Generic placeholder image

Current Neurovascular Research

Editor-in-Chief

ISSN (Print): 1567-2026
ISSN (Online): 1875-5739

Research Article

Pharmacological Inhibition of Transient Receptor Potential Melastatin 2 (TRPM2) Channels Attenuates Diabetes-induced Cognitive Deficits in Rats: A Mechanistic Study

Author(s): Pavan Thapak, Mahendra Bishnoi and Shyam S. Sharma*

Volume 17, Issue 3, 2020

Page: [249 - 258] Pages: 10

DOI: 10.2174/1567202617666200415142211

Price: $65

Abstract

Background: Diabetes is a chronic metabolic disorder affecting the central nervous system. A growing body of evidence has depicted that high glucose level leads to the activation of the transient receptor potential melastatin 2 (TRPM2) channels. However, there are no studies targeting TRPM2 channels in diabetes-induced cognitive decline using a pharmacological approach.

Objective: The present study intended to investigate the effects of 2-aminoethoxydiphenyl borate (2-APB), a TRPM2 inhibitor, in diabetes-induced cognitive impairment.

Methods: Streptozotocin (STZ, 50 mg/kg, i.p.) was used to induce diabetes in rats. Animals were randomly divided into the treatment group, model group and age-matched control and pre se group. 2-APB treatment was given for three weeks to the animals. After 10 days of behavioural treatment, parameters were performed. Animals were sacrificed at 10th week of diabetic induction and the hippocampus and cortex were isolated. After that, protein and mRNA expression study was performed in the hippocampus. Acetylcholinesterase (AchE) activity was done in the cortex.

Results: Our study showed the 10th week diabetic animals developed cognitive impairment, which was evident from the behavioural parameters. Diabetic animals depicted an increase in the TRPM2 mRNA and protein expression in the hippocampus as well as increased AchE activity in the cortex. However, memory associated proteins were down-regulated, namely Ca2+/calmodulin-dependent protein kinase II (CaMKII-Thr286), glycogen synthase kinase 3 beta (GSK-3β-Ser9), cAMP response element-binding protein (CREB-Ser133), and postsynaptic density protein 95 (PSD-95). Gene expression of parvalbumin, calsequestrin and brain-derived neurotrophic factor (BDNF) were down-regulated while mRNA level of calcineurin A/ protein phosphatase 3 catalytic subunit alpha (PPP3CA) was upregulated in the hippocampus of diabetic animals. A three-week treatment with 2-APB significantly ameliorated the alteration in behavioural cognitive parameters in diabetic rats. Moreover, 2-APB also down-regulated the expression of TRPM2 mRNA and protein in the hippocampus as well as AchE activity in the cortex of diabetic animals as compared to diabetic animals. Moreover, the 2-APB treatment also upregulated the CaMKII (Thr-286), GSK-3β (Ser9), CREB (Ser133), and PSD-95 expression and mRNA levels of parvalbumin, calsequestrin, and BDNF while mRNA level of calcineurin A was down-regulated in the hippocampus of diabetic animals.

Conclusion: This study confirms the ameliorative effect of TRPM2 channel inhibitor in the diabetes- induced cognitive deficits. Inhibition of TRPM2 channels reduced the calcium associated downstream signaling and showed a neuroprotective effect of TRPM2 channels in diabetesinduced cognitive impairment.

Keywords: TRPM2, metabolic disorder, behavioural treatment, neuroprotective effect, diabetes, neuropathy.

[1]
Sima AA. Encephalopathies: The emerging diabetic complications. Acta Diabetol 2010; 47(4): 279-93.
[http://dx.doi.org/10.1007/s00592-010-0218-0] [PMID: 20798963]
[2]
Chen R, Shi J, Yin Q, et al. Morphological and pathological characteristics of brain in diabetic encephalopathy. J Alzheimers Dis 2018; 65(1): 15-28.
[http://dx.doi.org/10.3233/JAD-180314] [PMID: 30040723]
[3]
Gispen WH, Biessels GJ. Cognition and synaptic plasticity in diabetes mellitus. Trends Neurosci 2000; 23(11): 542-9.
[http://dx.doi.org/10.1016/S0166-2236(00)01656-8] [PMID: 11074263]
[4]
Tomlinson DR, Gardiner NJ. Glucose neurotoxicity. Nat Rev Neurosci 2008; 9(1): 36-45.
[http://dx.doi.org/10.1038/nrn2294] [PMID: 18094705]
[5]
Thibault O, Anderson KL, DeMoll C, Brewer LD, Landfield PW, Porter NM. Hippocampal calcium dysregulation at the nexus of diabetes and brain aging. Eur J Pharmacol 2013; 719(1-3): 34-43.
[http://dx.doi.org/10.1016/j.ejphar.2013.07.024] [PMID: 23872402]
[6]
Kumar A. Long-term potentiation at CA3-CA1 hippocampal synapses with special emphasis on aging, disease, and stress. Front Aging Neurosci 2011; 3: 7.
[http://dx.doi.org/10.3389/fnagi.2011.00007] [PMID: 21647396]
[7]
Rababa’h AM, Alzoubi KH, Baydoun S, Khabour OF. Levosimendan prevents memory impairment induced by diabetes in rats: Role of oxidative stress. Curr Alzheimer Res 2019; 16(14): 1300-8.
[http://dx.doi.org/10.2174/1567205017666200102153239] [PMID: 31894746]
[8]
Fonfria E, Mattei C, Hill K, et al. TRPM2 is elevated in the tMCAO stroke model, transcriptionally regulated, and functionally expressed in C13 microglia. J Recept Signal Transduct Res 2006; 26(3): 179-98.
[http://dx.doi.org/10.1080/10799890600637522] [PMID: 16777714]
[9]
Adhya P, Sharma SS. Redox TRPs in diabetes and diabetic complications: Mechanisms and pharmacological modulation. Pharmacol Res 2019; 2019: 146104271
[http://dx.doi.org/10.1016/j.phrs.2019.104271] [PMID: 31096011]
[10]
Sita G, Hrelia P, Graziosi A, Ravegnini G, Morroni F. TRPM2 in the brain: Role in health and disease. Cells 2018; 7(7): E82
[http://dx.doi.org/10.3390/cells7070082] [PMID: 30037128]
[11]
Turlova E, Feng Z-P, Sun H-S. 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.
[http://dx.doi.org/10.1038/aps.2017.194] [PMID: 29542681]
[12]
Fonfria E, Marshall IC, Boyfield I, et al. Amyloid beta-peptide(1-42) and hydrogen peroxide-induced toxicity are mediated by TRPM2 in rat primary striatal cultures. J Neurochem 2005; 95(3): 715-23.
[http://dx.doi.org/10.1111/j.1471-4159.2005.03396.x] [PMID: 16104849]
[13]
Chung KK, Freestone PS, Lipski J. Expression and functional properties of TRPM2 channels in dopaminergic neurons of the substantia nigra of the rat. J Neurophysiol 2011; 106(6): 2865-75.
[http://dx.doi.org/10.1152/jn.00994.2010] [PMID: 21900507]
[14]
Kahya MC, Nazıroğlu M, Övey IS. Modulation of diabetes-induced oxidative stress, apoptosis, and Ca2+ entry through TRPM2 and TRPV1 channels in dorsal root ganglion and hippocampus of diabetic rats by melatonin and selenium. Mol Neurobiol 2017; 54(3): 2345-60.
[http://dx.doi.org/10.1007/s12035-016-9727-3] [PMID: 26957303]
[15]
Markó L, Mannaa M, Haschler TN, Krämer S, Gollasch M. Renoprotection: focus on TRPV1, TRPV4, TRPC6 and TRPM2. Acta Physiol (Oxf) 2017; 219(3): 589-612.
[http://dx.doi.org/10.1111/apha.12828] [PMID: 28028935]
[16]
Tseng HH, Vong CT, Kwan YW, Lee SM, Hoi MP. TRPM2 regulates TXNIP-mediated NLRP3 inflammasome activation via interaction with p47 phox under high glucose in human monocytic cells. Sci Rep 2016; 6: 35016.
[http://dx.doi.org/10.1038/srep35016] [PMID: 27731349]
[17]
Negi G, Kumar A, Sharma SS. Melatonin modulates neuroinflammation and oxidative stress in experimental diabetic neuropathy: effects on NF-κB and Nrf2 cascades. J Pineal Res 2011; 50(2): 124-31.
[PMID: 21062351]
[18]
Togashi K, Inada H, Tominaga M. Inhibition of the transient receptor potential cation channel TRPM2 by 2-aminoethoxydiphenyl borate (2-APB). Br J Pharmacol 2008; 153(6): 1324-30.
[http://dx.doi.org/10.1038/sj.bjp.0707675] [PMID: 18204483]
[19]
Lu Y, Wu X, Dong Y, Xu Z, Zhang Y, Xie Z. Anesthetic sevoflurane causes neurotoxicity differently in neonatal naïve and Alzheimer disease transgenic mice. Anesthesiology 2010; 112(6): 1404-16.
[http://dx.doi.org/10.1097/ALN.0b013e3181d94de1] [PMID: 20460993]
[20]
Oka J, Suzuki E, Kondo Y. Endogenous GLP-1 is involved in β-amyloid protein-induced memory impairment and hippocampal neuronal death in rats. Brain Res 2000; 878(1-2): 194-8.
[http://dx.doi.org/10.1016/S0006-8993(00)02741-4] [PMID: 10996151]
[21]
Datusalia AK, Sharma SS. NF-κB Inhibition resolves cognitive deficits in experimental Type 2 diabetes mellitus through CREB and Glutamate/GABA neurotransmitters pathway. Curr Neurovasc Res 2016; 13(1): 22-32.
[http://dx.doi.org/10.2174/1567202612666151030104810] [PMID: 26517200]
[22]
Kumar P, Kaundal RK, More S, Sharma SS. Beneficial effects of pioglitazone on cognitive impairment in MPTP model of Parkinson’s disease. Behav Brain Res 2009; 197(2): 398-403.
[http://dx.doi.org/10.1016/j.bbr.2008.10.010] [PMID: 18983875]
[23]
Datusalia AK, Sharma SS. Amelioration of diabetes-induced cognitive deficits by GSK-3β inhibition is attributed to modulation of neurotransmitters and neuroinflammation. Mol Neurobiol 2014; 50(2): 390-405.
[http://dx.doi.org/10.1007/s12035-014-8632-x] [PMID: 24420785]
[24]
Ellman GL, Courtney KD, Andres V Jr, Feather-Stone RM. A new and rapid colorimetric determination of acetylcholinesterase activity. Biochem Pharmacol 1961; 7(2): 88-95.
[http://dx.doi.org/10.1016/0006-2952(61)90145-9] [PMID: 13726518]
[25]
Khare P, Mangal P, Baboota RK, et al. Involvement of glucagon in preventive effect of menthol against high fat diet induced obesity in mice. Front Pharmacol 2018; 9: 1244.
[http://dx.doi.org/10.3389/fphar.2018.01244] [PMID: 30505271]
[26]
Resham K, Sharma SS. Pharmacologic inhibition of porcupine, disheveled, and β-Catenin in Wnt signaling pathway ameliorates diabetic peripheral neuropathy in rats. J Pain 2019; 20(11): 1338-52.
[http://dx.doi.org/10.1016/j.jpain.2019.04.010] [PMID: 31075529]
[27]
Bulani Y, Srinivasan K, Sharma SS. Attenuation of type-1 diabetes-induced cardiovascular dysfunctions by direct thrombin inhibitor in rats: A mechanistic study. Mol Cell Biochem 2019; 451(1-2): 69-78.
[http://dx.doi.org/10.1007/s11010-018-3394-9] [PMID: 29971544]
[28]
Kuhad A, Chopra K. Curcumin attenuates diabetic encephalopathy in rats: behavioral and biochemical evidences. Eur J Pharmacol 2007; 576(1-3): 34-42.
[http://dx.doi.org/10.1016/j.ejphar.2007.08.001] [PMID: 17822693]
[29]
Iwai T, Suzuki M, Kobayashi K, Mori K, Mogi Y, Oka J. The influences of juvenile diabetes on memory and hippocampal plasticity in rats: Improving effects of glucagon-like peptide-1. Neurosci Res 2009; 64(1): 67-74.
[http://dx.doi.org/10.1016/j.neures.2009.01.013] [PMID: 19321133]
[30]
Esmaeili MH, Enayati M, Khabbaz Abkenar F, Ebrahimian F, Salari AA. Glibenclamide mitigates cognitive impairment and hippocampal neuroinflammation in rats with type 2 diabetes and sporadic Alzheimer-like disease. Behav Brain Res 2020.: 379112359
[http://dx.doi.org/10.1016/j.bbr.2019.112359] [PMID: 31733313]
[31]
Hardigan T, Ward R, Ergul A. Cerebrovascular complications of diabetes: focus on cognitive dysfunction. Clin Sci (Lond) 2016; 130(20): 1807-22.
[http://dx.doi.org/10.1042/CS20160397] [PMID: 27634842]
[32]
Omidi G, Karimi SA, Shahidi S, Faraji N, Komaki A. Coenzyme Q10 supplementation reverses diabetes-related impairments in long-term potentiation induction in hippocampal dentate gyrus granular cells: An in vivo study. Brain Res 2020; •••: 1726146475
