1
|
Ruiz-Castelan JE, Villa-Díaz F, Castro ME, Melendez FJ, Scior T. The α/β3 complex of human voltage-gated sodium channel hNa v1.7 to study mechanistic differences in presence and absence of auxiliary subunit β3. J Mol Model 2025; 31:168. [PMID: 40397258 PMCID: PMC12095431 DOI: 10.1007/s00894-025-06378-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2024] [Accepted: 04/25/2025] [Indexed: 05/22/2025]
Abstract
CONTEXT In the context of structural interactomics, we generated a 3D model between α and β3 subunits for the hitherto unknown human voltage-gated sodium channel complex (hNa 1.7α/β3). We embedded our 3D model in a membrane lipid bilayer for molecular dynamics (MD) simulations of the sodium cation passage from the outer vestibule through the inner pore segment of our hNa 1.7 complex in presence and absence of auxiliary subunit β3 with remarkable changes close to electrophysiological study results. A complete passage could not be expected due to because the inactivated state of the underlying 3D template. A complete sodium ion passage would require an open state of the channel. The computed observations concerning side chain rearrangements for favorable cooperativity under evolutionary neighborhood conditions, favorable and unfavorable amino acid interactions, proline kink, loop, and helix displacements were all found in excellent keeping with the extant literature without any exception nor contradiction. Complex-stabilizing pairs of interacting amino acids with evolutionary neighborhood complementary were identified. METHODS The following tools were used: sequence search and alignment by FASTA and Clustal Omega; 3D model visualization and homology modeling by Vega ZZ, SPDBV, Chimera and Modeller, respectively; missing sections (loops) by Alphafold; geometry optimization prior to MD runs by GROMACS 2021.4 under the CHARMM 36 force field; local healing of bad contacts by SPDBV based on its Ramachandran plots; protein-protein docking by HDOCK 2.4; membrane insertion assisted by OPM; Berendsen V-rescaling for NVT; Parrinello-Rahman and Nose-Hoover for MPT; MD analyses by VMD and XMGRACE.
Collapse
Grants
- 100256733-VIEP2024 Vicerrectoría de Investigación y Estudios de Posgrado (VIEP-BUAP, Mexico)
- 100256733-VIEP2024 Vicerrectoría de Investigación y Estudios de Posgrado (VIEP-BUAP, Mexico)
- 100256733-VIEP2024 Vicerrectoría de Investigación y Estudios de Posgrado (VIEP-BUAP, Mexico)
- 100256733-VIEP2024 Vicerrectoría de Investigación y Estudios de Posgrado (VIEP-BUAP, Mexico)
- 100256733-VIEP2024 Vicerrectoría de Investigación y Estudios de Posgrado (VIEP-BUAP, Mexico)
- BUAP-CA-263 PRODEP Academic Group (SEP, Mexico)
- BUAP-CA-263 PRODEP Academic Group (SEP, Mexico)
- BUAP-CA-263 PRODEP Academic Group (SEP, Mexico)
- BUAP-CA-263 PRODEP Academic Group (SEP, Mexico)
- BUAP-CA-263 PRODEP Academic Group (SEP, Mexico)
Collapse
Affiliation(s)
| | - Fernando Villa-Díaz
- Laboratory of Basical Science, Tecnologico Nacional de Mexico, Campus Guaymas, C.P. 85480, Sonora, Mexico
| | | | - Francisco J Melendez
- Laboratory of Theoretical Chemistry, Faculty of Chemical Sciences, BUAP, C.P. 72570, Puebla, Mexico.
| | - Thomas Scior
- Laboratory of Computational Molecular Simulations, Faculty of Chemical Sciences, BUAP, C.P. 72570, Puebla, Mexico.
| |
Collapse
|
2
|
Liu Y, Yuan Z, He H, Liu H, Wu Y, Xue S, Diao Z, Qiao H. TTX-R and TTX-S Sodium Channels in CGRP-Positive Dorsal Root Ganglia Neurons Mediate Referred Somatic Hyperalgesia in Ulcerative Colitis Mice. Neurogastroenterol Motil 2025:e70051. [PMID: 40273371 DOI: 10.1111/nmo.70051] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/15/2024] [Revised: 03/11/2025] [Accepted: 03/31/2025] [Indexed: 04/26/2025]
Abstract
BACKGROUND Ulcerative colitis (UC) frequently co-exists with referred somatic hyperalgesia in clinical presentations. However, the peripheral neurophysiological mechanisms of visceral referred pain remain unclear. This study aimed to clarify the neurobiological mechanisms that underpin the referred somatic hyperalgesia associated with UC. METHODS A UC mouse model was constructed via the administration of dextran sulfate sodium (DSS). Referred somatic regions in these mice were identified by measuring the number of Evans blue extravasations and pain threshold levels. Electrophysiological and immunofluorescent staining approaches were applied to evaluate the alterations in kinetic properties and expression of TTX-R (Nav1.8) and TTX-S (Nav1.7) channels in calcitonin gene-related peptide (CGRP)-positive dorsal root ganglion (DRG) neurons in the referred regions. Pharmacological methods were utilized to elucidate the necessary role of the Nav1.8 and Nav1.7 channels in somatic referred hyperalgesia. KEY RESULTS Oral administration of DSS to mice for 7 days resulted in significant colon damage, neurogenic inflammation, and referred somatic hyperalgesia. The mechanisms underlying these effects may involve the activation of TTX-R and TTX-S channels, and the upregulation of co-expressed Nav1.8 and Nav1.7 with CGRP, resulting in an increased excitability of CGRP+ DRG neurons in sensitized regions. Selectively inhibiting either Nav1.8 or Nav1.7 channels could mitigate the referred somatic hyperalgesia induced by DSS. CONCLUSIONS AND INFERENCES The functional alterations in Nav1.8 and Nav1.7 channels within CGRP+ DRG neurons are pivotal to the development of neurogenic inflammation and referred somatic hyperalgesia. These findings lay a foundation for exploring novel therapeutic targets to relieve visceral referred pain.
Collapse
Affiliation(s)
- Yongbin Liu
- Shaanxi Key Laboratory of Integrative Acupuncture and Medicine, Shaanxi University of Chinese Medicine, Xianyang, Shaanxi Province, China
- Key Laboratory of Acupuncture and Neurobiology, Shaanxi Administration of Traditional Chinese Medicine, Xianyang, Shaanxi Province, China
- College of Acupuncture-Moxibustion and Tuina, Shaanxi University of Chinese Medicine, Xianyang, Shaanxi Province, China
| | - Ziyan Yuan
- Shaanxi Key Laboratory of Integrative Acupuncture and Medicine, Shaanxi University of Chinese Medicine, Xianyang, Shaanxi Province, China
- Key Laboratory of Acupuncture and Neurobiology, Shaanxi Administration of Traditional Chinese Medicine, Xianyang, Shaanxi Province, China
- College of Acupuncture-Moxibustion and Tuina, Shaanxi University of Chinese Medicine, Xianyang, Shaanxi Province, China
| | - Hongzhou He
- Shaanxi Key Laboratory of Integrative Acupuncture and Medicine, Shaanxi University of Chinese Medicine, Xianyang, Shaanxi Province, China
- Key Laboratory of Acupuncture and Neurobiology, Shaanxi Administration of Traditional Chinese Medicine, Xianyang, Shaanxi Province, China
- College of Acupuncture-Moxibustion and Tuina, Shaanxi University of Chinese Medicine, Xianyang, Shaanxi Province, China
| | - Huanhuan Liu
- Shaanxi Key Laboratory of Integrative Acupuncture and Medicine, Shaanxi University of Chinese Medicine, Xianyang, Shaanxi Province, China
- Key Laboratory of Acupuncture and Neurobiology, Shaanxi Administration of Traditional Chinese Medicine, Xianyang, Shaanxi Province, China
- College of Acupuncture-Moxibustion and Tuina, Shaanxi University of Chinese Medicine, Xianyang, Shaanxi Province, China
| | - Yuwei Wu
- College of Acupuncture-Moxibustion and Tuina, Shaanxi University of Chinese Medicine, Xianyang, Shaanxi Province, China
| | - Simeng Xue
- Shaanxi Key Laboratory of Integrative Acupuncture and Medicine, Shaanxi University of Chinese Medicine, Xianyang, Shaanxi Province, China
| | - Zhijun Diao
- Shaanxi Key Laboratory of Integrative Acupuncture and Medicine, Shaanxi University of Chinese Medicine, Xianyang, Shaanxi Province, China
- Key Laboratory of Acupuncture and Neurobiology, Shaanxi Administration of Traditional Chinese Medicine, Xianyang, Shaanxi Province, China
| | - Haifa Qiao
- Shaanxi Key Laboratory of Integrative Acupuncture and Medicine, Shaanxi University of Chinese Medicine, Xianyang, Shaanxi Province, China
- Key Laboratory of Acupuncture and Neurobiology, Shaanxi Administration of Traditional Chinese Medicine, Xianyang, Shaanxi Province, China
- Shaanxi Collaborative Innovation Center of TCM Technologies and Devices, Shaanxi University of Chinese Medicine, Xianyang, Shaanxi Province, China
| |
Collapse
|
3
|
Antunes A, Montnach J, Khakh K, Lopez L, Thomas B, Ribeiro Oliveira-Mendes B, Jaquillard L, Servent D, Béroud R, Cohen CJ, Benoit E, De Waard M. The venom of Cyriopagopus schmidti spider contains a natural huwentoxin-IV analogue with unexpected improved analgesic potential. Front Pharmacol 2025; 16:1566312. [PMID: 40276610 PMCID: PMC12019880 DOI: 10.3389/fphar.2025.1566312] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2025] [Accepted: 03/07/2025] [Indexed: 04/26/2025] Open
Abstract
The venom of Cyriopagopus schmidti spider has been extensively investigated, thereby allowing the identification of numerous new natural peptides. Many of these peptides are active on ion channels and several of them occur from post-translational processing. In order to further identify new entities, we screened this venom against five different human voltage-gated sodium (hNav) channels. We illustrate the unusual richness of this venom in targeting this wide variety of hNav channels. We confirm the identity of previously discovered peptides active on these ion channels type (huwentoxin (HwTx)-I, HwTx-II and HwTx-IV), indicating the efficacy of the screening process by automated patch-clamp. We also identified a novel analogue of HwTx-IV that differs by the absence of amidation and the presence of an extra C-terminal Gly residue. Interestingly, this analogue is less potent than HwTx-IV itself in blocking hNav1.7 in cell lines, but turns out to be significantly more potent in TTX-sensitive dorsal root ganglia neurons. Because of this unexpected finding, this novel analogue turns out to be a more potent analgesic than HwTx-IV itself without presenting most of the Nav1.6-related toxic effects of HwTx-IV.
Collapse
Affiliation(s)
- Aurélie Antunes
- Université Paris-Saclay, CEA, Département Médicaments et Technologies pour la Santé (DMTS), Service d’Ingénierie Moléculaire pour la Santé (SIMoS), EMR Centre National de la Recherche Scientifique, Gif-sur-Yvette, France
- Smartox Biotechnology, Saint-Egrève, France
- Nantes Université, Centre National de la Recherche Scientifique, Institut National de la Santé et de la Recherche Médicale, l’institut du thorax, Nantes, France
| | - Jérôme Montnach
- Nantes Université, Centre National de la Recherche Scientifique, Institut National de la Santé et de la Recherche Médicale, l’institut du thorax, Nantes, France
| | - Kuldip Khakh
- Department of Biology, Xenon Pharmaceuticals, Burnaby, BC, Canada
| | - Ludivine Lopez
- Nantes Université, Centre National de la Recherche Scientifique, Institut National de la Santé et de la Recherche Médicale, l’institut du thorax, Nantes, France
| | | | - Barbara Ribeiro Oliveira-Mendes
- Nantes Université, Centre National de la Recherche Scientifique, Institut National de la Santé et de la Recherche Médicale, l’institut du thorax, Nantes, France
| | | | - Denis Servent
- Université Paris-Saclay, CEA, Département Médicaments et Technologies pour la Santé (DMTS), Service d’Ingénierie Moléculaire pour la Santé (SIMoS), EMR Centre National de la Recherche Scientifique, Gif-sur-Yvette, France
| | - Rémy Béroud
- Smartox Biotechnology, Saint-Egrève, France
- Smartbioscience-Peptide, Saint-Egrève, France
| | - Charles J. Cohen
- Department of Biology, Xenon Pharmaceuticals, Burnaby, BC, Canada
| | - Evelyne Benoit
- Université Paris-Saclay, CEA, Département Médicaments et Technologies pour la Santé (DMTS), Service d’Ingénierie Moléculaire pour la Santé (SIMoS), EMR Centre National de la Recherche Scientifique, Gif-sur-Yvette, France
| | - Michel De Waard
- Smartox Biotechnology, Saint-Egrève, France
- Nantes Université, Centre National de la Recherche Scientifique, Institut National de la Santé et de la Recherche Médicale, l’institut du thorax, Nantes, France
- LabEx “Ion Channels, Science and Therapeutics”, Valbonne, France
| |
Collapse
|
4
|
Yang J, Xie YF, Smith R, Ratté S, Prescott SA. Discordance between preclinical and clinical testing of Na V 1.7-selective inhibitors for pain. Pain 2025; 166:481-501. [PMID: 39928833 PMCID: PMC11808711 DOI: 10.1097/j.pain.0000000000003425] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2024] [Revised: 07/19/2024] [Accepted: 08/13/2024] [Indexed: 10/26/2024]
Abstract
ABSTRACT The voltage-gated sodium channel Na V 1.7 plays an important role in pain processing according to genetic data. Those data made Na V 1.7 a popular drug target, especially since its relatively selective expression in nociceptors promised pain relief without the adverse effects associated with broader sodium channel blockade. Despite encouraging preclinical data in rodents, Na V 1.7-selective inhibitors have not yet proven effective in clinical trials. Discrepancies between preclinical and clinical results should raise alarms. We reviewed preclinical and clinical reports on the analgesic efficacy of Na V 1.7-selective inhibitors and found critical differences in several factors. Putting aside species differences, most preclinical studies tested young male rodents with limited genetic variability, inconsistent with the clinical population. Inflammatory pain was the most common preclinical chronic pain model whereas nearly all clinical trials focused on neuropathic pain despite some evidence suggesting Na V 1.7 channels are not essential for neuropathic pain. Preclinical studies almost exclusively measured evoked pain whereas most clinical trials assessed average pain intensity without distinguishing between evoked and spontaneous pain. Nearly all preclinical studies gave a single dose of drug unlike the repeat dosing used clinically, thus precluding preclinical data from demonstrating whether tolerance or other slow processes occur. In summary, preclinical testing of Na V 1.7-selective inhibitors aligned poorly with clinical testing. Beyond issues that have already garnered widespread attention in the pain literature, our results highlight the treatment regimen and choice of pain model as areas for improvement.
Collapse
Affiliation(s)
- Jane Yang
- Neurosciences and Mental Health, The Hospital for Sick Children, Toronto, ON, Canada
- Institute of Biomedical Engineering, University of Toronto, Toronto, ON, Canada
| | - Yu-Feng Xie
- Neurosciences and Mental Health, The Hospital for Sick Children, Toronto, ON, Canada
| | - Russell Smith
- Neurosciences and Mental Health, The Hospital for Sick Children, Toronto, ON, Canada
| | - Stéphanie Ratté
- Neurosciences and Mental Health, The Hospital for Sick Children, Toronto, ON, Canada
| | - Steven A. Prescott
- Neurosciences and Mental Health, The Hospital for Sick Children, Toronto, ON, Canada
- Institute of Biomedical Engineering, University of Toronto, Toronto, ON, Canada
- Department of Physiology, University of Toronto, Toronto, ON, Canada
| |
Collapse
|
5
|
Shehab S, Hamad MIK, Emerald BS. A novel approach to completely alleviate peripheral neuropathic pain in human patients: insights from preclinical data. Front Neuroanat 2025; 18:1523095. [PMID: 39839257 PMCID: PMC11747518 DOI: 10.3389/fnana.2024.1523095] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2024] [Accepted: 12/23/2024] [Indexed: 01/23/2025] Open
Abstract
Neuropathic pain is a pervasive health concern worldwide, posing significant challenges to both clinicians and neuroscientists. While acute pain serves as a warning signal for potential tissue damage, neuropathic pain represents a chronic pathological condition resulting from injury or disease affecting sensory pathways of the nervous system. Neuropathic pain is characterized by long-lasting ipsilateral hyperalgesia (increased sensitivity to pain), allodynia (pain sensation in response to stimuli that are not normally painful), and spontaneous unprovoked pain. Current treatments for neuropathic pain are generally inadequate, and prevention remains elusive. In this review, we provide an overview of current treatments, their limitations, and a discussion on the potential of capsaicin and its analog, resiniferatoxin (RTX), for complete alleviation of nerve injury-induced neuropathic pain. In an animal model of neuropathic pain where the fifth lumbar (L5) spinal nerve is unilaterally ligated and cut, resulting in ipsilateral hyperalgesia, allodynia, and spontaneous pain akin to human neuropathic pain. The application of capsaicin or RTX to the adjacent uninjured L3 and L4 nerves completely alleviated and prevented mechanical and thermal hyperalgesia following the L5 nerve injury. The effects of this treatment were specific to unmyelinated fibers (responsible for pain sensation), while thick myelinated nerve fibers (responsible for touch and mechanoreceptor sensations) remained intact. Here, we propose to translate these promising preclinical results into effective therapeutic interventions in humans by direct application of capsaicin or RTX to adjacent uninjured nerves in patients who suffer from neuropathic pain due to peripheral nerve injury, following surgical interventions, diabetic neuropathy, trauma, vertebral disc herniation, nerve entrapment, ischemia, postherpetic lesion, and spinal cord injury.
