1
|
Zhang J, Zhu Y, Zhang M, Yan J, Zheng Y, Yao L, Li Z, Shao Z, Chen Y. Potassium channels in depression: emerging roles and potential targets. Cell Biosci 2024; 14:136. [PMID: 39529121 PMCID: PMC11555980 DOI: 10.1186/s13578-024-01319-0] [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: 09/13/2024] [Accepted: 10/31/2024] [Indexed: 11/16/2024] Open
Abstract
Potassium ion channels play a fundamental role in regulating cell membrane repolarization, modulating the frequency and shape of action potentials, and maintaining the resting membrane potential. A growing number of studies have indicated that dysfunction in potassium channels associates with the pathogenesis and treatment of depression. However, the involvement of potassium channels in the onset and treatment of depression has not been thoroughly summarized. In this review, we performed a comprehensive analysis of the association between multiple potassium channels and their roles in depression, and compiles the SNP loci of potassium channels associated with depression, as well as antidepressant drugs that target these channels. We discussed the pivotal role of potassium channels in the treatment of depression, provide valuable insights into new therapeutic targets for antidepressant treatment and critical clues to future drug discovery.
Collapse
Affiliation(s)
- Jiahao Zhang
- Institute of Acupuncture and Moxibustion, Shandong University of Traditional Chinese Medicine, Jinan, 250355, China
| | - Yao Zhu
- Institute of Acupuncture and Moxibustion, Shandong University of Traditional Chinese Medicine, Jinan, 250355, China
| | - Meng Zhang
- Institute of Acupuncture and Moxibustion, Shandong University of Traditional Chinese Medicine, Jinan, 250355, China
- Shandong Key Laboratory of Innovation and Application Research in Basic Theory of Traditional Chinese Medicine, Shandong University of Traditional Chinese Medicine, Jinan, 250355, China
| | - Jinglan Yan
- Institute of Acupuncture and Moxibustion, Shandong University of Traditional Chinese Medicine, Jinan, 250355, China
- Key Laboratory of Traditional Chinese Medicine Classical Theory, Ministry of Education, Shandong University of Traditional Chinese Medicine, Jinan, 250355, China
| | - Yuanjia Zheng
- Institute of Acupuncture and Moxibustion, Shandong University of Traditional Chinese Medicine, Jinan, 250355, China
- Key Laboratory of Traditional Chinese Medicine Classical Theory, Ministry of Education, Shandong University of Traditional Chinese Medicine, Jinan, 250355, China
| | - Lin Yao
- Institute of Acupuncture and Moxibustion, Shandong University of Traditional Chinese Medicine, Jinan, 250355, China
- Shandong Provincial Engineering Research Center for the Prevention and Treatment of Major Brain Diseases with Traditional Chinese Medicine, Shandong University of Traditional Chinese Medicine, Shandong University of Traditional Chinese Medicine, Jinan, 250355, China
| | - Ziwei Li
- Institute of Acupuncture and Moxibustion, Shandong University of Traditional Chinese Medicine, Jinan, 250355, China
| | - Zihan Shao
- Institute of Acupuncture and Moxibustion, Shandong University of Traditional Chinese Medicine, Jinan, 250355, China
| | - Yongjun Chen
- Institute of Acupuncture and Moxibustion, Shandong University of Traditional Chinese Medicine, Jinan, 250355, China.
- Shandong Key Laboratory of Innovation and Application Research in Basic Theory of Traditional Chinese Medicine, Shandong University of Traditional Chinese Medicine, Jinan, 250355, China.
- Shandong Provincial Engineering Research Center for the Prevention and Treatment of Major Brain Diseases with Traditional Chinese Medicine, Shandong University of Traditional Chinese Medicine, Shandong University of Traditional Chinese Medicine, Jinan, 250355, China.
| |
Collapse
|
2
|
Chakraborty P, Hasan G. ER-Ca 2+ stores and the regulation of store-operated Ca 2+ entry in neurons. J Physiol 2024; 602:1463-1474. [PMID: 36691983 DOI: 10.1113/jp283827] [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: 11/18/2022] [Accepted: 01/16/2023] [Indexed: 01/25/2023] Open
Abstract
Key components of endoplasmic reticulum (ER) Ca2+ release and store-operated Ca2+ entry (SOCE) are likely expressed in all metazoan cells. Due to the complexity of canonical Ca2+ entry mechanisms in neurons, the functional significance of ER-Ca2+ release and SOCE has been difficult to identify and establish. In this review we present evidence of how these two related mechanisms of Ca2+ signalling impact multiple aspects of neuronal physiology and discuss their interaction with the better understood classes of ion channels that are gated by either voltage changes or extracellular ligands in neurons. Given how a small imbalance in Ca2+ homeostasis can have strongly detrimental effects on neurons, leading to cell death, it is essential that neuronal SOCE is carefully regulated. We go on to discuss some mechanisms of SOCE regulation that have been identified in Drosophila and mammalian neurons. These include specific splice variants of stromal interaction molecules, different classes of membrane-interacting proteins and an ER-Ca2+ channel. So far these appear distinct from the mechanisms of SOCE regulation identified in non-excitable cells. Finally, we touch upon the significance of these studies in the context of certain human neurodegenerative diseases.
Collapse
Affiliation(s)
- Pragnya Chakraborty
- National Centre for Biological Sciences, Tata Institute of Fundamental Research, Bangalore, India
- SASTRA University, Thanjavur, Tamil Nadu, India
| | - Gaiti Hasan
- National Centre for Biological Sciences, Tata Institute of Fundamental Research, Bangalore, India
| |
Collapse
|
3
|
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
|
4
|
Zhang XQ, Xu L, Zhu XY, Tang ZH, Dong YB, Yu ZP, Shang Q, Wang ZC, Shen HW. D-serine reconstitutes synaptic and intrinsic inhibitory control of pyramidal neurons in a neurodevelopmental mouse model for schizophrenia. Nat Commun 2023; 14:8255. [PMID: 38086803 PMCID: PMC10716516 DOI: 10.1038/s41467-023-43930-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2022] [Accepted: 11/24/2023] [Indexed: 12/18/2023] Open
Abstract
The hypothesis of N-methyl-D-aspartate receptor (NMDAR) dysfunction for cognitive impairment in schizophrenia constitutes the theoretical basis for the translational application of NMDAR co-agonist D-serine or its analogs. However, the cellular mechanism underlying the therapeutic effect of D-serine remains unclear. In this study, we utilize a mouse neurodevelopmental model for schizophrenia that mimics prenatal pathogenesis and exhibits hypoexcitability of parvalbumin-positive (PV) neurons, as well as PV-preferential NMDAR dysfunction. We find that D-serine restores excitation/inhibition balance by reconstituting both synaptic and intrinsic inhibitory control of cingulate pyramidal neurons through facilitating PV excitability and activating small-conductance Ca2+-activated K+ (SK) channels in pyramidal neurons, respectively. Either amplifying inhibitory drive via directly strengthening PV neuron activity or inhibiting pyramidal excitability via activating SK channels is sufficient to improve cognitive function in this model. These findings unveil a dual mechanism for how D-serine improves cognitive function in this model.
Collapse
Affiliation(s)
- Xiao-Qin Zhang
- Department of Pharmacology, School of Medicine, Ningbo University, 818 Fenghua Rd, Ningbo, Zhejiang, 315211, China
| | - Le Xu
- Department of Pharmacology, School of Medicine, Ningbo University, 818 Fenghua Rd, Ningbo, Zhejiang, 315211, China
| | - Xin-Yi Zhu
- Department of Pharmacology, School of Medicine, Ningbo University, 818 Fenghua Rd, Ningbo, Zhejiang, 315211, China
| | - Zi-Hang Tang
- Department of Pharmacology, School of Medicine, Ningbo University, 818 Fenghua Rd, Ningbo, Zhejiang, 315211, China
| | - Yi-Bei Dong
- Department of Pharmacology, School of Medicine, Ningbo University, 818 Fenghua Rd, Ningbo, Zhejiang, 315211, China
| | - Zhi-Peng Yu
- Department of Pharmacology, School of Medicine, Ningbo University, 818 Fenghua Rd, Ningbo, Zhejiang, 315211, China
| | - Qing Shang
- Department of Neurology, The First Affiliated Hospital of Ningbo University, 59 Liuting Street, Haishu District, Ningbo, Zhejiang, 315211, China
| | - Zheng-Chun Wang
- Department of Pharmacology, School of Medicine, Ningbo University, 818 Fenghua Rd, Ningbo, Zhejiang, 315211, China
| | - Hao-Wei Shen
- Department of Pharmacology, School of Medicine, Ningbo University, 818 Fenghua Rd, Ningbo, Zhejiang, 315211, China.
| |
Collapse
|
5
|
Rodrigues YE, Tigaret CM, Marie H, O'Donnell C, Veltz R. A stochastic model of hippocampal synaptic plasticity with geometrical readout of enzyme dynamics. eLife 2023; 12:e80152. [PMID: 37589251 PMCID: PMC10435238 DOI: 10.7554/elife.80152] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2022] [Accepted: 03/22/2023] [Indexed: 08/18/2023] Open
Abstract
Discovering the rules of synaptic plasticity is an important step for understanding brain learning. Existing plasticity models are either (1) top-down and interpretable, but not flexible enough to account for experimental data, or (2) bottom-up and biologically realistic, but too intricate to interpret and hard to fit to data. To avoid the shortcomings of these approaches, we present a new plasticity rule based on a geometrical readout mechanism that flexibly maps synaptic enzyme dynamics to predict plasticity outcomes. We apply this readout to a multi-timescale model of hippocampal synaptic plasticity induction that includes electrical dynamics, calcium, CaMKII and calcineurin, and accurate representation of intrinsic noise sources. Using a single set of model parameters, we demonstrate the robustness of this plasticity rule by reproducing nine published ex vivo experiments covering various spike-timing and frequency-dependent plasticity induction protocols, animal ages, and experimental conditions. Our model also predicts that in vivo-like spike timing irregularity strongly shapes plasticity outcome. This geometrical readout modelling approach can be readily applied to other excitatory or inhibitory synapses to discover their synaptic plasticity rules.
Collapse
Affiliation(s)
- Yuri Elias Rodrigues
- Université Côte d’AzurNiceFrance
- Institut de Pharmacologie Moléculaire et Cellulaire (IPMC), CNRSValbonneFrance
- Inria Center of University Côte d’Azur (Inria)Sophia AntipolisFrance
| | - Cezar M Tigaret
- Neuroscience and Mental Health Research Innovation Institute, Division of Psychological Medicine and Clinical Neurosciences,School of Medicine, Cardiff UniversityCardiffUnited Kingdom
| | - Hélène Marie
- Université Côte d’AzurNiceFrance
- Institut de Pharmacologie Moléculaire et Cellulaire (IPMC), CNRSValbonneFrance
| | - Cian O'Donnell
- School of Computing, Engineering, and Intelligent Systems, Magee Campus, Ulster UniversityLondonderryUnited Kingdom
- School of Computer Science, Electrical and Electronic Engineering, and Engineering Mathematics, University of BristolBristolUnited Kingdom
| | - Romain Veltz
- Inria Center of University Côte d’Azur (Inria)Sophia AntipolisFrance
| |
Collapse
|
6
|
Liu T, Li T, Xu D, Wang Y, Zhou Y, Wan J, Huang CLH, Tan X. Small-conductance calcium-activated potassium channels in the heart: expression, regulation and pathological implications. Philos Trans R Soc Lond B Biol Sci 2023; 378:20220171. [PMID: 37122223 PMCID: PMC10150224 DOI: 10.1098/rstb.2022.0171] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2022] [Accepted: 12/25/2022] [Indexed: 05/02/2023] Open
Abstract
Ca2+-activated K+ channels are critical to cellular Ca2+ homeostasis and excitability; they couple intracellular Ca2+ and membrane voltage change. Of these, the small, 4-14 pS, conductance SK channels include three, KCNN1-3 encoded, SK1/KCa2.1, SK2/KCa2.2 and SK3/KCa2.3, channel subtypes with characteristic, EC50 ∼ 10 nM, 40 pM, 1 nM, apamin sensitivities. All SK channels, particularly SK2 channels, are expressed in atrial, ventricular and conducting system cardiomyocytes. Pharmacological and genetic modification results have suggested that SK channel block or knockout prolonged action potential durations (APDs) and effective refractory periods (ERPs) particularly in atrial, but also in ventricular, and sinoatrial, atrioventricular node and Purkinje myocytes, correspondingly affect arrhythmic tendency. Additionally, mitochondrial SK channels may decrease mitochondrial Ca2+ overload and reactive oxygen species generation. SK channels show low voltage but marked Ca2+ dependences (EC50 ∼ 300-500 nM) reflecting their α-subunit calmodulin (CaM) binding domains, through which they may be activated by voltage-gated or ryanodine-receptor Ca2+ channel activity. SK function also depends upon complex trafficking and expression processes and associations with other ion channels or subunits from different SK subtypes. Atrial and ventricular clinical arrhythmogenesis may follow both increased or decreased SK expression through decreased or increased APD correspondingly accelerating and stabilizing re-entrant rotors or increasing incidences of triggered activity. This article is part of the theme issue 'The heartbeat: its molecular basis and physiological mechanisms'.
Collapse
Affiliation(s)
- Ting Liu
- Key Laboratory of Medical Electrophysiology of Ministry of Education, Institute of Cardiovascular Research, Southwest Medical University, Luzhou, Sichuan 646000, People's Republic of China
| | - Tao Li
- Key Laboratory of Medical Electrophysiology of Ministry of Education, Institute of Cardiovascular Research, Southwest Medical University, Luzhou, Sichuan 646000, People's Republic of China
- Department of Cardiology, the Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan 646000, People's Republic of China
| | - Dandi Xu
- Key Laboratory of Medical Electrophysiology of Ministry of Education, Institute of Cardiovascular Research, Southwest Medical University, Luzhou, Sichuan 646000, People's Republic of China
| | - Yan Wang
- Key Laboratory of Medical Electrophysiology of Ministry of Education, Institute of Cardiovascular Research, Southwest Medical University, Luzhou, Sichuan 646000, People's Republic of China
| | - Yafei Zhou
- Key Laboratory of Medical Electrophysiology of Ministry of Education, Institute of Cardiovascular Research, Southwest Medical University, Luzhou, Sichuan 646000, People's Republic of China
| | - Juyi Wan
- Department of Cardiovascular Surgery, the Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan 646000, People's Republic of China
| | - Christopher L.-H. Huang
- Key Laboratory of Medical Electrophysiology of Ministry of Education, Institute of Cardiovascular Research, Southwest Medical University, Luzhou, Sichuan 646000, People's Republic of China
- Physiological Laboratory and Department of Biochemistry, University of Cambridge, Cambridge CB2 3EG, UK
| | - Xiaoqiu Tan
- Key Laboratory of Medical Electrophysiology of Ministry of Education, Institute of Cardiovascular Research, Southwest Medical University, Luzhou, Sichuan 646000, People's Republic of China
- Department of Cardiology, the Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan 646000, People's Republic of China
- Department of Physiology, School of Basic Medical Sciences, Southwest Medical University, Luzhou, Sichuan 646000, People's Republic of China
| |
Collapse
|
7
|
Vasileva VY, Khairullina ZM, Sudarikova AV, Chubinskiy-Nadezhdin VI. Role of Calcium-Activated Potassium Channels in Proliferation, Migration and Invasion of Human Chronic Myeloid Leukemia K562 Cells. MEMBRANES 2023; 13:583. [PMID: 37367787 DOI: 10.3390/membranes13060583] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/28/2023] [Revised: 06/02/2023] [Accepted: 06/02/2023] [Indexed: 06/28/2023]
Abstract
Calcium-activated potassium channels (KCa) are important participants in calcium signaling pathways due to their ability to be activated by an increase in intracellular free calcium concentration. KCa channels are involved in the regulation of cellular processes in both normal and pathophysiological conditions, including oncotransformation. Previously, using patch-clamp, we registered the KCa currents in the plasma membrane of human chronic myeloid leukemia K562 cells, whose activity was controlled by local Ca2+ entry via mechanosensitive calcium-permeable channels. Here, we performed the molecular and functional identification of KCa channels and have uncovered their role in the proliferation, migration and invasion of K562 cells. Using a combined approach, we identified the functional activity of SK2, SK3 and IK channels in the plasma membrane of the cells. Selective SK and IK channel inhibitors, apamin and TRAM-34, respectively, reduced the proliferative, migratory and invasive capabilities of human myeloid leukemia cells. At the same time, the viability of K562 cells was not affected by KCa channel inhibitors. Ca2+ imaging showed that both SK and IK channel inhibitors affect Ca2+ entry and this could underlie the observed suppression of pathophysiological reactions of K562 cells. Our data imply that SK/IK channel inhibitors could be used to slow down the proliferation and spreading of chronic myeloid leukemia K562 cells that express functionally active KCa channels in the plasma membrane.
