1
|
Neureiter EG, Erickson-Oberg MQ, Nigam A, Johnson JW. Inhibition of NMDA receptors and other ion channel types by membrane-associated drugs. Front Pharmacol 2025; 16:1561956. [PMID: 40371334 PMCID: PMC12075551 DOI: 10.3389/fphar.2025.1561956] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2025] [Accepted: 04/15/2025] [Indexed: 05/16/2025] Open
Abstract
N-methyl-D-aspartate receptors (NMDARs) are ligand-gated ion channels present at most excitatory synapses in the brain that play essential roles in cognitive functions including learning and memory consolidation. However, NMDAR dysregulation is implicated in many nervous system disorders. Diseases that involve pathological hyperactivity of NMDARs can be treated clinically through inhibition by channel blocking drugs. NMDAR channel block can occur via two known mechanisms. First, in traditional block, charged drug molecules can enter the channel directly from the extracellular solution after NMDAR activation and channel opening. Second, uncharged molecules of channel blocking drug can enter the hydrophobic plasma membrane, and upon NMDAR activation the membrane-associated drug can transit into the channel through a fenestration within the NMDAR. This membrane-associated mechanism of action is called membrane to channel inhibition (MCI) and is not well understood despite the clinical importance of NMDAR channel blocking drugs. Intriguingly, a hydrophobic route of access for drugs is not unique to NMDARs. Our review will address inhibition of NMDARs and other ion channels by membrane-associated drugs and consider how the path of access may affect a drug's therapeutic potential.
Collapse
Affiliation(s)
| | | | | | - Jon W. Johnson
- Department of Neuroscience and Center for Neuroscience, University of Pittsburgh, Pittsburgh, PA, United States
| |
Collapse
|
2
|
Tang R, Perez R, Brogan DM, Berezin MY, McCarthy JE. Imaging Peripheral Nerves In Vivo with CT Neurogram Using Novel 2,4,6-Tri-Iodinated Lidocaine Contrast Agent. Bioengineering (Basel) 2025; 12:422. [PMID: 40281782 PMCID: PMC12024922 DOI: 10.3390/bioengineering12040422] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2025] [Revised: 04/07/2025] [Accepted: 04/11/2025] [Indexed: 04/29/2025] Open
Abstract
Peripheral nerve injuries are a significant concern in surgical procedures, often leading to chronic pain and functional impairment. Despite advancements in imaging, preoperative and intraoperative visualization of peripheral nerves remains a challenge. This study introduces and evaluates a novel tri-iodinated lidocaine-based contrast agent for computed tomography neurography, aiming to enhance the intraoperative visibility of peripheral nerves in vivo. A tri-iodinated lidocaine analogue was synthesized and characterized for its radiodensity, sodium channel binding and nerve affinity. Sodium channel affinity was performed using molecular docking. In vitro contrast enhancement was assessed by comparing the agent's Hounsfield unit (HU) values with those of Omnipaque, a clinically approved contrast medium. In vivo imaging was conducted on rat sciatic nerves using micro-CT, followed by ex vivo validation. Nerve conduction blockade was assessed via electrical stimulation and histological analysis was performed to evaluate neurotoxicity. Experimental results revealed the tri-iodinated lidocaine analogue to have similar or higher affinity toward voltage-gated sodium channels than the parent lidocaine and a radiodensity comparable to the commercial CT contrast agent Omnipaque in vitro. In vivo, the contrast agent provided CT visualization of the sciatic nerve, with a significant increase in HU values compared to untreated nerves. Electrical stimulation confirmed transient nerve conduction blockade without observable histological damage, supporting its dual role as an imaging and nerve-blocking agent. This study presents a novel tri-iodinated lidocaine-based contrast agent that enables clear CT visualization of peripheral nerves while maintaining reversible nerve inhibition. These findings support its potential application in preoperative planning and intraoperative nerve protection to reduce surgical nerve injuries. Further studies are warranted to optimize imaging conditions and evaluate its clinical feasibility.
Collapse
Affiliation(s)
- Rui Tang
- Department of Radiology, School of Medicine, Washington University, St. Louis, MO 63110, USA;
| | - Ron Perez
- Department of Orthopedic Surgery, School of Medicine, Washington University, St. Louis, MO 63110, USA; (R.P.); (D.M.B.)
| | - David M. Brogan
- Department of Orthopedic Surgery, School of Medicine, Washington University, St. Louis, MO 63110, USA; (R.P.); (D.M.B.)
| | - Mikhail Y. Berezin
- Department of Radiology, School of Medicine, Washington University, St. Louis, MO 63110, USA;
| | | |
Collapse
|
3
|
Tao E, Corry B. Drugs exhibit diverse binding modes and access routes in the Nav1.5 cardiac sodium channel pore. J Gen Physiol 2025; 157:e202413658. [PMID: 39774837 PMCID: PMC11706274 DOI: 10.1085/jgp.202413658] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2024] [Revised: 11/05/2024] [Accepted: 12/07/2024] [Indexed: 01/11/2025] Open
Abstract
Small molecule inhibitors of the sodium channel are common pharmacological agents used to treat a variety of cardiac and nervous system pathologies. They act on the channel via binding within the pore to directly block the sodium conduction pathway and/or modulate the channel to favor a non-conductive state. Despite their abundant clinical use, we lack specific knowledge of their protein-drug interactions and the subtle variations between different compound structures. This study investigates the binding and accessibility of nine different compounds in the pore cavity of the Nav1.5 sodium channel using enhanced sampling simulations. We find that most compounds share a common location of pore binding-near the mouth of the DII-III fenestration-associated with the high number of aromatic residues in this region. In contrast, some other compounds prefer binding within the lateral fenestrations where they compete with lipids, rather than binding in the central cavity. Overall, our simulation results suggest that the drug binding within the pore is highly promiscuous, with most drugs having multiple low-affinity binding sites. Access to the pore interior via two out of four of the hydrophobic fenestrations is favorable for the majority of compounds. Our results indicate that the polyspecific and diffuse binding of inhibitors in the pore contributes to the varied nature of their inhibitory effects and can be exploited for future drug discovery and optimization.
Collapse
Affiliation(s)
- Elaine Tao
- Division of Biomedical Science and Biochemistry, Research School of Biology, Australian National University, Canberra, Australia
| | - Ben Corry
- Division of Biomedical Science and Biochemistry, Research School of Biology, Australian National University, Canberra, Australia
| |
Collapse
|
4
|
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
|
5
|
Pukkanasut P, Jaskula-Sztul R, Gomora JC, Velu SE. Therapeutic targeting of voltage-gated sodium channel Na V1.7 for cancer metastasis. Front Pharmacol 2024; 15:1416705. [PMID: 39045054 PMCID: PMC11263763 DOI: 10.3389/fphar.2024.1416705] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2024] [Accepted: 06/12/2024] [Indexed: 07/25/2024] Open
Abstract
This review focuses on the expression and function of voltage-gated sodium channel subtype NaV1.7 in various cancers and explores its impact on the metastasis driving cell functions such as proliferation, migration, and invasiveness. An overview of its structural characteristics, drug binding sites, inhibitors and their likely mechanisms of action are presented. Despite the lack of clarity on the precise mechanism by which NaV1.7 contributes to cancer progression and metastasis; many studies have suggested a connection between NaV1.7 and proteins involved in multiple signaling pathways such as PKA and EGF/EGFR-ERK1/2. Moreover, the functional activity of NaV1.7 appears to elevate the expression levels of MACC1 and NHE-1, which are controlled by p38 MAPK activity, HGF/c-MET signaling and c-Jun activity. This cascade potentially enhances the secretion of extracellular matrix proteases, such as MMPs which play critical roles in cell migration and invasion activities. Furthermore, the NaV1.7 activity may indirectly upregulate Rho GTPases Rac activity, which is critical for cytoskeleton reorganization, cell adhesion, and actin polymerization. The relationship between NaV1.7 and cancer progression has prompted researchers to investigate the therapeutic potential of targeting NaV1.7 using inhibitors. The positive outcome of such studies resulted in the discovery of several inhibitors with the ability to reduce cancer cell migration, invasion, and tumor growth underscoring the significance of NaV1.7 as a promising pharmacological target for attenuating cancer cell proliferation and metastasis. The research findings summarized in this review suggest that the regulation of NaV1.7 expression and function by small molecules and/or by genetic engineering is a viable approach to discover novel therapeutics for the prevention and treatment of metastasis of cancers with elevated NaV1.7 expression.
