1
|
Guo W, Yang H, Wang Y, Liu T, Pan Y, Chen X, Xu Q, Zhao D, Shan Z, Cai S. Small-molecule natural product sophoricoside reduces peripheral neuropathic pain via directly blocking of NaV1.6 in dorsal root ganglion nociceptive neurons. Neuropsychopharmacology 2024:10.1038/s41386-024-01998-w. [PMID: 39414988 DOI: 10.1038/s41386-024-01998-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/14/2024] [Revised: 08/23/2024] [Accepted: 09/16/2024] [Indexed: 10/18/2024]
Abstract
Peripheral neuropathic pain poses a significant global health challenge. Current drugs for peripheral neuropathic pain often fall short in efficacy or come with severe side effects, emphasizing the critical need for the development of highly effective and well-tolerated alternatives. Sophoricoside (SOP) is a nature product-derived isoflavone that possesses various pharmacological effects on inflammatory and neuropathy diseases. Here, in this study, analgesic effect was investigated by intrathecally administration of SOP/vehicle to spared nerve injury (SNI) or paclitaxel-induced peripheral neuropathic pain (PINP) rodent models, and mechanical allodynia was measured in Von Frey tests. Ipsilateral L4-L6 dorsal root ganglia (DRG) were used for protein expression. In silico molecular docking analysis was applied for assessing compound-target binding affinity. Primary cultured DRG neurons were utilized to investigate SOP's effect on veratridine-triggered nociceptor activities and its selective inhibition of voltage-gated sodium channels subtype 1.6 (NaV1.6). The results showed SOP treatment alleviated mechanical allodynia in SNI and PINP rodent models (paw withdrawal threshold after 1 h of injection: SNI-vehicle: 1.385 ± 0.338 g; SNI-SOP: 9.963 ± 2.029 g, P < 0.001; PINP-vehicle: 5.040 ± 0.985 g; PINP-SOP: 8.287 ± 3.812 g, P = 0.004). SOP presented effects on both inhibiting veratridine-triggered nociceptor activities (oscillatory population: vehicle: 39.9 ± 7.3%; SOP: 30.7 ± 9.8%, P = 0.021) and selectively blocking NaV1.6 in DRG sensory neurons. Molecular docking analysis indicated direct binding between SOP and NaV1.6, leading to its endocytosis in DRG Sensory Neurons. In conclusion, SOP alleviated nociceptive allodynia induced by peripheral nerve injury via selectively blocking of NaV1.6 in DRG nociceptive neurons. we highlight its potential as an analgesic and elucidate its mechanism involving NaV1.6 endocytosis. This research opens avenues for exploring the analgesic effects of SOP and its potential impact on neuropathic pain therapy.
Collapse
Affiliation(s)
- Weijie Guo
- Health Science Center, Shenzhen University, Shenzhen, China
| | - Haoyi Yang
- Department of Anesthesiology, Shenzhen People's Hospital (The First Affiliated Hospital, Southern University of Science and Technology), Shenzhen, China
| | - Yuwei Wang
- Health Science Center, Shenzhen University, Shenzhen, China
| | - Tao Liu
- Health Science Center, Shenzhen University, Shenzhen, China
| | - Yunping Pan
- Department of Periodontology & Oral Mucosa, Shenzhen Stomatology Hospital, Shenzhen, China
| | - Xiying Chen
- Department of Anesthesiology, Shenzhen People's Hospital (The First Affiliated Hospital, Southern University of Science and Technology), Shenzhen, China
| | - Qiuyin Xu
- Department of Anesthesiology, Shenzhen People's Hospital (The First Affiliated Hospital, Southern University of Science and Technology), Shenzhen, China
| | - Dizhou Zhao
- Department of Anesthesiology, Shenzhen People's Hospital (The First Affiliated Hospital, Southern University of Science and Technology), Shenzhen, China
| | - Zhiming Shan
- Department of Anesthesiology, Shenzhen People's Hospital (The First Affiliated Hospital, Southern University of Science and Technology), Shenzhen, China.
- Laboratory and Clinical Research Institute for Pain, Department of Anaesthesiology, School of Clinical Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, China.
| | - Song Cai
- Health Science Center, Shenzhen University, Shenzhen, China.
| |
Collapse
|
2
|
Baeza-Loya S, Eatock RA. Effects of transient, persistent, and resurgent sodium currents on excitability and spike regularity in vestibular ganglion neurons. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2023.11.28.569044. [PMID: 38076890 PMCID: PMC10705474 DOI: 10.1101/2023.11.28.569044] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/20/2023]
Abstract
Vestibular afferent neurons occur as two populations with differences in spike timing regularity that are independent of rate. The more excitable regular afferents have lower current thresholds and sustained spiking responses to injected currents, while irregular afferent neurons have higher thresholds and transient responses. Differences in expression of low-voltage-activated potassium (K LV ) channels are emphasized in models of spiking regularity and excitability in these neurons, leaving open the potential contributions of the voltage-gated sodium (Na V ) channels responsible for the spike upstroke. We investigated the impact of different Na V current modes (transient, persistent, and resurgent) with whole-cell patch clamp experiments in mouse vestibular ganglion neurons (VGNs), the cultured and dissociated cell bodies of afferents. All VGNs had transient Na V current, many had a small persistent (non-inactivating) Na V current, and a few had resurgent current, which flows after the spike peak when Na V channels that were blocked are unblocked. Na V 1.6 channels conducted most or all of each Na V current mode, and a Na V 1.6-selective blocker decreased spike rate and altered spike waveforms in both sustained and transient VGNs. A Na V channel agonist enhanced persistent current and increased spike rate and regularity. We hypothesized that persistent and resurgent currents have different effects on sustained (regular) VGNs vs. transient (irregular) VGNs. Lacking blockers specific for the different current modes, we used modeling to isolate their effects on spiking of simulated transient and sustained VGNs, driven by simulated current steps and noisy trains of simulated EPSCs. In all simulated neurons, increasing transient Na V current increased spike rate and rate-independent regularity. In simulated sustained VGNs, adding persistent current increased both rate and rate-independent regularity, while adding resurgent current had limited impact. In transient VGNs, adding persistent current had little impact, while adding resurgent current increased both rate and rate-independent irregularity by enhancing sensitivity to synaptic noise. These experiments show that the small Na V current modes may enhance the differentiation of afferent populations, with persistent currents selectively making regular afferents more regular and resurgent currents selectively making irregular afferents less regular.
Collapse
|
3
|
Yuan T, Wang Y, Jin Y, Yang H, Xu S, Zhang H, Chen Q, Li N, Ma X, Song H, Peng C, Geng Z, Dong J, Duan G, Sun Q, Yang Y, Yang F, Huang Z. Coupling of Slack and Na V1.6 sensitizes Slack to quinidine blockade and guides anti-seizure strategy development. eLife 2024; 12:RP87559. [PMID: 38289338 PMCID: PMC10942592 DOI: 10.7554/elife.87559] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/01/2024] Open
Abstract
Quinidine has been used as an anticonvulsant to treat patients with KCNT1-related epilepsy by targeting gain-of-function KCNT1 pathogenic mutant variants. However, the detailed mechanism underlying quinidine's blockade against KCNT1 (Slack) remains elusive. Here, we report a functional and physical coupling of the voltage-gated sodium channel NaV1.6 and Slack. NaV1.6 binds to and highly sensitizes Slack to quinidine blockade. Homozygous knockout of NaV1.6 reduces the sensitivity of native sodium-activated potassium currents to quinidine blockade. NaV1.6-mediated sensitization requires the involvement of NaV1.6's N- and C-termini binding to Slack's C-terminus and is enhanced by transient sodium influx through NaV1.6. Moreover, disrupting the Slack-NaV1.6 interaction by viral expression of Slack's C-terminus can protect against SlackG269S-induced seizures in mice. These insights about a Slack-NaV1.6 complex challenge the traditional view of 'Slack as an isolated target' for anti-epileptic drug discovery efforts and can guide the development of innovative therapeutic strategies for KCNT1-related epilepsy.
Collapse
Affiliation(s)
- Tian Yuan
- State Key Laboratory of Natural and Biomimetic Drugs, Department of Molecular and Cellular Pharmacology, School of Pharmaceutical Sciences, Peking University Health Science CenterBeijingChina
| | - Yifan Wang
- State Key Laboratory of Natural and Biomimetic Drugs, Department of Molecular and Cellular Pharmacology, School of Pharmaceutical Sciences, Peking University Health Science CenterBeijingChina
| | - Yuchen Jin
- State Key Laboratory of Natural and Biomimetic Drugs, Department of Molecular and Cellular Pharmacology, School of Pharmaceutical Sciences, Peking University Health Science CenterBeijingChina
| | - Hui Yang
- State Key Laboratory of Natural and Biomimetic Drugs, Department of Molecular and Cellular Pharmacology, School of Pharmaceutical Sciences, Peking University Health Science CenterBeijingChina
| | - Shuai Xu
- State Key Laboratory of Natural and Biomimetic Drugs, Department of Molecular and Cellular Pharmacology, School of Pharmaceutical Sciences, Peking University Health Science CenterBeijingChina
| | - Heng Zhang
- NHC and CAMS Key Laboratory of Medical Neurobiology, MOE Frontier Science Center for Brain Research and Brain-Machine Integration, School of Brain Science and Brain Medicine, Zhejiang UniversityZhejiangChina
| | - Qian Chen
- State Key Laboratory of Natural and Biomimetic Drugs, Department of Molecular and Cellular Pharmacology, School of Pharmaceutical Sciences, Peking University Health Science CenterBeijingChina
| | - Na Li
- State Key Laboratory of Natural and Biomimetic Drugs, Department of Molecular and Cellular Pharmacology, School of Pharmaceutical Sciences, Peking University Health Science CenterBeijingChina
| | - Xinyue Ma
- State Key Laboratory of Natural and Biomimetic Drugs, Department of Molecular and Cellular Pharmacology, School of Pharmaceutical Sciences, Peking University Health Science CenterBeijingChina
| | - Huifang Song
- State Key Laboratory of Natural and Biomimetic Drugs, Department of Molecular and Cellular Pharmacology, School of Pharmaceutical Sciences, Peking University Health Science CenterBeijingChina
| | - Chao Peng
- State Key Laboratory of Natural and Biomimetic Drugs, Department of Molecular and Cellular Pharmacology, School of Pharmaceutical Sciences, Peking University Health Science CenterBeijingChina
| | - Ze Geng
- State Key Laboratory of Natural and Biomimetic Drugs, Department of Molecular and Cellular Pharmacology, School of Pharmaceutical Sciences, Peking University Health Science CenterBeijingChina
| | - Jie Dong
- State Key Laboratory of Natural and Biomimetic Drugs, Department of Molecular and Cellular Pharmacology, School of Pharmaceutical Sciences, Peking University Health Science CenterBeijingChina
| | - Guifang Duan
- State Key Laboratory of Natural and Biomimetic Drugs, Department of Molecular and Cellular Pharmacology, School of Pharmaceutical Sciences, Peking University Health Science CenterBeijingChina
| | - Qi Sun
- State Key Laboratory of Natural and Biomimetic Drugs, Department of Molecular and Cellular Pharmacology, School of Pharmaceutical Sciences, Peking University Health Science CenterBeijingChina
| | - Yang Yang
- Department of Medicinal Chemistry and Molecular Pharmacology, College of Pharmacy, Purdue UniversityWest LafayetteUnited States
| | - Fan Yang
- NHC and CAMS Key Laboratory of Medical Neurobiology, MOE Frontier Science Center for Brain Research and Brain-Machine Integration, School of Brain Science and Brain Medicine, Zhejiang UniversityZhejiangChina
- Department of Biophysics, Kidney Disease Center of the First Affiliated Hospital, Zhejiang University School of Medicine, HangzhouZhejiangChina
| | - Zhuo Huang
- State Key Laboratory of Natural and Biomimetic Drugs, Department of Molecular and Cellular Pharmacology, School of Pharmaceutical Sciences, Peking University Health Science CenterBeijingChina
- IDG/McGovern Institute for Brain Research, Peking UniversityBeijingChina
| |
Collapse
|
4
|
Drouillas B, Brocard C, Zanella S, Bos R, Brocard F. Persistent Nav1.1 and Nav1.6 currents drive spinal locomotor functions through nonlinear dynamics. Cell Rep 2023; 42:113085. [PMID: 37665666 DOI: 10.1016/j.celrep.2023.113085] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2023] [Revised: 06/29/2023] [Accepted: 08/16/2023] [Indexed: 09/06/2023] Open
Abstract
Persistent sodium current (INaP) in the spinal locomotor network promotes two distinct nonlinear firing patterns: a self-sustained spiking triggered by a brief excitation in bistable motoneurons and bursting oscillations in interneurons of the central pattern generator (CPG). Here, we identify the NaV channels responsible for INaP and their role in motor behaviors. We report the axonal Nav1.6 as the main molecular player for INaP in lumbar motoneurons. The inhibition of Nav1.6, but not of Nav1.1, in motoneurons impairs INaP, bistability, postural tone, and locomotor performance. In interneurons of the rhythmogenic CPG region, both Nav1.6 and Nav1.1 equally mediate INaP. Inhibition of both channels is required to abolish oscillatory bursting activities and the locomotor rhythm. Overall, Nav1.6 plays a significant role both in posture and locomotion by governing INaP-dependent bistability in motoneurons and working in tandem with Nav1.1 to provide INaP-dependent rhythmogenic properties of the CPG.
Collapse
Affiliation(s)
- Benoît Drouillas
- Institut de Neurosciences de la Timone, UMR 7289, Aix-Marseille Université and Centre National de la Recherche Scientifique (CNRS), Marseille, France
| | - Cécile Brocard
- Institut de Neurosciences de la Timone, UMR 7289, Aix-Marseille Université and Centre National de la Recherche Scientifique (CNRS), Marseille, France
| | - Sébastien Zanella
- Institut de Neurosciences de la Timone, UMR 7289, Aix-Marseille Université and Centre National de la Recherche Scientifique (CNRS), Marseille, France
| | - Rémi Bos
- Institut de Neurosciences de la Timone, UMR 7289, Aix-Marseille Université and Centre National de la Recherche Scientifique (CNRS), Marseille, France
| | - Frédéric Brocard
- Institut de Neurosciences de la Timone, UMR 7289, Aix-Marseille Université and Centre National de la Recherche Scientifique (CNRS), Marseille, France.
| |
Collapse
|
5
|
Someck S, Levi A, Sloin HE, Spivak L, Gattegno R, Stark E. Positive and biphasic extracellular waveforms correspond to return currents and axonal spikes. Commun Biol 2023; 6:950. [PMID: 37723241 PMCID: PMC10507124 DOI: 10.1038/s42003-023-05328-6] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2023] [Accepted: 09/06/2023] [Indexed: 09/20/2023] Open
Abstract
Multiple biophysical mechanisms may generate non-negative extracellular waveforms during action potentials, but the origin and prevalence of positive spikes and biphasic spikes in the intact brain are unknown. Using extracellular recordings from densely-connected cortical networks in freely-moving mice, we find that a tenth of the waveforms are non-negative. Positive phases of non-negative spikes occur in synchrony or just before wider same-unit negative spikes. Narrow positive spikes occur in isolation in the white matter. Isolated biphasic spikes are narrower than negative spikes, occurring right after spikes of verified inhibitory units. In CA1, units with dominant non-negative spikes exhibit place fields, phase precession, and phase-locking to ripples. Thus, near-somatic narrow positive extracellular potentials correspond to return currents, and isolated non-negative spikes correspond to axonal potentials. Identifying non-negative extracellular waveforms that correspond to non-somatic compartments during spikes can enhance the understanding of physiological and pathological neural mechanisms in intact animals.
Collapse
Affiliation(s)
- Shirly Someck
- Sagol School of Neuroscience, Tel Aviv University, Tel Aviv, 6997801, Israel
- Department of Physiology and Pharmacology, Faculty of Medicine, Tel Aviv University, Tel Aviv, 6997801, Israel
| | - Amir Levi
- Sagol School of Neuroscience, Tel Aviv University, Tel Aviv, 6997801, Israel
- Department of Physiology and Pharmacology, Faculty of Medicine, Tel Aviv University, Tel Aviv, 6997801, Israel
| | - Hadas E Sloin
- Sagol School of Neuroscience, Tel Aviv University, Tel Aviv, 6997801, Israel
- Department of Physiology and Pharmacology, Faculty of Medicine, Tel Aviv University, Tel Aviv, 6997801, Israel
| | - Lidor Spivak
- Sagol School of Neuroscience, Tel Aviv University, Tel Aviv, 6997801, Israel
- Department of Physiology and Pharmacology, Faculty of Medicine, Tel Aviv University, Tel Aviv, 6997801, Israel
| | - Roni Gattegno
- Sagol School of Neuroscience, Tel Aviv University, Tel Aviv, 6997801, Israel
| | - Eran Stark
- Sagol School of Neuroscience, Tel Aviv University, Tel Aviv, 6997801, Israel.
- Department of Physiology and Pharmacology, Faculty of Medicine, Tel Aviv University, Tel Aviv, 6997801, Israel.
