Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 21
Filtrar
1.
Atheroscler Plus ; 44: 43-50, 2021 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-36644668

RESUMO

Background and aims: High-density lipoproteins (HDL) of patients with type 2 diabetes mellitus (T2DM) have impaired anti-inflammatory activities. The anti-inflammatory activity of HDL has been determined ex vivo after isolation by different methods from blood mostly obtained after overnight fasting. We first determined the effect of the HDL isolation method, and subsequently the effect of food intake on the anti-inflammatory function of HDL from T2DM patients. Methods: Blood was collected from healthy controls and T2DM patients after an overnight fast, and from T2DM patients 3 h after breakfast (n = 17 each). HDL was isolated by a two-step density gradient ultracentrifugation in iodixanol (HDLDGUC2), by sequential salt density flotation (HDLSEQ) or by PEG precipitation (HDLPEG). The anti-inflammatory function of HDL was determined by the reduction of the TNFα-induced expression of VCAM-1 in human coronary artery endothelial cells (HCAEC) and retinal endothelial cells (REC). Results: HDL isolated by the three different methods from healthy controls inhibited TNFα-induced VCAM-1 expression in HCAEC. With apoA-I at 0.7 µM, HDLDGUC2 and HDLSEQ were similarly effective (16% versus 14% reduction; n = 3; p > 0.05) but less effective than HDLPEG (28%, p < 0.05). Since ultracentrifugation removes most of the unbound plasma proteins, we used HDLDGUC2 for further experiments. With apoA-I at 3.2 µM, HDL from fasting healthy controls and T2DM patients reduced TNFα-induced VCAM-1 expression in HCAEC by 58 ± 13% and 51 ± 20%, respectively (p = 0.35), and in REC by 42 ± 13% and 25 ± 18%, respectively (p < 0.05). Compared to preprandial HDL, postprandial HDL from T2DM patients reduced VCAM-1 expression by 56 ± 16% (paired test: p < 0.001) in HCAEC and by 34 ± 13% (paired test: p < 0.05) in REC. Conclusions: The ex vivo anti-inflammatory activity of HDL is affected by the HDL isolation method. Two-step ultracentrifugation in an iodixanol gradient is a suitable method for HDL isolation when testing HDL anti-inflammatory function. The anti-inflammatory activity of HDL from overnight fasted T2DM patients is significantly impaired in REC but not in HCAEC. The anti-inflammatory function of HDL is partly restored by food intake.

2.
J Neurol ; 266(12): 2987-2996, 2019 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-31471688

RESUMO

OBJECTIVE: To study scapular winging or other forms of scapular dyskinesis (condition of alteration of the normal position and motion of the scapula) in myotonic dystrophy type 1 (DM1), which is generally considered to be a distal myopathy, we performed an observational cohort study. METHODS: We performed a prospective cohort study on the clinical features and progression over time of 33 patients with DM1 and pronounced, mostly asymmetric scapular winging or other forms of scapular dyskinesis. We also explored if scapular dyskinesis in DM1 has the same genetic background as in facioscapulohumeral muscular dystrophy type 1 (FSHD1). RESULTS: The cohort included patients with congenital (n = 3), infantile (n = 6) and adult-onset DM1 (n = 24). Scapular girdle examination showed moderate shoulder girdle weakness (mean MRC 3) and atrophy of trapezius, infraspinatus, and rhomboid major, seemingly similar as in FSHD1. Shoulder abduction and forward flexion were limited (50-70°). In five patients, scapular dyskinesis was the initial disease symptom; in the others it appeared 1-24 years after disease onset. Follow-up data were available in 29 patients (mean 8 years) and showed mild to severe increase of scapular dyskinesis over time. In only three patients, DM1 coexisted with a FSHD mutation. In all other patients, FSHD was genetically excluded. DM2 was genetically excluded in nine patients. The clinical features of the patients with both DM1 and FSHD1 mutations were similar to those with DM1 only. CONCLUSION: Scapular dyskinesis can be considered to be part of DM1 in a small proportion of patients. In spite of the clinical overlap, FSHD can explain scapular dyskinesis only in a small minority. This study is expected to improve the recognition of shoulder girdle involvement in DM1, which will contribute to the management of these patients.


