1,463
Views
0
CrossRef citations to date
0
Altmetric
Review

Roles of TRPM channels in glioma

, , , , , , , , ORCID Icon & show all
Article: 2338955 | Received 03 Dec 2023, Accepted 01 Apr 2024, Published online: 29 Apr 2024

References

  • Lee A, Fakler B, Kaczmarek LK, Isom LL. More than a pore: ion channel signaling complexes. J Neurosci. 2014;34(46):15159–16. doi:10.1523/JNEUROSCI.3275-14.2014.
  • Jimenez I, Prado Y, Marchant F, Otero C, Eltit F, Cabello-Verrugio C, Cerda O, Simon F. TRPM channels in human diseases. Cells-Basel. 2020;9(12):2604. doi:10.3390/cells9122604.
  • Litan A, Langhans SA. Cancer as a channelopathy: ion channels and pumps in tumor development and progression. Front Cell Neurosci. 2015;9:86. doi:10.3389/fncel.2015.00086.
  • Qian LL, Sun X, Yang J, Wang X-L, Ackerman MJ, Wang R-X, Xu X, Lee H-C, Lu T. Changes in ion channel expression and function associated with cardiac arrhythmogenic remodeling by sorbs2. Biochim Biophys Acta Mol Basis Dis. 2021;1867(12):166247. doi:10.1016/j.bbadis.2021.166247.
  • Brierley S. Altered ion channel/receptor expression and function in extrinsic sensory neurons: the cause of and solution to chronic visceral pain? Adv Exp Med Biol. 2016;891:75–90. doi:10.1007/978-3-319-27592-5_9.
  • Sakaguchi R, Mori Y. Transient receptor potential (trp) channels: biosensors for redox environmental stimuli and cellular status. Free Radic Biol Med. 2020;146:36–44. doi:10.1016/j.freeradbiomed.2019.10.415.
  • Zhang Z, Toth B, Szollosi A, Chen J, Csanády L. Structure of a TRPM2 channel in complex with Ca(2+) explains unique gating regulation. Elife. 2018;7. doi:10.7554/eLife.36409.
  • Chen Y, Zhang X, Yang T, Bi R, Huang Z, Ding H, Li J, Zhang J. Emerging structural biology of trpm subfamily channels. Cell Calcium. 2019;79:75–9. doi:10.1016/j.ceca.2019.02.011.
  • Lopez-Romero AE, Hernandez-Araiza I, Torres-Quiroz F, Tovar-Y-Romo LB, Islas LD, Rosenbaum T. Trp ion channels: proteins with conformational flexibility. Channels (Austin). 2019;13(1):207–26. doi:10.1080/19336950.2019.1626793.
  • Huang Y, Fliegert R, Guse AH, Lü W, Du J. A structural overview of the ion channels of the trpm family. Cell Calcium. 2020;85:102111. doi:10.1016/j.ceca.2019.102111.
  • Held K, Voets T, Vriens J. TRPM3 in temperature sensing and beyond. Temp (Austin). 2015;2(2):201–13. doi:10.4161/23328940.2014.988524.
  • Liu Z, Wu H, Wei Z, Wang X, Shen P, Wang S, Wang A, Chen W, Lu Y. Trpm8: a potential target for cancer treatment. J Cancer Res Clin Oncol. 2016;142(9):1871–81. doi:10.1007/s00432-015-2112-1.
  • Zholos A, Johnson C, Burdyga T, Melanaphy D. TRPM channels in the vasculature. Adv Exp Med Biol. 2011;704:707–29. 10.1007/978-94-007-0265-3_37
  • Belrose JC, Jackson MF. TRPM2: a candidate therapeutic target for treating neurological diseases. Acta Pharmacol Sin. 2018;39(5):722–32. doi:10.1038/aps.2018.31.
  • Zielinska W, Zabrzynski J, Gagat M, Grzanka A. The role of trpm2 in endothelial function and dysfunction. Int J Mol Sci. 2021;22(14):7635. doi:10.3390/ijms22147635.
  • Schneider FM, Mohr F, Behrendt M, Oberwinkler J. Properties and functions of trpm1 channels in the dendritic tips of retinal on-bipolar cells. European Journal of Cell Biology. 2015;94(7–9):420–7. doi:10.1016/j.ejcb.2015.06.005.
  • Simon F, Varela D, Cabello-Verrugio C. Oxidative stress-modulated trpm ion channels in cell dysfunction and pathological conditions in humans. Cell Signal. 2013;25(7):1614–24. doi:10.1016/j.cellsig.2013.03.023.
  • Wong KK, Banham AH, Yaacob NS, Nur Husna SM. The oncogenic roles of TRPM ion channels in cancer. J Cell Physiol. 2019;234(9):14556–73. doi:10.1002/jcp.28168.
  • Santoni G, Maggi F, Morelli MB, Santoni M, Marinelli O. Transient receptor potential cation channels in cancer therapy. Med Sci (Basel). 2019;7(12):108. doi:10.3390/medsci7120108.
  • Hantute-Ghesquier A, Haustrate A, Prevarskaya N, Lehen’kyi V. Trpm family channels in cancer. Pharmaceuticals (Basel). 2018;11(2):11. doi:10.3390/ph11020058.
  • Shahmir S, Zahmatkesh N, Mirzaahmadi S, Asaadi Tehrani G. LncRNA CASC2 inhibits progression of glioblastoma by regulating the expression of AKT in T98G cell line, treated by TMZ and thiosemicarbazone complex. Asian Pac J Cancer Prev. 2023;24(5):1553–60. doi:10.31557/APJCP.2023.24.5.1553.
  • Persano F, Gigli G, Leporatti S. Natural compounds as promising adjuvant agents in the treatment of gliomas. Int J Mol Sci. 2022;23(6):3360. doi:10.3390/ijms23063360.
  • Kim BJ. Involvement of transient receptor potential melastatin 7 channels in sophorae radix-induced apoptosis in cancer cells: sophorae radix and trpm7. J Pharmacopuncture. 2012;15(3):31–8. doi:10.3831/KPI.2012.15.003.
