1
|
Palmisano VF, Anguita-Ortiz N, Faraji S, Nogueira JJ. Voltage-Gated Ion Channels: Structure, Pharmacology and Photopharmacology. Chemphyschem 2024; 25:e202400162. [PMID: 38649320 DOI: 10.1002/cphc.202400162] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2024] [Revised: 04/21/2024] [Accepted: 04/22/2024] [Indexed: 04/25/2024]
Abstract
Voltage-gated ion channels are transmembrane proteins responsible for the generation and propagation of action potentials in excitable cells. Over the last decade, advancements have enabled the elucidation of crystal structures of ion channels. This progress in structural understanding, particularly in identifying the binding sites of local anesthetics, opens avenues for the design of novel compounds capable of modulating ion conduction. However, many traditional drugs lack selectivity and come with adverse side effects. The emergence of photopharmacology has provided an orthogonal way of controlling the activity of compounds, enabling the regulation of ion conduction with light. In this review, we explore the central pore region of voltage-gated sodium and potassium channels, providing insights from both structural and pharmacological perspectives. We discuss the different binding modes of synthetic compounds that can physically occlude the pore and, therefore, block ion conduction. Moreover, we examine recent advances in the photopharmacology of voltage-gated ion channels, introducing molecular approaches aimed at controlling their activity by using photosensitive drugs.
Collapse
Affiliation(s)
- Vito F Palmisano
- Department of Chemistry, Universidad Autónoma de Madrid, 28049, Madrid, Spain
- Theoretical Chemistry Group, Zernike Institute for Advanced Materials, University of Groningen, 9747 AG, Groningen, The Netherlands
| | - Nuria Anguita-Ortiz
- Department of Chemistry, Universidad Autónoma de Madrid, 28049, Madrid, Spain
| | - Shirin Faraji
- Theoretical Chemistry Group, Zernike Institute for Advanced Materials, University of Groningen, 9747 AG, Groningen, The Netherlands
| | - Juan J Nogueira
- Department of Chemistry, Universidad Autónoma de Madrid, 28049, Madrid, Spain
- IADCHEM, Institute for Advanced Research in Chemistry, Universidad Autónoma de Madrid, 28049, Madrid, Spain
| |
Collapse
|
2
|
Zhou Q, Hao G, Xie W, Chen B, Lu W, Wang G, Zhong R, Chen J, Ye J, Shen J, Cao P. Exenatide reduces atrial fibrillation susceptibility by inhibiting hKv1.5 and hNav1.5 channels. J Biol Chem 2024; 300:107294. [PMID: 38636665 PMCID: PMC11109313 DOI: 10.1016/j.jbc.2024.107294] [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: 06/15/2023] [Revised: 04/02/2024] [Accepted: 04/12/2024] [Indexed: 04/20/2024] Open
Abstract
Exenatide, a promising cardioprotective agent, protects against cardiac structural remodeling and diastolic dysfunction. Combined blockade of sodium and potassium channels is valuable for managing atrial fibrillation (AF). Here, we explored whether exenatide displayed anti-AF effects by inhibiting human Kv1.5 and Nav1.5 channels. We used the whole-cell patch-clamp technique to investigate the effects of exenatide on hKv1.5 and hNav1.5 channels expressed in human embryonic kidney 293 cells and studied the effects of exenatide on action potential (AP) and other cardiac ionic currents in rat atrial myocytes. Additionally, an electrical mapping system was used to explore the effects of exenatide on electrical properties and AF activity in isolated rat hearts. Finally, a rat AF model, established using acetylcholine and calcium chloride, was employed to evaluate the anti-AF potential of exenatide in rats. Exenatide reversibly suppressed IKv1.5 with IC50 of 3.08 μM, preferentially blocked the hKv1.5 channel in its closed state, and positively shifted the voltage-dependent activation curve. Exenatide also reversibly inhibited INav1.5 with IC50 of 3.30 μM, negatively shifted the voltage-dependent inactivation curve, and slowed its recovery from inactivation with significant use-dependency at 5 and 10 Hz. Furthermore, exenatide prolonged AP duration and suppressed the sustained K+ current (Iss) and transient outward K+ current (Ito), but without inhibition of L-type Ca2+ current (ICa,L) in rat atrial myocytes. Exenatide prevented AF incidence and duration in rat hearts and rats. These findings demonstrate that exenatide inhibits IKv1.5 and INav1.5in vitro and reduces AF susceptibility in isolated rat hearts and rats.
Collapse
Affiliation(s)
- Qian Zhou
- Jiangsu Provincial Medical Innovation Center, Affiliated Hospital of Integrated Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing, China
| | - Guoliang Hao
- Institute of Electrophysiology, Henan Academy of Innovations in Medical Science, Zhengzhou, China; Henan SCOPE Research Institute of Electrophysiology Co Ltd, Kaifeng, China; Burdon Sanderson Cardiac Science Centre and BHF Centre of Research Excellence, Department of Physiology, Anatomy and Genetics, University of Oxford, Oxford, UK
| | - Wensen Xie
- Jiangsu Provincial Medical Innovation Center, Affiliated Hospital of Integrated Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing, China
| | - Bin Chen
- Jiangsu Provincial Medical Innovation Center, Affiliated Hospital of Integrated Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing, China; Nanjing Research Institute for Comprehensive Utilization of Wild Plants, Nanjing, China
| | - Wuguang Lu
- Jiangsu Provincial Medical Innovation Center, Affiliated Hospital of Integrated Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing, China
| | - Gongxin Wang
- Institute of Electrophysiology, Henan Academy of Innovations in Medical Science, Zhengzhou, China; Henan SCOPE Research Institute of Electrophysiology Co Ltd, Kaifeng, China
| | - Rongling Zhong
- Jiangsu Provincial Medical Innovation Center, Affiliated Hospital of Integrated Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing, China
| | - Jiao Chen
- Jiangsu Provincial Medical Innovation Center, Affiliated Hospital of Integrated Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing, China
| | - Juan Ye
- Jiangsu Provincial Medical Innovation Center, Affiliated Hospital of Integrated Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing, China
| | - Jianping Shen
- Jiangsu Provincial Medical Innovation Center, Affiliated Hospital of Integrated Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing, China
| | - Peng Cao
- Jiangsu Provincial Medical Innovation Center, Affiliated Hospital of Integrated Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing, China; State Key Laboratory on Technologies for Chinese Medicine Pharmaceutical Process Control and Intelligent Manufacture, Nanjing University of Chinese Medicine, Nanjing, China; Animal-Derived Chinese Medicine and Functional Peptides International Collaboration Joint Laboratory, Nanjing University of Chinese Medicine, Nanjing, China; Shandong Academy of Chinese Medicine, Jinan, China.
| |
Collapse
|
3
|
Goodchild SJ, Shuart NG, Williams AD, Ye W, Parrish RR, Soriano M, Thouta S, Mezeyova J, Waldbrook M, Dean R, Focken T, Ghovanloo MR, Ruben PC, Scott F, Cohen CJ, Empfield J, Johnson JP. Molecular Pharmacology of Selective Na V1.6 and Dual Na V1.6/Na V1.2 Channel Inhibitors that Suppress Excitatory Neuronal Activity Ex Vivo. ACS Chem Neurosci 2024; 15:1169-1184. [PMID: 38359277 PMCID: PMC10958515 DOI: 10.1021/acschemneuro.3c00757] [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: 11/21/2023] [Revised: 01/25/2024] [Accepted: 01/30/2024] [Indexed: 02/17/2024] Open
Abstract
Voltage-gated sodium channel (NaV) inhibitors are used to treat neurological disorders of hyperexcitability such as epilepsy. These drugs act by attenuating neuronal action potential firing to reduce excitability in the brain. However, all currently available NaV-targeting antiseizure medications nonselectively inhibit the brain channels NaV1.1, NaV1.2, and NaV1.6, which potentially limits the efficacy and therapeutic safety margins of these drugs. Here, we report on XPC-7724 and XPC-5462, which represent a new class of small molecule NaV-targeting compounds. These compounds specifically target inhibition of the NaV1.6 and NaV1.2 channels, which are abundantly expressed in excitatory pyramidal neurons. They have a > 100-fold molecular selectivity against NaV1.1 channels, which are predominantly expressed in inhibitory neurons. Sparing NaV1.1 preserves the inhibitory activity in the brain. These compounds bind to and stabilize the inactivated state of the channels thereby reducing the activity of excitatory neurons. They have higher potency, with longer residency times and slower off-rates, than the clinically used antiseizure medications carbamazepine and phenytoin. The neuronal selectivity of these compounds is demonstrated in brain slices by inhibition of firing in cortical excitatory pyramidal neurons, without impacting fast spiking inhibitory interneurons. XPC-5462 also suppresses epileptiform activity in an ex vivo brain slice seizure model, whereas XPC-7224 does not, suggesting a possible requirement of Nav1.2 inhibition in 0-Mg2+- or 4-AP-induced brain slice seizure models. The profiles of these compounds will facilitate pharmacological dissection of the physiological roles of NaV1.2 and NaV1.6 in neurons and help define the role of specific channels in disease states. This unique selectivity profile provides a new approach to potentially treat disorders of neuronal hyperexcitability by selectively downregulating excitatory circuits.
Collapse
Affiliation(s)
- Samuel J. Goodchild
- Department
of Cellular and Molecular Biology, Xenon
Pharmaceuticals, Burnaby, BC V5G 4W8, Canada
| | - Noah Gregory Shuart
- Department
of Cellular and Molecular Biology, Xenon
Pharmaceuticals, Burnaby, BC V5G 4W8, Canada
| | - Aaron D. Williams
- Department
of Cellular and Molecular Biology, Xenon
Pharmaceuticals, Burnaby, BC V5G 4W8, Canada
| | - Wenlei Ye
- Neurocrine
Biosciences, San Diego, California 92130, United States
| | - R. Ryley Parrish
- Department
of Cellular and Molecular Biology, Xenon
Pharmaceuticals, Burnaby, BC V5G 4W8, Canada
| | - Maegan Soriano
- Department
of Cellular and Molecular Biology, Xenon
Pharmaceuticals, Burnaby, BC V5G 4W8, Canada
| | - Samrat Thouta
- Department
of Cellular and Molecular Biology, Xenon
Pharmaceuticals, Burnaby, BC V5G 4W8, Canada
| | - Janette Mezeyova
- Department
of Cellular and Molecular Biology, Xenon
Pharmaceuticals, Burnaby, BC V5G 4W8, Canada
| | - Matthew Waldbrook
- Department
of Cellular and Molecular Biology, Xenon
Pharmaceuticals, Burnaby, BC V5G 4W8, Canada
| | - Richard Dean
- Department
of Cellular and Molecular Biology, Xenon
Pharmaceuticals, Burnaby, BC V5G 4W8, Canada
| | - Thilo Focken
- Department
of Cellular and Molecular Biology, Xenon
Pharmaceuticals, Burnaby, BC V5G 4W8, Canada
| | - Mohammad-Reza Ghovanloo
- Department
of Cellular and Molecular Biology, Xenon
Pharmaceuticals, Burnaby, BC V5G 4W8, Canada
- Department
of Biomedical Physiology and Kinesiology, Simon Fraser University, Burnaby, BC V5A 1S6, Canada
- Department
of Neurology, Yale University, New Haven, Connecticut 06519, United States
| | - Peter C. Ruben
- Department
of Biomedical Physiology and Kinesiology, Simon Fraser University, Burnaby, BC V5A 1S6, Canada
| | - Fiona Scott
- Neurocrine
Biosciences, San Diego, California 92130, United States
| | - Charles J. Cohen
- Department
of Cellular and Molecular Biology, Xenon
Pharmaceuticals, Burnaby, BC V5G 4W8, Canada
| | - James Empfield
- Department
of Cellular and Molecular Biology, Xenon
Pharmaceuticals, Burnaby, BC V5G 4W8, Canada
| | - JP Johnson
- Department
of Cellular and Molecular Biology, Xenon
Pharmaceuticals, Burnaby, BC V5G 4W8, Canada
| |
Collapse
|
4
|
Zhuang W, Mun SY, Park M, Jeong J, Park H, Na S, Lee SJ, Jung WK, Choi IW, Li H, Park WS. Lurasidone blocks the voltage-gated potassium channels of coronary arterial smooth muscle cells. Eur J Pharmacol 2023; 957:176005. [PMID: 37611842 DOI: 10.1016/j.ejphar.2023.176005] [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: 05/29/2023] [Revised: 08/10/2023] [Accepted: 08/18/2023] [Indexed: 08/25/2023]
Abstract
Lurasidone is a second-generation antipsychotic drug used to treat schizophrenia, mania, and bipolar disorder. The drug is an antagonist of the 5-HT2A and D2 receptors. No effect of lurasidone on the voltage-gated K+ (Kv) channels has yet been identified. Here, we show that lurasidone inhibits the vascular Kv channels of rabbit coronary arterial smooth muscle cells in a dose-dependent manner with an IC50 of 1.88 ± 0.21 μM and a Hill coefficient of 0.98 ± 0.09. Although lurasidone (3 μM) did not affect the activation kinetics, the drug negatively shifted the inactivation curve, suggesting that the drug interacted with the voltage sensors of Kv channels. Application of 1 or 2 Hz train steps in the presence of lurasidone significantly increased Kv current inhibition. The recovery time after channel inactivation increased in the presence of lurasidone. These results suggest that the inhibitory action of lurasidone is use (state)-dependent. Pretreatment with a Kv 1.5 subtype inhibitor effectively reduced the inhibitory effect of lurasidone. However, the inhibitory effect on Kv channels did not markedly change after pretreatment with a Kv 2.1 or a Kv7 subtype inhibitor. In summary, lurasidone inhibits vascular Kv channels (primarily the Kv1.5 subtype) in a concentration- and use (state)-dependent manner by shifting the steady-state inactivation curve.