Collapse
Affiliation(s)
- Valeria Y Vasileva
- Institute of Cytology, Russian Academy of Sciences, Tikhoretsky Ave. 4, 194064 Saint-Petersburg, Russia
| | - Zuleikha M Khairullina
- Institute of Cytology, Russian Academy of Sciences, Tikhoretsky Ave. 4, 194064 Saint-Petersburg, Russia
| | - Anastasia V Sudarikova
- Institute of Cytology, Russian Academy of Sciences, Tikhoretsky Ave. 4, 194064 Saint-Petersburg, Russia
| | | |
Collapse
|
8
|
Moreno M, Minjarez C, Todorovic SM, Quillinan N. Distinct excitability of thalamocortical neurons correlates with the presence of cerebellar afferents. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.05.26.542536. [PMID: 37292810 PMCID: PMC10246008 DOI: 10.1101/2023.05.26.542536] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/10/2023]
Abstract
Thalamocortical (TC) neurons within the ventrolateral thalamus (VL) receive projections from the cerebellum and the basal ganglia (BG) to facilitate motor and non-motor functions. Tonic and rebound firing patterns in response to excitatory cerebellar and inhibitory BG inputs, respectively, are a canonical feature of TC neurons and plays a key role in signal processing. The intrinsic excitability of TC neurons has a strong influence on how they respond to synaptic inputs, however, it is unknown whether their afferents influence their firing properties. Understanding the input-specific firing patterns could shed light into movement disorders with cerebellar or BG involvement. Here, we used whole-cell electrophysiology in brain slices from C57BL/6 mice to investigate the firing of TC neurons with optogenetic confirmation of cerebellar or BG afferents. TC neurons with cerebellar afferents exhibited higher tonic and rebound firing rates than those with BG afferents. This increased firing was associated with faster action potential depolarization kinetics and a smaller afterhyperpolarization potential. We also found differences in the passive membrane properties and sag currents during hyperpolarization. Despite higher rebound firing in TC neurons with cerebellar afferents, there were no differences in T-type calcium channel function compared to those with BG inputs. These data suggest input-specific differences in sodium and SK, but not T-type calcium channels, impact firing properties in TC populations. Altogether, we showed that the pronounced divergence observed in TC neuron firing properties correlate with its heterogeneous anatomical connectivity, which could signify a distinct signal integration and processing by these neurons. Keypoints Thalamocortical neurons in the VL with cerebellar afferents have higher intrinsic tonic and rebound firing properties than those with basal ganglia afferents.Membrane resistance and action potential depolarization slope were different based on the presence of cerebellar afferents.Despite elevated rebound burst firing, T-type mediated currents did not correlate with increased firing in neurons with cerebellar afferents.
Collapse
|
9
|
Guo R, Cui M, Li X, Wu M, Xu F, Zhang Y, Wang C, Feng P, Wang J, Huo S, Luo Z, Xing R, Gu J, Shi X, Liu Y, Wang L. Design, synthesis and biological evaluation of pyrrolopyrimidine derivatives as novel and selective positive modulator of the small conductance Ca 2+-activated K + channels. Eur J Med Chem 2023; 254:115353. [PMID: 37068385 DOI: 10.1016/j.ejmech.2023.115353] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2023] [Revised: 03/31/2023] [Accepted: 04/04/2023] [Indexed: 04/19/2023]
Abstract
The type 2 small conductance Ca2+-activated K+ channels (SK2) have been considered as one of the most promising therapeutic targets for spinocerebellar ataxias type 2 (SCA2) by playing a critical role in the control of normal purkinje cells (PCs) pacemaking. Herein, a novel series of pyrrolopyrimidine derivatives were designed and synthesized from the lead compound NS13001 as subtype-selective modulators of SK channels. Among them, the halogen-substituted compound 12b (EC50 = 0.34 ± 0.044 μM) was identified with a ∼5.4-fold higher potency on potentiating SK2-a channels at submicromolar concentrations as compared to NS13001 (EC50 = 1.83 ± 0.50 μM). Furthermore, compound 12b exhibited selectivity on SK2-a/SK3 subtype by displaying 93.33 ± 3.26% efficacies on SK2-a channels, and 84.54% ± 7.49% on SK3 channels. In addition, compound 12b demonstrated the potential to cross the blood-brain barrier (BBB) with suitable pharmacokinetic properties and low cytotoxicity. Molecular docking study also unveiled the binding interactions of compound 12b with SK2-CaM protein complex. Overall, the novel pyrrolopyrimidines provide an insightful guidance for future structural optimization of SK channel agonists.
Collapse
Affiliation(s)
- Ran Guo
- Department of Medicinal Chemistry, School of Pharmacy, Hebei Medical University, Shijiazhuang, 050017, PR China
| | - Miao Cui
- Department of Medicinal Chemistry, School of Pharmacy, Hebei Medical University, Shijiazhuang, 050017, PR China
| | - Xiaojing Li
- Department of Medicinal Chemistry, School of Pharmacy, Hebei Medical University, Shijiazhuang, 050017, PR China
| | - Mengqi Wu
- Department of Medicinal Chemistry, School of Pharmacy, Hebei Medical University, Shijiazhuang, 050017, PR China
| | - Fei Xu
- Department of Medicinal Chemistry, School of Pharmacy, Hebei Medical University, Shijiazhuang, 050017, PR China
| | - Yining Zhang
- Department of Medicinal Chemistry, School of Pharmacy, Hebei Medical University, Shijiazhuang, 050017, PR China
| | - Chun Wang
- Department of Medicinal Chemistry, School of Pharmacy, Hebei Medical University, Shijiazhuang, 050017, PR China
| | - Penglei Feng
- Department of Medicinal Chemistry, School of Pharmacy, Hebei Medical University, Shijiazhuang, 050017, PR China
| | - Jianchao Wang
- Department of Medicinal Chemistry, School of Pharmacy, Hebei Medical University, Shijiazhuang, 050017, PR China
| | - Sijia Huo
- Department of Medicinal Chemistry, School of Pharmacy, Hebei Medical University, Shijiazhuang, 050017, PR China
| | - Zijun Luo
- Department of Medicinal Chemistry, School of Pharmacy, Hebei Medical University, Shijiazhuang, 050017, PR China
| | - Ruijuan Xing
- Department of Medicinal Chemistry, School of Pharmacy, Hebei Medical University, Shijiazhuang, 050017, PR China
| | - Jianmin Gu
- Department of Medicinal Chemistry, School of Pharmacy, Hebei Medical University, Shijiazhuang, 050017, PR China
| | - Xiaowei Shi
- Department of Medicinal Chemistry, School of Pharmacy, Hebei Medical University, Shijiazhuang, 050017, PR China.
| | - Yi Liu
- Department of Toxicology, School of Public Health, Hebei Medical University, Shijiazhuang, 050017, PR China; Hebei Province Key Laboratory of Environment and Human Health, Shijiazhuang, 050017, PR China.
| | - Lei Wang
- Department of Medicinal Chemistry, School of Pharmacy, Hebei Medical University, Shijiazhuang, 050017, PR China; Hebei Province Key Laboratory of Innovative Drug Research and Evaluation, Shijiazhuang, 050017, PR China.
| |
Collapse
|
10
|
van Herck IGM, Seutin V, Bentzen BH, Marrion NV, Edwards AG. Gating kinetics and pharmacological properties of small-conductance Ca 2+-activated potassium channels. Biophys J 2023; 122:1143-1157. [PMID: 36760125 PMCID: PMC10111258 DOI: 10.1016/j.bpj.2023.02.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2022] [Revised: 11/20/2022] [Accepted: 02/06/2023] [Indexed: 02/11/2023] Open
Abstract
Small-conductance (SK) calcium-activated potassium channels are a promising treatment target in atrial fibrillation. However, the functional properties that differentiate SK inhibitors remain poorly understood. The objective of this study was to determine how two unrelated SK channel inhibitors, apamin and AP14145, impact SK channel function in excised inside-out single-channel recordings. Surprisingly, both apamin and AP14145 exert much of their inhibition by inducing a class of very-long-lived channel closures (apamin: τc,vl = 11.8 ± 7.1 s, and AP14145: τc,vl = 10.3 ± 7.2 s), which were never observed under control conditions. Both inhibitors also induced changes to the three closed and two open durations typical of normal SK channel gating. AP14145 shifted the open duration distribution to favor longer open durations, whereas apamin did not alter open-state kinetics. AP14145 also prolonged the two shortest channel closed durations (AP14145: τc,s = 3.50 ± 0.81 ms, and τc,i = 32.0 ± 6.76 ms versus control: τc,s = 1.59 ± 0.19 ms, and τc,i = 13.5 ± 1.17 ms), thus slowing overall gating kinetics within bursts of channel activity. In contrast, apamin accelerated intraburst gating kinetics by shortening the two shortest closed durations (τc,s = 0.75 ± 0.10 ms and τc,i = 5.08 ± 0.49 ms) and inducing periods of flickery activity. Finally, AP14145 introduced a unique form of inhibition by decreasing unitary current amplitude. SK channels exhibited two clearly distinguishable amplitudes (control: Ahigh = 0.76 ± 0.03 pA, and Alow = 0.54 ± 0.03 pA). AP14145 both reduced the fraction of patches exhibiting the higher amplitude (AP14145: 4/9 patches versus control: 16/16 patches) and reduced the mean low amplitude (0.38 ± 0.03 pA). Here, we have demonstrated that both inhibitors introduce very long channel closures but that each also exhibits unique effects on other components of SK gating kinetics and unitary current. The combination of these effects is likely to be critical for understanding the functional differences of each inhibitor in the context of cyclical Ca2+-dependent channel activation in vivo.
Collapse
Affiliation(s)
- Ilsbeth G M van Herck
- Computational Physiology Department, Simula Research Laboratory, Oslo, Norway; Institute of Informatics, University of Oslo, Oslo, Norway
| | - Vincent Seutin
- Neurophysiology Unit, GIGA Neurosciences, University of Liège, Liège, Belgium
| | - Bo H Bentzen
- Acesion Pharma, Copenhagen, Denmark; Biomedical Institute, University of Copenhagen, Copenhagen, Denmark
| | - Neil V Marrion
- School of Physiology, Pharmacology and Neuroscience, University of Bristol, Bristol, UK
| | - Andrew G Edwards
- Computational Physiology Department, Simula Research Laboratory, Oslo, Norway; Department of Pharmacology, University of California, Davis, California.
| |
Collapse
|
11
|
Soret B, Hense J, Lüdtke S, Thale I, Schwab A, Düfer M. Pancreatic K Ca3.1 channels in health and disease. Biol Chem 2023; 404:339-353. [PMID: 36571487 DOI: 10.1515/hsz-2022-0232] [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: 07/15/2022] [Accepted: 11/24/2022] [Indexed: 12/27/2022]
Abstract
Ion channels play an important role for regulation of the exocrine and the endocrine pancreas. This review focuses on the Ca2+-regulated K+ channel KCa3.1, encoded by the KCNN4 gene, which is present in both parts of the pancreas. In the islets of Langerhans, KCa3.1 channels are involved in the regulation of membrane potential oscillations characterizing nutrient-stimulated islet activity. Channel upregulation is induced by gluco- or lipotoxic conditions and might contribute to micro-inflammation and impaired insulin release in type 2 diabetes mellitus as well as to diabetes-associated renal and vascular complications. In the exocrine pancreas KCa3.1 channels are expressed in acinar and ductal cells. They are thought to play a role for anion secretion during digestion but their physiological role has not been fully elucidated yet. Pancreatic carcinoma, especially pancreatic ductal adenocarcinoma (PDAC), is associated with drastic overexpression of KCa3.1. For pharmacological targeting of KCa3.1 channels, we are discussing the possible benefits KCa3.1 channel inhibitors might provide in the context of diabetes mellitus and pancreatic cancer, respectively. We are also giving a perspective for the use of a fluorescently labeled derivative of the KCa3.1 blocker senicapoc as a tool to monitor channel distribution in pancreatic tissue. In summary, modulating KCa3.1 channel activity is a useful strategy for exo-and endocrine pancreatic disease but further studies are needed to evaluate its clinical suitability.
Collapse
Affiliation(s)
- Benjamin Soret
- University of Münster, Institute of Physiology II, Robert-Koch-Straße 27b, D-48149 Münster, Germany
| | - Jurek Hense
- University of Münster, Institute of Pharmaceutical and Medicinal Chemistry, Department of Pharmacology, Corrensstraße 48, D-48149 Münster, Germany
| | - Simon Lüdtke
- University of Münster, Institute of Pharmaceutical and Medicinal Chemistry, Department of Pharmacology, Corrensstraße 48, D-48149 Münster, Germany
| | - Insa Thale
- University of Münster, Institute of Pharmaceutical and Medicinal Chemistry, Corrensstraße 48, D-48149 Münster, Germany
| | - Albrecht Schwab
- University of Münster, Institute of Physiology II, Robert-Koch-Straße 27b, D-48149 Münster, Germany
| | - Martina Düfer
- University of Münster, Institute of Pharmaceutical and Medicinal Chemistry, Department of Pharmacology, Corrensstraße 48, D-48149 Münster, Germany
| |
Collapse
|
12
|
Numata T, Sato-Numata K, Yoshino M. Intermediate conductance Ca 2+-activated potassium channels are activated by functional coupling with stretch-activated nonselective cation channels in cricket myocytes. FRONTIERS IN INSECT SCIENCE 2023; 2:1100671. [PMID: 38468799 PMCID: PMC10926553 DOI: 10.3389/finsc.2022.1100671] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 11/17/2022] [Accepted: 12/20/2022] [Indexed: 03/13/2024]
Abstract
Cooperative gating of localized ion channels ranges from fine-tuning excitation-contraction coupling in muscle cells to controlling pace-making activity in the heart. Membrane deformation resulting from muscle contraction activates stretch-activated (SA) cation channels. The subsequent Ca2+ influx activates spatially localized Ca2+-sensitive K+ channels to fine-tune spontaneous muscle contraction. To characterize endogenously expressed intermediate conductance Ca2+-activated potassium (IK) channels and assess the functional relevance of the extracellular Ca2+ source leading to IK channel activity, we performed patch-clamp techniques on cricket oviduct myocytes and recorded single-channel data. In this study, we first investigated the identification of IK channels that could be distinguished from endogenously expressed large-conductance Ca2+-activated potassium (BK) channels by adding extracellular Ba2+. The single-channel conductance of the IK channel was 62 pS, and its activity increased with increasing intracellular Ca2+ concentration but was not voltage-dependent. These results indicated that IK channels are endogenously expressed in cricket oviduct myocytes. Second, the Ca2+ influx pathway that activates the IK channel was investigated. The absence of extracellular Ca2+ or the presence of Gd3+ abolished the activity of IK channels. Finally, we investigated the proximity between SA and IK channels. The removal of extracellular Ca2+, administration of Ca2+ to the microscopic region in a pipette, and application of membrane stretching stimulation increased SA channel activity, followed by IK channel activity. Membrane stretch-induced SA and IK channel activity were positively correlated. However, the emergence of IK channel activity and its increase in response to membrane mechanical stretch was not observed without Ca2+ in the pipette. These results strongly suggest that IK channels are endogenously expressed in cricket oviduct myocytes and that IK channel activity is regulated by neighboring SA channel activity. In conclusion, functional coupling between SA and IK channels may underlie the molecular basis of spontaneous rhythmic contractions.
Collapse
Affiliation(s)
- Tomohiro Numata
- Department of Integrative Physiology, Graduate School of Medicine, Akita University, Akita, Japan
- Department of Biology, Tokyo Gakugei University, Tokyo, Japan
| | - Kaori Sato-Numata
- Department of Integrative Physiology, Graduate School of Medicine, Akita University, Akita, Japan
- Department of Biology, Tokyo Gakugei University, Tokyo, Japan
| | - Masami Yoshino
- Department of Biology, Tokyo Gakugei University, Tokyo, Japan
| |
Collapse
|
13
|
Thale I, Maskri S, Grey L, Todesca LM, Budde T, Maisuls I, Strassert CA, Koch O, Schwab A, Wünsch B. Imaging of K Ca 3.1 Channels in Tumor Cells with PET and Small-Molecule Fluorescent Probes. ChemMedChem 2023; 18:e202200551. [PMID: 36315933 PMCID: PMC10098740 DOI: 10.1002/cmdc.202200551] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2022] [Revised: 10/31/2022] [Indexed: 01/20/2023]
Abstract
The Ca2+ activated K+ channel KCa 3.1 is overexpressed in several human tumor cell lines, e. g. clear cell renal carcinoma, prostate cancer, non-small cell lung cancer. Highly aggressive cancer cells use this ion channel for key processes of the metastatic cascade such as migration, extravasation and invasion. Therefore, small molecules, which are able to image this KCa 3.1 channel in vitro and in vivo represent valuable diagnostic and prognostic tool compounds. The [18 F]fluoroethyltriazolyl substituted senicapoc was used as positron emission tomography (PET) tracer and showed promising properties for imaging of KCa 3.1 channels in lung adenocarcinoma cells in mice. The novel senicapoc BODIPY conjugates with two F-atoms (9 a) and with a F-atom and a methoxy moiety (9 b) at the B-atom led to the characteristic punctate staining pattern resulting from labeling of single KCa 3.1 channels in A549-3R cells. This punctate pattern was completely removed by preincubation with an excess of senicapoc confirming the high specificity of KCa 3.1 labeling. Due to the methoxy moiety at the B-atom and the additional oxyethylene unit in the spacer, 9 b exhibits higher polarity, which improves solubility and handling without reduction of fluorescence quantum yield. Docking studies using a cryo-electron microscopy (EM) structure of the KCa 3.1 channel confirmed the interaction of 9 a and 9 b with a binding pocket in the channel pore.