Assuntos
Progressão da Doença , Discinesias/fisiopatologia , Distrofia Miotônica/fisiopatologia , Escápula/fisiopatologia , Adulto , Idade de Início , Idoso , Discinesias/classificação , Discinesias/etiologia , Feminino , Humanos , Masculino , Pessoa de Meia-Idade , Distrofia Muscular Facioescapuloumeral/complicações , Distrofia Muscular Facioescapuloumeral/genética , Distrofia Muscular Facioescapuloumeral/fisiopatologia , Distrofia Miotônica/complicações , Distrofia Miotônica/genética , Estudos Prospectivos , Índice de Gravidade de Doença , Adulto Jovem
3.
Neurology ; 75(17): 1548-54, 2010 Oct 26.
Artigo em Inglês | MEDLINE | ID: mdl-20975055

RESUMO

OBJECTIVE: In some 5% of patients with facioscapulohumeral muscular dystrophy (FSHD), no D4Z4 repeat contraction on chromosome 4q35 is observed. Such patients, termed patients with FSHD2, show loss of DNA methylation and heterochromatin markers at the D4Z4 repeat that are similar to patients with D4Z4 contractions (FSHD1). This commonality suggests that a change in D4Z4 chromatin structure unifies FSHD1 and FSHD2. The aim of our study was to critically evaluate the clinical features in patients with FSHD2 in order to establish whether these patients are phenotypically identical to FSHD1 and to establish the effects of the (epi-) genotype on the phenotype. METHODS: This cross-sectional study studied 33 patients with FSHD2 from 27 families, the largest cohort described to date. All patients were clinically assessed using a standardized clinical evaluation form. Genotype analysis was performed by pulsed field gel electrophoresis and PCR; D4Z4 methylation was studied by methylation-sensitive Southern blot analysis. RESULTS: FSHD2 is identical to FSHD1 in its clinical presentation. Notable differences include a higher incidence (67%) of sporadic cases and the absence of gender differences in disease severity in FSHD2. Overall, average disease severity in FSHD2 was similar to that reported in FSHD1 and was not influenced by D4Z4 repeat size. In FSHD2, a small effect of the degree of hypomethylation on disease severity was observed. CONCLUSIONS: Clinically, patients with FSHD2 are indistinguishable from patients with FSHD1. The present data suggest that FSHD1 and FSHD2 are the result of the same pathophysiologic process.


Assuntos
Distrofia Muscular Facioescapuloumeral/genética , Distrofia Muscular Facioescapuloumeral/fisiopatologia , Proteínas Nucleares/genética , Adolescente , Adulto , Criança , Pré-Escolar , Cromossomos Humanos Par 4 , Estudos de Coortes , Estudos Transversais , Metilação de DNA/genética , Expansão das Repetições de DNA/genética , Saúde da Família , Feminino , Genótipo , Humanos , Lactente , Recém-Nascido , Masculino , Proteínas dos Microfilamentos , Pessoa de Meia-Idade , Fenótipo , Polimorfismo Genético/genética , Proteínas de Ligação a RNA , Adulto Jovem
4.
Neurology ; 68(8): 578-82, 2007 Feb 20.
Artigo em Inglês | MEDLINE | ID: mdl-17229919

RESUMO

BACKGROUND: In the majority of facioscapulohumeral muscular dystrophy (FSHD) cases, the molecular basis of the disease is due to loss of subtelomeric D4Z4 repeat units at 4q35. Occasionally, an apparent absence of the contracted D4Z4 repeat is associated with FSHD. One explanation for this finding is a deletion in the region proximal to the D4Z4 repeat array that encompasses the p13E-11 (D4F104S1) probe-binding site used in the DNA diagnosis. The frequency of such proximally extended deletions is unknown, and to date, few patients have been described due to the difficulties in the molecular identification of such cases. METHODS: We describe a family (DUK 2531) in which a contracted D4Z4 allele and a large proximal deletion of approximately 75 kb are segregating to 11 individuals. This is the largest deletion identified to date. Family DUK 2531 was initially thought to have normal D4Z4 fragment size and therefore unlinked to the 4q35 region (FSHD1B). RESULTS: Further molecular analysis of DUK 2531 reveals the presence of 10 repeat units (33 kb). The extended deletion includes the probe p13E-11 and B31 binding sites, the inverted repeat D4S2463, and genes FRG2 and TUBB4Q. CONCLUSION: Despite the length of the proximal deletion in this family, the range and severity of the clinical manifestations are typical for the disorder. Because such deletions can lead to misinterpretation in the diagnostic setting, this suggests the need for additional diagnostic tests in facioscapulohumeral muscular dystrophy.


