Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 14 de 14
Filter
1.
Methods Mol Biol ; 2614: 369-381, 2023.
Article in English | MEDLINE | ID: mdl-36587136

ABSTRACT

Our increased understanding of how a cell's microenvironment influences its behavior has fueled an interest in three-dimensional (3D) cell cultures for drug discovery. Particularly, scaffold-based 3D cultures are expected to recapitulate in vivo tissue stiffness and extracellular matrix composition more accurately than standard two-dimensional (2D) monolayer cultures. Here we present a 3D hydrogel cell culture setup suitable for automated screening with standard high-throughput screening (HTS) liquid handling equipment commonly found in a drug discovery laboratory. Further, we describe the steps required to validate the assay system for compound screening.


Subject(s)
Drug Discovery , Hydrogels , Drug Discovery/methods , High-Throughput Screening Assays/methods , Cell Culture Techniques/methods , Extracellular Matrix
2.
Genome Med ; 11(1): 80, 2019 12 09.
Article in English | MEDLINE | ID: mdl-31818324

ABSTRACT

BACKGROUND: We investigated the features of the genomic rearrangements in a cohort of 50 male individuals with proteolipid protein 1 (PLP1) copy number gain events who were ascertained with Pelizaeus-Merzbacher disease (PMD; MIM: 312080). We then compared our new data to previous structural variant mutagenesis studies involving the Xq22 region of the human genome. The aggregate data from 159 sequenced join-points (discontinuous sequences in the reference genome that are joined during the rearrangement process) were studied. Analysis of these data from 150 individuals enabled the spectrum and relative distribution of the underlying genomic mutational signatures to be delineated. METHODS: Genomic rearrangements in PMD individuals with PLP1 copy number gain events were investigated by high-density customized array or clinical chromosomal microarray analysis and breakpoint junction sequence analysis. RESULTS: High-density customized array showed that the majority of cases (33/50; ~ 66%) present with single duplications, although complex genomic rearrangements (CGRs) are also frequent (17/50; ~ 34%). Breakpoint mapping to nucleotide resolution revealed further previously unknown structural and sequence complexities, even in single duplications. Meta-analysis of all studied rearrangements that occur at the PLP1 locus showed that single duplications were found in ~ 54% of individuals and that, among all CGR cases, triplication flanked by duplications is the most frequent CGR array CGH pattern observed. Importantly, in ~ 32% of join-points, there is evidence for a mutational signature of microhomeology (highly similar yet imperfect sequence matches). CONCLUSIONS: These data reveal a high frequency of CGRs at the PLP1 locus and support the assertion that replication-based mechanisms are prominent contributors to the formation of CGRs at Xq22. We propose that microhomeology can facilitate template switching, by stabilizing strand annealing of the primer using W-C base complementarity, and is a mutational signature for replicative repair.


Subject(s)
DNA Copy Number Variations , Gene Rearrangement , Mutation , Myelin Proteolipid Protein/genetics , Chromosome Breakpoints , Comparative Genomic Hybridization , Gene Duplication , Genetic Association Studies , Genetic Predisposition to Disease , Genome, Human , Genomic Instability , Genomics/methods , Humans , Polymorphism, Single Nucleotide
3.
Mol Ther Nucleic Acids ; 12: 420-432, 2018 Sep 07.
Article in English | MEDLINE | ID: mdl-30195779

ABSTRACT

DNA variants of the proteolipid protein 1 gene (PLP1) that shift PLP1/DM20 alternative splicing away from the PLP1 form toward DM20 cause the allelic X-linked leukodystrophies Pelizaeus-Merzbacher disease (PMD), spastic paraplegia 2 (SPG2), and hypomyelination of early myelinating structures (HEMS). We designed a morpholino oligomer (MO-PLP) to block use of the DM20 5' splice donor site, thereby shifting alternative splicing toward the PLP1 5' splice site. Treatment of an immature oligodendrocyte cell line with MO-PLP significantly shifted alternative splicing toward PLP1 expression from the endogenous gene and from transfected human minigene splicing constructs harboring patient variants known to reduce the amount of the PLP1 spliced product. Additionally, a single intracerebroventricular injection of MO-PLP into the brains of neonatal mice, carrying a deletion of an intronic splicing enhancer identified in a PMD patient that reduces the Plp1 spliced form, corrected alternative splicing at both RNA and protein levels in the CNS. The effect lasted to post-natal day 90, well beyond the early post-natal spike in myelination and PLP production. Further, the single injection produced a sustained reduction of inflammatory markers in the brains of the mice. Our results suggest that morpholino oligomers have therapeutic potential for the treatment of PMD, SPG2, and HEMS.

4.
Ann Clin Transl Neurol ; 2(6): 648-61, 2015 Jun.
Article in English | MEDLINE | ID: mdl-26125040

ABSTRACT

OBJECTIVE: The objective of this study was to investigate the genetic etiology of the X-linked disorder "Hypomyelination of Early Myelinating Structures" (HEMS). METHODS: We included 16 patients from 10 families diagnosed with HEMS by brain MRI criteria. Exome sequencing was used to search for causal mutations. In silico analysis of effects of the mutations on splicing and RNA folding was performed. In vitro gene splicing was examined in RNA from patients' fibroblasts and an immortalized immature oligodendrocyte cell line after transfection with mutant minigene splicing constructs. RESULTS: All patients had unusual hemizygous mutations of PLP1 located in exon 3B (one deletion, one missense and two silent), which is spliced out in isoform DM20, or in intron 3 (five mutations). The deletion led to truncation of PLP1, but not DM20. Four mutations were predicted to affect PLP1/DM20 alternative splicing by creating exonic splicing silencer motifs or new splice donor sites or by affecting the local RNA structure of the PLP1 splice donor site. Four deep intronic mutations were predicted to destabilize a long-distance interaction structure in the secondary PLP1 RNA fragment involved in regulating PLP1/DM20 alternative splicing. Splicing studies in fibroblasts and transfected cells confirmed a decreased PLP1/DM20 ratio. INTERPRETATION: Brain structures that normally myelinate early are poorly myelinated in HEMS, while they are the best myelinated structures in Pelizaeus-Merzbacher disease, also caused by PLP1 alterations. Our data extend the phenotypic spectrum of PLP1-related disorders indicating that normal PLP1/DM20 alternative splicing is essential for early myelination and support the need to include intron 3 in diagnostic sequencing.

5.
PLoS Genet ; 11(3): e1005050, 2015 Mar.
Article in English | MEDLINE | ID: mdl-25749076

ABSTRACT

Inverted repeats (IRs) can facilitate structural variation as crucibles of genomic rearrangement. Complex duplication-inverted triplication-duplication (DUP-TRP/INV-DUP) rearrangements that contain breakpoint junctions within IRs have been recently associated with both MECP2 duplication syndrome (MIM#300260) and Pelizaeus-Merzbacher disease (PMD, MIM#312080). We investigated 17 unrelated PMD subjects with copy number gains at the PLP1 locus including triplication and quadruplication of specific genomic intervals-16/17 were found to have a DUP-TRP/INV-DUP rearrangement product. An IR distal to PLP1 facilitates DUP-TRP/INV-DUP formation as well as an inversion structural variation found frequently amongst normal individuals. We show that a homology-or homeology-driven replicative mechanism of DNA repair can apparently mediate template switches within stretches of microhomology. Moreover, we provide evidence that quadruplication and potentially higher order amplification of a genomic interval can occur in a manner consistent with rolling circle amplification as predicted by the microhomology-mediated break induced replication (MMBIR) model.


Subject(s)
Gene Duplication , Myelin Proteolipid Protein/genetics , Pelizaeus-Merzbacher Disease/genetics , Chromosome Breakpoints , Chromosome Inversion , Gene Dosage , Humans
6.
Hum Mol Genet ; 23(20): 5464-78, 2014 Oct 15.
Article in English | MEDLINE | ID: mdl-24890387

ABSTRACT

Alternative splicing of the proteolipid protein 1 gene (PLP1) produces two forms, PLP1 and DM20, due to alternative use of 5' splice sites with the same acceptor site in intron 3. The PLP1 form predominates in central nervous system RNA. Mutations that reduce the ratio of PLP1 to DM20, whether mutant or normal protein is formed, result in the X-linked leukodystrophy Pelizaeus-Merzbacher disease (PMD). We investigated the ability of sequences throughout PLP1 intron 3 to regulate alternative splicing using a splicing minigene construct transfected into the oligodendrocyte cell line, Oli-neu. Our data reveal that the alternative splice of PLP1 is regulated by a long-distance interaction between two highly conserved elements that are separated by 581 bases within the 1071-base intron 3. Further, our data suggest that a base-pairing secondary structure forms between these two elements, and we demonstrate that mutations of either element designed to destabilize the secondary structure decreased the PLP1/DM20 ratio, while swap mutations designed to restore the structure brought the PLP1/DM20 ratio to near normal levels. Sequence analysis of intron 3 in families with clinical symptoms of PMD who did not have coding-region mutations revealed mutations that segregated with disease in three families. We showed that these patient mutations, which potentially destabilize the secondary structure, also reduced the PLP1/DM20 ratio. This is the first report of patient mutations causing disease by disruption of a long-distance intronic interaction controlling alternative splicing. This finding has important implications for molecular diagnostics of PMD.


