Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 8 de 8
Filtrar
1.
Hum Genet ; 141(8): 1385-1407, 2022 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-35089417

RESUMO

Coloboma, a congenital disorder characterized by gaps in ocular tissues, is caused when the choroid fissure fails to close during embryonic development. Several loci have been associated with coloboma, but these represent less than 40% of those that are involved with this disease. Here, we describe a novel coloboma-causing locus, BMP3. Whole exome sequencing and Sanger sequencing of patients with coloboma identified three variants in BMP3, two of which are predicted to be disease causing. Consistent with this, bmp3 mutant zebrafish have aberrant fissure closure. bmp3 is expressed in the ventral head mesenchyme and regulates phosphorylated Smad3 in a population of cells adjacent to the choroid fissure. Furthermore, mutations in bmp3 sensitize embryos to Smad3 inhibitor treatment resulting in open choroid fissures. Micro CT scans and Alcian blue staining of zebrafish demonstrate that mutations in bmp3 cause midface hypoplasia, suggesting that bmp3 regulates cranial neural crest cells. Consistent with this, we see active Smad3 in a population of periocular neural crest cells, and bmp3 mutant zebrafish have reduced neural crest cells in the choroid fissure. Taken together, these data suggest that Bmp3 controls Smad3 phosphorylation in neural crest cells to regulate early craniofacial and ocular development.


Assuntos
Coloboma , Animais , Coloboma/genética , Olho , Crista Neural , Peixe-Zebra/genética , Proteínas de Peixe-Zebra/genética
2.
Hum Mol Genet ; 22(7): 1432-42, 2013 Apr 01.
Artigo em Inglês | MEDLINE | ID: mdl-23307924

RESUMO

Retinal dystrophies are predominantly caused by mutations affecting the visual phototransduction system and cilia, with few genes identified that function to maintain photoreceptor survival. We reasoned that growth factors involved with early embryonic retinal development would represent excellent candidates for such diseases. Here we show that mutations in the transforming growth factor-ß (TGF-ß) ligand Growth Differentiation Factor 6, which specifies the dorso-ventral retinal axis, contribute to Leber congenital amaurosis. Furthermore, deficiency of gdf6 results in photoreceptor degeneration, so demonstrating a connection between Gdf6 signaling and photoreceptor survival. In addition, in both murine and zebrafish mutant models, we observe retinal apoptosis, a characteristic feature of human retinal dystrophies. Treatment of gdf6-deficient zebrafish embryos with a novel aminopropyl carbazole, P7C3, rescued the retinal apoptosis without evidence of toxicity. These findings implicate for the first time perturbed TGF-ß signaling in the genesis of retinal dystrophies, support the study of related morphogenetic genes for comparable roles in retinal disease and may offer additional therapeutic opportunities for genetically heterogeneous disorders presently only treatable with gene therapy.


Assuntos
Sobrevivência Celular , Fator 6 de Diferenciação de Crescimento/genética , Amaurose Congênita de Leber/genética , Retinose Pigmentar/genética , Sequência de Aminoácidos , Animais , Apoptose , Análise Mutacional de DNA , Estudos de Associação Genética , Fator 6 de Diferenciação de Crescimento/fisiologia , Humanos , Amaurose Congênita de Leber/patologia , Camundongos , Camundongos Transgênicos , Dados de Sequência Molecular , Mutação de Sentido Incorreto , Linhagem , Células Fotorreceptoras de Vertebrados/metabolismo , Células Fotorreceptoras de Vertebrados/fisiologia , Retina/patologia , Retinose Pigmentar/patologia , Peixe-Zebra
3.
Transl Anim Sci ; 7(1): txad029, 2023 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-36970312

RESUMO

The mature peptide of growth differentiation factor 11 (GDF11) in Bos taurus breeds, shares 90% amino acid sequence similarity to myostatin (MSTN), where loss-of-function mutations result in muscular hyperplasia causing a phenotype known as double-muscling. Mutations in the MSTN coding sequence increase muscle mass and reduce fat and bone tissues, but also confer poor fertility, reduced stress tolerance, and increased calf mortality. GDF11 influences skeletal muscle development in mice, and muscular atrophy can be induced by exogenous GDF11 treatment. To date, there are no reports of GDF11's role in bovine carcass traits. To determine associations between GDF11 and carcass quality in beef cattle, bovine GDF11 was examined in crossbred Canadian beef cattle populations during finishing. Few coding variants were found in this functionally important gene, but an upstream variant c.1-1951C > T (rs136619751) with a minor allele frequency of 0.31 was identified and further genotyped in two separate populations of crossbred steers (n = 415 and 450). CC animals had lower backfat thickness, marbling percentage, and yield score than CT or TT animals (P < 0.001 and < 0.05). These data suggest a role of GDF11 in carcass quality in beef cattle and may provide a selection tool to improve carcass traits in cattle.

4.
Hum Mol Genet ; 19(2): 287-98, 2010 Jan 15.
Artigo em Inglês | MEDLINE | ID: mdl-19864492

RESUMO

Ocular mal-development results in heterogeneous and frequently visually disabling phenotypes that include coloboma and microphthalmia. Due to the contribution of bone morphogenetic proteins to such processes, the function of the paralogue Growth Differentiation Factor 3 was investigated. Multiple mis-sense variants were identified in patients with ocular and/or skeletal (Klippel-Feil) anomalies including one individual with heterozygous alterations in GDF3 and GDF6. These variants were characterized, individually and in combination, through integrated biochemical and zebrafish model organism analyses, demonstrating appreciable effects with western blot analyses, luciferase based reporter assays and antisense morpholino inhibition. Notably, inhibition of the zebrafish co-orthologue of GDF3 accurately recapitulates patient phenotypes. By demonstrating the pleiotropic effects of GDF3 mutation, these results extend the contribution of perturbed BMP signaling to human disease and potentially implicate multi-allelic inheritance of BMP variants in developmental disorders.


Assuntos
Anormalidades do Olho/genética , Fator 3 de Diferenciação de Crescimento/genética , Músculo Esquelético/anormalidades , Mutação , Sequência de Aminoácidos , Animais , Linhagem Celular , Anormalidades do Olho/metabolismo , Feminino , Fator 3 de Diferenciação de Crescimento/química , Fator 3 de Diferenciação de Crescimento/metabolismo , Humanos , Masculino , Dados de Sequência Molecular , Músculo Esquelético/metabolismo , Linhagem , Alinhamento de Sequência , Fator de Crescimento Transformador beta/genética , Fator de Crescimento Transformador beta/metabolismo , Peixe-Zebra/genética , Peixe-Zebra/crescimento & desenvolvimento , Peixe-Zebra/metabolismo , Proteínas de Peixe-Zebra/genética , Proteínas de Peixe-Zebra/metabolismo
5.
Hum Mol Genet ; 18(6): 1110-21, 2009 Mar 15.
Artigo em Inglês | MEDLINE | ID: mdl-19129173

RESUMO

Proteins of the bone morphogenetic protein (BMP) family are known to have a role in ocular and skeletal development; however, because of their widespread expression and functional redundancy, less progress has been made identifying the roles of individual BMPs in human disease. We identified seven heterozygous mutations in growth differentiation factor 6 (GDF6), a member of the BMP family, in patients with both ocular and vertebral anomalies, characterized their effects with a SOX9-reporter assay and western analysis, and demonstrated comparable phenotypes in model organisms with reduced Gdf6 function. We observed a spectrum of ocular and skeletal anomalies in morphant zebrafish, the latter encompassing defective tail formation and altered expression of somite markers noggin1 and noggin2. Gdf6(+/-) mice exhibited variable ocular phenotypes compatible with phenotypes observed in patients and zebrafish. Key differences evident between patients and animal models included pleiotropic effects, variable expressivity and incomplete penetrance. These data establish the important role of this determinant in ocular and vertebral development, demonstrate the complex genetic inheritance of these phenotypes, and further understanding of BMP function and its contributions to human disease.


Assuntos
Anormalidades Múltiplas/genética , Anormalidades Múltiplas/patologia , Fator 6 de Diferenciação de Crescimento/genética , Penetrância , Sequência de Aminoácidos , Animais , Análise Mutacional de DNA , Genes Reporter , Fator 6 de Diferenciação de Crescimento/química , Humanos , Camundongos , Modelos Animais , Dados de Sequência Molecular , Proteínas Mutantes/química , Proteínas Mutantes/genética , Mutação/genética , Oligonucleotídeos Antissenso/farmacologia , Peixe-Zebra , Proteínas de Peixe-Zebra/química , Proteínas de Peixe-Zebra/genética
6.
Hum Mol Genet ; 17(22): 3446-58, 2008 Nov 15.
Artigo em Inglês | MEDLINE | ID: mdl-18694899

RESUMO

The factors that mediate chromosomal rearrangement remain incompletely defined. Among regions prone to structural variant formation, chromosome 6p25 is one of the few in which disease-associated segmental duplications and segmental deletions have been identified, primarily through gene dosage attributable ocular phenotypes. Using array comparative genome hybridization, we studied ten 6p25 duplication and deletion pedigrees and amplified junction fragments from each. Analysis of the breakpoint architecture revealed that all the rearrangements were non-recurrent, and in contrast to most previous examples the majority of the segmental duplications and deletions utilized coupled homologous and non-homologous recombination mechanisms. One junction fragment exhibited an unprecedented 367 bp insert derived from tandemly arranged breakpoint elements. While this accorded with a recently described replication-based mechanism, it differed from the previous example in being unassociated with template switching, and occurring in a segmental deletion. These results extend the mechanisms involved in structural variant formation, provide strong evidence that a spectrum of recombination, DNA repair and replication underlie 6p25 rearrangements, and have implications for genesis of copy number variations in other genomic regions. These findings highlight the benefits of undertaking the extensive studies necessary to characterize structural variants at the base pair level.


Assuntos
Aberrações Cromossômicas , Cromossomos Humanos Par 6/genética , Dosagem de Genes , Glaucoma/genética , Quebra Cromossômica , Deleção Cromossômica , Fatores de Transcrição Forkhead , Haplótipos , Humanos , Hibridização de Ácido Nucleico , Análise de Sequência com Séries de Oligonucleotídeos , Fenótipo , Polimorfismo de Nucleotídeo Único , Recombinação Genética
7.
Invest Ophthalmol Vis Sci ; 47(11): 4905-9, 2006 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-17065505

RESUMO

PURPOSE: Axenfeld-Rieger malformations of the anterior segment are clinically heterogeneous, and up to 50% of cases are attributable to PITX2 or FOXC1 mutation. In view of PITX2's contribution to corneal development and the altered CCT in some FOXC1-related cases, this study was undertaken to investigate whether a related phenotype is associated with the PITX2/Pitx2 mutation. METHODS: Central corneal thickness (CCT) was measured in patients and mice with PITX2/Pitx2 mutations. CCT in affected individuals and unaffected first-degree relatives from a large PITX2 mutation pedigree was measured with ultrasonic pachymetry. For murine measurements, the optical coherence tomogram (OCT) was calibrated against plastic films whose thickness had been determined with scanning electron microscopy (SEM). Subsequently, CCT was measured in ex vivo eyes from Pitx2(+/-) and wild-type murine littermates by using OCT. RESULTS: CCT in individuals with the PITX2 mutation (mean 484 microm; range, 425-519; n = 8) was significantly lower than in their unaffected first-degree relatives (mean 582 microm; range, 550-590; n = 5; P = 0.0002, t-test). Scanning electron microscopy (SEM) and OCT measurements of reference films correlated closely (r = 0.9995) and subsequent OCT analysis of murine eyes revealed a significant reduction in CCT in Pitx2(+/-) compared with wild-type littermates (Pitx2(+/-): mean, 72 microm; range, 57-87, n = 6; wt: mean, 88 microm; range, 63-100; n = 6, P = 0.035, t-test). CONCLUSIONS: The results show that PITX2/Pitx2 mutation results in reduced corneal thickness and provides the first example of reduced CCT in a genetic subtype of glaucoma. These data will facilitate management of developmental glaucoma and offer potential for guiding molecular genetic testing in patients with Axenfeld-Rieger. The similar CCT reduction observed in patients and mice with comparable mutations emphasizes the utility of this murine model. The technical advance of optical murine CCT measurement also provides scope for serial in vivo imaging of the developing anterior segment and determining the effects of altered CCT on measured IOP.


Assuntos
Córnea/anormalidades , Córnea/patologia , Anormalidades do Olho/diagnóstico , Glaucoma/diagnóstico , Proteínas de Homeodomínio/genética , Mutação , Fatores de Transcrição/genética , Animais , Pesos e Medidas Corporais , Córnea/diagnóstico por imagem , Anormalidades do Olho/genética , Glaucoma/genética , Humanos , Camundongos , Camundongos Endogâmicos C57BL , Microscopia Eletrônica de Varredura , Síndrome , Tomografia de Coerência Óptica , Ultrassonografia , Proteína Homeobox PITX2
8.
Am J Hum Genet ; 80(2): 306-15, 2007 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-17236135

RESUMO

Colobomata represent visually impairing ocular closure defects that are associated with a diverse range of developmental anomalies. Characterization of a chromosome 8q21.2-q22.1 segmental deletion in a patient with chorioretinal coloboma revealed elements of nonallelic homologous recombination and nonhomologous end joining. This genomic architecture extends the range of chromosomal rearrangements associated with human disease and indicates that a broader spectrum of human chromosomal rearrangements may use coupled homologous and nonhomologous mechanisms. We also demonstrate that the segmental deletion encompasses GDF6, encoding a member of the bone-morphogenetic protein family, and that inhibition of gdf6a in a model organism accurately recapitulates the proband's phenotype. The spectrum of disorders generated by morpholino inhibition and the more severe defects (microphthalmia and anophthalmia) observed at higher doses illustrate the key role of GDF6 in ocular development. These results underscore the value of integrated clinical and molecular investigation of patients with chromosomal anomalies.


Assuntos
Proteínas Morfogenéticas Ósseas/genética , Aberrações Cromossômicas , Coloboma/genética , Predisposição Genética para Doença , Recombinação Genética , Animais , Proteínas Morfogenéticas Ósseas/metabolismo , Cromossomos Humanos Par 8/genética , Modelos Animais de Doenças , Embrião não Mamífero/metabolismo , Feminino , Fator 6 de Diferenciação de Crescimento , Humanos , Cariotipagem , Masculino , Fenótipo , Peixe-Zebra/embriologia , Peixe-Zebra/genética , Proteínas de Peixe-Zebra/genética , Proteínas de Peixe-Zebra/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA