101
|
Pollak U, Bronicki RA, Achuff BJ, Checchia PA. Postoperative Pain Management in Pediatric Patients Undergoing Cardiac Surgery: Where Are We Heading? J Intensive Care Med 2019:885066619871432. [PMID: 31446831 DOI: 10.1177/0885066619871432] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
OBJECTIVES Adequate postoperative pain management is crucial in pediatric patients undergoing cardiac surgery because pain can lead to devastating short- and long-term consequences. This review discusses the limitations of current postoperative pain assessment and management in children after cardiac surgery, the obstacles to providing optimal treatment, and concepts to consider that may overcome these barriers. DATA SOURCE MEDLINE and PubMed. CONCLUSIONS Effective pain management in infants and young children undergoing cardiac surgery continues to evolve with innovative methods of both assessment and therapy using newer drugs or novel routes of administration. Artificial intelligence- and machine learning-based pain assessment and patient-tailored management in both pain measurement and prevention are already being integrated into the routine of current practice.
Collapse
Affiliation(s)
- Uri Pollak
- 1 Pediatric Cardiac Critical Care Unit, Hadassah University Medical Center, Ein Kerem, Jerusalem, Israel
- 2 Pediatric Cardiology, Hadassah University Medical Center, Ein Kerem, Jerusalem, Israel
- 3 Pediatric Extracorporeal Support Program, Hadassah University Medical Center, Ein Kerem, Jerusalem, Israel
- 4 The Hebrew University Hadassah Medical School, Jerusalem, Israel
| | - Ronald A Bronicki
- 5 Department of Pediatrics, Critical Care Medicine and Cardiology, Baylor College of Medicine, Houston, TX, USA
- 6 Pediatric Cardiovascular Intensive Care Unit, Texas Children's Hospital, Houston, TX, USA
| | - Barbara-Jo Achuff
- 5 Department of Pediatrics, Critical Care Medicine and Cardiology, Baylor College of Medicine, Houston, TX, USA
- 6 Pediatric Cardiovascular Intensive Care Unit, Texas Children's Hospital, Houston, TX, USA
| | - Paul A Checchia
- 5 Department of Pediatrics, Critical Care Medicine and Cardiology, Baylor College of Medicine, Houston, TX, USA
- 6 Pediatric Cardiovascular Intensive Care Unit, Texas Children's Hospital, Houston, TX, USA
| |
Collapse
|
102
|
Evaluation of the Spider ( Phlogiellus genus) Phlotoxin 1 and Synthetic Variants as Antinociceptive Drug Candidates. Toxins (Basel) 2019; 11:toxins11090484. [PMID: 31443554 PMCID: PMC6784069 DOI: 10.3390/toxins11090484] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2019] [Revised: 08/09/2019] [Accepted: 08/15/2019] [Indexed: 11/16/2022] Open
Abstract
Over the two last decades, venom toxins have been explored as alternatives to opioids to treat chronic debilitating pain. At present, approximately 20 potential analgesic toxins, mainly from spider venoms, are known to inhibit with high affinity the NaV1.7 subtype of voltage-gated sodium (NaV) channels, the most promising genetically validated antinociceptive target identified so far. The present study aimed to consolidate the development of phlotoxin 1 (PhlTx1), a 34-amino acid and 3-disulfide bridge peptide of a Phlogiellus genus spider, as an antinociceptive agent by improving its affinity and selectivity for the human (h) NaV1.7 subtype. The synthetic homologue of PhlTx1 was generated and equilibrated between two conformers on reverse-phase liquid chromatography and exhibited potent analgesic effects in a mouse model of NaV1.7-mediated pain. The effects of PhlTx1 and 8 successfully synthetized alanine-substituted variants were studied (by automated whole-cell patch-clamp electrophysiology) on cell lines stably overexpressing hNaV subtypes, as well as two cardiac targets, the hCaV1.2 and hKV11.1 subtypes of voltage-gated calcium (CaV) and potassium (KV) channels, respectively. PhlTx1 and D7A-PhlTx1 were shown to inhibit hNaV1.1-1.3 and 1.5-1.7 subtypes at hundred nanomolar concentrations, while their affinities for hNaV1.4 and 1.8, hCaV1.2 and hKV11.1 subtypes were over micromolar concentrations. Despite similar analgesic effects in the mouse model of NaV1.7-mediated pain and selectivity profiles, the affinity of D7A-PhlTx1 for the NaV1.7 subtype was at least five times higher than that of the wild-type peptide. Computational modelling was performed to deduce the 3D-structure of PhlTx1 and to suggest the amino acids involved in the efficiency of the molecule. In conclusion, the present structure-activity relationship study of PhlTx1 results in a low improved affinity of the molecule for the NaV1.7 subtype, but without any marked change in the molecule selectivity against the other studied ion channel subtypes. Further experiments are therefore necessary before considering the development of PhlTx1 or synthetic variants as antinociceptive drug candidates.
Collapse
|
103
|
Marine Toxins and Nociception: Potential Therapeutic Use in the Treatment of Visceral Pain Associated with Gastrointestinal Disorders. Toxins (Basel) 2019; 11:toxins11080449. [PMID: 31370176 PMCID: PMC6723473 DOI: 10.3390/toxins11080449] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2019] [Revised: 07/24/2019] [Accepted: 07/26/2019] [Indexed: 12/12/2022] Open
Abstract
Visceral pain, of which the pathogenic basis is currently largely unknown, is a hallmark symptom of both functional disorders, such as irritable bowel syndrome, and inflammatory bowel disease. Intrinsic sensory neurons in the enteric nervous system and afferent sensory neurons of the dorsal root ganglia, connecting with the central nervous system, represent the primary neuronal pathways transducing gut visceral pain. Current pharmacological therapies have several limitations, owing to their partial efficacy and the generation of severe adverse effects. Numerous cellular targets of visceral nociception have been recognized, including, among others, channels (i.e., voltage-gated sodium channels, VGSCs, voltage-gated calcium channels, VGCCs, Transient Receptor Potential, TRP, and Acid-sensing ion channels, ASICs) and neurotransmitter pathways (i.e., GABAergic pathways), which represent attractive targets for the discovery of novel drugs. Natural biologically active compounds, such as marine toxins, able to bind with high affinity and selectivity to different visceral pain molecular mediators, may represent a useful tool (1) to improve our knowledge of the physiological and pathological relevance of each nociceptive target, and (2) to discover therapeutically valuable molecules. In this review we report the most recent literature describing the effects of marine toxin on gastrointestinal visceral pain pathways and the possible clinical implications in the treatment of chronic pain associated with gut diseases.
Collapse
|
104
|
Stumpf A, Cheng ZK, Beaudry D, Angelaud R, Gosselin F. Improved Synthesis of the Nav1.7 Inhibitor GDC-0276 via a Highly Regioselective SNAr Reaction. Org Process Res Dev 2019. [DOI: 10.1021/acs.oprd.9b00082] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023]
Affiliation(s)
- Andreas Stumpf
- Department of Small Molecule Process Chemistry, Genentech, Inc., 1 DNA Way, South San Francisco, California 94080, United States
| | - Zhigang Ken Cheng
- Department of Small Molecule Process Chemistry, Genentech, Inc., 1 DNA Way, South San Francisco, California 94080, United States
| | - Danial Beaudry
- Department of Small Molecule Process Chemistry, Genentech, Inc., 1 DNA Way, South San Francisco, California 94080, United States
| | - Remy Angelaud
- Department of Small Molecule Process Chemistry, Genentech, Inc., 1 DNA Way, South San Francisco, California 94080, United States
| | - Francis Gosselin
- Department of Small Molecule Process Chemistry, Genentech, Inc., 1 DNA Way, South San Francisco, California 94080, United States
| |
Collapse
|
105
|
Nicolas S, Zoukimian C, Bosmans F, Montnach J, Diochot S, Cuypers E, De Waard S, Béroud R, Mebs D, Craik D, Boturyn D, Lazdunski M, Tytgat J, De Waard M. Chemical Synthesis, Proper Folding, Na v Channel Selectivity Profile and Analgesic Properties of the Spider Peptide Phlotoxin 1. Toxins (Basel) 2019; 11:toxins11060367. [PMID: 31234412 PMCID: PMC6628435 DOI: 10.3390/toxins11060367] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2019] [Revised: 06/11/2019] [Accepted: 06/16/2019] [Indexed: 12/19/2022] Open
Abstract
Phlotoxin-1 (PhlTx1) is a peptide previously identified in tarantula venom (Phlogius species) that belongs to the inhibitory cysteine-knot (ICK) toxin family. Like many ICK-based spider toxins, the synthesis of PhlTx1 appears particularly challenging, mostly for obtaining appropriate folding and concomitant suitable disulfide bridge formation. Herein, we describe a procedure for the chemical synthesis and the directed sequential disulfide bridge formation of PhlTx1 that allows for a straightforward production of this challenging peptide. We also performed extensive functional testing of PhlTx1 on 31 ion channel types and identified the voltage-gated sodium (Nav) channel Nav1.7 as the main target of this toxin. Moreover, we compared PhlTx1 activity to 10 other spider toxin activities on an automated patch-clamp system with Chinese Hamster Ovary (CHO) cells expressing human Nav1.7. Performing these analyses in reproducible conditions allowed for classification according to the potency of the best natural Nav1.7 peptide blockers. Finally, subsequent in vivo testing revealed that intrathecal injection of PhlTx1 reduces the response of mice to formalin in both the acute pain and inflammation phase without signs of neurotoxicity. PhlTx1 is thus an interesting toxin to investigate Nav1.7 involvement in cellular excitability and pain.
Collapse
Affiliation(s)
- Sébastien Nicolas
- Institut du Thorax, Inserm UMR 1087/CNRS UMR 6291, LabEx "Ion Channels, Science & Therapeutics", F-44007 Nantes, France.
| | - Claude Zoukimian
- Smartox Biotechnology, 6 rue des Platanes, F-38120 Saint-Egrève, France.
- Department of Molecular Chemistry, Univ. Grenoble Alpes, CNRS, 570 rue de la chimie, CS 40700, 38000 Grenoble, France.
| | - Frank Bosmans
- Faculty of Medicine and Health Sciences, Department of Basic and Applied Medical Sciences, 9000 Gent, Belgium.
- Toxicology and Pharmacology, University of Leuven, Campus Gasthuisberg, P.O. Box 922, Herestraat 49, 3000 Leuven, Belgium.
| | - Jérôme Montnach
- Institut du Thorax, Inserm UMR 1087/CNRS UMR 6291, LabEx "Ion Channels, Science & Therapeutics", F-44007 Nantes, France.
| | - Sylvie Diochot
- Université Côte d'Azur, CNRS UMR7275, Institut de Pharmacologie Moléculaire et Cellulaire, 660 route des lucioles, 6560 Valbonne, France.
| | - Eva Cuypers
- Toxicology and Pharmacology, University of Leuven, Campus Gasthuisberg, P.O. Box 922, Herestraat 49, 3000 Leuven, Belgium.
| | - Stephan De Waard
- Institut du Thorax, Inserm UMR 1087/CNRS UMR 6291, LabEx "Ion Channels, Science & Therapeutics", F-44007 Nantes, France.
| | - Rémy Béroud
- Smartox Biotechnology, 6 rue des Platanes, F-38120 Saint-Egrève, France.
| | - Dietrich Mebs
- Institute of Legal Medicine, University of Frankfurt, Kennedyallee 104, Frankfurt, Germany.
| | - David Craik
- Institute for Molecular Bioscience, University of Queensland, Brisbane 4072, Australia.
| | - Didier Boturyn
- Department of Molecular Chemistry, Univ. Grenoble Alpes, CNRS, 570 rue de la chimie, CS 40700, 38000 Grenoble, France.
| | - Michel Lazdunski
- Université Côte d'Azur, CNRS UMR7275, Institut de Pharmacologie Moléculaire et Cellulaire, 660 route des lucioles, 6560 Valbonne, France.
| | - Jan Tytgat
- Toxicology and Pharmacology, University of Leuven, Campus Gasthuisberg, P.O. Box 922, Herestraat 49, 3000 Leuven, Belgium.
| | - Michel De Waard
- Institut du Thorax, Inserm UMR 1087/CNRS UMR 6291, LabEx "Ion Channels, Science & Therapeutics", F-44007 Nantes, France.
- Smartox Biotechnology, 6 rue des Platanes, F-38120 Saint-Egrève, France.
| |
Collapse
|
106
|
Abstract
Neuropathic pain (NP) is an increasingly common chronic pain state and a major health burden, affecting approximately 7% to 10% of the general population. Emerging evidence suggests that genetic factors could partially explain individual susceptibility to NP and the estimated heritability in twins is 37%. The aim of this study was to systematically review and summarize the studies in humans that have investigated the influence of genetic factors associated with NP. We conducted a comprehensive literature search and performed meta-analyses of all the potential genetic variants associated with NP. We reviewed 29 full-text articles and identified 28 genes that were significantly associated with NP, mainly involved in neurotransmission, immune response, and metabolism. Genetic variants in HLA genes, COMT, OPRM1, TNFA, IL6, and GCH1, were found to have an association with NP in more than one study. In the meta-analysis, polymorphisms in HLA-DRB1*13 (odds ratio [OR], 2.96; confidence interval [CI], 1.93-4.56), HLA-DRB1*04 (OR, 1.40; CI, 1.02-1.93), HLA-DQB1*03 (OR, 2.86; CI, 1.57-5.21), HLA-A*33 (OR, 2.32; CI, 1.42-3.80), and HLA-B*44 (OR, 3.17; CI, 2.22-4.55) were associated with significantly increased risk of developing NP, whereas HLA-A*02 (OR, 0.64; CI, 0.47-0.87) conferred reduced risk and neither rs1799971 in OPRM1 (OR, 0.55; CI, 0.27-1.11) nor rs4680 in COMT (OR, 0.95; CI, 0.81-1.13) were significantly associated with NP. These findings demonstrate an important and specific contribution of genetic factors to the risk of developing NP. However, large-scale replication studies are required to validate these candidate genes. Our review also highlights the need for genome-wide association studies with consistent case definition to elucidate the genetic architecture underpinning NP.