  • Leng TD, Li MH, Shen JF, Liu M-L, Li X-B, Sun H-W, Branigan D, Zeng Z, Si H-F, Li J, et al. Suppression of trpm7 inhibits proliferation, migration, and invasion of malignant human glioma cells. CNS Neurosci Ther. 2015; 21(3):252–261. doi:10.1111/cns.12354.
  • Elias AF, Lin BC, Piggott BJ. Ion channels in gliomas—from molecular basis to treatment. Int J Mol Sci. 2023;24(3):2530. doi:10.3390/ijms24032530.
  • Lefranc F. Transient receptor potential (trp) ion channels involved in malignant glioma cell death and therapeutic perspectives. Front Cell Dev Biol. 2021;9:618961. doi:10.3389/fcell.2021.618961.
  • Chen WL, Barszczyk A, Turlova E, Deurloo M, Liu B, Yang BB, Rutka JT, Feng Z-P, Sun H-S. Inhibition of trpm7 by carvacrol suppresses glioblastoma cell proliferation, migration and invasion. Oncotarget. 2015;6(18):16321–40. doi:10.18632/oncotarget.3872.
  • So JS, Kim H, Han KS. Mechanisms of invasion in glioblastoma: extracellular matrix, Ca(2+) signaling, and glutamate. Front Cell Neurosci. 2021;15:663092. doi:10.3389/fncel.2021.663092.
  • Samanta A, Hughes T, Moiseenkova-Bell VY. Transient receptor potential (trp) channels. Subcell Biochem. 2018;87:141–65. doi:10.1007/978-981-10-7757-9_6.
  • Kaneko Y, Szallasi A. Transient receptor potential (TRP) channels: a clinical perspective. Br J Pharmacol. 2014;171(10):2474–507. doi:10.1111/bph.12414.
  • Katz B, Payne R, Minke B. TRP channels in vision. 2017:27–63. 10.4324/9781315152837-3.
  • Fleisch VC, Jametti T, Neuhauss SC. Electroretinogram (ERG) measurements in larval zebrafish. CSH Protoc. 2008;2008(3):t4973. doi:10.1101/pdb.prot4973.
  • Venkatraman P, Mills-Henry I, Padmanabhan KR, Pascuzzi P, Hassan M, Zhang J, Zhang X, Ma P, Pang CP, Dowling JE, et al. Rods contribute to visual behavior in larval zebrafish. Invest Ophthalmol Vis Sci. 2020; 61(12):11. doi:10.1167/iovs.61.12.11.
  • Montell C, Rubin GM. Molecular characterization of the drosophila trp locus: a putative integral membrane protein required for phototransduction. Neuron. 1989;2(4):1313–23. doi:10.1016/0896-6273(89)90069-x.
  • Minke B. The history of the drosophila trp channel: the birth of a new channel superfamily. J Neurogenet. 2010;24(4):216–33. doi:10.3109/01677063.2010.514369.
  • Wes PD, Chevesich J, Jeromin A, Rosenberg C, Stetten G, Montell C. Trpc1, a human homolog of a drosophila store-operated channel. Proc Natl Acad Sci USA. 1995;92(21):9652–6. doi:10.1073/pnas.92.21.9652.
  • Chen S, Chiu CN, McArthur KL, Fetcho JR, Prober DA. Trp channel mediated neuronal activation and ablation in freely behaving zebrafish. Nat Methods. 2016;13(2):147–50. doi:10.1038/nmeth.3691.
  • Bernardini M, Fiorio PA, Prevarskaya N, Gkika D. Human transient receptor potential (trp) channel expression profiling in carcinogenesis. Int J Dev Biol. 2015;59(7–8–9):399–406. doi:10.1387/ijdb.150232dg.
  • Desai BN, Clapham DE. Trp channels and mice deficient in trp channels. Pflugers Arch Eur J Physiol. 2005;451(1):11–18. doi:10.1007/s00424-005-1429-z.
  • Venkatachalam K, Luo J, Montell C. Evolutionarily conserved, multitasking trp channels: lessons from worms and flies. Handb Exp Pharmacol. 2014;223:937–62. doi:10.1007/978-3-319-05161-1_9.
  • Pedersen SF, Owsianik G, Nilius B. Trp channels: an overview. Cell Calcium. 2005;38(3–4):233–52. doi:10.1016/j.ceca.2005.06.028.
  • Clapham DE, Runnels LW, Strubing C. The trp ion channel family. Nat Rev Neurosci. 2001;2(6):387–96. doi:10.1038/35077544.
  • Liao M, Cao E, Julius D, Cheng Y. Structure of the TRPV1 ion channel determined by electron cryo-microscopy. Nature. 2013;504(7478):107–12. doi:10.1038/nature12822.
  • Miller BA. TRPC2. Handb Exp Pharmacol. 2014;222:53–65. doi:10.1007/978-3-642-54215-2_3.
  • Dietrich A. Transient receptor potential (trp) channels in health and disease. Cells-Basel. 2019;8. doi:10.3390/cells8050413.
  • Haustrate A, Prevarskaya N, Lehen’kyi V. Role of the trpv channels in the endoplasmic reticulum calcium homeostasis. Cells-Basel. 2020;9(2):317. doi:10.3390/cells9020317.
  • Smani T, Shapovalov G, Skryma R, Prevarskaya N, Rosado JA. Functional and physiopathological implications of trp channels. Biochimica et Biophysica Acta (BBA) – Mol Cell Res. 2015;1853(8):1772–82. doi:10.1016/j.bbamcr.2015.04.016.
  • Takahashi N, Mori Y. Trp channels as sensors and signal integrators of redox status changes. Front Pharmacol. 2011;2:58. doi:10.3389/fphar.2011.00058.
  • Theile JW, Cummins TR. Recent developments regarding voltage-gated sodium channel blockers for the treatment of inherited and acquired neuropathic pain syndromes. Front Pharmacol. 2011;2:54. doi:10.3389/fphar.2011.00054.
  • Choi JH, Jeong SY, Oh MR, Allen PD, Lee EH. Trpcs: influential mediators in skeletal muscle. Cells-Basel. 2020;9(4):850. doi:10.3390/cells9040850.
  • Cao E. Structural mechanisms of transient receptor potential ion channels. J Gen Physiol. 2020;152(3):152. doi:10.1085/jgp.201811998.
  • Gwanyanya A, Mubagwa K. Emerging role of transient receptor potential (trp) ion channels in cardiac fibroblast pathophysiology. Front Physiol. 2022;13:968393. doi:10.3389/fphys.2022.968393.
  • Nagarkatti N, Deshpande LS, DeLorenzo RJ. Development of the calcium plateau following status epilepticus: role of calcium in epileptogenesis. Expert Rev Neurother. 2009;9:813–24. doi:10.1586/ern.09.21.
  • Nishimura H, Kawasaki M, Tsukamoto M, Menuki K, Suzuki H, Matsuura T, Baba K, Motojima Y, Fujitani T, Ohnishi H, et al. Transient receptor potential vanilloid 1 and 4 double knockout leads to increased bone mass in mice. Bone Rep. 2020;12:100268. doi:10.1016/j.bonr.2020.100268.
  • Marquez-Nogueras KM, Hortua TM, Chasen NM, Kuo IY, Moreno SN. Calcium signaling through a transient receptor channel is important for toxoplasma gondii growth. Elife. 2021;10. doi:10.7554/eLife.63417.
  • Young A, Machacek DW, Dhara SK, MacLeish PR, Benveniste M, Dodla MC, Sturkie CD, Stice SL. Ion channels and ionotropic receptors in human embryonic stem cell derived neural progenitors. Neuroscience. 2011;192:793–805. doi:10.1016/j.neuroscience.2011.04.039.
  • Caterina MJ, Pang Z. Trp channels in skin biology and pathophysiology. Pharmaceuticals (Basel). 2016;9(4):77. doi:10.3390/ph9040077.
  • Peralvarez-Marin A, Donate-Macian P, Gaudet R. What do we know about the transient receptor potential vanilloid 2 (trpv2) ion channel? FEBS J. 2013;280(21):5471–87. doi:10.1111/febs.12302.
  • Nilius B, Owsianik G. The transient receptor potential family of ion channels. Genome Biol. 2011;12(3):218. doi:10.1186/gb-2011-12-3-218.
  • Xu X, Yu C, Xu L, Xu J. Emerging roles of keratinocytes in nociceptive transduction and regulation. Front Mol Neurosci. 2022;15:982202. doi:10.3389/fnmol.2022.982202.
  • Wan J, Guo AA, Chowdhury I, Guo S, Hibbert J, Wang G, Liu M. TRPM7 induces mechanistic target of rap1b through the downregulation of mir-28-5p in glioma proliferation and invasion. Front Oncol. 2019;9:1413. doi:10.3389/fonc.2019.01413.
  • Santoni G, Amantini C, Maggi F, Marinelli O, Santoni M, Nabissi M, Morelli MB. The trpv2 cation channels: from urothelial cancer invasiveness to glioblastoma multiforme interactome signature. Lab Invest. 2020;100(2):186–98. doi:10.1038/s41374-019-0333-7.
  • Chinigo G, Castel H, Chever O, Gkika D. Trp channels in brain tumors. Front Cell Dev Biol. 2021;9:617801. doi:10.3389/fcell.2021.617801.
  • Aroke EN, Powell-Roach KL, Jaime-Lara RB, Tesfaye M, Roy A, Jackson P, Joseph P. Taste the pain: the role of trp channels in pain and taste perception. Int J Mol Sci. 2020;21(16):5929. doi:10.3390/ijms21165929.
  • Ishimaru Y, Matsunami H. Transient receptor potential (trp) channels and taste sensation. J Dent Res. 2009;88(3):212–8. doi:10.1177/0022034508330212.
  • Shi R, Fu Y, Zhao D, Boczek T, Wang W, Guo F. Cell death modulation by transient receptor potential melastatin channels trpm2 and trpm7 and their underlying molecular mechanisms. Biochem Pharmacol. 2021;190:114664. doi:10.1016/j.bcp.2021.114664.
  • Castillo K, Diaz-Franulic I, Canan J, Gonzalez-Nilo F, Latorre R. Thermally activated trp channels: molecular sensors for temperature detection. Phys Biol. 2018;15(2):21001. doi:10.1088/1478-3975/aa9a6f.
  • Li T, Chen J, Zeng Z. Pathophysiological role of calcium channels and transporters in the multiple myeloma. Cell Commun Signal. 2021;19(1):99. doi:10.1186/s12964-021-00781-4.
  • Kunert-Keil C, Bisping F, Kruger J, Brinkmeier H. Tissue-specific expression of trp channel genes in the mouse and its variation in three different mouse strains. BMC Genomics. 2006;7(1):159. doi:10.1186/1471-2164-7-159.
  • Cohen MR, Moiseenkova-Bell VY. Structure of thermally activated trp channels. Curr Top Membr. 2014;74:181–211. doi:10.1016/B978-0-12-800181-3.00007-5.
  • Mei ZZ, Xia R, Beech DJ, Jiang L-H. Intracellular coiled-coil domain engaged in subunit interaction and assembly of melastatin-related transient receptor potential channel 2. J Biol Chem. 2006;281(50):38748–56. doi:10.1074/jbc.M607591200.
  • Guo J, She J, Zeng W, Chen Q, Bai X-C, Jiang Y. Structures of the calcium-activated, non-selective cation channel trpm4. Nature. 2017;552(7684):205–9. doi:10.1038/nature24997.
  • Malko P, Jiang LH. Trpm2 channel-mediated cell death: an important mechanism linking oxidative stress-inducing pathological factors to associated pathological conditions. Redox Biol. 2020;37:101755. doi:10.1016/j.redox.2020.101755.