Collapse
Affiliation(s)
- Wenwen Zhuang
- Institute of Medical Sciences, Department of Physiology, Kangwon National University School of Medicine, Chuncheon, 24341, South Korea
| | - Seo-Yeong Mun
- Institute of Medical Sciences, Department of Physiology, Kangwon National University School of Medicine, Chuncheon, 24341, South Korea
| | - Minju Park
- Institute of Medical Sciences, Department of Physiology, Kangwon National University School of Medicine, Chuncheon, 24341, South Korea
| | - Junsu Jeong
- Institute of Medical Sciences, Department of Physiology, Kangwon National University School of Medicine, Chuncheon, 24341, South Korea
| | - Hongzoo Park
- Institute of Medical Sciences, Department of Urology, Kangwon National University Hospital, Kangwon National University School of Medicine, Chuncheon, 24341, South Korea
| | - Sunghun Na
- Institute of Medical Sciences, Department of Obstetrics and Gynecology, Kangwon National University Hospital, Kangwon National University School of Medicine, Chuncheon, 24341, South Korea
| | - Se Jin Lee
- Institute of Medical Sciences, Department of Obstetrics and Gynecology, Kangwon National University Hospital, Kangwon National University School of Medicine, Chuncheon, 24341, South Korea
| | - Won-Kyo Jung
- Department of Biomedical Engineering, Center for Marine-Integrated Biomedical Technology (BK21 Plus), Pukyong National University, Busan, 48513, South Korea
| | - Il-Whan Choi
- Department of Microbiology, College of Medicine, Inje University, Busan, 48516, South Korea
| | - Hongliang Li
- Institute of Translational Medicine, Medical College, Laboratory of Integrated Traditional Chinese and Western Medicine for Prevention and Treatment for Senile Diseases, Yangzhou University, Yangzhou, 225001, Jiangsu, China
| | - Won Sun Park
- Institute of Medical Sciences, Department of Physiology, Kangwon National University School of Medicine, Chuncheon, 24341, South Korea.
| |
Collapse
|
5
|
Marques LP, Santos-Miranda A, Joviano-Santos JV, Teixeira-Fonseca JL, Alcântara FDS, Sarmento JO, Roman-Campos D. The fungicide tebuconazole modulates the sodium current of human Na V1.5 channels expressed in HEK293 cells. Food Chem Toxicol 2023; 180:113992. [PMID: 37633639 DOI: 10.1016/j.fct.2023.113992] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2022] [Revised: 08/09/2023] [Accepted: 08/16/2023] [Indexed: 08/28/2023]
Abstract
The fungicide Tebuconazole is a widely used pesticide in agriculture and may cause cardiotoxicity. In our present investigation the effect of Tebuconazole on the sodium current (INa) of human cardiac sodium channels (NaV1.5) was studied using a heterologous expression system and whole-cell patch-clamp techniques. Tebuconazole reduced the amplitude of the peak INa in a concentration- and voltage-dependent manner. At the holding potential of -120 mV the IC50 was estimated at 204.1 ± 34.3 μM, while at -80 mV the IC50 was 0.3 ± 0.1 μM. The effect of the fungicide is more pronounced at more depolarized potentials, indicating a state-dependent interaction. Tebuconazole caused a negative shift in the half-maximal inactivation voltage and delayed recovery from fast inactivation of INa. Also, it enhanced closed-state inactivation, exhibited use-dependent block in a voltage-dependent manner. Furthermore, Tebuconazole reduced the increase in late sodium current induced by the pyrethroid insecticide β-Cyfluthrin. These results suggest that Tebuconazole can interact with NaV1.5 channels and modulate INa. The observed effects may lead to decreased cardiac excitability through reduced INa availability, which could be a new mechanism of cardiotoxicity to be attributed to the fungicide.
Collapse
Affiliation(s)
- Leisiane Pereira Marques
- Laboratory of Cardiobiology, Department of Biophysics, Escola Paulista de Medicina, Universidade Federal de São Paulo, São Paulo, SP, Brazil
| | - Artur Santos-Miranda
- Department of Physiology and Biophysics, Universidade Federal de Minas Gerais, Belo Horizonte, MG, Brazil
| | | | - Jorge Lucas Teixeira-Fonseca
- Laboratory of Cardiobiology, Department of Biophysics, Escola Paulista de Medicina, Universidade Federal de São Paulo, São Paulo, SP, Brazil
| | - Fabiana da Silva Alcântara
- Laboratory of Cardiobiology, Department of Biophysics, Escola Paulista de Medicina, Universidade Federal de São Paulo, São Paulo, SP, Brazil
| | - Jaqueline Oliveira Sarmento
- Laboratory of Cardiobiology, Department of Biophysics, Escola Paulista de Medicina, Universidade Federal de São Paulo, São Paulo, SP, Brazil
| | - Danilo Roman-Campos
- Laboratory of Cardiobiology, Department of Biophysics, Escola Paulista de Medicina, Universidade Federal de São Paulo, São Paulo, SP, Brazil.
| |
Collapse
|
6
|
Patel N, B Urolagin S, Haq MA, Patel C, Bhatt R, Girdhar G, Sinha S, Haque M, Kumar S. Anesthetic Effect of 2% Amitriptyline Versus 2% Lidocaine: A Comparative Evaluation. Cureus 2023; 15:e43405. [PMID: 37581201 PMCID: PMC10423460 DOI: 10.7759/cureus.43405] [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] [Accepted: 08/13/2023] [Indexed: 08/16/2023] Open
Abstract
Introduction A common dental problem is the fear of pain during needle prick for giving local anesthesia (LA). The needle prick pain during dental procedures often varies with sex and age. Perception of pain depends on various factors, which can be psychological and biological. This perception of pain may change the behavior of patients toward dental treatments. Traditionally, lidocaine gel formulation was utilized before the parenteral dosage form. The lidocaine gel formulation is considered the drug of choice for LA in dental surgery. Currently, amitriptyline has been utilized in dental practice because of its beneficial pharmacology. Hence, the present study has been undertaken to compare the anesthetic ability of amitriptyline as an intraoral topical anesthetic agent with lidocaine gel. Methods This study was a comparative clinical study between two medications' anesthetic properties. This study included 120 patients indicated for bilateral orthodontics (the subdivision of dentistry that emphasizes identifying necessary interventions for the malocclusion of teeth) procedures. All the subjects were divided into amitriptyline and lidocaine groups. Both anesthetic gels were applied at separate sites before the injection of LA. The time of the onset of anesthesia was noted and analyzed. Patients were selected on the basis of inclusion and exclusion criteria. Individuals aged 18 to 30 years who were systemically healthy and orthodontically indicated for bilateral premolar extraction were included in this study. Again, patients with a history of neurological disorders and allergies to amitriptyline and lidocaine were excluded from the current study. Results Significant differences emerged between groups at five and 10 minutes, with amitriptyline-induced partial numbness (36.7% and 6.7%). At 40 and 45 minutes, both groups showed varied partial and complete numbness, with amitriptyline leading to partial recovery (23.3% and 73.3% complete numbness, 23.3% partial recovery) and lidocaine resulting in partial recovery (81.7%). When comparing the visual analog scale (VAS) scores, both groups exhibited a similar simultaneous effect at 15 minutes. Nonetheless, amitriptyline displayed significantly lower scores at 25 and 35 minutes (p < 0.001) in comparison to lidocaine. Similar observations were made when controlling for pain intensity. Conclusion It was concluded that amitriptyline holds both anesthetic and analgesic properties. Nevertheless, this study was unable to generalize the study findings because of the small sample size and being a single-center study. However, the VAS scores of anesthetic and analgesic pharmacodynamics properties of amitriptyline were statistically significantly lower than lidocaine, particularly at 25 and 35 minutes. Additionally, amitriptyline-induced anesthetic and analgesic pharmacology, especially pharmacokinetics properties, depends on the location and pattern of pain.
Collapse
Affiliation(s)
- Nirav Patel
- Department of Oral and Maxillofacial Surgery, Goenka Research Institute of Dental Science, Gandhinagar, IND
| | - Sarvesh B Urolagin
- Department of Oral and Maxillofacial Surgery, Subbaiah Institute of Dental Sciences, Shimoga, IND
| | - Md Ahsanul Haq
- Department of Biostatistics, Infectious Diseases Division, International Centre for Diarrhoeal Disease Research, Bangladesh (icddr,b), Dhaka, BGD
| | - Chhaya Patel
- Department of Pedodontics and Preventive Dentistry, Karnavati School of Dentistry, Karnavati University, Gandhinagar, IND
| | - Rohan Bhatt
- Department of Pediatric Dentistry, Karnavati School of Dentistry, Karnavati University, Gandhinagar, IND
| | - Gaurav Girdhar
- Department of Periodontology and Implantology, Karnavati University, Gandhinagar, IND
| | - Susmita Sinha
- Department of Physiology, Khulna City Medical College and Hospital, Khulna, BGD
| | - Mainul Haque
- Karnavati Scientific Research Center, Karnavati School of Dentistry, Karnavati University, Gandhinagar, IND
- Department of Pharmacology and Therapeutics, National Defence University of Malaysia, Kuala Lumpur, MYS
| | - Santosh Kumar
- Department of Periodontology and Implantology, Karnavati School of Dentistry, Karnavati University, Gandhinagar, IND
| |
Collapse
|
7
|
Stable expression of human Nav1.5 for high-throughput cardiac safety assessment. Mol Cell Toxicol 2023. [DOI: 10.1007/s13273-023-00331-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
|
8
|
Körner J, Albani S, Sudha Bhagavath Eswaran V, Roehl AB, Rossetti G, Lampert A. Sodium Channels and Local Anesthetics-Old Friends With New Perspectives. Front Pharmacol 2022; 13:837088. [PMID: 35418860 PMCID: PMC8996304 DOI: 10.3389/fphar.2022.837088] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2021] [Accepted: 02/16/2022] [Indexed: 12/11/2022] Open
Abstract
The long history of local anesthetics (LAs) starts out in the late 19th century when the content of coca plant leaves was discovered to alleviate pain. Soon after, cocaine was established and headed off to an infamous career as a substance causing addiction. Today, LAs and related substances-in modified form-are indispensable in our clinical everyday life for pain relief during and after minor and major surgery, and dental practices. In this review, we elucidate on the interaction of modern LAs with their main target, the voltage-gated sodium channel (Navs), in the light of the recently published channel structures. Knowledge of the 3D interaction sites of the drug with the protein will allow to mechanistically substantiate the comprehensive data available on LA gating modification. In the 1970s it was suggested that LAs can enter the channel pore from the lipid phase, which was quite prospective at that time. Today we know from cryo-electron microscopy structures and mutagenesis experiments, that indeed Navs have side fenestrations facing the membrane, which are likely the entrance for LAs to induce tonic block. In this review, we will focus on the effects of LA binding on fast inactivation and use-dependent inhibition in the light of the proposed new allosteric mechanism of fast inactivation. We will elaborate on subtype and species specificity and provide insights into modelling approaches that will help identify the exact molecular binding orientation, access pathways and pharmacokinetics. With this comprehensive overview, we will provide new perspectives in the use of the drug, both clinically and as a tool for basic ion channel research.
Collapse
Affiliation(s)
- Jannis Körner
- Institute of Physiology, Aachen, Germany.,Clinic of Anesthesiology, Medical Faculty, Uniklinik RWTH Aachen, Aachen, Germany
| | - Simone Albani
- Institute for Neuroscience and Medicine (INM-9/IAS-5), Forschungszentrum Jülich, Jülich, Germany.,Faculty of Mathematics, Computer Science and Natural Sciences, RWTH Aachen, Aachen, Germany
| | | | - Anna B Roehl
- Clinic of Anesthesiology, Medical Faculty, Uniklinik RWTH Aachen, Aachen, Germany
| | - Giulia Rossetti
- Institute for Neuroscience and Medicine (INM-9/IAS-5), Forschungszentrum Jülich, Jülich, Germany.,Jülich Supercomputing Center (JSC), Forschungszentrum Jülich, Aachen, Germany.,Department of Neurology, University Hospital Aachen, RWTH Aachen University, Aachen, Germany
| | | |
Collapse
|
9
|
Zhu W, Wang W, Angsutararux P, Mellor RL, Isom LL, Nerbonne JM, Silva JR. Modulation of the effects of class Ib antiarrhythmics on cardiac NaV1.5-encoded channels by accessory NaVβ subunits. JCI Insight 2021; 6:e143092. [PMID: 34156986 PMCID: PMC8410097 DOI: 10.1172/jci.insight.143092] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2020] [Accepted: 06/17/2021] [Indexed: 01/28/2023] Open
Abstract
Native myocardial voltage-gated sodium (NaV) channels function in macromolecular complexes comprising a pore-forming (α) subunit and multiple accessory proteins. Here, we investigated the impact of accessory NaVβ1 and NaVβ3 subunits on the functional effects of 2 well-known class Ib antiarrhythmics, lidocaine and ranolazine, on the predominant NaV channel α subunit, NaV1.5, expressed in the mammalian heart. We showed that both drugs stabilized the activated conformation of the voltage sensor of domain-III (DIII-VSD) in NaV1.5. In the presence of NaVβ1, the effect of lidocaine on the DIII-VSD was enhanced, whereas the effect of ranolazine was abolished. Mutating the main class Ib drug-binding site, F1760, affected but did not abolish the modulation of drug block by NaVβ1/β3. Recordings from adult mouse ventricular myocytes demonstrated that loss of Scn1b (NaVβ1) differentially affected the potencies of lidocaine and ranolazine. In vivo experiments revealed distinct ECG responses to i.p. injection of ranolazine or lidocaine in WT and Scn1b-null animals, suggesting that NaVβ1 modulated drug responses at the whole-heart level. In the human heart, we found that SCN1B transcript expression was 3 times higher in the atria than ventricles, differences that could, in combination with inherited or acquired cardiovascular disease, dramatically affect patient response to class Ib antiarrhythmic therapies.
Collapse
Affiliation(s)
- Wandi Zhu
- Department of Biomedical Engineering, McKelvey School of Engineering, Washington University in St. Louis, St. Louis, Missouri, USA.,Department of Medicine, Brigham and Women's Hospital, Boston, Massachusetts, USA
| | - Wei Wang
- Department of Internal Medicine, Washington University School of Medicine in St. Louis, St. Louis, Missouri, USA
| | - Paweorn Angsutararux
- Department of Biomedical Engineering, McKelvey School of Engineering, Washington University in St. Louis, St. Louis, Missouri, USA
| | - Rebecca L Mellor
- Department of Internal Medicine, Washington University School of Medicine in St. Louis, St. Louis, Missouri, USA
| | - Lori L Isom
- Department of Pharmacology, University of Michigan Medical School, Ann Arbor, Michigan, USA
| | - Jeanne M Nerbonne
- Department of Internal Medicine, Washington University School of Medicine in St. Louis, St. Louis, Missouri, USA.,Department of Developmental Biology, Washington University School of Medicine in St. Louis, St. Louis, Missouri, USA
| | - Jonathan R Silva
- Department of Biomedical Engineering, McKelvey School of Engineering, Washington University in St. Louis, St. Louis, Missouri, USA
| |
Collapse
|
10
|
Földi MC, Pesti K, Zboray K, Toth AV, Hegedűs T, Málnási-Csizmadia A, Lukacs P, Mike A. The mechanism of non-blocking inhibition of sodium channels revealed by conformation-selective photolabeling. Br J Pharmacol 2021; 178:1200-1217. [PMID: 33450052 DOI: 10.1111/bph.15365] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2020] [Revised: 12/10/2020] [Accepted: 01/03/2021] [Indexed: 11/30/2022] Open
Abstract
BACKGROUND AND PURPOSE Sodium channel inhibitors can be used to treat hyperexcitability-related diseases, including epilepsies, pain syndromes, neuromuscular disorders and cardiac arrhythmias. The applicability of these drugs is limited by their nonspecific effect on physiological function. They act mainly by sodium channel block and in addition by modulation of channel kinetics. While channel block inhibits healthy and pathological tissue equally, modulation can preferentially inhibit pathological activity. An ideal drug designed to target the sodium channels of pathological tissue would act predominantly by modulation. Thus far, no such drug has been described. EXPERIMENTAL APPROACH Patch-clamp experiments with ultra-fast solution exchange and photolabeling-coupled electrophysiology were applied to describe the unique mechanism of riluzole on Nav1.4 sodium channels. In silico docking experiments were used to study the molecular details of binding. KEY RESULTS We present evidence that riluzole acts predominantly by non-blocking modulation. We propose that, being a relatively small molecule, riluzole is able to stay bound to the binding site, but nonetheless stay off the conduction pathway, by residing in one of the fenestrations. We demonstrate how this mechanism can be recognized. CONCLUSIONS AND IMPLICATIONS Our results identify riluzole as the prototype of this new class of sodium channel inhibitors. Drugs of this class are expected to selectively prevent hyperexcitability, while having minimal effect on cells firing at a normal rate from a normal resting potential.
Collapse
Affiliation(s)
- Mátyás C Földi
- MTA-ELTE NAP B Opto-Neuropharmacology Group, Budapest, Hungary.,Plant Protection Institute, Centre for Agricultural Research, Martonvásár, Hungary
| | - Krisztina Pesti
- MTA-ELTE NAP B Opto-Neuropharmacology Group, Budapest, Hungary.,Department of Biochemistry, Eötvös Loránd University, Budapest, Hungary.,School of PhD Studies, Semmelweis University, Budapest, Hungary
| | - Katalin Zboray
- Plant Protection Institute, Centre for Agricultural Research, Martonvásár, Hungary
| | - Adam V Toth
- Plant Protection Institute, Centre for Agricultural Research, Martonvásár, Hungary
| | - Tamás Hegedűs
- Department of Biophysics and Radiation Biology, Semmelweis University, Budapest, Hungary
| | - András Málnási-Csizmadia
- Motor Pharmacology Research Group, Department of Biochemistry, Eötvös Loránd University, Budapest, Hungary
| | - Peter Lukacs
- MTA-ELTE NAP B Opto-Neuropharmacology Group, Budapest, Hungary.,Plant Protection Institute, Centre for Agricultural Research, Martonvásár, Hungary
| | - Arpad Mike
- MTA-ELTE NAP B Opto-Neuropharmacology Group, Budapest, Hungary.,Plant Protection Institute, Centre for Agricultural Research, Martonvásár, Hungary.,Department of Biochemistry, Eötvös Loránd University, Budapest, Hungary
| |
Collapse
|
11
|
Patyk-Kaźmierczak E, Kaźmierczak M. A new high-pressure benzocaine polymorph - towards understanding the molecular aggregation in crystals of an important active pharmaceutical ingredient (API). ACTA CRYSTALLOGRAPHICA SECTION B, STRUCTURAL SCIENCE, CRYSTAL ENGINEERING AND MATERIALS 2020; 76:56-64. [PMID: 32831241 DOI: 10.1107/s2052520619016548] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/20/2019] [Accepted: 12/08/2019] [Indexed: 06/11/2023]
Abstract
Benzocaine (BZC), an efficient and highly permeable anaesthetic and an active pharmaceutical ingredient of many commercially available drugs, was studied under high pressure up to 0.78 GPa. As a result, new BZC polymorph (IV) was discovered. The crystallization of polymorph (IV) can be initiated by heating crystals of polymorph (I) at a pressure of at least 0.45 GPa or by their compression to 0.60 GPa. However, no phase transition from polymorph (I) to (IV) was observed. Although polymorph (IV) exhibits the same main aggregation motif as in previously reported BZC polymorphs (I)-(III), i.e. a hydrogen-bonded ribbon, its molecular packing and hydrogen-bonding pattern differ considerably. The N-H...N hydrogen bonds joining parallel BZC ribbons in crystals at ambient pressure are eliminated in polymorph (IV), and BZC ribbons become positioned at an angle of about 80°. Unfortunately, crystals of polymorph (IV) were not preserved on pressure release, and depending on the decompression protocol they transformed into polymorph (II) or (I).
Collapse
Affiliation(s)
- Ewa Patyk-Kaźmierczak
- Faculty of Chemistry, Adam Mickiewicz University in Poznań, Uniwersytetu Poznańskiego 8, Poznań, 61-614, Poland
| | - Michał Kaźmierczak
- Faculty of Chemistry, Adam Mickiewicz University in Poznań, Uniwersytetu Poznańskiego 8, Poznań, 61-614, Poland
| |
Collapse
|
12
|
Zhu W, Mazzanti A, Voelker TL, Hou P, Moreno JD, Angsutararux P, Naegle KM, Priori SG, Silva JR. Predicting Patient Response to the Antiarrhythmic Mexiletine Based on Genetic Variation. Circ Res 2019; 124:539-552. [PMID: 30566038 DOI: 10.1161/circresaha.118.314050] [Citation(s) in RCA: 33] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
RATIONALE Mutations in the SCN5A gene, encoding the α subunit of the Nav1.5 channel, cause a life-threatening form of cardiac arrhythmia, long QT syndrome type 3 (LQT3). Mexiletine, which is structurally related to the Na+ channel-blocking anesthetic lidocaine, is used to treat LQT3 patients. However, the patient response is variable, depending on the genetic mutation in SCN5A. OBJECTIVE The goal of this study is to understand the molecular basis of patients' variable responses and build a predictive statistical model that can be used to personalize mexiletine treatment based on patient's genetic variant. METHODS AND RESULTS We monitored the cardiac Na+ channel voltage-sensing domain (VSD) conformational dynamics simultaneously with other gating properties for the LQT3 variants. To systematically identify the relationship between mexiletine block and channel biophysical properties, we used a system-based statistical modeling approach to connect the multivariate properties to patient phenotype. We found that mexiletine altered the conformation of the Domain III VSD, which is the same VSD that many tested LQT3 mutations affect. Analysis of 15 LQT3 variants showed a strong correlation between the activation of the Domain III-VSD and the strength of the inhibition of the channel by mexiletine. Based on this improved molecular-level understanding, we generated a systems-based model based on a dataset of 32 LQT3 patients, which then successfully predicted the response of 7 out of 8 patients to mexiletine in a blinded, retrospective trial. CONCLUSIONS Our results imply that the modulated receptor theory of local anesthetic action, which confines local anesthetic binding effects to the channel pore, should be revised to include drug interaction with the Domain III-VSD. Using an algorithm that incorporates this mode of action, we can predict patient-specific responses to mexiletine, improving therapeutic decision making.
Collapse
Affiliation(s)
- Wandi Zhu
- From the Department of Biomedical Engineering (W.Z., T.L.V., P.H., J.D.M., P.A., K.M.N., J.R.S.), Washington University in St Louis, MO
| | - Andrea Mazzanti
- Molecular Cardiology, IRCCS Salvatore Maugeri Foundation, Pavia, Italy (A.M., S.G.P.)
| | - Taylor L Voelker
- From the Department of Biomedical Engineering (W.Z., T.L.V., P.H., J.D.M., P.A., K.M.N., J.R.S.), Washington University in St Louis, MO
| | - Panpan Hou
- From the Department of Biomedical Engineering (W.Z., T.L.V., P.H., J.D.M., P.A., K.M.N., J.R.S.), Washington University in St Louis, MO
| | - Jonathan D Moreno
- From the Department of Biomedical Engineering (W.Z., T.L.V., P.H., J.D.M., P.A., K.M.N., J.R.S.), Washington University in St Louis, MO.,Division of Cardiology, Department of Medicine (J.D.M.), Washington University in St Louis, MO
| | - Paweorn Angsutararux
- From the Department of Biomedical Engineering (W.Z., T.L.V., P.H., J.D.M., P.A., K.M.N., J.R.S.), Washington University in St Louis, MO
| | - Kristen M Naegle
- From the Department of Biomedical Engineering (W.Z., T.L.V., P.H., J.D.M., P.A., K.M.N., J.R.S.), Washington University in St Louis, MO
| | - Silvia G Priori
- Molecular Cardiology, IRCCS Salvatore Maugeri Foundation, Pavia, Italy (A.M., S.G.P.).,Department of Molecular Medicine, University of Pavia, Italy (S.G.P.)
| | - Jonathan R Silva
- From the Department of Biomedical Engineering (W.Z., T.L.V., P.H., J.D.M., P.A., K.M.N., J.R.S.), Washington University in St Louis, MO
| |
Collapse
|
13
|
Elajnaf T, Baptista-Hon DT, Hales TG. Potent Inactivation-Dependent Inhibition of Adult and Neonatal NaV1.5 Channels by Lidocaine and Levobupivacaine. Anesth Analg 2019; 127:650-660. [PMID: 29958221 DOI: 10.1213/ane.0000000000003597] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
BACKGROUND Cardiotoxic effects of local anesthetics (LAs) involve inhibition of NaV1.5 voltage-gated Na channels. Metastatic breast and colon cancer cells also express NaV1.5, predominantly the neonatal splice variant (nNaV1.5) and their inhibition by LAs reduces invasion and migration. It may be advantageous to target cancer cells while sparing cardiac function through selective blockade of nNaV1.5 and/or by preferentially affecting inactivated NaV1.5, which predominate in cancer cells. We tested the hypotheses that lidocaine and levobupivacaine differentially affect (1) adult (aNaV1.5) and nNaV1.5 and (2) the resting and inactivated states of NaV1.5. METHODS The whole-cell voltage-clamp technique was used to evaluate the actions of lidocaine and levobupivacaine on recombinant NaV1.5 channels expressed in HEK-293 cells. Cells were transiently transfected with cDNAs encoding either aNaV1.5 or nNaV1.5. Voltage protocols were applied to determine depolarizing potentials that either activated or inactivated 50% of maximum conductance (V½ activation and V½ inactivation, respectively). RESULTS Lidocaine and levobupivacaine potently inhibited aNaV1.5 (IC50 mean [SD]: 20 [22] and 1 [0.6] μM, respectively) and nNaV1.5 (IC50 mean [SD]: 17 [10] and 3 [1.6] μM, respectively) at a holding potential of -80 mV. IC50s differed significantly between lidocaine and levobupivacaine with no influence of splice variant. Levobupivacaine induced a statistically significant depolarizing shift in the V½ activation for aNaV1.5 (mean [SD] from -32 [4.6] mV to -26 [8.1] mV) but had no effect on the voltage dependence of activation of nNaV1.5. Lidocaine had no effect on V½ activation of either variant but caused a significantly greater depression of maximum current mediated by nNaV1.5 compared to aNaV1.5. Similar statistically significant shifts in the V½ inactivation (approximately -10 mV) occurred for both LAs and NaV1.5 variants. Levobupivacaine (1 μM) caused a significantly greater slowing of recovery from inactivation of both variants than did lidocaine (10 μM). Both LAs caused approximately 50% tonic inhibition of aNaV1.5 or nNaV1.5 when holding at -80 mV. Neither LA caused tonic block at a holding potential of either -90 or -120 mV, voltages at which there was little steady-state inactivation. Higher concentrations of either lidocaine (300 μM) or levobupivacaine (100 μM) caused significantly more tonic block at -120 mV. CONCLUSIONS These data demonstrate that low concentrations of the LAs exhibit inactivation-dependent block of NaV1.5, which may provide a rationale for their use to safely inhibit migration and invasion by metastatic cancer cells without cardiotoxicity.
