51
|
A Buthus martensii Karsch scorpion sting targets Nav1.7 in mice and mimics a phenotype of human chronic pain. Pain 2021; 163:e202-e214. [PMID: 34252912 DOI: 10.1097/j.pain.0000000000002397] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2021] [Accepted: 06/28/2021] [Indexed: 11/25/2022]
Abstract
GAIN and loss-of-function mutations in Nav1.7 cause chronic pain and pain insensitivity, respectively. The preferential expression of Nav1.7 in peripheral nervous system and its role in human pain signaling make Nav1.7 a promising target for next-generation pain therapeutics. However, pharmacological agents have not fully recapitulated these pain phenotypes, and, due to the lack of subtype-selective molecular modulators, the role of Nav1.7 in the perception of pain remains poorly understood. Scorpion venom is an excellent source of bioactive peptides that modulate various ion channels, including voltage-gated sodium (Nav) channels . Here, we demonstrate that Buthus martensii Karsch scorpion venom (BV) elicits pain responses in mice through direct enhancement of Nav1.7 activity, and have identified that Makatoxin-3, an α-like toxin as a critical component for BV-mediated effects on Nav1.7. Blocking other Nav subtypes did not eliminate BV-evoked pain responses, supporting the pivotal role of Nav1.7 in BV-induced pain . Makatoxin-3 acts on the S3-S4 loop of voltage sensor domain IV (VSD4) of Nav1.7, which causes a hyperpolarizing shift in the steady-state fast inactivation and impairs inactivation kinetics. We also determined the key residues and structure-function relationships for the toxin-channel interactions, which are distinct from those of other well-studied α-toxins. This study not only reveals a new mechanism underlying BV-evoked pain, but also enriches our knowledge of key structural elements of scorpion toxins that are pivotal for toxin-Nav1.7 interaction, which facilitates the design of novel Nav1.7 selective modulators.
Collapse
|
52
|
Sloan G, Selvarajah D, Tesfaye S. Pathogenesis, diagnosis and clinical management of diabetic sensorimotor peripheral neuropathy. Nat Rev Endocrinol 2021; 17:400-420. [PMID: 34050323 DOI: 10.1038/s41574-021-00496-z] [Citation(s) in RCA: 222] [Impact Index Per Article: 55.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 04/15/2021] [Indexed: 02/08/2023]
Abstract
Diabetic sensorimotor peripheral neuropathy (DSPN) is a serious complication of diabetes mellitus and is associated with increased mortality, lower-limb amputations and distressing painful neuropathic symptoms (painful DSPN). Our understanding of the pathophysiology of the disease has largely been derived from animal models, which have identified key potential mechanisms. However, effective therapies in preclinical models have not translated into clinical trials and we have no universally accepted disease-modifying treatments. Moreover, the condition is generally diagnosed late when irreversible nerve damage has already taken place. Innovative point-of-care devices have great potential to enable the early diagnosis of DSPN when the condition might be more amenable to treatment. The management of painful DSPN remains less than optimal; however, studies suggest that a mechanism-based approach might offer an enhanced benefit in certain pain phenotypes. The management of patients with DSPN involves the control of individualized cardiometabolic targets, a multidisciplinary approach aimed at the prevention and management of foot complications, and the timely diagnosis and management of neuropathic pain. Here, we discuss the latest advances in the mechanisms of DSPN and painful DSPN, originating both from the periphery and the central nervous system, as well as the emerging diagnostics and treatments.
Collapse
Affiliation(s)
- Gordon Sloan
- Diabetes Research Unit, Royal Hallamshire Hospital, Sheffield Teaching Hospitals NHS Foundation Trust, Sheffield, UK
| | - Dinesh Selvarajah
- Diabetes Research Unit, Royal Hallamshire Hospital, Sheffield Teaching Hospitals NHS Foundation Trust, Sheffield, UK
- Department of Oncology and Human Metabolism, University of Sheffield, Sheffield, UK
| | - Solomon Tesfaye
- Diabetes Research Unit, Royal Hallamshire Hospital, Sheffield Teaching Hospitals NHS Foundation Trust, Sheffield, UK.
| |
Collapse
|
53
|
Differences in the antinociceptive effects of serotonin-noradrenaline reuptake inhibitors via sodium channel blockade using the veratrine test in mice. Neuroreport 2021; 32:797-802. [PMID: 33994525 DOI: 10.1097/wnr.0000000000001658] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
Antidepressants exert their analgesic effects by inhibiting the reuptake of noradrenaline. Several antidepressants have been shown to block the sodium channels, which might contribute to their analgesic potency. The aim of this study was to determine whether serotonin-noradrenaline reuptake inhibitors (SNRIs) could produce antinociceptive effects via sodium channel blockade using the veratrine test in mice. Furthermore, the effects of these agents on the veratrine test were examined to elucidate the effects of several antidepressants and tramadol on sodium channels. The administration of duloxetine (10 mg/kg) and venlafaxine (30 mg/kg) suppressed cuff-induced mechanical allodynia; however, these antinociceptive effects were only partially suppressed by atipamezole. Furthermore, duloxetine and venlafaxine demonstrated antinociceptive effects via sodium channel blockade, as assayed by the veratrine test. In addition, several antidepressants, including amitriptyline, paroxetine and mirtazapine, reduced veratrine-induced nociception. In contrast, milnacipran and tramadol did not alter the veratrine-induced nociception. These results indicated that, in addition to the primary action of SNRIs on monoamine transporters, sodium channel blockade might be involved in the antinociceptive activities of duloxetine and venlafaxine, but not milnacipran.
Collapse
|
54
|
Mehboob R, Marchenkova A, van den Maagdenberg AMJM, Nistri A. Overexpressed Na V 1.7 Channels Confer Hyperexcitability to in vitro Trigeminal Sensory Neurons of Ca V 2.1 Mutant Hemiplegic Migraine Mice. Front Cell Neurosci 2021; 15:640709. [PMID: 34113237 PMCID: PMC8185157 DOI: 10.3389/fncel.2021.640709] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2020] [Accepted: 04/09/2021] [Indexed: 11/21/2022] Open
Abstract
Trigeminal sensory neurons of transgenic knock-in (KI) mice expressing the R192Q missense mutation in the α1A subunit of neuronal voltage-gated CaV2.1 Ca2+ channels, which leads to familial hemiplegic migraine type 1 (FHM1) in patients, exhibit a hyperexcitability phenotype. Here, we show that the expression of NaV1.7 channels, linked to pain states, is upregulated in KI primary cultures of trigeminal ganglia (TG), as shown by increased expression of its α1 subunit. In the majority of TG neurons, NaV1.7 channels are co-expressed with ATP-gated P2X3 receptors (P2X3R), which are important nociceptive sensors. Reversing the trigeminal phenotype with selective CaV2.1 channel inhibitor ω-agatoxin IVA inhibited NaV1.7 overexpression. Functionally, KI neurons revealed a TTX-sensitive inward current of larger amplitude that was partially inhibited by selective NaV1.7 blocker Tp1a. Under current-clamp condition, Tp1a raised the spike threshold of both wild-type (WT) and KI neurons with decreased firing rate in KI cells. NaV1.7 activator OD1 accelerated firing in WT and KI neurons, a phenomenon blocked by Tp1a. Enhanced expression and function of NaV1.7 channels in KI TG neurons resulted in higher excitability and facilitated nociceptive signaling. Co-expression of NaV1.7 channels and P2X3Rs in TGs may explain how hypersensitivity to local stimuli can be relevant to migraine.
Collapse
Affiliation(s)
- Riffat Mehboob
- Department of Neuroscience, International School for Advanced Studies (SISSA), Trieste, Italy.,Research Unit, Faculty of Allied Health Sciences, University of Lahore, Lahore, Pakistan
| | - Anna Marchenkova
- Department of Neuroscience, International School for Advanced Studies (SISSA), Trieste, Italy
| | - Arn M J M van den Maagdenberg
- Department of Neurology, Leiden University Medical Center, Leiden, Netherlands.,Department of Human Genetics, University Medical Center, Leiden, Netherlands
| | - Andrea Nistri
- Department of Neuroscience, International School for Advanced Studies (SISSA), Trieste, Italy
| |
Collapse
|
55
|
MacDonald DI, Sikandar S, Weiss J, Pyrski M, Luiz AP, Millet Q, Emery EC, Mancini F, Iannetti GD, Alles SRA, Arcangeletti M, Zhao J, Cox JJ, Brownstone RM, Zufall F, Wood JN. A central mechanism of analgesia in mice and humans lacking the sodium channel Na V1.7. Neuron 2021; 109:1497-1512.e6. [PMID: 33823138 PMCID: PMC8110947 DOI: 10.1016/j.neuron.2021.03.012] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2019] [Revised: 02/05/2020] [Accepted: 03/08/2021] [Indexed: 11/18/2022]
Abstract
Deletion of SCN9A encoding the voltage-gated sodium channel NaV1.7 in humans leads to profound pain insensitivity and anosmia. Conditional deletion of NaV1.7 in sensory neurons of mice also abolishes pain, suggesting that the locus of analgesia is the nociceptor. Here we demonstrate, using in vivo calcium imaging and extracellular recording, that NaV1.7 knockout mice have essentially normal nociceptor activity. However, synaptic transmission from nociceptor central terminals in the spinal cord is greatly reduced by an opioid-dependent mechanism. Analgesia is also reversed substantially by central but not peripheral application of opioid antagonists. In contrast, the lack of neurotransmitter release from olfactory sensory neurons is opioid independent. Male and female humans with NaV1.7-null mutations show naloxone-reversible analgesia. Thus, inhibition of neurotransmitter release is the principal mechanism of anosmia and analgesia in mouse and human Nav1.7-null mutants.
Collapse
Affiliation(s)
- Donald Iain MacDonald
- Molecular Nociception Group, Wolfson Institute for Biomedical Research, University College London, Gower Street, London WC1E 6BT, UK.
| | - Shafaq Sikandar
- Centre for Experimental Medicine & Rheumatology, Queen Mary University of London, Charterhouse Square, London EC1M 6BQ, UK
| | - Jan Weiss
- Center for Integrative Physiology and Molecular Medicine, Saarland University, 66421 Homburg, Germany
| | - Martina Pyrski
- Center for Integrative Physiology and Molecular Medicine, Saarland University, 66421 Homburg, Germany
| | - Ana P Luiz
- Molecular Nociception Group, Wolfson Institute for Biomedical Research, University College London, Gower Street, London WC1E 6BT, UK
| | - Queensta Millet
- Molecular Nociception Group, Wolfson Institute for Biomedical Research, University College London, Gower Street, London WC1E 6BT, UK
| | - Edward C Emery
- Molecular Nociception Group, Wolfson Institute for Biomedical Research, University College London, Gower Street, London WC1E 6BT, UK
| | - Flavia Mancini
- Department of Neuroscience, Physiology and Pharmacology, University College London, Gower Street, London WC1E 6BT, UK
| | - Gian D Iannetti
- Department of Neuroscience, Physiology and Pharmacology, University College London, Gower Street, London WC1E 6BT, UK; Neuroscience and Behaviour Laboratory, Istituto Italiano di Tecnologia, Rome, Italy
| | - Sascha R A Alles
- Molecular Nociception Group, Wolfson Institute for Biomedical Research, University College London, Gower Street, London WC1E 6BT, UK
| | - Manuel Arcangeletti
- Molecular Nociception Group, Wolfson Institute for Biomedical Research, University College London, Gower Street, London WC1E 6BT, UK
| | - Jing Zhao
- Molecular Nociception Group, Wolfson Institute for Biomedical Research, University College London, Gower Street, London WC1E 6BT, UK
| | - James J Cox
- Molecular Nociception Group, Wolfson Institute for Biomedical Research, University College London, Gower Street, London WC1E 6BT, UK
| | | | - Frank Zufall
- Center for Integrative Physiology and Molecular Medicine, Saarland University, 66421 Homburg, Germany
| | - John N Wood
- Molecular Nociception Group, Wolfson Institute for Biomedical Research, University College London, Gower Street, London WC1E 6BT, UK.
| |
Collapse
|
56
|
Shen H, Yan N, Pan X. Structural determination of human Na v1.4 and Na v1.7 using single particle cryo-electron microscopy. Methods Enzymol 2021; 653:103-120. [PMID: 34099168 DOI: 10.1016/bs.mie.2021.03.010] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
Voltage-gated sodium (Nav) channels are responsible for the initiation and propagation of action potentials. Their abnormal functions are associated with numerous diseases, such as epilepsy, cardiac arrhythmia, and pain syndromes. Therefore, these channels represent important drug targets. Even in the post-resolution revolution era, a lack of structural information continues to impede structure-based drug discovery. The limiting factor for the structural determination of Nav channels using single particle cryo-electron microscopy (cryo-EM) resides in the generation of sufficient high-quality recombinant proteins. After extensive trials, we have been successful in determining a series of high-resolution structures of Nav channels, including NavPaS from American cockroach, Nav1.4 from electric eel, and human Nav1.1, Nav1.2, Nav1.4, Nav1.5, and Nav1.7, with distinct strategies. These structures established the framework for understanding the electromechanical coupling and disease mechanism of Nav channels, and for facilitating drug discovery. Here, we exemplify these methods with two specific cases, human Nav1.4 and Nav1.7, which may shed light on the structural determination of other membrane proteins.
Collapse
Affiliation(s)
- Huaizong Shen
- Key Laboratory of Structural Biology of Zhejiang Province, School of Life Sciences, Westlake University, Hangzhou, Zhejiang, China; Westlake Laboratory of Life Sciences and Biomedicine, Hangzhou, Zhejiang, China; Institute of Biology, Westlake Institute for Advanced Study, Hangzhou, Zhejiang, China.
| | - Nieng Yan
- Department of Molecular Biology, Princeton University, Princeton, NJ, United States.
| | - Xiaojing Pan
- State Key Laboratory of Membrane Biology, Beijing Advanced Innovation Center for Structural Biology, Tsinghua-Peking Joint Center for Life Sciences, School of Life Sciences, Tsinghua University, Beijing, China.
| |
Collapse
|
57
|
Mohammed ZA, Kaloyanova K, Nassar MA. An unbiased and efficient assessment of excitability of sensory neurons for analgesic drug discovery. Pain 2021; 161:1100-1108. [PMID: 31929383 PMCID: PMC7170445 DOI: 10.1097/j.pain.0000000000001802] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
Distinct veratridine-induced calcium responses in nociceptors and non-nociceptors allow for unbiased and efficient assessment of drugs' action on both populations separately but simultaneously. Alleviating chronic pain is challenging, due to lack of drugs that effectively inhibit nociceptors without off-target effects on motor or central neurons. Dorsal root ganglia (DRG) contain nociceptive and non-nociceptive neurons. Drug screening on cultured DRG neurons, rather than cell lines, allows for the identification of drugs most potent on nociceptors with no effects on non-nociceptors (as a proxy for unwanted side effects on central nervous system and motor neurons). However, screening using DRG neurons is currently a low-throughput process, and there is a need for assays to speed this process for analgesic drug discovery. We previously showed that veratridine elicits distinct response profiles in sensory neurons. Here, we show evidence that a veratridine-based calcium assay allows for an unbiased and efficient assessment of a drug effect on nociceptors (targeted neurons) and non-nociceptors (nontargeted neurons). We confirmed the link between the oscillatory profile and nociceptors, and the slow-decay profile and non-nociceptors using 3 transgenic mouse lines of known pain phenotypes. We used the assay to show that blockers for Nav1.7 and Nav1.8 channels, which are validated targets for analgesics, affect non-nociceptors at concentrations needed to effectively inhibit nociceptors. However, a combination of low doses of both blockers had an additive effect on nociceptors without a significant effect on non-nociceptors, indicating that the assay can also be used to screen for combinations of existing or novel drugs for the greatest selective inhibition of nociceptors.
Collapse
Affiliation(s)
- Zainab A Mohammed
- Department of Biomedical Science, University of Sheffield, Sheffield, United Kingdom
| | | | | |
Collapse
|
58
|
Raja SN, Ringkamp M, Guan Y, Campbell JN. John J. Bonica Award Lecture: Peripheral neuronal hyperexcitability: the "low-hanging" target for safe therapeutic strategies in neuropathic pain. Pain 2021; 161 Suppl 1:S14-S26. [PMID: 33090736 DOI: 10.1097/j.pain.0000000000001838] [Citation(s) in RCA: 42] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Affiliation(s)
| | - Matthias Ringkamp
- Neurological Surgery, Department of Neurosurgery, Johns Hopkins University, School of Medicine, Baltimore, MD, United States
| | - Yun Guan
- Departments of Anesthesiology and Critical Care Medicine and.,Neurological Surgery, Department of Neurosurgery, Johns Hopkins University, School of Medicine, Baltimore, MD, United States
| | - James N Campbell
- Neurological Surgery, Department of Neurosurgery, Johns Hopkins University, School of Medicine, Baltimore, MD, United States
| |
Collapse
|
59
|
Malfait AM, Miller RE, Miller RJ. Basic Mechanisms of Pain in Osteoarthritis: Experimental Observations and New Perspectives. Rheum Dis Clin North Am 2021; 47:165-180. [PMID: 33781488 DOI: 10.1016/j.rdc.2020.12.002] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
The specific changes in the peripheral neuronal pathways underlying joint pain in osteoarthritis are the focus of this review. The plasticity of the nociceptive system in osteoarthritis and how this involves changes in the structural, physiologic, and genetic properties of neurons in pain pathways are discussed. The role of the neurotrophin, nerve growth factor, in these pathogenic processes is discussed. Finally, how neuronal pathways are modified by interaction with the degenerating joint tissues they innervate and with the innate immune system is considered. These extensive cellular interactions provide a substrate for identification of targets for osteoarthritis pain.
Collapse
Affiliation(s)
- Anne-Marie Malfait
- Department of Internal Medicine, Division of Rheumatology, Rush University Medical Center, Room 714, 1735 W Harrison Street, Chicago, IL 60612, USA.
| | - Rachel E Miller
- Department of Internal Medicine, Division of Rheumatology, Rush University Medical Center, Room 714, 1735 W Harrison Street, Chicago, IL 60612, USA
| | - Richard J Miller
- Department of Pharmacology, Northwestern University, Searle Building Room 8-510, 320 E Superior Street, Chicago, IL 60611, USA
| |
Collapse
|
60
|
Studies on CRMP2 SUMOylation-deficient transgenic mice identify sex-specific Nav1.7 regulation in the pathogenesis of chronic neuropathic pain. Pain 2021; 161:2629-2651. [PMID: 32569093 DOI: 10.1097/j.pain.0000000000001951] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
The sodium channel Nav1.7 is a master regulator of nociceptive input into the central nervous system. Mutations in this channel can result in painful conditions and produce insensitivity to pain. Despite being recognized as a "poster child" for nociceptive signaling and human pain, targeting Nav1.7 has not yet produced a clinical drug. Recent work has illuminated the Nav1.7 interactome, offering insights into the regulation of these channels and identifying potentially new druggable targets. Among the regulators of Nav1.7 is the cytosolic collapsin response mediator protein 2 (CRMP2). CRMP2, modified at lysine 374 (K374) by addition of a small ubiquitin-like modifier (SUMO), bound Nav1.7 to regulate its membrane localization and function. Corollary to this, preventing CRMP2 SUMOylation was sufficient to reverse mechanical allodynia in rats with neuropathic pain. Notably, loss of CRMP2 SUMOylation did not compromise other innate functions of CRMP2. To further elucidate the in vivo role of CRMP2 SUMOylation in pain, we generated CRMP2 K374A knock-in (CRMP2) mice in which Lys374 was replaced with Ala. CRMP2 mice had reduced Nav1.7 membrane localization and function in female, but not male, sensory neurons. Behavioral appraisal of CRMP2 mice demonstrated no changes in depressive or repetitive, compulsive-like behaviors and a decrease in noxious thermal sensitivity. No changes were observed in CRMP2 mice to inflammatory, acute, or visceral pain. By contrast, in a neuropathic model, CRMP2 mice failed to develop persistent mechanical allodynia. Our study suggests that CRMP2 SUMOylation-dependent control of peripheral Nav1.7 is a hallmark of chronic, but not physiological, neuropathic pain.
Collapse
|
61
|
Qi S, Zhang C, Yu H, Zhang J, Yan T, Lin Z, Yang B, Dong Z. Foldamer-Based Potassium Channels with High Ion Selectivity and Transport Activity. J Am Chem Soc 2021; 143:3284-3288. [PMID: 33645973 DOI: 10.1021/jacs.0c12128] [Citation(s) in RCA: 56] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
Small molecules that independently perform natural channel-like functions show greatly potential in the treatment of human diseases. Taking advantage of aromatic helical scaffolds, we develop a kind of foldamer-based ion channels with lumen size varying from 3.8 to 2.3 Å through a sequence substitution strategy. Our results clearly elucidate the importance of channel size in ion transport selectivity in molecular detail, eventually leading to the discoveries of the best artificial K+ channel by far and a rare sodium-preferential channel as well. High K+ selectivity and transport activity together make foldamers promising in therapeutic applications.
Collapse
Affiliation(s)
- Shuaiwei Qi
- State Key Laboratory of Supramolecular Structure and Materials, College of Chemistry, Jilin University, Changchun 130012, China
| | - Chenyang Zhang
- State Key Laboratory of Supramolecular Structure and Materials, College of Chemistry, Jilin University, Changchun 130012, China
| | - Hao Yu
- State Key Laboratory of Supramolecular Structure and Materials, College of Chemistry, Jilin University, Changchun 130012, China
| | - Jing Zhang
- State Key Laboratory of Supramolecular Structure and Materials, College of Chemistry, Jilin University, Changchun 130012, China
| | - Tengfei Yan
- State Key Laboratory of Supramolecular Structure and Materials, College of Chemistry, Jilin University, Changchun 130012, China
| | - Ze Lin
- State Key Laboratory of Supramolecular Structure and Materials, College of Chemistry, Jilin University, Changchun 130012, China
| | - Bing Yang
- State Key Laboratory of Supramolecular Structure and Materials, College of Chemistry, Jilin University, Changchun 130012, China
| | - Zeyuan Dong
- State Key Laboratory of Supramolecular Structure and Materials, College of Chemistry, Jilin University, Changchun 130012, China
| |
Collapse
|
62
|
Hasan MM, Ragnarsson L, Cardoso FC, Lewis RJ. Transfection methods for high-throughput cellular assays of voltage-gated calcium and sodium channels involved in pain. PLoS One 2021; 16:e0243645. [PMID: 33667217 PMCID: PMC7935312 DOI: 10.1371/journal.pone.0243645] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2020] [Accepted: 11/25/2020] [Indexed: 11/24/2022] Open
Abstract
Chemical transfection is broadly used to transiently transfect mammalian cells, although often associated with cellular stress and membrane instability, which imposes challenges for most cellular assays, including high-throughput (HT) assays. In the current study, we compared the effectiveness of calcium phosphate, FuGENE and Lipofectamine 3000 to transiently express two key voltage-gated ion channels critical in pain pathways, CaV2.2 and NaV1.7. The expression and function of these channels were validated using two HT platforms, the Fluorescence Imaging Plate Reader FLIPRTetra and the automated patch clamp QPatch 16X. We found that all transfection methods tested demonstrated similar effectiveness when applied to FLIPRTetra assays. Lipofectamine 3000-mediated transfection produced the largest peak currents for automated patch clamp QPatch assays. However, the FuGENE-mediated transfection was the most effective for QPatch assays as indicated by the superior number of cells displaying GΩ seal formation in whole-cell patch clamp configuration, medium to large peak currents, and higher rates of accomplished assays for both CaV2.2 and NaV1.7 channels. Our findings can facilitate the development of HT automated patch clamp assays for the discovery and characterization of novel analgesics and modulators of pain pathways, as well as assisting studies examining the pharmacology of mutated channels.
