151
|
Spider venom-derived peptide induces hyperalgesia in Na v1.7 knockout mice by activating Na v1.9 channels. Nat Commun 2020; 11:2293. [PMID: 32385249 PMCID: PMC7210961 DOI: 10.1038/s41467-020-16210-y] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2019] [Accepted: 04/21/2020] [Indexed: 01/05/2023] Open
Abstract
The sodium channels Nav1.7, Nav1.8 and Nav1.9 are critical for pain perception in peripheral nociceptors. Loss of function of Nav1.7 leads to congenital insensitivity to pain in humans. Here we show that the spider peptide toxin called HpTx1, first identified as an inhibitor of Kv4.2, restores nociception in Nav1.7 knockout (Nav1.7-KO) mice by enhancing the excitability of dorsal root ganglion neurons. HpTx1 inhibits Nav1.7 and activates Nav1.9 but does not affect Nav1.8. This toxin produces pain in wild-type (WT) and Nav1.7-KO mice, and attenuates nociception in Nav1.9-KO mice, but has no effect in Nav1.8-KO mice. These data indicate that HpTx1-induced hypersensitivity is mediated by Nav1.9 activation and offers pharmacological insight into the relationship of the three Nav channels in pain signalling. Loss of function of Nav1.7 leads to congenital insensitivity to pain in humans. Here the authors found that activation of Nav1.9 can restore nociception in Nav1.7 knockout mice, revealed by a venom-derived peptide as a probe.
Collapse
|
152
|
Dendritic Spine Dynamics after Peripheral Nerve Injury: An Intravital Structural Study. J Neurosci 2020; 40:4297-4308. [PMID: 32371602 DOI: 10.1523/jneurosci.2858-19.2020] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2019] [Revised: 02/10/2020] [Accepted: 02/28/2020] [Indexed: 11/21/2022] Open
Abstract
Neuropathic pain is an intractable medical condition with few or no options for effective treatment. Emerging evidence shows a strong structure-function relationship between dendritic spine dysgenesis and the presence of neuropathic pain. Postmortem tissue analyses can only imply dynamic structural changes associated with injury-induced pain. Here, we profiled the in vivo dynamics of dendritic spines over time on the same superficial dorsal horn (lamina II) neurons before and after peripheral nerve injury-induced pain. We used a two-photon, whole-animal imaging paradigm that permitted repeat imaging of the same dendritic branches of these neurons in C57/Bl6 Thy1-YFP male mice. Our study demonstrates, for the first time, the ongoing, steady-state changes in dendritic spine dynamics in the dorsal horn associated with peripheral nerve injury and pain. Ultimately, the relationship between altered dendritic spine dynamics and neuropathic pain may serve as a structure-based opportunity to investigate mechanisms of pain following injury and disease.SIGNIFICANCE STATEMENT This work is important because it demonstrates for the first time: (1) the powerful utility of intravital study of dendritic spine dynamics in the superficial dorsal horn; (2) that nerve injury-induced pain triggers changes in dendritic spine steady-state behavior in the spinal cord dorsal horn; and (3) this work opens the door to further investigations in vivo of spinal cord dendritic spine dynamics in the context of injury and disease.
Collapse
|
153
|
Tesfaye S, Sloan G. Diabetic Polyneuropathy - Advances in Diagnosis and Intervention Strategies. EUROPEAN ENDOCRINOLOGY 2020; 16:15-20. [PMID: 32595764 DOI: 10.17925/ee.2020.16.1.15] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/10/2020] [Accepted: 01/28/2020] [Indexed: 12/19/2022]
Abstract
Over half of people with diabetes mellitus develop diabetic polyneuropathy (DPN), which is a major cause of reduced quality of life due to disabling neuropathic pain, sensory loss, gait instability, fall-related injury, and foot ulceration and amputation. The latter represents a major health and economic burden, with lower limb amputation rates related to diabetes increasing in the UK. There is a need for early diagnosis of DPN so that early management strategies may be instigated, such as achieving tight glucose control and management of cardiovascular risk factors, in an attempt to slow its progression. To this end, a one-stop microvascular assessment involving a combined eye, foot and renal screening clinic has proven feasible in the UK. Unfortunately, there are currently no approved disease-modifying therapies for DPN. Some disease-modifying agents have demonstrated efficacy, but further large trials using appropriate clinical endpoints are required before these treatments can be routinely recommended. There has been emerging evidence highlighting a reduction in vitamin D levels in cases of painful DPN and the potential for vitamin D supplementation in deficient individuals to improve neuropathic pain; however, this needs to be proved in randomised clinical trials. The use of established agents for neuropathic pain in DPN is limited by poor efficacy and adverse effects, but patient stratification using methods such as pain phenotyping are being tested to determine whether this improves the outcomes of such agents in clinical studies. In addition, innovative approaches such as the topical 8% capsaicin patch, new methods of electrical stimulation and novel therapeutic targets such as NaV1.7 offer promise for the future. This article aims to discuss the challenges of diagnosing and managing DPN and to review current and emerging lifestyle interventions and therapeutic options.
Collapse
Affiliation(s)
- Solomon Tesfaye
- Diabetes Research Unit, Royal Hallamshire Hospital, Sheffield Teaching Hospitals NHS Foundation Trust, Sheffield, UK
| | - Gordon Sloan
- Diabetes Research Unit, Royal Hallamshire Hospital, Sheffield Teaching Hospitals NHS Foundation Trust, Sheffield, UK
| |
Collapse
|
154
|
Tanabe Y, Shiraishi S, Hashimoto K, Ikeda K, Nishizawa D, Hasegawa J, Shimomura A, Ozaki Y, Tamura N, Yunokawa M, Yonemori K, Takano T, Kawabata H, Tamura K, Fujiwara Y, Shimizu C. Taxane-induced sensory peripheral neuropathy is associated with an SCN9A single nucleotide polymorphism in Japanese patients. BMC Cancer 2020; 20:325. [PMID: 32295642 PMCID: PMC7161266 DOI: 10.1186/s12885-020-06834-0] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2019] [Accepted: 04/06/2020] [Indexed: 12/26/2022] Open
Abstract
Background Sodium channels located in the dorsal root ganglion, particularly Nav1.7 and Nav1.8, encoded by SCN9A and SCN10A, respectively, act as molecular gatekeepers for pain detection. Our aim was to determine the association between TIPN and SCN9A and SCN10A polymorphisms. Methods Three single nucleotide polymorphisms (SNPs) in SCN9A and two in SCN10A were investigated using whole-genome genotyping data from 186 Japanese breast or ovarian cancer patients classified into two groups as follows: cases that developed taxane-induced grade 2–3 neuropathy (N = 108) and controls (N = 78) with grade 0–1 neuropathy. Multiple logistic regression analyses were conducted to evaluate associations between TIPN and SNP genotypes. Results SCN9A-rs13017637 was a significant predictor of grade 2 or higher TIPN (odds ratio (OR) = 3.463; P = 0.0050) after correction for multiple comparisons, and precision was improved when only breast cancer patients were included (OR 5.053, P = 0.0029). Moreover, rs13017637 was a significant predictor of grade 2 or higher TIPN 1 year after treatment (OR 3.906, P = 0.037), indicating its contribution to TIPN duration. Conclusion SCN9A rs13017637 was associated with the severity and duration of TIPN. These findings are highly exploratory and require replication and validation prior to any consideration of clinical use.
Collapse
Affiliation(s)
- Yuko Tanabe
- Department of Breast and Medical Oncology, National Cancer Center Hospital, 5-1-1 Tsukiji, Chuo-ku, Tokyo, 104-0045, Japan. .,Department of Medical Oncology, Toranomon Hospital, 2-2-2 Toranomon, Minato-ku, Tokyo, 105-8470, Japan.
| | - Seiji Shiraishi
- Department of Anesthesiology, Kohnodai Hospital, National Center for Global Health and Medicine, 1-7-1, Kohnodai, Ichikawa-shi, Chiba, 272-8516, Japan
| | - Kenji Hashimoto
- Department of Breast and Medical Oncology, National Cancer Center Hospital, 5-1-1 Tsukiji, Chuo-ku, Tokyo, 104-0045, Japan
| | - Kazutaka Ikeda
- Addictive Substance Project, Tokyo Metropolitan Institute of Medical Science, 2-1-6 Kamikitazawa, Setagaya-ku, Tokyo, 156-8506, Japan
| | - Daisuke Nishizawa
- Addictive Substance Project, Tokyo Metropolitan Institute of Medical Science, 2-1-6 Kamikitazawa, Setagaya-ku, Tokyo, 156-8506, Japan
| | - Junko Hasegawa
- Addictive Substance Project, Tokyo Metropolitan Institute of Medical Science, 2-1-6 Kamikitazawa, Setagaya-ku, Tokyo, 156-8506, Japan
| | - Akihiko Shimomura
- Department of Breast and Medical Oncology, National Cancer Center Hospital, 5-1-1 Tsukiji, Chuo-ku, Tokyo, 104-0045, Japan.,Department of Medical Oncology, Toranomon Hospital, 2-2-2 Toranomon, Minato-ku, Tokyo, 105-8470, Japan
| | - Yukinori Ozaki
- Department of Medical Oncology, Toranomon Hospital, 2-2-2 Toranomon, Minato-ku, Tokyo, 105-8470, Japan
| | - Nobuko Tamura
- Department of Breast and Endocrine Surgery, Toranomon Hospital, 2-2-2 Toranomon, Minato-ku, Tokyo, 105-8470, Japan
| | - Mayu Yunokawa
- Department of Breast and Medical Oncology, National Cancer Center Hospital, 5-1-1 Tsukiji, Chuo-ku, Tokyo, 104-0045, Japan
| | - Kan Yonemori
- Department of Breast and Medical Oncology, National Cancer Center Hospital, 5-1-1 Tsukiji, Chuo-ku, Tokyo, 104-0045, Japan
| | - Toshimi Takano
- Department of Medical Oncology, Toranomon Hospital, 2-2-2 Toranomon, Minato-ku, Tokyo, 105-8470, Japan
| | - Hidetaka Kawabata
- Department of Breast and Endocrine Surgery, Toranomon Hospital, 2-2-2 Toranomon, Minato-ku, Tokyo, 105-8470, Japan
| | - Kenji Tamura
- Department of Breast and Medical Oncology, National Cancer Center Hospital, 5-1-1 Tsukiji, Chuo-ku, Tokyo, 104-0045, Japan
| | - Yasuhiro Fujiwara
- Department of Breast and Medical Oncology, National Cancer Center Hospital, 5-1-1 Tsukiji, Chuo-ku, Tokyo, 104-0045, Japan
| | - Chikako Shimizu
- Department of Breast Medical Oncology, Comprehensive Cancer Center, National Center for Global Health and Medicine, 1-21-1 Toyama, Shinjuku-ku, Tokyo, 162-8655, Japan
| |
Collapse
|
155
|
Zhang H, Moyer BD, Yu V, McGivern JG, Jarosh M, Werley CA, Hecht VC, Babcock RJ, Dong K, Dempsey GT, McManus OB, Hempel CM. Correlation of Optical and Automated Patch Clamp Electrophysiology for Identification of Na V1.7 Inhibitors. SLAS DISCOVERY 2020; 25:434-446. [PMID: 32292096 DOI: 10.1177/2472555220914532] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
The voltage-gated sodium channel Nav1.7 is a genetically validated target for pain; pharmacological blockers are promising as a new class of nonaddictive therapeutics. The search for Nav1.7 subtype selective inhibitors requires a reliable, scalable, and sensitive assay. Previously, we developed an all-optical electrophysiology (Optopatch) Spiking HEK platform to study activity-dependent modulation of Nav1.7 in a format compatible with high-throughput screening. In this study, we benchmarked the Optopatch Spiking HEK assay with an existing validated automated electrophysiology assay on the IonWorks Barracuda (IWB) platform. In a pilot screen of 3520 compounds, which included compound plates from a random library as well as compound plates enriched for Nav1.7 inhibitors, the Optopatch Spiking HEK assay identified 174 hits, of which 143 were confirmed by IWB. The Optopatch Spiking HEK assay maintained the high reliability afforded by traditional fluorescent assays and further demonstrated comparable sensitivity to IWB measurements. We speculate that the Optopatch assay could provide an affordable high-throughput screening platform to identify novel Nav1.7 subtype selective inhibitors with diverse mechanisms of action, if coupled with a multiwell parallel optogenetic recording instrument.
Collapse
Affiliation(s)
| | - Bryan D Moyer
- Neuroscience, Amgen Research, Thousand Oaks, CA, USA
| | - Violeta Yu
- Neuroscience, Amgen Research, Cambridge, MA, USA
| | - Joseph G McGivern
- Discovery Technologies, Amgen Research, South San Francisco, CA, USA
| | | | | | - Vivian C Hecht
- Q-State Biosciences, Cambridge, MA, USA.,Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Ryan J Babcock
- Q-State Biosciences, Cambridge, MA, USA.,Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Kevin Dong
- Q-State Biosciences, Cambridge, MA, USA.,Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | | | | | - Chris M Hempel
- Q-State Biosciences, Cambridge, MA, USA.,Expressive Neuroscience, Syracuse, NY, USA
| |
Collapse
|
156
|
Agwa AJ, Tran P, Mueller A, Tran HNT, Deuis JR, Israel MR, McMahon KL, Craik DJ, Vetter I, Schroeder CI. Manipulation of a spider peptide toxin alters its affinity for lipid bilayers and potency and selectivity for voltage-gated sodium channel subtype 1.7. J Biol Chem 2020; 295:5067-5080. [PMID: 32139508 PMCID: PMC7152767 DOI: 10.1074/jbc.ra119.012281] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2019] [Revised: 03/03/2020] [Indexed: 02/05/2023] Open
Abstract
Huwentoxin-IV (HwTx-IV) is a gating modifier peptide toxin from spiders that has weak affinity for the lipid bilayer. As some gating modifier toxins have affinity for model lipid bilayers, a tripartite relationship among gating modifier toxins, voltage-gated ion channels, and the lipid membrane surrounding the channels has been proposed. We previously designed an HwTx-IV analogue (gHwTx-IV) with reduced negative charge and increased hydrophobic surface profile, which displays increased lipid bilayer affinity and in vitro activity at the voltage-gated sodium channel subtype 1.7 (NaV1.7), a channel targeted in pain management. Here, we show that replacements of the positively-charged residues that contribute to the activity of the peptide can improve gHwTx-IV's potency and selectivity for NaV1.7. Using HwTx-IV, gHwTx-IV, [R26A]gHwTx-IV, [K27A]gHwTx-IV, and [R29A]gHwTx-IV variants, we examined their potency and selectivity at human NaV1.7 and their affinity for the lipid bilayer. [R26A]gHwTx-IV consistently displayed the most improved potency and selectivity for NaV1.7, examined alongside off-target NaVs, compared with HwTx-IV and gHwTx-IV. The lipid affinity of each of the three novel analogues was weaker than that of gHwTx-IV, but stronger than that of HwTx-IV, suggesting a possible relationship between in vitro potency at NaV1.7 and affinity for lipid bilayers. In a murine NaV1.7 engagement model, [R26A]gHwTx-IV exhibited an efficacy comparable with that of native HwTx-IV. In summary, this study reports the development of an HwTx-IV analogue with improved in vitro selectivity for the pain target NaV1.7 and with an in vivo efficacy similar to that of native HwTx-IV.
