메뉴 건너뛰기




Volumn 71, Issue 1, 2014, Pages 1-20

Novel siRNA delivery strategy: A new "strand" in CNS translational medicine?

Author keywords

Gene therapy; Nanoparticles; Neurodegeneration; RNA interference

Indexed keywords

ADENOVIRUS VECTOR; AGN 211745; LENTIVIRUS VECTOR; NANOPARTICLE; PARVOVIRUS VECTOR; PEPTIDE; PF 04523655; SMALL INTERFERING RNA; SYL 1001; UNCLASSIFIED DRUG;

EID: 84891878822     PISSN: 1420682X     EISSN: 14209071     Source Type: Journal    
DOI: 10.1007/s00018-013-1310-8     Document Type: Review
Times cited : (16)

References (196)
  • 1
    • 0032545933 scopus 로고    scopus 로고
    • Potent and specific genetic interference by double-stranded RNA in caenorhabditis elegans
    • DOI 10.1038/35888
    • Fire A, Xu S, Montgomery MK, Kostas SA, Driver SE, Mello CC (1998) Potent and specific genetic interference by double-stranded RNA in Caenorhabditis elegans. Nature 391(6669):806-811. doi: 10.1038/35888 (Pubitemid 28099681)
    • (1998) Nature , vol.391 , Issue.6669 , pp. 806-811
    • Fire, A.1    Xu, S.2    Montgomery, M.K.3    Kostas, S.A.4    Driver, S.E.5    Mello, C.C.6
  • 2
    • 79960657081 scopus 로고    scopus 로고
    • Cortical plasticity as a new endpoint measurement for chronic pain
    • 10.1186/1744-8069-7-54 3157449 21798042
    • Zhuo M (2011) Cortical plasticity as a new endpoint measurement for chronic pain. Mol Pain 7:54. doi: 10.1186/1744-8069-7-54
    • (2011) Mol Pain , vol.7 , pp. 54
    • Zhuo, M.1
  • 3
    • 77949354685 scopus 로고    scopus 로고
    • Increasing tPA activity in astrocytes induced by multipotent mesenchymal stromal cells facilitate neurite outgrowth after stroke in the mouse
    • 10.1371/journal.pone.0009027 2815778 20140248
    • Xin H, Li Y, Shen LH, Liu X, Wang X, Zhang J, Pourabdollah-Nejad DS, Zhang C, Zhang L, Jiang H, Zhang ZG, Chopp M (2010) Increasing tPA activity in astrocytes induced by multipotent mesenchymal stromal cells facilitate neurite outgrowth after stroke in the mouse. PLoS ONE 5(2):e9027. doi: 10.1371/journal.pone.0009027
    • (2010) PLoS ONE , vol.5 , Issue.2 , pp. 9027
    • Xin, H.1    Li, Y.2    Shen, L.H.3    Liu, X.4    Wang, X.5    Zhang, J.6    Pourabdollah-Nejad, D.S.7    Zhang, C.8    Zhang, L.9    Jiang, H.10    Zhang, Z.G.11    Chopp, M.12
  • 4
    • 79960104144 scopus 로고    scopus 로고
    • MicroRNAs in development and disease
    • 10.1152/physrev.00006.2010 1:CAS:528:DC%2BC3MXhtFyjur%2FK 21742789
    • Sayed D, Abdellatif M (2011) MicroRNAs in development and disease. Physiol Rev 91(3):827-887. doi: 10.1152/physrev.00006.2010
    • (2011) Physiol Rev , vol.91 , Issue.3 , pp. 827-887
    • Sayed, D.1    Abdellatif, M.2
  • 5
    • 84863229335 scopus 로고    scopus 로고
    • Direct evidence of nuclear argonaute distribution during transcriptional silencing links the actin cytoskeleton to nuclear RNAi machinery in human cells
    • 10.1093/nar/gkr891 1:CAS:528:DC%2BC38XjtlGktrY%3D 3287199 22064859
    • Ahlenstiel CL, Lim HG, Cooper DA, Ishida T, Kelleher AD, Suzuki K (2012) Direct evidence of nuclear argonaute distribution during transcriptional silencing links the actin cytoskeleton to nuclear RNAi machinery in human cells. Nucleic Acids Res 40(4):1579-1595. doi: 10.1093/nar/gkr891
    • (2012) Nucleic Acids Res , vol.40 , Issue.4 , pp. 1579-1595
    • Ahlenstiel, C.L.1    Lim, H.G.2    Cooper, D.A.3    Ishida, T.4    Kelleher, A.D.5    Suzuki, K.6
  • 6
    • 84863116114 scopus 로고    scopus 로고
    • Expanding the action of duplex RNAs into the nucleus: Redirecting alternative splicing
    • 10.1093/nar/gkr780 1:CAS:528:DC%2BC38Xit1Ogtb8%3D 3273794 21948593
    • Liu J, Hu J, Corey DR (2012) Expanding the action of duplex RNAs into the nucleus: redirecting alternative splicing. Nucleic Acids Res 40(3):1240-1250. doi: 10.1093/nar/gkr780
    • (2012) Nucleic Acids Res , vol.40 , Issue.3 , pp. 1240-1250
    • Liu, J.1    Hu, J.2    Corey, D.R.3
  • 7
    • 24344500241 scopus 로고    scopus 로고
    • Sequence, chemical, and structural variation of small interfering RNAs and short hairpin RNAs and the effect on mammalian gene silencing
    • 10.1089/108729003321629638 1:CAS:528:DC%2BD3sXjsFejtLw%3D 12804036
    • Harborth J, Elbashir SM, Vandenburgh K, Manninga H, Scaringe SA, Weber K, Tuschl T (2003) Sequence, chemical, and structural variation of small interfering RNAs and short hairpin RNAs and the effect on mammalian gene silencing. Antisense Nucleic Acid Drug Dev 13(2):83-105. doi: 10.1089/108729003321629638
    • (2003) Antisense Nucleic Acid Drug Dev , vol.13 , Issue.2 , pp. 83-105
    • Harborth, J.1    Elbashir, S.M.2    Vandenburgh, K.3    Manninga, H.4    Scaringe, S.A.5    Weber, K.6    Tuschl, T.7
  • 9
    • 70350130799 scopus 로고    scopus 로고
    • Comparative assessment of siRNA and shRNA off target effects: What is slowing clinical development
    • 10.1038/cgt.2009.53 1:CAS:528:DC%2BD1MXhtVKjsbbJ 19713999
    • Rao DD, Senzer N, Cleary MA, Nemunaitis J (2009) Comparative assessment of siRNA and shRNA off target effects: what is slowing clinical development. Cancer Gene Ther 16(11):807-809. doi: 10.1038/cgt.2009.53
    • (2009) Cancer Gene Ther , vol.16 , Issue.11 , pp. 807-809
    • Rao, D.D.1    Senzer, N.2    Cleary, M.A.3    Nemunaitis, J.4
  • 10
    • 64549083734 scopus 로고    scopus 로고
    • The therapeutic potential of LNA-modified siRNAs: Reduction of off-target effects by chemical modification of the siRNA sequence
    • 10.1007/978-1-60327-547-7-9 1:CAS:528:DC%2BD1MXhtlertr0%3D 19301648
    • Fluiter K, Mook OR, Baas F (2009) The therapeutic potential of LNA-modified siRNAs: reduction of off-target effects by chemical modification of the siRNA sequence. Methods Mol Biol 487:189-203. doi: 10.1007/978-1-60327-547- 7-9
    • (2009) Methods Mol Biol , vol.487 , pp. 189-203
    • Fluiter, K.1    Mook, O.R.2    Baas, F.3
  • 11
    • 84867347162 scopus 로고    scopus 로고
    • SiRNA and cancer immunotherapy
    • 10.2217/imt.12.87 1:CAS:528:DC%2BC38XhsVOhsbvI 23046235
    • Ghafouri-Fard S (2012) siRNA and cancer immunotherapy. Immunotherapy 4(9):907-917. doi: 10.2217/imt.12.87
    • (2012) Immunotherapy , vol.4 , Issue.9 , pp. 907-917
    • Ghafouri-Fard, S.1
  • 12
    • 84872398430 scopus 로고    scopus 로고
    • Modification of small interfering RNAs to prevent off-target effects by the sense strand
    • 10.1016/j.nbt.2012.10.001 23124048
    • Nolte A, Ott K, Rohayem J, Walker T, Schlensak C, Wendel HP (2012) Modification of small interfering RNAs to prevent off-target effects by the sense strand. N Biotechnol. doi: 10.1016/j.nbt.2012.10.001
    • (2012) N Biotechnol
    • Nolte, A.1    Ott, K.2    Rohayem, J.3    Walker, T.4    Schlensak, C.5    Wendel, H.P.6
  • 13
    • 84868203868 scopus 로고    scopus 로고
    • Silica nanogelling of environment-responsive PEGylated polyplexes for enhanced stability and intracellular delivery of siRNA
    • 10.1016/j.biomaterials.2012.09.077 1:CAS:528:DC%2BC38XhsFant7nJ 23083934
    • Gouda N, Miyata K, Christie RJ, Suma T, Kishimura A, Fukushima S, Nomoto T, Liu X, Nishiyama N, Kataoka K (2013) Silica nanogelling of environment-responsive PEGylated polyplexes for enhanced stability and intracellular delivery of siRNA. Biomaterials 34(2):562-570. doi: 10.1016/j.biomaterials.2012.09.077
    • (2013) Biomaterials , vol.34 , Issue.2 , pp. 562-570
    • Gouda, N.1    Miyata, K.2    Christie, R.J.3    Suma, T.4    Kishimura, A.5    Fukushima, S.6    Nomoto, T.7    Liu, X.8    Nishiyama, N.9    Kataoka, K.10
  • 14
    • 22944466675 scopus 로고    scopus 로고
    • Delivering RNA interference to the mammalian brain
    • DOI 10.2174/1566523054546206
    • Fountaine TM, Wood MJ, Wade-Martins R (2005) Delivering RNA interference to the mammalian brain. Curr Gene Ther 5(4):399-410 (Pubitemid 41052431)
    • (2005) Current Gene Therapy , vol.5 , Issue.4 , pp. 399-410
    • Fountaine, T.M.1    Wood, M.J.A.2    Wade-Martins, R.3
  • 15
    • 79961168855 scopus 로고    scopus 로고
    • Barriers to non-viral vector-mediated gene delivery in the nervous system
    • 10.1007/s11095-010-0364-7 1:CAS:528:DC%2BC3MXktFWjsw%3D%3D 3130907 21225319
    • Perez-Martinez FC, Guerra J, Posadas I, Cena V (2011) Barriers to non-viral vector-mediated gene delivery in the nervous system. Pharm Res 28(8):1843-1858. doi: 10.1007/s11095-010-0364-7
    • (2011) Pharm Res , vol.28 , Issue.8 , pp. 1843-1858
    • Perez-Martinez, F.C.1    Guerra, J.2    Posadas, I.3    Cena, V.4
  • 16
    • 79956028473 scopus 로고    scopus 로고
    • RNAi medicine for the brain: Progresses and challenges
    • doi: 10.1093/hmg/ddr137
    • Boudreau RL, Rodriguez-Lebron E, Davidson BL (2011) RNAi medicine for the brain: progresses and challenges. Hum Mol Genet 20 (R1):R21-R27. doi: 10.1093/hmg/ddr137
    • (2011) Hum Mol Genet , vol.20 , Issue.1
    • Boudreau, R.L.1    Rodriguez-Lebron, E.2    Davidson, B.L.3
  • 17
    • 82955233704 scopus 로고    scopus 로고
    • Rational design of therapeutic siRNAs: Minimizing off-targeting potential to improve the safety of RNAi therapy for Huntington's disease
    • 10.1038/mt.2011.185 1:CAS:528:DC%2BC3MXht1anu7%2FI 21952166
    • Boudreau RL, Spengler RM, Davidson BL (2011) Rational design of therapeutic siRNAs: minimizing off-targeting potential to improve the safety of RNAi therapy for Huntington's disease. Mol Ther 19(12):2169-2177. doi: 10.1038/mt.2011.185
    • (2011) Mol Ther , vol.19 , Issue.12 , pp. 2169-2177
    • Boudreau, R.L.1    Spengler, R.M.2    Davidson, B.L.3
  • 19
    • 0036787671 scopus 로고    scopus 로고
    • SiRNA-mediated gene silencing in vitro and in vivo
    • 1:CAS:528:DC%2BD38XnsVGkt74%3D 12244328
    • Xia H, Mao Q, Paulson HL, Davidson BL (2002) siRNA-mediated gene silencing in vitro and in vivo. Nat Biotechnol 20(10):1006-1010
    • (2002) Nat Biotechnol , vol.20 , Issue.10 , pp. 1006-1010
    • Xia, H.1    Mao, Q.2    Paulson, H.L.3    Davidson, B.L.4
  • 21
    • 34447535345 scopus 로고    scopus 로고
    • Engineering targeted viral vectors for gene therapy
    • DOI 10.1038/nrg2141, PII NRG2141
    • Waehler R, Russell SJ, Curiel DT (2007) Engineering targeted viral vectors for gene therapy. Nat Rev Genet 8(8):573-587 (Pubitemid 47077278)
    • (2007) Nature Reviews Genetics , vol.8 , Issue.8 , pp. 573-587
    • Waehler, R.1    Russell, S.J.2    Curiel, D.T.3
  • 22
    • 4544344036 scopus 로고    scopus 로고
    • Immune responses against adenoviral vectors and their transgene products: A review of strategies for evasion
    • DOI 10.1016/S1040-8428(03)00172-0, PII S1040842803001720
    • Schagen FH, Ossevoort M, Toes RE, Hoeben RC (2004) Immune responses against adenoviral vectors and their transgene products: a review of strategies for evasion. Crit Rev Oncol Hematol 50(1):51-70. doi: 10.1016/S1040-8428(03) 00172-0 (Pubitemid 38501617)
    • (2004) Critical Reviews in Oncology/Hematology , vol.50 , Issue.1 , pp. 51-70
    • Schagen, F.H.E.1    Ossevoort, M.2    Toes, R.E.M.3    Hoeben, R.C.4
  • 23
    • 83255177161 scopus 로고    scopus 로고
    • Using non-coding small RNAs to develop therapies for Huntington's disease
    • 10.1038/gt.2011.170 1:CAS:528:DC%2BC3MXhsF2jt7nM 22158031
    • Zhang Y, Friedlander RM (2011) Using non-coding small RNAs to develop therapies for Huntington's disease. Gene Ther 18(12):1139-1149. doi: 10.1038/gt.2011.170
    • (2011) Gene Ther , vol.18 , Issue.12 , pp. 1139-1149
    • Zhang, Y.1    Friedlander, R.M.2
  • 24
    • 33745299723 scopus 로고    scopus 로고
    • Fatality in mice due to oversaturation of cellular microRNA/short hairpin RNA pathways
    • DOI 10.1038/nature04791, PII NATURE04791
    • Grimm D, Streetz KL, Jopling CL, Storm TA, Pandey K, Davis CR, Marion P, Salazar F, Kay MA (2006) Fatality in mice due to oversaturation of cellular microRNA/short hairpin RNA pathways. Nature 441(7092):537-541. doi: 10.1038/nature04791 (Pubitemid 44050158)
