메뉴 건너뛰기




Volumn 70, Issue 11, 2011, Pages 944-959

Aβ-degrading enzymes: Potential for treatment of alzheimer disease

Author keywords

A degrading enzymes; Alzheimer disease treatment; Cathepsin B; Convection enhanced delivery,Mouse models; Neprilysin

Indexed keywords

AMYLOID BETA PROTEIN; ENDOTHELIN CONVERTING ENZYME; GREEN TEA EXTRACT; IMATINIB; INSULINASE; MEMBRANE METALLOENDOPEPTIDASE;

EID: 80054891306     PISSN: 00223069     EISSN: 15546578     Source Type: Journal    
DOI: 10.1097/NEN.0b013e3182345e46     Document Type: Review
Times cited : (223)

References (278)
  • 2
    • 0030761094 scopus 로고    scopus 로고
    • The presenilins and Alzheimer's disease
    • DOI 10.1093/hmg/6.10.1639
    • Hutton M, Hardy J. The presenilins and Alzheimer's disease. Hum Mol Genet 1997;6:1639-46 (Pubitemid 27401606)
    • (1997) Human Molecular Genetics , vol.6 , Issue.10 REV. ISS. , pp. 1639-1646
    • Hutton, M.1    Hardy, J.2
  • 3
    • 0031052381 scopus 로고    scopus 로고
    • Amyloid, the presenilins and Alzheimer's disease
    • DOI 10.1016/S0166-2236(96)01030-2, PII S0166223696010302
    • Hardy J. Amyloid, the presenilins and Alzheimer's disease. Trends Neurosci 1997;20:154-59 (Pubitemid 27116286)
    • (1997) Trends in Neurosciences , vol.20 , Issue.4 , pp. 154-159
    • Hardy, J.1
  • 7
    • 78650678688 scopus 로고    scopus 로고
    • Decreased clearance of CNS A-amyloid in Alzheimer's disease
    • Mawuenyega KG, Sigurdson W, Ovod V, et al. Decreased clearance of CNS A-amyloid in Alzheimer's disease. Science 2010;330:1774
    • (2010) Science , vol.330 , pp. 1774
    • Mawuenyega, K.G.1    Sigurdson, W.2    Ovod, V.3
  • 8
    • 78049254530 scopus 로고    scopus 로고
    • Cerebral amyloid angiopathy in the aetiology and immunotherapy of Alzheimer disease
    • Weller RO, Preston SD, Subash M, et al. Cerebral amyloid angiopathy in the aetiology and immunotherapy of Alzheimer disease. Alzheimers Res Ther 2009;1:6
    • (2009) Alzheimers Res Ther , vol.1 , pp. 6
    • Weller, R.O.1    Preston, S.D.2    Subash, M.3
  • 9
    • 0034095485 scopus 로고    scopus 로고
    • Cerebral amyloid angiopathy: Accumulation of Aβ in interstitial fluid drainage pathways in Alzheimer's disease
    • Weller RO, Massey A, Kuo YM, et al. Cerebral amyloid angiopathy: Accumulation of AA in interstitial fluid drainage pathways in Alzheimer's disease. Ann N Y Acad Sci 2000;903:110-17 (Pubitemid 30251546)
    • (2000) Annals of the New York Academy of Sciences , vol.903 , pp. 110-117
    • Weller, R.O.1    Massey, A.2    Kuo, Y.-M.3    Roher, A.E.4
  • 10
    • 0037385370 scopus 로고    scopus 로고
    • Capillary and arterial cerebral amyloid angiopathy in Alzheimer's disease: Defining the perivascular route for the elimination of amyloid β from the human brain
    • DOI 10.1046/j.1365-2990.2003.00424.x
    • Preston SD, Steart PV, Wilkinson A, et al. Capillary and arterial cerebral amyloid angiopathy in Alzheimer's disease: Defining the perivascular route for the elimination of amyloid A from the human brain. Neuropathol Appl Neurobiol 2003;29:106-17 (Pubitemid 36408329)
    • (2003) Neuropathology and Applied Neurobiology , vol.29 , Issue.2 , pp. 106-117
    • Preston, S.D.1    Steart, P.V.2    Wilkinson, A.3    Nicoll, J.A.R.4    Weller, R.O.5
  • 13
    • 0035112889 scopus 로고    scopus 로고
    • A-Amyloid efflux mediated by P-glycoprotein
    • Lam FC, Liu R, Lu P, et al. A-Amyloid efflux mediated by P-glycoprotein. J Neurochem 2001;76:1121-28
    • (2001) J Neurochem , vol.76 , pp. 1121-1128
    • Lam, F.C.1    Liu, R.2    Lu, P.3
  • 16
    • 0034814056 scopus 로고    scopus 로고
    • Microglial chemotaxis, activation, and phagocytosis of amyloid β-peptide as linked phenomena in Alzheimer's disease
    • DOI 10.1016/S0197-0186(01)00040-7, PII S0197018601000407
    • Rogers J, Lue LF. Microglial chemotaxis, activation, and phagocytosis of amyloid A-peptide as linked phenomena in Alzheimer's disease. Neurochem Int 2001;39:333-40 (Pubitemid 32907397)
    • (2001) Neurochemistry International , vol.39 , Issue.5-6 , pp. 333-340
    • Rogers, J.1    Lue, L.-F.2
  • 17
    • 33644853531 scopus 로고    scopus 로고
    • Metabolism of amyloid β peptide and pathogenesis of Alzheimer's disease: Towards presymptomatic diagnosis, prevention and therapy
    • DOI 10.1016/j.neures.2005.12.015, PII S0168010205003330
    • Saido TC, Iwata N. Metabolism of amyloid A peptide and pathogenesis of Alzheimer's disease. Towards presymptomatic diagnosis, prevention and therapy. Neurosci Res 2006;54:235-53 (Pubitemid 43374441)
    • (2006) Neuroscience Research , vol.54 , Issue.4 , pp. 235-253
    • Saido, T.C.1    Iwata, N.2
  • 18
    • 41149149615 scopus 로고    scopus 로고
    • AA-degrading enzymes in Alzheimer's disease
    • Miners JS, Baig S, Palmer J, et al. AA-degrading enzymes in Alzheimer's disease. Brain Pathol 2008;18:240-52
    • (2008) Brain Pathol , vol.18 , pp. 240-252
    • Miners, J.S.1    Baig, S.2    Palmer, J.3
  • 19
    • 57649178322 scopus 로고    scopus 로고
    • The AACs of AA-cleaving proteases
    • Leissring MA. The AACs of AA-cleaving proteases. J Biol Chem 2008; 283:29645-49
    • (2008) J Biol Chem , vol.283 , pp. 29645-29649
    • Leissring, M.A.1
  • 20
    • 0031679480 scopus 로고    scopus 로고
    • Cerebral amyloid angiopathy: Amyloid β accumulates in putative interstitial fluid drainage pathways in Alzheimer's disease
    • Weller RO, Massey A, Newman TA, et al. Cerebral amyloid angiopathy: Amyloid A accumulates in putative interstitial fluid drainage pathways in Alzheimer's disease. Am J Pathol 1998;153:725-33 (Pubitemid 28419027)
    • (1998) American Journal of Pathology , vol.153 , Issue.3 , pp. 725-733
    • Weller, R.O.1    Massey, A.2    Newman, T.A.3    Hutchings, M.4    Kuo, Y.-M.5    Roher, A.E.6
  • 21
    • 48949117577 scopus 로고    scopus 로고
    • The role of the cell surface LRP and soluble LRP in blood-brain barrier Aβ clearance in Alzheimer's disease
    • DOI 10.2174/138161208784705487
    • Deane R, Sagare A, Zlokovic BV. The role of the cell surface LRP and soluble LRP in blood-brain barrier AA clearance in Alzheimer's disease. Curr Pharm Des 2008;14:1601-5 (Pubitemid 352002987)
    • (2008) Current Pharmaceutical Design , vol.14 , Issue.16 , pp. 1601-1605
    • Dearie, R.1    Sagare, A.2    Zlokovic, B.V.3
  • 22
    • 65649105035 scopus 로고    scopus 로고
    • Clearance of amyloid-A peptide across the blood-brain barrier: Implication for therapies in Alzheimer's disease
    • Deane R, Bell RD, Sagare A, et al. Clearance of amyloid-A peptide across the blood-brain barrier: Implication for therapies in Alzheimer's disease. CNS Neurol Disord Drug Targets 2009;8:16-30
    • (2009) CNS Neurol Disord Drug Targets , vol.8 , pp. 16-30
    • Deane, R.1    Bell, R.D.2    Sagare, A.3
  • 23
    • 80052022534 scopus 로고    scopus 로고
    • Neprilysin protects against cerebral amyloid angiopathy and AA-induced degeneration of cerebrovascular smooth muscle cells
    • Miners JS, Kehoe P, Love S. Neprilysin protects against cerebral amyloid angiopathy and AA-induced degeneration of cerebrovascular smooth muscle cells. Brain Pathol 2011;21:594-605
    • (2011) Brain Pathol , vol.21 , pp. 594-605
    • Miners, J.S.1    Kehoe, P.2    Love, S.3
  • 24
    • 0036849021 scopus 로고    scopus 로고
    • Microglia and inflammatory mechanisms in the clearance of amyloid β peptide
    • DOI 10.1002/glia.10153
    • Rogers J, Strohmeyer R, Kovelowski CJ, et al. Microglia and inflammatory mechanisms in the clearance of amyloid A peptide. Glia 2002; 40:260-69 (Pubitemid 35337383)
    • (2002) GLIA , vol.40 , Issue.2 , pp. 260-269
    • Rogers, J.1    Strohmeyer, R.2    Kovelowski, C.J.3    Li, R.4
  • 25
    • 78651374915 scopus 로고    scopus 로고
    • Accumulation of insoluble amyloid-A in Down's syndrome is associated with increased BACE-1 and neprilysin activities
    • Miners JS, Morris S, Love S, Kehoe PG. Accumulation of insoluble amyloid-A in Down's syndrome is associated with increased BACE-1 and neprilysin activities. J Alzheimers Dis 2011;23:101-8
    • (2011) J Alzheimers Dis , vol.23 , pp. 101-8
    • Miners, J.S.1    Morris, S.2    Love, S.3    Kehoe, P.G.4
  • 26
    • 69449102820 scopus 로고    scopus 로고
    • Neprilysin and insulin-degrading enzyme levels are increased in Alzheimer disease in relation to disease severity
    • Miners JS, Baig S, Tayler H, et al. Neprilysin and insulin-degrading enzyme levels are increased in Alzheimer disease in relation to disease severity. J Neuropathol Exp Neurol 2009;68:902-14
    • (2009) J Neuropathol Exp Neurol , vol.68 , pp. 902-914
    • Miners, J.S.1    Baig, S.2    Tayler, H.3
  • 30
    • 67849110360 scopus 로고    scopus 로고
    • Effects of 4-hydroxy-nonenal and amyloid-A on expression and activity of endothelin converting enzyme and insulin degrading enzyme in SH-SY5Y cells
    • Wang R, Wang S, Malter JS, et al. Effects of 4-hydroxy-nonenal and amyloid-A on expression and activity of endothelin converting enzyme and insulin degrading enzyme in SH-SY5Y cells. J Alzheimers Dis 2009;17:489-501
    • (2009) J Alzheimers Dis , vol.17 , pp. 489-501
    • Wang, R.1    Wang, S.2    Malter, J.S.3
  • 31
    • 67649984550 scopus 로고    scopus 로고
    • Endothelin-converting enzyme-2 is increased in Alzheimer's disease and up-regulated by AA
    • Palmer JC, Baig S, Kehoe PG, et al. Endothelin-converting enzyme-2 is increased in Alzheimer's disease and up-regulated by AA. Am J Pathol 2009;175:262-70
    • (2009) Am J Pathol , vol.175 , pp. 262-270
    • Palmer, J.C.1    Baig, S.2    Kehoe, P.G.3
  • 33
    • 78650213691 scopus 로고    scopus 로고
    • ACE variants and association with brain AA levels in Alzheimer's disease
    • Miners JS, van Helmond Z, Raiker M, et al. ACE variants and association with brain AA levels in Alzheimer's disease. Am J Transl Res 2010;3:73-80
    • (2010) Am J Transl Res , vol.3 , pp. 73-80
    • Miners, J.S.1    Van Helmond, Z.2    Raiker, M.3
  • 35
    • 0038241040 scopus 로고    scopus 로고
    • Pathogenic Aβ induces the expression and activation of matrix metalloproteinase-2 in human cerebrovascular smooth muscle cells
    • DOI 10.1046/j.1471-4159.2003.01745.x
    • Jung SS, Zhang W, Van Nostrand WE. Pathogenic AA induces the expression and activation of matrix metalloproteinase-2 in human cerebrovascular smooth muscle cells. J Neurochem 2003;85:1208-15 (Pubitemid 36613269)
    • (2003) Journal of Neurochemistry , vol.85 , Issue.5 , pp. 1208-1215
    • Jung, S.S.1    Zhang, W.2    Van Nostrand, W.E.3
  • 36
    • 0032900361 scopus 로고    scopus 로고
    • Activated isoforms of MMP-2 are induced in U87 human glioma cells in response to β-amyloid peptide
    • DOI 10.1002/(SICI)1097-4547(19990101)55:1<44::AID-JNR6>3.0.CO;2-G
    • Deb S, Zhang JW, Gottschall PE. Activated isoforms of MMP-2 are induced in U87 human glioma cells in response to A-amyloid peptide. J Neurosci Res 1999;55:44-53 (Pubitemid 29014410)
    • (1999) Journal of Neuroscience Research , vol.55 , Issue.1 , pp. 44-53
    • Deb, S.1    Zhang, J.W.2    Gottschall, P.E.3
  • 37
    • 0035066332 scopus 로고    scopus 로고
    • Alzheimer's disease: Genes, proteins, and therapy
    • Selkoe DJ. Alzheimer's disease: Genes, proteins, and therapy. Physiol Rev 2001;81:741-66 (Pubitemid 32267077)
    • (2001) Physiological Reviews , vol.81 , Issue.2 , pp. 741-766
    • Selkoe, D.J.1
  • 38
    • 3943092621 scopus 로고    scopus 로고
    • Pathways towards and away from Alzheimer's disease
    • DOI 10.1038/nature02621
    • Mattson MP. Pathways towards and away from Alzheimer's disease. Nature 2004;430:631-39 (Pubitemid 39061671)
    • (2004) Nature , vol.430 , Issue.7000 , pp. 631-639
    • Mattson, M.P.1
  • 39
    • 0035997231 scopus 로고    scopus 로고
    • Biogenesis and metabolism of Alzheimer's disease Aβ amyloid peptides
    • DOI 10.1016/S0196-9781(02)00063-3, PII S0196978102000633
    • Evin G, Weidemann A. Biogenesis and metabolism of Alzheimer's disease AA amyloid peptides. Peptides 2002;23:1285-97 (Pubitemid 34786707)
    • (2002) Peptides , vol.23 , Issue.7 , pp. 1285-1297
    • Evin, G.1    Weidemann, A.2
  • 43
    • 0027258525 scopus 로고
    • The carboxy terminus of the β amyloid protein is critical for the seeding of amyloid formation: Implications for the pathogenesis of Alzheimer's disease
    • DOI 10.1021/bi00069a001
    • Jarrett JT, Berger EP, Lansbury PT Jr. The carboxy terminus of the A amyloid protein is critical for the seeding of amyloid formation: Implications for the pathogenesis of Alzheimer's disease. Biochemistry 1993; 32:4693-97 (Pubitemid 23162022)
