1
|
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
|
2
|
Liu N, Liang H, Hong Y, Lu X, Jin X, Li Y, Tang S, Li Y, Cao W. Gallic acid pretreatment mitigates parathyroid ischemia-reperfusion injury through signaling pathway modulation. Sci Rep 2024; 14:12971. [PMID: 38839854 PMCID: PMC11153493 DOI: 10.1038/s41598-024-63470-5] [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: 01/01/2024] [Accepted: 05/29/2024] [Indexed: 06/07/2024] Open
Abstract
Thyroid surgery often results in ischemia-reperfusion injury (IRI) to the parathyroid glands, yet the mechanisms underlying this and how to ameliorate IRI remain incompletely explored. Our study identifies a polyphenolic herbal extract-gallic acid (GA)-with antioxidative properties against IRI. Through flow cytometry and CCK8 assays, we investigate the protective effects of GA pretreatment on a parathyroid IRI model and decode its potential mechanisms via RNA-seq and bioinformatics analysis. Results reveal increased apoptosis, pronounced G1 phase arrest, and significantly reduced cell proliferation in the hypoxia/reoxygenation group compared to the hypoxia group, which GA pretreatment mitigates. RNA-seq and bioinformatics analysis indicate GA's modulation of various signaling pathways, including IL-17, AMPK, MAPK, transient receptor potential channels, cAMP, and Rap1. In summary, GA pretreatment demonstrates potential in protecting parathyroid cells from IRI by influencing various genes and signaling pathways. These findings offer a promising therapeutic strategy for hypoparathyroidism treatment.
Collapse
Affiliation(s)
- Nianqiu Liu
- Departments of Breast Surgery, Yunnan Cancer Center, The Third Affiliated Hospital of Kunming Medical University, Kunming, 650000, Yunnan, People's Republic of China
| | - Hongmin Liang
- Department of Ultrasound, The First Affiliated Hospital of Kunming Medical University, 295 Xichang Road, Kunming, 650000, Yunnan, People's Republic of China
| | - Yuan Hong
- Departments of Laboratory, Yunnan Cancer Center, The Third Affiliated Hospital of Kunming Medical University, Kunming, 650000, Yunnan, People's Republic of China
| | - Xiaokai Lu
- Departments of Ultrasound, Yunnan Cancer Center, The Third Affiliated Hospital of Kunming Medical University, Kunming, 650000, Yunnan, People's Republic of China
| | - Xin Jin
- Department of Ultrasound, The First Affiliated Hospital of Kunming Medical University, 295 Xichang Road, Kunming, 650000, Yunnan, People's Republic of China
| | - Yuting Li
- Department of Ultrasound, The First Affiliated Hospital of Kunming Medical University, 295 Xichang Road, Kunming, 650000, Yunnan, People's Republic of China
| | - Shiying Tang
- Department of Ultrasound, The First Affiliated Hospital of Kunming Medical University, 295 Xichang Road, Kunming, 650000, Yunnan, People's Republic of China
| | - Yihang Li
- Department of Ultrasound, The First Affiliated Hospital of Kunming Medical University, 295 Xichang Road, Kunming, 650000, Yunnan, People's Republic of China
| | - Weihan Cao
- Department of Ultrasound, The First Affiliated Hospital of Kunming Medical University, 295 Xichang Road, Kunming, 650000, Yunnan, People's Republic of China.
| |
Collapse
|
3
|
Chen Z, Cheng Z, Ding C, Cao T, Chen L, Wang H, Li J, Huang X. ROS-Activated TRPM2 Channel: Calcium Homeostasis in Cardiovascular/renal System and Speculation in Cardiorenal Syndrome. Cardiovasc Drugs Ther 2023:10.1007/s10557-023-07531-3. [PMID: 38108918 DOI: 10.1007/s10557-023-07531-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 11/20/2023] [Indexed: 12/19/2023]
Abstract
The transient receptor potential melastatin 2 (TRPM2) channel is a nonselective calcium channel that is sensitive to oxidative stress (OS), and is widely expressed in multiple organs, such as the heart, kidney, and brain, which is inextricably related to calcium dyshomeostasis and downstream pathological events. Due to the increasing global burden of kidney or cardiovascular diseases (CVDs), safe and efficient drugs specific to novel targets are imperatively needed. Notably, investigation of the possibility to regard the TRPM2 channel as a new therapeutic target in ROS-related CVDs or renal diseases is urgently required because the roles of the TRPM2 channel in heart or kidney diseases have not received enough attention and thus have not been fully elaborated. Therefore, we aimed to review the involvement of the TRPM2 channel in cardiovascular disorders related to kidney or typical renal diseases and attempted to speculate about TRPM2-mediated mechanisms of cardiorenal syndrome (CRS) to provide representative perspectives for future research about novel and effective therapeutic strategies.
Collapse
Affiliation(s)
- Zihan Chen
- Department of Cardiology, the Second Affiliated Hospital of Nanchang University, Nanchang, China
- Queen Mary School, Medical Department, Nanchang University, Nanchang, China
| | - Zaihua Cheng
- Department of Cardiology, the Second Affiliated Hospital of Nanchang University, Nanchang, China
| | - Congcong Ding
- Department of Cardiology, the Second Affiliated Hospital of Nanchang University, Nanchang, China
| | - Tianyu Cao
- Biological anthropology, University of California, Santa Barbara, CA, USA
| | - Ling Chen
- Department of Cardiology, the First People's Hospital of Jiujiang, Jiujiang, China
| | - Hong Wang
- Temple University Lewis Katz School of Medicine, Philadelphia, PA, USA
| | - Junpei Li
- Department of Cardiology, the Second Affiliated Hospital of Nanchang University, Nanchang, China.
| | - Xiao Huang
- Department of Cardiology, the Second Affiliated Hospital of Nanchang University, Nanchang, China.
| |
Collapse
|
4
|
Marini M, Titiz M, Souza Monteiro de Araújo D, Geppetti P, Nassini R, De Logu F. TRP Channels in Cancer: Signaling Mechanisms and Translational Approaches. Biomolecules 2023; 13:1557. [PMID: 37892239 PMCID: PMC10605459 DOI: 10.3390/biom13101557] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2023] [Revised: 10/16/2023] [Accepted: 10/19/2023] [Indexed: 10/29/2023] Open
Abstract
Ion channels play a crucial role in a wide range of biological processes, including cell cycle regulation and cancer progression. In particular, the transient receptor potential (TRP) family of channels has emerged as a promising therapeutic target due to its involvement in several stages of cancer development and dissemination. TRP channels are expressed in a large variety of cells and tissues, and by increasing cation intracellular concentration, they monitor mechanical, thermal, and chemical stimuli under physiological and pathological conditions. Some members of the TRP superfamily, namely vanilloid (TRPV), canonical (TRPC), melastatin (TRPM), and ankyrin (TRPA), have been investigated in different types of cancer, including breast, prostate, lung, and colorectal cancer. TRP channels are involved in processes such as cell proliferation, migration, invasion, angiogenesis, and drug resistance, all related to cancer progression. Some TRP channels have been mechanistically associated with the signaling of cancer pain. Understanding the cellular and molecular mechanisms by which TRP channels influence cancer provides new opportunities for the development of targeted therapeutic strategies. Selective inhibitors of TRP channels are under initial scrutiny in experimental animals as potential anti-cancer agents. In-depth knowledge of these channels and their regulatory mechanisms may lead to new therapeutic strategies for cancer treatment, providing new perspectives for the development of effective targeted therapies.
Collapse
Affiliation(s)
| | | | | | | | - Romina Nassini
- Department of Health Sciences, Clinical Pharmacology and Oncology Section, University of Florence, 50139 Florence, Italy; (M.M.); (M.T.); (D.S.M.d.A.); (P.G.); (F.D.L.)
| | | |
Collapse
|
5
|
Gianò M, Franco C, Castrezzati S, Rezzani R. Involvement of Oxidative Stress and Nutrition in the Anatomy of Orofacial Pain. Int J Mol Sci 2023; 24:13128. [PMID: 37685933 PMCID: PMC10487620 DOI: 10.3390/ijms241713128] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2023] [Revised: 08/13/2023] [Accepted: 08/17/2023] [Indexed: 09/10/2023] Open
Abstract
Pain is a very important problem of our existence, and the attempt to understand it is one the oldest challenges in the history of medicine. In this review, we summarize what has been known about pain, its pathophysiology, and neuronal transmission. We focus on orofacial pain and its classification and features, knowing that is sometimes purely subjective and not well defined. We consider the physiology of orofacial pain, evaluating the findings on the main neurotransmitters; in particular, we describe the roles of glutamate as approximately 30-80% of total peripheric neurons associated with the trigeminal ganglia are glutamatergic. Moreover, we describe the important role of oxidative stress and its association with inflammation in the etiogenesis and modulation of pain in orofacial regions. We also explore the warning and protective function of orofacial pain and the possible action of antioxidant molecules, such as melatonin, and the potential influence of nutrition and diet on its pathophysiology. Hopefully, this will provide a solid background for future studies that would allow better treatment of noxious stimuli and for opening new avenues in the management of pain.
Collapse
Affiliation(s)
- Marzia Gianò
- Anatomy and Physiopathology Division, Department of Clinical and Experimental Sciences, University of Brescia, 25123 Brescia, Italy; (M.G.); (C.F.); (S.C.)
| | - Caterina Franco
- Anatomy and Physiopathology Division, Department of Clinical and Experimental Sciences, University of Brescia, 25123 Brescia, Italy; (M.G.); (C.F.); (S.C.)
| | - Stefania Castrezzati
- Anatomy and Physiopathology Division, Department of Clinical and Experimental Sciences, University of Brescia, 25123 Brescia, Italy; (M.G.); (C.F.); (S.C.)
| | - Rita Rezzani
- Anatomy and Physiopathology Division, Department of Clinical and Experimental Sciences, University of Brescia, 25123 Brescia, Italy; (M.G.); (C.F.); (S.C.)
- Interdipartimental University Center of Research “Adaption and Regeneration of Tissues and Organs (ARTO)”, University of Brescia, 25123 Brescia, Italy
- Italian Society for the Study of Orofacial Pain (Società Italiana Studio Dolore Orofacciale—SISDO), 25123 Brescia, Italy
| |
Collapse
|
6
|
Zheng D, Long S, Xi M. Identification of TRPM2 as a Potential Therapeutic Target Associated with Immune Infiltration: A Comprehensive Pan-Cancer Analysis and Experimental Verification in Ovarian Cancer. Int J Mol Sci 2023; 24:11912. [PMID: 37569287 PMCID: PMC10418504 DOI: 10.3390/ijms241511912] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2023] [Revised: 07/13/2023] [Accepted: 07/19/2023] [Indexed: 08/13/2023] Open
Abstract
The exact role of Transient receptor potential melastatin 2 (TRPM2) in tumor progression and immunomodulation remains elusive. We comprehensively investigated the expression pattern, diagnostic value, prognostic impact, genetic and epigenetic alterations of TRPM2 in pan-cancer. Then, we explored underlying pathways associated with TRPM2 and immune-related signatures. Ovarian cancer (OV) specimens were enrolled to test the expression of TRPM2 by immunohistochemistry and RT-qPCR. OV cell A2780 transfected with shRNA targeting TRPM2 was used in subsequent experiments. TRPM2 was aberrantly expressed and associated with unfavorable prognosis across various cancers. It possesses significant diagnostic values with AUC > 0.90. TRPM2 participated in pathways mediating immunoregulation and tumorigenesis. The expression of TRPM2 was significantly correlated with tumor microenvironment scores, tumor-stemness index, macrophages infiltration, immune checkpoints, and immune-related genes. OV single-cell datasets also indicated that TRPM2 was predominantly distributed on macrophages and malignancies. The overexpressed TRPM2 in OV tissues was validated at both the mRNA and protein levels. TRPM2 expression was significantly correlated with type2 macrophage marker CD206. Knockdown of TRPM2 inhibited OV cell proliferation and promoted apoptosis. Overall, TRPM2 has relevance to an immunosuppressive tumor microenvironment by modulating macrophage. It could serve as a powerful biomarker for tumor screening and prognosis, and a potential therapeutic target for tumor treatment, especially for OV.
Collapse
Affiliation(s)
- Danxi Zheng
- Department of Gynecology and Obstetrics, West China Second University Hospital, Sichuan University, No. 20, Third Section of People’s South Road, Chengdu 610041, China;
- Key Laboratory of Birth Defects and Related Diseases of Women and Children, Sichuan University, Chengdu 610041, China
| | - Siyu Long
- Laboratory of Molecular Translational Medicine, Center for Translational Medicine, Key Laboratory of Birth Defects and Related Diseases of Women and Children, West China Second Hospital, Sichuan University, Chengdu 610041, China;
| | - Mingrong Xi
- Department of Gynecology and Obstetrics, West China Second University Hospital, Sichuan University, No. 20, Third Section of People’s South Road, Chengdu 610041, China;
| |
Collapse
|
7
|
Shin S, Gombedza FC, Awuah Boadi E, Yiu AJ, Roy SK, Bandyopadhyay BC. Reduction of TRPC1/TRPC3 mediated Ca 2+-signaling protects oxidative stress-induced COPD. Cell Signal 2023; 107:110681. [PMID: 37062436 PMCID: PMC10542863 DOI: 10.1016/j.cellsig.2023.110681] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2022] [Revised: 03/14/2023] [Accepted: 04/13/2023] [Indexed: 04/18/2023]
Abstract
Oxidative stress is a predisposing factor in Chronic Obstructive Pulmonary Disease (COPD). Specifically, pulmonary epithelial (PE) cells reduce antioxidant capacity during COPD because of the continuous production of reactive oxygen species (ROS). However, the molecular pathogenesis that governs such ROS activity is unclear. Here we show that the dysregulation of intracellular calcium concentration ([Ca2+]i) in PE cells from COPD patients, compared to the healthy PE cells, is associated with the robust functional expressions of Transient Receptor Potential Canonical (TRPC)1 and TRPC3 channels, and Ca2+ entry (SOCE) components, Stromal Interaction Molecule 1 (STIM1) and ORAI1 channels. Additionally, the elevated expression levels of fibrotic, inflammatory, oxidative, and apoptotic markers in cells from COPD patients suggest detrimental pathway activation, thereby reducing the ability of lung remodeling. To further delineate the mechanism, we used human lung epithelial cell line, A549, since the behavior of SOCE and the expression patterns of TRPC1/C3, STIM1, and ORAI1 were much like PE cells. Notably, the knockdown of TRPC1/C3 in A549 cells substantially reduced the SOCE-induced [Ca2+]i rise, and reversed the ROS-mediated oxidative, fibrotic, inflammatory, and apoptotic responses, thus confirming the role of TRPC1/C3 in SOCE driven COPD-like condition. Higher TRPC1/C3, STIM1, and ORAI1 expressions, along with a greater Ca2+ entry, via SOCE in ROS-induced A549 cells, led to the rise in oxidative, fibrotic, inflammatory, and apoptotic gene expression, specifically through the extracellular signal-regulated kinase (ERK) pathway. Abatement of TRPC1 and/or TRPC3 reduced the mobilization of [Ca2+]i and reversed apoptotic gene expression and ERK activation, signifying the involvement of TRPC1/C3. Together these data suggest that TRPC1/C3 and SOCE facilitate the COPD condition through ROS-mediated cell death, thus implicating their likely roles as potential therapeutic targets for COPD. SUMMARY: Alterations in Ca2+ signaling modalities in normal pulmonary epithelial cells exhibit COPD through oxidative stress and cellular injury, compromising repair, which was alleviated through inhibition of store-operated calcium entry. SUBJECT AREA: Calcium, ROS, Cellular signaling, lung disease.
