1
|
Mai Y, Flechsig J, Warr J, Hummel T. Responses to the activation of different intranasal trigeminal receptors: Evidence from behavioral, peripheral and central levels. Behav Brain Res 2025; 480:115371. [PMID: 39638052 DOI: 10.1016/j.bbr.2024.115371] [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: 09/23/2024] [Revised: 11/18/2024] [Accepted: 12/02/2024] [Indexed: 12/07/2024]
Abstract
AIM There are various receptors that mediate intranasal trigeminal sensations. However, few studies compare the response patterns across different receptor activations. METHODS We recorded negative mucosal potentials (NMPs) in 24 healthy participants and event-related potentials (ERPs) in 17 participants during exposure to five odors that trigger trigeminal sensations and one olfactory stimulus. Additionally, 10 participants completed a continuous odor intensity rating task. RESULTS We observed a significant effect of odor type on NMP amplitudes (F=13.51-21.88, p's < 0.01), with cyclohexanone (TRPV1) and CO2 (TRPV1 +A1) inducing greater N1 and/or P1N1 amplitudes than other stimuli (t = 3.28-7.54, p's < 0.05). Similar differences were seen in ERP amplitudes (F=3.69-12.25, p's < 0.05), with cyclohexanone showing greater P2 and/or N1P2 amplitudes than PEA (odorant), carvacrol (TRPV3 +A1), and perillaldehyde (TRPA1) (t = 3.13-4.10, p's < 0.05). CO2 also produced greater amplitudes than carvacrol (t = 3.53-4.42, p's < 0.05). In the odor intensity rating task, cyclohexanone, CO2, and isopulegol (TRPM8 +TRPA1) had higher peak ratings, steeper slopes, and/or shorter latencies (F=6.15-13.86, p's < 0.01; t = 3.14-7.76, p's < 0.05). CONCLUSIONS Activation of different intranasal trigeminal receptors yields varied responses. Notably, stimuli involving TRPV1 activation, linked to irritation or pain, elicited stronger behavioral and neural activity compared to stimuli involving other receptors, even when controlling for rated stimulus intensity. This emphasizes TRPV1's significance in survival adaptation. Future studies should test different sets of stimulants to verify the robustness of these findings.
Collapse
Affiliation(s)
- Yiling Mai
- Smell and Taste Clinic, Department of Otorhinolaryngology, Faculty of Medicine Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany.
| | - Johanna Flechsig
- Smell and Taste Clinic, Department of Otorhinolaryngology, Faculty of Medicine Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany
| | | | - Thomas Hummel
- Smell and Taste Clinic, Department of Otorhinolaryngology, Faculty of Medicine Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany
| |
Collapse
|
2
|
Ma L, Ma C, Wang Z, Wei Y, Li N, Wang J, Li M, Wu Z, Du Y. Unraveling the Synergistic Neuroprotective Mechanism of Natural Drug Candidates Targeting TRPV1 and TRPM8 on an Ischemic Stroke. Anal Chem 2025. [PMID: 39789730 DOI: 10.1021/acs.analchem.4c04442] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2025]
Abstract
The development of multitargeted drugs is urgent for ischemic stroke. TRPV1 and TRPM8 are important targets of ischemic stroke. Previous drug candidate screening has identified that muscone, l-borneol, and ferulic acid may target TRPV1 and TRPM8 for ischemic stroke. However, the mechanisms of these drug candidates on targets were ill-informed. Therefore, firstly, a tongue-tissue biosensor was constructed. It explored the activation or inhibition mechanisms of drug candidates targeting TRPV1 and TRPM8 in a near-physiological environment. It was found that muscone could specifically inhibit TRPM8 and selectively activate TRPV1, while l-borneol exhibited the opposite effect. It suggested a synergistic network between these two drug candidates. Furthermore, more selective protein biosensors were developed to delve deeper into the synergistic mechanisms. A strong synergistic effect of muscone and l-borneol was proved. Molecular docking revealed that the synergistic effect was caused by different action sites, respectively. Subsequently, the synergistic effect of muscone and l-borneol was further confirmed by hypoxic nerve injury models of Caenorhabditis elegans (C. elegans) and antithrombus and anti-ischemic models of zebrafish. Ultimately, through nontargeted metabolomics, it was found that muscone and l-borneol mainly regulated Ca2+ concentration and energy metabolism by pathways such as purine and amino acid metabolisms. In conclusion, this research identified critical targets and synergistic drug candidates for multitarget neuroprotection of ischemic stroke. In addition, it has systemically demonstrated the feasibility of the integration of tissue/protein biosensors and metabolomics for the research and development of multitarget drugs. Compared to other screening and validation methods for drugs and targets, the biosensors we developed not only achieved higher sensitivity and specificity in complex physiological environments, ensuring a wider detection range, but also greatly saved biological samples. Simultaneously, they could be extended to other complex systems, such as biomarker screening in clinical samples and exosomes isolated from stem cells.
Collapse
Affiliation(s)
- Lijuan Ma
- Kobilka Institute of Innovative Drug Discovery, School of Medicine, The Chinese University of Hong Kong, Shenzhen 518172, China
| | - Chaofu Ma
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing 100029, China
| | - Zijian Wang
- China Beijing Tongrentang Group Co., Ltd., Beijing 100062, China
| | - Yunan Wei
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing 100029, China
| | - Nan Li
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing 100029, China
| | - Jing Wang
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing 100029, China
| | - Mingshuang Li
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing 100029, China
| | - Zhisheng Wu
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing 100029, China
| | - Yang Du
- Kobilka Institute of Innovative Drug Discovery, School of Medicine, The Chinese University of Hong Kong, Shenzhen 518172, China
| |
Collapse
|
3
|
Rodriguez R, Müller S, Colombeau L, Solier S, Sindikubwabo F, Cañeque T. Metal Ion Signaling in Biomedicine. Chem Rev 2025. [PMID: 39746035 DOI: 10.1021/acs.chemrev.4c00577] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2025]
Abstract
Complex multicellular organisms are composed of distinct tissues involving specialized cells that can perform specific functions, making such life forms possible. Species are defined by their genomes, and differences between individuals within a given species directly result from variations in their genetic codes. While genetic alterations can give rise to disease-causing acquisitions of distinct cell identities, it is now well-established that biochemical imbalances within a cell can also lead to cellular dysfunction and diseases. Specifically, nongenetic chemical events orchestrate cell metabolism and transcriptional programs that govern functional cell identity. Thus, imbalances in cell signaling, which broadly defines the conversion of extracellular signals into intracellular biochemical changes, can also contribute to the acquisition of diseased cell states. Metal ions exhibit unique chemical properties that can be exploited by the cell. For instance, metal ions maintain the ionic balance within the cell, coordinate amino acid residues or nucleobases altering folding and function of biomolecules, or directly catalyze specific chemical reactions. Thus, metals are essential cell signaling effectors in normal physiology and disease. Deciphering metal ion signaling is a challenging endeavor that can illuminate pathways to be targeted for therapeutic intervention. Here, we review key cellular processes where metal ions play essential roles and describe how targeting metal ion signaling pathways has been instrumental to dissecting the biochemistry of the cell and how this has led to the development of effective therapeutic strategies.
Collapse
Affiliation(s)
- Raphaël Rodriguez
- Institut Curie, CNRS, INSERM, PSL Research University, 75005 Paris, France
| | - Sebastian Müller
- Institut Curie, CNRS, INSERM, PSL Research University, 75005 Paris, France
| | - Ludovic Colombeau
- Institut Curie, CNRS, INSERM, PSL Research University, 75005 Paris, France
| | - Stéphanie Solier
- Institut Curie, CNRS, INSERM, PSL Research University, 75005 Paris, France
- Université Paris-Saclay, UVSQ, 78180 Montigny-le-Bretonneux, France
| | | | - Tatiana Cañeque
- Institut Curie, CNRS, INSERM, PSL Research University, 75005 Paris, France
| |
Collapse
|
4
|
Binkle-Ladisch L, Pironet A, Zaliani A, Alcouffe C, Mensching D, Haferkamp U, Willing A, Woo MS, Erdmann A, Jessen T, Hess SD, Gribbon P, Pless O, Vennekens R, Friese MA. Identification and development of TRPM4 antagonists to counteract neuronal excitotoxicity. iScience 2024; 27:111425. [PMID: 39687019 PMCID: PMC11648915 DOI: 10.1016/j.isci.2024.111425] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2024] [Revised: 09/21/2024] [Accepted: 11/15/2024] [Indexed: 12/18/2024] Open
Abstract
Neurodegeneration in central nervous system disorders is linked to dysregulated neuronal calcium. Direct inhibition of glutamate-induced neuronal calcium influx, particularly via N-methyl-D-aspartate receptors (NMDAR), has led to adverse effects and clinical trial failures. A more feasible approach is to modulate NMDAR activity or calcium signaling indirectly. In this respect, the calcium-activated non-selective cation channel transient receptor potential melastatin 4 (TRPM4) has been identified as a promising target. However, high affinity and specific antagonists are lacking. Here, we conducted high-throughput screening of a compound library to identify high affinity TRPM4 antagonists. This yielded five lead compound series with nanomolar half-maximal inhibitory concentration values. Through medicinal chemistry optimization of two series, we established detailed structure-activity relationships and inhibition of excitotoxicity in neurons. Moreover, we identified their potential binding site supported by electrophysiological measurements. These potent TRPM4 antagonists are promising drugs for treating neurodegenerative disorders and TRPM4-related pathologies, potentially overcoming previous therapeutic challenges.
Collapse
Affiliation(s)
- Lars Binkle-Ladisch
- Institute of Neuroimmunology and Multiple Sclerosis, University Medical Center Hamburg-Eppendorf, 20251 Hamburg, Germany
| | - Andy Pironet
- Laboratory of Ion Channel Research, Department of Cellular and Molecular Medicine, Katholieke Universiteit Leuven, Campus Gasthuisberg O/N1, Herestraat 49-Bus 802, 3000 Leuven, Belgium
| | - Andrea Zaliani
- Fraunhofer Institute for Translational Medicine and Pharmacology ITMP, 22525 Hamburg, Germany
| | - Chantal Alcouffe
- Department of Chemistry, Evotec SE, 195 Route D'Espagne, 31036 Toulouse, France
| | - Daniel Mensching
- Institute of Neuroimmunology and Multiple Sclerosis, University Medical Center Hamburg-Eppendorf, 20251 Hamburg, Germany
| | - Undine Haferkamp
- Fraunhofer Institute for Translational Medicine and Pharmacology ITMP, 22525 Hamburg, Germany
| | - Anne Willing
- Institute of Neuroimmunology and Multiple Sclerosis, University Medical Center Hamburg-Eppendorf, 20251 Hamburg, Germany
| | - Marcel S. Woo
- Institute of Neuroimmunology and Multiple Sclerosis, University Medical Center Hamburg-Eppendorf, 20251 Hamburg, Germany
| | - Alexandre Erdmann
- Department of Chemistry, Evotec SE, 195 Route D'Espagne, 31036 Toulouse, France
| | | | - Stephen D. Hess
- Evotec Asia Pte Ltd, 79 Science Park Drive, #04-05 Cintech IV, Singapore 118264, Singapore
| | - Philip Gribbon
- Fraunhofer Institute for Translational Medicine and Pharmacology ITMP, 22525 Hamburg, Germany
| | - Ole Pless
- Fraunhofer Institute for Translational Medicine and Pharmacology ITMP, 22525 Hamburg, Germany
| | - Rudi Vennekens
- Laboratory of Ion Channel Research, Department of Cellular and Molecular Medicine, Katholieke Universiteit Leuven, Campus Gasthuisberg O/N1, Herestraat 49-Bus 802, 3000 Leuven, Belgium
| | - Manuel A. Friese
- Institute of Neuroimmunology and Multiple Sclerosis, University Medical Center Hamburg-Eppendorf, 20251 Hamburg, Germany
| |
Collapse
|
5
|
Guo L, Fan W, Li D, Hao Z, Liu P, Liu C, Cui K, Zhang W, Liu X, Zhang Q, Mao J, Xie J. Metabolic pathways, pharmacokinetic, and brain neurochemicals effects of capsaicin: Comprehensively insights from in vivo studies. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2024; 135:156212. [PMID: 39522253 DOI: 10.1016/j.phymed.2024.156212] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/10/2024] [Revised: 10/13/2024] [Accepted: 11/01/2024] [Indexed: 11/16/2024]
Abstract
BACKGROUND Capsaicin (CAP), a prominent component of chili pepper known for its potent agonistic effects on TRPV1, has attracted significant attention for its diverse physiological effects. Nevertheless, there remains a paucity of data concerning its in vivo distribution, metabolism, pharmacodynamic properties, and influence on the metabolic profile of the brain. METHODS Stable isotope tracing, in vitro enzyme incubation, microdialysis coupled with UHPLC-MS/MS techniques were employed to investigate the in vivo metabolic pathways, distribution, and pharmacokinetic properties of CAP, and the potential biases in metabolic pathways was elucidate through molecular docking. Furthermore, the effect of CAP on brain metabolic profiles was assessed using untargeted metabolomics, and spatial visualization analysis was conducted through mass spectrometry imaging. RESULTS CAP was distributed predominantly in the kidneys, with lower content in the liver, heart, lungs, brain, and spleen following peripheral administration, and the absorption half-life in the body was about 20 min. CAP primarily underwent alkyl terminal dehydrogenation, hydroxylation, and macrocyclization metabolic pathways under the action of CYP2C9, CYP2C19 and CYP2D6, resulting in at least four metabolites. Among them, the hydroxylation products were main metabolites and the dehydrogenation product 16,17-dihydrocapsaicin could interact with the key binding sites Leu515 and Thr550 of TRPV1 like CAP. CAP quickly diffused to various brain regions and the metabolic characteristics in the striatum were relatively different from that in the blood. The distribution of CAP in the brain primarily triggered the release of neurotransmitters in areas associated with reward, cognition, and memory. Both acute and chronic exposure to CAP elevated amino acid levels in cortical regions, while producing contrasting effects on nucleotide metabolites. CONCLUSION This study offers an initial in-depth analysis of the distribution patterns, metabolic pathways and pharmacodynamic properties of CAP in the body and brain. These findings established a basis for further studies on CAP's pharmacology properties and its influence on the central nervous system.
Collapse
Affiliation(s)
- Lulu Guo
- Department of Nutrition and Health, Beijing Advanced Innovation Center for Food Nutrition and Human Health, China Agricultural University, Beijing 100080, PR China; Beijing Life Science Academy, Beijing 102209, PR China
| | - Wu Fan
- Beijing Life Science Academy, Beijing 102209, PR China; Flavour Science Research Center, Food Laboratory of Zhongyuan, Zhengzhou University, Zhengzhou 450001, PR China
| | - Die Li
- Flavour Science Research Center, Food Laboratory of Zhongyuan, Zhengzhou University, Zhengzhou 450001, PR China
| | - Zhilin Hao
- Beijing Life Science Academy, Beijing 102209, PR China
| | - Pingping Liu
- Flavour Science Research Center, Food Laboratory of Zhongyuan, Zhengzhou University, Zhengzhou 450001, PR China
| | - Chang Liu
- Beijing Life Science Academy, Beijing 102209, PR China
| | - Kun Cui
- Beijing Life Science Academy, Beijing 102209, PR China
| | - Wenjuan Zhang
- Beijing Life Science Academy, Beijing 102209, PR China
| | - Xingyu Liu
- Beijing Life Science Academy, Beijing 102209, PR China
| | - Qidong Zhang
- Beijing Life Science Academy, Beijing 102209, PR China; Flavour Science Research Center, Food Laboratory of Zhongyuan, Zhengzhou University, Zhengzhou 450001, PR China
| | - Jian Mao
- Beijing Life Science Academy, Beijing 102209, PR China; Flavour Science Research Center, Food Laboratory of Zhongyuan, Zhengzhou University, Zhengzhou 450001, PR China.
| | - Jianping Xie
- Department of Nutrition and Health, Beijing Advanced Innovation Center for Food Nutrition and Human Health, China Agricultural University, Beijing 100080, PR China; Beijing Life Science Academy, Beijing 102209, PR China; Flavour Science Research Center, Food Laboratory of Zhongyuan, Zhengzhou University, Zhengzhou 450001, PR China.
| |
Collapse
|
6
|
Moriyama S, Tatematsu K, Hinuma S, Kuroda S. Role of phosphorylation and vanilloid ligand structure in ligand-dependent differential activations of transient receptor potential vanilloid 1. Biosci Biotechnol Biochem 2024; 88:1316-1325. [PMID: 39217101 DOI: 10.1093/bbb/zbae119] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2024] [Accepted: 08/26/2024] [Indexed: 09/04/2024]
Abstract
Vanilloid analogs, which can activate transient receptor potential vanilloid 1 (TRPV1), have been classified into two types based on susceptibility to forskolin (FSK). Treatment of cells expressing TRPV1 with FSK enhances TRPV1 responses to capsaicin-type ligands while diminishing the responses to eugenol-type ligands. In this study, we determined the effect of FSK on the activation of TRPV1 stimulated with vanilloid ligands, through the influx of Ca2+ in HEK293T cells expressing TRPV1. Our findings suggest that the effects of FSK can be attributed to the phosphorylation of TRPV1, as evidenced by using a protein kinase A inhibitor and TRPV1 mutants at potential phosphorylation sites. Furthermore, we examined the structure-activity relationship of 13 vanilloid analogs. Our results indicated that vanilloid compounds could be classified into three types, that is the previously reported two types and a novel type of 10-shogaol, by which TRPV1 activation was insusceptible to the FSK treatment.
Collapse
Affiliation(s)
- Sakura Moriyama
- SANKEN, Osaka University, Ibaraki, Osaka, Japan
- Graduate School of Frontier Biosciences, Osaka University, Suita, Osaka, Japan
| | | | | | - Shun'ichi Kuroda
- SANKEN, Osaka University, Ibaraki, Osaka, Japan
- Graduate School of Frontier Biosciences, Osaka University, Suita, Osaka, Japan
| |
Collapse
|
7
|
Zhang B, Xie B, Xu W, Wei D, Zhang L, Sun J, Shi Y, Feng J, Yang F, Zhang H, Song X. Inhibition of transient receptor potential vanilloid 3 channels by antimalarial hydroxychloroquine alleviates TRPV3-dependent dermatitis. J Biol Chem 2024; 300:107733. [PMID: 39233228 PMCID: PMC11460631 DOI: 10.1016/j.jbc.2024.107733] [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: 06/08/2024] [Revised: 08/15/2024] [Accepted: 08/18/2024] [Indexed: 09/06/2024] Open
Abstract
Transient receptor potential vanilloid 3 channel (TRPV3) is closely associated with skin inflammation, but there is a lack of effective and specific inhibitors for clinical use. In this study, we identified antimalarial hydroxychloroquine (HCQ) as a selective TRPV3 inhibitor following the prediction by network pharmacology data analysis. In whole-cell patch-clamp recordings, HCQ inhibited the current of the TRPV3 channel, with an IC50 of 51.69 ± 4.78 μM. At the single-channel level, HCQ reduced the open probability of TRPV3 and decreased single-channel conductance. Molecular docking and site-directed mutagenesis confirmed that residues in the pore domain were critical for the activity of HCQ. In vivo, HCQ effectively reduced carvacrol-induced epidermal thickening, erythema, and desquamation. Additionally, the serum immunoglobulin E and inflammatory factors such as tumor necrosis factor-α and interleukin-6 were markedly decreased in the dorsal skin tissues in the HCQ treatment group, as compared to the model group. Our results suggested the antimalarial HCQ may represent a potential alleviator for treating skin inflammation by inhibiting TRPV3 channels.
Collapse
Affiliation(s)
- Beilei Zhang
- Department of Dermatology, Hangzhou Third People's Hospital, Zhejiang Chinese Medical University, Hangzhou, China
| | - Bo Xie
- Department of Dermatology, Hangzhou Third People's Hospital, Affiliated Hangzhou Dermatology Hospital of Zhejiang University School of Medicine, Hangzhou, China
| | - Wen Xu
- Department of Dermatology, Affiliated Hangzhou Dermatology Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Dongfan Wei
- Department of Dermatology, Affiliated Hangzhou Dermatology Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Li Zhang
- Department of Dermatology, Hangzhou Third People's Hospital, Zhejiang Chinese Medical University, Hangzhou, China
| | - Jiayi Sun
- Department of Dermatology, Hangzhou Third People's Hospital, Affiliated Hangzhou Dermatology Hospital of Zhejiang University School of Medicine, Hangzhou, China
| | - Yetan Shi
- Department of Dermatology, Hangzhou Third People's Hospital, Zhejiang Chinese Medical University, Hangzhou, China
| | - Jiangfeng Feng
- Department of Dermatology, Hangzhou Third People's Hospital, Zhejiang Chinese Medical University, Hangzhou, China
| | - Fan Yang
- Department of Biophysics, Kidney Disease Center of the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China; Liangzhu Laboratory, Zhejiang University Medical Center, Hangzhou, Zhejiang, China.
| | - Heng Zhang
- Department of Biophysics, Kidney Disease Center of the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China; Liangzhu Laboratory, Zhejiang University Medical Center, Hangzhou, Zhejiang, China.
| | - Xiuzu Song
- Department of Dermatology, Hangzhou Third People's Hospital, Affiliated Hangzhou Dermatology Hospital of Zhejiang University School of Medicine, Hangzhou, China.
| |
Collapse
|
8
|
Gualdani R, Barbeau S, Yuan JH, Jacobs DS, Gailly P, Dib-Hajj SD, Waxman SG. TRPV1 corneal neuralgia mutation: Enhanced pH response, bradykinin sensitization, and capsaicin desensitization. Proc Natl Acad Sci U S A 2024; 121:e2406186121. [PMID: 39226353 PMCID: PMC11406256 DOI: 10.1073/pnas.2406186121] [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: 03/26/2024] [Accepted: 07/30/2024] [Indexed: 09/05/2024] Open
Abstract
The factors that contribute to pain after nerve injury remain incompletely understood. Laser-assisted in situ keratomileusis (LASIK) and photorefractive keratectomy (PRK) are common surgical techniques to correct refractive errors. After LASIK or PRK, a subset of patients suffers intense and persistent pain, of unknown origin, described by patients as feeling like shards of glass in their eye. Here, we evaluated a TRPV1 variant, p.V527M, found in a 49-y-old woman who developed corneal pain after LASIK and subsequent PRK enhancement, reporting an Ocular Surface Disease Index score of 100. Using patch-clamp and Ca2+ imaging, we found that the V527M mutation enhances the response to acidic pH. Increasing proton concentration induced a stronger leftward shift in the activation curve of V527M compared to WT, resulting in channel activity of the mutant in acidic pH at more physiological membrane potentials. Finally, comparing the responses to consecutive applications of different agonists, we found in V527M channels a reduced capsaicin-induced desensitization and increased sensitization by the arachidonic acid metabolite 12-hydroxyeicosatetraenoic acid (12-HETE). We hypothesize that the increased response in V527M channels to protons and enhanced sensitization by 12-HETE, two inflammatory mediators released in the cornea after tissue damage, may contribute to the pathogenesis of corneal neuralgia after refractive surgery.
Collapse
Affiliation(s)
- Roberta Gualdani
- Laboratory of Cell Physiology, Institute of Neuroscience, Université catholique de Louvain, BrusselsB-1200, Belgium
| | - Solène Barbeau
- Laboratory of Cell Physiology, Institute of Neuroscience, Université catholique de Louvain, BrusselsB-1200, Belgium
| | - Jun-Hui Yuan
- Department of Neurology, Yale School of Medicine, New Haven, CT06520
- Center for Neuroscience and Regeneration Research, Yale School of Medicine, New Haven, CT06520
- Neurorehabilitation Research Center, Veterans Affairs Medical Center, West Haven, CT06516
| | - Deborah S. Jacobs
- Department of Ophthalmology, Massachusetts Eye and Ear, Boston, MA02114
| | - Philippe Gailly
- Laboratory of Cell Physiology, Institute of Neuroscience, Université catholique de Louvain, BrusselsB-1200, Belgium
| | - Sulayman D. Dib-Hajj
- Department of Neurology, Yale School of Medicine, New Haven, CT06520
- Center for Neuroscience and Regeneration Research, Yale School of Medicine, New Haven, CT06520
- Neurorehabilitation Research Center, Veterans Affairs Medical Center, West Haven, CT06516
| | - Stephen G. Waxman
- Department of Neurology, Yale School of Medicine, New Haven, CT06520
- Center for Neuroscience and Regeneration Research, Yale School of Medicine, New Haven, CT06520
- Neurorehabilitation Research Center, Veterans Affairs Medical Center, West Haven, CT06516
| |
Collapse
|
9
|
Wang S, Chen Y, Lu Y, Jiang D, Lin H, Jiang Z, Tang J, Dong W, Zhao J. Interaction of pepper numbing substances with myofibrillar proteins and numbness perception under thermal conditions: A structural mechanism analysis. Food Chem 2024; 449:139203. [PMID: 38599105 DOI: 10.1016/j.foodchem.2024.139203] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2024] [Revised: 03/18/2024] [Accepted: 03/29/2024] [Indexed: 04/12/2024]
Abstract
This study examined the interaction between myofibrillar proteins (MPs) and the numbing substance hydroxy-α-sanshool (α-SOH) in a thermal environment, and provided an explanation of the numbness perception mechanism through muti-spectroscopic and molecular dynamics simulation methodology. Results showed that addition of α-SOH could reduce the particle size and molecular weight of MPs, accompanied by changes in the tertiary and secondary structure, causing the α-helix of MPs transitioned to β-sheet and β-turn due to the reorganization of hydrogen bonds. After a moderate heating (60 or 70 °C), MPs could form the stable complexes with α-SOH that were associated with attachment sites and protein wrapping. The thermal process might convert a portion of α-SOH' into hydroxy-β-sanshool' (β-SOH'). When docking with the sensory receptor TRPV1, the RMSD, RMSF and binding free energy all showed that β-SOH' demonstrated a low affinity, thereby reducing the numbing perception. These findings can provide a theoretical foundation for the advanced processing of numbing meat products.
Collapse
Affiliation(s)
- Shuaiqian Wang
- School of Food and Bioengineering, Xihua University, Chengdu 610039, China
| | - Yu Chen
- School of Food and Bioengineering, Xihua University, Chengdu 610039, China
| | - Yan Lu
- School of Food and Bioengineering, Xihua University, Chengdu 610039, China
| | - Diandian Jiang
- School of Food and Bioengineering, Xihua University, Chengdu 610039, China
| | - Hongbin Lin
- School of Food and Bioengineering, Xihua University, Chengdu 610039, China; Chongqing Key Laboratory of Specialty Food Co-Built by Sichuan and Chongqing, Chengdu 610039, China
| | - Zhenju Jiang
- School of Food and Bioengineering, Xihua University, Chengdu 610039, China; Chongqing Key Laboratory of Specialty Food Co-Built by Sichuan and Chongqing, Chengdu 610039, China
| | - Jie Tang
- School of Food and Bioengineering, Xihua University, Chengdu 610039, China; Chongqing Key Laboratory of Specialty Food Co-Built by Sichuan and Chongqing, Chengdu 610039, China
| | - Wei Dong
- School of Food and Bioengineering, Xihua University, Chengdu 610039, China; Beijing Laboratory of Food Quality and Safety/Key Laboratory of Alcoholic Beverages Quality and Safety of China Light Industry, Beijing Technology and Business University, Beijing 100048, China
| | - Jie Zhao
- School of Food and Bioengineering, Xihua University, Chengdu 610039, China; Chongqing Key Laboratory of Specialty Food Co-Built by Sichuan and Chongqing, Chengdu 610039, China.
| |
Collapse
|
10
|
Fan J, Ke H, Lei J, Wang J, Tominaga M, Lei X. Structural basis of TRPV1 inhibition by SAF312 and cholesterol. Nat Commun 2024; 15:6689. [PMID: 39107321 PMCID: PMC11303535 DOI: 10.1038/s41467-024-51085-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2023] [Accepted: 07/29/2024] [Indexed: 08/10/2024] Open
Abstract
Transient Receptor Potential Vanilloid 1 (TRPV1) plays a central role in pain sensation and is thus an attractive pharmacological drug target. SAF312 is a potent, selective, and non-competitive antagonist of TRPV1 and shows promising potential in treating ocular surface pain. However, the precise mechanism by which SAF312 inhibits TRPV1 remains poorly understood. Here, we present the cryo-EM structure of human TRPV1 in complex with SAF312, elucidating the structural foundation of its antagonistic effects on TRPV1. SAF312 binds to the vanilloid binding pocket, preventing conformational changes in S4 and S5 helices, which are essential for channel gating. Unexpectedly, a putative cholesterol was found to contribute to SAF312's inhibition. Complemented by mutagenesis experiments and molecular dynamics simulations, our research offers substantial mechanistic insights into the regulation of TRPV1 by SAF312, highlighting the interplay between the antagonist and cholesterol in modulating TRPV1 function. This work not only expands our understanding of TRPV1 inhibition by SAF312 but also lays the groundwork for further developments in the design and optimization of TRPV1-related therapies.
Collapse
Affiliation(s)
- Junping Fan
- Beijing National Laboratory for Molecular Sciences, Key Laboratory of Bioorganic Chemistry and Molecular Engineering of Ministry of Education, Institute of Organic Chemistry, College of Chemistry and Molecular Engineering, Peking University, Beijing, 100871, China.
| | - Han Ke
- Beijing National Laboratory for Molecular Sciences, Key Laboratory of Bioorganic Chemistry and Molecular Engineering of Ministry of Education, Institute of Organic Chemistry, College of Chemistry and Molecular Engineering, Peking University, Beijing, 100871, China
| | - Jing Lei
- Division of Cell Signaling, National Institute for Physiological Sciences, National Institutes of Natural Sciences, Okazaki, 444-8787, Japan
- Thermal Biology Group, Exploratory Research Center on Life and Living Systems, National Institutes of Natural Sciences, Okazaki, 444-8787, Japan
| | - Jin Wang
- Beijing National Laboratory for Molecular Sciences, Key Laboratory of Bioorganic Chemistry and Molecular Engineering of Ministry of Education, Institute of Organic Chemistry, College of Chemistry and Molecular Engineering, Peking University, Beijing, 100871, China
| | - Makoto Tominaga
- Division of Cell Signaling, National Institute for Physiological Sciences, National Institutes of Natural Sciences, Okazaki, 444-8787, Japan
- Thermal Biology Group, Exploratory Research Center on Life and Living Systems, National Institutes of Natural Sciences, Okazaki, 444-8787, Japan
- Thermal Biology Research Group, Nagoya Advanced Research and Development Center, Nagoya City University, Nagoya, 467-8601, Japan
| | - Xiaoguang Lei
- Beijing National Laboratory for Molecular Sciences, Key Laboratory of Bioorganic Chemistry and Molecular Engineering of Ministry of Education, Institute of Organic Chemistry, College of Chemistry and Molecular Engineering, Peking University, Beijing, 100871, China.
- Peking-Tsinghua Center for Life Sciences, Peking University, Beijing, 100871, China.
- Institute for Cancer Research, Shenzhen Bay Laboratory, Shenzhen, 518107, China.
| |
Collapse
|
11
|
Moriyama S, Takita Y, Hinuma S, Kuroda S. Divergent effects of olfactory receptors on transient receptor potential vanilloid 1 activation by capsaicin and eugenol. Biosci Biotechnol Biochem 2024; 88:908-917. [PMID: 38734894 DOI: 10.1093/bbb/zbae060] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2024] [Accepted: 04/30/2024] [Indexed: 05/13/2024]
Abstract
We analyzed the effects of olfactory receptors (ORs) on transient receptor potential vanilloid 1 (TRPV1) activation using HEK293T cells co-expressing TRPV1 and OR51E1. We demonstrate here that the effect of OR51E1 on TRPV1 activation varies depending on the two TRPV1 ligands: capsaicin and eugenol. Notably, both of these ligands are vanilloid analogs. OR51E1 enhanced the response of TRPV1 to capsaicin but diminished that to eugenol. OR51E2 also showed similar effects. Based on the susceptibility to the OR's modulatory effects, various TRPV1 ligands could be classified into capsaicin and eugenol types. Activation of OR51E1 enhanced cAMP production. In addition, forskolin exhibited almost identical effects as ORs on TRPV1 responses to both types of ligands. These results suggest that OR51E1-induced cAMP elevation leads to a modification of TRPV1, presumably phosphorylation of TRPV1, which amplifies the susceptibility of TRPV1 to the two types of ligands differently.
Collapse
Affiliation(s)
- Sakura Moriyama
- SANKEN, Osaka University, Mihogaoka 8-1, Ibaraki, Osaka, Japan
- Graduate School of Frontier Biosciences, Osaka University, Yamadaoka 1-3, Suita, Osaka, Japan
| | - Yukie Takita
- SANKEN, Osaka University, Mihogaoka 8-1, Ibaraki, Osaka, Japan
- Graduate School of Frontier Biosciences, Osaka University, Yamadaoka 1-3, Suita, Osaka, Japan
| | - Shuji Hinuma
- SANKEN, Osaka University, Mihogaoka 8-1, Ibaraki, Osaka, Japan
| | - Shun'ichi Kuroda
- SANKEN, Osaka University, Mihogaoka 8-1, Ibaraki, Osaka, Japan
- Graduate School of Frontier Biosciences, Osaka University, Yamadaoka 1-3, Suita, Osaka, Japan
| |
Collapse
|
12
|
Li S, Zheng J. How much does TRPV1 deviate from an ideal MWC-type protein? Biophys J 2024; 123:2136-2144. [PMID: 38582967 PMCID: PMC11309965 DOI: 10.1016/j.bpj.2024.04.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2023] [Revised: 12/06/2023] [Accepted: 04/03/2024] [Indexed: 04/08/2024] Open
Abstract
Many ion channels are known to behave as an allosteric protein, coupling environmental stimuli captured by specialized sensing domains to the opening of a central pore. The classic Monod-Wyman-Changeux (MWC) model, originally proposed to describe binding of gas molecules to hemoglobin, has been widely used as a framework for analyzing ion channel gating. Here, we address the issue of how accurately the MWC model predicts activation of the capsaicin receptor TRPV1 by vanilloids. Taking advantage of a concatemeric design that makes it possible to lock TRPV1 in states with zero to four bound vanilloid molecules, we showed quantitatively that the overall gating behavior is satisfactorily predicted by the MWC model. There is, however, a small yet detectable subunit position effect: ligand binding to two kitty-corner subunits is 0.3-0.4 kcal/mol more effective in inducing opening than binding to two neighbor subunits. This difference-less than 10% of the overall energetic contribution from ligand binding-might be due to the restriction on subunit arrangement imposed by the planar membrane; if this is the case, then the position effect is not expected in hemoglobin, in which each subunit is related equivalently to all the other subunits.