    • (1993) Biochemistry , vol.32 , Issue.18 , pp. 4693-4697
    • Jarrett, J.T.1    Berger, E.P.2    Lansbury Jr., P.T.3
  • 44
    • 0347755563 scopus 로고    scopus 로고
    • Contribution of cerebral amyloid angiopathy to Alzheimer's disease
    • Love S. Contribution of cerebral amyloid angiopathy to Alzheimer's disease. J Neurol Neurosurg Psychiatry 2004;75:1-4 (Pubitemid 38091257)
    • (2004) Journal of Neurology, Neurosurgery and Psychiatry , vol.75 , Issue.1 , pp. 1-4
    • Love, S.1
  • 46
    • 0035947207 scopus 로고    scopus 로고
    • Metabolic regulation of brain AA by neprilysin
    • Iwata N, Tsubuki S, Takaki Y, et al. Metabolic regulation of brain AA by neprilysin. Science 2001;292:1550-52
    • (2001) Science , vol.292 , pp. 1550-1552
    • Iwata, N.1    Tsubuki, S.2    Takaki, Y.3
  • 47
    • 0346101885 scopus 로고    scopus 로고
    • Enhanced proteolysis of β-amyloid in APP transgenic mice prevents plaque formation, secondary pathology, and premature death
    • DOI 10.1016/S0896-6273(03)00787-6
    • Leissring MA, Farris W, Chang AY, et al. Enhanced proteolysis of A-amyloid in APP transgenic mice prevents plaque formation, secondary pathology, and premature death. Neuron 2003;40:1087-93 (Pubitemid 38032786)
    • (2003) Neuron , vol.40 , Issue.6 , pp. 1087-1093
    • Leissring, M.A.1    Farris, W.2    Chang, A.Y.3    Walsh, D.M.4    Wu, X.5    Sun, X.6    Frosch, M.P.7    Selkoe, D.J.8
  • 48
    • 0029061685 scopus 로고
    • Neutral endopeptidase can hydrolyze A-amyloid(1-40) but shows no effect on A-amyloid precursor protein metabolism
    • Howell S, Nalbantoglu J, Crine P. Neutral endopeptidase can hydrolyze A-amyloid(1-40) but shows no effect on A-amyloid precursor protein metabolism. Peptides 1995;16:647-52
    • (1995) Peptides , vol.16 , pp. 647-652
    • Howell, S.1    Nalbantoglu, J.2    Crine, P.3
  • 49
    • 0033548194 scopus 로고    scopus 로고
    • Hydrolysis of peptide hormones by endothelin-converting enzyme-1. A comparison with neprilysin
    • Johnson GD, Stevenson T, Ahn K. Hydrolysis of peptide hormones by endothelin-converting enzyme-1. A comparison with neprilysin. J Biol Chem 1999;274:4053-58
    • (1999) J Biol Chem , vol.274 , pp. 4053-4058
    • Johnson, G.D.1    Stevenson, T.2    Ahn, K.3
  • 50
    • 0026721943 scopus 로고
    • A-Amyloid precursor protein cleavage by a membranebound protease
    • Sisodia SS. A-Amyloid precursor protein cleavage by a membranebound protease. Proc Natl Acad Sci USA 1992;89:6075-79
    • (1992) Proc Natl Acad Sci USA , vol.89 , pp. 6075-6079
    • Sisodia, S.S.1
  • 51
    • 0035816707 scopus 로고    scopus 로고
    • Degradation of the Alzheimer's amyloid A peptide by endothelin-converting enzyme
    • Eckman EA, Reed DK, Eckman CB. Degradation of the Alzheimer's amyloid A peptide by endothelin-converting enzyme. J Biol Chem 2001; 276:24540-48
    • (2001) J Biol Chem , vol.276 , pp. 24540-24548
    • Eckman, E.A.1    Reed, D.K.2    Eckman, C.B.3
  • 52
    • 0034551718 scopus 로고    scopus 로고
    • Insulysin hydrolyzes amyloid A peptides to products that are neither neurotoxic nor deposit on amyloid plaques
    • Mukherjee A, Song E, Kihiko-Ehmann M, et al. Insulysin hydrolyzes amyloid A peptides to products that are neither neurotoxic nor deposit on amyloid plaques. J Neurosci 2000;20:8745-49
    • (2000) J Neurosci , vol.20 , pp. 8745-8749
    • Mukherjee, A.1    Song, E.2    Kihiko-Ehmann, M.3
  • 53
    • 0028176821 scopus 로고
    • Alzheimer's A-amyloid peptide specifically interacts with and is degraded by insulin degrading enzyme
    • Kurochkin IV, Goto S. Alzheimer's A-amyloid peptide specifically interacts with and is degraded by insulin degrading enzyme. FEBS Lett 1994;345:33-37
    • (1994) FEBS Lett , vol.345 , pp. 33-37
    • Kurochkin, I.V.1    Goto, S.2
  • 56
    • 0035930538 scopus 로고    scopus 로고
    • Angiotensin-converting enzyme degrades Alzheimer amyloid A-peptide (AA); Retards AA aggregation, deposition, fibril formation; And inhibits cytotoxicity
    • Hu J, Igarashi A, Kamata M, et al. Angiotensin-converting enzyme degrades Alzheimer amyloid A-peptide (AA); retards AA aggregation, deposition, fibril formation; and inhibits cytotoxicity. J Biol Chem 2001;276:47863-68
    • (2001) J Biol Chem , vol.276 , pp. 47863-47868
    • Hu, J.1    Igarashi, A.2    Kamata, M.3
  • 57
    • 14944340658 scopus 로고    scopus 로고
    • The N-terminal active centre of human angiotensin-converting enzyme degrades Alzheimer amyloid β-peptide
    • DOI 10.1111/j.1460-9568.2005.03912.x
    • Oba R, Igarashi A, Kamata M, et al. The N-terminal active centre of human angiotensin-converting enzyme degrades Alzheimer amyloid A-peptide. Eur J Neurosci 2005;21:733-40 (Pubitemid 40363508)
    • (2005) European Journal of Neuroscience , vol.21 , Issue.3 , pp. 733-740
    • Oba, R.1    Igarashi, A.2    Kamata, M.3    Nagata, K.4    Takano, S.5    Nakagawa, H.6
  • 58
    • 44249110915 scopus 로고    scopus 로고
    • Catabolic attacks of membranebound angiotensin-converting enzyme on the N-terminal part of speciesspecific amyloid-A peptides
    • Sun X, Becker M, Pankow K, et al. Catabolic attacks of membranebound angiotensin-converting enzyme on the N-terminal part of speciesspecific amyloid-A peptides. Eur J Pharmacol 2008;588:18-25
    • (2008) Eur J Pharmacol , vol.588 , pp. 18-25
    • Sun, X.1    Becker, M.2    Pankow, K.3
  • 59
    • 38549117383 scopus 로고    scopus 로고
    • The N-domain of angiotensin-converting enzyme specifically hydrolyzes the Arg-5-His-6 bond of Alzheimer's Aβ-(1-16) peptide and its isoAsp-7 analogue with different efficiency as evidenced by quantitative matrix-assisted laser desorption/ionization time-of-flight mass spectrometry
    • DOI 10.1002/rcm.3357
    • Toropygin IY, Kugaevskaya EV, Mirgorodskaya OA, et al. The N-domain of angiotensin-converting enzyme specifically hydrolyzes the Arg-5-His-6 bond of Alzheimer's AA-(1-16) peptide and its isoAsp-7 analogue with different efficiency as evidenced by quantitative matrixassisted laser desorption/ionization time-of-flight mass spectrometry. Rapid Commun Mass Spectrom 2008;22:231-39 (Pubitemid 351156136)
    • (2008) Rapid Communications in Mass Spectrometry , vol.22 , Issue.2 , pp. 231-239
    • Toropygin, I.Yu.1    Kugaevskaya, E.V.2    Mirgorodskaya, O.A.3    Elisseeva, Y.E.4    Kozmin, Y.P.5    Popov, I.A.6    Nikolaev, E.N.7    Makarov, A.A.8    Kozin, S.A.9
  • 60
    • 70450257558 scopus 로고    scopus 로고
    • AA42-to-AA40-and angiotensinconverting activities in different domains of angiotensin-converting enzyme
    • Zou K, Maeda T, Watanabe A, et al. AA42-to-AA40-and angiotensinconverting activities in different domains of angiotensin-converting enzyme. J Biol Chem 2009;284:31914-20
    • (2009) J Biol Chem , vol.284 , pp. 31914-31920
    • Zou, K.1    Maeda, T.2    Watanabe, A.3
  • 61
    • 59449101792 scopus 로고    scopus 로고
    • Restored degradation of the Alzheimer's amyloid-A peptide by targeting amyloid formation
    • Crouch PJ, Tew DJ, Du T, et al. Restored degradation of the Alzheimer's amyloid-A peptide by targeting amyloid formation. J Neurochem 2009;108:1198-207
    • (2009) J Neurochem , vol.108 , pp. 1198-207
    • Crouch, P.J.1    Tew, D.J.2    Du, T.3
  • 62
    • 0028788221 scopus 로고
    • White matter microglia produce membrane-type matrix metalloprotease, an activator of gelatinase A, in human brain tissues
    • Yamada T, Yoshiyama Y, Sato H, et al. White matter microglia produce membrane-type matrix metalloprotease, an activator of gelatinase A, in human brain tissues. Acta Neuropathol 1995;90:421-24
    • (1995) Acta Neuropathol , vol.90 , pp. 421-424
    • Yamada, T.1    Yoshiyama, Y.2    Sato, H.3
  • 63
    • 0028670204 scopus 로고
    • Proteolysis of A A peptide from Alzheimer disease brain by gelatinase A
    • Roher AE, Kasunic TC, Woods AS, et al. Proteolysis of A A peptide from Alzheimer disease brain by gelatinase A. Biochem Biophys Res Commun 1994;205:1755-61
    • (1994) Biochem Biophys Res Commun , vol.205 , pp. 1755-1761
    • Roher, A.E.1    Kasunic, T.C.2    Woods, A.S.3
  • 65
    • 0029852887 scopus 로고    scopus 로고
    • Matrix metalloproteinase-9 (MMP-9) is synthesized in neurons of the human hippocampus and is capable of degrading the amyloid-β peptide (1-40)
    • Backstrom JR, Lim GP, Cullen MJ, et al. Matrix metalloproteinase-9 (MMP-9) is synthesized in neurons of the human hippocampus and is capable of degrading the amyloid-A peptide (1-40). J Neurosci 1996;16: 7910-19 (Pubitemid 26405054)
    • (1996) Journal of Neuroscience , vol.16 , Issue.24 , pp. 7910-7919
    • Backstrom, J.R.1    Lim, G.P.2    Cullen, M.J.3    Tokes, Z.A.4
  • 66
    • 0033621048 scopus 로고    scopus 로고
    • Plasmin cleavage of the amyloid A-protein: Alteration of secondary structure and stimulation of tissue plasminogen activator activity
    • Van Nostrand WE, Porter M. Plasmin cleavage of the amyloid A-protein: Alteration of secondary structure and stimulation of tissue plasminogen activator activity. Biochemistry 1999;38:11570-76
    • (1999) Biochemistry , vol.38 , pp. 11570-11576
    • Van Nostrand, W.E.1    Porter, M.2
  • 67
    • 0034573026 scopus 로고    scopus 로고
    • Brain plasmin enhances APP α-cleavage and AA degradation and is reduced in Alzheimer's disease brains
    • Ledesma MD, Da Silva JS, Crassaerts K, et al. Brain plasmin enhances APP α-cleavage and AA degradation and is reduced in Alzheimer's disease brains. EMBO Rep 2000;1:530-35
    • (2000) EMBO Rep , vol.1 , pp. 530-535
    • Ledesma, M.D.1    Da Silva, J.S.2    Crassaerts, K.3
  • 68
    • 76749099393 scopus 로고    scopus 로고
    • Degradation of soluble and fibrillar amyloid A-protein by matrix metalloproteinase (MT1-MMP) in vitro
    • Liao MC, Van Nostrand WE. Degradation of soluble and fibrillar amyloid A-protein by matrix metalloproteinase (MT1-MMP) in vitro. Biochemistry 2010;49:1127-36
    • (2010) Biochemistry , vol.49 , pp. 1127-1136
    • Liao, M.C.1    Van Nostrand, W.E.2
  • 69
    • 70350366775 scopus 로고    scopus 로고
    • Degradation of amyloid A protein by purified myelin basic protein
    • Liao MC, Ahmed M, Smith SO, et al. Degradation of amyloid A protein by purified myelin basic protein. J Biol Chem 2009;284:28917-25
    • (2009) J Biol Chem , vol.284 , pp. 28917-28925
    • Liao, M.C.1    Ahmed, M.2    Smith, S.O.3
  • 70
    • 69049097135 scopus 로고    scopus 로고
    • Acyl peptide hydrolase degrades monomeric and oligomeric amyloid-A peptide
    • Yamin R, Zhao C, O'Connor PB, et al. Acyl peptide hydrolase degrades monomeric and oligomeric amyloid-A peptide. Mol Neurodegener 2009;4:33
    • (2009) Mol Neurodegener , vol.4 , pp. 33
    • Yamin, R.1    Zhao, C.2    O'Connor, P.B.3
  • 71
    • 33845763797 scopus 로고    scopus 로고
    • Acyl peptide hydrolase, a serine proteinase isolated from conditioned medium of neuroblastoma cells, degrades the amyloid-β peptide
    • DOI 10.1111/j.1471-4159.2006.04251.x
    • Yamin R, Bagchi S, Hildebrant R, et al. Acyl peptide hydrolase, a serine proteinase isolated from conditioned medium of neuroblastoma cells, degrades the amyloid-A peptide. J Neurochem 2007;100:458-67 (Pubitemid 46011006)
    • (2007) Journal of Neurochemistry , vol.100 , Issue.2 , pp. 458-467
    • Yamin, R.1    Bagchi, S.2    Hildebrant, R.3    Scaloni, A.4    Widom, R.L.5    Abraham, C.R.6
  • 72
    • 47249153453 scopus 로고    scopus 로고
    • Human Membrane Metalloendopeptidase-like Protein Degrades Both A-amyloid 42 and A-amyloid 40
    • Huang JY, Bruno AM, Patel CA, et al. Human membrane metalloendopeptidase- like protein degrades both A-amyloid 42 and A-amyloid40 Neuroscience 2008;155:258-62
    • (2008) Neuroscience , vol.155 , pp. 258-262
    • Huang, J.Y.1    Bruno, A.M.2    Patel, C.A.3
  • 73
    • 77950924188 scopus 로고    scopus 로고
    • Mechanisms of enzymatic degradation of amyloid A microfibrils generating nanofilaments and nanospheres related to cytotoxicity
    • Numata K, Kaplan DL. Mechanisms of enzymatic degradation of amyloid A microfibrils generating nanofilaments and nanospheres related to cytotoxicity. Biochemistry 2010;49:3254-60
    • (2010) Biochemistry , vol.49 , pp. 3254-3260
    • Numata, K.1    Kaplan, D.L.2
  • 74
    • 0036077536 scopus 로고    scopus 로고
    • β-amyloid catabolism: Roles for neprilysin (NEP) and other metallopeptidases?