Collapse
Affiliation(s)
- Insa Thale
- Westfälische Wilhelms-Universität Münster, GRK 2515, Chemical biology of ion channels (Chembion), Corrensstraße 48, 48149, Münster, Germany.,Westfälische Wilhelms-Universität Münster, Institut für Pharmazeutische und Medizinische Chemie, Corrensstraße 48, 48149, Münster, Germany
| | - Sarah Maskri
- Westfälische Wilhelms-Universität Münster, GRK 2515, Chemical biology of ion channels (Chembion), Corrensstraße 48, 48149, Münster, Germany.,Westfälische Wilhelms-Universität Münster, Institut für Pharmazeutische und Medizinische Chemie, Corrensstraße 48, 48149, Münster, Germany
| | - Lucie Grey
- Westfälische Wilhelms-Universität Münster, Institut für Pharmazeutische und Medizinische Chemie, Corrensstraße 48, 48149, Münster, Germany
| | - Luca Matteo Todesca
- Westfälische Wilhelms-Universität Münster, GRK 2515, Chemical biology of ion channels (Chembion), Corrensstraße 48, 48149, Münster, Germany.,Westfälische Wilhelms-Universität Münster, Universitätsklinikum Münster, Institute of Physiology II, Robert-Koch-Straße 27b, 48149, Münster, Germany
| | - Thomas Budde
- Westfälische Wilhelms-Universität Münster, GRK 2515, Chemical biology of ion channels (Chembion), Corrensstraße 48, 48149, Münster, Germany.,Westfälische Wilhelms-Universität Münster, Universitätsklinikum Münster, Institute of Physiology I, Robert-Koch-Straße 27a, 48149, Münster, Germany
| | - Ivan Maisuls
- Westfälische Wilhelms-Universität Münster, Institut für Anorganische und Analytische Chemie CiMIC, SoN, Corrensstraße 28, 48149, Münster, Germany.,Westfälische Wilhelms-Universität Münster, CeNTech, Heisenbergstraße 11, 48149, Münster, Germany
| | - Cristian A Strassert
- Westfälische Wilhelms-Universität Münster, Institut für Anorganische und Analytische Chemie CiMIC, SoN, Corrensstraße 28, 48149, Münster, Germany.,Westfälische Wilhelms-Universität Münster, CeNTech, Heisenbergstraße 11, 48149, Münster, Germany
| | - Oliver Koch
- Westfälische Wilhelms-Universität Münster, GRK 2515, Chemical biology of ion channels (Chembion), Corrensstraße 48, 48149, Münster, Germany.,Westfälische Wilhelms-Universität Münster, Institut für Pharmazeutische und Medizinische Chemie, Corrensstraße 48, 48149, Münster, Germany
| | - Albrecht Schwab
- Westfälische Wilhelms-Universität Münster, GRK 2515, Chemical biology of ion channels (Chembion), Corrensstraße 48, 48149, Münster, Germany.,Westfälische Wilhelms-Universität Münster, Universitätsklinikum Münster, Institute of Physiology II, Robert-Koch-Straße 27b, 48149, Münster, Germany
| | - Bernhard Wünsch
- Westfälische Wilhelms-Universität Münster, GRK 2515, Chemical biology of ion channels (Chembion), Corrensstraße 48, 48149, Münster, Germany.,Westfälische Wilhelms-Universität Münster, Institut für Pharmazeutische und Medizinische Chemie, Corrensstraße 48, 48149, Münster, Germany
| |
Collapse
|
14
|
Ca 2+-Activated K + Channels and the Regulation of the Uteroplacental Circulation. Int J Mol Sci 2023; 24:ijms24021349. [PMID: 36674858 PMCID: PMC9867535 DOI: 10.3390/ijms24021349] [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: 12/14/2022] [Revised: 01/06/2023] [Accepted: 01/08/2023] [Indexed: 01/13/2023] Open
Abstract
Adequate uteroplacental blood supply is essential for the development and growth of the placenta and fetus during pregnancy. Aberrant uteroplacental perfusion is associated with pregnancy complications such as preeclampsia, fetal growth restriction (FGR), and gestational diabetes. The regulation of uteroplacental blood flow is thus vital to the well-being of the mother and fetus. Ca2+-activated K+ (KCa) channels of small, intermediate, and large conductance participate in setting and regulating the resting membrane potential of vascular smooth muscle cells (VSMCs) and endothelial cells (ECs) and play a critical role in controlling vascular tone and blood pressure. KCa channels are important mediators of estrogen/pregnancy-induced adaptive changes in the uteroplacental circulation. Activation of the channels hyperpolarizes uteroplacental VSMCs/ECs, leading to attenuated vascular tone, blunted vasopressor responses, and increased uteroplacental blood flow. However, the regulation of uteroplacental vascular function by KCa channels is compromised in pregnancy complications. This review intends to provide a comprehensive overview of roles of KCa channels in the regulation of the uteroplacental circulation under physiological and pathophysiological conditions.
Collapse
|
15
|
Dantas CG, da Paixão AO, Nunes TLGM, Silva IJF, dos S. Lima B, Araújo AAS, de Albuquerque-Junior RLC, Gramacho KP, Padilha FF, da Costa LP, Severino P, Cardoso JC, Souto EB, Gomes MZ. Africanized Bee Venom ( Apis mellifera Linnaeus): Neuroprotective Effects in a Parkinson's Disease Mouse Model Induced by 6-hydroxydopamine. TOXICS 2022; 10:583. [PMID: 36287863 PMCID: PMC9609968 DOI: 10.3390/toxics10100583] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 08/13/2022] [Revised: 09/24/2022] [Accepted: 09/29/2022] [Indexed: 06/16/2023]
Abstract
This study evaluated the neuroprotective effects of the Africanized bee venom (BV) and its mechanisms of action after 6-hydroxydopamine-(6-OHDA)-induced lesion in a mice model. Prior to BV treatment, mice received intrastriatal microinjections of 6-OHDA (no induced dopaminergic neuronal death) or ascorbate saline (as a control). BV was administered subcutaneously at different dosages (0.01, 0.05 or 0.1 mg·Kg-1) once every two days over a period of 3 weeks. The open field test was carried out, together with the immunohistochemical and histopathological analysis. The chemical composition of BV was also assessed, identifying the highest concentrations of apamin, phospholipase A2 and melittin. In the behavioral evaluation, the BV (0.1 mg·Kg-1) counteracted the 6-OHDA-induced decrease in crossings and rearing. 6-OHDA caused loss of dopaminergic cell bodies in the substantia nigra pars compacta and fibers in striatum (STR). Mice that received 0.01 mg·Kg-1 showed significant increase in the mean survival of dopaminergic cell bodies. Increased astrocytic infiltration occurred in the STR of 6-OHDA injected mice, differently from those of the groups treated with BV. The results suggested that Africanized BV has neuroprotective activity in an animal model of Parkinson's disease.
Collapse
Affiliation(s)
- Camila G. Dantas
- Institute of Research and Technology, Tiradentes University, Av. Murilo Dantas, 300, Aracaju 49032-490, Sergipe, Brazil
| | - Ailma O. da Paixão
- Institute of Research and Technology, Tiradentes University, Av. Murilo Dantas, 300, Aracaju 49032-490, Sergipe, Brazil
| | - Tássia L. G. M. Nunes
- Institute of Research and Technology, Tiradentes University, Av. Murilo Dantas, 300, Aracaju 49032-490, Sergipe, Brazil
| | - Italo J. F. Silva
- Institute of Research and Technology, Tiradentes University, Av. Murilo Dantas, 300, Aracaju 49032-490, Sergipe, Brazil
| | - Bruno dos S. Lima
- Department of Pharmacy, Federal University of Sergipe (U.F.S.), Cidade Universitária Prof. José Aloísio de Campos, Av. Marechal Rondon, Jardim Rosa Elze, São Cristóvão 49100-000, Sergipe, Brazil
| | - Adriano A. S. Araújo
- Department of Pharmacy, Federal University of Sergipe (U.F.S.), Cidade Universitária Prof. José Aloísio de Campos, Av. Marechal Rondon, Jardim Rosa Elze, São Cristóvão 49100-000, Sergipe, Brazil
| | | | - Kátia P. Gramacho
- Department of Animal Science, Rural Federal University of Semi-Árido (U.F.E.R.S.A), Av. Francisco Mota, Costa e Silva, Mossoró 49032-490, Natal, Brazil
| | - Francine F. Padilha
- Institute of Research and Technology, Tiradentes University, Av. Murilo Dantas, 300, Aracaju 49032-490, Sergipe, Brazil
| | - Luiz P. da Costa
- Post-Graduation Program in Chemistry, Federal University of Sergipe (U.F.S.), Cidade Universitária Prof. José Aloísio de Campos, Av. Marechal Rondon, Jardim Rosa Elze, São Cristóvão 49100-000, Sergipe, Brazil
| | - Patricia Severino
- Institute of Research and Technology, Tiradentes University, Av. Murilo Dantas, 300, Aracaju 49032-490, Sergipe, Brazil
| | - Juliana C. Cardoso
- Institute of Research and Technology, Tiradentes University, Av. Murilo Dantas, 300, Aracaju 49032-490, Sergipe, Brazil
| | - Eliana B. Souto
- Department of Pharmaceutical Technology, Faculty of Pharmacy, University of Porto, Rua de Jorge Viterbo Ferreira, 228, 4050-313 Porto, Portugal
- REQUIMTE/UCIBIO, Faculty of Pharmacy, University of Porto, Rua de Jorge Viterbo Ferreira, 228, 4050-313 Porto, Portugal
| | - Margarete Z. Gomes
- Department of Animal Science, Rural Federal University of Semi-Árido (U.F.E.R.S.A), Av. Francisco Mota, Costa e Silva, Mossoró 49032-490, Natal, Brazil
| |
Collapse
|
16
|
Dorman DB, Blackwell KT. Synaptic Plasticity Is Predicted by Spatiotemporal Firing Rate Patterns and Robust to In Vivo-like Variability. Biomolecules 2022; 12:1402. [PMID: 36291612 PMCID: PMC9599115 DOI: 10.3390/biom12101402] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2022] [Revised: 09/13/2022] [Accepted: 09/28/2022] [Indexed: 11/22/2022] Open
Abstract
Synaptic plasticity, the experience-induced change in connections between neurons, underlies learning and memory in the brain. Most of our understanding of synaptic plasticity derives from in vitro experiments with precisely repeated stimulus patterns; however, neurons exhibit significant variability in vivo during repeated experiences. Further, the spatial pattern of synaptic inputs to the dendritic tree influences synaptic plasticity, yet is not considered in most synaptic plasticity rules. Here, we investigate how spatiotemporal synaptic input patterns produce plasticity with in vivo-like conditions using a data-driven computational model with a plasticity rule based on calcium dynamics. Using in vivo spike train recordings as inputs to different size clusters of spines, we show that plasticity is strongly robust to trial-to-trial variability of spike timing. In addition, we derive general synaptic plasticity rules describing how spatiotemporal patterns of synaptic inputs control the magnitude and direction of plasticity. Synapses that strongly potentiated have greater firing rates and calcium concentration later in the trial, whereas strongly depressing synapses have hiring firing rates early in the trial. The neighboring synaptic activity influences the direction and magnitude of synaptic plasticity, with small clusters of spines producing the greatest increase in synaptic strength. Together, our results reveal that calcium dynamics can unify diverse plasticity rules and reveal how spatiotemporal firing rate patterns control synaptic plasticity.
Collapse
Affiliation(s)
- Daniel B. Dorman
- Interdisciplinary Program in Neuroscience, George Mason University, Fairfax, VA 22030, USA
| | - Kim T. Blackwell
- Interdisciplinary Program in Neuroscience, George Mason University, Fairfax, VA 22030, USA
- Department of Bioengineering, Volgenau School of Engineering, George Mason University, Fairfax, VA 22030, USA
| |
Collapse
|
17
|
Metabolic regulation and dysregulation of endothelial small conductance calcium activated potassium channels. Eur J Cell Biol 2022; 101:151208. [DOI: 10.1016/j.ejcb.2022.151208] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2021] [Revised: 02/06/2022] [Accepted: 02/07/2022] [Indexed: 12/13/2022] Open
|
18
|
Tiffner A, Hopl V, Schober R, Sallinger M, Grabmayr H, Höglinger C, Fahrner M, Lunz V, Maltan L, Frischauf I, Krivic D, Bhardwaj R, Schindl R, Hediger MA, Derler I. Orai1 Boosts SK3 Channel Activation. Cancers (Basel) 2021; 13:6357. [PMID: 34944977 PMCID: PMC8699475 DOI: 10.3390/cancers13246357] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2021] [Accepted: 12/13/2021] [Indexed: 12/20/2022] Open
Abstract
The interplay of SK3, a Ca2+ sensitive K+ ion channel, with Orai1, a Ca2+ ion channel, has been reported to increase cytosolic Ca2+ levels, thereby triggering proliferation of breast and colon cancer cells, although a molecular mechanism has remained elusive to date. We show in the current study, via heterologous protein expression, that Orai1 can enhance SK3 K+ currents, in addition to constitutively bound calmodulin (CaM). At low cytosolic Ca2+ levels that decrease SK3 K+ permeation, co-expressed Orai1 potentiates SK3 currents. This positive feedback mechanism of SK3 and Orai1 is enabled by their close co-localization. Remarkably, we discovered that loss of SK3 channel activity due to overexpressed CaM mutants could be restored by Orai1, likely via its interplay with the SK3-CaM binding site. Mapping for interaction sites within Orai1, we identified that the cytosolic strands and pore residues are critical for a functional communication with SK3. Moreover, STIM1 has a bimodal role in SK3-Orai1 regulation. Under physiological ionic conditions, STIM1 is able to impede SK3-Orai1 interplay by significantly decreasing their co-localization. Forced STIM1-Orai1 activity and associated Ca2+ influx promote SK3 K+ currents. The dynamic regulation of Orai1 to boost endogenous SK3 channels was also determined in the human prostate cancer cell line LNCaP.
Collapse
Affiliation(s)
- Adéla Tiffner
- JKU Life Science Center, Institute of Biophysics, Johannes Kepler University Linz, A-4020 Linz, Austria; (A.T.); (V.H.); (R.S.); (M.S.); (H.G.); (C.H.); (M.F.); (V.L.); (L.M.); (I.F.)
| | - Valentina Hopl
- JKU Life Science Center, Institute of Biophysics, Johannes Kepler University Linz, A-4020 Linz, Austria; (A.T.); (V.H.); (R.S.); (M.S.); (H.G.); (C.H.); (M.F.); (V.L.); (L.M.); (I.F.)
| | - Romana Schober
- JKU Life Science Center, Institute of Biophysics, Johannes Kepler University Linz, A-4020 Linz, Austria; (A.T.); (V.H.); (R.S.); (M.S.); (H.G.); (C.H.); (M.F.); (V.L.); (L.M.); (I.F.)
- Gottfried Schatz Research Centre, Medical University of Graz, A-8010 Graz, Austria; (D.K.); (R.S.)
| | - Matthias Sallinger
- JKU Life Science Center, Institute of Biophysics, Johannes Kepler University Linz, A-4020 Linz, Austria; (A.T.); (V.H.); (R.S.); (M.S.); (H.G.); (C.H.); (M.F.); (V.L.); (L.M.); (I.F.)
| | - Herwig Grabmayr
- JKU Life Science Center, Institute of Biophysics, Johannes Kepler University Linz, A-4020 Linz, Austria; (A.T.); (V.H.); (R.S.); (M.S.); (H.G.); (C.H.); (M.F.); (V.L.); (L.M.); (I.F.)
| | - Carmen Höglinger
- JKU Life Science Center, Institute of Biophysics, Johannes Kepler University Linz, A-4020 Linz, Austria; (A.T.); (V.H.); (R.S.); (M.S.); (H.G.); (C.H.); (M.F.); (V.L.); (L.M.); (I.F.)
| | - Marc Fahrner
- JKU Life Science Center, Institute of Biophysics, Johannes Kepler University Linz, A-4020 Linz, Austria; (A.T.); (V.H.); (R.S.); (M.S.); (H.G.); (C.H.); (M.F.); (V.L.); (L.M.); (I.F.)
| | - Victoria Lunz
- JKU Life Science Center, Institute of Biophysics, Johannes Kepler University Linz, A-4020 Linz, Austria; (A.T.); (V.H.); (R.S.); (M.S.); (H.G.); (C.H.); (M.F.); (V.L.); (L.M.); (I.F.)
| | - Lena Maltan
- JKU Life Science Center, Institute of Biophysics, Johannes Kepler University Linz, A-4020 Linz, Austria; (A.T.); (V.H.); (R.S.); (M.S.); (H.G.); (C.H.); (M.F.); (V.L.); (L.M.); (I.F.)
| | - Irene Frischauf
- JKU Life Science Center, Institute of Biophysics, Johannes Kepler University Linz, A-4020 Linz, Austria; (A.T.); (V.H.); (R.S.); (M.S.); (H.G.); (C.H.); (M.F.); (V.L.); (L.M.); (I.F.)
| | - Denis Krivic
- Gottfried Schatz Research Centre, Medical University of Graz, A-8010 Graz, Austria; (D.K.); (R.S.)
| | - Rajesh Bhardwaj
- Department of Nephrology and Hypertension, University of Bern, Inselspital, Freiburgstrasse 15, CH-3010 Bern, Switzerland; (R.B.); (M.A.H.)