Collapse
Affiliation(s)
- Shisheng Li
- Department of Physiology and Membrane Biology, University of California at Davis, School of Medicine, Davis, California
| | - Jie Zheng
- Department of Physiology and Membrane Biology, University of California at Davis, School of Medicine, Davis, California.
| |
Collapse
|
13
|
Ren C, Ma Y, Wang Y, Luo D, Hong Y, Zhang X, Mei H, Liu W. Palmitoylethanolamide-Incorporated Elastic Nano-Liposomes for Enhanced Transdermal Delivery and Anti-Inflammation. Pharmaceutics 2024; 16:876. [PMID: 39065574 PMCID: PMC11280357 DOI: 10.3390/pharmaceutics16070876] [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: 05/12/2024] [Revised: 06/24/2024] [Accepted: 06/26/2024] [Indexed: 07/28/2024] Open
Abstract
Palmitoylethanolamide (PEA) exhibits multiple skincare functions such as anti-nociceptive and anti-inflammatory effects. However, its topical application is limited due to its difficulty in bypassing the stratum corneum barrier, relatively low bioavailability, and low stability. Herein, elastic nano-liposomes (ENLs) with excellent deformability and elasticity were utilized as a novel drug delivery system to encapsulate PEA to overcome the abovementioned issues and enhance the biological effects on the skin. ENL was prepared with phosphatidylcholine, cholesterol, and cetyl-PG hydroxyethyl palmitamide with a molar ratio mimicking skin epidermal lipids, and PEA was loaded. The PEA-loaded ENL (PEA-ENL) demonstrated efficient transdermal delivery and enhanced skin retention, with negligible cytotoxicity toward HaCaT cells and no allergic reaction in the human skin patch test. Notably, PEA-ENL treatment increased cell migration and induced significant regulation in the expression of genes associated with anti-nociceptive, anti-inflammatory, and skin barrier repair. The mechanism of the anti-nociceptive and anti-inflammatory effects of PEA was further investigated and explained by molecular docking site analysis. This novel PEA-ENL, with efficient transdermal delivery efficiency and multiple skincare functionalities, is promising for topical application.
Collapse
Affiliation(s)
- Chuanpeng Ren
- The Institute of Biocelline Precision Dermatology, Shanghai 200031, China; (Y.W.); (H.M.)
| | - Yanyun Ma
- Human Phenome Institute, Fudan University, Shanghai 201210, China;
- Institute for Six-Sector Economy, Fudan University, Shanghai 201203, China
| | - Yizhen Wang
- The Institute of Biocelline Precision Dermatology, Shanghai 200031, China; (Y.W.); (H.M.)
| | - Dan Luo
- Wuhan Bestcarrier Biotechnology Co., Ltd., Wuhan 430075, China; (D.L.); (Y.H.)
| | - Yanhan Hong
- Wuhan Bestcarrier Biotechnology Co., Ltd., Wuhan 430075, China; (D.L.); (Y.H.)
| | - Xinyuan Zhang
- Shanghai Skinshield Clinical Testing and Technological Research Ltd., Shanghai 201210, China;
| | - Hexiang Mei
- The Institute of Biocelline Precision Dermatology, Shanghai 200031, China; (Y.W.); (H.M.)
| | - Wei Liu
- National Engineering Research Center for Nanomedicine, Huazhong University of Science and Technology, Wuhan 430074, China
| |
Collapse
|
14
|
Harris BJ, Nguyen PT, Zhou G, Wulff H, DiMaio F, Yarov-Yarovoy V. Toward high-resolution modeling of small molecule-ion channel interactions. Front Pharmacol 2024; 15:1411428. [PMID: 38919257 PMCID: PMC11196768 DOI: 10.3389/fphar.2024.1411428] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2024] [Accepted: 05/13/2024] [Indexed: 06/27/2024] Open
Abstract
Ion channels are critical drug targets for a range of pathologies, such as epilepsy, pain, itch, autoimmunity, and cardiac arrhythmias. To develop effective and safe therapeutics, it is necessary to design small molecules with high potency and selectivity for specific ion channel subtypes. There has been increasing implementation of structure-guided drug design for the development of small molecules targeting ion channels. We evaluated the performance of two RosettaLigand docking methods, RosettaLigand and GALigandDock, on the structures of known ligand-cation channel complexes. Ligands were docked to voltage-gated sodium (NaV), voltage-gated calcium (CaV), and transient receptor potential vanilloid (TRPV) channel families. For each test case, RosettaLigand and GALigandDock methods frequently sampled a ligand-binding pose within a root mean square deviation (RMSD) of 1-2 Å relative to the experimental ligand coordinates. However, RosettaLigand and GALigandDock scoring functions cannot consistently identify experimental ligand coordinates as top-scoring models. Our study reveals that the proper scoring criteria for RosettaLigand and GALigandDock modeling of ligand-ion channel complexes should be assessed on a case-by-case basis using sufficient ligand and receptor interface sampling, knowledge about state-specific interactions of the ion channel, and inherent receptor site flexibility that could influence ligand binding.
Collapse
Affiliation(s)
- Brandon J. Harris
- Department of Physiology and Membrane Biology, University of California, Davis, Davis, CA, United States
- Biophysics Graduate Group, University of California, Davis, Davis, CA, United States
| | - Phuong T. Nguyen
- Department of Physiology and Membrane Biology, University of California, Davis, Davis, CA, United States
| | - Guangfeng Zhou
- Department of Biochemistry, University of Washington, Seattle, WA, United States
- Institute for Protein Design, University of Washington, Seattle, WA, United States
| | - Heike Wulff
- Department of Pharmacology, School of Medicine, University of California, Davis, Davis, CA, United States
| | - Frank DiMaio
- Department of Biochemistry, University of Washington, Seattle, WA, United States
| | - Vladimir Yarov-Yarovoy
- Department of Physiology and Membrane Biology, University of California, Davis, Davis, CA, United States
- Biophysics Graduate Group, University of California, Davis, Davis, CA, United States
- Department of Anesthesiology and Pain Medicine, University of California, Davis, Davis, CA, United States
| |
Collapse
|
15
|
Su T, Chen YH, Wu KK, Xu XH. Anti-cancer agent piperlongumine is an inhibitor of transient receptor potential melastatin 7 channel in oral squamous cell carcinoma. J Oral Biosci 2024; 66:430-438. [PMID: 38452870 DOI: 10.1016/j.job.2024.03.002] [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: 11/30/2023] [Revised: 03/04/2024] [Accepted: 03/05/2024] [Indexed: 03/09/2024]
Abstract
OBJECTIVES To elucidate the association between the anticancer activities of piperlongumine (PL) and its potential target, transient receptor potential melastatin 7 channel (TRPM7), in oral squamous cell carcinoma (OSCC). METHODS The expression levels and electrical characteristics of TRPM7 as well as cell viability in response to various PL treatments were investigated in the OSCC cell line Cal27. RESULTS PL treatment resulted in a concentration- and time-dependent reduction in TRPM7 mRNA and protein expression in Cal27 cells. Furthermore, PL treatment inhibited TRPM7-like rectifying currents in Cal27 cells; however, this inhibition was less effective than that of the TRPM7 antagonist waixenicin A. Rapid perfusion and washout experiments revealed an immediate inhibitory effect of PL on TRPM7-like currents. The antagonistic effect of PL occurred within 1 min and was not completely reversed following washout. Notably, the extracellular Ca2+ concentration still influenced PL-induced changes in the TRPM7-like current, indicating that PL can directly but gently antagonize the TRPM7 channel. Functional changes in TRPM7 correlated with the observed antiproliferative and cytotoxic effects of PL in Cal27 cells. CONCLUSIONS These findings suggest that PL exhibits potent inhibitory effects on TRPM7 and exerts its anti-cancer effects by downregulating TRPM7 expression and antagonizing channel currents.
Collapse
Affiliation(s)
- Tao Su
- Department of Stomatology, the Second Affiliated Hospital of Guangzhou Medical University, Guangzhou, China; Institute of Neuroscience, the Second Affiliated Hospital of Guangzhou Medical University, Guangzhou, China; Key Laboratory of Neurogenetics and Channelopathies of the Ministry of Education of China, Guangzhou, China
| | - Yi-Hui Chen
- Department of Prevention and Health-Care, the Second Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Kan-Kui Wu
- Department of Stomatology, the Second Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Xiao-Hong Xu
- Department of Stomatology, the Second Affiliated Hospital of Guangzhou Medical University, Guangzhou, China; Key Laboratory of Neurogenetics and Channelopathies of the Ministry of Education of China, Guangzhou, China.
| |
Collapse
|
16
|
Maximiano TKE, Carneiro JA, Fattori V, Verri WA. TRPV1: Receptor structure, activation, modulation and role in neuro-immune interactions and pain. Cell Calcium 2024; 119:102870. [PMID: 38531262 DOI: 10.1016/j.ceca.2024.102870] [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: 11/30/2023] [Revised: 03/06/2024] [Accepted: 03/06/2024] [Indexed: 03/28/2024]
Abstract
In the 1990s, the identification of a non-selective ion channel, especially responsive to capsaicin, revolutionized the studies of somatosensation and pain that were to follow. The TRPV1 channel is expressed mainly in neuronal cells, more specifically, in sensory neurons responsible for the perception of noxious stimuli. However, its presence has also been detected in other non-neuronal cells, such as immune cells, β- pancreatic cells, muscle cells and adipocytes. Activation of the channel occurs in response to a wide range of stimuli, such as noxious heat, low pH, gasses, toxins, endocannabinoids, lipid-derived endovanilloid, and chemical agents, such as capsaicin and resiniferatoxin. This activation results in an influx of cations through the channel pore, especially calcium. Intracellular calcium triggers different responses in sensory neurons. Dephosphorylation of the TRPV1 channel leads to its desensitization, which disrupts its function, while its phosphorylation increases the channel's sensitization and contributes to the channel's rehabilitation after desensitization. Kinases, phosphoinositides, and calmodulin are the main signaling pathways responsible for the channel's regulation. Thus, in this review we provide an overview of TRPV1 discovery, its tissue expression as well as on the mechanisms by which TRPV1 activation (directly or indirectly) induces pain in different disease models.
Collapse
Affiliation(s)
- Thaila Kawane Euflazio Maximiano
- Laboratory of Pain, Inflammation, Neuropathy, and Cancer, Department of Pathology, Center of Biological Sciences, Londrina State University, Londrina, Paraná, Brazil
| | - Jessica Aparecida Carneiro
- Laboratory of Pain, Inflammation, Neuropathy, and Cancer, Department of Pathology, Center of Biological Sciences, Londrina State University, Londrina, Paraná, Brazil
| | - Victor Fattori
- Vascular Biology Program, Department of Surgery, Boston Children's Hospital-Harvard Medical School, Karp Research Building, 300 Longwood Ave, 02115, Boston, Massachusetts, United States.
| | - Waldiceu A Verri
- Laboratory of Pain, Inflammation, Neuropathy, and Cancer, Department of Pathology, Center of Biological Sciences, Londrina State University, Londrina, Paraná, Brazil.
| |
Collapse
|
17
|
Zhang H, Wang J, Yang F. Design, synthesis and functional validation of peptide inhibitors based on TRPV1 ion channel agonist RhTx. Zhejiang Da Xue Xue Bao Yi Xue Ban 2024; 53:201-207. [PMID: 38501280 PMCID: PMC11057985 DOI: 10.3724/zdxbyxb-2023-0465] [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: 09/27/2023] [Accepted: 03/12/2024] [Indexed: 03/20/2024]
Abstract
OBJECTIVES To design and synthesize peptide inhibitors targeting transient receptor potential vanilloid 1 (TRPV1) ion channel, and to validate their function. METHODS Based on previous studies on the relation of molecular structure and function of red head toxin (RhTx), a series of peptides were rationally designed and synthesized, with positive charged amino acids linked to the N terminus of RhTx. These Nplus-RhTx peptides were functionally validated by patch-clamp recordings in live cells. RESULTS Among the 8 synthesized Nplus-RhTx peptides, four inhibited TRPV1 ion channel activated by capsaicin with IC50 of (188.3±4.7), (193.6±18.0), (282.8±11.9) and (299.5±6.4) µmol/L, respectively. CONCLUSIONS It is feasible to develop TRPV1 peptide inhibitors by using rational design based on N terminal residues of RhTx.
Collapse
Affiliation(s)
- Heng Zhang
- Department of Biophysics, School of Basic Medical Sciences, Zhejiang University School of Medicine, Hangzhou 310058, China.
| | - Jiawei Wang
- Department of Biophysics, School of Basic Medical Sciences, Zhejiang University School of Medicine, Hangzhou 310058, China
| | - Fan Yang
- Department of Biophysics, School of Basic Medical Sciences, Zhejiang University School of Medicine, Hangzhou 310058, China.
| |
Collapse
|
18
|
Amaya-Rodriguez CA, Carvajal-Zamorano K, Bustos D, Alegría-Arcos M, Castillo K. A journey from molecule to physiology and in silico tools for drug discovery targeting the transient receptor potential vanilloid type 1 (TRPV1) channel. Front Pharmacol 2024; 14:1251061. [PMID: 38328578 PMCID: PMC10847257 DOI: 10.3389/fphar.2023.1251061] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2023] [Accepted: 12/14/2023] [Indexed: 02/09/2024] Open
Abstract
The heat and capsaicin receptor TRPV1 channel is widely expressed in nerve terminals of dorsal root ganglia (DRGs) and trigeminal ganglia innervating the body and face, respectively, as well as in other tissues and organs including central nervous system. The TRPV1 channel is a versatile receptor that detects harmful heat, pain, and various internal and external ligands. Hence, it operates as a polymodal sensory channel. Many pathological conditions including neuroinflammation, cancer, psychiatric disorders, and pathological pain, are linked to the abnormal functioning of the TRPV1 in peripheral tissues. Intense biomedical research is underway to discover compounds that can modulate the channel and provide pain relief. The molecular mechanisms underlying temperature sensing remain largely unknown, although they are closely linked to pain transduction. Prolonged exposure to capsaicin generates analgesia, hence numerous capsaicin analogs have been developed to discover efficient analgesics for pain relief. The emergence of in silico tools offered significant techniques for molecular modeling and machine learning algorithms to indentify druggable sites in the channel and for repositioning of current drugs aimed at TRPV1. Here we recapitulate the physiological and pathophysiological functions of the TRPV1 channel, including structural models obtained through cryo-EM, pharmacological compounds tested on TRPV1, and the in silico tools for drug discovery and repositioning.