Subject(s)
Alternative Splicing , Introns , Myelin Proteolipid Protein/genetics , Pelizaeus-Merzbacher Disease/genetics , RNA, Messenger/chemistry , Base Pairing , Cell Line , Female , Humans , Male , Models, Molecular , Mutation , Myelin Proteolipid Protein/metabolism , Nucleic Acid Conformation , Oligodendroglia/metabolism , Pedigree , RNA, Messenger/metabolism , Sequence Analysis, DNA
7.
J Neurosci ; 33(29): 11788-99, 2013 Jul 17.
Article in English | MEDLINE | ID: mdl-23864668

ABSTRACT

Pelizaeus-Merzbacher disease (PMD) is a hypomyelinating leukodystrophy caused by mutations of the proteolipid protein 1 gene (PLP1), which is located on the X chromosome and encodes the most abundant protein of myelin in the central nervous sytem. Approximately 60% of PMD cases result from genomic duplications of a region of the X chromosome that includes the entire PLP1 gene. The duplications are typically in a head-to-tail arrangement, and they vary in size and gene content. Although rodent models with extra copies of Plp1 have been developed, none contains an actual genomic rearrangement that resembles those found in PMD patients. We used mutagenic insertion chromosome engineering resources to generate the Plp1dup mouse model by introducing an X chromosome duplication in the mouse genome that contains Plp1 and five neighboring genes that are also commonly duplicated in PMD patients. The Plp1dup mice display progressive gait abnormalities compared with wild-type littermates. The single duplication leads to increased transcript levels of Plp1 and four of the five other duplicated genes over wild-type levels in the brain beginning the second postnatal week. The Plp1dup mice also display altered transcript levels of other important myelin proteins leading to a progressive degeneration of myelin. Our results show that a single duplication of the Plp1 gene leads to a phenotype similar to the pattern seen in human PMD patients with duplications.


Subject(s)
Demyelinating Diseases/physiopathology , Gait/genetics , Lameness, Animal/physiopathology , Myelin Proteolipid Protein/genetics , Myelin Sheath/pathology , Pelizaeus-Merzbacher Disease/physiopathology , Animals , Demyelinating Diseases/genetics , Demyelinating Diseases/pathology , Disease Models, Animal , Disease Progression , Genotype , Lameness, Animal/genetics , Lameness, Animal/pathology , Mice , Mice, Transgenic , Mutation , Myelin Sheath/genetics , Pelizaeus-Merzbacher Disease/genetics , Pelizaeus-Merzbacher Disease/pathology
8.
Exp Neurol ; 214(2): 322-30, 2008 Dec.
Article in English | MEDLINE | ID: mdl-18835559

ABSTRACT

PLP1 and DM20, major myelin proteins, are generated by developmentally regulated alternative splicing. In the post-natal brain, PLP1 is the predominant product. Deletion of a splicing enhancer in PLP1 intron 3 causes a mild form of Pelizaeus-Merzbacher disease and reduces PLP1 specific splicing in vitro (Hobson, G. M., Huang, Z., Sperle, K., Stabley, D. L., Marks, H. G., and Cambi, F., 2002. A PLP splicing abnormality is associated with an unusual presentation of PMD. Ann. Neurol. 52, 477-488). We sought to investigate the pathogenic role of the mutation and to determine the consequences on the developmental regulation of PLP1 alternative splicing and myelin stability and function in vivo. We have generated a knockin mouse that carries deletion of the intronic splicing enhancer and have characterized the PLP1/DM20 ratio by Real Time RT-PCR and Western blot analysis in the developing and mature brain and examined the clinical and pathological phenotype by motor testing and electron microscopy. The deletion impairs the increase in the PLP1/DM20 transcript and protein ratio at the time of myelination and in adulthood and results in a PLP1 hypomorph. Electron microscopy shows abnormal myelin wraps with fragmented myelin whorls, which are progressive with age, suggesting a defect in myelin stability. Phenotypic characterization of the knockin mouse shows a defect in motor coordination. The data indicate that the intronic splicing enhancer is necessary for the developmental increase in PLP1/DM20 ratio and that full PLP1 dosage is necessary for myelin stability and brain function. This knockin mouse represents a useful model to investigate the mechanisms of disease in human disorders in which PLP1 expression is reduced.


