1
|
Gui T, Liu Y, Fu M, Wu H, Su P, Feng X, Zheng M, Huang Z, Luo X, Boron WF, Chen LM. Redox state of NAD modulates the activation of Na-bicarbonate cotransporter NBCe1-B via IRBIT and L-IRBIT. SCIENCE CHINA. LIFE SCIENCES 2025; 68:1452-1462. [PMID: 39985648 PMCID: PMC12097937 DOI: 10.1007/s11427-024-2750-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 08/01/2024] [Accepted: 09/29/2024] [Indexed: 02/24/2025]
Abstract
Nicotinamide adenine dinucleotide (NAD) is well known as a coenzyme involved in many redox reactions in cellular energy metabolism, or as a substrate for many NAD+-consuming enzymes, including those that generate the second messenger cyclic ADP-ribose or deacetylate proteins (e.g., histones). The role of NAD in non-catalytic proteins is poorly understood. IRBIT and L-IRBIT (the IRBITs) are two cytosolic proteins that are structurally related to dehydrogenases but lack catalytic activity. Instead, by interacting directly with their targets, the IRBITs modulate the function of numerous proteins with important roles, ranging from Ca2+ signaling and intracellular pH (pHi) regulation to DNA metabolism to autophagy. Among the targets of the IRBITs is the Na+-HCO3- cotransporter NBCe1-B, which plays a central role in intracellular pH (pHi) regulation and epithelial electrolyte transport. Here, we demonstrate that NAD modulates NBCe1-B activation by serving as a cofactor of IRBIT or L-IRBIT. Blocking NAD salvage pathway greatly decreases NBCe1-B activation by the IRBITs. Administration of the oxidized form NAD+ enhances, whereas the reduced form NADH decreases NBCe1-B activity. Our study represents the first example in which the redox state of NAD, via IRBIT or L-IRBIT, modulates the function of a membrane transport protein. Our findings reveal a new role of NAD and greatly expand our understanding of NAD biology. Because the NAD redox state fluctuates greatly with metabolic status, our work provides insight into how, via the IRBITs, energy metabolism could affect pHi regulation and many other IRBIT-dependent processes.
Collapse
Affiliation(s)
- Tianxiang Gui
- Key Laboratory of Molecular Biophysics of Ministry of Education, College of Life Science & Technology, Huazhong University of Science & Technology, Wuhan, 430074, China
| | - Ying Liu
- Key Laboratory of Molecular Biophysics of Ministry of Education, College of Life Science & Technology, Huazhong University of Science & Technology, Wuhan, 430074, China.
| | - Mingfeng Fu
- Key Laboratory of Molecular Biophysics of Ministry of Education, College of Life Science & Technology, Huazhong University of Science & Technology, Wuhan, 430074, China
| | - Han Wu
- Key Laboratory of Molecular Biophysics of Ministry of Education, College of Life Science & Technology, Huazhong University of Science & Technology, Wuhan, 430074, China
| | - Pan Su
- Key Laboratory of Molecular Biophysics of Ministry of Education, College of Life Science & Technology, Huazhong University of Science & Technology, Wuhan, 430074, China
| | - Xuhui Feng
- Key Laboratory of Molecular Biophysics of Ministry of Education, College of Life Science & Technology, Huazhong University of Science & Technology, Wuhan, 430074, China
| | - Mengmeng Zheng
- Key Laboratory of Molecular Biophysics of Ministry of Education, College of Life Science & Technology, Huazhong University of Science & Technology, Wuhan, 430074, China
| | - Zixuan Huang
- Key Laboratory of Molecular Biophysics of Ministry of Education, College of Life Science & Technology, Huazhong University of Science & Technology, Wuhan, 430074, China
| | - Xudong Luo
- Institute of Biomedicine and Hubei Key Laboratory of Embryonic Stem Cell Research, College of Basic Medicine, Hubei University of Medicine, Shiyan, 442000, China
| | - Walter F Boron
- Department of Physiology and Biophysics, Case Western Reserve University School of Medicine, Cleveland, OH, 44106, USA
| | - Li-Ming Chen
- Key Laboratory of Molecular Biophysics of Ministry of Education, College of Life Science & Technology, Huazhong University of Science & Technology, Wuhan, 430074, China.
- Research Institute of Huazhong University of Science and Technology in Shenzhen, Shenzhen, 518063, China.
| |
Collapse
|
2
|
Chiang WT, Chang YK, Hui WH, Chang SW, Liao CY, Chang YC, Chen CJ, Wang WC, Lai CC, Wang CH, Luo SY, Huang YP, Chou SH, Horng TL, Hou MH, Muench SP, Chen RS, Tsai MD, Hu NJ. Structural basis and synergism of ATP and Na + activation in bacterial K + uptake system KtrAB. Nat Commun 2024; 15:3850. [PMID: 38719864 PMCID: PMC11078986 DOI: 10.1038/s41467-024-48057-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2023] [Accepted: 04/17/2024] [Indexed: 05/12/2024] Open
Abstract
The K+ uptake system KtrAB is essential for bacterial survival in low K+ environments. The activity of KtrAB is regulated by nucleotides and Na+. Previous studies proposed a putative gating mechanism of KtrB regulated by KtrA upon binding to ATP or ADP. However, how Na+ activates KtrAB and the Na+ binding site remain unknown. Here we present the cryo-EM structures of ATP- and ADP-bound KtrAB from Bacillus subtilis (BsKtrAB) both solved at 2.8 Å. A cryo-EM density at the intra-dimer interface of ATP-KtrA was identified as Na+, as supported by X-ray crystallography and ICP-MS. Thermostability assays and functional studies demonstrated that Na+ binding stabilizes the ATP-bound BsKtrAB complex and enhances its K+ flux activity. Comparing ATP- and ADP-BsKtrAB structures suggests that BsKtrB Arg417 and Phe91 serve as a channel gate. The synergism of ATP and Na+ in activating BsKtrAB is likely applicable to Na+-activated K+ channels in central nervous system.
Collapse
Affiliation(s)
- Wesley Tien Chiang
- Graduate Institute of Biochemistry, National Chung Hsing University, Taichung, 402202, Taiwan
| | - Yao-Kai Chang
- Institute of Biological Chemistry, Academia Sinica, Taipei, 115201, Taiwan
| | - Wei-Han Hui
- Department of Civil Engineering, National Taiwan University, Taipei, 106319, Taiwan
| | - Shu-Wei Chang
- Department of Civil Engineering, National Taiwan University, Taipei, 106319, Taiwan
- Department of Biomedical Engineering, National Taiwan University, Taipei, 10663, Taiwan
| | - Chen-Yi Liao
- Graduate Institute of Biochemistry, National Chung Hsing University, Taichung, 402202, Taiwan
| | - Yi-Chuan Chang
- Graduate Institute of Biochemistry, National Chung Hsing University, Taichung, 402202, Taiwan
| | - Chun-Jung Chen
- Life Science Group, Scientific Research Division, National Synchrotron Radiation Research Center, Hsinchu, 30092, Taiwan
| | - Wei-Chen Wang
- Institute of Molecular Biology, National Chung Hsing University, Taichung, 402202, Taiwan
| | - Chien-Chen Lai
- Institute of Molecular Biology, National Chung Hsing University, Taichung, 402202, Taiwan
- Graduate Institute of Chinese Medical Science, China Medical University, Taichung, 406040, Taiwan
| | - Chun-Hsiung Wang
- Institute of Biological Chemistry, Academia Sinica, Taipei, 115201, Taiwan
| | - Siou-Ying Luo
- Institute of Biological Chemistry, Academia Sinica, Taipei, 115201, Taiwan
| | - Ya-Ping Huang
- Institute of Biological Chemistry, Academia Sinica, Taipei, 115201, Taiwan
| | - Shan-Ho Chou
- Graduate Institute of Biochemistry, National Chung Hsing University, Taichung, 402202, Taiwan
| | - Tzyy-Leng Horng
- Department of Applied Mathematics, Feng Chia University, Taichung, 407102, Taiwan
| | - Ming-Hon Hou
- Institute of Genomics and Bioinformatics, National Chung Hsing University, Taichung, 402202, Taiwan
| | - Stephen P Muench
- School of Biomedical Sciences, Faculty of Biological Sciences and the Astbury Centre for Structural Molecular Biology, University of Leeds, Leeds, LS2 9JT, UK
| | - Ren-Shiang Chen
- Department of Life Science, Tunghai University, Taichung, 407224, Taiwan
| | - Ming-Daw Tsai
- Institute of Biological Chemistry, Academia Sinica, Taipei, 115201, Taiwan.
- Institute of Biochemical Sciences, National Taiwan University, Taipei, 106319, Taiwan.
| | - Nien-Jen Hu
- Graduate Institute of Biochemistry, National Chung Hsing University, Taichung, 402202, Taiwan.
- Ph.D Program in Translational Medicine, National Chung Hsing University, Taichung, 402202, Taiwan.
| |
Collapse
|
3
|
Balzulat A, Zhu WF, Flauaus C, Hernandez‐Olmos V, Heering J, Sethumadhavan S, Dubiel M, Frank A, Menge A, Hebchen M, Metzner K, Lu R, Lukowski R, Ruth P, Knapp S, Müller S, Steinhilber D, Hänelt I, Stark H, Proschak E, Schmidtko A. Discovery of a Small Molecule Activator of Slack (Kcnt1) Potassium Channels That Significantly Reduces Scratching in Mouse Models of Histamine-Independent and Chronic Itch. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2307237. [PMID: 38350720 PMCID: PMC11022729 DOI: 10.1002/advs.202307237] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/29/2023] [Revised: 01/15/2024] [Indexed: 02/15/2024]
Abstract
Various disorders are accompanied by histamine-independent itching, which is often resistant to the currently available therapies. Here, it is reported that the pharmacological activation of Slack (Kcnt1, KNa1.1), a potassium channel highly expressed in itch-sensitive sensory neurons, has therapeutic potential for the treatment of itching. Based on the Slack-activating antipsychotic drug, loxapine, a series of new derivatives with improved pharmacodynamic and pharmacokinetic profiles is designed that enables to validate Slack as a pharmacological target in vivo. One of these new Slack activators, compound 6, exhibits negligible dopamine D2 and D3 receptor binding, unlike loxapine. Notably, compound 6 displays potent on-target antipruritic activity in multiple mouse models of acute histamine-independent and chronic itch without motor side effects. These properties make compound 6 a lead molecule for the development of new antipruritic therapies targeting Slack.
Collapse
Affiliation(s)
- Annika Balzulat
- Institute of Pharmacology and Clinical PharmacyGoethe University FrankfurtMax‐von‐Laue‐Str. 960438Frankfurt am MainGermany
| | - W. Felix Zhu
- Institute of Pharmaceutical ChemistryGoethe University FrankfurtMax‐von‐Laue‐Str. 960438Frankfurt am MainGermany
| | - Cathrin Flauaus
- Institute of Pharmacology and Clinical PharmacyGoethe University FrankfurtMax‐von‐Laue‐Str. 960438Frankfurt am MainGermany
| | - Victor Hernandez‐Olmos
- Fraunhofer Institute for Translational Medicine and Pharmacology ITMPTheodor‐Stern‐Kai 760596Frankfurt am MainGermany
| | - Jan Heering
- Fraunhofer Institute for Translational Medicine and Pharmacology ITMPTheodor‐Stern‐Kai 760596Frankfurt am MainGermany
| | - Sunesh Sethumadhavan
- Institute of BiochemistryGoethe University FrankfurtMax‐von‐Laue‐Str. 960438Frankfurt am MainGermany
| | - Mariam Dubiel
- Institute of Pharmaceutical and Medicinal ChemistryHeinrich Heine University DüsseldorfUniversitätsstr. 140225DüsseldorfGermany
| | - Annika Frank
- Institute of Pharmaceutical and Medicinal ChemistryHeinrich Heine University DüsseldorfUniversitätsstr. 140225DüsseldorfGermany
| | - Amelie Menge
- Institute of Pharmaceutical ChemistryGoethe University FrankfurtMax‐von‐Laue‐Str. 960438Frankfurt am MainGermany
- Structural Genomics Consortium (SGC)Buchmann Institute for Molecular Life SciencesGoethe University FrankfurtMax‐von‐Laue‐Str. 1560438Frankfurt am MainGermany
| | - Maureen Hebchen
- Institute of Pharmacology and Clinical PharmacyGoethe University FrankfurtMax‐von‐Laue‐Str. 960438Frankfurt am MainGermany
| | - Katharina Metzner
- Institute of Pharmacology and Clinical PharmacyGoethe University FrankfurtMax‐von‐Laue‐Str. 960438Frankfurt am MainGermany
| | - Ruirui Lu
- Institute of Pharmacology and Clinical PharmacyGoethe University FrankfurtMax‐von‐Laue‐Str. 960438Frankfurt am MainGermany
| | - Robert Lukowski
- Department of PharmacologyToxicology and Clinical PharmacyInstitute of Pharmacy University of TübingenAuf der Morgenstelle 872076TübingenGermany
| | - Peter Ruth
- Department of PharmacologyToxicology and Clinical PharmacyInstitute of Pharmacy University of TübingenAuf der Morgenstelle 872076TübingenGermany
| | - Stefan Knapp
- Institute of Pharmaceutical ChemistryGoethe University FrankfurtMax‐von‐Laue‐Str. 960438Frankfurt am MainGermany
- Structural Genomics Consortium (SGC)Buchmann Institute for Molecular Life SciencesGoethe University FrankfurtMax‐von‐Laue‐Str. 1560438Frankfurt am MainGermany
| | - Susanne Müller
- Institute of Pharmaceutical ChemistryGoethe University FrankfurtMax‐von‐Laue‐Str. 960438Frankfurt am MainGermany
- Structural Genomics Consortium (SGC)Buchmann Institute for Molecular Life SciencesGoethe University FrankfurtMax‐von‐Laue‐Str. 1560438Frankfurt am MainGermany
| | - Dieter Steinhilber
- Institute of Pharmaceutical ChemistryGoethe University FrankfurtMax‐von‐Laue‐Str. 960438Frankfurt am MainGermany
- Fraunhofer Institute for Translational Medicine and Pharmacology ITMPTheodor‐Stern‐Kai 760596Frankfurt am MainGermany
| | - Inga Hänelt
- Institute of BiochemistryGoethe University FrankfurtMax‐von‐Laue‐Str. 960438Frankfurt am MainGermany
| | - Holger Stark
- Institute of Pharmaceutical and Medicinal ChemistryHeinrich Heine University DüsseldorfUniversitätsstr. 140225DüsseldorfGermany
| | - Ewgenij Proschak
- Institute of Pharmaceutical ChemistryGoethe University FrankfurtMax‐von‐Laue‐Str. 960438Frankfurt am MainGermany
- Fraunhofer Institute for Translational Medicine and Pharmacology ITMPTheodor‐Stern‐Kai 760596Frankfurt am MainGermany
| | - Achim Schmidtko
- Institute of Pharmacology and Clinical PharmacyGoethe University FrankfurtMax‐von‐Laue‐Str. 960438Frankfurt am MainGermany
| |
Collapse
|
4
|
Willemen HLDM, Santos Ribeiro PS, Broeks M, Meijer N, Versteeg S, Tiggeler A, de Boer TP, Małecki JM, Falnes PØ, Jans J, Eijkelkamp N. Inflammation-induced mitochondrial and metabolic disturbances in sensory neurons control the switch from acute to chronic pain. Cell Rep Med 2023; 4:101265. [PMID: 37944527 PMCID: PMC10694662 DOI: 10.1016/j.xcrm.2023.101265] [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: 11/16/2022] [Revised: 07/24/2023] [Accepted: 10/10/2023] [Indexed: 11/12/2023]
Abstract
Pain often persists in patients with an inflammatory disease, even when inflammation has subsided. The molecular mechanisms leading to this failure in pain resolution and the transition to chronic pain are poorly understood. Mitochondrial dysfunction in sensory neurons links to chronic pain, but its role in resolution of inflammatory pain is unclear. Transient inflammation causes neuronal plasticity, called hyperalgesic priming, which impairs resolution of pain induced by a subsequent inflammatory stimulus. We identify that hyperalgesic priming in mice increases the expression of a mitochondrial protein (ATPSc-KMT) and causes mitochondrial and metabolic disturbances in sensory neurons. Inhibition of mitochondrial respiration, knockdown of ATPSCKMT expression, or supplementation of the affected metabolite is sufficient to restore resolution of inflammatory pain and prevents chronic pain development. Thus, inflammation-induced mitochondrial-dependent disturbances in sensory neurons predispose to a failure in resolution of inflammatory pain and development of chronic pain.
Collapse
Affiliation(s)
- Hanneke L D M Willemen
- Center for Translational Immunology, Department of Immunology, University Medical Center Utrecht, Utrecht University, 3508 Utrecht, the Netherlands
| | - Patrícia Silva Santos Ribeiro
- Center for Translational Immunology, Department of Immunology, University Medical Center Utrecht, Utrecht University, 3508 Utrecht, the Netherlands
| | - Melissa Broeks
- Section Metabolic Diagnostics, Department of Genetics, University Medical Center Utrecht, Utrecht University, 3508 Utrecht, the Netherlands
| | - Nils Meijer
- Section Metabolic Diagnostics, Department of Genetics, University Medical Center Utrecht, Utrecht University, 3508 Utrecht, the Netherlands
| | - Sabine Versteeg
- Center for Translational Immunology, Department of Immunology, University Medical Center Utrecht, Utrecht University, 3508 Utrecht, the Netherlands
| | - Annefien Tiggeler
- Center for Translational Immunology, Department of Immunology, University Medical Center Utrecht, Utrecht University, 3508 Utrecht, the Netherlands
| | - Teun P de Boer
- Department of Medical Physiology, Division of Heart & Lungs, University Medical Center Utrecht, Yalelaan 50, 3584 Utrecht, the Netherlands
| | - Jędrzej M Małecki
- Department of Biosciences, Faculty of Mathematics and Natural Sciences, University of Oslo, Oslo, Norway; CRES-O - Centre for Embryology and Healthy Development, University of Oslo and Oslo University Hospital, Oslo, Norway
| | - Pål Ø Falnes
- Department of Biosciences, Faculty of Mathematics and Natural Sciences, University of Oslo, Oslo, Norway; CRES-O - Centre for Embryology and Healthy Development, University of Oslo and Oslo University Hospital, Oslo, Norway
| | - Judith Jans
- Section Metabolic Diagnostics, Department of Genetics, University Medical Center Utrecht, Utrecht University, 3508 Utrecht, the Netherlands
| | - Niels Eijkelkamp
- Center for Translational Immunology, Department of Immunology, University Medical Center Utrecht, Utrecht University, 3508 Utrecht, the Netherlands.
| |
Collapse
|
5
|
Zhang J, Liu S, Fan J, Yan R, Huang B, Zhou F, Yuan T, Gong J, Huang Z, Jiang D. Structural basis of human Slo2.2 channel gating and modulation. Cell Rep 2023; 42:112858. [PMID: 37494189 DOI: 10.1016/j.celrep.2023.112858] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2022] [Revised: 06/16/2023] [Accepted: 07/07/2023] [Indexed: 07/28/2023] Open
Abstract
The sodium-activated Slo2.2 channel is abundantly expressed in the brain, playing a critical role in regulating neuronal excitability. The Na+-binding site and the underlying mechanisms of Na+-dependent activation remain unclear. Here, we present cryoelectron microscopy (cryo-EM) structures of human Slo2.2 in closed, open, and inhibitor-bound form at resolutions of 2.6-3.2 Å, revealing gating mechanisms of Slo2.2 regulation by cations and a potent inhibitor. The cytoplasmic gating ring domain of the closed Slo2.2 harbors multiple K+ and Zn2+ sites, which stabilize the channel in the closed conformation. The open Slo2.2 structure reveals at least two Na+-sensitive sites where Na+ binding induces expansion and rotation of the gating ring that opens the inner gate. Furthermore, a potent inhibitor wedges into a pocket formed by pore helix and S6 helix and blocks the pore. Together, our results provide a comprehensive structural framework for the investigation of Slo2.2 channel gating, Na+ sensation, and inhibition.
