AccScience Publishing / AN / Volume 1 / Issue 1 / DOI: 10.36922/an.v1i1.3
Cite this article
143
Download
2132
Views
Journal Browser
Volume | Year
Issue
Search
News and Announcements
View All
REVIEW

Transient receptor potential melastatin 2 channels in neurological disorders: Mechanisms and animal models

Joe Steinman1† Andrea Ovcjak1† Zhengwei Luo1,2† Xinyang Zhang1,2 Luiz Roberto Britto3 Jeffrey T. Henderson4 Hong-Shuo Sun1,2,4* Zhong-Ping Feng1*
Show Less
1 Departments of Physiology, Temerty Faculty of Medicine, University of Toronto, Toronto, Ontario, Canada
2 Departments of Surgery, Temerty Faculty of Medicine, University of Toronto, Toronto, Ontario, Canada
3 Department of Physiology and Biophysics, Institute of Biomedical Sciences, University of São Paulo, Sao Paulo, Brazil
4 Leslie Dan Faculty of Pharmacy, University of Toronto, Toronto, Ontario, Canada
Advanced Neurology 2022, 1(1), 3 https://doi.org/10.36922/an.v1i1.3
Submitted: 18 December 2021 | Accepted: 17 March 2022 | Published: 8 April 2022
© 2022 by the Authors. This article is an open access article distributed under the terms and conditions of the Creative Commons Attribution 4.0 International License ( https://creativecommons.org/licenses/by/4.0/ )
Abstract

Transient receptor potential melastatin 2 (TRPM2) is a calcium-permeable ion channel implicated in neurodegenerative disorders and conditions. It is activated in response to reactive oxygen species (ROS) and thereby alters Ca2+ homeostasis and initiates pathways that lead to apoptosis and cell dysfunction. This review summarizes the current role of TRPM2 in neurological disorders, including Parkinson’s disease, Alzheimer’s disease, ischemia, traumatic brain injury, and depressive disorders (bipolar disease and depression). It describes the distribution and function of the TRPM2 channel across the brain and highlights common mechanisms between diseases. Specific animal and cell culture studies using TRPM2 inhibitors or genetic knockouts are discussed, including strategies to reduce the effect of ROS in disease through TRPM2 inhibition.

Keywords
Transient receptor potential melastatin 2
Ion channel
Neurological disorders
Brain injury
Stroke
Funding
Natural Sciences and Engineering Research Council of Canada
Canadian Institutes of Health Research
References
[1]

Steinman J, Sun HS, Feng ZP, 2020, Microvascular alterations in Alzheimer’s disease. Front Cell Neurosci, 14: 618986. https://doi.org/10.3389/fncel.2020.618986 

[2]

Wang Q, Liu N, Ni YS, et al., 2021, TRPM2 in ischemic stroke: Structure, molecular mechanisms, and drug intervention. Channels (Austin), 15(1): 136–154. https://doi.org/10.1080/19336950.2020.1870088

[3]

Steinman J, Cahill LS, Koletar MM, et al., 2019, Acute and chronic stage adaptations of vascular architecture and cerebral blood flow in a mouse model of TBI. Neuroimage, 202: 116101. https://doi.org/10.1016/j.neuroimage.2019.116101

[4]

Bolton AN, Saatman KE, 2014, Regional neurodegeneration and gliosis are amplified by mild traumatic brain injury repeated at 24-hour intervals. J Neuropathol Exp Neurol, 73(10): 933–947. https://doi.org/10.1097/NEN.0000000000000115

[5]

Braidy N, Muñoz P, Palacios AG, et al., 2012, Recent rodent models for Alzheimer’s disease: Clinical implications and basic research. J Neural Transm (Vienna), 119(2): 173–195. https://doi.org/10.1007/s00702-011-0731-5

[6]

Wang Y, Kilic E, Kilic U, et al., 2005, VEGF overexpression induces post-ischaemic neuroprotection, but facilitates haemodynamic steal phenomena. Brain, 128(Pt 1): 52–63. https://doi.org/10.1093/brain/awh325

[7]

Lange I, Yamamoto S, Partida-Sanchez S, et al., 2009, TRPM2 functions as a lysosomal Ca2+-release channel in beta cells. Sci Signal, 2(71): ra23. https://doi.org/10.1126/scisignal.2000278

[8]

Fleig A, Penner R, 2004, The TRPM ion channel subfamily: Molecular, biophysical and functional features. Trends Pharmacol Sci, 25(12): 633–639. https://doi.org/10.1016/j.tips.2004.10.004

[9]

Belrose JC, Jackson MF, 2018, TRPM2: A candidate therapeutic target for treating neurological diseases. Acta Pharmacol Sin, 39(5): 722–732. https://doi.org/10.1038/aps.2018.31

[10]

Ji D, Luo ZW, Ovcjak A, et al., 2021, Role of TRPM2 in brain tumours and potential as a drug target. Acta Pharmacol Sin, online ahead of print. https://doi.org/10.1038/s41401-021-00679-4

[11]

Huang S, Turlova E, Li F, et al., 2017, Transient receptor potential melastatin 2 channels (TRPM2) mediate neonatal hypoxic-ischemic brain injury in mice. Exp Neurol, 296: 32–40. https://doi.org/10.1016/j.expneurol.2017.06.023

[12]

Li F, Wong R, Luo Z, et al., 2019, Neuroprotective effects of AG490 in neonatal hypoxic-ischemic brain injury. Mol Neurobiol, 56(12): 8109–8123. https://doi.org/10.1007/s12035-019-01656-z

[13]

Allen Institute for Brain Science, 2004, Allen Mouse Brain Atlas, 2011 [dataset]. Available from: https://mouse.brain-map.org [Last accessed on 2021 Dec 16].

[14]

Allen Reference Atlas Mouse Brain [Brain Atlas]. Available from: https://www.atlas.brain-map.org [Last accessed on 2021 Dec 16].

[15]

Lein ES, Hawrylycz MJ, Ao N, et al., 2007, Genome-wide atlas of gene expression in the adult mouse brain. Nature, 445(7124): 168–176. https://doi.org/10.1038/nature05453

[16]

Turlova E, Feng ZP, Sun HS, 2018, The role of TRPM2 channels in neurons, glial cells and the blood-brain barrier in cerebral ischemia and hypoxia. Acta Pharmacol Sin, 39(5): 713–721. https://doi.org/10.1038/aps.2017.194

[17]

Fonfria E, Murdock PR, Cusdin FS, et al., 2006, Tissue distribution profiles of the human TRPM cation channel family. J Recept Signal Transduct Res, 26(3): 159–178. https://doi.org/10.1080/10799890600637506

[18]

Uemura T, Kudoh J, Noda S, et al., 2005, Characterization of human and mouse TRPM2 genes: Identification of a novel N-terminal truncated protein specifically expressed in human striatum. Biochem Biophys Res Commun, 328(4): 1232–1243. https://doi.org/10.1016/j.bbrc.2005.01.086

[19]

Emir TLR, 2017, Neurobiology of TRP Channels. CRC Press, Boca Raton, Florida. 

[20]

Jia J, Verma S, Nakayama S, et al., 2011, Sex differences in neuroprotection provided by inhibition of TRPM2 channels following experimental stroke. J Cereb Blood Flow Metab, 31(11): 2160–2168. https://doi.org/10.1038/jcbfm.2011.77

[21]

Belrose JC, Xie YF, Gierszewski LJ, et al., 2012, Loss of glutathione homeostasis associated with neuronal senescence facilitates TRPM2 channel activation in cultured hippocampal pyramidal neurons. Mol Brain, 5: 11. https://doi.org/10.1186/1756-6606-5-11

[22]

Song K, Wang H, Kamm GB, et al., 2016, The TRPM2 channel is a hypothalamic heat sensor that limits fever and can drive hypothermia. Science, 353(6306): 1393–1398. https://doi.org/10.1126/science.aaf7537

[23]

Kamm GB, Siemens J, 2017, The TRPM2 channel in temperature detection and thermoregulation. Temperature (Austin), 4(1): 21–23. https://doi.org/10.1080/23328940.2016.1258445

[24]

Xie YF, Belrose JC, Lei G, et al., 2011, Dependence of NMDA/ GSK-3β mediated metaplasticity on TRPM2 channels at hippocampal CA3-CA1 synapses. Mol Brain, 4: 44. https://doi.org/10.1186/1756-6606-4-44

[25]

Chung KK, Freestone PS, Lipski J, 2011, Expression and functional properties of TRPM2 channels in dopaminergic neurons of the substantia nigra of the rat. J Neurophysiol, 106(6): 2865–2875. https://doi.org/10.1152/jn.00994.2010

[26]

Roedding AS, Tong SY, Au-Yeung W, et al., 2013, Chronic oxidative stress modulates TRPC3 and TRPM2 channel expression and function in rat primary cortical neurons: Relevance to the pathophysiology of bipolar disorder. Brain Res, 1517: 16–27. https://doi.org/10.1016/j.brainres.2013.04.025

[27]

Kraft R, Grimm C, Grosse K, et al., 2004, Hydrogen peroxide and ADP-ribose induce TRPM2-mediated calcium influx and cation currents in microglia. Am J Physiol Cell Physiol, 286(1): C129–C137. https://doi.org/10.1152/ajpcell.00331.2003

[28]

Lee M, Cho T, Jantaratnotai N, et al., 2010, Depletion of GSH in glial cells induces neurotoxicity: Relevance to aging and degenerative neurological diseases. FASEB J, 24(7): 2533–2545. https://doi.org/10.1096/fj.09-149997

[29]

Heiner I, Eisfeld J, Warnstedt M, et al., 2006, Endogenous ADP-ribose enables calcium-regulated cation currents through TRPM2 channels in neutrophil granulocytes. Biochem J, 398(2): 225–232. https://doi.org/10.1042/BJ20060183

[30]

Yamamoto S, Shimizu S, Kiyonaka S, et al., 2008, TRPM2- mediated Ca2+ influx induces chemokine production in monocytes that aggravates inflammatory neutrophil infiltration. Nat Med, 14(7): 738–747. https://doi.org/10.1038/nm1758

[31]

Kashio M, Sokabe T, Shintaku K, et al., 2012, Redox signal-mediated sensitization of transient receptor potential melastatin 2 (TRPM2) to temperature affects macrophage functions. Proc Natl Acad Sci U S A, 109(17): 6745–6750. https://doi.org/10.1073/pnas.1114193109

[32]

Roedding AS, Gao AF, Au-Yeung W, et al., 2012, Effect of oxidative stress on TRPM2 and TRPC3 channels in B lymphoblast cells in bipolar disorder. Bipolar Disord, 14(2): 151–161. https://doi.org/10.1111/j.1399-5618.2012.01003.x

[33]

Hecquet CM, Zhang M, Mittal M, et al., 2014, Cooperative interaction of Trp melastatin channel transient receptor potential (TRPM2) with its splice variant TRPM2 short variant is essential for endothelial cell apoptosis. Circ Res, 114(3): 469–479. https://doi.org/10.1161/CIRCRESAHA.114.302414

[34]

Brown RC, Wu L, Hicks K, et al., 2008, Regulation of blood-brain barrier permeability by transient receptor potential Type C and Type V calcium-permeable channels.  Microcirculation, 15(4): 359–371. https://doi.org/10.1080/10739680701762656

[35]

Sumoza-Toledo A, Lange I, Cortado H, et al., 2011, Dendritic cell maturation and chemotaxis is regulated by TRPM2- mediated Lysosomal Ca2+ release. FASEB J, 25(10): 3529–3542. https://doi.org/10.1096/fj.10-178483

[36]

DeMaagd G, Philip A, 2015, Parkinson’s disease and its management: Part 1: Disease entity, risk factors, pathophysiology, clinical presentation, and diagnosis. P T, 40(8): 504–532.

[37]

Kin K, Yasuhara T, Kameda M, et al., 2019, Animal models for Parkinson’s disease research: Trends in the 2000s. Int J Mol Sci, 20(21): 5402. https://doi.org/10.3390/ijms20215402

[38]

Sun Y, Sukumaran P, Selvaraj S, et al., 2018, TRPM2 Promotes neurotoxin MPP+/MPTP-induced cell death. Mol Neurobiol, 55(1): 409–420. https://doi.org/10.1007/s12035-016-0338-9

[39]

Meredith GE, Rademacher DJ, 2011, MPTP mouse models of Parkinson’s disease: An update. J Parkinsons Dis, 1(1): 19–33. https://doi.org/10.3233/JPD-2011-11023

[40]

Malko P, Mortadza SA, McWilliam J, et al., 2019, TRPM2 channel in microglia as a new player in neuroinflammation associated with a spectrum of central nervous system pathologies. Front Pharmacol, 10: 239. https://doi.org/10.3389/fphar.2019.00239

[41]

Hermosura MC, Garruto RM, 2007, TRPM7 and TRPM2- candidate susceptibility genes for Western Pacific ALS and PD? Biochim Biophys Acta, 1772(8): 822–835. https://doi.org/10.1016/j.bbadis.2007.02.008

[42]

Hermosura MC, Cui AM, Go RC, et al., Altered functional properties of a TRPM2 variant in guamanian ALS and PD. Proc Natl Acad Sci U S A, 105(46): 18029–18034. https://doi.org/10.1073/pnas.0808218105

[43]

Vaidya B, Kaur H, Thapak P, et al., 2022, Pharmacological modulation of TRPM2 channels via PARP pathway leads to neuroprotection in MPTP-induced Parkinson’s disease in Sprague dawley rats. Mol Neurobiol, 59(3): 1528-1542.

[44]

Yu Y, Xie A, Yang S, 2014, TrkA pathway(s) are involves in the regulation of TRPM2 and TRPM7 expression in the substantia Nigra of the Parkinson’s disease rat model induced by 6-hydroxydopamine. Adv Res, 2: 782–796.

[45]

Nazıroğlu M, Özgül C, Çelik O, et al., 2011, Aminoethoxydiphenyl borate and flufenamic acid inhibit Ca2+ influx through TRPM2 channels in rat dorsal root ganglion neurons activated by ADP-ribose and rotenone. J Membr Biol, 241(2): 69–75. https://doi.org/10.1007/s00232-011-9363-9

[46]

Xicoy H, Wieringa B, Martens GJ, 2017, The SH-SY5Y cell line in Parkinson’s disease research: A systematic review. Mol Neurodegener, 12(1): 10. https://doi.org/10.1186/s13024-017-0149-0

[47]

Yıldızhan K, Nazıroğlu M, 2021, Protective role of selenium on MPP+ and homocysteine-induced TRPM2 channel activation in SH-SY5Y cells. J Recept Signal Transduct Res, online ahead of print. https://doi.org/10.1080/10799893.2021.1981381

[48]

An X, Fu Z, Mai C, et al., 2019, Increasing the TRPM2 channel expression in human neuroblastoma SH-SY5Y cells augments the susceptibility to ROS-induced cell death. Cells, 8(1): 1–8. https://doi.org/10.3390/cells8010028

[49]

