251
|
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
|
252
|
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
|
253
|
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
|
254
|
Naimo GD, Guarnaccia M, Sprovieri T, Ungaro C, Conforti FL, Andò S, Cavallaro S. A Systems Biology Approach for Personalized Medicine in Refractory Epilepsy. Int J Mol Sci 2019; 20:E3717. [PMID: 31366017 PMCID: PMC6695675 DOI: 10.3390/ijms20153717] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2019] [Revised: 07/22/2019] [Accepted: 07/28/2019] [Indexed: 02/01/2023] Open
Abstract
Epilepsy refers to a common chronic neurological disorder that affects all age groups. Unfortunately, antiepileptic drugs are ineffective in about one-third of patients. The complex interindividual variability influences the response to drug treatment rendering the therapeutic failure one of the most relevant problems in clinical practice also for increased hospitalizations and healthcare costs. Recent advances in the genetics and neurobiology of epilepsies are laying the groundwork for a new personalized medicine, focused on the reversal or avoidance of the pathophysiological effects of specific gene mutations. This could lead to a significant improvement in the efficacy and safety of treatments for epilepsy, targeting the biological mechanisms responsible for epilepsy in each individual. In this review article, we focus on the mechanism of the epilepsy pharmacoresistance and highlight the use of a systems biology approach for personalized medicine in refractory epilepsy.
Collapse
Affiliation(s)
- Giuseppina Daniela Naimo
- Institute for Biomedical Research and Innovation, National Research Council, Contrada Burga, Piano Lago, 87050 Mangone (CS) and Via Paolo Gaifami 18, 95126 Catania, Italy
| | - Maria Guarnaccia
- Institute for Biomedical Research and Innovation, National Research Council, Contrada Burga, Piano Lago, 87050 Mangone (CS) and Via Paolo Gaifami 18, 95126 Catania, Italy
| | - Teresa Sprovieri
- Institute for Biomedical Research and Innovation, National Research Council, Contrada Burga, Piano Lago, 87050 Mangone (CS) and Via Paolo Gaifami 18, 95126 Catania, Italy
| | - Carmine Ungaro
- Institute for Biomedical Research and Innovation, National Research Council, Contrada Burga, Piano Lago, 87050 Mangone (CS) and Via Paolo Gaifami 18, 95126 Catania, Italy
| | - Francesca Luisa Conforti
- Department of Pharmacy, Health and Nutritional Sciences, University of Calabria, Rende, 87036 Cosenza, Italy
| | - Sebastiano Andò
- Department of Pharmacy, Health and Nutritional Sciences, University of Calabria, Rende, 87036 Cosenza, Italy
- Centro Sanitario, University of Calabria, Via Pietro Bucci, 87036 Arcavacata di Rende (CS), Italy
| | - Sebastiano Cavallaro
- Institute for Biomedical Research and Innovation, National Research Council, Contrada Burga, Piano Lago, 87050 Mangone (CS) and Via Paolo Gaifami 18, 95126 Catania, Italy.
| |
Collapse
|
255
|
Hompoonsup S, Chambers D, Doherty P, Williams G. No transcriptional evidence for active Na v channels in two classes of cancer cell. Channels (Austin) 2019; 13:311-320. [PMID: 31329011 PMCID: PMC6682260 DOI: 10.1080/19336950.2019.1644858] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/26/2022] Open
Abstract
Voltage-gated sodium channel (Nav) expression in non-excitable cells has raised questions regarding their non-canonical roles. Interestingly, a growing body of evidence also points towards the prevalence of aberrant Nav expression in malignant tumors, potentially opening a new therapeutic window. In this study, the transcriptional consequences of channel inhibition were investigated in non-small cell lung carcinoma H460 and neuroblastoma SH-SYSY cell lines, that both express Nav1.7. Channel activity was blocked by the application of both selective, ProTx-II, and non-selective, tetrodotoxin, inhibitors. Global gene expression profiling did not point to any statistically significant inhibition-associated perturbation of the transcriptome. A small subset of genes that showed relatively consistent changes across multiple treatments were further assayed in the context of a multiplex bead expression array which failed to recapitulate the changes seen in the global array. We conclude that there is no robust transcriptional signature associated with the inhibition of two sodium channel expressing cancer cell lines and consequently sodium channel inhibition will not lend itself to therapeutic approaches such as transcription-based drug repurposing.
Collapse
Affiliation(s)
- Supanida Hompoonsup
- a Wolfson Centre for Age-Related Diseases, King's College London , London , UK.,b Learning Institute, King Mongkut's University of Technology Thonburi , Bangkok , Thailand
| | - David Chambers
- a Wolfson Centre for Age-Related Diseases, King's College London , London , UK
| | - Patrick Doherty
- a Wolfson Centre for Age-Related Diseases, King's College London , London , UK
| | - Gareth Williams
- a Wolfson Centre for Age-Related Diseases, King's College London , London , UK
| |
Collapse
|
256
|
Morales Duque H, Campos Dias S, Franco OL. Structural and Functional Analyses of Cone Snail Toxins. Mar Drugs 2019; 17:md17060370. [PMID: 31234371 PMCID: PMC6628382 DOI: 10.3390/md17060370] [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: 05/27/2019] [Revised: 06/16/2019] [Accepted: 06/17/2019] [Indexed: 12/12/2022] Open
Abstract
Cone snails are marine gastropod mollusks with one of the most powerful venoms in nature. The toxins, named conotoxins, must act quickly on the cone snails´ prey due to the fact that snails are extremely slow, reducing their hunting capability. Therefore, the characteristics of conotoxins have become the object of investigation, and as a result medicines have been developed or are in the trialing process. Conotoxins interact with transmembrane proteins, showing specificity and potency. They target ion channels and ionotropic receptors with greater regularity, and when interaction occurs, there is immediate physiological decompensation. In this review we aimed to evaluate the structural features of conotoxins and the relationship with their target types.
Collapse
Affiliation(s)
- Harry Morales Duque
- Centro de Análises Proteômicas e Bioquímicas, Programa de Pós-Graduação em Ciências Genômicas e Biotecnologia, Universidade Católica de Brasília, Brasília-DF 70.790-160, Brazil.
| | - Simoni Campos Dias
- Centro de Análises Proteômicas e Bioquímicas, Programa de Pós-Graduação em Ciências Genômicas e Biotecnologia, Universidade Católica de Brasília, Brasília-DF 70.790-160, Brazil.
| | - Octávio Luiz Franco
- Centro de Análises Proteômicas e Bioquímicas, Programa de Pós-Graduação em Ciências Genômicas e Biotecnologia, Universidade Católica de Brasília, Brasília-DF 70.790-160, Brazil.
- S-inova Biotech, Programa de Pós-Graduação em Biotecnologia, Universidade Católica Dom Bosco, Campo Grande-MS 79.117-900, Brazil.
| |
Collapse
|
257
|
Rothenberg ME, Tagen M, Chang JH, Boyce-Rustay J, Friesenhahn M, Hackos DH, Hains A, Sutherlin D, Ward M, Cho W. Safety, Tolerability, and Pharmacokinetics of GDC-0276, a Novel NaV1.7 Inhibitor, in a First-in-Human, Single- and Multiple-Dose Study in Healthy Volunteers. Clin Drug Investig 2019; 39:873-887. [DOI: 10.1007/s40261-019-00807-3] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/26/2022]
|
258
|
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: 36] [Impact Index Per Article: 6.0] [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
|
259
|
Turlik A, Chen Y, Scruse AC, Newhouse TR. Convergent Total Synthesis of Principinol D, a Rearranged Kaurane Diterpenoid. J Am Chem Soc 2019; 141:8088-8092. [PMID: 31042866 PMCID: PMC7192013 DOI: 10.1021/jacs.9b03751] [Citation(s) in RCA: 32] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
The total synthesis of principinol D, a rearranged kaurane diterpenoid, is reported. This grayanane natural product is constructed via a convergent fragment coupling approach, wherein the central seven-membered ring is synthesized at a late stage. The bicyclo[3.2.1]octane fragment is accessed by a Ni-catalyzed α-vinylation reaction. Strategic reductions include a diastereoselective SmI2-mediated ketone reduction with PhSH and a new protocol for selective ester reduction in the presence of ketones. The convergent strategy reported herein may be an entry point to the larger class of kaurane diterpenoids.
Collapse
Affiliation(s)
- Aneta Turlik
- Department of Chemistry, Yale University, 225 Prospect Street, New Haven, Connecticut 06520-8107, United States
| | - Yifeng Chen
- Department of Chemistry, Yale University, 225 Prospect Street, New Haven, Connecticut 06520-8107, United States
| | - Anthony C. Scruse
- Department of Chemistry, Yale University, 225 Prospect Street, New Haven, Connecticut 06520-8107, United States
| | - Timothy R. Newhouse
- Department of Chemistry, Yale University, 225 Prospect Street, New Haven, Connecticut 06520-8107, United States
| |
Collapse
|
260
|
Trier AM, Mack MR, Kim BS. The Neuroimmune Axis in Skin Sensation, Inflammation, and Immunity. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2019; 202:2829-2835. [PMID: 31061146 PMCID: PMC6563610 DOI: 10.4049/jimmunol.1801473] [Citation(s) in RCA: 43] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/02/2018] [Accepted: 02/21/2019] [Indexed: 12/21/2022]
Abstract
Although connections between the immune and nervous systems have long been recognized, the precise mechanisms that underlie this relationship are just starting to be elucidated. Advances in sensory biology have unveiled novel mechanisms by which inflammatory cytokines promote itch and pain sensations to coordinate host-protective behavioral responses. Conversely, new evidence has emphasized the importance of immune cell regulation by sensory neurons. By focusing on itch biology and how it has been informed by the more established field of pain research, we highlight recent interdisciplinary studies that demonstrate how novel neuroimmune interactions underlie a diversity of sensory, inflammatory, and infectious diseases.
Collapse
Affiliation(s)
- Anna M Trier
- Center for the Study of Itch, Washington University School of Medicine, St. Louis, MO 63110
- Division of Dermatology, Department of Medicine, Washington University School of Medicine, St. Louis, MO 63110
| | - Madison R Mack
- Center for the Study of Itch, Washington University School of Medicine, St. Louis, MO 63110
- Division of Dermatology, Department of Medicine, Washington University School of Medicine, St. Louis, MO 63110
| | - Brian S Kim
- Center for the Study of Itch, Washington University School of Medicine, St. Louis, MO 63110;
- Division of Dermatology, Department of Medicine, Washington University School of Medicine, St. Louis, MO 63110
- Department of Anesthesiology, Washington University School of Medicine, St. Louis, MO 63110; and
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO 63110
| |
Collapse
|
261
|
Wulff H, Christophersen P, Colussi P, Chandy KG, Yarov-Yarovoy V. Antibodies and venom peptides: new modalities for ion channels. Nat Rev Drug Discov 2019; 18:339-357. [PMID: 30728472 PMCID: PMC6499689 DOI: 10.1038/s41573-019-0013-8] [Citation(s) in RCA: 108] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
Abstract
Ion channels play fundamental roles in both excitable and non-excitable tissues and therefore constitute attractive drug targets for myriad neurological, cardiovascular and metabolic diseases as well as for cancer and immunomodulation. However, achieving selectivity for specific ion channel subtypes with small-molecule drugs has been challenging, and there currently is a growing trend to target ion channels with biologics. One approach is to improve the pharmacokinetics of existing or novel venom-derived peptides. In parallel, after initial studies with polyclonal antibodies demonstrated the technical feasibility of inhibiting channel function with antibodies, multiple preclinical programmes are now using the full spectrum of available technologies to generate conventional monoclonal and engineered antibodies or nanobodies against extracellular loops of ion channels. After a summary of the current state of ion channel drug discovery, this Review discusses recent developments using the purinergic receptor channel P2X purinoceptor 7 (P2X7), the voltage-gated potassium channel KV1.3 and the voltage-gated sodium channel NaV1.7 as examples of targeting ion channels with biologics.