Collapse
Affiliation(s)
- Akello J Agwa
- Institute for Molecular Bioscience, Centre for Pain Research, The University of Queensland, Brisbane, Queensland 4072, Australia
| | - Poanna Tran
- Institute for Molecular Bioscience, Centre for Pain Research, The University of Queensland, Brisbane, Queensland 4072, Australia
| | - Alexander Mueller
- Institute for Molecular Bioscience, Centre for Pain Research, The University of Queensland, Brisbane, Queensland 4072, Australia
| | - Hue N T Tran
- Institute for Molecular Bioscience, Centre for Pain Research, The University of Queensland, Brisbane, Queensland 4072, Australia
| | - Jennifer R Deuis
- Institute for Molecular Bioscience, Centre for Pain Research, The University of Queensland, Brisbane, Queensland 4072, Australia
| | - Mathilde R Israel
- Institute for Molecular Bioscience, Centre for Pain Research, The University of Queensland, Brisbane, Queensland 4072, Australia
| | - Kirsten L McMahon
- Institute for Molecular Bioscience, Centre for Pain Research, The University of Queensland, Brisbane, Queensland 4072, Australia
| | - David J Craik
- Institute for Molecular Bioscience, Centre for Pain Research, The University of Queensland, Brisbane, Queensland 4072, Australia
| | - Irina Vetter
- Institute for Molecular Bioscience, Centre for Pain Research, The University of Queensland, Brisbane, Queensland 4072, Australia
- School of Pharmacy, The University of Queensland, Woolloongabba, Queensland 4103, Australia
| | - Christina I Schroeder
- Institute for Molecular Bioscience, Centre for Pain Research, The University of Queensland, Brisbane, Queensland 4072, Australia
- National Cancer Institute, National Institutes of Health, Frederick, Maryland 21702, USA
| |
Collapse
|
157
|
Abstract
Chemotherapy-induced peripheral neuropathy (CIPN) is a major challenge, with increasing impact as oncological treatments, using potentially neurotoxic chemotherapy, improve cancer cure and survival. Acute CIPN occurs during chemotherapy, sometimes requiring dose reduction or cessation, impacting on survival. Around 30% of patients will still have CIPN a year, or more, after finishing chemotherapy. Accurate assessment is essential to improve knowledge around prevalence and incidence of CIPN. Consensus is needed to standardize assessment and diagnosis, with use of well-validated tools, such as the EORTC-CIPN 20. Detailed phenotyping of the clinical syndrome moves toward a precision medicine approach, to individualize treatment. Understanding significant risk factors and pre-existing vulnerability may be used to improve strategies for CIPN prevention, or to use targeted treatment for established CIPN. No preventive therapies have shown significant clinical efficacy, although there are promising novel agents such as histone deacetylase 6 (HDAC6) inhibitors, currently in early phase clinical trials for cancer treatment. Drug repurposing, eg, metformin, may offer an alternative therapeutic avenue. Established treatment for painful CIPN is limited. Following recommendations for general neuropathic pain is logical, but evidence for agents such as gabapentinoids and amitriptyline is weak. The only agent currently recommended by the American Society of Clinical Oncology is duloxetine. Mechanisms are complex with changes in ion channels (sodium, potassium, and calcium), transient receptor potential channels, mitochondrial dysfunction, and immune cell interactions. Improved understanding is essential to advance CIPN management. On a positive note, there are many potential sites for modulation, with novel analgesic approaches.
Collapse
Affiliation(s)
- Lesley A Colvin
- Chair of Pain Medicine, Division of Population Health and Genomics, Ninewells Hospital and Medical School, University of Dundee, Dundee, Scotland
| |
Collapse
|
158
|
Huang Z, Zhou X, Zhang J, Mai CL, Mai JZ, Liu C, Zhang H, Liu XG. Bulleyaconitine A Inhibits Itch and Itch Sensitization Induced by Histamine and Chloroquine. Neuroscience 2020; 429:68-77. [DOI: 10.1016/j.neuroscience.2019.12.048] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2019] [Revised: 12/11/2019] [Accepted: 12/31/2019] [Indexed: 02/07/2023]
|
159
|
Wang JT, Zheng YM, Chen YT, Gu M, Gao ZB, Nan FJ. Discovery of aryl sulfonamide-selective Nav1.7 inhibitors with a highly hydrophobic ethanoanthracene core. Acta Pharmacol Sin 2020; 41:293-302. [PMID: 31316182 PMCID: PMC7471454 DOI: 10.1038/s41401-019-0267-z] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2019] [Accepted: 05/30/2019] [Indexed: 01/19/2023] Open
Abstract
Nav1.7 channels are mainly distributed in the peripheral nervous system. Blockade of Nav1.7 channels with small-molecule inhibitors in humans might provide pain relief without affecting the central nervous system. Based on the facts that many reported Nav1.7-selective inhibitors contain aryl sulfonamide fragments, as well as a tricyclic antidepressant, maprotiline, has been found to inhibit Nav1.7 channels, we designed and synthesized a series of compounds with ethanoanthracene and aryl sulfonamide moieties. Their inhibitory activity on sodium channels were detected with electrophysiological techniques. We found that compound 10o potently inhibited Nav1.7 channels stably expressed in HEK293 cells (IC50 = 0.64 ± 0.30 nmol/L) and displayed a high Nav1.7/Nav1.5 selectivity. In mouse small-sized dorsal root ganglion neurons, compound 10o (10, 100 nmol/L) dose-dependently decreased the sodium currents and dramatically suppressed depolarizing current-elicited neuronal discharge. Preliminary in vivo experiments showed that compound 10o possessed good analgesic activity: in a mouse visceral pain model, administration of compound 10o (30−100 mg/kg, i.p.) effectively and dose-dependently suppressed acetic acid-induced writhing.
Collapse
|
160
|
Kushnarev M, Pirvulescu IP, Candido KD, Knezevic NN. Neuropathic pain: preclinical and early clinical progress with voltage-gated sodium channel blockers. Expert Opin Investig Drugs 2020; 29:259-271. [PMID: 32070160 DOI: 10.1080/13543784.2020.1728254] [Citation(s) in RCA: 32] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Introduction: Neuropathic pain is a chronic condition that significantly affects the quality of life of millions of people globally. Most of the pharmacologic treatments currently in use demonstrate modest efficacy and over half of all patients do not respond to medical management. Hence, there is a need for new, efficacious drugs. Evidence points toward voltage-gated sodium channels as a key target for novel analgesics.Area covered: The role of voltage-gated sodium channels in pain pathophysiology is illuminated and the preclinical and clinical data for new sodium channel blockers and toxin-derived lead compounds are examined. The expansion of approved sodium channel blockers is discussed along with the limitations of current research, trends in drug development, and the potential of personalized medicine.Expert opinion: The transition from preclinical to clinical studies can be difficult because of the inherent inability of animal models to express the complexities of pain states. Pain pathways are notoriously intricate and may be pharmacologically modulated at a variety of targets; it is unlikely that action at a single target could completely abolish a pain response because pain is rarely unifactorial. Combination therapy may be necessary and this could further confound the discovery of novel agents.
Collapse
Affiliation(s)
- Mikhail Kushnarev
- Department of Anesthesiology, Advocate Illinois Masonic Medical Center, Chicago, IL, USA
| | - Iulia Paula Pirvulescu
- Department of Anesthesiology, Advocate Illinois Masonic Medical Center, Chicago, IL, USA
| | - Kenneth D Candido
- Department of Anesthesiology, Advocate Illinois Masonic Medical Center, Chicago, IL, USA.,Department of Anesthesiology, College of Medicine, University of Illinois, Chicago, IL, USA.,Department of Surgery, College of Medicine, University of Illinois, Chicago, IL, USA
| | - Nebojsa Nick Knezevic
- Department of Anesthesiology, Advocate Illinois Masonic Medical Center, Chicago, IL, USA.,Department of Anesthesiology, College of Medicine, University of Illinois, Chicago, IL, USA.,Department of Surgery, College of Medicine, University of Illinois, Chicago, IL, USA
| |
Collapse
|
161
|
Pharmacological characterization of a rat Nav1.7 loss-of-function model with insensitivity to pain. Pain 2020; 161:1350-1360. [DOI: 10.1097/j.pain.0000000000001807] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
|
162
|
Li ZM, Chen LX, Li H. Voltage-gated Sodium Channels and Blockers: An Overview and Where Will They Go? Curr Med Sci 2019; 39:863-873. [PMID: 31845216 DOI: 10.1007/s11596-019-2117-0] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2018] [Revised: 09/02/2019] [Indexed: 11/27/2022]
Abstract
Voltage-gated sodium (Nav) channels are critical players in the generation and propagation of action potentials by triggering membrane depolarization. Mutations in Nav channels are associated with a variety of channelopathies, which makes them relevant targets for pharmaceutical intervention. So far, the cryoelectron microscopic structure of the human Nav1.2, Nav1.4, and Nav1.7 has been reported, which sheds light on the molecular basis of functional mechanism of Nav channels and provides a path toward structure-based drug discovery. In this review, we focus on the recent advances in the structure, molecular mechanism and modulation of Nav channels, and state updated sodium channel blockers for the treatment of pathophysiology disorders and briefly discuss where the blockers may be developed in the future.
Collapse
Affiliation(s)
- Zhi-Mei Li
- Hubei Key Laboratory of Natural Medicinal Chemistry and Resource Evaluation, School of Pharmacy, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Li-Xia Chen
- Wuya College of Innovation, Key Laboratory of Structure-Based Drug Design & Discovery, Ministry of Education, Shenyang Pharmaceutical University, Shenyang, 110016, China.
| | - Hua Li
- Hubei Key Laboratory of Natural Medicinal Chemistry and Resource Evaluation, School of Pharmacy, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China.
| |
Collapse
|
163
|
Verma P, Kienle A, Flockerzi D, Ramkrishna D. Using Bifurcation Theory for Exploring Pain. Ind Eng Chem Res 2019. [DOI: 10.1021/acs.iecr.9b04495] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Affiliation(s)
- Parul Verma
- Davidson School of Chemical Engineering, Purdue University, West Lafayette, Indiana 47907, United States
| | - Achim Kienle
- Max Planck Institute for Dynamics of Complex Technical Systems, Magdeburg 39106, Germany
- Otto von Guericke University, Magdeburg 39106, Germany
| | - Dietrich Flockerzi
- Max Planck Institute for Dynamics of Complex Technical Systems, Magdeburg 39106, Germany
- Otto von Guericke University, Magdeburg 39106, Germany
| | - Doraiswami Ramkrishna
- Davidson School of Chemical Engineering, Purdue University, West Lafayette, Indiana 47907, United States
| |
Collapse
|
164
|
Tran HNT, Tran P, Deuis JR, Agwa AJ, Zhang AH, Vetter I, Schroeder CI. Enzymatic Ligation of a Pore Blocker Toxin and a Gating Modifier Toxin: Creating Double-Knotted Peptides with Improved Sodium Channel NaV1.7 Inhibition. Bioconjug Chem 2019; 31:64-73. [DOI: 10.1021/acs.bioconjchem.9b00744] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Affiliation(s)
- Hue N. T. Tran
- Institute for Molecular Bioscience, The University of Queensland, Brisbane, Queensland 4072, Australia
| | - Poanna Tran
- 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
| | - Akello J. Agwa
- Institute for Molecular Bioscience, The University of Queensland, Brisbane, Queensland 4072, Australia
| | - Alan H. Zhang
- Center for Advanced Imaging, 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
| | - Christina I. Schroeder
- Institute for Molecular Bioscience, The University of Queensland, Brisbane, Queensland 4072, Australia
| |
Collapse
|
165
|
Grotle AK, Stone AJ. Exaggerated exercise pressor reflex in type 2 diabetes: Potential role of oxidative stress. Auton Neurosci 2019; 222:102591. [PMID: 31669797 PMCID: PMC6858935 DOI: 10.1016/j.autneu.2019.102591] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2019] [Revised: 10/08/2019] [Accepted: 10/08/2019] [Indexed: 02/08/2023]
Abstract
Type 2 diabetes mellitus (T2DM) leads to exaggerated cardiovascular responses to exercise, in part due to an exaggerated exercise pressor reflex. Accumulating data suggest excessive oxidative stress contributes to an exaggerated exercise pressor reflex in cardiovascular-related diseases. Excessive oxidative stress is also a primary underlying mechanism for the development and progression of T2DM. However, whether oxidative stress plays a role in mediating the exaggerated exercise pressor reflex in T2DM is not known. Therefore, this review explores the potential role of oxidative stress leading to increased activation of the afferent arm of the exercise pressor reflex. Several lines of evidence support direct and indirect effects of oxidative stress on the exercise pressor reflex. For example, intramuscular ROS may directly and indirectly (by attenuating contracting muscle blood flow) increase group III and IV afferent activity. Oxidative stress is a primary underlying mechanism for the development of neuropathic pain, which in turn is associated with increased group III and IV afferent activity. These are the same type of afferents that evoke muscle pain and the exercise pressor reflex. Furthermore, oxidative stress-induced release of inflammatory mediators may modulate afferent activity. Collectively, these alterations may result in a positive feedback loop that further amplifies the exercise pressor reflex. An exaggerated reflex increases the risk of adverse cardiovascular events. Thus, identifying the contribution of oxidative stress could provide a potential therapeutic target to reduce this risk in T2DM.
Collapse
Affiliation(s)
- Ann-Katrin Grotle
- Department of Kinesiology and Health Education, The University of Texas at Austin, Austin, TX 78712, United States of America
| | - Audrey J Stone
- Department of Kinesiology and Health Education, The University of Texas at Austin, Austin, TX 78712, United States of America.
| |
Collapse
|
166
|
Newly Discovered Action of HpTx3 from Venom of Heteropoda venatoria on Na v1.7 and Its Pharmacological Implications in Analgesia. Toxins (Basel) 2019; 11:toxins11120680. [PMID: 31757020 PMCID: PMC6950750 DOI: 10.3390/toxins11120680] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2019] [Revised: 11/17/2019] [Accepted: 11/18/2019] [Indexed: 02/07/2023] Open
Abstract
It has been reported that Heteropodatoxin3 (HpTx3), a peptidic neurotoxin purified from the venom of the spider species Heteropoda venatoria, could inhibit Kv4.2 channels. Our present study newly found that HpTx3 also has potent and selective inhibitory action on Nav1.7, with an IC50 of 135.61 ± 12.98 nM. Without effect on the current–voltage (I-V) relationship of Nav1.7, HpTx3 made minor alternation in the voltage-dependence of activation and steady-state inactivation of Nav1.7 (4.15 mV and 7.29 mV, respectively) by interacting with the extracellular S3–S4 loop (S3b–S4 sequence) in domain II and the domain IV of the Nav channel subtype, showing the characteristics of both pore blocker and gate modifier toxin. During the interaction of HpTx3 with the S3b–S4 sequence of Nav1.7, the amino acid residue D in the sequence played a key role. When administered intraperitoneally or intramuscularly, HpTx3 displayed potent analgesic activity in a dose-dependent manner in different mouse pain models induced by formalin, acetic acid, complete Freund’s adjuvant, hot plate, or spared nerve injury, demonstrating that acute, inflammatory, and neuropathic pains were all effectively inhibited by the toxin. In most cases HpTx3 at doses of ≥ 1mg/kg could produce the analgesic effect comparable to that of 1 mg/kg morphine. These results suggest that HpTx3 not only can be used as a molecular probe to investigate ion channel function and pain mechanism, but also has potential in the development of the drugs that treat the Nav1.7 channel-related pain.
Collapse
|
167
|
Tzakoniati F, Xu H, Li T, Garcia N, Kugel C, Payandeh J, Koth CM, Tate EW. Development of Photocrosslinking Probes Based on Huwentoxin-IV to Map the Site of Interaction on Nav1.7. Cell Chem Biol 2019; 27:306-313.e4. [PMID: 31732432 PMCID: PMC7083225 DOI: 10.1016/j.chembiol.2019.10.011] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2019] [Revised: 08/31/2019] [Accepted: 10/24/2019] [Indexed: 02/07/2023]
Abstract
Voltage-gated sodium (Nav) channels respond to changes in the membrane potential of excitable cells through the concerted action of four voltage-sensor domains (VSDs). Subtype Nav1.7 plays an important role in the propagation of signals in pain-sensing neurons and is a target for the clinical development of novel analgesics. Certain inhibitory cystine knot (ICK) peptides produced by venomous animals potently modulate Nav1.7; however, the molecular mechanisms underlying their selective binding and activity remain elusive. This study reports on the design of a library of photoprobes based on the potent spider toxin Huwentoxin-IV and the determination of the toxin binding interface on VSD2 of Nav1.7 through a photocrosslinking and tandem mass spectrometry approach. Our Huwentoxin-IV probes selectively crosslink to extracellular loop S1-S2 and helix S3 of VSD2 in a chimeric channel system. Our results provide a strategy that will enable mapping of sites of interaction of other ICK peptides on Nav channels. Development of six potent diazirine-containing photoprobes based on Huwentoxin-IV Photoprobes specifically photolabel purified bacterial-Nav1.7 VSD2 chimeric channels Proteomic mass spectrometry identifies binding site on S1-S2 loop and S3 helix Proposed model of HwTx-IV binding reveals importance of K27 and R29
Collapse
Affiliation(s)
| | - Hui Xu
- Department of Structural Biology, Genentech, South San Francisco, CA 94080, USA
| | - Tianbo Li
- Department of Biochemical and Cellular Pharmacology, Genentech, South San Francisco, CA 94080, USA
| | - Natalie Garcia
- Department of Protein Analytical Chemistry, Genentech, South San Francisco, CA 94080, USA
| | - Christine Kugel
- Department of Biomolecular Resources, Genentech, South San Francisco, CA 94080, USA
| | - Jian Payandeh
- Department of Structural Biology, Genentech, South San Francisco, CA 94080, USA
| | - Christopher M Koth
- Department of Structural Biology, Genentech, South San Francisco, CA 94080, USA
| | - Edward W Tate
- Department of Chemistry, Imperial College London, London W12 0BZ, UK.
| |
Collapse
|
168
|
Kuffler DP. Injury-Induced Effectors of Neuropathic Pain. Mol Neurobiol 2019; 57:51-66. [PMID: 31701439 DOI: 10.1007/s12035-019-01756-w] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2019] [Revised: 08/29/2019] [Indexed: 02/07/2023]
Abstract
Injuries typically result in the development of neuropathic pain, which decreases in parallel with wound healing. However, the pain may remain after the injury appears to have healed, which is generally associated with an ongoing underlying pro-inflammatory state. Injury induces many cells to release factors that contribute to the development of a pro-inflammatory state, which is considered an essential first step towards wound healing. However, pain elimination requires a transition of the injury site from pro- to anti-inflammatory. Therefore, developing techniques that eliminate chronic pain require an understanding of the cells resident at and recruited to injury sites, the factors they release, that promote a pro-inflammatory state, and promote the subsequent transition of that site to be anti-inflammatory. Although a relatively large number of cells, factors, and gene expression changes are involved in these processes, it may be possible to control a relatively small number of them leading to the reduction and elimination of chronic neuropathic pain. This first of two papers examines the roles of the most salient cells and mediators associated with the development and maintenance of chronic neuropathic pain. The following paper examines the cells and mediators involved in reducing and eliminating chronic neuropathic pain.
Collapse
Affiliation(s)
- Damien P Kuffler
- Institute of Neurobiology, Medical Sciences Campus, University of Puerto Rico, 201 Blvd. del Valle, San Juan, PR, 00901, USA.
| |
Collapse
|
169
|
Jovanović O, Škulj S, Pohl EE, Vazdar M. Covalent modification of phosphatidylethanolamine by 4-hydroxy-2-nonenal increases sodium permeability across phospholipid bilayer membranes. Free Radic Biol Med 2019; 143:433-440. [PMID: 31461663 PMCID: PMC7115857 DOI: 10.1016/j.freeradbiomed.2019.08.027] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/17/2019] [Revised: 08/12/2019] [Accepted: 08/23/2019] [Indexed: 12/25/2022]
Abstract
Reactive aldehydes (RAs), such as 4-hydroxy-2-nonenal (HNE) and 4-oxo-2-nonenal (ONE), produced by cells under conditions of oxidative stress, were shown to react with phosphatidylethanolamine (PE) in biological and artificial membranes. They form RA-PE adducts, which affect the function of membrane proteins by modifying various biophysical properties of the membrane. The ratio of protein to lipid in biological membranes is different, but can reach 0.25 in the membranes of oligodendrocytes. However, the impact of RA-PE adducts on permeability (P) of the neat lipid phase and molecular mechanism of their action are poorly understood. In this study, we showed that HNE increased the membrane P for ions, and in particular for sodium. This effect depended on the presence of DOPE, and was not recorded for the more toxic compound, ONE. Molecular dynamics simulations suggested that HNE-PE and ONE-PE adducts anchored different positions in the lipid bilayer, and thus changed the membrane lipid area and bilayer thickness in different ways. Sodium permeability, calculated in the presence of double HNE-PE adducts, was increased by three to four orders of magnitude when compared to PNa in adduct - free membranes. A novel mechanism by which HNE alters permeability of the lipid membrane may explain the multiple toxic or regulative effects of HNE on the function of excitable cells, such as neurons, cardiomyocytes and neurosensory cells under conditions of oxidative stress.
Collapse
Affiliation(s)
- Olga Jovanović
- Institute of Physiology, Pathophysiology and Biophysics, Department of Biomedical Sciences, University of Veterinary Medicine, Vienna, Austria
| | - Sanja Škulj
- Division of Organic Chemistry and Biochemistry, Rudjer Bošković Institute, Zagreb, Croatia
| | - Elena E Pohl
- Institute of Physiology, Pathophysiology and Biophysics, Department of Biomedical Sciences, University of Veterinary Medicine, Vienna, Austria.
| | - Mario Vazdar
- Division of Organic Chemistry and Biochemistry, Rudjer Bošković Institute, Zagreb, Croatia.
| |
Collapse
|
170
|
Spider Knottin Pharmacology at Voltage-Gated Sodium Channels and Their Potential to Modulate Pain Pathways. Toxins (Basel) 2019; 11:toxins11110626. [PMID: 31671792 PMCID: PMC6891507 DOI: 10.3390/toxins11110626] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2019] [Revised: 10/24/2019] [Accepted: 10/24/2019] [Indexed: 12/15/2022] Open
Abstract
Voltage-gated sodium channels (NaVs) are a key determinant of neuronal signalling. Neurotoxins from diverse taxa that selectively activate or inhibit NaV channels have helped unravel the role of NaV channels in diseases, including chronic pain. Spider venoms contain the most diverse array of inhibitor cystine knot (ICK) toxins (knottins). This review provides an overview on how spider knottins modulate NaV channels and describes the structural features and molecular determinants that influence their affinity and subtype selectivity. Genetic and functional evidence support a major involvement of NaV subtypes in various chronic pain conditions. The exquisite inhibitory properties of spider knottins over key NaV subtypes make them the best lead molecules for the development of novel analgesics to treat chronic pain.
Collapse
|
171
|
Yamashita N. Retrograde signaling via axonal transport through signaling endosomes. J Pharmacol Sci 2019; 141:91-96. [PMID: 31679963 DOI: 10.1016/j.jphs.2019.10.001] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2019] [Revised: 10/07/2019] [Accepted: 10/10/2019] [Indexed: 11/28/2022] Open
Abstract
Neurons extend axons far from cell bodies, and retrograde communications from distal axons to cell bodies and/or dendrites play critical roles in the development and maintenance of neuronal circuits. In neurotrophin signaling, the retrograde axonal transport of endosomes containing active ligand-receptor complexes from distal axons to somatodendrite compartments mediates retrograde signaling. However, the generality and specificity of these endosome-based transportations called "signaling endosomes" remain to be elucidated. Here, I summarize the discovery of semaphorin3A signaling endosomes, the first example other than neurotrophins to regulate dendritic development via AMPA receptor GluA2 localization in dendrites. The molecular components of Sema3A and neurotrophin signaling endosomes are distinct, but partially overlap to regulate specific and common cellular events. Because receptors are transported back to the cell bodies, neurons must replenish receptors on the growth cone surface to ensure continued response to the target-derived ligands. Recent findings have demonstrated that retrograde signaling endosomes also induce anterograde delivery of nascent receptors in neurotrophin signaling. The coupling between anterograde and retrograde axonal transport via signaling endosomes therefore plays a critical role in regulating proper neuronal network formation.
Collapse
Affiliation(s)
- Naoya Yamashita
- Department of Pharmacology, Juntendo University School of Medicine, Hongo 2-1-1, Bunkyo-ku, Tokyo, 113-8421, Japan.
| |
Collapse
|
172
|
Mulcahy JV, Pajouhesh H, Beckley JT, Delwig A, Bois JD, Hunter JC. Challenges and Opportunities for Therapeutics Targeting the Voltage-Gated Sodium Channel Isoform Na V1.7. J Med Chem 2019; 62:8695-8710. [PMID: 31012583 PMCID: PMC6786914 DOI: 10.1021/acs.jmedchem.8b01906] [Citation(s) in RCA: 58] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Voltage-gated sodium ion channel subtype 1.7 (NaV1.7) is a high interest target for the discovery of non-opioid analgesics. Compelling evidence from human genetic data, particularly the finding that persons lacking functional NaV1.7 are insensitive to pain, has spurred considerable effort to develop selective inhibitors of this Na+ ion channel target as analgesic medicines. Recent clinical setbacks and disappointing performance of preclinical compounds in animal pain models, however, have led to skepticism around the potential of selective NaV1.7 inhibitors as human therapeutics. In this Perspective, we discuss the attributes and limitations of recently disclosed investigational drugs targeting NaV1.7 and review evidence that, by better understanding the requirements for selectivity and target engagement, the opportunity to deliver effective analgesic medicines targeting NaV1.7 endures.
Collapse
Affiliation(s)
- John V. Mulcahy
- SiteOne Therapeutics, 280 Utah Ave, Suite 250, South San Francisco, CA 94080
| | - Hassan Pajouhesh
- SiteOne Therapeutics, 280 Utah Ave, Suite 250, South San Francisco, CA 94080
| | - Jacob T. Beckley
- SiteOne Therapeutics, 351 Evergreen Drive, Suite B1, Bozeman, MT 59715
| | - Anton Delwig
- SiteOne Therapeutics, 280 Utah Ave, Suite 250, South San Francisco, CA 94080
| | - J. Du Bois
- Stanford University, Lokey Chemistry and Biology, 337 Campus Drive, Stanford, CA 94305
| | - John C. Hunter
- SiteOne Therapeutics, 280 Utah Ave, Suite 250, South San Francisco, CA 94080
| |
Collapse
|
173
|
Foti RS, Biswas K, Aral J, Be X, Berry L, Cheng Y, Conner K, Falsey JR, Glaus C, Herberich B, Hickman D, Ikotun T, Li H, Long J, Huang L, Miranda LP, Murray J, Moyer B, Netirojjanakul C, Nixey TE, Sham K, Soto M, Tegley CM, Tran L, Wu B, Yin L, Rock DA. Use of Cryopreserved Hepatocytes as Part of an Integrated Strategy to Characterize In Vivo Clearance for Peptide-Antibody Conjugate Inhibitors of Nav1.7 in Preclinical Species. Drug Metab Dispos 2019; 47:1111-1121. [PMID: 31387871 DOI: 10.1124/dmd.119.087742] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2019] [Accepted: 07/08/2019] [Indexed: 02/13/2025] Open
Abstract
The identification of nonopioid alternatives to treat chronic pain has received a great deal of interest in recent years. Recently, the engineering of a series of Nav1.7 inhibitory peptide-antibody conjugates has been reported, and herein, the preclinical efforts to identify novel approaches to characterize the pharmacokinetic properties of the peptide conjugates are described. A cryopreserved plated mouse hepatocyte assay was designed to measure the depletion of the peptide-antibody conjugates from the media, with a correlation being observed between percentage remaining in the media and in vivo clearance (Pearson r = -0.5525). Physicochemical (charge and hydrophobicity), receptor-binding [neonatal Fc receptor (FcRn)], and in vivo pharmacokinetic data were generated and compared with the results from our in vitro hepatocyte assay, which was hypothesized to encompass all of the aforementioned properties. Correlations were observed among hydrophobicity; FcRn binding; depletion rates from the hepatocyte assay; and ultimately, in vivo clearance. Subsequent studies identified potential roles for the low-density lipoprotein and mannose/galactose receptors in the association of the Nav1.7 peptide conjugates with mouse hepatocytes, although in vivo studies suggested that FcRn was still the primary receptor involved in determining the pharmacokinetics of the peptide conjugates. Ultimately, the use of the cryopreserved hepatocyte assay along with FcRn binding and hydrophobic interaction chromatography provided an efficient and integrated approach to rapidly triage molecules for advancement while reducing the number of in vivo pharmacokinetic studies. SIGNIFICANCE STATEMENT: Although multiple in vitro and in silico tools are available in small-molecule drug discovery, pharmacokinetic characterization of protein therapeutics is still highly dependent upon the use of in vivo studies in preclinical species. The current work demonstrates the combined use of cryopreserved hepatocytes, hydrophobic interaction chromatography, and neonatal Fc receptor binding to characterize a series of Nav1.7 peptide-antibody conjugates prior to conducting in vivo studies, thus providing a means to rapidly evaluate novel protein therapeutic platforms while concomitantly reducing the number of in vivo studies conducted in preclinical species.
Collapse
Affiliation(s)
- Robert S Foti
- Pharmacokinetics and Drug Metabolism, Amgen Research, Cambridge, Massachusetts (R.S.F., X.B., L.B., D.H., L.H.); Therapeutic Discovery (K.B., J.A., Y.C., J.R.F., C.G., B.H., T.I., J.L., L.P.M., J.M., C.N., T.E.N., K.S., C.M.T., B.W., L.Y.), Neuroscience (B.M.), and Pharmacokinetics and Drug Metabolism (H.L., M.S., L.T.), Amgen Research, Thousand Oaks, California; and Pharmacokinetics and Drug Metabolism, Amgen Research, South San Francisco, California (K.C., D.A.R.)
| | - Kaustav Biswas
- Pharmacokinetics and Drug Metabolism, Amgen Research, Cambridge, Massachusetts (R.S.F., X.B., L.B., D.H., L.H.); Therapeutic Discovery (K.B., J.A., Y.C., J.R.F., C.G., B.H., T.I., J.L., L.P.M., J.M., C.N., T.E.N., K.S., C.M.T., B.W., L.Y.), Neuroscience (B.M.), and Pharmacokinetics and Drug Metabolism (H.L., M.S., L.T.), Amgen Research, Thousand Oaks, California; and Pharmacokinetics and Drug Metabolism, Amgen Research, South San Francisco, California (K.C., D.A.R.)
| | - Jennifer Aral
- Pharmacokinetics and Drug Metabolism, Amgen Research, Cambridge, Massachusetts (R.S.F., X.B., L.B., D.H., L.H.); Therapeutic Discovery (K.B., J.A., Y.C., J.R.F., C.G., B.H., T.I., J.L., L.P.M., J.M., C.N., T.E.N., K.S., C.M.T., B.W., L.Y.), Neuroscience (B.M.), and Pharmacokinetics and Drug Metabolism (H.L., M.S., L.T.), Amgen Research, Thousand Oaks, California; and Pharmacokinetics and Drug Metabolism, Amgen Research, South San Francisco, California (K.C., D.A.R.)
| | - Xuhai Be
- Pharmacokinetics and Drug Metabolism, Amgen Research, Cambridge, Massachusetts (R.S.F., X.B., L.B., D.H., L.H.); Therapeutic Discovery (K.B., J.A., Y.C., J.R.F., C.G., B.H., T.I., J.L., L.P.M., J.M., C.N., T.E.N., K.S., C.M.T., B.W., L.Y.), Neuroscience (B.M.), and Pharmacokinetics and Drug Metabolism (H.L., M.S., L.T.), Amgen Research, Thousand Oaks, California; and Pharmacokinetics and Drug Metabolism, Amgen Research, South San Francisco, California (K.C., D.A.R.)
| | - Loren Berry
- Pharmacokinetics and Drug Metabolism, Amgen Research, Cambridge, Massachusetts (R.S.F., X.B., L.B., D.H., L.H.); Therapeutic Discovery (K.B., J.A., Y.C., J.R.F., C.G., B.H., T.I., J.L., L.P.M., J.M., C.N., T.E.N., K.S., C.M.T., B.W., L.Y.), Neuroscience (B.M.), and Pharmacokinetics and Drug Metabolism (H.L., M.S., L.T.), Amgen Research, Thousand Oaks, California; and Pharmacokinetics and Drug Metabolism, Amgen Research, South San Francisco, California (K.C., D.A.R.)
| | - Yuan Cheng
- Pharmacokinetics and Drug Metabolism, Amgen Research, Cambridge, Massachusetts (R.S.F., X.B., L.B., D.H., L.H.); Therapeutic Discovery (K.B., J.A., Y.C., J.R.F., C.G., B.H., T.I., J.L., L.P.M., J.M., C.N., T.E.N., K.S., C.M.T., B.W., L.Y.), Neuroscience (B.M.), and Pharmacokinetics and Drug Metabolism (H.L., M.S., L.T.), Amgen Research, Thousand Oaks, California; and Pharmacokinetics and Drug Metabolism, Amgen Research, South San Francisco, California (K.C., D.A.R.)
| | - Kip Conner
- Pharmacokinetics and Drug Metabolism, Amgen Research, Cambridge, Massachusetts (R.S.F., X.B., L.B., D.H., L.H.); Therapeutic Discovery (K.B., J.A., Y.C., J.R.F., C.G., B.H., T.I., J.L., L.P.M., J.M., C.N., T.E.N., K.S., C.M.T., B.W., L.Y.), Neuroscience (B.M.), and Pharmacokinetics and Drug Metabolism (H.L., M.S., L.T.), Amgen Research, Thousand Oaks, California; and Pharmacokinetics and Drug Metabolism, Amgen Research, South San Francisco, California (K.C., D.A.R.)
| | - James R Falsey
- Pharmacokinetics and Drug Metabolism, Amgen Research, Cambridge, Massachusetts (R.S.F., X.B., L.B., D.H., L.H.); Therapeutic Discovery (K.B., J.A., Y.C., J.R.F., C.G., B.H., T.I., J.L., L.P.M., J.M., C.N., T.E.N., K.S., C.M.T., B.W., L.Y.), Neuroscience (B.M.), and Pharmacokinetics and Drug Metabolism (H.L., M.S., L.T.), Amgen Research, Thousand Oaks, California; and Pharmacokinetics and Drug Metabolism, Amgen Research, South San Francisco, California (K.C., D.A.R.)
| | - Charles Glaus
- Pharmacokinetics and Drug Metabolism, Amgen Research, Cambridge, Massachusetts (R.S.F., X.B., L.B., D.H., L.H.); Therapeutic Discovery (K.B., J.A., Y.C., J.R.F., C.G., B.H., T.I., J.L., L.P.M., J.M., C.N., T.E.N., K.S., C.M.T., B.W., L.Y.), Neuroscience (B.M.), and Pharmacokinetics and Drug Metabolism (H.L., M.S., L.T.), Amgen Research, Thousand Oaks, California; and Pharmacokinetics and Drug Metabolism, Amgen Research, South San Francisco, California (K.C., D.A.R.)
| | - Brad Herberich
- Pharmacokinetics and Drug Metabolism, Amgen Research, Cambridge, Massachusetts (R.S.F., X.B., L.B., D.H., L.H.); Therapeutic Discovery (K.B., J.A., Y.C., J.R.F., C.G., B.H., T.I., J.L., L.P.M., J.M., C.N., T.E.N., K.S., C.M.T., B.W., L.Y.), Neuroscience (B.M.), and Pharmacokinetics and Drug Metabolism (H.L., M.S., L.T.), Amgen Research, Thousand Oaks, California; and Pharmacokinetics and Drug Metabolism, Amgen Research, South San Francisco, California (K.C., D.A.R.)
| | - Dean Hickman
- Pharmacokinetics and Drug Metabolism, Amgen Research, Cambridge, Massachusetts (R.S.F., X.B., L.B., D.H., L.H.); Therapeutic Discovery (K.B., J.A., Y.C., J.R.F., C.G., B.H., T.I., J.L., L.P.M., J.M., C.N., T.E.N., K.S., C.M.T., B.W., L.Y.), Neuroscience (B.M.), and Pharmacokinetics and Drug Metabolism (H.L., M.S., L.T.), Amgen Research, Thousand Oaks, California; and Pharmacokinetics and Drug Metabolism, Amgen Research, South San Francisco, California (K.C., D.A.R.)
| | - Tayo Ikotun
- Pharmacokinetics and Drug Metabolism, Amgen Research, Cambridge, Massachusetts (R.S.F., X.B., L.B., D.H., L.H.); Therapeutic Discovery (K.B., J.A., Y.C., J.R.F., C.G., B.H., T.I., J.L., L.P.M., J.M., C.N., T.E.N., K.S., C.M.T., B.W., L.Y.), Neuroscience (B.M.), and Pharmacokinetics and Drug Metabolism (H.L., M.S., L.T.), Amgen Research, Thousand Oaks, California; and Pharmacokinetics and Drug Metabolism, Amgen Research, South San Francisco, California (K.C., D.A.R.)
| | - Hongyan Li
- Pharmacokinetics and Drug Metabolism, Amgen Research, Cambridge, Massachusetts (R.S.F., X.B., L.B., D.H., L.H.); Therapeutic Discovery (K.B., J.A., Y.C., J.R.F., C.G., B.H., T.I., J.L., L.P.M., J.M., C.N., T.E.N., K.S., C.M.T., B.W., L.Y.), Neuroscience (B.M.), and Pharmacokinetics and Drug Metabolism (H.L., M.S., L.T.), Amgen Research, Thousand Oaks, California; and Pharmacokinetics and Drug Metabolism, Amgen Research, South San Francisco, California (K.C., D.A.R.)
| | - Jason Long
- Pharmacokinetics and Drug Metabolism, Amgen Research, Cambridge, Massachusetts (R.S.F., X.B., L.B., D.H., L.H.); Therapeutic Discovery (K.B., J.A., Y.C., J.R.F., C.G., B.H., T.I., J.L., L.P.M., J.M., C.N., T.E.N., K.S., C.M.T., B.W., L.Y.), Neuroscience (B.M.), and Pharmacokinetics and Drug Metabolism (H.L., M.S., L.T.), Amgen Research, Thousand Oaks, California; and Pharmacokinetics and Drug Metabolism, Amgen Research, South San Francisco, California (K.C., D.A.R.)
| | - Liyue Huang
- Pharmacokinetics and Drug Metabolism, Amgen Research, Cambridge, Massachusetts (R.S.F., X.B., L.B., D.H., L.H.); Therapeutic Discovery (K.B., J.A., Y.C., J.R.F., C.G., B.H., T.I., J.L., L.P.M., J.M., C.N., T.E.N., K.S., C.M.T., B.W., L.Y.), Neuroscience (B.M.), and Pharmacokinetics and Drug Metabolism (H.L., M.S., L.T.), Amgen Research, Thousand Oaks, California; and Pharmacokinetics and Drug Metabolism, Amgen Research, South San Francisco, California (K.C., D.A.R.)
| | - Les P Miranda
- Pharmacokinetics and Drug Metabolism, Amgen Research, Cambridge, Massachusetts (R.S.F., X.B., L.B., D.H., L.H.); Therapeutic Discovery (K.B., J.A., Y.C., J.R.F., C.G., B.H., T.I., J.L., L.P.M., J.M., C.N., T.E.N., K.S., C.M.T., B.W., L.Y.), Neuroscience (B.M.), and Pharmacokinetics and Drug Metabolism (H.L., M.S., L.T.), Amgen Research, Thousand Oaks, California; and Pharmacokinetics and Drug Metabolism, Amgen Research, South San Francisco, California (K.C., D.A.R.)
| | - Justin Murray
- Pharmacokinetics and Drug Metabolism, Amgen Research, Cambridge, Massachusetts (R.S.F., X.B., L.B., D.H., L.H.); Therapeutic Discovery (K.B., J.A., Y.C., J.R.F., C.G., B.H., T.I., J.L., L.P.M., J.M., C.N., T.E.N., K.S., C.M.T., B.W., L.Y.), Neuroscience (B.M.), and Pharmacokinetics and Drug Metabolism (H.L., M.S., L.T.), Amgen Research, Thousand Oaks, California; and Pharmacokinetics and Drug Metabolism, Amgen Research, South San Francisco, California (K.C., D.A.R.)
| | - Bryan Moyer
- Pharmacokinetics and Drug Metabolism, Amgen Research, Cambridge, Massachusetts (R.S.F., X.B., L.B., D.H., L.H.); Therapeutic Discovery (K.B., J.A., Y.C., J.R.F., C.G., B.H., T.I., J.L., L.P.M., J.M., C.N., T.E.N., K.S., C.M.T., B.W., L.Y.), Neuroscience (B.M.), and Pharmacokinetics and Drug Metabolism (H.L., M.S., L.T.), Amgen Research, Thousand Oaks, California; and Pharmacokinetics and Drug Metabolism, Amgen Research, South San Francisco, California (K.C., D.A.R.)
| | - Chawita Netirojjanakul
- Pharmacokinetics and Drug Metabolism, Amgen Research, Cambridge, Massachusetts (R.S.F., X.B., L.B., D.H., L.H.); Therapeutic Discovery (K.B., J.A., Y.C., J.R.F., C.G., B.H., T.I., J.L., L.P.M., J.M., C.N., T.E.N., K.S., C.M.T., B.W., L.Y.), Neuroscience (B.M.), and Pharmacokinetics and Drug Metabolism (H.L., M.S., L.T.), Amgen Research, Thousand Oaks, California; and Pharmacokinetics and Drug Metabolism, Amgen Research, South San Francisco, California (K.C., D.A.R.)
| | - Thomas E Nixey
- Pharmacokinetics and Drug Metabolism, Amgen Research, Cambridge, Massachusetts (R.S.F., X.B., L.B., D.H., L.H.); Therapeutic Discovery (K.B., J.A., Y.C., J.R.F., C.G., B.H., T.I., J.L., L.P.M., J.M., C.N., T.E.N., K.S., C.M.T., B.W., L.Y.), Neuroscience (B.M.), and Pharmacokinetics and Drug Metabolism (H.L., M.S., L.T.), Amgen Research, Thousand Oaks, California; and Pharmacokinetics and Drug Metabolism, Amgen Research, South San Francisco, California (K.C., D.A.R.)
| | - Kelvin Sham
- Pharmacokinetics and Drug Metabolism, Amgen Research, Cambridge, Massachusetts (R.S.F., X.B., L.B., D.H., L.H.); Therapeutic Discovery (K.B., J.A., Y.C., J.R.F., C.G., B.H., T.I., J.L., L.P.M., J.M., C.N., T.E.N., K.S., C.M.T., B.W., L.Y.), Neuroscience (B.M.), and Pharmacokinetics and Drug Metabolism (H.L., M.S., L.T.), Amgen Research, Thousand Oaks, California; and Pharmacokinetics and Drug Metabolism, Amgen Research, South San Francisco, California (K.C., D.A.R.)
| | - Marcus Soto
- Pharmacokinetics and Drug Metabolism, Amgen Research, Cambridge, Massachusetts (R.S.F., X.B., L.B., D.H., L.H.); Therapeutic Discovery (K.B., J.A., Y.C., J.R.F., C.G., B.H., T.I., J.L., L.P.M., J.M., C.N., T.E.N., K.S., C.M.T., B.W., L.Y.), Neuroscience (B.M.), and Pharmacokinetics and Drug Metabolism (H.L., M.S., L.T.), Amgen Research, Thousand Oaks, California; and Pharmacokinetics and Drug Metabolism, Amgen Research, South San Francisco, California (K.C., D.A.R.)
| | - Christopher M Tegley
- Pharmacokinetics and Drug Metabolism, Amgen Research, Cambridge, Massachusetts (R.S.F., X.B., L.B., D.H., L.H.); Therapeutic Discovery (K.B., J.A., Y.C., J.R.F., C.G., B.H., T.I., J.L., L.P.M., J.M., C.N., T.E.N., K.S., C.M.T., B.W., L.Y.), Neuroscience (B.M.), and Pharmacokinetics and Drug Metabolism (H.L., M.S., L.T.), Amgen Research, Thousand Oaks, California; and Pharmacokinetics and Drug Metabolism, Amgen Research, South San Francisco, California (K.C., D.A.R.)
| | - Linh Tran
- Pharmacokinetics and Drug Metabolism, Amgen Research, Cambridge, Massachusetts (R.S.F., X.B., L.B., D.H., L.H.); Therapeutic Discovery (K.B., J.A., Y.C., J.R.F., C.G., B.H., T.I., J.L., L.P.M., J.M., C.N., T.E.N., K.S., C.M.T., B.W., L.Y.), Neuroscience (B.M.), and Pharmacokinetics and Drug Metabolism (H.L., M.S., L.T.), Amgen Research, Thousand Oaks, California; and Pharmacokinetics and Drug Metabolism, Amgen Research, South San Francisco, California (K.C., D.A.R.)
| | - Bin Wu
- Pharmacokinetics and Drug Metabolism, Amgen Research, Cambridge, Massachusetts (R.S.F., X.B., L.B., D.H., L.H.); Therapeutic Discovery (K.B., J.A., Y.C., J.R.F., C.G., B.H., T.I., J.L., L.P.M., J.M., C.N., T.E.N., K.S., C.M.T., B.W., L.Y.), Neuroscience (B.M.), and Pharmacokinetics and Drug Metabolism (H.L., M.S., L.T.), Amgen Research, Thousand Oaks, California; and Pharmacokinetics and Drug Metabolism, Amgen Research, South San Francisco, California (K.C., D.A.R.)
| | - Lin Yin
- Pharmacokinetics and Drug Metabolism, Amgen Research, Cambridge, Massachusetts (R.S.F., X.B., L.B., D.H., L.H.); Therapeutic Discovery (K.B., J.A., Y.C., J.R.F., C.G., B.H., T.I., J.L., L.P.M., J.M., C.N., T.E.N., K.S., C.M.T., B.W., L.Y.), Neuroscience (B.M.), and Pharmacokinetics and Drug Metabolism (H.L., M.S., L.T.), Amgen Research, Thousand Oaks, California; and Pharmacokinetics and Drug Metabolism, Amgen Research, South San Francisco, California (K.C., D.A.R.)
| | - Dan A Rock
- Pharmacokinetics and Drug Metabolism, Amgen Research, Cambridge, Massachusetts (R.S.F., X.B., L.B., D.H., L.H.); Therapeutic Discovery (K.B., J.A., Y.C., J.R.F., C.G., B.H., T.I., J.L., L.P.M., J.M., C.N., T.E.N., K.S., C.M.T., B.W., L.Y.), Neuroscience (B.M.), and Pharmacokinetics and Drug Metabolism (H.L., M.S., L.T.), Amgen Research, Thousand Oaks, California; and Pharmacokinetics and Drug Metabolism, Amgen Research, South San Francisco, California (K.C., D.A.R.)
| |
Collapse
|
174
|
Blass BE. Sodium Channel Modulators and Their Method of Use. ACS Med Chem Lett 2019; 10:1247-1248. [PMID: 31531190 DOI: 10.1021/acsmedchemlett.9b00341] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2019] [Indexed: 11/29/2022] Open
Affiliation(s)
- Benjamin E. Blass
- Temple University School of Pharmacy, Moulder Center for Drug Discovery Research, Philadelphia, Pennsylvania 19140, United States
| |
Collapse
|
175
|
Abstract
Sodium channel NaV1.7 is a major target in pain research, largely because of genetic validation. In this issue of Neuron, McDermott et al. (2019) now present a comprehensive assessment of congenital pain insensitivity due to NaV1.7 loss of function. This work and studies on NaV1.7 gain of function raise important questions about how channelopathies produce disease.
Collapse
|
176
|
Yamamoto PA, Conchon Costa AC, Lauretti GR, de Moraes NV. Pharmacogenomics in chronic pain therapy: from disease to treatment and challenges for clinical practice. Pharmacogenomics 2019; 20:971-982. [DOI: 10.2217/pgs-2019-0066] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Pharmacogenomics (PGx) has emerged as an encouraging tool in chronic pain therapy. Genetic variations associated with drug effectiveness or adverse reactions (amitriptyline/nortriptyline/codeine/oxycodone/tramadol-CYP2D6, amitriptyline-CYP2C19, carbamazepine-HLA-A, carbamazepine/oxcarbazepine-HLA-B) can be used to guide chronic pain management. Despite this evidence, many obstacles still need to be overcome for the effective clinical implementation of PGx. To translate the pharmacogenetic testing into actionable clinical decisions, the Clinical Pharmacogenetics Implementation Consortium has been developing guidelines for several drug–gene pairs. This review will show the applicability of PGx in chronic pain from disease to treatment; report the drug–gene pairs with strongest evidences in the clinic; and the challenges for the clinical implementation of PGx.
Collapse
Affiliation(s)
- Priscila Akemi Yamamoto
- São Paulo State University, UNESP - School of Pharmaceutical Sciences, Araraquara, SP, Brazil
| | - Ana Carolina Conchon Costa
- São Paulo University, USP – School of Pharmaceutical Sciences of Ribeirão Preto, Ribeirão Preto, SP, Brazil
| | - Gabriela Rocha Lauretti
- São Paulo University, USP – School of Medicine of Ribeirão Preto, Ribeirão Preto, SP, Brazil
| | - Natália Valadares de Moraes
- São Paulo State University, UNESP - School of Pharmaceutical Sciences, Araraquara, SP, Brazil
- Queen's University Belfast, Belfast, UK
| |
Collapse
|
177
|
Effraim PR, Huang J, Lampert A, Stamboulian S, Zhao P, Black JA, Dib-Hajj SD, Waxman SG. Fibroblast growth factor homologous factor 2 (FGF-13) associates with Nav1.7 in DRG neurons and alters its current properties in an isoform-dependent manner. NEUROBIOLOGY OF PAIN (CAMBRIDGE, MASS.) 2019; 6:100029. [PMID: 31223136 PMCID: PMC6565799 DOI: 10.1016/j.ynpai.2019.100029] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/18/2019] [Revised: 03/22/2019] [Accepted: 03/22/2019] [Indexed: 11/29/2022]
Abstract
Fibroblast Growth Factor Homologous Factors (FHF) constitute a subfamily of FGF proteins with four prototypes (FHF1-4; also known as FGF11-14). FHF proteins have been shown to bind directly to the membrane-proximal segment of the C-terminus in voltage-gated sodium channels (Nav), and regulate current density, availability, and frequency-dependent inhibition of sodium currents. Members of the FHF2 subfamily, FHF2A and FHF2B, differ in the length and sequence of their N-termini, and, importantly, differentially regulate Nav1.6 gating properties. Using immunohistochemistry, we show that FHF2 isoforms are expressed in adult dorsal root ganglion (DRG) neurons where they co-localize with Nav1.6 and Nav1.7. FHF2A and FHF2B show differential localization in neuronal compartments in DRG neurons, and levels of expression of FHF2 factors are down-regulated following sciatic nerve axotomy. Because Nav1.7 in nociceptors plays a critical role in pain, we reasoned that its interaction with FHF2 isoforms might regulate its current properties. Using whole-cell patch clamp in heterologous expression systems, we show that the expression of FHF2A in HEK293 cell line stably expressing Nav1.7 channels causes no change in activation, whereas FHF2B depolarizes activation. Both FHF2 isoforms depolarize fast-inactivation. Additionally, FHF2A causes an accumulation of inactivated channels at all frequencies tested due to a slowing of recovery from inactivation, whereas FHF2B has little effect on these properties of Nav1.7. Measurements of the Nav1.7 current in DRG neurons in which FHF2 levels are knocked down confirmed the effects of FHF2A on repriming, and FHF2B on activation, however FHF2A and B did not have an effect on fast inactivation. Our data demonstrates that FHF2 does indeed regulate the current properties of Nav1.7 and does so in an isoform and cell-specific manner.
Collapse
Affiliation(s)
- Philip R. Effraim
- Department of Anesthesiology, Yale University School of Medicine, New Haven, CT 06510, USA
- Center for Neuroscience and Regeneration Research, Yale University School of Medicine, New Haven, CT 06510, USA
- Rehabilitation Research Center, Veterans Affairs Connecticut Healthcare System, West Haven, CT 06516, USA
| | - Jianying Huang
- Department of Neurology, Yale University School of Medicine, New Haven, CT 06510, USA
- Center for Neuroscience and Regeneration Research, Yale University School of Medicine, New Haven, CT 06510, USA
- Rehabilitation Research Center, Veterans Affairs Connecticut Healthcare System, West Haven, CT 06516, USA
| | - Angelika Lampert
- Institute of Physiology, Uniklinik RWTH Aachen University, Aachen, Germany
| | - Severine Stamboulian
- Department of Neurology, Yale University School of Medicine, New Haven, CT 06510, USA
- Center for Neuroscience and Regeneration Research, Yale University School of Medicine, New Haven, CT 06510, USA
- Rehabilitation Research Center, Veterans Affairs Connecticut Healthcare System, West Haven, CT 06516, USA
| | - Peng Zhao
- Department of Neurology, Yale University School of Medicine, New Haven, CT 06510, USA
- Center for Neuroscience and Regeneration Research, Yale University School of Medicine, New Haven, CT 06510, USA
- Rehabilitation Research Center, Veterans Affairs Connecticut Healthcare System, West Haven, CT 06516, USA
| | - Joel A. Black
- Department of Neurology, Yale University School of Medicine, New Haven, CT 06510, USA
- Center for Neuroscience and Regeneration Research, Yale University School of Medicine, New Haven, CT 06510, USA
- Rehabilitation Research Center, Veterans Affairs Connecticut Healthcare System, West Haven, CT 06516, USA
| | - Sulayman D. Dib-Hajj
- Department of Neurology, Yale University School of Medicine, New Haven, CT 06510, USA
- Center for Neuroscience and Regeneration Research, Yale University School of Medicine, New Haven, CT 06510, USA
- Rehabilitation Research Center, Veterans Affairs Connecticut Healthcare System, West Haven, CT 06516, USA
| | - Stephen G. Waxman
- Department of Neurology, Yale University School of Medicine, New Haven, CT 06510, USA
- Center for Neuroscience and Regeneration Research, Yale University School of Medicine, New Haven, CT 06510, USA
- Rehabilitation Research Center, Veterans Affairs Connecticut Healthcare System, West Haven, CT 06516, USA
| |
Collapse
|
178
|
Chinn G, Guan Z. Case Report and Literature Review: Interventional Management of Erythromelalgia. ACTA ACUST UNITED AC 2019; 6:91-97. [PMID: 31592193 PMCID: PMC6779334 DOI: 10.31480/2330-4871/094] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/01/2022]
Abstract
Erythromelalgia is a rare and very difficult to treat pain syndrome that usually presents as severe bilateral burning pain in the extremities. Here we present a case of a 34-year-old female with erythromelalgia who we treated successfully with a lumbar epidural infusion of ropivacaine and fentanyl. The patient had complete relief shortly after the epidural infusion, and she remained stable with only minor pain two weeks and nine months later. With this case, we have reviewed the interventional treatments of erythromelalgia. We suggest epidural infusion as the first line interventional management, followed by sympathetic block. Spinal cord stimulation can be considered if other interventional managements fail.
