1
|
Qiu Q, Yang M, Gong D, Liang H, Chen T. Potassium and calcium channels in different nerve cells act as therapeutic targets in neurological disorders. Neural Regen Res 2025; 20:1258-1276. [PMID: 38845230 PMCID: PMC11624876 DOI: 10.4103/nrr.nrr-d-23-01766] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2023] [Revised: 02/18/2024] [Accepted: 04/07/2024] [Indexed: 07/31/2024] Open
Abstract
The central nervous system, information integration center of the body, is mainly composed of neurons and glial cells. The neuron is one of the most basic and important structural and functional units of the central nervous system, with sensory stimulation and excitation conduction functions. Astrocytes and microglia belong to the glial cell family, which is the main source of cytokines and represents the main defense system of the central nervous system. Nerve cells undergo neurotransmission or gliotransmission, which regulates neuronal activity via the ion channels, receptors, or transporters expressed on nerve cell membranes. Ion channels, composed of large transmembrane proteins, play crucial roles in maintaining nerve cell homeostasis. These channels are also important for control of the membrane potential and in the secretion of neurotransmitters. A variety of cellular functions and life activities, including functional regulation of the central nervous system, the generation and conduction of nerve excitation, the occurrence of receptor potential, heart pulsation, smooth muscle peristalsis, skeletal muscle contraction, and hormone secretion, are closely related to ion channels associated with passive transmembrane transport. Two types of ion channels in the central nervous system, potassium channels and calcium channels, are closely related to various neurological disorders, including Alzheimer's disease, Parkinson's disease, and epilepsy. Accordingly, various drugs that can affect these ion channels have been explored deeply to provide new directions for the treatment of these neurological disorders. In this review, we focus on the functions of potassium and calcium ion channels in different nerve cells and their involvement in neurological disorders such as Parkinson's disease, Alzheimer's disease, depression, epilepsy, autism, and rare disorders. We also describe several clinical drugs that target potassium or calcium channels in nerve cells and could be used to treat these disorders. We concluded that there are few clinical drugs that can improve the pathology these diseases by acting on potassium or calcium ions. Although a few novel ion-channel-specific modulators have been discovered, meaningful therapies have largely not yet been realized. The lack of target-specific drugs, their requirement to cross the blood-brain barrier, and their exact underlying mechanisms all need further attention. This review aims to explain the urgent problems that need research progress and provide comprehensive information aiming to arouse the research community's interest in the development of ion channel-targeting drugs and the identification of new therapeutic targets for that can increase the cure rate of nervous system diseases and reduce the occurrence of adverse reactions in other systems.
Collapse
Affiliation(s)
- Qing Qiu
- Department of Pharmacology, School of Pharmacy, Nantong University, Nantong, Jiangsu Province, China
- Jiangsu Province Key Laboratory of Inflammation and Molecular Drug Target, Nantong, Jiangsu Province, China
| | - Mengting Yang
- Department of Pharmacology, School of Pharmacy, Nantong University, Nantong, Jiangsu Province, China
- Jiangsu Province Key Laboratory of Inflammation and Molecular Drug Target, Nantong, Jiangsu Province, China
| | - Danfeng Gong
- Department of Pharmacology, School of Pharmacy, Nantong University, Nantong, Jiangsu Province, China
- Jiangsu Province Key Laboratory of Inflammation and Molecular Drug Target, Nantong, Jiangsu Province, China
| | - Haiying Liang
- Department of Pharmacy, Longyan First Affiliated Hospital of Fujian Medical University, Longyan, Fujian Province, China
| | - Tingting Chen
- Department of Pharmacology, School of Pharmacy, Nantong University, Nantong, Jiangsu Province, China
- Jiangsu Province Key Laboratory of Inflammation and Molecular Drug Target, Nantong, Jiangsu Province, China
| |
Collapse
|
2
|
Pham DL, Cox K, Ko ML, Ko GYP. Peptide Lv and Angiogenesis: A Newly Discovered Angiogenic Peptide. Biomedicines 2024; 12:2851. [PMID: 39767758 PMCID: PMC11672992 DOI: 10.3390/biomedicines12122851] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2024] [Revised: 12/09/2024] [Accepted: 12/13/2024] [Indexed: 01/11/2025] Open
Abstract
Peptide Lv is a small endogenous secretory peptide with ~40 amino acids and is highly conserved among certain several species. While it was first discovered that it augments L-type voltage-gated calcium channels (LTCCs) in neurons, thus it was named peptide "Lv", it can bind to vascular endothelial growth factor receptor 2 (VEGFR2) and has VEGF-like activities, including eliciting vasodilation and promoting angiogenesis. Not only does peptide Lv augment LTCCs in neurons and cardiomyocytes, but it also promotes the expression of intermediate-conductance KCa channels (KCa3.1) in vascular endothelial cells. Peptide Lv is upregulated in the retinas of patients with early proliferative diabetic retinopathy, a disease involving pathological angiogenesis. This review will provide an overview of peptide Lv, its known bioactivities in vitro and in vivo, and its clinical relevance, with a focus on its role in angiogenesis. As there is more about peptide Lv to be explored, this article serves as a foundation for possible future developments of peptide Lv-related therapeutics to treat or prevent diseases.
Collapse
Affiliation(s)
- Dylan L. Pham
- Department of Veterinary Integrative Biosciences, College of Veterinary Medicine and Biomedical Sciences, Texas A&M University, College Station, TX 77843, USA
- Department of Medical Physiology, School of Medicine, Texas A&M University, Bryan, TX 77807, USA
| | - Kelsey Cox
- Department of Veterinary Integrative Biosciences, College of Veterinary Medicine and Biomedical Sciences, Texas A&M University, College Station, TX 77843, USA
| | - Michael L. Ko
- Department of Veterinary Integrative Biosciences, College of Veterinary Medicine and Biomedical Sciences, Texas A&M University, College Station, TX 77843, USA
- Department of Biology, Division of Natural and Physical Sciences, Blinn College, Bryan, TX 77802, USA
| | - Gladys Y.-P. Ko
- Department of Veterinary Integrative Biosciences, College of Veterinary Medicine and Biomedical Sciences, Texas A&M University, College Station, TX 77843, USA
- Texas A&M Institute for Neuroscience, Texas A&M University, College Station, TX 77843, USA
| |
Collapse
|
3
|
Thi Hong Van N, Hyun Nam J. Intermediate conductance calcium-activated potassium channel (KCa3.1) in cancer: Emerging roles and therapeutic potentials. Biochem Pharmacol 2024; 230:116573. [PMID: 39396649 DOI: 10.1016/j.bcp.2024.116573] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2024] [Revised: 10/02/2024] [Accepted: 10/10/2024] [Indexed: 10/15/2024]
Abstract
The KCa3.1 channel (also known as the KCNN4, IK1, or SK4 channel) is an intermediate-conductance calcium-activated potassium channel that regulates the membrane potential and maintains calcium homeostasis. Recently, KCa3.1 channels have attracted increasing attention because of their diverse roles in various types of cancers. In cancer cells, KCa3.1 channels regulate key processes, including cell proliferation, cell cycle, migration, invasion, tumor microenvironments, and therapy resistance. In addition, abnormal KCa3.1 expression in cancers is utilized to distinguish between tumor and normal tissues, classify cancer stages, and predict patient survival outcomes. This review comprehensively examines the current understanding of the contribution of KCa3.1 channels to tumor formation, metastasis, and its mechanisms. We evaluated the potential of KCa3.1 as a biomarker for cancer diagnosis and prognosis. Finally, we discuss the advances and challenges of applying KCa3.1 modulators in cancer treatment and propose approaches to overcome these obstacles. In summary, this review highlights the importance of this ion channel as a potent therapeutic target and prognostic biomarker of cancer.
Collapse
Affiliation(s)
- Nhung Thi Hong Van
- Department of Physiology, Dongguk University College of Medicine, Gyeongju 38066, Republic of Korea; Channelopathy Research Center (CRC), Dongguk University College of Medicine, Goyang 10326, Republic of Korea
| | - Joo Hyun Nam
- Department of Physiology, Dongguk University College of Medicine, Gyeongju 38066, Republic of Korea; Channelopathy Research Center (CRC), Dongguk University College of Medicine, Goyang 10326, Republic of Korea.
| |
Collapse
|
4
|
Oblasov I, Bal NV, Shvadchenko AM, Fortygina P, Idzhilova OS, Balaban PM, Nikitin ES. Ca 2+-permeable AMPA receptor-dependent silencing of neurons by KCa3.1 channels during epileptiform activity. Biochem Biophys Res Commun 2024; 733:150434. [PMID: 39068818 DOI: 10.1016/j.bbrc.2024.150434] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2024] [Accepted: 07/20/2024] [Indexed: 07/30/2024]
Abstract
Ca2+-activated KCa3.1 channels are known to contribute to slow afterhyperpolarization in pyramidal neurons of several brain areas, while Ca2+-permeable AMPA receptors (CP-AMPARs) may provide a subthreshold source of Ca2+ elevation in the cytoplasm. The functionality of these two types of channels has also been shown to be altered by epileptic disorders. However, the link between KCa3.1 channels and CP-AMPARs is poorly understood, and their potential interaction in epilepsy remains unclear. Here, we address this issue by overexpressing the KCNN4 gene, which encodes the KCa3.1 channel, using patch clamp, imaging, and channel blockers in an in vitro model of epilepsy in neuronal culture. We show that KCNN4 overexpression causes strong hyperpolarization and substantial silencing of neurons during epileptiform activity events, which also prevents KCNN4-positive neurons from firing action potentials (APs) during experimentally induced status epilepticus. Intracellular blocker application experiments showed that the amplitude of hyperpolarization was strongly dependent on CP-AMPARs, but not on NMDA receptors. Taken together, our data strongly suggest that subthreshold Ca2+ elevation produced by CP-AMPARs can trigger KCa3.1 channels to hyperpolarize neurons and protect them from seizures.
Collapse
Affiliation(s)
- Ilya Oblasov
- Institute of Higher Nervous Activity and Neurophysiology, Russian Academy of Sciences, 5a Butlerova str., Moscow, Russia, 117485
| | - Natalia V Bal
- Institute of Higher Nervous Activity and Neurophysiology, Russian Academy of Sciences, 5a Butlerova str., Moscow, Russia, 117485
| | - Anastasya M Shvadchenko
- Institute of Higher Nervous Activity and Neurophysiology, Russian Academy of Sciences, 5a Butlerova str., Moscow, Russia, 117485
| | - Polina Fortygina
- Institute of Higher Nervous Activity and Neurophysiology, Russian Academy of Sciences, 5a Butlerova str., Moscow, Russia, 117485
| | - Olga S Idzhilova
- Institute of Higher Nervous Activity and Neurophysiology, Russian Academy of Sciences, 5a Butlerova str., Moscow, Russia, 117485
| | - Pavel M Balaban
- Institute of Higher Nervous Activity and Neurophysiology, Russian Academy of Sciences, 5a Butlerova str., Moscow, Russia, 117485
| | - Evgeny S Nikitin
- Institute of Higher Nervous Activity and Neurophysiology, Russian Academy of Sciences, 5a Butlerova str., Moscow, Russia, 117485.
| |
Collapse
|
5
|
Di C, Wu T, Gao K, Li N, Song H, Wang L, Sun H, Yi J, Zhang X, Chen J, Shah M, Jiang Y, Huang Z. Carvedilol inhibits neuronal hyperexcitability caused by epilepsy-associated KCNT1 mutations. Br J Pharmacol 2024. [PMID: 39370580 DOI: 10.1111/bph.17360] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2024] [Revised: 08/27/2024] [Accepted: 09/03/2024] [Indexed: 10/08/2024] Open
Abstract
BACKGROUND AND PURPOSE KCNT1 encodes a sodium-activated potassium channel (Slack channel), and its mutation can cause several forms of epilepsy. Traditional antiepileptic medications have limited efficacy in treating patients with KCNT1 mutations. Here, we describe one heterozygous KCNT1 mutation, M267T, in a patient with EIMFS. The pathological channel properties of this mutation and its effect on neuronal excitability were investigated. Additionally, this study aimed to develop a medication for effective prevention of KCNT1 mutation-induced seizures. EXPERIMENTAL APPROACH Wild-type or mutant KCNT1 plasmids were expressed heterologously in Xenopus laevis oocytes, and channel property assessment and drug screening were performed based on two-electrode voltage-clamp recordings. The single-channel properties were investigated using the excised inside-out patches from HEK293T cells. Through in utero electroporation, WT and M267T Slack channels were expressed in the hippocampal CA1 pyramidal neurons in male mice, followed by the examination of the electrical properties using the whole-cell current-clamp technique. The kainic acid-induced epilepsy model in male mice was used to evalute the antiseizure effects of carvedilol. KEY RESULTS The KCNT1 M267T mutation enhanced Slack channel function by increasing single-channel open probability. Through screening 16 FDA-approved ion channel blockers, we found that carvedilol effectively reversed the mutation-induced gain-of-function channel properties. Notably, the KCNT1 M267T mutation in the mouse hippocampal CA1 pyramidal neurons affected afterhyperpolarization properties and induced neuronal hyperexcitability, which was inhibited by carvedilol. Additionally, carvedilol exhibited antiseizure effects in the kainic acid-induced epilepsy model. CONCLUSION AND IMPLICATION Our findings suggest carvedilol as a new potential candidate for treatment of epilepsies.
Collapse
Affiliation(s)
- Chang Di
- State Key Laboratory of Natural and Biomimetic Drugs, Department of Molecular and Cellular Pharmacology, School of Pharmaceutical Sciences, Peking University Health Science Center, Beijing, China
- Department of Pharmacology, School of Basic Medical Sciences, Peking University Health Science Center; State Key Laboratory of Vascular Homeostasis and Remodeling, Peking University, Beijing, China
| | - Tong Wu
- State Key Laboratory of Natural and Biomimetic Drugs, Department of Molecular and Cellular Pharmacology, School of Pharmaceutical Sciences, Peking University Health Science Center, Beijing, China
| | - Kai Gao
- Department of Pediatrics, Peking University First Hospital, Beijing, China
- Beijing Key Laboratory of Molecular Diagnosis and Study on Pediatric Genetic Diseases, Beijing, China
- Children Epilepsy Center, Peking University First Hospital, Beijing, China
- Key Laboratory for Neuroscience, Ministry of Education/National Health and Family Planning Commission, Peking University, Beijing, China
| | - Na Li
- State Key Laboratory of Natural and Biomimetic Drugs, Department of Molecular and Cellular Pharmacology, School of Pharmaceutical Sciences, Peking University Health Science Center, Beijing, China
| | - Huifang Song
- State Key Laboratory of Natural and Biomimetic Drugs, Department of Molecular and Cellular Pharmacology, School of Pharmaceutical Sciences, Peking University Health Science Center, Beijing, China
| | - Lili Wang
- State Key Laboratory of Natural and Biomimetic Drugs, Department of Molecular and Cellular Pharmacology, School of Pharmaceutical Sciences, Peking University Health Science Center, Beijing, China
| | - Haojie Sun
- UCL School of Pharmacy, University College London, London, UK
| | - Jingyun Yi
- State Key Laboratory of Natural and Biomimetic Drugs, Department of Molecular and Cellular Pharmacology, School of Pharmaceutical Sciences, Peking University Health Science Center, Beijing, China
| | - Xinran Zhang
- State Key Laboratory of Natural and Biomimetic Drugs, Department of Molecular and Cellular Pharmacology, School of Pharmaceutical Sciences, Peking University Health Science Center, Beijing, China
| | - Jiexin Chen
- State Key Laboratory of Natural and Biomimetic Drugs, Department of Molecular and Cellular Pharmacology, School of Pharmaceutical Sciences, Peking University Health Science Center, Beijing, China
| | - Mala Shah
- UCL School of Pharmacy, University College London, London, UK
| | - Yuwu Jiang
- Department of Pediatrics, Peking University First Hospital, Beijing, China
- Beijing Key Laboratory of Molecular Diagnosis and Study on Pediatric Genetic Diseases, Beijing, China
- Children Epilepsy Center, Peking University First Hospital, Beijing, China
- Key Laboratory for Neuroscience, Ministry of Education/National Health and Family Planning Commission, Peking University, Beijing, 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, China
- IDG/McGovern Institute for Brain Research, Peking University, Beijing, China
| |
Collapse
|
6
|
Bertagna F, Ahmad S, Lewis R, Silva SRP, McFadden J, Huang CLH, Matthews HR, Jeevaratnam K. Loose-patch clamp analysis applied to voltage-gated ionic currents following pharmacological ryanodine receptor modulation in murine hippocampal cornu ammonis-1 pyramidal neurons. Front Physiol 2024; 15:1359560. [PMID: 38720787 PMCID: PMC11076846 DOI: 10.3389/fphys.2024.1359560] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2023] [Accepted: 04/04/2024] [Indexed: 05/12/2024] Open
Abstract
Introduction The loose-patch clamp technique was first developed and used in native amphibian skeletal muscle (SkM), offering useful features complementing conventional sharp micro-electrode, gap, or conventional patch voltage clamping. It demonstrated the feedback effects of pharmacological modification of ryanodine receptor (RyR)-mediated Ca2+ release on the Na+ channel (Nav1.4) currents, initiating excitation-contraction coupling in native murine SkM. The effects of the further RyR and Ca2+-ATPase (SERCA) antagonists, dantrolene and cyclopiazonic acid (CPA), additionally implicated background tubular-sarcoplasmic Ca2+ domains in these actions. Materials and methods We extend the loose-patch clamp approach to ion current measurements in murine hippocampal brain slice cornu ammonis-1 (CA1) pyramidal neurons. We explored the effects on Na+ currents of pharmacologically manipulating RyR and SERCA-mediated intracellular store Ca2+ release and reuptake. We adopted protocols previously applied to native skeletal muscle. These demonstrated Ca2+-mediated feedback effects on the Na+ channel function. Results Experiments applying depolarizing 15 ms duration loose-patch clamp steps to test voltages ranging from -40 to 120 mV positive to the resting membrane potential demonstrated that 0.5 mM caffeine decreased inward current amplitudes, agreeing with the previous SkM findings. It also decreased transient but not prolonged outward current amplitudes. However, 2 mM caffeine affected neither inward nor transient outward but increased prolonged outward currents, in contrast to its increasing inward currents in SkM. Furthermore, similarly and in contrast to previous SkM findings, both dantrolene (10 μM) and CPA (1 μM) pre-administration left both inward and outward currents unchanged. Nevertheless, dantrolene pretreatment still abrogated the effects of subsequent 0.5- and 2-mM caffeine challenges on both inward and outward currents. Finally, CPA abrogated the effects of 0.5 mM caffeine on both inward and outward currents, but with 2 mM caffeine, inward and transient outward currents were unchanged, but sustained outward currents increased. Conclusion We, thus, extend loose-patch clamping to establish pharmacological properties of murine CA1 pyramidal neurons and their similarities and contrasts with SkM. Here, evoked though not background Ca2+-store release influenced Nav and Kv excitation, consistent with smaller contributions of background store Ca2+ release to resting [Ca2+]. This potential non-canonical mechanism could modulate neuronal membrane excitability or cellular firing rates.
Collapse
Affiliation(s)
- Federico Bertagna
- Leverhulme Quantum Biology Doctoral Training Centre, University of Surrey, Guildford, United Kingdom
- School of Veterinary Medicine, Faculty of Health and Medical Sciences, University of Surrey, Guildford, United Kingdom
| | - Shiraz Ahmad
- School of Veterinary Medicine, Faculty of Health and Medical Sciences, University of Surrey, Guildford, United Kingdom
| | - Rebecca Lewis
- Leverhulme Quantum Biology Doctoral Training Centre, University of Surrey, Guildford, United Kingdom
- School of Veterinary Medicine, Faculty of Health and Medical Sciences, University of Surrey, Guildford, United Kingdom
| | - S. Ravi P. Silva
- Leverhulme Quantum Biology Doctoral Training Centre, University of Surrey, Guildford, United Kingdom
- Advanced Technology Institute, University of Surrey, Guildford, United Kingdom
| | - Johnjoe McFadden
- Leverhulme Quantum Biology Doctoral Training Centre, University of Surrey, Guildford, United Kingdom
- School of Biosciences and Medicine, Faculty of Health and Medical Sciences, University of Surrey, Guildford, United Kingdom
| | - Christopher L.-H. Huang
- School of Veterinary Medicine, Faculty of Health and Medical Sciences, University of Surrey, Guildford, United Kingdom
- Physiological Laboratory, University of Cambridge, Cambridge, United Kingdom
- Department of Biochemistry, University of Cambridge, Cambridge, United Kingdom
| | - Hugh R. Matthews
- Physiological Laboratory, University of Cambridge, Cambridge, United Kingdom
| | - Kamalan Jeevaratnam
- Leverhulme Quantum Biology Doctoral Training Centre, University of Surrey, Guildford, United Kingdom
- School of Veterinary Medicine, Faculty of Health and Medical Sciences, University of Surrey, Guildford, United Kingdom
| |
Collapse
|
7
|
Constantin S, Quignon C, Pizano K, Shostak DM, Wray S. Vasoactive intestinal peptide excites GnRH neurons via KCa3.1, a potential player in the slow afterhyperpolarization current. Front Cell Neurosci 2024; 18:1354095. [PMID: 38633445 PMCID: PMC11021707 DOI: 10.3389/fncel.2024.1354095] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2023] [Accepted: 03/05/2024] [Indexed: 04/19/2024] Open
Abstract
Vasoactive intestinal peptide (VIP) is an important component of the suprachiasmatic nucleus (SCN) which relays circadian information to neuronal populations, including GnRH neurons. Human and animal studies have shown an impact of disrupted daily rhythms (chronic shift work, temporal food restriction, clock gene disruption) on both male and female reproduction and fertility. To date, how VIP modulates GnRH neurons remains unknown. Calcium imaging and electrophysiology on primary GnRH neurons in explants and adult mouse brain slice, respectively, were used to address this question. We found VIP excites GnRH neurons via the VIP receptor, VPAC2. The downstream signaling pathway uses both Gs protein/adenylyl cyclase/protein kinase A (PKA) and phospholipase C/phosphatidylinositol 4,5-bisphosphate (PIP2) depletion. Furthermore, we identified a UCL2077-sensitive target, likely contributing to the slow afterhyperpolarization current (IAHP), as the PKA and PIP2 depletion target, and the KCa3.1 channel as a specific target. Thus, VIP/VPAC2 provides an example of Gs protein-coupled receptor-triggered excitation in GnRH neurons, modulating GnRH neurons likely via the slow IAHP. The possible identification of KCa3.1 in the GnRH neuron slow IAHP may provide a new therapeutical target for fertility treatments.
Collapse
Affiliation(s)
| | | | | | | | - Susan Wray
- Cellular and Developmental Neurobiology Section, National Institute of Neurological Disorders and Stroke/National Institutes of Health, Bethesda, MD, United States
| |
Collapse
|
8
|
Van NTH, Kim WK, Nam JH. Challenges in the Therapeutic Targeting of KCa Channels: From Basic Physiology to Clinical Applications. Int J Mol Sci 2024; 25:2965. [PMID: 38474212 PMCID: PMC10932353 DOI: 10.3390/ijms25052965] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2023] [Revised: 01/23/2024] [Accepted: 01/25/2024] [Indexed: 03/14/2024] Open
Abstract
Calcium-activated potassium (KCa) channels are ubiquitously expressed throughout the body and are able to regulate membrane potential and intracellular calcium concentrations, thereby playing key roles in cellular physiology and signal transmission. Consequently, it is unsurprising that KCa channels have been implicated in various diseases, making them potential targets for pharmaceutical interventions. Over the past two decades, numerous studies have been conducted to develop KCa channel-targeting drugs, including those for disorders of the central and peripheral nervous, cardiovascular, and urinary systems and for cancer. In this review, we synthesize recent findings regarding the structure and activating mechanisms of KCa channels. We also discuss the role of KCa channel modulators in therapeutic medicine. Finally, we identify the major reasons behind the delay in bringing these modulators to the pharmaceutical market and propose new strategies to promote their application.
Collapse
Affiliation(s)
- Nhung Thi Hong Van
- Department of Physiology, Dongguk University College of Medicine, Gyeongju 38066, Republic of Korea;
- Channelopathy Research Center (CRC), Dongguk University College of Medicine, Goyang 10326, Republic of Korea
| | - Woo Kyung Kim
- Channelopathy Research Center (CRC), Dongguk University College of Medicine, Goyang 10326, Republic of Korea
- Department of Internal Medicine, Graduate School of Medicine, Dongguk University, Goyang 10326, Republic of Korea
| | - Joo Hyun Nam
- Department of Physiology, Dongguk University College of Medicine, Gyeongju 38066, Republic of Korea;
- Channelopathy Research Center (CRC), Dongguk University College of Medicine, Goyang 10326, Republic of Korea
| |
Collapse
|
9
|
Nikitin ES, Postnikova TY, Proskurina EY, Borodinova AA, Ivanova V, Roshchin MV, Smirnova MP, Kelmanson I, Belousov VV, Balaban PM, Zaitsev AV. Overexpression of KCNN4 channels in principal neurons produces an anti-seizure effect without reducing their coding ability. Gene Ther 2024; 31:144-153. [PMID: 37968509 DOI: 10.1038/s41434-023-00427-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2023] [Revised: 10/13/2023] [Accepted: 10/24/2023] [Indexed: 11/17/2023]
Abstract
Gene therapy offers a potential alternative to the surgical treatment of epilepsy, which affects millions of people and is pharmacoresistant in ~30% of cases. Aimed at reducing the excitability of principal neurons, the engineered expression of K+ channels has been proposed as a treatment due to the outstanding ability of K+ channels to hyperpolarize neurons. However, the effects of K+ channel overexpression on cell physiology remain to be investigated. Here we report an adeno-associated virus (AAV) vector designed to reduce epileptiform activity specifically in excitatory pyramidal neurons by expressing the human Ca2+-gated K+ channel KCNN4 (KCa3.1). Electrophysiological and pharmacological experiments in acute brain slices showed that KCNN4-transduced cells exhibited a Ca2+-dependent slow afterhyperpolarization that significantly decreased the ability of KCNN4-positive neurons to generate high-frequency spike trains without affecting their lower-frequency coding ability and action potential shapes. Antiepileptic activity tests showed potent suppression of pharmacologically induced seizures in vitro at both single cell and local field potential levels with decreased spiking during ictal discharges. Taken together, our findings strongly suggest that the AAV-based expression of the KCNN4 channel in excitatory neurons is a promising therapeutic intervention as gene therapy for epilepsy.
Collapse
Affiliation(s)
- Evgeny S Nikitin
- Institute of Higher Nervous Activity and Neurophysiology, RAS, 117485, Moscow, Russia.
| | - Tatiana Y Postnikova
- Sechenov Institute of Evolutionary Physiology and Biochemistry RAS, 194223, Saint Petersburg, Russia
| | - Elena Y Proskurina
- Sechenov Institute of Evolutionary Physiology and Biochemistry RAS, 194223, Saint Petersburg, Russia
| | | | - Violetta Ivanova
- Institute of Higher Nervous Activity and Neurophysiology, RAS, 117485, Moscow, Russia
| | - Matvey V Roshchin
- Institute of Higher Nervous Activity and Neurophysiology, RAS, 117485, Moscow, Russia
| | - Maria P Smirnova
- Institute of Higher Nervous Activity and Neurophysiology, RAS, 117485, Moscow, Russia
| | - Ilya Kelmanson
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry RAS, 117997, Moscow, Russia
- Center for Precision Genome Editing and Genetic Technologies for Biomedicine, Pirogov Russian National Research Medical University, 117997, Moscow, Russia
| | - Vsevolod V Belousov
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry RAS, 117997, Moscow, Russia
- Center for Precision Genome Editing and Genetic Technologies for Biomedicine, Pirogov Russian National Research Medical University, 117997, Moscow, Russia
- Federal Center of Brain Research and Neurotechnologies, Federal Medical Biological Agency, 117997, Moscow, Russia
- Life Improvement by Future Technologies (LIFT) Center, 143025, Moscow, Russia
| | - Pavel M Balaban
- Institute of Higher Nervous Activity and Neurophysiology, RAS, 117485, Moscow, Russia
| | - Aleksey V Zaitsev
- Sechenov Institute of Evolutionary Physiology and Biochemistry RAS, 194223, Saint Petersburg, Russia.
| |
Collapse
|
10
|
Charlick JN, Bozadzhieva D, Butler AS, Wilkinson KA, Marrion NV. A single coiled-coil domain mutation in hIKCa channel subunits disrupts preferential formation of heteromeric hSK1:hIKCa channels. Eur J Neurosci 2024; 59:3-16. [PMID: 38018635 PMCID: PMC10952195 DOI: 10.1111/ejn.16189] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2023] [Revised: 09/22/2023] [Accepted: 10/25/2023] [Indexed: 11/30/2023]
Abstract
The expression of IKCa (SK4) channel subunits overlaps with that of SK channel subunits, and it has been proposed that the two related subunits prefer to co-assemble to form heteromeric hSK1:hIKCa channels. This implicates hSK1:hIKCa heteromers in physiological roles that might have been attributed to activation of SK channels. We have used a mutation approach to confirm formation of heterometric hSK1:hIKCa channels. Introduction of residues within hSK1 that were predicted to impart sensitivity to the hIKCa current blocker TRAM-34 changed the pharmacology of functional heteromers. Heteromeric channels formed between wildtype hIKCa and mutant hSK1 subunits displayed a significantly higher sensitivity and maximum block to addition of TRAM-34 than heteromers formed between wildtype subunits. Heteromer formation was disrupted by a single point mutation within one COOH-terminal coiled-coil domain of the hIKCa channel subunit. This mutation only disrupted the formation of hSK1:hIKCa heteromeric channels, without affecting the formation of homomeric hIKCa channels. Finally, the Ca2+ gating sensitivity of heteromeric hSK1:hIKCa channels was found to be significantly lower than the Ca2+ gating sensitivity of homomeric hIKCa channels. These data confirmed the preferred formation of heteromeric channels that results from COOH-terminal interactions between subunits. The distinct sensitivity of the heteromer to activation by Ca2+ suggests that heteromeric channels fulfil a distinct function within those neurons that express both subunits.
Collapse
Affiliation(s)
- James N. Charlick
- School of Physiology, Pharmacology and NeuroscienceUniversity of BristolBristolUK
| | - Daniella Bozadzhieva
- School of Physiology, Pharmacology and NeuroscienceUniversity of BristolBristolUK
| | - Andrew S. Butler
- School of Physiology, Pharmacology and NeuroscienceUniversity of BristolBristolUK
| | - Kevin A. Wilkinson
- School of Physiology, Pharmacology and NeuroscienceUniversity of BristolBristolUK
| | - Neil V. Marrion
- School of Physiology, Pharmacology and NeuroscienceUniversity of BristolBristolUK
| |
Collapse
|
11
|
Viteri JA, Schulz DJ. Motor neurons within a network use cell-type specific feedback mechanisms to constrain relationships among ion channel mRNAs. J Neurophysiol 2023; 130:569-584. [PMID: 37529838 PMCID: PMC11550874 DOI: 10.1152/jn.00098.2023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2023] [Revised: 07/27/2023] [Accepted: 07/29/2023] [Indexed: 08/03/2023] Open
Abstract
Recently, activity has been proposed as a primary feedback mechanism used by continuously bursting neurons to coordinate ion channel mRNA relationships that underlie stable output. However, some neuron types only have intermittent periods of activity and so may require alternative mechanisms that induce and constrain the appropriate ion channel profile in different states of activity. To address this, we used the pyloric dilator (PD; constitutively active) and the lateral gastric (LG; periodically active) neurons of the stomatogastric ganglion (STG) of the crustacean Cancer borealis. We experimentally stimulated descending inputs to the STG to cause release of neuromodulators known to elicit the active state of LG neurons and quantified the mRNA abundances and pairwise relationships of 11 voltage-gated ion channels in active and silent LG neurons. The same stimulus does not significantly alter PD activity. Activation of LG upregulated ion channel mRNAs and lead to a greater number of positively correlated pairwise channel mRNA relationships. Conversely, this stimulus did not induce major changes in ion channel mRNA abundances and relationships of PD cells, suggesting their ongoing activity is sufficient to maintain channel mRNA relationships even under changing modulatory conditions. In addition, we found that ion channel mRNA correlations induced by the active state of LG are influenced by a combination of activity- and neuromodulator-dependent feedback mechanisms. Interestingly, some of these same correlations are maintained by distinct mechanisms in PD, suggesting that these motor networks use distinct feedback mechanisms to coordinate the same mRNA relationships across neuron types.NEW & NOTEWORTHY Neurons use various feedback mechanisms to adjust and maintain their output. Here, we demonstrate that different neurons within the same network can use distinct signaling mechanisms to regulate the same ion channel mRNA relationships.
Collapse
Affiliation(s)
- Jose A Viteri
- Division of Biological Sciences, University of Missouri-Columbia, Columbia, Missouri, United States
| | - David J Schulz
- Division of Biological Sciences, University of Missouri-Columbia, Columbia, Missouri, United States
| |
Collapse
|
12
|
Maziar A, Critch TNRHY, Ghosh S, Rajani V, Flynn CM, Qin T, Reinhardt C, Man KNM, Lee A, Hell JW, Yuan Q. Aging differentially affects LTCC function in hippocampal CA1 and piriform cortex pyramidal neurons. Cereb Cortex 2023; 33:1489-1503. [PMID: 35437602 PMCID: PMC9930631 DOI: 10.1093/cercor/bhac152] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2022] [Revised: 03/25/2022] [Accepted: 03/27/2022] [Indexed: 11/14/2022] Open
Abstract
Aging is associated with cognitive decline and memory loss in humans. In rats, aging-associated neuronal excitability changes and impairments in learning have been extensively studied in the hippocampus. Here, we investigated the roles of L-type calcium channels (LTCCs) in the rat piriform cortex (PC), in comparison with those of the hippocampus. We employed spatial and olfactory tasks that involve the hippocampus and PC. LTCC blocker nimodipine administration impaired spontaneous location recognition in adult rats (6-9 months). However, the same blocker rescued the spatial learning deficiency in aged rats (19-23 months). In an odor-associative learning task, infusions of nimodipine into either the PC or dorsal CA1 impaired the ability of adult rats to learn a positive odor association. Again, in contrast, nimodipine rescued odor associative learning in aged rats. Aged CA1 neurons had higher somatic expression of LTCC Cav1.2 subunits, exhibited larger afterhyperpolarization (AHP) and lower excitability compared with adult neurons. In contrast, PC neurons from aged rats showed higher excitability and no difference in AHP. Cav1.2 expression was similar in adult and aged PC somata, but relatively higher in PSD95- puncta in aged dendrites. Our data suggest unique features of aging-associated changes in LTCCs in the PC and hippocampus.
Collapse
Affiliation(s)
- Aida Maziar
- Division of Biomedical Sciences, Faculty of Medicine, Memorial University, St. John's, NL A1B 3V6, Canada
| | - Tristian N R H Y Critch
- Division of Biomedical Sciences, Faculty of Medicine, Memorial University, St. John's, NL A1B 3V6, Canada
| | - Sourav Ghosh
- Division of Biomedical Sciences, Faculty of Medicine, Memorial University, St. John's, NL A1B 3V6, Canada
| | - Vishaal Rajani
- Division of Biomedical Sciences, Faculty of Medicine, Memorial University, St. John's, NL A1B 3V6, Canada
| | - Cassandra M Flynn
- Division of Biomedical Sciences, Faculty of Medicine, Memorial University, St. John's, NL A1B 3V6, Canada
| | - Tian Qin
- Division of Biomedical Sciences, Faculty of Medicine, Memorial University, St. John's, NL A1B 3V6, Canada
| | - Camila Reinhardt
- Division of Biomedical Sciences, Faculty of Medicine, Memorial University, St. John's, NL A1B 3V6, Canada
| | - Kwun Nok Mimi Man
- Department of Pharmacology, School of Medicine, University of California-Davis, Sacramento, CA 95817, United States
| | - Amy Lee
- Department of Neuroscience, University of Texas-Austin, Austin, TX 78712, United States
| | - Johannes W Hell
- Department of Pharmacology, School of Medicine, University of California-Davis, Sacramento, CA 95817, United States
| | - Qi Yuan
- Division of Biomedical Sciences, Faculty of Medicine, Memorial University, St. John's, NL A1B 3V6, Canada
| |
Collapse
|
13
|
Lin Y, Zhao YJ, Zhang HL, Hao WJ, Zhu RD, Wang Y, Hu W, Zhou RP. Regulatory role of KCa3.1 in immune cell function and its emerging association with rheumatoid arthritis. Front Immunol 2022; 13:997621. [PMID: 36275686 PMCID: PMC9580404 DOI: 10.3389/fimmu.2022.997621] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2022] [Accepted: 09/16/2022] [Indexed: 11/25/2022] Open
Abstract
Rheumatoid arthritis (RA) is a common autoimmune disease characterized by chronic inflammation. Immune dysfunction is an essential mechanism in the pathogenesis of RA and directly linked to synovial inflammation and cartilage/bone destruction. Intermediate conductance Ca2+-activated K+ channel (KCa3.1) is considered a significant regulator of proliferation, differentiation, and migration of immune cells by mediating Ca2+ signal transduction. Earlier studies have demonstrated abnormal activation of KCa3.1 in the peripheral blood and articular synovium of RA patients. Moreover, knockout of KCa3.1 reduced the severity of synovial inflammation and cartilage damage to a significant extent in a mouse collagen antibody-induced arthritis (CAIA) model. Accumulating evidence implicates KCa3.1 as a potential therapeutic target for RA. Here, we provide an overview of the KCa3.1 channel and its pharmacological properties, discuss the significance of KCa3.1 in immune cells and feasibility as a drug target for modulating the immune balance, and highlight its emerging role in pathological progression of RA.
Collapse
Affiliation(s)
- Yi Lin
- Department of Clinical Pharmacology, The Second Hospital of Anhui Medical University, Hefei, China
- Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Anhui Institute of Innovative Drugs, School of Pharmacy, Anhui Medical University, Hefei, China
| | - Ying-Jie Zhao
- Department of Clinical Pharmacology, The Second Hospital of Anhui Medical University, Hefei, China
| | - Hai-Lin Zhang
- Department of Clinical Pharmacology, The Second Hospital of Anhui Medical University, Hefei, China
- Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Anhui Institute of Innovative Drugs, School of Pharmacy, Anhui Medical University, Hefei, China
| | - Wen-Juan Hao
- Department of Clinical Pharmacology, The Second Hospital of Anhui Medical University, Hefei, China
- Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Anhui Institute of Innovative Drugs, School of Pharmacy, Anhui Medical University, Hefei, China
| | - Ren-Di Zhu
- Department of Clinical Pharmacology, The Second Hospital of Anhui Medical University, Hefei, China
- Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Anhui Institute of Innovative Drugs, School of Pharmacy, Anhui Medical University, Hefei, China
| | - Yan Wang
- Department of Clinical Pharmacology, The Second Hospital of Anhui Medical University, Hefei, China
- Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Anhui Institute of Innovative Drugs, School of Pharmacy, Anhui Medical University, Hefei, China
| | - Wei Hu
- Department of Clinical Pharmacology, The Second Hospital of Anhui Medical University, Hefei, China
- The Key Laboratory of Anti-inflammatory and Immune Medicine, Ministry of Education, Anhui Medical University, Hefei, China
- *Correspondence: Wei Hu, ; Ren-Peng Zhou,
| | - Ren-Peng Zhou
- Department of Clinical Pharmacology, The Second Hospital of Anhui Medical University, Hefei, China
- The Key Laboratory of Anti-inflammatory and Immune Medicine, Ministry of Education, Anhui Medical University, Hefei, China
- *Correspondence: Wei Hu, ; Ren-Peng Zhou,
| |
Collapse
|
14
|
Awasthi R, Chandra N, Barkai E. Olfactory rule learning-induced enhancement in intrinsic neuronal excitability is maintained by shutdown of the cholinergic M-current. Front Cell Neurosci 2022; 16:934838. [PMID: 36246520 PMCID: PMC9556983 DOI: 10.3389/fncel.2022.934838] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2022] [Accepted: 09/08/2022] [Indexed: 11/17/2022] Open
Abstract
Training rats in a particularly difficult olfactory discrimination task initiates a period of accelerated learning, manifested as a dramatic increase in the rats' capacity to discriminate between pairs of odors once they have learned the discrimination task, implying that rule learning has taken place. At the cellular biophysical level, rule learning is maintained by reduction in the conductance of the slow current (sIAHP) simultaneously in most piriform cortex layer II pyramidal neurons. Such sIAHP reduction is expressed in attenuation of the post-burst afterhyperpolarization (AHP) potential and thus in enhanced repetitive action potential firing. Previous studies have shown that a causal relationship exists between long-lasting post-burst AHP reduction and rule learning. A specific channel through which the sIAHP flows has not been identified. The sIAHP in pyramidal cells is critically dependent on membrane phosphatidylinositol 4,5-bisphosphate [PtdIns(4,5)P(2)]. PtdIns(4,5)P(2) regulates the calcium sensitivity of the sIAHP by acting downstream from the rise in intracellular calcium. These findings led to the interesting hypothesis that PtdIns(4,5)P(2) activates a variety of potassium channels. Thus, the sIAHP would not represent a unitary ionic current but the embodiment of a generalized potassium channel gating mechanism. We thus hypothesized that the learning-induced increase in intrinsic excitability is mediated by reduced conductance of one or more of the currents that contribute to the sIAHP. Here we first show, using current-clamp recordings, that the post-burst AHP in piriform cortex pyramidal neurons is also mediated by the Ih, and the contribution of this current to the post-burst AHP is also affected by learning. We also show, using whole-cell patch-clamp recordings, that the sIAHP in neurons from trained rats is not sensitive to blocking membrane phosphatidylinositol 4,5-bisphosphate [PtdIns(4,5)P(2)], and to the blocking of the current mediated by the cholinergic muscarinic acetylcholine receptor (M-current). Further current-clamp recordings also show that blocking PtdIns(4,5)P(2) synthesis and application of a specific IKCa blocker have no effect on the post-burst AHP in neurons from trained as well as control rats. Taken together with results from our previous studies, these data suggest that rule learning-induced long-lasting enhancement in intrinsic neuronal excitability results from reduced conductance of the M-current and thus the slow potassium currents, which control repetitive spike firing.
Collapse
Affiliation(s)
| | | | - Edi Barkai
- Sagol Department of Neurobiology, Faculty of Natural Sciences, University of Haifa, Haifa, Israel
| |
Collapse
|
15
|
Tau isoform-specific enhancement of L-type calcium current and augmentation of afterhyperpolarization in rat hippocampal neurons. Sci Rep 2022; 12:15231. [PMID: 36075936 PMCID: PMC9458744 DOI: 10.1038/s41598-022-18648-0] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2022] [Accepted: 08/17/2022] [Indexed: 11/08/2022] Open
Abstract
Accumulation of tau is observed in dementia, with human tau displaying 6 isoforms grouped by whether they display either 3 or 4 C-terminal repeat domains (3R or 4R) and exhibit no (0N), one (1N) or two (2N) N terminal repeats. Overexpression of 4R0N-tau in rat hippocampal slices enhanced the L-type calcium (Ca2+) current-dependent components of the medium and slow afterhyperpolarizations (AHPs). Overexpression of both 4R0N-tau and 4R2N-tau augmented CaV1.2-mediated L-type currents when expressed in tsA-201 cells, an effect not observed with the third 4R isoform, 4R1N-tau. Current enhancement was only observed when the pore-forming subunit was co-expressed with CaVβ3 and not CaVβ2a subunits. Non-stationary noise analysis indicated that enhanced Ca2+ channel current arose from a larger number of functional channels. 4R0N-tau and CaVβ3 were found to be physically associated by co-immunoprecipitation. In contrast, the 4R1N-tau isoform that did not augment expressed macroscopic L-type Ca2+ current exhibited greatly reduced binding to CaVβ3. These data suggest that physical association between tau and the CaVβ3 subunit stabilises functional L-type channels in the membrane, increasing channel number and Ca2+ influx. Enhancing the Ca2+-dependent component of AHPs would produce cognitive impairment that underlie those seen in the early phases of tauopathies.
Collapse
|
16
|
Allosteric inhibitors targeting the calmodulin-PIP2 interface of SK4 K + channels for atrial fibrillation treatment. Proc Natl Acad Sci U S A 2022; 119:e2202926119. [PMID: 35969786 PMCID: PMC9407317 DOI: 10.1073/pnas.2202926119] [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] [Indexed: 11/18/2022] Open
Abstract
The Ca2+-activated SK4 K+ channel is gated by Ca2+-calmodulin (CaM) and is expressed in immune cells, brain, and heart. A cryoelectron microscopy (cryo-EM) structure of the human SK4 K+ channel recently revealed four CaM molecules per channel tetramer, where the apo CaM C-lobe and the holo CaM N-lobe interact with the proximal carboxyl terminus and the linker S4-S5, respectively, to gate the channel. Here, we show that phosphatidylinositol 4-5 bisphosphate (PIP2) potently activates SK4 channels by docking to the boundary of the CaM-binding domain. An allosteric blocker, BA6b9, was designed to act to the CaM-PIP2-binding domain, a previously untargeted region of SK4 channels, at the interface of the proximal carboxyl terminus and the linker S4-S5. Site-directed mutagenesis, molecular docking, and patch-clamp electrophysiology indicate that BA6b9 inhibits SK4 channels by interacting with two specific residues, Arg191 and His192 in the linker S4-S5, not conserved in SK1-SK3 subunits, thereby conferring selectivity and preventing the Ca2+-CaM N-lobe from properly interacting with the channel linker region. Immunohistochemistry of the SK4 channel protein in rat hearts showed a widespread expression in the sarcolemma of atrial myocytes, with a sarcomeric striated Z-band pattern, and a weaker occurrence in the ventricle but a marked incidence at the intercalated discs. BA6b9 significantly prolonged atrial and atrioventricular effective refractory periods in rat isolated hearts and reduced atrial fibrillation induction ex vivo. Our work suggests that inhibition of SK4 K+ channels by targeting drugs to the CaM-PIP2-binding domain provides a promising anti-arrhythmic therapy.
Collapse
|
17
|
Ierusalimsky VN, Balaban PM, Nikitin ES. Nav1.6 but not KCa3.1 channels contribute to heterogeneity in coding abilities and dynamics of action potentials in the L5 neocortical pyramidal neurons. Biochem Biophys Res Commun 2022; 615:102-108. [DOI: 10.1016/j.bbrc.2022.05.050] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2022] [Accepted: 05/14/2022] [Indexed: 12/16/2022]
|
18
|
Nikitin ES, Balaban PM, Zaitsev AV. Prospects for Gene Therapy of Epilepsy Using Calcium-Acivated Potassium Channel Vectors. J EVOL BIOCHEM PHYS+ 2022. [DOI: 10.1134/s0022093022040111] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
|
19
|
Ion Channel Partnerships: Odd and Not-So-Odd Couples Controlling Neuronal Ion Channel Function. Int J Mol Sci 2022; 23:ijms23041953. [PMID: 35216068 PMCID: PMC8878034 DOI: 10.3390/ijms23041953] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2022] [Revised: 02/05/2022] [Accepted: 02/08/2022] [Indexed: 12/04/2022] Open
Abstract
The concerted function of the large number of ion channels expressed in excitable cells, including brain neurons, shapes diverse signaling events by controlling the electrical properties of membranes. It has long been recognized that specific groups of ion channels are functionally coupled in mediating ionic fluxes that impact membrane potential, and that these changes in membrane potential impact ion channel gating. Recent studies have identified distinct sets of ion channels that can also physically and functionally associate to regulate the function of either ion channel partner beyond that afforded by changes in membrane potential alone. Here, we review canonical examples of such ion channel partnerships, in which a Ca2+ channel is partnered with a Ca2+-activated K+ channel to provide a dedicated route for efficient coupling of Ca2+ influx to K+ channel activation. We also highlight examples of non-canonical ion channel partnerships between Ca2+ channels and voltage-gated K+ channels that are not intrinsically Ca2+ sensitive, but whose partnership nonetheless yields enhanced regulation of one or the other ion channel partner. We also discuss how these ion channel partnerships can be shaped by the subcellular compartments in which they are found and provide perspectives on how recent advances in techniques to identify proteins in close proximity to one another in native cells may lead to an expanded knowledge of other ion channel partnerships.
Collapse
|
20
|
Klemz A, Wildner F, Tütüncü E, Gerevich Z. Regulation of Hippocampal Gamma Oscillations by Modulation of Intrinsic Neuronal Excitability. Front Neural Circuits 2022; 15:778022. [PMID: 35177966 PMCID: PMC8845518 DOI: 10.3389/fncir.2021.778022] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2021] [Accepted: 12/21/2021] [Indexed: 11/16/2022] Open
Abstract
Ion channels activated around the subthreshold membrane potential determine the likelihood of neuronal firing in response to synaptic inputs, a process described as intrinsic neuronal excitability. Long-term plasticity of chemical synaptic transmission is traditionally considered the main cellular mechanism of information storage in the brain; however, voltage- and calcium-activated channels modulating the inputs or outputs of neurons are also subjects of plastic changes and play a major role in learning and memory formation. Gamma oscillations are associated with numerous higher cognitive functions such as learning and memory, but our knowledge of their dependence on intrinsic plasticity is by far limited. Here we investigated the roles of potassium and calcium channels activated at near subthreshold membrane potentials in cholinergically induced persistent gamma oscillations measured in the CA3 area of rat hippocampal slices. Among potassium channels, which are responsible for the afterhyperpolarization in CA3 pyramidal cells, we found that blockers of SK (KCa2) and KV7.2/7.3 (KCNQ2/3), but not the BK (KCa1.1) and IK (KCa3.1) channels, increased the power of gamma oscillations. On the contrary, activators of these channels had an attenuating effect without affecting the frequency. Pharmacological blockade of the low voltage-activated T-type calcium channels (CaV3.1–3.3) reduced gamma power and increased the oscillation peak frequency. Enhancement of these channels also inhibited the peak power without altering the frequency of the oscillations. The presented data suggest that voltage- and calcium-activated ion channels involved in intrinsic excitability strongly regulate the power of hippocampal gamma oscillations. Targeting these channels could represent a valuable pharmacological strategy against cognitive impairment.
Collapse
|
21
|
Sahu G, Turner RW. The Molecular Basis for the Calcium-Dependent Slow Afterhyperpolarization in CA1 Hippocampal Pyramidal Neurons. Front Physiol 2022; 12:759707. [PMID: 35002757 PMCID: PMC8730529 DOI: 10.3389/fphys.2021.759707] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2021] [Accepted: 11/01/2021] [Indexed: 12/02/2022] Open
Abstract
Neuronal signal transmission depends on the frequency, pattern, and timing of spike output, each of which are shaped by spike afterhyperpolarizations (AHPs). There are classically three post-spike AHPs of increasing duration categorized as fast, medium and slow AHPs that hyperpolarize a cell over a range of 10 ms to 30 s. Intensive early work on CA1 hippocampal pyramidal cells revealed that all three AHPs incorporate activation of calcium-gated potassium channels. The ionic basis for a fAHP was rapidly attributed to the actions of big conductance (BK) and the mAHP to small conductance (SK) or Kv7 potassium channels. In stark contrast, the ionic basis for a prominent slow AHP of up to 30 s duration remained an enigma for over 30 years. Recent advances in pharmacological, molecular, and imaging tools have uncovered the expression of a calcium-gated intermediate conductance potassium channel (IK, KCa3.1) in central neurons that proves to contribute to the slow AHP in CA1 hippocampal pyramidal cells. Together the data show that the sAHP arises in part from a core tripartite complex between Cav1.3 (L-type) calcium channels, ryanodine receptors, and IK channels at endoplasmic reticulum-plasma membrane junctions. Work on the sAHP in CA1 pyramidal neurons has again quickened pace, with identified contributions by both IK channels and the Na-K pump providing answers to several mysteries in the pharmacological properties of the sAHP.
Collapse
Affiliation(s)
- Giriraj Sahu
- National Institute of Pharmaceutical Education and Research Ahmedabad, Ahmedabad, India
| | - Ray W Turner
- Department Cell Biology & Anatomy, Cumming School of Medicine, Hotchkiss Brain Institute, University of Calgary, Calgary, AB, Canada
| |
Collapse
|
22
|
Laker D, Tolle F, Stegen M, Heerdegen M, Köhling R, Kirschstein T, Wolfart J. K v7 and K ir6 Channels Shape the Slow AHP in Mouse Dentate Gyrus Granule Cells and Control Burst-like Firing Behavior. Neuroscience 2021; 467:56-72. [PMID: 34048798 DOI: 10.1016/j.neuroscience.2021.05.025] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2020] [Revised: 05/18/2021] [Accepted: 05/19/2021] [Indexed: 11/29/2022]
Abstract
The slow afterhyperpolarizing potential (sAHP) can silence a neuron for hundreds of milliseconds. Thereby, the sAHP determines the discharge behavior of many types of neurons. In dentate granule cells (DGCs), serving as a filter into the hippocampal network, mostly tonic or adapting discharge properties have been described. As under standard whole-cell recording conditions the sAHP is inhibited, we reevaluated the intrinsic functional phenotype of DGCs and the conductances underlying the sAHP, using gramicidine-perforated patch-clamp technique. We found that in 97/113 (86%) of the DGCs, a burst of action potentials (APs) to excitation ended by a large sAHP, despite continued depolarization. This result suggests that burst-like firing is the default functional phenotype of DGCs and that sAHPs are important for it. Indeed, burst-like firing DGCs showed a significantly higher sAHP-current (IsAHP) amplitude compared to spike-frequency adapting cells (16/113 = 14%). The IsAHP was mediated by Kv7 and Kir6 channels by pharmacological inhibition using XE991 and tolbutamide, although heterogeneously among DGCs. The percent inhibition of IsAHP by these compounds also correlated with the AP number and AP burst length. Application of 100 µM nickel after XE991 and tolbutamide detected a third conductance contributing to burst-like firing and the sAHP, most likely mediated by T-type calcium channels. Lastly, medial perforant path-dentate gyrus long-term potentiation was amplified by XE991 and tolbutamide. In conclusion, the sAHP shapes intrinsic burst-like firing which, under physiological circumstances, could be controlled via cholinergic afferents and ATP metabolism.
Collapse
Affiliation(s)
- Debora Laker
- Oscar Langendorff Institute of Physiology, University Medicine Rostock, Rostock, Germany
| | - Frederik Tolle
- Oscar Langendorff Institute of Physiology, University Medicine Rostock, Rostock, Germany
| | - Michael Stegen
- Department of Neurosurgery, University of Freiburg, Germany
| | - Marco Heerdegen
- Oscar Langendorff Institute of Physiology, University Medicine Rostock, Rostock, Germany
| | - Rüdiger Köhling
- Oscar Langendorff Institute of Physiology, University Medicine Rostock, Rostock, Germany
| | - Timo Kirschstein
- Oscar Langendorff Institute of Physiology, University Medicine Rostock, Rostock, Germany.
| | - Jakob Wolfart
- Oscar Langendorff Institute of Physiology, University Medicine Rostock, Rostock, Germany
| |
Collapse
|
23
|
Bacon TJ, Pickering AE, Mellor JR. Noradrenaline Release from Locus Coeruleus Terminals in the Hippocampus Enhances Excitation-Spike Coupling in CA1 Pyramidal Neurons Via β-Adrenoceptors. Cereb Cortex 2020; 30:6135-6151. [PMID: 32607551 PMCID: PMC7609922 DOI: 10.1093/cercor/bhaa159] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2020] [Revised: 05/13/2020] [Accepted: 05/13/2020] [Indexed: 12/29/2022] Open
Abstract
Release of the neuromodulator noradrenaline signals salience during wakefulness, flagging novel or important experiences to reconfigure information processing and memory representations in the hippocampus. Noradrenaline is therefore expected to enhance hippocampal responses to synaptic input; however, noradrenergic agonists have been found to have mixed and sometimes contradictory effects on Schaffer collateral synapses and the resulting CA1 output. Here, we examine the effects of endogenous, optogenetically driven noradrenaline release on synaptic transmission and spike output in mouse hippocampal CA1 pyramidal neurons. We show that endogenous noradrenaline release enhances the probability of CA1 pyramidal neuron spiking without altering feedforward excitatory or inhibitory synaptic inputs in the Schaffer collateral pathway. β-adrenoceptors mediate this enhancement of excitation-spike coupling by reducing the charge required to initiate action potentials, consistent with noradrenergic modulation of voltage-gated potassium channels. Furthermore, we find the likely effective concentration of endogenously released noradrenaline is sub-micromolar. Surprisingly, although comparable concentrations of exogenous noradrenaline cause robust depression of slow afterhyperpolarization currents, endogenous release of noradrenaline does not, indicating that endogenous noradrenaline release is targeted to specific cellular locations. These findings provide a mechanism by which targeted endogenous release of noradrenaline can enhance information transfer in the hippocampus in response to salient events.
Collapse
Affiliation(s)
- Travis J Bacon
- Centre for Synaptic Plasticity, University of Bristol, Bristol, UK
- School of Physiology, Pharmacology & Neuroscience, University of Bristol, Bristol BS8 1TD, UK
| | - Anthony E Pickering
- School of Physiology, Pharmacology & Neuroscience, University of Bristol, Bristol BS8 1TD, UK
- Bristol Anaesthesia, Pain & Critical Care Sciences, Translational Health Sciences, Bristol Medical School, University of Bristol, Bristol BS2 8HW, UK
| | - Jack R Mellor
- Centre for Synaptic Plasticity, University of Bristol, Bristol, UK
- School of Physiology, Pharmacology & Neuroscience, University of Bristol, Bristol BS8 1TD, UK
| |
Collapse
|
24
|
Severin D, Gallagher M, Kirkwood A. Afterhyperpolarization amplitude in CA1 pyramidal cells of aged Long-Evans rats characterized for individual differences. Neurobiol Aging 2020; 96:43-48. [PMID: 32932137 DOI: 10.1016/j.neurobiolaging.2020.07.022] [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: 04/09/2020] [Revised: 07/06/2020] [Accepted: 07/25/2020] [Indexed: 11/18/2022]
Abstract
Altered neural excitability is considered a prominent contributing factor to cognitive decline during aging. A clear example is the excess neural activity observed in several temporal lobe structures of cognitively impaired older individuals in rodents and humans. At a cellular level, aging-related changes in mechanisms regulating intrinsic excitability have been well examined in pyramidal cells of the CA1 hippocampal subfield. Studies in the inbred Fisher 344 rat strain document an age-related increase in the slow afterhyperpolarization (AHP) that normally occurs after a burst of action potentials, and serves to reduce subsequent firing. We evaluated the status of the AHP in the outbred Long-Evans rat, a well-established model for studying individual differences in neurocognitive aging. In contrast to the findings reported in the Fisher 344 rats, in the Long-Evan rats we detected a selective reduction in AHP in cognitively impaired aged individuals. We discuss plausible scenarios to account for these differences and also discuss possible implications of these differences.
Collapse
Affiliation(s)
- Daniel Severin
- Department of Neurosciences, Mind/Brain Institute, Johns Hopkins University, Baltimore, MD, USA
| | - Michela Gallagher
- Department of Psychological and Brain Sciences, Johns Hopkins University, Baltimore, MD, USA
| | - Alfredo Kirkwood
- Department of Neurosciences, Mind/Brain Institute, Johns Hopkins University, Baltimore, MD, USA.
| |
Collapse
|
25
|
Pousinha PA, Mouska X, Bianchi D, Temido-Ferreira M, Rajão-Saraiva J, Gomes R, Fernandez SP, Salgueiro-Pereira AR, Gandin C, Raymond EF, Barik J, Goutagny R, Bethus I, Lopes LV, Migliore M, Marie H. The Amyloid Precursor Protein C-Terminal Domain Alters CA1 Neuron Firing, Modifying Hippocampus Oscillations and Impairing Spatial Memory Encoding. Cell Rep 2020; 29:317-331.e5. [PMID: 31597094 DOI: 10.1016/j.celrep.2019.08.103] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2018] [Revised: 08/09/2019] [Accepted: 08/29/2019] [Indexed: 12/15/2022] Open
Abstract
There is a growing consensus that Alzheimer's disease (AD) involves failure of the homeostatic machinery, which underlies the firing stability of neural circuits. What are the culprits leading to neuron firing instability? The amyloid precursor protein (APP) is central to AD pathogenesis, and we recently showed that its intracellular domain (AICD) could modify synaptic signal integration. We now hypothesize that AICD modifies neuron firing activity, thus contributing to the disruption of memory processes. Using cellular, electrophysiological, and behavioral techniques, we show that pathological AICD levels weaken CA1 neuron firing activity through a gene-transcription-dependent mechanism. Furthermore, increased AICD production in hippocampal neurons modifies oscillatory activity, specifically in the γ-frequency range, and disrupts spatial memory task. Collectively, our data suggest that AICD pathological levels, observed in AD mouse models and in human patients, might contribute to progressive neuron homeostatic failure, driving the shift from normal aging to AD.
Collapse
Affiliation(s)
| | - Xavier Mouska
- Université Côte d'Azur, CNRS UMR 7275, IPMC, Valbonne, France
| | - Daniela Bianchi
- Institute of Biophysics, National Research Council, Palermo, Italy
| | - Mariana Temido-Ferreira
- Instituto de Medicina Molecular, Faculdade de Medicina de Lisboa, Universidade de Lisboa, Lisboa, Portugal
| | - Joana Rajão-Saraiva
- Instituto de Medicina Molecular, Faculdade de Medicina de Lisboa, Universidade de Lisboa, Lisboa, Portugal
| | - Rui Gomes
- Instituto de Medicina Molecular, Faculdade de Medicina de Lisboa, Universidade de Lisboa, Lisboa, Portugal
| | | | | | - Carine Gandin
- Université Côte d'Azur, CNRS UMR 7275, IPMC, Valbonne, France
| | | | - Jacques Barik
- Université Côte d'Azur, CNRS UMR 7275, IPMC, Valbonne, France
| | - Romain Goutagny
- Université de Strasbourg, CNRS UMR 7364, LNCA, Strasbourg, France
| | - Ingrid Bethus
- Université Côte d'Azur, CNRS UMR 7275, IPMC, Valbonne, France
| | - Luisa V Lopes
- Instituto de Medicina Molecular, Faculdade de Medicina de Lisboa, Universidade de Lisboa, Lisboa, Portugal
| | - Michele Migliore
- Institute of Biophysics, National Research Council, Palermo, Italy
| | - Hélène Marie
- Université Côte d'Azur, CNRS UMR 7275, IPMC, Valbonne, France
| |
Collapse
|
26
|
Ca 2+-activated KCa3.1 potassium channels contribute to the slow afterhyperpolarization in L5 neocortical pyramidal neurons. Sci Rep 2020; 10:14484. [PMID: 32879404 PMCID: PMC7468258 DOI: 10.1038/s41598-020-71415-x] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2020] [Accepted: 08/07/2020] [Indexed: 01/15/2023] Open
Abstract
Layer 5 neocortical pyramidal neurons are known to display slow Ca2+-dependent afterhyperpolarization (sAHP) after bursts of spikes, which is similar to the sAHP in CA1 hippocampal cells. However, the mechanisms of sAHP in the neocortex remain poorly understood. Here, we identified the Ca2+-gated potassium KCa3.1 channels as contributors to sAHP in ER81-positive neocortical pyramidal neurons. Moreover, our experiments strongly suggest that the relationship between sAHP and KCa3.1 channels in a feedback mechanism underlies the adaptation of the spiking frequency of layer 5 pyramidal neurons. We demonstrated the relationship between KCa3.1 channels and sAHP using several parallel methods: electrophysiology, pharmacology, immunohistochemistry, and photoactivatable probes. Our experiments demonstrated that ER81 immunofluorescence in layer 5 co-localized with KCa3.1 immunofluorescence in the soma. Targeted Ca2+ uncaging confirmed two major features of KCa3.1 channels: preferential somatodendritic localization and Ca2+-driven gating. In addition, both the sAHP and the slow Ca2+-induced hyperpolarizing current were sensitive to TRAM-34, a selective blocker of KCa3.1 channels.
Collapse
|
27
|
Kopach O, Esteras N, Wray S, Rusakov DA, Abramov AY. Maturation and phenotype of pathophysiological neuronal excitability of human cells in tau-related dementia. J Cell Sci 2020; 133:jcs241687. [PMID: 32299835 PMCID: PMC7272359 DOI: 10.1242/jcs.241687] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2019] [Accepted: 04/01/2020] [Indexed: 01/23/2023] Open
Abstract
Frontotemporal dementia and parkinsonism (FTDP-17) caused by the 10+16 splice-site mutation in the gene encoding microtubule-associated protein tau (MAPT) provides an established platform to model tau-related dementia in vitro Neurons derived from human induced pluripotent stem cells (iPSCs) have been shown to recapitulate the neurodevelopmental profile of tau pathology during in vitro corticogenesis, as in the adult human brain. However, the neurophysiological phenotype of these cells has remained unknown, leaving unanswered questions regarding the functional relevance and the gnostic power of this disease model. In this study, we used electrophysiology to explore the membrane properties and intrinsic excitability of the generated neurons and found that human cells mature by ∼150 days of neurogenesis to become compatible with matured cortical neurons. In earlier FTDP-17, however, neurons exhibited a depolarized resting membrane potential associated with increased resistance and reduced voltage-gated Na+- and K+-channel-mediated conductance. Expression of the Nav1.6 protein was reduced in FTDP-17. These effects led to reduced cell capability of induced firing and changed the action potential waveform in FTDP-17. The revealed neuropathology might thus contribute to the clinicopathological profile of the disease. This sheds new light on the significance of human in vitro models of dementia.
Collapse
Affiliation(s)
- Olga Kopach
- Department of Clinical and Experimental Epilepsy, UCL Queen Square Institute of Neurology, London, WC1N 3BG, UK
| | - Noemí Esteras
- Department of Clinical and Movement Neurosciences, UCL Queen Square Institute of Neurology, London, WC1N 3BG, UK
| | - Selina Wray
- Department of Neurodegenerative Disease, UCL Queen Square Institute of Neurology, London, WC1N 1PJ, UK
| | - Dmitri A Rusakov
- Department of Clinical and Experimental Epilepsy, UCL Queen Square Institute of Neurology, London, WC1N 3BG, UK
| | - Andrey Y Abramov
- Department of Clinical and Movement Neurosciences, UCL Queen Square Institute of Neurology, London, WC1N 3BG, UK
| |
Collapse
|
28
|
Moore SJ, Murphy GG. The role of L-type calcium channels in neuronal excitability and aging. Neurobiol Learn Mem 2020; 173:107230. [PMID: 32407963 DOI: 10.1016/j.nlm.2020.107230] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2019] [Revised: 03/09/2020] [Accepted: 04/12/2020] [Indexed: 12/11/2022]
Abstract
Over the last two decades there has been significant progress towards understanding the neural substrates that underlie age-related cognitive decline. Although many of the exact molecular and cellular mechanisms have yet to be fully understood, there is consensus that alterations in neuronal calcium homeostasis contribute to age-related deficits in learning and memory. Furthermore, it is thought that the age-related changes in calcium homeostasis are driven, at least in part, by changes in calcium channel expression. In this review, we focus on the role of a specific class of calcium channels: L-type voltage-gated calcium channels (LVGCCs). We provide the reader with a general introduction to voltage-gated calcium channels, followed by a more detailed description of LVGCCs and how they serve to regulate neuronal excitability via the post burst afterhyperpolarization (AHP). We conclude by reviewing studies that link the slow component of the AHP to learning and memory, and discuss how age-related increases in LVGCC expression may underlie cognitive decline by mediating a decrease in neuronal excitability.
Collapse
Affiliation(s)
- Shannon J Moore
- Michigan Neuroscience Institute, University of Michigan Medical School, 109 Zina Pitcher Place, Ann Arbor, MI 48109, United States; Department of Molecular and Integrative Physiology, University of Michigan Medical School, 109 Zina Pitcher Place, Ann Arbor, MI 48109, United States
| | - Geoffrey G Murphy
- Michigan Neuroscience Institute, University of Michigan Medical School, 109 Zina Pitcher Place, Ann Arbor, MI 48109, United States; Department of Molecular and Integrative Physiology, University of Michigan Medical School, 109 Zina Pitcher Place, Ann Arbor, MI 48109, United States.
| |
Collapse
|
29
|
Tubert C, Murer MG. What’s wrong with the striatal cholinergic interneurons in Parkinson’s disease? Focus on intrinsic excitability. Eur J Neurosci 2020; 53:2100-2116. [DOI: 10.1111/ejn.14742] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2020] [Revised: 04/04/2020] [Accepted: 04/05/2020] [Indexed: 12/14/2022]
Affiliation(s)
- Cecilia Tubert
- Instituto de Fisiología y Biofísica “Bernardo Houssay”, (IFIBIO‐Houssay) Grupo de Neurociencia de Sistemas Universidad de Buenos Aires y Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET) Buenos Aires Argentina
| | - Mario Gustavo Murer
- Instituto de Fisiología y Biofísica “Bernardo Houssay”, (IFIBIO‐Houssay) Grupo de Neurociencia de Sistemas Universidad de Buenos Aires y Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET) Buenos Aires Argentina
| |
Collapse
|
30
|
Characterization of Convergent Suppression by UCL-2077 (3-(Triphenylmethylaminomethyl)pyridine), Known to Inhibit Slow Afterhyperpolarization, of erg-Mediated Potassium Currents and Intermediate-Conductance Calcium-Activated Potassium Channels. Int J Mol Sci 2020; 21:ijms21041441. [PMID: 32093314 PMCID: PMC7073080 DOI: 10.3390/ijms21041441] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2020] [Revised: 02/15/2020] [Accepted: 02/18/2020] [Indexed: 01/06/2023] Open
Abstract
UCL-2077 (triphenylmethylaminomethyl)pyridine) was previously reported to suppress slow afterhyperpolarization in neurons. However, the information with respect to the effects of UCL-2077 on ionic currents is quite scarce. The addition of UCL-2077 decreased the amplitude of erg-mediated K+ current (IK(erg)) together with an increased deactivation rate of the current in pituitary GH3 cells. The IC50 and KD values of UCL-2077-induced inhibition of IK(erg) were 4.7 and 5.1 μM, respectively. UCL-2077 (10 μM) distinctly shifted the midpoint in the activation curve of IK(erg) to less hyperpolarizing potentials by 17 mV. Its presence decreased the degree of voltage hysteresis for IK(erg) elicitation by long-lasting triangular ramp pulse. It also diminished the probability of the opening of intermediate-conductance Ca2+-activated K+ channels. In cell-attached current recordings, UCL-2077 raised the frequency of action currents. When KCNH2 mRNA was knocked down, a UCL-2077-mediated increase in AC firing was attenuated. Collectively, the actions elaborated herein conceivably contribute to the perturbating effects of this compound on electrical behaviors of excitable cells.
Collapse
|
31
|
Oh MM, Disterhoft JF. Learning and aging affect neuronal excitability and learning. Neurobiol Learn Mem 2019; 167:107133. [PMID: 31786311 DOI: 10.1016/j.nlm.2019.107133] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2019] [Revised: 11/21/2019] [Accepted: 11/27/2019] [Indexed: 11/20/2022]
Abstract
The first study that demonstrated a change in intrinsic neuronal excitability after learning in ex vivo brain tissue slices from a mammal was published over thirty years ago. Numerous other manuscripts describing similar learning-related changes have followed over the years since the original paper demonstrating the postburst afterhyperpolarization (AHP) reduction in CA1 pyramidal neurons from rabbits that learned delay eyeblink conditioning was published. In addition to the learning-related changes, aging-related enlargement of the postburst AHP in CA1 pyramidal neurons have been reported. Extensive work has been done relating slow afterhyperpolarization enhancement in CA1 hippocampus to slowed learning in some aging animals. These reproducible findings strongly implicate modulation of the postburst AHP as an essential cellular mechanism necessary for successful learning, at least in learning tasks that engage CA1 hippocampal pyramidal neurons.
Collapse
Affiliation(s)
- M Matthew Oh
- Department of Physiology, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611-3008, United States
| | - John F Disterhoft
- Department of Physiology, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611-3008, United States.
| |
Collapse
|
32
|
Protein Kinase A-Mediated Suppression of the Slow Afterhyperpolarizing KCa3.1 Current in Temporal Lobe Epilepsy. J Neurosci 2019; 39:9914-9926. [PMID: 31672789 DOI: 10.1523/jneurosci.1603-19.2019] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2019] [Revised: 09/24/2019] [Accepted: 10/07/2019] [Indexed: 01/01/2023] Open
Abstract
Brain insults, such as trauma, stroke, anoxia, and status epilepticus (SE), cause multiple changes in synaptic function and intrinsic properties of surviving neurons that may lead to the development of epilepsy. Experimentally, a single SE episode, induced by the convulsant pilocarpine, initiates the development of an epileptic condition resembling human temporal lobe epilepsy (TLE). Principal hippocampal neurons from such epileptic animals display enhanced spike output in response to excitatory stimuli compared with neurons from nonepileptic animals. This enhanced firing is negatively related to the size of the slow afterhyperpolarization (sAHP), which is reduced in the epileptic neurons. The sAHP is an intrinsic neuronal negative feedback mechanism consisting normally of two partially overlapping components produced by disparate mechanisms. One component is generated by activation of Ca2+-gated K+ (KCa) channels, likely KCa3.1, consequent to spike Ca2+ influx (the KCa-sAHP component). The second component is generated by enhancement of the electrogenic Na+/K+ ATPase (NKA) by spike Na+ influx (NKA-sAHP component). Here we show that the KCa-sAHP component is markedly reduced in male rat epileptic neurons, whereas the NKA-sAHP component is not altered. The KCa-sAHP reduction is due to the downregulation of KCa3.1 channels, mediated by cAMP-dependent protein kinase A (PKA). This sustained effect can be acutely reversed by applying PKA inhibitors, leading also to normalization of the spike output of epileptic neurons. We propose that the novel "acquired channelopathy" described here, namely, PKA-mediated downregulation of KCa3.1 activity, provides an innovative target for developing new treatments for TLE, hopefully overcoming the pharmacoresistance to traditional drugs.SIGNIFICANCE STATEMENT Epilepsy, a common neurological disorder, often develops following a brain insult. Identifying key molecular and cellular mechanisms underlying acquired epilepsy is critical for developing effective antiepileptic therapies. In an experimental model of acquired epilepsy, we show that principal hippocampal neurons become intrinsically hyperexcitable. This alteration is due predominantly to the downregulation of a ubiquitous class of potassium ion channels, KCa3.1, whose main function is to dampen neuronal excitability. KCa3.1 downregulation is mediated by the cAMP-dependent protein kinase A (PKA) signaling pathway. Most importantly, it can be acutely reversed by PKA inhibitors, leading to recovery of KCa3.1 function and normalization of neuronal excitability. The discovery of this novel epileptogenic mechanism hopefully will facilitate the development of more efficient pharmacotherapy for acquired epilepsy.
Collapse
|
33
|
Capo Rangel G, Prezioso J, Gerardo-Giorda L, Somersalo E, Calvetti D. Brain energetics plays a key role in the coordination of electrophysiology, metabolism and hemodynamics: Evidence from an integrated computational model. J Theor Biol 2019; 478:26-39. [DOI: 10.1016/j.jtbi.2019.06.003] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2019] [Revised: 06/01/2019] [Accepted: 06/04/2019] [Indexed: 10/26/2022]
|
34
|
Dunn AR, Kaczorowski CC. Regulation of intrinsic excitability: Roles for learning and memory, aging and Alzheimer's disease, and genetic diversity. Neurobiol Learn Mem 2019; 164:107069. [PMID: 31442579 DOI: 10.1016/j.nlm.2019.107069] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2019] [Revised: 08/09/2019] [Accepted: 08/17/2019] [Indexed: 12/28/2022]
Abstract
Plasticity of intrinsic neuronal excitability facilitates learning and memory across multiple species, with aberrant modulation of this process being linked to the development of neurological symptoms in models of cognitive aging and Alzheimer's disease. Learning-related increases in intrinsic excitability of neurons occurs in a variety of brain regions, and is generally thought to promote information processing and storage through enhancement of synaptic throughput and induction of synaptic plasticity. Experience-dependent changes in intrinsic neuronal excitability rely on activity-dependent gene expression patterns, which can be influenced by genetic and environmental factors, aging, and disease. Reductions in baseline intrinsic excitability, as well as aberrant plasticity of intrinsic neuronal excitability and in some cases pathological hyperexcitability, have been associated with cognitive deficits in animal models of both normal cognitive aging and Alzheimer's disease. Genetic factors that modulate plasticity of intrinsic excitability likely underlie individual differences in cognitive function and susceptibility to cognitive decline. Thus, targeting molecular mediators that either control baseline intrinsic neuronal excitability, subserve learning-related intrinsic neuronal plasticity, and/or promote resilience may be a promising therapeutic strategy for maintaining cognitive function in aging and disease. In this review, we discuss the complementary relationship between intrinsic excitability and learning, with a particular focus on how this relationship varies as a function of age, disease state, and genetic make-up, and how targeting these factors may help to further elucidate our understanding of the role of intrinsic excitability in cognitive function and cognitive decline.
Collapse
Affiliation(s)
- Amy R Dunn
- The Jackson Laboratory, Bar Harbor, ME 04609, USA
| | | |
Collapse
|
35
|
Junctophilin Proteins Tether a Cav1-RyR2-KCa3.1 Tripartite Complex to Regulate Neuronal Excitability. Cell Rep 2019; 28:2427-2442.e6. [DOI: 10.1016/j.celrep.2019.07.075] [Citation(s) in RCA: 32] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2019] [Revised: 06/20/2019] [Accepted: 07/19/2019] [Indexed: 12/12/2022] Open
|
36
|
Brown BM, Shim H, Christophersen P, Wulff H. Pharmacology of Small- and Intermediate-Conductance Calcium-Activated Potassium Channels. Annu Rev Pharmacol Toxicol 2019; 60:219-240. [PMID: 31337271 DOI: 10.1146/annurev-pharmtox-010919-023420] [Citation(s) in RCA: 67] [Impact Index Per Article: 11.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
The three small-conductance calcium-activated potassium (KCa2) channels and the related intermediate-conductance KCa3.1 channel are voltage-independent K+ channels that mediate calcium-induced membrane hyperpolarization. When intracellular calcium increases in the channel vicinity, it calcifies the flexible N lobe of the channel-bound calmodulin, which then swings over to the S4-S5 linker and opens the channel. KCa2 and KCa3.1 channels are highly druggable and offer multiple binding sites for venom peptides and small-molecule blockers as well as for positive- and negative-gating modulators. In this review, we briefly summarize the physiological role of KCa channels and then discuss the pharmacophores and the mechanism of action of the most commonly used peptidic and small-molecule KCa2 and KCa3.1 modulators. Finally, we describe the progress that has been made in advancing KCa3.1 blockers and KCa2.2 negative- and positive-gating modulators toward the clinic for neurological and cardiovascular diseases and discuss the remaining challenges.
Collapse
Affiliation(s)
- Brandon M Brown
- Department of Pharmacology, University of California, Davis, California 95616, USA;
| | - Heesung Shim
- Department of Pharmacology, University of California, Davis, California 95616, USA;
| | | | - Heike Wulff
- Department of Pharmacology, University of California, Davis, California 95616, USA;
| |
Collapse
|
37
|
Higham J, Sahu G, Wazen RM, Colarusso P, Gregorie A, Harvey BSJ, Goudswaard L, Varley G, Sheppard DN, Turner RW, Marrion NV. Preferred Formation of Heteromeric Channels between Coexpressed SK1 and IKCa Channel Subunits Provides a Unique Pharmacological Profile of Ca 2+-Activated Potassium Channels. Mol Pharmacol 2019; 96:115-126. [PMID: 31048549 DOI: 10.1124/mol.118.115634] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2018] [Accepted: 04/27/2019] [Indexed: 01/19/2023] Open
Abstract
Three small conductance calcium-activated potassium channel (SK) subunits have been cloned and found to preferentially form heteromeric channels when expressed in a heterologous expression system. The original cloning of the gene encoding the intermediate conductance calcium-activated potassium channel (IKCa) was termed SK4 because of the high homology between channel subtypes. Recent immunovisualization suggests that IKCa is expressed in the same subcellular compartments of some neurons as SK channel subunits. Stochastic optical reconstruction microscopy super-resolution microscopy revealed that coexpressed IKCa and SK1 channel subunits were closely associated, a finding substantiated by measurement of fluorescence resonance energy transfer between coexpressed fluorophore-tagged subunits. Expression of homomeric SK1 channels produced current that displayed typical sensitivity to SK channel inhibitors, while expressed IKCa channel current was inhibited by known IKCa channel blockers. Expression of both SK1 and IKCa subunits gave a current that displayed no sensitivity to SK channel inhibitors and a decreased sensitivity to IKCa current inhibitors. Single channel recording indicated that coexpression of SK1 and IKCa subunits produced channels with properties intermediate between those observed for homomeric channels. These data indicate that SK1 and IKCa channel subunits preferentially combine to form heteromeric channels that display pharmacological and biophysical properties distinct from those seen with homomeric channels.
Collapse
Affiliation(s)
- James Higham
- School of Physiology, Pharmacology and Neuroscience, University of Bristol, Bristol, United Kingdom (J.H., A.G., B.S.J.H., L.G., G.V., D.N.S., N.V.M.); and Hotchkiss Brain Institute (G.S., R.W.T.) and Snyder Institute for Chronic Diseases (R.-M.W., P.C.), Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
| | - Giriraj Sahu
- School of Physiology, Pharmacology and Neuroscience, University of Bristol, Bristol, United Kingdom (J.H., A.G., B.S.J.H., L.G., G.V., D.N.S., N.V.M.); and Hotchkiss Brain Institute (G.S., R.W.T.) and Snyder Institute for Chronic Diseases (R.-M.W., P.C.), Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
| | - Rima-Marie Wazen
- School of Physiology, Pharmacology and Neuroscience, University of Bristol, Bristol, United Kingdom (J.H., A.G., B.S.J.H., L.G., G.V., D.N.S., N.V.M.); and Hotchkiss Brain Institute (G.S., R.W.T.) and Snyder Institute for Chronic Diseases (R.-M.W., P.C.), Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
| | - Pina Colarusso
- School of Physiology, Pharmacology and Neuroscience, University of Bristol, Bristol, United Kingdom (J.H., A.G., B.S.J.H., L.G., G.V., D.N.S., N.V.M.); and Hotchkiss Brain Institute (G.S., R.W.T.) and Snyder Institute for Chronic Diseases (R.-M.W., P.C.), Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
| | - Alice Gregorie
- School of Physiology, Pharmacology and Neuroscience, University of Bristol, Bristol, United Kingdom (J.H., A.G., B.S.J.H., L.G., G.V., D.N.S., N.V.M.); and Hotchkiss Brain Institute (G.S., R.W.T.) and Snyder Institute for Chronic Diseases (R.-M.W., P.C.), Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
| | - Bartholomew S J Harvey
- School of Physiology, Pharmacology and Neuroscience, University of Bristol, Bristol, United Kingdom (J.H., A.G., B.S.J.H., L.G., G.V., D.N.S., N.V.M.); and Hotchkiss Brain Institute (G.S., R.W.T.) and Snyder Institute for Chronic Diseases (R.-M.W., P.C.), Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
| | - Lucy Goudswaard
- School of Physiology, Pharmacology and Neuroscience, University of Bristol, Bristol, United Kingdom (J.H., A.G., B.S.J.H., L.G., G.V., D.N.S., N.V.M.); and Hotchkiss Brain Institute (G.S., R.W.T.) and Snyder Institute for Chronic Diseases (R.-M.W., P.C.), Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
| | - Gemma Varley
- School of Physiology, Pharmacology and Neuroscience, University of Bristol, Bristol, United Kingdom (J.H., A.G., B.S.J.H., L.G., G.V., D.N.S., N.V.M.); and Hotchkiss Brain Institute (G.S., R.W.T.) and Snyder Institute for Chronic Diseases (R.-M.W., P.C.), Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
| | - David N Sheppard
- School of Physiology, Pharmacology and Neuroscience, University of Bristol, Bristol, United Kingdom (J.H., A.G., B.S.J.H., L.G., G.V., D.N.S., N.V.M.); and Hotchkiss Brain Institute (G.S., R.W.T.) and Snyder Institute for Chronic Diseases (R.-M.W., P.C.), Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
| | - Ray W Turner
- School of Physiology, Pharmacology and Neuroscience, University of Bristol, Bristol, United Kingdom (J.H., A.G., B.S.J.H., L.G., G.V., D.N.S., N.V.M.); and Hotchkiss Brain Institute (G.S., R.W.T.) and Snyder Institute for Chronic Diseases (R.-M.W., P.C.), Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
| | - Neil V Marrion
- School of Physiology, Pharmacology and Neuroscience, University of Bristol, Bristol, United Kingdom (J.H., A.G., B.S.J.H., L.G., G.V., D.N.S., N.V.M.); and Hotchkiss Brain Institute (G.S., R.W.T.) and Snyder Institute for Chronic Diseases (R.-M.W., P.C.), Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
| |
Collapse
|
38
|
Cancer-Associated Intermediate Conductance Ca 2+-Activated K⁺ Channel K Ca3.1. Cancers (Basel) 2019; 11:cancers11010109. [PMID: 30658505 PMCID: PMC6357066 DOI: 10.3390/cancers11010109] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2018] [Revised: 01/10/2019] [Accepted: 01/13/2019] [Indexed: 12/14/2022] Open
Abstract
Several tumor entities have been reported to overexpress KCa3.1 potassium channels due to epigenetic, transcriptional, or post-translational modifications. By modulating membrane potential, cell volume, or Ca2+ signaling, KCa3.1 has been proposed to exert pivotal oncogenic functions in tumorigenesis, malignant progression, metastasis, and therapy resistance. Moreover, KCa3.1 is expressed by tumor-promoting stroma cells such as fibroblasts and the tumor vasculature suggesting a role of KCa3.1 in the adaptation of the tumor microenvironment. Combined, this features KCa3.1 as a candidate target for innovative anti-cancer therapy. However, immune cells also express KCa3.1 thereby contributing to T cell activation. Thus, any strategy targeting KCa3.1 in anti-cancer therapy may also modulate anti-tumor immune activity and/or immunosuppression. The present review article highlights the potential of KCa3.1 as an anti-tumor target providing an overview of the current knowledge on its function in tumor pathogenesis with emphasis on vasculo- and angiogenesis as well as anti-cancer immune responses.
Collapse
|
39
|
Church TW, Brown JT, Marrion NV. β 3-Adrenergic receptor-dependent modulation of the medium afterhyperpolarization in rat hippocampal CA1 pyramidal neurons. J Neurophysiol 2019; 121:773-784. [PMID: 30625002 DOI: 10.1152/jn.00334.2018] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023] Open
Abstract
Action potential firing in hippocampal pyramidal neurons is regulated by generation of an afterhyperpolarization (AHP). Three phases of AHP are recognized, with the fast AHP regulating action potential firing at the onset of a burst and the medium and slow AHPs supressing action potential firing over hundreds of milliseconds and seconds, respectively. Activation of β-adrenergic receptors suppresses the slow AHP by a protein kinase A-dependent pathway. However, little is known regarding modulation of the medium AHP. Application of the selective β-adrenergic receptor agonist isoproterenol suppressed both the medium and slow AHPs evoked in rat CA1 hippocampal pyramidal neurons recorded from slices maintained in organotypic culture. Suppression of the slow AHP was mimicked by intracellular application of cAMP, with the suppression of the medium AHP by isoproterenol still being evident in cAMP-dialyzed cells. Suppression of both the medium and slow AHPs was antagonized by the β-adrenergic receptor antagonist propranolol. The effect of isoproterenol to suppress the medium AHP was mimicked by two β3-adrenergic receptor agonists, BRL37344 and SR58611A. The medium AHP was mediated by activation of small-conductance calcium-activated K+ channels and deactivation of H channels at the resting membrane potential. Suppression of the medium AHP by isoproterenol was reduced by pretreating cells with the H-channel blocker ZD7288. These data suggest that activation of β3-adrenergic receptors inhibits H channels, which suppresses the medium AHP in CA1 hippocampal neurons by utilizing a pathway that is independent of a rise in intracellular cAMP. This finding highlights a potential new target in modulating H-channel activity and thereby neuronal excitability. NEW & NOTEWORTHY The noradrenergic input into the hippocampus is involved in modulating long-term synaptic plasticity and is implicated in learning and memory. We demonstrate that activation of functional β3-adrenergic receptors suppresses the medium afterhyperpolarization in hippocampal pyramidal neurons. This finding provides an additional mechanism to increase action potential firing frequency, where neuronal excitability is likely to be crucial in cognition and memory.
Collapse
Affiliation(s)
- Timothy W Church
- School of Physiology, Pharmacology and Neuroscience, University of Bristol , Bristol , United Kingdom
| | - Jon T Brown
- University of Exeter Medical School , Exeter , United Kingdom
| | - Neil V Marrion
- School of Physiology, Pharmacology and Neuroscience, University of Bristol , Bristol , United Kingdom
| |
Collapse
|
40
|
Medlock L, Shute L, Fry M, Standage D, Ferguson AV. Ionic mechanisms underlying tonic and burst firing behavior in subfornical organ neurons: a combined experimental and modeling study. J Neurophysiol 2018; 120:2269-2281. [PMID: 30089060 DOI: 10.1152/jn.00340.2018] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022] Open
Abstract
Subfornical organ (SFO) neurons exhibit heterogeneity in current expression and spiking behavior, where the two major spiking phenotypes appear as tonic and burst firing. Insight into the mechanisms behind this heterogeneity is critical for understanding how the SFO, a sensory circumventricular organ, integrates and selectively influences physiological function. To integrate efficient methods for studying this heterogeneity, we built a single-compartment, Hodgkin-Huxley-type model of an SFO neuron that is parameterized by SFO-specific in vitro patch-clamp data. The model accounts for the membrane potential distribution and spike train variability of both tonic and burst firing SFO neurons. Analysis of model dynamics confirms that a persistent Na+ and Ca2+ currents are required for burst initiation and maintenance and suggests that a slow-activating K+ current may be responsible for burst termination in SFO neurons. Additionally, the model suggests that heterogeneity in current expression and subsequent influence on spike afterpotential underlie the behavioral differences between tonic and burst firing SFO neurons. Future use of this model in coordination with single neuron patch-clamp electrophysiology provides a platform for explaining and predicting the response of SFO neurons to various combinations of circulating signals, thus elucidating the mechanisms underlying physiological signal integration within the SFO. NEW & NOTEWORTHY Our understanding of how the subfornical organ (SFO) selectively influences autonomic nervous system function remains incomplete but theoretically results from the electrical responses of SFO neurons to physiologically important signals. We have built a computational model of SFO neurons, derived from and supported by experimental data, which explains how SFO neurons produce different electrical patterns. The model provides an efficient system to theoretically and experimentally explore how changes in the essential features of SFO neurons affect their electrical activity.
Collapse
Affiliation(s)
- Laura Medlock
- Center for Neuroscience Studies, Queen's University , Kingston, Ontario , Canada
| | - Lauren Shute
- Department of Biological Sciences, University of Manitoba , Winnipeg, Manitoba , Canada
| | - Mark Fry
- Department of Biological Sciences, University of Manitoba , Winnipeg, Manitoba , Canada
| | - Dominic Standage
- Center for Neuroscience Studies, Queen's University , Kingston, Ontario , Canada
| | - Alastair V Ferguson
- Center for Neuroscience Studies, Queen's University , Kingston, Ontario , Canada
| |
Collapse
|
41
|
Kshatri AS, Gonzalez-Hernandez A, Giraldez T. Physiological Roles and Therapeutic Potential of Ca 2+ Activated Potassium Channels in the Nervous System. Front Mol Neurosci 2018; 11:258. [PMID: 30104956 PMCID: PMC6077210 DOI: 10.3389/fnmol.2018.00258] [Citation(s) in RCA: 84] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2018] [Accepted: 07/06/2018] [Indexed: 12/21/2022] Open
Abstract
Within the potassium ion channel family, calcium activated potassium (KCa) channels are unique in their ability to couple intracellular Ca2+ signals to membrane potential variations. KCa channels are diversely distributed throughout the central nervous system and play fundamental roles ranging from regulating neuronal excitability to controlling neurotransmitter release. The physiological versatility of KCa channels is enhanced by alternative splicing and co-assembly with auxiliary subunits, leading to fundamental differences in distribution, subunit composition and pharmacological profiles. Thus, understanding specific KCa channels’ mechanisms in neuronal function is challenging. Based on their single channel conductance, KCa channels are divided into three subtypes: small (SK, 4–14 pS), intermediate (IK, 32–39 pS) and big potassium (BK, 200–300 pS) channels. This review describes the biophysical characteristics of these KCa channels, as well as their physiological roles and pathological implications. In addition, we also discuss the current pharmacological strategies and challenges to target KCa channels for the treatment of various neurological and psychiatric disorders.
Collapse
Affiliation(s)
- Aravind S Kshatri
- Department of Basic Medical Sciences, Medical School, Universidad de La Laguna, Tenerife, Spain.,Instituto de Tecnologias Biomedicas, Universidad de La Laguna, Tenerife, Spain
| | - Alberto Gonzalez-Hernandez
- Department of Basic Medical Sciences, Medical School, Universidad de La Laguna, Tenerife, Spain.,Instituto de Tecnologias Biomedicas, Universidad de La Laguna, Tenerife, Spain
| | - Teresa Giraldez
- Department of Basic Medical Sciences, Medical School, Universidad de La Laguna, Tenerife, Spain.,Instituto de Tecnologias Biomedicas, Universidad de La Laguna, Tenerife, Spain
| |
Collapse
|
42
|
Tiwari MN, Mohan S, Biala Y, Yaari Y. Differential contributions of Ca 2+ -activated K + channels and Na + /K + -ATPases to the generation of the slow afterhyperpolarization in CA1 pyramidal cells. Hippocampus 2018; 28:338-357. [PMID: 29431274 PMCID: PMC5947627 DOI: 10.1002/hipo.22836] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2017] [Revised: 12/17/2017] [Accepted: 02/05/2018] [Indexed: 12/13/2022]
Abstract
In many types of CNS neurons, repetitive spiking produces a slow afterhyperpolarization (sAHP), providing sustained, intrinsically generated negative feedback to neuronal excitation. Changes in the sAHP have been implicated in learning behaviors, in cognitive decline in aging, and in epileptogenesis. Despite its importance in brain function, the mechanisms generating the sAHP are still controversial. Here we have addressed the roles of M-type K+ current (IM ), Ca2+ -gated K+ currents (ICa(K) 's) and Na+ /K+ -ATPases (NKAs) current to sAHP generation in adult rat CA1 pyramidal cells maintained at near-physiological temperature (35 °C). No evidence for IM contribution to the sAHP was found in these neurons. Both ICa(K) 's and NKA current contributed to sAHP generation, the latter being the predominant generator of the sAHP, particularly when evoked with short trains of spikes. Of the different NKA isoenzymes, α1 -NKA played the key role, endowing the sAHP a steep voltage-dependence. Thus normal and pathological changes in α1 -NKA expression or function may affect cognitive processes by modulating the inhibitory efficacy of the sAHP.
Collapse
Affiliation(s)
- Manindra Nath Tiwari
- Department of Medical Neurobiology; Institute for Medical Research Israel‐CanadaThe Hebrew University‐Hadassah School of MedicineJerusalem91120Israel
| | - Sandesh Mohan
- Department of Medical Neurobiology; Institute for Medical Research Israel‐CanadaThe Hebrew University‐Hadassah School of MedicineJerusalem91120Israel
| | - Yoav Biala
- Department of Medical Neurobiology; Institute for Medical Research Israel‐CanadaThe Hebrew University‐Hadassah School of MedicineJerusalem91120Israel
| | - Yoel Yaari
- Department of Medical Neurobiology; Institute for Medical Research Israel‐CanadaThe Hebrew University‐Hadassah School of MedicineJerusalem91120Israel
| |
Collapse
|
43
|
Calvetti D, Capo Rangel G, Gerardo Giorda L, Somersalo E. A computational model integrating brain electrophysiology and metabolism highlights the key role of extracellular potassium and oxygen. J Theor Biol 2018. [PMID: 29530764 DOI: 10.1016/j.jtbi.2018.02.029] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
The human brain is a small organ which uses a disproportionate amount of the total metabolic energy production in the body. While it is well understood that the most significant energy sink is the maintenance of the neuronal membrane potential during the brain signaling activity, the role of astrocytes in the energy balance continues to be the topic of a lot of research. A key function of astrocytes, besides clearing glutamate from the synaptic clefts, is the potassium clearing after neuronal activation. Extracellular potassium plays a significant role in triggering neuronal firing, and elevated concentration of potassium may lead to abnormal firing patterns, e.g., seizures, thus emphasizing the importance of the glial K+ buffering role. The predictive mathematical model proposed in this paper elucidates the role of glial potassium clearing in brain energy metabolism, integrating a detailed model of the ion dynamics which regulates neuronal firing with a four compartment metabolic model. Because of the very different characteristic time scales of electrophysiology and metabolism, care must be taken when coupling the two models to ensure that the predictions, e.g., neuronal firing frequencies and the oxygen-glucose index (OGI) of the brain during activation and rest, are in agreement with empirical observations. The temporal multi-scale nature of the problem requires the design of new computational tools to ensure a stable and accurate numerical treatment. The model predictions for different protocols, including combinations of elevated activation and ischemic episodes, are in good agreement with experimental observations reported in the literature.
Collapse
Affiliation(s)
- D Calvetti
- Department of Mathematics, Applied Mathematics and Statistics, Case Western Reserve University, USA
| | | | | | - E Somersalo
- Department of Mathematics, Applied Mathematics and Statistics, Case Western Reserve University, USA; Basque Center for Applied Mathematics, Spain.
| |
Collapse
|
44
|
Reboreda A, Theissen FM, Valero-Aracama MJ, Arboit A, Corbu MA, Yoshida M. Do TRPC channels support working memory? Comparing modulations of TRPC channels and working memory through G-protein coupled receptors and neuromodulators. Behav Brain Res 2018; 354:64-83. [PMID: 29501506 DOI: 10.1016/j.bbr.2018.02.042] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2017] [Revised: 02/27/2018] [Accepted: 02/27/2018] [Indexed: 12/11/2022]
Abstract
Working memory is a crucial ability we use in daily life. However, the cellular mechanisms supporting working memory still remain largely unclear. A key component of working memory is persistent neural firing which is believed to serve short-term (hundreds of milliseconds up to tens of seconds) maintenance of necessary information. In this review, we will focus on the role of transient receptor potential canonical (TRPC) channels as a mechanism underlying persistent firing. Many years of in vitro work have been suggesting a crucial role of TRPC channels in working memory and temporal association tasks. If TRPC channels are indeed a central mechanism for working memory, manipulations which impair or facilitate working memory should have a similar effect on TRPC channel modulation. However, modulations of working memory and TRPC channels were never systematically compared, and it remains unanswered whether TRPC channels indeed contribute to working memory in vivo or not. In this article, we review the effects of G-protein coupled receptors (GPCR) and neuromodulators, including acetylcholine, noradrenalin, serotonin and dopamine, on working memory and TRPC channels. Based on comparisons, we argue that GPCR and downstream signaling pathways that activate TRPC, generally support working memory, while those that suppress TRPC channels impair it. However, depending on the channel types, areas, and systems tested, this is not the case in all studies. Further work to clarify involvement of specific TRPC channels in working memory tasks and how they are affected by neuromodulators is still necessary in the future.
Collapse
Affiliation(s)
- Antonio Reboreda
- Leibniz Institute for Neurobiology (LIN) Magdeburg, Brenneckestraße 6, 39118 Magdeburg, Germany; German Center for Neurodegenerative Diseases (DZNE) Magdeburg, Leipziger Str. 44/Haus 64, 39120, Magdeburg, Germany.
| | - Frederik M Theissen
- German Center for Neurodegenerative Diseases (DZNE) Magdeburg, Leipziger Str. 44/Haus 64, 39120, Magdeburg, Germany
| | - Maria J Valero-Aracama
- Institute of Physiology and Pathophysiology, Friedrich-Alexander-Universität Erlangen-Nürnberg, Universitätsstraße 17, 91054 Erlangen, Germany
| | - Alberto Arboit
- German Center for Neurodegenerative Diseases (DZNE) Magdeburg, Leipziger Str. 44/Haus 64, 39120, Magdeburg, Germany
| | - Mihaela A Corbu
- Ruhr University Bochum (RUB), Universitätsstraße 150, 44801, Bochum, Germany
| | - Motoharu Yoshida
- Leibniz Institute for Neurobiology (LIN) Magdeburg, Brenneckestraße 6, 39118 Magdeburg, Germany; German Center for Neurodegenerative Diseases (DZNE) Magdeburg, Leipziger Str. 44/Haus 64, 39120, Magdeburg, Germany; Center for Behavioral Brain Sciences, 39106, Magdeburg, Germany.
| |
Collapse
|
45
|
Brown BM, Pressley B, Wulff H. KCa3.1 Channel Modulators as Potential Therapeutic Compounds for Glioblastoma. Curr Neuropharmacol 2018; 16:618-626. [PMID: 28676010 PMCID: PMC5997873 DOI: 10.2174/1570159x15666170630164226] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2017] [Revised: 06/01/2017] [Accepted: 06/22/2017] [Indexed: 12/31/2022] Open
Abstract
BACKGROUND The intermediate-conductance Ca2+-activated K+ channel KCa3.1 is widely expressed in cells of the immune system such as T- and B-lymphocytes, mast cells, macrophages and microglia, but also found in dedifferentiated vascular smooth muscle cells, fibroblasts and many cancer cells including pancreatic, prostate, leukemia and glioblastoma. In all these cell types KCa3.1 plays an important role in cellular activation, migration and proliferation by regulating membrane potential and Ca2+ signaling. METHODS AND RESULTS KCa3.1 therefore constitutes an attractive therapeutic target for diseases involving excessive proliferation or activation of one more of these cell types and researchers both in academia and in the pharmaceutical industry have developed several potent and selective small molecule inhibitors of KCa3.1. This article will briefly review the available compounds (TRAM-34, senicapoc, NS6180), their binding sites and mechanisms of action, and then discuss the potential usefulness of these compounds for the treatment of brain tumors based on their brain penetration and their efficacy in reducing microglia activation in animal models of ischemic stroke and Alzheimer's disease. CONCLUSION Senicapoc, which has previously been in Phase III clinical trials, would be available for repurposing, and could be used to quickly translate findings made with other KCa3.1 blocking tool compounds into clinical trials.
Collapse
Affiliation(s)
- Brandon M Brown
- Department of Pharmacology, School of Medicine, University of California, Davis, CA 95616, United States
| | - Brandon Pressley
- Department of Pharmacology, School of Medicine, University of California, Davis, CA 95616, United States
| | - Heike Wulff
- Department of Pharmacology, School of Medicine, University of California, Davis, CA 95616, United States
| |
Collapse
|
46
|
A non-synaptic mechanism of complex learning: Modulation of intrinsic neuronal excitability. Neurobiol Learn Mem 2017; 154:30-36. [PMID: 29196146 DOI: 10.1016/j.nlm.2017.11.015] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2017] [Revised: 11/06/2017] [Accepted: 11/27/2017] [Indexed: 11/24/2022]
Abstract
Training rats in a particularly difficult olfactory discrimination task initiates a period of accelerated learning of other odors, manifested as a dramatic increase in the rats' capacity to acquire memories for new odors once they have learned the first discrimination task, implying that rule learning has taken place. At the cellular level, pyramidal neurons in the piriform cortex, hippocampus and bsolateral amygdala of olfactory-discrimination trained rats show enhanced intrinsic neuronal excitability that lasts for several days after rule learning. Such enhanced intrinsic excitability is mediated by long-term reduction in the post-burst after-hyperpolarization (AHP) which is generated by repetitive spike firing, and is maintained by persistent activation of key second messenger systems. Much like late-LTP, the induction of long-term modulation of intrinsic excitability is protein synthesis dependent. Learning-induced modulation of intrinsic excitability can be bi-directional, pending of the valance of the outcome of the learned task. In this review we describe the physiological and molecular mechanisms underlying the rule learning-induced long-term enhancement in neuronal excitability and discuss the functional significance of such a wide spread modulation of the neurons' ability to sustain repetitive spike generation.
Collapse
|
47
|
Activity-Dependent Facilitation of Ca V1.3 Calcium Channels Promotes KCa3.1 Activation in Hippocampal Neurons. J Neurosci 2017; 37:11255-11270. [PMID: 29038242 DOI: 10.1523/jneurosci.0967-17.2017] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2017] [Revised: 10/02/2017] [Accepted: 10/07/2017] [Indexed: 11/21/2022] Open
Abstract
CaV1 L-type calcium channels are key to regulating neuronal excitability, with the range of functional roles enhanced by interactions with calmodulin, accessory proteins, or CaMKII that modulate channel activity. In hippocampal pyramidal cells, a prominent elevation of CaV1 activity is apparent in late channel openings that can last for seconds following a depolarizing stimulus train. The current study tested the hypothesis that a reported interaction among CaV1.3 channels, the scaffolding protein densin, and CaMKII could generate a facilitation of channel activity that outlasts a depolarizing stimulus. We found that CaV1.3 but not CaV1.2 channels exhibit a long-duration calcium-dependent facilitation (L-CDF) that lasts up to 8 s following a brief 50 Hz stimulus train, but only when coexpressed with densin and CaMKII. To test the physiological role for CaV1.3 L-CDF, we coexpressed the intermediate-conductance KCa3.1 potassium channel, revealing a strong functional coupling to CaV1.3 channel activity that was accentuated by densin and CaMKII. Moreover, the CaV1.3-densin-CaMKII interaction gave rise to an outward tail current of up to 8 s duration following a depolarizing stimulus in both tsA-201 cells and male rat CA1 pyramidal cells. A slow afterhyperpolarization in pyramidal cells was reduced by a selective block of CaV1 channels by isradipine, a CaMKII blocker, and siRNA knockdown of densin, and spike frequency increased upon selective block of CaV1 channel conductance. The results are important in revealing a CaV1.3-densin-CaMKII interaction that extends the contribution of CaV1.3 calcium influx to a time frame well beyond a brief input train.SIGNIFICANCE STATEMENT CaV1 L-type calcium channels play a key role in regulating the output of central neurons by providing calcium influx during repetitive inputs. This study identifies a long-duration calcium-dependent facilitation (L-CDF) of CaV1.3 channels that depends on the scaffolding protein densin and CaMKII and that outlasts a depolarizing stimulus by seconds. We further show a tight functional coupling between CaV1.3 calcium influx and the intermediate-conductance KCa3.1 potassium channel that promotes an outward tail current of up to 8 s following a depolarizing stimulus. Tests in CA1 hippocampal pyramidal cells reveal that a slow AHP is reduced by blocking different components of the CaV1.3-densin-CaMKII interaction, identifying an important role for CaV1.3 L-CDF in regulating neuronal excitability.
Collapse
|
48
|
McKiernan EC, Marrone DF. CA1 pyramidal cells have diverse biophysical properties, affected by development, experience, and aging. PeerJ 2017; 5:e3836. [PMID: 28948109 PMCID: PMC5609525 DOI: 10.7717/peerj.3836] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2017] [Accepted: 08/31/2017] [Indexed: 12/04/2022] Open
Abstract
Neuron types (e.g., pyramidal cells) within one area of the brain are often considered homogeneous, despite variability in their biophysical properties. Here we review literature demonstrating variability in the electrical activity of CA1 hippocampal pyramidal cells (PCs), including responses to somatic current injection, synaptic stimulation, and spontaneous network-related activity. In addition, we describe how responses of CA1 PCs vary with development, experience, and aging, and some of the underlying ionic currents responsible. Finally, we suggest directions that may be the most impactful in expanding this knowledge, including the use of text and data mining to systematically study cellular heterogeneity in more depth; dynamical systems theory to understand and potentially classify neuron firing patterns; and mathematical modeling to study the interaction between cellular properties and network output. Our goals are to provide a synthesis of the literature for experimentalists studying CA1 PCs, to give theorists an idea of the rich diversity of behaviors models may need to reproduce to accurately represent these cells, and to provide suggestions for future research.
Collapse
Affiliation(s)
- Erin C McKiernan
- Departamento de Física, Facultad de Ciencias, Universidad Nacional Autónoma de México, Ciudad de México, México
| | - Diano F Marrone
- Department of Psychology, Wilfrid Laurier University, Waterloo, Ontario, Canada.,McKnight Brain Institute, University of Arizona, Tucson, AZ, United States of America
| |
Collapse
|
49
|
Abstract
The misfolding of the cellular prion protein (PrPC) causes fatal neurodegenerative diseases. Yet PrPC is highly conserved in mammals, suggesting that it exerts beneficial functions preventing its evolutionary elimination. Ablation of PrPC in mice results in well-defined structural and functional alterations in the peripheral nervous system. Many additional phenotypes were ascribed to the lack of PrPC, but some of these were found to arise from genetic artifacts of the underlying mouse models. Here, we revisit the proposed physiological roles of PrPC in the central and peripheral nervous systems and highlight the need for their critical reassessment using new, rigorously controlled animal models.
Collapse
Affiliation(s)
- Marie-Angela Wulf
- Institute of Neuropathology, University of Zurich, Rämistrasse 100, CH-8091, Zürich, Switzerland
| | - Assunta Senatore
- Institute of Neuropathology, University of Zurich, Rämistrasse 100, CH-8091, Zürich, Switzerland
| | - Adriano Aguzzi
- Institute of Neuropathology, University of Zurich, Rämistrasse 100, CH-8091, Zürich, Switzerland.
| |
Collapse
|
50
|
Chakraborty D, Fedorova OV, Bagrov AY, Kaphzan H. Selective ligands for Na+/K+-ATPase α isoforms differentially and cooperatively regulate excitability of pyramidal neurons in distinct brain regions. Neuropharmacology 2017; 117:338-351. [DOI: 10.1016/j.neuropharm.2017.02.016] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2016] [Revised: 01/24/2017] [Accepted: 02/17/2017] [Indexed: 11/28/2022]
|