1
|
Martina M, Banderali U, Yogi A, Arbabi Ghahroudi M, Liu H, Sulea T, Durocher Y, Hussack G, van Faassen H, Chakravarty B, Liu QY, Iqbal U, Ling B, Lessard E, Sheff J, Robotham A, Callaghan D, Moreno M, Comas T, Ly D, Stanimirovic D. A Novel Antigen Design Strategy to Isolate Single-Domain Antibodies that Target Human Nav1.7 and Reduce Pain in Animal Models. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2405432. [PMID: 39206821 PMCID: PMC11516162 DOI: 10.1002/advs.202405432] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/17/2024] [Revised: 08/01/2024] [Indexed: 09/04/2024]
Abstract
Genetic studies have identified the voltage-gated sodium channel 1.7 (Nav1.7) as pain target. Due to the ineffectiveness of small molecules and monoclonal antibodies as therapeutics for pain, single-domain antibodies (VHHs) are developed against the human Nav1.7 (hNav1.7) using a novel antigen presentation strategy. A 70 amino-acid peptide from the hNav1.7 protein is identified as a target antigen. A recombinant version of this peptide is grafted into the complementarity determining region 3 (CDR3) loop of an inert VHH in order to maintain the native 3D conformation of the peptide. This antigen is used to isolate one VHH able to i) bind hNav1.7, ii) slow the deactivation of hNav1.7, iii) reduce the ability of eliciting action potentials in nociceptors, and iv) reverse hyperalgesia in in vivo rat and mouse models. This VHH exhibits the potential to be developed as a therapeutic capable of suppressing pain. This novel antigen presentation strategy can be applied to develop biologics against other difficult targets such as ion channels, transporters and GPCRs.
Collapse
Affiliation(s)
- Marzia Martina
- Human Health Therapeutics Research CenterNational Research Council Canada1200 Montreal Road, Building M54OttawaONK1A 0R6Canada
| | - Umberto Banderali
- Human Health Therapeutics Research CenterNational Research Council Canada1200 Montreal Road, Building M54OttawaONK1A 0R6Canada
| | - Alvaro Yogi
- Human Health Therapeutics Research CenterNational Research Council Canada1200 Montreal Road, Building M54OttawaONK1A 0R6Canada
| | - Mehdi Arbabi Ghahroudi
- Human Health Therapeutics Research CentreNational Research Council Canada100 Sussex DriveOttawaONK1N 5A2Canada
| | - Hong Liu
- Human Health Therapeutics Research CenterNational Research Council Canada1200 Montreal Road, Building M54OttawaONK1A 0R6Canada
| | - Traian Sulea
- Human Health Therapeutics Research CentreNational Research Council Canada6100 Royalmount Avenue MontréalQuebecH4P 2R2Canada
| | - Yves Durocher
- Human Health Therapeutics Research CentreNational Research Council Canada6100 Royalmount Avenue MontréalQuebecH4P 2R2Canada
| | - Greg Hussack
- Human Health Therapeutics Research CentreNational Research Council Canada100 Sussex DriveOttawaONK1N 5A2Canada
| | - Henk van Faassen
- Human Health Therapeutics Research CentreNational Research Council Canada100 Sussex DriveOttawaONK1N 5A2Canada
| | - Balu Chakravarty
- Human Health Therapeutics Research CenterNational Research Council Canada1200 Montreal Road, Building M54OttawaONK1A 0R6Canada
| | - Qing Yan Liu
- Human Health Therapeutics Research CenterNational Research Council Canada1200 Montreal Road, Building M54OttawaONK1A 0R6Canada
| | - Umar Iqbal
- Human Health Therapeutics Research CenterNational Research Council Canada1200 Montreal Road, Building M54OttawaONK1A 0R6Canada
| | - Binbing Ling
- Human Health Therapeutics Research CenterNational Research Council Canada1200 Montreal Road, Building M54OttawaONK1A 0R6Canada
| | - Etienne Lessard
- Human Health Therapeutics Research CentreNational Research Council Canada6100 Royalmount Avenue MontréalQuebecH4P 2R2Canada
| | - Joey Sheff
- Human Health Therapeutics Research CentreNational Research Council Canada100 Sussex DriveOttawaONK1N 5A2Canada
| | - Anna Robotham
- Human Health Therapeutics Research CentreNational Research Council Canada100 Sussex DriveOttawaONK1N 5A2Canada
| | - Debbie Callaghan
- Human Health Therapeutics Research CentreNational Research Council Canada100 Sussex DriveOttawaONK1N 5A2Canada
| | - Maria Moreno
- Human Health Therapeutics Research CenterNational Research Council Canada1200 Montreal Road, Building M54OttawaONK1A 0R6Canada
| | - Tanya Comas
- Human Health Therapeutics Research CenterNational Research Council Canada1200 Montreal Road, Building M54OttawaONK1A 0R6Canada
| | - Dao Ly
- Human Health Therapeutics Research CenterNational Research Council Canada1200 Montreal Road, Building M54OttawaONK1A 0R6Canada
| | - Danica Stanimirovic
- Human Health Therapeutics Research CenterNational Research Council Canada1200 Montreal Road, Building M54OttawaONK1A 0R6Canada
| |
Collapse
|
2
|
Sunaga M, Tsuboi Y, Kaizu A, Shinoda M. Role of macrophages in trigeminal ganglia in ectopic orofacial pain associated with pulpitis. J Oral Biosci 2024; 66:145-150. [PMID: 38342297 DOI: 10.1016/j.job.2024.02.001] [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: 11/27/2023] [Revised: 01/31/2024] [Accepted: 02/01/2024] [Indexed: 02/13/2024]
Abstract
OBJECTIVES This study aimed to elucidate the role of macrophages in the trigeminal ganglia (TG) in developing pulpitis-associated ectopic orofacial pain. METHODS Rats underwent maxillary pulp exposure, and Fluoro-Gold (FG) was administered in the ipsilateral whisker pad (WP). Head withdrawal threshold (HWT) upon mechanical stimulation of the WP was recorded, and liposomal clodronate clophosome-A (LCCA; macrophage depletion agent) was administered to the TG at three and four days after pulp exposure. Immunohistochemically, TG sections were stained with anti-Iba1 (a macrophage marker) and anti-Nav1.7 antibodies. RESULTS Pulp exposure decreased HWT and increased the number of Iba1-IR cells near FG-labelled TG neurons. LCCA inhibited the decrease in HWT and stopped the increase of FG-labelled Nav1.7-IR TG neurons in the pulpitis group. CONCLUSIONS Activation of macrophages by pulpitis induces the overexpression of Nav1.7 in TG neurons receiving inputs from WP, resulting in pulpitis-induced ectopic facial mechanical allodynia.
Collapse
Affiliation(s)
- Miki Sunaga
- Department of Physiology, Nihon University School of Dentistry, Tokyo, Japan
| | - Yoshiyuki Tsuboi
- Department of Physiology, Nihon University School of Dentistry, Tokyo, Japan.
| | - Akihiro Kaizu
- Department of Physiology, Nihon University School of Dentistry, Tokyo, Japan
| | - Masamichi Shinoda
- Department of Physiology, Nihon University School of Dentistry, Tokyo, Japan
| |
Collapse
|
3
|
Banderali U, Moreno M, Martina M. The elusive Na v1.7: From pain to cancer. CURRENT TOPICS IN MEMBRANES 2023; 92:47-69. [PMID: 38007269 DOI: 10.1016/bs.ctm.2023.09.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/27/2023]
Abstract
Voltage-gated sodium channels (Nav) are protein complexes that play fundamental roles in the transmission of signals in the nervous system, at the neuromuscular junction and in the heart. They are mainly present in excitable cells where they are responsible for triggering action potentials. Dysfunctions in Nav ion conduction give rise to a wide range of conditions, including neurological disorders, hypertension, arrhythmia, pain and cancer. Nav family 1 is composed of nine members, named numerically from 1 to 9. A Nax family also exists and is involved in body-fluid homeostasis. Of particular interest is Nav1.7 which is highly expressed in the sensory neurons of the dorsal root ganglions, where it is involved in the propagation of pain sensation. Gain-of-function mutations in Nav1.7 cause pathologies associated with increased pain sensitivity, while loss-of-function mutations cause reduced sensitivity to pain. The last decade has seen considerable effort in developing highly specific Nav1.7 blockers as pain medications, nonetheless, sufficient efficacy has yet to be achieved. Evidence is now conclusively showing that Navs are also present in many types of cancer cells, where they are involved in cell migration and invasiveness. Nav1.7 is anomalously expressed in endometrial, ovarian and lung cancers. Nav1.7 is also involved in Chemotherapy Induced Peripheral Neuropathy (CIPN). We propose that the knowledge and tools developed to study the role of Nav1.7 in pain can be exploited to develop novel cancer therapies. In this chapter, we illustrate the various aspects of Nav1.7 function in pain, cancer and CIPN, and outline therapeutic approaches.
Collapse
Affiliation(s)
- Umberto Banderali
- Human Health Therapeutics Research Centre, National Research Council of Canada, Montreal road, Ottawa, ON, Canada.
| | - Maria Moreno
- Human Health Therapeutics Research Centre, National Research Council of Canada, Montreal road, Ottawa, ON, Canada
| | - Marzia Martina
- Human Health Therapeutics Research Centre, National Research Council of Canada, Montreal road, Ottawa, ON, Canada
| |
Collapse
|
4
|
Szczerbowska-Boruchowska M, Piana K, Surowka AD, Czyzycki M, Wrobel P, Szymkowski M, Ziomber-Lisiak A. A combined X-ray fluorescence and infrared microspectroscopy study for new insights into elemental-biomolecular obesity-induced changes in rat brain structures. SPECTROCHIMICA ACTA. PART A, MOLECULAR AND BIOMOLECULAR SPECTROSCOPY 2023; 293:122478. [PMID: 36801735 DOI: 10.1016/j.saa.2023.122478] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/28/2022] [Revised: 01/28/2023] [Accepted: 02/07/2023] [Indexed: 06/18/2023]
Abstract
The objective of our research was to determine the brain changes at the molecular and elemental levels typical of early-stage obesity. Therefore a combined approach using Fourier transform infrared micro-spectroscopy (FTIR-MS) and synchrotron radiation induced X-ray fluorescence (SRXRF) was introduced to evaluate some brain macromolecular and elemental parameters in high-calorie diet (HCD)- induced obese rats (OB, n = 6) and in their lean counterparts (L, n = 6). A HCD was found to alter the lipid- and protein- related structure and elemental composition of the certain brain areas important for energy homeostasis. The increased lipid unsaturation in the frontal cortex and ventral tegmental area, the increased fatty acyl chain length in the lateral hypothalamus and substantia nigra as well as the decreased both protein α helix to protein β- sheet ratio and the percentage fraction of β-turns and β-sheets in the nucleus accumbens were revealed in the OB group reflecting obesity-related brain biomolecular aberrations. In addition, the certain brain elements including P, K and Ca were found to differentiate the lean and obese groups at the best extent. We can conclude that HCD-induced obesity triggers lipid- and protein- related structural changes as well as elemental redistribution within various brain structures important for energy homeostasis. In addition, an approach applying combined X-ray and infrared spectroscopy was shown to be a reliable tool for identifying elemental-biomolecular rat brain changes for better understanding the interplay between the chemical and structural processes involved in appetite control.