Collapse
Affiliation(s)
- Gregory Chinn
- Department of Anesthesia and Perioperative Care, University of California San Francisco, San Francisco, California, USA
| | - Zhonghui Guan
- Department of Anesthesia and Perioperative Care, University of California San Francisco, San Francisco, California, USA
| |
Collapse
|
179
|
Bisphenol A Regulates Sodium Ramp Currents in Mouse Dorsal Root Ganglion Neurons and Increases Nociception. Sci Rep 2019; 9:10306. [PMID: 31312012 PMCID: PMC6635372 DOI: 10.1038/s41598-019-46769-6] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2019] [Accepted: 07/05/2019] [Indexed: 12/02/2022] Open
Abstract
17β-Estradiol mediates the sensitivity to pain and is involved in sex differences in nociception. The widespread environmental disrupting chemical bisphenol A (BPA) has estrogenic activity, but its implications in pain are mostly unknown. Here we show that treatment of male mice with BPA (50 µg/kg/day) during 8 days, decreases the latency to pain behavior in response to heat, suggesting increased pain sensitivity. We demonstrate that incubation of dissociated dorsal root ganglia (DRG) nociceptors with 1 nM BPA increases the frequency of action potential firing. SCN9A encodes the voltage-gated sodium channel Nav1.7, which is present in DRG nociceptors and is essential in pain signaling. Nav1.7 and other voltage-gated sodium channels in mouse DRG are considered threshold channels because they produce ramp currents, amplifying small depolarizations and enhancing electrical activity. BPA increased Nav-mediated ramp currents elicited with slow depolarizations. Experiments using pharmacological tools as well as DRG from ERβ−/− mice indicate that this BPA effect involves ERα and phosphoinositide 3-kinase. The mRNA expression and biophysical properties other than ramp currents of Nav channels, were unchanged by BPA. Our data suggest that BPA at environmentally relevant doses affects the ability to detect noxious stimuli and therefore should be considered when studying the etiology of pain conditions.
Collapse
|
180
|
An iPSC Model Reveals Mechanisms of Interindividual Differences in Pain. J Neurosci 2019; 39:5422-5423. [PMID: 31292212 DOI: 10.1523/jneurosci.0602-19.2019] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2019] [Revised: 05/09/2019] [Accepted: 05/14/2019] [Indexed: 11/21/2022] Open
|
181
|
Abstract
Acute pain is adaptive, but chronic pain is a global challenge. Many chronic pain syndromes are peripheral in origin and reflect hyperactivity of peripheral pain-signaling neurons. Current treatments are ineffective or only partially effective and in some cases can be addictive, underscoring the need for better therapies. Molecular genetic studies have now linked multiple human pain disorders to voltage-gated sodium channels, including disorders characterized by insensitivity or reduced sensitivity to pain and others characterized by exaggerated pain in response to normally innocuous stimuli. Here, we review recent developments that have enhanced our understanding of pathophysiological mechanisms in human pain and advances in targeting sodium channels in peripheral neurons for the treatment of pain using novel and existing sodium channel blockers.
Collapse
Affiliation(s)
- Sulayman D Dib-Hajj
- Department of Neurology and Center for Neuroscience and Regeneration Research, Yale University School of Medicine, New Haven, Connecticut 06510, USA; .,Rehabilitation Research Center, Veterans Affairs, Connecticut Healthcare System, West Haven, Connecticut 06516, USA
| | - Stephen G Waxman
- Department of Neurology and Center for Neuroscience and Regeneration Research, Yale University School of Medicine, New Haven, Connecticut 06510, USA; .,Rehabilitation Research Center, Veterans Affairs, Connecticut Healthcare System, West Haven, Connecticut 06516, USA
| |
Collapse
|
182
|
Abstract
The global epidemic of prediabetes and diabetes has led to a corresponding epidemic of complications of these disorders. The most prevalent complication is neuropathy, of which distal symmetric polyneuropathy (for the purpose of this Primer, referred to as diabetic neuropathy) is very common. Diabetic neuropathy is a loss of sensory function beginning distally in the lower extremities that is also characterized by pain and substantial morbidity. Over time, at least 50% of individuals with diabetes develop diabetic neuropathy. Glucose control effectively halts the progression of diabetic neuropathy in patients with type 1 diabetes mellitus, but the effects are more modest in those with type 2 diabetes mellitus. These findings have led to new efforts to understand the aetiology of diabetic neuropathy, along with new 2017 recommendations on approaches to prevent and treat this disorder that are specific for each type of diabetes. In parallel, new guidelines for the treatment of painful diabetic neuropathy using distinct classes of drugs, with an emphasis on avoiding opioid use, have been issued. Although our understanding of the complexities of diabetic neuropathy has substantially evolved over the past decade, the distinct mechanisms underlying neuropathy in type 1 and type 2 diabetes remains unknown. Future discoveries on disease pathogenesis will be crucial to successfully address all aspects of diabetic neuropathy, from prevention to treatment.
Collapse
|
183
|
Feldman EL, Callaghan BC, Pop-Busui R, Zochodne DW, Wright DE, Bennett DL, Bril V, Russell JW, Viswanathan V. Diabetic neuropathy. Nat Rev Dis Primers 2019; 5:42. [PMID: 31197183 PMCID: PMC7096070 DOI: 10.1038/s41572-019-0097-9] [Citation(s) in RCA: 107] [Impact Index Per Article: 17.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
The global epidemic of prediabetes and diabetes has led to a corresponding epidemic of complications of these disorders. The most prevalent complication is neuropathy, of which distal symmetric polyneuropathy (for the purpose of this Primer, referred to as diabetic neuropathy) is very common. Diabetic neuropathy is a loss of sensory function beginning distally in the lower extremities that is also characterized by pain and substantial morbidity. Over time, at least 50% of individuals with diabetes develop diabetic neuropathy. Glucose control effectively halts the progression of diabetic neuropathy in patients with type 1 diabetes mellitus, but the effects are more modest in those with type 2 diabetes mellitus. These findings have led to new efforts to understand the aetiology of diabetic neuropathy, along with new 2017 recommendations on approaches to prevent and treat this disorder that are specific for each type of diabetes. In parallel, new guidelines for the treatment of painful diabetic neuropathy using distinct classes of drugs, with an emphasis on avoiding opioid use, have been issued. Although our understanding of the complexities of diabetic neuropathy has substantially evolved over the past decade, the distinct mechanisms underlying neuropathy in type 1 and type 2 diabetes remains unknown. Future discoveries on disease pathogenesis will be crucial to successfully address all aspects of diabetic neuropathy, from prevention to treatment.
Collapse
Affiliation(s)
- Eva L. Feldman
- Department of Neurology, University of Michigan, Ann Arbor, MI, USA.,
| | | | - Rodica Pop-Busui
- Department of Internal Medicine, Division of Metabolism, Endocrinology and Diabetes (MEND), University of Michigan, Ann Arbor, MI, USA
| | - Douglas W. Zochodne
- Division of Neurology, Department of Medicine and the Neuroscience and Mental Health Institute, University of Alberta, Edmonton, Alberta, Canada
| | - Douglas E. Wright
- Department of Anatomy and Cell Biology, University of Kansas Medical Center, Kansas City, KS, USA
| | - David L. Bennett
- Nuffield Department of Clinical Neuroscience, University of Oxford, Oxford, UK
| | - Vera Bril
- Division of Neurology, Department of Medicine, University of Toronto and University Health Network, Toronto, Ontario, Canada.,Institute for Research and Medical Consultations, Imam Abdulrahman Bin Faisal University, Dammam, Saudi Arabia
| | - James W. Russell
- Department of Neurology, University of Maryland and VA Maryland Health Care System, Baltimore, MD, USA
| | | |
Collapse
|
184
|
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: 4.3] [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
|
185
|
|
186
|
Murray JK, Wu B, Tegley CM, Nixey TE, Falsey JR, Herberich B, Yin L, Sham K, Long J, Aral J, Cheng Y, Netirojjanakul C, Doherty L, Glaus C, Ikotun T, Li H, Tran L, Soto M, Salimi-Moosavi H, Ligutti J, Amagasu S, Andrews KL, Be X, Lin MHJ, Foti RS, Ilch CP, Youngblood B, Kornecook TJ, Karow M, Walker KW, Moyer BD, Biswas K, Miranda LP. Engineering Na V1.7 Inhibitory JzTx-V Peptides with a Potency and Basicity Profile Suitable for Antibody Conjugation To Enhance Pharmacokinetics. ACS Chem Biol 2019; 14:806-818. [PMID: 30875193 DOI: 10.1021/acschembio.9b00183] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Drug discovery research on new pain targets with human genetic validation, including the voltage-gated sodium channel NaV1.7, is being pursued to address the unmet medical need with respect to chronic pain and the rising opioid epidemic. As part of early research efforts on this front, we have previously developed NaV1.7 inhibitory peptide-antibody conjugates with tarantula venom-derived GpTx-1 toxin peptides with an extended half-life (80 h) in rodents but only moderate in vitro activity (hNaV1.7 IC50 = 250 nM) and without in vivo activity. We identified the more potent peptide JzTx-V from our natural peptide collection and improved its selectivity against other sodium channel isoforms through positional analogueing. Here we report utilization of the JzTx-V scaffold in a peptide-antibody conjugate and architectural variations in the linker, peptide loading, and antibody attachment site. We found conjugates with 100-fold improved in vitro potency relative to those of complementary GpTx-1 analogues, but pharmacokinetic and bioimaging analyses of these JzTx-V conjugates revealed a shorter than expected plasma half-life in vivo with accumulation in the liver. In an attempt to increase circulatory serum levels, we sought the reduction of the net +6 charge of the JzTx-V scaffold while retaining a desirable NaV in vitro activity profile. The conjugate of a JzTx-V peptide analogue with a +2 formal charge maintained NaV1.7 potency with 18-fold improved plasma exposure in rodents. Balancing the loss of peptide and conjugate potency associated with the reduction of net charge necessary for improved target exposure resulted in a compound with moderate activity in a NaV1.7-dependent pharmacodynamic model but requires further optimization to identify a conjugate that can fully engage NaV1.7 in vivo.
Collapse
|
187
|
Emerging therapies in clinical development and new contributions for neuropathic pain. ACTA ACUST UNITED AC 2019; 66:324-334. [PMID: 31010688 DOI: 10.1016/j.redar.2019.02.003] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2019] [Revised: 02/11/2019] [Accepted: 02/17/2019] [Indexed: 12/19/2022]
Abstract
Neuropathic pain is very challenging to manage because of the heterogeneity of aetiologies, symptoms, and underlying mechanisms. Conventional oral therapies have been limited by negative factors such as systemic side effects, drug-drug interactions, slow onset of action, the need for titration, multiple daily dosing, as well as the potential risk of addiction, dependence, withdrawal symptoms and abuse. Therefore, new therapeutic perspectives are justified. New drugs that act on different therapeutic targets are currently in preclinical development or in their first phases of clinical development. In this review, focus will be directed specifically on new pharmacological treatments for neuropathic pain for which clinical data are already available, including older and known drugs with new data on their anti-neuropathic activity.
Collapse
|
188
|
Zhao F, Tang Q, Xu J, Wang S, Li S, Zou X, Cao Z. Dehydrocrenatidine Inhibits Voltage-Gated Sodium Channels and Ameliorates Mechanic Allodia in a Rat Model of Neuropathic Pain. Toxins (Basel) 2019; 11:E229. [PMID: 31003411 PMCID: PMC6521113 DOI: 10.3390/toxins11040229] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2019] [Revised: 04/10/2019] [Accepted: 04/15/2019] [Indexed: 12/27/2022] Open
Abstract
Picrasma quassioides (D. Don) Benn, a medical plant, is used in clinic to treat inflammation, pain, sore throat, and eczema. The alkaloids are the main active components in P. quassioides. In this study, we examined the analgesic effect of dehydrocrenatidine (DHCT), a β-carboline alkaloid abundantly found in P. quassioides in a neuropathic pain rat model of a sciatic nerve chronic constriction injury. DHCT dose-dependently attenuated the mechanic allodynia. In acutely isolated dorsal root ganglion, DHCT completely suppressed the action potential firing. Further electrophysiological characterization demonstrated that DHCT suppressed both tetrodotoxin-resistant (TTX-R) and sensitive (TTX-S) voltage-gated sodium channel (VGSC) currents with IC50 values of 12.36 μM and 4.87 µM, respectively. DHCT shifted half-maximal voltage (V1/2) of inactivation to hyperpolarizing direction by ~16.7 mV in TTX-S VGSCs. In TTX-R VGSCs, DHCT shifted V1/2 of inactivation voltage to hyperpolarizing direction and V1/2 of activation voltage to more depolarizing potential by ~23.9 mV and ~12.2 mV, respectively. DHCT preferred to interact with an inactivated state of VGSCs and prolonged the repriming time in both TTX-S and TTX-R VGSCs, transiting the channels into a slow inactivated state from a fast inactivated state. Considered together, these data demonstrated that the analgesic effect of DHCT was likely though the inhibition of neuronal excitability.
