메뉴 건너뛰기




Volumn 16, Issue 11, 2015, Pages 26417-26451

Alzheimer’s disease: Mechanism and approach to cell therapy

Author keywords

Alzheimer s disease; Amyloid ; Mesenchymal stem cells; Neural stem cells; Tau

Indexed keywords

ALZHEIMER DISEASE; ASTROCYTE; CANCER IMMUNIZATION; CANCER IMMUNOTHERAPY; DOWNSTREAM PROCESSING; GENE MUTATION; GLUCOSE METABOLISM; HUMAN; MESENCHYMAL STEM CELL; MICROGLIA; NEURAL STEM CELL; NONHUMAN; REVIEW; TUMOR MICROENVIRONMENT; AGING; AMYLOID PLAQUE; ANIMAL; BIOLOGICAL THERAPY; CYTOLOGY; DISEASE MODEL; ENERGY METABOLISM; GENETICS; GLIA; IMMUNOLOGY; IMMUNOTHERAPY; METABOLISM; MUTATION; ONSET AGE; PATHOLOGY; PROCEDURES; STEM CELL; STEM CELL TRANSPLANTATION;

EID: 84946594802     PISSN: 16616596     EISSN: 14220067     Source Type: Journal    
DOI: 10.3390/ijms161125961     Document Type: Review
Times cited : (84)

References (355)
  • 1
    • 0030570313 scopus 로고    scopus 로고
    • Auguste D. And Alzheimer’s disease
    • [CrossRef] [PubMed]
    • O’Brien, C. Auguste D. and Alzheimer’s disease. Science 1996, 273, 28. [CrossRef] [PubMed]
    • (1996) Science , vol.273 , pp. 28
    • O’Brien, C.1
  • 2
    • 25144501662 scopus 로고    scopus 로고
    • Intraneuronal Aβ accumulation and origin of plaques in Alzheimer’s disease
    • [CrossRef] [PubMed]
    • Gouras, G.K.; Almeida, C.G.; Takahashi, R.H. Intraneuronal Aβ accumulation and origin of plaques in Alzheimer’s disease. Neurobiol. Aging 2005, 26, 1235-1244. [CrossRef] [PubMed]
    • (2005) Neurobiol. Aging , vol.26 , pp. 1235-1244
    • Gouras, G.K.1    Almeida, C.G.2    Takahashi, R.H.3
  • 3
    • 65249180827 scopus 로고    scopus 로고
    • Oskar Fischer and the study of dementia
    • [CrossRef] [PubMed]
    • Goedert, M. Oskar Fischer and the study of dementia. Brain 2009, 132, 1102-1111. [CrossRef] [PubMed]
    • (2009) Brain , vol.132 , pp. 1102-1111
    • Goedert, M.1
  • 4
    • 0030981585 scopus 로고    scopus 로고
    • Augaste D and Alzheimer’s disease
    • [CrossRef]
    • Maurer, K.; Volk, S.; Gerbaldo, H. Augaste D and Alzheimer’s disease. Lancet 1997, 349, 1546-1549. [CrossRef]
    • (1997) Lancet , vol.349 , pp. 1546-1549
    • Maurer, K.1    Volk, S.2    Gerbaldo, H.3
  • 6
    • 0032008710 scopus 로고    scopus 로고
    • Histopathology and APOE genotype of the first Alzheimer disease patient, Auguste D
    • [CrossRef] [PubMed]
    • Graeber, M.B.; Kosel, S.; Grasbon-Frodl, E.; Moller, H.J.; Mehraein, P. Histopathology and APOE genotype of the first Alzheimer disease patient, Auguste D. Neurogenetics 1998, 1, 223-228. [CrossRef] [PubMed]
    • (1998) Neurogenetics , vol.1 , pp. 223-228
    • Graeber, M.B.1    Kosel, S.2    Grasbon-Frodl, E.3    Moller, H.J.4    Mehraein, P.5
  • 7
    • 84872424224 scopus 로고    scopus 로고
    • A presenilin 1 mutation in the first case of Alzheimer’s disease
    • [CrossRef]
    • Muller, U.; Winter, P.; Graeber, M.B. A presenilin 1 mutation in the first case of Alzheimer’s disease. Lancet Neurol. 2013, 12, 129-130. [CrossRef]
    • (2013) Lancet Neurol , vol.12 , pp. 129-130
    • Muller, U.1    Winter, P.2    Graeber, M.B.3
  • 8
    • 84927175488 scopus 로고    scopus 로고
    • Presenilin 1 mutation in the first case of Alzheimer’s disease: Revised
    • [CrossRef] [PubMed]
    • Rupp, C.; Beyreuther, K.; Maurer, K.; Kins, S. A presenilin 1 mutation in the first case of Alzheimer’s disease: Revised. Alzheimers Dement. 2014, 10, 869-872. [CrossRef] [PubMed]
    • (2014) Alzheimers Dement , vol.10 , pp. 869-872
    • Rupp, C.1    Beyreuther, K.2    Maurer, K.3    Kins, S.A.4
  • 9
    • 84886600387 scopus 로고    scopus 로고
    • Therapeutics of Alzheimer’s disease: Past, present and future
    • [CrossRef] [PubMed]
    • Anand, R.; Gill, K.D.; Mahdi, A.A. Therapeutics of Alzheimer’s disease: Past, present and future. Neuropharmacology 2014, 76, 27-50. [CrossRef] [PubMed]
    • (2014) Neuropharmacology , vol.76 , pp. 27-50
    • Anand, R.1    Gill, K.D.2    Mahdi, A.A.3
  • 10
    • 84897954480 scopus 로고    scopus 로고
    • Future direction in Alzheimer’s disease from risk factors to prevention
    • [CrossRef] [PubMed]
    • Imtiaz, B.; Tolppanen, A.M.; Kivipelto, M.; Soininen, H. Future direction in Alzheimer’s disease from risk factors to prevention. Biochem. Pharmacol. 2014, 88, 661-670. [CrossRef] [PubMed]
    • (2014) Biochem. Pharmacol , vol.88 , pp. 661-670
    • Imtiaz, B.1    Tolppanen, A.M.2    Kivipelto, M.3    Soininen, H.4
  • 11
    • 84860015622 scopus 로고    scopus 로고
    • Alzheimer’s disease-linked mutations in presenilin-1 result in a drastic loss of activity in purified γ-secretase complexes
    • [CrossRef] [PubMed]
    • Cacquevel, M.; Aeschbach, L.; Houacine, J.; Fraering, P.C. Alzheimer’s disease-linked mutations in presenilin-1 result in a drastic loss of activity in purified γ-secretase complexes. PLoS ONE 2012, 7, e35133. [CrossRef] [PubMed]
    • (2012) Plos ONE , vol.7
    • Cacquevel, M.1    Aeschbach, L.2    Houacine, J.3    Fraering, P.C.4
  • 13
    • 16944362157 scopus 로고    scopus 로고
    • Mutant presenilins of Alzheimer’s disease increase production of 42-residue amyloid β-protein in both transfected cells and transgenic mice
    • [CrossRef] [PubMed]
    • Citron, M.; Westaway, D.; Xia, W.; Carlson, G.; Diehl, T.; Levesque, G.; Johnson-Wood, K.; Lee, M.; Seubert, P.; Davis, A.; et al. Mutant presenilins of Alzheimer’s disease increase production of 42-residue amyloid β-protein in both transfected cells and transgenic mice. Nat. Med. 1997, 3, 67-72. [CrossRef] [PubMed]
    • (1997) Nat. Med , vol.3 , pp. 67-72
    • Citron, M.1    Westaway, D.2    Xia, W.3    Carlson, G.4    Diehl, T.5    Levesque, G.6    Johnson-Wood, K.7    Lee, M.8    Seubert, P.9    Davis, A.10
  • 14
    • 0035066332 scopus 로고    scopus 로고
    • Alzheimer’s disease: Genes, proteins, and therapy
    • [CrossRef] [PubMed]
    • Selkoe, D.J. Alzheimer’s disease: Genes, proteins, and therapy. Physiol. Rev. 2001, 81, 741-766. [CrossRef] [PubMed]
    • (2001) Physiol. Rev , vol.81 , pp. 741-766
    • Selkoe, D.J.1
  • 15
    • 84871417031 scopus 로고    scopus 로고
    • Role of mitochondrial homeostasis and dynamics in Alzheimer’s disease
    • [CrossRef] [PubMed]
    • Selfridge, J.E.; Lezi, E.; Lu, J.; Swerdlow, R.H. Role of mitochondrial homeostasis and dynamics in Alzheimer’s disease. Neurobiol. Dis. 2013, 51, 3-12. [CrossRef] [PubMed]
    • (2013) Neurobiol. Dis , vol.51 , pp. 3-12
    • Selfridge, J.E.1    Lezi, E.2    Lu, J.3    Swerdlow, R.H.4
  • 16
    • 0029924266 scopus 로고    scopus 로고
    • Apolipoprotein E. Structure, function, and possible roles in Alzheimer’s disease
    • [CrossRef] [PubMed]
    • Mahley, R.W.; Nathan, B.P.; Pitas, R.E. Apolipoprotein E. Structure, function, and possible roles in Alzheimer’s disease. Ann. N. Y. Acad. Sci. 1996, 777, 139-145. [CrossRef] [PubMed]
    • (1996) Ann. N. Y. Acad. Sci , vol.777 , pp. 139-145
    • Mahley, R.W.1    Nathan, B.P.2    Pitas, R.E.3
  • 18
    • 1542409285 scopus 로고    scopus 로고
    • Functions of lipoprotein receptors in neurons
    • [CrossRef] [PubMed]
    • Beffert, U.; Stolt, P.C.; Herz, J. Functions of lipoprotein receptors in neurons. J. Lipid Res. 2004, 45, 403-409. [CrossRef] [PubMed]
    • (2004) J. Lipid Res , vol.45 , pp. 403-409
    • Beffert, U.1    Stolt, P.C.2    Herz, J.3
  • 19
    • 38349146483 scopus 로고    scopus 로고
    • Molecular mechanisms and therapeutic opportunities
    • [CrossRef] [PubMed]
    • Cedazo-Minguez, A. Apolipoprotein E and Alzheimer’s disease: Molecular mechanisms and therapeutic opportunities. J. Cell. Mol. Med. 2007, 11, 1227-1238. [CrossRef] [PubMed]
    • (2007) J. Cell. Mol. Med , vol.11 , pp. 1227-1238
    • Cedazo-Minguez, A.1
  • 20
    • 0030005627 scopus 로고    scopus 로고
    • Apolipoprotein E alleles as risk factors in Alzheimer’s disease
    • [CrossRef] [PubMed]
    • Roses, A.D. Apolipoprotein E alleles as risk factors in Alzheimer’s disease. Annu. Rev. Med. 1996, 47, 387-400[CrossRef] [PubMed]
    • (1996) Annu. Rev. Med , vol.47 , pp. 387-400
    • Roses, A.D.1
  • 21
    • 84859584667 scopus 로고    scopus 로고
    • The genetics of Alzheimer’s disease
    • [PubMed]
    • Bertram, L.; Tanzi, R.E. The genetics of Alzheimer’s disease. Prog. Mol. Biol. Transl. Sci. 2012, 107, 79-100. [PubMed]
    • (2012) Prog. Mol. Biol. Transl. Sci , vol.107 , pp. 79-100
    • Bertram, L.1    Tanzi, R.E.2
  • 22
    • 0031839545 scopus 로고
    • Risk estimates of dementia by apolipoprotein E genotypes from a population-based incidence study: The Rotterdam study
    • [CrossRef]
    • Slooter, A.J.; Cruts, M.; Kalmijn, S.; Hofman, A.; Breteler, M.M.; van Broeckhoven, C.; van Duijn, C.M. Risk estimates of dementia by apolipoprotein E genotypes from a population-based incidence study: The Rotterdam study. Arch. Neurol. 1988, 55, 964-968. [CrossRef]
    • (1988) Arch. Neurol , vol.55 , pp. 964-968
    • Slooter, A.J.1    Cruts, M.2    Kalmijn, S.3    Hofman, A.4    Breteler, M.M.5    Van Broeckhoven, C.6    Van Duijn, C.M.7
  • 26
    • 77954122221 scopus 로고    scopus 로고
    • Genome-wide association studies: The key to unlocking neurodegeneration?
    • [CrossRef] [PubMed]
    • Gandhi, S.; Wood, N.W. Genome-wide association studies: The key to unlocking neurodegeneration? Nat. Neurosci. 2010, 13, 789-794. [CrossRef] [PubMed]
    • (2010) Nat. Neurosci , vol.13 , pp. 789-794
    • Gandhi, S.1    Wood, N.W.2
  • 33
    • 84870584668 scopus 로고    scopus 로고
    • Expression of novel Alzheimer’s disease risk genes in control and Alzheimer’s disease brains
    • [CrossRef] [PubMed]
    • Karch, C.M.; Jeng, A.T.; Nowotny, P.; Cady, J.; Cruchaga, C.; Goate, A.M. Expression of novel Alzheimer’s disease risk genes in control and Alzheimer’s disease brains. PLoS ONE 2012, 7, e50976. [CrossRef] [PubMed]
    • (2012) Plos ONE , vol.7
    • Karch, C.M.1    Jeng, A.T.2    Nowotny, P.3    Cady, J.4    Cruchaga, C.5    Goate, A.M.6
  • 39
    • 77951060145 scopus 로고    scopus 로고
    • Proteases and proteolysis in Alzheimer disease: A multifactorial view on the disease process
    • [CrossRef] [PubMed]
    • De Strooper, B. Proteases and proteolysis in Alzheimer disease: A multifactorial view on the disease process. Physiol. Rev. 2010, 90, 465-494. [CrossRef] [PubMed]
    • (2010) Physiol. Rev , vol.90 , pp. 465-494
    • De Strooper, B.1
  • 41
    • 60549089207 scopus 로고    scopus 로고
    • APP binds DR6 to trigger axon pruning and neuron death via distinct caspases
    • [CrossRef] [PubMed]
    • Nikolaev, A.; McLaughlin, T.; O’Leary, D.D.; Tessier-Lavigne, M. APP binds DR6 to trigger axon pruning and neuron death via distinct caspases. Nature 2009, 457, 981-989. [CrossRef] [PubMed]
    • (2009) Nature , vol.457 , pp. 981-989
    • Nikolaev, A.1    McLaughlin, T.2    O’Leary, D.D.3    Tessier-Lavigne, M.4
  • 42
    • 0025899041 scopus 로고
    • Amyloid deposition as the central event in the aetiology of Alzheimer’s disease
    • [CrossRef]
    • Hardy, J.; Allsop, D. Amyloid deposition as the central event in the aetiology of Alzheimer’s disease. Trends Pharmacol. Sci. 1991, 12, 383-388. [CrossRef]
    • (1991) Trends Pharmacol. Sci , vol.12 , pp. 383-388
    • Hardy, J.1    Allsop, D.2
  • 43
    • 0025753852 scopus 로고
    • The molecular pathology of Alzheimer’s disease
    • [CrossRef]
    • Selkoe, D.J. The molecular pathology of Alzheimer’s disease. Neuron 1991, 6, 487-498. [CrossRef]
    • (1991) Neuron , vol.6 , pp. 487-498
    • Selkoe, D.J.1
  • 44
    • 0026597063 scopus 로고
    • Alzheimer’s disease: The amyloid cascade hypothesis
    • [CrossRef] [PubMed]
    • Hardy, J.A. Higgins, G.A. Alzheimer’s disease: The amyloid cascade hypothesis. Science 1992, 256, 184-185. [CrossRef] [PubMed]
    • (1992) Science , vol.256 , pp. 184-185
    • Hardy, J.A.1    Higgins, G.A.2
  • 45
    • 0037135111 scopus 로고    scopus 로고
    • The amyloid hypothesis of Alzheimer’s disease: Progress and problems on the road to therapeutics
    • [CrossRef] [PubMed]
    • Hardy, J.; Selkoe, D.J. The amyloid hypothesis of Alzheimer’s disease: Progress and problems on the road to therapeutics. Science 2002, 297, 353-356. [CrossRef] [PubMed]
    • (2002) Science , vol.297 , pp. 353-356
    • Hardy, J.1    Selkoe, D.J.2
  • 47
    • 0038117796 scopus 로고    scopus 로고
    • Correlation between elevated levels of amyloid ß-peptide in the brain and cognitive decline
    • [CrossRef] [PubMed]
    • Naslund, J.; Haroutunian, V.; Mohs, R.; Davis, K.L.; Davies, P.; Greengard, P.; Buxbaum, J.D. Correlation between elevated levels of amyloid ß-peptide in the brain and cognitive decline. JAMA 2000, 283, 1571-1577. [CrossRef] [PubMed]
    • (2000) JAMA , vol.283 , pp. 1571-1577
    • Naslund, J.1    Haroutunian, V.2    Mohs, R.3    Davis, K.L.4    Davies, P.5    Greengard, P.6    Buxbaum, J.D.7
  • 48
  • 49
    • 0037041426 scopus 로고    scopus 로고
    • Naturally secreted oligomers of amyloid ß protein potently inhibit hippocampal long-term potentiation in vivo
    • [CrossRef] [PubMed]
    • Walsh, D.M.; Klyubin, I.; Fadeeva, J.V.; Cullen, W.K.; Anwyl, R.; Wolfe, M.S.; Rowan, M.J.; Selkoe, D.J. Naturally secreted oligomers of amyloid ß protein potently inhibit hippocampal long-term potentiation in vivo. Nature 2002, 416, 535-539. [CrossRef] [PubMed]
    • (2002) Nature , vol.416 , pp. 535-539
    • Walsh, D.M.1    Klyubin, I.2    Fadeeva, J.V.3    Cullen, W.K.4    Anwyl, R.5    Wolfe, M.S.6    Rowan, M.J.7    Selkoe, D.J.8
  • 51
    • 22144462135 scopus 로고    scopus 로고
    • Neurotoxic protein oligomers—What you see is not always what you get
    • Bitan, G.; Fradinger, E.A.; Spring, S.M.; Teplow, D.B. Neurotoxic protein oligomers—What you see is not always what you get. Amyloid 2005, 12, 88-95.
    • (2005) Amyloid , vol.12 , pp. 88-95
    • Bitan, G.1    Fradinger, E.A.2    Spring, S.M.3    Teplow, D.B.4
  • 53
    • 0035947044 scopus 로고    scopus 로고
    • Role of insulin and insulin receptor in learning and memory
    • [CrossRef]
    • Zhao, W.Q. Alkon, D.L. Role of insulin and insulin receptor in learning and memory. Mol. Cell. Endocrinol.2001, 177, 125-134. [CrossRef]
    • (2001) Mol. Cell. Endocrinol , vol.177 , pp. 125-134
    • Zhao, W.Q.1    Alkon, D.L.2
  • 54
    • 79956302348 scopus 로고    scopus 로고
    • Alzheimer’s disease brain-derived amyloid-ß-mediated inhibition of LTP in vivo is prevented by immunotargeting cellular prion protein
    • [CrossRef] [PubMed]
    • Barry, A.E.; Klyubin, I.; McDonald, J.M.; Mably, A.J.; Farrell, M.A.; Scott, M.; Walsh, D.M.; Rowan, M.J. Alzheimer’s disease brain-derived amyloid-ß-mediated inhibition of LTP in vivo is prevented by immunotargeting cellular prion protein. J. Neurosci. 2011, 31, 7259-7263. [CrossRef] [PubMed]
    • (2011) J. Neurosci , vol.31 , pp. 7259-7263
    • Barry, A.E.1    Klyubin, I.2    McDonald, J.M.3    Mably, A.J.4    Farrell, M.A.5    Scott, M.6    Walsh, D.M.7    Rowan, M.J.8
  • 56
    • 79955044494 scopus 로고    scopus 로고
    • Oluble amyloid ß-protein dimers isolated from Alzheimer cortex directly induce Tau hyperphosphorylation and neuritic degeneration
    • [CrossRef] [PubMed]
    • Jin, M.; Shepardson, N.; Yang, T.; Chen, G.; Walsh, D.; Selkoe, D.J. Soluble amyloid ß-protein dimers isolated from Alzheimer cortex directly induce Tau hyperphosphorylation and neuritic degeneration. Proc. Natl. Acad. Sci. USA 2011, 108, 5819-5824. [CrossRef] [PubMed]
    • (2011) Proc. Natl. Acad. Sci. USA , vol.108 , pp. 5819-5824
    • Jin, M.1    Shepardson, N.2    Yang, T.3    Chen, G.4    Walsh, D.5    Selkoe, D.J.6
  • 57
    • 84872559990 scopus 로고    scopus 로고
    • Molecular mechanisms of amyloid oligomers toxicity
    • [PubMed]
    • Kayed, R.; Lasagna-Reeves, C.A. Molecular mechanisms of amyloid oligomers toxicity. J. Alzheimers Dis.2013, 33, S67-S78. [PubMed]
    • (2013) J. Alzheimers Dis , vol.33 , pp. S67-S78
    • Kayed, R.1    Lasagna-Reeves, C.A.2
  • 58
    • 58149463424 scopus 로고    scopus 로고
    • MTOR-dependent signalling in Alzheimer’s disease
    • [CrossRef] [PubMed]
    • Pei, J.J.; Hugon, J. mTOR-dependent signalling in Alzheimer’s disease. J. Cell. Mol. Med. 2008, 12, 2525-2532. [CrossRef] [PubMed]
    • (2008) J. Cell. Mol. Med , vol.12 , pp. 2525-2532
    • Pei, J.J.1    Hugon, J.2
  • 59
    • 60549112186 scopus 로고    scopus 로고
    • Immunization in Alzheimer’s disease: Naive hope or realistic clinical potential?
