Generic placeholder image

Current Alzheimer Research

Editor-in-Chief

ISSN (Print): 1567-2050
ISSN (Online): 1875-5828

Review Article

GIRK2 Channels in Down Syndrome and Alzheimer’s Disease

Author(s): Alexander Kleschevnikov*

Volume 19, Issue 12, 2022

Published on: 05 January, 2022

Page: [819 - 829] Pages: 11

DOI: 10.2174/1567205020666221223122110

Price: $65

Abstract

Cognitive impairment in Down syndrome (DS) results from the abnormal expression of hundreds of genes. However, the impact of KCNJ6, a gene located in the middle of the ‘Down syndrome critical region’ of chromosome 21, seems to stand out. KCNJ6 encodes GIRK2 (KIR3.2) subunits of G protein-gated inwardly rectifying potassium channels, which serve as effectors for GABAB, m2, 5HT1A, A1, and many other postsynaptic metabotropic receptors. GIRK2 subunits are heavily expressed in neocortex, cerebellum, and hippocampus. By controlling resting membrane potential and neuronal excitability, GIRK2 channels may thus affect both synaptic plasticity and stability of neural circuits in the brain regions important for learning and memory. Here, we discuss recent experimental data regarding the role of KCNJ6/GIRK2 in neuronal abnormalities and cognitive impairment in models of DS and Alzheimer’s disease (AD). The results compellingly show that signaling through GIRK2 channels is abnormally enhanced in mouse genetic models of Down syndrome and that partial suppression of GIRK2 channels with pharmacological or genetic means can restore synaptic plasticity and improve impaired cognitive functions. On the other hand, signaling through GIRK2 channels is downregulated in AD models, such as models of early amyloidopathy. In these models, reduced GIRK2 channel signaling promotes neuronal hyperactivity, causing excitatory-inhibitory imbalance and neuronal death. Accordingly, activation of GABAB/GIRK2 signaling by GIRK channel activators or GABAB receptor agonists may reduce Aβ-induced hyperactivity and subsequent neuronal death, thereby exerting a neuroprotective effect in models of AD.

Keywords: Down syndrome, Alzheimer’s disease, GIRK2 channels, genetic disorders, neuronal hyperactivity, KCNJ6.