Collapse
Affiliation(s)
- Piyasuda Pukkanasut
- Department of Chemistry, The University of Alabama at Birmingham, Birmingham, AL, United States
| | - Renata Jaskula-Sztul
- Department of Surgery, The University of Alabama at Birmingham, Birmingham, AL, United States
- O’Neal Comprehensive Cancer Center, The University of Alabama at Birmingham, Birmingham, AL, United States
| | - Juan Carlos Gomora
- Departamento de Neuropatología Molecular, Instituto de Fisiología Celular, Universidad Nacional Autónoma de México, Mexico City, Mexico
| | - Sadanandan E. Velu
- Department of Chemistry, The University of Alabama at Birmingham, Birmingham, AL, United States
- O’Neal Comprehensive Cancer Center, The University of Alabama at Birmingham, Birmingham, AL, United States
| |
Collapse
|
6
|
Palmisano VF, Agnorelli C, Fagiolini A, Erritzoe D, Nutt D, Faraji S, Nogueira JJ. Membrane Permeation of Psychedelic Tryptamines by Dynamic Simulations. Biochemistry 2024. [PMID: 38324395 PMCID: PMC10882957 DOI: 10.1021/acs.biochem.3c00598] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/09/2024]
Abstract
Renewed scientific interest in psychedelic compounds represents one of the most promising avenues for addressing the current burden of mental health disorders. Classic psychedelics are a group of compounds that exhibit structural similarities to the naturally occurring neurotransmitter serotonin (5-HT). Acting on the 5-HT type 2A receptors (HT2ARs), psychedelics induce enduring neurophysiological changes that parallel their therapeutic psychological and behavioral effects. Recent preclinical evidence suggests that the ability of psychedelics to exert their action is determined by their ability to permeate the neuronal membrane to target a pool of intracellular 5-HT2ARs. In this computational study, we employ classical molecular dynamics simulations and umbrella sampling techniques to investigate the permeation behavior of 12 selected tryptamines and to characterize the interactions that drive the process. We aim at elucidating the impact of N-alkylation, indole ring substitution and positional modifications, and protonation on their membrane permeability. Dimethylation of the primary amine group and the introduction of a methoxy group at position 5 exhibited an increase in permeability. Moreover, there is a significant influence of positional substitutions on the indole groups, and the protonation of the molecules substantially increases the energy barrier at the center of the bilayer, making the compounds highly impermeable. All the information extracted from the trends predicted by the simulations can be applied in future drug design projects to develop psychedelics with enhanced activity.
Collapse
Affiliation(s)
- Vito F Palmisano
- Department of Chemistry, Universidad Autonoma de Madrid, Madrid 28049, Spain
- Theoretical Chemistry Group, Zernike Institute for Advanced Materials, University of Groningen, Groningen 9747 AG, The Netherlands
| | - Claudio Agnorelli
- Center for Psychedelic Research, Division of Psychiatry, Department of Brain Science, Imperial College of London, London SW7 2BX, U.K
- Unit of Psychiatry, Department of Molecular Medicine, University of Siena, Siena 53100, Italy
| | - Andrea Fagiolini
- Unit of Psychiatry, Department of Molecular Medicine, University of Siena, Siena 53100, Italy
| | - David Erritzoe
- Center for Psychedelic Research, Division of Psychiatry, Department of Brain Science, Imperial College of London, London SW7 2BX, U.K
| | - David Nutt
- Center for Psychedelic Research, Division of Psychiatry, Department of Brain Science, Imperial College of London, London SW7 2BX, U.K
| | - Shirin Faraji
- Theoretical Chemistry Group, Zernike Institute for Advanced Materials, University of Groningen, Groningen 9747 AG, The Netherlands
| | - Juan J Nogueira
- Department of Chemistry, Universidad Autonoma de Madrid, Madrid 28049, Spain
- IADCHEM, Institute for Advanced Research in Chemistry, Universidad Autonoma de Madrid, Madrid 28049, Spain
| |
Collapse
|
7
|
Wang G, Zhu L, Wu X, Qian Z. Influence of Protonation on the Norepinephrine Inhibiting α-Synuclein 71-82 Oligomerization. J Phys Chem B 2023; 127:7848-7857. [PMID: 37683121 DOI: 10.1021/acs.jpcb.3c03270] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/10/2023]
Abstract
The pathogenesis of Parkinson's disease (PD) is closely linked to the massive presence of Lewy vesicles and Lewy axons in the cytoplasm of neurons, mainly consisting of α-synuclein (αS). Norepinephrine (NE), whose secretion can be increased by exercise, has been demonstrated to prevent the fibrillation of αS and to break down the mature αS fibrils. In this work, we focus on the influence of protonation on the inhibitory ability of NE by using amyloid core fragment αS71-82 as a template. All-atom replica-exchange molecular dynamics simulations (accumulating to 33.6 μs) in explicit water were performed to explore the inhibitory effect of protonated and nonprotonated NE on αS oligomerization. Our results show that NE/NE+ can lead to a significant decrease in β-sheet content with increasing temperature, while isolated αS maintains relatively higher β-sheet conformations until 363 K, implying that both NE and NE+ can lower the critical temperature required for αS fibril decomposition. NE and NE+ also lead to the formation of less compact αS oligomers by preventing the backbone hydrogen bonds and the side-chain packing. The protonation would affect the binding affinity, interaction modes, and binding intensity of NE with αS. Interesting, NE and NE+ have a distinct binding free energy in the electrostatic and solvation terms, which mostly counter each other and produce a weak binding intensity with αS. Our work contributes to a better understanding of the inhibitory mechanism of NE and NE+ on αS oligomerization relevant to PD pathogenesis, which may provide clues for the design of antiamyloid medicine.
Collapse
Affiliation(s)
- Gang Wang
- Key Laboratory of Exercise and Health Sciences (Ministry of Education), Shanghai Frontiers Science Research Base of Exercise and Metabolic Health, and School of Exercise and Health, Shanghai University of Sport, 399 Changhai Road, Shanghai 200438, China
| | - Lili Zhu
- Key Laboratory of Exercise and Health Sciences (Ministry of Education), Shanghai Frontiers Science Research Base of Exercise and Metabolic Health, and School of Exercise and Health, Shanghai University of Sport, 399 Changhai Road, Shanghai 200438, China
- Shang Xing School, 6 Shangli Road, Shenzhen 518100, Guangdong, China
| | - Xiaoxiao Wu
- Key Laboratory of Exercise and Health Sciences (Ministry of Education), Shanghai Frontiers Science Research Base of Exercise and Metabolic Health, and School of Exercise and Health, Shanghai University of Sport, 399 Changhai Road, Shanghai 200438, China
| | - Zhenyu Qian
- Key Laboratory of Exercise and Health Sciences (Ministry of Education), Shanghai Frontiers Science Research Base of Exercise and Metabolic Health, and School of Exercise and Health, Shanghai University of Sport, 399 Changhai Road, Shanghai 200438, China
| |
Collapse
|
8
|
Wang G, Xu L, Chen H, Liu Y, Pan P, Hou T. Recent advances in computational studies on voltage‐gated sodium channels: Drug design and mechanism studies. WIRES COMPUTATIONAL MOLECULAR SCIENCE 2023. [DOI: 10.1002/wcms.1641] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/18/2023]
Affiliation(s)
- Gaoang Wang
- Innovation Institute for Artificial Intelligence in Medicine of Zhejiang University College of Pharmaceutical Sciences, Zhejiang University Hangzhou Zhejiang China
| | - Lei Xu
- Institute of Bioinformatics and Medical Engineering School of Electrical and Information Engineering, Jiangsu University of Technology Changzhou Jiangsu China
| | - Haiyi Chen
- Innovation Institute for Artificial Intelligence in Medicine of Zhejiang University College of Pharmaceutical Sciences, Zhejiang University Hangzhou Zhejiang China
| | - Yifei Liu
- Innovation Institute for Artificial Intelligence in Medicine of Zhejiang University College of Pharmaceutical Sciences, Zhejiang University Hangzhou Zhejiang China
| | - Peichen Pan
- Innovation Institute for Artificial Intelligence in Medicine of Zhejiang University College of Pharmaceutical Sciences, Zhejiang University Hangzhou Zhejiang China
| | - Tingjun Hou
- Innovation Institute for Artificial Intelligence in Medicine of Zhejiang University College of Pharmaceutical Sciences, Zhejiang University Hangzhou Zhejiang China
| |
Collapse
|
9
|
Choudhury K, Howard RJ, Delemotte L. An α-π transition in S6 shapes the conformational cycle of the bacterial sodium channel NavAb. J Gen Physiol 2022; 155:213748. [PMID: 36515966 PMCID: PMC9754703 DOI: 10.1085/jgp.202213214] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2022] [Revised: 10/17/2022] [Accepted: 11/28/2022] [Indexed: 12/15/2022] Open
Abstract
Voltage-gated sodium channels play an important role in electrical signaling in excitable cells. In response to changes in membrane potential, they cycle between nonconducting and conducting conformations. With recent advances in structural biology, structures of sodium channels have been captured in several distinct conformations, which are thought to represent different functional states. However, it has been difficult to capture the intrinsically transient open state. We recently showed that a proposed open state of the bacterial sodium channel NavMs was not conductive and that a conformational change involving a transition to a π-helix in the pore-lining S6 helix converted this structure into a conducting state. However, the relevance of this structural feature in other sodium channels, and its implications for the broader gating cycle, remained unclear. Here, we propose a comparable open state of another class of bacterial channel from Aliarcobacter butzleri (NavAb) with characteristic pore hydration, ion permeation, and drug binding properties. Furthermore, we show that a π-helix transition can lead to pore opening and that such a conformational change blocks fenestrations in the inner helix bundle. We also discover that a region in the C-terminal domain can undergo a disordering transition proposed to be important for pore opening. These results support a role for a π-helix transition in the opening of NavAb, enabling new proposals for the structural annotation and drug modulation mechanisms in this important sodium channel model.
Collapse
Affiliation(s)
- Koushik Choudhury
- Science for Life Laboratory, Department of Applied Physics, KTH Royal Institute of Technology, Solna, Sweden
| | - Rebecca J. Howard
- Science for Life Laboratory, Department of Biochemistry and Biophysics, Stockholm University, Stockholm, Sweden
| | - Lucie Delemotte
- Science for Life Laboratory, Department of Applied Physics, KTH Royal Institute of Technology, Solna, Sweden,Correspondence to Lucie Delemotte:
| |
Collapse
|
10
|
Zhorov BS, Dong K. Pyrethroids in an AlphaFold2 Model of the Insect Sodium Channel. INSECTS 2022; 13:745. [PMID: 36005370 PMCID: PMC9409284 DOI: 10.3390/insects13080745] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/17/2022] [Revised: 08/11/2022] [Accepted: 08/15/2022] [Indexed: 05/13/2023]
Abstract
Pyrethroid insecticides stabilize the open state of insect sodium channels. Previous mutational, electrophysiological, and computational analyses led to the development of homology models predicting two pyrethroid receptor sites, PyR1 and PyR2. Many of the naturally occurring sodium channel mutations, which confer knockdown resistance (kdr) to pyrethroids, are located within or close to these receptor sites, indicating that these mutations impair pyrethroid binding. However, the mechanism of the state-dependent action of pyrethroids and the mechanisms by which kdr mutations beyond the receptor sites confer resistance remain unclear. Recent advances in protein structure prediction using the AlphaFold2 (AF2) neural network allowed us to generate a new model of the mosquito sodium channel AaNav1-1, with the activated voltage-sensing domains (VSMs) and the presumably inactivated pore domain (PM). We further employed Monte Carlo energy minimizations to open PM and deactivate VSM-I and VSM-II to generate additional models. The docking of a Type II pyrethroid deltamethrin in the models predicted its interactions with many known pyrethroid-sensing residues in the PyR1 and PyR2 sites and revealed ligand-channel interactions that stabilized the open PM and activated VSMs. Our study confirms the predicted two pyrethroid receptor sites, explains the state-dependent action of pyrethroids, and proposes the mechanisms of the allosteric effects of various kdr mutations on pyrethroid action. The AF2-based models may assist in the structure-based design of new insecticides.
Collapse
Affiliation(s)
- Boris S. Zhorov
- Department of Biochemistry and Biomedical Sciences, McMaster University, Hamilton, ON L8S 4K1, Canada
- Sechenov Institute of Evolutionary Physiology & Biochemistry, Russian Academy of Sciences, Saint Petersburg 194223, Russia
- Almazov National Medical Research Centre, Saint Petersburg 197341, Russia
| | - Ke Dong
- Department of Biology, Duke University, Durham, NC 27708, USA
| |
Collapse
|
11
|
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: 20] [Impact Index Per Article: 6.7] [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
|
12
|
Tikhonov DB, Zhorov BS. P-Loop Channels: Experimental Structures, and Physics-Based and Neural Networks-Based Models. MEMBRANES 2022; 12:membranes12020229. [PMID: 35207150 PMCID: PMC8876033 DOI: 10.3390/membranes12020229] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/27/2021] [Revised: 02/09/2022] [Accepted: 02/09/2022] [Indexed: 01/27/2023]
Abstract
The superfamily of P-loop channels includes potassium, sodium, and calcium channels, as well as TRP channels and ionotropic glutamate receptors. A rapidly increasing number of crystal and cryo-EM structures have revealed conserved and variable elements of the channel structures. Intriguing differences are seen in transmembrane helices of channels, which may include π-helical bulges. The bulges reorient residues in the helices and thus strongly affect their intersegment contacts and patterns of ligand-sensing residues. Comparison of the experimental structures suggests that some π-bulges are dynamic: they may appear and disappear upon channel gating and ligand binding. The AlphaFold2 models represent a recent breakthrough in the computational prediction of protein structures. We compared some crystal and cryo-EM structures of P-loop channels with respective AlphaFold2 models. Folding of the regions, which are resolved experimentally, is generally similar to that predicted in the AlphaFold2 models. The models also reproduce some subtle but significant differences between various P-loop channels. However, patterns of π-bulges do not necessarily coincide in the experimental and AlphaFold2 structures. Given the importance of dynamic π-bulges, further studies involving experimental and theoretical approaches are necessary to understand the cause of the discrepancy.
Collapse
|
13
|
Tao E, Corry B. Characterizing fenestration size in sodium channel subtypes and their accessibility to inhibitors. Biophys J 2022; 121:193-206. [PMID: 34958776 PMCID: PMC8790208 DOI: 10.1016/j.bpj.2021.12.025] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2021] [Revised: 12/07/2021] [Accepted: 12/16/2021] [Indexed: 01/21/2023] Open
Abstract
Voltage-gated sodium channels (Nav) underlie the electrical activity of nerve and muscle cells. Humans have nine different subtypes of these channels, which are the target of small-molecule inhibitors commonly used to treat a range of conditions. Structural studies have identified four lateral fenestrations within the Nav pore module that have been shown to influence Nav pore blocker access during resting-state inhibition. However, the structural differences among the nine subtypes are still unclear. In particular, the dimensions of the four individual fenestrations across the Nav subtypes and their differential accessibility to pore blockers is yet to be characterized. To address this, we applied classical molecular dynamics simulations to study the recently published structures of Nav1.1, Nav1.2, Nav1.4, Nav1.5, and Nav1.7. Although there is significant variability in the bottleneck sizes of the Nav fenestrations, the subtypes follow a common pattern, with wider DI-II and DIII-IV fenestrations, a more restricted DII-III fenestration, and the most restricted DI-IV fenestration. We further identify the key bottleneck residues in each fenestration and show that the motions of aromatic residue sidechains govern the bottleneck radii. Well-tempered metadynamics simulations of Nav1.4 and Nav1.5 in the presence of the pore blocker lidocaine also support the DI-II fenestration being the most likely access route for drugs. Our computational results provide a foundation for future in vitro experiments examining the route of drug access to sodium channels. Understanding the fenestrations and their accessibility to drugs is critical for future analyses of diseases mutations across different sodium channel subtypes, with the potential to inform pharmacological development of resting-state inhibitors and subtype-selective drug design.
Collapse
Affiliation(s)
- Elaine Tao
- Research School of Biology, Australian National University, Canberra, Australia
| | - Ben Corry
- Research School of Biology, Australian National University, Canberra, Australia.
| |
Collapse
|
14
|
Choudhury K, Kasimova MA, McComas S, Howard RJ, Delemotte L. An open state of a voltage-gated sodium channel involving a π-helix and conserved pore-facing asparagine. Biophys J 2022; 121:11-22. [PMID: 34890580 PMCID: PMC8758419 DOI: 10.1016/j.bpj.2021.12.010] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2021] [Revised: 11/26/2021] [Accepted: 12/06/2021] [Indexed: 01/07/2023] Open
Abstract
Voltage-gated sodium (Nav) channels play critical roles in propagating action potentials and otherwise manipulating ionic gradients in excitable cells. These channels open in response to membrane depolarization, selectively permeating sodium ions until rapidly inactivating. Structural characterization of the gating cycle in this channel family has proved challenging, particularly due to the transient nature of the open state. A structure from the bacterium Magnetococcus marinus Nav (NavMs) was initially proposed to be open, based on its pore diameter and voltage-sensor conformation. However, the functional annotation of this model, and the structural details of the open state, remain disputed. In this work, we used molecular modeling and simulations to test possible open-state models of NavMs. The full-length experimental structure, termed here the α-model, was consistently dehydrated at the activation gate, indicating an inability to conduct ions. Based on a spontaneous transition observed in extended simulations, and sequence/structure comparison to other Nav channels, we built an alternative π-model featuring a helix transition and the rotation of a conserved asparagine residue into the activation gate. Pore hydration, ion permeation, and state-dependent drug binding in this model were consistent with an open functional state. This work thus offers both a functional annotation of the full-length NavMs structure and a detailed model for a stable Nav open state, with potential conservation in diverse ion-channel families.