Collapse
Affiliation(s)
- Taha Elajnaf
- From The Institute of Academic Anaesthesia, Division of Neuroscience, School of Medicine, Ninewells Hospital, University of Dundee, Dundee, United Kingdom
| | | | | |
Collapse
|
14
|
Non-blocking modulation contributes to sodium channel inhibition by a covalently attached photoreactive riluzole analog. Sci Rep 2018; 8:8110. [PMID: 29802266 PMCID: PMC5970139 DOI: 10.1038/s41598-018-26444-y] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2017] [Accepted: 05/04/2018] [Indexed: 11/08/2022] Open
Abstract
Sodium channel inhibitor drugs decrease pathological hyperactivity in various diseases including pain syndromes, myotonia, arrhythmias, nerve injuries and epilepsies. Inhibiting pathological but not physiological activity, however, is a major challenge in drug development. Sodium channel inhibitors exert their effects by a dual action: they obstruct ion flow ("block"), and they alter the energetics of channel opening and closing ("modulation"). Ideal drugs would be modulators without blocking effect, because modulation is inherently activity-dependent, therefore selective for pathological hyperactivity. Can block and modulation be separated? It has been difficult to tell, because the effect of modulation is obscured by conformation-dependent association/dissociation of the drug. To eliminate dynamic association/dissociation, we used a photoreactive riluzole analog which could be covalently bound to the channel; and found, unexpectedly, that drug-bound channels could still conduct ions, although with modulated gating. The finding that non-blocking modulation is possible, may open a novel avenue for drug development because non-blocking modulators could be more specific in treating hyperactivity-linked diseases.
Collapse
|
15
|
Salvage SC, Chandrasekharan KH, Jeevaratnam K, Dulhunty AF, Thompson AJ, Jackson AP, Huang CL. Multiple targets for flecainide action: implications for cardiac arrhythmogenesis. Br J Pharmacol 2018; 175:1260-1278. [PMID: 28369767 PMCID: PMC5866987 DOI: 10.1111/bph.13807] [Citation(s) in RCA: 38] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2017] [Revised: 03/27/2017] [Accepted: 03/29/2017] [Indexed: 12/19/2022] Open
Abstract
Flecainide suppresses cardiac tachyarrhythmias including paroxysmal atrial fibrillation, supraventricular tachycardia and arrhythmic long QT syndromes (LQTS), as well as the Ca2+ -mediated, catecholaminergic polymorphic ventricular tachycardia (CPVT). However, flecainide can also exert pro-arrhythmic effects most notably following myocardial infarction and when used to diagnose Brugada syndrome (BrS). These divergent actions result from its physiological and pharmacological actions at multiple, interacting levels of cellular organization. These were studied in murine genetic models with modified Nav channel or intracellular ryanodine receptor (RyR2)-Ca2+ channel function. Flecainide accesses its transmembrane Nav 1.5 channel binding site during activated, open, states producing a use-dependent antagonism. Closing either activation or inactivation gates traps flecainide within the pore. An early peak INa related to activation of Nav channels followed by rapid de-activation, drives action potential (AP) upstrokes and their propagation. This is diminished in pro-arrhythmic conditions reflecting loss of function of Nav 1.5 channels, such as BrS, accordingly exacerbated by flecainide challenge. Contrastingly, pro-arrhythmic effects attributed to prolonged AP recovery by abnormal late INaL following gain-of-function modifications of Nav 1.5 channels in LQTS3 are reduced by flecainide. Anti-arrhythmic effects of flecainide that reduce triggering in CPVT models mediated by sarcoplasmic reticular Ca2+ release could arise from its primary actions on Nav channels indirectly decreasing [Ca2+ ]i through a reduced [Na+ ]i and/or direct open-state RyR2-Ca2+ channel antagonism. The consequent [Ca2+ ]i alterations could also modify AP propagation velocity and therefore arrhythmic substrate through its actions on Nav 1.5 channel function. This is consistent with the paradoxical differences between flecainide actions upon Na+ currents, AP conduction and arrhythmogenesis under circumstances of normal and increased RyR2 function. LINKED ARTICLES This article is part of a themed section on Spotlight on Small Molecules in Cardiovascular Diseases. To view the other articles in this section visit http://onlinelibrary.wiley.com/doi/10.1111/bph.v175.8/issuetoc.
Collapse
Affiliation(s)
- Samantha C Salvage
- Department of BiochemistryUniversity of CambridgeCambridgeUK
- Physiological LaboratoryUniversity of CambridgeCambridgeUK
| | | | - Kamalan Jeevaratnam
- Faculty of Health and Medical SciencesUniversity of SurreyGuildfordUK
- School of MedicinePerdana University – Royal College of Surgeons IrelandSerdangSelangor Darul EhsanMalaysia
| | - Angela F Dulhunty
- Muscle Research Group, Eccles Institute of Neuroscience, John Curtin School of Medical ResearchAustralian National UniversityActonAustralia
| | | | | | - Christopher L‐H Huang
- Department of BiochemistryUniversity of CambridgeCambridgeUK
- Physiological LaboratoryUniversity of CambridgeCambridgeUK
| |
Collapse
|
16
|
Ahmed M, Jalily Hasani H, Ganesan A, Houghton M, Barakat K. Modeling the human Na v1.5 sodium channel: structural and mechanistic insights of ion permeation and drug blockade. Drug Des Devel Ther 2017; 11:2301-2324. [PMID: 28831242 PMCID: PMC5552146 DOI: 10.2147/dddt.s133944] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Abnormalities in the human Nav1.5 (hNav1.5) voltage-gated sodium ion channel (VGSC) are associated with a wide range of cardiac problems and diseases in humans. Current structural models of hNav1.5 are still far from complete and, consequently, their ability to study atomistic interactions of this channel is very limited. Here, we report a comprehensive atomistic model of the hNav1.5 ion channel, constructed using homology modeling technique and refined through long molecular dynamics simulations (680 ns) in the lipid membrane bilayer. Our model was comprehensively validated by using reported mutagenesis data, comparisons with previous models, and binding to a panel of known hNav1.5 blockers. The relatively long classical MD simulation was sufficient to observe a natural sodium permeation event across the channel's selectivity filters to reach the channel's central cavity, together with the identification of a unique role of the lysine residue. Electrostatic potential calculations revealed the existence of two potential binding sites for the sodium ion at the outer selectivity filters. To obtain further mechanistic insight into the permeation event from the central cavity to the intracellular region of the channel, we further employed "state-of-the-art" steered molecular dynamics (SMD) simulations. Our SMD simulations revealed two different pathways through which a sodium ion can be expelled from the channel. Further, the SMD simulations identified the key residues that are likely to control these processes. Finally, we discuss the potential binding modes of a panel of known hNav1.5 blockers to our structural model of hNav1.5. We believe that the data presented here will enhance our understanding of the structure-property relationships of the hNav1.5 ion channel and the underlying molecular mechanisms in sodium ion permeation and drug interactions. The results presented here could be useful for designing safer drugs that do not block the hNav1.5 channel.
Collapse
Affiliation(s)
| | | | | | - Michael Houghton
- Li Ka Shing Institute of Virology
- Li Ka Shing Applied Virology Institute
- Department of Medical Microbiology and Immunology, Katz Centre for Health Research, University of Alberta, Edmonton, AB, Canada
| | - Khaled Barakat
- Faculty of Pharmacy and Pharmaceutical Sciences
- Li Ka Shing Institute of Virology
- Li Ka Shing Applied Virology Institute
| |
Collapse
|
17
|
Jang YJ, Lee JH, Seo TB, Oh SH. Lidocaine/multivalent ion complex as a potential strategy for prolonged local anesthesia. Eur J Pharm Biopharm 2017; 115:113-121. [DOI: 10.1016/j.ejpb.2017.02.007] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2016] [Revised: 01/31/2017] [Accepted: 02/13/2017] [Indexed: 01/17/2023]
|
18
|
Föhr KJ, Knippschild U, Herkommer A, Fauler M, Peifer C, Georgieff M, Adolph O. State-dependent block of voltage-gated sodium channels by the casein-kinase 1 inhibitor IC261. Invest New Drugs 2017; 35:277-289. [PMID: 28164251 DOI: 10.1007/s10637-017-0429-0] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2016] [Accepted: 01/12/2017] [Indexed: 12/13/2022]
Abstract
Background and Purpose IC261 (3-[(2,4,6-trimethoxyphenyl)methylidenyl]-indolin-2-one) has previously been introduced as an isoform specific inhibitor of casein kinase 1 (CK1) causing cell cycle arrest or cell death of established tumor cell lines. However, it is reasonable to assume that not all antitumor activities of IC261 are mediated by the inhibition of CK1. Meanwhile there is growing evidence that functional voltage-gated sodium channels are also implicated in the progression of tumors as their blockage suppresses tumor migration and invasion of different tumor cell lines. Thus, we asked whether IC261 functionally inhibits voltage-gated sodium channels. Experimental Approach Electrophysiological experiments were performed using the patch-clamp technique at human heart muscle sodium channels heterologously expressed in human TsA cells. Key Results IC261 inhibits sodium channels in a state-dependent manner. IC261 does not interact with the open channel and has only a low affinity for the resting state of the hNav1.5 (human voltage-gated sodium channel; Kr: 120 μM). The efficacy of IC261 strongly increases with membrane depolarisation, indicating that the inactivated state is an important target. The results of different experimental approaches finally revealed an affinity of IC261 to the inactivated state between 1 and 2 μM. Conclusion and Implications IC261 inhibits sodium channels at a similar concentration necessary to reduce CK1δ/ε activity by 50% (IC50 value 1 μM). Thus, inhibition of sodium channels might contribute to the antitumor activity of IC261.
Collapse
Affiliation(s)
- Karl J Föhr
- Department of Anesthesiology, University Hospital of Ulm, Albert-Einstein-Allee 23, D-89075, Ulm, Germany.
| | - Uwe Knippschild
- Department of General and Visceral Surgery, University Hospital of Ulm, Albert-Einstein-Allee 23, D-89081, Ulm, Germany
| | - Anna Herkommer
- Department of Anesthesiology, University Hospital of Ulm, Albert-Einstein-Allee 23, D-89081, Ulm, Germany
| | - Michael Fauler
- Institute of General Physiology, University of Ulm, Albert-Einstein-Allee 11, D-89081, Ulm, Germany
| | - Christian Peifer
- Institute of Pharmacy, University of Kiel, Gutenbergstr. 76, D-24118, Kiel, Germany
| | - Michael Georgieff
- Department of Anesthesiology, University Hospital of Ulm, Albert-Einstein-Allee 23, D-89075, Ulm, Germany
| | - Oliver Adolph
- Department of Anesthesiology, University Hospital of Ulm, Albert-Einstein-Allee 23, D-89075, Ulm, Germany
| |
Collapse
|
19
|
Bohnen MS, Peng G, Robey SH, Terrenoire C, Iyer V, Sampson KJ, Kass RS. Molecular Pathophysiology of Congenital Long QT Syndrome. Physiol Rev 2017; 97:89-134. [PMID: 27807201 PMCID: PMC5539372 DOI: 10.1152/physrev.00008.2016] [Citation(s) in RCA: 114] [Impact Index Per Article: 16.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
Ion channels represent the molecular entities that give rise to the cardiac action potential, the fundamental cellular electrical event in the heart. The concerted function of these channels leads to normal cyclical excitation and resultant contraction of cardiac muscle. Research into cardiac ion channel regulation and mutations that underlie disease pathogenesis has greatly enhanced our knowledge of the causes and clinical management of cardiac arrhythmia. Here we review the molecular determinants, pathogenesis, and pharmacology of congenital Long QT Syndrome. We examine mechanisms of dysfunction associated with three critical cardiac currents that comprise the majority of congenital Long QT Syndrome cases: 1) IKs, the slow delayed rectifier current; 2) IKr, the rapid delayed rectifier current; and 3) INa, the voltage-dependent sodium current. Less common subtypes of congenital Long QT Syndrome affect other cardiac ionic currents that contribute to the dynamic nature of cardiac electrophysiology. Through the study of mutations that cause congenital Long QT Syndrome, the scientific community has advanced understanding of ion channel structure-function relationships, physiology, and pharmacological response to clinically employed and experimental pharmacological agents. Our understanding of congenital Long QT Syndrome continues to evolve rapidly and with great benefits: genotype-driven clinical management of the disease has improved patient care as precision medicine becomes even more a reality.