- Sagol Department of Neurobiology, Haifa University, Haifa, 3103301, Israel.
| |
Collapse
|
6
|
Structure of human Na V1.6 channel reveals Na + selectivity and pore blockade by 4,9-anhydro-tetrodotoxin. Nat Commun 2023; 14:1030. [PMID: 36823201 PMCID: PMC9950489 DOI: 10.1038/s41467-023-36766-9] [Citation(s) in RCA: 9] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2022] [Accepted: 02/15/2023] [Indexed: 02/25/2023] Open
Abstract
The sodium channel NaV1.6 is widely expressed in neurons of the central and peripheral nervous systems, which plays a critical role in regulating neuronal excitability. Dysfunction of NaV1.6 has been linked to epileptic encephalopathy, intellectual disability and movement disorders. Here we present cryo-EM structures of human NaV1.6/β1/β2 alone and complexed with a guanidinium neurotoxin 4,9-anhydro-tetrodotoxin (4,9-ah-TTX), revealing molecular mechanism of NaV1.6 inhibition by the blocker. The apo-form structure reveals two potential Na+ binding sites within the selectivity filter, suggesting a possible mechanism for Na+ selectivity and conductance. In the 4,9-ah-TTX bound structure, 4,9-ah-TTX binds to a pocket similar to the tetrodotoxin (TTX) binding site, which occupies the Na+ binding sites and completely blocks the channel. Molecular dynamics simulation results show that subtle conformational differences in the selectivity filter affect the affinity of TTX analogues. Taken together, our results provide important insights into NaV1.6 structure, ion conductance, and inhibition.
Collapse
|
7
|
Riluzole and novel naphthalenyl substituted aminothiazole derivatives prevent acute neural excitotoxic injury in a rat model of temporal lobe epilepsy. Neuropharmacology 2023; 224:109349. [PMID: 36436594 PMCID: PMC9843824 DOI: 10.1016/j.neuropharm.2022.109349] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2022] [Revised: 11/07/2022] [Accepted: 11/17/2022] [Indexed: 11/25/2022]
Abstract
Epileptogenic seizures, or status epilepticus (SE), leads to excitotoxic injury in hippocampal and limbic neurons in the kainic acid (KA) animal model of temporal lobe epilepsy (TLE). Here, we have further characterized neural activity regulated methylaminoisobutryic acid (MeAIB)/glutamine transport activity in mature rat hippocampal neurons in vitro that is inhibited by riluzole (IC50 = 1 μM), an anti-convulsant benzothiazole agent. We screened a library of riluzole derivatives and identified SKA-41 followed by a second screen and synthesized several novel chlorinated aminothiazoles (SKA-377, SKA-378, SKA-379) that are also potent MeAIB transport inhibitors in vitro, and brain penetrant following systemic administration. When administered before KA, SKA-378 did not prevent seizures but still protected the hippocampus and several other limbic areas against SE-induced neurodegeneration at 3d. When SKA-377 - 379, (30 mg/kg) were administered after KA-induced SE, acute neural injury in the CA3, CA1 and CA4/hilus was also largely attenuated. Riluzole (10 mg/kg) blocks acute neural injury. Kinetic analysis of SKA-378 and riluzoles' blockade of Ca2+-regulated MeAIB transport in neurons in vitro indicates that inhibition occurs via a non-competitive, indirect mechanism. Sodium channel NaV1.6 antagonism blocks neural activity regulated MeAIB/Gln transport in vitro (IC50 = 60 nM) and SKA-378 is the most potent inhibitor of NaV1.6 (IC50 = 28 μM) compared to NaV1.2 (IC50 = 118 μM) in heterologous cells. However, pharmacokinetic analysis suggests that sodium channel blockade may not be the predominant mechanism of neuroprotection here. Riluzole and our novel aminothiazoles are agents that attenuate acute neural hippocampal injury following KA-induced SE and may help to understand mechanisms involved in the progression of epileptic disease.
Collapse
|
8
|
Thouta S, Waldbrook MG, Lin S, Mahadevan A, Mezeyova J, Soriano M, Versi P, Goodchild SJ, Parrish RR. Pharmacological determination of the fractional block of Nav channels required to impair neuronal excitability and ex vivo seizures. Front Cell Neurosci 2022; 16:964691. [PMID: 36246527 PMCID: PMC9557217 DOI: 10.3389/fncel.2022.964691] [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: 06/09/2022] [Accepted: 08/31/2022] [Indexed: 11/13/2022] Open
Abstract
Voltage-gated sodium channels (Nav) are essential for the initiation and propagation of action potentials in neurons. Of the nine human channel subtypes, Nav1.1, Nav1.2 and Nav1.6 are prominently expressed in the adult central nervous system (CNS). All three of these sodium channel subtypes are sensitive to block by the neurotoxin tetrodotoxin (TTX), with TTX being almost equipotent on all three subtypes. In the present study we have used TTX to determine the fractional block of Nav channels required to impair action potential firing in pyramidal neurons and reduce network seizure-like activity. Using automated patch-clamp electrophysiology, we first determined the IC50s of TTX on mouse Nav1.1, Nav1.2 and Nav1.6 channels expressed in HEK cells, demonstrating this to be consistent with previously published data on human orthologs. We then compared this data to the potency of block of Nav current measured in pyramidal neurons from neocortical brain slices. Interestingly, we found that it requires nearly 10-fold greater concentration of TTX over the IC50 to induce significant block of action potentials using a current-step protocol. In contrast, concentrations near the IC50 resulted in a significant reduction in AP firing and increase in rheobase using a ramp protocol. Surprisingly, a 20% reduction in action potential generation observed with 3 nM TTX resulted in significant block of seizure-like activity in the 0 Mg2+ model of epilepsy. Additionally, we found that approximately 50% block in pyramidal cell intrinsic excitability is sufficient to completely block all seizure-like events. Furthermore, we also show that the anticonvulsant drug phenytoin blocked seizure-like events in a manner similar to TTX. These data serve as a critical starting point in understanding how fractional block of Nav channels affect intrinsic neuronal excitability and seizure-like activity. It further suggests that seizures can be controlled without significantly compromising intrinsic neuronal activity and determines the required fold over IC50 for novel and clinically relevant Nav channel blockers to produce efficacy and limit side effects.
Collapse
Affiliation(s)
- Samrat Thouta
- Department of Cellular and Molecular Biology, Xenon Pharmaceuticals Inc., Burnaby, BC, Canada
| | - Matthew G. Waldbrook
- Department of Cellular and Molecular Biology, Xenon Pharmaceuticals Inc., Burnaby, BC, Canada
| | - Sophia Lin
- Department of Cellular and Molecular Biology, Xenon Pharmaceuticals Inc., Burnaby, BC, Canada
| | - Arjun Mahadevan
- Department of Cellular and Molecular Biology, Xenon Pharmaceuticals Inc., Burnaby, BC, Canada
| | - Janette Mezeyova
- Department of Cellular and Molecular Biology, Xenon Pharmaceuticals Inc., Burnaby, BC, Canada
| | - Maegan Soriano
- Department of Cellular and Molecular Biology, Xenon Pharmaceuticals Inc., Burnaby, BC, Canada
| | - Pareesa Versi
- Department of Cellular and Molecular Biology, Xenon Pharmaceuticals Inc., Burnaby, BC, Canada
| | - Samuel J. Goodchild
- Department of Cellular and Molecular Biology, Xenon Pharmaceuticals Inc., Burnaby, BC, Canada
| | - R. Ryley Parrish
- Department of Cellular and Molecular Biology, Xenon Pharmaceuticals Inc., Burnaby, BC, Canada
- Department of Cell Biology and Physiology, Brigham Young University, Provo, UT, United States
| |
Collapse
|
9
|
Matthews RM, Bradley E, Griffin CS, Lim XR, Mullins ND, Hollywood MA, Lundy FT, McGarvey LP, Sergeant GP, Thornbury KD. Functional expression of Na V1.7 channels in freshly dispersed mouse bronchial smooth muscle cells. Am J Physiol Cell Physiol 2022; 323:C749-C762. [DOI: 10.1152/ajpcell.00011.2022] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Isolated smooth muscle cells (SMC) from mouse bronchus were studied using the whole-cell patch clamp technique at ~21oC. Stepping from -100 mV to -20 mV evoked inward currents of mean amplitude -275 pA. These inactivated (tau=1.1 ms) and were abolished when external Na+ was substituted with N-Methyl-D-glucamine. In current-voltage protocols, current peaked at -10 mV and reversed between +20 and +30 mV. The V1/2s of activation and inactivation were -25 & -86 mV, respectively. The current was highly sensitive to tetrodotoxin (IC50=1.5 nM) and the NaV1.7 subtype selective blocker, PF-05089771 (IC50=8.6 nM), consistent with NaV1.7 as the underlying pore-forming a subunit. Two NaV1.7-selective antibodies caused membrane-delineated staining of isolated SMC, as did a non-selective pan-NaVantibody. RT-PCR, performed on groups of ~15 isolated SMC, revealed transcripts for NaV1.7 in 7/8 samples. Veratridine (30 mM), a non-selective NaV channel activator, reduced peak current evoked by depolarization but induced a sustained current of 40 pA. Both effects were reversed by tetrodotoxin (100 nM). In tension experiments veratridine (10 mM) induced contractions that were entirely blocked by atropine (1 mM). However, in the presence of atropine, veratridine was able to modulate the pattern of activity induced by a combination of U-46619 (a thromboxane A2 mimetic) & PGE2(prostaglandin E2), by eliminating bursts in favour of sustained phasic contractions. These effects were readily reversed to control-like activity by tetrodotoxin (100 nM). In conclusion, mouse bronchial SMC functionally express NaV1.7 channels that are capable of modulating contractile activity, at least under experimental conditions.
Collapse
Affiliation(s)
- Ruth M. Matthews
- Smooth Muscle Research Centre, Dundalk Institute of Technology, Dundalk, County Louth, Ireland
| | - Eamonn Bradley
- Smooth Muscle Research Centre, Dundalk Institute of Technology, Dundalk, County Louth, Ireland
| | - Caoimhin S. Griffin
- Smooth Muscle Research Centre, Dundalk Institute of Technology, Dundalk, County Louth, Ireland
| | - Xin Rui Lim
- Smooth Muscle Research Centre, Dundalk Institute of Technology, Dundalk, County Louth, Ireland
| | - Nicolas D. Mullins
- Smooth Muscle Research Centre, Dundalk Institute of Technology, Dundalk, County Louth, Ireland
| | - Mark A. Hollywood
- Smooth Muscle Research Centre, Dundalk Institute of Technology, Dundalk, County Louth, Ireland
| | - Fionnuala T. Lundy
- Smooth Muscle Research Centre, Dundalk Institute of Technology, Dundalk, County Louth, Ireland
| | - Lorcan P. McGarvey
- Smooth Muscle Research Centre, Dundalk Institute of Technology, Dundalk, County Louth, Ireland
| | - Gerard P. Sergeant
- Smooth Muscle Research Centre, Dundalk Institute of Technology, Dundalk, County Louth, Ireland
| | - Keith D. Thornbury
- Smooth Muscle Research Centre, Dundalk Institute of Technology, Dundalk, County Louth, Ireland
| |
Collapse
|
10
|
Wienbar S, Schwartz GW. Differences in spike generation instead of synaptic inputs determine the feature selectivity of two retinal cell types. Neuron 2022; 110:2110-2123.e4. [PMID: 35508174 PMCID: PMC9262831 DOI: 10.1016/j.neuron.2022.04.012] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2021] [Revised: 03/21/2022] [Accepted: 04/11/2022] [Indexed: 12/19/2022]
Abstract
Retinal ganglion cells (RGCs) are the spiking projection neurons of the eye that encode different features of the visual environment. The circuits providing synaptic input to different RGC types to drive feature selectivity have been studied extensively, but there has been less research aimed at understanding the intrinsic properties and how they impact feature selectivity. We introduce an RGC type in the mouse, the Bursty Suppressed-by-Contrast (bSbC) RGC, and compared it to the OFF sustained alpha (OFFsA). Differences in their contrast response functions arose from differences not in synaptic inputs but in their intrinsic properties. Spike generation was the key intrinsic property behind this functional difference; the bSbC RGC undergoes depolarization block while the OFFsA RGC maintains a high spike rate. Our results demonstrate that differences in intrinsic properties allow these two RGC types to detect and relay distinct features of an identical visual stimulus to the brain.
Collapse
Affiliation(s)
- Sophia Wienbar
- Department of Ophthalmology, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA; Northwestern University Interdepartmental Neuroscience Program, Northwestern University, Evanston, IL 60208, USA
| | - Gregory William Schwartz
- Department of Ophthalmology, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA; Department of Neuroscience, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA; Department of Neurobiology, Weinberg College of Arts and Sciences, Northwestern University, Evanston, IL 60208, USA.
| |
Collapse
|
11
|
Elleman AV, Du Bois J. Chemical and Biological Tools for the Study of Voltage-Gated Sodium Channels in Electrogenesis and Nociception. Chembiochem 2022; 23:e202100625. [PMID: 35315190 PMCID: PMC9359671 DOI: 10.1002/cbic.202100625] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2021] [Revised: 02/22/2022] [Indexed: 12/17/2022]
Abstract
The malfunction and misregulation of voltage-gated sodium channels (NaV s) underlie in large part the electrical hyperexcitability characteristic of chronic inflammatory and neuropathic pain. NaV s are responsible for the initiation and propagation of electrical impulses (action potentials) in cells. Tissue and nerve injury alter the expression and localization of multiple NaV isoforms, including NaV 1.1, 1.3, and 1.6-1.9, resulting in aberrant action potential firing patterns. To better understand the role of NaV regulation, localization, and trafficking in electrogenesis and pain pathogenesis, a number of chemical and biological reagents for interrogating NaV function have been advanced. The development and application of such tools for understanding NaV physiology are the focus of this review.
Collapse
Affiliation(s)
- Anna V Elleman
- Department of Chemistry, Stanford University, Stanford, CA 94305, USA
| | - J Du Bois
- Department of Chemistry, Stanford University, Stanford, CA 94305, USA
| |
Collapse
|
12
|
Goodwin G, McMurray S, Stevens EB, Denk F, McMahon SB. Examination of the contribution of Nav1.7 to axonal propagation in nociceptors. Pain 2022; 163:e869-e881. [PMID: 34561392 DOI: 10.1097/j.pain.0000000000002490] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2021] [Accepted: 09/09/2021] [Indexed: 11/25/2022]
Abstract
ABSTRACT Nav1.7 is a promising drug target for the treatment of pain. However, there is a mismatch between the analgesia produced by Nav1.7 loss-of-function and the peripherally restricted Nav1.7 inhibitors, which may reflect a lack of understanding of the function of Nav1.7 in the transmission of nociceptive information. In the periphery, the role of Nav1.7 in transduction at nociceptive peripheral terminals has been comprehensively examined, but its role in axonal propagation in these neurons is less clearly defined. In this study, we examined the contribution of Nav1.7 to axonal propagation in nociceptors using sodium channel blockers in in vivo electrophysiological and calcium imaging recordings in mice. Using the sodium channel blocker tetrodotoxin (TTX) (1-10 µM) to inhibit Nav1.7 and other tetrodotoxin-sensitive sodium channels along the sciatic nerve, we first showed that around two-thirds of nociceptive L4 dorsal root ganglion neurons innervating the skin, but a lower proportion innervating the muscle (45%), are blocked by TTX. By contrast, nearly all large-sized cutaneous afferents (95%-100%) were blocked by axonal TTX. Many cutaneous nociceptors resistant to TTX were polymodal (57%) and capsaicin sensitive (57%). Next, we applied PF-05198007 (300 nM-1 µM) to the sciatic nerve between stimulating and recording sites to selectively block axonal Nav1.7 channels. One hundred to three hundred nanomolar PF-05198007 blocked propagation in 63% of C-fiber sensory neurons, whereas similar concentrations produced minimal block (5%) in rapidly conducting A-fiber neurons. We conclude that Nav1.7 is essential for axonal propagation in around two-thirds of nociceptive cutaneous C-fiber neurons and a lower proportion (≤45%) of nociceptive neurons innervating muscle.
Collapse
Affiliation(s)
- George Goodwin
- Neurorestoration Group, Wolfson Centre for Age-Related Diseases, King's College London, United Kingdom
| | | | | | - Franziska Denk
- Neurorestoration Group, Wolfson Centre for Age-Related Diseases, King's College London, United Kingdom
| | - Stephen B McMahon
- Neurorestoration Group, Wolfson Centre for Age-Related Diseases, King's College London, United Kingdom
| |
Collapse
|
13
|
Pearson KC, Tarvin RD. A review of chemical defense in harlequin toads (Bufonidae: Atelopus). Toxicon X 2022; 13:100092. [PMID: 35146414 PMCID: PMC8801762 DOI: 10.1016/j.toxcx.2022.100092] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2021] [Revised: 01/12/2022] [Accepted: 01/17/2022] [Indexed: 12/29/2022] Open
Abstract
Toads of the genus Atelopus are chemically defended by a unique combination of endogenously synthesized cardiotoxins (bufadienolides) and neurotoxins which may be sequestered (guanidinium alkaloids). Investigation into Atelopus small-molecule chemical defenses has been primarily concerned with identifying and characterizing various forms of these toxins while largely overlooking their ecological roles and evolutionary implications. In addition to describing the extent of knowledge about Atelopus toxin structures, pharmacology, and biological sources, we review the detection, identification, and quantification methods used in studies of Atelopus toxins to date and conclude that many known toxin profiles are unlikely to be comprehensive because of methodological and sampling limitations. Patterns in existing data suggest that both environmental (toxin availability) and genetic (capacity to synthesize or sequester toxins) factors influence toxin profiles. From an ecological and evolutionary perspective, we summarize the possible selective pressures acting on Atelopus toxicity and toxin profiles, including predation, intraspecies communication, disease, and reproductive status. Ultimately, we intend to provide a basis for future ecological, evolutionary, and biochemical research on Atelopus.