- Department of Biomedical Research, University of Bern, Inselspital, Freiburgstrasse 15, CH-3010 Bern, Switzerland
| | - Rainer Schindl
- Gottfried Schatz Research Centre, Medical University of Graz, A-8010 Graz, Austria; (D.K.); (R.S.)
| | - Matthias A. Hediger
- Department of Nephrology and Hypertension, University of Bern, Inselspital, Freiburgstrasse 15, CH-3010 Bern, Switzerland; (R.B.); (M.A.H.)
- Department of Biomedical Research, University of Bern, Inselspital, Freiburgstrasse 15, CH-3010 Bern, Switzerland
| | - Isabella Derler
- JKU Life Science Center, Institute of Biophysics, Johannes Kepler University Linz, A-4020 Linz, Austria; (A.T.); (V.H.); (R.S.); (M.S.); (H.G.); (C.H.); (M.F.); (V.L.); (L.M.); (I.F.)
| |
Collapse
|
19
|
Daniel NH, Aravind A, Thakur P. Are ion channels potential therapeutic targets for Parkinson's disease? Neurotoxicology 2021; 87:243-257. [PMID: 34699791 DOI: 10.1016/j.neuro.2021.10.008] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2021] [Revised: 10/15/2021] [Accepted: 10/21/2021] [Indexed: 01/31/2023]
Abstract
Parkinson's disease (PD) is primarily associated with the progressive neurodegeneration of the dopaminergic neurons in the substantia nigra region of the brain. The resulting motor symptoms are managed with the help of dopamine replacement therapies. However, these therapeutics do not prevent the neurodegeneration underlying the disease and therefore lose their effectiveness in managing disease symptoms over time. Thus, there is an urgent need to develop newer therapeutics for the benefit of patients. The release of dopamine and the firing activity of substantia nigra neurons is regulated by several ion channels that act in concert. Dysregulations of these channels cause the aberrant movement of various ions in the intracellular milieu. This eventually leads to disruption of intracellular signalling cascades, alterations in cellular homeostasis, and bioenergetic deficits. Therefore, ion channels play a central role in driving the high vulnerability of dopaminergic neurons to degenerate during PD. Targeting ion channels offers an attractive mechanistic strategy to combat the process of neurodegeneration. In this review, we highlight the evidence pointing to the role of various ion channels in driving the PD processes. In addition, we also discuss the various drugs or compounds that target the ion channels and have shown neuroprotective potential in the in-vitro and in-vivo models of PD. We also discuss the current clinical status of various drugs targeting the ion channels in the context of PD.
Collapse
Affiliation(s)
- Neha Hanna Daniel
- School of Biology, Indian Institute of Science Education and Research (IISER)-Thiruvananthapuram, Kerala, 695551, India
| | - Ananya Aravind
- School of Biology, Indian Institute of Science Education and Research (IISER)-Thiruvananthapuram, Kerala, 695551, India
| | - Poonam Thakur
- School of Biology, Indian Institute of Science Education and Research (IISER)-Thiruvananthapuram, Kerala, 695551, India.
| |
Collapse
|
20
|
Rahm AK, Gramlich D, Wieder T, Müller ME, Schoeffel A, El Tahry FA, Most P, Heimberger T, Sandke S, Weis T, Ullrich ND, Korff T, Lugenbiel P, Katus HA, Thomas D. Trigger-Specific Remodeling of K Ca2 Potassium Channels in Models of Atrial Fibrillation. Pharmgenomics Pers Med 2021; 14:579-590. [PMID: 34045886 PMCID: PMC8144362 DOI: 10.2147/pgpm.s290291] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2020] [Accepted: 04/06/2021] [Indexed: 11/23/2022] Open
Abstract
AIM Effective antiarrhythmic treatment of atrial fibrillation (AF) constitutes a major challenge, in particular, when concomitant heart failure (HF) is present. HF-associated atrial arrhythmogenesis is distinctly characterized by prolonged atrial refractoriness. Small-conductance, calcium-activated K+ (KCa, SK, KCNN) channels contribute to cardiac action potential repolarization and are implicated in AF susceptibility and therapy. The mechanistic impact of AF/HF-related triggers on atrial KCa channels is not known. We hypothesized that tachycardia, stretch, β-adrenergic stimulation, and hypoxia differentially determine KCa2.1-2.3 channel remodeling in atrial cells. METHODS KCNN1-3 transcript levels were assessed in AF/HF patients and in a pig model of atrial tachypacing-induced AF with reduced left ventricular function. HL-1 atrial myocytes were subjected to proarrhythmic triggers to investigate the effects on Kcnn mRNA and KCa channel protein. RESULTS Atrial KCNN1-3 expression was reduced in AF/HF patients. KCNN2 and KCNN3 suppression was recapitulated in the corresponding pig model. In contrast to human AF, KCNN1 remained unchanged in pigs. Channel- and stressor-specific remodeling was revealed in vitro. Lower expression levels of KCNN1/KCa2.1 were linked to stretch and β-adrenergic stimulation. Furthermore, KCNN3/KCa2.3 expression was suppressed upon tachypacing and hypoxia. Finally, KCNN2/KCa2.2 abundance was specifically enhanced by hypoxia. CONCLUSION Reduction of KCa2.1-2.3 channel expression might contribute to the action potential prolongation in AF complicated by HF. Subtype-specific KCa2 channel remodeling induced by tachypacing, stretch, β-adrenergic stimulation, or hypoxia is expected to differentially determine atrial remodeling, depending on patient-specific activation of each triggering factor. Stressor-dependent KCa2 regulation in atrial myocytes provides a starting point for mechanism-based antiarrhythmic therapy.
Collapse
Affiliation(s)
- Ann-Kathrin Rahm
- Department of Cardiology, Medical University Hospital Heidelberg, Heidelberg, 69120, Germany
- HCR (Heidelberg Center for Heart Rhythm Disorders), University Hospital Heidelberg, Heidelberg, 69120, Germany
- DZHK (German Centre for Cardiovascular Research), Partner Site Heidelberg/Mannheim, University of Heidelberg, Heidelberg, 69120, Germany
| | - Dominik Gramlich
- Department of Cardiology, Medical University Hospital Heidelberg, Heidelberg, 69120, Germany
- HCR (Heidelberg Center for Heart Rhythm Disorders), University Hospital Heidelberg, Heidelberg, 69120, Germany
- DZHK (German Centre for Cardiovascular Research), Partner Site Heidelberg/Mannheim, University of Heidelberg, Heidelberg, 69120, Germany
| | - Teresa Wieder
- Department of Cardiology, Medical University Hospital Heidelberg, Heidelberg, 69120, Germany
- HCR (Heidelberg Center for Heart Rhythm Disorders), University Hospital Heidelberg, Heidelberg, 69120, Germany
| | - Mara Elena Müller
- Department of Cardiology, Medical University Hospital Heidelberg, Heidelberg, 69120, Germany
- HCR (Heidelberg Center for Heart Rhythm Disorders), University Hospital Heidelberg, Heidelberg, 69120, Germany
- DZHK (German Centre for Cardiovascular Research), Partner Site Heidelberg/Mannheim, University of Heidelberg, Heidelberg, 69120, Germany
| | - Axel Schoeffel
- Department of Cardiology, Medical University Hospital Heidelberg, Heidelberg, 69120, Germany
- HCR (Heidelberg Center for Heart Rhythm Disorders), University Hospital Heidelberg, Heidelberg, 69120, Germany
| | - Fadwa A El Tahry
- Department of Cardiology, Medical University Hospital Heidelberg, Heidelberg, 69120, Germany
| | - Patrick Most
- Department of Cardiology, Medical University Hospital Heidelberg, Heidelberg, 69120, Germany
- HCR (Heidelberg Center for Heart Rhythm Disorders), University Hospital Heidelberg, Heidelberg, 69120, Germany
| | - Tanja Heimberger
- Department of Cardiology, Medical University Hospital Heidelberg, Heidelberg, 69120, Germany
- DZHK (German Centre for Cardiovascular Research), Partner Site Heidelberg/Mannheim, University of Heidelberg, Heidelberg, 69120, Germany
| | - Steffi Sandke
- Department of Cardiology, Medical University Hospital Heidelberg, Heidelberg, 69120, Germany
- DZHK (German Centre for Cardiovascular Research), Partner Site Heidelberg/Mannheim, University of Heidelberg, Heidelberg, 69120, Germany
| | - Tanja Weis
- Department of Cardiology, Medical University Hospital Heidelberg, Heidelberg, 69120, Germany
- DZHK (German Centre for Cardiovascular Research), Partner Site Heidelberg/Mannheim, University of Heidelberg, Heidelberg, 69120, Germany
| | - Nina D Ullrich
- Institute of Physiology and Pathophysiology, Department of Cardiovascular Physiology, Heidelberg University, Heidelberg, 69120, Germany
| | - Thomas Korff
- Institute of Physiology and Pathophysiology, Department of Cardiovascular Physiology, Heidelberg University, Heidelberg, 69120, Germany
- European Center for Angioscience (ECAS), Medical Faculty Mannheim, Heidelberg University, Mannheim, 68167, Germany
| | - Patrick Lugenbiel
- Department of Cardiology, Medical University Hospital Heidelberg, Heidelberg, 69120, Germany
- HCR (Heidelberg Center for Heart Rhythm Disorders), University Hospital Heidelberg, Heidelberg, 69120, Germany
- DZHK (German Centre for Cardiovascular Research), Partner Site Heidelberg/Mannheim, University of Heidelberg, Heidelberg, 69120, Germany
| | - Hugo A Katus
- Department of Cardiology, Medical University Hospital Heidelberg, Heidelberg, 69120, Germany
- HCR (Heidelberg Center for Heart Rhythm Disorders), University Hospital Heidelberg, Heidelberg, 69120, Germany
- DZHK (German Centre for Cardiovascular Research), Partner Site Heidelberg/Mannheim, University of Heidelberg, Heidelberg, 69120, Germany
| | - Dierk Thomas
- Department of Cardiology, Medical University Hospital Heidelberg, Heidelberg, 69120, Germany
- HCR (Heidelberg Center for Heart Rhythm Disorders), University Hospital Heidelberg, Heidelberg, 69120, Germany
- DZHK (German Centre for Cardiovascular Research), Partner Site Heidelberg/Mannheim, University of Heidelberg, Heidelberg, 69120, Germany
| |
Collapse
|
21
|
Zhang XD, Thai PN, Lieu DK, Chiamvimonvat N. Cardiac small-conductance calcium-activated potassium channels in health and disease. Pflugers Arch 2021; 473:477-489. [PMID: 33624131 PMCID: PMC7940285 DOI: 10.1007/s00424-021-02535-0] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2020] [Revised: 02/02/2021] [Accepted: 02/03/2021] [Indexed: 12/22/2022]
Abstract
Small-conductance Ca2+-activated K+ (SK, KCa2) channels are encoded by KCNN genes, including KCNN1, 2, and 3. The channels play critical roles in the regulation of cardiac excitability and are gated solely by beat-to-beat changes in intracellular Ca2+. The family of SK channels consists of three members with differential sensitivity to apamin. All three isoforms are expressed in human hearts. Studies over the past two decades have provided evidence to substantiate the pivotal roles of SK channels, not only in healthy heart but also with diseases including atrial fibrillation (AF), ventricular arrhythmia, and heart failure (HF). SK channels are prominently expressed in atrial myocytes and pacemaking cells, compared to ventricular cells. However, the channels are significantly upregulated in ventricular myocytes in HF and pulmonary veins in AF models. Interests in cardiac SK channels are further fueled by recent studies suggesting the possible roles of SK channels in human AF. Therefore, SK channel may represent a novel therapeutic target for atrial arrhythmias. Furthermore, SK channel function is significantly altered by human calmodulin (CaM) mutations, linked to life-threatening arrhythmia syndromes. The current review will summarize recent progress in our understanding of cardiac SK channels and the roles of SK channels in the heart in health and disease.
Collapse
Affiliation(s)
- Xiao-Dong Zhang
- Division of Cardiovascular Medicine, Department of Internal Medicine, School of Medicine, University of California, Davis, One Shields Avenue, GBSF 6315, Davis, CA, 95616, USA.
- Department of Veterans Affairs, Northern California Health Care System, 10535 Hospital Way, Mather, CA, 95655, USA.
| | - Phung N Thai
- Division of Cardiovascular Medicine, Department of Internal Medicine, School of Medicine, University of California, Davis, One Shields Avenue, GBSF 6315, Davis, CA, 95616, USA
- Department of Veterans Affairs, Northern California Health Care System, 10535 Hospital Way, Mather, CA, 95655, USA
| | - Deborah K Lieu
- Division of Cardiovascular Medicine, Department of Internal Medicine, School of Medicine, University of California, Davis, One Shields Avenue, GBSF 6315, Davis, CA, 95616, USA
| | - Nipavan Chiamvimonvat
- Division of Cardiovascular Medicine, Department of Internal Medicine, School of Medicine, University of California, Davis, One Shields Avenue, GBSF 6315, Davis, CA, 95616, USA.
- Department of Veterans Affairs, Northern California Health Care System, 10535 Hospital Way, Mather, CA, 95655, USA.
- Department of Pharmacology, School of Medicine, University of California, Davis, Davis, CA, 95616, USA.
| |
Collapse
|
22
|
Prager EM, Dorman DB, Hobel ZB, Malgady JM, Blackwell KT, Plotkin JL. Dopamine Oppositely Modulates State Transitions in Striosome and Matrix Direct Pathway Striatal Spiny Neurons. Neuron 2020; 108:1091-1102.e5. [PMID: 33080228 PMCID: PMC7769890 DOI: 10.1016/j.neuron.2020.09.028] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2018] [Revised: 07/29/2020] [Accepted: 09/21/2020] [Indexed: 12/18/2022]
Abstract
Corticostriatal synaptic integration is partitioned among striosome (patch) and matrix compartments of the dorsal striatum, allowing compartmentalized control of discrete aspects of behavior. Despite the significance of such organization, it's unclear how compartment-specific striatal output is dynamically achieved, particularly considering new evidence that overlap of afferents is substantial. We show that dopamine oppositely shapes responses to convergent excitatory inputs in mouse striosome and matrix striatal spiny projection neurons (SPNs). Activation of postsynaptic D1 dopamine receptors promoted the generation of long-lasting synaptically evoked "up-states" in matrix SPNs but opposed it in striosomes, which were more excitable under basal conditions. Differences in dopaminergic modulation were mediated, in part, by dendritic voltage-gated calcium channels (VGCCs): pharmacological manipulation of L-type VGCCs reversed compartment-specific responses to D1 receptor activation. These results support a novel mechanism for the selection of striatal circuit components, where fluctuating levels of dopamine shift the balance of compartment-specific striatal output.
Collapse
Affiliation(s)
- Eric M Prager
- Department of Neurobiology and Behavior, Center for Nervous System Disorders, Stony Brook University School of Medicine, Stony Brook, NY 11794, USA
| | - Daniel B Dorman
- Interdisciplinary Program in Neuroscience, George Mason University, Fairfax, VA 22030, USA
| | - Zachary B Hobel
- Department of Neurobiology and Behavior, Center for Nervous System Disorders, Stony Brook University School of Medicine, Stony Brook, NY 11794, USA
| | - Jeffrey M Malgady
- Department of Neurobiology and Behavior, Center for Nervous System Disorders, Stony Brook University School of Medicine, Stony Brook, NY 11794, USA
| | - Kim T Blackwell
- Interdisciplinary Program in Neuroscience, George Mason University, Fairfax, VA 22030, USA; Bioengineering Department, Volgenau School of Engineering, George Mason University, Fairfax, VA 22030, USA
| | - Joshua L Plotkin
- Department of Neurobiology and Behavior, Center for Nervous System Disorders, Stony Brook University School of Medicine, Stony Brook, NY 11794, USA.
| |
Collapse
|
23
|
Gong D, Yan N, Ledford HA. Structural Basis for the Modulation of Ryanodine Receptors. Trends Biochem Sci 2020; 46:489-501. [PMID: 33353849 DOI: 10.1016/j.tibs.2020.11.009] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2020] [Revised: 11/16/2020] [Accepted: 11/20/2020] [Indexed: 12/11/2022]
Abstract
Historically, ryanodine receptors (RyRs) have presented unique challenges for high-resolution structural determination despite long-standing interest in their role in excitation-contraction coupling. Owing to their large size (nearly 2.2 MDa), high-resolution structures remained elusive until the advent of cryogenic electron microscopy (cryo-EM) techniques. In recent years, structures for both RyR1 and RyR2 have been solved at near-atomic resolution. Furthermore, recent reports have delved into their more complex structural associations with key modulators - proteins such as the dihydropyridine receptor (DHPR), FKBP12/12.6, and calmodulin (CaM), as well as ions and small molecules including Ca2+, ATP, caffeine, and PCB95. This review addresses the modulation of RyR1 and RyR2, in addition to the impact of such discoveries on intracellular Ca2+ dynamics and biophysical properties.
