1
|
Ren C, Xu Q, Luo Q, Qiao X, Ding T, Wang W, Zeng X, Chen C, Xiao Y, Hong X. Benzothiazole amide analogues as antagonists of TRPC 6 channels: A therapeutic approach for kidney fibrosis. Eur J Med Chem 2025; 291:117628. [PMID: 40267878 DOI: 10.1016/j.ejmech.2025.117628] [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: 02/06/2025] [Revised: 04/09/2025] [Accepted: 04/10/2025] [Indexed: 04/25/2025]
Abstract
Transient receptor potential canonical 6 (TRPC6) channels, which function as receptor-operated, non-selective cation channels, are widely expressed in the kidney, lungs, and brain. Within these organs, they play crucial roles in regulating diverse physiological processes and contribute to the pathogenesis of various disorders. The resolution of the cryo-electron microscopy structure of TRPC6 has significantly advanced our understanding of its molecular mechanisms, thereby providing a robust platform for structure-based drug design. Building upon compound 1S as a lead, we developed and synthesized a series of benzothiazole derivatives, ultimately identifying compound X26 as a potent TRPC6 antagonist with an IC50 of 0.97 μM. In vitro administration of X26 significantly suppressed TGF-β1-induced myofibroblast differentiation in HK-2 cells, as evidenced by a reduced expression of α-SMA, collagen I, and fibronectin. Furthermore, in a unilateral ureteral obstruction (UUO)-induced kidney fibrosis mouse model, treatment with X26 resulted in a substantial reduction in serum urea nitrogen, serum creatinine, and urinary protein levels, as well as a decrease in renal collagen deposition. These findings establish X26 as a promising lead for the development of TRPC6 antagonists and therapeutic interventions for kidney fibrosis.
Collapse
Affiliation(s)
- Chunlin Ren
- Department of Cardiology, Zhongnan Hospital of Wuhan University, School of Pharmaceutical Sciences, Wuhan University, Wuhan, 430071, China; State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China; Shandong Laboratory of Yantai Drug Discovery, Bohai Rim Advanced Research Institute for Drug Discovery, Yantai, 264117, China
| | - Qiding Xu
- Department of Cardiology, Zhongnan Hospital of Wuhan University, School of Pharmaceutical Sciences, Wuhan University, Wuhan, 430071, China; State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China; Shandong Laboratory of Yantai Drug Discovery, Bohai Rim Advanced Research Institute for Drug Discovery, Yantai, 264117, China
| | - Qiusi Luo
- Department of Cardiology, Zhongnan Hospital of Wuhan University, School of Pharmaceutical Sciences, Wuhan University, Wuhan, 430071, China; State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China; Shandong Laboratory of Yantai Drug Discovery, Bohai Rim Advanced Research Institute for Drug Discovery, Yantai, 264117, China
| | - Xue Qiao
- Department of Cardiology, Zhongnan Hospital of Wuhan University, School of Pharmaceutical Sciences, Wuhan University, Wuhan, 430071, China; State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China; Shandong Laboratory of Yantai Drug Discovery, Bohai Rim Advanced Research Institute for Drug Discovery, Yantai, 264117, China
| | - Taotao Ding
- Department of Cardiology, Zhongnan Hospital of Wuhan University, School of Pharmaceutical Sciences, Wuhan University, Wuhan, 430071, China; State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China; Shandong Laboratory of Yantai Drug Discovery, Bohai Rim Advanced Research Institute for Drug Discovery, Yantai, 264117, China
| | - Wumei Wang
- Department of Cardiology, Zhongnan Hospital of Wuhan University, School of Pharmaceutical Sciences, Wuhan University, Wuhan, 430071, China; Shenzhen Institute of Wuhan University, Shenzhen, 518057, China
| | - Xiaodong Zeng
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China; Shandong Laboratory of Yantai Drug Discovery, Bohai Rim Advanced Research Institute for Drug Discovery, Yantai, 264117, China.
| | - Cheng Chen
- Division of Nephrology, Renmin Hospital of Wuhan University, Wuhan, 430060, China.
| | - Yuling Xiao
- Department of Cardiology, Zhongnan Hospital of Wuhan University, School of Pharmaceutical Sciences, Wuhan University, Wuhan, 430071, China; State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China; Shandong Laboratory of Yantai Drug Discovery, Bohai Rim Advanced Research Institute for Drug Discovery, Yantai, 264117, China.
| | - Xuechuan Hong
- Department of Cardiology, Zhongnan Hospital of Wuhan University, School of Pharmaceutical Sciences, Wuhan University, Wuhan, 430071, China; State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China; Shandong Laboratory of Yantai Drug Discovery, Bohai Rim Advanced Research Institute for Drug Discovery, Yantai, 264117, China; Shenzhen Institute of Wuhan University, Shenzhen, 518057, China.
| |
Collapse
|
2
|
Bai L, Xiang Y, Shen M, Han Y, Li P, Zuo Z, Li Y. Design, synthesis and activity evaluation of novel quinazolinone compounds as TRPC5 inhibitors. Bioorg Chem 2025; 155:108147. [PMID: 39817997 DOI: 10.1016/j.bioorg.2025.108147] [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: 11/16/2024] [Revised: 12/31/2024] [Accepted: 01/05/2025] [Indexed: 01/18/2025]
Abstract
The TRPC5 channel plays an important role in regulating various physiological processes, which is related to various human diseases, especially psychiatric and kidney diseases. Although the TRPC5 channel is one of the essential potential target, no drugs against TRPC5 channels have been granted in the market to date. In this study, based on the structure of hit compound ph1, we further synthesied 49 compounds of novel quinazolinone and heterocyclic fusion pyrimidinone derivatives, and their activities were evaluated by electrophysiological assays. After extensive screening, 21 compounds exhibited significant TRPC5 inhibitory activity, and compounds ph8 and ph14 displayed strong inhibitory with IC50 of 1.28 and 2.16 μM, respectively. These identified potential TRPC5 inhibitor may provide lead compounds and experimental evidence for the development of novel TRPC5 inhibitors with potential treatment for anxiety, depression, and progressive kidney disease.
Collapse
Affiliation(s)
- Longhui Bai
- School of Pharmacology Science and Technology, Hangzhou Institute for Advanced Study, University of Chinese Academy of Sciences, Hangzhou 310024 China; State Key Laboratory of Phytochemistry and Plant Resources in West China, Kunming Institute of Botany, Chinese Academy of Sciences, Kunming 650201 China; University of the Chinese Academy of Sciences, Beijing 100049 China
| | - Yu Xiang
- School of Chinese Materia Medica, Nanjing University of Chinese Medicine, Nanjing 210023 China; State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203 China
| | - Meiling Shen
- School of Pharmacology Science and Technology, Hangzhou Institute for Advanced Study, University of Chinese Academy of Sciences, Hangzhou 310024 China; State Key Laboratory of Phytochemistry and Plant Resources in West China, Kunming Institute of Botany, Chinese Academy of Sciences, Kunming 650201 China; University of the Chinese Academy of Sciences, Beijing 100049 China
| | - Yujun Han
- School of Chinese Materia Medica, Nanjing University of Chinese Medicine, Nanjing 210023 China; State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203 China
| | - Penghua Li
- School of Pharmacology Science and Technology, Hangzhou Institute for Advanced Study, University of Chinese Academy of Sciences, Hangzhou 310024 China; State Key Laboratory of Phytochemistry and Plant Resources in West China, Kunming Institute of Botany, Chinese Academy of Sciences, Kunming 650201 China; University of the Chinese Academy of Sciences, Beijing 100049 China
| | - Zhili Zuo
- School of Pharmacology Science and Technology, Hangzhou Institute for Advanced Study, University of Chinese Academy of Sciences, Hangzhou 310024 China; State Key Laboratory of Phytochemistry and Plant Resources in West China, Kunming Institute of Botany, Chinese Academy of Sciences, Kunming 650201 China; University of the Chinese Academy of Sciences, Beijing 100049 China.
| | - Yang Li
- School of Chinese Materia Medica, Nanjing University of Chinese Medicine, Nanjing 210023 China; State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203 China; University of the Chinese Academy of Sciences, Beijing 100049 China; National Clinical Research Center for Aging and Medicine, Huashan Hospital, Fudan University, Shanghai 200040 China.
| |
Collapse
|
3
|
Khare P, Chand J, Ptakova A, Liguori R, Ferrazzi F, Bishnoi M, Vlachova V, Zimmermann K. The TRPC5 receptor as pharmacological target for pain and metabolic disease. Pharmacol Ther 2024; 263:108727. [PMID: 39384022 DOI: 10.1016/j.pharmthera.2024.108727] [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: 06/06/2024] [Revised: 09/11/2024] [Accepted: 10/03/2024] [Indexed: 10/11/2024]
Abstract
The transient receptor potential canonical (TRPC) channels are a group of highly homologous nonselective cation channels from the larger TRP channel family. They have the ability to form homo- and heteromers with varying degrees of calcium (Ca2+) permeability and signalling properties. TRPC5 is the one cold-sensitive among them and likewise facilitates the influx of extracellular Ca2+ into cells to modulate neuronal depolarization and integrate various intracellular signalling pathways. Recent research with cryo-electron microscopy revealed its structure, along with clear insight into downstream signalling and protein-protein interaction sites. Investigations using global and conditional deficient mice revealed the involvement of TRPC5 in metabolic diseases, energy balance, thermosensation and conditions such as osteoarthritis, rheumatoid arthritis, and inflammatory pain including opioid-induced hyperalgesia and hyperalgesia following tooth decay and pulpitis. This review provides an update on recent advances in our understanding of the role of TRPC5 with focus on metabolic diseases and pain.