Collapse
Affiliation(s)
| | - Kaja Piana
- AGH University of Science and Technology, Faculty of Physics and Applied Computer Science, Al. A. Mickiewicza 30, 30-059 Krakow, Poland
| | - Artur D Surowka
- Elettra-Sincrotrone Trieste SCpA, SS 14, km 163.5, Basovizza, TS 34149 Trieste, Italy
| | - Mateusz Czyzycki
- AGH University of Science and Technology, Faculty of Physics and Applied Computer Science, Al. A. Mickiewicza 30, 30-059 Krakow, Poland; Karlsruhe Institute of Technology, Institute for Photon Science and Synchrotron Radiation, Hermann-von-Helmholtz-Platz 1, 76344 Eggenstein-Leopoldshafen, Germany; International Atomic Energy Agency, Nuclear Science and Instrumentation Laboratory, Friedensstrasse 1, 2444 Seibersdorf, Austria
| | - Pawel Wrobel
- AGH University of Science and Technology, Faculty of Physics and Applied Computer Science, Al. A. Mickiewicza 30, 30-059 Krakow, Poland
| | - Maciej Szymkowski
- Bialystok University of Technology, Faculty of Computer Science, ul. Wiejska 45A, 15-351 Białystok, Poland
| | - Agata Ziomber-Lisiak
- Chair of Pathophysiology, Faculty of Medicine, Jagiellonian University Medical College, ul. Czysta 18, 31-121 Krakow, Poland
| |
Collapse
|
5
|
Xin L, Liang Y, Yang S, Jiang F, Yu F, Zhang M, Chang W, Wang W, Yu C, Liu G, Lu Y. Simple and fast determination of tetrodotoxin in human plasma based on hydrophilic-interaction/ion-exchange mixed-mode solid phase extraction combined with liquid chromatography-tandem mass spectroscopy. J Chromatogr A 2022; 1684:463567. [DOI: 10.1016/j.chroma.2022.463567] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2022] [Revised: 10/11/2022] [Accepted: 10/12/2022] [Indexed: 11/30/2022]
|
6
|
Proteomic profiling reveals neuronal ion channel dysregulation and cellular responses to DNA damage-induced cell cycle arrest and senescence in human neuroblastoma SH-SY5Y cells exposed to cypermethrin. Neurotoxicology 2022; 93:71-83. [PMID: 36063984 DOI: 10.1016/j.neuro.2022.08.015] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2022] [Revised: 07/27/2022] [Accepted: 08/28/2022] [Indexed: 11/20/2022]
Abstract
Cypermethrin (CYP), a synthetic pyrethroid of class II, is widely used as a pesticide worldwide. The primary target of cypermethrin is a voltage-gated sodium channel. The neurotoxicity of CYP has been extensively studied in terms of affecting neuronal development, increasing cellular oxidative stress, and apoptosis. However, little is known about how it affects the expression of channel proteins involved in synaptic transmission, as well as the effects of cypermethrin on DNA damage and cell cycle processes. We found that the ligand and voltage-gated calcium channels and proteins involved in synaptic transmission including NMDA 1 receptor subunit, alpha 1A-voltage-dependent calcium channel, synaptotagmin-17, and synaptojanin-2 were downregulated in CYP-treated cells. After 48h of CYP exposure, cell viability was reduced with flattened and enlarged morphology. The levels of 23 proteins regulating cell cycle processes were altered in CYP-treated cells, according to a proteomic study. The cell cycle analysis showed elevated G0/G1 cell cycle arrest and DNA fragmentation at the sub-G0 stage after CYP exposure. CYP treatment also increased senescence-associated β-galactosidase positive cells, DNA damage, and apoptotic markers. Taken together, the current study showed that cypermethrin exposure caused DNA damage and hastened cellular senescence and apoptosis via disrupting cell cycle regulation. In addition, despite its primary target sodium channel, CYP might cause synaptic dysfunction via the downregulation of synaptic proteins and dysregulation of synapse-associated ion channels.
Collapse
|
7
|
Ngum NM, Aziz MYA, Latif ML, Wall RJ, Duce IR, Mellor IR. Non-canonical endogenous expression of voltage-gated sodium channel NaV1.7 subtype by the TE671 rhabdomyosarcoma cell line. J Physiol 2022; 600:2499-2513. [PMID: 35413129 PMCID: PMC9325523 DOI: 10.1113/jp283055] [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: 03/07/2022] [Accepted: 04/05/2022] [Indexed: 11/29/2022] Open
Abstract
Abstract The human TE671 cell line was originally used as a model of medulloblastoma but has since been reassigned as rhabdomyosarcoma. Despite the characterised endogenous expression of voltage‐sensitive sodium currents in these cells, the specific voltage‐gated sodium channel (VGSC) subtype underlying these currents remains unknown. To profile the VGSC subtype in undifferentiated TE671 cells, endpoint and quantitative reverse transcription–PCR (qRT‐PCR), western blot and whole‐cell patch clamp electrophysiology were performed. qRT‐PCR profiling revealed that expression of the SCN9A gene was ∼215‐fold greater than the SCN4A gene and over 400‐fold greater than any of the other VGSC genes, while western blot confirmed that the dominant SCN9A RNA was translated to a protein with a molecular mass of ∼250 kDa. Elicited sodium currents had a mean amplitude of 2.6 ± 0.7 nA with activation and fast inactivation V50 values of −31.9 ± 1.1 and −69.6 ± 1.0 mV, respectively. The currents were completely and reversibly blocked by tetrodotoxin at concentrations greater than 100 nm (IC50 = 22.3 nm). They were also very susceptible to the NaV1.7 specific blockers Huwentoxin‐IV and Protoxin‐II with IC50 values of 14.6 nm and 0.8 nm, respectively, characteristic of those previously determined for NaV1.7. Combined, the results revealed the non‐canonical and highly dominant expression of NaV1.7 in the human TE671 rhabdomyosarcoma cell line. We show that the TE671 cell line is an easy to maintain and cost‐effective model for the study of NaV1.7, a major target for the development of analgesic drugs and more generally for the study of pain. Key points Undifferentiated TE671 cells produce a voltage‐sensitive sodium current when depolarised. The voltage‐gated sodium channel isoform expressed in undifferentiated TE671 cells was previously unknown.
Through qRT‐PCR, western blot and toxin pharmacology, it is shown that undifferentiated TE671 cells dominantly (>99.5%) express the NaV1.7 isoform that is strongly associated with pain.
The TE671 cell line is, therefore, a very easy to maintain and cost‐effective model to study NaV1.7‐targeting drugs.
Collapse
Affiliation(s)
- Neville M Ngum
- School of Life Sciences, University of Nottingham, University Park, Nottingham, NG7 2RD, UK
| | - Muhammad Y A Aziz
- School of Life Sciences, University of Nottingham, University Park, Nottingham, NG7 2RD, UK
| | - M Liaque Latif
- School of Life Sciences, University of Nottingham, University Park, Nottingham, NG7 2RD, UK
| | - Richard J Wall
- School of Life Sciences, University of Nottingham, University Park, Nottingham, NG7 2RD, UK
| | - Ian R Duce
- School of Life Sciences, University of Nottingham, University Park, Nottingham, NG7 2RD, UK
| | - Ian R Mellor
- School of Life Sciences, University of Nottingham, University Park, Nottingham, NG7 2RD, UK
| |
Collapse
|
8
|
Martínez AL, Brea J, Domínguez E, Varela MJ, Allegue C, Cruz R, Monroy X, Merlos M, Burgueño J, Carracedo Á, Loza MI. Identification of Sodium Transients Through NaV1.5 Channels as Regulators of Differentiation in Immortalized Dorsal Root Ganglia Neurons. Front Cell Neurosci 2022; 16:816325. [PMID: 35465610 PMCID: PMC9018981 DOI: 10.3389/fncel.2022.816325] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2021] [Accepted: 03/07/2022] [Indexed: 11/13/2022] Open
Abstract
Neuronal differentiation is a complex process through which newborn neurons acquire the morphology of mature neurons and become excitable. We employed a combination of functional and transcriptomic approaches to deconvolute and identify key regulators of the differentiation process of a DRG neuron-derived cell line, and we focused our study on the NaV1.5 ion channel (encoded by Scn5a) as a channel involved in the acquisition of DRG neuronal features. Overexpression of Scn5a enhances the acquisition of neuronal phenotypic features and increases the KCl-elicited hyperexcitability response in a DRG-derived cell line. Moreover, pharmacologic inhibition of the NaV1.5 channel during differentiation hinders the acquisition of phenotypic features of neuronal cells and the hyperexcitability increase in response to changes in the extracellular medium ionic composition. Taken together, these data highlight the relevance of sodium transients in regulating the neuronal differentiation process in a DRG neuron-derived cell line.
Collapse
Affiliation(s)
- Antón L. Martínez
- BioFarma Research Group, Centro Singular de Investigación en Medicina Molecular y Enfermedades Crónicas (CIMUS), Universidade de Santiago de Compostela, Santiago de Compostela, Spain
| | - José Brea
- BioFarma Research Group, Centro Singular de Investigación en Medicina Molecular y Enfermedades Crónicas (CIMUS), Universidade de Santiago de Compostela, Santiago de Compostela, Spain
| | - Eduardo Domínguez
- BioFarma Research Group, Centro Singular de Investigación en Medicina Molecular y Enfermedades Crónicas (CIMUS), Universidade de Santiago de Compostela, Santiago de Compostela, Spain
| | - María J. Varela
- BioFarma Research Group, Centro Singular de Investigación en Medicina Molecular y Enfermedades Crónicas (CIMUS), Universidade de Santiago de Compostela, Santiago de Compostela, Spain
| | - Catarina Allegue
- Grupo de Medicina Xenómica, Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER), Universidade de Santiago de Compostela, Santiago de Compostela, Spain
| | - Raquel Cruz
- Grupo de Medicina Xenómica, Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER), Universidade de Santiago de Compostela, Santiago de Compostela, Spain
| | - Xavier Monroy
- WeLab Barcelona, Parc Científic de Barcelona, Barcelona, Spain
| | - Manuel Merlos
- WeLab Barcelona, Parc Científic de Barcelona, Barcelona, Spain
| | - Javier Burgueño
- WeLab Barcelona, Parc Científic de Barcelona, Barcelona, Spain
- *Correspondence: Javier Burgueño,
| | - Ángel Carracedo
- Grupo de Medicina Xenómica, Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER), Universidade de Santiago de Compostela, Santiago de Compostela, Spain
- Fundación Pública Galega de Medicina Xenómica, Instituto de Investigación Sanitaria de Santiago de Compostela (IDIS), SERGAS, Santiago de Compostela, Spain
| | - María Isabel Loza
- BioFarma Research Group, Centro Singular de Investigación en Medicina Molecular y Enfermedades Crónicas (CIMUS), Universidade de Santiago de Compostela, Santiago de Compostela, Spain
- María Isabel Loza,
| |
Collapse
|
9
|
Bhandari R, Sharma A, Kuhad A. Novel Nanotechnological Approaches for Targeting Dorsal Root Ganglion (DRG) in Mitigating Diabetic Neuropathic Pain (DNP). Front Endocrinol (Lausanne) 2022; 12:790747. [PMID: 35211091 PMCID: PMC8862660 DOI: 10.3389/fendo.2021.790747] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/07/2021] [Accepted: 12/17/2021] [Indexed: 11/28/2022] Open
Abstract
Diabetic neuropathy is the most entrenched complication of diabetes. Usually, it affects the distal foot and toes, which then gradually approaches the lower part of the legs. Diabetic foot ulcer (DFU) could be one of the worst complications of diabetes mellitus. Long-term diabetes leads to hyperglycemia, which is the utmost contributor to neuropathic pain. Hyperglycemia causing an upregulation of voltage-gated sodium channels in the dorsal root ganglion (DRG) was often observed in models of neuropathic pain. DRG opening frequency increases intracellular sodium ion levels, which further causes increased calcium channel opening and stimulates other pathways leading to diabetic peripheral neuropathy (DPN). Currently, pain due to diabetic neuropathy is managed via antidepressants, opioids, gamma-aminobutyric acid (GABA) analogs, and topical agents such as capsaicin. Despite the availability of various treatment strategies, the percentage of patients achieving adequate pain relief remains low. Many factors contribute to this condition, such as lack of specificity and adverse effects such as light-headedness, languidness, and multiple daily doses. Therefore, nanotechnology outperforms in every aspect, providing several benefits compared to traditional therapy such as site-specific and targeted drug delivery. Nanotechnology is the branch of science that deals with the development of nanoscale materials and products, even smaller than 100 nm. Carriers can improve their efficacy with reduced side effects by incorporating drugs into the novel delivery systems. Thus, the utilization of nanotechnological approaches such as nanoparticles, polymeric nanoparticles, inorganic nanoparticles, lipid nanoparticles, gene therapy (siRNA and miRNA), and extracellular vesicles can extensively contribute to relieving neuropathic pain.