Collapse
Affiliation(s)
- Safa Shehab
- Department of Anatomy, College of Medicine and Health Sciences, United Arab Emirates University, Al Ain, United Arab Emirates
| | | | | |
Collapse
|
6
|
Freuville L, Matthys C, Quinton L, Gillet JP. Venom-derived peptides for breaking through the glass ceiling of drug development. Front Chem 2024; 12:1465459. [PMID: 39398192 PMCID: PMC11468230 DOI: 10.3389/fchem.2024.1465459] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2024] [Accepted: 09/04/2024] [Indexed: 10/15/2024] Open
Abstract
Venoms are complex mixtures produced by animals and consist of hundreds of components including small molecules, peptides, and enzymes selected for effectiveness and efficacy over millions of years of evolution. With the development of venomics, which combines genomics, transcriptomics, and proteomics to study animal venoms and their effects deeply, researchers have identified molecules that selectively and effectively act against membrane targets, such as ion channels and G protein-coupled receptors. Due to their remarkable physico-chemical properties, these molecules represent a credible source of new lead compounds. Today, not less than 11 approved venom-derived drugs are on the market. In this review, we aimed to highlight the advances in the use of venom peptides in the treatment of diseases such as neurological disorders, cardiovascular diseases, or cancer. We report on the origin and activity of the peptides already approved and provide a comprehensive overview of those still in development.
Collapse
Affiliation(s)
- Lou Freuville
- Laboratory of Mass Spectrometry, MolSys Research Unit, University of Liège, Liège, Belgium
| | - Chloé Matthys
- Laboratory of Molecular Cancer Biology, URPhyM, NARILIS, University of Namur, Namur, Belgium
| | - Loïc Quinton
- Laboratory of Mass Spectrometry, MolSys Research Unit, University of Liège, Liège, Belgium
| | - Jean-Pierre Gillet
- Laboratory of Molecular Cancer Biology, URPhyM, NARILIS, University of Namur, Namur, Belgium
| |
Collapse
|
7
|
Kamei T, Kudo T, Yamane H, Ishibashi F, Takada Y, Honda S, Maezawa Y, Ikeda K, Oyamada Y. Unique electrophysiological property of a novel Nav1.7, Nav1.8, and Nav1.9 sodium channel blocker, ANP-230. Biochem Biophys Res Commun 2024; 721:150126. [PMID: 38776832 DOI: 10.1016/j.bbrc.2024.150126] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2024] [Revised: 04/28/2024] [Accepted: 05/13/2024] [Indexed: 05/25/2024]
Abstract
Voltage-gated sodium channel subtypes, Nav1.7, Nav1.8, and Nav1.9 are predominantly expressed in peripheral sensory neurons. Recent genetic studies have revealed that they are involved in pathological pain processing and that the blockade of Nav1.7, Nav1.8, or Nav1.9 will become a promising pharmacotherapy especially for neuropathic pain. A growing number of drug discovery programs have targeted either of the subtypes to obtain a selective inhibitor which can provide pain relief without affecting the cardiovascular and central nervous systems, though none of them has been approved yet. Here we describe the in vitro characteristics of ANP-230, a novel sodium channel blocker under clinical development. Surprisingly, ANP-230 was shown to block three pain-related subtypes, human Nav1.7, Nav1.8, and Nav1.9 with similar potency, but had only low inhibitory activity to human cardiac Nav1.5 channel and rat central Nav channels. The voltage clamp experiments using different step pulse protocols revealed that ANP-230 had a "tonic block" mode of action without state- and use-dependency. In addition, ANP-230 caused a depolarizing shift of the activation curve and decelerated gating kinetics in human Nav1.7-stably expressing cells. The depolarizing shift of activation curve was commonly observed in human Nav1.8-stably expressing cells as well as rat dorsal root ganglion neurons. These data suggested a quite unique mechanism of Nav channel inhibition by ANP-230. Finally, ANP-230 reduced excitability of rat dorsal root ganglion neurons in a concentration dependent manner. Collectively, these promising results indicate that ANP-230 could be a potent drug for neuropathic pain.
Collapse
Affiliation(s)
- Tatsuya Kamei
- Pharmacology Research Unit, Research Division, Sumitomo Pharma Co., Ltd., Osaka, 554-0022, Japan; Regenerative & Cellular Medicine Kobe Center, Sumitomo Pharma Co., Ltd., Kobe, 650-0047, Japan.
| | - Takehiro Kudo
- Pharmacology Research Unit, Research Division, Sumitomo Pharma Co., Ltd., Osaka, 554-0022, Japan
| | - Hana Yamane
- Pharmacology Research Unit, Research Division, Sumitomo Pharma Co., Ltd., Osaka, 554-0022, Japan; Regenerative & Cellular Medicine Kobe Center, Sumitomo Pharma Co., Ltd., Kobe, 650-0047, Japan
| | - Fumiaki Ishibashi
- Pharmacology Research Unit, Research Division, Sumitomo Pharma Co., Ltd., Osaka, 554-0022, Japan; Platform Technology Research Unit, Research Division, Sumitomo Pharma Co., Ltd., Osaka, 554-0022, Japan
| | - Yoshinori Takada
- Pharmacology Research Unit, Research Division, Sumitomo Pharma Co., Ltd., Osaka, 554-0022, Japan; Global Corporate Strategy, Sumitomo Pharma Co., Ltd., Tokyo, 104-8356, Japan
| | - Shigeyuki Honda
- Pharmacology Research Unit, Research Division, Sumitomo Pharma Co., Ltd., Osaka, 554-0022, Japan; Sumika Chemical Analysis Service, Ltd., Osaka, 554-0022, Japan
| | - Yasuyo Maezawa
- Pharmacology Research Unit, Research Division, Sumitomo Pharma Co., Ltd., Osaka, 554-0022, Japan
| | - Kazuhito Ikeda
- Pharmacology Research Unit, Research Division, Sumitomo Pharma Co., Ltd., Osaka, 554-0022, Japan; Platform Technology Research Unit, Research Division, Sumitomo Pharma Co., Ltd., Osaka, 554-0022, Japan
| | - Yoshihiro Oyamada
- Pharmacology Research Unit, Research Division, Sumitomo Pharma Co., Ltd., Osaka, 554-0022, Japan; AlphaNavi Pharma Inc., Osaka, 564-0053, Japan
| |
Collapse
|
8
|
Antunes A, Robin P, Mourier G, Béroud R, De Waard M, Servent D, Benoit E. Rattlesnake Crotalphine Analgesic Active on Tetrodotoxin-Sensitive Na + Current in Mouse Dorsal Root Ganglion Neurons. Toxins (Basel) 2024; 16:359. [PMID: 39195769 PMCID: PMC11359915 DOI: 10.3390/toxins16080359] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2024] [Revised: 08/04/2024] [Accepted: 08/13/2024] [Indexed: 08/29/2024] Open
Abstract
Crotalphine is an analgesic peptide identified from the venom of the South American rattlesnake Crotalus durissus terrificus. Although its antinociceptive effect is well documented, its direct mechanisms of action are still unclear. The aim of the present work was to study the action of the crotalid peptide on the NaV1.7 channel subtype, a genetically validated pain target. To this purpose, the effects of crotalphine were evaluated on the NaV1.7 component of the tetrodotoxin-sensitive Na+ current in the dorsal root ganglion neurons of adult mice, using the whole-cell patch-clamp configuration, and on cell viability, using propidium iodide fluorescence and trypan blue assays. The results show that 18.7 µM of peptide inhibited 50% of the Na+ current. The blocking effect occurred without any marked change in the current activation and inactivation kinetics, but it was more important as the membrane potential was more positive. In addition, crotalphine induced an increase in the leakage current amplitude of approximately 150% and led to a maximal 31% decrease in cell viability at a high 50 µM concentration. Taken together, these results point out, for the first time, the effectiveness of crotalphine in acting on the NaV1.7 channel subtype, which may be an additional target contributing to the peptide analgesic properties and, also, although less efficiently, on a second cell plasma membrane component, leading to cell loss.
Collapse
Affiliation(s)
- Aurélie Antunes
- Département Médicaments et Technologies pour la Santé (DMTS), Institut des Sciences du Vivant Frédéric Joliot, Université Paris-Saclay, CEA, Service d’Ingénierie Moléculaire pour la Santé (SIMoS), EMR CNRS/CEA 9004, F-91191 Gif-sur-Yvette, France; (A.A.); (P.R.); (G.M.); (D.S.)
- Smartox Biotechnology, F-38120 Saint-Egrève, France; (R.B.); (M.D.W.)
| | - Philippe Robin
- Département Médicaments et Technologies pour la Santé (DMTS), Institut des Sciences du Vivant Frédéric Joliot, Université Paris-Saclay, CEA, Service d’Ingénierie Moléculaire pour la Santé (SIMoS), EMR CNRS/CEA 9004, F-91191 Gif-sur-Yvette, France; (A.A.); (P.R.); (G.M.); (D.S.)
| | - Gilles Mourier
- Département Médicaments et Technologies pour la Santé (DMTS), Institut des Sciences du Vivant Frédéric Joliot, Université Paris-Saclay, CEA, Service d’Ingénierie Moléculaire pour la Santé (SIMoS), EMR CNRS/CEA 9004, F-91191 Gif-sur-Yvette, France; (A.A.); (P.R.); (G.M.); (D.S.)
| | - Rémy Béroud
- Smartox Biotechnology, F-38120 Saint-Egrève, France; (R.B.); (M.D.W.)
| | - Michel De Waard
- Smartox Biotechnology, F-38120 Saint-Egrève, France; (R.B.); (M.D.W.)
- L’Institut du Thorax, INSERM, CNRS, Université de Nantes, F-44007 Nantes, France
- LabEx “Ion Channels, Science and Therapeutics”, F-06560 Valbonne, France
| | - Denis Servent
- Département Médicaments et Technologies pour la Santé (DMTS), Institut des Sciences du Vivant Frédéric Joliot, Université Paris-Saclay, CEA, Service d’Ingénierie Moléculaire pour la Santé (SIMoS), EMR CNRS/CEA 9004, F-91191 Gif-sur-Yvette, France; (A.A.); (P.R.); (G.M.); (D.S.)
| | - Evelyne Benoit
- Département Médicaments et Technologies pour la Santé (DMTS), Institut des Sciences du Vivant Frédéric Joliot, Université Paris-Saclay, CEA, Service d’Ingénierie Moléculaire pour la Santé (SIMoS), EMR CNRS/CEA 9004, F-91191 Gif-sur-Yvette, France; (A.A.); (P.R.); (G.M.); (D.S.)
| |
Collapse
|
9
|
Vieira WF, Coelho DRA, Litwiler ST, McEachern KM, Clancy JA, Morales-Quezada L, Cassano P. Neuropathic pain, mood, and stress-related disorders: A literature review of comorbidity and co-pathogenesis. Neurosci Biobehav Rev 2024; 161:105673. [PMID: 38614452 DOI: 10.1016/j.neubiorev.2024.105673] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2024] [Revised: 04/02/2024] [Accepted: 04/10/2024] [Indexed: 04/15/2024]
Abstract
Neuropathic pain can be caused by multiple factors, and its prevalence can reach 10% of the global population. It is becoming increasingly evident that limited or short-lasting response to treatments for neuropathic pain is associated with psychological factors, which include psychiatric comorbidities known to affect quality of life. It is estimated that 60% of patients with neuropathic pain also experience depression, anxiety, and stress symptoms. Altered mood, including stress, can be a consequence of several painful conditions but can also favor pain chronicization when preexisting. Despite the apparent tight connection between clinical pain and mood/stress disorders, the exact physiological mechanisms remain unclear. This review aims to provide an overview of state-of-the-art research on the mechanisms of pain related to the pathophysiology of depression, anxiety, and stress disorders.
Collapse
Affiliation(s)
- Willians Fernando Vieira
- Division of Neuropsychiatry and Neuromodulation, Massachusetts General Hospital (MGH), Boston, USA; Department of Psychiatry, Harvard Medical School (HMS), Boston, USA; Department of Anatomy, Institute of Biomedical Sciences (ICB), University of São Paulo (USP), São Paulo, Brazil.
| | - David Richer Araujo Coelho
- Division of Neuropsychiatry and Neuromodulation, Massachusetts General Hospital (MGH), Boston, USA; Department of Psychiatry, Harvard Medical School (HMS), Boston, USA; Harvard T. H. Chan School of Public Health (HSPH), Boston, USA
| | - Scott Thomas Litwiler
- Center for Computational and Integrative Biology (CCIB), Massachusetts General Hospital (MGH), Boston, USA
| | - Kayla Marie McEachern
- Division of Neuropsychiatry and Neuromodulation, Massachusetts General Hospital (MGH), Boston, USA
| | - Julie A Clancy
- Division of Neuropsychiatry and Neuromodulation, Massachusetts General Hospital (MGH), Boston, USA
| | - Leon Morales-Quezada
- Department of Physical Medicine and Rehabilitation, Spaulding Rehabilitation Hospital, Boston, USA
| | - Paolo Cassano
- Division of Neuropsychiatry and Neuromodulation, Massachusetts General Hospital (MGH), Boston, USA; Department of Psychiatry, Harvard Medical School (HMS), Boston, USA
| |
Collapse
|
10
|
Zhao F, Liu Y, Liu Y, Ye Q, Yang H, Gui M, Song Y. The road to evolution of ProTx2: how to be a subtype-specific inhibition of human Na v1.7. Front Pharmacol 2024; 15:1374183. [PMID: 38756380 PMCID: PMC11096480 DOI: 10.3389/fphar.2024.1374183] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2024] [Accepted: 03/29/2024] [Indexed: 05/18/2024] Open
Abstract
The human voltage-gated sodium channel Nav1.7 is a widely proven target for analgesic drug studies. ProTx2, a 30-residue polypeptide from Peruvian green tarantula venom, shows high specificity to activity against human Nav1.7, suggesting its potential to become a non-addictive analgesic. However, its high sensitivity to human Nav1.4 raises concerns about muscle side effects. Here, we engineered three mutants (R13A, R13D, and K27Y) of ProTx2 to evaluate their pharmacological activities toward Nav1.7 and Nav1.4. It is demonstrated that the mutant R13D maintained the analgesic effect in mice while dramatically reducing its muscle toxicity compared with ProTx2. The main reason is the formation of a strong electrostatic interaction between R13D and the negatively charged amino acid residues in DII/S3-S4 of Nav1.7, which is absent in Nav1.4. This study advances our understanding and insights on peptide toxins, paving the way for safer, effective non-addictive analgesic development.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Yongbo Song
- School of Life Science and Biopharmaceutics, Shenyang Pharmaceutical University, Shenyang, China
| |
Collapse
|
11
|
Becker J, Effraim PR, Dib-Hajj S, Rittner HL. Lessons learned in translating pain knowledge into practice. Pain Rep 2023; 8:e1100. [PMID: 37928204 PMCID: PMC10624476 DOI: 10.1097/pr9.0000000000001100] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2022] [Accepted: 05/05/2023] [Indexed: 11/07/2023] Open
Abstract
Introduction During the past 2 decades, basic research deciphering the underlying mechanisms of nociception and chronic pain was thought to finally step beyond opioids and nonsteroidals and provide patients with new analgesics. But apart from calcitonin gene-related peptide antagonists, nothing arrived in hands of clinicians. Objectives To present existing evidence of 3 representative target molecules in the development of novel pain treatment that, so far, did not result in approved drugs. Methods This Clinical Update aligns with the 2022 IASP Global Year Translating Pain Knowledge into Practice and selectively reviews best available evidence and practice. Results We highlight 3 targets: a ion channel, a neuronal growth factor, and a neuropeptide to explore why these drug targets have been dropped in clinical phase II-III trials. Antibodies to nerve growth factor had very good effects in musculoskeletal pain but resulted into more patients requiring joint replacements. Blockers of NaV1.7 were often not effective enough-at least if patients were not stratified. Blockers of neurokinin receptor were similarly not successful enough. In general, failure was most often to the result of a lack of effect and to a lesser extend because of unexpected severe side effects. However, all studies and trials lead to an enormous move in the scientific community to better preclinical models and testing as well as revised methods to molecularly phenotype and stratify patients. Conclusion All stakeholders in the process can help in the future: better preclinical studies, phenotyping and stratifying patients, and participation in clinical trials to move the discovery of analgesics forward.