Collapse
Affiliation(s)
- Deshun Gong
- Zhejiang Provincial Laboratory of Life Sciences and Biomedicine, Key Laboratory of Structural Biology of Zhejiang Province/Key Laboratory of Growth Regulation and Transformation Research of Zhejiang Province, School of Life Sciences, Westlake University, Hangzhou 310024, Zhejiang, China; Institute of Biology, Westlake Institute for Advanced Study, Hangzhou 310024, Zhejiang Province, China.
| | - Nieng Yan
- Department of Molecular Biology, Princeton University, Princeton, NJ 08544, USA.
| | - Hannah A Ledford
- Department of Molecular Biology, Princeton University, Princeton, NJ 08544, USA.
| |
Collapse
|
24
|
Tiffner A, Derler I. Molecular Choreography and Structure of Ca 2+ Release-Activated Ca 2+ (CRAC) and K Ca2+ Channels and Their Relevance in Disease with Special Focus on Cancer. MEMBRANES 2020; 10:E425. [PMID: 33333945 PMCID: PMC7765462 DOI: 10.3390/membranes10120425] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/04/2020] [Revised: 12/04/2020] [Accepted: 12/07/2020] [Indexed: 12/16/2022]
Abstract
Ca2+ ions play a variety of roles in the human body as well as within a single cell. Cellular Ca2+ signal transduction processes are governed by Ca2+ sensing and Ca2+ transporting proteins. In this review, we discuss the Ca2+ and the Ca2+-sensing ion channels with particular focus on the structure-function relationship of the Ca2+ release-activated Ca2+ (CRAC) ion channel, the Ca2+-activated K+ (KCa2+) ion channels, and their modulation via other cellular components. Moreover, we highlight their roles in healthy signaling processes as well as in disease with a special focus on cancer. As KCa2+ channels are activated via elevations of intracellular Ca2+ levels, we summarize the current knowledge on the action mechanisms of the interplay of CRAC and KCa2+ ion channels and their role in cancer cell development.
Collapse
Affiliation(s)
| | - Isabella Derler
- Institute of Biophysics, JKU Life Science Center, Johannes Kepler University Linz, A-4020 Linz, Austria;
| |
Collapse
|
25
|
Weisbrod D. Small and Intermediate Calcium Activated Potassium Channels in the Heart: Role and Strategies in the Treatment of Cardiovascular Diseases. Front Physiol 2020; 11:590534. [PMID: 33329039 PMCID: PMC7719780 DOI: 10.3389/fphys.2020.590534] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2020] [Accepted: 10/02/2020] [Indexed: 12/11/2022] Open
Abstract
Calcium-activated potassium channels are a heterogeneous family of channels that, despite their different biophysical characteristics, structures, and pharmacological signatures, play a role of transducer between the ubiquitous intracellular calcium signaling and the electric variations of the membrane. Although this family of channels was extensively described in various excitable and non-excitable tissues, an increasing amount of evidences shows their functional role in the heart. This review aims to focus on the physiological role and the contribution of the small and intermediate calcium-activated potassium channels in cardiac pathologies.
Collapse
|
26
|
Predisposition of Neonatal Maternal Separation to Visceral Hypersensitivity via Downregulation of Small-Conductance Calcium-Activated Potassium Channel Subtype 2 (SK2) in Mice. Neural Plast 2020; 2020:8876230. [PMID: 33029124 PMCID: PMC7528131 DOI: 10.1155/2020/8876230] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2020] [Revised: 08/31/2020] [Accepted: 09/06/2020] [Indexed: 12/13/2022] Open
Abstract
Background Visceral hypersensitivity is a common occurrence of gastrointestinal diseases such as irritable bowel syndrome (IBS), wherein early-life stress (ELS) may have a high predisposition to the development of visceral hypersensitivity in adulthood, with the specific underlying mechanism still elusive. Herein, we assessed the potential effect of small-conductance calcium-activated potassium channel subtype 2 (SK2) in the spinal dorsal horn (DH) on the pathogenesis of visceral hypersensitivity induced by maternal separation (MS) in mice. Methods Neonatal mice were subjected to the MS paradigm, an established ELS model. In adulthood, the visceral pain threshold and the abdominal withdrawal reflex (AWR) were measured with an inflatable balloon. The elevated plus maze, open field test, sucrose preference test, and forced swim test were employed to evaluate the anxiety- and depression-like behaviors. The expression levels of SK2 in the spinal DH were determined by immunofluorescence and western blotting. The mRNA of SK2 and membrane palmitoylated protein 2 (MPP2) were determined by quantitative real-time polymerase chain reaction (qRT-PCR). Electrophysiology was applied to evaluate the neuronal firing rates and SK2 channel-mediated afterhyperpolarization current (I AHP). The interaction between MPP2 and SK2 was validated by coimmunoprecipitation. Results In contrast to the naïve mice, ethological findings in MS mice revealed lowered visceral pain threshold, more evident anxiety- and depression-like behaviors, and downregulated expression of membrane SK2 protein and MPP2 protein. Moreover, electrophysiological results indicated increased neuronal firing rates and decreased I AHP in the spinal DH neurons. Nonetheless, intrathecal injection of the SK2 channel activator 1-ethyl-2-benzimidazolinone (1-EBIO) in MS mice could reverse the electrophysiological alterations and elevate the visceral pain threshold. In the naïve mice, administration of the SK2 channel blocker apamin abated I AHP and elevated spontaneous neuronal firing rates in the spinal DH neurons, reducing the visceral pain threshold. Finally, disruption of the MPP2 expression by small interfering RNA (siRNA) could amplify visceral hypersensitivity in naïve mice. Conclusions ELS-induced visceral pain and visceral hypersensitivity are associated with the underfunction of SK2 channels in the spinal DH.
Collapse
|
27
|
Evans RC, Twedell EL, Zhu M, Ascencio J, Zhang R, Khaliq ZM. Functional Dissection of Basal Ganglia Inhibitory Inputs onto Substantia Nigra Dopaminergic Neurons. Cell Rep 2020; 32:108156. [PMID: 32937133 PMCID: PMC9887718 DOI: 10.1016/j.celrep.2020.108156] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2019] [Revised: 07/11/2020] [Accepted: 08/25/2020] [Indexed: 02/02/2023] Open
Abstract
Substantia nigra (SNc) dopaminergic neurons respond to aversive stimuli with inhibitory pauses in firing followed by transient rebound activation. We tested integration of inhibitory synaptic inputs onto SNc neurons from genetically defined populations in dorsal striatum (striosome and matrix) and external globus pallidus (GPe; parvalbumin- and Lhx6-positive), and examined their contribution to pause-rebound firing. Activation of striosome projections, which target "dendron bouquets" in the pars reticulata (SNr), consistently quiets firing and relief from striosome inhibition triggers rebound activity. Striosomal inhibitory postsynaptic currents (IPSCs) display a prominent GABA-B receptor-mediated component that strengthens the impact of SNr dendrite synapses on somatic excitability and enables rebounding. By contrast, GPe projections activate GABA-A receptors on the soma and proximal dendrites but do not result in rebounding. Lastly, optical mapping shows that dorsal striatum selectively inhibits the ventral population of SNc neurons, which are intrinsically capable of rebounding. Therefore, we define a distinct striatonigral circuit for generating dopamine rebound.
Collapse
Affiliation(s)
- Rebekah C. Evans
- Cellular Neurophysiology Section, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD 20892, USA
| | - Emily L. Twedell
- Cellular Neurophysiology Section, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD 20892, USA
| | - Manhua Zhu
- Cellular Neurophysiology Section, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD 20892, USA
| | - Jefferson Ascencio
- Cellular Neurophysiology Section, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD 20892, USA
| | - Renshu Zhang
- Cellular Neurophysiology Section, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD 20892, USA
| | - Zayd M. Khaliq
- Cellular Neurophysiology Section, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD 20892, USA,Lead Contact,Correspondence:
| |
Collapse
|
28
|
Shrestha A, Sultana R, Adeniyi PA, Lee CC, Ogundele OM. Positive Modulation of SK Channel Impedes Neuron-Specific Cytoskeletal Organization and Maturation. Dev Neurosci 2020; 42:59-71. [PMID: 32580196 PMCID: PMC7486235 DOI: 10.1159/000507989] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2019] [Accepted: 04/15/2020] [Indexed: 01/01/2023] Open
Abstract
N-methyl-D-aspartate receptor (NMDAR) modulates the structural plasticity of dendritic spines by impacting cytoskeletal organization and kinase signaling. In the developing nervous system, activation of NMDAR is pertinent for neuronal migration, neurite differentiation, and cellular organization. Given that small conductance potassium channels (SK2/3) repress NMDAR ionotropic signaling, this study highlights the impact of neonatal SK channel potentiation on adult cortical and hippocampal organization. Neonatal SK channel potentiation was performed by one injection of SK2/3 agonist (CyPPA) into the pallium of mice on postnatal day 2 (P2). When the animals reached adulthood (P55), the hippocampus and cortex were examined to assess neuronal maturation, lamination, and the distribution of synaptic cytoskeletal proteins. Immunodetection of neuronal markers in the brain of P2-treated P55 mice revealed the presence of immature neurons in the upper cortical layers (layers II-IV) and CA1 (hippocampus). Also, layer-dependent cortical-cell density was attenuated due to the ectopic localization of mature (NeuN+) and immature (Doublecortin+ [DCX+]) neurons in cortical layers II-IV. Similarly, the decreased count of NeuN+ neurons in the CA1 is accompanied by an increase in the number of immature DCX+ neurons. Ectopic localization of neurons in the upper cortex and CA1 caused the dramatic expression of neuron-specific cytoskeletal proteins. In line with this, structural deformity of neuronal projections and the loss of postsynaptic densities suggests that postsynaptic integrity is compromised in the SK2/3+ brain. From these results, we deduced that SK channel activity in the developing brain likely impacts neuronal maturation through its effects on cytoskeletal formation.
Collapse
Affiliation(s)
- Amita Shrestha
- Department of Comparative Biomedical Sciences, Louisiana State University School of Veterinary Medicine, Baton Rouge, Louisiana, USA
| | - Razia Sultana
- Department of Comparative Biomedical Sciences, Louisiana State University School of Veterinary Medicine, Baton Rouge, Louisiana, USA
| | - Philip A Adeniyi
- Department of Comparative Biomedical Sciences, Louisiana State University School of Veterinary Medicine, Baton Rouge, Louisiana, USA
| | - Charles C Lee
- Department of Comparative Biomedical Sciences, Louisiana State University School of Veterinary Medicine, Baton Rouge, Louisiana, USA
| | - Olalekan M Ogundele
- Department of Comparative Biomedical Sciences, Louisiana State University School of Veterinary Medicine, Baton Rouge, Louisiana, USA,
| |
Collapse
|
29
|
Trombetta-Lima M, Krabbendam IE, Dolga AM. Calcium-activated potassium channels: implications for aging and age-related neurodegeneration. Int J Biochem Cell Biol 2020; 123:105748. [PMID: 32353429 DOI: 10.1016/j.biocel.2020.105748] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2020] [Revised: 04/11/2020] [Accepted: 04/14/2020] [Indexed: 12/16/2022]
Abstract
Population aging, as well as the handling of age-associated diseases, is a worldwide increasing concern. Among them, Alzheimer's disease stands out as the major cause of dementia culminating in full dependence on other people for basic functions. However, despite numerous efforts, in the last decades, there was no new approved therapeutic drug for the treatment of the disease. Calcium-activated potassium channels have emerged as a potential tool for neuronal protection by modulating intracellular calcium signaling. Their subcellular localization is determinant of their functional effects. When located on the plasma membrane of neuronal cells, they can modulate synaptic function, while their activation at the inner mitochondrial membrane has a neuroprotective potential via the attenuation of mitochondrial reactive oxygen species in conditions of oxidative stress. Here we review the dual role of these channels in the aging phenotype and Alzheimer's disease pathology and discuss their potential use as a therapeutic tool.
Collapse
Affiliation(s)
- Marina Trombetta-Lima
- Faculty of Science and Engineering, Department of Molecular Pharmacology, Groningen Research Institute of Pharmacy (GRIP), University of Groningen, 9713 AV Groningen, the Netherlands; Medical School, Neurology Department, University of São Paulo (USP), 01246903 São Paulo, Brazil
| | - Inge E Krabbendam
- Faculty of Science and Engineering, Department of Molecular Pharmacology, Groningen Research Institute of Pharmacy (GRIP), University of Groningen, 9713 AV Groningen, the Netherlands
| | - Amalia M Dolga
- Faculty of Science and Engineering, Department of Molecular Pharmacology, Groningen Research Institute of Pharmacy (GRIP), University of Groningen, 9713 AV Groningen, the Netherlands.
| |
Collapse
|
30
|
Shrestha A, Sultana R, Lee CC, Ogundele OM. SK Channel Modulates Synaptic Plasticity by Tuning CaMKIIα/β Dynamics. Front Synaptic Neurosci 2019; 11:18. [PMID: 31736736 PMCID: PMC6834780 DOI: 10.3389/fnsyn.2019.00018] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2019] [Accepted: 05/20/2019] [Indexed: 11/13/2022] Open
Abstract
N-Methyl-D-Aspartate Receptor 1 (NMDAR)-linked Ca++ current represents a significant percentage of post-synaptic transient that modulates synaptic strength and is pertinent to dendritic spine plasticity. In the hippocampus, Ca++ transient produced by glutamatergic ionotropic neurotransmission facilitates Ca++-Calmodulin-dependent kinase 2 (CaMKII) Thr286 phosphorylation and promote long-term potentiation (LTP) expression. At CA1 post-synaptic densities, Ca++ transients equally activate small conductance (SK2) channel which regulates excitability by suppressing Ca++ movement. Here, we demonstrate that upstream attenuation of GluN1 function in the hippocampus led to a decrease in Thr286 CaMKIIα phosphorylation, and increased SK2 expression. Consistent with the loss of GluN1 function, potentiation of SK channel in wild type hippocampus reduced CaMKIIα expression and abrogate synaptic localization of T286 pCaMKIIα. Our results demonstrate that positive modulation of SK channel at hippocampal synapses likely refine GluN1-linked plasticity by tuning dendritic localization of CaMKIIα.
Collapse
Affiliation(s)
| | | | | | - Olalekan M. Ogundele
- Department of Comparative Biomedical Sciences, Louisiana State University School of Veterinary Medicine, Baton Rouge, LA, United States
| |
Collapse
|
31
|
Archer CR, Enslow BT, Taylor AB, De la Rosa V, Bhattacharya A, Shapiro MS. A mutually induced conformational fit underlies Ca 2+-directed interactions between calmodulin and the proximal C terminus of KCNQ4 K + channels. J Biol Chem 2019; 294:6094-6112. [PMID: 30808708 PMCID: PMC6463706 DOI: 10.1074/jbc.ra118.006857] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2018] [Revised: 02/24/2019] [Indexed: 11/06/2022] Open
Abstract
Calmodulin (CaM) conveys intracellular Ca2+ signals to KCNQ (Kv7, "M-type") K+ channels and many other ion channels. Whether this "calmodulation" involves a dramatic structural rearrangement or only slight perturbations of the CaM/KCNQ complex is as yet unclear. A consensus structural model of conformational shifts occurring between low nanomolar and physiologically high intracellular [Ca2+] is still under debate. Here, we used various techniques of biophysical chemical analyses to investigate the interactions between CaM and synthetic peptides corresponding to the A and B domains of the KCNQ4 subtype. We found that in the absence of CaM, the peptides are disordered, whereas Ca2+/CaM imposed helical structure on both KCNQ A and B domains. Isothermal titration calorimetry revealed that Ca2+/CaM has higher affinity for the B domain than for the A domain of KCNQ2-4 and much higher affinity for the B domain when prebound with the A domain. X-ray crystallography confirmed that these discrete peptides spontaneously form a complex with Ca2+/CaM, similar to previous reports of CaM binding KCNQ-AB domains that are linked together. Microscale thermophoresis and heteronuclear single-quantum coherence NMR spectroscopy indicated the C-lobe of Ca2+-free CaM to interact with the KCNQ4 B domain (Kd ∼10-20 μm), with increasing Ca2+ molar ratios shifting the CaM-B domain interactions via only the CaM C-lobe to also include the N-lobe. Our findings suggest that in response to increased Ca2+, CaM undergoes lobe switching that imposes a dramatic mutually induced conformational fit to both the proximal C terminus of KCNQ4 channels and CaM, likely underlying Ca2+-dependent regulation of KCNQ gating.