    • (2006) Nature , vol.441 , Issue.7092 , pp. 537-541
    • Grimm, D.1    Streetz, K.L.2    Jopling, C.L.3    Storm, T.A.4    Pandey, K.5    Davis, C.R.6    Marion, P.7    Salazar, F.8    Kay, M.A.9
  • 26
    • 44949222522 scopus 로고    scopus 로고
    • Results from a phase i safety trial of hAADC gene therapy for Parkinson disease
    • 10.1212/01.wnl.0000312381.29287.ff 1:CAS:528:DC%2BD1cXlvVCmtro%3D 18401019
    • Eberling JL, Jagust WJ, Christine CW, Starr P, Larson P, Bankiewicz KS, Aminoff MJ (2008) Results from a phase I safety trial of hAADC gene therapy for Parkinson disease. Neurology 70(21):1980-1983. doi: 10.1212/01.wnl.0000312381. 29287.ff
    • (2008) Neurology , vol.70 , Issue.21 , pp. 1980-1983
    • Eberling, J.L.1    Jagust, W.J.2    Christine, C.W.3    Starr, P.4    Larson, P.5    Bankiewicz, K.S.6    Aminoff, M.J.7
  • 27
    • 77955175906 scopus 로고    scopus 로고
    • Eight years of clinical improvement in MPTP-lesioned primates after gene therapy with AAV2-hAADC
    • 10.1038/mt.2010.106 1:CAS:528:DC%2BC3cXntFSmtrk%3D 20531394
    • Hadaczek P, Eberling JL, Pivirotto P, Bringas J, Forsayeth J, Bankiewicz KS (2010) Eight years of clinical improvement in MPTP-lesioned primates after gene therapy with AAV2-hAADC. Mol Ther 18(8):1458-1461. doi: 10.1038/mt.2010.106
    • (2010) Mol Ther , vol.18 , Issue.8 , pp. 1458-1461
    • Hadaczek, P.1    Eberling, J.L.2    Pivirotto, P.3    Bringas, J.4    Forsayeth, J.5    Bankiewicz, K.S.6
  • 31
    • 34447114618 scopus 로고    scopus 로고
    • Transvascular delivery of small interfering RNA to the central nervous system
    • DOI 10.1038/nature05901, PII NATURE05901
    • Kumar P, Wu H, McBride JL, Jung KE, Kim MH, Davidson BL, Lee SK, Shankar P, Manjunath N (2007) Transvascular delivery of small interfering RNA to the central nervous system. Nature 448(7149):39-43. doi: 10.1038/nature05901 (Pubitemid 47037001)
    • (2007) Nature , vol.448 , Issue.7149 , pp. 39-43
    • Kumar, P.1    Wu, H.2    McBride, J.L.3    Jung, K.-E.4    Hee Kim, M.5    Davidson, B.L.6    Kyung Lee, S.7    Shankar, P.8    Manjunath, N.9
  • 35
    • 65249105506 scopus 로고    scopus 로고
    • Nanotechnology approach for drug addiction therapy: Gene silencing using delivery of gold nanorod-siRNA nanoplex in dopaminergic neurons
    • 1:CAS:528:DC%2BD1MXkvFGhtLg%3D 19307583
    • Bonoiu AC, Mahajan SD, Ding H, Roy I, Yong KT, Kumar R, Hu R, Bergey EJ, Schwartz SA, Prasad PN (2009) Nanotechnology approach for drug addiction therapy: gene silencing using delivery of gold nanorod-siRNA nanoplex in dopaminergic neurons. Proc Natl Acad Sci USA 106(14):5546-5550
    • (2009) Proc Natl Acad Sci USA , vol.106 , Issue.14 , pp. 5546-5550
    • Bonoiu, A.C.1    Mahajan, S.D.2    Ding, H.3    Roy, I.4    Yong, K.T.5    Kumar, R.6    Hu, R.7    Bergey, E.J.8    Schwartz, S.A.9    Prasad, P.N.10
  • 36
    • 78149346495 scopus 로고    scopus 로고
    • Nonviral vectors for the delivery of small interfering RNAs to the CNS
    • 10.2217/nnm.10.105 1:CAS:528:DC%2BC3cXhtlOhtr%2FF
    • Posadas I, Guerra FJ, Cena V (2010) Nonviral vectors for the delivery of small interfering RNAs to the CNS. Nanomedicine (Lond) 5(8):1219-1236. doi: 10.2217/nnm.10.105
    • (2010) Nanomedicine (Lond) , vol.5 , Issue.8 , pp. 1219-1236
    • Posadas, I.1    Guerra, F.J.2    Cena, V.3
  • 37
    • 84861224359 scopus 로고    scopus 로고
    • Delivery of cationic polymer-siRNA nanoparticles for gene therapies in neural regeneration
    • 10.1016/j.bbrc.2012.03.155 1:CAS:528:DC%2BC38Xmslygsr8%3D 22542938
    • Liang Y, Liu Z, Shuai X, Wang W, Liu J, Bi W, Wang C, Jing X, Liu Y, Tao E (2012) Delivery of cationic polymer-siRNA nanoparticles for gene therapies in neural regeneration. Biochem Biophys Res Commun 421(4):690-695. doi: 10.1016/j.bbrc.2012.03.155
    • (2012) Biochem Biophys Res Commun , vol.421 , Issue.4 , pp. 690-695
    • Liang, Y.1    Liu, Z.2    Shuai, X.3    Wang, W.4    Liu, J.5    Bi, W.6    Wang, C.7    Jing, X.8    Liu, Y.9    Tao, E.10
  • 38
    • 77957820261 scopus 로고    scopus 로고
    • RNAi and small interfering RNAs in human disease therapeutic applications
    • 10.1016/j.tibtech.2010.07.009 1:CAS:528:DC%2BC3cXht12qurvJ 2955826 20833440
    • Lares MR, Rossi JJ, Ouellet DL (2010) RNAi and small interfering RNAs in human disease therapeutic applications. Trends Biotechnol 28(11):570-579. doi: 10.1016/j.tibtech.2010.07.009
    • (2010) Trends Biotechnol , vol.28 , Issue.11 , pp. 570-579
    • Lares, M.R.1    Rossi, J.J.2    Ouellet, D.L.3
  • 39
    • 84860129964 scopus 로고    scopus 로고
    • Dendrimer-based postnatal therapy for neuroinflammation and cerebral palsy in a rabbit model
    • doi: 10.1126/scitranslmed.3003162
    • Kannan S, Dai H, Navath RS, Balakrishnan B, Jyoti A, Janisse J, Romero R, Kannan RM (2012) Dendrimer-based postnatal therapy for neuroinflammation and cerebral palsy in a rabbit model. Sci Transl Med 4 (130):130ra146. doi: 10.1126/scitranslmed.3003162
    • (2012) Sci Transl Med , vol.4 , Issue.130
    • Kannan, S.1    Dai, H.2    Navath, R.S.3    Balakrishnan, B.4    Jyoti, A.5    Janisse, J.6    Romero, R.7    Kannan, R.M.8
  • 40
    • 67649887150 scopus 로고    scopus 로고
    • Brain-targeting gene delivery and cellular internalization mechanisms for modified rabies virus glycoprotein RVG29 nanoparticles
    • 10.1016/j.biomaterials.2009.02.051 1:CAS:528:DC%2BD1MXotlOkt7c%3D 19467700
    • Liu Y, Huang R, Han L, Ke W, Shao K, Ye L, Lou J, Jiang C (2009) Brain-targeting gene delivery and cellular internalization mechanisms for modified rabies virus glycoprotein RVG29 nanoparticles. Biomaterials 30(25):4195-4202. doi: 10.1016/j.biomaterials.2009.02.051
    • (2009) Biomaterials , vol.30 , Issue.25 , pp. 4195-4202
    • Liu, Y.1    Huang, R.2    Han, L.3    Ke, W.4    Shao, K.5    Ye, L.6    Lou, J.7    Jiang, C.8
  • 41
    • 39749086433 scopus 로고    scopus 로고
    • Polymeric nanoparticles for the drug delivery to the central nervous system
    • DOI 10.1517/17425247.5.2.155
    • Tosi G, Costantino L, Ruozi B, Forni F, Vandelli MA (2008) Polymeric nanoparticles for the drug delivery to the central nervous system. Expert Opin Drug Deliv 5(2):155-174. doi: 10.1517/17425247.5.2.155 (Pubitemid 351308309)
    • (2008) Expert Opinion on Drug Delivery , vol.5 , Issue.2 , pp. 155-174
    • Tosi, G.1    Costantino, L.2    Ruozi, B.3    Forni, F.4    Vandelli, M.A.5
  • 42
    • 0032976670 scopus 로고    scopus 로고
    • Hydrolytic degradation characteristics of aliphatic polyesters derived from lactic and glycolic acids
    • DOI 10.1002/(SICI)1097-4636(1999) 48:3<342::AID-JBM20>3.0.CO;2-7
    • Li S (1999) Hydrolytic degradation characteristics of aliphatic polyesters derived from lactic and glycolic acids. J Biomed Mater Res 48(3):342-353. doi: 10.1002/(SICI)1097-4636(1999)48:3<342:AID-JBM20>3.0. CO;2-7 (Pubitemid 29242707)
    • (1999) Journal of Biomedical Materials Research , vol.48 , Issue.3 , pp. 342-353
    • Li, S.1
  • 43
    • 0027070540 scopus 로고
    • 14C]-poly(lactic acid) nanoparticles coated with albumin after parenteral administration to rats
    • DOI 10.1016/0142-9612(92)90142-B
    • Bazile DV, Ropert C, Huve P, Verrecchia T, Marlard M, Frydman A, Veillard M, Spenlehauer G (1992) Body distribution of fully biodegradable [14C]-poly(lactic acid) nanoparticles coated with albumin after parenteral administration to rats. Biomaterials 13(15):1093-1102 (Pubitemid 23022908)
    • (1992) Biomaterials , vol.13 , Issue.15 , pp. 1093-1102
    • Bazile, D.V.1    Ropert, C.2    Huve, P.3    Verrecchia, T.4    Marlard, M.5    Frydman, A.6    Veillard, M.7    Spenlehauer, G.8
  • 44
    • 77955175216 scopus 로고    scopus 로고
    • Strategies in the design of nanoparticles for therapeutic applications
    • 10.1038/nrd2591 1:CAS:528:DC%2BC3cXosFeltbk%3D 20616808
    • Petros RA, DeSimone JM (2010) Strategies in the design of nanoparticles for therapeutic applications. Nat Rev Drug Discov 9(8):615-627. doi: 10.1038/nrd2591
    • (2010) Nat Rev Drug Discov , vol.9 , Issue.8 , pp. 615-627
    • Petros, R.A.1    Desimone, J.M.2
  • 45
    • 79959971282 scopus 로고    scopus 로고
    • Nanoparticle PEGylation for imaging and therapy
    • 10.2217/nnm.11.19 1:CAS:528:DC%2BC3MXpsVajtro%3D
    • Jokerst JV, Lobovkina T, Zare RN, Gambhir SS (2011) Nanoparticle PEGylation for imaging and therapy. Nanomedicine (Lond) 6(4):715-728. doi: 10.2217/nnm.11.19
    • (2011) Nanomedicine (Lond) , vol.6 , Issue.4 , pp. 715-728
    • Jokerst, J.V.1    Lobovkina, T.2    Zare, R.N.3    Gambhir, S.S.4
  • 46
    • 33847099637 scopus 로고    scopus 로고
    • Lipid-based nanoparticles for nucleic acid delivery
    • DOI 10.1007/s11095-006-9180-5
    • Li W, Szoka FC Jr (2007) Lipid-based nanoparticles for nucleic acid delivery. Pharm Res 24(3):438-449. doi: 10.1007/s11095-006-9180-5 (Pubitemid 46295166)
    • (2007) Pharmaceutical Research , vol.24 , Issue.3 , pp. 438-449
    • Li, W.1    Szoka Jr., F.C.2
  • 47
    • 71949099757 scopus 로고    scopus 로고
    • Lipid-based nanotherapeutics for siRNA delivery
    • 10.1111/j.1365-2796.2009.02189.x 1:CAS:528:DC%2BC3cXhtFOmtro%3D 20059641
    • Schroeder A, Levins CG, Cortez C, Langer R, Anderson DG (2010) Lipid-based nanotherapeutics for siRNA delivery. J Intern Med 267(1):9-21. doi: 10.1111/j.1365-2796.2009.02189.x
    • (2010) J Intern Med , vol.267 , Issue.1 , pp. 9-21
    • Schroeder, A.1    Levins, C.G.2    Cortez, C.3    Langer, R.4    Anderson, D.G.5
  • 48
    • 72949119713 scopus 로고    scopus 로고
    • Lipidic systems for in vivo siRNA delivery
    • 10.1208/s12248-009-9140-1 1:CAS:528:DC%2BD1MXhsFGmur%2FF 2782074 19757082
    • Wu SY, McMillan NA (2009) Lipidic systems for in vivo siRNA delivery. AAPS J 11(4):639-652. doi: 10.1208/s12248-009-9140-1
    • (2009) AAPS J , vol.11 , Issue.4 , pp. 639-652
    • Wu, S.Y.1    McMillan, N.A.2
  • 49
    • 34248679586 scopus 로고    scopus 로고
    • SHRNA and siRNA delivery to the brain
    • DOI 10.1016/j.addr.2007.03.008, PII S0169409X07000221, Opportunities and Challenges for Therapeutic Gene Silencing using RNAi and microRNA Technologies
    • Pardridge WM (2007) shRNA and siRNA delivery to the brain. Adv Drug Deliv Rev 59(2-3):141-152. doi: 10.1016/j.addr.2007.03.008 (Pubitemid 46773051)
    • (2007) Advanced Drug Delivery Reviews , vol.59 , Issue.2-3 , pp. 141-152
    • Pardridge, W.M.1
  • 50
    • 79953858598 scopus 로고    scopus 로고
    • Delivery of siRNA to the mouse brain by systemic injection of targeted exosomes
    • 10.1038/nbt.1807 1:CAS:528:DC%2BC3MXjsVKqsLw%3D 21423189
    • Alvarez-Erviti L, Seow Y, Yin H, Betts C, Lakhal S, Wood MJ (2011) Delivery of siRNA to the mouse brain by systemic injection of targeted exosomes. Nat Biotechnol 29(4):341-345. doi: 10.1038/nbt.1807
    • (2011) Nat Biotechnol , vol.29 , Issue.4 , pp. 341-345
    • Alvarez-Erviti, L.1    Seow, Y.2    Yin, H.3    Betts, C.4    Lakhal, S.5    Wood, M.J.6
  • 51
    • 79953899274 scopus 로고    scopus 로고
    • SiRNA delivery with exosome nanoparticles
    • 10.1038/nbt.1830 21478846
    • van den Boorn JG, Schlee M, Coch C, Hartmann G (2011) SiRNA delivery with exosome nanoparticles. Nat Biotechnol 29(4):325-326. doi: 10.1038/nbt.1830
    • (2011) Nat Biotechnol , vol.29 , Issue.4 , pp. 325-326
    • Van Den Boorn, J.G.1    Schlee, M.2    Coch, C.3    Hartmann, G.4
  • 52
    • 84855706467 scopus 로고    scopus 로고
    • A rapid, targeted, neuron-selective, in vivo knockdown following a single intracerebroventricular injection of a novel chemically modified siRNA in the adult rat brain
    • 10.1016/j.jbiotec.2011.10.003 1:CAS:528:DC%2BC38XpsFahug%3D%3D 22079868
    • Nakajima H, Kubo T, Semi Y, Itakura M, Kuwamura M, Izawa T, Azuma YT, Takeuchi T (2012) A rapid, targeted, neuron-selective, in vivo knockdown following a single intracerebroventricular injection of a novel chemically modified siRNA in the adult rat brain. J Biotechnol 157(2):326-333. doi: 10.1016/j.jbiotec.2011.10.003
    • (2012) J Biotechnol , vol.157 , Issue.2 , pp. 326-333
    • Nakajima, H.1    Kubo, T.2    Semi, Y.3    Itakura, M.4    Kuwamura, M.5    Izawa, T.6    Azuma, Y.T.7    Takeuchi, T.8
  • 53
    • 76949090691 scopus 로고    scopus 로고
    • Nonviral transfection of mouse calvarial organ in vitro using Accell-modified siRNA
    • 10.1097/PRS.0b013e3181c82df1 1:CAS:528:DC%2BC3cXitlajsrs%3D 19910849
    • Gupta AK, Eshraghi Y, Gliniak C, Gosain AK (2010) Nonviral transfection of mouse calvarial organ in vitro using Accell-modified siRNA. Plast Reconstr Surg 125(2):494-501. doi: 10.1097/PRS.0b013e3181c82df1
    • (2010) Plast Reconstr Surg , vol.125 , Issue.2 , pp. 494-501
    • Gupta, A.K.1    Eshraghi, Y.2    Gliniak, C.3    Gosain, A.K.4
  • 54
    • 80054709331 scopus 로고    scopus 로고
    • Nonviral transfection of leukemic primary cells and cells lines by siRNA-a direct comparison between Nucleofection and Accell delivery
    • 10.1016/j.exphem.2011.08.003 1:CAS:528:DC%2BC3MXhtlChtrjM 21856272
    • Larsen HO, Roug AS, Nielsen K, Sondergaard CS, Hokland P (2011) Nonviral transfection of leukemic primary cells and cells lines by siRNA-a direct comparison between Nucleofection and Accell delivery. Exp Hematol 39(11):1081-1089. doi: 10.1016/j.exphem.2011.08.003
    • (2011) Exp Hematol , vol.39 , Issue.11 , pp. 1081-1089
    • Larsen, H.O.1    Roug, A.S.2    Nielsen, K.3    Sondergaard, C.S.4    Hokland, P.5
  • 55
    • 57649134261 scopus 로고    scopus 로고
    • Intranasal delivery bypasses the blood-brain barrier to target therapeutic agents to the central nervous system and treat neurodegenerative disease
    • 10.1186/1471-2202-9-S3-S5 2604883 19091002
    • Hanson LR, Frey WH 2nd (2008) Intranasal delivery bypasses the blood-brain barrier to target therapeutic agents to the central nervous system and treat neurodegenerative disease. BMC Neurosci 9(Suppl 3):S5. doi: 10.1186/1471-2202-9-S3-S5
    • (2008) BMC Neurosci , vol.9 , Issue.SUPPL.. 3 , pp. 5
    • Hanson, L.R.1    Frey II, W.H.2
  • 56
    • 80054823427 scopus 로고    scopus 로고
    • Olfactory targeting through intranasal delivery of biopharmaceutical drugs to the brain: Current development
    • 21712015
    • Wen MM (2011) Olfactory targeting through intranasal delivery of biopharmaceutical drugs to the brain: current development. Discov Med 11(61):497-503
    • (2011) Discov Med , vol.11 , Issue.61 , pp. 497-503
    • Wen, M.M.1
  • 57
    • 60349115456 scopus 로고    scopus 로고
    • Delivery of NGF to the brain: Intranasal versus ocular administration in anti-NGF transgenic mice
    • 10.3233/JAD-2009-0953 1:CAS:528:DC%2BD1MXhvVCku7Y%3D 19221427
    • Capsoni S, Covaceuszach S, Ugolini G, Spirito F, Vignone D, Stefanini B, Amato G, Cattaneo A (2009) Delivery of NGF to the brain: intranasal versus ocular administration in anti-NGF transgenic mice. J Alzheimers Dis 16(2):371-388. doi: 10.3233/JAD-2009-0953
    • (2009) J Alzheimers Dis , vol.16 , Issue.2 , pp. 371-388
    • Capsoni, S.1    Covaceuszach, S.2    Ugolini, G.3    Spirito, F.4    Vignone, D.5    Stefanini, B.6    Amato, G.7    Cattaneo, A.8
  • 58
    • 84861614275 scopus 로고    scopus 로고
    • Intranasal "painless" human nerve growth factors slows amyloid neurodegeneration and prevents memory deficits in App X PS1 mice
    • 10.1371/journal.pone.0037555 1:CAS:528:DC%2BC38XosVels7s%3D 3364340 22666365
    • Capsoni S, Marinelli S, Ceci M, Vignone D, Amato G, Malerba F, Paoletti F, Meli G, Viegi A, Pavone F, Cattaneo A (2012) Intranasal "painless" human nerve growth factors slows amyloid neurodegeneration and prevents memory deficits in App X PS1 mice. PLoS ONE 7(5):e37555. doi: 10.1371/journal.pone. 0037555
    • (2012) PLoS ONE , vol.7 , Issue.5 , pp. 37555
    • Capsoni, S.1    Marinelli, S.2    Ceci, M.3    Vignone, D.4    Amato, G.5    Malerba, F.6    Paoletti, F.7    Meli, G.8    Viegi, A.9    Pavone, F.10    Cattaneo, A.11
  • 59
    • 84857424742 scopus 로고    scopus 로고
    • Mature and precursor brain-derived neurotrophic factor have individual roles in the mouse olfactory bulb
    • 10.1371/journal.pone.0031978 1:CAS:528:DC%2BC38XjsVaqsLo%3D 3283713 22363780
    • Mast TG, Fadool DA (2012) Mature and precursor brain-derived neurotrophic factor have individual roles in the mouse olfactory bulb. PLoS ONE 7(2):e31978. doi: 10.1371/journal.pone.0031978
    • (2012) PLoS ONE , vol.7 , Issue.2 , pp. 31978
    • Mast, T.G.1    Fadool, D.A.2
  • 60
    • 84860854802 scopus 로고    scopus 로고
    • Delivery of brain-derived neurotrophic factor via nose-to-brain pathway
    • 10.1007/s11095-011-0572-9 1:CAS:528:DC%2BC3MXhtV2gtr3N 21879386
    • Vaka SR, Murthy SN, Balaji A, Repka MA (2012) Delivery of brain-derived neurotrophic factor via nose-to-brain pathway. Pharm Res 29(2):441-447. doi: 10.1007/s11095-011-0572-9
    • (2012) Pharm Res , vol.29 , Issue.2 , pp. 441-447
    • Vaka, S.R.1    Murthy, S.N.2    Balaji, A.3    Repka, M.A.4
  • 61
    • 34147183745 scopus 로고    scopus 로고
    • RNAi silencing in mouse models of neurodegenerative diseases
    • DOI 10.2174/156720107780362276
    • Farah MH (2007) RNAi silencing in mouse models of neurodegenerative diseases. Curr Drug Deliv 4(2):161-167 (Pubitemid 46557492)
    • (2007) Current Drug Delivery , vol.4 , Issue.2 , pp. 161-167
    • Farah, M.H.1
  • 63
    • 84859000580 scopus 로고    scopus 로고
    • Intranasal delivery of siRNA to the olfactory bulbs of mice via the olfactory nerve pathway
    • 10.1016/j.neulet.2012.02.037 1:CAS:528:DC%2BC38Xjs1Gqurw%3D 22387067
    • Renner DB, Frey WH 2nd, Hanson LR (2012) Intranasal delivery of siRNA to the olfactory bulbs of mice via the olfactory nerve pathway. Neurosci Lett 513(2):193-197. doi: 10.1016/j.neulet.2012.02.037
    • (2012) Neurosci Lett , vol.513 , Issue.2 , pp. 193-197
    • Renner, D.B.1    Frey II, W.H.2    Hanson, L.R.3
  • 64
    • 84862315161 scopus 로고    scopus 로고
    • Increased brain radioactivity by intranasal P-labeled siRNA dendriplexes within in situ-forming mucoadhesive gels
    • 10.2147/IJN.S28261 1:CAS:528:DC%2BC38XkslamsLk%3D
    • Perez AP, Mundina-Weilenmann C, Romero EL, Morilla MJ (2012) Increased brain radioactivity by intranasal P-labeled siRNA dendriplexes within in situ-forming mucoadhesive gels. Int J Nanomed 7:1373-1385. doi: 10.2147/IJN.S28261
    • (2012) Int J Nanomed , vol.7 , pp. 1373-1385
    • Perez, A.P.1    Mundina-Weilenmann, C.2    Romero, E.L.3    Morilla, M.J.4
  • 65
    • 77349107167 scopus 로고    scopus 로고
    • Neuroprotection by biodegradable PAMAM ester (e-PAM-R)-mediated HMGB1 siRNA delivery in primary cortical cultures and in the postischemic brain
    • 10.1016/j.jconrel.2009.11.011 1:CAS:528:DC%2BC3cXisV2rtrc%3D 19944723
    • Kim ID, Lim CM, Kim JB, Nam HY, Nam K, Kim SW, Park JS, Lee JK (2010) Neuroprotection by biodegradable PAMAM ester (e-PAM-R)-mediated HMGB1 siRNA delivery in primary cortical cultures and in the postischemic brain. J Control Release 142(3):422-430. doi: 10.1016/j.jconrel.2009.11.011
    • (2010) J Control Release , vol.142 , Issue.3 , pp. 422-430
    • Kim, I.D.1    Lim, C.M.2    Kim, J.B.3    Nam, H.Y.4    Nam, K.5    Kim, S.W.6    Park, J.S.7    Lee, J.K.8
  • 66
    • 38449104482 scopus 로고    scopus 로고
    • The therapeutic potential of siRNA in gene therapy of neurodegenerative disorders
    • 1:CAS:528:DC%2BD2sXhsVKns7jP 17982877
    • Koutsilieri E, Rethwilm A, Scheller C (2007) The therapeutic potential of siRNA in gene therapy of neurodegenerative disorders. J Neural Transm Suppl 72:43-49
    • (2007) J Neural Transm Suppl , vol.72 , pp. 43-49
    • Koutsilieri, E.1    Rethwilm, A.2    Scheller, C.3
  • 67
    • 77953949978 scopus 로고    scopus 로고
    • Gene therapy in Alzheimer's disease - Potential for disease modification
    • 10.1111/j.1582-4934.2010.01038.x 1:CAS:528:DC%2BC3cXnvVagtr8%3D 20158567
    • Nilsson P, Iwata N, Muramatsu S, Tjernberg LO, Winblad B, Saido TC (2010) Gene therapy in Alzheimer's disease - potential for disease modification. J Cell Mol Med 14(4):741-757. doi: 10.1111/j.1582-4934.2010.01038.x
    • (2010) J Cell Mol Med , vol.14 , Issue.4 , pp. 741-757
    • Nilsson, P.1    Iwata, N.2    Muramatsu, S.3    Tjernberg, L.O.4    Winblad, B.5    Saido, T.C.6
  • 68
    • 28944450147 scopus 로고    scopus 로고
    • Therapeutic potential of small interfering RNA for central nervous system diseases
    • DOI 10.1001/archneur.62.12.1810
    • Lovett-Racke AE, Cravens PD, Gocke AR, Racke MK, Stuve O (2005) Therapeutic potential of small interfering RNA for central nervous system diseases. Arch Neurol 62(12):1810-1813. doi: 10.1001/archneur.62.12.1810 (Pubitemid 41785073)
    • (2005) Archives of Neurology , vol.62 , Issue.12 , pp. 1810-1813
    • Lovett-Racke, A.E.1    Cravens, P.D.2    Gocke, A.R.3    Racke, M.K.4    Stuve, O.5
  • 69
    • 33645841354 scopus 로고    scopus 로고
    • Silencing neurodegenerative disease: Bringing RNA interference to the clinic
    • 10.1586/14737175.6.2.223 1:CAS:528:DC%2BD28XislGisbo%3D 16466302
    • Rodriguez-Lebron E, Gonzalez-Alegre P (2006) Silencing neurodegenerative disease: bringing RNA interference to the clinic. Expert Rev Neurother 6(2):223-233. doi: 10.1586/14737175.6.2.223
    • (2006) Expert Rev Neurother , vol.6 , Issue.2 , pp. 223-233
    • Rodriguez-Lebron, E.1    Gonzalez-Alegre, P.2
  • 71
    • 70649097387 scopus 로고    scopus 로고
    • RNAi applications in therapy development for neurodegenerative disease
    • 1:CAS:528:DC%2BD1MXhsFGqtLjE 19751205
    • Maxwell MM (2009) RNAi applications in therapy development for neurodegenerative disease. Curr Pharm Des 15(34):3977-3991
    • (2009) Curr Pharm des , vol.15 , Issue.34 , pp. 3977-3991
    • Maxwell, M.M.1
  • 72
    • 84856882305 scopus 로고    scopus 로고
    • Regenerating islet-derived 1alpha (Reg-1alpha) protein is new neuronal secreted factor that stimulates neurite outgrowth via exostosin tumor-like 3 (EXTL3) receptor
    • 10.1074/jbc.M111.260349 22158612
    • Acquatella-Tran Van Ba I, Marchal S, Francois F, Silhol M, Lleres C, Michel B, Benyamin Y, Verdier JM, Trousse F, Marcilhac A (2012) Regenerating islet-derived 1alpha (Reg-1alpha) protein is new neuronal secreted factor that stimulates neurite outgrowth via exostosin tumor-like 3 (EXTL3) receptor. J Biol Chem 287(7):4726-4739. doi: 10.1074/jbc.M111.260349
    • (2012) J Biol Chem , vol.287 , Issue.7 , pp. 4726-4739
    • Acquatella-Tran Van Ba, I.1    Marchal, S.2    Francois, F.3    Silhol, M.4    Lleres, C.5    Michel, B.6    Benyamin, Y.7    Verdier, J.M.8    Trousse, F.9    Marcilhac, A.10
  • 74
    • 83555172246 scopus 로고    scopus 로고
    • Endoplasmic reticulum stress-induced CHOP activation mediates the down-regulation of leptin in human neuroblastoma SH-SY5Y cells treated with the oxysterol 27-hydroxycholesterol
    • 10.1016/j.cellsig.2011.09.029 1:CAS:528:DC%2BC3MXhs1Slt7fN 3237961 21983012
    • Marwarha G, Dasari B, Ghribi O (2012) Endoplasmic reticulum stress-induced CHOP activation mediates the down-regulation of leptin in human neuroblastoma SH-SY5Y cells treated with the oxysterol 27-hydroxycholesterol. Cell Signal 24(2):484-492. doi: 10.1016/j.cellsig.2011.09.029
    • (2012) Cell Signal , vol.24 , Issue.2 , pp. 484-492
    • Marwarha, G.1    Dasari, B.2    Ghribi, O.3
  • 75
    • 3242785569 scopus 로고    scopus 로고
    • Targeting BACE with small inhibitory nucleic acids - A future for Alzheimer's disease therapy?
    • Nawrot B (2004) Targeting BACE with small inhibitory nucleic acids - a future for Alzheimer's disease therapy? Acta Biochim Pol 51(2):431-444. doi: 035001431 (Pubitemid 38969305)
    • (2004) Acta Biochimica Polonica , vol.51 , Issue.2 , pp. 431-444
    • Nawrot, B.1
  • 76
    • 33751206536 scopus 로고    scopus 로고
    • Genetic and pharmacological basis for therapeutic inhibition of β-and γ-secretases in mouse models of Alzheimer's memory deficits
    • Ohno M (2006) Genetic and pharmacological basis for therapeutic inhibition of beta- and gamma-secretases in mouse models of Alzheimer's memory deficits. Rev Neurosci 17(4):429-454 (Pubitemid 44787022)
    • (2006) Reviews in the Neurosciences , vol.17 , Issue.4 , pp. 429-454
    • Ohno, M.1
  • 77
    • 77953922271 scopus 로고    scopus 로고
    • Lentivirus-expressed siRNA vectors against Alzheimer disease
    • 10.1007/978-1-60761-533-0-15 1:CAS:528:DC%2BC3cXnvFWks74%3D 20225047
    • Peng KA, Masliah E (2010) Lentivirus-expressed siRNA vectors against Alzheimer disease. Methods Mol Biol 614:215-224. doi: 10.1007/978-1-60761-533-0- 15
    • (2010) Methods Mol Biol , vol.614 , pp. 215-224
    • Peng, K.A.1    Masliah, E.2
  • 78
    • 80054680024 scopus 로고    scopus 로고
    • Silencing GADD153/CHOP gene expression protects against Alzheimer's disease-like pathology induced by 27-hydroxycholesterol in rabbit hippocampus
    • 10.1371/journal.pone.0026420 1:CAS:528:DC%2BC3MXhsVWqs7vI 3194795 22046282
    • Prasanthi JR, Larson T, Schommer J, Ghribi O (2011) Silencing GADD153/CHOP gene expression protects against Alzheimer's disease-like pathology induced by 27-hydroxycholesterol in rabbit hippocampus. PLoS ONE 6(10):e26420. doi: 10.1371/journal.pone.0026420
    • (2011) PLoS ONE , vol.6 , Issue.10 , pp. 26420
    • Prasanthi, J.R.1    Larson, T.2    Schommer, J.3    Ghribi, O.4
  • 79
    • 79953897022 scopus 로고    scopus 로고
    • Gene therapy in mouse models of Huntington disease
    • 10.1177/1073858410386236 1:CAS:528:DC%2BC3MXltVGqsbs%3D 3131092 21489966
    • Southwell AL, Patterson PH (2011) Gene therapy in mouse models of Huntington disease. Neuroscientist 17(2):153-162. doi: 10.1177/1073858410386236
    • (2011) Neuroscientist , vol.17 , Issue.2 , pp. 153-162
    • Southwell, A.L.1    Patterson, P.H.2
  • 81
    • 84891883697 scopus 로고    scopus 로고
    • Protein interactions and target discovery in Huntington's disease
    • D.C. Lo R.E. Hughes (eds) Frontiers in Neuroscience Boca Raton (FL)
    • Miller JP, Hughes RE (2011) Protein interactions and target discovery in Huntington's disease. In: Lo DC, Hughes RE (eds) Neurobiology of Huntington's disease: applications to drug discovery. Frontiers in Neuroscience, Boca Raton (FL)
    • (2011) Neurobiology of Huntington's Disease: Applications to Drug Discovery
    • Miller, J.P.1    Hughes, R.E.2
  • 82
    • 72149084425 scopus 로고    scopus 로고
    • Huntington disease: Pathogenesis, biomarkers, and approaches to experimental therapeutics
    • 10.1016/S1353-8020(09)70800-4 20082975
    • Ross CA, Shoulson I (2009) Huntington disease: pathogenesis, biomarkers, and approaches to experimental therapeutics. Parkinsonism Relat Disord 15(Suppl 3):S135-S138. doi: 10.1016/S1353-8020(09)70800-4
    • (2009) Parkinsonism Relat Disord , vol.15 , Issue.SUPPL. 3
    • Ross, C.A.1    Shoulson, I.2
  • 84
    • 78650855101 scopus 로고    scopus 로고
    • NMDA receptor-dependent regulation of dendritic spine morphology by SAP102 splice variants
    • 10.1523/JNEUROSCI.1034-10.2011 3030119 21209193
    • Chen BS, Thomas EV, Sanz-Clemente A, Roche KW (2011) NMDA receptor-dependent regulation of dendritic spine morphology by SAP102 splice variants. J Neurosci 31(1):89-96. doi: 10.1523/JNEUROSCI.1034-10.2011
    • (2011) J Neurosci , vol.31 , Issue.1 , pp. 89-96
    • Chen, B.S.1    Thomas, E.V.2    Sanz-Clemente, A.3    Roche, K.W.4
  • 85
    • 80054013637 scopus 로고    scopus 로고
    • Spatial and temporal requirements for huntingtin (Htt) in neuronal migration and survival during brain development
    • 10.1523/JNEUROSCI.2774-11.2011 1:CAS:528:DC%2BC3MXhtlCgtrvI 3407803 21994396
    • Tong Y, Ha TJ, Liu L, Nishimoto A, Reiner A, Goldowitz D (2011) Spatial and temporal requirements for huntingtin (Htt) in neuronal migration and survival during brain development. J Neurosci 31(41):14794-14799. doi: 10.1523/JNEUROSCI.2774-11.2011
    • (2011) J Neurosci , vol.31 , Issue.41 , pp. 14794-14799
    • Tong, Y.1    Ha, T.J.2    Liu, L.3    Nishimoto, A.4    Reiner, A.5    Goldowitz, D.6
  • 86
    • 67349263503 scopus 로고    scopus 로고
    • A majority of Huntington's disease patients may be treatable by individualized allele-specific RNA interference
    • 10.1016/j.expneurol.2009.03.004 1:CAS:528:DC%2BD1MXlvF2ktrg%3D 19289118
    • Lombardi MS, Jaspers L, Spronkmans C, Gellera C, Taroni F, Di Maria E, Donato SD, Kaemmerer WF (2009) A majority of Huntington's disease patients may be treatable by individualized allele-specific RNA interference. Exp Neurol 217(2):312-319. doi: 10.1016/j.expneurol.2009.03.004
    • (2009) Exp Neurol , vol.217 , Issue.2 , pp. 312-319
    • Lombardi, M.S.1    Jaspers, L.2    Spronkmans, C.3    Gellera, C.4    Taroni, F.5    Di Maria, E.6    Donato, S.D.7    Kaemmerer, W.F.8
  • 87
    • 67349159137 scopus 로고    scopus 로고
    • Five siRNAs targeting three SNPs may provide therapy for three-quarters of Huntington's disease patients
    • 10.1016/j.cub.2009.03.030 1:CAS:528:DC%2BD1MXlslWmt7w%3D 2746439 19361997
    • Pfister EL, Kennington L, Straubhaar J, Wagh S, Liu W, DiFiglia M, Landwehrmeyer B, Vonsattel JP, Zamore PD, Aronin N (2009) Five siRNAs targeting three SNPs may provide therapy for three-quarters of Huntington's disease patients. Curr Biol 19(9):774-778. doi: 10.1016/j.cub.2009.03.030
    • (2009) Curr Biol , vol.19 , Issue.9 , pp. 774-778
    • Pfister, E.L.1    Kennington, L.2    Straubhaar, J.3    Wagh, S.4    Liu, W.5    Difiglia, M.6    Landwehrmeyer, B.7    Vonsattel, J.P.8    Zamore, P.D.9    Aronin, N.10
  • 88
    • 78649359013 scopus 로고    scopus 로고
    • Allele-selective inhibition of huntingtin expression by switching to an miRNA-like RNAi mechanism
    • 10.1016/j.chembiol.2010.10.013 1:CAS:528:DC%2BC3cXhsVGntrfN 3021381 21095568
    • Hu J, Liu J, Corey DR (2010) Allele-selective inhibition of huntingtin expression by switching to an miRNA-like RNAi mechanism. Chem Biol 17(11):1183-1188. doi: 10.1016/j.chembiol.2010.10.013
    • (2010) Chem Biol , vol.17 , Issue.11 , pp. 1183-1188
    • Hu, J.1    Liu, J.2    Corey, D.R.3
  • 90
    • 82955199935 scopus 로고    scopus 로고
    • Preclinical safety of RNAi-mediated HTT suppression in the rhesus macaque as a potential therapy for Huntington's disease
    • 10.1038/mt.2011.219 1:CAS:528:DC%2BC3MXhtlOnur3I 22031240
    • McBride JL, Pitzer MR, Boudreau RL, Dufour B, Hobbs T, Ojeda SR, Davidson BL (2011) Preclinical safety of RNAi-mediated HTT suppression in the rhesus macaque as a potential therapy for Huntington's disease. Mol Ther 19(12):2152-2162. doi: 10.1038/mt.2011.219
    • (2011) Mol Ther , vol.19 , Issue.12 , pp. 2152-2162
    • McBride, J.L.1    Pitzer, M.R.2    Boudreau, R.L.3    Dufour, B.4    Hobbs, T.5    Ojeda, S.R.6    Davidson, B.L.7
  • 92
    • 33645159024 scopus 로고    scopus 로고
    • RNA knockdown as a potential therapeutic strategy in Parkinson's disease
    • 10.1038/sj.gt.3302669 1:CAS:528:DC%2BD28XitVGnurc%3D 16267570
    • Manfredsson FP, Lewin AS, Mandel RJ (2006) RNA knockdown as a potential therapeutic strategy in Parkinson's disease. Gene Ther 13(6):517-524. doi: 10.1038/sj.gt.3302669
    • (2006) Gene Ther , vol.13 , Issue.6 , pp. 517-524
    • Manfredsson, F.P.1    Lewin, A.S.2    Mandel, R.J.3
  • 93
    • 60649116702 scopus 로고    scopus 로고
    • Applications of lentiviral vectors for biology and gene therapy of neurological disorders
    • 1:CAS:528:DC%2BD1MXhtVSkt7w%3D 19075629
    • Lundberg C, Bjorklund T, Carlsson T, Jakobsson J, Hantraye P, Deglon N, Kirik D (2008) Applications of lentiviral vectors for biology and gene therapy of neurological disorders. Curr Gene Ther 8(6):461-473
    • (2008) Curr Gene Ther , vol.8 , Issue.6 , pp. 461-473
    • Lundberg, C.1    Bjorklund, T.2    Carlsson, T.3    Jakobsson, J.4    Hantraye, P.5    Deglon, N.6    Kirik, D.7
  • 94
    • 37549021547 scopus 로고    scopus 로고
    • Preclinical development of gene therapy for Parkinson's disease
    • 10.1016/j.expneurol.2007.08.003 1:CAS:528:DC%2BD1cXivFeguw%3D%3D 17904121
    • Porras G, Bezard E (2008) Preclinical development of gene therapy for Parkinson's disease. Exp Neurol 209(1):72-81. doi: 10.1016/j.expneurol.2007.08. 003
    • (2008) Exp Neurol , vol.209 , Issue.1 , pp. 72-81
    • Porras, G.1    Bezard, E.2
  • 95
    • 12744251557 scopus 로고    scopus 로고
    • The aggravating role of the ubiquitin-proteasome system in neurodegeneration
    • 10.1016/j.febslet.2004.12.058 1:CAS:528:DC%2BD2MXmsVGmsg%3D%3D 15670810
    • Ardley HC, Hung CC, Robinson PA (2005) The aggravating role of the ubiquitin-proteasome system in neurodegeneration. FEBS Lett 579(3):571-576. doi: 10.1016/j.febslet.2004.12.058
    • (2005) FEBS Lett , vol.579 , Issue.3 , pp. 571-576
    • Ardley, H.C.1    Hung, C.C.2    Robinson, P.A.3
  • 98
    • 81755185877 scopus 로고    scopus 로고
    • Alpha-Synuclein fate is determined by USP9X-regulated monoubiquitination
    • 10.1073/pnas.1105725108 1:CAS:528:DC%2BC3MXhs1WjtL%2FO 22065755
    • Rott R, Szargel R, Haskin J, Bandopadhyay R, Lees AJ, Shani V, Engelender S (2011) alpha-Synuclein fate is determined by USP9X-regulated monoubiquitination. Proc Natl Acad Sci USA 108(46):18666-18671. doi: 10.1073/pnas.1105725108
    • (2011) Proc Natl Acad Sci USA , vol.108 , Issue.46 , pp. 18666-18671
    • Rott, R.1    Szargel, R.2    Haskin, J.3    Bandopadhyay, R.4    Lees, A.J.5    Shani, V.6    Engelender, S.7
  • 100
    • 34249006580 scopus 로고    scopus 로고
    • Protein kinase Cδ negatively regulates tyrosine hydroxylase activity and dopamine synthesis by enhancing protein phosphatase-2A activity in dopaminergic neurons
    • DOI 10.1523/JNEUROSCI.4107-06.2007
    • Zhang D, Kanthasamy A, Yang Y, Anantharam V, Kanthasamy A (2007) Protein kinase C delta negatively regulates tyrosine hydroxylase activity and dopamine synthesis by enhancing protein phosphatase-2A activity in dopaminergic neurons. J Neurosci 27(20):5349-5362. doi: 10.1523/JNEUROSCI.4107-06.2007 (Pubitemid 46790158)
    • (2007) Journal of Neuroscience , vol.27 , Issue.20 , pp. 5349-5362
    • Zhang, D.1    Kanthasamy, A.2    Yang, Y.3    Anantharam, V.4    Kanthasamy, A.5
  • 101
    • 80855132749 scopus 로고    scopus 로고
    • Dopaminergic neurotoxicant 6-OHDA induces oxidative damage through proteolytic activation of PKCdelta in cell culture and animal models of Parkinson's disease
    • 10.1016/j.taap.2011.07.021 1:CAS:528:DC%2BC3MXhtl2ntbrK 3205342 21846476
    • Latchoumycandane C, Anantharam V, Jin H, Kanthasamy A, Kanthasamy A (2011) Dopaminergic neurotoxicant 6-OHDA induces oxidative damage through proteolytic activation of PKCdelta in cell culture and animal models of Parkinson's disease. Toxicol Appl Pharmacol 256(3):314-323. doi: 10.1016/j.taap.2011.07.021
    • (2011) Toxicol Appl Pharmacol , vol.256 , Issue.3 , pp. 314-323
    • Latchoumycandane, C.1    Anantharam, V.2    Jin, H.3    Kanthasamy, A.4    Kanthasamy, A.5
  • 103
    • 71849084134 scopus 로고    scopus 로고
    • Mitochondrial dysfunction in Parkinson's disease
    • 10.1016/j.bbadis.2009.08.013 1:CAS:528:DC%2BD1MXhsFanurnE 19733240
    • Winklhofer KF, Haass C (2010) Mitochondrial dysfunction in Parkinson's disease. Biochim Biophys Acta 1802(1):29-44. doi: 10.1016/j.bbadis.2009.08.013
    • (2010) Biochim Biophys Acta , vol.1802 , Issue.1 , pp. 29-44
    • Winklhofer, K.F.1    Haass, C.2
  • 104
    • 79955129749 scopus 로고    scopus 로고
    • Knockdown of GAD67 protein levels normalizes neuronal activity in a rat model of Parkinson's disease
    • 1:CAS:528:DC%2BC3MXmslWksbk%3D 21449035
    • Horvath L, van Marion I, Taï K, Nielsen TT, Lundberg C (2011) Knockdown of GAD67 protein levels normalizes neuronal activity in a rat model of Parkinson's disease. J Gene Med 13(3):188-197
    • (2011) J Gene Med , vol.13 , Issue.3 , pp. 188-197
    • Horvath, L.1    Van Marion, I.2    Taï, K.3    Nielsen, T.T.4    Lundberg, C.5
  • 105
    • 78649483815 scopus 로고    scopus 로고
    • Nature and nurture in neuropsychiatric genetics: Where do we stand?