Vaidya B, Sharma SS, 2020, Transient receptor potential channels as an emerging target for the treatment of Parkinson’s disease: An insight into role of pharmacological interventions. Front Cell Dev Biol, 8: 584513. https://doi.org/10.3389/fcell.2020.584513

[50]

Ding XM, Zhao LJ, Qiao HY, et al., 2019, Long Non-coding RNA-p21 regulates MPP+-induced neuronal injury by targeting miR-625 and depressing TRPM2 in SH-SY5Y cells. Chem Biol Interact, 307: 73–81. https://doi.org/10.1016/j.cbi.2019.04.017

[51]

Manoharan S, Guillemin GJ, Abiramasundari RS, et al., 2016, The role of reactive oxygen species in the pathogenesis of Alzheimer’s disease, Parkinson’s disease, and Huntington’s disease: A mini review. Oxid Med Cell Longev, 2016: 8590578. https://doi.org/10.1155/2016/8590578

[52]

Drummond E, Wisniewski T, 2017, Alzheimer’s disease: Experimental models and reality. Acta Neuropathol, 133(2): 155–175. https://doi.org/10.1007/s00401-016-1662-x

[53]

Chishti MA, Yang DS, Janus C, et al., 2001, Early-onset amyloid deposition and cognitive deficits in transgenic mice expressing a double mutant form of amyloid precursor protein 695. J Biol Chem, 276(24): 21562–21570. https://doi.org/10.1074/jbc.M100710200

[54]

Radde R, Bolmont T, Kaeser SA, et al., 2006, Abeta42-driven cerebral amyloidosis in transgenic mice reveals early and robust pathology. EMBO Rep, 7(9): 940–946. https://doi.org/10.1038/sj.embor.7400784

[55]

Lewis J, McGowan E, Rockwood J, et al., 2000, Neurofibrillary tangles, amyotrophy and progressive motor disturbance in mice expressing mutant (P301L) tau protein. Nat Genet, 25(4): 402–405. https://doi.org/10.1038/78078

[56]

Lewis J, Dickson DW, Lin WL, et al., 2001, Enhanced neurofibrillary degeneration in transgenic mice expressing mutant tau and APP. Science, 293(5534): 1487–1491. https://doi.org/10.1126/science.1058189

[57]

Kitazawa M, Medeiros R, Laferla FM, 2012, Transgenic mouse models of alzheimer disease: Developing a better model as a tool for therapeutic interventions. Curr Pharm Des, 18(8): 1131–1147. https://doi.org/10.2174/138161212799315786

[58]

Li X, Jiang LH, 2018, 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, 9(2): 195. https://doi.org/10.1038/s41419-018-0270-1 

[59]

Fonfria E, Marshall IC, Boyfield I, et al., 2005, Amyloid beta-peptide(1-42) and hydrogen peroxide-induced toxicity are mediated by TRPM2 in rat primary striatal cultures. J Neurochem, 95(3): 715–723. https://doi.org/10.1111/j.1471-4159.2005.03396.x

[60]

Li F, Abuarab N, Sivaprasadarao A, 2017, TRPM2: Shredding the mitochondrial network. Channels (Austin), 11(6): 507–509. https://doi.org/10.1080/19336950.2017.1376982

[61]

Ostapchenko VG, Chen M, Guzman MS, et al., 2015, The transient receptor potential melastatin 2 (TRPM2) channel contributes to β-amyloid oligomer-related neurotoxicity and memory impairment. J Neurosci, 35(45): 15157–15169. https://doi.org/10.1523/JNEUROSCI.4081-14.2015

[62]

Mortadza SA, Sim JA, Neubrand VE, et al., 2018, A critical role of TRPM2 channel in Aβ42-induced microglial activation and generation of tumor necrosis factor-α. Glia, 66(3): 562–575. https://doi.org/10.1002/glia.23265

[63]

Kakae M, Miyanohara J, Morishima M, et al., 2019, Pathophysiological role of TRPM2 in age-related cognitive impairment in mice. Neuroscience, 408: 204–213. https://doi.org/10.1016/j.neuroscience.2019.04.012

[64]

Jiang LH, Li X, Mortadza SA, et al., 2018, The TRPM2 channel nexus from oxidative damage to Alzheimer’s pathologies: An emerging novel intervention target for age-related dementia. Ageing Res Rev, 47: 67–79. https://doi.org/10.1016/j.arr.2018.07.002

[65]

Chugh C, 2019, Acute ischemic stroke: Management approach. Indian J Crit Care Med, 23(Suppl 2): S140–S146 https://doi.org/10.5005/jp-journals-10071-23192

[66]

McBride DW, Zhang JH, 2017, Precision stroke animal models: The permanent mcao model should be the primary model, not transient MCAO. Transl Stroke Res, 10: 554. https://doi.org/10.1007/s12975-017-0554-2

[67]

Fluri F, Schuhmann MK, Kleinschnitz C, 2015, Animal models of ischemic stroke and their application in clinical research. Drug Des Devel Ther, 9: 3445–3454. https://doi.org/10.2147/DDDT.S56071

[68]

Orset C, Macrez R, Young AR, et al., 2007, Mouse model of in situ thromboembolic stroke and reperfusion. Stroke, 38(10): 2771–2778. https://doi.org/10.1161/STROKEAHA.107.487520

