1
|
Wang T, Yang H, Wang X, Li R, Jiang Y, Ye J. Anti-arthritic effect of spirocyclopiperazinium bromide DXL-A-24 in CFA-induced arthritic rats and its mechanism. Eur J Pharmacol 2024; 984:177051. [PMID: 39393667 DOI: 10.1016/j.ejphar.2024.177051] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2024] [Revised: 09/05/2024] [Accepted: 10/07/2024] [Indexed: 10/13/2024]
Abstract
This study aimed to investigate the effect of spirocyclopiperazinium bromide DXL-A-24 on complete Freund's adjuvant (CFA)-induced arthritis and its underlying mechanism in rats. A rheumatoid arthritis model was established by the intradermal injection of CFA into the paws of rats. Mechanical withdrawal threshold (MWT), thermal withdrawal latency (TWL), ankle swelling and paw edema were used to evaluate the effects of DXL-A-24. Bone erosion and bone mineral density (BMD) were observed using micro-computed tomography. Receptor blocking test, western blotting, and enzyme-linked immunosorbent assay were performed to explore the mechanisms. Administration of DXL-A-24 (1, 0.5, 0.25 mg/kg, i.g.) dose-dependently increased the MWT and TWL, while alleviating ankle and paw swelling in CFA rats. The effects were blocked by peripheral α7 nicotinic or M4 muscarinic receptor antagonists. DXL-A-24 improved bone erosion and BMD, as well as downregulated the overexpression of Cav3.2, pJAK2, pSTAT3, pIκBα, pNF-κB p65, c-Fos and TNF-α proteins that were induced by CFA. In conclusion, this study shows, for the first time, that DXL-A-24 improves bone erosion and BMD and exhibits obvious anti-arthritic effects in CFA rats. The mechanism may be related to activating the peripheral α7 nicotinic and M4 muscarinic receptors, reducing Cav3.2 expression, and suppressing JAK2/STAT3 and IκBα/NF-κB p65 inflammatory signaling pathways, ultimately inhibiting inflammation-related proteins TNF-α and c-Fos.
Collapse
Affiliation(s)
- Tianyu Wang
- Department of Molecular and Cellular Pharmacology, School of Pharmaceutical Sciences, Peking University, Beijing, China
| | - Hua Yang
- Department of Molecular and Cellular Pharmacology, School of Pharmaceutical Sciences, Peking University, Beijing, China; Beijing Tsinghua Changgung Hospital, School of Clinical Medicine, Tsinghua University, Beijing, China
| | - Xin Wang
- Department of Chemical Biology, School of Pharmaceutical Sciences, Peking University, Beijing, China
| | - Runtao Li
- Department of Chemical Biology, School of Pharmaceutical Sciences, Peking University, Beijing, China
| | - Yimin Jiang
- Medical and Healthy Analysis Center, Peking University, Beijing, China
| | - Jia Ye
- Department of Molecular and Cellular Pharmacology, School of Pharmaceutical Sciences, Peking University, Beijing, China.
| |
Collapse
|
2
|
Antunes FTT, Campos MM, Carvalho VDPR, da Silva Junior CA, Magno LAV, de Souza AH, Gomez MV. Current Drug Development Overview: Targeting Voltage-Gated Calcium Channels for the Treatment of Pain. Int J Mol Sci 2023; 24:ijms24119223. [PMID: 37298174 DOI: 10.3390/ijms24119223] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2023] [Revised: 04/29/2023] [Accepted: 05/02/2023] [Indexed: 06/12/2023] Open
Abstract
Voltage-gated calcium channels (VGCCs) are targeted to treat pain conditions. Since the discovery of their relation to pain processing control, they are investigated to find new strategies for better pain control. This review provides an overview of naturally based and synthetic VGCC blockers, highlighting new evidence on the development of drugs focusing on the VGCC subtypes as well as mixed targets with pre-clinical and clinical analgesic effects.
Collapse
Affiliation(s)
- Flavia Tasmin Techera Antunes
- Department of Physiology and Pharmacology, University of Calgary, Calgary, AB T2N 1N4, Canada
- Hotchkiss Brain Institute, University of Calgary, Calgary, AB T2N 1N4, Canada
| | - Maria Martha Campos
- Programa de Pós-Graduação em Odontologia, Escola de Ciências da Saúde e da Vida, Pontifícia Universidade Católica do Rio Grande do Sul, Porto Alegre 90619-900, RS, Brazil
| | | | | | - Luiz Alexandre Viana Magno
- Programa de Pós-Graduação em Ciências da Saúde, Faculdade Ciências Médicas de Minas Gerais (FCMMG), Belo Horizonte 30110-005, MG, Brazil
| | - Alessandra Hubner de Souza
- Programa de Pós-Graduação em Ciências da Saúde, Faculdade Ciências Médicas de Minas Gerais (FCMMG), Belo Horizonte 30110-005, MG, Brazil
| | | |
Collapse
|
3
|
Sharma A, Rahman G, Gorelik J, Bhargava A. Voltage-Gated T-Type Calcium Channel Modulation by Kinases and Phosphatases: The Old Ones, the New Ones, and the Missing Ones. Cells 2023; 12:461. [PMID: 36766802 PMCID: PMC9913649 DOI: 10.3390/cells12030461] [Citation(s) in RCA: 10] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2022] [Revised: 01/14/2023] [Accepted: 01/29/2023] [Indexed: 02/04/2023] Open
Abstract
Calcium (Ca2+) can regulate a wide variety of cellular fates, such as proliferation, apoptosis, and autophagy. More importantly, changes in the intracellular Ca2+ level can modulate signaling pathways that control a broad range of physiological as well as pathological cellular events, including those important to cellular excitability, cell cycle, gene-transcription, contraction, cancer progression, etc. Not only intracellular Ca2+ level but the distribution of Ca2+ in the intracellular compartments is also a highly regulated process. For this Ca2+ homeostasis, numerous Ca2+ chelating, storage, and transport mechanisms are required. There are also specialized proteins that are responsible for buffering and transport of Ca2+. T-type Ca2+ channels (TTCCs) are one of those specialized proteins which play a key role in the signal transduction of many excitable and non-excitable cell types. TTCCs are low-voltage activated channels that belong to the family of voltage-gated Ca2+ channels. Over decades, multiple kinases and phosphatases have been shown to modulate the activity of TTCCs, thus playing an indirect role in maintaining cellular physiology. In this review, we provide information on the kinase and phosphatase modulation of TTCC isoforms Cav3.1, Cav3.2, and Cav3.3, which are mostly described for roles unrelated to cellular excitability. We also describe possible potential modulations that are yet to be explored. For example, both mitogen-activated protein kinase and citron kinase show affinity for different TTCC isoforms; however, the effect of such interaction on TTCC current/kinetics has not been studied yet.
