1
|
Fan YG, Wu TY, Zhao LX, Jia RJ, Ren H, Hou WJ, Wang ZY. From zinc homeostasis to disease progression: Unveiling the neurodegenerative puzzle. Pharmacol Res 2024; 199:107039. [PMID: 38123108 DOI: 10.1016/j.phrs.2023.107039] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/07/2023] [Revised: 11/16/2023] [Accepted: 12/10/2023] [Indexed: 12/23/2023]
Abstract
Zinc is a crucial trace element in the human body, playing a role in various physiological processes such as oxidative stress, neurotransmission, protein synthesis, and DNA repair. The zinc transporters (ZnTs) family members are responsible for exporting intracellular zinc, while Zrt- and Irt-like proteins (ZIPs) are involved in importing extracellular zinc. These processes are essential for maintaining cellular zinc homeostasis. Imbalances in zinc metabolism have been linked to the development of neurodegenerative diseases. Disruptions in zinc levels can impact the survival and activity of neurons, thereby contributing to the progression of neurodegenerative diseases through mechanisms like cell apoptosis regulation, protein phase separation, ferroptosis, oxidative stress, and neuroinflammation. Therefore, conducting a systematic review of the regulatory network of zinc and investigating the relationship between zinc dysmetabolism and neurodegenerative diseases can enhance our understanding of the pathogenesis of these diseases. Additionally, it may offer new insights and approaches for the treatment of neurodegenerative diseases.
Collapse
Affiliation(s)
- Yong-Gang Fan
- Key Laboratory of Medical Cell Biology of Ministry of Education, Key Laboratory of Major Chronic Diseases of Nervous System of Liaoning Province, Health Sciences Institute of China Medical University, Shenyang 110122, China.
| | - Ting-Yao Wu
- First Affiliated Hospital of Jinzhou Medical University, Jinzhou 121000, China
| | - Ling-Xiao Zhao
- Key Laboratory of Medical Cell Biology of Ministry of Education, Key Laboratory of Major Chronic Diseases of Nervous System of Liaoning Province, Health Sciences Institute of China Medical University, Shenyang 110122, China
| | - Rong-Jun Jia
- Key Laboratory of Medical Cell Biology of Ministry of Education, Key Laboratory of Major Chronic Diseases of Nervous System of Liaoning Province, Health Sciences Institute of China Medical University, Shenyang 110122, China
| | - Hang Ren
- Key Laboratory of Medical Cell Biology of Ministry of Education, Key Laboratory of Major Chronic Diseases of Nervous System of Liaoning Province, Health Sciences Institute of China Medical University, Shenyang 110122, China
| | - Wen-Jia Hou
- Key Laboratory of Medical Cell Biology of Ministry of Education, Key Laboratory of Major Chronic Diseases of Nervous System of Liaoning Province, Health Sciences Institute of China Medical University, Shenyang 110122, China
| | - Zhan-You Wang
- Key Laboratory of Medical Cell Biology of Ministry of Education, Key Laboratory of Major Chronic Diseases of Nervous System of Liaoning Province, Health Sciences Institute of China Medical University, Shenyang 110122, China.
| |
Collapse
|
2
|
Okada Y, Numata T, Sabirov RZ, Kashio M, Merzlyak PG, Sato-Numata K. Cell death induction and protection by activation of ubiquitously expressed anion/cation channels. Part 3: the roles and properties of TRPM2 and TRPM7. Front Cell Dev Biol 2023; 11:1246955. [PMID: 37842082 PMCID: PMC10576435 DOI: 10.3389/fcell.2023.1246955] [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: 06/25/2023] [Accepted: 09/15/2023] [Indexed: 10/17/2023] Open
Abstract
Cell volume regulation (CVR) is a prerequisite for animal cells to survive and fulfill their functions. CVR dysfunction is essentially involved in the induction of cell death. In fact, sustained normotonic cell swelling and shrinkage are associated with necrosis and apoptosis, and thus called the necrotic volume increase (NVI) and the apoptotic volume decrease (AVD), respectively. Since a number of ubiquitously expressed ion channels are involved in the CVR processes, these volume-regulatory ion channels are also implicated in the NVI and AVD events. In Part 1 and Part 2 of this series of review articles, we described the roles of swelling-activated anion channels called VSOR or VRAC and acid-activated anion channels called ASOR or PAC in CVR and cell death processes. Here, Part 3 focuses on therein roles of Ca2+-permeable non-selective TRPM2 and TRPM7 cation channels activated by stress. First, we summarize their phenotypic properties and molecular structure. Second, we describe their roles in CVR. Since cell death induction is tightly coupled to dysfunction of CVR, third, we focus on their participation in the induction of or protection against cell death under oxidative, acidotoxic, excitotoxic, and ischemic conditions. In this regard, we pay attention to the sensitivity of TRPM2 and TRPM7 to a variety of stress as well as to their capability to physicall and functionally interact with other volume-related channels and membrane enzymes. Also, we summarize a large number of reports hitherto published in which TRPM2 and TRPM7 channels are shown to be involved in cell death associated with a variety of diseases or disorders, in some cases as double-edged swords. Lastly, we attempt to describe how TRPM2 and TRPM7 are organized in the ionic mechanisms leading to cell death induction and protection.
Collapse
Affiliation(s)
- Yasunobu Okada
- National Institute for Physiological Sciences (NIPS), Okazaki, Japan
- Department of Integrative Physiology, Graduate School of Medicine, AkitaUniversity, Akita, Japan
- Department of Physiology, School of Medicine, Aichi Medical Uniersity, Nagakute, Japan
- Department of Physiology, Kyoto Prefectural University of Medicine, Kyoto, Japan
- Cardiovascular Research Institute, Yokohama City University, Yokohama, Japan
| | - Tomohiro Numata
- Department of Integrative Physiology, Graduate School of Medicine, AkitaUniversity, Akita, Japan
| | - Ravshan Z. Sabirov
- Institute of Biophysics and Biochemistry, National University of Uzbekistan, Tashkent, Uzbekistan
| | - Makiko Kashio
- National Institute for Physiological Sciences (NIPS), Okazaki, Japan
- Department of Physiology, School of Medicine, Aichi Medical Uniersity, Nagakute, Japan
| | - Peter G. Merzlyak
- Institute of Biophysics and Biochemistry, National University of Uzbekistan, Tashkent, Uzbekistan
| | - Kaori Sato-Numata
- Department of Integrative Physiology, Graduate School of Medicine, AkitaUniversity, Akita, Japan
| |
Collapse
|
3
|
Ciaglia T, Vestuto V, Bertamino A, González-Muñiz R, Gómez-Monterrey I. On the modulation of TRPM channels: Current perspectives and anticancer therapeutic implications. Front Oncol 2023; 12:1065935. [PMID: 36844925 PMCID: PMC9948629 DOI: 10.3389/fonc.2022.1065935] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2022] [Accepted: 12/15/2022] [Indexed: 02/11/2023] Open
Abstract
The transient melastatin receptor potential (TRPM) ion channel subfamily functions as cellular sensors and transducers of critical biological signal pathways by regulating ion homeostasis. Some members of TRPM have been cloned from cancerous tissues, and their abnormal expressions in various solid malignancies have been correlated with cancer cell growth, survival, or death. Recent evidence also highlights the mechanisms underlying the role of TRPMs in tumor epithelial-mesenchymal transition (EMT), autophagy, and cancer metabolic reprogramming. These implications support TRPM channels as potential molecular targets and their modulation as an innovative therapeutic approach against cancer. Here, we discuss the general characteristics of the different TRPMs, focusing on current knowledge about the connection between TRPM channels and critical features of cancer. We also cover TRPM modulators used as pharmaceutical tools in biological trials and an indication of the only clinical trial with a TRPM modulator about cancer. To conclude, the authors describe the prospects for TRPM channels in oncology.
Collapse
Affiliation(s)
- Tania Ciaglia
- Dipartimento di Farmacia (DIFARMA), Università degli Studi di Salerno, Fisciano, Italy
| | - Vincenzo Vestuto
- Dipartimento di Farmacia (DIFARMA), Università degli Studi di Salerno, Fisciano, Italy
| | - Alessia Bertamino
- Dipartimento di Farmacia (DIFARMA), Università degli Studi di Salerno, Fisciano, Italy
| | | | | |
Collapse
|
4
|
Leong IL, Yu CM, Shiao LR, Chan P, Wu KC, Leung YM. Sensitivity of Ca 2+-sensing receptor-transient receptor potential-mediated Ca 2+ influx to extracellular acidity in bEND.3 endothelial cells. CHINESE J PHYSIOL 2022; 65:277-281. [PMID: 36588353 DOI: 10.4103/0304-4920.365460] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022] Open
Abstract
Ca2+-sensing receptors (CaSRs) are G protein-coupled receptors activated by elevated concentrations of extracellular Ca2+. In our previous works, we showed protein and functional expression of CaSR in mouse cerebral endothelial cell (EC) (bEND.3); the CaSR response (high Ca2+-elicited cytosolic [Ca2+] elevation) was unaffected by suppression of phospholipase C but in part involved Ca2+ influx through transient receptor potential V1 (TRPV1) channels. In this work, we investigated if extracellular acidity affected CaSR-mediated Ca2+ influx triggered by high (3 mM) Ca2+ (CaSR agonist), 3 mM spermine (CaSR agonist), and 10 mM cinacalcet (positive allosteric modulator of CaSR). Extracellular acidosis (pH 6.8 and pH 6.0) strongly suppressed cytosolic [Ca2+] elevation triggered by high Ca2+, spermine, and cinacalcet; acidosis also inhibited Mn2+ influx stimulated by high Ca2+ and cinacalcet. Purinoceptor-triggered Ca2+ response, however, was not suppressed by acidosis. Extracellular acidity also did not affect membrane potential, suggesting suppressed CaSR-mediated Ca2+ influx in acidity did not result from the reduced electrical driving force for Ca2+. Our results suggest Ca2+ influx through a putative CaSR-TRP complex in bEND.3 EC was sensitive to extracellular pH.
Collapse
Affiliation(s)
- Iat-Lon Leong
- Division of Cardiology, Department of Internal Medicine, Kiang Wu Hospital, Macau, China
| | - Chung-Ming Yu
- Department of Anesthesiology, Chang Gung Memorial Hospital, Chiayi, Taiwan
| | - Lian-Ru Shiao
- Department of Physiology, China Medical University, Taichung, Taiwan
| | - Paul Chan
- Division of Cardiology, Department of Medicine, Taipei Medical University Wan Fang Hospital, Taipei, Taiwan
| | - King-Chuen Wu
- Department of Anesthesiology, Chang Gung Memorial Hospital, Chiayi; Department of Nursing, Chang Gung University of Science and Technology, Chiayi; Department of Information Management, Shu-Zen Junior College of Medicine and Management, Kaohsiung, Taiwan
| | - Yuk-Man Leung
- Department of Physiology, China Medical University, Taichung, Taiwan
| |
Collapse
|
5
|
Li D, Wang T, Lai J, Zeng D, Chen W, Zhang X, Zhu X, Zhang G, Hu Z. Silencing TRPM2 enhanced erastin- and RSL3-induced ferroptosis in gastric cancer cells through destabilizing HIF-1α and Nrf2 proteins. Cytotechnology 2022; 74:559-577. [PMID: 36238268 PMCID: PMC9525503 DOI: 10.1007/s10616-022-00545-z] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2021] [Accepted: 07/25/2022] [Indexed: 11/03/2022] Open
Abstract
Ferroptosis is a regulated form of cell death driven by small molecules or conditions that induce lipid-based reactive oxygen species (ROS) accumulation. Cation channel transient receptor potential melastatin-2 (TRPM2) is crucial for cancer cell survival. Our bioinformatic analysis revealed that TRPM2 is associated with cellular responses to chemical stimulus and oxidative stress, implying the potential role of TRPM2 in ferroptosis. Gastric cancer cells were treated with the ferroptosis-inducer, Erastin and RSL3. siRNA transfection was used to silence TRPM2. The levels of GSH, Fe2+, ROS and lipid peroxidation, and the activity of GPx activity were evaluated by flow cytometry and spectrophotometer. The effect of TRPM2 on ubiquitination of HIF-1α and Nrf2 were evaluated by co-immunoprecipitation. Erastin and RSL3 induced the up-regulation of TRPM2 in gastric cancer cell lines, especially in SGC7901 and MGC803. These two cells also showed stronger resistance to Erastin and RSL3 than the other cell lines. TRPM2 knockdown reduced the concentration of GSH and GPx activity, but enhanced the concentration of Fe2+, ROS and lipid peroxidation, which are significant indicators of ferroptosis. Importantly, silencing TRPM2 enhanced the inhibitory effects of Erastin and RSL3 on gastric cancer cell viability, migration, and invasion. TRPM2 stabilized and finally elevated the abundance of HIF-1α and Nrf2 in SGC7901 and MGC803 cells upon Erastin and RSL3. Activation of HIF-1α impaired Erastin- and RSL3-induced ferroptosis after TRPM2 knockdown. Collectively, silencing TRPM2 enhanced Erastin- and RSL3-induced ferroptosis in gastric cancer cells through destabilizing HIF-1α and Nrf2 proteins. Supplementary Information The online version contains supplementary material available at 10.1007/s10616-022-00545-z.