    • [CrossRef] [PubMed]
    • Foster, J.K.; Verdile, G.; Bates, K.A.; Martins, R.N. Immunization in Alzheimer’s disease: Naive hope or realistic clinical potential? Mol. Psychiatry 2009, 14, 239-251. [CrossRef] [PubMed]
    • (2009) Mol. Psychiatry , vol.14 , pp. 239-251
    • Foster, J.K.1    Verdile, G.2    Bates, K.A.3    Martins, R.N.4
  • 61
    • 84904204899 scopus 로고    scopus 로고
    • Targeting the proper amyloid-ß neuronal toxins: A path forward for Alzheimer’s disease immunotherapeutics
    • [CrossRef] [PubMed]
    • Goure, W.F.; Krafft, G.A.; Jerecic, J.; Hefti, F. Targeting the proper amyloid-ß neuronal toxins: A path forward for Alzheimer’s disease immunotherapeutics. Alzheimers Res. Ther. 2014, 6, 42. [CrossRef] [PubMed]
    • (2014) Alzheimers Res. Ther , vol.6 , pp. 42
    • Goure, W.F.1    Krafft, G.A.2    Jerecic, J.3    Hefti, F.4
  • 62
    • 84892748542 scopus 로고    scopus 로고
    • Alzheimer’s Disease Cooperative Study Steering Committee, Solanezumab Study Group. Phase 3 trials of solanezumab for mild-to-moderate Alzheimer’s disease
    • [CrossRef] [PubMed]
    • Doody, R.S.; Thomas, R.G.; Farlow, M.; Iwatsubo, T.; Vellas, B.; Joffe, S.; Kieburtz, K.; Raman, R.; Sun, X.; Aisen, P.S.; et al. Alzheimer’s Disease Cooperative Study Steering Committee, Solanezumab Study Group. Phase 3 trials of solanezumab for mild-to-moderate Alzheimer’s disease. N. Engl. J. Med. 2014, 370, 311-321. [CrossRef] [PubMed]
    • (2014) N. Engl. J. Med , vol.370 , pp. 311-321
    • Doody, R.S.1    Thomas, R.G.2    Farlow, M.3    Iwatsubo, T.4    Vellas, B.5    Joffe, S.6    Kieburtz, K.7    Raman, R.8    Sun, X.9    Aisen, P.S.10
  • 64
    • 84897954317 scopus 로고    scopus 로고
    • Immunotherapy for Alzheimer’s disease
    • [CrossRef] [PubMed]
    • Wisniewski, T.; Goni, F. Immunotherapy for Alzheimer’s disease. Biochem. Pharmacol. 2014, 88, 499-507. [CrossRef] [PubMed]
    • (2014) Biochem. Pharmacol , vol.88 , pp. 499-507
    • Wisniewski, T.1    Goni, F.2
  • 66
    • 0021338217 scopus 로고
    • Phosphorylation affects the ability of Tau protein to promote microtubule assembly
    • [PubMed]
    • Lindwall, G.; Cole, R.D. Phosphorylation affects the ability of Tau protein to promote microtubule assembly. J. Biol. Chem. 1984, 259, 5301-5305. [PubMed]
    • (1984) J. Biol. Chem , vol.259 , pp. 5301-5305
    • Lindwall, G.1    Cole, R.D.2
  • 67
    • 0025098891 scopus 로고
    • Inhibition of neurite polarity by Tau antisense oligonucleotides in primary cerebellar neurons
    • [CrossRef] [PubMed]
    • Caceres, A.; Kosik, K.S. Inhibition of neurite polarity by Tau antisense oligonucleotides in primary cerebellar neurons. Nature 1990, 343, 461-463. [CrossRef] [PubMed]
    • (1990) Nature , vol.343 , pp. 461-463
    • Caceres, A.1    Kosik, K.S.2
  • 68
    • 0034940160 scopus 로고    scopus 로고
    • Neurodegenerative tauopathies. Annu
    • [CrossRef] [PubMed]
    • Lee, V.M.; Goedert, M.; Trojanowski, J.Q. Neurodegenerative tauopathies. Annu. Rev. Neurosci. 2001, 24,1121-1159. [CrossRef] [PubMed]
    • (2001) Rev. Neurosci , vol.24 , pp. 1121-1159
    • Lee, V.M.1    Goedert, M.2    Trojanowski, J.Q.3
  • 70
    • 0035140975 scopus 로고    scopus 로고
    • Going new places using an old MAP: Tau, microtubules and humanneurodegenerative disease
    • [CrossRef]
    • Garcia, M.L.; Cleveland, D.W. Going new places using an old MAP: Tau, microtubules and humanneurodegenerative disease. Curr. Opin. Cell Biol. 2001, 13, 41-48. [CrossRef]
    • (2001) Curr. Opin. Cell Biol , vol.13 , pp. 41-48
    • Garcia, M.L.1    Cleveland, D.W.2
  • 74
    • 34548036227 scopus 로고    scopus 로고
    • Tau-mediated neurodegeneration in Alzheimer’s disease andrelated disorders
    • [CrossRef] [PubMed]
    • Ballatore, C.; Lee, V.M.; Trojanowski, J.Q. Tau-mediated neurodegeneration in Alzheimer’s disease andrelated disorders. Nat. Rev. Neurosci. 2007, 8, 663-672. [CrossRef] [PubMed]
    • (2007) Nat. Rev. Neurosci , vol.8 , pp. 663-672
    • Ballatore, C.1    Lee, V.M.2    Trojanowski, J.Q.3
  • 76
    • 10944223484 scopus 로고    scopus 로고
    • Tau protein as a differential biomarker of tauopathies
    • [CrossRef] [PubMed]
    • Sergeant, N.; Delacourte, A.; Buee, L. Tau protein as a differential biomarker of tauopathies.Biochim. Biophys. Acta 2005, 1739, 179-197. [CrossRef] [PubMed]
    • (2005) Biochim. Biophys. Acta , vol.1739 , pp. 179-197
    • Sergeant, N.1    Delacourte, A.2    Buee, L.3
  • 77
    • 0028073692 scopus 로고
    • Localization of disinhibition-dementia-parkinsonism-amyotrophy complex to 17q2-22
    • [PubMed]
    • Wilhelmsen, K.C.; Lynch, T.; Pavlou, E.; Higgins, M.; Nygaard, T.G. Localization of disinhibition-dementia-parkinsonism-amyotrophy complex to 17q2-22. Am. J. Hum. Genet. 1994, 55, 1159-1165. [PubMed]
    • (1994) Am. J. Hum. Genet , vol.55 , pp. 1159-1165
    • Wilhelmsen, K.C.1    Lynch, T.2    Pavlou, E.3    Higgins, M.4    Nygaard, T.G.5
  • 78
    • 26444581827 scopus 로고    scopus 로고
    • Tau gene mutations and their effects
    • Goedert, M. Tau gene mutations and their effects. Mov. Disord. 2005, 12, 45-52.
    • (2005) Mov. Disord , vol.12 , pp. 45-52
    • Goedert, M.1
  • 79
    • 56349119351 scopus 로고    scopus 로고
    • Linking Aß and Tau in late-onset Alzheimer’s disease: A dual pathway hypothesis
    • [CrossRef] [PubMed]
    • Small, S.A.; Duff, K. Linking Aß and Tau in late-onset Alzheimer’s disease: A dual pathway hypothesis.Neuron 2008, 60, 534-542. [CrossRef] [PubMed]
    • (2008) Neuron , vol.60 , pp. 534-542
    • Small, S.A.1    Duff, K.2
  • 80
    • 84897954670 scopus 로고    scopus 로고
    • The complexities of the pathology—pathogenesis relationship in Alzheimerdisease
    • [CrossRef] [PubMed]
    • Castellani, R.J.; Perry, G. The complexities of the pathology—pathogenesis relationship in Alzheimerdisease. Biochem. Pharmacol. 2014, 88, 671-676. [CrossRef] [PubMed]
    • (2014) Biochem. Pharmacol , vol.88 , pp. 671-676
    • Castellani, R.J.1    Perry, G.2
  • 81
    • 47149112621 scopus 로고    scopus 로고
    • Long-term effects of Aß42 immunisation in Alzheimer’s disease: Follow-up of a randomised, placebo-controlled phase I trial
    • [CrossRef]
    • Holmes, C.; Boche, D.; Wilkinson, D.; Yadegarfar, G.; Hopkins, V.; Bayer, A.; Jones, R.W.; Bullock, R.;Love, S.; Neal, J.W.; et al. Long-term effects of Aß42 immunisation in Alzheimer’s disease: Follow-up of a randomised, placebo-controlled phase I trial. Lancet 2008, 372, 216-223. [CrossRef]
    • (2008) Lancet , vol.372 , pp. 216-223
    • Holmes, C.1    Boche, D.2    Wilkinson, D.3    Yadegarfar, G.4    Hopkins, V.5    Bayer, A.6    Jones, R.W.7    Bullock, R.8    Love, S.9    Neal, J.W.10
  • 82
    • 33749614555 scopus 로고    scopus 로고
    • Tauopathy-like abnormalities and neurologic deficits in mice immunized with neuronal Tau protein
    • [CrossRef] [PubMed]
    • Rosenmann, H.; Grigoriadis, N.; Karussis, D.; Boimel, M.; Touloumi, O.; Ovadia, H.; Abramsky, O. Tauopathy-like abnormalities and neurologic deficits in mice immunized with neuronal Tau protein. Arch. Neurol. 2006, 63, 1459-1467. [CrossRef] [PubMed]
    • (2006) Arch. Neurol , vol.63 , pp. 1459-1467
    • Rosenmann, H.1    Grigoriadis, N.2    Karussis, D.3    Boimel, M.4    Touloumi, O.5    Ovadia, H.6    Abramsky, O.7
  • 83
    • 79960563632 scopus 로고    scopus 로고
    • Passive immunization targeting pathologicalphospho-Tau protein in a mouse model reduces functional decline and clears Tau aggregates from the brain
    • [CrossRef] [PubMed]
    • Boutajangout, A.; Ingadottir, J.; Davies, P.; Sigurdsson, E.M. Passive immunization targeting pathologicalphospho-Tau protein in a mouse model reduces functional decline and clears Tau aggregates from the brain. J. Neurochem. 2011, 118, 658-667. [CrossRef] [PubMed]
    • (2011) J. Neurochem , vol.118 , pp. 658-667
    • Boutajangout, A.1    Ingadottir, J.2    Davies, P.3    Sigurdsson, E.M.4
  • 84
    • 80053202160 scopus 로고    scopus 로고
    • Passive immunization with anti-Tau antibodies in two transgenic models: Reduction of Tau pathology and delay of disease progression
    • [CrossRef] [PubMed]
    • Chai, X.; Wu, S.; Murray, T.K.; Kinley, R.; Cella, C.V.; Sims, H.; Buckner, N.; Hanmer, J.; Davies, P.;O’Neill, M.J.; et al. Passive immunization with anti-Tau antibodies in two transgenic models: Reduction of Tau pathology and delay of disease progression. J. Biol. Chem. 2011, 286, 34457-34467. [CrossRef] [PubMed]
    • (2011) J. Biol. Chem , vol.286 , pp. 34457-34467
    • Chai, X.1    Wu, S.2    Murray, T.K.3    Kinley, R.4    Cella, C.V.5    Sims, H.6    Buckner, N.7    Hanmer, J.8    Davies, P.9    O’Neill, M.J.10
  • 85
    • 84897954162 scopus 로고    scopus 로고
    • The role of amyloid-ß in the regulation of memory
    • [CrossRef] [PubMed]
    • Morley, J.E.; Farr, S.A. The role of amyloid-ß in the regulation of memory. Biochem. Pharmacol. 2014, 88, 479-485. [CrossRef] [PubMed]
    • (2014) Biochem , vol.88 , pp. 479-485
    • Morley, J.E.1    Farr, S.A.2
  • 86
    • 84878956537 scopus 로고    scopus 로고
    • Physiological functions of APP family proteins
    • [CrossRef] [PubMed]
    • Muller, U.C.; Zheng, H. Physiological functions of APP family proteins. Cold Spring Harb. Perspect. Med. 2012, 2. [CrossRef] [PubMed]
    • (2012) Cold Spring Harb. Perspect. Med , pp. 2
    • Muller, U.C.1    Zheng, H.2
  • 88
    • 84863651655 scopus 로고    scopus 로고
    • Aging as an event of proteostasis collapse
    • [CrossRef] [PubMed]
    • Taylor, R.C.; Dillin, A. Aging as an event of proteostasis collapse. Cold Spring Harb. Perspect. Biol. 2011, 3. [CrossRef] [PubMed]
    • (2011) Cold Spring Harb. Perspect. Biol , pp. 3
    • Taylor, R.C.1    Dillin, A.2
  • 89
    • 22244446505 scopus 로고    scopus 로고
    • The mammalian unfolded protein response
    • [CrossRef] [PubMed]
    • Schroder, M.; Kaufman, R.J. The mammalian unfolded protein response. Annu. Rev. Biochem. 2005, 74, 739-789. [CrossRef] [PubMed]
    • (2005) Annu. Rev. Biochem , vol.74 , pp. 739-789
    • Schroder, M.1    Kaufman, R.J.2
  • 91
    • 74549169836 scopus 로고    scopus 로고
    • Stress in Alzheimer’s disease: A novel neuronal trigger for inflammation and Alzheimer’s pathology
    • [CrossRef] [PubMed]
    • Salminen, A.; Kauppinen, A.; Suuronen, T.; Kaarniranta, K.; Ojala, J. ER stress in Alzheimer’s disease: A novel neuronal trigger for inflammation and Alzheimer’s pathology. J. Neuroinflamm. 2009, 6, 41. [CrossRef] [PubMed]
    • (2009) J. Neuroinflamm , vol.6 , pp. 41
    • Salminen, A.1    Kauppinen, A.2    Suuronen, T.3    Kaarniranta, K.4    Ojala, J.5
  • 92
    • 70349905357 scopus 로고    scopus 로고
    • Role of ERO1-a-mediated stimulation of inositol 1,4,5-tirphoate receptor activity in endoplasmic reticulum stress-induced apoptosis
    • [CrossRef] [PubMed]
    • Li, G.; Mongillo, M.; Chin, K.T.; Harding, H.; Ron, D.; Marks, A.R.; Tabas, I. Role of ERO1-a-mediated stimulation of inositol 1,4,5-tirphoate receptor activity in endoplasmic reticulum stress-induced apoptosis. J. Cell Biol. 2009, 186, 783-792. [CrossRef] [PubMed]
    • (2009) J. Cell Biol , vol.186 , pp. 783-792
    • Li, G.1    Mongillo, M.2    Chin, K.T.3    Harding, H.4    Ron, D.5    Marks, A.R.6    Tabas, I.7
  • 93
    • 0034723235 scopus 로고    scopus 로고
    • Coupling of stress in the ER to activation of JNK protein kinases by transmembrane protein kinase IRE1
    • [CrossRef] [PubMed]
    • Urano, F.; Wang, X.; Bertolotti, A.; Zhang, Y.; Chung, P.; Harding, H.P.; Ron, D. Coupling of stress in the ER to activation of JNK protein kinases by transmembrane protein kinase IRE1. Science 2000, 287, 664-666. [CrossRef] [PubMed]
    • (2000) Science , vol.287 , pp. 664-666
    • Urano, F.1    Wang, X.2    Bertolotti, A.3    Zhang, Y.4    Chung, P.5    Harding, H.P.6    Ron, D.7
  • 94
    • 33646190885 scopus 로고    scopus 로고
    • The ASK1-MAP kinase signaling in ER stress and neurodegenerative diseases
    • [CrossRef] [PubMed]
    • Sekine, Y.; Takeda, K.; Ichijo, H. The ASK1-MAP kinase signaling in ER stress and neurodegenerative diseases. Curr. Mol. Med. 2006, 6, 87-97. [CrossRef] [PubMed]
    • (2006) Curr. Mol. Med , vol.6 , pp. 87-97
    • Sekine, Y.1    Takeda, K.2    Ichijo, H.3
  • 95
    • 36448936747 scopus 로고    scopus 로고
    • Kinase activities increase during the development of tauopathy in htau mice
    • [CrossRef] [PubMed]
    • Kelleher, I.; Garwood, C.; Hanger, D.P.; Anderton, B.H.; Noble, W. Kinase activities increase during the development of tauopathy in htau mice. J. Neurochem. 2007, 103, 2256-2267. [CrossRef] [PubMed]
    • (2007) J. Neurochem , vol.103 , pp. 2256-2267
    • Kelleher, I.1    Garwood, C.2    Hanger, D.P.3    Anderton, B.H.4    Noble, W.5
  • 97
    • 84890085638 scopus 로고    scopus 로고
    • Role of protein misfolding and proteostasis deficiency in protein misfolding diseases and aging
    • [CrossRef] [PubMed]
    • Cuanalo-Contreras, K.; Mukherjee, A.; Soto, C. Role of protein misfolding and proteostasis deficiency in protein misfolding diseases and aging. Int. J. Cell Biol. 2013, 2013. [CrossRef] [PubMed]
    • (2013) Int. J. Cell Biol , pp. 2013
    • Cuanalo-Contreras, K.1    Mukherjee, A.2    Soto, C.3
  • 98
    • 67650724069 scopus 로고    scopus 로고
    • Regulation and function of NF-kappaB transcription factors in the immune system
    • [CrossRef] [PubMed]
    • Vallabhapurapu, S.; Karin, M. Regulation and function of NF-kappaB transcription factors in the immune system. Annu. Rev. Immunol. 2009, 27, 693-733. [CrossRef] [PubMed]
    • (2009) Annu. Rev. Immunol , vol.27 , pp. 693-733
    • Vallabhapurapu, S.1    Karin, M.2
  • 99
    • 33747347875 scopus 로고    scopus 로고
    • Transcriptional and translational regulation of BACE1 expression—Implications for Alzheimer’s disease
    • [CrossRef] [PubMed]
    • Rossner, S.; Sastre, M.; Bourne, K.; Lichtenthaler, S.F. Transcriptional and translational regulation of BACE1 expression—Implications for Alzheimer’s disease. Prog. Neurobiol. 2006, 79, 95-111. [CrossRef] [PubMed]
    • (2006) Prog. Neurobiol , vol.79 , pp. 95-111
    • Rossner, S.1    Sastre, M.2    Bourne, K.3    Lichtenthaler, S.F.4
  • 101
    • 84868115008 scopus 로고    scopus 로고
    • Protein homeostasis, aging and Alzheimer’s disease
    • [CrossRef] [PubMed]
    • Morawe, T.; Hiebel, C.; Kern, A.; Behl, C. Protein homeostasis, aging and Alzheimer’s disease. Mol. Neurobiol. 2012, 46, 41-54. [CrossRef] [PubMed]
    • (2012) Mol. Neurobiol , vol.46 , pp. 41-54
    • Morawe, T.1    Hiebel, C.2    Kern, A.3    Behl, C.4
  • 102
    • 84863337843 scopus 로고    scopus 로고
    • Alzheimer mechanisms and therapeutic strategies
    • [CrossRef] [PubMed]
    • Huang, Y.; Mucke, L. Alzheimer mechanisms and therapeutic strategies. Cell 2012, 148, 1204-1222. [CrossRef] [PubMed]
    • (2012) Cell , vol.148 , pp. 1204-1222
    • Huang, Y.1    Mucke, L.2
  • 105
    • 36248968668 scopus 로고    scopus 로고
    • Heat shock proteins and amateur chaperones in amyloid-ß accumulation and clearance in Alzheimer’s disease
    • [CrossRef] [PubMed]
    • Wilhelmus, M.M.; de Waal, R.M.; Verbeek, M.M. Heat shock proteins and amateur chaperones in amyloid-ß accumulation and clearance in Alzheimer’s disease. Mol. Neurobiol. 2007, 35, 203-216. [CrossRef] [PubMed]
    • (2007) Mol. Neurobiol , vol.35 , pp. 203-216
    • Wilhelmus, M.M.1    De Waal, R.M.2    Verbeek, M.M.3
  • 109
    • 77953269718 scopus 로고    scopus 로고
    • Brain cholesterol in normal and pathological aging
    • [CrossRef] [PubMed]
    • Martin, M.; Dotti, C.G.; Ledesma, M.D. Brain cholesterol in normal and pathological aging. Biochim. Biophys. Acta 2010, 1801, 934-944. [CrossRef] [PubMed]
    • (2010) Biochim. Biophys. Acta , vol.1801 , pp. 934-944
    • Martin, M.1    Dotti, C.G.2    Ledesma, M.D.3
  • 110
    • 84890987858 scopus 로고    scopus 로고
    • What are lipoproteins doing in the brain?