Collapse
Affiliation(s)
- Samuel Shin
- From Calcium Signaling Laboratory, Research Service, Veterans Affairs Medical Center, 50 Irving Street, NW, Washington, DC 20422, United States of America
| | - Farai C Gombedza
- From Calcium Signaling Laboratory, Research Service, Veterans Affairs Medical Center, 50 Irving Street, NW, Washington, DC 20422, United States of America
| | - Eugenia Awuah Boadi
- From Calcium Signaling Laboratory, Research Service, Veterans Affairs Medical Center, 50 Irving Street, NW, Washington, DC 20422, United States of America
| | - Allen J Yiu
- From Calcium Signaling Laboratory, Research Service, Veterans Affairs Medical Center, 50 Irving Street, NW, Washington, DC 20422, United States of America
| | - Sanjit K Roy
- From Calcium Signaling Laboratory, Research Service, Veterans Affairs Medical Center, 50 Irving Street, NW, Washington, DC 20422, United States of America
| | - Bidhan C Bandyopadhyay
- From Calcium Signaling Laboratory, Research Service, Veterans Affairs Medical Center, 50 Irving Street, NW, Washington, DC 20422, United States of America.
| |
Collapse
|
8
|
Molot J, Sears M, Anisman H. Multiple Chemical Sensitivity: It's time to catch up to the science. Neurosci Biobehav Rev 2023; 151:105227. [PMID: 37172924 DOI: 10.1016/j.neubiorev.2023.105227] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2022] [Accepted: 05/06/2023] [Indexed: 05/15/2023]
Abstract
Multiple chemical sensitivity (MCS) is a complex medical condition associated with low dose chemical exposures. MCS is characterized by diverse features and common comorbidities, including fibromyalgia, cough hypersensitivity, asthma, and migraine, and stress/anxiety, with which the syndrome shares numerous neurobiological processes and altered functioning within diverse brain regions. Predictive factors linked to MCS comprise genetic influences, gene-environment interactions, oxidative stress, systemic inflammation, cell dysfunction, and psychosocial influences. The development of MCS may be attributed to the sensitization of transient receptor potential (TRP) receptors, notably TRPV1 and TRPA1. Capsaicin inhalation challenge studies demonstrated that TRPV1 sensitization is manifested in MCS, and functional brain imaging studies revealed that TRPV1 and TRPA1 agonists promote brain-region specific neuronal variations. Unfortunately, MCS has often been inappropriately viewed as stemming exclusively from psychological disturbances, which has fostered patients being stigmatized and ostracized, and often being denied accommodation for their disability. Evidence-based education is essential to provide appropriate support and advocacy. Greater recognition of receptor-mediated biological mechanisms should be incorporated in laws, and regulation of environmental exposures.
Collapse
Affiliation(s)
- John Molot
- Family Medicine, University of Ottawa Faculty of Medicine, Ottawa ON Canada; Ottawa Hospital Research Institute, Ottawa, ON, Canada; Department of Neuroscience, Carleton University, Ottawa Canada.
| | - Margaret Sears
- Family Medicine, University of Ottawa Faculty of Medicine, Ottawa ON Canada; Ottawa Hospital Research Institute, Ottawa, ON, Canada; Department of Neuroscience, Carleton University, Ottawa Canada.
| | - Hymie Anisman
- Family Medicine, University of Ottawa Faculty of Medicine, Ottawa ON Canada; Ottawa Hospital Research Institute, Ottawa, ON, Canada; Department of Neuroscience, Carleton University, Ottawa Canada.
| |
Collapse
|
9
|
Aydın B, Nazıroğlu M. Involvement of TRPM7 Channel on the Induction of Diabetic Neuropathic Pain in Mice: Protective Role of Selenium and Curcumin. Biol Trace Elem Res 2023; 201:2377-2395. [PMID: 36567422 DOI: 10.1007/s12011-022-03518-7] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/19/2022] [Accepted: 12/05/2022] [Indexed: 12/26/2022]
Abstract
Excessive levels of the mitochondrial reactive oxygen radical (mitSOX) and Ca2+ influx were found to cause neuropathic pain in patients with diabetes mellitus (DM). Naltriben (NLT) and mitSOX activate the transient receptor (TRP) melastatin 7 (TRPM7) channel, but antioxidants and carvacrol inhibit it. Selenium (Se) and curcumin (CRC) have been thoroughly studied for their modulator effects on streptozotocin (STZ)-induced neuropathic pain, apoptosis, and oxidative stress through the blockage of TRP channels in dorsal root ganglion (DRG) neurons. It has not yet been fully understood how Se and CRC protect against STZ-induced neuropathic pain by modulating TRPM7. Here, we assessed how Se and CRC affected the Ca2+ influx, mitSOX-mediated oxidative damage, and apoptosis in the DRGs of mice through modifying TRPM7 activity. Seven groups (control, Se, CRC, STZ, STZ + Se, STZ + CRC, and STZ + Se + CRC) were induced from the 56 male mice. We observed that the STZ-induced stimulation of TRPM7 increased mechanical neuropathic pain (von Frey), thermal neuropathic pain (hot plate), cytosolic Ca2+, TRPM7 current density, TRPM7 expression, lipid peroxidation, mitSOX, cytosolic ROS, apoptosis, caspase-3, caspase-8, and caspase-9 concentrations, whereas Se and CRC therapies diminished the alterations. The STZ-mediated decreases of DRG viability, brain glutathione, glutathione peroxidase, vitamin A, and vitamin E concentrations were also upregulated in the treatment groups by the therapies. These findings collectively imply that an imbalance of neuropathic pain, oxidative neurotoxicity, and apoptosis in the mice is caused by the STZ-mediated activation of TRPM7. However, the downregulation of TRPM7 activity caused by the injections of Se and CRC reduced the neurotoxicity and apoptosis.
Collapse
Affiliation(s)
- Bünyamin Aydın
- Division of Endocrinology and Metabolism, Department of Internal Medicine, Faculty of Medicine, Kutahya Health Sciences University, Kutahya Evliya Çelebi Training and Research Hospital, TR-64100, Kutahya, Turkey
| | - Mustafa Nazıroğlu
- Neuroscience Research Center (NOROBAM), Suleyman Demirel University, TR-32260, Isparta, Turkey.
- Drug Discovery Unit, Analyses, Innov, BSN Health, Org., Agricul., Ltd, Consult, TR-32260, Isparta, Turkey.
| |
Collapse
|
10
|
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
|
11
|
Transient receptor potential (TRP) channels in the Manila clam (Ruditapes philippinarum): Characterization and expression patterns of the TRP gene family under heat stress in Manila clams based on genome-wide identification. Gene 2023; 854:147112. [PMID: 36513188 DOI: 10.1016/j.gene.2022.147112] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2022] [Revised: 11/25/2022] [Accepted: 12/06/2022] [Indexed: 12/14/2022]
Abstract
In this study, we identified a total of 40 transient receptor potential genes (RpTRP) in Manila clam by genome-wide identification and classified them into four categories (TRPV, TRPA, TRPM, TRPC) based on gene structure and subfamily relationships. The protein length of RpTRP genes ranges from 281 amino acids to 1601 amino acids. Molecular weight and theoretical PI values range from 182.82 kDa to 32.43 kDa, respectively, with PI values between 5.17 and 9.25. By comparing the expression profiles of TRP genes during heat stress in Manila clams at different latitudes, we found that most genes in the TRP gene family were up-regulated in expression during heat challenge. Therefore, we determined that TRP genes have an important role in the heat stress of Manila clams. This work provides a basis for further studies on the molecular mechanisms of TRP-mediated heat tolerance in Manila clam and for explaining differences in heat tolerance in Manila clam at different latitudes through key differential TRP genes at the molecular level.
Collapse
|
12
|
Wang S, Wang W, Ye X. Bibliometric Analysis of Global Research on Transient Receptor Potential Vanilloid 1 in the Field of Pain. J Pain Res 2023; 16:1517-1532. [PMID: 37193358 PMCID: PMC10182809 DOI: 10.2147/jpr.s407384] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2023] [Accepted: 05/03/2023] [Indexed: 05/18/2023] Open
Abstract
Background Transient Receptor Potential Vanilloid 1 (TRPV1) is a heat-activated cation channel modulated by inflammatory mediators, which is closely related to pain and serves as a potential analgesic target. However, the bibliometric analyses summarizing TRPV1 in the field of pain are scarce. This study aims to summarize the current status of TRPV1 in pain and the potential research direction. Methods Articles regarding TRPV1 in the pain field between 2013 and 2022 were extracted from the Web of Science core collection database on 31 December 2022. Scientometric software (VOSviewer and CiteSpace 6.1.R6) were used to perform bibliometric analysis. This study provided data on the trend of the annual outputs, countries/regions, institutions, journals, authors, co-cited references and keywords. Results A total of 2462 publications related to TRPV1 in the field of pain were extracted from 2013 to 2022, which were written by 12,005 authors of 2304 institutions, 68 countries/regions in 686 journals, with 48,723 citations totally. The number of publications has grown rapidly over the past 10 years. Most publications were from the USA and China; the Seoul Natl Univ was the most active institution; Tominaga M published the most papers and Caterina MJ was the most productive co-cited author; The top-contributing journal was Pain; The most cited references was the article authored by Julius D. "Neuropathic pain", "inflammatory pain", "visceral pain" and "migraine" were the most common types of pain in this field. The mechanism of TRPV1 in pain was one of the main research directions. Conclusion This study presented an overview of the major research directions of TRPV1 in the pain field by bibliometric methods over the past decade. The results could reveal the research trends and the hotspots in the field and provide helpful information for clinical treatments of pain.
Collapse
Affiliation(s)
- Sisi Wang
- Center for Rehabilitation Medicine, Rehabilitation & Sports Medicine Research Institute of Zhejiang Province, Department of Rehabilitation Medicine, Zhejiang Provincial People’s Hospital, Affiliated People’s Hospital, Hangzhou Medical College, Hangzhou, Zhejiang, People’s Republic of China
| | - Wen Wang
- Department of Preventive Treatment Center, Quzhou Hospital of Traditional Chinese Medicine, Quzhou, Zhejiang, People’s Republic of China
| | - Xiangming Ye
- Center for Rehabilitation Medicine, Rehabilitation & Sports Medicine Research Institute of Zhejiang Province, Department of Rehabilitation Medicine, Zhejiang Provincial People’s Hospital, Affiliated People’s Hospital, Hangzhou Medical College, Hangzhou, Zhejiang, People’s Republic of China
- Correspondence: Xiangming Ye, Department of Rehabilitation Medicine, Zhejiang Provincial People’s Hospital, 158 Shangtang Road, Gongshu District, Hangzhou City, Zhejiang, People’s Republic of China, Tel +86 571 87692748, Email
| |
Collapse
|
13
|
Molot J, Sears M, Marshall LM, Bray RI. Neurological susceptibility to environmental exposures: pathophysiological mechanisms in neurodegeneration and multiple chemical sensitivity. REVIEWS ON ENVIRONMENTAL HEALTH 2022; 37:509-530. [PMID: 34529912 DOI: 10.1515/reveh-2021-0043] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/06/2021] [Accepted: 08/13/2021] [Indexed: 05/23/2023]
Abstract
The World Health Organization lists air pollution as one of the top five risks for developing chronic non-communicable disease, joining tobacco use, harmful use of alcohol, unhealthy diets and physical inactivity. This review focuses on how host defense mechanisms against adverse airborne exposures relate to the probable interacting and overlapping pathophysiological features of neurodegeneration and multiple chemical sensitivity. Significant long-term airborne exposures can contribute to oxidative stress, systemic inflammation, transient receptor subfamily vanilloid 1 (TRPV1) and subfamily ankyrin 1 (TRPA1) upregulation and sensitization, with impacts on olfactory and trigeminal nerve function, and eventual loss of brain mass. The potential for neurologic dysfunction, including decreased cognition, chronic pain and central sensitization related to airborne contaminants, can be magnified by genetic polymorphisms that result in less effective detoxification. Onset of neurodegenerative disorders is subtle, with early loss of brain mass and loss of sense of smell. Onset of MCS may be gradual following long-term low dose airborne exposures, or acute following a recognizable exposure. Upregulation of chemosensitive TRPV1 and TRPA1 polymodal receptors has been observed in patients with neurodegeneration, and chemically sensitive individuals with asthma, migraine and MCS. In people with chemical sensitivity, these receptors are also sensitized, which is defined as a reduction in the threshold and an increase in the magnitude of a response to noxious stimulation. There is likely damage to the olfactory system in neurodegeneration and trigeminal nerve hypersensitivity in MCS, with different effects on olfactory processing. The associations of low vitamin D levels and protein kinase activity seen in neurodegeneration have not been studied in MCS. Table 2 presents a summary of neurodegeneration and MCS, comparing 16 distinctive genetic, pathophysiological and clinical features associated with air pollution exposures. There is significant overlap, suggesting potential comorbidity. Canadian Health Measures Survey data indicates an overlap between neurodegeneration and MCS (p < 0.05) that suggests comorbidity, but the extent of increased susceptibility to the other condition is not established. Nevertheless, the pathways to the development of these conditions likely involve TRPV1 and TRPA1 receptors, and so it is hypothesized that manifestation of neurodegeneration and/or MCS and possibly why there is divergence may be influenced by polymorphisms of these receptors, among other factors.
Collapse
Affiliation(s)
- John Molot
- Family Medicine, University of Ottawa Faculty of Medicine, North York, ON, Canada
| | | | | | - Riina I Bray
- Family and Community Medicine, University of Toronto, Toronto, ON, Canada
| |
Collapse
|
14
|
Khanahmad H, Mirbod SM, Karimi F, Kharazinejad E, Owjfard M, Najaflu M, Tavangar M. Pathological Mechanisms Induced by TRPM2 Ion Channels Activation in Renal Ischemia-Reperfusion Injury. Mol Biol Rep 2022; 49:11071-11079. [PMID: 36104583 DOI: 10.1007/s11033-022-07836-w] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2022] [Revised: 07/26/2022] [Accepted: 08/01/2022] [Indexed: 10/14/2022]
Abstract
Renal ischemia-reperfusion (IR) injury triggers a cascade of signaling reactions involving an increase in Ca2 + charge and reactive oxygen species (ROS) levels resulting in necrosis, inflammation, apoptosis, and subsequently acute kidney injury (AKI).Transient receptor potential (TRP) channels include an essential class of Ca2+ permeable cation channels, which are segregated into six main channels: the canonical channel (TRPC), the vanilloid-related channel (TRPV), the melastatin-related channel (TRPM), the ankyrin-related channel (TRPA), the mucolipin-related channel (TRPML) and polycystin-related channel (TRPP) or polycystic kidney disease protein (PKD2). TRP channels are involved in adjusting vascular tone, vascular permeability, cell volume, proliferation, secretion, angiogenesis and apoptosis.TRPM channels include eight isoforms (TRPM1-TRPM8) and TRPM2 is the second member of this subfamily that has been expressed in various tissues and organs such as the brain, heart, kidney and lung. Renal TRPM2 channels have an important role in renal IR damage. So that TRPM2 deficient mice are resistant to renal IR injury. TRPM2 channels are triggered by several chemicals including hydrogen peroxide, Ca2+, and cyclic adenosine diphosphate (ADP) ribose (cADPR) that are generated during AKI caused by IR injury, as well as being implicated in cell death caused by oxidative stress, inflammation, and apoptosis.
Collapse
Affiliation(s)
- Hossein Khanahmad
- Department of Genetics and Molecular biology, School of Medicine, Isfahan University of medical science, Isfahan, Iran
- Department of Genetics and Molecular biology, School of Medicine, Isfahan University of Medical sciences, Isfahan, Iran, Isfahan University of Medical sciences, Isfahan, Iran
| | - Seyedeh Mahnaz Mirbod
- Resident of Cardiology, Department of cardiology, Isfahan University of Medical Science, Isfahan, Iran
- Department of Cardiology, Isfahan University of Medical Sciences, Isfahan, Iran., Isfahan University of Medical Sciences, Isfahan, Iran
| | - Farzaneh Karimi
- Behbahan Faculty of Medical Sciences, Behbahan, Iran.