Collapse
Affiliation(s)
- Md. Mahadhi Hasan
- Division of Chemistry and Structural Biology, Institute for Molecular Bioscience, The University of Queensland, Brisbane, Qld, Australia
| | - Lotten Ragnarsson
- Division of Chemistry and Structural Biology, Institute for Molecular Bioscience, The University of Queensland, Brisbane, Qld, Australia
| | - Fernanda C. Cardoso
- Division of Chemistry and Structural Biology, Institute for Molecular Bioscience, The University of Queensland, Brisbane, Qld, Australia
- * E-mail: (FCC); (RJL)
| | - Richard J. Lewis
- Division of Chemistry and Structural Biology, Institute for Molecular Bioscience, The University of Queensland, Brisbane, Qld, Australia
- * E-mail: (FCC); (RJL)
| |
Collapse
|
63
|
Voltage-gated sodium channel blockers: new perspectives in the treatment of neuropathic pain. NEUROLOGÍA (ENGLISH EDITION) 2021. [DOI: 10.1016/j.nrleng.2020.02.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
|
64
|
Glasser M, Chen J, Alzarah M, Wallace M. Non-opioid Analgesics and Emerging Therapies. Cancer Treat Res 2021; 182:125-142. [PMID: 34542880 DOI: 10.1007/978-3-030-81526-4_9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/13/2023]
Abstract
Pain is a common and debilitating symptom of cancer. Cancer-related pain can occur at any point along the continuum from diagnosis to treatment to survivorship1. A systematic review published in 2016 estimated the prevalence of cancer pain to be 55% in those undergoing antineoplastic treatment, 66.4% in advanced cancer, and 39.3% in the post-treatment population. Thirty-eight percent of cancer patients in this pooled analysis experienced moderate to severe pain2.
Collapse
Affiliation(s)
- Marga Glasser
- Department of Anesthesiology, Center for Pain Medicine, UC San Diego Health System, 9300 Campus Point Dr, MC 7651, San Diego, USA
| | - Jeffrey Chen
- Department of Anesthesiology, Center for Pain Medicine, UC San Diego Health System, 9300 Campus Point Dr, MC 7651, San Diego, USA.
| | - Mohammed Alzarah
- Department of Anesthesiology, Center for Pain Medicine, UC San Diego Health System, 9300 Campus Point Dr, MC 7651, San Diego, USA
| | - Mark Wallace
- Department of Anesthesiology, Center for Pain Medicine, UC San Diego Health System, 9300 Campus Point Dr, MC 7651, San Diego, USA
| |
Collapse
|
65
|
Chen M, Peng S, Wang L, Yang L, Si Y, Zhou X, Zhang Y, Liu Z. Recombinant PaurTx-3, a spider toxin, inhibits sodium channels and decreases membrane excitability in DRG neurons. Biochem Biophys Res Commun 2020; 533:958-964. [PMID: 33004176 DOI: 10.1016/j.bbrc.2020.09.103] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2020] [Accepted: 09/23/2020] [Indexed: 10/23/2022]
Abstract
Voltage-gated sodium channels are critical for the generation and propagation of action potentials. Gating modifier toxins from spider venom can modulate the gating mechanism of sodium channels and thus have potential as drug leads. Here, we established expression of the gating modifier toxin PaurTx-3, a sodium channel inhibitor found in the venom of the spider Phrixotrichus auratus. Whole-cell voltage-clamp recordings indicated that recombinant PaurTx-3 (rPaurTx-3) inhibited Nav1.4, Nav1.5, and Nav1.7 currents with IC50 values of 61 nM, 72 nM, and 25 nM, respectively. Furthermore, rPaurTx-3 irreversibly inhibited Nav1.7 currents, but had 60-70% recovery in Nav1.4 and Nav1.5 after washing with a bath solution. rPaurTx-3 also hyperpolarized the voltage-dependent steady-state inactivation curve and significantly slowed recovery from fast inactivation of Nav1.7. Current-clamp recordings showed that rPaurTx-3 suppressed small DRG neuron activity. The biological activity assay findings for rPaurTx-3 support its potent pharmacological effect in Nav1.7 and small DRG neurons.
Collapse
Affiliation(s)
- Minzhi Chen
- The National and Local Joint Engineering Laboratory of Animal Peptide Drug Development, College of Life Sciences, Hunan Normal University, Changsha, 410081, China
| | - Shuijiao Peng
- The National and Local Joint Engineering Laboratory of Animal Peptide Drug Development, College of Life Sciences, Hunan Normal University, Changsha, 410081, China
| | - Li Wang
- The National and Local Joint Engineering Laboratory of Animal Peptide Drug Development, College of Life Sciences, Hunan Normal University, Changsha, 410081, China
| | - Li Yang
- The National and Local Joint Engineering Laboratory of Animal Peptide Drug Development, College of Life Sciences, Hunan Normal University, Changsha, 410081, China
| | - Yuxin Si
- The National and Local Joint Engineering Laboratory of Animal Peptide Drug Development, College of Life Sciences, Hunan Normal University, Changsha, 410081, China
| | - Xi Zhou
- The National and Local Joint Engineering Laboratory of Animal Peptide Drug Development, College of Life Sciences, Hunan Normal University, Changsha, 410081, China.
| | - Yunxiao Zhang
- Key Laboratory of Hunan Province for Advanced Carbon-based Functional Materials, School of Chemistry and Chemical Engineering, Hunan Institute of Science and Technology, Yueyang, 414006, Hunan, China.
| | - Zhonghua Liu
- The National and Local Joint Engineering Laboratory of Animal Peptide Drug Development, College of Life Sciences, Hunan Normal University, Changsha, 410081, China.
| |
Collapse
|
66
|
Wang M, Thyagarajan B. Pain pathways and potential new targets for pain relief. Biotechnol Appl Biochem 2020; 69:110-123. [PMID: 33316085 DOI: 10.1002/bab.2086] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2020] [Accepted: 12/06/2020] [Indexed: 12/15/2022]
Abstract
Pain is an unpleasant sensory and emotional experience that affects a sizable percentage of people on a daily basis. Sensory neurons known as nociceptors built specifically to detect damaging stimuli can be found throughout the body. They transmit information about noxious stimuli from mechanical, thermal, and chemical sources to the central nervous system and higher brain centers via electrical signals. Nociceptors express various channels and receptors such as voltage-gated sodium and calcium channels, transient receptor potential channels, and opioid receptors that allow them to respond in a highly specific manner to noxious stimuli. Attenuating the pain response can be achieved by inhibiting or altering the expression of these pain targets. Achieving a deeper understanding of how these receptors can be affected at the molecular level can lead to the development of novel pain therapies. This review will discuss the mechanisms of pain, introduce the various receptors that are responsible for detecting pain, and future directions in pharmacological therapies.
Collapse
Affiliation(s)
- Menglan Wang
- Department of Pharmaceutical Sciences, School of Pharmacy, University of Wyoming, Laramie, WY, USA
| | - Baskaran Thyagarajan
- Department of Pharmaceutical Sciences, School of Pharmacy, University of Wyoming, Laramie, WY, USA
| |
Collapse
|
67
|
Naik H, Steiner DJ, Versavel M, Palmer J, Fong R. Safety, Tolerability and Pharmacokinetics of Single and Repeat Doses of Vixotrigine in Healthy Volunteers. Clin Transl Sci 2020; 14:1272-1279. [PMID: 33278330 PMCID: PMC8301589 DOI: 10.1111/cts.12935] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2020] [Accepted: 11/02/2020] [Indexed: 11/30/2022] Open
Abstract
Neuropathic pain affects ~ 6.9–10% of the general population and leads to loss of function, anxiety, depression, sleep disturbance, and impaired cognition. Here, we report the safety, tolerability, and pharmacokinetics of a voltage‐dependent and use‐dependent sodium channel blocker, vixotrigine, currently under investigation for the treatment of neuropathic pain conditions. The randomized, placebo‐controlled, phase I clinical trials were split into single ascending dose (SAD) and multiple ascending dose (MAD) studies. Healthy volunteers received oral vixotrigine as either single doses followed by a ≥ 7‐day washout period for up to 5 dosing sessions (SAD, n = 30), or repeat doses (once or twice daily) for 14 and 28 days (MAD, n = 51). Adverse events (AEs), maximum observed vixotrigine plasma concentration (Cmax), area under the concentration‐time curve from predose to 24 hours postdose (AUC0–24), time to Cmax (Tmax), and terminal half‐life (t1/2), among others, were assessed. Drug‐related AEs were reported in 47% and 53% of volunteers in the SAD and MAD studies, respectively, with dizziness as the most commonly reported drug‐related AE. SAD results showed that Cmax and AUC increased with dose, Tmax was 1–2 hours, and t1/2 was ~ 11 hours. A twofold increase in accumulation was observed when vixotrigine was taken twice vs. once daily (MAD). Steady‐state was achieved from day 5 onward. These data indicate that oral vixotrigine is well‐tolerated when administered as single doses up to 825 mg and multiple doses up to 450 mg twice daily.
Collapse
Affiliation(s)
| | | | - Mark Versavel
- Biogen, Cambridge, Massachusetts, USA.,Convergence Pharmaceuticals Ltd., a Biogen company, Cambridge, UK
| | | | - Regan Fong
- GlaxoSmithKline, King of Prussia, Pennsylvania, USA
| |
Collapse
|
68
|
Li GZ, Hu YH, Li DY, Zhang Y, Guo HL, Li YM, Chen F, Xu J. Vincristine-induced peripheral neuropathy: A mini-review. Neurotoxicology 2020; 81:161-171. [DOI: 10.1016/j.neuro.2020.10.004] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2020] [Revised: 09/29/2020] [Accepted: 10/07/2020] [Indexed: 12/11/2022]
|
69
|
Controlling the "Opioid Epidemic": A Novel Chemical Entity (NCE) to Reduce or Supplant Opiate Use for Chronic Pain. ACTA ACUST UNITED AC 2020; 5. [PMID: 33117893 PMCID: PMC7591148 DOI: 10.20900/jpbs.20200022] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
We report on the ongoing project “A Novel Therapeutic to Ameliorate Chronic Pain and Reduce Opiate Use.” Over 100 million adults in the U.S. suffer from intermittent or constant chronic pain, and chronic pain affects at least 10% of the world’s population. The primary pharmaceuticals for treatment of chronic pain have been natural or synthetic opioids and the use of opioids for pain treatment has resulted in what has been called an “epidemic” of opioid abuse, addiction and lethal overdoses. We have, through a process of rational drug design, generated a novel chemical entity (NCE) and have given it the name Kindolor. Kindolor is a non-opiate, non-addicting molecule that was developed specifically to simultaneously control the aberrant activity of three targets on the peripheral sensory system that are integral in the development and propagation of chronic pain. In our initial preclinical studies, we demonstrated the efficacy of Kindolor to reduce or eliminate chronic pain in five animal models. The overall goal of the project is to complete the investigational new drug (IND)-enabling preclinical studies of Kindolor, and once IND approval is gained, we will proceed to the clinical Phase Ia and 1b safety studies and a Phase 2a efficacy study. The work is in its second year, and the present report describes progress toward our overall goal of bringing our compound to a full Phase 2 ready stage.
Collapse
|
70
|
Toffano AA, Chiarot G, Zamuner S, Marchi M, Salvi E, Waxman SG, Faber CG, Lauria G, Giacometti A, Simeoni M. Computational pipeline to probe NaV1.7 gain-of-function variants in neuropathic painful syndromes. Sci Rep 2020; 10:17930. [PMID: 33087732 PMCID: PMC7578092 DOI: 10.1038/s41598-020-74591-y] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2020] [Accepted: 09/30/2020] [Indexed: 01/09/2023] Open
Abstract
Applications of machine learning and graph theory techniques to neuroscience have witnessed an increased interest in the last decade due to the large data availability and unprecedented technology developments. Their employment to investigate the effect of mutational changes in genes encoding for proteins modulating the membrane of excitable cells, whose biological correlates are assessed at electrophysiological level, could provide useful predictive clues. We apply this concept to the analysis of variants in sodium channel NaV1.7 subunit found in patients with chronic painful syndromes, by the implementation of a dedicated computational pipeline empowering different and complementary techniques including homology modeling, network theory, and machine learning. By testing three templates of different origin and sequence identities, we provide an optimal condition for its use. Our findings reveal the usefulness of our computational pipeline in supporting the selection of candidates for cell electrophysiology assay and with potential clinical applications.
