1
|
Mishra S, Mishra Y, Kumar A. Marine-derived bioactive compounds for neuropathic pain: pharmacology and therapeutic potential. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2025:10.1007/s00210-024-03667-7. [PMID: 39797987 DOI: 10.1007/s00210-024-03667-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/25/2024] [Accepted: 11/22/2024] [Indexed: 01/13/2025]
Abstract
Neuropathic pain, a challenging condition often associated with diabetes, trauma, or chemotherapy, impairs patients' quality of life. Current treatments often provide inconsistent relief and notable adverse effects, highlighting the urgent need for safer and more effective alternatives. This review investigates marine-derived bioactive compounds as potential novel therapies for neuropathic pain management. Marine organisms, including fungi, algae, cone snails, sponges, soft corals, tunicates, and fish, produce a diverse range of secondary metabolites with significant pharmacological properties. These include peptides (e.g., conopeptides, piscidin 1), non-peptides (e.g., guanidinium toxins, astaxanthin, docosahexaenoic acid, fucoidan, apigenin, fumagillin, aaptamine, flexibilide, excavatolide B, capnellenes, austrasulfones, lemnalol), and crude extracts (e.g., Spirulina platensis, Dunaliella salina, Cliothosa aurivilli). These compounds exhibit diverse mechanisms of action, such as modulating ion channels (e.g., transient receptor potential channels, voltage-gated sodium, calcium, and potassium channels, and G protein-coupled inwardly rectifying potassium channels), interacting with cell-surface receptors (e.g., nicotinic acetylcholine, NMDA, kainate, GABAB, and neurotensin receptors), inhibiting norepinephrine transporters, reducing oxidative stress, and attenuating neuroinflammation. These effects collectively contribute to alleviating nerve degeneration and symptoms of neuropathic pain, including hyperalgesia, allodynia, and associated psychomotor disturbances. Marine-derived bioactive compounds represent promising alternatives to conventional neuropathic pain treatments, to advance their development and assess their integration into neuropathic pain management strategies.
Collapse
Affiliation(s)
- Swapnil Mishra
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research (NIPER), Kolkata, West Bengal, India
| | - Yogesh Mishra
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research (NIPER), S.A.S. Nagar, Punjab, India
| | - Ashutosh Kumar
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research (NIPER), S.A.S. Nagar, Punjab, India.
| |
Collapse
|
2
|
Hemajha L, Singh S, Biji CA, Balde A, Benjakul S, Nazeer RA. A review on inflammation modulating venom proteins/peptide therapeutics and their delivery strategies: A review. Int Immunopharmacol 2024; 142:113130. [PMID: 39278056 DOI: 10.1016/j.intimp.2024.113130] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2024] [Revised: 09/02/2024] [Accepted: 09/06/2024] [Indexed: 09/17/2024]
Abstract
Inflammation is an initial biological reaction that occurs in response to infection caused by foreign pathogens or injury. This process involves a tightly controlled series of signaling events at the molecular and cellular levels, with the ultimate goal of restoring tissue balance and protecting against invading pathogens. Malfunction in the process of inflammation can result in a diverse array of diseases, such as cardiovascular, neurological, and autoimmune disorders. Therefore, the management of inflammation is of utmost importance in modern medicine. Nonsteroidal anti-inflammatory drugs (NSAIDs) and corticosteroids have long been the mainstays of pharmacological treatment for inflammation, effectively alleviating symptoms in many patients. Recently, toxins and venom, formerly seen as mostly harmful to the human body, have been recognized as possible medicinal substances for treating inflammation. Organisms that are venomous, such as spiders, scorpions, snakes, and certain marine species, have developed a wide range of powerful toxins that can effectively disable or discourage predators. Remarkably, the majority of these poisons and venoms consist of proteins and peptides, which are acknowledged as significant bioactive compounds with medicinal potential. The goal of this review is to investigate the medicinal potential of peptides derived from venoms and their complex mechanism of action in suppressing inflammation. This review also discusses various challenges and future prospects for effective venom delivery.
Collapse
Affiliation(s)
- Lakshmikanthan Hemajha
- Biopharmaceuticals Lab, Department of Biotechnology, School of Bioengineering, SRM Institute of Science and Technology, Kattankulathur, Chennai 603203, Tamilnadu, India
| | - Simran Singh
- Biopharmaceuticals Lab, Department of Biotechnology, School of Bioengineering, SRM Institute of Science and Technology, Kattankulathur, Chennai 603203, Tamilnadu, India
| | - Catherin Ann Biji
- Biopharmaceuticals Lab, Department of Biotechnology, School of Bioengineering, SRM Institute of Science and Technology, Kattankulathur, Chennai 603203, Tamilnadu, India
| | - Akshad Balde
- Biopharmaceuticals Lab, Department of Biotechnology, School of Bioengineering, SRM Institute of Science and Technology, Kattankulathur, Chennai 603203, Tamilnadu, India
| | - Soottawat Benjakul
- International Center of Excellence in Seafood Science and Innovation, Faculty of Agro Industry, Prince of Songkla University, Hat Yai, Songkhla 90110, Thailand; Department of Food and Nutrition, Kyung Hee University, Seoul 02447, Republic of Korea
| | - Rasool Abdul Nazeer
- Biopharmaceuticals Lab, Department of Biotechnology, School of Bioengineering, SRM Institute of Science and Technology, Kattankulathur, Chennai 603203, Tamilnadu, India.
| |
Collapse
|
3
|
Xu C, Wang N, Ma T, Pei S, Wang M, Yu J, Zhangsun D, Zhu X, Luo S. The α3β4 nAChR tissue distribution identified by fluorescent α-conotoxin [D11A]LvIA. Int J Biol Macromol 2024; 281:136220. [PMID: 39362420 DOI: 10.1016/j.ijbiomac.2024.136220] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2024] [Revised: 09/29/2024] [Accepted: 09/30/2024] [Indexed: 10/05/2024]
Abstract
α3β4, a vital subtype of neuronal nicotinic acetylcholine receptors (nAChRs), is widely distributed in the brain, ganglia, and adrenal glands, associated with addiction and neurological diseases. However, the lack of specific imaging tools for α3β4 nAChRs has hindered the investigation of their tissue distribution and functions. [D11A]LvIA, a peptide derived from marine cone snails, demonstrates high affinity and potency for α3β4 nAChRs, making it a valuable pharmacological tool for studying this receptor subtype. In this study, three fluorescent conjugates of [D11A]LvIA were synthesized using 6-TAMRA-SE (R), Cy3-NHS-ester (Cy3), and BODIPY-FL NHS ester (BDP) dyes. The electrophysiological activities were assessed in Xenopus laevis oocytes by two-electrodes voltage clamp (TEVC). [D11A]LvIA-Cy3 and [D11A]LvIA-BDP show improved selectivity and affinity, with IC50 values of 512.70 nM and 343.50 nM, respectively, and [D11A]LvIA-Cy3 exhibits better stability in cerebrospinal fluid (CSF). Utilizing [D11A]LvIA-Cy3, we successfully visualized the distribution of α3β4 nAChRs in rat trigeminal ganglia, retina, adrenal glands, and various brain regions. This novel fluorescent peptide provides a significant pharmacological tool for the exploration and visualization in-situ distribution of α3β4 nAChRs in different tissues and also assists in clarifying the function.
Collapse
Affiliation(s)
- Chenxing Xu
- Guangxi Key Laboratory of Special Biomedicine, School of Medicine, Guangxi University, Nanning 530004, China
| | - Nan Wang
- Guangxi Key Laboratory of Special Biomedicine, School of Medicine, Guangxi University, Nanning 530004, China
| | - Tao Ma
- Guangxi Key Laboratory of Special Biomedicine, School of Medicine, Guangxi University, Nanning 530004, China
| | - Shengrong Pei
- Guangxi Key Laboratory of Special Biomedicine, School of Medicine, Guangxi University, Nanning 530004, China
| | - Meiting Wang
- Guangxi Key Laboratory of Special Biomedicine, School of Medicine, Guangxi University, Nanning 530004, China
| | - Jinpeng Yu
- Guangxi Key Laboratory of Special Biomedicine, School of Medicine, Guangxi University, Nanning 530004, China
| | - Dongting Zhangsun
- Guangxi Key Laboratory of Special Biomedicine, School of Medicine, Guangxi University, Nanning 530004, China; Key Laboratory of Tropical Biological Resources of Ministry of Education, Hainan University, Haikou 570228, China
| | - Xiaopeng Zhu
- Guangxi Key Laboratory of Special Biomedicine, School of Medicine, Guangxi University, Nanning 530004, China.
| | - Sulan Luo
- Guangxi Key Laboratory of Special Biomedicine, School of Medicine, Guangxi University, Nanning 530004, China; Key Laboratory of Tropical Biological Resources of Ministry of Education, Hainan University, Haikou 570228, China.
| |
Collapse
|
4
|
Montigné E, Balayssac D. Exploring Cholinergic Compounds for Peripheral Neuropathic Pain Management: A Comprehensive Scoping Review of Rodent Model Studies. Pharmaceuticals (Basel) 2023; 16:1363. [PMID: 37895835 PMCID: PMC10609809 DOI: 10.3390/ph16101363] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2023] [Revised: 09/01/2023] [Accepted: 09/14/2023] [Indexed: 10/29/2023] Open
Abstract
Neuropathic pain affects about 7-8% of the population, and its management still poses challenges with unmet needs. Over the past decades, researchers have explored the cholinergic system (muscarinic and nicotinic acetylcholine receptors: mAChR and nAChR) and compounds targeting these receptors as potential analgesics for neuropathic pain management. This scoping review aims to provide an overview of studies on peripheral neuropathic pain (PNP) in rodent models, exploring compounds targeting cholinergic neurotransmission. The inclusion criteria were original articles on PNP in rodent models that explored the use of compounds directly targeting cholinergic neurotransmission and reported results of nociceptive behavioral assays. The literature search was performed in the PubMed and Web of Science databases (1 January 2000-22 April 2023). The selection process yielded 82 publications, encompassing 62 compounds. The most studied compounds were agonists of α4β2 nAChR and α7 nAChR, and antagonists of α9/α10 nAChR, along with those increasing acetylcholine and targeting mAChRs. Studies mainly reported antinociceptive effects in traumatic PNP models, and to a lesser extent, chemotherapy-induced neuropathy or diabetic models. These preclinical studies underscore the considerable potential of cholinergic compounds in the management of PNP, warranting the initiation of clinical trials.
Collapse
Affiliation(s)
- Edouard Montigné
- INSERM, U1107, NEURO-DOL, Université Clermont Auvergne, F-63000 Clermont-Ferrand, France;
| | - David Balayssac
- INSERM, U1107, NEURO-DOL, Université Clermont Auvergne, Direction de la Recherche Clinique et de l’Innovation, CHU Clermont-Ferrand, F-63000 Clermont-Ferrand, France
| |
Collapse
|
5
|
Shelukhina I, Siniavin A, Kasheverov I, Ojomoko L, Tsetlin V, Utkin Y. α7- and α9-Containing Nicotinic Acetylcholine Receptors in the Functioning of Immune System and in Pain. Int J Mol Sci 2023; 24:ijms24076524. [PMID: 37047495 PMCID: PMC10095066 DOI: 10.3390/ijms24076524] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2023] [Revised: 03/26/2023] [Accepted: 03/28/2023] [Indexed: 04/03/2023] Open
Abstract
Nicotinic acetylcholine receptors (nAChRs) present as many different subtypes in the nervous and immune systems, muscles and on the cells of other organs. In the immune system, inflammation is regulated via the vagus nerve through the activation of the non-neuronal α7 nAChR subtype, affecting the production of cytokines. The analgesic properties of α7 nAChR-selective compounds are mostly based on the activation of the cholinergic anti-inflammatory pathway. The molecular mechanism of neuropathic pain relief mediated by the inhibition of α9-containing nAChRs is not fully understood yet, but the role of immune factors in this process is becoming evident. To obtain appropriate drugs, a search of selective agonists, antagonists and modulators of α7- and α9-containing nAChRs is underway. The naturally occurring three-finger snake α-neurotoxins and mammalian Ly6/uPAR proteins, as well as neurotoxic peptides α-conotoxins, are not only sophisticated tools in research on nAChRs but are also considered as potential medicines. In particular, the inhibition of the α9-containing nAChRs by α-conotoxins may be a pathway to alleviate neuropathic pain. nAChRs are involved in the inflammation processes during AIDS and other viral infections; thus they can also be means used in drug design. In this review, we discuss the role of α7- and α9-containing nAChRs in the immune processes and in pain.
Collapse
Affiliation(s)
| | | | | | | | | | - Yuri Utkin
- Correspondence: or ; Tel.: +7-495-3366522
| |
Collapse
|
6
|
Margiotta F, Micheli L, Ciampi C, Ghelardini C, McIntosh JM, Di Cesare Mannelli L. Conus regius-Derived Conotoxins: Novel Therapeutic Opportunities from a Marine Organism. Mar Drugs 2022; 20:773. [PMID: 36547920 PMCID: PMC9783627 DOI: 10.3390/md20120773] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2022] [Revised: 12/03/2022] [Accepted: 12/06/2022] [Indexed: 12/14/2022] Open
Abstract
Conus regius is a marine venomous mollusk of the Conus genus that captures its prey by injecting a rich cocktail of bioactive disulfide bond rich peptides called conotoxins. These peptides selectively target a broad range of ion channels, membrane receptors, transporters, and enzymes, making them valuable pharmacological tools and potential drug leads. C. regius-derived conotoxins are particularly attractive due to their marked potency and selectivity against specific nicotinic acetylcholine receptor subtypes, whose signalling is involved in pain, cognitive disorders, drug addiction, and cancer. However, the species-specific differences in sensitivity and the low stability and bioavailability of these conotoxins limit their clinical development as novel therapeutic agents for these disorders. Here, we give an overview of the main pharmacological features of the C. regius-derived conotoxins described so far, focusing on the molecular mechanisms underlying their potential therapeutic effects. Additionally, we describe adoptable chemical engineering solutions to improve their pharmacological properties for future potential clinical translation.