Collapse
Affiliation(s)
- Cesar A. Amaya-Rodriguez
- Centro Interdisciplinario de Neurociencia de Valparaíso, Facultad de Ciencias, Universidad de Valparaíso, Valparaíso, Chile
- Departamento de Fisiología y Comportamiento Animal, Facultad de Ciencias Naturales, Exactas y Tecnología, Universidad de Panamá, Ciudad de Panamá, Panamá
| | - Karina Carvajal-Zamorano
- Centro Interdisciplinario de Neurociencia de Valparaíso, Facultad de Ciencias, Universidad de Valparaíso, Valparaíso, Chile
| | - Daniel Bustos
- Centro de Investigación de Estudios Avanzados del Maule (CIEAM), Vicerrectoría de Investigación y Postgrado Universidad Católica del Maule, Talca, Chile
- Laboratorio de Bioinformática y Química Computacional, Departamento de Medicina Traslacional, Facultad de Medicina, Universidad Católica del Maule, Talca, Chile
| | - Melissa Alegría-Arcos
- Núcleo de Investigación en Data Science, Facultad de Ingeniería y Negocios, Universidad de las Américas, Santiago, Chile
| | - Karen Castillo
- Centro Interdisciplinario de Neurociencia de Valparaíso, Facultad de Ciencias, Universidad de Valparaíso, Valparaíso, Chile
- Centro de Investigación de Estudios Avanzados del Maule (CIEAM), Vicerrectoría de Investigación y Postgrado Universidad Católica del Maule, Talca, Chile
| |
Collapse
|
19
|
Musolino M, D’Agostino M, Zicarelli M, Andreucci M, Coppolino G, Bolignano D. Spice Up Your Kidney: A Review on the Effects of Capsaicin in Renal Physiology and Disease. Int J Mol Sci 2024; 25:791. [PMID: 38255865 PMCID: PMC10815060 DOI: 10.3390/ijms25020791] [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: 12/10/2023] [Revised: 12/31/2023] [Accepted: 01/06/2024] [Indexed: 01/24/2024] Open
Abstract
Capsaicin, the organic compound which attributes the spicy flavor and taste of red peppers and chili peppers, has been extensively studied for centuries as a potential natural remedy for the treatment of several illnesses. Indeed, this compound exerts well-known systemic pleiotropic effects and may thus bring important benefits against various pathological conditions like neuropathic pain, rhinitis, itching, or chronic inflammation. Yet, little is known about the possible biological activity of capsaicin at the kidney level, as this aspect has only been addressed by sparse experimental investigations. In this paper, we aimed to review the available evidence focusing specifically on the effects of capsaicin on renal physiology, as well as its potential benefits for the treatment of various kidney disorders. Capsaicin may indeed modulate various aspects of renal function and renal nervous activity. On the other hand, the observed experimental benefits in preventing acute kidney injury, slowing down the progression of diabetic and chronic kidney disease, ameliorating hypertension, and even delaying renal cancer growth may set the stage for future human trials of capsaicin administration as an adjuvant or preventive therapy for different, difficult-to-treat renal diseases.
Collapse
Affiliation(s)
- Michela Musolino
- Nephrology and Dialysis Unit, Magna Graecia University Hospital, 88100 Catanzaro, Italy; (M.M.); (M.D.); (M.A.); (G.C.)
- Department of Health Sciences, Magna Graecia University, 88100 Catanzaro, Italy;
| | - Mario D’Agostino
- Nephrology and Dialysis Unit, Magna Graecia University Hospital, 88100 Catanzaro, Italy; (M.M.); (M.D.); (M.A.); (G.C.)
| | | | - Michele Andreucci
- Nephrology and Dialysis Unit, Magna Graecia University Hospital, 88100 Catanzaro, Italy; (M.M.); (M.D.); (M.A.); (G.C.)
- Department of Health Sciences, Magna Graecia University, 88100 Catanzaro, Italy;
| | - Giuseppe Coppolino
- Nephrology and Dialysis Unit, Magna Graecia University Hospital, 88100 Catanzaro, Italy; (M.M.); (M.D.); (M.A.); (G.C.)
- Department of Health Sciences, Magna Graecia University, 88100 Catanzaro, Italy;
| | - Davide Bolignano
- Nephrology and Dialysis Unit, Magna Graecia University Hospital, 88100 Catanzaro, Italy; (M.M.); (M.D.); (M.A.); (G.C.)
- Department of Medical and Surgical Sciences, Magna Graecia University, 88100 Catanzaro, Italy
| |
Collapse
|
20
|
Wei X, Huang T, Yang Z, Pan L, Wang L, Ding J. Quantitative Predictive Studies of Multiple Biological Activities of TRPV1 Modulators. Molecules 2024; 29:295. [PMID: 38257208 PMCID: PMC10820055 DOI: 10.3390/molecules29020295] [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: 12/03/2023] [Revised: 12/25/2023] [Accepted: 12/27/2023] [Indexed: 01/24/2024] Open
Abstract
TRPV1 channel agonists and antagonists, which have powerful analgesic effects without the addictive qualities associated with traditional analgesics, have become a focus area for the development of novel analgesics. In this study, quantitative structure-activity relationship (QSAR) models for three bioactive endpoints (Ki, IC50, and EC50) were successfully constructed using four machine learning algorithms: SVM, Bagging, GBDT, and XGBoost. These models were based on 2922 TRPV1 modulators and incorporated four types of molecular descriptors: Daylight, E-state, ECFP4, and MACCS. After the rigorous five-fold cross-validation and external test set validation, the optimal models for the three endpoints were obtained. For the Ki endpoint, the Bagging-ECFP4 model had a Q2 value of 0.778 and an R2 value of 0.780. For the IC50 endpoint, the XGBoost-ECFP4 model had a Q2 value of 0.806 and an R2 value of 0.784. For the EC50 endpoint, the SVM-Daylight model had a Q2 value of 0.784 and an R2 value of 0.809. These results demonstrate that the constructed models exhibit good predictive performance. In addition, based on the model feature importance analysis, the influence between substructure and biological activity was also explored, which can provide important theoretical guidance for the efficient virtual screening and structural optimization of novel TRPV1 analgesics. And subsequent studies on novel TRPV1 modulators will be based on the feature substructures of the three endpoints.
Collapse
Affiliation(s)
- Xinmiao Wei
- State Key Laboratory of NBC Protection for Civilian, Beijing 102205, China; (X.W.); (T.H.); (Z.Y.); (L.P.)
| | - Tengxin Huang
- State Key Laboratory of NBC Protection for Civilian, Beijing 102205, China; (X.W.); (T.H.); (Z.Y.); (L.P.)
- School of Physics and Electronic Engineering, Sichuan University of Science & Engineering, Zigong 643000, China
| | - Zhijiang Yang
- State Key Laboratory of NBC Protection for Civilian, Beijing 102205, China; (X.W.); (T.H.); (Z.Y.); (L.P.)
| | - Li Pan
- State Key Laboratory of NBC Protection for Civilian, Beijing 102205, China; (X.W.); (T.H.); (Z.Y.); (L.P.)
| | - Liangliang Wang
- State Key Laboratory of NBC Protection for Civilian, Beijing 102205, China; (X.W.); (T.H.); (Z.Y.); (L.P.)
| | - Junjie Ding
- State Key Laboratory of NBC Protection for Civilian, Beijing 102205, China; (X.W.); (T.H.); (Z.Y.); (L.P.)
| |
Collapse
|
21
|
Zhu K, Wang L, Liao T, Li W, Zhou J, You Y, Shi J. Progress in the development of TRPV1 small-molecule antagonists: Novel Strategies for pain management. Eur J Med Chem 2023; 261:115806. [PMID: 37713804 DOI: 10.1016/j.ejmech.2023.115806] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2023] [Revised: 09/06/2023] [Accepted: 09/07/2023] [Indexed: 09/17/2023]
Abstract
Transient receptor potential vanilloid 1 (TRPV1) channels are widely distributed in sensory nerve endings, the central nervous system, and other tissues, functioning as ion channel proteins responsive to thermal pain and chemical stimuli. In recent years, the TRPV1 receptor has garnered significant interest as a potential therapeutic approach for various pain-related disorders, particularly TRPV1 antagonists. The present review offers a comprehensive, systematic exploration of both first- and second-generation TRPV1 antagonists in the context of pain management. Antagonists are categorized and explicated according to their structural characteristics. Detailed examination of binding modes, structural features, and pharmacological activities, alongside a critical appraisal of the advantages and limitations inherent to typical compounds within each structural category, are undertaken. Detailed discussions of the binding modes, structural features, pharmacological activities, advantages, and limitations of typical compounds within each structural category offer valuable insights and guidance for the future research and development of safer, more effective, and more targeted TRPV1 antagonists.
Collapse
Affiliation(s)
- Kun Zhu
- Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, 610072, China
| | - Lin Wang
- Department of Pharmacy, Personalized Drug Therapy Key Laboratory of Sichuan Province, Sichuan Academy of Medical Science & Sichuan Provincial People's Hospital, Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, 610072, China; State Key Laboratory of Biotherapy, Collaborative Innovation Center of Biotherapy and Cancer Center, West China Hospital of Sichuan University, Chengdu, 610041, China
| | - TingTing Liao
- Department of Endocrinology, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, 610072, China
| | - Wen Li
- Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, 610072, China
| | - Jing Zhou
- Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, 610072, China
| | - Yaodong You
- Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, 610072, China; TCM Regulating Metabolic Diseases Key Laboratory of Sichuan Province, Chengdu, 610072, China.
| | - Jianyou Shi
- Department of Pharmacy, Personalized Drug Therapy Key Laboratory of Sichuan Province, Sichuan Academy of Medical Science & Sichuan Provincial People's Hospital, Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, 610072, China; State Key Laboratory of Southwestern Chinese Medicine Resources, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, China.
| |
Collapse
|
22
|
Dewaker V, Sharma AR, Debnath U, Park ST, Kim HS. Insights from molecular dynamics simulations of TRPV1 channel modulators in pain. Drug Discov Today 2023; 28:103798. [PMID: 37838068 DOI: 10.1016/j.drudis.2023.103798] [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: 07/28/2023] [Revised: 09/28/2023] [Accepted: 10/09/2023] [Indexed: 10/16/2023]
Abstract
TRPV1 is a nonselective cation channel vital for detecting noxious stimuli (heat, acid, capsaicin). Its role in pain makes it a potential drug target for chronic pain management, migraines, and related disorders. This review updates molecular dynamics (MD) simulation studies on the TRPV1 channel, focusing on its gating mechanism, ligand-binding sites, and implications for drug design. The article also explores challenges in developing modulators, SAR optimization, and clinical trial studies. Efforts have been undertaken to concisely present MD simulation findings, with a focus on their relevance to drug discovery.
Collapse
Affiliation(s)
- Varun Dewaker
- Institute of New Frontier Research Team, Hallym University, Chuncheon-si 24252, Gangwon-do, Republic of Korea
| | - Ashish R Sharma
- Institute for Skeletal Aging & Orthopedic Surgery, Hallym University-Chuncheon Sacred Heart Hospital, Chuncheon-si 24252, Gangwon-do, Republic of Korea
| | - Utsab Debnath
- School of Health Sciences & Technology, UPES, Dehradun, Uttarakhand 248007, India
| | - Sung Taek Park
- Institute of New Frontier Research Team, Hallym University, Chuncheon-si 24252, Gangwon-do, Republic of Korea; Department of Obstetrics and Gynecology, Kangnam Sacred-Heart Hospital, Hallym University Medical Center, Hallym University College of Medicine, Seoul 07441, Republic of Korea; EIONCELL Inc., Chuncheon 24252, Republic of Korea
| | - Hyeong Su Kim
- Institute of New Frontier Research Team, Hallym University, Chuncheon-si 24252, Gangwon-do, Republic of Korea; Division of Hemato-Oncology, Department of Internal Medicine, Kangnam Sacred-Heart Hospital, Hallym University Medical Center, Hallym University College of Medicine, Seoul 07441, Republic of Korea; EIONCELL Inc., Chuncheon 24252, Republic of Korea.
| |
Collapse
|
23
|
Luján-Méndez F, Roldán-Padrón O, Castro-Ruíz JE, López-Martínez J, García-Gasca T. Capsaicinoids and Their Effects on Cancer: The "Double-Edged Sword" Postulate from the Molecular Scale. Cells 2023; 12:2573. [PMID: 37947651 PMCID: PMC10650825 DOI: 10.3390/cells12212573] [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: 09/18/2023] [Revised: 10/30/2023] [Accepted: 11/01/2023] [Indexed: 11/12/2023] Open
Abstract
Capsaicinoids are a unique chemical species resulting from a particular biosynthesis pathway of hot chilies (Capsicum spp.) that gives rise to 22 analogous compounds, all of which are TRPV1 agonists and, therefore, responsible for the pungency of Capsicum fruits. In addition to their human consumption, numerous ethnopharmacological uses of chili have emerged throughout history. Today, more than 25 years of basic research accredit a multifaceted bioactivity mainly to capsaicin, highlighting its antitumor properties mediated by cytotoxicity and immunological adjuvancy against at least 74 varieties of cancer, while non-cancer cells tend to have greater tolerance. However, despite the progress regarding the understanding of its mechanisms of action, the benefit and safety of capsaicinoids' pharmacological use remain subjects of discussion, since CAP also promotes epithelial-mesenchymal transition, in an ambivalence that has been referred to as "the double-edge sword". Here, we update the comparative discussion of relevant reports about capsaicinoids' bioactivity in a plethora of experimental models of cancer in terms of selectivity, efficacy, and safety. Through an integration of the underlying mechanisms, as well as inherent aspects of cancer biology, we propose mechanistic models regarding the dichotomy of their effects. Finally, we discuss a selection of in vivo evidence concerning capsaicinoids' immunomodulatory properties against cancer.
Collapse
Affiliation(s)
- Francisco Luján-Méndez
- Laboratorio de Biología Celular y Molecular, Facultad de Ciencias Naturales, Universidad Autónoma de Querétaro, Av. De las Ciencias s/n, Juriquilla, Querétaro 76230, Querétaro, Mexico; (F.L.-M.); (O.R.-P.); (J.L.-M.)
| | - Octavio Roldán-Padrón
- Laboratorio de Biología Celular y Molecular, Facultad de Ciencias Naturales, Universidad Autónoma de Querétaro, Av. De las Ciencias s/n, Juriquilla, Querétaro 76230, Querétaro, Mexico; (F.L.-M.); (O.R.-P.); (J.L.-M.)
| | - J. Eduardo Castro-Ruíz
- Escuela de Odontología, Facultad de Medicina, Universidad Autónoma de Querétaro, Querétaro 76176, Querétaro, Mexico;
| | - Josué López-Martínez
- Laboratorio de Biología Celular y Molecular, Facultad de Ciencias Naturales, Universidad Autónoma de Querétaro, Av. De las Ciencias s/n, Juriquilla, Querétaro 76230, Querétaro, Mexico; (F.L.-M.); (O.R.-P.); (J.L.-M.)
| | - Teresa García-Gasca
- Laboratorio de Biología Celular y Molecular, Facultad de Ciencias Naturales, Universidad Autónoma de Querétaro, Av. De las Ciencias s/n, Juriquilla, Querétaro 76230, Querétaro, Mexico; (F.L.-M.); (O.R.-P.); (J.L.-M.)
| |
Collapse
|
24
|
Li S, Zheng J. The capsaicin binding affinity of wildtype and mutant TRPV1 ion channels. J Biol Chem 2023; 299:105268. [PMID: 37734552 PMCID: PMC10616419 DOI: 10.1016/j.jbc.2023.105268] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2023] [Revised: 09/03/2023] [Accepted: 09/12/2023] [Indexed: 09/23/2023] Open
Abstract
Vanilloids such as capsaicin and resiniferatoxin are highly selective and potent activators for transient receptor potential vanilloid subfamily, member 1, a nociceptor for heat and pain perception. However, the intrinsic vanilloid binding affinity, key for understanding transient receptor potential vanilloid subfamily, member 1 function, remains unknown despite intensive investigations by electrophysiological, structural, and computational methods. In this study, we determined capsaicin binding affinity under physiological conditions by isolating individual binding steps to each subunit with concatemers. We estimated the capsaicin association constant of a wildtype subunit to be in the order of 106 M-1 and that of the Y511A mutant subunit to be a hundred times lower, in the order of 104 M-1. The Y511A mutation, located at the entrance of the vanilloid binding pocket, reduces binding affinity without a noticeable effect on activation gating. We further affirmed that there is little cooperativity between vanilloid binding steps. Models based on independent binding and equally cooperative subunit gating can accurately describe capsaicin activation.
Collapse
Affiliation(s)
- Shisheng Li
- Department of Physiology and Membrane Biology, University of California at Davis, School of Medicine, Davis California, USA
| | - Jie Zheng
- Department of Physiology and Membrane Biology, University of California at Davis, School of Medicine, Davis California, USA.
| |
Collapse
|
25
|
Li S, Zheng J. How Much Does TRPV1 Deviate from An Ideal MWC-Type Protein. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.10.26.564268. [PMID: 37961345 PMCID: PMC10634860 DOI: 10.1101/2023.10.26.564268] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/15/2023]
Abstract
Many ion channels are known to behave as an allosteric protein, coupling environmental stimuli captured by specialized sensing domains to the opening of a central pore. The classic Monod-Wyman-Changeux (MWC) model, originally proposed to describe binding of gas molecules to hemoglobin, has been widely used for analyzing ion channel gating. Here we address the issue of how accurate the MWC model predicts activation of the capsaicin receptor TRPV1 by vanilloids. Taking advantage of a concatemeric design that makes it possible to lock TRPV1 in states with zero-to-four bound vanilloid molecules, we showed quantitatively that the overall gating behavior is satisfactorily predicted by the MWC model. There is however a small yet detectable subunit position effect: ligand binding to two kitty-corner subunits is 0.4-to-0.6 kcal/mol more effective in inducing opening than binding to two neighbor subunits. This difference, less than 10% of the overall energetic contribution from ligand binding, is not expected in hemoglobin, in which each subunit is related equivalently to all the other subunits. Significance The MWC model, proposed more than 50 years ago, is elegantly simple yet powerful in predicting the behavior of allosteric proteins like hemoglobin. Its prediction power for ion channel gating has been beautifully demonstrated in the studies of BK channels. Our present work aims to determine how accurate the MWC model predicts TRPV1 activation induced by vanilloids. Our findings support the notion that the evolutionary drive upon allosteric proteins applies generally to multi-subunit proteins.
Collapse
|
26
|
Silva JL, Santos EA, Alvarez-Leite JI. Are We Ready to Recommend Capsaicin for Disorders Other Than Neuropathic Pain? Nutrients 2023; 15:4469. [PMID: 37892544 PMCID: PMC10609899 DOI: 10.3390/nu15204469] [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: 09/25/2023] [Revised: 10/15/2023] [Accepted: 10/19/2023] [Indexed: 10/29/2023] Open
Abstract
Capsaicin, a lipophilic, volatile compound, is responsible for the pungent properties of chili peppers. In recent years, a significant increase in investigations into its properties has allowed the production of new formulations and the development of tools with biotechnological, diagnostic, and potential therapeutic applications. Most of these studies show beneficial effects, improving antioxidant and anti-inflammatory status, inducing thermogenesis, and reducing white adipose tissue. Other mechanisms, including reducing food intake and improving intestinal dysbiosis, are also described. In this way, the possible clinical application of such compound is expanding every year. This opinion article aims to provide a synthesis of recent findings regarding the mechanisms by which capsaicin participates in the control of non-communicable diseases such as obesity, diabetes, and dyslipidemia.
Collapse
Affiliation(s)
| | | | - Jacqueline I. Alvarez-Leite
- Departamento de Bioquímica e Imunologia, Universidade Federal de Minas Gerais, Belo Horizonte 30161-970, MG, Brazil; (J.L.S.); (E.A.S.)
| |
Collapse
|
27
|
Szallasi A. Resiniferatoxin: Nature's Precision Medicine to Silence TRPV1-Positive Afferents. Int J Mol Sci 2023; 24:15042. [PMID: 37894723 PMCID: PMC10606200 DOI: 10.3390/ijms242015042] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2023] [Revised: 09/27/2023] [Accepted: 10/07/2023] [Indexed: 10/29/2023] Open
Abstract
Resiniferatoxin (RTX) is an ultrapotent capsaicin analog with a unique spectrum of pharmacological actions. The therapeutic window of RTX is broad, allowing for the full desensitization of pain perception and neurogenic inflammation without causing unacceptable side effects. Intravesical RTX was shown to restore continence in a subset of patients with idiopathic and neurogenic detrusor overactivity. RTX can also ablate sensory neurons as a "molecular scalpel" to achieve permanent analgesia. This targeted (intrathecal or epidural) RTX therapy holds great promise in cancer pain management. Intra-articular RTX is undergoing clinical trials to treat moderate-to-severe knee pain in patients with osteoarthritis. Similar targeted approaches may be useful in the management of post-operative pain or pain associated with severe burn injuries. The current state of this field is reviewed, from preclinical studies through veterinary medicine to clinical trials.
Collapse
Affiliation(s)
- Arpad Szallasi
- Department of Pathology and Experimental Cancer Research, Semmelweis University, 1083 Budapest, Hungary
| |
Collapse
|
28
|
Zhao P, Tang C, Yang Y, Xiao Z, Perez-Miller S, Zhang H, Luo G, Liu H, Li Y, Liao Q, Yang F, Dong H, Khanna R, Liu Z. A new polymodal gating model of the proton-activated chloride channel. PLoS Biol 2023; 21:e3002309. [PMID: 37713449 PMCID: PMC10529583 DOI: 10.1371/journal.pbio.3002309] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2023] [Revised: 09/27/2023] [Accepted: 08/23/2023] [Indexed: 09/17/2023] Open
Abstract
The proton-activated chloride (PAC) channel plays critical roles in ischemic neuron death, but its activation mechanisms remain elusive. Here, we investigated the gating of PAC channels using its novel bifunctional modulator C77304. C77304 acted as a weak activator of the PAC channel, causing moderate activation by acting on its proton gating. However, at higher concentrations, C77304 acted as a weak inhibitor, suppressing channel activity. This dual function was achieved by interacting with 2 modulatory sites of the channel, each with different affinities and dependencies on the channel's state. Moreover, we discovered a protonation-independent voltage activation of the PAC channel that appears to operate through an ion-flux gating mechanism. Through scanning-mutagenesis and molecular dynamics simulation, we confirmed that E181, E257, and E261 in the human PAC channel serve as primary proton sensors, as their alanine mutations eliminated the channel's proton gating while sparing the voltage-dependent gating. This proton-sensing mechanism was conserved among orthologous PAC channels from different species. Collectively, our data unveils the polymodal gating and proton-sensing mechanisms in the PAC channel that may inspire potential drug development.
Collapse
Affiliation(s)
- Piao Zhao
- The National and Local Joint Engineering Laboratory of Animal Peptide Drug Development, College of Life Sciences, Hunan Normal University, Changsha, China
- Peptide and small molecule drug R&D platform, Furong Laboratory, Hunan Normal University, Changsha, China
| | - Cheng Tang
- The National and Local Joint Engineering Laboratory of Animal Peptide Drug Development, College of Life Sciences, Hunan Normal University, Changsha, China
- Peptide and small molecule drug R&D platform, Furong Laboratory, Hunan Normal University, Changsha, China
| | - Yuqin Yang
- Kuang Yaming Honors School, State Key Laboratory of Analytical Chemistry for Life Science, Engineering Research Center of Protein and Peptide Medicine of Ministry of Education, & Institute for Brain Sciences, Nanjing University, Nanjing, Jiangsu, China
| | - Zhen Xiao
- The National and Local Joint Engineering Laboratory of Animal Peptide Drug Development, College of Life Sciences, Hunan Normal University, Changsha, China
| | - Samantha Perez-Miller
- Department of Molecular Pathobiology and NYU Pain Research Center, College of Dentistry, New York University, New York, New York, United States of America
| | - Heng Zhang
- Department of Biophysics and Kidney Disease Center, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Guoqing Luo
- The National and Local Joint Engineering Laboratory of Animal Peptide Drug Development, College of Life Sciences, Hunan Normal University, Changsha, China
| | - Hao Liu
- The National and Local Joint Engineering Laboratory of Animal Peptide Drug Development, College of Life Sciences, Hunan Normal University, Changsha, China
| | - Yaqi Li
- The National and Local Joint Engineering Laboratory of Animal Peptide Drug Development, College of Life Sciences, Hunan Normal University, Changsha, China
| | - Qingyi Liao
- The National and Local Joint Engineering Laboratory of Animal Peptide Drug Development, College of Life Sciences, Hunan Normal University, Changsha, China
| | - Fan Yang
- Department of Biophysics and Kidney Disease Center, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Hao Dong
- Kuang Yaming Honors School, State Key Laboratory of Analytical Chemistry for Life Science, Engineering Research Center of Protein and Peptide Medicine of Ministry of Education, & Institute for Brain Sciences, Nanjing University, Nanjing, Jiangsu, China
| | - Rajesh Khanna
- Department of Molecular Pathobiology and NYU Pain Research Center, College of Dentistry, New York University, New York, New York, United States of America
- Department of Neuroscience and Physiology and Neuroscience Institute, School of Medicine, New York University, New York, New York, United States of America
| | - Zhonghua Liu
- The National and Local Joint Engineering Laboratory of Animal Peptide Drug Development, College of Life Sciences, Hunan Normal University, Changsha, China
- Peptide and small molecule drug R&D platform, Furong Laboratory, Hunan Normal University, Changsha, China
| |
Collapse
|
29
|
Wei X, Yang Q, Yang Z, Huang T, Yang H, Wang L, Pan L, Ding J. Discovery of novel TRPV1 modulators through machine learning-based molecular docking and molecular similarity searching. Chem Biol Drug Des 2023; 102:409-423. [PMID: 37489095 DOI: 10.1111/cbdd.14270] [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: 04/11/2023] [Revised: 05/04/2023] [Accepted: 05/09/2023] [Indexed: 07/26/2023]
Abstract
The transient receptor potential vanilloid 1 (TRPV1) channel belongs to the transient receptor potential channel superfamily and participates in many physiological processes. TRPV1 modulators (both agonists and antagonists) can effectively inhibit pain caused by various factors and have curative effects in various diseases, such as itch, cancer, and cardiovascular diseases. Therefore, the development of TRPV1 channel modulators is of great importance. In this study, the structure-based virtual screening and ligand-based virtual screening methods were used to screen compound databases respectively. In the structure-based virtual screening route, a full-length human TRPV1 protein was first constructed, three molecular docking methods with different precisions were performed based on the hTRPV1 structure, and a machine learning-based rescoring model by the XGBoost algorithm was constructed to enrich active compounds. In the ligand-based virtual screening route, the ROCS program was used for 3D shape similarity searching and the EON program was used for electrostatic similarity searching. Final 77 compounds were selected from two routes for in vitro assays. The results showed that 8 of them were identified as active compounds, including three hits with IC50 values close to capsazepine. In addition, one hit is a partial agonist with both agonistic and antagonistic activity. The mechanisms of some active compounds were investigated by molecular dynamics simulation, which explained their agonism or antagonism.
Collapse
Affiliation(s)
- Xinmiao Wei
- State Key Laboratory of NBC Protection for Civilian, Beijing, China
| | - Qifan Yang
- State Key Laboratory of NBC Protection for Civilian, Beijing, China
| | - Zhijiang Yang
- State Key Laboratory of NBC Protection for Civilian, Beijing, China
| | - Tengxin Huang
- State Key Laboratory of NBC Protection for Civilian, Beijing, China
- School of Physics and Electronic Engineering, Sichuan University of Science & Engineering, Zigong, China
| | - Hang Yang
- State Key Laboratory of NBC Protection for Civilian, Beijing, China
- School of Physics and Electronic Engineering, Sichuan University of Science & Engineering, Zigong, China
| | - Liangliang Wang
- State Key Laboratory of NBC Protection for Civilian, Beijing, China
| | - Li Pan
- State Key Laboratory of NBC Protection for Civilian, Beijing, China
| | - Junjie Ding
- State Key Laboratory of NBC Protection for Civilian, Beijing, China
| |
Collapse
|
30
|
Li X, Hu J, Yin P, Liu L, Chen Y. Mechanotransduction in the urothelium: ATP signalling and mechanoreceptors. Heliyon 2023; 9:e19427. [PMID: 37674847 PMCID: PMC10477517 DOI: 10.1016/j.heliyon.2023.e19427] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2023] [Revised: 08/10/2023] [Accepted: 08/22/2023] [Indexed: 09/08/2023] Open
Abstract
The urothelium, which covers the inner surface of the bladder, is continuously exposed to a complex physical environment where it is stimulated by, and responds to, a wide range of mechanical cues. Mechanically activated ion channels endow the urothelium with functioning in the conversion of mechanical stimuli into biochemical events that influence the surface of the urothelium itself as well as suburothelial tissues, including afferent nerve fibres, interstitial cells of Cajal and detrusor smooth muscle cells, to ensure normal urinary function during the cycle of filling and voiding. However, under prolonged and abnormal loading conditions, the urothelial sensory system can become maladaptive, leading to the development of bladder dysfunction. In this review, we summarize developments in the understanding of urothelial mechanotransduction from two perspectives: first, with regard to the functions of urothelial mechanotransduction, particularly stretch-mediated ATP signalling and the regulation of urothelial surface area; and secondly, with regard to the mechanoreceptors present in the urothelium, primarily transient receptor potential channels and mechanosensitive Piezo channels, and the potential pathophysiological role of these channels in the bladder. A more thorough understanding of urothelial mechanotransduction function may inspire the development of new therapeutic strategies for lower urinary tract diseases.
Collapse
Affiliation(s)
| | | | - Ping Yin
- Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, 200032, China
| | - Lumin Liu
- Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, 200032, China
| | - Yuelai Chen
- Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, 200032, China
| |
Collapse
|
31
|
Mio K, Ohkubo T, Sasaki D, Arai T, Sugiura M, Fujimura S, Nozawa S, Sekiguchi H, Kuramochi M, Sasaki YC. Real-Time Observation of Capsaicin-Induced Intracellular Domain Dynamics of TRPV1 Using the Diffracted X-ray Tracking Method. MEMBRANES 2023; 13:708. [PMID: 37623769 PMCID: PMC10456751 DOI: 10.3390/membranes13080708] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/08/2023] [Revised: 07/27/2023] [Accepted: 07/28/2023] [Indexed: 08/26/2023]
Abstract
The transient receptor potential vanilloid type 1 (TRPV1) is a multimodal receptor which responds to various stimuli, including capsaicin, protons, and heat. Recent advances in cryo-electron microscopy have revealed the structures of TRPV1. However, due to the large size of TRPV1 and its structural complexity, the detailed process of channel gating has not been well documented. In this study, we applied the diffracted X-ray tracking (DXT) technique to analyze the intracellular domain dynamics of the TRPV1 protein. DXT enables the capture of intramolecular motion through the analysis of trajectories of Laue spots generated from attached gold nanocrystals. Diffraction data were recorded at two different frame rates: 100 μs/frame and 12.5 ms/frame. The data from the 100 μs/frame recording were further divided into two groups based on the moving speed, using the lifetime filtering technique, and they were analyzed separately. Capsaicin increased the slope angle of the MSD curve of the C-terminus in 100 μs/frame recording, which accompanied a shifting of the rotational bias toward the counterclockwise direction, as viewed from the cytoplasmic side. This capsaicin-induced fluctuation was not observed in the 12.5 ms/frame recording, indicating that it is a high-frequency fluctuation. An intrinsiccounterclockwise twisting motion was observed in various speed components at the N-terminus, regardless of the capsaicin administration. Additionally, the competitive inhibitor AMG9810 induced a clockwise twisting motion, which is the opposite direction to capsaicin. These findings contribute to our understanding of the activation mechanisms of the TRPV1 channel.
Collapse
Affiliation(s)
- Kazuhiro Mio
- AIST-UTokyo Advanced Operando-Measurement Technology Open Innovation Laboratory (OPERANDO-OIL), National Institute of Advanced Industrial Science and Technology (AIST), 6-2-3 Kashiwanoha, Chiba 277-0882, Japan
- Graduate School of Medical Life Science, Yokohama City University, 1-7-29 Suehiro-cho, Tsurumi-ku, Yokohama 230-0045, Japan
| | - Tatsunari Ohkubo
- AIST-UTokyo Advanced Operando-Measurement Technology Open Innovation Laboratory (OPERANDO-OIL), National Institute of Advanced Industrial Science and Technology (AIST), 6-2-3 Kashiwanoha, Chiba 277-0882, Japan
- Graduate School of Medical Life Science, Yokohama City University, 1-7-29 Suehiro-cho, Tsurumi-ku, Yokohama 230-0045, Japan
| | - Daisuke Sasaki
- Graduate School of Frontier Sciences, The University of Tokyo, 5-1-5 Kashiwanoha, Chiba 277-8561, Japan (T.A.)
| | - Tatsuya Arai
- Graduate School of Frontier Sciences, The University of Tokyo, 5-1-5 Kashiwanoha, Chiba 277-8561, Japan (T.A.)
| | - Mayui Sugiura
- AIST-UTokyo Advanced Operando-Measurement Technology Open Innovation Laboratory (OPERANDO-OIL), National Institute of Advanced Industrial Science and Technology (AIST), 6-2-3 Kashiwanoha, Chiba 277-0882, Japan
| | - Shoko Fujimura
- AIST-UTokyo Advanced Operando-Measurement Technology Open Innovation Laboratory (OPERANDO-OIL), National Institute of Advanced Industrial Science and Technology (AIST), 6-2-3 Kashiwanoha, Chiba 277-0882, Japan
| | - Shunsuke Nozawa
- Photon Factory, Institute of Materials Structure Science, High Energy Accelerator Research Organization, 1-1 Oho, Tsukuba 305-0801, Japan;
| | - Hiroshi Sekiguchi
- Center for Synchrotron Radiation Research, Japan Synchrotron Radiation Research Institute, 1-1-1 Kouto, Sayo-cho 679-5198, Japan
| | - Masahiro Kuramochi
- Graduate School of Science and Engineering, Ibaraki University, Hitachi 316-8511, Japan
| | - Yuji C. Sasaki
- AIST-UTokyo Advanced Operando-Measurement Technology Open Innovation Laboratory (OPERANDO-OIL), National Institute of Advanced Industrial Science and Technology (AIST), 6-2-3 Kashiwanoha, Chiba 277-0882, Japan
- Graduate School of Frontier Sciences, The University of Tokyo, 5-1-5 Kashiwanoha, Chiba 277-8561, Japan (T.A.)