    • [CrossRef] [PubMed]
    • Wang, H.; Eckel, R.H. What are lipoproteins doing in the brain? Trends Endocrinol. Metab. 2014, 25, 8-14. [CrossRef] [PubMed]
    • (2014) Trends Endocrinol. Metab , vol.25 , pp. 8-14
    • Wang, H.1    Eckel, R.H.2
  • 112
    • 79952176116 scopus 로고    scopus 로고
    • The pathological roles of ganglioside metabolism in Alzheimer’s disease: Effects of gangliosides on neurogenesis
    • [CrossRef] [PubMed]
    • Ariga, T.; Wakade, C.; Yu, R.K. The pathological roles of ganglioside metabolism in Alzheimer’s disease: Effects of gangliosides on neurogenesis. Int. J. Alzheimers Dis. 2011, 2011, 193618. [CrossRef] [PubMed]
    • (2011) Int. J. Alzheimers Dis
    • Ariga, T.1    Wakade, C.2    Yu, R.K.3
  • 113
    • 0035816709 scopus 로고    scopus 로고
    • Cholesterol-dependent formation on GM1 ganglioside-bound amyloid ß-protein, an endogenous seed for Alzheimer amyloid
    • [CrossRef] [PubMed]
    • Kakio, A.; Nishimoto, S.I.; Yanagisawa, K.; Kazutsumi, Y.; Matsuzaki, K. Cholesterol-dependent formation on GM1 ganglioside-bound amyloid ß-protein, an endogenous seed for Alzheimer amyloid. J. Biol. Chem. 2001, 276, 24985-24990. [CrossRef] [PubMed]
    • (2001) J. Biol. Chem , vol.276 , pp. 24985-24990
    • Kakio, A.1    Nishimoto, S.I.2    Yanagisawa, K.3    Kazutsumi, Y.4    Matsuzaki, K.5
  • 114
    • 84884538771 scopus 로고    scopus 로고
    • Cholesterol accelerates the binding of Alzheimer’s ß-amyloid peptide to ganglioside GM1 through a universal hydrogen-bond-dependent sterol tuning of glycolipid conformation
    • [CrossRef] [PubMed]
    • Fantini, J.; Yahi, N.; Garmy, N. Cholesterol accelerates the binding of Alzheimer’s ß-amyloid peptide to ganglioside GM1 through a universal hydrogen-bond-dependent sterol tuning of glycolipid conformation. Front. Physiol. 2013, 4, 120. [CrossRef] [PubMed]
    • (2013) Front. Physiol , vol.4 , pp. 120
    • Fantini, J.1    Yahi, N.2    Garmy, N.3
  • 115
    • 77955341552 scopus 로고    scopus 로고
    • Sphingomyelin accumulation provides a favorable milieu for GM1 ganglioside-induced assembly of amyloid ß-protein
    • [CrossRef] [PubMed]
    • Yuyama, K.; Yanagisawa, K. Sphingomyelin accumulation provides a favorable milieu for GM1 ganglioside-induced assembly of amyloid ß-protein. Neurosci. Lett. 2010, 481, 168-172. [CrossRef] [PubMed]
    • (2010) Neurosci. Lett , vol.481 , pp. 168-172
    • Yuyama, K.1    Yanagisawa, K.2
  • 116
    • 33846419488 scopus 로고    scopus 로고
    • Lipid raft microdomains and neurotransmitter signalling
    • [CrossRef] [PubMed]
    • Allen, J.A.; Halverson-Tamboli, R.A.; Rasenick, M.M. Lipid raft microdomains and neurotransmitter signalling. Nat. Rev. Neurosci. 2007, 8, 128-140. [CrossRef] [PubMed]
    • (2007) Nat. Rev. Neurosci , vol.8 , pp. 128-140
    • Allen, J.A.1    Halverson-Tamboli, R.A.2    Rasenick, M.M.3
  • 117
    • 34248195469 scopus 로고    scopus 로고
    • Lipid rats and membrane traffic
    • [CrossRef] [PubMed]
    • Hanzal-Bayer, M.F.; Hancock, J.F. Lipid rats and membrane traffic. FEBS Lett. 2007, 581, 2098-2104. [CrossRef] [PubMed]
    • (2007) FEBS Lett , vol.581 , pp. 2098-2104
    • Hanzal-Bayer, M.F.1    Hancock, J.F.2
  • 118
    • 33645299023 scopus 로고    scopus 로고
    • The involvement of lipid rafts in Alzheimer’s disease
    • [CrossRef] [PubMed]
    • Cordy, J.M.; Hooper, N.M.; Turner, A.J. The involvement of lipid rafts in Alzheimer’s disease. Mol. Membr. Biol. 2006, 23, 111-122. [CrossRef] [PubMed]
    • (2006) Mol. Membr. Biol , vol.23 , pp. 111-122
    • Cordy, J.M.1    Hooper, N.M.2    Turner, A.J.3
  • 119
    • 0035826909 scopus 로고    scopus 로고
    • Low cholesterol stimulates the nonamyloidogenic pathway by its effect on the a-secretase ADAM 10
    • [CrossRef] [PubMed]
    • Kojro, E.; Gimpl, G.; Lammich, S.; Marz, W.; Fahrenholz, F. Low cholesterol stimulates the nonamyloidogenic pathway by its effect on the a-secretase ADAM 10. Proc. Natl. Acad. Sci. USA 2001, 98, 5815-5820. [CrossRef] [PubMed]
    • (2001) Proc. Natl. Acad. Sci. USA , vol.98 , pp. 5815-5820
    • Kojro, E.1    Gimpl, G.2    Lammich, S.3    Marz, W.4    Fahrenholz, F.5
  • 120
    • 0037421206 scopus 로고    scopus 로고
    • Amyloidogenic processing of the Alzheimer ß-amyloid precursor protein depends on lipid rafts
    • [CrossRef] [PubMed]
    • Ehehalt, R.; Keller, P.; Haass, C.; Thiele, C.; Simons, K. Amyloidogenic processing of the Alzheimer ß-amyloid precursor protein depends on lipid rafts. J. Cell Biol. 2003, 160, 113-123. [CrossRef] [PubMed]
    • (2003) J. Cell Biol , vol.160 , pp. 113-123
    • Ehehalt, R.1    Keller, P.2    Haass, C.3    Thiele, C.4    Simons, K.5
  • 121
    • 84866430180 scopus 로고    scopus 로고
    • Lipid rafts and Alzheimer’s disease: Protein-lipid interactions and perturbation of signaling
    • [CrossRef] [PubMed]
    • Hicks, D.A.; Nalivaeva, N.N.; Turner, A.J. Lipid rafts and Alzheimer’s disease: Protein-lipid interactions and perturbation of signaling. Front. Physiol. 2012, 3, 189. [CrossRef] [PubMed]
    • (2012) Front. Physiol , vol.3 , pp. 189
    • Hicks, D.A.1    Nalivaeva, N.N.2    Turner, A.J.3
  • 122
    • 79851471469 scopus 로고    scopus 로고
    • Plasmalogen synthesis is regulated via alkyl-dihydorxyacetonephosphate-synthase by amyloid precursor protein processing and is affected in Alzheimer’s disease
    • [CrossRef] [PubMed]
    • Grimm, M.O.; Kuchenbecker, J.; Rothhaar, T.L.; Grosgen, S.; Hundsdorfer, B.; Burg, V.K.; Friess, P.; Muller, U.; Grimm, H.S.; Riemenschneider, M.; et al. Plasmalogen synthesis is regulated via alkyl-dihydorxyacetonephosphate-synthase by amyloid precursor protein processing and is affected in Alzheimer’s disease. J. Neurochem. 2011, 116, 916-925. [CrossRef] [PubMed]
    • (2011) J. Neurochem , vol.116 , pp. 916-925
    • Grimm, M.O.1    Kuchenbecker, J.2    Rothhaar, T.L.3    Grosgen, S.4    Hundsdorfer, B.5    Burg, V.K.6    Friess, P.7    Muller, U.8    Grimm, H.S.9    Riemenschneider, M.10
  • 124
    • 70450235497 scopus 로고    scopus 로고
    • Alzheimer’s disease-like pathological features in transgenic mice expressing the APP intracellular domain
    • [PubMed]
    • Ghosal, K.; Vogt, D.L.; Liang, M.; Shen, Y.; Lamb, B.T.; Pimplikar, S.W. Alzheimer’s disease-like pathological features in transgenic mice expressing the APP intracellular domain. Proc. Natl. Acad. Sci. USA 2009, 106, 18367-18372. [PubMed]
    • (2009) Proc. Natl. Acad. Sci. USA , vol.106 , pp. 18367-18372
    • Ghosal, K.1    Vogt, D.L.2    Liang, M.3    Shen, Y.4    Lamb, B.T.5    Pimplikar, S.W.6
  • 125
    • 84899966426 scopus 로고    scopus 로고
    • Altered lipid composition in cortical lipid rafts occurs at early stages of sporadic Alzheimer’s disease and facilitates APP/BACE1 interactions
    • [CrossRef] [PubMed]
    • Fabelo, N.; Marin, V.; Marin, R.; Moreno, D.; Ferrer, I.; Diaz, M. Altered lipid composition in cortical lipid rafts occurs at early stages of sporadic Alzheimer’s disease and facilitates APP/BACE1 interactions. Neurobiol. Aging 2014, 35, 1801-1812. [CrossRef] [PubMed]
    • (2014) Neurobiol. Aging , vol.35 , pp. 1801-1812
    • Fabelo, N.1    Marin, V.2    Marin, R.3    Moreno, D.4    Ferrer, I.5    Diaz, M.6
  • 126
    • 84877633354 scopus 로고    scopus 로고
    • Inhibition of serine palmitoyltransferase reduces Aß and Tau hyperphosphorylation in a murine model: A safe therapeutic strategy for Alzheimer’s disease
    • [PubMed]
    • Geekiyanage, H.; Upadhye, A.; Chan, C. Inhibition of serine palmitoyltransferase reduces Aß and Tau hyperphosphorylation in a murine model: A safe therapeutic strategy for Alzheimer’s disease. Neurobiol. Aging 2013, 34, 2037-2051. [PubMed]
    • (2013) Neurobiol. Aging , vol.34 , pp. 2037-2051
    • Geekiyanage, H.1    Upadhye, A.2    Chan, C.3
  • 128
    • 11144356498 scopus 로고    scopus 로고
    • Dimeric amyloid ß protein rapidly accumulates in lipid rafts followed by apolipoprotein E and phosphorylated Tau accumulation in the Tg2576 mouse model of Alzheimer’s disease
    • [CrossRef] [PubMed]
    • Kawarabayashi, T.; Shoji, M.; Younkin, L.H.; Wen-Lang, L.; Dickson, D.W.; Murakami, T.; Matsubara, E.; Abe, K.; Ashe, K.H.; Younkin, S.G. Dimeric amyloid ß protein rapidly accumulates in lipid rafts followed by apolipoprotein E and phosphorylated Tau accumulation in the Tg2576 mouse model of Alzheimer’s disease. J. Neurosci. 2004, 24, 3801-3809. [CrossRef] [PubMed]
    • (2004) J. Neurosci , vol.24 , pp. 3801-3809
    • Kawarabayashi, T.1    Shoji, M.2    Younkin, L.H.3    Wen-Lang, L.4    Dickson, D.W.5    Murakami, T.6    Matsubara, E.7    Abe, K.8    Ashe, K.H.9    Younkin, S.G.10
  • 129
    • 61849129656 scopus 로고    scopus 로고
    • Detergent resistant membrane-associated IDE in brain tissue and cultured cells: Relevance to Aß and insulin degradation
    • [CrossRef] [PubMed]
    • Bulloj, A.; Leal, M.C.; Surace, E.I.; Zhang, X.; Xu, H.; Ledesma, M.D.; Castano, E.M.; Morelli, L. Detergent resistant membrane-associated IDE in brain tissue and cultured cells: Relevance to Aß and insulin degradation. Mol. Neurodegener. 2008, 3, 22. [CrossRef] [PubMed]
    • (2008) Mol. Neurodegener , vol.3 , pp. 22
    • Bulloj, A.1    Leal, M.C.2    Surace, E.I.3    Zhang, X.4    Xu, H.5    Ledesma, M.D.6    Castano, E.M.7    Morelli, L.8
  • 131
    • 24644491679 scopus 로고    scopus 로고
    • The synaptic localization of NR2B-containing NMDA receptors is controlled by interactions with PDZ proteins and AP-2
    • [CrossRef] [PubMed]
    • Prybylowski, K.; Chang, K.; Sans, N.; Kan, L.; Vicini, S.; Wenthold, R.J. The synaptic localization of NR2B-containing NMDA receptors is controlled by interactions with PDZ proteins and AP-2. Neuron 2005, 47, 845-857. [CrossRef] [PubMed]
    • (2005) Neuron , vol.47 , pp. 845-857
    • Prybylowski, K.1    Chang, K.2    Sans, N.3    Kan, L.4    Vicini, S.5    Wenthold, R.J.6
  • 132
    • 84874585213 scopus 로고    scopus 로고
    • Amyloid-ß induced signaling by cellular prion protein and Fyn kinase in Alzheimer disease
    • [CrossRef] [PubMed]
    • Um, J.W.; Strittmatter, S.M. Amyloid-ß induced signaling by cellular prion protein and Fyn kinase in Alzheimer disease. Prion 2013, 7, 37-41. [CrossRef] [PubMed]
    • (2013) Prion , vol.7 , pp. 37-41
    • Um, J.W.1    Strittmatter, S.M.2
  • 135
    • 43249114144 scopus 로고    scopus 로고
    • Membrane-bound ß-amyloid oligomers are recruited into lipid rafts by a fyn-dependent mechanism
    • [CrossRef] [PubMed]
    • Williamson, R.; Usardi, A.; Hanger, D.P.; Anderton, B.H. Membrane-bound ß-amyloid oligomers are recruited into lipid rafts by a fyn-dependent mechanism. FASEB J. 2008, 22, 1552-1559. [CrossRef] [PubMed]
    • (2008) FASEB J , vol.22 , pp. 1552-1559
    • Williamson, R.1    Usardi, A.2    Hanger, D.P.3    Anderton, B.H.4
  • 140
    • 84863011560 scopus 로고    scopus 로고
    • Cellular prion protein is essential for oligomeric amyloid-ß-induced neuronal cell death
    • [CrossRef] [PubMed]
    • Kudo, W.; Lee, H.P.; Zou, W.Q.; Wang, X.; Perry, G.; Zhu, X.; Smith, M.A.; Petersen, R.B.; Lee, H.G. Cellular prion protein is essential for oligomeric amyloid-ß-induced neuronal cell death. Hum. Mol. Genet. 2012, 21, 1138-1144. [CrossRef] [PubMed]
    • (2012) Hum. Mol. Genet , vol.21 , pp. 1138-1144
    • Kudo, W.1    Lee, H.P.2    Zou, W.Q.3    Wang, X.4    Perry, G.5    Zhu, X.6    Smith, M.A.7    Petersen, R.B.8    Lee, H.G.9
  • 142
    • 58849084518 scopus 로고    scopus 로고
    • Au phosphorylation by Cdk5 and Fyn in response to amyloid peptide Abera (25-35): Involvement of lipid rafts
    • [PubMed]
    • Hernandez, P.; Lee, G.; Sjoberg, M.; Maccioni, R.B. Tau phosphorylation by Cdk5 and Fyn in response to amyloid peptide Abera (25-35): Involvement of lipid rafts. J. Alzheimers Dis. 2009, 16, 149-156. [PubMed]
    • (2009) J. Alzheimers Dis , vol.16 , pp. 149-156
    • Hernandez, P.1    Lee, G.2    Sjoberg, M.3    Maccioni, R.B.4
  • 143
    • 33744515765 scopus 로고    scopus 로고
    • Recruitment of active glycogen synthase kinase-3 into neuronal lipid rafts
    • [CrossRef] [PubMed]
    • Sui, Z.; Kovacs, A.D.; Maggirwar, S.B. Recruitment of active glycogen synthase kinase-3 into neuronal lipid rafts. Biochem. Biophys. Res. Commun. 2006, 345, 1643-1648. [CrossRef] [PubMed]
    • (2006) Biochem. Biophys. Res. Commun , vol.345 , pp. 1643-1648
    • Sui, Z.1    Kovacs, A.D.2    Maggirwar, S.B.3
  • 144
    • 0029966469 scopus 로고    scopus 로고
    • The Cdk5/p35 kinase is essential for neurite outgrowth during neuronal differentiation
    • [CrossRef] [PubMed]
    • Nikolic, M.; Dudek, H.; Kwon, Y.T.; Ramos, Y.F.; Tsai, L.H. The Cdk5/p35 kinase is essential for neurite outgrowth during neuronal differentiation. Genes Dev. 1996, 10, 816-825. [CrossRef] [PubMed]
    • (1996) Genes Dev , vol.10 , pp. 816-825
    • Nikolic, M.1    Dudek, H.2    Kwon, Y.T.3    Ramos, Y.F.4    Tsai, L.H.5
  • 145
    • 84904333334 scopus 로고    scopus 로고
    • Physiological and pathological phosphorylation of Tau by Cdk5
    • [CrossRef] [PubMed]
    • Kimura, T.; Ishiguro, K.; Hisanaga, S. Physiological and pathological phosphorylation of Tau by Cdk5. Front. Mol. Neurosci. 2014, 7, 65. [CrossRef] [PubMed]
    • (2014) Front. Mol. Neurosci , vol.7 , pp. 65
    • Kimura, T.1    Ishiguro, K.2    Hisanaga, S.3
  • 146
    • 84871649258 scopus 로고    scopus 로고
    • Deregulated Cdk5 activity is involved in inducing Alzheimer’s disease
    • [CrossRef] [PubMed]
    • Shukla, V.; Skuntz, S.; Pant, H.C. Deregulated Cdk5 activity is involved in inducing Alzheimer’s disease. Arch. Med. Res. 2012, 43, 655-662. [CrossRef] [PubMed]
    • (2012) Arch. Med. Res , vol.43 , pp. 655-662
    • Shukla, V.1    Skuntz, S.2    Pant, H.C.3
  • 147
    • 34447503455 scopus 로고    scopus 로고
    • Untangling Tau hyperphosphorylation in drug design for neurodegenerative diseases
    • [CrossRef] [PubMed]
    • Mazanetz, M.P.; Fischer, P.M. Untangling Tau hyperphosphorylation in drug design for neurodegenerative diseases. Nat. Rev. Drug Discov. 2007, 6, 464-479. [CrossRef] [PubMed]
    • (2007) Nat. Rev. Drug Discov , vol.6 , pp. 464-479
    • Mazanetz, M.P.1    Fischer, P.M.2
  • 149
    • 27444437758 scopus 로고    scopus 로고
    • Disease-related modifications in Tau affect the interaction between Fyn and Tau
    • [CrossRef] [PubMed]
    • Bhaskar, K.; Yen, S.H.; Lee, G. Disease-related modifications in Tau affect the interaction between Fyn and Tau. J. Biol. Chem. 2005, 280, 35119-35125. [CrossRef] [PubMed]
    • (2005) J. Biol. Chem , vol.280 , pp. 35119-35125
    • Bhaskar, K.1    Yen, S.H.2    Lee, G.3
  • 151
    • 79961025441 scopus 로고    scopus 로고
    • Tyrosine phosphorylation of Tau regulates its interactions with Fyn SH2 domains, but not SH3 domains, altering the cellular localization of tau
    • [CrossRef] [PubMed]
    • Usardi, A.; Pooler, A.M.; Seereeram, A.M.; Reynolds, C.H.; Derkinderen, P.; Anderton, B.; Hanger, D.P.; Noble, W.; Williamson, R. Tyrosine phosphorylation of Tau regulates its interactions with Fyn SH2 domains, but not SH3 domains, altering the cellular localization of tau. FEBS J. 2011, 278, 2927-2937. [CrossRef] [PubMed]
    • (2011) FEBS J , vol.278 , pp. 2927-2937
    • Usardi, A.1    Pooler, A.M.2    Seereeram, A.M.3    Reynolds, C.H.4    Derkinderen, P.5    Anderton, B.6    Hanger, D.P.7    Noble, W.8    Williamson, R.9
  • 152
    • 67650072530 scopus 로고    scopus 로고
    • Reduced O-GlcNAcylation links lower brain glucose metabolism and Tau pathology in Alzheimer’s disease
    • [CrossRef] [PubMed]
    • Liu, F.; Shi, J.; Tranimukai, H.; Gu, J.; Gu, J.; Grundke-Iqbal, I.; Iqbal, K.; Gong, C.X. Reduced O-GlcNAcylation links lower brain glucose metabolism and Tau pathology in Alzheimer’s disease. Brain 2009, 132, 1820-1832. [CrossRef] [PubMed]
    • (2009) Brain , vol.132 , pp. 1820-1832
    • Liu, F.1    Shi, J.2    Tranimukai, H.3    Gu, J.4    Gu, J.5    Grundke-Iqbal, I.6    Iqbal, K.7    Gong, C.X.8
  • 153
    • 28044458281 scopus 로고    scopus 로고
    • Energy inhibition elevates ß-secretase levels and activity and is potentially amyloidogenic in APP transgenic mice: Possible early events in Alzheimer’s disease pathogenesis
    • [CrossRef] [PubMed]
    • Velliquette, R.A.; O’Connor, T.; Vassar, R. Energy inhibition elevates ß-secretase levels and activity and is potentially amyloidogenic in APP transgenic mice: Possible early events in Alzheimer’s disease pathogenesis. J. Neurosci. 2005, 25, 10874-10883. [CrossRef] [PubMed]
    • (2005) J. Neurosci , vol.25 , pp. 10874-10883
    • Velliquette, R.A.1    O’Connor, T.2    Vassar, R.3
  • 154
  • 155
    • 77956219269 scopus 로고    scopus 로고
    • Rethinking Alzheimer’s disease therapy: Are mitochondria the key
    • [PubMed]
    • Ankarcrona, M.; Mangialasche, F.; Winblad, B. Rethinking Alzheimer’s disease therapy: Are mitochondria the key? J. Alzheimers Dis. 2010, 20, S579-S590.[PubMed]
    • (2010) J. Alzheimers Dis , vol.20 , pp. S579-S590
    • Ankarcrona, M.1    Mangialasche, F.2    Winblad, B.3
  • 156
    • 78751620383 scopus 로고    scopus 로고
    • O-GlcNAcylation increases non-amyloidogenic processing of the amyloid-ß precursor protein (APP)
    • [CrossRef] [PubMed]
    • Jacobsen, K.T.; Iverfeldt, K. O-GlcNAcylation increases non-amyloidogenic processing of the amyloid-ß precursor protein (APP). Biochem. Biophys. Res. Commun. 2011, 404, 882-886. [CrossRef] [PubMed]
    • (2011) Biochem. Biophys. Res. Commun , vol.404 , pp. 882-886
    • Jacobsen, K.T.1    Iverfeldt, K.2
  • 157
    • 2542446200 scopus 로고    scopus 로고
    • Accumulation of protein O-GlcNAc modification inhibits proteasomes in the brain and coincides with neuronal apoptosis in brain areas with high O-GlcNAc metabolism
    • [CrossRef] [PubMed]
    • Liu, K.; Paterson, A.J.; Zhang, F.; McAndrew, J.; Fukuchi, K.; Wyss, J.M.; Peng, L.; Hu, Y.; Kudlow, J.E. Accumulation of protein O-GlcNAc modification inhibits proteasomes in the brain and coincides with neuronal apoptosis in brain areas with high O-GlcNAc metabolism. J. Neurochem. 2004, 89, 1044-1055. [CrossRef] [PubMed]
    • (2004) J. Neurochem , vol.89 , pp. 1044-1055
    • Liu, K.1    Paterson, A.J.2    Zhang, F.3    McAndrew, J.4    Fukuchi, K.5    Wyss, J.M.6    Peng, L.7    Hu, Y.8    Kudlow, J.E.9
  • 158
    • 84896271270 scopus 로고    scopus 로고
    • OGlcNAcylation and phosphorylation have opposing structural effects in Tau: Phosphothreonine induces particular conformational order
    • [CrossRef] [PubMed]
    • Brister, M.A.; Pandey, A.K.; Bielska, A.A.; Zondlo, N.J. OGlcNAcylation and phosphorylation have opposing structural effects in Tau: Phosphothreonine induces particular conformational order. J. Am. Chem. Soc. 2014, 136, 3803-3816. [CrossRef] [PubMed]
