1
|
Lu T, Zhang Y, Li J, Dai M, Liu H, Zhu H, Fu S, Dong X, Sun F, Lin H, Zhang X, Yang W, Yu P, Zou H. Indolizine Derivatives Inhibit TRPM2 and Protect against Ischemic Brain Injury with an Extended Treatment Window. J Med Chem 2025; 68:7642-7661. [PMID: 40168472 DOI: 10.1021/acs.jmedchem.5c00186] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/03/2025]
Abstract
Ischemic stroke, a major cause of disability and death worldwide, lacks effective treatments due to the complexity of brain ischemia/reperfusion (I/R) injury. The transient receptor potential melastatin 2 (TRPM2) channel is a promising therapeutic target. In this study, an extracellular TRPM2 inhibitor A1 with an indolizine scaffold was identified through chemical library screening. Four series of indolizine derivatives were synthesized, yielding four compounds with TRPM2 inhibitory activity comparable to or superior to A1, as confirmed by calcium fluorescence and electrophysiological assays. These compounds demonstrated significant neuroprotective effects in vitro. Among them, D10 showed robust efficacy in reducing cerebral infarction in a transient middle cerebral artery occlusion (tMCAO) model, surpassing edaravone. When administered 24 h postreperfusion and continued for 7 days, D10 exhibited sustained in vivo antistroke activity and improved survival rates compared to edaravone and vehicle controls. D10 represents a promising lead compound for ischemic stroke therapy.
Collapse
Affiliation(s)
- Tinghao Lu
- College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, PR China
| | - Yi Zhang
- Zhejiang University School of Medicine, Hangzhou 310058, PR China
| | - Jinbiao Li
- College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, PR China
| | - MeiJie Dai
- College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, PR China
- Institute of Pharmacology and Toxicology, College of Pharmaceutical Sciences, Key Laboratory of Medical Neurobiology of the Ministry of Health of China, Zhejiang University, Hangzhou 310058, PR China
| | - Huan Liu
- Zhejiang University School of Medicine, Hangzhou 310058, PR China
- Department of Biophysics, and Department of Neurosurgery of the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310058, PR China
| | - Huajian Zhu
- College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, PR China
| | - Shaozi Fu
- Zhejiang University School of Medicine, Hangzhou 310058, PR China
- Department of Thoracic Surgery, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310058, PR China
| | - Xianhao Dong
- College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, PR China
| | - Fenghao Sun
- Zhongshan hospital, Fudan University, Shanghai 200000, PR China
| | - Hongwei Lin
- Zhejiang University School of Medicine, Hangzhou 310058, PR China
- Department of Neurosurgery, Sir Run Run Shaw Hospital, Affiliated with the Zhejiang University School of Medicine, Hangzhou, Zhejiang 310058, PR China
| | - Xiangnan Zhang
- College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, PR China
- Institute of Pharmacology and Toxicology, State Key Laboratory of Advanced Drug Delivery and Release Systems, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
| | - Wei Yang
- Zhejiang University School of Medicine, Hangzhou 310058, PR China
- Department of Biophysics, and Department of Neurosurgery of the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310058, PR China
| | - Peilin Yu
- Zhejiang University School of Medicine, Hangzhou 310058, PR China
- Department of Toxicology, and Department of Medical Oncology of the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310058, PR China
| | - Hongbin Zou
- College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, PR China
| |
Collapse
|
2
|
Ferreira AFF, Ulrich H, Mori Y, Feng ZP, Sun HS, Britto LR. Deletion of the Transient Receptor Potential Melastatin 2 Gene Mitigates the 6-Hydroxydopamine-Induced Parkinson's Disease-Like Pathology. Mol Neurobiol 2025; 62:5333-5346. [PMID: 39541072 DOI: 10.1007/s12035-024-04611-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2024] [Accepted: 11/04/2024] [Indexed: 11/16/2024]
Abstract
Pharmacological inhibition of the transient receptor potential melastatin 2 (TRPM2), an oxidative stress-activated calcium channel, was previously reported to be protective in Parkinson's disease (PD). However, the inhibitors used were not TRPM2 specific, so the involvement of this channel in PD remains unclear. Here, for the first time, Trpm2 partial (+ / -) and complete (- / -) knockout mice underwent stereotaxic surgery for PD induction. Six-hydroxydopamine was injected in the right striatum. On days 3 and 6, motor behavior tests (cylinder, apomorphine, and pole test) were performed. On day 7, brains were collected for dopaminergic neuron immunostaining. Our results showed that Trpm2 + / - male and female mice had reduced motor impairment and dopaminergic neuron death after PD induction. In addition, Trpm2 - / - male and female mice showed absent or lesser motor deficit and the dopaminergic neuronal loss was no longer observed. These findings suggest that TRPM2 is involved in the PD-like pathology and that targeting TRPM2 may possibly represent a potential neuroprotective strategy for PD.
Collapse
Affiliation(s)
- Ana Flavia F Ferreira
- Department of Physiology and Biophysics, Institute of Biomedical Sciences, University of São Paulo, São Paulo, Brazil.
- Department of Physiology, Temerty Faculty of Medicine, University of Toronto, Toronto, Canada.
| | - Henning Ulrich
- Department of Biochemistry, Institute of Chemistry, University of São Paulo, São Paulo, Brazil
| | - Yasuo Mori
- Department of Synthetic Chemistry and Biological Chemistry, Graduate School of Engineering, Kyoto University, Katsura Campus, Nishikyo-Ku, Japan
| | - Zhong-Ping Feng
- Department of Physiology, Temerty Faculty of Medicine, University of Toronto, Toronto, Canada
| | - Hong-Shuo Sun
- Department of Physiology, Temerty Faculty of Medicine, University of Toronto, Toronto, Canada
- Department of Surgery, Temerty Faculty of Medicine, University of Toronto, Toronto, Canada
| | - Luiz Roberto Britto
- Department of Physiology and Biophysics, Institute of Biomedical Sciences, University of São Paulo, São Paulo, Brazil
| |
Collapse
|
3
|
Xua R, Bai Y, Liang J, Wang N, He Y, Meng L, Ming D. The effects of different challenge-level balance tasks on stroke cortical responses and balance assessment using EEG. IEEE Trans Neural Syst Rehabil Eng 2025; PP:640-652. [PMID: 40030936 DOI: 10.1109/tnsre.2025.3529890] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/05/2025]
Abstract
Previous studies have validated that different balance tasks induce different cortical responses, which are key indexes of balance assessment. Assessing balance is crucial for stroke survivors to prevent falls and improve rehabilitation outcomes. However, it was unclear whether these tasks may affect the balance assessment, particularly regarding the relationship between task difficulty and the corresponding cortical responses involved in balance control. Therefore, we sought to explore the effects of different challenge-level balance tasks on balance assessment. Eighteen participants with stroke and thirteen healthy individuals were recruited in this study. The EEG was collected during sitting, standing and perturbation tasks. The pairwise-derived Brain Symmetry Index (pdBSI), and Granger Causality (GC) were analyzed with a two-way (task ×group) RMANOVA. Finally, a multiple linear regression analysis was applied to predict the BBS score with the above parameters. We found a significant interaction effect on pdBSI and GC. In the frontal lobe, participants with stroke exhibited significantly higher pdBSI (standing: p=0.042, perturbation: p=0.013) and lower GC (standing: p<0.001, perturbation: p=0.028) compared to healthy controls. Similarly, in the parietal lobe, stroke survivors showed markedly higher pdBSI (standing: p = 0.006, perturbation: p=0.012) and lower GC (standing: p=0.030, perturbation: p=0.011). Finally, The Berg Balance Scale (BBS) scores could be reliably predicted using parietal BSI and frontal GC metrics recorded during standing (p<0.001, adjusted R²=0.938) and perturbation tasks (p=0.001, adjusted R²=0.644). It was discovered that the more challenging balance tasks better revealed the difference in the power distribution and the directional functional connection between groups. The pdBSI and GC during standing and perturbation tasks, could be used as biomarkers for stroke balance assessment.
Collapse
|
4
|
Li YS, Ren HC, Li H, Xing M, Cao JH. From oxidative stress to metabolic dysfunction: The role of TRPM2. Int J Biol Macromol 2025; 284:138081. [PMID: 39603285 DOI: 10.1016/j.ijbiomac.2024.138081] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2024] [Revised: 11/14/2024] [Accepted: 11/24/2024] [Indexed: 11/29/2024]
Abstract
Metabolic syndromes including atherosclerosis, diabetes, obesity, and hypertension are increasingly prevalent worldwide. The disorders are the primary attributes of oxidative stress and inflammation. The transient receptor potential M2 (TRPM2) channel is a pivotal mediator linking oxidative stress to metabolic dysfunction. TRPM2, a non-selective cation channel activated by reactive oxygen species (ROS) and adenosine diphosphate ribose (ADPR), regulates calcium influx, inflammation, and cell death across various tissues. This review explores the structural and activation mechanisms of TRPM2, emphasizing its significance in metabolic diseases. Elevated levels of TRPM2 play a vital role in the disease progression by influencing physiological and cellular processes such as endothelial dysfunction, immune cell activation, and mitochondrial impairment. In conditions such as atherosclerosis, ischemic stroke, diabetes, obesity, and hypertension; TRPM2 exacerbates oxidative damage, amplifies inflammatory responses, and disrupts metabolic homeostasis. Recent research highlights the potential of TRPM2 as a therapeutic target, developing specified inhibitors. This review underscores the multifaceted role of TRPM2 in metabolic disorders and its promise as a target for therapeutic interventions.
Collapse
Affiliation(s)
- Ying-Shuang Li
- Intravenous Drug Administration Center, Department of Pharmacy, Qingdao Third People's Hospital affiliated with Qingdao University, Qingdao, Shandong 266041, PR China
| | - Hua-Cheng Ren
- Intravenous Drug Administration Center, Department of Pharmacy, Qingdao Third People's Hospital affiliated with Qingdao University, Qingdao, Shandong 266041, PR China
| | - Hui Li
- Intravenous Drug Administration Center, Department of Pharmacy, Qingdao Third People's Hospital affiliated with Qingdao University, Qingdao, Shandong 266041, PR China
| | - Man Xing
- Intravenous Drug Administration Center, Department of Pharmacy, Qingdao Third People's Hospital affiliated with Qingdao University, Qingdao, Shandong 266041, PR China
| | - Jian-Hua Cao
- Intravenous Drug Administration Center, Department of Pharmacy, Qingdao Third People's Hospital affiliated with Qingdao University, Qingdao, Shandong 266041, PR China.
| |
Collapse
|
5
|
Hu F, Lin C. TRPM2 knockdown attenuates myocardial apoptosis and promotes autophagy in HFD/STZ-induced diabetic mice via regulating the MEK/ERK and mTORC1 signaling pathway. Mol Cell Biochem 2024; 479:3307-3328. [PMID: 38308007 PMCID: PMC11511773 DOI: 10.1007/s11010-024-04926-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2023] [Accepted: 01/05/2024] [Indexed: 02/04/2024]
Abstract
Diabetic cardiomyopathy (DCM) is a major complication of diabetes. Transient receptor potential melastatin 2 (TRPM2) activity increases in diabetic oxidative stress state, and it is involved in myocardial damage and repair. We explore the protective effect of TRPM2 knockdown on the progression of DCM. A type 2 diabetes animal model was established in C57BL/6N mice by long-term high-fat diet (HFD) feeding combined with a single injection of 100-mg/kg streptozotocin (STZ). Genetic knockdown of TRPM2 in heart was accomplished by the intravenous injection via the tail vein of adeno-associated virus type 9 carrying TRPM2 shRNA. Neonatal rat ventricular myocytes was exposed to 45 mM of high-glucose (HG) stimulation for 72 h in vitro to mimic the in vivo conditions. Western blot, real-time quantitative PCR (RT-qPCR), immunohistochemistry and fluorescence, electron, CCK-8, and flow cytometry were used to evaluate the phenotype of cardiac inflammation, fibrosis, apoptosis, and autophagy. Mice with HFD/STZ-induced diabetes exhibited systolic and diastolic dysfunction, as demonstrated by increased myocardial apoptosis and autophagy inhibition in the heart. Compared to control group, the protein expression of TRPM2, bax, cleaved caspase-3, and P62 was significantly elevated, and the protein expression of bcl-2 and LC3-II was significantly decreased in the myocardial tissues of the HFD/STZ-induced diabetes group. Knockdown of TRPM2 significantly reversed the HFD/STZ-induced myocardial apoptosis and autophagy inhibition. TRPM2 silencing attenuated HG-induced apoptosis and autophagy inhibition in primary cardiomyocytes via regulating the MEK/ERK mTORC1 signaling pathway. TRPM2 knockdown attenuates hyperglycemia-induced myocardial apoptosis and promotes autophagy in HFD/STZ-induced diabetic mice or HG-stimulated cardiomyocytes via regulating the MEK/ERK and mTORC1 signaling pathway.
Collapse
Affiliation(s)
- Feng Hu
- Department of Cardiology, Fujian Medical University Union Hospital, Fuzhou, 350001, Fujian, China.
| | - Chaoyang Lin
- Department of Cardiology, Fujian Medical University Union Hospital, Fuzhou, 350001, Fujian, China
| |
Collapse
|
6
|
Mandlem VKK, Rivera A, Khan Z, Quazi SH, Deba F. TLR4 induced TRPM2 mediated neuropathic pain. Front Pharmacol 2024; 15:1472771. [PMID: 39329114 PMCID: PMC11424904 DOI: 10.3389/fphar.2024.1472771] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2024] [Accepted: 09/02/2024] [Indexed: 09/28/2024] Open
Abstract
Ion channels play an important role in mediating pain through signal transduction, regulation, and control of responses, particularly in neuropathic pain. Transient receptor potential channel superfamily plays an important role in cation permeability and cellular signaling. Transient receptor potential channel Melastatin 2 (TRPM2) subfamily regulates Ca2+ concentration in response to various chemicals and signals from the surrounding environment. TRPM2 has a role in several physiological functions such as cellular osmosis, temperature sensing, cellular proliferation, as well as the manifestation of many disease processes such as pain process, cancer, apoptosis, endothelial dysfunction, angiogenesis, renal and lung fibrosis, and cerebral ischemic stroke. Toll-like Receptor 4 (TLR4) is a critical initiator of the immune response to inflammatory stimuli, particularly those triggered by Lipopolysaccharide (LPS). It activates downstream pathways leading to the production of oxidative molecules and inflammatory cytokines, which are modulated by basal and store-operated calcium ion signaling. The cytokine production and release cause an imbalance of antioxidant enzymes and redox potential in the Endoplasmic Reticulum and mitochondria due to oxidative stress, which results from TLR-4 activation and consequently induces the production of inflammatory cytokines in neuronal cells, exacerbating the pain process. Very few studies have reported the role of TRPM2 and its association with Toll-like receptors in the context of neuropathic pain. However, the molecular mechanism underlying the interaction between TRPM2 and TLR-4 and the quantum of impact in acute and chronic neuropathic pain remains unclear. Understanding the link between TLR-4 and TRPM2 will provide more insights into pain regulation mechanisms for the development of new therapeutic molecules to address neuropathic pain.
