1
|
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
|
2
|
de Lima Conceição MR, Teixeira-Fonseca JL, Marques LP, Souza DS, Roman-Campos D. Interaction of the antiarrhythmic drug Amiodarone with the sodium channel Na v1.5 depends on the extracellular pH. Eur J Pharmacol 2023; 960:176127. [PMID: 37858835 DOI: 10.1016/j.ejphar.2023.176127] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2023] [Revised: 09/21/2023] [Accepted: 10/16/2023] [Indexed: 10/21/2023]
Abstract
INTRODUCTION Amiodarone (AMD) is a clinically used drug to treat arrhythmias with significant effect upon the cardiac sodium channel Nav1.5. AMD has a pKa of 6.56, and changes in extracellular pH (pHe) may alter its pharmacological properties. Here we explored how changes in pHe impacts the pharmacological properties of AMD upon human-Nav1.5-sodium-current (INa) and in ex vivo rat hearts. METHODS Embryonic-human-kidney-cells (HEK293) were used to transiently express the human alpha-subunit of NaV1.5 channels and the isolated heart of Wistar rats were used. Patch-Clamp technique was deployed to study INa and for electrocardiogram (ECG) evaluation the ex vivo heart preparation in the Langendorff system was applied. RESULTS The potency of AMD upon peak INa was ∼25x higher in pHe 7.0 when compared to pHe 7.4. Voltage dependence for activation did not differ among all groups. AMD shifted the steady-state inactivation curve to more hyperpolarized potentials, with similar magnitudes for both pHes. The recovery from INa inactivation was delayed in the presence of AMD with similar profile in both pHes. Interestingly, the use-dependent properties of AMD was distinct at pHe 7.0 and 7.4. Finally, AMD was able to change the ex vivo ECG profile, however at pHe 7.0+AMD a larger increase in the RR and QRS duration and in the QT interval when compared to pHe 7.4 was found. CONCLUSIONS The pharmacological properties of AMD upon NaV1.5 and isolated heart preparation depends on the pHe and its use in vivo during extracellular acidosis may cause a distinct biological response in the heart tissue.
Collapse
Affiliation(s)
| | - Jorge Lucas Teixeira-Fonseca
- Laboratory of CardioBiology, Department of Biophysics, Paulista School of Medicine, Federal University of Sao Paulo, Brazil
| | - Leisiane Pereira Marques
- Laboratory of CardioBiology, Department of Biophysics, Paulista School of Medicine, Federal University of Sao Paulo, Brazil
| | - Diego Santos Souza
- Laboratory of CardioBiology, Department of Biophysics, Paulista School of Medicine, Federal University of Sao Paulo, Brazil; Department of Physiology, Federal University of Sergipe, São Cristóvão, Brazil
| | - Danilo Roman-Campos
- Laboratory of CardioBiology, Department of Biophysics, Paulista School of Medicine, Federal University of Sao Paulo, Brazil.
| |
Collapse
|
3
|
Teixeira-Fonseca JL, Santos-Miranda A, Marques ILS, Marques LP, Alcantara F, de Lima Conceição MR, Souza DS, Santana Gondim AN, Roman-Campos D. Eugenol delays the onset of ouabain-induced ventricular cardiac arrhythmias in guinea pigs. Basic Clin Pharmacol Toxicol 2023; 133:565-575. [PMID: 37675641 DOI: 10.1111/bcpt.13941] [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: 04/06/2023] [Revised: 08/22/2023] [Accepted: 09/01/2023] [Indexed: 09/08/2023]
Abstract
Eugenol is an aromatic compound used in the manufacture of medicines, perfumes, cosmetics and as an anaesthetic due to the ability of the drug to block the neuronal isoform of voltage-gated Na+ channels (NaV ). Some arrhythmias are associated with gain of function in the sodium current (INa ) found in cardiomyocytes, and antiarrhythmic sodium channel blockers are commonly used in the clinical practice. This study sought to elucidate the potential mechanisms of eugenol's protection in the arrhythmic model of ouabain-induced arrhythmias in guinea pig heart. Ex vivo arrhythmias were induced using 50 μM of ouabain. The antiarrhythmic properties of eugenol were evaluated in the ex vivo heart preparation and isolated ventricular cardiomyocytes. The compound's effects on cardiac sodium current and action potential using the patch-clamp technique were evaluated. In all, eugenol decreased the ex vivo cardiac arrhythmias induced by ouabain. Furthermore, eugenol showed concentration dependent effect upon peak INa , left-shifted the stationary inactivation curve and delayed the recovery from inactivation of the INa . All these aspects are considered to be antiarrhythmic. Our findings demonstrate that eugenol has antiarrhythmic activity, which may be partially explained by the ability of eugenol to change de biophysical properties of INa of cardiomyocytes.