Collapse
Affiliation(s)
- Juliane Becker
- Department of Anesthesiology, Intensive Care, Emergency and Pain Medicine, Center for Interdisciplinary Pain Medicine, University Hospital Würzburg, Würzburg, Germany
| | - Philip R. Effraim
- Department of Anesthesiology, Yale School of Medicine, New Haven, CT, USA
- Department of Neurology, Center for Neuroscience & Regeneration Research, Yale School of Medicine, New Haven, CT, USA
| | - Sulayman Dib-Hajj
- Department of Neurology, Center for Neuroscience & Regeneration Research, Yale School of Medicine, New Haven, CT, USA
- Rehabilitation Research Center, VA Connecticut Healthcare System, West Haven, CT, USA
| | - Heike L. Rittner
- Department of Anesthesiology, Intensive Care, Emergency and Pain Medicine, Center for Interdisciplinary Pain Medicine, University Hospital Würzburg, Würzburg, Germany
| |
Collapse
|
12
|
Dongol Y, Wilson DT, Daly NL, Cardoso FC, Lewis RJ. Structure-function and rational design of a spider toxin Ssp1a at human voltage-gated sodium channel subtypes. Front Pharmacol 2023; 14:1277143. [PMID: 38034993 PMCID: PMC10682951 DOI: 10.3389/fphar.2023.1277143] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2023] [Accepted: 10/23/2023] [Indexed: 12/02/2023] Open
Abstract
The structure-function and optimization studies of NaV-inhibiting spider toxins have focused on developing selective inhibitors for peripheral pain-sensing NaV1.7. With several NaV subtypes emerging as potential therapeutic targets, structure-function analysis of NaV-inhibiting spider toxins at such subtypes is warranted. Using the recently discovered spider toxin Ssp1a, this study extends the structure-function relationships of NaV-inhibiting spider toxins beyond NaV1.7 to include the epilepsy target NaV1.2 and the pain target NaV1.3. Based on these results and docking studies, we designed analogues for improved potency and/or subtype-selectivity, with S7R-E18K-rSsp1a and N14D-P27R-rSsp1a identified as promising leads. S7R-E18K-rSsp1a increased the rSsp1a potency at these three NaV subtypes, especially at NaV1.3 (∼10-fold), while N14D-P27R-rSsp1a enhanced NaV1.2/1.7 selectivity over NaV1.3. This study highlights the challenge of developing subtype-selective spider toxin inhibitors across multiple NaV subtypes that might offer a more effective therapeutic approach. The findings of this study provide a basis for further rational design of Ssp1a and related NaSpTx1 homologs targeting NaV1.2, NaV1.3 and/or NaV1.7 as research tools and therapeutic leads.
Collapse
Affiliation(s)
- Yashad Dongol
- Centre for Chemistry and Drug Discovery, Institute for Molecular Bioscience, The University of Queensland, Brisbane, QLD, Australia
| | - David T. Wilson
- Australian Institute of Tropical Health and Medicine, James Cook University, Cairns, QLD, Australia
| | - Norelle L. Daly
- Australian Institute of Tropical Health and Medicine, James Cook University, Cairns, QLD, Australia
| | - Fernanda C. Cardoso
- Centre for Chemistry and Drug Discovery, Institute for Molecular Bioscience, The University of Queensland, Brisbane, QLD, Australia
| | - Richard J. Lewis
- Centre for Chemistry and Drug Discovery, Institute for Molecular Bioscience, The University of Queensland, Brisbane, QLD, Australia
| |
Collapse
|
13
|
Cho PG, Jang JH, Ko S, Shin DA, Chung S, Chang MC. The Effect of Evogliptin Tartrate on Controlling Inflammatory Pain. Biomedicines 2023; 11:2990. [PMID: 38001990 PMCID: PMC10669149 DOI: 10.3390/biomedicines11112990] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2023] [Revised: 10/31/2023] [Accepted: 11/03/2023] [Indexed: 11/26/2023] Open
Abstract
Background: Evogliptin tartrate inhibits dipeptidyl peptidase-4 (DPP-4), boosting glucagon-like peptide 1 (GLP-1) secretion and improving insulin release and glucose tolerance, while also exerting anti-inflammatory effects. We investigated its anti-inflammatory and analgesic effects. Methods: Forty male Sprague Dawley rats were divided into (N = 10 in each): (1) naïve, (2) complete Freund's adjuvant (CFA) inflammation + evogliptin tartrate (once for 10 mg/kg) (CFAE), (3) CFA + vehicle (same volume with normal saline with evogliptin tartrate/once) (CFAV), and (4) CFA + indomethacin (5 mg/mL/kg/1 time) (CFAI) groups. CFA was injected subcutaneously into rat plantar regions, and medications (evogliptin tartrate, vehicle, and indomethacin) were administered orally for 5 days. Post treatment, blood from the heart and plantar inflammatory tissue were collected to assess inflammatory cytokines. Evogliptin tartrate effects on controlling inflammation and pain were evaluated by measuring rat plantar paw thickness, paw withdrawal threshold, dorsal root ganglion (DRG) resting membrane potential, DRG action potential firing, and cytokine (TNF-α and IL-1β) levels. Results: Compared with the naïve group, plantar paw thickness, cytokine (TNF-α and IL-1β) levels, DRG resting membrane potential, and DRG action potential firing increased, whereas the paw withdrawal threshold decreased in all CFA groups. However, CFAE and CFAI rats showed recovery. The degree of CFAE recovery resembled that observed in the CFAI group. Conclusions: Evogliptin tartrate mirrored the anti-inflammatory pain relief of indomethacin. We aim to broaden its use as an anti-inflammatory drug or pain relief drug.
Collapse
Affiliation(s)
- Pyung Goo Cho
- Department of Neurosurgery, Ajou University Medical Center, Suwon-si 16499, Republic of Korea;
| | - Jun Ho Jang
- BnH Research Co., Ltd., Goyang-si 10594, Gyeonggi-do, Republic of Korea;
| | - Sukjin Ko
- Department of Physiology, Graduate School of Medical Science, Brain Korea 21 Project, Yonsei University College of Medicine, Seoul 03722, Republic of Korea;
| | - Dong Ah Shin
- Department of Neurosurgery, Yonsei University College of Medicine, Seoul 03722, Republic of Korea;
| | - Seungsoo Chung
- Department of Physiology, Graduate School of Medical Science, Brain Korea 21 Project, Yonsei University College of Medicine, Seoul 03722, Republic of Korea;
| | - Min Cheol Chang
- Department of Physical Medicine & Rehabilitation, College of Medicine, Yeungnam University, Daegu 42415, Republic of Korea
| |
Collapse
|
14
|
Yaksh TL, Santos GGD, Borges Paes Lemes J, Malange K. Neuraxial drug delivery in pain management: An overview of past, present, and future. Best Pract Res Clin Anaesthesiol 2023; 37:243-265. [PMID: 37321769 DOI: 10.1016/j.bpa.2023.04.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2023] [Accepted: 04/11/2023] [Indexed: 06/17/2023]
Abstract
Activation of neuraxial nociceptive linkages leads to a high level of encoding of the message that is transmitted to the brain and that can initiate a pain state with its attendant emotive covariates. As we review here, the encoding of this message is subject to a profound regulation by pharmacological targeting of dorsal root ganglion and dorsal horn systems. Though first shown with the robust and selective modulation by spinal opiates, subsequent work has revealed the pharmacological and biological complexity of these neuraxial systems and points to several regulatory targets. Novel therapeutic delivery platforms, such as viral transfection, antisense and targeted neurotoxins, point to disease-modifying approaches that can selectively address the acute and chronic pain phenotype. Further developments are called for in delivery devices to enhance local distribution and to minimize concentration gradients, as frequently occurs with the poorly mixed intrathecal space. The field has advanced remarkably since the mid-1970s, but these advances must always address the issues of safety and tolerability of neuraxial therapy.
Collapse
Affiliation(s)
- Tony L Yaksh
- Department of Anesthesiology University of California, San Diego, San Diego CA, 92103, USA.
| | | | | | - Kaue Malange
- Department of Anesthesiology University of California, San Diego, San Diego CA, 92103, USA
| |
Collapse
|
15
|
Tran P, Tran HNT, McMahon KL, Deuis JR, Ragnarsson L, Norman A, Sharpe SJ, Payne RJ, Vetter I, Schroeder CI. Changes in Potency and Subtype Selectivity of Bivalent Na V Toxins are Knot-Specific. Bioconjug Chem 2023. [PMID: 37262436 DOI: 10.1021/acs.bioconjchem.3c00135] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/03/2023]
Abstract
Disulfide-rich peptide toxins have long been studied for their ability to inhibit voltage-gated sodium channel subtype NaV1.7, a validated target for the treatment of pain. In this study, we sought to combine the pore blocking activity of conotoxins with the gating modifier activity of spider toxins to design new bivalent inhibitors of NaV1.7 with improved potency and selectivity. To do this, we created an array of heterodimeric toxins designed to target human NaV1.7 by ligating a conotoxin to a spider toxin and assessed the potency and selectivity of the resulting bivalent toxins. A series of spider-derived gating modifier toxins (GpTx-1, ProTx-II, gHwTx-IV, JzTx-V, CcoTx-1, and Pn3a) and two pore-blocker μ-conotoxins, SxIIIC and KIIIA, were used for this study. We employed either enzymatic ligation with sortase A for C- to N-terminal ligation or click chemistry for N- to N-terminal ligation. The bivalent peptide resulting from ligation of ProTx-II and SxIIIC (Pro[LPATG6]Sx) was shown to be the best combination as native ProTx-II potency at hNaV1.7 was conserved following ligation. At hNaV1.4, a synergistic effect between the pore blocker and gating modifier toxin moieties was observed, resulting in altered sodium channel subtype selectivity compared to the parent peptides. Further studies including mutant bivalent peptides and mutant hNaV1.7 channels suggested that gating modifier toxins have a greater contribution to the potency of the bivalent peptides than pore blockers. This study delineated potential benefits and drawbacks of designing pharmacological hybrid peptides targeting hNaV1.7.
Collapse
Affiliation(s)
- Poanna Tran
- Institute for Molecular Bioscience, The University of Queensland, St Lucia, Queensland 4072, Australia
| | - Hue N T Tran
- Institute for Molecular Bioscience, The University of Queensland, St Lucia, Queensland 4072, Australia
| | - Kirsten L McMahon
- Institute for Molecular Bioscience, The University of Queensland, St Lucia, Queensland 4072, Australia
| | - Jennifer R Deuis
- Institute for Molecular Bioscience, The University of Queensland, St Lucia, Queensland 4072, Australia
| | - Lotten Ragnarsson
- Institute for Molecular Bioscience, The University of Queensland, St Lucia, Queensland 4072, Australia
| | - Alexander Norman
- School of Chemistry, The University of Sydney, Camperdown, New South Wales 2006, Australia
- Australian Research Council Centre of Excellence for Innovations in Peptide and Protein Science, The University of Sydney, Camperdown, New South Wales 2006, Australia
| | - Simon J Sharpe
- Molecular Medicine Program, Research Institute, The Hospital for Sick Children, Toronto, Ontario M5G 0A4, Canada
| | - Richard J Payne
- School of Chemistry, The University of Sydney, Camperdown, New South Wales 2006, Australia
- Australian Research Council Centre of Excellence for Innovations in Peptide and Protein Science, The University of Sydney, Camperdown, New South Wales 2006, Australia
| | - Irina Vetter
- Institute for Molecular Bioscience, The University of Queensland, St Lucia, Queensland 4072, Australia
- School of Pharmacy, The University of Queensland, Woolloongabba, Queensland 4102, Australia
| | - Christina I Schroeder
- Institute for Molecular Bioscience, The University of Queensland, St Lucia, Queensland 4072, Australia
- Center for Cancer Research, National Cancer Institute, Frederick, Maryland 21702-1201, United States
- Genentech, 1 DNA Way South San Francisco, California 94080, United States
| |
Collapse
|
16
|
Mateos DL, Yarov-Yarovoy V. Structural modeling of peptide toxin-ion channel interactions using RosettaDock. Proteins 2023. [PMID: 36729043 DOI: 10.1002/prot.26474] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2022] [Revised: 12/09/2022] [Accepted: 01/30/2023] [Indexed: 02/03/2023]
Abstract
Voltage-gated ion channels play essential physiological roles in action potential generation and propagation. Peptidic toxins from animal venoms target ion channels and provide useful scaffolds for the rational design of novel channel modulators with enhanced potency and subtype selectivity. Despite recent progress in obtaining experimental structures of peptide toxin-ion channel complexes, structural determination of peptide toxins bound to ion channels in physiologically important states remains challenging. Here we describe an application of RosettaDock approach to the structural modeling of peptide toxins interactions with ion channels. We tested this approach on 10 structures of peptide toxin-ion channel complexes and demonstrated that it can sample near-native structures in all tested cases. Our approach will be useful for improving the understanding of the molecular mechanism of natural peptide toxin modulation of ion channel gating and for the structural modeling of novel peptide-based ion channel modulators.
Collapse
Affiliation(s)
- Diego Lopez Mateos
- Department of Physiology and Membrane Biology, University of California Davis, Davis, California, USA.,Biophysics Graduate Group, University of California Davis, Davis, California, USA
| | - Vladimir Yarov-Yarovoy
- Department of Physiology and Membrane Biology, University of California Davis, Davis, California, USA.,Biophysics Graduate Group, University of California Davis, Davis, California, USA.,Department of Anesthesiology and Pain Medicine, University of California Davis, Davis, California, USA
| |
Collapse
|
17
|
Zhao F, Fang L, Wang Q, Ye Q, He Y, Xu W, Song Y. Exploring the Pivotal Components Influencing the Side Effects Induced by an Analgesic-Antitumor Peptide from Scorpion Venom on Human Voltage-Gated Sodium Channels 1.4 and 1.5 through Computational Simulation. Toxins (Basel) 2022; 15:33. [PMID: 36668853 PMCID: PMC9864070 DOI: 10.3390/toxins15010033] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2022] [Revised: 12/28/2022] [Accepted: 12/29/2022] [Indexed: 01/04/2023] Open
Abstract
Voltage-gated sodium channels (VGSCs, or Nav) are important determinants of action potential generation and propagation. Efforts are underway to develop medicines targeting different channel subtypes for the treatment of related channelopathies. However, a high degree of conservation across its nine subtypes could lead to the off-target adverse effects on skeletal and cardiac muscles due to acting on primary skeletal muscle sodium channel Nav1.4 and cardiac muscle sodium channel Nav1.5, respectively. For a long evolutionary process, some peptide toxins from venoms have been found to be highly potent yet selective on ion channel subtypes and, therefore, hold the promising potential to be developed into therapeutic agents. In this research, all-atom molecular dynamic methods were used to elucidate the selective mechanisms of an analgesic-antitumor β-scorpion toxin (AGAP) with human Nav1.4 and Nav1.5 in order to unravel the primary reason for the production of its adverse reactions on the skeletal and cardiac muscles. Our results suggest that the rational distribution of residues with ring structures near position 38 and positive residues in the C-terminal on AGAP are critical factors to ensure its analgesic efficacy. Moreover, the substitution for residues with benzene is beneficial to reduce its side effects.
Collapse
Affiliation(s)
- Fan Zhao
- School of Life Science and Biopharmaceutics, Shenyang Pharmaceutical University, 103 Wenhua Road, Shenyang 110016, China
| | - Liangyi Fang
- School of Life Science and Biopharmaceutics, Shenyang Pharmaceutical University, 103 Wenhua Road, Shenyang 110016, China
| | - Qi Wang
- School of Life Science and Biopharmaceutics, Shenyang Pharmaceutical University, 103 Wenhua Road, Shenyang 110016, China
| | - Qi Ye
- School of Life Science and Biopharmaceutics, Shenyang Pharmaceutical University, 103 Wenhua Road, Shenyang 110016, China
| | - Yanan He
- School of Life Science and Biopharmaceutics, Shenyang Pharmaceutical University, 103 Wenhua Road, Shenyang 110016, China
| | - Weizhuo Xu
- Faculty of Functional Food and Wine, Shenyang Pharmaceutical University, 103 Wenhua Road, Shenyang 110016, China
| | - Yongbo Song
- School of Life Science and Biopharmaceutics, Shenyang Pharmaceutical University, 103 Wenhua Road, Shenyang 110016, China
| |
Collapse
|
18
|
Ageitos L, Torres MDT, de la Fuente-Nunez C. Biologically Active Peptides from Venoms: Applications in Antibiotic Resistance, Cancer, and Beyond. Int J Mol Sci 2022; 23:ijms232315437. [PMID: 36499761 PMCID: PMC9740984 DOI: 10.3390/ijms232315437] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2022] [Revised: 10/04/2022] [Accepted: 10/06/2022] [Indexed: 12/12/2022] Open
Abstract
Peptides are potential therapeutic alternatives against global diseases, such as antimicrobial-resistant infections and cancer. Venoms are a rich source of bioactive peptides that have evolved over time to act on specific targets of the prey. Peptides are one of the main components responsible for the biological activity and toxicity of venoms. South American organisms such as scorpions, snakes, and spiders are important producers of a myriad of peptides with different biological activities. In this review, we report the main venom-derived peptide families produced from South American organisms and their corresponding activities and biological targets.
