A large genomic deletion leads to enhancer adoption by the lamin B1 gene: a second path to autosomal dominant adult-onset demyelinating leukodystrophy (ADLD)

Détails

Ressource 1Télécharger: 25701871_BIB_683F5A0EB797.pdf (621.16 [Ko])
Etat: Public
Version: Final published version
Licence: CC BY 4.0
ID Serval
serval:BIB_683F5A0EB797
Type
Article: article d'un périodique ou d'un magazine.
Collection
Publications
Institution
Titre
A large genomic deletion leads to enhancer adoption by the lamin B1 gene: a second path to autosomal dominant adult-onset demyelinating leukodystrophy (ADLD)
Périodique
Hum Mol Genet
Auteur⸱e⸱s
Giorgio E., Robyr D., Spielmann M., Ferrero E., Di Gregorio E., Imperiale D., Vaula G., Stamoulis G., Santoni F., Atzori C., Gasparini L., Ferrera D., Canale C., Guipponi M., Pennacchio L. A., Antonarakis S. E., Brussino A., Brusco A.
ISSN
1460-2083 (Electronic)
ISSN-L
0964-6906
Statut éditorial
Publié
Date de publication
2015
Volume
24
Numéro
11
Pages
3143-54
Langue
anglais
Notes
Giorgio, Elisa
Robyr, Daniel
Spielmann, Malte
Ferrero, Enza
Di Gregorio, Eleonora
Imperiale, Daniele
Vaula, Giovanna
Stamoulis, Georgios
Santoni, Federico
Atzori, Cristiana
Gasparini, Laura
Ferrera, Denise
Canale, Claudio
Guipponi, Michel
Pennacchio, Len A
Antonarakis, Stylianos E
Brussino, Alessandro
Brusco, Alfredo
eng
GGP10184/Telethon/Italy
R01 HG003988/HG/NHGRI NIH HHS/
HG003988/HG/NHGRI NIH HHS/
U54HG006997/HG/NHGRI NIH HHS/
Research Support, N.I.H., Extramural
Research Support, U.S. Gov't, Non-P.H.S.
England
Hum Mol Genet. 2015 Jun 1;24(11):3143-54. doi: 10.1093/hmg/ddv065. Epub 2015 Feb 20.
Résumé
Chromosomal rearrangements with duplication of the lamin B1 (LMNB1) gene underlie autosomal dominant adult-onset demyelinating leukodystrophy (ADLD), a rare neurological disorder in which overexpression of LMNB1 causes progressive central nervous system demyelination. However, we previously reported an ADLD family (ADLD-1-TO) without evidence of duplication or other mutation in LMNB1 despite linkage to the LMNB1 locus and lamin B1 overexpression. By custom array-CGH, we further investigated this family and report here that patients carry a large ( approximately 660 kb) heterozygous deletion that begins 66 kb upstream of the LMNB1 promoter. Lamin B1 overexpression was confirmed in further ADLD-1-TO tissues and in a postmortem brain sample, where lamin B1 was increased in the frontal lobe. Through parallel studies, we investigated both loss of genetic material and chromosomal rearrangement as possible causes of LMNB1 overexpression, and found that ADLD-1-TO plausibly results from an enhancer adoption mechanism. The deletion eliminates a genome topological domain boundary, allowing normally forbidden interactions between at least three forebrain-directed enhancers and the LMNB1 promoter, in line with the observed mainly cerebral localization of lamin B1 overexpression and myelin degeneration. This second route to LMNB1 overexpression and ADLD is a new example of the relevance of regulatory landscape modifications in determining Mendelian phenotypes.
Mots-clé
Animals, Base Sequence, Cells, Cultured, DNA Mutational Analysis, *Enhancer Elements, Genetic, Female, Gene Expression, Gene Expression Regulation, Genetic Association Studies, Humans, Lamin Type B/*genetics/metabolism, Male, Mice, Transgenic, Middle Aged, Molecular Sequence Data, Pedigree, Pelizaeus-Merzbacher Disease/*genetics, *Sequence Deletion
Pubmed
Création de la notice
20/05/2019 13:52
Dernière modification de la notice
13/01/2021 8:09
Données d'usage