    • DOI 10.1046/j.1471-4159.2002.00855.x
    • Carson JA, Turner AJ. A-Amyloid catabolism: Roles for neprilysin (NEP) and other metallopeptidases? J Neurochem 2002;81:1-8 (Pubitemid 34808849)
    • (2002) Journal of Neurochemistry , vol.81 , Issue.1 , pp. 1-8
    • Carson, J.A.1    Turner, A.J.2
  • 76
    • 27844442718 scopus 로고    scopus 로고
    • Aβ-degrading enzymes: Modulators of Alzheimer's disease pathogenesis and targets for therapeutic intervention
    • DOI 10.1042/BST20051101
    • Eckman EA, Eckman CB. AA-degrading enzymes: Modulators of Alzheimer's disease pathogenesis and targets for therapeutic intervention. Biochem Soc Trans 2005;33:1101-5 (Pubitemid 41659123)
    • (2005) Biochemical Society Transactions , vol.33 , Issue.5 , pp. 1101-1105
    • Eckman, E.A.1    Eckman, C.B.2
  • 77
    • 77951975748 scopus 로고    scopus 로고
    • Structural conversion of neurotoxic amyloid-A (1-42) oligomers to fibrils
    • Ahmed M, Davis J, Aucoin D, et al. Structural conversion of neurotoxic amyloid-A (1-42) oligomers to fibrils. Nat Struct Mol Biol 2010;17: 561-67
    • (2010) Nat Struct Mol Biol , vol.17 , pp. 561-567
    • Ahmed, M.1    Davis, J.2    Aucoin, D.3
  • 79
    • 0028952265 scopus 로고
    • Endopeptidase-24.11 is the integral membrane peptidase initiating degradation of somatostatin in the hippocampus
    • Barnes K, Doherty S, Turner AJ. Endopeptidase-24.11 is the integral membrane peptidase initiating degradation of somatostatin in the hippocampus. J Neurochem 1995;64:1826-32
    • (1995) J Neurochem , vol.64 , pp. 1826-1832
    • Barnes, K.1    Doherty, S.2    Turner, A.J.3
  • 81
    • 0036794008 scopus 로고    scopus 로고
    • Declining expression of neprilysin in Alzheimer disease vasculature: Possible involvement in cerebral amyloid angiopathy
    • Carpentier M, Robitaille Y, DesGroseillers L, et al. Declining expression of neprilysin in Alzheimer disease vasculature: Possible involvement in cerebral amyloid angiopathy. J Neuropathol Exp Neurol 2002; 61:849-56
    • (2002) J Neuropathol Exp Neurol , vol.61 , pp. 849-856
    • Carpentier, M.1    Robitaille, Y.2    Desgroseillers, L.3
  • 83
    • 0042827324 scopus 로고    scopus 로고
    • Human neprilysin is capable of degrading amyloid β peptide not only in the monomeric form but also the pathological oligomeric form
    • DOI 10.1016/S0304-3940(03)00898-X
    • Kanemitsu H, Tomiyama T, Mori H. Human neprilysin is capable of degrading amyloid A peptide not only in the monomeric form but also the pathological oligomeric form. Neurosci Lett 2003;350:113-16 (Pubitemid 37101425)
    • (2003) Neuroscience Letters , vol.350 , Issue.2 , pp. 113-116
    • Kanemitsu, H.1    Tomiyama, T.2    Mori, H.3
  • 85
    • 45549087494 scopus 로고    scopus 로고
    • Human neprilysin-2 (NEP2) and NEP display distinct subcellular localisations and substrate preferences
    • Whyteside AR, Turner AJ. Human neprilysin-2 (NEP2) and NEP display distinct subcellular localisations and substrate preferences. FEBS Lett 2008;582:2382-86
    • (2008) FEBS Lett , vol.582 , pp. 2382-2386
    • Whyteside, A.R.1    Turner, A.J.2
  • 86
    • 0036566092 scopus 로고    scopus 로고
    • Cell-specific activity of neprilysin 2 isoforms and enzymic specificity compared with neprilysin
    • DOI 10.1042/0264-6021:3630697
    • Rose C, Voisin S, Gros C, et al. Cell-specific activity of neprilysin 2 isoforms and enzymic specificity compared with neprilysin. Biochem J 2002;363:697-705 (Pubitemid 34526390)
    • (2002) Biochemical Journal , vol.363 , Issue.3 , pp. 697-705
    • Rose, C.1    Voisin, S.2    Gros, C.3    Schwartz, J.-C.4    Ouimet, T.5
  • 87
    • 0033527579 scopus 로고    scopus 로고
    • Molecular identification and characterization of novel membrane-bound metalloprotease, the soluble secreted form of which hydrolyzes a variety of vasoactive peptides
    • Ikeda K, Emoto N, Raharjo SB, et al. Molecular identification and characterization of novel membrane-bound metalloprotease, the soluble secreted form of which hydrolyzes a variety of vasoactive peptides. J Biol Chem 1999;274:32469-77
    • (1999) J Biol Chem , vol.274 , pp. 32469-32477
    • Ikeda, K.1    Emoto, N.2    Raharjo, S.B.3
  • 88
    • 0031440109 scopus 로고    scopus 로고
    • Human endothelin-converting enzyme (ECE-1): Three isoforms with distinct subcellular localizations
    • Schweizer A, Valdenaire O, Nelbock P, et al. Human endothelinconverting enzyme (ECE-1): Three isoforms with distinct subcellular localizations. Biochem J 1997;328(Pt 3):871-77 (Pubitemid 28005788)
    • (1997) Biochemical Journal , vol.328 , Issue.3 , pp. 871-877
    • Schweizer, A.1    Valdenaire, O.2    Nelbock, P.3
  • 89
    • 77956497978 scopus 로고    scopus 로고
    • Endothelin-converting enzyme-1 in Alzheimer's disease and vascular dementia
    • Palmer JC, Kehoe PG, Love S. Endothelin-converting enzyme-1 in Alzheimer's disease and vascular dementia. Neuropathol Appl Neurobiol 2010;36:487-97
    • (2010) Neuropathol Appl Neurobiol , vol.36 , pp. 487-497
    • Palmer, J.C.1    Kehoe, P.G.2    Love, S.3
  • 90
    • 0031627512 scopus 로고    scopus 로고
    • Endothelin-converting enzyme in the human vasculature: Evidence for differential conversion of big endothelin-3 by endothelial and smooth-muscle cells
    • Davenport AP, Kuc RE, Mockridge JW. Endothelin-converting enzyme in the human vasculature: Evidence for differential conversion of big endothelin-3 by endothelial and smooth-muscle cells. J Cardiovasc Pharmacol 1998;31(Suppl 1):S1-S3 (Pubitemid 128709852)
    • (1998) Journal of Cardiovascular Pharmacology , vol.31 , Issue.SUPPL. 1
    • Davenport, A.P.1    Kuc, R.E.2    Mockridge, J.W.3
  • 91
    • 0027992642 scopus 로고
    • ECE-1: A membrane-bound metalloprotease that catalyzes the proteolytic activation of big endothelin-1
    • DOI 10.1016/0092-8674(94)90425-1
    • Xu D, Emoto N, Giaid A, et al. ECE-1: A membrane-bound metalloprotease that catalyzes the proteolytic activation of big endothelin-1. Cell 1994;78:473-85 (Pubitemid 24250811)
    • (1994) Cell , vol.78 , Issue.3 , pp. 473-485
    • Xu, D.1    Emoto, N.2    Giaid, A.3    Slaughter, C.4    Kaw, S.5    DeWit, D.6    Yanagisawa, M.7
  • 92
    • 0037462769 scopus 로고    scopus 로고
    • Alzheimer's disease β-amyloid peptide is increased in mice deficient in endothelin-converting enzyme
    • DOI 10.1074/jbc.C200642200
    • Eckman EA, Watson M, Marlow L, et al. Alzheimer's disease A-amyloid peptide is increased in mice deficient in endothelin-converting enzyme. J Biol Chem 2003;278:2081-84 (Pubitemid 36801269)
    • (2003) Journal of Biological Chemistry , vol.278 , Issue.4 , pp. 2081-2084
    • Eckman, E.A.1    Watson, M.2    Marlow, L.3    Sambamurti, K.4    Eckman, C.B.5
  • 94
    • 2342544395 scopus 로고    scopus 로고
    • Immunolocalisation of endothelin-1 in human brain
    • DOI 10.1016/j.jchemneu.2004.03.007, PII S0891061804000377
    • Naidoo V, Naidoo S, Raidoo DM. Immunolocalisation of endothelin-1 in human brain. J Chem Neuroanat 2004;27:193-200 (Pubitemid 38739358)
    • (2004) Journal of Chemical Neuroanatomy , vol.27 , Issue.3 , pp. 193-200
    • Naidoo, V.1    Naidoo, S.2    Raidoo, D.M.3
  • 95
    • 0030700341 scopus 로고    scopus 로고
    • Novel activity of endothelin-converting enzyme: Hydrolysis of bradykinin
    • Hoang MV, Turner AJ. Novel activity of endothelin-converting enzyme: Hydrolysis of bradykinin. Biochem J 1997;327:23-26 (Pubitemid 27455455)
    • (1997) Biochemical Journal , vol.327 , Issue.1 , pp. 23-26
    • Hoang, M.V.1    Turner, A.J.2
  • 96
    • 0033761812 scopus 로고    scopus 로고
    • Cellular expression of isoforms of endothelinconverting enzyme-1 (ECE-1c, ECE-1b and ECE-1a) and endothelinconverting enzyme-2
    • Davenport AP, Kuc RE. Cellular expression of isoforms of endothelinconverting enzyme-1 (ECE-1c, ECE-1b and ECE-1a) and endothelinconverting enzyme-2. J Cardiovasc Pharmacol 2000;36:S12-14
    • (2000) J Cardiovasc Pharmacol , vol.36
    • Davenport, A.P.1    Kuc, R.E.2
  • 97
    • 0038013935 scopus 로고    scopus 로고
    • Characterization of endothelin-converting enzyme-2: Implication for a role in the nonclassical processing of regulatory peptides
    • DOI 10.1074/jbc.M211242200
    • Mzhavia N, Pan H, Che FY, et al. Characterization of endothelinconverting enzyme-2. Implication for a role in the nonclassical processing of regulatory peptides. J Biol Chem 2003;278:14704-11 (Pubitemid 36799793)
    • (2003) Journal of Biological Chemistry , vol.278 , Issue.17 , pp. 14704-14711
    • Mzhavia, N.1    Pan, H.2    Che, F.-Y.3    Fricker, L.D.4    Devi, L.A.5
  • 98
    • 0029017876 scopus 로고
    • Endothelin-converting enzyme-2 is a membranebound, phosphoramidon- sensitive metalloprotease with acidic pH optimum
    • Emoto N, Yanagisawa M. Endothelin-converting enzyme-2 is a membranebound, phosphoramidon-sensitive metalloprotease with acidic pH optimum. J Biol Chem 1995;270:15262-68
    • (1995) J Biol Chem , vol.270 , pp. 15262-15268
    • Emoto, N.1    Yanagisawa, M.2
  • 99
    • 0034974108 scopus 로고    scopus 로고
    • Cortical alterations of angiotensin converting enzyme, angiotensin II and AT1 receptor in Alzheimer's dementia
    • DOI 10.1016/S0197-4580(00)00259-1, PII S0197458000002591
    • Savaskan E, Hock C, Olivieri G, et al. Cortical alterations of angiotensin converting enzyme, angiotensin II and AT1 receptor in Alzheimer's dementia. Neurobiol Aging 2001;22:541-46 (Pubitemid 32607737)
    • (2001) Neurobiology of Aging , vol.22 , Issue.4 , pp. 541-546
    • Savaskan, E.1    Hock, C.2    Olivieri, G.3    Bruttel, S.4    Rosenberg, C.5    Hulette, C.6    Muller-Spahn, F.7
  • 100
    • 40149093247 scopus 로고    scopus 로고
    • Angiotensin-converting enzyme (ACE) levels and activity in Alzheimer's disease, and relationship of perivascular ACE-1 to cerebral amyloid angiopathy
    • DOI 10.1111/j.1365-2990.2007.00885.x
    • Miners JS, Ashby E, Van Helmond Z, et al. Angiotensin-converting enzyme (ACE) levels and activity in Alzheimer's disease, and relationship of perivascular ACE-1 to cerebral amyloid angiopathy. Neuropathol Appl Neurobiol 2008;34:181-93 (Pubitemid 351327960)
    • (2008) Neuropathology and Applied Neurobiology , vol.34 , Issue.2 , pp. 181-193
    • Miners, J.S.1    Ashby, E.2    Van Helmond, Z.3    Chalmers, K.A.4    Palmer, L.E.5    Love, S.6    Kehoe, P.G.7
  • 101
    • 27844534846 scopus 로고    scopus 로고
    • Amyloid β-protein is degraded by cellular angiotensin-converting enzyme (ACE) and elevated by an ACE inhibitor
    • DOI 10.1074/jbc.M508460200
    • Hemming ML, Selkoe DJ. Amyloid A-protein is degraded by cellular angiotensin-converting enzyme (ACE) and elevated by an ACE inhibitor. J Biol Chem 2005;280:37644-50 (Pubitemid 41642372)
    • (2005) Journal of Biological Chemistry , vol.280 , Issue.45 , pp. 37644-37650
    • Hemming, M.L.1    Selkoe, D.J.2
  • 102
    • 2342537711 scopus 로고    scopus 로고
    • Angiotensin converting enzyme (ACE) and neprilysin hydrolyze neuropeptides: A brief history, the beginning and follow-ups to early studies
    • DOI 10.1016/j.peptides.2003.12.010, PII S0196978103004327
    • Skidgel RA, Erdos EG. Angiotensin converting enzyme (ACE) and neprilysin hydrolyze neuropeptides: A brief history, the beginning and follow-ups to early studies. Peptides 2004;25:521-25 (Pubitemid 38591559)
    • (2004) Peptides , vol.25 , Issue.3 , pp. 521-525
    • Skidgel, R.A.1    Erdos, E.G.2
  • 103
    • 0028102743 scopus 로고