[1]
Rafii MS, Kleschevnikov AM, Sawa M, Mobley WC. Down syndrome. Handb Clin Neurol 2019; 167: 321-36.
[http://dx.doi.org/10.1016/B978-0-12-804766-8.00017-0] [PMID: 31753140]
[2]
Dierssen M. Down syndrome: The brain in trisomic mode. Nat Rev Neurosci 2012; 13(12): 844-58.
[http://dx.doi.org/10.1038/nrn3314] [PMID: 23165261]
[3]
Lott IT, Head E. Dementia in Down syndrome: Unique insights for Alzheimer disease research. Nat Rev Neurol 2019; 15(3): 135-47.
[http://dx.doi.org/10.1038/s41582-018-0132-6] [PMID: 30733618]
[4]
Antonarakis SE, Skotko BG, Rafii MS, et al. Down syndrome. Nat Rev Dis Primers 2020; 6(1): 9.
[http://dx.doi.org/10.1038/s41572-019-0143-7] [PMID: 32029743]
[5]
Sturgeon X, Le T, Ahmed MM, Gardiner KJ. Pathways to cognitive deficits in Down syndrome. Prog Brain Res 2012; 197: 73-100.
[http://dx.doi.org/10.1016/B978-0-444-54299-1.00005-4] [PMID: 22541289]
[6]
Letourneau A, Antonarakis SE. Genomic determinants in the phenotypic variability of Down syndrome. Prog Brain Res 2012; 197: 15-28.
[http://dx.doi.org/10.1016/B978-0-444-54299-1.00002-9] [PMID: 22541286]
[7]
Antonarakis SE. Down syndrome and the complexity of genome dosage imbalance. Nat Rev Genet 2017; 18(3): 147-63.
[http://dx.doi.org/10.1038/nrg.2016.154] [PMID: 28029161]
[8]
Korenberg JR, Chen XN, Schipper R, et al. Down syndrome phenotypes: The consequences of chromosomal imbalance. Proc Natl Acad Sci USA 1994; 91(11): 4997-5001.
[http://dx.doi.org/10.1073/pnas.91.11.4997] [PMID: 8197171]
[9]
Delabar JM, Theophile D, Rahmani Z, et al. Molecular mapping of twenty-four features of Down syndrome on chromosome 21. Eur J Hum Genet 1993; 1(2): 114-24.
[http://dx.doi.org/10.1159/000472398] [PMID: 8055322]
[10]
Rahmani Z, Blouin JL, Créau-Goldberg N, et al. Down syndrome critical region around D21S55 on proximal 21q22.3. Am J Med Genet 1990; 37(S7): 98-103.
[http://dx.doi.org/10.1002/ajmg.1320370720] [PMID: 2149984]
[11]
Pelleri MC, Cicchini E, Locatelli C, et al. Systematic reanalysis of partial trisomy 21 cases with or without Down syndrome suggests a small region on 21q22.13 as critical to the phenotype. Hum Mol Genet 2016; 25(12): ddw116.
[http://dx.doi.org/10.1093/hmg/ddw116] [PMID: 27106104]
[12]
Pelleri MC, Cicchini E, Petersen MB, et al. Partial trisomy 21 map: Ten cases further supporting the highly restricted Down syndrome critical region (HR‐DSCR) on human chromosome 21. Mol Genet Genomic Med 2019; 7(8): e797.
[http://dx.doi.org/10.1002/mgg3.797] [PMID: 31237416]
[13]
Antonaros F, Pitocco M, Abete D, et al. Structural Characterization of the Highly Restricted Down Syndrome Critical Region on 21q22.13: New KCNJ6 and DSCR4 Transcript Isoforms. Front Genet 2021; 12: 770359.
[http://dx.doi.org/10.3389/fgene.2021.770359] [PMID: 34956324]
[14]
Asai S, Yamaki A, Kudoh J, Shimizu N, Shimizu Y. Analysis of the promoter region of human placenta-specific DSCR4 gene. Biochim Biophys Acta Gene Regul Mech 2008; 1779(1): 40-50.
[http://dx.doi.org/10.1016/j.bbagrm.2007.09.005] [PMID: 18086574]
[15]
Saber MM, Karimiavargani M, Uzawa T, et al. Possible roles for the hominoid-specific DSCR4 gene in human cells. Genes Genet Syst 2021; 96(1): 1-11.
[http://dx.doi.org/10.1266/ggs.20-00012] [PMID: 33762515]
[16]
Murer G, Adelbrecht C, Lauritzen I, et al. An immunocytochemical study on the distribution of two G-protein-gated inward rectifier potassium channels (GIRK2 and GIRK4) in the adult rat brain. Neuroscience 1997; 80(2): 345-57.
[http://dx.doi.org/10.1016/S0306-4522(97)00001-8] [PMID: 9284339]
[17]
Wei J, Dlouhy SR, Bayer S, et al. In situ hybridization analysis of Girk2 expression in the developing central nervous system in normal and weaver mice. J Neuropathol Exp Neurol 1997; 56(7): 762-71.
[http://dx.doi.org/10.1097/00005072-199756070-00002] [PMID: 9210872]
[18]
Harashima C, Jacobowitz DM, Witta J, et al. Abnormal expression of the G-protein-activated inwardly rectifying potassium channel 2 (GIRK2) in hippocampus, frontal cortex, and substantia nigra of Ts65Dn mouse: A model of Down syndrome. J Comp Neurol 2006; 494(5): 815-33.
[http://dx.doi.org/10.1002/cne.20844] [PMID: 16374808]
[19]
Fernández-Alacid L, Watanabe M, Molnár E, Wickman K, Luján R. Developmental regulation of G protein-gated inwardly-rectifying K+ (GIRK/Kir3) channel subunits in the brain. Eur J Neurosci 2011; 34(11): 1724-36.
[http://dx.doi.org/10.1111/j.1460-9568.2011.07886.x] [PMID: 22098295]
[20]
Liao YJ, Jan YN, Jan LY. Heteromultimerization of G-protein-gated inwardly rectifying K+ channel proteins GIRK1 and GIRK2 and their altered expression in weaver brain. J Neurosci 1996; 16(22): 7137-50.
[http://dx.doi.org/10.1523/JNEUROSCI.16-22-07137.1996] [PMID: 8929423]
[21]
Siarey RJ, Villar AJ, Epstein CJ, Galdzicki Z. Abnormal synaptic plasticity in the Ts1Cje segmental trisomy 16 mouse model of Down syndrome. Neuropharmacology 2005; 49(1): 122-8.
[http://dx.doi.org/10.1016/j.neuropharm.2005.02.012] [PMID: 15992587]
[22]
Belichenko NP, Belichenko PV, Kleschevnikov AM, Salehi A, Reeves RH, Mobley WC. The “Down syndrome critical region” is sufficient in the mouse model to confer behavioral, neurophysiological, and synaptic phenotypes characteristic of Down syndrome. J Neurosci 2009; 29(18): 5938-48.
[http://dx.doi.org/10.1523/JNEUROSCI.1547-09.2009] [PMID: 19420260]
[23]
Jiang X, Liu C, Yu T, et al. Genetic dissection of the Down syndrome critical region. Hum Mol Genet 2015; 24(22): 6540-51.
[http://dx.doi.org/10.1093/hmg/ddv364] [PMID: 26374847]
[24]
Isomoto S, Kondo C, Kurachi Y. Inwardly rectifying potassium channels: Their molecular heterogeneity and function. Jpn J Physiol 1997; 47(1): 11-39.
[http://dx.doi.org/10.2170/jjphysiol.47.11] [PMID: 9159640]
[25]
Yamada M, Inanobe A, Kurachi Y. G protein regulation of potassium ion channels. Pharmacol Rev 1998; 50(4): 723-60.
[PMID: 9860808]
[26]
Mark MD, Herlitze S. G-protein mediated gating of inward-rectifier K+ channels. Eur J Biochem 2000; 267(19): 5830-6.
[http://dx.doi.org/10.1046/j.1432-1327.2000.01670.x] [PMID: 10998041]
[27]
Glaaser IW, Slesinger PA. Structural Insights into GIRK Channel Function. Int Rev Neurobiol 2015; 123: 117-60.
[http://dx.doi.org/10.1016/bs.irn.2015.05.014] [PMID: 26422984]
[28]
Rosenhouse-Dantsker A. Cholesterol binding sites in inwardly rectifying potassium channels. Adv Exp Med Biol 2019; 1135: 119-38.
[http://dx.doi.org/10.1007/978-3-030-14265-0_7] [PMID: 31098814]
[29]
Bichet D, Haass FA, Jan LY. Merging functional studies with structures of inward-rectifier K+ channels. Nat Rev Neurosci 2003; 4(12): 957-67.
[http://dx.doi.org/10.1038/nrn1244] [PMID: 14618155]
[30]
Koyrakh L, Luján R, Colón J, et al. Molecular and cellular diversity of neuronal G-protein-gated potassium channels. J Neurosci 2005; 25(49): 11468-78.
[http://dx.doi.org/10.1523/JNEUROSCI.3484-05.2005] [PMID: 16339040]
[31]
Cramer NP, Best TK, Stoffel M, Siarey RJ, Galdzicki Z. GABAB-GIRK2-mediated signaling in Down syndrome. Adv Pharmacol 2010; 58: 397-426.
[http://dx.doi.org/10.1016/S1054-3589(10)58015-3] [PMID: 20655490]
[32]
Hibino H, Inanobe A, Furutani K, Murakami S, Findlay I, Kurachi Y. Inwardly rectifying potassium channels: Their structure, function, and physiological roles. Physiol Rev 2010; 90(1): 291-366.
[http://dx.doi.org/10.1152/physrev.00021.2009] [PMID: 20086079]
[33]
Dascal N, Kahanovitch U. The roles of Gβγ and Gα in gating and regulation of GIRK channels. Int Rev Neurobiol 2015; 123: 27-85.
[http://dx.doi.org/10.1016/bs.irn.2015.06.001] [PMID: 26422982]
[34]
Jeremic D, Sanchez-Rodriguez I, Jimenez-Diaz L, Navarro-Lopez JD. Therapeutic potential of targeting G protein-gated inwardly rectifying potassium (GIRK) channels in the central nervous system. Pharmacol Ther 2021; 223: 107808.
[http://dx.doi.org/10.1016/j.pharmthera.2021.107808] [PMID: 33476640]
[35]
Kleschevnikov AM. Enhanced GIRK2 channel signaling in Down syndrome: A feasible role in the development of abnormal nascent neural circuits. Front Genet 2022; 13: 1006068.
[http://dx.doi.org/10.3389/fgene.2022.1006068] [PMID: 36171878]
[36]
Marron Fernandez de Velasco E, Zhang L, N Vo B, et al. GIRK2 splice variants and neuronal G protein-gated K+ channels: Implications for channel function and behavior. Sci Rep 2017; 7(1): 1639.
[http://dx.doi.org/10.1038/s41598-017-01820-2] [PMID: 28487514]
[37]
Whorton MR, MacKinnon R. Crystal structure of the mammalian GIRK2 K+ channel and gating regulation by G proteins, PIP2, and sodium. Cell 2011; 147(1): 199-208.
[http://dx.doi.org/10.1016/j.cell.2011.07.046] [PMID: 21962516]
[38]
Chen X, Johnston D. Constitutively active G-protein-gated inwardly rectifying K+ channels in dendrites of hippocampal CA1 pyramidal neurons. J Neurosci 2005; 25(15): 3787-92.
[http://dx.doi.org/10.1523/JNEUROSCI.5312-04.2005] [PMID: 15829630]
[39]
Gonzalez JC, Epps SA, Markwardt SJ, Wadiche JI, Overstreet-Wadiche L. Constitutive and synaptic activation of GIRK channels differentiates mature and newborn dentate granule cells. J Neurosci 2018; 38(29): 6513-26.
[http://dx.doi.org/10.1523/JNEUROSCI.0674-18.2018] [PMID: 29915136]
[40]
Lüscher C, Jan LY, Stoffel M, Malenka RC, Nicoll RA. G protein-coupled inwardly rectifying K+ channels (GIRKs) mediate postsynaptic but not presynaptic transmitter actions in hippocampal neurons. Neuron 1997; 19(3): 687-95.
[http://dx.doi.org/10.1016/S0896-6273(00)80381-5] [PMID: 9331358]
[41]
Wang W, Whorton MR, MacKinnon R. Quantitative analysis of mammalian GIRK2 channel regulation by G proteins, the signaling lipid PIP2 and Na+ in a reconstituted system. eLife 2014; 3: e03671.
[http://dx.doi.org/10.7554/eLife.03671] [PMID: 25049222]
[42]
Djebari S, Iborra-Lázaro G, Temprano-Carazo S, et al. G-protein-gated inwardly rectifying potassium (Kir3/GIRK) channels govern synaptic plasticity that supports hippocampal-dependent cognitive functions in male mice. J Neurosci 2021; 41(33): 7086-102.
[http://dx.doi.org/10.1523/JNEUROSCI.2849-20.2021] [PMID: 34261700]
[43]