Assuntos
Cromossomos Humanos Par 4/genética , Deleção de Genes , Predisposição Genética para Doença/genética , Distrofia Muscular Facioescapuloumeral/genética , Distrofia Muscular Facioescapuloumeral/fisiopatologia , Mutação/genética , Adulto , Aberrações Cromossômicas , Análise Mutacional de DNA , Feminino , Dosagem de Genes/genética , Frequência do Gene/genética , Testes Genéticos , Genótipo , Humanos , Masculino , Debilidade Muscular/diagnóstico , Debilidade Muscular/genética , Debilidade Muscular/fisiopatologia , Músculo Esquelético/patologia , Músculo Esquelético/fisiopatologia , Distrofia Muscular Facioescapuloumeral/diagnóstico , Linhagem , Fenótipo
5.
Neurology ; 67(8): 1464-6, 2006 Oct 24.
Artigo em Inglês | MEDLINE | ID: mdl-17060574

RESUMO

The diagnosis of facioscapulohumeral muscular dystrophy (FSHD) can be difficult due to its clinical variability and complex genetic cause. We present three challenging cases: one misdiagnosis of FSHD, one patient with FSHD resembling mitochondrial myopathy, and one patient with combined FSHD and limb girdle muscular dystrophy 2A. Detailed clinical and genetic evaluation, including 4qA/4qB allele determination, may be needed for the diagnosis of FSHD.


Assuntos
Distrofia Muscular Facioescapuloumeral/diagnóstico , Adulto , Idoso , Alelos , Cromossomos Humanos Par 4 , Diagnóstico Diferencial , Erros de Diagnóstico , Deleção de Genes , Dosagem de Genes , Humanos , Masculino , Pessoa de Meia-Idade , Miopatias Mitocondriais/diagnóstico , Distrofia Muscular do Cíngulo dos Membros/complicações , Distrofia Muscular Facioescapuloumeral/complicações , Distrofia Muscular Facioescapuloumeral/genética , Distrofia Muscular Facioescapuloumeral/patologia , Mutação , Polimorfismo Genético , Sequências de Repetição em Tandem
7.
J Med Genet ; 41(11): 826-36, 2004 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-15520407

RESUMO

BACKGROUND: Autosomal dominant facioscapulohumeral muscular dystrophy (FSHD) is associated with partial deletion of the subtelomeric D4Z4 repeat array on chromosome 4qter. This chromosomal rearrangement may result in regional chromatin relaxation and transcriptional deregulation of genes nearby. METHODS AND RESULTS: Here we describe the isolation and characterisation of FRG2, a member of a chromosomally dispersed gene family, mapping only 37 kb proximal to the D4Z4 repeat array. Homology and motif searches yielded no clues to the function of the predicted protein. FRG2 expression is undetectable in all tissues tested except for differentiating myoblasts of FSHD patients, which display low, yet distinct levels of FRG2 expression, partly from chromosome 4 but predominantly originating from its homologue on chromosome 10. However, in non-FSHD myopathy patients only distantly related FRG2 homologues are transcribed, while differentiating myoblasts from healthy controls fail to express any member of this gene family. Moreover, fibroblasts of FSHD patients and control individuals undergoing forced Ad5-MyoD mediated myogenesis show expression of FRG2 mainly originating from chromosome 10. Luciferase reporter assays show that the FRG2 promoter region can direct high levels of expression but is inhibited by increasing numbers of D4Z4 repeat units. Transient transfection experiments with FRG2 fusion-protein constructs reveal nuclear localisation and apparently FRG2 overexpression causes a wide range of morphological changes. CONCLUSION: The localisation of FRG2 genes close to the D4Z4 repeats on chromosome 4 and 10, their transcriptional upregulation specifically in FSHD myoblast cultures, potential involvement in myogenesis, and promoter properties qualify FRG2 as an attractive candidate for FSHD pathogenesis.


Assuntos
Distrofia Muscular Facioescapuloumeral/genética , Mioblastos Esqueléticos/metabolismo , Proteínas/genética , Ativação Transcricional , Sequência de Aminoácidos , Sequência de Bases , Diferenciação Celular , Células Cultivadas , Cromossomos Humanos Par 10/genética , Cromossomos Humanos Par 4/genética , Feminino , Predisposição Genética para Doença , Humanos , Masculino , Dados de Sequência Molecular , Desenvolvimento Muscular , Mioblastos Esqueléticos/química , Mioblastos Esqueléticos/citologia , Proteínas Nucleares , Regiões Promotoras Genéticas , Proteínas/análise , Proteínas/metabolismo , Regulação para Cima
9.
Neurology ; 61(7): 909-13, 2003 Oct 14.
Artigo em Inglês | MEDLINE | ID: mdl-14557558

RESUMO

OBJECTIVE: Autosomal dominant facioscapulohumeral muscular dystrophy (FSHD) is associated with a contraction of the D4Z4 repeat array on chromosome 4. So far, homozygosity or compound heterozygosity for FSHD alleles has not been described, and it has been debated whether the absence of such subjects is because of the rarity or the lethality of the disorder. METHODS: Two unrelated families in which the probands are compound heterozygous for two FSHD-sized alleles were studied. Clinical examination, pulsed-field gel electrophoresis (PFGE) studies of DNA with probes proximal and distal to D4Z4, and cytogenetic analysis of metaphase chromosomes by FISH were performed. RESULTS: Complementary molecular and cytogenetic approaches confirmed the chromosome 4qA origin of all FSHD-sized repeat arrays that segregate in the families. CONCLUSIONS: Heterozygosity for FSHD-sized alleles is compatible with life in men and women. A possible dosage effect was observed in both probands in whom each 4qA allele contributed to the FSHD phenotype. Because at least one of the FSHD alleles in both families showed an unusual low penetrance, the authors propose that susceptibility for FSHD is partly determined by intrinsic properties of the disease allele other than the residual D4Z4 repeat size alone.


Assuntos
Alelos , Cromossomos Humanos Par 4/genética , Dosagem de Genes , Distrofia Muscular Facioescapuloumeral/genética , Fenótipo , Idoso , Análise Citogenética , Eletroforese em Gel de Campo Pulsado , Feminino , Genes Dominantes , Heterozigoto , Humanos , Hibridização in Situ Fluorescente , Masculino , Linhagem , Penetrância , Sequências Repetitivas de Ácido Nucleico , Mapeamento por Restrição
10.
Neurology ; 61(2): 178-83, 2003 Jul 22.
Artigo em Inglês | MEDLINE | ID: mdl-12874395

RESUMO

BACKGROUND: The facioscapulohumeral muscular dystrophy (FSHD) locus maps to 4q35 where it is closely linked to D4F104S1 (p13E-11), a probe that recognizes the pathognomonic FSHD deletion involving the subtelomeric D4Z4 tandem repeat array. Extended deletions that include both the more proximal D4F104S1 region and the D4Z4 repeat array proper do, however, occur, albeit rarely, and such deletions can lead to difficulties of interpretation in the diagnostic setting. OBJECTIVE: To devise a means to determine the true frequency of proximally extended deletions in individuals with FSHD. METHODS: Three families selected for this study were originally identified during routine FSHD analysis on the basis that the affected individuals in each family had failed to exhibit a small (<38-kb) EcoRI fragment. High molecular weight DNA from these families was analyzed with both conventional and pulsed-field gel electrophoresis using DNA markers p13E-11, 9B6A, B31, 4qA, and 4qB. RESULTS: Large genomic deletions were identified involving both D4Z4 and D4F104S1. The precise number of D4Z4 repeat units borne by the p13E11 deletion allele was established by the use of an additional restriction enzyme (MseI) digest. All three cases carry different sizes of deletion proximal to the D4Z4 repeat units. With use of a recently described telomeric probe, 4qA, a method was developed that identifies large genomic deletions involving both D4Z4 and D4F104S1 using conventional gel electrophoresis. CONCLUSION: Proximally extended deletions can be found in patients with a normal spectrum of the disease. This assay promises to allow estimation of the true frequency of proximally extended deletions and should improve the accuracy and reliability of molecular diagnostic testing for FSHD.


Assuntos
Cromossomos Humanos Par 4/genética , Distrofia Muscular Facioescapuloumeral/genética , Deleção de Sequência , Adulto , Alelos , Criança , Análise Mutacional de DNA , Sondas de DNA , Desoxirribonuclease EcoRI , Eletroforese em Gel de Campo Pulsado , Feminino , Genes Dominantes , Marcadores Genéticos , Haplótipos/genética , Humanos , Hibridização in Situ Fluorescente , Masculino , Proteínas dos Microfilamentos , Repetições Minissatélites , Distrofia Muscular Facioescapuloumeral/diagnóstico , Proteínas Nucleares , Linhagem , Fenótipo , Polimorfismo de Fragmento de Restrição , Proteínas , Proteínas de Ligação a RNA
11.
Nat Genet ; 29(4): 383-8, 2001 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-11704758

RESUMO

Leukoencephalopathy with vanishing white matter (VWM) is an inherited brain disease that occurs mainly in children. The course is chronic-progressive with additional episodes of rapid deterioration following febrile infection or minor head trauma. We have identified mutations in EIF2B5 and EIF2B2, encoding the epsilon- and beta-subunits of the translation initiation factor eIF2B and located on chromosomes 3q27 and 14q24, respectively, as causing VWM. We found 16 different mutations in EIF2B5 in 29 patients from 23 families. We also found two distantly related individuals who were homozygous with respect to a missense mutation in EIF2B2, affecting a conserved amino acid. Three other patients also had mutations in EIF2B2. As eIF2B has an essential role in the regulation of translation under different conditions, including stress, this may explain the rapid deterioration of people with VWM under stress. Mutant translation initiation factors have not previously been implicated in disease.