Collapse
Affiliation(s)
| | | | - Anurag Kuhad
- University Institute of Pharmaceutical Sciences, Panjab University, Chandigarh, India
| |
Collapse
|
10
|
Bucciarelli GM, Lechner M, Fontes A, Kats LB, Eisthen HL, Shaffer HB. From Poison to Promise: The Evolution of Tetrodotoxin and Its Potential as a Therapeutic. Toxins (Basel) 2021; 13:toxins13080517. [PMID: 34437388 PMCID: PMC8402337 DOI: 10.3390/toxins13080517] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2021] [Revised: 07/17/2021] [Accepted: 07/20/2021] [Indexed: 11/16/2022] Open
Abstract
Tetrodotoxin (TTX) is a potent neurotoxin that was first identified in pufferfish but has since been isolated from an array of taxa that host TTX-producing bacteria. However, determining its origin, ecosystem roles, and biomedical applications has challenged researchers for decades. Recognized as a poison and for its lethal effects on humans when ingested, TTX is primarily a powerful sodium channel inhibitor that targets voltage-gated sodium channels, including six of the nine mammalian isoforms. Although lethal doses for humans range from 1.5-2.0 mg TTX (blood level 9 ng/mL), when it is administered at levels far below LD50, TTX exhibits therapeutic properties, especially to treat cancer-related pain, neuropathic pain, and visceral pain. Furthermore, TTX can potentially treat a variety of medical ailments, including heroin and cocaine withdrawal symptoms, spinal cord injuries, brain trauma, and some kinds of tumors. Here, we (i) describe the perplexing evolution and ecology of tetrodotoxin, (ii) review its mechanisms and modes of action, and (iii) offer an overview of the numerous ways it may be applied as a therapeutic. There is much to be explored in these three areas, and we offer ideas for future research that combine evolutionary biology with therapeutics. The TTX system holds great promise as a therapeutic and understanding the origin and chemical ecology of TTX as a poison will only improve its general benefit to humanity.
Collapse
Affiliation(s)
- Gary M. Bucciarelli
- Department of Ecology and Evolutionary Biology & UCLA La Kretz Center for California Conservation Science, Institute of the Environment and Sustainability, University of California, Los Angeles, CA 90095, USA; (M.L.); (H.B.S.)
- Correspondence:
| | - Maren Lechner
- Department of Ecology and Evolutionary Biology & UCLA La Kretz Center for California Conservation Science, Institute of the Environment and Sustainability, University of California, Los Angeles, CA 90095, USA; (M.L.); (H.B.S.)
| | - Audrey Fontes
- Natural Science Division, Pepperdine University, Malibu, CA 90263, USA; (A.F.); (L.B.K.)
| | - Lee B. Kats
- Natural Science Division, Pepperdine University, Malibu, CA 90263, USA; (A.F.); (L.B.K.)
| | - Heather L. Eisthen
- Department of Integrative Biology, Michigan State University, East Lansing, MI 48824, USA;
| | - H. Bradley Shaffer
- Department of Ecology and Evolutionary Biology & UCLA La Kretz Center for California Conservation Science, Institute of the Environment and Sustainability, University of California, Los Angeles, CA 90095, USA; (M.L.); (H.B.S.)
| |
Collapse
|
11
|
Mehboob R, Marchenkova A, van den Maagdenberg AMJM, Nistri A. Overexpressed Na V 1.7 Channels Confer Hyperexcitability to in vitro Trigeminal Sensory Neurons of Ca V 2.1 Mutant Hemiplegic Migraine Mice. Front Cell Neurosci 2021; 15:640709. [PMID: 34113237 PMCID: PMC8185157 DOI: 10.3389/fncel.2021.640709] [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: 12/11/2020] [Accepted: 04/09/2021] [Indexed: 11/21/2022] Open
Abstract
Trigeminal sensory neurons of transgenic knock-in (KI) mice expressing the R192Q missense mutation in the α1A subunit of neuronal voltage-gated CaV2.1 Ca2+ channels, which leads to familial hemiplegic migraine type 1 (FHM1) in patients, exhibit a hyperexcitability phenotype. Here, we show that the expression of NaV1.7 channels, linked to pain states, is upregulated in KI primary cultures of trigeminal ganglia (TG), as shown by increased expression of its α1 subunit. In the majority of TG neurons, NaV1.7 channels are co-expressed with ATP-gated P2X3 receptors (P2X3R), which are important nociceptive sensors. Reversing the trigeminal phenotype with selective CaV2.1 channel inhibitor ω-agatoxin IVA inhibited NaV1.7 overexpression. Functionally, KI neurons revealed a TTX-sensitive inward current of larger amplitude that was partially inhibited by selective NaV1.7 blocker Tp1a. Under current-clamp condition, Tp1a raised the spike threshold of both wild-type (WT) and KI neurons with decreased firing rate in KI cells. NaV1.7 activator OD1 accelerated firing in WT and KI neurons, a phenomenon blocked by Tp1a. Enhanced expression and function of NaV1.7 channels in KI TG neurons resulted in higher excitability and facilitated nociceptive signaling. Co-expression of NaV1.7 channels and P2X3Rs in TGs may explain how hypersensitivity to local stimuli can be relevant to migraine.
Collapse
Affiliation(s)
- Riffat Mehboob
- Department of Neuroscience, International School for Advanced Studies (SISSA), Trieste, Italy.,Research Unit, Faculty of Allied Health Sciences, University of Lahore, Lahore, Pakistan
| | - Anna Marchenkova
- Department of Neuroscience, International School for Advanced Studies (SISSA), Trieste, Italy
| | - Arn M J M van den Maagdenberg
- Department of Neurology, Leiden University Medical Center, Leiden, Netherlands.,Department of Human Genetics, University Medical Center, Leiden, Netherlands
| | - Andrea Nistri
- Department of Neuroscience, International School for Advanced Studies (SISSA), Trieste, Italy
| |
Collapse
|
12
|
Loser D, Schaefer J, Danker T, Möller C, Brüll M, Suciu I, Ückert AK, Klima S, Leist M, Kraushaar U. Human neuronal signaling and communication assays to assess functional neurotoxicity. Arch Toxicol 2021; 95:229-252. [PMID: 33269408 PMCID: PMC7811517 DOI: 10.1007/s00204-020-02956-3] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2020] [Accepted: 11/16/2020] [Indexed: 01/08/2023]
Abstract
Prediction of drug toxicity on the human nervous system still relies mainly on animal experiments. Here, we developed an alternative system allowing assessment of complex signaling in both individual human neurons and on the network level. The LUHMES cultures used for our approach can be cultured in 384-well plates with high reproducibility. We established here high-throughput quantification of free intracellular Ca2+ concentrations [Ca2+]i as broadly applicable surrogate of neuronal activity and verified the main processes by patch clamp recordings. Initially, we characterized the expression pattern of many neuronal signaling components and selected the purinergic receptors to demonstrate the applicability of the [Ca2+]i signals for quantitative characterization of agonist and antagonist responses on classical ionotropic neurotransmitter receptors. This included receptor sub-typing and the characterization of the anti-parasitic drug suramin as modulator of the cellular response to ATP. To exemplify potential studies on ion channels, we characterized voltage-gated sodium channels and their inhibition by tetrodotoxin, saxitoxin and lidocaine, as well as their opening by the plant alkaloid veratridine and the food-relevant marine biotoxin ciguatoxin. Even broader applicability of [Ca2+]i quantification as an end point was demonstrated by measurements of dopamine transporter activity based on the membrane potential-changing activity of this neurotransmitter carrier. The substrates dopamine or amphetamine triggered [Ca2+]i oscillations that were synchronized over the entire culture dish. We identified compounds that modified these oscillations by interfering with various ion channels. Thus, this new test system allows multiple types of neuronal signaling, within and between cells, to be assessed, quantified and characterized for their potential disturbance.
Collapse
Affiliation(s)
- Dominik Loser
- NMI Natural and Medical Sciences Institute at the University of Tuebingen, 72770, Reutlingen, Germany
- NMI TT GmbH, 72770, Reutlingen, Germany
- Life Sciences Faculty, Albstadt-Sigmaringen University, 72488, Sigmaringen, Germany
| | - Jasmin Schaefer
- NMI Natural and Medical Sciences Institute at the University of Tuebingen, 72770, Reutlingen, Germany
- NMI TT GmbH, 72770, Reutlingen, Germany
| | | | - Clemens Möller
- Life Sciences Faculty, Albstadt-Sigmaringen University, 72488, Sigmaringen, Germany
| | - Markus Brüll
- In Vitro Toxicology and Biomedicine, Department Inaugurated by the Doerenkamp-Zbinden Foundation, University of Konstanz, Universitaetsstr. 10, 78457, Constance, Germany
| | - Ilinca Suciu
- In Vitro Toxicology and Biomedicine, Department Inaugurated by the Doerenkamp-Zbinden Foundation, University of Konstanz, Universitaetsstr. 10, 78457, Constance, Germany
| | - Anna-Katharina Ückert
- In Vitro Toxicology and Biomedicine, Department Inaugurated by the Doerenkamp-Zbinden Foundation, University of Konstanz, Universitaetsstr. 10, 78457, Constance, Germany
| | - Stefanie Klima
- In Vitro Toxicology and Biomedicine, Department Inaugurated by the Doerenkamp-Zbinden Foundation, University of Konstanz, Universitaetsstr. 10, 78457, Constance, Germany
| | - Marcel Leist
- In Vitro Toxicology and Biomedicine, Department Inaugurated by the Doerenkamp-Zbinden Foundation, University of Konstanz, Universitaetsstr. 10, 78457, Constance, Germany.
| | - Udo Kraushaar
- NMI Natural and Medical Sciences Institute at the University of Tuebingen, 72770, Reutlingen, Germany
| |
Collapse
|
13
|
Chemometric Models of Differential Amino Acids at the Na vα and Na vβ Interface of Mammalian Sodium Channel Isoforms. Molecules 2020; 25:molecules25153551. [PMID: 32756517 PMCID: PMC7435598 DOI: 10.3390/molecules25153551] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2020] [Revised: 07/21/2020] [Accepted: 07/22/2020] [Indexed: 12/19/2022] Open
Abstract
(1) Background: voltage-gated sodium channels (Navs) are integral membrane proteins that allow the sodium ion flux into the excitable cells and initiate the action potential. They comprise an α (Navα) subunit that forms the channel pore and are coupled to one or more auxiliary β (Navβ) subunits that modulate the gating to a variable extent. (2) Methods: after performing homology in silico modeling for all nine isoforms (Nav1.1α to Nav1.9α), the Navα and Navβ protein-protein interaction (PPI) was analyzed chemometrically based on the primary and secondary structures as well as topological or spatial mapping. (3) Results: our findings reveal a unique isoform-specific correspondence between certain segments of the extracellular loops of the Navα subunits. Precisely, loop S5 in domain I forms part of the PPI and assists Navβ1 or Navβ3 on all nine mammalian isoforms. The implied molecular movements resemble macroscopic springs, all of which explains published voltage sensor effects on sodium channel fast inactivation in gating. (4) Conclusions: currently, the specific functions exerted by the Navβ1 or Navβ3 subunits on the modulation of Navα gating remain unknown. Our work determined functional interaction in the extracellular domains on theoretical grounds and we propose a schematic model of the gating mechanism of fast channel sodium current inactivation by educated guessing.
Collapse
|
14
|
Mechanisms of dynamical complexity changes in patterns of sensory neurons under antinociceptive effect emergence. Neurocomputing 2020. [DOI: 10.1016/j.neucom.2019.10.004] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
|
15
|
Abstract
Essential tremor (ET) is a neurological movement disorder characterised by bilateral limb kinetic/postural tremor, with or without tremor in other body parts including head, voice and lower limbs. Since no causative genes for ET have been identified, it is likely that the disorder occurs as a result of complex genetic factors interacting with various cellular and environmental factors that can result in abnormal function of circuitry involving the cerebello-thalamo-cortical pathway. Genetic analyses have uncovered at least 14 loci and 11 genes that are related to ET, as well as various risk or protective genetic factors. Limitations in ET genetic analyses include inconsistent disease definition, small sample size, varied ethnic backgrounds and many other factors that may contribute to paucity of relevant genetic data in ET. Genetic analyses, coupled with functional and animal studies, have led to better insights into possible pathogenic mechanisms underlying ET. These genetic studies may guide the future development of genetic testing and counselling, and specific, pathogenesis-targeted, therapeutic strategies.