Collapse
Affiliation(s)
- Jorge Lucas Teixeira-Fonseca
- Laboratório de Cardiobiologia, Departamento de Biofísica, Universidade Federal de São Paulo (UNIFESP), São Paulo, São Paulo, Brazil
| | - Artur Santos-Miranda
- Departamento de Fisiologia, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | | | - Leisiane Pereira Marques
- Laboratório de Cardiobiologia, Departamento de Biofísica, Universidade Federal de São Paulo (UNIFESP), São Paulo, São Paulo, Brazil
| | - Fabiana Alcantara
- Laboratório de Cardiobiologia, Departamento de Biofísica, Universidade Federal de São Paulo (UNIFESP), São Paulo, São Paulo, Brazil
| | - Michael Ramon de Lima Conceição
- Laboratório de Cardiobiologia, Departamento de Biofísica, Universidade Federal de São Paulo (UNIFESP), São Paulo, São Paulo, Brazil
| | - Diego Santos Souza
- Laboratório de Cardiobiologia, Departamento de Biofísica, Universidade Federal de São Paulo (UNIFESP), São Paulo, São Paulo, Brazil
| | - Antonio Nei Santana Gondim
- Laboratório de Biofísica e Farmacologia do Coração, Departamento de Educação (Campus-XII), Universidade do Estado da Bahia (UNEB), Guanambi, Brazil
| | - Danilo Roman-Campos
- Laboratório de Cardiobiologia, Departamento de Biofísica, Universidade Federal de São Paulo (UNIFESP), São Paulo, São Paulo, Brazil
| |
Collapse
|
4
|
Wang D, Long YQ, Sun Y, Zhu YJ, Feng XM, Liu H, Ji FH, Peng K. Opioid-free total intravenous anesthesia for thyroid and parathyroid surgery: Protocol for a randomized, double-blind, controlled trial. Front Med (Lausanne) 2022; 9:939098. [PMID: 36111120 PMCID: PMC9468489 DOI: 10.3389/fmed.2022.939098] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2022] [Accepted: 08/12/2022] [Indexed: 11/13/2022] Open
Abstract
BackgroundOpioid-free anesthesia (OFA) may improve postoperative outcomes by reducing opioid-related adverse effects. This study aims to evaluate the effects of OFA on postoperative nausea and vomiting (PONV), postoperative pain, and 30-day outcomes after thyroid and parathyroid surgery.MethodsThis two-center, randomized, double-blind, controlled trial will include 400 adult patients scheduled for thyroid and parathyroid surgery. Patients will be randomly assigned, 1:1 and stratified by sex and site, to an OFA group (esketamine, lidocaine, and dexmedetomidine) or a control group (opioid-based anesthesia with sufentanil). All patients will receive propofol-based total intravenous anesthesia and PONV prophylaxis with dexamethasone and ondansetron. The primary outcome is the incidence of PONV (defined as experiencing any event of nausea, retching, or vomiting) during the first 48 h postoperatively. The secondary outcomes include the severity of PONV, antiemetic rescue therapy, pain scores at rest and while coughing, need for rescue analgesia, perioperative adverse effects related to anesthetics or analgesics (hypotension, bradycardia, hypertension, tachycardia, desaturation, dizziness, headache, hallucination, and nightmare), time to extubation, length of post-anesthesia care unit stay, length of postoperative hospital stay, patient satisfaction, and a composite of 30-day major adverse events (myocardial infarction, cardiac arrest, cerebrovascular accident, coma, acute renal failure, pulmonary embolism, sepsis, septic shock, deep neck space infection, reintubation, reoperation, blood transfusion, failure to wean off ventilator, and death). Analyses will be performed in the modified intention-to-treat population.DiscussionWe hypothesize that our OFA regimen reduces PONV after thyroid and parathyroid surgery. We will also investigate whether OFA leads to improvements in postoperative pain and major adverse events. Our results will offer evidence for optimizing anesthesia regimens in patients who undergo thyroid and parathyroid surgical procedures.Clinical trial registrationhttp://www.chictr.org.cn, identifier: ChiCTR2200059656.
Collapse
Affiliation(s)
- Dan Wang
- Department of Anesthesiology, First Affiliated Hospital of Soochow University, Suzhou, China
- Institute of Anesthesiology, Soochow University, Suzhou, China
| | - Yu-qin Long
- Department of Anesthesiology, First Affiliated Hospital of Soochow University, Suzhou, China
- Institute of Anesthesiology, Soochow University, Suzhou, China
| | - Yan Sun
- Department of Anesthesiology, First Affiliated Hospital of Soochow University, Suzhou, China
- Institute of Anesthesiology, Soochow University, Suzhou, China
| | - Ya-juan Zhu
- Department of Anesthesiology, First Affiliated Hospital of Soochow University, Suzhou, China
- Institute of Anesthesiology, Soochow University, Suzhou, China
| | - Xiao-mei Feng
- Department of Anesthesiology, University of Utah Health, Salt Lake City, UT, United States
| | - Hong Liu
- Department of Anesthesiology and Pain Medicine, University of California, Sacramento, Sacramento, CA, United States
| | - Fu-hai Ji
- Department of Anesthesiology, First Affiliated Hospital of Soochow University, Suzhou, China
- Institute of Anesthesiology, Soochow University, Suzhou, China
- *Correspondence: Fu-hai Ji
| | - Ke Peng
- Department of Anesthesiology, First Affiliated Hospital of Soochow University, Suzhou, China
- Institute of Anesthesiology, Soochow University, Suzhou, China
- Ke Peng
| |
Collapse
|
5
|
Oung SW, Kremer N, Ben Amara S, Zaidi A, Koslowski T. Protonation and orientation: a computational approach to cocaine diffusion through a model membrane. Phys Chem Chem Phys 2022; 24:14219-14227. [DOI: 10.1039/d2cp01140a] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
We study the diffusion of cocaine through a DMPC lipid bilayer as an example of a protonable, amphiphilic molecule passing a biological membrane. Using classical molecular dynamics simulations, the free...