Collapse
Affiliation(s)
- Heike Wulff
- Department of Pharmacology, University of California Davis, Davis, CA, USA.
| | | | | | - K George Chandy
- Molecular Physiology Laboratory, Infection and Immunity Theme, Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore, Singapore
| | - Vladimir Yarov-Yarovoy
- Department of Physiology & Membrane Biology, University of California Davis, Davis, CA, USA
| |
Collapse
|
262
|
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
|
263
|
Gonçalves TC, Benoit E, Kurz M, Lucarain L, Fouconnier S, Combemale S, Jaquillard L, Schombert B, Chambard JM, Boukaiba R, Hessler G, Bohme A, Bialy L, Hourcade S, Béroud R, De Waard M, Servent D, Partiseti M. From identification to functional characterization of cyriotoxin-1a, an antinociceptive toxin from the spider Cyriopagopus schioedtei. Br J Pharmacol 2019; 176:1298-1314. [PMID: 30784059 DOI: 10.1111/bph.14628] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2018] [Revised: 01/24/2019] [Accepted: 01/31/2019] [Indexed: 12/18/2022] Open
Abstract
BACKGROUND AND PURPOSE The NaV 1.7 channel is highly expressed in dorsal root ganglia of the sensory nervous system and plays a central role in the pain signalling process. We investigated a library prepared from original venoms of 117 different animals to identify new selective inhibitors of this target. EXPERIMENTAL APPROACH We used high throughput screening of a large venom collection using automated patch-clamp experiments on human voltage-gated sodium channel subtypes and then in vitro and in vivo electrophysiological experiments to characterize the active peptides that have been purified, sequenced, and chemically synthesized. Analgesic effects were evaluated in vivo in mice models. KEY RESULTS We identified cyriotoxin-1a (CyrTx-1a), a novel peptide isolated from Cyriopagopus schioedtei spider venom, as a candidate for further characterization. This 33 amino acids toxin belongs to the inhibitor cystine knot structural family and inhibits hNaV 1.1-1.3 and 1.6-1.7 channels in the low nanomolar range, compared to the micromolar range for hNaV 1.4-1.5 and 1.8 channels. CyrTx-1a was 920 times more efficient at inhibiting tetrodotoxin (TTX)-sensitive than TTX-resistant sodium currents recorded from adult mouse dorsal root ganglia neurons and in vivo electrophysiological experiments showed that CyrTx-1a was approximately 170 times less efficient than huwentoxin-IV at altering mouse skeletal neuromuscular excitability properties. CyrTx-1a exhibited an analgesic effect in mice by increasing reaction time in the hot-plate assay. CONCLUSIONS AND IMPLICATIONS The pharmacological profile of CyrTx-1a paves the way for further molecular engineering aimed to optimize the potential antinociceptive properties of this peptide.
Collapse
Affiliation(s)
- Tânia C Gonçalves
- Integrated Drug Discovery-High Content Biology, Sanofi R&D, Vitry-sur-Seine, France.,Service d'Ingénierie Moléculaire des Protéines (SIMOPRO), CEA, Université Paris-Saclay, Gif-sur-Yvette, France
| | - Evelyne Benoit
- Service d'Ingénierie Moléculaire des Protéines (SIMOPRO), CEA, Université Paris-Saclay, Gif-sur-Yvette, France.,Institut des Neurosciences Paris-Saclay (Neuro-PSI), UMR CNRS/Université Paris-Sud 9197, Université Paris-Saclay, Gif-sur-Yvette, France
| | - Michael Kurz
- Integrated Drug Discovery-Synthetic Molecular Design, Sanofi R&D, Frankfurt, Germany
| | - Laetitia Lucarain
- Integrated Drug Discovery-High Content Biology, Sanofi R&D, Vitry-sur-Seine, France
| | - Sophie Fouconnier
- Integrated Drug Discovery-High Content Biology, Sanofi R&D, Vitry-sur-Seine, France
| | | | | | - Brigitte Schombert
- Integrated Drug Discovery-High Content Biology, Sanofi R&D, Vitry-sur-Seine, France
| | - Jean-Marie Chambard
- Integrated Drug Discovery-High Content Biology, Sanofi R&D, Vitry-sur-Seine, France
| | - Rachid Boukaiba
- Integrated Drug Discovery-High Content Biology, Sanofi R&D, Vitry-sur-Seine, France
| | - Gerhard Hessler
- Integrated Drug Discovery-Synthetic Molecular Design, Sanofi R&D, Frankfurt, Germany
| | - Andrees Bohme
- Integrated Drug Discovery-High Content Biology, Sanofi R&D, Vitry-sur-Seine, France
| | - Laurent Bialy
- Integrated Drug Discovery-Synthetic Molecular Design, Sanofi R&D, Frankfurt, Germany
| | - Stéphane Hourcade
- Neuroscience Therapeutic Area, Neurodegeneration Research, Sanofi R&D, Chilly-Mazarin, France
| | | | - Michel De Waard
- Smartox Biotechnology, Saint-Egrève, France.,Institut du Thorax, Inserm UMR 1087/CNRS UMR 6291, LabEx "Ion Channels, Science and Therapeutics", Nantes, France
| | - Denis Servent
- Service d'Ingénierie Moléculaire des Protéines (SIMOPRO), CEA, Université Paris-Saclay, Gif-sur-Yvette, France
| | - Michel Partiseti
- Integrated Drug Discovery-High Content Biology, Sanofi R&D, Vitry-sur-Seine, France
| |
Collapse
|
264
|
Nastou KC, Batskinis MA, Litou ZI, Hamodrakas SJ, Iconomidou VA. Analysis of Single-Nucleotide Polymorphisms in Human Voltage-Gated Ion Channels. J Proteome Res 2019; 18:2310-2320. [DOI: 10.1021/acs.jproteome.9b00121] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Affiliation(s)
- Katerina C. Nastou
- Section of Cell Biology and Biophysics, Department of Biology, School of Sciences, National and Kapodistrian University of Athens, Panepistimiopolis, Athens 15701, Greece
| | - Michail A. Batskinis
- Section of Cell Biology and Biophysics, Department of Biology, School of Sciences, National and Kapodistrian University of Athens, Panepistimiopolis, Athens 15701, Greece
| | - Zoi I. Litou
- Section of Cell Biology and Biophysics, Department of Biology, School of Sciences, National and Kapodistrian University of Athens, Panepistimiopolis, Athens 15701, Greece
| | - Stavros J. Hamodrakas
- Section of Cell Biology and Biophysics, Department of Biology, School of Sciences, National and Kapodistrian University of Athens, Panepistimiopolis, Athens 15701, Greece
| | - Vassiliki A. Iconomidou
- Section of Cell Biology and Biophysics, Department of Biology, School of Sciences, National and Kapodistrian University of Athens, Panepistimiopolis, Athens 15701, Greece
| |
Collapse
|
265
|
Sun JF, Xu YJ, Kong XH, Su Y, Wang ZY. Fenamates inhibit human sodium channel Nav1.7 and Nav1.8. Neurosci Lett 2019; 696:67-73. [DOI: 10.1016/j.neulet.2018.12.008] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2018] [Revised: 11/06/2018] [Accepted: 12/04/2018] [Indexed: 11/27/2022]
|
266
|
Maatuf Y, Geron M, Priel A. The Role of Toxins in the Pursuit for Novel Analgesics. Toxins (Basel) 2019; 11:toxins11020131. [PMID: 30813430 PMCID: PMC6409898 DOI: 10.3390/toxins11020131] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2019] [Revised: 02/17/2019] [Accepted: 02/20/2019] [Indexed: 12/19/2022] Open
Abstract
Chronic pain is a major medical issue which reduces the quality of life of millions and inflicts a significant burden on health authorities worldwide. Currently, management of chronic pain includes first-line pharmacological therapies that are inadequately effective, as in just a portion of patients pain relief is obtained. Furthermore, most analgesics in use produce severe or intolerable adverse effects that impose dose restrictions and reduce compliance. As the majority of analgesic agents act on the central nervous system (CNS), it is possible that blocking pain at its source by targeting nociceptors would prove more efficient with minimal CNS-related side effects. The development of such analgesics requires the identification of appropriate molecular targets and thorough understanding of their structural and functional features. To this end, plant and animal toxins can be employed as they affect ion channels with high potency and selectivity. Moreover, elucidation of the toxin-bound ion channel structure could generate pharmacophores for rational drug design while favorable safety and analgesic profiles could highlight toxins as leads or even as valuable therapeutic compounds themselves. Here, we discuss the use of plant and animal toxins in the characterization of peripherally expressed ion channels which are implicated in pain.
Collapse
Affiliation(s)
- Yossi Maatuf
- The Institute for Drug Research (IDR), School of Pharmacy, Faculty of Medicine, The Hebrew University of Jerusalem, Jerusalem 9112001, Israel.
| | - Matan Geron
- The Institute for Drug Research (IDR), School of Pharmacy, Faculty of Medicine, The Hebrew University of Jerusalem, Jerusalem 9112001, Israel.
| | - Avi Priel
- The Institute for Drug Research (IDR), School of Pharmacy, Faculty of Medicine, The Hebrew University of Jerusalem, Jerusalem 9112001, Israel.
| |
Collapse
|
267
|
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
|
268
|
Lawrence N, Wu B, Ligutti J, Cheneval O, Agwa AJ, Benfield AH, Biswas K, Craik DJ, Miranda LP, Henriques ST, Schroeder CI. Peptide-Membrane Interactions Affect the Inhibitory Potency and Selectivity of Spider Toxins ProTx-II and GpTx-1. ACS Chem Biol 2019; 14:118-130. [PMID: 30507158 DOI: 10.1021/acschembio.8b00989] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Gating modifier toxins (GMTs) from spider venom can inhibit voltage gated sodium channels (NaVs) involved in pain signal transmission, including the NaV1.7 subtype. GMTs have a conserved amphipathic structure that allow them to interact with membranes and also with charged residues in regions of NaV that are exposed at the cell surface. ProTx-II and GpTx-1 are GMTs able to inhibit NaV1.7 with high potency, but they differ in their ability to bind to membranes and in their selectivity over other NaV subtypes. To explore these differences and gain detailed information on their membrane-binding ability and how this relates to potency and selectivity, we examined previously described NaV1.7 potent/selective GpTx-1 analogues and new ProTx-II analogues designed to reduce membrane binding and improve selectivity for NaV1.7. Our studies reveal that the number and type of hydrophobic residues as well as how they are presented at the surface determine the affinity of ProTx-II and GpTx-1 for membranes and that altering these residues can have dramatic effects on NaV inhibitory activity. We demonstrate that strong peptide-membrane interactions are not essential for inhibiting NaV1.7 and propose that hydrophobic interactions instead play an important role in positioning the GMT at the membrane surface proximal to exposed NaV residues, thereby affecting peptide-channel interactions. Our detailed structure-activity relationship study highlights the challenges of designing GMT-based molecules that simultaneously achieve high potency and selectivity for NaV1.7, as single mutations can induce local changes in GMT structure that can have a major impact on NaV-inhibitory activity.
Collapse
Affiliation(s)
- Nicole Lawrence
- Institute for Molecular Bioscience, The University of Queensland, Brisbane, Queensland 4072, Australia
| | | | - Joseph Ligutti
- Institute for Molecular Bioscience, The University of Queensland, Brisbane, Queensland 4072, Australia
| | - Olivier Cheneval
- Institute for Molecular Bioscience, The University of Queensland, Brisbane, Queensland 4072, Australia
| | - Akello Joanna Agwa
- Institute for Molecular Bioscience, The University of Queensland, Brisbane, Queensland 4072, Australia
| | - Aurélie H. Benfield
- Institute for Molecular Bioscience, The University of Queensland, Brisbane, Queensland 4072, Australia
| | | | | | | | - Sónia Troeira Henriques
- Institute for Molecular Bioscience, The University of Queensland, Brisbane, Queensland 4072, Australia
| | - Christina I. Schroeder
- Institute for Molecular Bioscience, The University of Queensland, Brisbane, Queensland 4072, Australia
| |
Collapse
|
269
|
Luo G, Chen L, Easton A, Newton A, Bourin C, Shields E, Mosure K, Soars MG, Knox RJ, Matchett M, Pieschl RL, Post-Munson DJ, Wang S, Herrington J, Graef J, Newberry K, Sivarao DV, Senapati A, Bristow LJ, Meanwell NA, Thompson LA, Dzierba C. Discovery of Indole- and Indazole-acylsulfonamides as Potent and Selective Na V1.7 Inhibitors for the Treatment of Pain. J Med Chem 2019; 62:831-856. [PMID: 30576602 DOI: 10.1021/acs.jmedchem.8b01550] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
3-Aryl-indole and 3-aryl-indazole derivatives were identified as potent and selective Nav1.7 inhibitors. Compound 29 was shown to be efficacious in the mouse formalin assay and also reduced complete Freund's adjuvant (CFA)-induced thermal hyperalgesia and chronic constriction injury (CCI) induced cold allodynia and models of inflammatory and neuropathic pain, respectively, following intraperitoneal (IP) doses of 30 mg/kg. The observed efficacy could be correlated with the mouse dorsal root ganglion exposure and NaV1.7 potency associated with 29.
Collapse
Affiliation(s)
- Guanglin Luo
- Bristol-Myers Squibb Research and Development , PO Box 4000, Princeton , New Jersey 08543-4000 , United States
| | - Ling Chen
- Bristol-Myers Squibb Research and Development , PO Box 4000, Princeton , New Jersey 08543-4000 , United States
| | - Amy Easton
- Bristol-Myers Squibb Research and Development , PO Box 4000, Princeton , New Jersey 08543-4000 , United States
| | - Amy Newton
- Bristol-Myers Squibb Research and Development , PO Box 4000, Princeton , New Jersey 08543-4000 , United States
| | - Clotilde Bourin
- Bristol-Myers Squibb Research and Development , PO Box 4000, Princeton , New Jersey 08543-4000 , United States
| | - Eric Shields
- Bristol-Myers Squibb Research and Development , PO Box 4000, Princeton , New Jersey 08543-4000 , United States
| | - Kathy Mosure
- Bristol-Myers Squibb Research and Development , PO Box 4000, Princeton , New Jersey 08543-4000 , United States
| | - Matthew G Soars
- Bristol-Myers Squibb Research and Development , PO Box 4000, Princeton , New Jersey 08543-4000 , United States
| | - Ronald J Knox
- Bristol-Myers Squibb Research and Development , PO Box 4000, Princeton , New Jersey 08543-4000 , United States
| | - Michele Matchett
- Bristol-Myers Squibb Research and Development , PO Box 4000, Princeton , New Jersey 08543-4000 , United States
| | - Rick L Pieschl
- Bristol-Myers Squibb Research and Development , PO Box 4000, Princeton , New Jersey 08543-4000 , United States
| | - Debra J Post-Munson
- Bristol-Myers Squibb Research and Development , PO Box 4000, Princeton , New Jersey 08543-4000 , United States
| | - Shuya Wang
- Bristol-Myers Squibb Research and Development , PO Box 4000, Princeton , New Jersey 08543-4000 , United States
| | - James Herrington
- Bristol-Myers Squibb Research and Development , PO Box 4000, Princeton , New Jersey 08543-4000 , United States
| | - John Graef
- Bristol-Myers Squibb Research and Development , PO Box 4000, Princeton , New Jersey 08543-4000 , United States
| | - Kimberly Newberry
- Bristol-Myers Squibb Research and Development , PO Box 4000, Princeton , New Jersey 08543-4000 , United States
| | - Digavalli V Sivarao
- Bristol-Myers Squibb Research and Development , PO Box 4000, Princeton , New Jersey 08543-4000 , United States
| | - Arun Senapati
- Bristol-Myers Squibb Research and Development , PO Box 4000, Princeton , New Jersey 08543-4000 , United States
| | - Linda J Bristow
- Bristol-Myers Squibb Research and Development , PO Box 4000, Princeton , New Jersey 08543-4000 , United States
| | - Nicholas A Meanwell
- Bristol-Myers Squibb Research and Development , PO Box 4000, Princeton , New Jersey 08543-4000 , United States
| | - Lorin A Thompson
- Bristol-Myers Squibb Research and Development , PO Box 4000, Princeton , New Jersey 08543-4000 , United States
| | - Carolyn Dzierba
- Bristol-Myers Squibb Research and Development , PO Box 4000, Princeton , New Jersey 08543-4000 , United States
| |
Collapse
|
270
|
Domokos D, Ducza E, Gáspár R. RhoA and Rho-kinase inhibitors modulate cervical resistance: The possible role of RhoA/Rho-kinase signalling pathway in cervical ripening and contractility. Eur J Pharmacol 2019; 843:27-33. [DOI: 10.1016/j.ejphar.2018.11.017] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2018] [Revised: 11/11/2018] [Accepted: 11/12/2018] [Indexed: 12/14/2022]
|
271
|
Liu Z, Wadsworth P, Singh AK, Chen H, Wang P, Folorunso O, Scaduto P, Ali SR, Laezza F, Zhou J. Identification of peptidomimetics as novel chemical probes modulating fibroblast growth factor 14 (FGF14) and voltage-gated sodium channel 1.6 (Nav1.6) protein-protein interactions. Bioorg Med Chem Lett 2018; 29:413-419. [PMID: 30587448 DOI: 10.1016/j.bmcl.2018.12.031] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2018] [Revised: 12/11/2018] [Accepted: 12/13/2018] [Indexed: 11/26/2022]
Abstract
The voltage-gated sodium (Nav) channel is the molecular determinant of action potential in neurons. Protein-protein interactions (PPI) between the intracellular Nav1.6 C-tail and its regulatory protein fibroblast growth factor 14 (FGF14) provide an ideal and largely untapped opportunity for development of neurochemical probes. Based on a previously identified peptide FLPK, mapped to the FGF14:FGF14 PPI interface, we have designed and synthesized a series of peptidomimetics with the intent of increasing clogP values and improving cell permeability relative to the parental lead peptide. In-cell screening using the split-luciferase complementation (LCA) assay identified ZL0177 (13) as the most potent inhibitor of the FGF14:Nav1.6 channel complex assembly with an apparent IC50 of 11 μM. Whole-cell patch-clamp recordings demonstrated that ZL0177 significantly reduced Nav1.6-mediated transient current density and induced a depolarizing shift of the channel voltage-dependence of activation. Docking studies revealed strong interactions between ZL0177 and Nav1.6, mediated by hydrogen bonds, cation-π interactions and hydrophobic contacts. All together these results suggest that ZL0177 retains some key features of FGF14-dependent modulation of Nav1.6 currents. Overall, ZL0177 provides a chemical scaffold for developing Nav channel modulators as pharmacological probes with therapeutic potential of interest for a broad range of CNS and PNS disorders.
Collapse
Affiliation(s)
- Zhiqing Liu
- Department of Pharmacology and Toxicology, University of Texas Medical Branch, 301 University Blvd, Galveston, Galveston, TX 77555, United States
| | - Paul Wadsworth
- Department of Pharmacology and Toxicology, University of Texas Medical Branch, 301 University Blvd, Galveston, Galveston, TX 77555, United States
| | - Aditya K Singh
- Department of Pharmacology and Toxicology, University of Texas Medical Branch, 301 University Blvd, Galveston, Galveston, TX 77555, United States
| | - Haiying Chen
- Department of Pharmacology and Toxicology, University of Texas Medical Branch, 301 University Blvd, Galveston, Galveston, TX 77555, United States
| | - Pingyuan Wang
- Department of Pharmacology and Toxicology, University of Texas Medical Branch, 301 University Blvd, Galveston, Galveston, TX 77555, United States
| | - Oluwarotimi Folorunso
- Department of Pharmacology and Toxicology, University of Texas Medical Branch, 301 University Blvd, Galveston, Galveston, TX 77555, United States
| | - Pietro Scaduto
- Department of Pharmacology and Toxicology, University of Texas Medical Branch, 301 University Blvd, Galveston, Galveston, TX 77555, United States
| | - Syed R Ali
- Department of Pharmacology and Toxicology, University of Texas Medical Branch, 301 University Blvd, Galveston, Galveston, TX 77555, United States
| | - Fernanda Laezza
- Department of Pharmacology and Toxicology, University of Texas Medical Branch, 301 University Blvd, Galveston, Galveston, TX 77555, United States.
| | - Jia Zhou
- Department of Pharmacology and Toxicology, University of Texas Medical Branch, 301 University Blvd, Galveston, Galveston, TX 77555, United States.
| |
Collapse
|
272
|
Wu YJ, Venables B, Guernon J, Chen J, Sit SY, Rajamani R, Knox RJ, Matchett M, Pieschl RL, Herrington J, Bristow LJ, Meanwell NA, Thompson LA, Dzierba C. Discovery of new indole-based acylsulfonamide Na v1.7 inhibitors. Bioorg Med Chem Lett 2018; 29:659-663. [PMID: 30638874 DOI: 10.1016/j.bmcl.2018.12.013] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2018] [Revised: 12/03/2018] [Accepted: 12/05/2018] [Indexed: 01/05/2023]
Abstract
Screening of 100 acylsulfonamides from the Bristol-Myers Squibb compound collection identified the C3-cyclohexyl indole 6 as a potent Nav1.7 inhibitor. Replacement of the C2 furanyl ring of 6 with a heteroaryl moiety or truncation of this group led to the identification of 4 analogs with hNav1.7 IC50 values under 50 nM. Fluorine substitution of the truncated compound 12 led to 34 with improved potency and isoform selectivity. The inverted indole 36 also maintained good activity. Both 34 and 36 exhibited favorable CYP inhibition profiles, good membrane permeability and a low efflux ratio and, therefore, represent new leads in the search for potent and selective Nav1.7 inhibitors to treat pain.
Collapse
Affiliation(s)
- Yong-Jin Wu
- Bristol-Myers Squibb Research and Development, 5 Research Parkway, Wallingford, CT 06492-7660, USA.