Collapse
|
107
|
Xu L, Ding X, Wang T, Mou S, Sun H, Hou T. Voltage-gated sodium channels: structures, functions, and molecular modeling. Drug Discov Today 2019; 24:1389-1397. [PMID: 31129313 DOI: 10.1016/j.drudis.2019.05.014] [Citation(s) in RCA: 39] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2019] [Revised: 04/02/2019] [Accepted: 05/17/2019] [Indexed: 10/26/2022]
Abstract
Voltage-gated sodium channels (VGSCs), formed by 24 transmembrane segments arranged into four domains, have a key role in the initiation and propagation of electrical signaling in excitable cells. VGSCs are involved in a variety of diseases, including epilepsy, cardiac arrhythmias, and neuropathic pain, and therefore have been regarded as appealing therapeutic targets for the development of anticonvulsant, antiarrhythmic, and local anesthetic drugs. In this review, we discuss recent advances in understanding the structures and biological functions of VGSCs. In addition, we systematically summarize eight pharmacologically distinct ligand-binding sites in VGSCs and representative isoform-selective VGSC modulators in clinical trials. Finally, we review studies on molecular modeling and computer-aided drug design (CADD) for VGSCs to help understanding of biological processes involving VGSCs.
Collapse
Affiliation(s)
- Lei Xu
- Institute of Bioinformatics and Medical Engineering, School of Electrical and Information Engineering, Jiangsu University of Technology, Changzhou 213001, China
| | - Xiaoqin Ding
- Beijing Institute of Pharmaceutical Chemistry, Beijing 102205, China
| | - Tianhu Wang
- Institute of Bioinformatics and Medical Engineering, School of Electrical and Information Engineering, Jiangsu University of Technology, Changzhou 213001, China
| | - Shanzhi Mou
- School of Mathematics and Physics, Jiangsu University of Technology, Changzhou 213001, China
| | - Huiyong Sun
- Department of Medicinal Chemistry, School of Pharmacy, State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing 210009, China.
| | - Tingjun Hou
- College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China.
| |
Collapse
|
108
|
|
109
|
Murray JK, Wu B, Tegley CM, Nixey TE, Falsey JR, Herberich B, Yin L, Sham K, Long J, Aral J, Cheng Y, Netirojjanakul C, Doherty L, Glaus C, Ikotun T, Li H, Tran L, Soto M, Salimi-Moosavi H, Ligutti J, Amagasu S, Andrews KL, Be X, Lin MHJ, Foti RS, Ilch CP, Youngblood B, Kornecook TJ, Karow M, Walker KW, Moyer BD, Biswas K, Miranda LP. Engineering Na V1.7 Inhibitory JzTx-V Peptides with a Potency and Basicity Profile Suitable for Antibody Conjugation To Enhance Pharmacokinetics. ACS Chem Biol 2019; 14:806-818. [PMID: 30875193 DOI: 10.1021/acschembio.9b00183] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Drug discovery research on new pain targets with human genetic validation, including the voltage-gated sodium channel NaV1.7, is being pursued to address the unmet medical need with respect to chronic pain and the rising opioid epidemic. As part of early research efforts on this front, we have previously developed NaV1.7 inhibitory peptide-antibody conjugates with tarantula venom-derived GpTx-1 toxin peptides with an extended half-life (80 h) in rodents but only moderate in vitro activity (hNaV1.7 IC50 = 250 nM) and without in vivo activity. We identified the more potent peptide JzTx-V from our natural peptide collection and improved its selectivity against other sodium channel isoforms through positional analogueing. Here we report utilization of the JzTx-V scaffold in a peptide-antibody conjugate and architectural variations in the linker, peptide loading, and antibody attachment site. We found conjugates with 100-fold improved in vitro potency relative to those of complementary GpTx-1 analogues, but pharmacokinetic and bioimaging analyses of these JzTx-V conjugates revealed a shorter than expected plasma half-life in vivo with accumulation in the liver. In an attempt to increase circulatory serum levels, we sought the reduction of the net +6 charge of the JzTx-V scaffold while retaining a desirable NaV in vitro activity profile. The conjugate of a JzTx-V peptide analogue with a +2 formal charge maintained NaV1.7 potency with 18-fold improved plasma exposure in rodents. Balancing the loss of peptide and conjugate potency associated with the reduction of net charge necessary for improved target exposure resulted in a compound with moderate activity in a NaV1.7-dependent pharmacodynamic model but requires further optimization to identify a conjugate that can fully engage NaV1.7 in vivo.
Collapse
|
110
|
Emerging therapies in clinical development and new contributions for neuropathic pain. ACTA ACUST UNITED AC 2019; 66:324-334. [PMID: 31010688 DOI: 10.1016/j.redar.2019.02.003] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2019] [Revised: 02/11/2019] [Accepted: 02/17/2019] [Indexed: 12/19/2022]
Abstract
Neuropathic pain is very challenging to manage because of the heterogeneity of aetiologies, symptoms, and underlying mechanisms. Conventional oral therapies have been limited by negative factors such as systemic side effects, drug-drug interactions, slow onset of action, the need for titration, multiple daily dosing, as well as the potential risk of addiction, dependence, withdrawal symptoms and abuse. Therefore, new therapeutic perspectives are justified. New drugs that act on different therapeutic targets are currently in preclinical development or in their first phases of clinical development. In this review, focus will be directed specifically on new pharmacological treatments for neuropathic pain for which clinical data are already available, including older and known drugs with new data on their anti-neuropathic activity.
Collapse
|
111
|
Glare P, Aubrey KR, Myles PS. Transition from acute to chronic pain after surgery. Lancet 2019; 393:1537-1546. [PMID: 30983589 DOI: 10.1016/s0140-6736(19)30352-6] [Citation(s) in RCA: 496] [Impact Index Per Article: 82.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/13/2018] [Revised: 10/20/2018] [Accepted: 02/07/2019] [Indexed: 12/14/2022]
Abstract
Over the past decade there has been an increasing reliance on strong opioids to treat acute and chronic pain, which has been associated with a rising epidemic of prescription opioid misuse, abuse, and overdose-related deaths. Deaths from prescription opioids have more than quadrupled in the USA since 1999, and this pattern is now occurring globally. Inappropriate opioid prescribing after surgery, particularly after discharge, is a major cause of this problem. Chronic postsurgical pain, occurring in approximately 10% of patients who have surgery, typically begins as acute postoperative pain that is difficult to control, but soon transitions into a persistent pain condition with neuropathic features that are unresponsive to opioids. Research into how and why this transition occurs has led to a stronger appreciation of opioid-induced hyperalgesia, use of more effective and safer opioid-sparing analgesic regimens, and non-pharmacological interventions for pain management. This Series provides an overview of the epidemiology and societal effect, basic science, and current recommendations for managing persistent postsurgical pain. We discuss the advances in the prevention of this transitional pain state, with the aim to promote safer analgesic regimens to better manage patients with acute and chronic pain.
Collapse
Affiliation(s)
- Paul Glare
- Pain Management Research Institute, University of Sydney, Sydney, NSW, Australia
| | - Karin R Aubrey
- Pain Management Research Institute, University of Sydney, Sydney, NSW, Australia; Kolling Institute, Royal North Shore Hospital, St Leonards, NSW, Australia
| | - Paul S Myles
- Department of Anaesthesiology and Perioperative Medicine, Alfred Hospital, Melbourne, VIC.
| |
Collapse
|
112
|
Shen H, Liu D, Wu K, Lei J, Yan N. Structures of human Na v1.7 channel in complex with auxiliary subunits and animal toxins. Science 2019; 363:1303-1308. [PMID: 30765606 DOI: 10.1126/science.aaw2493] [Citation(s) in RCA: 307] [Impact Index Per Article: 51.2] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2018] [Accepted: 01/29/2019] [Indexed: 12/18/2022]
Abstract
Voltage-gated sodium channel Nav1.7 represents a promising target for pain relief. Here we report the cryo-electron microscopy structures of the human Nav1.7-β1-β2 complex bound to two combinations of pore blockers and gating modifier toxins (GMTs), tetrodotoxin with protoxin-II and saxitoxin with huwentoxin-IV, both determined at overall resolutions of 3.2 angstroms. The two structures are nearly identical except for minor shifts of voltage-sensing domain II (VSDII), whose S3-S4 linker accommodates the two GMTs in a similar manner. One additional protoxin-II sits on top of the S3-S4 linker in VSDIV The structures may represent an inactivated state with all four VSDs "up" and the intracellular gate closed. The structures illuminate the path toward mechanistic understanding of the function and disease of Nav1.7 and establish the foundation for structure-aided development of analgesics.
Collapse
Affiliation(s)
- Huaizong Shen
- State Key Laboratory of Membrane Biology, Tsinghua University, Beijing 100084, China.,Beijing Advanced Innovation Center for Structural Biology, Tsinghua University, Beijing 100084, China.,Tsinghua-Peking Joint Center for Life Sciences, School of Life Sciences and School of Medicine, Tsinghua University, Beijing 100084, China
| | - Dongliang Liu
- State Key Laboratory of Membrane Biology, Tsinghua University, Beijing 100084, China.,Beijing Advanced Innovation Center for Structural Biology, Tsinghua University, Beijing 100084, China.,Tsinghua-Peking Joint Center for Life Sciences, School of Life Sciences and School of Medicine, Tsinghua University, Beijing 100084, China
| | - Kun Wu
- Medical Research Center, Beijing Key Laboratory of Cardiopulmonary Cerebral Resuscitation, Beijing Chao-Yang Hospital, Capital Medical University, Beijing 100020, China
| | - Jianlin Lei
- Beijing Advanced Innovation Center for Structural Biology, Tsinghua University, Beijing 100084, China.,Technology Center for Protein Sciences, Ministry of Education Key Laboratory of Protein Sciences, School of Life Sciences, Tsinghua University, Beijing 100084, China
| | - Nieng Yan
- State Key Laboratory of Membrane Biology, Tsinghua University, Beijing 100084, China. .,Beijing Advanced Innovation Center for Structural Biology, Tsinghua University, Beijing 100084, China.,Tsinghua-Peking Joint Center for Life Sciences, School of Life Sciences and School of Medicine, Tsinghua University, Beijing 100084, China
| |
Collapse
|
113
|
Coates MD, Vrana KE, Ruiz-Velasco V. The influence of voltage-gated sodium channels on human gastrointestinal nociception. Neurogastroenterol Motil 2019; 31:e13460. [PMID: 30216585 DOI: 10.1111/nmo.13460] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/13/2018] [Revised: 08/01/2018] [Accepted: 08/07/2018] [Indexed: 12/14/2022]
Abstract
BACKGROUND Abdominal pain is a frequent and persistent problem in the most common gastrointestinal disorders, including irritable bowel syndrome and inflammatory bowel disease. Pain adversely impacts quality of life, incurs significant healthcare expenditures, and remains a challenging issue to manage with few safe therapeutic options currently available. It is imperative that new methods are developed for identifying and treating this symptom. A variety of peripherally active neuroendocrine signaling elements have the capability to influence gastrointestinal pain perception. A large and growing body of evidence suggests that voltage-gated sodium channels (VGSCs) play a critical role in the development and modulation of nociceptive signaling associated with the gut. Several VGSC isoforms demonstrate significant promise as potential targets for improved diagnosis and treatment of gut-based disorders associated with hyper- and hyposensitivity to abdominal pain. PURPOSE In this article, we critically review key investigations that have evaluated the potential role that VGSCs play in visceral nociception and discuss recent advances related to this topic. Specifically, we discuss the following: (a) what is known about the structure and basic function of VGSCs, (b) the role that each VGSC plays in gut nociception, particularly as it relates to human physiology, and (c) potential diagnostic and therapeutic uses of VGSCs to manage disorders associated with chronic abdominal pain.
Collapse
Affiliation(s)
- Matthew D Coates
- Division of Gastroenterology & Hepatology, Department of Medicine, Penn State University College of Medicine, Hershey, Pennsylvania
| | - Kent E Vrana
- Department of Pharmacology, Penn State University College of Medicine, Hershey, Pennsylvania
| | - Victor Ruiz-Velasco
- Department of Anesthesiology and Perioperative Medicine, Penn State University College of Medicine, Hershey, Pennsylvania
| |
Collapse
|
114
|
Chew LA, Bellampalli SS, Dustrude ET, Khanna R. Mining the Na v1.7 interactome: Opportunities for chronic pain therapeutics. Biochem Pharmacol 2019; 163:9-20. [PMID: 30699328 DOI: 10.1016/j.bcp.2019.01.018] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2019] [Accepted: 01/24/2019] [Indexed: 12/14/2022]
Abstract
The peripherally expressed voltage-gated sodium NaV1.7 (gene SCN9A) channel boosts small stimuli to initiate firing of pain-signaling dorsal root ganglia (DRG) neurons and facilitates neurotransmitter release at the first synapse within the spinal cord. Mutations in SCN9A produce distinct human pain syndromes. Widely acknowledged as a "gatekeeper" of pain, NaV1.7 has been the focus of intense investigation but, to date, no NaV1.7-selective drugs have reached the clinic. Elegant crystallographic studies have demonstrated the potential of designing highly potent and selective NaV1.7 compounds but their therapeutic value remains untested. Transcriptional silencing of NaV1.7 by a naturally expressed antisense transcript has been reported in rodents and humans but whether this represents a viable opportunity for designing NaV1.7 therapeutics is currently unknown. The demonstration that loss of NaV1.7 function is associated with upregulation of endogenous opioids and potentiation of mu- and delta-opioid receptor activities, suggests that targeting only NaV1.7 may be insufficient for analgesia. However, the link between opioid-dependent analgesic mechanisms and function of sodium channels and intracellular sodium-dependent signaling remains controversial. Thus, additional new targets - regulators, modulators - are needed. In this context, we mine the literature for the known interactome of NaV1.7 with a focus on protein interactors that affect the channel's trafficking or link it to opioid signaling. As a case study, we present antinociceptive evidence of allosteric regulation of NaV1.7 by the cytosolic collapsin response mediator protein 2 (CRMP2). Throughout discussions of these possible new targets, we offer thoughts on the therapeutic implications of modulating NaV1.7 function in chronic pain.