Collapse
Affiliation(s)
- M S Bohnen
- Department of Pharmacology, Columbia University Medical Center, New York, New York; and The New York Stem Cell Foundation Research Institute, New York, New York
| | - G Peng
- Department of Pharmacology, Columbia University Medical Center, New York, New York; and The New York Stem Cell Foundation Research Institute, New York, New York
| | - S H Robey
- Department of Pharmacology, Columbia University Medical Center, New York, New York; and The New York Stem Cell Foundation Research Institute, New York, New York
| | - C Terrenoire
- Department of Pharmacology, Columbia University Medical Center, New York, New York; and The New York Stem Cell Foundation Research Institute, New York, New York
| | - V Iyer
- Department of Pharmacology, Columbia University Medical Center, New York, New York; and The New York Stem Cell Foundation Research Institute, New York, New York
| | - K J Sampson
- Department of Pharmacology, Columbia University Medical Center, New York, New York; and The New York Stem Cell Foundation Research Institute, New York, New York
| | - R S Kass
- Department of Pharmacology, Columbia University Medical Center, New York, New York; and The New York Stem Cell Foundation Research Institute, New York, New York
| |
Collapse
|
20
|
Sheets MF, Fozzard HA, Hanck DA. Important Role of Asparagines in Coupling the Pore and Votage-Sensor Domain in Voltage-Gated Sodium Channels. Biophys J 2016; 109:2277-86. [PMID: 26636939 DOI: 10.1016/j.bpj.2015.10.012] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2015] [Revised: 09/24/2015] [Accepted: 10/08/2015] [Indexed: 11/26/2022] Open
Abstract
Voltage-gated sodium (NaV) channels contain an α-subunit incorporating the channel's pore and gating machinery composed of four homologous domains (DI-DIV), with a pore domain formed by the S5 and S6 segments and a voltage-sensor domain formed by the S1-S4 segments. During a membrane depolarization movement, the S4s in the voltage-sensor domains exert downstream effects on the S6 segments to control ionic conductance through the pore domain. We used lidocaine, a local anesthetic and antiarrhythmic drug, to probe the role of conserved Asn residues in the S6s of DIII and DIV in NaV1.5 and NaV1.4. Previous studies have shown that lidocaine binding to the pore domain causes a decrease in the maximum gating (Qmax) charge of ∼38%, and three-fourths of this decrease results from the complete stabilization of DIII-S4 (contributing a 30% reduction in Qmax) and one-fourth is due to partial stabilization of DIV-S4 (a reduction of 8-10%). Even though substitutions for the Asn in DIV-S6 in NaV1.5, N1764A and N1764C, produce little ionic current in transfected mammalian cells, they both express robust gating currents. Anthopleurin-A toxin, which inhibits movement of DIV-S4, still reduced Qmax by nearly 30%, a value similar to that observed in wild-type channels, in both N1764A and N1764C. By applying lidocaine and measuring the gating currents, we demonstrated that Asn residues in the S6s of DIII and DIV are important for coupling their pore domains to their voltage-sensor domains, and that Ala and Cys substitutions for Asn in both S6s result in uncoupling of the pore domains from their voltage-sensor domains. Similar observations were made for NaV1.4, although substitutions for Asn in DIII-S6 showed somewhat less uncoupling.
Collapse
Affiliation(s)
- Michael F Sheets
- Nora Eccles Harrison Cardiovascular Research and Training Institute (CVRTI), University of Utah, Salt Lake City, Utah.
| | | | | |
Collapse
|
21
|
Gingrich KJ, Wagner LE. Fast-onset lidocaine block of rat Na V1.4 channels suggests involvement of a second high-affinity open state. BIOCHIMICA ET BIOPHYSICA ACTA-BIOMEMBRANES 2016; 1858:1175-88. [DOI: 10.1016/j.bbamem.2016.02.033] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/23/2015] [Revised: 02/04/2016] [Accepted: 02/24/2016] [Indexed: 11/25/2022]
|
22
|
Testa B, Vistoli G, Pedretti A. Mechanisms and pharmaceutical consequences of processes of stereoisomerisation — A didactic excursion. Eur J Pharm Sci 2016; 88:101-23. [DOI: 10.1016/j.ejps.2016.04.007] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2016] [Revised: 04/02/2016] [Accepted: 04/05/2016] [Indexed: 11/29/2022]
|
23
|
Smith NE, Corry B. Mutant bacterial sodium channels as models for local anesthetic block of eukaryotic proteins. Channels (Austin) 2016; 10:225-37. [PMID: 26852716 DOI: 10.1080/19336950.2016.1148224] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Voltage gated sodium channels are the target of a range of local anesthetic, anti-epileptic and anti-arrhythmic compounds. But, gaining a molecular level understanding of their mode of action is difficult as we only have atomic resolution structures of bacterial sodium channels not their eukaryotic counterparts. In this study we used molecular dynamics simulations to demonstrate that the binding sites of both the local anesthetic benzocaine and the anti-epileptic phenytoin to the bacterial sodium channel NavAb can be altered significantly by the introduction of point mutations. Free energy techniques were applied to show that increased aromaticity in the pore of the channel, used to emulate the aromatic residues observed in eukaryotic Nav1.2, led to changes in the location of binding and dissociation constants of each drug relative to wild type NavAb. Further, binding locations and dissociation constants obtained for both benzocaine (660 μM) and phenytoin (1 μM) in the mutant channels were within the range expected from experimental values obtained from drug binding to eukaryotic sodium channels, indicating that these mutant NavAb may be a better model for drug binding to eukaryotic channels than the wild type.
Collapse
Affiliation(s)
- Natalie E Smith
- a Research School of Biology, Australian National University , Canberra , ACT , Australia
| | - Ben Corry
- a Research School of Biology, Australian National University , Canberra , ACT , Australia
| |
Collapse
|
24
|
Moczydlowski E. On the Natural and Unnatural History of the Voltage-Gated Na+ Channel. NA CHANNELS FROM PHYLA TO FUNCTION 2016; 78:3-36. [DOI: 10.1016/bs.ctm.2016.06.002] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/23/2023]
|
25
|
Zhou Y, Lingle CJ. Paxilline inhibits BK channels by an almost exclusively closed-channel block mechanism. ACTA ACUST UNITED AC 2015; 144:415-40. [PMID: 25348413 PMCID: PMC4210426 DOI: 10.1085/jgp.201411259] [Citation(s) in RCA: 105] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Abstract
Paxilline, a tremorogenic fungal alkaloid, potently inhibits large conductance Ca(2+)- and voltage-activated K(+) (BK)-type channels, but little is known about the mechanism underlying this inhibition. Here we show that inhibition is inversely dependent on BK channel open probability (Po), and is fully relieved by conditions that increase Po, even in the constant presence of paxilline. Manipulations that shift BK gating to more negative potentials reduce inhibition by paxilline in accordance with the increase in channel Po. Measurements of Po times the number of channels at negative potentials support the idea that paxilline increases occupancy of closed states, effectively reducing the closed-open equilibrium constant, L(0). Gating current measurements exclude an effect of paxilline on voltage sensors. Steady-state inhibition by multiple paxilline concentrations was determined for four distinct equilibration conditions, each with a distinct Po. The IC50 for paxilline shifted from around 10 nM when channels were largely closed to near 10 µM as maximal Po was approached. Model-dependent analysis suggests a mechanism of inhibition in which binding of a single paxilline molecule allosterically alters the intrinsic L(0) favoring occupancy of closed states, with affinity for the closed conformation being >500-fold greater than affinity for the open conformation. The rate of inhibition of closed channels was linear up through 2 µM paxilline, with a slope of 2 × 10(6) M(-1)s(-1). Paxilline inhibition was hindered by either the bulky cytosolic blocker, bbTBA, or by concentrations of cytosolic sucrose that hinder ion permeation. However, paxilline does not hinder MTSET modification of the inner cavity residue, A313C. We conclude that paxilline binds more tightly to the closed conformation, favoring occupancy of closed-channel conformations, and propose that it binds to a superficial position near the entrance to the central cavity, but does not hinder access of smaller molecules to this cavity.
Collapse
Affiliation(s)
- Yu Zhou
- Department of Anesthesiology, Washington University School of Medicine, St. Louis, MO 63110
| | - Christopher J Lingle
- Department of Anesthesiology, Washington University School of Medicine, St. Louis, MO 63110
| |
Collapse
|
26
|
Voltage-gated Na+ Channel Activity Increases Colon Cancer Transcriptional Activity and Invasion Via Persistent MAPK Signaling. Sci Rep 2015; 5:11541. [PMID: 26096612 PMCID: PMC4476109 DOI: 10.1038/srep11541] [Citation(s) in RCA: 63] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2015] [Accepted: 05/22/2015] [Indexed: 12/27/2022] Open
Abstract
Functional expression of voltage-gated Na+ channels (VGSCs) has been demonstrated in multiple cancer cell types where channel activity induces invasive activity. The signaling mechanisms by which VGSCs promote oncogenesis remain poorly understood. We explored the signal transduction process critical to VGSC-mediated invasion on the basis of reports linking channel activity to gene expression changes in excitable cells. Coincidentally, many genes transcriptionally regulated by the SCN5A isoform in colon cancer have an over-representation of cis-acting sites for transcription factors phosphorylated by ERK1/2 MAPK. We hypothesized that VGSC activity promotes MAPK activation to induce transcriptional changes in invasion-related genes. Using pharmacological inhibitors/activators and siRNA-mediated gene knockdowns, we correlated channel activity with Rap1-dependent persistent MAPK activation in the SW620 human colon cancer cell line. We further demonstrated that VGSC activity induces downstream changes in invasion-related gene expression via a PKA/ERK/c-JUN/ELK-1/ETS-1 transcriptional pathway. This is the first study illustrating a molecular mechanism linking functional activity of VGSCs to transcriptional activation of invasion-related genes.
Collapse
|
27
|
Wang Y, Mi J, Lu K, Lu Y, Wang K. Comparison of Gating Properties and Use-Dependent Block of Nav1.5 and Nav1.7 Channels by Anti-Arrhythmics Mexiletine and Lidocaine. PLoS One 2015; 10:e0128653. [PMID: 26068619 PMCID: PMC4465899 DOI: 10.1371/journal.pone.0128653] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2015] [Accepted: 04/29/2015] [Indexed: 12/19/2022] Open
Abstract
Mexiletine and lidocaine are widely used class IB anti-arrhythmic drugs that are considered to act by blocking voltage-gated open sodium currents for treatment of ventricular arrhythmias and relief of pain. To gain mechanistic insights into action of anti-arrhythmics, we characterized biophysical properties of Nav1.5 and Nav1.7 channels stably expressed in HEK293 cells and compared their use-dependent block in response to mexiletine and lidocaine using whole-cell patch clamp recordings. While the voltage-dependent activation of Nav1.5 or Nav1.7 was not affected by mexiletine and lidocaine, the steady-state fast and slow inactivation of Nav1.5 and Nav1.7 were significantly shifted to hyperpolarized direction by either mexiletine or lidocaine in dose-dependent manner. Both mexiletine and lidocaine enhanced the slow component of closed-state inactivation, with mexiletine exerting stronger inhibition on either Nav1.5 or Nav1.7. The recovery from inactivation of Nav1.5 or Nav1.7 was significantly prolonged by mexiletine compared to lidocaine. Furthermore, mexiletine displayed a pronounced and prominent use-dependent inhibition of Nav1.5 than lidocaine, but not Nav1.7 channels. Taken together, our findings demonstrate differential responses to blockade by mexiletine and lidocaine that preferentially affect the gating of Nav1.5, as compared to Nav1.7; and mexiletine exhibits stronger use-dependent block of Nav1.5. The differential gating properties of Nav1.5 and Nav1.7 in response to mexiletine and lidocaine may help explain the drug effectiveness and advance in new designs of safe and specific sodium channel blockers for treatment of cardiac arrhythmia or pain.
Collapse
Affiliation(s)
- Ying Wang
- Biomedical Research Institute, Shenzhen Peking University-Hong Kong University of Science and Technology Medical Center, Shenzhen 518036, China
| | - Jianxun Mi
- Key Laboratory of Computational Intelligence, College of Computer Science and Technology, Chongqing University of Posts and Telecommunications, Chongqing 400065, China
| | - Ka Lu
- Department of Molecular and Cellular Pharmacology, State Key Laboratory of Natural and Biomimetic Drugs, Peking University School of Pharmaceutical Sciences, Beijing 100191, China
| | - Yanxin Lu
- Biomedical Research Institute, Shenzhen Peking University-Hong Kong University of Science and Technology Medical Center, Shenzhen 518036, China
- Department of Molecular and Cellular Pharmacology, State Key Laboratory of Natural and Biomimetic Drugs, Peking University School of Pharmaceutical Sciences, Beijing 100191, China
| | - KeWei Wang
- Biomedical Research Institute, Shenzhen Peking University-Hong Kong University of Science and Technology Medical Center, Shenzhen 518036, China
- Department of Molecular and Cellular Pharmacology, State Key Laboratory of Natural and Biomimetic Drugs, Peking University School of Pharmaceutical Sciences, Beijing 100191, China
- Department of Pharmacology, Qingdao University School of Pharmacy, Qingdao 266021, China
- * E-mail:
| |
Collapse
|
28
|
Kamp TJ, January CT. Harry A. Fozzard, MD: 1931–2014. Circ Res 2015; 116:552-3. [PMID: 25821880 DOI: 10.1161/circresaha.115.306038] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Affiliation(s)
- Timothy J Kamp
- Division of Cardiovascular Medicine, 8459 WIMR2, 1111 Highland Ave, University of Wisconsin-Madison, Madison, WI 53705. E-mail
| | | |
Collapse
|
29
|
Yoshida K, Takashima A, Nishio I. Effect of dibucaine hydrochloride on raft-like lipid domains in model membrane systems. MEDCHEMCOMM 2015. [DOI: 10.1039/c5md00108k] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
To clarify the biophysical and/or physicochemical mechanism of anaesthesia, we investigated the influence of dibucaine hydrochloride (DC·HCl), a local anaesthetic, on raft-like domains in ternary liposomes composed of dioleoylphosphatidylcholine (DOPC), dipalmitoylphosphatidylcholine (DPPC) and cholesterol (Chol).