- Center for Synchrotron Radiation Research, Japan Synchrotron Radiation Research Institute, 1-1-1 Kouto, Sayo-cho 679-5198, Japan
| |
Collapse
|
32
|
Li S, Nguyen PT, Vu S, Yarov-Yarovoy V, Zheng J. Opening of capsaicin receptor TRPV1 is stabilized equally by its four subunits. J Biol Chem 2023; 299:104828. [PMID: 37196769 PMCID: PMC10318505 DOI: 10.1016/j.jbc.2023.104828] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2023] [Revised: 05/04/2023] [Accepted: 05/10/2023] [Indexed: 05/19/2023] Open
Abstract
Capsaicin receptor TRPV1 is a nociceptor for vanilloid molecules, such as capsaicin and resiniferatoxin (RTX). Even though cryo-EM structures of TRPV1 in complex with these molecules are available, how their binding energetically favors the open conformation is not known. Here, we report an approach to control the number of bound RTX molecules (0-4) in functional rat TRPV1. The approach allowed direct measurements of each of the intermediate open states under equilibrium conditions at both macroscopic and single-molecule levels. We found that RTX binding to each of the four subunits contributes virtually the same activation energy, which we estimated to be 1.70 to 1.86 kcal/mol and found to arise predominately from destabilizing the closed conformation. We further showed that sequential bindings of RTX increase open probability without altering single-channel conductance, confirming that there is likely a single open-pore conformation for TRPV1 activated by RTX.
Collapse
Affiliation(s)
- Shisheng Li
- Department of Physiology and Membrane Biology, School of Medicine, University of California at Davis, Davis, California, USA
| | - Phuong T Nguyen
- Department of Physiology and Membrane Biology, School of Medicine, University of California at Davis, Davis, California, USA
| | - Simon Vu
- Department of Physiology and Membrane Biology, School of Medicine, University of California at Davis, Davis, California, USA
| | - Vladimir Yarov-Yarovoy
- Department of Physiology and Membrane Biology, School of Medicine, University of California at Davis, Davis, California, USA
| | - Jie Zheng
- Department of Physiology and Membrane Biology, School of Medicine, University of California at Davis, Davis, California, USA.
| |
Collapse
|
33
|
Wang B, Zhang XY, Yuan S, Fu HP, Wang CZ, Wang DH. Genetic Diversity of a Heat Activated Channel-TRPV1 in Two Desert Gerbil Species with Different Heat Sensitivity. Int J Mol Sci 2023; 24:ijms24119123. [PMID: 37298074 DOI: 10.3390/ijms24119123] [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: 03/24/2023] [Revised: 05/16/2023] [Accepted: 05/16/2023] [Indexed: 06/12/2023] Open
Abstract
Heat sensation and tolerance are crucial for determining species' survival and distribution range of small mammals. As a member of the transmembrane proteins, transient receptor potential vanniloid 1 (TRPV1) is involved in the sensation and thermoregulation of heat stimuli; however, the associations between animal's heat sensitivity and TRPV1 in wild rodents are less studied. Here, we found that Mongolian gerbils (Meriones unguiculatus), a rodent species living in Mongolia grassland, showed an attenuated sensitivity to heat compared with sympatrically distributed mid-day gerbils (M. meridianus) based on a temperature preference test. To explain this phenotypical difference, we measured the TRPV1 mRNA expression of two gerbil species in the hypothalamus, brown adipose tissue, and liver, and no statistical difference was detected between two species. However, according to the bioinformatics analysis of TRPV1 gene, we identified two single amino acid mutations on two TRPV1 orthologs in these two species. Further Swiss-model analyses of two TRPV1 protein sequences indicated the disparate conformations at amino acid mutation sites. Additionally, we confirmed the haplotype diversity of TRPV1 in both species by expressing TRPV1 genes ectopicly in Escherichia coli system. Taken together, our findings supplemented genetic cues to the association between the discrepancy of heat sensitivity and the functional differentiation of TRPV1 using two wild congener gerbils, promoting the comprehension of the evolutionary mechanisms of the TRPV1 gene for heat sensitivity in small mammals.
Collapse
Affiliation(s)
- Bing Wang
- State key Laboratory of Integrated Management of Pests Insects and Rodents, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, China
- CAS Center for Excellence in Biotic Interactions, University of Chinese Academy of Sciences, Beijing 100049, China
| | - Xue-Ying Zhang
- State key Laboratory of Integrated Management of Pests Insects and Rodents, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, China
- CAS Center for Excellence in Biotic Interactions, University of Chinese Academy of Sciences, Beijing 100049, China
| | - Shuai Yuan
- College of Grassland, Resources and Environment, Inner Mongolia Agricultural University, Hohhot 010018, China
| | - He-Ping Fu
- College of Grassland, Resources and Environment, Inner Mongolia Agricultural University, Hohhot 010018, China
| | - Chen-Zhu Wang
- State key Laboratory of Integrated Management of Pests Insects and Rodents, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, China
- CAS Center for Excellence in Biotic Interactions, University of Chinese Academy of Sciences, Beijing 100049, China
| | - De-Hua Wang
- State key Laboratory of Integrated Management of Pests Insects and Rodents, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, China
- CAS Center for Excellence in Biotic Interactions, University of Chinese Academy of Sciences, Beijing 100049, China
- School of Life Sciences, Shandong University, Qingdao 266237, China
| |
Collapse
|
34
|
Seo SE, Lim SG, Kim KH, Kim J, Shin CJ, Kim S, Kim L, Lee SH, Jang SY, Oh HW, Lee HA, Kim WK, Park YM, Lee KG, Lee SH, Ha S, Kwon OS. Reusable Electronic Tongue Based on Transient Receptor Potential Vanilloid 1 Nanodisc-Conjugated Graphene Field-Effect Transistor for a Spiciness-Related Pain Evaluation. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2023; 35:e2206198. [PMID: 36856042 DOI: 10.1002/adma.202206198] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/08/2022] [Revised: 02/15/2023] [Indexed: 05/12/2023]
Abstract
The sense of spiciness is related to the stimulation of vanilloid compounds contained in the foods. Although, the spiciness is commonly considered as the part of taste, it is more classified to the sense of pain stimulated on a tongue, namely, pungency, which is described as a tingling or burning on the tongue. Herein, first, a reusable electronic tongue based on a transient receptor potential vanilloid 1 (TRPV1) nanodisc conjugated graphene field-effect transistor is fabricated and spiciness-related pain evaluation with reusable electrode is demonstrated. The pungent compound reactive receptor TRPV1 is synthesized in the form of nanodiscs to maintain stability and reusability. The newly developed platform shows highly selective and sensitive performance toward each spiciness related vanilloid compound repeatably: 1 aM capsaicin, 10 aM dihydrocapsaicin, 1 fM piperine, 10 nM allicin, and 1 pM AITC. The binding mechanism is also examined by simulation. Furthermore, the elimination of the burning sensation on the tongue after eating spicy foods is not investigated. Based on the synthesis of micelles composed of casein protein (which is contained in skim milk) that remove pungent compounds bound to TRPV1 nanodisc, the deactivation of TRPV1 is investigated, and the electrode is reusable that mimics electronic tongue.
Collapse
Affiliation(s)
- Sung Eun Seo
- Infectious Disease Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), 125 Gwahak-ro, Yuseong-gu, Daejeon, 34141, South Korea
- Department of Nano Engineering, SKKU Advanced Institute of Nanotechnology (SAINT), Sungkyunkwan University (SKKU), 2066 Seobu-ro, Jangan-gu, Suwon, 16419, South Korea
| | - Seong Gi Lim
- Infectious Disease Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), 125 Gwahak-ro, Yuseong-gu, Daejeon, 34141, South Korea
- Department of Nano Engineering, SKKU Advanced Institute of Nanotechnology (SAINT), Sungkyunkwan University (SKKU), 2066 Seobu-ro, Jangan-gu, Suwon, 16419, South Korea
| | - Kyung Ho Kim
- Infectious Disease Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), 125 Gwahak-ro, Yuseong-gu, Daejeon, 34141, South Korea
- Department of Nano Engineering, SKKU Advanced Institute of Nanotechnology (SAINT), Sungkyunkwan University (SKKU), 2066 Seobu-ro, Jangan-gu, Suwon, 16419, South Korea
| | - Jinyeong Kim
- Infectious Disease Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), 125 Gwahak-ro, Yuseong-gu, Daejeon, 34141, South Korea
| | - Chan Jae Shin
- Department of Nano Engineering, SKKU Advanced Institute of Nanotechnology (SAINT), Sungkyunkwan University (SKKU), 2066 Seobu-ro, Jangan-gu, Suwon, 16419, South Korea
| | - Soomin Kim
- Infectious Disease Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), 125 Gwahak-ro, Yuseong-gu, Daejeon, 34141, South Korea
| | - Lina Kim
- Infectious Disease Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), 125 Gwahak-ro, Yuseong-gu, Daejeon, 34141, South Korea
| | - Seung Hwan Lee
- Department of Bionano Engineering, Center for Bionano Intelligence Education and Research, Hanyang University, 55 Hanyangdaehak-ro, Sangnok-gu, Ansan, 15588, South Korea
- Department of Chemical and Biological Engineering, Hanbat National University, 125 Dongseo-daero, Yuseong-gu, Daejeon, 34158, South Korea
| | - Song Yee Jang
- Core Research Facility & Analysis Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), 125 Gwahak-ro, Yuseong-gu, Daejeon, 34141, South Korea
- School of Pharmacy, University of Maryland Eastern Shore, Princess Anne, MD, 21853, United States
| | - Hyun Woo Oh
- Core Research Facility & Analysis Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), 125 Gwahak-ro, Yuseong-gu, Daejeon, 34141, South Korea
- School of Pharmacy, University of Maryland Eastern Shore, Princess Anne, MD, 21853, United States
| | - Hyang-Ae Lee
- Department of Predictive Toxicology, Korea Institute of Toxicology, 141 Gajeong-ro, Yuseong-gu, Daejeon, 34114, South Korea
| | - Woo-Keun Kim
- Department of Predictive Toxicology, Korea Institute of Toxicology, 141 Gajeong-ro, Yuseong-gu, Daejeon, 34114, South Korea
| | - Yoo Min Park
- Center for NanoBio Development, National NanoFab Center, 291 Daehak-ro, Yuseong-gu, Daejeon, 34141, South Korea
| | - Kyoung G Lee
- Center for NanoBio Development, National NanoFab Center, 291 Daehak-ro, Yuseong-gu, Daejeon, 34141, South Korea
| | - Sang Hun Lee
- Department of Chemical and Biological Engineering, Hanbat National University, 125 Dongseo-daero, Yuseong-gu, Daejeon, 34158, South Korea
| | - Siyoung Ha
- School of Pharmacy, University of Maryland Eastern Shore, Princess Anne, MD, 21853, United States
| | - Oh Seok Kwon
- Infectious Disease Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), 125 Gwahak-ro, Yuseong-gu, Daejeon, 34141, South Korea
- Department of Nano Engineering, SKKU Advanced Institute of Nanotechnology (SAINT), Sungkyunkwan University (SKKU), 2066 Seobu-ro, Jangan-gu, Suwon, 16419, South Korea
| |
Collapse
|
35
|
Neuberger A, Oda M, Nikolaev YA, Nadezhdin KD, Gracheva EO, Bagriantsev SN, Sobolevsky AI. Human TRPV1 structure and inhibition by the analgesic SB-366791. Nat Commun 2023; 14:2451. [PMID: 37117175 PMCID: PMC10147690 DOI: 10.1038/s41467-023-38162-9] [Citation(s) in RCA: 38] [Impact Index Per Article: 19.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2022] [Accepted: 04/18/2023] [Indexed: 04/30/2023] Open
Abstract
Pain therapy has remained conceptually stagnant since the opioid crisis, which highlighted the dangers of treating pain with opioids. An alternative addiction-free strategy to conventional painkiller-based treatment is targeting receptors at the origin of the pain pathway, such as transient receptor potential (TRP) ion channels. Thus, a founding member of the vanilloid subfamily of TRP channels, TRPV1, represents one of the most sought-after pain therapy targets. The need for selective TRPV1 inhibitors extends beyond pain treatment, to other diseases associated with this channel, including psychiatric disorders. Here we report the cryo-electron microscopy structures of human TRPV1 in the apo state and in complex with the TRPV1-specific nanomolar-affinity analgesic antagonist SB-366791. SB-366791 binds to the vanilloid site and acts as an allosteric hTRPV1 inhibitor. SB-366791 binding site is supported by mutagenesis combined with electrophysiological recordings and can be further explored to design new drugs targeting TRPV1 in disease conditions.
Collapse
Affiliation(s)
- Arthur Neuberger
- Department of Biochemistry and Molecular Biophysics, Columbia University, New York, NY, USA
| | - Mai Oda
- Department of Cellular and Molecular Physiology, Yale University School of Medicine, New Haven, CT, 06510, USA
| | - Yury A Nikolaev
- Department of Cellular and Molecular Physiology, Yale University School of Medicine, New Haven, CT, 06510, USA
| | - Kirill D Nadezhdin
- Department of Biochemistry and Molecular Biophysics, Columbia University, New York, NY, USA
| | - Elena O Gracheva
- Department of Cellular and Molecular Physiology, Yale University School of Medicine, New Haven, CT, 06510, USA
- Department of Neuroscience, Yale University School of Medicine, New Haven, CT, 06510, USA
- Program in Cellular Neuroscience, Neurodegeneration and Repair, Yale University School of Medicine, New Haven, CT, 06510, USA
- Kavli Institute for Neuroscience, Yale University School of Medicine, New Haven, CT, 06510, USA
| | - Sviatoslav N Bagriantsev
- Department of Cellular and Molecular Physiology, Yale University School of Medicine, New Haven, CT, 06510, USA
| | - Alexander I Sobolevsky
- Department of Biochemistry and Molecular Biophysics, Columbia University, New York, NY, USA.
| |
Collapse
|
36
|
Bustos D, Galarza C, Ordoñez W, Brauchi S, Benso B. Cost-Effective Pipeline for a Rational Design and Selection of Capsaicin Analogues Targeting TRPV1 Channels. ACS OMEGA 2023; 8:11736-11749. [PMID: 37033853 PMCID: PMC10077575 DOI: 10.1021/acsomega.2c05672] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/01/2022] [Accepted: 11/25/2022] [Indexed: 06/19/2023]
Abstract
Transient receptor potential (TRP) channels constitute a large group of membrane receptors associated with sensory pathways in vertebrates. One of the most studied is TRPV1, a polymodal receptor tuned for detecting heat and pungent compounds. Specific inhibition of the nociceptive transduction at the peripheral nerve represents a convenient approach to pain relief. While acting as a chemoreceptor, TRPV1 shows high sensitivity and selectivity for capsaicin. In contrast to the drugs available on the market that target the inflammatory system, TRPV1 antagonists act as negative modulators of nociceptive transduction. Therefore, the development of compounds modulating TRPV1 activity has expanded dramatically over time. Experimental data suggest that most agonist and antagonist drugs interact at or near capsaicin's binding site. In particular, the properties of capsaicin's head play an essential role in modulating potency and affinity. Here, we explored a cost-efficient pipeline to predict the effects of introducing chemical modifications into capsaicin's head region. An extensive set of molecules was selected by first considering the geometrical properties of capsaicin's binding site and then molecular docking. Finally, the novel ligands were ranked by combining molecular and pharmacokinetic predictions.
Collapse
Affiliation(s)
- Daniel Bustos
- Centro
de Investigación de Estudios Avanzados del Maule (CIEAM), Vicerrectoría
de Investigación y Postgrado, Universidad
Católica del Maule, Talca 3460000, Chile
- Laboratorio
de Bioinformática y Química Computacional, Departamento
de Medicina Traslacional, Facultad de Medicina, Universidad Católica del Maule, Talca 3480094, Chile
| | - Christian Galarza
- Facultad
de Ciencias Naturales y Matemáticas, Escuela Superior Politécnica del Litoral, ESPOL, Guayaquil 090703, Ecuador
| | - Wilson Ordoñez
- Facultad
de Ciencias Naturales y Matemáticas, Escuela Superior Politécnica del Litoral, ESPOL, Guayaquil 090703, Ecuador
| | - Sebastian Brauchi
- Department
of Physiology, Faculty of Medicine, Universidad
Austral de Chile, Valdivia 5090000, Chile
- Millennium
Nucleus of Ion Channels Associated Diseases (MiNICAD), 8330024, Chile
| | - Bruna Benso
- Millennium
Nucleus of Ion Channels Associated Diseases (MiNICAD), 8330024, Chile
- School of
Dentistry, Faculty of Medicine, Pontificia
Universidad Católica de Chile, Santiago 8330024, Chile
| |
Collapse
|
37
|
Oz M, Lorke DE, Howarth FC. Transient receptor potential vanilloid 1 (TRPV1)-independent actions of capsaicin on cellular excitability and ion transport. Med Res Rev 2023. [PMID: 36916676 DOI: 10.1002/med.21945] [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: 06/14/2022] [Revised: 01/17/2023] [Accepted: 02/26/2023] [Indexed: 03/15/2023]
Abstract
Capsaicin is a naturally occurring alkaloid derived from chili pepper that is responsible for its hot pungent taste. Capsaicin is known to exert multiple pharmacological actions, including analgesia, anticancer, anti-inflammatory, antiobesity, and antioxidant effects. The transient receptor potential vanilloid subfamily member 1 (TRPV1) is the main receptor mediating the majority of the capsaicin effects. However, numerous studies suggest that the TRPV1 receptor is not the only target for capsaicin. An increasing number of studies indicates that capsaicin, at low to mid µM ranges, not only indirectly through TRPV1-mediated Ca2+ increases, but also directly modulates the functions of voltage-gated Na+ , K+ , and Ca2+ channels, as well as ligand-gated ion channels and other ion transporters and enzymes involved in cellular excitability. These TRPV1-independent effects are mediated by alterations of the biophysical properties of the lipid membrane and subsequent modulation of the functional properties of ion channels and by direct binding of capsaicin to the channels. The present study, for the first time, systematically categorizes this diverse range of non-TRPV1 targets and discusses cellular and molecular mechanisms mediating TRPV1-independent effects of capsaicin in excitable, as well as nonexcitable cells.