Collapse
Affiliation(s)
- Francesco Margiotta
- Department of Neuroscience, Psychology, Drug Research and Child Health—NEUROFARBA, Pharmacology and Toxicology Section, University of Florence, 50139 Florence, Italy
| | - Laura Micheli
- Department of Neuroscience, Psychology, Drug Research and Child Health—NEUROFARBA, Pharmacology and Toxicology Section, University of Florence, 50139 Florence, Italy
| | - Clara Ciampi
- Department of Neuroscience, Psychology, Drug Research and Child Health—NEUROFARBA, Pharmacology and Toxicology Section, University of Florence, 50139 Florence, Italy
| | - Carla Ghelardini
- Department of Neuroscience, Psychology, Drug Research and Child Health—NEUROFARBA, Pharmacology and Toxicology Section, University of Florence, 50139 Florence, Italy
| | - J. Michael McIntosh
- George E. Wohlen Veterans Affairs Medical Center, Salt Lake City, UT 84148, USA
- Department of Psychiatry, University of Utah, Salt Lake City, UT 84108, USA
- School of Biological Sciences University of Utah, Salt Lake City, UT 84112, USA
| | - Lorenzo Di Cesare Mannelli
- Department of Neuroscience, Psychology, Drug Research and Child Health—NEUROFARBA, Pharmacology and Toxicology Section, University of Florence, 50139 Florence, Italy
| |
Collapse
|
7
|
A Single Amino Acid Replacement Boosts the Analgesic Activity of α-Conotoxin AuIB through the Inhibition of the GABA BR-Coupled N-Type Calcium Channel. Mar Drugs 2022; 20:md20120750. [PMID: 36547897 PMCID: PMC9781320 DOI: 10.3390/md20120750] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2022] [Revised: 11/17/2022] [Accepted: 11/22/2022] [Indexed: 11/30/2022] Open
Abstract
α-conotoxin AuIB is the only one of the 4/6 type α-conotoxins (α-CTxs) that inhibits the γ-aminobutyric acid receptor B (GABABR)-coupled N-type calcium channel (CaV2.2). To improve its inhibitory activity, a series of variants were synthesized and evaluated according to the structure-activity relationships of 4/7 type α-CTxs targeting GABABR-coupled CaV2.2. Surprisingly, only the substitution of Pro7 with Arg results in a 2-3-fold increase in the inhibition of GABABR-coupled CaV2.2 (IC50 is 0.74 nM); substitutions of position 9-12 with basic or hydrophobic amino acid and the addition of hydrophobic amino acid Leu or Ile at the second loop to mimic 4/7 type α-CTxs all failed to improve the inhibitory activity of AuIB against GABABR-coupled CaV2.2. Interestingly, the most potent form of AuIB[P7R] has disulfide bridges of "1-4, 2-3" (ribbon), which differs from the "1-3, 2-4" (globular) in the isoforms of wildtype AuIB. In addition, AuIB[P7R](globular) displays potent analgesic activity in the acetic acid writhing model and the partial sciatic nerve injury (PNL) model. Our study demonstrated that 4/6 type α-CTxs, with the disulfide bridge connectivity "1-4, 2-3," are also potent inhibitors for GABABR-coupled CaV2.2, exhibiting potent analgesic activity.
Collapse
|
8
|
Wang S, Bartels P, Zhao C, Yousuf A, Liu Z, Yu S, Bony AR, Ma X, Dai Q, Sun T, Liu N, Yang M, Yu R, Du W, Adams DJ, Dai Q. A 4/8 Subtype α-Conotoxin Vt1.27 Inhibits N-Type Calcium Channels With Potent Anti-Allodynic Effect. Front Pharmacol 2022; 13:881732. [PMID: 35754473 PMCID: PMC9230573 DOI: 10.3389/fphar.2022.881732] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2022] [Accepted: 04/08/2022] [Indexed: 11/22/2022] Open
Abstract
A novel 4/8 subtype α-conotoxin, Vt1.27 (NCCMFHTCPIDYSRFNC-NH2), was identified from Conus vitulinus in the South China Sea by RACE methods. The peptide was synthesized and structurally characterized. Similar to other α-conotoxins that target neuronal nicotinic acetylcholine receptor (nAChR) subtypes, Vt1.27 inhibited the rat α3β2 nAChR subtype (IC50 = 1160 nM) and was inactive at voltage-gated sodium and potassium channels in rat sensory neurons. However, Vt1.27 inhibited high voltage-activated N-type (CaV2.2) calcium channels expressed in HEK293T cells with an IC50 of 398 nM. An alanine scan of the peptide showed that residues Phe5, Pro9, Ile10, and Ser13 contribute significantly to the inhibitory activity of Vt1.27. The molecular dockings indicate that Vt1.27 inhibits the transmembrane region of CaV2.2, which is different from that of ω-conotoxins. Furthermore, Vt1.27 exhibited potent anti-allodynic effect in rat partial sciatic nerve injury (PNL) and chronic constriction injury (CCI) pain models at 10 nmol/kg level with the intramuscular injection. The pain threshold elevation of Vt1.27 groups was higher than that of α-conotoxin Vc1.1 in CCI rat models. These findings expand our knowledge of targets of α-conotoxins and potentially provide a potent, anti-allodynic peptide for the treatment of neuropathic pain.
Collapse
Affiliation(s)
- Shuo Wang
- Beijing Institute of Biotechnology, Beijing, China
- Department of Pharmacy, PLA Rocket Force Characteristic Medical Center, Beijing, China
| | - Peter Bartels
- Illawarra Health and Medical Research Institute (IHMRI), University of Wollongong, Wollongong, NSW, Australia
| | - Cong Zhao
- Department of Chemistry, Renmin University of China, Beijing, China
| | - Arsalan Yousuf
- Illawarra Health and Medical Research Institute (IHMRI), University of Wollongong, Wollongong, NSW, Australia
| | - Zhuguo Liu
- Beijing Institute of Biotechnology, Beijing, China
| | - Shuo Yu
- Beijing Institute of Biotechnology, Beijing, China
| | - Anuja R. Bony
- Illawarra Health and Medical Research Institute (IHMRI), University of Wollongong, Wollongong, NSW, Australia
| | - Xiaoli Ma
- Beijing Institute of Biotechnology, Beijing, China
| | - Qin Dai
- Beijing Institute of Biotechnology, Beijing, China
| | - Ting Sun
- Beijing Institute of Biotechnology, Beijing, China
| | - Na Liu
- Beijing Institute of Biotechnology, Beijing, China
| | - Mengke Yang
- School of Medicine and Pharmacy, Ocean University of China, Qingdao, China
| | - Rilei Yu
- School of Medicine and Pharmacy, Ocean University of China, Qingdao, China
| | - Weihong Du
- Department of Chemistry, Renmin University of China, Beijing, China
- *Correspondence: Qiuyun Dai, ; David J. Adams, ; Weihong Du,
| | - David J. Adams
- Illawarra Health and Medical Research Institute (IHMRI), University of Wollongong, Wollongong, NSW, Australia
- *Correspondence: Qiuyun Dai, ; David J. Adams, ; Weihong Du,
| | - Qiuyun Dai
- Beijing Institute of Biotechnology, Beijing, China
- *Correspondence: Qiuyun Dai, ; David J. Adams, ; Weihong Du,
| |
Collapse
|
9
|
Marine Origin Ligands of Nicotinic Receptors: Low Molecular Compounds, Peptides and Proteins for Fundamental Research and Practical Applications. Biomolecules 2022; 12:biom12020189. [PMID: 35204690 PMCID: PMC8961598 DOI: 10.3390/biom12020189] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2021] [Revised: 01/18/2022] [Accepted: 01/19/2022] [Indexed: 02/05/2023] Open
Abstract
The purpose of our review is to briefly show what different compounds of marine origin, from low molecular weight ones to peptides and proteins, offer for understanding the structure and mechanism of action of nicotinic acetylcholine receptors (nAChRs) and for finding novel drugs to combat the diseases where nAChRs may be involved. The importance of the mentioned classes of ligands has changed with time; a protein from the marine snake venom was the first excellent tool to characterize the muscle-type nAChRs from the electric ray, while at present, muscle and α7 receptors are labeled with the radioactive or fluorescent derivatives prepared from α-bungarotoxin isolated from the many-banded krait. The most sophisticated instruments to distinguish muscle from neuronal nAChRs, and especially distinct subtypes within the latter, are α-conotoxins. Such information is crucial for fundamental studies on the nAChR revealing the properties of their orthosteric and allosteric binding sites and mechanisms of the channel opening and closure. Similar data are provided by low-molecular weight compounds of marine origin, but here the main purpose is drug design. In our review we tried to show what has been obtained in the last decade when the listed classes of compounds were used in the nAChR research, applying computer modeling, synthetic analogues and receptor mutants, X-ray and electron-microscopy analyses of complexes with the nAChRs, and their models which are acetylcholine-binding proteins and heterologously-expressed ligand-binding domains.
Collapse
|
10
|
Yang Y, Tan Y, Zhangsun D, Zhu X, Luo S. Design, Synthesis, and Activity of an α-Conotoxin LtIA Fluorescent Analogue. ACS Chem Neurosci 2021; 12:3662-3671. [PMID: 34523332 DOI: 10.1021/acschemneuro.1c00392] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Nicotinic acetylcholine receptors (nAChRs) are essential pentameric ligand-gated ion channels that are distributed throughout the central and peripheral nervous systems and non-neuronal tissues in mammalian species that play critical roles in a variety of neural and mental activities. The α3β2 nAChR subtype participates in pain, addiction to nicotine, and other neurophysiological and pathological activities. Owing to the lack of highly selective pharmacological tools targeting α3β2, related research on its tissue distribution and function has been hindered. α-Conotoxin (α-CTx) LtIA, discovered from Conus literatus in our lab, potently and selectively blocks α3β2 nAChR, providing an important molecular probe to study the α3β2 nAChR structure and function. We used the fluorescent molecule 5-carboxytetramethylrhodamine succinimidyl ester, which can react with the N-terminus of LtIA, to obtain a novel fluorescent analogue of LtIA (LtIA-F). The potency and selectivity of LtIA-F were tested using a two-electrode voltage clamp recording on various nAChRs expressed in Xenopus laevis oocytes. LtIA-F potently inhibited ACh-evoked currents at the α3β2 nAChR, with an IC50 value of 90.66 nM, displaying a ∼4-fold decrease in potency compared with native LtIA without a change in selectivity. The serum stability results indicated that LtIA-F exhibited stability similar to that of native LtIA. This study on an α-CTx LtIA fluorescent analogue provides a wealth of pharmacological tools to explore the structure-function relationship, distribution, and ligand binding domain of the α3β2 nAChR subtype.
Collapse
Affiliation(s)
- Yishuai Yang
- Key Laboratory of Tropical Biological Resources of Ministry of Education, School of Life and Pharmaceutical Sciences, Hainan University, Haikou 570228, China
| | - Yao Tan
- Key Laboratory of Tropical Biological Resources of Ministry of Education, School of Life and Pharmaceutical Sciences, Hainan University, Haikou 570228, China
| | - Dongting Zhangsun
- Key Laboratory of Tropical Biological Resources of Ministry of Education, School of Life and Pharmaceutical Sciences, Hainan University, Haikou 570228, China
| | - Xiaopeng Zhu
- Medical School, Guangxi University, Nanning 530004, China
| | - Sulan Luo
- Key Laboratory of Tropical Biological Resources of Ministry of Education, School of Life and Pharmaceutical Sciences, Hainan University, Haikou 570228, China
- Medical School, Guangxi University, Nanning 530004, China
| |
Collapse
|
11
|
Jimenez EC. Post-translationally modified conopeptides: Biological activities and pharmacological applications. Peptides 2021; 139:170525. [PMID: 33684482 DOI: 10.1016/j.peptides.2021.170525] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/30/2020] [Revised: 02/20/2021] [Accepted: 02/24/2021] [Indexed: 10/25/2022]
Abstract
Conus venoms comprise a large variety of biologically active peptides (conopeptides or conotoxins) that are employed for prey capture and other biological functions. Throughout the course of evolution of the cone snails, they have developed an envenomation scheme that necessitates a potent mixture of peptides, most of which are highly post-translationally modified, that can cause rapid paralysis of their prey. The great diversity of these peptides defines the ecological interactions and evolutionary strategy of cone snails. Such scheme has led to some pharmacological applications for pain, epilepsy, and myocardial infarction, that could be further explored to ultimately find unique peptide-based therapies. This review focuses on ∼ 60 representative post-translationally modified conopeptides that were isolated from Conus venoms. Various conopeptides reveal post-translational modifications of specific amino acids, such as hydroxylation of proline and lysine, gamma-carboxylation of glutamate, formation of N-terminal pyroglutamate, isomerization of l- to d-amino acid, bromination of tryptophan, O-glycosylation of threonine or serine, sulfation of tyrosine, and cysteinylation of cysteine, other than the more common disulfide crosslinking and C-terminal amidation. Many of the post-translationally modified peptides paved the way for the characterization, by alternative analytical methods, of other pharmacologically important peptides that are classified under 27 conopeptide families denoting pharmacological classes.
Collapse
Affiliation(s)
- Elsie C Jimenez
- Department of Physical Sciences, College of Science, University of the Philippines Baguio, Baguio City, 2600, Philippines.
| |
Collapse
|
12
|
Belgi A, Burnley JV, MacRaild CA, Chhabra S, Elnahriry KA, Robinson SD, Gooding SG, Tae HS, Bartels P, Sadeghi M, Zhao FY, Wei H, Spanswick D, Adams DJ, Norton RS, Robinson AJ. Alkyne-Bridged α-Conotoxin Vc1.1 Potently Reverses Mechanical Allodynia in Neuropathic Pain Models. J Med Chem 2021; 64:3222-3233. [PMID: 33724033 DOI: 10.1021/acs.jmedchem.0c02151] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Several Conus-derived venom peptides are promising lead compounds for the management of neuropathic pain, with α-conotoxins being of particular interest. Modification of the interlocked disulfide framework of α-conotoxin Vc1.1 has been achieved using on-resin alkyne metathesis. Although introduction of a metabolically stable alkyne motif significantly disrupts backbone topography, the structural modification generates a potent and selective GABAB receptor agonist that inhibits Cav2.2 channels and exhibits dose-dependent reversal of mechanical allodynia in a behavioral rat model of neuropathic pain. The findings herein support the hypothesis that analgesia can be achieved via activation of GABABRs expressed in dorsal root ganglion (DRG) sensory neurons.