Subject(s)
Myelin Proteolipid Protein/genetics , Myelin Sheath/physiology , Pelizaeus-Merzbacher Disease/genetics , Pelizaeus-Merzbacher Disease/physiopathology , Alternative Splicing , Animals , Behavior, Animal , Brain/growth & development , Brain/pathology , Disease Models, Animal , Enhancer Elements, Genetic , Gene Deletion , Gene Knock-In Techniques , Introns/genetics , Male , Mice , Mice, Inbred C57BL , Mice, Mutant Strains , Microscopy, Electron , Motor Activity , Myelin Proteins/genetics , Myelin Proteins/metabolism , Myelin Proteolipid Protein/metabolism , Myelin Sheath/pathology , Myelin Sheath/ultrastructure , Optic Nerve/growth & development , Optic Nerve/pathology , Optic Nerve/ultrastructure , Pelizaeus-Merzbacher Disease/pathology
9.
Ann Neurol ; 59(2): 398-403, 2006 Feb.
Article in English | MEDLINE | ID: mdl-16374829

ABSTRACT

OBJECTIVE: To report an association between spastic paraplegia type 2 with axonal peripheral neuropathy and apparent proteolipid protein gene (PLP1) silencing in a family. METHODS: Pulsed-field gel electrophoresis, custom array comparative genomic hybridization, and semi-quantitative multiplex polymerase chain reaction analyses were used to examine the PLP1 genomic region. RESULTS: Electrodiagnostic studies and a sural nerve biopsy showed features of a dystrophic axonal neuropathy. Molecular studies identified a small duplication downstream of PLP1. INTERPRETATION: We propose the duplication to result in PLP1 gene silencing by virtue of a position effect. Our observations suggest that genomic rearrangements that do not include PLP1 coding sequences should be considered as yet another potential mutational mechanism underlying PLP1-related dysmyelinating disorders.


Subject(s)
Membrane Proteins/genetics , Mutation , Pelizaeus-Merzbacher Disease/genetics , Peripheral Nervous System Diseases/genetics , DNA Mutational Analysis/methods , Electrophoresis, Polyacrylamide Gel/methods , Humans , MARVEL Domain-Containing Proteins , Magnetic Resonance Imaging/methods , Male , Microscopy, Electron, Transmission/methods , Neural Conduction/physiology , Nucleic Acid Hybridization/methods , Pelizaeus-Merzbacher Disease/complications , Pelizaeus-Merzbacher Disease/pathology , Peripheral Nervous System Diseases/complications , Peripheral Nervous System Diseases/pathology , Proteolipids , Reverse Transcriptase Polymerase Chain Reaction/methods , Sural Nerve/metabolism , Sural Nerve/pathology , Sural Nerve/ultrastructure
10.
Hum Mutat ; 27(1): 69-77, 2006 Jan.
Article in English | MEDLINE | ID: mdl-16287154

ABSTRACT

Mutations in the proteolipid protein 1 (PLP1) gene cause the X-linked dysmyelinating diseases Pelizaeus-Merzbacher disease (PMD) and spastic paraplegia 2 (SPG2). We examined the severity of the following mutations that were suspected of affecting levels of PLP1 and DM20 RNA, the alternatively spliced products of PLP1: c.453G>A, c.453G>T, c.453G>C, c.453+2T>C, c.453+4A>G, c.347C>A, and c.453+28_+46del (the old nomenclature did not include the methionine codon: G450A, G450T, G450C, IVS3+2T>C, IVS3+4A>G, C344A, and IVS3+28-+46del). These mutations were evaluated by information theory-based analysis and compared with mRNA expression of the alternatively spliced products. The results are discussed relative to the clinical severity of disease. We conclude that the observed PLP1 and DM20 splicing patterns correlated well with predictions of information theory-based analysis, and that the relative strength of the PLP1 and DM20 donor splice sites plays an important role in PLP1 alternative splicing.


Subject(s)
Alternative Splicing/genetics , Membrane Proteins/genetics , Myelin Proteolipid Protein/genetics , Oligodendroglia/metabolism , RNA Splice Sites/genetics , Animals , Cells, Cultured , Exons/genetics , Fibroblasts/metabolism , Humans , Information Theory , Mutation/genetics , Phenotype , RNA, Messenger/genetics , RNA, Messenger/metabolism , Rats , Rats, Sprague-Dawley , Skin/cytology
11.
J Cell Biochem ; 97(5): 999-1016, 2006 Apr 01.
Article in English | MEDLINE | ID: mdl-16288477

ABSTRACT

Proteolipid protein (PLP) and DM20 are generated by alternative splicing of exon 3B of PLP1 transcript in differentiating oligodendrocytes. We investigated the role of exonic splicing enhancers (ESE) in the selection of PLP 5' donor site, focusing on putative ASF/SF2, and SC35 binding motifs in exon 3B on the basis of mutations that cause disease in humans. Mutations in a putative ASF/SF2 binding motif (nucleotides 406-412) reduced PLP 5' donor site selection, whereas a mutation in a putative SC35 binding motif (nucleotides 382-389) had no effect. UV crosslinking and immunoprecipitation (IP) assays using an antibody to ASF/SF2 showed that the ASF/SF2 protein specifically binds to the ESE (nucleotides 406-412). The single nucleotide mutations that reduced PLP splice site selection greatly diminished ASF/SF2 protein binding to this motif. We next tested the effect of overexpressed ASF/SF2 on PLP 5'splice selection in differentiating oligodendrocytes. ASF/SF2 positively regulates PLP splice site selection in a concentration-dependent manner. Disruption of the putative ASF/SF2 binding site in exon 3B reduced the positive effect of ASF/SF2 on PLP splicing. We conclude that an ESE in exon3B regulates PLP 5' donor site selection and that ASF/SF2 protein participates in the regulation of PLP alternative splicing in oligodendrocytes.


Subject(s)
Alternative Splicing , Enhancer Elements, Genetic , Exons , Myelin Proteolipid Protein/genetics , Nerve Tissue Proteins/genetics , Oligodendroglia/physiology , Amino Acid Motifs , Animals , Binding Sites , Cell Differentiation , Cell Lineage/physiology , Cells, Cultured , Humans , Mutation , Promoter Regions, Genetic , RNA Splice Sites , Rats , Rats, Sprague-Dawley , Transfection
12.
Am J Hum Genet ; 77(6): 966-87, 2005 Dec.
Article in English | MEDLINE | ID: mdl-16380909

ABSTRACT

We describe genomic structures of 59 X-chromosome segmental duplications that include the proteolipid protein 1 gene (PLP1) in patients with Pelizaeus-Merzbacher disease. We provide the first report of 13 junction sequences, which gives insight into underlying mechanisms. Although proximal breakpoints were highly variable, distal breakpoints tended to cluster around low-copy repeats (LCRs) (50% of distal breakpoints), and each duplication event appeared to be unique (100 kb to 4.6 Mb in size). Sequence analysis of the junctions revealed no large homologous regions between proximal and distal breakpoints. Most junctions had microhomology of 1-6 bases, and one had a 2-base insertion. Boundaries between single-copy and duplicated DNA were identical to the reference genomic sequence in all patients investigated. Taken together, these data suggest that the tandem duplications are formed by a coupled homologous and nonhomologous recombination mechanism. We suggest repair of a double-stranded break (DSB) by one-sided homologous strand invasion of a sister chromatid, followed by DNA synthesis and nonhomologous end joining with the other end of the break. This is in contrast to other genomic disorders that have recurrent rearrangements formed by nonallelic homologous recombination between LCRs. Interspersed repetitive elements (Alu elements, long interspersed nuclear elements, and long terminal repeats) were found at 18 of the 26 breakpoint sequences studied. No specific motif that may predispose to DSBs was revealed, but single or alternating tracts of purines and pyrimidines that may cause secondary structures were common. Analysis of the 2-Mb region susceptible to duplications identified proximal-specific repeats and distal LCRs in addition to the previously reported ones, suggesting that the unique genomic architecture may have a role in nonrecurrent rearrangements by promoting instability.