Collapse
Affiliation(s)
- Kannon C. Pearson
- Museum of Vertebrate Zoology and Department of Integrative Biology, University of California Berkeley, Berkeley, CA, 94720, USA
| | - Rebecca D. Tarvin
- Museum of Vertebrate Zoology and Department of Integrative Biology, University of California Berkeley, Berkeley, CA, 94720, USA
| |
Collapse
|
14
|
Mulpuri Y, Yamamoto T, Nishimura I, Spigelman I. Role of voltage-gated sodium channels in axonal signal propagation of trigeminal ganglion neurons after infraorbital nerve entrapment. NEUROBIOLOGY OF PAIN 2022; 11:100084. [PMID: 35128176 PMCID: PMC8803652 DOI: 10.1016/j.ynpai.2022.100084] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/15/2021] [Revised: 01/17/2022] [Accepted: 01/17/2022] [Indexed: 11/25/2022]
Abstract
Infraorbital nerve entrapment (IoNE) induces mechanical allodynia and enhances signal propagation in primary afferent A- and C-fibers. IoNE increases sensitivity of A- and C-fibers to conduction block by tetrodotoxin (TTX) and selective voltage-gated sodium channel 1.8 (NaV1.8) inhibitor, A-803467. IoNE increases signal propagation in vibrissal pad Ad -, but not Aβ-fibers, and their sensitivity to conduction block by the selective NaV1.8 inhibitor. IoNE increases membrane excitability of dissociated small and medium sized trigeminal neurons. IoNE increases nerve, but not ganglion, levels of NaV1.3, NaV1.7, and NaV1.8 mRNAs, and NaV1.8 protein.
Chronic pain arising from peripheral nerve injuries represents a significant clinical challenge because even the most efficacious anticonvulsant drug treatments are limited by their side effects profile. We investigated pain behavior, changes in axonal signal conduction and excitability of trigeminal neurons, and expression of voltage-gated sodium channels (NaVs) in the infraorbital nerve and trigeminal ganglion (TG) after infraorbital nerve entrapment (IoNE). Compared to Sham, IoNE rats had increased A- and C-fiber compound action potentials (CAPs) and Aδ component of A-CAP area from fibers innervating the vibrissal pad. After IoNE, A- and C-fiber CAPs were more sensitive to blockade by tetrodotoxin (TTX), and those fibers that were TTX-resistant were more sensitive to blockade by the NaV1.8 selective blocker, A-803467. Although NaV1.7 blocker, ICA-121431 alone, did not affect Aδ-fiber signal propagation, cumulative application with A-803467 and 4,9-anhydro-TTX significantly reduced the Aδ-fiber CAP in IoNE rats. In patch clamp recordings from small- and medium-sized TG neurons, IoNE resulted in reduced action potential (AP) depolarizing current threshold, hyperpolarized AP voltage threshold, increased AP duration, and a more depolarized membrane potential. While the transcripts of most NaVs were reduced in the ipsilateral TG after IoNE, NaV1.3, NaV1.7, and NaV1.8 mRNAs, and NaV1.8 protein, were significantly increased in the nerve. Altogether, our data suggest that axonal redistribution of NaV1.8, and to a lesser extent NaV1.3, and NaV1.7 contributes to enhanced nociceptive signal propagation in peripheral nerve after IoNE.
Collapse
|
15
|
Lim XR, Bradley E, Griffin CS, Hollywood MA, Sergeant GP, Thornbury KD. Fast voltage-dependent sodium (Na V ) currents are functionally expressed in mouse corpus cavernosum smooth muscle cells. Br J Pharmacol 2021; 179:1082-1101. [PMID: 34767251 DOI: 10.1111/bph.15728] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2021] [Revised: 10/17/2021] [Accepted: 10/19/2021] [Indexed: 12/01/2022] Open
Abstract
BACKGROUND AND PURPOSE Corpus cavernosum smooth muscle (CCSM) exhibits phasic contractions that are coordinated by ion channels. Mouse models are commonly used to study erectile dysfunction, but there are few published electrophysiological studies of mouse CCSM. We describe, for the first time, voltage-dependent sodium (NaV ) currents in mouse CCSM and investigate their function. EXPERIMENTAL APPROACH Electrophysiological, pharmacological, and immunocytochemical studies on isolated CCSM cells. Tension measurements in whole tissue. KEY RESULTS A fast, voltage-dependent sodium current was induced by depolarising steps. Steady-state activation and inactivation curves revealed a window current between -60 and -30 mV. Two populations of NaV currents, ('TTX-sensitive') and ('TTX-insensitive'), were distinguished. TTX-sensitive current showed 48% block with the NaV -subtype-specific blockers ICA-121431 (NaV 1.1-1.3), PF-05089771 (NaV 1.7), and 4,9-anhydro-TTX (NaV 1.6). TTX-insensitive current was insensitive to A803467, a NaV 1.8 blocker. Immunocytochemistry confirmed the expression of NaV 1.5 and NaV 1.4 in freshly dispersed CCSM cells. Veratridine, a NaV activator, reduced time-dependent inactivation of the current and increased the duration of evoked action potentials. Veratridine induced phasic contractions in CCSM strips. This effect was reversible with TTX and nifedipine but not by KB-R7943. CONCLUSION AND IMPLICATIONS We report, for the first time, a fast voltage-dependent sodium current in mouse CCSM. Stimulation of this current increases the contractility of corpus cavernosum in vitro, suggesting that it may contribute to the mechanisms of detumescence, and potentially serve as a clinically relevant target for pharmaceutical intervention in erectile dysfunction. Further work will be necessary to define its role.
Collapse
Affiliation(s)
| | | | | | | | | | - Keith D Thornbury
- Smooth Muscle Research Centre Dundalk Institute of Technology, Dublin, Ireland
| |
Collapse
|
16
|
Mechanisms Underlying the Selective Therapeutic Efficacy of Carbamazepine for Attenuation of Trigeminal Nerve Injury Pain. J Neurosci 2021; 41:8991-9007. [PMID: 34446571 DOI: 10.1523/jneurosci.0547-21.2021] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2021] [Revised: 08/02/2021] [Accepted: 08/03/2021] [Indexed: 11/21/2022] Open
Abstract
Different peripheral nerve injuries cause neuropathic pain through distinct mechanisms. Even the site of injury may impact underlying mechanisms, as indicated by the clinical finding that the antiseizure drug carbamazepine (CBZ) relieves pain because of compression injuries of trigeminal but not somatic nerves. We leveraged this observation in the present study hypothesizing that because CBZ blocks voltage-gated sodium channels (VGSCs), its therapeutic selectivity reflects differences between trigeminal and somatic nerves with respect to injury-induced changes in VGSCs. CBZ diminished ongoing and evoked pain behavior in rats with chronic constriction injury (CCI) to the infraorbital nerve (ION) but had minimal effect in rats with sciatic nerve CCI. This difference in behavior was associated with a selective increase in the potency of CBZ-induced inhibition of compound action potentials in the ION, an effect mirrored in human trigeminal versus somatic nerves. The increase in potency was associated with a selective increase in the efficacy of the NaV1.1 channel blocker ICA-121431 and NaV1.1 protein in the ION, but no change in NaV1.1 mRNA in trigeminal ganglia. Importantly, local ICA-121431 administration reversed ION CCI-induced hypersensitivity. Our results suggest a novel therapeutic target for the treatment of trigeminal neuropathic pain.SIGNIFICANCE STATEMENT This study is based on evidence of differences in pain and its treatment depending on whether the pain is above (trigeminal) or below (somatic) the neck, as well as evidence that voltage-gated sodium channels (VGSCs) may contribute to these differences. The focus of the present study was on channels underlying action potential propagation in peripheral nerves. There were differences between somatic and trigeminal nerves in VGSC subtypes underlying action potential propagation both in the absence and presence of injury. Importantly, because the local block of NaV1.1 in the trigeminal nerve reverses nerve injury-induced mechanical hypersensitivity, the selective upregulation of NaV1.1 in trigeminal nerves suggests a novel therapeutic target for the treatment of pain associated with trigeminal nerve injury.
Collapse
|
17
|
A Buthus martensii Karsch scorpion sting targets Nav1.7 in mice and mimics a phenotype of human chronic pain. Pain 2021; 163:e202-e214. [PMID: 34252912 DOI: 10.1097/j.pain.0000000000002397] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2021] [Accepted: 06/28/2021] [Indexed: 11/25/2022]
Abstract
GAIN and loss-of-function mutations in Nav1.7 cause chronic pain and pain insensitivity, respectively. The preferential expression of Nav1.7 in peripheral nervous system and its role in human pain signaling make Nav1.7 a promising target for next-generation pain therapeutics. However, pharmacological agents have not fully recapitulated these pain phenotypes, and, due to the lack of subtype-selective molecular modulators, the role of Nav1.7 in the perception of pain remains poorly understood. Scorpion venom is an excellent source of bioactive peptides that modulate various ion channels, including voltage-gated sodium (Nav) channels . Here, we demonstrate that Buthus martensii Karsch scorpion venom (BV) elicits pain responses in mice through direct enhancement of Nav1.7 activity, and have identified that Makatoxin-3, an α-like toxin as a critical component for BV-mediated effects on Nav1.7. Blocking other Nav subtypes did not eliminate BV-evoked pain responses, supporting the pivotal role of Nav1.7 in BV-induced pain . Makatoxin-3 acts on the S3-S4 loop of voltage sensor domain IV (VSD4) of Nav1.7, which causes a hyperpolarizing shift in the steady-state fast inactivation and impairs inactivation kinetics. We also determined the key residues and structure-function relationships for the toxin-channel interactions, which are distinct from those of other well-studied α-toxins. This study not only reveals a new mechanism underlying BV-evoked pain, but also enriches our knowledge of key structural elements of scorpion toxins that are pivotal for toxin-Nav1.7 interaction, which facilitates the design of novel Nav1.7 selective modulators.
Collapse
|
18
|
Mohammed ZA, Kaloyanova K, Nassar MA. An unbiased and efficient assessment of excitability of sensory neurons for analgesic drug discovery. Pain 2021; 161:1100-1108. [PMID: 31929383 PMCID: PMC7170445 DOI: 10.1097/j.pain.0000000000001802] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
Distinct veratridine-induced calcium responses in nociceptors and non-nociceptors allow for unbiased and efficient assessment of drugs' action on both populations separately but simultaneously. Alleviating chronic pain is challenging, due to lack of drugs that effectively inhibit nociceptors without off-target effects on motor or central neurons. Dorsal root ganglia (DRG) contain nociceptive and non-nociceptive neurons. Drug screening on cultured DRG neurons, rather than cell lines, allows for the identification of drugs most potent on nociceptors with no effects on non-nociceptors (as a proxy for unwanted side effects on central nervous system and motor neurons). However, screening using DRG neurons is currently a low-throughput process, and there is a need for assays to speed this process for analgesic drug discovery. We previously showed that veratridine elicits distinct response profiles in sensory neurons. Here, we show evidence that a veratridine-based calcium assay allows for an unbiased and efficient assessment of a drug effect on nociceptors (targeted neurons) and non-nociceptors (nontargeted neurons). We confirmed the link between the oscillatory profile and nociceptors, and the slow-decay profile and non-nociceptors using 3 transgenic mouse lines of known pain phenotypes. We used the assay to show that blockers for Nav1.7 and Nav1.8 channels, which are validated targets for analgesics, affect non-nociceptors at concentrations needed to effectively inhibit nociceptors. However, a combination of low doses of both blockers had an additive effect on nociceptors without a significant effect on non-nociceptors, indicating that the assay can also be used to screen for combinations of existing or novel drugs for the greatest selective inhibition of nociceptors.
Collapse
Affiliation(s)
- Zainab A Mohammed
- Department of Biomedical Science, University of Sheffield, Sheffield, United Kingdom
| | | | | |
Collapse
|
19
|
Nishikawa K, Noguchi T, Kikuchi S, Maruyama T, Araki Y, Yotsu-Yamashita M, Morimoto Y. Tetrodotoxin Framework Construction from Linear Substrates Utilizing a Hg(OTf)2-Catalyzed Cycloisomerization Reaction: Synthesis of the Unnatural Analogue 11-nor-6,7,8-Trideoxytetrodotoxin. Org Lett 2021; 23:1703-1708. [DOI: 10.1021/acs.orglett.1c00125] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2023]
Affiliation(s)
- Keisuke Nishikawa
- Department of Chemistry, Graduate School of Science, Osaka City University, Osaka, Osaka 558-8585, Japan
| | - Takayuki Noguchi
- Department of Chemistry, Graduate School of Science, Osaka City University, Osaka, Osaka 558-8585, Japan
| | - Seiho Kikuchi
- Department of Chemistry, Graduate School of Science, Osaka City University, Osaka, Osaka 558-8585, Japan
| | - Takahiro Maruyama
- Department of Chemistry, Graduate School of Science, Osaka City University, Osaka, Osaka 558-8585, Japan
| | - Yusuke Araki
- Department of Chemistry, Graduate School of Science, Osaka City University, Osaka, Osaka 558-8585, Japan
| | - Mari Yotsu-Yamashita
- Graduate School of Agricultural Science, Tohoku University, Sendai, Miyagi 980-8572, Japan
| | - Yoshiki Morimoto
- Department of Chemistry, Graduate School of Science, Osaka City University, Osaka, Osaka 558-8585, Japan
| |
Collapse
|
20
|
Ryczko D, Hanini‐Daoud M, Condamine S, Bréant BJB, Fougère M, Araya R, Kolta A. S100β‐mediated astroglial control of firing and input processing in layer 5 pyramidal neurons of the mouse visual cortex. J Physiol 2020; 599:677-707. [DOI: 10.1113/jp280501] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2020] [Accepted: 11/23/2020] [Indexed: 12/18/2022] Open
Affiliation(s)
- Dimitri Ryczko
- Département de Neurosciences Université de Montréal Montréal QC Canada
- Département de Pharmacologie‐Physiologie Université de Sherbrooke Sherbrooke QC Canada
- Centre de recherche du CHUS Sherbrooke QC Canada
- Institut de Pharmacologie de Sherbrooke Sherbrooke QC Canada
- Centre d'excellence en neurosciences de l'Université de Sherbrooke Sherbrooke QC Canada
| | | | - Steven Condamine
- Département de Neurosciences Université de Montréal Montréal QC Canada
| | | | - Maxime Fougère
- Département de Pharmacologie‐Physiologie Université de Sherbrooke Sherbrooke QC Canada
| | - Roberto Araya
- Département de Neurosciences Université de Montréal Montréal QC Canada
| | - Arlette Kolta
- Département de Neurosciences Université de Montréal Montréal QC Canada
- Faculté de Médecine Dentaire Université de Montréal Montréal QC Canada
| |
Collapse
|
21
|
Davis H, Herring N, Paterson DJ. Downregulation of M Current Is Coupled to Membrane Excitability in Sympathetic Neurons Before the Onset of Hypertension. Hypertension 2020; 76:1915-1923. [PMID: 33040619 PMCID: PMC8360673 DOI: 10.1161/hypertensionaha.120.15922] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Supplemental Digital Content is available in the text. Neurohumoral activation is an early hallmark of cardiovascular disease and contributes to the etiology of the pathophysiology. Stellectomy has reemerged as a positive therapeutic intervention to modify the progression of dysautonomia, although the biophysical properties underpinning abnormal activity of this ganglia are not fully understood in the initial stages of the disease. We investigated whether stellate ganglia neurons from prehypertensive SHRs (spontaneously hypertensive rats) are hyperactive and describe their electrophysiological phenotype guided by single-cell RNA sequencing, molecular biology, and perforated patch clamp to uncover the mechanism of abnormal excitability. We demonstrate the contribution of a plethora of ion channels, in particular inhibition of M current to stellate ganglia neuronal firing, and confirm the conservation of expression of key ion channel transcripts in human stellate ganglia. We show that hyperexcitability was curbed by M-current activators, nonselective sodium current blockers, or inhibition of Nav1.1-1.3, Nav1.6, or INaP. We conclude that reduced activity of M current contributes significantly to abnormal firing of stellate neurons, which, in part, contributes to the hyperexcitability from rats that have a predisposition to hypertension. Targeting these channels could provide a therapeutic opportunity to minimize the consequences of excessive sympathetic activation.