Chung HJ, Ge WP, Qian X, Wiser O, Jan YN, Jan LY. G protein-activated inwardly rectifying potassium channels mediate depotentiation of long-term potentiation. Proc Natl Acad Sci USA 2009; 106(2): 635-40.
[http://dx.doi.org/10.1073/pnas.0811685106] [PMID: 19118199]
[44]
Signorini S, Liao YJ, Duncan SA, Jan LY, Stoffel M. Normal cerebellar development but susceptibility to seizures in mice lacking G protein-coupled, inwardly rectifying K + channel GIRK2. Proc Natl Acad Sci USA 1997; 94(3): 923-7.
[http://dx.doi.org/10.1073/pnas.94.3.923] [PMID: 9023358]
[45]
Blednov YA, Stoffel M, Chang SR, Adron Harris R. GIRK2 deficient mice. Physiol Behav 2001; 74(1-2): 109-17.
[http://dx.doi.org/10.1016/S0031-9384(01)00555-8] [PMID: 11564458]
[46]
Victoria NC, Marron Fernandez de Velasco E, Ostrovskaya O, et al. G Protein-Gated K+ channel ablation in forebrain pyramidal neurons selectively impairs fear learning. Biol Psychiatry 2016; 80(10): 796-806.
[http://dx.doi.org/10.1016/j.biopsych.2015.10.004] [PMID: 26612516]
[47]
Lüscher C, Slesinger PA. Emerging roles for G protein-gated inwardly rectifying potassium (GIRK) channels in health and disease. Nat Rev Neurosci 2010; 11(5): 301-15.
[http://dx.doi.org/10.1038/nrn2834] [PMID: 20389305]
[48]
Kessi M, Chen B, Peng J, et al. Intellectual disability and potassium channelopathies: A systematic review. Front Genet 2020; 11: 614.
[http://dx.doi.org/10.3389/fgene.2020.00614] [PMID: 32655623]
[49]
Basel-Vanagaite L, Shaffer L, Chitayat D. Keppen-Lubinsky syndrome: Expanding the phenotype. Am J Med Genet A 2009; 149A(8): 1827-9.
[http://dx.doi.org/10.1002/ajmg.a.32975] [PMID: 19610118]
[50]
Masotti A, Uva P, Davis-Keppen L, et al. Keppen-Lubinsky syndrome is caused by mutations in the inwardly rectifying K+ channel encoded by KCNJ6. Am J Hum Genet 2015; 96(2): 295-300.
[http://dx.doi.org/10.1016/j.ajhg.2014.12.011] [PMID: 25620207]
[51]
Horvath GA, Zhao Y, Tarailo-Graovac M, et al. Gain-of-function KCNJ6 mutation in a severe hyperkinetic movement disorder phenotype. Neuroscience 2018; 384: 152-64.
[http://dx.doi.org/10.1016/j.neuroscience.2018.05.031] [PMID: 29852244]
[52]
Patil N, Cox DR, Bhat D, Faham M, Myers RM, Peterson AS. A potassium channel mutation in weaver mice implicates membrane excitability in granule cell differentiation. Nat Genet 1995; 11(2): 126-9.
[http://dx.doi.org/10.1038/ng1095-126] [PMID: 7550338]
[53]
Schein JC, Wang JKT, Roffler-Tarlov SK. The effect of GIRK2wv on neurite growth, protein expression, and viability in the CNS-derived neuronal cell line, CATH.A-differentiated. Neuroscience 2005; 134(1): 21-32.
[http://dx.doi.org/10.1016/j.neuroscience.2005.03.043] [PMID: 15953684]
[54]
Ozaki M, Hashikawa T, Ikeda K, et al. Degeneration of pontine mossy fibres during cerebellar development in weaver mutant mice. Eur J Neurosci 2002; 16(4): 565-74.
[http://dx.doi.org/10.1046/j.1460-9568.2002.02111.x] [PMID: 12270032]
[55]
Cooper A, Grigoryan G, Guy-David L, Tsoory MM, Chen A, Reuveny E. Trisomy of the G protein-coupled K+ channel gene, Kcnj6, affects reward mechanisms, cognitive functions, and synaptic plasticity in mice. Proc Natl Acad Sci USA 2012; 109(7): 2642-7.
[http://dx.doi.org/10.1073/pnas.1109099109] [PMID: 22308328]
[56]
Mojabi FS, Fahimi A, Moghadam S, et al. GABAergic hyperinnervation of dentate granule cells in the Ts65Dn mouse model of down syndrome: Exploring the role of App. Hippocampus 2016; 26(12): 1641-54.
[http://dx.doi.org/10.1002/hipo.22664] [PMID: 27701794]
[57]
Chen XQ, Salehi A, Pearn ML, et al. Targeting increased levels of APP in Down syndrome: Posiphen-mediated reductions in APP and its products reverse endosomal phenotypes in the Ts65Dn mouse model. Alzheimers Dement 2021; 17(2): 271-92.
[PMID: 32975365]
[58]
Lysenko LV, Kim J, Madamba F, Tyrtyshnaia AA, Ruparelia A, Kleschevnikov AM. Developmental excitatory-to-inhibitory GABA polarity switch is delayed in Ts65Dn mice, a genetic model of Down syndrome. Neurobiol Dis 2018; 115: 1-8.
[http://dx.doi.org/10.1016/j.nbd.2018.03.005] [PMID: 29550538]
[59]
Kleschevnikov AM, Yu J, Kim J, et al. Evidence that increased Kcnj6 gene dose is necessary for deficits in behavior and dentate gyrus synaptic plasticity in the Ts65Dn mouse model of Down syndrome. Neurobiol Dis 2017; 103: 1-10.
[http://dx.doi.org/10.1016/j.nbd.2017.03.009] [PMID: 28342823]
[60]
Belichenko PV, Madani R, Rey-Bellet L, et al. An Anti-β-amyloid vaccine for treating cognitive deficits in a mouse model of down syndrome. PLoS One 2016; 11(3): e0152471.
[http://dx.doi.org/10.1371/journal.pone.0152471] [PMID: 27023444]
[61]
Belichenko PV, Kleschevnikov AM, Becker A, et al. Down syndrome cognitive phenotypes modeled in mice trisomic for All HSA 21 homologues. PLoS One 2015; 10(7): e0134861.
[http://dx.doi.org/10.1371/journal.pone.0134861] [PMID: 26230397]
[62]
Zhang L, Meng K, Jiang X, et al. Human chromosome 21 orthologous region on mouse chromosome 17 is a major determinant of Down syndrome-related developmental cognitive deficits. Hum Mol Genet 2014; 23(3): 578-89.
[http://dx.doi.org/10.1093/hmg/ddt446] [PMID: 24041763]
[63]
Lysenko LV, Kim J, Henry C, et al. Monoacylglycerol lipase inhibitor JZL184 improves behavior and neural properties in Ts65Dn mice, a model of down syndrome. PLoS One 2014; 9(12): e114521.
[http://dx.doi.org/10.1371/journal.pone.0114521] [PMID: 25474204]
[64]
Zhang L, Fu D, Belichenko PV, et al. Genetic analysis of Down syndrome facilitated by mouse chromosome engineering. Bioeng Bugs 2012; 3(1): 8-12.
[PMID: 22126738]
[65]
Kleschevnikov AM, Belichenko PV, Salehi A, Wu C. Discoveries in Down syndrome. Prog Brain Res 2012; 197: 199-221.
[http://dx.doi.org/10.1016/B978-0-444-54299-1.00010-8] [PMID: 22541294]
[66]
Kleschevnikov AM, Belichenko PV, Gall J, et al. Increased efficiency of the GABAA and GABAB receptor-mediated neurotransmission in the Ts65Dn mouse model of Down syndrome. Neurobiol Dis 2012; 45(2): 683-91.
[http://dx.doi.org/10.1016/j.nbd.2011.10.009] [PMID: 22062771]
[67]
Kleschevnikov AM, Belichenko PV, Faizi M, et al. Deficits in cognition and synaptic plasticity in a mouse model of Down syndrome ameliorated by GABAB receptor antagonists. J Neurosci 2012; 32(27): 9217-27.
[http://dx.doi.org/10.1523/JNEUROSCI.1673-12.2012] [PMID: 22764230]
[68]
Popov VI, Kleschevnikov AM, Klimenko OA, Stewart MG, Belichenko PV. Three-dimensional synaptic ultrastructure in the dentate gyrus and hippocampal area CA3 in the Ts65Dn mouse model of down syndrome. J Comp Neurol 2011; 519(7): 1338-54.
[http://dx.doi.org/10.1002/cne.22573] [PMID: 21452200]
[69]
Liu C, Belichenko PV, Zhang L, et al. Mouse models for Down syndrome-associated developmental cognitive disabilities. Dev Neurosci 2011; 33(5): 404-13.
[http://dx.doi.org/10.1159/000329422] [PMID: 21865664]
[70]
Faizi M, Bader PL, Tun C, et al. Comprehensive behavioral phenotyping of Ts65Dn mouse model of Down syndrome: Activation of β1-adrenergic receptor by xamoterol as a potential cognitive enhancer. Neurobiol Dis 2011; 43(2): 397-413.
[http://dx.doi.org/10.1016/j.nbd.2011.04.011] [PMID: 21527343]
[71]
Belichenko PV, Kleschevnikov AM. Deficiency of adult neurogenesis in the Ts65Dn mouse model of Down syndrome. In: Dey S, Ed. Genetics and Etiology of Down syndrome. InTech 2011; pp. 177-92.
[72]
Yu T, Liu C, Belichenko P, et al. Effects of individual segmental trisomies of human chromosome 21 syntenic regions on hippocampal long-term potentiation and cognitive behaviors in mice. Brain Res 2010; 1366: 162-71.
[http://dx.doi.org/10.1016/j.brainres.2010.09.107] [PMID: 20932954]
[73]
Salehi A, Faizi M, Colas D, et al. Restoration of norepinephrine-modulated contextual memory in a mouse model of Down syndrome. Sci Transl Med 2009; 1(7): 7ra17.
[http://dx.doi.org/10.1126/scitranslmed.3000258] [PMID: 20368182]
[74]
Belichenko PV, Kleschevnikov AM, Masliah E, et al. Excitatory-inhibitory relationship in the fascia dentata in the Ts65Dn mouse model of down syndrome. J Comp Neurol 2009; 512(4): 453-66.
[http://dx.doi.org/10.1002/cne.21895] [PMID: 19034952]
[75]
Belichenko PV, Kleschevnikov AM, Salehi A, Epstein CJ, Mobley WC. Synaptic and cognitive abnormalities in mouse models of down syndrome: Exploring genotype-phenotype relationships. J Comp Neurol 2007; 504(4): 329-45.
[http://dx.doi.org/10.1002/cne.21433] [PMID: 17663443]
[76]
Salehi A, Delcroix JD, Belichenko PV, et al. Increased App expression in a mouse model of Down’s syndrome disrupts NGF transport and causes cholinergic neuron degeneration. Neuron 2006; 51(1): 29-42.
[http://dx.doi.org/10.1016/j.neuron.2006.05.022] [PMID: 16815330]
[77]
Kleschevnikov AM, Van Volkinburg J, Zhan K, Mobley WC. Expression of GIRK2 and related proteins after chronic administrations of Prozac in Ts65Dn mice, a genetic model of Down syndrome
[78]
Kleschevnikov AM, Belichenko PV, Villar AJ, Epstein CJ, Malenka RC, Mobley WC. Hippocampal long-term potentiation suppressed by increased inhibition in the Ts65Dn mouse, a genetic model of Down syndrome. J Neurosci 2004; 24(37): 8153-60.
[http://dx.doi.org/10.1523/JNEUROSCI.1766-04.2004] [PMID: 15371516]
[79]
Belichenko PV, Masliah E, Kleschevnikov AM, et al. Synaptic structural abnormalities in the Ts65Dn mouse model of down syndrome. J Comp Neurol 2004; 480(3): 281-98.
[http://dx.doi.org/10.1002/cne.20337] [PMID: 15515178]
[80]
Martínez-Cué C, Delatour B, Potier MC. Treating enhanced GABAergic inhibition in Down syndrome: Use of GABA α5-selective inverse agonists. Neurosci Biobehav Rev 2014; 46(Pt 2): 218-27.
[http://dx.doi.org/10.1016/j.neubiorev.2013.12.008] [PMID: 24412222]
[81]
Ruparelia A, Pearn ML, Mobley WC. Cognitive and pharmacological insights from the Ts65Dn mouse model of Down syndrome. Curr Opin Neurobiol 2012; 22(5): 880-6.
[http://dx.doi.org/10.1016/j.conb.2012.05.002] [PMID: 22658745]
[82]
Rueda N, Flórez J, Martínez-Cué C. Mouse models of Down syndrome as a tool to unravel the causes of mental disabilities. Neural Plast 2012; 2012: 584071.