Subject(s)
Chromosomes, Human, X , Gene Duplication , Genetic Heterogeneity , Pelizaeus-Merzbacher Disease/genetics , Recombination, Genetic , Base Sequence , Chromosome Breakage , Chromosome Mapping , Cohort Studies , Computational Biology , Dosage Compensation, Genetic , Humans , In Situ Hybridization, Fluorescence , Membrane Proteins/genetics , Molecular Sequence Data , Myelin Proteolipid Protein/genetics , Polymerase Chain Reaction , Sequence Analysis, DNA , Sequence Homology, Nucleic Acid , Tandem Repeat Sequences
13.
Ann Neurol ; 53(3): 354-65, 2003 Mar.
Article in English | MEDLINE | ID: mdl-12601703

ABSTRACT

Proteolipid protein (PLP1) and its alternatively spliced isoform, DM20, are the major myelin proteins in the CNS, but are also expressed in the PNS. The proteins have an identical sequence except for 35 amino acids in PLP1 (the PLP1-specific domain) not present in DM20. Mutations of PLP1/DM20 cause Pelizaeus-Merzbacher Disease (PMD), a leukodystrophy, and in some instances, a peripheral neuropathy. To identify which mutations cause neuropathy, we have evaluated a cohort of patients with PMD and PLP1 mutations for the presence of neuropathy. As shown previously, all patients with PLP1 null mutations had peripheral neuropathy. We also identified 4 new PLP1 point mutations that cause both PMD and peripheral neuropathy, three of which truncate PLP1 expression within the PLP1-specific domain, but do not alter DM20. The fourth, a splicing mutation, alters both PLP1 and DM20, and is probably a null mutation. Six PLP1 point mutations predicted to produce proteins with an intact PLP1-specific domain do not cause peripheral neuropathy. Sixty-one individuals with PLP1 duplications also had normal peripheral nerve function. These data demonstrate that expression of PLP1 but not DMSO is necessary to prevent neuropathy, and suggest that the 35 amino acid PLP1-specific domain plays an important role in normal peripheral nerve function.


Subject(s)
Myelin Proteolipid Protein/biosynthesis , Nerve Tissue Proteins , Pelizaeus-Merzbacher Disease/metabolism , Peripheral Nervous System Diseases/metabolism , Schwann Cells/metabolism , Adolescent , Adult , Amino Acid Sequence , Animals , COS Cells , Child , Chlorocebus aethiops , Cohort Studies , Female , Humans , Male , Molecular Sequence Data , Mutation/genetics , Myelin Proteolipid Protein/genetics , Pelizaeus-Merzbacher Disease/genetics , Pelizaeus-Merzbacher Disease/pathology , Peripheral Nervous System Diseases/genetics , Peripheral Nervous System Diseases/pathology , Rats
14.
Ann Neurol ; 52(4): 477-88, 2002 Oct.
Article in English | MEDLINE | ID: mdl-12325077

ABSTRACT

We report that a deletion of 19 base pairs (bp) in intron 3 of the proteolipid protein (PLP/DM20) gene causes a neurological disease characterized by mild developmental delay, followed by progressive decline of acquired motor and cognitive milestones. The clinical features are associated with mild delay in myelination demonstrated by magnetic resonance imaging studies and with ongoing demyelination and axonal loss demonstrated by magnetic resonance spectroscopy. We demonstrate that the purine-rich 19bp element regulates PLP-specific splice site selection in transient transfections of chimeric constructs into cultured oligodendrocytes. Runs of 4 and 5 Gs centered in the 19bp element are critical for efficient PLP-specific splicing. The intronic element is sequence specific in oligodendrocytes and is not a repressor of PLP-specific splicing in nonglial cells. These data support the conclusion that deletion of the 19bp purine-rich region in PLP intron 3 causes a reduction in PLP message and protein, which affects myelin stability and axonal integrity.


Subject(s)
Gene Deletion , Myelin Proteolipid Protein/genetics , Pelizaeus-Merzbacher Disease/genetics , RNA Splicing , Animals , Base Sequence , Brain/pathology , COS Cells , Chromosome Mapping , Female , Humans , Introns/genetics , Magnetic Resonance Imaging , Molecular Sequence Data , Pelizaeus-Merzbacher Disease/pathology , Phenotype , Regulatory Sequences, Nucleic Acid/genetics , Transfection
SELECTION OF CITATIONS
SEARCH DETAIL
...