Collapse
Affiliation(s)
- Dingyun Li
- Department of Gastrointestinal Surgery, Yue Bei People’s Hospital, No.133 Huimin South Road, Wujiang District, Shaoguan, 512026 Guangdong China
| | - Ting Wang
- Department of Physical Diagnosis, Yue Bei People’s Hospital, No. 133 Huimin South Road, Wujiang District, Shaoguan, 512026 Guangdong China
| | - Jiajun Lai
- Department of Gastrointestinal Surgery, Yue Bei People’s Hospital, No.133 Huimin South Road, Wujiang District, Shaoguan, 512026 Guangdong China
| | - Deqiang Zeng
- Department of Gastrointestinal Surgery, Yue Bei People’s Hospital, No.133 Huimin South Road, Wujiang District, Shaoguan, 512026 Guangdong China
| | - Weijuan Chen
- Clinical Laboratory, Yue Bei People’s Hospital, No. 133 Huimin South Road, Wujiang District, Shaoguan, 512026 Guangdong China
| | - Xiaochong Zhang
- Department of Gastrointestinal Surgery, Yue Bei People’s Hospital, No.133 Huimin South Road, Wujiang District, Shaoguan, 512026 Guangdong China
| | - Xiaofeng Zhu
- Department of Gastrointestinal Surgery, Yue Bei People’s Hospital, No.133 Huimin South Road, Wujiang District, Shaoguan, 512026 Guangdong China
| | - Guoxiong Zhang
- Department of Gastrointestinal Surgery, Yue Bei People’s Hospital, No.133 Huimin South Road, Wujiang District, Shaoguan, 512026 Guangdong China
| | - Zhiwei Hu
- Department of Gastrointestinal Surgery, Yue Bei People’s Hospital, No.133 Huimin South Road, Wujiang District, Shaoguan, 512026 Guangdong China
| |
Collapse
|
6
|
Audero MM, Prevarskaya N, Fiorio Pla A. Ca 2+ Signalling and Hypoxia/Acidic Tumour Microenvironment Interplay in Tumour Progression. Int J Mol Sci 2022; 23:7377. [PMID: 35806388 PMCID: PMC9266881 DOI: 10.3390/ijms23137377] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2022] [Revised: 06/26/2022] [Accepted: 06/27/2022] [Indexed: 01/18/2023] Open
Abstract
Solid tumours are characterised by an altered microenvironment (TME) from the physicochemical point of view, displaying a highly hypoxic and acidic interstitial fluid. Hypoxia results from uncontrolled proliferation, aberrant vascularization and altered cancer cell metabolism. Tumour cellular apparatus adapts to hypoxia by altering its metabolism and behaviour, increasing its migratory and metastatic abilities by the acquisition of a mesenchymal phenotype and selection of aggressive tumour cell clones. Extracellular acidosis is considered a cancer hallmark, acting as a driver of cancer aggressiveness by promoting tumour metastasis and chemoresistance via the selection of more aggressive cell phenotypes, although the underlying mechanism is still not clear. In this context, Ca2+ channels represent good target candidates due to their ability to integrate signals from the TME. Ca2+ channels are pH and hypoxia sensors and alterations in Ca2+ homeostasis in cancer progression and vascularization have been extensively reported. In the present review, we present an up-to-date and critical view on Ca2+ permeable ion channels, with a major focus on TRPs, SOCs and PIEZO channels, which are modulated by tumour hypoxia and acidosis, as well as the consequent role of the altered Ca2+ signals on cancer progression hallmarks. We believe that a deeper comprehension of the Ca2+ signalling and acidic pH/hypoxia interplay will break new ground for the discovery of alternative and attractive therapeutic targets.
Collapse
Affiliation(s)
- Madelaine Magalì Audero
- U1003—PHYCEL—Laboratoire de Physiologie Cellulaire, Inserm, University of Lille, Villeneuve d’Ascq, 59000 Lille, France; (M.M.A.); (N.P.)
- Laboratory of Cellular and Molecular Angiogenesis, Department of Life Sciences and Systems Biology, University of Turin, 10123 Turin, Italy
| | - Natalia Prevarskaya
- U1003—PHYCEL—Laboratoire de Physiologie Cellulaire, Inserm, University of Lille, Villeneuve d’Ascq, 59000 Lille, France; (M.M.A.); (N.P.)
| | - Alessandra Fiorio Pla
- U1003—PHYCEL—Laboratoire de Physiologie Cellulaire, Inserm, University of Lille, Villeneuve d’Ascq, 59000 Lille, France; (M.M.A.); (N.P.)
- Laboratory of Cellular and Molecular Angiogenesis, Department of Life Sciences and Systems Biology, University of Turin, 10123 Turin, Italy
| |
Collapse
|
7
|
Zong P, Feng J, Yue Z, Li Y, Wu G, Sun B, He Y, Miller B, Yu AS, Su Z, Xie J, Mori Y, Hao B, Yue L. Functional coupling of TRPM2 and extrasynaptic NMDARs exacerbates excitotoxicity in ischemic brain injury. Neuron 2022; 110:1944-1958.e8. [PMID: 35421327 DOI: 10.1016/j.neuron.2022.03.021] [Citation(s) in RCA: 41] [Impact Index Per Article: 20.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2021] [Revised: 02/12/2022] [Accepted: 03/14/2022] [Indexed: 12/18/2022]
Abstract
Excitotoxicity induced by NMDA receptor (NMDAR) activation is a major cause of neuronal death in ischemic stroke. However, past efforts of directly targeting NMDARs have unfortunately failed in clinical trials. Here, we reveal an unexpected mechanism underlying NMDAR-mediated neurotoxicity, which leads to the identification of a novel target and development of an effective therapeutic peptide for ischemic stroke. We show that NMDAR-induced excitotoxicity is enhanced by physical and functional coupling of NMDAR to an ion channel TRPM2 upon ischemic insults. TRPM2-NMDAR association promotes the surface expression of extrasynaptic NMDARs, leading to enhanced NMDAR activity and increased neuronal death. We identified a specific NMDAR-interacting motif on TRPM2 and designed a membrane-permeable peptide to uncouple the TRPM2-NMDAR interaction. This disrupting peptide protects neurons against ischemic injury in vitro and protects mice against ischemic stroke in vivo. These findings provide an unconventional strategy to mitigate excitotoxic neuronal death without directly targeting NMDARs.
Collapse
Affiliation(s)
- Pengyu Zong
- Department of Cell Biology, Calhoun Cardiology Center, University of Connecticut School of Medicine (UConn Health), Farmington, CT 06030, USA
| | - Jianlin Feng
- Department of Cell Biology, Calhoun Cardiology Center, University of Connecticut School of Medicine (UConn Health), Farmington, CT 06030, USA
| | - Zhichao Yue
- Department of Cell Biology, Calhoun Cardiology Center, University of Connecticut School of Medicine (UConn Health), Farmington, CT 06030, USA
| | - Yunfeng Li
- Department of Molecular Biology and Biophysics, University of Connecticut School of Medicine (UConn Health), Farmington, CT 06030, USA
| | - Gongxiong Wu
- Department of Medicine, Brigham and Women's Hospital, Laboratory for Translational Research, Harvard Medical School, Cambridge, MA 02139, USA
| | - Baonan Sun
- Department of Cell Biology, Calhoun Cardiology Center, University of Connecticut School of Medicine (UConn Health), Farmington, CT 06030, USA
| | - Yanlin He
- Department of Cell Biology, Calhoun Cardiology Center, University of Connecticut School of Medicine (UConn Health), Farmington, CT 06030, USA
| | - Barbara Miller
- Departments of Pediatrics and Biochemistry and Molecular Biology, The Pennsylvania State University College of Medicine, P.O. Box 850, Hershey, PA 17033, USA
| | - Albert S Yu
- Department of Cell Biology, Calhoun Cardiology Center, University of Connecticut School of Medicine (UConn Health), Farmington, CT 06030, USA
| | - Zhongping Su
- Department of Cell Biology, Calhoun Cardiology Center, University of Connecticut School of Medicine (UConn Health), Farmington, CT 06030, USA
| | - Jia Xie
- Department of Cell Biology, Calhoun Cardiology Center, University of Connecticut School of Medicine (UConn Health), Farmington, CT 06030, USA
| | - Yasuo Mori
- Laboratory of Molecular Biology, Department of Synthetic Chemistry and Biological Chemistry, Graduate School of Engineering, Kyoto University, Kyoto 615-8510, Japan; The World Premier International Research Initiative, Institute for Integrated Cell-Material Sciences, Kyoto University, Kyoto 615-8510, Japan
| | - Bing Hao
- Department of Molecular Biology and Biophysics, University of Connecticut School of Medicine (UConn Health), Farmington, CT 06030, USA
| | - Lixia Yue
- Department of Cell Biology, Calhoun Cardiology Center, University of Connecticut School of Medicine (UConn Health), Farmington, CT 06030, USA.
| |
Collapse
|
8
|
Zhao S, Zhang H, Jin H, Cai X, Zhang R, Jin Z, Yang W, Yu P, Zhang L, Liu Z. Design, synthesis and biological activities of benzo[d]imidazo[1,2-a]imidazole derivatives as TRPM2-specfic inhibitors. Eur J Med Chem 2021; 225:113750. [PMID: 34416664 DOI: 10.1016/j.ejmech.2021.113750] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2021] [Revised: 07/26/2021] [Accepted: 08/04/2021] [Indexed: 10/20/2022]
Abstract
Transient receptor potential melastatin 2 (TRPM2) channel is associated with ischemia/reperfusion injury, inflammation, cancer and neurodegenerative diseases. However, the lack of specific inhibitors impedes the development of TRPM2 targeted therapeutic agents. To develop a selective TRPM2 inhibitor, three-dimensional similarity-based screening strategy was employed using the energy-minimized conformation of non-selective TRPM2 inhibitor 2-APB as the query structure, which resulted in the discovery of a novel tricyclic TRPM2 inhibitor Z-4 with benzo[d]imidazo[1,2-a]imidazole skeleton. A series of Z-4 derivatives were subsequently synthesized and evaluated using calcium imaging and electrophysiology approaches. Among them, preferred compounds ZA10 and ZA18 inhibited the TRPM2 channel with micromolar half-maximal inhibitory concentration values and exhibited TRPM2 selectivity over the TRPM8 channel, TRPV1 channel, InsP3 receptor and Orai channel. The analysis of structure-activity relationship provides valuable insights for further development of selective TRPM2 inhibitors. Neuroprotection assay showed that ZA10 and ZA18 could effectively reduce the mortality of SH-SY5Y cells induced by H2O2. These findings enrich the structure types of existing TRPM2 inhibitors and might provide a new tool for the study of TRPM2 function in Reactive oxygen species (ROS) -related diseases.
Collapse
Affiliation(s)
- Siqi Zhao
- State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, Beijing, 100191, PR China
| | - Han Zhang
- State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, Beijing, 100191, PR China
| | - Hongwei Jin
- State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, Beijing, 100191, PR China
| | - Xiaobo Cai
- Department of Biophysics, Zhejiang University School of Medicine, Hangzhou, Zhejiang, 310058, PR China
| | - Rongxue Zhang
- State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, Beijing, 100191, PR China
| | - Zefang Jin
- State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, Beijing, 100191, PR China
| | - Wei Yang
- Department of Biophysics, Zhejiang University School of Medicine, Hangzhou, Zhejiang, 310058, PR China
| | - Peilin Yu
- Department of Toxicology, School of Public Health, Zhejiang University, Hangzhou, Zhejiang, 310058, PR China.
| | - Liangren Zhang
- State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, Beijing, 100191, PR China.
| | - Zhenming Liu
- State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, Beijing, 100191, PR China.
| |
Collapse
|
9
|
Wang X, Xiao Y, Huang M, Shen B, Xue H, Wu K. Effect of TRPM2-Mediated Calcium Signaling on Cell Proliferation and Apoptosis in Esophageal Squamous Cell Carcinoma. Technol Cancer Res Treat 2021; 20:15330338211045213. [PMID: 34605693 PMCID: PMC8642046 DOI: 10.1177/15330338211045213] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
Esophageal squamous cell carcinoma (ESCC) is the sixth leading cause of death due to
cancer, indicating that finding new therapeutic targets or approaches for ESCC treatment
is imperative. Transient Receptor Potential cation channel subfamily M, member 2 (TRPM2)
is a calcium-permeable, nonselective cation channel that responds to reactive oxygen
species (ROS), which are found in the tumor microenvironment and are important regulators
of tumorigenesis, cell proliferation, apoptosis, and the therapeutic response. Here, we
used immunohistochemical analysis of tumor tissue derived from patients with ESCC to find
that the TRPM2 channel protein expression level was increased in tumor tissue compared
with adjacent normal tissue. Intracellular calcium concentration measurements, western
blotting, and ROS and cell viability assays were used with a human ESCC cell line (TE-1
cells) to find that TRPM2 participated in the ROS hydrogen peroxide-induced increase in
intracellular calcium. This increased calcium inhibited cell proliferation and enhanced
apoptosis. Pretreatment of cells with the anticancer agent 5-fluorouracil (5-FU)
significantly increased ROS production, which potentiated TRPM2-mediated calcium
signaling, decreased cell proliferation, and increased apoptosis in TE-1 cells, suggesting
that the therapeutic effect of 5-FU in ESCC cells may be mediated by the TRPM2
channel-mediated calcium influx. These findings offer a potential treatment target and
provide mechanistic insight into the therapeutic effects of 5-FU in patients with
ESCC.