    • A direct inhibitory effect of insulin on a cytosolic proteolytic complex containing insulin-degrading enzyme and multicatalytic proteinase
    • Duckworth WC, Bennett RG, Hamel FG. A direct inhibitory effect of insulin on a cytosolic proteolytic complex containing insulin-degrading enzyme and multicatalytic proteinase. J Biol Chem 1994;269:24575-80 (Pubitemid 24311707)
    • (1994) Journal of Biological Chemistry , vol.269 , Issue.40 , pp. 24575-24580
    • Duckworth, W.C.1    Bennett, R.G.2    Hamel, F.G.3
  • 105
    • 0023766957 scopus 로고
    • Cellular localization of insulin insulindegrading enzyme in rat liver using monoclonal antibodies specific for this enzyme
    • Akiyama H, Shii K, Yokono K, et al. Cellular localization of insulin insulindegrading enzyme in rat liver using monoclonal antibodies specific for this enzyme. Biochem Biophys Res Commun 1988;155:914-22
    • (1988) Biochem Biophys Res Commun , vol.155 , pp. 914-922
    • Akiyama, H.1    Shii, K.2    Yokono, K.3
  • 107
    • 0033605607 scopus 로고    scopus 로고
    • Insulin-degrading enzyme in the Alzheimer's disease brain: Prominent localization in neurons and senile plaques
    • DOI 10.1016/S0304-3940(99)00135-4, PII S0304394099001354
    • Bernstein HG, Ansorge S, Riederer P, et al. Insulin-degrading enzyme in the Alzheimer's disease brain: Prominent localization in neurons and senile plaques. Neurosci Lett 1999;263:161-64 (Pubitemid 29158953)
    • (1999) Neuroscience Letters , vol.263 , Issue.2-3 , pp. 161-164
    • Bernstein, H.-G.1    Ansorge, S.2    Riederer, P.3    Reiser, M.4    Frolich, L.5    Bogerts, B.6
  • 108
    • 0031690490 scopus 로고    scopus 로고
    • Insulin degradation: Progress and potential
    • DOI 10.1210/er.19.5.608
    • Duckworth WC, Bennett RG, Hamel FG. Insulin degradation: Progress and potential. Endocr Rev 1998;19:608-24 (Pubitemid 28467714)
    • (1998) Endocrine Reviews , vol.19 , Issue.5 , pp. 608-624
    • Duckworth, W.C.1    Bennett, R.G.2    Hamel, F.G.3
  • 109
    • 0021353578 scopus 로고
    • Degradation of insulin by isolated mouse pancreatic acini. Evidence for cell surface protease activity
    • Goldfine ID, Williams JA, Bailey AC, et al. Degradation of insulin by isolated mouse pancreatic acini. Evidence for cell surface protease activity. Diabetes 1984;33:64-72 (Pubitemid 14187205)
    • (1984) Diabetes , vol.33 , Issue.1 , pp. 64-72
    • Goldfine, I.D.1    Williams, J.A.2    Bailey, A.C.3
  • 110
    • 0024595382 scopus 로고
    • 125]monoiodoinsulin
    • Misbin RI, Almira EC. Degradation of insulin and insulin-like growth factors by enzyme purified from human erythrocytes. Comparison of degradation products observed with A14-and B26-[125I] monoiodoinsulin. Diabetes 1989;38:152-58 (Pubitemid 19054794)
    • (1989) Diabetes , vol.38 , Issue.2 , pp. 152-158
    • Misbin, R.I.1    Almira, E.C.2
  • 111
    • 75149166216 scopus 로고    scopus 로고
    • Insulin-degrading enzyme sorting in exosomes: A secretory pathway for a key brain amyloid-A degrading protease
    • Bulloj A, Leal MC, Xu H, et al. Insulin-degrading enzyme sorting in exosomes: A secretory pathway for a key brain amyloid-A degrading protease. J Alzheimers Dis 2010;19:79-95
    • (2010) J Alzheimers Dis , vol.19 , pp. 79-95
    • Bulloj, A.1    Leal, M.C.2    Xu, H.3
  • 116
    • 11244276934 scopus 로고    scopus 로고
    • Insulin-degrading enzyme in brain microvessels: Proteolysis of amyloid β vasculotropic variants and reduced activity in cerebral amyloid angiopathy
    • DOI 10.1074/jbc.M407283200
    • Morelli L, Llovera RE, Mathov I, et al. Insulin-degrading enzyme in brain microvessels: Proteolysis of amyloid {A} vasculotropic variants and reduced activity in cerebral amyloid angiopathy. J Biol Chem 2004; 279:56004-13 (Pubitemid 40066608)
    • (2004) Journal of Biological Chemistry , vol.279 , Issue.53 , pp. 56004-56013
    • Morelli, L.1    Llovera, R.E.2    Mathov, I.3    Lue, L.-F.4    Frangione, B.5    Ghiso, J.6    Castano, E.M.7
  • 118
    • 0031550507 scopus 로고    scopus 로고
    • Overexpression of insulin degrading enzyme: Cellular localization and effects on insulin signaling
    • DOI 10.1006/bbrc.1997.6066
    • Seta KA, Roth RA, Gottschail PE. Overexpression of insulin degrading enzyme: Cellular localization and effects on insulin signaling. Biochem Biophys Res Commun 1997;231:167-71 (Pubitemid 27212131)
    • (1997) Biochemical and Biophysical Research Communications , vol.231 , Issue.1 , pp. 167-171
    • Seta, K.A.1    Roth, R.A.2
  • 119
    • 0037466395 scopus 로고    scopus 로고
    • β-amyloid induces the production of active, matrix-degrading proteases in cultured rat astrocytes
    • DOI 10.1016/S0006-8993(03)02344-8
    • Deb S, Wenjun Zhang J, et al. A-Amyloid induces the production of active, matrix-degrading proteases in cultured rat astrocytes. Brain Res 2003;970:205-13 (Pubitemid 36423196)
    • (2003) Brain Research , vol.970 , Issue.1-2 , pp. 205-213
    • Deb, S.1    Zhang, J.W.2    Gottschall, P.E.3
  • 120
    • 40149101997 scopus 로고    scopus 로고
    • MMP-2, -3 and -9 levels and activity are not related to Aβ load in the frontal cortex in Alzheimer's disease
    • DOI 10.1111/j.1365-2990.2007.00897.x
    • Baig S, Kehoe PG, Love S. MMP-2,-3 and-9 levels and activity are not related to AA load in the frontal cortex in Alzheimer's disease. Neuropathol Appl Neurobiol 2008;34:205-15 (Pubitemid 351327964)
    • (2008) Neuropathology and Applied Neurobiology , vol.34 , Issue.2 , pp. 205-215
    • Baig, S.1    Kehoe, P.G.2    Love, S.3
  • 121
    • 77956232864 scopus 로고    scopus 로고
    • Matrix metalloproteinase 2 (MMP-2) degrades soluble vasculotropic amyloid-A E22Q and L34V mutants, delaying their toxicity for human brain microvascular endothelial cells
    • Hernandez-Guillamon M, Mawhirt S, Fossati S, et al. Matrix metalloproteinase 2 (MMP-2) degrades soluble vasculotropic amyloid-A E22Q and L34V mutants, delaying their toxicity for human brain microvascular endothelial cells. J Biol Chem 2010;285:27144-58
    • (2010) J Biol Chem , vol.285 , pp. 27144-27158
    • Hernandez-Guillamon, M.1    Mawhirt, S.2    Fossati, S.3
  • 122
    • 0021971494 scopus 로고
    • Increase in vesicle permeability mediated by myelin basic protein: Effect of phosphorylation of basic protein
    • DOI 10.1021/bi00340a032
    • Cheifetz S, Boggs JM, Moscarello MA. Increase in vesicle permeability mediated by myelin basic protein: Effect of phosphorylation of basic protein. Biochemistry 1985;24:5170-75 (Pubitemid 15242623)
    • (1985) Biochemistry , vol.24 , Issue.19 , pp. 5170-5175
    • Cheifetz, S.1    Boggs, J.M.2    Moscarello, M.A.3
  • 123
    • 25444524820 scopus 로고    scopus 로고
    • Autocatalytic cleavage of myelin basic protein: An alternative to molecular mimicry
    • DOI 10.1021/bi051152f
    • D'Souza CA, Wood DD, She YM, et al. Autocatalytic cleavage of myelin basic protein: An alternative to molecular mimicry. Biochemistry 2005;44:12905-13 (Pubitemid 41377330)
    • (2005) Biochemistry , vol.44 , Issue.38 , pp. 12905-12913
    • D'Souza, C.A.1    Wood, D.D.2    She, Y.-M.3    Moscarello, M.A.4
  • 124
    • 77956613606 scopus 로고    scopus 로고
    • Plasminogen and plasmin in Alzheimer's disease
    • Barker R, Love S, Kehoe PG. Plasminogen and plasmin in Alzheimer's disease. Brain Res 2010;1355:7-15
    • (2010) Brain Res , vol.1355 , pp. 7-15
    • Barker, R.1    Love, S.2    Kehoe, P.G.3
  • 125
    • 0033567263 scopus 로고    scopus 로고
    • Exocytosis of active cathepsin B: Enzyme activity at pH 7.0, inhibition and molecular mass
    • DOI 10.1046/j.1432-1327.1999.00582.x
    • Linebaugh BE, Sameni M, Day NA, et al. Exocytosis of active cathepsin B enzyme activity at pH 7.0, inhibition and molecular mass. Eur J Biochem 1999;264:100-9 (Pubitemid 29385872)
    • (1999) European Journal of Biochemistry , vol.264 , Issue.1 , pp. 100-109
    • Linebaugh, B.E.1    Sameni, M.2    Day, N.A.3    Sloane, B.F.4    Keppler, D.5
  • 127
    • 0030899822 scopus 로고    scopus 로고
    • Mammalian membrane metallopeptidases: NEP, ECE, KELL, and PEX
    • Turner AJ, Tanzawa K. Mammalian membrane metallopeptidases: NEP, ECE, KELL, and PEX. FASEB J 1997;11:355-64 (Pubitemid 27193940)
    • (1997) FASEB Journal , vol.11 , Issue.5 , pp. 355-364
    • Turner, A.J.1    Tanzawa, K.2
  • 128
    • 0035112440 scopus 로고    scopus 로고
    • The neprilysin (NEP) family of zinc metalloendopeptidases: Genomics and function
    • DOI 10.1002/1521-1878(200103)23:3<261::AID-BIES1036>3.0.CO;2-K
    • Turner AJ, Isaac RE, Coates D. The neprilysin (NEP) family of zinc metalloendopeptidases: Genomics and function. Bioessays 2001;23:261-69 (Pubitemid 32192145)
    • (2001) BioEssays , vol.23 , Issue.3 , pp. 261-269
    • Turner, A.J.1    Elwyn Isaac, R.2    Coates, D.3
  • 131
    • 33645505550 scopus 로고    scopus 로고
    • Effects of secreted oligomers of amyloid A-protein on hippocampal synaptic plasticity: A potent role for trimers
    • Townsend M, Shankar GM, Mehta T, et al. Effects of secreted oligomers of amyloid A-protein on hippocampal synaptic plasticity: A potent role for trimers. J Physiol 2006;572:477-92
    • (2006) J Physiol , vol.572 , pp. 477-492
    • Townsend, M.1    Shankar, G.M.2    Mehta, T.3
  • 132
    • 49149124343 scopus 로고    scopus 로고
    • Amyloid-A protein dimers isolated directly from Alzheimer's brains impair synaptic plasticity and memory
    • Shankar GM, Li S, Mehta TH, et al. Amyloid-A protein dimers isolated directly from Alzheimer's brains impair synaptic plasticity and memory. Nat Med 2008;14:837-42
    • (2008) Nat Med , vol.14 , pp. 837-842
    • Shankar, G.M.1    Li, S.2    Mehta, T.H.3
  • 133
    • 33645038471 scopus 로고    scopus 로고
    • A specific amyloid-A protein assembly in the brain impairs memory
    • Lesne S, Koh MT, Kotilinek L, et al. A specific amyloid-A protein assembly in the brain impairs memory. Nature 2006;440:352-57
    • (2006) Nature , vol.440 , pp. 352-357
    • Lesne, S.1    Koh, M.T.2    Kotilinek, L.3
  • 134
    • 60549107033 scopus 로고    scopus 로고
    • A specific enzyme-linked immunosorbent assay for measuring A-amyloid protein oligomers in human plasma and brain tissue of patients with Alzheimer disease
    • Xia W, Yang T, Shankar G, et al. A specific enzyme-linked immunosorbent assay for measuring A-amyloid protein oligomers in human plasma and brain tissue of patients with Alzheimer disease. Arch Neurol 2009;66:190-99
    • (2009) Arch Neurol , vol.66 , pp. 190-199
    • Xia, W.1    Yang, T.2    Shankar, G.3
  • 135
    • 75949101330 scopus 로고    scopus 로고
    • Oligomeric AA in Alzheimer's disease: Relationship to plaque and tangle pathology. APOE genotype and cerebral amyloid angiopathy
    • van Helmond Z, Miners JS, Kehoe PG, Love S. Oligomeric AA in Alzheimer's disease: Relationship to plaque and tangle pathology, APOE genotype and cerebral amyloid angiopathy. Brain Pathol 2010; 20:468-80
    • (2010) Brain Pathol , vol.20 , pp. 468-480
    • Van Helmond, Z.1    Miners, J.S.2    Kehoe, P.G.3    Love, S.4
  • 136
    • 80051569682 scopus 로고    scopus 로고
    • Amyloid-dependent and amyloid-independent stages of Alzheimer disease
    • Hyman BT. Amyloid-dependent and amyloid-independent stages of Alzheimer disease. Arch Neurol 2011;68:1062-64
    • (2011) Arch Neurol , vol.68 , pp. 1062-1064
    • Hyman, B.T.1
  • 138
    • 79954632126 scopus 로고    scopus 로고
    • Mechanism mediating oligomeric AA clearance by naive primary microglia
    • Yang CN, Shiao YJ, Shie FS, et al. Mechanism mediating oligomeric AA clearance by naive primary microglia. Neurobiol Dis 2011;42: 221-30
    • (2011) Neurobiol Dis , vol.42 , pp. 221-230
    • Yang, C.N.1    Shiao, Y.J.2    Shie, F.S.3
  • 140
    • 0028981219 scopus 로고
    • Structureactivity analyses of A-amyloid peptides: Contributions of the A 25-35 region to aggregation and neurotoxicity
    • Pike CJ, Walencewicz-Wasserman AJ, Kosmoski J, et al. Structureactivity analyses of A-amyloid peptides: Contributions of the A 25-35 region to aggregation and neurotoxicity. J Neurochem 1995;64:253-65
    • (1995) J Neurochem , vol.64 , pp. 253-265
    • Pike, C.J.1    Walencewicz-Wasserman, A.J.2    Kosmoski, J.3
  • 141
    • 33646117738 scopus 로고    scopus 로고
    • Amyloid A peptide (25-35) activates protein kinase C leading to cyclooxygenase-2 induction and prostaglandin E2 release in primary midbrain astrocytes
    • Hull M, Muksch B, Akundi RS, et al. Amyloid A peptide (25-35) activates protein kinase C leading to cyclooxygenase-2 induction and prostaglandin E2 release in primary midbrain astrocytes. Neurochem Int 2006;48:663-72
    • (2006) Neurochem Int , vol.48 , pp. 663-672
    • Hull, M.1    Muksch, B.2    Akundi, R.S.3
  • 143
    • 0346099098 scopus 로고    scopus 로고
    • Residues 17-20 and 30-35 of Beta-Amyloid Play Critical Roles in Aggregation
    • DOI 10.1002/jnr.10859
    • Liu R, McAllister C, Lyubchenko Y, et al. Residues 17-20 and 30-35 of A-amyloid play critical roles in aggregation. J Neurosci Res 2004;75: 162-71 (Pubitemid 38049387)
    • (2004) Journal of Neuroscience Research , vol.75 , Issue.2 , pp. 162-171
    • Liu, R.1    McAllister, C.2    Lyubchenko, Y.3    Sierks, M.R.4
  • 144
    • 0035877747 scopus 로고    scopus 로고
    • Neprilysin degrades both amyloid A peptides 1-40 and 1-42 most rapidly and efficiently among thiorphan-and phosphoramidon-sensitive endopeptidases
    • Shirotani K, Tsubuki S, Iwata N, et al. Neprilysin degrades both amyloid A peptides 1-40 and 1-42 most rapidly and efficiently among thiorphan-and phosphoramidon-sensitive endopeptidases. J Biol Chem 2001;276:21895-901
    • (2001) J Biol Chem , vol.276 , pp. 21895-901
    • Shirotani, K.1    Tsubuki, S.2    Iwata, N.3
  • 145
    • 79251469086 scopus 로고    scopus 로고
    • Neprilysin-2 is an important A-amyloid degrading enzyme
    • Hafez D, Huang JY, Huynh AM, et al. Neprilysin-2 is an important A-amyloid degrading enzyme. Am J Pathol 2011;178:306-12
    • (2011) Am J Pathol , vol.178 , pp. 306-312
    • Hafez, D.1    Huang, J.Y.2    Huynh, A.M.3
  • 147
    • 40449136127 scopus 로고    scopus 로고
    • Activation of the amyloid cascade by intracerebroventricular injection of the protease inhibitor phosphoramidon
    • DOI 10.1159/000113692
    • Nisemblat Y, Belinson H, Dolev I, Michaelson DM. Activation of the amyloid cascade by intracerebroventricular injection of the protease inhibitor phosphoramidon. Neurodegener Dis 2008;5:166-69 (Pubitemid 351347840)
    • (2008) Neurodegenerative Diseases , vol.5 , Issue.3-4 , pp. 166-169
    • Nisemblat, Y.1    Belinson, H.2    Dolev, I.3    Michaelson, D.M.4
  • 149
    • 33947245344 scopus 로고    scopus 로고
    • Effects of prolonged angiotensin-converting enzyme inhibitor treatment on amyloid β-protein metabolism in mouse models of Alzheimer disease
    • DOI 10.1016/j.nbd.2007.01.004, PII S096999610700023X
    • Hemming ML, Selkoe DJ, Farris W. Effects of prolonged angiotensinconverting enzyme inhibitor treatment on amyloid A-protein metabolism in mouse models of Alzheimer disease. Neurobiol Dis 2007;26:273-81 (Pubitemid 46436503)
    • (2007) Neurobiology of Disease , vol.26 , Issue.1 , pp. 273-281
    • Hemming, M.L.1    Selkoe, D.J.2    Farris, W.3
  • 153
    • 33645114238 scopus 로고    scopus 로고
    • Inhibition of neprilysin by infusion of thiorphan into the hippocampus
    • Zou LB, Mouri A, Iwata N, et al. Inhibition of neprilysin by infusion of thiorphan into the hippocampus causes an accumulation of amyloid-A and impairment of learning and memory. J Pharmacol Exp Ther 2006; 317:334-40
    • (2006) J Pharmacol Exp Ther , vol.317 , pp. 334-340
    • Zou, L.B.1    Mouri, A.2    Iwata, N.3
  • 154
    • 31444448250 scopus 로고    scopus 로고
    • Inhibition of neprilysin by thiorphan (i.c.v.) causes an accumulation of amyloid β and impairment of learning and memory
    • DOI 10.1016/j.bbr.2005.10.014, PII S0166432805004584
    • Mouri A, Zou LB, Iwata N, et al. Inhibition of neprilysin by thiorphan (i.c.v.) causes an accumulation of amyloid A and impairment of learning and memory. Behav Brain Res 2006;168:83-91 (Pubitemid 43152056)
    • (2006) Behavioural Brain Research , vol.168 , Issue.1 , pp. 83-91
    • Mouri, A.1    Zou, L.-B.2    Iwata, N.3    Saido, T.C.4    Wang, D.5    Wang, M.-W.6    Noda, Y.7    Nabeshima, T.8
  • 156
    • 60849097262 scopus 로고    scopus 로고
    • Neprilysin overexpression inhibits plaque formation but fails to reduce pathogenic AA oligomers and associated cognitive deficits in human amyloid precursor protein transgenic mice
    • Meilandt WJ, Cisse M, Ho K, et al. Neprilysin overexpression inhibits plaque formation but fails to reduce pathogenic AA oligomers and associated cognitive deficits in human amyloid precursor protein transgenic mice. J Neurosci 2009;29:1977-86
    • (2009) J Neurosci , vol.29 , pp. 1977-1986
    • Meilandt, W.J.1    Cisse, M.2    Ho, K.3
  • 161
    • 57449117668 scopus 로고    scopus 로고
    • Long-term neprilysin gene transfer is associated with reduced levels of intracellular AA and behavioral improvement in APP transgenic mice
    • Spencer B, Marr RA, Rockenstein E, et al. Long-term neprilysin gene transfer is associated with reduced levels of intracellular AA and behavioral improvement in APP transgenic mice. BMC Neurosci 2008; 9:109
    • (2008) BMC Neurosci , vol.9 , pp. 109
    • Spencer, B.1    Marr, R.A.2    Rockenstein, E.3
  • 163
    • 0035846826 scopus 로고    scopus 로고
    • Reduced neprilysin in high plaque areas of Alzheimer brain: A possible relationship to deficient degradation of A-amyloid peptide
    • Yasojima K, Akiyama H, McGeer EG, et al. Reduced neprilysin in high plaque areas of Alzheimer brain: A possible relationship to deficient degradation of A-amyloid peptide. Neurosci Lett 2001;297:97-100
    • (2001) Neurosci Lett , vol.297 , pp. 97-100
    • Yasojima, K.1    Akiyama, H.2    McGeer, E.G.3
  • 164
    • 27544432457 scopus 로고    scopus 로고
    • Neprylisin decreases uniformly in Alzheimer's disease and in normal aging
    • DOI 10.1016/j.febslet.2005.09.054, PII S0014579305011725
    • Russo R, Borghi R, Markesbery W, et al. Neprylisin decreases uniformly in Alzheimer's disease and in normal aging. FEBS Lett 2005; 579:6027-30 (Pubitemid 41546241)
    • (2005) FEBS Letters , vol.579 , Issue.27 , pp. 6027-6030
    • Russo, R.1    Borghi, R.2    Markesbery, W.3    Tabaton, M.4    Piccini, A.5
  • 165
    • 0035369189 scopus 로고    scopus 로고
    • Immunohistochemical localization of neprilysin in the human cerebral cortex: Inverse association with vulnerability to amyloid β-protein (Aβ) deposition
    • DOI 10.1016/S0006-8993(01)02390-3, PII S0006899301023903
    • Akiyama H, Kondo H, Ikeda K, et al. Immunohistochemical localization of neprilysin in the human cerebral cortex: Inverse association with vulnerability to amyloid A-protein (AA) deposition. Brain Res 2001; 902:277-81 (Pubitemid 32510877)
    • (2001) Brain Research , vol.902 , Issue.2 , pp. 277-281
    • Akiyama, H.1    Kondo, H.2    Ikeda, K.3    Kato, M.4    McGeer, P.L.5
  • 166
    • 77956583512 scopus 로고    scopus 로고
    • Expression and functional profiling of neprilysin, insulin-degrading enzyme, and endothelin-converting enzyme in prospectively studied elderly and Alzheimer's brain
    • Wang S, Wang R, Chen L, et al. Expression and functional profiling of neprilysin, insulin-degrading enzyme, and endothelin-converting enzyme in prospectively studied elderly and Alzheimer's brain. J Neurochem 2010;115:47-57
    • (2010) J Neurochem , vol.115 , pp. 47-57
    • Wang, S.1    Wang, R.2    Chen, L.3
  • 168
    • 37549012213 scopus 로고    scopus 로고
    • Age-dependent decline of neprilysin in Alzheimer's disease and normal brain: Inverse correlation with AA levels
    • Hellstrom-Lindahl E, Ravid R, Nordberg A. Age-dependent decline of neprilysin in Alzheimer's disease and normal brain: Inverse correlation with AA levels. Neurobiol Aging 2008;29:210-21
    • (2008) Neurobiol Aging , vol.29 , pp. 210-221
    • Hellstrom-Lindahl, E.1    Ravid, R.2    Nordberg, A.3
  • 169
    • 37049011221 scopus 로고    scopus 로고
    • Immunocapture-based fluorometric assay for the measurement of neprilysin-specific enzyme activity in brain tissue homogenates and cerebrospinal fluid
    • DOI 10.1016/j.jneumeth.2007.08.012, PII S0165027007004207
    • Miners JS, Verbeek MM, Rikkert MO, et al. Immunocapture-based fluorometric assay for the measurement of neprilysin-specific enzyme activity in brain tissue homogenates and cerebrospinal fluid. J Neurosci Methods 2008;167:229-36 (Pubitemid 350245527)
    • (2008) Journal of Neuroscience Methods , vol.167 , Issue.2 , pp. 229-236
    • Miners, J.S.1    Verbeek, M.M.2    Rikkert, M.O.3    Kehoe, P.G.4    Love, S.5
  • 170
    • 39749194421 scopus 로고    scopus 로고
    • Immunocapture-based fluorometric assay for the measurement of insulin-degrading enzyme activity in brain tissue homogenates
    • Miners JS, Kehoe PG, Love S. Immunocapture-based fluorometric assay for the measurement of insulin-degrading enzyme activity in brain tissue homogenates. J Neurosci Methods 2008;169:177-81
    • (2008) J Neurosci Methods , vol.169 , pp. 177-181
    • Miners, J.S.1    Kehoe, P.G.2    Love, S.3
  • 171
    • 77953193048 scopus 로고    scopus 로고
    • Changes with age in the activities of A-secretase and the AA-degrading enzymes neprilysin, insulin-degrading enzyme and angiotensin-converting enzyme
    • Miners JS, van Helmond Z, Kehoe PG, et al. Changes with age in the activities of A-secretase and the AA-degrading enzymes neprilysin, insulin-degrading enzyme and angiotensin-converting enzyme. Brain Pathol 2010;20:794-802
    • (2010) Brain Pathol , vol.20 , pp. 794-802
    • Miners, J.S.1    Van Helmond, Z.2    Kehoe, P.G.3
  • 173
    • 73349089956 scopus 로고    scopus 로고
    • Increased matrix metalloproteinase 9 activity in mild cognitive impairment
    • Bruno MA, Mufson EJ, Wuu J, et al. Increased matrix metalloproteinase 9 activity in mild cognitive impairment. J Neuropathol Exp Neurol 2009;68:1309-18
    • (2009) J Neuropathol Exp Neurol , vol.68 , pp. 1309-1318
    • Bruno, M.A.1    Mufson, E.J.2    Wuu, J.3
  • 174
    • 79751521177 scopus 로고    scopus 로고
    • Higher cathepsin B levels in plasma in Alzheimer's disease compared to healthy controls
    • Sundelof J, Sundstrom J, Hansson O, et al. Higher cathepsin B levels in plasma in Alzheimer's disease compared to healthy controls. J Alzheimers Dis 2010;22:1223-30
    • (2010) J Alzheimers Dis , vol.22 , pp. 1223-1230
    • Sundelof, J.1    Sundstrom, J.2    Hansson, O.3
  • 176
    • 58549086590 scopus 로고    scopus 로고
    • Effects of HNE-modification induced by AA on neprilysin expression and activity in SH-SY5Y cells
    • Wang R, Wang S, Malter JS, et al. Effects of HNE-modification induced by AA on neprilysin expression and activity in SH-SY5Y cells. J Neurochem 2009;108:1072-82