Collapse
|
16
|
µ-TRTX-Ca1a: a novel neurotoxin from Cyriopagopus albostriatus with analgesic effects. Acta Pharmacol Sin 2019; 40:859-866. [PMID: 30382183 DOI: 10.1038/s41401-018-0181-9] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2018] [Accepted: 09/30/2018] [Indexed: 12/16/2022] Open
Abstract
Human genetic and pharmacological studies have demonstrated that voltage-gated sodium channels (VGSCs) are promising therapeutic targets for the treatment of pain. Spider venom contains many toxins that modulate the activity of VGSCs. To date, only 0.01% of such spider toxins has been explored, and thus there is a great potential for discovery of novel VGSC modulators as useful pharmacological tools or potential therapeutics. In the current study, we identified a novel peptide, µ-TRTX-Ca1a (Ca1a), in the venom of the tarantula Cyriopagopus albostriatus. This peptide consisted of 38 residues, including 6 cysteines, i.e. IFECSISCEIEKEGNGKKCKPKKCKGGWKCKFNICVKV. In HEK293T or ND7/23 cells expressing mammalian VGSCs, this peptide exhibited the strongest inhibitory activity on Nav1.7 (IC50 378 nM), followed by Nav1.6 (IC50 547 nM), Nav1.2 (IC50 728 nM), Nav1.3 (IC50 2.2 µM) and Nav1.4 (IC50 3.2 µM), and produced negligible inhibitory effect on Nav1.5, Nav1.8, and Nav1.9, even at high concentrations of up to 10 µM. Furthermore, this peptide did not significantly affect the activation and inactivation of Nav1.7. Using site-directed mutagenesis of Nav1.7 and Nav1.4, we revealed that its binding site was localized to the DIIS3-S4 linker region involving the D816 and E818 residues. In three different mouse models of pain, pretreatment with Cala (100, 200, 500 µg/kg) dose-dependently suppressed the nociceptive responses induced by formalin, acetic acid or heat. These results suggest that Ca1a is a novel neurotoxin against VGSCs and has a potential to be developed as a novel analgesic.
Collapse
|
17
|
Khodashenas M, Baghdadi G, Towhidkhah F. A modified Hodgkin-Huxley model to show the effect of motor cortex stimulation on the trigeminal neuralgia network. JOURNAL OF MATHEMATICAL NEUROSCIENCE 2019; 9:4. [PMID: 31152270 PMCID: PMC6544710 DOI: 10.1186/s13408-019-0072-5] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 11/02/2018] [Accepted: 05/21/2019] [Indexed: 06/09/2023]
Abstract
BACKGROUND Trigeminal neuralgia (TN) is a severe neuropathic pain, which has an electric shock-like characteristic. There are some common treatments for this pain such as medicine, microvascular decompression or radio frequency. In this regard, transcranial direct current stimulation (tDCS) is another therapeutic method to reduce pain, which has been recently attracting the therapists' attention. The positive effect of tDCS on TN was shown in many previous studies. However, the mechanism of the tDCS effect has remained unclear. OBJECTIVE This study aims to model the neuronal behavior of the main known regions of the brain participating in TN pathways to study the effect of transcranial direct current stimulation. METHOD The proposed model consists of several blocks: (1) trigeminal nerve, (2) trigeminal ganglion, (3) PAG (periaqueductal gray in the brainstem), (4) thalamus, (5) motor cortex (M1) and (6) somatosensory cortex (S1). Each of these components is represented by a modified Hodgkin-Huxley (HH) model. The modification of the HH model was done based on some neurological facts of pain sodium channels. The input of the model involves any stimuli to the 'trigeminal nerve,' which cause the pain, and the output is the activity of the somatosensory cortex. An external current, which is considered as an electrical current, was applied to the motor cortex block of the model. RESULT The results showed that by decreasing the conductivity of the slow sodium channels (pain channels) and applying tDCS over the M1, the activity of the somatosensory cortex would be reduced. This reduction can cause pain relief. CONCLUSION The proposed model provided some possible suggestions about the relationship between the effects of tDCS and associated components in TN, and also the relationship between the pain measurement index, somatosensory cortex activity, and the strength of tDCS.
Collapse
Affiliation(s)
| | - Golnaz Baghdadi
- Department of Biomedical Engineering, Amirkabir University of Technology, Tehran, Iran
| | - Farzad Towhidkhah
- Department of Biomedical Engineering, Amirkabir University of Technology, Tehran, Iran
| |
Collapse
|
18
|
Addressing the Issue of Tetrodotoxin Targeting. Mar Drugs 2018; 16:md16100352. [PMID: 30261623 PMCID: PMC6212850 DOI: 10.3390/md16100352] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2018] [Revised: 09/21/2018] [Accepted: 09/24/2018] [Indexed: 12/27/2022] Open
Abstract
This review is devoted to the medical application of tetrodotoxin (TTX), a potent non-protein specific blocker of voltage-gated sodium (NaV) channels. The selectivity of action, lack of affinity with the heart muscle NaV channels, and the inability to penetrate the blood–brain barrier make this toxin an attractive candidate for anesthetic and analgesic drug design. The efficacy of TTX was shown in neuropathic, acute and inflammatory pain models. The main emphasis of the review is on studies focused on the improvement of TTX efficacy and safety in conjunction with additional substances and drug delivery systems. A significant improvement in the effectiveness of the toxin was demonstrated when used in tandem with vasoconstrictors, local anesthetics and chemical permeation enhancers, with the best results obtained with the encapsulation of TTX in microparticles and liposomes conjugated to gold nanorods.
Collapse
|
19
|
Sun W, Ma M, Yu H, Yu H. Inhibition of lncRNA X inactivate-specific transcript ameliorates inflammatory pain by suppressing satellite glial cell activation and inflammation by acting as a sponge of miR-146a to inhibit Na v 1.7. J Cell Biochem 2018; 119:9888-9898. [PMID: 30129228 DOI: 10.1002/jcb.27310] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2018] [Accepted: 06/25/2018] [Indexed: 12/28/2022]
Abstract
Long noncoding RNAs (lncRNA) has been validated to participate in nociception in inflammatory pain, presenting as a potential target against anesthesia. Previous research work confirmed the correlation between lncRNA X inactivate-specific transcript (XIST) and inflammation. However, its role in inflammatory pain is undefined. In animal pain models, voltage-gated sodium channels (VGSCs) reportedly participate in neural excitation. In this study, we observed the high expression of XIST and VGSC 1.7 (Nav 1.7) in the dorsal root ganglion (DRG) of the complete Freund's adjuvant (CFA)-induced rat inflammatory pain model. Furthermore, XIST inhibition alleviated pain behavior and the activation of DRG satellite glial cells by suppressing glial fibrillary acidic protein (GFAP) expression, as well as inflammatory cytokine levels of interleukin-6 and tumor necrosis factor-α. XIST downregulation increased the mechanical pain threshold in an inflammatory pain model. Moreover, the expression of miR-146a was decreased in CFA rats. In vitro, XIST acted as a sponge of miR-146a, which targeted Nav 1.7 via bioinformatic prediction, luciferase reporter, and pull-down assay. More importantly, activation of the Nav 1.7 pathway or miR-146 depression both reversed XIST knockdown-inhibited satellite glial cell activation and inflammatory pain in CFA rats. These results suggest that cessation of XIST may ameliorate inflammatory pain by acting as a sponge of miR-146a to inhibit Nav1.7, implying a promising strategy against inflammatory pain.
Collapse
Affiliation(s)
- Wenbo Sun
- Department of Anesthesia, Cangzhou Central Hospital, CangZhou, China
| | - Meina Ma
- Department of Anesthesia, Cangzhou Central Hospital, CangZhou, China
| | - Hongmei Yu
- Department of Anesthesia, Cangzhou Central Hospital, CangZhou, China
| | - Hong Yu
- Department of Anesthesia, Cangzhou Central Hospital, CangZhou, China
| |
Collapse
|
20
|
Zhou Z, Tang X, Chen H, Wang Y. Comparative studies of saxitoxin (STX) -induced cytotoxicity in Neuro-2a and RTG-2 cell lines: An explanation with respect to changes in ROS. CHEMOSPHERE 2018; 192:66-74. [PMID: 29100123 DOI: 10.1016/j.chemosphere.2017.10.083] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/28/2017] [Revised: 10/06/2017] [Accepted: 10/14/2017] [Indexed: 06/07/2023]
Abstract
Saxitoxin (STX), a paralytic shellfish toxin (PST) produced from toxic bloom-forming dinoflagellates, was selected to comparatively investigate the induction of cytotoxicity and apoptosis and a possible mechanism based on changes in the antioxidant defence system of two cellular strains: the mouse neuroblastoma cell line Neuro-2a and the rainbow trout fish cell line RTG-2. Increasing concentrations of STX (0-256 nM) presented little cytotoxic or apoptotic effects on the two cell lines. Measurements of cellular viability, lethal ratio and LDH leakage showed slight changes in Neuro-2a and RTG-2 cells (p > 0.05), and similar results were observed for cellular morphology and apoptotic rates. The contents of the main reactive oxygen species (ROS) components, superoxide anion (O2-) and hydrogen peroxide (H2O2), were markedly increased in Neuro-2a cell with STX exposure at middle (15 nM) and high (150 nM) concentrations (p < 0.05), and the simultaneous increase of the ratio of reduced/oxidized glutathione (GSH/GSSG) (p < 0.05) inferred the occurrence of oxidative stress. However, little difference was observed in all treated groups of RTG-2 cells. The activities of three antioxidant enzymes, superoxide dismutase (SOD), catalase (CAT) and glutathione reductase (GR), were significantly enhanced in Neuro-2a cells in the middle and high concentration groups (p < 0.05), while glutathione peroxidase (GPX) obviously decreased (p < 0.05) in all treated groups. Little change was found in RTG-2 cells with the same exposures. These results provided evidence that STX exposure altered the redox status of Neuro-2a cells and resulted in oxidative stress, but the same exposure exerted little effect on RTG-2 cells. Therefore, Neuro-2a cells are more sensitive than reproductive cells to STX exposure, and the antioxidant systems appears to be partly responsible for this differentiation response.
Collapse
Affiliation(s)
- Zhongyuan Zhou
- Department of Marine Ecology, College of Marine Life Science, Ocean University of China, Qingdao, 266003, China.
| | - Xuexi Tang
- Department of Marine Ecology, College of Marine Life Science, Ocean University of China, Qingdao, 266003, China; Laboratory for Marine Ecology and Environmental Science, Qingdao National Laboratory for Marine Science and Technology, Qingdao, 266071, China.
| | - Hongmei Chen
- Key Laboratory of Xinjiang Endemic Phytomedicine Resources, Ministry of Education, Pharmacology Department, School of Pharmacy, Shihezi University, Shihezi, 832002, China.
| | - You Wang
- Department of Marine Ecology, College of Marine Life Science, Ocean University of China, Qingdao, 266003, China; Laboratory for Marine Ecology and Environmental Science, Qingdao National Laboratory for Marine Science and Technology, Qingdao, 266071, China.
| |
Collapse
|
21
|
Tsutsui Y, Sunada K. Dexmedetomidine Increases the Latency of Thermal Antinociception in Rats. Anesth Prog 2017; 64:230-234. [PMID: 29200374 DOI: 10.2344/anpr-65-01-08] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022] Open
Abstract
Recent reports have stated that dexmedetomidine (DEX), an α2-adrenoreceptor agonist, enhances the local anesthetic effects of ropivacaine and prolongs its effective duration. However, little is known about the effect of a combination of DEX and lidocaine on anesthetic duration. Therefore, we investigated whether DEX can prolong the local anesthetic effect of lidocaine, using the thermal paw withdrawal test in Wistar rats in order to measure local anesthetic duration. We subcutaneously injected 50 μL of either normal saline, 2% lidocaine, a combination of 0.5 μg/kg DEX and 2% lidocaine, or a combination of 2% lidocaine with 1:80,000 epinephrine into the plantar surface of the left hind paw of the rats. The plantar region was stimulated using heat. We measured the perceived acute pain according to paw movement in response to stimulation. We found DEX significantly prolonged the paw withdrawal latency of lidocaine. Moreover, we found that DEX can prolong the local anesthetic duration of lidocaine as much as 1:80,000 epinephrine, up to 35 minutes after injection. In conclusion, this study concluded that a combination of DEX and lidocaine may be useful as a local anesthetic, similar to a combination of epinephrine and lidocaine, for short procedures.