Collapse
Affiliation(s)
- Venkata Kiran Kumar Mandlem
- Departmental of Pharmaceutical Sciences and Health Outcomes, The Ben and Maytee Fisch College of Pharmacy, University of Texas at Tyler, Tyler, TX, United States
| | - Ana Rivera
- Departmental of Pharmaceutical Sciences and Health Outcomes, The Ben and Maytee Fisch College of Pharmacy, University of Texas at Tyler, Tyler, TX, United States
| | - Zaina Khan
- Departmental of Pharmaceutical Sciences and Health Outcomes, The Ben and Maytee Fisch College of Pharmacy, University of Texas at Tyler, Tyler, TX, United States
- Departmental of Neuroscience, University of Texas at Dallas, Richardson, TX, United States
| | - Sohel H Quazi
- Departmental of Pharmaceutical Sciences and Health Outcomes, The Ben and Maytee Fisch College of Pharmacy, University of Texas at Tyler, Tyler, TX, United States
- Department of Biology, Division of Natural and Computation Sciences, Texas College, Tyler, TX, United States
| | - Farah Deba
- Departmental of Pharmaceutical Sciences and Health Outcomes, The Ben and Maytee Fisch College of Pharmacy, University of Texas at Tyler, Tyler, TX, United States
| |
Collapse
|
7
|
Takeda A, Tamano H. Insight into brain metallothioneins from bidirectional Zn2+ signaling in synaptic dynamics. Metallomics 2024; 16:mfae039. [PMID: 39223100 DOI: 10.1093/mtomcs/mfae039] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2024] [Accepted: 09/01/2024] [Indexed: 09/04/2024]
Abstract
The basal levels as the labile Zn2+ pools in the extracellular and intracellular compartments are in the range of ∼10 nM and ∼100 pM, respectively. The influx of extracellular Zn2+ is used for memory via cognitive activity and is regulated for synaptic plasticity, a cellular mechanism of memory. When Zn2+ influx into neurons excessively occurs, however, it becomes a critical trigger for cognitive decline and neurodegeneration, resulting in acute and chronic pathogenesis. Aging, a biological process, generally accelerates vulnerability to neurodegenerative disorders such as Alzheimer's disease (AD) and Parkinson's disease (PD). The basal level of extracellular Zn2+ is age relatedly increased in the rat hippocampus, and the influx of extracellular Zn2+ contributes to accelerating vulnerability to the AD and PD pathogenesis in experimental animals with aging. Metallothioneins (MTs) are Zn2+-binding proteins for cellular Zn2+ homeostasis and involved in not only supplying functional Zn2+ required for cognitive activity, but also capturing excess (toxic) Zn2+ involved in cognitive decline and neurodegeneration. Therefore, it is estimated that regulation of MT synthesis is involved in both neuronal activity and neuroprotection. The present report provides recent knowledge regarding the protective/preventive potential of MT synthesis against not only normal aging but also the AD and PD pathogenesis in experimental animals, focused on MT function in bidirectional Zn2+ signaling in synaptic dynamics.
Collapse
Affiliation(s)
- Atsushi Takeda
- School of Pharmaceutical Sciences, University of Shizuoka, 52-1 Yada, Suruga-ku, Shizuoka 422-8526, Japan
| | - Haruna Tamano
- School of Pharmaceutical Sciences, University of Shizuoka, 52-1 Yada, Suruga-ku, Shizuoka 422-8526, Japan
- Shizuoka Tohto Medical College, 1949 Minamiema, Izunokuni, Shizuoka 410-2221, Japan
| |
Collapse
|
8
|
Zong P, Li CX, Feng J, Cicchetti M, Yue L. TRP Channels in Stroke. Neurosci Bull 2024; 40:1141-1159. [PMID: 37995056 PMCID: PMC11306852 DOI: 10.1007/s12264-023-01151-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2023] [Accepted: 09/11/2023] [Indexed: 11/24/2023] Open
Abstract
Ischemic stroke is a devastating disease that affects millions of patients worldwide. Unfortunately, there are no effective medications for mitigating brain injury after ischemic stroke. TRP channels are evolutionally ancient biosensors that detect external stimuli as well as tissue or cellular injury. To date, many members of the TRP superfamily have been reported to contribute to ischemic brain injury, including the TRPC subfamily (1, 3, 4, 5, 6, 7), TRPV subfamily (1, 2, 3, 4) and TRPM subfamily (2, 4, 7). These TRP channels share structural similarities but have distinct channel functions and properties. Their activation during ischemic stroke can be beneficial, detrimental, or even both. In this review, we focus on discussing the interesting features of stroke-related TRP channels and summarizing the underlying cellular and molecular mechanisms responsible for their involvement in ischemic brain injury.
Collapse
Affiliation(s)
- Pengyu Zong
- Department of Cell Biology, Calhoun Cardiology Center, School of Medicine (UConn Health), University of Connecticut, Farmington, CT, 06030, USA.
- Institute for the Brain and Cognitive Sciences, University of Connecticut, 337 Mansfield Road, Unit 1272, Storrs, CT, 06269, USA.
| | - Cindy X Li
- Department of Cell Biology, Calhoun Cardiology Center, School of Medicine (UConn Health), University of Connecticut, Farmington, CT, 06030, USA
| | - Jianlin Feng
- Department of Cell Biology, Calhoun Cardiology Center, School of Medicine (UConn Health), University of Connecticut, Farmington, CT, 06030, USA
| | - Mara Cicchetti
- Department of Cell Biology, Calhoun Cardiology Center, School of Medicine (UConn Health), University of Connecticut, Farmington, CT, 06030, USA
- Department of Neuroscience, University of Pittsburgh, 4200 Fifth Ave, Pittsburgh, PA, 15260, USA
| | - Lixia Yue
- Department of Cell Biology, Calhoun Cardiology Center, School of Medicine (UConn Health), University of Connecticut, Farmington, CT, 06030, USA.
| |
Collapse
|
9
|
Huang P, Qu C, Rao Z, Wu D, Zhao J. Bidirectional regulation mechanism of TRPM2 channel: role in oxidative stress, inflammation and ischemia-reperfusion injury. Front Immunol 2024; 15:1391355. [PMID: 39007141 PMCID: PMC11239348 DOI: 10.3389/fimmu.2024.1391355] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2024] [Accepted: 06/17/2024] [Indexed: 07/16/2024] Open
Abstract
Transient receptor potential melastatin 2 (TRPM2) is a non-selective cation channel that exhibits Ca2+ permeability. The TRPM2 channel is expressed in various tissues and cells and can be activated by multiple factors, including endogenous ligands, Ca2+, reactive oxygen species (ROS) and temperature. This article reviews the multiple roles of the TRPM2 channel in physiological and pathological processes, particularly on oxidative stress, inflammation and ischemia-reperfusion (I/R) injury. In oxidative stress, the excessive influx of Ca2+ caused by the activation of the TRPM2 channel may exacerbate cellular damage. However, under specific conditions, activating the TRPM2 channel can have a protective effect on cells. In inflammation, the activation of the TRPM2 channel may not only promote inflammatory response but also inhibit inflammation by regulating ROS production and bactericidal ability of macrophages and neutrophils. In I/R, the activation of the TRPM2 channel may worsen I/R injury to various organs, including the brain, heart, kidney and liver. However, activating the TRPM2 channel may protect the myocardium from I/R injury by regulating calcium influx and phosphorylating proline-rich tyrosine kinase 2 (Pyk2). A thorough investigation of the bidirectional role and regulatory mechanism of the TRPM2 channel in these physiological and pathological processes will aid in identifying new targets and strategies for treatment of related diseases.
Collapse
Affiliation(s)
- Peng Huang
- School of Kinesiology, Shanghai University of Sport, Shanghai, China
- Exercise Biological Center, China Institute of Sport Science, Beijing, China
| | - Chaoyi Qu
- Physical Education College, Hebei Normal University, Shijiazhuang, China
| | - Zhijian Rao
- Exercise Biological Center, China Institute of Sport Science, Beijing, China
- College of Physical Education, Shanghai Normal University, Shanghai, China
| | - Dongzhe Wu
- Exercise Biological Center, China Institute of Sport Science, Beijing, China
- Department of Exercise Physiology, Beijing Sport University, Beijing, China
| | - Jiexiu Zhao
- School of Kinesiology, Shanghai University of Sport, Shanghai, China
- Exercise Biological Center, China Institute of Sport Science, Beijing, China
| |
Collapse
|
10
|
Benarroch E. What Are the Functions of Zinc in the Nervous System? Neurology 2023; 101:714-720. [PMID: 37845046 PMCID: PMC10585682 DOI: 10.1212/wnl.0000000000207912] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2023] [Accepted: 08/16/2023] [Indexed: 10/18/2023] Open
|
11
|
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] [Download PDF] [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
|
12
|
Yu B, Jin L, Yao X, Zhang Y, Zhang G, Wang F, Su X, Fang Q, Xiao L, Yang Y, Jiang LH, Chen J, Yang W, Lin W, Han F. TRPM2 protects against cisplatin-induced acute kidney injury and mitochondrial dysfunction via modulating autophagy. Theranostics 2023; 13:4356-4375. [PMID: 37649595 PMCID: PMC10465213 DOI: 10.7150/thno.84655] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2023] [Accepted: 07/26/2023] [Indexed: 09/01/2023] Open
Abstract
Background: Cisplatin is a widely used anti-tumor agent but its use is frequently limited by nephrotoxicity. Transient receptor potential melastatin 2 (TRPM2) is a non-selective cation channel which is generally viewed as a sensor of oxidative stress, and increasing evidence supports its link with autophagy, a critical process for organelle homeostasis. Methods: Cisplatin-induced cell injury and mitochondrial damage were both assessed in WT and Trpm2-knockout mice and primary cells. RNA sequencing, immunofluorescence staining, immunoblotting and flowcytometry were applied to interpret the mechanism of TRPM2 in cisplatin nephrotoxicity. Results: Knockout of TRPM2 exacerbates renal dysfunction, tubular injury and cell apoptosis in a model of acute kidney injury (AKI) induced by treatment with cisplatin. Cisplatin-caused tubular mitochondrial damage is aggravated in TRPM2-deficient mice and cells and, conversely, alleviated by treatment with Mito-TEMPO, a mitochondrial ROS scavenger. TRPM2 deficiency hinders cisplatin-induced autophagy via blockage of Ca2+ influx and subsequent up-regulation of AKT-mTOR signaling. Consistently, cisplatin-induced tubular mitochondrial damage, cell apoptosis and renal dysfunction in TRPM2-deficient mice are mitigated by treatment with a mTOR inhibitor. Conclusion: Our results suggest that the TRPM2 channel plays a protective role in cisplatin-induced AKI via modulating the Ca2+-AKT-mTOR signaling pathway and autophagy, providing novel insights into the pathogenesis of kidney injury.
Collapse
Affiliation(s)
- Binfeng Yu
- Kidney Disease Center, The First Affiliated Hospital, Zhejiang University School of Medicine; Institute of Nephrology, Zhejiang University; Key Laboratory of Kidney Disease Prevention and Control Technology, Zhejiang Province; Zhejiang Clinical Research Center of Kidney and Urinary System Disease, Hangzhou 310003, China
- Department of Infectious Disease, Sir Run Run Shaw Hospital, Zhejiang University School of medicine, Hangzhou 310003, China
| | - Lini Jin
- Kidney Disease Center, The First Affiliated Hospital, Zhejiang University School of Medicine; Institute of Nephrology, Zhejiang University; Key Laboratory of Kidney Disease Prevention and Control Technology, Zhejiang Province; Zhejiang Clinical Research Center of Kidney and Urinary System Disease, Hangzhou 310003, China
- International Institutes of Medicine, The Fourth Affiliated Hospital, Zhejiang University School of Medicine, Yiwu 322000, China
| | - Xi Yao
- Kidney Disease Center, The First Affiliated Hospital, Zhejiang University School of Medicine; Institute of Nephrology, Zhejiang University; Key Laboratory of Kidney Disease Prevention and Control Technology, Zhejiang Province; Zhejiang Clinical Research Center of Kidney and Urinary System Disease, Hangzhou 310003, China
| | - Yi Zhang
- Department of Biophysics, and Department of Neurology of the Fourth Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310003, China
| | - Gensheng Zhang
- Department of Biophysics, and Department of Neurology of the Fourth Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310003, China
- The Children's Hospital, Zhejiang University School of medicine, Hangzhou 310003, China
| | - Fangqin Wang
- The Children's Hospital, Zhejiang University School of medicine, Hangzhou 310003, China
| | - Xinwan Su
- International Institutes of Medicine, The Fourth Affiliated Hospital, Zhejiang University School of Medicine, Yiwu 322000, China
| | - Qiuyuan Fang
- Department of Biophysics, and Department of Neurology of the Fourth Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310003, China
| | - Liang Xiao
- Kidney Disease Center, The First Affiliated Hospital, Zhejiang University School of Medicine; Institute of Nephrology, Zhejiang University; Key Laboratory of Kidney Disease Prevention and Control Technology, Zhejiang Province; Zhejiang Clinical Research Center of Kidney and Urinary System Disease, Hangzhou 310003, China
| | - Yi Yang
- International Institutes of Medicine, The Fourth Affiliated Hospital, Zhejiang University School of Medicine, Yiwu 322000, China
| | - Lin-Hua Jiang
- Sino-UK Joint Laboratory of Brain Function and Injury of Henan Province, and Department of Physiology and Pathophysiology, Xinxiang Medical University, P.R. China
- A4245-Transplantation, Immunology and Inflammation, Faculty of Medicine, University of Tours, France
- School of Biomedical Sciences, Faculty of Biological Sciences, University of Leeds, UK
| | - Jianghua Chen
- Kidney Disease Center, The First Affiliated Hospital, Zhejiang University School of Medicine; Institute of Nephrology, Zhejiang University; Key Laboratory of Kidney Disease Prevention and Control Technology, Zhejiang Province; Zhejiang Clinical Research Center of Kidney and Urinary System Disease, Hangzhou 310003, China
| | - Wei Yang
- Department of Biophysics, and Department of Neurology of the Fourth Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310003, China
| | - Weiqiang Lin
- International Institutes of Medicine, The Fourth Affiliated Hospital, Zhejiang University School of Medicine, Yiwu 322000, China
| | - Fei Han
- Kidney Disease Center, The First Affiliated Hospital, Zhejiang University School of Medicine; Institute of Nephrology, Zhejiang University; Key Laboratory of Kidney Disease Prevention and Control Technology, Zhejiang Province; Zhejiang Clinical Research Center of Kidney and Urinary System Disease, Hangzhou 310003, China
| |
Collapse
|
13
|
Ali ES, Chakrabarty B, Ramproshad S, Mondal B, Kundu N, Sarkar C, Sharifi-Rad J, Calina D, Cho WC. TRPM2-mediated Ca 2+ signaling as a potential therapeutic target in cancer treatment: an updated review of its role in survival and proliferation of cancer cells. Cell Commun Signal 2023; 21:145. [PMID: 37337283 PMCID: PMC10278317 DOI: 10.1186/s12964-023-01149-6] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2023] [Accepted: 04/28/2023] [Indexed: 06/21/2023] Open
Abstract
The transient receptor potential melastatin subfamily member 2 (TRPM2), a thermo and reactive oxygen species (ROS) sensitive Ca2+-permeable cation channel has a vital role in surviving the cell as well as defending the adaptability of various cell groups during and after oxidative stress. It shows higher expression in several cancers involving breast, pancreatic, prostate, melanoma, leukemia, and neuroblastoma, indicating it raises the survivability of cancerous cells. In various cancers including gastric cancers, and neuroblastoma, TRPM2 is known to conserve viability, and several underlying mechanisms of action have been proposed. Transcription factors are thought to activate TRPM2 channels, which is essential for cell proliferation and survival. In normal physiological conditions with an optimal expression of TRPM2, mitochondrial ROS is produced in optimal amounts while regulation of antioxidant expression is carried on. Depletion of TRPM2 overexpression or activity has been shown to improve ischemia-reperfusion injury in organ levels, reduce tumor growth and/or viability of various malignant cancers like breast, gastric, pancreatic, prostate, head and neck cancers, melanoma, neuroblastoma, T-cell and acute myelogenous leukemia. This updated and comprehensive review also analyzes the mechanisms by which TRPM2-mediated Ca2+ signaling can regulate the growth and survival of different types of cancer cells. Based on the discussion of the available data, it can be concluded that TRPM2 may be a unique therapeutic target in the treatment of several types of cancer. Video Abstract.