- Behbahan Faculty of Medical Sciences, No.8, Shahid Zibaei Blvd. Behbahan city, Behbahan, Khozestan province, Iran.
- Department of Physiology, Behbahan Faculty of Medical Sciences, Behbahan, Iran., Behbahan Faculty of Medical Sciences, Behbahan, Iran.
| | - Ebrahim Kharazinejad
- Abadan University of Medical Sciences, Abadan, Iran
- Department of Anatomy, Abadan University of Medical Sciences, Abadan, Iran, Abadan University of Medical Sciences, Abadan , Iran
| | - Maryam Owjfard
- Clinical Neurology Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
- Shiraz University of Applied Science and Technology (UAST), Shiraz, Iran
- Clinical Neurology Research Center, Shiraz University of Medical Sciences, Shiraz, Iran. Shiraz University of Applied Science and Technology (UAST), Shiraz, Iran, Shiraz University of Applied Science and Technology (UAST), Shiraz, Iran
| | - Malihe Najaflu
- Department of Genetics and Molecular biology, School of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran
- Department of Genetics and Molecular biology, School of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Mehrsa Tavangar
- Department of Genetics and Molecular biology, School of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran
- Department of Genetics and Molecular biology, School of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran, Isfahan University of Medical Sciences, Isfahan, Iran
| |
Collapse
|
15
|
Orozco Morato E, Knight B, Nair LS. Transcriptional profiling of neuronal ion channels in dorsal root ganglion-derived immortal cell line (F-11) under different culture conditions. IN VITRO MODELS 2022; 1:385-395. [PMID: 38107764 PMCID: PMC10723754 DOI: 10.1007/s44164-022-00036-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/12/2022] [Revised: 09/29/2022] [Accepted: 10/03/2022] [Indexed: 12/19/2023]
Abstract
Pathological pain is a prevalent condition that affects majority of adults with a variety of underlying disease conditions. Current available pharmacological pain treatments have several negative and potentially life-threatening side effects associated with their long-term use. Due to the heterogeneity of pain perception and the diversity of neuronal mechanisms that contribute to pain, high-throughput screening of small molecules that may have underlying analgesic properties is essential for identifying new analgesic treatments that are both effective and safe. The F-11 hybrid immortalized cell line is one of the currently available dorsal root ganglion (DRG) cell lines used for drug screening. While F-11 cells are commonly used as analogs to primary DRG sensory neurons, they differ significantly in physiological properties. The present study investigated the impact of differentiation protocols on the expression of mature neuron ion channels and receptors in F-11 cells. Using a customized gene array of more than eighty neuronal ion channels and receptors including voltage-gated ion channels, transient receptor potential channels, and cannabinoid receptors, we assessed the following groups: control F-11 cells; F-11 cells cultured under different culture conditions, and murine DRG tissue. The expression profiles of majority of the investigated ion channels and receptors in F-11 cells were found to be lower compared to primary mouse DRG neurons. F-11 cells cultured under low serum (LSM) conditions had increased expression of several investigated targets including voltage-gated ion channels and cannabinoid receptors when compared to control F-11 cells. The study showed that the culture conditions significantly modulated the transcriptional expression of studied ion channels and receptors, and that long-term culture (21 days) may adversely affect the expression of many of the studied targets.
Collapse
Affiliation(s)
- Erick Orozco Morato
- The Connecticut Convergence Institute for Translation in Regenerative Engineering, University of Connecticut Health Center, E-7041, MC-3711, 263 Farmington Avenue, Farmington, CT 06030 USA
| | - Brittany Knight
- The Connecticut Convergence Institute for Translation in Regenerative Engineering, University of Connecticut Health Center, E-7041, MC-3711, 263 Farmington Avenue, Farmington, CT 06030 USA
| | - Lakshmi S. Nair
- The Connecticut Convergence Institute for Translation in Regenerative Engineering, University of Connecticut Health Center, E-7041, MC-3711, 263 Farmington Avenue, Farmington, CT 06030 USA
- Department of Orthopaedic Surgery, University of Connecticut Health Center, Farmington, CT 06030 USA
- Department of Biomedical Engineering, Department of Material Science and Engineering, Institute of Material Science, University of Connecticut, Storrs, CT 06269 USA
| |
Collapse
|
16
|
Li D, Wang T, Lai J, Zeng D, Chen W, Zhang X, Zhu X, Zhang G, Hu Z. Silencing TRPM2 enhanced erastin- and RSL3-induced ferroptosis in gastric cancer cells through destabilizing HIF-1α and Nrf2 proteins. Cytotechnology 2022; 74:559-577. [PMID: 36238268 PMCID: PMC9525503 DOI: 10.1007/s10616-022-00545-z] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2021] [Accepted: 07/25/2022] [Indexed: 11/03/2022] Open
Abstract
Ferroptosis is a regulated form of cell death driven by small molecules or conditions that induce lipid-based reactive oxygen species (ROS) accumulation. Cation channel transient receptor potential melastatin-2 (TRPM2) is crucial for cancer cell survival. Our bioinformatic analysis revealed that TRPM2 is associated with cellular responses to chemical stimulus and oxidative stress, implying the potential role of TRPM2 in ferroptosis. Gastric cancer cells were treated with the ferroptosis-inducer, Erastin and RSL3. siRNA transfection was used to silence TRPM2. The levels of GSH, Fe2+, ROS and lipid peroxidation, and the activity of GPx activity were evaluated by flow cytometry and spectrophotometer. The effect of TRPM2 on ubiquitination of HIF-1α and Nrf2 were evaluated by co-immunoprecipitation. Erastin and RSL3 induced the up-regulation of TRPM2 in gastric cancer cell lines, especially in SGC7901 and MGC803. These two cells also showed stronger resistance to Erastin and RSL3 than the other cell lines. TRPM2 knockdown reduced the concentration of GSH and GPx activity, but enhanced the concentration of Fe2+, ROS and lipid peroxidation, which are significant indicators of ferroptosis. Importantly, silencing TRPM2 enhanced the inhibitory effects of Erastin and RSL3 on gastric cancer cell viability, migration, and invasion. TRPM2 stabilized and finally elevated the abundance of HIF-1α and Nrf2 in SGC7901 and MGC803 cells upon Erastin and RSL3. Activation of HIF-1α impaired Erastin- and RSL3-induced ferroptosis after TRPM2 knockdown. Collectively, silencing TRPM2 enhanced Erastin- and RSL3-induced ferroptosis in gastric cancer cells through destabilizing HIF-1α and Nrf2 proteins. Supplementary Information The online version contains supplementary material available at 10.1007/s10616-022-00545-z.
Collapse
Affiliation(s)
- Dingyun Li
- Department of Gastrointestinal Surgery, Yue Bei People’s Hospital, No.133 Huimin South Road, Wujiang District, Shaoguan, 512026 Guangdong China
| | - Ting Wang
- Department of Physical Diagnosis, Yue Bei People’s Hospital, No. 133 Huimin South Road, Wujiang District, Shaoguan, 512026 Guangdong China
| | - Jiajun Lai
- Department of Gastrointestinal Surgery, Yue Bei People’s Hospital, No.133 Huimin South Road, Wujiang District, Shaoguan, 512026 Guangdong China
| | - Deqiang Zeng
- Department of Gastrointestinal Surgery, Yue Bei People’s Hospital, No.133 Huimin South Road, Wujiang District, Shaoguan, 512026 Guangdong China
| | - Weijuan Chen
- Clinical Laboratory, Yue Bei People’s Hospital, No. 133 Huimin South Road, Wujiang District, Shaoguan, 512026 Guangdong China
| | - Xiaochong Zhang
- Department of Gastrointestinal Surgery, Yue Bei People’s Hospital, No.133 Huimin South Road, Wujiang District, Shaoguan, 512026 Guangdong China
| | - Xiaofeng Zhu
- Department of Gastrointestinal Surgery, Yue Bei People’s Hospital, No.133 Huimin South Road, Wujiang District, Shaoguan, 512026 Guangdong China
| | - Guoxiong Zhang
- Department of Gastrointestinal Surgery, Yue Bei People’s Hospital, No.133 Huimin South Road, Wujiang District, Shaoguan, 512026 Guangdong China
| | - Zhiwei Hu
- Department of Gastrointestinal Surgery, Yue Bei People’s Hospital, No.133 Huimin South Road, Wujiang District, Shaoguan, 512026 Guangdong China
| |
Collapse
|
17
|
Kazandzhieva K, Mammadova-Bach E, Dietrich A, Gudermann T, Braun A. TRP channel function in platelets and megakaryocytes: basic mechanisms and pathophysiological impact. Pharmacol Ther 2022; 237:108164. [PMID: 35247518 DOI: 10.1016/j.pharmthera.2022.108164] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2021] [Revised: 01/29/2022] [Accepted: 02/28/2022] [Indexed: 12/30/2022]
Abstract
Transient receptor potential (TRP) proteins form a superfamily of cation channels that are expressed in a wide range of tissues and cell types. During the last years, great progress has been made in understanding the molecular complexity and the functions of TRP channels in diverse cellular processes, including cell proliferation, migration, adhesion and activation. The diversity of functions depends on multiple regulatory mechanisms by which TRP channels regulate Ca2+ entry mechanisms and intracellular Ca2+ dynamics, either through membrane depolarization involving cation influx or store- and receptor-operated mechanisms. Abnormal function or expression of TRP channels results in vascular pathologies, including hypertension, ischemic stroke and inflammatory disorders through effects on vascular cells, including the components of blood vessels and platelets. Moreover, some TRP family members also regulate megakaryopoiesis and platelet production, indicating a complex role of TRP channels in pathophysiological conditions. In this review, we describe potential roles of TRP channels in megakaryocytes and platelets, as well as their contribution to diseases such as thrombocytopenia, thrombosis and stroke. We also critically discuss the potential of TRP channels as possible targets for disease prevention and treatment.
Collapse
Affiliation(s)
- Kalina Kazandzhieva
- Walther-Straub-Institute for Pharmacology and Toxicology, Ludwig-Maximilians-University, Munich, Germany
| | - Elmina Mammadova-Bach
- Walther-Straub-Institute for Pharmacology and Toxicology, Ludwig-Maximilians-University, Munich, Germany; Division of Nephrology, Department of Medicine IV, Ludwig-Maximilians-University Hospital, Munich, Germany
| | - Alexander Dietrich
- Walther-Straub-Institute for Pharmacology and Toxicology, Ludwig-Maximilians-University, Munich, Germany; German Center for Lung Research (DZL), Munich, Germany
| | - Thomas Gudermann
- Walther-Straub-Institute for Pharmacology and Toxicology, Ludwig-Maximilians-University, Munich, Germany; German Center for Lung Research (DZL), Munich, Germany.
| | - Attila Braun
- Walther-Straub-Institute for Pharmacology and Toxicology, Ludwig-Maximilians-University, Munich, Germany.
| |
Collapse
|
18
|
The human ion channel TRPM2 modulates cell survival in neuroblastoma through E2F1 and FOXM1. Sci Rep 2022; 12:6311. [PMID: 35428820 PMCID: PMC9012789 DOI: 10.1038/s41598-022-10385-8] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2021] [Accepted: 04/05/2022] [Indexed: 12/15/2022] Open
Abstract
Transient receptor potential channel melastatin 2 (TRPM2) is highly expressed in cancer and has an essential function in preserving viability through maintenance of mitochondrial function and antioxidant response. Here, the role of TRPM2 in cell survival was examined in neuroblastoma cells with TRPM2 deletion with CRISPR technology. Viability was significantly decreased in TRPM2 knockout after doxorubicin treatment. RNA sequence analysis and RT-qPCR revealed reduced RNAs encoding master transcription regulators FOXM1 and E2F1/2 and downstream cell cycle targets including Cyclin B1, CDK1, PLK1, and CKS1. CHIP analysis demonstrated decreased FOXM1 binding to their promoters. Western blotting confirmed decreased expression, and increased expression of CDK inhibitor p21, a CKS1 target. In cells with TRPM2 deletion, cell cycle progression to S and G2/M phases was reduced after treatment with doxorubicin. RNA sequencing also identified decreased DNA repair proteins in cells with TRPM2 deletion after doxorubicin treatment, and DNA damage was increased. Wild type TRPM2, but not Ca2+-impermeable mutant E960D, restored live cell number and reconstituted expression of E2F1, FOXM1, and cell cycle/DNA repair proteins. FOXM1 expression alone restored viability. TRPM2 is a potential therapeutic target to reduce tumor proliferation and increase doxorubicin sensitivity through modulation of FOXM1, E2F1, and cell cycle/DNA repair proteins.
Collapse
|
19
|
Roy P, Martinelli I, Moruzzi M, Maggi F, Amantini C, Micioni Di Bonaventura MV, Cifani C, Amenta F, Tayebati SK, Tomassoni D. Ion channels alterations in the forebrain of high-fat diet fed rats. Eur J Histochem 2021; 65:3305. [PMID: 34814650 PMCID: PMC8636841 DOI: 10.4081/ejh.2021.3305] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2021] [Accepted: 10/27/2021] [Indexed: 12/22/2022] Open
Abstract
Evidence suggests that transient receptor potential (TRP) ion channels dysfunction significantly contributes to the physiopathology of metabolic and neurological disorders. Dysregulation in functions and expression in genes encoding the TRP channels cause several inherited diseases in humans (the so-called 'TRP channelopathies'), which affect the cardiovascular, renal, skeletal, and nervous systems. This study aimed to evaluate the expression of ion channels in the forebrain of rats with diet-induced obesity (DIO). DIO rats were studied after 17 weeks under a hypercaloric diet (high-fat diet, HFD) and were compared to the control rats with a standard diet (CHOW). To determine the systemic effects of HFD exposure, we examined food intake, fat mass content, fasting glycemia, insulin levels, cholesterol, and triglycerides. qRT-PCR, Western blot, and immunochemistry analysis were performed in the frontal cortex (FC) and hippocampus (HIP). After 17 weeks of HFD, DIO rats increased their body weight significantly compared to the CHOW rats. In DIO rats, TRPC1 and TRPC6 were upregulated in the HIP, while they were downregulated in the FC. In the case of TRPM2 expression, instead was increased both in the HIP and in the FC. These could be related to the increase of proteins and nucleic acid oxidation. TRPV1 and TRPV2 gene expression showed no differences both in the FC and HIP. In general, qRT-PCR analyses were confirmed by Western blot analysis. Immunohistochemical procedures highlighted the expression of the channels in the cell body of neurons and axons, particularly for the TRPC1 and TRPC6. The alterations of TRP channel expression could be related to the activation of glial cells or the neurodegenerative process presented in the brain of the DIO rat highlighted with post synaptic protein (PSD 95) alterations. The availability of suitable animal models may be useful for studying possible pharmacological treatments to counter obesity-induced brain injury. The identified changes in DIO rats may represent the first insight to characterize the neuronal alterations occurring in obesity. Further investigations are necessary to characterize the role of TRP channels in the regulation of synaptic plasticity and obesity-related cognitive decline.