Collapse
Affiliation(s)
- Pragyanshu Khare
- Department of Anesthesiology, Friedrich Alexander Universität Erlangen-Nürnberg, Erlangen, Germany; Department of Pharmacy, Birla Institute of Technology and Science Pilani, Pilani Campus, Rajasthan 333031, India
| | - Jagdish Chand
- Department of Pharmacy, Birla Institute of Technology and Science Pilani, Pilani Campus, Rajasthan 333031, India
| | - Alexandra Ptakova
- Department of Cellular Neurophysiology, Institute of Physiology, Czech Academy of Sciences, Prague, Czech Republic
| | - Renato Liguori
- Department of Nephropathology, Institute of Pathology, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany; Institute of Pathology, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
| | - Fulvia Ferrazzi
- Department of Nephropathology, Institute of Pathology, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany; Institute of Pathology, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
| | - Mahendra Bishnoi
- TR(i)P for Health Laboratory Centre for Excellence in Functional Foods, Food & Nutrition Biotechnology Division, National Agri-Food Biotechnology Institute, S.A.S Nagar, Sector (Knowledge City), Punjab, India
| | - Viktorie Vlachova
- Department of Cellular Neurophysiology, Institute of Physiology, Czech Academy of Sciences, Prague, Czech Republic
| | - Katharina Zimmermann
- Department of Anesthesiology, Friedrich Alexander Universität Erlangen-Nürnberg, Erlangen, Germany.
| |
Collapse
|
4
|
Xu Y, Ren Y, Zhang J, Niu B, Liu M, Xu T, Zhang X, Shen J, Wang K, Cao Z. Discovery of pyridazinone derivatives bearing tetrahydroimidazo[1,2-a]pyrazine scaffold as potent inhibitors of transient receptor potential canonical 5 to ameliorate hypertension-induced renal injury in rats. Eur J Med Chem 2024; 275:116565. [PMID: 38878518 DOI: 10.1016/j.ejmech.2024.116565] [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: 03/13/2024] [Revised: 05/30/2024] [Accepted: 06/01/2024] [Indexed: 07/12/2024]
Abstract
Transient receptor potential canonical 5 (TRPC5) is a calcium-permeable non-selective cation channel involved in various pathophysiological processes, including renal injury. Recently, GFB-887, an investigational pyridazinone TRPC5 inhibitor, demonstrated significant therapeutic potential in a Phase II clinical trial for focal segmental glomerulosclerosis (FSGS), a rare and severe form of chronic kidney disease (CKD). In the current study, based on the structure of GFB-887, we conducted extensive structural modification to explore novel TRPC5 inhibitors with desirable drug-like properties and robust nephroprotective efficacy. A series of pyridazinone derivatives featuring a novel tetrahydroimidazo[1,2-a]pyrazine scaffold were synthesized and their activities were evaluated in HEK-293 cells stably expressing TRPC5 using a fluorescence-based Ca2+ mobilization assay. Among these compounds, compound 12 is turned out to be a potent TRPC5 inhibitor with apparent affinity comparable to the parent compound GBF-887. Compound 12 is highly selective on TRPC4/5 over TRPC3/6/7 and hERG channels, along with acceptable pharmacokinetic properties and a favorable safety profile. More importantly, in a rat model of hypertension-induced renal injury, oral administration of compound 12 (10 mg/kg, BID) efficaciously reduced mean blood pressure, inhibited proteinuria, and protected podocyte damage. These findings further confirmed the potential of TRPC5 inhibitors on the CKD treatment and provided compound 12 to be a valuable tool for exploring TRPC4/5 pathophysiology.
Collapse
Affiliation(s)
- Yuanyuan Xu
- School of Chinese Materia Medica, Nanjing University of Chinese Medicine, Nanjing, 210046, China; State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China
| | - Younan Ren
- State Key Laboratory of Natural Medicines and Jiangsu Provincial Key Laboratory for TCM Evaluation and Translational Development, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing, Jiangsu, 211198, China
| | - Jie Zhang
- State Key Laboratory of Natural Medicines and Jiangsu Provincial Key Laboratory for TCM Evaluation and Translational Development, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing, Jiangsu, 211198, China
| | - Bo Niu
- State Key Laboratory of Natural Medicines and Jiangsu Provincial Key Laboratory for TCM Evaluation and Translational Development, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing, Jiangsu, 211198, China
| | - Mengru Liu
- State Key Laboratory of Natural Medicines and Jiangsu Provincial Key Laboratory for TCM Evaluation and Translational Development, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing, Jiangsu, 211198, China
| | - Tifei Xu
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China
| | - Xian Zhang
- State Key Laboratory of Natural Medicines and Jiangsu Provincial Key Laboratory for TCM Evaluation and Translational Development, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing, Jiangsu, 211198, China
| | - Jianhua Shen
- School of Chinese Materia Medica, Nanjing University of Chinese Medicine, Nanjing, 210046, China; State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China
| | - Kai Wang
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China.
| | - Zhengyu Cao
- State Key Laboratory of Natural Medicines and Jiangsu Provincial Key Laboratory for TCM Evaluation and Translational Development, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing, Jiangsu, 211198, China.
| |
Collapse
|
5
|
Umlauf F, Diebolt CM, Englisch CN, Flockerzi F, Tschernig T. Distribution of TRPC5 in the human lung: A study in body donors. Exp Ther Med 2024; 28:363. [PMID: 39071908 PMCID: PMC11273251 DOI: 10.3892/etm.2024.12652] [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] [Received: 02/07/2024] [Accepted: 06/13/2024] [Indexed: 07/30/2024] Open
Abstract
Transient receptor potential channel canonical 5 (TRPC5) is a non-selective ion channel; ion influx through TRPC5 causes activation of downstream signaling pathways. In addition, TRPC5 has been identified as having a potential role in pathological processes, particularly in diseases caused by cellular cation homeostasis dysregulation, such as bronchial asthma or pulmonary hypertension. However, the expression and distribution of TRPC5 in the human lung remain unclear. To date, TRPC5 has only been detected in a few cell types in the human lung, such as airway, pulmonary venous and arterial smooth muscle cells. The present study therefore aimed to investigate the protein expression of TRPC5 in the human lung and to evaluate its histological distribution. Human lung samples were obtained from six preserved body donors. After processing, both hematoxylin & eosin staining, as well as immunohistochemistry were performed. Microscopic analysis revealed medium to strong immunostaining signals in all lung structures examined, including the pleura, pulmonary arteries and veins, bronchioles, alveolar septa, type 1 and 2 pneumocytes, as well as alveolar macrophages. Current research suggests that TRPC5 may be involved in various pathological processes in the human lung and some pharmacological compounds have already been identified that affect the function of TRPC5. Therefore, TRPC5 may present a novel drug target for therapeutic intervention in various lung diseases. The results of the present study indicate that the TRPC5 protein is expressed in all examined histological structures of the human lung. These findings suggest that TRPC5 may be more important for physiological cell function and pathophysiological cell dysfunction in the lung than is currently known. Further research is needed to explore the role and therapeutic target potential of TRPC5 in the human lung.
Collapse
Affiliation(s)
- Frederik Umlauf
- Institute of Anatomy and Cell Biology, Saarland University, Faculty of Medicine, D-66421 Homburg, Germany
| | - Coline M. Diebolt
- Institute of Anatomy and Cell Biology, Saarland University, Faculty of Medicine, D-66421 Homburg, Germany
| | - Colya N. Englisch
- Institute of Anatomy and Cell Biology, Saarland University, Faculty of Medicine, D-66421 Homburg, Germany
| | - Fidelis Flockerzi
- Institute of Pathology, Saarland University, Faculty of Medicine, D-66421 Homburg, Germany
| | - Thomas Tschernig
- Institute of Anatomy and Cell Biology, Saarland University, Faculty of Medicine, D-66421 Homburg, Germany
| |
Collapse
|
6
|
Huang B, Zhang Y, Sun P, Yuan Y, Wang C. MiR-138-5p Inhibits Thyroid Cancer Cell Growth and Stemness by Targeting TRPC5/Wnt/β-Catenin Pathway. Mol Biotechnol 2024; 66:544-553. [PMID: 37278959 DOI: 10.1007/s12033-023-00782-3] [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: 01/24/2023] [Accepted: 05/24/2023] [Indexed: 06/07/2023]
Abstract
MicroRNAs play a key role in the pathogenesis of many types of cancer, including thyroid cancer (TC). MiR-138-5p has been confirmed to be abnormally expressed in TC tissues. However, the role of miR-138-5p in TC progression and its potential molecular mechanism need to be further explored. In this study, quantitative real-time PCR was used to examine miR-138-5p and TRPC5 expression, and western blot analysis was performed to examine the protein levels of TRPC5, stemness-related markers, and Wnt pathway-related markers. Dual-luciferase reporter assay was used to assess the interaction between miR-138-5p and TRPC5. Cell proliferation, stemness, and apoptosis were examined using colony formation assay, sphere formation assay, and flow cytometry. Our data showed that miR-138-5p could target TRPC5 and its expression was negatively correlated with TRPC5 expression in TC tumor tissues. MiR-138-5p decreased proliferation, stemness, and promoted gemcitabine-induced apoptosis in TC cells, and this effect could be reversed by TRPC5 overexpression. Moreover, TRPC5 overexpression abolished the inhibitory effect of miR-138-5p on the activity of Wnt/β-catenin pathway. In conclusion, our data showed that miR-138-5p suppressed TC cell growth and stemness via the regulation of TRPC5/Wnt/β-catenin pathway, which provided some guidance for studying the potential function of miR-138-5p in TC progression.
Collapse
Affiliation(s)
- Bo Huang
- Department of General Surgery, The First Affiliated Hospital of Jinan University, No.613, Huangpu Street, Guangzhou, 510000, China
| | - YiChao Zhang
- Department of General Surgery, The First Affiliated Hospital of Jinan University, No.613, Huangpu Street, Guangzhou, 510000, China
| | - Peng Sun
- Department of General Surgery, The First Affiliated Hospital of Jinan University, No.613, Huangpu Street, Guangzhou, 510000, China
| | - YuanYuan Yuan
- Department of General Surgery, The First Affiliated Hospital of Jinan University, No.613, Huangpu Street, Guangzhou, 510000, China
| | - CunChuan Wang
- Department of General Surgery, The First Affiliated Hospital of Jinan University, No.613, Huangpu Street, Guangzhou, 510000, China.
| |
Collapse
|
7
|
Saqib U, Demaree IS, Obukhov AG, Baig MS, Khan MS, Altwaijry N, Nasution MAF, Mizuguchi K, Hajela K. Structural and accessibility studies highlight the differential binding of clemizole to TRPC5 and TRPC6. J Biomol Struct Dyn 2024:1-14. [PMID: 38279926 PMCID: PMC11412694 DOI: 10.1080/07391102.2024.2306198] [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: 10/10/2023] [Accepted: 01/07/2024] [Indexed: 01/29/2024]
Abstract
Transient Receptor Potential Canonical 5 (T RP C5) and T RP C6 channels play critical physiological roles in various cell types. Their involvement in numerous disease progression mechanisms has led to extensive searches for their inhibitors. Although several potent T RP C inhibitors have been developed and the structure of their binding sites were mapped using cryo electron microscopy, a comprehensive understanding of the molecular interactions within the inhibitor binding site of T RP Cs remains elusive. This study aimed to decipher the structural determinants and molecular mechanisms contributing to the differential binding of clemizole to T RP C5 and T RP C6, with a particular focus on the accessibility of binding site residues. This information can help better understand what molecular features allow for selective binding, which is a key characteristic of clinically effective pharmacological agents. Using computational methodologies, we conducted an in-depth molecular docking analysis of clemizole with T RP C5 and T RP C6 channels. The protein structures were retrieved from publicly accessible protein databases. Discovery Studio 2020 Client Visualizer and Chimera software facilitated our in-silico mutation experiments and enabled us to identify the critical structural elements influencing clemizole binding. Our study reveals key molecular determinants at the clemizole binding site, specifically outlining the role of residues' Accessible Surface Area (ASA) and Relative Accessible Surface Area (RASA) in differential binding. We found that lower accessibility of T RP C6 binding site residues, compared to those in T RP C5, could account for the lower affinity binding of clemizole to T RP C6. This work illuminates the pivotal role of binding site residue accessibility in determining the affinity of clemizole to T RP C5 and T RP C6. A nuanced understanding of the distinct binding properties between these homologous proteins may pave the way for the development of more selective inhibitors, promising improved therapeutic efficacy and fewer off-target effects. By demystifying the structural and molecular subtleties of T RP C inhibitors, this research could significantly accelerate the drug discovery process, offering hope to patients afflicted with T RP C-related diseases.