Collapse
|
6
|
Xiao J, Chen Z, Yu B. A Potential Mechanism of Sodium Channel Mediating the General Anesthesia Induced by Propofol. Front Cell Neurosci 2020; 14:593050. [PMID: 33343303 PMCID: PMC7746837 DOI: 10.3389/fncel.2020.593050] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2020] [Accepted: 11/10/2020] [Indexed: 12/19/2022] Open
Abstract
General anesthesia has revolutionized healthcare over the past 200 years and continues to show advancements. However, many phenomena induced by general anesthetics including paradoxical excitation are still poorly understood. Voltage-gated sodium channels (NaV) were believed to be one of the proteins targeted during general anesthesia. Based on electrophysiological measurements before and after propofol treatments of different concentrations, we mathematically modified the Hodgkin–Huxley sodium channel formulations and constructed a thalamocortical model to investigate the potential roles of NaV. The ion channels of individual neurons were modeled using the Hodgkin–Huxley type equations. The enhancement of propofol-induced GABAa current was simulated by increasing the maximal conductance and the time-constant of decay. Electroencephalogram (EEG) was evaluated as the post-synaptic potential from pyramidal (PY) cells. We found that a left shift in activation of NaV was induced primarily by a low concentration of propofol (0.3–10 μM), while a left shift in inactivation of NaV was induced by an increasing concentration (0.3–30 μM). Mathematical simulation indicated that a left shift of NaV activation produced a Hopf bifurcation, leading to cell oscillations. Left shift of NaV activation around a value of 5.5 mV in the thalamocortical models suppressed normal bursting of thalamocortical (TC) cells by triggering its chaotic oscillations. This led to irregular spiking of PY cells and an increased frequency in EEG readings. This observation suggests a mechanism leading to paradoxical excitation during general anesthesia. While a left shift in inactivation led to light hyperpolarization in individual cells, it inhibited the activity of the thalamocortical model after a certain depth of anesthesia. This finding implies that high doses of propofol inhibit the network partly by accelerating NaV toward inactivation. Additionally, this result explains why the application of sodium channel blockers decreases the requirement for general anesthetics. Our study provides an insight into the roles that NaV plays in the mechanism of general anesthesia. Since the activation and inactivation of NaV are structurally independent, it should be possible to avoid side effects by state-dependent binding to the NaV to achieve precision medicine in the future.
Collapse
Affiliation(s)
- Jinglei Xiao
- Department of Anesthesiology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Zhengguo Chen
- College of Computer, National University of Defence Technology, Changsha, China
| | - Buwei Yu
- Department of Anesthesiology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| |
Collapse
|
7
|
Bajaj S, Ong ST, Chandy KG. Contributions of natural products to ion channel pharmacology. Nat Prod Rep 2020; 37:703-716. [PMID: 32065187 DOI: 10.1039/c9np00056a] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Covering: Up to 2020Ion channels are a vast super-family of membrane proteins that play critical physiological roles in excitable and non-excitable cells. Their biomedical importance makes them valuable and attractive drug targets for neurological, cardiovascular, gastrointestinal and metabolic diseases, and for cancer therapy and immune modulation. Current therapeutics target only a minor subset of ion channels, leaving a large unexploited space within the ion channel field. Natural products harnessed from the almost unlimited and diverse universe of compounds within the bioenvironment have been used to modulate channels for decades. In this review we highlight the impact made by natural products on ion channel pharmacology, specifically on K+, NaV and CaV channels, and use case studies to describe the development of ion channel-modulating drugs from natural sources for the treatment of pain, heart disease and autoimmune diseases.
Collapse
Affiliation(s)
- Saumya Bajaj
- Lee Kong Chian School of Medicine, Nanyang Technological University Singapore, Experimental Medicine Building, 59 Nanyang Drive, 636921, Singapore.
| | | | | |
Collapse
|
8
|
Mulcahy JV, Pajouhesh H, Beckley JT, Delwig A, Bois JD, Hunter JC. Challenges and Opportunities for Therapeutics Targeting the Voltage-Gated Sodium Channel Isoform Na V1.7. J Med Chem 2019; 62:8695-8710. [PMID: 31012583 PMCID: PMC6786914 DOI: 10.1021/acs.jmedchem.8b01906] [Citation(s) in RCA: 57] [Impact Index Per Article: 11.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Voltage-gated sodium ion channel subtype 1.7 (NaV1.7) is a high interest target for the discovery of non-opioid analgesics. Compelling evidence from human genetic data, particularly the finding that persons lacking functional NaV1.7 are insensitive to pain, has spurred considerable effort to develop selective inhibitors of this Na+ ion channel target as analgesic medicines. Recent clinical setbacks and disappointing performance of preclinical compounds in animal pain models, however, have led to skepticism around the potential of selective NaV1.7 inhibitors as human therapeutics. In this Perspective, we discuss the attributes and limitations of recently disclosed investigational drugs targeting NaV1.7 and review evidence that, by better understanding the requirements for selectivity and target engagement, the opportunity to deliver effective analgesic medicines targeting NaV1.7 endures.