Collapse
Affiliation(s)
- Proshanta Roy
- School of Biosciences and Veterinary Medicine, University of Camerino.
| | | | | | - Federica Maggi
- Department of Molecular Medicine, La Sapienza University of Rome.
| | - Consuelo Amantini
- School of Biosciences and Veterinary Medicine, University of Camerino.
| | | | | | | | | | - Daniele Tomassoni
- School of Biosciences and Veterinary Medicine, University of Camerino.
| |
Collapse
|
20
|
Hsu WL, Noda M, Yoshioka T, Ito E. A novel strategy for treating cancer: understanding the role of Ca2+ signaling from nociceptive TRP channels in regulating cancer progression. EXPLORATION OF TARGETED ANTI-TUMOR THERAPY 2021; 2:401-415. [PMID: 36045706 PMCID: PMC9400763 DOI: 10.37349/etat.2021.00053] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2021] [Accepted: 08/12/2021] [Indexed: 11/19/2022] Open
Abstract
Cancer is an aging-associated disease and caused by genomic instability that is driven by the accumulation of mutations and epimutations in the aging process. Although Ca2+ signaling, reactive oxygen species (ROS) accumulation, DNA damage response (DDR) and senescence inflammation response (SIR) are processed during genomic instability, the underlying mechanism for the cause of genomic instability and cancer development is still poorly understood and needs to be investigated. Nociceptive transient receptor potential (TRP) channels, which firstly respond to environmental stimuli, such as microbes, chemicals or physical injuries, potentiate regulation of the aging process by Ca2+ signaling. In this review, the authors provide an explanation of the dual role of nociceptive TRP channels in regulating cancer progression, initiating cancer progression by aging-induced genomic instability, and promoting malignancy by epigenetic regulation. Thus, therapeutically targeting nociceptive TRP channels seems to be a novel strategy for treating cancers.
Collapse
Affiliation(s)
- Wen-Li Hsu
- Department of Dermatology, Kaohsiung Municipal Ta-Tung Hospital, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung 80145, Taiwan; Regenerative Medicine and Cell Therapy Research Center, Kaohsiung Medical University, Kaohsiung 80708, Taiwan
| | - Mami Noda
- Laboratory of Pathophysiology, Graduate School of Pharmaceutical Sciences, Kyushu University, Fukuoka 812-8582, Japan
| | - Tohru Yoshioka
- Regenerative Medicine and Cell Therapy Research Center, Kaohsiung Medical University, Kaohsiung 80708, Taiwan; Graduate Institute of Medicine, School of Medicine, Kaohsiung Medical University, Kaohsiung 80708, Taiwan
| | - Etsuro Ito
- Graduate Institute of Medicine, School of Medicine, Kaohsiung Medical University, Kaohsiung 80708, Taiwan; Waseda Research Institute for Science and Engineering, Waseda University, Tokyo 162-8480, Japan; Department of Biology, Waseda University, Tokyo 162-8480, Japan
| |
Collapse
|
21
|
Bussulo SKD, Ferraz CR, Carvalho TT, Verri WA, Borghi SM. Redox interactions of immune cells and muscle in the regulation of exercise-induced pain and analgesia: implications on the modulation of muscle nociceptor sensory neurons. Free Radic Res 2021; 55:757-775. [PMID: 34238089 DOI: 10.1080/10715762.2021.1953696] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022]
Abstract
The mechanistic interactions among redox status of leukocytes, muscle, and exercise in pain regulation are still poorly understood and limit targeted treatment. Exercise benefits are numerous, including the treatment of chronic pain. However, unaccustomed exercise may be reported as undesirable as it may contribute to pain. The aim of the present review is to evaluate the relationship between oxidative metabolism and acute exercise-induced pain, and as to whether improved antioxidant capacity underpins the analgesic effects of regular exercise. Preclinical and clinical studies addressing relevant topics on mechanisms by which exercise modulates the nociceptive activity and how redox status can outline pain and analgesia are discussed, in sense of translating into refined outcomes. Emerging evidence points to the role of oxidative stress-induced signaling in sensitizing nociceptor sensory neurons. In response to acute exercise, there is an increase in oxidative metabolism, and consequently, pain. Instead, regular exercise can modulate redox status in favor of antioxidant capacity and repair mechanisms, which have consequently increased resistance to oxidative stress, damage, and pain. Data indicate that acute sessions of unaccustomed prolonged and/or intense exercise increase oxidative metabolism and regulate exercise-induced pain in the post-exercise recovery period. Further, evidence demonstrates regular exercise improves antioxidant status, indicating its therapeutic utility for chronic pain disorders. An improved comprehension of the role of redox status in exercise can provide helpful insights into immune-muscle communication during pain modulatory effects of exercise and support new therapeutic efforts and rationale for the promotion of exercise.
Collapse
Affiliation(s)
- Sylvia K D Bussulo
- Center for Research in Health Sciences, University of Northern Paraná, Londrina, Brazil
| | - Camila R Ferraz
- Department of Pathology, Biological Sciences Center, Rodovia Celso Garcia Cid, State University of Londrina, Londrina, Brazil
| | - Thacyana T Carvalho
- Department of Pathology, Biological Sciences Center, Rodovia Celso Garcia Cid, State University of Londrina, Londrina, Brazil
| | - Waldiceu A Verri
- Department of Pathology, Biological Sciences Center, Rodovia Celso Garcia Cid, State University of Londrina, Londrina, Brazil
| | - Sergio M Borghi
- Center for Research in Health Sciences, University of Northern Paraná, Londrina, Brazil.,Department of Pathology, Biological Sciences Center, Rodovia Celso Garcia Cid, State University of Londrina, Londrina, Brazil
| |
Collapse
|
22
|
Zhang D, Yang B, Chang SQ, Ma SS, Sun JX, Yi L, Li X, Shi HM, Jing B, Zheng YC, Zhang CL, Chen FG, Zhao GP. Protective effect of paeoniflorin on H 2O 2 induced Schwann cells injury based on network pharmacology and experimental validation. Chin J Nat Med 2021; 19:90-99. [PMID: 33641788 DOI: 10.1016/s1875-5364(21)60010-9] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2020] [Indexed: 02/07/2023]
Abstract
This study was to investigate the protective effect of paeoniflorin (PF) on hydrogen peroxide-induced injury. Firstly, "SMILES" of PF was searched in Pubchem and further was used for reverse molecular docking in Swiss Target Prediction database to obtain potential targets. Injury-related molecules were obtained from GeenCards database, and the predicted targets of PF for injury treatment were selected by Wayne diagram. For mechanism analysis, the protein-protein interactions were constructed by String, and the KEGG analysis was conducted in Webgestalt. Then, cell viability and cytotoxicity assay were established by CCK8 assay. Also, the experimental cells were allocated to control, model (200 μmol·L-1 H2O2), SB203580 10 μmol·L-1 (200 μmol·L-1 H2O2+ SB203580 10 μmol·L-1), PF 50 μmol·L-1 (200 μmol·L-1 H2O2+ PF 50 μmol·L-1), and PF 100 μmol·L-1 (200 μmol·L-1 H2O2+ PF 100 μmol·L-1) groups. We measured the intracellular ROS, Hoechst 33258 staining, cell apoptosis, the levels of Bcl-xl, Bcl-2, Caspase-3, Cleaved-caspase3, Cleaved-caspase7, TRPA1, TRPV1, and the phosphorylation expression of p38MAPK. There are 96 potential targets that may be associated with PF for injury treatment. Then, we chose the "Inflammatory mediator regulation of TRP channels" pathway for the experimental verification from the first 10 KEGG pathway. In experimental verification, H2O2 decreased the cell viability moderately (P < 0.05), and 100 μmol·L -1 PF increased the cell viability significantly (P < 0.05). Depending on the difference of intracellular ROS fluorescence intensity, PF inhibited H 2O2-induced reactive oxygen species production in Schwann cells. In Hoechst 33258 staining, PF reversed the condensed chromatin and apoptotic nuclei following H2O2 treatment. Moreover, Flow cytometry results showed that PF could substantially inhibit H2O2 induced apoptosis (P < 0.05). Pretreatment with PF obviously reduced the levels of Caspase3, Cleaved-caspase3, Cleaved-caspase7, TRPA1, TRPV1, and the phosphorylation expression of p38MAPK after H 2O2 treatment (P < 0.05), increased the levels of Bcl-2, and Bcl-xl ( P < 0.05). PF inhibited Schwann cell injury and apoptosis induced by hydrogen peroxide, which mechanism was linked to the inhibition of phosphorylation of p38MAPK.
Collapse
Affiliation(s)
- Di Zhang
- College of Traditional Chinese Medicine, Jinan University, Guangzhou 510630, China
| | - Bing Yang
- College of Traditional Chinese Medicine, Jinan University, Guangzhou 510630, China
| | - Shi-Quan Chang
- College of Traditional Chinese Medicine, Jinan University, Guangzhou 510630, China
| | - Sheng-Suo Ma
- College of Traditional Chinese Medicine, Jinan University, Guangzhou 510630, China
| | - Jian-Xin Sun
- College of Traditional Chinese Medicine, Jinan University, Guangzhou 510630, China
| | - Lin Yi
- College of Traditional Chinese Medicine, Jinan University, Guangzhou 510630, China
| | - Xing Li
- College of Traditional Chinese Medicine, Jinan University, Guangzhou 510630, China
| | - Hui-Mei Shi
- College of Traditional Chinese Medicine, Jinan University, Guangzhou 510630, China
| | - Bei Jing
- College of Traditional Chinese Medicine, Jinan University, Guangzhou 510630, China
| | - Ya-Chun Zheng
- College of Traditional Chinese Medicine, Jinan University, Guangzhou 510630, China
| | - Chun-Lan Zhang
- College of Traditional Chinese Medicine, Jinan University, Guangzhou 510630, China
| | - Feng-Guo Chen
- LiWan Hospital of Traditional Chinese Medicine, Guangzhou 510665, China
| | - Guo-Ping Zhao
- College of Traditional Chinese Medicine, Jinan University, Guangzhou 510630, China.
| |
Collapse
|
23
|
Hu F, Li M, Han F, Zhang Q, Zeng Y, Zhang W, Cheng X. Role of TRPM7 in cardiac fibrosis: A potential therapeutic target (Review). Exp Ther Med 2020; 21:173. [PMID: 33456540 PMCID: PMC7792474 DOI: 10.3892/etm.2020.9604] [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: 04/04/2020] [Accepted: 12/04/2020] [Indexed: 02/06/2023] Open
Abstract
Cardiac fibrosis is a hallmark of cardiac remodeling associated with nearly all forms of heart disease. Clinically, no effective therapeutic drugs aim to inhibit cardiac fibrosis, owing to the complex etiological heterogeneity and pathogenesis of this disease. A two-in-one protein structure, a ubiquitous expression profile and unique biophysical characteristics enable the involvement of transient receptor potential melastatin-subfamily member 7 (TRPM7) in the pathogenesis and development of fibrosis-related cardiac diseases, such as heart failure (HF), cardiomyopathies, arrhythmia and hyperaldosteronism. In response to a variety of stimuli, multiple bioactive molecules can activate TRPM7 and related signaling pathways, leading to fibroblast proliferation, differentiation and extracellular matrix production in cardiac fibroblasts. TRPM7-mediated Ca2+ signaling and TGF-β1 signaling pathways are critical for the formation of fibrosis. Accumulating evidence has demonstrated that TRPM7 is a potential pharmacological target for halting the development of fibrotic cardiac diseases. Reliable drug-like molecules for further development of high-affinity in vivo drugs targeting TRPM7 are urgently needed. The present review discusses the widespread and significant role of TRPM7 in cardiac fibrosis and focuses on its potential as a therapeutic target for alleviating heart fibrogenesis.
Collapse
Affiliation(s)
- Feng Hu
- Department of Cardiovascular Medicine, The Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi 330006, P.R. China
| | - Meiyong Li
- Department of Laboratory Medicine, The Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi 330006, P.R. China
| | - Fengyu Han
- Department of Cardiovascular Medicine, The Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi 330006, P.R. China
| | - Qing Zhang
- Department of Cardiology, The Union Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430022, P.R. China
| | - Yuhao Zeng
- Department of Medical Education, The Second Clinical Medical College of Nanchang University, Nanchang, Jiangxi 330006, P.R. China
| | - Weifang Zhang
- Department of Pharmacy, The Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi 330006, P.R. China
| | - Xiaoshu Cheng
- Department of Cardiovascular Medicine, The Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi 330006, P.R. China.,Center for Prevention and Treatment of Cardiovascular Diseases, The Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi 330006, P.R. China
| |
Collapse
|
24
|
Effects of Vitamin D on doxorubucin-induced lung injury and TRPM2 immunoreactivity in rats. JOURNAL OF SURGERY AND MEDICINE 2020. [DOI: 10.28982/josam.842133] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/14/2022] Open
|
25
|
Övey İS, Nazıroğlu M. Effects of homocysteine and memantine on oxidative stress related TRP cation channels in in-vitro model of Alzheimer’s disease. J Recept Signal Transduct Res 2020; 41:273-283. [DOI: 10.1080/10799893.2020.1806321] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/24/2022]
Affiliation(s)
- İshak Suat Övey
- Department of Physiology, School of Medicine, Alanya Alaaddin Keykubat University, Alanya, Turkey
- Department of Neuroscience, Institute of Health Sciences, Suleyman Demirel University, Isparta, Turkey
| | - Mustafa Nazıroğlu
- Department of Neuroscience, Institute of Health Sciences, Suleyman Demirel University, Isparta, Turkey
- Neuroscience Research Center, Suleyman Demirel University, Isparta, Turkey
| |
Collapse
|
26
|
Novel Therapeutic Approaches of Ion Channels and Transporters in Cancer. Rev Physiol Biochem Pharmacol 2020; 183:45-101. [PMID: 32715321 DOI: 10.1007/112_2020_28] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
The expression and function of many ion channels and transporters in cancer cells display major differences in comparison to those from healthy cells. These differences provide the cancer cells with advantages for tumor development. Accordingly, targeting ion channels and transporters have beneficial anticancer effects including inhibition of cancer cell proliferation, migration, invasion, metastasis, tumor vascularization, and chemotherapy resistance, as well as promoting apoptosis. Some of the molecular mechanisms associating ion channels and transporters with cancer include the participation of oxidative stress, immune response, metabolic pathways, drug synergism, as well as noncanonical functions of ion channels. This diversity of mechanisms offers an exciting possibility to suggest novel and more effective therapeutic approaches to fight cancer. Here, we review and discuss most of the current knowledge suggesting novel therapeutic approaches for cancer therapy targeting ion channels and transporters. The role and regulation of ion channels and transporters in cancer provide a plethora of exceptional opportunities in drug design, as well as novel and promising therapeutic approaches that may be used for the benefit of cancer patients.
Collapse
|
27
|
Özkal B, Övey İS. Selenium enhances TRPA1 channel-mediated activity of temozolomide in SH-SY5Y neuroblastoma cells. Childs Nerv Syst 2020; 36:1283-1292. [PMID: 32146544 DOI: 10.1007/s00381-020-04567-w] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/23/2019] [Accepted: 02/28/2020] [Indexed: 12/24/2022]
Abstract
PURPOSE Neuroblastoma is a malignant solid tumor that originates from the sympathetic nervous system in early childhood. Temozolomide is used for treatment in high-risk groups with low treatment response of neuroblastomas. TRPA1 channels in neuroblastoma cells are calcium permeable channels that can be activated by reactive oxygen species (ROT). In this study, we aimed to evaluate the level of activity of temozolomide and selenium in neuroblastoma cells via TRPA1 channels. METHOD Seven main groups were formed using SH-SY5Y neuroblastoma cells. The control was divided into temozolomide (TMZ) (100 μM, 24 h), TMZ+SEL+AP18, SEL (sodium selenite, 100 μM, 24 h), and SEL+AP18 groups. Intergroup calcium signaling, intracellular reactive oxygen species, caspase-3 and caspase-9, and mitochondrial depolarization analyses were performed by channel activation with TRPA1 agonist cinnamaldehyde in all groups. RESULTS Cytosolic calcium concentration, apoptosis, caspase-3 and caspase-9 activation, mitochondrial membrane depolarization, and ROT levels were higher in TMZ (p < 0.001), TMZ+SEL (p < 0.001), and SEL (p < 0.05) groups than the control group. TRPA1 was lower in TTMZ+AP18, TMZ+SEL+AP18, and SEL+AP18 groups with channel blockers than respectively TMZ, TMZ+SEL, and SEL groups without channel blockers (p < 0.05). CONCLUSION The use of selenium with temozolomide increased the apoptotic efficacy of temozolomide via TRPA1 channels on tumor cells.