Collapse
Affiliation(s)
- Lindsey A Chew
- Department of Pharmacology, College of Medicine, University of Arizona, Tucson, AZ 85724, USA
| | - Shreya S Bellampalli
- Department of Pharmacology, College of Medicine, University of Arizona, Tucson, AZ 85724, USA
| | - Erik T Dustrude
- Stark Neurosciences Research Institute, Indiana University School of Medicine, Indianapolis, IN, USA; Department of Psychiatry, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Rajesh Khanna
- Department of Pharmacology, College of Medicine, University of Arizona, Tucson, AZ 85724, USA; Graduate Interdisciplinary Program in Neuroscience, College of Medicine, University of Arizona, Tucson, AZ, USA; The Center for Innovation in Brain Sciences, The University of Arizona Health Sciences, Tucson, AZ 85724, USA.
| |
Collapse
|
115
|
Hameed S. Na v1.7 and Na v1.8: Role in the pathophysiology of pain. Mol Pain 2019; 15:1744806919858801. [PMID: 31172839 PMCID: PMC6589956 DOI: 10.1177/1744806919858801] [Citation(s) in RCA: 115] [Impact Index Per Article: 19.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2019] [Revised: 05/25/2019] [Accepted: 05/30/2019] [Indexed: 01/25/2023] Open
Abstract
Chronic pain is a significant unmet medical problem. Current research regarding sodium channel function in pathological pain is advancing with the hope that it will enable the development of isoform-specific sodium channel blockers, a promising treatment for chronic pain. Before advancements in the pharmacological field, an elucidation of the roles of Nav1.7 and Nav1.8 in the pathophysiology of pain states is required. Thus, the aim of this report is to present what is currently known about the contributions of these sodium channel subtypes in the pathophysiology of neuropathic and inflammatory pain. The electrophysiological properties and localisation of sodium channel isoforms is discussed. Research concerning the genetic links of Nav1.7 and Nav1.8 in acquired neuropathic and inflammatory pain states from the scientific literature in this field is reported. The role of Nav1.7 and Nav1.8 in the generation and maintenance of abnormal neuronal electrogenesis and hyperexcitability highlights the importance of these channels in the development of pathological pain. However, further research in this area is required to fully elucidate the roles of Nav1.7 and Nav1.8 in the pathophysiology of pain for the development of subtype-specific sodium channel blockers.
Collapse
Affiliation(s)
- Shaila Hameed
- Department of Physiology, King’s College London, London, UK
| |
Collapse
|
116
|
Hutchings CJ, Colussi P, Clark TG. Ion channels as therapeutic antibody targets. MAbs 2018; 11:265-296. [PMID: 30526315 PMCID: PMC6380435 DOI: 10.1080/19420862.2018.1548232] [Citation(s) in RCA: 74] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2018] [Revised: 11/01/2018] [Accepted: 11/03/2018] [Indexed: 12/12/2022] Open
Abstract
It is now well established that antibodies have numerous potential benefits when developed as therapeutics. Here, we evaluate the technical challenges of raising antibodies to membrane-spanning proteins together with enabling technologies that may facilitate the discovery of antibody therapeutics to ion channels. Additionally, we discuss the potential targeting opportunities in the anti-ion channel antibody landscape, along with a number of case studies where functional antibodies that target ion channels have been reported. Antibodies currently in development and progressing towards the clinic are highlighted.
Collapse
Affiliation(s)
| | | | - Theodore G. Clark
- TetraGenetics Inc, Arlington Massachusetts, USA
- Department of Microbiology and Immunology, Cornell University, Ithaca New York, USA
| |
Collapse
|
117
|
Englert C, Brendel JC, Majdanski TC, Yildirim T, Schubert S, Gottschaldt M, Windhab N, Schubert US. Pharmapolymers in the 21st century: Synthetic polymers in drug delivery applications. Prog Polym Sci 2018. [DOI: 10.1016/j.progpolymsci.2018.07.005] [Citation(s) in RCA: 70] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
|
118
|
|
119
|
Meredith FL, Rennie KJ. Regional and Developmental Differences in Na + Currents in Vestibular Primary Afferent Neurons. Front Cell Neurosci 2018; 12:423. [PMID: 30487736 PMCID: PMC6246661 DOI: 10.3389/fncel.2018.00423] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2018] [Accepted: 10/29/2018] [Indexed: 02/04/2023] Open
Abstract
The vestibular system relays information about head position via afferent nerve fibers to the brain in the form of action potentials. Voltage-gated Na+ channels in vestibular afferents drive the initiation and propagation of action potentials, but their expression during postnatal development and their contributions to firing in diverse mature afferent populations are unknown. Electrophysiological techniques were used to determine Na+ channel subunit types in vestibular calyx-bearing afferents at different stages of postnatal development. We used whole cell patch clamp recordings in thin slices of gerbil crista neuroepithelium to investigate Na+ channels and firing patterns in central zone (CZ) and peripheral zone (PZ) afferents. PZ afferents are exclusively dimorphic, innervating type I and type II hair cells, whereas CZ afferents can form dimorphs or calyx-only terminals which innervate type I hair cells alone. All afferents expressed tetrodotoxin (TTX)-sensitive Na+ currents, but TTX-sensitivity varied with age. During the fourth postnatal week, 200–300 nM TTX completely blocked sodium currents in PZ and CZ calyces. By contrast, in immature calyces [postnatal day (P) 5–11], a small component of peak sodium current remained in 200 nM TTX. Application of 1 μM TTX, or Jingzhaotoxin-III plus 200 nM TTX, abolished sodium current in immature calyces, suggesting the transient expression of voltage-gated sodium channel 1.5 (Nav1.5) during development. A similar TTX-insensitive current was found in early postnatal crista hair cells (P5–9) and constituted approximately one third of the total sodium current. The Nav1.6 channel blocker, 4,9-anhydrotetrodotoxin, reduced a component of sodium current in immature and mature calyces. At 100 nM 4,9-anhydrotetrodotoxin, peak sodium current was reduced on average by 20% in P5–14 calyces, by 37% in mature dimorphic PZ calyces, but by less than 15% in mature CZ calyx-only terminals. In mature PZ calyces, action potentials became shorter and broader in the presence of 4,9-anhydrotetrodotoxin implicating a role for Nav1.6 channels in firing in dimorphic afferents.
Collapse
Affiliation(s)
- Frances L Meredith
- Department of Otolaryngology, University of Colorado School of Medicine, Aurora, CO, United States
| | - Katherine J Rennie
- Department of Otolaryngology, University of Colorado School of Medicine, Aurora, CO, United States.,Department of Physiology & Biophysics, University of Colorado School of Medicine, Aurora, CO, United States
| |
Collapse
|
120
|
Abstract
In 2000, with the completion of the human genome project, nine related channels were found to comprise the complete voltage-gated sodium gene family and they were renamed NaV1.1–NaV1.9. This millennial event reflected the extraordinary impact of molecular genetics on our understanding of electrical signalling in the nervous system. In this review, studies of animal electricity from the time of Galvani to the present day are described. The seminal experiments and models of Hodgkin and Huxley coupled with the discovery of the structure of DNA, the genetic code and the application of molecular genetics have resulted in an appreciation of the extraordinary diversity of sodium channels and their surprisingly broad repertoire of functions. In the present era, unsuspected roles for sodium channels in a huge range of pathologies have become apparent.
Collapse
Affiliation(s)
- John N Wood
- Molecular Nociception Group, Wolfson Institute for Biomedical Research, University College London, London, UK
| | - Federico Iseppon
- Molecular Nociception Group, Wolfson Institute for Biomedical Research, University College London, London, UK
| |
Collapse
|
121
|
Majeed MH, Ubaidulhaq M, Rugnath A, Eriator I. Extreme Ends of Pain Sensitivity in SCN9A Mutation Variants: Case Report and Literature Review. INNOVATIONS IN CLINICAL NEUROSCIENCE 2018; 15:33-35. [PMID: 30834170 PMCID: PMC6380612] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 06/09/2023]
Abstract
Pain insensitivity disorders are rare; however, when individuals are insensitive to pain, they are significantly more vulnerable to physical injuries, with higher morbidity and mortality rates, compared with the general population. The authors present the case of an 11-month-old male infant with SCN 9A gene mutation that resulted in congenital insensitivity to pain, while his mother, with a different mutation of the same gene, had hypersensitivity to pain. This is a rare familial presentation of the extreme ends of pain sensitivity, and might be the first such example in medical literature. There is little available information regarding the treatment of pain insensitivity disorders. The authors provide a brief discussion regarding diagnosis (including differentials), known etiology, and treatment of congenital insensitivity to pain, of which a multidisciplinary treatment approach is recommended.
Collapse
Affiliation(s)
- Muhammad Hassan Majeed
- Dr. Majeed is Attending Psychiatrist with the Department of Psychiatry at Natchaug Hospital in Norwich, Connecticut
- Dr. Ubaidulhaq is Pain Medicine Fellow with the Department of Anesthesiology at University of Mississippi Medical Center in Jackson, Mississippi
- Dr. Rugnath is Attending Physician with the Department of Anesthesiology & Pain Medicine at University of Mississippi Medical Center in Jackson, Mississippi
- Dr. Eriator is Chairman with the Department of Anesthesiology & Pain Medicine at University of Mississippi Medical Center in Jackson, Mississippi
| | - Muhammad Ubaidulhaq
- Dr. Majeed is Attending Psychiatrist with the Department of Psychiatry at Natchaug Hospital in Norwich, Connecticut
- Dr. Ubaidulhaq is Pain Medicine Fellow with the Department of Anesthesiology at University of Mississippi Medical Center in Jackson, Mississippi
- Dr. Rugnath is Attending Physician with the Department of Anesthesiology & Pain Medicine at University of Mississippi Medical Center in Jackson, Mississippi
- Dr. Eriator is Chairman with the Department of Anesthesiology & Pain Medicine at University of Mississippi Medical Center in Jackson, Mississippi
| | - Anesh Rugnath
- Dr. Majeed is Attending Psychiatrist with the Department of Psychiatry at Natchaug Hospital in Norwich, Connecticut
- Dr. Ubaidulhaq is Pain Medicine Fellow with the Department of Anesthesiology at University of Mississippi Medical Center in Jackson, Mississippi
- Dr. Rugnath is Attending Physician with the Department of Anesthesiology & Pain Medicine at University of Mississippi Medical Center in Jackson, Mississippi
- Dr. Eriator is Chairman with the Department of Anesthesiology & Pain Medicine at University of Mississippi Medical Center in Jackson, Mississippi
| | - Ike Eriator
- Dr. Majeed is Attending Psychiatrist with the Department of Psychiatry at Natchaug Hospital in Norwich, Connecticut
- Dr. Ubaidulhaq is Pain Medicine Fellow with the Department of Anesthesiology at University of Mississippi Medical Center in Jackson, Mississippi
- Dr. Rugnath is Attending Physician with the Department of Anesthesiology & Pain Medicine at University of Mississippi Medical Center in Jackson, Mississippi
- Dr. Eriator is Chairman with the Department of Anesthesiology & Pain Medicine at University of Mississippi Medical Center in Jackson, Mississippi
| |
Collapse
|
122
|
François-Moutal L, Dustrude ET, Wang Y, Brustovetsky T, Dorame A, Ju W, Moutal A, Perez-Miller S, Brustovetsky N, Gokhale V, Khanna M, Khanna R. Inhibition of the Ubc9 E2 SUMO-conjugating enzyme-CRMP2 interaction decreases NaV1.7 currents and reverses experimental neuropathic pain. Pain 2018; 159:2115-2127. [PMID: 29847471 PMCID: PMC6150792 DOI: 10.1097/j.pain.0000000000001294] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
We previously reported that destruction of the small ubiquitin-like modifier (SUMO) modification site in the axonal collapsin response mediator protein 2 (CRMP2) was sufficient to selectively decrease trafficking of the voltage-gated sodium channel NaV1.7 and reverse neuropathic pain. Here, we further interrogate the biophysical nature of the interaction between CRMP2 and the SUMOylation machinery, and test the hypothesis that a rationally designed CRMP2 SUMOylation motif (CSM) peptide can interrupt E2 SUMO-conjugating enzyme Ubc9-dependent modification of CRMP2 leading to a similar suppression of NaV1.7 currents. Microscale thermophoresis and amplified luminescent proximity homogeneous alpha assay revealed a low micromolar binding affinity between CRMP2 and Ubc9. A heptamer peptide harboring CRMP2's SUMO motif, also bound with similar affinity to Ubc9, disrupted the CRMP2-Ubc9 interaction in a concentration-dependent manner. Importantly, incubation of a tat-conjugated cell-penetrating peptide (t-CSM) decreased sodium currents, predominantly NaV1.7, in a model neuronal cell line. Dialysis of t-CSM peptide reduced CRMP2 SUMOylation and blocked surface trafficking of NaV1.7 in rat sensory neurons. Fluorescence dye-based imaging in rat sensory neurons demonstrated inhibition of sodium influx in the presence of t-CSM peptide; by contrast, calcium influx was unaffected. Finally, t-CSM effectively reversed persistent mechanical and thermal hypersensitivity induced by a spinal nerve injury, a model of neuropathic pain. Structural modeling has now identified a pocket-harboring CRMP2's SUMOylation motif that, when targeted through computational screening of ligands/molecules, is expected to identify small molecules that will biochemically and functionally target CRMP2's SUMOylation to reduce NaV1.7 currents and reverse neuropathic pain.