Collapse
Affiliation(s)
- Eunus S. Ali
- College of Medicine and Public Health, Flinders University, Bedford Park, 5042 Australia
- Gaco Pharmaceuticals, Dhaka, 1000 Bangladesh
- Present Address: Department of Biochemistry and Molecular Genetics, and Simpson Querrey Institute for Epigenetics, Northwestern University Feinberg School of Medicine, 303 E Superior St, Chicago, IL 60611 USA
| | | | - Sarker Ramproshad
- Department of Pharmacy, Ranada Prasad Shaha University, Narayanganj, 1400 Bangladesh
| | - Banani Mondal
- Department of Pharmacy, Ranada Prasad Shaha University, Narayanganj, 1400 Bangladesh
| | - Neloy Kundu
- Pharmacy Discipline, Khulna University, Khulna, 9208 Bangladesh
| | - Chandan Sarkar
- Department of Pharmacy, Bangabandhu Sheikh Mujibur Rahman Science and Technology University, Gopalganj, 8100 Bangladesh
| | | | - Daniela Calina
- Department of Clinical Pharmacy, University of Medicine and Pharmacy of Craiova, Craiova, 200349 Romania
| | - William C. Cho
- Department of Clinical Oncology, Queen Elizabeth Hospital, Kowloon, Hong Kong, China
| |
Collapse
|
14
|
Zhong C, Yang J, Zhang Y, Fan X, Fan Y, Hua N, Li D, Jin S, Li Y, Chen P, Chen Y, Cai X, Zhang Y, Jiang L, Yang W, Yu P, Lin H. TRPM2 Mediates Hepatic Ischemia-Reperfusion Injury via Ca 2+-Induced Mitochondrial Lipid Peroxidation through Increasing ALOX12 Expression. RESEARCH (WASHINGTON, D.C.) 2023; 6:0159. [PMID: 37275121 PMCID: PMC10232356 DOI: 10.34133/research.0159] [Citation(s) in RCA: 22] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/28/2023] [Accepted: 05/03/2023] [Indexed: 06/07/2023]
Abstract
Hepatic ischemia-reperfusion (IR) injury is a serious clinical problem that complicates liver resection and transplantation. Despite recent advances in understanding of the pathophysiology of hepatic IR injury, effective interventions and therapeutics are still lacking. Here, we examined the role of transient receptor potential melastatin 2 (TRPM2), a Ca2+-permeable, non-selective cation channel, in mediating hepatic IR injury. Our data showed that TRPM2 deficiency attenuated IR-induced liver dysfunction, inflammation, and cell death in mice. Moreover, RNA sequencing analysis indicated that TRPM2-induced IR injury occurs via ferroptosis-related pathways. Consistently, as a ferroptosis inducer, (1S,3R)-RSL3 treatment induced mitochondrial dysfunction in hepatocytes and a TRPM2 inhibitor suppressed this. Interestingly, TRPM2-mediated calcium influx caused mitochondrial calcium accumulation via the mitochondrial Ca2+-selective uniporter and increased the expression level of arachidonate 12-lipoxygenase (ALOX12), which results in mitochondrial lipid peroxidation during hepatic IR injury. Furthermore, hepatic IR injury-induced ferroptosis was obviously relieved by a TRPM2 inhibitor or calcium depletion, both in vitro and in vivo. Collectively, these findings demonstrate a crucial role for TRPM2-mediated ferroptosis in hepatic IR injury via increased Ca2+-induced ALOX12 expression, indicating that pharmacological inhibition of TRPM2 may provide an effective therapeutic strategy for hepatic IR injury-related diseases, such as during liver resection and transplantation.
Collapse
Affiliation(s)
- Cheng Zhong
- Department of General Surgery, Sir Run Run Shaw Hospital, School of Medicine,
Zhejiang University, Hangzhou, P.R. China
| | - Jing Yang
- Department of General Surgery, Sir Run Run Shaw Hospital, School of Medicine,
Zhejiang University, Hangzhou, P.R. China
| | - Yiyin Zhang
- Department of General Surgery, Sir Run Run Shaw Hospital, School of Medicine,
Zhejiang University, Hangzhou, P.R. China
| | - Xiaoxiao Fan
- Department of General Surgery, Sir Run Run Shaw Hospital, School of Medicine,
Zhejiang University, Hangzhou, P.R. China
| | - Yang Fan
- Department of Toxicology and Department of Medical Oncology of Second Affiliated Hospital,
Zhejiang University School of Medicine, Hangzhou, P.R. China
| | - Ning Hua
- Department of Physiology and Pathophysiology and Sino-UK Joint Laboratory of Brain Function and Injury of Henan Province,
Xinxiang Medical University, 453003 Xinxiang, Henan, P.R. China
| | - Duguang Li
- Department of General Surgery, Sir Run Run Shaw Hospital, School of Medicine,
Zhejiang University, Hangzhou, P.R. China
| | - Shengxi Jin
- Department of General Surgery, Sir Run Run Shaw Hospital, School of Medicine,
Zhejiang University, Hangzhou, P.R. China
| | - Yirun Li
- Department of General Surgery, Sir Run Run Shaw Hospital, School of Medicine,
Zhejiang University, Hangzhou, P.R. China
| | - Peng Chen
- Department of General Surgery, Sir Run Run Shaw Hospital, School of Medicine,
Zhejiang University, Hangzhou, P.R. China
| | - Yongle Chen
- Department of General Surgery, Sir Run Run Shaw Hospital, School of Medicine,
Zhejiang University, Hangzhou, P.R. China
| | - Xiaobo Cai
- Department of Biophysics and Department of Neurology of the Fourth Affiliated Hospital,
Zhejiang University School of Medicine, Hangzhou 310000, P.R. China
| | - Yi Zhang
- Department of Biophysics and Department of Neurology of the Fourth Affiliated Hospital,
Zhejiang University School of Medicine, Hangzhou 310000, P.R. China
| | - Linhua Jiang
- Department of Physiology and Pathophysiology and Sino-UK Joint Laboratory of Brain Function and Injury of Henan Province,
Xinxiang Medical University, 453003 Xinxiang, Henan, P.R. China
- School of Biomedical Sciences, Faculty of Biological Sciences, University of Leeds, LS2 9JT Leeds, UK
| | - Wei Yang
- Department of Biophysics and Department of Neurology of the Fourth Affiliated Hospital,
Zhejiang University School of Medicine, Hangzhou 310000, P.R. China
| | - Peilin Yu
- Department of Toxicology and Department of Medical Oncology of Second Affiliated Hospital,
Zhejiang University School of Medicine, Hangzhou, P.R. China
| | - Hui Lin
- Department of General Surgery, Sir Run Run Shaw Hospital, School of Medicine,
Zhejiang University, Hangzhou, P.R. China
- Zhejiang Engineering Research Center of Cognitive Healthcare, Sir Run Run Shaw Hospital,
School of Medicine, Zhejiang University, Hangzhou 310020, P.R. China
- College of Biomedical Engineering and Instrument Science,
Zhejiang University, Hangzhou 310058, P.R. China
| |
Collapse
|
15
|
Xu J, Zhang W, Dong J, Cao L, Huang Z. A New Potential Strategy for Treatment of Ischemic Stroke: Targeting TRPM2-NMDAR Association. Neurosci Bull 2023; 39:703-706. [PMID: 36342656 PMCID: PMC10073358 DOI: 10.1007/s12264-022-00971-1] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2022] [Accepted: 08/09/2022] [Indexed: 11/09/2022] Open
Affiliation(s)
- Jiayun Xu
- School of Pharmacy, Hangzhou Normal University, Hangzhou, 311121, China
| | - Wei Zhang
- School of Pharmacy, Hangzhou Normal University, Hangzhou, 311121, China
| | - Jianhong Dong
- School of Pharmacy, Hangzhou Normal University, Hangzhou, 311121, China
| | - Liying Cao
- School of Pharmacy, Hangzhou Normal University, Hangzhou, 311121, China.
- Laboratory of Aging and Cancer Biology of Zhejiang Province, School of Medicine, Hangzhou Normal University, Hangzhou, 311121, China.
| | - Zhihui Huang
- School of Pharmacy, Hangzhou Normal University, Hangzhou, 311121, China.
| |
Collapse
|
16
|
Zhang XM, Song Y, Zhu XY, Wang WJ, Fan XL, El-Aziz TMA. MITOCHONDRIA: The dual function of the transient receptor potential melastatin 2 channels from cytomembrane to mitochondria. Int J Biochem Cell Biol 2023; 157:106374. [PMID: 36708986 DOI: 10.1016/j.biocel.2023.106374] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2022] [Revised: 12/20/2022] [Accepted: 01/24/2023] [Indexed: 01/26/2023]
Abstract
Mitochondria are closely related to oxidative stress and play an important role in maintaining cell functional homeostasis and meeting cell energy demand. The transient receptor potential melastatin 2 (TRPM2) channel affects the occurrence and progression of diseases by regulating mitochondrial function. TRPM2 channel promotes Ca2+ influx to affect 18 kDa translocator protein (TSPO), mitochondrial membrane potential (MMP), reactive oxygen species (ROS), adenosine triphosphate (ATP) production, and mitochondrial autophagy. The mechanism of Ca2+ influx into the mitochondria by TRPM2 is abundant. Interestingly, the TRPM2 channel inhibits the production of mitochondrial ROS in cancer cells and promotes the production of mitochondrial ROS in normal cells, which induces cell death in normal cells but proliferation in cancer cells. TRPM2 can be a potential target for the treatment of various diseases due to its role as a molecular link between mitochondria and Ca2+ signals.
Collapse
Affiliation(s)
- Xiao-Min Zhang
- Department of Pharmacology, Zhejiang University of Technology, Hangzhou, Zhejiang 310014, China
| | - Ying Song
- Department of Pharmacology, Zhejiang University of Technology, Hangzhou, Zhejiang 310014, China.
| | - Xin-Yi Zhu
- Department of Pharmacology, Zhejiang University of Technology, Hangzhou, Zhejiang 310014, China
| | - Wen-Jun Wang
- Department of Pharmacology, Zhejiang University of Technology, Hangzhou, Zhejiang 310014, China
| | - Xu-Li Fan
- Department of Pharmacology, Zhejiang University of Technology, Hangzhou, Zhejiang 310014, China
| | - Tarek Mohamed Abd El-Aziz
- Department of Cellular and Integrative Physiology, University of Texas Health Science Center at San Antonio, San Antonio, TX 78229-3900, USA; Zoology Department, Faculty of Science, Minia University, El-Minia 61519, Egypt.
| |
Collapse
|
17
|
Çınar R, Nazıroğlu M. TRPM2 Channel Inhibition Attenuates Amyloid β42-Induced Apoptosis and Oxidative Stress in the Hippocampus of Mice. Cell Mol Neurobiol 2023; 43:1335-1353. [PMID: 35840808 PMCID: PMC11414446 DOI: 10.1007/s10571-022-01253-0] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2022] [Accepted: 07/01/2022] [Indexed: 01/16/2023]
Abstract
Alzheimer's disease (AD) is characterized by the increase of hippocampal Ca2+ influx-induced apoptosis and mitochondrial oxidative stress (OS). The OS is a stimulator of TRPM2, although N-(p-amylcinnamoyl)anthranilic acid (ACA), 2-aminoethyl diphenylborinate (2/APB), and glutathione (GSH) are non-specific antagonists of TRPM2. In the present study, we investigated the protective roles of GSH and TRPM2 antagonist treatments on the amyloid β42 peptide (Aβ)-caused oxidative neurotoxicity and apoptosis in the hippocampus of mice with AD model. After the isolation of hippocampal neurons from the newborn mice, they were divided into five incubation groups as follows: control, ACA, Aβ, Aβ+ACA, and Aβ+GSH. The levels of apoptosis, hippocampus death, cytosolic ROS, cytosolic Zn2+, mitochondrial ROS, caspase-3, caspase-9, lipid peroxidation, and cytosolic Ca2+ were increased in the primary hippocampus cultures by treatments of Aβ, although their levels were decreased in the neurons by the treatments of GSH, PARP-1 inhibitors (PJ34 and DPQ), and TRPM2 blockers (ACA and 2/APB). The Aβ-induced decreases of cell viability, cytosolic GSH, reduced GSH, and GSH peroxidase levels were also increased in the groups of Aβ+ACA and Aβ+GSH by the treatments of ACA and GSH. However, the Aβ-caused changes were not observed in the hippocampus of TRPM2-knockout mice. In conclusion, the present data demonstrate that maintaining the activation of TRPM2 is not only important for the quenching OS and neurotoxicity in the hippocampal neurons of mice with experimental AD but also equally critical to the modulation of Aβ-induced apoptosis. The possible positive effects of GSH and TRPM2 antagonist treatments on the amyloid-beta (Aβ)-induced oxidative toxicity in the hippocampus of mice. The ADP-ribose (ADPR) is produced via the stimulation of PARP-1 in the nucleus of neurons. The NUT9 in the C terminus of TRPM2 channel acts as a key role for the activation of TRPM2. The antagonists of TRPM2 are glutathione (GSH), ACA, and 2/APB in the hippocampus. The Aβ incubation-mediated TRPM2 stimulation increases the concentration of cytosolic-free Ca2+ and Zn2+ in the hippocampus. In turn, the increased concentration causes the increase of mitochondrial membrane potential (ΔΨm), which causes the excessive generations of mitochondria ROS and the decrease of cytosolic GSH and GSH peroxidase (GSH-Px). The ROS production and GSH depletion are two main causes in the neurobiology of Alzheimer's disease. However, the effect of Aβ was not shown in the hippocampus of TRPM2-knockout mice. The Aβ and TRPM2 stimulation-caused overload Ca2+ entry cause apoptosis and cell death via the activations of caspase-3 (Casp/3) and caspase-9 (Casp/9) in the hippocampus. The actions of Aβ-induced oxidative toxicity were modulated in the primary hippocampus by the incubations of ACA, GSH, 2/APB, and PARP-1 inhibitors (PJ34 and DPQ). (↑) Increase. (↓) Decrease.
Collapse
Affiliation(s)
- Ramazan Çınar
- Department of Neuroscience, Health Science Institute, Suleyman Demirel University, Isparta, Turkey
| | - Mustafa Nazıroğlu
- Department of Neuroscience, Health Science Institute, Suleyman Demirel University, Isparta, Turkey.
- Neuroscience Research Center, Suleyman Demirel University, Isparta, Turkey.
- Drug Discovery Unit, BSN Health, Analyses, Innovation, Consultancy, Organization, Agriculture and Industry Ltd., Isparta, Turkey.
- Department of Biophysics, School of Medicine, University of Suleyman Demirel, 32260, Isparta, Turkey.
| |
Collapse
|
18
|
Zhou Q, Fu X, Xu J, Dong S, Liu C, Cheng D, Gao C, Huang M, Liu Z, Ni X, Hua R, Tu H, Sun H, Shen Q, Chen B, Zhang J, Zhang L, Yang H, Hu J, Yang W, Pei W, Yao Q, Sheng X, Zhang J, Yang WZ, Shen WL. Hypothalamic warm-sensitive neurons require TRPC4 channel for detecting internal warmth and regulating body temperature in mice. Neuron 2023; 111:387-404.e8. [PMID: 36476978 DOI: 10.1016/j.neuron.2022.11.008] [Citation(s) in RCA: 18] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2021] [Revised: 06/28/2022] [Accepted: 11/07/2022] [Indexed: 12/12/2022]
Abstract
Precise monitoring of internal temperature is vital for thermal homeostasis in mammals. For decades, warm-sensitive neurons (WSNs) within the preoptic area (POA) were thought to sense internal warmth, using this information as feedback to regulate body temperature (Tcore). However, the cellular and molecular mechanisms by which WSNs measure temperature remain largely undefined. Via a pilot genetic screen, we found that silencing the TRPC4 channel in mice substantially attenuated hypothermia induced by light-mediated heating of the POA. Loss-of-function studies of TRPC4 confirmed its role in warm sensing in GABAergic WSNs, causing additional defects in basal temperature setting, warm defense, and fever responses. Furthermore, TRPC4 antagonists and agonists bidirectionally regulated Tcore. Thus, our data indicate that TRPC4 is essential for sensing internal warmth and that TRPC4-expressing GABAergic WSNs function as a novel cellular sensor for preventing Tcore from exceeding set-point temperatures. TRPC4 may represent a potential therapeutic target for managing Tcore.