Collapse
Affiliation(s)
- Jiangtao Zhang
- Laboratory of Soft Matter Physics, Institute of Physics, Chinese Academy of Sciences, Beijing 100190, China; College of Life Science and Technology, Key Laboratory of Molecular Biophysics of MOE, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Shiqi Liu
- State Key Laboratory of Natural and Biomimetic Drugs, Department of Molecular and Cellular Pharmacology, School of Pharmaceutical Sciences, Peking University Health Science Center, Beijing 100191, China; IDG/McGovern Institute for Brain Research, Peking University, Beijing 100871, China
| | - Junping Fan
- Beijing National Laboratory for Molecular Sciences, Key Laboratory of Bioorganic Chemistry and Molecular Engineering of Ministry of Education, Department of Chemical Biology, College of Chemistry and Molecular Engineering, Synthetic and Functional Biomolecules Center, and Peking-Tsinghua Center for Life Sciences, Peking University, Beijing, China
| | - Rui Yan
- Laboratory of Soft Matter Physics, Institute of Physics, Chinese Academy of Sciences, Beijing 100190, China
| | - Bo Huang
- Beijing StoneWise Technology Co Ltd., Haidian District, Beijing, China
| | - Feng Zhou
- Beijing StoneWise Technology Co Ltd., Haidian District, Beijing, China
| | - Tian Yuan
- State Key Laboratory of Natural and Biomimetic Drugs, Department of Molecular and Cellular Pharmacology, School of Pharmaceutical Sciences, Peking University Health Science Center, Beijing 100191, China; IDG/McGovern Institute for Brain Research, Peking University, Beijing 100871, China
| | - Jianke Gong
- College of Life Science and Technology, Key Laboratory of Molecular Biophysics of MOE, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Zhuo Huang
- State Key Laboratory of Natural and Biomimetic Drugs, Department of Molecular and Cellular Pharmacology, School of Pharmaceutical Sciences, Peking University Health Science Center, Beijing 100191, China; IDG/McGovern Institute for Brain Research, Peking University, Beijing 100871, China.
| | - Daohua Jiang
- Laboratory of Soft Matter Physics, Institute of Physics, Chinese Academy of Sciences, Beijing 100190, China; University of Chinese Academy of Sciences, Beijing 100049, China.
| |
Collapse
|
6
|
Potassium channelopathies associated with epilepsy-related syndromes and directions for therapeutic intervention. Biochem Pharmacol 2023; 208:115413. [PMID: 36646291 DOI: 10.1016/j.bcp.2023.115413] [Citation(s) in RCA: 19] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2022] [Revised: 01/06/2023] [Accepted: 01/09/2023] [Indexed: 01/15/2023]
Abstract
A number of mutations to members of several CNS potassium (K) channel families have been identified which result in rare forms of neonatal onset epilepsy, or syndromes of which one prominent characteristic is a form of epilepsy. Benign Familial Neonatal Convulsions or Seizures (BFNC or BFNS), also referred to as Self-Limited Familial Neonatal Epilepsy (SeLNE), results from mutations in 2 members of the KV7 family (KCNQ) of K channels; while generally self-resolving by about 15 weeks of age, these mutations significantly increase the probability of generalized seizure disorders in the adult, in some cases they result in more severe developmental syndromes. Epilepsy of Infancy with Migrating Focal Seizures (EIMSF), or Migrating Partial Seizures of Infancy (MMPSI), is a rare severe form of epilepsy linked primarily to gain of function mutations in a member of the sodium-dependent K channel family, KCNT1 or SLACK. Finally, KCNMA1 channelopathies, including Liang-Wang syndrome (LIWAS), are rare combinations of neurological symptoms including seizure, movement abnormalities, delayed development and intellectual disabilities, with Liang-Wang syndrome an extremely serious polymalformative syndrome with a number of neurological sequelae including epilepsy. These are caused by mutations in the pore-forming subunit of the large-conductance calcium-activated K channel (BK channel) KCNMA1. The identification of these rare but significant channelopathies has resulted in a resurgence of interest in their treatment by direct pharmacological or genetic modulation. We will briefly review the genetics, biophysics and pharmacology of these K channels, their linkage with the 3 syndromes described above, and efforts to more effectively target these syndromes.
Collapse
|
7
|
Therapeutic Drug Monitoring of Quinidine in Pediatric Patients with KCNT1 Genetic Variants. Pharmaceutics 2022; 14:pharmaceutics14102230. [DOI: 10.3390/pharmaceutics14102230] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2022] [Revised: 10/10/2022] [Accepted: 10/11/2022] [Indexed: 11/16/2022] Open
Abstract
Quinidine (QND) is an old antimalarial drug that was used in the early 20th century as an antiarrhythmic agent. Currently, QND is receiving attention for its use in epilepsy of infancy with migrating focal seizures (EIMFS) due to potassium sodium-activated channel subfamily T member 1 (KCNT1) genetic variants. Here, we report the application of Therapeutic Drug Monitoring (TDM) in pediatric patients carrying KCNT1 genetic variants and orally treated with QND for developmental and epileptic encephalopathies (DEE). We measured plasma levels of QND and its metabolite hydroquinidine (H-QND) by using a validated method based on liquid chromatography coupled with mass spectrometry (LC-MS/MS). Three pediatric patients (median age 4.125 years, IQR 2.375–4.125) received increasing doses of QND. Cardiac toxicity was monitored at every dose change. Reduction in seizure frequency ranged from 50 to 90%. Our results show that QND is a promising drug for pediatric patients with DEE due to KCNT1 genetic variants. Although QND blood levels were significantly lower than the therapeutic range as an anti-arrhythmic drug, patients showed a significant improvement in seizure burden. These data underlie the utility of TDM for QND not only to monitor its toxic effects but also to evaluate possible drug–drug interactions.
Collapse
|
8
|
Liang M, Liu H, Yin X, Gong L, Jie H, Wang L, Shi H, He J, Chen P, Lu J, Yin S, Yang J. NAD+ attenuates bilirubin-induced augmentation of voltage-gated calcium currents in neurons of the ventral cochlear nucleus. Neurosci Lett 2022; 784:136747. [PMID: 35724761 DOI: 10.1016/j.neulet.2022.136747] [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: 11/02/2021] [Revised: 04/26/2022] [Accepted: 06/15/2022] [Indexed: 11/30/2022]
Abstract
Nicotinamide adenine dinucleotide (NAD+) is a ubiquitous molecule with wide-ranging roles in several cell processes, such as regulation of calcium homeostasis and protection against cell injuries. However, the roles of NAD+ in neuroprotection is poorly understood. The main neurons in ventral cochlear nucleus (VCN) are highly susceptible to bilirubin-associated excitotoxicity. We investigated the effects of NAD+ on VCN neurons by whole cell patch-clamp recordings. We found that NAD+ effectively reverses and inhibits bilirubin-mediated enhancement of voltage-gated calcium (VGCC) currents in VCN neurons. Moreover, NAD+ itself did not affect VGCC currents. These results collectively suggest that NAD+ may be neuroprotective by attenuating Ca2+ influx to suppress bilirubin-induced intracellular Ca2+ overloads. Our research provides a basis for evaluation of NAD+ as a promising therapeutic target for bilirubin encephalopathy and excitotoxicity associated with other neurological disorders.
Collapse
Affiliation(s)
- Min Liang
- Department of Otorhinolaryngology-Head and Neck Surgery, Xinhua Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Hanwei Liu
- Department of Otorhinolaryngology, the Sixth People's Hospital, Shanghai Jiao Tong University, Shanghai, China
| | - Xinlu Yin
- Department of Otorhinolaryngology-Head and Neck Surgery, Renji Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Lina Gong
- Department of Otorhinolaryngology, the Sixth People's Hospital, Shanghai Jiao Tong University, Shanghai, China
| | - Huiqun Jie
- Department of Otorhinolaryngology-Head and Neck Surgery, Xinhua Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Luyang Wang
- Program in Neurosciences & Mental Health, SickKids Research Institute
| | - Haibo Shi
- Department of Otorhinolaryngology, the Sixth People's Hospital, Shanghai Jiao Tong University, Shanghai, China
| | - Jingchun He
- Department of Otorhinolaryngology-Head and Neck Surgery, Xinhua Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Penghui Chen
- Department of Otorhinolaryngology-Head and Neck Surgery, Xinhua Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Jingrong Lu
- Department of Physiology, University of Toronto, Toronto, ON, Canada
| | - Shankai Yin
- Department of Otorhinolaryngology, the Sixth People's Hospital, Shanghai Jiao Tong University, Shanghai, China
| | - Jun Yang
- Department of Otorhinolaryngology-Head and Neck Surgery, Xinhua Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China.
| |
Collapse
|
9
|
Liu R, Sun L, Wang Y, Jia M, Wang Q, Cai X, Wu J. Double-edged Role of K Na Channels in Brain Tuning: Identifying Epileptogenic Network Micro-Macro Disconnection. Curr Neuropharmacol 2022; 20:916-928. [PMID: 34911427 PMCID: PMC9881102 DOI: 10.2174/1570159x19666211215104829] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2021] [Revised: 11/09/2021] [Accepted: 12/10/2021] [Indexed: 11/22/2022] Open
Abstract
Epilepsy is commonly recognized as a disease driven by generalized hyperexcited and hypersynchronous neural activity. Sodium-activated potassium channels (KNa channels), which are encoded by the Slo 2.2 and Slo 2.1 genes, are widely expressed in the central nervous system and considered as "brakes" to adjust neuronal adaptation through regulating action potential threshold or after-hyperpolarization under physiological condition. However, the variants in KNa channels, especially gain-of-function variants, have been found in several childhood epileptic conditions. Most previous studies focused on mapping the epileptic network on the macroscopic scale while ignoring the value of microscopic changes. Notably, paradoxical role of KNa channels working on individual neuron/microcircuit and the macroscopic epileptic expression highlights the importance of understanding epileptogenic network through combining microscopic and macroscopic methods. Here, we first illustrated the molecular and physiological function of KNa channels on preclinical seizure models and patients with epilepsy. Next, we summarized current hypothesis on the potential role of KNa channels during seizures to provide essential insight into what emerged as a micro-macro disconnection at different levels. Additionally, we highlighted the potential utility of KNa channels as therapeutic targets for developing innovative anti-seizure medications.
Collapse
Affiliation(s)
- Ru Liu
- Beijing Tiantan Hospital, Capital Medical University, Beijing, China;,Advanced Innovation Center for Human Brain Protection, Capital Medical University, Beijing, China;,China National Clinical Research Center for Neurological Diseases, Beijing, China
| | - Lei Sun
- Beijing Tiantan Hospital, Capital Medical University, Beijing, China;,Advanced Innovation Center for Human Brain Protection, Capital Medical University, Beijing, China;,China National Clinical Research Center for Neurological Diseases, Beijing, China
| | | | - Meng Jia
- Beijing Tiantan Hospital, Capital Medical University, Beijing, China;,Advanced Innovation Center for Human Brain Protection, Capital Medical University, Beijing, China;,China National Clinical Research Center for Neurological Diseases, Beijing, China
| | - Qun Wang
- Beijing Tiantan Hospital, Capital Medical University, Beijing, China;,China National Clinical Research Center for Neurological Diseases, Beijing, China
| | - Xiang Cai
- Advanced Innovation Center for Human Brain Protection, Capital Medical University, Beijing, China;,Address correspondence to these authors at the Beijing Tiantan Hospital, Capital Medical University, Beijing, China; Tel: +0086-18062552085; E-mail: Advanced Innovation Center for Human Brain Protection, Capital Medical University, Beijing, China; Tel: +0086-13319285082; E-mail:
| | - Jianping Wu
- Beijing Tiantan Hospital, Capital Medical University, Beijing, China;,Advanced Innovation Center for Human Brain Protection, Capital Medical University, Beijing, China;,China National Clinical Research Center for Neurological Diseases, Beijing, China;,Address correspondence to these authors at the Beijing Tiantan Hospital, Capital Medical University, Beijing, China; Tel: +0086-18062552085; E-mail: Advanced Innovation Center for Human Brain Protection, Capital Medical University, Beijing, China; Tel: +0086-13319285082; E-mail:
| |
Collapse
|
10
|
Liu Y, Zhang FF, Song Y, Wang R, Zhang Q, Shen ZS, Zhang FF, Zhong DY, Wang XH, Guo Q, Tang QY, Zhang Z. The Slack Channel Deletion Causes Mechanical Pain Hypersensitivity in Mice. Front Mol Neurosci 2022; 15:811441. [PMID: 35359569 PMCID: PMC8963359 DOI: 10.3389/fnmol.2022.811441] [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: 11/08/2021] [Accepted: 02/08/2022] [Indexed: 11/13/2022] Open
Abstract
The role of the Slack (also known as Slo2.2, KNa1.1, or KCNT1) channel in pain-sensing is still in debate on which kind of pain it regulates. In the present study, we found that the Slack–/– mice exhibited decreased mechanical pain threshold but normal heat and cold pain sensitivity. Subsequently, X-gal staining, in situ hybridization, and immunofluorescence staining revealed high expression of the Slack channel in Isolectin B4 positive (IB4+) neurons in the dorsal root ganglion (DRG) and somatostatin-positive (SOM+) neurons in the spinal cord. Patch-clamp recordings indicated the firing frequency was increased in both small neurons in DRG and spinal SOM+ neurons in the Slack–/– mice whereas no obvious slow afterhyperpolarization was observed in both WT mice and Slack–/– mice. Furthermore, we found Kcnt1 gene expression in spinal SOM+ neurons in Slack–/– mice partially relieved the mechanical pain hypersensitivity of Slack–/– mice and decreased AP firing rates of the spinal SOM+ neurons. Finally, deletion of the Slack channel in spinal SOM+ neurons is sufficient to result in mechanical pain hypersensitivity in mice. In summary, our results suggest the important role of the Slack channel in the regulation of mechanical pain-sensing both in small neurons in DRG and SOM+ neurons in the spinal dorsal horn.
Collapse
Affiliation(s)
- Ye Liu
- Jiangsu Province Key Laboratory of Anesthesiology, Xuzhou Medical University, Xuzhou, China
- Jiangsu Province Key Laboratory of Anesthesia and Analgesia Application Technology, Xuzhou Medical University, Xuzhou, China
- NMPA Key Laboratory for Research and Evaluation of Narcotic and Psychotropic Drugs, Xuzhou Medical University, Xuzhou, China
| | - Fang-Fang Zhang
- Jiangsu Province Key Laboratory of Anesthesiology, Xuzhou Medical University, Xuzhou, China
- Jiangsu Province Key Laboratory of Anesthesia and Analgesia Application Technology, Xuzhou Medical University, Xuzhou, China
- NMPA Key Laboratory for Research and Evaluation of Narcotic and Psychotropic Drugs, Xuzhou Medical University, Xuzhou, China
| | - Ying Song
- Jiangsu Province Key Laboratory of Anesthesiology, Xuzhou Medical University, Xuzhou, China
- Jiangsu Province Key Laboratory of Anesthesia and Analgesia Application Technology, Xuzhou Medical University, Xuzhou, China
- NMPA Key Laboratory for Research and Evaluation of Narcotic and Psychotropic Drugs, Xuzhou Medical University, Xuzhou, China
| | - Ran Wang
- Jiangsu Province Key Laboratory of Anesthesiology, Xuzhou Medical University, Xuzhou, China
- Jiangsu Province Key Laboratory of Anesthesia and Analgesia Application Technology, Xuzhou Medical University, Xuzhou, China
| | - Qi Zhang
- Jiangsu Province Key Laboratory of Anesthesiology, Xuzhou Medical University, Xuzhou, China
- Jiangsu Province Key Laboratory of Anesthesia and Analgesia Application Technology, Xuzhou Medical University, Xuzhou, China
- NMPA Key Laboratory for Research and Evaluation of Narcotic and Psychotropic Drugs, Xuzhou Medical University, Xuzhou, China
| | - Zhong-Shan Shen
- Jiangsu Province Key Laboratory of Anesthesiology, Xuzhou Medical University, Xuzhou, China
- Jiangsu Province Key Laboratory of Anesthesia and Analgesia Application Technology, Xuzhou Medical University, Xuzhou, China
- NMPA Key Laboratory for Research and Evaluation of Narcotic and Psychotropic Drugs, Xuzhou Medical University, Xuzhou, China
| | - Fei-Fei Zhang
- Jiangsu Province Key Laboratory of Anesthesiology, Xuzhou Medical University, Xuzhou, China
- Jiangsu Province Key Laboratory of Anesthesia and Analgesia Application Technology, Xuzhou Medical University, Xuzhou, China
- NMPA Key Laboratory for Research and Evaluation of Narcotic and Psychotropic Drugs, Xuzhou Medical University, Xuzhou, China
| | - Dan-Ya Zhong
- Jiangsu Province Key Laboratory of Anesthesiology, Xuzhou Medical University, Xuzhou, China
- Jiangsu Province Key Laboratory of Anesthesia and Analgesia Application Technology, Xuzhou Medical University, Xuzhou, China
- NMPA Key Laboratory for Research and Evaluation of Narcotic and Psychotropic Drugs, Xuzhou Medical University, Xuzhou, China
| | - Xiao-Hui Wang
- Jiangsu Province Key Laboratory of Anesthesiology, Xuzhou Medical University, Xuzhou, China
- Jiangsu Province Key Laboratory of Anesthesia and Analgesia Application Technology, Xuzhou Medical University, Xuzhou, China
- NMPA Key Laboratory for Research and Evaluation of Narcotic and Psychotropic Drugs, Xuzhou Medical University, Xuzhou, China
| | - Qing Guo
- Jiangsu Province Key Laboratory of Anesthesiology, Xuzhou Medical University, Xuzhou, China
- Jiangsu Province Key Laboratory of Anesthesia and Analgesia Application Technology, Xuzhou Medical University, Xuzhou, China
- NMPA Key Laboratory for Research and Evaluation of Narcotic and Psychotropic Drugs, Xuzhou Medical University, Xuzhou, China
| | - Qiong-Yao Tang
- Jiangsu Province Key Laboratory of Anesthesiology, Xuzhou Medical University, Xuzhou, China
- Jiangsu Province Key Laboratory of Anesthesia and Analgesia Application Technology, Xuzhou Medical University, Xuzhou, China
- NMPA Key Laboratory for Research and Evaluation of Narcotic and Psychotropic Drugs, Xuzhou Medical University, Xuzhou, China
- *Correspondence: Qiong-Yao Tang,
| | - Zhe Zhang
- Jiangsu Province Key Laboratory of Anesthesiology, Xuzhou Medical University, Xuzhou, China
- Jiangsu Province Key Laboratory of Anesthesia and Analgesia Application Technology, Xuzhou Medical University, Xuzhou, China
- NMPA Key Laboratory for Research and Evaluation of Narcotic and Psychotropic Drugs, Xuzhou Medical University, Xuzhou, China
- Zhe Zhang,
| |
Collapse
|
11
|
Zhang Q, Liu Y, Xu J, Teng Y, Zhang Z. The Functional Properties, Physiological Roles, Channelopathy and Pharmacological Characteristics of the Slack (KCNT1) Channel. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2022; 1349:387-400. [PMID: 35138624 DOI: 10.1007/978-981-16-4254-8_18] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
The KCNT1 gene encodes the sodium-activated potassium channel that is abundantly expressed in the central nervous system of mammalians and plays an important role in reducing neuronal excitability. Structurally, the KCNT1 channel is absent of voltage sensor but possesses a long C-terminus including RCK1 and RCK2domain, to which the intracellular sodium and chloride bind to activate the channel. Recent publications using electron cryo-microscopy (cryo-EM) revealed the open and closed structural characteristics of the KCNT1 channel and co-assembly of functional domains. The activation of the KCNT1 channel regulates various physiological processes including nociceptive behavior, itch, spatial learning. Meanwhile, malfunction of this channel causes important pathophysiological consequences, including Fragile X syndrome and a wide spectrum of seizure disorders. This review comprehensively describes the structure, expression patterns, physiological functions of the KCNT1 channel and emphasizes the channelopathy of gain-of-function KCNT1 mutations in epilepsy.
Collapse
Affiliation(s)
- Qi Zhang
- Jiangsu Province Key Laboratory of Anesthesiology, Xuzhou Medical University, Xuzhou, Jiangsu Province, China
- Jiangsu Province Key Laboratory of Anesthesia and Analgesia Application Technology, Xuzhou Medical University, Xuzhou, Jiangsu Province, China
| | - Ye Liu
- Jiangsu Province Key Laboratory of Anesthesiology, Xuzhou Medical University, Xuzhou, Jiangsu Province, China
- Jiangsu Province Key Laboratory of Anesthesia and Analgesia Application Technology, Xuzhou Medical University, Xuzhou, Jiangsu Province, China
| | - Jie Xu
- Jiangsu Province Key Laboratory of Anesthesiology, Xuzhou Medical University, Xuzhou, Jiangsu Province, China
- Jiangsu Province Key Laboratory of Anesthesia and Analgesia Application Technology, Xuzhou Medical University, Xuzhou, Jiangsu Province, China
| | - Yue Teng
- Jiangsu Province Key Laboratory of Anesthesiology, Xuzhou Medical University, Xuzhou, Jiangsu Province, China
- Jiangsu Province Key Laboratory of Anesthesia and Analgesia Application Technology, Xuzhou Medical University, Xuzhou, Jiangsu Province, China
| | - Zhe Zhang
- Jiangsu Province Key Laboratory of Anesthesiology, Xuzhou Medical University, Xuzhou, Jiangsu Province, China.
- Jiangsu Province Key Laboratory of Anesthesia and Analgesia Application Technology, Xuzhou Medical University, Xuzhou, Jiangsu Province, China.
| |
Collapse
|
12
|
Slo2/K Na Channels in Drosophila Protect against Spontaneous and Induced Seizure-like Behavior Associated with an Increased Persistent Na + Current. J Neurosci 2021; 41:9047-9063. [PMID: 34544836 DOI: 10.1523/jneurosci.0290-21.2021] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2021] [Revised: 08/20/2021] [Accepted: 09/13/2021] [Indexed: 11/21/2022] Open
Abstract
Na+ sensitivity is a unique feature of Na+-activated K+ (KNa) channels, making them naturally suited to counter a sudden influx in Na+ ions. As such, it has long been suggested that KNa channels may serve a protective function against excessive excitation associated with neuronal injury and disease. This hypothesis, however, has remained largely untested. Here, we examine KNa channels encoded by the Drosophila Slo2 (dSlo2) gene in males and females. We show that dSlo2/KNa channels are selectively expressed in cholinergic neurons in the adult brain, as well as in glutamatergic motor neurons, where dampening excitation may function to inhibit global hyperactivity and seizure-like behavior. Indeed, we show that effects of feeding Drosophila a cholinergic agonist are exacerbated by the loss of dSlo2/KNa channels. Similar to mammalian Slo2/KNa channels, we show that dSlo2/KNa channels encode a TTX-sensitive K+ conductance, indicating that dSlo2/KNa channels can be activated by Na+ carried by voltage-dependent Na+ channels. We then tested the role of dSlo2/KNa channels in established genetic seizure models in which the voltage-dependent persistent Na+ current (INap) is elevated. We show that the absence of dSlo2/KNa channels increased susceptibility to mechanically induced seizure-like behavior. Similar results were observed in WT flies treated with veratridine, an enhancer of INap Finally, we show that loss of dSlo2/KNa channels in both genetic and pharmacologically primed seizure models resulted in the appearance of spontaneous seizures. Together, our results support a model in which dSlo2/KNa channels, activated by neuronal overexcitation, contribute to a protective threshold to suppress the induction of seizure-like activity.SIGNIFICANCE STATEMENT Slo2/KNa channels are unique in that they constitute a repolarizing K+ pore that is activated by the depolarizing Na+ ion, making them naturally suited to function as a protective "brake" against overexcitation and Na+ overload. Here, we test this hypothesis in vivo by examining how a null mutation of the Drosophila Slo2 (dSlo2)/KNa gene affects seizure-like behavior in genetic and pharmacological models of epilepsy. We show that indeed the loss of dSlo2/KNa channels results in increased incidence and severity of induced seizure behavior, as well as the appearance of spontaneous seizure activity. Our results advance our understanding of neuronal excitability and protective mechanisms that preserve normal physiology and the suppression of seizure susceptibility.
Collapse
|
13
|
Ehinger R, Kuret A, Matt L, Frank N, Wild K, Kabagema-Bilan C, Bischof H, Malli R, Ruth P, Bausch AE, Lukowski R. Slack K + channels attenuate NMDA-induced excitotoxic brain damage and neuronal cell death. FASEB J 2021; 35:e21568. [PMID: 33817875 DOI: 10.1096/fj.202002308rr] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2020] [Revised: 03/14/2021] [Accepted: 03/17/2021] [Indexed: 12/13/2022]
Abstract
The neuronal Na+ -activated K+ channel Slack (aka Slo2.2, KNa 1.1, or Kcnt1) has been implicated in setting and maintaining the resting membrane potential and defining excitability and firing patterns, as well as in the generation of the slow afterhyperpolarization following bursts of action potentials. Slack activity increases significantly under conditions of high intracellular Na+ levels, suggesting this channel may exert important pathophysiological functions. To address these putative roles, we studied whether Slack K+ channels contribute to pathological changes and excitotoxic cell death caused by glutamatergic overstimulation of Ca2+ - and Na+ -permeable N-methyl-D-aspartic acid receptors (NMDAR). Slack-deficient (Slack KO) and wild-type (WT) mice were subjected to intrastriatal microinjections of the NMDAR agonist NMDA. NMDA-induced brain lesions were significantly increased in Slack KO vs WT mice, suggesting that the lack of Slack renders neurons particularly susceptible to excitotoxicity. Accordingly, excessive neuronal cell death was seen in Slack-deficient primary cerebellar granule cell (CGC) cultures exposed to glutamate and NMDA. Differences in neuronal survival between WT and Slack KO CGCs were largely abolished by the NMDAR antagonist MK-801, but not by NBQX, a potent and highly selective competitive antagonist of α-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid (AMPA)-type ionotropic glutamate receptors. Interestingly, NMDAR-evoked Ca2+ signals did not differ with regard to Slack genotype in CGCs. However, real-time monitoring of K+ following NMDAR activation revealed a significant contribution of this channel to the intracellular drop in K+ . Finally, TrkB and TrkC neurotrophin receptor transcript levels were elevated in NMDA-exposed Slack-proficient CGCs, suggesting a mechanism by which this K+ channel contributes to the activation of the extracellular-signal-regulated kinase (Erk) pathway and thereby to neuroprotection. Combined, our findings suggest that Slack-dependent K+ signals oppose the NMDAR-mediated excitotoxic neuronal injury by promoting pro-survival signaling via the BDNF/TrkB and Erk axis.
Collapse
Affiliation(s)
- Rebekka Ehinger
- Department of Pharmacology, Toxicology and Clinical Pharmacy, Institute of Pharmacy, University of Tübingen, Tübingen, Germany
| | - Anna Kuret
- Department of Pharmacology, Toxicology and Clinical Pharmacy, Institute of Pharmacy, University of Tübingen, Tübingen, Germany
| | - Lucas Matt
- Department of Pharmacology, Toxicology and Clinical Pharmacy, Institute of Pharmacy, University of Tübingen, Tübingen, Germany
| | - Nadine Frank
- Department of Pharmacology, Toxicology and Clinical Pharmacy, Institute of Pharmacy, University of Tübingen, Tübingen, Germany
| | - Katharina Wild
- Department of Pharmacology, Toxicology and Clinical Pharmacy, Institute of Pharmacy, University of Tübingen, Tübingen, Germany
| | - Clement Kabagema-Bilan
- Department of Pharmacology, Toxicology and Clinical Pharmacy, Institute of Pharmacy, University of Tübingen, Tübingen, Germany
| | - Helmut Bischof
- Department of Pharmacology, Toxicology and Clinical Pharmacy, Institute of Pharmacy, University of Tübingen, Tübingen, Germany
| | - Roland Malli
- Gottfried Schatz Research Center, Molecular Biology and Biochemistry, Medical University of Graz, Graz, Austria
| | - Peter Ruth
- Department of Pharmacology, Toxicology and Clinical Pharmacy, Institute of Pharmacy, University of Tübingen, Tübingen, Germany
| | - Anne E Bausch
- Department of Pharmacology, Toxicology and Clinical Pharmacy, Institute of Pharmacy, University of Tübingen, Tübingen, Germany
| | - Robert Lukowski
- Department of Pharmacology, Toxicology and Clinical Pharmacy, Institute of Pharmacy, University of Tübingen, Tübingen, Germany
| |
Collapse
|
14
|
Bonardi CM, Heyne HO, Fiannacca M, Fitzgerald MP, Gardella E, Gunning B, Olofsson K, Lesca G, Verbeek N, Stamberger H, Striano P, Zara F, Mancardi MM, Nava C, Syrbe S, Buono S, Baulac S, Coppola A, Weckhuysen S, Schoonjans AS, Ceulemans B, Sarret C, Baumgartner T, Muhle H, des Portes V, Toulouse J, Nougues MC, Rossi M, Demarquay G, Ville D, Hirsch E, Maurey H, Willems M, de Bellescize J, Altuzarra CD, Villeneuve N, Bartolomei F, Picard F, Hornemann F, Koolen DA, Kroes HY, Reale C, Fenger CD, Tan WH, Dibbens L, Bearden DR, Møller RS, Rubboli G. KCNT1-related epilepsies and epileptic encephalopathies: phenotypic and mutational spectrum. Brain 2021; 144:3635-3650. [PMID: 34114611 DOI: 10.1093/brain/awab219] [Citation(s) in RCA: 51] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2020] [Revised: 03/02/2021] [Accepted: 05/11/2021] [Indexed: 11/15/2022] Open
Abstract
Variants in KCNT1, encoding a sodium-gated potassium channel (subfamily T member 1), have been associated with a spectrum of epilepsies and neurodevelopmental disorders. These range from familial autosomal dominant or sporadic sleep-related hypermotor epilepsy ((AD)SHE) to epilepsy of infancy with migrating focal seizures (EIMFS) and include developmental and epileptic encephalopathies (DEE). This study aims to provide a comprehensive overview of the phenotypic and genotypic spectrum of KCNT1 mutation-related epileptic disorders in 248 individuals, including 66 unpreviously published and 182 published cases, the largest cohort reported so far. Four phenotypic groups emerged from our analysis: i) EIMFS (152 individuals, 33 previously unpublished); ii) DEE other than EIMFS (non-EIMFS DEE) (37 individuals, 17 unpublished); iii) (AD)SHE (53 patients, 14 unpublished); iv) other phenotypes (6 individuals, 2 unpublished). In our cohort of 66 new cases, the most common phenotypic features were: a) in EIMFS, heterogeneity of seizure types, including epileptic spasms, epilepsy improvement over time, no epilepsy-related deaths; b) in non-EIMFS DEE, possible onset with West syndrome, occurrence of atypical absences, possible evolution to DEE with SHE features; one case of sudden unexplained death in epilepsy (SUDEP); c) in (AD)SHE, we observed a high prevalence of drug-resistance, although seizure frequency improved with age in some individuals, appearance of cognitive regression after seizure onset in all patients, no reported severe psychiatric disorders, although behavioural/psychiatric comorbidities were reported in about 50% of the patients, SUDEP in one individual; d) other phenotypes in individuals with mutation of KCNT1 included temporal lobe epilepsy, and epilepsy with tonic-clonic seizures and cognitive regression. Genotypic analysis of the whole cohort of 248 individuals showed only missense mutations and one inframe deletion in KCNT1. Although the KCNT1 mutations in affected individuals were seen to be distributed among the different domains of the KCNT1 protein, genotype-phenotype considerations showed many of the (AD)SHE-associated mutations to be clustered around the RCK2 domain in the C-terminus, distal to the NADP domain. Mutations associated with EIMFS/non-EIMFS DEE did not show a particular pattern of distribution in the KCNT1 protein. Recurrent KCNT1 mutations were seen to be associated with both severe and less severe phenotypes. Our study further defines and broadens the phenotypic and genotypic spectrums of KCNT1-related epileptic conditions and emphasizes the increasingly important role of this gene in the pathogenesis of early onset DEEs as well as in focal epilepsies, namely (AD)SHE.
Collapse
Affiliation(s)
- Claudia M Bonardi
- Department of Epilepsy Genetics and Precision Medicine, Danish Epilepsy Centre, member of the ERN EpiCARE, 4293 Dianalund, Denmark.,Department of Woman's and Child's Health, University Hospital of Padua, 35100 Padua, Italy
| | - Henrike O Heyne
- Finnish Institute for Molecular Medicine: FIMM, University of Helsinki, 00290 Helsinki, Finland.,Program for Medical and Population Genetics, Broad Institute of MIT and Harvard, 02142 Cambridge, MA, USA
| | | | - Mark P Fitzgerald
- Division of Neurology, Departments of Neurology and Pediatrics, The Children's Hospital of Philadelphia and the Perelman School of Medicine at the University of Pennsylvania, Philadelphia PA, USA
| | - Elena Gardella
- Department of Epilepsy Genetics and Precision Medicine, Danish Epilepsy Centre, member of the ERN EpiCARE, 4293 Dianalund, Denmark.,Institute of Regional Health Research, University of Southern Denmark, 5230 Odense, Denmark
| | - Boudewijn Gunning
- Stichting Epilepsie Instellingen Nederland, Zwolle, 8025 BV, The Netherlands
| | - Kern Olofsson
- Department of Pediatric Neurology, Danish Epilepsy Center, 4293 Dianalund, Denmark
| | - Gaétan Lesca
- Department of Genetics, Hospices Civils de Lyon, 69002 Bron, France.,Institut NeuroMyoGène, CNRS UMR 5310 - INSERM U1217, Université Claude Bernard Lyon 1, 69008 Lyon, France
| | - Nienke Verbeek
- Department of Genetics, University Medical Center, 3584 CX Utrecht, Netherlands
| | - Hannah Stamberger
- Neurogenetics Group, VIB-Center for Molecular Neurology, B-2610 Antwerp, Belgium.,Department of Neurology, University Hospital, 2650 Antwerp, Belgium
| | - Pasquale Striano
- IRCCS "G. Gaslini" Institute, University of Genoa, 16147 Genoa, Italy
| | - Federico Zara
- IRCCS "G. Gaslini" Institute, University of Genoa, 16147 Genoa, Italy
| | - Maria M Mancardi
- Child Neuropsychiatry Unit, IRCCS Istituto Giannina Gaslini, 16147 Genoa, Italy
| | - Caroline Nava
- Département de Génétique, APHP, GH Pitié-Salpêtrière, 75013 Paris, France
| | - Steffen Syrbe
- Division of Pediatric Epileptology, Centre for Paediatrics and Adolescent Medicine, University Hospital Heidelberg, 69120 Heidelberg, Germany
| | - Salvatore Buono
- Neurology Division, Hospital of National Relevance (AORN), Santobono Pausilipon, 80122 Naples, Italy
| | - Stephanie Baulac
- Sorbonne Université, Institut du Cerveau - Paris Brain Institute - ICM, Inserm, CNRS, F-75013, Paris, France
| | - Antonietta Coppola
- Department of Neuroscience and Reproductive and Odontostomatological Sciences, Federico II University, 80138 Naples, Italy
| | - Sarah Weckhuysen
- Neurogenetics Group, VIB-Center for Molecular Neurology, B-2610 Antwerp, Belgium.,Department of Neurology, University Hospital, 2650 Antwerp, Belgium
| | - An-Sofie Schoonjans
- Department of Pediatric Neurology, Antwerp University Hospital, University of Antwerp, 2650 Edegem, Belgium
| | - Berten Ceulemans
- Department of Pediatric Neurology, Antwerp University Hospital, University of Antwerp, 2650 Edegem, Belgium
| | - Catherine Sarret
- Service de Neuropédiatrie, CHU de Clermont-Ferrand, 6310 Clermont-Ferrand, France
| | | | - Hiltrud Muhle
- Department of Neuropediatrics, University Medical Center Schleswig Holstein, 24105 Kiel, Germany
| | - Vincent des Portes
- Neuropaediatrics Department, Femme Mère Enfant Hospital, 69500 Lyon, France
| | - Joseph Toulouse
- Epileptology, Sleep Disorders and Functional Pediatric Neurology CHU Lyon, 69500 Bron, France
| | | | - Massimiliano Rossi
- Department of Genetics, Hospices Civils de Lyon, 69002 Bron, France.,Lyon Neuroscience Research Center (CRNL), INSERM U1028, CNRS UMR5292, GENDEV Team, Claude Bernard Lyon 1 University, 69675 Bron, France
| | - Geneviève Demarquay
- Service de neurologie fonctionnelle et épileptologie, Neurological Hospital, 69677 Bron, France.,Lyon Neuroscience Research Center (CRNL), INSERM U1028, CNRS UMR5292, NeuroPain, 69677 Bron, France
| | - Dorothée Ville
- Pediatric Neurology Department, Lyon University Hospital, 69500 Bron, France
| | - Edouard Hirsch
- Epilepsy Unit, Hautepierre Hospital, University of Strasbourg, 67100 Strasbourg, France
| | - Hélène Maurey
- Department of Pediatric Neurology, Hopital Bicêtre, Le Kremlin-Bicêtre, 94270 Paris, France
| | - Marjolaine Willems
- Department of Clinical Genetics, Arnaud de Villeneuve Hospital, 34090 Montpellier, France
| | - Julitta de Bellescize
- Department of Pediatric Clinical Epileptology, Sleep Disorders and Functional Neurology, Hospices Civils de Lyon, 69677 Bron, Lyon, France
| | | | - Nathalie Villeneuve
- Pediatric Neurology Department, Timone Children Hospital, 13005 Marseille, France
| | - Fabrice Bartolomei
- Epileptology Department, Timone Hospital, Public Assistance Hospitals of Marseille, Aix-Marseille University, 13005 Marseille, France
| | - Fabienne Picard
- Department of Clinical Neurosciences, University Hospitals and Faculty of Medicine, CH-1211 Geneva, Switzerland
| | - Frauke Hornemann
- Centre of Pediatric Research, Hospital for Children and Adolescents, 04103 Leipzig, Germany
| | - David A Koolen
- Department of Human Genetics, Donders Institute for Brain, Cognition and Behaviour, Radboud university medical center (Radboudumc), 6525 GA Nijmegen, The Netherlands
| | - Hester Y Kroes
- Department of Genetics, University Medical Center, 3584 CX Utrecht, Netherlands
| | - Chiara Reale
- Department of Epilepsy Genetics and Precision Medicine, Danish Epilepsy Centre, member of the ERN EpiCARE, 4293 Dianalund, Denmark.,Department of Clinical and Experimental Medicine, University of Messina, 98122 Messina, Italy
| | - Christina D Fenger
- Department of Epilepsy Genetics and Precision Medicine, Danish Epilepsy Centre, member of the ERN EpiCARE, 4293 Dianalund, Denmark
| | - Wen-Hann Tan
- Division of Genetics and Genomics, Boston Children's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Leanne Dibbens
- Epilepsy Research Group, UniSA Clinical and Health Sciences, University of South Australia, and Australian Centre for Precision Health, SA 5001 Adelaide, Australia
| | - David R Bearden
- Division of Child Neurology, Department of Neurology, University of Rochester School of Medicine, Rochester, NY14642, USA
| | - Rikke S Møller
- Department of Epilepsy Genetics and Precision Medicine, Danish Epilepsy Centre, member of the ERN EpiCARE, 4293 Dianalund, Denmark.,Institute of Regional Health Research, University of Southern Denmark, 5230 Odense, Denmark
| | - Guido Rubboli
- Department of Epilepsy Genetics and Precision Medicine, Danish Epilepsy Centre, member of the ERN EpiCARE, 4293 Dianalund, Denmark.,Institute of Clinical Medicine, University of Copenhagen, 2200 Copenhagen, Denmark
| |
Collapse
|
15
|
Cole BA, Clapcote SJ, Muench SP, Lippiat JD. Targeting K Na1.1 channels in KCNT1-associated epilepsy. Trends Pharmacol Sci 2021; 42:700-713. [PMID: 34074526 DOI: 10.1016/j.tips.2021.05.003] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2021] [Revised: 04/30/2021] [Accepted: 05/05/2021] [Indexed: 12/19/2022]
Abstract
Gain-of-function (GOF) pathogenic variants of KCNT1, the gene encoding the largest known potassium channel subunit, KNa1.1, are associated with developmental and epileptic encephalopathies accompanied by severe psychomotor and intellectual disabilities. Blocking hyperexcitable KNa1.1 channels with quinidine, a class I antiarrhythmic drug, has shown variable success in patients in part because of dose-limiting off-target effects, poor blood-brain barrier (BBB) penetration, and low potency. In recent years, high-resolution cryogenic electron microscopy (cryo-EM) structures of the chicken KNa1.1 channel in different activation states have been determined, and animal models of the diseases have been generated. Alongside increasing information about the functional effects of GOF pathogenic variants on KNa1.1 channel behaviour and how they lead to hyperexcitability, these tools will facilitate the development of more effective treatment strategies. We review the range of KCNT1 variants and their functional effects, the challenges posed by current treatment strategies, and recent advances in finding more potent and selective therapeutic interventions for KCNT1-related epilepsies.
