and epileptic disorders. Prog Neurobiol 2018; 162:1-16. [DOI: 10.1016/j.pneurobio.2017.11.002] [Citation(s) in RCA: 42] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2017] [Revised: 11/09/2017] [Accepted: 11/28/2017] [Indexed: 12/31/2022]
|
49
|
Hershfinkel M. The Zinc Sensing Receptor, ZnR/GPR39, in Health and Disease. Int J Mol Sci 2018; 19:ijms19020439. [PMID: 29389900 PMCID: PMC5855661 DOI: 10.3390/ijms19020439] [Citation(s) in RCA: 79] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2018] [Revised: 01/24/2018] [Accepted: 01/29/2018] [Indexed: 02/07/2023] Open
Abstract
A distinct G-protein coupled receptor that senses changes in extracellular Zn2+, ZnR/GPR39, was found in cells from tissues in which Zn2+ plays a physiological role. Most prominently, ZnR/GPR39 activity was described in prostate cancer, skin keratinocytes, and colon epithelial cells, where zinc is essential for cell growth, wound closure, and barrier formation. ZnR/GPR39 activity was also described in neurons that are postsynaptic to vesicular Zn2+ release. Activation of ZnR/GPR39 triggers Gαq-dependent signaling and subsequent cellular pathways associated with cell growth and survival. Furthermore, ZnR/GPR39 was shown to regulate the activity of ion transport mechanisms that are essential for the physiological function of epithelial and neuronal cells. Thus, ZnR/GPR39 provides a unique target for therapeutically modifying the actions of zinc in a specific and selective manner.
Collapse
Affiliation(s)
- Michal Hershfinkel
- Department of Physiology and Cell Biology and The Zlotowski Center for Neuroscience, Faculty of Health Sciences, POB 653, Ben-Gurion Ave. Ben-Gurion University of the Negev, Beer Sheva 84105, Israel.
| |
Collapse
|
50
|
Kitayama T. The Role of K +-Cl --Cotransporter-2 in Neuropathic Pain. Neurochem Res 2018; 43:110-115. [PMID: 28677029 DOI: 10.1007/s11064-017-2344-3] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2017] [Revised: 06/09/2017] [Accepted: 06/26/2017] [Indexed: 01/21/2023]
Abstract
The pain sensory system normally functions under a fine balance between excitation and inhibition. When this balance is perturbed for some reason, it leads to neuropathic pain. There is accumulating evidence that attributes this pain generation to specific dysfunctions of the inhibitory system in the spinal cord. One possible mechanism leading to the induction of these dysfunctions is the down-regulation of K+-Cl--cotransporter-2 (KCC2) expression. In fact, various neuropathic pain models indicate a decrease of KCC2 expression in the spinal cord. The alteration of KCC2 expression affects GABAergic and glycinergic neurotransmissions, because KCC2 is a potassium-chloride exporter and serves to maintain intracellular chloride concentration. When there is a low level of KCC2 expression, GABAergic and glycinergic neurotransmissions transform from inhibitory signals to excitatory signals. In this review, the hypothesis that an alteration of KCC2 expression has a crucial influence on the initiation/development or maintenance of neuropathic pain is discussed. In addition, it is suggested that the alteration of inhibitory signals is dependent on the time after peripheral nerve injury.
Collapse
Affiliation(s)
- Tomoya Kitayama
- Department of Pharmacy, School of Pharmacy and Pharmaceutical Science, Mukogawa Women's University, 11-68 Koshien-Kyubanmachi, Nishinomiya, Hyogo, 663-8179, Japan.
| |
Collapse
|