Assuntos
Encefalopatias/genética , Fator de Iniciação 2B em Eucariotos/genética , Biossíntese de Proteínas/fisiologia , Sequência de Bases , Encefalopatias/patologia , Cromossomos Humanos Par 14 , Cromossomos Humanos Par 3 , Fator de Iniciação 2B em Eucariotos/fisiologia , Humanos , Dados de Sequência Molecular
12.
Hum Mol Genet ; 9(19): 2879-84, 2000 Nov 22.
Artigo em Inglês | MEDLINE | ID: mdl-11092764

RESUMO

Chromosomal rearrangements occur more frequently in subtelomeric domains than in other regions of the genome and are often associated with human pathology. To further elucidate the plasticity of subtelomeric domains, we examined the 3.3 kb D4Z4 repeat array on chromosome 4 and its homologue on chromosome 10 in 208 Dutch blood donors by pulsed field gel electrophoresis. These subtelomeric repeats are known to rearrange and partial deletions of this polymorphic array on chromosome 4 are associated with facioscapulohumeral muscular dystrophy (FSHD), an autosomal dominant myopathy. Our results show that mitotic rearrangements occur frequently as 3% of individuals display somatic mosaicism for a repeat expansion or contraction explaining the high variability of subtelomeric repeat array sizes. Translocated 4-type repeat arrays on chromosome 10 and the reverse configuration of 10-type repeat arrays on chromosome 4 are observed in 21% of individuals. The translocated repeat arrays on chromosome 4 tend to be more heterogeneous than 4-type repeats on chromosome 10. The repeat length on chromosome 4 is on average larger than on chromosome 10. But on both chromosomes we observe a multi-modal repeat length distribution with equidistant peaks at intervals of 65 kb, possibly reflecting a higher-order chromatin structure. Interestingly, in as many as six random blood donors (3%) we identified FSHD-sized 4-type repeat arrays. Assuming that these individuals are clinically unaffected, these results imply an incomplete penetrance in the upper range of FSHD alleles. Overall, the observed dynamic characteristics of these homologous domains may serve as a model for subtelomeric plasticity.


Assuntos
Cromossomos Humanos Par 10/metabolismo , Cromossomos Humanos Par 4/metabolismo , Distrofia Muscular Facioescapuloumeral/genética , Sequências Repetitivas de Ácido Nucleico/genética , Telômero/genética , Translocação Genética/genética , Cromossomos Humanos Par 10/genética , Cromossomos Humanos Par 4/genética , Eletroforese em Gel de Campo Pulsado , Feminino , Humanos , Masculino , Mitose/genética , Mosaicismo/genética , Países Baixos , Hibridização de Ácido Nucleico
13.
Cytogenet Cell Genet ; 88(3-4): 316-21, 2000.
Artigo em Inglês | MEDLINE | ID: mdl-10828619

RESUMO

The human beta-tubulin supergene family consists of several isotypes with many associated pseudogenes. Here we report the identification of yet another beta-tubulin sequence designated TUBB4Q. This tubulin maps 80 kb proximal to the facioscapulohumeral muscular dystrophy (FSHD1) associated D4Z4 repeats on chromosome 4q35. The genomic structure contains four exons encoding a putative protein of 434 amino acids. The TUBB4Q nucleotide and protein sequence show 87% and 86% homology to beta2-tubulin, respectively. Although the genomic structure shows all functional aspects of a genuine gene, no transcript could be detected. TUBB4Q-related sequences were identified on multiple chromosomes. Since these sequences mutually exhibit a high nucleotide sequence homology, they presumably belong to a novel subfamily of beta-tubulin genes. Although the chromosome 4q35 tubulin-member probably represents a pseudogene, ectopic expression due to a postulated position effect variegation (PEV), makes TUBB4Q an ideal dominant-negative candidate gene for FSHD1.


Assuntos
Cromossomos Humanos Par 4/genética , Família Multigênica/genética , Telômero/genética , Tubulina (Proteína)/genética , Alelos , Sequência de Aminoácidos , Substituição de Aminoácidos , Sequência de Bases , Clonagem Molecular , Éxons/genética , Ligação Genética/genética , Humanos , Íntrons/genética , Dados de Sequência Molecular , Distrofia Muscular Facioescapuloumeral/genética , Mapeamento Físico do Cromossomo , Reação em Cadeia da Polimerase , Regiões Promotoras Genéticas/genética , Estrutura Terciária de Proteína , Pseudogenes/genética , RNA Mensageiro/análise , RNA Mensageiro/genética , Tubulina (Proteína)/química
14.
Am J Hum Genet ; 66(1): 26-35, 2000 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-10631134

