메뉴 건너뛰기




Volumn 4, Issue 4, 2003, Pages 296-307

Modelling brain diseases in mice: The challenges of design and analysis

Author keywords

[No Author keywords available]

Indexed keywords

ACONITATE HYDRATASE; AMYLOID PRECURSOR PROTEIN; CRE RECOMBINASE; DNA; POLYGLUTAMINE; PRESENILIN 1; PRESENILIN 2; SYNUCLEIN;

EID: 0037379663     PISSN: 14710056     EISSN: None     Source Type: Journal    
DOI: 10.1038/nrg1045     Document Type: Review
Times cited : (77)

References (149)
  • 1
    • 0029014350 scopus 로고
    • Acid sphingomyelinase-deficient mice mimic the neurovisceral form of human lysosomal storage disease (Niemann-Pick disease)
    • Otterbach, B. & Stoffel, W. Acid sphingomyelinase-deficient mice mimic the neurovisceral form of human lysosomal storage disease (Niemann-Pick disease). Cell 81, 1053-1061(1995).
    • (1995) Cell , vol.81 , pp. 1053-1061
    • Otterbach, B.1    Stoffel, W.2
  • 2
    • 0029012443 scopus 로고
    • Acid sphingomyelinase deficient mice: A model of types A and B Niemann-Pick disease
    • Horinouchi, K. et al. Acid sphingomyelinase deficient mice: a model of types A and B Niemann-Pick disease Nature Genet. 10, 288-293 (1995).
    • (1995) Nature Genet. , vol.10 , pp. 288-293
    • Horinouchi, K.1
  • 3
    • 0031764610 scopus 로고    scopus 로고
    • Progressive ataxia, myoclonic epilepsy, and cerebellar apoptosis in cystatin B-deficient mice
    • Pennachio, L. A et al. Progressive ataxia, myoclonic epilepsy, and cerebellar apoptosis in cystatin B-deficient mice. Nature Genet. 20, 251-258 (1998).
    • (1998) Nature Genet. , vol.20 , pp. 251-258
    • Pennachio, L.A.1
  • 4
    • 0032491408 scopus 로고    scopus 로고
    • Genetic classification of primary neurodegenerative disease
    • Hardy, J. & Gwinn-Hardy, K. Genetic classification of primary neurodegenerative disease. Science 282, 1075-1079 (1998).
    • (1998) Science , vol.282 , pp. 1075-1079
    • Hardy, J.1    Gwinn-Hardy, K.2
  • 5
    • 0037077040 scopus 로고    scopus 로고
    • Toxic proteins in neurodegenerative disease
    • Taylor, J. P., Hardy, J. & Fischbeck, K. H. Toxic proteins in neurodegenerative disease. Science 296, 1991-1995 (2002).
    • (2002) Science , vol.296 , pp. 1991-1995
    • Taylor, J.P.1    Hardy, J.2    Fischbeck, K.H.3
  • 6
    • 0034329159 scopus 로고    scopus 로고
    • Molecular genetics: Unmasking polyglutamine triggers in neurodegnerative disease
    • Gusella, J. F. & MacDonald, M. E. Molecular genetics: unmasking polyglutamine triggers in neurodegnerative disease. Nature Rev. Neurosci 1, 109-115 (2000).
    • (2000) Nature Rev. Neurosci. , vol.1 , pp. 109-115
    • Gusella, J.F.1    MacDonald, M.E.2
  • 7
    • 0035475788 scopus 로고    scopus 로고
    • Y SCA1 molecular genetics: A history of a 13 year collaboration against glutamines
    • Orr, H. T. & Zoghbi, H. Y SCA1 molecular genetics: a history of a 13 year collaboration against glutamines, Hum. Mol. Genet. 10, 2307-2311 (2001).
    • (2001) Hum. Mol. Genet. , vol.10 , pp. 2307-2311
    • Orr, H.T.1    Zoghbi, H.2
  • 8
    • 0029163222 scopus 로고
    • SCA1 transgenic mice: A model for neurodegeneration caused by an expanded CAG trinucleotide repeat
    • Burright, E. et al. SCA1 transgenic mice: a model for neurodegeneration caused by an expanded CAG trinucleotide repeat. Cell 82, 937-948 (1995).
    • (1995) Cell , vol.82 , pp. 937-948
    • Burright, E.1
  • 9
    • 0032528167 scopus 로고    scopus 로고
    • Mice lacking ataxin-1 display learning deficits and decreased hippocampal paired-pulse facilitation
    • Matilla, A. et al. Mice lacking ataxin-1 display learning deficits and decreased hippocampal paired-pulse facilitation. J. Neurosci. 18, 5508-5516 (1998).
    • (1998) J. Neurosci. , vol.18 , pp. 5508-5516
    • Matilla, A.1
  • 10
    • 0034597833 scopus 로고    scopus 로고
    • Identification of genes that modify ataxin-1-induced neurodegeneration
    • Fernandez-Funez, P. et al. Identification of genes that modify ataxin-1-induced neurodegeneration. Nature 408, 101-106 (2000).
    • (2000) Nature , vol.408 , pp. 101-106
    • Fernandez-Funez, P.1
  • 11
    • 0032475941 scopus 로고    scopus 로고
    • Ataxin-1 nuclear localization and aggregation: Role in polyglutamine-induced disease in SCA1 transgenic mice
    • Klement, I, A. et al. Ataxin-1 nuclear localization and aggregation: role in polyglutamine-induced disease in SCA1 transgenic mice Cell 95, 41-53 (1998).
    • (1998) Cell , vol.95 , pp. 41-53
    • Klement, I.A.1
  • 12
    • 0034701278 scopus 로고    scopus 로고
    • Repeat instability and motor incoordination in mice with a targeted expanded CAG repeat in the Sca1 locus
    • Lorenzetti, D. et al. Repeat instability and motor incoordination in mice with a targeted expanded CAG repeat in the Sca1 locus, Hum. Mol. Genet. 9, 779-785 (2000).
    • (2000) Hum. Mol. Genet. , vol.9 , pp. 779-785
    • Lorenzetti, D.1
  • 13
    • 18444386197 scopus 로고    scopus 로고
    • A long CAG repeat in the mouse Scal locus replicates SCA1 features and reveals the impact of protein solubility on selective neurodegeneration
    • Watase, K. et al. A long CAG repeat in the mouse Scal locus replicates SCA1 features and reveals the impact of protein solubility on selective neurodegeneration Neuron 34, 905-919 (2002) This paper reports on mice that express the mutant protein with endogenous patterns and replicate the selective neuronal vulnerability that is found in polyglutamine disorders.
    • (2002) Neuron , vol.34 , pp. 905-919
    • Watase, K.1
  • 14
    • 0027291710 scopus 로고
    • Subclinical phenotypic expressionsin heterozygous females of X-linked recessive bulbospinal neuronopathy
    • Sobue, G. et al. Subclinical phenotypic expressionsin heterozygous females of X-linked recessive bulbospinal neuronopathy J. Neurol. Sci. 117, 74-78 (1993).
    • (1993) J. Neurol. Sci. , vol.117 , pp. 74-78
    • Sobue, G.1
  • 15
    • 0034793311 scopus 로고    scopus 로고
    • Clinical features and skewed X-chromosome inactivation in female carriers of X-linked recessive spinal and bulbar muscular atrophy
    • Ishihara, H. et al. Clinical features and skewed X-chromosome inactivation in female carriers of X-linked recessive spinal and bulbar muscular atrophy. J. Neurol 248. 856-860 (2001).
    • (2001) J. Neurol. , vol.248 , pp. 856-860
    • Ishihara, H.1
  • 16
    • 0028952350 scopus 로고
    • Androgen receptor (CAG)n repeat analysis in the differential diagnosis between Kennedy disease and other motor neuron disorders
    • Ferlini, A. et al. Androgen receptor (CAG)n repeat analysis in the differential diagnosis between Kennedy disease and other motor neuron disorders Am. J. Med. Genet. 55, 105-111(1995).
    • (1995) Am. J. Med. Genet. , vol.55 , pp. 105-111
    • Ferlini, A.1
  • 18
    • 0028878844 scopus 로고
    • Stability of an expanded trinucleotide repeat in the androgen receptor gene in transgenic mice
    • Bingham, P. M. et al. Stability of an expanded trinucleotide repeat in the androgen receptor gene in transgenic mice. Nature Genet. 9, 191-196 (1995).