[69]

Li H, Yang Z, Tang Q, et al., 2021, Embolic stroke model with magnetic nanoparticles. ACS Appl Mater Interfaces, 13(37): 43993–44001. https://doi.org/10.1021/acsami.1c11377

[70]

Uzdensky AB, 2018, Photothrombotic stroke as a model of ischemic stroke. Transl Stroke Res, 9(5): 437–451. https://doi.org/10.1007/s12975-017-0593-8

[71]

Traystman RJ, 2003, Animal models of focal and global cerebral ischemia. ILAR J, 44(2): 85–95. https://doi.org/10.1093/ilar.44.2.85

[72]

Smith ML, Bendek G, Dahlgren N, et al., 1984, Models for studying long-term recovery following forebrain ischemia in the rat. 2. A 2-vessel occlusion model. Acta Neurol Scand, 69(6): 385–401. https://doi.org/10.1111/j.1600-0404.1984.tb07822.x

[73]

Bacigaluppi M, Comi G, Hermann DM, 2010, Animal models of ischemic stroke. Part two: Modeling cerebral ischemia. Open Neurol J, 4: 34–38. https://doi.org/10.2174/1874205X01004020034

[74]

Fonfria E, Mattei C, Hill K, et al., 2006, TRPM2 is elevated in the tMCAO stroke model, transcriptionally regulated, and functionally expressed in C13 microglia. J Recept Signal Transduct Res, 26(3): 179–198. https://doi.org/10.1080/10799890600637522

[75]

Alim I, Teves L, Li R, et al., 2013, Modulation of NMDAR subunit expression by TRPM2 channels regulates neuronal vulnerability to ischemic cell death. J Neurosci, 33(44): 17264–17277. https://doi.org/10.1523/JNEUROSCI.1729-13.2013

[76]

Toda T, Yamamoto S, Umehara N, et al., 2019, Protective effects of duloxetine against cerebral ischemia-reperfusion injury via transient receptor potential melastatin 2 inhibition. J Pharmacol Exp Ther, 368(2): 246–254. https://doi.org/10.1124/jpet.118.253922

[77]

Gelderblom M, Melzer N, Schattling B, et al., 2014, Transient receptor potential melastatin subfamily member 2 cation channel regulates detrimental immune cell invasion in ischemic stroke. Stroke, 45(11): 3395–3402. https://doi.org/10.1161/STROKEAHA.114.005836

[78]

Wang GJ, Deng HY, Maier CM, et al., 2002, Mild hypothermia reduces ICAM-1 expression, neutrophil infiltration and microglia/monocyte accumulation following experimental stroke. Neuroscience, 114(4): 1081–1090. https://doi.org/10.1016/s0306-4522(02)00350-0 

[79]

Zhang Z, Chopp M, Powers C, 1997, Temporal profile of microglial response following transient (2h) middle cerebral artery occlusion. Brain Res, 744(2): 189–198. https://doi.org/10.1016/S0006-8993(96)01085-2

[80]

Shirakawa H, Sakimoto S, Nakagawa T, et al., 2014, Pathophysiology of immune cells during the progression of cerebral ischemic injury involvement of TRPM2-mediated induction of iNOS in microglia/macrophage. Nihon Yakurigaku Zasshi, 144(3): 104–109. https://doi.org/10.1254/fpj.144.104

[81]

Alkayed NJ, Harukuni I, Kimes AS, et al., 1998, Gender-linked brain injury in experimental stroke. Stroke, 29(1): 159–165; discussion 166. https://doi.org/10.1161/01.str.29.1.159

[82]

Shimizu T, Dietz RM, Cruz-Torres I, et al., 2016, Extended therapeutic window of a novel peptide inhibitor of TRPM2 channels following focal cerebral ischemia. Exp Neurol, 283(Pt A): 151–156. https://doi.org/10.1016/j.expneurol.2016.06.015

[83]

Shimizu T, Macey TA, Quillinan N, et al., 2013, Androgen and PARP-1 regulation of TRPM2 channels after ischemic injury. J Cereb Blood Flow Metab, 33(10): 1549–1555. https://doi.org/10.1038/jcbfm.2013.105

[84]

McCullough LD, Zeng Z, Blizzard KK, et al., 2005, Ischemic nitric oxide and poly (ADP-ribose) polymerase-1 in cerebral ischemia: Male toxicity, female protection. J Cereb Blood Flow Metab, 25(4): 502–512. https://doi.org/10.1038/sj.jcbfm.9600059

[85]

Quillinan N, Grewal H, Klawitter J, et al., 2014, Sex steroids do not modulate TRPM2-mediated injury in females following middle cerebral artery occlusion. eNeuro, 1(1): ENEURO.0022-14.2014. https://doi.org/10.1523/ENEURO.0022-14.2014

[86]

Ye M, Yang W, Ainscough JF, et al., 2014, TRPM2 Channel deficiency prevents delayed cytosolic Zn2+ accumulation and CA1 pyramidal neuronal death after transient global ischemia. Cell Death Dis, 5: e1541. https://doi.org/10.1038/cddis.2014.494

[87]