    • (2014) J. Am. Chem. Soc , vol.136 , pp. 3803-3816
    • Brister, M.A.1    Pandey, A.K.2    Bielska, A.A.3    Zondlo, N.J.4
  • 160
    • 3242739968 scopus 로고    scopus 로고
    • O-GlcNAcylation regulates phosphorylation ofTau: A mechanism involved in Alzheimer’s disease
    • [CrossRef] [PubMed]
    • Liu, F.; Iqbal, K.; Grundke-Igbal, I.; Hart, G.W.; Gong, C.X. O-GlcNAcylation regulates phosphorylation ofTau: A mechanism involved in Alzheimer’s disease. Proc. Natl. Acad. Sci. USA 2004, 101, 10804-10809. [CrossRef] [PubMed]
    • (2004) Proc. Natl. Acad. Sci. USA , vol.101 , pp. 10804-10809
    • Liu, F.1    Iqbal, K.2    Grundke-Igbal, I.3    Hart, G.W.4    Gong, C.X.5
  • 161
    • 58649120835 scopus 로고    scopus 로고
    • In vivo modulation of O-GlcNAc levels regulates hippocampal synaptic plasticity through interplay with phosphorylation
    • [CrossRef] [PubMed]
    • Tallent, M.K.; Varghis, N.; Skorobogatko, Y.; Hernandez-Cuebas, L.; Whelan, K.; Vocadlo, D.J.; Vosseller, K. In vivo modulation of O-GlcNAc levels regulates hippocampal synaptic plasticity through interplay with phosphorylation. J. Biol. Chem. 2009, 284, 174-181. [CrossRef] [PubMed]
    • (2009) J. Biol. Chem , vol.284 , pp. 174-181
    • Tallent, M.K.1    Varghis, N.2    Skorobogatko, Y.3    Hernandez-Cuebas, L.4    Whelan, K.5    Vocadlo, D.J.6    Vosseller, K.7
  • 162
    • 67049134478 scopus 로고    scopus 로고
    • Oxidative stress in Alzheimer disease
    • [CrossRef] [PubMed]
    • Gella, A.; Durany, N. Oxidative stress in Alzheimer disease. Cell Adh. Migr. 2009, 3, 88-93. [CrossRef] [PubMed]
    • (2009) Cell Adh. Migr , vol.3 , pp. 88-93
    • Gella, A.1    Durany, N.2
  • 163
    • 84858161403 scopus 로고    scopus 로고
    • Elevation of glutathione as a therapeutic strategy in Alzheimer disease
    • [PubMed]
    • Pocernich, C.B.; Butterfield, D.A. Elevation of glutathione as a therapeutic strategy in Alzheimer disease. Biochim. Biophys. Acta 2012, 1822, 625-630. [PubMed]
    • (2012) Biochim. Biophys. Acta , vol.1822 , pp. 625-630
    • Pocernich, C.B.1    Butterfield, D.A.2
  • 164
    • 0034652426 scopus 로고    scopus 로고
    • Age-associated decline in ?-glutamylcysteine synthetase gene expression in rats
    • [CrossRef]
    • Liu, R.; Choi, J. Age-associated decline in ?-glutamylcysteine synthetase gene expression in rats. Free Radic.Biol. Med. 2000, 28, 566-574. [CrossRef]
    • (2000) Free Radic.Biol. Med , vol.28 , pp. 566-574
    • Liu, R.1    Choi, J.2
  • 165
    • 84901911564 scopus 로고    scopus 로고
    • The emerging role of glutathione in Alzheimer’s disease
    • [PubMed]
    • Saharan, S.; Mandal, P.K. The emerging role of glutathione in Alzheimer’s disease. J. Alzheimers Dis. 2014, 40, 519-529. [PubMed]
    • (2014) J. Alzheimers Dis , vol.40 , pp. 519-529
    • Saharan, S.1    Mandal, P.K.2
  • 166
    • 77954452230 scopus 로고    scopus 로고
    • Depletion of GSH in glial cells induces neurotoxicity: Relevance to aging and degenerative neurological diseases
    • [CrossRef] [PubMed]
    • Lee, M.; Cho, T.; Jantaratnotai, N.; Wang, Y.T.; McGeer, E.; McGeer, P.L. Depletion of GSH in glial cells induces neurotoxicity: Relevance to aging and degenerative neurological diseases. FASEB J. 2010, 24, 2533-2545. [CrossRef] [PubMed]
    • (2010) FASEB J , vol.24 , pp. 2533-2545
    • Lee, M.1    Cho, T.2    Jantaratnotai, N.3    Wang, Y.T.4    McGeer, E.5    McGeer, P.L.6
  • 167
    • 47749147258 scopus 로고    scopus 로고
    • Hydrogen peroxide promotes Aß production through JNK-dependent activation of ?-secretase
    • [CrossRef] [PubMed]
    • Shen, C.; Chen, Y.; Liu, H.; Zhang, K.; Zhang, T.; Lin, A.; Jing, N. Hydrogen peroxide promotes Aß production through JNK-dependent activation of ?-secretase. J. Biol. Chem. 2008, 283, 17721-17730. [CrossRef] [PubMed]
    • (2008) J. Biol. Chem , vol.283 , pp. 17721-17730
    • Shen, C.1    Chen, Y.2    Liu, H.3    Zhang, K.4    Zhang, T.5    Lin, A.6    Jing, N.7
  • 168
    • 33646534207 scopus 로고    scopus 로고
    • St.; Clair, D.K. ß-amyloid mediated nitration of manganese superoxide dismutase: Implication for oxidative stress in a APPNLH/NLH X PS-1P264L/P264L double knock-in mouse model of Alzheimer’s disease
    • [CrossRef] [PubMed]
    • Anantharaman, M.; Tangpong, J.; Keller, J.N.; Murphy, M.P.; Markesbery, W.R.; Kiningham, K.K., St.; Clair, D.K. ß-amyloid mediated nitration of manganese superoxide dismutase: Implication for oxidative stress in a APPNLH/NLH X PS-1P264L/P264L double knock-in mouse model of Alzheimer’s disease. Am. J. Pathol. 2006, 168, 1608-1617. [CrossRef] [PubMed]
    • (2006) Am. J. Pathol , vol.168 , pp. 1608-1617
    • Anantharaman, M.1    Tangpong, J.2    Keller, J.N.3    Murphy, M.P.4    Markesbery, W.R.5    Kiningham, K.K.6
  • 170
    • 0033844944 scopus 로고    scopus 로고
    • Characterization of copper interactions with Alzheimer amyloid ß peptides: Identification of an attomolar-affinity copper binding site on amyloid ß1-42
    • [CrossRef] [PubMed]
    • Atwood, C.S.; Scarpa, R.C.; Huang, X.; Moir, R.D.; Jones, W.D.; Fairlie, D.P.; Tanzi, R.E.; Bush, A.I. Characterization of copper interactions with Alzheimer amyloid ß peptides: Identification of an attomolar-affinity copper binding site on amyloid ß1-42. J. Neurochem. 2000, 75, 1219-1233. [CrossRef] [PubMed]
    • (2000) J. Neurochem , vol.75 , pp. 1219-1233
    • Atwood, C.S.1    Scarpa, R.C.2    Huang, X.3    Moir, R.D.4    Jones, W.D.5    Fairlie, D.P.6    Tanzi, R.E.7    Bush, A.I.8
  • 173
    • 0032531735 scopus 로고    scopus 로고
    • Electrochemical analysis of protein nitrotyrosine and dityrosine in the Alzheimer brain indicates region-specific accumulation
    • [PubMed]
    • Hensley, K.; Maidt, M.L.; Yu, Z.; Sang, H.; Markesbery, W.R.; Floyd, R.A. Electrochemical analysis of protein nitrotyrosine and dityrosine in the Alzheimer brain indicates region-specific accumulation. J. Neurosci. 1998, 18, 8126-8132. [PubMed]
    • (1998) J. Neurosci , vol.18 , pp. 8126-8132
    • Hensley, K.1    Maidt, M.L.2    Yu, Z.3    Sang, H.4    Markesbery, W.R.5    Floyd, R.A.6
  • 174
    • 0034733705 scopus 로고    scopus 로고
    • Evidence that the ß-amyloid plaques of Alzheimer’s disease represent the redox-silencing and entombment of Aß by zinc
    • [CrossRef] [PubMed]
    • Cuajungco, M.P.; Goldstein, L.E.; Nunomura, A.; Smith, M.A.; Lim, J.T.; Atwood, C.S.; Huang, X.; Farrag, Y.W.; Perry, G.; Bush, A.I. Evidence that the ß-amyloid plaques of Alzheimer’s disease represent the redox-silencing and entombment of Aß by zinc. J. Biol. Chem. 2000, 275, 19439-19442. [CrossRef] [PubMed]
    • (2000) J. Biol. Chem , vol.275 , pp. 19439-19442
    • Cuajungco, M.P.1    Goldstein, L.E.2    Nunomura, A.3    Smith, M.A.4    Lim, J.T.5    Atwood, C.S.6    Huang, X.7    Farrag, Y.W.8    Perry, G.9    Bush, A.I.10
  • 178
    • 79954585505 scopus 로고    scopus 로고
    • Advanced glycation endproducts and their receptor RAGE in Alzheimer’s disease
    • [CrossRef] [PubMed]
    • Srikanth, V.; Maczurek, A.; Phan, T.; Steele, M.; Westcott, B.; Juskiw, D.; Munch, G. Advanced glycation endproducts and their receptor RAGE in Alzheimer’s disease. Neurobiol. Aging 2011, 32, 763-777. [CrossRef] [PubMed]
    • (2011) Neurobiol. Aging , vol.32 , pp. 763-777
    • Srikanth, V.1    Maczurek, A.2    Phan, T.3    Steele, M.4    Westcott, B.5    Juskiw, D.6    Munch, G.7
  • 179
    • 70449732132 scopus 로고    scopus 로고
    • Immunohistochemical analysis ofhuman brain suggests pathological synergism of Alzheimer’s disease and diabetes mellitus
    • [CrossRef] [PubMed]
    • Valente, T.; Gella, A.; Fernandez-Busquets, X.; Unzeta, M.; Durany, N. Immunohistochemical analysis ofhuman brain suggests pathological synergism of Alzheimer’s disease and diabetes mellitus. Neurobiol. Dis. 2010, 37, 67-76. [CrossRef] [PubMed]
    • (2010) Neurobiol. Dis , vol.37 , pp. 67-76
    • Valente, T.1    Gella, A.2    Fernandez-Busquets, X.3    Unzeta, M.4    Durany, N.5
  • 180
    • 68849129707 scopus 로고    scopus 로고
    • BACE1 and Aß generation via NFAT1 activation in Alzheimer’s disease animal model
    • [CrossRef] [PubMed]
    • Cho, H.J.; Son, S.M.; Jin, S.M.; Hong, H.S.; Shin, D.H.; Kim, S.J.; Huh, K.; Mook-Jung, I. RAGE regulates BACE1 and Aß generation via NFAT1 activation in Alzheimer’s disease animal model. FASEB J. 2009, 23, 2639-2649. [CrossRef] [PubMed]
    • (2009) FASEB J , vol.23 , pp. 2639-2649
    • Cho, H.J.1    Son, S.M.2    Jin, S.M.3    Hong, H.S.4    Shin, D.H.5    Kim, S.J.6    Huh, K.7    Mook-Jung, I.8
  • 181
    • 67649739088 scopus 로고    scopus 로고
    • A progression factor foramyloid-ß-induced cellular perturbation
    • [PubMed]
    • Yan, S.D.; Bierhaus, A.; Nawroth, P.P.; Stern, D.M. RAGE and Alzheimer’s disease: A progression factor foramyloid-ß-induced cellular perturbation? J. Alzheimers Dis. 2009, 16, 833-843. [PubMed]
    • (2009) J. Alzheimers Dis , vol.16 , pp. 833-843
    • Yan, S.D.1    Bierhaus, A.2    Nawroth, P.P.3    Stern, D.4
  • 182
    • 0029870240 scopus 로고    scopus 로고
    • Blood-brain barrier uptake of the 40 and 42 amino acid sequences of circulating Alzheimer’s amyloid ß in guinea pigs
    • [CrossRef]
    • Martel, C.L.; Mackic, J.B.; McComb, J.G.; Ghiso, J.; Zlokovic, B.V. Blood-brain barrier uptake of the 40 and 42 amino acid sequences of circulating Alzheimer’s amyloid ß in guinea pigs. Neurosci. Lett. 1996, 206, 157-160. [CrossRef]
    • (1996) Neurosci. Lett , vol.206 , pp. 157-160
    • Martel, C.L.1    Mackic, J.B.2    McComb, J.G.3    Ghiso, J.4    Zlokovic, B.V.5
  • 183
    • 0037703255 scopus 로고    scopus 로고
    • RAGE mediates amyloid-ß peptide transport across the blood-brain barrier and accumlation in brain
    • [CrossRef] [PubMed]
    • Deane, R.; du Yan, S.; Submamaryan, R.K.; LaRue, B.; Jovanovic, S.; Hogg, E.; Welch, D.; Manness, L.; Lin, C.; Yu, J.; et al. RAGE mediates amyloid-ß peptide transport across the blood-brain barrier and accumlation in brain. Nat. Med. 2003, 9, 907-913. [CrossRef] [PubMed]
    • (2003) Nat. Med , vol.9 , pp. 907-913
    • Deane, R.1    Du Yan, S.2    Submamaryan, R.K.3    Larue, B.4    Jovanovic, S.5    Hogg, E.6    Welch, D.7    Manness, L.8    Lin, C.9    Yu, J.10
  • 184
    • 77958494422 scopus 로고    scopus 로고
    • Linking Alzheimer’s disease to insulin resistance: The FoxO response to oxidative stress
    • [CrossRef] [PubMed]
    • Manolopoulos, K.N.; Klotz, L.O.; Korsten, P.; Bornstein, S.R.; Barthel, A. Linking Alzheimer’s disease to insulin resistance: The FoxO response to oxidative stress. Mol. Psychiatry 2010, 15, 1046-1052. [CrossRef] [PubMed]
    • (2010) Mol. Psychiatry , vol.15 , pp. 1046-1052
    • Manolopoulos, K.N.1    Klotz, L.O.2    Korsten, P.3    Bornstein, S.R.4    Barthel, A.5
  • 186
    • 33846949357 scopus 로고    scopus 로고
    • The oxidative stress metabolite 4-hydroxynonenal promotes Alzheimer protofibril formation
    • [CrossRef] [PubMed]
    • Siegel, S.J.; Bieschke, J.; Powers, E.T.; Kelly, J.W. The oxidative stress metabolite 4-hydroxynonenal promotes Alzheimer protofibril formation. Biochemistry 2007, 46, 1503-1510. [CrossRef] [PubMed]
    • (2007) Biochemistry , vol.46 , pp. 1503-1510
    • Siegel, S.J.1    Bieschke, J.2    Powers, E.T.3    Kelly, J.W.4
  • 187
    • 0031980065 scopus 로고    scopus 로고
    • Four-hydroxynonenal, a product of lipid peroxidation, is increased in the brain in Alzheimer’s disease
    • [CrossRef]
    • Markesbery, W.R.; Lovell, M.A. Four-hydroxynonenal, a product of lipid peroxidation, is increased in the brain in Alzheimer’s disease. Neurobiol. Aging 1998, 19, 33-36. [CrossRef]
    • (1998) Neurobiol. Aging , vol.19 , pp. 33-36
    • Markesbery, W.R.1    Lovell, M.A.2
  • 188
    • 0033664539 scopus 로고    scopus 로고
    • Insulin receptors and insulin action in the brain:Review and clinical implications
    • [CrossRef]
    • Schulingkamp, R.J.; Pagano, T.C.; Hung, D.; Raffa, R.B. Insulin receptors and insulin action in the brain:Review and clinical implications. Neurosci. Biobehav. Rev. 2000, 24, 855-872. [CrossRef]
    • (2000) Neurosci. Biobehav. Rev , vol.24 , pp. 855-872
    • Schulingkamp, R.J.1    Pagano, T.C.2    Hung, D.3    Raffa, R.B.4
  • 189
    • 0030902595 scopus 로고    scopus 로고
    • Insulin-like growth factor I protects and rescues hippocampal neurons against ß-amyloid- and human amylin-induced toxicity
    • [CrossRef] [PubMed]
    • Dore, S.; Kar, S.; Quirion, R. Insulin-like growth factor I protects and rescues hippocampal neurons against ß-amyloid- and human amylin-induced toxicity. Proc. Natl. Acad. Sci. USA 1997, 94, 4772-4777. [CrossRef] [PubMed]
    • (1997) Proc. Natl. Acad. Sci. USA , vol.94 , pp. 4772-4777
    • Dore, S.1    Kar, S.2    Quirion, R.3
  • 190
    • 84858631674 scopus 로고    scopus 로고
    • The many faces of insulin-like peptide signalling in the brain
    • [CrossRef] [PubMed]
    • Fernandez, A.M.; Torres-Aleman, I. The many faces of insulin-like peptide signalling in the brain. Nat. Rev. Neurosci. 2012, 13, 225-239. [CrossRef] [PubMed]
    • (2012) Nat. Rev. Neurosci , vol.13 , pp. 225-239
    • Fernandez, A.M.1    Torres-Aleman, I.2
  • 191
    • 0027391758 scopus 로고
    • The cellular and physiological actions of insulin in the central nervous system
    • [CrossRef]
    • Wozniak, M.; Rydzewski, B.; Baker, S.P.; Raizada, M.K. The cellular and physiological actions of insulin in the central nervous system. Neurochem. Int. 1993, 22, 1-10. [CrossRef]
    • (1993) Neurochem. Int , vol.22 , pp. 1-10
    • Wozniak, M.1    Rydzewski, B.2    Baker, S.P.3    Raizada, M.K.4
  • 192
    • 0034810894 scopus 로고    scopus 로고
    • Defects of the isnulin receptor substrate (IRS) system in human metabolic disorders
    • [CrossRef] [PubMed]
    • Sesti, G.; Federici, M.; Hribal, M.L.; Lauro, D.; Sbraccia, P.; Lauro, R. Defects of the isnulin receptor substrate (IRS) system in human metabolic disorders. FASEB J. 2001, 15, 2099-2111. [CrossRef] [PubMed]
    • (2001) FASEB J , vol.15 , pp. 2099-2111
    • Sesti, G.1    Federici, M.2    Hribal, M.L.3    Lauro, D.4    Sbraccia, P.5    Lauro, R.6
  • 193
    • 0344305782 scopus 로고    scopus 로고
    • Insulin signaling in health and disease
    • [CrossRef] [PubMed]
    • White, M.F. Insulin signaling in health and disease. Science 2003, 302, 1710-1711. [CrossRef] [PubMed]
    • (2003) Science , vol.302 , pp. 1710-1711
    • White, M.F.1
  • 196
    • 27844537831 scopus 로고    scopus 로고
    • Tau is hyperphosphorylated in the insulin-like growth factor-I null brain
    • [CrossRef] [PubMed]
    • Cheng, C.M.; Tseng, V.; Wang, J.; Wang, D.; Matyakhina, L.; Bondy, C.A. Tau is hyperphosphorylated in the insulin-like growth factor-I null brain. Endocrinology 2005, 146, 5086-5091. [CrossRef] [PubMed]
    • (2005) Endocrinology , vol.146 , pp. 5086-5091
    • Cheng, C.M.1    Tseng, V.2    Wang, J.3    Wang, D.4    Matyakhina, L.5    Bondy, C.A.6
  • 198
    • 71349085957 scopus 로고    scopus 로고
    • Defects in IGF-1 receptor, insulin receptor and IRS-1/2 in Alzheimer’s disease indicate possible resistance to IGF-1 and insulin signalling
    • [CrossRef] [PubMed]
    • Moloney, A.M.; Griffin, R.J.; Timmons, S.; O’Connor, R.; Ravid, R.; O’Neil, C. Defects in IGF-1 receptor, insulin receptor and IRS-1/2 in Alzheimer’s disease indicate possible resistance to IGF-1 and insulin signalling. Neurobiol. Aging 2010, 31, 224-243. [CrossRef] [PubMed]
    • (2010) Neurobiol. Aging , vol.31 , pp. 224-243
    • Moloney, A.M.1    Griffin, R.J.2    Timmons, S.3    O’Connor, R.4    Ravid, R.5    O’Neil, C.6
  • 199
    • 0029587224 scopus 로고
    • Inhibition of glycogen synthase kinase-3 by insulin mediated by protein kinase B
    • [CrossRef] [PubMed]
    • Cross, D.A.; Alessi, D.R.; Cohen, P.; Andjelkovich, M.; Hemmings, B.A. Inhibition of glycogen synthase kinase-3 by insulin mediated by protein kinase B. Nature 1995, 378, 785-789. [CrossRef] [PubMed]
    • (1995) Nature , vol.378 , pp. 785-789
    • Cross, D.A.1    Alessi, D.R.2    Cohen, P.3    Andjelkovich, M.4    Hemmings, B.A.5
  • 200
    • 0031913246 scopus 로고    scopus 로고
    • Mechanism of activation and function of protein kinase B
    • [CrossRef]
    • Alessi, D.R.; Cohen, P. Mechanism of activation and function of protein kinase B. Curr. Opin. Genet. Dev.1998, 8, 55-62. [CrossRef]
    • (1998) Curr. Opin. Genet. Dev , vol.8 , pp. 55-62
    • Alessi, D.R.1    Cohen, P.2
  • 202
    • 0030061922 scopus 로고    scopus 로고
    • IRS-1-mediated inhibition of insulin receptor tyrosine kinase activity in TNF-a- and obesity-induced insulin resistance
    • [CrossRef] [PubMed]
    • Hotamisligil, G.S.; Peraldi, P.; Budavari, A.; Ellis, R.; White, M.F.; Spiegelman, B.M. IRS-1-mediated inhibition of insulin receptor tyrosine kinase activity in TNF-a- and obesity-induced insulin resistance. Science 1996, 271, 665-668. [CrossRef] [PubMed]
    • (1996) Science , vol.271 , pp. 665-668
    • Hotamisligil, G.S.1    Peraldi, P.2    Budavari, A.3    Ellis, R.4    White, M.F.5    Spiegelman, B.M.6
  • 203
    • 0037059330 scopus 로고    scopus 로고
    • Phosphorylation of Ser307 in insulin receptor substrate-1 blocks interactions with the insulin receptor and inhibits insulin action
    • [CrossRef] [PubMed]
    • Aguirre, V.; Werner, E.D.; Giraud, J.; Lee, Y.H.; Shoelson, S.E.; White, M.F. Phosphorylation of Ser307 in insulin receptor substrate-1 blocks interactions with the insulin receptor and inhibits insulin action. J. Biol. Chem. 2002, 277, 1531-1537. [CrossRef] [PubMed]
    • (2002) J. Biol. Chem , vol.277 , pp. 1531-1537
    • Aguirre, V.1    Werner, E.D.2    Giraud, J.3    Lee, Y.H.4    Shoelson, S.E.5    White, M.F.6
  • 204
    • 0030669392 scopus 로고    scopus 로고
    • Molecular basis for insulin resistance. Elevated serine/threonine phosphorylation of IRS-1 and IRS-2 inhibits their binding to the juxtamembrane region of the insulin and impairs their ability to undergo insulin-induced tyrosine phosphorylation
    • [CrossRef] [PubMed]
    • Paz, K.; Hemi, R.; LeRoith, D.; Karasik, A.; Elhanany, E.; Kanety, H.; Zick, Y. A molecular basis for insulin resistance. Elevated serine/threonine phosphorylation of IRS-1 and IRS-2 inhibits their binding to the juxtamembrane region of the insulin and impairs their ability to undergo insulin-induced tyrosine phosphorylation. J. Biol. Chem. 1997, 272, 29911-29918. [CrossRef] [PubMed]
    • (1997) J. Biol. Chem , vol.272 , pp. 29911-29918
    • Paz, K.1    Hemi, R.2    Leroith, D.3    Karasik, A.4    Elhanany, E.5    Kanety, H.6    Zick, Y.A.7
  • 205
    • 84897907030 scopus 로고    scopus 로고
    • How does brain insulin resistance develop in Alzheimer’s disease?