Collapse
Affiliation(s)
- Ankush Sharma
- Department of Biotechnology, Indian Institute of Technology Hyderabad (IITH), Kandi 502284, Telangana, India
| | - Ghazala Rahman
- Department of Biotechnology, Indian Institute of Technology Hyderabad (IITH), Kandi 502284, Telangana, India
| | - Julia Gorelik
- National Heart and Lung Institute, Faculty of Medicine, Imperial College London, London W12 0NN, UK
| | - Anamika Bhargava
- Department of Biotechnology, Indian Institute of Technology Hyderabad (IITH), Kandi 502284, Telangana, India
| |
Collapse
|
4
|
Zhou K, Luo W, Liu T, Ni Y, Qin Z. Neurotoxins Acting at Synaptic Sites: A Brief Review on Mechanisms and Clinical Applications. Toxins (Basel) 2022; 15:18. [PMID: 36668838 PMCID: PMC9865788 DOI: 10.3390/toxins15010018] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2022] [Revised: 12/20/2022] [Accepted: 12/21/2022] [Indexed: 12/29/2022] Open
Abstract
Neurotoxins generally inhibit or promote the release of neurotransmitters or bind to receptors that are located in the pre- or post-synaptic membranes, thereby affecting physiological functions of synapses and affecting biological processes. With more and more research on the toxins of various origins, many neurotoxins are now widely used in clinical treatment and have demonstrated good therapeutic outcomes. This review summarizes the structural properties and potential pharmacological effects of neurotoxins acting on different components of the synapse, as well as their important clinical applications, thus could be a useful reference for researchers and clinicians in the study of neurotoxins.
Collapse
Affiliation(s)
- Kunming Zhou
- Department of Pharmacology and Laboratory of Aging and Nervous Diseases, College of Pharmaceutical Sciences, Suzhou Medical College of Soochow University, Suzhou 215123, China
| | - Weifeng Luo
- Department of Neurology and Clinical Research Center of Neurological Disease, The Second Affiliated Hospital of Soochow University, Suzhou 215004, China
| | - Tong Liu
- Institute of Pain Medicine and Special Environmental Medicine, Nantong University, Nantong 226019, China
| | - Yong Ni
- Department of Neurology and Clinical Research Center of Neurological Disease, The Second Affiliated Hospital of Soochow University, Suzhou 215004, China
| | - Zhenghong Qin
- Department of Pharmacology and Laboratory of Aging and Nervous Diseases, College of Pharmaceutical Sciences, Suzhou Medical College of Soochow University, Suzhou 215123, China
| |
Collapse
|
5
|
Talukdar A, Maddhesiya P, Namsa ND, Doley R. Snake venom toxins targeting the central nervous system. TOXIN REV 2022. [DOI: 10.1080/15569543.2022.2084418] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/17/2022]
Affiliation(s)
- Amit Talukdar
- Department of Molecular Biology and Biotechnology, Tezpur University, Assam, India
| | - Priya Maddhesiya
- Cell Biology and Anatomy, Ludwig Maximilian University (LMU), Munich, Germany
| | - Nima Dondu Namsa
- Department of Molecular Biology and Biotechnology, Tezpur University, Assam, India
| | - Robin Doley
- Department of Molecular Biology and Biotechnology, Tezpur University, Assam, India
| |
Collapse
|
6
|
Xu L, Wang S, Zhang L, Liu B, Zheng S, Yao M. Cobratoxin Alleviates Cancer-Induced Bone Pain in Rats via Inhibiting CaMKII Signaling Pathway after Acting on M4 Muscarinic Cholinergic Receptors. ACS Chem Neurosci 2022; 13:1422-1432. [PMID: 35420768 DOI: 10.1021/acschemneuro.2c00048] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022] Open
Abstract
Cancer-induced bone pain (CIBP) is a common pain in clinics, which can reduce the quality of life and increase the mortality of patients, but the treatment of CIBP is limited. This study was designed to investigate the analgesic effect of α-cobratoxin on CIBP and further to explore the molecular target and potential signal pathway. As shown by the mechanical allodynia test in a CIBP rat model, administration of α-cobratoxin produced significant analgesia in a dose-dependent manner, and the analgesic effects were blocked by pretreatment with an intrathecal injection of M4 mAChR-siRNA or intraperitoneal injection of tropicamide, an antagonist of M4 muscarinic cholinergic receptor. Whole-cell patch-clamp recording showed that α-cobratoxin can decrease the spontaneous firing and spontaneous excitatory postsynaptic currents of SDH neurons in CIBP rats. In primary lumber SDH neurons, intracellular calcium measurement revealed that α-cobratoxin decreased intracellular calcium concentration, and immunofluorescence demonstrated that M4 muscarinic cholinergic receptor and CaMKII/CREB were co-expressed. In the CIBP model and primary SDH neurons, Western blot showed that the levels of p-CaMKII and p-CREB were increased by α-cobratoxin and the effect of α-cobratoxin was antagonized by M4 mAChR-siRNA. The quantitative polymerase chain reaction (qPCR) results showed that α-cobratoxin downregulated the expression of proinflammatory cytokines through M4 muscarinic cholinergic receptor in SDH. These results suggest that α-cobratoxin may activate M4 muscarinic cholinergic receptor, triggering the inhibition of SDH neuronal excitability via CaMKII signaling pathway, thereby resulting in antagonistic effects in the CIBP rat model.