Collapse
Affiliation(s)
- Xingbang Wang
- Lu'an People's Hospital, Lu'an Affiliated Hospital of Anhui Medical University, Lu'an, China
| | - Yong Xiao
- Lu'an People's Hospital, Lu'an Affiliated Hospital of Anhui Medical University, Lu'an, China
| | - Mingming Huang
- Lu'an People's Hospital, Lu'an Affiliated Hospital of Anhui Medical University, Lu'an, China
| | - Bing Shen
- School of Basic Medical Sciences, Anhui Medical University, Hefei, China
| | - Haowei Xue
- 36639The First Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Kaile Wu
- 36639The First Affiliated Hospital of Anhui Medical University, Hefei, China
| |
Collapse
|
10
|
Zhang H, Yu P, Lin H, Jin Z, Zhao S, Zhang Y, Xu Q, Jin H, Liu Z, Yang W, Zhang L. The Discovery of Novel ACA Derivatives as Specific TRPM2 Inhibitors that Reduce Ischemic Injury Both In Vitro and In Vivo. J Med Chem 2021; 64:3976-3996. [PMID: 33784097 DOI: 10.1021/acs.jmedchem.0c02129] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
The transient receptor potential melastatin 2 (TRPM2) channel is associated with ischemia/reperfusion injury, inflammation, cancer, and neurodegenerative diseases. However, the limit of specific inhibitors impedes the development of TRPM2-targeted therapeutic agents. To discover more potent and selective TRPM2 inhibitors, 59 N-(p-amylcinnamoyl) anthranilic acid (ACA) derivatives were synthesized and evaluated using calcium imaging and electrophysiology approaches. Systematic structure-activity relationship studies resulted in some potent compounds inhibiting the TRPM2 channel with sub-micromolar half-maximal inhibitory concentration values. Among them, the preferred compound A23 exhibited TRPM2 selectivity over TRPM8 and TRPV1 channels as well as phospholipase A2 and showed neuroprotective activity in vitro. Following pharmacokinetic studies, A23 was further evaluated in a transient middle cerebral artery occlusion model in vivo, which significantly reduced cerebral infarction. These data indicate that A23 might serve as a useful tool for TRPM2-related research as well as a lead compound for the development of therapeutic agents for ischemic injury.
Collapse
Affiliation(s)
- Han Zhang
- State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, Beijing 100191, P. R. China
| | - Peilin Yu
- Department of Toxicology, and Department of Medical Oncology of Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310058, Zhejiang, P. R. China
| | - Hongwei Lin
- Department of Biophysics, and Department of Neurosurgery of the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310058, P. R. China
| | - Zefang Jin
- State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, Beijing 100191, P. R. China
| | - Siqi Zhao
- State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, Beijing 100191, P. R. China
| | - Yi Zhang
- Department of Biophysics, and Department of Neurosurgery of the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310058, P. R. China
| | - Qingxia Xu
- State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, Beijing 100191, P. R. China
| | - Hongwei Jin
- State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, Beijing 100191, P. R. China
| | - Zhenming Liu
- State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, Beijing 100191, P. R. China
| | - Wei Yang
- Department of Biophysics, and Department of Neurosurgery of the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310058, P. R. China
| | - Liangren Zhang
- State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, Beijing 100191, P. R. China
| |
Collapse
|
11
|
Hossain Saad MZ, Xiang L, Liao YS, Reznikov LR, Du J. The Underlying Mechanism of Modulation of Transient Receptor Potential Melastatin 3 by protons. Front Pharmacol 2021; 12:632711. [PMID: 33603674 PMCID: PMC7884864 DOI: 10.3389/fphar.2021.632711] [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: 11/23/2020] [Accepted: 01/04/2021] [Indexed: 12/03/2022] Open
Abstract
Transient receptor potential melastatin 3 channel (TRPM3) is a calcium-permeable nonselective cation channel that plays an important role in modulating glucose homeostasis in the pancreatic beta cells. However, how TRPM3 is regulated under physiological and pathological conditions is poorly understood. In this study, we found that both intracellular and extracellular protons block TRPM3 through its binding sites in the pore region. We demonstrated that external protons block TRPM3 with an inhibitory pH50 of 5.5. whereas internal protons inhibit TRPM3 with an inhibitory pH50 of 6.9. We identified three titratable residues, D1059, D1062, and D1073, at the vestibule of the channel pore that contributes to pH sensitivity. The mutation of D1073Q reduced TRPM3 current by low external pH 5.5 from 62 ± 3% in wildtype to 25 ± 6.0% in D1073Q mutant. These results indicate that D1073 is essential for pH sensitivity. In addition, we found that a single mutation of D1059 or D1062 enhanced pH sensitivity. In summary, our findings identify molecular determinants respionsible for the pH regulation of TRPM3. The inhibition of TRPM3 by protons may indicate an endogenous mechanism governing TRPM3 gating and its physiological/pathological functions.
Collapse
Affiliation(s)
- Md Zubayer Hossain Saad
- Department of Anatomy and Neurobiology, University of Tennessee Health Science Center, Memphis, TN, United States.,Department of Biological Sciences, University of Toledo, Toledo, OH, United States
| | - Liuruimin Xiang
- Department of Anatomy and Neurobiology, University of Tennessee Health Science Center, Memphis, TN, United States.,Department of Biological Sciences, University of Toledo, Toledo, OH, United States.,Program of Neuroscience, University of Maryland School of Medicine, Baltimore, MD, United States
| | - Yan-Shin Liao
- Department of Physiological Sciences, University of Florida, Gainesville, FL, United States
| | - Leah R Reznikov
- Department of Physiological Sciences, University of Florida, Gainesville, FL, United States
| | - Jianyang Du
- Department of Anatomy and Neurobiology, University of Tennessee Health Science Center, Memphis, TN, United States.,Neuroscience Institute, University of Tennessee Health Science Center, Memphis, TN, United States
| |
Collapse
|
12
|
Pala S, Atilgan R, Kuloglu T, Yalçın E, Kaya N, Etem E. The decrease in hippocampal transient receptor potential M2 (TRPM2) channel and muscarinic acetylcholine receptor 1 (CHRM1) is associated with memory loss in a surgical menopause rat model. Arch Med Sci 2021; 17:228-235. [PMID: 33488875 PMCID: PMC7811316 DOI: 10.5114/aoms.2019.83760] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/15/2018] [Accepted: 03/01/2019] [Indexed: 11/17/2022] Open
Abstract
INTRODUCTION The aim of the study was to investigate the association of transient receptor potential M2 (TRPM2) channel and muscarinic acetylcholine receptor 1 (CHRM1) activity with the memorial functions that are deteriorated in surgical menopause. MATERIAL AND METHODS A total of 14 female rats were randomly divided into 2 groups: group (G)1: sham group; group (G)2: surgical menopause group, the group in which bilateral ovariectomy was performed. Fourteen days after the surgical procedure, learning and memorial tests were performed in G1 and G2 for a totally 13 days. The time required for the rats to find the cheese in the labyrinth was recorded and statistical evaluation of it was performed between groups. On the 14th day of the memory test, the rats were decapitated and the brain tissues were fixed in 10% formalin. Hippocampal TRPM2 and CHRM1 gene expression was evaluated with RNA isolation, complementary DNA (cDNA) synthesis and quantitative real-time PCR (qRT-PCR) analysis. TRPM2 and CHRM1 immunoreactivity was evaluated in hippocampal tissue with the immunohistochemical method. Histo-score was calculated regarding the diffuseness of and severity of the staining; and statistical analyses were performed. RESULTS In the ovariectomized group, the mean time required for the rats to find the cheese was statistically significantly elongated (39.29 ±4.0 s vs. 29.86 ±2.6 s). When the hippocampal TRPM2 and CHRM1 gene expression and immunoreactivity were compared with the sham group, there was a statistically significant decrease in the surgical menopause group (p < 0.05). CONCLUSIONS In surgical menopause, in deterioration of memorial functions, hippocampal TRPM2 channel and CHRM1 activity plays an important role.
Collapse
Affiliation(s)
- Sehmus Pala
- Department of Obstetrics and Gynecology, School of Medicine, Firat University, Elazig, Turkey
| | - Remzi Atilgan
- Department of Obstetrics and Gynecology, School of Medicine, Firat University, Elazig, Turkey
| | - Tuncay Kuloglu
- Department of Histology and Embryology, School of Medicine, Firat University, Elazig, Turkey
| | - Emre Yalçın
- Department of Obstetrics and Gynecology, School of Medicine, Firat University, Elazig, Turkey
| | - Nalan Kaya
- Department of Histology and Embryology, School of Medicine, Firat University, Elazig, Turkey
| | - Ebru Etem
- Department of Medical Biology, School of Medicine, Firat University, Elazig, Turkey
| |
Collapse
|
13
|
Yu P, Cai X, Liang Y, Wang M, Yang W. Roles of NAD + and Its Metabolites Regulated Calcium Channels in Cancer. Molecules 2020; 25:molecules25204826. [PMID: 33092205 PMCID: PMC7587972 DOI: 10.3390/molecules25204826] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2020] [Revised: 10/11/2020] [Accepted: 10/16/2020] [Indexed: 02/08/2023] Open
Abstract
Nicotinamide adenine dinucleotide (NAD+) is an essential cofactor for redox enzymes, but also moonlights as a regulator for ion channels, the same as its metabolites. Ca2+ homeostasis is dysregulated in cancer cells and affects processes such as tumorigenesis, angiogenesis, autophagy, progression, and metastasis. Herein, we summarize the regulation of the most common calcium channels (TRPM2, TPCs, RyRs, and TRPML1) by NAD+ and its metabolites, with a particular focus on their roles in cancers. Although the mechanisms of NAD+ metabolites in these pathological processes are yet to be clearly elucidated, these ion channels are emerging as potential candidates of alternative targets for anticancer therapy.
Collapse
Affiliation(s)
- Peilin Yu
- Department of Toxicology, and Department of Medical Oncology of Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310058, Zhejiang, China; (P.Y.); (Y.L.)
| | - Xiaobo Cai
- Department of Biophysics, and Department of Neurosurgery of the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310058, Zhejiang, China;
| | - Yan Liang
- Department of Toxicology, and Department of Medical Oncology of Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310058, Zhejiang, China; (P.Y.); (Y.L.)
| | - Mingxiang Wang
- BrioPryme Biologics, Inc., Hangzhou 310058, Zhejiang, China;
| | - Wei Yang
- Department of Biophysics, and Department of Neurosurgery of the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310058, Zhejiang, China;
- Correspondence: ; Tel.: +86-571-8820-8713
| |
Collapse
|
14
|
Pethő Z, Najder K, Carvalho T, McMorrow R, Todesca LM, Rugi M, Bulk E, Chan A, Löwik CWGM, Reshkin SJ, Schwab A. pH-Channeling in Cancer: How pH-Dependence of Cation Channels Shapes Cancer Pathophysiology. Cancers (Basel) 2020; 12:E2484. [PMID: 32887220 PMCID: PMC7565548 DOI: 10.3390/cancers12092484] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2020] [Revised: 08/25/2020] [Accepted: 08/26/2020] [Indexed: 12/20/2022] Open
Abstract
Tissue acidosis plays a pivotal role in tumor progression: in particular, interstitial acidosis promotes tumor cell invasion, and is a major contributor to the dysregulation of tumor immunity and tumor stromal cells. The cell membrane and integral membrane proteins commonly act as important sensors and transducers of altered pH. Cell adhesion molecules and cation channels are prominent membrane proteins, the majority of which is regulated by protons. The pathophysiological consequences of proton-sensitive ion channel function in cancer, however, are scarcely considered in the literature. Thus, the main focus of this review is to highlight possible events in tumor progression and tumor immunity where the pH sensitivity of cation channels could be of great importance.
Collapse
Affiliation(s)
- Zoltán Pethő
- Institute of Physiology II, University Münster, 48147 Münster, Germany; (K.N.); (L.M.T.); (M.R.); (E.B.); (A.S.)
| | - Karolina Najder
- Institute of Physiology II, University Münster, 48147 Münster, Germany; (K.N.); (L.M.T.); (M.R.); (E.B.); (A.S.)
| | - Tiago Carvalho
- Department of Biosciences, Biotechnologies, and Biopharmaceutics, University of Bari, 90126 Bari, Italy; (T.C.); (S.J.R.)
| | - Roisin McMorrow
- Department of Radiology and Nuclear Medicine, Erasmus Medical Center, 3035 GD Rotterdam, The Netherlands; (R.M.); (C.W.G.M.L.)
| | - Luca Matteo Todesca
- Institute of Physiology II, University Münster, 48147 Münster, Germany; (K.N.); (L.M.T.); (M.R.); (E.B.); (A.S.)
| | - Micol Rugi
- Institute of Physiology II, University Münster, 48147 Münster, Germany; (K.N.); (L.M.T.); (M.R.); (E.B.); (A.S.)
| | - Etmar Bulk
- Institute of Physiology II, University Münster, 48147 Münster, Germany; (K.N.); (L.M.T.); (M.R.); (E.B.); (A.S.)
| | - Alan Chan
- Percuros B.V., 2333 CL Leiden, The Netherlands;
| | - Clemens W. G. M. Löwik
- Department of Radiology and Nuclear Medicine, Erasmus Medical Center, 3035 GD Rotterdam, The Netherlands; (R.M.); (C.W.G.M.L.)
- Department of Oncology CHUV, UNIL and Ludwig Cancer Center, 1011 Lausanne, Switzerland
| | - Stephan J. Reshkin
- Department of Biosciences, Biotechnologies, and Biopharmaceutics, University of Bari, 90126 Bari, Italy; (T.C.); (S.J.R.)
| | - Albrecht Schwab
- Institute of Physiology II, University Münster, 48147 Münster, Germany; (K.N.); (L.M.T.); (M.R.); (E.B.); (A.S.)
| |
Collapse
|
15
|
Yu P, Liu Z, Yu X, Ye P, Liu H, Xue X, Yang L, Li Z, Wu Y, Fang C, Zhao YJ, Yang F, Luo JH, Jiang LH, Zhang L, Zhang L, Yang W. Direct Gating of the TRPM2 Channel by cADPR via Specific Interactions with the ADPR Binding Pocket. Cell Rep 2020; 27:3684-3695.e4. [PMID: 31216484 DOI: 10.1016/j.celrep.2019.05.067] [Citation(s) in RCA: 47] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2018] [Revised: 02/05/2019] [Accepted: 05/18/2019] [Indexed: 12/29/2022] Open
Abstract
cADPR is a well-recognized signaling molecule by modulating the RyRs, but considerable debate exists regarding whether cADPR can bind to and gate the TRPM2 channel, which mediates oxidative stress signaling in diverse physiological and pathological processes. Here, we show that purified cADPR evoked TRPM2 channel currents in both whole-cell and cell-free single-channel recordings and specific binding of cADPR to the purified NUDT9-H domain of TRPM2 by surface plasmon resonance. Furthermore, by combining computational modeling with electrophysiological recordings, we show that the TRPM2 channels carrying point mutations at H1346, T1347, L1379, S1391, E1409, and L1484 possess distinct sensitivity profiles for ADPR and cADPR. These results clearly indicate cADPR is a bona fide activator at the TRPM2 channel and clearly delineate the structural basis for cADPR binding, which not only lead to a better understanding in the gating mechanism of TRPM2 channel but also shed light on a cADPR-induced RyRs-independent Ca2+ signaling mechanism.
Collapse
Affiliation(s)
- Peilin Yu
- Department of Biophysics, Institute of Neuroscience, NHC and CAMS Key Laboratory of Medical Neurobiology, Zhejiang University School of Medicine, Hangzhou 310058, P.R. China; Department of Toxicology, School of Public Health, Zhejiang University, Hangzhou, Zhejiang 310058, P.R. China
| | - Zhenming Liu
- State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, Beijing 100191, P.R. China
| | - Xiafei Yu
- Department of Biophysics, Institute of Neuroscience, NHC and CAMS Key Laboratory of Medical Neurobiology, Zhejiang University School of Medicine, Hangzhou 310058, P.R. China
| | - Peiwu Ye
- Department of Biophysics, Institute of Neuroscience, NHC and CAMS Key Laboratory of Medical Neurobiology, Zhejiang University School of Medicine, Hangzhou 310058, P.R. China
| | - Huan Liu
- Department of Biophysics, Institute of Neuroscience, NHC and CAMS Key Laboratory of Medical Neurobiology, Zhejiang University School of Medicine, Hangzhou 310058, P.R. China
| | - Xiwen Xue
- State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, Beijing 100191, P.R. China
| | - Lixin Yang
- State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, Beijing 100191, P.R. China
| | - Zhongtang Li
- State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, Beijing 100191, P.R. China
| | - Yang Wu
- Laboratory of Cytophysiology, State Key Laboratory of Chemical Oncogenomics, Key Laboratory of Chemical Genomics, Peking University Shenzhen Graduate School, Shenzhen, Guangdong 518055, P.R. China
| | - Cheng Fang
- Laboratory of Cytophysiology, State Key Laboratory of Chemical Oncogenomics, Key Laboratory of Chemical Genomics, Peking University Shenzhen Graduate School, Shenzhen, Guangdong 518055, P.R. China
| | - Yong Juan Zhao
- Laboratory of Cytophysiology, State Key Laboratory of Chemical Oncogenomics, Key Laboratory of Chemical Genomics, Peking University Shenzhen Graduate School, Shenzhen, Guangdong 518055, P.R. China
| | - Fan Yang
- Department of Biophysics, Institute of Neuroscience, NHC and CAMS Key Laboratory of Medical Neurobiology, Zhejiang University School of Medicine, Hangzhou 310058, P.R. China; Kidney Disease Center, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, Zhejiang 310058, P.R. China
| | - Jian Hong Luo
- Department of Neurobiology, Institute of Neuroscience, NHC and CAMS Key Laboratory of Medical Neurobiology, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310058, P.R. China
| | - Lin-Hua Jiang
- School of Biomedical Sciences, Faculty of Biological Sciences, University of Leeds, Leeds LS2 9JT, UK; Sino-UK Laboratory of Brain Function and Injury of Henan Province and Department of Physiology and Neurobiology, Xinxiang Medical University, Henan 453003, P.R. China
| | - Liangren Zhang
- State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, Beijing 100191, P.R. China
| | - Lihe Zhang
- State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, Beijing 100191, P.R. China
| | - Wei Yang
- Department of Biophysics, Institute of Neuroscience, NHC and CAMS Key Laboratory of Medical Neurobiology, Zhejiang University School of Medicine, Hangzhou 310058, P.R. China; Department of Neurosurgery, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, Zhejiang 310058, P.R. China.
| |
Collapse
|
16
|
Luo YH, Yu XF, Ma C, Yang F, Yang W. Effects of calcium-binding sites in the S2-S3 loop on human and Nematostella vectensis TRPM2 channel gating processes. J Zhejiang Univ Sci B 2020; 20:972-982. [PMID: 31749344 DOI: 10.1631/jzus.b1900477] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
As a crucial signaling molecule, calcium plays a critical role in many physiological and pathological processes by regulating ion channel activity. Recently, one study resolved the structure of the transient receptor potential melastatin 2 (TRPM2) channel from Nematostella vectensis (nvTRPM2). This identified a calcium-binding site in the S2-S3 loop, while its effect on channel gating remains unclear. Here, we investigated the role of this calcium-binding site in both nvTRPM2 and human TRPM2 (hTRPM2) by mutagenesis and patch-clamp recording. Unlike hTRPM2, nvTRPM2 cannot be activated by calcium alone. Moreover, the inactivation rate of nvTRPM2 was decreased as intracellular calcium concentration was increased. In addition, our results showed that the four key residues in the calcium-binding site of S2-S3 loop have similar effects on the gating processes of nvTRPM2 and hTRPM2. Among them, the mutations at negatively charged residues (glutamate and aspartate) substantially decreased the currents of nvTRPM2 and hTRPM2. This suggests that these sites are essential for calcium-dependent channel gating. For the charge-neutralizing residues (glutamine and asparagine) in the calcium-binding site, our data showed that glutamine mutating to alanine or glutamate did not affect the channel activity, but glutamine mutating to lysine caused loss of function. Asparagine mutating to aspartate still remained functional, while asparagine mutating to alanine or lysine led to little channel activity. These results suggest that the side chain of glutamine has a less contribution to channel gating than does asparagine. However, our data indicated that both glutamine mutating to alanine or glutamate and asparagine mutating to aspartate accelerated the channel inactivation rate, suggesting that the calcium-binding site in the S2-S3 loop is important for calcium-dependent channel inactivation. Taken together, our results uncovered the effect of four key residues in the S2-S3 loop of TRPM2 on the TRPM2 gating process.
Collapse
Affiliation(s)
- Yu-Huan Luo
- Department of Biophysics, Institute of Neuroscience, NHC and CAMS Key Laboratory of Medical Neurobiology, School of Medicine, Zhejiang University, Hangzhou 310058, China.,Department of Pediatric, Affiliated Hangzhou First People's Hospital, Zhejiang University School of Medicine, Hangzhou 310006, China.,Translational Medicine Research Center, Key Laboratory of Clinical Cancer Pharmacology and Toxicology Research of Zhejiang Province, Affiliated Hangzhou First People's Hospital, Zhejiang University School of Medicine, Hangzhou 310006, China
| | - Xia-Fei Yu
- Department of Biophysics, Institute of Neuroscience, NHC and CAMS Key Laboratory of Medical Neurobiology, School of Medicine, Zhejiang University, Hangzhou 310058, China
| | - Cheng Ma
- Co-facility Center, Zhejiang University School of Medicine, Hangzhou 310058, China
| | - Fan Yang
- Department of Biophysics, Institute of Neuroscience, NHC and CAMS Key Laboratory of Medical Neurobiology, School of Medicine, Zhejiang University, Hangzhou 310058, China
| | - Wei Yang
- Department of Biophysics, Institute of Neuroscience, NHC and CAMS Key Laboratory of Medical Neurobiology, School of Medicine, Zhejiang University, Hangzhou 310058, China.,Department of Neurosurgery, the First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou 310003, China
| |
Collapse
|
17
|
Luo Y, Yu X, Ma C, Luo J, Yang W. Identification of a Novel EF-Loop in the N-terminus of TRPM2 Channel Involved in Calcium Sensitivity. Front Pharmacol 2018; 9:581. [PMID: 29915540 PMCID: PMC5994415 DOI: 10.3389/fphar.2018.00581] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2018] [Accepted: 05/15/2018] [Indexed: 01/10/2023] Open
Abstract
As an oxidative stress sensor, transient receptor potential melastatin 2 (TRPM2) channel is involved in many physiological and pathological processes including warmth sensing, ischemia injury, inflammatory diseases and diabetes. Intracellular calcium is critical for TRPM2 channel activation and the IQ-like motif in the N-terminus has been shown to be important by mediating calmodulin binding. Sequence analysis predicted two potential EF-loops in the N-terminus of TRPM2. Site-directed mutagenesis combining with functional assay showed that substitution with alanine of several residues, most of which are conserved in the typical EF-loop, including D267, D278, D288, and E298 dramatically reduced TRPM2 channel currents. By further changing the charges or side chain length of these conserved residues, our results indicate that the negative charge of D267 and the side chain length of D278 are critical for calcium-induced TRPM2 channel activation. G272I mutation also dramatically reduced the channel currents, suggesting that this site is critical for calcium-induced TRPM2 channel activation. Furthermore, D267A mutant dramatically reduced the currents induced by calcium alone compared with that by ADPR, indicating that D267 residue in D267–D278 motif is the most important site for calcium sensitivity of TRPM2. In addition, inside-out recordings showed that mutations at D267, G272, D278, and E298 had no effect on single-channel conductance. Taken together, our data indicate that D267–D278 motif in the N-terminus as a novel EF-loop is critical for calcium-induced TRPM2 channel activation.
Collapse
Affiliation(s)
- Yuhuan Luo
- Department of Neurobiology, Institute of Neuroscience, NHC and CAMS Key Laboratory of Medical Neurobiology, Zhejiang University School of Medicine, Hangzhou, China
| | - Xiafei Yu
- Department of Neurobiology, Institute of Neuroscience, NHC and CAMS Key Laboratory of Medical Neurobiology, Zhejiang University School of Medicine, Hangzhou, China
| | - Cheng Ma
- Co-facility Center, Zhejiang University School of Medicine, Hangzhou, China
| | - Jianhong Luo
- Department of Neurobiology, Institute of Neuroscience, NHC and CAMS Key Laboratory of Medical Neurobiology, Zhejiang University School of Medicine, Hangzhou, China
| | - Wei Yang
- Department of Neurobiology, Institute of Neuroscience, NHC and CAMS Key Laboratory of Medical Neurobiology, Zhejiang University School of Medicine, Hangzhou, China
| |
Collapse
|
18
|
Belrose JC, Jackson MF. TRPM2: a candidate therapeutic target for treating neurological diseases. Acta Pharmacol Sin 2018; 39:722-732. [PMID: 29671419 PMCID: PMC5943913 DOI: 10.1038/aps.2018.31] [Citation(s) in RCA: 77] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2017] [Accepted: 02/24/2018] [Indexed: 12/25/2022] Open
Abstract
Transient receptor potential melastatin 2 (TRPM2) is a calcium (Ca2+)-permeable non-selective cation channel belonging to the TRP ion channel family. Oxidative stress-induced TRPM2 activation provokes aberrant intracellular Ca2+ accumulation and cell death in a variety of cell types, including neurons. Aberrant TRPM2 function has been implicated in several neurological disorders including ischemia/stroke, Alzheimer's disease, neuropathic pain, Parkinson's disease and bipolar disorder. In addition to research identifying a role for TRPM2 in disease, progress has been made in the identification of physiological functions of TRPM2 in the brain, including recent evidence that TRPM2 is necessary for the induction of N-methyl-D-aspartate (NMDA) receptor-dependent long-term depression, an important form of synaptic plasticity at glutamate synapses. Here, we summarize recent evidence on the role of TRPM2 in the central nervous system (CNS) in health and disease and discuss the potential therapeutic implications of targeting TRPM2. Collectively, these studies suggest that TRPM2 represents a prospective novel therapeutic target for neurological disorders.
Collapse
Affiliation(s)
- Jillian Corinne Belrose
- Department of Anesthesia & Perioperative Medicine, Schulich Medicine & Dentistry, Western University, London, Ontario, N6A 5A5, Canada
- E-mail
| | - Michael Frederick Jackson
- Department of Pharmacology and Therapeutics, Rady Faculty of Health Sciences, Max Rady College of Medicine, University of Manitoba, Winnipeg, Manitoba, R3E 0T6, Canada
- Neuroscience Research Program, Kleysen Institute for Advanced Medicine, Health Sciences Centre, Winnipeg, Manitoba, R3E 3J7, Canada
| |
Collapse
|
19
|
The role of TRPM2 channels in neurons, glial cells and the blood-brain barrier in cerebral ischemia and hypoxia. Acta Pharmacol Sin 2018. [PMID: 29542681 PMCID: PMC5943904 DOI: 10.1038/aps.2017.194] [Citation(s) in RCA: 43] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
Stroke is one of the major causes of mortality and morbidity worldwide, yet novel therapeutic treatments for this condition are lacking. This review focuses on the roles of the transient receptor potential melastatin 2 (TRPM2) ion channels in cellular damage following hypoxia-ischemia and their potential as a future therapeutic target for stroke. Here, we highlight the complex molecular signaling that takes place in neurons, glial cells and the blood-brain barrier following ischemic insult. We also describe the evidence of TRPM2 involvement in these processes, as shown from numerous in vitro and in vivo studies that utilize genetic and pharmacological approaches. This evidence implicates TRPM2 in a broad range of pathways that take place every stage of cerebral ischemic injury, thus making TRPM2 a promising target for drug development for stroke and other neurodegenerative conditions of the central nervous system.