    • [CrossRef] [PubMed]
    • De Felice, F.G.; Lourenco, M.V.; Ferreira, S.T. How does brain insulin resistance develop in Alzheimer’s disease? Alzheimers Dement. 2014, 10, S26-S32. [CrossRef] [PubMed]
    • (2014) Alzheimers Dement , vol.10 , pp. S26-S32
    • De Felice, F.G.1    Lourenco, M.V.2    Ferreira, S.T.3
  • 206
    • 84904113244 scopus 로고    scopus 로고
    • Elevated risk of type 2 diabetes for development of Alzheimer disease: A key role for oxidative stress in brain. Biochim. Biophys
    • [CrossRef] [PubMed]
    • Butterfield, D.A.; di Domenico, F.; Barone, E. Elevated risk of type 2 diabetes for development of Alzheimer disease: A key role for oxidative stress in brain. Biochim. Biophys. Acta 2014, 1842, 1693-1706. [CrossRef] [PubMed]
    • (2014) Acta , vol.1842 , pp. 1693-1706
    • Butterfield, D.A.1    Di Domenico, F.2    Barone, E.3
  • 207
    • 28844495644 scopus 로고    scopus 로고
    • Insulin receptor signaling in long-term memory consolidation following spatial learning
    • [CrossRef] [PubMed]
    • Dou, J.T.; Chen, M.; Dufour, F.; Alkon, D.L.; Zhao, W.Q. Insulin receptor signaling in long-term memory consolidation following spatial learning. Learn. Mem. 2005, 12, 646-655. [CrossRef] [PubMed]
    • (2005) Learn. Mem , vol.12 , pp. 646-655
    • Dou, J.T.1    Chen, M.2    Dufour, F.3    Alkon, D.L.4    Zhao, W.Q.5
  • 208
    • 33845577779 scopus 로고    scopus 로고
    • The role of insulin and neurotrophic factor signaling in brain aging and Alzheimer’s disease
    • [CrossRef] [PubMed]
    • Cole, G.M.; Frautschy, S.A. The role of insulin and neurotrophic factor signaling in brain aging and Alzheimer’s disease. Exp. Gerontol. 2007, 42, 10-21. [CrossRef] [PubMed]
    • (2007) Exp. Gerontol , vol.42 , pp. 10-21
    • Cole, G.M.1    Frautschy, S.A.2
  • 209
    • 67650544970 scopus 로고    scopus 로고
    • Insulin receptor dysfunction impairs cellular clearance of neurotoxic oligomeric Aß
    • [CrossRef] [PubMed]
    • Zhao, W.Q.; Lacor, P.N.; Chen, H.; Lambert, M.P.; Quon, M.J.; Krafft, G.A.; Klein, W.L. Insulin receptor dysfunction impairs cellular clearance of neurotoxic oligomeric Aß. J. Biol. Chem. 2009, 284, 18742-18753. [CrossRef] [PubMed]
    • (2009) J. Biol. Chem , vol.284 , pp. 18742-18753
    • Zhao, W.Q.1    Lacor, P.N.2    Chen, H.3    Lambert, M.P.4    Quon, M.J.5    Krafft, G.A.6    Klein, W.L.7
  • 210
    • 0034161516 scopus 로고    scopus 로고
    • Neurons regulate extracellular levels of amyloid ß-protein via proteolysis by insulin-degrading enzyme
    • [PubMed]
    • Vekrellis, K.; Ye, Z.; Qiu, W.Q.; Walsh, D.; Hartley, D.; Chesneau, V.; Rosner, M.R.; Selkoe, D.J. Neurons regulate extracellular levels of amyloid ß-protein via proteolysis by insulin-degrading enzyme. J. Neurosci. 2000, 20, 1657-1665. [PubMed]
    • (2000) J. Neurosci , vol.20 , pp. 1657-1665
    • Vekrellis, K.1    Ye, Z.2    Qiu, W.Q.3    Walsh, D.4    Hartley, D.5    Chesneau, V.6    Rosner, M.R.7    Selkoe, D.J.8
  • 211
    • 10944228564 scopus 로고    scopus 로고
    • Insulin-degrading enzyme as a downstream target of insulin receptor signaling cascade: Implications for Alzheimer’s disease intervention
    • [CrossRef] [PubMed]
    • Zhao, L.; Teter, B.; Morihara, T.; Lim, G.P.; Ambegaokar, S.S.; Ubeda, O.J.; Frautschy, S.A.; Cole, G.M. Insulin-degrading enzyme as a downstream target of insulin receptor signaling cascade: Implications for Alzheimer’s disease intervention. J. Neurosci. 2004, 24, 11120-11126. [CrossRef] [PubMed]
    • (2004) J. Neurosci , vol.24 , pp. 11120-11126
    • Zhao, L.1    Teter, B.2    Morihara, T.3    Lim, G.P.4    Ambegaokar, S.S.5    Ubeda, O.J.6    Frautschy, S.A.7    Cole, G.M.8
  • 214
    • 51449109656 scopus 로고    scopus 로고
    • Neocortical glial cell numbers in human brains
    • [CrossRef] [PubMed]
    • Pelvig, D.P.; Pakkenberg, H.; Stark, A.K.; Pakkenberg, B. Neocortical glial cell numbers in human brains. Neurobiol. Aging 2008, 29, 1754-1762. [CrossRef] [PubMed]
    • (2008) Neurobiol. Aging , vol.29 , pp. 1754-1762
    • Pelvig, D.P.1    Pakkenberg, H.2    Stark, A.K.3    Pakkenberg, B.4
  • 215
    • 84906307352 scopus 로고    scopus 로고
    • Microglia as a critical player in both developmental and late-life CNS pathogenesis
    • [CrossRef] [PubMed]
    • Derecki, N.C.; Katzmarski, N.; Kipnis, J.; Meyer-Luehmann, M. Microglia as a critical player in both developmental and late-life CNS pathogenesis. Acta Neuropathol. 2014, 128, 333-345. [CrossRef] [PubMed]
    • (2014) Acta Neuropathol , vol.128 , pp. 333-345
    • Derecki, N.C.1    Katzmarski, N.2    Kipnis, J.3    Meyer-Luehmann, M.4
  • 216
  • 217
    • 0345189366 scopus 로고    scopus 로고
    • Inflammatory blockade restores adult hippocampal neurogenesis
    • [CrossRef] [PubMed]
    • Monje, M.L.; Toda, H.; Palmer, T.D. Inflammatory blockade restores adult hippocampal neurogenesis. Science 2003, 302, 1760-1765. [CrossRef] [PubMed]
    • (2003) Science , vol.302 , pp. 1760-1765
    • Monje, M.L.1    Toda, H.2    Palmer, T.D.3
  • 218
    • 31544460682 scopus 로고    scopus 로고
    • Immune cells contribute to the maintenance of neurogenesis and spatial learning abilities in adulthood
    • [CrossRef] [PubMed]
    • Ziv, Y.; Ron, N.; Butovsky, O.; Landa, G.; Greenberg, N.; Cohen, H.; Kipnis, J.; Schwartz, M. Immune cells contribute to the maintenance of neurogenesis and spatial learning abilities in adulthood. Nat. Neurosci. 2006, 9, 268-275. [CrossRef] [PubMed]
    • (2006) Nat. Neurosci , vol.9 , pp. 268-275
    • Ziv, Y.1    Ron, N.2    Butovsky, O.3    Landa, G.4    Greenberg, N.5    Cohen, H.6    Kipnis, J.7    Schwartz, M.8
  • 219
    • 0037336701 scopus 로고    scopus 로고
    • Interleukin-1 mediates pathological effects of microglia on Tau phosphorylation and on synaptophysin synthesis in cortical neurons through a p38-MAPK pathway
    • [PubMed]
    • Li, Y.; Liu, L.; Barger, S.W.; Griffin, W.S. Interleukin-1 mediates pathological effects of microglia on Tau phosphorylation and on synaptophysin synthesis in cortical neurons through a p38-MAPK pathway. J. Neurosci. 2003, 23, 1605-1611. [PubMed]
    • (2003) J. Neurosci , vol.23 , pp. 1605-1611
    • Li, Y.1    Liu, L.2    Barger, S.W.3    Griffin, W.S.4
  • 221
    • 0036682239 scopus 로고    scopus 로고
    • Fractalkine and fractalkine receptors in human neurons and glial cells
    • [CrossRef] [PubMed]
    • Hatori, H.; Nagai, A.; Heisel, R.; Ryu, J.K.; Kim, S.U. Fractalkine and fractalkine receptors in human neurons and glial cells. J. Neurosci. Res. 2002, 69, 418-426. [CrossRef] [PubMed]
    • (2002) J. Neurosci. Res , vol.69 , pp. 418-426
    • Hatori, H.1    Nagai, A.2    Heisel, R.3    Ryu, J.K.4    Kim, S.U.5
  • 222
    • 33745630948 scopus 로고    scopus 로고
    • Fractalkine: Moving from chemotaxis to neuroprotection. Nat
    • [PubMed]
    • Re, D.B.; Przedborski, S. Fractalkine: Moving from chemotaxis to neuroprotection. Nat. Neurosci. 2006, 9, 859-861. [PubMed]
    • (2006) Neurosci , vol.9 , pp. 859-861
    • Re, D.B.1    Przedborski, S.2
  • 223
    • 68949200462 scopus 로고    scopus 로고
    • Fractalkine-induced activation of the phosphatidylinositol-3 kinase pathway attenuates microglial activation in vivo and in vitro
    • [CrossRef] [PubMed]
    • Lyons, A.; Lynch, A.M.; Downer, E.J.; Hanley, R.; O’Sullivan, J.B.; Smith, A.; Lynch, M.A. Fractalkine-induced activation of the phosphatidylinositol-3 kinase pathway attenuates microglial activation in vivo and in vitro. J. Neurochem. 2009, 110, 1547-1556. [CrossRef] [PubMed]
    • (2009) J. Neurochem , vol.110 , pp. 1547-1556
    • Lyons, A.1    Lynch, A.M.2    Downer, E.J.3    Hanley, R.4    O’Sullivan, J.B.5    Smith, A.6    Lynch, M.A.7
  • 224
    • 10944260096 scopus 로고    scopus 로고
    • Fractalkine reduces N-methyl-D-aspartate-induced calcium flux and apoptosis in human neurons through extracellular signal-regulated kinase activation
    • [CrossRef] [PubMed]
    • Deiva, K.; Geeraerts, T.; Salim, H.; Leclerc, P.; Hery, C.; Hugel, B.; Freyssinet, J.M.; Tardieu, M. Fractalkine reduces N-methyl-D-aspartate-induced calcium flux and apoptosis in human neurons through extracellular signal-regulated kinase activation. Eur. J. Neurosci. 2004, 20, 3222-3232. [CrossRef] [PubMed]
    • (2004) Eur. J. Neurosci , vol.20 , pp. 3222-3232
    • Deiva, K.1    Geeraerts, T.2    Salim, H.3    Leclerc, P.4    Hery, C.5    Hugel, B.6    Freyssinet, J.M.7    Tardieu, M.8
  • 226
    • 41949125406 scopus 로고    scopus 로고
    • Changes in the levels of plasma soluble fractalkine in patients with mild cognitive impairment and Alzheimer’s disease
    • [CrossRef] [PubMed]
    • Kim, T.S.; Lim, H.K.; Lee, J.Y.; Kim, D.J.; Park, S.; Lee, C.; Lee, C.U. Changes in the levels of plasma soluble fractalkine in patients with mild cognitive impairment and Alzheimer’s disease. Neurosci. Lett. 2008, 436, 196-200. [CrossRef] [PubMed]
    • (2008) Neurosci. Lett , vol.436 , pp. 196-200
    • Kim, T.S.1    Lim, H.K.2    Lee, J.Y.3    Kim, D.J.4    Park, S.5    Lee, C.6    Lee, C.U.7
  • 227
    • 80052714655 scopus 로고    scopus 로고
    • CX3CR1 protein signaling modulates microglial activation and protects against plaque-independent cognitive deficits in a mouse model of Alzheimer disease
    • [CrossRef] [PubMed]
    • Cho, S.H.; Sun, B.; Zhou, Y.; Kauppinen, T.M.; Halabisky, B.; Wes, P.; Ransohoff, R.M.; Gan, L. CX3CR1 protein signaling modulates microglial activation and protects against plaque-independent cognitive deficits in a mouse model of Alzheimer disease. J. Biol. Chem. 2011, 286, 32713-32722. [CrossRef] [PubMed]
    • (2011) J. Biol. Chem , vol.286 , pp. 32713-32722
    • Cho, S.H.1    Sun, B.2    Zhou, Y.3    Kauppinen, T.M.4    Halabisky, B.5    Wes, P.6    Ransohoff, R.M.7    Gan, L.8
  • 228
    • 34547680192 scopus 로고    scopus 로고
    • CD200 ligand receptor interaction modulates microglial activation in vivo and in vitro: A role for IL-4
    • [CrossRef] [PubMed]
    • Lyons, A.; Downer, E.J.; Crotty, S.; Nolan, Y.M.; Mills, K.H.; Lynch, M.A. CD200 ligand receptor interaction modulates microglial activation in vivo and in vitro: A role for IL-4. J. Neurosci. 2007, 27, 8309-8313. [CrossRef] [PubMed]
    • (2007) J. Neurosci , vol.27 , pp. 8309-8313
    • Lyons, A.1    Downer, E.J.2    Crotty, S.3    Nolan, Y.M.4    Mills, K.H.5    Lynch, M.A.6
  • 229
    • 84906252683 scopus 로고    scopus 로고
    • Brain innate immunity in the regulation of neuroinflammation: Therapeutic strategies by modulating CD200-CD200R interaction involve the cannabinoid system
    • [CrossRef] [PubMed]
    • Hernangomez, M.; Carrillo-Salinas, F.J.; Mecha, M.; Correa, F.; Mestre, L.; Loria, F.; Feliu, A.; Docagne, F.; Guaza, C. Brain innate immunity in the regulation of neuroinflammation: Therapeutic strategies by modulating CD200-CD200R interaction involve the cannabinoid system. Curr. Pharm. Des. 2014, 20,4707-4722. [CrossRef] [PubMed]
    • (2014) Curr. Pharm. Des , vol.20 , pp. 4707-4722
    • Hernangomez, M.1    Carrillo-Salinas, F.J.2    Mecha, M.3    Correa, F.4    Mestre, L.5    Loria, F.6    Feliu, A.7    Docagne, F.8    Guaza, C.9
  • 230
    • 84861993323 scopus 로고    scopus 로고
    • CD200 fusion protein decreases microglial activation in the hippocampus of aged rats
    • [CrossRef] [PubMed]
    • Cox, F.F.; Carney, D.; Miller, A.M.; Lynch, M.A. CD200 fusion protein decreases microglial activation in the hippocampus of aged rats. Brain Behav. Immun. 2012, 26, 789-796. [CrossRef] [PubMed]
    • (2012) Brain Behav. Immun , vol.26 , pp. 789-796
    • Cox, F.F.1    Carney, D.2    Miller, A.M.3    Lynch, M.A.4
  • 231
    • 33646006075 scopus 로고    scopus 로고
    • MRNA up-regulation of MHC II and pivotal pro-inflammatory genes in normal brain aging
    • [CrossRef] [PubMed]
    • Frank, M.G.; Barrientos, R.M.; Biedenkapp, J.C.; Rudy, J.W.; Watkins, L.R.; Maier, S.F. mRNA up-regulation of MHC II and pivotal pro-inflammatory genes in normal brain aging. Neurobiol. Aging 2006, 27, 717-722. [CrossRef] [PubMed]
    • (2006) Neurobiol. Aging , vol.27 , pp. 717-722
    • Frank, M.G.1    Barrientos, R.M.2    Biedenkapp, J.C.3    Rudy, J.W.4    Watkins, L.R.5    Maier, S.F.6
  • 232
    • 57449100832 scopus 로고    scopus 로고
    • Decreased expression of CD200 and CD200 receptor in Alzheimer’s disease: A potential mechanism leading to chronic inflammation
    • [CrossRef] [PubMed]
    • Walker, D.G.; Dalsing-Hernandez, J.E.; Campbell, N.A.; Lue, L.F. Decreased expression of CD200 and CD200 receptor in Alzheimer’s disease: A potential mechanism leading to chronic inflammation. Exp. Neurol. 2009, 215, 5-19. [CrossRef] [PubMed]
    • (2009) Exp. Neurol , vol.215 , pp. 5-19
    • Walker, D.G.1    Dalsing-Hernandez, J.E.2    Campbell, N.A.3    Lue, L.F.4
  • 233
    • 80053390173 scopus 로고    scopus 로고
    • Long term potentiation is impaired in membrane glycoprotein CD200-deficient mice: A role for Toll-like receptor activation
    • [CrossRef] [PubMed]
    • Costello, D.A.; Lyons, A.; Denieffe, S.; Browne, T.C.; Cox, F.F.; Lynch, M.A. Long term potentiation is impaired in membrane glycoprotein CD200-deficient mice: A role for Toll-like receptor activation. J. Biol. Chem. 2011, 286, 34722-34732. [CrossRef] [PubMed]
    • (2011) J. Biol. Chem , vol.286 , pp. 34722-34732
    • Costello, D.A.1    Lyons, A.2    Denieffe, S.3    Browne, T.C.4    Cox, F.F.5    Lynch, M.A.6
  • 234
    • 77956393132 scopus 로고    scopus 로고
    • Involvement of Fc receptors in disorders of the central nervoussystem
    • [CrossRef] [PubMed]
    • Okun, E.; Mattson, M.P.; Arumugam, T.V. Involvement of Fc receptors in disorders of the central nervoussystem. Neuromol. Med. 2010, 12, 164-178. [CrossRef] [PubMed]
    • (2010) Neuromol. Med , vol.12 , pp. 164-178
    • Okun, E.1    Mattson, M.P.2    Arumugam, T.V.3
  • 235
    • 84893657950 scopus 로고    scopus 로고
    • The function of Fc? Receptors in dendritic cells and macrophages
    • [CrossRef] [PubMed]
    • Guilliams, M.; Bruhns, P.; Saeys, Y.; Hammad, H.; Lambrecht, B.N. The function of Fc? receptors in dendritic cells and macrophages. Nat. Rev. Immunol. 2014, 14, 94-108. [CrossRef] [PubMed]
    • (2014) Nat. Rev. Immunol , vol.14 , pp. 94-108
    • Guilliams, M.1    Bruhns, P.2    Saeys, Y.3    Hammad, H.4    Lambrecht, B.N.5
  • 236
    • 73549091923 scopus 로고    scopus 로고
    • Signal regulatory protein-ß1: A microglial modulator of phagocytosis in Alzheimer’s disease
    • [CrossRef] [PubMed]
    • Gaikwad, S.; Larionov, S.; Wang, Y.; Dannenberg, H.; Matozaki, T.; Monsonego, A.; Thal, D.R.; Neumann, H. Signal regulatory protein-ß1: A microglial modulator of phagocytosis in Alzheimer’s disease. Am. J. Pathol. 2009, 175, 2528-2539. [CrossRef] [PubMed]
    • (2009) Am. J. Pathol , vol.175 , pp. 2528-2539
    • Gaikwad, S.1    Larionov, S.2    Wang, Y.3    Dannenberg, H.4    Matozaki, T.5    Monsonego, A.6    Thal, D.R.7    Neumann, H.8
  • 237
    • 84897935089 scopus 로고    scopus 로고
    • Microglia receptors and their implications in the response to amyloid ß for Alzheimer’s disease pathogenesis
    • [CrossRef] [PubMed]
    • Doens, D.; Fernandez, P.L. Microglia receptors and their implications in the response to amyloid ß for Alzheimer’s disease pathogenesis. J. Neuroinflamm. 2014, 11, 48. [CrossRef] [PubMed]
    • (2014) J. Neuroinflamm , vol.11 , pp. 48
    • Doens, D.1    Fernandez, P.L.2
  • 238
    • 1842367306 scopus 로고    scopus 로고
    • Degradation of amyloid ß-protein by a metalloprotease secreted by microglia and other neural and non-neural cells
    • [CrossRef] [PubMed]
    • Qiu, W.Q.; Ye, Z.; Kholodenko, D.; Seubert, P.; Selkoe, D.J. Degradation of amyloid ß-protein by a metalloprotease secreted by microglia and other neural and non-neural cells. J. Biol. Chem. 1997, 272, 6641-6646. [CrossRef] [PubMed]
    • (1997) J. Biol. Chem , vol.272 , pp. 6641-6646
    • Qiu, W.Q.1    Ye, Z.2    Kholodenko, D.3    Seubert, P.4    Selkoe, D.J.5
  • 240
    • 0036077536 scopus 로고    scopus 로고
    • ß-amyloid catabolism: Roles for neprilysin (NEP) and other metallopeptidases
    • [CrossRef] [PubMed]
    • Carson, J.A.; Turner, A.J. ß-amyloid catabolism: Roles for neprilysin (NEP) and other metallopeptidases? J. Neurochem. 2002, 81, 1-8. [CrossRef] [PubMed]
    • (2002) J. Neurochem , vol.81 , pp. 1-8
    • Carson, J.A.1    Turner, A.J.2
  • 241
    • 48749086761 scopus 로고    scopus 로고
    • Possible involvement of cathepsin B released by microglia in methylmercury-induced cerebellar pathological changes in the adult rat
    • [CrossRef] [PubMed]
    • Sakamoto, M.; Miyamoto, K.; Wu, Z.; Nakanishi, H. Possible involvement of cathepsin B released by microglia in methylmercury-induced cerebellar pathological changes in the adult rat. Neurosci. Lett. 2008, 442, 292-296. [CrossRef] [PubMed]
    • (2008) Neurosci. Lett , vol.442 , pp. 292-296
    • Sakamoto, M.1    Miyamoto, K.2    Wu, Z.3    Nakanishi, H.4
  • 242
    • 84897954445 scopus 로고    scopus 로고
    • Microglial dysfunction in brain aging and Alzheimer’s disease
    • [CrossRef] [PubMed]
    • Mosher, K.I.; Wyss-Coray, T. Microglial dysfunction in brain aging and Alzheimer’s disease. Biochem. Pharmacol. 2014, 88, 594-604. [CrossRef] [PubMed]
    • (2014) Biochem. Pharmacol , vol.88 , pp. 594-604
    • Mosher, K.I.1    Wyss-Coray, T.2
  • 243
  • 244
    • 80052181853 scopus 로고    scopus 로고
    • Deregulated inflammasome signaling in disease
    • [CrossRef] [PubMed]
    • Lamkanfi, M.; Vande Walle, L.; Kanneganti, T.D. Deregulated inflammasome signaling in disease. Immunol. Rev. 2011, 243, 163-173. [CrossRef] [PubMed]
    • (2011) Immunol. Rev , vol.243 , pp. 163-173
    • Lamkanfi, M.1    Vande Walle, L.2    Kanneganti, T.D.3
  • 245
    • 24744467358 scopus 로고    scopus 로고
    • Microglial phagocytosis induced by fibrillar ß-amyloid and IgGs are differentially regulated by proinflammatory cytokines
    • [CrossRef] [PubMed]
    • Koenigsknecht-Talboo, J.; Landreth, G.E. Microglial phagocytosis induced by fibrillar ß-amyloid and IgGs are differentially regulated by proinflammatory cytokines. J. Neurosci. 2005, 25, 8240-8249. [CrossRef] [PubMed]
    • (2005) J. Neurosci , vol.25 , pp. 8240-8249
    • Koenigsknecht-Talboo, J.1    Landreth, G.E.2
  • 248
    • 84898879651 scopus 로고    scopus 로고
    • Human astrocytes: Secretome profiles of cytokines and chemokines
    • [CrossRef] [PubMed]
    • Choi, S.S.; Lee, H.J.; Lim, I.; Satoh, J.; Kim, S.U. Human astrocytes: Secretome profiles of cytokines and chemokines. PLoS ONE 2014, 9, e92325. [CrossRef] [PubMed]
    • (2014) Plos ONE , vol.9
    • Choi, S.S.1    Lee, H.J.2    Lim, I.3    Satoh, J.4    Kim, S.U.5
  • 249
    • 0035783762 scopus 로고    scopus 로고
    • Glial diffusion barriers during aging and pathological states
    • [PubMed]
    • Sykova, E. Glial diffusion barriers during aging and pathological states. Prog. Brain Res. 2001, 132, 339-363.[PubMed]
    • (2001) Prog. Brain Res , vol.132 , pp. 339-363
    • Sykova, E.1
  • 250
    • 84907223865 scopus 로고    scopus 로고
    • Astrocytes and extracellular matrix in extrasynaptic volume transmission. Philos
    • [CrossRef] [PubMed]
    • Vargova, L.; Sykova, E. Astrocytes and extracellular matrix in extrasynaptic volume transmission. Philos. Trans. R. Soc. Lond. B 2014, 369. [CrossRef] [PubMed]
    • (2014) Trans. R. Soc. Lond. B , pp. 369
    • Vargova, L.1    Sykova, E.2
  • 251
    • 1842587600 scopus 로고    scopus 로고
    • Astrocyte influences on ischemic neuronal death
    • [CrossRef] [PubMed]
    • Swanson, R.A.; Ying, W.; Kauppinen, T.M. Astrocyte influences on ischemic neuronal death. Curr. Mol. Med. 2004, 4, 193-205. [CrossRef] [PubMed]
    • (2004) Curr. Mol. Med , vol.4 , pp. 193-205
    • Swanson, R.A.1    Ying, W.2    Kauppinen, T.M.3
  • 252
    • 34547381007 scopus 로고    scopus 로고
    • The role of astrocytes and complement system in neural plasticity
    • [PubMed]
    • Pekny, M.; Wilhelmsson, U.; Bogestal, Y.R.; Pekna, M. The role of astrocytes and complement system in neural plasticity. Int. Rev. Neurobiol. 2007, 82, 95-111. [PubMed]
    • (2007) Int. Rev. Neurobiol , vol.82 , pp. 95-111
    • Pekny, M.1    Wilhelmsson, U.2    Bogestal, Y.R.3    Pekna, M.4
  • 253
    • 0035831189 scopus 로고    scopus 로고
    • ß-amyloid-induced glial expression of both pro- and anti-inflammatory cytokines in cerebral cortex of aged transgenic Tg2576 mice with Alzheimer plaque pathology
    • [CrossRef]
    • Apelt, J.; Schliebs, R. ß-amyloid-induced glial expression of both pro- and anti-inflammatory cytokines in cerebral cortex of aged transgenic Tg2576 mice with Alzheimer plaque pathology. Brain Res. 2001, 894, 21-30. [CrossRef]
    • (2001) Brain Res , vol.894 , pp. 21-30
    • Apelt, J.1    Schliebs, R.2
  • 254
    • 84875414010 scopus 로고    scopus 로고
    • Aquaporin water channels in the nervous system
    • [CrossRef] [PubMed]
    • Papadopoulos, M.C.; Verkman, A.S. Aquaporin water channels in the nervous system. Nat. Rev. Neurosci. 2013, 14, 265-277. [CrossRef] [PubMed]
    • (2013) Nat. Rev. Neurosci , vol.14 , pp. 265-277
    • Papadopoulos, M.C.1    Verkman, A.S.2
  • 255
    • 0043172479 scopus 로고    scopus 로고
    • The glutamatergic system and Alzheimer’s disease: Therapeutic implications
    • [CrossRef] [PubMed]
    • Butterfield, D.A.; Pocernich, C.B. The glutamatergic system and Alzheimer’s disease: Therapeutic implications. CNS Drugs 2003, 17, 641-652. [CrossRef] [PubMed]
    • (2003) CNS Drugs , vol.17 , pp. 641-652
    • Butterfield, D.A.1    Pocernich, C.B.2
  • 256
    • 0034925118 scopus 로고    scopus 로고
    • The glial glutamate transporter, GLT-1, is oxidatively modified by 4-hydroxy-2-nonenal in the Alzheimer’s disease brain: The role of Aß1-42
    • [CrossRef] [PubMed]
    • Lauderback, C.M.; Hackett, J.M.; Huang, F.F.; Keller, J.N.; Szweda, L.I.; Markesbery, W.R.; Butterfield, D.A. The glial glutamate transporter, GLT-1, is oxidatively modified by 4-hydroxy-2-nonenal in the Alzheimer’s disease brain: The role of Aß1-42. J. Neurochem. 2001, 78, 413-416. [CrossRef] [PubMed]
    • (2001) J. Neurochem , vol.78 , pp. 413-416
    • Lauderback, C.M.1    Hackett, J.M.2    Huang, F.F.3    Keller, J.N.4    Szweda, L.I.5    Markesbery, W.R.6    Butterfield, D.A.7
  • 257
    • 0029811226 scopus 로고    scopus 로고
    • Deficient glutamate transport is associated with neurodegeneration in Alzheimer’s disease
    • [CrossRef] [PubMed]
    • Masliah, E.; Alford, M.; de Teresa, R.; Mallory, M.; Hansen, L. Deficient glutamate transport is associated with neurodegeneration in Alzheimer’s disease. Ann. Neurol. 1996, 40, 759-766. [CrossRef] [PubMed]
    • (1996) Ann. Neurol , vol.40 , pp. 759-766
    • Masliah, E.1    Alford, M.2    De Teresa, R.3    Mallory, M.4    Hansen, L.5
  • 258
    • 27744437016 scopus 로고    scopus 로고
    • Autoantigen specific T cells inhibit glutamate uptake in astrocytes by decreasing expression of astrocytic glutamate transporter GLAST: A mechanism mediated by tumor necrosis factor-a
    • [CrossRef] [PubMed]
    • Korn, T.; Magnus, T.; Jung, S. Autoantigen specific T cells inhibit glutamate uptake in astrocytes by decreasing expression of astrocytic glutamate transporter GLAST: A mechanism mediated by tumor necrosis factor-a. FASEB J. 2005, 19, 1878-1880. [CrossRef] [PubMed]
    • (2005) FASEB J , vol.19 , pp. 1878-1880
    • Korn, T.1    Magnus, T.2    Jung, S.3
  • 259
    • 77249122078 scopus 로고    scopus 로고
    • Glutamine synthetase down-regulation reduces astrocyte protection against glutamate excitotoxicity to neurons
    • [CrossRef] [PubMed]
    • Zou, J.; Wang, Y.X.; Dou, F.F.; Lu, H.Z.; Ma, Z.W.; Lu, P.H.; Xu, X.M. Glutamine synthetase down-regulation reduces astrocyte protection against glutamate excitotoxicity to neurons. Neurochem. Int. 2010, 56, 577-584. [CrossRef] [PubMed]
    • (2010) Neurochem. Int , vol.56 , pp. 577-584
    • Zou, J.1    Wang, Y.X.2    Dou, F.F.3    Lu, H.Z.4    Ma, Z.W.5    Lu, P.H.6    Xu, X.M.7
  • 260
    • 79960887452 scopus 로고    scopus 로고
    • Age-dependent decrease in glutamine synthetase expression in the hippocampal astroglia of the triple transgenic Alzheimer’s disease mouse model: Mechanism of deficient glutamategic transmission?
    • [CrossRef] [PubMed]
    • Olabarria, M.; Noristani, H.N.; Verkhratsky, A.; Rodriguez, J.J. Age-dependent decrease in glutamine synthetase expression in the hippocampal astroglia of the triple transgenic Alzheimer’s disease mouse model: Mechanism of deficient glutamategic transmission? Mol. Neurodegener. 2011, 6, 55. [CrossRef] [PubMed]
    • (2011) Mol. Neurodegener , vol.6 , pp. 55
    • Olabarria, M.1    Noristani, H.N.2    Verkhratsky, A.3    Rodriguez, J.J.4
  • 261
    • 84864359502 scopus 로고    scopus 로고
    • Astrocytic cytoskeletal atrophy in the medial prefrontal cortex of a triple transgenic mouse model of Alzheimer’s disease
    • [CrossRef] [PubMed]
    • Kulijewicz-Nawrot, M.; Verkhratsky, A.; Chvatal, A.; Sykova, E.; Rodriguez, J.J. Astrocytic cytoskeletal atrophy in the medial prefrontal cortex of a triple transgenic mouse model of Alzheimer’s disease. J. Anat. 2012, 221, 252-262. [CrossRef] [PubMed]
    • (2012) J. Anat , vol.221 , pp. 252-262
    • Kulijewicz-Nawrot, M.1    Verkhratsky, A.2    Chvatal, A.3    Sykova, E.4    Rodriguez, J.J.5
  • 263
    • 33751112725 scopus 로고    scopus 로고
    • Matrix metalloproteinases expressed by astrocytes mediate extracellular amyloid-ß peptide catabolism
    • [CrossRef] [PubMed]
    • Yin, K.J.; Cirrito, J.R.; Yan, P.; Hu, X.; Xiao, Q.; Pan, X.; Bateman, R.; Song, H.; Hsu, F.F.; Turk, J.; et al. Matrix metalloproteinases expressed by astrocytes mediate extracellular amyloid-ß peptide catabolism. J. Neurosci. 2006, 26, 10939-10948. [CrossRef] [PubMed]
    • (2006) J. Neurosci , vol.26 , pp. 10939-10948
    • Yin, K.J.1    Cirrito, J.R.2    Yan, P.3    Hu, X.4    Xiao, Q.5    Pan, X.6    Bateman, R.7    Song, H.8    Hsu, F.F.9    Turk, J.10
  • 264
    • 0042827324 scopus 로고    scopus 로고
    • Human neprilysin is capable of degrading amyloid ß peptide not only in the monomeric form but also the pathological oligomeric form
    • [CrossRef]
    • Kanemitsu, H.; Tomiyama, T.; Mori, H. Human neprilysin is capable of degrading amyloid ß peptide not only in the monomeric form but also the pathological oligomeric form. Neurosci. Lett. 2003, 350, 113-116. [CrossRef]
    • (2003) Neurosci. Lett , vol.350 , pp. 113-116
    • Kanemitsu, H.1    Tomiyama, T.2    Mori, H.3
  • 265
    • 27844442718 scopus 로고    scopus 로고
    • Aß-degrading enzymes: Modulators of Alzheimer’s disease pathogenesis and targets for therapeutic intervention
    • [CrossRef] [PubMed]
    • Eckman, E.A.; Eckman, C.B. Aß-degrading enzymes: Modulators of Alzheimer’s disease pathogenesis and targets for therapeutic intervention. Biochem. Soc. Trans. 2005, 33, 1101-1105. [CrossRef] [PubMed]
    • (2005) Biochem. Soc. Trans , vol.33 , pp. 1101-1105
    • Eckman, E.A.1    Eckman, C.B.2
  • 266
    • 0004982358 scopus 로고
    • Apolipoprotein E mRNA is abundant in the brain and adrenals, as well as in the liver, and is present in other peripheral tissues of rats and marmosets
    • [CrossRef] [PubMed]
    • Elshourbagy, N.A.; Liao, W.S.; Mahley, R.W.; Taylor, J.M. Apolipoprotein E mRNA is abundant in the brain and adrenals, as well as in the liver, and is present in other peripheral tissues of rats and marmosets. Proc. Natl. Acad. Sci. USA 1985, 82, 203-207. [CrossRef] [PubMed]
    • (1985) Proc. Natl. Acad. Sci. USA , vol.82 , pp. 203-207
    • Elshourbagy, N.A.1    Liao, W.S.2    Mahley, R.W.3    Taylor, J.M.4
  • 267
    • 0023080585 scopus 로고
    • Astrocytes synthesize apolipoprotein E and metabolize apolipoprotein E-containing lipoproteins
    • [CrossRef]
    • Pitas, R.E.; Boyles, J.K.; Lee, S.H.; Foss, D.; Mahley, R.W. Astrocytes synthesize apolipoprotein E and metabolize apolipoprotein E-containing lipoproteins. Biochim. Biophys. Acta 1987, 917, 148-161. [CrossRef]
    • (1987) Biochim. Biophys. Acta , vol.917 , pp. 148-161
    • Pitas, R.E.1    Boyles, J.K.2    Lee, S.H.3    Foss, D.4    Mahley, R.W.5
  • 268
    • 0030582979 scopus 로고    scopus 로고
    • Expression of apolipoprotein E mRNA in rat microglia
    • [CrossRef]
    • Nakai, M.; Kawamata, T.; Taniguchi, T.; Maeda, K.; Tanaka, C. Expression of apolipoprotein E mRNA in rat microglia. Neurosci. Lett. 1996, 211, 41-44. [CrossRef]
    • (1996) Neurosci. Lett , vol.211 , pp. 41-44
    • Nakai, M.1    Kawamata, T.2    Taniguchi, T.3    Maeda, K.4    Tanaka, C.5
  • 269
    • 33746311471 scopus 로고    scopus 로고
    • Apolipoprotein E structure: Insights into function
    • [CrossRef] [PubMed]
    • Hatters, D.M.; Peters-Libeu, C.A.; Weisgraber, K.H. Apolipoprotein E structure: Insights into function. Trends Biochem. Sci. 2006, 31, 445-454. [CrossRef] [PubMed]
    • (2006) Trends Biochem. Sci , vol.31 , pp. 445-454
    • Hatters, D.M.1    Peters-Libeu, C.A.2    Weisgraber, K.H.3
  • 270
    • 84896726518 scopus 로고    scopus 로고
    • ApoE and Aß in Alzheimer’s disease: Accidental encounters or partners?