  • Combe CL, Upchurch CM, Canavier CC, Gasparini S. Cholinergic modulation shifts the response of ca1 pyramidal cells to depolarizing ramps via trpm4 channels with potential implications for place field firing. Elife. 2023;12. doi:10.7554/eLife.84387.
  • Guo H, Carlson JA, Slominski A. Role of trpm in melanocytes and melanoma. Exp Dermatol. 2012;21(9):650–4. doi:10.1111/j.1600-0625.2012.01565.x.
  • Canales J, Morales D, Blanco C, Rivas J, Cerda O. A TR(I)P to cell migration: new roles of trp channels in mechanotransduction and cancer. Front Physiol. 2019;10:757. doi:10.3389/fphys.2019.00757.
  • Bouron A, Kiselyov K, Oberwinkler J. Permeation, regulation and control of expression of trp channels by trace metal ions. Pflugers Arch Eur J Physiol. 2015;467(6):1143–164. doi:10.1007/s00424-014-1590-3.
  • Turlova E, Feng ZP, Sun HS. The role of trpm2 channels in neurons, glial cells and the blood-brain barrier in cerebral ischemia and hypoxia. Acta Pharmacol Sin. 2018;39(5):713–21. doi:10.1038/aps.2017.194.
  • Yu P, Cai X, Liang Y, Wang M, Yang W. Roles of NAD(+) and its metabolites regulated calcium channels in cancer. Molecules. 2020;25(20):4826. doi:10.3390/molecules25204826.
  • Rosenbaum T. Activators of trpm2: getting it right. J Gen Physiol. 2015;145(6):485–7. doi:10.1085/jgp.201511405.
  • Pires PW, Earley S. Redox regulation of transient receptor potential channels in the endothelium. Microcirculation. 2017;24(3). doi:10.1111/micc.12329.
  • Miller BA. Trpm2 in cancer. Cell Calcium. 2019;80:8–17. doi:10.1016/j.ceca.2019.03.002.
  • Syed MS, Wang L, Li D, Jiang L-H. Trpm2 channel-mediated ros-sensitive CA(2+) signaling mechanisms in immune cells. Front Immunol. 2015;6:407. doi:10.3389/fimmu.2015.00407.
  • Hirschler-Laszkiewicz I, Chen SJ, Bao L, Wang J, Zhang X-Q, Shanmughapriya S, Keefer K, Madesh M, Cheung JY, Miller BA, et al. 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(4):C571–C586. doi:10.1152/ajpcell.00098.2018.
  • Tan CH, McNaughton PA. Trpm2 and warmth sensation. Pflugers Arch - Eur J Physiol. 2018;470(5):787–798. doi:10.1007/s00424-018-2139-7.
  • Starkus J, Beck A, Fleig A, Penner R. Regulation of trpm2 by extra- and intracellular calcium. J Gen Physiol. 2007;130(4):427–440. doi:10.1085/jgp.200709836.
  • Jiang LH, Gamper N, Beech DJ. Properties and therapeutic potential of transient receptor potential channels with putative roles in adversity: focus on TRPC5, TRPM2 and TRPA1. Curr Drug Targets. 2011;12(5):724–736. doi:10.2174/138945011795378568.
  • Hu X, Wu L, Liu X, Zhang Y, Xu M, Fang Q, Lu L, Niu J, Abd El-Aziz TM, Jiang L-H, et al. Deficiency of ros-activated trpm2 channel protects neurons from cerebral ischemia-reperfusion injury through upregulating autophagy. Oxid Med Cell Longev. 2021;2021:1–12. doi:10.1155/2021/7356266.
  • Ferrandiz-Huertas C, Mathivanan S, Wolf CJ, Devesa I, Ferrer-Montiel A. Trafficking of thermotrp channels. Membranes (Basel). 2014;4(3):525–564. doi:10.3390/membranes4030525.
  • Cabanas H, Muraki K, Staines D, Marshall-Gradisnik S. Naltrexone restores impaired transient receptor potential melastatin 3 ion channel function in natural killer cells from myalgic encephalomyelitis/chronic fatigue syndrome patients. Front Immunol. 2019;10:2545. doi:10.3389/fimmu.2019.02545.
  • Becker A, Gotz C, Montenarh M, Philipp SE. Control of TRPM3 Ion Channels by Protein Kinase CK2-Mediated Phosphorylation in Pancreatic β-Cells of the Line INS-1. Int J Mol Sci. 2021;22(23):22. doi:10.3390/ijms222313133.
  • Liu Y, Lyu Y, Wang H. Trp channels as molecular targets to relieve endocrine-related diseases. Front Mol Biosci. 2022;9:895814. doi:10.3389/fmolb.2022.895814.
  • Nilius B, Vennekens R. From cardiac cation channels to the molecular dissection of the transient receptor potential channel trpm4. Pflugers Arch Eur J Physiol. 2006;453(3):313–321. doi:10.1007/s00424-006-0088-z.
  • Liman ER. Trpm5 and taste transduction. Handb Exp Pharmacol. 2007:287–298. doi:10.1007/978-3-540-34891-7_17.
  • Kurland DB, Gerzanich V, Karimy JK, Woo SK, Vennekens R, Freichel M, Nilius B, Bryan J, Simard JM. The SUR1-TRPM4 channel regulates nos2 transcription in tlr4-activated microglia. J Neuroinflammation. 2016;13(1):130. doi:10.1186/s12974-016-0599-2.
  • Chubanov V, Mittermeier L, Gudermann T. Role of kinase-coupled trp channels in mineral homeostasis. Pharmacol Ther. 2018;184:159–176. doi:10.1016/j.pharmthera.2017.11.003.
  • Ryazanova LV, Hu Z, Suzuki S, Chubanov V, Fleig A, Ryazanov AG. Elucidating the role of the trpm7 alpha-kinase: trpm7 kinase inactivation leads to magnesium deprivation resistance phenotype in mice. Sci Rep. 2014;4(1):7599. doi:10.1038/srep07599.
  • Demeuse P, Penner R, Fleig A. Trpm7 channel is regulated by magnesium nucleotides via its kinase domain. J Gen Physiol. 2006;127(4):421–434. doi:10.1085/jgp.200509410.
  • Wong R, Turlova E, Feng ZP, Rutka JT, Sun H-S. Activation of trpm7 by naltriben enhances migration and invasion of glioblastoma cells. Oncotarget. 2017;8(7):11239–11248. doi:10.18632/oncotarget.14496.
  • Andersson KE. Trp channels as lower urinary tract sensory targets. Med Sci (Basel). 2019;7(5):67. doi:10.3390/medsci7050067.
  • Yin Y, Lee SY. Current view of ligand and lipid recognition by the menthol receptor trpm8. Trends Biochem Sci. 2020;45(9):806–819. doi:10.1016/j.tibs.2020.05.008.
  • Yin Y, Le SC, Hsu AL, Borgnia MJ, Yang H, Lee S-Y. Structural basis of cooling agent and lipid sensing by the cold-activated trpm8 channel. Science. 2019;363(6430). doi:10.1126/science.aav9334.
  • Duvoisin RM, Haley TL, Ren G, Strycharska-Orczyk I, Bonaparte JP, Morgans CW. Autoantibodies in melanoma-associated retinopathy recognize an epitope conserved between trpm1 and trpm3. Invest Ophthalmol Vis Sci. 2017;58(5):2732–2738. doi:10.1167/iovs.17-21443.
  • Maddodi N, Setaluri V. Prognostic significance of melanoma differentiation and trans-differentiation. Cancers Basel. 2010;2(2):989–999. doi:10.3390/cancers2020989.
  • Shiels A. TRPM3_MIR-204: a complex locus for eye development and disease. Hum Genomics. 2020;14(1):7. doi:10.1186/s40246-020-00258-4.
  • Sita G, Hrelia P, Graziosi A, Ravegnini G, Morroni F. Trpm2 in the brain: role in health and disease. Cells-Basel. 2018;7(7):82. doi:10.3390/cells7070082.
  • Xie YF, Belrose JC, Lei G, Tymianski M, Mori Y, MacDonald JF, Jackson MF. Dependence of NMDA/GSK-3β mediated metaplasticity on TRPM2 channels at Hippocampal CA3-CA1 synapses. Mol Brain. 2011;4(1):44. doi:10.1186/1756-6606-4-44.
  • Du J, Xie J, Yue L. Modulation of trpm2 by acidic PH and the underlying mechanisms for PH sensitivity. J Gen Physiol. 2009;134(6):471–488. doi:10.1085/jgp.200910254.
  • McQueen CM, Whitfield-Cargile CM, Konganti K, Blodgett GP, Dindot SV, Cohen ND. Trpm2 SNP genotype previously associated with susceptibility to rhodococcus equi pneumonia in quarter horse foals displays differential gene expression identified using RNA-seq. BMC Genomics. 2016;17(1):993. doi:10.1186/s12864-016-3345-3.
  • Kondratskyi A, Yassine M, Kondratska K, Skryma R, Slomianny C, Prevarskaya N. Calcium-permeable ion channels in control of autophagy and cancer. Front Physiol. 2013;4:272. doi:10.3389/fphys.2013.00272.
  • Brown RL, Xiong WH, Peters JH, Tekmen-Clark M, Strycharska-Orczyk I, Reed BT, Morgans CW, Duvoisin RM. Trpm3 expression in mouse retina. PLoS One. 2015;10(2):e117615. doi:10.1371/journal.pone.0117615.
  • Wagner TF, Drews A, Loch S, Mohr F, Philipp SE, Lambert S, Oberwinkler J. Trpm3 channels provide a regulated influx pathway for zinc in pancreatic beta cells. Pflugers Arch. 2010;460(4):755–765. doi:10.1007/s00424-010-0838-9.
  • Lee N, Chen J, Sun L, Wu S, Gray KR, Rich A, Huang M, Lin J-H, Feder JN, Janovitz EB, et al. Expression and characterization of human transient receptor potential melastatin 3 (htrpm3). J Biol Chem. 2003; 278(23):20890–20897. doi:10.1074/jbc.M211232200.
  • Vriens J, Voets T. Sensing the heat with trpm3. Pflugers Arch Eur J Physiol. 2018;470(5):799–807. doi:10.1007/s00424-017-2100-1.
  • Choudhary S, Kashyap SS, Martin RJ, Robertson AP. Advances in our understanding of nematode ion channels as potential anthelmintic targets. Int J Parasitol Drugs Drug Resist. 2022;18:52–86. doi:10.1016/j.ijpddr.2021.12.001.
  • Gonzales AL, Earley S. Regulation of cerebral artery smooth muscle membrane potential by Ca 2+-activated cation channels. Microcirculation. 2013;20(4):337–347. doi:10.1111/micc.12023.
  • Zhang H, Zhang X, Wang X, Sun H, Hou C, Yu Y, Wang S, Yin F, Yang Z. Comprehensive analysis of TRP channel–related genes in patients with triple-negative breast cancer for guiding prognostic prediction. Front Oncol. 2022;12:941283. doi:10.3389/fonc.2022.941283.
  • Kaske S, Krasteva G, Konig P, Kummer W, Hofmann T, Gudermann T, Chubanov V. Trpm5, a taste-signaling transient receptor potential ion-channel, is a ubiquitous signaling component in chemosensory cells. BMC Neurosci. 2007;8(1):49. doi:10.1186/1471-2202-8-49.
  • Dhakal S, Lee Y. Transient receptor potential channels and metabolism. Mol Cells. 2019;42(8):569–578. doi:10.14348/molcells.2019.0007.
  • Gaudet R. Divide and conquer: high resolution structural information on trp channel fragments. J Gen Physiol. 2009;133(3):231–7. doi:10.1085/jgp.200810137.
  • Chen WL, Turlova E, Sun CL, Kim J-S, Huang S, Zhong X, Guan Y-Y, Wang G-L, Rutka J, Feng Z-P, et al. Xyloketal b suppresses glioblastoma cell proliferation and migration in vitro through inhibiting trpm7-regulated pi3k/akt and mek/erk signaling pathways. Mar Drugs. 2015; 13(4):2505–25. doi:10.3390/md13042505.