Collapse
|
20
|
Zhang H, Liu H, Luo X, Wang Y, Liu Y, Jin H, Liu Z, Yang W, Yu P, Zhang L, Zhang L. Design, synthesis and biological activities of 2,3-dihydroquinazolin-4(1H)-one derivatives as TRPM2 inhibitors. Eur J Med Chem 2018; 152:235-252. [PMID: 29723786 DOI: 10.1016/j.ejmech.2018.04.045] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2018] [Revised: 04/06/2018] [Accepted: 04/21/2018] [Indexed: 12/26/2022]
Abstract
Transient receptor potential melastatin 2 (TRPM2), a Ca2+-permeable cationic channel, plays critical roles in insulin release, cytokine production, body temperature regulation and cell death as a reactive oxygen species (ROS) and temperature sensor. However, few TRPM2 inhibitors have been reported, especially TRP-subtype selective inhibitors, which hampers the investigation and validation of TRPM2 as a drug target. To discover novel TRPM2 inhibitors, 3D similarity-based virtual screening method was employed, by which 2,3-dihydroquinazolin-4(1H)-one derivative H1 was identified as a TRPM2 inhibitor. A series of novel 2,3-dihydroquinazolin-4(1H)-one derivatives were subsequently synthesized and characterized. Their inhibitory activities against the TRPM2 channel were evaluated by calcium imaging and electrophysiology approaches. Some of the compounds exhibited significant inhibitory activity, especially D9 which showed an IC50 of 3.7 μM against TRPM2 and did not affect the TRPM8 channel. The summarized structure-activity relationship (SAR) provides valuable insights for further development of specific TRPM2 targeted inhibitors.
Collapse
Affiliation(s)
- Han Zhang
- State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, Beijing, 100191, PR China
| | - Huan Liu
- Department of Neurobiology, Key Laboratory of Medical Neurobiology of the Ministry of Health of China, Zhejiang University School of Medicine, Hangzhou, Zhejiang, 310058, PR China
| | - Xiao Luo
- State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, Beijing, 100191, PR China
| | - Yuxi Wang
- State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, Beijing, 100191, PR China
| | - Yuan Liu
- State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, Beijing, 100191, PR China
| | - Hongwei Jin
- State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, Beijing, 100191, PR China
| | - Zhenming Liu
- State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, Beijing, 100191, PR China
| | - Wei Yang
- Department of Neurobiology, Key Laboratory of Medical Neurobiology of the Ministry of Health of China, Zhejiang University School of Medicine, Hangzhou, Zhejiang, 310058, PR China
| | - Peilin Yu
- Department of Toxicology, School of Public Health, Zhejiang University, Hangzhou, Zhejiang, 310058, PR China.
| | - Liangren Zhang
- State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, Beijing, 100191, PR China.
| | - Lihe Zhang
- State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, Beijing, 100191, PR China
| |
Collapse
|
21
|
Luo X, Li M, Zhan K, Yang W, Zhang L, Wang K, Yu P, Zhang L. Selective inhibition of TRPM2 channel by two novel synthesized ADPR analogues. Chem Biol Drug Des 2018; 91:552-566. [PMID: 29034580 PMCID: PMC5813235 DOI: 10.1111/cbdd.13119] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2017] [Revised: 07/04/2017] [Accepted: 08/21/2017] [Indexed: 01/02/2023]
Abstract
Transient receptor potential melastatin-2 (TRPM2) channel critical for monitoring internal body temperature is implicated in the pathological processes such as neurodegeneration. However, lacking selective and potent TRPM2 inhibitors impedes investigation and validation of the channel as a drug target. To discover novel and selective TRPM2 inhibitors, a series of adenosine 5'-diphosphoribose analogues were synthesized, and their activities and selectivity were evaluated. Whole-cell patch-clamp recordings were employed for screen and evaluation of synthesized compounds. Two compounds, 7i and 8a, were identified as TRPM2 inhibitors with IC50 of 5.7 and 5.4 μm, respectively. Both 7i and 8a inhibited TRPM2 current without affecting TRPM7, TRPM8, TRPV1 and TRPV3. These two TRPM2 inhibitors can serve as new pharmacological tools for further investigation and validation of TRPM2 channel as a drug target, and the summarized structure-activity relationship (SAR) may also provide insights into further improving existing inhibitors as potential lead compounds.
Collapse
Affiliation(s)
- Xiao Luo
- State Key Laboratory of Natural and Biomimetic DrugsPeking UniversityBeijingChina
| | - Meng Li
- Department of NeurobiologyNeuroscience Research InstitutePeking University Health Science CenterPeking University School of Pharmaceutical SciencesBeijingChina
| | - Kaiyu Zhan
- Department of NeurobiologyZhejiang University School of MedicineHangzhouZhejiangChina
| | - Wei Yang
- Department of NeurobiologyZhejiang University School of MedicineHangzhouZhejiangChina
| | - Lihe Zhang
- State Key Laboratory of Natural and Biomimetic DrugsPeking UniversityBeijingChina
| | - KeWei Wang
- Department of NeurobiologyNeuroscience Research InstitutePeking University Health Science CenterPeking University School of Pharmaceutical SciencesBeijingChina
- Department of PharmacologySchool of PharmacyQingdao UniversityQingdaoChina
| | - Peilin Yu
- Department of ToxicologySchool of Public HealthZhejiang UniversityHangzhouZhejiangChina
| | - Liangren Zhang
- State Key Laboratory of Natural and Biomimetic DrugsPeking UniversityBeijingChina
| |
Collapse
|
22
|
Almasi S, Kennedy BE, El-Aghil M, Sterea AM, Gujar S, Partida-Sánchez S, El Hiani Y. TRPM2 channel-mediated regulation of autophagy maintains mitochondrial function and promotes gastric cancer cell survival via the JNK-signaling pathway. J Biol Chem 2018; 293:3637-3650. [PMID: 29343514 DOI: 10.1074/jbc.m117.817635] [Citation(s) in RCA: 85] [Impact Index Per Article: 14.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2017] [Revised: 12/22/2017] [Indexed: 12/16/2022] Open
Abstract
A lack of effective treatment is one of the main factors contributing to gastric cancer-related death. Discovering effective targets and understanding their underlying anti-cancer mechanism are key to achieving the best response to treatment and to limiting side effects. Although recent studies have shown that the cation channel transient receptor potential melastatin-2 (TRPM2) is crucial for cancer cell survival, the exact mechanism remains unclear, limiting its therapeutic potential. Here, using molecular and functional assays, we investigated the role of TRPM2 in survival of gastric cancer cells. Our results indicated that TRPM2 knockdown in AGS and MKN-45 cells decreases cell proliferation and enhances apoptosis. We also observed that the TRPM2 knockdown impairs mitochondrial metabolism, indicated by a decrease in basal and maximal mitochondrial oxygen consumption rates and ATP production. These mitochondrial defects coincided with a decrease in autophagy and mitophagy, indicated by reduced levels of autophagy- and mitophagy-associated proteins (i.e. ATGs, LC3A/B II, and BNIP3). Moreover, we found that TRPM2 modulates autophagy through a c-Jun N-terminal kinase (JNK)-dependent and mechanistic target of rapamycin-independent pathway. We conclude that in the absence of TRPM2, down-regulation of the JNK-signaling pathway impairs autophagy, ultimately causing the accumulation of damaged mitochondria and death of gastric cancer cells. Of note, by inhibiting cell proliferation and promoting apoptosis, the TRPM2 down-regulation enhanced the efficacy of paclitaxel and doxorubicin in gastric cancer cells. Collectively, we provide compelling evidence that TRPM2 inhibition may benefit therapeutic approaches for managing gastric cancer.
Collapse
Affiliation(s)
| | | | | | - Andra M Sterea
- Physiology, Biophysics Faculty of Life Science, Dalhousie University, Halifax and
| | - Shashi Gujar
- Pathology.,Microbiology and Immunology, and.,the Centre for Innovative and Collaborative Health Services Research, Quality and System Performance, IWK Health Centre, Halifax, Nova Scotia B3H 4R2, Canada
| | - Santiago Partida-Sánchez
- Center for Microbial Pathogenesis, Research Institute at Nationwide Children's Hospital and.,the Department of Pediatrics, College of Medicine, Ohio State University, Columbus, Ohio 43205
| | - Yassine El Hiani
- Physiology, Biophysics Faculty of Life Science, Dalhousie University, Halifax and
| |
Collapse
|
23
|
Fliegert R, Watt JM, Schöbel A, Rozewitz MD, Moreau C, Kirchberger T, Thomas MP, Sick W, Araujo AC, Harneit A, Potter BVL, Guse AH. Ligand-induced activation of human TRPM2 requires the terminal ribose of ADPR and involves Arg1433 and Tyr1349. Biochem J 2017; 474:2159-2175. [PMID: 28515263 PMCID: PMC5473349 DOI: 10.1042/bcj20170091] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2017] [Revised: 05/09/2017] [Accepted: 05/15/2017] [Indexed: 12/23/2022]
Abstract
TRPM2 (transient receptor potential channel, subfamily melastatin, member 2) is a Ca2+-permeable non-selective cation channel activated by the binding of adenosine 5'-diphosphoribose (ADPR) to its cytoplasmic NUDT9H domain (NUDT9 homology domain). Activation of TRPM2 by ADPR downstream of oxidative stress has been implicated in the pathogenesis of many human diseases, rendering TRPM2 an attractive novel target for pharmacological intervention. However, the structural basis underlying this activation is largely unknown. Since ADP (adenosine 5'-diphosphate) alone did not activate or antagonize the channel, we used a chemical biology approach employing synthetic analogues to focus on the role of the ADPR terminal ribose. All novel ADPR derivatives modified in the terminal ribose, including that with the seemingly minor change of methylating the anomeric-OH, abolished agonist activity at TRPM2. Antagonist activity improved as the terminal substituent increasingly resembled the natural ribose, indicating that gating by ADPR might require specific interactions between hydroxyl groups of the terminal ribose and the NUDT9H domain. By mutating amino acid residues of the NUDT9H domain, predicted by modelling and docking to interact with the terminal ribose, we demonstrate that abrogating hydrogen bonding of the amino acids Arg1433 and Tyr1349 interferes with activation of the channel by ADPR. Taken together, using the complementary experimental approaches of chemical modification of the ligand and site-directed mutagenesis of TRPM2, we demonstrate that channel activation critically depends on hydrogen bonding of Arg1433 and Tyr1349 with the terminal ribose. Our findings allow for a more rational design of novel TRPM2 antagonists that may ultimately lead to compounds of therapeutic potential.
Collapse
Affiliation(s)
- Ralf Fliegert
- The Calcium Signalling Group, Department of Biochemistry and Molecular Cell Biology, University Medical Center Hamburg-Eppendorf, Martinistrasse 52, 20246 Hamburg, Germany
| | - Joanna M Watt
- Wolfson Laboratory of Medicinal Chemistry, Department of Pharmacy and Pharmacology, University of Bath, Bath BA2 7AY, U.K
- Medicinal Chemistry & Drug Discovery, Department of Pharmacology, University of Oxford, Mansfield Road, Oxford OX1 3QT, U.K
| | - Anja Schöbel
- The Calcium Signalling Group, Department of Biochemistry and Molecular Cell Biology, University Medical Center Hamburg-Eppendorf, Martinistrasse 52, 20246 Hamburg, Germany
| | - Monika D Rozewitz
- The Calcium Signalling Group, Department of Biochemistry and Molecular Cell Biology, University Medical Center Hamburg-Eppendorf, Martinistrasse 52, 20246 Hamburg, Germany
| | - Christelle Moreau
- Wolfson Laboratory of Medicinal Chemistry, Department of Pharmacy and Pharmacology, University of Bath, Bath BA2 7AY, U.K
| | - Tanja Kirchberger
- The Calcium Signalling Group, Department of Biochemistry and Molecular Cell Biology, University Medical Center Hamburg-Eppendorf, Martinistrasse 52, 20246 Hamburg, Germany
| | - Mark P Thomas
- Wolfson Laboratory of Medicinal Chemistry, Department of Pharmacy and Pharmacology, University of Bath, Bath BA2 7AY, U.K
| | - Wiebke Sick
- The Calcium Signalling Group, Department of Biochemistry and Molecular Cell Biology, University Medical Center Hamburg-Eppendorf, Martinistrasse 52, 20246 Hamburg, Germany
| | - Andrea C Araujo
- The Calcium Signalling Group, Department of Biochemistry and Molecular Cell Biology, University Medical Center Hamburg-Eppendorf, Martinistrasse 52, 20246 Hamburg, Germany
| | - Angelika Harneit
- The Calcium Signalling Group, Department of Biochemistry and Molecular Cell Biology, University Medical Center Hamburg-Eppendorf, Martinistrasse 52, 20246 Hamburg, Germany
| | - Barry V L Potter
- Wolfson Laboratory of Medicinal Chemistry, Department of Pharmacy and Pharmacology, University of Bath, Bath BA2 7AY, U.K
- Medicinal Chemistry & Drug Discovery, Department of Pharmacology, University of Oxford, Mansfield Road, Oxford OX1 3QT, U.K
| | - Andreas H Guse
- The Calcium Signalling Group, Department of Biochemistry and Molecular Cell Biology, University Medical Center Hamburg-Eppendorf, Martinistrasse 52, 20246 Hamburg, Germany
| |
Collapse
|
24
|
Yu P, Xue X, Zhang J, Hu X, Wu Y, Jiang LH, Jin H, Luo J, Zhang L, Liu Z, Yang W. Identification of the ADPR binding pocket in the NUDT9 homology domain of TRPM2. J Gen Physiol 2017; 149:219-235. [PMID: 28108595 PMCID: PMC5299621 DOI: 10.1085/jgp.201611675] [Citation(s) in RCA: 37] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2016] [Revised: 10/12/2016] [Accepted: 11/30/2016] [Indexed: 12/21/2022] Open
Abstract
Activation of the transient receptor potential melastatin 2 (TRPM2) channel occurs during the response to oxidative stress under physiological conditions as well as in pathological processes such as ischemia and diabetes. Accumulating evidence indicates that adenosine diphosphate ribose (ADPR) is the most important endogenous ligand of TRPM2. However, although it is known that ADPR binds to the NUDT9 homology (NUDT9-H) domain in the intracellular C-terminal region, the molecular mechanism underlying ADPR binding and activation of TRPM2 remains unknown. In this study, we generate a structural model of the NUDT9-H domain and identify the binding pocket for ADPR using induced docking and molecular dynamics simulation. We find a subset of 11 residues-H1346, T1347, T1349, L1379, G1389, S1391, E1409, D1431, R1433, L1484, and H1488-that are most likely to directly interact with ADPR. Results from mutagenesis and electrophysiology approaches support the predicted binding mechanism, indicating that ADPR binds tightly to the NUDT9-H domain, and suggest that the most significant interactions are the van der Waals forces with S1391 and L1484, polar solvation interaction with E1409, and electronic interactions (including π-π interactions) with H1346, T1347, Y1349, D1431, and H1488. These findings not only clarify the roles of a range of newly identified residues involved in ADPR binding in the TRPM2 channel, but also reveal the binding pocket for ADPR in the NUDT9-H domain, which should facilitate structure-based drug design for the TRPM2 channel.