Collapse
Affiliation(s)
- Liberty François-Moutal
- Department of Pharmacology, The University of Arizona Health Sciences, Tucson, Arizona 85724
| | - Erik T. Dustrude
- Department of Pharmacology, The University of Arizona Health Sciences, Tucson, Arizona 85724
| | - Yue Wang
- Department of Pharmacology, The University of Arizona Health Sciences, Tucson, Arizona 85724
| | - Tatiana Brustovetsky
- Department of Pharmacology and Toxicology, and Stark Neurosciences Research Institute, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | - Angie Dorame
- Department of Pharmacology, The University of Arizona Health Sciences, Tucson, Arizona 85724
| | - Weina Ju
- Department of Neurology, First Hospital of Jilin University, Jilin University, 71 Xinmin Street, Changchun, 130021, Jilin Province, China
- Department of Pharmacology, First Hospital of Jilin University, Jilin University, 71 Xinmin Street, Changchun, 130021, Jilin Province, China
| | - Aubin Moutal
- Department of Pharmacology, The University of Arizona Health Sciences, Tucson, Arizona 85724
| | - Samantha Perez-Miller
- Department of Pharmacology, The University of Arizona Health Sciences, Tucson, Arizona 85724
| | - Nickolay Brustovetsky
- Department of Pharmacology and Toxicology, and Stark Neurosciences Research Institute, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | - Vijay Gokhale
- Department of Pharmacology and Toxicology, College of Pharmacy, The University of Arizona Health Sciences, Tucson, Arizona 85724
| | - May Khanna
- Department of Pharmacology, The University of Arizona Health Sciences, Tucson, Arizona 85724
- The Center for Innovation in Brain Sciences, The University of Arizona Health Sciences, Tucson, Arizona 85724
| | - Rajesh Khanna
- Department of Pharmacology, The University of Arizona Health Sciences, Tucson, Arizona 85724
- Neuroscience Graduate Interdisciplinary Program, College of Medicine, The University of Arizona Health Sciences, Tucson, Arizona 85724
- The Center for Innovation in Brain Sciences, The University of Arizona Health Sciences, Tucson, Arizona 85724
| |
Collapse
|
123
|
Engineering Gain-of-Function Analogues of the Spider Venom Peptide HNTX-I, A Potent Blocker of the hNa V1.7 Sodium Channel. Toxins (Basel) 2018; 10:toxins10090358. [PMID: 30181499 PMCID: PMC6162447 DOI: 10.3390/toxins10090358] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2018] [Revised: 08/30/2018] [Accepted: 08/31/2018] [Indexed: 11/17/2022] Open
Abstract
Pain is a medical condition that interferes with normal human life and work and reduces human well-being worldwide. The voltage-gated sodium channel (VGSC) human NaV1.7 (hNaV1.7) is a compelling target that plays a key role in human pain signaling. The 33-residue peptide µ-TRTX-Hhn2b (HNTX-I), a member of NaV-targeting spider toxin (NaSpTx) family 1, has shown negligible activity on mammalian VGSCs, including the hNaV1.7 channel. We engineered analogues of HNTX-I based on sequence conservation in NaSpTx family 1. Substitution of Asn for Ser at position 23 or Asp for His at position 26 conferred potent activity against hNaV1.7. Moreover, multiple site mutations combined together afforded improvements in potency. Ultimately, we generated an analogue E1G⁻N23S⁻D26H⁻L32W with >300-fold improved potency compared with wild-type HNTX-1 on hNaV1.7 (IC50 0.036 ± 0.007 µM). Structural simulation suggested that the charged surface and the hydrophobic surface of the modified peptide are responsible for binding affinity to the hNaV1.7 channel, while variable residues may determine pharmacological specificity. Therefore, this study provides a profile for drug design targeting the hNaV1.7 channel.
Collapse
|
124
|
Pereira V, Millet Q, Aramburu J, Lopez-Rodriguez C, Gaveriaux-Ruff C, Wood JN. Analgesia linked to Nav1.7 loss of function requires µ- and δ-opioid receptors. Wellcome Open Res 2018; 3:101. [PMID: 30271888 PMCID: PMC6134336 DOI: 10.12688/wellcomeopenres.14687.1] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/07/2018] [Indexed: 01/08/2023] Open
Abstract
Background: Functional deletion of the Scn9a (sodium voltage-gated channel alpha subunit 9) gene encoding sodium channel Nav1.7 makes humans and mice pain-free. Opioid signalling contributes to this analgesic state. We have used pharmacological and genetic approaches to identify the opioid receptors involved in this form of analgesia. We also examined the regulation of proenkephalin expression by the transcription factor Nfat5 that binds upstream of the Penk gene. Methods: We used specific µ-, δ- and κ-opioid receptor antagonists alone or in combination to examine which opioid receptors were necessary for Nav1.7 loss-associated analgesia in mouse behavioural assays of thermal pain. We also used µ- and δ-opioid receptor null mutant mice alone and in combination in behavioural assays to examine the role of these receptors in Nav1.7 knockouts pain free phenotype. Finally, we examined the levels of Penk mRNA in Nfat5-null mutant mice, as this transcription factor binds to consensus sequences upstream of the Penk gene. Results: The pharmacological block or deletion of both µ- and δ-opioid receptors was required to abolish Nav1.7-null opioid-related analgesia. κ-opioid receptor antagonists were without effect. Enkephalins encoded by the Penk gene are upregulated in Nav1.7 nulls. Deleting Nfat5, a transcription factor with binding motifs upstream of Penk, induces the same level of enkephalin mRNA expression as found in Nav1 .7 nulls, but without consequent analgesia. These data confirm that a combination of events linked to Scn9a gene loss is required for analgesia. Higher levels of endogenous enkephalins, potentiated opioid receptors, diminished electrical excitability and loss of neurotransmitter release together contribute to the analgesic phenotype found in Nav1.7-null mouse and human mutants. Conclusions: These observations help explain the failure of Nav1.7 channel blockers alone to produce analgesia and suggest new routes for analgesic drug development.
Collapse
Affiliation(s)
- Vanessa Pereira
- Molecular Nociception Group, WIBR, University College London, Gower Street, WC1E 6BT, UK
| | - Queensta Millet
- Molecular Nociception Group, WIBR, University College London, Gower Street, WC1E 6BT, UK
| | - Jose Aramburu
- Immunology Unit, Department of Experimental and Health Sciences, Universitat Pompeu Fabra, Carrer Doctor Aiguader No88, 08003 Barcelona, Spain
| | - Cristina Lopez-Rodriguez
- Immunology Unit, Department of Experimental and Health Sciences, Universitat Pompeu Fabra, Carrer Doctor Aiguader No88, 08003 Barcelona, Spain
| | - Claire Gaveriaux-Ruff
- Institut de Génétique et de Biologie Moléculaire et Cellulaire, Université de Strasbourg, Centre National de la Recherche Scientifique , UMR7104, INSERM U1258, Ecole Supérieure de Biotechnologie de Strasbourg, Ilkirch, Strasbourg, France
| | - John N. Wood
- Molecular Nociception Group, WIBR, University College London, Gower Street, WC1E 6BT, UK
| |
Collapse
|
125
|
Yoshizawa K, Arai N, Suzuki Y, Nakamura T, Takeuchi K, Sakamoto R, Masuda R. Evaluation of the antinociceptive activities of several sodium channel blockers using veratrine test in mice. Synapse 2018; 72. [PMID: 29993143 DOI: 10.1002/syn.22056] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2018] [Revised: 06/11/2018] [Accepted: 06/21/2018] [Indexed: 12/19/2022]
Abstract
An important role of voltage-gated sodium channels (VGSCs) in many different pain states has been established in animal models and humans wherein sodium channel blockers partially ameliorate pain. However, behavioral tests for screening analgesics that exhibit pharmacologic action by acting on VGSCs are rarely reported, and there are no studies on antinociception using veratrine as a nociceptive agent. The aim of the present study was to examine the amount of nociceptive behavior evoked by subcutaneous administration of veratrine into the hind paw and investigate whether veratrine can be used as a VGSC agonist to test the pharmacological properties of candidate analgesics via sodium channel blockade. We report for the first time that intraplantar injection of veratrine produced a reproducible nociceptive response in mice. Furthermore, several sodium channel blockers, namely carbamazepine, valproate, mexiletine, and the selective Nav1.7 inhibitor PF-04856264, but not flecainide or pilsicainide, reduced veratrine-induced nociception. In contrast, calcium channel blockers gabapentin and ethosuximide did not change veratrine-induced nociception. The veratrine test in mice might be a useful tool, at least in part, to evaluate the potential analgesic effect of sodium channel blockers.
Collapse
Affiliation(s)
- Kazumi Yoshizawa
- Laboratory of Pharmacology and Therapeutics, Faculty of Pharmaceutical Sciences, Tokyo University of Science, Noda, Japan
| | - Narumaki Arai
- Laboratory of Pharmacology and Therapeutics, Faculty of Pharmaceutical Sciences, Tokyo University of Science, Noda, Japan
| | - Yukina Suzuki
- Laboratory of Pharmacology and Therapeutics, Faculty of Pharmaceutical Sciences, Tokyo University of Science, Noda, Japan
| | - Toka Nakamura
- Laboratory of Pharmacology and Therapeutics, Faculty of Pharmaceutical Sciences, Tokyo University of Science, Noda, Japan
| | - Kota Takeuchi
- Laboratory of Pharmacology and Therapeutics, Faculty of Pharmaceutical Sciences, Tokyo University of Science, Noda, Japan
| | - Reinii Sakamoto
- Department of Anesthesiology, Tokai University Hachioji Hospital, Tokyo, Japan
| | - Ritsuko Masuda
- Department of Anesthesiology, Tokai University Hachioji Hospital, Tokyo, Japan
| |
Collapse
|
126
|
Insights into the Contribution of Voltage-Gated Sodium Channel 1.7 to Paclitaxel-Induced Neuropathy. J Neurosci 2018; 38:6025-6027. [PMID: 29973416 DOI: 10.1523/jneurosci.0692-18.2018] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2018] [Revised: 05/18/2018] [Accepted: 05/24/2018] [Indexed: 11/21/2022] Open
|
127
|
A complicated complex: Ion channels, voltage sensing, cell membranes and peptide inhibitors. Neurosci Lett 2018; 679:35-47. [DOI: 10.1016/j.neulet.2018.04.030] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2018] [Revised: 04/11/2018] [Accepted: 04/17/2018] [Indexed: 01/04/2023]
|
128
|
Tao J, Jiang F, Liu C, Liu Z, Zhu Y, Xu J, Ge Y, Xu K, Yin P. Modulatory effects of bufalin, an active ingredient from toad venom on voltage-gated sodium channels. Mol Biol Rep 2018; 45:721-740. [PMID: 29931533 DOI: 10.1007/s11033-018-4213-9] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2018] [Accepted: 06/11/2018] [Indexed: 12/22/2022]
Abstract
Chan-su (toad venom) has been used as an analgesic agent in China from ancient to modern times. Bufalin, a non-peptide toxin extracted from toad venom, is considered as one of the analgesic components. The molecular mechanism underlying the anti-nociceptive effects of bufalin remains unclear so far. In this study, we investigated the pharmacological effects of bufalin on pain-related ion channels as well as animal models through patch clamping, calcium imaging and animal behavior observation. Using the whole-cell recording, bufalin caused remarkable suppressive effect on the peak currents of Nav channels (voltage gated sodium channels, VGSCs) of dorsal root ganglion neuroblastomas (ND7-23 cell) in a dose-dependent manner. Bufalin facilitated the voltage-dependent activation and induced a negative shift on the fast inactivation of VGSCs. The recovery kinetics of VGSCs were significantly slowed and the recovery proportion were reduced after administering bufalin. However, bufalin prompted no significant effect not only on Kv4.2, Kv4.3 and BK channels heterologously expressed in HEK293T cells, but also on the capsaicin and allyl isothiocyanate induced Ca2+ influx. What's more, bufalin could observably relieve formalin-induced spontaneous flinching and licking response as well as carrageenan-induced thermal and mechanical hyperalgesia in dose-dependent manner in agreement with the results of in vitro experiments. The present results imply that the remarkable anti-nociceptive effects produced by bufalin are probably ascribed to its specific regulation on Nav channels. Bufalin inhibits the Nav channels in a dose-dependent manner, which will provide references for the optimal dose selection of analgesia drugs.
Collapse
Affiliation(s)
- Jie Tao
- Department of Central Laboratory and Neurosurgery, Putuo Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China.
| | - Feng Jiang
- Xinhua Hospital (Chongming) Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai Chongming Xinhua Translational Medical Institute for Cancer Pain, Shanghai, China
| | - Cheng Liu
- Department of Central Laboratory and Neurosurgery, Putuo Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Zhirui Liu
- Department of Pharmacology, Institute of Medical Science, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yudan Zhu
- Department of Central Laboratory and Neurosurgery, Putuo Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Jian Xu
- Department of Central Laboratory and Neurosurgery, Putuo Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Yiqin Ge
- Department of Central Laboratory and Neurosurgery, Putuo Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Kan Xu
- Department of Central Laboratory and Neurosurgery, Putuo Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China.
| | - Peihao Yin
- Department of Central Laboratory and Neurosurgery, Putuo Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China.
| |
Collapse
|
129
|
Abstract
The mechanisms by which noxious stimuli produce the sensation of pain in animals are complex. Noxious stimuli are transduced at the periphery and transmitted to the CNS, where this information is subject to considerable modulation. Finally, the information is projected to the brain where it is perceived as pain. Additionally, plasticity can develop in the pain pathway and hyperalgesia and allodynia may develop through sensitisation both peripherally and centrally. A large number of different ion channels, receptors, and cell types are involved in pain perception, and it is hoped that through a better understanding of these, new and refined treatments for pain will result.
Collapse
Affiliation(s)
- A Bell
- School of Veterinary Medicine, University of Glasgow, Glasgow, G61 1QH, UK.
| |
Collapse
|
130
|
Co-expression of β Subunits with the Voltage-Gated Sodium Channel Na V1.7: the Importance of Subunit Association and Phosphorylation and Their Effects on Channel Pharmacology and Biophysics. J Mol Neurosci 2018; 65:154-166. [PMID: 29744740 DOI: 10.1007/s12031-018-1082-6] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2018] [Accepted: 05/01/2018] [Indexed: 12/19/2022]
Abstract
The voltage-gated sodium ion channel NaV1.7 is crucial in pain signaling. We examined how auxiliary β2 and β3 subunits and the phosphorylation state of the channel influence its biophysical properties and pharmacology. The human NaV1.7α subunit was co-expressed with either β2 or β3 subunits in HEK-293 cells. The β2 subunits and the NaV1.7α, however, were barely associated as evidenced by immunoprecipitation. Therefore, the β2 subunits did not change the biophysical properties of the channel. In contrast, β3 subunit was clearly associated with NaV1.7α. This subunit had a significant degree of glycosylation, and only the fully glycosylated β3 subunit was associated with the NaV1.7α. Electrophysiological characterisation revealed that the β3 subunit had small but consistent effects: a right-hand shift of the steady-state inactivation and faster recovery from inactivation. Furthermore, the β3 subunit reduced the susceptibility of NaV1.7α to several sodium channel blockers. In addition, we assessed the functional effect of NaV1.7α phosphorylation. Inhibition of kinase activity increased channel inactivation, while the blocking phosphatases produced the opposite effect. In conclusion, co-expression of β subunits with NaV1.7α, to better mimic the native channel properties, may be ineffective in cases when subunits are not associated, as shown in our experiments with β2. The β3 subunit significantly influences the function of NaV1.7α and, together with the phosphorylation of the channel, regulates its biophysical and pharmacological properties. These are important findings to take into account when considering the role of NaV1.7 channel in pain signaling.