Collapse
Affiliation(s)
- John V. Mulcahy
- SiteOne Therapeutics, 280 Utah Ave, Suite 250, South San Francisco, CA 94080
| | - Hassan Pajouhesh
- SiteOne Therapeutics, 280 Utah Ave, Suite 250, South San Francisco, CA 94080
| | - Jacob T. Beckley
- SiteOne Therapeutics, 351 Evergreen Drive, Suite B1, Bozeman, MT 59715
| | - Anton Delwig
- SiteOne Therapeutics, 280 Utah Ave, Suite 250, South San Francisco, CA 94080
| | - J. Du Bois
- Stanford University, Lokey Chemistry and Biology, 337 Campus Drive, Stanford, CA 94305
| | - John C. Hunter
- SiteOne Therapeutics, 280 Utah Ave, Suite 250, South San Francisco, CA 94080
| |
Collapse
|
9
|
Abstract
Balanced general anesthesia, the most common management strategy used in anesthesia care, entails the administration of different drugs together to create the anesthetic state. Anesthesiologists developed this approach to avoid sole reliance on ether for general anesthesia maintenance. Balanced general anesthesia uses less of each drug than if the drug were administered alone, thereby increasing the likelihood of its desired effects and reducing the likelihood of its side effects. To manage nociception intraoperatively and pain postoperatively, the current practice of balanced general anesthesia relies almost exclusively on opioids. While opioids are the most effective antinociceptive agents, they have undesirable side effects. Moreover, overreliance on opioids has contributed to the opioid epidemic in the United States. Spurred by concern of opioid overuse, balanced general anesthesia strategies are now using more agents to create the anesthetic state. Under these approaches, called “multimodal general anesthesia,” the additional drugs may include agents with specific central nervous system targets such as dexmedetomidine and ones with less specific targets, such as magnesium. It is postulated that use of more agents at smaller doses further maximizes desired effects while minimizing side effects. Although this approach appears to maximize the benefit-to-side effect ratio, no rational strategy has been provided for choosing the drug combinations. Nociception induced by surgery is the primary reason for placing a patient in a state of general anesthesia. Hence, any rational strategy should focus on nociception control intraoperatively and pain control postoperatively. In this Special Article, we review the anatomy and physiology of the nociceptive and arousal circuits, and the mechanisms through which commonly used anesthetics and anesthetic adjuncts act in these systems. We propose a rational strategy for multimodal general anesthesia predicated on choosing a combination of agents that act at different targets in the nociceptive system to control nociception intraoperatively and pain postoperatively. Because these agents also decrease arousal, the doses of hypnotics and/or inhaled ethers needed to control unconsciousness are reduced. Effective use of this strategy requires simultaneous monitoring of antinociception and level of unconsciousness. We illustrate the application of this strategy by summarizing anesthetic management for 4 representative surgeries.
Collapse
|
10
|
Iqbal F, Thompson AJ, Riaz S, Pehar M, Rice T, Syed NI. Anesthetics: from modes of action to unconsciousness and neurotoxicity. J Neurophysiol 2019; 122:760-787. [PMID: 31242059 DOI: 10.1152/jn.00210.2019] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Modern anesthetic compounds and advanced monitoring tools have revolutionized the field of medicine, allowing for complex surgical procedures to occur safely and effectively. Faster induction times and quicker recovery periods of current anesthetic agents have also helped reduce health care costs significantly. Moreover, extensive research has allowed for a better understanding of anesthetic modes of action, thus facilitating the development of more effective and safer compounds. Notwithstanding the realization that anesthetics are a prerequisite to all surgical procedures, evidence is emerging to support the notion that exposure of the developing brain to certain anesthetics may impact future brain development and function. Whereas the data in support of this postulate from human studies is equivocal, the vast majority of animal research strongly suggests that anesthetics are indeed cytotoxic at multiple brain structure and function levels. In this review, we first highlight various modes of anesthetic action and then debate the evidence of harm from both basic science and clinical studies perspectives. We present evidence from animal and human studies vis-à-vis the possible detrimental effects of anesthetic agents on both the young developing and the elderly aging brain while discussing potential ways to mitigate these effects. We hope that this review will, on the one hand, invoke debate vis-à-vis the evidence of anesthetic harm in young children and the elderly, and on the other hand, incentivize the search for better and less toxic anesthetic compounds.