Collapse
Affiliation(s)
- Birol Özkal
- Department of Neurosurgery, School of Medicine, Alanya Alaaddin Keykubat University, 07400, Alanya, Antalya, Turkey.
| | - İshak Suat Övey
- Department of Pysiology, School of Medicine, Alanya Alaaddin Keykubat University, 07400, Alanya, Antalya, Turkey
| |
Collapse
|
28
|
Transient receptor potential ion channel TRPM2 promotes AML proliferation and survival through modulation of mitochondrial function, ROS, and autophagy. Cell Death Dis 2020; 11:247. [PMID: 32312983 PMCID: PMC7170900 DOI: 10.1038/s41419-020-2454-8] [Citation(s) in RCA: 41] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2019] [Revised: 04/02/2020] [Accepted: 04/06/2020] [Indexed: 01/01/2023]
Abstract
Transient receptor potential melastatin 2 (TRPM2) ion channel has an essential function in maintaining cell survival following oxidant injury. Here, we show that TRPM2 is highly expressed in acute myeloid leukemia (AML). The role of TRPM2 in AML was studied following depletion with CRISPR/Cas9 technology in U937 cells. In in vitro experiments and in xenografts, depletion of TRPM2 in AML inhibited leukemia proliferation, and doxorubicin sensitivity was increased. Mitochondrial function including oxygen consumption rate and ATP production was reduced, impairing cellular bioenergetics. Mitochondrial membrane potential and mitochondrial calcium uptake were significantly decreased in depleted cells. Mitochondrial reactive oxygen species (ROS) were significantly increased, and Nrf2 was decreased, reducing the antioxidant response. In TRPM2-depleted cells, ULK1, Atg7, and Atg5 protein levels were decreased, leading to autophagy inhibition. Consistently, ATF4 and CREB, two master transcription factors for autophagosome biogenesis, were reduced in TRPM2-depleted cells. In addition, Atg13 and FIP200, which are known to stabilize ULK1 protein, were decreased. Reconstitution with TRPM2 fully restored proliferation, viability, and autophagy; ATF4 and CREB fully restored proliferation and viability but only partially restored autophagy. TRPM2 expression reduced the elevated ROS found in depleted cells. These data show that TRPM2 has an important role in AML proliferation and survival through regulation of key transcription factors and target genes involved in mitochondrial function, bioenergetics, the antioxidant response, and autophagy. Targeting TRPM2 may represent a novel therapeutic approach to inhibit myeloid leukemia growth and enhance susceptibility to chemotherapeutic agents through multiple pathways.
Collapse
|
29
|
Antioxidative and Anti-Inflammatory Properties of Cannabidiol. Antioxidants (Basel) 2019; 9:antiox9010021. [PMID: 31881765 PMCID: PMC7023045 DOI: 10.3390/antiox9010021] [Citation(s) in RCA: 417] [Impact Index Per Article: 69.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2019] [Revised: 12/21/2019] [Accepted: 12/23/2019] [Indexed: 12/21/2022] Open
Abstract
Cannabidiol (CBD) is one of the main pharmacologically active phytocannabinoids of Cannabis sativa L. CBD is non-psychoactive but exerts a number of beneficial pharmacological effects, including anti-inflammatory and antioxidant properties. The chemistry and pharmacology of CBD, as well as various molecular targets, including cannabinoid receptors and other components of the endocannabinoid system with which it interacts, have been extensively studied. In addition, preclinical and clinical studies have contributed to our understanding of the therapeutic potential of CBD for many diseases, including diseases associated with oxidative stress. Here, we review the main biological effects of CBD, and its synthetic derivatives, focusing on the cellular, antioxidant, and anti-inflammatory properties of CBD.
Collapse
|
30
|
The Human Transient Receptor Potential Melastatin 2 Ion Channel Modulates ROS Through Nrf2. Sci Rep 2019; 9:14132. [PMID: 31575956 PMCID: PMC6773863 DOI: 10.1038/s41598-019-50661-8] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2019] [Accepted: 09/16/2019] [Indexed: 12/20/2022] Open
Abstract
Transient receptor potential melastatin channel subfamily member 2 (TRPM2) has an essential role in protecting cell viability through modulation of oxidative stress. TRPM2 is highly expressed in cancer. When TRPM2 is inhibited, mitochondria are dysfunctional, ROS levels are increased, and cell viability is reduced. Here, the importance of NF-E2-related factor (Nrf2) in TRPM2-mediated suppression of oxidant stress was explored. In TRPM2 depleted cells, antioxidant cofactors glutathione, NADPH, and NADH were significantly reduced. Cytoplasmic and nuclear expression of Nrf2 and of IQGAP1, a modulator of Nrf2 stability regulated by intracellular calcium, were decreased. Antioxidant enzymes transcriptionally regulated by Nrf2 and involved in GSH, NADPH, and NADH generation were significantly lower including PRX1 and PRX3, GPX4, GSTP1, GCLC, and MTHFD2. The glutamine pathway leading to GSH production was suppressed, and ATP and GTP levels were impaired. Reconstitution with wild type TRPM2 or Nrf2, but not TRPM2 pore mutant E960D, rescued expression of enzymes downstream of Nrf2 and restored GSH and GTP. Cell viability, ROS, NADPH, NADH, and ATP levels were fully rescued by TRPM2 and partially by Nrf2. These data show that TRPM2 maintains cell survival following oxidative stress through modulation of antioxidant pathways and cofactors regulated by Nrf2.
Collapse
|
31
|
Miller BA, Wang J, Song J, Zhang XQ, Hirschler-Laszkiewicz I, Shanmughapriya S, Tomar D, Rajan S, Feldman AM, Madesh M, Sheu SS, Cheung JY. Trpm2 enhances physiological bioenergetics and protects against pathological oxidative cardiac injury: Role of Pyk2 phosphorylation. J Cell Physiol 2019; 234:15048-15060. [PMID: 30637731 PMCID: PMC6626587 DOI: 10.1002/jcp.28146] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2018] [Accepted: 01/03/2019] [Indexed: 01/25/2023]
Abstract
The mechanisms by which Trpm2 channels enhance mitochondrial bioenergetics and protect against oxidative stress-induced cardiac injury remain unclear. Here, the role of proline-rich tyrosine kinase 2 (Pyk2) in Trpm2 signaling is explored. Activation of Trpm2 in adult myocytes with H2 O2 resulted in 10- to 21-fold increases in Pyk2 phosphorylation in wild-type (WT) myocytes which was significantly lower (~40%) in Trpm2 knockout (KO) myocytes. Pyk2 phosphorylation was inhibited (~54%) by the Trpm2 blocker clotrimazole. Buffering Trpm2-mediated Ca2+ increase with 1,2-bis(2-aminophenoxy)ethane-N,N,N',N'-tetraacetic acid (BAPTA) resulted in significantly reduced pPyk2 in WT but not in KO myocytes, indicating Ca2+ influx through activated Trpm2 channels phosphorylated Pyk2. Part of phosphorylated Pyk2 translocated from cytosol to mitochondria which has been previously shown to augment mitochondrial Ca2+ uptake and enhance adenosine triphosphate generation. Although Trpm2-mediated Ca2+ influx phosphorylated Ca2+ -calmodulin kinase II (CaMKII), the CaMKII inhibitor KN93 did not significantly affect Pyk2 phosphorylation in H2 O2 -treated WT myocytes. After ischemia/reperfusion (I/R), Pyk2 phosphorylation and its downstream prosurvival signaling molecules (pERK1/2 and pAkt) were significantly lower in KO-I/R when compared with WT-I/R hearts. After hypoxia/reoxygenation, mitochondrial membrane potential was lower and superoxide level was higher in KO myocytes, and were restored to WT values by the mitochondria-targeted superoxide scavenger MitoTempo. Our results suggested that Ca2+ influx via tonically activated Trpm2 phosphorylated Pyk2, part of which translocated to mitochondria, resulting in better mitochondrial bioenergetics to maintain cardiac health. After I/R, Pyk2 activated prosurvival signaling molecules and prevented excessive increases in reactive oxygen species, thereby affording protection from I/R injury.
Collapse
Affiliation(s)
- Barbara A. Miller
- Department of Pediatrics, The Pennsylvania State University College of Medicine, Hershey, Pennsylvania 17033
| | - JuFang Wang
- Center of Translational Medicine, Lewis Katz School of Medicine of Temple University, Philadelphia, PA 19140
| | - Jianliang Song
- Center of Translational Medicine, Lewis Katz School of Medicine of Temple University, Philadelphia, PA 19140
| | - Xue-Qian Zhang
- Center of Translational Medicine, Lewis Katz School of Medicine of Temple University, Philadelphia, PA 19140
| | - Iwona Hirschler-Laszkiewicz
- Department of Pediatrics, The Pennsylvania State University College of Medicine, Hershey, Pennsylvania 17033
| | - Santhanam Shanmughapriya
- Center of Translational Medicine, Lewis Katz School of Medicine of Temple University, Philadelphia, PA 19140,Department of Biochemistry, Lewis Katz School of Medicine of Temple University, Philadelphia, PA 19140
| | - Dhanendra Tomar
- Center of Translational Medicine, Lewis Katz School of Medicine of Temple University, Philadelphia, PA 19140,Department of Biochemistry, Lewis Katz School of Medicine of Temple University, Philadelphia, PA 19140
| | - Sudasan Rajan
- Center of Translational Medicine, Lewis Katz School of Medicine of Temple University, Philadelphia, PA 19140,Department of Biochemistry, Lewis Katz School of Medicine of Temple University, Philadelphia, PA 19140
| | - Arthur M. Feldman
- Department of Medicine, Lewis Katz School of Medicine of Temple University, Philadelphia, PA 19140
| | - Muniswamy Madesh
- Center of Translational Medicine, Lewis Katz School of Medicine of Temple University, Philadelphia, PA 19140,Department of Biochemistry, Lewis Katz School of Medicine of Temple University, Philadelphia, PA 19140
| | - Shey-Shing Sheu
- Center for Translational Medicine, Sidney Kimmel Medical College, Thomas Jefferson University, Philadelphia, Pennsylvania 19107
| | - Joseph Y. Cheung
- Center of Translational Medicine, Lewis Katz School of Medicine of Temple University, Philadelphia, PA 19140,Department of Medicine, Lewis Katz School of Medicine of Temple University, Philadelphia, PA 19140
| |
Collapse
|
32
|
Dietz RM, Cruz-Torres I, Orfila JE, Patsos OP, Shimizu K, Chalmers N, Deng G, Tiemeier E, Quillinan N, Herson PS. Reversal of Global Ischemia-Induced Cognitive Dysfunction by Delayed Inhibition of TRPM2 Ion Channels. Transl Stroke Res 2019; 11:254-266. [PMID: 31250378 DOI: 10.1007/s12975-019-00712-z] [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] [Received: 04/23/2019] [Revised: 06/05/2019] [Accepted: 06/06/2019] [Indexed: 12/23/2022]
Abstract
Hippocampal injury and cognitive impairments are common after cardiac arrest and stroke and do not have an effective intervention despite much effort. Therefore, we developed a new approach aimed at reversing synaptic dysfunction by targeting TRPM2 channels. Cardiac arrest/cardiopulmonary resuscitation (CA/CPR) in mice was used to investigate cognitive deficits and the role of the calcium-permeable ion channel transient receptor potential-M2 (TRPM2) in ischemia-induced synaptic dysfunction. Our data indicates that absence (TRPM2-/-) or acute inhibition of TRPM2 channels with tatM2NX reduced hippocampal cell death in males only, but prevented synaptic plasticity deficits in both sexes. Remarkably, administration of tatM2NX weeks after injury reversed hippocampal plasticity and memory deficits. Finally, TRPM2-dependent activation of calcineurin-GSK3β pathway contributes to synaptic plasticity impairments. These data suggest persistent TRPM2 activity following ischemia contributes to impairments of the surviving hippocampal network and that inhibition of TRPM2 channels at chronic time points may represent a novel strategy to improve functional recovery following cerebral ischemia that is independent of neuroprotection.
Collapse
Affiliation(s)
- Robert M Dietz
- Department of Pediatrics, University of Colorado School of Medicine, Aurora, CO, USA.,Neuronal Injury & Plasticity Program, University of Colorado School of Medicine, Aurora, CO, USA
| | - Ivelisse Cruz-Torres
- Neuronal Injury & Plasticity Program, University of Colorado School of Medicine, Aurora, CO, USA.,Department of Pharmacology, University of Colorado School of Medicine, Aurora, CO, USA
| | - James E Orfila
- Neuronal Injury & Plasticity Program, University of Colorado School of Medicine, Aurora, CO, USA.,Department of Anesthesiology, University of Colorado School of Medicine, Aurora, CO, USA
| | - Olivia P Patsos
- Neuronal Injury & Plasticity Program, University of Colorado School of Medicine, Aurora, CO, USA.,Department of Anesthesiology, University of Colorado School of Medicine, Aurora, CO, USA
| | - Kaori Shimizu
- Neuronal Injury & Plasticity Program, University of Colorado School of Medicine, Aurora, CO, USA.,Department of Anesthesiology, University of Colorado School of Medicine, Aurora, CO, USA
| | - Nicholas Chalmers
- Neuronal Injury & Plasticity Program, University of Colorado School of Medicine, Aurora, CO, USA.,Department of Anesthesiology, University of Colorado School of Medicine, Aurora, CO, USA
| | - Guiying Deng
- Neuronal Injury & Plasticity Program, University of Colorado School of Medicine, Aurora, CO, USA.,Department of Anesthesiology, University of Colorado School of Medicine, Aurora, CO, USA
| | - Erika Tiemeier
- Neuronal Injury & Plasticity Program, University of Colorado School of Medicine, Aurora, CO, USA.,Department of Anesthesiology, University of Colorado School of Medicine, Aurora, CO, USA
| | - Nidia Quillinan
- Neuronal Injury & Plasticity Program, University of Colorado School of Medicine, Aurora, CO, USA.,Department of Anesthesiology, University of Colorado School of Medicine, Aurora, CO, USA
| | - Paco S Herson
- Neuronal Injury & Plasticity Program, University of Colorado School of Medicine, Aurora, CO, USA. .,Department of Pharmacology, University of Colorado School of Medicine, Aurora, CO, USA. .,Department of Anesthesiology, University of Colorado School of Medicine, Aurora, CO, USA.
| |
Collapse
|
33
|
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: 37] [Impact Index Per Article: 6.2] [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
|
34
|
Abstract
The TRP ion channel TRPM2 has an essential function in cell survival and protects the viability of a number of cell types after oxidative stress. It is highly expressed in many cancers including breast, prostate, and pancreatic cancer, melanoma, leukemia, and neuroblastoma, suggesting it promotes cancer cell survival. TRPM2 is activated by production of ADP-ribose (ADPR) following oxidative stress, which binds to the C-terminus of TRPM2, resulting in channel opening. In a number of cancers including neuroblastoma, TRPM2 has been shown to preserve viability and mechanisms have been identified. Activation of TRPM2 results in expression of transcription factors and kinases important in cell proliferation and survival including HIF-1/2α, CREB, nuclear factor (erythroid-derived 2)-related factor-2 (Nrf2), and Pyk2, and Src phosphorylation. Together, HIF-1/2α and CREB regulate expression of genes encoding proteins with roles in mitochondrial function including members of the electron transport complex involved in ATP production. These contribute to lower mitochondrial ROS production while expression of antioxidants regulated by HIF-1/2α, FOXO3a, CREB, and Nrf2 is maintained. CREB is also important in control of expression of key proteins involved in autophagy. When TRPM2-mediated calcium influx is inhibited, mitochondria are dysfunctional, cellular bioenergetics are reduced, production of ROS is increased, and autophagy and DNA repair are impaired, decreasing tumor growth and increasing chemotherapy sensitivity. Inhibition of TRPM2 expression or function results in decreased tumor proliferation and/or viability in many malignancies including breast, gastric, pancreatic, prostate, head and neck cancers, melanoma, neuroblastoma, and T-cell and acute myelogenous leukemia. However, in a small number of malignancies, activation of TRPM2 rather than inhibition has been reported to reduce tumor cell survival. Here, TRPM2-mediated Ca2+ signaling and mechanisms of regulation of cancer cell growth and survival are reviewed and controversies discussed. Evidence suggests that targeting TRPM2 may be a novel therapeutic approach in many cancers.