RESUMO

Autosomal dominant facioscapulohumeral muscular dystrophy (FSHD) is caused by deletion of most copies of the 3.3-kb subtelomeric D4Z4 repeat array on chromosome 4q. The molecular mechanisms behind the deletion and the high proportion of new mutations have remained elusive. We surveyed 35 de novo FSHD families and found somatic mosaicism in 40% of cases, in either the patient or an asymptomatic parent. Mosaic males were typically affected; mosaic females were more often the unaffected parent of a nonmosaic de novo patient. A genotypic-severity score, composed of the residual repeat size and the degree of somatic mosaicism, yields a consistent relationship with severity and age at onset of disease. Mosaic females had a higher proportion of somatic mosaicism than did mosaic males. The repeat deletion is significantly enhanced by supernumerary homologous repeat arrays. In 10% of normal chromosomes, 4-type repeat arrays are present on chromosome 10. In mosaic individuals, 4-type repeats on chromosome 10 are almost five times more frequent. The reverse configuration, also 10% in normal chromosomes, was not found, indicating that mutations may arise from transchromosomal interaction, to which the increase in 4-type repeat clusters is a predisposing factor. The somatic mosaicism suggests a mainly mitotic origin; mitotic interchromosomal gene conversion or translocation between fully homologous 4-type repeat arrays may be a major mechanism for FSHD mutations.


Assuntos
Cromossomos Humanos Par 10/genética , Cromossomos Humanos Par 4/genética , Mosaicismo/genética , Distrofia Muscular Facioescapuloumeral/genética , Idade de Início , DNA/análise , Eletroforese em Gel de Campo Pulsado , Feminino , Humanos , Masculino , Mitose , Linhagem , Fenótipo , Sequências Repetitivas de Ácido Nucleico , Fatores Sexuais
15.
Gene ; 240(2): 389-98, 1999 Nov 29.
Artigo em Inglês | MEDLINE | ID: mdl-10580159

RESUMO

We report the cloning of a cDNA for the mouse unconventional myosin Myo9b, the orthologue of the rat myr5 and human MYOIXb genes. A full-length spleen cDNA of 7087bp encoding a protein of 1961 amino acids was isolated. By RT-PCR, we show that Myo9b is expressed in a wide range of tissues, including heart, brain, muscle and inner ear. In addition, we have identified two alternatively spliced exons. Equivalent exons have not been previously reported for either the human or rat homologues. These exons are located in the Myo9b specific actin-binding site insert of the head domain and in the tail region. A third splice form utilizing an alternative reading frame within the 3'UTR is also described. Several polymorphisms within the coding region were identified; of interest is an in-frame 33bp imperfect duplication within the tail region that was observed only in the C57Bl/6 strain. Myo9b has been previously mapped to mouse chromosome 8 and is a candidate for the mouse mutations myodystrophy and quinky.


Assuntos
Processamento Alternativo , Miosinas/genética , Regiões 3' não Traduzidas/genética , Sequência de Aminoácidos , Animais , Sequência de Bases , Clonagem Molecular , DNA Complementar/química , DNA Complementar/genética , DNA Complementar/isolamento & purificação , Orelha Interna/embriologia , Orelha Interna/metabolismo , Regulação da Expressão Gênica no Desenvolvimento , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Endogâmicos CBA , Camundongos Endogâmicos , Dados de Sequência Molecular , Muridae , Polimorfismo Genético , Isoformas de Proteínas/genética , RNA/genética , RNA/metabolismo , Alinhamento de Sequência , Análise de Sequência de DNA , Homologia de Sequência de Aminoácidos , Distribuição Tecidual
16.
J Med Genet ; 36(11): 823-8, 1999 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-10544225

RESUMO

Facioscapulohumeral muscular dystrophy (FSHD) is caused by the size reduction of a polymorphic repeat array on 4q35. Probe p13E-11 recognises this chromosomal rearrangement and is generally used for diagnosis. However, diagnosis of FSHD is complicated by three factors. First, the probe cross hybridises to a highly homologous repeat array locus on chromosome 10q26. Second, although a BlnI polymorphism allows discrimination between the repeat units on chromosomes 4 and 10 and greatly facilitates FSHD diagnosis, the occurrence of translocations between chromosomes 4 and 10 further complicates accurate FSHD diagnosis. Third, the recent identification of deletions of p13E-11 in both control and FSHD populations is an additional complicating factor. Although pulsed field gel electrophoresis is very useful and sometimes necessary to detect these rearrangements, this technique is not operational in most FSHD diagnostic laboratories. Moreover, repeat arrays >200 kb are often difficult to detect and can falsely suggest a deletion of p13E-11. Therefore, we have developed an easy and reliable Southern blotting method to identify exchanges between 4 type and 10 type repeat arrays and deletions of p13E-11. This BglII-BlnI dosage test addresses all the above mentioned complicating factors and can be carried out in addition to the standard Southern blot analysis for FSHD diagnosis as performed in most laboratories. It will enhance the specificity and sensitivity of conventional FSHD diagnosis to the values obtained by PFGE based diagnosis of FSHD. Moreover, this study delimits the FSHD candidate gene region by mapping the 4;10 translocation breakpoint proximal to the polymorphic BlnI site in the first repeat unit.