Collapse
Affiliation(s)
- Harvey Davis
- From the Burdon Sanderson Cardiac Science Centre (H.D., N.H., D.J.P.), University of Oxford, United Kingdom.,Department of Physiology, Anatomy and Genetics, Wellcome Trust OXION Initiative in Ion Channels and Disease (H.D., D.J.P.), University of Oxford, United Kingdom
| | - Neil Herring
- From the Burdon Sanderson Cardiac Science Centre (H.D., N.H., D.J.P.), University of Oxford, United Kingdom.,Oxford Heart Centre, Oxford University Hospitals NHS Foundation Trust, John Radcliffe Hospital, United Kingdom (N.H.)
| | - David J Paterson
- From the Burdon Sanderson Cardiac Science Centre (H.D., N.H., D.J.P.), University of Oxford, United Kingdom.,Department of Physiology, Anatomy and Genetics, Wellcome Trust OXION Initiative in Ion Channels and Disease (H.D., D.J.P.), University of Oxford, United Kingdom
| |
Collapse
|
22
|
Meredith FL, Rennie KJ. Persistent and resurgent Na + currents in vestibular calyx afferents. J Neurophysiol 2020; 124:510-524. [PMID: 32667253 DOI: 10.1152/jn.00124.2020] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Vestibular afferent neurons convey information from hair cells in the peripheral vestibular end organs to central nuclei. Primary vestibular afferent neurons can fire action potentials at high rates and afferent firing patterns vary with the position of nerve terminal endings in vestibular neuroepithelia. Terminals contact hair cells as small bouton or large calyx endings. To investigate the role of Na+ currents (INa) in firing mechanisms, we investigated biophysical properties of INa in calyx-bearing afferents. Whole cell patch-clamp recordings were made from calyx terminals in thin slices of gerbil crista at different postnatal ages: immature [postnatal day (P)5-P8, young (P13-P15), and mature (P30-P45)]. A large transient Na+ current (INaT) was completely blocked by 300 nM tetrodotoxin (TTX) in mature calyces. In addition, INaT was accompanied by much smaller persistent Na+ currents (INaP) and distinctive resurgent Na+ currents (INaR), which were also blocked by TTX. ATX-II, a toxin that slows Na+ channel inactivation, enhanced INaP in immature and mature calyces. 4,9-Anhydro-TTX (4,9-ah-TTX), which selectively blocks Nav1.6 channels, abolished the enhanced INa in mature, but not immature, calyces. Therefore, Nav1.6 channels mediate a component of INaT and INaP in mature calyces, but are minimally expressed at early postnatal days. INaR was expressed in less than one-third of calyces at P6-P8, but expression increased with development, and in mature cristae INaR was frequently found in peripheral calyces. INaR served to increase the availability of Na+ channels following brief membrane depolarizations. In current clamp, the rate and regularity of action potential firing decreased in mature peripheral calyces following 4,9-ah-TTX application. Therefore, Nav1.6 channels are upregulated during development, contribute to INaT, INaP, and INaR, and may regulate excitability by enabling higher mean discharge rates in a subpopulation of mature calyx afferents.NEW & NOTEWORTHY Action potential firing patterns differ between groups of afferent neurons innervating vestibular epithelia. We investigated the biophysical properties of Na+ currents in specialized vestibular calyx afferent terminals during postnatal development. Mature calyces express Na+ currents with transient, persistent, and resurgent components. Nav1.6 channels contribute to resurgent Na+ currents and may enhance firing in peripheral calyx afferents. Understanding Na+ channels that contribute to vestibular nerve responses has implications for developing new treatments for vestibular dysfunction.
Collapse
Affiliation(s)
- Frances L Meredith
- Department of Otolaryngology, University of Colorado School of Medicine, Aurora, Colorado
| | - Katherine J Rennie
- Department of Otolaryngology, University of Colorado School of Medicine, Aurora, Colorado.,Department of Physiology & Biophysics, University of Colorado School of Medicine, Aurora, Colorado
| |
Collapse
|
23
|
Zybura AS, Baucum AJ, Rush AM, Cummins TR, Hudmon A. CaMKII enhances voltage-gated sodium channel Nav1.6 activity and neuronal excitability. J Biol Chem 2020; 295:11845-11865. [PMID: 32611770 DOI: 10.1074/jbc.ra120.014062] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2020] [Revised: 06/30/2020] [Indexed: 11/06/2022] Open
Abstract
Nav1.6 is the primary voltage-gated sodium channel isoform expressed in mature axon initial segments and nodes, making it critical for initiation and propagation of neuronal impulses. Thus, Nav1.6 modulation and dysfunction may have profound effects on input-output properties of neurons in normal and pathological conditions. Phosphorylation is a powerful and reversible mechanism regulating ion channel function. Because Nav1.6 and the multifunctional Ca2+/CaM-dependent protein kinase II (CaMKII) are independently linked to excitability disorders, we sought to investigate modulation of Nav1.6 function by CaMKII signaling. We show that inhibition of CaMKII, a Ser/Thr protein kinase associated with excitability, synaptic plasticity, and excitability disorders, with the CaMKII-specific peptide inhibitor CN21 reduces transient and persistent currents in Nav1.6-expressing Purkinje neurons by 87%. Using whole-cell voltage clamp of Nav1.6, we show that CaMKII inhibition in ND7/23 and HEK293 cells significantly reduces transient and persistent currents by 72% and produces a 5.8-mV depolarizing shift in the voltage dependence of activation. Immobilized peptide arrays and nanoflow LC-electrospray ionization/MS of Nav1.6 reveal potential sites of CaMKII phosphorylation, specifically Ser-561 and Ser-641/Thr-642 within the first intracellular loop of the channel. Using site-directed mutagenesis to test multiple potential sites of phosphorylation, we show that Ala substitutions of Ser-561 and Ser-641/Thr-642 recapitulate the depolarizing shift in activation and reduction in current density. Computational simulations to model effects of CaMKII inhibition on Nav1.6 function demonstrate dramatic reductions in spontaneous and evoked action potentials in a Purkinje cell model, suggesting that CaMKII modulation of Nav1.6 may be a powerful mechanism to regulate neuronal excitability.
Collapse
Affiliation(s)
- Agnes S Zybura
- Program in Medical Neuroscience, Paul and Carole Stark Neurosciences Research Institute, Indiana University School of Medicine, Indianapolis, Indiana, USA
| | - Anthony J Baucum
- Program in Medical Neuroscience, Paul and Carole Stark Neurosciences Research Institute, Indiana University School of Medicine, Indianapolis, Indiana, USA.,Biology Department, Indiana University-Purdue University Indianapolis, School of Science, Indianapolis, Indiana, USA
| | | | - Theodore R Cummins
- Program in Medical Neuroscience, Paul and Carole Stark Neurosciences Research Institute, Indiana University School of Medicine, Indianapolis, Indiana, USA.,Biology Department, Indiana University-Purdue University Indianapolis, School of Science, Indianapolis, Indiana, USA
| | - Andy Hudmon
- Program in Medical Neuroscience, Paul and Carole Stark Neurosciences Research Institute, Indiana University School of Medicine, Indianapolis, Indiana, USA .,Department of Medicinal Chemistry and Molecular Pharmacology, Purdue University, West Lafayette, Indiana, USA
| |
Collapse
|
24
|
Denomme N, Lukowski AL, Hull JM, Jameson MB, Bouza AA, Narayan ARH, Isom LL. The voltage-gated sodium channel inhibitor, 4,9-anhydrotetrodotoxin, blocks human Na v1.1 in addition to Na v1.6. Neurosci Lett 2020; 724:134853. [PMID: 32114117 PMCID: PMC7096269 DOI: 10.1016/j.neulet.2020.134853] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2019] [Revised: 02/12/2020] [Accepted: 02/18/2020] [Indexed: 11/23/2022]
Abstract
Voltage-gated sodium channels (VGSCs) are responsible for the initiation and propagation of action potentials in neurons. The human genome includes ten human VGSC α-subunit genes, SCN(X)A, encoding Nav1.1-1.9 plus Nax. To understand the unique role that each VGSC plays in normal and pathophysiological function in neural networks, compounds with high affinity and selectivity for specific VGSC subtypes are required. Toward that goal, a structural analog of the VGSC pore blocker tetrodotoxin, 4,9-anhydrotetrodotoxin (4,9-ah-TTX), has been reported to be more selective in blocking Na+ current mediated by Nav1.6 than other TTX-sensitive VGSCs, including Nav1.2, Nav1.3, Nav1.4, and Nav1.7. While SCN1A, encoding Nav1.1, has been implicated in several neurological diseases, the effects of 4,9-ah-TTX on Nav1.1-mediated Na+ current have not been tested. Here, we compared the binding of 4,9-ah-TTX for human and mouse brain preparations, and the effects of 4,9-ah-TTX on human Nav1.1-, Nav1.3- and Nav1.6-mediated Na+ currents using the whole-cell patch clamp technique in heterologous cells. We show that, while 4,9-ah-TTX administration results in significant blockade of Nav1.6-mediated Na+ current in the nanomolar range, it also has significant effects on Nav1.1-mediated Na+ current. Thus, 4,9-ah-TTX is not a useful tool in identifying Nav1.6-specific effects in human brain networks.
Collapse
Affiliation(s)
- Nicholas Denomme
- Department of Pharmacology, University of Michigan, Ann Arbor, Michigan, 48109 United States; Center for Consciousness Science, University of Michigan, Ann Arbor, Michigan, 48109 United States
| | - April L Lukowski
- Program in Chemical Biology, University of Michigan, Ann Arbor, Michigan, 48109 United States; Life Sciences Institute, University of Michigan, Ann Arbor, Michigan, 48109 United States
| | - Jacob M Hull
- Neuroscience Graduate Program, University of Michigan, Ann Arbor, Michigan, 48109 United States
| | - Margaret B Jameson
- Department of Pharmacology, University of Michigan, Ann Arbor, Michigan, 48109 United States; Molecular and Cellular Pharmacology Training Program, University of Wisconsin School of Medicine and Public Health, Madison, WI 53705 United States
| | - Alexandra A Bouza
- Department of Pharmacology, University of Michigan, Ann Arbor, Michigan, 48109 United States
| | - Alison R H Narayan
- Program in Chemical Biology, University of Michigan, Ann Arbor, Michigan, 48109 United States; Life Sciences Institute, University of Michigan, Ann Arbor, Michigan, 48109 United States; Department of Chemistry, University of Michigan, Ann Arbor, Michigan, 48109 United States
| | - Lori L Isom
- Department of Pharmacology, University of Michigan, Ann Arbor, Michigan, 48109 United States; Department of Molecular and Integrative Physiology, University of Michigan, Ann Arbor, Michigan, 48109 United States; Neuroscience Graduate Program, University of Michigan, Ann Arbor, Michigan, 48109 United States; Department of Neurology, University of Michigan, Ann Arbor, Michigan, 48109 United States.
| |
Collapse
|
25
|
Affiliation(s)
- Keigo Murakami
- Graduate School of Pharmaceutical SciencesNagoya University Furo-cho, Chikusa-ku Nagoya 464-8601 Japan
| | - Tatsuya Toma
- Graduate School of Pharmaceutical SciencesNagoya University Furo-cho, Chikusa-ku Nagoya 464-8601 Japan
| | - Tohru Fukuyama
- Graduate School of Pharmaceutical SciencesNagoya University Furo-cho, Chikusa-ku Nagoya 464-8601 Japan
| | - Satoshi Yokoshima
- Graduate School of Pharmaceutical SciencesNagoya University Furo-cho, Chikusa-ku Nagoya 464-8601 Japan
| |
Collapse
|
26
|
Weuring WJ, Singh S, Volkers L, Rook MB, van ‘t Slot RH, Bosma M, Inserra M, Vetter I, Verhoeven-Duif NM, Braun KPJ, Rivara M, Koeleman BPC. NaV1.1 and NaV1.6 selective compounds reduce the behavior phenotype and epileptiform activity in a novel zebrafish model for Dravet Syndrome. PLoS One 2020; 15:e0219106. [PMID: 32134913 PMCID: PMC7058281 DOI: 10.1371/journal.pone.0219106] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2019] [Accepted: 02/04/2020] [Indexed: 12/03/2022] Open
Abstract
Dravet syndrome is caused by dominant loss-of-function mutations in SCN1A which cause reduced activity of Nav1.1 leading to lack of neuronal inhibition. On the other hand, gain-of-function mutations in SCN8A can lead to a severe epileptic encephalopathy subtype by over activating NaV1.6 channels. These observations suggest that Nav1.1 and Nav1.6 represent two opposing sides of the neuronal balance between inhibition and activation. Here, we hypothesize that Dravet syndrome may be treated by either enhancing Nav1.1 or reducing Nav1.6 activity. To test this hypothesis we generated and characterized a novel DS zebrafish model and tested new compounds that selectively activate or inhibit the human NaV1.1 or NaV1.6 channel respectively. We used CRISPR/Cas9 to generate two separate Scn1Lab knockout lines as an alternative to previous zebrafish models generated by random mutagenesis or morpholino oligomers. Using an optimized locomotor assay, spontaneous burst movements were detected that were unique to Scn1Lab knockouts and disappear when introducing human SCN1A mRNA. Besides the behavioral phenotype, Scn1Lab knockouts show sudden, electrical discharges in the brain that indicate epileptic seizures in zebrafish. Scn1Lab knockouts showed increased sensitivity to the GABA antagonist pentylenetetrazole and a reduction in whole organism GABA levels. Drug screenings further validated a Dravet syndrome phenotype. We tested the NaV1.1 activator AA43279 and two novel NaV1.6 inhibitors MV1369 and MV1312 in the Scn1Lab knockouts. Both type of compounds significantly reduced the number of spontaneous burst movements and seizure activity. Our results show that selective inhibition of NaV1.6 could be just as efficient as selective activation of NaV1.1 and these approaches could prove to be novel potential treatment strategies for Dravet syndrome and other (genetic) epilepsies. Compounds tested in zebrafish however, should always be further validated in other model systems for efficacy in mammals and to screen for potential side effects.
Collapse
Affiliation(s)
- Wout J. Weuring
- Department of Genetics, Center for Molecular Medicine, UMC Utrecht Brain Center, University Medical Center Utrecht, Utrecht University, Utrecht, The Netherlands
| | - Sakshi Singh
- Department of Genetics, Center for Molecular Medicine, UMC Utrecht Brain Center, University Medical Center Utrecht, Utrecht University, Utrecht, The Netherlands
| | - Linda Volkers
- Department of Cardiology, Laboratory of Experimental Cardiology, University Medical Centre Leiden, Leiden, the Netherlands
| | - Martin B. Rook
- Department of Medical Physiology, University Medical Centre Utrecht, Utrecht, the Netherlands
| | - Ruben H. van ‘t Slot
- Department of Genetics, Center for Molecular Medicine, UMC Utrecht Brain Center, University Medical Center Utrecht, Utrecht University, Utrecht, The Netherlands
| | - Marjolein Bosma
- Department of Genetics, Center for Molecular Medicine, UMC Utrecht Brain Center, University Medical Center Utrecht, Utrecht University, Utrecht, The Netherlands
| | - Marco Inserra
- Centre for Pain Research & School of Pharmacy, University of Queensland, Brisbane, Australia
| | - Irina Vetter
- Centre for Pain Research & School of Pharmacy, University of Queensland, Brisbane, Australia
| | - Nanda M. Verhoeven-Duif
- Department of Genetics, Center for Molecular Medicine, UMC Utrecht Brain Center, University Medical Center Utrecht, Utrecht University, Utrecht, The Netherlands
| | - Kees P. J. Braun
- Department of Neurology, University Medical Centre Utrecht, Utrecht, the Netherlands
| | - Mirko Rivara
- Food and Drug Department, University of Parma, Parma, Italy
| | - Bobby P. C. Koeleman
- Department of Genetics, Center for Molecular Medicine, UMC Utrecht Brain Center, University Medical Center Utrecht, Utrecht University, Utrecht, The Netherlands
| |
Collapse
|
27
|
Murakami K, Toma T, Fukuyama T, Yokoshima S. Total Synthesis of Tetrodotoxin. Angew Chem Int Ed Engl 2020; 59:6253-6257. [DOI: 10.1002/anie.201916611] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2019] [Indexed: 01/03/2023]
Affiliation(s)
- Keigo Murakami
- Graduate School of Pharmaceutical SciencesNagoya University Furo-cho, Chikusa-ku Nagoya 464-8601 Japan
| | - Tatsuya Toma
- Graduate School of Pharmaceutical SciencesNagoya University Furo-cho, Chikusa-ku Nagoya 464-8601 Japan
| | - Tohru Fukuyama
- Graduate School of Pharmaceutical SciencesNagoya University Furo-cho, Chikusa-ku Nagoya 464-8601 Japan
| | - Satoshi Yokoshima
- Graduate School of Pharmaceutical SciencesNagoya University Furo-cho, Chikusa-ku Nagoya 464-8601 Japan
| |
Collapse
|
28
|
Li N, Kalyanasundaram A, Hansen BJ, Artiga EJ, Sharma R, Abudulwahed SH, Helfrich KM, Rozenberg G, Wu PJ, Zakharkin S, Gyorke S, Janssen PM, Whitson BA, Mokadam NA, Biesiadecki BJ, Accornero F, Hummel JD, Mohler PJ, Dobrzynski H, Zhao J, Fedorov VV. Impaired neuronal sodium channels cause intranodal conduction failure and reentrant arrhythmias in human sinoatrial node. Nat Commun 2020; 11:512. [PMID: 31980605 PMCID: PMC6981137 DOI: 10.1038/s41467-019-14039-8] [Citation(s) in RCA: 30] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2019] [Accepted: 12/16/2019] [Indexed: 01/26/2023] Open
Abstract
Mechanisms for human sinoatrial node (SAN) dysfunction are poorly understood and whether human SAN excitability requires voltage-gated sodium channels (Nav) remains controversial. Here, we report that neuronal (n)Nav blockade and selective nNav1.6 blockade during high-resolution optical mapping in explanted human hearts depress intranodal SAN conduction, which worsens during autonomic stimulation and overdrive suppression to conduction failure. Partial cardiac (c)Nav blockade further impairs automaticity and intranodal conduction, leading to beat-to-beat variability and reentry. Multiple nNav transcripts are higher in SAN vs atria; heterogeneous alterations of several isoforms, specifically nNav1.6, are associated with heart failure and chronic alcohol consumption. In silico simulations of Nav distributions suggest that INa is essential for SAN conduction, especially in fibrotic failing hearts. Our results reveal that not only cNav but nNav are also integral for preventing disease-induced failure in human SAN intranodal conduction. Disease-impaired nNav may underlie patient-specific SAN dysfunctions and should be considered to treat arrhythmias. The role of of voltage-gated sodium channels (Nav) in pacemaking and conduction of the human sinoatrial node is unclear. Here, the authors investigate existence and function of neuronal and cardiac Nav in human sinoatrial nodes, and demonstrate their alterations in explanted human diseased hearts.