Collapse
Affiliation(s)
- Bethan A Cole
- School of Biomedical Sciences, University of Leeds, Leeds LS2 9JT, UK
| | - Steven J Clapcote
- School of Biomedical Sciences, University of Leeds, Leeds LS2 9JT, UK
| | - Stephen P Muench
- School of Biomedical Sciences, University of Leeds, Leeds LS2 9JT, UK
| | | |
Collapse
|
16
|
Stautz J, Hellmich Y, Fuss MF, Silberberg JM, Devlin JR, Stockbridge RB, Hänelt I. Molecular Mechanisms for Bacterial Potassium Homeostasis. J Mol Biol 2021; 433:166968. [PMID: 33798529 DOI: 10.1016/j.jmb.2021.166968] [Citation(s) in RCA: 79] [Impact Index Per Article: 19.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2021] [Revised: 03/11/2021] [Accepted: 03/22/2021] [Indexed: 10/21/2022]
Abstract
Potassium ion homeostasis is essential for bacterial survival, playing roles in osmoregulation, pH homeostasis, regulation of protein synthesis, enzyme activation, membrane potential adjustment and electrical signaling. To accomplish such diverse physiological tasks, it is not surprising that a single bacterium typically encodes several potassium uptake and release systems. To understand the role each individual protein fulfills and how these proteins work in concert, it is important to identify the molecular details of their function. One needs to understand whether the systems transport ions actively or passively, and what mechanisms or ligands lead to the activation or inactivation of individual systems. Combining mechanistic information with knowledge about the physiology under different stress situations, such as osmostress, pH stress or nutrient limitation, one can identify the task of each system and deduce how they are coordinated with each other. By reviewing the general principles of bacterial membrane physiology and describing the molecular architecture and function of several bacterial K+-transporting systems, we aim to provide a framework for microbiologists studying bacterial potassium homeostasis and the many K+-translocating systems that are still poorly understood.
Collapse
Affiliation(s)
- Janina Stautz
- Institute of Biochemistry, Goethe University Frankfurt, Frankfurt am Main, Germany
| | - Yvonne Hellmich
- Institute of Biochemistry, Goethe University Frankfurt, Frankfurt am Main, Germany
| | - Michael F Fuss
- Institute of Biochemistry, Goethe University Frankfurt, Frankfurt am Main, Germany
| | - Jakob M Silberberg
- Institute of Biochemistry, Goethe University Frankfurt, Frankfurt am Main, Germany
| | - Jason R Devlin
- Department of Molecular, Cellular, and Developmental Biology, University of Michigan, Ann Arbor, MI, United States
| | - Randy B Stockbridge
- Department of Molecular, Cellular, and Developmental Biology, University of Michigan, Ann Arbor, MI, United States.
| | - Inga Hänelt
- Institute of Biochemistry, Goethe University Frankfurt, Frankfurt am Main, Germany.
| |
Collapse
|
17
|
A Role for Transmembrane Protein 16C/Slack Impairment in Excitatory Nociceptive Synaptic Plasticity in the Pathogenesis of Remifentanil-induced Hyperalgesia in Rats. Neurosci Bull 2021; 37:669-683. [PMID: 33779892 DOI: 10.1007/s12264-021-00652-5] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2020] [Accepted: 09/09/2020] [Indexed: 10/21/2022] Open
Abstract
Remifentanil is widely used to control intraoperative pain. However, its analgesic effect is limited by the generation of postoperative hyperalgesia. In this study, we investigated whether the impairment of transmembrane protein 16C (TMEM16C)/Slack is required for α-amino-3-hydroxy-5-methyl-4-isoxazolepropionic receptor (AMPAR) activation in remifentanil-induced postoperative hyperalgesia. Remifentanil anesthesia reduced the paw withdrawal threshold from 2 h to 48 h postoperatively, with a decrease in the expression of TMEM16C and Slack in the dorsal root ganglia (DRG) and spinal cord. Knockdown of TMEM16C in the DRG reduced the expression of Slack and elevated the basal peripheral sensitivity and AMPAR expression and function. Overexpression of TMEM16C in the DRG impaired remifentanil-induced ERK1/2 phosphorylation and behavioral hyperalgesia. AMPAR-mediated current and neuronal excitability were downregulated by TMEM16C overexpression in the spinal cord. Taken together, these findings suggest that TMEM16C/Slack regulation of excitatory synaptic plasticity via GluA1-containing AMPARs is critical in the pathogenesis of remifentanil-induced postoperative hyperalgesia in rats.
Collapse
|
18
|
Functional Coupling of Slack Channels and P2X3 Receptors Contributes to Neuropathic Pain Processing. Int J Mol Sci 2021; 22:ijms22010405. [PMID: 33401689 PMCID: PMC7795269 DOI: 10.3390/ijms22010405] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2020] [Revised: 12/19/2020] [Accepted: 12/29/2020] [Indexed: 12/16/2022] Open
Abstract
The sodium-activated potassium channel Slack (KNa1.1, Slo2.2, or Kcnt1) is highly expressed in populations of sensory neurons, where it mediates the sodium-activated potassium current (IKNa) and modulates neuronal activity. Previous studies suggest that Slack is involved in the processing of neuropathic pain. However, mechanisms underlying the regulation of Slack activity in this context are poorly understood. Using whole-cell patch-clamp recordings we found that Slack-mediated IKNa in sensory neurons of mice is reduced after peripheral nerve injury, thereby contributing to neuropathic pain hypersensitivity. Interestingly, Slack is closely associated with ATP-sensitive P2X3 receptors in a population of sensory neurons. In vitro experiments revealed that Slack-mediated IKNa may be bidirectionally modulated in response to P2X3 activation. Moreover, mice lacking Slack show altered nocifensive responses to P2X3 stimulation. Our study identifies P2X3/Slack signaling as a mechanism contributing to hypersensitivity after peripheral nerve injury and proposes a potential novel strategy for treatment of neuropathic pain.
Collapse
|
19
|
Spitznagel BD, Mishra NM, Qunies AM, Prael FJ, Du Y, Kozek KA, Lazarenko RM, Denton JS, Emmitte KA, Weaver CD. VU0606170, a Selective Slack Channels Inhibitor, Decreases Calcium Oscillations in Cultured Cortical Neurons. ACS Chem Neurosci 2020; 11:3658-3671. [PMID: 33143429 DOI: 10.1021/acschemneuro.0c00583] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022] Open
Abstract
Malignant migrating partial seizures of infancy is a rare, devastating form of epilepsy most commonly associated with gain-of-function mutations in the potassium channel, Slack. Not only is this condition almost completely pharmacoresistant, there are not even selective drug-like tools available to evaluate whether inhibition of these overactivated, mutant Slack channels may represent a viable path forward toward new antiepileptic therapies. Therefore, we used a high-throughput thallium flux assay to screen a drug-like, 100 000-compound library in search of inhibitors of both wild-type and a disease-associated mutant Slack channel. Using this approach, we discovered VU0606170, a selective Slack channel inhibitor with low micromolar potency. Critically, VU0606170 also proved effective at significantly decreasing the firing rate in overexcited, spontaneously firing cortical neuron cultures. Taken together, our data provide compelling evidence that selective inhibition of Slack channel activity can be achieved with small molecules and that inhibition of Slack channel activity in neurons produces efficacy consistent with an antiepileptic effect. Thus, the identification of VU0606170 provides a much-needed tool for advancing our understanding of the role of the Slack channel in normal physiology and disease as well as its potential as a target for therapeutic intervention.
Collapse
Affiliation(s)
- Brittany D. Spitznagel
- Department of Pharmacology, Vanderbilt University, Nashville, Tennessee 37232, United States
| | - Nigam M. Mishra
- Department of Pharmaceutical Sciences, UNT System College of Pharmacy, University of North Texas Health Science Center, Fort Worth, Texas 76107, United States
| | - Alshaima’a M. Qunies
- Department of Pharmaceutical Sciences, UNT System College of Pharmacy, University of North Texas Health Science Center, Fort Worth, Texas 76107, United States
- Graduate School of Biomedical Sciences, University of North Texas Health Science Center, Fort Worth, Texas 76107, United States
| | - Francis J. Prael
- Department of Pharmacology, Vanderbilt University, Nashville, Tennessee 37232, United States
- Vanderbilt Institute of Chemical Biology, Vanderbilt University, Nashville, Tennessee 37240, United States
| | - Yu Du
- Department of Pharmacology, Vanderbilt University, Nashville, Tennessee 37232, United States
- Vanderbilt Institute of Chemical Biology, Vanderbilt University, Nashville, Tennessee 37240, United States
| | - Krystian A. Kozek
- Department of Pharmacology, Vanderbilt University, Nashville, Tennessee 37232, United States
- Vanderbilt Institute of Chemical Biology, Vanderbilt University, Nashville, Tennessee 37240, United States
- Vanderbilt Medical Scientist Training Program, Vanderbilt University, Nashville, Tennessee 37232, United States
| | - Roman M. Lazarenko
- Department of Anesthesiology, Vanderbilt University, Nashville, Tennessee 37212, United States
| | - Jerod S. Denton
- Department of Pharmacology, Vanderbilt University, Nashville, Tennessee 37232, United States
- Department of Anesthesiology, Vanderbilt University, Nashville, Tennessee 37212, United States
| | - Kyle A. Emmitte
- Department of Pharmaceutical Sciences, UNT System College of Pharmacy, University of North Texas Health Science Center, Fort Worth, Texas 76107, United States
| | - C. David Weaver
- Department of Pharmacology, Vanderbilt University, Nashville, Tennessee 37232, United States
- Vanderbilt Institute of Chemical Biology, Vanderbilt University, Nashville, Tennessee 37240, United States
| |
Collapse
|
20
|
Wang X, Mick G, McCormick K. Pyridine nucleotide regulation of hepatic endoplasmic reticulum calcium uptake. Physiol Rep 2020; 7:e14151. [PMID: 31222964 PMCID: PMC6586769 DOI: 10.14814/phy2.14151] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2019] [Accepted: 05/31/2019] [Indexed: 01/03/2023] Open
Abstract
Pyridine nucleotides serve an array of intracellular metabolic functions such as, to name a few, shuttling electrons in enzymatic reactions, safeguarding the redox state against reactive oxygen species, cytochrome P450 (CYP) enzyme detoxification pathways and, relevant to this study, the regulation of ion fluxes. In particular, the maintenance of a steep calcium gradient between the cytosol and endoplasmic reticulum (ER), without which apoptosis ensues, is achieved by an elaborate combination of energy–requiring ER membrane pumps and efflux channels. In liver microsomes, net calcium uptake was inhibited by physiological concentrations of NADP. In the presence of 1 mmol/L NADP, calcium uptake was attenuated by nearly 80%, additionally, this inhibitory effect was blunted by concomitant addition of NADPH. No other nicotinamide containing compounds ‐save a slight inhibition by NAADP‐hindered calcium uptake; thus, only oxidized pyridine nucleotides, or related compounds with a phosphate moiety, had an imposing effect. Moreover, the NADP inhibition was evident even after selectively blocking ER calcium efflux channels. Given the fundamental role of endoplasmic calcium homeostasis, it is plausible that changes in cytosolic NADP concentration, for example, during anabolic processes, could regulate net ER calcium uptake.
Collapse
Affiliation(s)
- Xudong Wang
- Department of Pediatrics, University of Alabama at Birmingham, Birmingham, Alabama
| | - Gail Mick
- Department of Pediatrics, University of Alabama at Birmingham, Birmingham, Alabama
| | - Kenneth McCormick
- Department of Pediatrics, University of Alabama at Birmingham, Birmingham, Alabama
| |
Collapse
|
21
|
Logan RW, Parekh PK, Kaplan G, Becker-Krail D, Williams W, Yamaguchi S, Yoshino J, Shelton MA, Zhu X, Zhang H, Waplinger S, Fitzgerald E, Oliver-Smith J, Sundarvelu P, Enwright JF, Huang YH, McClung CA. NAD+ cellular redox and SIRT1 regulate the diurnal rhythms of tyrosine hydroxylase and conditioned cocaine reward. Mol Psychiatry 2019; 24:1668-1684. [PMID: 29728703 PMCID: PMC6215755 DOI: 10.1038/s41380-018-0061-1] [Citation(s) in RCA: 39] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/05/2017] [Revised: 01/12/2018] [Accepted: 02/19/2018] [Indexed: 12/21/2022]
Abstract
The diurnal regulation of dopamine is important for normal physiology and diseases such as addiction. Here we find a novel role for the CLOCK protein to antagonize CREB-mediated transcriptional activity at the tyrosine hydroxylase (TH) promoter, which is mediated by the interaction with the metabolic sensing protein, Sirtuin 1 (SIRT1). Additionally, we demonstrate that the transcriptional activity of TH is modulated by the cellular redox state, and daily rhythms of redox balance in the ventral tegmental area (VTA), along with TH transcription, are highly disrupted following chronic cocaine administration. Furthermore, CLOCK and SIRT1 are important for regulating cocaine reward and dopaminergic (DAergic) activity, with interesting differences depending on whether DAergic activity is in a heightened state and if there is a functional CLOCK protein. Taken together, we find that rhythms in cellular metabolism and circadian proteins work together to regulate dopamine synthesis and the reward value for drugs of abuse.
Collapse
Affiliation(s)
- Ryan W. Logan
- Translational Neuroscience Program, Department of Psychiatry, University of Pittsburgh Medical School, Pittsburgh, PA, 15219, USA,Center for Neuroscience, University of Pittsburgh, Pittsburgh, PA, 15260, USA,Center for Systems Neurogenetics of Addiction, The Jackson Laboratory, Bar Harbor, ME, 04609
| | - Puja K. Parekh
- Translational Neuroscience Program, Department of Psychiatry, University of Pittsburgh Medical School, Pittsburgh, PA, 15219, USA,Center for Neuroscience, University of Pittsburgh, Pittsburgh, PA, 15260, USA
| | - Gabrielle Kaplan
- Translational Neuroscience Program, Department of Psychiatry, University of Pittsburgh Medical School, Pittsburgh, PA, 15219, USA,Center for Neuroscience, University of Pittsburgh, Pittsburgh, PA, 15260, USA
| | - Darius Becker-Krail
- Translational Neuroscience Program, Department of Psychiatry, University of Pittsburgh Medical School, Pittsburgh, PA, 15219, USA,Center for Neuroscience, University of Pittsburgh, Pittsburgh, PA, 15260, USA
| | - Wilbur Williams
- Translational Neuroscience Program, Department of Psychiatry, University of Pittsburgh Medical School, Pittsburgh, PA, 15219, USA
| | - Shintaro Yamaguchi
- Center for Systems Neurogenetics of Addiction, The Jackson Laboratory, Bar Harbor, ME, 04609
| | - Jun Yoshino
- Center for Systems Neurogenetics of Addiction, The Jackson Laboratory, Bar Harbor, ME, 04609
| | - Micah A. Shelton
- Translational Neuroscience Program, Department of Psychiatry, University of Pittsburgh Medical School, Pittsburgh, PA, 15219, USA
| | - Xiyu Zhu
- Translational Neuroscience Program, Department of Psychiatry, University of Pittsburgh Medical School, Pittsburgh, PA, 15219, USA,Center for Neuroscience, University of Pittsburgh, Pittsburgh, PA, 15260, USA
| | - Hui Zhang
- Translational Neuroscience Program, Department of Psychiatry, University of Pittsburgh Medical School, Pittsburgh, PA, 15219, USA,School of Medicine, Peking Union Medical College, Tsinghua University, Beijing, China
| | - Spencer Waplinger
- Translational Neuroscience Program, Department of Psychiatry, University of Pittsburgh Medical School, Pittsburgh, PA, 15219, USA
| | - Ethan Fitzgerald
- Translational Neuroscience Program, Department of Psychiatry, University of Pittsburgh Medical School, Pittsburgh, PA, 15219, USA
| | - Jeffrey Oliver-Smith
- Translational Neuroscience Program, Department of Psychiatry, University of Pittsburgh Medical School, Pittsburgh, PA, 15219, USA
| | - Poornima Sundarvelu
- Translational Neuroscience Program, Department of Psychiatry, University of Pittsburgh Medical School, Pittsburgh, PA, 15219, USA
| | - John F. Enwright
- Translational Neuroscience Program, Department of Psychiatry, University of Pittsburgh Medical School, Pittsburgh, PA, 15219, USA
| | | | - Colleen A. McClung
- Translational Neuroscience Program, Department of Psychiatry, University of Pittsburgh Medical School, Pittsburgh, PA, 15219, USA,Center for Neuroscience, University of Pittsburgh, Pittsburgh, PA, 15260, USA,Center for Systems Neurogenetics of Addiction, The Jackson Laboratory, Bar Harbor, ME, 04609,Correspondence: (C.A.M.)
| |
Collapse
|
22
|
An Epilepsy-Associated KCNT1 Mutation Enhances Excitability of Human iPSC-Derived Neurons by Increasing Slack K Na Currents. J Neurosci 2019; 39:7438-7449. [PMID: 31350261 DOI: 10.1523/jneurosci.1628-18.2019] [Citation(s) in RCA: 62] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2018] [Revised: 07/08/2019] [Accepted: 07/17/2019] [Indexed: 12/26/2022] Open
Abstract
Mutations in the KCNT1 (Slack, KNa1.1) sodium-activated potassium channel produce severe epileptic encephalopathies. Expression in heterologous systems has shown that the disease-causing mutations give rise to channels that have increased current amplitude. It is not known, however, whether such gain of function occurs in human neurons, nor whether such increased KNa current is expected to suppress or increase the excitability of cortical neurons. Using genetically engineered human induced pluripotent stem cell (iPSC)-derived neurons, we have now found that sodium-dependent potassium currents are increased several-fold in neurons bearing a homozygous P924L mutation. In current-clamp recordings, the increased KNa current in neurons with the P924L mutation acts to shorten the duration of action potentials and to increase the amplitude of the afterhyperpolarization that follows each action potential. Strikingly, the number of action potentials that were evoked by depolarizing currents as well as maximal firing rates were increased in neurons expressing the mutant channel. In networks of spontaneously active neurons, the mean firing rate, the occurrence of rapid bursts of action potentials, and the intensity of firing during the burst were all increased in neurons with the P924L Slack mutation. The feasibility of an increased KNa current to increase firing rates independent of any compensatory changes was validated by numerical simulations. Our findings indicate that gain-of-function in Slack KNa channels causes hyperexcitability in both isolated neurons and in neural networks and occurs by a cell-autonomous mechanism that does not require network interactions.SIGNIFICANCE STATEMENT KCNT1 mutations lead to severe epileptic encephalopathies for which there are no effective treatments. This study is the first demonstration that a KCNT1 mutation increases the Slack current in neurons. It also provides the first explanation for how this increased potassium current induces hyperexcitability, which could be the underlining factor causing seizures.
Collapse
|
23
|
Fitzgerald MP, Fiannacca M, Smith DM, Gertler TS, Gunning B, Syrbe S, Verbeek N, Stamberger H, Weckhuysen S, Ceulemans B, Schoonjans AS, Rossi M, Demarquay G, Lesca G, Olofsson K, Koolen DA, Hornemann F, Baulac S, Rubboli G, Minks KQ, Lee B, Helbig I, Dlugos D, Møller RS, Bearden D. Treatment Responsiveness in KCNT1-Related Epilepsy. Neurotherapeutics 2019; 16:848-857. [PMID: 31054119 PMCID: PMC6694367 DOI: 10.1007/s13311-019-00739-y] [Citation(s) in RCA: 62] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/26/2022] Open
Abstract
Pathogenic variants in KCNT1 represent an important cause of treatment-resistant epilepsy, for which an effective therapy has been elusive. Reports about the effectiveness of quinidine, a candidate precision therapy, have been mixed. We sought to evaluate the treatment responsiveness of patients with KCNT1-related epilepsy. We performed an observational study of 43 patients using a collaborative KCNT1 patient registry. We assessed treatment efficacy based upon clinical seizure reduction, side effects of quinidine therapy, and variant-specific responsiveness to treatment. Quinidine treatment resulted in a > 50% seizure reduction in 20% of patients, with rare patients achieving transient seizure freedom. Multiple other therapies demonstrated some success in reducing seizure frequency, including the ketogenic diet and vigabatrin, the latter particularly in patients with epileptic spasms. Patients with the best quinidine response had variants that clustered distal to the NADP domain within the RCK2 domain of the protein. Half of patients did not receive a quinidine trial. In those who did, nearly half did not achieve therapeutic blood levels. More favorable response to quinidine in patients with KCNT1 variants distal to the NADP domain within the RCK2 domain may suggest a variant-specific response.
Collapse
Affiliation(s)
- Mark P Fitzgerald
- Division of Neurology, Departments of Neurology and Pediatrics, The Children's Hospital of Philadelphia and the Perelman School of Medicine at the University of Pennsylvania, 3501 Civic Center Blvd, Philadelphia, PA, 19104, USA.
| | | | | | - Tracy S Gertler
- Division of Neurology, Department of Pediatrics, Ann and Robert H. Lurie Children's Hospital of Chicago, Chicago, IL, USA
| | - Boudewijn Gunning
- Stichting Epilepsie Instellingen Nederland, Zwolle, 8025 BV, Netherlands
| | - Steffen Syrbe
- Division of Child Neurology and Inherited Metabolic Diseases, Department of General Paediatrics, Centre for Paediatrics and Adolescent Medicine, University Hospital Heidelberg, Im Neuenheimer Feld 430, 69120, Heidelberg, Germany
| | - Nienke Verbeek
- Department of Genetics, University Medical Center Utrecht, Utrecht, 3584 CX, The Netherlands
| | - Hannah Stamberger
- Neurogenetics group, Center for Molecular Neurology, Vlaams Instituut voor Biotechnologie, and Institute Born Bunge, University of Antwerp, Antwerp, 2000, Belgium
- Department of Neurology, Antwerp University Hospital, Antwerp, 2650, Belgium
| | - Sarah Weckhuysen
- Neurogenetics group, Center for Molecular Neurology, Vlaams Instituut voor Biotechnologie, and Institute Born Bunge, University of Antwerp, Antwerp, 2000, Belgium
- Department of Neurology, Antwerp University Hospital, Antwerp, 2650, Belgium
| | - Berten Ceulemans
- Department of Paediatric Neurology, Antwerp University Hospital, University of Antwerp, Antwerp, 2650, Belgium
| | - An-Sofie Schoonjans
- Neurogenetics Research Group, Vrije Universiteit Brussel (VUB), Universitair Ziekenhuis Brussel (UZ Brussel), Laarbeeklaan 101, 1090, Brussels, Belgium
| | - Massimiliano Rossi
- Genetics department, Hospices Civils de Lyon, and Institut National de la Santé et de la Recherche Médicale U1028, Centre national de la recherche scientifique Unité Mixte de Recherche 5292, Lyon Neuroscience Research Center, GENDEV Team, Claude Bernard Lyon 1 University, Bron, 69500, France
| | - Geneviève Demarquay
- Department of Functional Neurology and Epileptology, Hospices Civils de Lyon and Centre national de la recherche scientifique, Unité Mixte de Recherche 5292, Lyon Neuroscience Research Center, Auditory Cognition and Psychoacoustics Team, Lyon, 69003, France
| | - Gaetan Lesca
- Genetics department, Hospices Civils de Lyon, and Institut National de la Santé et de la Recherche Médicale U1028, Centre national de la recherche scientifique Unité Mixte de Recherche 5292, Lyon Neuroscience Research Center, GENDEV Team, Claude Bernard Lyon 1 University, Bron, 69500, France
| | - Kern Olofsson
- Danish Epilepsy Centre, Filadelfia, Dianalund,, DK 4293, Denmark
| | - D A Koolen
- Department of Human Genetics, Radboud University Medical Center, Nijmegen, 6525 GA, The Netherlands
| | - Frauke Hornemann
- Centre of Pediatric Research, Hospital for Children and Adolescents, 04103, Leipzig, Germany
| | - Stephanie Baulac
- Sorbonne Université, UPMC Univ Paris 06, Unité Mixte de Recherche S 1127, F-75013, Paris, France
- Institut National de la Santé et de la Recherche Médicale, U1127, F-75013, Paris, France
- Centre national de la recherche scientifique, Unité Mixte de Recherche 7225, F-75013, Paris, France
- Institut du Cerveau et de la Moelle épinière (ICM), Hôpital Pitié-Salpêtrière, F-75013, Paris, France
- Department of Genetics, Assistance Publique des Hôpitaux de Paris (AP-HP), Hôpital Pitié-Salpêtrière, F-75013, Paris, France
| | - Guido Rubboli
- Danish Epilepsy Centre, Filadelfia, Dianalund,, DK 4293, Denmark
- University of Copenhagen, Copenhagen, 1165, Denmark
| | - Kelly Q Minks
- Division of Child Neurology, Department of Neurology, University of Rochester School of Medicine, Rochester, NY, USA
| | - Bohoon Lee
- Division of Child Neurology, Department of Neurology, University of Rochester School of Medicine, Rochester, NY, USA
| | - Ingo Helbig
- Division of Neurology, Departments of Neurology and Pediatrics, The Children's Hospital of Philadelphia and the Perelman School of Medicine at the University of Pennsylvania, 3501 Civic Center Blvd, Philadelphia, PA, 19104, USA
| | - Dennis Dlugos
- Division of Neurology, Departments of Neurology and Pediatrics, The Children's Hospital of Philadelphia and the Perelman School of Medicine at the University of Pennsylvania, 3501 Civic Center Blvd, Philadelphia, PA, 19104, USA
| | - Rikke S Møller
- Danish Epilepsy Centre, Filadelfia, Dianalund,, DK 4293, Denmark
- Institute for Regional Health Research, University of Southern Denmark, Odense, 5230, Denmark
| | - David Bearden
- Division of Child Neurology, Department of Neurology, University of Rochester School of Medicine, Rochester, NY, USA
| |
Collapse
|
24
|
Sodium-activated potassium channels shape peripheral auditory function and activity of the primary auditory neurons in mice. Sci Rep 2019; 9:2573. [PMID: 30796290 PMCID: PMC6384918 DOI: 10.1038/s41598-019-39119-z] [Citation(s) in RCA: 30] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2018] [Accepted: 01/17/2019] [Indexed: 11/08/2022] Open
Abstract
Potassium (K+) channels shape the response properties of neurons. Although enormous progress has been made to characterize K+ channels in the primary auditory neurons, the molecular identities of many of these channels and their contributions to hearing in vivo remain unknown. Using a combination of RNA sequencing and single molecule fluorescent in situ hybridization, we localized expression of transcripts encoding the sodium-activated potassium channels KNa1.1 (SLO2.2/Slack) and KNa1.2 (SLO2.1/Slick) to the primary auditory neurons (spiral ganglion neurons, SGNs). To examine the contribution of these channels to function of the SGNs in vivo, we measured auditory brainstem responses in KNa1.1/1.2 double knockout (DKO) mice. Although auditory brainstem response (wave I) thresholds were not altered, the amplitudes of suprathreshold responses were reduced in DKO mice. This reduction in amplitude occurred despite normal numbers and molecular architecture of the SGNs and their synapses with the inner hair cells. Patch clamp electrophysiology of SGNs isolated from DKO mice displayed altered membrane properties, including reduced action potential thresholds and amplitudes. These findings show that KNa1 channel activity is essential for normal cochlear function and suggest that early forms of hearing loss may result from physiological changes in the activity of the primary auditory neurons.
Collapse
|
25
|
Dilena R, DiFrancesco JC, Soldovieri MV, Giacobbe A, Ambrosino P, Mosca I, Galli MA, Guez S, Fumagalli M, Miceli F, Cattaneo D, Darra F, Gennaro E, Zara F, Striano P, Castellotti B, Gellera C, Varesio C, Veggiotti P, Taglialatela M. Early Treatment with Quinidine in 2 Patients with Epilepsy of Infancy with Migrating Focal Seizures (EIMFS) Due to Gain-of-Function KCNT1 Mutations: Functional Studies, Clinical Responses, and Critical Issues for Personalized Therapy. Neurotherapeutics 2018; 15:1112-1126. [PMID: 30112700 PMCID: PMC6277296 DOI: 10.1007/s13311-018-0657-9] [Citation(s) in RCA: 57] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/28/2022] Open
Abstract
Epilepsy of infancy with migrating focal seizures (EIMFS) is a rare early-onset developmental epileptic encephalopathy resistant to anti-epileptic drugs. The most common cause for EIMFS is a gain-of-function mutation in the KCNT1 potassium channel gene, and treatment with the KCNT1 blocker quinidine has been suggested as a rational approach for seizure control in EIMFS patients. However, variable results on the clinical efficacy of quinidine have been reported. In the present study, we provide a detailed description of the clinical, genetic, in vitro, and in vivo electrophysiological profile and pharmacological responses to quinidine of 2 EIMFS unrelated patients with a heterozygous de novo KCNT1 mutation: c.2849G>A (p.R950Q) in patient 1 and c.2677G>A (p.E893K) in patient 2. When expressed heterologously in CHO cells, KCNT1 channels carrying each variant showed gain-of-function effects, and were more effectively blocked by quinidine when compared to wild-type KCNT1 channels. On the basis of these in vitro results, add-on quinidine treatment was started at 3 and 16 months of age in patients 1 and 2, respectively. The results obtained reveal that quinidine significantly reduced seizure burden (by about 90%) and improved quality of life in both patients, but failed to normalize developmental milestones, which persisted as severely delayed. Based on the present experience, early quinidine intervention associated with heart monitoring and control of blood levels is among the critical factors for therapy effectiveness in EIMFS patients with KCNT1 gain-of-function mutations. Multicenter studies are needed to establish a consensus protocol for patient recruitment, quinidine treatment modalities, and outcome evaluation, to optimize clinical efficacy and reduce risks as well as variability associated to quinidine use in such severe developmental encephalopathy.
Collapse
Affiliation(s)
- Robertino Dilena
- Pediatric Epileptology and Neurophysiology (RD), Infantile Neuropsichiatry (AG), Cardiology (MAG), High Intensity Pediatric Care (SG), Neonatology (MF), Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, 20122, Milan, Italy
| | - Jacopo C DiFrancesco
- Clinical Neurophysiology and Epilepsy Center, Fondazione IRCCS Istituto Neurologico Carlo Besta, 20133, Milan, Italy
- Department of Neurology, San Gerardo Hospital, School of Medicine and Surgery, Milan Center for Neuroscience (NeuroMi), University of Milano-Bicocca, 20900, Monza, Italy
| | | | - Antonella Giacobbe
- Pediatric Epileptology and Neurophysiology (RD), Infantile Neuropsichiatry (AG), Cardiology (MAG), High Intensity Pediatric Care (SG), Neonatology (MF), Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, 20122, Milan, Italy
| | - Paolo Ambrosino
- Department of Medicine and Health Science, University of Molise, 86100, Campobasso, Italy
| | - Ilaria Mosca
- Department of Medicine and Health Science, University of Molise, 86100, Campobasso, Italy
| | - Maria Albina Galli
- Pediatric Epileptology and Neurophysiology (RD), Infantile Neuropsichiatry (AG), Cardiology (MAG), High Intensity Pediatric Care (SG), Neonatology (MF), Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, 20122, Milan, Italy
| | - Sophie Guez
- Pediatric Epileptology and Neurophysiology (RD), Infantile Neuropsichiatry (AG), Cardiology (MAG), High Intensity Pediatric Care (SG), Neonatology (MF), Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, 20122, Milan, Italy
| | - Monica Fumagalli
- Pediatric Epileptology and Neurophysiology (RD), Infantile Neuropsichiatry (AG), Cardiology (MAG), High Intensity Pediatric Care (SG), Neonatology (MF), Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, 20122, Milan, Italy
| | - Francesco Miceli
- Division of Pharmacology, Department of Neuroscience, University of Naples "Federico II", 80131, Naples, Italy
| | - Dario Cattaneo
- Unit of Clinical Pharmacology, ASST Fatebenefratelli Sacco, 20157, Milan, Italy
| | - Francesca Darra
- Department of Surgical, Odontostomatological, and Maternal-Infantile Sciences, University of Verona, 37134, Verona, Italy
| | - Elena Gennaro
- Laboratory of Genetics, E.O. Ospedali Galliera, 16128, Genoa, Italy
| | - Federico Zara
- Laboratory of Genetics, E.O. Ospedali Galliera, 16128, Genoa, Italy
| | - Pasquale Striano
- Pediatric Neurology and Muscular Diseases Unit, Department of Neurosciences, Rehabilitation, Ophthalmology, Genetics, Maternal and Child Health, University of Genoa, "G. Gaslini" Institute, 16147, Genoa, Italy
| | - Barbara Castellotti
- Unit of Genetics of Neurodegenerative and Metabolic Diseases, Fondazione IRCCS Istituto Neurologico Carlo Besta, 20133, Milan, Italy
| | - Cinzia Gellera
- Unit of Genetics of Neurodegenerative and Metabolic Diseases, Fondazione IRCCS Istituto Neurologico Carlo Besta, 20133, Milan, Italy
| | - Costanza Varesio
- Department of Child Neurology and Psychiatry, "C. Mondino" National Neurological Institute, 27100, Pavia, Italy
| | - Pierangelo Veggiotti
- Department of Biomedical and Clinical Sciences, Children's Hospital Vittore Buzzi, University of Milan, and Pediatric Neurology, 20154, Milan, Italy
| | - Maurizio Taglialatela
- Department of Medicine and Health Science, University of Molise, 86100, Campobasso, Italy.
- Division of Pharmacology, Department of Neuroscience, University of Naples "Federico II", 80131, Naples, Italy.
- Department of Neuroscience, University of Naples "Federico II", Via Pansini 5, 80131, Naples, Italy.
| |
Collapse
|
26
|
Kumar J, Solaiman A, Mahakkanukrauh P, Mohamed R, Das S. Sleep Related Epilepsy and Pharmacotherapy: An Insight. Front Pharmacol 2018; 9:1088. [PMID: 30319421 PMCID: PMC6171479 DOI: 10.3389/fphar.2018.01088] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2018] [Accepted: 09/07/2018] [Indexed: 01/26/2023] Open
Abstract
In the last several decades, sleep-related epilepsy has drawn considerable attention among epileptologists and neuroscientists in the interest of new paradigms of the disease etiology, pathogenesis and management. Sleep-related epilepsy is nocturnal seizures that manifest solely during the sleep state. Sleep comprises two distinct stages i.e., non-rapid eye movement (NREM) and rapid eye movement (REM) that alternate every 90 min with NREM preceding REM. Current findings indicate that the sleep-related epilepsy manifests predominantly during the synchronized stages of sleep; NREM over REM stage. Sleep related hypermotor epilepsy (SHE), benign partial epilepsy with centrotemporal spikes or benign rolandic epilepsy (BECTS), and Panayiotopoulos Syndrome (PS) are three of the most frequently implicated epilepsies occurring during the sleep state. Although some familial types are described, others are seemingly sporadic occurrences. In the present review, we aim to discuss the predominance of sleep-related epilepsy during NREM, established familial links to the pathogenesis of SHE, BECTS and PS, and highlight the present available pharmacotherapy options.