    • (1995) Nature Genet. , vol.9 , pp. 191-196
    • Bingham, P.M.1
  • 19
    • 7144256250 scopus 로고    scopus 로고
    • Androgen receptor YAC transgenic mice carrying CAG 45 alleles show trinuclectide repeat instability
    • La Spada, A. R. et al. Androgen receptor YAC transgenic mice carrying CAG 45 alleles show trinuclectide repeat instability. Hum Mol Genet. 7, 959-967 (1998).
    • (1998) Hum. Mol. Genet. , vol.7 , pp. 959-967
    • La Spada, A.R.1
  • 20
    • 0035862754 scopus 로고    scopus 로고
    • Expression of expanded repeat androgen receptor produces neurologic disease in transgenic mice
    • Abel, A. et al. Expression of expanded repeat androgen receptor produces neurologic disease in transgenic mice. Hum. Mol. Genet, 10, 107-116 (2001).
    • (2001) Hum. Mol. Genet. , vol.10 , pp. 107-116
    • Abel, A.1
  • 21
    • 0030058208 scopus 로고    scopus 로고
    • Expanded polyglutamine in the Machado-Joseph disease protein induces cell death in vitro and in vivo
    • Ikeda, H. et al. Expanded polyglutamine in the Machado-Joseph disease protein induces cell death in vitro and in vivo. Nature Genet. 13, 196-202 (1996).
    • (1996) Nature Genet. , vol.13 , pp. 196-202
    • Ikeda, H.1
  • 22
    • 16044373842 scopus 로고    scopus 로고
    • Exon 1 of the HD gene with an expanded CAG repeat is sufficient to cause a progressive neurological phenotype in transgenic mice
    • Mangiarini, L. et al. Exon 1 of the HD gene with an expanded CAG repeat is sufficient to cause a progressive neurological phenotype in transgenic mice. Cell 87, 493-506 (1996).
    • (1996) Cell , vol.87 , pp. 493-506
    • Mangiarini, L.1
  • 23
    • 0036713608 scopus 로고    scopus 로고
    • A mouse model of spinal and bulbar muscular atrophy
    • McManamny, P. et al. A mouse model of spinal and bulbar muscular atrophy. Hum. Mol. Genet. 18, 2103-2111 (2002).
    • (2002) Hum. Mol. Genet. , vol.18 , pp. 2103-2111
    • McManamny, P.1
  • 24
    • 18644379256 scopus 로고    scopus 로고
    • Testosterone reduction prevents phenotypic expression in a transgenic mouse model of spinal and bulbar muscular atrophy
    • Katsuno, M. et al. Testosterone reduction prevents phenotypic expression in a transgenic mouse model of spinal and bulbar muscular atrophy. Neuron 35, 843-854 (2002) Through the creation of a transgenic animal model expressing full-length mutant AR cDNA, this study indicated that nuclear translocation of the mutant AR by testosterone might contribute to the profound gender differences of phenotypes in SBMA and showed the therapeutic potential of hormonal intervention.
    • (2002) Neuron , vol.35 , pp. 843-854
    • Katsuno, M.1
  • 25
    • 0037421691 scopus 로고    scopus 로고
    • SCA7 knock-in mice reproduce features of human SCA7 and reveal that mutant ataxin-7 gradually accumulates in neurons and interferes with short-term synaptic plasticity
    • Yoo, S. Y. et al. SCA7 knock-in mice reproduce features of human SCA7 and reveal that mutant ataxin-7 gradually accumulates in neurons and interferes with short-term synaptic plasticity. Neuron 37, 383401 (2003). By inserting highly expanded CAG repeat seen in juvenile cases of SCA7 into the corresponding mouse locus, the authors succeeded in generating an accurate model for this polyglutamine disease.
    • (2003) Neuron , vol.37 , pp. 383-401
    • Yoo, S.Y.1
  • 26
    • 0037154184 scopus 로고    scopus 로고
    • Recent advances in the genetics and pathogenesis of Parkinson disease
    • Mouradian, M. M. Recent advances in the genetics and pathogenesis of Parkinson disease. Neurology 58, 179-185 (2002).
    • (2002) Neurology , vol.58 , pp. 179-185
    • Mouradian, M.M.1
  • 27
    • 0032543684 scopus 로고    scopus 로고
    • Association of missense and 5′-splice-site mutations in tau with the inherited dementia FTDP-17
    • Hutton, M. et al. Association of missense and 5′-splice-site mutations in tau with the inherited dementia FTDP-17. Nature 393, 702-705 (1998).
    • (1998) Nature , vol.393 , pp. 702-705
    • Hutton, M.1
  • 28
    • 0034681471 scopus 로고    scopus 로고
    • Dopaminergic loss and inclusion body formation in α-synuclein mice: Implications for neurodegenerative disorders
    • Masliah, E. et al. Dopaminergic loss and inclusion body formation in α-synuclein mice: implications for neurodegenerative disorders. Science 287, 1265-1269 (2000). This paper shows that the accumulation of wild-type α-synuclein causes dopaminergic neuron dysfunction and loss in transgenic mice.
    • (2000) Science , vol.287 , pp. 1265-1269
    • Masliah, E.1
  • 29
    • 0034663176 scopus 로고    scopus 로고
    • Neuropathology in mice expressing human α-synuclein
    • van der Putten, H. et al. Neuropathology in mice expressing human α-synuclein. J. Neurosci. 20, 6021-6029 (2000).
    • (2000) J. Neurosci. , vol.20 , pp. 6021-6029
    • Van der Putten, H.1
  • 30
    • 0037118259 scopus 로고    scopus 로고
    • Neuronal α-synucleinopathy with severe movement disorder in mice expressing A53T human α-synuclein
    • Giasson, B. I. et al. Neuronal α-synucleinopathy with severe movement disorder in mice expressing A53T human α-synuclein. Neuron 34, 521-533 (2002).
    • (2002) Neuron , vol.34 , pp. 521-533
    • Giasson, B.I.1
  • 31
    • 0036855635 scopus 로고    scopus 로고
    • Misfolded proteinase-K resistant hyperphosphorylated α-synuclein in aged transgenic mice with locomotor deterioration and in α-synucleinopathies
    • Neumann, M. et al. Misfolded proteinase-K resistant hyperphosphorylated α-synuclein in aged transgenic mice with locomotor deterioration and in α-synucleinopathies. J. Clin. Invest. 110, 1429-1439 (2002).
    • (2002) J. Clin. Invest. , vol.110 , pp. 1429-1439
    • Neumann, M.1
  • 32
    • 0037333642 scopus 로고    scopus 로고
    • Motor dysfunction and gliosis with preserved dopaminergic markers in human α-synuclein A30P transgenic mice
    • Gomez-Isla, T. et al. Motor dysfunction and gliosis with preserved dopaminergic markers in human α-synuclein A30P transgenic mice. Neurobiol Aging 24, 245-258 (2003).
    • (2003) Neurobiol Aging , vol.24 , pp. 245-258
    • Gomez-Isla, T.1
  • 33
    • 0035651255 scopus 로고    scopus 로고
    • BAC to the future: The use of BAC transgenic mice for neuroscience research
    • Heintz, N. BAC to the future: the use of BAC transgenic mice for neuroscience research. Nature Rev Neurosci. 2, 861-870 (2001). An excellent review of the usefulness of BAC transgenes as a tool for in vivo functional studies of genes implicated in nervous-system function and dysfunction.
    • (2001) Nature Rev. Neurosci. , vol.2 , pp. 861-870
    • Heintz, N.1
  • 34
    • 0035487293 scopus 로고    scopus 로고
    • Recombineering: A powerful new tool for mouse functional genomics
    • Copeland, N. G., Jenkins, N. A. & Court, D. L. Recombineering: a powerful new tool for mouse functional genomics. Nature Rev. Genet. 2, 769-779 (2001). This review gives full details of the recent technological advances in a phage-based homologous recombination system that enables the modification and subcloning of genomic DNA in BACs.
    • (2001) Nature Rev. Genet. , vol.2 , pp. 769-779
    • Copeland, N.G.1    Jenkins, N.A.2    Court, D.L.3
  • 35
    • 0019782799 scopus 로고
    • Friedreich ataxia: A clinical and genetic study of 90 families with an analysis of early diagnostic criteria and intrafamilial clustering of clinical features
    • Harding, A. E. Friedreich ataxia: a clinical and genetic study of 90 families with an analysis of early diagnostic criteria and intrafamilial clustering of clinical features. Brain 104, 589-620 (1981).