Collapse
Affiliation(s)
- Alessia Belgi
- School of Chemistry, Monash University, Clayton, Victoria 3800, Australia
| | - James V Burnley
- School of Chemistry, Monash University, Clayton, Victoria 3800, Australia
| | - Christopher A MacRaild
- Medicinal Chemistry, Monash Institute of Pharmaceutical Science, Monash University, 381 Royal Parade, Parkville, Victoria 3052, Australia
| | - Sandeep Chhabra
- Medicinal Chemistry, Monash Institute of Pharmaceutical Science, Monash University, 381 Royal Parade, Parkville, Victoria 3052, Australia
| | - Khaled A Elnahriry
- Medicinal Chemistry, Monash Institute of Pharmaceutical Science, Monash University, 381 Royal Parade, Parkville, Victoria 3052, Australia
| | - Samuel D Robinson
- Medicinal Chemistry, Monash Institute of Pharmaceutical Science, Monash University, 381 Royal Parade, Parkville, Victoria 3052, Australia
| | - Simon G Gooding
- School of Chemistry, Monash University, Clayton, Victoria 3800, Australia
| | - Han-Shen Tae
- Illawarra Health & Medical Research Institute (IHMRI), University of Wollongong, Wollongong, New South Wales 2522, Australia
| | - Peter Bartels
- Illawarra Health & Medical Research Institute (IHMRI), University of Wollongong, Wollongong, New South Wales 2522, Australia
| | - Mahsa Sadeghi
- Illawarra Health & Medical Research Institute (IHMRI), University of Wollongong, Wollongong, New South Wales 2522, Australia
| | | | | | - David Spanswick
- NeuroSolutions Ltd., Coventry CV4 7AL, U.K
- Biomedicine Discovery Institute and the Department of Physiology, Monash University, Clayton, Victoria 3800, Australia
- Warwick Medical School, University of Warwick, Coventry CV4 7AL, U.K
| | - David J Adams
- Illawarra Health & Medical Research Institute (IHMRI), University of Wollongong, Wollongong, New South Wales 2522, Australia
| | - Raymond S Norton
- Medicinal Chemistry, Monash Institute of Pharmaceutical Science, Monash University, 381 Royal Parade, Parkville, Victoria 3052, Australia
- ARC Centre for Fragment-Based Design, Monash University, Parkville, Victoria 3052, Australia
| | - Andrea J Robinson
- School of Chemistry, Monash University, Clayton, Victoria 3800, Australia
| |
Collapse
|
13
|
Li X, Tae HS, Chu Y, Jiang T, Adams DJ, Yu R. Medicinal chemistry, pharmacology, and therapeutic potential of α-conotoxins antagonizing the α9α10 nicotinic acetylcholine receptor. Pharmacol Ther 2020; 222:107792. [PMID: 33309557 DOI: 10.1016/j.pharmthera.2020.107792] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2020] [Accepted: 12/07/2020] [Indexed: 12/20/2022]
Abstract
α-Conotoxins are disulfide-rich and well-structured peptides, most of which can block nicotinic acetylcholine receptors (nAChRs) with exquisite selectivity and potency. There are various nAChR subtypes, of which the α9α10 nAChR functions as a heteromeric ionotropic receptor in the mammalian cochlea and mediates postsynaptic transmission from the medial olivocochlear. The α9α10 nAChR subtype has also been proposed as a target for the treatment of neuropathic pain and the suppression of breast cancer cell proliferation. Therefore, α-conotoxins targeting the α9α10 nAChR are potentially useful in the development of specific therapeutic drugs and pharmacological tools. Despite dissimilarities in their amino acid sequence and structures, these conopeptides are potent antagonists of the α9α10 nAChR subtype. Consequently, the activity and stability of these peptides have been subjected to chemical modifications. The resulting synthetic analogues have not only functioned as molecular probes to explore ligand binding sites of the α9α10 nAChR, but also have the potential to become candidates for drug development. From the perspectives of medicinal chemistry and pharmacology, we highlight the structure and function of the α9α10 nAChR and review studies of α-conotoxins targeting it, including their three-dimensional structures, structure optimization strategies, and binding modes at the α9α10 nAChR, as well as their therapeutic potential.
Collapse
Affiliation(s)
- Xiao Li
- Key Laboratory of Marine Drugs, Chinese Ministry of Education, School of Medicine and Pharmacy, Ocean University of China, 5 Yushan Road, Qingdao 266003, China; Laboratory for Marine Drugs and Bioproducts, Qingdao National Laboratory for Marine Science and Technology, Qingdao 266003, China
| | - Han-Shen Tae
- Illawarra Health and Medical Research Institute (IHMRI), University of Wollongong, Wollongong, New South Wales 2522, Australia
| | - Yanyan Chu
- Key Laboratory of Marine Drugs, Chinese Ministry of Education, School of Medicine and Pharmacy, Ocean University of China, 5 Yushan Road, Qingdao 266003, China; Laboratory for Marine Drugs and Bioproducts, Qingdao National Laboratory for Marine Science and Technology, Qingdao 266003, China; Innovation Platform of Marine Drug Screening & Evaluation, Qingdao National Laboratory for Marine Science and Technology, Qingdao, Shandong 266100, China
| | - Tao Jiang
- Key Laboratory of Marine Drugs, Chinese Ministry of Education, School of Medicine and Pharmacy, Ocean University of China, 5 Yushan Road, Qingdao 266003, China; Laboratory for Marine Drugs and Bioproducts, Qingdao National Laboratory for Marine Science and Technology, Qingdao 266003, China
| | - David J Adams
- Illawarra Health and Medical Research Institute (IHMRI), University of Wollongong, Wollongong, New South Wales 2522, Australia.
| | - Rilei Yu
- Key Laboratory of Marine Drugs, Chinese Ministry of Education, School of Medicine and Pharmacy, Ocean University of China, 5 Yushan Road, Qingdao 266003, China; Laboratory for Marine Drugs and Bioproducts, Qingdao National Laboratory for Marine Science and Technology, Qingdao 266003, China; Innovation Platform of Marine Drug Screening & Evaluation, Qingdao National Laboratory for Marine Science and Technology, Qingdao, Shandong 266100, China.
| |
Collapse
|
14
|
Li X, Wang S, Zhu X, Zhangsun D, Wu Y, Luo S. Effects of Cyclization on Activity and Stability of α-Conotoxin TxIB. Mar Drugs 2020; 18:E180. [PMID: 32235388 PMCID: PMC7230940 DOI: 10.3390/md18040180] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2020] [Revised: 03/25/2020] [Accepted: 03/25/2020] [Indexed: 11/17/2022] Open
Abstract
α-Conotoxin TxIB specifically blocked α6/α3β2β3 acetylcholine receptors (nAChRs), and it could be a potential probe for studying addiction and other diseases related to α6/α3β2β3 nAChRs. However, as a peptide, TxIB may suffer from low stability, short half-life, and poor bioavailability. In this study, cyclization of TxIB was used to improve its stability. Four cyclic mutants of TxIB (cTxIB) were synthesized, and the inhibition of these analogues on α6/α3β2β3 nAChRs as well as their stability in human serum were measured. All cyclized analogues had similar activity compared to wild-type TxIB, which indicated that backbone cyclization of TxIB had no significant effect on its activity. Cyclization of TxIB with a seven-residue linker improved its stability significantly in human serum. Besides this, the results showed that cyclization maintained the activity of α-conotoxin TxIB, which is conducive to its future application.
Collapse
Affiliation(s)
- Xincan Li
- Key Laboratory of Tropical Biological Resources of Ministry of Education, Key Laboratory for Marine Drugs of Haikou, School of Life and Pharmaceutical Sciences, Hainan University, Haikou 570228, China; (X.L.); (S.W.); (X.Z.); (D.Z.)
| | - Shuai Wang
- Key Laboratory of Tropical Biological Resources of Ministry of Education, Key Laboratory for Marine Drugs of Haikou, School of Life and Pharmaceutical Sciences, Hainan University, Haikou 570228, China; (X.L.); (S.W.); (X.Z.); (D.Z.)
| | - Xiaopeng Zhu
- Key Laboratory of Tropical Biological Resources of Ministry of Education, Key Laboratory for Marine Drugs of Haikou, School of Life and Pharmaceutical Sciences, Hainan University, Haikou 570228, China; (X.L.); (S.W.); (X.Z.); (D.Z.)
| | - Dongting Zhangsun
- Key Laboratory of Tropical Biological Resources of Ministry of Education, Key Laboratory for Marine Drugs of Haikou, School of Life and Pharmaceutical Sciences, Hainan University, Haikou 570228, China; (X.L.); (S.W.); (X.Z.); (D.Z.)
- Medical School, Guangxi University, Nanning 530004, China
| | - Yong Wu
- Key Laboratory of Tropical Biological Resources of Ministry of Education, Key Laboratory for Marine Drugs of Haikou, School of Life and Pharmaceutical Sciences, Hainan University, Haikou 570228, China; (X.L.); (S.W.); (X.Z.); (D.Z.)
- Medical School, Guangxi University, Nanning 530004, China
| | - Sulan Luo
- Key Laboratory of Tropical Biological Resources of Ministry of Education, Key Laboratory for Marine Drugs of Haikou, School of Life and Pharmaceutical Sciences, Hainan University, Haikou 570228, China; (X.L.); (S.W.); (X.Z.); (D.Z.)
- Medical School, Guangxi University, Nanning 530004, China
| |
Collapse
|
15
|
Toma W, Ulker E, Alqasem M, AlSharari SD, McIntosh JM, Damaj MI. Behavioral and Molecular Basis of Cholinergic Modulation of Pain: Focus on Nicotinic Acetylcholine Receptors. Curr Top Behav Neurosci 2020; 45:153-166. [PMID: 32468494 DOI: 10.1007/7854_2020_135] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/11/2023]
Abstract
Nicotinic acetylcholine receptors (nAChRs) have emerged as a novel therapeutic strategy for pain and inflammatory disorders. In particular, α4β2∗, α7, and α9α10 nAChR subtypes have been investigated as potential targets to treat pain. The nAChRs are distributed on the pain transmission pathways, including central and peripheral nervous systems and immune cells as well. Several agonists for α4β2∗ nAChR subtypes have been investigated in multiple animal pain models with promising results. However, studies in human indicated a narrow therapeutic window for α4β2∗ agonists. Furthermore, animal studies suggest that using agonists for α7 nAChR subtype and antagonists for α9α10 nAChR subtypes are potential novel therapies for chronic pain management, including inflammatory and neuropathic pain. More recently, alternative nAChRs ligands such as positive allosteric modulators and silent agonists have shown potential to develop into new treatments for chronic pain.
Collapse
Affiliation(s)
- Wisam Toma
- Department of Pharmacology and Toxicology, Virginia Commonwealth University, Richmond, VA, USA
| | - Esad Ulker
- Department of Pharmacology and Toxicology, Virginia Commonwealth University, Richmond, VA, USA
| | - Mashael Alqasem
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, Riyadh, Saudi Arabia
| | - Shakir D AlSharari
- Department of Pharmacology and Toxicology, Virginia Commonwealth University, Richmond, VA, USA
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, Riyadh, Saudi Arabia
| | - J Michael McIntosh
- Departments of Psychiatry and Biology, University of Utah, Salt Lake City, UT, USA
| | - M Imad Damaj
- Department of Pharmacology and Toxicology, Virginia Commonwealth University, Richmond, VA, USA.
| |
Collapse
|
16
|
Xu P, Xiong Y, Liu Y, Yu S, Zhangsun D, Wu Y, Luo S. Degradation kinetics of α-conotoxin TxID. FEBS Open Bio 2019; 9:1561-1572. [PMID: 31278882 PMCID: PMC6722883 DOI: 10.1002/2211-5463.12697] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2019] [Revised: 06/27/2019] [Accepted: 07/04/2019] [Indexed: 12/05/2022] Open
Abstract
α‐Conotoxin (CTx) TxID is a potent α3β4 nicotinic acetylcholine receptor (nAChR) antagonist that has been suggested as a potential drug candidate to treat addiction and small cell lung cancer. The function and structure of TxID have been well‐studied, but analyses of its stability have not previously been reported. The purpose of this study was to analyze the stability and forced degradation of TxID under various conditions: acid, alkali, water hydrolysis, oxidation, light, thiols, temperature, ionic strength and buffer pH. Different degradation products were formed under various conditions, and the degradation patterns of TxID showed pseudo‐first‐order kinetics. TxID degraded slowest at pH 3 within a pH range of 2–8. The major degradation products were analyzed using liquid chromatography–tandem mass spectrometry and the activity of the main product with α3β4 nAChR was analyzed using electrophysiological methods. Our analysis of TxID stability may aid the selection of appropriate conditions for peptide production, packaging and storage.
Collapse
Affiliation(s)
- Pan Xu
- Key Laboratory of Tropical Biological Resources of Ministry of Education, Key Lab for Marine Drugs of Haikou, School of Life and Pharmaceutical Sciences, Hainan University, Haikou, Hainan, China
| | - Yang Xiong
- Key Laboratory of Tropical Biological Resources of Ministry of Education, Key Lab for Marine Drugs of Haikou, School of Life and Pharmaceutical Sciences, Hainan University, Haikou, Hainan, China
| | - Yiqiao Liu
- Key Laboratory of Tropical Biological Resources of Ministry of Education, Key Lab for Marine Drugs of Haikou, School of Life and Pharmaceutical Sciences, Hainan University, Haikou, Hainan, China
| | - Shurun Yu
- Key Laboratory of Tropical Biological Resources of Ministry of Education, Key Lab for Marine Drugs of Haikou, School of Life and Pharmaceutical Sciences, Hainan University, Haikou, Hainan, China
| | - Dongting Zhangsun
- Key Laboratory of Tropical Biological Resources of Ministry of Education, Key Lab for Marine Drugs of Haikou, School of Life and Pharmaceutical Sciences, Hainan University, Haikou, Hainan, China
| | - Yong Wu
- Key Laboratory of Tropical Biological Resources of Ministry of Education, Key Lab for Marine Drugs of Haikou, School of Life and Pharmaceutical Sciences, Hainan University, Haikou, Hainan, China
| | - Sulan Luo
- Key Laboratory of Tropical Biological Resources of Ministry of Education, Key Lab for Marine Drugs of Haikou, School of Life and Pharmaceutical Sciences, Hainan University, Haikou, Hainan, China
| |
Collapse
|
17
|
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
|
18
|
Abstract
In order to improve stability of a peptide marine drug lead, α-conotoxin TxID, we synthesized and modified TxID at the N-terminal with DSPE-PEG-NHS by a nucleophilic substitution reaction to prepare the DSPE-PEG-TxID for the first time. The reaction conditions, including solvent, ratio, pH, and reaction time, were optimized systematically and the optimal one was reacted in dimethyl formamide at pH 8.2 with triethylamine at room temperature for 120 h. The in vitro stabilities in serum, simulated gastric juice, and intestinal fluid were tested, and improved dramatically compared with TxID. The PEG-modified peptide was functionally tested on α3β4 nicotinic acetylcholine receptor (nAChR) heterologously expressed in Xenopus laevis oocytes. The DSPE-PEG-TxID showed an obvious inhibition effect on α3β4 nAChR. All in all, the PEG modification of TxID was improved in stability, resistance to enzymatic degradation, and may prolong the half-life in vivo, which may pave the way for the future application in smoking cessation and drug rehabilitation, as well as small cell lung cancer.
Collapse
|
19
|
Cai F, Xu N, Liu Z, Ding R, Yu S, Dong M, Wang S, Shen J, Tae HS, Adams DJ, Zhang X, Dai Q. Targeting of N-Type Calcium Channels via GABAB-Receptor Activation by α-Conotoxin Vc1.1 Variants Displaying Improved Analgesic Activity. J Med Chem 2018; 61:10198-10205. [DOI: 10.1021/acs.jmedchem.8b01343] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Affiliation(s)
- Fengtao Cai
- Beijing Institute of Biotechnology, Beijing 100071, China
- School of Preclinical Medicine, Guangxi Medical University, Nanning 530021, China
| | - Ning Xu
- Beijing Institute of Biotechnology, Beijing 100071, China
| | - Zhuguo Liu
- Beijing Institute of Biotechnology, Beijing 100071, China
| | - Rong Ding
- Beijing Institute of Biotechnology, Beijing 100071, China
| | - Shuo Yu
- Beijing Institute of Biotechnology, Beijing 100071, China
| | - Mingxin Dong
- Beijing Institute of Biotechnology, Beijing 100071, China
| | - Shuo Wang
- Beijing Institute of Biotechnology, Beijing 100071, China
| | - Jintao Shen
- Beijing Institute of Biotechnology, Beijing 100071, China
| | - Han-Shen Tae
- Illawarra Health and Medical Research Institute (IHMRI), University of Wollongong, Wollongong, NSW 2522, Australia
| | - David J. Adams
- Illawarra Health and Medical Research Institute (IHMRI), University of Wollongong, Wollongong, NSW 2522, Australia
| | - Xuerong Zhang
- School of Preclinical Medicine, Guangxi Medical University, Nanning 530021, China
| | - Qiuyun Dai
- Beijing Institute of Biotechnology, Beijing 100071, China
| |
Collapse
|
20
|
Hamad MK, He K, Abdulrazeq HF, Mustafa AM, Luceri R, Kamal N, Ali M, Nakhla J, Herzallah MM, Mammis A. Potential Uses of Isolated Toxin Peptides in Neuropathic Pain Relief: A Literature Review. World Neurosurg 2018; 113:333-347.e5. [DOI: 10.1016/j.wneu.2018.01.116] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2017] [Revised: 01/15/2018] [Accepted: 01/16/2018] [Indexed: 01/31/2023]
|
21
|
Chen J, Liang L, Ning H, Cai F, Liu Z, Zhang L, Zhou L, Dai Q. Cloning, Synthesis and Functional Characterization of a Novel α-Conotoxin Lt1.3. Mar Drugs 2018; 16:md16040112. [PMID: 29614714 PMCID: PMC5923399 DOI: 10.3390/md16040112] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2018] [Revised: 03/16/2018] [Accepted: 03/22/2018] [Indexed: 01/28/2023] Open
Abstract
α-Conotoxins (α-CTxs) are small peptides composed of 11 to 20 amino acid residues with two disulfide bridges. Most of them potently and selectively target nicotinic acetylcholine receptor (nAChR) subtypes, and a few were found to inhibit the GABAB receptor (GABABR)-coupled N-type calcium channels (Cav2.2). However, in all of α-CTxs targeting both receptors, the disulfide connectivity arrangement "C¹-C³, C²-C⁴" is present. In this work, a novel α4/7-CTx named Lt1.3 (GCCSHPACSGNNPYFC-NH₂) was cloned from the venom ducts of Conus litteratus (C. litteratus) in the South China Sea. Lt1.3 was then chemically synthesized and two isomers with disulfide bridges "C¹-C³, C²-C⁴" and "C¹-C⁴, C²-C³" were found and functionally characterized. Electrophysiological experiments showed that Lt1.3 containing the common disulfide bridges "C¹-C³, C²-C⁴" potently and selectively inhibited α3β2 nAChRs and not GABABR-coupled Cav2.2. Surprisingly, but the isomer with the disulfide bridges "C¹-C⁴, C²-C³" showed exactly the opposite inhibitory activity, inhibiting only GABABR-coupled Cav2.2 and not α3β2 nAChRs. These findings expand the knowledge of the targets and selectivity of α-CTxs and provide a new structural motif to inhibit the GABABR-coupled Cav2.2.
Collapse
Affiliation(s)
- Jinqin Chen
- Beijing Institute of Biotechnology, Beijing 100071, China.
- Institute of Physical Science and Information Technology, Anhui University, Hefei 236041, China.
| | - Li Liang
- Beijing Institute of Biotechnology, Beijing 100071, China.
| | - Huying Ning
- Beijing Institute of Biotechnology, Beijing 100071, China.
| | - Fengtao Cai
- Beijing Institute of Biotechnology, Beijing 100071, China.
| | - Zhuguo Liu
- Beijing Institute of Biotechnology, Beijing 100071, China.
| | - Longxiao Zhang
- Beijing Institute of Biotechnology, Beijing 100071, China.
| | - Liangyi Zhou
- Beijing Institute of Biotechnology, Beijing 100071, China.
| | - Qiuyun Dai
- Beijing Institute of Biotechnology, Beijing 100071, China.
| |
Collapse
|
22
|
A novel α-conopeptide Eu1.6 inhibits N-type (Ca V2.2) calcium channels and exhibits potent analgesic activity. Sci Rep 2018; 8:1004. [PMID: 29343689 PMCID: PMC5772529 DOI: 10.1038/s41598-017-18479-4] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2017] [Accepted: 12/06/2017] [Indexed: 01/13/2023] Open
Abstract
We here describe a novel α-conopeptide, Eu1.6 from Conus eburneus, which exhibits strong anti-nociceptive activity by an unexpected mechanism of action. Unlike other α-conopeptides that largely target nicotinic acetylcholine receptors (nAChRs), Eu1.6 displayed only weak inhibitory activity at the α3β4 and α7 nAChR subtypes and TTX-resistant sodium channels, and no activity at TTX-sensitive sodium channels in rat dorsal root ganglion (DRG) neurons, or opiate receptors, VR1, KCNQ1, L- and T-type calcium channels expressed in HEK293 cells. However, Eu1.6 inhibited high voltage-activated N-type calcium channel currents in isolated mouse DRG neurons which was independent of GABAB receptor activation. In HEK293 cells expressing CaV2.2 channels alone, Eu1.6 reversibly inhibited depolarization-activated Ba2+ currents in a voltage- and state-dependent manner. Inhibition of CaV2.2 by Eu1.6 was concentration-dependent (IC50 ~1 nM). Significantly, systemic administration of Eu1.6 at doses of 2.5–5.0 μg/kg exhibited potent analgesic activities in rat partial sciatic nerve injury and chronic constriction injury pain models. Furthermore, Eu1.6 had no significant side-effect on spontaneous locomotor activity, cardiac and respiratory function, and drug dependence in mice. These findings suggest α-conopeptide Eu1.6 is a potent analgesic for the treatment of neuropathic and chronic pain and opens a novel option for future analgesic drug design.
Collapse
|
23
|
α-Conotoxins to explore the molecular, physiological and pathophysiological functions of neuronal nicotinic acetylcholine receptors. Neurosci Lett 2017; 679:24-34. [PMID: 29199094 DOI: 10.1016/j.neulet.2017.11.063] [Citation(s) in RCA: 47] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2017] [Revised: 11/28/2017] [Accepted: 11/29/2017] [Indexed: 12/22/2022]
Abstract
The vast diversity of neuronal nicotinic acetylcholine subunits expressed in the central and peripheral nervous systems, as well as in non-neuronal tissues, constitutes a formidable challenge for researchers and clinicians to decipher the role of particular subtypes, including complex subunit associations, in physiological and pathophysiological functions. Many natural products target the nAChRs, but there is no richer source of nicotinic ligands than the venom of predatory gastropods known as cone snails. Indeed, every single species of cone snail was shown to produce at least one type of such α-conotoxins. These tiny peptides (10-25 amino acids), constrained by disulfide bridges, proved to be unvaluable tools to investigate the structure and function of nAChRs, some of them having also therapeutic potential. In this review, we provide a recent update on the pharmacology and subtype specificity of several major α-conotoxins.
Collapse
|
24
|
G-Protein Coupled Receptors Targeted by Analgesic Venom Peptides. Toxins (Basel) 2017; 9:toxins9110372. [PMID: 29144441 PMCID: PMC5705987 DOI: 10.3390/toxins9110372] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2017] [Revised: 11/13/2017] [Accepted: 11/13/2017] [Indexed: 12/14/2022] Open
Abstract
Chronic pain is a complex and debilitating condition associated with a large personal and socioeconomic burden. Current pharmacological approaches to treating chronic pain such as opioids, antidepressants and anticonvulsants exhibit limited efficacy in many patients and are associated with dose-limiting side effects that hinder their clinical use. Therefore, improved strategies for the pharmacological treatment of pathological pain are urgently needed. G-protein coupled receptors (GPCRs) are ubiquitously expressed on the surface of cells and act to transduce extracellular signals and regulate physiological processes. In the context of pain, numerous and diverse families of GPCRs expressed in pain pathways regulate most aspects of physiological and pathological pain and are thus implicated as potential targets for therapy of chronic pain. In the search for novel compounds that produce analgesia via GPCR modulation, animal venoms offer an enormous and virtually untapped source of potent and selective peptide molecules. While many venom peptides target voltage-gated and ligand-gated ion channels to inhibit neuronal excitability and blunt synaptic transmission of pain signals, only a small proportion are known to interact with GPCRs. Of these, only a few have shown analgesic potential in vivo. Here we review the current state of knowledge regarding venom peptides that target GPCRs to produce analgesia, and their development as therapeutic compounds.
Collapse
|
25
|
Leffler AE, Kuryatov A, Zebroski HA, Powell SR, Filipenko P, Hussein AK, Gorson J, Heizmann A, Lyskov S, Tsien RW, Poget SF, Nicke A, Lindstrom J, Rudy B, Bonneau R, Holford M. Discovery of peptide ligands through docking and virtual screening at nicotinic acetylcholine receptor homology models. Proc Natl Acad Sci U S A 2017; 114:E8100-E8109. [PMID: 28874590 PMCID: PMC5617267 DOI: 10.1073/pnas.1703952114] [Citation(s) in RCA: 48] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Venom peptide toxins such as conotoxins play a critical role in the characterization of nicotinic acetylcholine receptor (nAChR) structure and function and have potential as nervous system therapeutics as well. However, the lack of solved structures of conotoxins bound to nAChRs and the large size of these peptides are barriers to their computational docking and design. We addressed these challenges in the context of the α4β2 nAChR, a widespread ligand-gated ion channel in the brain and a target for nicotine addiction therapy, and the 19-residue conotoxin α-GID that antagonizes it. We developed a docking algorithm, ToxDock, which used ensemble-docking and extensive conformational sampling to dock α-GID and its analogs to an α4β2 nAChR homology model. Experimental testing demonstrated that a virtual screen with ToxDock correctly identified three bioactive α-GID mutants (α-GID[A10V], α-GID[V13I], and α-GID[V13Y]) and one inactive variant (α-GID[A10Q]). Two mutants, α-GID[A10V] and α-GID[V13Y], had substantially reduced potency at the human α7 nAChR relative to α-GID, a desirable feature for α-GID analogs. The general usefulness of the docking algorithm was highlighted by redocking of peptide toxins to two ion channels and a binding protein in which the peptide toxins successfully reverted back to near-native crystallographic poses after being perturbed. Our results demonstrate that ToxDock can overcome two fundamental challenges of docking large toxin peptides to ion channel homology models, as exemplified by the α-GID:α4β2 nAChR complex, and is extendable to other toxin peptides and ion channels. ToxDock is freely available at rosie.rosettacommons.org/tox_dock.
Collapse
Affiliation(s)
- Abba E Leffler
- Neuroscience Graduate Program, Sackler Institute of Graduate Biomedical Sciences, New York University School of Medicine, New York, NY 10016
| | - Alexander Kuryatov
- Department of Neuroscience, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA 19104
| | - Henry A Zebroski
- Proteomics Resource Center, The Rockefeller University, New York, NY 10065
| | - Susan R Powell
- Proteomics Resource Center, The Rockefeller University, New York, NY 10065
| | - Petr Filipenko
- Department of Chemistry, Belfer Research Center-Hunter College, New York, NY 10021
- Division of Invertebrate Zoology, The American Museum of Natural History, New York, NY 10024
- Department of Biochemistry, Weill Cornell Medical College, Cornell University, New York, NY 10021
| | - Adel K Hussein
- Department of Chemistry, College of Staten Island, Staten Island, NY 10314
- Program in Biochemistry, The Graduate Center, City University of New York, New York, NY 10016
| | - Juliette Gorson
- Department of Chemistry, Belfer Research Center-Hunter College, New York, NY 10021
- Division of Invertebrate Zoology, The American Museum of Natural History, New York, NY 10024
- Department of Biochemistry, Weill Cornell Medical College, Cornell University, New York, NY 10021
- Program in Biochemistry, The Graduate Center, City University of New York, New York, NY 10016
| | - Anna Heizmann
- Walther Straub Institute of Pharmacology and Toxicology, LMU Munich, 80336 Munich, Germany
| | - Sergey Lyskov
- Department of Chemical and Biomolecular Engineering, Johns Hopkins University, Baltimore, MD 21218
| | - Richard W Tsien
- NYU Neuroscience Institute and the Department of Neuroscience and Physiology, New York University School of Medicine, New York, NY 10016;
| | - Sébastien F Poget
- Department of Chemistry, College of Staten Island, Staten Island, NY 10314
- Program in Biochemistry, The Graduate Center, City University of New York, New York, NY 10016
| | - Annette Nicke
- Walther Straub Institute of Pharmacology and Toxicology, LMU Munich, 80336 Munich, Germany
| | - Jon Lindstrom
- Department of Neuroscience, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA 19104
| | - Bernardo Rudy
- NYU Neuroscience Institute and the Department of Neuroscience and Physiology, New York University School of Medicine, New York, NY 10016
| | - Richard Bonneau
- Department of Biology, New York University, New York, NY 10003
- Courant Institute of Mathematical Sciences, New York University, New York, NY 10012
- Center for Computational Biology, Simons Foundation, New York, NY 10010
| | - Mandë Holford
- Department of Chemistry, Belfer Research Center-Hunter College, New York, NY 10021;
- Division of Invertebrate Zoology, The American Museum of Natural History, New York, NY 10024
- Department of Biochemistry, Weill Cornell Medical College, Cornell University, New York, NY 10021
- Program in Biochemistry, The Graduate Center, City University of New York, New York, NY 10016
| |
Collapse
|
26
|
Tabassum N, Tae HS, Jia X, Kaas Q, Jiang T, Adams DJ, Yu R. Role of Cys I-Cys III Disulfide Bond on the Structure and Activity of α-Conotoxins at Human Neuronal Nicotinic Acetylcholine Receptors. ACS OMEGA 2017; 2:4621-4631. [PMID: 30023726 PMCID: PMC6044955 DOI: 10.1021/acsomega.7b00639] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 05/23/2017] [Accepted: 08/04/2017] [Indexed: 06/08/2023]
Abstract
α-Conotoxins preferentially antagonize muscle and neuronal nicotinic acetylcholine receptors (nAChRs). Native α-conotoxins have two disulfide links, CI-CIII and CII-CIV, and owing to the inherent properties of disulfide bonds, α-conotoxins have been systematically engineered to improve their chemical and biological properties. In this study, we explored the possibility of simplifying the disulfide framework of α-conotoxins Vc1.1, BuIA, ImI, and AuIB, by introducing [C2H,C8F] modification to the CI-CIII bond. We therefore explored the possibility of using hydrophobic packing of standard amino acid side chains to replace disulfide bonds as an alternative strategy to nonnatural amino acid cross-links. The impact of CI-CIII disulfide bond replacement on the conformation of the α-conotoxins was investigated using molecular dynamics (MD) simulations and nuclear magnetic resonance chemical shift index study. Two-electrode voltage clamp techniques and MD simulations were used to study the impact of disulfide bond deletion on the activities of the peptides at human neuronal nAChRs. All disulfide-deleted variants except ImI[C2H,C8F] had reduced potency for inhibiting nAChRs. Our results suggest that the CI-CIII disulfide bond is important to stabilize the secondary structure of α-conotoxins as well as their interaction with neuronal nAChR targets. Results from this study enrich our understanding of the function of the CI-CIII disulfide bond and are useful in guiding future structural engineering of the α-conotoxins.
Collapse
Affiliation(s)
- Nargis Tabassum
- Key
Laboratory of Marine Drugs, Chinese Ministry of Education, School
of Medicine and Pharmacy, Ocean University
of China, Qingdao 266003, China
- Laboratory
for Marine Drugs and Bioproducts of Qingdao National Laboratory for
Marine Science and Technology, Qingdao 266003, China
| | - Han-Shen Tae
- Illawarra
Health and Medical Research Institute (IHMRI), University of Wollongong, Wollongong, New South Wales 2522, Australia
| | - Xinying Jia
- The Centre for Advanced Imaging and Institute for Molecular Bioscience, The University of Queensland, Brisbane, Queensland 4072 Australia
| | - Quentin Kaas
- The Centre for Advanced Imaging and Institute for Molecular Bioscience, The University of Queensland, Brisbane, Queensland 4072 Australia
| | - Tao Jiang
- Key
Laboratory of Marine Drugs, Chinese Ministry of Education, School
of Medicine and Pharmacy, Ocean University
of China, Qingdao 266003, China
| | - David J. Adams
- Illawarra
Health and Medical Research Institute (IHMRI), University of Wollongong, Wollongong, New South Wales 2522, Australia
| | - Rilei Yu
- Key
Laboratory of Marine Drugs, Chinese Ministry of Education, School
of Medicine and Pharmacy, Ocean University
of China, Qingdao 266003, China
- Laboratory
for Marine Drugs and Bioproducts of Qingdao National Laboratory for
Marine Science and Technology, Qingdao 266003, China
| |
Collapse
|
27
|
Hone AJ, Servent D, McIntosh JM. α9-containing nicotinic acetylcholine receptors and the modulation of pain. Br J Pharmacol 2017; 175:1915-1927. [PMID: 28662295 DOI: 10.1111/bph.13931] [Citation(s) in RCA: 62] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2017] [Revised: 06/09/2017] [Accepted: 06/13/2017] [Indexed: 01/01/2023] Open
Abstract
Neuropathic pain is a complex and debilitating syndrome for which there are few effective pharmacological treatments. Opioid-based medications are initially effective for acute pain, but tolerance to their analgesic effects quickly develops, and long-term use often leads to physical dependence and addiction. Furthermore, neuropathic pain is generally resistant to non-steroidal anti-inflammatory drugs. Other classes of medications including antidepressants, antiepileptics and voltage-gated calcium channel inhibitors are only partially effective in most patients, may be associated with significant side effects and have few disease-modifying effects on the underlying pathology. Medications that act through new mechanisms of action, and particularly ones that have disease-modifying properties, would be highly desirable. In the last decade, a potential new target for the treatment of neuropathic pain has emerged: the α9-containing nicotinic acetylcholine receptor (nAChR). Recent studies indicate that antagonists of α9-containing nAChRs are analgesic in animal models of neuropathic pain. These nerve injury models include chronic constriction injury, partial sciatic nerve ligation, streptozotocin-induced diabetic neuropathy and chemotherapeutic-induced neuropathy. This review details the history and state of the field regarding the role that α9-containing nAChRs may play in neuropathic pain. An alternative hypothesis that α-conotoxins exert their therapeutic effect through blocking N-type calcium channels via activation of GABAB receptors is also reviewed. Understanding how antagonists of α9-containing nAChRs exert their therapeutic effects may ultimately result in the development of medications that not only treat but also prevent the development of neuropathic pain states. LINKED ARTICLES This article is part of a themed section on Nicotinic Acetylcholine Receptors. To view the other articles in this section visit http://onlinelibrary.wiley.com/doi/10.1111/bph.v175.11/issuetoc.
Collapse
Affiliation(s)
- Arik J Hone
- Department of Biology, University of Utah, Salt Lake City, UT, USA
| | - Denis Servent
- Service d'Ingénierie Moléculaire des Protéines (SIMOPRO), IBITECS, CEA, Université Paris-Saclay, Gif-sur-Yvette, France
| | - J Michael McIntosh
- Department of Biology, University of Utah, Salt Lake City, UT, USA.,George E. Whalen Veterans Affairs Medical Center, Salt Lake City, UT, USA.,Department of Psychiatry, University of Utah, Salt Lake City, UT, USA
| |
Collapse
|
28
|
Christensen SB, Hone AJ, Roux I, Kniazeff J, Pin JP, Upert G, Servent D, Glowatzki E, McIntosh JM. RgIA4 Potently Blocks Mouse α9α10 nAChRs and Provides Long Lasting Protection against Oxaliplatin-Induced Cold Allodynia. Front Cell Neurosci 2017; 11:219. [PMID: 28785206 PMCID: PMC5519620 DOI: 10.3389/fncel.2017.00219] [Citation(s) in RCA: 50] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2017] [Accepted: 07/06/2017] [Indexed: 01/12/2023] Open
Abstract
Transcripts for α9 and α10 nicotinic acetylcholine receptor (nAChR) subunits are found in diverse tissues. The function of α9α10 nAChRs is best known in mechanosensory cochlear hair cells, but elsewhere their roles are less well-understood. α9α10 nAChRs have been implicated as analgesic targets and α-conotoxins that block α9α10 nAChRs produce analgesia. However, some of these peptides show large potency differences between species. Additionally several studies have indicated that these conotoxins may also activate GABAB receptors (GABABRs). To further address these issues, we cloned the cDNAs of mouse α9 and α10 nAChR subunits. When heterologously expressed in Xenopus oocytes, the resulting α9α10 nAChRs had the expected pharmacology of being activated by acetylcholine and choline but not by nicotine. A conotoxin analog, RgIA4, potently, and selectively blocked mouse α9α10 nAChRs with low nanomolar affinity indicating that RgIA4 may be effectively used to study murine α9α10 nAChR function. Previous reports indicated that RgIA4 attenuates chemotherapy-induced cold allodynia. Here we demonstrate that RgIA4 analgesic effects following oxaliplatin treatment are sustained for 21 days after last RgIA4 administration indicating that RgIA4 may provide enduring protection against nerve damage. RgIA4 lacks activity at GABAB receptors; a bioluminescence resonance energy transfer assay was used to demonstrate that two other analgesic α-conotoxins, Vc1.1 and AuIB, also do not activate GABABRs expressed in HEK cells. Together these findings further support the targeting of α9α10 nAChRs in the treatment of pain.
Collapse
Affiliation(s)
- Sean B Christensen
- Department of Biology, University of UtahSalt Lake City, UT, United States
| | - Arik J Hone
- Department of Biology, University of UtahSalt Lake City, UT, United States
| | - Isabelle Roux
- Department of Otolaryngology, Head and Neck Surgery, The Center for Hearing and Balance and the Center for Sensory Biology, The Johns Hopkins University School of MedicineBaltimore, MD, United States
| | - Julie Kniazeff
- IGF, Centre National de la Recherche Scientifique, Institut National de la Santé et de la Recherche Médicale, Université MontpellierMontpellier, France
| | - Jean-Philippe Pin
- IGF, Centre National de la Recherche Scientifique, Institut National de la Santé et de la Recherche Médicale, Université MontpellierMontpellier, France
| | - Grégory Upert
- Service d'Ingénierie Moléculaire des Protéines, CEA, Université Paris-SaclayGif-sur-Yvette, France
| | - Denis Servent
- Service d'Ingénierie Moléculaire des Protéines, CEA, Université Paris-SaclayGif-sur-Yvette, France
| | - Elisabeth Glowatzki
- Department of Otolaryngology, Head and Neck Surgery, The Center for Hearing and Balance and the Center for Sensory Biology, The Johns Hopkins University School of MedicineBaltimore, MD, United States.,Department of Neuroscience, The Johns Hopkins University School of MedicineBaltimore, MD, United States
| | - J Michael McIntosh
- Department of Biology, University of UtahSalt Lake City, UT, United States.,George E. Whalen Veterans Affairs Medical CenterSalt Lake City, UT, United States.,Department of Psychiatry, University of UtahSalt Lake City, UT, United States
| |
Collapse
|
29
|
Dutertre S, Nicke A, Tsetlin VI. Nicotinic acetylcholine receptor inhibitors derived from snake and snail venoms. Neuropharmacology 2017. [PMID: 28623170 DOI: 10.1016/j.neuropharm.2017.06.011] [Citation(s) in RCA: 54] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
The nicotinic acetylcholine receptor (nAChR) represents the prototype of ligand-gated ion channels. It is vital for neuromuscular transmission and an important regulator of neurotransmission. A variety of toxic compounds derived from diverse species target this receptor and have been of elemental importance in basic and applied research. They enabled milestone discoveries in pharmacology and biochemistry ranging from the original formulation of the receptor concept, the first isolation and structural analysis of a receptor protein (the nAChR) to the identification, localization, and differentiation of its diverse subtypes and their validation as a target for therapeutic intervention. Among the venom-derived compounds, α-neurotoxins and α-conotoxins provide the largest families and still represent indispensable pharmacological tools. Application of modified α-neurotoxins provided substantial structural and functional details of the nAChR long before high resolution structures were available. α-bungarotoxin represents not only a standard pharmacological tool and label in nAChR research but also for unrelated proteins tagged with a minimal α-bungarotoxin binding motif. A major advantage of α-conotoxins is their smaller size, as well as superior selectivity for diverse nAChR subtypes that allows their development into ligands with optimized pharmacological and chemical properties and potentially novel drugs. In the following, these two groups of nAChR antagonists will be described focusing on their respective roles in the structural and functional characterization of nAChRs and their development into research tools. In addition, we provide a comparative overview of the diverse α-conotoxin selectivities that can serve as a practical guide for both structure activity studies and subtype classification. This article is part of the Special Issue entitled 'Venom-derived Peptides as Pharmacological Tools.'