Hong DK, Kho AR, Lee SH, et al., 2020, Transient receptor potential melastatin 2 (TRPM2) inhibition by antioxidant, N-acetyl-l-cysteine, reduces global cerebral ischemia-induced neuronal death. Int J Mol Sci, 21(17): 6026. https://doi.org/10.3390/ijms21176026

[88]

Dietz RM, Cruz-Torres I, Orfila JE, et al., 2020, Reversal of global ischemia-induced cognitive dysfunction by delayed inhibition of TRPM2 ion channels. Transl Stroke Res, 11(2): 254–266. https://doi.org/10.1007/s12975-019-00712-z

[89]

Nakayama S, Vest R, Traystman RJ, et al., 2013, Sexually dimorphic response of TRPM2 inhibition following cardiac arrest-induced global cerebral ischemia in mice. J Mol Neurosci, 51(1): 92–98. https://doi.org/10.1007/s12031-013-0005-9

[90]

Werner C, Engelhard K, 2007, Pathophysiology of traumatic brain injury. Br J Anaesth, 99(1): 4–9. https://doi.org/10.1093/bja/aem131

[91]

Dewan MC, Rattani A, Gupta S, et al., 2018, Estimating the global incidence of traumatic brain injury. J Neurosurg, online ahead of print. https://doi.org/10.3171/2017.10.JNS17352

[92]

Xiong Y, Mahmood A, Chopp M, 2013, Animal models of traumatic brain injury. Nat Rev Neurosci, 14(2): 128–142. https://doi.org/10.1038/nrn3407

[93]

Hicks R, Soares H, Smith D, et al., 1996, Temporal and spatial characterization of neuronal injury following lateral fluid-percussion brain injury in the rat. Acta Neuropathol, 91(3): 236–246. https://doi.org/10.1007/s004010050421

[94]

Yürüker V, Nazıroğlu M, Şenol N, 2015, Reduction in traumatic brain injury-induced oxidative stress, apoptosis, and calcium entry in rat hippocampus by melatonin: Possible involvement of TRPM2 channels. Metab Brain Dis, 30(1): 223–231. https://doi.org/10.1007/s11011-014-9623-3

[95]

Cook NL, Vink R, Helps SC, et al., 2010, Transient receptor potential melastatin 2 expression is increased following experimental traumatic brain injury in rats. J Mol Neurosci, 42(2): 192–199. https://doi.org/10.1007/s12031-010-9347-8

[96]

Rubin TG, Lipton ML, 2019, Sex differences in animal models of traumatic brain injury. J Exp Neurosci, 13: 1179069519844020. https://doi.org/10.1177/1179069519844020

[97]

Yoon IS, Li PP, Siu KP, et al., 2001, Altered TRPC7 gene expression in bipolar-I disorder. Biol Psychiatry, 50(8): 620–626. https://doi.org/10.1016/s0006-3223(01)01077-0

[98]

Gao Y, Lei Z, Lu C, et al., 2010, Effect of ionic stress on apoptosis and the expression of TRPM2 in human olfactory neuroepithelial-derived progenitors. World J Biol Psychiatry, 11(8): 972–984. https://doi.org/10.3109/15622975.2010.507784

[99]

Jang Y, Lee SH, Lee B, et al. TRPM2, a susceptibility gene for bipolar disorder, regulates glycogen synthase kinase-3 activity in the brain. J Neurosci, 35(34): 11811–1123. https://doi.org/10.1523/JNEUROSCI.5251-14.2015

[100]

Ko SY, Wang SE, Lee HK, et al., 2019, Transient receptor potential melastatin 2 governs stress-induced depressive-like behaviors. Proc Natl Acad Sci U S A, 116(5): 1770–1775. https://doi.org/10.1073/pnas.1814335116

[101]

Akpinar A, Uğuz AC, Nazıroğlu M, 2014, Agomelatine and duloxetine synergistically modulates apoptotic pathway by inhibiting oxidative stress triggered intracellular calcium entry in neuronal PC12 cells: Role of TRPM2 and voltage-gated calcium channels. J Membr Biol, 247(5): 451–459. https://doi.org/10.1007/s00232-014-9652-1

[102]

Anderson IM, Haddad PM, Scott J, 2012, Bipolar disorder. BMJ, 345: e8508. https://doi.org/10.1136/bmj.e8508

[103]

Goldberg JF, Perlis RH, Bowden CL, et al., 2009, Manic symptoms during depressive episodes in 1,380 patients with bipolar disorder: Findings from the STEP-BD. Am J Psychiatry, 166(2): 173–181. https://doi.org/10.1176/appi.ajp.2008.08050746

[104]

Merikangas KR, Jin R, He JP, et al., 2011, Prevalence and correlates of bipolar spectrum disorder in the world mental health survey initiative. Arch Gen Psychiatry, 68(3): 241–251. https://doi.org/10.1001/archgenpsychiatry.2011.12

[105]

Sullivan PF, Daly MJ, O’Donovan M, 2012, Genetic architectures of psychiatric disorders: The emerging picture and its implications. Nat Rev Genet, 13(8): 537–551. https://doi.org/10.1038/nrg3240

[106]