Collapse
Affiliation(s)
- Peilin Yu
- Department of Neurobiology, Key Laboratory of Medical Neurobiology of the Ministry of Health of China, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310058, China.,Department of Toxicology, School of Public Health, Zhejiang University, Hangzhou, Zhejiang 310058, China
| | - Xiwen Xue
- State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, Beijing 100191, China
| | - Jianmin Zhang
- Department of Neurobiology, Key Laboratory of Medical Neurobiology of the Ministry of Health of China, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310058, China
| | - Xupang Hu
- Department of Neurobiology, Key Laboratory of Medical Neurobiology of the Ministry of Health of China, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310058, China
| | - Yan Wu
- Department of Neurobiology, Key Laboratory of Medical Neurobiology of the Ministry of Health of China, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310058, China
| | - Lin-Hua Jiang
- School of Biomedical Sciences, Faculty of Biological Sciences, University of Leeds, Leeds LS2 9JT, England, UK.,Department of Physiology and Neurobiology, Xinxiang Medical University, Henan 453003, China.,Sino-UK Brain Function Laboratory, Xinxiang Medical University, Henan 453003, China
| | - Hongwei Jin
- State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, Beijing 100191, China
| | - Jianhong Luo
- Department of Neurobiology, Key Laboratory of Medical Neurobiology of the Ministry of Health of China, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310058, China
| | - Liangren Zhang
- State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, Beijing 100191, China
| | - Zhenming Liu
- State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, Beijing 100191, China
| | - Wei Yang
- Department of Neurobiology, Key Laboratory of Medical Neurobiology of the Ministry of Health of China, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310058, China
| |
Collapse
|
25
|
Cruz-Rangel S, De Jesús-Pérez JJ, Aréchiga-Figueroa IA, Rodríguez-Menchaca AA, Pérez-Cornejo P, Hartzell HC, Arreola J. Extracellular protons enable activation of the calcium-dependent chloride channel TMEM16A. J Physiol 2017; 595:1515-1531. [PMID: 27859335 DOI: 10.1113/jp273111] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2016] [Accepted: 10/27/2016] [Indexed: 12/13/2022] Open
Abstract
KEY POINTS The calcium-activated chloride channel TMEM16A provides a pathway for chloride ion movements that are key in preventing polyspermy, allowing fluid secretion, controlling blood pressure, and enabling gastrointestinal activity. TMEM16A is opened by voltage-dependent calcium binding and regulated by permeant anions and intracellular protons. Here we show that a low proton concentration reduces TMEM16A activity while maximum activation is obtained when the external proton concentration is high. In addition, protonation conditions determine the open probability of TMEM16A without changing its calcium sensitivity. External glutamic acid 623 (E623) is key for TMEM16A's ability to respond to external protons. At physiological pH, E623 is un-protonated and TMEM16A is activated when intracellular calcium increases; however, under acidic conditions E623 is partially protonated and works synergistically with intracellular calcium to activate the channel. These findings are critical for understanding physiological and pathological processes that involve changes in pH and chloride flux via TMEM16A. ABSTRACT Transmembrane protein 16A (TMEM16A), also known as ANO1, the pore-forming subunit of a Ca2+ -dependent Cl- channel (CaCC), is activated by direct, voltage-dependent, binding of intracellular Ca2+ . Endogenous CaCCs are regulated by extracellular protons; however, the molecular basis of such regulation remains unidentified. Here, we evaluated the effects of different extracellular proton concentrations ([H+ ]o ) on mouse TMEM16A expressed in HEK-293 cells using whole-cell and inside-out patch-clamp recordings. We found that increasing the [H+ ]o from 10-10 to 10-5.5 m caused a progressive increase in the chloride current (ICl ) that is described by titration of a protonatable site with pK = 7.3. Protons regulate TMEM16A in a voltage-independent manner, regardless of channel state (open or closed), and without altering its apparent Ca2+ sensitivity. Noise analysis showed that protons regulate TMEM16A by tuning its open probability without modifying the single channel current. We found a robust reduction of the proton effect at high [Ca2+ ]i . To identify protonation targets we mutated all extracellular glutamate and histidine residues and 4 of 11 aspartates. Most mutants were sensitive to protons. However, mutation that substituted glutamic acid (E) for glutamine (Q) at amino acid position 623 (E623Q) displayed a titration curve shifted to the left relative to wild type channels and the ICl was nearly insensitive to proton concentrations between 10-5.5 and 10-9.0 m. Additionally, ICl of the mutant containing an aspartic acid (D) to asparagine (N) substitution at position 405 (D405N) mutant was partially inhibited by a proton concentration of 10-5.5 m, but 10-9.0 m produced the same effect as in wild type. Based on our findings we propose that external protons titrate glutamic acid 623, which enables voltage activation of TMEM16A at non-saturating [Ca2+ ]i .
Collapse
Affiliation(s)
- Silvia Cruz-Rangel
- Physics Institute, Universidad Autónoma de San Luis Potosí, Ave. Dr. Manuel Nava #6, San Luis Potosí, SLP, 78290, México
| | - José J De Jesús-Pérez
- Physics Institute, Universidad Autónoma de San Luis Potosí, Ave. Dr. Manuel Nava #6, San Luis Potosí, SLP, 78290, México
| | - Iván A Aréchiga-Figueroa
- CONACYT-Universidad Autónoma de San Luis Potosí School of Medicine, Ave. V. Carranza 2405, San Luis Potosí, SLP, 78290, México
| | - Aldo A Rodríguez-Menchaca
- Department of Physiology and Biophysics, Universidad Autónoma de San Luis Potosí School of Medicine, Ave. V. Carranza 2405, San Luis Potosí, SLP, 78290, México
| | - Patricia Pérez-Cornejo
- Department of Physiology and Biophysics, Universidad Autónoma de San Luis Potosí School of Medicine, Ave. V. Carranza 2405, San Luis Potosí, SLP, 78290, México
| | - H Criss Hartzell
- Department of Cell Biology, Emory University School of Medicine, Atlanta, GA, 30322, USA
| | - Jorge Arreola
- Physics Institute, Universidad Autónoma de San Luis Potosí, Ave. Dr. Manuel Nava #6, San Luis Potosí, SLP, 78290, México
| |
Collapse
|
26
|
Zhao LY, Xu WL, Xu ZQ, Qi C, Li Y, Cheng J, Liu LK, Wu YN, Gao J, Ye JH. The overexpressed functional transient receptor potential channel TRPM2 in oral squamous cell carcinoma. Sci Rep 2016; 6:38471. [PMID: 28008929 PMCID: PMC5180100 DOI: 10.1038/srep38471] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2016] [Accepted: 11/10/2016] [Indexed: 12/20/2022] Open
Abstract
TRPM2, one member of the transient receptor potential (TRP) protein super-family, is a Ca2+-permeable channel that is activated by oxidative stress and confers susceptibility to cell death. In the human tongue specimens of carcinoma and the tongue carcinoma SCC cell lines, we observed the enhanced expression of TRPM2. By means of the whole-cell electrophysiological recording, the ADPR-induced currents mediated by TRPM2 were recorded in cultured SCC9 cells. Moreover, after H2O2 treatment for 24 hours, the apoptotic number of SCC9 cells was significantly increased. However, the selectively knocked-down TRPM2 with the small interfering RNA technique inhibited the survival and migration of the SCC9 cancer cells, which was independent of the p53-p21 pathway, since the expression of p21 was enhanced after TRPM2 knockdown. Furthermore, the sub-cellular localization of TRPM2 was remarkably different between cancerous and non-cancerous cells. A significant amount of the TRPM2 proteins were located in the nuclei in cancer cells. All these data suggest that TRPM2 is essential for the survival and migration of SCC cancer cells and may be a potential target for the selective treatment of tongue cancer.
Collapse
Affiliation(s)
- Ling-Yan Zhao
- Jiangsu Key Laboratory of Oral Diseases and Department of Oral and maxillofacial surgery, Affiliated Hospital of Stomatology, Nanjing Medical University, 136 Hanzhong Road, Nanjing, 210029, China
| | - Wan-Lin Xu
- Jiangsu Key Laboratory of Oral Diseases and Department of Oral and maxillofacial surgery, Affiliated Hospital of Stomatology, Nanjing Medical University, 136 Hanzhong Road, Nanjing, 210029, China.,Shanghai Key Laboratory of Stomatology and Shanghai Research Institute of Stomatology, Department of Oral and Maxillofacial-Head and Neck Oncology, Ninth People's Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200011, China
| | - Zeng-Qi Xu
- Jiangsu Key Laboratory of Oral Diseases and Department of Oral and maxillofacial surgery, Affiliated Hospital of Stomatology, Nanjing Medical University, 136 Hanzhong Road, Nanjing, 210029, China
| | - Cui Qi
- Key Laboratory of Human Functional Genomics of Jiangsu, Department of Neurobiology, Nanjing Medical University, 101 Longmian Road, Nanjing, Jiangsu, 211166, China
| | - Yang Li
- Jiangsu Key Laboratory of Oral Diseases and Department of Oral and maxillofacial surgery, Affiliated Hospital of Stomatology, Nanjing Medical University, 136 Hanzhong Road, Nanjing, 210029, China
| | - Jie Cheng
- Jiangsu Key Laboratory of Oral Diseases and Department of Oral and maxillofacial surgery, Affiliated Hospital of Stomatology, Nanjing Medical University, 136 Hanzhong Road, Nanjing, 210029, China
| | - Lai-Kui Liu
- Jiangsu Key Laboratory of Oral Diseases and Department of Oral and maxillofacial surgery, Affiliated Hospital of Stomatology, Nanjing Medical University, 136 Hanzhong Road, Nanjing, 210029, China
| | - Yu-Nong Wu
- Jiangsu Key Laboratory of Oral Diseases and Department of Oral and maxillofacial surgery, Affiliated Hospital of Stomatology, Nanjing Medical University, 136 Hanzhong Road, Nanjing, 210029, China
| | - Jun Gao
- Key Laboratory of Human Functional Genomics of Jiangsu, Department of Neurobiology, Nanjing Medical University, 101 Longmian Road, Nanjing, Jiangsu, 211166, China
| | - Jin-Hai Ye
- Jiangsu Key Laboratory of Oral Diseases and Department of Oral and maxillofacial surgery, Affiliated Hospital of Stomatology, Nanjing Medical University, 136 Hanzhong Road, Nanjing, 210029, China
| |
Collapse
|
27
|
Detrimental or beneficial: the role of TRPM2 in ischemia/reperfusion injury. Acta Pharmacol Sin 2016; 37:4-12. [PMID: 26725732 DOI: 10.1038/aps.2015.141] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2015] [Accepted: 10/14/2015] [Indexed: 12/30/2022] Open
Abstract
Ischemia/reperfusion (I/R) injury is the main cause of tissue damage and dysfunction. I/R injury is characterized by Ca(2+) overload and production of reactive oxygen species (ROS), which play critical roles in the process of I/R injury to the brain, heart and kidney, but the underlying mechanisms are largely elusive. Recent evidence demonstrates that TRPM2, a Ca(2+)-permeable cationic channel and ROS sensor, is involved in I/R injury, but whether TRPM2 plays a protective or detrimental role in this process remains controversial. In this review, we discuss the recent progress in understanding the role of TRPM2 in reperfusion process after brain, heart and kidney ischemia and the potential of targeting TRPM2 for the development of therapeutic drugs to treat I/R injury.
Collapse
|
28
|
Tsujikawa H, Yu AS, Xie J, Yue Z, Yang W, He Y, Yue L. Identification of key amino acid residues responsible for internal and external pH sensitivity of Orai1/STIM1 channels. Sci Rep 2015; 5:16747. [PMID: 26576490 PMCID: PMC4649748 DOI: 10.1038/srep16747] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2014] [Accepted: 10/20/2015] [Indexed: 12/27/2022] Open
Abstract
Changes of intracellular and extracellular pH are involved in a variety of physiological and pathological processes, in which regulation of the Ca2+ release activated Ca2+ channel (ICRAC) by pH has been implicated. Ca2+ entry mediated by ICRAC has been shown to be regulated by acidic or alkaline pH. Whereas several amino acid residues have been shown to contribute to extracellular pH (pHo) sensitivity, the molecular mechanism for intracellular pH (pHi) sensitivity of Orai1/STIM1 is not fully understood. By investigating a series of mutations, we find that the previously identified residue E106 is responsible for pHo sensitivity when Ca2+ is the charge carrier. Unexpectedly, we identify that the residue E190 is responsible for pHo sensitivity when Na+ is the charge carrier. Furthermore, the intracellular mutant H155F markedly diminishes the response to acidic and alkaline pHi, suggesting that H155 is responsible for pHi sensitivity of Orai1/STIM1. Our results indicate that, whereas H155 is the intracellular pH sensor of Orai1/STIM1, the molecular mechanism of external pH sensitivity varies depending on the permeant cations. As changes of pH are involved in various physiological/pathological functions, Orai/STIM channels may be an important mediator for various physiological and pathological processes associated with acidosis and alkalinization.