    • (2009) J Neurochem , vol.108 , pp. 1072-1082
    • Wang, R.1    Wang, S.2    Malter, J.S.3
  • 177
    • 0037144078 scopus 로고    scopus 로고
    • AA42-induced increase in neprilysin is associated with prevention of amyloid plaque formation in vivo
    • Mohajeri MH, Wollmer MA, Nitsch RM. AA42-induced increase in neprilysin is associated with prevention of amyloid plaque formation in vivo. J Biol Chem 2002;277:35460-65
    • (2002) J Biol Chem , vol.277 , pp. 35460-35465
    • Mohajeri, M.H.1    Wollmer, M.A.2    Nitsch, R.M.3
  • 178
    • 1642359902 scopus 로고    scopus 로고
    • Anti-amyloid activity of neprilysin in plaque-bearing mouse models of Alzheimer's disease
    • DOI 10.1016/S0014-5793(04)00169-3, PII S0014579304001693
    • Mohajeri MH, Kuehnle K, Li H, et al. Anti-amyloid activity of neprilysin in plaque-bearing mouse models of Alzheimer's disease. FEBS Lett 2004;562:16-21 (Pubitemid 38388441)
    • (2004) FEBS Letters , vol.562 , Issue.1-3 , pp. 16-21
    • Mohajeri, M.H.1    Kuehnle, K.2    Li, H.3    Poirier, R.4    Tracy, J.5    Nitsch, R.M.6
  • 179
    • 43849097891 scopus 로고    scopus 로고
    • Increased expression of AA degrading enzyme IDE in the cortex of transgenic mice with Alzheimer's disease-like neuropathology
    • Vepsalainen S, Hiltunen M, Helisalmi S, et al. Increased expression of AA degrading enzyme IDE in the cortex of transgenic mice with Alzheimer's disease-like neuropathology. Neurosci Lett 2008;438:216-20
    • (2008) Neurosci Lett , vol.438 , pp. 216-220
    • Vepsalainen, S.1    Hiltunen, M.2    Helisalmi, S.3
  • 182
    • 63849115094 scopus 로고    scopus 로고
    • Insights into the pathogenesis and pathogenicity of cerebral amyloid angiopathy
    • Love S, Miners S, Palmer J, et al. Insights into the pathogenesis and pathogenicity of cerebral amyloid angiopathy. Front Biosci 2009;14: 4778-92
    • (2009) Front Biosci , vol.14 , pp. 4778-4792
    • Love, S.1    Miners, S.2    Palmer, J.3
  • 183
    • 61649094266 scopus 로고    scopus 로고
    • Population studies of sporadic cerebral amyloid angiopathy and dementia: A systematic review
    • Keage HA, Carare RO, Friedland RP, et al. Population studies of sporadic cerebral amyloid angiopathy and dementia: A systematic review. BMC Neurol 2009;9:3
    • (2009) BMC Neurol , vol.9 , pp. 3
    • Keage, H.A.1    Carare, R.O.2    Friedland, R.P.3
  • 184
    • 0033809909 scopus 로고    scopus 로고
    • Prominent cerebral amyloid angiopathy in transgenic mice overexpressing the London mutant of human APP in neurons
    • Van Dorpe J, Smeijers L, Dewachter I, et al. Prominent cerebral amyloid angiopathy in transgenic mice overexpressing the London mutant of human APP in neurons. Am J Pathol 2000;157:1283-98
    • (2000) Am J Pathol , vol.157 , pp. 1283-1298
    • Van Dorpe, J.1    Smeijers, L.2    Dewachter, I.3
  • 185
    • 13044265226 scopus 로고    scopus 로고
    • Neuronal overexpression of mutant amyloid precursor protein results in prominent deposition of cerebrovascular amyloid
    • Calhoun ME, Burgermeister P, Phinney AL, et al. Neuronal overexpression of mutant amyloid precursor protein results in prominent deposition of cerebrovascular amyloid. Proc Natl Acad Sci USA 1999; 96:14088-93
    • (1999) Proc Natl Acad Sci USA , vol.96 , pp. 14088-14093
    • Calhoun, M.E.1    Burgermeister, P.2    Phinney, A.L.3
  • 187
    • 0028915895 scopus 로고
    • Amyloid A protein (AA) in Alzheimer's disease brain. Biochemical and immunocytochemical analysis with antibodies specific for forms ending at AA40 or AA42(43)
    • Gravina SA, Ho L, Eckman CB, et al. Amyloid A protein (AA) in Alzheimer's disease brain. Biochemical and immunocytochemical analysis with antibodies specific for forms ending at AA40 or AA42(43). J Biol Chem 1995;270:7013-16
    • (1995) J Biol Chem , vol.270 , pp. 7013-7016
    • Gravina, S.A.1    Ho, L.2    Eckman, C.B.3
  • 188
    • 67349118757 scopus 로고    scopus 로고
    • Microvasculature changes and cerebral amyloid angiopathy in Alzheimer's disease and their potential impact on therapy
    • Weller RO, Boche D, Nicoll JA. Microvasculature changes and cerebral amyloid angiopathy in Alzheimer's disease and their potential impact on therapy. Acta Neuropathol 2009;118:87-102
    • (2009) Acta Neuropathol , vol.118 , pp. 87-102
    • Weller, R.O.1    Boche, D.2    Nicoll, J.A.3
  • 189
    • 79954623282 scopus 로고    scopus 로고
    • Perivascular drainage of solutes is impaired in the ageing mouse brain and in the presence of cerebral amyloid angiopathy
    • Hawkes CA, Hartig W, Kacza J, et al. Perivascular drainage of solutes is impaired in the ageing mouse brain and in the presence of cerebral amyloid angiopathy. Acta Neuropathol 2011;121:431-43
    • (2011) Acta Neuropathol , vol.121 , pp. 431-443
    • Hawkes, C.A.1    Hartig, W.2    Kacza, J.3
  • 191
    • 80054912279 scopus 로고    scopus 로고
    • LRP-1Ymediated uptake of AA by cerebrovascular smooth muscle cells: Relationship to cytotoxicity
    • Ruzali W, Kehoe P, Love S. LRP-1Ymediated uptake of AA by cerebrovascular smooth muscle cells: Relationship to cytotoxicity. Neuropathol Appl Neurobiol 2011;37:10
    • (2011) Neuropathol Appl Neurobiol , vol.37 , pp. 10
    • Ruzali, W.1    Kehoe, P.2    Love, S.3
  • 192
    • 77956955758 scopus 로고    scopus 로고
    • The role of microglia in amyloid clearance from the AD brain
    • Lee CY, Landreth GE. The role of microglia in amyloid clearance from the AD brain. J Neural Transm 2010;117:949-60
    • (2010) J Neural Transm , vol.117 , pp. 949-960
    • Lee, C.Y.1    Landreth, G.E.2
  • 193
    • 32344440522 scopus 로고    scopus 로고
    • Bone marrow-derived microglia play a critical role in restricting senile plaque formation in Alzheimer's disease
    • DOI 10.1016/j.neuron.2006.01.022, PII S0896627306000754
    • Simard AR, Soulet D, Gowing G, et al. Bone marrowYderived microglia play a critical role in restricting senile plaque formation in Alzheimer's disease. Neuron 2006;49:489-502 (Pubitemid 43221886)
    • (2006) Neuron , vol.49 , Issue.4 , pp. 489-502
    • Simard, A.R.1    Soulet, D.2    Gowing, G.3    Julien, J.-P.4    Rivest, S.5
  • 194
    • 51149120624 scopus 로고    scopus 로고
    • Microglial dysfunction and defective A-amyloid clearance pathways in aging Alzheimer's disease mice
    • Hickman SE, Allison EK, El Khoury J. Microglial dysfunction and defective A-amyloid clearance pathways in aging Alzheimer's disease mice. J Neurosci 2008;28:8354-60
    • (2008) J Neurosci , vol.28 , pp. 8354-8360
    • Hickman, S.E.1    Allison, E.K.2    El Khoury, J.3
  • 199
    • 0037531198 scopus 로고    scopus 로고
    • Intracranially administered anti-Aβ antibodies reduce β-amyloid deposition by mechanisms both independent of and associated with microglial activation
    • Wilcock DM, DiCarlo G, Henderson D, et al. Intracranially administered anti-AA antibodies reduce A-amyloid deposition by mechanisms both independent of and associated with microglial activation. J Neurosci 2003;23:3745-51 (Pubitemid 36958481)
    • (2003) Journal of Neuroscience , vol.23 , Issue.9 , pp. 3745-3751
    • Wilcock, D.M.1    DiCarlo, G.2    Henderson, D.3    Jackson, J.4    Clarke, K.5    Ugen, K.E.6    Gordon, M.N.7    Morgan, D.8
  • 201
    • 0035695987 scopus 로고    scopus 로고
    • Gene expression profiling of amyloid beta peptide-stimulated human post-mortem brain microglia
    • DOI 10.1016/S0197-4580(01)00306-2, PII S0197458001003062
    • Walker DG, Lue LF, Beach TG. Gene expression profiling of amyloid A peptideYstimulated human post-mortem brain microglia. Neurobiol Aging 2001;22:957-66 (Pubitemid 34066564)
    • (2001) Neurobiology of Aging , vol.22 , Issue.6 , pp. 957-966
    • Walker, D.G.1    Lue, L.-F.2    Beach, T.G.3
  • 202
    • 0037215645 scopus 로고    scopus 로고
    • Role of microglia in inflammation-mediated neurodegenerative diseases: Mechanisms and strategies for therapeutic intervention
    • DOI 10.1124/jpet.102.035048
    • Liu B, Hong JS. Role of microglia in inflammation-mediated neurodegenerative diseases: Mechanisms and strategies for therapeutic intervention. J Pharmacol Exp Ther 2003;304:1-7 (Pubitemid 36008905)
    • (2003) Journal of Pharmacology and Experimental Therapeutics , vol.304 , Issue.1 , pp. 1-7
    • Liu, B.1    Hong, J.-S.2
  • 205
    • 0030222037 scopus 로고    scopus 로고
    • Microglia: A sensor for pathological events in the CNS
    • DOI 10.1016/0166-2236(96)10049-7
    • Kreutzberg GW. Microglia: A sensor for pathological events in the CNS. Trends Neurosci 1996;19:312-18 (Pubitemid 26349059)
    • (1996) Trends in Neurosciences , vol.19 , Issue.8 , pp. 312-318
    • Kreutzberg, G.W.1
  • 206
    • 0037265240 scopus 로고    scopus 로고
    • Alternative activation of macrophages
    • DOI 10.1038/nri978
    • Gordon S. Alternative activation of macrophages. Nat Rev Immunol 2003;3:23-35 (Pubitemid 37328697)
    • (2003) Nature Reviews Immunology , vol.3 , Issue.1 , pp. 23-35
    • Gordon, S.1
  • 207
    • 24744467358 scopus 로고    scopus 로고
    • Microglial phagocytosis induced by fibrillar β-amyloid and IgGs are differentially regulated by proinflammatory cytokines
    • DOI 10.1523/JNEUROSCI.1808-05.2005
    • Koenigsknecht-Talboo J, Landreth GE. Microglial phagocytosis induced by fibrillar A-amyloid and IgGs are differentially regulated by proinflammatory cytokines. J Neurosci 2005;25:8240-49 (Pubitemid 41292118)
    • (2005) Journal of Neuroscience , vol.25 , Issue.36 , pp. 8240-8249
    • Koenigsknecht-Talboo, J.1    Landreth, G.E.2
  • 208
    • 77952423753 scopus 로고    scopus 로고
    • Rosiglitazone rescues memory impairment in Alzheimer's transgenic mice: Mechanisms involving a reduced amyloid and tau pathology
    • Escribano L, Simon AM, Gimeno E, et al. Rosiglitazone rescues memory impairment in Alzheimer's transgenic mice: Mechanisms involving a reduced amyloid and tau pathology. Neuropsychopharmacology 2010;35: 1593-604
    • (2010) Neuropsychopharmacology , vol.35 , pp. 1593-604
    • Escribano, L.1    Simon, A.M.2    Gimeno, E.3
  • 209
    • 77950547609 scopus 로고    scopus 로고
    • Locus ceruleus controls Alzheimer's disease pathology by modulating microglial functions through norepinephrine
    • Heneka MT, Nadrigny F, Regen T, et al. Locus ceruleus controls Alzheimer's disease pathology by modulating microglial functions through norepinephrine. Proc Natl Acad Sci USA 2010;107:6058-63
    • (2010) Proc Natl Acad Sci USA , vol.107 , pp. 6058-6063
    • Heneka, M.T.1    Nadrigny, F.2    Regen, T.3
  • 211
    • 58749093072 scopus 로고    scopus 로고
    • IL-4Yinduced selective clearance of oligomeric A-amyloid peptide( 1-42) by rat primary type 2 microglia
    • Shimizu E, Kawahara K, Kajizono M, et al. IL-4Yinduced selective clearance of oligomeric A-amyloid peptide(1-42) by rat primary type 2 microglia. J Immunol 2008;181:6503-13
    • (2008) J Immunol , vol.181 , pp. 6503-6513
    • Shimizu, E.1    Kawahara, K.2    Kajizono, M.3
  • 212
    • 0030248270 scopus 로고    scopus 로고
    • Microglial cells internalize aggregates of the Alzheimer's disease amyloid β-protein via a scavenger receptor
    • DOI 10.1016/S0896-6273(00)80187-7
    • Paresce DM, Ghosh RN, Maxfield FR. Microglial cells internalize aggregates of the Alzheimer's disease amyloid A-protein via a scavenger receptor. Neuron 1996;17:553-65 (Pubitemid 26322226)
    • (1996) Neuron , vol.17 , Issue.3 , pp. 553-565
    • Paresce, D.M.1    Ghosh, R.N.2    Maxfield, F.R.3