    • (1981) Brain , vol.104 , pp. 589-620
    • Harding, A.E.1
  • 36
    • 13344270899 scopus 로고    scopus 로고
    • Friedreich ataxia: Autosomal recessive disease caused by an intronic GAA triplet repeat expansion
    • Campuzano, V. et al Friedreich ataxia: autosomal recessive disease caused by an intronic GAA triplet repeat expansion. Science 271, 1423-1427 (1996).
    • (1996) Science , vol.271 , pp. 1423-1427
    • Campuzano, V.1
  • 37
    • 9844222853 scopus 로고    scopus 로고
    • Frataxin is reduced in Friedreich ataxia patients and is associated with mitochondrial membranes
    • Campuzano, V. et al. Frataxin is reduced in Friedreich ataxia patients and is associated with mitochondrial membranes. Hum. Mol. Genet, 8, 1771-1780 (1997).
    • (1997) Hum. Mol. Genet. , vol.8 , pp. 1771-1780
    • Campuzano, V.1
  • 38
    • 0031253821 scopus 로고    scopus 로고
    • Frataxin gene expansion causes aconitase and mitochondrial iron-sulfer protein deficiency in Friedreich ataxia
    • Rötig, A. et al. Frataxin gene expansion causes aconitase and mitochondrial iron-sulfer protein deficiency in Friedreich ataxia. Nature Genet. 17, 215-217 (1997).
    • (1997) Nature Genet. , vol.17 , pp. 215-217
    • Rötig, A.1
  • 39
    • 0034192352 scopus 로고    scopus 로고
    • Inactivation of Friedreich ataxia mouse gene leads to early embryonic lethality without iron accumulation
    • Cossée, M. et al. Inactivation of Friedreich ataxia mouse gene leads to early embryonic lethality without iron accumulation. Hum. Mol. Genet. 9, 1219-1226 (2000).
    • (2000) Hum. Mol. Genet. , vol.9 , pp. 1219-1226
    • Cossée, M.1
  • 40
    • 0035138072 scopus 로고    scopus 로고
    • Mouse models for Friedreich ataxia exhibit cardiomyopathy, sensory nerve defects and Fe-S enzyme deficiency followed by mitochondrial iron deposits
    • Puccio, H. et al. Mouse models for Friedreich ataxia exhibit cardiomyopathy, sensory nerve defects and Fe-S enzyme deficiency followed by mitochondrial iron deposits Nature Genet. 27, 181-186 (2001). This paper reports on the creation of FRDA mouse models using conditional gene-targeting approaches. The mice reproduced both pathological and biochemical features of the human disease.
    • (2001) Nature Genet. , vol.27 , pp. 181-186
    • Puccio, H.1
  • 41
    • 18244408334 scopus 로고    scopus 로고
    • Frataxin knock-in mouse
    • Miranda, C. J. et al. Frataxin knock-in mouse. FEBS Lett 512, 291-297 (2002).
    • (2002) FEBS Lett. , vol.512 , pp. 291-297
    • Miranda, C.J.1
  • 42
    • 0018238065 scopus 로고
    • Incidence, prevalence, and gene frequency studies of chronic spinal muscular atrophy
    • Pearn, J. Incidence, prevalence, and gene frequency studies of chronic spinal muscular atrophy. J. Med. Genet. 15, 409-413 (1978).
    • (1978) J. Med. Genet. , vol.15 , pp. 409-413
    • Pearn, J.1
  • 43
    • 0033983258 scopus 로고    scopus 로고
    • An exonic enhancer is required for inclusion of an essential exon in the SMA-determining gene SMN
    • Lorson, C. L. & Androphy, E. J. An exonic enhancer is required for inclusion of an essential exon in the SMA-determining gene SMN. Hum. Mol. Genet. 9, 259-265 (2000).
    • (2000) Hum. Mol. Genet. , vol.9 , pp. 259-265
    • Lorson, C.L.1    Androphy, E.J.2
  • 45
    • 0029954338 scopus 로고    scopus 로고
    • A novel nuclear structure containing the survival motor neurons protein
    • Liu, O. & Dreyfuss, G, A novel nuclear structure containing the survival motor neurons protein. EMBO J. 15, 3555-3564, (1996).
    • (1996) EMBO J. , vol.15 , pp. 3555-3564
    • Liu, O.1    Dreyfuss, G.2
  • 46
    • 0012624586 scopus 로고    scopus 로고
    • Inactivation of the survival motor neuron gene, a candidate gene for human spinal muscular atrophy, leads to massive cell death in early mouse embryos
    • Schrank, B. et al. Inactivation of the survival motor neuron gene, a candidate gene for human spinal muscular atrophy, leads to massive cell death in early mouse embryos Proc. Natl Acad. Sci. USA 96, 6307-6311 (1997).
    • (1997) Proc. Natl. Acad. Sci. USA , vol.96 , pp. 6307-6311
    • Schrank, B.1
  • 47
    • 0033987669 scopus 로고    scopus 로고
    • A mouse model for spinal muscular atrophy
    • Hsieh-Li, H. M. et al. A mouse model for spinal muscular atrophy, Nature Genet. 24, 66-70 (2000).
    • (2000) Nature Genet. , vol.24 , pp. 66-70
    • Hsieh-Li, H.M.1
  • 48
    • 0034639645 scopus 로고    scopus 로고
    • -/- mice and results in a mouse with spinal muscular atrophy
    • -/- mice and results in a mouse with spinal muscular atrophy Hum Mol. Genet. 9, 331-339 (2000). References 47 and 48 report the creation of transgenic mice carrying the entire human SMN2 gene on the Smn-null background. These approaches overcame the embryonic lethality seen in Smn-null mice, providing accurate mouse models for SMA and confirming that the disease severity is dependent on the copy number of SMN2 or levels of SMN protein.
    • (2000) Hum. Mol. Genet. , vol.9 , pp. 331-339
    • Monani, U.R.1
  • 49
    • 0014979663 scopus 로고
    • Histological and histochemical changes from cases of chronic juvenile and early adult muscular spinal atrophy (the Kugelberg-Welander syndrome)
    • Mastaglia, F. L. & Walton, J. N. Histological and histochemical changes from cases of chronic juvenile and early adult muscular spinal atrophy (the Kugelberg-Welander syndrome). J. Neurol. Sci. 12, 15-44 (1971).
    • (1971) J. Neurol. Sci. , vol.12 , pp. 15-44
    • Mastaglia, F.L.1    Walton, J.N.2
  • 50
    • 0034701295 scopus 로고    scopus 로고
    • Nuclear targeting defect of SMN lacking the C-terminus in a mouse model of spinal muscluar atrophy
    • Frugier, T. et al. Nuclear targeting defect of SMN lacking the C-terminus in a mouse model of spinal muscluar atrophy. Hum. Mol. Genet. 9, 849-858 (2000).
    • (2000) Hum. Mol. Genet. , vol.9 , pp. 849-858
    • Frugier, T.1
  • 51
    • 0035809926 scopus 로고    scopus 로고
    • Deletion of munne Smn exon 7 directed to skeletal muscle leads to severe muscular atrophy
    • Cifuentes-Diaz, C. et al. Deletion of munne Smn exon 7 directed to skeletal muscle leads to severe muscular atrophy J. Cell. Biol. 152, 1107-1114 (2001).
    • (2001) J. Cell. Biol. , vol.152 , pp. 1107-1114
    • Cifuentes-Diaz, C.1
  • 52
    • 0035859952 scopus 로고    scopus 로고
    • Treatment of spinal muscular atrophy by sodium butyrate
    • Chang, J.-G. et al. Treatment of spinal muscular atrophy by sodium butyrate. Proc. Natl Acad Sci. USA 98, 9808-9813 (2001). This paper shows that sodium butyrate treatment of SMA-like mice resulted in the increased expression of SMN protein in motor neurons and improvement of the phenotypes, indicating that sodium butyrate might be an effective drug for treating SMA patients.
    • (2001) Proc. Natl. Acad. Sci. USA , vol.98 , pp. 9808-9813
    • Chang, J.-G.1
  • 53
    • 0027958509 scopus 로고
    • Mierophthalmia with linear skin defects (MLS) syndrome: Clinical, cytogenetic, and molecular characterization
    • Lindsay, E. A. et al. Mierophthalmia with linear skin defects (MLS) syndrome: clinical, cytogenetic, and molecular characterization Am. J. Med. Genet. 49, 229-234 (1994).