Assuntos
Cromossomos Humanos Par 10 , Cromossomos Humanos Par 4 , Análise Citogenética , Distrofia Muscular Facioescapuloumeral/diagnóstico , Distrofia Muscular Facioescapuloumeral/genética , Translocação Genética , Southern Blotting/métodos , Humanos , Sensibilidade e Especificidade
17.
Hum Mol Genet ; 7(8): 1207-14, 1998 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-9668160

RESUMO

The autosomal dominant myopathy facioscapulohumeral muscular dystrophy (FSHD) is causally related to a short Eco RI fragment detected by probe p13E-11. This remnant fragment is the result of a deletion of an integral number of tandemly arrayed 3.3 kb repeat units (D4Z4) on 4q35. Despite intensive efforts, no transcribed sequences have been identified within this array. Previously, we have shown that these repeats on 4q35 have been exchanged for a similar highly homologous repeat locus on 10q26 in 20% of the population and that a short chromosome 10-like array on 4q35 also results in FSHD. Here, we describe the hybrid structure of some of these repeat arrays, reflecting additional sub-telomeric instability. In three healthy individuals carrying a 4-like repeat on chromosome 10 or vice versa, one repeat array was shown to consist of hybrid clusters of 4-derived and 10-derived repeat units. Moreover, employing pulsed field gel electrophoresis analysis, we identified two unrelated individuals carrying deletions of a chromosomal segment (p13E-11) proximal to the repeat locus. These deletions were not associated with FSHD. In one of these cases, however, an expansion of the deletion into the repeat array was observed in one of his children suffering from FSHD. These data provide additional evidence for instability of this sub-telomeric region and suggests that the length of the repeat, and not its intrinsic properties, is crucial to FSHD. Moreover, they are in agreement with the hypothesis that FSHD is caused by a position effect in which the repeat structure influences the expression of genes nearby. Therefore, the region deleted proximal to the repeat locus in healthy individuals can be instrumental to refine the critical region for FSHD1.


Assuntos
Cromossomos Humanos Par 4 , Rearranjo Gênico , Distrofias Musculares/diagnóstico , Distrofias Musculares/genética , Mapeamento Cromossômico , Feminino , Humanos , Hibridização in Situ Fluorescente , Masculino , Telômero/genética
18.
Mamm Genome ; 8(6): 394-8, 1997 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-9166581

RESUMO

The human autosomal dominant neuromuscular disorder facioscapulohumeral muscular dystrophy (FSHD) is associated with deletions within a complex tandem DNA repeat (D4Z4) on Chromosome (Chr) 4q35. The molecular mechanism underlying this association of FSHD with DNA rearrangements is unknown, and, thus far, no gene has been identified within the repeat. We isolated a gene mapping 100 kb proximal to D4Z4 (FSHD Region Gene 1:FRG1), but were unable to detect any alterations in total or allele-specific mRNA levels of FRG1 in FSHD patients. Human Chr 4q35 exhibits synteny homology with the region of mouse Chr 8 containing the gene for the myodystrophy mutation (myd), a possible mouse homolog of FSHD. We report the cloning of the mouse gene (Frg1) and show that it maps to mouse Chr 8. Using a cross segregating the myd mutation and the European Collaborative Interspecific Backcross, we showed that Frg1 maps proximal to the myd locus and to the Clc3 and Ant1 genes.


Assuntos
Mapeamento Cromossômico , Distrofias Musculares/genética , Mutação , Proteínas/genética , Homologia de Sequência de Aminoácidos , Sequência de Aminoácidos , Animais , Sequência de Bases , Northern Blotting , Cromossomos , Clonagem Molecular , Cricetinae , Cruzamentos Genéticos , Face , Humanos , Camundongos , Camundongos Endogâmicos C57BL , Proteínas dos Microfilamentos , Dados de Sequência Molecular , Distrofia Muscular Animal/genética , Proteínas Nucleares , Proteínas de Ligação a RNA , Distribuição Tecidual
19.
Hum Mol Genet ; 5(12): 1997-2003, 1996 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-8968754