Collapse
Affiliation(s)
- Fang Zhao
- Jiangsu Key Laboratory of TCM Evaluation and Translational Research, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing 211198, China.
| | - Qinglian Tang
- Jiangsu Key Laboratory of TCM Evaluation and Translational Research, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing 211198, China.
| | - Jian Xu
- Jiangsu Key Laboratory of TCM Evaluation and Translational Research, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing 211198, China.
| | - Shuangyan Wang
- Jiangsu Key Laboratory of TCM Evaluation and Translational Research, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing 211198, China.
| | - Shaoheng Li
- Jiangsu Key Laboratory of TCM Evaluation and Translational Research, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing 211198, China.
| | - Xiaohan Zou
- Jiangsu Key Laboratory of TCM Evaluation and Translational Research, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing 211198, China.
| | - Zhengyu Cao
- Jiangsu Key Laboratory of TCM Evaluation and Translational Research, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing 211198, China.
| |
Collapse
|
189
|
Gonçalves TC, Benoit E, Kurz M, Lucarain L, Fouconnier S, Combemale S, Jaquillard L, Schombert B, Chambard JM, Boukaiba R, Hessler G, Bohme A, Bialy L, Hourcade S, Béroud R, De Waard M, Servent D, Partiseti M. From identification to functional characterization of cyriotoxin-1a, an antinociceptive toxin from the spider Cyriopagopus schioedtei. Br J Pharmacol 2019; 176:1298-1314. [PMID: 30784059 DOI: 10.1111/bph.14628] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2018] [Revised: 01/24/2019] [Accepted: 01/31/2019] [Indexed: 12/18/2022] Open
Abstract
BACKGROUND AND PURPOSE The NaV 1.7 channel is highly expressed in dorsal root ganglia of the sensory nervous system and plays a central role in the pain signalling process. We investigated a library prepared from original venoms of 117 different animals to identify new selective inhibitors of this target. EXPERIMENTAL APPROACH We used high throughput screening of a large venom collection using automated patch-clamp experiments on human voltage-gated sodium channel subtypes and then in vitro and in vivo electrophysiological experiments to characterize the active peptides that have been purified, sequenced, and chemically synthesized. Analgesic effects were evaluated in vivo in mice models. KEY RESULTS We identified cyriotoxin-1a (CyrTx-1a), a novel peptide isolated from Cyriopagopus schioedtei spider venom, as a candidate for further characterization. This 33 amino acids toxin belongs to the inhibitor cystine knot structural family and inhibits hNaV 1.1-1.3 and 1.6-1.7 channels in the low nanomolar range, compared to the micromolar range for hNaV 1.4-1.5 and 1.8 channels. CyrTx-1a was 920 times more efficient at inhibiting tetrodotoxin (TTX)-sensitive than TTX-resistant sodium currents recorded from adult mouse dorsal root ganglia neurons and in vivo electrophysiological experiments showed that CyrTx-1a was approximately 170 times less efficient than huwentoxin-IV at altering mouse skeletal neuromuscular excitability properties. CyrTx-1a exhibited an analgesic effect in mice by increasing reaction time in the hot-plate assay. CONCLUSIONS AND IMPLICATIONS The pharmacological profile of CyrTx-1a paves the way for further molecular engineering aimed to optimize the potential antinociceptive properties of this peptide.
Collapse
Affiliation(s)
- Tânia C Gonçalves
- Integrated Drug Discovery-High Content Biology, Sanofi R&D, Vitry-sur-Seine, France.,Service d'Ingénierie Moléculaire des Protéines (SIMOPRO), CEA, Université Paris-Saclay, Gif-sur-Yvette, France
| | - Evelyne Benoit
- Service d'Ingénierie Moléculaire des Protéines (SIMOPRO), CEA, Université Paris-Saclay, Gif-sur-Yvette, France.,Institut des Neurosciences Paris-Saclay (Neuro-PSI), UMR CNRS/Université Paris-Sud 9197, Université Paris-Saclay, Gif-sur-Yvette, France
| | - Michael Kurz
- Integrated Drug Discovery-Synthetic Molecular Design, Sanofi R&D, Frankfurt, Germany
| | - Laetitia Lucarain
- Integrated Drug Discovery-High Content Biology, Sanofi R&D, Vitry-sur-Seine, France
| | - Sophie Fouconnier
- Integrated Drug Discovery-High Content Biology, Sanofi R&D, Vitry-sur-Seine, France
| | | | | | - Brigitte Schombert
- Integrated Drug Discovery-High Content Biology, Sanofi R&D, Vitry-sur-Seine, France
| | - Jean-Marie Chambard
- Integrated Drug Discovery-High Content Biology, Sanofi R&D, Vitry-sur-Seine, France
| | - Rachid Boukaiba
- Integrated Drug Discovery-High Content Biology, Sanofi R&D, Vitry-sur-Seine, France
| | - Gerhard Hessler
- Integrated Drug Discovery-Synthetic Molecular Design, Sanofi R&D, Frankfurt, Germany
| | - Andrees Bohme
- Integrated Drug Discovery-High Content Biology, Sanofi R&D, Vitry-sur-Seine, France
| | - Laurent Bialy
- Integrated Drug Discovery-Synthetic Molecular Design, Sanofi R&D, Frankfurt, Germany
| | - Stéphane Hourcade
- Neuroscience Therapeutic Area, Neurodegeneration Research, Sanofi R&D, Chilly-Mazarin, France
| | | | - Michel De Waard
- Smartox Biotechnology, Saint-Egrève, France.,Institut du Thorax, Inserm UMR 1087/CNRS UMR 6291, LabEx "Ion Channels, Science and Therapeutics", Nantes, France
| | - Denis Servent
- Service d'Ingénierie Moléculaire des Protéines (SIMOPRO), CEA, Université Paris-Saclay, Gif-sur-Yvette, France
| | - Michel Partiseti
- Integrated Drug Discovery-High Content Biology, Sanofi R&D, Vitry-sur-Seine, France
| |
Collapse
|
190
|
|
191
|
Abstract
Diabetic peripheral neuropathy (DPN) is a common disabling complication of diabetes. Almost half of the patients with DPN develop neuropathic pain (NeuP) for which current analgesic treatments are inadequate. Understanding the role of genetic variability in the development of painful DPN is needed for improved understanding of pain pathogenesis for better patient stratification in clinical trials and to target therapy more appropriately. Here, we examined the relationship between variants in the voltage-gated sodium channel NaV1.7 and NeuP in a deeply phenotyped cohort of patients with DPN. Although no rare variants were found in 78 participants with painless DPN, we identified 12 rare NaV1.7 variants in 10 (out of 111) study participants with painful DPN. Five of these variants had previously been described in the context of other NeuP disorders and 7 have not previously been linked to NeuP. Those patients with rare variants reported more severe pain and greater sensitivity to pressure stimuli on quantitative sensory testing. Electrophysiological characterization of 2 of the novel variants (M1852T and T1596I) demonstrated that gain of function changes as a consequence of markedly impaired channel fast inactivation. Using a structural model of NaV1.7, we were also able to provide further insight into the structural mechanisms underlying fast inactivation and the role of the C-terminal domain in this process. Our observations suggest that rare NaV1.7 variants contribute to the development NeuP in patients with DPN. Their identification should aid understanding of sensory phenotype, patient stratification, and help target treatments effectively.
Collapse
|
192
|
Li YF, Zheng YM, Yu Y, Gan Y, Gao ZB. Inhibitory effects of lappaconitine on the neuronal isoforms of voltage-gated sodium channels. Acta Pharmacol Sin 2019; 40:451-459. [PMID: 29991710 PMCID: PMC6461957 DOI: 10.1038/s41401-018-0067-x] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2018] [Accepted: 05/17/2018] [Indexed: 11/09/2022] Open
Abstract
Lappaconitine (LA) has been widely used for postoperative and cancer pain control. LA exhibits excellent analgesic activity with a longer effective time than common local anesthetics such as tetracaine and bupivacaine. However, the mechanisms underlying the featured analgesic activity of LA remain largely unknown. Here, we report that LA is an inhibitor of voltage-gated sodium channel 1.7 (Nav1.7) stably expressed in human embryonic kidney (HEK293) cells. LA inhibited Nav1.7 in a voltage-dependent manner with an IC50 value (with 95% confidence limits) of 27.67 (15.68-39.66) µmol/L when the cell was clamped at -70 mV. In comparison with the quick and reversible inhibition of Nav1.7 by tetracaine and bupivacaine, the inhibitory effect of LA was rather slow and irreversible. It took more than 10 min to achieve steady-state inhibition when LA (300 µmol/L) was administered. Unlike tetracaine and bupivacaine, LA affected neither the voltage-dependent activation nor the inactivation of the channels. Five residues in domain III and domain IV have been reported to be critical for the effects of the two local anesthetics on Nav channels. But our mutant study revealed that only two residues (F1737, N1742) located in domain IV were necessary for the inhibitory activity of LA. The slow onset, irreversibility, and lack of influence on channel activation and inactivation accompanied with the different molecular determinants suggest that LA may inhibit Nav1.7 channels in a manner different from local anesthetics. These results may help to understand the featured analgesic activity of LA, thus benefiting its application in the clinic and future drug development.
Collapse
Affiliation(s)
- Yan-Fen Li
- Shanghai Key Laboratory of Bio-Energy Crops, School of Life Sciences, Shanghai University, Shanghai, 200444, China
- CAS Key Laboratory of Receptor Research, State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China
| | - Yue-Ming Zheng
- CAS Key Laboratory of Receptor Research, State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China
- University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Yong Yu
- Department of Neurosurgery, Zhongshan Hospital, Fudan University, Shanghai, 200032, China
| | - Yong Gan
- CAS Key Laboratory of Receptor Research, State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China.
- University of Chinese Academy of Sciences, Beijing, 100049, China.
| | - Zhao-Bing Gao
- CAS Key Laboratory of Receptor Research, State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China.
- University of Chinese Academy of Sciences, Beijing, 100049, China.
| |
Collapse
|
193
|
Antisense oligonucleotides selectively suppress target RNA in nociceptive neurons of the pain system and can ameliorate mechanical pain. Pain 2019; 159:139-149. [PMID: 28976422 DOI: 10.1097/j.pain.0000000000001074] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
There is an urgent need for better treatments for chronic pain, which affects more than 1 billion people worldwide. Antisense oligonucleotides (ASOs) have proven successful in treating children with spinal muscular atrophy, a severe infantile neurological disorder, and several ASOs are currently being tested in clinical trials for various neurological disorders. Here, we characterize the pharmacodynamic activity of ASOs in spinal cord and dorsal root ganglia (DRG), key tissues for pain signaling. We demonstrate that activity of ASOs lasts up to 2 months after a single intrathecal bolus dose. Interestingly, comparison of subcutaneous, intracerebroventricular, and intrathecal administration shows that DRGs are targetable by systemic and central delivery of ASOs, while target reduction in the spinal cord is achieved only after direct central delivery. Upon detailed characterization of ASO activity in individual cell populations in DRG, we observe robust target suppression in all neuronal populations, thereby establishing that ASOs are effective in the cell populations involved in pain propagation. Furthermore, we confirm that ASOs are selective and do not modulate basal pain sensation. We also demonstrate that ASOs targeting the sodium channel Nav1.7 induce sustained analgesia up to 4 weeks. Taken together, our findings support the idea that ASOs possess the required pharmacodynamic properties, along with a long duration of action beneficial for treating pain.
Collapse
|
194
|
McDermott LA, Weir GA, Themistocleous AC, Segerdahl AR, Blesneac I, Baskozos G, Clark AJ, Millar V, Peck LJ, Ebner D, Tracey I, Serra J, Bennett DL. Defining the Functional Role of Na V1.7 in Human Nociception. Neuron 2019; 101:905-919.e8. [PMID: 30795902 PMCID: PMC6424805 DOI: 10.1016/j.neuron.2019.01.047] [Citation(s) in RCA: 125] [Impact Index Per Article: 20.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2018] [Revised: 12/03/2018] [Accepted: 01/18/2019] [Indexed: 12/17/2022]
Abstract
Loss-of-function mutations in NaV1.7 cause congenital insensitivity to pain (CIP); this voltage-gated sodium channel is therefore a key target for analgesic drug development. Utilizing a multi-modal approach, we investigated how NaV1.7 mutations lead to human pain insensitivity. Skin biopsy and microneurography revealed an absence of C-fiber nociceptors in CIP patients, reflected in a reduced cortical response to capsaicin on fMRI. Epitope tagging of endogenous NaV1.7 revealed the channel to be localized at the soma membrane, axon, axon terminals, and the nodes of Ranvier of induced pluripotent stem cell (iPSC) nociceptors. CIP patient-derived iPSC nociceptors exhibited an inability to properly respond to depolarizing stimuli, demonstrating that NaV1.7 is a key regulator of excitability. Using this iPSC nociceptor platform, we found that some NaV1.7 blockers undergoing clinical trials lack specificity. CIP, therefore, arises due to a profound loss of functional nociceptors, which is more pronounced than that reported in rodent models, or likely achievable following acute pharmacological blockade. VIDEO ABSTRACT.