Berk M, Dodd S, Kauer-Sant’anna M, et al., 2007, Dopamine dysregulation syndrome: Implications for a dopamine hypothesis of bipolar disorder. Acta Psychiatr Scand Suppl, 434: 41–49. https://doi.org/10.1111/j.1600-0447.2007.01058.x

[107]

Rao JS, Harry GJ, Rapoport SI, et al., Increased excitotoxicity and neuroinflammatory markers in postmortem frontal cortex from bipolar disorder patients. Mol Psychiatry, 15(4): 384–392. https://doi.org/10.1038/mp.2009.47

[108]

Andreazza AC, Wang JF, Salmasi F, et al., 2013, Specific subcellular changes in oxidative stress in prefrontal cortex from patients with bipolar disorder. J Neurochem, 127(4): 552–561. https://doi.org/10.1111/jnc.12316

[109]

Berk M, Kapczinski F, Andreazza AC, et al., 2011, pathways underlying neuroprogression in bipolar disorder: Focus on inflammation, oxidative stress and neurotrophic factors. Neurosci Biobehav Rev, 35(3): 804–817. https://doi.org/10.1016/j.neubiorev.2010.10.001

[110]

Xu C, Macciardi F, Li PP, et al., 2006, Association of the putative susceptibility gene, transient receptor potential protein melastatin Type 2, with bipolar disorder. Am J Med Genet B Neuropsychiatr Genet, 141B(1): 36–43. https://doi.org/10.1002/ajmg.b.30239

[111]

van Enkhuizen J, Minassian A, Young JW, 2013, Further evidence for ClockΔ19 mice as a model for bipolar disorder mania using cross-species tests of exploration and sensorimotor gating. Behav Brain Res, 249: 44–54. https:// doi.org/10.1016/j.bbr.2013.04.023

[112]

van Enkhuizen J, Henry BL, Minassian A, et al., 2014, Reduced dopamine transporter functioning induces high-reward risk-preference consistent with bipolar disorder. Neuropsychopharmacology, 39(13): 3112–3122. https://doi.org/10.1038/npp.2014.170

[113]

Young JW, Cope ZA, Romoli B, et al., 2018, Mice with reduced DAT levels recreate seasonal-induced switching between states in bipolar disorder. Neuropsychopharmacology, 43(8): 1721–1731. https://doi.org/10.1038/s41386-018-0031-y

[114]

Sonntag KC, Brenhouse HC, Freund N, et al., 2014, Viral over-expression of D1 dopamine receptors in the prefrontal cortex increase high-risk behaviors in adults: Comparison with adolescents. Psychopharmacology (Berl), 231(8): 1615–1626. https://doi.org/10.1007/s00213-013-3399-8

[115]

Borison RL, Sabelli HC, Maple PJ, et al., 1978, Lithium prevention of amphetamine-induced “manic” excitement and of reserpine-induced “depression” in mice: Possible role of 2-phenylethylamine. Psychopharmacology (Berl), 59(3): 259–262. https://doi.org/10.1007/BF00426631

[116]

Rezin GT, Furlanetto CB, Scaini G, et al., 2014, Fenproporex increases locomotor activity and alters energy metabolism, and mood stabilizers reverse these changes: A proposal for a new animal model of mania. Mol Neurobiol, 49(2): 877–892. https://doi.org/10.1007/s12035-013-8566-8

[117]

Markou A, Koob GF, 1991, Postcocaine anhedonia. An animal model of cocaine withdrawal. Neuropsychopharmacology, 4(1): 17–26.

[118]

Harrison PJ, Cipriani A, Harmer CJ, et al., 2016, Innovative approaches to bipolar disorder and its treatment. Ann N Y Acad Sci, 1366(1): 76–89. https://doi.org/10.1111/nyas.13048

[119]

Freund N, Juckel G, 2019, Bipolar disorder: Its etiology and how to model in rodents. Methods Mol Biol, 2011: 61–77. https://doi.org/10.1007/978-1-4939-9554-7_4

[120]

Kostyrko A, Hauser J, Rybakowski JK, et al., 2006, Screening of chromosomal region 21q22.3 for mutations in genes associated with neuronal Ca2+ signalling in bipolar affective disorder. Acta Biochim Pol, 53(2): 317–320.

[121]

McQuillin A, Bass NJ, Kalsi G, et al., 2006, Fine mapping of a susceptibility locus for bipolar and genetically related unipolar affective disorders, to a region containing the C21ORF29 and TRPM2 genes on chromosome 21q22.3. Mol Psychiatry, 11(2): 134–142. https://doi.org/10.1038/sj.mp.4001759

[122]

Xu C, Li PP, Cooke RG, et al., 2009, TRPM2 Variants and bipolar disorder risk: Confirmation in a family-based association study. Bipolar Disord, 11(1): 1–10. https://doi.org/10.1111/j.1399-5618.2008.00655.x

[123]

Xu C, Warsh JJ, Wang KS, et al., 2013, Association of the iPLA2β gene with bipolar disorder and assessment of its interaction with TRPM2 gene polymorphisms. Psychiatr Genet, 23(2): 86–89. https://doi.org/10.1097/YPG.0b013e32835d700d 

[124]

Depression. Available from: https://www.who.int/news-room/fact-sheets/detail/depression [Last accessed on 2021 Dec 16].