    • (1994) Am. J. Med. Genet. , vol.49 , pp. 229-234
    • Lindsay, E.A.1
  • 54
    • 0027403249 scopus 로고
    • Incontinentia pigmenti (Bloch-Sulzberger syndrome)
    • Landy, S. J. & Donnai, D Incontinentia pigmenti (Bloch-Sulzberger syndrome). J. Med Genet. 30, 53-59 (1993).
    • (1993) J. Med. Genet. , vol.30 , pp. 53-59
    • Landy, S.J.1    Donnai, D.2
  • 55
    • 0036900297 scopus 로고    scopus 로고
    • Loss of holocytochrome c-type synthetase causes the male lethality of X-linked dominant microphtalmia with linear skin defects (MLS) syndrome
    • Prakash, S. K. et al. Loss of holocytochrome c-type synthetase causes the male lethality of X-linked dominant microphtalmia with linear skin defects (MLS) syndrome. Hum. Mol. Genet. 11, 3237-3248 (2002).
    • (2002) Hum. Mol. Genet. , vol.11 , pp. 3237-3248
    • Prakash, S.K.1
  • 56
    • 0033634663 scopus 로고    scopus 로고
    • Female mice heterozygous for iKKγ/NEMO deficiencies develop a dermatopathy similar to the human X-linked disorder incontinentia pigmenti
    • Makns, C. et al. Female mice heterozygous for iKKγ/NEMO deficiencies develop a dermatopathy similar to the human X-linked disorder incontinentia pigmenti. Mol. Cell 5, 969-979 (2000).
    • (2000) Mol. Cell , vol.5 , pp. 969-979
    • Makns, C.1
  • 57
    • 0036301947 scopus 로고    scopus 로고
    • A decade of molecular studies of fragile X syndrome
    • O'Donnel, W. T. & Warren, S. T. A. decade of molecular studies of fragile X syndrome. Annu. Rev Neurosci. 25, 315-338 (2002).
    • (2002) Annu. Rev. Neurosci. , vol.25 , pp. 315-338
    • O'Donnel, W.T.1    Warren, S.T.2
  • 58
    • 0028246435 scopus 로고
    • Dutch-Belgian Fragile X Consortium. Fmr1 knockout mice: A model to study fragile X mental retardation
    • Dutch-Belgian Fragile X Consortium. Fmr1 knockout mice: a model to study fragile X mental retardation. Cell 78, 23-33 (1994).
    • (1994) Cell , vol.78 , pp. 23-33
  • 59
    • 0034194228 scopus 로고    scopus 로고
    • (Over)correction of FMR1 deficiency with YAC transgenics: Behavioral and physical features
    • Peier, A. M. et al. (Over)correction of FMR1 deficiency with YAC transgenics: behavioral and physical features Hum. Mol. Genet. 9, 1145-1159 (2000). By analysing YAC transgenic mice that overexpress FMR1 protein on a Fmr1-null background, the authors suggested that the levels of FMR1 protein must be regulated to enable gene therapy to be considered for fragile X syndrome.
    • (2000) Hum. Mol. Genet. , vol.9 , pp. 1145-1159
    • Peier, A.M.1
  • 60
  • 61
    • 0021837533 scopus 로고
    • Adult fragile X syndrome: Cliniconeuropathologic findings
    • Rudelli, R. D. et al. Adult fragile X syndrome: cliniconeuropathologic findings. Acta. Neuropathol. 67, 289-295 (1985).
    • (1985) Acta. Neuropathol. , vol.67 , pp. 289-295
    • Rudelli, R.D.1
  • 62
    • 0030986183 scopus 로고    scopus 로고
    • Abnormal dendritic spines in fragile X knockouts mice: Maturation and pruning deficits
    • Comery et al. Abnormal dendritic spines in fragile X knockouts mice: maturation and pruning deficits. Proc. Natl Acad. Sci USA 94, 5401-5404 (1997).
    • (1997) Proc. Natl. Acad. Sci USA , vol.94 , pp. 5401-5404
    • Comery1
  • 63
    • 0035879180 scopus 로고    scopus 로고
    • Abnormal development of dendritic spine in FMR1 knockout mice
    • Nimchinsky, E. A., Oberlander, A. M. & Svoboda, K. Abnormal development of dendritic spine in FMR1 knockout mice J. Neurosci. 21, 5139-5146 (2001).
    • (2001) J. Neurosci. , vol.21 , pp. 5139-5146
    • Nimchinsky, E.A.1    Oberlander, A.M.2    Svoboda, K.3
  • 64
    • 0033365401 scopus 로고    scopus 로고
    • Rett syndrome and beyond: Recurrent spontaneous and familial MECP2 mutations at CpG hotspots
    • Wan, M. et al. Rett syndrome and beyond: recurrent spontaneous and familial MECP2 mutations at CpG hotspots. Am. J. Hum. Genet. 65, 1520-1529 (1999).
    • (1999) Am. J. Hum. Genet. , vol.65 , pp. 1520-1529
    • Wan, M.1
  • 65
    • 0035093830 scopus 로고    scopus 로고
    • Deficiency of methyl-CpG binding protein-2 in CNS neurons results in a Rett-fike phenotype in mice
    • Chen, R. Z. et al. Deficiency of methyl-CpG binding protein-2 in CNS neurons results in a Rett-fike phenotype in mice. Nature Genet. 27, 327-331 (2001).
    • (2001) Nature Genet. , vol.27 , pp. 327-331
    • Chen, R.Z.1
  • 66
    • 0035094767 scopus 로고    scopus 로고
    • A mouse Mecp2-null mutation causes neurological symptoms that mimic Rett syndrome
    • Guy, J. et al. A mouse Mecp2-null mutation causes neurological symptoms that mimic Rett syndrome. Nature Genet. 27, 322-326 (2001). References 65 and 66 indicate that total Mecp2 deficiency in male mice models the more severe neonatal encephalopathy seen in humans. Importantly, these papers show that the phenotype is a result of the loss of Mecp2 function in neurons.
    • (2001) Nature Genet. , vol.27 , pp. 322-326
    • Guy, J.1
  • 67
    • 0037130455 scopus 로고    scopus 로고
    • Mice with truncated MeCP2 recapitulate many Rett syndrome features and display hyperecetylation of histone H3
    • Shahbazian, M. D. et al. Mice with truncated MeCP2 recapitulate many Rett syndrome features and display hyperecetylation of histone H3. Neuron 35, 243-254 (2002). The authors generate a hypomorphic allele of Mecp2 to avoid lethality and establish a Rett phenotype in male mice, avoiding the confounding effect of XCI.
    • (2002) Neuron , vol.35 , pp. 243-254
    • Shahbazian, M.D.1
  • 68
    • 0032076307 scopus 로고    scopus 로고
    • Imprinting in Prader-Willi and Angelman syndrome
    • Nicholls, R. D., Saitoh, S. & Horsthemke, B. Imprinting in Prader-Willi and Angelman syndrome. Trends Genet, 14, 194-200 (1998).
    • (1998) Trends Genet. , vol.14 , pp. 194-200
    • Nicholls, R.D.1    Saitoh, S.2    Horsthemke, B.3
  • 69
    • 0029348110 scopus 로고
    • Angelman syndrome
    • Williams, C. A. et al. Angelman syndrome, Curr. Prob. Pediatr. 25, 216-231 (1995).
    • (1995) Curr. Prob. Pediatr. , vol.25 , pp. 216-231
    • Williams, C.A.1
  • 70
    • 0032192481 scopus 로고    scopus 로고
    • Mutation of the Angelman ubiquitin ligase in mice causes increased cytoplasmic p53 and deficits of contextual learning and long-term potentiation
    • Jiang, Y. et al. Mutation of the Angelman ubiquitin ligase in mice causes increased cytoplasmic p53 and deficits of contextual learning and long-term potentiation. Neuron 21, 799-811 (1998).
    • (1998) Neuron , vol.21 , pp. 799-811
    • Jiang, Y.1
  • 71
    • 0036197031 scopus 로고    scopus 로고
    • Neurobehavioral and electroencephalographic abnormalities in Ube3a maternal-deficient mice
    • Miura, K. et al. Neurobehavioral and electroencephalographic abnormalities in Ube3a maternal-deficient mice, Neurobiol. Dis. 9, 149-159 (2002).
    • (2002) Neurobiol. Dis. , vol.9 , pp. 149-159
    • Miura, K.1
  • 72
    • 0027176708 scopus 로고
    • Isolation of a Miller-Dieker lissencephaly gene containing G protein β-subunit-like repeats
    • Reiner, O. et al. Isolation of a Miller-Dieker lissencephaly gene containing G protein β-subunit-like repeats. Nature 364, 717-721 (1993).
    • (1993) Nature , vol.364 , pp. 717-721
    • Reiner, O.1
  • 73
    • 0031848149 scopus 로고    scopus 로고
    • Graded reduction of Pafah1b1 (Lis1) activity results in neuronal migration defects and early embryonic lethality
    • Hirotsune, S. et al. Graded reduction of Pafah1b1 (Lis1) activity results in neuronal migration defects and early embryonic lethality. Nature Genet. 19, 333-339 (1998). This work clearly indicates that Lis1 controls neuronal migration throughout the mouse brain in a dosage-dependent manner, by taking advantage of both null and hypomorphic alleles of the gene.
    • (1998) Nature Genet. , vol.19 , pp. 333-339
    • Hirotsune, S.1
  • 74
    • 0032536030 scopus 로고    scopus 로고
    • A potassium channel mutation in neonatal human epilepsy
    • Biervert, C. et al. A potassium channel mutation in neonatal human epilepsy. Science 279, 403-436 (1998).
    • (1998) Science , vol.279 , pp. 403-436
    • Biervert, C.1
  • 75
    • 17344372328 scopus 로고    scopus 로고
    • A novel potassium channel gene, KCNQ2, is mutated in an inherited epilepsy of newborns
    • Singh, N. A. et al. A novel potassium channel gene, KCNQ2, is mutated in an inherited epilepsy of newborns. Nature Genet. 18, 25-29 (1998).
    • (1998) Nature Genet. , vol.18 , pp. 25-29
    • Singh, N.A.1
  • 76
    • 0031974209 scopus 로고    scopus 로고
    • Pore mutation in a novel KQT-like potassium channel gene in an idiopathic epilepsy family
    • Chalier, C. et al. pore mutation in a novel KQT-like potassium channel gene in an idiopathic epilepsy family. Nature Genet. 18, 53-55 (1998).
    • (1998) Nature Genet. , vol.18 , pp. 53-55
    • Chalier, C.1
  • 77
    • 0028980028 scopus 로고
    • A missense mutation in the neuronal nicotinic acetylcholine receptor α4 subunit is associated with autosomal dominant nocturnal frontal lobe epilepsy
    • Steinlein, O. K. et al. A missense mutation in the neuronal nicotinic acetylcholine receptor α4 subunit is associated with autosomal dominant nocturnal frontal lobe epilepsy. Nature Genet, 11, 201-203 (1995).
    • (1995) Nature Genet. , vol.11 , pp. 201-203
    • Steinlein, O.K.1
  • 78
    • 0033763090 scopus 로고    scopus 로고
    • The nicotinic receptor β2 subunit is mutant in nocturnal frontal lobe epilepsy
    • De Fusco, M. et al. The nicotinic receptor β2 subunit is mutant in nocturnal frontal lobe epilepsy Nature Genet. 26, 276-276 (2000).
    • (2000) Nature Genet. , vol.26 , pp. 276
    • De Fusco, M.1
  • 81
    • 0035163074 scopus 로고    scopus 로고
    • CHRNB2 is the second acetylcholine receptor subunit associated with autosomal dominant nocturnal frontal lobe epilepsy
    • Phillips, H. A. et al. CHRNB2 is the second acetylcholine receptor subunit associated with autosomal dominant nocturnal frontal lobe epilepsy. Am. J. Hum. Genet. 68, 226-231 (2001).
    • (2001) Am. J. Hum. Genet. , vol.68 , pp. 226-231
    • Phillips, H.A.1
  • 82
    • 0034214261 scopus 로고    scopus 로고
    • Nicotinic receptor function: New perspectives from knockout mice
    • Cordero-Erauquin, M. et al. Nicotinic receptor function: new perspectives from knockout mice. Trends Pharmacol. Sci. 21, 211-217 (2000).
    • (2000) Trends Pharmacol. Sci. , vol.21 , pp. 211-217
    • Cordero-Erauquin, M.1
  • 83
    • 0035030766 scopus 로고    scopus 로고
    • A receptor dysfunction in epilepsy: A mutation in the γ2-subunit gene
    • A receptor dysfunction in epilepsy: a mutation in the γ2-subunit gene. Nature Genet. 28, 46-48 (2001).
    • (2001) Nature Genet. , vol.28 , pp. 46-48
    • Baulac, S.1
  • 84
    • 0035033520 scopus 로고    scopus 로고
    • A receptor γ-subunit in childhood absence epilepsy and febrile seizures
    • A receptor γ-subunit in childhood absence epilepsy and febrile seizures. Nature Genet. 28, 49-52 (2001).
    • (2001) Nature Genet. , vol.28 , pp. 49-52
    • Wallace, R.H.1
  • 85
    • 0029097465 scopus 로고
    • Benzodiazepine-insensitive mice generated by targeted disruption of the γ2 subunit of γ-aminobutyric acid type A receptors
    • Günther, U. et al. Benzodiazepine-insensitive mice generated by targeted disruption of the γ2 subunit of γ-aminobutyric acid type A receptors. Proc. Natl. Acad. Sci. USA 92, 7795-7753 (1995).
    • (1995) Proc. Natl. Acad. Sci. USA , vol.92 , pp. 7795-7753
    • Günther, U.1
  • 86
    • 0032968683 scopus 로고    scopus 로고
    • Voltage-dependent calcium channel mutations in neurological disease
    • Burgess, D. L. & Noebels, J. Voltage-dependent calcium channel mutations in neurological disease. Ann. NY Acad. Sci. 868, 199-212 (1999).
    • (1999) Ann. NY Acad. Sci. , vol.868 , pp. 199-212
    • Burgess, D.L.1    Noebels, J.2
  • 87
    • 0033593005 scopus 로고    scopus 로고
    • 2+ channel currents, altered synaptic transmission, and progressive ataxia in mice lacking α1A subunit
    • 2+ channel currents, altered synaptic transmission, and progressive ataxia in mice lacking α1A subunit Proc, Natl Acad. Sci. USA 96, 15246-15250 (1999).
    • (1999) Proc, Natl. Acad. Sci. USA , vol.96 , pp. 15246-15250
    • Jun, K.1
  • 88
    • 0035828406 scopus 로고    scopus 로고
    • Human epilepsy associated with dysfunction of the brain P/Q type calcium channel
    • Jouvenceau, A. et al. Human epilepsy associated with dysfunction of the brain P/Q type calcium channel. Lancet 358, 801-807 (2001).
    • (2001) Lancet , vol.358 , pp. 801-807
    • Jouvenceau, A.1
  • 89
    • 0033910736 scopus 로고    scopus 로고
    • Coding and noncoding variation of the human calcium-channel β4-subunit gene CACNB4 in patients with idiopathic generalized epilepsy and episodic ataxia
    • Escayg, A. et al. Coding and noncoding variation of the human calcium-channel β4-subunit gene CACNB4 in patients with idiopathic generalized epilepsy and episodic ataxia. Am J. Hum Genet. 66, 1531-1539 (2000).
    • (2000) Am. J. Hum. Genet. , vol.66 , pp. 1531-1539
    • Escayg, A.1
  • 90
    • 0035464960 scopus 로고    scopus 로고
    • Monogenic causes of X-linked mental retardation
    • Chelly, J. & Mandel, J.-L. Monogenic causes of X-linked mental retardation. Nature Rev. Genet 2, 669-680 (2001).
    • (2001) Nature Rev. Genet. , vol.2 , pp. 669-680
    • Chelly, J.1    Mandel, J.-L.2
  • 91
    • 0036798191 scopus 로고    scopus 로고
    • Deletion of the mental retardation gene Gdi1 impairs associative memory and alters social behavior in mice
    • D'Adamo, P. et al. Deletion of the mental retardation gene Gdi1 impairs associative memory and alters social behavior in mice. Hum. Mol. Genet 11, 2567-2580 (2002).
    • (2002) Hum. Mol. Genet. , vol.11 , pp. 2567-2580
    • D'Adamo, P.1
  • 92
    • 0036549187 scopus 로고    scopus 로고
    • Impaired conditioned fear and enhanced long-term potentiation in Fmr2 knock-out mice
    • Gu, Y. et al. Impaired conditioned fear and enhanced long-term potentiation in Fmr2 knock-out mice. J. Neurosci, 22, 2753-2763 (2002).