    • 3181950 20373663
    • Dick DM, Riley B, Kendler KS (2010) Nature and nurture in neuropsychiatric genetics: where do we stand? Dialogues Clin Neurosci 12(1):7-23
    • (2010) Dialogues Clin Neurosci , vol.12 , Issue.1 , pp. 7-23
    • Dick, D.M.1    Riley, B.2    Kendler, K.S.3
  • 106
    • 84860179774 scopus 로고    scopus 로고
    • Is dopamine neurotransmission altered in prodromal schizophrenia? A review of the evidence
    • 1:CAS:528:DC%2BC38XkslWjs78%3D 22280434
    • Bauer M, Praschak-Rieder N, Kasper S, Willeit M (2012) Is dopamine neurotransmission altered in prodromal schizophrenia? A review of the evidence. Curr Pharm Des 18(12):1568-1579
    • (2012) Curr Pharm des , vol.18 , Issue.12 , pp. 1568-1579
    • Bauer, M.1    Praschak-Rieder, N.2    Kasper, S.3    Willeit, M.4
  • 107
    • 84870057403 scopus 로고    scopus 로고
    • Pharmacological treatment of schizophrenia: A critical review of the pharmacology and clinical effects of current and future therapeutic agents
    • 10.1038/mp.2012.47 22584864
    • Miyamoto S, Miyake N, Jarskog LF, Fleischhacker WW, Lieberman JA (2012) Pharmacological treatment of schizophrenia: a critical review of the pharmacology and clinical effects of current and future therapeutic agents. Mol Psychiatry. doi: 10.1038/mp.2012.47
    • (2012) Mol Psychiatry
    • Miyamoto, S.1    Miyake, N.2    Jarskog, L.F.3    Fleischhacker, W.W.4    Lieberman, J.A.5
  • 108
    • 80052019393 scopus 로고    scopus 로고
    • The effects of siRNA-mediated RGS4 gene silencing on the whole genome transcription profile: Implications for schizophrenia
    • 1:CAS:528:DC%2BC3MXht1SrurnN 21712773
    • Vrajova M, Pekova S, Horacek J, Hoschl C (2011) The effects of siRNA-mediated RGS4 gene silencing on the whole genome transcription profile: implications for schizophrenia. Neuro Endocrinol Lett 32(3):246-252
    • (2011) Neuro Endocrinol Lett , vol.32 , Issue.3 , pp. 246-252
    • Vrajova, M.1    Pekova, S.2    Horacek, J.3    Hoschl, C.4
  • 109
    • 33646517484 scopus 로고    scopus 로고
    • Insulin receptor deficits in schizophrenia and in cellular and animal models of insulin receptor dysfunction
    • 10.1016/j.schres.2006.02.009 16581231
    • Zhao Z, Ksiezak-Reding H, Riggio S, Haroutunian V, Pasinetti GM (2006) Insulin receptor deficits in schizophrenia and in cellular and animal models of insulin receptor dysfunction. Schizophr Res 84(1):1-14. doi: 10.1016/j.schres.2006.02.009
    • (2006) Schizophr Res , vol.84 , Issue.1 , pp. 1-14
    • Zhao, Z.1    Ksiezak-Reding, H.2    Riggio, S.3    Haroutunian, V.4    Pasinetti, G.M.5
  • 111
    • 85027925126 scopus 로고    scopus 로고
    • Dysbindin-1, a schizophrenia-related protein, facilitates neurite outgrowth by promoting the transcriptional activity of p53
    • 10.1038/mp.2011.43 1:CAS:528:DC%2BC3MXhtlKjt7zO 21502952
    • Ma X, Fei E, Fu C, Ren H, Wang G (2011) Dysbindin-1, a schizophrenia-related protein, facilitates neurite outgrowth by promoting the transcriptional activity of p53. Mol Psychiatry 16(11):1105-1116. doi: 10.1038/mp.2011.43
    • (2011) Mol Psychiatry , vol.16 , Issue.11 , pp. 1105-1116
    • Ma, X.1    Fei, E.2    Fu, C.3    Ren, H.4    Wang, G.5
  • 112
    • 84860834078 scopus 로고    scopus 로고
    • Behavioral effects of non-viral mediated RNA interference of synapsin II in the medial prefrontal cortex of the rat
    • 10.1016/j.schres.2012.01.029 22341900
    • Dyck BA, Tan ML, Daya RP, Basu D, Sookram CD, Thomas N, Mishra RK (2012) Behavioral effects of non-viral mediated RNA interference of synapsin II in the medial prefrontal cortex of the rat. Schizophr Res 137(1-3):32-38. doi: 10.1016/j.schres.2012.01.029
    • (2012) Schizophr Res , vol.137 , Issue.1-3 , pp. 32-38
    • Dyck, B.A.1    Tan, M.L.2    Daya, R.P.3    Basu, D.4    Sookram, C.D.5    Thomas, N.6    Mishra, R.K.7
  • 114
    • 42249102078 scopus 로고    scopus 로고
    • Transgenics, toxicity and therapeutics in rodent models of mutant SOD1-mediated familial ALS
    • 10.1016/j.pneurobio.2008.01.001 1:CAS:528:DC%2BD1cXls1yksbY%3D 18282652
    • Turner BJ, Talbot K (2008) Transgenics, toxicity and therapeutics in rodent models of mutant SOD1-mediated familial ALS. Prog Neurobiol 85(1):94-134. doi: 10.1016/j.pneurobio.2008.01.001
    • (2008) Prog Neurobiol , vol.85 , Issue.1 , pp. 94-134
    • Turner, B.J.1    Talbot, K.2
  • 115
    • 0242379146 scopus 로고    scopus 로고
    • Selective silencing by RNAi of a dominant allele that causes amyotrophic lateral sclerosis
    • 1:CAS:528:DC%2BD3sXmt1CrsbY%3D 12934714
    • Ding H, Schwarz DS, Keene A, el Affar B, Fenton L, Xia X, Shi Y, Zamore PD, Xu Z (2003) Selective silencing by RNAi of a dominant allele that causes amyotrophic lateral sclerosis. Aging Cell 2(4):209-217
    • (2003) Aging Cell , vol.2 , Issue.4 , pp. 209-217
    • Ding, H.1    Schwarz, D.S.2    Keene, A.3    El Affar, B.4    Fenton, L.5    Xia, X.6    Shi, Y.7    Zamore, P.D.8    Xu, Z.9
  • 116
    • 78651237824 scopus 로고    scopus 로고
    • Design of functional small interfering RNAs targeting amyotrophic lateral sclerosis-associated mutant alleles
    • 1:CAS:528:DC%2BC3MXisFakt7g%3D
    • Geng CM, Ding HL (2011) Design of functional small interfering RNAs targeting amyotrophic lateral sclerosis-associated mutant alleles. Chin Med J (Engl) 124(1):106-110
    • (2011) Chin Med J (Engl) , vol.124 , Issue.1 , pp. 106-110
    • Geng, C.M.1    Ding, H.L.2
  • 118
    • 78649473387 scopus 로고    scopus 로고
    • An examination of wild-type SOD1 in modulating the toxicity and aggregation of ALS-associated mutant SOD1
    • 10.1093/hmg/ddq408 1:CAS:528:DC%2BC3cXhsVyhsr3M 20871097
    • Prudencio M, Durazo A, Whitelegge JP, Borchelt DR (2010) An examination of wild-type SOD1 in modulating the toxicity and aggregation of ALS-associated mutant SOD1. Hum Mol Genet 19(24):4774-4789. doi: 10.1093/hmg/ddq408
    • (2010) Hum Mol Genet , vol.19 , Issue.24 , pp. 4774-4789
    • Prudencio, M.1    Durazo, A.2    Whitelegge, J.P.3    Borchelt, D.R.4
  • 119
    • 84984774109 scopus 로고    scopus 로고
    • Motor neuron disease: Misfolded wild-type SOD1 may link sporadic and familial ALS
    • 10.1038/nrneurol.2010.169 21188749
    • Yates D (2010) Motor neuron disease: misfolded wild-type SOD1 may link sporadic and familial ALS. Nat Rev Neurol 6(12):645. doi: 10.1038/nrneurol.2010. 169
    • (2010) Nat Rev Neurol , vol.6 , Issue.12 , pp. 645
    • Yates, D.1
  • 120
    • 33747201641 scopus 로고    scopus 로고
    • Allele-specific RNAi selectively silences mutant SOD1 and achieves significant therapeutic benefit in vivo
    • DOI 10.1016/j.nbd.2006.04.019, PII S0969996106001021
    • Xia X, Zhou H, Huang Y, Xu Z (2006) Allele-specific RNAi selectively silences mutant SOD1 and achieves significant therapeutic benefit in vivo. Neurobiol Dis 23(3):578-586. doi: 10.1016/j.nbd.2006.04.019 (Pubitemid 44234094)
    • (2006) Neurobiology of Disease , vol.23 , Issue.3 , pp. 578-586
    • Xia, X.1    Zhou, H.2    Huang, Y.3    Xu, Z.4
  • 122
    • 17644383664 scopus 로고    scopus 로고
    • Lentiviral-mediated silencing of SOD1 through RNA interference retards disease onset and progression in a mouse model of ALS
    • DOI 10.1038/nm1207
    • Raoul C, Abbas-Terki T, Bensadoun JC, Guillot S, Haase G, Szulc J, Henderson CE, Aebischer P (2005) Lentiviral-mediated silencing of SOD1 through RNA interference retards disease onset and progression in a mouse model of ALS. Nat Med 11(4):423-428. doi: 10.1038/nm1207 (Pubitemid 40562333)
    • (2005) Nature Medicine , vol.11 , Issue.4 , pp. 423-428
    • Raoul, C.1    Abbas-Terki, T.2    Bensadoun, J.-C.3    Guillot, S.4    Haase, G.5    Szulc, J.6    Henderson, C.E.7    Aebischer, P.8
  • 123
    • 79551641133 scopus 로고    scopus 로고
    • Neuroprotection by gene therapy targeting mutant SOD1 in individual pools of motor neurons does not translate into therapeutic benefit in fALS mice
    • 10.1038/mt.2010.260 1:CAS:528:DC%2BC3cXhsVGnu7zE 21102563
    • Towne C, Setola V, Schneider BL, Aebischer P (2011) Neuroprotection by gene therapy targeting mutant SOD1 in individual pools of motor neurons does not translate into therapeutic benefit in fALS mice. Mol Ther 19(2):274-283. doi: 10.1038/mt.2010.260
    • (2011) Mol Ther , vol.19 , Issue.2 , pp. 274-283
    • Towne, C.1    Setola, V.2    Schneider, B.L.3    Aebischer, P.4
  • 124
    • 61349134134 scopus 로고    scopus 로고
    • Retrogradely transported siRNA silences human mutant SOD1 in spinal cord motor neurons
    • 10.1007/s00221-009-1742-4 1:CAS:528:DC%2BD1MXlt1Sgsb8%3D 19259649
    • Rizvanov AA, Mukhamedyarov MA, Palotas A, Islamov RR (2009) Retrogradely transported siRNA silences human mutant SOD1 in spinal cord motor neurons. Exp Brain Res 195(1):1-4. doi: 10.1007/s00221-009-1742-4
    • (2009) Exp Brain Res , vol.195 , Issue.1 , pp. 1-4
    • Rizvanov, A.A.1    Mukhamedyarov, M.A.2    Palotas, A.3    Islamov, R.R.4
  • 125
    • 84856007240 scopus 로고    scopus 로고
    • Cdk5/p25-induced cytosolic PLA2-mediated lysophosphatidylcholine production regulates neuroinflammation and triggers neurodegeneration
    • 10.1523/JNEUROSCI.5177-11.2012 1:CAS:528:DC%2BC38XhsVOgtrk%3D 22262900
    • Sundaram JR, Chan ES, Poore CP, Pareek TK, Cheong WF, Shui G, Tang N, Low CM, Wenk MR, Kesavapany S (2012) Cdk5/p25-induced cytosolic PLA2-mediated lysophosphatidylcholine production regulates neuroinflammation and triggers neurodegeneration. J Neurosci 32(3):1020-1034. doi: 10.1523/JNEUROSCI.5177-11. 2012
    • (2012) J Neurosci , vol.32 , Issue.3 , pp. 1020-1034
    • Sundaram, J.R.1    Chan, E.S.2    Poore, C.P.3    Pareek, T.K.4    Cheong, W.F.5    Shui, G.6    Tang, N.7    Low, C.M.8    Wenk, M.R.9    Kesavapany, S.10
  • 126
    • 81255143014 scopus 로고    scopus 로고
    • Epigenetic regulation of motor neuron cell death through DNA methylation
    • 10.1523/JNEUROSCI.1639-11.2011 1:CAS:528:DC%2BC3MXhsFaksLzO 3238138 22090490
    • Chestnut BA, Chang Q, Price A, Lesuisse C, Wong M, Martin LJ (2011) Epigenetic regulation of motor neuron cell death through DNA methylation. J Neurosci 31(46):16619-16636. doi: 10.1523/JNEUROSCI.1639-11.2011
    • (2011) J Neurosci , vol.31 , Issue.46 , pp. 16619-16636
    • Chestnut, B.A.1    Chang, Q.2    Price, A.3    Lesuisse, C.4    Wong, M.5    Martin, L.J.6
  • 127
    • 78651368514 scopus 로고    scopus 로고
    • New therapeutic targets for amyotrophic lateral sclerosis
    • 10.1517/14728222.2011.542152 1:CAS:528:DC%2BC3MXktVehtw%3D%3D 21133819
    • Kuzma-Kozakiewicz M, Kwiecinski H (2011) New therapeutic targets for amyotrophic lateral sclerosis. Expert Opin Ther Targets 15(2):127-143. doi: 10.1517/14728222.2011.542152
    • (2011) Expert Opin Ther Targets , vol.15 , Issue.2 , pp. 127-143
    • Kuzma-Kozakiewicz, M.1    Kwiecinski, H.2
  • 128
    • 78650515010 scopus 로고    scopus 로고
    • Lentivirus-mediated transfer of MMP-9 shRNA provides neuroprotection following focal ischemic brain injury in rats
    • 10.1016/j.brainres.2010.10.002 1:CAS:528:DC%2BC3cXhsFyltbjM 20950592
    • Hu Q, Chen C, Khatibi NH, Li L, Yang L, Wang K, Han J, Duan W, Zhang JH, Zhou C (2011) Lentivirus-mediated transfer of MMP-9 shRNA provides neuroprotection following focal ischemic brain injury in rats. Brain Res 1367:347-359. doi: 10.1016/j.brainres.2010.10.002
    • (2011) Brain Res , vol.1367 , pp. 347-359
    • Hu, Q.1    Chen, C.2    Khatibi, N.H.3    Li, L.4    Yang, L.5    Wang, K.6    Han, J.7    Duan, W.8    Zhang, J.H.9    Zhou, C.10
  • 129
    • 59749100986 scopus 로고    scopus 로고
    • Therapeutic application of gene silencing MMP-9 in a middle cerebral artery occlusion-induced focal ischemia rat model
    • 10.1016/j.expneurol.2008.11.007 1:CAS:528:DC%2BD1MXhvFGntL8%3D 19073180
    • Hu Q, Chen C, Yan J, Yang X, Shi X, Zhao J, Lei J, Yang L, Wang K, Chen L, Huang H, Han J, Zhang JH, Zhou C (2009) Therapeutic application of gene silencing MMP-9 in a middle cerebral artery occlusion-induced focal ischemia rat model. Exp Neurol 216(1):35-46. doi: 10.1016/j.expneurol.2008.11.007
    • (2009) Exp Neurol , vol.216 , Issue.1 , pp. 35-46
    • Hu, Q.1    Chen, C.2    Yan, J.3    Yang, X.4    Shi, X.5    Zhao, J.6    Lei, J.7    Yang, L.8    Wang, K.9    Chen, L.10    Huang, H.11    Han, J.12    Zhang, J.H.13    Zhou, C.14
  • 130
    • 84863230646 scopus 로고    scopus 로고
    • Matrix metalloproteinase-2-mediated occludin degradation and caveolin-1-mediated claudin-5 redistribution contribute to blood-brain barrier damage in early ischemic stroke stage
    • 10.1523/JNEUROSCI.6409-11.2012 1:CAS:528:DC%2BC38XjslKjtLY%3D 3339570 22378877
    • Liu J, Jin X, Liu KJ, Liu W (2012) Matrix metalloproteinase-2-mediated occludin degradation and caveolin-1-mediated claudin-5 redistribution contribute to blood-brain barrier damage in early ischemic stroke stage. J Neurosci 32(9):3044-3057. doi: 10.1523/JNEUROSCI.6409-11.2012
    • (2012) J Neurosci , vol.32 , Issue.9 , pp. 3044-3057
    • Liu, J.1    Jin, X.2    Liu, K.J.3    Liu, W.4
  • 131
    • 60149094194 scopus 로고    scopus 로고
    • The Notch pathway mediates expansion of a progenitor pool and neuronal differentiation in adult neural progenitor cells after stroke
    • 10.1016/j.neuroscience.2008.10.064 1:CAS:528:DC%2BD1MXit1OjtLc%3D 2757073 19059466
    • Wang L, Chopp M, Zhang RL, Zhang L, Letourneau Y, Feng YF, Jiang A, Morris DC, Zhang ZG (2009) The Notch pathway mediates expansion of a progenitor pool and neuronal differentiation in adult neural progenitor cells after stroke. Neuroscience 158(4):1356-1363. doi: 10.1016/j.neuroscience.2008.10.064
    • (2009) Neuroscience , vol.158 , Issue.4 , pp. 1356-1363
    • Wang, L.1    Chopp, M.2    Zhang, R.L.3    Zhang, L.4    Letourneau, Y.5    Feng, Y.F.6    Jiang, A.7    Morris, D.C.8    Zhang, Z.G.9
  • 132
    • 79957873952 scopus 로고    scopus 로고
    • SDF-1alpha secreted by human CD133-derived multipotent stromal cells promotes neural progenitor cell survival through CXCR7
    • 10.1089/scd.2010.0198 1:CAS:528:DC%2BC3MXmslaju7s%3D 20854168
    • Bakondi B, Shimada IS, Peterson BM, Spees JL (2011) SDF-1alpha secreted by human CD133-derived multipotent stromal cells promotes neural progenitor cell survival through CXCR7. Stem Cells Dev 20(6):1021-1029. doi: 10.1089/scd.2010.0198
    • (2011) Stem Cells Dev , vol.20 , Issue.6 , pp. 1021-1029
    • Bakondi, B.1    Shimada, I.S.2    Peterson, B.M.3    Spees, J.L.4
  • 133
    • 84857873046 scopus 로고    scopus 로고
    • Minocycline-preconditioned neural stem cells enhance neuroprotection after ischemic stroke in rats
    • 10.1523/JNEUROSCI.5686-11.2012 1:CAS:528:DC%2BC38XjvFyntbs%3D 3315362 22399769
    • Sakata H, Niizuma K, Yoshioka H, Kim GS, Jung JE, Katsu M, Narasimhan P, Maier CM, Nishiyama Y, Chan PH (2012) Minocycline-preconditioned neural stem cells enhance neuroprotection after ischemic stroke in rats. J Neurosci 32(10):3462-3473. doi: 10.1523/JNEUROSCI.5686-11.2012
    • (2012) J Neurosci , vol.32 , Issue.10 , pp. 3462-3473
    • Sakata, H.1    Niizuma, K.2    Yoshioka, H.3    Kim, G.S.4    Jung, J.E.5    Katsu, M.6    Narasimhan, P.7    Maier, C.M.8    Nishiyama, Y.9    Chan, P.H.10
  • 135
    • 78650903873 scopus 로고    scopus 로고
    • The NCX3 isoform of the Na+/Ca2+ exchanger contributes to neuroprotection elicited by ischemic postconditioning
    • 10.1038/jcbfm.2010.100 1:CAS:528:DC%2BC3MXhtlCisw%3D%3D 20628398
    • Pignataro G, Esposito E, Cuomo O, Sirabella R, Boscia F, Guida N, Di Renzo G, Annunziato L (2011) The NCX3 isoform of the Na+/Ca2+ exchanger contributes to neuroprotection elicited by ischemic postconditioning. J Cereb Blood Flow Metab 31(1):362-370. doi: 10.1038/jcbfm.2010.100
    • (2011) J Cereb Blood Flow Metab , vol.31 , Issue.1 , pp. 362-370
    • Pignataro, G.1    Esposito, E.2    Cuomo, O.3    Sirabella, R.4    Boscia, F.5    Guida, N.6    Di Renzo, G.7    Annunziato, L.8
  • 137
    • 80455174695 scopus 로고    scopus 로고
    • Multipotent mesenchymal stromal cells increase tPA expression and concomitantly decrease PAI-1 expression in astrocytes through the sonic hedgehog signaling pathway after stroke (in vitro study)
    • 10.1038/jcbfm.2011.116 1:CAS:528:DC%2BC3MXhsVags73O 21829213
    • Xin H, Li Y, Shen LH, Liu X, Hozeska-Solgot A, Zhang RL, Zhang ZG, Chopp M (2011) Multipotent mesenchymal stromal cells increase tPA expression and concomitantly decrease PAI-1 expression in astrocytes through the sonic hedgehog signaling pathway after stroke (in vitro study). J Cereb Blood Flow Metab 31(11):2181-2188. doi: 10.1038/jcbfm.2011.116
    • (2011) J Cereb Blood Flow Metab , vol.31 , Issue.11 , pp. 2181-2188
    • Xin, H.1    Li, Y.2    Shen, L.H.3    Liu, X.4    Hozeska-Solgot, A.5    Zhang, R.L.6    Zhang, Z.G.7    Chopp, M.8
  • 138
    • 77952579169 scopus 로고    scopus 로고
    • SiRNA delivery to CNS cells using a membrane translocation peptide
    • 10.1021/bc900488e 1:CAS:528:DC%2BC3cXltFChtrs%3D 2874326 20420373
    • Ifediba MA, Medarova Z, Ng SW, Yang J, Moore A (2010) siRNA delivery to CNS cells using a membrane translocation peptide. Bioconjug Chem 21(5):803-806. doi: 10.1021/bc900488e
    • (2010) Bioconjug Chem , vol.21 , Issue.5 , pp. 803-806
    • Ifediba, M.A.1    Medarova, Z.2    Ng, S.W.3    Yang, J.4    Moore, A.5
  • 139
    • 85028103629 scopus 로고    scopus 로고
    • Vascular adhesion protein-1 inhibition provides antiinflammatory protection after an intracerebral hemorrhagic stroke in mice
    • 10.1038/jcbfm.2010.167 1:CAS:528:DC%2BC3MXis1CgtL0%3D 20877383
    • Ma Q, Manaenko A, Khatibi NH, Chen W, Zhang JH, Tang J (2011) Vascular adhesion protein-1 inhibition provides antiinflammatory protection after an intracerebral hemorrhagic stroke in mice. J Cereb Blood Flow Metab 31(3):881-893. doi: 10.1038/jcbfm.2010.167
    • (2011) J Cereb Blood Flow Metab , vol.31 , Issue.3 , pp. 881-893
    • Ma, Q.1    Manaenko, A.2    Khatibi, N.H.3    Chen, W.4    Zhang, J.H.5    Tang, J.6
  • 140
    • 80052023031 scopus 로고    scopus 로고
    • Apoptosis signal-regulating kinase 1 (Ask1) targeted small interfering RNA on ischemic neuronal cell death
    • 10.1016/j.brainres.2011.07.018 1:CAS:528:DC%2BC3MXhtVGltb3K 21803338
    • Kim HW, Cho KJ, Lee SK, Kim GW (2011) Apoptosis signal-regulating kinase 1 (Ask1) targeted small interfering RNA on ischemic neuronal cell death. Brain Res 1412:73-78. doi: 10.1016/j.brainres.2011.07.018
    • (2011) Brain Res , vol.1412 , pp. 73-78
    • Kim, H.W.1    Cho, K.J.2    Lee, S.K.3    Kim, G.W.4
  • 141
    • 8544283103 scopus 로고    scopus 로고
    • CHOP is involved in endoplasmic reticulum stress-induced apoptosis by enhancing DR5 expression in human carcinoma cells
    • DOI 10.1074/jbc.M406933200
    • Yamaguchi H, Wang HG (2004) CHOP is involved in endoplasmic reticulum stress-induced apoptosis by enhancing DR5 expression in human carcinoma cells. J Biol Chem 279(44):45495-45502. doi: 10.1074/jbc.M406933200 (Pubitemid 39491535)
    • (2004) Journal of Biological Chemistry , vol.279 , Issue.44 , pp. 45495-45502
    • Yamaguchi, H.1    Wang, H.-G.2
  • 142
    • 77954377779 scopus 로고    scopus 로고
    • The endoplasmic reticulum stress response factor CHOP-10 protects against hypoxia-induced neuronal death
    • 10.1074/jbc.M109.095299 1:CAS:528:DC%2BC3cXos1Wrsbs%3D 20448044
    • Halterman MW, Gill M, DeJesus C, Ogihara M, Schor NF, Federoff HJ (2010) The endoplasmic reticulum stress response factor CHOP-10 protects against hypoxia-induced neuronal death. J Biol Chem 285(28):21329-21340. doi: 10.1074/jbc.M109.095299
    • (2010) J Biol Chem , vol.285 , Issue.28 , pp. 21329-21340
    • Halterman, M.W.1    Gill, M.2    Dejesus, C.3    Ogihara, M.4    Schor, N.F.5    Federoff, H.J.6
  • 143
    • 84856433369 scopus 로고    scopus 로고
    • CHOP silencing reduces acute brain injury in the rat model of subarachnoid hemorrhage
    • 10.1161/STROKEAHA.111.626432 3265655 22180248
    • He Z, Ostrowski RP, Sun X, Ma Q, Huang B, Zhan Y, Zhang JH (2012) CHOP silencing reduces acute brain injury in the rat model of subarachnoid hemorrhage. Stroke 43(2):484-490. doi: 10.1161/STROKEAHA.111.626432
    • (2012) Stroke , vol.43 , Issue.2 , pp. 484-490
    • He, Z.1    Ostrowski, R.P.2    Sun, X.3    Ma, Q.4    Huang, B.5    Zhan, Y.6    Zhang, J.H.7
  • 144
    • 4744345481 scopus 로고    scopus 로고
    • Neurovascular protection reduces early brain injury after subarachnoid hemorrhage
    • DOI 10.1161/01.STR.0000141162.29864.e9
    • Park S, Yamaguchi M, Zhou C, Calvert JW, Tang J, Zhang JH (2004) Neurovascular protection reduces early brain injury after subarachnoid hemorrhage. Stroke 35(10):2412-2417. doi: 10.1161/01.STR.0000141162.29864.e9 (Pubitemid 39311273)
    • (2004) Stroke , vol.35 , Issue.10 , pp. 2412-2417
    • Park, S.1    Yamaguchi, M.2    Zhou, C.3    Calvert, J.W.4    Tang, J.5    Zhang, J.H.6
  • 145
    • 59849110117 scopus 로고    scopus 로고
    • Pluronic-coated carbon nanotubes do not induce degeneration of cortical neurons in vivo and in vitro
    • 10.1016/j.nano.2008.06.008 1:CAS:528:DC%2BC3cXmtVCgtg%3D%3D 18693142
    • Bardi G, Tognini P, Ciofani G, Raffa V, Costa M, Pizzorusso T (2009) Pluronic-coated carbon nanotubes do not induce degeneration of cortical neurons in vivo and in vitro. Nanomedicine 5(1):96-104. doi: 10.1016/j.nano.2008.06.008
    • (2009) Nanomedicine , vol.5 , Issue.1 , pp. 96-104
    • Bardi, G.1    Tognini, P.2    Ciofani, G.3    Raffa, V.4    Costa, M.5    Pizzorusso, T.6
  • 146
    • 84155164308 scopus 로고    scopus 로고
    • Dendrimer-mediated siRNA delivery knocks down Beclin 1 and potentiates NMDA-mediated toxicity in rat cortical neurons
    • 10.1111/j.1471-4159.2011.07556.x 1:CAS:528:DC%2BC38XhsFygu7s%3D 22035151
    • Perez-Carrion MD, Perez-Martinez FC, Merino S, Sanchez-Verdu P, Martinez-Hernandez J, Lujan R, Cena V (2012) Dendrimer-mediated siRNA delivery knocks down Beclin 1 and potentiates NMDA-mediated toxicity in rat cortical neurons. J Neurochem 120(2):259-268. doi: 10.1111/j.1471-4159.2011.07556.x
    • (2012) J Neurochem , vol.120 , Issue.2 , pp. 259-268
    • Perez-Carrion, M.D.1    Perez-Martinez, F.C.2    Merino, S.3    Sanchez-Verdu, P.4    Martinez-Hernandez, J.5    Lujan, R.6    Cena, V.7
  • 149
    • 81555222837 scopus 로고    scopus 로고
    • Identification of chemosensitivity nodes for vinblastine through small interfering RNA high-throughput screens
    • 10.1124/jpet.111.184879 1:CAS:528:DC%2BC38XjtVSksL0%3D 21880871
    • Kitchens CA, McDonald PR, Shun TY, Pollack IF, Lazo JS (2011) Identification of chemosensitivity nodes for vinblastine through small interfering RNA high-throughput screens. J Pharmacol Exp Ther 339(3):851-858. doi: 10.1124/jpet.111.184879
    • (2011) J Pharmacol Exp Ther , vol.339 , Issue.3 , pp. 851-858
    • Kitchens, C.A.1    McDonald, P.R.2    Shun, T.Y.3    Pollack, I.F.4    Lazo, J.S.5
  • 150
    • 80755159102 scopus 로고    scopus 로고
    • RNA interference targeting survivin exerts antitumoral effects in vitro and in established glioma xenografts in vivo
    • 10.1093/neuonc/nor098 1:CAS:528:DC%2BC38XhtVSgt7%2FO 3177660 21788344
    • Hendruschk S, Wiedemuth R, Aigner A, Topfer K, Cartellieri M, Martin D, Kirsch M, Ikonomidou C, Schackert G, Temme A (2011) RNA interference targeting survivin exerts antitumoral effects in vitro and in established glioma xenografts in vivo. Neuro Oncol 13(10):1074-1089. doi: 10.1093/neuonc/nor098
    • (2011) Neuro Oncol , vol.13 , Issue.10 , pp. 1074-1089
    • Hendruschk, S.1    Wiedemuth, R.2    Aigner, A.3    Topfer, K.4    Cartellieri, M.5    Martin, D.6    Kirsch, M.7    Ikonomidou, C.8    Schackert, G.9    Temme, A.10
  • 151
    • 84555220669 scopus 로고    scopus 로고
    • In vivo specific delivery of c-Met siRNA to glioblastoma using cationic solid lipid nanoparticles
    • 10.1021/bc200406n 1:CAS:528:DC%2BC3MXhsVelt73F 22070554