Collapse
Affiliation(s)
- Hiroto Tsujikawa
- Calhoun Cardiology Center, Department of Cell Biology, University of Connecticut Health Center, Farmington, CT, USA
| | - Albert S Yu
- Calhoun Cardiology Center, Department of Cell Biology, University of Connecticut Health Center, Farmington, CT, USA
| | - Jia Xie
- Calhoun Cardiology Center, Department of Cell Biology, University of Connecticut Health Center, Farmington, CT, USA
| | - Zhichao Yue
- Calhoun Cardiology Center, Department of Cell Biology, University of Connecticut Health Center, Farmington, CT, USA
| | - Wenzhong Yang
- Calhoun Cardiology Center, Department of Cell Biology, University of Connecticut Health Center, Farmington, CT, USA
| | - Yanlin He
- Calhoun Cardiology Center, Department of Cell Biology, University of Connecticut Health Center, Farmington, CT, USA
| | - Lixia Yue
- Calhoun Cardiology Center, Department of Cell Biology, University of Connecticut Health Center, Farmington, CT, USA
| |
Collapse
|
29
|
PKD2L1/PKD1L3 channel complex with an alkali-activated mechanism and calcium-dependent inactivation. EUROPEAN BIOPHYSICS JOURNAL: EBJ 2015; 44:483-92. [PMID: 26066678 DOI: 10.1007/s00249-015-1040-y] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/10/2014] [Revised: 04/20/2015] [Accepted: 05/07/2015] [Indexed: 10/23/2022]
Abstract
Polycystic kidney disease-like (PKDL) genes that are expressed in sour taste cells have been proposed to be involved in the transduction of sourness by producing off-responses, which shows a large inward current after withdrawing the acid stimuli. However, the underlying mechanisms of off-responses are still unclear. Here, we demonstrate that an alkali-activated mechanism is responsible for eliciting off-responses, as evidenced by both experimental and theoretical analyses. In addition, we showed that the decaying phase of offset responses in PKD2L1/PKD1L3 channels was substantially accelerated by extracellular Ca(2+).
Collapse
|
30
|
Yu W, Jiang LH, Zheng Y, Hu X, Luo J, Yang W. Inactivation of TRPM2 channels by extracellular divalent copper. PLoS One 2014; 9:e112071. [PMID: 25386648 PMCID: PMC4227687 DOI: 10.1371/journal.pone.0112071] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2014] [Accepted: 10/11/2014] [Indexed: 12/02/2022] Open
Abstract
Cu2+ is an essential metal ion that plays a critical role in the regulation of a number of ion channels and receptors in addition to acting as a cofactor in a variety of enzymes. Here, we showed that human melastatin transient receptor potential 2 (hTRPM2) channel is sensitive to inhibition by extracellular Cu2+. Cu2+ at concentrations as low as 3 µM inhibited the hTRPM2 channel completely and irreversibly upon washing or using Cu2+ chelators, suggesting channel inactivation. The Cu2+-induced inactivation was similar when the channels conducted inward or outward currents, indicating the permeating ions had little effect on Cu2+-induced inactivation. Furthermore, Cu2+ had no effect on singe channel conductance. Alanine substitution by site-directed mutagenesis of His995 in the pore-forming region strongly attenuated Cu2+-induced channel inactivation, and mutation of several other pore residues to alanine altered the kinetics of channel inactivation by Cu2+. In addition, while introduction of the P1018L mutation is known to result in channel inactivation, exposure to Cu2+ accelerated the inactivation of this mutant channel. In contrast with the hTRPM2, the mouse TRPM2 (mTRPM2) channel, which contains glutamine at the position equivalent to His995, was insensitive to Cu2+. Replacement of His995 with glutamine in the hTRPM2 conferred loss of Cu2+-induced channel inactivation. Taken together, these results suggest that Cu2+ inactivates the hTRPM2 channel by interacting with the outer pore region. Our results also indicate that the amino acid residue difference in this region gives rise to species-dependent effect by Cu2+ on the human and mouse TRPM2 channels.
Collapse
Affiliation(s)
- Wenyue Yu
- Key Laboratory of Medical Neurobiology of the Ministry of Health of China, Zhejiang Province Key Laboratory of Neurobiology, Department of Neurobiology, School of Medicine, Zhejiang University, Hangzhou, Zhejiang Province, China
| | - Lin-Hua Jiang
- Department of Physiology and Neurobiology, Xinxiang Medical University, Xinxiang, Henan Province, China
- School of Biomedical Sciences, Faculty of Biological Sciences, University of Leeds, Leeds, United Kingdom
| | - Yang Zheng
- Key Laboratory of Medical Neurobiology of the Ministry of Health of China, Zhejiang Province Key Laboratory of Neurobiology, Department of Neurobiology, School of Medicine, Zhejiang University, Hangzhou, Zhejiang Province, China
| | - Xupang Hu
- Key Laboratory of Medical Neurobiology of the Ministry of Health of China, Zhejiang Province Key Laboratory of Neurobiology, Department of Neurobiology, School of Medicine, Zhejiang University, Hangzhou, Zhejiang Province, China
| | - Jianhong Luo
- Key Laboratory of Medical Neurobiology of the Ministry of Health of China, Zhejiang Province Key Laboratory of Neurobiology, Department of Neurobiology, School of Medicine, Zhejiang University, Hangzhou, Zhejiang Province, China
- * E-mail: (JHL); (WY)
| | - Wei Yang
- Key Laboratory of Medical Neurobiology of the Ministry of Health of China, Zhejiang Province Key Laboratory of Neurobiology, Department of Neurobiology, School of Medicine, Zhejiang University, Hangzhou, Zhejiang Province, China
- * E-mail: (JHL); (WY)
| |
Collapse
|
31
|
Abstract
TRPM2 (transient receptor potential melastatin 2) is a non-selective Ca2+-permeable cation channel activated by ADPR (adenosine diphosphoribose) and H2O2. It is widely expressed in mammalian cells and plays an important role in the regulation of various cell functions. However, the mechanisms of TRPM2 channel activation are not fully understood. Previously, we reported that TRPM2 channel activation is induced by high intracellular Cl- concentration. In the present study, we investigated the functional role of Lys1110 in the membrane-proximal C-terminal region by site-directed mutagenesis. Replacement of the positively charged amino acid lysine (Lys1110) with the neutrally charged amino acid asparagine (K1110N) or the negatively charged amino acid glutamic acid (K1110E) generated mutants that failed to induce an increase in free cytosolic calcium concentration ([Ca2+]i) not only by intracellular injection of Cl-, but also by H2O2 or ADPR. However, a mutant generated by replacing the lysine residue with a positively charged amino acid arginine (K1110R) displayed channel activity similar to wild-type TRPM2. Interestingly, in the K1107N/K1110N double-point mutant, the impaired function of the K1110N mutant in response to ADPR and H2O2, but not to Cl-, was recovered. There were no changes in protein expression, membrane trafficking and oligomerization of the mutant channels. The extent of [Ca2+]i increase by H2O2 in HEK (human embryonic kidney)-293 cells expressing TRPM2 mutants was well correlated with the degree of susceptibility to H2O2-induced cell death. These results display the crucial role of a positively charged amino acid residue at position 1110 for TRPM2 channel activity.
Collapse
|
32
|
Abstract
TRPM2 is the second member of the transient receptor potential melastatin-related (TRPM) family of cation channels. The protein is widely expressed including in the brain, immune system, endocrine cells, and endothelia. It embodies both ion channel functionality and enzymatic ADP-ribose (ADPr) hydrolase activity. TRPM2 is a Ca(2+)-permeable nonselective cation channel embedded in the plasma membrane and/or lysosomal compartments that is primarily activated in a synergistic fashion by intracellular ADP-ribose (ADPr) and Ca(2+). It is also activated by reactive oxygen and nitrogen species (ROS/NOS) and enhanced by additional factors, such as cyclic ADPr and NAADP, while inhibited by permeating protons (acidic pH) and adenosine monophosphate (AMP). Activation of TRPM2 leads to increases in intracellular Ca(2+) levels, which can serve signaling roles in inflammatory and secretory cells through release of vesicular mediators (e.g., cytokines, neurotransmitters, insulin) and in extreme cases can induce apoptotic and necrotic cell death under oxidative stress.
Collapse
Affiliation(s)
- Malika Faouzi
- Center for Biomedical Research, The Queen's Medical Center, 1301 Punchbowl Street, Honolulu, HI, 96813, USA,
| | | |
Collapse
|
33
|
Lockwood TD. Lysosomal metal, redox and proton cycles influencing the CysHis cathepsin reaction. Metallomics 2013; 5:110-24. [PMID: 23302864 DOI: 10.1039/c2mt20156a] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
Abstract
In the 1930's pioneers discovered that maximal autolysis in tissue homogenates requires metal chelator, sulfhydryl reducing agent and acid pH. However, metals, reducing equivalents and protons (MR&P) have been overlooked as combined catalytic controls. Three categories of lysosomal machinery drive three distinguishable cycles importing and exporting MR&P. Zn(2+) preemptively inhibits CysHis catalysis under otherwise optimal protonation and reduction. Protein-bound cell Zn(2+) concentration is 200-2000 times the non-sequestered inhibitory concentration. Following autophagy, lysosomal proteolysis liberates much inhibitory Zn(2+). The vacuolar proton pump is the driving force for Zn(2+) export, as well as protonation of the peptidolytic mechanism. Other machinery of lysosomal cycles includes proton-driven Zn(2+) exporters (e.g. SLC11A1), Zn(2+) channels (e.g. TRPML-1), lysosomal thiol reductase, etc. The CysHis dyad is a sensor of the vacuolar environment of MR&P, an integrator of these simultaneous variables, and a catalytic responder. Rate-determination can shift between autophagic substrate acquisition (swallowing) and substrate degradation (digesting). Zn(2+) recycling from degraded proteins to new proteins is a fourth cycle that might pace lysosomal function under some conditions. Heritable insufficient or excess functions of CysHis cathepsins are associated with dysfunctional inflammation and immunity/auto-immunity, including diabetic pathogenesis.
Collapse
Affiliation(s)
- Thomas D Lockwood
- Dept. of Pharmacology, School of Medicine, Wright State University, Dayton, Ohio 45435, USA.
| |
Collapse
|
34
|
Sumoza-Toledo A, Fleig A, Penner R. TRPM2 channels are not required for acute airway inflammation in OVA-induced severe allergic asthma in mice. JOURNAL OF INFLAMMATION-LONDON 2013; 10:19. [PMID: 23631390 PMCID: PMC3648402 DOI: 10.1186/1476-9255-10-19] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/19/2013] [Accepted: 04/23/2013] [Indexed: 11/23/2022]
Abstract
Background Airway inflammation and asthma have been linked to oxidative stress and the melastatin-related transient receptor potential cation channel, member 2 (TRPM2), which can be activated by reactive oxygen species (ROS), has emerged as a potential therapeutic target for inflammatory diseases. Objective Using TRPM2 deficient (TRPM2-/-) mice, we investigated whether the TRPM2 ion channel, which mediates calcium (Ca2+) influx and lysosomal Ca2+ release, plays a role in the pathophysiology of severe allergic asthma in mouse. Methods Severe allergic asthma was initiated in wild type (WT) and TRPM2-/- mice by repeated sensitization with ovalbumin (OVA)/aluminum hydroxide on Days 0, 7 and 14, followed by intranasal challenge on Days 21, 22 and 23. Mice were investigated for the presence of airway responsiveness, airway inflammation, production of allergen-specific antibodies, cytokine response and lung pathology. Results The absence of TRPM2 channels has no obvious effect on major etiologic markers of severe allergic asthma in this mouse model. Neither airway resistance nor mucus production are affected in TRPM2-/- mice. TRPM2 channel ablation also does not alter airway inflammation or immunocyte infiltration and does not affect antibody response or cytokine levels. Conclusions TRPM2 is not required for airway inflammation in OVA-induced severe allergic asthma in mice. Accordingly, TRPM2 might not be a suitable therapeutic target for airway inflammation caused by allergens in humans.
Collapse
Affiliation(s)
- Adriana Sumoza-Toledo
- Laboratory for Cellular and Molecular Signaling, Center for Biomedical Research at The Queen's Medical Center and John A, Burns School of Medicine, University of Hawai'i, 1301 Punchbowl St, Honolulu, HI, 96813, USA.
| | | | | |
Collapse
|
35
|
Zou J, Ainscough JF, Yang W, Sedo A, Yu SP, Mei ZZ, Sivaprasadarao A, Beech DJ, Jiang LH. A differential role of macrophage TRPM2 channels in Ca²⁺ signaling and cell death in early responses to H₂O₂. Am J Physiol Cell Physiol 2013; 305:C61-9. [PMID: 23596170 DOI: 10.1152/ajpcell.00390.2012] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/30/2023]
Abstract
Reactive oxygen species such as H₂O₂ elevates the cytosolic Ca²⁺ concentration ([Ca²⁺]c) and causes cell death via poly(ADPR) polymerase (PARP) activation, which also represents the primary mechanism by which H₂O₂ activate the transient receptor potential melastatin-related 2 (TRPM2) channel as a Ca²⁺-permeable channel present in the plasma membrane or an intracellular Ca²⁺-release channel. The present study aimed to define the contribution and mechanisms of the TRPM2 channels in macrophage cells in mediating Ca²⁺ signaling and cell death during initial response to H₂O₂, using mouse peritoneal macrophage, RAW264.7, and differentiated THP-1 cells. H₂O₂ evoked robust increases in the [Ca²⁺]c, and such Ca²⁺ responses were significantly greater at body temperature than room temperature. H₂O₂-induced Ca²⁺ responses were strongly inhibited by pretreatment with PJ-34, a PARP inhibitor, and largely prevented by removal of extracellular Ca²⁺. Furthermore, H₂O₂-induced increases in the [Ca²⁺]c were completely abolished in macrophage cells isolated from trpm2-/- mice. H₂O₂ reduced macrophage cell viability in a duration- and concentration-dependent manner. H₂O₂-induced cell death was significantly attenuated by pretreatment with PJ-34 and TRPM2 channel deficiency but remained significant and persistent. Taken together, these results show that the TRPM2 channel in macrophage cells functions as a cell surface Ca²⁺-permeable channel that mediates Ca²⁺ influx and constitutes the principal Ca²⁺ signaling mechanism but has a limited, albeit significant, role in cell death during early exposure to H₂O₂.