Collapse
|
131
|
Moyer BD, Murray JK, Ligutti J, Andrews K, Favreau P, Jordan JB, Lee JH, Liu D, Long J, Sham K, Shi L, Stöcklin R, Wu B, Yin R, Yu V, Zou A, Biswas K, Miranda LP. Pharmacological characterization of potent and selective NaV1.7 inhibitors engineered from Chilobrachys jingzhao tarantula venom peptide JzTx-V. PLoS One 2018; 13:e0196791. [PMID: 29723257 PMCID: PMC5933747 DOI: 10.1371/journal.pone.0196791] [Citation(s) in RCA: 36] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2017] [Accepted: 04/19/2018] [Indexed: 11/18/2022] Open
Abstract
Identification of voltage-gated sodium channel NaV1.7 inhibitors for chronic pain therapeutic development is an area of vigorous pursuit. In an effort to identify more potent leads compared to our previously reported GpTx-1 peptide series, electrophysiology screening of fractionated tarantula venom discovered the NaV1.7 inhibitory peptide JzTx-V from the Chinese earth tiger tarantula Chilobrachys jingzhao. The parent peptide displayed nominal selectivity over the skeletal muscle NaV1.4 channel. Attribute-based positional scan analoging identified a key Ile28Glu mutation that improved NaV1.4 selectivity over 100-fold, and further optimization yielded the potent and selective peptide leads AM-8145 and AM-0422. NMR analyses revealed that the Ile28Glu substitution changed peptide conformation, pointing to a structural rationale for the selectivity gains. AM-8145 and AM-0422 as well as GpTx-1 and HwTx-IV competed for ProTx-II binding in HEK293 cells expressing human NaV1.7, suggesting that these NaV1.7 inhibitory peptides interact with a similar binding site. AM-8145 potently blocked native tetrodotoxin-sensitive (TTX-S) channels in mouse dorsal root ganglia (DRG) neurons, exhibited 30- to 120-fold selectivity over other human TTX-S channels and exhibited over 1,000-fold selectivity over other human tetrodotoxin-resistant (TTX-R) channels. Leveraging NaV1.7-NaV1.5 chimeras containing various voltage-sensor and pore regions, AM-8145 mapped to the second voltage-sensor domain of NaV1.7. AM-0422, but not the inactive peptide analog AM-8374, dose-dependently blocked capsaicin-induced DRG neuron action potential firing using a multi-electrode array readout and mechanically-induced C-fiber spiking in a saphenous skin-nerve preparation. Collectively, AM-8145 and AM-0422 represent potent, new engineered NaV1.7 inhibitory peptides derived from the JzTx-V scaffold with improved NaV selectivity and biological activity in blocking action potential firing in both DRG neurons and C-fibers.
Collapse
Affiliation(s)
- Bryan D. Moyer
- Neuroscience, Amgen Discovery Research, Thousand Oaks, California, United States of America
- * E-mail:
| | - Justin K. Murray
- Therapeutic Discovery, Amgen Discovery Research, Thousand Oaks, California, United States of America
| | - Joseph Ligutti
- Neuroscience, Amgen Discovery Research, Thousand Oaks, California, United States of America
| | - Kristin Andrews
- Molecular Engineering, Amgen Discovery Research, Cambridge, Massachusetts, United States of America
| | | | - John B. Jordan
- Discovery Attribute Sciences, Amgen Discovery Research, Thousand Oaks, California, United States of America
| | - Josie H. Lee
- Neuroscience, Amgen Discovery Research, Cambridge, Massachusetts, United States of America
| | - Dong Liu
- Neuroscience, Amgen Discovery Research, Thousand Oaks, California, United States of America
| | - Jason Long
- Therapeutic Discovery, Amgen Discovery Research, Thousand Oaks, California, United States of America
| | - Kelvin Sham
- Therapeutic Discovery, Amgen Discovery Research, Thousand Oaks, California, United States of America
| | - Licheng Shi
- Neuroscience, Amgen Discovery Research, Thousand Oaks, California, United States of America
| | - Reto Stöcklin
- Atheris Laboratories, CH Bernex, Geneva, Switzerland
| | - Bin Wu
- Therapeutic Discovery, Amgen Discovery Research, Thousand Oaks, California, United States of America
| | - Ruoyuan Yin
- Neuroscience, Amgen Discovery Research, Thousand Oaks, California, United States of America
| | - Violeta Yu
- Neuroscience, Amgen Discovery Research, Cambridge, Massachusetts, United States of America
| | - Anruo Zou
- Neuroscience, Amgen Discovery Research, Thousand Oaks, California, United States of America
| | - Kaustav Biswas
- Therapeutic Discovery, Amgen Discovery Research, Thousand Oaks, California, United States of America
| | - Les P. Miranda
- Therapeutic Discovery, Amgen Discovery Research, Thousand Oaks, California, United States of America
| |
Collapse
|
132
|
Zhou X, Zhang Y, Tang D, Liang S, Chen P, Tang C, Liu Z. A Chimeric NaV1.8 Channel Expression System Based on HEK293T Cell Line. Front Pharmacol 2018; 9:337. [PMID: 29686617 PMCID: PMC5900924 DOI: 10.3389/fphar.2018.00337] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2017] [Accepted: 03/22/2018] [Indexed: 11/13/2022] Open
Abstract
Among the nine voltage-gated sodium channel (NaV) subtypes, NaV1.8 is an attractive therapeutic target for pain. The heterologous expression of recombinant NaV1.8 currents is of particular importance for its electrophysiological and pharmacological studies. However, NaV1.8 expresses no or low-level functional currents when transiently transfected into non-neuronal cell lines. The present study aims to explore the molecular determinants limiting its functional expression and accordingly establish a functional NaV1.8 expression system. We conducted screening analysis of the NaV1.8 intracellular loops by constructing NaV chimeric channels and confirmed that the NaV1.8 C-terminus was the only limiting factor. Replacing this sequence with that of NaV1.4, NaV1.5, or NaV1.7 constructed functional channels (NaV1.8/1.4L5, NaV1.8/1.5L5, and NaV1.8/1.7L5, respectively), which expressed high-level NaV1.8-like currents in HEK293T cells. The chimeric channel NaV1.8/1.7L5 displayed much faster inactivation of its macroscopic currents than NaV1.8/1.4L5 and NaV1.8/1.5L5, and it was the most similar to wild-type NaV1.8 expressed in ND7/23 cells. Its currents were very stable during repetitive depolarizations, while its repriming kinetic was different from wild-type NaV1.8. Most importantly, NaV1.8/1.7L5 pharmacologically resembled wild-type NaV1.8 as revealed by testing their susceptibility to two NaV1.8 selective antagonists, APETx-2 and MrVIB. NaV chimeras study showed that at least the domain 2 and domain 4 of NaV1.8 were involved in binding with APETx-2. Our study provided new insights into the function of NaV1.8 intracellular loops, as well as a reliable and convenient expression system which could be useful in NaV1.8 studies.
Collapse
Affiliation(s)
| | | | | | | | | | - Cheng Tang
- The National and Local Joint Engineering Laboratory of Animal Peptide Drug Development, College of Life Sciences, Hunan Normal University, Changsha, China
| | - Zhonghua Liu
- The National and Local Joint Engineering Laboratory of Animal Peptide Drug Development, College of Life Sciences, Hunan Normal University, Changsha, China
| |
Collapse
|
133
|
Zheng YM, Wang WF, Li YF, Yu Y, Gao ZB. Enhancing inactivation rather than reducing activation of Nav1.7 channels by a clinically effective analgesic CNV1014802. Acta Pharmacol Sin 2018; 39:587-596. [PMID: 29094728 PMCID: PMC5888685 DOI: 10.1038/aps.2017.151] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/19/2017] [Accepted: 09/03/2017] [Indexed: 12/16/2022]
Abstract
The Nav1.7 channel represents a promising target for pain relief. In the recent decades, a number of Nav1.7 channel inhibitors have been developed. According to the effects on channel kinetics, these inhibitors could be divided into two major classes: reducing activation or enhancing inactivation. To date, however, only several inhibitors have moved forward into phase 2 clinical trials and most of them display a less than ideal analgesic efficacy, thus intensifying the controversy regarding if an ideal candidate should preferentially affect the activation or inactivation state. In the present study, we investigated the action mechanisms of a recently clinically confirmed inhibitor CNV1014802 using both electrophysiology and site-directed mutagenesis. We found that CNV1014802 inhibited Nav1.7 channels through stabilizing a nonconductive inactivated state. When the cells expressing Nav1.7 channels were hold at 70 mV or 120 mV, the half maximal inhibitory concentration (IC50) values (with 95% confidence limits) were 1.77 (1.20-2.33) and 71.66 (46.85-96.48) μmol/L, respectively. This drug caused dramatic hyperpolarizing shift of channel inactivation but did not affect activation. Moreover, CNV1014802 accelerated the onset of inactivation and delayed the recovery from inactivation. Notably, application of CNV1014802 (30 μmol/L) could rescue the Nav1.7 mutations expressed in CHO cells that cause paroxysmal extreme pain disorder (PEPD), thereby restoring the impaired inactivation to those of the wild-type channel. Our study demonstrates that CNV1014802 enhances the inactivation but does not reduce the activation of Nav1.7 channels, suggesting that identifying inhibitors that preferentially affect inactivation is a promising approach for developing drugs targeting Nav1.7.
Collapse
Affiliation(s)
- Yue-ming Zheng
- CAS Key Laboratory of Receptor Research, State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
| | - Wan-fu Wang
- CAS Key Laboratory of Receptor Research, State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
- Shanghai Key Laboratory of New Drug Design, School of Pharmacy, East China University of Science and Technology, Shanghai 200237, China
| | - Yan-fen Li
- CAS Key Laboratory of Receptor Research, State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
- Shanghai Key Laboratory of Bio-Energy Crops, School of Life Sciences, Shanghai University, Shanghai 200444, China
| | - Yong Yu
- Department of Neurosurgery, Zhongshan Hospital, Fudan University, Shanghai 200032, China
| | - Zhao-bing Gao
- CAS Key Laboratory of Receptor Research, State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
| |
Collapse
|
134
|
Foadi N. Modulation of sodium channels as pharmacological tool for pain therapy-highlights and gaps. Naunyn Schmiedebergs Arch Pharmacol 2018; 391:481-488. [PMID: 29572558 DOI: 10.1007/s00210-018-1487-3] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2018] [Accepted: 03/14/2018] [Indexed: 02/06/2023]
Abstract
Voltage-gated sodium channels are crucially involved in the transduction and transmission of nociceptive signals and pathological pain states. In the past decades, a lot of effort has been spent examining and characterizing biophysical properties of the different sodium channels and their role in signaling pathways. Several gains of function mutations of the sodium channels Nav1.7, Nav1.8, and Nav1.9 are associated with pain disorders. Due to their critical role in nociceptive pathways voltage-gated sodium channels are regarded interesting targets for pharmacological pain treatment. However we still need to fill the gap that exists in the translation of efficacy in preclinical in vitro experiments and in models of pain into the clinic. This review summarizes biological and electrophysiological properties of voltage-gated sodium channels and aims to discuss limitations and promising pharmacological strategies in sodium channel research in the context of pain therapy.
Collapse
Affiliation(s)
- Nilufar Foadi
- Clinic for Anaesthesia and Critical Care Medicine, Hannover Medical School, 30625, Hannover, Germany.
| |
Collapse
|
135
|
Efficacy of the Nav1.7 blocker PF-05089771 in a randomised, placebo-controlled, double-blind clinical study in subjects with painful diabetic peripheral neuropathy. Pain 2018; 159:1465-1476. [DOI: 10.1097/j.pain.0000000000001227] [Citation(s) in RCA: 90] [Impact Index Per Article: 12.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
|
136
|
Affiliation(s)
- Benjamin E. Blass
- Temple University School of Pharmacy, Moulder Center for Drug Discovery Research, Philadelphia, Pennsylvania 19140, United States
| |
Collapse
|
137
|
Zeng X, Li P, Chen B, Huang J, Lai R, Liu J, Rong M. Selective Closed-State Nav1.7 Blocker JZTX-34 Exhibits Analgesic Effects against Pain. Toxins (Basel) 2018; 10:toxins10020064. [PMID: 29393892 PMCID: PMC5848165 DOI: 10.3390/toxins10020064] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2017] [Revised: 01/26/2018] [Accepted: 01/31/2018] [Indexed: 12/11/2022] Open
Abstract
Jingzhaotoxin-34 (JZTX-34) is a selective inhibitor of tetrodotoxin-sensitive (TTX-S) sodium channels. In this study, we found that JZTX-34 selectively acted on Nav1.7 with little effect on other sodium channel subtypes including Nav1.5. If the DIIS3-S4 linker of Nav1.5 is substituted by the correspond linker of Nav1.7, the sensitivity of Nav1.5 to JZTX-34 extremely increases to 1.05 µM. Meanwhile, a mutant D816R in the DIIS3-S4 linker of Nav1.7 decreases binding affinity of Nav1.7 to JZTX-34 about 32-fold. The reverse mutant R800D at the corresponding position in Nav1.5 greatly increased its binding affinity to JZTX-34. This implies that JZTX-34 binds to DIIS3-S4 linker of Nav1.7 and the critical residue of Nav1.7 is D816. Unlike β-scorpion toxin trapping sodium channel in an open state, activity of JZTX-34 requires the sodium channel to be in a resting state. JZTX-34 exhibits an obvious analgesic effect in a rodent pain model. Especially, it shows a longer duration and is more effective than morphine in hot pain models. In a formalin-induced pain model, JZTX-34 at dose of 2 mg/kg is equipotent with morphine (5 mg/kg) in the first phase and several-fold more effective than morphine in second phase. Taken together, our data indicate that JZTX-34 releases pain by selectively binding to the domain II voltage sensor of Nav1.7 in a closed configuration.