Collapse
Affiliation(s)
- Kazunari Yoshida
- New Industry Creation Hatchery Center
- Tohoku University
- 6-6-10 Aoba
- Aoba-ku
- Japan
| | - Akito Takashima
- Department of Physics and Mathematics
- College of Science and Engineering
- Aoyama Gakuin University
- 5-10-1 Fuchinobe
- Sagamihara
| | - Izumi Nishio
- Department of Physics and Mathematics
- College of Science and Engineering
- Aoyama Gakuin University
- 5-10-1 Fuchinobe
- Sagamihara
| |
Collapse
|
30
|
Zhang H, Ji H, Liu Z, Ji Y, You X, Ding G, Cheng Z. Voltage-dependent blockade by bupivacaine of cardiac sodium channels expressed in Xenopus oocytes. Neurosci Bull 2014; 30:697-710. [PMID: 25008571 DOI: 10.1007/s12264-013-1449-1] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2013] [Accepted: 03/24/2014] [Indexed: 11/29/2022] Open
Abstract
Bupivacaine ranks as the most potent and efficient drug among class I local anesthetics, but its high potential for toxic reactions severely limits its clinical use. Although bupivacaine-induced toxicity is mainly caused by substantial blockade of voltage-gated sodium channels (VGSCs), how these hydrophobic molecules interact with the receptor sites to which they bind remains unclear. Nav1.5 is the dominant isoform of VGSCs expressed in cardiac myocytes, and its dysfunction may be the cause of bupivacaine-triggered arrhythmia. Here, we investigated the effect of bupivacaine on Nav1.5 within the clinical concentration range. The electrophysiological measurements on Nav1.5 expressed in Xenopus oocytes showed that bupivacaine induced a voltage- and concentration-dependent blockade on the peak of I Na and the half-maximal inhibitory dose was 4.51 μmol/L. Consistent with other local anesthetics, bupivacaine also induced a use-dependent blockade on Nav1.5 currents. The underlying mechanisms of this blockade may contribute to the fact that bupivacaine not only dose-dependently affected the gating kinetics of Nav1.5 but also accelerated the development of its open-state slow inactivation. These results extend our knowledge of the action of bupivacaine on cardiac sodium channels, and therefore contribute to the safer and more efficient clinical use of bupivacaine.
Collapse
Affiliation(s)
- Heng Zhang
- Lab of Neuropharmacology and Neurotoxicology, Shanghai University, Shanghai, 200444, China
| | | | | | | | | | | | | |
Collapse
|
31
|
Ranolazine inhibition of hERG potassium channels: drug-pore interactions and reduced potency against inactivation mutants. J Mol Cell Cardiol 2014; 74:220-30. [PMID: 24877995 PMCID: PMC4121676 DOI: 10.1016/j.yjmcc.2014.05.013] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/17/2014] [Revised: 05/14/2014] [Accepted: 05/19/2014] [Indexed: 01/06/2023]
Abstract
The antianginal drug ranolazine, which combines inhibitory actions on rapid and sustained sodium currents with inhibition of the hERG/IKr potassium channel, shows promise as an antiarrhythmic agent. This study investigated the structural basis of hERG block by ranolazine, with lidocaine used as a low potency, structurally similar comparator. Recordings of hERG current (IhERG) were made from cell lines expressing wild-type (WT) or mutant hERG channels. Docking simulations were performed using homology models built on MthK and KvAP templates. In conventional voltage clamp, ranolazine inhibited IhERG with an IC50 of 8.03 μM; peak IhERG during ventricular action potential clamp was inhibited ~ 62% at 10 μM. The IC50 values for ranolazine inhibition of the S620T inactivation deficient and N588K attenuated inactivation mutants were respectively ~ 73-fold and ~ 15-fold that for WT IhERG. Mutations near the bottom of the selectivity filter (V625A, S624A, T623A) exhibited IC50s between ~ 8 and 19-fold that for WT IhERG, whilst the Y652A and F656A S6 mutations had IC50s ~ 22-fold and 53-fold WT controls. Low potency lidocaine was comparatively insensitive to both pore helix and S6 mutations, but was sensitive to direction of K+ flux and particularly to loss of inactivation, with an IC50 for S620T-hERG ~ 49-fold that for WT IhERG. Docking simulations indicated that the larger size of ranolazine gives it potential for a greater range of interactions with hERG pore side chains compared to lidocaine, in particular enabling interaction of its two aromatic groups with side chains of both Y652 and F656. The N588K mutation is responsible for the SQT1 variant of short QT syndrome and our data suggest that ranolazine is unlikely to be effective against IKr/hERG in SQT1 patients. hERG K+ channels regulate cardiac action potential repolarization. The molecular basis of hERG block by ranolazine and structurally related lidocaine was studied. S6 Y652A and F656A mutations affected greatly ranolazine but not lidocaine binding. T623 and S624 residues may directly interact with ranolazine but not lidocaine. N588K and S620T attenuated inactivation mutants had reduced sensitivity to both drugs.
Collapse
|
32
|
Prokaryotic NavMs channel as a structural and functional model for eukaryotic sodium channel antagonism. Proc Natl Acad Sci U S A 2014; 111:8428-33. [PMID: 24850863 DOI: 10.1073/pnas.1406855111] [Citation(s) in RCA: 100] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Voltage-gated sodium channels are important targets for the development of pharmaceutical drugs, because mutations in different human sodium channel isoforms have causal relationships with a range of neurological and cardiovascular diseases. In this study, functional electrophysiological studies show that the prokaryotic sodium channel from Magnetococcus marinus (NavMs) binds and is inhibited by eukaryotic sodium channel blockers in a manner similar to the human Nav1.1 channel, despite millions of years of divergent evolution between the two types of channels. Crystal complexes of the NavMs pore with several brominated blocker compounds depict a common antagonist binding site in the cavity, adjacent to lipid-facing fenestrations proposed to be the portals for drug entry. In silico docking studies indicate the full extent of the blocker binding site, and electrophysiology studies of NavMs channels with mutations at adjacent residues validate the location. These results suggest that the NavMs channel can be a valuable tool for screening and rational design of human drugs.
Collapse
|
33
|
Aguado E, León I, Millán J, Cocinero EJ, Jaeqx S, Rijs AM, Lesarri A, Fernández JA. Unraveling the Benzocaine–Receptor Interaction at Molecular Level Using Mass-Resolved Spectroscopy. J Phys Chem B 2013; 117:13472-80. [DOI: 10.1021/jp4068944] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Affiliation(s)
- Edurne Aguado
- Departamento de Química
Física, Facultad de Ciencia
y Tecnología, Universidad del País Vasco (UPV/EHU), B°
Sarriena s/n, 48940 Leioa, Spain
| | - Iker León
- Departamento de Química
Física, Facultad de Ciencia
y Tecnología, Universidad del País Vasco (UPV/EHU), B°
Sarriena s/n, 48940 Leioa, Spain
| | - Judith Millán
- Departamento
de Química, Facultad de
Ciencias, Estudios Agroalimentarios
e Informática, Universidad de La Rioja, Madre de Dios,
51, 26006 Logroño, Spain
| | - Emilio J. Cocinero
- Departamento de Química
Física, Facultad de Ciencia
y Tecnología, Universidad del País Vasco (UPV/EHU), B°
Sarriena s/n, 48940 Leioa, Spain
| | - Sander Jaeqx
- Radboud
University Nijmegen, Institute for Molecules and Materials, FELIX Facility, Toernooiveld 7, 6525 ED Nijmegen, The Netherlands
| | - Anouk M. Rijs
- Radboud
University Nijmegen, Institute for Molecules and Materials, FELIX Facility, Toernooiveld 7, 6525 ED Nijmegen, The Netherlands
| | - Alberto Lesarri
- Departamento de Química Física y Química
Inorgánica, Facultad de
Ciencias, Universidad de Valladolid, E-47011 Valladolid, Spain
| | - José A. Fernández
- Departamento de Química
Física, Facultad de Ciencia
y Tecnología, Universidad del País Vasco (UPV/EHU), B°
Sarriena s/n, 48940 Leioa, Spain
| |
Collapse
|
34
|
Abstract
Recent structural breakthroughs with the voltage-gated sodium channel from Arcobacter butzleri suggest that such bacterial channels may provide a structural platform to advance the understanding of eukaryotic sodium channel gating and pharmacology. We therefore set out to determine whether compounds known to interact with eukaryotic NaVs could also inhibit the bacterial channel from Bacillus halodurans and NaChBac and whether they did so through similar mechanisms as in their eukaryotic homologues. The data show that the archetypal local anesthetic (LA) lidocaine inhibits resting NaChBac channels with a dissociation constant (Kd) of 260 µM, and channels displayed a left-shifted steady-state inactivation gating relationship in the presence of the drug. Extracellular application of QX-314 to expressed NaChBac channels had no effect on sodium current, whereas internal exposure via injection of a bolus of the quaternary derivative rapidly reduced sodium conductance, consistent with a hydrophilic cytoplasmic access pathway to an internal binding site. However, the neutral derivative benzocaine applied externally inhibited NaChBac channels, suggesting that hydrophobic pathways can also provide drug access to inhibit channels. Alternatively, ranolazine, a putative preopen state blocker of eukaryotic NaVs, displayed a Kd of 60 µM and left-shifted the NaChBac activation-voltage relationship. In each case, block enhanced entry into the inactivated state of the channel, an effect that is well described by a simple kinetic scheme. The data suggest that although significant differences exist, LA block of eukaryotic NaVs also occurs in bacterial sodium channels and that NaChBac shares pharmacological homology to the resting state of vertebrate NaV homologues.
Collapse
Affiliation(s)
- Sora Lee
- Department of Anesthesiology, Pharmacology, and Therapeutics, University of British Columbia, Vancouver, British Columbia V6T 1Z3, Canada
| | | | | |
Collapse
|
35
|
Huang CJ, Schild L, Moczydlowski EG. Use-dependent block of the voltage-gated Na(+) channel by tetrodotoxin and saxitoxin: effect of pore mutations that change ionic selectivity. ACTA ACUST UNITED AC 2013; 140:435-54. [PMID: 23008436 PMCID: PMC3457692 DOI: 10.1085/jgp.201210853] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Voltage-gated Na(+) channels (NaV channels) are specifically blocked by guanidinium toxins such as tetrodotoxin (TTX) and saxitoxin (STX) with nanomolar to micromolar affinity depending on key amino acid substitutions in the outer vestibule of the channel that vary with NaV gene isoforms. All NaV channels that have been studied exhibit a use-dependent enhancement of TTX/STX affinity when the channel is stimulated with brief repetitive voltage depolarizations from a hyperpolarized starting voltage. Two models have been proposed to explain the mechanism of TTX/STX use dependence: a conformational mechanism and a trapped ion mechanism. In this study, we used selectivity filter mutations (K1237R, K1237A, and K1237H) of the rat muscle NaV1.4 channel that are known to alter ionic selectivity and Ca(2+) permeability to test the trapped ion mechanism, which attributes use-dependent enhancement of toxin affinity to electrostatic repulsion between the bound toxin and Ca(2+) or Na(+) ions trapped inside the channel vestibule in the closed state. Our results indicate that TTX/STX use dependence is not relieved by mutations that enhance Ca(2+) permeability, suggesting that ion-toxin repulsion is not the primary factor that determines use dependence. Evidence now favors the idea that TTX/STX use dependence arises from conformational coupling of the voltage sensor domain or domains with residues in the toxin-binding site that are also involved in slow inactivation.
Collapse
|
36
|
Foadi N, Pilawski I, Buchholz V, de la Roche J, Wegner F, Leffler A, Ahrens J, Kästner S. Interaction of alfaxalone with the neuronal and the skeletal muscle sodium channel. Pharmacology 2012; 89:295-302. [PMID: 22538831 DOI: 10.1159/000337737] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2011] [Accepted: 02/29/2012] [Indexed: 11/19/2022]
Abstract
The neurosteroid alfaxalone exerts potent anesthetic activity in humans and animals. In former studies on myelinated axons, alfaxalone was assumed to produce a local anesthetic-like effect on the peripheral nervous system. Therefore,the present in vitro study aimed to characterize possible modulatory actions of alfaxalone on voltage-gated sodium channels. -Subunits of voltage-gated neuronal (Nav1.2)and skeletal muscle (Nav1.4) sodium channels were stably expressed in human embryonic kidney cells, and in vitro effects of alfaxalone were compared with lidocaine by means of the patch clamp technique. Alfaxalone preferentially blocked slow inactivated channels and therefore could provide membrane-stabilizing effects in ischemic/hypoxic tissues where slow inactivation is regarded to play a crucial role.