Collapse
Affiliation(s)
- Yukako Tsutsui
- Department of Dental Anesthesiology, The Nippon Dental University School of Life Dentistry at Tokyo, Japan
| | - Katsuhisa Sunada
- Department of Dental Anesthesiology, The Nippon Dental University School of Life Dentistry at Tokyo, Japan
| |
Collapse
|
22
|
González-Cano R, Tejada MÁ, Artacho-Cordón A, Nieto FR, Entrena JM, Wood JN, Cendán CM. Effects of Tetrodotoxin in Mouse Models of Visceral Pain. Mar Drugs 2017; 15:E188. [PMID: 28635651 PMCID: PMC5484138 DOI: 10.3390/md15060188] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2017] [Revised: 06/07/2017] [Accepted: 06/16/2017] [Indexed: 12/11/2022] Open
Abstract
Visceral pain is very common and represents a major unmet clinical need for which current pharmacological treatments are often insufficient. Tetrodotoxin (TTX) is a potent neurotoxin that exerts analgesic actions in both humans and rodents under different somatic pain conditions, but its effect has been unexplored in visceral pain. Therefore, we tested the effects of systemic TTX in viscero-specific mouse models of chemical stimulation of the colon (intracolonic instillation of capsaicin and mustard oil) and intraperitoneal cyclophosphamide-induced cystitis. The subcutaneous administration of TTX dose-dependently inhibited the number of pain-related behaviors in all evaluated pain models and reversed the referred mechanical hyperalgesia (examined by stimulation of the abdomen with von Frey filaments) induced by capsaicin and cyclophosphamide, but not that induced by mustard oil. Morphine inhibited both pain responses and the referred mechanical hyperalgesia in all tests. Conditional nociceptor‑specific Nav1.7 knockout mice treated with TTX showed the same responses as littermate controls after the administration of the algogens. No motor incoordination after the administration of TTX was observed. These results suggest that blockade of TTX-sensitive sodium channels, but not Nav1.7 subtype alone, by systemic administration of TTX might be a potential therapeutic strategy for the treatment of visceral pain.
Collapse
Affiliation(s)
- Rafael González-Cano
- Department of Pharmacology, Biomedical Research Centre and Institute of Neuroscience, Faculty of Medicine, University of Granada, 18016 Granada, Spain.
- Biosanitary Research Institute, University Hospital Complex of Granada, 18012 Granada, Spain.
| | - Miguel Ángel Tejada
- Department of Pharmacology, Biomedical Research Centre and Institute of Neuroscience, Faculty of Medicine, University of Granada, 18016 Granada, Spain.
- Biosanitary Research Institute, University Hospital Complex of Granada, 18012 Granada, Spain.
| | - Antonia Artacho-Cordón
- Department of Pharmacology, Biomedical Research Centre and Institute of Neuroscience, Faculty of Medicine, University of Granada, 18016 Granada, Spain.
- Biosanitary Research Institute, University Hospital Complex of Granada, 18012 Granada, Spain.
| | - Francisco Rafael Nieto
- Department of Pharmacology, Biomedical Research Centre and Institute of Neuroscience, Faculty of Medicine, University of Granada, 18016 Granada, Spain.
- Biosanitary Research Institute, University Hospital Complex of Granada, 18012 Granada, Spain.
| | - José Manuel Entrena
- Animal Behavior Research Unit, Scientific Instrumentation Center, University of Granada, Armilla, 18100 Granada, Spain.
| | - John N Wood
- Molecular Nociception Group, Wolfson Institute for Biomedical Research, University College London, London WC1E 6BT, UK.
| | - Cruz Miguel Cendán
- Department of Pharmacology, Biomedical Research Centre and Institute of Neuroscience, Faculty of Medicine, University of Granada, 18016 Granada, Spain.
- Biosanitary Research Institute, University Hospital Complex of Granada, 18012 Granada, Spain.
| |
Collapse
|
23
|
Leng XR, Qi XH, Zhou YT, Wang YP. Gain-of-function mutation p.Arg225Cys in SCN11A causes familial episodic pain and contributes to essential tremor. J Hum Genet 2017; 62:641-646. [DOI: 10.1038/jhg.2017.21] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2016] [Revised: 01/16/2017] [Accepted: 01/19/2017] [Indexed: 02/01/2023]
|
24
|
An Updated Review of Ciguatera Fish Poisoning: Clinical, Epidemiological, Environmental, and Public Health Management. Mar Drugs 2017; 15:md15030072. [PMID: 28335428 PMCID: PMC5367029 DOI: 10.3390/md15030072] [Citation(s) in RCA: 186] [Impact Index Per Article: 23.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2016] [Revised: 02/13/2017] [Accepted: 02/13/2017] [Indexed: 01/07/2023] Open
Abstract
Ciguatera Fish Poisoning (CFP) is the most frequently reported seafood-toxin illness in the world. It causes substantial human health, social, and economic impacts. The illness produces a complex array of gastrointestinal, neurological and neuropsychological, and cardiovascular symptoms, which may last days, weeks, or months. This paper is a general review of CFP including the human health effects of exposure to ciguatoxins (CTXs), diagnosis, human pathophysiology of CFP, treatment, detection of CTXs in fish, epidemiology of the illness, global dimensions, prevention, future directions, and recommendations for clinicians and patients. It updates and expands upon the previous review of CFP published by Friedman et al. (2008) and addresses new insights and relevant emerging global themes such as climate and environmental change, international market issues, and socioeconomic impacts of CFP. It also provides a proposed universal case definition for CFP designed to account for the variability in symptom presentation across different geographic regions. Information that is important but unchanged since the previous review has been reiterated. This article is intended for a broad audience, including resource and fishery managers, commercial and recreational fishers, public health officials, medical professionals, and other interested parties.
Collapse
|
25
|
Contribution of concentration-sensitive sodium channels to the absorption of alveolar fluid in mice. Respir Physiol Neurobiol 2016; 231:45-54. [PMID: 27259686 DOI: 10.1016/j.resp.2016.05.017] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2016] [Revised: 04/29/2016] [Accepted: 05/29/2016] [Indexed: 01/11/2023]
Abstract
The concentration-sensitive sodium channel (Nac) is activated by an increase in the extracellular sodium concentration. Although the expression of Nac in alveolar type II epithelial cells (AEC II) has been reported previously, the physiological role of Nac in the lung has not been established. We characterized Nac expression and examined amiloride-insensitive sodium transport mediated by Nac in mouse lung. Immunofluorescence studies revealed that Nac did not colocalize with either aquaporin 5 or cystic fibrosis transmembrane conductance regulator, but partially colocalized with the epithelial sodium channel γ-subunit. Immunoelectron microscopy studies showed that Nac localized at the basolateral membrane of pulmonary microvascular endothelial cells (PMVECs). Nac mRNA and protein were expressed in PMVECs isolated from the lungs of mice. Image analysis indicated that sodium influx into the alveolar wall was dependent on increases in extracellular sodium concentration. We conclude that Nac expressed in PMVECs and AEC II contributes to the reabsorption of sodium via an amiloride-insensitive pathway during alveolar fluid clearance.
Collapse
|
26
|
Shcherbatko A, Rossi A, Foletti D, Zhu G, Bogin O, Galindo Casas M, Rickert M, Hasa-Moreno A, Bartsevich V, Crameri A, Steiner AR, Henningsen R, Gill A, Pons J, Shelton DL, Rajpal A, Strop P. Engineering Highly Potent and Selective Microproteins against Nav1.7 Sodium Channel for Treatment of Pain. J Biol Chem 2016; 291:13974-13986. [PMID: 27129258 DOI: 10.1074/jbc.m116.725978] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2016] [Indexed: 12/19/2022] Open
Abstract
The prominent role of voltage-gated sodium channel 1.7 (Nav1.7) in nociception was revealed by remarkable human clinical and genetic evidence. Development of potent and subtype-selective inhibitors of this ion channel is crucial for obtaining therapeutically useful analgesic compounds. Microproteins isolated from animal venoms have been identified as promising therapeutic leads for ion channels, because they naturally evolved to be potent ion channel blockers. Here, we report the engineering of highly potent and selective inhibitors of the Nav1.7 channel based on tarantula ceratotoxin-1 (CcoTx1). We utilized a combination of directed evolution, saturation mutagenesis, chemical modification, and rational drug design to obtain higher potency and selectivity to the Nav1.7 channel. The resulting microproteins are highly potent (IC50 to Nav1.7 of 2.5 nm) and selective. We achieved 80- and 20-fold selectivity over the closely related Nav1.2 and Nav1.6 channels, respectively, and the IC50 on skeletal (Nav1.4) and cardiac (Nav1.5) sodium channels is above 3000 nm The lead molecules have the potential for future clinical development as novel therapeutics in the treatment of pain.
Collapse
Affiliation(s)
| | - Andrea Rossi
- Rinat Laboratories, Pfizer Inc., South San Francisco, California 94080
| | - Davide Foletti
- Rinat Laboratories, Pfizer Inc., South San Francisco, California 94080
| | - Guoyun Zhu
- Rinat Laboratories, Pfizer Inc., South San Francisco, California 94080
| | | | | | - Mathias Rickert
- Rinat Laboratories, Pfizer Inc., South San Francisco, California 94080
| | - Adela Hasa-Moreno
- Rinat Laboratories, Pfizer Inc., South San Francisco, California 94080
| | | | | | | | | | - Avinash Gill
- Sutro Biopharma, South San Francisco, California 94080
| | - Jaume Pons
- Rinat Laboratories, Pfizer Inc., South San Francisco, California 94080
| | - David L Shelton
- Rinat Laboratories, Pfizer Inc., South San Francisco, California 94080
| | - Arvind Rajpal
- Rinat Laboratories, Pfizer Inc., South San Francisco, California 94080
| | - Pavel Strop
- Rinat Laboratories, Pfizer Inc., South San Francisco, California 94080,.
| |
Collapse
|
27
|
Emery EC, Luiz AP, Wood JN. Nav1.7 and other voltage-gated sodium channels as drug targets for pain relief. Expert Opin Ther Targets 2016; 20:975-83. [PMID: 26941184 PMCID: PMC4950419 DOI: 10.1517/14728222.2016.1162295] [Citation(s) in RCA: 141] [Impact Index Per Article: 15.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Introduction: Chronic pain is a massive clinical problem. We discuss the potential of subtype selective sodium channel blockers that may provide analgesia with limited side effects. Areas covered: Sodium channel subtypes have been linked to human pain syndromes through genetic studies. Gain of function mutations in Nav1.7, 1.8 and 1.9 can cause pain, whilst loss of function Nav1.7 mutations lead to loss of pain in otherwise normal people. Intriguingly, both human and mouse Nav1.7 null mutants have increased opioid drive, because naloxone, an opioid antagonist, can reverse the analgesia associated with the loss of Nav1.7 expression. Expert Opinion: We believe there is a great future for sodium channel antagonists, particularly Nav1.7 antagonists in treating most pain syndromes. This review deals with recent attempts to develop specific sodium channel blockers, the mechanisms that underpin the Nav1.7 null pain-free phenotype and new routes to analgesia using, for example, gene therapy or combination therapy with subtype specific sodium channel blockers and opioids. The use of selective Nav1.7 antagonists together with either enkephalinase inhibitors or low dose opioids has the potential for side effect-free analgesia, as well as an important opioid sparing function that may be clinically very significant.
Collapse
Affiliation(s)
- Edward C Emery
- a Molecular Nociception Group, Department of Medicine , WIBR, University College London , London WC1E 6BT , UK
| | - Ana Paula Luiz
- a Molecular Nociception Group, Department of Medicine , WIBR, University College London , London WC1E 6BT , UK
| | - John N Wood
- a Molecular Nociception Group, Department of Medicine , WIBR, University College London , London WC1E 6BT , UK
| |
Collapse
|
28
|
Jeevaratnam K, Guzadhur L, Goh YM, Grace AA, Huang CLH. Sodium channel haploinsufficiency and structural change in ventricular arrhythmogenesis. Acta Physiol (Oxf) 2016; 216:186-202. [PMID: 26284956 DOI: 10.1111/apha.12577] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2015] [Revised: 06/11/2015] [Accepted: 07/24/2015] [Indexed: 12/19/2022]
Abstract
Normal cardiac excitation involves orderly conduction of electrical activation and recovery dependent upon surface membrane, voltage-gated, sodium (Na(+) ) channel α-subunits (Nav 1.5). We summarize experimental studies of physiological and clinical consequences of loss-of-function Na(+) channel mutations. Of these conditions, Brugada syndrome (BrS) and progressive cardiac conduction defect (PCCD) are associated with sudden, often fatal, ventricular tachycardia (VT) or fibrillation. Mouse Scn5a(+/-) hearts replicate important clinical phenotypes modelling these human conditions. The arrhythmic phenotype is associated not only with the primary biophysical change but also with additional, anatomical abnormalities, in turn dependent upon age and sex, each themselves exerting arrhythmic effects. Available evidence suggests a unified binary scheme for the development of arrhythmia in both BrS and PCCD. Previous biophysical studies suggested that Nav 1.5 deficiency produces a background electrophysiological defect compromising conduction, thereby producing an arrhythmic substrate unmasked by flecainide or ajmaline challenge. More recent reports further suggest a progressive decline in conduction velocity and increase in its dispersion particularly in ageing male Nav 1.5 haploinsufficient compared to WT hearts. This appears to involve a selective appearance of slow conduction at the expense of rapidly conducting pathways with changes in their frequency distributions. These changes were related to increased cardiac fibrosis. It is thus the combination of the structural and biophysical changes both accentuating arrhythmic substrate that may produce arrhythmic tendency. This binary scheme explains the combined requirement for separate, biophysical and structural changes, particularly occurring in ageing Nav 1.5 haploinsufficient males in producing clinical arrhythmia.