Collapse
Affiliation(s)
- Jie Zou
- School of Biomedical Sciences, Faculty of Biological Sciences, University of Leeds, Leeds, United Kingdom
| | | | | | | | | | | | | | | | | |
Collapse
|
36
|
Site-directed mutagenesis to study the structure-function relationships of ion channels. Methods Mol Biol 2013; 998:257-66. [PMID: 23529436 DOI: 10.1007/978-1-62703-351-0_20] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Ion channels mediate a wide variety of physiological processes by forming small pores across the membranes that allow regulated flow of ions into or out of the cell. The primary linear sequences of ion channel proteins, like any proteins, are composed by 20 different amino acids, each of which is determined by specific triplet codon in their genes. Site-directed mutagenesis is a widely used molecular biology method to change the triplet in the coding sequence and thereby the amino acid residue in the protein sequence. Functional characterization of the ion channels carrying point mutations allows us to interrogate the structure-function relationships of the ion channels. Here, we will describe the site-directed mutagenesis procedures, in which the wide-type cDNA or plasmid is used as a template to synthesize the complementary mutation-containing DNAs from two mutagenic primers in the polymerase chain reaction.
Collapse
|
37
|
Abstract
Transient potential receptor melastatin-2 (TRPM2) is a non-selective Ca2+-permeable cation channel of the TRPM channel subfamily and is mainly activated by intracellular adenosine diphosphate ribose (ADPR). Here we synthesized a 1-(2-nitrophenyl)ethyl caged ADPR (NPE-ADPR) and found that uncaging of NPE-ADPR efficiently stimulated Ca2+, Mg2+, and Zn2+ influx in a concentration-dependent manner in intact human Jurkat T-lymphocytes. The cation influx was inhibited by inhibitors or knockdown of TRPM2. Likewise, uncaging of NPE-ADPR markedly induced cation entry in HEK 293 cells that overexpress TRPM2. As expected, high temperature increased the ability of the photolyzed NPE-ADPR to induce cation entry, whereas acidic pH inhibited. Moreover, the absence of extracellular Ca2+ significantly inhibited Mg2+ and Zn2+ influx after uncaging NPE-ADPR. On the other hand, the absence of extracellular Na+ or Mg2+ had no effect on photolyzed NPE-ADPR induced Ca2+ entry. Taken together, our results indicated that NPE-ADPR is a cell permeable ADPR analogue that is useful for studying TRPM2-mediated cation entry in intact cells.
Collapse
|
38
|
Pore collapse underlies irreversible inactivation of TRPM2 cation channel currents. Proc Natl Acad Sci U S A 2012; 109:13440-5. [PMID: 22847436 DOI: 10.1073/pnas.1204702109] [Citation(s) in RCA: 53] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
The Ca(2+)-permeable cation channel transient receptor potential melastatin 2 (TRPM2) plays a key role in pathogen-evoked phagocyte activation, postischemic neuronal apoptosis, and glucose-evoked insulin secretion, by linking these cellular responses to oxidative stress. TRPM2 channels are coactivated by binding of intracellular ADP ribose and Ca(2+) to distinct cytosolically accessible sites on the channels. These ligands likely regulate the activation gate, conserved in the voltage-gated cation channel superfamily, that comprises a helix bundle formed by the intracellular ends of transmembrane helix six of each subunit. For several K(+) and TRPM family channels, activation gate opening requires the presence of phosphatidylinositol-bisphosphate (PIP(2)) in the inner membrane leaflet. Most TRPM family channels inactivate upon prolonged stimulation in inside-out patches; this "rundown" is due to PIP(2) depletion. TRPM2 currents also run down within minutes, but the molecular mechanism of this process is unknown. Here we report that high-affinity PIP(2) binding regulates Ca(2+) sensitivity of TRPM2 activation. Nevertheless, TRPM2 inactivation is not due to PIP(2) depletion; rather, it is state dependent, sensitive to permeating ions, and can be completely prevented by mutations in the extracellular selectivity filter. Introduction of two negative charges plus a single-residue insertion, to mimic the filter sequence of TRPM5, results in TRPM2 channels that maintain unabated maximal activity for over 1 h, and display altered permeation properties but intact ADP ribose/Ca(2+)-dependent gating. Thus, upon prolonged stimulation, the TRPM2 selectivity filter undergoes a conformational change reminiscent of that accompanying C-type inactivation of voltage-gated K(+) channels. The noninactivating TRPM2 variant will be invaluable for gating studies.
Collapse
|
39
|
Kajma A, Szewczyk A. A new pH-sensitive rectifying potassium channel in mitochondria from the embryonic rat hippocampus. BIOCHIMICA ET BIOPHYSICA ACTA-BIOENERGETICS 2012; 1817:1867-78. [PMID: 22406520 DOI: 10.1016/j.bbabio.2012.02.029] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/08/2011] [Revised: 02/22/2012] [Accepted: 02/24/2012] [Indexed: 12/13/2022]
Abstract
Patch-clamp single-channel studies on mitochondria isolated from embryonic rat hippocampus revealed the presence of two different potassium ion channels: a large-conductance (288±4pS) calcium-activated potassium channel and second potassium channel with outwardly rectifying activity under symmetric conditions (150/150mM KCl). At positive voltages, this channel displayed a conductance of 67.84pS and a strong voltage dependence at holding potentials from -80mV to +80mV. The open probability was higher at positive than at negative voltages. Patch-clamp studies at the mitoplast-attached mode showed that the channel was not sensitive to activators and inhibitors of mitochondrial potassium channels but was regulated by pH. Moreover, we demonstrated that the channel activity was not affected by the application of lidocaine, an inhibitor of two-pore domain potassium channels, or by tertiapin, an inhibitor of inwardly rectifying potassium channels. In summary, based on the single-channel recordings, we characterised for the first time mitochondrial pH-sensitive ion channel that is selective for cations, permeable to potassium ions, displays voltage sensitivity and does not correspond to any previously described potassium ion channels in the inner mitochondrial membrane. This article is part of a Special Issue entitled: 17th European Bioenergetics Conference (EBEC 2012).
Collapse
Affiliation(s)
- Anna Kajma
- Polish Academy of Sciences, Warsaw, Poland
| | | |
Collapse
|
40
|
Sumoza-Toledo A, Lange I, Cortado H, Bhagat H, Mori Y, Fleig A, Penner R, Partida-Sánchez S. Dendritic cell maturation and chemotaxis is regulated by TRPM2-mediated lysosomal Ca2+ release. FASEB J 2011; 25:3529-42. [PMID: 21753080 DOI: 10.1096/fj.10-178483] [Citation(s) in RCA: 111] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
Chemokines induce calcium (Ca(2+)) signaling and chemotaxis in dendritic cells (DCs), but the molecular players involved in shaping intracellular Ca(2+) changes remain to be characterized. Using siRNA and knockout mice, we show that in addition to inositol 1,4,5-trisphosphate (IP(3))-mediated Ca(2+) release and store-operated Ca(2+) entry (SOCE), the transient receptor potential melastatin 2 (TRPM2) channel contributes to Ca(2+) release but not Ca(2+) influx in mouse DCs. Consistent with these findings, TRPM2 expression in DCs is restricted to endolysosomal vesicles, whereas in neutrophils, the channel localizes to the plasma membrane. TRPM2-deficient DCs show impaired maturation and severely compromised chemokine-activated directional migration as well as bacterial-induced DC trafficking to the draining lymph nodes. Defective DC chemotaxis is due to perturbed chemokine-receptor-initiated Ca(2+) signaling mechanisms, which include suppression of TRPM2-mediated Ca(2+) release and secondary modification of SOCE. DCs deficient in both TRPM2 and IP(3) receptor signaling lose their ability to perform chemotaxis entirely. These results highlight TRPM2 as a key player regulating DC chemotaxis through its function as Ca(2+) release channel and confirm ADP-ribose as a novel second messenger for intracellular Ca(2+) mobilization.
Collapse
Affiliation(s)
- Adriana Sumoza-Toledo
- Laboratory of Cell and Molecular Signaling, Center for Biomedical Research, Queen's Medical Center, Honolulu, Hawaii, USA
| | | | | | | | | | | | | | | |
Collapse
|
41
|
Yang W, Manna PT, Zou J, Luo J, Beech DJ, Sivaprasadarao A, Jiang LH. Zinc inactivates melastatin transient receptor potential 2 channels via the outer pore. J Biol Chem 2011; 286:23789-98. [PMID: 21602277 PMCID: PMC3129160 DOI: 10.1074/jbc.m111.247478] [Citation(s) in RCA: 37] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2011] [Revised: 05/19/2011] [Indexed: 10/18/2022] Open
Abstract
Zinc ion (Zn(2+)) is an endogenous allosteric modulator that regulates the activity of a wide variety of ion channels in a reversible and concentration-dependent fashion. Here we used patch clamp recording to study the effects of Zn(2+) on the melastatin transient receptor potential 2 (TRPM2) channel. Zn(2+) inhibited the human (h) TRPM2 channel currents, and the steady-state inhibition was largely not reversed upon washout and concentration-independent in the range of 30-1000 μM, suggesting that Zn(2+) induces channel inactivation. Zn(2+) inactivated the channels fully when they conducted inward currents, but only by half when they passed outward currents, indicating profound influence of the permeant ion on Zn(2+) inactivation. Alanine substitution scanning mutagenesis of 20 Zn(2+)-interacting candidate residues in the outer pore region of the hTRPM2 channel showed that mutation of Lys(952) in the extracellular end of the fifth transmembrane segment and Asp(1002) in the large turret strongly attenuated or abolished Zn(2+) inactivation, and mutation of several other residues dramatically changed the inactivation kinetics. The mouse (m) TRPM2 channels were also inactivated by Zn(2+), but the kinetics were remarkably slower. Reciprocal mutation of His(995) in the hTRPM2 channel and the equivalent Gln(992) in the mTRPM2 channel completely swapped the kinetics, but no such opposing effects resulted from exchanging another pair of species-specific residues, Arg(961)/Ser(958). We conclude from these results that Zn(2+) inactivates the TRPM2 channels and that residues in the outer pore are critical determinants of the inactivation.
Collapse
Affiliation(s)
- Wei Yang
- From the Institute of Membrane and Systems Biology, Faculty of Biological Sciences, University of Leeds, Leeds LS2 9JT, United Kingdom and
- the Department of Neurobiology, Zhejiang University School of Medicine, Zhejiang 310058, China
| | - Paul T. Manna
- From the Institute of Membrane and Systems Biology, Faculty of Biological Sciences, University of Leeds, Leeds LS2 9JT, United Kingdom and
| | - Jie Zou
- From the Institute of Membrane and Systems Biology, Faculty of Biological Sciences, University of Leeds, Leeds LS2 9JT, United Kingdom and
| | - Jianhong Luo
- the Department of Neurobiology, Zhejiang University School of Medicine, Zhejiang 310058, China
| | - David J. Beech
- From the Institute of Membrane and Systems Biology, Faculty of Biological Sciences, University of Leeds, Leeds LS2 9JT, United Kingdom and
| | - Asipu Sivaprasadarao
- From the Institute of Membrane and Systems Biology, Faculty of Biological Sciences, University of Leeds, Leeds LS2 9JT, United Kingdom and
| | - Lin-Hua Jiang
- From the Institute of Membrane and Systems Biology, Faculty of Biological Sciences, University of Leeds, Leeds LS2 9JT, United Kingdom and
| |
Collapse
|
42
|
A residue in the TRPM2 channel outer pore is crucial in determining species-dependent sensitivity to extracellular acidic pH. Pflugers Arch 2011; 462:293-302. [DOI: 10.1007/s00424-011-0957-y] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2011] [Revised: 03/07/2011] [Accepted: 03/09/2011] [Indexed: 10/18/2022]
|
43
|
Abstract
The transient potential receptor melastatin-2 (TRPM2) channel has emerged as an important Ca(2+) signalling mechanism in a variety of cells, contributing to cellular functions that include cytokine production, insulin release, cell motility and cell death. Its ability to respond to reactive oxygen species has made TRPM2 a potential therapeutic target for chronic inflammation, neurodegenerative diseases, and oxidative stress-related pathologies. TRPM2 is a non-selective, calcium (Ca(2+))-permeable cation channel of the melastatin-related transient receptor potential (TRPM) ion channel subfamily. It is activated by intracellular adenosine diphosphate ribose (ADPR) through a diphosphoribose hydrolase domain in its C-terminus and regulated through a variety of factors, including synergistic facilitation by [Ca(2+)](i), cyclic ADPR, H(2)O(2), NAADP, and negative feedback regulation by AMP and permeating protons (pH). In addition to its role mediating Ca(2+) influx into the cells, TRPM2 can also function as a lysosomal Ca(2+) release channel, contributing to cell death. The physiological and pathophysiological context of ROS-mediated events makes TRPM2 a promising target for the development of therapeutic tools of inflammatory and degenerative diseases.
Collapse
Affiliation(s)
- Adriana Sumoza-Toledo
- Center for Biomedical Research, The Queen's Medical Center, University of Hawaii, 1301 Punchbowl Street - UHT 8, HI 96813, USA
| | | |
Collapse
|