    • (2002) J. Neurosci. , vol.22 , pp. 2753-2763
    • Gu, Y.1
  • 94
    • 0023150598 scopus 로고
    • HPRT-deficient (Lesch-Nyhan) mouse embryos derived from germline colonization by cultured cells
    • Hooper, M., Hardy, K., Handyside, A, Hunter, S. & Monk, M. HPRT-deficient (Lesch-Nyhan) mouse embryos derived from germline colonization by cultured cells. Nature 326, 292-295 (1987).
    • (1987) Nature , vol.326 , pp. 292-295
    • Hooper, M.1    Hardy, K.2    Handyside, A.3    Hunter, S.4    Monk, M.5
  • 95
    • 0023090920 scopus 로고
    • A potential animal model for Lesch-Nyhan syndrome through introduction of HPRT mutations in mice
    • Kuehn, M. R., Bradley, A, Robertson, E. J. & Evans, M. J. A potential animal model for Lesch-Nyhan syndrome through introduction of HPRT mutations in mice. Nature 326, 295-298 (1987).
    • (1987) Nature , vol.326 , pp. 295-298
    • Kuehn, M.R.1    Bradley, A.2    Robertson, E.J.3    Evans, M.J.4
  • 96
    • 0028299278 scopus 로고
    • Characterization of benzodiazepine-sensitive behaviors in the A/J and C57BL/6J inbred strains of mice
    • Mathis, C., Paul, S. N. & Crawley, J. N. Characterization of benzodiazepine-sensitive behaviors in the A/J and C57BL/6J inbred strains of mice. Behav. Genet. 2, 171-180 (1994).
    • (1994) Behav. Genet. , vol.2 , pp. 171-180
    • Mathis, C.1    Paul, S.N.2    Crawley, J.N.3
  • 97
    • 0029940557 scopus 로고    scopus 로고
    • Gene-targeting studies of the mammalian behavior: Is it the mutation or the background phenotype?
    • Gerlai, R. Gene-targeting studies of the mammalian behavior: is it the mutation or the background phenotype? Trends Neurosci. 19, 177-181 (1996).
    • (1996) Trends Neurosci. , vol.19 , pp. 177-181
    • Gerlai, R.1
  • 98
    • 0031714678 scopus 로고    scopus 로고
    • Contextual learning and cue association in fear conditioning in mice: A strain comparison and lesion study
    • Gerlai, R. Contextual learning and cue association in fear conditioning in mice: a strain comparison and lesion study. Behav. Brain Res. 95, 191-203 (1998).
    • (1998) Behav. Brain Res. , vol.95 , pp. 191-203
    • Gerlai, R.1
  • 99
    • 0025959353 scopus 로고
    • FVB/N: An inbred mouse strain preferable for transgenic analysis
    • Taketo, M. et al. FVB/N: an inbred mouse strain preferable for transgenic analysis. Proc. Natl Acad. Sci USA 88, 2065-2069 (1991).
    • (1991) Proc. Natl. Acad. Sci. USA , vol.88 , pp. 2065-2069
    • Taketo, M.1
  • 100
    • 0036148883 scopus 로고    scopus 로고
    • Behavioral and neuroanatomical characterization of FVB/N inbred mice
    • Mineur, Y. S. & Crusio, W. E. Behavioral and neuroanatomical characterization of FVB/N inbred mice. Brain Res. Bull 57, 41-47 (2002).
    • (2002) Brain Res. Bull. , vol.57 , pp. 41-47
    • Mineur, Y.S.1    Crusio, W.E.2
  • 101
    • 9844255568 scopus 로고    scopus 로고
    • Genetic modification of the phenotypes produced by amyloid precursor protein overexpression in transgenic mice
    • Carlson, G. A. et al. Genetic modification of the phenotypes produced by amyloid precursor protein overexpression in transgenic mice. Hum. Mol. Genet. 6, 1951-1959 (1997).
    • (1997) Hum. Mol. Genet. , vol.6 , pp. 1951-1959
    • Carlson, G.A.1
  • 102
    • 0034537966 scopus 로고    scopus 로고
    • Genetic mapping of a mouse modifier gene that can prevent ALS onset
    • Kunst, C. B., Messer, L., Gordon, J., Haines, J. & Patterson, D. Genetic mapping of a mouse modifier gene that can prevent ALS onset. Genomics, 70, 181-189 (2000).
    • (2000) Genomics , vol.70 , pp. 181-189
    • Kunst, C.B.1    Messer, L.2    Gordon, J.3    Haines, J.4    Patterson, D.5
  • 103
    • 15844393658 scopus 로고    scopus 로고
    • Motor neurons in Cu/Zn superoxide dismutase deficient mice develop normally but exhibit enhanced cell death after axonal injury
    • Reaume, A. G, et al. Motor neurons in Cu/Zn superoxide dismutase deficient mice develop normally but exhibit enhanced cell death after axonal injury. Nature Genet. 13, 43-47 (1996).
    • (1996) Nature Genet. , vol.13 , pp. 43-47
    • Reaume, A.G.1
  • 104
    • 0028284779 scopus 로고
    • Motor neuron degeneration in mice that express a human Cu, Zn superoxide dismutase mutation
    • Gurney, M. E. et al. Motor neuron degeneration in mice that express a human Cu, Zn superoxide dismutase mutation. Science 264, 1772-1775 (1994).
    • (1994) Science , vol.264 , pp. 1772-1775
    • Gurney, M.E.1
  • 105
    • 0029053881 scopus 로고
    • An adverse property of familial ALS linked SOD1 mutation causes motor neuron disease characterized by vacuclar degeneration of mitochondria
    • Wong, P. C. et al. An adverse property of familial ALS linked SOD1 mutation causes motor neuron disease characterized by vacuclar degeneration of mitochondria. Neuron 14, 1105-1116 (1995).
    • (1995) Neuron , vol.14 , pp. 1105-1116
    • Wong, P.C.1
  • 106
    • 0028888945 scopus 로고
    • Transgenic mice expressing superoxide dismutase gene provide an animal model of amyotrephic lateral sclerosis
    • Ripps, M. E., Huntley, G. W., Hof, P. R., Mornson, J. H. & Gordon, J. W. Transgenic mice expressing superoxide dismutase gene provide an animal model of amyotrephic lateral sclerosis. Proc. Natl Acad Sci. USA, 92, 689-693 (1995).
    • (1995) Proc. Natl. Acad Sci. USA , vol.92 , pp. 689-693
    • Ripps, M.E.1    Huntley, G.W.2    Hof, P.R.3    Mornson, J.H.4    Gordon, J.W.5
  • 107
    • 0031051485 scopus 로고    scopus 로고
    • ALS linked SOD1 mutant G85R mediates damage to astrocyte and promotes rapidly progressive disease with SOD1 containing inclusions
    • Bruljn, L. I. et al. ALS linked SOD1 mutant G85R mediates damage to astrocyte and promotes rapidly progressive disease with SOD1 containing inclusions. Neuron 18, 327-338 (1997).
    • (1997) Neuron , vol.18 , pp. 327-338
    • Bruljn, L.I.1
  • 108
    • 0037014426 scopus 로고    scopus 로고
    • Protein misfolding, amyloid formation, and neurodegeneration: A critical role for molecular chaperones?
    • Muchowski, P. J. Protein misfolding, amyloid formation, and neurodegeneration: a critical role for molecular chaperones? Neuron 35, 9-12 (2002).
    • (2002) Neuron , vol.35 , pp. 9-12
    • Muchowski, P.J.1
  • 109
    • 0033946710 scopus 로고    scopus 로고
    • Prions, peptides and protein misfolding
    • Cohen, F. E. Prions, peptides and protein misfolding. Mol. Med. Today 6, 292-293 (2000).
    • (2000) Mol. Med. Today , vol.6 , pp. 292-293
    • Cohen, F.E.1
  • 110
    • 18544410106 scopus 로고    scopus 로고
    • Formation of neuronal intranuclear inclusions underlies the neurological dysfunction in mice transgenic for the HD mutation
    • Davies, S. W. et al. Formation of neuronal intranuclear inclusions underlies the neurological dysfunction in mice transgenic for the HD mutation. Cell 90, 537-548 (1997).
    • (1997) Cell , vol.90 , pp. 537-548
    • Davies, S.W.1
  • 111
    • 12644284502 scopus 로고    scopus 로고
    • α-Galactosidase A deficient mice: A model of Fabry disease
    • Oshima, T. et al. α-galactosidase A deficient mice: a model of Fabry disease. Proc. Natl Acad. Sci. USA 94, 2540-2544 (1997).