Collapse
Affiliation(s)
- Qian Zhou
- School of Life Science and Technology, Shanghai Clinical Research and Trial Center, Shanghai Institute for Advanced Immunochemical Studies, ShanghaiTech University, Shanghai 201210, China; Institute of Neuroscience, State Key Laboratory of Neuroscience, CAS Center for Excellence in Brain Science and Intelligence Technology, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai 200031, China; University of Chinese Academy of Sciences, Beijing 100049, China
| | - Xin Fu
- School of Life Science and Technology, Shanghai Clinical Research and Trial Center, Shanghai Institute for Advanced Immunochemical Studies, ShanghaiTech University, Shanghai 201210, China; Institute of Neuroscience, State Key Laboratory of Neuroscience, CAS Center for Excellence in Brain Science and Intelligence Technology, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai 200031, China; University of Chinese Academy of Sciences, Beijing 100049, China
| | - Jianhui Xu
- Thermoregulation and Inflammation Laboratory, Chengdu Medical College, Chengdu, Sichuan 610500, China
| | - Shiming Dong
- University of Chinese Academy of Sciences, Beijing 100049, China; Shanghai Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences (CAS), Shanghai 200031, China
| | - Changhao Liu
- School of Life Science and Technology, Shanghai Clinical Research and Trial Center, Shanghai Institute for Advanced Immunochemical Studies, ShanghaiTech University, Shanghai 201210, China
| | - Dali Cheng
- Department of Electronic Engineering, Tsinghua University, Beijing 100084, China
| | - Cuicui Gao
- School of Life Science and Technology, Shanghai Clinical Research and Trial Center, Shanghai Institute for Advanced Immunochemical Studies, ShanghaiTech University, Shanghai 201210, China; Institute of Neuroscience, State Key Laboratory of Neuroscience, CAS Center for Excellence in Brain Science and Intelligence Technology, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai 200031, China; University of Chinese Academy of Sciences, Beijing 100049, China
| | - Minhua Huang
- Department of Biophysics, NHC and CAMS Key Laboratory of Medical Neurobiology, Zhejiang University School of Medicine, Hangzhou 310058, China
| | - Zhiduo Liu
- University of Chinese Academy of Sciences, Beijing 100049, China; State Key Laboratory of Integrated Optoelectronics, Institute of Semiconductors, Chinese Academy of Sciences, Beijing 100083, China
| | - Xinyan Ni
- School of Life Science and Technology, Shanghai Clinical Research and Trial Center, Shanghai Institute for Advanced Immunochemical Studies, ShanghaiTech University, Shanghai 201210, China
| | - Rong Hua
- Department of General Surgery, Huashan Hospital, Fudan University, Shanghai 200433, China
| | - Hongqing Tu
- School of Life Science and Technology, Shanghai Clinical Research and Trial Center, Shanghai Institute for Advanced Immunochemical Studies, ShanghaiTech University, Shanghai 201210, China
| | - Hongbin Sun
- School of Life Science and Technology, Shanghai Clinical Research and Trial Center, Shanghai Institute for Advanced Immunochemical Studies, ShanghaiTech University, Shanghai 201210, China
| | - Qiwei Shen
- Department of General Surgery, Huashan Hospital, Fudan University, Shanghai 200433, China
| | - Baoting Chen
- School of Life Science and Technology, Shanghai Clinical Research and Trial Center, Shanghai Institute for Advanced Immunochemical Studies, ShanghaiTech University, Shanghai 201210, China; Institute of Neuroscience, State Key Laboratory of Neuroscience, CAS Center for Excellence in Brain Science and Intelligence Technology, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai 200031, China; University of Chinese Academy of Sciences, Beijing 100049, China
| | - Jin Zhang
- School of Basic Medical Sciences, Nanchang University, Nanchang 330031, China
| | - Liye Zhang
- School of Life Science and Technology, Shanghai Clinical Research and Trial Center, Shanghai Institute for Advanced Immunochemical Studies, ShanghaiTech University, Shanghai 201210, China
| | - Haitao Yang
- School of Life Science and Technology, Shanghai Clinical Research and Trial Center, Shanghai Institute for Advanced Immunochemical Studies, ShanghaiTech University, Shanghai 201210, China
| | - Ji Hu
- School of Life Science and Technology, Shanghai Clinical Research and Trial Center, Shanghai Institute for Advanced Immunochemical Studies, ShanghaiTech University, Shanghai 201210, China
| | - Wei Yang
- Department of Biophysics, NHC and CAMS Key Laboratory of Medical Neurobiology, Zhejiang University School of Medicine, Hangzhou 310058, China
| | - Weihua Pei
- State Key Laboratory of Integrated Optoelectronics, Institute of Semiconductors, Chinese Academy of Sciences, Beijing 100083, China
| | - Qiyuan Yao
- Department of General Surgery, Huashan Hospital, Fudan University, Shanghai 200433, China
| | - Xing Sheng
- Department of Electronic Engineering, Tsinghua University, Beijing 100084, China
| | - Jie Zhang
- Thermoregulation and Inflammation Laboratory, Chengdu Medical College, Chengdu, Sichuan 610500, China.
| | - Wen Z Yang
- School of Life Science and Technology, Shanghai Clinical Research and Trial Center, Shanghai Institute for Advanced Immunochemical Studies, ShanghaiTech University, Shanghai 201210, China.
| | - Wei L Shen
- School of Life Science and Technology, Shanghai Clinical Research and Trial Center, Shanghai Institute for Advanced Immunochemical Studies, ShanghaiTech University, Shanghai 201210, China.
| |
Collapse
|
19
|
Xu Q, Zou Y, Miao Z, Jiang L, Zhao X. Transient receptor potential ion channels and cerebral stroke. Brain Behav 2023; 13:e2843. [PMID: 36527242 PMCID: PMC9847613 DOI: 10.1002/brb3.2843] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/18/2022] [Accepted: 11/23/2022] [Indexed: 12/23/2022] Open
Abstract
METHODS The databases Pubmed, and the National Library of Medicine were searched for literature. All papers on celebral stroke and transient receptor potential ion channels were considered. RESULTS Stroke is the second leading cause of death and disability, with an increasing incidence in developing countries. About 75 per cent of strokes are caused by occlusion of cerebral arteries, and substantial advances have been made in elucidating mechanisms how stroke affects the brain. Transient receptor potential (TRP) ion channels are calcium-permeable channels highly expressed in brain that drives Ca2+ entry into multiple cellular compartments. TRPC1/3/4/6, TRPV1/2/4, and TRPM2/4/7 channels have been implicated in stroke pathophysiology. CONCLUSIONS Although the precise mechanism of transient receptor potential ion channels in cerebral stroke is still unclear, it has the potential to be a therapeutic target for patients with stroke if developed appropriately. Hence, more research is needed to prove its efficacy in this context.
Collapse
Affiliation(s)
- Qin'yi Xu
- Department of Neurosurgery, The Affiliated Wuxi No. 2 Hospital of Nanjing Medical University, Wuxi, China
| | - Yan Zou
- Department of Neurosurgery, The Affiliated Jinling Hospital, Medical School of Nanjing University, Nanjing, China
| | - Zeng'li Miao
- Department of Neurosurgery, The Affiliated Wuxi No. 2 Hospital of Nanjing Medical University, Wuxi, China
| | - Lei Jiang
- Department of Neurosurgery, The Affiliated Wuxi No. 2 Hospital of Nanjing Medical University, Wuxi, China
| | - Xu'dong Zhao
- Department of Neurosurgery, The Affiliated Wuxi No. 2 Hospital of Nanjing Medical University, Wuxi, China
| |
Collapse
|
20
|
Ying Y, Gong L, Tao X, Ding J, Chen N, Yao Y, Liu J, Chen C, Zhu T, Jiang P. Genetic Knockout of TRPM2 Increases Neuronal Excitability of Hippocampal Neurons by Inhibiting Kv7 Channel in Epilepsy. Mol Neurobiol 2022; 59:6918-6933. [PMID: 36053438 DOI: 10.1007/s12035-022-02993-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2022] [Accepted: 08/07/2022] [Indexed: 11/30/2022]
Abstract
Epilepsy is a chronic brain disease that makes serious cognitive and motor retardation. Ion channels affect the occurrence of epilepsy in various ways, but the mechanisms have not yet been fully elucidated. Transient receptor potential melastain2 (TRPM2) ion channel is a non-selective cationic channel that can permeate Ca2+ and critical for epilepsy. Here, TRPM2 gene knockout mice were used to generate a chronic kindling epilepsy model by PTZ administration in mice. We found that TRPM2 knockout mice were more susceptible to epilepsy than WT mice. Furthermore, the neuronal excitability in the hippocampal CA1 region of TRPM2 knockout mice was significantly increased. Compared with WT group, there were no significant differences in the input resistance and after hyperpolarization of CA1 neurons in TRPM2 knockout mice. Firing adaptation rate of hippocampal CA1 pyramidal neurons of TRPM2 knockout mice was lower than that of WT mice. We also found that activation of Kv7 channel by retigabine reduced the firing frequency of action potential in the hippocampal pyramidal neurons of TRPM2 knockout mice. However, inhibiting Kv7 channel increased the firing frequency of action potential in hippocampal pyramidal neurons of WT mice. The data suggest that activation of Kv7 channel can effectively reduce epileptic seizures in TRPM2 knockout mice. We conclude that genetic knockout of TRPM2 in hippocampal CA1 pyramidal neurons may increase neuronal excitability by inhibiting Kv7 channel, affecting the susceptibility to epilepsy. These findings may provide a potential therapeutic target for epilepsy.
Collapse
Affiliation(s)
- Yingchao Ying
- Department of Neurology, Children's Hospital, Zhejiang University School of Medicine, National Clinical Research Center for Child Health, Hangzhou, China
| | - Lifen Gong
- Department of Neurology, Children's Hospital, Zhejiang University School of Medicine, National Clinical Research Center for Child Health, Hangzhou, China
| | - Xiaohan Tao
- Department of Neurology, Children's Hospital, Zhejiang University School of Medicine, National Clinical Research Center for Child Health, Hangzhou, China
| | - Junchao Ding
- Department of Neurology, Children's Hospital, Zhejiang University School of Medicine, National Clinical Research Center for Child Health, Hangzhou, China
- Department of Pediatrics, Yiwu Maternal and Child Health Care Hospital, Yiwu, China
| | - Nannan Chen
- Department of Neurology, Children's Hospital, Zhejiang University School of Medicine, National Clinical Research Center for Child Health, Hangzhou, China
| | - Yinping Yao
- Department of Neurology, Children's Hospital, Zhejiang University School of Medicine, National Clinical Research Center for Child Health, Hangzhou, China
- Department of Pediatrics, Shaoxing People's Hospital, Shaoxing, China
| | - Jiajing Liu
- Department of Neurology, Children's Hospital, Zhejiang University School of Medicine, National Clinical Research Center for Child Health, Hangzhou, China
| | - Chen Chen
- Department of Neurology, Children's Hospital, Zhejiang University School of Medicine, National Clinical Research Center for Child Health, Hangzhou, China
| | - Tao Zhu
- Department of Critical Care Medicine, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, China.
| | - Peifang Jiang
- Department of Neurology, Children's Hospital, Zhejiang University School of Medicine, National Clinical Research Center for Child Health, Hangzhou, China.
| |
Collapse
|
21
|
Wang Y, Liu J, Yu B, Jin Y, Li J, Ma X, Yu J, Niu J, Liang X. Umbilical cord-derived mesenchymal stem cell conditioned medium reverses neuronal oxidative injury by inhibition of TRPM2 activation and the JNK signaling pathway. Mol Biol Rep 2022; 49:7337-7345. [PMID: 35585377 PMCID: PMC9304044 DOI: 10.1007/s11033-022-07524-9] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2022] [Revised: 04/20/2022] [Accepted: 04/26/2022] [Indexed: 11/30/2022]
Abstract
Background The mechanism by which MSC-CM protects neuronal cells against ischemic injury remains to be elucidated. In this study, we aimed to clarify the protective effect of umbilical cord-derived mesenchymal stem cell conditioned medium (UC-MSC-CM) on neuronal oxidative injury and its potential mechanism. Methods and Results Neuronal oxidative damage was mimicked by H2O2 treatment of the HT22 cell line. The numbers of cleaved-Caspase-3-positive cells and protein expression of Caspase-9 induced by H2O2 treatment were decreased by UC-MSC-CM treatment. Furthermore, SOD protein expression was increased in the MSC-CM group compared with that in the H2O2 group. The H2O2-induced TRPM2-like currents in HT22 cells were attenuated by MSC-CM treatment. In addition, H2O2 treatment downregulated the expression of p-JNK protein in HT22 cells, and this the downward trend was reversed by incubation with MSC-CM. Conclusions UC-MSC-CM protects neurons against oxidative injury, possibly by inhibiting activation of TRPM2 and the JNK signaling pathway.
Collapse
Affiliation(s)
- Yan Wang
- Key Laboratory of Ningxia Stem Cell and Regenerative Medicine, General Hospital of Ningxia Medical University, 750001, Yinchuan, China
| | - Jiaxin Liu
- Key Laboratory of Ningxia Stem Cell and Regenerative Medicine, General Hospital of Ningxia Medical University, 750001, Yinchuan, China
| | - Baocong Yu
- Ningxia Key Laboratory of Cerebrocranial Diseases, Ningxia Medical University, 750004, Yinchuan, China
| | - Yiran Jin
- Key Laboratory of Ningxia Stem Cell and Regenerative Medicine, General Hospital of Ningxia Medical University, 750001, Yinchuan, China
| | - Jiahui Li
- Key Laboratory of Ningxia Stem Cell and Regenerative Medicine, General Hospital of Ningxia Medical University, 750001, Yinchuan, China
| | - Xiaona Ma
- Key Laboratory of Ningxia Stem Cell and Regenerative Medicine, General Hospital of Ningxia Medical University, 750001, Yinchuan, China
| | - Jianqiang Yu
- School of Pharmacology, Ningxia Medical University, 750004, Yinchuan, China.
| | - Jianguo Niu
- Ningxia Key Laboratory of Cerebrocranial Diseases, Ningxia Medical University, 750004, Yinchuan, China.
| | - Xueyun Liang
- Key Laboratory of Ningxia Stem Cell and Regenerative Medicine, General Hospital of Ningxia Medical University, 750001, Yinchuan, China.