Collapse
Affiliation(s)
- Ning Li
- Department of Physiology and Cell Biology, The Ohio State University Wexner Medical Center, Columbus, OH, USA.,Bob and Corrine Frick Center for Heart Failure and Arrhythmia, Davis Heart and Lung Research Institute, The Ohio State University Wexner Medical Center, Columbus, OH, USA
| | - Anuradha Kalyanasundaram
- Department of Physiology and Cell Biology, The Ohio State University Wexner Medical Center, Columbus, OH, USA.,Bob and Corrine Frick Center for Heart Failure and Arrhythmia, Davis Heart and Lung Research Institute, The Ohio State University Wexner Medical Center, Columbus, OH, USA
| | - Brian J Hansen
- Department of Physiology and Cell Biology, The Ohio State University Wexner Medical Center, Columbus, OH, USA.,Bob and Corrine Frick Center for Heart Failure and Arrhythmia, Davis Heart and Lung Research Institute, The Ohio State University Wexner Medical Center, Columbus, OH, USA
| | - Esthela J Artiga
- Department of Physiology and Cell Biology, The Ohio State University Wexner Medical Center, Columbus, OH, USA.,Bob and Corrine Frick Center for Heart Failure and Arrhythmia, Davis Heart and Lung Research Institute, The Ohio State University Wexner Medical Center, Columbus, OH, USA
| | - Roshan Sharma
- Auckland Bioengineering Institute, The University of Auckland, Auckland, New Zealand
| | - Suhaib H Abudulwahed
- Department of Physiology and Cell Biology, The Ohio State University Wexner Medical Center, Columbus, OH, USA.,Bob and Corrine Frick Center for Heart Failure and Arrhythmia, Davis Heart and Lung Research Institute, The Ohio State University Wexner Medical Center, Columbus, OH, USA
| | - Katelynn M Helfrich
- Department of Physiology and Cell Biology, The Ohio State University Wexner Medical Center, Columbus, OH, USA.,Bob and Corrine Frick Center for Heart Failure and Arrhythmia, Davis Heart and Lung Research Institute, The Ohio State University Wexner Medical Center, Columbus, OH, USA
| | - Galina Rozenberg
- Department of Physiology and Cell Biology, The Ohio State University Wexner Medical Center, Columbus, OH, USA.,Bob and Corrine Frick Center for Heart Failure and Arrhythmia, Davis Heart and Lung Research Institute, The Ohio State University Wexner Medical Center, Columbus, OH, USA
| | - Pei-Jung Wu
- Department of Physiology and Cell Biology, The Ohio State University Wexner Medical Center, Columbus, OH, USA.,Bob and Corrine Frick Center for Heart Failure and Arrhythmia, Davis Heart and Lung Research Institute, The Ohio State University Wexner Medical Center, Columbus, OH, USA
| | - Stanislav Zakharkin
- Department of Physiology and Cell Biology, The Ohio State University Wexner Medical Center, Columbus, OH, USA
| | - Sandor Gyorke
- Department of Physiology and Cell Biology, The Ohio State University Wexner Medical Center, Columbus, OH, USA.,Bob and Corrine Frick Center for Heart Failure and Arrhythmia, Davis Heart and Lung Research Institute, The Ohio State University Wexner Medical Center, Columbus, OH, USA
| | - Paul Ml Janssen
- Department of Physiology and Cell Biology, The Ohio State University Wexner Medical Center, Columbus, OH, USA.,Bob and Corrine Frick Center for Heart Failure and Arrhythmia, Davis Heart and Lung Research Institute, The Ohio State University Wexner Medical Center, Columbus, OH, USA
| | - Bryan A Whitson
- Bob and Corrine Frick Center for Heart Failure and Arrhythmia, Davis Heart and Lung Research Institute, The Ohio State University Wexner Medical Center, Columbus, OH, USA.,Department of Surgery, The Ohio State University Wexner Medical Center, Columbus, OH, USA
| | - Nahush A Mokadam
- Bob and Corrine Frick Center for Heart Failure and Arrhythmia, Davis Heart and Lung Research Institute, The Ohio State University Wexner Medical Center, Columbus, OH, USA.,Department of Surgery, The Ohio State University Wexner Medical Center, Columbus, OH, USA
| | - Brandon J Biesiadecki
- Department of Physiology and Cell Biology, The Ohio State University Wexner Medical Center, Columbus, OH, USA.,Bob and Corrine Frick Center for Heart Failure and Arrhythmia, Davis Heart and Lung Research Institute, The Ohio State University Wexner Medical Center, Columbus, OH, USA
| | - Federica Accornero
- Department of Physiology and Cell Biology, The Ohio State University Wexner Medical Center, Columbus, OH, USA.,Bob and Corrine Frick Center for Heart Failure and Arrhythmia, Davis Heart and Lung Research Institute, The Ohio State University Wexner Medical Center, Columbus, OH, USA
| | - John D Hummel
- Bob and Corrine Frick Center for Heart Failure and Arrhythmia, Davis Heart and Lung Research Institute, The Ohio State University Wexner Medical Center, Columbus, OH, USA.,Department of Internal Medicine, The Ohio State University Wexner Medical Center, Columbus, OH, USA
| | - Peter J Mohler
- Department of Physiology and Cell Biology, The Ohio State University Wexner Medical Center, Columbus, OH, USA.,Bob and Corrine Frick Center for Heart Failure and Arrhythmia, Davis Heart and Lung Research Institute, The Ohio State University Wexner Medical Center, Columbus, OH, USA
| | - Halina Dobrzynski
- Division of Cardiovascular Sciences, The University of Manchester, Manchester, UK.,Department of Anatomy, Jagiellonian University Medical College, Cracow, Poland
| | - Jichao Zhao
- Auckland Bioengineering Institute, The University of Auckland, Auckland, New Zealand
| | - Vadim V Fedorov
- Department of Physiology and Cell Biology, The Ohio State University Wexner Medical Center, Columbus, OH, USA. .,Bob and Corrine Frick Center for Heart Failure and Arrhythmia, Davis Heart and Lung Research Institute, The Ohio State University Wexner Medical Center, Columbus, OH, USA.
| |
Collapse
|
29
|
Plantier V, Sanchez-Brualla I, Dingu N, Brocard C, Liabeuf S, Gackière F, Brocard F. Calpain fosters the hyperexcitability of motoneurons after spinal cord injury and leads to spasticity. eLife 2019; 8:e51404. [PMID: 31815668 PMCID: PMC6927741 DOI: 10.7554/elife.51404] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2019] [Accepted: 12/08/2019] [Indexed: 12/12/2022] Open
Abstract
Up-regulation of the persistent sodium current (INaP) and down-regulation of the potassium/chloride extruder KCC2 lead to spasticity after spinal cord injury (SCI). We here identified calpain as the driver of the up- and down-regulation of INaP and KCC2, respectively, in neonatal rat lumbar motoneurons. Few days after SCI, neonatal rats developed behavioral signs of spasticity with the emergence of both hyperreflexia and abnormal involuntary muscle contractions on hindlimbs. At the same time, in vitro isolated lumbar spinal cords became hyperreflexive and displayed numerous spontaneous motor outputs. Calpain-I expression paralleled with a proteolysis of voltage-gated sodium (Nav) channels and KCC2. Acute inhibition of calpains reduced this proteolysis, restored the motoneuronal expression of Nav and KCC2, normalized INaP and KCC2 function, and curtailed spasticity. In sum, by up- and down-regulating INaP and KCC2, the calpain-mediated proteolysis of Nav and KCC2 drives the hyperexcitability of motoneurons which leads to spasticity after SCI.
Collapse
Affiliation(s)
- Vanessa Plantier
- Institut de Neurosciences de la Timone (UMR7289), Aix-Marseille Université and CNRSMarseilleFrance
| | - Irene Sanchez-Brualla
- Institut de Neurosciences de la Timone (UMR7289), Aix-Marseille Université and CNRSMarseilleFrance
| | - Nejada Dingu
- Institut de Neurosciences de la Timone (UMR7289), Aix-Marseille Université and CNRSMarseilleFrance
| | - Cécile Brocard
- Institut de Neurosciences de la Timone (UMR7289), Aix-Marseille Université and CNRSMarseilleFrance
| | - Sylvie Liabeuf
- Institut de Neurosciences de la Timone (UMR7289), Aix-Marseille Université and CNRSMarseilleFrance
| | - Florian Gackière
- Institut de Neurosciences de la Timone (UMR7289), Aix-Marseille Université and CNRSMarseilleFrance
| | - Frédéric Brocard
- Institut de Neurosciences de la Timone (UMR7289), Aix-Marseille Université and CNRSMarseilleFrance
| |
Collapse
|
30
|
Alrashdi B, Dawod B, Schampel A, Tacke S, Kuerten S, Marshall JS, Côté PD. Nav1.6 promotes inflammation and neuronal degeneration in a mouse model of multiple sclerosis. J Neuroinflammation 2019; 16:215. [PMID: 31722722 PMCID: PMC6852902 DOI: 10.1186/s12974-019-1622-1] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2019] [Accepted: 10/22/2019] [Indexed: 02/07/2023] Open
Abstract
BACKGROUND In multiple sclerosis (MS) and in the experimental autoimmune encephalomyelitis (EAE) model of MS, the Nav1.6 voltage-gated sodium (Nav) channel isoform has been implicated as a primary contributor to axonal degeneration. Following demyelination Nav1.6, which is normally co-localized with the Na+/Ca2+ exchanger (NCX) at the nodes of Ranvier, associates with β-APP, a marker of neural injury. The persistent influx of sodium through Nav1.6 is believed to reverse the function of NCX, resulting in an increased influx of damaging Ca2+ ions. However, direct evidence for the role of Nav1.6 in axonal degeneration is lacking. METHODS In mice floxed for Scn8a, the gene that encodes the α subunit of Nav1.6, subjected to EAE we examined the effect of eliminating Nav1.6 from retinal ganglion cells (RGC) in one eye using an AAV vector harboring Cre and GFP, while using the contralateral either injected with AAV vector harboring GFP alone or non-targeted eye as control. RESULTS In retinas, the expression of Rbpms, a marker for retinal ganglion cells, was found to be inversely correlated to the expression of Scn8a. Furthermore, the gene expression of the pro-inflammatory cytokines Il6 (IL-6) and Ifng (IFN-γ), and of the reactive gliosis marker Gfap (GFAP) were found to be reduced in targeted retinas. Optic nerves from targeted eyes were shown to have reduced macrophage infiltration and improved axonal health. CONCLUSION Taken together, our results are consistent with Nav1.6 promoting inflammation and contributing to axonal degeneration following demyelination.
Collapse
Affiliation(s)
- Barakat Alrashdi
- Department of Biology, Dalhousie University, Halifax, NS, B3H 4R2, Canada.,Department of Biology, Al-Jouf University, Sakaka, Saudi Arabia
| | - Bassel Dawod
- Department of Pathology, Dalhousie University, Halifax, NS, B3H 4R2, Canada
| | - Andrea Schampel
- Institute of Anatomy and Cell Biology Friedrich Alexander University Erlangen-Nürnberg (FAU), Erlangen, Germany
| | - Sabine Tacke
- Institute of Anatomy and Cell Biology Friedrich Alexander University Erlangen-Nürnberg (FAU), Erlangen, Germany
| | - Stefanie Kuerten
- Institute of Anatomy and Cell Biology Friedrich Alexander University Erlangen-Nürnberg (FAU), Erlangen, Germany
| | - Jean S Marshall
- Department of Pathology, Dalhousie University, Halifax, NS, B3H 4R2, Canada.,Department of Microbiology and Immunology, Dalhousie University, Halifax, NS, B3H 4R2, Canada
| | - Patrice D Côté
- Department of Biology, Dalhousie University, Halifax, NS, B3H 4R2, Canada. .,Department of Ophthalmology and Visual Sciences, Dalhousie University, Halifax, NS, B3H 4R2, Canada.
| |
Collapse
|
31
|
Yin N, Yang YL, Cheng S, Wang HN, Hu X, Miao Y, Li F, Wang Z. Dopamine D2 Receptor-Mediated Modulation of Rat Retinal Ganglion Cell Excitability. Neurosci Bull 2019; 36:230-242. [PMID: 31606861 DOI: 10.1007/s12264-019-00431-3] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2019] [Accepted: 06/09/2019] [Indexed: 12/19/2022] Open
Abstract
Ganglion cells (RGCs) are the sole output neurons of the retinal circuity. Here, we investigated whether and how dopamine D2 receptors modulate the excitability of dissociated rat RGCs. Application of the selective D2 receptor agonist quinpirole inhibited outward K+ currents, which were mainly mediated by glybenclamide- and 4-aminopyridine-sensitive channels, but not the tetraethylammonium-sensitive channel. In addition, quinpirole selectively enhanced Nav1.6 voltage-gated Na+ currents. The intracellular cAMP/protein kinase A, Ca2+/calmodulin-dependent protein kinase II, and mitogen-activated protein kinase/extracellular signal-regulated kinase signaling pathways were responsible for the effects of quinpirole on K+ and Na+ currents, while phospholipase C/protein kinase C signaling was not involved. Under current-clamp conditions, the number of action potentials evoked by positive current injection was increased by quinpirole. Our results suggest that D2 receptor activation increases RGC excitability by suppressing outward K+ currents and enhancing Nav1.6 currents, which may affect retinal visual information processing.
Collapse
Affiliation(s)
- Ning Yin
- Department of Neurology, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institutes of Brain Science, Zhongshan Hospital, Fudan University, Shanghai, 200032, China
| | - Yu-Long Yang
- Department of Neurology, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institutes of Brain Science, Zhongshan Hospital, Fudan University, Shanghai, 200032, China
| | - Shuo Cheng
- Department of Neurology, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institutes of Brain Science, Zhongshan Hospital, Fudan University, Shanghai, 200032, China
| | - Hong-Ning Wang
- Department of Neurology, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institutes of Brain Science, Zhongshan Hospital, Fudan University, Shanghai, 200032, China
| | - Xin Hu
- Department of Neurology, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institutes of Brain Science, Zhongshan Hospital, Fudan University, Shanghai, 200032, China
| | - Yanying Miao
- Department of Neurology, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institutes of Brain Science, Zhongshan Hospital, Fudan University, Shanghai, 200032, China
| | - Fang Li
- Department of Neurology, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institutes of Brain Science, Zhongshan Hospital, Fudan University, Shanghai, 200032, China
| | - Zhongfeng Wang
- Department of Neurology, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institutes of Brain Science, Zhongshan Hospital, Fudan University, Shanghai, 200032, China.
| |
Collapse
|
32
|
White HV, Brown ST, Bozza TC, Raman IM. Effects of FGF14 and Na Vβ4 deletion on transient and resurgent Na current in cerebellar Purkinje neurons. J Gen Physiol 2019; 151:1300-1318. [PMID: 31558566 PMCID: PMC6829560 DOI: 10.1085/jgp.201912390] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2019] [Accepted: 09/03/2019] [Indexed: 11/20/2022] Open
Abstract
Voltage-gated Na channels of Purkinje cells are specialized to maintain high availability during high-frequency repetitive firing. They enter fast-inactivated states relatively slowly and undergo a voltage-dependent open-channel block by an intracellular protein (or proteins) that prevents stable fast inactivation and generates resurgent Na current. These properties depend on the pore-forming α subunits, as well as modulatory subunits within the Na channel complex. The identity of the factors responsible for open-channel block remains a question. Here we investigate the effects of genetic mutation of two Na channel auxiliary subunits highly expressed in Purkinje cells, NaVβ4 and FGF14, on modulating Na channel blocked as well as inactivated states. We find that although both NaVβ4 and the FGF14 splice variant FGF14-1a contain sequences that can generate resurgent-like currents when applied to Na channels in peptide form, deletion of either protein, or both proteins simultaneously, does not eliminate resurgent current in acutely dissociated Purkinje cell bodies. Loss of FGF14 expression does, however, reduce resurgent current amplitude and leads to an acceleration and stabilization of inactivation that is not reversed by application of the site-3 toxin, anemone toxin II (ATX). Tetrodotoxin (TTX) sensitivity is higher for resurgent than transient components of Na current, and loss of FGF14 preferentially affects a highly TTX-sensitive subset of Purkinje α subunits. The data suggest that NaV1.6 channels, which are known to generate the majority of Purkinje cell resurgent current, bind TTX with high affinity and are modulated by FGF14 to facilitate open-channel block.