Collapse
Affiliation(s)
- Alberto A Toffano
- Dipartimento di Scienze Molecolari e Nanosistemi, Universitá Ca' Foscari Venezia, Venezia-Mestre, Italy
| | - Giacomo Chiarot
- Dipartimento di Scienze Ambientali, Informatica e Statistica, Universitá Ca' Foscari Venezia, Venezia-Mestre, Italy
| | - Stefano Zamuner
- Laboratory of Statistical Biophysics, Institute of Physics, School of Basic Sciences, Ècole Polytechnique Fèdèrale de Lausanne (EPFL), Lausanne, Switzerland
| | - Margherita Marchi
- Neuroalgology Unit, Fondazione IRCCS Istituto Neurologico "Carlo Besta", Milan, Italy
| | - Erika Salvi
- Neuroalgology Unit, Fondazione IRCCS Istituto Neurologico "Carlo Besta", Milan, Italy
| | - Stephen G Waxman
- Center for Neuroscience and Regeneration Research, VA Connecticut Healthcare System and Yale Medical School, West Haven, USA
| | - Catharina G Faber
- MHeNs school for Mental Health and Neuroscience, Maastricht University, Maastricht, The Netherlands.,Department of Neurology, Maastricht University Medical Center, Maastricht, The Netherlands
| | - Giuseppe Lauria
- Neuroalgology Unit, Fondazione IRCCS Istituto Neurologico "Carlo Besta", Milan, Italy.,Department of Biomedical and Clinical Sciences "Luigi Sacco", University of Milan, Milan, Italy
| | - Achille Giacometti
- Dipartimento di Scienze Molecolari e Nanosistemi, Universitá Ca' Foscari Venezia, Venezia-Mestre, Italy.,European Centre for Living Technology (ECLT), Venice, Italy
| | - Marta Simeoni
- Dipartimento di Scienze Ambientali, Informatica e Statistica, Universitá Ca' Foscari Venezia, Venezia-Mestre, Italy. .,European Centre for Living Technology (ECLT), Venice, Italy.
| |
Collapse
|
71
|
Liao P, Qiu Y, Mo Y, Fu J, Song Z, Huang L, Bai S, Wang Y, Zhu JJ, Tian F, Chen Z, Pan N, Sun EY, Yang L, Lan X, Chen Y, Huang D, Sun P, Zhao L, Yang D, Lu W, Yang T, Xiao J, Li WG, Gao Z, Shen B, Zhang Q, Liu J, Jiang H, Jiang R, Yang H. Selective activation of TWIK-related acid-sensitive K + 3 subunit-containing channels is analgesic in rodent models. Sci Transl Med 2020; 11:11/519/eaaw8434. [PMID: 31748231 DOI: 10.1126/scitranslmed.aaw8434] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2019] [Revised: 06/27/2019] [Accepted: 09/19/2019] [Indexed: 02/05/2023]
Abstract
The paucity of selective agonists for TWIK-related acid-sensitive K+ 3 (TASK-3) channel, a member of two-pore domain K+ (K2P) channels, has contributed to our limited understanding of its biological functions. By targeting a druggable transmembrane cavity using a structure-based drug design approach, we discovered a biguanide compound, CHET3, as a highly selective allosteric activator for TASK-3-containing K2P channels, including TASK-3 homomers and TASK-3/TASK-1 heteromers. CHET3 displayed potent analgesic effects in vivo in a variety of acute and chronic pain models in rodents that could be abolished pharmacologically or by genetic ablation of TASK-3. We further found that TASK-3-containing channels anatomically define a unique population of small-sized, transient receptor potential cation channel subfamily M member 8 (TRPM8)-, transient receptor potential cation channel subfamily V member 1 (TRPV1)-, or tyrosine hydroxylase (TH)-positive nociceptive sensory neurons and functionally regulate their membrane excitability, supporting CHET3 analgesic effects in thermal hyperalgesia and mechanical allodynia under chronic pain. Overall, our proof-of-concept study reveals TASK-3-containing K2P channels as a druggable target for treating pain.
Collapse
Affiliation(s)
- Ping Liao
- Laboratory of Anesthesia and Critical Care Medicine, Department of Anesthesiology, National-Local Joint Engineering Research Center of Translational Medicine of Anesthesiology, West China Hospital of Sichuan University, Chengdu, Sichuan 610000, China
| | - Yunguang Qiu
- State Key Laboratory of Drug Research and CAS Key Laboratory of Receptor Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China.,University of Chinese Academy of Sciences, Beijing 100049, China
| | - Yiqing Mo
- Shanghai Key Laboratory of Regulatory Biology, Institute of Biomedical Sciences, School of Life Sciences, East China Normal University, Shanghai 200241, China
| | - Jie Fu
- Shanghai Key Laboratory of Regulatory Biology, Institute of Biomedical Sciences, School of Life Sciences, East China Normal University, Shanghai 200241, China
| | - Zhenpeng Song
- Laboratory of Anesthesia and Critical Care Medicine, Department of Anesthesiology, National-Local Joint Engineering Research Center of Translational Medicine of Anesthesiology, West China Hospital of Sichuan University, Chengdu, Sichuan 610000, China
| | - Lu Huang
- Laboratory of Anesthesia and Critical Care Medicine, Department of Anesthesiology, National-Local Joint Engineering Research Center of Translational Medicine of Anesthesiology, West China Hospital of Sichuan University, Chengdu, Sichuan 610000, China
| | - Suwen Bai
- Department of Physiology, Anhui Medical University, Hefei, Anhui 230032, China
| | - Yang Wang
- Department of Physiology, Anhui Medical University, Hefei, Anhui 230032, China
| | - Jia-Jie Zhu
- Collaborative Innovation Center for Brain Science, Department of Anatomy and Physiology, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Fuyun Tian
- State Key Laboratory of Drug Research and CAS Key Laboratory of Receptor Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
| | - Zhuo Chen
- Laboratory of Anesthesia and Critical Care Medicine, Department of Anesthesiology, National-Local Joint Engineering Research Center of Translational Medicine of Anesthesiology, West China Hospital of Sichuan University, Chengdu, Sichuan 610000, China
| | - Nanfang Pan
- Laboratory of Anesthesia and Critical Care Medicine, Department of Anesthesiology, National-Local Joint Engineering Research Center of Translational Medicine of Anesthesiology, West China Hospital of Sichuan University, Chengdu, Sichuan 610000, China
| | - Er-Yi Sun
- Laboratory of Anesthesia and Critical Care Medicine, Department of Anesthesiology, National-Local Joint Engineering Research Center of Translational Medicine of Anesthesiology, West China Hospital of Sichuan University, Chengdu, Sichuan 610000, China
| | - Linghui Yang
- Laboratory of Anesthesia and Critical Care Medicine, Department of Anesthesiology, National-Local Joint Engineering Research Center of Translational Medicine of Anesthesiology, West China Hospital of Sichuan University, Chengdu, Sichuan 610000, China
| | - Xi Lan
- State Key Laboratory of Drug Research and CAS Key Laboratory of Receptor Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
| | - Yinbin Chen
- Shanghai Key Laboratory of Regulatory Biology, Institute of Biomedical Sciences, School of Life Sciences, East China Normal University, Shanghai 200241, China
| | - Dongping Huang
- Shanghai Key Laboratory of Regulatory Biology, Institute of Biomedical Sciences, School of Life Sciences, East China Normal University, Shanghai 200241, China
| | - Peihua Sun
- Jiangsu Key Laboratory of Neuropsychiatric Diseases and College of Pharmaceutical Sciences, Soochow University, Suzhou, Jiangsu 215123, China
| | - Lifen Zhao
- State Key Laboratory of Drug Research and CAS Key Laboratory of Receptor Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
| | - Dehua Yang
- State Key Laboratory of Drug Research and CAS Key Laboratory of Receptor Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
| | - Weiqiang Lu
- Shanghai Key Laboratory of Regulatory Biology, Institute of Biomedical Sciences, School of Life Sciences, East China Normal University, Shanghai 200241, China
| | - Tingting Yang
- Cardiac Regeneration and Ageing Lab, School of Life Science, Shanghai University, Shanghai 200444, China
| | - Junjie Xiao
- Cardiac Regeneration and Ageing Lab, School of Life Science, Shanghai University, Shanghai 200444, China
| | - Wei-Guang Li
- Collaborative Innovation Center for Brain Science, Department of Anatomy and Physiology, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Zhaobing Gao
- State Key Laboratory of Drug Research and CAS Key Laboratory of Receptor Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
| | - Bing Shen
- Department of Physiology, Anhui Medical University, Hefei, Anhui 230032, China
| | - Qiansen Zhang
- Shanghai Key Laboratory of Regulatory Biology, Institute of Biomedical Sciences, School of Life Sciences, East China Normal University, Shanghai 200241, China
| | - Jin Liu
- Laboratory of Anesthesia and Critical Care Medicine, Department of Anesthesiology, National-Local Joint Engineering Research Center of Translational Medicine of Anesthesiology, West China Hospital of Sichuan University, Chengdu, Sichuan 610000, China
| | - Hualiang Jiang
- State Key Laboratory of Drug Research and CAS Key Laboratory of Receptor Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China.,University of Chinese Academy of Sciences, Beijing 100049, China
| | - Ruotian Jiang
- Laboratory of Anesthesia and Critical Care Medicine, Department of Anesthesiology, National-Local Joint Engineering Research Center of Translational Medicine of Anesthesiology, West China Hospital of Sichuan University, Chengdu, Sichuan 610000, China.
| | - Huaiyu Yang
- Shanghai Key Laboratory of Regulatory Biology, Institute of Biomedical Sciences, School of Life Sciences, East China Normal University, Shanghai 200241, China.
| |
Collapse
|
72
|
Robles-Gómez E, Benítez-Villalobos F, Soriano-García M, Antúnez-Argüelles E. Non-peptide molecules in the pedicellariae of Toxopneustes roseus. Toxicon 2020; 184:143-151. [DOI: 10.1016/j.toxicon.2020.06.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2019] [Revised: 05/25/2020] [Accepted: 06/02/2020] [Indexed: 11/30/2022]
|
73
|
Computational analysis of a 9D model for a small DRG neuron. J Comput Neurosci 2020; 48:429-444. [PMID: 32862338 DOI: 10.1007/s10827-020-00761-6] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2020] [Revised: 07/20/2020] [Accepted: 08/03/2020] [Indexed: 10/23/2022]
Abstract
Small dorsal root ganglion (DRG) neurons are primary nociceptors which are responsible for sensing pain. Elucidation of their dynamics is essential for understanding and controlling pain. To this end, we present a numerical bifurcation analysis of a small DRG neuron model in this paper. The model is of Hodgkin-Huxley type and has 9 state variables. It consists of a Nav1.7 and a Nav1.8 sodium channel, a leak channel, a delayed rectifier potassium, and an A-type transient potassium channel. The dynamics of this model strongly depend on the maximal conductances of the voltage-gated ion channels and the external current, which can be adjusted experimentally. We show that the neuron dynamics are most sensitive to the Nav1.8 channel maximal conductance ([Formula: see text]). Numerical bifurcation analysis shows that depending on [Formula: see text] and the external current, different parameter regions can be identified with stable steady states, periodic firing of action potentials, mixed-mode oscillations (MMOs), and bistability between stable steady states and stable periodic firing of action potentials. We illustrate and discuss the transitions between these different regimes. We further analyze the behavior of MMOs. As the external current is decreased, we find that MMOs appear after a cyclic limit point. Within this region, bifurcation analysis shows a sequence of isolated periodic solution branches with one large action potential and a number of small amplitude peaks per period. For decreasing external current, the number of small amplitude peaks is increasing and the distance between the large amplitude action potentials is growing, finally tending to infinity and thereby leading to a stable steady state. A closer inspection reveals more complex concatenated MMOs in between these periodic MMO branches, forming Farey sequences. Lastly, we also find small solution windows with aperiodic oscillations which seem to be chaotic. The dynamical patterns found here-as consequences of bifurcation points regulated by different parameters-have potential translational significance as repetitive firing of action potentials imply pain of some form and intensity; manipulating these patterns by regulating the different parameters could aid in investigating pain dynamics.