Collapse
Affiliation(s)
- Koushik Choudhury
- Science for Life Laboratory, Department of Applied Physics, KTH Royal Institute of Technology, Solna, Sweden
| | - Marina A. Kasimova
- Science for Life Laboratory, Department of Applied Physics, KTH Royal Institute of Technology, Solna, Sweden
| | - Sarah McComas
- Science for Life Laboratory, Department of Biochemistry and Biophysics, Stockholm University, Stockholm, Sweden
| | - Rebecca J. Howard
- Science for Life Laboratory, Department of Biochemistry and Biophysics, Stockholm University, Stockholm, Sweden
| | - Lucie Delemotte
- Science for Life Laboratory, Department of Applied Physics, KTH Royal Institute of Technology, Solna, Sweden,Corresponding author
| |
Collapse
|
15
|
Manzur-Villalobos I, Contreras-Puentes N, Díaz-Caballero A, Durán-Lengua M, Alviz-Amador A. In silico study of local anesthetics analogues on sodium channel Nav 1.7 a pharmacological target on inflamed dental pulp. INFORMATICS IN MEDICINE UNLOCKED 2022. [DOI: 10.1016/j.imu.2022.101117] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022] Open
|
16
|
Buyan A, Corry B. Initiating Coarse-Grained MD Simulations for Membrane-Bound Proteins. Methods Mol Biol 2022; 2402:131-141. [PMID: 34854041 DOI: 10.1007/978-1-0716-1843-1_11] [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] [Indexed: 06/13/2023]
Abstract
Molecular dynamics (MD) simulations have become a widely used tool in the scientific community for understanding molecular scale phenomena that are challenging to address with wet-lab techniques. Coarse-grained simulations, in which multiple atoms are combined into single beads, allow for larger systems and longer time scales to be explored than atomistic techniques. Here, we describe the procedures and equipment required to set up coarse-grained simulations of membrane-bound proteins in a lipid bilayer that can mimic many membrane environments.
Collapse
Affiliation(s)
- Amanda Buyan
- Research School of Biology, Australian National University, Canberra, ACT, Australia.
| | - Ben Corry
- Research School of Biology, Australian National University, Canberra, ACT, Australia
| |
Collapse
|
17
|
Differences in local anaesthetic and antiepileptic binding in the inactivated state of human sodium channel Nav1.4. Biophys J 2021; 120:5553-5563. [PMID: 34774501 DOI: 10.1016/j.bpj.2021.11.014] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2021] [Revised: 09/27/2021] [Accepted: 11/05/2021] [Indexed: 11/22/2022] Open
Abstract
Voltage-gated sodium channels play a vital role in nerve and muscle cells, enabling them to encode and transmit electrical signals. Currently, there exist several classes of drugs that aim to inhibit these channels for therapeutic purposes, including local anesthetics, antiepileptics and antiarrhythmics. However, sodium-channel-inhibiting drugs lack subtype specificity; instead, they inhibit all sodium channels in the human body. Improving understanding of the mechanisms of binding of existing nonselective drugs is important in providing insight into how subtype-selective drugs could be developed. This study used molecular dynamics simulations to investigate the binding of the antiepileptics carbamazepine and lamotrigine and the local anesthetic lidocaine in neutral and charged states to the recently resolved human Nav1.4 channel. Replica exchange solute tempering was used to enable greater sampling of each compound within the pore. It was found that all four compounds show similarities in their binding sites within the pore. However, the positions of the carbamazepine and lamotrigine did not occlude the center of the pore but preferentially bound to homologous domain DII and DIII. The charged and neutral forms of lidocaine positioned themselves more centrally in the pore, with more common interactions with DIV. The best localized binding site was for charged lidocaine, whose aromatic moiety interacted with Y1593, whereas the amine projected toward the selectivity filter. Comparisons with our previous simulations and published structures highlight potential differences between tonic and use-dependent block related to conformational changes occurring in the pore.
Collapse
|
18
|
Grage SL, Culetto A, Ulrich AS, Weinschenk S. Membrane-Mediated Activity of Local Anesthetics. Mol Pharmacol 2021; 100:502-512. [PMID: 34475108 DOI: 10.1124/molpharm.121.000252] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2021] [Accepted: 07/19/2021] [Indexed: 01/06/2023] Open
Abstract
The activity of local anesthetics (LAs) has been attributed to the inhibition of ion channels, causing anesthesia. However, there is a growing body of research showing that LAs act on a wide range of receptors and channel proteins far beyond simple analgesia. The current concept of ligand recognition may no longer explain the multitude of protein targets influenced by LAs. We hypothesize that LAs can cause anesthesia without directly binding to the receptor proteins just by changing the physical properties of the lipid bilayer surrounding these proteins and ion channels based on LAs' amphiphilicity. It is possible that LAs act in one of the following ways: They 1) dissolve raft-like membrane microdomains, 2) impede nerve impulse propagation by lowering the lipid phase transition temperature, or 3) modulate the lateral pressure profile of the lipid bilayer. This could also explain the numerous additional effects of LAs besides anesthesia. Furthermore, the concepts of membrane-mediated activity and binding to ion channels do not have to exclude each other. If we were to consider LA as the middle part of a continuum between unspecific membrane-mediated activity on one end and highly specific ligand binding on the other end, we could describe LA as the link between the unspecific action of general anesthetics and toxins with their highly specific receptor binding. This comprehensive membrane-mediated model offers a fresh perspective to clinical and pharmaceutical research and therapeutic applications of local anesthetics. SIGNIFICANCE STATEMENT: Local anesthetics, according to the World Health Organization, belong to the most important drugs available to mankind. Their rediscovery as therapeutics and not only anesthetics marks a milestone in global pain therapy. The membrane-mediated mechanism of action proposed in this review can explain their puzzling variety of target proteins and their thus far inexplicable therapeutic effects. The new concept presented here places LAs on a continuum of structures and molecular mechanisms in between small general anesthetics and the more complex molecular toxins.
Collapse
Affiliation(s)
- Stephan L Grage
- Institute of Biological Interfaces (IBG-2), Karlsruhe Institute of Technology (KIT), Karlsruhe, Germany (S.L.G., A.C., A.S.U.); Institute of Organic Chemistry, KIT, Karlsruhe, Germany (A.C., A.S.U.); Women's Hospital, Department of Gynecological Endocrinology and Fertility Disorders, Heidelberg, Germany (S.W.); and The HUNTER Group, Heidelberg University, Women's Hospital, Im Neuenheimer Feld 440, 69120 Heidelberg, Germany, Heidelberg University, Women's Hospital, Neural Therapy Education & Research Group (S.W.)
| | - Anke Culetto
- Institute of Biological Interfaces (IBG-2), Karlsruhe Institute of Technology (KIT), Karlsruhe, Germany (S.L.G., A.C., A.S.U.); Institute of Organic Chemistry, KIT, Karlsruhe, Germany (A.C., A.S.U.); Women's Hospital, Department of Gynecological Endocrinology and Fertility Disorders, Heidelberg, Germany (S.W.); and The HUNTER Group, Heidelberg University, Women's Hospital, Im Neuenheimer Feld 440, 69120 Heidelberg, Germany, Heidelberg University, Women's Hospital, Neural Therapy Education & Research Group (S.W.)
| | - Anne S Ulrich
- Institute of Biological Interfaces (IBG-2), Karlsruhe Institute of Technology (KIT), Karlsruhe, Germany (S.L.G., A.C., A.S.U.); Institute of Organic Chemistry, KIT, Karlsruhe, Germany (A.C., A.S.U.); Women's Hospital, Department of Gynecological Endocrinology and Fertility Disorders, Heidelberg, Germany (S.W.); and The HUNTER Group, Heidelberg University, Women's Hospital, Im Neuenheimer Feld 440, 69120 Heidelberg, Germany, Heidelberg University, Women's Hospital, Neural Therapy Education & Research Group (S.W.)
| | - Stefan Weinschenk
- Institute of Biological Interfaces (IBG-2), Karlsruhe Institute of Technology (KIT), Karlsruhe, Germany (S.L.G., A.C., A.S.U.); Institute of Organic Chemistry, KIT, Karlsruhe, Germany (A.C., A.S.U.); Women's Hospital, Department of Gynecological Endocrinology and Fertility Disorders, Heidelberg, Germany (S.W.); and The HUNTER Group, Heidelberg University, Women's Hospital, Im Neuenheimer Feld 440, 69120 Heidelberg, Germany, Heidelberg University, Women's Hospital, Neural Therapy Education & Research Group (S.W.)