Collapse
Affiliation(s)
- Longsheng Xu
- Department of Anesthesia and Pain Medicine, Affiliated Hospital of Jiaxing University, Jiaxing 314000, China
| | - Shizhen Wang
- Department of Basic Medicine, Jiangsu Vocational College of Nursing, Huaian 223001, China
| | - Ling Zhang
- Department of central laboratory, Affiliated Zhangjiagang Hospital of Suzhou University, Zhangjiagang 215600, China
| | - Beibei Liu
- Department of Anesthesia and Pain Medicine, Affiliated Hospital of Jiaxing University, Jiaxing 314000, China
| | - Shang Zheng
- Department of Anesthesia and Pain Medicine, Affiliated Hospital of Jiaxing University, Jiaxing 314000, China
| | - Ming Yao
- Department of Anesthesia and Pain Medicine, Affiliated Hospital of Jiaxing University, Jiaxing 314000, China
| |
Collapse
|
7
|
Trevisan G, Oliveira SM. Animal Venom Peptides Cause Antinociceptive Effects by Voltage-gated Calcium Channels Activity Blockage. Curr Neuropharmacol 2022; 20:1579-1599. [PMID: 34259147 PMCID: PMC9881091 DOI: 10.2174/1570159x19666210713121217] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2021] [Revised: 06/02/2021] [Accepted: 06/09/2021] [Indexed: 11/22/2022] Open
Abstract
Pain is a complex phenomenon that is usually unpleasant and aversive. It can range widely in intensity, quality, and duration and has diverse pathophysiologic mechanisms and meanings. Voltage-gated sodium and calcium channels are essential to transmitting painful stimuli from the periphery until the dorsal horn of the spinal cord. Thus, blocking voltage-gated calcium channels (VGCCs) can effectively control pain refractory to treatments currently used in the clinic, such as cancer and neuropathic pain. VGCCs blockers isolated of cobra Naja naja kaouthia (α-cobratoxin), spider Agelenopsis aperta (ω-Agatoxin IVA), spider Phoneutria nigriventer (PhTx3.3, PhTx3.4, PhTx3.5, PhTx3.6), spider Hysterocrates gigas (SNX-482), cone snails Conus geographus (GVIA), Conus magus (MVIIA or ziconotide), Conus catus (CVID, CVIE and CVIF), Conus striatus (SO- 3), Conus fulmen (FVIA), Conus moncuri (MoVIA and MoVIB), Conus regularis (RsXXIVA), Conus eburneus (Eu1.6), Conus victoriae (Vc1.1.), Conus regius (RgIA), and spider Ornithoctonus huwena (huwentoxin-I and huwentoxin-XVI) venoms caused antinociceptive effects in different acute and chronic pain models. Currently, ziconotide is the only clinical used N-type VGCCs blocker peptide for chronic intractable pain. However, ziconotide causes different adverse effects, and the intrathecal route of administration also impairs its use in a more significant number of patients. In this sense, peptides isolated from animal venoms or their synthetic forms that act by modulating or blocking VGCCs channels seem to be a relevant prototype for developing new analgesics efficacious and well tolerated by patients.
Collapse
Affiliation(s)
- Gabriela Trevisan
- Graduated Program in Pharmacology, Federal University of Santa Maria (UFSM), Santa Maria, RS 97105-900, Brazil
| | - Sara Marchesan Oliveira
- Graduated Program in Biological Sciences: Toxicological Biochemistry, Federal University of Santa Maria (UFSM), Santa Maria, RS 97105-900, Brazil
| |
Collapse
|
8
|
Zhang Y, Qian Z, Jiang D, Sun Y, Gao S, Jiang X, Wang H, Tao J. Neuromedin B receptor stimulation of Cav3.2 T-type Ca 2+ channels in primary sensory neurons mediates peripheral pain hypersensitivity. Theranostics 2021; 11:9342-9357. [PMID: 34646374 PMCID: PMC8490515 DOI: 10.7150/thno.62255] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2021] [Accepted: 09/01/2021] [Indexed: 01/21/2023] Open
Abstract
Background: Neuromedin B (Nmb) is implicated in the regulation of nociception of sensory neurons. However, the underlying cellular and molecular mechanisms remain unknown. Methods: Using patch clamp recording, western blot analysis, immunofluorescent labelling, enzyme-linked immunosorbent assays, adenovirus-mediated shRNA knockdown and animal behaviour tests, we studied the effects of Nmb on the sensory neuronal excitability and peripheral pain sensitivity mediated by Cav3.2 T-type channels. Results: Nmb reversibly and concentration-dependently increased T-type channel currents (IT) in small-sized trigeminal ganglion (TG) neurons through the activation of neuromedin B receptor (NmbR). This NmbR-mediated IT response was Gq protein-coupled, but independent of protein kinase C activity. Either intracellular application of the QEHA peptide or shRNA-mediated knockdown of Gβ abolished the NmbR-induced IT response. Inhibition of protein kinase A (PKA) or AMP-activated protein kinase (AMPK) completely abolished the Nmb-induced IT response. Analysis of phospho-AMPK (p-AMPK) revealed that Nmb significantly activated AMPK, while AMPK inhibition prevented the Nmb-induced increase in PKA activity. In a heterologous expression system, activation of NmbR significantly enhanced the Cav3.2 channel currents, while the Cav3.1 and Cav3.3 channel currents remained unaffected. Nmb induced TG neuronal hyperexcitability and concomitantly induced mechanical and thermal hypersensitivity, both of which were attenuated by T-type channel blockade. Moreover, blockade of NmbR signalling prevented mechanical hypersensitivity in a mouse model of complete Freund's adjuvant-induced inflammatory pain, and this effect was attenuated by siRNA knockdown of Cav3.2. Conclusions: Our study reveals a novel mechanism by which NmbR stimulates Cav3.2 channels through a Gβγ-dependent AMPK/PKA pathway. In mouse models, this mechanism appears to drive the hyperexcitability of TG neurons and induce pain hypersensitivity.
Collapse
|
9
|
Han Z, Chai W, Wang Z, Xiao F, Dai J. Quantum energy levels of glutamate modulate neural biophotonic signals. Photochem Photobiol Sci 2021; 20:343-356. [PMID: 33721274 DOI: 10.1007/s43630-021-00022-0] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2020] [Accepted: 02/03/2021] [Indexed: 02/07/2023]
Abstract
Glutamate is the most abundant excitatory neurotransmitter in the brain, and it plays an essential and important role in neural functions. Current studies have shown that glutamate can induce neural biophotonic activity and transmission, which may involve the mechanism of photon quantum brain; however, it is unclear whether such a mechanism follows the principle of quantum mechanics. Here we show that the action of glutamate on its receptors leads to a decrease in its quantum energy levels, and glutamate then partially or completely loses its function to further induce the biophotonic activity in mouse brain slices. The reduced quantum energy levels of glutamate can be restored by direct-current electrical discharges and the use of energy transfer of chloroplast photosynthesis; hence, the quantum energy recovered glutamate can again induce significant biophotonic activity. Furthermore, the changes in quantum energy levels of glutamate are related to the exchange and transfer of electron energy on its active hydrogen atom. These findings suggest that the glutamate-induced neural biophotonic signals may be involved in the transfer of the quantum energy levels of glutamate, which implies a quantum mechanism of neurotransmitter action.
Collapse
Affiliation(s)
- Zhengrong Han
- Wuhan Institute for Neuroscience and Neuroengineering (WINN), South-Central University for Nationalities, Minzu Dadao 182, Wuhan, 430074, China.,Department of Neurobiology, College of Life Sciences, South-Central University for Nationalities, Wuhan, 430074, China
| | - Weitai Chai
- Wuhan Institute for Neuroscience and Neuroengineering (WINN), South-Central University for Nationalities, Minzu Dadao 182, Wuhan, 430074, China.,Department of Neurobiology, College of Life Sciences, South-Central University for Nationalities, Wuhan, 430074, China
| | - Zhuo Wang
- Wuhan Institute for Neuroscience and Neuroengineering (WINN), South-Central University for Nationalities, Minzu Dadao 182, Wuhan, 430074, China.,Department of Neurobiology, College of Life Sciences, South-Central University for Nationalities, Wuhan, 430074, China
| | - Fangyan Xiao
- Wuhan Institute for Neuroscience and Neuroengineering (WINN), South-Central University for Nationalities, Minzu Dadao 182, Wuhan, 430074, China.,Department of Neurobiology, College of Life Sciences, South-Central University for Nationalities, Wuhan, 430074, China
| | - Jiapei Dai
- Wuhan Institute for Neuroscience and Neuroengineering (WINN), South-Central University for Nationalities, Minzu Dadao 182, Wuhan, 430074, China. .,Department of Neurobiology, College of Life Sciences, South-Central University for Nationalities, Wuhan, 430074, China.
| |
Collapse
|
10
|
Nirthanan S. Snake three-finger α-neurotoxins and nicotinic acetylcholine receptors: molecules, mechanisms and medicine. Biochem Pharmacol 2020; 181:114168. [PMID: 32710970 DOI: 10.1016/j.bcp.2020.114168] [Citation(s) in RCA: 36] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2020] [Revised: 07/20/2020] [Accepted: 07/20/2020] [Indexed: 12/13/2022]
Abstract
Snake venom three-finger α-neurotoxins (α-3FNTx) act on postsynaptic nicotinic acetylcholine receptors (nAChRs) at the neuromuscular junction (NMJ) to produce skeletal muscle paralysis. The discovery of the archetypal α-bungarotoxin (α-BgTx), almost six decades ago, exponentially expanded our knowledge of membrane receptors and ion channels. This included the localisation, isolation and characterization of the first receptor (nAChR); and by extension, the pathophysiology and pharmacology of neuromuscular transmission and associated pathologies such as myasthenia gravis, as well as our understanding of the role of α-3FNTxs in snakebite envenomation leading to novel concepts of targeted treatment. Subsequent studies on a variety of animal venoms have yielded a plethora of novel toxins that have revolutionized molecular biomedicine and advanced drug discovery from bench to bedside. This review provides an overview of nAChRs and their subtypes, classification of α-3FNTxs and the challenges of typifying an increasing arsenal of structurally and functionally unique toxins, and the three-finger protein (3FP) fold in the context of the uPAR/Ly6/CD59/snake toxin superfamily. The pharmacology of snake α-3FNTxs including their mechanisms of neuromuscular blockade, variations in reversibility of nAChR interactions, specificity for nAChR subtypes or for distinct ligand-binding interfaces within a subtype and the role of α-3FNTxs in neurotoxic envenomation are also detailed. Lastly, a reconciliation of structure-function relationships between α-3FNTx and nAChRs, derived from historical mutational and biochemical studies and emerging atomic level structures of nAChR models in complex with α-3FNTxs is discussed.
Collapse
Affiliation(s)
- Selvanayagam Nirthanan
- School of Medical Science, Griffith Health Group, Griffith University, Gold Coast, Queensland, Australia.
| |
Collapse
|
11
|
Trento MVC, Sales TA, de Abreu TS, Braga MA, Cesar PHS, Marques TR, Marcussi S. Exploring the structural and functional aspects of the phospholipase A 2 from Naja spp. Int J Biol Macromol 2019; 140:49-58. [PMID: 31421173 DOI: 10.1016/j.ijbiomac.2019.08.125] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2019] [Revised: 08/13/2019] [Accepted: 08/14/2019] [Indexed: 01/22/2023]
Abstract
Naja spp. venom is a natural source of active compounds with therapeutic application potential. Phospholipase A2 (PLA2) is abundant in the venom of Naja spp. and can perform neurotoxicity, cytotoxicity, cardiotoxicity, and hematological disorders. The PLA2s from Naja spp. venoms are Asp 49 isoenzymes with the exception of PLA2 Cys 49 from Naja sagittifera. When looking at the functional aspects, the neurotoxicity occurs by PLA2 called β-toxins that have affinity for phosphatidylcholine in nerve endings and synaptosomes membranes, and by α-toxins that block the nicotinic acetylcholine receptors in the neuromuscular junctions. In addition, these neurotoxins may inhibit K+ and Ca++ channels or even interfere with the Na+/K+/ATPase enzyme. The disturbance in the membrane fluidity also results in inhibition of the release of acetylcholine. The PLA2 can act as anticoagulants or procoagulant. The cytotoxicity exerted by PLA2s result from changes in the cardiomyocyte membranes, triggering cardiac failure and hemolysis. The antibacterial activity, however, is the result of alterations that decrease the stability of the lipid bilayer. Thus, the understanding of the structural and functional aspects of PLA2s can contribute to studies on the toxic and therapeutic mechanisms involved in the envenomation by Naja spp. and in the treatment of pathologies.
Collapse
Affiliation(s)
- Marcus Vinícius Cardoso Trento
- Biochemistry Laboratory, Department of Chemistry, Universidade Federal de Lavras (UFLA), Lavras, Minas Gerais 37200-000, Brazil.
| | - Thais Aparecida Sales
- Computational Chemistry Laboratory, Department of Chemistry, Universidade Federal de Lavras (UFLA), Lavras, Minas Gerais 37200-000, Brazil
| | - Tatiane Silva de Abreu
- Biochemistry Laboratory, Department of Chemistry, Universidade Federal de Lavras (UFLA), Lavras, Minas Gerais 37200-000, Brazil
| | - Mariana Aparecida Braga
- Biochemistry Laboratory, Department of Chemistry, Universidade Federal de Lavras (UFLA), Lavras, Minas Gerais 37200-000, Brazil
| | - Pedro Henrique Souza Cesar
- Biochemistry Laboratory, Department of Chemistry, Universidade Federal de Lavras (UFLA), Lavras, Minas Gerais 37200-000, Brazil
| | - Tamara Rezende Marques
- Biochemistry Laboratory, Department of Chemistry, Universidade Federal de Lavras (UFLA), Lavras, Minas Gerais 37200-000, Brazil
| | - Silvana Marcussi
- Biochemistry Laboratory, Department of Chemistry, Universidade Federal de Lavras (UFLA), Lavras, Minas Gerais 37200-000, Brazil
| |
Collapse
|
12
|
Muratspahić E, Freissmuth M, Gruber CW. Nature-Derived Peptides: A Growing Niche for GPCR Ligand Discovery. Trends Pharmacol Sci 2019; 40:309-326. [PMID: 30955896 DOI: 10.1016/j.tips.2019.03.004] [Citation(s) in RCA: 35] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2018] [Revised: 02/26/2019] [Accepted: 03/06/2019] [Indexed: 12/11/2022]
Abstract
G protein-coupled receptors (GPCRs) represent important drug targets, as they regulate pivotal physiological processes and they have proved to be readily druggable. Natural products have been and continue to be amongst the most valuable sources for drug discovery and development. Here, we surveyed small molecules and (poly-)peptides derived from plants, animals, fungi, and bacteria, which modulate GPCR signaling. Among naturally occurring compounds, peptides from plants, cone-snails, snakes, spiders, scorpions, fungi, and bacteria are of particular interest as lead compounds for the development of GPCR ligands, since they cover a chemical space, which differs from that of synthetic small molecules. Peptides, however, face challenges, some of which can be overcome by studying plant-derived compounds. We argue here that the opportunities outweigh the challenges.
Collapse
Affiliation(s)
- Edin Muratspahić
- Institute of Pharmacology, Center for Physiology and Pharmacology, Medical University of Vienna, Austria
| | - Michael Freissmuth
- Institute of Pharmacology, Center for Physiology and Pharmacology, Medical University of Vienna, Austria; Gaston H. Glock Research Laboratories for Exploratory Drug Development, Center for Physiology and Pharmacology, Medical University of Vienna, Austria
| | - Christian W Gruber
- Institute of Pharmacology, Center for Physiology and Pharmacology, Medical University of Vienna, Austria.
| |
Collapse
|
13
|
Zhang Y, Ji H, Wang J, Sun Y, Qian Z, Jiang X, Snutch TP, Sun Y, Tao J. Melatonin-mediated inhibition of Cav3.2 T-type Ca 2+ channels induces sensory neuronal hypoexcitability through the novel protein kinase C-eta isoform. J Pineal Res 2018; 64:e12476. [PMID: 29437250 DOI: 10.1111/jpi.12476] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/01/2017] [Accepted: 02/04/2018] [Indexed: 12/29/2022]
Abstract
Recent studies implicate melatonin in the antinociceptive activity of sensory neurons. However, the underlying mechanisms are still largely unknown. Here, we identify a critical role of melatonin in functionally regulating Cav3.2 T-type Ca2+ channels (T-type channel) in trigeminal ganglion (TG) neurons. Melatonin inhibited T-type channels in small TG neurons via the melatonin receptor 2 (MT2 receptor) and a pertussis toxin-sensitive G-protein pathway. Immunoprecipitation analyses revealed that the intracellular subunit of the MT2 receptor coprecipitated with Gαo . Both shRNA-mediated knockdown of Gαo and intracellular application of QEHA peptide abolished the inhibitory effects of melatonin. Protein kinase C (PKC) antagonists abolished the melatonin-induced T-type channel response, whereas inhibition of conventional PKC isoforms elicited no effect. Furthermore, application of melatonin increased membrane abundance of PKC-eta (PKCη ) while antagonism of PKCη or shRNA targeting PKCη prevented the melatonin-mediated effects. In a heterologous expression system, activation of MT2 receptor strongly inhibited Cav3.2 T-type channel currents but had no effect on Cav3.1 and Cav3.3 current amplitudes. The selective Cav3.2 response was PKCη dependent and was accompanied by a negative shift in the steady-state inactivation curve. Furthermore, melatonin decreased the action potential firing rate of small TG neurons and attenuated the mechanical hypersensitivity in a mouse model of complete Freund's adjuvant-induced inflammatory pain. These actions were inhibited by T-type channel blockade. Together, our results demonstrated that melatonin inhibits Cav3.2 T-type channel activity through the MT2 receptor coupled to novel Gβγ -mediated PKCη signaling, subsequently decreasing the membrane excitability of TG neurons and pain hypersensitivity in mice.
Collapse
Affiliation(s)
- Yuan Zhang
- Department of Physiology and Neurobiology & Centre for Ion Channelopathy, Medical College of Soochow University, Suzhou, China
- Department of Geriatrics & Institute of Neuroscience, the Second Affiliated Hospital of Soochow University, Suzhou, China
| | - Heyi Ji
- Department of Physiology and Neurobiology & Centre for Ion Channelopathy, Medical College of Soochow University, Suzhou, China
| | - Jiangong Wang
- Department of Physiology and Neurobiology & Centre for Ion Channelopathy, Medical College of Soochow University, Suzhou, China
| | - Yufang Sun
- Department of Physiology and Neurobiology & Centre for Ion Channelopathy, Medical College of Soochow University, Suzhou, China
| | - Zhiyuan Qian
- Department of Geriatrics & Institute of Neuroscience, the Second Affiliated Hospital of Soochow University, Suzhou, China
| | - Xinghong Jiang
- Department of Physiology and Neurobiology & Centre for Ion Channelopathy, Medical College of Soochow University, Suzhou, China
| | - Terrance P Snutch
- Michael Smith Laboratories and Djavad Mowafaghian Centre for Brain Health, University of British Columbia, Vancouver, BC, Canada
| | - Yangang Sun
- Institute of Neuroscience, State Key Laboratory of Neuroscience, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai, China
| | - Jin Tao
- Department of Physiology and Neurobiology & Centre for Ion Channelopathy, Medical College of Soochow University, Suzhou, China
| |
Collapse
|
14
|
Si W, Zhang Y, Chen K, Hu D, Qian Z, Gong S, Li H, Hao Y, Tao J. Fibroblast growth factor type 1 receptor stimulation of T-type Ca2+ channels in sensory neurons requires the phosphatidylinositol 3-kinase and protein kinase A pathways, independently of Akt. Cell Signal 2018; 45:93-101. [DOI: 10.1016/j.cellsig.2018.01.024] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2017] [Revised: 01/25/2018] [Accepted: 01/25/2018] [Indexed: 02/08/2023]
|
15
|
Bailly-Chouriberry L, Garcia P, Cormant F, Loup B, Popot MA, Bonnaire Y. Use of split-free nano-liquid chromatography-mass spectrometry/high resolution mass spectrometry interface to improve the detection of α
-cobratoxin in equine plasma for doping control. Drug Test Anal 2018; 10:880-885. [DOI: 10.1002/dta.2348] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2016] [Revised: 11/22/2017] [Accepted: 11/28/2017] [Indexed: 11/12/2022]
Affiliation(s)
| | - Patrice Garcia
- Laboratoire des Courses Hippiques (LCH); Verrières le Buisson France
| | - Florence Cormant
- Laboratoire des Courses Hippiques (LCH); Verrières le Buisson France
| | - Benoit Loup
- Laboratoire des Courses Hippiques (LCH); Verrières le Buisson France
| | - Marie-Agnès Popot
- Laboratoire des Courses Hippiques (LCH); Verrières le Buisson France
| | - Yves Bonnaire
- Laboratoire des Courses Hippiques (LCH); Verrières le Buisson France
| |
Collapse
|
16
|
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: 7.7] [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
|
17
|
Kudryavtsev DS, Shelukhina IV, Son LV, Ojomoko LO, Kryukova EV, Lyukmanova EN, Zhmak MN, Dolgikh DA, Ivanov IA, Kasheverov IE, Starkov VG, Ramerstorfer J, Sieghart W, Tsetlin VI, Utkin YN. Neurotoxins from snake venoms and α-conotoxin ImI inhibit functionally active ionotropic γ-aminobutyric acid (GABA) receptors. J Biol Chem 2015. [PMID: 26221036 DOI: 10.1074/jbc.m115.648824] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
Ionotropic receptors of γ-aminobutyric acid (GABAAR) regulate neuronal inhibition and are targeted by benzodiazepines and general anesthetics. We show that a fluorescent derivative of α-cobratoxin (α-Ctx), belonging to the family of three-finger toxins from snake venoms, specifically stained the α1β3γ2 receptor; and at 10 μm α-Ctx completely blocked GABA-induced currents in this receptor expressed in Xenopus oocytes (IC50 = 236 nm) and less potently inhibited α1β2γ2 ≈ α2β2γ2 > α5β2γ2 > α2β3γ2 and α1β3δ GABAARs. The α1β3γ2 receptor was also inhibited by some other three-finger toxins, long α-neurotoxin Ls III and nonconventional toxin WTX. α-Conotoxin ImI displayed inhibitory activity as well. Electrophysiology experiments showed mixed competitive and noncompetitive α-Ctx action. Fluorescent α-Ctx, however, could be displaced by muscimol indicating that most of the α-Ctx-binding sites overlap with the orthosteric sites at the β/α subunit interface. Modeling and molecular dynamic studies indicated that α-Ctx or α-bungarotoxin seem to interact with GABAAR in a way similar to their interaction with the acetylcholine-binding protein or the ligand-binding domain of nicotinic receptors. This was supported by mutagenesis studies and experiments with α-conotoxin ImI and a chimeric Naja oxiana α-neurotoxin indicating that the major role in α-Ctx binding to GABAAR is played by the tip of its central loop II accommodating under loop C of the receptors.