    • (1997) Proc. Natl. Acad. Sci. USA , vol.94 , pp. 2540-2544
    • Oshima, T.1
  • 112
    • 0030689779 scopus 로고    scopus 로고
    • Targeted inactivation of the X-linked adrenoleukodystrophy gene in mice
    • Forss-Petter, S. et al. Targeted inactivation of the X-linked adrenoleukodystrophy gene in mice. J Neurosci. Res. 50, 829-843 (1997).
    • (1997) J. Neurosci. Res. , vol.50 , pp. 829-843
    • Forss-Petter, S.1
  • 113
    • 0035150547 scopus 로고    scopus 로고
    • Gene therapy for Fabry disease
    • Siatskas, C. & Medin, J. A. Gene therapy for Fabry disease. J Inherit. Metab. Dis. 24 (Suppl.) 25-41 (2001).
    • (2001) J. Inherit. Metab. Dis. , vol.24 , Issue.SUPPL. , pp. 25-41
    • Siatskas, C.1    Medin, J.A.2
  • 114
    • 0031730433 scopus 로고    scopus 로고
    • Gene redundancy and pharmacological gene therapy: Implications for X-linked adrenoleukodystrophy
    • Kemp, S. et. al. Gene redundancy and pharmacological gene therapy: implications for X-linked adrenoleukodystrophy, Nature Med, 4, 1261-1268 (1998).
    • (1998) Nature Med. , vol.4 , pp. 1261-1268
    • Kemp, S.1
  • 115
    • 0032924991 scopus 로고    scopus 로고
    • Adrenoleukodystrophy-related protein can compensate functionally for adrenoleukodystrophy protein deficiency (X-ALD): Implications for therapy
    • Netik, A. et al. Adrenoleukodystrophy-related protein can compensate functionally for adrenoleukodystrophy protein deficiency (X-ALD): implications for therapy. Hum. Mol. Genet. 8, 907-913 (1999).
    • (1999) Hum. Mol. Genet. , vol.8 , pp. 907-913
    • Netik, A.1
  • 116
    • 0037015889 scopus 로고    scopus 로고
    • Tools for targeted manipulation of the mouse genome
    • van der Weyden, L., Adams, D. J. & Bradley, A. Tools for targeted manipulation of the mouse genome. Physiol. Genomics, 11, 133-164 (2002). This review covers recent technological advances in mouse genomic engineering.
    • (2002) Physiol. Genomics , vol.11 , pp. 133-164
    • Van der Weyden, L.1    Adams, D.J.2    Bradley, A.3
  • 117
    • 0035491607 scopus 로고    scopus 로고
    • Conditional control of gene expression in mouse
    • Lewandoski, M Conditional control of gene expression in mouse, Nature Rev. Genet. 2, 743-755 (2001).
    • (2001) Nature Rev. Genet. , vol.2 , pp. 743-755
    • Lewandoski, M.1
  • 118
    • 0034766425 scopus 로고    scopus 로고
    • Application of magnetic resonance to animal models of cerebral ischemia
    • Hoehn, M. et al. Application of magnetic resonance to animal models of cerebral ischemia, J. Magn, Reson. Imaging 14, 491-509 (2001).
    • (2001) J. Magn. Reson. Imaging , vol.14 , pp. 491-509
    • Hoehn, M.1
  • 119
    • 0037022288 scopus 로고    scopus 로고
    • β-Amyloid precursor transgenic mice that harbor diffuse Aβ deposits but do not form plaques show increased ischemic vulnerability: Role of inflammation
    • Koistinaho, M et al. β-amyloid precursor transgenic mice that harbor diffuse Aβ deposits but do not form plaques show increased ischemic vulnerability: role of inflammation, Proc. Natl Acad. Sci. USA 99, 1610-1615 (2002).
    • (2002) Proc. Natl. Acad. Sci. USA , vol.99 , pp. 1610-1615
    • Koistinaho, M.1
  • 120
    • 0037104754 scopus 로고    scopus 로고
    • Compromised hemodynamic response in amyloid precursor protein transgenic mice
    • Mueggler, T. et al. Compromised hemodynamic response in amyloid precursor protein transgenic mice. J. Neurosci 22, 7218-7224(2002).
    • (2002) J. Neurosci. , vol.22 , pp. 7218-7224
    • Mueggler, T.1
  • 121
    • 0034517627 scopus 로고    scopus 로고
    • Imaging physiologic dysfunction of individual hippocampal subregions in humans and genetically modified mice
    • Small, S. A. et al. Imaging physiologic dysfunction of individual hippocampal subregions in humans and genetically modified mice. Neuron 28, 653-664 (2000).
    • (2000) Neuron , vol.28 , pp. 653-664
    • Small, S.A.1
  • 122
    • 0034691124 scopus 로고    scopus 로고
    • In vivo detection of amyloid plaques in a mouse model of Alzheimer's disease
    • Skovronsky, D. M. et al. In vivo detection of amyloid plaques in a mouse model of Alzheimer's disease. Proc. Natl Acad. Sci. USA 97, 7609-7614 (2000).
    • (2000) Proc. Natl. Acad. Sci. USA , vol.97 , pp. 7609-7614
    • Skovronsky, D.M.1
  • 124
    • 0035106780 scopus 로고    scopus 로고
    • Imaging of amyloid-β deposits in brains of living mice permits direct observation of clearance of plaques with immunotherapy
    • Bacskai, B. J. et al. Imaging of amyloid-β deposits in brains of living mice permits direct observation of clearance of plaques with immunotherapy. Nature Med. 7, 369-372 (2001).
    • (2001) Nature Med. , vol.7 , pp. 369-372
    • Bacskai, B.J.1
  • 125
    • 0036965918 scopus 로고    scopus 로고
    • Molecular targeting of Alzheimer's amyloid plaques for contrast-enhanced magnetic resonance imaging
    • Poduslo, J. F. et al. Molecular targeting of Alzheimer's amyloid plaques for contrast-enhanced magnetic resonance imaging. Neurobiol. Dis. 11, 315-329 (2002).
    • (2002) Neurobiol. Dis. , vol.11 , pp. 315-329
    • Poduslo, J.F.1
  • 126
    • 0034660457 scopus 로고    scopus 로고
    • Neuroprotective effects of creatine in a transgenic mouse model of Huntington's disease
    • Ferrante, R. J. et al. Neuroprotective effects of creatine in a transgenic mouse model of Huntington's disease. J. Neurosci. 20, 4389-4397 (2000).
    • (2000) J. Neurosci. , vol.20 , pp. 4389-4397
    • Ferrante, R.J.1
  • 127
    • 0034743672 scopus 로고    scopus 로고
    • Creatine increases survival and delays motor symptoms in a transgenic animal model of Huntington's disease (2001)
    • Andreassen, O. A. et al. Creatine increases survival and delays motor symptoms in a transgenic animal model of Huntington's disease (2001). Neurobiol. Dis. 8, 479-491 (2001).
    • (2001) Neurobiol. Dis. , vol.8 , pp. 479-491
    • Andreassen, O.A.1
  • 128
    • 0036523110 scopus 로고    scopus 로고
    • Therapeutic effects of coenzyme Q10 and remacemide in transgenic mouse models of Huntington's disease
    • Ferrante, R. J. et al. Therapeutic effects of coenzyme Q10 and remacemide in transgenic mouse models of Huntington's disease. J. Neurosci. 22 1592-1599 (2002).
    • (2002) J. Neurosci. , vol.22 , pp. 1592-1599
    • Ferrante, R.J.1
  • 129
    • 0033587128 scopus 로고    scopus 로고
    • Inhibition of caspase-1 slows disease progression in a mouse model of Huntington's disease
    • Ona, V. O. et al. Inhibition of caspase-1 slows disease progression in a mouse model of Huntington's disease. Nature 399, 263-267 (1999).
    • (1999) Nature , vol.399 , pp. 263-267
    • Ona, V.O.1
  • 130
    • 0034955984 scopus 로고    scopus 로고
    • Dichloroacetate exerts therapeutic effects in transgenic mouse models of Huntington's disease
    • Andreassen O. A. et al. Dichloroacetate exerts therapeutic effects in transgenic mouse models of Huntington's disease. Ann. Neurol 50, 112-116 (2001).
    • (2001) Ann. Neurol. , vol.50 , pp. 112-116
    • Andreassen, O.A.1
  • 131
    • 0037109665 scopus 로고    scopus 로고
    • Therapeutic effects of cystamine in a murine model of Huntington's disease
    • Dedeoglu, A. et al. Therapeutic effects of cystamine in a murine model of Huntington's disease. J, Neurosci, 22, 8942-8950 (2002).