| |
Collapse
|
19
|
Zhorov BS. Structure of Sodium and Calcium Channels
with Ligands. J EVOL BIOCHEM PHYS+ 2021. [DOI: 10.1134/s0022093021020150] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
|
20
|
Palmisano VF, Gómez-Rodellar C, Pollak H, Cárdenas G, Corry B, Faraji S, Nogueira JJ. Binding of azobenzene and p-diaminoazobenzene to the human voltage-gated sodium channel Na v1.4. Phys Chem Chem Phys 2021; 23:3552-3564. [PMID: 33514952 DOI: 10.1039/d0cp06140a] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
The activity of voltage-gated ion channels can be controlled by the binding of photoswitches inside their internal cavity and subsequent light irradiation. We investigated the binding of azobenzene and p-diaminoazobenzene to the human Nav1.4 channel in the inactivated state by means of Gaussian accelerated molecular dynamics simulations and free-energy computations. Three stable binding pockets were identified for each of the two photoswitches. In all the cases, the binding is controlled by the balance between the favorable hydrophobic interactions of the ligands with the nonpolar residues of the protein and the unfavorable polar solvation energy. In addition, electrostatic interactions between the ligand and the polar aminoacids are also relevant for p-diaminoazobenzene due to the presence of the amino groups on the benzene moieties. These groups participate in hydrogen bonding in the most favorable binding pocket and in long-range electrostatic interactions in the other pockets. The thermodinamically preferred binding sites found for both photoswitches are close to the selectivity filter of the channel. Therefore, it is very likely that the binding of these ligands will induce alterations in the ion conduction through the channel.
Collapse
Affiliation(s)
- Vito F Palmisano
- Department of Chemistry, Universidad Autónoma de Madrid, Calle Francisco Tomás y Valiente, 7, 28049, Madrid, Spain.
| | | | | | | | | | | | | |
Collapse
|
21
|
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: 1.5] [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
|
22
|
Left Ventricular Hypertrophy Increases Susceptibility to Bupivacaine-induced Cardiotoxicity through Overexpression of Transient Receptor Potential Canonical Channels in Rats. Anesthesiology 2020; 133:1077-1092. [PMID: 32915958 DOI: 10.1097/aln.0000000000003554] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
BACKGROUND Local anesthetics, particularly potent long acting ones such as bupivacaine, can cause cardiotoxicity by inhibiting sodium ion channels; however, the impact of left ventricular hypertrophy on the cardiotoxicity and the underlying mechanisms remain undetermined. Transient receptor potential canonical (TRPC) channels are upregulated in left ventricular hypertrophy. Some transient receptor potential channel subtypes have been reported to pass relatively large cations, including protonated local anesthetics; this is known as the "pore phenomenon." The authors hypothesized that bupivacaine-induced cardiotoxicity is more severe in left ventricular hypertrophy due to upregulated TRPC channels. METHODS The authors used a modified transverse aortic constriction model as a left ventricular hypertrophy. Cardiotoxicity caused by bupivacaine was compared between sham and aortic constriction male rats, and the underlying mechanisms were investigated by recording sodium ion channel currents and immunocytochemistry of TRPC protein in cardiomyocytes. RESULTS The time to cardiac arrest by bupivacaine was shorter in aortic constriction rats (n =11) than in sham rats (n = 12) (mean ± SD, 1,302 ± 324 s vs. 1,034 ± 211 s; P = 0.030), regardless of its lower plasma concentration. The half-maximal inhibitory concentrations of bupivacaine toward sodium ion currents were 4.5 and 4.3 μM, which decreased to 3.9 and 2.6 μM in sham and aortic constriction rats, respectively, upon coapplication of 1-oleoyl-2-acetyl-sn-glycerol, a TRPC3 channel activator. In both groups, sodium ion currents were unaffected by QX-314, a positively charged lidocaine derivative, that hardly permeates the cell membrane, but was significantly decreased with QX-314 and 1-oleoyl-2-acetyl-sn-glycerol coapplication (sham: 79 ± 10% of control; P = 0.004; aortic constriction: 47± 27% of control; P = 0.020; n = 5 cells per group). Effects of 1-oleoyl-2-acetyl-sn-glycerol were antagonized by a specific TRPC3 channel inhibitor. CONCLUSIONS Left ventricular hypertrophy exacerbated bupivacaine-induced cardiotoxicity, which could be a consequence of the "pore phenomenon" of TRPC3 channels upregulated in left ventricular hypertrophy. EDITOR’S PERSPECTIVE
Collapse
|
23
|
Robles-Gómez E, Benítez-Villalobos F, Soriano-García M, Antúnez-Argüelles E. Non-peptide molecules in the pedicellariae of Toxopneustes roseus. Toxicon 2020; 184:143-151. [DOI: 10.1016/j.toxicon.2020.06.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2019] [Revised: 05/25/2020] [Accepted: 06/02/2020] [Indexed: 11/30/2022]
|
24
|
Tikhonov DB, Lin L, Yang DSC, Yuchi Z, Zhorov BS. Phenylalkylamines in calcium channels: computational analysis of experimental structures. J Comput Aided Mol Des 2020; 34:1157-1169. [PMID: 32648151 DOI: 10.1007/s10822-020-00330-0] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2020] [Accepted: 07/03/2020] [Indexed: 12/16/2022]
Abstract
Experimental 3D structures of calcium channels with phenylalkylamines (PAAs) provide basis for further analysis of atomic mechanisms of these important cardiovascular drugs. In the crystal structure of the engineered calcium channel CavAb with Br-verapamil and in the cryo-EM structure of the Cav1.1 channel with verapamil, the ligands bind in the inner pore. However, there are significant differences between these structures. In the crystal structure the ligand ammonium group is much closer to the ion in the selectivity-filter region Site 3, which is most proximal to the inner pore, than in the cryo-EM structure. Here we used Monte Carlo energy minimizations to dock PAAs in calcium channels. Our computations suggest that in the crystal structure Site 3 is occupied by a water molecule rather than by a calcium ion. Analysis of the published electron density map does not rule out this possibility. In the cryo-EM structures the ammonium group of verapamil is shifted from the calcium ion in Site 3 either along the pore axis, towards the cytoplasm or away from the axis. Our unbiased docking reproduced these binding modes. However, in the cryo-EM structures detergent and lipid molecules interact with verapamil. When we removed these molecules, the nitrile group of verapamil bound to the calcium ion in Site 3. Models of Cav1.2 with different PAAs suggest similar binding modes and direct contacts of the ligands electronegative atoms with the calcium ion in Site 3. Such interactions explain paradoxes in structure-activity relationships of PAAs.
Collapse
Affiliation(s)
- Denis B Tikhonov
- Sechenov Institute of Evolutionary Physiology and Biochemistry, Russian Academy of Sciences, St. Petersburg, Russian Federation.
| | - Lianyun Lin
- Tianjin Key Laboratory for Modern Drug Delivery & High-Efficiency, Collaborative Innovation Center of Chemical Science and Engineering, School of Pharmaceutical Science and Technology, Tianjin University, Tianjin, China
| | - Daniel S C Yang
- Almazov National Medical Research Centre, St. Petersburg, Russia, 197341
| | - Zhiguang Yuchi
- Tianjin Key Laboratory for Modern Drug Delivery & High-Efficiency, Collaborative Innovation Center of Chemical Science and Engineering, School of Pharmaceutical Science and Technology, Tianjin University, Tianjin, China.
| | - Boris S Zhorov
- Sechenov Institute of Evolutionary Physiology and Biochemistry, Russian Academy of Sciences, St. Petersburg, Russian Federation.
- Almazov National Medical Research Centre, St. Petersburg, Russia, 197341.
- Department of Biochemistry and Biomedical Sciences, McMaster University, Hamilton, ON, Canada.
| |
Collapse
|
25
|
Lee S, Jo S, Talbot S, Zhang HXB, Kotoda M, Andrews NA, Puopolo M, Liu PW, Jacquemont T, Pascal M, Heckman LM, Jain A, Lee J, Woolf CJ, Bean BP. Novel charged sodium and calcium channel inhibitor active against neurogenic inflammation. eLife 2019; 8:48118. [PMID: 31765298 PMCID: PMC6877086 DOI: 10.7554/elife.48118] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2019] [Accepted: 11/13/2019] [Indexed: 12/12/2022] Open
Abstract
Voltage-dependent sodium and calcium channels in pain-initiating nociceptor neurons are attractive targets for new analgesics. We made a permanently charged cationic derivative of an N-type calcium channel-inhibitor. Unlike cationic derivatives of local anesthetic sodium channel blockers like QX-314, this cationic compound inhibited N-type calcium channels more effectively with extracellular than intracellular application. Surprisingly, the compound is also a highly effective sodium channel inhibitor when applied extracellularly, producing more potent inhibition than lidocaine or bupivacaine. The charged inhibitor produced potent and long-lasting analgesia in mouse models of incisional wound and inflammatory pain, inhibited release of the neuropeptide calcitonin gene-related peptide (CGRP) from dorsal root ganglion neurons, and reduced inflammation in a mouse model of allergic asthma, which has a strong neurogenic component. The results show that some cationic molecules applied extracellularly can powerfully inhibit both sodium channels and calcium channels, thereby blocking both nociceptor excitability and pro-inflammatory peptide release.