Collapse
Affiliation(s)
- Denis S Kudryavtsev
- From the Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, 16/10 Miklukho-Maklaya Street, Moscow 117997, Russia
| | - Irina V Shelukhina
- From the Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, 16/10 Miklukho-Maklaya Street, Moscow 117997, Russia
| | - Lina V Son
- the Moscow Institute of Physics and Technology, Institutsky Per. 9, Dolgoprudny, Moscow Region 141700, Russia
| | - Lucy O Ojomoko
- From the Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, 16/10 Miklukho-Maklaya Street, Moscow 117997, Russia
| | - Elena V Kryukova
- From the Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, 16/10 Miklukho-Maklaya Street, Moscow 117997, Russia
| | - Ekaterina N Lyukmanova
- From the Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, 16/10 Miklukho-Maklaya Street, Moscow 117997, Russia, the Lomonosov Moscow State University, Moscow 119991, Russia, and
| | - Maxim N Zhmak
- From the Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, 16/10 Miklukho-Maklaya Street, Moscow 117997, Russia, the Syneuro OOO, ul. Miklukho-Maklaya 16/10, Moscow 117997, Russia
| | - Dmitry A Dolgikh
- From the Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, 16/10 Miklukho-Maklaya Street, Moscow 117997, Russia, the Lomonosov Moscow State University, Moscow 119991, Russia, and
| | - Igor A Ivanov
- From the Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, 16/10 Miklukho-Maklaya Street, Moscow 117997, Russia
| | - Igor E Kasheverov
- From the Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, 16/10 Miklukho-Maklaya Street, Moscow 117997, Russia
| | - Vladislav G Starkov
- From the Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, 16/10 Miklukho-Maklaya Street, Moscow 117997, Russia
| | - Joachim Ramerstorfer
- the Department of Molecular Neurosciences, Center for Brain Research, Medical University Vienna, A-1090 Vienna, Austria
| | - Werner Sieghart
- the Department of Molecular Neurosciences, Center for Brain Research, Medical University Vienna, A-1090 Vienna, Austria
| | - Victor I Tsetlin
- From the Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, 16/10 Miklukho-Maklaya Street, Moscow 117997, Russia
| | - Yuri N Utkin
- From the Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, 16/10 Miklukho-Maklaya Street, Moscow 117997, Russia,
| |
Collapse
|
18
|
Zhang Y, Li H, Pu Y, Gong S, Liu C, Jiang X, Tao J. Melatonin-mediated inhibition of Purkinje neuron P-type Ca²⁺ channels in vitro induces neuronal hyperexcitability through the phosphatidylinositol 3-kinase-dependent protein kinase C delta pathway. J Pineal Res 2015; 58:321-34. [PMID: 25707622 DOI: 10.1111/jpi.12218] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/25/2014] [Accepted: 02/20/2015] [Indexed: 12/18/2022]
Abstract
Although melatonin receptors are widely expressed in the mammalian central nervous system and peripheral tissues, there are limited data regarding the functions of melatonin in cerebellar Purkinje cells. Here, we identified a novel functional role of melatonin in modulating P-type Ca(2+) channels and action-potential firing in rat Purkinje neurons. Melatonin at 0.1 μm reversibly decreased peak currents (I(Ba)) by 32.9%. This effect was melatonin receptor 1 (MT(R1)) dependent and was associated with a hyperpolarizing shift in the voltage dependence of inactivation. Pertussis toxin pretreatment, intracellular application of QEHA peptide, and a selective antibody raised against the Gβ subunit prevented the inhibitory effects of melatonin. Pretreatment with phosphatidylinositol 3-kinase (PI3K) inhibitors abolished the melatonin-induced decrease in I(Ba). Surprisingly, melatonin responses were not regulated by Akt, a common downstream target of PI3K. Melatonin treatment significantly increased protein kinase C (PKC) activity 2.1-fold. Antagonists of PKC, but not of protein kinase A, abolished the melatonin-induced decrease in I(Ba). Melatonin application increased the membrane abundance of PKCδ, and PKCδ inhibition (either pharmacologically or genetically) abolished the melatonin-induced IBa response. Functionally, melatonin increased spontaneous action-potential firing by 53.0%; knockdown of MT(R1) and blockade of P-type channels abolished this effect. Thus, our results suggest that melatonin inhibits P-type channels through MT(R1) activation, which is coupled sequentially to the βγ subunits of G(i/o)-protein and to downstream PI3K-dependent PKCδ signaling. This likely contributes to its physiological functions, including spontaneous firing of cerebellar Purkinje neurons.
Collapse
Affiliation(s)
- Yuan Zhang
- Department of Neurobiology, Medical College of Soochow University, Suzhou, China; Department of Geriatrics and Institute of Neuroscience, The Second Affiliated Hospital of Soochow University, Suzhou, China
| | | | | | | | | | | | | |
Collapse
|
19
|
Gong S, Liang Q, Zhu Q, Ding D, Yin Q, Tao J, Jiang X. Nicotinic acetylcholine receptor α7 subunit is involved in the cobratoxin-induced antinociception in an animal model of neuropathic pain. Toxicon 2014; 93:31-6. [PMID: 25447771 DOI: 10.1016/j.toxicon.2014.11.222] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2014] [Revised: 05/19/2014] [Accepted: 11/05/2014] [Indexed: 11/17/2022]
Abstract
In this study we report that cobratoxin (CbTX), a long-chain postsynaptic α-neurotoxin isolated from the Thailand cobra, Naja naja kaouthia, has antinociceptive effect in rats with neuropathic pain. The neuropathic pain model was established in rats with partial sciatic nerve ligature (PSNL) method. The pain response was examined behaviorally with mechanical paw withdrawal and thermal paw withdrawal method. Different doses (0.56, 1.12 and 4.50 μg/kg) of CbTX were injected intrathecally. Injection of CbTX resulted in a significant dose-dependent antinociception as evidenced by increased mechanical withdrawal threshold and thermal withdrawal latency. CbTX also induces a significant dose-dependent inhibition of pain-evoked unit discharges of thalamic parafascicular neurons. Both the behavioral mechanical and thermal antinociception and the inhibition of pain-evoked discharges of neurons in thalamic parafascicular nucleus in PSNL model could be mimicked by PUN282987, selective α7 nicotinic AChR (α7 nAChR) agonist and reversed by methyllycaconitine (MLA) selective α7 nAChR antagonist. In summary, these results suggested that AChR α7 subunit was involved in the antinociceptive action of CbTX for neuropathic pain and might be the candidate target for analgesic drug design.