  • 213
    • 7744235869 scopus 로고    scopus 로고
    • 1 integrin-dependent mechanism
    • DOI 10.1523/JNEUROSCI.2557-04.2004
    • Koenigsknecht J, Landreth G. Microglial phagocytosis of fibrillar A-amyloid through a A1 integrinYdependent mechanism. J Neurosci 2004;24:9838-46 (Pubitemid 39463564)
    • (2004) Journal of Neuroscience , vol.24 , Issue.44 , pp. 9838-9846
    • Koenigsknecht, J.1    Landreth, G.2
  • 215
    • 2542431100 scopus 로고    scopus 로고
    • The microglial phagocytic role with specific plaque types in the Alzheimer disease brain
    • DOI 10.1016/j.neurobiolaging.2003.12.026, PII S0197458004001046
    • D'Andrea MR, Cole GM, Ard MD. The microglial phagocytic role with specific plaque types in the Alzheimer disease brain. Neurobiol Aging 2004;25:675-83 (Pubitemid 38686491)
    • (2004) Neurobiology of Aging , vol.25 , Issue.5 , pp. 675-683
    • D'Andrea, M.R.1    Cole, G.M.2    Ard, M.D.3
  • 216
    • 0030664076 scopus 로고    scopus 로고
    • Slow degradation of aggregates of the Alzheimer's disease amyloid β- protein by microglial cells
    • DOI 10.1074/jbc.272.46.29390
    • Paresce DM, Chung H, Maxfield FR. Slow degradation of aggregates of the Alzheimer's disease amyloid A-protein by microglial cells. J Biol Chem 1997;272:29390-97 (Pubitemid 27498234)
    • (1997) Journal of Biological Chemistry , vol.272 , Issue.46 , pp. 29390-29397
    • Paresce, D.M.1    Chung, H.2    Maxfield, F.R.3
  • 217
    • 0033527637 scopus 로고    scopus 로고
    • Uptake, degradation, and release of fibrillar and soluble forms of Alzheimer's amyloid A-peptide by microglial cells
    • Chung H, Brazil MI, Soe TT, et al. Uptake, degradation, and release of fibrillar and soluble forms of Alzheimer's amyloid A-peptide by microglial cells. J Biol Chem 1999;274:32301-8
    • (1999) J Biol Chem , vol.274 , pp. 32301-8
    • Chung, H.1    Brazil, M.I.2    Soe, T.T.3
  • 220
    • 0017709822 scopus 로고
    • Degradation of myofibrillar proteins by cathepsins B and D
    • Schwartz W, Bird JW. Degradation of myofibrillar proteins by cathepsins B and D. Biochem J 1977;167:811-20 (Pubitemid 8240997)
    • (1977) Biochemical Journal , vol.167 , Issue.3 , pp. 811-820
    • Schwartz, W.N.1    Bird, J.W.C.2
  • 221
    • 0029804989 scopus 로고    scopus 로고
    • Degradation of Alzheimer's β-amyloid protein by human cathepsin D
    • McDermott JR, Gibson AM. Degradation of Alzheimer's A-amyloid protein by human cathepsin D. Neuroreport 1996;7:2163-66 (Pubitemid 26361838)
    • (1996) NeuroReport , vol.7 , Issue.13 , pp. 2163-2166
    • McDermott, J.R.1    Gibson, A.M.2
  • 222
    • 0030569293 scopus 로고    scopus 로고
    • Cathepsin D is involved in the clearance of Alzheimer's β-amyloid protein
    • DOI 10.1016/0014-5793(96)01087-3, PII S0014579396010873
    • Hamazaki H. Cathepsin D is involved in the clearance of Alzheimer's A-amyloid protein. FEBS Lett 1996;396:139-42 (Pubitemid 26377812)
    • (1996) FEBS Letters , vol.396 , Issue.2-3 , pp. 139-142
    • Hamazaki, H.1
  • 223
    • 65249170245 scopus 로고    scopus 로고
    • Microglia mediate the clearance of soluble AA through fluid phase macropinocytosis
    • Mandrekar S, Jiang Q, Lee CY, et al. Microglia mediate the clearance of soluble AA through fluid phase macropinocytosis. J Neurosci 2009;29: 4252-62
    • (2009) J Neurosci , vol.29 , pp. 4252-4262
    • Mandrekar, S.1    Jiang, Q.2    Lee, C.Y.3
  • 224
    • 78549284024 scopus 로고    scopus 로고
    • Statins promote the degradation of extracellular amyloid {A}-peptide by microglia via stimulation of exosome-associated insulin-degrading enzyme (IDE) secretion
    • Tamboli IY, Barth E, Christian L, et al. Statins promote the degradation of extracellular amyloid {A}-peptide by microglia via stimulation of exosome-associated insulin-degrading enzyme (IDE) secretion. J Biol Chem 2010;285:37405-14
    • (2010) J Biol Chem , vol.285 , pp. 37405-37414
    • Tamboli, I.Y.1    Barth, E.2    Christian, L.3
  • 225
    • 77956238695 scopus 로고    scopus 로고
    • Norepinephrine promotes microglia to uptake and degrade amyloid A peptide through upregulation of mouse formyl peptide receptor 2 and induction of insulin-degrading enzyme
    • Kong Y, Ruan L, Qian L, et al. Norepinephrine promotes microglia to uptake and degrade amyloid A peptide through upregulation of mouse formyl peptide receptor 2 and induction of insulin-degrading enzyme. J Neurosci 2010;30:11848-57
    • (2010) J Neurosci , vol.30 , pp. 11848-11857
    • Kong, Y.1    Ruan, L.2    Qian, L.3
  • 226
    • 27344441173 scopus 로고    scopus 로고
    • Metabolism of amyloid-β peptide and Alzheimer's disease
    • DOI 10.1016/j.pharmthera.2005.03.010, PII S0163725805000999
    • Iwata N, Higuchi M, Saido TC. Metabolism of amyloid-A peptide and Alzheimer's disease. Pharmacol Ther 2005;108:129-48 (Pubitemid 41527082)
    • (2005) Pharmacology and Therapeutics , vol.108 , Issue.2 , pp. 129-148
    • Iwata, N.1    Higuchi, M.2    Saido, T.C.3
  • 227
    • 23444452105 scopus 로고    scopus 로고
    • Expression of somatostatin receptor subtypes (SSTR1-5) in Alzheimer's disease brain: An immunohistochemical analysis
    • DOI 10.1016/j.neuroscience.2005.04.001, PII S030645220500401X
    • Kumar U. Expression of somatostatin receptor subtypes (SSTR1-5) in Alzheimer's disease brain: An immunohistochemical analysis. Neuroscience 2005;134:525-38 (Pubitemid 41111781)
    • (2005) Neuroscience , vol.134 , Issue.2 , pp. 525-538
    • Kumar, U.1
  • 228
    • 77954493503 scopus 로고    scopus 로고
    • Expression of somatostatin, cortistatin, and their receptors, as well as dopamine receptors, but not of neprilysin, are reduced in the temporal lobe of Alzheimer's disease patients
    • Gahete MD, Rubio A, Duran-Prado M, et al. Expression of somatostatin, cortistatin, and their receptors, as well as dopamine receptors, but not of neprilysin, are reduced in the temporal lobe of Alzheimer's disease patients. J Alzheimers Dis 2010;20:465-75
    • (2010) J Alzheimers Dis , vol.20 , pp. 465-475
    • Gahete, M.D.1    Rubio, A.2    Duran-Prado, M.3
  • 230
    • 13244292389 scopus 로고    scopus 로고
    • Protective effects of insulin-like growth factor-I on the somatostatinergic system in the temporal cortex of β-amyloid-treated rats
    • DOI 10.1111/j.1471-4159.2004.02889.x
    • Aguado-Llera D, Arilla-Ferreiro E, Campos-Barros A, et al. Protective effects of insulin-like growth factor-I on the somatostatinergic system in the temporal cortex of A-amyloidYtreated rats. J Neurochem 2005;92: 607-15 (Pubitemid 40189463)
    • (2005) Journal of Neurochemistry , vol.92 , Issue.3 , pp. 607-615
    • Aguado-Llera, D.1    Arilla-Ferreiro, E.2    Campos-Barros, A.3    Puebla-Jimenez, L.4    Barrios, V.5
  • 231
    • 58349118489 scopus 로고    scopus 로고
    • Minocycline prevents AA(25-35)Yinduced reduction of somatostatin and neprilysin content in rat temporal cortex
    • Burgos-Ramos E, Puebla-Jimenez L, Arilla-Ferreiro E. Minocycline prevents AA(25-35)Yinduced reduction of somatostatin and neprilysin content in rat temporal cortex. Life Sci 2009;84:205-10
    • (2009) Life Sci , vol.84 , pp. 205-210
    • Burgos-Ramos, E.1    Puebla-Jimenez, L.2    Arilla-Ferreiro, E.3
  • 232
    • 62649107650 scopus 로고    scopus 로고
    • The N-terminal tripeptide of insulin-like growth factor-I protects against A-amyloidY induced somatostatin depletion by calcium and glycogen synthase kinase 3A modulation
    • Burgos-Ramos E, Martos-Moreno GA, Lopez MG, et al. The N-terminal tripeptide of insulin-like growth factor-I protects against A-amyloidY induced somatostatin depletion by calcium and glycogen synthase kinase 3A modulation. J Neurochem 2009;109:360-70
    • (2009) J Neurochem , vol.109 , pp. 360-370
    • Burgos-Ramos, E.1    Martos-Moreno, G.A.2    Lopez, M.G.3
  • 234
    • 58049089518 scopus 로고    scopus 로고
    • Neprilysin gene expression requires binding of the amyloid precursor protein intracellular domain to its promoter: Implications for Alzheimer disease
    • Belyaev ND, Nalivaeva NN, Makova NZ, et al. Neprilysin gene expression requires binding of the amyloid precursor protein intracellular domain to its promoter: Implications for Alzheimer disease. EMBO Rep 2009;10:94-100
    • (2009) EMBO Rep , vol.10 , pp. 94-100
    • Belyaev, N.D.1    Nalivaeva, N.N.2    Makova, N.Z.3
  • 235
    • 34548515503 scopus 로고    scopus 로고
    • Gleevec increases levels of the amyloid precursor protein intracellular domain and of the amyloid-β- degrading enzyme neprilysin
    • DOI 10.1091/mbc.E07-01-0035
    • Eisele YS, Baumann M, Klebl B, et al. Gleevec increases levels of the amyloid precursor protein intracellular domain and of the amyloid-A degrading enzyme neprilysin. Mol Biol Cell 2007;18:3591-600 (Pubitemid 47378696)
    • (2007) Molecular Biology of the Cell , vol.18 , Issue.9 , pp. 3591-3600
    • Eisele, Y.S.1    Baumann, M.2    Klebl, B.3    Nordhammer, C.4    Jucker, M.5    Kilger, E.6
  • 237
    • 33847034860 scopus 로고    scopus 로고
    • Demethylation-linked activation of urokinase plasminogen activator is involved in progression of prostate cancer
    • Pulukuri SM, Estes N, Patel J, et al. Demethylation-linked activation of urokinase plasminogen activator is involved in progression of prostate cancer. Cancer Res 2007;67:930-39
    • (2007) Cancer Res , vol.67 , pp. 930-939
    • Pulukuri, S.M.1    Estes, N.2    Patel, J.3
  • 239
  • 241
    • 74049100935 scopus 로고    scopus 로고
    • Estrogen stimulates degradation of A-amyloid peptide by up-regulating neprilysin
    • Liang K, Yang L, Yin C, et al. Estrogen stimulates degradation of A-amyloid peptide by up-regulating neprilysin. J Biol Chem 2010;285: 935-42
    • (2010) J Biol Chem , vol.285 , pp. 935-942
    • Liang, K.1    Yang, L.2    Yin, C.3
  • 242
    • 70350221898 scopus 로고    scopus 로고
    • In vivo metabolism study of ginsenoside Re in rat using high-performance liquid chromatography coupled with tandem mass spectrometry
    • Yang L, Xu S, Liu C, et al. In vivo metabolism study of ginsenoside Re in rat using high-performance liquid chromatography coupled with tandem mass spectrometry. Anal Bioanal Chem 2009;395:1441-51
    • (2009) Anal Bioanal Chem , vol.395 , pp. 1441-1451
    • Yang, L.1    Xu, S.2    Liu, C.3
  • 243
    • 33645562327 scopus 로고    scopus 로고
    • Induction of neutral endopeptidase (NEP) activity of SK-N-SH cells by natural compounds from green tea
    • Ayoub S, Melzig MF. Induction of neutral endopeptidase (NEP) activity of SK-N-SH cells by natural compounds from green tea. J Pharm Pharmacol 2006;58:495-501
    • (2006) J Pharm Pharmacol , vol.58 , pp. 495-501
    • Ayoub, S.1    Melzig, M.F.2
  • 244
    • 0036674111 scopus 로고    scopus 로고
    • Induction of neutral endopeptidase and angiotensinconverting enzyme activity of SK-N-SH cells in vitro by quercetin and resveratrol
    • Melzig MF, Escher F. Induction of neutral endopeptidase and angiotensinconverting enzyme activity of SK-N-SH cells in vitro by quercetin and resveratrol. Pharmazie 2002;57:556-58
    • (2002) Pharmazie , vol.57 , pp. 556-558
    • Melzig, M.F.1    Escher, F.2
  • 245
    • 77953949978 scopus 로고    scopus 로고
    • Gene therapy in Alzheimer's diseaseVpotential for disease modification
    • Nilsson P, Iwata N, Muramatsu S, et al. Gene therapy in Alzheimer's diseaseVpotential for disease modification. J Cell Mol Med 2010;14: 741-57
    • (2010) J Cell Mol Med , vol.14 , pp. 741-757