    • (2002) J. Neurosci. , vol.22 , pp. 8942-8950
    • Dedeoglu, A.1
  • 132
    • 0033912716 scopus 로고    scopus 로고
    • Minocycline inhibits caspase-1 and caspase-3 expression and delays mortality in a transgenic mouse model of Huntington disease
    • Chen M. et aL Minocycline inhibits caspase-1 and caspase-3 expression and delays mortality in a transgenic mouse model of Huntington disease. Nature Med. 6, 797-801 (2000).
    • (2000) Nature Med. , vol.6 , pp. 797-801
    • Chen, M.1
  • 133
    • 85009226418 scopus 로고    scopus 로고
    • A randomized, placebo-controlled trial of coenzyme Q10 and remacemide in Huntington's disease
    • Huntington Study Group. A randomized, placebo-controlled trial of coenzyme Q10 and remacemide in Huntington's disease Neurology 57, 397-404 (2001).
    • (2001) Neurology , vol.57 , pp. 397-404
  • 134
    • 0031171578 scopus 로고    scopus 로고
    • A proposed test battery and constellations of specific behavioral paradigms to investigate the behavioral phenotypes of traragenic and knockout mice
    • Crawley, J. N. & Paylor, R. E. A proposed test battery and constellations of specific behavioral paradigms to investigate the behavioral phenotypes of traragenic and knockout mice. Horm. Behav. 31, 197-211 (1997).
    • (1997) Horm. Behav. , vol.31 , pp. 197-211
    • Crawley, J.N.1    Paylor, R.E.2
  • 135
    • 0034639931 scopus 로고    scopus 로고
    • Towards new models of disease and physiology in the neurosciences: The role of induced and naturally occurring mutations
    • Hunter, A. J. Nolan, P. M. & Brown, S. D. M. Towards new models of disease and physiology in the neurosciences: the role of induced and naturally occurring mutations. Hum. Mol. Genet. 9, 893-900 (2000).
    • (2000) Hum. Mol. Genet. , vol.9 , pp. 893-900
    • Hunter, A.J.1    Nolan, P.M.2    Brown, S.D.M.3
  • 136
    • 0034068758 scopus 로고    scopus 로고
    • List of transgenic and knockout mice: Behavioral profiles
    • Bolivar, V., Cook, M. & Flaherty, L. List of transgenic and knockout mice: behavioral profiles. Mamm. Genome 11, 260-274 (2000).
    • (2000) Mamm. Genome , vol.11 , pp. 260-274
    • Bolivar, V.1    Cook, M.2    Flaherty, L.3
  • 137
    • 0034910512 scopus 로고    scopus 로고
    • Long-term consequences of developmental exposure to aluminium in a suboptimal diet for growth and behavior of Swiss Webster mice
    • Golub, M. S. & Germann, S. L Long-term consequences of developmental exposure to aluminium in a suboptimal diet for growth and behavior of Swiss Webster mice. Neurotoxicol. Teratol. 23, 365-372 (2001).
    • (2001) Neurotoxicol. Teratol. , vol.23 , pp. 365-372
    • Golub, M.S.1    Germann, S.L.2
  • 138
    • 0030864463 scopus 로고    scopus 로고
    • Purkinje cell expression of a mutant allele of SCA 1 in transgenic mice leads to disparate effects on motor behaviors, followed by a progressive cerebellar dysfunction and histological alterations
    • Clark, H. B. et al. Purkinje cell expression of a mutant allele of SCA 1 in transgenic mice leads to disparate effects on motor behaviors, followed by a progressive cerebellar dysfunction and histological alterations. J. Neurosci. 17, 7385-7395 (1997).
    • (1997) J. Neurosci. , vol.17 , pp. 7385-7395
    • Clark, H.B.1
  • 139
    • 0034234894 scopus 로고    scopus 로고
    • Conditional bialleic Nf2 mutation in the mouse promotes manifestations of human neurofibromatosis type 2
    • Giovannini M. et al. Conditional bialleic Nf2 mutation in the mouse promotes manifestations of human neurofibromatosis type 2. Genes. Dev. 14, 1617-1630 (2000).
    • (2000) Genes. Dev. , vol.14 , pp. 1617-1630
    • Giovannini, M.1
  • 140
    • 0036571383 scopus 로고    scopus 로고
    • Nf2 gene inactivation in arachnoidal cells is rate-limiting for meningioma development in the mouse
    • Kalamarides M. et al. Nf2 gene inactivation in arachnoidal cells is rate-limiting for meningioma development in the mouse. Genes. Dev. 16, 1060-1065 (2002).
    • (2002) Genes. Dev. , vol.16 , pp. 1060-1065
    • Kalamarides, M.1
  • 141
    • 0029113867 scopus 로고
    • Mouse models of Tay-Sachs and Sandhoff diseases differ in neurologic phenotype and ganglloside metabolism
    • Sango, K. et al. Mouse models of Tay-Sachs and Sandhoff diseases differ in neurologic phenotype and ganglloside metabolism. Nature Genet. 11, 170-176 (1995).
    • (1995) Nature Genet. , vol.11 , pp. 170-176
    • Sango, K.1
  • 142
    • 9044236158 scopus 로고    scopus 로고
    • Dramatically different phenotypes in mouse models of human Tay-Sachs and Sandhoff diseases
    • Phaneuf, D. et al. Dramatically different phenotypes in mouse models of human Tay-Sachs and Sandhoff diseases. Hum. Mol. Genet. 5, 1-14 (1996).
    • (1996) Hum. Mol. Genet. , vol.5 , pp. 1-14
    • Phaneuf, D.1
  • 143
    • 0035910076 scopus 로고    scopus 로고
    • Delayed-onset ataxia in mice lacking α-tocopherol transfer protein: Model for neuronal degeneration caused by chronic oxidative stress
    • Yokota, T. et al. Delayed-onset ataxia in mice lacking α-tocopherol transfer protein: model for neuronal degeneration caused by chronic oxidative stress. Proc. Natl Acad. Sci. USA 98, 15185-15190 (2001).
    • (2001) Proc. Natl. Acad. Sci. USA , vol.98 , pp. 15185-15190
    • Yokota, T.1
  • 145
    • 0028133486 scopus 로고
    • Glial cell degeneration and hypomyelination caused by overexpression of myelin proteolipid protein gene
    • Kagawa, T. et al. Glial cell degeneration and hypomyelination caused by overexpression of myelin proteolipid protein gene, Neuron 13, 427-442 (1994).
    • (1994) Neuron , vol.13 , pp. 427-442
    • Kagawa, T.1
  • 146
    • 0031037761 scopus 로고    scopus 로고
    • Assembly of CNS myelin in the absence of proteolipid protein
    • Klugmann, M. et al. Assembly of CNS myelin in the absence of proteolipid protein. Neuron 18, 59-70 (1997).
    • (1997) Neuron , vol.18 , pp. 59-70
    • Klugmann, M.1
  • 147
    • 0032486428 scopus 로고    scopus 로고
    • Axonal swellings and degeneration in mice lacking the major proteolipid of myelin
    • Griffiths, I. et al. Axonal swellings and degeneration in mice lacking the major proteolipid of myelin. Science 280, 1610-1613 (1998).
    • (1998) Science , vol.280 , pp. 1610-1613
    • Griffiths, I.1
  • 148
    • 0035862896 scopus 로고    scopus 로고
    • Neurological abnormalities in a knock-in mouse model of Huntington's disease
    • Lin, C. H. et al. Neurological abnormalities in a knock-in mouse model of Huntington's disease. Hum. Mol. Genet. 15, 137-144, (2001).
    • (2001) Hum. Mol. Genet. , vol.15 , pp. 137-144
    • Lin, C.H.1
  • 149
    • 0037108766 scopus 로고    scopus 로고
    • Generation and characterization of androgen receptor knockout (ARKO) mice: An in vivo model for the study of androgen functions in selective tissues
    • Yeh, S. et al. Generation and characterization of androgen receptor knockout (ARKO) mice: an in vivo model for the study of androgen functions in selective tissues. Proc. Natl Acad. Sci. USA 99, 13498-13503 (2002).
    • (2002) Proc. Natl. Acad. Sci. USA , vol.99 , pp. 13498-13503
    • Yeh, S.1


* 이 정보는 Elsevier사의 SCOPUS DB에서 KISTI가 분석하여 추출한 것입니다.