Collapse
Affiliation(s)
- Xiongzhi Zeng
- The National & Local Joint Engineering Laboratory of Animal Peptide Drug Development, College of Life Sciences, Hunan Normal University, Changsha 410081, Hunan, China.
- The Key Laboratory of Protein Chemistry and Developmental Biology of Ministry of Education, College of Life Sciences, Hunan Normal University, Changsha 410081, Hunan, China.
| | - Pengpeng Li
- Life Sciences College of Nanjing Agricultural University, 210095, Jiangsu, China.
| | - Bo Chen
- The National & Local Joint Engineering Laboratory of Animal Peptide Drug Development, College of Life Sciences, Hunan Normal University, Changsha 410081, Hunan, China.
- The Key Laboratory of Protein Chemistry and Developmental Biology of Ministry of Education, College of Life Sciences, Hunan Normal University, Changsha 410081, Hunan, China.
| | - Juan Huang
- The National & Local Joint Engineering Laboratory of Animal Peptide Drug Development, College of Life Sciences, Hunan Normal University, Changsha 410081, Hunan, China.
- The Key Laboratory of Protein Chemistry and Developmental Biology of Ministry of Education, College of Life Sciences, Hunan Normal University, Changsha 410081, Hunan, China.
| | - Ren Lai
- Life Sciences College of Nanjing Agricultural University, 210095, Jiangsu, China.
- Key Laboratory of Animal Models and Human Disease Mechanisms of Chinese Academy of Sciences & Yunnan Province, Kunming Institute of Zoology, Kunming 650223, Yunnan, China.
| | - Jingze Liu
- Key Laboratory of Animal Physiology, Biochemistry and Molecular Biology of Hebei Province, College of Life Science, Hebei Normal University, Shijiazhuang 050024, Hebei, China.
| | - Mingqiang Rong
- The National & Local Joint Engineering Laboratory of Animal Peptide Drug Development, College of Life Sciences, Hunan Normal University, Changsha 410081, Hunan, China.
- The Key Laboratory of Protein Chemistry and Developmental Biology of Ministry of Education, College of Life Sciences, Hunan Normal University, Changsha 410081, Hunan, China.
| |
Collapse
|
138
|
Kanellopoulos AH, Koenig J, Huang H, Pyrski M, Millet Q, Lolignier S, Morohashi T, Gossage SJ, Jay M, Linley JE, Baskozos G, Kessler BM, Cox JJ, Dolphin AC, Zufall F, Wood JN, Zhao J. Mapping protein interactions of sodium channel Na V1.7 using epitope-tagged gene-targeted mice. EMBO J 2018; 37:427-445. [PMID: 29335280 PMCID: PMC5793798 DOI: 10.15252/embj.201796692] [Citation(s) in RCA: 49] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2017] [Revised: 11/30/2017] [Accepted: 12/05/2017] [Indexed: 11/24/2022] Open
Abstract
The voltage-gated sodium channel NaV1.7 plays a critical role in pain pathways. We generated an epitope-tagged NaV1.7 mouse that showed normal pain behaviours to identify channel-interacting proteins. Analysis of NaV1.7 complexes affinity-purified under native conditions by mass spectrometry revealed 267 proteins associated with Nav1.7 in vivo The sodium channel β3 (Scn3b), rather than the β1 subunit, complexes with Nav1.7, and we demonstrate an interaction between collapsing-response mediator protein (Crmp2) and Nav1.7, through which the analgesic drug lacosamide regulates Nav1.7 current density. Novel NaV1.7 protein interactors including membrane-trafficking protein synaptotagmin-2 (Syt2), L-type amino acid transporter 1 (Lat1) and transmembrane P24-trafficking protein 10 (Tmed10) together with Scn3b and Crmp2 were validated by co-immunoprecipitation (Co-IP) from sensory neuron extract. Nav1.7, known to regulate opioid receptor efficacy, interacts with the G protein-regulated inducer of neurite outgrowth (Gprin1), an opioid receptor-binding protein, demonstrating a physical and functional link between Nav1.7 and opioid signalling. Further information on physiological interactions provided with this normal epitope-tagged mouse should provide useful insights into the many functions now associated with the NaV1.7 channel.
Collapse
Affiliation(s)
| | - Jennifer Koenig
- Molecular Nociception Group, WIBR, University College London, London, UK
| | - Honglei Huang
- TDI Mass Spectrometry Laboratory, Target Discovery Institute, University of Oxford, Oxford, UK
| | - Martina Pyrski
- Center for Integrative Physiology and Molecular Medicine, Saarland University, Homburg, Germany
| | - Queensta Millet
- Molecular Nociception Group, WIBR, University College London, London, UK
| | - Stéphane Lolignier
- Molecular Nociception Group, WIBR, University College London, London, UK
- Université Clermont Auvergne, Inserm U1107 Neuro-Dol, Pharmacologie Fondamentale et Clinique de la Douleur, Clermont-Ferrand, France
| | - Toru Morohashi
- Molecular Nociception Group, WIBR, University College London, London, UK
| | - Samuel J Gossage
- Molecular Nociception Group, WIBR, University College London, London, UK
| | - Maude Jay
- Molecular Nociception Group, WIBR, University College London, London, UK
| | - John E Linley
- Molecular Nociception Group, WIBR, University College London, London, UK
- Neuroscience, IMED Biotech Unit, AstraZeneca, Cambridge, UK
| | | | - Benedikt M Kessler
- TDI Mass Spectrometry Laboratory, Target Discovery Institute, University of Oxford, Oxford, UK
| | - James J Cox
- Molecular Nociception Group, WIBR, University College London, London, UK
| | - Annette C Dolphin
- Department of Neuroscience, Physiology and Pharmacology, University College London, London, UK
| | - Frank Zufall
- Center for Integrative Physiology and Molecular Medicine, Saarland University, Homburg, Germany
| | - John N Wood
- Molecular Nociception Group, WIBR, University College London, London, UK
| | - Jing Zhao
- Molecular Nociception Group, WIBR, University College London, London, UK
| |
Collapse
|
139
|
Kato AS, Witkin JM. Protein complexes as psychiatric and neurological drug targets. Biochem Pharmacol 2018; 151:263-281. [PMID: 29330067 DOI: 10.1016/j.bcp.2018.01.018] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2017] [Accepted: 01/05/2018] [Indexed: 12/25/2022]
Abstract
The need for improved medications for psychiatric and neurological disorders is clear. Difficulties in finding such drugs demands that all strategic means be utilized for their invention. The discovery of forebrain specific AMPA receptor antagonists, which selectively block the specific combinations of principal and auxiliary subunits present in forebrain regions but spare targets in the cerebellum, was recently disclosed. This discovery raised the possibility that other auxiliary protein systems could be utilized to help identify new medicines. Discussion of the TARP-dependent AMPA receptor antagonists has been presented elsewhere. Here we review the diversity of protein complexes of neurotransmitter receptors in the nervous system to highlight the broad range of protein/protein drug targets. We briefly outline the structural basis of protein complexes as drug targets for G-protein-coupled receptors, voltage-gated ion channels, and ligand-gated ion channels. This review highlights heterodimers, subunit-specific receptor constructions, multiple signaling pathways, and auxiliary proteins with an emphasis on the later. We conclude that the use of auxiliary proteins in chemical compound screening could enhance the detection of specific, targeted drug searches and lead to novel and improved medicines for psychiatric and neurological disorders.
Collapse
Affiliation(s)
- Akihiko S Kato
- Neuroscience Discovery, Lilly Research Labs, Eli Lilly and Company, Indianapolis, IN, USA.
| | - Jeffrey M Witkin
- Neuroscience Discovery, Lilly Research Labs, Eli Lilly and Company, Indianapolis, IN, USA
| |
Collapse
|
140
|
Abstract
Voltage-gated sodium channels (VGSCs) are critical in generation and conduction of electrical signals in multiple excitable tissues. Natural toxins, produced by animal, plant, and microorganisms, target VGSCs through diverse strategies developed over millions of years of evolutions. Studying of the diverse interaction between VGSC and VGSC-targeting toxins has been contributing to the increasing understanding of molecular structure and function, pharmacology, and drug development potential of VGSCs. This chapter aims to summarize some of the current views on the VGSC-toxin interaction based on the established receptor sites of VGSC for natural toxins.
Collapse
Affiliation(s)
- Yonghua Ji
- Laboratory of Neuropharmacology and Neurotoxicology, Shanghai University, Shanghai, China.
| |
Collapse
|
141
|
Schaefer CP, Tome ME, Davis TP. The opioid epidemic: a central role for the blood brain barrier in opioid analgesia and abuse. Fluids Barriers CNS 2017; 14:32. [PMID: 29183383 PMCID: PMC5706290 DOI: 10.1186/s12987-017-0080-3] [Citation(s) in RCA: 51] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2017] [Accepted: 10/30/2017] [Indexed: 12/12/2022] Open
Abstract
Opioids are currently the primary treatment method used to manage both acute and chronic pain. In the past two to three decades, there has been a surge in the use, abuse and misuse of opioids. The mechanism by which opioids relieve pain and induce euphoria is dependent on the drug crossing the blood-brain barrier and accessing the central nervous system. This suggests the blood brain barrier plays a central role in both the benefits and risks of opioid use. The complex physiological responses to opioids that provide the benefits and drive the abuse also needs to be considered in the resolution of the opioid epidemic.
Collapse
Affiliation(s)
- Charles P. Schaefer
- Department of Pharmacology, University of Arizona, P.O. Box 245050, Tucson, AZ 85724 USA
| | - Margaret E. Tome
- Department of Pharmacology, University of Arizona, P.O. Box 245050, Tucson, AZ 85724 USA
| | - Thomas P. Davis
- Department of Pharmacology, University of Arizona, P.O. Box 245050, Tucson, AZ 85724 USA
| |
Collapse
|
142
|
Fouillet A, Watson JF, Piekarz AD, Huang X, Li B, Priest B, Nisenbaum E, Sher E, Ursu D. Characterisation of Nav1.7 functional expression in rat dorsal root ganglia neurons by using an electrical field stimulation assay. Mol Pain 2017; 13:1744806917745179. [PMID: 29166836 PMCID: PMC5731621 DOI: 10.1177/1744806917745179] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Background The Nav1.7 subtype of voltage-gated sodium channels is specifically expressed in sensory and sympathetic ganglia neurons where it plays an important role in the generation and transmission of information related to pain sensation. Human loss or gain-of-function mutations in the gene encoding Nav1.7 channels (SCN9A) are associated with either absence of pain, as reported for congenital insensitivity to pain, or with exacerbation of pain, as reported for primary erythromelalgia and paroxysmal extreme pain disorder. Based on this important human genetic evidence, numerous drug discovery efforts are ongoing in search for Nav1.7 blockers as a novel therapeutic strategy to treat pain conditions. Results We are reporting here a novel approach to study Nav1.7 function in cultured rat sensory neurons. We used live cell imaging combined with electrical field stimulation to evoke and record action potential-driven calcium transients in the neurons. We have shown that the tarantula venom peptide Protoxin-II, a known Nav1.7 subtype selective blocker, inhibited electrical field stimulation-evoked calcium responses in dorsal root ganglia neurons with an IC50 of 72 nM, while it had no activity in embryonic hippocampal neurons. The results obtained in the live cell imaging assay were supported by patch-clamp studies as well as by quantitative PCR and Western blotting experiments that confirmed the presence of Nav1.7 mRNA and protein in dorsal root ganglia but not in embryonic hippocampal neurons. Conclusions The findings presented here point to a selective effect of Protoxin-II in sensory neurons and helped to validate a new method for investigating and comparing Nav1.7 pharmacology in sensory versus central nervous system neurons. This will help in the characterisation of the selectivity of novel Nav1.7 modulators using native ion channels and will provide the basis for the development of higher throughput models for enabling pain-relevant phenotypic screening.
Collapse
Affiliation(s)
- Antoine Fouillet
- 1 Lilly Research Centre, 1539 Eli Lilly and Company , Windlesham, UK
| | - Jake F Watson
- 1 Lilly Research Centre, 1539 Eli Lilly and Company , Windlesham, UK
| | - Andrew D Piekarz
- 2 Lilly Research Laboratories, 1539 Eli Lilly and Company , IN, USA
| | - Xiaofang Huang
- 2 Lilly Research Laboratories, 1539 Eli Lilly and Company , IN, USA
| | - Baolin Li
- 2 Lilly Research Laboratories, 1539 Eli Lilly and Company , IN, USA
| | - Birgit Priest
- 2 Lilly Research Laboratories, 1539 Eli Lilly and Company , IN, USA
| | - Eric Nisenbaum
- 2 Lilly Research Laboratories, 1539 Eli Lilly and Company , IN, USA
| | - Emanuele Sher
- 1 Lilly Research Centre, 1539 Eli Lilly and Company , Windlesham, UK
| | - Daniel Ursu
- 1 Lilly Research Centre, 1539 Eli Lilly and Company , Windlesham, UK
| |
Collapse
|
143
|
Hilder TA, Robinson A, Chung SH. Functionalized Fullerene Targeting Human Voltage-Gated Sodium Channel, hNa v1.7. ACS Chem Neurosci 2017; 8:1747-1755. [PMID: 28586206 DOI: 10.1021/acschemneuro.7b00099] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/24/2023] Open
Abstract
Mutations of hNav1.7 that cause its activities to be enhanced contribute to severe neuropathic pain. Only a small number of hNav1.7 specific inhibitors have been identified, most of which interact with the voltage-sensing domain of the voltage-activated sodium ion channel. In our previous computational study, we demonstrated that a [Lys6]-C84 fullerene binds tightly (affinity of 46 nM) to NavAb, the voltage-gated sodium channel from the bacterium Arcobacter butzleri. Here, we extend this work and, using molecular dynamics simulations, demonstrate that the same [Lys6]-C84 fullerene binds strongly (2.7 nM) to the pore of a modeled human sodium ion channel hNav1.7. In contrast, the fullerene binds only weakly to a mutated model of hNav1.7 (I1399D) (14.5 mM) and a model of the skeletal muscle hNav1.4 (3.7 mM). Comparison of one representative sequence from each of the nine human sodium channel isoforms shows that only hNav1.7 possesses residues that are critical for binding the fullerene derivative and blocking the channel pore.