    • Jin J, Bae KH, Yang H, Lee SJ, Kim H, Kim Y, Joo KM, Seo SW, Park TG, Nam DH (2011) In vivo specific delivery of c-Met siRNA to glioblastoma using cationic solid lipid nanoparticles. Bioconjug Chem 22(12):2568-2572. doi: 10.1021/bc200406n
    • (2011) Bioconjug Chem , vol.22 , Issue.12 , pp. 2568-2572
    • Jin, J.1    Bae, K.H.2    Yang, H.3    Lee, S.J.4    Kim, H.5    Kim, Y.6    Joo, K.M.7    Seo, S.W.8    Park, T.G.9    Nam, D.H.10
  • 152
    • 67649317011 scopus 로고    scopus 로고
    • Delivery of small-interfering RNA (siRNA) to the brain
    • 10.1517/13543770802680195 1:CAS:528:DC%2BD1MXhsVCksrs%3D 2698461 19441914
    • Mathupala SP (2009) Delivery of small-interfering RNA (siRNA) to the brain. Expert Opin Ther Pat 19(2):137-140. doi: 10.1517/13543770802680195
    • (2009) Expert Opin Ther Pat , vol.19 , Issue.2 , pp. 137-140
    • Mathupala, S.P.1
  • 153
    • 79951550507 scopus 로고    scopus 로고
    • Angiogenesis and invasion in glioma
    • 10.1007/s10014-010-0007-z 1:CAS:528:DC%2BC3MXhvV2lsbk%3D 21221826
    • Onishi M, Ichikawa T, Kurozumi K, Date I (2011) Angiogenesis and invasion in glioma. Brain Tumor Pathol 28(1):13-24. doi: 10.1007/s10014-010-0007-z
    • (2011) Brain Tumor Pathol , vol.28 , Issue.1 , pp. 13-24
    • Onishi, M.1    Ichikawa, T.2    Kurozumi, K.3    Date, I.4
  • 154
    • 9144232309 scopus 로고    scopus 로고
    • Molecular mechanisms of glioma cell migration and invasion
    • DOI 10.1007/s11060-004-2751-6
    • Demuth T, Berens ME (2004) Molecular mechanisms of glioma cell migration and invasion. J Neurooncol 70(2):217-228. doi: 10.1007/s11060-004-2751-6 (Pubitemid 39545733)
    • (2004) Journal of Neuro-Oncology , vol.70 , Issue.2 , pp. 217-228
    • Demuth, T.1    Berens, M.E.2
  • 155
    • 84863503702 scopus 로고    scopus 로고
    • High-grade glioma motility reduced by genetic knockdown of KCC3
    • 10.1159/000339040 1:CAS:528:DC%2BC38XhtVKlt7nE 22776998
    • Gagnon KB (2012) High-grade glioma motility reduced by genetic knockdown of KCC3. Cell Physiol Biochem 30(2):466-476. doi: 10.1159/000339040
    • (2012) Cell Physiol Biochem , vol.30 , Issue.2 , pp. 466-476
    • Gagnon, K.B.1
  • 156
    • 84862705620 scopus 로고    scopus 로고
    • MiRNA expression profiling in migrating glioblastoma cells: Regulation of cell migration and invasion by miR-23b via targeting of Pyk2
    • 10.1371/journal.pone.0039818 3382150 22745829
    • Loftus JC, Ross JT, Paquette KM, Paulino VM, Nasser S, Yang Z, Kloss J, Kim S, Berens ME, Tran NL (2012) miRNA expression profiling in migrating glioblastoma cells: regulation of cell migration and invasion by miR-23b via targeting of Pyk2. PLoS ONE 7(6):e39818. doi: 10.1371/journal.pone.0039818
    • (2012) PLoS ONE , vol.7 , Issue.6 , pp. 39818
    • Loftus, J.C.1    Ross, J.T.2    Paquette, K.M.3    Paulino, V.M.4    Nasser, S.5    Yang, Z.6    Kloss, J.7    Kim, S.8    Berens, M.E.9    Tran, N.L.10
  • 157
    • 84856275188 scopus 로고    scopus 로고
    • Knockdown of ubiquitin ligases in glioblastoma cancer stem cells leads to cell death and differentiation
    • 10.1177/1087057111422565 1:CAS:528:DC%2BC38XjtVKlt7o%3D 21956171
    • Low J, Blosser W, Dowless M, Ricci-Vitiani L, Pallini R, de Maria R, Stancato L (2012) Knockdown of ubiquitin ligases in glioblastoma cancer stem cells leads to cell death and differentiation. J Biomol Screen 17(2):152-162. doi: 10.1177/1087057111422565
    • (2012) J Biomol Screen , vol.17 , Issue.2 , pp. 152-162
    • Low, J.1    Blosser, W.2    Dowless, M.3    Ricci-Vitiani, L.4    Pallini, R.5    De Maria, R.6    Stancato, L.7
  • 158
    • 84862200200 scopus 로고    scopus 로고
    • UPAR and cathepsin B inhibition enhanced radiation-induced apoptosis in gliomainitiating cells
    • 10.1093/neuonc/nos088 1:CAS:528:DC%2BC38Xhtl2hsb%2FL 3367854 22573309
    • Malla RR, Gopinath S, Alapati K, Gorantla B, Gondi CS, Rao JS (2012) uPAR and cathepsin B inhibition enhanced radiation-induced apoptosis in gliomainitiating cells. Neuro Oncol 14(6):745-760. doi: 10.1093/neuonc/nos088
    • (2012) Neuro Oncol , vol.14 , Issue.6 , pp. 745-760
    • Malla, R.R.1    Gopinath, S.2    Alapati, K.3    Gorantla, B.4    Gondi, C.S.5    Rao, J.S.6
  • 159
    • 84863602857 scopus 로고    scopus 로고
    • UPAR and cathepsin B knockdown inhibits radiation-induced PKC integrated integrin signaling to the cytoskeleton of glioma-initiating cells
    • 10.3892/ijo.2012.1496 1:CAS:528:DC%2BC38Xht1WltL%2FK 22641287
    • Alapati K, Gopinath S, Malla RR, Dasari VR, Rao JS (2012) uPAR and cathepsin B knockdown inhibits radiation-induced PKC integrated integrin signaling to the cytoskeleton of glioma-initiating cells. Int J Oncol 41(2):599-610. doi: 10.3892/ijo.2012.1496
    • (2012) Int J Oncol , vol.41 , Issue.2 , pp. 599-610
    • Alapati, K.1    Gopinath, S.2    Malla, R.R.3    Dasari, V.R.4    Rao, J.S.5
  • 160
    • 84855692830 scopus 로고    scopus 로고
    • Metabolic remodeling precedes mitochondrial outer membrane permeabilization in human glioma xenograft cells
    • 10.3892/ijo.2011.1255 1:CAS:528:DC%2BC38XitVWhtrw%3D 3232313 22076676
    • Ponnala S, Chetty C, Veeravalli KK, Dinh DH, Klopfenstein JD, Rao JS (2012) Metabolic remodeling precedes mitochondrial outer membrane permeabilization in human glioma xenograft cells. Int J Oncol 40(2):509-518. doi: 10.3892/ijo.2011.1255
    • (2012) Int J Oncol , vol.40 , Issue.2 , pp. 509-518
    • Ponnala, S.1    Chetty, C.2    Veeravalli, K.K.3    Dinh, D.H.4    Klopfenstein, J.D.5    Rao, J.S.6
  • 161
    • 77953194234 scopus 로고    scopus 로고
    • An extensive invasive intracranial human glioblastoma xenograft model: Role of high level matrix metalloproteinase 9
    • 10.2353/ajpath.2010.090571 1:CAS:528:DC%2BC3cXptFKrtro%3D 20413683
    • Zhao Y, Xiao A, diPierro CG, Carpenter JE, Abdel-Fattah R, Redpath GT, Lopes MB, Hussaini IM (2010) An extensive invasive intracranial human glioblastoma xenograft model: role of high level matrix metalloproteinase 9. Am J Pathol 176(6):3032-3049. doi: 10.2353/ajpath.2010.090571
    • (2010) Am J Pathol , vol.176 , Issue.6 , pp. 3032-3049
    • Zhao, Y.1    Xiao, A.2    Dipierro, C.G.3    Carpenter, J.E.4    Abdel-Fattah, R.5    Redpath, G.T.6    Lopes, M.B.7    Hussaini, I.M.8
  • 162
    • 84863377918 scopus 로고    scopus 로고
    • Effective adoptive transfer of haploidentical tumor-specific T cells in B16-melanoma bearing mice
    • 1:CAS:528:DC%2BC38XlvFSgsr0%3D
    • Cui NP, Xie SJ, Han JS, Ma ZF, Chen BP, Cai JH (2012) Effective adoptive transfer of haploidentical tumor-specific T cells in B16-melanoma bearing mice. Chin Med J (Engl) 125(5):794-800
    • (2012) Chin Med J (Engl) , vol.125 , Issue.5 , pp. 794-800
    • Cui, N.P.1    Xie, S.J.2    Han, J.S.3    Ma, Z.F.4    Chen, B.P.5    Cai, J.H.6
  • 164
    • 84859478908 scopus 로고    scopus 로고
    • Notch signaling pathway and cancer metastasis
    • 10.1007/978-1-4614-0899-4-14 1:CAS:528:DC%2BC38Xht12mt7fL 22399348
    • Hu YY, Zheng MH, Zhang R, Liang YM, Han H (2012) Notch signaling pathway and cancer metastasis. Adv Exp Med Biol 727:186-198. doi: 10.1007/978-1-4614- 0899-4-14
    • (2012) Adv Exp Med Biol , vol.727 , pp. 186-198
    • Hu, Y.Y.1    Zheng, M.H.2    Zhang, R.3    Liang, Y.M.4    Han, H.5
  • 165
    • 78651265055 scopus 로고    scopus 로고
    • IL-8 is a mediator of NF-kappaB induced invasion by gliomas
    • 10.1007/s11060-010-0261-2 1:CAS:528:DC%2BC3cXhsFajsL7N 20577780
    • Raychaudhuri B, Vogelbaum MA (2011) IL-8 is a mediator of NF-kappaB induced invasion by gliomas. J Neurooncol 101(2):227-235. doi: 10.1007/s11060-010-0261-2
    • (2011) J Neurooncol , vol.101 , Issue.2 , pp. 227-235
    • Raychaudhuri, B.1    Vogelbaum, M.A.2
  • 166
    • 72949116366 scopus 로고    scopus 로고
    • Regulation of cell signalling by uPAR
    • 10.1038/nrm2821 1:CAS:528:DC%2BD1MXhsFymtb7M 20027185
    • Smith HW, Marshall CJ (2010) Regulation of cell signalling by uPAR. Nat Rev Mol Cell Biol 11(1):23-36. doi: 10.1038/nrm2821
    • (2010) Nat Rev Mol Cell Biol , vol.11 , Issue.1 , pp. 23-36
    • Smith, H.W.1    Marshall, C.J.2
  • 167
    • 84884816982 scopus 로고    scopus 로고
    • Knockdown of cathepsin B and uPAR inhibits CD151 and alpha3beta1 integrin-mediated cell adhesion and invasion in glioma
    • 10.1002/mc.21915 22495828
    • Rao Malla R, Gopinath S, Alapati K, Gorantla B, Gondi CS, Rao JS (2012) Knockdown of cathepsin B and uPAR inhibits CD151 and alpha3beta1 integrin-mediated cell adhesion and invasion in glioma. Mol Carcinog. doi: 10.1002/mc.21915
    • (2012) Mol Carcinog
    • Rao Malla, R.1    Gopinath, S.2    Alapati, K.3    Gorantla, B.4    Gondi, C.S.5    Rao, J.S.6
  • 168
    • 80053529166 scopus 로고    scopus 로고
    • Synergistic induction of apoptosis in brain cancer cells by targeted codelivery of siRNA and anticancer drugs
    • 10.1021/mp100460h 1:CAS:528:DC%2BC3MXpsl2ntb0%3D 3185194 21793576
    • Kim C, Shah BP, Subramaniam P, Lee KB (2011) Synergistic induction of apoptosis in brain cancer cells by targeted codelivery of siRNA and anticancer drugs. Mol Pharm 8(5):1955-1961. doi: 10.1021/mp100460h
    • (2011) Mol Pharm , vol.8 , Issue.5 , pp. 1955-1961
    • Kim, C.1    Shah, B.P.2    Subramaniam, P.3    Lee, K.B.4
  • 169
    • 77955663516 scopus 로고    scopus 로고
    • Interaction of Beclin 1 with survivin regulates sensitivity of human glioma cells to TRAIL-induced apoptosis
    • 10.1016/j.febslet.2010.07.018 1:CAS:528:DC%2BC3cXhtVehtrbO 3210451 20638385
    • Niu TK, Cheng Y, Ren X, Yang JM (2010) Interaction of Beclin 1 with survivin regulates sensitivity of human glioma cells to TRAIL-induced apoptosis. FEBS Lett 584(16):3519-3524. doi: 10.1016/j.febslet.2010.07.018
    • (2010) FEBS Lett , vol.584 , Issue.16 , pp. 3519-3524
    • Niu, T.K.1    Cheng, Y.2    Ren, X.3    Yang, J.M.4
  • 170
    • 38449106717 scopus 로고    scopus 로고
    • Short hairpin RNA targeting survivin inhibits growth and angiogenesis of glioma U251 cells
    • 1:CAS:528:DC%2BD2sXhtlKhsrfI 17912437
    • Zhen HN, Li LW, Zhang W, Fei Z, Shi CH, Yang TT, Bai WT, Zhang X (2007) Short hairpin RNA targeting survivin inhibits growth and angiogenesis of glioma U251 cells. Int J Oncol 31(5):1111-1117
    • (2007) Int J Oncol , vol.31 , Issue.5 , pp. 1111-1117
    • Zhen, H.N.1    Li, L.W.2    Zhang, W.3    Fei, Z.4    Shi, C.H.5    Yang, T.T.6    Bai, W.T.7    Zhang, X.8
  • 171
    • 80155211502 scopus 로고    scopus 로고
    • Glioma growth inhibition in vitro and in vivo by single chain variable fragments of the transferrin receptor conjugated to survivin small interfering RNA
    • 22117970
    • Wang F, Bai HR, Wang J, Bai YZ, Dou CW (2011) Glioma growth inhibition in vitro and in vivo by single chain variable fragments of the transferrin receptor conjugated to survivin small interfering RNA. J Int Med Res 39(5):1701-1712
    • (2011) J Int Med Res , vol.39 , Issue.5 , pp. 1701-1712
    • Wang, F.1    Bai, H.R.2    Wang, J.3    Bai, Y.Z.4    Dou, C.W.5
  • 172
    • 78751483373 scopus 로고    scopus 로고
    • Bortezomib sensitizes malignant human glioma cells to TRAIL, mediated by inhibition of the NF-{kappa}B signaling pathway
    • 10.1158/1535-7163.MCT-10-0725 1:CAS:528:DC%2BC3MXjtFektw%3D%3D 3075591 21220502
    • Jane EP, Premkumar DR, Pollack IF (2011) Bortezomib sensitizes malignant human glioma cells to TRAIL, mediated by inhibition of the NF-{kappa}B signaling pathway. Mol Cancer Ther 10(1):198-208. doi: 10.1158/1535-7163.MCT-10-0725
    • (2011) Mol Cancer Ther , vol.10 , Issue.1 , pp. 198-208
    • Jane, E.P.1    Premkumar, D.R.2    Pollack, I.F.3
  • 173
    • 12344269275 scopus 로고    scopus 로고
    • RNA interference and nonviral targeted gene therapy of experimental brain cancer
    • DOI 10.1602/neurorx.2.1.139
    • Boado RJ (2005) RNA interference and nonviral targeted gene therapy of experimental brain cancer. NeuroRx 2(1):139-150. doi: 10.1602/neurorx.2.1.139 (Pubitemid 40128034)
    • (2005) NeuroRx , vol.2 , Issue.1 , pp. 139-150
