81
views
0
recommends
+1 Recommend
0 collections
    0
    shares
      • Record: found
      • Abstract: found
      • Article: found
      Is Open Access

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

      research-article

      Read this article at

      Bookmark
          There is no author summary for this article yet. Authors can add summaries to their articles on ScienceOpen to make them more accessible to a non-specialist audience.

          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.

          Related collections

          Most cited references37

          • Record: found
          • Abstract: found
          • Article: not found

          Reprogramming of human somatic cells to pluripotency with defined factors.

          Pluripotency pertains to the cells of early embryos that can generate all of the tissues in the organism. Embryonic stem cells are embryo-derived cell lines that retain pluripotency and represent invaluable tools for research into the mechanisms of tissue formation. Recently, murine fibroblasts have been reprogrammed directly to pluripotency by ectopic expression of four transcription factors (Oct4, Sox2, Klf4 and Myc) to yield induced pluripotent stem (iPS) cells. Using these same factors, we have derived iPS cells from fetal, neonatal and adult human primary cells, including dermal fibroblasts isolated from a skin biopsy of a healthy research subject. Human iPS cells resemble embryonic stem cells in morphology and gene expression and in the capacity to form teratomas in immune-deficient mice. These data demonstrate that defined factors can reprogramme human cells to pluripotency, and establish a method whereby patient-specific cells might be established in culture.
            Bookmark
            • Record: found
            • Abstract: found
            • Article: not found

            Elevated levels of FMR1 mRNA in carrier males: a new mechanism of involvement in the fragile-X syndrome.

            Fragile-X syndrome is a trinucleotide-repeat-expansion disorder in which the clinical phenotype is believed to result from transcriptional silencing of the fragile-X mental retardation 1 (FMR1) gene as the number of CGG repeats exceeds approximately 200. For premutation alleles ( approximately 55-200 repeats), no abnormalities in FMR1-gene expression have been described, despite growing evidence of clinical involvement in premutation carriers. To address this (apparent) paradox, we have determined, for 16 carrier males (55-192 repeats), the relative levels of leukocyte FMR1 mRNA, by use of automated fluorescence-detection reverse transcriptase-PCR, and the percent of lymphocytes that are immunoreactive for FMR1 protein (FMRP). For some alleles with>100 repeats, there was a reduction in the number of FMRP-positive cells. Unexpectedly, FMR1 mRNA levels were elevated at least fivefold within this same range. No significant increase in FMR1 mRNA stability was observed in a lymphoblastoid cell line (160 repeats) derived from one of the carrier males, suggesting that the increased message levels are due to an increased rate of transcription. Current results support a mechanism of involvement in premutation carriers, in which reduced translational efficiency is at least partially compensated through increased transcriptional activity. Thus, diminished translational efficiency may be important throughout much of the premutation range, with a mechanistic switch occurring in the full-mutation range as the FMR1 gene is silenced.
              Bookmark
              • Record: found
              • Abstract: found
              • Article: not found

              Advances in the treatment of fragile X syndrome.

              The FMR1 mutations can cause a variety of disabilities, including cognitive deficits, attention-deficit/hyperactivity disorder, autism, and other socioemotional problems, in individuals with the full mutation form (fragile X syndrome) and distinct difficulties, including primary ovarian insufficiency, neuropathy and the fragile X-associated tremor/ataxia syndrome, in some older premutation carriers. Therefore, multigenerational family involvement is commonly encountered when a proband is identified with a FMR1 mutation. Studies of metabotropic glutamate receptor 5 pathway antagonists in animal models of fragile X syndrome have demonstrated benefits in reducing seizures, improving behavior, and enhancing cognition. Trials of metabotropic glutamate receptor 5 antagonists are beginning with individuals with fragile X syndrome. Targeted treatments, medical and behavioral interventions, genetic counseling, and family supports are reviewed here.
                Bookmark

                Author and article information

                Contributors
                Role: Editor
                Journal
                PLoS One
                plos
                plosone
                PLoS ONE
                Public Library of Science (San Francisco, USA )
                1932-6203
                2011
                12 October 2011
                : 6
                : 10
                : e26203
                Affiliations
                [1 ]Center for Human Genetic Research, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, United States of America
                [2 ]Stanley Center for Psychiatric Research, Broad Institute of Harvard and Massachusetts Institute of Technology, Cambridge, Massachusetts, United States of America
                [3 ]Harvard Stem Cell Institute, Harvard University, Cambridge, Massachusetts, United States of America
                [4 ]Center for Regenerative Medicine, Massachusetts General Hospital, Boston, Massachusetts, United States of America
                [5 ]Center for Regenerative Medicine, Department of Chemical Physiology, The Scripps Research Institute, La Jolla, California, United States of America
                National Institutes of Health, United States of America
                Author notes

                Conceived and designed the experiments: SS KT SR SH. Performed the experiments: SS KT FZ. Analyzed the data: SS KT SH SR JL. Contributed reagents/materials/analysis tools: SS KT FZ JM SH. Wrote the paper: SS KT JL SH. Coordinated the study: SS. Performed iPSC characterization, neural differentiation and phenotypic characterization: SS. Performed iPSC characterization, neural differentiation and phenotypic characterization, performed iPSC characterization, FMR1 promoter methylation and expression analysis, FMRP western analysis: KT. Derived a control iPSC line used in the study and contributed advice on characterizing differentiated neurons: FZ JM. Contributed unpublished data on other FXS iPSC lines: JL. Provided technical guidance, training and reagents for iPSC derivation: LD. Contributed financial support: SH.

                Article
                PONE-D-11-13642
                10.1371/journal.pone.0026203
                3192166
                22022567
                f724245e-48a3-4c57-8e78-c9dd1b537d2c
                Sheridan et al. This is an open-access article distributed under the terms of the Creative Commons Attribution License, which permits unrestricted use, distribution, and reproduction in any medium, provided the original author and source are credited.
                History
                : 17 July 2011
                : 22 September 2011
                Page count
                Pages: 13
                Categories
                Research Article
                Biology
                Developmental Biology
                Stem Cells
                Adult Stem Cells
                Cell Potency
                Embryonic Stem Cells
                Induced Pluripotent Stem Cells
                Neural Stem Cells
                Stem Cell Lines
                Cell Differentiation
                Cell Fate Determination
                Genomic Imprinting
                Genetics
                Epigenetics
                DNA modification
                Gene Function
                Neuroscience
                Cellular Neuroscience
                Neuronal Morphology
                Developmental Neuroscience
                Neural Stem Cells
                Neurogenesis
                Neuroglial Development
                Medicine
                Neurology
                Huntington Disease
                Neuropharmacology

                Uncategorized
                Uncategorized

                Comments

                Comment on this article