Collapse
Affiliation(s)
- Shan Gong
- Department of Physiology and Neurobiology, Key Laboratory of Pain Research and Therapy, Medical College of Soochow University, PR China
| | - Qian Liang
- Department of Physiology and Neurobiology, Key Laboratory of Pain Research and Therapy, Medical College of Soochow University, PR China
| | - Qi Zhu
- Department of Physiology and Neurobiology, Key Laboratory of Pain Research and Therapy, Medical College of Soochow University, PR China
| | - Dayong Ding
- Department of Physiology and Neurobiology, Key Laboratory of Pain Research and Therapy, Medical College of Soochow University, PR China
| | - Qizhang Yin
- Department of Physiology and Neurobiology, Key Laboratory of Pain Research and Therapy, Medical College of Soochow University, PR China
| | - Jin Tao
- Department of Physiology and Neurobiology, Key Laboratory of Pain Research and Therapy, Medical College of Soochow University, PR China
| | - Xinghong Jiang
- Department of Physiology and Neurobiology, Key Laboratory of Pain Research and Therapy, Medical College of Soochow University, PR China.
| |
Collapse
|
20
|
Kerckhove N, Mallet C, François A, Boudes M, Chemin J, Voets T, Bourinet E, Alloui A, Eschalier A. Ca(v)3.2 calcium channels: the key protagonist in the supraspinal effect of paracetamol. Pain 2014; 155:764-772. [PMID: 24447516 DOI: 10.1016/j.pain.2014.01.015] [Citation(s) in RCA: 43] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2013] [Revised: 12/25/2013] [Accepted: 01/14/2014] [Indexed: 12/20/2022]
Abstract
To exert its analgesic action, paracetamol requires complex metabolism to produce a brain-specific lipoamino acid compound, AM404, which targets central transient receptor potential vanilloid receptors (TRPV1). Lipoamino acids are also known to induce analgesia through T-type calcium-channel inhibition (Ca(v)3.2). In this study we show that the antinociceptive effect of paracetamol in mice is lost when supraspinal Ca(v)3.2 channels are inhibited. Therefore, we hypothesized a relationship between supraspinal Ca(v)3.2 and TRPV1, via AM404, which mediates the analgesic effect of paracetamol. AM404 is able to activate TRPV1 and weakly inhibits Ca(v)3.2. Interestingly, activation of TRPV1 induces a strong inhibition of Ca(v)3.2 current. Supporting this, intracerebroventricular administration of AM404 or capsaicin produces antinociception that is lost in Ca(v)3.2(-/-) mice. Our study, for the first time, (1) provides a molecular mechanism for the supraspinal antinociceptive effect of paracetamol; (2) identifies the relationship between TRPV1 and the Ca(v)3.2 channel; and (3) suggests supraspinal Ca(v)3.2 inhibition as a potential pharmacological strategy to alleviate pain.
Collapse
Affiliation(s)
- Nicolas Kerckhove
- Clermont Université, Université d'Auvergne, Pharmacologie Fondamentale et Clinique de la Douleur, 63000 Clermont-Ferrand, France INSERM, U 1107, Neuro-Dol, 63000 Clermont-Ferrand, France CHU Clermont-Ferrand, Service de Pharmacologie, 63003 Clermont-Ferrand, France Laboratories of Excellence, Ion Channel Science and Therapeutics, Institut de Génomique Fonctionnelle, 141 rue de la Cardonille, 34094 Montpellier, France CNRS UMR5203, Montpellier, France INSERM, U661, Montpellier, France IFR3 Universités Montpellier I & II, Montpellier, France Laboratory of Ion Channel Research, Department of Cellular and Molecular Medicine, KU Leuven, Leuven, Belgium
| | | | | | | | | | | | | | | | | |
Collapse
|
21
|
Bailly-Chouriberry L, Cormant F, Garcia P, Kind A, Popot MA, Bonnaire Y. Identification of α-Cobratoxin in Equine Plasma by LC-MS/MS for Doping Control. Anal Chem 2013; 85:5219-25. [DOI: 10.1021/ac4006342] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Affiliation(s)
| | - Florence Cormant
- Laboratoire des Courses Hippiques (LCH), 15 rue de Paradis, 91370 Verrières
le Buisson, France
| | - Patrice Garcia
- Laboratoire des Courses Hippiques (LCH), 15 rue de Paradis, 91370 Verrières
le Buisson, France
| | - Albert Kind
- CYCADS
Laboratory, Iowa State College of Veterinary Medicine, 1600 S 16th Street, Ames, Iowa 50011, United States
| | - Marie-Agnès Popot
- Laboratoire des Courses Hippiques (LCH), 15 rue de Paradis, 91370 Verrières
le Buisson, France
| | - Yves Bonnaire
- Laboratoire des Courses Hippiques (LCH), 15 rue de Paradis, 91370 Verrières
le Buisson, France
| |
Collapse
|
22
|
Modulation of low-voltage-activated T-type Ca²⁺ channels. BIOCHIMICA ET BIOPHYSICA ACTA-BIOMEMBRANES 2012; 1828:1550-9. [PMID: 22975282 DOI: 10.1016/j.bbamem.2012.08.032] [Citation(s) in RCA: 51] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/25/2012] [Revised: 08/29/2012] [Accepted: 08/30/2012] [Indexed: 12/16/2022]
Abstract
Low-voltage-activated T-type Ca²⁺ channels contribute to a wide variety of physiological functions, most predominantly in the nervous, cardiovascular and endocrine systems. Studies have documented the roles of T-type channels in sleep, neuropathic pain, absence epilepsy, cell proliferation and cardiovascular function. Importantly, novel aspects of the modulation of T-type channels have been identified over the last few years, providing new insights into their physiological and pathophysiological roles. Although there is substantial literature regarding modulation of native T-type channels, the underlying molecular mechanisms have only recently begun to be addressed. This review focuses on recent evidence that the Ca(v)3 subunits of T-type channels, Ca(v)3.1, Ca(v)3.2 and Ca(v)3.3, are differentially modulated by a multitude of endogenous ligands including anandamide, monocyte chemoattractant protein-1, endostatin, and redox and oxidizing agents. The review also provides an overview of recent knowledge gained concerning downstream pathways involving G-protein-coupled receptors. This article is part of a Special Issue entitled: Calcium channels.
Collapse
|