| | - Brian Venables
- Bristol-Myers Squibb Research and Development, 5 Research Parkway, Wallingford, CT 06492-7660, USA
| | - Jason Guernon
- Bristol-Myers Squibb Research and Development, 5 Research Parkway, Wallingford, CT 06492-7660, USA
| | - Jie Chen
- Bristol-Myers Squibb Research and Development, 5 Research Parkway, Wallingford, CT 06492-7660, USA
| | - Sing-Yuen Sit
- Bristol-Myers Squibb Research and Development, 5 Research Parkway, Wallingford, CT 06492-7660, USA
| | - Ramkumar Rajamani
- Bristol-Myers Squibb Research and Development, 5 Research Parkway, Wallingford, CT 06492-7660, USA
| | - Ronald J Knox
- Bristol-Myers Squibb Research and Development, 5 Research Parkway, Wallingford, CT 06492-7660, USA
| | - Michele Matchett
- Bristol-Myers Squibb Research and Development, 5 Research Parkway, Wallingford, CT 06492-7660, USA
| | - Rick L Pieschl
- Bristol-Myers Squibb Research and Development, 5 Research Parkway, Wallingford, CT 06492-7660, USA
| | - James Herrington
- Bristol-Myers Squibb Research and Development, 5 Research Parkway, Wallingford, CT 06492-7660, USA
| | - Linda J Bristow
- Bristol-Myers Squibb Research and Development, 5 Research Parkway, Wallingford, CT 06492-7660, USA
| | - Nicholas A Meanwell
- Bristol-Myers Squibb Research and Development, 5 Research Parkway, Wallingford, CT 06492-7660, USA
| | - Lorin A Thompson
- Bristol-Myers Squibb Research and Development, 5 Research Parkway, Wallingford, CT 06492-7660, USA
| | - Carolyn Dzierba
- Bristol-Myers Squibb Research and Development, 5 Research Parkway, Wallingford, CT 06492-7660, USA
| |
Collapse
|
273
|
Pérez de Vega MJ, Ferrer-Montiel A, González-Muñiz R. Recent progress in non-opioid analgesic peptides. Arch Biochem Biophys 2018; 660:36-52. [DOI: 10.1016/j.abb.2018.10.011] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2018] [Revised: 10/15/2018] [Accepted: 10/16/2018] [Indexed: 02/08/2023]
|
274
|
Sun S, Jia Q, Zenova AY, Wilson MS, Chowdhury S, Focken T, Li J, Decker S, Grimwood ME, Andrez JC, Hemeon I, Sheng T, Chen CA, White A, Hackos DH, Deng L, Bankar G, Khakh K, Chang E, Kwan R, Lin S, Nelkenbrecher K, Sellers BD, DiPasquale AG, Chang J, Pang J, Sojo L, Lindgren A, Waldbrook M, Xie Z, Young C, Johnson JP, Robinette CL, Cohen CJ, Safina BS, Sutherlin DP, Ortwine DF, Dehnhardt CM. Identification of Selective Acyl Sulfonamide–Cycloalkylether Inhibitors of the Voltage-Gated Sodium Channel (NaV) 1.7 with Potent Analgesic Activity. J Med Chem 2018; 62:908-927. [DOI: 10.1021/acs.jmedchem.8b01621] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Affiliation(s)
- Shaoyi Sun
- Xenon Pharmaceuticals Inc., 200-3650 Gilmore Way, Burnaby, British Columbia V5G 4W8, Canada
| | - Qi Jia
- Xenon Pharmaceuticals Inc., 200-3650 Gilmore Way, Burnaby, British Columbia V5G 4W8, Canada
| | - Alla Y. Zenova
- Xenon Pharmaceuticals Inc., 200-3650 Gilmore Way, Burnaby, British Columbia V5G 4W8, Canada
| | - Michael S. Wilson
- Xenon Pharmaceuticals Inc., 200-3650 Gilmore Way, Burnaby, British Columbia V5G 4W8, Canada
| | - Sultan Chowdhury
- Xenon Pharmaceuticals Inc., 200-3650 Gilmore Way, Burnaby, British Columbia V5G 4W8, Canada
| | - Thilo Focken
- Xenon Pharmaceuticals Inc., 200-3650 Gilmore Way, Burnaby, British Columbia V5G 4W8, Canada
| | - Jun Li
- Genentech Inc., 1 DNA Way, South San Francisco, California 94080-4990, United States
| | - Shannon Decker
- Xenon Pharmaceuticals Inc., 200-3650 Gilmore Way, Burnaby, British Columbia V5G 4W8, Canada
| | - Michael E. Grimwood
- Xenon Pharmaceuticals Inc., 200-3650 Gilmore Way, Burnaby, British Columbia V5G 4W8, Canada
| | - Jean-Christophe Andrez
- Xenon Pharmaceuticals Inc., 200-3650 Gilmore Way, Burnaby, British Columbia V5G 4W8, Canada
| | - Ivan Hemeon
- Xenon Pharmaceuticals Inc., 200-3650 Gilmore Way, Burnaby, British Columbia V5G 4W8, Canada
| | - Tao Sheng
- Xenon Pharmaceuticals Inc., 200-3650 Gilmore Way, Burnaby, British Columbia V5G 4W8, Canada
| | - Chien-An Chen
- ChemPartner, Building No. 5, 998 Halei Road, Zhangjiang Hi-Tech Park, Pudong New Area, Shanghai 201203, P. R. China
| | - Andy White
- ChemPartner, Building No. 5, 998 Halei Road, Zhangjiang Hi-Tech Park, Pudong New Area, Shanghai 201203, P. R. China
| | - David H. Hackos
- Genentech Inc., 1 DNA Way, South San Francisco, California 94080-4990, United States
| | - Lunbin Deng
- Genentech Inc., 1 DNA Way, South San Francisco, California 94080-4990, United States
| | - Girish Bankar
- Xenon Pharmaceuticals Inc., 200-3650 Gilmore Way, Burnaby, British Columbia V5G 4W8, Canada
| | - Kuldip Khakh
- Xenon Pharmaceuticals Inc., 200-3650 Gilmore Way, Burnaby, British Columbia V5G 4W8, Canada
| | - Elaine Chang
- Xenon Pharmaceuticals Inc., 200-3650 Gilmore Way, Burnaby, British Columbia V5G 4W8, Canada
| | - Rainbow Kwan
- Xenon Pharmaceuticals Inc., 200-3650 Gilmore Way, Burnaby, British Columbia V5G 4W8, Canada
| | - Sophia Lin
- Xenon Pharmaceuticals Inc., 200-3650 Gilmore Way, Burnaby, British Columbia V5G 4W8, Canada
| | - Karen Nelkenbrecher
- Xenon Pharmaceuticals Inc., 200-3650 Gilmore Way, Burnaby, British Columbia V5G 4W8, Canada
| | - Benjamin D. Sellers
- Genentech Inc., 1 DNA Way, South San Francisco, California 94080-4990, United States
| | - Antonio G. DiPasquale
- Genentech Inc., 1 DNA Way, South San Francisco, California 94080-4990, United States
| | - Jae Chang
- Genentech Inc., 1 DNA Way, South San Francisco, California 94080-4990, United States
| | - Jodie Pang
- Genentech Inc., 1 DNA Way, South San Francisco, California 94080-4990, United States
| | - Luis Sojo
- Xenon Pharmaceuticals Inc., 200-3650 Gilmore Way, Burnaby, British Columbia V5G 4W8, Canada
| | - Andrea Lindgren
- Xenon Pharmaceuticals Inc., 200-3650 Gilmore Way, Burnaby, British Columbia V5G 4W8, Canada
| | - Matthew Waldbrook
- Xenon Pharmaceuticals Inc., 200-3650 Gilmore Way, Burnaby, British Columbia V5G 4W8, Canada
| | - Zhiwei Xie
- Xenon Pharmaceuticals Inc., 200-3650 Gilmore Way, Burnaby, British Columbia V5G 4W8, Canada
| | - Clint Young
- Xenon Pharmaceuticals Inc., 200-3650 Gilmore Way, Burnaby, British Columbia V5G 4W8, Canada
| | - James P. Johnson
- Xenon Pharmaceuticals Inc., 200-3650 Gilmore Way, Burnaby, British Columbia V5G 4W8, Canada
| | - C. Lee Robinette
- Xenon Pharmaceuticals Inc., 200-3650 Gilmore Way, Burnaby, British Columbia V5G 4W8, Canada
| | - Charles J. Cohen
- Xenon Pharmaceuticals Inc., 200-3650 Gilmore Way, Burnaby, British Columbia V5G 4W8, Canada
| | - Brian S. Safina
- Genentech Inc., 1 DNA Way, South San Francisco, California 94080-4990, United States
| | - Daniel P. Sutherlin
- Genentech Inc., 1 DNA Way, South San Francisco, California 94080-4990, United States
| | - Daniel F. Ortwine
- Genentech Inc., 1 DNA Way, South San Francisco, California 94080-4990, United States
| | - Christoph M. Dehnhardt
- Xenon Pharmaceuticals Inc., 200-3650 Gilmore Way, Burnaby, British Columbia V5G 4W8, Canada
| |
Collapse
|
275
|
Niitsu A, Egawa A, Ikeda K, Tachibana K, Fujiwara T. Veratridine binding to a transmembrane helix of sodium channel Na v1.4 determined by solid-state NMR. Bioorg Med Chem 2018; 26:5644-5653. [PMID: 30389410 DOI: 10.1016/j.bmc.2018.10.012] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2018] [Revised: 10/13/2018] [Accepted: 10/16/2018] [Indexed: 11/30/2022]
Abstract
The multi-step ligand action to a target protein is an important aspect when understanding mechanisms of ligand binding and discovering new drugs. However, structurally capturing such complex mechanisms is challenging. This is particularly true for interactions between large membrane proteins and small molecules. One such large membrane of interest is Nav1.4, a eukaryotic voltage-gated sodium channel. Domain 4 segment 6 (D4S6) of Nav1.4 is a transmembrane α-helical segment playing a key role in channel gating regulation, and is targeted by a neurotoxin, veratridine (VTD). VTD has been suggested to exhibit a two-step action to activate Nav1.4. Here, we determine the NMR structure of a selectively 13C-labeled peptide corresponding to D4S6 and its VTD binding site in lipid bilayers determined by using magic-angle spinning solid-state NMR. By 13C NMR, we obtain NMR structural constraints as 13C chemical shifts and the 1H-2H dipolar couplings between the peptide and deuterated lipids. The peptide backbone structure and its location with respect to the membrane are determined under the obtained NMR structural constraints aided by replica exchange molecular dynamics simulations with an implicit membrane/solvent system. Further, by measuring the 1H-2H dipolar couplings to monitor the peptide-lipid interaction, we identify a VTD binding site on D4S6. When superimposed to a crystal structure of a bacterial sodium channel NavRh, the determined binding site is the only surface exposed to the protein exterior and localizes beside the second-step binding site reported in the past. Based on these results, we propose that VTD initially binds to these newly-determined residues on D4S6 from the membrane hydrophobic domain, which induces the first-step channel opening followed by the second-step blocking of channel inactivation of Nav1.4. Our findings provide new detailed insights of the VTD action mechanism, which could be useful in designing new drugs targeting D4S6.
Collapse
Affiliation(s)
- Ai Niitsu
- RIKEN, 2-1 Hirosawa, Wako, Saitama 351-0198, Japan
| | - Ayako Egawa
- Institute for Protein Research, Osaka University, 3-2 Yamadaoka, Suita, Osaka 565-0871, Japan
| | - Keisuke Ikeda
- Institute for Protein Research, Osaka University, 3-2 Yamadaoka, Suita, Osaka 565-0871, Japan; Graduate School of Medicine and Pharmaceutical Sciences, University of Toyama, 2630 Sugitani, Toyama 930-0194, Japan
| | - Kazuo Tachibana
- School of Science, Department of Chemistry, University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-8654, Japan
| | - Toshimichi Fujiwara
- Institute for Protein Research, Osaka University, 3-2 Yamadaoka, Suita, Osaka 565-0871, Japan.
| |
Collapse
|
276
|
Atanasoska M, Vazharova R, Ivanov I, Balabanski L, Andonova S, Ivanov S, Pacheva I, Malinov M, Toncheva D. SCN8A p.Arg1872Gln mutation in early infantile epileptic encephalopathy type 13: Review and case report. BIOTECHNOL BIOTEC EQ 2018. [DOI: 10.1080/13102818.2018.1532815] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/27/2022] Open
Affiliation(s)
- Maya Atanasoska
- Genetic Laboratory, Gynecology and Assisted Reproduction Hospital “Dr Malinov D.M.”, Sofia, Bulgaria
| | - Radoslava Vazharova
- Genetic Laboratory, Gynecology and Assisted Reproduction Hospital “Dr Malinov D.M.”, Sofia, Bulgaria
- Department of Biology, Medical Genetics and Microbiology, Faculty of Medicine, Sofia University “St. Kliment Ohridski”, Sofia, Bulgaria
| | - Ivan Ivanov
- Department of Paediatrics and Medical Genetics, St. George University Hospital, Medical University - Plovdiv, Plovdiv, Bulgaria
| | - Lubomir Balabanski
- Genetic Laboratory, Gynecology and Assisted Reproduction Hospital “Dr Malinov D.M.”, Sofia, Bulgaria
| | - Silvia Andonova
- National Genetic Laboratory, University Hospital of Obstetrics and Gynecology “Maichin dom”, Medical University of Sofia, Sofia, Bulgaria
| | - Samuil Ivanov
- Genetic Laboratory, Gynecology and Assisted Reproduction Hospital “Dr Malinov D.M.”, Sofia, Bulgaria
| | - Iliana Pacheva
- Department of Paediatrics and Medical Genetics, St. George University Hospital, Medical University - Plovdiv, Plovdiv, Bulgaria
| | - Maxim Malinov
- Genetic Laboratory, Gynecology and Assisted Reproduction Hospital “Dr Malinov D.M.”, Sofia, Bulgaria
| | - Draga Toncheva
- Genetic Laboratory, Gynecology and Assisted Reproduction Hospital “Dr Malinov D.M.”, Sofia, Bulgaria
- Department of Medical Genetics, Faculty of Medicine, Medical University of Sofia, Sofia, Bulgaria
| |
Collapse
|
277
|
Lin MH, Chou HC, Chen YF, Liu W, Lee CC, Liu LYM, Chuang YJ. Development of a rapid and economic in vivo electrocardiogram platform for cardiovascular drug assay and electrophysiology research in adult zebrafish. Sci Rep 2018; 8:15986. [PMID: 30375400 PMCID: PMC6207748 DOI: 10.1038/s41598-018-33577-7] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2018] [Accepted: 09/27/2018] [Indexed: 11/09/2022] Open
Abstract
Zebrafish is a popular and favorable model organism for cardiovascular research, with an increasing number of studies implementing functional assays in the adult stage. For example, the application of electrocardiography (ECG) in adult zebrafish has emerged as an important tool for cardiac pathophysiology, toxicity, and chemical screen studies. However, few laboratories are able to perform such functional analyses due to the high cost and limited availability of a convenient in vivo ECG recording system. In this study, an inexpensive ECG recording platform and operation protocol that has been optimized for adult zebrafish ECG research was developed. The core hardware includes integration of a ready-to-use portable ECG kit with a set of custom-made needle electrode probes. A combined anesthetic formula of MS-222 and isoflurane was first tested to determine the optimal assay conditions to minimize the interference to zebrafish cardiac physiology under sedation. For demonstration, we treated wild-type zebrafish with different pharmacological agents known to affect cardiac rhythms in humans. Conserved electrophysiological responses to these drugs were induced in adult zebrafish and recorded in real time. This economic ECG platform has the potential to facilitate teaching and training in cardiac electrophysiology with adult zebrafish and to promote future translational applications in cardiovascular medicine.