Collapse
Affiliation(s)
- Jaya Kumar
- Department of Physiology, Faculty of Medicine, Universiti Kebangsaan Malaysia Medical Centre, Kuala Lumpur, Malaysia
| | - Amro Solaiman
- Department of Anatomy, Universiti Kebangsaan Malaysia Medical Centre, Kuala Lumpur, Malaysia
| | - Pasuk Mahakkanukrauh
- Department of Anatomy, Faculty of Medicine, Chiang Mai University, Chiang Mai, Thailand
- Excellence Centre in Forensic Osteology Research Center, Faculty of Medicine, Chiang Mai University, Chiang Mai, Thailand
| | - Rashidi Mohamed
- Department of Familty Medicine, Universiti Kebangsaan Malaysia Medical Centre, Kuala Lumpur, Malaysia
| | - Srijit Das
- Department of Anatomy, Universiti Kebangsaan Malaysia Medical Centre, Kuala Lumpur, Malaysia
| |
Collapse
|
27
|
Evely KM, Pryce KD, Bausch AE, Lukowski R, Ruth P, Haj-Dahmane S, Bhattacharjee A. Slack K Na Channels Influence Dorsal Horn Synapses and Nociceptive Behavior. Mol Pain 2018; 13:1744806917714342. [PMID: 28604221 PMCID: PMC5486487 DOI: 10.1177/1744806917714342] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
The sodium-activated potassium channel Slack (Kcnt1, Slo2.2) is highly expressed in dorsal root ganglion neurons where it regulates neuronal firing. Several studies have implicated the Slack channel in pain processing, but the precise mechanism or the levels within the sensory pathway where channels are involved remain unclear. Here, we furthered the behavioral characterization of Slack channel knockout mice and for the first time examined the role of Slack channels in the superficial, pain-processing lamina of the dorsal horn. We performed whole-cell recordings from spinal cord slices to examine the intrinsic and synaptic properties of putative inhibitory and excitatory lamina II interneurons. Slack channel deletion altered intrinsic properties and synaptic drive to favor an overall enhanced excitatory tone. We measured the amplitudes and paired pulse ratio of paired excitatory post-synaptic currents at primary afferent synapses evoked by electrical stimulation of the dorsal root entry zone. We found a substantial decrease in the paired pulse ratio at synapses in Slack deleted neurons compared to wildtype, indicating increased presynaptic release from primary afferents. Corroborating these data, plantar test showed Slack knockout mice have an enhanced nociceptive responsiveness to localized thermal stimuli compared to wildtype mice. Our findings suggest that Slack channels regulate synaptic transmission within the spinal cord dorsal horn and by doing so establishes the threshold for thermal nociception.
Collapse
Affiliation(s)
- Katherine M Evely
- Program in Neuroscience, University at Buffalo, The State University of New York, NY, USA
| | - Kerri D Pryce
- Department of Pharmacology and Toxicology, University at Buffalo, The State University of New York, NY, USA
| | - Anne E Bausch
- Institut für Pharmazie, Pharmakologie, Toxikologie und Klinische Pharmazie der Universität Tübingen, Germany
| | - Robert Lukowski
- Institut für Pharmazie, Pharmakologie, Toxikologie und Klinische Pharmazie der Universität Tübingen, Germany
| | - Peter Ruth
- Institut für Pharmazie, Pharmakologie, Toxikologie und Klinische Pharmazie der Universität Tübingen, Germany
| | - Samir Haj-Dahmane
- Program in Neuroscience, University at Buffalo, The State University of New York, NY, USA
| | | |
Collapse
|
28
|
Gao L, Ortega-Sáenz P, López-Barneo J. Acute oxygen sensing-Role of metabolic specifications in peripheral chemoreceptor cells. Respir Physiol Neurobiol 2018; 265:100-111. [PMID: 30172779 DOI: 10.1016/j.resp.2018.08.007] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2018] [Revised: 08/17/2018] [Accepted: 08/29/2018] [Indexed: 12/30/2022]
Abstract
Acute oxygen sensing is essential for humans under hypoxic environments or pathologic conditions. This is achieved by the carotid body (CB), the key arterial chemoreceptor, along with other peripheral chemoreceptor organs, such as the adrenal medulla (AM). Although it is widely accepted that inhibition of K+ channels in the plasma membrane of CB cells during acute hypoxia results in the activation of cardiorespiratory reflexes, the molecular mechanisms by which the hypoxic signal is detected to modulate ion channel activity are not fully understood. Using conditional knockout mice lacking mitochondrial complex I (MCI) subunit NDUFS2, we have found that MCI generates reactive oxygen species and pyridine nucleotides, which signal K+ channels during acute hypoxia. Comparing the transcriptomes from CB and AM, which are O2-sensitive, with superior cervical ganglion, which is practically O2-insensitive, we have found that CB and AM contain unique metabolic gene expression profiles. The "signature metabolic profile" and their biophysical characteristics could be essential for acute O2 sensing by chemoreceptor cells.
Collapse
Affiliation(s)
- Lin Gao
- Instituto de Biomedicina de Sevilla (IBiS), Hospital Universitario Virgen del Rocío/CSIC/Universidad de Sevilla, Spain; Departamento de Fisiología Médica y Biofísica, Facultad de Medicina, Universidad de Sevilla, Spain; Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), Madrid, Spain.
| | - Patricia Ortega-Sáenz
- Instituto de Biomedicina de Sevilla (IBiS), Hospital Universitario Virgen del Rocío/CSIC/Universidad de Sevilla, Spain; Departamento de Fisiología Médica y Biofísica, Facultad de Medicina, Universidad de Sevilla, Spain; Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), Madrid, Spain
| | - José López-Barneo
- Instituto de Biomedicina de Sevilla (IBiS), Hospital Universitario Virgen del Rocío/CSIC/Universidad de Sevilla, Spain; Departamento de Fisiología Médica y Biofísica, Facultad de Medicina, Universidad de Sevilla, Spain; Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), Madrid, Spain.
| |
Collapse
|
29
|
Bausch AE, Ehinger R, Straubinger J, Zerfass P, Nann Y, Lukowski R. Loss of Sodium-Activated Potassium Channel Slack and FMRP Differentially Affect Social Behavior in Mice. Neuroscience 2018; 384:361-374. [DOI: 10.1016/j.neuroscience.2018.05.040] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2017] [Revised: 05/22/2018] [Accepted: 05/24/2018] [Indexed: 12/31/2022]
|
30
|
Abstract
Exome and targeted sequencing have revolutionized clinical diagnosis. This has been particularly striking in epilepsy and neurodevelopmental disorders, for which new genes or new variants of preexisting candidate genes are being continuously identified at increasing rates every year. A surprising finding of these efforts is the recognition that gain of function potassium channel variants are actually associated with certain types of epilepsy, such as malignant migrating partial seizures of infancy or early-onset epileptic encephalopathy. This development has been difficult to understand as traditionally potassium channel loss-of-function, not gain-of-function, has been associated with hyperexcitability disorders. In this article, we describe the current state of the field regarding the gain-of-function potassium channel variants associated with epilepsy (KCNA2, KCNB1, KCND2, KCNH1, KCNH5, KCNJ10, KCNMA1, KCNQ2, KCNQ3, and KCNT1) and speculate on the possible cellular mechanisms behind the development of seizures and epilepsy in these patients. Understanding how potassium channel gain-of-function leads to epilepsy will provide new insights into the inner working of neural circuits and aid in developing new therapies.
Collapse
Affiliation(s)
- Zachary Niday
- Dept. of Physiology and Neurobiology, University of Connecticut, Storrs, CT 06269, USA
| | | |
Collapse
|
31
|
Gururaj S, Palmer EE, Sheehan GD, Kandula T, Macintosh R, Ying K, Morris P, Tao J, Dias KR, Zhu Y, Dinger ME, Cowley MJ, Kirk EP, Roscioli T, Sachdev R, Duffey ME, Bye A, Bhattacharjee A. A De Novo Mutation in the Sodium-Activated Potassium Channel KCNT2 Alters Ion Selectivity and Causes Epileptic Encephalopathy. Cell Rep 2018; 21:926-933. [PMID: 29069600 DOI: 10.1016/j.celrep.2017.09.088] [Citation(s) in RCA: 36] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2016] [Revised: 06/12/2017] [Accepted: 09/26/2017] [Indexed: 12/31/2022] Open
Abstract
Early infantile epileptic encephalopathies (EOEE) are a debilitating spectrum of disorders associated with cognitive impairments. We present a clinical report of a KCNT2 mutation in an EOEE patient. The de novo heterozygous variant Phe240Leu SLICK was identified by exome sequencing and confirmed by Sanger sequencing. Phe240Leu rSlick and hSLICK channels were electrophysiologically, heterologously characterized to reveal three significant alterations to channel function. First, [Cl-]i sensitivity was reversed in Phe240Leu channels. Second, predominantly K+-selective WT channels were made to favor Na+ over K+ by Phe240Leu. Third, and consequent to altered ion selectivity, Phe240Leu channels had larger inward conductance. Further, rSlick channels induced membrane hyperexcitability when expressed in primary neurons, resembling the cellular seizure phenotype. Taken together, our results confirm that Phe240Leu is a "change-of-function" KCNT2 mutation, demonstrating unusual altered selectivity in KNa channels. These findings establish pathogenicity of the Phe240Leu KCNT2 mutation in the reported EOEE patient.
Collapse
Affiliation(s)
- Sushmitha Gururaj
- Pharmacology and Toxicology, University at Buffalo - The State University of New York, Buffalo, NY 14214, USA
| | - Elizabeth Emma Palmer
- Sydney Children's Hospital, Randwick, NSW 2031, Australia; University of New South Wales, Sydney, NSW 2031, Australia; Genetics of Learning Disability Service, Waratah, NSW 2298, Australia
| | - Garrett D Sheehan
- Pharmacology and Toxicology, University at Buffalo - The State University of New York, Buffalo, NY 14214, USA
| | - Tejaswi Kandula
- Sydney Children's Hospital, Randwick, NSW 2031, Australia; University of New South Wales, Sydney, NSW 2031, Australia
| | | | - Kevin Ying
- Kinghorn Centre for Clinical Genomics, Garvan Institute of Medical Research, Sydney, NSW 2298, Australia
| | - Paula Morris
- Kinghorn Centre for Clinical Genomics, Garvan Institute of Medical Research, Sydney, NSW 2298, Australia
| | - Jiang Tao
- Kinghorn Centre for Clinical Genomics, Garvan Institute of Medical Research, Sydney, NSW 2298, Australia
| | - Kerith-Rae Dias
- Kinghorn Centre for Clinical Genomics, Garvan Institute of Medical Research, Sydney, NSW 2298, Australia
| | - Ying Zhu
- Genetics of Learning Disability Service, Waratah, NSW 2298, Australia; SEALS Pathology, Randwick, NSW 2031, Australia
| | - Marcel E Dinger
- University of New South Wales, Sydney, NSW 2031, Australia; Kinghorn Centre for Clinical Genomics, Garvan Institute of Medical Research, Sydney, NSW 2298, Australia
| | - Mark J Cowley
- University of New South Wales, Sydney, NSW 2031, Australia; Kinghorn Centre for Clinical Genomics, Garvan Institute of Medical Research, Sydney, NSW 2298, Australia
| | - Edwin P Kirk
- Sydney Children's Hospital, Randwick, NSW 2031, Australia; University of New South Wales, Sydney, NSW 2031, Australia; SEALS Pathology, Randwick, NSW 2031, Australia
| | - Tony Roscioli
- Sydney Children's Hospital, Randwick, NSW 2031, Australia; University of New South Wales, Sydney, NSW 2031, Australia; SEALS Pathology, Randwick, NSW 2031, Australia
| | - Rani Sachdev
- Sydney Children's Hospital, Randwick, NSW 2031, Australia; University of New South Wales, Sydney, NSW 2031, Australia
| | - Michael E Duffey
- Physiology and Biophysics, University at Buffalo - The State University of New York, Buffalo, NY 14214, USA
| | - Ann Bye
- Sydney Children's Hospital, Randwick, NSW 2031, Australia; University of New South Wales, Sydney, NSW 2031, Australia
| | - Arin Bhattacharjee
- Pharmacology and Toxicology, University at Buffalo - The State University of New York, Buffalo, NY 14214, USA; Program for Neuroscience, University at Buffalo - The State University of New York, Buffalo, NY 14214, USA.
| |
Collapse
|
32
|
Gururaj S, Evely KM, Pryce KD, Li J, Qu J, Bhattacharjee A. Protein kinase A-induced internalization of Slack channels from the neuronal membrane occurs by adaptor protein-2/clathrin-mediated endocytosis. J Biol Chem 2017; 292:19304-19314. [PMID: 28982974 DOI: 10.1074/jbc.m117.804716] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2017] [Revised: 09/13/2017] [Indexed: 02/01/2023] Open
Abstract
The sodium-activated potassium (KNa) channel Kcnt1 (Slack) is abundantly expressed in nociceptor (pain-sensing) neurons of the dorsal root ganglion (DRG), where they transmit the large outward conductance IKNa and arbitrate membrane excitability. Slack channel expression at the DRG membrane is necessary for their characteristic firing accommodation during maintained stimulation, and reduced membrane channel density causes hyperexcitability. We have previously shown that in a pro-inflammatory state, a decrease in membrane channel expression leading to reduced Slack-mediated IKNa expression underlies DRG neuronal sensitization. An important component of the inflammatory milieu, PKA internalizes Slack channels from the DRG membrane, reduces IKNa, and produces DRG neuronal hyperexcitability when activated in cultured primary DRG neurons. Here, we show that this PKA-induced retrograde trafficking of Slack channels also occurs in intact spinal cord slices and that it is carried out by adaptor protein-2 (AP-2) via clathrin-mediated endocytosis. We provide mass spectrometric and biochemical evidence of an association of native neuronal AP-2 adaptor proteins with Slack channels, facilitated by a dileucine motif housed in the cytoplasmic Slack C terminus that binds AP-2. By creating a competitive peptide blocker of AP-2-Slack binding, we demonstrated that this interaction is essential for clathrin recruitment to the DRG membrane, Slack channel endocytosis, and DRG neuronal hyperexcitability after PKA activation. Together, these findings uncover AP-2 and clathrin as players in Slack channel regulation. Given the significant role of Slack in nociceptive neuronal excitability, the AP-2 clathrin-mediated endocytosis trafficking mechanism may enable targeting of peripheral and possibly, central neuronal sensitization.
Collapse
Affiliation(s)
| | - Katherine M Evely
- the Program for Neuroscience, University at Buffalo, State University of New York, Buffalo, New York 14214 and
| | - Kerri D Pryce
- From the Department of Pharmacology and Toxicology and
| | - Jun Li
- the New York State Center of Excellence in Bioinformatics and Life Sciences, Buffalo, New York 14203
| | - Jun Qu
- the New York State Center of Excellence in Bioinformatics and Life Sciences, Buffalo, New York 14203
| | - Arin Bhattacharjee
- From the Department of Pharmacology and Toxicology and .,the Program for Neuroscience, University at Buffalo, State University of New York, Buffalo, New York 14214 and
| |
Collapse
|
33
|
Tomasello DL, Hurley E, Wrabetz L, Bhattacharjee A. Slick (Kcnt2) Sodium-Activated Potassium Channels Limit Peptidergic Nociceptor Excitability and Hyperalgesia. J Exp Neurosci 2017; 11:1179069517726996. [PMID: 28943756 PMCID: PMC5602212 DOI: 10.1177/1179069517726996] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2017] [Accepted: 07/25/2017] [Indexed: 01/21/2023] Open
Abstract
The Slick (Kcnt2) sodium-activated potassium (KNa) channel is a rapidly gating and weakly voltage-dependent and sodium-dependent potassium channel with no clearly defined physiological function. Within the dorsal root ganglia (DRGs), we show Slick channels are exclusively expressed in small-sized and medium-sized calcitonin gene–related peptide (CGRP)-containing DRG neurons, and a pool of channels are localized to large dense-core vesicles (LDCV)-containing CGRP. We stimulated DRG neurons for CGRP release and found Slick channels contained within CGRP-positive LDCV translocated to the neuronal membrane. Behavioral studies in Slick knockout (KO) mice indicated increased basal heat detection and exacerbated thermal hyperalgesia compared with wild-type littermate controls during neuropathic and chronic inflammatory pain. Electrophysiologic recordings of DRG neurons from Slick KO mice revealed that Slick channels contribute to outward current, propensity to fire action potentials (APs), and to AP properties. Our data suggest that Slick channels restrain the excitability of CGRP-containing neurons, diminishing pain behavior after inflammation and injury.
Collapse
Affiliation(s)
- Danielle L Tomasello
- Neuroscience Program, The State University of New York - University at Buffalo, Buffalo, NY, USA
| | - Edward Hurley
- Department of Neurology, Jacobs School of Medicine and Biomedical Sciences, The State University of New York - University at Buffalo, Buffalo, NY, USA
| | - Lawrence Wrabetz
- Department of Neurology, Jacobs School of Medicine and Biomedical Sciences, The State University of New York - University at Buffalo, Buffalo, NY, USA
| | - Arin Bhattacharjee
- Neuroscience Program, The State University of New York - University at Buffalo, Buffalo, NY, USA.,Department of Pharmacology and Toxicology, Jacobs School of Medicine and Biomedical Sciences, The State University of New York - University at Buffalo, Buffalo, NY, USA
| |
Collapse
|
34
|
The Slo(w) path to identifying the mitochondrial channels responsible for ischemic protection. Biochem J 2017; 474:2067-2094. [PMID: 28600454 DOI: 10.1042/bcj20160623] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2017] [Revised: 02/10/2017] [Accepted: 02/13/2017] [Indexed: 12/19/2022]
Abstract
Mitochondria play an important role in tissue ischemia and reperfusion (IR) injury, with energetic failure and the opening of the mitochondrial permeability transition pore being the major causes of IR-induced cell death. Thus, mitochondria are an appropriate focus for strategies to protect against IR injury. Two widely studied paradigms of IR protection, particularly in the field of cardiac IR, are ischemic preconditioning (IPC) and volatile anesthetic preconditioning (APC). While the molecular mechanisms recruited by these protective paradigms are not fully elucidated, a commonality is the involvement of mitochondrial K+ channel opening. In the case of IPC, research has focused on a mitochondrial ATP-sensitive K+ channel (mitoKATP), but, despite recent progress, the molecular identity of this channel remains a subject of contention. In the case of APC, early research suggested the existence of a mitochondrial large-conductance K+ (BK, big conductance of potassium) channel encoded by the Kcnma1 gene, although more recent work has shown that the channel that underlies APC is in fact encoded by Kcnt2 In this review, we discuss both the pharmacologic and genetic evidence for the existence and identity of mitochondrial K+ channels, and the role of these channels both in IR protection and in regulating normal mitochondrial function.