Collapse
Affiliation(s)
- Uzma Saqib
- School of Life Sciences, Devi Ahilya Vishwavidyalaya, Indore, MP, India
| | - Isaac S Demaree
- Department of Anatomy, Cell Biology & Physiology, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Alexander G Obukhov
- Department of Anatomy, Cell Biology & Physiology, Indiana University School of Medicine, Indianapolis, IN, USA
- Stark Neurosciences Research Institute, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Mirza S Baig
- Department of Biosciences and Biomedical Engineering (BSBE), Indian Institute of Technology Indore (IITI), Indore, India
| | - Mohd Shahnawaz Khan
- Department of Biochemistry, College of Science, King Saud University, Riyadh, Saudi Arabia
| | - Nojood Altwaijry
- Department of Biochemistry, College of Science, King Saud University, Riyadh, Saudi Arabia
| | - Mochammad Arfin Fardiansyah Nasution
- Institute for Protein Research, Osaka University, Osaka, Japan
- Artificial Intelligence Center for Health and Biomedical Research, National Institutes of Biomedical Innovation, Health and Nutrition, Osaka, Japan
| | - Kenji Mizuguchi
- Institute for Protein Research, Osaka University, Osaka, Japan
- Artificial Intelligence Center for Health and Biomedical Research, National Institutes of Biomedical Innovation, Health and Nutrition, Osaka, Japan
| | - Krishnan Hajela
- School of Life Sciences, Devi Ahilya Vishwavidyalaya, Indore, MP, India
| |
Collapse
|
8
|
Anand S, Rajagopal S. A Comprehensive Review on the Regulatory Action of TRP Channels: A Potential Therapeutic Target for Nociceptive Pain. Neurosci Insights 2023; 18:26331055231220340. [PMID: 38146332 PMCID: PMC10749524 DOI: 10.1177/26331055231220340] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2023] [Accepted: 11/28/2023] [Indexed: 12/27/2023] Open
Abstract
The transient receptor potential (TRP) superfamily of ion channels in humans comprises voltage-gated, non-selective cation channels expressed both in excitable as well as non-excitable cells. Four TRP channel subunits associate to create functional homo- or heterotetramers that allow the influx of calcium, sodium, and/or potassium. These channels are highly abundant in the brain and kidney and are important mediators of diverse biological functions including thermosensation, vascular tone, flow sensing in the kidney and irritant stimuli sensing. Inherited or acquired dysfunction of TRP channels influences cellular functions and signaling pathways resulting in multifaceted disorders affecting skeletal, renal, cardiovascular, and nervous systems. Studies have demonstrated the involvement of these channels in the generation and transduction of pain. Based on the multifaceted role orchestrated by these TRP channels, modulation of the activity of these channels presents an important strategy to influence cellular function by regulating intracellular calcium levels as well as membrane excitability. Therefore, there has been a remarkable pharmaceutical inclination toward TRP channels as therapeutic interventions. Several candidate drugs influencing the activity of these channels are already in the clinical trials pipeline. The present review encompasses the current understanding of TRP channels and TRP modulators in pain and pain management.
Collapse
Affiliation(s)
- Santosh Anand
- Department of Biotechnology, School of Applied Sciences, REVA University, Bengaluru, Karnataka, India
| | - Senthilkumar Rajagopal
- Department of Biotechnology, School of Applied Sciences, REVA University, Bengaluru, Karnataka, India
| |
Collapse
|
9
|
Shen M, Li L, Li Y, Gu X, Bai L, Xia C, Xiong W, Zuo Z. Discovery of potential novel TRPC5 inhibitors by virtual screening and bioassay. Bioorg Med Chem 2023; 94:117477. [PMID: 37738708 DOI: 10.1016/j.bmc.2023.117477] [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: 07/03/2023] [Revised: 08/22/2023] [Accepted: 09/13/2023] [Indexed: 09/24/2023]
Abstract
The transient receptor potential canonical channel 5 (TRPC5), a member of the TRPC family, plays a crucial role in the regulation of various physiological activities and diseases, including those related to the central nervous system, cardiovascular system, kidney, and cancer. As a nonselective cation channel, TRPC5 mainly controls the influx of extracellular Ca2+ into cells, thereby modulating cellular depolarization and intracellular ion concentration. Inhibition of TRPC5 by small molecules presents a promising approach for the treatment of TRPC5-associated diseases. In this study, we conducted a comprehensive virtual screening of more than 1.5 million molecules from the Chemdiv database (https://www.chemdiv.com) to identify potential inhibitors of hTRPC5, utilizing the published structures and binding sites of hTRPC5 as a basis. Lipinski's rule, Veber's rule, PAINS filters, pharmacophore analysis, molecular docking, ADMET evaluation and cluster analysis methods were applied for the screening. From this rigorous screening process, 18 candidates exhibiting higher affinities to hTRPC5 were subsequently evaluated for their inhibitory effects on Ca2+ influx using a fluorescence-based assay. Notably, two molecules, namely SML-1 and SML-13, demonstrated significant inhibition of intracellular Ca2+ levels in hTRPC5-overexpressing HEK 293T cells, with IC50 values of 10.2 μM and 10.3 μM, respectively. These findings highlight SML-1 and SML-13 as potential lead molecules for the development of therapeutics targeting hTRPC5 and its associated physiological activities and diseases.
Collapse
Affiliation(s)
- Meiling Shen
- State Key Laboratory of Phytochemistry and Plant Resources in West China, Kunming Institute of Botany, Chinese Academy of Sciences, Kunming 650201, China; University of the Chinese Academy of Sciences, Beijing 100049, China
| | - Lingfeng Li
- Key Laboratory of Medicinal Chemistry for Natural Resource (Ministry of Education), School of Pharmacy, Yunnan University, Kunming 650091, China
| | - Yue Li
- State Key Laboratory of Phytochemistry and Plant Resources in West China, Kunming Institute of Botany, Chinese Academy of Sciences, Kunming 650201, China
| | - Xi Gu
- State Key Laboratory of Phytochemistry and Plant Resources in West China, Kunming Institute of Botany, Chinese Academy of Sciences, Kunming 650201, China; University of the Chinese Academy of Sciences, Beijing 100049, China
| | - Longhui Bai
- State Key Laboratory of Phytochemistry and Plant Resources in West China, Kunming Institute of Botany, Chinese Academy of Sciences, Kunming 650201, China; University of the Chinese Academy of Sciences, Beijing 100049, China
| | - Chengfeng Xia
- Key Laboratory of Medicinal Chemistry for Natural Resource (Ministry of Education), School of Pharmacy, Yunnan University, Kunming 650091, China
| | - Wenyong Xiong
- Key Laboratory of Medicinal Chemistry for Natural Resource (Ministry of Education), School of Pharmacy, Yunnan University, Kunming 650091, China.
| | - Zhili Zuo
- State Key Laboratory of Phytochemistry and Plant Resources in West China, Kunming Institute of Botany, Chinese Academy of Sciences, Kunming 650201, China; University of the Chinese Academy of Sciences, Beijing 100049, China.
| |
Collapse
|
10
|
Yu Y, Jiang H, Liang Q, Qiu L, Huang T, Hu H, Bolshakov VY, Perlmutter JS, Tu Z. Radiosynthesis and Evaluation of a C-11 Radiotracer for Transient Receptor Potential Canonical 5 in the Brain. Mol Imaging Biol 2023; 25:334-342. [PMID: 35951211 PMCID: PMC9918595 DOI: 10.1007/s11307-022-01760-y] [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: 04/20/2022] [Revised: 07/12/2022] [Accepted: 07/14/2022] [Indexed: 10/15/2022]
Abstract
PURPOSE TRPC5 belongs to the mammalian superfamily of transient receptor potential (TRP) Ca2+-permeable cationic channels and it has been implicated in various CNS disorders. As part of our ongoing interest in the development of a PET radiotracer for imaging TRPC5, herein, we explored the radiosynthesis, and in vitro and in vivo evaluation of a new C-11 radiotracer [11C]HC070 in rodents and nonhuman primates. PROCEDURES [11C]HC070 was radiolabeled utilizing the corresponding precursor and [11C]CH3I via N-methylation protocol. Ex vivo biodistribution study of [11C]HC070 was performed in Sprague-Dawley rats. In vitro autoradiography study was conducted for the rat brain sections to characterize the radiotracer distribution in the brain regionals. MicroPET brain imaging studies of [11C]HC070 were done for 129S1/SvImJ wild-type mice and 129S1/SvImJ TRPC5 knockout mice for 0-60-min dynamic data acquisition after intravenous administration of the radiotracer. Dynamic PET scans (0-120 min) for the brain of cynomolgus male macaques were performed after the radiotracer injection. RESULTS [11C]HC070 was efficiently prepared with good radiochemical yield (45 ± 5%, n = 15), high chemical and radiochemical purity (> 99%), and high molar activity (320.6 ± 7.4 GBq/μmol, 8.6 ± 0.2 Ci/μmol) at the end of bombardment (EOB). Radiotracer [11C]HC070 has good solubility in the aqueous dose solution. The ex vivo biodistribution study showed that [11C]HC070 had a quick rat brain clearance. Autoradiography demonstrated that [11C]HC070 specifically binds to TRPC5-enriched regions in rat brain. MicroPET study showed the peak brain uptake (SUV value) was 0.63 in 129S1/SvImJ TRPC5 knockout mice compared to 1.13 in 129S1/SvImJ wild-type mice. PET study showed that [11C]HC070 has good brain uptake with maximum SUV of ~ 2.2 in the macaque brain, followed by rapid clearance. CONCLUSIONS Our data showed that [11C]HC070 is a TRPC5-specific radiotracer with high brain uptake and good brain washout pharmacokinetics in both rodents and nonhuman primates. The radiotracer is worth further investigating of its suitability to be a PET radiotracer for imaging TRPC5 in animals and human subjects in vivo.