Collapse
Affiliation(s)
- Seungkyu Lee
- FM Kirby Neurobiology Research Center, Boston Children's Hospital, Boston, United States
| | - Sooyeon Jo
- Department of Neurobiology, Harvard Medical School, Boston, United States
| | - Sébastien Talbot
- Département de Pharmacologie et Physiologie, Université de Montréal, Montréal, Canada
| | | | - Masakazu Kotoda
- FM Kirby Neurobiology Research Center, Boston Children's Hospital, Boston, United States
| | - Nick A Andrews
- FM Kirby Neurobiology Research Center, Boston Children's Hospital, Boston, United States
| | - Michelino Puopolo
- Department of Anesthesiology, Renaissance School of Medicine at Stony Brook University, Stony Brook, United States
| | - Pin W Liu
- Department of Neurobiology, Harvard Medical School, Boston, United States
| | - Thomas Jacquemont
- FM Kirby Neurobiology Research Center, Boston Children's Hospital, Boston, United States
| | - Maud Pascal
- FM Kirby Neurobiology Research Center, Boston Children's Hospital, Boston, United States
| | - Laurel M Heckman
- FM Kirby Neurobiology Research Center, Boston Children's Hospital, Boston, United States
| | - Aakanksha Jain
- FM Kirby Neurobiology Research Center, Boston Children's Hospital, Boston, United States
| | - Jinbo Lee
- Sage Partner International, Andover, United States
| | - Clifford J Woolf
- FM Kirby Neurobiology Research Center, Boston Children's Hospital, Boston, United States.,Department of Neurobiology, Harvard Medical School, Boston, United States
| | - Bruce P Bean
- Department of Neurobiology, Harvard Medical School, Boston, United States
| |
Collapse
|
26
|
Bedoya M, Rinné S, Kiper AK, Decher N, González W, Ramírez D. TASK Channels Pharmacology: New Challenges in Drug Design. J Med Chem 2019; 62:10044-10058. [PMID: 31260312 DOI: 10.1021/acs.jmedchem.9b00248] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
Abstract
Rational drug design targeting ion channels is an exciting and always evolving research field. New medicinal chemistry strategies are being implemented to explore the wild chemical space and unravel the molecular basis of the ion channels modulators binding mechanisms. TASK channels belong to the two-pore domain potassium channel family and are modulated by extracellular acidosis. They are extensively distributed along the cardiovascular and central nervous systems, and their expression is up- and downregulated in different cancer types, which makes them an attractive therapeutic target. However, TASK channels remain unexplored, and drugs designed to target these channels are poorly selective. Here, we review TASK channels properties and their known blockers and activators, considering the new challenges in ion channels drug design and focusing on the implementation of computational methodologies in the drug discovery process.
Collapse
Affiliation(s)
- Mauricio Bedoya
- Centro de Bioinformática y Simulación Molecular (CBSM) , Universidad de Talca , 1 Poniente No. 1141 , 3460000 Talca , Chile
| | - Susanne Rinné
- Institute for Physiology and Pathophysiology, Vegetative Physiology and Marburg Center for Mind, Brain and Behavior, MCMBB , Philipps-University of Marburg , Deutschhausstraße 2 , Marburg 35037 , Germany
| | - Aytug K Kiper
- Institute for Physiology and Pathophysiology, Vegetative Physiology and Marburg Center for Mind, Brain and Behavior, MCMBB , Philipps-University of Marburg , Deutschhausstraße 2 , Marburg 35037 , Germany
| | - Niels Decher
- Institute for Physiology and Pathophysiology, Vegetative Physiology and Marburg Center for Mind, Brain and Behavior, MCMBB , Philipps-University of Marburg , Deutschhausstraße 2 , Marburg 35037 , Germany
| | - Wendy González
- Centro de Bioinformática y Simulación Molecular (CBSM) , Universidad de Talca , 1 Poniente No. 1141 , 3460000 Talca , Chile.,Millennium Nucleus of Ion Channels-Associated Diseases (MiNICAD) , Universidad de Talca , 1 Poniente No. 1141 , 3460000 Talca , Chile
| | - David Ramírez
- Instituto de Ciencias Biomédicas, Facultad de Ciencias de la Salud , Universidad Autónoma de Chile , El Llano Subercaseaux 2801, Piso 6 , 8900000 Santiago , Chile
| |
Collapse
|
27
|
Flood E, Boiteux C, Lev B, Vorobyov I, Allen TW. Atomistic Simulations of Membrane Ion Channel Conduction, Gating, and Modulation. Chem Rev 2019; 119:7737-7832. [DOI: 10.1021/acs.chemrev.8b00630] [Citation(s) in RCA: 65] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Affiliation(s)
- Emelie Flood
- School of Science, RMIT University, Melbourne, Victoria 3000, Australia
| | - Céline Boiteux
- School of Science, RMIT University, Melbourne, Victoria 3000, Australia
| | - Bogdan Lev
- School of Science, RMIT University, Melbourne, Victoria 3000, Australia
| | - Igor Vorobyov
- Department of Physiology & Membrane Biology/Department of Pharmacology, University of California, Davis, 95616, United States
| | - Toby W. Allen
- School of Science, RMIT University, Melbourne, Victoria 3000, Australia
| |
Collapse
|
28
|
Belinskaia DA, Belinskaia MA, Barygin OI, Vanchakova NP, Shestakova NN. Psychotropic Drugs for the Management of Chronic Pain and Itch. Pharmaceuticals (Basel) 2019; 12:ph12020099. [PMID: 31238561 PMCID: PMC6631469 DOI: 10.3390/ph12020099] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2019] [Revised: 06/19/2019] [Accepted: 06/21/2019] [Indexed: 12/11/2022] Open
Abstract
Clinical observations have shown that patients with chronic neuropathic pain or itch exhibit symptoms of increased anxiety, depression and cognitive impairment. Such patients need corrective therapy with antidepressants, antipsychotics or anticonvulsants. It is known that some psychotropic drugs are also effective for the treatment of neuropathic pain and pruritus syndromes due to interaction with the secondary molecular targets. Our own clinical studies have identified antipruritic and/or analgesic efficacy of the following compounds: tianeptine (atypical tricyclic antidepressant), citalopram (selective serotonin reuptake inhibitor), mianserin (tetracyclic antidepressant), carbamazepine (anticonvulsant), trazodone (serotonin antagonist and reuptake inhibitor), and chlorprothixene (antipsychotic). Venlafaxine (serotonin-norepinephrine reuptake inhibitor) is known to have an analgesic effect too. The mechanism of such effect of these drugs is not fully understood. Herein we review and correlate the literature data on analgesic/antipruritic activity with pharmacological profile of these compounds.
Collapse
Affiliation(s)
- Daria A Belinskaia
- Sechenov Institute of Evolutionary Physiology and Biochemistry, Russian Academy of Sciences, pr. Torez 44, St. Petersburg 194223, Russia.
| | - Mariia A Belinskaia
- International Centre for Neurotherapeutics, Dublin City University, Glasnevin, Dublin 9, Ireland.
| | - Oleg I Barygin
- Sechenov Institute of Evolutionary Physiology and Biochemistry, Russian Academy of Sciences, pr. Torez 44, St. Petersburg 194223, Russia.
| | - Nina P Vanchakova
- Department of Pedagogy and Psychology, Faculty of Postgraduate Education, First Pavlov State Medical University, L'va Tolstogo str. 6-8, St. Petersburg 197022, Russia.
| | - Natalia N Shestakova
- Sechenov Institute of Evolutionary Physiology and Biochemistry, Russian Academy of Sciences, pr. Torez 44, St. Petersburg 194223, Russia.
| |
Collapse
|
29
|
Xu L, Ding X, Wang T, Mou S, Sun H, Hou T. Voltage-gated sodium channels: structures, functions, and molecular modeling. Drug Discov Today 2019; 24:1389-1397. [PMID: 31129313 DOI: 10.1016/j.drudis.2019.05.014] [Citation(s) in RCA: 39] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2019] [Revised: 04/02/2019] [Accepted: 05/17/2019] [Indexed: 10/26/2022]
Abstract
Voltage-gated sodium channels (VGSCs), formed by 24 transmembrane segments arranged into four domains, have a key role in the initiation and propagation of electrical signaling in excitable cells. VGSCs are involved in a variety of diseases, including epilepsy, cardiac arrhythmias, and neuropathic pain, and therefore have been regarded as appealing therapeutic targets for the development of anticonvulsant, antiarrhythmic, and local anesthetic drugs. In this review, we discuss recent advances in understanding the structures and biological functions of VGSCs. In addition, we systematically summarize eight pharmacologically distinct ligand-binding sites in VGSCs and representative isoform-selective VGSC modulators in clinical trials. Finally, we review studies on molecular modeling and computer-aided drug design (CADD) for VGSCs to help understanding of biological processes involving VGSCs.