Collapse
Affiliation(s)
- Murat Oz
- Department of Pharmacology and Therapeutics, Faculty of Pharmacy, Kuwait University, Safat, Kuwait
| | - Dietrich E Lorke
- Department of Anatomy and Cellular Biology, College of Medicine and Health Sciences, Khalifa University, Abu Dhabi, United Arab Emirates.,Center for Biotechnology, Khalifa University of Science and Technology, Abu Dhabi, United Arab Emirates
| | - Frank C Howarth
- Department of Physiology, College of Medicine and Health Sciences, UAE University, Al Ain, United Arab Emirates
| |
Collapse
|
38
|
Zhu KF, Yuan C, Du YM, Sun KL, Zhang XK, Vogel H, Jia XD, Gao YZ, Zhang QF, Wang DP, Zhang HW. Applications and prospects of cryo-EM in drug discovery. Mil Med Res 2023; 10:10. [PMID: 36872349 PMCID: PMC9986049 DOI: 10.1186/s40779-023-00446-y] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/09/2022] [Accepted: 02/14/2023] [Indexed: 03/07/2023] Open
Abstract
Drug discovery is a crucial part of human healthcare and has dramatically benefited human lifespan and life quality in recent centuries, however, it is usually time- and effort-consuming. Structural biology has been demonstrated as a powerful tool to accelerate drug development. Among different techniques, cryo-electron microscopy (cryo-EM) is emerging as the mainstream of structure determination of biomacromolecules in the past decade and has received increasing attention from the pharmaceutical industry. Although cryo-EM still has limitations in resolution, speed and throughput, a growing number of innovative drugs are being developed with the help of cryo-EM. Here, we aim to provide an overview of how cryo-EM techniques are applied to facilitate drug discovery. The development and typical workflow of cryo-EM technique will be briefly introduced, followed by its specific applications in structure-based drug design, fragment-based drug discovery, proteolysis targeting chimeras, antibody drug development and drug repurposing. Besides cryo-EM, drug discovery innovation usually involves other state-of-the-art techniques such as artificial intelligence (AI), which is increasingly active in diverse areas. The combination of cryo-EM and AI provides an opportunity to minimize limitations of cryo-EM such as automation, throughput and interpretation of medium-resolution maps, and tends to be the new direction of future development of cryo-EM. The rapid development of cryo-EM will make it as an indispensable part of modern drug discovery.
Collapse
Affiliation(s)
- Kong-Fu Zhu
- Department of Biomedical Engineering, Southern University of Science and Technology, Shenzhen, 518055 Guangdong China
| | - Chuang Yuan
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Peking University, Beijing, 100191 China
| | - Yong-Ming Du
- Department of Structural Biology, St. Jude Children’s Research Hospital, Memphis, TN 38105 USA
| | - Kai-Lei Sun
- Center for Protein Science and Crystallography, School of Life Sciences, Faculty of Science, Chinese University of Hong Kong, Hong Kong, 999077 China
| | - Xiao-Kang Zhang
- Interdisciplinary Center for Brain Information, the Brain Cognition and Brain Disease Institute, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, 518055 Guangdong China
- Faculty of Life and Health Sciences, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, 518055 Guangdong China
- Shenzhen-Hong Kong Institute of Brain Science-Shenzhen Fundamental Research Institutions, Shenzhen, 518055 Guangdong China
| | - Horst Vogel
- Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, 518055 Guangdong China
| | - Xu-Dong Jia
- State Key Lab for Biocontrol, School of Life Sciences, Sun Yat-Sen University, Guangzhou, 510275 China
| | - Yuan-Zhu Gao
- Cryo-EM Facility Center, Southern University of Science and Technology, Shenzhen, 518055 Guangdong China
| | - Qin-Fen Zhang
- State Key Lab for Biocontrol, School of Life Sciences, Sun Yat-Sen University, Guangzhou, 510275 China
| | - Da-Ping Wang
- Department of Biomedical Engineering, Southern University of Science and Technology, Shenzhen, 518055 Guangdong China
- Department of Orthopedics, Shenzhen Intelligent Orthopaedics and Biomedical Innovation Platform, Guangdong Provincial Research Center for Artificial Intelligence and Digital Orthopedic Technology, Shenzhen Second People’s Hospital, The First Affiliated Hospital of Shenzhen University, Shenzhen, 518000 Guangdong China
| | - Hua-Wei Zhang
- Department of Biomedical Engineering, Southern University of Science and Technology, Shenzhen, 518055 Guangdong China
- Guangdong Provincial Key Laboratory of Advanced Biomaterials, Southern University of Science and Technology, Shenzhen, 518055 Guangdong China
| |
Collapse
|
39
|
Mateos DL, Yarov-Yarovoy V. Structural modeling of peptide toxin-ion channel interactions using RosettaDock. Proteins 2023. [PMID: 36729043 DOI: 10.1002/prot.26474] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2022] [Revised: 12/09/2022] [Accepted: 01/30/2023] [Indexed: 02/03/2023]
Abstract
Voltage-gated ion channels play essential physiological roles in action potential generation and propagation. Peptidic toxins from animal venoms target ion channels and provide useful scaffolds for the rational design of novel channel modulators with enhanced potency and subtype selectivity. Despite recent progress in obtaining experimental structures of peptide toxin-ion channel complexes, structural determination of peptide toxins bound to ion channels in physiologically important states remains challenging. Here we describe an application of RosettaDock approach to the structural modeling of peptide toxins interactions with ion channels. We tested this approach on 10 structures of peptide toxin-ion channel complexes and demonstrated that it can sample near-native structures in all tested cases. Our approach will be useful for improving the understanding of the molecular mechanism of natural peptide toxin modulation of ion channel gating and for the structural modeling of novel peptide-based ion channel modulators.
Collapse
Affiliation(s)
- Diego Lopez Mateos
- Department of Physiology and Membrane Biology, University of California Davis, Davis, California, USA.,Biophysics Graduate Group, University of California Davis, Davis, California, USA
| | - Vladimir Yarov-Yarovoy
- Department of Physiology and Membrane Biology, University of California Davis, Davis, California, USA.,Biophysics Graduate Group, University of California Davis, Davis, California, USA.,Department of Anesthesiology and Pain Medicine, University of California Davis, Davis, California, USA
| |
Collapse
|
40
|
Li S, Nguyen PT, Vu S, Yarov-Yarovoy V, Zheng J. TRPV1 Opening is Stabilized Equally by Its Four Subunits. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.01.26.525787. [PMID: 36747729 PMCID: PMC9900918 DOI: 10.1101/2023.01.26.525787] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/30/2023]
Abstract
Capsaicin receptor TRPV1 is a nociceptor for vanilloid molecules such as capsaicin and resiniferatoxin (RTX). Even though cryo-EM structures of TRPV1 in complex with these molecules are available, how their binding energetically favors the open conformation is not known. Here we report an approach to control the number of bound RTX molecules (0-to-4) in functional mouse TRPV1. The approach allowed direct measurements of each of the intermediate open states under equilibrium conditions at both macroscopic and single-molecule levels. We found that RTX binding to each of the four subunits contributes virtually the same activation energy, which we estimated to be 1.86 kcal/mol and found to arise predominately from destabilizing the closed conformation. We further showed that sequential bindings of RTX increase open probability without altering single-channel conductance, confirming that there is likely a single open-pore conformation for TRPV1 activated by RTX.
Collapse
|
41
|
Andrei C, Zanfirescu A, Nițulescu GM, Olaru OT, Negreș S. Natural Active Ingredients and TRPV1 Modulation: Focus on Key Chemical Moieties Involved in Ligand-Target Interaction. PLANTS (BASEL, SWITZERLAND) 2023; 12:339. [PMID: 36679051 PMCID: PMC9860573 DOI: 10.3390/plants12020339] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 11/25/2022] [Revised: 01/06/2023] [Accepted: 01/08/2023] [Indexed: 06/17/2023]
Abstract
Diseases such as cancer, neurological pathologies and chronic pain represent currently unmet needs. The existing pharmacotherapeutic options available for treating these conditions are limited by lack of efficiency and/or side effects. Transient receptor potential vanilloid 1 ion channel emerged as an attractive therapeutic target for developing new analgesic, anti-cancer and antiepileptic agents. Furthermore, various natural ingredients were shown to have affinity for this receptor. The aim of this narrative review was to summarize the diverse natural scaffolds of TRPV1 modulators based on their agonistic/antagonistic properties and to analyze the structure-activity relationships between the ligands and molecular targets based on the results of the existing molecular docking, mutagenesis and in vitro studies. We present here an exhaustive collection of TRPV1 modulators grouped by relevant chemical features: vanilloids, guaiacols, phenols, alkylbenzenes, monoterpenes, sesquiterpenoids, alkaloids, etc. The information herein is useful for understanding the key structural elements mediating the interaction with TRPV1 and how their structural variation impacts the interaction between the ligand and receptor. We hope this data will contribute to the design of novel effective and safe TRPV1 modulators, to help overcome the lack of effective therapeutic agents against pathologies with high morbidity and mortality.
Collapse
|
42
|
Structure-guided peptide engineering of a positive allosteric modulator targeting the outer pore of TRPV1 for long-lasting analgesia. Nat Commun 2023; 14:4. [PMID: 36596769 PMCID: PMC9810691 DOI: 10.1038/s41467-022-34817-1] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2022] [Accepted: 11/08/2022] [Indexed: 01/05/2023] Open
Abstract
Transient receptor potential vanilloid 1 (TRPV1) ion channel is a classic analgesic target, but antagonists of TRPV1 failed in clinical trials due to their side effects like hyperthermia. Here we rationally engineer a peptide s-RhTx as a positive allosteric modulator (PAM) of TRPV1. Patch-clamp recordings demonstrate s-RhTx selectively potentiated TRPV1 activation. s-RhTx also slows down capsaicin-induced desensitization of TRPV1 in the presence of calcium to cause more calcium influx in TRPV1-expressing cells. In addition, our thermodynamic mutant cycle analysis shows that E652 in TRPV1 outer pore specifically interacts with R12 and K22 in s-RhTx. Furthermore, we demonstrate in vivo that s-RhTx exhibits long-lasting analgesic effects in noxious heat hyperalgesia and CFA-induced chronic inflammatory pain by promoting the reversible degeneration of intra-epidermal nerve fiber (IENF) expressing TRPV1 channels in mice, while their body temperature remains unaffected. Our results suggest s-RhTx is an analgesic agent as a PAM of TRPV1.
Collapse
|
43
|
Tatsumi M, Kishi T, Ishida S, Kawana H, Uwamizu A, Ono Y, Kawakami K, Aoki J, Inoue A. Ectodomain shedding of EGFR ligands serves as an activation readout for TRP channels. PLoS One 2023; 18:e0280448. [PMID: 36668668 PMCID: PMC9858409 DOI: 10.1371/journal.pone.0280448] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2022] [Accepted: 12/29/2022] [Indexed: 01/21/2023] Open
Abstract
Transient receptor potential (TRP) channels are activated by various extracellular and intracellular stimuli and are involved in many physiological events. Because compounds that act on TRP channels are potential candidates for therapeutic agents, a simple method for evaluating TRP channel activation is needed. In this study, we demonstrated that a transforming growth factor alpha (TGFα) shedding assay, previously developed for detecting G-protein-coupled receptor (GPCR) activation, can also detect TRP channel activation. This assay is a low-cost, easily accessible method that requires only an absorbance microplate reader. Mechanistically, TRP-channel-triggered TGFα shedding is achieved by both of a disintegrin and metalloproteinase domain-containing protein 10 (ADAM10) and 17 (ADAM17), whereas the GPCR-induced TGFα shedding response depends solely on ADAM17. This difference may be the result of qualitative or quantitative differences in intracellular Ca2+ kinetics between TRP channels and GPCRs. Use of epidermal growth factor (EGF) and betacellulin (BTC), substrates of ADAM10, improved the specificity of the shedding assay by reducing background responses mediated by endogenously expressed GPCRs. This assay for TRP channel measurement will not only facilitate the high-throughput screening of TRP channel ligands but also contribute to understanding the roles played by TRP channels as regulators of membrane protein ectodomain shedding.
Collapse
Affiliation(s)
- Manae Tatsumi
- Molecular and Cellular Biochemistry, Graduate School of Pharmaceutical Sciences, Tohoku University, Sendai, Japan
| | - Takayuki Kishi
- Molecular and Cellular Biochemistry, Graduate School of Pharmaceutical Sciences, Tohoku University, Sendai, Japan
| | - Satoru Ishida
- Molecular and Cellular Biochemistry, Graduate School of Pharmaceutical Sciences, Tohoku University, Sendai, Japan
| | - Hiroki Kawana
- Department of Health Chemistry, Graduate School of Pharmaceutical Sciences, The University of Tokyo, Bunkyo-ku, Tokyo, Japan
| | - Akiharu Uwamizu
- Department of Health Chemistry, Graduate School of Pharmaceutical Sciences, The University of Tokyo, Bunkyo-ku, Tokyo, Japan
| | - Yuki Ono
- Molecular and Cellular Biochemistry, Graduate School of Pharmaceutical Sciences, Tohoku University, Sendai, Japan
| | - Kouki Kawakami
- Molecular and Cellular Biochemistry, Graduate School of Pharmaceutical Sciences, Tohoku University, Sendai, Japan
| | - Junken Aoki
- Department of Health Chemistry, Graduate School of Pharmaceutical Sciences, The University of Tokyo, Bunkyo-ku, Tokyo, Japan
| | - Asuka Inoue
- Molecular and Cellular Biochemistry, Graduate School of Pharmaceutical Sciences, Tohoku University, Sendai, Japan
- * E-mail:
| |
Collapse
|
44
|
Structural mechanisms of TRPV2 modulation by endogenous and exogenous ligands. Nat Chem Biol 2023; 19:72-80. [PMID: 36163384 DOI: 10.1038/s41589-022-01139-8] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2022] [Accepted: 08/10/2022] [Indexed: 12/31/2022]
Abstract
The transient receptor potential vanilloid 2 (TRPV2) ion channel is a polymodal receptor widely involved in many physiological and pathological processes. Despite many TRPV2 modulators being identified, whether and how TRPV2 is regulated by endogenous lipids remains elusive. Here, we report an endogenous cholesterol molecule inside the vanilloid binding pocket (VBP) of TRPV2, with a 'head down, tail up' configuration, resolved at 3.2 Å using cryo-EM. Cholesterol binding antagonizes ligand activation of TRPV2, which is removed from VBP by methyl-β-cyclodextrin (MβCD) as resolved at 2.9 Å. We also observed that estradiol (E2) potentiated TRPV2 activation by 2-aminoethoxydiphenyl borate (2-APB), a classic tool compound for TRP channels. Our cryo-EM structures (resolved at 2.8-3.3 Å) further suggest how E2 disturbed cholesterol binding and how 2-APB bound within the VBP with E2 or without both E2 and endogenous cholesterol, respectively. Therefore, our study has established the structural basis for ligand recognition of the inhibitory endogenous cholesterol and excitatory exogenous 2-APB in TRPV2.
Collapse
|
45
|
Peng JW, Gu YY, Wei J, Sun Y, Zhu CL, Zhang L, Song Y, Chen L, Chen X, Wang Q, Zhang HL. LncRNA MEG3-TRPV1 signaling regulates chronic inflammatory pain in rats. Mol Pain 2022; 18:17448069221144246. [PMID: 36424837 PMCID: PMC9726848 DOI: 10.1177/17448069221144246] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022] Open
Abstract
Osteoarthritis (OA) is a common osteoarthropathy with chronic inflammatory pain as the core symptom in middle-aged and elderly people. LncRNA MEG3 (Maternally expressed gene 3) is involved in the development of OA via regulation of angiogenesis, which causes the activation and overexpression of transient receptor potential vanilloid type-1 (TRPV1). In this study, we investigated the mechanism of MEG3-TRPV1 signaling in chronic inflammatory pain (CIP) of rat model. Chronic inflammatory pain was modeled using subcutaneous microinjection of complete Freund's adjuvant (CFA) into the left hind paw of rats. We showed that TRPV1 mRNA and protein were significantly increased, while MEG3 mRNA was significantly decreased, in the DRG and SDH of CFA-induced rats. In addition, intrathecal injection of MEG3-overexpressing lentivirus significantly downregulated TRPV1 expression and alleviated chronic inflammatory pain in CFA-induced rats. Treatment with a TRPV1 antagonist also significantly relieved chronic inflammatory pain in CFA-induced rats. In general, our results reveal that MEG3 alleviates chronic inflammatory pain by downregulating TRPV1 expression. These findings may provide new therapeutic targets in the treatment of patients with OA.