[http://dx.doi.org/10.1016/j.brainres.2019.146475] [PMID: 31560865]
[33]
Shimizu T, Dietz RM, Cruz-Torres I, et al. Extended therapeutic window of a novel peptide inhibitor of TRPM2 channels following focal cerebral ischemia. Exp Neurol 2016; 283(Pt A): 151-6.
[http://dx.doi.org/10.1016/j.expneurol.2016.06.015]
[34]
Sun Y, Sukumaran P, Selvaraj S, et al. TRPM2 promotes neurotoxin MPP+/MPTP-induced cell death. Mol Neurobiol 2018; 55(1): 409-20.
[http://dx.doi.org/10.1007/s12035-016-0338-9] [PMID: 27957685]
[35]
Kaneko S, Kawakami S, Hara Y, et al. A critical role of TRPM2 in neuronal cell death by hydrogen peroxide. J Pharmacol Sci 2006; 101(1): 66-76.
[http://dx.doi.org/10.1254/jphs.FP0060128] [PMID: 16651700]
[36]
An X, Fu Z, Mai C, et al. Increasing the TRPM2 channel expression in human neuroblastoma SH-SY5Y cells augments the susceptibility to ROS-induced cell death. Cells 2019; 8(1): E28
[http://dx.doi.org/10.3390/cells8010028] [PMID: 30625984]
[37]
Wang S, Kobayashi K, Kogure Y, et al. Negative regulation of TRPA1 by AMPK in primary sensory neurons as a potential mechanism of painful diabetic neuropathy. Diabetes 2018; 67(1): 98-109.
[http://dx.doi.org/10.2337/db17-0503] [PMID: 29025860]
[38]
Liao MH, Xiang YC, Huang JY, et al. The disturbance of hippocampal CaMKII/PKA/PKC phosphorylation in early experimental diabetes mellitus. CNS Neurosci Ther 2013; 19(5): 329-36.
[http://dx.doi.org/10.1111/cns.12084] [PMID: 23490331]
[39]
Bhardwaj SK, Kaur G. Effect of diabetes on calcium/calmodulin dependent protein kinase-II from rat brain. Neurochem Int 1999; 35(4): 329-35.
[http://dx.doi.org/10.1016/S0197-0186(99)00066-2] [PMID: 10482353]
[40]
Wang Z, Ge Q, Wu Y, Zhang J, Gu Q, Han J. Impairment of long-term memory by a short-term high-fat diet via hippocampal oxidative stress and alterations in synaptic plasticity. Neuroscience 2020; 424: 24-33.
[http://dx.doi.org/10.1016/j.neuroscience.2019.10.050] [PMID: 31711814]
[41]
Wang J, Gong B, Zhao W, et al. Epigenetic mechanisms linking diabetes and synaptic impairments. Diabetes 2014; 63(2): 645-54.
[http://dx.doi.org/10.2337/db13-1063] [PMID: 24154559]
[42]
Hu T, Lu XY, Shi JJ, et al. Quercetin protects against diabetic encephalopathy via SIRT1/NLRP3 pathway in db/db mice. J Cell Mol Med 2020; 24(6): 3449-59.
[http://dx.doi.org/10.1111/jcmm.15026] [PMID: 32000299]
[43]
Chen X, Levy JM, Hou A, et al. PSD-95 family MAGUKs are essential for anchoring AMPA and NMDA receptor complexes at the postsynaptic density. Proc Natl Acad Sci USA 2015; 112(50): E6983-92.
[http://dx.doi.org/10.1073/pnas.1517045112] [PMID: 26604311]
[44]
Bartolotti N, Bennett DA, Lazarov O. Reduced pCREB in Alzheimer’s disease prefrontal cortex is reflected in peripheral blood mononuclear cells. Mol Psychiatry 2016; 21(9): 1158-66.
[http://dx.doi.org/10.1038/mp.2016.111]
[45]
Miao Y, He T, Zhu Y, Li W, Wang B, Zhong Y. Activation of hippocampal CREB by rolipram partially recovers balance between TNF-α and IL-10 levels and improves cognitive deficits in diabetic rats. Cell Mol Neurobiol 2015; 35(8): 1157-64.
[http://dx.doi.org/10.1007/s10571-015-0209-3] [PMID: 26001770]
[46]
Khandkar MA, Mukherjee E, Parmar DV, Katyare SS. Alloxan-diabetes alters kinetic properties of the membrane-bound form, but not of the soluble form, of acetylcholinesterase in rat brain. Biochem J 1995; 307(3): 647-9.
[http://dx.doi.org/10.1042/bj3070647]
[47]
Suhail M, Rizvi SI. Erythrocyte membrane acetylcholinesterase in type 1 (insulin-dependent) diabetes mellitus. Biochem J 1989; 259(3): 897-9.
[http://dx.doi.org/10.1042/bj2590897] [PMID: 2658981]

Rights & Permissions Print Cite
© 2024 Bentham Science Publishers | Privacy Policy