Collapse
|
74
|
Bertamino A, Ostacolo C, Medina A, Di Sarno V, Lauro G, Ciaglia T, Vestuto V, Pepe G, Basilicata MG, Musella S, Smaldone G, Cristiano C, Gonzalez-Rodriguez S, Fernandez-Carvajal A, Bifulco G, Campiglia P, Gomez-Monterrey I, Russo R. Exploration of TRPM8 Binding Sites by β-Carboline-Based Antagonists and Their In Vitro Characterization and In Vivo Analgesic Activities. J Med Chem 2020; 63:9672-9694. [PMID: 32787109 PMCID: PMC8009520 DOI: 10.1021/acs.jmedchem.0c00816] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
![]()
Transient
receptor potential melastatin 8 (TRPM8) ion channel represents
a valuable pharmacological option for several therapeutic areas. Here,
a series of conformationally restricted derivatives of the previously
described TRPM8 antagonist N,N′-dibenzyl
tryptophan 4 were prepared and characterized in vitro
by Ca2+-imaging and patch-clamp electrophysiology assays.
Molecular modeling studies led to identification of a broad and well-defined
interaction network of these derivatives inside the TRPM8 binding
site, underlying their antagonist activity. The (5R,11aS)-5-(4-chlorophenyl)-2-(4-fluorobenzyl)-5,6,11,11a-tetrahydro-1H-imidazo[1′,5′:1,6]pyrido[3,4-b]indole-1,3(2H)-dione (31a) emerged as a potent (IC50 = 4.10 ± 1.2 nM), selective,
and metabolically stable TRPM8 antagonist. In vivo, 31a showed significant target coverage in an icilin-induced WDS (at
11.5 mg/kg ip), an oxaliplatin-induced cold allodynia (at 10–30
μg sc), and CCI-induced thermal hyperalgesia (at 11.5 mg/kg
ip) mice models. These results confirm the tryptophan moiety as a
solid pharmacophore template for the design of highly potent modulators
of TRPM8-mediated activities.
Collapse
Affiliation(s)
- Alessia Bertamino
- Department of Pharmacy, University of Salerno, Via G. Paolo II 132, 84084 Fisciano, Salerno, Italy
| | - Carmine Ostacolo
- Department of Pharmacy, University Federico II of Naples, Via D. Montesano 49, 80131 Naples, Italy
| | - Alicia Medina
- IDiBE, Universitas Miguel Herna'ndez, Avda de la Universidad, 032020 Elche, Spain
| | - Veronica Di Sarno
- Department of Pharmacy, University of Salerno, Via G. Paolo II 132, 84084 Fisciano, Salerno, Italy
| | - Gianluigi Lauro
- Department of Pharmacy, University of Salerno, Via G. Paolo II 132, 84084 Fisciano, Salerno, Italy
| | - Tania Ciaglia
- Department of Pharmacy, University of Salerno, Via G. Paolo II 132, 84084 Fisciano, Salerno, Italy
| | - Vincenzo Vestuto
- Department of Pharmacy, University of Salerno, Via G. Paolo II 132, 84084 Fisciano, Salerno, Italy
| | - Giacomo Pepe
- Department of Pharmacy, University of Salerno, Via G. Paolo II 132, 84084 Fisciano, Salerno, Italy
| | | | - Simona Musella
- European Biomedical Research Institute (EBRIS), Via S. De Renzi 50, 84125 Salerno, Italy
| | - Gerardina Smaldone
- Department of Pharmacy, University of Salerno, Via G. Paolo II 132, 84084 Fisciano, Salerno, Italy
| | - Claudia Cristiano
- Department of Pharmacy, University Federico II of Naples, Via D. Montesano 49, 80131 Naples, Italy
| | | | | | - Giuseppe Bifulco
- Department of Pharmacy, University of Salerno, Via G. Paolo II 132, 84084 Fisciano, Salerno, Italy
| | - Pietro Campiglia
- Department of Pharmacy, University of Salerno, Via G. Paolo II 132, 84084 Fisciano, Salerno, Italy.,European Biomedical Research Institute (EBRIS), Via S. De Renzi 50, 84125 Salerno, Italy
| | - Isabel Gomez-Monterrey
- Department of Pharmacy, University Federico II of Naples, Via D. Montesano 49, 80131 Naples, Italy
| | - Roberto Russo
- Department of Pharmacy, University Federico II of Naples, Via D. Montesano 49, 80131 Naples, Italy
| |
Collapse
|
75
|
Sonvane S, Choudhari P, Bhusnuare O. In silico analysis of polyphenols and flavonoids for design of human Nav1.7 inhibitors. J Biomol Struct Dyn 2020; 39:4472-4479. [PMID: 32686994 DOI: 10.1080/07391102.2020.1777902] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/23/2022]
Abstract
Neuropathic pain is commonly associated with lesion or disease of the somatosensory system and often reflected as indicator of impaired life. Although the central nervous system is main regulator of pain but for initiation and maintenance of the neuropathic pain is regulated by peripheral nervous system. Sodium channels particularly Nav1.7, Nav1.8, Nav 1.9 are key stake holders in the peripheral neuropathy, activation of these sodium channels might lead to genesis and propagation. Flavonoids and polyphenols showed promising effects in neuropathic pain. Here we are reporting In silico analysis of some selected flavonoids and polyphenols on sodium activated voltage channel 1.7 to explore the structural fragments required for binding. Results indicated Baicalin, Butrin, Dihydromonospermoside, Icariin, Isocoreopsin and Isosaponarin are showing promising docking score with sodium activated voltage channel 1.7 than other compounds. Structural modification of these promising leads keeping pharamcophoric requirement intact may yield potent Nav1.7 inhibitors for peripheral pain management.Communicated by Ramaswamy H. Sarma.
Collapse
Affiliation(s)
- Sameep Sonvane
- Channabasweshwar Pharmacy College (Degree), Latur, India.,Department of Pharmaceutical Chemistry, Dayanand College of Pharmacy, Latur, India
| | - Prafulla Choudhari
- Department of Pharmaceutical Chemistry, Bharati Vidyapeeth College of Pharmacy, Kolhapur, India
| | - Omprakash Bhusnuare
- Channabasweshwar Pharmacy College (Degree), Latur, India.,Department of Pharmaceutical Chemistry, Dayanand College of Pharmacy, Latur, India
| |
Collapse
|
76
|
Painful and painless mutations of SCN9A and SCN11A voltage-gated sodium channels. Pflugers Arch 2020; 472:865-880. [PMID: 32601768 PMCID: PMC7351857 DOI: 10.1007/s00424-020-02419-9] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2020] [Revised: 05/25/2020] [Accepted: 06/10/2020] [Indexed: 12/11/2022]
Abstract
Chronic pain is a global problem affecting up to 20% of the world’s population and has a significant economic, social and personal cost to society. Sensory neurons of the dorsal root ganglia (DRG) detect noxious stimuli and transmit this sensory information to regions of the central nervous system (CNS) where activity is perceived as pain. DRG neurons express multiple voltage-gated sodium channels that underlie their excitability. Research over the last 20 years has provided valuable insights into the critical roles that two channels, NaV1.7 and NaV1.9, play in pain signalling in man. Gain of function mutations in NaV1.7 cause painful conditions while loss of function mutations cause complete insensitivity to pain. Only gain of function mutations have been reported for NaV1.9. However, while most NaV1.9 mutations lead to painful conditions, a few are reported to cause insensitivity to pain. The critical roles these channels play in pain along with their low expression in the CNS and heart muscle suggest they are valid targets for novel analgesic drugs.
Collapse
|
77
|
Employing NaChBac for cryo-EM analysis of toxin action on voltage-gated Na + channels in nanodisc. Proc Natl Acad Sci U S A 2020; 117:14187-14193. [PMID: 32513729 DOI: 10.1073/pnas.1922903117] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
NaChBac, the first bacterial voltage-gated Na+ (Nav) channel to be characterized, has been the prokaryotic prototype for studying the structure-function relationship of Nav channels. Discovered nearly two decades ago, the structure of NaChBac has not been determined. Here we present the single particle electron cryomicroscopy (cryo-EM) analysis of NaChBac in both detergent micelles and nanodiscs. Under both conditions, the conformation of NaChBac is nearly identical to that of the potentially inactivated NavAb. Determining the structure of NaChBac in nanodiscs enabled us to examine gating modifier toxins (GMTs) of Nav channels in lipid bilayers. To study GMTs in mammalian Nav channels, we generated a chimera in which the extracellular fragment of the S3 and S4 segments in the second voltage-sensing domain from Nav1.7 replaced the corresponding sequence in NaChBac. Cryo-EM structures of the nanodisc-embedded chimera alone and in complex with HuwenToxin IV (HWTX-IV) were determined to 3.5 and 3.2 Å resolutions, respectively. Compared to the structure of HWTX-IV-bound human Nav1.7, which was obtained at an overall resolution of 3.2 Å, the local resolution of the toxin has been improved from ∼6 to ∼4 Å. This resolution enabled visualization of toxin docking. NaChBac can thus serve as a convenient surrogate for structural studies of the interactions between GMTs and Nav channels in a membrane environment.
Collapse
|
78
|
Ren WJ, Ulrich H, Semyanov A, Illes P, Tang Y. TASK-3: New Target for Pain-Relief. Neurosci Bull 2020; 36:951-954. [PMID: 32458273 DOI: 10.1007/s12264-020-00516-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2020] [Accepted: 04/24/2020] [Indexed: 11/26/2022] Open
Affiliation(s)
- Wen-Jing Ren
- International Collaborative Centre on Big Science Plan for Purine Signaling, Chengdu University of Traditional Chinese Medicine, Chengdu, 610075, China
- Acupuncture and Chronobiology Key Laboratory of Sichuan Province, Chengdu, 610075, China
| | - Henning Ulrich
- Department of Biochemistry, Institute of Chemistry, University of São Paulo, São Paulo, 05508, Brazil
| | - Alexey Semyanov
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, Moscow, Russia, 117997
| | - Peter Illes
- International Collaborative Centre on Big Science Plan for Purine Signaling, Chengdu University of Traditional Chinese Medicine, Chengdu, 610075, China.
- Rudolf-Boehm-Institut für Pharmakologie und Toxikologie, Universitaet Leipzig, 04109, Leipzig, Sachsen, Germany.
| | - Yong Tang
- International Collaborative Centre on Big Science Plan for Purine Signaling, Chengdu University of Traditional Chinese Medicine, Chengdu, 610075, China.
- Acupuncture and Chronobiology Key Laboratory of Sichuan Province, Chengdu, 610075, China.
| |
Collapse
|
79
|
Drissi I, Woods WA, Woods CG. Understanding the genetic basis of congenital insensitivity to pain. Br Med Bull 2020; 133:65-78. [PMID: 32219415 PMCID: PMC7227775 DOI: 10.1093/bmb/ldaa003] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/18/2019] [Revised: 01/21/2020] [Accepted: 01/31/2020] [Indexed: 12/15/2022]
Abstract
INTRODUCTION OR BACKGROUND Congenital insensitivity to pain (CIP) is caused by extremely rare Mendelian genetic disorders. CIP individuals demonstrate the unexpectedly severe consequences of painlessness. Although only a small number of causative conditions and genes are known, most have led to profound insights into human nociception. CIP gene discovery is catalyzing the manufacture of completely new classes of analgesics, and these are needed as alternatives to synthetic highly potent opioids. SOURCES OF DATA Pubmed.gov peer-reviewed journal articles and reviews. AREAS OF AGREEMENT The importance of nerve growth factor-tropomyosin receptor kinase A (NGF-TRKA) signalling for nociceptor genesis and subsequent pain sensing.New analgesics can be generated from knowledge of the NGF-TRKA nociceptor pathway.Increased susceptibility to Staphylococcus aureus infection is a consequence of deficient NGF-TRKA signalling.Mutations in the voltage-gated sodium channels SCN9A and SCN11A can cause congenital painlessness, and in contradistinction, other mutations can cause episodic neuropathic pain. SCN9A/Nav1.7 is an analgesic target. SCN11A/Nav1.9 is unlikely to be an analgesic target.There are further Mendelian causes of painlessness to be discovered. AREAS OF CONTROVERSY Which NGF-TRKA intracellular signalling pathways operate in nociceptor development and which in post-natal pain sensing?Why have no clinically effective Nav1.7 antagonist been generated? SCN9A-CIP causes analgesia, at least in part, through endogenous opioids.Why do all CIP phenotypes involve a complete loss of all types of nociception? AREAS TIMELY FOR DEVELOPING RESEARCH PRDM12 as an analgesic target.Discovery of the function and analgesic potential of new CIP genes.Can NGF-TRKA be used in the treatment of S. aureus?
Collapse
Affiliation(s)
- Ichrak Drissi
- Cambridge Institute for Medical Research, University of Cambridge, Cambridge Biomedical Campus, The Keith Peters Building, Hills Road, Cambridge CB2 0XY, UK
| | - William Aidan Woods
- School of Medicine, David Weatherall building, University Road, Keele University, Staffordshire ST5 5BG, UK
| | - Christopher Geoffrey Woods
- Cambridge Institute for Medical Research, University of Cambridge, Cambridge Biomedical Campus, The Keith Peters Building, Hills Road, Cambridge CB2 0XY, UK
| |
Collapse
|
80
|
Chu Y, Qiu P, Yu R. Centipede Venom Peptides Acting on Ion Channels. Toxins (Basel) 2020; 12:toxins12040230. [PMID: 32260499 PMCID: PMC7232367 DOI: 10.3390/toxins12040230] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2020] [Revised: 03/30/2020] [Accepted: 04/01/2020] [Indexed: 12/19/2022] Open
Abstract
Centipedes are among the oldest venomous arthropods that use their venom to subdue the prey. The major components of centipede venom are a variety of low-molecular-weight peptide toxins that have evolved to target voltage-gated ion channels to interfere with the central system of prey and produce pain or paralysis for efficient hunting. Peptide toxins usually contain several intramolecular disulfide bonds, which confer chemical, thermal and biological stability. In addition, centipede peptides generally have novel structures and high potency and specificity and therefore hold great promise both as diagnostic tools and in the treatment of human disease. Here, we review the centipede peptide toxins with reported effects on ion channels, including Nav, Kv, Cav and the nonselective cation channel polymodal transient receptor potential vanilloid 1 (TRPV1).
Collapse
Affiliation(s)
- YanYan Chu
- School of Medicine and Pharmacy, Ocean University of China, 5 Yushan Road, Qingdao 266003, China;
- Laboratory for Marine Drugs and Bioproducts, Qingdao National Laboratory for Marine Science and Technology, Qingdao 266003, China
- Innovation Center for Marine Drug Screening & Evaluation, Qingdao National Laboratory for Marine Science and Technology, Qingdao 266003, China
- Marine Biomedical Research Institute of Qingdao, Qingdao 266071, China
- Correspondence: (Y.C.); (R.Y.)
| | - PeiJu Qiu
- School of Medicine and Pharmacy, Ocean University of China, 5 Yushan Road, Qingdao 266003, China;
- Laboratory for Marine Drugs and Bioproducts, Qingdao National Laboratory for Marine Science and Technology, Qingdao 266003, China
- Innovation Center for Marine Drug Screening & Evaluation, Qingdao National Laboratory for Marine Science and Technology, Qingdao 266003, China
- Marine Biomedical Research Institute of Qingdao, Qingdao 266071, China
| | - RiLei Yu
- School of Medicine and Pharmacy, Ocean University of China, 5 Yushan Road, Qingdao 266003, China;
- Laboratory for Marine Drugs and Bioproducts, Qingdao National Laboratory for Marine Science and Technology, Qingdao 266003, China
- Innovation Center for Marine Drug Screening & Evaluation, Qingdao National Laboratory for Marine Science and Technology, Qingdao 266003, China
- Correspondence: (Y.C.); (R.Y.)
| |
Collapse
|
81
|
Alcántara Montero A, Sánchez Carnerero CI. [Voltage-gated sodium channel blockers: New perspectives in the treatment of neuropathic pain]. Neurologia 2020; 36:169-171. [PMID: 32253051 DOI: 10.1016/j.nrl.2020.02.004] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2019] [Revised: 02/10/2020] [Accepted: 02/27/2020] [Indexed: 01/10/2023] Open
Affiliation(s)
- A Alcántara Montero
- Centro de Salud Manuel Encinas, Consultorio de Malpartida de Cáceres, Cáceres, España.
| | - C I Sánchez Carnerero
- Complejo Hospitalario Universitario de Cáceres, Hospital Universitario, Cáceres, España
| |
Collapse
|
82
|
Busserolles J, Lolignier S, Kerckhove N, Bertin C, Authier N, Eschalier A. Replacement of current opioid drugs focusing on MOR-related strategies. Pharmacol Ther 2020; 210:107519. [PMID: 32165137 DOI: 10.1016/j.pharmthera.2020.107519] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2019] [Accepted: 02/24/2020] [Indexed: 12/12/2022]
Abstract
The scarcity and limited risk/benefit ratio of painkillers available on the market, in addition to the opioid crisis, warrant reflection on new innovation strategies. The pharmacopoeia of analgesics is based on products that are often old and derived from clinical empiricism, with limited efficacy or spectrum of action, or resulting in an unsatisfactory tolerability profile. Although they are reference analgesics for nociceptive pain, opioids are subject to the same criticism. The use of opium as an analgesic is historical. Morphine was synthesized at the beginning of the 19th century. The efficacy of opioids is limited in certain painful contexts and these drugs can induce potentially serious and fatal adverse effects. The current North American opioid crisis, with an ever-rising number of deaths by opioid overdose, is a tragic illustration of this. It is therefore legitimate to develop research into molecules likely to maintain or increase opioid efficacy while improving their tolerability. Several avenues are being explored including targeting of the mu opioid receptor (MOR) splice variants, developing biased agonists or targeting of other receptors such as heteromers with MOR. Ion channels acting as MOR effectors, are also targeted in order to offer compounds without MOR-dependent adverse effects. Another route is to develop opioid analgesics with peripheral action or limited central nervous system (CNS) access. Finally, endogenous opioids used as drugs or compounds that modify the metabolism of endogenous opioids (Dual ENKephalinase Inhibitors) are being developed. The aim of the present review is to present these various targets/strategies with reference to current indications for opioids, concerns about their widespread use, particularly in chronic non-cancer pains, and ways of limiting the risk of opioid abuse and misuse.
Collapse
Affiliation(s)
- Jérôme Busserolles
- Université Clermont Auvergne, INSERM, CHU, NEURO-DOL Pharmacologie Fondamentale et Clinique de la douleur, F-63000 Clermont-Ferrand, France; Institut ANALGESIA, Faculté de Médecine, F-63000 Clermont-Ferrand, France
| | - Stéphane Lolignier
- Université Clermont Auvergne, INSERM, CHU, NEURO-DOL Pharmacologie Fondamentale et Clinique de la douleur, F-63000 Clermont-Ferrand, France; Institut ANALGESIA, Faculté de Médecine, F-63000 Clermont-Ferrand, France
| | - Nicolas Kerckhove
- Université Clermont Auvergne, INSERM, CHU, NEURO-DOL Pharmacologie Fondamentale et Clinique de la douleur, F-63000 Clermont-Ferrand, France; Institut ANALGESIA, Faculté de Médecine, F-63000 Clermont-Ferrand, France; Observatoire Français des Médicaments Antalgiques (OFMA), French monitoring centre for analgesic drugs, CHU, F-63000 Clermont-Ferrand, France
| | - Célian Bertin
- Université Clermont Auvergne, INSERM, CHU, NEURO-DOL Pharmacologie Fondamentale et Clinique de la douleur, F-63000 Clermont-Ferrand, France; Institut ANALGESIA, Faculté de Médecine, F-63000 Clermont-Ferrand, France; Observatoire Français des Médicaments Antalgiques (OFMA), French monitoring centre for analgesic drugs, CHU, F-63000 Clermont-Ferrand, France
| | - Nicolas Authier
- Université Clermont Auvergne, INSERM, CHU, NEURO-DOL Pharmacologie Fondamentale et Clinique de la douleur, F-63000 Clermont-Ferrand, France; Institut ANALGESIA, Faculté de Médecine, F-63000 Clermont-Ferrand, France; Observatoire Français des Médicaments Antalgiques (OFMA), French monitoring centre for analgesic drugs, CHU, F-63000 Clermont-Ferrand, France
| | - Alain Eschalier
- Université Clermont Auvergne, INSERM, CHU, NEURO-DOL Pharmacologie Fondamentale et Clinique de la douleur, F-63000 Clermont-Ferrand, France; Institut ANALGESIA, Faculté de Médecine, F-63000 Clermont-Ferrand, France.
| |
Collapse
|
83
|
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] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/05/2019] [Accepted: 05/30/2019] [Indexed: 01/19/2023]
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
|
84
|
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: 33] [Impact Index Per Article: 6.6] [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
|
85
|
Complementary roles of murine Na V1.7, Na V1.8 and Na V1.9 in acute itch signalling. Sci Rep 2020; 10:2326. [PMID: 32047194 PMCID: PMC7012836 DOI: 10.1038/s41598-020-59092-2] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2019] [Accepted: 01/17/2020] [Indexed: 12/19/2022] Open
Abstract
Acute pruritus occurs in various disorders. Despite severe repercussions on quality of life treatment options remain limited. Voltage-gated sodium channels (NaV) are indispensable for transformation and propagation of sensory signals implicating them as drug targets. Here, NaV1.7, 1.8 and 1.9 were compared for their contribution to itch by analysing NaV-specific knockout mice. Acute pruritus was induced by a comprehensive panel of pruritogens (C48/80, endothelin, 5-HT, chloroquine, histamine, lysophosphatidic acid, trypsin, SLIGRL, β-alanine, BAM8-22), and scratching was assessed using a magnet-based recording technology. We report an unexpected stimulus-dependent diversity in NaV channel-mediated itch signalling. NaV1.7−/− showed substantial scratch reduction mainly towards strong pruritogens. NaV1.8−/− impaired histamine and 5-HT-induced scratching while NaV1.9 was involved in itch signalling towards 5-HT, C48/80 and SLIGRL. Furthermore, similar microfluorimetric calcium responses of sensory neurons and expression of itch-related TRP channels suggest no change in sensory transduction but in action potential transformation and conduction. The cumulative sum of scratching over all pruritogens confirmed a leading role of NaV1.7 and indicated an overall contribution of NaV1.9. Beside the proposed general role of NaV1.7 and 1.9 in itch signalling, scrutiny of time courses suggested NaV1.8 to sustain prolonged itching. Therefore, NaV1.7 and 1.9 may represent targets in pruritus therapy.
Collapse
|
86
|
Ghasemi M, Rajabally YA. Small fiber neuropathy in unexpected clinical settings: a review. Muscle Nerve 2020; 62:167-175. [PMID: 31943276 DOI: 10.1002/mus.26808] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2019] [Revised: 01/07/2020] [Accepted: 01/11/2020] [Indexed: 12/12/2022]
Abstract
Small fiber neuropathy (SFN) is being recognized with increasing frequency in neuromuscular practice due to improved diagnostic techniques. Although there are some common etiologies, up to one-third of cases are considered idiopathic. In recent years, several disorders have unexpectedly been reported in association with SFN, on clinical grounds and complementary investigations, including quantitative sensory testing, intraepidermal nerve fiber density and confocal corneal microscopy. Knowledge of these disorders is important in clinical practice as increased awareness enables prompt diagnosis of SFN in these settings and early optimal therapeutic management of affected patients. Furthermore, these new developments may lead to a better understanding of the pathophysiologic mechanisms underlying SFN in these different disorders as well as, in some cases, an expanded spectrum of affected organs and systems. This article reviews these reported associations, their possible pathophysiologic bases, and the potential resulting management implications.
