↓ Skip to main content

PLOS

Epigenetic Characterization of the FMR1 Gene and Aberrant Neurodevelopment in Human Induced Pluripotent Stem Cell Models of Fragile X Syndrome

Overview of attention for article published in PLOS ONE, October 2011
Altmetric Badge

Mentioned by

twitter
2 X users
patent
2 patents
facebook
3 Facebook pages
wikipedia
1 Wikipedia page

Citations

dimensions_citation
281 Dimensions

Readers on

mendeley
322 Mendeley
Title
Epigenetic Characterization of the FMR1 Gene and Aberrant Neurodevelopment in Human Induced Pluripotent Stem Cell Models of Fragile X Syndrome
Published in
PLOS ONE, October 2011
DOI 10.1371/journal.pone.0026203
Pubmed ID
Authors

Steven D. Sheridan, Kraig M. Theriault, Surya A. Reis, Fen Zhou, Jon M. Madison, Laurence Daheron, Jeanne F. Loring, Stephen J. Haggarty

Abstract

Fragile X syndrome (FXS) is the most common inherited cause of intellectual disability. In addition to cognitive deficits, FXS patients exhibit hyperactivity, attention deficits, social difficulties, anxiety, and other autistic-like behaviors. FXS is caused by an expanded CGG trinucleotide repeat in the 5' untranslated region of the Fragile X Mental Retardation (FMR1) gene leading to epigenetic silencing and loss of expression of the Fragile X Mental Retardation protein (FMRP). Despite the known relationship between FMR1 CGG repeat expansion and FMR1 silencing, the epigenetic modifications observed at the FMR1 locus, and the consequences of the loss of FMRP on human neurodevelopment and neuronal function remain poorly understood. To address these limitations, we report on the generation of induced pluripotent stem cell (iPSC) lines from multiple patients with FXS and the characterization of their differentiation into post-mitotic neurons and glia. We show that clones from reprogrammed FXS patient fibroblast lines exhibit variation with respect to the predominant CGG-repeat length in the FMR1 gene. In two cases, iPSC clones contained predominant CGG-repeat lengths shorter than measured in corresponding input population of fibroblasts. In another instance, reprogramming a mosaic patient having both normal and pre-mutation length CGG repeats resulted in genetically matched iPSC clonal lines differing in FMR1 promoter CpG methylation and FMRP expression. Using this panel of patient-specific, FXS iPSC models, we demonstrate aberrant neuronal differentiation from FXS iPSCs that is directly correlated with epigenetic modification of the FMR1 gene and a loss of FMRP expression. Overall, these findings provide evidence for a key role for FMRP early in human neurodevelopment prior to synaptogenesis and have implications for modeling of FXS using iPSC technology. By revealing disease-associated cellular phenotypes in human neurons, these iPSC models will aid in the discovery of novel therapeutics for FXS and other autism-spectrum disorders sharing common pathophysiology.

X Demographics

X Demographics

The data shown below were collected from the profiles of 2 X users who shared this research output. Click here to find out more about how the information was compiled.
Mendeley readers

Mendeley readers

The data shown below were compiled from readership statistics for 322 Mendeley readers of this research output. Click here to see the associated Mendeley record.

Geographical breakdown

Country Count As %
Portugal 3 <1%
Brazil 3 <1%
United States 2 <1%
Italy 1 <1%
United Kingdom 1 <1%
Australia 1 <1%
Mexico 1 <1%
Canada 1 <1%
Unknown 309 96%

Demographic breakdown

Readers by professional status Count As %
Student > Ph. D. Student 70 22%
Researcher 59 18%
Student > Bachelor 49 15%
Student > Master 37 11%
Student > Doctoral Student 17 5%
Other 56 17%
Unknown 34 11%
Readers by discipline Count As %
Agricultural and Biological Sciences 103 32%
Biochemistry, Genetics and Molecular Biology 67 21%
Neuroscience 41 13%
Medicine and Dentistry 26 8%
Psychology 14 4%
Other 27 8%
Unknown 44 14%