Collapse
Affiliation(s)
- Lucía Ageitos
- Machine Biology Group, Departments of Psychiatry and Microbiology, Institute for Biomedical Informatics, Institute for Translational Medicine and Therapeutics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
- Departments of Bioengineering and Chemical and Biomolecular Engineering, School of Engineering and Applied Science, University of Pennsylvania, Philadelphia, PA 19104, USA
- Penn Institute for Computational Science, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Marcelo D. T. Torres
- Machine Biology Group, Departments of Psychiatry and Microbiology, Institute for Biomedical Informatics, Institute for Translational Medicine and Therapeutics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
- Departments of Bioengineering and Chemical and Biomolecular Engineering, School of Engineering and Applied Science, University of Pennsylvania, Philadelphia, PA 19104, USA
- Penn Institute for Computational Science, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Cesar de la Fuente-Nunez
- Machine Biology Group, Departments of Psychiatry and Microbiology, Institute for Biomedical Informatics, Institute for Translational Medicine and Therapeutics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
- Departments of Bioengineering and Chemical and Biomolecular Engineering, School of Engineering and Applied Science, University of Pennsylvania, Philadelphia, PA 19104, USA
- Penn Institute for Computational Science, University of Pennsylvania, Philadelphia, PA 19104, USA
- Correspondence:
| |
Collapse
|
19
|
Kitano Y, Shinozuka T. Inhibition of Na V1.7: the possibility of ideal analgesics. RSC Med Chem 2022; 13:895-920. [PMID: 36092147 PMCID: PMC9384491 DOI: 10.1039/d2md00081d] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2022] [Accepted: 07/25/2022] [Indexed: 08/03/2023] Open
Abstract
The selective inhibition of NaV1.7 is a promising strategy for developing novel analgesic agents with fewer adverse effects. Although the potent selective inhibition of NaV1.7 has been recently achieved, multiple NaV1.7 inhibitors failed in clinical development. In this review, the relationship between preclinical in vivo efficacy and NaV1.7 coverage among three types of voltage-gated sodium channel (VGSC) inhibitors, namely conventional VGSC inhibitors, sulphonamides and acyl sulphonamides, is discussed. By demonstrating the PK/PD discrepancy of preclinical studies versus in vivo models and clinical results, the potential reasons behind the disconnect between preclinical results and clinical outcomes are discussed together with strategies for developing ideal analgesic agents.
Collapse
Affiliation(s)
- Yutaka Kitano
- R&D Division, Daiichi Sankyo Co., Ltd. 1-2-58 Hiromachi Shinagawa-ku Tokyo 140-8710 Japan
| | - Tsuyoshi Shinozuka
- R&D Division, Daiichi Sankyo Co., Ltd. 1-2-58 Hiromachi Shinagawa-ku Tokyo 140-8710 Japan
| |
Collapse
|
20
|
Borja GB, Zhang H, Harwood BN, Jacques J, Grooms J, Chantre RO, Zhang D, Barnett A, Werley CA, Lu Y, Nagle SF, McManus OB, Dempsey GT. Highly Parallelized, Multicolor Optogenetic Recordings of Cellular Activity for Therapeutic Discovery Applications in Ion Channels and Disease-Associated Excitable Cells. Front Mol Neurosci 2022; 15:896320. [PMID: 35860501 PMCID: PMC9289666 DOI: 10.3389/fnmol.2022.896320] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2022] [Accepted: 06/06/2022] [Indexed: 11/13/2022] Open
Abstract
Optogenetic assays provide a flexible, scalable, and information rich approach to probe compound effects for ion channel drug targets in both heterologous expression systems and associated disease relevant cell types. Despite the potential utility and growing adoption of optogenetics, there remains a critical need for compatible platform technologies with the speed, sensitivity, and throughput to enable their application to broader drug screening applications. To address this challenge, we developed the SwarmTM, a custom designed optical instrument for highly parallelized, multicolor measurements in excitable cells, simultaneously recording changes in voltage and calcium activities at high temporal resolution under optical stimulation. The compact design featuring high power LEDs, large numerical aperture optics, and fast photodiode detection enables all-optical individual well readout of 24-wells simultaneously from multi-well plates while maintaining sufficient temporal resolution to probe millisecond response dynamics. The Swarm delivers variable intensity blue-light optogenetic stimulation to enable membrane depolarization and red or lime-light excitation to enable fluorescence detection of the resulting changes in membrane potential or calcium levels, respectively. The Swarm can screen ~10,000 wells/day in 384-well format, probing complex pharmacological interactions via a wide array of stimulation protocols. To evaluate the Swarm screening system, we optimized a series of heterologous optogenetic spiking HEK293 cell assays for several voltage-gated sodium channel subtypes including Nav1.2, Nav1.5, and Nav1.7. The Swarm was able to record pseudo-action potentials stably across all 24 objectives and provided pharmacological characterization of diverse sodium channel blockers. We performed a Nav1.7 screen of 200,000 small molecules in a 384-well plate format with all 560 plates reaching a Z' > 0.5. As a demonstration of the versatility of the Swarm, we also developed an assay measuring cardiac action potential and calcium waveform properties simultaneously under paced conditions using human induced pluripotent stem (iPS) cell-derived cardiomyocytes as an additional counter screen for cardiac toxicity. In summary, the Swarm is a novel high-throughput all-optical system capable of collecting information-dense data from optogenetic assays in both heterologous and iPS cell-derived models, which can be leveraged to drive diverse therapeutic discovery programs for nervous system disorders and other disease areas involving excitable cells.
Collapse
|
21
|
A Drug Discovery Approach for an Effective Pain Therapy through Selective Inhibition of Nav1.7. Int J Mol Sci 2022; 23:ijms23126793. [PMID: 35743236 PMCID: PMC9223482 DOI: 10.3390/ijms23126793] [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: 05/17/2022] [Revised: 06/14/2022] [Accepted: 06/16/2022] [Indexed: 12/10/2022] Open
Abstract
Chronic pain is a widespread disorder affecting millions of people and is insufficiently addressed by current classes of analgesics due to significant long-term or high dosage side effects. A promising approach that was recently proposed involves the systemic inhibition of the voltage-gated sodium channel Nav1.7, capable of cancelling pain perception completely. Notwithstanding numerous attempts, currently no drugs have been approved for the inhibition of Nav1.7. The task is complicated by the difficulty of creating a selective drug for Nav1.7, and avoiding binding to the many human paralogs performing fundamental physiological functions. In our work, we obtained a promising set of ligands with up to 5-40-fold selectivity and reaching 5.2 nanomolar binding affinity by employing a proper treatment of the problem and an innovative differential in silico screening procedure to discriminate for affinity and selectivity against the Nav paralogs. The absorption, distribution, metabolism, and excretion (ADME) properties of our top-scoring ligands were also evaluated, with good to excellent results. Additionally, our study revealed that the top-scoring ligand is a stereoisomer of an already-approved drug. These facts could reduce the time required to bring a new effective and selective Nav1.7 inhibitor to the market.
Collapse
|
22
|
Wisedchaisri G, Gamal El-Din TM. Druggability of Voltage-Gated Sodium Channels-Exploring Old and New Drug Receptor Sites. Front Pharmacol 2022; 13:858348. [PMID: 35370700 PMCID: PMC8968173 DOI: 10.3389/fphar.2022.858348] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2022] [Accepted: 03/01/2022] [Indexed: 01/12/2023] Open
Abstract
Voltage-gated ion channels are important drug targets because they play crucial physiological roles in both excitable and non-excitable cells. About 15% of clinical drugs used for treating human diseases target ion channels. However, most of these drugs do not provide sufficient specificity to a single subtype of the channels and their off-target side effects can be serious and sometimes fatal. Recent advancements in imaging techniques have enabled us for the first time to visualize unique and hidden parts of voltage-gated sodium channels in different structural conformations, and to develop drugs that further target a selected functional state in each channel subtype with the potential for high precision and low toxicity. In this review we describe the druggability of voltage-gated sodium channels in distinct functional states, which could potentially be used to selectively target the channels. We review classical drug receptors in the channels that have recently been structurally characterized by cryo-electron microscopy with natural neurotoxins and clinical drugs. We further examine recent drug discoveries for voltage-gated sodium channels and discuss opportunities to use distinct, state-dependent receptor sites in the voltage sensors as unique drug targets. Finally, we explore potential new receptor sites that are currently unknown for sodium channels but may be valuable for future drug discovery. The advancement presented here will help pave the way for drug development that selectively targets voltage-gated sodium channels.
Collapse
Affiliation(s)
- Goragot Wisedchaisri
- Department of Pharmacology, University of Washington, Seattle, WA, United States
| | - Tamer M Gamal El-Din
- Department of Pharmacology, University of Washington, Seattle, WA, United States
| |
Collapse
|
23
|
Nguyen PT, Yarov-Yarovoy V. Towards Structure-Guided Development of Pain Therapeutics Targeting Voltage-Gated Sodium Channels. Front Pharmacol 2022; 13:842032. [PMID: 35153801 PMCID: PMC8830516 DOI: 10.3389/fphar.2022.842032] [Citation(s) in RCA: 27] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2021] [Accepted: 01/12/2022] [Indexed: 12/19/2022] Open
Abstract
Voltage-gated sodium (NaV) channels are critical molecular determinants of action potential generation and propagation in excitable cells. Normal NaV channel function disruption can affect physiological neuronal signaling and lead to increased sensitivity to pain, congenital indifference to pain, uncoordinated movement, seizures, or paralysis. Human genetic studies have identified human NaV1.7 (hNaV1.7), hNaV1.8, and hNaV1.9 channel subtypes as crucial players in pain signaling. The premise that subtype selective NaV inhibitors can reduce pain has been reinforced through intensive target validation and therapeutic development efforts. However, an ideal therapeutic has yet to emerge. This review is focused on recent progress, current challenges, and future opportunities to develop NaV channel targeting small molecules and peptides as non-addictive therapeutics to treat pain.
Collapse
Affiliation(s)
- Phuong T Nguyen
- Department of Physiology and Membrane Biology, University of California, Davis, Davis, CA, United States
| | - Vladimir Yarov-Yarovoy
- Department of Physiology and Membrane Biology, University of California, Davis, Davis, CA, United States.,Department of Anesthesiology and Pain Medicine, University of California, Davis, Davis, CA, United States
| |
Collapse
|
24
|
Nguyen PT, Nguyen HM, Wagner KM, Stewart RG, Singh V, Thapa P, Chen YJ, Lillya MW, Ton AT, Kondo R, Ghetti A, Pennington MW, Hammock B, Griffith TN, Sack JT, Wulff H, Yarov-Yarovoy V. Computational design of peptides to target Na V1.7 channel with high potency and selectivity for the treatment of pain. eLife 2022; 11:81727. [PMID: 36576241 PMCID: PMC9831606 DOI: 10.7554/elife.81727] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2022] [Accepted: 12/23/2022] [Indexed: 12/29/2022] Open
Abstract
The voltage-gated sodium NaV1.7 channel plays a key role as a mediator of action potential propagation in C-fiber nociceptors and is an established molecular target for pain therapy. ProTx-II is a potent and moderately selective peptide toxin from tarantula venom that inhibits human NaV1.7 activation. Here we used available structural and experimental data to guide Rosetta design of potent and selective ProTx-II-based peptide inhibitors of human NaV1.7 channels. Functional testing of designed peptides using electrophysiology identified the PTx2-3127 and PTx2-3258 peptides with IC50s of 7 nM and 4 nM for hNaV1.7 and more than 1000-fold selectivity over human NaV1.1, NaV1.3, NaV1.4, NaV1.5, NaV1.8, and NaV1.9 channels. PTx2-3127 inhibits NaV1.7 currents in mouse and human sensory neurons and shows efficacy in rat models of chronic and thermal pain when administered intrathecally. Rationally designed peptide inhibitors of human NaV1.7 channels have transformative potential to define a new class of biologics to treat pain.
Collapse
Affiliation(s)
- Phuong T Nguyen
- Department of Physiology and Membrane Biology, University of California DavisDavisUnited States
| | - Hai M Nguyen
- Department of Pharmacology, University of California DavisDavisUnited States
| | - Karen M Wagner
- Department of Entomology and Nematology & Comprehensive Cancer Center, University of California DavisDavisUnited States
| | - Robert G Stewart
- Department of Physiology and Membrane Biology, University of California DavisDavisUnited States
| | - Vikrant Singh
- Department of Pharmacology, University of California DavisDavisUnited States
| | - Parashar Thapa
- Department of Physiology and Membrane Biology, University of California DavisDavisUnited States
| | - Yi-Je Chen
- Department of Pharmacology, University of California DavisDavisUnited States
| | - Mark W Lillya
- Department of Physiology and Membrane Biology, University of California DavisDavisUnited States
| | | | | | | | | | - Bruce Hammock
- Department of Entomology and Nematology & Comprehensive Cancer Center, University of California DavisDavisUnited States
| | - Theanne N Griffith
- Department of Physiology and Membrane Biology, University of California DavisDavisUnited States
| | - Jon T Sack
- Department of Physiology and Membrane Biology, University of California DavisDavisUnited States,Department of Anesthesiology and Pain Medicine, University of California DavisDavisUnited States
| | - Heike Wulff
- Department of Pharmacology, University of California DavisDavisUnited States
| | - Vladimir Yarov-Yarovoy
- Department of Physiology and Membrane Biology, University of California DavisDavisUnited States,Department of Anesthesiology and Pain Medicine, University of California DavisDavisUnited States,Biophysics Graduate Group, University of California DavisDavisUnited States
| |
Collapse
|
25
|
Dongol Y, Choi PM, Wilson DT, Daly NL, Cardoso FC, Lewis RJ. Voltage-Gated Sodium Channel Modulation by a New Spider Toxin Ssp1a Isolated From an Australian Theraphosid. Front Pharmacol 2021; 12:795455. [PMID: 35002728 PMCID: PMC8740163 DOI: 10.3389/fphar.2021.795455] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2021] [Accepted: 11/22/2021] [Indexed: 11/13/2022] Open
Abstract
Given the important role of voltage-gated sodium (NaV) channel-modulating spider toxins in elucidating the function, pharmacology, and mechanism of action of therapeutically relevant NaV channels, we screened the venom from Australian theraphosid species against the human pain target hNaV1.7. Using assay-guided fractionation, we isolated a 33-residue inhibitor cystine knot (ICK) peptide (Ssp1a) belonging to the NaSpTx1 family. Recombinant Ssp1a (rSsp1a) inhibited neuronal hNaV subtypes with a rank order of potency hNaV1.7 > 1.6 > 1.2 > 1.3 > 1.1. rSsp1a inhibited hNaV1.7, hNaV1.2 and hNaV1.3 without significantly altering the voltage-dependence of activation, inactivation, or delay in recovery from inactivation. However, rSsp1a demonstrated voltage-dependent inhibition at hNaV1.7 and rSsp1a-bound hNaV1.7 opened at extreme depolarizations, suggesting rSsp1a likely interacted with voltage-sensing domain II (VSD II) of hNaV1.7 to trap the channel in its resting state. Nuclear magnetic resonance spectroscopy revealed key structural features of Ssp1a, including an amphipathic surface with hydrophobic and charged patches shown by docking studies to comprise the interacting surface. This study provides the basis for future structure-function studies to guide the development of subtype selective inhibitors.
