메뉴 건너뛰기




Volumn 122, Issue , 2016, Pages 1-11

Toward more predictive genetic mouse models of Alzheimer's disease

Author keywords

Alzheimer's disease; Biomarkers; Genetic model; Genomics; Mouse model; Neuroinflammation; Preclinical model; Translational assays

Indexed keywords

BIOLOGICAL MARKER; TAU PROTEIN;

EID: 84951762738     PISSN: 03619230     EISSN: 18732747     Source Type: Journal    
DOI: 10.1016/j.brainresbull.2015.12.003     Document Type: Review
Times cited : (126)

References (82)
  • 1
    • 33845892752 scopus 로고    scopus 로고
    • Systematic meta-analyses of Alzheimer disease genetic association studies: the AlzGene database
    • Bertram L., et al. Systematic meta-analyses of Alzheimer disease genetic association studies: the AlzGene database. Nat. Genet. 2007, 39(1):17-23.
    • (2007) Nat. Genet. , vol.39 , Issue.1 , pp. 17-23
    • Bertram, L.1
  • 2
    • 84860460906 scopus 로고    scopus 로고
    • Mouse models of Alzheimer's disease
    • Hall A.M., Roberson E.D. Mouse models of Alzheimer's disease. Brain Res. Bull 2012, 88(1):3-12.
    • (2012) Brain Res. Bull , vol.88 , Issue.1 , pp. 3-12
    • Hall, A.M.1    Roberson, E.D.2
  • 3
    • 84951836404 scopus 로고    scopus 로고
    • Keeping score, strengthening policy and fighting bad actors over access to research tools
    • Bubela T., Cook-Deegan R. Keeping score, strengthening policy and fighting bad actors over access to research tools. Nat. Biotechnol. 2015, 33(2):143-147.
    • (2015) Nat. Biotechnol. , vol.33 , Issue.2 , pp. 143-147
    • Bubela, T.1    Cook-Deegan, R.2
  • 4
    • 84872826790 scopus 로고    scopus 로고
    • Amyloid deposition and advanced age fails to induce Alzheimer's type progression in a double knock-in mouse model
    • Malthankar-Phatak G.H., et al. Amyloid deposition and advanced age fails to induce Alzheimer's type progression in a double knock-in mouse model. Aging Dis. 2012, 3(2):141-155.
    • (2012) Aging Dis. , vol.3 , Issue.2 , pp. 141-155
    • Malthankar-Phatak, G.H.1
  • 5
    • 84899647552 scopus 로고    scopus 로고
    • Single App knock-in mouse models of Alzheimer's disease
    • Saito T., et al. Single App knock-in mouse models of Alzheimer's disease. Nat. Neurosci. 2014, 17(5):661-663.
    • (2014) Nat. Neurosci. , vol.17 , Issue.5 , pp. 661-663
    • Saito, T.1
  • 6
    • 84905706814 scopus 로고    scopus 로고
    • Knock-in of human BACE1 cleaves murine APP and reiterates Alzheimer-like phenotypes
    • Plucinska K., et al. Knock-in of human BACE1 cleaves murine APP and reiterates Alzheimer-like phenotypes. J. Neurosci. 2014, 34(32):10710-10728.
    • (2014) J. Neurosci. , vol.34 , Issue.32 , pp. 10710-10728
    • Plucinska, K.1
  • 7
    • 84874605021 scopus 로고    scopus 로고
    • Reversible pathologic and cognitive phenotypes in an inducible model of Alzheimer-amyloidosis
    • Melnikova T., et al. Reversible pathologic and cognitive phenotypes in an inducible model of Alzheimer-amyloidosis. J. Neurosci. 2013, 33(9):3765-3779.
    • (2013) J. Neurosci. , vol.33 , Issue.9 , pp. 3765-3779
    • Melnikova, T.1
  • 8
    • 84864382491 scopus 로고    scopus 로고
    • Cognitive defects are reversible in inducible mice expressing pro-aggregant full-length human Tau
    • Van der Jeugd A., et al. Cognitive defects are reversible in inducible mice expressing pro-aggregant full-length human Tau. Acta Neuropathol. 2012, 123(6):787-805.
    • (2012) Acta Neuropathol. , vol.123 , Issue.6 , pp. 787-805
    • Van der Jeugd, A.1
  • 9
    • 84901779689 scopus 로고    scopus 로고
    • Genetic modulation of soluble Abeta rescues cognitive and synaptic impairment in a mouse model of Alzheimer's disease
    • Fowler S.W., et al. Genetic modulation of soluble Abeta rescues cognitive and synaptic impairment in a mouse model of Alzheimer's disease. J. Neurosci. 2014, 34(23):7871-7885.