    • [CrossRef] [PubMed]
    • Kanekiyo, T.; Xu, H.; Bu, G. ApoE and Aß in Alzheimer’s disease: Accidental encounters or partners? Neuron 2014, 81, 740-754. [CrossRef] [PubMed]
    • (2014) Neuron , vol.81 , pp. 740-754
    • Kanekiyo, T.1    Xu, H.2    Bu, G.3
  • 271
    • 84863504256 scopus 로고    scopus 로고
    • Apolipoprotein E and apolipoprotein E receptors: Normal biology and role in Alzheimer disease
    • [CrossRef] [PubMed]
    • Holtzman, D.M.; Herz, J.; Bu, G. Apolipoprotein E and apolipoprotein E receptors: Normal biology and role in Alzheimer disease. Cold Spring Harb. Perspect. Med. 2012, 2. [CrossRef] [PubMed]
    • (2012) Cold Spring Harb. Perspect. Med , pp. 2
    • Holtzman, D.M.1    Herz, J.2    Bu, G.3
  • 272
    • 84859980871 scopus 로고    scopus 로고
    • Low-density lipoprotein receptor represent an apolipoprotein E-independent pathway of Aß uptake and degradation by astrocytes
    • [CrossRef] [PubMed]
    • Basak, J.M.; Verghese, P.B.; Yoon, H.; Kim, J.; Hotzman, D.M. Low-density lipoprotein receptor represent an apolipoprotein E-independent pathway of Aß uptake and degradation by astrocytes. J. Biol. Chem. 2012, 287, 13959-13971. [CrossRef] [PubMed]
    • (2012) J. Biol. Chem , vol.287 , pp. 13959-13971
    • Basak, J.M.1    Verghese, P.B.2    Yoon, H.3    Kim, J.4    Hotzman, D.M.5
  • 273
  • 275
    • 33749521100 scopus 로고    scopus 로고
    • Intraneuronal ß-amyloid aggregates, neurodegeneration, and neuron loss in transgenic mice with five familial Alzheimer’s disease mutations: Potential factors in amyloid plaque formation
    • [CrossRef] [PubMed]
    • Oakley, H.; Cole, S.L.; Logan, S.; Maus, E.; Shao, P.; Craft, J.; Guillozet-Bongaarts, A.; Ohno, M.; Disterhoft, J.; van Eldik, L.; et al. Intraneuronal ß-amyloid aggregates, neurodegeneration, and neuron loss in transgenic mice with five familial Alzheimer’s disease mutations: Potential factors in amyloid plaque formation. J. Neurosci. 2006, 26, 10129-10140. [CrossRef] [PubMed]
    • (2006) J. Neurosci , vol.26 , pp. 10129-10140
    • Oakley, H.1    Cole, S.L.2    Logan, S.3    Maus, E.4    Shao, P.5    Craft, J.6    Guillozet-Bongaarts, A.7    Ohno, M.8    Disterhoft, J.9    Van Eldik, L.10
  • 276
    • 0344845132 scopus 로고    scopus 로고
    • Amyloid deposition precedes tangle formation in a triple transgenic model of Alzheimer’s disease. Neurobiol
    • [CrossRef] [PubMed]
    • Oddo, S.; Caccamo, A.; Kitazawa, M.; Tseng, B.P.; LaFerla, F.M. Amyloid deposition precedes tangle formation in a triple transgenic model of Alzheimer’s disease. Neurobiol. Aging 2003, 24, 1063-1070. [CrossRef] [PubMed]
    • (2003) Aging , vol.24 , pp. 1063-1070
    • Oddo, S.1    Caccamo, A.2    Kitazawa, M.3    Tseng, B.P.4    Laferla, F.M.5
  • 277
    • 41149176407 scopus 로고    scopus 로고
    • Lack of pathology in a triple transgenic mouse model of Alzheimer’s disease after overexpression of the anti-apoptotic protein Bcl-2
    • [CrossRef] [PubMed]
    • Rohn, T.T.; Vyas, V.; Hernandez-Estrada, T.; Nichol, K.E.; Christie, L.A.; Head, E. Lack of pathology in a triple transgenic mouse model of Alzheimer’s disease after overexpression of the anti-apoptotic protein Bcl-2. J. Neurosci. 2008, 28, 3051-3059. [CrossRef] [PubMed]
    • (2008) J. Neurosci , vol.28 , pp. 3051-3059
    • Rohn, T.T.1    Vyas, V.2    Hernandez-Estrada, T.3    Nichol, K.E.4    Christie, L.A.5    Head, E.6
  • 279
    • 0030805736 scopus 로고    scopus 로고
    • Behavioural performance in three substrains of mouse strain 129
    • [CrossRef]
    • Montkowski, A.; Poetting, M.; Mederer, A.; Holsboer, F. Behavioural performance in three substrains of mouse strain 129. Brain Res. 1997, 762, 12-18. [CrossRef]
    • (1997) Brain Res , vol.762 , pp. 12-18
    • Montkowski, A.1    Poetting, M.2    Mederer, A.3    Holsboer, F.4
  • 280
    • 0033620883 scopus 로고    scopus 로고
    • A behavioral and neuroanatomical assessment of an inbred substrain of 129 mice with behavioral comparisons to C57BL/6J mice
    • [CrossRef]
    • Balogh, S.A.; McDowell, C.S.; Stavnezer, A.J.; Denenberg, V.H. A behavioral and neuroanatomical assessment of an inbred substrain of 129 mice with behavioral comparisons to C57BL/6J mice. Brain Res.1999, 836, 38-48. [CrossRef]
    • (1999) Brain Res , vol.836 , pp. 38-48
    • Balogh, S.A.1    McDowell, C.S.2    Stavnezer, A.J.3    Denenberg, V.H.4
  • 281
    • 0029940557 scopus 로고    scopus 로고
    • Is it the mutation or the background genotype?
    • Gerlai, R. Gene-targeting studies of mammalian behavior, [CrossRef]
    • Gerlai, R. Gene-targeting studies of mammalian behavior: Is it the mutation or the background genotype? Trends Neurosci. 1996, 19, 177-181. [CrossRef]
    • (1996) Trends Neurosci , vol.19 , pp. 177-181
  • 282
    • 0038025431 scopus 로고    scopus 로고
    • Assessing the effects of the 129/Sc genetic background on swimming navigation learning in transgenic mutants: A study using mice with a modified ß-amyloid precursor protein gene
    • [CrossRef]
    • Wolfer, D.P.; Muller, U.; Stagliar, M.; Lipp, H.P. Assessing the effects of the 129/Sc genetic background on swimming navigation learning in transgenic mutants: A study using mice with a modified ß-amyloid precursor protein gene. Brain Res. 1997, 771, 1-13. [CrossRef]
    • (1997) Brain Res , vol.771 , pp. 1-13
    • Wolfer, D.P.1    Muller, U.2    Stagliar, M.3    Lipp, H.P.4
  • 283
    • 62349123068 scopus 로고    scopus 로고
    • Senescence-accelerated mouse (SAM) with special references to neurodegeneration models, SAMP8 and SAMP10 mice
    • [CrossRef] [PubMed]
    • Takeda, T. Senescence-accelerated mouse (SAM) with special references to neurodegeneration models, SAMP8 and SAMP10 mice. Neurochem. Res. 2009, 34, 639-659. [CrossRef] [PubMed]
    • (2009) Neurochem. Res , vol.34 , pp. 639-659
    • Takeda, T.1
  • 284
    • 79956324801 scopus 로고    scopus 로고
    • Age-related autophagy alterations in the brain of senescence accelerated mouse prone 8 (SAMP8) mice
    • [CrossRef] [PubMed]
    • Ma, Q.; Qiang, J.; Gu, P.; Wang, Y.; Geng, Y.; Wang, M. Age-related autophagy alterations in the brain of senescence accelerated mouse prone 8 (SAMP8) mice. Exp. Gerontol. 2011, 46, 533-541. [CrossRef] [PubMed]
    • (2011) Exp. Gerontol , vol.46 , pp. 533-541
    • Ma, Q.1    Qiang, J.2    Gu, P.3    Wang, Y.4    Geng, Y.5    Wang, M.6
  • 286
    • 84857125516 scopus 로고    scopus 로고
    • The SAMP8 mouse: A model to develop therapeutic interventions for Alzheimer’s disease
    • [CrossRef] [PubMed]
    • Morley, J.E.; Farr, S.A.; Kumar, V.B.; Armbrecht, H.J. The SAMP8 mouse: A model to develop therapeutic interventions for Alzheimer’s disease. Curr. Pharm. Des. 2012, 18, 1123-1130. [CrossRef] [PubMed]
    • (2012) Curr. Pharm. Des , vol.18 , pp. 1123-1130
    • Morley, J.E.1    Farr, S.A.2    Kumar, V.B.3    Armbrecht, H.J.4
  • 287
    • 70349154192 scopus 로고    scopus 로고
    • Amelioration of cognitive ability in senescence-accelerated mouse prone 8 (SAMP8) by intra-bone marrow-bone marrow transplantation
    • [CrossRef] [PubMed]
    • Li, M.; Inaba, M.; Guo, K.; Abraham, N.G.; Ikehara, S. Amelioration of cognitive ability in senescence-accelerated mouse prone 8 (SAMP8) by intra-bone marrow-bone marrow transplantation. Neurosci. Lett. 2009, 465, 36-40. [CrossRef] [PubMed]
    • (2009) Neurosci. Lett , vol.465 , pp. 36-40
    • Li, M.1    Inaba, M.2    Guo, K.3    Abraham, N.G.4    Ikehara, S.5
  • 289
    • 84855805035 scopus 로고    scopus 로고
    • Human umbilical cord blood-derived mesenchymal stem cells improve neuropathology and cognitive impairment in an Alzheimer’s disease mouse model through modulation of neuroinflammation
    • [CrossRef] [PubMed]
    • Lee, H.J.; Lee, J.K.; Lee, H.; Carter, J.E.; Chang, J.W.; Oh, W.; Yang, Y.S.; Suh, J.G.; Lee, B.H.; Jin, H.K.; et al. Human umbilical cord blood-derived mesenchymal stem cells improve neuropathology and cognitive impairment in an Alzheimer’s disease mouse model through modulation of neuroinflammation. Neurobiol. Aging 2012, 33, 588-602. [CrossRef] [PubMed]
    • (2012) Neurobiol. Aging , vol.33 , pp. 588-602
    • Lee, H.J.1    Lee, J.K.2    Lee, H.3    Carter, J.E.4    Chang, J.W.5    Oh, W.6    Yang, Y.S.7    Suh, J.G.8    Lee, B.H.9    Jin, H.K.10
  • 290
    • 84868136186 scopus 로고    scopus 로고
    • Mesenchymal stem cells augment neurogenesis in the subventricular zone and enhance differentiation of neural precursor cells into dopaminergic neurons in the substantia nigra of a parkinsonian model
    • [CrossRef] [PubMed]
    • Park, H.J.; Shin, J.Y.; Lee, B.R.; Kim, H.O.; Lee, P.H. Mesenchymal stem cells augment neurogenesis in the subventricular zone and enhance differentiation of neural precursor cells into dopaminergic neurons in the substantia nigra of a parkinsonian model. Cell Transpl. 2012, 21, 1629-1640. [CrossRef] [PubMed]
    • (2012) Cell Transpl , vol.21 , pp. 1629-1640
    • Park, H.J.1    Shin, J.Y.2    Lee, B.R.3    Kim, H.O.4    Lee, P.H.5
  • 292
    • 84912074929 scopus 로고    scopus 로고
    • Intrathecal delivery of mesenchymal stromal cells protects the structure of altered perineuronal nets in SOD1 rats and amends the course of ALS
    • [CrossRef] [PubMed]
    • Forostyak, S.; Homola, A.; Turnovcova, K.; Svitil, P.; Jendelova, P.; Sykova, E. Intrathecal delivery of mesenchymal stromal cells protects the structure of altered perineuronal nets in SOD1 rats and amends the course of ALS. Stem Cells 2014, 32, 3163-3172. [CrossRef] [PubMed]
    • (2014) Stem Cells , vol.32 , pp. 3163-3172
    • Forostyak, S.1    Homola, A.2    Turnovcova, K.3    Svitil, P.4    Jendelova, P.5    Sykova, E.6
  • 295
    • 84876743127 scopus 로고    scopus 로고
    • Mesenchymal stem cells and neuroregeneration in Parkinson’s disease
    • [CrossRef] [PubMed]
    • Glavaski-Joksimovic, A.; Bohn, M.C. Mesenchymal stem cells and neuroregeneration in Parkinson’s disease. Exp. Neurol. 2013, 247, 25-38. [CrossRef] [PubMed]
    • (2013) Exp. Neurol , vol.247 , pp. 25-38
    • Glavaski-Joksimovic, A.1    Bohn, M.C.2
  • 296
    • 33745437684 scopus 로고    scopus 로고
    • Comparative analysis of mesenchymal stem cells from bone marrow, umbilical cord blood, or adipose tissue
    • [CrossRef] [PubMed]
    • Kern, S.; Eichler, H.; Stoeve, J.; Kluter, H.; Bieback, K. Comparative analysis of mesenchymal stem cells from bone marrow, umbilical cord blood, or adipose tissue. Stem Cells 2006, 24, 1294-1301. [CrossRef] [PubMed]
    • (2006) Stem Cells , vol.24 , pp. 1294-1301
    • Kern, S.1    Eichler, H.2    Stoeve, J.3    Kluter, H.4    Bieback, K.5
  • 297
    • 37249086723 scopus 로고    scopus 로고
    • Human bone-marrow-derived mesenchymal stem cells: Biological characteristics and potential role in therapy of degenerative diseases
    • [CrossRef] [PubMed]
    • Kassem, M. Abdallah, B.M. Human bone-marrow-derived mesenchymal stem cells: Biological characteristics and potential role in therapy of degenerative diseases. Cell Tissue Res. 2008, 331, 157-163. [CrossRef] [PubMed]
    • (2008) Cell Tissue Res , vol.331 , pp. 157-163
    • Kassem, M.1    Abdallah, B.M.2
  • 298
    • 4444300615 scopus 로고    scopus 로고
    • Human placenta-derived cells have mesenchymal stem/progenitor cell potential
    • [CrossRef] [PubMed]
    • Fukuchi, Y.; Nakajima, H.; Sugiyama, D.; Hirose, I.; Kitamura, T.; Tsuji, K. Human placenta-derived cells have mesenchymal stem/progenitor cell potential. Stem Cells 2004, 22, 649-658. [CrossRef] [PubMed]
    • (2004) Stem Cells , vol.22 , pp. 649-658
    • Fukuchi, Y.1    Nakajima, H.2    Sugiyama, D.3    Hirose, I.4    Kitamura, T.5    Tsuji, K.6
  • 299
    • 84892987332 scopus 로고    scopus 로고
    • Use of genetically modified mesenchymal stem cells to treat neurodegenerative diseases
    • Wyse, R.D.; Dunbar, G.L.; Rossignol, J. Use of genetically modified mesenchymal stem cells to treat neurodegenerative diseases. Int. J. Mol. Sci. 2014, 15, 1719-1745.
    • (2014) Int. J. Mol. Sci , vol.15 , pp. 1719-1745
    • Wyse, R.D.1    Dunbar, G.L.2    Rossignol, J.3
  • 300
  • 302
    • 64749093912 scopus 로고    scopus 로고
    • Grafting of human bone marrow stromal cells into spinal cord injury: A comparison of delivery methods
    • [CrossRef] [PubMed]
    • Paul, C.; Samdani, A.F.; Betz, R.R.; Fischer, I.; Neuhuber, B. Grafting of human bone marrow stromal cells into spinal cord injury: A comparison of delivery methods. Spine 2009, 34, 328-334. [CrossRef] [PubMed]
    • (2009) Spine , vol.34 , pp. 328-334
    • Paul, C.1    Samdani, A.F.2    Betz, R.R.3    Fischer, I.4    Neuhuber, B.5
  • 304
    • 84891752308 scopus 로고    scopus 로고
    • Placenta-derived mesenchymal stem cells improve memory dysfunction in an Aß1-42-induced mouse model of Alzheimer’s disease
    • [CrossRef] [PubMed]
    • Yun, H.M.; Kim, H.S.; Park, K.R.; Shin, J.M.; Kang, A.R.; Lee, K.; Song, S.; Kim, Y.B.; Han, S.B.; Chung, H.M.; et al. Placenta-derived mesenchymal stem cells improve memory dysfunction in an Aß1-42-induced mouse model of Alzheimer’s disease. Cell Death Dis. 2013, 4, e958. [CrossRef] [PubMed]
    • (2013) Cell Death Dis , vol.4
    • Yun, H.M.1    Kim, H.S.2    Park, K.R.3    Shin, J.M.4    Kang, A.R.5    Lee, K.6    Song, S.7    Kim, Y.B.8    Han, S.B.9    Chung, H.M.10
  • 305
    • 84930439146 scopus 로고    scopus 로고
    • Stem cell transplantation improves aging-related disease
    • [CrossRef] [PubMed]
    • Ikehara, S.; Li, M. Stem cell transplantation improves aging-related disease. Front. Cell Dev. Biol. 2014, 2, 16. [CrossRef] [PubMed]
    • (2014) Front. Cell Dev. Biol , vol.2 , pp. 16
    • Ikehara, S.1    Li, M.2
  • 306
    • 58149333136 scopus 로고    scopus 로고
    • Bone marrow-derived mesenchymal stem cells reduce brain amyloid-ß deposition and accelerate the activation of microglia in an acutely induced Alzheimer’s disease mouse model
    • [CrossRef] [PubMed]
    • Lee, J.K.; Jin, H.K.; Bae, J.S. Bone marrow-derived mesenchymal stem cells reduce brain amyloid-ß deposition and accelerate the activation of microglia in an acutely induced Alzheimer’s disease mouse model. Neurosci. Lett. 2009, 450, 136-141. [CrossRef] [PubMed]
    • (2009) Neurosci. Lett , vol.450 , pp. 136-141
    • Lee, J.K.1    Jin, H.K.2    Bae, J.S.3
  • 307
    • 77149146814 scopus 로고    scopus 로고
    • Intracerebral transplantation of bone marrow-derived mesenchymal stem cells reduces amyloid-ß deposition and rescues memory deficits in Alzheimer’s disease mice by modulation of immune responses
    • [CrossRef] [PubMed]
    • Lee, J.K.; Jin, H.K.; Endo, S.; Schuchman, E.H.; Carter, J.E.; Bae, J.S. Intracerebral transplantation of bone marrow-derived mesenchymal stem cells reduces amyloid-ß deposition and rescues memory deficits in Alzheimer’s disease mice by modulation of immune responses. Stem Cells 2010, 28, 329-343. [CrossRef] [PubMed]
    • (2010) Stem Cells , vol.28 , pp. 329-343
    • Lee, J.K.1    Jin, H.K.2    Endo, S.3    Schuchman, E.H.4    Carter, J.E.5    Bae, J.S.6
  • 308
    • 20444364844 scopus 로고    scopus 로고
    • Activation of microglia by aggregated ß-amyloid or lipopolysaccharide impairs MHC-II expression and renders them cytotoxic whereas IFN-? And IL-4 render them protective
    • [CrossRef] [PubMed]
    • Butovsky, O.; Talpalar, A.E.; Ben-Yaakov, K.; Schwartz, M. Activation of microglia by aggregated ß-amyloid or lipopolysaccharide impairs MHC-II expression and renders them cytotoxic whereas IFN-? and IL-4 render them protective. Mol. Cell. Neurosci. 2005, 29, 381-393. [CrossRef] [PubMed]
    • (2005) Mol. Cell. Neurosci , vol.29 , pp. 381-393
    • Butovsky, O.1    Talpalar, A.E.2    Ben-Yaakov, K.3    Schwartz, M.4
  • 309
    • 33746827434 scopus 로고    scopus 로고
    • Glatiramer acetate fights against Alzheimer’s disease by inducing dendritic-like microglia expression insulin-like growth factor 1
    • [CrossRef] [PubMed]
    • Butovsky, O.; Koronyo-Hamaoui, M.; Kunis, G.; Ophir, E.; Landa, G.; Cohen, H.; Schwartz, M. Glatiramer acetate fights against Alzheimer’s disease by inducing dendritic-like microglia expression insulin-like growth factor 1. Proc. Natl. Acad. Sci. USA 2006, 103, 11784-11789. [CrossRef] [PubMed]
    • (2006) Proc. Natl. Acad. Sci. USA , vol.103 , pp. 11784-11789
    • Butovsky, O.1    Koronyo-Hamaoui, M.2    Kunis, G.3    Ophir, E.4    Landa, G.5    Cohen, H.6    Schwartz, M.7
  • 310
    • 58149326737 scopus 로고    scopus 로고
    • Brown, J.M.; et al. Bone marrow stromal cells attenuate sepsis via prostaglandin E(2)-dependent reprogramming of host macrophages to increase their interleukin-10 production
    • [CrossRef] [PubMed]
    • Nemeth, K.; Leelahavanichkul, A.; Yuen, P.S.; Mayer, B.; Parmelee, A.; Doi, K.; Robey, P.G.; Leelahavanichkul, K.; Koller, B.H.; Brown, J.M.; et al. Bone marrow stromal cells attenuate sepsis via prostaglandin E(2)-dependent reprogramming of host macrophages to increase their interleukin-10 production. Nat. Med. 2009, 15, 42-49. [CrossRef] [PubMed]
    • (2009) Nat. Med , vol.15 , pp. 42-49
    • Nemeth, K.1    Leelahavanichkul, A.2    Yuen, P.S.3    Mayer, B.4    Parmelee, A.5    Doi, K.6    Robey, P.G.7    Leelahavanichkul, K.8    Koller, B.H.9
  • 311
    • 84870374627 scopus 로고    scopus 로고
    • Can stem cells be used to treat or model Alzheimer disease?