Collapse
Affiliation(s)
- Fahad Iqbal
- Vi Riddell Pain Program, Alberta Children's Hospital Research Institute, Hotchkiss Brain Institute, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
| | - Andrew J Thompson
- Vi Riddell Pain Program, Alberta Children's Hospital Research Institute, Hotchkiss Brain Institute, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada.,Department of Neuroscience, Faculty of Science, University of Calgary, Calgary, Alberta, Canada
| | - Saba Riaz
- Vi Riddell Pain Program, Alberta Children's Hospital Research Institute, Hotchkiss Brain Institute, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
| | - Marcus Pehar
- Vi Riddell Pain Program, Alberta Children's Hospital Research Institute, Hotchkiss Brain Institute, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
| | - Tiffany Rice
- Department of Anesthesiology, Perioperative and Pain Medicine, Alberta Children's Hospital, University of Calgary, Calgary, Alberta, Canada
| | - Naweed I Syed
- Vi Riddell Pain Program, Alberta Children's Hospital Research Institute, Hotchkiss Brain Institute, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
| |
Collapse
|
11
|
Bownik A, Ślaska B, Bochra J, Gumieniak K, Gałek K. Procaine penicillin alters swimming behaviour and physiological parameters of Daphnia magna. ENVIRONMENTAL SCIENCE AND POLLUTION RESEARCH INTERNATIONAL 2019; 26:18662-18673. [PMID: 31055748 PMCID: PMC6570677 DOI: 10.1007/s11356-019-05255-2] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/07/2018] [Revised: 03/15/2019] [Accepted: 04/22/2019] [Indexed: 05/23/2023]
Abstract
Procaine penicillin (PP) is a β-lactam antibiotic widely used in human and veterinary medicine. Although PP is detected in surface water, little is known on its effects on aquatic invertebrates. Our aim was to determine the influence of PP on swimming behaviour (track density, swimming speed, turning angle, hopping frequency) and physiological activity (oxygen consumption, heart rate, thoracic limb movement) of a freshwater invertebrate Daphnia magna exposed to PP at concentrations of 11.79 mg/L, 117.9 mg/L and 1179 mg/L for 2 h and 24 h. The results showed no mortality; however, reduction of swimming activity manifested by the decreased track density, swimming speed and turning angle noted in Daphnia exposed to all the concentrations of PP. Increase of oxygen consumption was observed after 2-h exposure; however, decrease of this parameter was found after 24 h. PP also reduced heart rate and thoracic limb movement in a concentration-dependent manner. The results suggest that the antibiotic should not induce mortality; however, it may affect swimming behaviour and physiological parameters of Daphnia magna particularly inhabiting aquaculture facilities with intensive antibiotic treatment. On the basis of the present results, we also suggest higher sensitivity of behavioural and physiological parameters of cladocerans than the commonly used endpoints: mortality or immobilisation and their possible application as a part of early warning systems in monitoring of surface water toxicity.
Collapse
Affiliation(s)
- Adam Bownik
- Institute of Biological Basis of Animal Production, Faculty of Biology, Animal Science and Bioeconomy, University of Life Sciences in Lublin, Akademicka 13 Str, 20-950, Lublin, Poland.
| | - Brygida Ślaska
- Institute of Biological Basis of Animal Production, Faculty of Biology, Animal Science and Bioeconomy, University of Life Sciences in Lublin, Akademicka 13 Str, 20-950, Lublin, Poland
| | - Justyna Bochra
- Institute of Biological Basis of Animal Production, Faculty of Biology, Animal Science and Bioeconomy, University of Life Sciences in Lublin, Akademicka 13 Str, 20-950, Lublin, Poland
| | - Katarzyna Gumieniak
- Institute of Biological Basis of Animal Production, Faculty of Biology, Animal Science and Bioeconomy, University of Life Sciences in Lublin, Akademicka 13 Str, 20-950, Lublin, Poland
| | - Kinga Gałek
- Institute of Biological Basis of Animal Production, Faculty of Biology, Animal Science and Bioeconomy, University of Life Sciences in Lublin, Akademicka 13 Str, 20-950, Lublin, Poland
| |
Collapse
|
12
|
Oliveira JD, Ribeiro LNDM, Rodrigues da Silva GH, Casadei BR, Couto VM, Martinez EF, de Paula E. Sustained Release from Ionic-Gradient Liposomes Significantly Decreases ETIDOCAINE Cytotoxicity. Pharm Res 2018; 35:229. [DOI: 10.1007/s11095-018-2512-4] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2018] [Accepted: 09/27/2018] [Indexed: 12/18/2022]
|
13
|
Zhao L, Yang XX, Yin YQ, Wu H, Kang Y, Lou JS. Acute and chronic effects of taurine magnesium coordination compound on cardiac sodium channel Nav1.5. Mol Med Rep 2017; 16:4259-4264. [PMID: 28765943 DOI: 10.3892/mmr.2017.7117] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2016] [Accepted: 04/12/2017] [Indexed: 11/05/2022] Open
Abstract
It has been previously demonstrated that taurine magnesium coordination compound (TMCC) produces antiarrhythmic effects in vivo. The present study investigated the acute and chronic effect of TMCC on sodium channels in HEK cells stably expressing human cardiac Nav1.5 sodium channels. The current amplitude, activation and inactivation kinetics, recovery time from inactivation, and use‑dependent block of sodium channels were analyzed using the whole‑cell patch clamp technique. Western blotting was used to analyze Nav1.5 expression following chronic TMCC treatment. In HEK cells expressing Nav1.5 channels, TMCC acutely inhibited Na+ currents in a dose‑dependent manner. In addition, acute application of TMCC shifted the activation and inactivation curves, and prolonged the recovery time from inactivation, but did not exhibit a use‑dependent block of Nav1.5. By contrast, chronic TMCC treatment only produced a use‑dependent block of Nav1.5 and downregulated Nav1.5 expression. The results of the present study suggested that TMCC may produce antiarrhythmic actions via acute inhibition of sodium channel currents and chronic downregulation of Nav1.5 expression.