Collapse
Affiliation(s)
- Tamsyn A. Hilder
- School
of Chemical and Physical Sciences, Victoria University of Wellington, Wellington 6040, New Zealand
- Computational
Biophysics Group, Research School of Biology, Australian National University, Acton, ACT 2601, Australia
| | - Anna Robinson
- Computational
Biophysics Group, Research School of Biology, Australian National University, Acton, ACT 2601, Australia
| | - Shin-Ho Chung
- Computational
Biophysics Group, Research School of Biology, Australian National University, Acton, ACT 2601, Australia
| |
Collapse
|
144
|
Antihyperalgesic effect by herpes vector-mediated knockdown of NaV1.7 sodium channels after skin incision. Neuroreport 2017; 28:661-665. [DOI: 10.1097/wnr.0000000000000814] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
|
145
|
Kornecook TJ, Yin R, Altmann S, Be X, Berry V, Ilch CP, Jarosh M, Johnson D, Lee JH, Lehto SG, Ligutti J, Liu D, Luther J, Matson D, Ortuno D, Roberts J, Taborn K, Wang J, Weiss MM, Yu V, Zhu DXD, Fremeau RT, Moyer BD. Pharmacologic Characterization of AMG8379, a Potent and Selective Small Molecule Sulfonamide Antagonist of the Voltage-Gated Sodium Channel Na V1.7. J Pharmacol Exp Ther 2017; 362:146-160. [PMID: 28473457 DOI: 10.1124/jpet.116.239590] [Citation(s) in RCA: 44] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2016] [Accepted: 05/03/2017] [Indexed: 03/08/2025] Open
Abstract
Potent and selective antagonists of the voltage-gated sodium channel NaV1.7 represent a promising avenue for the development of new chronic pain therapies. We generated a small molecule atropisomer quinolone sulfonamide antagonist AMG8379 and a less active enantiomer AMG8380. Here we show that AMG8379 potently blocks human NaV1.7 channels with an IC50 of 8.5 nM and endogenous tetrodotoxin (TTX)-sensitive sodium channels in dorsal root ganglion (DRG) neurons with an IC50 of 3.1 nM in whole-cell patch clamp electrophysiology assays using a voltage protocol that interrogates channels in a partially inactivated state. AMG8379 was 100- to 1000-fold selective over other NaV family members, including NaV1.4 expressed in muscle and NaV1.5 expressed in the heart, as well as TTX-resistant NaV channels in DRG neurons. Using an ex vivo mouse skin-nerve preparation, AMG8379 blocked mechanically induced action potential firing in C-fibers in both a time-dependent and dose-dependent manner. AMG8379 similarly reduced the frequency of thermally induced C-fiber spiking, whereas AMG8380 affected neither mechanical nor thermal responses. In vivo target engagement of AMG8379 in mice was evaluated in multiple NaV1.7-dependent behavioral endpoints. AMG8379 dose-dependently inhibited intradermal histamine-induced scratching and intraplantar capsaicin-induced licking, and reversed UVB radiation skin burn-induced thermal hyperalgesia; notably, behavioral effects were not observed with AMG8380 at similar plasma exposure levels. AMG8379 is a potent and selective NaV1.7 inhibitor that blocks sodium current in heterologous cells as well as DRG neurons, inhibits action potential firing in peripheral nerve fibers, and exhibits pharmacodynamic effects in translatable models of both itch and pain.
Collapse
Affiliation(s)
- Thomas J Kornecook
- Department of Neuroscience (T.J.K., R.Y., S.A, C.P.I., M.J., D.J., J.H.L., S.G.L., J.Li., D.L., J.Lu., D.M., D.O., K.T., J.W., V.Y., D.X.D.Z., R.T.F., B.D.M.), Department of Medicinal Chemistry (M.M.W.), and Department of Pharmacokinetics and Drug Metabolism (X.B., V.B., J.R.), Amgen Inc., Cambridge, Massachusetts and Thousand Oaks, California
| | - Ruoyuan Yin
- Department of Neuroscience (T.J.K., R.Y., S.A, C.P.I., M.J., D.J., J.H.L., S.G.L., J.Li., D.L., J.Lu., D.M., D.O., K.T., J.W., V.Y., D.X.D.Z., R.T.F., B.D.M.), Department of Medicinal Chemistry (M.M.W.), and Department of Pharmacokinetics and Drug Metabolism (X.B., V.B., J.R.), Amgen Inc., Cambridge, Massachusetts and Thousand Oaks, California
| | - Stephen Altmann
- Department of Neuroscience (T.J.K., R.Y., S.A, C.P.I., M.J., D.J., J.H.L., S.G.L., J.Li., D.L., J.Lu., D.M., D.O., K.T., J.W., V.Y., D.X.D.Z., R.T.F., B.D.M.), Department of Medicinal Chemistry (M.M.W.), and Department of Pharmacokinetics and Drug Metabolism (X.B., V.B., J.R.), Amgen Inc., Cambridge, Massachusetts and Thousand Oaks, California
| | - Xuhai Be
- Department of Neuroscience (T.J.K., R.Y., S.A, C.P.I., M.J., D.J., J.H.L., S.G.L., J.Li., D.L., J.Lu., D.M., D.O., K.T., J.W., V.Y., D.X.D.Z., R.T.F., B.D.M.), Department of Medicinal Chemistry (M.M.W.), and Department of Pharmacokinetics and Drug Metabolism (X.B., V.B., J.R.), Amgen Inc., Cambridge, Massachusetts and Thousand Oaks, California
| | - Virginia Berry
- Department of Neuroscience (T.J.K., R.Y., S.A, C.P.I., M.J., D.J., J.H.L., S.G.L., J.Li., D.L., J.Lu., D.M., D.O., K.T., J.W., V.Y., D.X.D.Z., R.T.F., B.D.M.), Department of Medicinal Chemistry (M.M.W.), and Department of Pharmacokinetics and Drug Metabolism (X.B., V.B., J.R.), Amgen Inc., Cambridge, Massachusetts and Thousand Oaks, California
| | - Christopher P Ilch
- Department of Neuroscience (T.J.K., R.Y., S.A, C.P.I., M.J., D.J., J.H.L., S.G.L., J.Li., D.L., J.Lu., D.M., D.O., K.T., J.W., V.Y., D.X.D.Z., R.T.F., B.D.M.), Department of Medicinal Chemistry (M.M.W.), and Department of Pharmacokinetics and Drug Metabolism (X.B., V.B., J.R.), Amgen Inc., Cambridge, Massachusetts and Thousand Oaks, California
| | - Michael Jarosh
- Department of Neuroscience (T.J.K., R.Y., S.A, C.P.I., M.J., D.J., J.H.L., S.G.L., J.Li., D.L., J.Lu., D.M., D.O., K.T., J.W., V.Y., D.X.D.Z., R.T.F., B.D.M.), Department of Medicinal Chemistry (M.M.W.), and Department of Pharmacokinetics and Drug Metabolism (X.B., V.B., J.R.), Amgen Inc., Cambridge, Massachusetts and Thousand Oaks, California
| | - Danielle Johnson
- Department of Neuroscience (T.J.K., R.Y., S.A, C.P.I., M.J., D.J., J.H.L., S.G.L., J.Li., D.L., J.Lu., D.M., D.O., K.T., J.W., V.Y., D.X.D.Z., R.T.F., B.D.M.), Department of Medicinal Chemistry (M.M.W.), and Department of Pharmacokinetics and Drug Metabolism (X.B., V.B., J.R.), Amgen Inc., Cambridge, Massachusetts and Thousand Oaks, California
| | - Josie H Lee
- Department of Neuroscience (T.J.K., R.Y., S.A, C.P.I., M.J., D.J., J.H.L., S.G.L., J.Li., D.L., J.Lu., D.M., D.O., K.T., J.W., V.Y., D.X.D.Z., R.T.F., B.D.M.), Department of Medicinal Chemistry (M.M.W.), and Department of Pharmacokinetics and Drug Metabolism (X.B., V.B., J.R.), Amgen Inc., Cambridge, Massachusetts and Thousand Oaks, California
| | - Sonya G Lehto
- Department of Neuroscience (T.J.K., R.Y., S.A, C.P.I., M.J., D.J., J.H.L., S.G.L., J.Li., D.L., J.Lu., D.M., D.O., K.T., J.W., V.Y., D.X.D.Z., R.T.F., B.D.M.), Department of Medicinal Chemistry (M.M.W.), and Department of Pharmacokinetics and Drug Metabolism (X.B., V.B., J.R.), Amgen Inc., Cambridge, Massachusetts and Thousand Oaks, California
| | - Joseph Ligutti
- Department of Neuroscience (T.J.K., R.Y., S.A, C.P.I., M.J., D.J., J.H.L., S.G.L., J.Li., D.L., J.Lu., D.M., D.O., K.T., J.W., V.Y., D.X.D.Z., R.T.F., B.D.M.), Department of Medicinal Chemistry (M.M.W.), and Department of Pharmacokinetics and Drug Metabolism (X.B., V.B., J.R.), Amgen Inc., Cambridge, Massachusetts and Thousand Oaks, California
| | - Dong Liu
- Department of Neuroscience (T.J.K., R.Y., S.A, C.P.I., M.J., D.J., J.H.L., S.G.L., J.Li., D.L., J.Lu., D.M., D.O., K.T., J.W., V.Y., D.X.D.Z., R.T.F., B.D.M.), Department of Medicinal Chemistry (M.M.W.), and Department of Pharmacokinetics and Drug Metabolism (X.B., V.B., J.R.), Amgen Inc., Cambridge, Massachusetts and Thousand Oaks, California
| | - Jason Luther
- Department of Neuroscience (T.J.K., R.Y., S.A, C.P.I., M.J., D.J., J.H.L., S.G.L., J.Li., D.L., J.Lu., D.M., D.O., K.T., J.W., V.Y., D.X.D.Z., R.T.F., B.D.M.), Department of Medicinal Chemistry (M.M.W.), and Department of Pharmacokinetics and Drug Metabolism (X.B., V.B., J.R.), Amgen Inc., Cambridge, Massachusetts and Thousand Oaks, California
| | - David Matson
- Department of Neuroscience (T.J.K., R.Y., S.A, C.P.I., M.J., D.J., J.H.L., S.G.L., J.Li., D.L., J.Lu., D.M., D.O., K.T., J.W., V.Y., D.X.D.Z., R.T.F., B.D.M.), Department of Medicinal Chemistry (M.M.W.), and Department of Pharmacokinetics and Drug Metabolism (X.B., V.B., J.R.), Amgen Inc., Cambridge, Massachusetts and Thousand Oaks, California
| | - Danny Ortuno
- Department of Neuroscience (T.J.K., R.Y., S.A, C.P.I., M.J., D.J., J.H.L., S.G.L., J.Li., D.L., J.Lu., D.M., D.O., K.T., J.W., V.Y., D.X.D.Z., R.T.F., B.D.M.), Department of Medicinal Chemistry (M.M.W.), and Department of Pharmacokinetics and Drug Metabolism (X.B., V.B., J.R.), Amgen Inc., Cambridge, Massachusetts and Thousand Oaks, California
| | - John Roberts
- Department of Neuroscience (T.J.K., R.Y., S.A, C.P.I., M.J., D.J., J.H.L., S.G.L., J.Li., D.L., J.Lu., D.M., D.O., K.T., J.W., V.Y., D.X.D.Z., R.T.F., B.D.M.), Department of Medicinal Chemistry (M.M.W.), and Department of Pharmacokinetics and Drug Metabolism (X.B., V.B., J.R.), Amgen Inc., Cambridge, Massachusetts and Thousand Oaks, California
| | - Kristin Taborn
- Department of Neuroscience (T.J.K., R.Y., S.A, C.P.I., M.J., D.J., J.H.L., S.G.L., J.Li., D.L., J.Lu., D.M., D.O., K.T., J.W., V.Y., D.X.D.Z., R.T.F., B.D.M.), Department of Medicinal Chemistry (M.M.W.), and Department of Pharmacokinetics and Drug Metabolism (X.B., V.B., J.R.), Amgen Inc., Cambridge, Massachusetts and Thousand Oaks, California
| | - Jinti Wang
- Department of Neuroscience (T.J.K., R.Y., S.A, C.P.I., M.J., D.J., J.H.L., S.G.L., J.Li., D.L., J.Lu., D.M., D.O., K.T., J.W., V.Y., D.X.D.Z., R.T.F., B.D.M.), Department of Medicinal Chemistry (M.M.W.), and Department of Pharmacokinetics and Drug Metabolism (X.B., V.B., J.R.), Amgen Inc., Cambridge, Massachusetts and Thousand Oaks, California
| | - Matthew M Weiss
- Department of Neuroscience (T.J.K., R.Y., S.A, C.P.I., M.J., D.J., J.H.L., S.G.L., J.Li., D.L., J.Lu., D.M., D.O., K.T., J.W., V.Y., D.X.D.Z., R.T.F., B.D.M.), Department of Medicinal Chemistry (M.M.W.), and Department of Pharmacokinetics and Drug Metabolism (X.B., V.B., J.R.), Amgen Inc., Cambridge, Massachusetts and Thousand Oaks, California
| | - Violeta Yu
- Department of Neuroscience (T.J.K., R.Y., S.A, C.P.I., M.J., D.J., J.H.L., S.G.L., J.Li., D.L., J.Lu., D.M., D.O., K.T., J.W., V.Y., D.X.D.Z., R.T.F., B.D.M.), Department of Medicinal Chemistry (M.M.W.), and Department of Pharmacokinetics and Drug Metabolism (X.B., V.B., J.R.), Amgen Inc., Cambridge, Massachusetts and Thousand Oaks, California
| | - Dawn X D Zhu
- Department of Neuroscience (T.J.K., R.Y., S.A, C.P.I., M.J., D.J., J.H.L., S.G.L., J.Li., D.L., J.Lu., D.M., D.O., K.T., J.W., V.Y., D.X.D.Z., R.T.F., B.D.M.), Department of Medicinal Chemistry (M.M.W.), and Department of Pharmacokinetics and Drug Metabolism (X.B., V.B., J.R.), Amgen Inc., Cambridge, Massachusetts and Thousand Oaks, California
| | - Robert T Fremeau
- Department of Neuroscience (T.J.K., R.Y., S.A, C.P.I., M.J., D.J., J.H.L., S.G.L., J.Li., D.L., J.Lu., D.M., D.O., K.T., J.W., V.Y., D.X.D.Z., R.T.F., B.D.M.), Department of Medicinal Chemistry (M.M.W.), and Department of Pharmacokinetics and Drug Metabolism (X.B., V.B., J.R.), Amgen Inc., Cambridge, Massachusetts and Thousand Oaks, California
| | - Bryan D Moyer
- Department of Neuroscience (T.J.K., R.Y., S.A, C.P.I., M.J., D.J., J.H.L., S.G.L., J.Li., D.L., J.Lu., D.M., D.O., K.T., J.W., V.Y., D.X.D.Z., R.T.F., B.D.M.), Department of Medicinal Chemistry (M.M.W.), and Department of Pharmacokinetics and Drug Metabolism (X.B., V.B., J.R.), Amgen Inc., Cambridge, Massachusetts and Thousand Oaks, California
| |
Collapse
|
146
|
Berta T, Qadri Y, Tan PH, Ji RR. Targeting dorsal root ganglia and primary sensory neurons for the treatment of chronic pain. Expert Opin Ther Targets 2017; 21:695-703. [PMID: 28480765 DOI: 10.1080/14728222.2017.1328057] [Citation(s) in RCA: 162] [Impact Index Per Article: 20.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023]
Abstract
INTRODUCTION Currently the treatment of chronic pain is inadequate and compromised by debilitating central nervous system side effects. Here we discuss new therapeutic strategies that target dorsal root ganglia (DRGs) in the peripheral nervous system for a better and safer treatment of chronic pain. Areas covered: The DRGs contain the cell bodies of primary sensory neurons including nociceptive neurons. After painful injuries, primary sensory neurons demonstrate maladaptive molecular changes in DRG cell bodies and in their axons. These changes result in hypersensitivity and hyperexcitability of sensory neurons (peripheral sensitization) and are crucial for the onset and maintenance of chronic pain. We discuss the following new strategies to target DRGs and primary sensory neurons as a means of alleviating chronic pain and minimizing side effects: inhibition of sensory neuron-expressing ion channels such as TRPA1, TRPV1, and Nav1.7, selective blockade of C- and Aβ-afferent fibers, gene therapy, and implantation of bone marrow stem cells. Expert opinion: These peripheral pharmacological treatments, as well as gene and cell therapies, aimed at DRG tissues and primary sensory neurons can offer better and safer treatments for inflammatory, neuropathic, cancer, and other chronic pain states.