  • Yee NS, Kazi AA, Yee RK. Cellular and developmental biology of trpm7 channel-kinase: implicated roles in cancer. Cells-Basel. 2014;3(3):751–77. doi:10.3390/cells3030751.
  • Ledeganck KJ, Boulet GA, Bogers JJ, Verpooten GA, De Winter BY. The trpm6/egf pathway is downregulated in a rat model of cisplatin nephrotoxicity. PLoS One. 2013;8(2):e57016. doi:10.1371/journal.pone.0057016.
  • Bidaux G, Borowiec AS, Dubois C, Delcourt P, Schulz C, Abeele FV, Lepage G, Desruelles E, Bokhobza A, Dewailly E, et al. Targeting of short trpm8 isoforms induces 4TM-TRPM8-dependent apoptosis in prostate cancer cells. Oncotarget. 2016; 7(20):29063–80. doi:10.18632/oncotarget.8666.
  • Peng M, Wang Z, Yang Z, Tao L, Liu Q, Yi Lu, Wang X. Overexpression of short TRPM8 variant α promotes cell migration and invasion, and decreases starvation-induced apoptosis in prostate cancer LNCaP cells. Oncol Lett. 2015;10(3):1378–84. doi:10.3892/ol.2015.3373.
  • Iraci N, Ostacolo C, Medina-Peris A, Ciaglia T, Novoselov AM, Altieri A, Cabañero D, Fernandez-Carvajal A, Campiglia P, Gomez-Monterrey I, et al. In vitro and in vivo pharmacological characterization of a novel trpm8 inhibitor chemotype identified by small-scale preclinical screening. Int J Mol Sci. 2022; 23(4):2070. doi:10.3390/ijms23042070.
  • Yudin Y, Rohacs T. Regulation of trpm8 channel activity. Mol Cell Endocrinol. 2012;353(1–2):68–74. doi:10.1016/j.mce.2011.10.023.
  • Ishii M, Oyama A, Hagiwara T, Miyazaki A, Mori Y, Kiuchi Y, Shimizu S. Facilitation of H2O2-INDUCED A172 human glioblastoma cell death by insertion of oxidative stress-sensitive trpm2 channels. Anticancer Res. 2007;27(6B):3987–92.
  • Zhao Q, Li J, Ko WH, Kwan Y-W, Jiang L, Sun L, Yao X. Trpm2 promotes autophagic degradation in vascular smooth muscle cells. Sci Rep. 2020;10(1):20719. doi:10.1038/s41598-020-77620-y.
  • Park L, Wang G, Moore J, Girouard H, Zhou P, Anrather J, Iadecola C. The key role of transient receptor potential melastatin-2 channels in amyloid-β-induced neurovascular dysfunction. Nat Commun. 2014;5(1):5318. doi:10.1038/ncomms6318.
  • Mahmuda NA, Yokoyama S, Munesue T, Hayashi K, Yagi K, Tsuji C, Higashida H. One single nucleotide polymorphism of the trpm2 channel gene identified as a risk factor in bipolar disorder associates with autism spectrum disorder in a Japanese population. Diseases. 2020;8(1):4. doi:10.3390/diseases8010004.
  • Bao MH, Lv QL, Szeto V, Wong R, Zhu S-Z, Zhang Y-Y, Feng Z-P, Sun H-S. Trpm2-as inhibits the growth, migration, and invasion of gliomas through JNK, C-JUN, and RGS4. J Cell Physiol. 2020;235(5):4594–604. doi:10.1002/jcp.29336.
  • Yamamoto S, Ishii T, Mikami R, Numata T, Shimizu S. Short trpm2 prevents the targeting of full-length trpm2 to the surface transmembrane by hijacking to ER associated degradation. Biochem Biophys Res Commun. 2019;520(3):520–5. doi:10.1016/j.bbrc.2019.10.065.
  • Cui D, Feng Y, Shi K, Zhang H, Qian R. Long non-coding RNA trpm2-as sponges microRNA-138-5p to activate epidermal growth factor receptor and PI3K/AKT signaling in non-small cell lung cancer. Ann Transl Med. 2020;8(20):1313. doi:10.21037/atm-20-6331.
  • Shi T, Li R, Duan P, Guan Y, Zhang D, Ding Z, Ruan X. Trpm2-as promotes paclitaxel resistance in prostate cancer by regulating FOXK1 via sponging mir-497-5p. Drug Dev Res. 2022;83(4):967–78. doi:10.1002/ddr.21924.
  • Xu C, Huang Q, Zhang C, Xu W, Xu G, Zhao X, Liu X, Du Y. Long non-coding RNA trpm2-as as a potential biomarker for hepatocellular carcinoma. Ir J Med Sci. 2018;187(3):621–8. doi:10.1007/s11845-017-1692-y.
  • Guo J, Shan C, Xu J, Li M, Zhao J, Cheng W. New insights into trp ion channels in stem cells. Int J Mol Sci. 2022;23(14):7766. doi:10.3390/ijms23147766.
  • Fallah HP, Ahuja E, Lin H, Qi J, He Q, Gao S, An H, Zhang J, Xie Y, Liang D, et al. A review on the role of trp channels and their potential as drug targets_an insight into the trp channel drug discovery methodologies. Front Pharmacol. 2022;13:914499. doi:10.3389/fphar.2022.914499.
  • Chen X, Numata T, Li M, Mori Y, Orser BA, Jackson MF, Xiong Z-G, MacDonald JF. The modulation of trpm7 currents by nafamostat mesilate depends directly upon extracellular concentrations of divalent cations. Mol Brain. 2010;3(1):38. doi:10.1186/1756-6606-3-38.
  • Liu M, Inoue K, Leng T, Guo S, Xiong Z-G. Trpm7 channels regulate glioma stem cell through STAT3 and Notch signaling pathways. Cell Signalling. 2014;26(12):2773–81. doi:10.1016/j.cellsig.2014.08.020.