Collapse
Affiliation(s)
- Lei Xu
- Institute of Bioinformatics and Medical Engineering, School of Electrical and Information Engineering, Jiangsu University of Technology, Changzhou 213001, China
| | - Xiaoqin Ding
- Beijing Institute of Pharmaceutical Chemistry, Beijing 102205, China
| | - Tianhu Wang
- Institute of Bioinformatics and Medical Engineering, School of Electrical and Information Engineering, Jiangsu University of Technology, Changzhou 213001, China
| | - Shanzhi Mou
- School of Mathematics and Physics, Jiangsu University of Technology, Changzhou 213001, China
| | - Huiyong Sun
- Department of Medicinal Chemistry, School of Pharmacy, State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing 210009, China.
| | - Tingjun Hou
- College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China.
| |
Collapse
|
30
|
Ropp PJ, Kaminsky JC, Yablonski S, Durrant JD. Dimorphite-DL: an open-source program for enumerating the ionization states of drug-like small molecules. J Cheminform 2019; 11:14. [PMID: 30767086 PMCID: PMC6689865 DOI: 10.1186/s13321-019-0336-9] [Citation(s) in RCA: 33] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2018] [Accepted: 02/08/2019] [Indexed: 12/22/2022] Open
Abstract
Small-molecule protonation can promote or discourage protein binding by altering hydrogen-bond, electrostatic, and van-der-Waals interactions. To improve virtual-screen pose and affinity predictions, researchers must account for all major small-molecule ionization states. But existing programs for calculating these states have notable limitations such as high cost, restrictive licenses, slow execution times, and poor modularity. Here, we present dimorphite-DL 1.0, a fast, accurate, accessible, and modular open-source program for enumerating small-molecule ionization states. Dimorphite-DL uses a straightforward empirical algorithm that leverages substructure searching and draws on a database of experimentally characterized ionizable molecules. We have tested dimorphite-DL using several versions of Python and RDKit on all major operating systems. We release it under the terms of the Apache License, Version 2.0. A copy is available free of charge from http://durrantlab.com/dimorphite-dl/.
Collapse
Affiliation(s)
- Patrick J Ropp
- Department of Biological Sciences, University of Pittsburgh, 4249 Fifth Avenue, Pittsburgh, PA, 15260, USA
| | - Jesse C Kaminsky
- Department of Biological Sciences, University of Pittsburgh, 4249 Fifth Avenue, Pittsburgh, PA, 15260, USA
| | - Sara Yablonski
- Department of Biological Sciences, University of Pittsburgh, 4249 Fifth Avenue, Pittsburgh, PA, 15260, USA
| | - Jacob D Durrant
- Department of Biological Sciences, University of Pittsburgh, 4249 Fifth Avenue, Pittsburgh, PA, 15260, USA.
| |
Collapse
|
31
|
Structural basis for antiarrhythmic drug interactions with the human cardiac sodium channel. Proc Natl Acad Sci U S A 2019; 116:2945-2954. [PMID: 30728299 PMCID: PMC6386684 DOI: 10.1073/pnas.1817446116] [Citation(s) in RCA: 63] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Voltage-gated sodium channels play a central role in cellular excitability and are key targets for drug development. Recent breakthroughs in high-resolution cryo-electron microscopy protein structure determination, Rosetta computational protein structure modeling, and multimicrosecond molecular dynamics simulations are empowering advances in structural biology to study the atomistic details of channel−drug interactions. We used Rosetta structural computational modeling and molecular dynamics simulations to study the interactions of antiarrhythmic and local anesthetic drugs with cardiac sodium channel. Our results provide crucial atomic-scale mechanistic insights into the channel–drug interactions, necessary for the rational design of novel modulators of the human cardiac sodium channel to be used for the treatment of cardiac arrhythmias. The human voltage-gated sodium channel, hNaV1.5, is responsible for the rapid upstroke of the cardiac action potential and is target for antiarrhythmic therapy. Despite the clinical relevance of hNaV1.5-targeting drugs, structure-based molecular mechanisms of promising or problematic drugs have not been investigated at atomic scale to inform drug design. Here, we used Rosetta structural modeling and docking as well as molecular dynamics simulations to study the interactions of antiarrhythmic and local anesthetic drugs with hNaV1.5. These calculations revealed several key drug binding sites formed within the pore lumen that can simultaneously accommodate up to two drug molecules. Molecular dynamics simulations identified a hydrophilic access pathway through the intracellular gate and a hydrophobic access pathway through a fenestration between DIII and DIV. Our results advance the understanding of molecular mechanisms of antiarrhythmic and local anesthetic drug interactions with hNaV1.5 and will be useful for rational design of novel therapeutics.
Collapse
|
32
|
Pugsley MK, Yong SL, Goldin AL, Hayes ES, Walker MJA. Molecular charge associated with antiarrhythmic actions in a series of amino-2-cyclohexyl ester derivatives. Eur J Pharmacol 2019; 844:241-252. [PMID: 30571955 DOI: 10.1016/j.ejphar.2018.12.025] [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: 08/05/2018] [Revised: 11/12/2018] [Accepted: 12/12/2018] [Indexed: 11/15/2022]
Abstract
A series of amino-2-cyclohexyl ester derivatives were studied for their ion channel blocking and antiarrhythmic actions in the rat and a structure-activity analysis was conducted. The compounds are similar in chemical structure except for ionizable amine groups (pK values 6.1-8.9) and the positional arrangements of aromatic naphthyl moieties. Ventricular arrhythmias were produced in rats by coronary-artery occlusion or electrical stimulation. The electrophysiological effects of these compounds on rat heart sodium channels (Nav1.5) expressed in Xenopus laevis oocytes and transient outward potassium currents (Kv4.3) from isolated rat ventricular myocytes were examined. The compounds reduced the incidence of ischemia-related arrhythmias and increased current threshold for induction of ventricular fibrillo-flutter (VFt) dose-dependently. As pK increased compounds showed a diminished effectiveness against ischemia-induced arrhythmias, and were less selective for ischemia- versus electrically-induced arrhythmias. Where tested, compounds produced a concentration-dependent tonic block of Nav1.5 channels. An increased potency for inhibition of Nav1.5 occurred when the external pH (pHo) was reduced to 6.5. Some compounds inhibited Kv4.3 in a pH-independent manner. Overall, the differences in antiarrhythmic and ion channel blocking properties in this series of compounds can be explained by differences in chemical structure. Antiarrhythmic activity for the amino-2-cyclohexyl ester derivatives is likely a function of mixed ion channel blockade in ischemic myocardium. These studies show that drug inhibition of Nav1.5 occurred at lower concentrations than Kv4.3 and was more sensitive to changes in the ionizable amine groups rather than on positional arrangements of the naphthyl constituents. These results offer insight into antiarrhythmic mechanisms of drug-ion channel interactions.
Collapse
Affiliation(s)
- Michael K Pugsley
- Safety Pharmacology & Toxicology, Fairfield, CT 06825, USA; Department of Pharmacology & Therapeutics, Faculty of Medicine, The University of British Columbia, 2176 Health Sciences Mall, Vancouver, BC, Canada, V6T 1Z3.