Collapse
Affiliation(s)
- Crystal R Archer
- From the Departments of Cell and Integrative Physiology, University of Texas Health San Antonio, San Antonio, Texas 78229; Departments of Biochemistry and Structural Biology, University of Texas Health San Antonio, San Antonio, Texas 78229
| | - Benjamin T Enslow
- the Long School of Medicine, University of Texas Health San Antonio, San Antonio, Texas 78229
| | - Alexander B Taylor
- Departments of Biochemistry and Structural Biology, University of Texas Health San Antonio, San Antonio, Texas 78229
| | - Victor De la Rosa
- From the Departments of Cell and Integrative Physiology, University of Texas Health San Antonio, San Antonio, Texas 78229
| | - Akash Bhattacharya
- Departments of Biochemistry and Structural Biology, University of Texas Health San Antonio, San Antonio, Texas 78229
| | - Mark S Shapiro
- From the Departments of Cell and Integrative Physiology, University of Texas Health San Antonio, San Antonio, Texas 78229.
| |
Collapse
|
32
|
Dickerson MT, Dadi PK, Altman MK, Verlage KR, Thorson AS, Jordan KL, Vierra NC, Amarnath G, Jacobson DA. Glucose-mediated inhibition of calcium-activated potassium channels limits α-cell calcium influx and glucagon secretion. Am J Physiol Endocrinol Metab 2019; 316:E646-E659. [PMID: 30694690 PMCID: PMC6482666 DOI: 10.1152/ajpendo.00342.2018] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Pancreatic α-cells exhibit oscillations in cytosolic Ca2+ (Ca2+c), which control pulsatile glucagon (GCG) secretion. However, the mechanisms that modulate α-cell Ca2+c oscillations have not been elucidated. As β-cell Ca2+c oscillations are regulated in part by Ca2+-activated K+ (Kslow) currents, this work investigated the role of Kslow in α-cell Ca2+ handling and GCG secretion. α-Cells displayed Kslow currents that were dependent on Ca2+ influx through L- and P/Q-type voltage-dependent Ca2+ channels (VDCCs) as well as Ca2+ released from endoplasmic reticulum stores. α-Cell Kslow was decreased by small-conductance Ca2+-activated K+ (SK) channel inhibitors apamin and UCL 1684, large-conductance Ca2+-activated K+ (BK) channel inhibitor iberiotoxin (IbTx), and intermediate-conductance Ca2+-activated K+ (IK) channel inhibitor TRAM 34. Moreover, partial inhibition of α-cell Kslow with apamin depolarized membrane potential ( Vm) (3.8 ± 0.7 mV) and reduced action potential (AP) amplitude (10.4 ± 1.9 mV). Although apamin transiently increased Ca2+ influx into α-cells at low glucose (42.9 ± 10.6%), sustained SK (38.5 ± 10.4%) or BK channel inhibition (31.0 ± 11.7%) decreased α-cell Ca2+ influx. Total α-cell Ca2+c was similarly reduced (28.3 ± 11.1%) following prolonged treatment with high glucose, but it was not decreased further by SK or BK channel inhibition. Consistent with reduced α-cell Ca2+c following prolonged Kslow inhibition, apamin decreased GCG secretion from mouse (20.4 ± 4.2%) and human (27.7 ± 13.1%) islets at low glucose. These data demonstrate that Kslow activation provides a hyperpolarizing influence on α-cell Vm that sustains Ca2+ entry during hypoglycemic conditions, presumably by preventing voltage-dependent inactivation of P/Q-type VDCCs. Thus, when α-cell Ca2+c is elevated during secretagogue stimulation, Kslow activation helps to preserve GCG secretion.
Collapse
Affiliation(s)
- Matthew T Dickerson
- Department of Molecular Physiology and Biophysics, Vanderbilt University , Nashville, Tennessee
| | - Prasanna K Dadi
- Department of Molecular Physiology and Biophysics, Vanderbilt University , Nashville, Tennessee
| | - Molly K Altman
- Department of Molecular Physiology and Biophysics, Vanderbilt University , Nashville, Tennessee
| | - Kenneth R Verlage
- Department of Molecular Physiology and Biophysics, Vanderbilt University , Nashville, Tennessee
- School of Medicine, Texas Tech University Health Sciences Center , Lubbock, Texas
- Department of Urology, Oregon Health and Science University , Portland, Oregon
| | - Ariel S Thorson
- Department of Molecular Physiology and Biophysics, Vanderbilt University , Nashville, Tennessee
| | - Kelli L Jordan
- Department of Molecular Physiology and Biophysics, Vanderbilt University , Nashville, Tennessee
| | - Nicholas C Vierra
- Department of Molecular Physiology and Biophysics, Vanderbilt University , Nashville, Tennessee
- Department of Neurobiology, Physiology and Behavior University of California , Davis, California
| | - Gautami Amarnath
- Department of Molecular Physiology and Biophysics, Vanderbilt University , Nashville, Tennessee
- Experimental and Clinical Neurosciences, University of Regensburg , Regensburg , Germany
| | - David A Jacobson
- Department of Molecular Physiology and Biophysics, Vanderbilt University , Nashville, Tennessee
| |
Collapse
|
33
|
Hamilton S, Polina I, Terentyeva R, Bronk P, Kim TY, Roder K, Clements RT, Koren G, Choi BR, Terentyev D. PKA phosphorylation underlies functional recruitment of sarcolemmal SK2 channels in ventricular myocytes from hypertrophic hearts. J Physiol 2019; 598:2847-2873. [PMID: 30771223 PMCID: PMC7496687 DOI: 10.1113/jp277618] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2018] [Accepted: 02/08/2019] [Indexed: 12/11/2022] Open
Abstract
KEY POINTS Small-conductance Ca2+ -activated K+ (SK) channels expressed in ventricular myocytes are dormant in health, yet become functional in cardiac disease. SK channels are voltage independent and their gating is controlled by intracellular [Ca2+ ] in a biphasic manner. Submicromolar [Ca2+ ] activates the channel via constitutively-bound calmodulin, whereas higher [Ca2+ ] exerts inhibitory effect during depolarization. Using a rat model of cardiac hypertrophy induced by thoracic aortic banding, we found that functional upregulation of SK2 channels in hypertrophic rat ventricular cardiomyocytes is driven by protein kinase A (PKA) phosphorylation. Using site-directed mutagenesis, we identified serine-465 as the site conferring PKA-dependent effects on SK2 channel function. PKA phosphorylation attenuates ISK rectification by reducing the Ca2+ /voltage-dependent inhibition of SK channels without changing their sensitivity to activating submicromolar [Ca2+ ]i . This mechanism underlies the functional recruitment of SK channels not only in cardiac disease, but also in normal physiology, contributing to repolarization under conditions of enhanced adrenergic drive. ABSTRACT Small-conductance Ca2+ -activated K+ (SK) channels expressed in ventricular myocytes (VMs) are dormant in health, yet become functional in cardiac disease. We aimed to test the hypothesis that post-translational modification of SK channels under conditions accompanied by enhanced adrenergic drive plays a central role in disease-related activation of the channels. We investigated this phenomenon using a rat model of hypertrophy induced by thoracic aortic banding (TAB). Western blot analysis using anti-pan-serine/threonine antibodies demonstrated enhanced phosphorylation of immunoprecipitated SK2 channels in VMs from TAB rats vs. Shams, which was reversible by incubation of the VMs with PKA inhibitor H89 (1 μmol L-1 ). Patch clamped VMs under basal conditions from TABs but not Shams exhibited outward current sensitive to the specific SK inhibitor apamin (100 nmol L-1 ), which was eliminated by inhibition of PKA (1 μmol L-1 ). Beta-adrenergic stimulation (isoproterenol, 100 nmol L-1 ) evoked ISK in VMs from Shams, resulting in shortening of action potentials in VMs and ex vivo optically mapped Sham hearts. Using adenoviral gene transfer, wild-type and mutant SK2 channels were overexpressed in adult rat VMs, revealing serine-465 as the site that elicits PKA-dependent phosphorylation effects on SK2 channel function. Concurrent confocal Ca2+ imaging experiments established that PKA phosphorylation lessens rectification of ISK via reduction Ca2+ /voltage-dependent inhibition of the channels at high [Ca2+ ] without affecting their sensitivity to activation by Ca2+ in the submicromolar range. In conclusion, upregulation of SK channels in diseased VMs is mediated by hyperadrenergic drive in cardiac hypertrophy, with functional effects on the channel conferred by PKA-dependent phosphorylation at serine-465.
Collapse
Affiliation(s)
- Shanna Hamilton
- Department of Medicine, The Warren Alpert Medical School of Brown University, Rhode Island Hospital, Cardiovascular Research Center, Providence, RI, USA.,Dorothy M. Davis Heart and Lung Research Institute, College of Medicine, The Ohio State University, Columbus, OH, USA.,Department of Physiology and Cell Biology, College of Medicine, The Ohio State University, Columbus, OH, USA
| | - Iuliia Polina
- Department of Medicine, The Warren Alpert Medical School of Brown University, Rhode Island Hospital, Cardiovascular Research Center, Providence, RI, USA.,Medical University of South Carolina, Department of Medicine, Division of Nephrology, Charleston, SC, USA
| | - Radmila Terentyeva
- Department of Medicine, The Warren Alpert Medical School of Brown University, Rhode Island Hospital, Cardiovascular Research Center, Providence, RI, USA.,Dorothy M. Davis Heart and Lung Research Institute, College of Medicine, The Ohio State University, Columbus, OH, USA.,Department of Physiology and Cell Biology, College of Medicine, The Ohio State University, Columbus, OH, USA
| | - Peter Bronk
- Department of Medicine, The Warren Alpert Medical School of Brown University, Rhode Island Hospital, Cardiovascular Research Center, Providence, RI, USA
| | - Tae Yun Kim
- Department of Medicine, The Warren Alpert Medical School of Brown University, Rhode Island Hospital, Cardiovascular Research Center, Providence, RI, USA
| | - Karim Roder
- Department of Medicine, The Warren Alpert Medical School of Brown University, Rhode Island Hospital, Cardiovascular Research Center, Providence, RI, USA
| | - Richard T Clements
- Department of Surgery, The Warren Alpert Medical School of Brown University, Rhode Island Hospital, Cardiovascular Research Center, Providence, RI, USA.,Vascular Research Laboratory, Providence Veterans Affairs Medical Center, Providence, RI, USA
| | - Gideon Koren
- Department of Medicine, The Warren Alpert Medical School of Brown University, Rhode Island Hospital, Cardiovascular Research Center, Providence, RI, USA
| | - Bum-Rak Choi
- Department of Medicine, The Warren Alpert Medical School of Brown University, Rhode Island Hospital, Cardiovascular Research Center, Providence, RI, USA
| | - Dmitry Terentyev
- Department of Medicine, The Warren Alpert Medical School of Brown University, Rhode Island Hospital, Cardiovascular Research Center, Providence, RI, USA.,Dorothy M. Davis Heart and Lung Research Institute, College of Medicine, The Ohio State University, Columbus, OH, USA.,Department of Physiology and Cell Biology, College of Medicine, The Ohio State University, Columbus, OH, USA
| |
Collapse
|
34
|
Cantonero C, Sanchez-Collado J, Gonzalez-Nuñez MA, Salido GM, Lopez JJ, Jardin I, Rosado JA. Store-independent Orai1-mediated Ca 2+ entry and cancer. Cell Calcium 2019; 80:1-7. [PMID: 30921687 DOI: 10.1016/j.ceca.2019.02.012] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2019] [Revised: 02/28/2019] [Accepted: 02/28/2019] [Indexed: 12/22/2022]
Abstract
Ca2+ channels play an important role in the development of different types of cancer, and considerable progress has been made to understand the pathophysiological mechanisms underlying the role of Ca2+ influx in the development of different cancer hallmarks. Orai1 is among the most ubiquitous and multifunctional Ca2+ channels. Orai1 mediates the highly Ca2+-selective Ca2+ release-activated current (ICRAC) and participates in the less Ca2+-selective store-operated current (ISOC), along with STIM1 or STIM1 and TRPC1, respectively. Furthermore, Orai1 contributes to a variety of store-independent Ca2+ influx mechanisms, including the arachidonate-regulated Ca2+ current, together with Orai3 and the plasma membrane resident pool of STIM1, as well as the constitutive Ca2+ influx processes activated by the secretory pathway Ca2+-ATPase-2 (SPCA2) or supported by physical and functional interaction with the small conductance Ca2+-activated K+ channel 3 (SK3) or the voltage-dependent Kv10.1 channel. This review summarizes the current knowledge concerning the store-independent mechanisms of Ca2+ influx activation through Orai1 channels and their role in the development of different cancer features.
Collapse
Affiliation(s)
- C Cantonero
- Cellular Physiology Research Group, Department of Physiology, Institute of Molecular Pathology Biomarkers, University of Extremadura, 10003 Caceres, Spain
| | - J Sanchez-Collado
- Cellular Physiology Research Group, Department of Physiology, Institute of Molecular Pathology Biomarkers, University of Extremadura, 10003 Caceres, Spain
| | - M A Gonzalez-Nuñez
- Pathology Service, Hospital San Pedro de Alcantara, 10003 Cáceres, Spain
| | - G M Salido
- Cellular Physiology Research Group, Department of Physiology, Institute of Molecular Pathology Biomarkers, University of Extremadura, 10003 Caceres, Spain
| | - J J Lopez
- Cellular Physiology Research Group, Department of Physiology, Institute of Molecular Pathology Biomarkers, University of Extremadura, 10003 Caceres, Spain
| | - I Jardin
- Cellular Physiology Research Group, Department of Physiology, Institute of Molecular Pathology Biomarkers, University of Extremadura, 10003 Caceres, Spain
| | - J A Rosado
- Cellular Physiology Research Group, Department of Physiology, Institute of Molecular Pathology Biomarkers, University of Extremadura, 10003 Caceres, Spain.
| |
Collapse
|
35
|
Dorman DB, Jędrzejewska-Szmek J, Blackwell KT. Inhibition enhances spatially-specific calcium encoding of synaptic input patterns in a biologically constrained model. eLife 2018; 7:e38588. [PMID: 30355449 PMCID: PMC6235562 DOI: 10.7554/elife.38588] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2018] [Accepted: 10/24/2018] [Indexed: 11/13/2022] Open
Abstract
Synaptic plasticity, which underlies learning and memory, depends on calcium elevation in neurons, but the precise relationship between calcium and spatiotemporal patterns of synaptic inputs is unclear. Here, we develop a biologically realistic computational model of striatal spiny projection neurons with sophisticated calcium dynamics, based on data from rodents of both sexes, to investigate how spatiotemporally clustered and distributed excitatory and inhibitory inputs affect spine calcium. We demonstrate that coordinated excitatory synaptic inputs evoke enhanced calcium elevation specific to stimulated spines, with lower but physiologically relevant calcium elevation in nearby non-stimulated spines. Results further show a novel and important function of inhibition-to enhance the difference in calcium between stimulated and non-stimulated spines. These findings suggest that spine calcium dynamics encode synaptic input patterns and may serve as a signal for both stimulus-specific potentiation and heterosynaptic depression, maintaining balanced activity in a dendritic branch while inducing pattern-specific plasticity.
Collapse
Affiliation(s)
- Daniel B Dorman
- Interdisciplinary Program in NeuroscienceGeorge Mason UniversityFairfaxUnited States
| | | | - Kim T Blackwell
- Interdisciplinary Program in Neuroscience, Bioengineering DepartmentKrasnow Institute for Advanced Study, George Mason UniversityFairfaxUnited States
| |
Collapse
|
36
|
Computational modeling of the effect of temperature variations on human pancreatic β-cell activity. J Therm Biol 2018; 75:69-80. [PMID: 30017054 DOI: 10.1016/j.jtherbio.2018.05.006] [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] [Received: 01/30/2018] [Revised: 05/23/2018] [Accepted: 05/24/2018] [Indexed: 11/22/2022]
Abstract
The effect of temperature variations on the pancreatic β-cell activity and the role of different model compartments in temperature sensing have been investigated using a computational modeling approach. The results of our study show that temperature variations by several degrees can change the dynamical states of the β-cell system. In addition, temperature variations can alter the characteristic features of the membrane voltage, which correlates with insulin secretion. Simulation results show that the ion channels such as the L-type calcium, the hERG potassium, sodium channels and the glycolysis pathway are the possible sites for sensing temperature variation. Results indicate that for a small temperature change, even though the frequency and amplitude of electrical activity are altered, the area under the membrane potential curve remains almost unchanged, which implies the existence of a thermoregulatory mechanism for preserving the amount of insulin secretion. Furthermore, the computational analysis shows that the β-cell electrical activity exhibits a bursting pattern in physiological temperature (37 °C) while in vitro studies reported almost the spiking activity at lower temperatures. Since hormone-secreting systems work more efficient in bursting mode, we propose that the pancreatic β-cell works better in the physiological temperature compared with the reference temperature (33 °C).