Collapse
Affiliation(s)
- Yashad Dongol
- Centre for Pain Research, Institute for Molecular Bioscience, The University of Queensland, Brisbane, QLD, Australia
| | - Phil M. Choi
- Centre for Pain Research, Institute for Molecular Bioscience, The University of Queensland, Brisbane, QLD, Australia
| | - David T. Wilson
- Centre for Molecular Therapeutics, Australian Institute of Tropical Health and Medicine, James Cook University, Cairns, QLD, Australia
| | - Norelle L. Daly
- Centre for Molecular Therapeutics, Australian Institute of Tropical Health and Medicine, James Cook University, Cairns, QLD, Australia
| | - Fernanda C. Cardoso
- Centre for Pain Research, Institute for Molecular Bioscience, The University of Queensland, Brisbane, QLD, Australia
| | - Richard J. Lewis
- Centre for Pain Research, Institute for Molecular Bioscience, The University of Queensland, Brisbane, QLD, Australia
| |
Collapse
|
26
|
Adams GL, Pall PS, Grauer SM, Zhou X, Ballard JE, Vavrek M, Kraus RL, Morissette P, Li N, Colarusso S, Bianchi E, Palani A, Klein R, John CT, Wang D, Tudor M, Nolting AF, Biba M, Nowak T, Makarov AA, Reibarkh M, Buevich AV, Zhong W, Regalado EL, Wang X, Gao Q, Shahripour A, Zhu Y, de Simone D, Frattarelli T, Pasquini NM, Magotti P, Iaccarino R, Li Y, Solly K, Lee KJ, Wang W, Chen F, Zeng H, Wang J, Regan H, Amin RP, Regan CP, Burgey CS, Henze DA, Sun C, Tellers DM. Development of ProTx-II Analogues as Highly Selective Peptide Blockers of Na v1.7 for the Treatment of Pain. J Med Chem 2021; 65:485-496. [PMID: 34931831 DOI: 10.1021/acs.jmedchem.1c01570] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
Inhibitor cystine knot peptides, derived from venom, have evolved to block ion channel function but are often toxic when dosed at pharmacologically relevant levels in vivo. The article describes the design of analogues of ProTx-II that safely display systemic in vivo blocking of Nav1.7, resulting in a latency of response to thermal stimuli in rodents. The new designs achieve a better in vivo profile by improving ion channel selectivity and limiting the ability of the peptides to cause mast cell degranulation. The design rationale, structural modeling, in vitro profiles, and rat tail flick outcomes are disclosed and discussed.
Collapse
Affiliation(s)
- Gregory L Adams
- Merck & Co., Inc., West Point, Pennsylvania 19486, United States
| | - Parul S Pall
- Merck & Co., Inc., West Point, Pennsylvania 19486, United States
| | - Steven M Grauer
- Merck & Co., Inc., West Point, Pennsylvania 19486, United States
| | - Xiaoping Zhou
- Merck & Co., Inc., West Point, Pennsylvania 19486, United States
| | | | - Marissa Vavrek
- Merck & Co., Inc., West Point, Pennsylvania 19486, United States
| | - Richard L Kraus
- Merck & Co., Inc., West Point, Pennsylvania 19486, United States
| | | | - Nianyu Li
- Merck & Co., Inc., West Point, Pennsylvania 19486, United States
| | - Stefania Colarusso
- Peptides and Small Molecules R&D Department, IRBM Spa, Via Pontina km 30.600, 00071 Pomezia (RM), Italy
| | - Elisabetta Bianchi
- Peptides and Small Molecules R&D Department, IRBM Spa, Via Pontina km 30.600, 00071 Pomezia (RM), Italy
| | - Anandan Palani
- Merck & Co., Inc., Kenilworth, New Jersey 07033, United States
| | - Rebecca Klein
- Merck & Co., Inc., West Point, Pennsylvania 19486, United States
| | | | - Deping Wang
- Merck & Co., Inc., West Point, Pennsylvania 19486, United States
| | - Matthew Tudor
- Merck & Co., Inc., West Point, Pennsylvania 19486, United States
| | - Andrew F Nolting
- Merck & Co., Inc., West Point, Pennsylvania 19486, United States
| | - Mirlinda Biba
- Merck & Co., Inc., Rahway, New Jersey 07065, United States
| | - Timothy Nowak
- Merck & Co., Inc., Kenilworth, New Jersey 07033, United States
| | | | | | | | - Wendy Zhong
- Merck & Co., Inc., Rahway, New Jersey 07065, United States
| | | | - Xiao Wang
- Merck & Co., Inc., Rahway, New Jersey 07065, United States
| | - Qi Gao
- Merck & Co., Inc., Rahway, New Jersey 07065, United States
| | | | - Yuping Zhu
- Merck & Co., Inc., Kenilworth, New Jersey 07033, United States
| | - Daniele de Simone
- Peptides and Small Molecules R&D Department, IRBM Spa, Via Pontina km 30.600, 00071 Pomezia (RM), Italy
| | - Tommaso Frattarelli
- Peptides and Small Molecules R&D Department, IRBM Spa, Via Pontina km 30.600, 00071 Pomezia (RM), Italy
| | - Nicolo' Maria Pasquini
- Peptides and Small Molecules R&D Department, IRBM Spa, Via Pontina km 30.600, 00071 Pomezia (RM), Italy
| | - Paola Magotti
- Peptides and Small Molecules R&D Department, IRBM Spa, Via Pontina km 30.600, 00071 Pomezia (RM), Italy
| | - Roberto Iaccarino
- Peptides and Small Molecules R&D Department, IRBM Spa, Via Pontina km 30.600, 00071 Pomezia (RM), Italy
| | - Yuxing Li
- Merck & Co., Inc., West Point, Pennsylvania 19486, United States
| | - Kelli Solly
- Merck & Co., Inc., West Point, Pennsylvania 19486, United States
| | - Keun-Joong Lee
- Merck & Co., Inc., Rahway, New Jersey 07065, United States
| | - Weixun Wang
- Merck & Co., Inc., West Point, Pennsylvania 19486, United States
| | - Feifei Chen
- Merck & Co., Inc., West Point, Pennsylvania 19486, United States
| | - Haoyu Zeng
- Merck & Co., Inc., West Point, Pennsylvania 19486, United States
| | - Jixin Wang
- Merck & Co., Inc., West Point, Pennsylvania 19486, United States
| | - Hilary Regan
- Merck & Co., Inc., West Point, Pennsylvania 19486, United States
| | - Rupesh P Amin
- Merck & Co., Inc., West Point, Pennsylvania 19486, United States
| | | | | | - Darrell A Henze
- Merck & Co., Inc., West Point, Pennsylvania 19486, United States
| | - Chengzao Sun
- Merck & Co., Inc., West Point, Pennsylvania 19486, United States
| | - David M Tellers
- Merck & Co., Inc., West Point, Pennsylvania 19486, United States
| |
Collapse
|
27
|
Diochot S. Pain-related toxins in scorpion and spider venoms: a face to face with ion channels. J Venom Anim Toxins Incl Trop Dis 2021; 27:e20210026. [PMID: 34925480 PMCID: PMC8667759 DOI: 10.1590/1678-9199-jvatitd-2021-0026] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2021] [Revised: 05/10/2021] [Indexed: 12/12/2022] Open
Abstract
Pain is a common symptom induced during envenomation by spiders and scorpions.
Toxins isolated from their venom have become essential tools for studying the
functioning and physiopathological role of ion channels, as they modulate their
activity. In particular, toxins that induce pain relief effects can serve as a
molecular basis for the development of future analgesics in humans. This review
provides a summary of the different scorpion and spider toxins that directly
interact with pain-related ion channels, with inhibitory or stimulatory effects.
Some of these toxins were shown to affect pain modalities in different animal
models providing information on the role played by these channels in the pain
process. The close interaction of certain gating-modifier toxins with membrane
phospholipids close to ion channels is examined along with molecular approaches
to improve selectivity, affinity or bioavailability in vivo for
therapeutic purposes.
Collapse
Affiliation(s)
- Sylvie Diochot
- Institut de Pharmacologie Moléculaire et Cellulaire (IPMC), Centre National de la Recherche Scientifique (CNRS) UMR 7275 et Université Côte d'Azur (UCA), 06560 Valbonne, France. Institut de Pharmacologie Moléculaire et Cellulaire Centre National de la Recherche Scientifique Université Côte d'Azur Valbonne France
| |
Collapse
|
28
|
Lopez L, Montnach J, Oliveira-Mendes B, Khakh K, Thomas B, Lin S, Caumes C, Wesolowski S, Nicolas S, Servent D, Cohen C, Béroud R, Benoit E, De Waard M. Synthetic Analogues of Huwentoxin-IV Spider Peptide With Altered Human NaV1.7/NaV1.6 Selectivity Ratios. Front Cell Dev Biol 2021; 9:798588. [PMID: 34988086 PMCID: PMC8722715 DOI: 10.3389/fcell.2021.798588] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2021] [Accepted: 11/22/2021] [Indexed: 11/26/2022] Open
Abstract
Huwentoxin-IV (HwTx-IV), a peptide discovered in the venom of the Chinese bird spider Cyriopagopus schmidti, has been reported to be a potent antinociceptive compound due to its action on the genetically-validated NaV1.7 pain target. Using this peptide for antinociceptive applications in vivo suffers from one major drawback, namely its negative impact on the neuromuscular system. Although studied only recently, this effect appears to be due to an interaction between the peptide and the NaV1.6 channel subtype located at the presynaptic level. The aim of this work was to investigate how HwTx-IV could be modified in order to alter the original human (h) NaV1.7/NaV1.6 selectivity ratio of 23. Nineteen HwTx-IV analogues were chemically synthesized and tested for their blocking effects on the Na+ currents flowing through these two channel subtypes stably expressed in cell lines. Dose-response curves for these analogues were generated, thanks to the use of an automated patch-clamp system. Several key amino acid positions were targeted owing to the information provided by earlier structure-activity relationship (SAR) studies. Among the analogues tested, the potency of HwTx-IV E4K was significantly improved for hNaV1.6, leading to a decreased hNaV1.7/hNaV1.6 selectivity ratio (close to 1). Similar decreased selectivity ratios, but with increased potency for both subtypes, were observed for HwTx-IV analogues that combine a substitution at position 4 with a modification of amino acid 1 or 26 (HwTx-IV E1G/E4G and HwTx-IV E4K/R26Q). In contrast, increased selectivity ratios (>46) were obtained if the E4K mutation was combined to an additional double substitution (R26A/Y33W) or simply by further substituting the C-terminal amidation of the peptide by a carboxylated motif, linked to a marked loss of potency on hNaV1.6 in this latter case. These results demonstrate that it is possible to significantly modulate the selectivity ratio for these two channel subtypes in order to improve the potency of a given analogue for hNaV1.6 and/or hNaV1.7 subtypes. In addition, selective analogues for hNaV1.7, possessing better safety profiles, were produced to limit neuromuscular impairments.
Collapse
Affiliation(s)
- Ludivine Lopez
- L’institut du Thorax, INSERM, CNRS, UNIV NANTES, Nantes, France
| | - Jérôme Montnach
- L’institut du Thorax, INSERM, CNRS, UNIV NANTES, Nantes, France
| | | | | | | | - Sophia Lin
- Xenon Pharmaceuticals, Burnaby, BC, Canada
| | | | | | | | - Denis Servent
- Département Médicaments et Technologies pour La Santé (DMTS), Service d’Ingénierie Moléculaire pour La Santé (SIMoS), ERL CNRS/CEA, Institut des Sciences du Vivant Frédéric Joliot, CEA, Université Paris Saclay, Gif-sur-Yvette, France
| | | | | | - Evelyne Benoit
- Département Médicaments et Technologies pour La Santé (DMTS), Service d’Ingénierie Moléculaire pour La Santé (SIMoS), ERL CNRS/CEA, Institut des Sciences du Vivant Frédéric Joliot, CEA, Université Paris Saclay, Gif-sur-Yvette, France
| | - Michel De Waard
- L’institut du Thorax, INSERM, CNRS, UNIV NANTES, Nantes, France
- Smartox Biotechnology, Saint-Egrève, France
- LabEx « Ion Channels, Science and Therapeutics », Valbonne, France
- *Correspondence: Michel De Waard,
| |
Collapse
|
29
|
Alles SRA, Smith PA. Peripheral Voltage-Gated Cation Channels in Neuropathic Pain and Their Potential as Therapeutic Targets. FRONTIERS IN PAIN RESEARCH 2021; 2:750583. [PMID: 35295464 PMCID: PMC8915663 DOI: 10.3389/fpain.2021.750583] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2021] [Accepted: 11/10/2021] [Indexed: 11/25/2022] Open
Abstract
The persistence of increased excitability and spontaneous activity in injured peripheral neurons is imperative for the development and persistence of many forms of neuropathic pain. This aberrant activity involves increased activity and/or expression of voltage-gated Na+ and Ca2+ channels and hyperpolarization activated cyclic nucleotide gated (HCN) channels as well as decreased function of K+ channels. Because they display limited central side effects, peripherally restricted Na+ and Ca2+ channel blockers and K+ channel activators offer potential therapeutic approaches to pain management. This review outlines the current status and future therapeutic promise of peripherally acting channel modulators. Selective blockers of Nav1.3, Nav1.7, Nav1.8, Cav3.2, and HCN2 and activators of Kv7.2 abrogate signs of neuropathic pain in animal models. Unfortunately, their performance in the clinic has been disappointing; some substances fail to meet therapeutic end points whereas others produce dose-limiting side effects. Despite this, peripheral voltage-gated cation channels retain their promise as therapeutic targets. The way forward may include (i) further structural refinement of K+ channel activators such as retigabine and ASP0819 to improve selectivity and limit toxicity; use or modification of Na+ channel blockers such as vixotrigine, PF-05089771, A803467, PF-01247324, VX-150 or arachnid toxins such as Tap1a; the use of Ca2+ channel blockers such as TTA-P2, TTA-A2, Z 944, ACT709478, and CNCB-2; (ii) improving methods for assessing "pain" as opposed to nociception in rodent models; (iii) recognizing sex differences in pain etiology; (iv) tailoring of therapeutic approaches to meet the symptoms and etiology of pain in individual patients via quantitative sensory testing and other personalized medicine approaches; (v) targeting genetic and biochemical mechanisms controlling channel expression using anti-NGF antibodies such as tanezumab or re-purposed drugs such as vorinostat, a histone methyltransferase inhibitor used in the management of T-cell lymphoma, or cercosporamide a MNK 1/2 inhibitor used in treatment of rheumatoid arthritis; (vi) combination therapy using drugs that are selective for different channel types or regulatory processes; (vii) directing preclinical validation work toward the use of human or human-derived tissue samples; and (viii) application of molecular biological approaches such as clustered regularly interspaced short palindromic repeats (CRISPR) technology.
Collapse
Affiliation(s)
- Sascha R A Alles
- Department of Anesthesiology and Critical Care Medicine, University of New Mexico School of Medicine, Albuquerque, NM, United States
| | - Peter A Smith
- Department of Pharmacology, Neuroscience and Mental Health Institute, University of Alberta, Edmonton, AB, Canada
| |
Collapse
|
30
|
Neff RA, Wickenden AD. Selective Targeting of Nav1.7 with Engineered Spider Venom-Based Peptides. Channels (Austin) 2021; 15:179-193. [PMID: 33427574 PMCID: PMC7808416 DOI: 10.1080/19336950.2020.1860382] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2020] [Revised: 12/02/2020] [Accepted: 12/02/2020] [Indexed: 01/12/2023] Open
Abstract
A fundamental mechanism that drives the propagation of electrical signals in the nervous system is the activation of voltage-gated sodium channels. The sodium channel subtype Nav1.7 is critical for the transmission of pain-related signaling, with gain-of-function mutations in Nav1.7 resulting in various painful pathologies. Loss-of-function mutations cause complete insensitivity to pain and anosmia in humans that otherwise have normal nervous system function, rendering Nav1.7 an attractive target for the treatment of pain. Despite this, no Nav1.7 selective therapeutic has been approved for use as an analgesic to date. Here we present a summary of research that has focused on engineering peptides found in spider venoms to produce Nav1.7 selective antagonists. We discuss the progress that has been made on various scaffolds from different venom families and highlight the challenges that remain in the effort to produce a Nav1.7 selective, venom-based analgesic.
Collapse
Affiliation(s)
- Robert A. Neff
- Neuroscience Discovery, Janssen Research and Development, LLC, San Diego, CA, USA
| | - Alan D. Wickenden
- Molecular and Cellular Pharmacology, Janssen Research and Development, LLC, San Diego, CA, USA
| |
Collapse
|
31
|
Morissette P, Li N, Ballard JE, Vavrek M, Adams GL, Regan C, Regan H, Lee KJ, Wang W, Burton A, Chen F, Gerenser P, Li Y, Kraus RL, Tellers D, Palani A, Zhu Y, Sun C, Bianchi E, Colarusso S, De Simone D, Frattarelli T, Pasquini NM, Amin RP. Guiding Chemically Synthesized Peptide Drug Lead Optimization by Derisking Mast Cell Degranulation-Related Toxicities of a NaV1.7 Peptide Inhibitor. Toxicol Sci 2021; 185:170-183. [PMID: 34897513 DOI: 10.1093/toxsci/kfab138] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Studies have shown that some peptides and small molecules can induce non IgE-mediated anaphylactoid reactions through mast cell activation. Upon activation, mast cells degranulate and release vasoactive and proinflammatory mediators, from cytoplasmic granules into the extracellular environment which can induce a cascade of severe adverse reactions. This study describes a lead optimization strategy to select NaV1.7 inhibitor peptides that minimize acute mast cell degranulation (MCD) toxicities. Various in vitro, in vivo, and PKPD models were used to screen candidates and guide peptide chemical modifications to mitigate this risk. Anesthetized rats dosed with peptides demonstrated treatment-related decreases in blood pressure and increases in plasma histamine concentrations which were reversible with a mast cell stabilizer, supporting the MCD mechanism. In vitro testing in rat mast cells with NaV1.7 peptides demonstrated a concentration-dependent increase in histamine. Pharmacodynamic modeling facilitated establishing an in vitro to in vivo correlation for histamine as a biomarker for blood pressure decline via the MCD mechanism. These models enabled assessment of structure-activity relationship (SAR) to identify substructures that contribute to peptide-mediated MCD. Peptides with hydrophobic and cationic characteristics were determined to have an elevated risk for MCD, which could be reduced or avoided by incorporating anionic residues into the protoxin II scaffold. Our analyses support that in vitro MCD assessment in combination with PKPD modeling can guide SAR to improve peptide lead optimization and ensure an acceptable early in vivo tolerability profile with reduced resources, cycle time, and animal use.