Collapse
Affiliation(s)
- K. Jeevaratnam
- Faculty of Health and Medical Science; University of Surrey; Guilford UK
- Perdana University - Royal College of Surgeons Ireland; Serdang Selangor Darul Ehsan Malaysia
| | - L. Guzadhur
- Division of Cardiovascular Biology; Department of Biochemistry; University of Cambridge; Cambridge UK
- Niche Science & Technology; Richmond UK
| | - Y. M. Goh
- Department of Preclinical Sciences; Faculty of Veterinary Medicine; University Putra Malaysia; Serdang Selangor Darul Ehsan Malaysia
| | - A. A. Grace
- Division of Cardiovascular Biology; Department of Biochemistry; University of Cambridge; Cambridge UK
| | - C. L.-H. Huang
- Division of Cardiovascular Biology; Department of Biochemistry; University of Cambridge; Cambridge UK
- Physiological Laboratory; University of Cambridge; Cambridge UK
| |
Collapse
|
29
|
Luiz AP, Wood JN. Sodium Channels in Pain and Cancer: New Therapeutic Opportunities. ADVANCES IN PHARMACOLOGY (SAN DIEGO, CALIF.) 2016; 75:153-78. [PMID: 26920012 DOI: 10.1016/bs.apha.2015.12.006] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/25/2023]
Abstract
Voltage-gated sodium channels (VGSCs) underpin electrical activity in the nervous system through action potential propagation. First predicted by the modeling studies of Hodgkin and Huxley, they were subsequently identified at the molecular level by groups led by Catterall and Numa. VGSC dysfunction has long been linked to neuronal and cardiac disorders with some nonselective sodium channel blockers in current use in the clinic. The lack of selectivity means that side effect issues are a major impediment to the use of broad spectrum sodium channel blockers. Nine different sodium channels are known to exist, and selective blockers are now being developed. The potential utility of these drugs to target diseases ranging from migraine, multiple sclerosis, muscle, and immune system disorders, to cancer and pain is being explored. Four channels are potential targets for pain disorders. This conclusion comes from mouse knockout studies and human mutations that prove the involvement of Nav1.3, Nav1.7, Nav1.8, and Nav1.9 in the development and maintenance of acute and chronic pain. In this chapter, we present a short overview of the possible role of Nav1.3, Nav1.7, Nav1.8, and Nav1.9 in human pain and the emerging and unexpected role of sodium channels in cancer pathogenesis.
Collapse
Affiliation(s)
- Ana Paula Luiz
- Molecular Nociception Group, Wolfson Institute for Biomedical Research, University College London, London, United Kingdom
| | - John N Wood
- Molecular Nociception Group, Wolfson Institute for Biomedical Research, University College London, London, United Kingdom.
| |
Collapse
|
30
|
Zhang F, Liu C, Tan H, Wang H, Jiang Y, Liang S, Zhang F, Liu Z. A survey of the venom of the spider Lycosa vittata by biochemical, pharmacological and transcriptomic analyses. Toxicon 2015; 107:335-43. [DOI: 10.1016/j.toxicon.2015.05.004] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2015] [Revised: 05/04/2015] [Accepted: 05/07/2015] [Indexed: 11/26/2022]
|
31
|
Gu H, Fang YJ, Liu DD, Chen P, Mei YA. cAMP/PKA Pathways and S56 Phosphorylation Are Involved in AA/PGE2-Induced Increases in rNaV1.4 Current. PLoS One 2015; 10:e0140715. [PMID: 26485043 PMCID: PMC4618696 DOI: 10.1371/journal.pone.0140715] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2015] [Accepted: 09/28/2015] [Indexed: 12/19/2022] Open
Abstract
Arachidonic acid (AA) and its metabolites are important second messengers for ion channel modulation. The effects of extracellular application of AA and its non-metabolized analogue on muscle rNaV1.4 Na+ current has been studied, but little is known about the effects of intracellular application of AA on this channel isoform. Here, we report that intracellular application of AA significantly augmented the rNaV1.4 current peak without modulating the steady-state activation and inactivation properties of the rNaV1.4 channel. These results differed from the effects of extracellular application of AA on rNaV1.4 current. The effects of intracellular AA were mimicked by prostaglandin E2 but not eicosatetraynoic acid (ETYA), the non-metabolized analogue of AA, and were eliminated by treatment with cyclooxygenase inhibitors, flufenamic acid, or indomethacin. AA/PGE2-induced activation of rNaV1.4 channels was mimicked by a cAMP analogue (db-cAMP) and eliminated by a PKA inhibitor, PKAi. Furthermore, inhibition of EP2 and EP4 (PGE2 receptors) with AH6809 and AH23848 reduced the intracellular AA/PGE2-induced increase of rNaV1.4 current. Two mutated channels, rNaV1.4S56A and rNaV1.4T21A, were designed to investigate the role of predicted phosphorylation sites in the AA/PGE2–mediated regulation of rNaV1.4 currents. In rNaV1.4S56A, the effects of intracellular db-cAMP, AA, and PGE2 were significantly reduced. The results of the present study suggest that intracellular AA augments rNaV1.4 current by PGE2/EP receptor-mediated activation of the cAMP/PKA pathway, and that the S56 residue on the channel protein is important for this process.
Collapse
Affiliation(s)
- Hua Gu
- School of Life Science and Technology, Tongji University, Shanghai 200092, PR China
- * E-mail: (HG); (YAM)
| | - Yan-Jia Fang
- School of Life Sciences, Institute of Brain Science and State Key Laboratory of Medical Neurobiology, Fudan University, Shanghai 200433, PR China
| | - Dong-Dong Liu
- School of Life Sciences, Institute of Brain Science and State Key Laboratory of Medical Neurobiology, Fudan University, Shanghai 200433, PR China
| | - Ping Chen
- School of Life Science and Technology, Tongji University, Shanghai 200092, PR China
| | - Yan-Ai Mei
- School of Life Sciences, Institute of Brain Science and State Key Laboratory of Medical Neurobiology, Fudan University, Shanghai 200433, PR China
- * E-mail: (HG); (YAM)
| |
Collapse
|
32
|
Sun LH, Yan ML, Hu XL, Peng LW, Che H, Bao YN, Guo F, Liu T, Chen X, Zhang R, Ban T, Wang N, Liu HL, Hou X, Ai J. MicroRNA-9 induces defective trafficking of Nav1.1 and Nav1.2 by targeting Navβ2 protein coding region in rat with chronic brain hypoperfusion. Mol Neurodegener 2015; 10:36. [PMID: 26259688 PMCID: PMC4530481 DOI: 10.1186/s13024-015-0032-9] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2014] [Accepted: 07/24/2015] [Indexed: 01/09/2023] Open
Abstract
Background Previous studies have demonstrated that the trafficking defects of Nav1.1/Nav1.2 are involved in the dementia pathophysiology. However, the detailed mechanisms are not fully understood. Moreover, whether the impaired miRNAs regulation linked to dementia is a key player in sodium channel trafficking disturbance remains unclear. The cognitive impairment induced by chronic cerebral ischemia through chronic brain hypoperfusion (CBH) is likely reason to precede dementia. Therefore, our goal in the present study was to examine the role of microRNA-9 (miR-9) in regulating Nav1.1/Nav1.2 trafficking under CBH generated by bilateral common carotid artery occlusion (2VO). Results The impairment of Nav1.1/Nav1.2 trafficking and decreased expression of Navβ2 were found in the hippocampi and cortices of rats following CBH generated by bilateral 2VO. MiR-9 was increased in both the hippocampi and cortices of rats following CBH by qRT-PCR. Intriguingly, miR-9 suppressed, while AMO-miR-9 enhanced, the trafficking of Nav1.1/Nav1.2 from cytoplasm to cell membrane. Further study showed that overexpression of miR-9 inhibited the Navβ2 expression by targeting on its coding sequence (CDS) domain by dual luciferase assay. However, binding-site mutation or miR-masks failed to influence Navβ2 expression as well as Nav1.1/Nav1.2 trafficking process, indicating that Navβ2 is a potential target for miR-9. Lentivirus-mediated miR-9 overexpression also inhibited Navβ2 expression and elicited translocation deficits to cell membrane of Nav1.1/Nav1.2 in rats, whereas injection of lentivirus-mediated miR-9 knockdown could reverse the impaired trafficking of Nav1.1/Nav1.2 triggered by 2VO. Conclusions We conclude that miR-9 may play a key role in regulating the process of Nav1.1/Nav1.2 trafficking via targeting on Navβ2 protein in 2VO rats at post-transcriptional level, and inhibition of miR-9 may be a potentially valuable approach to prevent Nav1.1/Nav1.2 trafficking disturbance induced by CBH.
Collapse
Affiliation(s)
- Li-Hua Sun
- Department of Pharmacology, Harbin Medical University, No.157 Baojian Road, Nangang District,Harbin, Heilongjiang Province, 15008, China.
| | - Mei-Ling Yan
- Department of Pharmacology, Harbin Medical University, No.157 Baojian Road, Nangang District,Harbin, Heilongjiang Province, 15008, China
| | - Xue-Ling Hu
- Department of Pharmacology, Harbin Medical University, No.157 Baojian Road, Nangang District,Harbin, Heilongjiang Province, 15008, China
| | - Li-Wei Peng
- Department of Pharmacology, Harbin Medical University, No.157 Baojian Road, Nangang District,Harbin, Heilongjiang Province, 15008, China
| | - Hui Che
- Department of Pharmacology, Harbin Medical University, No.157 Baojian Road, Nangang District,Harbin, Heilongjiang Province, 15008, China
| | - Ya-Nan Bao
- Department of Pharmacology, Harbin Medical University, No.157 Baojian Road, Nangang District,Harbin, Heilongjiang Province, 15008, China
| | - Fei Guo
- Department of Pharmacology, Harbin Medical University, No.157 Baojian Road, Nangang District,Harbin, Heilongjiang Province, 15008, China
| | - Tong Liu
- Department of Pharmacology, Harbin Medical University, No.157 Baojian Road, Nangang District,Harbin, Heilongjiang Province, 15008, China
| | - Xin Chen
- Department of Pharmacology, Harbin Medical University, No.157 Baojian Road, Nangang District,Harbin, Heilongjiang Province, 15008, China
| | - Rong Zhang
- Department of Pharmacology, Harbin Medical University, No.157 Baojian Road, Nangang District,Harbin, Heilongjiang Province, 15008, China
| | - Tao Ban
- Department of Pharmacology, Harbin Medical University, No.157 Baojian Road, Nangang District,Harbin, Heilongjiang Province, 15008, China
| | - Ning Wang
- Department of Pharmacology, Harbin Medical University, No.157 Baojian Road, Nangang District,Harbin, Heilongjiang Province, 15008, China
| | - Huai-Lei Liu
- Department of Neurosurgery, The First Affiliated Hospital of Harbin Medical University, No.157 Baojian Road, Nangang District, Harbin, Heilongjiang Province, 150081, China
| | - Xu Hou
- Department of Neurosurgery, The First Affiliated Hospital of Harbin Medical University, No.157 Baojian Road, Nangang District, Harbin, Heilongjiang Province, 150081, China
| | - Jing Ai
- Department of Pharmacology, Harbin Medical University, No.157 Baojian Road, Nangang District,Harbin, Heilongjiang Province, 15008, China.
| |
Collapse
|
33
|
Heng LJ, Qi R, Yang RH, Xu GZ. Docosahexaenoic acid inhibits mechanical allodynia and thermal hyperalgesia in diabetic rats by decreasing the excitability of DRG neurons. Exp Neurol 2015; 271:291-300. [PMID: 26118950 DOI: 10.1016/j.expneurol.2015.06.022] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2015] [Revised: 06/07/2015] [Accepted: 06/24/2015] [Indexed: 11/16/2022]
Abstract
Diabetes mellitus is a common metabolic disease in human beings with characteristic symptoms of hyperglycemia, chronic inflammation and insulin resistance. One of the most common complications of early-onset diabetes mellitus is peripheral diabetic neuropathy, which is manifested either by loss of nociception or by allodynia and hyperalgesia. Dietary fatty acids, especially polyunsaturated fatty acids, have been shown the potential of anti-inflammation and modulating neuron excitability. The present study investigated the effects of docosahexaenoic acid (DHA) on the excitability of dorsal root ganglion (DRG) neurons in streptozotocin (STZ)-induced diabetes rats. The effects of DHA on the allodynia and hyperalgesia of diabetic rats were also evaluated. Dietary DHA supplementation effectively attenuated both allodynia and hyperalgesia induced by STZ injection. DHA supplementation decreased the excitability of DRG neurons by decreasing the sodium currents and increasing potassium currents, which may contribute to the effect of alleviating allodynia and hyperalgesia in diabetic rats. The results suggested that DHA might be useful as an adjuvant therapy for the prevention and treatment of painful diabetic neuropathy.
Collapse
Affiliation(s)
- Li-Jun Heng
- Department of Neurosurgery, Wuhan General Hospital of Guangzhou Military Command, Wuhan, Hubei 430070, China; Department of Neurosurgery, Tangdu Hospital of Fourth Military Medical University, Xi'an, Shaanxi 710038, China
| | - Rui Qi
- Department of Nutrition and Food Hygiene, School of Public Health, The Fourth Military Medical University, Xi'an 710032, China
| | - Rui-Hua Yang
- Department of Nutrition and Food Hygiene, School of Public Health, The Fourth Military Medical University, Xi'an 710032, China.
| | - Guo-Zheng Xu
- Department of Neurosurgery, Wuhan General Hospital of Guangzhou Military Command, Wuhan, Hubei 430070, China.
| |
Collapse
|
34
|
León Ariza HH, Valenzuela Faccini N, Rojas Ortega AC, Botero Rosas DA. Nav1.5 cardiac sodium channels, regulation and clinical implications. REVISTA DE LA FACULTAD DE MEDICINA 2015. [DOI: 10.15446/revfacmed.v62n4.44015] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
<p>Voltage-gated sodium channels constitute a group of membrane<br />proteins widely distributed thought the body. In the heart, there<br />are at least six different isoforms, being the Nav1.5 the most<br />abundant. The channel is composed of an α subunit that is formed<br />by four domains of six segments each, and four much smaller β<br />subunits that provide stability and integrate other channels into<br />the α subunit. The function of the Nav1.5 channel is modulated<br />by intracellular cytoskeleton proteins, extracellular proteins,<br />calcium concentration, free radicals, and medications, among<br />other things. The study of the channel and its alterations has<br />grown thanks to its association with pathogenic conditions such<br />as Long QT syndrome, Brugada syndrome, atrial fibrillation,<br />arrhythmogenic ventricular dysplasia and complications during<br />ischemic processes.</p>
Collapse
|
35
|
Schreiber AK, Nones CFM, Reis RC, Chichorro JG, Cunha JM. Diabetic neuropathic pain: Physiopathology and treatment. World J Diabetes 2015; 6:432-444. [PMID: 25897354 PMCID: PMC4398900 DOI: 10.4239/wjd.v6.i3.432] [Citation(s) in RCA: 262] [Impact Index Per Article: 26.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/29/2014] [Revised: 11/26/2014] [Accepted: 02/09/2015] [Indexed: 02/05/2023] Open
Abstract
Diabetic neuropathy is a common complication of both type 1 and type 2 diabetes, which affects over 90% of the diabetic patients. Although pain is one of the main symptoms of diabetic neuropathy, its pathophysiological mechanisms are not yet fully known. It is widely accepted that the toxic effects of hyperglycemia play an important role in the development of this complication, but several other hypotheses have been postulated. The management of diabetic neuropathic pain consists basically in excluding other causes of painful peripheral neuropathy, improving glycemic control as a prophylactic therapy and using medications to alleviate pain. First line drugs for pain relief include anticonvulsants, such as pregabalin and gabapentin and antidepressants, especially those that act to inhibit the reuptake of serotonin and noradrenaline. In addition, there is experimental and clinical evidence that opioids can be helpful in pain control, mainly if associated with first line drugs. Other agents, including for topical application, such as capsaicin cream and lidocaine patches, have also been proposed to be useful as adjuvants in the control of diabetic neuropathic pain, but the clinical evidence is insufficient to support their use. In conclusion, a better understanding of the mechanisms underlying diabetic neuropathic pain will contribute to the search of new therapies, but also to the improvement of the guidelines to optimize pain control with the drugs currently available.
Collapse
|
36
|
Yin R, Liu D, Chhoa M, Li CM, Luo Y, Zhang M, Lehto SG, Immke DC, Moyer BD. Voltage-gated sodium channel function and expression in injured and uninjured rat dorsal root ganglia neurons. Int J Neurosci 2015; 126:182-92. [DOI: 10.3109/00207454.2015.1004172] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
|
37
|
Mattei C, Molgó J, Benoit E. Involvement of both sodium influx and potassium efflux in ciguatoxin-induced nodal swelling of frog myelinated axons. Neuropharmacology 2014; 85:417-26. [PMID: 24950451 DOI: 10.1016/j.neuropharm.2014.06.001] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2014] [Revised: 05/09/2014] [Accepted: 06/02/2014] [Indexed: 01/22/2023]
Abstract
Ciguatoxins, mainly produced by benthic dinoflagellate Gambierdiscus species, are responsible for a complex human poisoning known as ciguatera. Previous pharmacological studies revealed that these toxins activate voltage-gated Na+ channels. In frog nodes of Ranvier, ciguatoxins induce spontaneous and repetitive action potentials (APs) and increase axonal volume that may explain alterations of nerve functioning in intoxicated humans. The present study aimed determining the ionic mechanisms involved in Pacific ciguatoxin-1B (P-CTX-1B)-induced membrane hyperexcitability and subsequent volume increase in frog nodes of Ranvier, using electrophysiology and confocal microscopy. The results reveal that P-CTX-1B action is not dependent on external Cl- ions since it was not affected by substituting Cl- by methylsulfate ions. In contrast, substitution of external Na+ by Li+ ions suppressed spontaneous APs and prevented nodal swelling. This suggests that P-CTX-1B-modified Na+ channels are not selective to Li+ ions and/or are blocked by these ions, and that Na+ influx through Na+ channels opened during spontaneous APs is required for axonal swelling. The fact that the K+ channel blocker tetraethylammonium modified, but did not suppress, spontaneous APs and greatly reduced nodal swelling induced by P-CTX-1B indicates that K+ efflux might also be involved. This is supported by the fact that P-CTX-1B, when tested in the presence of both tetraethylammonium and the K+ ionophore valinomycin, produced the characteristic nodal swelling. It is concluded that, during the action of P-CTX-1B, water movements responsible for axonal swelling depend on both Na+ influx and K+ efflux. These results pave the way for further studies regarding ciguatera treatment.
Collapse
Affiliation(s)
- César Mattei
- CNRS, Institut de Neurobiologie Alfred Fessard - FRC2118, Laboratoire de Neurobiologie et Développement - UPR3294, 91198 Gif-sur-Yvette cedex, France; Laboratoire Biologie Neurovasculaire et Mitochondriale Intégrée, UMR CNRS 6214 INSERM 1083, Université d'Angers, 49045 Angers cedex 01, France.
| | - Jordi Molgó
- CNRS, Institut de Neurobiologie Alfred Fessard - FRC2118, Laboratoire de Neurobiologie et Développement - UPR3294, 91198 Gif-sur-Yvette cedex, France
| | - Evelyne Benoit
- CNRS, Institut de Neurobiologie Alfred Fessard - FRC2118, Laboratoire de Neurobiologie et Développement - UPR3294, 91198 Gif-sur-Yvette cedex, France
| |
Collapse
|
38
|
Tanaka KI, Nakanishi Y, Sekino S, Ikegami M, Ikeda H, Kamei J. Fentanyl produces an anti-hyperalgesic effect through the suppression of sodium channels in mice with painful diabetic neuropathy. Eur J Pharmacol 2014; 733:68-74. [DOI: 10.1016/j.ejphar.2014.03.042] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2013] [Revised: 03/17/2014] [Accepted: 03/24/2014] [Indexed: 11/30/2022]
|
39
|
Yan Y, Li J, Zhang Y, Peng X, Guo T, Wang J, Hu W, Duan Z, Wang X. Physiological and biochemical characterization of egg extract of black widow spiders to uncover molecular basis of egg toxicity. Biol Res 2014; 47:17. [PMID: 25027663 PMCID: PMC4101730 DOI: 10.1186/0717-6287-47-17] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2014] [Accepted: 05/10/2014] [Indexed: 11/16/2022] Open
Abstract
Background Black widow spider (L. tredecimguttatus) has toxic components not only in the venomous glands, but also in other parts of the body and its eggs. It is biologically important to investigate the molecular basis of the egg toxicity. Results In the present work, an aqueous extract was prepared from the eggs of the spider and characterized using multiple physiological and biochemical strategies. Gel electrophoresis and mass spectrometry demonstrated that the eggs are rich in high-molecular-mass proteins and the peptides below 5 kDa. The lyophilized extract of the eggs had a protein content of 34.22% and was shown to have a strong toxicity towards mammals and insects. When applied at a concentration of 0.25 mg/mL, the extract could completely block the neuromuscular transmission in mouse isolated phrenic nerve-hemidiaphragm preparations within 12.0 ± 1.5 min. Using whole-cell patch-clamp technique, the egg extract was demonstrated to be able to inhibit the voltage-activated Na+, K+ and Ca2+ currents in rat DRG neurons. In addition, the extract displayed activities of multiple hydrolases. Finally, the molecular basis of the egg toxicity was discussed. Conclusions The eggs of black widow spiders are rich in proteinous compounds particularly the high-molecular-mass proteins with different types of biological activity The neurotoxic and other active compounds in the eggs are believed to play important roles in the eggs’ toxic actions.
Collapse
|
40
|
Li J, Yan Y, Yu H, Peng X, Zhang Y, Hu W, Duan Z, Wang X, Liang S. Isolation and identification of a sodium channel-inhibiting protein from eggs of black widow spiders. Int J Biol Macromol 2014; 65:115-20. [PMID: 24412150 DOI: 10.1016/j.ijbiomac.2014.01.004] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2013] [Revised: 01/01/2014] [Accepted: 01/02/2014] [Indexed: 11/30/2022]
|
41
|
Application of quinidine on rat sciatic nerve decreases the amplitude and increases the latency of evoked responses. J Anesth 2013; 28:559-68. [DOI: 10.1007/s00540-013-1754-x] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2013] [Accepted: 11/07/2013] [Indexed: 10/25/2022]
|
42
|
Zhang F, Xu X, Li T, Liu Z. Shellfish toxins targeting voltage-gated sodium channels. Mar Drugs 2013; 11:4698-723. [PMID: 24287955 PMCID: PMC3877881 DOI: 10.3390/md11124698] [Citation(s) in RCA: 41] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2013] [Revised: 11/10/2013] [Accepted: 11/12/2013] [Indexed: 12/19/2022] Open
Abstract
Voltage-gated sodium channels (VGSCs) play a central role in the generation and propagation of action potentials in excitable neurons and other cells and are targeted by commonly used local anesthetics, antiarrhythmics, and anticonvulsants. They are also common targets of neurotoxins including shellfish toxins. Shellfish toxins are a variety of toxic secondary metabolites produced by prokaryotic cyanobacteria and eukaryotic dinoflagellates in both marine and fresh water systems, which can accumulate in marine animals via the food chain. Consumption of shellfish toxin-contaminated seafood may result in potentially fatal human shellfish poisoning. This article provides an overview of the structure, bioactivity, and pharmacology of shellfish toxins that act on VGSCs, along with a brief discussion on their pharmaceutical potential for pain management.
Collapse
Affiliation(s)
- Fan Zhang
- Cooperative Innovation Center of Engineering and New Products for Developmental Biology, College of Life Sciences, Hunan Normal University, Changsha, Hunan 410081, China.
| | | | | | | |
Collapse
|
43
|
Zhang X, Wen J, Yang W, Wang C, Gao L, Zheng L, Wang T, Ran K, Li Y, Li X, Xu M, Luo J, Feng S, Ma X, Ma H, Chai Z, Zhou Z, Yao J, Zhang X, Liu J. Gain-of-function mutations in SCN11A cause familial episodic pain. Am J Hum Genet 2013; 93:957-66. [PMID: 24207120 DOI: 10.1016/j.ajhg.2013.09.016] [Citation(s) in RCA: 147] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2013] [Revised: 08/04/2013] [Accepted: 09/25/2013] [Indexed: 12/15/2022] Open
Abstract
Many ion channel genes have been associated with human genetic pain disorders. Here we report two large Chinese families with autosomal-dominant episodic pain. We performed a genome-wide linkage scan with microsatellite markers after excluding mutations in three known genes (SCN9A, SCN10A, and TRPA1) that cause similar pain syndrome to our findings, and we mapped the genetic locus to a 7.81 Mb region on chromosome 3p22.3-p21.32. By using whole-exome sequencing followed by conventional Sanger sequencing, we identified two missense mutations in the gene encoding voltage-gated sodium channel Nav1.9 (SCN11A): c.673C>T (p.Arg225Cys) and c.2423C>G (p.Ala808Gly) (one in each family). Each mutation showed a perfect cosegregation with the pain phenotype in the corresponding family, and neither of them was detected in 1,021 normal individuals. Both missense mutations were predicted to change a highly conserved amino acid residue of the human Nav1.9 channel. We expressed the two SCN11A mutants in mouse dorsal root ganglion (DRG) neurons and showed that both mutations enhanced the channel's electrical activities and induced hyperexcitablity of DRG neurons. Taken together, our results suggest that gain-of-function mutations in SCN11A can be causative of an autosomal-dominant episodic pain disorder.
Collapse
|
44
|
Fedosov AÉ, Moshkovskiĭ SA, Kuznetsova KG, Olivera BM. [Conotoxins: from the biodiversity of gastropods to new drugs]. BIOMEDIT︠S︡INSKAI︠A︡ KHIMII︠A︡ 2013; 59:267-94. [PMID: 23987066 DOI: 10.18097/pbmc20135903267] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
Abstract
A review describes general trends in research of conotoxins that are peptide toxins isolated from sea gastropods of the Conus genus, since the toxins were discovered in 1970th. There are disclosed a conotoxin classification, their structure diversity and different ways of action to their molecular targets, mainly, ion channels. In the applied aspect of conotoxin research, drug discovery and development is discussed, the drugs being based on conotoxin structure. A first exemplary drug is a ziconotide, which is an analgesic of new generation.
Collapse
|
45
|
Nelsen DR, Nisani Z, Cooper AM, Fox GA, Gren ECK, Corbit AG, Hayes WK. Poisons, toxungens, and venoms: redefining and classifying toxic biological secretions and the organisms that employ them. Biol Rev Camb Philos Soc 2013; 89:450-65. [PMID: 24102715 DOI: 10.1111/brv.12062] [Citation(s) in RCA: 65] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2012] [Revised: 08/15/2013] [Accepted: 08/16/2013] [Indexed: 01/02/2023]
Abstract
Despite extensive study of poisonous and venomous organisms and the toxins they produce, a review of the literature reveals inconsistency and ambiguity in the definitions of 'poison' and 'venom'. These two terms are frequently conflated with one another, and with the more general term, 'toxin.' We therefore clarify distinctions among three major classes of toxins (biological, environmental, and anthropogenic or man-made), evaluate prior definitions of venom which differentiate it from poison, and propose more rigorous definitions for poison and venom based on differences in mechanism of delivery. We also introduce a new term, 'toxungen', thereby partitioning toxic biological secretions into three categories: poisons lacking a delivery mechanism, i.e. ingested, inhaled, or absorbed across the body surface; toxungens delivered to the body surface without an accompanying wound; and venoms, delivered to internal tissues via creation of a wound. We further propose a system to classify toxic organisms with respect to delivery mechanism (absent versus present), source (autogenous versus heterogenous), and storage of toxins (aglandular versus glandular). As examples, a frog that acquires toxins from its diet, stores the secretion within cutaneous glands, and transfers the secretion upon contact or ingestion would be heteroglandular-poisonous; an ant that produces its own toxins, stores the secretion in a gland, and sprays it for defence would be autoglandular-toxungenous; and an anemone that produces its own toxins within specialized cells that deliver the secretion via a penetrating wound would be autoaglandular-venomous. Adoption of our scheme should benefit our understanding of both proximate and ultimate causes in the evolution of these toxins.
Collapse
Affiliation(s)
- David R Nelsen
- Department of Earth and Biological Sciences, Loma Linda University, 11065 Campus Street, Loma Linda, CA, 92350, U.S.A
| | | | | | | | | | | | | |
Collapse
|
46
|
Synthesis and biological characterization of synthetic analogs of Huwentoxin-IV (Mu-theraphotoxin-Hh2a), a neuronal tetrodotoxin-sensitive sodium channel inhibitor. Toxicon 2013; 71:57-65. [PMID: 23726857 DOI: 10.1016/j.toxicon.2013.05.015] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2013] [Revised: 05/12/2013] [Accepted: 05/15/2013] [Indexed: 11/22/2022]
Abstract
Huwentoxin-IV (HWTX-IV, also named Mu-theraphotoxin-Hh2a) is a typical inhibitor cystine knot peptide isolated from the venom of Chinese tarantula Ornithoctonus huwena and is found to inhibit tetrodotoxin-sensitive (TTX-S) sodium channels from mammalian sensory neurons. This peptide binds to neurotoxin receptor site 4 located at the extracellular S3-S4 linker of domain II in neuronal sodium channels. However, the molecular surface of HWTX-IV interaction with sodium channels remains unknown. In this study, we synthesized HWTX-IV and three mutants (T28D, R29A and Q34D) and characterized their functions on TTX-S sodium channels from adult rat dorsal root ganglion (DRG) neurons. Analysis of liquid chromatography, mass spectrometry and circular dichroism spectrum indicated that all four synthetic peptides are properly folded. Synthetic HWTX-IV exhibited the same activity as native HWTX-IV, while three mutations reduced toxin binding affinities by 10-200 fold, indicating that the basic or vicinal polar residues Thr²⁸, Arg²⁹, and Gln³⁴ in C-terminus might play critical roles in the interaction of HWTX-IV with TTX-S sodium channels.
Collapse
|
47
|
Xu J, Song D, Xue Z, Gu L, Hertz L, Peng L. Requirement of Glycogenolysis for Uptake of Increased Extracellular K+ in Astrocytes: Potential Implications for K+ Homeostasis and Glycogen Usage in Brain. Neurochem Res 2012; 38:472-85. [DOI: 10.1007/s11064-012-0938-3] [Citation(s) in RCA: 99] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2012] [Revised: 11/09/2012] [Accepted: 11/20/2012] [Indexed: 11/29/2022]
|
48
|
Savio-Galimberti E, Gollob MH, Darbar D. Voltage-gated sodium channels: biophysics, pharmacology, and related channelopathies. Front Pharmacol 2012; 3:124. [PMID: 22798951 PMCID: PMC3394224 DOI: 10.3389/fphar.2012.00124] [Citation(s) in RCA: 80] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2011] [Accepted: 06/11/2012] [Indexed: 12/19/2022] Open
Abstract
Voltage-gated sodium channels (VGSC) are multi-molecular protein complexes expressed in both excitable and non-excitable cells. They are primarily formed by a pore-forming multi-spanning integral membrane glycoprotein (α-subunit) that can be associated with one or more regulatory β-subunits. The latter are single-span integral membrane proteins that modulate the sodium current (INa) and can also function as cell adhesion molecules. In vitro some of the cell-adhesive functions of the β-subunits may play important physiological roles independently of the α-subunits. Other endogenous regulatory proteins named “channel partners” or “channel interacting proteins” (ChiPs) like caveolin-3 and calmodulin/calmodulin kinase II (CaMKII) can also interact and modulate the expression and/or function of VGSC. In addition to their physiological roles in cell excitability and cell adhesion, VGSC are the site of action of toxins (like tetrodotoxin and saxitoxin), and pharmacologic agents (like antiarrhythmic drugs, local anesthetics, antiepileptic drugs, and newly developed analgesics). Mutations in genes that encode α- and/or β-subunits as well as the ChiPs can affect the structure and biophysical properties of VGSC, leading to the development of diseases termed sodium “channelopathies”. This review will outline the structure, function, and biophysical properties of VGSC as well as their pharmacology and associated channelopathies and highlight some of the recent advances in this field.
Collapse
Affiliation(s)
- Eleonora Savio-Galimberti
- Division of Cardiovascular Medicine, Department of Medicine, Vanderbilt University Nashville, TN, USA
| | | | | |
Collapse
|
49
|
Spencer CI, Li N, Chen Q, Johnson J, Nevill T, Kammonen J, Ionescu-Zanetti C. Ion channel pharmacology under flow: automation via well-plate microfluidics. Assay Drug Dev Technol 2012; 10:313-24. [PMID: 22574656 DOI: 10.1089/adt.2011.414] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
Automated patch clamping addresses the need for high-throughput screening of chemical entities that alter ion channel function. As a result, there is considerable utility in the pharmaceutical screening arena for novel platforms that can produce relevant data both rapidly and consistently. Here we present results that were obtained with an innovative microfluidic automated patch clamp system utilizing a well-plate that eliminates the necessity of internal robotic liquid handling. Continuous recording from cell ensembles, rapid solution switching, and a bench-top footprint enable a number of assay formats previously inaccessible to automated systems. An electro-pneumatic interface was employed to drive the laminar flow of solutions in a microfluidic network that delivered cells in suspension to ensemble recording sites. Whole-cell voltage clamp was applied to linear arrays of 20 cells in parallel utilizing a 64-channel voltage clamp amplifier. A number of unique assays requiring sequential compound applications separated by a second or less, such as rapid determination of the agonist EC(50) for a ligand-gated ion channel or the kinetics of desensitization recovery, are enabled by the system. In addition, the system was validated via electrophysiological characterizations of both voltage-gated and ligand-gated ion channel targets: hK(V)2.1 and human Ether-à-go-go-related gene potassium channels, hNa(V)1.7 and 1.8 sodium channels, and (α1) hGABA(A) and (α1) human nicotinic acetylcholine receptor receptors. Our results show that the voltage dependence, kinetics, and interactions of these channels with pharmacological agents were matched to reference data. The results from these IonFlux™ experiments demonstrate that the system provides high-throughput automated electrophysiology with enhanced reliability and consistency, in a user-friendly format.
Collapse
Affiliation(s)
- C Ian Spencer
- Fluxion Biosciences, Inc., South San Francisco, California 94080, USA
| | | | | | | | | | | | | |
Collapse
|
50
|
Tetrodotoxin as a tool to elucidate sensory transduction mechanisms: the case for the arterial chemoreceptors of the carotid body. Mar Drugs 2011; 9:2683-2704. [PMID: 22363245 PMCID: PMC3280568 DOI: 10.3390/md9122683] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2011] [Revised: 11/22/2011] [Accepted: 12/01/2011] [Indexed: 12/17/2022] Open
Abstract
Carotid bodies (CBs) are secondary sensory receptors in which the sensing elements, chemoreceptor cells, are activated by decreases in arterial PO2 (hypoxic hypoxia). Upon activation, chemoreceptor cells (also known as Type I and glomus cells) increase their rate of release of neurotransmitters that drive the sensory activity in the carotid sinus nerve (CSN) which ends in the brain stem where reflex responses are coordinated. When challenged with hypoxic hypoxia, the physiopathologically most relevant stimulus to the CBs, they are activated and initiate ventilatory and cardiocirculatory reflexes. Reflex increase in minute volume ventilation promotes CO2 removal from alveoli and a decrease in alveolar PCO2 ensues. Reduced alveolar PCO2 makes possible alveolar and arterial PO2 to increase minimizing the intensity of hypoxia. The ventilatory effect, in conjunction the cardiocirculatory components of the CB chemoreflex, tend to maintain an adequate supply of oxygen to the tissues. The CB has been the focus of attention since the discovery of its nature as a sensory organ by de Castro (1928) and the discovery of its function as the origin of ventilatory reflexes by Heymans group (1930). A great deal of effort has been focused on the study of the mechanisms involved in O2 detection. This review is devoted to this topic, mechanisms of oxygen sensing. Starting from a summary of the main theories evolving through the years, we will emphasize the nature and significance of the findings obtained with veratridine and tetrodotoxin (TTX) in the genesis of current models of O2-sensing.
Collapse
|