Collapse
|
37
|
Cheung AC, Lorenzo Pisarello MJ, LaRusso NF. Pathobiology of biliary epithelia. Biochim Biophys Acta Mol Basis Dis 2018; 1864:1220-1231. [PMID: 28716705 PMCID: PMC5777905 DOI: 10.1016/j.bbadis.2017.06.024] [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: 05/11/2017] [Revised: 06/22/2017] [Accepted: 06/26/2017] [Indexed: 12/12/2022]
Abstract
Cholangiocytes are epithelial cells that line the intra- and extrahepatic biliary tree. They serve predominantly to mediate the content of luminal biliary fluid, which is controlled via numerous signaling pathways influenced by endogenous (e.g., bile acids, nucleotides, hormones, neurotransmitters) and exogenous (e.g., microbes/microbial products, drugs etc.) molecules. When injured, cholangiocytes undergo apoptosis/lysis, repair and proliferation. They also become senescent, a form of cell cycle arrest, which may prevent propagation of injury and/or malignant transformation. Senescent cholangiocytes can undergo further transformation to a senescence-associated secretory phenotype (SASP), where they begin secreting pro-inflammatory and pro-fibrotic signals that may contribute to disease initiation and progression. These and other concepts related to cholangiocyte pathobiology will be reviewed herein. This article is part of a Special Issue entitled: Cholangiocytes in Health and Disease edited by Jesus Banales, Marco Marzioni, Nicholas LaRusso and Peter Jansen.
Collapse
Affiliation(s)
- Angela C Cheung
- Division of Gastroenterology and Hepatology, Mayo Clinic Center for Cell Signaling in Gastroenterology, Mayo Clinic, Rochester, MN, United States
| | - Maria J Lorenzo Pisarello
- Division of Gastroenterology and Hepatology, Mayo Clinic Center for Cell Signaling in Gastroenterology, Mayo Clinic, Rochester, MN, United States
| | - Nicholas F LaRusso
- Division of Gastroenterology and Hepatology, Mayo Clinic Center for Cell Signaling in Gastroenterology, Mayo Clinic, Rochester, MN, United States.
| |
Collapse
|
38
|
Sforna L, Megaro A, Pessia M, Franciolini F, Catacuzzeno L. Structure, Gating and Basic Functions of the Ca2+-activated K Channel of Intermediate Conductance. Curr Neuropharmacol 2018; 16:608-617. [PMID: 28875832 PMCID: PMC5997868 DOI: 10.2174/1570159x15666170830122402] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2017] [Revised: 07/21/2017] [Accepted: 07/22/2017] [Indexed: 01/04/2023] Open
Abstract
BACKGROUND The KCa3.1 channel is the intermediate-conductance member of the Ca2+- activated K channel superfamily. It is widely expressed in excitable and non-excitable cells, where it plays a major role in a number of cell functions. This paper aims at illustrating the main structural, biophysical and modulatory properties of the KCa3.1 channel, and providing an account of experimental data on its role in volume regulation and Ca2+ signals. METHODS Research and online content related to the structure, structure/function relationship, and physiological role of the KCa3.1 channel are reviewed. RESULTS Expressed in excitable and non-excitable cells, the KCa3.1 channel is voltage independent, its opening being exclusively gated by the binding of intracellular Ca2+ to calmodulin, a Ca2+- binding protein constitutively associated with the C-terminus of each KCa3.1 channel α subunit. The KCa3.1 channel activates upon high affinity Ca2+ binding, and in highly coordinated fashion giving steep Hill functions and relatively low EC50 values (100-350 nM). This high Ca2+ sensitivity is physiologically modulated by closely associated kinases and phosphatases. The KCa3.1 channel is normally activated by global Ca2+ signals as resulting from Ca2+ released from intracellular stores, or by the refilling influx through store operated Ca2+ channels, but cases of strict functional coupling with Ca2+-selective channels are also found. KCa3.1 channels are highly expressed in many types of cells, where they play major roles in cell migration and death. The control of these complex cellular processes is achieved by KCa3.1 channel regulation of the driving force for Ca2+ entry from the extracellular medium, and by mediating the K+ efflux required for cell volume control. CONCLUSION Much work remains to be done to fully understand the structure/function relationship of the KCa3.1 channels. Hopefully, this effort will provide the basis for a beneficial modulation of channel activity under pathological conditions.
Collapse
Affiliation(s)
| | | | | | - Fabio Franciolini
- Address correspondence to these authors at the Department of Chemistry, Biology and Biotechnology, University of Perugia, Via Pascoli, 8-06123, Perugia; Tel: 39.075.585.5751; E-mails: and
| | - Luigi Catacuzzeno
- Address correspondence to these authors at the Department of Chemistry, Biology and Biotechnology, University of Perugia, Via Pascoli, 8-06123, Perugia; Tel: 39.075.585.5751; E-mails: and
| |
Collapse
|
39
|
Sultana R, Ghandi T, M. Davila A, Lee CC, Ogundele OM. Upregulated SK2 Expression and Impaired CaMKII Phosphorylation Are Shared Synaptic Defects Between 16p11.2del and 129S: Δdisc1 Mutant Mice. ASN Neuro 2018; 10:1759091418817641. [PMID: 33592687 PMCID: PMC6295693 DOI: 10.1177/1759091418817641] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2018] [Revised: 11/07/2018] [Accepted: 11/09/2018] [Indexed: 12/29/2022] Open
Abstract
Ion channel gating and kinase regulation of N-methyl-D-aspartate receptor 1 activity are fundamental mechanisms that govern synaptic plasticity. In this study, we showed that two mutant models (16p11.2del and Δdisc1 ) that recapitulate aspects of human cognitive disorders shared a similar defect in N-methyl-D-aspartate receptor 1-dependent synaptic function. Our results demonstrate that the expression of small-conductance potassium channels (SK2 or KCa2.2) was significantly upregulated in the hippocampus and prefrontal cortex of 16p11.2del and 129S:Δdisc1 mutant mice. Likewise, both mutant strains exhibited an impairment of T286 phosphorylation of calcium-calmodulin-dependent kinase II (CaMKII) in the hippocampus and prefrontal cortex. In vivo neural recordings revealed that increased SK2 expression and impaired T286 phosphorylation of CaMKII coincide with a prolonged interspike interval in the hippocampal cornu ammonis-1 (CA1) field for both 16p11.2del and 129S:Δdisc1 mutant mice. These findings suggest that alteration of small conductance channels and T286 phosphorylation of CaMKII are likely shared factors underlying behavioral changes in these two genetic mouse models.
Collapse
Affiliation(s)
- Razia Sultana
- Department of Comparative Biomedical Sciences, Louisiana State
University School of Veterinary Medicine, Baton Rouge, LA, USA
| | - Tanya Ghandi
- Department of Comparative Biomedical Sciences, Louisiana State
University School of Veterinary Medicine, Baton Rouge, LA, USA
| | - Alexandra M. Davila
- Department of Comparative Biomedical Sciences, Louisiana State
University School of Veterinary Medicine, Baton Rouge, LA, USA
| | - Charles C. Lee
- Department of Comparative Biomedical Sciences, Louisiana State
University School of Veterinary Medicine, Baton Rouge, LA, USA
| | - Olalekan M. Ogundele
- Department of Comparative Biomedical Sciences, Louisiana State
University School of Veterinary Medicine, Baton Rouge, LA, USA
| |
Collapse
|
40
|
Villalobo A, Ishida H, Vogel HJ, Berchtold MW. Calmodulin as a protein linker and a regulator of adaptor/scaffold proteins. BIOCHIMICA ET BIOPHYSICA ACTA-MOLECULAR CELL RESEARCH 2017; 1865:507-521. [PMID: 29247668 DOI: 10.1016/j.bbamcr.2017.12.004] [Citation(s) in RCA: 62] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/16/2017] [Revised: 12/07/2017] [Accepted: 12/08/2017] [Indexed: 01/29/2023]
Abstract
Calmodulin (CaM) is a universal regulator for a huge number of proteins in all eukaryotic cells. Best known is its function as a calcium-dependent modulator of the activity of enzymes, such as protein kinases and phosphatases, as well as other signaling proteins including membrane receptors, channels and structural proteins. However, less well known is the fact that CaM can also function as a Ca2+-dependent adaptor protein, either by bridging between different domains of the same protein or by linking two identical or different target proteins together. These activities are possible due to the fact that CaM contains two independently-folded Ca2+ binding lobes that are able to interact differentially and to some degree separately with targets proteins. In addition, CaM can interact with and regulates several proteins that function exclusively as adaptors. This review provides an overview over our present knowledge concerning the structural and functional aspects of the role of CaM as an adaptor protein and as a regulator of known adaptor/scaffold proteins.
Collapse
Affiliation(s)
- Antonio Villalobo
- Department of Cancer Biology, Instituto de Investigaciones Biomédicas, Consejo Superior de Investigaciones Científicas and Universidad Autónoma de Madrid, Arturo Duperier 4, E-28029 Madrid, Spain.
| | - Hiroaki Ishida
- Department of Biological Sciences, University of Calgary, 2500 University Dr. N.W., Calgary, Alberta T2N 1N4, Canada
| | - Hans J Vogel
- Department of Biological Sciences, University of Calgary, 2500 University Dr. N.W., Calgary, Alberta T2N 1N4, Canada.
| | - Martin W Berchtold
- Department of Biology, University of Copenhagen, 13 Universitetsparken, DK-2100 Copenhagen Ø, Denmark.
| |
Collapse
|
41
|
Yang M, Camara AKS, Aldakkak M, Kwok WM, Stowe DF. Identity and function of a cardiac mitochondrial small conductance Ca 2+-activated K + channel splice variant. BIOCHIMICA ET BIOPHYSICA ACTA. BIOENERGETICS 2017; 1858:442-458. [PMID: 28342809 PMCID: PMC5749404 DOI: 10.1016/j.bbabio.2017.03.005] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/11/2016] [Revised: 02/28/2017] [Accepted: 03/21/2017] [Indexed: 01/07/2023]
Abstract
We provide evidence for location and function of a small conductance, Ca2+-activated K+ (SKCa) channel isoform 3 (SK3) in mitochondria (m) of guinea pig, rat and human ventricular myocytes. SKCa agonists protected isolated hearts and mitochondria against ischemia/reperfusion (IR) injury; SKCa antagonists worsened IR injury. Intravenous infusion of a SKCa channel agonist/antagonist, respectively, in intact rats was effective in reducing/enhancing regional infarct size induced by coronary artery occlusion. Localization of SK3 in mitochondria was evidenced by Western blot of inner mitochondrial membrane, immunocytochemical staining of cardiomyocytes, and immunogold labeling of isolated mitochondria. We identified a SK3 splice variant in guinea pig (SK3.1, aka SK3a) and human ventricular cells (SK3.2) by amplifying mRNA, and show mitochondrial expression in mouse atrial tumor cells (HL-1) by transfection with full length and truncated SK3.1 protein. We found that the N-terminus is not required for mitochondrial trafficking but the C-terminus beyond the Ca2+ calmodulin binding domain is required for Ca2+ sensing to induce mK+ influx and/or promote mitochondrial localization. In isolated guinea pig mitochondria and in SK3 overexpressed HL-1 cells, mK+ influx was driven by adding CaCl2. Moreover, there was a greater fall in membrane potential (ΔΨm), and enhanced cell death with simulated cell injury after silencing SK3.1 with siRNA. Although SKCa channel opening protects the heart and mitochondria against IR injury, the mechanism for favorable bioenergetics effects resulting from SKCa channel opening remains unclear. SKCa channels could play an essential role in restraining cardiac mitochondria from inducing oxidative stress-induced injury resulting from mCa2+ overload.
Collapse
Affiliation(s)
- MeiYing Yang
- Department of Anesthesiology, Medical College of Wisconsin, Milwaukee, WI, USA
| | - Amadou K S Camara
- Department of Anesthesiology, Medical College of Wisconsin, Milwaukee, WI, USA; Cardiovascular Center, Medical College of Wisconsin, Milwaukee, WI, USA
| | - Mohammed Aldakkak
- Department of Anesthesiology, Medical College of Wisconsin, Milwaukee, WI, USA
| | - Wai-Meng Kwok
- Department of Anesthesiology, Medical College of Wisconsin, Milwaukee, WI, USA; Department of Pharmacology and Toxicology, Medical College of Wisconsin, Milwaukee, WI, USA; Cardiovascular Center, Medical College of Wisconsin, Milwaukee, WI, USA
| | - David F Stowe
- Department of Anesthesiology, Medical College of Wisconsin, Milwaukee, WI, USA; Department of Physiology, Medical College of Wisconsin, Milwaukee, WI, USA; Department of Biomedical Engineering, Medical College of Wisconsin and Marquette University, Milwaukee, WI, USA; Cardiovascular Center, Medical College of Wisconsin, Milwaukee, WI, USA; Research Service, Zablocki VA Medical Center, Milwaukee, WI, USA.
| |
Collapse
|
42
|
Enhanced Sensitivity to Hyperpolarizing Inhibition in Mesoaccumbal Relative to Nigrostriatal Dopamine Neuron Subpopulations. J Neurosci 2017; 37:3311-3330. [PMID: 28219982 DOI: 10.1523/jneurosci.2969-16.2017] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2016] [Revised: 02/02/2017] [Accepted: 02/10/2017] [Indexed: 11/21/2022] Open
Abstract
Midbrain dopamine neurons recorded in vivo pause their firing in response to reward omission and aversive stimuli. While the initiation of pauses typically involves synaptic or modulatory input, intrinsic membrane properties may also enhance or limit hyperpolarization, raising the question of how intrinsic conductances shape pauses in dopamine neurons. Using retrograde labeling and electrophysiological techniques combined with computational modeling, we examined the intrinsic conductances that shape pauses evoked by current injections and synaptic stimulation in subpopulations of dopamine neurons grouped according to their axonal projections to the nucleus accumbens or dorsal striatum in mice. Testing across a range of conditions and pulse durations, we found that mesoaccumbal and nigrostriatal neurons differ substantially in rebound properties with mesoaccumbal neurons displaying significantly longer delays to spiking following hyperpolarization. The underlying mechanism involves an inactivating potassium (IA) current with decay time constants of up to 225 ms, and small-amplitude hyperpolarization-activated currents (IH), characteristics that were most often observed in mesoaccumbal neurons. Pharmacological block of IA completely abolished rebound delays and, importantly, shortened synaptically evoked inhibitory pauses, thereby demonstrating the involvement of A-type potassium channels in prolonging pauses evoked by GABAergic inhibition. Therefore, these results show that mesoaccumbal and nigrostriatal neurons display differential responses to hyperpolarizing inhibitory stimuli that favors a higher sensitivity to inhibition in mesoaccumbal neurons. These findings may explain, in part, observations from in vivo experiments that ventral tegmental area neurons tend to exhibit longer aversive pauses relative to SNc neurons.SIGNIFICANCE STATEMENT Our study examines rebound, postburst, and synaptically evoked inhibitory pauses in subpopulations of midbrain dopamine neurons. We show that pauses in dopamine neuron firing, evoked by either stimulation of GABAergic inputs or hyperpolarizing current injections, are enhanced by a subclass of potassium conductances that are recruited at voltages below spike threshold. Importantly, A-type potassium currents recorded in mesoaccumbal neurons displayed substantially slower inactivation kinetics, which, combined with weaker expression of hyperpolarization-activated currents, lengthened hyperpolarization-induced delays in spiking relative to nigrostriatal neurons. These results suggest that input integration differs among dopamine neurons favoring higher sensitivity to inhibition in mesoaccumbal neurons and may partially explain in vivo observations that ventral tegmental area neurons exhibit longer aversive pauses relative to SNc neurons.
Collapse
|
43
|
Yi M, Yu P, Lu Q, Geller HM, Yu Z, Chen H. KCa3.1 constitutes a pharmacological target for astrogliosis associated with Alzheimer's disease. Mol Cell Neurosci 2016; 76:21-32. [PMID: 27567685 DOI: 10.1016/j.mcn.2016.08.008] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2016] [Revised: 07/28/2016] [Accepted: 08/23/2016] [Indexed: 01/08/2023] Open
Abstract
Alzheimer's disease (AD) is the most common type of dementia and is characterized by a progression from decline of episodic memory to a global impairment of cognitive function. Astrogliosis is a hallmark feature of AD, and reactive gliosis has been considered as an important target for intervention in various neurological disorders. We previously found in astrocyte cultures that the expression of the intermediate conductance calcium-activated potassium channel KCa3.1 was increased in reactive astrocytes induced by TGF-β, while pharmacological blockade or genetic deletion of KCa3.1 attenuated astrogliosis. In this study, we sought to suppress reactive gliosis in the context of AD by inhibiting KCa3.1 and evaluate its effects on the cognitive impairment using murine animal models such as the senescence-accelerated mouse prone 8 (SAMP8) model that exhibits some AD-like symptoms. We found KCa3.1 expression was increased in reactive astrocytes as well as neurons in the brains of both SAMP8 mice and Alzheimer's disease patients. Blockade of KCa3.1 with the selective inhibitor TRAM-34 in SAMP8 mice resulted in a decrease in astrogliosis as well as microglia activation, and moreover an attenuation of memory deficits. Using KCa3.1 knockout mice, we further confirmed that deletion of KCa3.1 reduced the activation of astrocytes and microglia, and rescued the memory loss induced by intrahippocampal Aβ1-42 peptide injection. We also found in astrocyte cultures that blockade of KCa3.1 or deletion of KCa3.1 suppressed Aβ oligomer-induced astrogliosis. Our data suggest that KCa3.1 inhibition might represent a promising therapeutic strategy for AD treatment.