RESUMO

Facioscapulohumeral muscular dystrophy (FSHD) is an autosomal dominant myopathy, clinically characterized by asymmetric weakness of muscles in the face, shoulder girdle and upper arm. Deletion of an integral number of 3.3 kb repeated units within a highly polymorphic EcoRI fragment at chromosome 4q35, generating a relatively short EcoRI fragment (< 35 kb), has been shown to cause FSHD1. Probe p13E-11 detects these short fragments in FSHD1 patients, and has therefore been used for diagnostic DNA analysis. However, the reliability of this analysis has been hampered by cross-hybridization of p13E-11 to chromosome 10q26-linked EcoRI fragments of comparable size, which also contain a variable number of 3.3 kb repeated units. Recently, a BinI restriction site was identified within each of the repeated units derived from chromosome 10q26, which enables differentiation of the two polymorphic p13E-11 loci in most cases without haplotype analysis. Remarkably, applying the differential analysis to screen DNA of 160 Dutch cases referred to us for FSHD1 diagnosis, we obtained evidence for subtelomeric exchange of 3.3 kb repeated units between chromosomes 4q35 and 10q26 in affected and unaffected individuals. Subsequently, analysis of 50 unrelated control samples indicated such exchange between chromosomes 4q35 and 10q26 in at least 20% of the population. These subtelomeric rearrangements have generated a novel interchromosomal polymorphism, which has implications for the specificity and sensitivity of the differential restriction analysis for diagnostic purposes. Moreover, the high frequency of the interchromosomal exchanges of 3.3 kb repeated units suggests that they probably do not contain (part of) the FSHD1 gene, and supports position effect variegation as the most likely mechanism for FSHD1.


Assuntos
Cromossomos Humanos Par 10 , Cromossomos Humanos Par 4 , Rearranjo Gênico , Distrofias Musculares/genética , Proteínas/genética , Sequências Repetitivas de Ácido Nucleico/genética , Feminino , Humanos , Masculino , Proteínas dos Microfilamentos , Proteínas Nucleares , Linhagem , Proteínas de Ligação a RNA , Telômero/genética
20.
Hum Mol Genet ; 5(5): 581-90, 1996 May.
Artigo em Inglês | MEDLINE | ID: mdl-8733123

RESUMO

Facioscapulohumeral muscular dystrophy (FSHD) is an autosomal dominant, neuromuscular disorder characterized by progressive weakness of muscles in the face, shoulder and upper arm. Deletion of integral copies of a 3.3 kb repeated unit from the subtelomeric region on chromosome 4q35 has been shown to be associated with FSHD. These repeated units which are apparently not transcribed, map very close to the 4q telomere and belong to a 3.3 kb repeat family dispersed over heterochromatic regions of the genome. Hence, position effect variegation (PEV), inducing allele-specific transcriptional repression of a gene located more centromeric, has been postulated as the underlying genetic mechanism of FSHD. This hypothesis has directed the search for the FSHD gene to the region centromeric to the repeated units. A CpG island was identified and found to be associated with the 5' untranslated region of a novel human gene, FRG1 (FSHD Region Gene 1). This evolutionary conserved gene is located about 100 kb proximal to the repeated units and belongs to a multigene family with FRG1 related sequences on multiple chromosomes. The mature chromosome 4 FRG1 transcript is 1042 bp in length and contains nine exons which encode a putative protein of 258 amino acid residues. Transcription of FRG1 was detected in several human tissues including placenta, lymphocytes, brain and muscle. To investigate a possible PEV mechanism, allele-specific FRG1 steady-state transcript levels were determined using RNA-based single-strand conformation polymorphism (SSCP) analysis. A polymorphic fragment contained within the first exon of FRG1 was amplified from reverse transcribed RNA from lymphocytes and muscle biopsies of patients and controls. No evidence for PEV mediated repression of allelic transcription was obtained in these tissues. However, detection of PEV in FSHD patients may require analysis of more specific cell types at particular developmental stages.


Assuntos
Cromossomos Humanos Par 4 , Distrofias Musculares/genética , Proteínas/genética , Alelos , Sequência de Aminoácidos , Animais , Sequência de Bases , Northern Blotting , Southern Blotting , Mapeamento Cromossômico , DNA Complementar , Regulação da Expressão Gênica , Haplorrinos , Humanos , Hibridização in Situ Fluorescente , Proteínas dos Microfilamentos , Dados de Sequência Molecular , Família Multigênica , Proteínas Nucleares , Linhagem , Reação em Cadeia da Polimerase , Polimorfismo Conformacional de Fita Simples , Proteínas de Ligação a RNA , Ratos , Mapeamento por Restrição , Ovinos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA