Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 4 de 4
Filtrar
1.
Life Sci Alliance ; 7(5)2024 May.
Artigo em Inglês | MEDLINE | ID: mdl-38418088

RESUMO

Detecting structural variants (SVs) in whole-genome sequencing poses significant challenges. We present a protocol for variant calling, merging, genotyping, sensitivity analysis, and laboratory validation for generating a high-quality SV call set in whole-genome sequencing from the Alzheimer's Disease Sequencing Project comprising 578 individuals from 111 families. Employing two complementary pipelines, Scalpel and Parliament, for SV/indel calling, we assessed sensitivity through sample replicates (N = 9) with in silico variant spike-ins. We developed a novel metric, D-score, to evaluate caller specificity for deletions. The accuracy of deletions was evaluated by Sanger sequencing. We generated a high-quality call set of 152,301 deletions of diverse sizes. Sanger sequencing validated 114 of 146 detected deletions (78.1%). Scalpel excelled in accuracy for deletions ≤100 bp, whereas Parliament was optimal for deletions >900 bp. Overall, 83.0% and 72.5% of calls by Scalpel and Parliament were validated, respectively, including all 11 deletions called by both Parliament and Scalpel between 101 and 900 bp. Our flexible protocol successfully generated a high-quality deletion call set and a truth set of Sanger sequencing-validated deletions with precise breakpoints spanning 1-17,000 bp.


Assuntos
Doença de Alzheimer , Humanos , Doença de Alzheimer/genética , Sequenciamento Completo do Genoma/métodos
2.
Sci Rep ; 9(1): 14166, 2019 10 02.
Artigo em Inglês | MEDLINE | ID: mdl-31578364

RESUMO

Congenital stationary night blindness (CSNB), in the complete form, is caused by dysfunctions in ON-bipolar cells (ON-BCs) which are secondary neurons of the retina. We describe the first disease causative variant associated with CSNB in the dog. A genome-wide association study using 12 cases and 11 controls from a research colony determined a 4.6 Mb locus on canine chromosome 32. Subsequent whole-genome sequencing identified a 1 bp deletion in LRIT3 segregating with CSNB. The canine mutant LRIT3 gives rise to a truncated protein with unaltered subcellular expression in vitro. Genetic variants in LRIT3 have been associated with CSNB in patients although there is limited evidence regarding its apparently critical function in the mGluR6 pathway in ON-BCs. We determine that in the canine CSNB retina, the mutant LRIT3 is correctly localized to the region correlating with the ON-BC dendritic tips, albeit with reduced immunolabelling. The LRIT3-CSNB canine model has direct translational potential enabling studies to help understand the CSNB pathogenesis as well as to develop new therapies targeting the secondary neurons of the retina.


Assuntos
Doenças do Cão/genética , Oftalmopatias Hereditárias/veterinária , Deleção de Genes , Doenças Genéticas Ligadas ao Cromossomo X/veterinária , Proteínas de Membrana/genética , Miopia/veterinária , Cegueira Noturna/veterinária , Animais , Cromossomos/genética , Cães , Oftalmopatias Hereditárias/genética , Feminino , Doenças Genéticas Ligadas ao Cromossomo X/genética , Estudo de Associação Genômica Ampla , Heterozigoto , Masculino , Proteínas de Membrana/metabolismo , Miopia/genética , Cegueira Noturna/genética , Retina/metabolismo , Retina/patologia , Sequenciamento Completo do Genoma
3.
Sci Rep ; 8(1): 13058, 2018 Aug 24.
Artigo em Inglês | MEDLINE | ID: mdl-30139995

RESUMO

A correction to this article has been published and is linked from the HTML and PDF versions of this paper. The error has not been fixed in the paper.

4.
Sci Rep ; 7(1): 12823, 2017 10 09.
Artigo em Inglês | MEDLINE | ID: mdl-28993665

RESUMO

Defects in the cilia gene RPGRIP1 cause Leber congenital amaurosis and cone-rod dystrophy in humans. A form of canine cone-rod dystrophy (cord1) was originally associated with a homozygous insertion in RPGRIP1 (RPGRIP1 ins/ins) as the primary disease locus while a homozygous deletion in MAP9 (MAP9 del/del) was later identified as a modifier associated with the early onset form. However, we find further variability in cone electroretinograms (ERGs) ranging from normal to absent in an extended RPGRIP1 ins/ins canine colony, irrespective of the MAP9 genotype. Ophthalmoscopically, cone ERGabsent RPGRIP1 ins/ins eyes show discolouration of the tapetal fundus with varying onset and disease progression, while sd-OCT reveals atrophic changes. Despite marked changes in cone ERG and retinal morphology, photopic vision-guided behaviour is comparable between normal and cone ERGabsent RPGRIP1 ins/ins littermates. Cone morphology of the dogs lacking cone ERG are truncated with shortened outer and inner segments. Immunohistochemically, cone ERGabsent RPGRIP1 ins/ins retinas have extensive L/M-opsin mislocalization, lack CNGB3 labelling in the L/M-cones, and lack GC1 in all cones. Our results indicate that cord1 is a multigenic disease in which mutations in neither RPGRIP1 nor MAP9 alone lead to visual deficits, and additional gene(s) contribute to cone-specific functional and morphologic defects.


Assuntos
Distrofias de Cones e Bastonetes/genética , Distrofias de Cones e Bastonetes/fisiopatologia , Proteínas do Olho/metabolismo , Herança Multifatorial/genética , Retina/patologia , Retina/fisiopatologia , Animais , Comportamento Animal , Dendritos/metabolismo , Modelos Animais de Doenças , Cães , Eletrorretinografia , Proteínas do Olho/genética , Feminino , Regulação da Expressão Gênica , Masculino , Linhagem , Transporte Proteico , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Retina/metabolismo , Células Fotorreceptoras Retinianas Cones/metabolismo , Segmento Externo das Células Fotorreceptoras da Retina/metabolismo , Células Fotorreceptoras Retinianas Bastonetes/metabolismo , Opsinas de Bastonetes/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA