Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 27
Filtrar
1.
Am J Hum Genet ; 109(12): 2270-2282, 2022 12 01.
Artigo em Inglês | MEDLINE | ID: mdl-36368327

RESUMO

An Xq22.2 region upstream of PLP1 has been proposed to underly a neurological disease trait when deleted in 46,XX females. Deletion mapping revealed that heterozygous deletions encompassing the smallest region of overlap (SRO) spanning six Xq22.2 genes (BEX3, RAB40A, TCEAL4, TCEAL3, TCEAL1, and MORF4L2) associate with an early-onset neurological disease trait (EONDT) consisting of hypotonia, intellectual disability, neurobehavioral abnormalities, and dysmorphic facial features. None of the genes within the SRO have been associated with monogenic disease in OMIM. Through local and international collaborations facilitated by GeneMatcher and Matchmaker Exchange, we have identified and herein report seven de novo variants involving TCEAL1 in seven unrelated families: three hemizygous truncating alleles; one hemizygous missense allele; one heterozygous TCEAL1 full gene deletion; one heterozygous contiguous deletion of TCEAL1, TCEAL3, and TCEAL4; and one heterozygous frameshift variant allele. Variants were identified through exome or genome sequencing with trio analysis or through chromosomal microarray. Comparison with previously reported Xq22 deletions encompassing TCEAL1 identified a more-defined syndrome consisting of hypotonia, abnormal gait, developmental delay/intellectual disability especially affecting expressive language, autistic-like behavior, and mildly dysmorphic facial features. Additional features include strabismus, refractive errors, variable nystagmus, gastroesophageal reflux, constipation, dysmotility, recurrent infections, seizures, and structural brain anomalies. An additional maternally inherited hemizygous missense allele of uncertain significance was identified in a male with hypertonia and spasticity without syndromic features. These data provide evidence that TCEAL1 loss of function causes a neurological rare disease trait involving significant neurological impairment with features overlapping the EONDT phenotype in females with the Xq22 deletion.


Assuntos
Transtorno Autístico , Deficiência Intelectual , Feminino , Humanos , Masculino , Transtorno Autístico/genética , Deficiência Intelectual/genética , Deficiência Intelectual/complicações , Hipotonia Muscular/genética , Hipotonia Muscular/complicações , Fenótipo , Síndrome , Fatores de Transcrição/genética
2.
Am J Med Genet A ; 182(8): 1906-1912, 2020 08.
Artigo em Inglês | MEDLINE | ID: mdl-32573057

RESUMO

Leukodystrophies are a heterogeneous group of heritable disorders characterized by abnormal brain white matter signal on magnetic resonance imaging (MRI) and primary involvement of the cellular components of myelin. Previous estimates suggest the incidence of leukodystrophies as a whole to be 1 in 7,000 individuals, however the frequency of specific diagnoses relative to others has not been described. Next generation sequencing approaches offer the opportunity to redefine our understanding of the relative frequency of different leukodystrophies. We assessed the relative frequency of all 30 leukodystrophies (associated with 55 genes) in more than 49,000 exomes. We identified a relatively high frequency of disorders previously thought of as very rare, including Aicardi Goutières Syndrome, TUBB4A-related leukodystrophy, Peroxisomal biogenesis disorders, POLR3-related Leukodystrophy, Vanishing White Matter, and Pelizaeus-Merzbacher Disease. Despite the relative frequency of these conditions, carrier-screening laboratories regularly test only 20 of the 55 leukodystrophy-related genes, and do not test at all, or test only one or a few, genes for some of the higher frequency disorders. Relative frequency of leukodystrophies previously considered very rare suggests these disorders may benefit from expanded carrier screening.


Assuntos
Doenças Autoimunes do Sistema Nervoso/genética , Doenças Desmielinizantes/genética , Malformações do Sistema Nervoso/genética , Doença de Pelizaeus-Merzbacher/genética , RNA Polimerase III/genética , Tubulina (Proteína)/genética , Doenças Autoimunes do Sistema Nervoso/patologia , Doenças Desmielinizantes/epidemiologia , Doenças Desmielinizantes/patologia , Exoma/genética , Feminino , Predisposição Genética para Doença , Heterozigoto , Sequenciamento de Nucleotídeos em Larga Escala , Humanos , Doenças por Armazenamento dos Lisossomos/epidemiologia , Doenças por Armazenamento dos Lisossomos/genética , Imageamento por Ressonância Magnética , Masculino , Bainha de Mielina/genética , Bainha de Mielina/metabolismo , Malformações do Sistema Nervoso/patologia , Doença de Pelizaeus-Merzbacher/epidemiologia , Doença de Pelizaeus-Merzbacher/patologia , Substância Branca/diagnóstico por imagem , Substância Branca/patologia
3.
Ann Clin Transl Neurol ; 7(1): 144-152, 2020 01.
Artigo em Inglês | MEDLINE | ID: mdl-31912665

RESUMO

Genetic white matter disorders have heterogeneous etiologies and overlapping clinical presentations. We performed a study of the diagnostic efficacy of genome sequencing in 41 unsolved cases with prior exome sequencing, resolving an additional 14 from an historical cohort (n = 191). Reanalysis in the context of novel disease-associated genes and improved variant curation and annotation resolved 64% of cases. The remaining diagnoses were directly attributable to genome sequencing, including cases with small and large copy number variants (CNVs) and variants in deep intronic and technically difficult regions. Genome sequencing, in combination with other methodologies, achieved a diagnostic yield of 85% in this retrospective cohort.


Assuntos
Leucoencefalopatias/diagnóstico , Leucoencefalopatias/genética , Sistema de Registros , Sequenciamento Completo do Genoma , Adolescente , Criança , Pré-Escolar , Feminino , Humanos , Leucoencefalopatias/patologia , Masculino , Linhagem
4.
Hum Mutat ; 41(1): 150-168, 2020 01.
Artigo em Inglês | MEDLINE | ID: mdl-31448840

RESUMO

Xq22 deletions that encompass PLP1 (Xq22-PLP1-DEL) are notable for variable expressivity of neurological disease traits in females ranging from a mild late-onset form of spastic paraplegia type 2 (MIM# 312920), sometimes associated with skewed X-inactivation, to an early-onset neurological disease trait (EONDT) of severe developmental delay, intellectual disability, and behavioral abnormalities. Size and gene content of Xq22-PLP1-DEL vary and were proposed as potential molecular etiologies underlying variable expressivity in carrier females where two smallest regions of overlap (SROs) were suggested to influence disease. We ascertained a cohort of eight unrelated patients harboring Xq22-PLP1-DEL and performed high-density array comparative genomic hybridization and breakpoint-junction sequencing. Molecular characterization of Xq22-PLP1-DEL from 17 cases (eight herein and nine published) revealed an overrepresentation of breakpoints that reside within repeats (11/17, ~65%) and the clustering of ~47% of proximal breakpoints in a genomic instability hotspot with characteristic non-B DNA density. These findings implicate a potential role for genomic architecture in stimulating the formation of Xq22-PLP1-DEL. The correlation of Xq22-PLP1-DEL gene content with neurological disease trait in female cases enabled refinement of the associated SROs to a single genomic interval containing six genes. Our data support the hypothesis that genes contiguous to PLP1 contribute to EONDT.


Assuntos
Deleção Cromossômica , Cromossomos Humanos X , Estudos de Associação Genética , Predisposição Genética para Doença , Doenças do Sistema Nervoso/diagnóstico , Doenças do Sistema Nervoso/genética , Característica Quantitativa Herdável , Criança , Pré-Escolar , Pontos de Quebra do Cromossomo , Mapeamento Cromossômico , Hibridização Genômica Comparativa , Feminino , Estudos de Associação Genética/métodos , Humanos , Masculino , Linhagem , Fenótipo , Sequências Repetitivas de Ácido Nucleico , Fatores Sexuais , Síndrome , Inativação do Cromossomo X
5.
Genome Med ; 11(1): 80, 2019 12 09.
Artigo em Inglês | MEDLINE | ID: mdl-31818324

RESUMO

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.


Assuntos
Variações do Número de Cópias de DNA , Rearranjo Gênico , Mutação , Proteína Proteolipídica de Mielina/genética , Pontos de Quebra do Cromossomo , Hibridização Genômica Comparativa , Duplicação Gênica , Estudos de Associação Genética , Predisposição Genética para Doença , Genoma Humano , Instabilidade Genômica , Genômica/métodos , Humanos , Polimorfismo de Nucleotídeo Único
6.
Mol Ther Nucleic Acids ; 12: 420-432, 2018 Sep 07.
Artigo em Inglês | MEDLINE | ID: mdl-30195779

RESUMO

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.

7.
J Neurol ; 265(7): 1580-1589, 2018 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-29725841

RESUMO

Pelizaeus-Merzbacher disease (PMD; MIM 312080), an inherited defect of central nervous system myelin formation, affects individuals in many ways, including their hearing and language abilities. The aim of this study was to assess the auditory abilities in 18 patients with PMD by examining the functional processes along the central auditory pathways using auditory brainstem responses (ABR) and cortical auditory evoked potentials (CAEP) in response to speech sounds. The significant ABR anomalies confirm the existence of dyssynchrony previously described at the level of the brainstem in patients with PMD. Despite the significant auditory dyssynchrony observed at the level of the brainstem, CAEPs were present in most patients, albeit somehow abnormal in terms of morphology and latency, resembling a type of auditory neuropathy spectrum disorder.


Assuntos
Doenças Auditivas Centrais/etiologia , Potenciais Evocados Auditivos do Tronco Encefálico/fisiologia , Doença de Pelizaeus-Merzbacher/complicações , Testes de Impedância Acústica , Estimulação Acústica , Adolescente , Adulto , Doenças Auditivas Centrais/diagnóstico , Doenças Auditivas Centrais/patologia , Vias Auditivas/fisiopatologia , Criança , Pré-Escolar , Eletroencefalografia , Feminino , Humanos , Lactente , Masculino , Emissões Otoacústicas Espontâneas , Otoscopia , Adulto Jovem
8.
Am J Hum Genet ; 100(4): 617-634, 2017 Apr 06.
Artigo em Inglês | MEDLINE | ID: mdl-28366443

RESUMO

Pelizaeus-Merzbacher disease (PMD) is a pediatric disease of myelin in the central nervous system and manifests with a wide spectrum of clinical severities. Although PMD is a rare monogenic disease, hundreds of mutations in the X-linked myelin gene proteolipid protein 1 (PLP1) have been identified in humans. Attempts to identify a common pathogenic process underlying PMD have been complicated by an incomplete understanding of PLP1 dysfunction and limited access to primary human oligodendrocytes. To address this, we generated panels of human induced pluripotent stem cells (hiPSCs) and hiPSC-derived oligodendrocytes from 12 individuals with mutations spanning the genetic and clinical diversity of PMD-including point mutations and duplication, triplication, and deletion of PLP1-and developed an in vitro platform for molecular and cellular characterization of all 12 mutations simultaneously. We identified individual and shared defects in PLP1 mRNA expression and splicing, oligodendrocyte progenitor development, and oligodendrocyte morphology and capacity for myelination. These observations enabled classification of PMD subgroups by cell-intrinsic phenotypes and identified a subset of mutations for targeted testing of small-molecule modulators of the endoplasmic reticulum stress response, which improved both morphologic and myelination defects. Collectively, these data provide insights into the pathogeneses of a variety of PLP1 mutations and suggest that disparate etiologies of PMD could require specific treatment approaches for subsets of individuals. More broadly, this study demonstrates the versatility of a hiPSC-based panel spanning the mutational heterogeneity within a single disease and establishes a widely applicable platform for genotype-phenotype correlation and drug screening in any human myelin disorder.


Assuntos
Oligodendroglia/patologia , Doença de Pelizaeus-Merzbacher/genética , Doença de Pelizaeus-Merzbacher/patologia , Técnicas de Cultura de Células , Criança , Pré-Escolar , Estresse do Retículo Endoplasmático , Feminino , Humanos , Células-Tronco Pluripotentes Induzidas/patologia , Masculino , Proteína Proteolipídica de Mielina , Oligodendroglia/metabolismo
10.
Ann Clin Transl Neurol ; 2(6): 648-61, 2015 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-26125040

RESUMO

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.

11.
PLoS Genet ; 11(3): e1005050, 2015 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-25749076

RESUMO

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.


Assuntos
Duplicação Gênica , Proteína Proteolipídica de Mielina/genética , Doença de Pelizaeus-Merzbacher/genética , Pontos de Quebra do Cromossomo , Inversão Cromossômica , Dosagem de Genes , Humanos
12.
Pulm Pharmacol Ther ; 30: 22-31, 2015 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-25445931

RESUMO

Mice with Plp1 gene duplication model the most common form of Pelizaeus-Merzbacher disease (PMD), a CNS disease in which patients may suffer respiratory complications. We hypothesized that affected mice would lack airway responsiveness compared to wild-type and carrier mice during methacholine challenge. Wild-type (n = 10), carrier female (n = 6) and affected male (n = 8) mice were anesthetized-paralyzed, tracheostomized and ventilated. Respiratory mechanics were recorded at baseline and during escalating doses of nebulized methacholine followed by albuterol. Lung resistance (RL) was the primary endpoint. Lung tissues were assayed for inflammatory and histological differences. At baseline, phase angles were higher in carrier and affected mice than wild-type. Dose-response RL curves in affected and carrier mice indicated a lack of methacholine response. Albuterol reduced RL in wild-type and carrier, but not affected mice. Affected mice exhibited lower interleukin (IL)-6 tissue levels and alveolar inflammatory infiltrates. Affected and carrier mice, compared to wild-type, lacked airway reactivity during methacholine challenge, but only affected mice exhibited decreased lung tissue levels of IL-6 and inflammation.


Assuntos
Duplicação Gênica , Proteína Proteolipídica de Mielina/genética , Doença de Pelizaeus-Merzbacher/fisiopatologia , Pneumonia/fisiopatologia , Albuterol/farmacologia , Animais , Modelos Animais de Doenças , Relação Dose-Resposta a Droga , Feminino , Interleucina-6/metabolismo , Masculino , Cloreto de Metacolina/administração & dosagem , Cloreto de Metacolina/farmacologia , Camundongos , Doença de Pelizaeus-Merzbacher/genética , Pneumonia/genética , Mecânica Respiratória/efeitos dos fármacos , Mecânica Respiratória/genética
13.
Hum Mol Genet ; 23(20): 5464-78, 2014 Oct 15.
Artigo em Inglês | MEDLINE | ID: mdl-24890387

RESUMO

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.


Assuntos
Processamento Alternativo , Íntrons , Proteína Proteolipídica de Mielina/genética , Doença de Pelizaeus-Merzbacher/genética , RNA Mensageiro/química , Pareamento de Bases , Linhagem Celular , Feminino , Humanos , Masculino , Modelos Moleculares , Mutação , Proteína Proteolipídica de Mielina/metabolismo , Conformação de Ácido Nucleico , Oligodendroglia/metabolismo , Linhagem , RNA Mensageiro/metabolismo , Análise de Sequência de DNA
14.
Mol Genet Metab ; 111(3): 393-398, 2014 03.
Artigo em Inglês | MEDLINE | ID: mdl-24374284

RESUMO

OBJECTIVE: Pelizaeus-Merzbacher-like disease is a rare hypomyelinating leukodystrophy caused by autosomal recessive mutations in GJC2, encoding a gap junction protein essential for production of a mature myelin sheath. A previously identified GJC2 mutation (c.-167A>G) in the promoter region is hypothesized to disrupt a putative SOX10 binding site; however, the lack of additional mutations in this region and contradictory functional data have limited the interpretation of this variant. METHODS: We describe two independent Pelizaeus-Merzbacher-like disease families with a novel promoter region mutation and updated in vitro functional assays. RESULTS: A novel GJC2 mutation (c.-170A>G) in the promoter region was identified in Pelizaeus-Merzbacher-like disease patients. In vitro functional assays using human GJC2 promoter constructs demonstrated that this mutation and the previously described c.-167A>G mutation similarly diminished the transcriptional activity driven by SOX10 and the binding affinity for SOX10. INTERPRETATION: These findings support the role of GJC2 promoter mutations in Pelizaeus-Merzbacher-like disease. GJC2 promoter region mutation screening should be included in the evaluation of patients with unexplained hypomyelinating leukodystrophies.


Assuntos
Conexinas/genética , Doenças Desmielinizantes Hereditárias do Sistema Nervoso Central/genética , Regiões Promotoras Genéticas , Fatores de Transcrição SOXE/metabolismo , Adulto , Sítios de Ligação , Criança , Conexinas/metabolismo , Feminino , Doenças Desmielinizantes Hereditárias do Sistema Nervoso Central/metabolismo , Doenças Desmielinizantes Hereditárias do Sistema Nervoso Central/patologia , Humanos , Masculino , Dados de Sequência Molecular , Mutação , Bainha de Mielina/patologia , Ligação Proteica , Fatores de Transcrição SOXE/genética
15.
J Neurosci ; 33(29): 11788-99, 2013 Jul 17.
Artigo em Inglês | MEDLINE | ID: mdl-23864668

RESUMO

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.


Assuntos
Doenças Desmielinizantes/fisiopatologia , Marcha/genética , Coxeadura Animal/fisiopatologia , Proteína Proteolipídica de Mielina/genética , Bainha de Mielina/patologia , Doença de Pelizaeus-Merzbacher/fisiopatologia , Animais , Doenças Desmielinizantes/genética , Doenças Desmielinizantes/patologia , Modelos Animais de Doenças , Progressão da Doença , Genótipo , Coxeadura Animal/genética , Coxeadura Animal/patologia , Camundongos , Camundongos Transgênicos , Mutação , Bainha de Mielina/genética , Doença de Pelizaeus-Merzbacher/genética , Doença de Pelizaeus-Merzbacher/patologia
16.
Semin Neurol ; 32(1): 62-7, 2012 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-22422208

RESUMO

The purpose of this article is to present contemporary information on the clinical and molecular diagnosis and the treatment of Pelizaeus-Merzbacher's disease (PMD) and related leukodystrophies. Various types of mutations of the X-linked proteolipid protein 1 gene (PLP1) that include copy number changes, point mutations, and insertions or deletions of a few bases lead to a clinical spectrum from the most severe connatal PMD, to the least severe spastic paraplegia 2 (SPG2). Signs of PMD include nystagmus, hypotonia, tremors, titubation, ataxia, spasticity, athetotic movements and cognitive impairment; the major findings in SPG2 are leg weakness and spasticity. A diffuse pattern of hypomyelination is seen on magnetic resonance imaging (MRI) of PMD/SPG2 patients. A similar constellation of signs and pattern of hypomyelination lead to the autosomal recessive disease called Pelizaeus-Merzbacher-like disease 1 (PMLD1) and the less-severe spastic paraplegia 44 (SPG44), caused by mutations of the gap junction protein, gamma-2 gene (GJC2), formerly known as the gap junction protein, α-12 gene (GJA12). Magnetic resonance spectroscopy (MRS) and brainstem auditory evoked potentials (BAEP) may assist with differential clinical diagnosis of PMD and PMLD1. Supportive therapy for patients with PMD/SPG2 and PMLD1/SPG44 includes medications for seizures and spasticity; physical therapy, exercise, and orthotics for spasticity management; surgery for contractures and scoliosis; gastrostomy for severe dysphagia; proper wheelchair seating, physical therapy, and orthotics to prevent or ameliorate the effects of scoliosis; special education; and assistive communication devices.


Assuntos
Doenças Desmielinizantes/genética , Paraplegia/genética , Doença de Pelizaeus-Merzbacher/diagnóstico , Doença de Pelizaeus-Merzbacher/genética , Doenças Desmielinizantes/diagnóstico , Feminino , Humanos , Imageamento por Ressonância Magnética , Espectroscopia de Ressonância Magnética , Masculino , Mutação/genética , Paraplegia/diagnóstico , Doença de Pelizaeus-Merzbacher/terapia
17.
Am J Med Genet A ; 149A(8): 1698-705, 2009 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-19610109

RESUMO

A female patient is described with clinical symptoms of both microphthalmia with linear skin defects (MLS or MIDAS) and dental enamel defects, having an appearance compatible with X-linked amelogenesis imperfecta (XAI). Genomic DNA was purified from the patient's blood and semiquantitative multiplex PCR revealed a deletion encompassing the amelogenin gene (AMELX). Because MLS is also localized to Xp22, genomic DNA was subjected to array comparative genomic hybridization, and a large heterozygous deletion was identified. Histopathology of one primary and one permanent molar tooth showed abnormalities in the dental enamel layer, and a third tooth had unusually high microhardness measurements, possibly due to its ultrastructural anomalies as seen by scanning electron microscopy. This is the first report of a patient with both of these rare conditions, and the first description of the phenotype resulting from a deletion encompassing the entire AMELX gene. More than 50 additional genes were monosomic in this patient.


Assuntos
Amelogênese Imperfeita/complicações , Amelogênese Imperfeita/genética , Deleção Cromossômica , Cromossomos Humanos X/genética , Microftalmia/complicações , Microftalmia/genética , Anormalidades da Pele/complicações , Adolescente , Densidade Óssea/genética , Criança , Pré-Escolar , Análise Mutacional de DNA , Esmalte Dentário/patologia , Esmalte Dentário/ultraestrutura , Dentina/metabolismo , Feminino , Dosagem de Genes , Predisposição Genética para Doença , Dureza , Humanos , Anormalidades da Pele/genética , Inativação do Cromossomo X , Adulto Jovem
18.
Acta Neuropathol ; 118(4): 531-9, 2009 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-19562355

RESUMO

Mutations affecting proteolipid protein 1 (PLP1), the major protein in central nervous system myelin, cause the X-linked leukodystrophy Pelizaeus-Merzbacher disease (PMD). We describe the neuropathologic findings in a series of eight male PMD subjects with confirmed PLP1 mutations, including duplications, complete gene deletion, missense and exon-skipping. While PLP1 mutations have effects on oligodendrocytes that result in mutation-specific degrees of dysmyelination, our findings indicate that there are also unexpected effects in the central nervous system resulting in neuronal loss. Although length-dependent axonal degeneration has been described in PLP1 null mutations, there have been no reports on neuronal degeneration in PMD patients. We now demonstrate widespread neuronal loss in PMD. The patterns of neuronal loss appear to be dependent on the mutation type, suggesting selective vulnerability of neuronal populations that depends on the nature of the PLP1 disturbance. Nigral neurons, which were not affected in patients with either null or severe misfolding mutations, and thalamic neurons appear particularly vulnerable in PLP1 duplication and deletion patients, while hippocampal neuronal loss was prominent in a patient with complete PLP1 gene deletion. All subjects showed cerebellar neuronal loss. The patterns of neuronal involvement may explain some clinical findings, such as ataxia, being more prominent in PMD than in other leukodystrophies. While the precise pathogenetic mechanisms are not known, these observations suggest that defective glial functions contribute to neuronal pathology.


Assuntos
Encéfalo/patologia , Morte Celular/genética , Proteína Proteolipídica de Mielina/genética , Neurônios/patologia , Doença de Pelizaeus-Merzbacher/genética , Doença de Pelizaeus-Merzbacher/patologia , Adulto , Fatores Etários , Cromossomos Humanos X , Marcadores Genéticos , Humanos , Imuno-Histoquímica , Masculino , Pessoa de Meia-Idade , Mutação , Bainha de Mielina/genética , Bainha de Mielina/patologia , Neuroglia/patologia
19.
J Child Neurol ; 24(5): 618-24, 2009 May.
Artigo em Inglês | MEDLINE | ID: mdl-19151366

RESUMO

Pelizaeus-Merzbacher disease is a rare X-linked disorder caused by mutations of the proteolipid protein 1 gene that encodes a structural component of myelin. It is characterized by progressive psychomotor delay, nystagmus, spastic quadriplegia, and cerebellar ataxia. Variable clinical expression was seen in 5 members of a family bearing a novel missense mutation in proteolipid protein 1, c.619T>C. Symptomatic patients included a 6-year-old girl, her younger brother, and their maternal uncle, a 29-year-old college graduate initially diagnosed with cerebral palsy; their brain magnetic resonance imaging studies showed diffuse dysmyelination. The mother had a history of delayed walking, achieved independently by age 3; she and the maternal grandmother were asymptomatic on presentation. Review of clinical information and family history led to consideration of Pelizaeus-Merzbacher disease. Subsequent identification of the causal mutation enabled preimplantation genetic diagnosis and the birth of an unaffected child.


Assuntos
Mutação de Sentido Incorreto , Proteína Proteolipídica de Mielina/genética , Doença de Pelizaeus-Merzbacher/genética , Adulto , Encéfalo/patologia , Paralisia Cerebral/diagnóstico , Criança , Pré-Escolar , Análise Mutacional de DNA , Diagnóstico Diferencial , Família , Feminino , Humanos , Masculino , Pessoa de Meia-Idade , Bainha de Mielina/patologia , Linhagem , Doença de Pelizaeus-Merzbacher/diagnóstico , Doença de Pelizaeus-Merzbacher/patologia , Mutação Puntual , Diagnóstico Pré-Implantação
20.
Exp Neurol ; 214(2): 322-30, 2008 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-18835559

RESUMO

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.


Assuntos
Proteína Proteolipídica de Mielina/genética , Bainha de Mielina/fisiologia , Doença de Pelizaeus-Merzbacher/genética , Doença de Pelizaeus-Merzbacher/fisiopatologia , Processamento Alternativo , Animais , Comportamento Animal , Encéfalo/crescimento & desenvolvimento , Encéfalo/patologia , Modelos Animais de Doenças , Elementos Facilitadores Genéticos , Deleção de Genes , Técnicas de Introdução de Genes , Íntrons/genética , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Mutantes , Microscopia Eletrônica , Atividade Motora , Proteínas da Mielina/genética , Proteínas da Mielina/metabolismo , Proteína Proteolipídica de Mielina/metabolismo , Bainha de Mielina/patologia , Bainha de Mielina/ultraestrutura , Nervo Óptico/crescimento & desenvolvimento , Nervo Óptico/patologia , Nervo Óptico/ultraestrutura , Doença de Pelizaeus-Merzbacher/patologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...