Collapse
Affiliation(s)
- Yanbo Yu
- Department of Radiology, Washington University School of Medicine, St. Louis, MO, 63110, USA
| | - Hao Jiang
- Department of Radiology, Washington University School of Medicine, St. Louis, MO, 63110, USA
| | - Qianwa Liang
- Department of Radiology, Washington University School of Medicine, St. Louis, MO, 63110, USA
| | - Lin Qiu
- Department of Radiology, Washington University School of Medicine, St. Louis, MO, 63110, USA
| | - Tianyu Huang
- Department of Radiology, Washington University School of Medicine, St. Louis, MO, 63110, USA
| | - Hongzhen Hu
- Center for the Study of Itch and Sensory Disorders, Department of Anesthesiology, Washington University School of Medicine, St. Louis, MO, 63110, USA
| | - Vadim Y Bolshakov
- Department of Psychiatry, McLean Hospital, Harvard Medical School, Boston, MA, 02115, USA
| | - Joel S Perlmutter
- Department of Radiology, Washington University School of Medicine, St. Louis, MO, 63110, USA
- Department of Neurology, Washington University School of Medicine, St. Louis, MO, 63110, USA
| | - Zhude Tu
- Department of Radiology, Washington University School of Medicine, St. Louis, MO, 63110, USA.
| |
Collapse
|
11
|
Müller M, Niemeyer K, Urban N, Ojha NK, Zufall F, Leinders‐Zufall T, Schaefer M, Thorn‐Seshold O. BTDAzo: A Photoswitchable TRPC5 Channel Activator. Angew Chem Int Ed Engl 2022; 61:e202201565. [PMID: 35713469 PMCID: PMC9542918 DOI: 10.1002/anie.202201565] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2022] [Indexed: 11/13/2022]
Abstract
Photoswitchable reagents can be powerful tools for high-precision biological control. TRPC5 is a Ca2+ -permeable cation channel with distinct tissue-specific roles, from synaptic function to hormone regulation. Reagents giving spatiotemporally-resolved control over TRPC5 activity may be key to understanding and harnessing its biology. Here we develop the first photoswitchable TRPC5-modulator, BTDAzo, to address this goal. BTDAzo can photocontrol TRPC5 currents in cell culture, as well as controlling endogenous TRPC5-based neuronal Ca2+ responses in mouse brain slices. BTDAzos are also the first reported azo-benzothiadiazines, an accessible and conveniently derivatised azoheteroarene with strong two-colour photoswitching. BTDAzo's ability to control TRPC5 across relevant channel biology settings makes it suitable for a range of dynamically reversible photoswitching studies in TRP channel biology, with the aim to decipher the various biological roles of this centrally important ion channel.
Collapse
Affiliation(s)
- Markus Müller
- Department of PharmacyLMU MunichButenandtstrasse 781377MunichGermany
| | - Konstantin Niemeyer
- Rudolf-Boehm-Institute of Pharmacology and ToxicologyLeipzig UniversityHärtelstraße 16–1804107LeipzigGermany
| | - Nicole Urban
- Rudolf-Boehm-Institute of Pharmacology and ToxicologyLeipzig UniversityHärtelstraße 16–1804107LeipzigGermany
| | - Navin K. Ojha
- Center for Integrative Physiology and Molecular MedicineSaarland UniversityKirrbergerstraße 10066421HomburgGermany
| | - Frank Zufall
- Center for Integrative Physiology and Molecular MedicineSaarland UniversityKirrbergerstraße 10066421HomburgGermany
| | - Trese Leinders‐Zufall
- Center for Integrative Physiology and Molecular MedicineSaarland UniversityKirrbergerstraße 10066421HomburgGermany
| | - Michael Schaefer
- Rudolf-Boehm-Institute of Pharmacology and ToxicologyLeipzig UniversityHärtelstraße 16–1804107LeipzigGermany
| | | |
Collapse
|
12
|
Interactions between the Polysialylated Neural Cell Adhesion Molecule and the Transient Receptor Potential Canonical Channels 1, 4, and 5 Induce Entry of Ca 2+ into Neurons. Int J Mol Sci 2022; 23:ijms231710027. [PMID: 36077460 PMCID: PMC9456277 DOI: 10.3390/ijms231710027] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2022] [Revised: 08/25/2022] [Accepted: 08/26/2022] [Indexed: 11/16/2022] Open
Abstract
The neural cell adhesion molecule (NCAM) plays important functional roles in the developing and mature nervous systems. Here, we show that the transient receptor potential canonical (TRPC) ion channels TRPC1, -4, and -5 not only interact with the intracellular domains of the transmembrane isoforms NCAM140 and NCAM180, but also with the glycan polysialic acid (PSA) covalently attached to the NCAM protein backbone. NCAM antibody treatment leads to the opening of TRPC1, -4, and -5 hetero- or homomers at the plasma membrane and to the influx of Ca2+ into cultured cortical neurons and CHO cells expressing NCAM, PSA, and TRPC1 and -4 or TRPC1 and -5. NCAM-stimulated Ca2+ entry was blocked by the TRPC inhibitor Pico145 or the bacterial PSA homolog colominic acid. NCAM-stimulated Ca2+ influx was detectable neither in NCAM-deficient cortical neurons nor in TRPC1/4- or TRPC1/5-expressing CHO cells that express NCAM, but not PSA. NCAM-induced neurite outgrowth was reduced by TRPC inhibitors and a function-blocking TRPC1 antibody. A characteristic signaling feature was that extracellular signal-regulated kinase 1/2 phosphorylation was also reduced by TRPC inhibitors. Our findings indicate that the interaction of NCAM with TRPC1, -4, and -5 contributes to the NCAM-stimulated and PSA-dependent Ca2+ entry into neurons thereby influencing essential neural functions.
Collapse
|
13
|
Strickland RG, Garner MA, Gross AK, Girkin CA. Remodeling of the Lamina Cribrosa: Mechanisms and Potential Therapeutic Approaches for Glaucoma. Int J Mol Sci 2022; 23:8068. [PMID: 35897642 PMCID: PMC9329908 DOI: 10.3390/ijms23158068] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2022] [Revised: 07/18/2022] [Accepted: 07/19/2022] [Indexed: 11/28/2022] Open
Abstract
Glaucomatous optic neuropathy is the leading cause of irreversible blindness in the world. The chronic disease is characterized by optic nerve degeneration and vision field loss. The reduction of intraocular pressure remains the only proven glaucoma treatment, but it does not prevent further neurodegeneration. There are three major classes of cells in the human optic nerve head (ONH): lamina cribrosa (LC) cells, glial cells, and scleral fibroblasts. These cells provide support for the LC which is essential to maintain healthy retinal ganglion cell (RGC) axons. All these cells demonstrate responses to glaucomatous conditions through extracellular matrix remodeling. Therefore, investigations into alternative therapies that alter the characteristic remodeling response of the ONH to enhance the survival of RGC axons are prevalent. Understanding major remodeling pathways in the ONH may be key to developing targeted therapies that reduce deleterious remodeling.
Collapse
Affiliation(s)
- Ryan G. Strickland
- Department of Neurobiology, University of Alabama at Birmingham, Birmingham, AL 35294, USA; (R.G.S.); (M.A.G.); (A.K.G.)
| | - Mary Anne Garner
- Department of Neurobiology, University of Alabama at Birmingham, Birmingham, AL 35294, USA; (R.G.S.); (M.A.G.); (A.K.G.)
| | - Alecia K. Gross
- Department of Neurobiology, University of Alabama at Birmingham, Birmingham, AL 35294, USA; (R.G.S.); (M.A.G.); (A.K.G.)
| | - Christopher A. Girkin
- Department of Ophthalmology and Vision Sciences, University of Alabama at Birmingham, Birmingham, AL 35294, USA
| |
Collapse
|
14
|
Yang Y, Wei M, Chen L. Structural identification of riluzole-binding site on human TRPC5. Cell Discov 2022; 8:67. [PMID: 35821012 PMCID: PMC9276808 DOI: 10.1038/s41421-022-00410-5] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2022] [Accepted: 03/27/2022] [Indexed: 11/09/2022] Open
Affiliation(s)
- Yaxiong Yang
- Key Laboratory of Biomechanics and Mechanobiology (Beihang University), Ministry of Education, Beijing Advanced Innovation Center for Biomedical Engineering, School of Biological Science and Medical Engineering, Beihang University, Beijing, China
| | - Miao Wei
- State Key Laboratory of Membrane Biology, College of Future Technology, Institute of Molecular Medicine, Peking University, Beijing Key Laboratory of Cardiometabolic Molecular Medicine, Beijing, China
| | - Lei Chen
- State Key Laboratory of Membrane Biology, College of Future Technology, Institute of Molecular Medicine, Peking University, Beijing Key Laboratory of Cardiometabolic Molecular Medicine, Beijing, China.
- Peking-Tsinghua Center for Life Sciences, Peking University, Beijing, China.
- Academy for Advanced Interdisciplinary Studies, Peking University, Beijing, China.
| |
Collapse
|
15
|
Müller M, Niemeyer K, Urban N, Ojha NK, Zufall F, Leinders-Zufall T, Schaefer M, Thorn-Seshold O. BTDAzo ‐ A Photoswitchable TRPC5 Channel Activator. Angew Chem Int Ed Engl 2022. [DOI: 10.1002/ange.202201565] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Affiliation(s)
- Markus Müller
- Ludwig Maximillians University Munich: Ludwig-Maximilians-Universitat Munchen Department of Pharmacy Butenandstr. 7 81377 Munich GERMANY
| | - Konstantin Niemeyer
- Leipzig University: Universitat Leipzig Rudolf-Boehm-Institut für Pharmakologie und Toxikologie Härtelstr. 16-18 04107 Leipzig GERMANY
| | - Nicole Urban
- Leipzig University: Universitat Leipzig Rudolf-Boehm-Institut für Pharmakologie und Toxikologie Härtelstraße 16-18 04107 Leipzig GERMANY
| | - Navin K. Ojha
- Saarland University: Universitat des Saarlandes Center for Integrative Physiology and Molecular Medicine Kirrbergerstraße 100 66421 Homburg GERMANY
| | - Frank Zufall
- Saarland University: Universitat des Saarlandes Center for Integrative Physiology and Molecular Medicine Kirrbergerstraße 100 66421 Homburg GERMANY
| | - Trese Leinders-Zufall
- Saarland University: Universitat des Saarlandes Center for Integrative Physiology and Molecular Medicine Kirrbergerstraße 100 66421 Homburg GERMANY
| | - Michael Schaefer
- Leipzig University: Universitat Leipzig Rudolf-Boehm-Institute of Pharmacology and Toxicology Härtelstraße 16-18 04107 Leipzig GERMANY
| | - Oliver Thorn-Seshold
- Ludwig-Maximilians-Universitat Munchen Dept Pharmacy Butenandtstr 5-13 81377 Munich GERMANY
| |
Collapse
|
16
|
Sharma S, Pablo JL, Tolentino KT, Gallegos W, Hinman J, Beninato M, Asche M, Greka A, Hopkins CR. Further exploration of the benzimidazole scaffold as TRPC5 inhibitors: identification of 1-alkyl-2-(pyrrolidin-1-yl)-1H-benzo[d]imidazoles as potent and selective inhibitors. ChemMedChem 2022; 17:e202200151. [PMID: 35557491 PMCID: PMC9308755 DOI: 10.1002/cmdc.202200151] [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] [Received: 03/24/2022] [Revised: 05/05/2022] [Indexed: 11/29/2022]
Abstract
The transient receptor potential cation channel 5 (TRPC5) plays an important role in numerous cellular processes. Due to this, it has gained considerable attention over the past few years as a potential therapeutic target. Recently, TRPC5 has been shown to be involved in the regulation of podocyte survival, indicating a potential treatment option for chronic kidney disease. In addition, a recent study has shown TRPC5 to be expressed in human sensory neurons and suggests that TRPC5 inhibition could be an effective treatment for spontaneous and tactile pain. To understand these processes more fully, potent and selective tool compounds are needed. Herein we report further exploration of the 2‐aminobenzimidazole scaffold as a potent TRPC5 inhibitor, culminating in the discovery of 16 f as a potent and selective TRPC5 inhibitor.
Collapse
Affiliation(s)
- Swagat Sharma
- University of Nebraska Medical Center College of Pharmacy, Pharmaceutical Sciences, UNITED STATES
| | | | - Kirsten T Tolentino
- University of Nebraska Medical Center College of Pharmacy, Pharmaceutical Sciences, UNITED STATES
| | - Wacey Gallegos
- University of Nebraska Medical Center College of Pharmacy, Pharmaceutical Sciences, UNITED STATES
| | - Jennifer Hinman
- University of Nebraska Medical Center College of Pharmacy, Pharmaceutical Sciences, UNITED STATES
| | - Madison Beninato
- University of Nebraska Medical Center College of Pharmacy, Pharmaceutical Sciences, UNITED STATES
| | - MacKenzie Asche
- University of Nebraska Medical Center College of Pharmacy, Pharmaceutical Sciences, UNITED STATES
| | - Anna Greka
- Broad Institute Harvard: Broad Institute, Pharmacology, UNITED STATES
| | - Corey R Hopkins
- University of Nebraska Medical Center College of Pharmacy, Pharmaceutical Sciences, 986125 Nebraska Medical Center, PDD 3015, 68198-6125, Omaha, UNITED STATES
| |
Collapse
|
17
|
Zhang Z, Chen L, Tian H, Liu M, Jiang S, Shen J, Wang K, Cao Z. Discovery of pyridazinone analogs as potent transient receptor potential canonical channel 5 inhibitors. Bioorg Med Chem Lett 2022; 61:128612. [PMID: 35143983 DOI: 10.1016/j.bmcl.2022.128612] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2021] [Revised: 01/09/2022] [Accepted: 02/04/2022] [Indexed: 11/17/2022]
Abstract
A deepening understanding of the relationship between transient receptor potential canonical channel 5 (TRPC5) and chronic kidney disease (CKD), has led to the emergence of several types of TRPC5 inhibitors displaying clear therapeutic effect. Herein, we report the synthesis and biological evaluation of a series of pyrroledione TRPC5 inhibitors, culminating in the discovery of compound 16g with subtype selectivity. Compared with GFB-8438, a potent TRPC5 inhibitor (Goldfinch Bio), compound 16g showed improved inhibition of TRPC5 and enhanced protective effect against protamine sulfates (PS)-induced podocyte injury in vitro. In addition, compound 16g did not induce cell death in primary cultured hepatocytes and immortalized podocytes in a preliminary toxicity assessment, indicating its utility as a potent and safe inhibitor for studying the function of TRPC5.
Collapse
Affiliation(s)
- Zhuang Zhang
- Jiangsu Key Laboratory of TCM Evaluation and Translational Research, School of Traditional Chinese Pharmacy, China Pharmaceutical University, No. 639 Long Mian Road, Nanjing, Jiangsu 211198, China
| | - Lili Chen
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica (SIMM), Chinese Academy of Sciences, No. 555 Zu Chong Zhi Road, Shanghai 201203, China; School of Pharmacy, University of Chinese Academy of Sciences, No. 19A Yuquan Road, Beijing 100049, China
| | - Hongtao Tian
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica (SIMM), Chinese Academy of Sciences, No. 555 Zu Chong Zhi Road, Shanghai 201203, China
| | - Mengru Liu
- Jiangsu Key Laboratory of TCM Evaluation and Translational Research, School of Traditional Chinese Pharmacy, China Pharmaceutical University, No. 639 Long Mian Road, Nanjing, Jiangsu 211198, China
| | - Shan Jiang
- Jiangsu Key Laboratory of TCM Evaluation and Translational Research, School of Traditional Chinese Pharmacy, China Pharmaceutical University, No. 639 Long Mian Road, Nanjing, Jiangsu 211198, China
| | - Jianhua Shen
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica (SIMM), Chinese Academy of Sciences, No. 555 Zu Chong Zhi Road, Shanghai 201203, China
| | - Kai Wang
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica (SIMM), Chinese Academy of Sciences, No. 555 Zu Chong Zhi Road, Shanghai 201203, China.
| | - Zhengyu Cao
- Jiangsu Key Laboratory of TCM Evaluation and Translational Research, School of Traditional Chinese Pharmacy, China Pharmaceutical University, No. 639 Long Mian Road, Nanjing, Jiangsu 211198, China.
| |
Collapse
|
18
|
Bon RS, Wright DJ, Beech DJ, Sukumar P. Pharmacology of TRPC Channels and Its Potential in Cardiovascular and Metabolic Medicine. Annu Rev Pharmacol Toxicol 2022; 62:427-446. [PMID: 34499525 DOI: 10.1146/annurev-pharmtox-030121-122314] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Transient receptor potential canonical (TRPC) proteins assemble to form homo- or heterotetrameric, nonselective cation channels permeable to K+, Na+, and Ca2+. TRPC channels are thought to act as complex integrators of physical and chemical environmental stimuli. Although the understanding of essential physiological roles of TRPC channels is incomplete, their implication in various pathological mechanisms and conditions of the nervous system, kidneys, and cardiovascular system in combination with the lack of major adverse effects of TRPC knockout or TRPC channel inhibition is driving the search of TRPC channel modulators as potential therapeutics. Here, we review the most promising small-molecule TRPC channel modulators, the understanding of their mode of action, and their potential in the study and treatment of cardiovascular and metabolic disease.
Collapse
Affiliation(s)
- Robin S Bon
- Discovery and Translational Science Department, Leeds Institute of Cardiovascular and Metabolic Medicine, University of Leeds, Leeds LS2 9JT, United Kingdom;
| | - David J Wright
- Discovery and Translational Science Department, Leeds Institute of Cardiovascular and Metabolic Medicine, University of Leeds, Leeds LS2 9JT, United Kingdom;
| | - David J Beech
- Discovery and Translational Science Department, Leeds Institute of Cardiovascular and Metabolic Medicine, University of Leeds, Leeds LS2 9JT, United Kingdom;
| | - Piruthivi Sukumar
- Discovery and Translational Science Department, Leeds Institute of Cardiovascular and Metabolic Medicine, University of Leeds, Leeds LS2 9JT, United Kingdom;
| |
Collapse
|
19
|
Downregulation of TRPC4 and TRPC5 Inhibits Smooth Muscle Cell Proliferation without Affecting Endothelial Cell Proliferation. Genet Res (Camb) 2021; 2021:2949986. [PMID: 34899056 PMCID: PMC8643255 DOI: 10.1155/2021/2949986] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2021] [Accepted: 11/09/2021] [Indexed: 12/04/2022] Open
Abstract
Aims The main treatment for coronary heart disease is percutaneous coronary intervention (PCI), and drug-eluting stents are designed to inhibit vascular smooth muscle cell (VSMCs) proliferation and migration causing restenosis by releasing pharmacological agents into the vessel wall. Once drug-eluting stents are deployed, these pharmacological agents exert many biological effects in the coronary circulation, not only inhibition of VSMCs but also extension to vascular endothelial cells (VECs). The purpose of this study was to explore target molecules that inhibit VSMCs proliferation without affecting VECs. Methods mRNA and protein expressions of transient receptor potential channels (TRPCs) in cultured VSMCs and VECs were determined by western blotting and RT-qPCR. VSMCs and VECs proliferation was evaluated using CCK-8 assays and western blotting of proliferating cell nuclear antigen (PCNA). Calcium backfilling assays were performed to detect intracellular calcium ion concentration in cultured VSMCs and VECs. Results The TRPC6 expression was more abundant in VECs than VSMCs, while TRPC4 and TRPC5 expressions were more abundant in VSMCs than VECs. Knockdown of TRPC4 or TRPC5 alone had no remarkable inhibitory effect on VSMC proliferation. Synergistic knockdown of TRPC4 and TRPC5 inhibited the proliferation of VSMCs, declined the expression of the PCNA, and reduced the intracellular calcium ion concentration but not VECs. Conclusion These data suggest that concurrent inhibition of TRPC4 and TRPC5 inhibits VSMCs proliferation without affecting VECs, thus providing novel targets for developing pharmacological agents for drug-eluting stents.
Collapse
|
20
|
Abstract
Beta cells of the pancreatic islet express many different types of ion channels. These channels reside in the β-cell plasma membrane as well as subcellular organelles and their coordinated activity and sensitivity to metabolism regulate glucose-dependent insulin secretion. Here, we review the molecular nature, expression patterns, and functional roles of many β-cell channels, with an eye toward explaining the ionic basis of glucose-induced insulin secretion. Our primary focus is on KATP and voltage-gated Ca2+ channels as these primarily regulate insulin secretion; other channels in our view primarily help to sculpt the electrical patterns generated by activated β-cells or indirectly regulate metabolism. Lastly, we discuss why understanding the physiological roles played by ion channels is important for understanding the secretory defects that occur in type 2 diabetes. © 2021 American Physiological Society. Compr Physiol 11:1-21, 2021.