    • Boado, R.J.1
  • 174
    • 84855186726 scopus 로고    scopus 로고
    • Bevacizumab in the treatment of high-grade gliomas: An overview
    • 10.1007/s10456-011-9232-2 1:CAS:528:DC%2BC3MXhsVKhu7rK 21858651
    • Kunnakkat S, Narayana A (2011) Bevacizumab in the treatment of high-grade gliomas: an overview. Angiogenesis 14(4):423-430. doi: 10.1007/s10456-011-9232- 2
    • (2011) Angiogenesis , vol.14 , Issue.4 , pp. 423-430
    • Kunnakkat, S.1    Narayana, A.2
  • 175
    • 66149135691 scopus 로고    scopus 로고
    • C-Met-targeted RNA interference inhibits growth and metastasis of glioma U251 cells in vitro
    • 10.1007/s11060-008-9772-5 1:CAS:528:DC%2BD1MXlvFOms7o%3D 19165419
    • Chu SH, Feng DF, Zhang H, Chen ET, Duan ZX, Li XY, Li J, Ma YB, Zhu ZA, Qiu JH (2009) c-Met-targeted RNA interference inhibits growth and metastasis of glioma U251 cells in vitro. J Neurooncol 93(2):183-189. doi: 10.1007/s11060-008-9772-5
    • (2009) J Neurooncol , vol.93 , Issue.2 , pp. 183-189
    • Chu, S.H.1    Feng, D.F.2    Zhang, H.3    Chen, E.T.4    Duan, Z.X.5    Li, X.Y.6    Li, J.7    Ma, Y.B.8    Zhu, Z.A.9    Qiu, J.H.10
  • 176
    • 78651273961 scopus 로고    scopus 로고
    • Phosphatase and tensin homolog reconstruction and vascular endothelial growth factor knockdown synergistically inhibit the growth of glioblastoma
    • 10.1089/cbr.2010.0821 1:CAS:528:DC%2BC3MXht1yhug%3D%3D 21204766
    • Chen H, Shen X, Guo C, Zhu H, Zhou L, Zhu Y, Wang H, Zheng Y, Huang L (2010) Phosphatase and tensin homolog reconstruction and vascular endothelial growth factor knockdown synergistically inhibit the growth of glioblastoma. Cancer Biother Radiopharm 25(6):713-721. doi: 10.1089/cbr.2010.0821
    • (2010) Cancer Biother Radiopharm , vol.25 , Issue.6 , pp. 713-721
    • Chen, H.1    Shen, X.2    Guo, C.3    Zhu, H.4    Zhou, L.5    Zhu, Y.6    Wang, H.7    Zheng, Y.8    Huang, L.9
  • 177
    • 70349105997 scopus 로고    scopus 로고
    • The epidermal growth factor receptor as a therapeutic target in glioblastoma multiforme and other malignant neoplasms
    • 1:CAS:528:DC%2BD1MXovVKgu7w%3D 19601750
    • Loew S, Schmidt U, Unterberg A, Halatsch ME (2009) The epidermal growth factor receptor as a therapeutic target in glioblastoma multiforme and other malignant neoplasms. Anticancer Agents Med Chem 9(6):703-715
    • (2009) Anticancer Agents Med Chem , vol.9 , Issue.6 , pp. 703-715
    • Loew, S.1    Schmidt, U.2    Unterberg, A.3    Halatsch, M.E.4
  • 178
    • 73949121242 scopus 로고    scopus 로고
    • Induction of in vivo synthetic lethal RNAi responses to treat glioblastoma
    • 19901546
    • Michiue H, Eguchi A, Scadeng M, Dowdy SF (2009) Induction of in vivo synthetic lethal RNAi responses to treat glioblastoma. Cancer Biol Ther 8(23):2306-2313
    • (2009) Cancer Biol Ther , vol.8 , Issue.23 , pp. 2306-2313
    • Michiue, H.1    Eguchi, A.2    Scadeng, M.3    Dowdy, S.F.4
  • 179
    • 80052844556 scopus 로고    scopus 로고
    • B4GALNT3 expression predicts a favorable prognosis and suppresses cell migration and invasion via beta(1) integrin signaling in neuroblastoma
    • 10.1016/j.ajpath.2011.05.025 1:CAS:528:DC%2BC3MXhtF2jsb%2FO 21741930
    • Hsu WM, Che MI, Liao YF, Chang HH, Chen CH, Huang YM, Jeng YM, Huang J, Quon MJ, Lee H, Huang HC, Huang MC (2011) B4GALNT3 expression predicts a favorable prognosis and suppresses cell migration and invasion via beta(1) integrin signaling in neuroblastoma. Am J Pathol 179(3):1394-1404. doi: 10.1016/j.ajpath.2011.05.025
    • (2011) Am J Pathol , vol.179 , Issue.3 , pp. 1394-1404
    • Hsu, W.M.1    Che, M.I.2    Liao, Y.F.3    Chang, H.H.4    Chen, C.H.5    Huang, Y.M.6    Jeng, Y.M.7    Huang, J.8    Quon, M.J.9    Lee, H.10    Huang, H.C.11    Huang, M.C.12
  • 180
    • 84861839598 scopus 로고    scopus 로고
    • 2-Methoxyestradiol induced bax phosphorylation and apoptosis in human retinoblastoma cells via p38 MAPK activation
    • 10.1002/mc.20825 1:CAS:528:DC%2BC38XnvFOqsrw%3D 21769948
    • Min H, Ghatnekar GS, Ghatnekar AV, You X, Bu M, Guo X, Bu S, Shen B, Huang Q (2012) 2-Methoxyestradiol induced bax phosphorylation and apoptosis in human retinoblastoma cells via p38 MAPK activation. Mol Carcinog 51(7):576-585. doi: 10.1002/mc.20825
    • (2012) Mol Carcinog , vol.51 , Issue.7 , pp. 576-585
    • Min, H.1    Ghatnekar, G.S.2    Ghatnekar, A.V.3    You, X.4    Bu, M.5    Guo, X.6    Bu, S.7    Shen, B.8    Huang, Q.9
  • 181
    • 80052811789 scopus 로고    scopus 로고
    • Reversal of stathmin-mediated microtubule destabilization sensitizes retinoblastoma cells to a low dose of antimicrotubule agents: A novel synergistic therapeutic intervention
    • 10.1167/iovs.10-6973 1:CAS:528:DC%2BC3MXhtFKqtrzO 21546534
    • Mitra M, Kandalam M, Sundaram CS, Verma RS, Maheswari UK, Swaminathan S, Krishnakumar S (2011) Reversal of stathmin-mediated microtubule destabilization sensitizes retinoblastoma cells to a low dose of antimicrotubule agents: a novel synergistic therapeutic intervention. Invest Ophthalmol Vis Sci 52(8):5441-5448. doi: 10.1167/iovs.10-6973
    • (2011) Invest Ophthalmol Vis Sci , vol.52 , Issue.8 , pp. 5441-5448
    • Mitra, M.1    Kandalam, M.2    Sundaram, C.S.3    Verma, R.S.4    Maheswari, U.K.5    Swaminathan, S.6    Krishnakumar, S.7
  • 182
    • 79952623093 scopus 로고    scopus 로고
    • Treatment with connexin 46 siRNA suppresses the growth of human Y79 retinoblastoma cell xenografts in vivo
    • 10.1016/j.exer.2011.02.003 1:CAS:528:DC%2BC3MXjsV2nsLY%3D 3060947 21320488
    • Burr DB, Molina SA, Banerjee D, Low DM, Takemoto DJ (2011) Treatment with connexin 46 siRNA suppresses the growth of human Y79 retinoblastoma cell xenografts in vivo. Exp Eye Res 92(4):251-259. doi: 10.1016/j.exer.2011.02.003
    • (2011) Exp Eye Res , vol.92 , Issue.4 , pp. 251-259
    • Burr, D.B.1    Molina, S.A.2    Banerjee, D.3    Low, D.M.4    Takemoto, D.J.5
  • 183
    • 84860511567 scopus 로고    scopus 로고
    • Age-related macular degeneration
    • 10.1016/S0140-6736(12)60282-7 22559899
    • Lim LS, Mitchell P, Seddon JM, Holz FG, Wong TY (2012) Age-related macular degeneration. Lancet 379(9827):1728-1738. doi: 10.1016/S0140-6736(12) 60282-7
    • (2012) Lancet , vol.379 , Issue.9827 , pp. 1728-1738
    • Lim, L.S.1    Mitchell, P.2    Seddon, J.M.3    Holz, F.G.4    Wong, T.Y.5
  • 186
    • 79956053371 scopus 로고    scopus 로고
    • Age-related macular degeneration and the other double helix the Cogan lecture
    • 10.1167/iovs.11-7328 21471430
    • Ambati J (2011) Age-related macular degeneration and the other double helix. The Cogan lecture. Invest Ophthalmol Vis Sci 52(5):2165-2169. doi: 10.1167/iovs.11-7328
    • (2011) Invest Ophthalmol Vis Sci , vol.52 , Issue.5 , pp. 2165-2169
    • Ambati, J.1
  • 187
    • 84857981475 scopus 로고    scopus 로고
    • Translational investigation and treatment of neuropathic pain
    • 10.1186/1744-8069-8-15 1:CAS:528:DC%2BC38Xns1GntLo%3D 3320533 22400804
    • Xu B, Descalzi G, Ye HR, Zhuo M, Wang YW (2012) Translational investigation and treatment of neuropathic pain. Mol Pain 8:15. doi: 10.1186/1744-8069-8-15
    • (2012) Mol Pain , vol.8 , pp. 15
    • Xu, B.1    Descalzi, G.2    Ye, H.R.3    Zhuo, M.4    Wang, Y.W.5
  • 189
    • 69249113423 scopus 로고    scopus 로고
    • Angiopoietin 2 mediates the differentiation and migration of neural progenitor cells in the subventricular zone after stroke
    • 10.1074/jbc.M109.006551 1:CAS:528:DC%2BD1MXpvFKit70%3D 19553662
    • Liu XS, Chopp M, Zhang RL, Hozeska-Solgot A, Gregg SC, Buller B, Lu M, Zhang ZG (2009) Angiopoietin 2 mediates the differentiation and migration of neural progenitor cells in the subventricular zone after stroke. J Biol Chem 284(34):22680-22689. doi: 10.1074/jbc.M109.006551
    • (2009) J Biol Chem , vol.284 , Issue.34 , pp. 22680-22689
    • Liu, X.S.1    Chopp, M.2    Zhang, R.L.3    Hozeska-Solgot, A.4    Gregg, S.C.5    Buller, B.6    Lu, M.7    Zhang, Z.G.8
  • 190
    • 34247373879 scopus 로고    scopus 로고
    • V1.8 tetrodotoxin-resistant sodium channel reverses mechanical allodynia in neuropathic rats
    • DOI 10.1016/j.neuroscience.2007.01.054, PII S0306452207001145
    • Dong XW, Goregoaker S, Engler H, Zhou X, Mark L, Crona J, Terry R, Hunter J, Priestley T (2007) Small interfering RNA-mediated selective knockdown of Na(V)1.8 tetrodotoxin-resistant sodium channel reverses mechanical allodynia in neuropathic rats. Neuroscience 146(2):812-821. doi: 10.1016/j.neuroscience.2007. 01.054 (Pubitemid 46628998)
    • (2007) Neuroscience , vol.146 , Issue.2 , pp. 812-821
    • Dong, X.-W.1    Goregoaker, S.2    Engler, H.3    Zhou, X.4    Mark, L.5    Crona, J.6    Terry, R.7    Hunter, J.8    Priestley, T.9
  • 191
    • 70350127316 scopus 로고    scopus 로고
    • Role of M2, M3, and M4 muscarinic receptor subtypes in the spinal cholinergic control of nociception revealed using siRNA in rats
    • 10.1111/j.1471-4159.2009.06396.x 1:CAS:528:DC%2BD1MXhsVSkurzI 19780895
    • Cai YQ, Chen SR, Han HD, Sood AK, Lopez-Berestein G, Pan HL (2009) Role of M2, M3, and M4 muscarinic receptor subtypes in the spinal cholinergic control of nociception revealed using siRNA in rats. J Neurochem 111(4):1000-1010. doi: 10.1111/j.1471-4159.2009.06396.x
    • (2009) J Neurochem , vol.111 , Issue.4 , pp. 1000-1010
    • Cai, Y.Q.1    Chen, S.R.2    Han, H.D.3    Sood, A.K.4    Lopez-Berestein, G.5    Pan, H.L.6
  • 193
    • 84865148347 scopus 로고    scopus 로고
    • Calcium regulation by thermo- and osmosensing transient receptor potential vanilloid channels (TRPVs) in human conjunctival epithelial cells
    • 10.1007/s00418-012-0924-5 1:CAS:528:DC%2BC38XmvF2ntr4%3D 22327830
    • Mergler S, Garreis F, Sahlmuller M, Lyras EM, Reinach PS, Dwarakanath A, Paulsen F, Pleyer U (2012) Calcium regulation by thermo- and osmosensing transient receptor potential vanilloid channels (TRPVs) in human conjunctival epithelial cells. Histochem Cell Biol 137(6):743-761. doi: 10.1007/s00418-012- 0924-5
    • (2012) Histochem Cell Biol , vol.137 , Issue.6 , pp. 743-761
    • Mergler, S.1    Garreis, F.2    Sahlmuller, M.3    Lyras, E.M.4    Reinach, P.S.5    Dwarakanath, A.6    Paulsen, F.7    Pleyer, U.8
  • 194
    • 79952275918 scopus 로고    scopus 로고
    • TRPV1 activation is required for hypertonicity-stimulated inflammatory cytokine release in human corneal epithelial cells
    • 10.1167/iovs.10-5801 1:CAS:528:DC%2BC3MXmsVShtLs%3D 20739465
    • Pan Z, Wang Z, Yang H, Zhang F, Reinach PS (2011) TRPV1 activation is required for hypertonicity-stimulated inflammatory cytokine release in human corneal epithelial cells. Invest Ophthalmol Vis Sci 52(1):485-493. doi: 10.1167/iovs.10-5801
    • (2011) Invest Ophthalmol Vis Sci , vol.52 , Issue.1 , pp. 485-493
    • Pan, Z.1    Wang, Z.2    Yang, H.3    Zhang, F.4    Reinach, P.S.5
  • 195
    • 84870355461 scopus 로고    scopus 로고
    • Brain-targeting study of stearic acid-grafted chitosan micelle drug-delivery system
    • 10.2147/IJN.S32701 1:CAS:528:DC%2BC38XhtVakur3M 3396390 22802685
    • Xie YT, Du YZ, Yuan H, Hu FQ (2012) Brain-targeting study of stearic acid-grafted chitosan micelle drug-delivery system. Int J Nanomedicine 7:3235-3244. doi: 10.2147/IJN.S32701
    • (2012) Int J Nanomedicine , vol.7 , pp. 3235-3244
    • Xie, Y.T.1    Du, Y.Z.2    Yuan, H.3    Hu, F.Q.4
  • 196
    • 84877067774 scopus 로고    scopus 로고
    • Protein functionalized tramadol-loaded PLGA nanoparticles: Preparation, optimization, stability and pharmacodynamic studies
    • 10.3109/03639045.2012.684390 22799442
    • Lalani J, Rathi M, Lalan M, Misra A (2012) Protein functionalized tramadol-loaded PLGA nanoparticles: preparation, optimization, stability and pharmacodynamic studies. Drug Dev Ind Pharm. doi: 10.3109/03639045.2012.684390
    • (2012) Drug Dev Ind Pharm
    • Lalani, J.1    Rathi, M.2    Lalan, M.3    Misra, A.4


* 이 정보는 Elsevier사의 SCOPUS DB에서 KISTI가 분석하여 추출한 것입니다.