Collapse
|
35
|
Liang M, Yin XL, Wang LY, Yin WH, Song NY, Shi HB, Li CY, Yin SK. NAD+ Attenuates Bilirubin-Induced Hyperexcitation in the Ventral Cochlear Nucleus by Inhibiting Excitatory Neurotransmission and Neuronal Excitability. Front Cell Neurosci 2017; 11:21. [PMID: 28217084 PMCID: PMC5289950 DOI: 10.3389/fncel.2017.00021] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2016] [Accepted: 01/23/2017] [Indexed: 02/05/2023] Open
Abstract
Nicotinamide adenine dinucleotide (NAD+) is an important molecule with extensive biological functions in various cellular processes, including protection against cell injuries. However, little is known regarding the roles of NAD+ in neuronal excitation and excitotoxicity associated with many neurodegenerative disorders and diseases. Using patch-clamp recordings, we studied its potential effects on principal neurons in the ventral cochlear nucleus (VCN), which is particularly vulnerable to bilirubin excitotoxicity. We found that NAD+ effectively decreased the size of evoked excitatory postsynaptic currents (eEPSCs), increased paired-pulse ratio (PPR) and reversed the effect of bilirubin on eEPSCs, implicating its inhibitory effects on the presynaptic release probability (Pr). Moreover, NAD+ not only decreased the basal frequency of miniature EPSCs (mEPSCs), but also reversed bilirubin-induced increases in the frequency of mEPSCs without affecting their amplitude under either condition. Furthermore, we found that NAD+ decreased the frequency of spontaneous firing of VCN neurons as well as bilirubin-induced increases in firing frequency. Whole-cell current-clamp recordings showed that NAD+ could directly decrease the intrinsic excitability of VCN neurons in the presence of synaptic blockers, suggesting NAD+ exerts its actions in both presynaptic and postsynaptic loci. Consistent with these observations, we found that the latency of the first postsynaptic spike triggered by high-frequency train stimulation of presynaptic afferents (i.e., the auditory nerve) was prolonged by NAD+. These results collectively indicate that NAD+ suppresses presynaptic transmitter release and postsynaptic excitability, jointly weakening excitatory neurotransmission. Our findings provide a basis for the exploration of NAD+ for the prevention and treatment of bilirubin encephalopathy and excitotoxicity associated with other neurological disorders.
Collapse
Affiliation(s)
- Min Liang
- Department of Otorhinolaryngology, The Sixth People's Hospital, Shanghai Jiao Tong University Shanghai, China
| | - Xin-Lu Yin
- Department of Otorhinolaryngology, The Sixth People's Hospital, Shanghai Jiao Tong University Shanghai, China
| | - Lu-Yang Wang
- Programs in Neurosciences and Mental Health, SickKids Research Institute and Department of Physiology, University of Toronto Toronto, ON, Canada
| | - Wei-Hai Yin
- Med-X Research Institute and School of Biomedical Engineering, Shanghai Jiao Tong University Shanghai Shanghai, China
| | - Ning-Ying Song
- Department of Otorhinolaryngology, West China Hospital, Sichuan University Chengdu, China
| | - Hai-Bo Shi
- Department of Otorhinolaryngology, The Sixth People's Hospital, Shanghai Jiao Tong University Shanghai, China
| | - Chun-Yan Li
- Department of Otorhinolaryngology, The Sixth People's Hospital, Shanghai Jiao Tong University Shanghai, China
| | - Shan-Kai Yin
- Department of Otorhinolaryngology, The Sixth People's Hospital, Shanghai Jiao Tong University Shanghai, China
| |
Collapse
|
36
|
Kaczmarek LK, Aldrich RW, Chandy KG, Grissmer S, Wei AD, Wulff H. International Union of Basic and Clinical Pharmacology. C. Nomenclature and Properties of Calcium-Activated and Sodium-Activated Potassium Channels. Pharmacol Rev 2017; 69:1-11. [PMID: 28267675 PMCID: PMC11060434 DOI: 10.1124/pr.116.012864] [Citation(s) in RCA: 68] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/02/2024] Open
Abstract
A subset of potassium channels is regulated primarily by changes in the cytoplasmic concentration of ions, including calcium, sodium, chloride, and protons. The eight members of this subfamily were originally all designated as calcium-activated channels. More recent studies have clarified the gating mechanisms for these channels and have documented that not all members are sensitive to calcium. This article describes the molecular relationships between these channels and provides an introduction to their functional properties. It also introduces a new nomenclature that differentiates between calcium- and sodium-activated potassium channels.
Collapse
Affiliation(s)
- Leonard K Kaczmarek
- Departments of Pharmacology and Cellular and Molecular Physiology, Yale School of Medicine, New Haven, Connecticut (L.K.K.); Center for Learning and Memory and Department of Neuroscience, University of Texas at Austin, Austin, Texas (R.W.A.); Laboratory of Molecular Physiology in the Infection and Immunity Theme, Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore (K.G.C.); Institute of Applied Physiology, Ulm University, Ulm, Germany (S.G.); Center for Integrative Brain Research, Seattle Children's Research Institute, Seattle, Washington (A.D.W.); and Department of Pharmacology, School of Medicine, University of California, Davis, California (H.W.)
| | - Richard W Aldrich
- Departments of Pharmacology and Cellular and Molecular Physiology, Yale School of Medicine, New Haven, Connecticut (L.K.K.); Center for Learning and Memory and Department of Neuroscience, University of Texas at Austin, Austin, Texas (R.W.A.); Laboratory of Molecular Physiology in the Infection and Immunity Theme, Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore (K.G.C.); Institute of Applied Physiology, Ulm University, Ulm, Germany (S.G.); Center for Integrative Brain Research, Seattle Children's Research Institute, Seattle, Washington (A.D.W.); and Department of Pharmacology, School of Medicine, University of California, Davis, California (H.W.)
| | - K George Chandy
- Departments of Pharmacology and Cellular and Molecular Physiology, Yale School of Medicine, New Haven, Connecticut (L.K.K.); Center for Learning and Memory and Department of Neuroscience, University of Texas at Austin, Austin, Texas (R.W.A.); Laboratory of Molecular Physiology in the Infection and Immunity Theme, Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore (K.G.C.); Institute of Applied Physiology, Ulm University, Ulm, Germany (S.G.); Center for Integrative Brain Research, Seattle Children's Research Institute, Seattle, Washington (A.D.W.); and Department of Pharmacology, School of Medicine, University of California, Davis, California (H.W.)
| | - Stephan Grissmer
- Departments of Pharmacology and Cellular and Molecular Physiology, Yale School of Medicine, New Haven, Connecticut (L.K.K.); Center for Learning and Memory and Department of Neuroscience, University of Texas at Austin, Austin, Texas (R.W.A.); Laboratory of Molecular Physiology in the Infection and Immunity Theme, Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore (K.G.C.); Institute of Applied Physiology, Ulm University, Ulm, Germany (S.G.); Center for Integrative Brain Research, Seattle Children's Research Institute, Seattle, Washington (A.D.W.); and Department of Pharmacology, School of Medicine, University of California, Davis, California (H.W.)
| | - Aguan D Wei
- Departments of Pharmacology and Cellular and Molecular Physiology, Yale School of Medicine, New Haven, Connecticut (L.K.K.); Center for Learning and Memory and Department of Neuroscience, University of Texas at Austin, Austin, Texas (R.W.A.); Laboratory of Molecular Physiology in the Infection and Immunity Theme, Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore (K.G.C.); Institute of Applied Physiology, Ulm University, Ulm, Germany (S.G.); Center for Integrative Brain Research, Seattle Children's Research Institute, Seattle, Washington (A.D.W.); and Department of Pharmacology, School of Medicine, University of California, Davis, California (H.W.)
| | - Heike Wulff
- Departments of Pharmacology and Cellular and Molecular Physiology, Yale School of Medicine, New Haven, Connecticut (L.K.K.); Center for Learning and Memory and Department of Neuroscience, University of Texas at Austin, Austin, Texas (R.W.A.); Laboratory of Molecular Physiology in the Infection and Immunity Theme, Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore (K.G.C.); Institute of Applied Physiology, Ulm University, Ulm, Germany (S.G.); Center for Integrative Brain Research, Seattle Children's Research Institute, Seattle, Washington (A.D.W.); and Department of Pharmacology, School of Medicine, University of California, Davis, California (H.W.)
| |
Collapse
|
37
|
Protective effect of P7C3 on retinal ganglion cells from optic nerve injury. Jpn J Ophthalmol 2016; 61:195-203. [DOI: 10.1007/s10384-016-0493-6] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2016] [Accepted: 11/07/2016] [Indexed: 10/20/2022]
|
38
|
Bansal V, Fisher TE. Na(+) -Activated K(+) Channels in Rat Supraoptic Neurones. J Neuroendocrinol 2016; 28. [PMID: 27091544 DOI: 10.1111/jne.12394] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/16/2016] [Revised: 04/06/2016] [Accepted: 04/15/2016] [Indexed: 11/29/2022]
Abstract
The magnocellular neurosecretory cells (MNCs) of the hypothalamus secrete the neurohormones vasopressin and oxytocin. The systemic release of these hormones depends on the rate and pattern of MNC firing and it is therefore important to identify the ion channels that contribute to the electrical behaviour of MNCs. In the present study, we report evidence for the presence of Na(+) -activated K(+) (KN a ) channels in rat MNCs. KN a channels mediate outwardly rectifying K(+) currents activated by the increases in intracellular Na(+) that occur during electrical activity. Although the molecular identity of native KN a channels is unclear, their biophysical properties are consistent with those of expressed Slick (slo 2.1) and Slack (slo 2.2) proteins. Using immunocytochemistry and Western blot experiments, we found that both Slick and Slack proteins are expressed in rat MNCs. Using whole cell voltage clamp techniques on acutely isolated rat MNCs, we found that inhibiting Na(+) influx by the addition of the Na(+) channel blocker tetrodotoxin or the replacement of Na(+) in the external solution with Li(+) caused a significant decrease in sustained outward currents. Furthermore, the evoked outward current density was significantly higher in rat MNCs using patch pipettes containing 60 mm Na(+) than it was when patch pipettes containing 0 mm Na(+) were used. Our data show that functional KN a channels are expressed in rat MNCs. These channels could contribute to the activity-dependent afterhyperpolarisations that have been identified in the MNCs and thereby play a role in the regulation of their electrical behaviour.
Collapse
Affiliation(s)
- V Bansal
- Department of Physiology, College of Medicine, University of Saskatchewan, Saskatoon, Saskatchewan, Canada
| | - T E Fisher
- Department of Physiology, College of Medicine, University of Saskatchewan, Saskatoon, Saskatchewan, Canada
| |
Collapse
|
39
|
Villa C, Combi R. Potassium Channels and Human Epileptic Phenotypes: An Updated Overview. Front Cell Neurosci 2016; 10:81. [PMID: 27064559 PMCID: PMC4811893 DOI: 10.3389/fncel.2016.00081] [Citation(s) in RCA: 95] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2015] [Accepted: 03/15/2016] [Indexed: 12/03/2022] Open
Abstract
Potassium (K+) channels are expressed in almost every cells and are ubiquitous in neuronal and glial cell membranes. These channels have been implicated in different disorders, in particular in epilepsy. K+ channel diversity depends on the presence in the human genome of a large number of genes either encoding pore-forming or accessory subunits. More than 80 genes encoding the K+ channels were cloned and they represent the largest group of ion channels regulating the electrical activity of cells in different tissues, including the brain. It is therefore not surprising that mutations in these genes lead to K+ channels dysfunctions linked to inherited epilepsy in humans and non-human model animals. This article reviews genetic and molecular progresses in exploring the pathogenesis of different human epilepsies, with special emphasis on the role of K+ channels in monogenic forms.
Collapse
Affiliation(s)
- Chiara Villa
- School of Medicine and Surgery, University of Milano-Bicocca Monza, Italy
| | - Romina Combi
- School of Medicine and Surgery, University of Milano-Bicocca Monza, Italy
| |
Collapse
|
40
|
Lim CX, Ricos MG, Dibbens LM, Heron SE. KCNT1mutations in seizure disorders: the phenotypic spectrum and functional effects. J Med Genet 2016; 53:217-25. [DOI: 10.1136/jmedgenet-2015-103508] [Citation(s) in RCA: 67] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2015] [Accepted: 12/06/2015] [Indexed: 01/01/2023]
|
41
|
Tang QY, Zhang FF, Xu J, Wang R, Chen J, Logothetis DE, Zhang Z. Epilepsy-Related Slack Channel Mutants Lead to Channel Over-Activity by Two Different Mechanisms. Cell Rep 2015; 14:129-139. [PMID: 26725113 DOI: 10.1016/j.celrep.2015.12.019] [Citation(s) in RCA: 54] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2015] [Revised: 08/11/2015] [Accepted: 11/23/2015] [Indexed: 01/07/2023] Open
Abstract
Twelve sodium-activated potassium channel (KCNT1, Slack) genetic mutants have been identified from severe early-onset epilepsy patients. The changes in biophysical properties of these mutants and the underlying mechanisms causing disease remain elusive. Here, we report that seven of the 12 mutations increase, whereas one mutation decreases, the channel's sodium sensitivity. Two of the mutants exhibit channel over-activity only when the intracellular Na(+) ([Na(+)]i) concentration is ∼80 mM. In contrast, single-channel data reveal that all 12 mutants increase the maximal open probability (Po). We conclude that these mutant channels lead to channel over-activity predominantly by increasing the ability of sodium binding to activate the channel, which is indicated by its maximal Po. The sodium sensitivity of these epilepsy causing mutants probably determines the [Na(+)]i concentration at which these mutants exert their pathological effects.
Collapse
Affiliation(s)
- Qiong-Yao Tang
- Jiangsu Province Key Laboratory of Anesthesiology, Xuzhou Medical College, Xuzhou, Jiangsu Province 221004, China; Jiangsu Province Key Laboratory of Anesthesia and Analgesia Application Technology, Xuzhou Medical College, Xuzhou, Jiangsu Province 221004, China
| | - Fei-Fei Zhang
- Jiangsu Province Key Laboratory of Anesthesiology, Xuzhou Medical College, Xuzhou, Jiangsu Province 221004, China; Jiangsu Province Key Laboratory of Anesthesia and Analgesia Application Technology, Xuzhou Medical College, Xuzhou, Jiangsu Province 221004, China
| | - Jie Xu
- Jiangsu Province Key Laboratory of Anesthesiology, Xuzhou Medical College, Xuzhou, Jiangsu Province 221004, China; Jiangsu Province Key Laboratory of Anesthesia and Analgesia Application Technology, Xuzhou Medical College, Xuzhou, Jiangsu Province 221004, China
| | - Ran Wang
- School of Anesthesiology, Xuzhou Medical College, Xuzhou, Jiangsu Province 221004, China
| | - Jian Chen
- School of Anesthesiology, Xuzhou Medical College, Xuzhou, Jiangsu Province 221004, China
| | - Diomedes E Logothetis
- Department of Physiology and Biophysics, Medical College of Virginia Campus, Virginia Commonwealth University, Richmond, VA 23298, USA
| | - Zhe Zhang
- Jiangsu Province Key Laboratory of Anesthesiology, Xuzhou Medical College, Xuzhou, Jiangsu Province 221004, China; Jiangsu Province Key Laboratory of Anesthesia and Analgesia Application Technology, Xuzhou Medical College, Xuzhou, Jiangsu Province 221004, China.
| |
Collapse
|
42
|
Suzuki T, Hansen A, Sanguinetti MC. Hydrophobic interactions between the S5 segment and the pore helix stabilizes the closed state of Slo2.1 potassium channels. BIOCHIMICA ET BIOPHYSICA ACTA-BIOMEMBRANES 2015; 1858:783-92. [PMID: 26724206 DOI: 10.1016/j.bbamem.2015.12.024] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/30/2015] [Revised: 11/26/2015] [Accepted: 12/21/2015] [Indexed: 11/28/2022]
Abstract
Under normal physiological conditions, Slo2.1K(+) channels are in a closed state unless activated by an elevation in [Na(+)]i. Fenamates such as niflumic acid also activate Slo2.1. Previous studies suggest that activation of Slo2.1 channels is mediated by a conformational change in the selectivity filter, and not a widening of the aperture formed by the S6 segment bundle crossing as occurs in voltage-gated K(+) channels. It is unclear how binding of Na(+) or fenamates is allosterically linked to opening of the presumed selectivity filter activation gate in Slo2.1. Here we examined the role of the S5 transmembrane segment in the activation of Slo2.1. Channels were heterologously expressed in Xenopus laevis oocytes and whole cell currents measured with the voltage-clamp technique. Ala substitution of five residues located on a single face of the S5 α-helical segment induced constitutive channel activity. Leu-209, predicted to face towards Phe-240 in the pore helix was investigated by further mutagenesis. Mutation of Leu-209 to Glu or Gln induced maximal channel activation as did the combined mutation to Ala of all three hydrophobic S5 residues predicted to be adjacent to Phe-240. Together these results suggest that hydrophobic interactions between residues in S5 and the C-terminal end of the pore helix stabilize Slo2.1 channels in a closed state.
Collapse
Affiliation(s)
- Tomoyuki Suzuki
- Nora Eccles Harrison Cardiovascular Research and Training Institute, University of Utah, Salt Lake City, UT 84112, USA
| | - Angela Hansen
- Nora Eccles Harrison Cardiovascular Research and Training Institute, University of Utah, Salt Lake City, UT 84112, USA
| | - Michael C Sanguinetti
- Nora Eccles Harrison Cardiovascular Research and Training Institute, University of Utah, Salt Lake City, UT 84112, USA; Department of Internal Medicine, Division of Cardiovascular Medicine, University of Utah, Salt Lake City, UT 84112, USA.
| |
Collapse
|
43
|
Gururaj S, Fleites J, Bhattacharjee A. Slack sodium-activated potassium channel membrane expression requires p38 mitogen-activated protein kinase phosphorylation. Neuropharmacology 2015; 103:279-89. [PMID: 26721627 DOI: 10.1016/j.neuropharm.2015.12.016] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2015] [Revised: 12/11/2015] [Accepted: 12/15/2015] [Indexed: 01/14/2023]
Abstract
p38 MAPK has long been understood as an inducible kinase under conditions of cellular stress, but there is now increasing evidence to support its role in the regulation of neuronal function. Several phosphorylation targets have been identified, an appreciable number of which are ion channels, implicating the possible involvement of p38 MAPK in neuronal excitability. The KNa channel Slack is an important protein to be studied as it is highly and ubiquitously expressed in DRG neurons and is important in the maintenance of their firing accommodation. We sought to examine if the Slack channel could be a substrate of p38 MAPK activity. First, we found that the Slack C-terminus contains two putative p38 MAPK phosphorylation sites that are highly conserved across species. Second, we show via electrophysiology experiments that KNa currents and further, Slack currents, are subject to tonic modulation by p38 MAPK. Third, biochemical approaches revealed that Slack channel regulation by p38 MAPK occurs through direct phosphorylation at the two putative sites of interaction, and mutating both sites prevented surface expression of Slack channels. Based on these results, we conclude that p38 MAPK is an obligate regulator of Slack channel function via the trafficking of channels into the membrane. The present study identifies Slack KNa channels as p38 MAPK substrates.
Collapse
Affiliation(s)
- Sushmitha Gururaj
- Department of Pharmacology and Toxicology, The State University of New York, Buffalo, USA
| | - John Fleites
- Program in Neuroscience, The State University of New York, Buffalo, USA
| | - Arin Bhattacharjee
- Department of Pharmacology and Toxicology, The State University of New York, Buffalo, USA; Program in Neuroscience, The State University of New York, Buffalo, USA.
| |
Collapse
|
44
|
Martinez-Espinosa PL, Wu J, Yang C, Gonzalez-Perez V, Zhou H, Liang H, Xia XM, Lingle CJ. Knockout of Slo2.2 enhances itch, abolishes KNa current, and increases action potential firing frequency in DRG neurons. eLife 2015; 4:e10013. [PMID: 26559620 PMCID: PMC4641468 DOI: 10.7554/elife.10013] [Citation(s) in RCA: 60] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2015] [Accepted: 10/06/2015] [Indexed: 02/02/2023] Open
Abstract
Two mammalian genes, Kcnt1 and Kcnt2, encode pore-forming subunits of Na(+)-dependent K(+) (KNa) channels. Progress in understanding KNa channels has been hampered by the absence of specific tools and methods for rigorous KNa identification in native cells. Here, we report the genetic disruption of both Kcnt1 and Kcnt2, confirm the loss of Slo2.2 and Slo2.1 protein, respectively, in KO animals, and define tissues enriched in Slo2 expression. Noting the prevalence of Slo2.2 in dorsal root ganglion, we find that KO of Slo2.2, but not Slo2.1, results in enhanced itch and pain responses. In dissociated small diameter DRG neurons, KO of Slo2.2, but not Slo2.1, abolishes KNa current. Utilizing isolectin B4+ neurons, the absence of KNa current results in an increase in action potential (AP) firing and a decrease in AP threshold. Activation of KNa acts as a brake to initiation of the first depolarization-elicited AP with no discernible effect on afterhyperpolarizations.
Collapse
Affiliation(s)
| | - Jianping Wu
- Department of Anesthesiology, Washington University School of Medicine, St. Louis, United States
| | - Chengtao Yang
- Department of Anesthesiology, Washington University School of Medicine, St. Louis, United States
| | - Vivian Gonzalez-Perez
- Department of Anesthesiology, Washington University School of Medicine, St. Louis, United States
| | - Huifang Zhou
- Department of Anesthesiology, Washington University School of Medicine, St. Louis, United States
| | - Hongwu Liang
- Department of Anesthesiology, Washington University School of Medicine, St. Louis, United States
| | - Xiao-Ming Xia
- Department of Anesthesiology, Washington University School of Medicine, St. Louis, United States
| | - Christopher J Lingle
- Department of Anesthesiology, Washington University School of Medicine, St. Louis, United States
| |
Collapse
|
45
|
Fernández-Agüera MC, Gao L, González-Rodríguez P, Pintado CO, Arias-Mayenco I, García-Flores P, García-Pergañeda A, Pascual A, Ortega-Sáenz P, López-Barneo J. Oxygen Sensing by Arterial Chemoreceptors Depends on Mitochondrial Complex I Signaling. Cell Metab 2015; 22:825-37. [PMID: 26437605 DOI: 10.1016/j.cmet.2015.09.004] [Citation(s) in RCA: 156] [Impact Index Per Article: 15.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/09/2015] [Revised: 07/17/2015] [Accepted: 09/08/2015] [Indexed: 12/30/2022]
Abstract
O2 sensing is essential for mammalian homeostasis. Peripheral chemoreceptors such as the carotid body (CB) contain cells with O2-sensitive K(+) channels, which are inhibited by hypoxia to trigger fast adaptive cardiorespiratory reflexes. How variations of O2 tension (PO2) are detected and the mechanisms whereby these changes are conveyed to membrane ion channels have remained elusive. We have studied acute O2 sensing in conditional knockout mice lacking mitochondrial complex I (MCI) genes. We inactivated Ndufs2, which encodes a protein that participates in ubiquinone binding. Ndufs2-null mice lose the hyperventilatory response to hypoxia, although they respond to hypercapnia. Ndufs2-deficient CB cells have normal functions and ATP content but are insensitive to changes in PO2. Our data suggest that chemoreceptor cells have a specialized succinate-dependent metabolism that induces an MCI state during hypoxia, characterized by the production of reactive oxygen species and accumulation of reduced pyridine nucleotides, which signal neighboring K(+) channels.
Collapse
Affiliation(s)
- M Carmen Fernández-Agüera
- Instituto de Biomedicina de Sevilla (IBiS), Hospital Universitario Virgen del Rocío/CSIC/Universidad de Sevilla, Campus Hospital Universitario Virgen del Rocío, Avenida Manuel Siurot, s/n, 41013 Seville, Spain; Departamento de Fisiología Médica y Biofísica, Facultad de Medicina, Universidad de Sevilla, Avenida Sánchez Pizjuan, 4, 41009 Seville, Spain; Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), Campus Hospital Universitario Virgen del Rocío, Avenida Manuel Siurot, s/n, 41013 Seville, Spain
| | - Lin Gao
- Instituto de Biomedicina de Sevilla (IBiS), Hospital Universitario Virgen del Rocío/CSIC/Universidad de Sevilla, Campus Hospital Universitario Virgen del Rocío, Avenida Manuel Siurot, s/n, 41013 Seville, Spain; Departamento de Fisiología Médica y Biofísica, Facultad de Medicina, Universidad de Sevilla, Avenida Sánchez Pizjuan, 4, 41009 Seville, Spain; Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), Campus Hospital Universitario Virgen del Rocío, Avenida Manuel Siurot, s/n, 41013 Seville, Spain
| | - Patricia González-Rodríguez
- Instituto de Biomedicina de Sevilla (IBiS), Hospital Universitario Virgen del Rocío/CSIC/Universidad de Sevilla, Campus Hospital Universitario Virgen del Rocío, Avenida Manuel Siurot, s/n, 41013 Seville, Spain; Departamento de Fisiología Médica y Biofísica, Facultad de Medicina, Universidad de Sevilla, Avenida Sánchez Pizjuan, 4, 41009 Seville, Spain; Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), Campus Hospital Universitario Virgen del Rocío, Avenida Manuel Siurot, s/n, 41013 Seville, Spain
| | - C Oscar Pintado
- Centro de Producción y Experimentación Animal, Universidad de Sevilla, Calle San Fernando, 4, 41004 Seville, Spain
| | - Ignacio Arias-Mayenco
- Instituto de Biomedicina de Sevilla (IBiS), Hospital Universitario Virgen del Rocío/CSIC/Universidad de Sevilla, Campus Hospital Universitario Virgen del Rocío, Avenida Manuel Siurot, s/n, 41013 Seville, Spain; Departamento de Fisiología Médica y Biofísica, Facultad de Medicina, Universidad de Sevilla, Avenida Sánchez Pizjuan, 4, 41009 Seville, Spain; Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), Campus Hospital Universitario Virgen del Rocío, Avenida Manuel Siurot, s/n, 41013 Seville, Spain
| | - Paula García-Flores
- Instituto de Biomedicina de Sevilla (IBiS), Hospital Universitario Virgen del Rocío/CSIC/Universidad de Sevilla, Campus Hospital Universitario Virgen del Rocío, Avenida Manuel Siurot, s/n, 41013 Seville, Spain; Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), Campus Hospital Universitario Virgen del Rocío, Avenida Manuel Siurot, s/n, 41013 Seville, Spain
| | - Antonio García-Pergañeda
- Instituto de Biomedicina de Sevilla (IBiS), Hospital Universitario Virgen del Rocío/CSIC/Universidad de Sevilla, Campus Hospital Universitario Virgen del Rocío, Avenida Manuel Siurot, s/n, 41013 Seville, Spain
| | - Alberto Pascual
- Instituto de Biomedicina de Sevilla (IBiS), Hospital Universitario Virgen del Rocío/CSIC/Universidad de Sevilla, Campus Hospital Universitario Virgen del Rocío, Avenida Manuel Siurot, s/n, 41013 Seville, Spain
| | - Patricia Ortega-Sáenz
- Instituto de Biomedicina de Sevilla (IBiS), Hospital Universitario Virgen del Rocío/CSIC/Universidad de Sevilla, Campus Hospital Universitario Virgen del Rocío, Avenida Manuel Siurot, s/n, 41013 Seville, Spain; Departamento de Fisiología Médica y Biofísica, Facultad de Medicina, Universidad de Sevilla, Avenida Sánchez Pizjuan, 4, 41009 Seville, Spain; Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), Campus Hospital Universitario Virgen del Rocío, Avenida Manuel Siurot, s/n, 41013 Seville, Spain
| | - José López-Barneo
- Instituto de Biomedicina de Sevilla (IBiS), Hospital Universitario Virgen del Rocío/CSIC/Universidad de Sevilla, Campus Hospital Universitario Virgen del Rocío, Avenida Manuel Siurot, s/n, 41013 Seville, Spain; Departamento de Fisiología Médica y Biofísica, Facultad de Medicina, Universidad de Sevilla, Avenida Sánchez Pizjuan, 4, 41009 Seville, Spain; Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), Campus Hospital Universitario Virgen del Rocío, Avenida Manuel Siurot, s/n, 41013 Seville, Spain.
| |
Collapse
|
46
|
Identification of potential novel interaction partners of the sodium-activated potassium channels Slick and Slack in mouse brain. Biochem Biophys Rep 2015; 4:291-298. [PMID: 29124216 PMCID: PMC5669359 DOI: 10.1016/j.bbrep.2015.09.024] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2015] [Revised: 09/18/2015] [Accepted: 09/29/2015] [Indexed: 02/06/2023] Open
Abstract
The sodium-activated potassium channels Slick (Slo2.1, KCNT2) and Slack (Slo2.2, KCNT1) are paralogous channels of the Slo family of high-conductance potassium channels. Slick and Slack channels are widely distributed in the mammalian CNS and they play a role in slow afterhyperpolarization, generation of depolarizing afterpotentials and in setting and stabilizing the resting potential. In the present study we used a combined approach of (co)-immunoprecipitation studies, Western blot analysis, double immunofluorescence and mass spectrometric sequencing in order to investigate protein-protein interactions of the Slick and Slack channels. The data strongly suggest that Slick and Slack channels co-assemble into identical cellular complexes. Double immunofluorescence experiments revealed that Slick and Slack channels co-localize in distinct mouse brain regions. Moreover, we identified the small cytoplasmic protein beta-synuclein and the transmembrane protein 263 (TMEM 263) as novel interaction partners of both, native Slick and Slack channels. In addition, the inactive dipeptidyl-peptidase (DPP 10) and the synapse associated protein 102 (SAP 102) were identified as constituents of the native Slick and Slack channel complexes in the mouse brain. This study presents new insights into protein-protein interactions of native Slick and Slack channels in the mouse brain.
Collapse
|
47
|
Tomasello DL, Gancarz-Kausch AM, Dietz DM, Bhattacharjee A. Transcriptional Regulation of the Sodium-activated Potassium Channel SLICK (KCNT2) Promoter by Nuclear Factor-κB. J Biol Chem 2015; 290:18575-83. [PMID: 26100633 DOI: 10.1074/jbc.m115.643536] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2015] [Indexed: 11/06/2022] Open
Abstract
Although recent studies have shown the sodium-activated potassium channel SLACK (KCNT1) can contribute to neuronal excitability, there remains little information on the physiological role of the closely related SLICK (KCNT2) channel. Activation of SLICK channels may be important during pathological states such as ischemia, in which an increase in intracellular sodium and chloride can perturb membrane potential and ion homeostasis. We have identified two NFκB-binding sites within the promoter region of the human SLICK (KCNT2) and orthologous rat Slick (Kcnt2) genes, suggesting that conditions in which NFκB transcriptional activity is elevated promote expression of this channel. NFκB binding to the rat Slick promoter was confirmed in vivo by ChIP analyses, and NFκB was found differentially bound to the two sites. We verified NFκB transcriptional regulation of SLICK/Slick by mutational analyses and studying gene expression by luciferase assay in P19 cells, where NFκB is constitutively active. For the rat gene, activation of the Slick promoter was found to be additive in single NFκB mutations and synergistic in double mutations. Unexpectedly, for the human gene, NFκB exhibited cooperativity in activating the SLICK promoter. The human SLICK promoter constructs were then tested under hypoxic conditions in PC-12 cells, where NFκB is not active. Only under hypoxic conditions could luciferase activity be detected; the double NFκB mutant construct failed to exhibit activity. Transcriptional regulation of Slick by NFκB was verified in primary neurons. The Slick transcript decreased 24 h after NFκB inhibition. Our data show SLICK expression is predominantly under the control of NFκB. Because neuronal NFκB activation occurs during stressful stimuli such as hypoxia and injury, our findings suggest that SLICK is a neuroprotective gene.
Collapse
Affiliation(s)
| | - Amy M Gancarz-Kausch
- Department of Pharmacology and Toxicology, The State University of New York at Buffalo, Buffalo, New York 14214
| | - David M Dietz
- From the Program in Neuroscience and Department of Pharmacology and Toxicology, The State University of New York at Buffalo, Buffalo, New York 14214
| | - Arin Bhattacharjee
- From the Program in Neuroscience and Department of Pharmacology and Toxicology, The State University of New York at Buffalo, Buffalo, New York 14214
| |
Collapse
|
48
|
Bausch AE, Dieter R, Nann Y, Hausmann M, Meyerdierks N, Kaczmarek LK, Ruth P, Lukowski R. The sodium-activated potassium channel Slack is required for optimal cognitive flexibility in mice. ACTA ACUST UNITED AC 2015; 22:323-35. [PMID: 26077685 PMCID: PMC4478330 DOI: 10.1101/lm.037820.114] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2014] [Accepted: 05/05/2015] [Indexed: 01/14/2023]
Abstract
Kcnt1 encoded sodium-activated potassium channels (Slack channels) are highly expressed throughout the brain where they modulate the firing patterns and general excitability of many types of neurons. Increasing evidence suggests that Slack channels may be important for higher brain functions such as cognition and normal intellectual development. In particular, recent findings have shown that human Slack mutations produce very severe intellectual disability and that Slack channels interact directly with the Fragile X mental retardation protein (FMRP), a protein that when missing or mutated results in Fragile X syndrome (FXS), the most common form of inherited intellectual disability and autism in humans. We have now analyzed a recently developed Kcnt1 null mouse model in several behavioral tasks to assess which aspects of memory and learning are dependent on Slack. We demonstrate that Slack deficiency results in mildly altered general locomotor activity, but normal working memory, reference memory, as well as cerebellar control of motor functions. In contrast, we find that Slack channels are required for cognitive flexibility, including reversal learning processes and the ability to adapt quickly to unfamiliar situations and environments. Our data reveal that hippocampal-dependent spatial learning capabilities require the proper function of Slack channels.
Collapse
Affiliation(s)
- Anne E Bausch
- Pharmakologie, Toxikologie und Klinische Pharmazie, Institut für Pharmazie, 72076 Tübingen, Germany
| | - Rebekka Dieter
- Pharmakologie, Toxikologie und Klinische Pharmazie, Institut für Pharmazie, 72076 Tübingen, Germany
| | - Yvette Nann
- Pharmakologie, Toxikologie und Klinische Pharmazie, Institut für Pharmazie, 72076 Tübingen, Germany
| | - Mario Hausmann
- Pharmakologie, Toxikologie und Klinische Pharmazie, Institut für Pharmazie, 72076 Tübingen, Germany
| | - Nora Meyerdierks
- Pharmakologie, Toxikologie und Klinische Pharmazie, Institut für Pharmazie, 72076 Tübingen, Germany
| | - Leonard K Kaczmarek
- Department of Pharmacology, Yale University School of Medicine, New Haven, Connecticut 06520, USA Department of Cellular and Molecular Physiology, Yale University School of Medicine, New Haven, Connecticut 06520, USA
| | - Peter Ruth
- Pharmakologie, Toxikologie und Klinische Pharmazie, Institut für Pharmazie, 72076 Tübingen, Germany
| | - Robert Lukowski
- Pharmakologie, Toxikologie und Klinische Pharmazie, Institut für Pharmazie, 72076 Tübingen, Germany
| |
Collapse
|
49
|
Kasten MR, Anderson MP. Self-regulation of adult thalamocortical neurons. J Neurophysiol 2015; 114:323-31. [PMID: 25948871 DOI: 10.1152/jn.00800.2014] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2014] [Accepted: 05/03/2015] [Indexed: 11/22/2022] Open
Abstract
The thalamus acts as a conduit for sensory and other information traveling to the cortex. In response to continuous sensory stimulation in vivo, the firing rate of thalamocortical neurons initially increases, but then within a minute firing rate decreases and T-type Ca(2+) channel-dependent action potential burst firing emerges. While neuromodulatory systems could play a role in this inhibitory response, we instead report a novel and cell-autonomous inhibitory mechanism intrinsic to the thalamic relay neuron. Direct intracellular stimulation of thalamocortical neuron firing initially triggered a continuous and high rate of action potential discharge, but within a minute membrane potential (Vm) was hyperpolarized and firing rate to the same stimulus was decreased. This self-inhibition was observed across a wide variety of thalamic nuclei, and in a subset firing mode switched from tonic to bursting. The self-inhibition resisted blockers of intracellular Ca(2+) signaling, Na(+)-K(+)-ATPases, and G protein-regulated inward rectifier (GIRK) channels as implicated in other neuron subtypes, but instead was in part inhibited by an ATP-sensitive K(+) channel blocker. The results identify a new homeostatic mechanism within the thalamus capable of gating excitatory signals at the single-cell level.
Collapse
Affiliation(s)
- Michael R Kasten
- Departments of Neurology and Pathology, Harvard Medical School and Beth Israel Deaconess Medical Center, Center for Life Science, Boston, Massachusetts
| | - Matthew P Anderson
- Departments of Neurology and Pathology, Harvard Medical School and Beth Israel Deaconess Medical Center, Center for Life Science, Boston, Massachusetts; Program in Neuroscience, Harvard Medical School, Boston, Massachusetts; and Children's Hospital Boston Intellectual and Developmental Disabilities Research Center, Children's Hospital Boston, Boston, Massachusetts
| |
Collapse
|
50
|
Ban Y, Smith BE, Markham MR. A highly polarized excitable cell separates sodium channels from sodium-activated potassium channels by more than a millimeter. J Neurophysiol 2015; 114:520-30. [PMID: 25925327 DOI: 10.1152/jn.00475.2014] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2014] [Accepted: 04/28/2015] [Indexed: 01/25/2023] Open
Abstract
The bioelectrical properties and resulting metabolic demands of electrogenic cells are determined by their morphology and the subcellular localization of ion channels. The electric organ cells (electrocytes) of the electric fish Eigenmannia virescens generate action potentials (APs) with Na(+) currents >10 μA and repolarize the AP with Na(+)-activated K(+) (KNa) channels. To better understand the role of morphology and ion channel localization in determining the metabolic cost of electrocyte APs, we used two-photon three-dimensional imaging to determine the fine cellular morphology and immunohistochemistry to localize the electrocytes' ion channels, ionotropic receptors, and Na(+)-K(+)-ATPases. We found that electrocytes are highly polarized cells ∼ 1.5 mm in anterior-posterior length and ∼ 0.6 mm in diameter, containing ∼ 30,000 nuclei along the cell periphery. The cell's innervated posterior region is deeply invaginated and vascularized with complex ultrastructural features, whereas the anterior region is relatively smooth. Cholinergic receptors and Na(+) channels are restricted to the innervated posterior region, whereas inward rectifier K(+) channels and the KNa channels that terminate the electrocyte AP are localized to the anterior region, separated by >1 mm from the only sources of Na(+) influx. In other systems, submicrometer spatial coupling of Na(+) and KNa channels is necessary for KNa channel activation. However, our computational simulations showed that KNa channels at a great distance from Na(+) influx can still terminate the AP, suggesting that KNa channels can be activated by distant sources of Na(+) influx and overturning a long-standing assumption that AP-generating ion channels are restricted to the electrocyte's posterior face.
Collapse
Affiliation(s)
- Yue Ban
- Department of Biology, The University of Oklahoma, Norman, Oklahoma; Cellular & Behavioral Neurobiology Graduate Program, The University of Oklahoma, Norman, Oklahoma; and
| | - Benjamin E Smith
- Samuel Roberts Noble Microscopy Laboratory, The University of Oklahoma, Norman, Oklahoma
| | - Michael R Markham
- Department of Biology, The University of Oklahoma, Norman, Oklahoma; Cellular & Behavioral Neurobiology Graduate Program, The University of Oklahoma, Norman, Oklahoma; and
| |
Collapse
|