Collapse
Affiliation(s)
- Sébastien Dutertre
- Institut des Biomolécules Max Mousseron, UMR 5247, Université Montpellier - CNRS, Place Eugène Bataillon, 34095 Montpellier Cedex 5, France
| | - Annette Nicke
- Walther Straub Institute for Pharmacology and Toxicology, Ludwig-Maximilians-Universität, Nußbaumstr. 26, 80336 Munich, Germany.
| | - Victor I Tsetlin
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, Miklukho-Maklaya str.16/10, Moscow 117999, Russian Federation
| |
Collapse
|
30
|
Cuny H, Yu R, Tae HS, Kompella SN, Adams DJ. α-Conotoxins active at α3-containing nicotinic acetylcholine receptors and their molecular determinants for selective inhibition. Br J Pharmacol 2017; 175:1855-1868. [PMID: 28477355 DOI: 10.1111/bph.13852] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2017] [Revised: 04/13/2017] [Accepted: 04/24/2017] [Indexed: 01/22/2023] Open
Abstract
Neuronal α3-containing nicotinic acetylcholine receptors (nAChRs) in the peripheral nervous system (PNS) and non-neuronal tissues are implicated in a number of severe disease conditions ranging from cancer to cardiovascular diseases and chronic pain. However, despite the physiological characterization of mouse models and cell lines, the precise pathophysiology of nAChRs outside the CNS remains not well understood, in part because there is a lack of subtype-selective antagonists. α-Conotoxins isolated from cone snail venom exhibit characteristic individual selectivity profiles for nAChRs and, therefore, are excellent tools to study the determinants for nAChR-antagonist interactions. Given that human α3β4 subtype selective α-conotoxins are scarce and this is a major nAChR subtype in the PNS, the design of new peptides targeting this nAChR subtype is desirable. Recent studies using α-conotoxins RegIIA and AuIB, in combination with nAChR site-directed mutagenesis and computational modelling, have shed light onto specific nAChR residues, which determine the selectivity of the α-conotoxins for the human α3β2 and α3β4 subtypes. Publications describing the selectivity profile and binding sites of other α-conotoxins confirm that subtype-selective nAChR antagonists often work through common mechanisms by interacting with the same structural components and sites on the receptor. LINKED ARTICLES This article is part of a themed section on Nicotinic Acetylcholine Receptors. To view the other articles in this section visit http://onlinelibrary.wiley.com/doi/10.1111/bph.v175.11/issuetoc.
Collapse
Affiliation(s)
- Hartmut Cuny
- Illawarra Health and Medical Research Institute (IHMRI), University of Wollongong, Wollongong, NSW, Australia.,Victor Chang Cardiac Research Institute, Developmental and Stem Cell Biology Division, Sydney, NSW, Australia
| | - Rilei Yu
- Key Laboratory of Marine Drugs, Chinese Ministry of Education, School of Medicine and Pharmacy, Ocean University of China, Qingdao, China.,Laboratory for Marine Drugs and Bioproducts of Qingdao National Laboratory for Marine Science and Technology, Qingdao, China
| | - Han-Shen Tae
- Illawarra Health and Medical Research Institute (IHMRI), University of Wollongong, Wollongong, NSW, Australia
| | - Shiva N Kompella
- Faculty of Pharmacy, University of Sydney, Sydney, NSW, Australia
| | - David J Adams
- Illawarra Health and Medical Research Institute (IHMRI), University of Wollongong, Wollongong, NSW, Australia
| |
Collapse
|
31
|
Castro J, Harrington AM, Garcia-Caraballo S, Maddern J, Grundy L, Zhang J, Page G, Miller PE, Craik DJ, Adams DJ, Brierley SM. α-Conotoxin Vc1.1 inhibits human dorsal root ganglion neuroexcitability and mouse colonic nociception via GABA B receptors. Gut 2017; 66:1083-1094. [PMID: 26887818 PMCID: PMC5532460 DOI: 10.1136/gutjnl-2015-310971] [Citation(s) in RCA: 72] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/27/2015] [Revised: 12/22/2015] [Accepted: 01/14/2016] [Indexed: 01/29/2023]
Abstract
OBJECTIVE α-Conotoxin Vc1.1 is a small disulfide-bonded peptide from the venom of the marine cone snail Conus victoriae. Vc1.1 has antinociceptive actions in animal models of neuropathic pain, but its applicability to inhibiting human dorsal root ganglion (DRG) neuroexcitability and reducing chronic visceral pain (CVP) is unknown. DESIGN We determined the inhibitory actions of Vc1.1 on human DRG neurons and on mouse colonic sensory afferents in healthy and chronic visceral hypersensitivity (CVH) states. In mice, visceral nociception was assessed by neuronal activation within the spinal cord in response to noxious colorectal distension (CRD). Quantitative-reverse-transcription-PCR, single-cell-reverse-transcription-PCR and immunohistochemistry determined γ-aminobutyric acid receptor B (GABABR) and voltage-gated calcium channel (CaV2.2, CaV2.3) expression in human and mouse DRG neurons. RESULTS Vc1.1 reduced the excitability of human DRG neurons, whereas a synthetic Vc1.1 analogue that is inactive at GABABR did not. Human DRG neurons expressed GABABR and its downstream effector channels CaV2.2 and CaV2.3. Mouse colonic DRG neurons exhibited high GABABR, CaV2.2 and CaV2.3 expression, with upregulation of the CaV2.2 exon-37a variant during CVH. Vc1.1 inhibited mouse colonic afferents ex vivo and nociceptive signalling of noxious CRD into the spinal cord in vivo, with greatest efficacy observed during CVH. A selective GABABR antagonist prevented Vc1.1-induced inhibition, whereas blocking both CaV2.2 and CaV2.3 caused inhibition comparable with Vc1.1 alone. CONCLUSIONS Vc1.1-mediated activation of GABABR is a novel mechanism for reducing the excitability of human DRG neurons. Vc1.1-induced activation of GABABR on the peripheral endings of colonic afferents reduces nociceptive signalling. The enhanced antinociceptive actions of Vc1.1 during CVH suggest it is a novel candidate for the treatment for CVP.
Collapse
Affiliation(s)
- Joel Castro
- Visceral Pain Group, Centre for Nutrition and Gastrointestinal Diseases, Discipline of Medicine, Faculty of Health Sciences, The University of Adelaide, South Australian Health and Medical Research Institute (SAHMRI), Adelaide, South Australia, Australia
| | - Andrea M Harrington
- Visceral Pain Group, Centre for Nutrition and Gastrointestinal Diseases, Discipline of Medicine, Faculty of Health Sciences, The University of Adelaide, South Australian Health and Medical Research Institute (SAHMRI), Adelaide, South Australia, Australia
| | - Sonia Garcia-Caraballo
- Visceral Pain Group, Centre for Nutrition and Gastrointestinal Diseases, Discipline of Medicine, Faculty of Health Sciences, The University of Adelaide, South Australian Health and Medical Research Institute (SAHMRI), Adelaide, South Australia, Australia
| | - Jessica Maddern
- Visceral Pain Group, Centre for Nutrition and Gastrointestinal Diseases, Discipline of Medicine, Faculty of Health Sciences, The University of Adelaide, South Australian Health and Medical Research Institute (SAHMRI), Adelaide, South Australia, Australia
| | - Luke Grundy
- Visceral Pain Group, Centre for Nutrition and Gastrointestinal Diseases, Discipline of Medicine, Faculty of Health Sciences, The University of Adelaide, South Australian Health and Medical Research Institute (SAHMRI), Adelaide, South Australia, Australia
| | | | - Guy Page
- Anabios, San Diego, California, USA
| | | | - David J Craik
- Division of Chemistry and Structural Biology, Institute for Molecular Bioscience, University of Queensland, Brisbane, Queensland, Australia
| | - David J Adams
- Illawarra Health & Medical Research Institute (IHMRI), University of Wollongong, Wollongong, NSW, Australia
| | - Stuart M Brierley
- Visceral Pain Group, Centre for Nutrition and Gastrointestinal Diseases, Discipline of Medicine, Faculty of Health Sciences, The University of Adelaide, South Australian Health and Medical Research Institute (SAHMRI), Adelaide, South Australia, Australia
| |
Collapse
|
32
|
Sadeghi M, McArthur JR, Finol-Urdaneta RK, Adams DJ. Analgesic conopeptides targeting G protein-coupled receptors reduce excitability of sensory neurons. Neuropharmacology 2017; 127:116-123. [PMID: 28533165 DOI: 10.1016/j.neuropharm.2017.05.020] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2017] [Revised: 05/16/2017] [Accepted: 05/18/2017] [Indexed: 01/28/2023]
Abstract
Conotoxins (conopeptides) are a diverse group of peptides isolated from the venom of marine cone snails. Conus peptides modulate pain by interacting with voltage-gated ion channels and G protein-coupled receptors (GPCRs). Opiate drugs targeting GPCRs have long been used, nonetheless, many undesirable side effects associated with opiates have been observed including addiction. Consequently, alternative avenues to pain management are a largely unmet need. It has been shown that various voltage-gated calcium channels (VGCCs) respond to GPCR modulation. Thus, regulation of VGCCs by GPCRs has become a valuable alternative in the management of pain. In this review, we focus on analgesic conotoxins that exert their effects via GPCR-mediated inhibition of ion channels involved in nociception and pain transmission. Specifically, α-conotoxin Vc1.1 activation of GABAB receptors and inhibition of voltage-gated calcium channels as a novel mechanism for reducing the excitability of dorsal root ganglion neurons is described. Vc1.1 and other α-conotoxins have been shown to be analgesic in different animal models of chronic pain. This review will outline the functional effects of conopeptide modulation of GPCRs and how their signalling is translated to downstream components of the pain pathways. Where available we present the proposed signalling mechanisms that couples metabotropic receptor activation to their downstream effectors to produce analgesia. This article is part of the Special Issue entitled 'Venom-derived Peptides as Pharmacological Tools.'
Collapse
Affiliation(s)
- Mahsa Sadeghi
- Illawarra Health and Medical Research Institute, University of Wollongong, Wollongong, New South Wales, 2522, Australia
| | - Jeffrey R McArthur
- Illawarra Health and Medical Research Institute, University of Wollongong, Wollongong, New South Wales, 2522, Australia
| | - Rocio K Finol-Urdaneta
- Illawarra Health and Medical Research Institute, University of Wollongong, Wollongong, New South Wales, 2522, Australia
| | - David J Adams
- Illawarra Health and Medical Research Institute, University of Wollongong, Wollongong, New South Wales, 2522, Australia.
| |
Collapse
|
33
|
Molecular Engineering of Conus Peptides as Therapeutic Leads. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2017; 1030:229-254. [DOI: 10.1007/978-3-319-66095-0_10] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
|
34
|
Carstens BB, Berecki G, Daniel JT, Lee HS, Jackson KAV, Tae H, Sadeghi M, Castro J, O'Donnell T, Deiteren A, Brierley SM, Craik DJ, Adams DJ, Clark RJ. Structure–Activity Studies of Cysteine‐Rich α‐Conotoxins that Inhibit High‐Voltage‐Activated Calcium Channels via GABA
B
Receptor Activation Reveal a Minimal Functional Motif. Angew Chem Int Ed Engl 2016. [DOI: 10.1002/ange.201600297] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Affiliation(s)
- Bodil B. Carstens
- Institute for Molecular Biosciences The University of Queensland Brisbane Qld 4072 Australia
| | - Géza Berecki
- Health Innovations Research Institute RMIT University Melbourne Vic 3083 Australia
| | - James T. Daniel
- School of Biomedical Science The University of Queensland Brisbane Qld 4072 Australia
| | - Han Siean Lee
- School of Biomedical Science The University of Queensland Brisbane Qld 4072 Australia
| | - Kathryn A. V. Jackson
- School of Biomedical Science The University of Queensland Brisbane Qld 4072 Australia
| | - Han‐Shen Tae
- Health Innovations Research Institute RMIT University Melbourne Vic 3083 Australia
- Illawarra Health and Medical Research Institute (IHMRI) University of Wollongong Wollongong NSW 2522 Australia
| | - Mahsa Sadeghi
- Health Innovations Research Institute RMIT University Melbourne Vic 3083 Australia
- Illawarra Health and Medical Research Institute (IHMRI) University of Wollongong Wollongong NSW 2522 Australia
| | - Joel Castro
- Visceral Pain Group, Centre for Nutrition and Gastrointestinal Diseases Discipline of Medicine The University of Adelaide South Australian Health and Medical Research Institute (SAHMRI) Adelaide SA 5000 Australia
| | - Tracy O'Donnell
- Visceral Pain Group, Centre for Nutrition and Gastrointestinal Diseases Discipline of Medicine The University of Adelaide South Australian Health and Medical Research Institute (SAHMRI) Adelaide SA 5000 Australia
| | - Annemie Deiteren
- Visceral Pain Group, Centre for Nutrition and Gastrointestinal Diseases Discipline of Medicine The University of Adelaide South Australian Health and Medical Research Institute (SAHMRI) Adelaide SA 5000 Australia
| | - Stuart M. Brierley
- Visceral Pain Group, Centre for Nutrition and Gastrointestinal Diseases Discipline of Medicine The University of Adelaide South Australian Health and Medical Research Institute (SAHMRI) Adelaide SA 5000 Australia
| | - David J. Craik
- Institute for Molecular Biosciences The University of Queensland Brisbane Qld 4072 Australia
| | - David J. Adams
- Health Innovations Research Institute RMIT University Melbourne Vic 3083 Australia
- Illawarra Health and Medical Research Institute (IHMRI) University of Wollongong Wollongong NSW 2522 Australia
| | - Richard J. Clark
- School of Biomedical Science The University of Queensland Brisbane Qld 4072 Australia
| |
Collapse
|
35
|
Carstens BB, Berecki G, Daniel JT, Lee HS, Jackson KAV, Tae H, Sadeghi M, Castro J, O'Donnell T, Deiteren A, Brierley SM, Craik DJ, Adams DJ, Clark RJ. Structure–Activity Studies of Cysteine‐Rich α‐Conotoxins that Inhibit High‐Voltage‐Activated Calcium Channels via GABA
B
Receptor Activation Reveal a Minimal Functional Motif. Angew Chem Int Ed Engl 2016; 55:4692-6. [DOI: 10.1002/anie.201600297] [Citation(s) in RCA: 45] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2016] [Indexed: 01/29/2023]
Affiliation(s)
- Bodil B. Carstens
- Institute for Molecular Biosciences The University of Queensland Brisbane Qld 4072 Australia
| | - Géza Berecki
- Health Innovations Research Institute RMIT University Melbourne Vic 3083 Australia
| | - James T. Daniel
- School of Biomedical Science The University of Queensland Brisbane Qld 4072 Australia
| | - Han Siean Lee
- School of Biomedical Science The University of Queensland Brisbane Qld 4072 Australia
| | - Kathryn A. V. Jackson
- School of Biomedical Science The University of Queensland Brisbane Qld 4072 Australia
| | - Han‐Shen Tae
- Health Innovations Research Institute RMIT University Melbourne Vic 3083 Australia
- Illawarra Health and Medical Research Institute (IHMRI) University of Wollongong Wollongong NSW 2522 Australia
| | - Mahsa Sadeghi
- Health Innovations Research Institute RMIT University Melbourne Vic 3083 Australia
- Illawarra Health and Medical Research Institute (IHMRI) University of Wollongong Wollongong NSW 2522 Australia
| | - Joel Castro
- Visceral Pain Group, Centre for Nutrition and Gastrointestinal Diseases Discipline of Medicine The University of Adelaide South Australian Health and Medical Research Institute (SAHMRI) Adelaide SA 5000 Australia
| | - Tracy O'Donnell
- Visceral Pain Group, Centre for Nutrition and Gastrointestinal Diseases Discipline of Medicine The University of Adelaide South Australian Health and Medical Research Institute (SAHMRI) Adelaide SA 5000 Australia
| | - Annemie Deiteren
- Visceral Pain Group, Centre for Nutrition and Gastrointestinal Diseases Discipline of Medicine The University of Adelaide South Australian Health and Medical Research Institute (SAHMRI) Adelaide SA 5000 Australia
| | - Stuart M. Brierley
- Visceral Pain Group, Centre for Nutrition and Gastrointestinal Diseases Discipline of Medicine The University of Adelaide South Australian Health and Medical Research Institute (SAHMRI) Adelaide SA 5000 Australia
| | - David J. Craik
- Institute for Molecular Biosciences The University of Queensland Brisbane Qld 4072 Australia
| | - David J. Adams
- Health Innovations Research Institute RMIT University Melbourne Vic 3083 Australia
- Illawarra Health and Medical Research Institute (IHMRI) University of Wollongong Wollongong NSW 2522 Australia
| | - Richard J. Clark
- School of Biomedical Science The University of Queensland Brisbane Qld 4072 Australia
| |
Collapse
|
36
|
Zamponi GW, Han C, Waxman SG. Voltage-Gated Ion Channels as Molecular Targets for Pain. Transl Neurosci 2016. [DOI: 10.1007/978-1-4899-7654-3_22] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/22/2022] Open
|
37
|
Li L, Liu N, Ding R, Wang S, Liu Z, Li H, Zheng X, Dai Q. A novel 4/6-type alpha-conotoxin ViIA selectively inhibits nAchR α3β2 subtype. Acta Biochim Biophys Sin (Shanghai) 2015; 47:1023-8. [PMID: 26511093 DOI: 10.1093/abbs/gmv105] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2015] [Accepted: 08/24/2015] [Indexed: 01/01/2023] Open
Abstract
Conotoxins (CTxs) are typically small peptides composed of 12-50 amino acid residues with 2-5 disulfide bridges. Most of them potently and selectively target a wide variety of ion channels and membrane receptors. They are highly valued as neuropharmacological probes and in pharmaceutical development. In this work, a novel α4/6-CTx named ViIA (RDCCSNPPCAHNNPDC-NH2) was identified from a cDNA library of the venom ducts of Conus virgo (C. virgo). ViIA was then synthesized chemically and its disulfide connectivity was identified as 'C(1)-C(3), C(2)-C(4)'. Its molecular targets were further assessed using two-electrode voltage clamping. The results indicated that ViIA selectively inhibited nicotinic acetylcholine receptor (nAChR) α3β2 subtype with an IC50 of 845.5 nM, but did not target dorsal root ganglion sodium (Na(+))-, potassium (K(+))- or calcium (Ca(2+))-ion channels. Further structure-activity relationship analysis demonstrated that Arg(1) and His(11) but not Asp(2) were the functional residues. To the best of our knowledge, ViIA is the first 4/6 α-CTx that selectively inhibits nAChR α3β2 subtype. This finding expands the knowledge of targets of α4/6-family CTxs.
Collapse
Affiliation(s)
- Liang Li
- Institute of Pharmacy and Pharmacology, University of South China, Hengyang 421001, China Beijing Institute of Biotechnology, Beijing 100071, China
| | - Na Liu
- Beijing Institute of Biotechnology, Beijing 100071, China
| | - Rong Ding
- Beijing Institute of Biotechnology, Beijing 100071, China
| | - Shuo Wang
- Beijing Institute of Biotechnology, Beijing 100071, China
| | - Zhuguo Liu
- Beijing Institute of Biotechnology, Beijing 100071, China
| | - Haiying Li
- Beijing Institute of Biotechnology, Beijing 100071, China
| | - Xing Zheng
- Institute of Pharmacy and Pharmacology, University of South China, Hengyang 421001, China
| | - Qiuyun Dai
- Beijing Institute of Biotechnology, Beijing 100071, China
| |
Collapse
|
38
|
Mohammadi SA, Christie MJ. Conotoxin Interactions with α9α10-nAChRs: Is the α9α10-Nicotinic Acetylcholine Receptor an Important Therapeutic Target for Pain Management? Toxins (Basel) 2015; 7:3916-32. [PMID: 26426047 PMCID: PMC4626711 DOI: 10.3390/toxins7103916] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2015] [Revised: 09/14/2015] [Accepted: 09/18/2015] [Indexed: 11/16/2022] Open
Abstract
The α9α10-nicotinic acetylcholine receptor (nAChR) has been implicated in pain and has been proposed to be a novel target for analgesics. However, the evidence to support the involvement of the α9α10-nAChR in pain is conflicted. This receptor was first implicated in pain with the characterisation of conotoxin Vc1.1, which is highly selective for α9α10-nAChRs and is an efficacious analgesic in chronic pain models with restorative capacities and no reported side effects. Numerous other analgesic conotoxin and non-conotoxin molecules have been subsequently characterised that also inhibit α9α10-nAChRs. However, there is evidence that α9α10-nAChR inhibition is neither necessary nor sufficient for analgesia. α9α10-nAChR-inhibiting analogues of Vc1.1 have no analgesic effects. Genetically-modified α9-nAChR knockout mice have a phenotype that is markedly different from the analgesic profile of Vc1.1 and similar conotoxins, suggesting that the conotoxin effects are largely independent of α9α10-nAChRs. Furthermore, an alternative mechanism of analgesia by Vc1.1 and other similar conotoxins involving non-canonical coupling of GABAB receptors to voltage-gated calcium channels is known. Additional incongruities regarding α9α10-nAChRs in analgesia are discussed. A more comprehensive characterisation of the role of α9α10-nAChRs in pain is crucial for understanding the analgesic action of conotoxins and for improved drug design.
Collapse
Affiliation(s)
- Sarasa A Mohammadi
- Discipline of Pharmacology, the University of Sydney, Sydney, NSW 2006, Australia.
| | - MacDonald J Christie
- Discipline of Pharmacology, the University of Sydney, Sydney, NSW 2006, Australia.
| |
Collapse
|
39
|
Wang S, Zhao C, Liu Z, Wang X, Liu N, Du W, Dai Q. Structural and Functional Characterization of a Novel α-Conotoxin Mr1.7 from Conus marmoreus Targeting Neuronal nAChR α3β2, α9α10 and α6/α3β2β3 Subtypes. Mar Drugs 2015; 13:3259-75. [PMID: 26023835 PMCID: PMC4483627 DOI: 10.3390/md13063259] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2015] [Accepted: 05/11/2015] [Indexed: 12/26/2022] Open
Abstract
In the present study, we synthesized and, structurally and functionally characterized a novel α4/7-conotoxin Mr1.7 (PECCTHPACHVSHPELC-NH2), which was previously identified by cDNA libraries from Conus marmoreus in our lab. The NMR solution structure showed that Mr1.7 contained a 310-helix from residues Pro7 to His10 and a type I β-turn from residues Pro14 to Cys17. Electrophysiological results showed that Mr1.7 selectively inhibited the α3β2, α9α10 and α6/α3β2β3 neuronal nicotinic acetylcholine receptors (nAChRs) with an IC50 of 53.1 nM, 185.7 nM and 284.2 nM, respectively, but showed no inhibitory activity on other nAChR subtypes. Further structure-activity studies of Mr1.7 demonstrated that the PE residues at the N-terminal sequence of Mr1.7 were important for modulating its selectivity, and the replacement of Glu2 by Ala resulted in a significant increase in potency and selectivity to the α3β2 nAChR. Furthermore, the substitution of Ser12 with Asn in the loop2 significantly increased the binding of Mr1.7 to α3β2, α3β4, α2β4 and α7 nAChR subtypes. Taken together, this work expanded our knowledge of selectivity and provided a new way to improve the potency and selectivity of inhibitors for nAChR subtypes.
Collapse
Affiliation(s)
- Shuo Wang
- Beijing Institute of Biotechnology, Beijing 100071, China.
| | - Cong Zhao
- Department of Chemistry, Renmin University of China, Beijing 100872, China.
| | - Zhuguo Liu
- Beijing Institute of Biotechnology, Beijing 100071, China.
| | - Xuesong Wang
- Department of Chemistry, Renmin University of China, Beijing 100872, China.
| | - Na Liu
- Beijing Institute of Biotechnology, Beijing 100071, China.
| | - Weihong Du
- Department of Chemistry, Renmin University of China, Beijing 100872, China.
| | - Qiuyun Dai
- Beijing Institute of Biotechnology, Beijing 100071, China.
| |
Collapse
|
40
|
Abstract
Chronic pain is very difficult to treat. Thus, novel analgesics are a critical area of research. Strong pre-clinical evidence supports the analgesic effects of α-conopeptides, Vc1.1 and RgIA, which block α9α10 nicotinic acetylcholine receptors (nAChRs). However, the analgesic mechanism is controversial. Some evidence supports the block of α9α10 nAChRs as an analgesic mechanism, while other evidence supports the inhibition of N-type CaV (CaV2.2) current via activation of GABAB receptors. Here we reassess the effect of Vc1.1 and RgIA on CaV current in rat sensory neurons. Unlike the previous findings, we found highly variable effects among individual sensory neurons, but on average only minimal inhibition induced by Vc1.1, and no significant effect on the current by RgIA. We also investigated the potential involvement of GABAB receptors in the Vc1.1 induced inhibition, and found no correlation between the size of CaV current inhibition induced by baclofen (GABAB agonist) vs. that induced by Vc1.1. Thus, GABAB receptors are unlikely to mediate the Vc1.1 induced CaV current inhibition. Based on the present findings, CaV current inhibition in dorsal root ganglia is unlikely to be the predominant mechanism by which either Vc1.1 or RgIA induce analgesia. SIGNIFICANCE STATEMENT Better analgesic drugs are desperately needed to help physicians to treat pain. While many pre-clinical studies support the analgesic effects of α-conopeptides, Vc1.1 and RgIA, the mechanism is controversial. The development of improved α-conopeptide analgesics would be greatly facilitated by a complete understanding of the analgesic mechanism. However, we show that we cannot reproduce one of the proposed analgesic mechanisms, which is an irreversible inhibition of CaV current in a majority of sensory neurons.
Collapse
|
41
|
α9-nicotinic acetylcholine receptors contribute to the maintenance of chronic mechanical hyperalgesia, but not thermal or mechanical allodynia. Mol Pain 2014; 10:64. [PMID: 25274008 PMCID: PMC4195954 DOI: 10.1186/1744-8069-10-64] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2014] [Accepted: 09/19/2014] [Indexed: 11/29/2022] Open
Abstract
Background The current pharmacological treatments for chronic pain are limited. The first analgesic drug approved for clinical use in decades that has a novel molecular target is the synthetic version of a naturally occurring conotoxin. Several conotoxins that target ion channels have progressed to clinical trials for the relief of pain. Vc1.1 and RgIA are analgesic α-conotoxins that target α9-subunit-containing nicotinic acetylcholine receptors (α9-nAChR) as well as GABAB receptor mechanisms. However, the evidence for the involvement of α9-nAChRs in pain is controversial. In the present study, the role of the α9-nAChR in pain was assessed using a battery of behavioural pain tests and pain models in α9-nAChR knockout (KO) mice. Results α9-nAChR KO mice showed normal responses to acute noxious thermal and mechanical stimuli, and developed normal chronic cold and mechanical allodynia in inflammatory and nerve injury pain models. However, KO animals developed mechanical hyperalgesia to a lesser extent than their wild type (WT) counterparts in both inflammatory and neuropathic pain models. Chronic neuropathic pain is sustained in WT mice for at least 21 days post injury, while KO mice show significant recovery by 14 days post injury. KO sham mice were also resistant to the repeated-measures effect of the noxious pain test that caused a gradual onset of mild mechanical hyperalgesia in WT sham animals. Conclusions The α9-nAChR is not involved in acute pain perception or chronic thermal or mechanical allodynia or thermal hyperalgesia but does contribute to the intensity and duration of chronic mechanical hyperalgesia, suggesting that pain-relieving actions of antagonists that target this site may be restricted to high threshold mechanosensation. The α9-nAChR appears to be a valid target for pharmacological compounds that alleviate long-term mechanical hyperalgesia and may be of use as a prophylactic drug to prevent the development of some symptoms of chronic pain.