Collapse
Affiliation(s)
- Lin Zhao
- International Medical School, Tianjin Medical University, Tianjin 300070, P.R. China
| | - Xiao-Xu Yang
- Department of Pharmacy, Tianjin Medical University Metabolic Disease Hospital, Tianjin 300070, P.R. China
| | - Yong-Qiang Yin
- Department of Pharmacology, Tianjin Medical University, Tianjin 300070, P.R. China
| | - Hong Wu
- Department of Pharmacology, Tianjin Medical University, Tianjin 300070, P.R. China
| | - Yi Kang
- Department of Pharmacology, Tianjin Medical University, Tianjin 300070, P.R. China
| | - Jian-Shi Lou
- Department of Pharmacology, Tianjin Medical University, Tianjin 300070, P.R. China
| |
Collapse
|
14
|
Sparling BA, Yi S, Able J, Bregman H, DiMauro EF, Foti RS, Gao H, Guzman-Perez A, Huang H, Jarosh M, Kornecook T, Ligutti J, Milgram BC, Moyer BD, Youngblood B, Yu VL, Weiss MM. Discovery and hit-to-lead evaluation of piperazine amides as selective, state-dependent Na V1.7 inhibitors. MEDCHEMCOMM 2017; 8:744-754. [PMID: 30108793 PMCID: PMC6072352 DOI: 10.1039/c6md00578k] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/14/2016] [Accepted: 11/30/2016] [Indexed: 11/21/2022]
Abstract
NaV1.7 is a particularly compelling target for the treatment of pain. Herein, we report the discovery and evaluation of a series of piperazine amides that exhibit state-dependent inhibition of NaV1.7. After demonstrating significant pharmacodynamic activity with early lead compound 14 in a NaV1.7-dependent behavioural mouse model, we systematically established SAR trends throughout each sector of the scaffold. The information gleaned from this modular analysis was then applied additively to quickly access analogues that encompass an optimal balance of properties, including NaV1.7 potency, selectivity over NaV1.5, aqueous solubility, and microsomal stability.
Collapse
Affiliation(s)
- Brian A Sparling
- Department of Medicinal Chemistry , Amgen Inc. , 360 Binney Street , Cambridge , MA 02142 , USA .
| | - S Yi
- Department of Medicinal Chemistry , Amgen Inc. , 360 Binney Street , Cambridge , MA 02142 , USA .
| | - J Able
- Department of Neuroscience , Amgen Inc. , One Amgen Center Drive , Thousand Oaks , CA 91320 , USA
| | - H Bregman
- Department of Medicinal Chemistry , Amgen Inc. , 360 Binney Street , Cambridge , MA 02142 , USA .
| | - Erin F DiMauro
- Department of Medicinal Chemistry , Amgen Inc. , 360 Binney Street , Cambridge , MA 02142 , USA .
| | - R S Foti
- Department of Pharmacokinetics and Drug Metabolism , Amgen Inc. , 360 Binney Street , Cambridge , MA 02142 , USA
| | - H Gao
- Department of Molecular Engineering, Amgen Inc. , 360 Binney Street , Cambridge , MA 02142 , USA
| | - A Guzman-Perez
- Department of Medicinal Chemistry , Amgen Inc. , 360 Binney Street , Cambridge , MA 02142 , USA .
| | - H Huang
- Department of Medicinal Chemistry , Amgen Inc. , 360 Binney Street , Cambridge , MA 02142 , USA .
| | - M Jarosh
- Department of Neuroscience , Amgen Inc. , 360 Binney Street , Cambridge , MA 02142 , USA
| | - T Kornecook
- Department of Neuroscience , Amgen Inc. , One Amgen Center Drive , Thousand Oaks , CA 91320 , USA
| | - J Ligutti
- Department of Neuroscience , Amgen Inc. , One Amgen Center Drive , Thousand Oaks , CA 91320 , USA
| | - B C Milgram
- Department of Medicinal Chemistry , Amgen Inc. , 360 Binney Street , Cambridge , MA 02142 , USA .
| | - B D Moyer
- Department of Neuroscience , Amgen Inc. , One Amgen Center Drive , Thousand Oaks , CA 91320 , USA
| | - B Youngblood
- Department of Neuroscience , Amgen Inc. , One Amgen Center Drive , Thousand Oaks , CA 91320 , USA
| | - V L Yu
- Department of Neuroscience , Amgen Inc. , 360 Binney Street , Cambridge , MA 02142 , USA
| | - M M Weiss
- Department of Medicinal Chemistry , Amgen Inc. , 360 Binney Street , Cambridge , MA 02142 , USA .
| |
Collapse
|
15
|
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
|
16
|
Abstract
Sodium bicarbonate is a well-known antidote for tricyclic antidepressant (TCA) poisoning. It has been used for over half a century to treat toxin-induced sodium channel blockade as evidenced by QRS widening on the electrocardiogram (ECG). The purpose of this review is to describe the literature regarding electrophysiological mechanisms and clinical use of this antidote after poisoning by tricyclic antidepressants and other agents. This article will also address the literature supporting an increased serum sodium concentration, alkalemia, or the combination of both as the responsible mechanism(s) for sodium bicarbonate's antidotal properties. While sodium bicarbonate has been used as a treatment for cardiac sodium channel blockade for multiple other agents including citalopram, cocaine, flecainide, diphenhydramine, propoxyphene, and lamotrigine, it has uncertain efficacy with bupropion, propranolol, and taxine-containing plants.