Collapse
Affiliation(s)
- Benjamin Thompson
- Department of Pharmacology, University of Michigan Medical School, Ann Arbor, Michigan, USA
- Brehm Diabetes Research Center, University of Michigan Medical School, Ann Arbor, Michigan, USA
| | | |
Collapse
|
21
|
de Sousa Valente J, Alawi KM, Bharde S, Zarban AA, Kodji X, Thapa D, Argunhan F, Barrett B, Nagy I, Brain SD. (-)-Englerin-A Has Analgesic and Anti-Inflammatory Effects Independent of TRPC4 and 5. Int J Mol Sci 2021; 22:6380. [PMID: 34203675 PMCID: PMC8232259 DOI: 10.3390/ijms22126380] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2021] [Revised: 06/10/2021] [Accepted: 06/11/2021] [Indexed: 12/13/2022] Open
Abstract
Recently, we found that the deletion of TRPC5 leads to increased inflammation and pain-related behaviour in two animal models of arthritis. (-)-Englerin A (EA), an extract from the East African plant Phyllanthus engleri has been identified as a TRPC4/5 agonist. Here, we studied whether or not EA has any anti-inflammatory and analgesic properties via TRPC4/5 in the carrageenan model of inflammation. We found that EA treatment in CD1 mice inhibited thermal hyperalgesia and mechanical allodynia in a dose-dependent manner. Furthermore, EA significantly reduced the volume of carrageenan-induced paw oedema and the mass of the treated paws. Additionally, in dorsal root ganglion (DRG) neurons cultured from WT 129S1/SvIm mice, EA induced a dose-dependent cobalt uptake that was surprisingly preserved in cultured DRG neurons from 129S1/SvIm TRPC5 KO mice. Likewise, EA-induced anti-inflammatory and analgesic effects were preserved in the carrageenan model in animals lacking TRPC5 expression or in mice treated with TRPC4/5 antagonist ML204.This study demonstrates that while EA activates a sub-population of DRG neurons, it induces a novel TRPC4/5-independent analgesic and anti-inflammatory effect in vivo. Future studies are needed to elucidate the molecular and cellular mechanisms underlying EA's anti-inflammatory and analgesic effects.
Collapse
Affiliation(s)
- João de Sousa Valente
- Section of Vascular Biology and Inflammation, BHF Cardiovascular Centre of Research Excellence, School of Cardiovascular Medicine and Sciences, King’s College London, Franklin-Wilkins Building, London SE1 9NH, UK; (K.M.A.); (S.B.); (A.A.Z.); (X.K.); (D.T.); (F.A.); (B.B.); (S.D.B.)
| | - Khadija M Alawi
- Section of Vascular Biology and Inflammation, BHF Cardiovascular Centre of Research Excellence, School of Cardiovascular Medicine and Sciences, King’s College London, Franklin-Wilkins Building, London SE1 9NH, UK; (K.M.A.); (S.B.); (A.A.Z.); (X.K.); (D.T.); (F.A.); (B.B.); (S.D.B.)
| | - Sabah Bharde
- Section of Vascular Biology and Inflammation, BHF Cardiovascular Centre of Research Excellence, School of Cardiovascular Medicine and Sciences, King’s College London, Franklin-Wilkins Building, London SE1 9NH, UK; (K.M.A.); (S.B.); (A.A.Z.); (X.K.); (D.T.); (F.A.); (B.B.); (S.D.B.)
| | - Ali A. Zarban
- Section of Vascular Biology and Inflammation, BHF Cardiovascular Centre of Research Excellence, School of Cardiovascular Medicine and Sciences, King’s College London, Franklin-Wilkins Building, London SE1 9NH, UK; (K.M.A.); (S.B.); (A.A.Z.); (X.K.); (D.T.); (F.A.); (B.B.); (S.D.B.)
- Department of Pharmacological Sciences, Faculty of Pharmacy, Jazan University, Jazan 45142, Saudi Arabia
| | - Xenia Kodji
- Section of Vascular Biology and Inflammation, BHF Cardiovascular Centre of Research Excellence, School of Cardiovascular Medicine and Sciences, King’s College London, Franklin-Wilkins Building, London SE1 9NH, UK; (K.M.A.); (S.B.); (A.A.Z.); (X.K.); (D.T.); (F.A.); (B.B.); (S.D.B.)
| | - Dibesh Thapa
- Section of Vascular Biology and Inflammation, BHF Cardiovascular Centre of Research Excellence, School of Cardiovascular Medicine and Sciences, King’s College London, Franklin-Wilkins Building, London SE1 9NH, UK; (K.M.A.); (S.B.); (A.A.Z.); (X.K.); (D.T.); (F.A.); (B.B.); (S.D.B.)
| | - Fulye Argunhan
- Section of Vascular Biology and Inflammation, BHF Cardiovascular Centre of Research Excellence, School of Cardiovascular Medicine and Sciences, King’s College London, Franklin-Wilkins Building, London SE1 9NH, UK; (K.M.A.); (S.B.); (A.A.Z.); (X.K.); (D.T.); (F.A.); (B.B.); (S.D.B.)
| | - Brentton Barrett
- Section of Vascular Biology and Inflammation, BHF Cardiovascular Centre of Research Excellence, School of Cardiovascular Medicine and Sciences, King’s College London, Franklin-Wilkins Building, London SE1 9NH, UK; (K.M.A.); (S.B.); (A.A.Z.); (X.K.); (D.T.); (F.A.); (B.B.); (S.D.B.)
| | - Istvan Nagy
- Nociception Group, Section of Anaesthetic, Pain Medicine and Intensive Care, Department of Surgery and Cancer, Imperial College London, London SW7 2AZ, UK;
| | - Susan D. Brain
- Section of Vascular Biology and Inflammation, BHF Cardiovascular Centre of Research Excellence, School of Cardiovascular Medicine and Sciences, King’s College London, Franklin-Wilkins Building, London SE1 9NH, UK; (K.M.A.); (S.B.); (A.A.Z.); (X.K.); (D.T.); (F.A.); (B.B.); (S.D.B.)
| |
Collapse
|
22
|
Ningoo M, Plant LD, Greka A, Logothetis DE. PIP 2 regulation of TRPC5 channel activation and desensitization. J Biol Chem 2021; 296:100726. [PMID: 33933453 PMCID: PMC8191310 DOI: 10.1016/j.jbc.2021.100726] [Citation(s) in RCA: 32] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2021] [Revised: 04/20/2021] [Accepted: 04/28/2021] [Indexed: 10/27/2022] Open
Abstract
Transient receptor potential canonical type 5 (TRPC5) ion channels are expressed in the brain and kidney and have been identified as promising therapeutic targets whose selective inhibition can protect against diseases driven by a leaky kidney filter, such as focal segmental glomerular sclerosis. TRPC5 channels are activated not only by elevated levels of extracellular Ca2+or lanthanide ions but also by G protein (Gq/11) stimulation. Phosphatidylinositol 4,5-bisphosphate (PIP2) hydrolysis by phospholipase C enzymes leads to PKC-mediated phosphorylation of TRPC5 channels and their subsequent desensitization. However, the roles of PIP2 in activation and maintenance of TRPC5 channel activity via its hydrolysis product diacyl glycerol (DAG), as well as the mechanism of desensitization of TRPC5 activity by DAG-stimulated PKC activity, remain unclear. Here, we designed experiments to distinguish between the processes underlying channel activation and inhibition. Employing whole-cell patch-clamp, we used an optogenetic tool to dephosphorylate PIP2 and assess channel-PIP2 interactions influenced by activators, such as DAG, or inhibitors, such as PKC phosphorylation. Using total internal reflection microscopy, we assessed channel cell surface density. We show that PIP2 controls both the PKC-mediated inhibition and the DAG- and lanthanide-mediated activation of TRPC5 currents via control of gating rather than channel cell surface density. These mechanistic insights promise to aid in the development of more selective and precise inhibitors to block TRPC5 channel activity and illuminate new opportunities for targeted therapies for a group of chronic kidney diseases for which there is currently a great unmet need.
Collapse
Affiliation(s)
- Mehek Ningoo
- Department of Pharmaceutical Sciences, School of Pharmacy, Bouvé College of Health Sciences, Northeastern University, Boston, Massachusetts, USA
| | - Leigh D Plant
- Department of Pharmaceutical Sciences, School of Pharmacy, Bouvé College of Health Sciences, Northeastern University, Boston, Massachusetts, USA; Center for Drug Discovery, Northeastern University, Boston, Massachusetts, USA
| | - Anna Greka
- Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, Massachusetts, USA; Broad Institute of MIT and Harvard, Cambridge, Massachusetts, USA
| | - Diomedes E Logothetis
- Department of Pharmaceutical Sciences, School of Pharmacy, Bouvé College of Health Sciences, Northeastern University, Boston, Massachusetts, USA; Center for Drug Discovery, Northeastern University, Boston, Massachusetts, USA; Department of Chemistry and Chemical Biology, College of Science, Northeastern University, Boston, Massachusetts, USA.
| |
Collapse
|
23
|
Sabnis RW. Novel Pyridazinones as TRPC5 Inhibitors for Treating Kidney Diseases. ACS Med Chem Lett 2021; 12:526-527. [PMID: 33859787 PMCID: PMC8040034 DOI: 10.1021/acsmedchemlett.1c00123] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2021] [Indexed: 11/30/2022] Open
Affiliation(s)
- Ram W. Sabnis
- Smith, Gambrell
& Russell LLP, 1230 Peachtree Street NE, Suite 3100, Atlanta, Georgia 30309, United States
| |
Collapse
|
24
|
Song K, Wei M, Guo W, Quan L, Kang Y, Wu JX, Chen L. Structural basis for human TRPC5 channel inhibition by two distinct inhibitors. eLife 2021; 10:63429. [PMID: 33683200 PMCID: PMC7987348 DOI: 10.7554/elife.63429] [Citation(s) in RCA: 49] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2020] [Accepted: 03/05/2021] [Indexed: 02/06/2023] Open
Abstract
TRPC5 channel is a nonselective cation channel that participates in diverse physiological processes. TRPC5 inhibitors show promise in the treatment of anxiety disorder, depression, and kidney disease. However, the binding sites and inhibitory mechanism of TRPC5 inhibitors remain elusive. Here, we present the cryo-EM structures of human TRPC5 in complex with two distinct inhibitors, namely clemizole and HC-070, to the resolution of 2.7 Å. The structures reveal that clemizole binds inside the voltage sensor-like domain of each subunit. In contrast, HC-070 is wedged between adjacent subunits and replaces the glycerol group of a putative diacylglycerol molecule near the extracellular side. Moreover, we found mutations in the inhibitor binding pockets altered the potency of inhibitors. These structures suggest that both clemizole and HC-070 exert the inhibitory functions by stabilizing the ion channel in a nonconductive closed state. These results pave the way for further design and optimization of inhibitors targeting human TRPC5.