| |
Collapse
|
22
|
Sander S, Pick J, Gattkowski E, Fliegert R, Tidow H. The crystal structure of
TRPM2 MHR1
/2 domain reveals a conserved Zn
2+
‐binding domain essential for structural integrity and channel activity. Protein Sci 2022; 31:e4320. [PMID: 35634784 PMCID: PMC9112350 DOI: 10.1002/pro.4320] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2022] [Revised: 03/02/2022] [Accepted: 04/10/2022] [Indexed: 11/12/2022]
Affiliation(s)
- Simon Sander
- Hamburg Advanced Research Centre for Bioorganic Chemistry (HARBOR) & Department of Chemistry Institute for Biochemistry and Molecular Biology, University of Hamburg Hamburg Germany
| | - Jelena Pick
- The Calcium Signaling Group, Department of Biochemistry and Molecular Cell Biology University Medical Center Hamburg‐Eppendorf Hamburg Germany
| | - Ellen Gattkowski
- Hamburg Advanced Research Centre for Bioorganic Chemistry (HARBOR) & Department of Chemistry Institute for Biochemistry and Molecular Biology, University of Hamburg Hamburg Germany
| | - Ralf Fliegert
- The Calcium Signaling Group, Department of Biochemistry and Molecular Cell Biology University Medical Center Hamburg‐Eppendorf Hamburg Germany
| | - Henning Tidow
- Hamburg Advanced Research Centre for Bioorganic Chemistry (HARBOR) & Department of Chemistry Institute for Biochemistry and Molecular Biology, University of Hamburg Hamburg Germany
| |
Collapse
|
23
|
Yin YL, Liu YH, Zhu ML, Wang HH, Qiu Y, Wan GR, Li P. Floralozone improves cognitive impairment in vascular dementia rats via regulation of TRPM2 and NMDAR signaling pathway. Physiol Behav 2022; 249:113777. [PMID: 35276121 DOI: 10.1016/j.physbeh.2022.113777] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2021] [Revised: 02/23/2022] [Accepted: 03/07/2022] [Indexed: 12/11/2022]
Abstract
Vascular dementia (VD) is the second largest type of dementia after Alzheimer's disease. At present, the pathogenesis is complex and there is no effective treatment. Floralozone has been shown to reduce atherosclerosis in rats caused by a high-fat diet. However, whether it plays a role in VD remains elusive. In the present study, the protective activities and relevant mechanisms of Floralozone were evaluated in rats with cognitive impairment, which were induced by bilateral occlusion of the common carotid arteries (BCCAO) in rats. Cognitive function, pathological changes and oxidative stress condition in the brains of VD rats were assessed using Neurobehavioral tests, Morris water maze tests, hematoxylin-eosin staining, Neu N staining, TUNEL staining, Golgi staining, Western blot assay and antioxidant assays (MDA, SOD, GSH), respectively. The results indicated that VD model was established successfully and BCCAO caused a decline in spatial learning and memory and hippocampal histopathological abnormalities of rats. Floralozone (50, 100, 150 mg/kg) dose-dependently alleviated the pathological changes, decreased oxidative stress injury, which eventually reduced cognitive impairment in BCCAO rats. The same results were shown in further experiments with neurobehavioral tests. At the molecular biological level, Floralozone decreased the protein level of transient receptor potential melastatin-related 2 (TRPM2) in VD and normal rats, and increased the protein level of NR2B in hippocampus of N-methyl-D-aspartate receptor (NMDAR). Notably, Floralozone could markedly improved learning and memory function of BCCAO rats in Morris water maze (MWM) and improved neuronal cell loss, synaptic structural plasticity. In conclusion, Floralozone has therapeutic potential for VD, increased synaptic structural plasticity and alleviating neuronal cell apoptosis, which may be related to the TRPM2/NMDAR pathway.
Collapse
Affiliation(s)
- Ya-Ling Yin
- School of Basic Medical Sciences, Department of Physiology and Pathophysiology, Sino-UK Joint Laboratory of Brain Function and Injury and Department of Physiology and Neurobiology, Xinxiang Medical University,Xinxiang, China, 453003; College of Pharmacy, Henan international joint laboratory of cardiovascular remodeling and drug intervention, Xinxiang key laboratory of vascular remodeling intervention and molecular targeted therapy drug development, Xinxiang Medical University,Xinxiang, China, 453003.
| | - Yan-Hua Liu
- College of Pharmacy, Henan international joint laboratory of cardiovascular remodeling and drug intervention, Xinxiang key laboratory of vascular remodeling intervention and molecular targeted therapy drug development, Xinxiang Medical University,Xinxiang, China, 453003.
| | - Mo-Li Zhu
- College of Pharmacy, Henan international joint laboratory of cardiovascular remodeling and drug intervention, Xinxiang key laboratory of vascular remodeling intervention and molecular targeted therapy drug development, Xinxiang Medical University,Xinxiang, China, 453003.
| | - Huan-Huan Wang
- College of Pharmacy, Henan international joint laboratory of cardiovascular remodeling and drug intervention, Xinxiang key laboratory of vascular remodeling intervention and molecular targeted therapy drug development, Xinxiang Medical University,Xinxiang, China, 453003.
| | - Yue Qiu
- College of Pharmacy, Henan international joint laboratory of cardiovascular remodeling and drug intervention, Xinxiang key laboratory of vascular remodeling intervention and molecular targeted therapy drug development, Xinxiang Medical University,Xinxiang, China, 453003.
| | - Guang-Rui Wan
- College of Pharmacy, Henan international joint laboratory of cardiovascular remodeling and drug intervention, Xinxiang key laboratory of vascular remodeling intervention and molecular targeted therapy drug development, Xinxiang Medical University,Xinxiang, China, 453003.
| | - Peng Li
- School of Basic Medical Sciences, Department of Physiology and Pathophysiology, Sino-UK Joint Laboratory of Brain Function and Injury and Department of Physiology and Neurobiology, Xinxiang Medical University,Xinxiang, China, 453003; College of Pharmacy, Henan international joint laboratory of cardiovascular remodeling and drug intervention, Xinxiang key laboratory of vascular remodeling intervention and molecular targeted therapy drug development, Xinxiang Medical University,Xinxiang, China, 453003.
| |
Collapse
|
24
|
The Role of Mitochondrial Dynamin in Stroke. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2022; 2022:2504798. [PMID: 35571256 PMCID: PMC9106451 DOI: 10.1155/2022/2504798] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/12/2021] [Accepted: 04/17/2022] [Indexed: 11/25/2022]
Abstract
Stroke is one of the leading causes of death and disability in the world. However, the pathophysiological process of stroke is still not fully clarified. Mitochondria play an important role in promoting nerve survival and are an important drug target for the treatment of stroke. Mitochondrial dysfunction is one of the hallmarks of stroke. Mitochondria are in a state of continuous fission and fusion, which are termed as mitochondrial dynamics. Mitochondrial dynamics are very important for maintaining various functions of mitochondria. In this review, we will introduce the structure and functions of mitochondrial fission and fusion related proteins and discuss their role in the pathophysiologic process of stroke. A better understanding of mitochondrial dynamin in stroke will pave way for the development of new therapeutic options.
Collapse
|
25
|
Zong P, Lin Q, Feng J, Yue L. A Systemic Review of the Integral Role of TRPM2 in Ischemic Stroke: From Upstream Risk Factors to Ultimate Neuronal Death. Cells 2022; 11:491. [PMID: 35159300 PMCID: PMC8834171 DOI: 10.3390/cells11030491] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2021] [Revised: 01/26/2022] [Accepted: 01/29/2022] [Indexed: 02/04/2023] Open
Abstract
Ischemic stroke causes a heavy health burden worldwide, with over 10 million new cases every year. Despite the high prevalence and mortality rate of ischemic stroke, the underlying molecular mechanisms for the common etiological factors of ischemic stroke and ischemic stroke itself remain unclear, which results in insufficient preventive strategies and ineffective treatments for this devastating disease. In this review, we demonstrate that transient receptor potential cation channel, subfamily M, member 2 (TRPM2), a non-selective ion channel activated by oxidative stress, is actively involved in all the important steps in the etiology and pathology of ischemic stroke. TRPM2 could be a promising target in screening more effective prophylactic strategies and therapeutic medications for ischemic stroke.
Collapse
Affiliation(s)
- Pengyu Zong
- Department of Cell Biology, Calhoun Cardiology Center, University of Connecticut School of Medicine (UConnHealth), Farmington, CT 06030, USA; (P.Z.); (J.F.)
| | - Qiaoshan Lin
- Department of Ecology and Evolutionary Biology, University of Connecticut, Storrs, CT 06269, USA;
| | - Jianlin Feng
- Department of Cell Biology, Calhoun Cardiology Center, University of Connecticut School of Medicine (UConnHealth), Farmington, CT 06030, USA; (P.Z.); (J.F.)
| | - Lixia Yue
- Department of Cell Biology, Calhoun Cardiology Center, University of Connecticut School of Medicine (UConnHealth), Farmington, CT 06030, USA; (P.Z.); (J.F.)
| |
Collapse
|
26
|
Vaidya B, Kaur H, Thapak P, Sharma SS, Singh JN. Pharmacological Modulation of TRPM2 Channels via PARP Pathway Leads to Neuroprotection in MPTP-induced Parkinson's Disease in Sprague Dawley Rats. Mol Neurobiol 2022; 59:1528-1542. [PMID: 34997907 DOI: 10.1007/s12035-021-02711-4] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2021] [Accepted: 12/21/2021] [Indexed: 12/13/2022]
Abstract
Transient receptor potential melastatin-2 (TRPM2) channels are cation channels activated by oxidative stress and ADP-ribose (ADPR). Role of TRPM2 channels has been postulated in several neurological disorders, but, it has not been explored in animal models of Parkinson's disease (PD). Thus, the role of TRPM2 and its associated poly (ADPR) polymerase (PARP) signaling pathways were investigated in the 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP)-induced PD rat model using TRPM2 inhibitor, 2-aminoethyl diphenyl borinate (2-APB), and PARP inhibitor, N-(6-Oxo-5,6-dihydrophenanthridin-2-yl)-(N,N-dimethylamino) acetamide hydrochloride (PJ-34). PD was induced by using a bilateral intranigral administration of MPTP in rats, and different parameters were evaluated. An increase in oxidative stress was observed, leading to locomotor and cognitive deficits in the PD rats. PD rats also showed an increased TRPM2 expression in the striatum and mid-brain accompanied by reduced expression of tyrosine hydroxylase (TH) in comparison to sham animals. Intraperitoneal administration of 2-APB and PJ-34 led to an improvement in the locomotor and cognitive deficits in comparison to MPTP-induced PD rats. These improvements were accompanied by a reduction in the levels of oxidative stress and an increase in TH levels in the striatum and mid-brain. In addition, these pharmacological interventions also led to a decrease in the expression of TRPM2 in PD in the striatum and mid-brain. Our results provide a rationale for the development of potent pharmacological agents targeting the TRPM2-PARP pathway to provide therapeutic benefits for the treatment of neurological diseases like PD.
Collapse
Affiliation(s)
- Bhupesh Vaidya
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research (NIPER), S.A.S. Nagar (Mohali), 160062, Punjab, India
| | - Harpinder Kaur
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research (NIPER), S.A.S. Nagar (Mohali), 160062, Punjab, India
| | - Pavan Thapak
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research (NIPER), S.A.S. Nagar (Mohali), 160062, Punjab, India
| | - Shyam Sunder Sharma
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research (NIPER), S.A.S. Nagar (Mohali), 160062, Punjab, India
| | - Jitendra Narain Singh
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research (NIPER), S.A.S. Nagar (Mohali), 160062, Punjab, India.
| |
Collapse
|
27
|
Wang Q, Liu N, Ni YS, Yang JM, Ma L, Lan XB, Wu J, Niu JG, Yu JQ. TRPM2 in ischemic stroke: Structure, molecular mechanisms, and drug intervention. Channels (Austin) 2021; 15:136-154. [PMID: 33455532 PMCID: PMC7833771 DOI: 10.1080/19336950.2020.1870088] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2020] [Revised: 12/23/2020] [Accepted: 12/23/2020] [Indexed: 01/14/2023] Open
Abstract
Ischemic stroke has a high lethality rate worldwide, and novel treatments are limited. Calcium overload is considered to be one of the mechanisms of cerebral ischemia. Transient receptor potential melastatin 2 (TRPM2) is a reactive oxygen species (ROS)-sensitive calcium channel. Cerebral ischemia-induced TRPM2 activation triggers abnormal intracellular Ca2+ accumulation and cell death, which in turn causes irreversible brain damage. Thus, TRPM2 has emerged as a new therapeutic target for ischemic stroke. This review provides data on the expression, structure, and function of TRPM2 and illustrates its cellular and molecular mechanisms in ischemic stroke. Natural and synthetic TRPM2 inhibitors (both specific and nonspecific) are also summarized. The three-dimensional protein structure of TRPM2 has been identified, and we speculate that molecular simulation techniques will be essential for developing new drugs that block TRPM2 channels. These insights about TRPM2 may be the key to find potent therapeutic approaches for the treatment of ischemic stroke.
Collapse
Affiliation(s)
- Qing Wang
- Department of Pharmacology, Ningxia Medical University, Yinchuan, China
| | - Ning Liu
- Department of Pharmacology, Ningxia Medical University, Yinchuan, China
| | - Yuan-Shu Ni
- Department of Pharmacology, Ningxia Medical University, Yinchuan, China
| | - Jia-Mei Yang
- Department of Pharmacology, Ningxia Medical University, Yinchuan, China
| | - Lin Ma
- Ningxia Key Laboratory of Craniocerebral Diseases of Ningxia Hui Autonomous Region, Ningxia Medical University, Yinchuan, China
| | - Xiao-Bing Lan
- Department of Pharmacology, Ningxia Medical University, Yinchuan, China
| | - Jing Wu
- Laboratory Animal Center, Ningxia Medical University, Yinchuan, China
| | - Jian-Guo Niu
- Ningxia Key Laboratory of Craniocerebral Diseases of Ningxia Hui Autonomous Region, Ningxia Medical University, Yinchuan, China
| | - Jian-Qiang Yu
- Department of Pharmacology, Ningxia Medical University, Yinchuan, China
- Ningxia Collaborative Innovation Center of Regional Characteristic Traditional Chinese Medicine, Ningxia Medical University, Yinchuan, Ningxia, China
| |
Collapse
|
28
|
Structural and functional basis of the selectivity filter as a gate in human TRPM2 channel. Cell Rep 2021; 37:110025. [PMID: 34788616 DOI: 10.1016/j.celrep.2021.110025] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2020] [Revised: 08/31/2021] [Accepted: 10/27/2021] [Indexed: 12/27/2022] Open
Abstract
Transient receptor potential melastatin 2 (TRPM2), a Ca2+-permeable cation channel, is gated by intracellular adenosine diphosphate ribose (ADPR), Ca2+, warm temperature, and oxidative stress. It is critically involved in physiological and pathological processes ranging from inflammation to stroke to neurodegeneration. At present, the channel's gating and ion permeation mechanisms, such as the location and identity of the selectivity filter, remain ambiguous. Here, we report the cryo-electron microscopy (cryo-EM) structure of human TRPM2 in nanodisc in the ligand-free state. Cryo-EM map-guided computational modeling and patch-clamp recording further identify a quadruple-residue motif as the ion selectivity filter, which adopts a restrictive conformation in the closed state and acts as a gate, profoundly contrasting with its widely open conformation in the Nematostella vectensis TRPM2. Our study reveals the gating of human TRPM2 by the filter and demonstrates the feasibility of using cryo-EM in conjunction with computational modeling and functional studies to garner structural information for intrinsically dynamic but functionally important domains.