Collapse
Affiliation(s)
- Pierre Morissette
- Nonclinical Drug Safety (NDS), Merck & Co., Inc, West Point, Pennsylvania 19486, USA
| | - Nianyu Li
- Nonclinical Drug Safety (NDS), Merck & Co., Inc, West Point, Pennsylvania 19486, USA
| | - Jeanine E Ballard
- Pharmacokinetics, Pharmacodynamics and Drug Metabolism (PPDM), Merck & Co., Inc, West Point, Pennsylvania 19486, USA
| | - Marissa Vavrek
- Pharmacokinetics, Pharmacodynamics and Drug Metabolism (PPDM), Merck & Co., Inc, West Point, Pennsylvania 19486, USA
| | - Gregory L Adams
- Discovery Chemistry Peptide, Merck & Co., Inc, West Point, Pennsylvania 19486, USA
| | - Chris Regan
- Nonclinical Drug Safety (NDS), Merck & Co., Inc, West Point, Pennsylvania 19486, USA
| | - Hillary Regan
- Nonclinical Drug Safety (NDS), Merck & Co., Inc, West Point, Pennsylvania 19486, USA
| | - K J Lee
- Pharmacokinetics, Pharmacodynamics and Drug Metabolism (PPDM), Merck & Co., Inc, West Point, Pennsylvania 19486, USA
| | - Weixun Wang
- Pharmacokinetics, Pharmacodynamics and Drug Metabolism (PPDM), Merck & Co., Inc, West Point, Pennsylvania 19486, USA
| | - Aimee Burton
- Nonclinical Drug Safety (NDS), Merck & Co., Inc, West Point, Pennsylvania 19486, USA
| | - Feifei Chen
- Nonclinical Drug Safety (NDS), Merck & Co., Inc, West Point, Pennsylvania 19486, USA
| | - Pamela Gerenser
- Nonclinical Drug Safety (NDS), Merck & Co., Inc, West Point, Pennsylvania 19486, USA
| | - Yuxing Li
- Nonclinical Drug Safety (NDS), Merck & Co., Inc, West Point, Pennsylvania 19486, USA
| | - Richard L Kraus
- Nonclinical Drug Safety (NDS), Merck & Co., Inc, West Point, Pennsylvania 19486, USA
| | - David Tellers
- Discovery Chemistry Peptide, Merck & Co., Inc, West Point, Pennsylvania 19486, USA
| | - Anand Palani
- Discovery Chemistry Peptide, Merck & Co., Inc, West Point, Pennsylvania 19486, USA
| | - Yuping Zhu
- Discovery Chemistry Peptide, Merck & Co., Inc, West Point, Pennsylvania 19486, USA
| | - Chengzao Sun
- Discovery Chemistry Peptide, Merck & Co., Inc, West Point, Pennsylvania 19486, USA
| | - Elisabetta Bianchi
- Peptides and Small Molecules R&D Department, IRBM Spa , Pomezia, Rome 00071, Italy
| | - Stefania Colarusso
- Peptides and Small Molecules R&D Department, IRBM Spa , Pomezia, Rome 00071, Italy
| | - Daniele De Simone
- Peptides and Small Molecules R&D Department, IRBM Spa , Pomezia, Rome 00071, Italy
| | - Tommaso Frattarelli
- Peptides and Small Molecules R&D Department, IRBM Spa , Pomezia, Rome 00071, Italy
| | | | - Rupesh P Amin
- Nonclinical Drug Safety (NDS), Merck & Co., Inc, West Point, Pennsylvania 19486, USA
| |
Collapse
|
32
|
Hu H, Mawlawi SE, Zhao T, Deuis JR, Jami S, Vetter I, Lewis RJ, Cardoso FC. Engineering of a Spider Peptide via Conserved Structure-Function Traits Optimizes Sodium Channel Inhibition In Vitro and Anti-Nociception In Vivo. Front Mol Biosci 2021; 8:742457. [PMID: 34621788 PMCID: PMC8490825 DOI: 10.3389/fmolb.2021.742457] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2021] [Accepted: 09/06/2021] [Indexed: 12/12/2022] Open
Abstract
Venom peptides are potent and selective modulators of voltage-gated ion channels that regulate neuronal function both in health and in disease. We previously identified the spider venom peptide Tap1a from the Venezuelan tarantula Theraphosa apophysis that targeted multiple voltage-gated sodium and calcium channels in visceral pain pathways and inhibited visceral mechano-sensing neurons contributing to irritable bowel syndrome. In this work, alanine scanning and domain activity analysis revealed Tap1a inhibited sodium channels by binding with nanomolar affinity to the voltage-sensor domain II utilising conserved structure-function features characteristic of spider peptides belonging to family NaSpTx1. In order to speed up the development of optimized NaV-targeting peptides with greater inhibitory potency and enhanced in vivo activity, we tested the hypothesis that incorporating residues identified from other optimized NaSpTx1 peptides into Tap1a could also optimize its potency for NaVs. Applying this approach, we designed the peptides Tap1a-OPT1 and Tap1a-OPT2 exhibiting significant increased potency for NaV1.1, NaV1.2, NaV1.3, NaV1.6 and NaV1.7 involved in several neurological disorders including acute and chronic pain, motor neuron disease and epilepsy. Tap1a-OPT1 showed increased potency for the off-target NaV1.4, while this off-target activity was absent in Tap1a-OPT2. This enhanced potency arose through a slowed off-rate mechanism. Optimized inhibition of NaV channels observed in vitro translated in vivo, with reversal of nocifensive behaviours in a murine model of NaV-mediated pain also enhanced by Tap1a-OPT. Molecular docking studies suggested that improved interactions within loops 3 and 4, and C-terminal of Tap1a-OPT and the NaV channel voltage-sensor domain II were the main drivers of potency optimization. Overall, the rationally designed peptide Tap1a-OPT displayed new and refined structure-function features which are likely the major contributors to its enhanced bioactive properties observed in vivo. This work contributes to the rapid engineering and optimization of potent spider peptides multi-targeting NaV channels, and the research into novel drugs to treat neurological diseases.
Collapse
Affiliation(s)
- H Hu
- Institute for Molecular Bioscience, The University of Queensland, Brisbane, QLD, Australia
| | - S E Mawlawi
- Institute for Molecular Bioscience, The University of Queensland, Brisbane, QLD, Australia
| | - T Zhao
- Institute for Molecular Bioscience, The University of Queensland, Brisbane, QLD, Australia
| | - J R Deuis
- Institute for Molecular Bioscience, The University of Queensland, Brisbane, QLD, Australia
| | - S Jami
- Institute for Molecular Bioscience, The University of Queensland, Brisbane, QLD, Australia
| | - I Vetter
- Institute for Molecular Bioscience, The University of Queensland, Brisbane, QLD, Australia.,School of Pharmacy, The University of Queensland, Brisbane, QLD, Australia
| | - R J Lewis
- Institute for Molecular Bioscience, The University of Queensland, Brisbane, QLD, Australia
| | - F C Cardoso
- Institute for Molecular Bioscience, The University of Queensland, Brisbane, QLD, Australia.,Centre for Innovations in Peptide and Protein Science, The University of Queensland, Brisbane, QLD, Australia
| |
Collapse
|
33
|
Yu L, Tsuji K, Ujihara I, Liu Q, Pavelkova N, Tsujimura T, Inoue M, Meeker S, Nisenbaum E, McDermott JS, Krajewski J, Undem BJ, Kollarik M, Canning BJ. Antitussive effects of Na V 1.7 blockade in Guinea pigs. Eur J Pharmacol 2021; 907:174192. [PMID: 34010618 PMCID: PMC11822864 DOI: 10.1016/j.ejphar.2021.174192] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2020] [Revised: 05/07/2021] [Accepted: 05/12/2021] [Indexed: 01/25/2023]
Abstract
Our previous studies implicated the voltage-gated sodium channel subtype NaV 1.7 in the transmission of action potentials by the vagal afferent nerves regulating cough and thus identified this channel as a rational therapeutic target for antitussive therapy. But it is presently unclear whether a systemically administered small molecule inhibitor of NaV 1.7 conductance can achieve therapeutic benefit in the absence of side effects on cardiovascular function, gastrointestinal motility or respiration. To this end, we have evaluated the antitussive effects of the NaV 1.7 selective blocker Compound 801 administered systemically in awake guinea pigs or administered topically in anesthetized guinea pigs. We also evaluated the antitussive effects of ambroxol, a low affinity NaV blocker modestly selective for tetrodotoxin resistant NaV subtypes. Both Compound 801 and ambroxol dose-dependently inhibited action potential conduction in guinea pig vagus nerves (assessed by compound potential), with ambroxol nearly 100-fold less potent than the NaV 1.7 selective Compound 801 in this and other NaV 1.7-dependent guinea pig and human tissue-based assays. Both drugs also inhibited citric acid evoked coughing in awake or anesthetized guinea pigs, with potencies supportive of an NaV 1.7-dependent mechanism. Notably, however, the antitussive effects of systemically administered Compound 801 were accompanied by hypotension and respiratory depression. Given the antitussive effects of topically administered Compound 801, we speculate that the likely insurmountable side effects on blood pressure and respiratory drive associated with systemic dosing make topical formulations a viable and perhaps unavoidable therapeutic strategy for targeting NaV 1.7 in cough.
Collapse
Affiliation(s)
- Li Yu
- Department of Pulmonary and Critical Care Medicine, Tongji Hospital, Tongii University School of Medicine, Shanghai, 200065, China
| | - Kojun Tsuji
- Division of Dysphagia Rehabilitation, Niigata University Graduate School of Medical and Dental Sciences, Niigata, Japan
| | - Izumi Ujihara
- Division of Dysphagia Rehabilitation, Niigata University Graduate School of Medical and Dental Sciences, Niigata, Japan
| | - Qi Liu
- Johns Hopkins Asthma and Allergy Center, 5501 Hopkins Bayview Circle, Baltimore, MD, 21224, USA
| | - Nikoleta Pavelkova
- Molecular Pharmacology and Physiology, Morsani College of Medicine, University of South Florida, Tampa, FL, USA
| | - Takanori Tsujimura
- Division of Dysphagia Rehabilitation, Niigata University Graduate School of Medical and Dental Sciences, Niigata, Japan
| | - Makoto Inoue
- Division of Dysphagia Rehabilitation, Niigata University Graduate School of Medical and Dental Sciences, Niigata, Japan
| | - Sonya Meeker
- Johns Hopkins Asthma and Allergy Center, 5501 Hopkins Bayview Circle, Baltimore, MD, 21224, USA
| | - Eric Nisenbaum
- Lilly Research Laboratories, Indianapolis, IN, 46285, USA
| | | | - Jeff Krajewski
- Lilly Research Laboratories, Indianapolis, IN, 46285, USA
| | - Bradley J Undem
- Johns Hopkins Asthma and Allergy Center, 5501 Hopkins Bayview Circle, Baltimore, MD, 21224, USA
| | - Marian Kollarik
- Molecular Pharmacology and Physiology, Morsani College of Medicine, University of South Florida, Tampa, FL, USA
| | - Brendan J Canning
- Johns Hopkins Asthma and Allergy Center, 5501 Hopkins Bayview Circle, Baltimore, MD, 21224, USA.
| |
Collapse
|
34
|
Jiang Y, Castro J, Blomster LV, Agwa AJ, Maddern J, Schober G, Herzig V, Chow CY, Cardoso FC, Demétrio De Souza França P, Gonzales J, Schroeder CI, Esche S, Reiner T, Brierley SM, King GF. Pharmacological Inhibition of the Voltage-Gated Sodium Channel Na V1.7 Alleviates Chronic Visceral Pain in a Rodent Model of Irritable Bowel Syndrome. ACS Pharmacol Transl Sci 2021; 4:1362-1378. [PMID: 34423271 DOI: 10.1021/acsptsci.1c00072] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2021] [Indexed: 12/12/2022]
Abstract
The human nociceptor-specific voltage-gated sodium channel 1.7 (hNaV1.7) is critical for sensing various types of somatic pain, but it appears not to play a primary role in acute visceral pain. However, its role in chronic visceral pain remains to be determined. We used assay-guided fractionation to isolate a novel hNaV1.7 inhibitor, Tsp1a, from tarantula venom. Tsp1a is 28-residue peptide that potently inhibits hNaV1.7 (IC50 = 10 nM), with greater than 100-fold selectivity over hNaV1.3-hNaV1.6, 45-fold selectivity over hNaV1.1, and 24-fold selectivity over hNaV1.2. Tsp1a is a gating modifier that inhibits NaV1.7 by inducing a hyperpolarizing shift in the voltage-dependence of channel inactivation and slowing recovery from fast inactivation. NMR studies revealed that Tsp1a adopts a classical knottin fold, and like many knottin peptides, it is exceptionally stable in human serum. Remarkably, intracolonic administration of Tsp1a completely reversed chronic visceral hypersensitivity in a mouse model of irritable bowel syndrome. The ability of Tsp1a to reduce visceral hypersensitivity in a model of irritable bowel syndrome suggests that pharmacological inhibition of hNaV1.7 at peripheral sensory nerve endings might be a viable approach for eliciting analgesia in patients suffering from chronic visceral pain.
Collapse
Affiliation(s)
- Yan Jiang
- Institute for Molecular Bioscience, The University of Queensland, St. Lucia, Queensland 4072, Australia
| | - Joel Castro
- Visceral Pain Research Group, College of Medicine and Public Health, Flinders Health and Medical Research Institute, Flinders University, Bedford Park, South Australia 5042, Australia.,Hopwood Centre for Neurobiology, Lifelong Health Theme, South Australian Health and Medical Research Institute, Adelaide, South Australia 5000, Australia
| | - Linda V Blomster
- Institute for Molecular Bioscience, The University of Queensland, St. Lucia, Queensland 4072, Australia
| | - Akello J Agwa
- Institute for Molecular Bioscience, The University of Queensland, St. Lucia, Queensland 4072, Australia
| | - Jessica Maddern
- Visceral Pain Research Group, College of Medicine and Public Health, Flinders Health and Medical Research Institute, Flinders University, Bedford Park, South Australia 5042, Australia.,Hopwood Centre for Neurobiology, Lifelong Health Theme, South Australian Health and Medical Research Institute, Adelaide, South Australia 5000, Australia
| | - Gudrun Schober
- Visceral Pain Research Group, College of Medicine and Public Health, Flinders Health and Medical Research Institute, Flinders University, Bedford Park, South Australia 5042, Australia.,Hopwood Centre for Neurobiology, Lifelong Health Theme, South Australian Health and Medical Research Institute, Adelaide, South Australia 5000, Australia
| | - Volker Herzig
- Institute for Molecular Bioscience, The University of Queensland, St. Lucia, Queensland 4072, Australia
| | - Chun Yuen Chow
- Institute for Molecular Bioscience, The University of Queensland, St. Lucia, Queensland 4072, Australia
| | - Fernanda C Cardoso
- Institute for Molecular Bioscience, The University of Queensland, St. Lucia, Queensland 4072, Australia
| | - Paula Demétrio De Souza França
- Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, New York 10065, United States.,Department of Otorhinolaryngology & Head and Neck Surgery, Federal University of São Paulo, São Paulo 04021-001, Brazil
| | - Junior Gonzales
- Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, New York 10065, United States
| | - Christina I Schroeder
- Institute for Molecular Bioscience, The University of Queensland, St. Lucia, Queensland 4072, Australia
| | | | - Thomas Reiner
- Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, New York 10065, United States.,Department of Radiology, Weill Cornell Medical College, New York, New York 10021, United States
| | - Stuart M Brierley
- Visceral Pain Research Group, College of Medicine and Public Health, Flinders Health and Medical Research Institute, Flinders University, Bedford Park, South Australia 5042, Australia.,Hopwood Centre for Neurobiology, Lifelong Health Theme, South Australian Health and Medical Research Institute, Adelaide, South Australia 5000, Australia.,Discipline of Medicine, University of Adelaide, Adelaide, South Australia 5000, Australia
| | - Glenn F King
- Institute for Molecular Bioscience, The University of Queensland, St. Lucia, Queensland 4072, Australia.,Australian Research Council Centre of Excellence for Innovations in Peptide and Protein Science, The University of Queensland, St. Lucia, Queensland 4072, Australia
| |
Collapse
|
35
|
Abd El-Aziz TM, Xiao Y, Kline J, Gridley H, Heaston A, Linse KD, Ward MJ, Rokyta DR, Stockand JD, Cummins TR, Fornelli L, Rowe AH. Identification and Characterization of Novel Proteins from Arizona Bark Scorpion Venom That Inhibit Nav1.8, a Voltage-Gated Sodium Channel Regulator of Pain Signaling. Toxins (Basel) 2021; 13:toxins13070501. [PMID: 34357973 PMCID: PMC8310189 DOI: 10.3390/toxins13070501] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2021] [Revised: 07/14/2021] [Accepted: 07/16/2021] [Indexed: 12/17/2022] Open
Abstract
The voltage-gated sodium channel Nav1.8 is linked to neuropathic and inflammatory pain, highlighting the potential to serve as a drug target. However, the biophysical mechanisms that regulate Nav1.8 activation and inactivation gating are not completely understood. Progress has been hindered by a lack of biochemical tools for examining Nav1.8 gating mechanisms. Arizona bark scorpion (Centruroides sculpturatus) venom proteins inhibit Nav1.8 and block pain in grasshopper mice (Onychomys torridus). These proteins provide tools for examining Nav1.8 structure–activity relationships. To identify proteins that inhibit Nav1.8 activity, venom samples were fractioned using liquid chromatography (reversed-phase and ion exchange). A recombinant Nav1.8 clone expressed in ND7/23 cells was used to identify subfractions that inhibited Nav1.8 Na+ current. Mass-spectrometry-based bottom-up proteomic analyses identified unique peptides from inhibitory subfractions. A search of the peptides against the AZ bark scorpion venom gland transcriptome revealed four novel proteins between 40 and 60% conserved with venom proteins from scorpions in four genera (Centruroides, Parabuthus, Androctonus, and Tityus). Ranging from 63 to 82 amino acids, each primary structure includes eight cysteines and a “CXCE” motif, where X = an aromatic residue (tryptophan, tyrosine, or phenylalanine). Electrophysiology data demonstrated that the inhibitory effects of bioactive subfractions can be removed by hyperpolarizing the channels, suggesting that proteins may function as gating modifiers as opposed to pore blockers.