  • Thuringer D, Chanteloup G, Winckler P, Garrido C. The vesicular transfer of CLIC1 from glioblastoma to microvascular endothelial cells requires trpm7. Oncotarget. 2018;9(70):33302–11. doi:10.18632/oncotarget.26048.
  • Zeitlmayr S, Zierler S, Staab-Weijnitz CA, Dietrich A, Geiger F, Horgen FD, Gudermann T, Breit A. TRPM7 restrains plasmin activity and promotes transforming growth factor-β1 signaling in primary human lung fibroblasts. Arch Toxicol. 2022;96(10):2767–83. doi:10.1007/s00204-022-03342-x.
  • Zalles M, Smith N, Saunders D, Lerner M, Fung K-M, Battiste J, Towner RA. A tale of two multi-focal therapies for glioblastoma: an antibody targeting ELTD1 and nitrone-based okn-007. J Cell Mol Med. 2022;26(2):570–82. doi:10.1111/jcmm.17133.
  • Klumpp D, Frank SC, Klumpp L, Sezgin EC, Eckert M, Edalat L, Bastmeyer M, Zips D, Ruth P, Huber SM, et al. Trpm8 is required for survival and radioresistance of glioblastoma cells. Oncotarget. 2017; 8(56):95896–913. doi:10.18632/oncotarget.21436.
  • Grolez GP, Gordiendko DV, Clarisse M, Hammadi M, Desruelles E, Fromont G, Prevarskaya N, Slomianny C, Gkika D. Trpm8-androgen receptor association within lipid rafts promotes prostate cancer cell migration. Cell Death Dis. 2019;10(9):652. doi:10.1038/s41419-019-1891-8.
  • Laterra J, Rosen E, Nam M, Ranganathan S, Fielding K, Johnston P. Scatter factor/hepatocyte growth factor expression enhances human glioblastoma tumorigenicity and growth. Biochem Biophys Res Commun. 1997;235(3):743–7. doi:10.1006/bbrc.1997.6853.
  • Wang X, Omar O, Vazirisani F, Thomsen P, Ekström K. Mesenchymal stem cell-derived exosomes have altered microRNA profiles and induce osteogenic differentiation depending on the stage of differentiation. PLoS One. 2018;13(2):e193059. doi:10.1371/journal.pone.0193059.
  • Li T, Pan H, Li R. The dual regulatory role of mir-204 in cancer. Tumour Biol. 2016;37(9):11667–77. doi:10.1007/s13277-016-5144-5.
  • Son JC, Jeong HO, Park D, No SG, Lee EK, Lee J, Chung HY. mir-10a and mir-204 as a potential prognostic indicator in low-grade gliomas. Cancer Inform. 2017;16:1885518114. doi:10.1177/1176935117702878.
  • Fan X, Zou X, Liu C, Cheng W, Zhang S, Geng X, Zhu W. microRNA expression profile in serum reveals novel diagnostic biomarkers for endometrial cancer. Biosci Rep. 2021;41(6):41. doi:10.1042/BSR20210111.
  • Xin J, Zheng LM, Sun DK, Li X, Xu P, Tian L. miR‑204 functions as a tumor suppressor gene, at least partly by suppressing CYP27A1 in glioblastoma. Oncol Lett. 2018;16:1439–48. doi:10.3892/ol.2018.8846.
  • Li L, Zhao GD, Shi Z, Qi L-L, Zhou L-Y, Fu Z-X. The RAS/RAF/MEK/ERK signaling pathway and its role in the occurrence and development of HCC. Oncol Lett. 2016;12(5):3045–3050. doi:10.3892/ol.2016.5110.
  • Wang SH, Wu HC, Badrealam KF, Kuo Y-H, Chao Y-P, Hsu H-H, Bau D-T, Viswanadha VP, Chen Y-H, Lio P-J, et al. Taiwanin E induces cell cycle arrest and apoptosis in Arecoline/4-NQO-induced oral cancer cells through modulation of the ERK signaling pathway. Front Oncol. 2019;9:1309. doi:10.3389/fonc.2019.01309.
  • Zou ZG, Rios FJ, Montezano AC, Touyz RM. Trpm7, magnesium, and signaling. Int J Mol Sci. 2019;20(8):1877. doi:10.3390/ijms20081877.
  • Lee EH, Chun SY, Kim B, Yoon BH, Lee JN, Kim BS, Yoo ES, Lee S, Song PH, Kwon TG, et al. Knockdown of trpm7 prevents tumor growth, migration, and invasion through the Src, Akt, and Jnk pathway in bladder cancer. BMC Urol. 2020; 20(1):145. doi:10.1186/s12894-020-00714-2.
  • Takayasu T, Kurisu K, Esquenazi Y, Ballester LY. Ion channels and their role in the pathophysiology of gliomas. Mol Cancer Ther. 2020;19(10):1959–1969. doi:10.1158/1535-7163.MCT-19-0929.
  • William D, Mokri P, Lamp N, Linnebacher M, Classen CF, Erbersdobler A, Schneider B. Amplification of the egfr gene can be maintained and modulated by variation of egf concentrations in in vitro models of glioblastoma multiforme. PLoS One. 2017;12(9):e185208. doi:10.1371/journal.pone.0185208.
  • Zhang Q, Shen J, Wu Y, Ruan W, Zhu F, Duan S. LINC00520: a potential diagnostic and prognostic biomarker in cancer. Front Immunol. 2022;13:845418. doi:10.3389/fimmu.2022.845418.
  • Zhang Y, Zhuang K, Yuan S, Si W, Li Y, Zhang J, Liu J. Effects of mir-195 targeted regulation of JAK2 on proliferation, invasion, and apoptosis of gastric cancer cells. Comput Math Methods Med. 2022;2022:1–8. doi:10.1155/2022/5873479.
  • Swiatek-Machado K, Kaminska B. Stat signaling in glioma cells. Adv Exp Med Biol. 2020;1202:203–222. doi:10.1007/978-3-030-30651-9_10.