[http://dx.doi.org/10.1155/2012/584071] [PMID: 22685678]
[83]
Möhler H. Cognitive enhancement by pharmacological and behavioral interventions: The murine Down syndrome model. Biochem Pharmacol 2012; 84(8): 994-9.
[http://dx.doi.org/10.1016/j.bcp.2012.06.028] [PMID: 22898099]
[84]
Dierssen M, Fillat C, Crnic L, Arbonés M, Flórez J, Estivill X. Murine models for Down syndrome. Physiol Behav 2001; 73(5): 859-71.
[http://dx.doi.org/10.1016/S0031-9384(01)00523-6] [PMID: 11566219]
[85]
Dierssen M, Herault Y, Estivill X. Aneuploidy: From a physiological mechanism of variance to Down syndrome. Physiol Rev 2009; 89(3): 887-920.
[http://dx.doi.org/10.1152/physrev.00032.2007] [PMID: 19584316]
[86]
Hamlett ED, Boger HA, Ledreux A, et al. Cognitive impairment, neuroimaging, and Alzheimer neuropathology in mouse models of down syndrome. Curr Alzheimer Res 2016; 13(1): 35-52.
[http://dx.doi.org/10.2174/1567205012666150921095505] [PMID: 26391050]
[87]
Haydar TF, Reeves RH. Trisomy 21 and early brain development. Trends Neurosci 2012; 35(2): 81-91.
[http://dx.doi.org/10.1016/j.tins.2011.11.001] [PMID: 22169531]
[88]
Lott IT. Neurological phenotypes for Down syndrome across the life span. Prog Brain Res 2012; 197: 101-21.
[http://dx.doi.org/10.1016/B978-0-444-54299-1.00006-6] [PMID: 22541290]
[89]
Reeves RH, Delabar J, Potier MC, et al. Paving the way for therapy: The second international conference of the trisomy 21 research society. Mol Syndromol 2018; 9(6): 279-86.
[http://dx.doi.org/10.1159/000494231] [PMID: 30800043]
[90]
Martínez Cué C, Dierssen M. Plasticity as a therapeutic target for improving cognition and behavior in Down syndrome. Prog Brain Res 2020; 251: 269-302.
[http://dx.doi.org/10.1016/bs.pbr.2019.11.001] [PMID: 32057310]
[91]
Costa ACS, Scott-McKean JJ. Prospects for improving brain function in individuals with Down syndrome. CNS Drugs 2013; 27(9): 679-702.
[http://dx.doi.org/10.1007/s40263-013-0089-3] [PMID: 23821040]
[92]
Zorrilla de San Martin J, Delabar JM, Bacci A, Potier MC. GABAergic over-inhibition, a promising hypothesis for cognitive deficits in Down syndrome. Free Radic Biol Med 2018; 114: 33-9.
[http://dx.doi.org/10.1016/j.freeradbiomed.2017.10.002] [PMID: 28993272]
[93]
Tosh J, Tybulewicz V, Fisher EMC. Mouse models of aneuploidy to understand chromosome disorders. Mamm Genome 2022; 33(1): 157-68.
[http://dx.doi.org/10.1007/s00335-021-09930-z] [PMID: 34719726]
[94]
Cannavo C, Tosh J, Fisher EMC, Wiseman FK. Using mouse models to understand Alzheimer’s disease mechanisms in the context of trisomy of chromosome 21. Prog Brain Res 2020; 251: 181-208.
[http://dx.doi.org/10.1016/bs.pbr.2019.10.004] [PMID: 32057307]
[95]
Herault Y, Delabar JM, Fisher EMC, Tybulewicz VLJ, Yu E, Brault V. Rodent models in Down syndrome research: Impact and future opportunities. Dis Model Mech 2017; 10(10): 1165-86.
[http://dx.doi.org/10.1242/dmm.029728] [PMID: 28993310]
[96]
Gardiner K, Fortna A, Bechtel L, Davisson MT. Mouse models of Down syndrome: How useful can they be? Comparison of the gene content of human chromosome 21 with orthologous mouse genomic regions. Gene 2003; 318: 137-47.
[http://dx.doi.org/10.1016/S0378-1119(03)00769-8] [PMID: 14585506]
[97]
Davisson MT, Schmidt C, Akeson EC. Segmental trisomy of murine chromosome 16: A new model system for studying Down syndrome. Prog Clin Biol Res 1990; 360: 263-80.
[PMID: 2147289]
[98]
Davisson MT, Schmidt C, Reeves RH, et al. Segmental trisomy as a mouse model for Down syndrome. Prog Clin Biol Res 1993; 384: 117-33.
[PMID: 8115398]
[99]
Edgin JO, Mason GM, Spanò G, Fernández A, Nadel L. Human and mouse model cognitive phenotypes in Down syndrome. Prog Brain Res 2012; 197: 123-51.
[http://dx.doi.org/10.1016/B978-0-444-54299-1.00007-8] [PMID: 22541291]
[100]
Reeves RH, Irving NG, Moran TH, et al. A mouse model for Down syndrome exhibits learning and behaviour deficits. Nat Genet 1995; 11(2): 177-84.
[http://dx.doi.org/10.1038/ng1095-177] [PMID: 7550346]
[101]
Holtzman DM, Santucci D, Kilbridge J, et al. Developmental abnormalities and age-related neurodegeneration in a mouse model of Down syndrome. Proc Natl Acad Sci USA 1996; 93(23): 13333-8.
[http://dx.doi.org/10.1073/pnas.93.23.13333] [PMID: 8917591]
[102]
Hyde LA, Frisone DF, Crnic LS. Ts65Dn mice, a model for Down syndrome, have deficits in context discrimination learning suggesting impaired hippocampal function. Behav Brain Res 2001; 118(1): 53-60.
[http://dx.doi.org/10.1016/S0166-4328(00)00313-2] [PMID: 11163633]
[103]
Costa ACS, Scott-McKean JJ, Stasko MR. Acute injections of the NMDA receptor antagonist memantine rescue performance deficits of the Ts65Dn mouse model of Down syndrome on a fear conditioning test. Neuropsychopharmacology 2008; 33(7): 1624-32.
[http://dx.doi.org/10.1038/sj.npp.1301535] [PMID: 17700645]
[104]
Harashima C, Jacobowitz DM, Stoffel M, et al. Elevated expression of the G-protein-activated inwardly rectifying potassium channel 2 (GIRK2) in cerebellar unipolar brush cells of a Down syndrome mouse model. Cell Mol Neurobiol 2006; 26(4-6): 717-32.
[http://dx.doi.org/10.1007/s10571-006-9066-4] [PMID: 16783527]
[105]
Wang X, Zhao Y, Zhang X, et al. Loss of sorting nexin 27 contributes to excitatory synaptic dysfunction by modulating glutamate receptor recycling in Down’s syndrome. Nat Med 2013; 19(4): 473-80.
[http://dx.doi.org/10.1038/nm.3117] [PMID: 23524343]
[106]
Lunn ML, Nassirpour R, Arrabit C, et al. A unique sorting nexin regulates trafficking of potassium channels via a PDZ domain interaction. Nat Neurosci 2007; 10(10): 1249-59.
[http://dx.doi.org/10.1038/nn1953] [PMID: 17828261]
[107]
Best TK, Siarey RJ, Galdzicki Z. Ts65Dn, a mouse model of Down syndrome, exhibits increased GABAB-induced potassium current. J Neurophysiol 2007; 97(1): 892-900.
[http://dx.doi.org/10.1152/jn.00626.2006] [PMID: 17093127]
[108]
Roncacé V, Burattini C, Stagni F, et al. Neuroanatomical alterations and synaptic plasticity impairment in the perirhinal cortex of the Ts65Dn mouse model of Down syndrome. Neurobiol Dis 2017; 106: 89-100.
[http://dx.doi.org/10.1016/j.nbd.2017.06.017] [PMID: 28651891]
[109]
Kobayashi T, Washiyama K, Ikeda K. Inhibition of G protein-activated inwardly rectifying K+ channels by fluoxetine (Prozac). Br J Pharmacol 2003; 138(6): 1119-28.
[http://dx.doi.org/10.1038/sj.bjp.0705172] [PMID: 12684268]
[110]
Sakai N, Tanaka C. Inhibitory modulation of long-term potentiation via the 5-HT1A receptor in slices of the rat hippocampal dentate gyrus. Brain Res 1993; 613(2): 326-30.
[http://dx.doi.org/10.1016/0006-8993(93)90921-9] [PMID: 8186986]
[111]
Fuller RW, Wong DT. Inhibition of serotonin reuptake. Fed Proc 1977; 36(8): 2154-8.
[PMID: 326579]
[112]
Guidi S, Stagni F, Bianchi P, et al. Prenatal pharmacotherapy rescues brain development in a Down’s syndrome mouse model. Brain 2014; 137(2): 380-401.
[http://dx.doi.org/10.1093/brain/awt340] [PMID: 24334313]
[113]
Bianchi P, Ciani E, Guidi S, et al. Early pharmacotherapy restores neurogenesis and cognitive performance in the Ts65Dn mouse model for Down syndrome. J Neurosci 2010; 30(26): 8769-79.
[http://dx.doi.org/10.1523/JNEUROSCI.0534-10.2010] [PMID: 20592198]
[114]
Stagni F, Giacomini A, Guidi S, et al. Long-term effects of neonatal treatment with fluoxetine on cognitive performance in Ts65Dn mice. Neurobiol Dis 2015; 74: 204-18.
[http://dx.doi.org/10.1016/j.nbd.2014.12.005] [PMID: 25497735]
[115]
Begenisic T, Baroncelli L, Sansevero G, et al. Fluoxetine in adulthood normalizes GABA release and rescues hippocampal synaptic plasticity and spatial memory in a mouse model of Down syndrome. Neurobiol Dis 2014; 63: 12-9.
[http://dx.doi.org/10.1016/j.nbd.2013.11.010] [PMID: 24269730]
[116]
Heinen M, Hettich MM, Ryan DP, Schnell S, Paesler K, Ehninger D. Adult-onset fluoxetine treatment does not improve behavioral impairments and may have adverse effects on the Ts65Dn mouse model of Down syndrome. Neural Plast 2012; 2012: 1-10.
[http://dx.doi.org/10.1155/2012/467251] [PMID: 22848851]
[117]
Luo H, Marron Fernandez de Velasco E, Wickman K. Neuronal G protein-gated K+ channels. Am J Physiol Cell Physiol 2022; 323(2): C439-60.
[http://dx.doi.org/10.1152/ajpcell.00102.2022] [PMID: 35704701]
[118]
Dal Prà I, Armato U, Chiarini A. Family C G-protein-coupled receptors in Alzheimer’s disease and therapeutic implications. Front Pharmacol 2019; 10: 1282.
[http://dx.doi.org/10.3389/fphar.2019.01282] [PMID: 31719824]
[119]
Li Y, Sun H, Chen Z, Xu H, Bu G, Zheng H. Implications of GABAergic neurotransmission in Alzheimer’s disease. Front Aging Neurosci 2016; 8: 31.
[http://dx.doi.org/10.3389/fnagi.2016.00031] [PMID: 26941642]
[120]
Nava-Mesa MO, Jiménez-Díaz L, Yajeya J, Navarro-Lopez JD. GABAergic neurotransmission and new strategies of neuromodulation to compensate synaptic dysfunction in early stages of Alzheimer’s disease. Front Cell Neurosci 2014; 8: 167.
[http://dx.doi.org/10.3389/fncel.2014.00167] [PMID: 24987334]
[121]
Thathiah A, De Strooper B. G protein-coupled receptors, cholinergic dysfunction, and Abeta toxicity in Alzheimer’s disease. Sci Signal 2009; 2(93): re8.
[http://dx.doi.org/10.1126/scisignal.293re8] [PMID: 19843960]
[122]
Jeremic D, Jiménez-Díaz L, Navarro-López JD. Past, present and future of therapeutic strategies against amyloid-β peptides in Alzheimer’s disease: A systematic review. Ageing Res Rev 2021; 72: 101496.
[http://dx.doi.org/10.1016/j.arr.2021.101496] [PMID: 34687956]
[123]
Calvo-Flores GB, Vinnakota C, Govindpani K, Waldvogel HJ, Faull RLM, Kwakowsky A. The GABAergic system as a therapeutic target for Alzheimer’s disease. J Neurochem 2018; 146(6): 649-69.
[http://dx.doi.org/10.1111/jnc.14345] [PMID: 29645219]
[124]
Chu DCM, Penney JB Jr, Young AB. Quantitative autoradiography of hippocampal GABAB and GABAA receptor changes in Alzheimer’s disease. Neurosci Lett 1987; 82(3): 246-52.
[http://dx.doi.org/10.1016/0304-3940(87)90264-3] [PMID: 2827074]
[125]
Chu DCM, Penney JB Jr, Young AB. Cortical GABAB and GABAA receptors in Alzheimer’s disease: A quantitative autoradiographic study. Neurology 1987; 37(9): 1454-9.
[http://dx.doi.org/10.1212/WNL.37.9.1454] [PMID: 2819782]
[126]
Iwakiri M, Mizukami K, Ikonomovic MD, et al. Changes in hippocampal GABABR1 subunit expression in Alzheimer’s patients: Association with Braak staging. Acta Neuropathol 2005; 109(5): 467-74.
[http://dx.doi.org/10.1007/s00401-005-0985-9] [PMID: 15759131]
[127]
Martín-Belmonte A, Aguado C, Alfaro-Ruíz R, et al. Reduction in the neuronal surface of post and presynaptic GABA B receptors in the hippocampus in a mouse model of Alzheimer’s disease. Brain Pathol 2020; 30(3): 554-75.
[http://dx.doi.org/10.1111/bpa.12802] [PMID: 31729777]
[128]
Martín-Belmonte A, Aguado C, Alfaro-Ruíz R, et al. Density of GABAB receptors is reduced in granule cells of the hippocampus in a mouse model of Alzheimer’s disease. Int J Mol Sci 2020; 21(7): 2459.
[http://dx.doi.org/10.3390/ijms21072459] [PMID: 32252271]
[129]
Schwenk J, Pérez-Garci E, Schneider A, et al. Modular composition and dynamics of native GABAB receptors identified by high-resolution proteomics. Nat Neurosci 2016; 19(2): 233-42.
[http://dx.doi.org/10.1038/nn.4198] [PMID: 26691831]
[130]
Jo S, Yarishkin O, Hwang YJ, et al. GABA from reactive astrocytes impairs memory in mouse models of Alzheimer’s disease. Nat Med 2014; 20(8): 886-96.
[http://dx.doi.org/10.1038/nm.3639] [PMID: 24973918]
[131]
Alfaro-Ruiz R, Martín-Belmonte A, Aguado C, et al. The expression and localisation of G-protein-coupled inwardly rectifying potassium (GIRK) channels is differentially altered in the hippocampus of two mouse models of Alzheimer’s disease. Int J Mol Sci 2021; 22(20): 11106.
[http://dx.doi.org/10.3390/ijms222011106] [PMID: 34681766]
[132]
Martín-Belmonte A, Aguado C, Alfaro-Ruiz R, et al. Nanoscale alterations in GABAB receptors and GIRK channel organization on the hippocampus of APP/PS1 mice. Alzheimers Res Ther 2022; 14(1): 136.
[http://dx.doi.org/10.1186/s13195-022-01078-5] [PMID: 36131327]
[133]
Temprano-Carazo S, Contreras A, Saura CA, Navarro-López JD, Jiménez-Díaz L. Spatial memory training counteracts hippocampal GIRK channel decrease in the transgenic APPSw,Ind J9 Alzheimer’s disease mouse model. Int J Mol Sci 2022; 23(21): 13444.
[http://dx.doi.org/10.3390/ijms232113444] [PMID: 36362230]
[134]
Nava-Mesa MO, Jiménez-Díaz L, Yajeya J, Navarro-Lopez JD. Amyloid-β induces synaptic dysfunction through G protein-gated inwardly rectifying potassium channels in the fimbria-CA3 hippocampal synapse. Front Cell Neurosci 2013; 7: 117.
[http://dx.doi.org/10.3389/fncel.2013.00117] [PMID: 23898239]
[135]
Mayordomo-Cava J, Yajeya J, Navarro-López JD, Jiménez-Díaz L. Amyloid-β(25-35) modulates the expression of GirK and KCNQ channel genes in the hippocampus. PLoS One 2015; 10(7): e0134385.
[http://dx.doi.org/10.1371/journal.pone.0134385] [PMID: 26218288]
[136]
Sánchez-Rodríguez I, Temprano-Carazo S, Nájera A, et al. Activation of G-protein-gated inwardly rectifying potassium (Kir3/GirK) channels rescues hippocampal functions in a mouse model of early amyloid-β pathology. Sci Rep 2017; 7(1): 14658.
[http://dx.doi.org/10.1038/s41598-017-15306-8] [PMID: 29116174]
[137]
Sánchez-Rodríguez I, Djebari S, Temprano-Carazo S, et al. Hippocampal long‐term synaptic depression and memory deficits induced in early amyloidopathy are prevented by enhancing G‐protein‐gated inwardly rectifying potassium channel activity. J Neurochem 2020; 153(3): 362-76.
[http://dx.doi.org/10.1111/jnc.14946] [PMID: 31875959]
[138]
Chumakov I, Nabirotchkin S, Cholet N, et al. Combining two repurposed drugs as a promising approach for Alzheimer’s disease therapy. Sci Rep 2015; 5(1): 7608.
[http://dx.doi.org/10.1038/srep07608] [PMID: 25566747]
[139]
Pilipenko V, Narbute K, Beitnere U, et al. Very low doses of muscimol and baclofen ameliorate cognitive deficits and regulate protein expression in the brain of a rat model of streptozocin-induced Alzheimer’s disease. Eur J Pharmacol 2018; 818: 381-99.
[http://dx.doi.org/10.1016/j.ejphar.2017.11.012] [PMID: 29133125]
[140]
Lee BY, Ban JY, Seong YH. Chronic stimulation of GABAA receptor with muscimol reduces amyloid β protein (25–35)-induced neurotoxicity in cultured rat cortical cells. Neurosci Res 2005; 52(4): 347-56.
[http://dx.doi.org/10.1016/j.neures.2005.04.008] [PMID: 15896866]
[141]
May LM, Anggono V, Gooch HM, et al. G-Protein-Coupled Inwardly Rectifying Potassium (GIRK) channel activation by the p75 neurotrophin receptor is required for amyloid β toxicity. Front Neurosci 2017; 11: 455.
[http://dx.doi.org/10.3389/fnins.2017.00455] [PMID: 28848381]
[142]
Shabala L, Howells C, West AK, Chung RS. Prolonged Aβ treatment leads to impairment in the ability of primary cortical neurons to maintain K+ and Ca2+ homeostasis. Mol Neurodegener 2010; 5(1): 30.
[http://dx.doi.org/10.1186/1750-1326-5-30] [PMID: 20704753]

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