Collapse
Affiliation(s)
- Temugin Berta
- a Pain Research Center, Department of Anesthesiology , University of Cincinnati Medical Center , Cincinnati , OH , USA
| | - Yawar Qadri
- b Department of Anesthesiology , Duke University Medical Center , Durham , NC , USA
| | - Ping-Heng Tan
- c Department of Anesthesiology, E-Da Hospital, School of Medicine , I-Shou University , Kaohsiung , Taiwan
| | - Ru-Rong Ji
- b Department of Anesthesiology , Duke University Medical Center , Durham , NC , USA.,d Department of Neurobiology , Duke University Medical Center , Durham , NC , USA
| |
Collapse
|
147
|
Feldman EL, Nave KA, Jensen TS, Bennett DLH. New Horizons in Diabetic Neuropathy: Mechanisms, Bioenergetics, and Pain. Neuron 2017; 93:1296-1313. [PMID: 28334605 PMCID: PMC5400015 DOI: 10.1016/j.neuron.2017.02.005] [Citation(s) in RCA: 602] [Impact Index Per Article: 75.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2016] [Revised: 02/02/2017] [Accepted: 02/02/2017] [Indexed: 12/13/2022]
Abstract
Pre-diabetes and diabetes are a global epidemic, and the associated neuropathic complications create a substantial burden on both the afflicted patients and society as a whole. Given the enormity of the problem and the lack of effective therapies, there is a pressing need to understand the mechanisms underlying diabetic neuropathy (DN). In this review, we present the structural components of the peripheral nervous system that underlie its susceptibility to metabolic insults and then discuss the pathways that contribute to peripheral nerve injury in DN. We also discuss systems biology insights gleaned from the recent advances in biotechnology and bioinformatics, emerging ideas centered on the axon-Schwann cell relationship and associated bioenergetic crosstalk, and the rapid expansion of our knowledge of the mechanisms contributing to neuropathic pain in diabetes. These recent advances in our understanding of DN pathogenesis are paving the way for critical mechanism-based therapy development.
Collapse
Affiliation(s)
- Eva L Feldman
- Department of Neurology, University of Michigan, Ann Arbor, MI 48109, USA.
| | - Klaus-Armin Nave
- Department of Neurogenetics, Max Planck Institute for Experimental Medicine, 37075 Göttingen, Germany
| | - Troels S Jensen
- Department of Neurology and Danish Pain Research Center, Aarhus University, 8000 Aarhus C, Denmark
| | - David L H Bennett
- Nuffield Department of Clinical Neuroscience, University of Oxford, Oxford OX3 9DU, UK
| |
Collapse
|
148
|
Inhibition of α9α10 nicotinic acetylcholine receptors prevents chemotherapy-induced neuropathic pain. Proc Natl Acad Sci U S A 2017; 114:E1825-E1832. [PMID: 28223528 DOI: 10.1073/pnas.1621433114] [Citation(s) in RCA: 124] [Impact Index Per Article: 15.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023] Open
Abstract
Opioids are first-line drugs for moderate to severe acute pain and cancer pain. However, these medications are associated with severe side effects, and whether they are efficacious in treatment of chronic nonmalignant pain remains controversial. Medications that act through alternative molecular mechanisms are critically needed. Antagonists of α9α10 nicotinic acetylcholine receptors (nAChRs) have been proposed as an important nonopioid mechanism based on studies demonstrating prevention of neuropathology after trauma-induced nerve injury. However, the key α9α10 ligands characterized to date are at least two orders of magnitude less potent on human vs. rodent nAChRs, limiting their translational application. Furthermore, an alternative proposal that these ligands achieve their beneficial effects by acting as agonists of GABAB receptors has caused confusion over whether blockade of α9α10 nAChRs is the fundamental underlying mechanism. To address these issues definitively, we developed RgIA4, a peptide that exhibits high potency for both human and rodent α9α10 nAChRs, and was at least 1,000-fold more selective for α9α10 nAChRs vs. all other molecular targets tested, including opioid and GABAB receptors. A daily s.c. dose of RgIA4 prevented chemotherapy-induced neuropathic pain in rats. In wild-type mice, oxaliplatin treatment produced cold allodynia that could be prevented by RgIA4. Additionally, in α9 KO mice, chemotherapy-induced development of cold allodynia was attenuated and the milder, temporary cold allodynia was not relieved by RgIA4. These findings establish blockade of α9-containing nAChRs as the basis for the efficacy of RgIA4, and that α9-containing nAChRs are a critical target for prevention of chronic cancer chemotherapy-induced neuropathic pain.
Collapse
|
149
|
Colloca L, Ludman T, Bouhassira D, Baron R, Dickenson AH, Yarnitsky D, Freeman R, Truini A, Attal N, Finnerup NB, Eccleston C, Kalso E, Bennett DL, Dworkin RH, Raja SN. Neuropathic pain. Nat Rev Dis Primers 2017; 3:17002. [PMID: 28205574 PMCID: PMC5371025 DOI: 10.1038/nrdp.2017.2] [Citation(s) in RCA: 1357] [Impact Index Per Article: 169.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Neuropathic pain is caused by a lesion or disease of the somatosensory system, including peripheral fibres (Aβ, Aδ and C fibres) and central neurons, and affects 7-10% of the general population. Multiple causes of neuropathic pain have been described and its incidence is likely to increase owing to the ageing global population, increased incidence of diabetes mellitus and improved survival from cancer after chemotherapy. Indeed, imbalances between excitatory and inhibitory somatosensory signalling, alterations in ion channels and variability in the way that pain messages are modulated in the central nervous system all have been implicated in neuropathic pain. The burden of chronic neuropathic pain seems to be related to the complexity of neuropathic symptoms, poor outcomes and difficult treatment decisions. Importantly, quality of life is impaired in patients with neuropathic pain owing to increased drug prescriptions and visits to health care providers, as well as the morbidity from the pain itself and the inciting disease. Despite challenges, progress in the understanding of the pathophysiology of neuropathic pain is spurring the development of new diagnostic procedures and personalized interventions, which emphasize the need for a multidisciplinary approach to the management of neuropathic pain.
Collapse
Affiliation(s)
- Luana Colloca
- Department of Pain and Translational Symptom Science, School of Nursing and Department of Anesthesiology School of Medicine, University of Maryland, 655 West Lombard Street, 21201 Baltimore, Maryland, USA
| | - Taylor Ludman
- Department of Pain and Translational Symptom Science, School of Nursing and Department of Anesthesiology School of Medicine, University of Maryland, 655 West Lombard Street, 21201 Baltimore, Maryland, USA
| | - Didier Bouhassira
- INSERM, Unit 987, Ambroise Paré Hospital, UVSQ, Boulogne Billancourt, France
| | - Ralf Baron
- Department of Neurology, Division of Neurological Pain Research and Therapy, Klinik fur Neurologie Christian-Albrechts-Universität Kiel, Kiel, Germany
| | - Anthony H Dickenson
- Department of Neuroscience, Physiology and Pharmacology, University College London, London, UK
| | - David Yarnitsky
- Department of Neurology, Rambam Health Care Campus, Technion Faculty of Medicine, Haifa, Israel
| | - Roy Freeman
- Department of Neurology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts, USA
| | - Andrea Truini
- Department of Neurology and Psychiatry, Sapienza University, Rome, Italy
| | - Nadine Attal
- Pain Evaluation and Treatment Centre of Hôpital Ambroise Paré, Paris, France
| | - Nanna B Finnerup
- Department of Clinical Medicine - The Danish Pain Research Center, Aarhus University, Aarhus, Denmark
| | - Christopher Eccleston
- Centre for Pain Research, University of Bath, Bath, UK
- Department of Clinical and Health Psychology, Ghent University, Ghent, Belgium
| | - Eija Kalso
- Division of Pain Medicine, Department of Anesthesiology, Intensive Care and Pain Medicine, University of Helsinki and Helsinki University Hospital, Helsinki, Finland
| | - David L Bennett
- Nuffield Department of Clinical Neuroscience, University of Oxford, Oxford, UK
| | - Robert H Dworkin
- Department of Anesthesiology, School of Medicine and Dentistry, University of Rochester, Rochester, New York, USA
| | - Srinivasa N Raja
- Department of Anesthesiology and Critical Care Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| |
Collapse
|
150
|
Colloca L, Ludman T, Bouhassira D, Baron R, Dickenson AH, Yarnitsky D, Freeman R, Truini A, Attal N, Finnerup NB, Eccleston C, Kalso E, Bennett DL, Dworkin RH, Raja SN. Neuropathic pain. Nat Rev Dis Primers 2017; 3:17002. [PMID: 28205574 DOI: 10.1038/nrdp.2017.3] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 05/25/2023]
Abstract
Neuropathic pain is caused by a lesion or disease of the somatosensory system, including peripheral fibres (Aβ, Aδ and C fibres) and central neurons, and affects 7-10% of the general population. Multiple causes of neuropathic pain have been described and its incidence is likely to increase owing to the ageing global population, increased incidence of diabetes mellitus and improved survival from cancer after chemotherapy. Indeed, imbalances between excitatory and inhibitory somatosensory signalling, alterations in ion channels and variability in the way that pain messages are modulated in the central nervous system all have been implicated in neuropathic pain. The burden of chronic neuropathic pain seems to be related to the complexity of neuropathic symptoms, poor outcomes and difficult treatment decisions. Importantly, quality of life is impaired in patients with neuropathic pain owing to increased drug prescriptions and visits to health care providers, as well as the morbidity from the pain itself and the inciting disease. Despite challenges, progress in the understanding of the pathophysiology of neuropathic pain is spurring the development of new diagnostic procedures and personalized interventions, which emphasize the need for a multidisciplinary approach to the management of neuropathic pain.
Collapse
Affiliation(s)
- Luana Colloca
- Department of Pain and Translational Symptom Science, School of Nursing and Department of Anesthesiology School of Medicine, University of Maryland, 655 West Lombard Street, 21201 Baltimore, Maryland, USA
| | - Taylor Ludman
- Department of Pain and Translational Symptom Science, School of Nursing and Department of Anesthesiology School of Medicine, University of Maryland, 655 West Lombard Street, 21201 Baltimore, Maryland, USA
| | - Didier Bouhassira
- INSERM, Unit 987, Ambroise Paré Hospital, UVSQ, Boulogne Billancourt, France
| | - Ralf Baron
- Department of Neurology, Division of Neurological Pain Research and Therapy, Klinik fur Neurologie Christian-Albrechts-Universität Kiel, Kiel, Germany
| | - Anthony H Dickenson
- Department of Neuroscience, Physiology and Pharmacology, University College London, London, UK
| | - David Yarnitsky
- Department of Neurology, Rambam Health Care Campus, Technion Faculty of Medicine, Haifa, Israel
| | - Roy Freeman
- Department of Neurology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts, USA
| | - Andrea Truini
- Department of Neurology and Psychiatry, Sapienza University, Rome, Italy
| | - Nadine Attal
- Pain Evaluation and Treatment Centre of Hôpital Ambroise Paré, Paris, France
| | - Nanna B Finnerup
- Department of Clinical Medicine - The Danish Pain Research Center, Aarhus University, Aarhus, Denmark
| | - Christopher Eccleston
- Centre for Pain Research, University of Bath, Bath, UK
- Department of Clinical and Health Psychology, Ghent University, Ghent, Belgium
| | - Eija Kalso
- Division of Pain Medicine, Department of Anesthesiology, Intensive Care and Pain Medicine, University of Helsinki and Helsinki University Hospital, Helsinki, Finland
| | - David L Bennett
- Nuffield Department of Clinical Neuroscience, University of Oxford, Oxford, UK
| | - Robert H Dworkin
- Department of Anesthesiology, School of Medicine and Dentistry, University of Rochester, Rochester, New York, USA
| | - Srinivasa N Raja
- Department of Anesthesiology and Critical Care Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| |
Collapse
|