  • Ciaglia T, Vestuto V, Bertamino A, González-Muñiz R, Gómez-Monterrey I. On the modulation of trpm channels: current perspectives and anticancer therapeutic implications. Front Oncol. 2022;12:1065935. doi:10.3389/fonc.2022.1065935.
  • Su Z, Han S, Jin Q, Zhou N, Lu J, Shangguan F, Yu S, Liu Y, Wang L, Lu J, et al. Ciclopirox and bortezomib synergistically inhibits glioblastoma multiforme growth via simultaneously enhancing JNK/p38 MAPK and NF-κB signaling. Cell Death Dis. 2021; 12(3):251. doi:10.1038/s41419-021-03535-9.
  • Yuan Z, Yang Z, Li W, Wu A, Su Z, Jiang B, Ganesan S. Triphlorethol-a attenuates u251 human glioma cancer cell proliferation and ameliorates apoptosis through Jak2/Stat3 and P38 Mapk/Erk signaling pathways. J Biochem Mol Toxicol. 2022;36(9):e23138. doi:10.1002/jbt.23138.
  • Maklad A, Sharma A, Azimi I. Calcium signaling in brain cancers: roles and therapeutic targeting. Cancers Basel. 2019;11(2):11. doi:10.3390/cancers11020145.
  • Higashimori H, Sontheimer H. Role of kir4.1 channels in growth control of glia. Glia. 2007;55(16):1668–1679. doi:10.1002/glia.20574.
  • Seker-Polat F, Pinarbasi DN, Solaroglu I, Bagci-Onder T. Tumor cell infiltration into the brain in glioblastoma: from mechanisms to clinical perspectives. Cancers Basel. 2022;14(2):14. doi:10.3390/cancers14020443.
  • Akpinar O, Ozsimsek A, Guzel M, Nazıroğlu M. Clostridium botulinum neurotoxin a induces apoptosis and mitochondrial oxidative stress via activation of trpm2 channel signaling pathway in neuroblastoma and glioblastoma tumor cells. J Recept Signal Transduct Res. 2020;40(6):620–632. doi:10.1080/10799893.2020.1781174.
  • Lee JE, Yoon SS, Moon EY. Curcumin-induced autophagy augments its antitumor effect against a172 human glioblastoma cells. Biomol & Therapeutics. 2019;27(5):484–491. doi:10.4062/biomolther.2019.107.
  • Gokce KS, Gokce G, Kutuk M, Gürses Cila HE, Nazıroğlu M. Curcumin enhances cisplatin-induced human laryngeal squamous cancer cell death through activation of trpm2 channel and mitochondrial oxidative stress. Sci Rep. 2019;9(1):17784. doi:10.1038/s41598-019-54284-x.
  • Sprouse AA, Herbert BS. Resveratrol augments paclitaxel treatment in MDA-MB-231 and paclitaxel-resistant Mda-Mb-231 breast cancer cells. Anticancer Res. 2014;34(10):5363–5374.
  • Ozturk Y, Gunaydin C, Yalcin F, Nazıroğlu M, Braidy N. Resveratrol enhances apoptotic and oxidant effects of paclitaxel through trpm2 channel activation in dbtrg glioblastoma cells. Oxid Med Cell Longevity. 2019;2019:1–13. doi:10.1155/2019/4619865.
  • Li T, Wu K, Yue Z, Wang Y, Zhang F, Shen H. Structural basis for the modulation of human KCNQ4 by small-molecule drugs. Mol Cell. 2021;81(1):25–37. doi:10.1016/j.molcel.2020.10.037.
  • Nabissi M, Morelli MB, Santoni M, Santoni G. Triggering of the trpv2 channel by cannabidiol sensitizes glioblastoma cells to cytotoxic chemotherapeutic agents. Carcinogenesis. 2013;34(1):48–57. doi:10.1093/carcin/bgs328.
  • Nikolova B, Antov G, Semkova S, Tsoneva I, Christova N, Nacheva L, Kardaleva P, Angelova S, Stoineva I, Ivanova J, et al. Bacterial natural disaccharide (Trehalose Tetraester): molecular modeling and in vitro study of anticancer activity on breast cancer cells. Polym (Basel). 2020; 12(2):12. doi:10.3390/polym12020499.
  • Chen J, Dou Y, Zheng X, Leng T, Lu X, Ouyang Y, Sun H, Xing F, Mai J, Gu J, et al. Trpm7 channel inhibition mediates midazolam-induced proliferation loss in human malignant glioma. Tumour Biol. 2016; 37(11):14721–14731. doi:10.1007/s13277-016-5317-2.
  • Sander P, Mostafa H, Soboh A, Schneider JM, Pala A, Baron A-K, Moepps B, Wirtz CR, Georgieff M, Schneider M, et al. Vacquinol-1 inducible cell death in glioblastoma multiforme is counter regulated by trpm7 activity induced by exogenous ATP. Oncotarget. 2017; 8(21):35124–35137. doi:10.18632/oncotarget.16703.
  • Voringer S, Schreyer L, Nadolni W, Meier MA, Woerther K, Mittermeier C, Ferioli S, Singer S, Holzer K, Zierler S, et al. Inhibition of TRPM7 blocks MRTF/SRF-dependent transcriptional and tumorigenic activity. Oncogene. 2020; 39(11):2328–2344. doi:10.1038/s41388-019-1140-8.
  • McFerrin MB, Sontheimer H. A role for ion channels in glioma cell invasion. Neuron Glia Biol. 2006;2(1):39–49. doi:10.1017/S17440925X06000044.
  • Ransom CB, Sontheimer H. BK channels in human glioma cells. J Neurophysiol. 2001;85(2):790–803. doi:10.1152/jn.2001.85.2.790.
  • Ratto D, Ferrari B, Roda E, Brandalise F, Siciliani S, De Luca F, Priori EC, Di Iorio C, Cobelli F, Veneroni P, et al. Squaring the circle: a new study of inward and outward-rectifying potassium currents in U251 GBM cells. Cell Mol Neurobiol. 2020; 40(5):813–828. doi:10.1007/s10571-019-00776-3.