| | - Sandro L Yong
- Department of Pharmacology & Therapeutics, Faculty of Medicine, The University of British Columbia, 2176 Health Sciences Mall, Vancouver, BC, Canada, V6T 1Z3
| | - Alan L Goldin
- Department of Microbiology & Molecular Genetics, University of California, Irvine, California, CA 92697-4025, USA
| | - Eric S Hayes
- BioCurate Pty Ltd., Parkville, Victoria, Australia
| | - Michael J A Walker
- Department of Pharmacology & Therapeutics, Faculty of Medicine, The University of British Columbia, 2176 Health Sciences Mall, Vancouver, BC, Canada, V6T 1Z3
| |
Collapse
|
33
|
Oakes V, Domene C. Capturing the Molecular Mechanism of Anesthetic Action by Simulation Methods. Chem Rev 2018; 119:5998-6014. [DOI: 10.1021/acs.chemrev.8b00366] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Affiliation(s)
- Victoria Oakes
- Department of Chemistry, University of Bath, Claverton Down, Bath BA2 7AY, United Kingdom
| | - Carmen Domene
- Department of Chemistry, University of Bath, Claverton Down, Bath BA2 7AY, United Kingdom
- Department of Chemistry, University of Oxford, Oxford OX1 3TA, United Kingdom
| |
Collapse
|
34
|
Radwański PB, Johnson CN, Györke S, Veeraraghavan R. Cardiac Arrhythmias as Manifestations of Nanopathies: An Emerging View. Front Physiol 2018; 9:1228. [PMID: 30233404 PMCID: PMC6131669 DOI: 10.3389/fphys.2018.01228] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2018] [Accepted: 08/14/2018] [Indexed: 12/21/2022] Open
Abstract
A nanodomain is a collection of proteins localized within a specialized, nanoscale structural environment, which can serve as the functional unit of macroscopic physiologic processes. We are beginning to recognize the key roles of cardiomyocyte nanodomains in essential processes of cardiac physiology such as electrical impulse propagation and excitation–contraction coupling (ECC). There is growing appreciation of nanodomain dysfunction, i.e., nanopathy, as a mechanistic driver of life-threatening arrhythmias in a variety of pathologies. Here, we offer an overview of current research on the role of nanodomains in cardiac physiology with particular emphasis on: (1) sodium channel-rich nanodomains within the intercalated disk that participate in cell-to-cell electrical coupling and (2) dyadic nanodomains located along transverse tubules that participate in ECC. The beat to beat function of cardiomyocytes involves three phases: the action potential, the calcium transient, and mechanical contraction/relaxation. In all these phases, cell-wide function results from the aggregation of the stochastic function of individual proteins. While it has long been known that proteins that exist in close proximity influence each other’s function, it is increasingly appreciated that there exist nanoscale structures that act as functional units of cardiac biophysical phenomena. Termed nanodomains, these structures are collections of proteins, localized within specialized nanoscale structural environments. The nano-environments enable the generation of localized electrical and/or chemical gradients, thereby conferring unique functional properties to these units. Thus, the function of a nanodomain is determined by its protein constituents as well as their local structural environment, adding an additional layer of complexity to cardiac biology and biophysics. However, with the emergence of experimental techniques that allow direct investigation of structure and function at the nanoscale, our understanding of cardiac physiology and pathophysiology at these scales is rapidly advancing. Here, we will discuss the structure and functions of multiple cardiomyocyte nanodomains, and novel strategies that target them for the treatment of cardiac arrhythmias.
Collapse
Affiliation(s)
- Przemysław B Radwański
- Bob and Corinne Frick Center for Heart Failure and Arrhythmia, The Ohio State University Wexner Medical Center, Columbus, OH, United States.,Dorothy M. Davis Heart and Lung Research Institute, College of Medicine, The Ohio State University Wexner Medical Center, Columbus, OH, United States.,Department of Physiology and Cell Biology, College of Medicine, The Ohio State University, Columbus, OH, United States.,Division of Pharmacy Practice and Science, College of Pharmacy, The Ohio State University, Columbus, OH, United States
| | - Christopher N Johnson
- Dorothy M. Davis Heart and Lung Research Institute, College of Medicine, The Ohio State University Wexner Medical Center, Columbus, OH, United States.,Vanderbilt Center for Arrhythmia Research and Therapeutics, Nashville, TN, United States
| | - Sándor Györke
- Bob and Corinne Frick Center for Heart Failure and Arrhythmia, The Ohio State University Wexner Medical Center, Columbus, OH, United States.,Dorothy M. Davis Heart and Lung Research Institute, College of Medicine, The Ohio State University Wexner Medical Center, Columbus, OH, United States.,Department of Physiology and Cell Biology, College of Medicine, The Ohio State University, Columbus, OH, United States
| | - Rengasayee Veeraraghavan
- Bob and Corinne Frick Center for Heart Failure and Arrhythmia, The Ohio State University Wexner Medical Center, Columbus, OH, United States.,Dorothy M. Davis Heart and Lung Research Institute, College of Medicine, The Ohio State University Wexner Medical Center, Columbus, OH, United States.,Department of Physiology and Cell Biology, College of Medicine, The Ohio State University, Columbus, OH, United States.,Department of Biomedical Engineering, The Ohio State University, Columbus, OH, United States
| |
Collapse
|
35
|
Tikhonov DB, Zhorov BS. Predicting Structural Details of the Sodium Channel Pore Basing on Animal Toxin Studies. Front Pharmacol 2018; 9:880. [PMID: 30131702 PMCID: PMC6090064 DOI: 10.3389/fphar.2018.00880] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2018] [Accepted: 07/20/2018] [Indexed: 12/25/2022] Open
Abstract
Eukaryotic voltage-gated sodium channels play key roles in physiology and are targets for many toxins and medically important drugs. Physiology, pharmacology, and general architecture of the channels has long been the subject of intensive research in academia and industry. In particular, animal toxins such as tetrodotoxin, saxitoxin, and conotoxins have been used as molecular probes of the channel structure. More recently, X-ray structures of potassium and prokaryotic sodium channels allowed elaborating models of the toxin-channel complexes that integrated data from biophysical, electrophysiological, and mutational studies. Atomic level cryo-EM structures of eukaryotic sodium channels, which became available in 2017, show that the selectivity filter structure and other important features of the pore domain have been correctly predicted. This validates further employments of toxins and other small molecules as sensitive probes of fine structural details of ion channels.
Collapse
Affiliation(s)
- Denis B Tikhonov
- Sechenov Institute of Evolutionary Physiology and Biochemistry, Russian Academy of Sciences, Saint Petersburg, Russia
| | - Boris S Zhorov
- Sechenov Institute of Evolutionary Physiology and Biochemistry, Russian Academy of Sciences, Saint Petersburg, Russia.,Department of Biochemistry and Biomedical Sciences, McMaster University, Hamilton, ON, Canada
| |
Collapse
|
36
|
Yang E, Granata D, Eckenhoff RG, Carnevale V, Covarrubias M. Propofol inhibits prokaryotic voltage-gated Na + channels by promoting activation-coupled inactivation. J Gen Physiol 2018; 150:1299-1316. [PMID: 30018038 PMCID: PMC6122921 DOI: 10.1085/jgp.201711924] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2017] [Accepted: 06/12/2018] [Indexed: 12/21/2022] Open
Abstract
Propofol is widely used in the clinic for the induction and maintenance of general anesthesia. As with most general anesthetics, however, our understanding of its mechanism of action remains incomplete. Local and general anesthetics largely inhibit voltage-gated Na+ channels (Navs) by inducing an apparent stabilization of the inactivated state, associated in some instances with pore block. To determine the biophysical and molecular basis of propofol action in Navs, we investigated NaChBac and NavMs, two prokaryotic Navs with distinct voltage dependencies and gating kinetics, by whole-cell patch clamp electrophysiology in the absence and presence of propofol at clinically relevant concentrations (2-10 µM). In both Navs, propofol induced a hyperpolarizing shift of the pre-pulse inactivation curve without any significant effects on recovery from inactivation at strongly hyperpolarized voltages, demonstrating that propofol does not stabilize the inactivated state. Moreover, there was no evidence of fast or slow pore block by propofol in a non-inactivating NaChBac mutant (T220A). Propofol also induced hyperpolarizing shifts of the conductance-voltage relationships with negligible effects on the time constants of deactivation at hyperpolarized voltages, indicating that propofol does not stabilize the open state. Instead, propofol decreases the time constants of macroscopic activation and inactivation. Adopting a kinetic scheme of Nav gating that assumes preferential closed-state recovery from inactivation, a 1.7-fold acceleration of the rate constant of activation and a 1.4-fold acceleration of the rate constant of inactivation were sufficient to reproduce experimental observations with computer simulations. In addition, molecular dynamics simulations and molecular docking suggest that propofol binding involves interactions with gating machinery in the S4-S5 linker and external pore regions. Our findings show that propofol is primarily a positive gating modulator of prokaryotic Navs, which ultimately inhibits the channels by promoting activation-coupled inactivation.
Collapse
Affiliation(s)
- Elaine Yang
- Vickie and Jack Farber Institute for Neuroscience and Department of Neuroscience, Sidney Kimmel Medical College and Jefferson College of Biomedical Sciences, Thomas Jefferson University, Philadelphia, PA
| | - Daniele Granata
- Institute for Computational Molecular Science, College of Science and Technology, Temple University, Philadelphia, PA
| | - Roderic G Eckenhoff
- Department of Anesthesiology and Critical Care, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA
| | - Vincenzo Carnevale
- Institute for Computational Molecular Science, College of Science and Technology, Temple University, Philadelphia, PA
| | - Manuel Covarrubias
- Vickie and Jack Farber Institute for Neuroscience and Department of Neuroscience, Sidney Kimmel Medical College and Jefferson College of Biomedical Sciences, Thomas Jefferson University, Philadelphia, PA
| |
Collapse
|
37
|
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: 1.4] [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
|
38
|
|