Collapse
Affiliation(s)
- Min-Hsuan Lin
- Department of Medical Science & Institute of Bioinformatics and Structural Biology, National Tsing Hua University, Hsinchu, 30013, Taiwan
| | - Huang-Cheng Chou
- Department of Electrical Engineering, National Tsing Hua University, Hsinchu, 30013, Taiwan
| | - Yu-Fu Chen
- Department of Medical Science, National Tsing Hua University, Hsinchu, 30013, Taiwan
| | - Wangta Liu
- Department of Biotechnology, College of Life Science, Kaohsiung Medical University, Kaohsiung, 80708, Taiwan
| | - Chi-Chun Lee
- Department of Electrical Engineering, National Tsing Hua University, Hsinchu, 30013, Taiwan
| | - Lawrence Yu-Min Liu
- Department of Medical Science & Institute of Bioinformatics and Structural Biology, National Tsing Hua University, Hsinchu, 30013, Taiwan. .,Division of Cardiology, Department of Internal Medicine, Hsinchu Mackay Memorial Hospital, Hsinchu, 30071, Taiwan.
| | - Yung-Jen Chuang
- Department of Medical Science & Institute of Bioinformatics and Structural Biology, National Tsing Hua University, Hsinchu, 30013, Taiwan.
| |
Collapse
|
278
|
Wu B, Murray JK, Andrews KL, Sham K, Long J, Aral J, Ligutti J, Amagasu S, Liu D, Zou A, Min X, Wang Z, Ilch CP, Kornecook TJ, Lin MHJ, Be X, Miranda LP, Moyer BD, Biswas K. Discovery of Tarantula Venom-Derived NaV1.7-Inhibitory JzTx-V Peptide 5-Br-Trp24 Analogue AM-6120 with Systemic Block of Histamine-Induced Pruritis. J Med Chem 2018; 61:9500-9512. [DOI: 10.1021/acs.jmedchem.8b00736] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
Affiliation(s)
| | | | | | | | | | | | | | | | | | | | - Xiaoshan Min
- Therapeutic Discovery, Amgen Research, Amgen Inc., 1120 Veterans Blvd, South San Francisco, California 94080, United States
| | - Zhulun Wang
- Therapeutic Discovery, Amgen Research, Amgen Inc., 1120 Veterans Blvd, South San Francisco, California 94080, United States
| | | | | | | | | | | | | | | |
Collapse
|
279
|
Liao Q, Li S, Siu SWI, Morlighem JÉRL, Wong CTT, Wang X, Rádis-Baptista G, Lee SMY. Novel neurotoxic peptides from Protopalythoa variabilis virtually interact with voltage-gated sodium channel and display anti-epilepsy and neuroprotective activities in zebrafish. Arch Toxicol 2018; 93:189-206. [DOI: 10.1007/s00204-018-2334-5] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2018] [Accepted: 10/10/2018] [Indexed: 02/06/2023]
|
280
|
McKerrall SJ, Sutherlin DP. Nav1.7 inhibitors for the treatment of chronic pain. Bioorg Med Chem Lett 2018; 28:3141-3149. [DOI: 10.1016/j.bmcl.2018.08.007] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2018] [Revised: 08/01/2018] [Accepted: 08/04/2018] [Indexed: 12/27/2022]
|
281
|
Gonçalves TC, Benoit E, Partiseti M, Servent D. The Na V1.7 Channel Subtype as an Antinociceptive Target for Spider Toxins in Adult Dorsal Root Ganglia Neurons. Front Pharmacol 2018; 9:1000. [PMID: 30233376 PMCID: PMC6131673 DOI: 10.3389/fphar.2018.01000] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2018] [Accepted: 08/14/2018] [Indexed: 12/11/2022] Open
Abstract
Although necessary for human survival, pain may sometimes become pathologic if long-lasting and associated with alterations in its signaling pathway. Opioid painkillers are officially used to treat moderate to severe, and even mild, pain. However, the consequent strong and not so rare complications that occur, including addiction and overdose, combined with pain management costs, remain an important societal and economic concern. In this context, animal venom toxins represent an original source of antinociceptive peptides that mainly target ion channels (such as ASICs as well as TRP, CaV, KV and NaV channels) involved in pain transmission. The present review aims to highlight the NaV1.7 channel subtype as an antinociceptive target for spider toxins in adult dorsal root ganglia neurons. It will detail (i) the characteristics of these primary sensory neurons, the first ones in contact with pain stimulus and conveying the nociceptive message, (ii) the electrophysiological properties of the different NaV channel subtypes expressed in these neurons, with a particular attention on the NaV1.7 subtype, an antinociceptive target of choice that has been validated by human genetic evidence, and (iii) the features of spider venom toxins, shaped of inhibitory cysteine knot motif, that present high affinity for the NaV1.7 subtype associated with evidenced analgesic efficacy in animal models.
Collapse
Affiliation(s)
- Tânia C Gonçalves
- Sanofi R&D, Integrated Drug Discovery - High Content Biology, Paris, France.,Service d'Ingénierie Moléculaire des Protéines, CEA de Saclay, Université Paris-Saclay, Gif-sur-Yvette, France
| | - Evelyne Benoit
- Service d'Ingénierie Moléculaire des Protéines, CEA de Saclay, Université Paris-Saclay, Gif-sur-Yvette, France.,Institut des Neurosciences Paris-Saclay, UMR CNRS/Université Paris-Sud 9197, Gif-sur-Yvette, France
| | - Michel Partiseti
- Sanofi R&D, Integrated Drug Discovery - High Content Biology, Paris, France
| | - Denis Servent
- Service d'Ingénierie Moléculaire des Protéines, CEA de Saclay, Université Paris-Saclay, Gif-sur-Yvette, France
| |
Collapse
|
282
|
Wang M, Li W, Wang Y, Song Y, Wang J, Cheng M. In silico insight into voltage-gated sodium channel 1.7 inhibition for anti-pain drug discovery. J Mol Graph Model 2018; 84:18-28. [DOI: 10.1016/j.jmgm.2018.05.006] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2018] [Revised: 05/14/2018] [Accepted: 05/14/2018] [Indexed: 12/31/2022]
|
283
|
Fang GM, Chen XX, Yang QQ, Zhu LJ, Li NN, Yu HZ, Meng XM. Discovery, structure, and chemical synthesis of disulfide-rich peptide toxins and their analogs. CHINESE CHEM LETT 2018. [DOI: 10.1016/j.cclet.2018.02.002] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
|
284
|
Ogawara H. Comparison of Strategies to Overcome Drug Resistance: Learning from Various Kingdoms. Molecules 2018; 23:E1476. [PMID: 29912169 PMCID: PMC6100412 DOI: 10.3390/molecules23061476] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2018] [Revised: 06/13/2018] [Accepted: 06/15/2018] [Indexed: 11/16/2022] Open
Abstract
Drug resistance, especially antibiotic resistance, is a growing threat to human health. To overcome this problem, it is significant to know precisely the mechanisms of drug resistance and/or self-resistance in various kingdoms, from bacteria through plants to animals, once more. This review compares the molecular mechanisms of the resistance against phycotoxins, toxins from marine and terrestrial animals, plants and fungi, and antibiotics. The results reveal that each kingdom possesses the characteristic features. The main mechanisms in each kingdom are transporters/efflux pumps in phycotoxins, mutation and modification of targets and sequestration in marine and terrestrial animal toxins, ABC transporters and sequestration in plant toxins, transporters in fungal toxins, and various or mixed mechanisms in antibiotics. Antibiotic producers in particular make tremendous efforts for avoiding suicide, and are more flexible and adaptable to the changes of environments. With these features in mind, potential alternative strategies to overcome these resistance problems are discussed. This paper will provide clues for solving the issues of drug resistance.
Collapse
Affiliation(s)
- Hiroshi Ogawara
- HO Bio Institute, Yushima-2, Bunkyo-ku, Tokyo 113-0034, Japan.
- Department of Biochemistry, Meiji Pharmaceutical University, Noshio-2, Kiyose, Tokyo 204-8588, Japan.
| |
Collapse
|
285
|
Tochitsky I, Kienzler MA, Isacoff E, Kramer RH. Restoring Vision to the Blind with Chemical Photoswitches. Chem Rev 2018; 118:10748-10773. [PMID: 29874052 DOI: 10.1021/acs.chemrev.7b00723] [Citation(s) in RCA: 89] [Impact Index Per Article: 12.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Degenerative retinal diseases such as retinitis pigmentosa (RP) and age-related macular degeneration (AMD) affect millions of people around the world and lead to irreversible vision loss if left untreated. A number of therapeutic strategies have been developed over the years to treat these diseases or restore vision to already blind patients. In this Review, we describe the development and translational application of light-sensitive chemical photoswitches to restore visual function to the blind retina and compare the translational potential of photoswitches with other vision-restoring therapies. This therapeutic strategy is enabled by an efficient fusion of chemical synthesis, chemical biology, and molecular biology and is broadly applicable to other biological systems. We hope this Review will be of interest to chemists as well as neuroscientists and clinicians.
Collapse
Affiliation(s)
- Ivan Tochitsky
- F.M. Kirby Neurobiology Center , Boston Children's Hospital , Boston , Massachusetts 02115 , United States.,Department of Neurobiology , Harvard Medical School , Boston , Massachusetts 02115 , United States
| | - Michael A Kienzler
- Department of Chemistry , University of Maine , Orono , Maine 04469 , United States
| | - Ehud Isacoff
- Department of Molecular and Cell Biology , University of California , Berkeley , California 94720 , United States.,Helen Wills Neuroscience Institute , University of California , Berkeley , California 94720 , United States.,Bioscience Division , Lawrence Berkeley National Laboratory , Berkeley , California 94720 , United States
| | - Richard H Kramer
- Department of Molecular and Cell Biology , University of California , Berkeley , California 94720 , United States.,Helen Wills Neuroscience Institute , University of California , Berkeley , California 94720 , United States
| |
Collapse
|
286
|
1,2,4-Triazolsulfone: A novel isosteric replacement of acylsulfonamides in the context of Na V 1.7 inhibition. Bioorg Med Chem Lett 2018; 28:2103-2108. [DOI: 10.1016/j.bmcl.2018.04.035] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2018] [Revised: 04/06/2018] [Accepted: 04/13/2018] [Indexed: 12/26/2022]
|
287
|
Xu L, Li D, Ding J, Pan L, Ding X. Insight into tetrodotoxin blockade and resistance mechanisms of Na v 1.2 sodium channel by theoretical approaches. Chem Biol Drug Des 2018; 92:1445-1457. [PMID: 29673065 DOI: 10.1111/cbdd.13310] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2018] [Revised: 03/08/2018] [Accepted: 03/17/2018] [Indexed: 11/30/2022]
Abstract
Nav 1.2, a member of voltage-gated sodium channels (Nav s) that are responsible for the generation and propagation of action potentials along the cell membrane, and play a vital role in the process of information transmission within the nervous system and muscle contraction, is preferentially expressed in the central nervous system. As a potent and selective blocker of Nav s, tetrodotoxin (TTX) has been extensively studied in biological and chemical sciences, whereas the detailed mechanism by which it blocks nine Nav 1 channel subtypes remain elusive. Despite the high structural similarity, the TTX metabolite 4,9-anhydro-TTX is 161 times less effective toward the mammalian Nav 1.2, which puzzled us to ask a question why such a subtle structural variation results in the largely binding affinity difference. In the current work, an integrated computational strategy, including homology modeling, induced fit docking, explicit-solvent MD simulations, and free energy calculations, was employed to investigate the binding mechanism and conformational determinants of TTX analogs. Based on the computational results, the H-bond interactions between C4-OH and C9-OH of TTX and the outer ring carboxylates of the selectivity-filter residues, and the cation-π interaction between the primary amine of guanidinium of TTX and Phe385 determine the difference of their binding affinities. Moreover, the computationally simulations were carried out for the D384N and E945K mutants of hNav 1.2-TTX, and the rank of the predicted binding free energies is in accordance with the experimental data. These observations provide a valuable model to design potent and selective neurotoxins of Nav 1.2 and shed light on the blocking mechanism of TTX to sodium channels.
Collapse
Affiliation(s)
- Lei Xu
- Institute of Bioinformatics and Medical Engineering, School of Electrical and Information Engineering, Jiangsu University of Technology, Changzhou, China
| | - Dayu Li
- Beijing Institute of Pharmaceutical Chemistry, Beijing, China
| | - Junjie Ding
- Beijing Institute of Pharmaceutical Chemistry, Beijing, China
| | - Li Pan
- Beijing Institute of Pharmaceutical Chemistry, Beijing, China
| | - Xiaoqin Ding
- Beijing Institute of Pharmaceutical Chemistry, Beijing, China
| |
Collapse
|
288
|
Focken T, Chowdhury S, Zenova A, Grimwood ME, Chabot C, Sheng T, Hemeon I, Decker SM, Wilson M, Bichler P, Jia Q, Sun S, Young C, Lin S, Goodchild SJ, Shuart NG, Chang E, Xie Z, Li B, Khakh K, Bankar G, Waldbrook M, Kwan R, Nelkenbrecher K, Karimi Tari P, Chahal N, Sojo L, Robinette CL, White AD, Chen CA, Zhang Y, Pang J, Chang JH, Hackos DH, Johnson JP, Cohen CJ, Ortwine DF, Sutherlin DP, Dehnhardt CM, Safina BS. Design of Conformationally Constrained Acyl Sulfonamide Isosteres: Identification of N-([1,2,4]Triazolo[4,3-a]pyridin-3-yl)methane-sulfonamides as Potent and Selective hNaV1.7 Inhibitors for the Treatment of Pain. J Med Chem 2018; 61:4810-4831. [DOI: 10.1021/acs.jmedchem.7b01826] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/27/2023]
Affiliation(s)
- Thilo Focken
- Xenon Pharmaceuticals, Inc., 200-3650 Gilmore Way, Burnaby, BC V5G 4W8, Canada
| | - Sultan Chowdhury
- Xenon Pharmaceuticals, Inc., 200-3650 Gilmore Way, Burnaby, BC V5G 4W8, Canada
| | - Alla Zenova
- Xenon Pharmaceuticals, Inc., 200-3650 Gilmore Way, Burnaby, BC V5G 4W8, Canada
| | - Michael E. Grimwood
- Xenon Pharmaceuticals, Inc., 200-3650 Gilmore Way, Burnaby, BC V5G 4W8, Canada
| | - Christine Chabot
- Genentech, Inc., 1 DNA Way, South San Francisco, California 94080, United States
| | - Tao Sheng
- Xenon Pharmaceuticals, Inc., 200-3650 Gilmore Way, Burnaby, BC V5G 4W8, Canada
| | - Ivan Hemeon
- Xenon Pharmaceuticals, Inc., 200-3650 Gilmore Way, Burnaby, BC V5G 4W8, Canada
| | - Shannon M. Decker
- Xenon Pharmaceuticals, Inc., 200-3650 Gilmore Way, Burnaby, BC V5G 4W8, Canada
| | - Michael Wilson
- Xenon Pharmaceuticals, Inc., 200-3650 Gilmore Way, Burnaby, BC V5G 4W8, Canada
| | - Paul Bichler
- Xenon Pharmaceuticals, Inc., 200-3650 Gilmore Way, Burnaby, BC V5G 4W8, Canada
| | - Qi Jia
- Xenon Pharmaceuticals, Inc., 200-3650 Gilmore Way, Burnaby, BC V5G 4W8, Canada
| | - Shaoyi Sun
- Xenon Pharmaceuticals, Inc., 200-3650 Gilmore Way, Burnaby, BC V5G 4W8, Canada
| | - Clint Young
- Xenon Pharmaceuticals, Inc., 200-3650 Gilmore Way, Burnaby, BC V5G 4W8, Canada
| | - Sophia Lin
- Xenon Pharmaceuticals, Inc., 200-3650 Gilmore Way, Burnaby, BC V5G 4W8, Canada
| | - Samuel J. Goodchild
- Xenon Pharmaceuticals, Inc., 200-3650 Gilmore Way, Burnaby, BC V5G 4W8, Canada
| | - Noah G. Shuart
- Xenon Pharmaceuticals, Inc., 200-3650 Gilmore Way, Burnaby, BC V5G 4W8, Canada
| | - Elaine Chang
- Xenon Pharmaceuticals, Inc., 200-3650 Gilmore Way, Burnaby, BC V5G 4W8, Canada
| | - Zhiwei Xie
- Xenon Pharmaceuticals, Inc., 200-3650 Gilmore Way, Burnaby, BC V5G 4W8, Canada
| | - Bowen Li
- Xenon Pharmaceuticals, Inc., 200-3650 Gilmore Way, Burnaby, BC V5G 4W8, Canada
| | - Kuldip Khakh
- Xenon Pharmaceuticals, Inc., 200-3650 Gilmore Way, Burnaby, BC V5G 4W8, Canada
| | - Girish Bankar
- Xenon Pharmaceuticals, Inc., 200-3650 Gilmore Way, Burnaby, BC V5G 4W8, Canada
| | - Matthew Waldbrook
- Xenon Pharmaceuticals, Inc., 200-3650 Gilmore Way, Burnaby, BC V5G 4W8, Canada
| | - Rainbow Kwan
- Xenon Pharmaceuticals, Inc., 200-3650 Gilmore Way, Burnaby, BC V5G 4W8, Canada
| | - Karen Nelkenbrecher
- Xenon Pharmaceuticals, Inc., 200-3650 Gilmore Way, Burnaby, BC V5G 4W8, Canada
| | - Parisa Karimi Tari
- Xenon Pharmaceuticals, Inc., 200-3650 Gilmore Way, Burnaby, BC V5G 4W8, Canada
| | - Navjot Chahal
- Xenon Pharmaceuticals, Inc., 200-3650 Gilmore Way, Burnaby, BC V5G 4W8, Canada
| | - Luis Sojo
- Xenon Pharmaceuticals, Inc., 200-3650 Gilmore Way, Burnaby, BC V5G 4W8, Canada
| | - C. Lee Robinette
- Xenon Pharmaceuticals, Inc., 200-3650 Gilmore Way, Burnaby, BC V5G 4W8, Canada
| | - Andrew D. White
- Chempartner, Building No. 5, 998 Halei Rd., Zhangjiang Hi-Tech
Park, Pudong New Area, Shanghai 201203, China
| | - Chien-An Chen
- Chempartner, Building No. 5, 998 Halei Rd., Zhangjiang Hi-Tech
Park, Pudong New Area, Shanghai 201203, China
| | - Yi Zhang
- Chempartner, Building No. 5, 998 Halei Rd., Zhangjiang Hi-Tech
Park, Pudong New Area, Shanghai 201203, China
| | - Jodie Pang
- Genentech, Inc., 1 DNA Way, South San Francisco, California 94080, United States
| | - Jae H. Chang
- Genentech, Inc., 1 DNA Way, South San Francisco, California 94080, United States
| | - David H. Hackos
- Genentech, Inc., 1 DNA Way, South San Francisco, California 94080, United States
| | - J. P. Johnson
- Xenon Pharmaceuticals, Inc., 200-3650 Gilmore Way, Burnaby, BC V5G 4W8, Canada
| | - Charles J. Cohen
- Xenon Pharmaceuticals, Inc., 200-3650 Gilmore Way, Burnaby, BC V5G 4W8, Canada
| | - Daniel F. Ortwine
- Genentech, Inc., 1 DNA Way, South San Francisco, California 94080, United States
| | - Daniel P. Sutherlin
- Genentech, Inc., 1 DNA Way, South San Francisco, California 94080, United States
| | | | - Brian S. Safina
- Genentech, Inc., 1 DNA Way, South San Francisco, California 94080, United States
| |
Collapse
|
289
|
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
|
290
|
Sanders SJ, Campbell AJ, Cottrell JR, Moller RS, Wagner FF, Auldridge AL, Bernier RA, Catterall WA, Chung WK, Empfield JR, George AL, Hipp JF, Khwaja O, Kiskinis E, Lal D, Malhotra D, Millichap JJ, Otis TS, Petrou S, Pitt G, Schust LF, Taylor CM, Tjernagel J, Spiro JE, Bender KJ. Progress in Understanding and Treating SCN2A-Mediated Disorders. Trends Neurosci 2018; 41:442-456. [PMID: 29691040 DOI: 10.1016/j.tins.2018.03.011] [Citation(s) in RCA: 203] [Impact Index Per Article: 29.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2018] [Revised: 03/09/2018] [Accepted: 03/14/2018] [Indexed: 01/20/2023]
Abstract
Advances in gene discovery for neurodevelopmental disorders have identified SCN2A dysfunction as a leading cause of infantile seizures, autism spectrum disorder, and intellectual disability. SCN2A encodes the neuronal sodium channel NaV1.2. Functional assays demonstrate strong correlation between genotype and phenotype. This insight can help guide therapeutic decisions and raises the possibility that ligands that selectively enhance or diminish channel function may improve symptoms. The well-defined function of sodium channels makes SCN2A an important test case for investigating the neurobiology of neurodevelopmental disorders more generally. Here, we discuss the progress made, through the concerted efforts of a diverse group of academic and industry scientists as well as policy advocates, in understanding and treating SCN2A-related disorders.
Collapse
Affiliation(s)
- Stephan J Sanders
- Department of Psychiatry, UCSF Weill Institute for Neurosciences, University of California, San Francisco, San Francisco, CA 94158, USA.
| | - Arthur J Campbell
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, 75 Ames Street, Cambridge, MA 02142, USA
| | - Jeffrey R Cottrell
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, 75 Ames Street, Cambridge, MA 02142, USA
| | - Rikke S Moller
- The Danish Epilepsy Centre, Dianalund, Denmark; Institute for Regional Health Services, University of Southern Denmark, Campusvej 55, 5230 Odense, Denmark
| | - Florence F Wagner
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, 75 Ames Street, Cambridge, MA 02142, USA
| | - Angie L Auldridge
- FamilieSCN2a Foundation, P.O. Box 82, East Longmeadow, MA 01028, USA
| | - Raphael A Bernier
- Department of Psychiatry and Behavioral Sciences, University of Washington, Seattle, WA 98195, USA
| | - William A Catterall
- Department of Pharmacology, University of Washington, Seattle, WA 98195-7280, USA
| | - Wendy K Chung
- Simons Foundation, New York, NY 10010, USA; Department of Pediatrics and Medicine, Columbia University, New York, NY 10032, USA
| | - James R Empfield
- Xenon Pharmaceuticals Inc., 3650 Gilmore Way, Burnaby, BC V5G 4W8, Canada
| | - Alfred L George
- Department of Pharmacology, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA
| | - Joerg F Hipp
- Roche Innovation Center Basel, F. Hoffmann-La Roche Ltd, Basel, Switzerland
| | - Omar Khwaja
- Roche Innovation Center Basel, F. Hoffmann-La Roche Ltd, Basel, Switzerland
| | - Evangelos Kiskinis
- Department of Neurology, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA; Department of Physiology, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA
| | - Dennis Lal
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, 75 Ames Street, Cambridge, MA 02142, USA
| | - Dheeraj Malhotra
- Roche Innovation Center Basel, F. Hoffmann-La Roche Ltd, Basel, Switzerland
| | - John J Millichap
- Department of Neurology, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA; Epilepsy Center and Division of Neurology, Ann & Robert H. Lurie Children's Hospital of Chicago, IL 60611, USA; Department of Pediatrics, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA
| | - Thomas S Otis
- Sainsbury Wellcome Centre for Neural Circuits and Behaviour, University College London, 25 Howland Street, London W1T 4JG, UK
| | - Steven Petrou
- Florey Institute of Neuroscience and Mental Health, The University of Melbourne, Parkville, VIC, Australia
| | - Geoffrey Pitt
- Cardiovascular Research Institute, Weill Cornell Medical College, Cornell University, New York, NY 10065, USA
| | - Leah F Schust
- FamilieSCN2a Foundation, P.O. Box 82, East Longmeadow, MA 01028, USA
| | - Cora M Taylor
- Geisinger Health System, 100 North Academy Avenue, Danville, PA 17822, USA
| | | | | | - Kevin J Bender
- Department of Neurology, UCSF Weill Institute for Neurosciences, University of California, San Francisco, San Francisco, CA 94158, USA.
| |
Collapse
|
291
|
Jayaraj ND, Bhattacharyya BJ, Belmadani AA, Ren D, Rathwell CA, Hackelberg S, Hopkins BE, Gupta HR, Miller RJ, Menichella DM. Reducing CXCR4-mediated nociceptor hyperexcitability reverses painful diabetic neuropathy. J Clin Invest 2018. [PMID: 29533926 DOI: 10.1172/jci92117] [Citation(s) in RCA: 59] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Painful diabetic neuropathy (PDN) is an intractable complication of diabetes that affects 25% of patients. PDN is characterized by neuropathic pain and small-fiber degeneration, accompanied by dorsal root ganglion (DRG) nociceptor hyperexcitability and loss of their axons within the skin. The molecular mechanisms underlying DRG nociceptor hyperexcitability and small-fiber degeneration in PDN are unknown. We hypothesize that chemokine CXCL12/CXCR4 signaling is central to this mechanism, as we have shown that CXCL12/CXCR4 signaling is necessary for the development of mechanical allodynia, a pain hypersensitivity behavior common in PDN. Focusing on DRG neurons expressing the sodium channel Nav1.8, we applied transgenic, electrophysiological, imaging, and chemogenetic techniques to test this hypothesis. In the high-fat diet mouse model of PDN, we were able to prevent and reverse mechanical allodynia and small-fiber degeneration by limiting CXCR4 signaling or neuronal excitability. This study reveals that excitatory CXCR4/CXCL12 signaling in Nav1.8-positive DRG neurons plays a critical role in the pathogenesis of mechanical allodynia and small-fiber degeneration in a mouse model of PDN. Hence, we propose that targeting CXCR4-mediated DRG nociceptor hyperexcitability is a promising therapeutic approach for disease-modifying treatments for this currently intractable and widespread affliction.
Collapse
Affiliation(s)
| | | | - Abdelhak A Belmadani
- Department of Pharmacology, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, USA
| | - Dongjun Ren
- Department of Pharmacology, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, USA
| | - Craig A Rathwell
- Department of Pharmacology, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, USA
| | | | - Brittany E Hopkins
- Department of Pharmacology, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, USA
| | - Herschel R Gupta
- Department of Pharmacology, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, USA
| | - Richard J Miller
- Department of Pharmacology, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, USA
| | - Daniela M Menichella
- Department of Neurology and.,Department of Pharmacology, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, USA
| |
Collapse
|
292
|
Abou Ziki MD, Seidelmann SB, Smith E, Atteya G, Jiang Y, Fernandes RG, Marieb MA, Akar JG, Mani A. Deleterious protein-altering mutations in the SCN10A voltage-gated sodium channel gene are associated with prolonged QT. Clin Genet 2018; 93:741-751. [PMID: 28407228 PMCID: PMC5640462 DOI: 10.1111/cge.13036] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2017] [Revised: 03/27/2017] [Accepted: 04/09/2017] [Indexed: 12/19/2022]
Abstract
BACKGROUND Long QT syndrome (LQT) is a pro-arrhythmogenic condition with life-threatening complications. Fifteen genes have been associated with congenital LQT, however, the genetic causes remain unknown in more than 20% of cases. MATERIALS AND METHODS Eighteen patients with history of palpitations, pre-syncope, syncope and prolonged QT were referred to the Yale Cardiovascular Genetics Program. All subjects underwent whole-exome sequencing (WES) followed by confirmatory Sanger sequencing. Mutation burden analysis was carried out using WES data from 16 subjects with no identifiable cause of LQT. RESULTS Deleterious and novel SCN10A mutations were identified in 3 of the 16 patients (19%) with idiopathic LQT. These included 2 frameshifts and 1 missense variants (p.G810fs, p.R1259Q, and p.P1877fs). Further analysis identified 2 damaging SCN10A mutations with allele frequencies of approximately 0.2% (p.R14L and p.R1268Q) in 2 independent cases. None of the SCN10A mutation carriers had mutations in known arrhythmia genes. Damaging SCN10A mutations (p.R209H and p.R485C) were also identified in the 2 subjects on QT prolonging medications. CONCLUSION Our findings implicate SCN10A in LQT. The presence of frameshift mutations suggests loss-of-function as the underlying disease mechanism. The common association with atrial fibrillation suggests a unique mechanism of disease for this LQT gene.
Collapse
Affiliation(s)
- Maen D. Abou Ziki
- Section of Cardiovascular Medicine, Department of Internal Medicine, Yale University School of Medicine, New Haven, CT, 06510
| | - Sara B. Seidelmann
- Section of Cardiovascular Medicine, Department of Internal Medicine, Yale University School of Medicine, New Haven, CT, 06510
- Cardiovascular Division, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA, 02115
| | - Emily Smith
- Section of Cardiovascular Medicine, Department of Internal Medicine, Yale University School of Medicine, New Haven, CT, 06510
| | - Gourg Atteya
- Section of Cardiovascular Medicine, Department of Internal Medicine, Yale University School of Medicine, New Haven, CT, 06510
| | - Yuexin Jiang
- Section of Cardiovascular Medicine, Department of Internal Medicine, Yale University School of Medicine, New Haven, CT, 06510
| | - Rodolfo Gil Fernandes
- Section of Cardiovascular Medicine, Department of Internal Medicine, Yale University School of Medicine, New Haven, CT, 06510
| | - Mark A. Marieb
- Section of Cardiovascular Medicine, Department of Internal Medicine, Yale University School of Medicine, New Haven, CT, 06510
| | - Joseph G. Akar
- Section of Cardiovascular Medicine, Department of Internal Medicine, Yale University School of Medicine, New Haven, CT, 06510
| | - Arya Mani
- Section of Cardiovascular Medicine, Department of Internal Medicine, Yale University School of Medicine, New Haven, CT, 06510
- Deparetment of Genetics, Yale University School of Medicine, New Haven, CT, 06510
| |
Collapse
|
293
|
Gonçalves TC, Boukaiba R, Molgó J, Amar M, Partiseti M, Servent D, Benoit E. Direct evidence for high affinity blockade of Na V1.6 channel subtype by huwentoxin-IV spider peptide, using multiscale functional approaches. Neuropharmacology 2018; 133:404-414. [PMID: 29474819 DOI: 10.1016/j.neuropharm.2018.02.016] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2017] [Revised: 01/16/2018] [Accepted: 02/19/2018] [Indexed: 12/13/2022]
Abstract
The Chinese bird spider huwentoxin-IV (HwTx-IV) is well-known to be a highly potent blocker of NaV1.7 subtype of voltage-gated sodium (NaV) channels, a genetically validated analgesic target, and thus promising as a potential lead molecule for the development of novel pain therapeutics. In the present study, the interaction between HwTx-IV and NaV1.6 channel subtype was investigated using multiscale (from in vivo to individual cell) functional approaches. HwTx-IV was approximatively 2 times more efficient than tetrodotoxin (TTX) to inhibit the compound muscle action potential recorded from the mouse skeletal neuromuscular system in vivo, and 30 times more effective to inhibit nerve-evoked than directly-elicited muscle contractile force of isolated mouse hemidiaphragms. These results strongly suggest that the inhibition of nerve-evoked skeletal muscle functioning, produced by HwTx-IV, resulted from a toxin-induced preferential blockade of NaV1.6, compared to NaV1.4, channel subtype. This was confirmed by whole-cell automated patch-clamp experiments performed on human embryonic kidney (HEK)-293 cells overexpressing hNaV1.1-1.8 channel subtypes. HwTx-IV was also approximatively 850 times more efficient to inhibit TTX-sensitive than TTX-resistant sodium currents recorded from mouse dorsal root ganglia neurons. Finally, based on our data, we predict that blockade of the NaV1.6 channel subtype was involved in the in vivo toxicity of HwTx-IV, although this toxicity was more than 2 times lower than that of TTX. In conclusion, our results provide detailed information regarding the effects of HwTx-IV and allow a better understanding of the side-effect mechanisms involved in vivo and of channel subtype interactions resulting from the toxin activity.
Collapse
Affiliation(s)
- Tânia C Gonçalves
- Sanofi R & D, Integrated Drug Discovery, In Vitro Biology & Pharmacology, F-94440, Vitry-sur-Seine, France; Service d'Ingénierie Moléculaire des Protéines (SIMOPRO), CEA, Université Paris-Saclay, F-91191, Gif sur Yvette, France
| | - Rachid Boukaiba
- Sanofi R & D, Integrated Drug Discovery, In Vitro Biology & Pharmacology, F-94440, Vitry-sur-Seine, France
| | - Jordi Molgó
- Service d'Ingénierie Moléculaire des Protéines (SIMOPRO), CEA, Université Paris-Saclay, F-91191, Gif sur Yvette, France; Institut des Neurosciences Paris-Saclay (Neuro-PSI), UMR CNRS/Université Paris-Sud 9197, Université Paris-Saclay, F-91198, Gif sur Yvette, France
| | - Muriel Amar
- Service d'Ingénierie Moléculaire des Protéines (SIMOPRO), CEA, Université Paris-Saclay, F-91191, Gif sur Yvette, France
| | - Michel Partiseti
- Sanofi R & D, Integrated Drug Discovery, In Vitro Biology & Pharmacology, F-94440, Vitry-sur-Seine, France
| | - Denis Servent
- Service d'Ingénierie Moléculaire des Protéines (SIMOPRO), CEA, Université Paris-Saclay, F-91191, Gif sur Yvette, France
| | - Evelyne Benoit
- Service d'Ingénierie Moléculaire des Protéines (SIMOPRO), CEA, Université Paris-Saclay, F-91191, Gif sur Yvette, France; Institut des Neurosciences Paris-Saclay (Neuro-PSI), UMR CNRS/Université Paris-Sud 9197, Université Paris-Saclay, F-91198, Gif sur Yvette, France.
| |
Collapse
|
294
|
Wu Y, Ma H, Zhang F, Zhang C, Zou X, Cao Z. Selective Voltage-Gated Sodium Channel Peptide Toxins from Animal Venom: Pharmacological Probes and Analgesic Drug Development. ACS Chem Neurosci 2018; 9:187-197. [PMID: 29161016 DOI: 10.1021/acschemneuro.7b00406] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Voltage-gated sodium channels (Navs) play critical roles in action potential generation and propagation. Nav channelopathy as well as pathological sensitization contribute to allodynia and hyperalgesia. Recent evidence has demonstrated the significant roles of Nav subtypes (Nav1.3, 1.7, 1.8, and 1.9) in nociceptive transduction, and therefore these Navs may represent attractive targets for analgesic drug discovery. Animal toxins are structurally diverse peptides that are highly potent yet selective on ion channel subtypes and therefore represent valuable probes to elucidate the structures, gating properties, and cellular functions of ion channels. In this review, we summarize recent advances on peptide toxins from animal venom that selectively target Nav1.3, 1.7, 1.8, and 1.9, along with their potential in analgesic drug discovery.