Collapse
Affiliation(s)
- Hayley V White
- Department of Neurobiology, Northwestern University, Evanston, IL.,Northwestern University Interdepartmental Neuroscience Program, Northwestern University, Evanston, IL
| | - Spencer T Brown
- Department of Neurobiology, Northwestern University, Evanston, IL.,Northwestern University Interdepartmental Neuroscience Program, Northwestern University, Evanston, IL
| | - Thomas C Bozza
- Department of Neurobiology, Northwestern University, Evanston, IL.,Northwestern University Interdepartmental Neuroscience Program, Northwestern University, Evanston, IL
| | - Indira M Raman
- Department of Neurobiology, Northwestern University, Evanston, IL .,Northwestern University Interdepartmental Neuroscience Program, Northwestern University, Evanston, IL
| |
Collapse
|
33
|
Vysokov N, McMahon SB, Raouf R. The role of Na V channels in synaptic transmission after axotomy in a microfluidic culture platform. Sci Rep 2019; 9:12915. [PMID: 31501450 PMCID: PMC6733904 DOI: 10.1038/s41598-019-49214-w] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2018] [Accepted: 08/20/2019] [Indexed: 01/07/2023] Open
Abstract
Voltage gated sodium channels are key players in aberrant pain signaling and sensitization of nociceptors after peripheral nerve injury. The extent to which sodium channel activity after injury contributes to synaptic transmission at the first pain synapse however remains unclear. To investigate the effect of axotomy on synaptic transmission between dorsal root ganglia neurons and dorsal horn neurons, we reconstructed the first pain synapse in a novel microfluidic based compartmentalized cell culture system, which recapitulates the connectivity of peripheral pain signaling. We show that following axotomy of the distal axons, inhibition of NaV1.7 and NaV1.8 sodium channels in incoming presynaptic DRG axons is no longer sufficient to block activation of these axons and the resulting synaptic transmission to dorsal horn neurons. We found that blockade of NaV1.6 activity is highly effective in reducing activation of incoming axons contributing to synaptic transmission after axotomy of DRG neurons. The microfluidic culture system described here offers an in vitro platform to recapitulate and study the first pain synapse.
Collapse
Affiliation(s)
- Nickolai Vysokov
- Wolfson Centre for Age-Related Diseases, Institute of Psychiatry, Psychology & Neuroscience, King's College London, London, SE1 1UL, United Kingdom
| | - Stephen B McMahon
- Wolfson Centre for Age-Related Diseases, Institute of Psychiatry, Psychology & Neuroscience, King's College London, London, SE1 1UL, United Kingdom
| | - Ramin Raouf
- Wolfson Centre for Age-Related Diseases, Institute of Psychiatry, Psychology & Neuroscience, King's College London, London, SE1 1UL, United Kingdom.
| |
Collapse
|
34
|
Tetrodotoxin-Sensitive Sodium Channels Mediate Action Potential Firing and Excitability in Menthol-Sensitive Vglut3-Lineage Sensory Neurons. J Neurosci 2019; 39:7086-7101. [PMID: 31300524 DOI: 10.1523/jneurosci.2817-18.2019] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2018] [Revised: 06/04/2019] [Accepted: 07/03/2019] [Indexed: 12/16/2022] Open
Abstract
Small-diameter vesicular glutamate transporter 3-lineage (Vglut3lineage) dorsal root ganglion (DRG) neurons play an important role in mechanosensation and thermal hypersensitivity; however, little is known about their intrinsic electrical properties. We therefore set out to investigate mechanisms of excitability within this population. Calcium microfluorimetry analysis of male and female mouse DRG neurons demonstrated that the cooling compound menthol selectively activates a subset of Vglut3lineage neurons. Whole-cell recordings showed that small-diameter Vglut3lineage DRG neurons fire menthol-evoked action potentials and exhibited robust, transient receptor potential melastatin 8 (TRPM8)-dependent discharges at room temperature. This heightened excitability was confirmed by current-clamp and action potential phase-plot analyses, which showed menthol-sensitive Vglut3lineage neurons to have more depolarized membrane potentials, lower firing thresholds, and higher evoked firing frequencies compared with menthol-insensitive Vglut3lineage neurons. A biophysical analysis revealed voltage-gated sodium channel (NaV) currents in menthol-sensitive Vglut3lineage neurons were resistant to entry into slow inactivation compared with menthol-insensitive neurons. Multiplex in situ hybridization showed similar distributions of tetrodotoxin (TTX)-sensitive NaV transcripts between TRPM8-positive and -negative Vglut3lineage neurons; however, NaV1.8 transcripts, which encode TTX-resistant channels, were more prevalent in TRPM8-negative neurons. Conversely, pharmacological analyses identified distinct functional contributions of NaV subunits, with NaV1.1 driving firing in menthol-sensitive neurons, whereas other small-diameter Vglut3lineage neurons rely primarily on TTX-resistant NaV channels. Additionally, when NaV1.1 channels were blocked, the remaining NaV current readily entered into slow inactivation in menthol-sensitive Vglut3lineage neurons. Thus, these data demonstrate that TTX-sensitive NaVs drive action potential firing in menthol-sensitive sensory neurons and contribute to their heightened excitability.SIGNIFICANCE STATEMENT Somatosensory neurons encode various sensory modalities including thermoreception, mechanoreception, nociception, and itch. This report identifies a previously unknown requirement for tetrodotoxin-sensitive sodium channels in action potential firing in a discrete subpopulation of small-diameter sensory neurons that are activated by the cooling agent menthol. Together, our results provide a mechanistic understanding of factors that control intrinsic excitability in functionally distinct subsets of peripheral neurons. Furthermore, as menthol has been used for centuries as an analgesic and anti-pruritic, these findings support the viability of NaV1.1 as a therapeutic target for sensory disorders.
Collapse
|
35
|
Saba L, Viscomi MT, Martini A, Caioli S, Mercuri NB, Guatteo E, Zona C. Modified age-dependent expression of NaV1.6 in an ALS model correlates with motor cortex excitability alterations. Neurobiol Dis 2019; 130:104532. [PMID: 31302244 DOI: 10.1016/j.nbd.2019.104532] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2019] [Revised: 06/28/2019] [Accepted: 07/10/2019] [Indexed: 12/13/2022] Open
Abstract
Cortical hyperexcitability is an early and intrinsic feature of Amyotrophic Lateral Sclerosis (ALS), but the mechanisms underlying this critical neuronal dysfunction are poorly understood. Recently, we have demonstrated that layer V pyramidal neurons (PNs) in the primary motor cortex (M1) of one-month old (P30) G93A ALS mice display an early hyperexcitability status compared to Control mice. In order to investigate the time-dependent evolution of the cortical excitability in the G93A ALS model, here we have performed an electrophysiological and immunohistochemical study at three different mouse ages. M1 PNs from 14-days old (P14) G93A mice have shown no excitability alterations, while M1 PNs from 3-months old (P90) G93A mice have shown a hypoexcitability status, compared to Control mice. These age-dependent cortical excitability dysfunctions correlate with a similar time-dependent trend of the persistent sodium current (INaP) amplitude alterations, suggesting that INaP may play a crucial role in the G93A cortical excitability aberrations. Specifically, immunohistochemistry experiments have indicated that the expression level of the NaV1.6 channel, one of the voltage-gated Na+ channels mainly distributed within the central nervous system, varies in G93A primary motor cortex during disease progression, according to the excitability and INaP alterations, but not in other cortical areas. Microfluorometry experiments, combined with electrophysiological recordings, have verified that P30 G93A PNs hyperexcitability is associated to a greater accumulation of intracellular calcium ([Ca2+]i) compared to Control PNs, and that this difference is still present when G93A and Control PNs fire action potentials at the same frequency. These results suggest that [Ca2+]i de-regulation in G93A PNs may contribute to neuronal demise and that the NaV1.6 channels could be a potential therapeutic target to ameliorate ALS disease progression.
Collapse
Affiliation(s)
- Luana Saba
- Department of Systems Medicine, University of Rome "Tor Vergata" via Montpellier 1, Rome 00133, Italy
| | - Maria Teresa Viscomi
- Università Cattolica del Sacro Cuore, Istituto di Istologia ed Embriologia, Fondazione Policlinico Universitario A. Gemelli, Largo F. Vito 1, Rome 00168, Italy
| | - Alessandro Martini
- IRCCS Fondazione Santa Lucia, via del Fosso di Fiorano 64, Rome 00143, Italy
| | - Silvia Caioli
- IRCCS Fondazione Santa Lucia, via del Fosso di Fiorano 64, Rome 00143, Italy
| | - Nicola Biagio Mercuri
- Department of Systems Medicine, University of Rome "Tor Vergata" via Montpellier 1, Rome 00133, Italy; IRCCS Fondazione Santa Lucia, via del Fosso di Fiorano 64, Rome 00143, Italy
| | - Ezia Guatteo
- IRCCS Fondazione Santa Lucia, via del Fosso di Fiorano 64, Rome 00143, Italy; Department of Motor Science and Wellness, University of Naples 'Parthenope', Via Medina 40, Naples 80133, Italy
| | - Cristina Zona
- Department of Systems Medicine, University of Rome "Tor Vergata" via Montpellier 1, Rome 00133, Italy; IRCCS Fondazione Santa Lucia, via del Fosso di Fiorano 64, Rome 00143, Italy.
| |
Collapse
|
36
|
Israel MR, Tanaka BS, Castro J, Thongyoo P, Robinson SD, Zhao P, Deuis JR, Craik DJ, Durek T, Brierley SM, Waxman SG, Dib-Hajj SD, Vetter I. Na V 1.6 regulates excitability of mechanosensitive sensory neurons. J Physiol 2019; 597:3751-3768. [PMID: 31087362 DOI: 10.1113/jp278148] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2019] [Accepted: 05/08/2019] [Indexed: 12/20/2022] Open
Abstract
KEY POINTS Voltage-gated sodium channels are critical for peripheral sensory neuron transduction and have been implicated in a number of painful and painless disorders. The β-scorpion toxin, Cn2, is selective for NaV 1.6 in dorsal root ganglion neurons. NaV 1.6 plays an essential role in peripheral sensory neurons, specifically at the distal terminals of mechanosensing fibres innervating the skin and colon. NaV 1.6 activation also leads to enhanced response to mechanical stimulus in vivo. This works highlights the use of toxins in elucidating pain pathways moreover the importance of non-peripherally restricted NaV isoforms in pain generation. ABSTRACT Peripheral sensory neurons express multiple voltage-gated sodium channels (NaV ) critical for the initiation and propagation of action potentials and transmission of sensory input. Three pore-forming sodium channel isoforms are primarily expressed in the peripheral nervous system (PNS): NaV 1.7, NaV 1.8 and NaV 1.9. These sodium channels have been implicated in painful and painless channelopathies and there has been intense interest in them as potential therapeutic targets in human pain. Emerging evidence suggests NaV 1.6 channels are an important isoform in pain sensing. This study aimed to assess, using pharmacological approaches, the function of NaV 1.6 channels in peripheral sensory neurons. The potent and NaV 1.6 selective β-scorpion toxin Cn2 was used to assess the effect of NaV 1.6 channel activation in the PNS. The multidisciplinary approach included Ca2+ imaging, whole-cell patch-clamp recordings, skin-nerve and gut-nerve preparations and in vivo behavioural assessment of pain. Cn2 facilitates NaV 1.6 early channel opening, and increased persistent and resurgent currents in large-diameter dorsal root ganglion (DRG) neurons. This promotes enhanced excitatory drive and tonic action potential firing in these neurons. In addition, NaV 1.6 channel activation in the skin and gut leads to increased response to mechanical stimuli. Finally, intra-plantar injection of Cn2 causes mechanical but not thermal allodynia. This study confirms selectivity of Cn2 on NaV 1.6 channels in sensory neurons. Activation of NaV 1.6 channels, in terminals of the skin and viscera, leads to profound changes in neuronal responses to mechanical stimuli. In conclusion, sensory neurons expressing NaV 1.6 are important for the transduction of mechanical information in sensory afferents innervating the skin and viscera.
Collapse
Affiliation(s)
- Mathilde R Israel
- IMB Centre for Pain Research, Institute for Molecular Bioscience, 306 Carmody Rd (Building 80), University of Queensland, Brisbane, Queensland, 4072, Australia
| | - Brian S Tanaka
- Center for Neuroscience and Regeneration Research, Yale School of Medicine, New Haven, CT, 06510, USA.,Department of Neurology, Yale School of Medicine, New Haven, CT, 06510, USA.,Rehabilitation Research Center, VA Connecticut Healthcare System, West Haven, CT, 06516, USA
| | - Joel Castro
- Visceral Pain Research Group, Centre for Neuroscience, College of Medicine and Public Health, Flinders University, Bedford Park, South Australia, 5042, Australia.,Hopwood Centre for Neurobiology and Centre for Nutrition and Gastrointestinal Diseases, Discipline of Medicine, University of Adelaide, South Australian Health and Medical Research Institute (SAHMRI), North Terrace, Adelaide, South Australia, 5000, Australia
| | - Panumart Thongyoo
- IMB Centre for Pain Research, Institute for Molecular Bioscience, 306 Carmody Rd (Building 80), University of Queensland, Brisbane, Queensland, 4072, Australia
| | - Samuel D Robinson
- IMB Centre for Pain Research, Institute for Molecular Bioscience, 306 Carmody Rd (Building 80), University of Queensland, Brisbane, Queensland, 4072, Australia
| | - Peng Zhao
- Center for Neuroscience and Regeneration Research, Yale School of Medicine, New Haven, CT, 06510, USA.,Department of Neurology, Yale School of Medicine, New Haven, CT, 06510, USA.,Rehabilitation Research Center, VA Connecticut Healthcare System, West Haven, CT, 06516, USA
| | - Jennifer R Deuis
- IMB Centre for Pain Research, Institute for Molecular Bioscience, 306 Carmody Rd (Building 80), University of Queensland, Brisbane, Queensland, 4072, Australia
| | - David J Craik
- IMB Centre for Pain Research, Institute for Molecular Bioscience, 306 Carmody Rd (Building 80), University of Queensland, Brisbane, Queensland, 4072, Australia
| | - Thomas Durek
- IMB Centre for Pain Research, Institute for Molecular Bioscience, 306 Carmody Rd (Building 80), University of Queensland, Brisbane, Queensland, 4072, Australia
| | - Stuart M Brierley
- Visceral Pain Research Group, Centre for Neuroscience, College of Medicine and Public Health, Flinders University, Bedford Park, South Australia, 5042, Australia.,Hopwood Centre for Neurobiology and Centre for Nutrition and Gastrointestinal Diseases, Discipline of Medicine, University of Adelaide, South Australian Health and Medical Research Institute (SAHMRI), North Terrace, Adelaide, South Australia, 5000, Australia
| | - Stephen G Waxman
- Center for Neuroscience and Regeneration Research, Yale School of Medicine, New Haven, CT, 06510, USA.,Department of Neurology, Yale School of Medicine, New Haven, CT, 06510, USA.,Rehabilitation Research Center, VA Connecticut Healthcare System, West Haven, CT, 06516, USA
| | - Sulayman D Dib-Hajj
- Center for Neuroscience and Regeneration Research, Yale School of Medicine, New Haven, CT, 06510, USA.,Department of Neurology, Yale School of Medicine, New Haven, CT, 06510, USA.,Rehabilitation Research Center, VA Connecticut Healthcare System, West Haven, CT, 06516, USA
| | - Irina Vetter
- IMB Centre for Pain Research, Institute for Molecular Bioscience, 306 Carmody Rd (Building 80), University of Queensland, Brisbane, Queensland, 4072, Australia.,School of Pharmacy, Pharmacy Australia Centre of Excellence, 20 Cornwall St, Woolloongabba, Queensland, 4102, Australia
| |
Collapse
|
37
|
Meredith FL, Rennie KJ. Regional and Developmental Differences in Na + Currents in Vestibular Primary Afferent Neurons. Front Cell Neurosci 2018; 12:423. [PMID: 30487736 PMCID: PMC6246661 DOI: 10.3389/fncel.2018.00423] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2018] [Accepted: 10/29/2018] [Indexed: 02/04/2023] Open
Abstract
The vestibular system relays information about head position via afferent nerve fibers to the brain in the form of action potentials. Voltage-gated Na+ channels in vestibular afferents drive the initiation and propagation of action potentials, but their expression during postnatal development and their contributions to firing in diverse mature afferent populations are unknown. Electrophysiological techniques were used to determine Na+ channel subunit types in vestibular calyx-bearing afferents at different stages of postnatal development. We used whole cell patch clamp recordings in thin slices of gerbil crista neuroepithelium to investigate Na+ channels and firing patterns in central zone (CZ) and peripheral zone (PZ) afferents. PZ afferents are exclusively dimorphic, innervating type I and type II hair cells, whereas CZ afferents can form dimorphs or calyx-only terminals which innervate type I hair cells alone. All afferents expressed tetrodotoxin (TTX)-sensitive Na+ currents, but TTX-sensitivity varied with age. During the fourth postnatal week, 200–300 nM TTX completely blocked sodium currents in PZ and CZ calyces. By contrast, in immature calyces [postnatal day (P) 5–11], a small component of peak sodium current remained in 200 nM TTX. Application of 1 μM TTX, or Jingzhaotoxin-III plus 200 nM TTX, abolished sodium current in immature calyces, suggesting the transient expression of voltage-gated sodium channel 1.5 (Nav1.5) during development. A similar TTX-insensitive current was found in early postnatal crista hair cells (P5–9) and constituted approximately one third of the total sodium current. The Nav1.6 channel blocker, 4,9-anhydrotetrodotoxin, reduced a component of sodium current in immature and mature calyces. At 100 nM 4,9-anhydrotetrodotoxin, peak sodium current was reduced on average by 20% in P5–14 calyces, by 37% in mature dimorphic PZ calyces, but by less than 15% in mature CZ calyx-only terminals. In mature PZ calyces, action potentials became shorter and broader in the presence of 4,9-anhydrotetrodotoxin implicating a role for Nav1.6 channels in firing in dimorphic afferents.