Collapse
Affiliation(s)
- Majid Ghasemi
- Regional Neuromuscular Service, University Hospitals Birmingham, Birmingham, United Kingdom.,Department of Neurology, School of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Yusuf A Rajabally
- Regional Neuromuscular Service, University Hospitals Birmingham, Birmingham, United Kingdom.,Aston Medical School, Aston University, Birmingham, United Kingdom
| |
Collapse
|
87
|
Ballas SK, Darbari DS. Review/overview of pain in sickle cell disease. Complement Ther Med 2020; 49:102327. [PMID: 32147066 DOI: 10.1016/j.ctim.2020.102327] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2019] [Revised: 01/22/2020] [Accepted: 01/22/2020] [Indexed: 12/28/2022] Open
Abstract
Sickle cell disease (SCD) is a highly complex inherited disorder of hemoglobin structure. Although the molecular lesion is a single-point mutation, the sickle gene is pleiotropic in nature causing multiple phenotypic expressions that constitute the various complications of the disease. Its manifestations could be acute, chronic, nociceptive, neuropathic that could occur singly or in various combinations. Pain continues to be the major factor of SCD phenotypic complications and the most common cause of admissions to the Emergency Department and/or the hospital. Although progress has been made in understanding the pathophysiology of SCD as well as in developing curative therapies such as hematopoietic stem cell transplantation and gene therapy, effective pain management continues to lag behind. Palliative therapies continue to be the major approach to the management of SCD and its complications. The advent of hydroxyurea made partial success in preventing the frequency of vaso-occlusive crises and l-glutamine awaits post-trial confirmation of benefits. The search for additional pharmacotherapeutic agents that could be used singly or in combination with hydroxyurea and/or l-glutamine awaits their dawn hopefully in the near future. The purpose of this review is to describe the various manifestations of SCD, their pathophysiology and their current management. Recent impressive advances in understanding the pathophysiology of pain promise the determination of agents that could replace or minimize the use of opioids.
Collapse
Affiliation(s)
- Samir K Ballas
- Cardeza Foundation for Hematologic Research, Department of Medicine, Sidney Kimmel Medical College, Thomas Jefferson University, 1020 Locust Street, Philadelphia, PA, USA.
| | - Deepika S Darbari
- Division of Hematology, Center for Cancer and Blood Disorders, Children's National Medical Center, Washington, DC, USA
| |
Collapse
|
88
|
Alles SRA, Nascimento F, Luján R, Luiz AP, Millet Q, Bangash MA, Santana-Varela S, Zhou X, Cox JJ, Okorokov AL, Beato M, Zhao J, Wood JN. Sensory neuron-derived Na V1.7 contributes to dorsal horn neuron excitability. SCIENCE ADVANCES 2020; 6:eaax4568. [PMID: 32128393 PMCID: PMC7030926 DOI: 10.1126/sciadv.aax4568] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/25/2019] [Accepted: 12/04/2019] [Indexed: 05/08/2023]
Abstract
Expression of the voltage-gated sodium channel NaV1.7 in sensory neurons is required for pain sensation. We examined the role of NaV1.7 in the dorsal horn of the spinal cord using an epitope-tagged NaV1.7 knock-in mouse. Immuno-electron microscopy showed the presence of NaV1.7 in dendrites of superficial dorsal horn neurons, despite the absence of mRNA. Rhizotomy of L5 afferent nerves lowered the levels of NaV1.7 in the dorsal horn. Peripheral nervous system-specific NaV1.7 null mutant mice showed central deficits, with lamina II dorsal horn tonic firing neurons more than halved and single spiking neurons more than doubled. NaV1.7 blocker PF05089771 diminished excitability in dorsal horn neurons but had no effect on NaV1.7 null mutant mice. These data demonstrate an unsuspected functional role of primary afferent neuron-generated NaV1.7 in dorsal horn neurons and an expression pattern that would not be predicted by transcriptomic analysis.
Collapse
Affiliation(s)
- Sascha R. A. Alles
- Molecular Nociception Group, Wolfson Institute for Biomedical Research, University College London, London WC1E 6BT, UK
| | - Filipe Nascimento
- Department of Neuroscience, Physiology and Pharmacology, University College London, London WC1E 6BT, UK
| | - Rafael Luján
- Synaptic Structure Laboratory, Instituto de Investigación en Discapacidades Neurológicas (IDINE), Department Ciencias Médicas, Facultad de Medicina, Universidad Castilla-La Mancha, Campus Biosanitario, C/Almansa 14, 02008 Albacete, Spain
| | - Ana P. Luiz
- Molecular Nociception Group, Wolfson Institute for Biomedical Research, University College London, London WC1E 6BT, UK
| | - Queensta Millet
- Molecular Nociception Group, Wolfson Institute for Biomedical Research, University College London, London WC1E 6BT, UK
| | - M. Ali Bangash
- Molecular Nociception Group, Wolfson Institute for Biomedical Research, University College London, London WC1E 6BT, UK
| | - Sonia Santana-Varela
- Molecular Nociception Group, Wolfson Institute for Biomedical Research, University College London, London WC1E 6BT, UK
| | - Xuelong Zhou
- Molecular Nociception Group, Wolfson Institute for Biomedical Research, University College London, London WC1E 6BT, UK
- Department of Anesthesiology, The First Hospital of Nanjing Medical University, Nanjing, Jiangsu 210029, China
| | - James J. Cox
- Molecular Nociception Group, Wolfson Institute for Biomedical Research, University College London, London WC1E 6BT, UK
| | - Andrei L. Okorokov
- Molecular Nociception Group, Wolfson Institute for Biomedical Research, University College London, London WC1E 6BT, UK
| | - Marco Beato
- Department of Neuroscience, Physiology and Pharmacology, University College London, London WC1E 6BT, UK
- Corresponding author. (M.B.); (J.Z.); (J.N.W.)
| | - Jing Zhao
- Molecular Nociception Group, Wolfson Institute for Biomedical Research, University College London, London WC1E 6BT, UK
- Corresponding author. (M.B.); (J.Z.); (J.N.W.)
| | - John N. Wood
- Molecular Nociception Group, Wolfson Institute for Biomedical Research, University College London, London WC1E 6BT, UK
- Corresponding author. (M.B.); (J.Z.); (J.N.W.)
| |
Collapse
|
89
|
Woolf CJ. Capturing Novel Non-opioid Pain Targets. Biol Psychiatry 2020; 87:74-81. [PMID: 31399256 PMCID: PMC6898770 DOI: 10.1016/j.biopsych.2019.06.017] [Citation(s) in RCA: 65] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/01/2019] [Revised: 06/07/2019] [Accepted: 06/24/2019] [Indexed: 12/29/2022]
Abstract
The relatively high efficacy of opioids, which have associated risks of addiction, tolerance, and dependence, for the management of acute and terminal pain has been a major driver of the opioid crisis, together with the availability, overprescription, and diversion of these drugs. Eliminating opioids without an effective replacement is, however, no solution, as it substitutes one major problem with another. To deal successfully with the opioid crisis, we need to discover novel analgesics whose mechanisms do not involve the mu opioid receptor but that have high analgesic potency and low risk of adverse effects, particularly no abuse liability. The question is how to achieve this. There are several necessary elements; first, we need to understand the nature of pain and the mechanisms responsible for it, and second, we need to adopt novel and unbiased approaches to the identification and validation of pain targets.
Collapse
Affiliation(s)
- Clifford J Woolf
- F.M. Kirby Neurobiology Center, Boston Children's Hospital and Harvard Medical School, Boston, Massachusetts.
| |
Collapse
|
90
|
Neff RA, Flinspach M, Gibbs A, Shih AY, Minassian NA, Liu Y, Fellows R, Libiger O, Young S, Pennington MW, Hunter MJ, Wickenden AD. Comprehensive engineering of the tarantula venom peptide huwentoxin-IV to inhibit the human voltage-gated sodium channel hNav1.7. J Biol Chem 2020. [DOI: 10.1016/s0021-9258(17)49888-7] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/22/2022] Open
|
91
|
Neff RA, Flinspach M, Gibbs A, Shih AY, Minassian NA, Liu Y, Fellows R, Libiger O, Young S, Pennington MW, Hunter MJ, Wickenden AD. Comprehensive engineering of the tarantula venom peptide huwentoxin-IV to inhibit the human voltage-gated sodium channel hNa v1.7. J Biol Chem 2019; 295:1315-1327. [PMID: 31871053 DOI: 10.1074/jbc.ra119.011318] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2019] [Revised: 12/20/2019] [Indexed: 02/04/2023] Open
Abstract
Pain is a significant public health burden in the United States, and current treatment approaches rely heavily on opioids, which often have limited efficacy and can lead to addiction. In humans, functional loss of the voltage-gated sodium channel Nav1.7 leads to pain insensitivity without deficits in the central nervous system. Accordingly, discovery of a selective Nav1.7 antagonist should provide an analgesic without abuse liability and an improved side-effect profile. Huwentoxin-IV, a component of tarantula venom, potently blocks sodium channels and is an attractive scaffold for engineering a Nav1.7-selective molecule. To define the functional impact of alterations in huwentoxin-IV sequence, we produced a library of 373 point mutants and tested them for Nav1.7 and Nav1.2 activity. We then combined favorable individual changes to produce combinatorial mutants that showed further improvements in Nav1.7 potency (E1N, E4D, Y33W, Q34S-Nav1.7 pIC50 = 8.1 ± 0.08) and increased selectivity over other Nav isoforms (E1N, R26K, Q34S, G36I, Nav1.7 pIC50 = 7.2 ± 0.1, Nav1.2 pIC50 = 6.1 ± 0.18, Nav1.3 pIC50 = 6.4 ± 1.0), Nav1.4 is inactive at 3 μm, and Nav1.5 is inactive at 10 μm We also substituted noncoded amino acids at select positions in huwentoxin-IV. Based on these results, we identify key determinants of huwentoxin's Nav1.7 inhibition and propose a model for huwentoxin-IV's interaction with Nav1.7. These findings uncover fundamental features of huwentoxin involved in Nav1.7 blockade, provide a foundation for additional optimization of this molecule, and offer a basis for the development of a safe and effective analgesic.
Collapse
Affiliation(s)
- Robert A Neff
- Neuroscience Discovery, Janssen Research and Development, LLC, San Diego, California 92121
| | - Mack Flinspach
- Biologics Research, Janssen Research and Development, LLC, San Diego, California 92121
| | - Alan Gibbs
- Structural Biology, Janssen Research and Development, LLC, Spring House, Pennsylvania 19477
| | - Amy Y Shih
- Discovery Chemistry-Computational Chemistry, Janssen Research and Development, LLC, San Diego, California 92121
| | - Natali A Minassian
- Neuroscience Discovery, Janssen Research and Development, LLC, San Diego, California 92121
| | - Yi Liu
- Neuroscience Discovery, Janssen Research and Development, LLC, San Diego, California 92121
| | - Ross Fellows
- Biologics Research, Janssen Research and Development, LLC, San Diego, California 92121
| | - Ondrej Libiger
- Translational Medicine and Early Development Statistics, Janssen Research and Development, LLC, San Diego, California 92121
| | - Stephanie Young
- Translational Medicine and Early Development Statistics, Janssen Research and Development, LLC, San Diego, California 92121
| | | | - Michael J Hunter
- Biologics Research, Janssen Research and Development, LLC, San Diego, California 92121
| | - Alan D Wickenden
- Molecular and Cellular Pharmacology, Janssen Research and Development, LLC, San Diego, California 92121
| |
Collapse
|
92
|
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
|
93
|
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
|
94
|
Siebenga P, van Amerongen G, Hay JL, McDonnell A, Gorman D, Butt R, Groeneveld GJ. Lack of Detection of the Analgesic Properties of PF-05089771, a Selective Na v 1.7 Inhibitor, Using a Battery of Pain Models in Healthy Subjects. Clin Transl Sci 2019; 13:318-324. [PMID: 31642607 PMCID: PMC7070789 DOI: 10.1111/cts.12712] [Citation(s) in RCA: 37] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2019] [Accepted: 09/06/2019] [Indexed: 12/12/2022] Open
Abstract
Sodium channel blockers are used for the treatment of pain, but this is limited by the lack of selectivity for different sodium channel subtypes, which can result in central nervous system and cardiovascular side effects. As such, there is special interest in the Nav 1.7 subtype, which is expressed predominantly in nociceptive and sympathetic neurons. The aim was to demonstrate analgesic properties of a potent selective Nav 1.7 sodium channel blocker, PF-05089771, alone and concomitantly with pregabalin in healthy subjects using a battery of human evoked pain models. This was a double-blind, double-dummy, randomized, placebo-controlled, five-period cross-over study with PF-05089771 alone and PF-05089771 concomitantly with pregabalin as treatment arms with pregabalin, ibuprofen, and placebo as control arms (NCT02349607). A battery of human evoked pain models was used to investigate analgesic properties of PF-05089771. Twenty-five subjects were enrolled in the study of which 23 subjects completed all five periods. PF-05089771 alone did not differ from placebo on the primary pain end points. The same holds when comparing PF-05089771 concomitantly with pregabalin and pregabalin alone. Pregabalin showed significant effects relative to placebo on thermal pain on the normal skin and UVB skin (least squares means with 90% confidence interval: 0.63 (0.32-0.93) and 0.53 (0.11-0.96)), pressure stimulation (1.10 (1.04-1.18)), and cold pressor (1.22 (1.14-1.32)). Ibuprofen demonstrated significant effects on thermal pain UVB skin (1.26 (0.82-1.70)) and pressure stimulation assessment (1.08 (1.01-1.15)), consistent with historical results. This study did not demonstrate analgesic properties of PF-05089771 alone or concomitantly with pregabalin in a battery of pain models.