    • [CrossRef] [PubMed]
    • Chen, W.W.; Blurton-Jones, M. Can stem cells be used to treat or model Alzheimer disease? Stem Cells 2012, 30, 2612-2618. [CrossRef] [PubMed]
    • (2012) Stem Cells , vol.30 , pp. 2612-2618
    • Chen, W.W.1    Blurton-Jones, M.2
  • 312
    • 84892433295 scopus 로고    scopus 로고
    • Immunobiology of mesenchymal stem cells
    • [CrossRef] [PubMed]
    • Ma, S.; Xie, N.; Li, W.; Yuan, B.; Shi, Y.; Wang, Y. Immunobiology of mesenchymal stem cells. Cell Death Differ. 2014, 21, 216-225. [CrossRef] [PubMed]
    • (2014) Cell Death Differ , vol.21 , pp. 216-225
    • Ma, S.1    Xie, N.2    Li, W.3    Yuan, B.4    Shi, Y.5    Wang, Y.6
  • 313
    • 34249039544 scopus 로고    scopus 로고
    • Transplantation of adult rat spinal cord stem/progenitor cells for spinal cord injury
    • [CrossRef] [PubMed]
    • Parr, A.M.; Kulbatski, I.; Tator, C.H. Transplantation of adult rat spinal cord stem/progenitor cells for spinal cord injury. J. Neurotrauma 2007, 24, 835-845. [CrossRef] [PubMed]
    • (2007) J. Neurotrauma , vol.24 , pp. 835-845
    • Parr, A.M.1    Kulbatski, I.2    Tator, C.H.3
  • 314
    • 29144460856 scopus 로고    scopus 로고
    • Human stem/progenitor cells from bone marrow promote neurogenesis of endogenous neural stem cells in the hippocampus of mice
    • [CrossRef] [PubMed]
    • Munoz, J.R.; Stoutenger, B.R.; Robinson, A.P.; Spees, J.L.; Prockop, D.J. Human stem/progenitor cells from bone marrow promote neurogenesis of endogenous neural stem cells in the hippocampus of mice. Proc. Natl. Acad. Sci. USA 2005, 102, 18171-18176. [CrossRef] [PubMed]
    • (2005) Proc. Natl. Acad. Sci. USA , vol.102 , pp. 18171-18176
    • Munoz, J.R.1    Stoutenger, B.R.2    Robinson, A.P.3    Spees, J.L.4    Prockop, D.J.5
  • 315
    • 79954999836 scopus 로고    scopus 로고
    • Mesenchymal stem cells stimulate endogenous neurogenesis in the subventricular zone of adult mice
    • [CrossRef] [PubMed]
    • Kan, I.; Barhum, Y.; Melamed, E.; Offen, D. Mesenchymal stem cells stimulate endogenous neurogenesis in the subventricular zone of adult mice. Stem Cell Rev. 2011, 7, 404-412. [CrossRef] [PubMed]
    • (2011) Stem Cell Rev , vol.7 , pp. 404-412
    • Kan, I.1    Barhum, Y.2    Melamed, E.3    Offen, D.4
  • 316
    • 84866862490 scopus 로고    scopus 로고
    • The preventive and therapeutic effects of intravenous human adipose-derived stem cells in Alzheimer’s disease mice
    • [CrossRef] [PubMed]
    • Kim, S.; Chang, K.A.; Kim, J.A.; Park, H.G.; Ra, J.C.; Kim, H.S.; Suh, Y.H. The preventive and therapeutic effects of intravenous human adipose-derived stem cells in Alzheimer’s disease mice. PLoS ONE 2012, 7, e45757. [CrossRef] [PubMed]
    • (2012) Plos ONE , vol.7
    • Kim, S.1    Chang, K.A.2    Kim, J.A.3    Park, H.G.4    Ra, J.C.5    Kim, H.S.6    Suh, Y.H.7
  • 317
    • 78650254124 scopus 로고    scopus 로고
    • Mesenchymal stem cells increase hippocampal neurogenesis and counteract depressive-like behavior
    • [CrossRef] [PubMed]
    • Tfilin, M.; Sudai, E.; Merenlender, A.; Gispan, I.; Yadid, G.; Turgeman, G. Mesenchymal stem cells increase hippocampal neurogenesis and counteract depressive-like behavior. Mol. Psychiatry 2010, 15, 1164-1175. [CrossRef] [PubMed]
    • (2010) Mol. Psychiatry , vol.15 , pp. 1164-1175
    • Tfilin, M.1    Sudai, E.2    Merenlender, A.3    Gispan, I.4    Yadid, G.5    Turgeman, G.6
  • 318
    • 78650019181 scopus 로고    scopus 로고
    • Adipose-derived mesenchymal stem cells protect PC12 cells from glutamate excitotoxicity-induced apoptosis by upregulation of XIAP through PI3-K/Akt activation
    • [CrossRef] [PubMed]
    • Lu, S.; Lu, C.; Han, Q.; Li, J.; Du, Z.; Liao, L.; Zhao, R.C. Adipose-derived mesenchymal stem cells protect PC12 cells from glutamate excitotoxicity-induced apoptosis by upregulation of XIAP through PI3-K/Akt activation. Toxicology 2011, 279, 189-195. [CrossRef] [PubMed]
    • (2011) Toxicology , vol.279 , pp. 189-195
    • Lu, S.1    Lu, C.2    Han, Q.3    Li, J.4    Du, Z.5    Liao, L.6    Zhao, R.C.7
  • 319
    • 0028361594 scopus 로고
    • Long-distance neuronal migration in the adult mammalian brain
    • [CrossRef] [PubMed]
    • Lois, C.; Alvarez-Buylla, A. Long-distance neuronal migration in the adult mammalian brain. Science 1994, 264, 1145-1148. [CrossRef] [PubMed]
    • (1994) Science , vol.264 , pp. 1145-1148
    • Lois, C.1    Alvarez-Buylla, A.2
  • 320
    • 0035320035 scopus 로고    scopus 로고
    • A unified hypothesis on the lineage of neuralstem cells
    • [CrossRef] [PubMed]
    • Alvarez-Buylla, A.; Garcia-Verdugo, J.M.; Tramontin, A.D. A unified hypothesis on the lineage of neuralstem cells. Nat. Rev. Neurosci. 2001, 2, 287-293. [CrossRef] [PubMed]
    • (2001) Nat. Rev. Neurosci , vol.2 , pp. 287-293
    • Alvarez-Buylla, A.1    Garcia-Verdugo, J.M.2    Tramontin, A.D.3
  • 321
    • 0035833068 scopus 로고    scopus 로고
    • Adult neurogenesis produces a large pool of new granule cells in the dentate gyrus
    • [CrossRef] [PubMed]
    • Cameron, H.A.; McKay, R.D. Adult neurogenesis produces a large pool of new granule cells in the dentate gyrus. J. Comp. Neurol. 2001, 435, 406-417. [CrossRef] [PubMed]
    • (2001) J. Comp. Neurol , vol.435 , pp. 406-417
    • Cameron, H.A.1    McKay, R.D.2
  • 322
    • 39149107803 scopus 로고    scopus 로고
    • Mechanisms and functional implications of adult neurogenesis
    • [CrossRef] [PubMed]
    • Zhao, C.; Deng, W.; Gage, F.H. Mechanisms and functional implications of adult neurogenesis. Cell 2008,132, 645-660. [CrossRef] [PubMed]
    • (2008) Cell , vol.132 , pp. 645-660
    • Zhao, C.1    Deng, W.2    Gage, F.H.3
  • 323
    • 65549125564 scopus 로고    scopus 로고
    • Neural stem and progenitor cells retain their potential for proliferation and differentiation into functional neurons despite lower number in aged brain
    • [CrossRef] [PubMed]
    • Ahlenius, H.; Visan, V.; Kokaia, M.; Lindvall, O.; Kokaia, Z. Neural stem and progenitor cells retain their potential for proliferation and differentiation into functional neurons despite lower number in aged brain.J. Neurosci. 2009, 29, 4408-4419. [CrossRef] [PubMed]
    • (2009) J. Neurosci , vol.29 , pp. 4408-4419
    • Ahlenius, H.1    Visan, V.2    Kokaia, M.3    Lindvall, O.4    Kokaia, Z.5
  • 324
    • 84860885387 scopus 로고    scopus 로고
    • Aging brain microenvironment decreases hippocampal neurogenesis through Wnt-mediated survivin signaling
    • [CrossRef] [PubMed]
    • Miranda, C.J.; Braun, L.; Jiang, Y.; Hester, M.E.; Zhang, L.; Riolo, M.; Wang, H.; Rao, M.; Altura, R.A.;Kaspar, B.K. Aging brain microenvironment decreases hippocampal neurogenesis through Wnt-mediated survivin signaling. Aging Cell 2012, 11, 542-552. [CrossRef] [PubMed]
    • (2012) Aging Cell , vol.11 , pp. 542-552
    • Miranda, C.J.1    Braun, L.2    Jiang, Y.3    Hester, M.E.4    Zhang, L.5    Riolo, M.6    Wang, H.7    Rao, M.8    Altura, R.A.9    Kaspar, B.K.10
  • 325
    • 84855977476 scopus 로고    scopus 로고
    • New neurons in an aged brain
    • [CrossRef] [PubMed]
    • Lee, S.W.; Clemenson, G.D.; Gage, F.H. New neurons in an aged brain. Behav. Brain Res. 2012, 227, 497-507. [CrossRef] [PubMed]
    • (2012) Behav. Brain Res , vol.227 , pp. 497-507
    • Lee, S.W.1    Clemenson, G.D.2    Gage, F.H.3
  • 326
    • 70949100121 scopus 로고    scopus 로고
    • Telomere shortening in neural stem cells disrupts neuronal differentiation and neuritogenesis
    • [CrossRef] [PubMed]
    • Ferron, S.R.; Marques-Torrejon, M.A.; Mira, H.; Flores, I.; Taylor, K.; Blasco, M.A.; Farinas, I. Telomere shortening in neural stem cells disrupts neuronal differentiation and neuritogenesis. J. Neurosci. 2009, 29, 14394-14407. [CrossRef] [PubMed]
    • (2009) J. Neurosci , vol.29 , pp. 14394-14407
    • Ferron, S.R.1    Marques-Torrejon, M.A.2    Mira, H.3    Flores, I.4    Taylor, K.5    Blasco, M.A.6    Farinas, I.7
  • 328
    • 22144469750 scopus 로고    scopus 로고
    • Possible use of autologous stem cell therapies for Alzheimer’s disease
    • [CrossRef] [PubMed]
    • Sugaya, K. Possible use of autologous stem cell therapies for Alzheimer’s disease. Curr. Alzheimer Res. 2005, 2, 367-376. [CrossRef] [PubMed]
    • (2005) Curr. Alzheimer Res , vol.2 , pp. 367-376
    • Sugaya, K.1
  • 329
    • 49249116020 scopus 로고    scopus 로고
    • Behavior of neural stem cells in the Alzheimer brain
    • [CrossRef] [PubMed]
    • Waldau, B.; Shetty, A.K. Behavior of neural stem cells in the Alzheimer brain. Cell. Mol. Life Sci. 2008, 65,2372-2384. [CrossRef] [PubMed]
    • (2008) Cell. Mol. Life Sci , vol.65 , pp. 2372-2384
    • Waldau, B.1    Shetty, A.K.2
  • 332
    • 70349323417 scopus 로고    scopus 로고
    • CD14 and Toll-like receptors 2 and 4 are required for fibrillar Aß-stimulated microglial activation
    • [CrossRef] [PubMed]
    • Reed-Geaghan, E.G.; Savage, J.C.; Hise, A.G.; Landreth, G.E. CD14 and Toll-like receptors 2 and 4 are required for fibrillar Aß-stimulated microglial activation. J. Neurosci. 2009, 29, 11982-11992. [CrossRef] [PubMed]
    • (2009) J. Neurosci , vol.29 , pp. 11982-11992
    • Reed-Geaghan, E.G.1    Savage, J.C.2    Hise, A.G.3    Landreth, G.E.4
  • 333
    • 80052836312 scopus 로고    scopus 로고
    • MyD88 deficiency ameliorates ß-amyloidosis in an animal model of AD
    • [CrossRef] [PubMed]
    • Lim, J.E.; Kou, J.; Song, M.; Pattanayak, A.; Jin, J.; Lalonde, R.; Fukuchi, K. MyD88 deficiency ameliorates ß-amyloidosis in an animal model of AD. Am. J. Pathol. 2011, 179, 1095-1103. [CrossRef] [PubMed]
    • (2011) Am. J. Pathol , vol.179 , pp. 1095-1103
    • Lim, J.E.1    Kou, J.2    Song, M.3    Pattanayak, A.4    Jin, J.5    Lalonde, R.6    Fukuchi, K.7
  • 334
    • 84855271475 scopus 로고    scopus 로고
    • Utilization of APPswe/PS1dE9 transgenic mice in research of Alzheimer’s disease: Focus on gene therapy and cell-based therapy applications
    • [CrossRef] [PubMed]
    • Malm, T.; Koistinaho, J.; Kanninen, K. Utilization of APPswe/PS1dE9 transgenic mice in research of Alzheimer’s disease: Focus on gene therapy and cell-based therapy applications. Int. J. Alzheimers Dis. 2011, 2011. [CrossRef] [PubMed]
    • (2011) Int. J. Alzheimers Dis , pp. 2011
    • Malm, T.1    Koistinaho, J.2    Kanninen, K.3
  • 335
    • 0023643241 scopus 로고
    • Amelioration of cholinergicneuron atrophy and spatial memory impairment in aged rats by nerve growth factor
    • [CrossRef] [PubMed]
    • Fischer, W.; Wictorin, K.; Bjorklund, A.; Williams, L.P.; Varon, S.; Gage, F.H. Amelioration of cholinergicneuron atrophy and spatial memory impairment in aged rats by nerve growth factor. Nature 1987, 329, 65-68. [CrossRef] [PubMed]
    • (1987) Nature , vol.329 , pp. 65-68
    • Fischer, W.1    Wictorin, K.2    Bjorklund, A.3    Williams, L.P.4    Varon, S.5    Gage, F.H.6
  • 336
    • 0032579839 scopus 로고    scopus 로고
    • Permeability of the blood-brain barrier to neurotrophins
    • [CrossRef]
    • Pan, W.; Banks, W.A.; Kastin, A.J. Permeability of the blood-brain barrier to neurotrophins. Brain Res. 1998,788, 87-94. [CrossRef]
    • (1998) Brain Res , vol.788 , pp. 87-94
    • Pan, W.1    Banks, W.A.2    Kastin, A.J.3
  • 337
    • 0025816941 scopus 로고
    • Hypophagia is induced by intracerebroventricular administration of nerve growth factor
    • [CrossRef]
    • Williams, L.R. Hypophagia is induced by intracerebroventricular administration of nerve growth factor. Exp. Neurol. 1991, 113, 31-37. [CrossRef]
    • (1991) Exp. Neurol , vol.113 , pp. 31-37
    • Williams, L.R.1
  • 340
    • 62049083420 scopus 로고    scopus 로고
    • Neuroprotective effects of brain-derived neurotrophic factor in rodent and primate models of Alzheimer’s disease
    • [CrossRef] [PubMed]
    • Nagahara, A.H.; Merrill, D.A.; Coppola, G.; Tsukada, S.; Schroeder, B.E.; Shaked, G.M.; Wang, L.; Blesch, A.; Kim, A.; Conner, J.M.; et al. Neuroprotective effects of brain-derived neurotrophic factor in rodent and primate models of Alzheimer’s disease. Nat. Med. 2009, 15, 331-337. [CrossRef] [PubMed]
    • (2009) Nat. Med , vol.15 , pp. 331-337
    • Nagahara, A.H.1    Merrill, D.A.2    Coppola, G.3    Tsukada, S.4    Schroeder, B.E.5    Shaked, G.M.6    Wang, L.7    Blesch, A.8    Kim, A.9    Conner, J.M.10
  • 341
    • 0030658839 scopus 로고    scopus 로고
    • Brain-derived neurotrophic factor is reduced in Alzheimer’s disease. Mol
    • [CrossRef]
    • Connor, B.; Young, D.; Yan, Q.; Faull, R.L.; Synek, B.; Dragunow, M. Brain-derived neurotrophic factor is reduced in Alzheimer’s disease. Mol. Brain Res. 1997, 49, 71-81. [CrossRef]
    • (1997) Brain Res , vol.49 , pp. 71-81
    • Connor, B.1    Young, D.2    Yan, Q.3    Faull, R.L.4    Synek, B.5    Dragunow, M.6
  • 343
    • 33747195353 scopus 로고    scopus 로고
    • Induction of pluripotent stem cells from mouse embryonic and adult fibroblast cultures by defined factors
    • [CrossRef] [PubMed]
    • Takahashi, K.; Yamanaka, S. Induction of pluripotent stem cells from mouse embryonic and adult fibroblast cultures by defined factors. Cell 2006, 126, 663-676. [CrossRef] [PubMed]
    • (2006) Cell , vol.126 , pp. 663-676
    • Takahashi, K.1    Yamanaka, S.2
  • 345
    • 84864330048 scopus 로고    scopus 로고
    • Establishment of induced pluripotent stem cells from centenarians for neurodegenerative disease research
    • [CrossRef] [PubMed]
    • Yagi, T.; Kosakai, A.; Ito, D.; Okada, Y.; Akamatsu, W.; Nihei, Y.; Nabetani, A.; Ishikawa, F.; Arai, Y.; Hirose, N.; et al. Establishment of induced pluripotent stem cells from centenarians for neurodegenerative disease research. PLoS ONE 2012, 7, e41572. [CrossRef] [PubMed]
    • (2012) Plos ONE , vol.7
    • Yagi, T.1    Kosakai, A.2    Ito, D.3    Okada, Y.4    Akamatsu, W.5    Nihei, Y.6    Nabetani, A.7    Ishikawa, F.8    Arai, Y.9    Hirose, N.10
  • 349
    • 34548667973 scopus 로고    scopus 로고
    • Telomere length, stem cells and aging
    • Blasco, M.A. Telomere length, stem cells and aging. Nat. Chem. Biol. 2007, 3, 640-649.
    • (2007) Nat. Chem. Biol , vol.3 , pp. 640-649
    • Blasco, M.A.1
  • 350
    • 58949094552 scopus 로고    scopus 로고
    • Telomeres acquire embryonic stem cell characteristics in induced pluripotent stem cells
    • [CrossRef] [PubMed]
    • Marion, R.M.; Strati, K.; Li, H.; Tejera, A.; Schoeftner, S.; Ortega, S.; Serrano, M.; Blasco, M.A. Telomeres acquire embryonic stem cell characteristics in induced pluripotent stem cells. Cell Stem Cell 2009, 4, 141-154. [CrossRef] [PubMed]
    • (2009) Cell Stem Cell , vol.4 , pp. 141-154
    • Marion, R.M.1    Strati, K.2    Li, H.3    Tejera, A.4    Schoeftner, S.5    Ortega, S.6    Serrano, M.7    Blasco, M.A.8
  • 351
    • 79955573452 scopus 로고    scopus 로고
    • Association of telomere length with authentic pluripotency of ES/iPS cells
    • [CrossRef] [PubMed]
    • Huang, J.; Wang, F.; Okuka, M.; Liu, N.; Ji, G.; Ye, X.; Zuo, B.; Li, M.; Liang, P.; Ge, W.W.; et al. Association of telomere length with authentic pluripotency of ES/iPS cells. Cell Res. 2011, 21, 779-792. [CrossRef] [PubMed]
    • (2011) Cell Res , vol.21 , pp. 779-792
    • Huang, J.1    Wang, F.2    Okuka, M.3    Liu, N.4    Ji, G.5    Ye, X.6    Zuo, B.7    Li, M.8    Liang, P.9    Ge, W.W.10
  • 352
    • 77953504104 scopus 로고    scopus 로고
    • Proteinopathy-induced neuronal senescence: A hypothesis for brain failure in Alzheimer’s and other neurodegenerative diseases
    • [CrossRef] [PubMed]
    • Golde, T.E.; Miller, V.M. Proteinopathy-induced neuronal senescence: A hypothesis for brain failure in Alzheimer’s and other neurodegenerative diseases. Alzheimers Res. Ther. 2009, 1, 5. [CrossRef] [PubMed]
    • (2009) Alzheimers Res. Ther , vol.1 , pp. 5
    • Golde, T.E.1    Miller, V.M.2
  • 353
    • 84887995472 scopus 로고    scopus 로고
    • The senescence hypothesis of disease progression in Alzheimer’s disease: An integrated matrix of disease pathways for FAD and SAD. Mol
    • [CrossRef] [PubMed]
    • Hunter, S.; Arendt, T.; Brayne, C. The senescence hypothesis of disease progression in Alzheimer’s disease: An integrated matrix of disease pathways for FAD and SAD. Mol. Neurobiol. 2013, 48, 556-570. [CrossRef] [PubMed]
    • (2013) Neurobiol , vol.48 , pp. 556-570
    • Hunter, S.1    Arendt, T.2    Brayne, C.3
  • 354
    • 0024535476 scopus 로고
    • Brain transplants enhance rather than reduce the impairment of spatial memory and olfaction in bulbectomized rats
    • [CrossRef] [PubMed]
    • Amemori, T.; Ermakova, I.V.; Buresova, O.; Zigova, T.; Racekova, E.; Bures, J. Brain transplants enhance rather than reduce the impairment of spatial memory and olfaction in bulbectomized rats. Behav. Neurosci. 1989, 103, 61-70. [CrossRef] [PubMed]
    • (1989) Behav. Neurosci , vol.103 , pp. 61-70
    • Amemori, T.1    Ermakova, I.V.2    Buresova, O.3    Zigova, T.4    Racekova, E.5    Bures, J.6
  • 355
    • 33846461062 scopus 로고    scopus 로고
    • Morris water maze: Procedures for assessing spatial and related forms of learning and memory
    • [CrossRef] [PubMed]
    • Vorhees, C.V.; Williams, M.T. Morris water maze: Procedures for assessing spatial and related forms of learning and memory. Nat. Protoc. 2006, 1, 848-858. [CrossRef] [PubMed]
    • (2006) Nat. Protoc , vol.1 , pp. 848-858
    • Vorhees, C.V.1    Williams, M.T.2


* 이 정보는 Elsevier사의 SCOPUS DB에서 KISTI가 분석하여 추출한 것입니다.