    • (2014) J. Neurosci. , vol.34 , Issue.23 , pp. 7871-7885
    • Fowler, S.W.1
  • 10
    • 84944324335 scopus 로고    scopus 로고
    • Eta-Secretase processing of APP inhibits neuronal activity in the hippocampus
    • Willem M., et al. eta-Secretase processing of APP inhibits neuronal activity in the hippocampus. Nature 2015, 526(7573):443-447.
    • (2015) Nature , vol.526 , Issue.7573 , pp. 443-447
    • Willem, M.1
  • 11
    • 84888093231 scopus 로고    scopus 로고
    • Clustering of transcriptional profiles identifies changes to insulin signaling as an early event in a mouse model of Alzheimer's disease
    • Jackson H.M., et al. Clustering of transcriptional profiles identifies changes to insulin signaling as an early event in a mouse model of Alzheimer's disease. BMC Genomics 2013, 14:831.
    • (2013) BMC Genomics , vol.14 , pp. 831
    • Jackson, H.M.1
  • 12
    • 84907173621 scopus 로고    scopus 로고
    • Temporal gene profiling of the 5XFAD transgenic mouse model highlights the importance of microglial activation in Alzheimer's disease
    • Landel V., et al. Temporal gene profiling of the 5XFAD transgenic mouse model highlights the importance of microglial activation in Alzheimer's disease. Mol. Neurodegener. 2014, 9:33.
    • (2014) Mol. Neurodegener. , vol.9 , pp. 33
    • Landel, V.1
  • 13
    • 84894350553 scopus 로고    scopus 로고
    • Transcriptome analysis of distinct mouse strains reveals kinesin light chain-1 splicing as an amyloid-beta accumulation modifier
    • Morihara T., et al. Transcriptome analysis of distinct mouse strains reveals kinesin light chain-1 splicing as an amyloid-beta accumulation modifier. Proc. Natl. Acad. Sci. U. S. A. 2014, 111(7):2638-2643.
    • (2014) Proc. Natl. Acad. Sci. U. S. A. , vol.111 , Issue.7 , pp. 2638-2643
    • Morihara, T.1
  • 14
    • 84937763848 scopus 로고    scopus 로고
    • Mouse models rarely mimic the transcriptome of human neurodegenerative diseases: a systematic bioinformatics-based critique of preclinical models
    • Burns T.C., et al. Mouse models rarely mimic the transcriptome of human neurodegenerative diseases: a systematic bioinformatics-based critique of preclinical models. Eur. J. Pharmacol. 2015, 759:101-117.
    • (2015) Eur. J. Pharmacol. , vol.759 , pp. 101-117
    • Burns, T.C.1
  • 15
    • 84936998627 scopus 로고    scopus 로고
    • Tau reduction prevents Abeta-induced axonal transport deficits by blocking activation of GSK3beta
    • Vossel K.A., et al. Tau reduction prevents Abeta-induced axonal transport deficits by blocking activation of GSK3beta. J. Cell Biol. 2015, 209(3):419-433.
    • (2015) J. Cell Biol. , vol.209 , Issue.3 , pp. 419-433
    • Vossel, K.A.1
  • 16
    • 84901513015 scopus 로고    scopus 로고
    • Cumulative effects of the ApoE genotype and gender on the synaptic proteome and oxidative stress in the mouse brain
    • Shi L., et al. Cumulative effects of the ApoE genotype and gender on the synaptic proteome and oxidative stress in the mouse brain. Int. J. Neuropsychopharmacol. 2014, 17(11):1863-1879.
    • (2014) Int. J. Neuropsychopharmacol. , vol.17 , Issue.11 , pp. 1863-1879
    • Shi, L.1
  • 17
    • 84988215565 scopus 로고    scopus 로고
    • Low plasma ApoE levels are associated with smaller hippocampal size in the Alzheimer's disease neuroimaging initiative cohort
    • Teng E., et al. Low plasma ApoE levels are associated with smaller hippocampal size in the Alzheimer's disease neuroimaging initiative cohort. Dement. Geriatr. Cogn. Disord. 2015, 39(3-4):154-166.
    • (2015) Dement. Geriatr. Cogn. Disord. , vol.39 , Issue.3-4 , pp. 154-166
    • Teng, E.1
  • 18
    • 84872057940 scopus 로고    scopus 로고
    • TREM2 variants in Alzheimer's disease
    • Guerreiro R., et al. TREM2 variants in Alzheimer's disease. N. Engl. J. Med. 2013, 368(2):117-127.
    • (2013) N. Engl. J. Med. , vol.368 , Issue.2 , pp. 117-127
    • Guerreiro, R.1
  • 19
    • 84945443544 scopus 로고    scopus 로고
    • CD33 modulates TREM2: convergence of Alzheimer loci
    • Chan G., et al. CD33 modulates TREM2: convergence of Alzheimer loci. Nat. Neurosci. 2015, 18(11):1556-1558.
    • (2015) Nat. Neurosci. , vol.18 , Issue.11 , pp. 1556-1558
    • Chan, G.1
  • 21
    • 84879113935 scopus 로고    scopus 로고
    • TREM2 in neurodegeneration: evidence for association of the p. R47H variant with frontotemporal dementia and Parkinson's disease
    • Rayaprolu S., et al. TREM2 in neurodegeneration: evidence for association of the p. R47H variant with frontotemporal dementia and Parkinson's disease. Mol. Neurodegener. 2013, 8:19.
    • (2013) Mol. Neurodegener. , vol.8 , pp. 19
    • Rayaprolu, S.1
  • 22
    • 85058205970 scopus 로고    scopus 로고
    • Heterozygous TREM2 mutations in frontotemporal dementia
    • e7-e10
    • Borroni B., et al. Heterozygous TREM2 mutations in frontotemporal dementia. Neurobiol. Aging 2014, 35(4):934. e7-10.
    • (2014) Neurobiol. Aging , vol.35 , Issue.4 , pp. 934
    • Borroni, B.1
  • 23
    • 84897398469 scopus 로고    scopus 로고
    • TREM2 variant p.R47H as a risk factor for sporadic amyotrophic lateral sclerosis
    • Cady J., et al. TREM2 variant p.R47H as a risk factor for sporadic amyotrophic lateral sclerosis. JAMA Neurol. 2014, 71(4):449-453.
    • (2014) JAMA Neurol. , vol.71 , Issue.4 , pp. 449-453
    • Cady, J.1
  • 24
    • 84902243410 scopus 로고    scopus 로고
    • Altered microglial response to Abeta plaques in APPPS1-21 mice heterozygous for TREM2
    • Ulrich J.D., et al. Altered microglial response to Abeta plaques in APPPS1-21 mice heterozygous for TREM2. Mol. Neurodegener. 2014, 9:20.
    • (2014) Mol. Neurodegener. , vol.9 , pp. 20
    • Ulrich, J.D.1
  • 25
    • 84924737031 scopus 로고    scopus 로고
    • TREM2 deficiency eliminates TREM2+ inflammatory macrophages and ameliorates pathology in Alzheimer's disease mouse models
    • Jay T.R., et al. TREM2 deficiency eliminates TREM2+ inflammatory macrophages and ameliorates pathology in Alzheimer's disease mouse models. J. Exp. Med. 2015, 212(3):287-295.
    • (2015) J. Exp. Med. , vol.212 , Issue.3 , pp. 287-295
    • Jay, T.R.1
  • 26
    • 84925464993 scopus 로고    scopus 로고
    • TREM2 lipid sensing sustains the microglial response in an Alzheimer's disease model
    • Wang Y., et al. TREM2 lipid sensing sustains the microglial response in an Alzheimer's disease model. Cell 2015, 160(6):1061-1071.
    • (2015) Cell , vol.160 , Issue.6 , pp. 1061-1071
    • Wang, Y.1
  • 27
    • 84939654955 scopus 로고    scopus 로고
    • Rare coding mutations identified by sequencing of Alzheimer disease genome-wide association studies loci
    • Vardarajan B.N., et al. Rare coding mutations identified by sequencing of Alzheimer disease genome-wide association studies loci. Ann. Neurol. 2015, 78(3):487-498.
    • (2015) Ann. Neurol. , vol.78 , Issue.3 , pp. 487-498
    • Vardarajan, B.N.1
  • 28
    • 84900314611 scopus 로고    scopus 로고
    • CRISPR-Cas systems for editing, regulating and targeting genomes
    • Sander J.D., Joung J.K. CRISPR-Cas systems for editing, regulating and targeting genomes. Nat. Biotechnol. 2014, 32(4):347-355.
    • (2014) Nat. Biotechnol. , vol.32 , Issue.4 , pp. 347-355
    • Sander, J.D.1    Joung, J.K.2
  • 29
    • 84906079358 scopus 로고    scopus 로고
    • Permanent alteration of PCSK9 with in vivo CRISPR-Cas9 genome editing
    • Ding Q., et al. Permanent alteration of PCSK9 with in vivo CRISPR-Cas9 genome editing. Circ. Res. 2014, 115(5):488-492.
    • (2014) Circ. Res. , vol.115 , Issue.5 , pp. 488-492
    • Ding, Q.1
  • 30
    • 84916209395 scopus 로고    scopus 로고
    • Duplication of TBK1 stimulates autophagy in iPSC-derived retinal cells from a patient with normal tension glaucoma
    • Tucker B.A., et al. Duplication of TBK1 stimulates autophagy in iPSC-derived retinal cells from a patient with normal tension glaucoma. J. Stem Cell Res. Ther. 2014, 3(5):161.
    • (2014) J. Stem Cell Res. Ther. , vol.3 , Issue.5 , pp. 161
    • Tucker, B.A.1
  • 31
    • 84867335289 scopus 로고    scopus 로고
    • The diversity outbred mouse population
    • Churchill G.A., et al. The diversity outbred mouse population. Mamm. Genome 2012, 23(9-10):713-718.
    • (2012) Mamm. Genome , vol.23 , Issue.9-10 , pp. 713-718
    • Churchill, G.A.1
  • 32
    • 79959735506 scopus 로고    scopus 로고
    • Subspecific origin and haplotype diversity in the laboratory mouse
    • Yang H., et al. Subspecific origin and haplotype diversity in the laboratory mouse. Nat. Genet. 2011, 43(7):648-655.
    • (2011) Nat. Genet. , vol.43 , Issue.7 , pp. 648-655
    • Yang, H.1
  • 33
    • 84857410370 scopus 로고    scopus 로고
    • High-resolution genetic mapping using the mouse diversity outbred population
    • Svenson K.L., et al. High-resolution genetic mapping using the mouse diversity outbred population. Genetics 2012, 190(2):437-447.
    • (2012) Genetics , vol.190 , Issue.2 , pp. 437-447
    • Svenson, K.L.1
  • 34
    • 84875845939 scopus 로고    scopus 로고
    • Cerebrospinal fluid biomarkers for pathological processes in Alzheimer's disease
    • Rosen C., Zetterberg H. Cerebrospinal fluid biomarkers for pathological processes in Alzheimer's disease. Curr. Opin. Psychiatry 2013, 26(3):276-282.
    • (2013) Curr. Opin. Psychiatry , vol.26 , Issue.3 , pp. 276-282
    • Rosen, C.1    Zetterberg, H.2
  • 35
    • 84937758712 scopus 로고    scopus 로고
    • Amyloid biomarkers in Alzheimer's disease
    • Blennow K., et al. Amyloid biomarkers in Alzheimer's disease. Trends Pharmacol. Sci. 2015, 36(5):297-309.
    • (2015) Trends Pharmacol. Sci. , vol.36 , Issue.5 , pp. 297-309
    • Blennow, K.1
  • 36
    • 79951761390 scopus 로고    scopus 로고
    • The culprit behind amyloid beta peptide related neurotoxicity in Alzheimer's disease: oligomer size or conformation?
    • Broersen K., Rousseau Schymkowitz F.J. The culprit behind amyloid beta peptide related neurotoxicity in Alzheimer's disease: oligomer size or conformation?. Alzheimers Res. Ther. 2010, 2(4):12.
    • (2010) Alzheimers Res. Ther. , vol.2 , Issue.4 , pp. 12
    • Broersen, K.1    Rousseau Schymkowitz, F.J.2
  • 37
    • 78649503285 scopus 로고    scopus 로고
    • Preparation and characterization of neurotoxic tau oligomers
    • Lasagna-Reeves C.A., et al. Preparation and characterization of neurotoxic tau oligomers. Biochemistry 2010, 49(47):10039-10041.
    • (2010) Biochemistry , vol.49 , Issue.47 , pp. 10039-10041
    • Lasagna-Reeves, C.A.1
  • 38
    • 84877271133 scopus 로고    scopus 로고
    • Correlation of specific amyloid-beta oligomers with tau in cerebrospinal fluid from cognitively normal older adults
    • Handoko M., et al. Correlation of specific amyloid-beta oligomers with tau in cerebrospinal fluid from cognitively normal older adults. JAMA Neurol. 2013, 70(5):594-599.
    • (2013) JAMA Neurol. , vol.70 , Issue.5 , pp. 594-599
    • Handoko, M.1
  • 39
    • 84925845438 scopus 로고    scopus 로고
    • Tau immunotherapy modulates both pathological tau and upstream amyloid pathology in an Alzheimer's disease mouse model
    • Castillo-Carranza D.L., et al. Tau immunotherapy modulates both pathological tau and upstream amyloid pathology in an Alzheimer's disease mouse model. J. Neurosci. 2015, 35(12):4857-4868.
    • (2015) J. Neurosci. , vol.35 , Issue.12 , pp. 4857-4868
    • Castillo-Carranza, D.L.1
  • 40
    • 84883578547 scopus 로고    scopus 로고
    • Beta-Amyloid oligomers in aging and Alzheimer's disease
    • Zahs K.R., Ashe K.H. beta-Amyloid oligomers in aging and Alzheimer's disease. Front. Aging Neurosci. 2013, 5:28.
    • (2013) Front. Aging Neurosci. , vol.5 , pp. 28
    • Zahs, K.R.1    Ashe, K.H.2
  • 41
    • 84893494322 scopus 로고    scopus 로고
    • CB2 receptor deficiency increases amyloid pathology and alters tau processing in a transgenic mouse model of Alzheimer's disease
    • Koppel J., et al. CB2 receptor deficiency increases amyloid pathology and alters tau processing in a transgenic mouse model of Alzheimer's disease. Mol. Med. 2014, 20:29-36.
    • (2014) Mol. Med. , vol.20 , pp. 29-36
    • Koppel, J.1
  • 42
    • 84929951298 scopus 로고    scopus 로고
    • Synaptic contacts enhance cell-to-cell tau pathology propagation
    • Calafate S., et al. Synaptic contacts enhance cell-to-cell tau pathology propagation. Cell Rep 2015, 11(8):1176-1183.
    • (2015) Cell Rep , vol.11 , Issue.8 , pp. 1176-1183
    • Calafate, S.1
  • 43
    • 84865123660 scopus 로고    scopus 로고
    • A paravascular pathway facilitates CSF flow through the brain parenchyma and the clearance of interstitial solutes, including amyloid beta
    • 147ra111
    • Iliff J.J., et al. A paravascular pathway facilitates CSF flow through the brain parenchyma and the clearance of interstitial solutes, including amyloid beta. Sci. Transl Med. 2012, 4(147):147ra111.
    • (2012) Sci. Transl Med. , vol.4 , Issue.147
    • Iliff, J.J.1
  • 44
    • 84949316085 scopus 로고    scopus 로고
    • The glymphatic system: a beginner's guide
    • Jessen N.A., et al. The glymphatic system: a beginner's guide. Neurochem. Res. 2015, 40(12):2583-2599.
    • (2015) Neurochem. Res. , vol.40 , Issue.12 , pp. 2583-2599
    • Jessen, N.A.1
  • 45
    • 84885775321 scopus 로고    scopus 로고
    • Sleep drives metabolite clearance from the adult brain
    • Xie L., et al. Sleep drives metabolite clearance from the adult brain. Science 2013, 342(6156):373-377.
    • (2013) Science , vol.342 , Issue.6156 , pp. 373-377
    • Xie, L.1
  • 46
    • 84862908271 scopus 로고    scopus 로고
    • Effects of age and amyloid deposition on Abeta dynamics in the human central nervous system
    • Huang Y., et al. Effects of age and amyloid deposition on Abeta dynamics in the human central nervous system. Arch. Neurol. 2012, 69(1):51-58.
    • (2012) Arch. Neurol. , vol.69 , Issue.1 , pp. 51-58
    • Huang, Y.1
  • 47
    • 79951674720 scopus 로고    scopus 로고
    • Sleep and sleep disorders in older adults
    • Crowley K. Sleep and sleep disorders in older adults. Neuropsychol. Rev. 2011, 21(1):41-53.
    • (2011) Neuropsychol. Rev. , vol.21 , Issue.1 , pp. 41-53
    • Crowley, K.1
  • 48
    • 79960069372 scopus 로고    scopus 로고
    • Age-related changes in the cognitive function of sleep
    • Pace-Schott E.F., Spencer R.M. Age-related changes in the cognitive function of sleep. Prog. Brain Res. 2011, 191:75-89.
    • (2011) Prog. Brain Res. , vol.191 , pp. 75-89
    • Pace-Schott, E.F.1    Spencer, R.M.2
  • 49
    • 84933049267 scopus 로고    scopus 로고
    • Beta-Amyloid disrupts human NREM slow waves and related hippocampus-dependent memory consolidation
    • Mander B.A., et al. beta-Amyloid disrupts human NREM slow waves and related hippocampus-dependent memory consolidation. Nat. Neurosci. 2015, 18(7):1051-1057.
    • (2015) Nat. Neurosci. , vol.18 , Issue.7 , pp. 1051-1057
    • Mander, B.A.1
  • 50
    • 79953645895 scopus 로고    scopus 로고
    • Evidence for neuroinflammatory and microglial changes in the cerebral response to sleep loss
    • Wisor J.P., Schmidt Clegern M.A.W.C. Evidence for neuroinflammatory and microglial changes in the cerebral response to sleep loss. Sleep 2011, 34(3):261-272.
    • (2011) Sleep , vol.34 , Issue.3 , pp. 261-272
    • Wisor, J.P.1    Schmidt Clegern, M.A.W.C.2
  • 51
    • 84897954445 scopus 로고    scopus 로고
    • Microglial dysfunction in brain aging and Alzheimer's disease
    • Mosher K.I., Wyss-Coray T. Microglial dysfunction in brain aging and Alzheimer's disease. Biochem. Pharmacol. 2014, 88(4):594-604.
    • (2014) Biochem. Pharmacol. , vol.88 , Issue.4 , pp. 594-604
    • Mosher, K.I.1    Wyss-Coray, T.2
  • 52
    • 84939186864 scopus 로고    scopus 로고
    • Cannabinoid CB2 receptors in a mouse model of abeta amyloidosis: immunohistochemical analysis and suitability as a PET biomarker of neuroinflammation
    • Savonenko A.V., et al. Cannabinoid CB2 receptors in a mouse model of abeta amyloidosis: immunohistochemical analysis and suitability as a PET biomarker of neuroinflammation. PLoS One 2015, 10(6):e0129618.
    • (2015) PLoS One , vol.10 , Issue.6 , pp. e0129618
    • Savonenko, A.V.1
  • 53
    • 84870574017 scopus 로고    scopus 로고
    • CB2 receptor and amyloid pathology in frontal cortex of Alzheimer's disease patients
    • Solas M., et al. CB2 receptor and amyloid pathology in frontal cortex of Alzheimer's disease patients. Neurobiol. Aging 2013, 34(3):805-808.
    • (2013) Neurobiol. Aging , vol.34 , Issue.3 , pp. 805-808
    • Solas, M.1
  • 54
    • 65549141230 scopus 로고    scopus 로고
    • Gene expression as peripheral biomarkers for sporadic Alzheimer's disease
    • Grunblatt E., et al. Gene expression as peripheral biomarkers for sporadic Alzheimer's disease. J. Alzheimers Dis. 2009, 16(3):627-634.
    • (2009) J. Alzheimers Dis. , vol.16 , Issue.3 , pp. 627-634
    • Grunblatt, E.1
  • 55
    • 37849049077 scopus 로고    scopus 로고
    • Glial expression of cannabinoid CB(2) receptors and fatty acid amide hydrolase are beta amyloid-linked events in Down's syndrome
    • Nunez E., et al. Glial expression of cannabinoid CB(2) receptors and fatty acid amide hydrolase are beta amyloid-linked events in Down's syndrome. Neuroscience 2008, 151(1):104-110.
    • (2008) Neuroscience , vol.151 , Issue.1 , pp. 104-110
    • Nunez, E.1
  • 56
    • 80052582895 scopus 로고    scopus 로고
    • Enhanced dentate gyrus synaptic plasticity but reduced neurogenesis in a mouse model of amyloidosis
    • Poirier R., et al. Enhanced dentate gyrus synaptic plasticity but reduced neurogenesis in a mouse model of amyloidosis. Neurobiol. Dis 2010, 40(2):386-393.
    • (2010) Neurobiol. Dis , vol.40 , Issue.2 , pp. 386-393
    • Poirier, R.1
  • 57
    • 33747817727 scopus 로고    scopus 로고
    • Altered neurogenesis in Alzheimer's disease
    • Ziabreva I., et al. Altered neurogenesis in Alzheimer's disease. J. Psychosom. Res. 2006, 61(3):311-316.
    • (2006) J. Psychosom. Res. , vol.61 , Issue.3 , pp. 311-316
    • Ziabreva, I.1
  • 58
    • 79952579942 scopus 로고    scopus 로고
    • Neurodegenerative disease and adult neurogenesis
    • Winner B., Kohl Z., Gage F.H. Neurodegenerative disease and adult neurogenesis. Eur. J. Neurosci. 2011, 33(6):1139-1151.
    • (2011) Eur. J. Neurosci. , vol.33 , Issue.6 , pp. 1139-1151
    • Winner, B.1    Kohl, Z.2    Gage, F.H.3
  • 59
    • 77950461116 scopus 로고    scopus 로고
    • APP transgenic modeling of Alzheimer's disease: mechanisms of neurodegeneration and aberrant neurogenesis
    • Crews L., Rockenstein Masliah E.E. APP transgenic modeling of Alzheimer's disease: mechanisms of neurodegeneration and aberrant neurogenesis. Brain Struct. Funct. 2010, 214(2-3):111-126.
    • (2010) Brain Struct. Funct. , vol.214 , Issue.2-3 , pp. 111-126
    • Crews, L.1    Rockenstein Masliah, E.E.2
  • 60
    • 79955077994 scopus 로고    scopus 로고
    • Compensatory network changes in the dentate gyrus restore long-term potentiation following ablation of neurogenesis in young-adult mice
    • Singer B.H., et al. Compensatory network changes in the dentate gyrus restore long-term potentiation following ablation of neurogenesis in young-adult mice. Proc. Natl. Acad. Sci. U. S. A. 2011, 108(13):5437-5442.
    • (2011) Proc. Natl. Acad. Sci. U. S. A. , vol.108 , Issue.13 , pp. 5437-5442
    • Singer, B.H.1
  • 61
    • 84877285151 scopus 로고    scopus 로고
    • Role of neuroinflammation in adult neurogenesis and Alzheimer disease: therapeutic approaches
    • Fuster-Matanzo A., et al. Role of neuroinflammation in adult neurogenesis and Alzheimer disease: therapeutic approaches. Mediators Inflamm. 2013, 2013:260925.