Collapse
|
42
|
Di Cesare Mannelli L, Cinci L, Micheli L, Zanardelli M, Pacini A, McIntosh JM, Ghelardini C. α-conotoxin RgIA protects against the development of nerve injury-induced chronic pain and prevents both neuronal and glial derangement. Pain 2014; 155:1986-95. [PMID: 25008370 DOI: 10.1016/j.pain.2014.06.023] [Citation(s) in RCA: 89] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2014] [Revised: 06/05/2014] [Accepted: 06/30/2014] [Indexed: 12/18/2022]
Abstract
Neuropathic pain affects millions of people worldwide, causing substantial disability and greatly impairing quality of life. Commonly used analgesics or antihyperalgesic compounds are generally characterized by limited therapeutic outcomes. Thus, there is a compelling need for novel therapeutic strategies able to prevent nervous tissue alterations responsible for chronic pain. The α9α10 nicotinic acetylcholine receptor antagonist α-conotoxin RgIA (RgIA), a peptide isolated from the venom of a carnivorous cone snail, induces relief in both acute and chronic pain models. To evaluate potential disease-modifying effects of RgIA, the compound was given to rats following chronic constriction injury (CCI) of the sciatic nerve. Two or 10 nmol RgIA injected intramuscularly once a day for 14 days reduced the painful response to suprathreshold stimulation, increased pain threshold to nonnoxious stimuli, and normalized alterations in hind limb weight bearing. Histological analysis of the sciatic nerve revealed that RgIA prevented CCI-induced decreases of axonal compactness and diameter, loss of myelin sheath, and decreases in the fiber number. Moreover, RgIA significantly reduced edema and inflammatory infiltrate, including a decrease of CD86(+) macrophages. In L4-L5 dorsal root ganglia, RgIA prevented morphometric changes and reduced the inflammatory infiltrate consistent with a disease-modifying effect. In the dorsal horn of the spinal cord, RgIA prevented CCI-induced activation of microglia and astrocytes. These data suggest that RgIA-like compounds may represent a novel class of therapeutics for neuropathic pain that protects peripheral nervous tissues as well as prevents central maladaptive plasticity by inhibiting glial cell activation.
Collapse
Affiliation(s)
- Lorenzo Di Cesare Mannelli
- Department of Neuroscience, Psychology, Drug Research and Child Health - Neurofarba - Pharmacology and Toxicology Section, University of Florence, Florence, Italy.
| | - Lorenzo Cinci
- Department of Neuroscience, Psychology, Drug Research and Child Health - Neurofarba - Pharmacology and Toxicology Section, University of Florence, Florence, Italy
| | - Laura Micheli
- Department of Neuroscience, Psychology, Drug Research and Child Health - Neurofarba - Pharmacology and Toxicology Section, University of Florence, Florence, Italy
| | - Matteo Zanardelli
- Department of Neuroscience, Psychology, Drug Research and Child Health - Neurofarba - Pharmacology and Toxicology Section, University of Florence, Florence, Italy
| | - Alessandra Pacini
- Department of Experimental and Clinical Medicine - DMSC - Anatomy and Histology Section, University of Florence, Florence, Italy
| | - J Michael McIntosh
- George E. Wahlen Veterans Affairs Medical Center, Salt Lake City, UT, USA; Department of Biology, University of Utah, Salt Lake City, UT, USA; Department of Psychiatry, University of Utah, Salt Lake City, UT, USA
| | - Carla Ghelardini
- Department of Neuroscience, Psychology, Drug Research and Child Health - Neurofarba - Pharmacology and Toxicology Section, University of Florence, Florence, Italy
| |
Collapse
|
43
|
Conotoxins targeting nicotinic acetylcholine receptors: an overview. Mar Drugs 2014; 12:2970-3004. [PMID: 24857959 PMCID: PMC4052327 DOI: 10.3390/md12052970] [Citation(s) in RCA: 113] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2014] [Revised: 04/24/2014] [Accepted: 04/28/2014] [Indexed: 12/19/2022] Open
Abstract
Marine snails of the genus Conus are a large family of predatory gastropods with an unparalleled molecular diversity of pharmacologically active compounds in their venom. Cone snail venom comprises of a rich and diverse cocktail of peptide toxins which act on a wide variety of ion channels such as voltage-gated sodium- (NaV), potassium- (KV), and calcium- (CaV) channels as well as nicotinic acetylcholine receptors (nAChRs) which are classified as ligand-gated ion channels. The mode of action of several conotoxins has been the subject of investigation, while for many others this remains unknown. This review aims to give an overview of the knowledge we have today on the molecular pharmacology of conotoxins specifically interacting with nAChRs along with the structure–function relationship data.
Collapse
|
44
|
Chang YP, Banerjee J, Dowell C, Wu J, Gyanda R, Houghten RA, Toll L, McIntosh JM, Armishaw CJ. Discovery of a potent and selective α3β4 nicotinic acetylcholine receptor antagonist from an α-conotoxin synthetic combinatorial library. J Med Chem 2014; 57:3511-21. [PMID: 24649848 PMCID: PMC4358631 DOI: 10.1021/jm500183r] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023]
Abstract
α-Conotoxins are disulfide-rich peptide neurotoxins that selectively inhibit neuronal nicotinic acetylcholine receptors (nAChRs). The α3β4 nAChR subtype has been identified as a novel target for managing nicotine addiction. Using a mixture-based positional-scanning synthetic combinatorial library (PS-SCL) with the α4/4-conotoxin BuIA framework, we discovered a highly potent and selective α3β4 nAChR antagonist. The initial PS-SCL consisted of a total of 113 379 904 sequences that were screened for α3β4 nAChR inhibition, which facilitated the design and synthesis of a second generation library of 64 individual α-conotoxin derivatives. Eleven analogues were identified as α3β4 nAChR antagonists, with TP-2212-59 exhibiting the most potent antagonistic activity and selectivity over the α3β2 and α4β2 nAChR subtypes. Final electrophysiological characterization demonstrated that TP-2212-59 inhibited acetylcholine evoked currents in α3β4 nAChRs heterogeneously expressed in Xenopus laevis oocytes with a calculated IC50 of 2.3 nM and exhibited more than 1000-fold selectivity over the α3β2 and α7 nAChR subtypes. As such, TP-2212-59 is among the most potent α3β4 nAChRs antagonists identified to date and further demonstrates the utility of mixture-based combinatorial libraries in the discovery of novel α-conotoxin derivatives with refined pharmacological activity.
Collapse
Affiliation(s)
- Yi-Pin Chang
- Torrey Pines Institute for Molecular Studies , 11350 SW Village Parkway, Port St. Lucie, Florida 34987, United States
| | | | | | | | | | | | | | | | | |
Collapse
|
45
|
Luo S, Zhangsun D, Zhu X, Wu Y, Hu Y, Christensen S, Harvey PJ, Akcan M, Craik DJ, McIntosh JM. Characterization of a novel α-conotoxin TxID from Conus textile that potently blocks rat α3β4 nicotinic acetylcholine receptors. J Med Chem 2013; 56:9655-63. [PMID: 24200193 DOI: 10.1021/jm401254c] [Citation(s) in RCA: 52] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
The α3β4 nAChRs are implicated in pain sensation in the PNS and addiction to nicotine in the CNS. We identified an α-4/6-conotoxin (CTx) TxID from Conus textile. The new toxin consists of 15 amino acid residues with two disulfide bonds. TxID was synthesized using solid phase methods, and the synthetic peptide was functionally tested on nAChRs heterologously expressed in Xenopus laevis oocytes. TxID blocked rat α3β4 nAChRs with a 12.5 nM IC50, which places it among the most potent α3β4 nAChR antagonists. TxID also blocked the closely related α6/α3β4 with a 94 nM IC50 but showed little activity on other nAChR subtypes. NMR analysis showed that two major structural isomers exist in solution, one of which adopts a regular α-CTx fold but with different surface charge distribution to other 4/6 family members. α-CTx TxID is a novel tool with which to probe the structure and function of α3β4 nAChRs.
Collapse
Affiliation(s)
- Sulan Luo
- Key Laboratory of Tropical Biological Resources, Ministry of Education, Key Lab for Marine Drug of Haikou, Hainan University , Haikou Hainan, 570228, China
| | | | | | | | | | | | | | | | | | | |
Collapse
|
46
|
Armishaw CJ, Banerjee J, Ganno ML, Reilley KJ, Eans SO, Mizrachi E, Gyanda R, Hoot MR, Houghten RA, McLaughlin JP. Discovery of novel antinociceptive α-conotoxin analogues from the direct in vivo screening of a synthetic mixture-based combinatorial library. ACS COMBINATORIAL SCIENCE 2013; 15:153-61. [PMID: 23414173 DOI: 10.1021/co300152x] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Marine cone snail venoms consist of large, naturally occurring combinatorial libraries of disulfide-constrained peptide neurotoxins known as conotoxins, which have profound potential in the development of analgesics. In this study, we report a synthetic combinatorial strategy that probes the hypervariable regions of conotoxin frameworks to discover novel analgesic agents by utilizing high diversity mixture-based positional-scanning synthetic combinatorial libraries (PS-SCLs). We hypothesized that the direct in vivo testing of these mixture-based combinatorial library samples during the discovery phase would facilitate the identification of novel individual compounds with desirable antinociceptive profiles while simultaneously eliminating many compounds with poor activity or liabilities of locomotion and respiration. A PS-SCL was designed based on the α-conotoxin RgIA-ΔR n-loop region and consisted of 10,648 compounds systematically arranged into 66 mixture samples. Mixtures were directly screened in vivo using the mouse 55 °C warm-water tail-withdrawal assay, which allowed deconvolution of amino acid residues at each position that confer antinociceptive activity. A second generation library of 36 individual α-conotoxin analogues was synthesized using systematic combinations of amino acids identified from PS-SCL deconvolution and further screened for antinociceptive activity. Six individual analogues exhibited comparable antinociceptive activity to that of the recognized analgesic α-conotoxin RgIA-ΔR, and were selected for further examination of antinociceptive, respiratory, and locomotor effects. Three lead compounds were identified that produced dose-dependent antinociception without significant respiratory depression or decreased locomotor activity. Our results represent a unique approach for rapidly developing novel lead α-conotoxin analogues as low-liability analgesics with promising therapeutic potential.
Collapse
Affiliation(s)
- Christopher J. Armishaw
- Torrey Pines Institute for Molecular Studies, 11350 SW Village Pkwy, Port St. Lucie, Florida
34987, United States
| | - Jayati Banerjee
- Torrey Pines Institute for Molecular Studies, 11350 SW Village Pkwy, Port St. Lucie, Florida
34987, United States
| | - Michelle L. Ganno
- Torrey Pines Institute for Molecular Studies, 11350 SW Village Pkwy, Port St. Lucie, Florida
34987, United States
| | - Kate J. Reilley
- Torrey Pines Institute for Molecular Studies, 11350 SW Village Pkwy, Port St. Lucie, Florida
34987, United States
| | - Shainnel O. Eans
- Torrey Pines Institute for Molecular Studies, 11350 SW Village Pkwy, Port St. Lucie, Florida
34987, United States
| | - Elisa Mizrachi
- Torrey Pines Institute for Molecular Studies, 11350 SW Village Pkwy, Port St. Lucie, Florida
34987, United States
| | - Reena Gyanda
- Torrey Pines Institute for Molecular Studies, 11350 SW Village Pkwy, Port St. Lucie, Florida
34987, United States
| | - Michelle R. Hoot
- Torrey Pines Institute for Molecular Studies, 11350 SW Village Pkwy, Port St. Lucie, Florida
34987, United States
| | - Richard A. Houghten
- Torrey Pines Institute for Molecular Studies, 11350 SW Village Pkwy, Port St. Lucie, Florida
34987, United States
| | - Jay P. McLaughlin
- Torrey Pines Institute for Molecular Studies, 11350 SW Village Pkwy, Port St. Lucie, Florida
34987, United States
| |
Collapse
|
47
|
Adams DJ, Berecki G. Mechanisms of conotoxin inhibition of N-type (Ca(v)2.2) calcium channels. BIOCHIMICA ET BIOPHYSICA ACTA-BIOMEMBRANES 2013; 1828:1619-28. [PMID: 23380425 DOI: 10.1016/j.bbamem.2013.01.019] [Citation(s) in RCA: 66] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/19/2012] [Revised: 01/16/2013] [Accepted: 01/19/2013] [Indexed: 12/27/2022]
Abstract
N-type (Ca(v)2.2) voltage-gated calcium channels (VGCC) transduce electrical activity into other cellular functions, regulate calcium homeostasis and play a major role in processing pain information. Although the distribution and function of these channels vary widely among different classes of neurons, they are predominantly expressed in nerve terminals, where they control neurotransmitter release. To date, genetic and pharmacological studies have identified that high-threshold, N-type VGCCs are important for pain sensation in disease models. This suggests that N-type VGCC inhibitors or modulators could be developed into useful drugs to treat neuropathic pain. This review discusses the role of N-type (Ca(v)2.2) VGCCs in nociception and pain transmission through primary sensory dorsal root ganglion (DRG) neurons (nociceptors). It also outlines the potent and selective inhibition of N-type VGCCs by conotoxins, small disulfide-rich peptides isolated from the venom of marine cone snails. Of these conotoxins, ω-conotoxins are selective N-type VGCC antagonists that preferentially block nociception in inflammatory pain models, and allodynia and/or hyperalgesia in neuropathic pain models. Another conotoxin family, α-conotoxins, were initially proposed as competitive antagonists of muscle and neuronal nicotinic acetylcholine receptors (nAChR). Surprisingly, however, α-conotoxins Vc1.1 and RgIA, also potently inhibit N-type VGCC currents in the sensory DRG neurons of rodents and α9 nAChR knockout mice, via intracellular signaling mediated by G protein-coupled GABAB receptors. Understanding how conotoxins inhibit VGCCs is critical for developing these peptides into analgesics and may result in better pain management. This article is part of a Special Issue entitled: Calcium channels.
Collapse
Affiliation(s)
- David J Adams
- Health Innovations Research Institute, RMIT University, Melbourne, Victoria, Australia.
| | | |
Collapse
|