Collapse
Affiliation(s)
- Jing-Wei Peng
- Department of Traditional Chinese Medicine, Affiliated Zhangjiagang Hospital of Soochow University, Zhangjiagang, China,Center for Translational Medicine, Affiliated Zhangjiagang Hospital of Soochow University, Zhangjiagang, China
| | - Yin-Yin Gu
- Department of Traditional Chinese Medicine, Affiliated Zhangjiagang Hospital of Soochow University, Zhangjiagang, China
| | - Jia Wei
- Department of Traditional Chinese Medicine, Affiliated Zhangjiagang Hospital of Soochow University, Zhangjiagang, China
| | - Ye Sun
- Department of Traditional Chinese Medicine, Affiliated Zhangjiagang Hospital of Soochow University, Zhangjiagang, China
| | - Chun-Long Zhu
- Department of Traditional Chinese Medicine, Affiliated Zhangjiagang Hospital of Soochow University, Zhangjiagang, China
| | - Ling Zhang
- Center for Translational Medicine, Affiliated Zhangjiagang Hospital of Soochow University, Zhangjiagang, China
| | - Yu Song
- Center for Translational Medicine, Affiliated Zhangjiagang Hospital of Soochow University, Zhangjiagang, China
| | - Long Chen
- Center for Translational Medicine, Affiliated Zhangjiagang Hospital of Soochow University, Zhangjiagang, China
| | - Xia Chen
- Department of Anesthesiology, Children’s Hospital of Soochow University, Suzhou, China
| | - Qian Wang
- Department of Anesthesiology, Children’s Hospital of Soochow University, Suzhou, China,Qian Wang, Department of Anesthesiology, Children’s Hospital of Soochow University, Suzhou 215123, China.
| | - Hai-Long Zhang
- Center for Translational Medicine, Affiliated Zhangjiagang Hospital of Soochow University, Zhangjiagang, China,Jiangsu Key Laboratory of Neuropsychiatric Diseases and Institute of Neuroscience, Soochow University, Suzhou, China,Hai-Long Zhang, Center for Translational Pain Medicine, Institute of Neuroscience, Soochow University, Suzhou, 215123, China.
| |
Collapse
|
46
|
Zhang L, Simonsen C, Zimova L, Wang K, Moparthi L, Gaudet R, Ekoff M, Nilsson G, Hellmich UA, Vlachova V, Gourdon P, Zygmunt PM. Cannabinoid non-cannabidiol site modulation of TRPV2 structure and function. Nat Commun 2022; 13:7483. [PMID: 36470868 PMCID: PMC9722916 DOI: 10.1038/s41467-022-35163-y] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2022] [Accepted: 11/21/2022] [Indexed: 12/12/2022] Open
Abstract
TRPV2 is a ligand-operated temperature sensor with poorly defined pharmacology. Here, we combine calcium imaging and patch-clamp electrophysiology with cryo-electron microscopy (cryo-EM) to explore how TRPV2 activity is modulated by the phytocannabinoid Δ9-tetrahydrocannabiorcol (C16) and by probenecid. C16 and probenecid act in concert to stimulate TRPV2 responses including histamine release from rat and human mast cells. Each ligand causes distinct conformational changes in TRPV2 as revealed by cryo-EM. Although the binding for probenecid remains elusive, C16 associates within the vanilloid pocket. As such, the C16 binding location is distinct from that of cannabidiol, partially overlapping with the binding site of the TRPV2 inhibitor piperlongumine. Taken together, we discover a new cannabinoid binding site in TRPV2 that is under the influence of allosteric control by probenecid. This molecular insight into ligand modulation enhances our understanding of TRPV2 in normal and pathophysiology.
Collapse
Affiliation(s)
- Liying Zhang
- grid.4514.40000 0001 0930 2361Department of Clinical Sciences Malmö, Lund University, Malmö, Sweden ,grid.4514.40000 0001 0930 2361Department of Experimental Medical Science, Lund University, Lund, Sweden ,grid.5254.60000 0001 0674 042XDepartment of Biomedical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Charlotte Simonsen
- grid.4514.40000 0001 0930 2361Department of Clinical Sciences Malmö, Lund University, Malmö, Sweden
| | - Lucie Zimova
- grid.418095.10000 0001 1015 3316Department of Cellular Neurophysiology, Institute of Physiology, Czech Academy of Sciences, Prague, Czech Republic
| | - Kaituo Wang
- grid.5254.60000 0001 0674 042XDepartment of Biomedical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Lavanya Moparthi
- grid.5640.70000 0001 2162 9922Wallenberg Centre for Molecular Medicine, Linköping University, Linköping, Sweden ,grid.5640.70000 0001 2162 9922Department of Biomedical and Clinical Sciences, Linköping University, Linköping, Sweden
| | - Rachelle Gaudet
- grid.38142.3c000000041936754XDepartment of Molecular and Cellular Biology, Harvard University, Cambridge, MA USA
| | - Maria Ekoff
- grid.24381.3c0000 0000 9241 5705Division Immunology and Allergy Unit, Department of Medicine Solna, Karolinska Institutet, Karolinska University Hospital, Solna, Sweden
| | - Gunnar Nilsson
- grid.24381.3c0000 0000 9241 5705Division Immunology and Allergy Unit, Department of Medicine Solna, Karolinska Institutet, Karolinska University Hospital, Solna, Sweden
| | - Ute A. Hellmich
- grid.9613.d0000 0001 1939 2794Faculty of Chemistry and Earth Sciences, Institute of Organic Chemistry and Macromolecular Chemistry and Cluster of Excellence “Balance of the Microverse”, Friedrich Schiller University Jena, Jena, Germany ,grid.7839.50000 0004 1936 9721Center for Biomolecular Magnetic Resonance, Goethe-University, Frankfurt/Main, Germany
| | - Viktorie Vlachova
- grid.418095.10000 0001 1015 3316Department of Cellular Neurophysiology, Institute of Physiology, Czech Academy of Sciences, Prague, Czech Republic
| | - Pontus Gourdon
- grid.4514.40000 0001 0930 2361Department of Experimental Medical Science, Lund University, Lund, Sweden ,grid.5254.60000 0001 0674 042XDepartment of Biomedical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Peter M. Zygmunt
- grid.4514.40000 0001 0930 2361Department of Clinical Sciences Malmö, Lund University, Malmö, Sweden
| |
Collapse
|
47
|
Chen X, Xu L, Zhang H, Wen H, Yang F. Differential Activation of TRPM8 by the Stereoisomers of Menthol. Front Pharmacol 2022; 13:898670. [PMID: 35800442 PMCID: PMC9253294 DOI: 10.3389/fphar.2022.898670] [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: 03/17/2022] [Accepted: 05/09/2022] [Indexed: 11/29/2022] Open
Abstract
The stereoisomers of menthol elicit cooling sensation to various levels. Though the high-resolution three-dimensional structures of the menthol receptor, the transient receptor potential melastatin 8 (TRPM8) ion channels, have been revolved in different states, the menthol-bound state structure is not determined and how the stereoisomers of menthol interact with TRPM8 remains largely elusive. Taking advantage of the identical atom composition but distinct spatial orientation of chemical groups in menthol stereoisomers, we performed thermodynamic mutant cycle analysis (TMCA) with patch-clamp recordings to probe the interaction between these ligands and TRPM8. By comparing (−)-menthol with (+)-neoisomenthol or (+)-neomenthol, we observed that the isopropyl or hydroxyl group in menthol interacts with the S4 or S3 helix in TRPM8, respectively. These interactions were also corroborated in our molecular docking of the stereoisomers, though the predicted structural details in the interactions of these ligands with TRPM8 residues are different. Therefore, we suggest similar molecular mechanisms of TRPM8 activation by the stereoisomers of menthol, while the binding configuration of an individual stereoisomer is varied.
Collapse
Affiliation(s)
- Xiaoying Chen
- Department of Biophysics, Kidney Disease Center of the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Lizhen Xu
- Department of Biophysics, Kidney Disease Center of the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Heng Zhang
- Department of Biophysics, Kidney Disease Center of the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Han Wen
- DP Technology, Beijing, China
| | - Fan Yang
- Department of Biophysics, Kidney Disease Center of the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
- Alibaba-Zhejiang University Joint Research Center of Future Digital Healthcare, Hangzhou, China
- *Correspondence: Fan Yang,
| |
Collapse
|
48
|
Sun MY, Zhang X, Yu PC, Liu D, Yang Y, Cui WW, Yang XN, Lei YT, Li XH, Wang WH, Cao P, Wang HS, Zhu MX, Li CZ, Wang R, Fan YZ, Yu Y. Vanilloid agonist-mediated activation of TRPV1 channels requires coordinated movement of the S1-S4 bundle rather than a quiescent state. Sci Bull (Beijing) 2022; 67:1062-1076. [PMID: 36546250 DOI: 10.1016/j.scib.2022.02.016] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2021] [Revised: 01/12/2022] [Accepted: 02/16/2022] [Indexed: 01/07/2023]
Abstract
Transient receptor potential vanilloid1 (TRPV1) channel plays an important role in a wide range of physiological and pathological processes, and a comprehensive understanding of TRPV1 gating will create opportunities for therapeutic intervention. Recent incredible advances in cryo-electron microscopy (cryo-EM) have yielded high-resolution structures of all TRPV subtypes (TRPV1-6) and all of them share highly conserved six transmembrane (TM) domains (S1-S6). As revealed by the open structures of TRPV1 in the presence of a bound vanilloid agonist (capsaicin or resiniferatoxin), TM helicesS1 to S4 form a bundle that remains quiescent during channel activation, highlighting differences in the gating mechanism of TRPV1 and voltage-gated ion channels. Here, however, we argue that the structural dynamics rather than quiescence of S1-S4 domains is necessary for capsaicin-mediated activation of TRPV1. Using fluorescent unnatural amino acid (flUAA) incorporation and voltage-clamp fluorometry (VCF) analysis, we directly observed allostery of the S1-S4 bundle upon capsaicin binding. Covalent occupation of VCF-identified sites, single-channel recording, cell apoptosis analysis, and exploration of the role of PSFL828, a novel non-vanilloid agonist we identified, have collectively confirmed the essential role of this coordinated S1-S4 motility in capsaicin-mediated activation of TRPV1. This study concludes that, in contrast to cryo-EM structural studies, vanilloid agonists are also required for S1-S4 movement during TRPV1 activation. Redefining the gating process of vanilloid agonists and the discovery of new non-vanilloid agonists will allow the evaluation of new strategies aimed at the development of TRPV1 modulators.
Collapse
Affiliation(s)
- Meng-Yang Sun
- School of Life Sciences and Key Laboratory of Preclinical Study for New Drugs of Gansu Province, School of Basic Medical Sciences, Lanzhou University, Lanzhou 730000, China; Department of Pharmacology and Chemical Biology, Institute of Medical Sciences, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China; Department of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing 210009, China
| | - Xue Zhang
- Department of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing 210009, China
| | - Peng-Cheng Yu
- Department of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing 210009, China
| | - Di Liu
- Department of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing 210009, China
| | - Yang Yang
- Department of Pharmacology and Chemical Biology, Institute of Medical Sciences, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China; Department of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing 210009, China
| | - Wen-Wen Cui
- Department of Pharmacology and Chemical Biology, Institute of Medical Sciences, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Xiao-Na Yang
- Department of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing 210009, China; College of Bioscience and Biotechnology, Hunan Agricultural University, Changsha 410128, China
| | - Yun-Tao Lei
- Department of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing 210009, China
| | - Xing-Hua Li
- Department of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing 210009, China
| | - Wen-Hui Wang
- Department of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing 210009, China
| | - Peng Cao
- Hospital of Integrated Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing 210023, China
| | - Heng-Shan Wang
- State Key Laboratory for Chemistry and Molecular Engineering of Medicinal Resources, Collaborative Innovation Center for Guangxi Ethnic Medicine, School of Chemistry and Pharmaceutical Sciences, Guangxi Normal University, Guilin 541004, China
| | - Michael X Zhu
- Department of Integrative Biology and Pharmacology, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, TX 77030, USA
| | - Chang-Zhu Li
- State Key Laboratory of Utilization of Woody Oil Resource, Hunan Academy of Forestry, Changsha 410004, China
| | - Rui Wang
- School of Life Sciences and Key Laboratory of Preclinical Study for New Drugs of Gansu Province, School of Basic Medical Sciences, Lanzhou University, Lanzhou 730000, China.
| | - Ying-Zhe Fan
- Putuo Hospital, Shanghai University of Chinese Traditional Medicine, Shanghai 200062, China.
| | - Ye Yu
- Department of Pharmacology and Chemical Biology, Institute of Medical Sciences, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China; Department of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing 210009, China.
| |
Collapse
|
49
|
Abstract
Native mass spectrometry (nMS) has emerged as an important tool in studying the structure and function of macromolecules and their complexes in the gas phase. In this review, we cover recent advances in nMS and related techniques including sample preparation, instrumentation, activation methods, and data analysis software. These advances have enabled nMS-based techniques to address a variety of challenging questions in structural biology. The second half of this review highlights recent applications of these technologies and surveys the classes of complexes that can be studied with nMS. Complementarity of nMS to existing structural biology techniques and current challenges in nMS are also addressed.
Collapse
Affiliation(s)
- Kelly R Karch
- Department of Chemistry and Biochemistry, The Ohio State University, Columbus, Ohio, USA;
- Resource for Native Mass Spectrometry Guided Structural Biology, The Ohio State University, Columbus, Ohio, USA
| | - Dalton T Snyder
- Resource for Native Mass Spectrometry Guided Structural Biology, The Ohio State University, Columbus, Ohio, USA
| | - Sophie R Harvey
- Department of Chemistry and Biochemistry, The Ohio State University, Columbus, Ohio, USA;
- Resource for Native Mass Spectrometry Guided Structural Biology, The Ohio State University, Columbus, Ohio, USA
| | - Vicki H Wysocki
- Department of Chemistry and Biochemistry, The Ohio State University, Columbus, Ohio, USA;
- Resource for Native Mass Spectrometry Guided Structural Biology, The Ohio State University, Columbus, Ohio, USA
| |
Collapse
|
50
|
Domene C, Darré L, Oakes V, Gonzalez-Resines S. A Potential Route of Capsaicin to Its Binding Site in the TRPV1 Ion Channel. J Chem Inf Model 2022; 62:2481-2489. [PMID: 35504659 PMCID: PMC9131452 DOI: 10.1021/acs.jcim.1c01441] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
Transient receptor potential (TRP) ion channels are important pharmacological targets because of their role in the perception of pain, and so, understanding their chemical regulation is essential for the development of analgesic drugs. Among the currently known TRP channel chemical agonists, capsaicin, the active compound of chili pepper, is probably the most exhaustively studied. The availability of the three-dimensional structure of the vanilloid receptor 1 (TRPV1) has fueled computational studies revealing the molecular details of capsaicin binding modes. Although this is a significant step, a comprehensible binding mechanism or pathway is invaluable for targeting TRP channels in modern pharmacology. In the present work, free-energy and enhanced sampling techniques have been used to explore a possible membrane-mediated pathway for capsaicin to enter the TRPV1 binding pocket where capsaicin accesses the protein starting at the extracellular milieu through the outer leaflet and into its binding site in the protein. The main states visited along this route have been characterized and include (i) a bound state in agreement with the binding mode "head-down, tail-up" and (ii) an alternative state corresponding to a "head-up, tail-down" binding mode. In agreement with previous reports, binding is mediated by both hydrogen bonds and van der Waals interactions, and residue Y511 is crucial for stabilizing the bound state and during the binding process. Together, these results provide a foundation to further understand TRPV channels, and they could be used to guide therapeutic design of selective inhibitors potentially leading to novel avenues for pharmacological applications targeting the TRPV1 channel.
Collapse
Affiliation(s)
- Carmen Domene
- Department of Chemistry, King's College London, Britannia House, 7 Trinity Street, London SE1 1DB, UK.,Department of Chemistry, University of Bath, 1 South Building, Claverton Down, Bath BA2 7AY, UK.,Chemistry Research Laboratory, University of Oxford, Mansfield Road, Oxford OX1 3TA, UK
| | - Leonardo Darré
- Department of Chemistry, King's College London, Britannia House, 7 Trinity Street, London SE1 1DB, UK
| | - Victoria Oakes
- Department of Chemistry, University of Bath, 1 South Building, Claverton Down, Bath BA2 7AY, UK
| | - Saul Gonzalez-Resines
- Department of Chemistry, University of Bath, 1 South Building, Claverton Down, Bath BA2 7AY, UK
| |
Collapse
|