Collapse
|
17
|
Pal K, Gangopadhyay G. Probing kinetic drug binding mechanism in voltage-gated sodium ion channel: open state versus inactive state blockers. Channels (Austin) 2016; 9:307-16. [PMID: 26274618 DOI: 10.1080/19336950.2015.1078950] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
The kinetics and nonequilibrium thermodynamics of open state and inactive state drug binding mechanisms have been studied here using different voltage protocols in sodium ion channel. We have found that for constant voltage protocol, open state block is more efficient in blocking ionic current than inactive state block. Kinetic effect comes through peak current for mexiletine as an open state blocker and in the tail part for lidocaine as an inactive state blocker. Although the inactivation of sodium channel is a free energy driven process, however, the two different kinds of drug affect the inactivation process in a different way as seen from thermodynamic analysis. In presence of open state drug block, the process initially for a long time remains entropy driven and then becomes free energy driven. However in presence of inactive state block, the process remains entirely entropy driven until the equilibrium is attained. For oscillating voltage protocol, the inactive state blocking is more efficient in damping the oscillation of ionic current. From the pulse train analysis it is found that inactive state blocking is less effective in restoring normal repolarisation and blocks peak ionic current. Pulse train protocol also shows that all the inactive states behave differently as one inactive state responds instantly to the test pulse in an opposite manner from the other two states.
Collapse
Affiliation(s)
- Krishnendu Pal
- a S.N. Bose National Center for Basic Sciences ; Block-JD; Sector-III; Salt Lake; Kolkata , India
| | - Gautam Gangopadhyay
- a S.N. Bose National Center for Basic Sciences ; Block-JD; Sector-III; Salt Lake; Kolkata , India
| |
Collapse
|
18
|
Vadhanan P, Tripaty DK, Adinarayanan S. Physiological and pharmacologic aspects of peripheral nerve blocks. J Anaesthesiol Clin Pharmacol 2015; 31:384-93. [PMID: 26330722 PMCID: PMC4541190 DOI: 10.4103/0970-9185.161679] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
A successful peripheral nerve block not only involves a proper technique, but also a thorough knowledge and understanding of the physiology of nerve conduction and pharmacology of local anesthetics (LAs). This article focuses on what happens after the block. Pharmacodynamics of LAs, underlying mechanisms of clinically observable phenomena such as differential blockade, tachyphylaxis, C fiber resistance, tonic and phasic blockade and effect of volume and concentration of LAs. Judicious use of additives along with LAs in peripheral nerve blocks can prolong analgesia. An entirely new group of drugs-neurotoxins has shown potential as local anesthetics. Various methods are available now to prolong the duration of peripheral nerve blocks.
Collapse
Affiliation(s)
- Prasanna Vadhanan
- Department of Anaesthesiology, Vinayaka Mission's Medical College and Hospital, Karaikal, Puducherry, India
| | | | - S Adinarayanan
- Department of Jawaharlal Nehru Institute of Postgraduate Medical Education and Research, Puducherry, India
| |
Collapse
|
19
|
Wang ZJ, Tabakoff B, Levinson SR, Heinbockel T. Inhibition of Nav1.7 channels by methyl eugenol as a mechanism underlying its antinociceptive and anesthetic actions. Acta Pharmacol Sin 2015; 36:791-9. [PMID: 26051112 DOI: 10.1038/aps.2015.26] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2014] [Accepted: 02/05/2015] [Indexed: 11/09/2022] Open
Abstract
AIM Methyl eugenol is a major active component extracted from the Chinese herb Asari Radix et Rhizoma, which has been used to treat toothache and other pain. Previous in vivo studies have shown that methyl eugenol has anesthetic and antinociceptive effects. The aim of this study was to determine the possible mechanism underlying its effect on nervous system disorders. METHODS The direct interaction of methyl eugenol with Na(+) channels was explored and characterized using electrophysiological recordings from Nav1.7-transfected CHO cells. RESULTS In whole-cell patch clamp mode, methyl eugenol tonically inhibited peripheral nerve Nav1.7 currents in a concentration- and voltage-dependent manner, with an IC50 of 295 μmol/L at a -100 mV holding potential. Functionally, methyl eugenol preferentially bound to Nav1.7 channels in the inactivated and/or open state, with weaker binding to channels in the resting state. Thus, in the presence of methyl eugenol, Nav1.7 channels exhibited reduced availability for activation in a steady-state inactivation protocol, strong use-dependent inhibition, enhanced binding kinetics, and slow recovery from inactivation compared to untreated channels. An estimation of the affinity of methyl eugenol for the resting and inactivated states of the channel also demonstrated that methyl eugenol preferentially binds to inactivated channels, with a 6.4 times greater affinity compared to channels in the resting state. The failure of inactivated channels to completely recover to control levels at higher concentrations of methyl eugenol implies that the drug may drive more drug-bound, fast-inactivated channels into drug-bound, slow-inactivated channels. CONCLUSION Methyl eugenol is a potential candidate as an effective local anesthetic and analgesic. The antinociceptive and anesthetic effects of methyl eugenol result from the inhibitory action of methyl eugenol on peripheral Na(+) channels.