[125]

DSM-5. Available from: https://www.psychiatry.org/ psychiatrists/practice/dsm [Last accessed on 2021 Dec 16]. 

[126]

Post RM, Denicoff KD, Leverich GS, et al., 2003, Morbidity in 258 bipolar outpatients followed for 1 year with daily prospective ratings on the NIMH life chart method. J Clin Psychiatry, 64(6): 680–690; quiz 738–739. https://doi.org/10.4088/jcp.v64n0610

[127]

Lee S, Jeong J, Kwak Y, et al., 2010, Depression research: Where are we now? Mol Brain, 3: 8. https://doi.org/10.1186/1756-6606-3-8

[128]

Morilak DA, Frazer A, 2004, Antidepressants and brain monoaminergic systems: A dimensional approach to understanding their behavioural effects in depression and anxiety disorders. Int J Neuropsychopharmacol, 7(2): 193–218. https://doi.org/10.1017/S1461145704004080

[129]

Juszczyk G, Mikulska J, Kasperek K, et al., 2021, Chronic stress and oxidative stress as common factors of the pathogenesis of depression and Alzheimer’s disease: The role of antioxidants in prevention and treatment. Antioxidants (Basel), 10(9): L1439. https://doi.org/10.3390/antiox10091439

[130]

Maeng SH, Hong H, 2019, Inflammation as the potential basis in depression. Int Neurourol J, 23(Suppl 2): S63–S71. https://doi.org/10.5213/inj.1938226.113

[131]

Willner P, 2017, The chronic mild stress (CMS) model of depression: History, evaluation and usage. Neurobiol Stress, 6: 78–93. https://doi.org/10.1016/j.ynstr.2016.08.002

[132]

Fox ME, Lobo MK, 2019, The molecular and cellular mechanisms of depression: A focus on reward circuitry. Mol Psychiatry, 24(12): 1798–1815. https://doi.org/10.1038/s41380-019-0415-3

[133]

Krishnan V, Nestler EJ, 2011, Animal models of depression: Molecular perspectives. Curr Top Behav Neurosci, 7: 121–147. https://doi.org/10.1007/7854_2010_108

[134]

Das R, Emon PZ, Shahriar M, et al., 2021, Higher levels of serum IL-1β and TNF-α are associated with an increased probability of major depressive disorder. Psychiatry Res, 295: 113568. https://doi.org/10.1016/j.psychres.2020.113568

[135]

Köhler O, Benros ME, Nordentoft M, et al., 2014, Effect of anti-inflammatory treatment on depression, depressive symptoms, and adverse effects: A systematic review and meta-analysis of randomized clinical trials. JAMA Psychiatry, 71(12): 1381–1391. https://doi.org/10.1001/jamapsychiatry.2014.1611

[136]

Colaianna M, Schiavone S, Zotti M, et al., 2013, Neuroendocrine profile in a rat model of psychosocial stress: Relation to oxidative stress. Antioxid Redox Signal, 18(12): 1385–1399. https://doi.org/10.1089/ars.2012.4569

[137]

Schiavone S, Sorce S, Dubois-Dauphin M, et al., 2009, Involvement of NOX2 in the Development of Behavioral and Pathologic Alterations in Isolated Rats. Biol Psychiatry, 66(4): 384–392. https://doi.org/10.1016/j.biopsych.2009.04.033

[138]

Luo X, Li M, Zhan K, et al., 2018, Selective inhibition of TRPM2 channel by two novel synthesized ADPR analogues. Chem Biol Drug Des, 91(2): 552–566. https://doi.org/10.1111/cbdd.13119

[139]

Hu HZ, Gu Q, Wang C, et al., 2004, 2-aminoethoxydiphenyl Borate is a Common Activator of TRPV1, TRPV2, and TRPV3. J Biol Chem, 279(34): 35741–35748. https://doi.org/10.1074/jbc.M404164200

[140]

Qi Z, Shi W, Zhao Y, et al., 2019, Zinc accumulation in mitochondria promotes ischemia-induced BBB disruption through Drp1-dependent mitochondria fission. Toxicol Appl Pharmacol, 377: 114601. https://doi.org/10.1016/j.taap.2019.114601

[141]

Manna PT, Munsey TS, Abuarab N, et al., 2015, TRPM2- mediated intracellular Zn2+ release triggers pancreatic β-cell death. Biochem J, 466(3): 537–546. https://doi.org/10.1042/BJ20140747

[142]

Feng Y, Wang X, 2012, Antioxidant therapies for Alzheimer’s disease. Oxid Med Cell Longev, 2012: 472932. https://doi.org/10.1155/2012/472932

[143]

Naziroğlu M, Lückhoff A, 2008, Effects of antioxidants on calcium influx through TRPM2 channels in transfected cells activated by hydrogen peroxide. J Neurol Sci, 270(1-2): 152–158. https://doi.org/10.1016/j.jns.2008.03.003

Conflict of interest
The authors have no competing interest to declare.
Share
Back to top
Advanced Neurology, Electronic ISSN: 2810-9619 Published by AccScience Publishing