Collapse
Affiliation(s)
| | | | - Justin L Hay
- Centre for Human Drug Research, Leiden, The Netherlands
| | | | - Donal Gorman
- Neuroscience and Pain Research Unit, Pfizer WRD, Cambridge, UK
| | - Richard Butt
- Neuroscience and Pain Research Unit, Pfizer WRD, Cambridge, UK
| | - Geert Jan Groeneveld
- Centre for Human Drug Research, Leiden, The Netherlands.,Leiden University Medical Centre, Leiden, The Netherlands
| |
Collapse
|
95
|
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
|
96
|
Kong DJ, Wang Y, Wang HX, Wang MX, Wang J, Cheng MS. Molecular determinants for ligand binding at Nav1.4 and Nav1.7 channels: Experimental affinity results analyzed by molecular modeling. Comput Biol Chem 2019; 83:107132. [PMID: 31563636 DOI: 10.1016/j.compbiolchem.2019.107132] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2019] [Revised: 09/11/2019] [Accepted: 09/18/2019] [Indexed: 12/16/2022]
Abstract
Here, we focused on exploring the selectivity mechanism against Nav1.7 over Nav1.4 due to different binding modes of two selected inhibitors. By the superposition of Nav1.7 and Nav1.4 proteins, we selected the most homologous chain of Nav1.7 with Nav1.4, defining the active site of Nav1.4-VSD4 based on the aryl sulfonamide binding site of Nav1.7-VSD4. Comparison of the conformations exhibited by Tyr1386 (Nav1.4) and Tyr1537 (Nav1.7) suggested that the steric hindrance caused by Tyr1386 owned primary influence on inhibition selectivity, which was further verified through molecular docking and MD simulation of two representative inhibitors. Our finding would be helpful for discovery of selective Nav1.7 inhibitors over Nav1.4.
Collapse
Affiliation(s)
- De-Jiang Kong
- Key Laboratory of Structure-Based Drug Design & Discovery of Ministry of Education, School of Pharmaceutical Engineering, Shenyang Pharmaceutical University, Shenyang 110016, PR China
| | - Ying Wang
- Key Laboratory of Structure-Based Drug Design & Discovery of Ministry of Education, School of Pharmaceutical Engineering, Shenyang Pharmaceutical University, Shenyang 110016, PR China
| | - Han-Xun Wang
- Key Laboratory of Structure-Based Drug Design & Discovery of Ministry of Education, School of Pharmaceutical Engineering, Shenyang Pharmaceutical University, Shenyang 110016, PR China
| | - Ming-Xing Wang
- Key Laboratory of Structure-Based Drug Design & Discovery of Ministry of Education, School of Pharmaceutical Engineering, Shenyang Pharmaceutical University, Shenyang 110016, PR China
| | - Jian Wang
- Key Laboratory of Structure-Based Drug Design & Discovery of Ministry of Education, School of Pharmaceutical Engineering, Shenyang Pharmaceutical University, Shenyang 110016, PR China.
| | - Mao-Sheng Cheng
- Key Laboratory of Structure-Based Drug Design & Discovery of Ministry of Education, School of Pharmaceutical Engineering, Shenyang Pharmaceutical University, Shenyang 110016, PR China
| |
Collapse
|
97
|
Ciotu CI, Tsantoulas C, Meents J, Lampert A, McMahon SB, Ludwig A, Fischer MJM. Noncanonical Ion Channel Behaviour in Pain. Int J Mol Sci 2019; 20:E4572. [PMID: 31540178 PMCID: PMC6770626 DOI: 10.3390/ijms20184572] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2019] [Revised: 09/09/2019] [Accepted: 09/12/2019] [Indexed: 12/19/2022] Open
Abstract
Ion channels contribute fundamental properties to cell membranes. Although highly diverse in conductivity, structure, location, and function, many of them can be regulated by common mechanisms, such as voltage or (de-)phosphorylation. Primarily considering ion channels involved in the nociceptive system, this review covers more novel and less known features. Accordingly, we outline noncanonical operation of voltage-gated sodium, potassium, transient receptor potential (TRP), and hyperpolarization-activated cyclic nucleotide (HCN)-gated channels. Noncanonical features discussed include properties as a memory for prior voltage and chemical exposure, alternative ion conduction pathways, cluster formation, and silent subunits. Complementary to this main focus, the intention is also to transfer knowledge between fields, which become inevitably more separate due to their size.
Collapse
Affiliation(s)
- Cosmin I Ciotu
- Center for Physiology and Pharmacology, Medical University of Vienna, 1090 Vienna, Austria
| | | | - Jannis Meents
- Institute of Physiology, University Hospital RWTH Aachen, 52074 Aachen, Germany
| | - Angelika Lampert
- Institute of Physiology, University Hospital RWTH Aachen, 52074 Aachen, Germany
| | - Stephen B McMahon
- Wolfson Centre for Age-Related Diseases, King's College London, London SE1 1UR, UK
| | - Andreas Ludwig
- Institute of Experimental and Clinical Pharmacology and Toxicology, Friedrich-Alexander-Universität Erlangen-Nürnberg, 91054 Erlangen, Germany
| | - Michael J M Fischer
- Center for Physiology and Pharmacology, Medical University of Vienna, 1090 Vienna, Austria.
| |
Collapse
|
98
|
Ma RSY, Kayani K, Whyte-Oshodi D, Whyte-Oshodi A, Nachiappan N, Gnanarajah S, Mohammed R. Voltage gated sodium channels as therapeutic targets for chronic pain. J Pain Res 2019; 12:2709-2722. [PMID: 31564962 PMCID: PMC6743634 DOI: 10.2147/jpr.s207610] [Citation(s) in RCA: 47] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2019] [Accepted: 08/02/2019] [Indexed: 01/23/2023] Open
Abstract
Being maladaptive and frequently unresponsive to pharmacotherapy, chronic pain presents a major unmet clinical need. While an intact central nervous system is required for conscious pain perception, nociceptor hyperexcitability induced by nerve injury in the peripheral nervous system (PNS) is sufficient and necessary to initiate and maintain neuropathic pain. The genesis and propagation of action potentials is dependent on voltage-gated sodium channels, in particular, Nav1.7, Nav1.8 and Nav1.9. However, nerve injury triggers changes in their distribution, expression and/or biophysical properties, leading to aberrant excitability. Most existing treatment for pain relief acts through non-selective, state-dependent sodium channel blockage and have narrow therapeutic windows. Natural toxins and developing subtype-specific and molecular-specific sodium channel blockers show promise for treatment of neuropathic pain with minimal side effects. New approaches to analgesia include combination therapy and gene therapy. Here, we review how individual sodium channel subtypes contribute to pain, and the attempts made to develop more effective analgesics for the treatment of chronic pain.
Collapse
Affiliation(s)
- Renee Siu Yu Ma
- Department of Medicine, University of Cambridge, Cambridge, UK
| | - Kayani Kayani
- Department of Medicine, University of Cambridge, Cambridge, UK
| | | | | | | | | | - Raihan Mohammed
- Department of Medicine, University of Cambridge, Cambridge, UK
| |
Collapse
|
99
|
Vysokov N, McMahon SB, Raouf R. The role of Na V channels in synaptic transmission after axotomy in a microfluidic culture platform. Sci Rep 2019; 9:12915. [PMID: 31501450 PMCID: PMC6733904 DOI: 10.1038/s41598-019-49214-w] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2018] [Accepted: 08/20/2019] [Indexed: 01/07/2023] Open
Abstract
Voltage gated sodium channels are key players in aberrant pain signaling and sensitization of nociceptors after peripheral nerve injury. The extent to which sodium channel activity after injury contributes to synaptic transmission at the first pain synapse however remains unclear. To investigate the effect of axotomy on synaptic transmission between dorsal root ganglia neurons and dorsal horn neurons, we reconstructed the first pain synapse in a novel microfluidic based compartmentalized cell culture system, which recapitulates the connectivity of peripheral pain signaling. We show that following axotomy of the distal axons, inhibition of NaV1.7 and NaV1.8 sodium channels in incoming presynaptic DRG axons is no longer sufficient to block activation of these axons and the resulting synaptic transmission to dorsal horn neurons. We found that blockade of NaV1.6 activity is highly effective in reducing activation of incoming axons contributing to synaptic transmission after axotomy of DRG neurons. The microfluidic culture system described here offers an in vitro platform to recapitulate and study the first pain synapse.
Collapse
Affiliation(s)
- Nickolai Vysokov
- Wolfson Centre for Age-Related Diseases, Institute of Psychiatry, Psychology & Neuroscience, King's College London, London, SE1 1UL, United Kingdom
| | - Stephen B McMahon
- Wolfson Centre for Age-Related Diseases, Institute of Psychiatry, Psychology & Neuroscience, King's College London, London, SE1 1UL, United Kingdom
| | - Ramin Raouf
- Wolfson Centre for Age-Related Diseases, Institute of Psychiatry, Psychology & Neuroscience, King's College London, London, SE1 1UL, United Kingdom.
| |
Collapse
|
100
|
Yin S, Liu J, Kang Y, Lin Y, Li D, Shao L. Interactions of nanomaterials with ion channels and related mechanisms. Br J Pharmacol 2019; 176:3754-3774. [PMID: 31290152 DOI: 10.1111/bph.14792] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2018] [Revised: 06/10/2019] [Accepted: 07/01/2019] [Indexed: 12/15/2022] Open
Abstract
The pharmacological potential of nanotechnology, especially in drug delivery and bioengineering, has developed rapidly in recent decades. Ion channels, which are easily targeted by external agents, such as nanomaterials (NMs) and synthetic drugs, due to their unique structures, have attracted increasing attention in the fields of nanotechnology and pharmacology for the treatment of ion channel-related diseases. NMs have significant effects on ion channels, and these effects are manifested in many ways, including changes in ion currents, kinetic characteristics and channel distribution. Subsequently, intracellular ion homeostasis, signalling pathways, and intracellular ion stores are affected, leading to the initiation of a range of biological processes. However, the effect of the interactions of NMs with ion channels is an interesting topic that remains obscure. In this review, we have summarized the recent research progress on the direct and indirect interactions between NMs and ion channels and discussed the related molecular mechanisms, which are crucial to the further development of ion channel-related nanotechnological applications.
Collapse
Affiliation(s)
- Suhan Yin
- Nanfang Hospital, Southern Medical University, Guangzhou, China.,Guangdong Provincial Key Laboratory of Construction and Detection in Tissue Engineering, Biomaterials Research Center, School of Biomedical Engineering, Southern Medical University, Guangzhou, 510515, China
| | - Jia Liu
- Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Yiyuan Kang
- Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Yuqing Lin
- Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Dongjian Li
- Liwan District Stomatology Hospital, Guangzhou, China
| | - Longquan Shao
- Nanfang Hospital, Southern Medical University, Guangzhou, China.,Guangdong Provincial Key Laboratory of Construction and Detection in Tissue Engineering, Biomaterials Research Center, School of Biomedical Engineering, Southern Medical University, Guangzhou, 510515, China
| |
Collapse
|