Collapse
Affiliation(s)
- Kangcheng Song
- State Key Laboratory of Membrane Biology, College of Future Technology, Institute of Molecular Medicine, Beijing Key Laboratory of Cardiometabolic Molecular Medicine, Peking University, Beijing, China
| | - Miao Wei
- State Key Laboratory of Membrane Biology, College of Future Technology, Institute of Molecular Medicine, Beijing Key Laboratory of Cardiometabolic Molecular Medicine, Peking University, Beijing, China
| | - Wenjun Guo
- State Key Laboratory of Membrane Biology, College of Future Technology, Institute of Molecular Medicine, Beijing Key Laboratory of Cardiometabolic Molecular Medicine, Peking University, Beijing, China
| | - Li Quan
- State Key Laboratory of Membrane Biology, College of Future Technology, Institute of Molecular Medicine, Beijing Key Laboratory of Cardiometabolic Molecular Medicine, Peking University, Beijing, China
| | - Yunlu Kang
- State Key Laboratory of Membrane Biology, College of Future Technology, Institute of Molecular Medicine, Beijing Key Laboratory of Cardiometabolic Molecular Medicine, Peking University, Beijing, China
| | - Jing-Xiang Wu
- State Key Laboratory of Membrane Biology, College of Future Technology, Institute of Molecular Medicine, Beijing Key Laboratory of Cardiometabolic Molecular Medicine, Peking University, Beijing, China.,Peking-Tsinghua Center for Life Sciences, Peking University, Beijing, China.,Academy for Advanced Interdisciplinary Studies, Peking University, Beijing, China
| | - Lei Chen
- State Key Laboratory of Membrane Biology, College of Future Technology, Institute of Molecular Medicine, Beijing Key Laboratory of Cardiometabolic Molecular Medicine, Peking University, Beijing, China.,Peking-Tsinghua Center for Life Sciences, Peking University, Beijing, China.,Academy for Advanced Interdisciplinary Studies, Peking University, Beijing, China
| |
Collapse
|
25
|
Wright DJ, Simmons KJ, Johnson RM, Beech DJ, Muench SP, Bon RS. Human TRPC5 structures reveal interaction of a xanthine-based TRPC1/4/5 inhibitor with a conserved lipid binding site. Commun Biol 2020; 3:704. [PMID: 33230284 PMCID: PMC7683545 DOI: 10.1038/s42003-020-01437-8] [Citation(s) in RCA: 40] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2020] [Accepted: 10/23/2020] [Indexed: 02/08/2023] Open
Abstract
TRPC1/4/5 channels are non-specific cation channels implicated in a wide variety of diseases, and TRPC1/4/5 inhibitors have recently entered clinical trials. However, fundamental and translational studies require a better understanding of TRPC1/4/5 channel regulation by endogenous and exogenous factors. Although several potent and selective TRPC1/4/5 modulators have been reported, the paucity of mechanistic insights into their modes-of-action remains a barrier to the development of new chemical probes and drug candidates. Xanthine-based modulators include the most potent and selective TRPC1/4/5 inhibitors described to date, as well as TRPC5 activators. Our previous studies suggest that xanthines interact with a, so far, elusive pocket of TRPC1/4/5 channels that is essential to channel gating. Here we report the structure of a small-molecule-bound TRPC1/4/5 channel-human TRPC5 in complex with the xanthine Pico145-to 3.0 Å. We found that Pico145 binds to a conserved lipid binding site of TRPC5, where it displaces a bound phospholipid. Our findings explain the mode-of-action of xanthine-based TRPC1/4/5 modulators, and suggest a structural basis for TRPC1/4/5 modulation by endogenous factors such as (phospho)lipids and Zn2+ ions. These studies lay the foundations for the structure-based design of new generations of TRPC1/4/5 modulators.
Collapse
Affiliation(s)
- David J Wright
- Discovery and Translational Science Department, Leeds Institute of Cardiovascular and Metabolic Medicine, University of Leeds, Leeds, LS2 9JT, UK
- Astbury Centre for Structural Molecular Biology, University of Leeds, Woodhouse Lane, Leeds, LS2 9JT, UK
| | - Katie J Simmons
- Discovery and Translational Science Department, Leeds Institute of Cardiovascular and Metabolic Medicine, University of Leeds, Leeds, LS2 9JT, UK
- Astbury Centre for Structural Molecular Biology, University of Leeds, Woodhouse Lane, Leeds, LS2 9JT, UK
| | - Rachel M Johnson
- Astbury Centre for Structural Molecular Biology, University of Leeds, Woodhouse Lane, Leeds, LS2 9JT, UK
- School of Biomedical Sciences, University of Leeds, Woodhouse Lane, Leeds, LS2 9JT, UK
| | - David J Beech
- Discovery and Translational Science Department, Leeds Institute of Cardiovascular and Metabolic Medicine, University of Leeds, Leeds, LS2 9JT, UK
| | - Stephen P Muench
- Astbury Centre for Structural Molecular Biology, University of Leeds, Woodhouse Lane, Leeds, LS2 9JT, UK.
- School of Biomedical Sciences, University of Leeds, Woodhouse Lane, Leeds, LS2 9JT, UK.
| | - Robin S Bon
- Discovery and Translational Science Department, Leeds Institute of Cardiovascular and Metabolic Medicine, University of Leeds, Leeds, LS2 9JT, UK.
- Astbury Centre for Structural Molecular Biology, University of Leeds, Woodhouse Lane, Leeds, LS2 9JT, UK.
| |
Collapse
|
26
|
Liu B, Zhang W, Guo S, Zuo Z. Discovery of novel modulators targeting human TRPC5: Docking-based virtual screening, molecular dynamics simulation and binding affinity predication. J Mol Graph Model 2020; 102:107795. [PMID: 33161371 DOI: 10.1016/j.jmgm.2020.107795] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2020] [Revised: 10/28/2020] [Accepted: 10/28/2020] [Indexed: 11/20/2022]
Abstract
Canonical transient receptor potential channel 5 (TRPC5) plays a key role in the regulation of central nervous system, cardiovascular system, kidney disease, cancer, and could be also involved in liver function, arthritis, diabetes-associated complications and so on. However, evidence of TRPC5 function on cellular or organismic levels is sparse. There is still a need for identifying novel and efficient TRPC5 channel modulators to study TRPC5 function. In this study, based on the hTRPC5 structure obtained by homology modeling and the predicted binding site, we have performed virtual screening of 212,736 compounds from the specs database(http://www.specs.net) to find potential hTRPC5 modulators. Lipinski and Veber rules, ADMET (Absorption, Distribution, Metabolism, Excretion, Toxicity) and PAINS (Pan Assay Interference structures) filters were used to screen the large database. Further, multi-software combination docking, cluster analysis and interaction analysis were used to select 20 potential active candidates with novel skeleton. 4 Hits, bearing appreciable binding affinity with hTRPC5 were selected for 40ns all-atom molecular dynamics (MD) simulations under explicit water conditions. The MD simulation results suggested that the 4 Hits binding induces a slight structural change and stabilizes the hTRPC5 structure. In addition, decomposition free energy demonstrated that residues TRP434, LEU437, MET438, ALA441, ILE484, ILE487, LEU488, LEU491, LEU515, ILE517, LEU518, LEU521, PHE531, THR607, VAL610, ILE611, VAL615 played the critical role on system stability. 4 Hits, as potential modulators of hTRPC5, may be potential leads to develop effective therapeutics hTRPC5-associated diseases.
Collapse
Affiliation(s)
- Bin Liu
- State Key Laboratory of Phytochemistry and Plant Resources in West China, Kunming Institute of Botany, Chinese Academy of Sciences, Kunming 650201, PR China; University of Chinese Academy of Sciences, Beijing 100049, PR China
| | - Wei Zhang
- State Key Laboratory of Phytochemistry and Plant Resources in West China, Kunming Institute of Botany, Chinese Academy of Sciences, Kunming 650201, PR China
| | - Sheng Guo
- State Key Laboratory of Phytochemistry and Plant Resources in West China, Kunming Institute of Botany, Chinese Academy of Sciences, Kunming 650201, PR China
| | - Zhili Zuo
- State Key Laboratory of Phytochemistry and Plant Resources in West China, Kunming Institute of Botany, Chinese Academy of Sciences, Kunming 650201, PR China; University of Chinese Academy of Sciences, Beijing 100049, PR China.
| |
Collapse
|
27
|
Yu Y, Liang Q, Du L, Jiang H, Gu J, Hu H, Tu Z. Synthesis and Characterization of a Specific Iodine-125-Labeled TRPC5 Radioligand. ChemMedChem 2020; 15:1854-1860. [PMID: 32717096 PMCID: PMC8544919 DOI: 10.1002/cmdc.202000339] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2020] [Indexed: 11/11/2022]
Abstract
The nonselective Ca2+ -permeable transient receptor potential channel subfamily member 5 (TRPC5) belongs to the transient receptor potential canonical (TRPC) superfamily and is widely expressed in the brain. Compelling evidence reveals that TRPC5 plays crucial roles in depression and other psychiatric disorders. To develop a TRPC5 radioligand, following up on our previous effort, we synthesized the iodine compound TZ66127 and its iodine-125-labeled counterpart [125 I]TZ66127. The synthesis of TZ66127 was achieved by replacing chloride with iodide in the structure of HC608, and the [125 I]TZ66127 was radiosynthesized using its corresponding tributylstannylated precursor. We established a stable human TRPC5-overexpressed HEK293-hTRPC5 cell line and performed Ca2+ imaging and a cell-binding assay study of TZ66127; these indicated that TZ66127 had good inhibition activity for TRPC5, and the inhibitory efficiency of TZ66127 toward TRPC5 presented in a dose-dependent manner. An in vitro autoradiography and immunohistochemistry study of rat brain sections suggested that [125 I]TZ66127 had binding specificity toward TRPC5. Altogether, [125 I]TZ66127 has high potential to serve as a radioligand for screening the binding activity of other new compounds toward TRPC5. The availability of [125 I]TZ66127 might facilitate the development of therapeutic drugs and PET imaging agents that target TRPC5.