Collapse
Affiliation(s)
- Tarek Mohamed Abd El-Aziz
- Department of Cellular & Integrative Physiology, University of Texas Health Science Center San Antonio, San Antonio, TX 78229, USA; (T.M.A.E.-A.); (J.D.S.)
- Zoology Department, Faculty of Science, Minia University, El-Minia 61519, Egypt
| | - Yucheng Xiao
- Department of Biology, School of Science, Indiana University-Purdue University Indianapolis, Indianapolis, IN 46202, USA; (Y.X.); (T.R.C.)
| | - Jake Kline
- Department of Biology, University of Oklahoma, 730 Van Vleet Oval, Norman, OK 73019, USA; (J.K.); (H.G.); (A.H.); (L.F.)
| | - Harold Gridley
- Department of Biology, University of Oklahoma, 730 Van Vleet Oval, Norman, OK 73019, USA; (J.K.); (H.G.); (A.H.); (L.F.)
| | - Alyse Heaston
- Department of Biology, University of Oklahoma, 730 Van Vleet Oval, Norman, OK 73019, USA; (J.K.); (H.G.); (A.H.); (L.F.)
| | - Klaus D. Linse
- Bio-Synthesis Inc., 612 E. Main Street, Lewisville, TX 75057, USA;
| | - Micaiah J. Ward
- Department of Biological Sciences, Florida State University, 319 Stadium Drive, Tallahassee, FL 32306, USA; (M.J.W.); (D.R.R.)
| | - Darin R. Rokyta
- Department of Biological Sciences, Florida State University, 319 Stadium Drive, Tallahassee, FL 32306, USA; (M.J.W.); (D.R.R.)
| | - James D. Stockand
- Department of Cellular & Integrative Physiology, University of Texas Health Science Center San Antonio, San Antonio, TX 78229, USA; (T.M.A.E.-A.); (J.D.S.)
| | - Theodore R. Cummins
- Department of Biology, School of Science, Indiana University-Purdue University Indianapolis, Indianapolis, IN 46202, USA; (Y.X.); (T.R.C.)
| | - Luca Fornelli
- Department of Biology, University of Oklahoma, 730 Van Vleet Oval, Norman, OK 73019, USA; (J.K.); (H.G.); (A.H.); (L.F.)
| | - Ashlee H. Rowe
- Department of Biology, University of Oklahoma, 730 Van Vleet Oval, Norman, OK 73019, USA; (J.K.); (H.G.); (A.H.); (L.F.)
- Correspondence: ; Tel.: +1-936-577-5782
| |
Collapse
|
36
|
Qin J, Cheng W, Duan YT, Yang H, Yao Y. Indazole as a Privileged Scaffold: The Derivatives and their Therapeutic Applications. Anticancer Agents Med Chem 2021; 21:839-860. [PMID: 32819234 DOI: 10.2174/1871520620999200818160350] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/29/2020] [Revised: 06/09/2020] [Accepted: 06/25/2020] [Indexed: 11/22/2022]
Abstract
BACKGROUND Heterocyclic compounds, also called heterocycles, are a major class of organic chemical compound that plays a vital role in the metabolism of all living cells. The heterocyclic compound, indazole, has attracted more attention in recent years and is widely present in numerous commercially available drugs. Indazole-containing derivatives, representing one of the most important heterocycles in drug molecules, are endowed with a broad range of biological properties. METHODS A literature search was conducted in PubMed, Google Scholar and Web of Science regarding articles related to indazole and its therapeutic application. RESULTS The mechanism and structure-activity relationship of indazole and its derivatives were described. Based on their versatile biological activities, the compounds were divided into six groups: anti-inflammatory, antibacterial, anti-HIV, antiarrhythmic, antifungal and antitumour. At least 43 indazole-based therapeutic agents were found to be used in clinical application or clinical trials. CONCLUSION This review is a guide for pharmacologists who are in search of valid preclinical/clinical drug compounds where the progress of approved marketed drugs containing indazole scaffold is examined from 1966 to the present day. Future direction involves more diverse bioactive moieties with indazole scaffold and greater insights into its mechanism.
Collapse
Affiliation(s)
- Jinling Qin
- School of Pharmaceutical Science, Zhengzhou University, Zhengzhou, Henan 450001, China
| | - Weyland Cheng
- Henan Provincial Key Laboratory of Children's Genetics and Metabolic Diseases, Children's Hospital Affilited to Zhengzhou University, Zhengzhou University, Henan 450018, China
| | - Yong-Tao Duan
- Henan Provincial Key Laboratory of Children's Genetics and Metabolic Diseases, Children's Hospital Affilited to Zhengzhou University, Zhengzhou University, Henan 450018, China
| | - Hua Yang
- School of Pharmaceutical Science, Zhengzhou University, Zhengzhou, Henan 450001, China
| | - Yongfang Yao
- Henan Provincial Key Laboratory of Children's Genetics and Metabolic Diseases, Children's Hospital Affilited to Zhengzhou University, Zhengzhou University, Henan 450018, China
| |
Collapse
|
37
|
Abstract
Since the introduction of insulin almost a century ago, more than 80 peptide drugs have reached the market for a wide range of diseases, including diabetes, cancer, osteoporosis, multiple sclerosis, HIV infection and chronic pain. In this Perspective, we summarize key trends in peptide drug discovery and development, covering the early efforts focused on human hormones, elegant medicinal chemistry and rational design strategies, peptide drugs derived from nature, and major breakthroughs in molecular biology and peptide chemistry that continue to advance the field. We emphasize lessons from earlier approaches that are still relevant today as well as emerging strategies such as integrated venomics and peptide-display libraries that create new avenues for peptide drug discovery. We also discuss the pharmaceutical landscape in which peptide drugs could be particularly valuable and analyse the challenges that need to be addressed for them to reach their full potential.
Collapse
|
38
|
Structural Pharmacology of Voltage-Gated Sodium Channels. J Mol Biol 2021; 433:166967. [PMID: 33794261 DOI: 10.1016/j.jmb.2021.166967] [Citation(s) in RCA: 39] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2021] [Revised: 03/22/2021] [Accepted: 03/22/2021] [Indexed: 12/19/2022]
Abstract
Voltage-gated sodium (NaV) channels initiate and propagate action potentials in excitable tissues to mediate key physiological processes including heart contraction and nervous system function. Accordingly, NaV channels are major targets for drugs, toxins and disease-causing mutations. Recent breakthroughs in cryo-electron microscopy have led to the visualization of human NaV1.1, NaV1.2, NaV1.4, NaV1.5 and NaV1.7 channel subtypes at high-resolution. These landmark studies have greatly advanced our structural understanding of channel architecture, ion selectivity, voltage-sensing, electromechanical coupling, fast inactivation, and the molecular basis underlying NaV channelopathies. NaV channel structures have also been increasingly determined in complex with toxin and small molecule modulators that target either the pore module or voltage sensor domains. These structural studies have provided new insights into the mechanisms of pharmacological action and opportunities for subtype-selective NaV channel drug design. This review will highlight the structural pharmacology of human NaV channels as well as the potential use of engineered and chimeric channels in future drug discovery efforts.
Collapse
|
39
|
Eagles DA, Chow CY, King GF. Fifteen years of Na
V
1.7 channels as an analgesic target: Why has excellent in vitro pharmacology not translated into in vivo analgesic efficacy? Br J Pharmacol 2020; 179:3592-3611. [DOI: 10.1111/bph.15327] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2020] [Revised: 09/14/2020] [Accepted: 10/23/2020] [Indexed: 12/16/2022] Open
Affiliation(s)
- David A. Eagles
- Institute for Molecular Bioscience The University of Queensland St Lucia QLD Australia
| | - Chun Yuen Chow
- Institute for Molecular Bioscience The University of Queensland St Lucia QLD Australia
| | - Glenn F. King
- Institute for Molecular Bioscience The University of Queensland St Lucia QLD Australia
| |
Collapse
|
40
|
FGF13 Is Required for Histamine-Induced Itch Sensation by Interaction with Na V1.7. J Neurosci 2020; 40:9589-9601. [PMID: 33172979 DOI: 10.1523/jneurosci.0599-20.2020] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2020] [Revised: 10/20/2020] [Accepted: 10/21/2020] [Indexed: 01/17/2023] Open
Abstract
Itch can be induced by activation of small-diameter DRG neurons, which express abundant intracellular fibroblast growth factor 13 (FGF13). Although FGF13 is revealed to be essential for heat nociception, its role in mediating itch remains to be investigated. Here, we reported that loss of FGF13 in mouse DRG neurons impaired the histamine-induced scratching behavior. Calcium imaging showed that the percentage of histamine-responsive DRG neurons was largely decreased in FGF13-deficient mice; and consistently, electrophysiological recording exhibited that histamine failed to evoke action potential firing in most DRG neurons from these mice. Given that the reduced histamine-evoked neuronal response was caused by knockdown of FGF13 but not by FGF13A deficiency, FGF13B was supposed to mediate this process. Furthermore, overexpression of histamine Type 1 receptor H1R, but not H2R, H3R, nor H4R, increased the percentage of histamine-responsive DRG neurons, and the scratching behavior in FGF13-deficient mice was highly reduced by selective activation of H1R, suggesting that H1R is mainly required for FGF13-mediated neuronal response and scratching behavior induced by histamine. However, overexpression of H1R failed to rescue the histamine-evoked neuronal response in FGF13-deficient mice. Histamine enhanced the FGF13 interaction with NaV1.7. Disruption of this interaction by a membrane-permeable competitive peptide, GST-Flag-NaV1.7CT-TAT, reduced the percentage of histamine-responsive DRG neurons, and impaired the histamine-induced scratching, indicating that the FGF13/NaV1.7 interaction is a key molecular determinant in the histamine-induced itch sensation. Therefore, our study reveals a novel role of FGF13 in mediating itch sensation via the interaction of NaV1.7 in the peripheral nervous system.SIGNIFICANCE STATEMENT Scratching induced by itch brings serious tissue damage in chronic itchy diseases, and targeting itch-sensing molecules is crucial for its therapeutic intervention. Here, we reveal that FGF13 is required for the neuronal excitation and scratching behavior induced by histamine. We further provide the evidence that the histamine-evoked neuronal response is mainly mediated by histamine Type 1 receptor H1R, and is largely attenuated in FGF13-deficent mice. Importantly, we identify that histamine enhances the FGF13/NaV1.7 interaction, and disruption of this interaction reduces histamine-evoked neuronal excitation and highly impairs histamine-induced scratching behavior. Additionally, we also find that FGF13 is involved in 5-hydroxytryptamine-induced scratching behavior and hapten 1-fluoro-2,4-dinitrobenzene-induced chronic itch.
Collapse
|
41
|
|
42
|
Alsaloum M, Higerd GP, Effraim PR, Waxman SG. Status of peripheral sodium channel blockers for non-addictive pain treatment. Nat Rev Neurol 2020; 16:689-705. [PMID: 33110213 DOI: 10.1038/s41582-020-00415-2] [Citation(s) in RCA: 110] [Impact Index Per Article: 22.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/10/2020] [Indexed: 12/15/2022]
Abstract
The effective and safe treatment of pain is an unmet health-care need. Current medications used for pain management are often only partially effective, carry dose-limiting adverse effects and are potentially addictive, highlighting the need for improved therapeutic agents. Most common pain conditions originate in the periphery, where dorsal root ganglion and trigeminal ganglion neurons feed pain information into the CNS. Voltage-gated sodium (NaV) channels drive neuronal excitability and three subtypes - NaV1.7, NaV1.8 and NaV1.9 - are preferentially expressed in the peripheral nervous system, suggesting that their inhibition might treat pain while avoiding central and cardiac adverse effects. Genetic and functional studies of human pain disorders have identified NaV1.7, NaV1.8 and NaV1.9 as mediators of pain and validated them as targets for pain treatment. Consequently, multiple NaV1.7-specific and NaV1.8-specific blockers have undergone clinical trials, with others in preclinical development, and the targeting of NaV1.9, although hampered by technical constraints, might also be moving ahead. In this Review, we summarize the clinical and preclinical literature describing compounds that target peripheral NaV channels and discuss the challenges and future prospects for the field. Although the potential of peripheral NaV channel inhibition for the treatment of pain has yet to be realized, this remains a promising strategy to achieve non-addictive analgesia for multiple pain conditions.
Collapse
Affiliation(s)
- Matthew Alsaloum
- Department of Neurology, Yale University School of Medicine, New Haven, CT, USA.,Center for Neuroscience & Regeneration Research, Yale University, West Haven, CT, USA.,Center for Rehabilitation Research, VA Connecticut Healthcare System, West Haven, CT, USA.,Yale Medical Scientist Training Program, Yale School of Medicine, New Haven, CT, USA.,Interdepartmental Neuroscience Program, Yale School of Medicine, New Haven, CT, USA
| | - Grant P Higerd
- Department of Neurology, Yale University School of Medicine, New Haven, CT, USA.,Center for Neuroscience & Regeneration Research, Yale University, West Haven, CT, USA.,Center for Rehabilitation Research, VA Connecticut Healthcare System, West Haven, CT, USA.,Yale Medical Scientist Training Program, Yale School of Medicine, New Haven, CT, USA
| | - Philip R Effraim
- Center for Neuroscience & Regeneration Research, Yale University, West Haven, CT, USA.,Center for Rehabilitation Research, VA Connecticut Healthcare System, West Haven, CT, USA.,Department of Anesthesiology, Yale School of Medicine, New Haven, CT, USA
| | - Stephen G Waxman
- Department of Neurology, Yale University School of Medicine, New Haven, CT, USA. .,Center for Neuroscience & Regeneration Research, Yale University, West Haven, CT, USA. .,Center for Rehabilitation Research, VA Connecticut Healthcare System, West Haven, CT, USA.
| |
Collapse
|
43
|
Shah B, Sindhikara D, Borrelli K, Leffler AE. Water Thermodynamics of Peptide Toxin Binding Sites on Ion Channels. Toxins (Basel) 2020; 12:toxins12100652. [PMID: 33053750 PMCID: PMC7599640 DOI: 10.3390/toxins12100652] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2020] [Revised: 09/21/2020] [Accepted: 10/09/2020] [Indexed: 12/20/2022] Open
Abstract
Peptide toxins isolated from venomous creatures, long prized as research tools due to their innate potency for ion channels, are emerging as drugs as well. However, it remains challenging to understand why peptide toxins bind with high potency to ion channels, to identify residues that are key for activity, and to improve their affinities via mutagenesis. We use WaterMap, a molecular dynamics simulation-based method, to gain computational insight into these three questions by calculating the locations and thermodynamic properties of water molecules in the peptide toxin binding sites of five ion channels. These include an acid-sensing ion channel, voltage-gated potassium channel, sodium channel in activated and deactivated states, transient-receptor potential channel, and a nicotinic receptor whose structures were recently determined by crystallography and cryo-electron microscopy (cryo-EM). All channels had water sites in the peptide toxin binding site, and an average of 75% of these sites were stable (low-energy), and 25% were unstable (medium or high energy). For the sodium channel, more unstable water sites were present in the deactivated state structure than the activated. Additionally, for each channel, unstable water sites coincided with the positions of peptide toxin residues that previous mutagenesis experiments had shown were important for activity. Finally, for the sodium channel in the deactivated state, unstable water sites were present in the peptide toxin binding pocket but did not overlap with the peptide toxin, suggesting that future experimental efforts could focus on targeting these sites to optimize potency.