Collapse
|
20
|
Gintant G. Cardiac Sodium Current (Na v1.5). METHODS AND PRINCIPLES IN MEDICINAL CHEMISTRY 2015. [DOI: 10.1002/9783527673643.ch12] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
|
21
|
Zhang H, Zou B, Du F, Xu K, Li M. Reporting sodium channel activity using calcium flux: pharmacological promiscuity of cardiac Nav1.5. Mol Pharmacol 2014; 87:207-17. [PMID: 25422141 DOI: 10.1124/mol.114.094789] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022] Open
Abstract
Voltage-gated sodium (Nav) channels are essential for membrane excitability and represent therapeutic targets for treating human diseases. Recent reports suggest that these channels, e.g., Nav1.3 and Nav1.5, are inhibited by multiple structurally distinctive small molecule drugs. These studies give reason to wonder whether these drugs collectively target a single site or multiple sites in manifesting such pharmacological promiscuity. We thus investigate the pharmacological profile of Nav1.5 through systemic analysis of its sensitivity to diverse compound collections. Here, we report a dual-color fluorescent method that exploits a customized Nav1.5 [calcium permeable Nav channel, subtype 5 (SoCal5)] with engineered-enhanced calcium permeability. SoCal5 retains wild-type (WT) Nav1.5 pharmacological profiles. WT SoCal5 and SoCal5 with the local anesthetics binding site mutated (F1760A) could be expressed in separate cells, each with a different-colored genetically encoded calcium sensor, which allows a simultaneous report of compound activity and site dependence. The pharmacological profile of SoCal5 reveals a hit rate (>50% inhibition) of around 13% at 10 μM, comparable to that of hERG. The channel activity is susceptible to blockage by known drugs and structurally diverse compounds. The broad inhibition profile is highly dependent on the F1760 residue in the inner cavity, which is a residue conserved among all nine subtypes of Nav channels. Both promiscuity and dependence on F1760 seen in Nav1.5 were replicated in Nav1.4. Our evidence of a broad inhibition profile of Nav channels suggests a need to consider off-target effects on Nav channels. The site-dependent promiscuity forms a foundation to better understand Nav channels and compound interactions.
Collapse
Affiliation(s)
- Hongkang Zhang
- The Solomon H. Snyder Department of Neuroscience, High Throughput Biology Center (H.Z., B.Z., F.D., K.X., M.L.); Johns Hopkins Ion Channel Center (H.Z., B.Z., F.D., K.X., M.L.), Johns Hopkins University, Baltimore, Maryland; and GlaxoSmithKline, King of Prussia, Pennsylvania (M.L.)
| | - Beiyan Zou
- The Solomon H. Snyder Department of Neuroscience, High Throughput Biology Center (H.Z., B.Z., F.D., K.X., M.L.); Johns Hopkins Ion Channel Center (H.Z., B.Z., F.D., K.X., M.L.), Johns Hopkins University, Baltimore, Maryland; and GlaxoSmithKline, King of Prussia, Pennsylvania (M.L.)
| | - Fang Du
- The Solomon H. Snyder Department of Neuroscience, High Throughput Biology Center (H.Z., B.Z., F.D., K.X., M.L.); Johns Hopkins Ion Channel Center (H.Z., B.Z., F.D., K.X., M.L.), Johns Hopkins University, Baltimore, Maryland; and GlaxoSmithKline, King of Prussia, Pennsylvania (M.L.)
| | - Kaiping Xu
- The Solomon H. Snyder Department of Neuroscience, High Throughput Biology Center (H.Z., B.Z., F.D., K.X., M.L.); Johns Hopkins Ion Channel Center (H.Z., B.Z., F.D., K.X., M.L.), Johns Hopkins University, Baltimore, Maryland; and GlaxoSmithKline, King of Prussia, Pennsylvania (M.L.)
| | - Min Li
- The Solomon H. Snyder Department of Neuroscience, High Throughput Biology Center (H.Z., B.Z., F.D., K.X., M.L.); Johns Hopkins Ion Channel Center (H.Z., B.Z., F.D., K.X., M.L.), Johns Hopkins University, Baltimore, Maryland; and GlaxoSmithKline, King of Prussia, Pennsylvania (M.L.).
| |
Collapse
|
22
|
Baptista-Hon D, Robertson F, Robertson G, Owen S, Rogers G, Lydon E, Lee N, Hales T. Potent inhibition by ropivacaine of metastatic colon cancer SW620 cell invasion and Na V 1.5 channel function. Br J Anaesth 2014; 113 Suppl 1:i39-i48. [DOI: 10.1093/bja/aeu104] [Citation(s) in RCA: 100] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/30/2023] Open
|
23
|
Altered sodium channel gating as molecular basis for pain: contribution of activation, inactivation, and resurgent currents. Handb Exp Pharmacol 2014; 221:91-110. [PMID: 24737233 DOI: 10.1007/978-3-642-41588-3_5] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
Mutations in voltage-gated sodium channels, especially Nav1.7, can cause the genetic pain syndromes inherited erythromelalgia, small fiber neuropathy, paroxysmal extreme pain disorder, and chronic insensitivity to pain. Functional analysis of these mutations offers the possibility of understanding the potential pathomechanisms of these disease patterns and also may help to explicate the molecular mechanisms underlying pain in normal conditions. The mutations are distributed over the whole channel protein, but nevertheless induce similar changes for each pain syndrome. In this review we focus on their impact on sodium channel gating, which may be conferred via modulation of (1) conformation (affecting all gating characteristics); (2) the amount of voltage-sensing charges (affecting mainly activation); (3) interaction within the protein (e.g., binding of the inactivation linker); and (4) interaction with other proteins (e.g., for generation of resurgent currents). Understanding the molecular basis for each gating mode and its impact on cellular excitability and nociception in each disease type may provide a basis for development of more specific and effective therapeutic tools.
Collapse
|