    • Nilsson, P.1    Iwata, N.2    Muramatsu, S.3
  • 246
    • 77955773499 scopus 로고    scopus 로고
    • Circulating neprilysin clears brain amyloid
    • Liu Y, Studzinski C, Beckett T, et al. Circulating neprilysin clears brain amyloid. Mol Cell Neurosci 2010;45:101-7
    • (2010) Mol Cell Neurosci , vol.45 , pp. 101-7
    • Liu, Y.1    Studzinski, C.2    Beckett, T.3
  • 247
    • 77954851584 scopus 로고    scopus 로고
    • Trafficking CD11b-positive blood cells deliver therapeutic genes to the brain of amyloid-depositing transgenic mice
    • Lebson L, Nash K, Kamath S, et al. Trafficking CD11b-positive blood cells deliver therapeutic genes to the brain of amyloid-depositing transgenic mice. J Neurosci 2010;30:9651-58
    • (2010) J Neurosci , vol.30 , pp. 9651-9658
    • Lebson, L.1    Nash, K.2    Kamath, S.3
  • 248
    • 79551635864 scopus 로고    scopus 로고
    • Peripheral delivery of a CNS targeted, metalo-protease reduces AA toxicity in a mouse model of Alzheimer's disease
    • Spencer B, Marr RA, Gindi R, et al. Peripheral delivery of a CNS targeted, metalo-protease reduces AA toxicity in a mouse model of Alzheimer's disease. PLoS One 2011;6:e16575
    • (2011) PLoS One , vol.6
    • Spencer, B.1    Marr, R.A.2    Gindi, R.3
  • 249
    • 35348984094 scopus 로고    scopus 로고
    • Immune responses to adenovirus and adeno-associated vectors used for gene therapy of brain diseases: The role of immunological synapses in understanding the cell biology of neuroimmune interactions
    • DOI 10.2174/156652307782151498
    • Lowenstein PR, Mandel RJ, Xiong WD, et al. Immune responses to adenovirus and adeno-associated vectors used for gene therapy of brain diseases: The role of immunological synapses in understanding the cell biology of neuroimmune interactions. Curr Gene Ther 2007;7:347-60 (Pubitemid 47595777)
    • (2007) Current Gene Therapy , vol.7 , Issue.5 , pp. 347-360
    • Lowenstein, P.R.1    Mandel, R.J.2    Xiong, W.-D.3    Kroeger, K.4    Castro, M.G.5
  • 250
    • 49649123892 scopus 로고    scopus 로고
    • Overexpression of neprilysin reduces Alzheimer amyloid-A42 (AA42)Yinduced neuron loss and intraneuronal AA42 deposits but causes a reduction in cAMP-responsive element-binding proteinYmediated transcription, age-dependent axon pathology, and premature death in Drosophila
    • Iijima-Ando K, Hearn SA, Granger L, et al. Overexpression of neprilysin reduces Alzheimer amyloid-A42 (AA42)Yinduced neuron loss and intraneuronal AA42 deposits but causes a reduction in cAMP-responsive element-binding proteinYmediated transcription, age-dependent axon pathology, and premature death in Drosophila. J Biol Chem 2008;283: 19066-76
    • (2008) J Biol Chem , vol.283 , pp. 19066-19076
    • Iijima-Ando, K.1    Hearn, S.A.2    Granger, L.3
  • 251
    • 84887212630 scopus 로고    scopus 로고
    • Improved learning and memory in aged mice deficient in amyloid A-degrading neutral endopeptidase
    • Walther T, Albrecht D, Becker M, et al. Improved learning and memory in aged mice deficient in amyloid A-degrading neutral endopeptidase. PLoS One 2009;4:e4590
    • (2009) PLoS One , vol.4
    • Walther, T.1    Albrecht, D.2    Becker, M.3
  • 253
    • 34548456959 scopus 로고    scopus 로고
    • Reducing amyloid plaque burden via ex vivo gene delivery of an AA-degrading protease: A novel therapeutic approach to Alzheimer disease
    • Hemming ML, Patterson M, Reske-Nielsen C, et al. Reducing amyloid plaque burden via ex vivo gene delivery of an AA-degrading protease: A novel therapeutic approach to Alzheimer disease. PLoS Med 2007;4: e262
    • (2007) PLoS Med , vol.4
    • Hemming, M.L.1    Patterson, M.2    Reske-Nielsen, C.3
  • 254
    • 65549114145 scopus 로고    scopus 로고
    • Peripherally expressed neprilysin reduces brain amyloid burden: A novel approach for treating Alzheimer's disease
    • Guan H, Liu Y, Daily A, et al. Peripherally expressed neprilysin reduces brain amyloid burden: A novel approach for treating Alzheimer's disease. J Neurosci Res 2009;87:1462-73
    • (2009) J Neurosci Res , vol.87 , pp. 1462-1473
    • Guan, H.1    Liu, Y.2    Daily, A.3
  • 257
    • 8844269115 scopus 로고    scopus 로고
    • Why do we need new gene therapy viral vectors? Characteristics, limitations and future perspectives of viral vector transduction
    • Tomanin R, Scarpa M. Why do we need new gene therapy viral vectors? Characteristics, limitations and future perspectives of viral vector transduction. Curr Gene Ther 2004;4:357-72 (Pubitemid 39534752)
    • (2004) Current Gene Therapy , vol.4 , Issue.4 , pp. 357-372
    • Tomanin, R.1    Scarpa, M.2
  • 260
    • 20044374196 scopus 로고    scopus 로고
    • Safety and feasibility of convection-enhanced delivery of Cotara for the treatment of malignant glioma: Initial experience in 51 patients
    • DOI 10.1227/01.NEU.0000159649.71890.30
    • Patel SJ, Shapiro WR, Laske DW, et al. Safety and feasibility of convection-enhanced delivery of Cotara for the treatment of malignant glioma: Initial experience in 51 patients. Neurosurgery 2005;56:1243-52, discussion 52-3 (Pubitemid 40770440)
    • (2005) Neurosurgery , vol.56 , Issue.6 , pp. 1243-1252
    • Patel, S.J.1    Shapiro, W.R.2    Laske, D.W.3    Jensen, R.L.4    Asher, A.L.5    Wessels, B.W.6    Carpenter, S.P.7    Shan, J.S.8
  • 261
    • 85052608961 scopus 로고    scopus 로고
    • GDNF delivery for Parkinson's disease
    • Patel NK, Gill SS. GDNF delivery for Parkinson's disease. Acta Neurochir Suppl 2007;97:135-54
    • (2007) Acta Neurochir Suppl , vol.97 , pp. 135-154
    • Patel, N.K.1    Gill, S.S.2
  • 262
    • 22544461907 scopus 로고    scopus 로고
    • Glial cell line-derived neurotrophic factor induces neuronal sprouting in human brain
    • DOI 10.1038/nm0705-703
    • Love S, Plaha P, Patel NK, et al. Glial cell lineYderived neurotrophic factor induces neuronal sprouting in human brain. Nat Med 2005;11: 703-4 (Pubitemid 41021193)
    • (2005) Nature Medicine , vol.11 , Issue.7 , pp. 703-704
    • Love, S.1    Plaha, P.2    Patel, N.K.3    Hotton, G.R.4    Brooks, D.J.5    Gill, S.S.6
  • 264
    • 33747163632 scopus 로고    scopus 로고
    • Safety of intraparenchymal convection-enhanced delivery of cintredekin besudotox in early-phase studies
    • Kunwar S, Chang SM, Prados MD, et al. Safety of intraparenchymal convection-enhanced delivery of cintredekin besudotox in early-phase studies. Neurosurg Focus 2006;20:E15
    • (2006) Neurosurg Focus , vol.20
    • Kunwar, S.1    Chang, S.M.2    Prados, M.D.3
  • 267
    • 0032904141 scopus 로고    scopus 로고
    • Delivery, distribution, and neuronal uptake of exogenous mannose- terminal glucocerebrosidase in the intact rat brain
    • DOI 10.1023/A:1022578424693
    • Zirzow GC, Sanchez OA, Murray GJ, et al. Delivery, distribution, and neuronal uptake of exogenous mannose-terminal glucocerebrosidase in the intact rat brain. Neurochem Res 1999;24:301-5 (Pubitemid 29067974)
    • (1999) Neurochemical Research , vol.24 , Issue.2 , pp. 301-305
    • Zirzow, G.C.1    Sanchez, O.A.2    Murray, G.J.3    Brady, R.O.4    Oldfield, E.H.5
  • 268
    • 0029001712 scopus 로고
    • Convection-enhanced distribution of large molecules in gray matter during interstitial drug infusion
    • Lieberman DM, Laske DW, Morrison PF, et al. Convection-enhanced distribution of large molecules in gray matter during interstitial drug infusion. J Neurosurg 1995;82:1021-29
    • (1995) J Neurosurg , vol.82 , pp. 1021-1029
    • Lieberman, D.M.1    Laske, D.W.2    Morrison, P.F.3
  • 269
    • 0029874655 scopus 로고    scopus 로고
    • Increasing volume of distribution to the brain with interstitial infusion: Dose, rather than convection, might be the most important factor
    • discussion 52-4
    • Kroll RA, Pagel MA, Muldoon LL, et al. Increasing volume of distribution to the brain with interstitial infusion: Dose, rather than convection, might be the most important factor. Neurosurgery 1996;38:746-52, discussion 52-4
    • (1996) Neurosurgery , vol.38 , pp. 746-752
    • Kroll, R.A.1    Pagel, M.A.2    Muldoon, L.L.3
  • 271
    • 67849085408 scopus 로고    scopus 로고
    • Controlled dissemination of AAV vectors in the primate brain
    • Varenika V, Kells AP, Valles F, et al. Controlled dissemination of AAV vectors in the primate brain. Prog Brain Res 2009;175:163-72
    • (2009) Prog Brain Res , vol.175 , pp. 163-172
    • Varenika, V.1    Kells, A.P.2    Valles, F.3
  • 272
    • 40149099870 scopus 로고    scopus 로고
    • Solutes, but not cells, drain from the brain parenchyma along basement membranes of capillaries and arteries: Significance for cerebral amyloid angiopathy and neuroimmunology
    • DOI 10.1111/j.1365-2990.2007.00926.x
    • Carare RO, Bernardes-Silva M, Newman TA, et al. Solutes, but not cells, drain from the brain parenchyma along basement membranes of capillaries and arteries: Significance for cerebral amyloid angiopathy and neuroimmunology. Neuropathol Appl Neurobiol 2008;34:131-44 (Pubitemid 351327967)
    • (2008) Neuropathology and Applied Neurobiology , vol.34 , Issue.2 , pp. 131-144
    • Carare, R.O.1    Bernardes-Silva, M.2    Newman, T.A.3    Page, A.M.4    Nicoll, J.A.R.5    Perry, V.H.6    Weller, R.O.7
  • 273
    • 41149157623 scopus 로고    scopus 로고
    • Perivascular drainage of amyloid-β peptides from the brain and its failure in cerebral amyloid angiopathy and Alzheimer's disease
    • DOI 10.1111/j.1750-3639.2008.00133.x
    • Weller RO, Subash M, Preston SD, et al. Perivascular drainage of amyloid-A peptides from the brain and its failure in cerebral amyloid angiopathy and Alzheimer's disease. Brain Pathol 2008;18:253-66 (Pubitemid 351430402)
    • (2008) Brain Pathology , vol.18 , Issue.2 , pp. 253-266
    • Weller, R.O.1    Subash, M.2    Preston, S.D.3    Mazanti, I.4    Carare, R.O.5
  • 274
    • 57049154546 scopus 로고    scopus 로고
    • Lymphatic drainage of the brain and the pathophysiology of neurological disease
    • Weller RO, Djuanda E, Yow HY, et al. Lymphatic drainage of the brain and the pathophysiology of neurological disease. Acta Neuropathol 2009;117:1-14
    • (2009) Acta Neuropathol , vol.117 , pp. 1-14
    • Weller, R.O.1    Djuanda, E.2    Yow, H.Y.3
  • 275
    • 77952930303 scopus 로고    scopus 로고
    • Efficient processing of Alzheimer's disease amyloid-A peptides by neuroectodermally converted mesenchymal stem cells
    • Habisch HJ, Schmid B, von Arnim CA, et al. Efficient processing of Alzheimer's disease amyloid-A peptides by neuroectodermally converted mesenchymal stem cells. Stem Cells Dev 2010;19:629-33
    • (2010) Stem Cells Dev , vol.19 , pp. 629-633
    • Habisch, H.J.1    Schmid, B.2    Von Arnim, C.A.3
  • 276
    • 74949087489 scopus 로고    scopus 로고
    • Stem cells in human neurodegenerative disordersV time for clinical translation?
    • Lindvall O, Kokaia Z. Stem cells in human neurodegenerative disordersV time for clinical translation? J Clin Invest 2010;120:29-40
    • (2010) J Clin Invest , vol.120 , pp. 29-40
    • Lindvall, O.1    Kokaia, Z.2
  • 277
    • 65349181409 scopus 로고    scopus 로고
    • Prospects of stem cell therapy for replacing dopamine neurons in Parkinson's disease
    • Lindvall O, Kokaia Z. Prospects of stem cell therapy for replacing dopamine neurons in Parkinson's disease. Trends Pharmacol Sci 2009; 30:260-67
    • (2009) Trends Pharmacol Sci , vol.30 , pp. 260-267
    • Lindvall, O.1    Kokaia, Z.2


* 이 정보는 Elsevier사의 SCOPUS DB에서 KISTI가 분석하여 추출한 것입니다.