Collapse
Affiliation(s)
- Binita Shah
- Schrödinger, Inc. 120 W. 45th St, New York, NY 10036, USA; (B.S.); (D.S.); (K.B.)
- PhD Program in Biological and Biomedical Sciences, Division of Medical Sciences, Harvard Medical School, Boston, MA 02115, USA
| | - Dan Sindhikara
- Schrödinger, Inc. 120 W. 45th St, New York, NY 10036, USA; (B.S.); (D.S.); (K.B.)
| | - Ken Borrelli
- Schrödinger, Inc. 120 W. 45th St, New York, NY 10036, USA; (B.S.); (D.S.); (K.B.)
| | - Abba E. Leffler
- Schrödinger, Inc. 120 W. 45th St, New York, NY 10036, USA; (B.S.); (D.S.); (K.B.)
- Correspondence:
| |
Collapse
|
44
|
Pajouhesh H, Beckley JT, Delwig A, Hajare HS, Luu G, Monteleone D, Zhou X, Ligutti J, Amagasu S, Moyer BD, Yeomans DC, Du Bois J, Mulcahy JV. Discovery of a selective, state-independent inhibitor of Na V1.7 by modification of guanidinium toxins. Sci Rep 2020; 10:14791. [PMID: 32908170 PMCID: PMC7481244 DOI: 10.1038/s41598-020-71135-2] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2020] [Accepted: 08/06/2020] [Indexed: 12/15/2022] Open
Abstract
The voltage-gated sodium channel isoform NaV1.7 is highly expressed in dorsal root ganglion neurons and is obligatory for nociceptive signal transmission. Genetic gain-of-function and loss-of-function NaV1.7 mutations have been identified in select individuals, and are associated with episodic extreme pain disorders and insensitivity to pain, respectively. These findings implicate NaV1.7 as a key pharmacotherapeutic target for the treatment of pain. While several small molecules targeting NaV1.7 have been advanced to clinical development, no NaV1.7-selective compound has shown convincing efficacy in clinical pain applications. Here we describe the discovery and characterization of ST-2262, a NaV1.7 inhibitor that blocks the extracellular vestibule of the channel with an IC50 of 72 nM and greater than 200-fold selectivity over off-target sodium channel isoforms, NaV1.1-1.6 and NaV1.8. In contrast to other NaV1.7 inhibitors that preferentially inhibit the inactivated state of the channel, ST-2262 is equipotent in a protocol that favors the resting state of the channel, a protocol that favors the inactivated state, and a high frequency protocol. In a non-human primate study, animals treated with ST-2262 exhibited reduced sensitivity to noxious heat. These findings establish the extracellular vestibule of the sodium channel as a viable receptor site for the design of selective ligands targeting NaV1.7.
Collapse
Affiliation(s)
- H Pajouhesh
- SiteOne Therapeutics, South San Francisco, CA, 94080, USA
| | - J T Beckley
- SiteOne Therapeutics, Bozeman, MT, 59715, USA
| | - A Delwig
- SiteOne Therapeutics, South San Francisco, CA, 94080, USA
| | - H S Hajare
- Department of Chemistry, Stanford University, Stanford, CA, 94305, USA
| | - G Luu
- SiteOne Therapeutics, South San Francisco, CA, 94080, USA
| | - D Monteleone
- SiteOne Therapeutics, South San Francisco, CA, 94080, USA
| | - X Zhou
- SiteOne Therapeutics, South San Francisco, CA, 94080, USA
| | - J Ligutti
- Neuroscience Department, Amgen Research, Thousand Oaks, CA, 91320, USA
| | - S Amagasu
- Neuroscience Department, Amgen Research, Thousand Oaks, CA, 91320, USA
| | - B D Moyer
- Neuroscience Department, Amgen Research, Thousand Oaks, CA, 91320, USA
| | - D C Yeomans
- Department of Anesthesiology, Perioperative, and Pain Medicine, Stanford University, Stanford, CA, 94305, USA
| | - J Du Bois
- Department of Chemistry, Stanford University, Stanford, CA, 94305, USA
| | - J V Mulcahy
- SiteOne Therapeutics, South San Francisco, CA, 94080, USA.
| |
Collapse
|
45
|
Cardoso FC. Multi-targeting sodium and calcium channels using venom peptides for the treatment of complex ion channels-related diseases. Biochem Pharmacol 2020; 181:114107. [PMID: 32579958 DOI: 10.1016/j.bcp.2020.114107] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2020] [Revised: 06/13/2020] [Accepted: 06/17/2020] [Indexed: 02/08/2023]
Abstract
Venom peptides are amongst the most exquisite group of bioactive molecules able to alter the normal physiology of organisms. These bioactive peptides penetrate tissues and blood vessels to encounter a number of receptors and ion channels to which they bind with high affinity and execute modulatory activities. Arachnid is the most diverse class of venomous animals often rich in peptides modulating voltage-gated sodium (NaV), calcium (CaV), and potassium (KV) channels. Spider venoms, in particular, contain potent and selective peptides targeting these channels, with a few displaying interesting multi-target properties for NaV and CaV channels underlying disease mechanisms such as in neuropathic pain, motor neuron disease and cancer. The elucidation of the pharmacology and structure-function properties of these venom peptides are invaluable for the development of effective drugs targeting NaV and CaV channels. This perspective discusses spider venom peptides displaying multi-target properties to modulate NaV and CaV channels in regard to their pharmacological features, structure-function relationships and potential to become the next generation of effective drugs to treat neurological disorders and other multi-ion channels related diseases.
Collapse
Affiliation(s)
- Fernanda C Cardoso
- Institute for Molecular Bioscience, The University of Queensland, 306 Carmody Rd., St Lucia, QLD AU 4072, Australia
| |
Collapse
|
46
|
Jo S, Bean BP. Lidocaine Binding Enhances Inhibition of Nav1.7 Channels by the Sulfonamide PF-05089771. Mol Pharmacol 2020; 97:377-383. [PMID: 32193331 PMCID: PMC7237866 DOI: 10.1124/mol.119.118380] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2019] [Accepted: 02/27/2020] [Indexed: 11/22/2022] Open
Abstract
PF-05089771 is an aryl sulfonamide Nav1.7 channel blocker that binds to the inactivated state of Nav1.7 channels with high affinity but binds only weakly to channels in the resting state. Such aryl sulfonamide Nav1.7 channel blockers bind to the extracellular surface of the S1-S4 voltage-sensor segment of homologous Domain 4, whose movement is associated with inactivation. This binding site is different from that of classic sodium channel inhibitors like lidocaine, which also bind with higher affinity to the inactivated state than the resting state but bind at a site within the pore of the channel. The common dependence on gating state with distinct binding sites raises the possibility that inhibition by aryl sulfonamides and by classic local anesthetics might show an interaction mediated by their mutual state dependence. We tested this possibility by examining the state-dependent inhibition by PF-05089771 and lidocaine of human Nav1.7 channels expressed in human embryonic kidney 293 cells. At -80 mV, where a small fraction of channels are in an inactivated state under drug-free conditions, inhibition by PF-05089771 was both enhanced and speeded in the presence of lidocaine. The results suggest that lidocaine binding to the channel enhances PF-05089771 inhibition by altering the equilibrium between resting states (with D4S4 in the inner position) and inactivated states (with D4S4 in the outer position). The gating state-mediated interaction between the compounds illustrates a principle applicable to many state-dependent agents. SIGNIFICANCE STATEMENT: The results show that lidocaine enhances the degree and rate of inhibition of Nav1.7 channels by the aryl sulfonamide compound PF-05089771, consistent with state-dependent binding by lidocaine increasing the fraction of channels presenting a high-affinity binding site for PF-05089771 and suggesting that combinations of agents targeted to the pore-region binding site of lidocaine and the external binding site of aryl sulfonamides may have synergistic actions.
Collapse
Affiliation(s)
- Sooyeon Jo
- Department of Neurobiology, Harvard Medical School, Boston, Massachusetts
| | - Bruce P Bean
- Department of Neurobiology, Harvard Medical School, Boston, Massachusetts
| |
Collapse
|
47
|
Kong W, Tu X, Huang W, Yang Y, Xie Z, Huang Z. Prediction and Optimization of NaV1.7 Sodium Channel Inhibitors Based on Machine Learning and Simulated Annealing. J Chem Inf Model 2020; 60:2739-2753. [DOI: 10.1021/acs.jcim.9b01180] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Affiliation(s)
- Weikaixin Kong
- Department of Molecular and Cellular Pharmacology, School of Pharmaceutical Sciences, Peking University Health Science Center, 38 Xueyuan Lu,
Haidian district, Beijing 100191, China
| | - Xinyu Tu
- Department of Molecular and Cellular Pharmacology, School of Pharmaceutical Sciences, Peking University Health Science Center, 38 Xueyuan Lu,
Haidian district, Beijing 100191, China
| | - Weiran Huang
- Department of Molecular and Cellular Pharmacology, School of Pharmaceutical Sciences, Peking University Health Science Center, 38 Xueyuan Lu,
Haidian district, Beijing 100191, China
| | - Yang Yang
- Department of Medicinal Chemistry and Molecular Pharmacology, College of Pharmacy, Purdue University, West Lafayette, Indiana 47907, United States
| | - Zhengwei Xie
- Peking University International Cancer Institute and Department of Pharmacology, School of Basic Medical Sciences, Peking University Health Science Center, 38 Xueyuan Lu, Haidian district, Beijing 100191, China
| | - Zhuo Huang
- Department of Molecular and Cellular Pharmacology, School of Pharmaceutical Sciences, Peking University Health Science Center, 38 Xueyuan Lu,
Haidian district, Beijing 100191, China
- State Key Laboratory of Natural and Biomimetic Drugs, Department of Molecular and Cellular Pharmacology, School of Pharmaceutical Sciences, Peking University Health Science Center, 38 Xueyuan Lu,
Haidian district, Beijing 100191, China
| |
Collapse
|
48
|
Antiallodynic effects of the selective NaV1.7 inhibitor Pn3a in a mouse model of acute postsurgical pain: evidence for analgesic synergy with opioids and baclofen. Pain 2020; 160:1766-1780. [PMID: 31335646 DOI: 10.1097/j.pain.0000000000001567] [Citation(s) in RCA: 43] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
Pain is the leading cause of disability in the developed world but remains a poorly treated condition. Specifically, postsurgical pain continues to be a frequent and undermanaged condition. Here, we investigate the analgesic potential of pharmacological NaV1.7 inhibition in a mouse model of acute postsurgical pain, based on incision of the plantar skin and underlying muscle of the hind paw. We demonstrate that local and systemic treatment with the selective NaV1.7 inhibitor μ-theraphotoxin-Pn3a is effectively antiallodynic in this model and completely reverses mechanical hypersensitivity in the absence of motor adverse effects. In addition, the selective NaV1.7 inhibitors ProTx-II and PF-04856264 as well as the clinical candidate CNV1014802 also reduced mechanical allodynia. Interestingly, co-administration of the opioid receptor antagonist naloxone completely reversed analgesic effects of Pn3a, indicating an involvement of endogenous opioids in the analgesic activity of Pn3a. In addition, we found superadditive antinociceptive effects of subtherapeutic Pn3a doses not only with the opioid oxycodone but also with the GABAB receptor agonist baclofen. Transcriptomic analysis of gene expression changes in dorsal root ganglia of mice after surgery did not reveal any changes in mRNA expression of endogenous opioids or opioid receptors; however, several genes involved in pain, including Runx1 (Runt related transcription factor 1), Cacna1a (CaV2.1), and Cacna1b (CaV2.2), were downregulated. In summary, these findings suggest that pain after surgery can be successfully treated with NaV1.7 inhibitors alone or in combination with baclofen or opioids, which may present a novel and safe treatment strategy for this frequent and poorly managed condition.
Collapse
|
49
|
Qian B, Park SH, Yu W. Screening Assay Protocols Targeting the Nav1.7 Channel Using Qube High‐Throughput Automated Patch‐Clamp System. ACTA ACUST UNITED AC 2020; 89:e74. [DOI: 10.1002/cpph.74] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Affiliation(s)
- Binbin Qian
- Sophion Bioscience A/S, Baltorpvej Ballerup Denmark
| | | | - Weifeng Yu
- Sophion Bioscience A/S, Baltorpvej Ballerup Denmark
| |
Collapse
|
50
|
Agwa AJ, Tran P, Mueller A, Tran HNT, Deuis JR, Israel MR, McMahon KL, Craik DJ, Vetter I, Schroeder CI. Manipulation of a spider peptide toxin alters its affinity for lipid bilayers and potency and selectivity for voltage-gated sodium channel subtype 1.7. J Biol Chem 2020; 295:5067-5080. [PMID: 32139508 PMCID: PMC7152767 DOI: 10.1074/jbc.ra119.012281] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2019] [Revised: 03/03/2020] [Indexed: 02/05/2023] Open
Abstract
Huwentoxin-IV (HwTx-IV) is a gating modifier peptide toxin from spiders that has weak affinity for the lipid bilayer. As some gating modifier toxins have affinity for model lipid bilayers, a tripartite relationship among gating modifier toxins, voltage-gated ion channels, and the lipid membrane surrounding the channels has been proposed. We previously designed an HwTx-IV analogue (gHwTx-IV) with reduced negative charge and increased hydrophobic surface profile, which displays increased lipid bilayer affinity and in vitro activity at the voltage-gated sodium channel subtype 1.7 (NaV1.7), a channel targeted in pain management. Here, we show that replacements of the positively-charged residues that contribute to the activity of the peptide can improve gHwTx-IV's potency and selectivity for NaV1.7. Using HwTx-IV, gHwTx-IV, [R26A]gHwTx-IV, [K27A]gHwTx-IV, and [R29A]gHwTx-IV variants, we examined their potency and selectivity at human NaV1.7 and their affinity for the lipid bilayer. [R26A]gHwTx-IV consistently displayed the most improved potency and selectivity for NaV1.7, examined alongside off-target NaVs, compared with HwTx-IV and gHwTx-IV. The lipid affinity of each of the three novel analogues was weaker than that of gHwTx-IV, but stronger than that of HwTx-IV, suggesting a possible relationship between in vitro potency at NaV1.7 and affinity for lipid bilayers. In a murine NaV1.7 engagement model, [R26A]gHwTx-IV exhibited an efficacy comparable with that of native HwTx-IV. In summary, this study reports the development of an HwTx-IV analogue with improved in vitro selectivity for the pain target NaV1.7 and with an in vivo efficacy similar to that of native HwTx-IV.
Collapse
Affiliation(s)
- Akello J Agwa
- Institute for Molecular Bioscience, Centre for Pain Research, The University of Queensland, Brisbane, Queensland 4072, Australia
| | - Poanna Tran
- Institute for Molecular Bioscience, Centre for Pain Research, The University of Queensland, Brisbane, Queensland 4072, Australia
| | - Alexander Mueller
- Institute for Molecular Bioscience, Centre for Pain Research, The University of Queensland, Brisbane, Queensland 4072, Australia
| | - Hue N T Tran
- Institute for Molecular Bioscience, Centre for Pain Research, The University of Queensland, Brisbane, Queensland 4072, Australia
| | - Jennifer R Deuis
- Institute for Molecular Bioscience, Centre for Pain Research, The University of Queensland, Brisbane, Queensland 4072, Australia
| | - Mathilde R Israel
- Institute for Molecular Bioscience, Centre for Pain Research, The University of Queensland, Brisbane, Queensland 4072, Australia
| | - Kirsten L McMahon
- Institute for Molecular Bioscience, Centre for Pain Research, The University of Queensland, Brisbane, Queensland 4072, Australia
| | - David J Craik
- Institute for Molecular Bioscience, Centre for Pain Research, The University of Queensland, Brisbane, Queensland 4072, Australia
| | - Irina Vetter
- Institute for Molecular Bioscience, Centre for Pain Research, The University of Queensland, Brisbane, Queensland 4072, Australia
- School of Pharmacy, The University of Queensland, Woolloongabba, Queensland 4103, Australia
| | - Christina I Schroeder
- Institute for Molecular Bioscience, Centre for Pain Research, The University of Queensland, Brisbane, Queensland 4072, Australia
- National Cancer Institute, National Institutes of Health, Frederick, Maryland 21702, USA
| |
Collapse
|