Collapse
Affiliation(s)
- Lucy A McDermott
- Nuffield Department of Clinical Neurosciences, University of Oxford, Oxford OX3 9DU, UK
| | - Greg A Weir
- Nuffield Department of Clinical Neurosciences, University of Oxford, Oxford OX3 9DU, UK
| | | | - Andrew R Segerdahl
- Nuffield Department of Clinical Neurosciences, University of Oxford, Oxford OX3 9DU, UK; Wellcome Centre for Integrative Neuroimaging, FMRIB, Nuffield Department of Clinical Neurosciences, University of Oxford, Oxford OX3 9DU, UK
| | - Iulia Blesneac
- Nuffield Department of Clinical Neurosciences, University of Oxford, Oxford OX3 9DU, UK
| | - Georgios Baskozos
- Nuffield Department of Clinical Neurosciences, University of Oxford, Oxford OX3 9DU, UK
| | - Alex J Clark
- Nuffield Department of Clinical Neurosciences, University of Oxford, Oxford OX3 9DU, UK
| | - Val Millar
- Target Discovery Institute, Nuffield Department of Medicine, University of Oxford, Oxford OX3 7FZ, UK
| | - Liam J Peck
- Nuffield Department of Clinical Neurosciences, University of Oxford, Oxford OX3 9DU, UK
| | - Daniel Ebner
- Target Discovery Institute, Nuffield Department of Medicine, University of Oxford, Oxford OX3 7FZ, UK
| | - Irene Tracey
- Nuffield Department of Clinical Neurosciences, University of Oxford, Oxford OX3 9DU, UK; Wellcome Centre for Integrative Neuroimaging, FMRIB, Nuffield Department of Clinical Neurosciences, University of Oxford, Oxford OX3 9DU, UK
| | - Jordi Serra
- Department of Clinical Neurophysiology, King's College Hospital, London SE5 9RS, UK
| | - David L Bennett
- Nuffield Department of Clinical Neurosciences, University of Oxford, Oxford OX3 9DU, UK.
| |
Collapse
|
195
|
Sun JF, Xu YJ, Kong XH, Su Y, Wang ZY. Fenamates inhibit human sodium channel Nav1.7 and Nav1.8. Neurosci Lett 2019; 696:67-73. [DOI: 10.1016/j.neulet.2018.12.008] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2018] [Revised: 11/06/2018] [Accepted: 12/04/2018] [Indexed: 11/27/2022]
|
196
|
Coates MD, Vrana KE, Ruiz-Velasco V. The influence of voltage-gated sodium channels on human gastrointestinal nociception. Neurogastroenterol Motil 2019; 31:e13460. [PMID: 30216585 DOI: 10.1111/nmo.13460] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/13/2018] [Revised: 08/01/2018] [Accepted: 08/07/2018] [Indexed: 12/14/2022]
Abstract
BACKGROUND Abdominal pain is a frequent and persistent problem in the most common gastrointestinal disorders, including irritable bowel syndrome and inflammatory bowel disease. Pain adversely impacts quality of life, incurs significant healthcare expenditures, and remains a challenging issue to manage with few safe therapeutic options currently available. It is imperative that new methods are developed for identifying and treating this symptom. A variety of peripherally active neuroendocrine signaling elements have the capability to influence gastrointestinal pain perception. A large and growing body of evidence suggests that voltage-gated sodium channels (VGSCs) play a critical role in the development and modulation of nociceptive signaling associated with the gut. Several VGSC isoforms demonstrate significant promise as potential targets for improved diagnosis and treatment of gut-based disorders associated with hyper- and hyposensitivity to abdominal pain. PURPOSE In this article, we critically review key investigations that have evaluated the potential role that VGSCs play in visceral nociception and discuss recent advances related to this topic. Specifically, we discuss the following: (a) what is known about the structure and basic function of VGSCs, (b) the role that each VGSC plays in gut nociception, particularly as it relates to human physiology, and (c) potential diagnostic and therapeutic uses of VGSCs to manage disorders associated with chronic abdominal pain.
Collapse
Affiliation(s)
- Matthew D Coates
- Division of Gastroenterology & Hepatology, Department of Medicine, Penn State University College of Medicine, Hershey, Pennsylvania
| | - Kent E Vrana
- Department of Pharmacology, Penn State University College of Medicine, Hershey, Pennsylvania
| | - Victor Ruiz-Velasco
- Department of Anesthesiology and Perioperative Medicine, Penn State University College of Medicine, Hershey, Pennsylvania
| |
Collapse
|
197
|
Chew LA, Bellampalli SS, Dustrude ET, Khanna R. Mining the Na v1.7 interactome: Opportunities for chronic pain therapeutics. Biochem Pharmacol 2019; 163:9-20. [PMID: 30699328 DOI: 10.1016/j.bcp.2019.01.018] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2019] [Accepted: 01/24/2019] [Indexed: 12/14/2022]
Abstract
The peripherally expressed voltage-gated sodium NaV1.7 (gene SCN9A) channel boosts small stimuli to initiate firing of pain-signaling dorsal root ganglia (DRG) neurons and facilitates neurotransmitter release at the first synapse within the spinal cord. Mutations in SCN9A produce distinct human pain syndromes. Widely acknowledged as a "gatekeeper" of pain, NaV1.7 has been the focus of intense investigation but, to date, no NaV1.7-selective drugs have reached the clinic. Elegant crystallographic studies have demonstrated the potential of designing highly potent and selective NaV1.7 compounds but their therapeutic value remains untested. Transcriptional silencing of NaV1.7 by a naturally expressed antisense transcript has been reported in rodents and humans but whether this represents a viable opportunity for designing NaV1.7 therapeutics is currently unknown. The demonstration that loss of NaV1.7 function is associated with upregulation of endogenous opioids and potentiation of mu- and delta-opioid receptor activities, suggests that targeting only NaV1.7 may be insufficient for analgesia. However, the link between opioid-dependent analgesic mechanisms and function of sodium channels and intracellular sodium-dependent signaling remains controversial. Thus, additional new targets - regulators, modulators - are needed. In this context, we mine the literature for the known interactome of NaV1.7 with a focus on protein interactors that affect the channel's trafficking or link it to opioid signaling. As a case study, we present antinociceptive evidence of allosteric regulation of NaV1.7 by the cytosolic collapsin response mediator protein 2 (CRMP2). Throughout discussions of these possible new targets, we offer thoughts on the therapeutic implications of modulating NaV1.7 function in chronic pain.
Collapse
Affiliation(s)
- Lindsey A Chew
- Department of Pharmacology, College of Medicine, University of Arizona, Tucson, AZ 85724, USA
| | - Shreya S Bellampalli
- Department of Pharmacology, College of Medicine, University of Arizona, Tucson, AZ 85724, USA
| | - Erik T Dustrude
- Stark Neurosciences Research Institute, Indiana University School of Medicine, Indianapolis, IN, USA; Department of Psychiatry, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Rajesh Khanna
- Department of Pharmacology, College of Medicine, University of Arizona, Tucson, AZ 85724, USA; Graduate Interdisciplinary Program in Neuroscience, College of Medicine, University of Arizona, Tucson, AZ, USA; The Center for Innovation in Brain Sciences, The University of Arizona Health Sciences, Tucson, AZ 85724, USA.
| |
Collapse
|
198
|
Luo G, Chen L, Easton A, Newton A, Bourin C, Shields E, Mosure K, Soars MG, Knox RJ, Matchett M, Pieschl RL, Post-Munson DJ, Wang S, Herrington J, Graef J, Newberry K, Sivarao DV, Senapati A, Bristow LJ, Meanwell NA, Thompson LA, Dzierba C. Discovery of Indole- and Indazole-acylsulfonamides as Potent and Selective Na V1.7 Inhibitors for the Treatment of Pain. J Med Chem 2019; 62:831-856. [PMID: 30576602 DOI: 10.1021/acs.jmedchem.8b01550] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
3-Aryl-indole and 3-aryl-indazole derivatives were identified as potent and selective Nav1.7 inhibitors. Compound 29 was shown to be efficacious in the mouse formalin assay and also reduced complete Freund's adjuvant (CFA)-induced thermal hyperalgesia and chronic constriction injury (CCI) induced cold allodynia and models of inflammatory and neuropathic pain, respectively, following intraperitoneal (IP) doses of 30 mg/kg. The observed efficacy could be correlated with the mouse dorsal root ganglion exposure and NaV1.7 potency associated with 29.
Collapse
Affiliation(s)
- Guanglin Luo
- Bristol-Myers Squibb Research and Development , PO Box 4000, Princeton , New Jersey 08543-4000 , United States
| | - Ling Chen
- Bristol-Myers Squibb Research and Development , PO Box 4000, Princeton , New Jersey 08543-4000 , United States
| | - Amy Easton
- Bristol-Myers Squibb Research and Development , PO Box 4000, Princeton , New Jersey 08543-4000 , United States
| | - Amy Newton
- Bristol-Myers Squibb Research and Development , PO Box 4000, Princeton , New Jersey 08543-4000 , United States
| | - Clotilde Bourin
- Bristol-Myers Squibb Research and Development , PO Box 4000, Princeton , New Jersey 08543-4000 , United States
| | - Eric Shields
- Bristol-Myers Squibb Research and Development , PO Box 4000, Princeton , New Jersey 08543-4000 , United States
| | - Kathy Mosure
- Bristol-Myers Squibb Research and Development , PO Box 4000, Princeton , New Jersey 08543-4000 , United States
| | - Matthew G Soars
- Bristol-Myers Squibb Research and Development , PO Box 4000, Princeton , New Jersey 08543-4000 , United States
| | - Ronald J Knox
- Bristol-Myers Squibb Research and Development , PO Box 4000, Princeton , New Jersey 08543-4000 , United States
| | - Michele Matchett
- Bristol-Myers Squibb Research and Development , PO Box 4000, Princeton , New Jersey 08543-4000 , United States
| | - Rick L Pieschl
- Bristol-Myers Squibb Research and Development , PO Box 4000, Princeton , New Jersey 08543-4000 , United States
| | - Debra J Post-Munson
- Bristol-Myers Squibb Research and Development , PO Box 4000, Princeton , New Jersey 08543-4000 , United States
| | - Shuya Wang
- Bristol-Myers Squibb Research and Development , PO Box 4000, Princeton , New Jersey 08543-4000 , United States
| | - James Herrington
- Bristol-Myers Squibb Research and Development , PO Box 4000, Princeton , New Jersey 08543-4000 , United States
| | - John Graef
- Bristol-Myers Squibb Research and Development , PO Box 4000, Princeton , New Jersey 08543-4000 , United States
| | - Kimberly Newberry
- Bristol-Myers Squibb Research and Development , PO Box 4000, Princeton , New Jersey 08543-4000 , United States
| | - Digavalli V Sivarao
- Bristol-Myers Squibb Research and Development , PO Box 4000, Princeton , New Jersey 08543-4000 , United States
| | - Arun Senapati
- Bristol-Myers Squibb Research and Development , PO Box 4000, Princeton , New Jersey 08543-4000 , United States
| | - Linda J Bristow
- Bristol-Myers Squibb Research and Development , PO Box 4000, Princeton , New Jersey 08543-4000 , United States
| | - Nicholas A Meanwell
- Bristol-Myers Squibb Research and Development , PO Box 4000, Princeton , New Jersey 08543-4000 , United States
| | - Lorin A Thompson
- Bristol-Myers Squibb Research and Development , PO Box 4000, Princeton , New Jersey 08543-4000 , United States
| | - Carolyn Dzierba
- Bristol-Myers Squibb Research and Development , PO Box 4000, Princeton , New Jersey 08543-4000 , United States
| |
Collapse
|
199
|
|
200
|
Wu YJ, Venables B, Guernon J, Chen J, Sit SY, Rajamani R, Knox RJ, Matchett M, Pieschl RL, Herrington J, Bristow LJ, Meanwell NA, Thompson LA, Dzierba C. Discovery of new indole-based acylsulfonamide Na v1.7 inhibitors. Bioorg Med Chem Lett 2018; 29:659-663. [PMID: 30638874 DOI: 10.1016/j.bmcl.2018.12.013] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2018] [Revised: 12/03/2018] [Accepted: 12/05/2018] [Indexed: 01/05/2023]
Abstract
Screening of 100 acylsulfonamides from the Bristol-Myers Squibb compound collection identified the C3-cyclohexyl indole 6 as a potent Nav1.7 inhibitor. Replacement of the C2 furanyl ring of 6 with a heteroaryl moiety or truncation of this group led to the identification of 4 analogs with hNav1.7 IC50 values under 50 nM. Fluorine substitution of the truncated compound 12 led to 34 with improved potency and isoform selectivity. The inverted indole 36 also maintained good activity. Both 34 and 36 exhibited favorable CYP inhibition profiles, good membrane permeability and a low efflux ratio and, therefore, represent new leads in the search for potent and selective Nav1.7 inhibitors to treat pain.
Collapse
Affiliation(s)
- Yong-Jin Wu
- Bristol-Myers Squibb Research and Development, 5 Research Parkway, Wallingford, CT 06492-7660, USA.
| | - Brian Venables
- Bristol-Myers Squibb Research and Development, 5 Research Parkway, Wallingford, CT 06492-7660, USA
| | - Jason Guernon
- Bristol-Myers Squibb Research and Development, 5 Research Parkway, Wallingford, CT 06492-7660, USA
| | - Jie Chen
- Bristol-Myers Squibb Research and Development, 5 Research Parkway, Wallingford, CT 06492-7660, USA
| | - Sing-Yuen Sit
- Bristol-Myers Squibb Research and Development, 5 Research Parkway, Wallingford, CT 06492-7660, USA
| | - Ramkumar Rajamani
- Bristol-Myers Squibb Research and Development, 5 Research Parkway, Wallingford, CT 06492-7660, USA
| | - Ronald J Knox
- Bristol-Myers Squibb Research and Development, 5 Research Parkway, Wallingford, CT 06492-7660, USA
| | - Michele Matchett
- Bristol-Myers Squibb Research and Development, 5 Research Parkway, Wallingford, CT 06492-7660, USA
| | - Rick L Pieschl
- Bristol-Myers Squibb Research and Development, 5 Research Parkway, Wallingford, CT 06492-7660, USA
| | - James Herrington
- Bristol-Myers Squibb Research and Development, 5 Research Parkway, Wallingford, CT 06492-7660, USA
| | - Linda J Bristow
- Bristol-Myers Squibb Research and Development, 5 Research Parkway, Wallingford, CT 06492-7660, USA
| | - Nicholas A Meanwell
- Bristol-Myers Squibb Research and Development, 5 Research Parkway, Wallingford, CT 06492-7660, USA
| | - Lorin A Thompson
- Bristol-Myers Squibb Research and Development, 5 Research Parkway, Wallingford, CT 06492-7660, USA
| | - Carolyn Dzierba
- Bristol-Myers Squibb Research and Development, 5 Research Parkway, Wallingford, CT 06492-7660, USA
| |
Collapse
|