Collapse
Affiliation(s)
- Ying Wu
- Jiangsu Provincial Key Laboratory for TCM Evaluation
and Translational Development, China Pharmaceutical University, Nanjing 211198, China
| | - Hui Ma
- Jiangsu Provincial Key Laboratory for TCM Evaluation
and Translational Development, China Pharmaceutical University, Nanjing 211198, China
| | - Fan Zhang
- Jiangsu Provincial Key Laboratory for TCM Evaluation
and Translational Development, China Pharmaceutical University, Nanjing 211198, China
| | - Chunlei Zhang
- Jiangsu Provincial Key Laboratory for TCM Evaluation
and Translational Development, China Pharmaceutical University, Nanjing 211198, China
| | - Xiaohan Zou
- Jiangsu Provincial Key Laboratory for TCM Evaluation
and Translational Development, China Pharmaceutical University, Nanjing 211198, China
| | - Zhengyu Cao
- Jiangsu Provincial Key Laboratory for TCM Evaluation
and Translational Development, China Pharmaceutical University, Nanjing 211198, China
| |
Collapse
|
295
|
Zhao F, Jin W, Ma L, Zhang JY, Wang JL, Zhang JH, Song YB. Investigation of the selectivity of one type of small-molecule inhibitor for three Na v channel isoforms based on the method of computer simulation. J Biomol Struct Dyn 2018; 37:702-713. [PMID: 29448911 DOI: 10.1080/07391102.2018.1438921] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
Abstract
Voltage-gated sodium (Nav) channels play a pivotal role for the changes in membrane potential and belong to large membrane proteins that compose four voltage sensor domains (VSD1-4). In this study, we describe the binding mode and selectivity of one of the aryl sulfonamide sodium channel inhibitors, PF-04856264, for the VSD4s in Nav1.4, Nav1.5 and Nav1.7, respectively, through molecular dynamics simulation and enhanced post-dynamics analyses. Our results show that there are three binding site regions (BSR1-3) in the combination of the ligand and receptors, of which BSR1 and BSR3 contribute to the selectivity and affinity of the ligand to the receptor. What's more, the 39th residue (Y39 in VSD4hNav1.4/ VSD4hNav1.7 and A39 in VSD4hNav1.5) and N42 in BSR1, the 84th residue (L84 in VSD4hNav1.4, T84 in VSD4hNav1.5, and M84 in VSD4hNav1.7) in BSR2 and the conserved positive charged residues in BSR3 have major contributions to the interaction between the ligand and receptor. Further analysis reveals that if the 39th residue has a benzene ring structure, the connection of BSR1 and the ligand would be much stronger through π-stacking interaction. On the other hand, the strength and number of the hydrogen bonds formed by the ligand and the conserved arginines on S4 determine the contribution of BSR3 to the total free binding energy. We anticipate this study pave the way for the design of more effective and safe treatment for pain that selectively target Nav1.7.
Collapse
Affiliation(s)
- Fan Zhao
- a School of Life Science and Bio-pharmaceutics , Shenyang Pharmaceutical University , 103 Wenhua Road, Shenyang 110016 , China
| | - Wei Jin
- a School of Life Science and Bio-pharmaceutics , Shenyang Pharmaceutical University , 103 Wenhua Road, Shenyang 110016 , China
| | - Lin Ma
- a School of Life Science and Bio-pharmaceutics , Shenyang Pharmaceutical University , 103 Wenhua Road, Shenyang 110016 , China
| | - Jian-Ye Zhang
- a School of Life Science and Bio-pharmaceutics , Shenyang Pharmaceutical University , 103 Wenhua Road, Shenyang 110016 , China
| | - Jin-Long Wang
- a School of Life Science and Bio-pharmaceutics , Shenyang Pharmaceutical University , 103 Wenhua Road, Shenyang 110016 , China
| | - Jing-Hai Zhang
- a School of Life Science and Bio-pharmaceutics , Shenyang Pharmaceutical University , 103 Wenhua Road, Shenyang 110016 , China
| | - Yong-Bo Song
- a School of Life Science and Bio-pharmaceutics , Shenyang Pharmaceutical University , 103 Wenhua Road, Shenyang 110016 , China
| |
Collapse
|
296
|
Rubio Ayala M, Syrovets T, Hafner S, Zablotskii V, Dejneka A, Simmet T. Spatiotemporal magnetic fields enhance cytosolic Ca 2+ levels and induce actin polymerization via activation of voltage-gated sodium channels in skeletal muscle cells. Biomaterials 2018; 163:174-184. [PMID: 29471128 DOI: 10.1016/j.biomaterials.2018.02.031] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2017] [Revised: 01/27/2018] [Accepted: 02/13/2018] [Indexed: 12/13/2022]
Abstract
Cellular function is modulated by the electric membrane potential controlling intracellular physiology and signal propagation from a motor neuron to a muscle fiber resulting in muscle contraction. Unlike electric fields, magnetic fields are not attenuated by biological materials and penetrate deep into the tissue. We used complex spatiotemporal magnetic fields (17-70 mT) to control intracellular signaling in skeletal muscle cells. By changing different parameters of the alternating magnetic field (amplitude, inversion time, rotation frequency), we induced transient depolarization of cellular membranes leading to i) Na+ influx through voltage-gated sodium channels (VGSC), ii) cytosolic calcium increase, and iii) VGSC- and ryanodine receptor-dependent increase of actin polymerization. The ion fluxes occurred only, when the field was applied and returned to baseline after the field was turned off. The 30-s-activation-cycle could be repeated without any loss of signal intensity. By contrast, static magnetic fields of the same strength exhibited no effect on myotube Ca2+ levels. Mathematical modeling suggested a role for the alternating magnetic field-induced eddy current, which mediates a local change in the membrane potential triggering the activation of VGSC. These findings might pave the way for the use of complex magnetic fields to improve function of skeletal muscles in myopathies.
Collapse
Affiliation(s)
- Mónica Rubio Ayala
- Institute of Pharmacology of Natural Products & Clinical Pharmacology, Ulm University, Ulm, 89081, Germany
| | - Tatiana Syrovets
- Institute of Pharmacology of Natural Products & Clinical Pharmacology, Ulm University, Ulm, 89081, Germany
| | - Susanne Hafner
- Institute of Pharmacology of Natural Products & Clinical Pharmacology, Ulm University, Ulm, 89081, Germany
| | - Vitalii Zablotskii
- Institute of Physics Academy of Sciences of the Czech Republic, Prague 8, Czech Republic
| | - Alexandr Dejneka
- Institute of Physics Academy of Sciences of the Czech Republic, Prague 8, Czech Republic
| | - Thomas Simmet
- Institute of Pharmacology of Natural Products & Clinical Pharmacology, Ulm University, Ulm, 89081, Germany.
| |
Collapse
|
297
|
Orsini A, Zara F, Striano P. Recent advances in epilepsy genetics. Neurosci Lett 2018; 667:4-9. [DOI: 10.1016/j.neulet.2017.05.014] [Citation(s) in RCA: 56] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2016] [Revised: 04/20/2017] [Accepted: 05/08/2017] [Indexed: 01/10/2023]
|
298
|
Wu YJ, Guernon J, McClure A, Venables B, Rajamani R, Robbins KJ, Knox RJ, Matchett M, Pieschl RL, Herrington J, Bristow LJ, Meanwell NA, Olson R, Thompson LA, Dzierba C. Discovery of morpholine-based aryl sulfonamides as Na v1.7 inhibitors. Bioorg Med Chem Lett 2018; 28:958-962. [PMID: 29439904 DOI: 10.1016/j.bmcl.2018.01.035] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2017] [Revised: 01/14/2018] [Accepted: 01/19/2018] [Indexed: 12/13/2022]
Abstract
Replacement of the piperidine ring in the lead benzenesulfonamide Nav1.7 inhibitor 1 with a weakly basic morpholine core resulted in a significant reduction in Nav1.7 inhibitory activity, but the activity was restored by shortening the linkage from methyleneoxy to oxygen. These efforts led to a series of morpholine-based aryl sulfonamides as isoform-selective Nav1.7 inhibitors. This report describes the synthesis and SAR of these analogs.
Collapse
Affiliation(s)
- Yong-Jin Wu
- Bristol-Myers Squibb Research and Development, 5 Research Parkway, Wallingford, CT 06492-7660, USA.
| | - Jason Guernon
- Bristol-Myers Squibb Research and Development, 5 Research Parkway, Wallingford, CT 06492-7660, USA
| | - Andrea McClure
- Bristol-Myers Squibb Research and Development, 5 Research Parkway, Wallingford, CT 06492-7660, USA
| | - Brian Venables
- Bristol-Myers Squibb Research and Development, 5 Research Parkway, Wallingford, CT 06492-7660, USA
| | - Ramkumar Rajamani
- Bristol-Myers Squibb Research and Development, 5 Research Parkway, Wallingford, CT 06492-7660, USA
| | - Kevin J Robbins
- Bristol-Myers Squibb Research and Development, 5 Research Parkway, Wallingford, CT 06492-7660, USA
| | - Ronald J Knox
- Bristol-Myers Squibb Research and Development, 5 Research Parkway, Wallingford, CT 06492-7660, USA
| | - Michele Matchett
- Bristol-Myers Squibb Research and Development, 5 Research Parkway, Wallingford, CT 06492-7660, USA
| | - Rick L Pieschl
- Bristol-Myers Squibb Research and Development, 5 Research Parkway, Wallingford, CT 06492-7660, USA
| | - James Herrington
- Bristol-Myers Squibb Research and Development, 5 Research Parkway, Wallingford, CT 06492-7660, USA
| | - Linda J Bristow
- Bristol-Myers Squibb Research and Development, 5 Research Parkway, Wallingford, CT 06492-7660, USA
| | - Nicholas A Meanwell
- Bristol-Myers Squibb Research and Development, 5 Research Parkway, Wallingford, CT 06492-7660, USA
| | - Richard Olson
- Bristol-Myers Squibb Research and Development, 5 Research Parkway, Wallingford, CT 06492-7660, USA
| | - Lorin A Thompson
- Bristol-Myers Squibb Research and Development, 5 Research Parkway, Wallingford, CT 06492-7660, USA
| | - Carolyn Dzierba
- Bristol-Myers Squibb Research and Development, 5 Research Parkway, Wallingford, CT 06492-7660, USA
| |
Collapse
|
299
|
Baumruck AC, Tietze D, Steinacker LK, Tietze AA. Chemical synthesis of membrane proteins: a model study on the influenza virus B proton channel. Chem Sci 2018; 9:2365-2375. [PMID: 29719709 PMCID: PMC5897842 DOI: 10.1039/c8sc00004b] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/01/2018] [Accepted: 01/21/2018] [Indexed: 12/16/2022] Open
Abstract
NCL results in the quantitative yield of a membrane protein, where a thioester peptide is formed from an oxo-ester with an in situ cleavable solubilizing tag.
In the present study we have developed and optimized a robust strategy for the synthesis of highly hydrophobic peptides, especially membrane proteins, exemplarily using the influenza B M2 proton channel (BM2(1–51)). This strategy is based on the native chemical ligation of two fragments, where the thioester fragment is formed from an oxo-ester peptide, which is synthesized using Fmoc-SPPS, and features an in situ cleavable solubilizing tag (ADO, ADO2 or ADO-Lys5). The nearly quantitative production of the ligation product was followed by an optimized work up protocol, resulting in almost quantitative desulfurization and Acm-group cleavage. Circular dichroism analysis in a POPC lipid membrane revealed that the synthetic BM2(1–51) construct adopts a helical structure similar to that of the previously characterized BM2(1–33).
Collapse
Affiliation(s)
- A C Baumruck
- Darmstadt University of Technology , Clemens-Schöpf Institute of Organic Chemistry and Biochemistry , Alarich-Weiss Str. 4 , 64287 Darmstadt , Germany .
| | - D Tietze
- Darmstadt University of Technology , Eduard-Zintl-Institute of Inorganic and Physical Chemistry , Alarich-Weiss-Str. 4 , 64287 Darmstadt , Germany
| | - L K Steinacker
- Darmstadt University of Technology , Clemens-Schöpf Institute of Organic Chemistry and Biochemistry , Alarich-Weiss Str. 4 , 64287 Darmstadt , Germany .
| | - A A Tietze
- Darmstadt University of Technology , Clemens-Schöpf Institute of Organic Chemistry and Biochemistry , Alarich-Weiss Str. 4 , 64287 Darmstadt , Germany .
| |
Collapse
|
300
|
Jones KD, Stewart SG. Recent Advances in Steroid Synthesis: A Tribute to Sir Derek Barton. Aust J Chem 2018. [DOI: 10.1071/ch18256] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023]
Abstract
The synthesis of steroids and gaining an ultimate understanding of their reactivity was one of Sir Derek Barton’s most notable research areas. This highlight will focus on the construction of the steroid ring system from 2016 to 2018, and will include pathways that eventually led to natural product synthesis. For example, efficient syntheses of ent-pregnanolone sulfate and oestradiol methyl ether will be explained along with the total synthesis of cannogenol-3-O-α-l-rhamnoside.
Collapse
|