Collapse
Affiliation(s)
- Frances L Meredith
- Department of Otolaryngology, University of Colorado School of Medicine, Aurora, CO, United States
| | - Katherine J Rennie
- Department of Otolaryngology, University of Colorado School of Medicine, Aurora, CO, United States.,Department of Physiology & Biophysics, University of Colorado School of Medicine, Aurora, CO, United States
| |
Collapse
|
38
|
Cardoso FC, Lewis RJ. Sodium channels and pain: from toxins to therapies. Br J Pharmacol 2018; 175:2138-2157. [PMID: 28749537 PMCID: PMC5980290 DOI: 10.1111/bph.13962] [Citation(s) in RCA: 64] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2017] [Revised: 07/11/2017] [Accepted: 07/17/2017] [Indexed: 12/16/2022] Open
Abstract
Voltage-gated sodium channels (NaV channels) are essential for the initiation and propagation of action potentials that critically influence our ability to respond to a diverse range of stimuli. Physiological and pharmacological studies have linked abnormal function of NaV channels to many human disorders, including chronic neuropathic pain. These findings, along with the description of the functional properties and expression pattern of NaV channel subtypes, are helping to uncover subtype specific roles in acute and chronic pain and revealing potential opportunities to target these with selective inhibitors. High-throughput screens and automated electrophysiology platforms have identified natural toxins as a promising group of molecules for the development of target-specific analgesics. In this review, the role of toxins in defining the contribution of NaV channels in acute and chronic pain states and their potential to be used as analgesic therapies are discussed. LINKED ARTICLES This article is part of a themed section on Recent Advances in Targeting Ion Channels to Treat Chronic Pain. To view the other articles in this section visit http://onlinelibrary.wiley.com/doi/10.1111/bph.v175.12/issuetoc.
Collapse
Affiliation(s)
- Fernanda C Cardoso
- Department of Chemistry and Structural Biology, Institute for Molecular BioscienceThe University of QueenslandBrisbaneQLDAustralia
| | - Richard J Lewis
- Department of Chemistry and Structural Biology, Institute for Molecular BioscienceThe University of QueenslandBrisbaneQLDAustralia
| |
Collapse
|
39
|
Koleske M, Bonilla I, Thomas J, Zaman N, Baine S, Knollmann BC, Veeraraghavan R, Györke S, Radwański PB. Tetrodotoxin-sensitive Na vs contribute to early and delayed afterdepolarizations in long QT arrhythmia models. J Gen Physiol 2018; 150:991-1002. [PMID: 29793933 PMCID: PMC6028491 DOI: 10.1085/jgp.201711909] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2017] [Revised: 03/30/2018] [Accepted: 04/30/2018] [Indexed: 01/08/2023] Open
Abstract
Neuronal Na+ channels contribute to catecholaminergic polymorphic ventricular tachycardia in the heart, but their role in other types of arrhythmias is unknown. Koleske et al. show that they contribute to early and delayed afterdepolarizations common to long QT, catecholaminergic polymorphic ventricular tachycardia, and overlap phenotypes. Recent evidence suggests that neuronal Na+ channels (nNavs) contribute to catecholamine-promoted delayed afterdepolarizations (DADs) and catecholaminergic polymorphic ventricular tachycardia (CPVT). The newly identified overlap between CPVT and long QT (LQT) phenotypes has stoked interest in the cross-talk between aberrant Na+ and Ca2+ handling and its contribution to early afterdepolarizations (EADs) and DADs. Here, we used Ca2+ imaging and electrophysiology to investigate the role of Na+ and Ca2+ handling in DADs and EADs in wild-type and cardiac calsequestrin (CASQ2)-null mice. In experiments, repolarization was impaired using 4-aminopyridine (4AP), whereas the L-type Ca2+ and late Na+ currents were augmented using Bay K 8644 (BayK) and anemone toxin II (ATX-II), respectively. The combination of 4AP and isoproterenol prolonged action potential duration (APD) and promoted aberrant Ca2+ release, EADs, and DADs in wild-type cardiomyocytes. Similarly, BayK in the absence of isoproterenol induced the same effects in CASQ2-null cardiomyocytes. In vivo, it prolonged the QT interval and, upon catecholamine challenge, precipitated wide QRS polymorphic ventricular tachycardia that resembled human torsades de pointes. Treatment with ATX-II produced similar effects at both the cellular level and in vivo. Importantly, nNav inhibition with riluzole or 4,9-anhydro-tetrodotoxin reduced the incidence of ATX-II–, BayK-, or 4AP-induced EADs, DADs, aberrant Ca2+ release, and VT despite only modestly mitigating APD prolongation. These data reveal the contribution of nNaVs to triggered arrhythmias in murine models of LQT and CPVT-LQT overlap phenotypes. We also demonstrate the antiarrhythmic impact of nNaV inhibition, independent of action potential and QT interval duration, and provide a basis for a mechanistically driven antiarrhythmic strategy.
Collapse
Affiliation(s)
- Megan Koleske
- Dorothy M. Davis Heart and Lung Research Institute, College of Medicine, The Ohio State University Wexner Medical Center, Columbus, OH
| | - Ingrid Bonilla
- Dorothy M. Davis Heart and Lung Research Institute, College of Medicine, The Ohio State University Wexner Medical Center, Columbus, OH.,Department of Physiology and Cell Biology, College of Medicine, The Ohio State University, Columbus, OH
| | - Justin Thomas
- Division of Pharmacy Practice and Sciences, College of Pharmacy, The Ohio State University, Columbus, OH
| | - Naveed Zaman
- Division of Pharmacology, College of Pharmacy, The Ohio State University, Columbus, OH
| | - Stephen Baine
- Division of Pharmacology, College of Pharmacy, The Ohio State University, Columbus, OH
| | - Bjorn C Knollmann
- Division of Clinical Pharmacology, Vanderbilt University Medical School, Nashville, TN
| | - Rengasayee Veeraraghavan
- Dorothy M. Davis Heart and Lung Research Institute, College of Medicine, The Ohio State University Wexner Medical Center, Columbus, OH.,Department of Biomedical Engineering, College of Engineering, The Ohio State University, Columbus, OH
| | - Sándor Györke
- Dorothy M. Davis Heart and Lung Research Institute, College of Medicine, The Ohio State University Wexner Medical Center, Columbus, OH.,Department of Physiology and Cell Biology, College of Medicine, The Ohio State University, Columbus, OH
| | - Przemysław B Radwański
- Dorothy M. Davis Heart and Lung Research Institute, College of Medicine, The Ohio State University Wexner Medical Center, Columbus, OH .,Division of Pharmacy Practice and Sciences, College of Pharmacy, The Ohio State University, Columbus, OH.,Division of Pharmacology, College of Pharmacy, The Ohio State University, Columbus, OH
| |
Collapse
|
40
|
Xu L, Li D, Ding J, Pan L, Ding X. Insight into tetrodotoxin blockade and resistance mechanisms of Na v 1.2 sodium channel by theoretical approaches. Chem Biol Drug Des 2018; 92:1445-1457. [PMID: 29673065 DOI: 10.1111/cbdd.13310] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2018] [Revised: 03/08/2018] [Accepted: 03/17/2018] [Indexed: 11/30/2022]
Abstract
Nav 1.2, a member of voltage-gated sodium channels (Nav s) that are responsible for the generation and propagation of action potentials along the cell membrane, and play a vital role in the process of information transmission within the nervous system and muscle contraction, is preferentially expressed in the central nervous system. As a potent and selective blocker of Nav s, tetrodotoxin (TTX) has been extensively studied in biological and chemical sciences, whereas the detailed mechanism by which it blocks nine Nav 1 channel subtypes remain elusive. Despite the high structural similarity, the TTX metabolite 4,9-anhydro-TTX is 161 times less effective toward the mammalian Nav 1.2, which puzzled us to ask a question why such a subtle structural variation results in the largely binding affinity difference. In the current work, an integrated computational strategy, including homology modeling, induced fit docking, explicit-solvent MD simulations, and free energy calculations, was employed to investigate the binding mechanism and conformational determinants of TTX analogs. Based on the computational results, the H-bond interactions between C4-OH and C9-OH of TTX and the outer ring carboxylates of the selectivity-filter residues, and the cation-π interaction between the primary amine of guanidinium of TTX and Phe385 determine the difference of their binding affinities. Moreover, the computationally simulations were carried out for the D384N and E945K mutants of hNav 1.2-TTX, and the rank of the predicted binding free energies is in accordance with the experimental data. These observations provide a valuable model to design potent and selective neurotoxins of Nav 1.2 and shed light on the blocking mechanism of TTX to sodium channels.
Collapse
Affiliation(s)
- Lei Xu
- Institute of Bioinformatics and Medical Engineering, School of Electrical and Information Engineering, Jiangsu University of Technology, Changzhou, China
| | - Dayu Li
- Beijing Institute of Pharmaceutical Chemistry, Beijing, China
| | - Junjie Ding
- Beijing Institute of Pharmaceutical Chemistry, Beijing, China
| | - Li Pan
- Beijing Institute of Pharmaceutical Chemistry, Beijing, China
| | - Xiaoqin Ding
- Beijing Institute of Pharmaceutical Chemistry, Beijing, China
| |
Collapse
|
41
|
Takahara K, Yamamoto T, Uchida K, Zhu HL, Shibata A, Inai T, Noguchi M, Yotsu-Yamashita M, Teramoto N. Effects of 4,9-anhydrotetrodotoxin on voltage-gated Na + channels of mouse vas deferens myocytes and recombinant Na V1.6 channels. Naunyn Schmiedebergs Arch Pharmacol 2018; 391:489-499. [PMID: 29453527 DOI: 10.1007/s00210-018-1476-6] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2017] [Accepted: 01/31/2018] [Indexed: 12/19/2022]
Abstract
Molecular investigations were performed in order to determine the major characteristics of voltage-gated Na+ channel β-subunits in mouse vas deferens. The use of real-time quantitative PCR showed that the expression of Scn1b was significantly higher than that of other β-subunit genes (Scn2b - Scn4b). Immunoreactivity of Scn1b proteins was also detected in the inner circular and outer longitudinal smooth muscle of mouse vas deferens. In whole-cell recordings, the actions of 4,9-anhydroTTX on voltage-gated Na+ current peak amplitude in myocytes (i.e., native INa) were compared with its inhibitory potency on recombinant NaV1.6 channels (expressed in HEK293 cells). A depolarizing rectangular voltage-pulse elicited a fast and transient inward native INa and recombinant NaV1.6 expressed in HEK293 cells (i.e., recombinant INa). The current decay of native INa was similar to the recombinant NaV1.6 current co-expressed with β1-subunits. The current-voltage (I-V) relationships of native INa were similar to those of recombinant NaV1.6 currents co-expressed with β1-subunits. Application of 4,9-anhydroTTX inhibited the peak amplitude of native INa (K i = 510 nM), recombinant INa (K i = 112 nM), and recombinant INa co-expressed with β1-subunits (K i = 92 nM). The half-maximal (Vhalf) activation and inactivation of native INa values were similar to those observed in recombinant INa co-expressed with β1-subunits. These results suggest that β1-subunit proteins are likely to be expressed mainly in the smooth muscle layers of murine vas deferens and that 4,9-anhydroTTX inhibited not only native INa but also recombinant INa and recombinant INa co-expressed with β1-subunits in a concentration-dependent manner.
Collapse
Affiliation(s)
- Kohei Takahara
- Department of Pharmacology, Faculty of Medicine, Saga University, Saga, 849-8501, Japan
| | - Tadashi Yamamoto
- Department of Pharmacology, Faculty of Medicine, Saga University, Saga, 849-8501, Japan
| | - Keiichiro Uchida
- Department of Pharmacology, Faculty of Medicine, Saga University, Saga, 849-8501, Japan
| | - Hai-Lei Zhu
- Department of Pharmacology, Faculty of Medicine, Saga University, Saga, 849-8501, Japan
| | - Atsushi Shibata
- Department of Pharmacology, Faculty of Medicine, Saga University, Saga, 849-8501, Japan
| | - Tetsuichiro Inai
- Department of Morphological Biology, Division of Biomedical Sciences, Fukuoka Dental College, Fukuoka, 814-0193, Japan
| | - Mitsuru Noguchi
- Department of Urology, Faculty of Medicine, Saga University, Saga, 849-8501, Japan
| | - Mari Yotsu-Yamashita
- Laboratory of Bioorganic Chemistry of Natural Products, Graduate School of Agricultural Science, Tohoku University, Sendai, 981-8555, Japan
| | - Noriyoshi Teramoto
- Department of Pharmacology, Faculty of Medicine, Saga University, Saga, 849-8501, Japan. .,Laboratory of Biomedical Engineering, Graduate School of Biomedical Engineering, Tohoku University, Sendai, 980-0845, Japan.
| |
Collapse
|
42
|
Israel MR, Thongyoo P, Deuis JR, Craik DJ, Vetter I, Durek T. The E15R Point Mutation in Scorpion Toxin Cn2 Uncouples Its Depressant and Excitatory Activities on Human NaV1.6. J Med Chem 2018; 61:1730-1736. [DOI: 10.1021/acs.jmedchem.7b01609] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/30/2023]
Affiliation(s)
- Mathilde R. Israel
- Institute
for Molecular Bioscience, The University of Queensland, Brisbane, Queensland 4072, Australia
| | - Panumart Thongyoo
- Institute
for Molecular Bioscience, The University of Queensland, Brisbane, Queensland 4072, Australia
| | - Jennifer R. Deuis
- Institute
for Molecular Bioscience, The University of Queensland, Brisbane, Queensland 4072, Australia
| | - David J. Craik
- Institute
for Molecular Bioscience, The University of Queensland, Brisbane, Queensland 4072, Australia
| | - Irina Vetter
- Institute
for Molecular Bioscience, The University of Queensland, Brisbane, Queensland 4072, Australia
- School
of Pharmacy, The University of Queensland, Woolloongabba, Queensland 4102, Australia
| | - Thomas Durek
- Institute
for Molecular Bioscience, The University of Queensland, Brisbane, Queensland 4072, Australia
| |
Collapse
|
43
|
Identification of Persistent and Resurgent Sodium Currents in Spiral Ganglion Neurons Cultured from the Mouse Cochlea. eNeuro 2017; 4:eN-NWR-0303-17. [PMID: 29138759 PMCID: PMC5684619 DOI: 10.1523/eneuro.0303-17.2017] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2017] [Revised: 10/13/2017] [Accepted: 10/17/2017] [Indexed: 01/11/2023] Open
Abstract
In spiral ganglion neurons (SGNs), the afferent single units of the auditory nerve, high spontaneous and evoked firing rates ensure preservation of the temporal code describing the key features of incoming sound. During postnatal development, the spatiotemporal distribution of ion channel subtypes contributes to the maturation of action potential generation in SGNs, and to their ability to generate spike patterns that follow rapidly changing inputs. Here we describe tetrodotoxin (TTX)-sensitive Na+ currents in SGNs cultured from mice, whose properties may support this fast spiking behavior. A subthreshold persistent Na+ current (INaP) and a resurgent Na+ current (INaR) both emerged prior to the onset of hearing and became more prevalent as hearing matured. Navβ4 subunits, which are proposed to play a key role in mediating INaR elsewhere in the nervous system, were immunolocalized to the first heminode where spikes are generated in the auditory nerve, and to perisomatic nodes of Ranvier. ATX-II, a sea anemone toxin that slows classical Na+ channel inactivation selectively, enhanced INaP five-fold and INaR three-fold in voltage clamp recordings. In rapidly-adapting SGNs under current clamp, ATX-II increased the likelihood of firing additional action potentials. The data identify INaP and INaR as novel regulators of excitability in SGNs, and consistent with their roles in other neuronal types, we suggest that these nonclassical Na+ currents may contribute to the control of refractoriness in the auditory nerve.