Collapse
|
29
|
Zhu T, Zhu M, Qiu Y, Wu Z, Huang N, Wan G, Xu J, Song P, Wang S, Yin Y, Li P. Puerarin Alleviates Vascular Cognitive Impairment in Vascular Dementia Rats. Front Behav Neurosci 2021; 15:717008. [PMID: 34720898 PMCID: PMC8554240 DOI: 10.3389/fnbeh.2021.717008] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2021] [Accepted: 09/21/2021] [Indexed: 11/13/2022] Open
Abstract
Cerebral ischemia triggers vascular dementia (VD), which is characterized by memory loss, cognitive deficits, and vascular injury in the brain. Puerarin (Pur) represents the major isoflavone glycoside of Radix Puerariae, with verified neuroprotective activity and cardiovascular protective effects. However, whether Pur ameliorates cognitive impairment and vascular injury in rats with permanent occlusion of bilateral common carotid arteries (BCCAO) remains unknown. This work aimed to assess Pur's effects on BCCAO-induced VD and to dissect the underlying mechanisms, especially examining the function of transient receptor potential melastatin-related 2 (TRPM2) in alleviating cognitive deficits and vascular injuries. Rats with BCCAO developed VD. Pur (50, 100, and 150 mg/kg) dose-dependently attenuated the pathological changes, increased synaptic structural plasticity in the dorsal CA1 hippocampal region and decreased oxidative stress, which eventually reduced cognitive impairment and vascular injury in BCCAO rats. Notably, Pur-improved neuronal cell loss, synaptic structural plasticity, and endothelial vasorelaxation function might be mediated by the reactive oxygen species (ROS)-dependent TRPM2/NMDAR pathway, evidenced by decreased levels of ROS, malondialdehyde (MDA), Bax, Bax/Bcl2, and TRPM2, and increased levels of superoxide dismutase (SOD), Bcl2, and NR2A. In conclusion, Pur has therapeutic potential for VD, alleviating neuronal cell apoptosis and vascular injury, which may be related to the ROS-dependent TRPM2/NMDAR pathway.
Collapse
Affiliation(s)
- Tiantian Zhu
- College of Pharmacy, Xinxiang Medical University, Xinxiang, China.,Henan International Joint Laboratory of Cardiovascular Remodeling and Drug Intervention, Xinxiang, China.,Xinxiang Key Laboratory of Vascular Remodeling Intervention and Molecular Targeted Therapy Drug Development, Xinxiang, China
| | - Moli Zhu
- College of Pharmacy, Xinxiang Medical University, Xinxiang, China.,Henan International Joint Laboratory of Cardiovascular Remodeling and Drug Intervention, Xinxiang, China.,Xinxiang Key Laboratory of Vascular Remodeling Intervention and Molecular Targeted Therapy Drug Development, Xinxiang, China
| | - Yue Qiu
- College of Pharmacy, Xinxiang Medical University, Xinxiang, China.,Henan International Joint Laboratory of Cardiovascular Remodeling and Drug Intervention, Xinxiang, China.,Xinxiang Key Laboratory of Vascular Remodeling Intervention and Molecular Targeted Therapy Drug Development, Xinxiang, China
| | - Zeqing Wu
- College of Pharmacy, Xinxiang Medical University, Xinxiang, China.,Henan International Joint Laboratory of Cardiovascular Remodeling and Drug Intervention, Xinxiang, China.,Xinxiang Key Laboratory of Vascular Remodeling Intervention and Molecular Targeted Therapy Drug Development, Xinxiang, China
| | - Ning Huang
- College of Pharmacy, Xinxiang Medical University, Xinxiang, China.,Henan International Joint Laboratory of Cardiovascular Remodeling and Drug Intervention, Xinxiang, China.,Xinxiang Key Laboratory of Vascular Remodeling Intervention and Molecular Targeted Therapy Drug Development, Xinxiang, China
| | - Guangrui Wan
- College of Pharmacy, Xinxiang Medical University, Xinxiang, China.,Henan International Joint Laboratory of Cardiovascular Remodeling and Drug Intervention, Xinxiang, China.,Xinxiang Key Laboratory of Vascular Remodeling Intervention and Molecular Targeted Therapy Drug Development, Xinxiang, China
| | - Jian Xu
- College of Pharmacy, Xinxiang Medical University, Xinxiang, China.,Henan International Joint Laboratory of Cardiovascular Remodeling and Drug Intervention, Xinxiang, China.,Xinxiang Key Laboratory of Vascular Remodeling Intervention and Molecular Targeted Therapy Drug Development, Xinxiang, China
| | - Ping Song
- College of Pharmacy, Xinxiang Medical University, Xinxiang, China.,Henan International Joint Laboratory of Cardiovascular Remodeling and Drug Intervention, Xinxiang, China.,Xinxiang Key Laboratory of Vascular Remodeling Intervention and Molecular Targeted Therapy Drug Development, Xinxiang, China
| | - Shuangxi Wang
- College of Pharmacy, Xinxiang Medical University, Xinxiang, China.,Henan International Joint Laboratory of Cardiovascular Remodeling and Drug Intervention, Xinxiang, China.,Xinxiang Key Laboratory of Vascular Remodeling Intervention and Molecular Targeted Therapy Drug Development, Xinxiang, China
| | - Yaling Yin
- Henan International Joint Laboratory of Cardiovascular Remodeling and Drug Intervention, Xinxiang, China.,Xinxiang Key Laboratory of Vascular Remodeling Intervention and Molecular Targeted Therapy Drug Development, Xinxiang, China.,School of Basic Medical Sciences, Xinxiang Medical University, Xinxiang, China
| | - Peng Li
- College of Pharmacy, Xinxiang Medical University, Xinxiang, China.,Henan International Joint Laboratory of Cardiovascular Remodeling and Drug Intervention, Xinxiang, China.,Xinxiang Key Laboratory of Vascular Remodeling Intervention and Molecular Targeted Therapy Drug Development, Xinxiang, China
| |
Collapse
|
30
|
Deficiency of ROS-Activated TRPM2 Channel Protects Neurons from Cerebral Ischemia-Reperfusion Injury through Upregulating Autophagy. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2021; 2021:7356266. [PMID: 34367466 PMCID: PMC8337124 DOI: 10.1155/2021/7356266] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/18/2021] [Accepted: 06/29/2021] [Indexed: 12/16/2022]
Abstract
Cerebral ischemia-reperfusion (I-R) transiently increased autophagy by producing excessively reactive oxygen species (ROS); on the other hand, activated autophagy would remove ROS-damaged mitochondria and proteins, which led to cell survival. However, the regulation mechanism of autophagy activity during cerebral I-R is still unclear. In this study, we found that deficiency of the TRPM2 channel which is a ROS sensor significantly decreased I-R-induced neuronal damage. I-R transiently increased autophagy activity both in vitro and in vivo. More importantly, TRPM2 deficiency decreased I-R-induced neurological deficit score and infarct volume. Interestingly, our results indicated that TRPM2 deficiency could further activate AMPK rather than Beclin1 activity, suggesting that TRPM2 inhibits autophagy by regulating the AMPK/mTOR pathway in I-R. In conclusion, our study reveals that ROS-activated TRPM2 inhibits autophagy by downregulating the AMPK/mTOR pathway, which results in neuronal death induced by cerebral I-R, further supporting that TRPM2 might be a potential drug target for cerebral ischemic injury therapy.
Collapse
|
31
|
Abstract
The transient receptor potential (TRP) channel superfamily consists of a large group of non-selective cation channels that serve as cellular sensors for a wide spectrum of physical and environmental stimuli. The 28 mammalian TRPs, categorized into six subfamilies, including TRPC (canonical), TRPV (vanilloid), TRPM (melastatin), TRPA (ankyrin), TRPML (mucolipin) and TRPP (polycystin), are widely expressed in different cells and tissues. TRPs exhibit a variety of unique features that not only distinguish them from other superfamilies of ion channels, but also confer diverse physiological functions. Located at the plasma membrane or in the membranes of intracellular organelles, TRPs are the cellular safeguards that sense various cell stresses and environmental stimuli and translate this information into responses at the organismal level. Loss- or gain-of-function mutations of TRPs cause inherited diseases and pathologies in different physiological systems, whereas up- or down-regulation of TRPs is associated with acquired human disorders. In this Cell Science at a Glance article and the accompanying poster, we briefly summarize the history of the discovery of TRPs, their unique features, recent advances in the understanding of TRP activation mechanisms, the structural basis of TRP Ca2+ selectivity and ligand binding, as well as potential roles in mammalian physiology and pathology.
Collapse
Affiliation(s)
- Lixia Yue
- Calhoun Cardiology Center, Department of Cell Biology, University of Connecticut School of Medicine (UConn Health), Farmington, CT 06030, USA
| | - Haoxing Xu
- Department of Molecular, Cellular, and Developmental Biology, University of Michigan, Ann Arbor, MI 48109, USA
| |
Collapse
|
32
|
Ying Y, Jiang P. Research progress on transient receptor potential melastatin 2 channel in nervous system diseases. Zhejiang Da Xue Xue Bao Yi Xue Ban 2021; 50:267-276. [PMID: 34137233 PMCID: PMC8710270 DOI: 10.3724/zdxbyxb-2021-0110] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2020] [Accepted: 01/30/2021] [Indexed: 11/25/2022]
Abstract
Transient receptor potential M2 (TRPM2) ion channel is a non-selective cationic channel that can permeate calcium ions, and plays an important role in neuroinflammation, ischemic reperfusion brain injury, neurodegenerative disease, neuropathic pain, epilepsy and other neurological diseases. In ischemic reperfusion brain injury, TRPM2 mediates neuronal death by modulating the different subunits of glutamate N-methyl-D-aspartic acid receptor in response to calcium/zinc signal. In Alzheimer's disease, TRPM2 is activated by reactive oxygen species generated by β-amyloid peptide to form a malignant positive feedback loop that induces neuronal death and is involved in the pathological process of glial cells by promoting inflammatory response and oxidative stress. In epilepsy, the TRPM2-knockout alleviates epilepsy induced neuronal degeneration by inhibiting autophagy and apoptosis related proteins. The roles of TRPM2 channel in the pathogenesis of various central nervous system diseases and its potential drug development and clinical application prospects are summarized in this review.
Collapse
|
33
|
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: 3.5] [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
|
34
|
TRPM2 channel in oxidative stress-induced mitochondrial dysfunction and apoptotic cell death. ADVANCES IN PROTEIN CHEMISTRY AND STRUCTURAL BIOLOGY 2021; 125:51-72. [PMID: 33931144 DOI: 10.1016/bs.apcsb.2020.12.001] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
Abstract
Mitochondria, conserved intracellular organelles best known as the powerhouse of cells for generating ATP, play an important role in apoptosis. Oxidative stress can induce mitochondrial dysfunction and activate mitochondria-mediated apoptotic cell death. TRPM2 is a Ca2+-permeable cation channel that is activated by pathologically relevant concentrations of reactive oxygen species (ROS) and one of its well-recognized roles is to confer susceptibility to ROS-induced cell death. Increasing evidence from recent studies supports TRPM2 channel-mediated cell death as an important cellular mechanism linking miscellaneous oxidative stress-inducing pathological factors to associated diseased conditions. In this chapter, we will discuss the role of the TRPM2 channel in neurons in the brain and pancreatic β-cells in mediating mitochondrial dysfunction and cell death, focusing mainly on apoptotic cell death, that are induced by pathological stimuli implicated in the pathogenesis of neurodegenerative diseases, ischemic stroke and diabetes.
Collapse
|
35
|
Krall RF, Tzounopoulos T, Aizenman E. The Function and Regulation of Zinc in the Brain. Neuroscience 2021; 457:235-258. [PMID: 33460731 DOI: 10.1016/j.neuroscience.2021.01.010] [Citation(s) in RCA: 73] [Impact Index Per Article: 18.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2020] [Revised: 01/05/2021] [Accepted: 01/08/2021] [Indexed: 12/31/2022]
Abstract
Nearly sixty years ago Fredrich Timm developed a histochemical technique that revealed a rich reserve of free zinc in distinct regions of the brain. Subsequent electron microscopy studies in Timm- stained brain tissue found that this "labile" pool of cellular zinc was highly concentrated at synaptic boutons, hinting a possible role for the metal in synaptic transmission. Although evidence for activity-dependent synaptic release of zinc would not be reported for another twenty years, these initial findings spurred decades of research into zinc's role in neuronal function and revealed a diverse array of signaling cascades triggered or regulated by the metal. Here, we delve into our current understanding of the many roles zinc plays in the brain, from influencing neurotransmission and sensory processing, to activating both pro-survival and pro-death neuronal signaling pathways. Moreover, we detail the many mechanisms that tightly regulate cellular zinc levels, including metal binding proteins and a large array of zinc transporters.
Collapse
Affiliation(s)
- Rebecca F Krall
- Department of Neurobiology, University of Pittsburgh School of Medicine, USA; Department of Otolaryngology, University of Pittsburgh School of Medicine, USA; Pittsburgh Hearing Research Center, University of Pittsburgh School of Medicine, USA; Pittsburgh Institute for Neurodegenerative Diseases, University of Pittsburgh School of Medicine, USA
| | - Thanos Tzounopoulos
- Department of Otolaryngology, University of Pittsburgh School of Medicine, USA; Pittsburgh Hearing Research Center, University of Pittsburgh School of Medicine, USA.
| | - Elias Aizenman
- Department of Neurobiology, University of Pittsburgh School of Medicine, USA; Pittsburgh Hearing Research Center, University of Pittsburgh School of Medicine, USA; Pittsburgh Institute for Neurodegenerative Diseases, University of Pittsburgh School of Medicine, USA.
| |
Collapse
|
36
|
The role of diurnal fluctuations in excitatory amino acid carrier 1 levels in post-ischemic hippocampal Zn 2+ accumulation. Exp Neurol 2020; 336:113538. [PMID: 33253705 DOI: 10.1016/j.expneurol.2020.113538] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2020] [Revised: 11/04/2020] [Accepted: 11/22/2020] [Indexed: 02/06/2023]
Abstract
Accumulating evidence indicates time-of-day variations in ischemic neuronal injury. Under ischemic conditions, Zn2+ is massively released from hippocampal glutamatergic neurons, and intracellular Zn2+ accumulation results in neuron death. Notably, excitatory amino acid carrier 1 (EAAC1), known as a cysteine transporter, is involved in Zn2+ homeostasis, and its expressions exhibit a diurnal fluctuation. This study aimed to investigate whether time of day of an ischemic insult affects Zn2+ accumulation and neuronal injury and determine whether altered Zn2+ accumulation is modulated by EAAC1 diurnal fluctuation in the hippocampus in a mouse model of ischemic stroke. Mice subjected to transient global ischemia for 40 min at Zeitgeber time 18 (ZT18) (23:00) exhibited reduced Zn2+ accumulation and neuronal death in the hilar region of the hippocampus compared to those at ZT4 (09:00). The EAAC1 protein expression in the hippocampus was increased at ZT18 relative to ZT4. Intracerebroventricular injection of a non-selective excitatory amino acid transporter inhibitor, DL-threo-β-benzyloxyaspartate, or a selective EAAC1 inhibitor, L-aspartic acid β-hydroxamate, increased ischemia-induced Zn2+ accumulation and neuronal death in the hilus at ZT18. These findings suggest that ischemia-induced Zn2+ accumulation displays circadian fluctuations through diurnal variations in EAAC1 expressions and affects susceptibility to ischemic neuronal injury in the hippocampal hilar region.
Collapse
|
37
|
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: 12] [Impact Index Per Article: 2.4] [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
|
38
|
Malko P, Jiang LH. TRPM2 channel-mediated cell death: An important mechanism linking oxidative stress-inducing pathological factors to associated pathological conditions. Redox Biol 2020; 37:101755. [PMID: 33130440 PMCID: PMC7600390 DOI: 10.1016/j.redox.2020.101755] [Citation(s) in RCA: 87] [Impact Index Per Article: 17.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2020] [Revised: 08/17/2020] [Accepted: 10/08/2020] [Indexed: 12/26/2022] Open
Abstract
Oxidative stress resulting from the accumulation of high levels of reactive oxygen species is a salient feature of, and a well-recognised pathological factor for, diverse pathologies. One common mechanism for oxidative stress damage is via the disruption of intracellular ion homeostasis to induce cell death. TRPM2 is a non-selective Ca2+-permeable cation channel with a wide distribution throughout the body and is highly sensitive to activation by oxidative stress. Recent studies have collected abundant evidence to show its important role in mediating cell death induced by miscellaneous oxidative stress-inducing pathological factors, both endogenous and exogenous, including ischemia/reperfusion and the neurotoxicants amyloid-β peptides and MPTP/MPP+ that cause neuronal demise in the brain, myocardial ischemia/reperfusion, proinflammatory mediators that disrupt endothelial function, diabetogenic agent streptozotocin and diabetes risk factor free fatty acids that induce loss of pancreatic β-cells, bile acids that damage pancreatic acinar cells, renal ischemia/reperfusion and albuminuria that are detrimental to kidney cells, acetaminophen that triggers hepatocyte death, and nanoparticles that injure pericytes. Studies have also shed light on the signalling mechanisms by which these pathological factors activate the TRPM2 channel to alter intracellular ion homeostasis leading to aberrant initiation of various cell death pathways. TRPM2-mediated cell death thus emerges as an important mechanism in the pathogenesis of conditions including ischemic stroke, neurodegenerative diseases, cardiovascular diseases, diabetes, pancreatitis, chronic kidney disease, liver damage and neurovascular injury. These findings raise the exciting perspective of targeting the TRPM2 channel as a novel therapeutic strategy to treat such oxidative stress-associated diseases.
Collapse
Affiliation(s)
- Philippa Malko
- School of Biomedical Sciences, Faculty of Biological Sciences, University of Leeds, UK
| | - Lin-Hua Jiang
- Sino-UK Joint Laboratory of Brain Function and Injury of Henan Province and Department of Physiology and Pathophysiology, Xinxiang Medical University, PR China; School of Biomedical Sciences, Faculty of Biological Sciences, University of Leeds, UK.
| |
Collapse
|
39
|
Transient Receptor Potential Melastatin 2 (TRPM2) Inhibition by Antioxidant, N-Acetyl-l-Cysteine, Reduces Global Cerebral Ischemia-Induced Neuronal Death. Int J Mol Sci 2020; 21:ijms21176026. [PMID: 32825703 PMCID: PMC7504640 DOI: 10.3390/ijms21176026] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2020] [Revised: 08/18/2020] [Accepted: 08/20/2020] [Indexed: 12/14/2022] Open
Abstract
A variety of pathogenic mechanisms, such as cytoplasmic calcium/zinc influx, reactive oxygen species production, and ionic imbalance, have been suggested to play a role in cerebral ischemia induced neurodegeneration. During the ischemic state that occurs after stroke or heart attack, it is observed that vesicular zinc can be released into the synaptic cleft, and then translocated into the cytoplasm via various cation channels. Transient receptor potential melastatin 2 (TRPM2) is highly distributed in the central nervous system and has high sensitivity to oxidative damage. Several previous studies have shown that TRPM2 channel activation contributes to neuroinflammation and neurodegeneration cascades. Therefore, we examined whether anti-oxidant treatment, such as with N-acetyl-l-cysteine (NAC), provides neuroprotection via regulation of TRPM2, following global cerebral ischemia (GCI). Experimental animals were then immediately injected with NAC (150 mg/kg/day) for 3 and 7 days, before sacrifice. We demonstrated that NAC administration reduced activation of GCI-induced neuronal death cascades, such as lipid peroxidation, microglia and astroglia activation, free zinc accumulation, and TRPM2 over-activation. Therefore, modulation of the TRPM2 channel can be a potential therapeutic target to prevent ischemia-induced neuronal death.
Collapse
|
40
|
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: 9.4] [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
|
41
|
Wang M, Li J, Dong S, Cai X, Simaiti A, Yang X, Zhu X, Luo J, Jiang LH, Du B, Yu P, Yang W. Silica nanoparticles induce lung inflammation in mice via ROS/PARP/TRPM2 signaling-mediated lysosome impairment and autophagy dysfunction. Part Fibre Toxicol 2020; 17:23. [PMID: 32513195 PMCID: PMC7281956 DOI: 10.1186/s12989-020-00353-3] [Citation(s) in RCA: 104] [Impact Index Per Article: 20.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2019] [Accepted: 05/26/2020] [Indexed: 01/26/2023] Open
Abstract
Background Wide applications of nanoparticles (NPs) have raised increasing concerns about safety to humans. Oxidative stress and inflammation are extensively investigated as mechanisms for NPs-induced toxicity. Autophagy and lysosomal dysfunction are emerging molecular mechanisms. Inhalation is one of the main pathways of exposing humans to NPs, which has been reported to induce severe pulmonary inflammation. However, the underlying mechanisms and, more specifically, the interplays of above-mentioned mechanisms in NPs-induced pulmonary inflammation are still largely obscure. Considered that NPs exposure in modern society is often unavoidable, it is highly desirable to develop effective strategies that could help to prevent nanomaterials-induced pulmonary inflammation. Results Pulmonary inflammation induced by intratracheal instillation of silica nanoparticles (SiNPs) in C57BL/6 mice was prevented by PJ34, a poly (ADP-ribose) polymerase (PARP) inhibitor. In human lung bronchial epithelial (BEAS-2B) cells, exposure to SiNPs reduced cell viability, and induced ROS generation, impairment in lysosome function and autophagic flux. Inhibition of ROS generation, PARP and TRPM2 channel suppressed SiNPs-induced lysosome impairment and autophagy dysfunction and consequent inflammatory responses. Consistently, SiNPs-induced pulmonary inflammation was prevented in TRPM2 deficient mice. Conclusion The ROS/PARP/TRPM2 signaling is critical in SiNPs-induced pulmonary inflammation, providing novel mechanistic insights into NPs-induced lung injury. Our study identifies TRPM2 channel as a new target for the development of preventive and therapeutic strategies to mitigate nanomaterials-induced lung inflammation. Graphical abstract ![]()
Collapse
Affiliation(s)
- Mingxiang Wang
- Department of Toxicology, and Department of Medical Oncology of Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, P. R. China
| | - Jin Li
- Department of Toxicology, and Department of Medical Oncology of Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, P. R. China
| | - Shunni Dong
- MOE Key Laboratory of Macromolecular Synthesis and Functionalization, Department of Polymer Science & Engineering, Zhejiang University, Hangzhou, China
| | - Xiaobo Cai
- Department of Toxicology, and Department of Medical Oncology of Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, P. R. China.,Department of Biophysics, and Department of Neurosurgery of the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, P. R. China
| | - Aili Simaiti
- Department of Toxicology, and Department of Medical Oncology of Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, P. R. China
| | - Xin Yang
- Department of Toxicology, and Department of Medical Oncology of Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, P. R. China
| | - Xinqiang Zhu
- Department of Toxicology, and Department of Medical Oncology of Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, P. R. China.,The Fourth Affiliated Hospital, Zhejiang University School of Medicine, Yiwu, P. R. China
| | - Jianhong Luo
- Institute of Neuroscience, NHC and CAMS Key Laboratory of Medical Neurobiology, Zhejiang University School of Medicine, Hangzhou, P. R. China
| | - Lin-Hua Jiang
- Sino-UK Joint Laboratory of Brain Function and Injury and Department of Physiology and Neurobiology, Xinxiang Medical University, Xinxiang, P. R. China.,School of Biomedical Sciences, Faculty of Biological Sciences, University of Leeds, Leeds, LS2 9JT, UK
| | - Binyang Du
- MOE Key Laboratory of Macromolecular Synthesis and Functionalization, Department of Polymer Science & Engineering, Zhejiang University, Hangzhou, China.
| | - Peilin Yu
- Department of Toxicology, and Department of Medical Oncology of Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, P. R. China.
| | - Wei Yang
- Department of Biophysics, and Department of Neurosurgery of the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, P. R. China.
| |
Collapse
|
42
|
Mai C, Mankoo H, Wei L, An X, Li C, Li D, Jiang LH. TRPM2 channel: A novel target for alleviating ischaemia-reperfusion, chronic cerebral hypo-perfusion and neonatal hypoxic-ischaemic brain damage. J Cell Mol Med 2019; 24:4-12. [PMID: 31568632 PMCID: PMC6933339 DOI: 10.1111/jcmm.14679] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2018] [Revised: 08/10/2019] [Accepted: 08/26/2019] [Indexed: 12/14/2022] Open
Abstract
The transient receptor potential melastatin-related 2 (TRPM2) channel, a reactive oxygen species (ROS)-sensitive cation channel, has been well recognized for being an important and common mechanism that confers the susceptibility to ROS-induced cell death. An elevated level of ROS is a salient feature of ischaemia-reperfusion, chronic cerebral hypo-perfusion and neonatal hypoxia-ischaemia. The TRPM2 channel is expressed in hippocampus, cortex and striatum, the brain regions that are critical for cognitive functions. In this review, we examine the recent studies that combine pharmacological and/or genetic interventions with using in vitro and in vivo models to demonstrate a crucial role of the TRPM2 channel in brain damage by ischaemia-reperfusion, chronic cerebral hypo-perfusion and neonatal hypoxic-ischaemia. We also discuss the current understanding of the underlying TRPM2-dependent cellular and molecular mechanisms. These new findings lead to the hypothesis of targeting the TRPM2 channel as a potential novel therapeutic strategy to alleviate brain damage and cognitive dysfunction caused by these conditions.
Collapse
Affiliation(s)
- Chendi Mai
- Sino-UK Joint Laboratory of Brian Function and Injury of Henan Province and Department of Physiology and Neurobiology, Xinxiang Medical University, Xinxiang, China.,Sanquan College of Xinxiang Medical University, Xinxiang, China
| | - Harneet Mankoo
- School of Biomedical Sciences, Faculty of Biological Sciences, University of Leeds, Leeds, UK
| | - Linyu Wei
- Sino-UK Joint Laboratory of Brian Function and Injury of Henan Province and Department of Physiology and Neurobiology, Xinxiang Medical University, Xinxiang, China
| | - Xinfang An
- Sino-UK Joint Laboratory of Brian Function and Injury of Henan Province and Department of Physiology and Neurobiology, Xinxiang Medical University, Xinxiang, China.,Xinxiang Maternal and Child Health Care Hospital, Xinxiang, China
| | - Chaokun Li
- Sino-UK Joint Laboratory of Brian Function and Injury of Henan Province and Department of Physiology and Neurobiology, Xinxiang Medical University, Xinxiang, China
| | - Dongliang Li
- Sino-UK Joint Laboratory of Brian Function and Injury of Henan Province and Department of Physiology and Neurobiology, Xinxiang Medical University, Xinxiang, China.,Sanquan College of Xinxiang Medical University, Xinxiang, China
| | - Lin-Hua Jiang
- Sino-UK Joint Laboratory of Brian Function and Injury of Henan Province and Department of Physiology and Neurobiology, Xinxiang Medical University, Xinxiang, China.,Sanquan College of Xinxiang Medical University, Xinxiang, China.,School of Biomedical Sciences, Faculty of Biological Sciences, University of Leeds, Leeds, UK
| |
Collapse
|
43
|
Locus coeruleus-CA1 projections are involved in chronic depressive stress-induced hippocampal vulnerability to transient global ischaemia. Nat Commun 2019; 10:2942. [PMID: 31270312 PMCID: PMC6610150 DOI: 10.1038/s41467-019-10795-9] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2018] [Accepted: 05/28/2019] [Indexed: 12/14/2022] Open
Abstract
Depression and transient ischaemic attack represent the common psychological and neurological diseases, respectively, and are tightly associated. However, studies of depression-affected ischaemic attack have been limited to epidemiological evidences, and the neural circuits underlying depression-modulated ischaemic injury remain unknown. Here, we find that chronic social defeat stress (CSDS) and chronic footshock stress (CFS) exacerbate CA1 neuron loss and spatial learning/memory impairment after a short transient global ischaemia (TGI) attack in mice. Whole-brain mapping of direct outputs of locus coeruleus (LC)-tyrosine hydroxylase (TH, Th:) positive neurons reveals that LC-CA1 projections are decreased in CSDS or CFS mice. Furthermore, using designer receptors exclusively activated by designer drugs (DREADDs)-based chemogenetic tools, we determine that Th:LC-CA1 circuit is necessary and sufficient for depression-induced aggravated outcomes of TGI. Collectively, we suggest that Th:LC-CA1 pathway plays a crucial role in depression-induced TGI vulnerability and offers a potential intervention for preventing depression-related transient ischaemic attack.
Collapse
|
44
|
Jiang W, Tian X, Yang P, Li J, Xiao L, Liu J, Liu C, Tan W, Tu H. Enolase1 Alleviates Cerebral Ischemia-Induced Neuronal Injury via Its Enzymatic Product Phosphoenolpyruvate. ACS Chem Neurosci 2019; 10:2877-2889. [PMID: 30943007 DOI: 10.1021/acschemneuro.9b00103] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022] Open
Abstract
Stroke is a leading cause of disability and the second leading cause of death among adults worldwide, while the mechanisms underlying neuronal death and dysfunction remain poorly understood. Here, we investigated the differential proteomic profiles of mouse brain homogenate with 3 h of middle cerebral artery occlusion (MCAO) ischemia, or sham, using Coomassie Brilliant Blue staining, followed by mass spectrometry. We identified enolase1 (ENO1), a key glycolytic enzyme, as a potential mediator of neuronal injury in MCAO ischemic model. Reverse transcription polymerase chain reaction and western blotting data showed that ENO1 was ubiquitously expressed in various tissues, distinct regions of brain, and different postnatal age. Immunohistochemical analysis revealed that ENO1 is localized in neuronal cytoplasm and dendrites. Interestingly, the expression level of ENO1 was significantly increased in the early stage, but dramatically decreased in the late stage, of cerebral ischemia in vivo. This dynamic change was consistent with our finding in cultured hippocampal neurons treated with oxygen/glucose deprivation (OGD) in vitro. Importantly, ENO1 overexpression in cultured neurons alleviated dendritic and spinal loss caused by OGD treatment. Furthermore, the enzymatic product of ENO1, phosphoenolpyruvate (PEP), was also synchronously changed along with the dynamic ENO1 level. The neuronal injury caused by OGD treatment in vitro or ischemia in vivo was mitigated by the application of PEP. Taken together, our data revealed that ENO1 plays a novel and protective role in cerebral ischemia-induced neuronal injury, highlighting a potential of ENO1 as a therapeutic target of neuronal protection from cerebral ischemia.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | - Weihong Tan
- Department of Chemistry, Department of Physiology and Functional Genomics, Center for Research at the Bio/Nano Interface, UF Health Cancer Center, UF Genetics Institute and McKnight Brain Institute University of Florida, Gainesville, Florida 32611, United States
| | - Haijun Tu
- Shenzhen Research Institute, Hunan University, Shenzhen, Guangdong 518000, China
| |
Collapse
|
45
|
Malko P, Syed Mortadza SA, McWilliam J, Jiang LH. TRPM2 Channel in Microglia as a New Player in Neuroinflammation Associated With a Spectrum of Central Nervous System Pathologies. Front Pharmacol 2019; 10:239. [PMID: 30914955 PMCID: PMC6423084 DOI: 10.3389/fphar.2019.00239] [Citation(s) in RCA: 47] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2019] [Accepted: 02/26/2019] [Indexed: 12/15/2022] Open
Abstract
Microglial cells in the central nervous system (CNS) are crucial in maintaining a healthy environment for neurons to function properly. However, aberrant microglial cell activation can lead to excessive generation of neurotoxic proinflammatory mediators and neuroinflammation, which represents a contributing factor in a wide spectrum of CNS pathologies, including ischemic stroke, traumatic brain damage, Alzheimer’s disease, Parkinson’s disease, multiple sclerosis, psychiatric disorders, autism spectrum disorders, and chronic neuropathic pain. Oxidative stress is a salient and common feature of these conditions and has been strongly implicated in microglial cell activation and neuroinflammation. The transient receptor potential melastatin-related 2 (TRPM2) channel, an oxidative stress-sensitive calcium-permeable cationic channel, is highly expressed in microglial cells. In this review, we examine the recent studies that provide evidence to support an important role for the TRPM2 channel, particularly TRPM2-mediated Ca2+ signaling, in mediating microglial cell activation, generation of proinflammatory mediators and neuroinflammation, which are of relevance to CNS pathologies. These findings lead to a growing interest in the TRPM2 channel, a new player in neuroinflammation, as a novel therapeutic target for CNS diseases.