Collapse
Affiliation(s)
- Barbara A Miller
- Departments of Pediatrics, and Biochemistry and Molecular Biology, The Pennsylvania State University College of Medicine, P.O. Box 850, Hershey, PA 17033, USA.
| |
Collapse
|
35
|
Park HJ, Shin KC, Yoou SK, Kang M, Kim JG, Sung DJ, Yu W, Lee Y, Kim SH, Bae YM, Park SW. Hydrogen peroxide constricts rat arteries by activating Na +-permeable and Ca 2+-permeable cation channels. Free Radic Res 2018; 53:94-103. [PMID: 30526150 DOI: 10.1080/10715762.2018.1556394] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/27/2022]
Abstract
Oxidative stress is associated with many cardiovascular diseases, such as hypertension and arteriosclerosis. Oxidative stress reportedly activates the L-type voltage-gated calcium channel (VDCCL) and elevates [Ca2+]i in many cells. However, how oxidative stress activates VDCCL under clinical setting and the consequence for arteries are unclear. Here, we examined the hypothesis that hydrogen peroxide (H2O2) regulates membrane potential (Em) by altering Na+ influx through cation channels, which consequently activates VDCCL to induce vasoconstriction in rat mesenteric arteries. To measure the tone of the endothelium-denuded arteries, a conventional isometric organ chamber was used. Membrane currents and Em were recorded by the patch-clamp technique. [Ca2+]i and [Na+]i were measured with microfluorometry using Fura2-AM and SBFI-AM, respectively. We found that H2O2 (10 and 100 µM) increased arterial contraction, and nifedipine blocked the effects of H2O2 on isometric contraction. H2O2 increased [Ca2+]i as well as [Na+]i, and depolarised Em. Gd3+ (1 µM) blocked all these H2O2-induced effects including Em depolarisation and increases in [Ca2+]i and [Na+]i. Although both nifedipine (30 nM) and low Na+ bath solution completely prevented the H2O2-induced increase in [Na+], they only partly inhibited the H2O2-induced effects on [Ca2+]i and Em. Taken together, the results suggested that H2O2 constricts rat arteries by causing Em depolarisation and VDCCL activation through activating Gd3+-and nifedipine-sensitive, Na+-permeable channels as well as Gd3+-sensitive Ca2+-permeable cation channels. We suggest that unidentified Na+-permeable cation channels as well as Ca2+-permeable cation channels may function as important mediators for oxidative stress-induced vascular dysfunction.
Collapse
Affiliation(s)
- Hyun Ji Park
- a Department of Physiology, KU Open Innovation Center , Research Institute of Medical Science, Konkuk University School of Medicine , Chungju , Republic of Korea
| | - Kyung Chul Shin
- a Department of Physiology, KU Open Innovation Center , Research Institute of Medical Science, Konkuk University School of Medicine , Chungju , Republic of Korea
| | - Soon-Kyu Yoou
- b Department of Emergency Medical Services , Eulji University , Seongnam , Republic of Korea
| | - Myeongsin Kang
- b Department of Emergency Medical Services , Eulji University , Seongnam , Republic of Korea
| | - Jae Gon Kim
- a Department of Physiology, KU Open Innovation Center , Research Institute of Medical Science, Konkuk University School of Medicine , Chungju , Republic of Korea
| | - Dong Jun Sung
- c Division of Sport and Health Science, College of Biomedical and Health Science , Konkuk University , Chungju , Republic of Korea
| | - Wonjong Yu
- d Department of Physical Therapy , Eulji University , Eulji , Republic of Korea
| | - Youngjin Lee
- e Department of Radiological Science , Gachon University , Yeonsu-gu , Republic of Korea
| | - Sung Hea Kim
- f Department of Cardiology , Konkuk University School of Medicine , Seoul , Republic of Korea
| | - Young Min Bae
- a Department of Physiology, KU Open Innovation Center , Research Institute of Medical Science, Konkuk University School of Medicine , Chungju , Republic of Korea
| | - Sang Woong Park
- b Department of Emergency Medical Services , Eulji University , Seongnam , Republic of Korea
| |
Collapse
|
36
|
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
|
37
|
Startek JB, Voets T, Talavera K. To flourish or perish: evolutionary TRiPs into the sensory biology of plant-herbivore interactions. Pflugers Arch 2018; 471:213-236. [PMID: 30229297 DOI: 10.1007/s00424-018-2205-1] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2018] [Revised: 08/31/2018] [Accepted: 09/06/2018] [Indexed: 12/18/2022]
Abstract
The interactions between plants and their herbivores are highly complex systems generating on one side an extraordinary diversity of plant protection mechanisms and on the other side sophisticated consumer feeding strategies. Herbivores have evolved complex, integrative sensory systems that allow them to distinguish between food sources having mere bad flavors from the actually toxic ones. These systems are based on the senses of taste, olfaction and somatosensation in the oral and nasal cavities, and on post-ingestive chemosensory mechanisms. The potential ability of plant defensive chemical traits to induce tissue damage in foragers is mainly encoded in the latter through chemesthetic sensations such as burning, pain, itch, irritation, tingling, and numbness, all of which induce innate aversive behavioral responses. Here, we discuss the involvement of transient receptor potential (TRP) channels in the chemosensory mechanisms that are at the core of complex and fascinating plant-herbivore ecological networks. We review how "sensory" TRPs are activated by a myriad of plant-derived compounds, leading to cation influx, membrane depolarization, and excitation of sensory nerve fibers of the oronasal cavities in mammals and bitter-sensing cells in insects. We also illustrate how TRP channel expression patterns and functionalities vary between species, leading to intriguing evolutionary adaptations to the specific habitats and life cycles of individual organisms.
Collapse
Affiliation(s)
- Justyna B Startek
- Laboratory of Ion Channel Research, Department of Cellular and Molecular Medicine, KU Leuven, Herestraat 49, Campus Gasthuisberg O&N1 bus 802, 3000, Leuven, Belgium. .,VIB Center for Brain & Disease Research, Leuven, Belgium.
| | - Thomas Voets
- Laboratory of Ion Channel Research, Department of Cellular and Molecular Medicine, KU Leuven, Herestraat 49, Campus Gasthuisberg O&N1 bus 802, 3000, Leuven, Belgium.,VIB Center for Brain & Disease Research, Leuven, Belgium
| | - Karel Talavera
- Laboratory of Ion Channel Research, Department of Cellular and Molecular Medicine, KU Leuven, Herestraat 49, Campus Gasthuisberg O&N1 bus 802, 3000, Leuven, Belgium.,VIB Center for Brain & Disease Research, Leuven, Belgium
| |
Collapse
|
38
|
Hirschler-Laszkiewicz I, Chen SJ, Bao L, Wang J, Zhang XQ, Shanmughapriya S, Keefer K, Madesh M, Cheung JY, Miller BA. The human ion channel TRPM2 modulates neuroblastoma cell survival and mitochondrial function through Pyk2, CREB, and MCU activation. Am J Physiol Cell Physiol 2018; 315:C571-C586. [PMID: 30020827 PMCID: PMC6230687 DOI: 10.1152/ajpcell.00098.2018] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
Transient receptor potential melastatin channel subfamily member 2 (TRPM2) has an essential function in cell survival and is highly expressed in many cancers. Inhibition of TRPM2 in neuroblastoma by depletion with CRISPR technology or expression of dominant negative TRPM2-S has been shown to significantly reduce cell viability. Here, the role of proline-rich tyrosine kinase 2 (Pyk2) in TRPM2 modulation of neuroblastoma viability was explored. In TRPM2-depleted cells, phosphorylation and expression of Pyk2 and cAMP-responsive element-binding protein (CREB), a downstream target, were significantly reduced after application of the chemotherapeutic agent doxorubicin. Overexpression of wild-type Pyk2 rescued cell viability. Reduction of Pyk2 expression with shRNA decreased cell viability and CREB phosphorylation and expression, demonstrating Pyk2 modulates CREB activation. TRPM2 depletion impaired phosphorylation of Src, an activator of Pyk2, and this may be a mechanism to reduce Pyk2 phosphorylation. TRPM2 inhibition was previously demonstrated to decrease mitochondrial function. Here, CREB, Pyk2, and phosphorylated Src were reduced in mitochondria of TRPM2-depleted cells, consistent with their role in modulating expression and activation of mitochondrial proteins. Phosphorylated Src and phosphorylated and total CREB were reduced in TRPM2-depleted nuclei. Expression and function of mitochondrial calcium uniporter (MCU), a target of phosphorylated Pyk2 and CREB, were significantly reduced. Wild-type TRPM2 but not Ca2+-impermeable mutant E960D reconstituted phosphorylation and expression of Pyk2 and CREB in TRPM2-depleted cells exposed to doxorubicin. Results demonstrate that TRPM2 expression protects the viability of neuroblastoma through Src, Pyk2, CREB, and MCU activation, which play key roles in maintaining mitochondrial function and cellular bioenergetics.
Collapse
Affiliation(s)
| | - Shu-Jen Chen
- Department of Pediatrics, The Pennsylvania State University College of Medicine , Hershey, Pennsylvania
| | - Lei Bao
- Department of Pediatrics, The Pennsylvania State University College of Medicine , Hershey, Pennsylvania
| | - JuFang Wang
- The Center of Translational Medicine, Lewis Katz School of Medicine of Temple University , Philadelphia, Pennsylvania
| | - Xue-Qian Zhang
- The Center of Translational Medicine, Lewis Katz School of Medicine of Temple University , Philadelphia, Pennsylvania
| | - Santhanam Shanmughapriya
- The Center of Translational Medicine, Lewis Katz School of Medicine of Temple University , Philadelphia, Pennsylvania.,Department of Biochemistry, Lewis Katz School of Medicine of Temple University , Philadelphia, Pennsylvania
| | - Kerry Keefer
- Department of Pediatrics, The Pennsylvania State University College of Medicine , Hershey, Pennsylvania
| | - Muniswamy Madesh
- The Center of Translational Medicine, Lewis Katz School of Medicine of Temple University , Philadelphia, Pennsylvania.,Department of Biochemistry, Lewis Katz School of Medicine of Temple University , Philadelphia, Pennsylvania
| | - Joseph Y Cheung
- The Center of Translational Medicine, Lewis Katz School of Medicine of Temple University , Philadelphia, Pennsylvania.,Department of Medicine, Lewis Katz School of Medicine of Temple University , Philadelphia, Pennsylvania
| | - Barbara A Miller
- Department of Pediatrics, The Pennsylvania State University College of Medicine , Hershey, Pennsylvania.,Department of Biochemistry and Molecular Biology, The Pennsylvania State University College of Medicine , Hershey, Pennsylvania
| |
Collapse
|
39
|
Li X, Jiang LH. Multiple molecular mechanisms form a positive feedback loop driving amyloid β42 peptide-induced neurotoxicity via activation of the TRPM2 channel in hippocampal neurons. Cell Death Dis 2018; 9:195. [PMID: 29416015 PMCID: PMC5833848 DOI: 10.1038/s41419-018-0270-1] [Citation(s) in RCA: 45] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2017] [Revised: 12/01/2017] [Accepted: 12/27/2017] [Indexed: 11/10/2022]
Abstract
Emerging evidence supports an important role for the ROS-sensitive TRPM2 channel in mediating age-related cognitive impairment in Alzheimer’s disease (AD), particularly neurotoxicity resulting from generation of excessive neurotoxic Aβ peptides. Here we examined the elusive mechanisms by which Aβ42 activates the TRPM2 channel to induce neurotoxicity in mouse hippocampal neurons. Aβ42-induced neurotoxicity was ablated by genetic knockout (TRPM2-KO) and attenuated by inhibition of the TRPM2 channel activity or activation through PARP-1. Aβ42-induced neurotoxicity was also inhibited by treatment with TPEN used as a Zn2+-specific chelator. Cell imaging revealed that Aβ42-induced lysosomal dysfunction, cytosolic Zn2+ increase, mitochondrial Zn2+ accumulation, loss of mitochondrial function, and mitochondrial generation of ROS. These effects were suppressed by TRPM2-KO, inhibition of TRPM2 or PARP-1, or treatment with TPEN. Bafilomycin-induced lysosomal dysfunction also resulted in TRPM2-dependent cytosolic Zn2+ increase, mitochondrial Zn2+ accumulation, and mitochondrial generation of ROS, supporting that lysosomal dysfunction and accompanying Zn2+ release trigger mitochondrial Zn2+ accumulation and generation of ROS. Aβ42-induced effects on lysosomal and mitochondrial functions besides neurotoxicity were also suppressed by inhibition of PKC and NOX. Furthermore, Aβ42-induced neurotoxicity was prevented by inhibition of MEK/ERK. Therefore, our study reveals multiple molecular mechanisms, including PKC/NOX-mediated generation of ROS, activation of MEK/ERK and PARP-1, lysosomal dysfunction and Zn2+ release, mitochondrial Zn2+ accumulation, loss of mitochondrial function, and mitochondrial generation of ROS, are critically engaged in forming a positive feedback loop that drives Aβ42-induced activation of the TRPM2 channel and neurotoxicity in hippocampal neurons. These findings shed novel and mechanistic insights into AD pathogenesis.
Collapse
Affiliation(s)
- Xin Li
- School of Biomedical Sciences, Faculty of Biological Sciences, University of Leeds, Leeds, UK
| | - Lin-Hua Jiang
- School of Biomedical Sciences, Faculty of Biological Sciences, University of Leeds, Leeds, UK. .,Sino-UK Joint Laboratory of Brain Function and Injury of Henan Province and Department of Physiology and Neurobiology, Xinxiang Medical University, Xinxiang, China.
| |
Collapse
|
40
|
Sachdeva R, Schlotterer A, Schumacher D, Matka C, Mathar I, Dietrich N, Medert R, Kriebs U, Lin J, Nawroth P, Birnbaumer L, Fleming T, Hammes HP, Freichel M. TRPC proteins contribute to development of diabetic retinopathy and regulate glyoxalase 1 activity and methylglyoxal accumulation. Mol Metab 2018; 9:156-167. [PMID: 29373286 PMCID: PMC5870093 DOI: 10.1016/j.molmet.2018.01.003] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/01/2017] [Revised: 12/18/2017] [Accepted: 01/02/2018] [Indexed: 12/19/2022] Open
Abstract
OBJECTIVE Diabetic retinopathy (DR) is induced by an accumulation of reactive metabolites such as ROS, RNS, and RCS species, which were reported to modulate the activity of cation channels of the TRPC family. In this study, we use Trpc1/4/5/6-/- compound knockout mice to analyze the contribution of these TRPC proteins to diabetic retinopathy. METHODS We used Nanostring- and qPCR-based analysis to determine mRNA levels of TRPC channels in control and diabetic retinae and retinal cell types. Chronic hyperglycemia was induced by Streptozotocin (STZ) treatment. To assess the development of diabetic retinopathy, vasoregression, pericyte loss, and thickness of individual retinal layers were analyzed. Plasma and cellular methylglyoxal (MG) levels, as well as Glyoxalase 1 (GLO1) enzyme activity and protein expression, were measured in WT and Trpc1/4/5/6-/- cells or tissues. MG-evoked toxicity in cells of both genotypes was compared by MTT assay. RESULTS We find that Trpc1/4/5/6-/- mice are protected from hyperglycemia-evoked vasoregression determined by the formation of acellular capillaries and pericyte drop-out. In addition, Trpc1/4/5/6-/- mice are resistant to the STZ-induced reduction in retinal layer thickness. The RCS metabolite methylglyoxal, which represents a key mediator for the development of diabetic retinopathy, was significantly reduced in plasma and red blood cells (RBCs) of STZ-treated Trpc1/4/5/6-/- mice compared to controls. GLO1 is the major MG detoxifying enzyme, and its activity and protein expression were significantly elevated in Trpc1/4/5/6-deficient cells, which led to significantly increased resistance to MG toxicity. GLO1 activity was also increased in retinal extracts from Trpc1/4/5/6-/- mice. The TRPCs investigated here are expressed at different levels in endothelial and glial cells of the retina. CONCLUSION The protective phenotype in diabetic retinopathy observed in Trpc1/4/5/6-/- mice is suggestive of a predominant action of TRPCs in Müller cells and microglia because of their central position in the retention of a proper homoeostasis of the neurovascular unit.
Collapse
Affiliation(s)
- Robin Sachdeva
- Institute of Pharmacology, Heidelberg University, Im Neuenheimer Feld 366, 69120 Heidelberg, Germany
| | - Andrea Schlotterer
- Vth Department of Medicine, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
| | - Dagmar Schumacher
- Institute of Pharmacology, Heidelberg University, Im Neuenheimer Feld 366, 69120 Heidelberg, Germany
| | - Christin Matka
- Institute of Pharmacology, Heidelberg University, Im Neuenheimer Feld 366, 69120 Heidelberg, Germany
| | - Ilka Mathar
- Institute of Pharmacology, Heidelberg University, Im Neuenheimer Feld 366, 69120 Heidelberg, Germany
| | - Nadine Dietrich
- Vth Department of Medicine, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
| | - Rebekka Medert
- Institute of Pharmacology, Heidelberg University, Im Neuenheimer Feld 366, 69120 Heidelberg, Germany
| | - Ulrich Kriebs
- Institute of Pharmacology, Heidelberg University, Im Neuenheimer Feld 366, 69120 Heidelberg, Germany
| | - Jihong Lin
- Vth Department of Medicine, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
| | - Peter Nawroth
- Department of Medicine I and Clinical Chemistry, University Hospital Heidelberg, Germany; German Center for Diabetes Research (DZD), Germany; Institute for Diabetes and Cancer IDC Helmholtz Center Munich, Neuherberg, Germany; Joint Heidelberg-IDC Translational Diabetes Program, Dept. of Medicine I, Heidelberg University Hospital, Heidelberg, Germany
| | - Lutz Birnbaumer
- Neurobiology Laboratory, National Institute of Environmental Health Sciences, North Carolina, USA; Institute for Biomedical Research (BIOMED), School of Medical sciences, Catholic University of Argentina, Buenos Aires, Argentina
| | - Thomas Fleming
- Department of Medicine I and Clinical Chemistry, University Hospital Heidelberg, Germany; German Center for Diabetes Research (DZD), Germany
| | - Hans-Peter Hammes
- Vth Department of Medicine, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
| | - Marc Freichel
- Institute of Pharmacology, Heidelberg University, Im Neuenheimer Feld 366, 69120 Heidelberg, Germany.
| |
Collapse
|
41
|
Kühn F, Kühn C, Lückhoff A. Different Principles of ADP-Ribose-Mediated Activation and Opposite Roles of the NUDT9 Homology Domain in the TRPM2 Orthologs of Man and Sea Anemone. Front Physiol 2017; 8:879. [PMID: 29163217 PMCID: PMC5671594 DOI: 10.3389/fphys.2017.00879] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2017] [Accepted: 10/18/2017] [Indexed: 01/31/2023] Open
Abstract
A decisive element in the human cation channel TRPM2 is a region in its cytosolic C-terminus named NUDT9H because of its homology to the NUDT9 enzyme, a pyrophosphatase degrading ADP-ribose (ADPR). In hTRPM2, however, the NUDT9H domain has lost its enzymatic activity but serves as a binding domain for ADPR. As consequence of binding, gating of the channel is initiated. Since ADPR is produced after oxidative DNA damage, hTRPM2 mediates Ca2+ influx in response to oxidative stress which may lead to cell death. In the genome of the sea anemone Nematostella vectensis (nv), a preferred model organism for the evolution of key bilaterian features, a TRPM2 ortholog has been identified that contains a NUDT9H domain as well. Heterologous expression of nvTRPM2 in HEK-293 cells reveals a cation channel with many close similarities to the human counterpart. Most notably, nvTRPM2 is activated by ADPR, and Ca2+ is a co-agonist. However, the intramolecular mechanisms of ADPR gating as well as the role of NUDT9H are strikingly different in the two species. Whereas already subtle changes of NUDT9H abolish ADPR gating in hTRPM2, the region can be completely removed from nvTRPM2 without loss of responses to ADPR. An alternative ADPR binding site seems to be present but has not yet been characterized. The ADP-ribose pyrophosphatase (ADPRase) function of nvNUDT9H has been preserved but can be abolished by numerous genetic manipulations. All these manipulations create channels that are sensitive to hydrogen peroxide which fails to induce channel activity in wild-type nvTRPM2. Therefore, the function of NUDT9H in nvTRPM2 is the degradation of ADPR, thereby reducing agonist concentration in the presence of oxidative stress. Thus, the two TRPM2 orthologs have evolved divergently but nevertheless gained analogous functional properties, i.e., gating by ADPR with Ca2+ as co-factor. Opposite roles are played by the respective NUDT9H domains, either binding of ADPR and mediating channel activity, or controlling the availability of ADPR at the binding site located in a different domain.
Collapse
Affiliation(s)
- Frank Kühn
- Medical Faculty, Institute of Physiology, RWTH Aachen University, Aachen, Germany
| | - Cornelia Kühn
- Medical Faculty, Institute of Physiology, RWTH Aachen University, Aachen, Germany
| | - Andreas Lückhoff
- Medical Faculty, Institute of Physiology, RWTH Aachen University, Aachen, Germany
| |
Collapse
|
42
|
Carvacrol promotes neuroprotection in the mouse hemiparkinsonian model. Neuroscience 2017; 356:176-181. [DOI: 10.1016/j.neuroscience.2017.05.013] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2017] [Revised: 05/04/2017] [Accepted: 05/08/2017] [Indexed: 12/12/2022]
|
43
|
Blatter LA. Tissue Specificity: SOCE: Implications for Ca 2+ Handling in Endothelial Cells. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2017; 993:343-361. [PMID: 28900923 DOI: 10.1007/978-3-319-57732-6_18] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Many cellular functions of the vascular endothelium are regulated by fine-tuned global and local, microdomain-confined changes of cytosolic free Ca2+ ([Ca2+]i). Vasoactive agonist-induced stimulation of vascular endothelial cells (VECs) typically induces Ca2+ release through IP3 receptor Ca2+ release channels embedded in the membrane of the endoplasmic reticulum (ER) Ca2+ store, followed by Ca2+ entry from the extracellular space elicited by Ca2+ store depletion and referred to as capacitative or store-operated Ca2+ entry (SOCE). In vascular endothelial cells, SOCE is graded with the degree of store depletion and controlled locally in the subcellular microdomain where depletion occurs. SOCE provides distinct Ca2+ signals that selectively control specific endothelial functions: in calf pulmonary artery endothelial cells, the SOCE Ca2+ signal drives nitric oxide (an endothelium-derived relaxing factor of the vascular smooth muscle) production and controls activation and nuclear translocation of the transcription factor NFAT. Both cellular events are not affected by Ca2+ signals of comparable magnitude arising directly from Ca2+ release from intracellular stores, clearly indicating that SOCE regulates specific Ca2+-dependent cellular tasks by a unique and exclusive mechanism. This review discusses the mechanisms of intracellular Ca2+ regulation in vascular endothelial cells and the role of store-operated Ca2+ entry for endothelium-dependent smooth muscle relaxation and nitric oxide signaling, endothelial oxidative stress response, and excitation-transcription coupling in the vascular endothelium.
Collapse
Affiliation(s)
- Lothar A Blatter
- Department of Physiology and Biophysics, Rush University Medical Center, 1750 W. Harrison St., Chicago, IL, 60612, USA.
| |
Collapse
|
44
|
Zhao LY, Xu WL, Xu ZQ, Qi C, Li Y, Cheng J, Liu LK, Wu YN, Gao J, Ye JH. The overexpressed functional transient receptor potential channel TRPM2 in oral squamous cell carcinoma. Sci Rep 2016; 6:38471. [PMID: 28008929 PMCID: PMC5180100 DOI: 10.1038/srep38471] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2016] [Accepted: 11/10/2016] [Indexed: 12/20/2022] Open
Abstract
TRPM2, one member of the transient receptor potential (TRP) protein super-family, is a Ca2+-permeable channel that is activated by oxidative stress and confers susceptibility to cell death. In the human tongue specimens of carcinoma and the tongue carcinoma SCC cell lines, we observed the enhanced expression of TRPM2. By means of the whole-cell electrophysiological recording, the ADPR-induced currents mediated by TRPM2 were recorded in cultured SCC9 cells. Moreover, after H2O2 treatment for 24 hours, the apoptotic number of SCC9 cells was significantly increased. However, the selectively knocked-down TRPM2 with the small interfering RNA technique inhibited the survival and migration of the SCC9 cancer cells, which was independent of the p53-p21 pathway, since the expression of p21 was enhanced after TRPM2 knockdown. Furthermore, the sub-cellular localization of TRPM2 was remarkably different between cancerous and non-cancerous cells. A significant amount of the TRPM2 proteins were located in the nuclei in cancer cells. All these data suggest that TRPM2 is essential for the survival and migration of SCC cancer cells and may be a potential target for the selective treatment of tongue cancer.
Collapse
Affiliation(s)
- Ling-Yan Zhao
- Jiangsu Key Laboratory of Oral Diseases and Department of Oral and maxillofacial surgery, Affiliated Hospital of Stomatology, Nanjing Medical University, 136 Hanzhong Road, Nanjing, 210029, China
| | - Wan-Lin Xu
- Jiangsu Key Laboratory of Oral Diseases and Department of Oral and maxillofacial surgery, Affiliated Hospital of Stomatology, Nanjing Medical University, 136 Hanzhong Road, Nanjing, 210029, China.,Shanghai Key Laboratory of Stomatology and Shanghai Research Institute of Stomatology, Department of Oral and Maxillofacial-Head and Neck Oncology, Ninth People's Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200011, China
| | - Zeng-Qi Xu
- Jiangsu Key Laboratory of Oral Diseases and Department of Oral and maxillofacial surgery, Affiliated Hospital of Stomatology, Nanjing Medical University, 136 Hanzhong Road, Nanjing, 210029, China
| | - Cui Qi
- Key Laboratory of Human Functional Genomics of Jiangsu, Department of Neurobiology, Nanjing Medical University, 101 Longmian Road, Nanjing, Jiangsu, 211166, China
| | - Yang Li
- Jiangsu Key Laboratory of Oral Diseases and Department of Oral and maxillofacial surgery, Affiliated Hospital of Stomatology, Nanjing Medical University, 136 Hanzhong Road, Nanjing, 210029, China
| | - Jie Cheng
- Jiangsu Key Laboratory of Oral Diseases and Department of Oral and maxillofacial surgery, Affiliated Hospital of Stomatology, Nanjing Medical University, 136 Hanzhong Road, Nanjing, 210029, China
| | - Lai-Kui Liu
- Jiangsu Key Laboratory of Oral Diseases and Department of Oral and maxillofacial surgery, Affiliated Hospital of Stomatology, Nanjing Medical University, 136 Hanzhong Road, Nanjing, 210029, China
| | - Yu-Nong Wu
- Jiangsu Key Laboratory of Oral Diseases and Department of Oral and maxillofacial surgery, Affiliated Hospital of Stomatology, Nanjing Medical University, 136 Hanzhong Road, Nanjing, 210029, China
| | - Jun Gao
- Key Laboratory of Human Functional Genomics of Jiangsu, Department of Neurobiology, Nanjing Medical University, 101 Longmian Road, Nanjing, Jiangsu, 211166, China
| | - Jin-Hai Ye
- Jiangsu Key Laboratory of Oral Diseases and Department of Oral and maxillofacial surgery, Affiliated Hospital of Stomatology, Nanjing Medical University, 136 Hanzhong Road, Nanjing, 210029, China
| |
Collapse
|
45
|
Nicholas S, Yuan SY, Brookes SJH, Spencer NJ, Zagorodnyuk VP. Hydrogen peroxide preferentially activates capsaicin-sensitive high threshold afferents via TRPA1 channels in the guinea pig bladder. Br J Pharmacol 2016; 174:126-138. [PMID: 27792844 DOI: 10.1111/bph.13661] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2016] [Revised: 09/22/2016] [Accepted: 10/19/2016] [Indexed: 11/28/2022] Open
Abstract
BACKGROUND AND PURPOSE There is increasing evidence suggesting that ROS play a major pathological role in bladder dysfunction induced by bladder inflammation and/or obstruction. The aim of this study was to determine the effect of H2 O2 on different types of bladder afferents and its mechanism of action on sensory neurons in the guinea pig bladder. EXPERIMENTAL APPROACH 'Close-to-target' single unit extracellular recordings were made from fine branches of pelvic nerves entering the guinea pig bladder, in flat sheet preparations, in vitro. KEY RESULTS H2 O2 (300-1000 μM) preferentially and potently activated capsaicin-sensitive high threshold afferents but not low threshold stretch-sensitive afferents, which were only activated by significantly higher concentrations of hydrogen peroxide. The TRPV1 channel agonist, capsaicin, excited 86% of high threshold afferents. The TRPA1 channel agonist, allyl isothiocyanate and the TRPM8 channel agonist, icilin activated 72% and 47% of capsaicin-sensitive high threshold afferents respectively. The TRPA1 channel antagonist, HC-030031, but not the TRPV1 channel antagonist, capsazepine or the TRPM8 channel antagonist, N-(2-aminoethyl)-N-[[3-methoxy-4-(phenylmethoxy)phenyl]methyl]thiophene-2-carboxamide, significantly inhibited the H2 O2 -induced activation of high threshold afferents. Dimethylthiourea and deferoxamine did not significantly change the effect of H2 O2 on high threshold afferents. CONCLUSIONS AND IMPLICATIONS The findings show that H2 O2 , in the concentration range detected in inflammation or reperfusion after ischaemia, evoked long-lasting activation of the majority of capsaicin-sensitive high threshold afferents, but not low threshold stretch-sensitive afferents. The data suggest that the TRPA1 channels located on these capsaicin-sensitive afferent fibres are probable targets of ROS released during oxidative stress.
Collapse
Affiliation(s)
- S Nicholas
- Discipline of Human Physiology & Centre for Neuroscience, Flinders University of South Australia, Adelaide, SA, Australia
| | - S Y Yuan
- Discipline of Anatomy and Histology & Centre for Neuroscience, Flinders University of South Australia, Adelaide, SA, Australia
| | - S J H Brookes
- Discipline of Human Physiology & Centre for Neuroscience, Flinders University of South Australia, Adelaide, SA, Australia
| | - N J Spencer
- Discipline of Human Physiology & Centre for Neuroscience, Flinders University of South Australia, Adelaide, SA, Australia
| | - V P Zagorodnyuk
- Discipline of Human Physiology & Centre for Neuroscience, Flinders University of South Australia, Adelaide, SA, Australia
| |
Collapse
|
46
|
Bao L, Chen SJ, Conrad K, Keefer K, Abraham T, Lee JP, Wang J, Zhang XQ, Hirschler-Laszkiewicz I, Wang HG, Dovat S, Gans B, Madesh M, Cheung JY, Miller BA. Depletion of the Human Ion Channel TRPM2 in Neuroblastoma Demonstrates Its Key Role in Cell Survival through Modulation of Mitochondrial Reactive Oxygen Species and Bioenergetics. J Biol Chem 2016; 291:24449-24464. [PMID: 27694440 DOI: 10.1074/jbc.m116.747147] [Citation(s) in RCA: 52] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2016] [Revised: 09/29/2016] [Indexed: 12/13/2022] Open
Abstract
Transient receptor potential melastatin 2 (TRPM2) ion channel has an essential function in modulating cell survival following oxidant injury and is highly expressed in many cancers including neuroblastoma. Here, in xenografts generated from neuroblastoma cells in which TRPM2 was depleted with CRISPR/Cas9 technology and in in vitro experiments, tumor growth was significantly inhibited and doxorubicin sensitivity increased. The hypoxia-inducible transcription factor 1/2α (HIF-1/2α) signaling cascade including proteins involved in oxidant stress, glycolysis, and mitochondrial function was suppressed by TRPM2 depletion. TRPM2-depleted SH-SY5Y neuroblastoma cells demonstrated reduced oxygen consumption and ATP production after doxorubicin, confirming impaired cellular bioenergetics. In cells in which TRPM2 was depleted, mitochondrial superoxide production was significantly increased, particularly following doxorubicin. Ectopic expression of superoxide dismutase 2 (SOD2) reduced ROS and preserved viability of TRPM2-depleted cells, however, failed to restore ATP levels. Mitochondrial reactive oxygen species (ROS) were also significantly increased in cells in which TRPM2 function was inhibited by TRPM2-S, and pretreatment of these cells with the antioxidant MitoTEMPO significantly reduced ROS levels in response to doxorubicin and protected cell viability. Expression of the TRPM2 pore mutant E960D, in which calcium entry through TRPM2 is abolished, also resulted in significantly increased mitochondrial ROS following doxorubicin treatment, showing the critical role of TRPM2-mediated calcium entry. These findings demonstrate the important function of TRPM2 in modulation of cell survival through mitochondrial ROS, and the potential of targeted inhibition of TRPM2 as a therapeutic approach to reduce cellular bioenergetics, tumor growth, and enhance susceptibility to chemotherapeutic agents.
Collapse
Affiliation(s)
- Lei Bao
- Departments of Pediatrics, The Pennsylvania State University College of Medicine, Hershey, Pennsylvania 17033
| | - Shu-Jen Chen
- Departments of Pediatrics, The Pennsylvania State University College of Medicine, Hershey, Pennsylvania 17033
| | - Kathleen Conrad
- Departments of Pediatrics, The Pennsylvania State University College of Medicine, Hershey, Pennsylvania 17033
| | - Kerry Keefer
- Departments of Pediatrics, The Pennsylvania State University College of Medicine, Hershey, Pennsylvania 17033
| | - Thomas Abraham
- Neural and Behavioral Sciences and Microscopy Imaging Facility
| | - John P Lee
- Departments of Pediatrics, The Pennsylvania State University College of Medicine, Hershey, Pennsylvania 17033
| | - JuFang Wang
- the Departments of Medicine, Temple University School of Medicine, Philadelphia, Pennsylvania 19140; The Center of Translational Medicine, Temple University School of Medicine, Philadelphia, Pennsylvania 19140
| | - Xue-Qian Zhang
- the Departments of Medicine, Temple University School of Medicine, Philadelphia, Pennsylvania 19140; The Center of Translational Medicine, Temple University School of Medicine, Philadelphia, Pennsylvania 19140
| | - Iwona Hirschler-Laszkiewicz
- Departments of Pediatrics, The Pennsylvania State University College of Medicine, Hershey, Pennsylvania 17033
| | - Hong-Gang Wang
- Departments of Pediatrics, The Pennsylvania State University College of Medicine, Hershey, Pennsylvania 17033; Departments of Pharmacology, The Pennsylvania State University College of Medicine, Hershey, Pennsylvania 17033
| | - Sinisa Dovat
- Departments of Pediatrics, The Pennsylvania State University College of Medicine, Hershey, Pennsylvania 17033; Biochemistry and Molecular Biology, The Pennsylvania State University College of Medicine, Hershey, Pennsylvania 17033 and
| | - Brian Gans
- Departments of Pediatrics, The Pennsylvania State University College of Medicine, Hershey, Pennsylvania 17033
| | - Muniswamy Madesh
- The Center of Translational Medicine, Temple University School of Medicine, Philadelphia, Pennsylvania 19140; Molecular Genetics and Medical Biochemistry, Temple University School of Medicine, Philadelphia, Pennsylvania 19140
| | - Joseph Y Cheung
- the Departments of Medicine, Temple University School of Medicine, Philadelphia, Pennsylvania 19140; The Center of Translational Medicine, Temple University School of Medicine, Philadelphia, Pennsylvania 19140
| | - Barbara A Miller
- Departments of Pediatrics, The Pennsylvania State University College of Medicine, Hershey, Pennsylvania 17033; Biochemistry and Molecular Biology, The Pennsylvania State University College of Medicine, Hershey, Pennsylvania 17033 and.
| |
Collapse
|
47
|
Kozai D, Sakaguchi R, Ohwada T, Mori Y. Deciphering Subtype-Selective Modulations in TRPA1 Biosensor Channels. Curr Neuropharmacol 2016; 13:266-78. [PMID: 26411770 PMCID: PMC4598439 DOI: 10.2174/1570159x1302150525122020] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
Abstract
The transient receptor potential (TRP) proteins are a family of ion channels that act as
cellular sensors. Several members of the TRP family are sensitive to oxidative stress mediators.
Among them, TRPA1 is remarkably susceptible to various oxidants, and is known to mediate
neuropathic pain and respiratory, vascular and gastrointestinal functions, making TRPA1 an
attractive therapeutic target. Recent studies have revealed a number of modulators (both activators and inhibitors) that act
on TRPA1. Endogenous mediators of oxidative stress and exogenous electrophiles activate TRPA1 through oxidative
modification of cysteine residues. Non-electrophilic compounds also activate TRPA1. Certain non-electrophilic
modulators may act on critical non-cysteine sites in TRPA1. However, a method to achieve selective modulation of
TRPA1 by small molecules has not yet been established. More recently, we found that a novel N-nitrosamine compound
activates TRPA1 by S-nitrosylation (the addition of a nitric oxide (NO) group to cysteine thiol), and does so with
significant selectivity over other NO-sensitive TRP channels. It is proposed that this subtype selectivity is conferred
through synergistic effects of electrophilic cysteine transnitrosylation and molecular recognition of the non-electrophilic
moiety on the N-nitrosamine. In this review, we describe the molecular pharmacology of these TRPA1 modulators and
discuss their modulatory mechanisms.
Collapse
Affiliation(s)
| | | | | | - Yasuo Mori
- Laboratory of Molecular Biology, Department of Synthetic Chemistry and Biological Chemistry, Graduate School of Engineering, Kyoto University, Katsura Campus, Nishikyoku, Kyoto 615-8510, Japan.
| |
Collapse
|
48
|
Saul S, Gibhardt CS, Schmidt B, Lis A, Pasieka B, Conrad D, Jung P, Gaupp R, Wonnenberg B, Diler E, Stanisz H, Vogt T, Schwarz EC, Bischoff M, Herrmann M, Tschernig T, Kappl R, Rieger H, Niemeyer BA, Bogeski I. A calcium-redox feedback loop controls human monocyte immune responses: The role of ORAI Ca2+ channels. Sci Signal 2016; 9:ra26. [PMID: 26956485 DOI: 10.1126/scisignal.aaf1639] [Citation(s) in RCA: 50] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
In phagocytes, pathogen recognition is followed by Ca(2+) mobilization and NADPH oxidase 2 (NOX2)-mediated "oxidative burst," which involves the rapid production of large amounts of reactive oxygen species (ROS). We showed that ORAI Ca(2+) channels control store-operated Ca(2+) entry, ROS production, and bacterial killing in primary human monocytes. ROS inactivate ORAI channels that lack an ORAI3 subunit. Staphylococcal infection of mice reduced the expression of the gene encoding the redox-sensitive Orai1 and increased the expression of the gene encoding the redox-insensitive Orai3 in the lungs or in bronchoalveolar lavages. A similar switch from ORAI1 to ORAI3 occurred in primary human monocytes exposed to bacterial peptides in culture. These alterations in ORAI1 and ORAI3 abundance shifted the channel assembly toward a more redox-insensitive configuration. Accordingly, silencing ORAI3 increased the redox sensitivity of the channel and enhanced oxidation-induced inhibition of NOX2. We generated a mathematical model that predicted additional features of the Ca(2+)-redox interplay. Our results identified the ORAI-NOX2 feedback loop as a determinant of monocyte immune responses.
Collapse
Affiliation(s)
- Stephanie Saul
- Department of Biophysics, Center for Integrative Physiology and Molecular Medicine (CIPPM), School of Medicine, Saarland University, Homburg 66421, Germany
| | - Christine S Gibhardt
- Department of Biophysics, Center for Integrative Physiology and Molecular Medicine (CIPPM), School of Medicine, Saarland University, Homburg 66421, Germany
| | - Barbara Schmidt
- Department of Biophysics, Center for Integrative Physiology and Molecular Medicine (CIPPM), School of Medicine, Saarland University, Homburg 66421, Germany. Department of Theoretical Physics, Saarland University, Saarbrücken 66123, Germany. Molecular Biophysics, CIPMM, School of Medicine, Saarland University, Homburg 66421, Germany
| | - Annette Lis
- Department of Biophysics, Center for Integrative Physiology and Molecular Medicine (CIPPM), School of Medicine, Saarland University, Homburg 66421, Germany
| | - Bastian Pasieka
- Department of Biophysics, Center for Integrative Physiology and Molecular Medicine (CIPPM), School of Medicine, Saarland University, Homburg 66421, Germany
| | - David Conrad
- Department of Biophysics, Center for Integrative Physiology and Molecular Medicine (CIPPM), School of Medicine, Saarland University, Homburg 66421, Germany
| | - Philipp Jung
- Institute of Medical Microbiology and Hygiene, Saarland University, Homburg 66421, Germany
| | - Rosmarie Gaupp
- Institute of Medical Microbiology and Hygiene, Saarland University, Homburg 66421, Germany
| | - Bodo Wonnenberg
- Department of Anatomy, School of Medicine, Saarland University, Homburg 66421, Germany
| | - Ebru Diler
- Department of Anatomy, School of Medicine, Saarland University, Homburg 66421, Germany
| | - Hedwig Stanisz
- Department of Dermatology, Venereology and Allergology, University Hospital of Saarland, Homburg 66421, Germany
| | - Thomas Vogt
- Department of Dermatology, Venereology and Allergology, University Hospital of Saarland, Homburg 66421, Germany
| | - Eva C Schwarz
- Department of Biophysics, Center for Integrative Physiology and Molecular Medicine (CIPPM), School of Medicine, Saarland University, Homburg 66421, Germany
| | - Markus Bischoff
- Institute of Medical Microbiology and Hygiene, Saarland University, Homburg 66421, Germany
| | - Mathias Herrmann
- Institute of Medical Microbiology and Hygiene, Saarland University, Homburg 66421, Germany
| | - Thomas Tschernig
- Department of Anatomy, School of Medicine, Saarland University, Homburg 66421, Germany
| | - Reinhard Kappl
- Department of Biophysics, Center for Integrative Physiology and Molecular Medicine (CIPPM), School of Medicine, Saarland University, Homburg 66421, Germany
| | - Heiko Rieger
- Department of Theoretical Physics, Saarland University, Saarbrücken 66123, Germany
| | - Barbara A Niemeyer
- Molecular Biophysics, CIPMM, School of Medicine, Saarland University, Homburg 66421, Germany
| | - Ivan Bogeski
- Department of Biophysics, Center for Integrative Physiology and Molecular Medicine (CIPPM), School of Medicine, Saarland University, Homburg 66421, Germany.
| |
Collapse
|
49
|
Detrimental or beneficial: the role of TRPM2 in ischemia/reperfusion injury. Acta Pharmacol Sin 2016; 37:4-12. [PMID: 26725732 DOI: 10.1038/aps.2015.141] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2015] [Accepted: 10/14/2015] [Indexed: 12/30/2022] Open
Abstract
Ischemia/reperfusion (I/R) injury is the main cause of tissue damage and dysfunction. I/R injury is characterized by Ca(2+) overload and production of reactive oxygen species (ROS), which play critical roles in the process of I/R injury to the brain, heart and kidney, but the underlying mechanisms are largely elusive. Recent evidence demonstrates that TRPM2, a Ca(2+)-permeable cationic channel and ROS sensor, is involved in I/R injury, but whether TRPM2 plays a protective or detrimental role in this process remains controversial. In this review, we discuss the recent progress in understanding the role of TRPM2 in reperfusion process after brain, heart and kidney ischemia and the potential of targeting TRPM2 for the development of therapeutic drugs to treat I/R injury.
Collapse
|
50
|
Miller BA, Cheung JY. TRPM2 protects against tissue damage following oxidative stress and ischaemia-reperfusion. J Physiol 2015; 594:4181-91. [PMID: 26420388 DOI: 10.1113/jp270934] [Citation(s) in RCA: 46] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2015] [Accepted: 09/13/2015] [Indexed: 12/17/2022] Open
Abstract
TRPM channels are a subgroup of the transient receptor potential (TRP) channel superfamily whose members have important roles in cell proliferation and survival. TRPM2, the second subfamily member to be cloned, is expressed in many tissues including brain, heart, vasculature and haematopoietic cells. TRPM2 is activated by oxidative stress and several other extracellular signals including tumour necrosis factor α (TNF-α) and amyloid β-peptide, which increase production of ADP-ribose (ADPR). ADPR binds to the TRPM2 C-terminal NUDT9-H domain, activating the channel. Early studies support the paradigm that TRPM2 activation induces cell death by sustained Ca(2+) influx or by enhancing cytokine production, aggravating inflammation and tissue injury. However, more recent data show that for a number of physiological processes, TRPM2 is protective. TRPM2 protects lungs from endotoxin-induced injury by reducing reactive oxygen species (ROS) production by phagocytes. It protects hearts from oxidative damage after ischaemia-reperfusion or hypoxia-reoxygenation by maintaining better mitochondrial bioenergetics and by decreasing ROS. Sustained Ca(2+) entry through TRPM2 is required to maintain cellular bioenergetics and protect against hypoxia-reoxygenation injury. TRPM2 also protects neuroblastoma from moderate oxidative stress by decreasing ROS through increased levels of forkhead box transcription factor 3a (FOXO3a) and a downstream effector, superoxide dismutase 2. TRPM2 is important for tumour growth and cell survival through modulation of hypoxia-inducible transcription factor expression, mitochondrial function and mitophagy. These findings in cardiac ischaemia and in neuroblastoma suggest that TRPM2 has a basic role in sustaining mitochondrial function and in cell survival that applies to a number of physiological systems and pathophysiological processes including ischaemia-reperfusion injury.
Collapse
Affiliation(s)
- Barbara A Miller
- Departments of Pediatrics and Biochemistry & Molecular Biology, Pennsylvania State University College of Medicine, Hershey, PA, 17033, USA
| | - Joseph Y Cheung
- Center of Translational Medicine and Department of Medicine, Temple University School of Medicine, Philadelphia, PA, 19140, USA
| |
Collapse
|