Collapse
Affiliation(s)
- Yanbo Yu
- Department of Radiology, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Qianwa Liang
- Department of Radiology, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Lixia Du
- Department of Anesthesiology, Center for the Study of Itch, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Hao Jiang
- Department of Radiology, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Jiwei Gu
- Department of Radiology, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Hongzhen Hu
- Department of Anesthesiology, Center for the Study of Itch, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Zhude Tu
- Department of Radiology, Washington University School of Medicine, St. Louis, MO 63110, USA
| |
Collapse
|
28
|
Epigallocatechin-3-gallate mobilizes intracellular Ca 2+ in prostate cancer cells through combined Ca 2+ entry and Ca 2+-induced Ca 2+ release. Life Sci 2020; 258:118232. [PMID: 32781066 DOI: 10.1016/j.lfs.2020.118232] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2020] [Revised: 07/31/2020] [Accepted: 08/05/2020] [Indexed: 12/28/2022]
Abstract
AIMS To elucidate the mechanism by which (-)-epigallocatechin-3-gallate (EGCG) mediates intracellular Ca2+ increase in androgen-independent prostate cancer (PCa) cells. MAIN METHODS Following exposure to different doses of EGCG, viability of DU145 and PC3 PCa cells was evaluated by MTT assay and the intracellular Ca2+ dynamics by the fluorescent Ca2+ chelator Fura-2. The expression of different channels was investigated by qPCR analysis and sulfhydryl bonds by Ellman's assay. KEY FINDINGS EGCG inhibited DU145 and PC3 proliferation with IC50 = 46 and 56 μM, respectively, and induced dose-dependent peaks of internal Ca2+ that were dependent on extracellular Ca2+. The expression of TRPC4 and TRPC6 channels was revealed by qPCR in PC3 cells, but lack of effect by modulators and blockers ruled out an exclusive role for these, as well as for voltage-dependent T-type Ca2+ channels. Application of dithiothreitol and catalase and sulfhydryl (SH) measurements showed that EGCG-induced Ca2+ rise depends on SH oxidation, while the effect of EGTA, dantrolene, and the PLC inhibitor U73122 suggested that EGCG-induced Ca2+ influx acts as a trigger for Ca2+-induced Ca2+ release, involving both ryanodine and IP3 receptors. Different from EGCG, ATP caused a rapid Ca2+ increase, which was independent of external Ca2+, but sensitive to U73122. SIGNIFICANCE EGCG induces an internal Ca2+ increase in PCa cells by a multi-step mechanism. As dysregulation of cytosolic Ca2+ is directly linked to apoptosis in PCa cells, these data confirm the possibility of using EGCG as a synergistic adjuvant in combined therapies for recalcitrant malignancies like androgen-independent PCa.
Collapse
|
29
|
Aroke EN, Powell-Roach KL, Jaime-Lara RB, Tesfaye M, Roy A, Jackson P, Joseph PV. Taste the Pain: The Role of TRP Channels in Pain and Taste Perception. Int J Mol Sci 2020; 21:E5929. [PMID: 32824721 PMCID: PMC7460556 DOI: 10.3390/ijms21165929] [Citation(s) in RCA: 31] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2020] [Revised: 08/12/2020] [Accepted: 08/16/2020] [Indexed: 12/11/2022] Open
Abstract
Transient receptor potential (TRP) channels are a superfamily of cation transmembrane proteins that are expressed in many tissues and respond to many sensory stimuli. TRP channels play a role in sensory signaling for taste, thermosensation, mechanosensation, and nociception. Activation of TRP channels (e.g., TRPM5) in taste receptors by food/chemicals (e.g., capsaicin) is essential in the acquisition of nutrients, which fuel metabolism, growth, and development. Pain signals from these nociceptors are essential for harm avoidance. Dysfunctional TRP channels have been associated with neuropathic pain, inflammation, and reduced ability to detect taste stimuli. Humans have long recognized the relationship between taste and pain. However, the mechanisms and relationship among these taste-pain sensorial experiences are not fully understood. This article provides a narrative review of literature examining the role of TRP channels on taste and pain perception. Genomic variability in the TRPV1 gene has been associated with alterations in various pain conditions. Moreover, polymorphisms of the TRPV1 gene have been associated with alterations in salty taste sensitivity and salt preference. Studies of genetic variations in TRP genes or modulation of TRP pathways may increase our understanding of the shared biological mediators of pain and taste, leading to therapeutic interventions to treat many diseases.
Collapse
Affiliation(s)
- Edwin N. Aroke
- School of Nursing, University of Alabama at Birmingham, Birmingham, AL 35294, USA; (E.N.A.); (P.J.)
| | | | - Rosario B. Jaime-Lara
- Sensory Science and Metabolism Unit (SenSMet), National Institute of Nursing Research, National Institutes of Health, Bethesda, MD 20892, USA; (R.B.J.-L.); (M.T.); (A.R.)
| | - Markos Tesfaye
- Sensory Science and Metabolism Unit (SenSMet), National Institute of Nursing Research, National Institutes of Health, Bethesda, MD 20892, USA; (R.B.J.-L.); (M.T.); (A.R.)
| | - Abhrabrup Roy
- Sensory Science and Metabolism Unit (SenSMet), National Institute of Nursing Research, National Institutes of Health, Bethesda, MD 20892, USA; (R.B.J.-L.); (M.T.); (A.R.)
| | - Pamela Jackson
- School of Nursing, University of Alabama at Birmingham, Birmingham, AL 35294, USA; (E.N.A.); (P.J.)
| | - Paule V. Joseph
- Sensory Science and Metabolism Unit (SenSMet), National Institute of Nursing Research, National Institutes of Health, Bethesda, MD 20892, USA; (R.B.J.-L.); (M.T.); (A.R.)
| |
Collapse
|
30
|
Synthesis of the quinazolinone derivatives using an acid-functionalized magnetic silica heterogeneous catalyst in terms of green chemistry. Mol Divers 2020; 25:889-897. [PMID: 32078143 DOI: 10.1007/s11030-020-10033-1] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2019] [Accepted: 01/04/2020] [Indexed: 01/08/2023]
Abstract
In this research, the synthesis of the quinazolinone derivatives by the reaction of diaminoglyoxime with anthranilic acid or methyl 2-amino benzoate over an acetic acid-functionalized magnetic silica-based catalyst in water was described. The acetic acid-functionalized catalyst was prepared using a three-step procedure from magnetite NPs that initially coated with a layer of silica through the sol-gel process, modified with an aminosilane layer and functionalized with bromoacetic acid. The catalyst was characterized by means of spectroscopic and microscopic techniques, and its activity was investigated for the synthesis of the quinazolinones, bisquinazolinone and oxadiazole quinazolinones obtained from diaminoglyoxime in water at room temperature.
Collapse
|
31
|
Bauer CC, Minard A, Pickles IB, Simmons KJ, Chuntharpursat-Bon E, Burnham MP, Kapur N, Beech DJ, Muench SP, Wright MH, Warriner SL, Bon RS. Xanthine-based photoaffinity probes allow assessment of ligand engagement by TRPC5 channels. RSC Chem Biol 2020. [DOI: 10.1039/d0cb00126k] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
Diazirine-containing photoaffinity probes, based on the potent and selective TRPC1/4/5 channel inhibitor Pico145, allowed the development of an assay to probe cellular interactions between TRPC5 protein and xanthine-based TRPC5 channel modulators.
Collapse
Affiliation(s)
- Claudia C. Bauer
- Leeds Institute of Cardiovascular and Metabolic Medicine
- LIGHT Laboratories
- University of Leeds
- Leeds LS2 9JT
- UK
| | - Aisling Minard
- Leeds Institute of Cardiovascular and Metabolic Medicine
- LIGHT Laboratories
- University of Leeds
- Leeds LS2 9JT
- UK
| | - Isabelle B. Pickles
- Leeds Institute of Cardiovascular and Metabolic Medicine
- LIGHT Laboratories
- University of Leeds
- Leeds LS2 9JT
- UK
| | - Katie J. Simmons
- Leeds Institute of Cardiovascular and Metabolic Medicine
- LIGHT Laboratories
- University of Leeds
- Leeds LS2 9JT
- UK
| | | | | | - Nikil Kapur
- School of Mechanical Engineering
- University of Leeds
- Leeds LS2 9JT
- UK
| | - David J. Beech
- Leeds Institute of Cardiovascular and Metabolic Medicine
- LIGHT Laboratories
- University of Leeds
- Leeds LS2 9JT
- UK
| | - Stephen P. Muench
- School of Biomedical Sciences
- University of Leeds
- Leeds LS2 9JT
- UK
- Astbury Centre for Structural Molecular Biology
| | - Megan H. Wright
- School of Chemistry
- University of Leeds
- Woodhouse Lane
- Leeds LS2 9JT
- UK
| | | | - Robin S. Bon
- Leeds Institute of Cardiovascular and Metabolic Medicine
- LIGHT Laboratories
- University of Leeds
- Leeds LS2 9JT
- UK
| |
Collapse
|
32
|
Pablo JL, Greka A. Charting a TRP to Novel Therapeutic Destinations for Kidney Diseases. Trends Pharmacol Sci 2019; 40:911-918. [PMID: 31704171 DOI: 10.1016/j.tips.2019.10.001] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2019] [Revised: 09/24/2019] [Accepted: 10/07/2019] [Indexed: 12/29/2022]
Abstract
Ion channels are critical to kidney function, and their dysregulation leads to several distinct kidney diseases. Of the diversity of ion channels in kidney cells, the transient receptor potential (TRP) superfamily of proteins plays important and varied roles in both maintaining homeostasis as well as in causing disease. Recent work showed that TRPC5 blockers could successfully protect critical components of the kidney filter both in vitro and in vivo, thus revealing TRPC5 as a tractable therapeutic target for focal and segmental glomerulosclerosis (FSGS), a common cause of kidney failure. Human genetics point to three additional TRP channels as plausible therapeutic targets: TRPC6 in FSGS, PKD2 in polycystic kidney disease, and TRPM6 in familial hypomagnesemia with secondary hypocalcemia (HSH). We conclude that targeting TRP channels could pave the way for much needed therapies for kidney diseases.
Collapse
Affiliation(s)
- Juan Lorenzo Pablo
- Broad Institute of MIT and Harvard, Massachusetts Institute of Technology, Cambridge, MA 02142, USA; Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA 02115, USA
| | - Anna Greka
- Broad Institute of MIT and Harvard, Massachusetts Institute of Technology, Cambridge, MA 02142, USA; Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA 02115, USA.
| |
Collapse
|