Collapse
|
44
|
Tsukamoto T, Chiba Y, Wakamori M, Yamada T, Tsunogae S, Cho Y, Sakakibara R, Imazu T, Tokoro S, Satake Y, Adachi M, Nishikawa T, Yotsu-Yamashita M, Konoki K. Differential binding of tetrodotoxin and its derivatives to voltage-sensitive sodium channel subtypes (Na v 1.1 to Na v 1.7). Br J Pharmacol 2017; 174:3881-3892. [PMID: 28832970 DOI: 10.1111/bph.13985] [Citation(s) in RCA: 45] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2016] [Revised: 07/28/2017] [Accepted: 08/01/2017] [Indexed: 12/19/2022] Open
Abstract
BACKGROUND AND PURPOSE The development of subtype-selective ligands to inhibit voltage-sensitive sodium channels (VSSCs) has been attempted with the aim of developing therapeutic compounds. Tetrodotoxin (TTX) is a toxin from pufferfish that strongly inhibits VSSCs. Many TTX analogues have been identified from marine and terrestrial sources, although their specificity for particular VSSC subtypes has not been investigated. Herein, we describe the binding of 11 TTX analogues to human VSSC subtypes Nav 1.1-Nav 1.7. EXPERIMENTAL APPROACH Each VSSC subtype was transiently expressed in HEK293T cells. The inhibitory effects of TTX analogues on each subtype were assessed using whole-cell patch-clamp recordings. KEY RESULTS The inhibitory effects of TTX on Nav 1.1-Nav 1.7 were observed in accordance with those reported in the literature; however, the 5-deoxy-10,7-lactone-type analogues and 4,9-anhydro-type analogues did not cause inhibition. Chiriquitoxin showed less binding to Nav 1.7 compared to the other TTX-sensitive subtypes. Two amino acid residues in the TTX binding site of Nav 1.7, Thr1425 and Ile1426 were mutated to Met and Asp, respectively, because these residues were found at the same positions in other subtypes. The two mutants, Nav 1.7 T1425M and Nav 1.7 I1426D, had a 16-fold and 5-fold increase in binding affinity for chiriquitoxin, respectively. CONCLUSIONS AND IMPLICATIONS The reduced binding of chiriquitoxin to Nav 1.7 was attributed to its C11-OH and/or C12-NH2 , based on reported models for the TTX-VSSC complex. Chiriquitoxin is a useful tool for probing the configuration of the TTX binding site until a crystal structure for the mammalian VSSC is solved.
Collapse
Affiliation(s)
- Tadaaki Tsukamoto
- Graduate School of Agricultural Science, Tohoku University, Sendai, Japan
| | - Yukie Chiba
- Graduate School of Agricultural Science, Tohoku University, Sendai, Japan
| | - Minoru Wakamori
- Graduate School of Dentistry, Tohoku University, Sendai, Japan
| | - Tomoshi Yamada
- Graduate School of Agricultural Science, Tohoku University, Sendai, Japan
| | - Shunsuke Tsunogae
- Graduate School of Agricultural Science, Tohoku University, Sendai, Japan
| | - Yuko Cho
- Graduate School of Agricultural Science, Tohoku University, Sendai, Japan
| | - Ryo Sakakibara
- Graduate School of Bioagricultural Sciences, Nagoya University, Nagoya, Japan
| | - Takuya Imazu
- Graduate School of Bioagricultural Sciences, Nagoya University, Nagoya, Japan
| | - Shouta Tokoro
- Graduate School of Bioagricultural Sciences, Nagoya University, Nagoya, Japan
| | - Yoshiki Satake
- Graduate School of Bioagricultural Sciences, Nagoya University, Nagoya, Japan
| | - Masaatsu Adachi
- Graduate School of Bioagricultural Sciences, Nagoya University, Nagoya, Japan
| | - Toshio Nishikawa
- Graduate School of Bioagricultural Sciences, Nagoya University, Nagoya, Japan
| | | | - Keiichi Konoki
- Graduate School of Agricultural Science, Tohoku University, Sendai, Japan
| |
Collapse
|
45
|
Sodium Channel β2 Subunits Prevent Action Potential Propagation Failures at Axonal Branch Points. J Neurosci 2017; 37:9519-9533. [PMID: 28871036 DOI: 10.1523/jneurosci.0891-17.2017] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2017] [Revised: 08/21/2017] [Accepted: 08/22/2017] [Indexed: 12/19/2022] Open
Abstract
Neurotransmitter release depends on voltage-gated Na+ channels (Navs) to propagate an action potential (AP) successfully from the axon hillock to a synaptic terminal. Unmyelinated sections of axon are very diverse structures encompassing branch points and numerous presynaptic terminals with undefined molecular partners of Na+ channels. Using optical recordings of Ca2+ and membrane voltage, we demonstrate here that Na+ channel β2 subunits (Navβ2s) are required to prevent AP propagation failures across the axonal arborization of cultured rat hippocampal neurons (mixed male and female). When Navβ2 expression was reduced, we identified two specific phenotypes: (1) membrane excitability and AP-evoked Ca2+ entry were impaired at synapses and (2) AP propagation was severely compromised with >40% of axonal branches no longer responding to AP-stimulation. We went on to show that a great deal of electrical signaling heterogeneity exists in AP waveforms across the axonal arborization independent of axon morphology. Therefore, Navβ2 is a critical regulator of axonal excitability and synaptic function in unmyelinated axons.SIGNIFICANCE STATEMENT Voltage-gated Ca2+ channels are fulcrums of neurotransmission that convert electrical inputs into chemical outputs in the form of vesicle fusion at synaptic terminals. However, the role of the electrical signal, the presynaptic action potential (AP), in modulating synaptic transmission is less clear. What is the fidelity of a propagating AP waveform in the axon and what molecules shape it throughout the axonal arborization? Our work identifies several new features of AP propagation in unmyelinated axons: (1) branches of a single axonal arborization have variable AP waveforms independent of morphology, (2) Na+ channel β2 subunits modulate AP-evoked Ca2+-influx, and (3) β2 subunits maintain successful AP propagation across the axonal arbor. These findings are relevant to understanding the flow of excitation in the brain.
Collapse
|
46
|
Pro-excitatory alterations in sodium channel activity facilitate subiculum neuron hyperexcitability in temporal lobe epilepsy. Neurobiol Dis 2017; 108:183-194. [PMID: 28860087 DOI: 10.1016/j.nbd.2017.08.018] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2017] [Revised: 08/07/2017] [Accepted: 08/26/2017] [Indexed: 11/23/2022] Open
Abstract
Temporal lobe epilepsy (TLE) is a common form of adult epilepsy involving the limbic structures of the temporal lobe. Subiculum neurons act to provide a major output from the hippocampus and consist of a large population of endogenously bursting excitatory neurons. In TLE, subiculum neurons are largely spared, become hyperexcitable and show spontaneous epileptiform activity. The basis for this hyperexcitability is unclear, but is likely to involve alterations in the expression levels and function of various ion channels. In this study, we sought to determine the importance of sodium channel currents in facilitating neuronal hyperexcitability of subiculum neurons in the continuous hippocampal stimulation (CHS) rat model of TLE. Subiculum neurons from TLE rats were hyperexcitable, firing a higher frequency of action potentials after somatic current injection and action potential (AP) bursts after synaptic stimulation. Voltage clamp recordings revealed increases in resurgent (INaR) and persistent (INaP) sodium channel currents and pro-excitatory shifts in sodium channel activation and inactivation parameters that would facilitate increases in AP generation. Attenuation of INaR and INaP currents with 4,9-anhydro-tetrodotoxin (4,9-ah TTX; 100nM), a toxin with increased potency against Nav1.6 channels, suppressed neuronal firing frequency and inhibited AP bursting induced by synaptic stimulation in TLE neurons. These findings support an important role of sodium channels, particularly Nav1.6, in facilitating subiculum neuron hyperexcitability in TLE and provide further support for the importance of INaR and INaP currents in establishing epileptiform activity of subiculum neurons.
Collapse
|
47
|
CDYL suppresses epileptogenesis in mice through repression of axonal Nav1.6 sodium channel expression. Nat Commun 2017; 8:355. [PMID: 28842554 PMCID: PMC5572458 DOI: 10.1038/s41467-017-00368-z] [Citation(s) in RCA: 37] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2016] [Accepted: 06/25/2017] [Indexed: 12/27/2022] Open
Abstract
Impairment of intrinsic plasticity is involved in a range of neurological disorders such as epilepsy. However, how intrinsic excitability is regulated is still not fully understood. Here we report that the epigenetic factor Chromodomain Y-like (CDYL) protein is a critical regulator of the initiation and maintenance of intrinsic neuroplasticity by regulating voltage-gated ion channels in mouse brains. CDYL binds to a regulatory element in the intron region of SCN8A and mainly recruits H3K27me3 activity for transcriptional repression of the gene. Knockdown of CDYL in hippocampal neurons results in augmented Nav1.6 currents, lower neuronal threshold, and increased seizure susceptibility, whereas transgenic mice over-expressing CDYL exhibit higher neuronal threshold and are less prone to epileptogenesis. Finally, examination of human brain tissues reveals decreased CDYL and increased SCN8A in the temporal lobe epilepsy group. Together, our findings indicate CDYL is a critical player for experience-dependent gene regulation in controlling intrinsic excitability. Alterations in intrinsic plasticity are important in epilepsy. Here the authors show that the epigenetic factor CDYL regulates the gene expression of the voltage gated sodium channel, Nav1.6, which contributes to seizures in a rat model of epilepsy.
Collapse
|
48
|
Knutsen HK, Alexander J, Barregård L, Bignami M, Brüschweiler B, Ceccatelli S, Cottrill B, Dinovi M, Edler L, Grasl-Kraupp B, Hogstrand C, Hoogenboom LR, Nebbia CS, Oswald IP, Rose M, Roudot AC, Schwerdtle T, Vleminckx C, Vollmer G, Wallace H, Arnich N, Benford D, Botana L, Viviani B, Arcella D, Binaglia M, Horvath Z, Steinkellner H, van Manen M, Petersen A. Risks for public health related to the presence of tetrodotoxin (TTX) and TTX analogues in marine bivalves and gastropods. EFSA J 2017; 15:e04752. [PMID: 32625458 PMCID: PMC7010203 DOI: 10.2903/j.efsa.2017.4752] [Citation(s) in RCA: 31] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022] Open
Abstract
Tetrodotoxin (TTX) and its analogues are produced by marine bacteria and have been detected in marine bivalves and gastropods from European waters. The European Commission asked EFSA for a scientific opinion on the risks to public health related to the presence of TTX and TTX analogues in marine bivalves and gastropods. The Panel on Contaminants in the Food Chain reviewed the available literature but did not find support for the minimum lethal dose for humans of 2 mg, mentioned in various reviews. Some human case reports describe serious effects at a dose of 0.2 mg, corresponding to 4 μg/kg body weight (bw). However, the uncertainties on the actual exposure in the studies preclude their use for derivation of an acute reference dose (ARfD). Instead, a group ARfD of 0.25 μg/kg bw, applying to TTX and its analogues, was derived based on a TTX dose of 25 μg/kg bw at which no apathy was observed in an acute oral study with mice, applying a standard uncertainty factor of 100. Estimated relative potencies for analogues are lower than that of TTX but are associated with a high degree of uncertainty. Based on the occurrence data submitted to EFSA and reported consumption days only, average and P95 exposures of 0.00-0.09 and 0.00-0.03 μg/kg bw, respectively, were calculated. Using a large portion size of 400 g bivalves and P95 occurrence levels of TTX, with exception of oysters, the exposure was below the group ARfD in all consumer groups. A concentration below 44 μg TTX equivalents/kg shellfish meat, based on a large portion size of 400 g, was considered not to result in adverse effects in humans. Liquid chromatography with tandem mass spectroscopy (LC-MS/MS) methods are the most suitable for identification and quantification of TTX and its analogues, with LOQs between 1 and 25 μg/kg.
Collapse
|
49
|
Rodríguez C, Rollins-Smith L, Ibáñez R, Durant-Archibold AA, Gutiérrez M. Toxins and pharmacologically active compounds from species of the family Bufonidae (Amphibia, Anura). JOURNAL OF ETHNOPHARMACOLOGY 2017; 198:235-254. [PMID: 28034659 DOI: 10.1016/j.jep.2016.12.021] [Citation(s) in RCA: 67] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/29/2016] [Revised: 12/12/2016] [Accepted: 12/14/2016] [Indexed: 05/25/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Among amphibians, 15 of the 47 species reported to be used in traditional medicines belong to the family Bufonidae, which demonstrates their potential in pharmacological and natural products research. For example, Asian and American tribes use the skin and the parotoid gland secretions of some common toads in the treatment of hemorrhages, bites and stings from venomous animals, skin and stomach disorders, as well as several types of cancers. OVERARCHING OBJECTIVE In addition to reviewing the occurrence of chemical constituents present in the family Bufonidae, the cytotoxic and biomedical potential of the active compounds produced by different taxa are presented. METHODOLOGY Available information on bioactive compounds isolated from species of the family Bufonidae was obtained from ACS Publications, Google, Google Scholar, Pubmed, Sciendirect and Springer. Papers written in Chinese, English, German and Spanish were considered. RESULTS Recent reports show more than 30% of amphibians are in decline and some of bufonid species are considered to be extinct. For centuries, bufonids have been used as traditional folk remedies to treat allergies, inflammation, cancer, infections and other ailments, highlighting their importance as a prolific source for novel drugs and therapies. Toxins and bioactive chemical constituents from skin and parotid gland secretions of bufonid species can be grouped in five families, the guanidine alkaloids isolated and characterized from Atelopus, the lipophilic alkaloids isolated from Melanophryniscus, the indole alkaloids and bufadienolides known to be synthesized by species of bufonids, and peptides and proteins isolated from the skin and gastrointestinal extracts of some common toads. Overall, the bioactive secretions of this family of anurans may have antimicrobial, protease inhibitor and anticancer properties, as well as being active at the neuromuscular level. CONCLUSION In this article, the traditional uses, toxicity and pharmacological potential of chemical compounds from bufonids have been summarized. In spite of being reported to be used to treat several diseases, neither extracts nor metabolites from bufonids have been tested in such illness like acne, osteoporosis, arthritis and other illnesses. However, the cytotoxicity of these metabolites needs to be evaluated on adequate animal models due to the limited conditions of in vitro assays. Novel qualitative and quantitative tools based on MS spectrometry and Nuclear Magnetic Resonance spectroscopy is now available to study the complex secretions of bufonids.
Collapse
Affiliation(s)
- Candelario Rodríguez
- Centro de Biodiversidad y Descubrimiento de Drogas, Instituto de Investigaciones, Científicas y Servicios de Alta Tecnología (INDICASAT-AIP), City of Knowledge, Panama 0843-01103, Republic of Panama; Department of Biotechnology, Acharya Nagarjuna University, Guntur 522510, India
| | - Louise Rollins-Smith
- Department of Pathology, Microbiology, and Immunology, and Department of Pediatrics, Vanderbilt University School of Medicine, Nashville, TN 37232, USA; Department of Biological Sciences, Vanderbilt University, Nashville, TN 37235, USA
| | - Roberto Ibáñez
- Smithsonian Tropical Research Institute, Ancon, Panama 0843-03092, Republic of Panama; Department of Zoology, College of Natural, Exact Sciences and Technology, University of Panama, Republic of Panama
| | - Armando A Durant-Archibold
- Centro de Biodiversidad y Descubrimiento de Drogas, Instituto de Investigaciones, Científicas y Servicios de Alta Tecnología (INDICASAT-AIP), City of Knowledge, Panama 0843-01103, Republic of Panama; Department of Biochemistry, College of Natural, Exact Sciences and Technology, University of Panama, Republic of Panama.
| | - Marcelino Gutiérrez
- Centro de Biodiversidad y Descubrimiento de Drogas, Instituto de Investigaciones, Científicas y Servicios de Alta Tecnología (INDICASAT-AIP), City of Knowledge, Panama 0843-01103, Republic of Panama.
| |
Collapse
|
50
|
Maehara T, Motoyama K, Toma T, Yokoshima S, Fukuyama T. Total Synthesis of (−)‐Tetrodotoxin and 11‐norTTX‐6(
R
)‐ol. Angew Chem Int Ed Engl 2017; 56:1549-1552. [DOI: 10.1002/anie.201611574] [Citation(s) in RCA: 36] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2016] [Indexed: 11/12/2022]
Affiliation(s)
- Tomoaki Maehara
- Graduate School of Pharmaceutical SciencesNagoya University Furo-cho, Chikusa-ku Nagoya 464-8601 Japan
| | - Keisuke Motoyama
- Graduate School of Pharmaceutical SciencesUniversity of Tokyo 7-3-1 Hongo, Bunkyo-ku Tokyo 113-0033 Japan
| | - Tatsuya Toma
- Graduate School of Pharmaceutical SciencesNagoya University Furo-cho, Chikusa-ku Nagoya 464-8601 Japan
| | - Satoshi Yokoshima
- Graduate School of Pharmaceutical SciencesNagoya University Furo-cho, Chikusa-ku Nagoya 464-8601 Japan
| | - Tohru Fukuyama
- Graduate School of Pharmaceutical SciencesNagoya University Furo-cho, Chikusa-ku Nagoya 464-8601 Japan
| |
Collapse
|