Collapse
Affiliation(s)
- Nilufar Foadi
- Clinic for Anaesthesia and Critical Care Medicine, Veterinary School of Medicine, Hannover, Germany
| | | | | | | | | | | | | | | |
Collapse
|
37
|
O’Reilly AO, Eberhardt E, Weidner C, Alzheimer C, Wallace BA, Lampert A. Bisphenol A binds to the local anesthetic receptor site to block the human cardiac sodium channel. PLoS One 2012; 7:e41667. [PMID: 22848561 PMCID: PMC3407203 DOI: 10.1371/journal.pone.0041667] [Citation(s) in RCA: 55] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2012] [Accepted: 06/23/2012] [Indexed: 12/24/2022] Open
Abstract
Bisphenol A (BPA) has attracted considerable public attention as it leaches from plastic used in food containers, is detectable in human fluids and recent epidemiologic studies link BPA exposure with diseases including cardiovascular disorders. As heart-toxicity may derive from modified cardiac electrophysiology, we investigated the interaction between BPA and hNav1.5, the predominant voltage-gated sodium channel subtype expressed in the human heart. Electrophysiology studies of heterologously-expressed hNav1.5 determined that BPA blocks the channel with a Kd of 25.4±1.3 µM. By comparing the effects of BPA and the local anesthetic mexiletine on wild type hNav1.5 and the F1760A mutant, we demonstrate that both compounds share an overlapping binding site. With a key binding determinant thus identified, an homology model of hNav1.5 was generated based on the recently-reported crystal structure of the bacterial voltage-gated sodium channel NavAb. Docking predictions position both ligands in a cavity delimited by F1760 and contiguous with the DIII–IV pore fenestration. Steered molecular dynamics simulations used to assess routes of ligand ingress indicate that the DIII–IV pore fenestration is a viable access pathway. Therefore BPA block of the human heart sodium channel involves the local anesthetic receptor and both BPA and mexiletine may enter the closed-state pore via membrane-located side fenestrations.
Collapse
Affiliation(s)
- Andrias O. O’Reilly
- Institute of Physiology and Pathophysiology, Friedrich-Alexander-Universität Erlangen-Nürnberg, Bavaria, Germany
- Department of Crystallography, Institute of Structural and Molecular Biology, Birkbeck College, University of London, London, United Kingdom
| | - Esther Eberhardt
- Institute of Physiology and Pathophysiology, Friedrich-Alexander-Universität Erlangen-Nürnberg, Bavaria, Germany
| | - Christian Weidner
- Institute of Physiology and Pathophysiology, Friedrich-Alexander-Universität Erlangen-Nürnberg, Bavaria, Germany
- Bavarian Health and Food Safety Authority, Erlangen, Germany
| | - Christian Alzheimer
- Institute of Physiology and Pathophysiology, Friedrich-Alexander-Universität Erlangen-Nürnberg, Bavaria, Germany
| | - B. A. Wallace
- Department of Crystallography, Institute of Structural and Molecular Biology, Birkbeck College, University of London, London, United Kingdom
| | - Angelika Lampert
- Institute of Physiology and Pathophysiology, Friedrich-Alexander-Universität Erlangen-Nürnberg, Bavaria, Germany
- * E-mail:
| |
Collapse
|
38
|
Song W, Xiao Y, Chen H, Ashpole NM, Piekarz AD, Ma P, Hudmon A, Cummins TR, Shou W. The human Nav1.5 F1486 deletion associated with long QT syndrome leads to impaired sodium channel inactivation and reduced lidocaine sensitivity. J Physiol 2012; 590:5123-39. [PMID: 22826127 DOI: 10.1113/jphysiol.2012.235374] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022] Open
Abstract
The deletion of phenylalanine 1486 (F1486del) in the human cardiac voltage-gated sodium channel (hNav1.5) is associated with fatal long QT (LQT) syndrome. In this study we determined how F1486del impairs the functional properties of hNav1.5 and alters action potential firing in heterologous expression systems (human embryonic kidney (HEK) 293 cells) and their native cardiomyocyte background. Cells expressing hNav1.5-F1486del exhibited a loss-of-function alteration, reflected by an 80% reduction of peak current density, and several gain-of-function alterations, including reduced channel inactivation, enlarged window current, substantial augmentation of persistent late sodium current and an increase in ramp current. We also observed substantial action potential duration (APD) prolongation and prominent early afterdepolarizations (EADs) in neonatal cardiomyocytes expressing the F1486del channels, as well as in computer simulations of myocyte activity. In addition, lidocaine sensitivity was dramatically reduced, which probably contributed to the poor therapeutic outcome observed in the patient carrying the hNav1.5-F1486del mutation. Therefore, despite the significant reduction in peak current density, the F1486del mutation also leads to substantial gain-of-function alterations that are sufficient to cause APD prolongation and EADs, the predominant characteristic of LQTs. These data demonstrate that hNav1.5 mutations can have complex functional consequences and highlight the importance of identifying the specific molecular defect when evaluating potential treatments for individuals with prolonged QT intervals.
Collapse
Affiliation(s)
- Weihua Song
- Riley Heart Research Center, Department of Pediatrics Herman B. Wells Center for Pediatric Research & Department of Pharmacology and Toxicology, Indiana University School of Medicine, 1044 W. Walnut Street, R4 W302D, Indianapolis, IN 46202, USA.
| | | | | | | | | | | | | | | | | |
Collapse
|
39
|
Beyder A, Strege PR, Bernard C, Farrugia G. Membrane permeable local anesthetics modulate Na(V)1.5 mechanosensitivity. Channels (Austin) 2012; 6:308-16. [PMID: 22874086 PMCID: PMC3508909 DOI: 10.4161/chan.21202] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Voltage-gated sodium selective ion channel NaV1.5 is expressed in the heart and the gastrointestinal tract, which are mechanically active organs. NaV1.5 is mechanosensitive at stimuli that gate other mechanosensitive ion channels. Local anesthetic and antiarrhythmic drugs act upon NaV1.5 to modulate activity by multiple mechanisms. This study examined whether NaV1.5 mechanosensitivity is modulated by local anesthetics. NaV1.5 channels wereexpressed in HEK-293 cells, and mechanosensitivity was tested in cell-attached and excised inside-out configurations. Using a novel protocol with paired voltage ladders and short pressure pulses, negative patch pressure (-30 mmHg) in both configurations produced a hyperpolarizing shift in the half-point of the voltage-dependence of activation (V1/2a) and inactivation (V1/2i) by about -10 mV. Lidocaine (50 µM) inhibited the pressure-induced shift of V1/2a but not V1/2i. Lidocaine inhibited the tonic increase in pressure-induced peak current in a use-dependence protocol, but it did not otherwise affect use-dependent block. The local anesthetic benzocaine, which does not show use-dependent block, also effectively blocked a pressure-induced shift in V1/2a. Lidocaine inhibited mechanosensitivity in NaV1.5 at the local anesthetic binding site mutated (F1760A). However, a membrane impermeable lidocaine analog QX-314 did not affect mechanosensitivity of F1760A NaV1.5 when applied from either side of the membrane. These data suggest that the mechanism of lidocaine inhibition of the pressure-induced shift in the half-point of voltage-dependence of activation is separate from the mechanisms of use-dependent block. Modulation of NaV1.5 mechanosensitivity by the membrane permeable local anesthetics may require hydrophobic access and may involve membrane-protein interactions.
Collapse
Affiliation(s)
- Arthur Beyder
- Division of Gastroenterology and Hepatology, Enteric Neuroscience Program, Mayo Clinic, Rochester, MN, USA
| | | | | | | |
Collapse
|
40
|
Desaphy JF, Dipalma A, Costanza T, Carbonara R, Dinardo MM, Catalano A, Carocci A, Lentini G, Franchini C, Camerino DC. Molecular Insights into the Local Anesthetic Receptor within Voltage-Gated Sodium Channels Using Hydroxylated Analogs of Mexiletine. Front Pharmacol 2012; 3:17. [PMID: 22403541 PMCID: PMC3279704 DOI: 10.3389/fphar.2012.00017] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2011] [Accepted: 01/30/2012] [Indexed: 12/26/2022] Open
Abstract
We previously showed that the β-adrenoceptor modulators, clenbuterol and propranolol, directly blocked voltage-gated sodium channels, whereas salbutamol and nadolol did not (Desaphy et al., 2003), suggesting the presence of two hydroxyl groups on the aromatic moiety of the drugs as a molecular requisite for impeding sodium channel block. To verify such an hypothesis, we synthesized five new mexiletine analogs by adding one or two hydroxyl groups to the aryloxy moiety of the sodium channel blocker and tested these compounds on hNav1.4 channels expressed in HEK293 cells. Concentration–response relationships were constructed using 25-ms-long depolarizing pulses at −30 mV applied from an holding potential of −120 mV at 0.1 Hz (tonic block) and 10 Hz (use-dependent block) stimulation frequencies. The half-maximum inhibitory concentrations (IC50) were linearly correlated to drug lipophilicity: the less lipophilic the drug, minor was the block. The same compounds were also tested on F1586C and Y1593C hNav1.4 channel mutants, to gain further information on the molecular interactions of mexiletine with its receptor within the sodium channel pore. In particular, replacement of Phe1586 and Tyr1593 by non-aromatic cysteine residues may help in the understanding of the role of π–π or π–cation interactions in mexiletine binding. Alteration of tonic block suggests that the aryloxy moiety of mexiletine may interact either directly or indirectly with Phe1586 in the closed sodium channel to produce low-affinity binding block, and that this interaction depends on the electrostatic potential of the drug aromatic tail. Alteration of use-dependent block suggests that addition of hydroxyl groups to the aryloxy moiety may modify high-affinity binding of the drug amine terminal to Phe1586 through cooperativity between the two pharmacophores, this effect being mainly related to drug lipophilicity. Mutation of Tyr1593 further impaired such cooperativity. In conclusion, these results confirm our former hypothesis by showing that the presence of hydroxyl groups to the aryloxy moiety of mexiletine greatly reduced sodium channel block, and provide molecular insights into the intimate interaction of local anesthetics with their receptor.
Collapse
Affiliation(s)
- Jean-François Desaphy
- Section of Pharmacology, Department of Pharmacobiology, Faculty of Pharmacy, University of Bari Bari, Italy
| | | | | | | | | | | | | | | | | | | |
Collapse
|
41
|
Fozzard HA, Sheets MF, Hanck DA. The sodium channel as a target for local anesthetic drugs. Front Pharmacol 2011; 2:68. [PMID: 22053156 PMCID: PMC3205381 DOI: 10.3389/fphar.2011.00068] [Citation(s) in RCA: 69] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2011] [Accepted: 10/14/2011] [Indexed: 01/08/2023] Open
Abstract
Na channels are the source of excitatory currents for the nervous system and muscle. They are the target for a class of drugs called local anesthetics (LA), which have been used for local and regional anesthesia and for excitatory problems such as epilepsy and cardiac arrhythmia. These drugs are prototypes for new analgesic drugs. The drug-binding site has been localized to the inner pore of the channel, where drugs interact mainly with a phenylalanine in domain IV S6. Drug affinity is both voltage- and use-dependent. Voltage-dependency is the result of changes in the conformation of the inner pore during channel activation and opening, allowing high energy interaction of drugs with the phenylalanine. LA drugs also reduce the gating current of Na channels, which represents the movement of charged residues in the voltage sensors. Specifically, drug binding to phenylalanine locks the domain III S4 in its outward (activated) position, and slows recovery of the domain IV S4. Although strongly affecting gating, LA drugs almost certainly also block by steric occlusion of the pore. Molecular definition of the binding and blocking interactions may help in new drug development.
Collapse
Affiliation(s)
- Harry A Fozzard
- Cardiac Electrophysiology Laboratory, Department of Medicine, The University of Chicago Chicago, IL, USA
| | | | | |
Collapse
|
42
|
A residue W756 in the P-loop segment of the sodium channel is critical for primaquine binding. Eur J Pharmacol 2011; 663:1-8. [DOI: 10.1016/j.ejphar.2011.04.025] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2010] [Revised: 03/17/2011] [Accepted: 04/06/2011] [Indexed: 11/18/2022]
|
43
|
Abstract
Preclinical strategies that are used to identify potential drug candidates include target-based screening, phenotypic screening, modification of natural substances and biologic-based approaches. To investigate whether some strategies have been more successful than others in the discovery of new drugs, we analysed the discovery strategies and the molecular mechanism of action (MMOA) for new molecular entities and new biologics that were approved by the US Food and Drug Administration between 1999 and 2008. Out of the 259 agents that were approved, 75 were first-in-class drugs with new MMOAs, and out of these, 50 (67%) were small molecules and 25 (33%) were biologics. The results also show that the contribution of phenotypic screening to the discovery of first-in-class small-molecule drugs exceeded that of target-based approaches - with 28 and 17 of these drugs coming from the two approaches, respectively - in an era in which the major focus was on target-based approaches. We postulate that a target-centric approach for first-in-class drugs, without consideration of an optimal MMOA, may contribute to the current high attrition rates and low productivity in pharmaceutical research and development.
Collapse
|
44
|
Molecular basis for class Ib anti-arrhythmic inhibition of cardiac sodium channels. Nat Commun 2011; 2:351. [PMID: 21673672 DOI: 10.1038/ncomms1351] [Citation(s) in RCA: 73] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2011] [Accepted: 05/12/2011] [Indexed: 12/19/2022] Open
Abstract
Cardiac sodium channels are established therapeutic targets for the management of inherited and acquired arrhythmias by class I anti-arrhythmic drugs (AADs). These drugs share a common target receptor bearing two highly conserved aromatic side chains, and are subdivided by the Vaughan-Williams classification system into classes Ia-c based on their distinct effects on the electrocardiogram. How can these drugs elicit distinct effects on the cardiac action potential by binding to a common receptor? Here we use fluorinated phenylalanine derivatives to test whether the electronegative surface potential of aromatic side chains contributes to inhibition by six class I AADs. Surprisingly, we find that class Ib AADs bind via a strong electrostatic cation-pi interaction, whereas class Ia and Ic AADs rely significantly less on this interaction. Our data shed new light on drug-target interactions underlying the inhibition of cardiac sodium channels by clinically relevant drugs and provide information for the directed design of AADs.
Collapse
|
45
|
Lenkey N, Karoly R, Lukacs P, Vizi ES, Sunesen M, Fodor L, Mike A. Classification of drugs based on properties of sodium channel inhibition: a comparative automated patch-clamp study. PLoS One 2010; 5:e15568. [PMID: 21187965 PMCID: PMC3004914 DOI: 10.1371/journal.pone.0015568] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2010] [Accepted: 11/15/2010] [Indexed: 01/28/2023] Open
Abstract
Background There is only one established drug binding site on sodium channels. However, drug binding of sodium channels shows extreme promiscuity: ∼25% of investigated drugs have been found to potently inhibit sodium channels. The structural diversity of these molecules suggests that they may not share the binding site, and/or the mode of action. Our goal was to attempt classification of sodium channel inhibitors by measuring multiple properties of inhibition in electrophysiology experiments. We also aimed to investigate if different properties of inhibition correlate with specific chemical properties of the compounds. Methodology/Principal Findings A comparative electrophysiological study of 35 compounds, including classic sodium channel inhibitors (anticonvulsants, antiarrhythmics and local anesthetics), as well as antidepressants, antipsychotics and neuroprotective agents, was carried out using rNav1.2 expressing HEK-293 cells and the QPatch automatic patch-clamp instrument. In the multi-dimensional space defined by the eight properties of inhibition (resting and inactivated affinity, potency, reversibility, time constants of onset and offset, use-dependence and state-dependence), at least three distinct types of inhibition could be identified; these probably reflect distinct modes of action. The compounds were clustered similarly in the multi-dimensional space defined by relevant chemical properties, including measures of lipophilicity, aromaticity, molecular size, polarity and electric charge. Drugs of the same therapeutic indication typically belonged to the same type. We identified chemical properties, which were important in determining specific properties of inhibition. State-dependence correlated with lipophilicity, the ratio of the neutral form of molecules, and aromaticity: We noticed that the highly state dependent inhibitors had at least two aromatic rings, logP>4.0, and pKa<8.0. Conclusions/Significance The correlations of inhibition properties both with chemical properties and therapeutic profiles would not have been evident through the sole determination of IC50; therefore, recording multiple properties of inhibition may allow improved prediction of therapeutic usefulness.
Collapse
Affiliation(s)
- Nora Lenkey
- Department of Pharmacology, Institute of Experimental Medicine, Hungarian Academy of Sciences, Budapest, Hungary
| | - Robert Karoly
- Department of Pharmacology, Institute of Experimental Medicine, Hungarian Academy of Sciences, Budapest, Hungary
| | - Peter Lukacs
- Department of Pharmacology, Institute of Experimental Medicine, Hungarian Academy of Sciences, Budapest, Hungary
| | - E. Sylvester Vizi
- Department of Pharmacology, Institute of Experimental Medicine, Hungarian Academy of Sciences, Budapest, Hungary
| | | | - Laszlo Fodor
- Pharmacology and Drug Safety Research, Gedeon Richter Plc., Budapest, Hungary
| | - Arpad Mike
- Department of Pharmacology, Institute of Experimental Medicine, Hungarian Academy of Sciences, Budapest, Hungary
- * E-mail:
| |
Collapse
|
46
|
Sheets MF, Fozzard HA, Lipkind GM, Hanck DA. Sodium channel molecular conformations and antiarrhythmic drug affinity. Trends Cardiovasc Med 2010; 20:16-21. [PMID: 20685573 DOI: 10.1016/j.tcm.2010.03.002] [Citation(s) in RCA: 43] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
Class I cardiac antiarrhythmic drugs, for example, lidocaine, mexiletine, flecainide, quinidine, and procainamide, continue to play an important role in the therapy for cardiac arrhythmias because of the presence of use-dependent block. Lidocaine, as well as related drugs such as mepivacaine, bupivacaine, and cocaine, also belong to the class of medications referred to as local anesthetics. In this review, we will consider lidocaine as the prototypical antiarrhythmic drug because it continues to be widely used both as an antiarrhythmic drug (first used as an antiarrhythmic drug in 1950) as well as a local anesthetic agent. Both of these clinical uses depend upon block of sodium current (I(Na)), but it is the presence of use-dependent I(Na) block, that is, an increasing amount of block at faster heart rates, which enables a local anesthetic agent to be a useful antiarrhythmic drug. Although many early studies investigated the action of antiarrhythmic drugs on Na currents, the availability of site-directed mutant Na channels has enabled for major advances in understanding their mechanisms of action based upon molecular conformations of the Na channel.
Collapse
Affiliation(s)
- Michael F Sheets
- University of Utah, The Nora Eccles Harrison Cardiovascular Research and Teaching Institute, Department of Internal Medicine, Salt Lake City, UT 84112, USA.
| | | | | | | |
Collapse
|
47
|
Sheets MF, Chen T, Hanck DA. Lidocaine partially depolarizes the S4 segment in domain IV of the sodium channel. Pflugers Arch 2010; 461:91-7. [PMID: 20981437 DOI: 10.1007/s00424-010-0894-1] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2010] [Revised: 08/10/2010] [Accepted: 10/06/2010] [Indexed: 01/16/2023]
Abstract
Previous studies have shown that lidocaine and other local anesthetic drugs (LAs) cause use-dependent block of sodium current (I (Na)), i.e., block that increases with membrane depolarization by allosteric coupling between drug binding in the inner pore and the S4s in domains III and IV. MTSET protection experiments have established that LAs stabilize DIIIS4 in an outward, depolarized position. Similar tests have not been reported for the DIVS4, although LAs have been shown to reduce DIV's contribution to total gating charge by about one third and to alter its movement such that it contributes more gating charge at negative potentials around -100 mV compared to non-drug-bound sodium (Na) channels. To investigate whether lidocaine reduces the gating charge of DIVS4 by causing it to adopt either a depolarized position at rest or by restricting its outward movement upon depolarization, we performed MTSET protection experiments on I (Na) of the mutant Na channel, R1628C (R3C-DIV), in the presence and absence of 10 mM lidocaine. The results indicate that lidocaine causes the DIVS4 to assume a more depolarized position, which facilitates its movement upon depolarization leading to the excess gating charge at potentials near -100 mV.
Collapse
Affiliation(s)
- Michael F Sheets
- The Nora Eccles Harrison Cardiovascular Research & Training Institute, University of Utah, 95 South 2000 East, Salt Lake City, UT 84112, USA.
| | | | | |
Collapse
|
48
|
Poulin P, Ekins S, Theil FP. A hybrid approach to advancing quantitative prediction of tissue distribution of basic drugs in human. Toxicol Appl Pharmacol 2010; 250:194-212. [PMID: 21034759 DOI: 10.1016/j.taap.2010.10.014] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2010] [Revised: 09/29/2010] [Accepted: 10/18/2010] [Indexed: 01/01/2023]
Abstract
A general toxicity of basic drugs is related to phospholipidosis in tissues. Therefore, it is essential to predict the tissue distribution of basic drugs to facilitate an initial estimate of that toxicity. The objective of the present study was to further assess the original prediction method that consisted of using the binding to red blood cells measured in vitro for the unbound drug (RBCu) as a surrogate for tissue distribution, by correlating it to unbound tissue:plasma partition coefficients (Kpu) of several tissues, and finally to predict volume of distribution at steady-state (V(ss)) in humans under in vivo conditions. This correlation method demonstrated inaccurate predictions of V(ss) for particular basic drugs that did not follow the original correlation principle. Therefore, the novelty of this study is to provide clarity on the actual hypotheses to identify i) the impact of pharmacological mode of action on the generic correlation of RBCu-Kpu, ii) additional mechanisms of tissue distribution for the outlier drugs, iii) molecular features and properties that differentiate compounds as outliers in the original correlation analysis in order to facilitate its applicability domain alongside the properties already used so far, and finally iv) to present a novel and refined correlation method that is superior to what has been previously published for the prediction of human V(ss) of basic drugs. Applying a refined correlation method after identifying outliers would facilitate the prediction of more accurate distribution parameters as key inputs used in physiologically based pharmacokinetic (PBPK) and phospholipidosis models.
Collapse
|
49
|
Desaphy JF, Dipalma A, Costanza T, Bruno C, Lentini G, Franchini C, George A, Conte Camerino D. Molecular determinants of state-dependent block of voltage-gated sodium channels by pilsicainide. Br J Pharmacol 2010; 160:1521-33. [PMID: 20590641 DOI: 10.1111/j.1476-5381.2010.00816.x] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
BACKGROUND AND PURPOSE Pilsicainide, an anti-arrhythmic drug used in Japan, is described as a pure sodium channel blocker. We examined the mechanisms by which it is able to block open channels, because these properties may be especially useful to reduce hyperexcitability in pathologies characterized by abnormal sodium channel opening. EXPERIMENTAL APPROACH The effects of pilsicainide on various heterologously expressed human sodium channel subtypes and mutants were investigated using the patch clamp technique. KEY RESULTS Pilsicainide exhibited tonic and use-dependent effects comparable to those of mexiletine and flecainide on hNav1.4 channels. These use-dependent effects were abolished in the mutations F1586C and Y1593C within segment 6 of domain IV, suggesting that the interaction of pilsicainide with these residues is critical for its local anaesthetic action. Its affinity constants for closed channels (K(R)) and channels inactivated from the closed state (K(I)) were high, suggesting that its use-dependent block (UDB) requires the channel to be open for it to reach a high-affinity blocking site. Accordingly, basic pH, which slightly increased the proportion of neutral drug, dramatically decreased K(R) and K(I) values. Effects of pilsicainide were similar on skeletal muscle hNav1.4, brain hNav1.1 and heart hNav1.5 channels. The myotonic R1448C and G1306E hNav1.4 mutants were more and less sensitive to pilsicainide, respectively, due to mutation-induced gating modifications. CONCLUSIONS AND IMPLICATIONS Although therapeutic concentrations of pilsicainide may have little effect on resting and closed-state inactivated channels, it induces a strong UDB due to channel opening, rendering the drug ideally suited for inhibition of high-frequency action potential firing.
Collapse
Affiliation(s)
- J-F Desaphy
- Department of Pharmacobiology, University of Bari, Bari, Italy.
| | | | | | | | | | | | | | | |
Collapse
|
50
|
Arcisio-Miranda M, Muroi Y, Chowdhury S, Chanda B. Molecular mechanism of allosteric modification of voltage-dependent sodium channels by local anesthetics. ACTA ACUST UNITED AC 2010; 136:541-54. [PMID: 20937693 PMCID: PMC2964522 DOI: 10.1085/jgp.201010438] [Citation(s) in RCA: 40] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
The hallmark of many intracellular pore blockers such as tetra-alkylammonium compounds and local anesthetics is their ability to allosterically modify the movement of the voltage sensors in voltage-dependent ion channels. For instance, the voltage sensor of domain III is specifically stabilized in the activated state when sodium currents are blocked by local anesthetics. The molecular mechanism underlying this long-range interaction between the blocker-binding site in the pore and voltage sensors remains poorly understood. Here, using scanning mutagenesis in combination with voltage clamp fluorimetry, we systematically evaluate the role of the internal gating interface of domain III of the sodium channel. We find that several mutations in the S4–S5 linker and S5 and S6 helices dramatically reduce the stabilizing effect of lidocaine on the activation of domain III voltage sensor without significantly altering use-dependent block at saturating drug concentrations. In the wild-type skeletal muscle sodium channel, local anesthetic block is accompanied by a 21% reduction in the total gating charge. In contrast, point mutations in this critical intracellular region reduce this charge modification by local anesthetics. Our analysis of a simple model suggests that these mutations in the gating interface are likely to disrupt the various coupling interactions between the voltage sensor and the pore of the sodium channel. These findings provide a molecular framework for understanding the mechanisms underlying allosteric interactions between a drug-binding site and voltage sensors.
Collapse
Affiliation(s)
- Manoel Arcisio-Miranda
- Department of Physiology, School of Medicine and Public Health, University of Wisconsin, Madison, WI 53706, USA
| | | | | | | |
Collapse
|