    • (2013) Mediators Inflamm. , vol.2013 , pp. 260925
    • Fuster-Matanzo, A.1
  • 62
    • 79952751974 scopus 로고    scopus 로고
    • Complement receptor 2 is expressed in neural progenitor cells and regulates adult hippocampal neurogenesis
    • Moriyama M., et al. Complement receptor 2 is expressed in neural progenitor cells and regulates adult hippocampal neurogenesis. J. Neurosci. 2011, 31(11):3981-3989.
    • (2011) J. Neurosci. , vol.31 , Issue.11 , pp. 3981-3989
    • Moriyama, M.1
  • 63
    • 84925549946 scopus 로고    scopus 로고
    • Endocannabinoid signalling and the deteriorating brain
    • Di Marzo V., Stella N., Zimmer A. Endocannabinoid signalling and the deteriorating brain. Nat. Rev. Neurosci. 2015, 16(1):30-42.
    • (2015) Nat. Rev. Neurosci. , vol.16 , Issue.1 , pp. 30-42
    • Di Marzo, V.1    Stella, N.2    Zimmer, A.3
  • 64
    • 0035716136 scopus 로고    scopus 로고
    • ApoE deficiency compromises the blood brain barrier especially after injury
    • Methia N., et al. ApoE deficiency compromises the blood brain barrier especially after injury. Mol. Med. 2001, 7(12):810-815.
    • (2001) Mol. Med. , vol.7 , Issue.12 , pp. 810-815
    • Methia, N.1
  • 65
    • 84861418194 scopus 로고    scopus 로고
    • Apolipoprotein E controls cerebrovascular integrity via cyclophilin A
    • Bell R.D., et al. Apolipoprotein E controls cerebrovascular integrity via cyclophilin A. Nature 2012, 485(7399):512-516.
    • (2012) Nature , vol.485 , Issue.7399 , pp. 512-516
    • Bell, R.D.1
  • 66
    • 84946066133 scopus 로고    scopus 로고
    • APOE stabilization by exercise prevents aging neurovascular dysfunction and complement induction
    • Soto I., et al. APOE stabilization by exercise prevents aging neurovascular dysfunction and complement induction. PLoS Biol. 2015, 13(10).
    • (2015) PLoS Biol. , vol.13 , Issue.10
    • Soto, I.1
  • 67
    • 24744457932 scopus 로고    scopus 로고
    • Role of the MEOX2 homeobox gene in neurovascular dysfunction in Alzheimer disease
    • Wu Z., et al. Role of the MEOX2 homeobox gene in neurovascular dysfunction in Alzheimer disease. Nat. Med. 2005, 11(9):959-965.
    • (2005) Nat. Med. , vol.11 , Issue.9 , pp. 959-965
    • Wu, Z.1
  • 68
    • 84889872057 scopus 로고    scopus 로고
    • Development, maintenance and disruption of the blood-brain barrier
    • Obermeier B., Daneman R., Ransohoff R.M. Development, maintenance and disruption of the blood-brain barrier. Nat. Med. 2013, 19(12):1584-1596.
    • (2013) Nat. Med. , vol.19 , Issue.12 , pp. 1584-1596
    • Obermeier, B.1    Daneman, R.2    Ransohoff, R.M.3
  • 69
    • 84862264187 scopus 로고    scopus 로고
    • Transgenic APP expression during postnatal development causes persistent locomotor hyperactivity in the adult
    • Rodgers S.P., et al. Transgenic APP expression during postnatal development causes persistent locomotor hyperactivity in the adult. Mol. Neurodegener. 2012, 7:28.
    • (2012) Mol. Neurodegener. , vol.7 , pp. 28
    • Rodgers, S.P.1
  • 70
    • 84901029497 scopus 로고    scopus 로고
    • Using mice to model Alzheimer's dementia: an overview of the clinical disease and the preclinical behavioral changes in 10 mouse models
    • Webster S.J., et al. Using mice to model Alzheimer's dementia: an overview of the clinical disease and the preclinical behavioral changes in 10 mouse models. Front. Genet. 2014, 5:88.
    • (2014) Front. Genet. , vol.5 , pp. 88
    • Webster, S.J.1
  • 71
    • 80053113373 scopus 로고    scopus 로고
    • New translational assays for preclinical modelling of cognition in schizophrenia: the touchscreen testing method for mice and rats
    • Bussey T.J., et al. New translational assays for preclinical modelling of cognition in schizophrenia: the touchscreen testing method for mice and rats. Neuropharmacology 2012, 62(3):1191-1203.