Collapse
Affiliation(s)
- Mengni Yi
- Department of Pharmacology, Institute of Medical Sciences, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Panpan Yu
- Guangdong-Hongkong-Macau Institute of CNS Regeneration; Ministry of Education Joint International Research Laboratory of CNS Regeneration, Jinan University, Guangzhou 510632, China
| | - Qin Lu
- Department of Pharmacology, Institute of Medical Sciences, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Herbert M Geller
- Developmental Neurobiology Section, Division of Intramural Research, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | - Zhihua Yu
- Department of Pharmacology, Institute of Medical Sciences, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China.
| | - Hongzhuan Chen
- Department of Pharmacology, Institute of Medical Sciences, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China.
| |
Collapse
|
44
|
Underwood EL, Thompson LT. High-fat diet impairs spatial memory and hippocampal intrinsic excitability and sex-dependently alters circulating insulin and hippocampal insulin sensitivity. Biol Sex Differ 2016; 7:9. [PMID: 26823968 PMCID: PMC4730722 DOI: 10.1186/s13293-016-0060-3] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/11/2015] [Accepted: 01/18/2016] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND High-fat diets promoting obesity/type-2 diabetes can impair physiology and cognitive performance, although sex-dependent comparisons of these impairments are rarely made. Transient reductions in Ca(2+)-dependent afterhyperpolarizations (AHPs) occur during memory consolidation, enhancing intrinsic excitability of hippocampal CA1 pyramidal neurons. In rats fed standard diets, insulin can enhance memory and reduce amplitude and duration of AHPs. METHODS Effects of chronic high-fat diet (HFD) on memory, circulating insulin, and neuronal physiology were compared between young adult male and female Long-Evans rats. Rats fed for 12 weeks (from weaning) a HFD or a control diet (CD) were then tested in vivo prior to in vitro recordings from CA1 pyramidal neurons. RESULTS The HFD significantly impaired spatial memory in both males and females. Significant sex differences occurred in circulating insulin and in the insulin sensitivity of hippocampal neurons. Circulating insulin significantly increased in HFD males but decreased in HFD females. While the HFD significantly reduced hippocampal intrinsic excitability in both sexes, CA1 neurons from HFD females remained insulin-sensitive but those from HFD males became insulin-insensitive. CONCLUSIONS Findings consistent with these have been characterized previously in HFD or senescent males, but the effects observed here in young females are unique. Loss of CA1 neuronal excitability, and sex-dependent loss of insulin sensitivity, can have significant cognitive consequences, over both the short term and the life span. These findings highlight needs for more research into sex-dependent differences, relating systemic and neural plasticity mechanisms in metabolic disorders.
Collapse
Affiliation(s)
- Erica L. Underwood
- Cognition & Neuroscience Program, School of Behavioral & Brain Sciences, University of Texas at Dallas, 800 W. Campbell Rd., Richardson, TX 75080 USA
| | - Lucien T. Thompson
- Cognition & Neuroscience Program, School of Behavioral & Brain Sciences, University of Texas at Dallas, 800 W. Campbell Rd., Richardson, TX 75080 USA
| |
Collapse
|
45
|
Abdulkareem ZA, Gee JMW, Cox CD, Wann KT. Knockdown of the small conductance Ca(2+) -activated K(+) channels is potently cytotoxic in breast cancer cell lines. Br J Pharmacol 2016; 173:177-90. [PMID: 26454020 PMCID: PMC4737296 DOI: 10.1111/bph.13357] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2015] [Revised: 08/27/2015] [Accepted: 09/24/2015] [Indexed: 12/31/2022] Open
Abstract
BACKGROUND AND PURPOSE Small conductance calcium-activated potassium (KCa 2.x) channels have a widely accepted canonical function in regulating cellular excitability. In this study, we address a potential non-canonical function of KCa 2.x channels in breast cancer cell survival, using in vitro models. EXPERIMENTAL APPROACH The expression of all KCa 2.x channel isoforms was initially probed using RT-PCR, Western blotting and microarray analysis in five widely studied breast cancer cell lines. In order to assess the effect of pharmacological blockade and siRNA-mediated knockdown of KCa 2.x channels on these cell lines, we utilized MTS proliferation assays and also followed the corresponding expression of apoptotic markers. KEY RESULTS All of the breast cancer cell lines, regardless of their lineage or endocrine responsiveness, were highly sensitive to KCa 2.x channel blockade. UCL1684 caused cytotoxicity, with LD50 values in the low nanomolar range, in all cell lines. The role of KCa 2.x channels was confirmed using pharmacological inhibition and siRNA-mediated knockdown. This reduced cell viability and also reduced expression of Bcl-2 but increased expression of active caspase-7 and caspase-9. Complementary to these results, a variety of cell lines can be protected from apoptosis induced by staurosporine using the KCa 2.x channel activator CyPPA. CONCLUSIONS AND IMPLICATIONS In addition to a well-established role for KCa 2.x channels in migration, blockade of these channels was potently cytotoxic in breast cancer cell lines, pointing to modulation of KCa 2.x channels as a potential therapeutic approach to breast cancer.
Collapse
Affiliation(s)
| | - Julia MW Gee
- School of Pharmacy and Pharmaceutical SciencesCardiff UniversityCardiffCF10 3NBUK
| | - Charles D Cox
- Victor Chang Cardiac Research InstituteDarlinghurstNSW2010Australia
| | - Kenneth T Wann
- School of Pharmacy and Pharmaceutical SciencesCardiff UniversityCardiffCF10 3NBUK
| |
Collapse
|
46
|
Sihn CR, Kim HJ, Woltz RL, Yarov-Yarovoy V, Yang PC, Xu J, Clancy CE, Zhang XD, Chiamvimonvat N, Yamoah EN. Mechanisms of Calmodulin Regulation of Different Isoforms of Kv7.4 K+ Channels. J Biol Chem 2015; 291:2499-509. [PMID: 26515070 DOI: 10.1074/jbc.m115.668236] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2015] [Indexed: 01/17/2023] Open
Abstract
Calmodulin (CaM), a Ca(2+)-sensing protein, is constitutively bound to IQ domains of the C termini of human Kv7 (hKv7, KCNQ) channels to mediate Ca(2+)-dependent reduction of Kv7 currents. However, the mechanism remains unclear. We report that CaM binds to two isoforms of the hKv7.4 channel in a Ca(2+)-independent manner but that only the long isoform (hKv7.4a) is regulated by Ca(2+)/CaM. Ca(2+)/CaM mediate reduction of the hKv7.4a channel by decreasing the channel open probability and altering activation kinetics. We took advantage of a known missense mutation (G321S) that has been linked to progressive hearing loss to further examine the inhibitory effects of Ca(2+)/CaM on the Kv7.4 channel. Using multidisciplinary techniques, we demonstrate that the G321S mutation may destabilize CaM binding, leading to a decrease in the inhibitory effects of Ca(2+) on the channels. Our study utilizes an expression system to dissect the biophysical properties of the WT and mutant Kv7.4 channels. This report provides mechanistic insights into the critical roles of Ca(2+)/CaM regulation of the Kv7.4 channel under physiological and pathological conditions.
Collapse
Affiliation(s)
- Choong-Ryoul Sihn
- From the Department of Physiology and Cell Biology, Program in Communication Science, School of Medicine, University of Nevada, Reno, Reno, Nevada 85997
| | - Hyo Jeong Kim
- the Department of Internal Medicine, Division of Cardiovascular Medicine
| | - Ryan L Woltz
- the Department of Internal Medicine, Division of Cardiovascular Medicine
| | - Vladimir Yarov-Yarovoy
- Department of Physiology and Membrane Biology, and the Northern California Health Care System, Department of Veterans Affairs, Mather, California 95655, and
| | - Pei-Chi Yang
- Department of Pharmacology, University of California, Davis, Davis, California 95616
| | - Jun Xu
- the Department of Engineering Technology, College of Science and Technology, Tarleton State University, Stephenville, Texas 76402
| | - Colleen E Clancy
- Department of Pharmacology, University of California, Davis, Davis, California 95616
| | - Xiao-Dong Zhang
- the Department of Internal Medicine, Division of Cardiovascular Medicine, the Northern California Health Care System, Department of Veterans Affairs, Mather, California 95655, and
| | - Nipavan Chiamvimonvat
- the Department of Internal Medicine, Division of Cardiovascular Medicine, the Northern California Health Care System, Department of Veterans Affairs, Mather, California 95655, and
| | - Ebenezer N Yamoah
- From the Department of Physiology and Cell Biology, Program in Communication Science, School of Medicine, University of Nevada, Reno, Reno, Nevada 85997,
| |
Collapse
|
47
|
Humphries ESA, Dart C. Neuronal and Cardiovascular Potassium Channels as Therapeutic Drug Targets: Promise and Pitfalls. JOURNAL OF BIOMOLECULAR SCREENING 2015; 20:1055-73. [PMID: 26303307 PMCID: PMC4576507 DOI: 10.1177/1087057115601677] [Citation(s) in RCA: 53] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/31/2015] [Revised: 07/26/2015] [Accepted: 07/28/2015] [Indexed: 12/21/2022]
Abstract
Potassium (K(+)) channels, with their diversity, often tissue-defined distribution, and critical role in controlling cellular excitability, have long held promise of being important drug targets for the treatment of dysrhythmias in the heart and abnormal neuronal activity within the brain. With the exception of drugs that target one particular class, ATP-sensitive K(+) (KATP) channels, very few selective K(+) channel activators or inhibitors are currently licensed for clinical use in cardiovascular and neurological disease. Here we review what a range of human genetic disorders have told us about the role of specific K(+) channel subunits, explore the potential of activators and inhibitors of specific channel populations as a therapeutic strategy, and discuss possible reasons for the difficulty in designing clinically relevant K(+) channel modulators.
Collapse
Affiliation(s)
| | - Caroline Dart
- Institute of Integrative Biology, University of Liverpool, UK
| |
Collapse
|
48
|
Loppini A, Braun M, Filippi S, Pedersen MG. Mathematical modeling of gap junction coupling and electrical activity in human β-cells. Phys Biol 2015; 12:066002. [PMID: 26403477 DOI: 10.1088/1478-3975/12/6/066002] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022]
Abstract
Coordinated insulin secretion is controlled by electrical coupling of pancreatic β-cells due to connexin-36 gap junctions. Gap junction coupling not only synchronizes the heterogeneous β-cell population, but can also modify the electrical behavior of the cells. These phenomena have been widely studied with mathematical models based on data from mouse β-cells. However, it is now known that human β-cell electrophysiology shows important differences to its rodent counterpart, and although human pancreatic islets express connexin-36 and show evidence of β-cell coupling, these aspects have been little investigated in human β-cells. Here we investigate theoretically, the gap junction coupling strength required for synchronizing electrical activity in a small cluster of cells simulated with a recent mathematical model of human β-cell electrophysiology. We find a lower limit for the coupling strength of approximately 20 pS (i.e., normalized to cell size, ∼2 pS pF(-1)) below which spiking electrical activity is asynchronous. To confront this theoretical lower bound with data, we use our model to estimate from an experimental patch clamp recording that the coupling strength is approximately 100-200 pS (10-20 pS pF(-1)), similar to previous estimates in mouse β-cells. We then investigate the role of gap junction coupling in synchronizing and modifying other forms of electrical activity in human β-cell clusters. We find that electrical coupling can prolong the period of rapid bursting electrical activity, and synchronize metabolically driven slow bursting, in particular when the metabolic oscillators are in phase. Our results show that realistic coupling conductances are sufficient to promote synchrony in small clusters of human β-cells as observed experimentally, and provide motivation for further detailed studies of electrical coupling in human pancreatic islets.
Collapse
Affiliation(s)
- Alessandro Loppini
- Nonlinear Physics and Mathematical Modeling Laboratory, University Campus Bio-Medico, I-00128, Rome, Italy
| | | | | | | |
Collapse
|
49
|
Mizukami K, Yokoshiki H, Mitsuyama H, Watanabe M, Tenma T, Takada S, Tsutsui H. Small-conductance Ca2+-activated K+ current is upregulated via the phosphorylation of CaMKII in cardiac hypertrophy from spontaneously hypertensive rats. Am J Physiol Heart Circ Physiol 2015; 309:H1066-74. [DOI: 10.1152/ajpheart.00825.2014] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/12/2014] [Accepted: 08/12/2015] [Indexed: 01/09/2023]
Abstract
Left ventricular hypertrophy is associated with an increased risk of ventricular arrhythmias. However, the underlying molecular basis is poorly understood. It has been reported that small-conductance Ca2+-activated K+ (SK) channels are involved in the pathogenesis of ventricular arrhythmias in heart failure. The present study aimed to test the hypothesis that SK channel activity is increased via the Ca2+/calmodulin-dependent protein kinase II (CaMKII)-dependent pathway in hypertensive cardiac hypertrophy. Normotensive Wistar-Kyoto (WKY) rats and spontaneous hypertensive rats (SHRs) were used. Whole cell membrane currents were recorded in isolated ventricular myocytes by the patch-clamp method, and apamin-sensitive K+ current ( IKAS), which is inhibited by apamin (100 nM), an SK channel blocker, was evaluated. IKAS at 40 mV was present in SHRs, whereas it was hardly detectable in WKY rats (0.579 ± 0.046 vs. 0.022 ± 0.062 pA/pF, both n = 6, P < 0.05). IKAS was almost completely abolished by 1 μM KN-93, a CaMKII inhibitor, in SHRs. Optical recordings of left ventricular anterior wall action potentials revealed that apamin prolonged the late phase of repolarization only in SHRs. Western blot analysis of SK channel protein isoforms demonstrated that SK2 was significantly increased in SHRs compared with WKY rats (SK2/GAPDH: 0.66 ± 0.07 vs. 0.40 ± 0.02, both n = 6, P < 0.05), whereas SK1 and SK3 did not differ between groups. In addition, autophosphorylated CaMKII was significantly increased in SHRs (phosphorylated CaMKII/GAPDH: 0.80 ± 0.06 vs. 0.58 ± 0.06, both n = 6, P < 0.05) despite a comparable total amount of CaMKII between groups. In conclusion, SK channels are upregulated via the enhanced activation of CaMKII in cardiac hypertrophy in SHRs.
Collapse
Affiliation(s)
- Kazuya Mizukami
- Department of Cardiovascular Medicine, Hokkaido University Graduate School of Medicine, Sapporo, Japan
| | - Hisashi Yokoshiki
- Department of Cardiovascular Medicine, Hokkaido University Graduate School of Medicine, Sapporo, Japan
| | - Hirofumi Mitsuyama
- Department of Cardiovascular Medicine, Hokkaido University Graduate School of Medicine, Sapporo, Japan
| | - Masaya Watanabe
- Department of Cardiovascular Medicine, Hokkaido University Graduate School of Medicine, Sapporo, Japan
| | - Taro Tenma
- Department of Cardiovascular Medicine, Hokkaido University Graduate School of Medicine, Sapporo, Japan
| | - Shingo Takada
- Department of Cardiovascular Medicine, Hokkaido University Graduate School of Medicine, Sapporo, Japan
| | - Hiroyuki Tsutsui
- Department of Cardiovascular Medicine, Hokkaido University Graduate School of Medicine, Sapporo, Japan
| |
Collapse
|
50
|
Zhang XD, Lieu DK, Chiamvimonvat N. Small-conductance Ca2+ -activated K+ channels and cardiac arrhythmias. Heart Rhythm 2015; 12:1845-51. [PMID: 25956967 PMCID: PMC4662728 DOI: 10.1016/j.hrthm.2015.04.046] [Citation(s) in RCA: 53] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/11/2015] [Indexed: 01/04/2023]
Abstract
Small-conductance Ca2+ -activated K+ (SK, KCa2) channels are unique in that they are gated solely by changes in intracellular Ca2+ and, hence, function to integrate intracellular Ca2+ and membrane potentials on a beat-to-beat basis. Recent studies have provided evidence for the existence and functional significance of SK channels in the heart. Indeed, our knowledge of cardiac SK channels has been greatly expanded over the past decade. Interests in cardiac SK channels are further driven by recent studies suggesting the critical roles of SK channels in human atrial fibrillation, the SK channel as a possible novel therapeutic target in atrial arrhythmias, and upregulation of SK channels in heart failure in animal models and in human heart failure. However, there remain critical gaps in our knowledge. Specifically, blockade of SK channels in cardiac arrhythmias has been shown to be both antiarrhythmic and proarrhythmic. This contemporary review provides an overview of the literature on the role of cardiac SK channels in cardiac arrhythmias and serves as a discussion platform for the current clinical perspectives. At the translational level, development of SK channel blockers as a new therapeutic strategy in the treatment of atrial fibrillation and the possible proarrhythmic effects merit further considerations and investigations.
Collapse
Affiliation(s)
- Xiao-Dong Zhang
- Division of Cardiovascular Medicine, Department of Internal Medicine, University of California, Davis, California.
| | - Deborah K Lieu
- Division of Cardiovascular Medicine, Department of Internal Medicine, University of California, Davis, California
| | - Nipavan Chiamvimonvat
- Division of Cardiovascular Medicine, Department of Internal Medicine, University of California, Davis, California; Department of Veterans Affairs, Northern California Health Care System, Mather, California.
| |
Collapse
|