Collapse
Affiliation(s)
- Philippa Malko
- School of Biomedical Sciences, Faculty of Biological Sciences, University of Leeds, Leeds, United Kingdom
| | - Sharifah A Syed Mortadza
- School of Biomedical Sciences, Faculty of Biological Sciences, University of Leeds, Leeds, United Kingdom.,Department of Biochemistry, Universiti Putra Malaysia, Seri Kembangan, Malaysia
| | - Joseph McWilliam
- School of Biomedical Sciences, Faculty of Biological Sciences, University of Leeds, Leeds, United Kingdom
| | - Lin-Hua Jiang
- School of Biomedical Sciences, Faculty of Biological Sciences, University of Leeds, Leeds, United Kingdom
| |
Collapse
|
46
|
Toda T, Yamamoto S, Umehara N, Mori Y, Wakamori M, Shimizu S. Protective Effects of Duloxetine against Cerebral Ischemia-Reperfusion Injury via Transient Receptor Potential Melastatin 2 Inhibition. J Pharmacol Exp Ther 2019; 368:246-254. [PMID: 30523061 DOI: 10.1124/jpet.118.253922] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2018] [Accepted: 12/03/2018] [Indexed: 03/08/2025] Open
Abstract
Activation of transient receptor potential melastatin 2 (TRPM2), an oxidative stress-sensitive Ca2+-permeable channel, contributes to the aggravation of cerebral ischemia-reperfusion (CIR) injury. Recent studies indicated that treatment with the antidepressant duloxetine for 24 hours (long term) attenuates TRPM2 activation in response to oxidative stress in neuronal cells. To examine the direct effects of antidepressants on TRPM2 activation, we examined their short-term (0-30 minutes) treatment effects on H2O2-induced TRPM2 activation in TRPM2-expressing human embryonic kidney 293 cells using the Ca2+ indicator fura-2. Duloxetine exerted the strongest inhibitory effects on TRPM2 activation among the seven antidepressants tested. These inhibitory effects appeared to be due to the inhibition of H2O2-induced TRPM2 activation via an open-channel blocking-like mechanism, because duloxetine reduced the sustained phase but not the initial phase of increases in intracellular Ca2+ concentrations. In a whole-cell patch-clamp study, duloxetine reduced the TRPM2-mediated inward current during the channel opening state. We also examined the effects of duloxetine in a mouse model of CIR injury. The administration of duloxetine to wild-type mice attenuated CIR injury, similar to that in Trpm2 knockout (KO) mice. The administration of duloxetine did not reduce CIR injury further in Trpm2 KO mice, suggesting that it exerts neuroprotective effects against CIR injury by inhibiting TRPM2 activation. Regarding drug repositioning, duloxetine may be a useful drug in reperfusion therapy for ischemic stroke because it has already been used clinically in therapeutics for several disorders, including depression.
Collapse
Affiliation(s)
- Takahiro Toda
- Division of Pharmacology, Faculty of Pharmaceutical Sciences, Teikyo Heisei University, Tokyo, Japan (T.T., S.Y., S.S.); Department of Oral Biology, Graduate School of Dentistry, Tohoku University, Sendai, Japan (N.U., M.W.); and Department of Synthetic Chemistry and Biological Chemistry, Graduate School of Engineering, Kyoto University, Kyoto, Japan (Y.M.)
| | - Shinichiro Yamamoto
- Division of Pharmacology, Faculty of Pharmaceutical Sciences, Teikyo Heisei University, Tokyo, Japan (T.T., S.Y., S.S.); Department of Oral Biology, Graduate School of Dentistry, Tohoku University, Sendai, Japan (N.U., M.W.); and Department of Synthetic Chemistry and Biological Chemistry, Graduate School of Engineering, Kyoto University, Kyoto, Japan (Y.M.)
| | - Noriko Umehara
- Division of Pharmacology, Faculty of Pharmaceutical Sciences, Teikyo Heisei University, Tokyo, Japan (T.T., S.Y., S.S.); Department of Oral Biology, Graduate School of Dentistry, Tohoku University, Sendai, Japan (N.U., M.W.); and Department of Synthetic Chemistry and Biological Chemistry, Graduate School of Engineering, Kyoto University, Kyoto, Japan (Y.M.)
| | - Yasuo Mori
- Division of Pharmacology, Faculty of Pharmaceutical Sciences, Teikyo Heisei University, Tokyo, Japan (T.T., S.Y., S.S.); Department of Oral Biology, Graduate School of Dentistry, Tohoku University, Sendai, Japan (N.U., M.W.); and Department of Synthetic Chemistry and Biological Chemistry, Graduate School of Engineering, Kyoto University, Kyoto, Japan (Y.M.)
| | - Minoru Wakamori
- Division of Pharmacology, Faculty of Pharmaceutical Sciences, Teikyo Heisei University, Tokyo, Japan (T.T., S.Y., S.S.); Department of Oral Biology, Graduate School of Dentistry, Tohoku University, Sendai, Japan (N.U., M.W.); and Department of Synthetic Chemistry and Biological Chemistry, Graduate School of Engineering, Kyoto University, Kyoto, Japan (Y.M.)
| | - Shunichi Shimizu
- Division of Pharmacology, Faculty of Pharmaceutical Sciences, Teikyo Heisei University, Tokyo, Japan (T.T., S.Y., S.S.); Department of Oral Biology, Graduate School of Dentistry, Tohoku University, Sendai, Japan (N.U., M.W.); and Department of Synthetic Chemistry and Biological Chemistry, Graduate School of Engineering, Kyoto University, Kyoto, Japan (Y.M.)
| |
Collapse
|
47
|
An X, Fu Z, Mai C, Wang W, Wei L, Li D, Li C, Jiang LH. Increasing the TRPM2 Channel Expression in Human Neuroblastoma SH-SY5Y Cells Augments the Susceptibility to ROS-Induced Cell Death. Cells 2019; 8:cells8010028. [PMID: 30625984 PMCID: PMC6356620 DOI: 10.3390/cells8010028] [Citation(s) in RCA: 31] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2018] [Revised: 12/22/2018] [Accepted: 12/30/2018] [Indexed: 12/31/2022] Open
Abstract
Human neuroblastoma SH-SY5Y cells are a widely-used human neuronal cell model in the study of neurodegeneration. A recent study shows that, 1-methyl-4-phenylpyridine ion (MPP), which selectively causes dopaminergic neuronal death leading to Parkinson’s disease-like symptoms, can reduce SH-SY5Y cell viability by inducing H2O2 generation and subsequent TRPM2 channel activation. MPP-induced cell death is enhanced by increasing the TRPM2 expression. By contrast, increasing the TRPM2 expression has also been reported to support SH-SY5Y cell survival after exposure to H2O2, leading to the suggestion of a protective role for the TRPM2 channel. To clarify the role of reactive oxygen species (ROS)-induced TRPM2 channel activation in SH-SY5Y cells, we generated a stable SH-SY5Y cell line overexpressing the human TRPM2 channel and examined cell death and cell viability after exposure to H2O2 in the wild-type and TRPM2-overexpressing SH-SY5Y cells. Exposure to H2O2 resulted in concentration-dependent cell death and reduction in cell viability in both cell types. TRPM2 overexpression remarkably augmented H2O2-induced cell death and reduction in cell viability. Furthermore, H2O2-induced cell death in both the wild-type and TRPM2-overexpressing cells was prevented by 2-APB, a TRPM2 inhibitor, and also by PJ34 and DPQ, poly(ADP-ribose) polymerase (PARP) inhibitors. Collectively, our results show that increasing the TRPM2 expression renders SH-SY5Y cells to be more susceptible to ROS-induced cell death and reinforce the notion that the TRPM2 channel plays a critical role in conferring ROS-induced cell death. It is anticipated that SH-SY5Y cells can be useful for better understanding the molecular and signaling mechanisms for ROS-induced TRPM2-mediated neurodegeneration in the pathogenesis of neurodegenerative diseases.
Collapse
Affiliation(s)
- Xinfang An
- Sino-UK Joint Laboratory for Brain Function and Injury and Department of Physiology and Neurobiology, Xinxiang Medical University, Xinxiang 453003, China.
| | - Zixing Fu
- Sino-UK Joint Laboratory for Brain Function and Injury and Department of Physiology and Neurobiology, Xinxiang Medical University, Xinxiang 453003, China.
| | - Chendi Mai
- Sino-UK Joint Laboratory for Brain Function and Injury and Department of Physiology and Neurobiology, Xinxiang Medical University, Xinxiang 453003, China.
| | - Weiming Wang
- Sino-UK Joint Laboratory for Brain Function and Injury and Department of Physiology and Neurobiology, Xinxiang Medical University, Xinxiang 453003, China.
| | - Linyu Wei
- Sino-UK Joint Laboratory for Brain Function and Injury and Department of Physiology and Neurobiology, Xinxiang Medical University, Xinxiang 453003, China.
| | - Dongliang Li
- Sino-UK Joint Laboratory for Brain Function and Injury and Department of Physiology and Neurobiology, Xinxiang Medical University, Xinxiang 453003, China.
| | - Chaokun Li
- Sino-UK Joint Laboratory for Brain Function and Injury and Department of Physiology and Neurobiology, Xinxiang Medical University, Xinxiang 453003, China.
| | - Lin-Hua Jiang
- Sino-UK Joint Laboratory for Brain Function and Injury and Department of Physiology and Neurobiology, Xinxiang Medical University, Xinxiang 453003, China.
- School of Biomedical Sciences, Faculty of Biological Sciences, University of Leeds, Leeds LS2 JT, UK.
| |
Collapse
|
48
|
The TRPM2 channel nexus from oxidative damage to Alzheimer's pathologies: An emerging novel intervention target for age-related dementia. Ageing Res Rev 2018; 47:67-79. [PMID: 30009973 DOI: 10.1016/j.arr.2018.07.002] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2018] [Revised: 07/05/2018] [Accepted: 07/09/2018] [Indexed: 12/11/2022]
Abstract
Alzheimer's disease (AD), an age-related neurodegenerative condition, is the most common cause of dementia among the elder people, but currently there is no treatment. A number of putative pathogenic events, particularly amyloid β peptide (Aβ) accumulation, are believed to be early triggers that initiate AD. However, thus far targeting Aβ generation/aggregation as the mainstay strategy of drug development has not led to effective AD-modifying therapeutics. Oxidative damage is a conspicuous feature of AD, but this remains poorly defined phenomenon and mechanistically ill understood. The TRPM2 channel has emerged as a potentially ubiquitous molecular mechanism mediating oxidative damage and thus plays a vital role in the pathogenesis and progression of diverse neurodegenerative diseases. This article will review the emerging evidence from recent studies and propose a novel 'hypothesis' that multiple TRPM2-mediated cellular and molecular mechanisms cascade Aβ and/or oxidative damage to AD pathologies. The 'hypothesis' based on these new findings discusses the prospect of considering the TRPM2 channel as a novel therapeutic target for intervening AD and age-related dementia.
Collapse
|
49
|
Li X, Jiang LH. A critical role of the transient receptor potential melastatin 2 channel in a positive feedback mechanism for reactive oxygen species-induced delayed cell death. J Cell Physiol 2018; 234:3647-3660. [PMID: 30229906 DOI: 10.1002/jcp.27134] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2018] [Accepted: 07/05/2018] [Indexed: 12/19/2022]
Abstract
Transient receptor potential melastatin 2 (TRPM2) channel activation by reactive oxygen species (ROS) plays a critical role in delayed neuronal cell death, responsible for postischemia brain damage via altering intracellular Zn2+ homeostasis, but a mechanistic understanding is still lacking. Here, we showed that H2 O2 induced neuroblastoma SH-SY5Y cell death with a significant delay, dependently of the TRPM2 channel and increased [Zn2+ ]i , and therefore used this cell model to investigate the mechanisms underlying ROS-induced TRPM2-mediated delayed cell death. H2 O2 increased concentration-dependently the [Zn2+ ]i and caused lysosomal dysfunction and Zn2+ loss and, furthermore, mitochondrial Zn2+ accumulation, fragmentation, and ROS generation. Such effects were suppressed by preventing poly(adenosine diphosphate ribose, ADPR) polymerase-1-dependent TRPM2 channel activation with PJ34 and 3,3',5,5'-tetra-tert-butyldiphenoquinone, inhibiting the TRPM2 channel with 2-aminoethoxydiphenyl borate (2-APB) and N-(p-amylcinnamoyl)anthranilic acid, or chelating Zn2+ with N,N,N,N-tetrakis(2-pyridylmethyl)-ethylenediamine (TPEN). Bafilomycin-induced lysosomal dysfunction also resulted in mitochondrial Zn2+ accumulation, fragmentation, and ROS generation that were inhibited by PJ34 or 2-APB, suggesting that these mitochondrial events are TRPM2 dependent and sequela of lysosomal dysfunction. Mitochondrial TRPM2 expression was detected and exposure to ADPR-induced Zn2+ uptake in isolated mitochondria, which was prevented by TPEN. H2 O2 -induced delayed cell death was inhibited by apocynin and diphenyleneiodonium, nicotinamide adenine dinucleotide phosphate hydrogen (NADPH) oxidase (NOX) inhibitors, GKT137831, an NOX1/4-specific inhibitor, or Gö6983, a protein kinase C (PKC) inhibitor. Moreover, inhibition of PKC/NOX prevented H2 O2 -induced ROS generation, lysosomal dysfunction and Zn2+ release, and mitochondrial Zn2+ accumulation, fragmentation and ROS generation. Collectively, these results support a critical role for the TRPM2 channel in coupling PKC/NOX-mediated ROS generation, lysosomal Zn2+ release, and mitochondrial Zn2+ accumulation, and ROS generation to form a vicious positive feedback signaling mechanism for ROS-induced delayed cell death.
Collapse
Affiliation(s)
- Xin Li
- Sino-UK Joint Laboratory of Brain Function and Injury, Xinxiang Medical University, Xinxiang, China.,Faculty of Biological Sciences, School of Biomedical Sciences, University of Leeds, Leeds, UK
| | - Lin-Hua Jiang
- Sino-UK Joint Laboratory of Brain Function and Injury, Xinxiang Medical University, Xinxiang, China.,Faculty of Biological Sciences, School of Biomedical Sciences, University of Leeds, Leeds, UK
| |
Collapse
|
50
|
Trpm2 Ablation Accelerates Protein Aggregation by Impaired ADPR and Autophagic Clearance in the Brain. Mol Neurobiol 2018; 56:3819-3832. [PMID: 30215158 PMCID: PMC6477016 DOI: 10.1007/s12035-018-1309-0] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2018] [Accepted: 08/08/2018] [Indexed: 01/10/2023]
Abstract
TRPM2 a cation channel is also known to work as an enzyme that hydrolyzes highly reactive, neurotoxic ADP-ribose (ADPR). Although ADPR is hydrolyzed by NUT9 pyrophosphatase in major organs, the enzyme is defective in the brain. The present study questions the role of TRPM2 in the catabolism of ADPR in the brain. Genetic ablation of Trpm2 results in the disruption of ADPR catabolism that leads to the accumulation of ADPR and reduction in AMP. Trpm2−/− mice elicit the reduction in autophagosome formation in the hippocampus. Trpm2−/− mice also show aggregations of proteins in the hippocampus, aberrant structural changes and neuronal connections in synapses, and neuronal degeneration. Trpm2−/− mice exhibit learning and memory impairment, enhanced neuronal intrinsic excitability, and imbalanced synaptic transmission. These results respond to long-unanswered questions regarding the potential role of the enzymatic function of TRPM2 in the brain, whose dysfunction evokes protein aggregation. In addition, the present finding answers to the conflicting reports such as neuroprotective or neurodegenerative phenotypes observed in Trpm2−/− mice.
Collapse
|