    • (2012) Neuropharmacology , vol.62 , Issue.3 , pp. 1191-1203
    • Bussey, T.J.1
  • 72
    • 84875234023 scopus 로고    scopus 로고
    • Paying more attention to attention: towards more comprehensive cognitive translation using mouse models of Alzheimer's disease
    • Romberg C., Bussey T.J., Saksida L.M. Paying more attention to attention: towards more comprehensive cognitive translation using mouse models of Alzheimer's disease. Brain Res. Bull. 2013, 92:49-55.
    • (2013) Brain Res. Bull. , vol.92 , pp. 49-55
    • Romberg, C.1    Bussey, T.J.2    Saksida, L.M.3
  • 73
    • 79952260574 scopus 로고    scopus 로고
    • Impaired attention in the 3xTgAD mouse model of Alzheimer's disease: rescue by donepezil (Aricept)
    • Romberg C., et al. Impaired attention in the 3xTgAD mouse model of Alzheimer's disease: rescue by donepezil (Aricept). J. Neurosci. 2011, 31(9):3500-3507.
    • (2011) J. Neurosci. , vol.31 , Issue.9 , pp. 3500-3507
    • Romberg, C.1
  • 74
    • 0001856264 scopus 로고    scopus 로고
    • The fallacy of behavioral phenotyping without standardisation
    • van der Staay F.J., Steckler T. The fallacy of behavioral phenotyping without standardisation. Genes Brain Behav. 2002, 1(1):9-13.
    • (2002) Genes Brain Behav. , vol.1 , Issue.1 , pp. 9-13
    • van der Staay, F.J.1    Steckler, T.2
  • 75
    • 84926450571 scopus 로고    scopus 로고
    • Preclinical data reproducibility for R&D-the challenge for neuroscience
    • Steckler T. Preclinical data reproducibility for R&D-the challenge for neuroscience. Psychopharmacology (Berl.) 2015, 232(2):317-320.
    • (2015) Psychopharmacology (Berl.) , vol.232 , Issue.2 , pp. 317-320
    • Steckler, T.1
  • 76
    • 76849104977 scopus 로고    scopus 로고
    • Survey of the quality of experimental design, statistical analysis and reporting of research using animals
    • Kilkenny C., et al. Survey of the quality of experimental design, statistical analysis and reporting of research using animals. PLoS One 2009, 4(11):e7824.
    • (2009) PLoS One , vol.4 , Issue.11 , pp. e7824
    • Kilkenny, C.1
  • 77
    • 79958140708 scopus 로고    scopus 로고
    • Improving bioscience research reporting: the ARRIVE guidelines for reporting animal research
    • Kilkenny C., et al. Improving bioscience research reporting: the ARRIVE guidelines for reporting animal research. J. Pharmacol. Pharmacother. 2010, 1(2):94-99.
    • (2010) J. Pharmacol. Pharmacother. , vol.1 , Issue.2 , pp. 94-99
    • Kilkenny, C.1
  • 78
    • 84867302438 scopus 로고    scopus 로고
    • A call for transparent reporting to optimize the predictive value of preclinical research
    • Landis S.C., et al. A call for transparent reporting to optimize the predictive value of preclinical research. Nature 2012, 490(7419):187-191.
    • (2012) Nature , vol.490 , Issue.7419 , pp. 187-191
    • Landis, S.C.1
  • 79
    • 84912105043 scopus 로고    scopus 로고
    • Rigor or mortis: best practices for preclinical research in neuroscience
    • Steward O., Balice-Gordon R. Rigor or mortis: best practices for preclinical research in neuroscience. Neuron 2014, 84(3):572-581.
    • (2014) Neuron , vol.84 , Issue.3 , pp. 572-581
    • Steward, O.1    Balice-Gordon, R.2
  • 80
    • 34547737350 scopus 로고    scopus 로고
    • All is not well in the world of translational research
    • Unger E.F. All is not well in the world of translational research. J. Am. Coll. Cardiol. 2007, 50(8):738-740..
    • (2007) J. Am. Coll. Cardiol. , vol.50 , Issue.8 , pp. 738-740
    • Unger, E.F.1
  • 81
    • 84951832639 scopus 로고    scopus 로고
    • Mouse Models for Drug Discovery
    • ed. P.a. Wiles.
    • Sukoff Rizzo, S.J., 2015. Mouse Models for Drug Discovery, ed. P.a. Wiles.
    • (2015)
    • Sukoff Rizzo, S.J.1
  • 82
    • 84878190712 scopus 로고    scopus 로고
    • Future viable models of psychiatry drug discovery in pharma
    • Rizzo S.J., et al. Future viable models of psychiatry drug discovery in pharma. J. Biomol. Screen 2013, 18(5):509-521.
    • (2013) J. Biomol. Screen , vol.18 , Issue.5 , pp. 509-521
    • Rizzo, S.J.1


* 이 정보는 Elsevier사의 SCOPUS DB에서 KISTI가 분석하여 추출한 것입니다.