Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 5 de 5
Filter
1.
J Biol Chem ; 294(6): 1846-1859, 2019 02 08.
Article in English | MEDLINE | ID: mdl-30552117

ABSTRACT

Friedreich ataxia (FRDA) is a neurodegenerative disorder caused by transcriptional silencing of the frataxin (FXN) gene, resulting in loss of the essential mitochondrial protein frataxin. Based on the knowledge that a GAA·TTC repeat expansion in the first intron of FXN induces heterochromatin, we previously showed that 2-aminobenzamide-type histone deacetylase inhibitors (HDACi) increase FXN mRNA levels in induced pluripotent stem cell (iPSC)-derived FRDA neurons and in circulating lymphocytes from patients after HDACi oral administration. How the reduced expression of frataxin leads to neurological and other systemic symptoms in FRDA patients remains unclear. Similar to other triplet-repeat disorders, it is unknown why FRDA affects only specific cell types, primarily the large sensory neurons of the dorsal root ganglia and cardiomyocytes. The combination of iPSC technology and genome-editing techniques offers the unique possibility to address these questions in a relevant cell model of FRDA, obviating confounding effects of variable genetic backgrounds. Here, using "scarless" gene-editing methods, we created isogenic iPSC lines that differ only in the length of the GAA·TTC repeats. To uncover the gene expression signatures due to the GAA·TTC repeat expansion in FRDA neuronal cells and the effect of HDACi on these changes, we performed RNA-seq-based transcriptomic analysis of iPSC-derived central nervous system (CNS) and isogenic sensory neurons. We found that cellular pathways related to neuronal function, regulation of transcription, extracellular matrix organization, and apoptosis are affected by frataxin loss in neurons of the CNS and peripheral nervous system and that these changes are partially restored by HDACi treatment.


Subject(s)
Friedreich Ataxia/genetics , Histone Deacetylase Inhibitors/pharmacology , Neurons/pathology , Transcriptome , Cells, Cultured , Friedreich Ataxia/pathology , Gene Editing/methods , Gene Expression Profiling , Humans , Induced Pluripotent Stem Cells/chemistry , Iron-Binding Proteins/genetics , Iron-Binding Proteins/metabolism , Neurons/chemistry , Trinucleotide Repeat Expansion/genetics , Frataxin
2.
Ann Neurol ; 76(4): 489-508, 2014 Oct.
Article in English | MEDLINE | ID: mdl-25159818

ABSTRACT

OBJECTIVE: To investigate whether a histone deacetylase inhibitor (HDACi) would be effective in an in vitro model for the neurodegenerative disease Friedreich ataxia (FRDA) and to evaluate safety and surrogate markers of efficacy in a phase I clinical trial in patients. METHODS: We used a human FRDA neuronal cell model, derived from patient induced pluripotent stem cells, to determine the efficacy of a 2-aminobenzamide HDACi (109) as a modulator of FXN gene expression and chromatin histone modifications. FRDA patients were dosed in 4 cohorts, ranging from 30mg/day to 240mg/day of the formulated drug product of HDACi 109, RG2833. Patients were monitored for adverse effects as well as for increases in FXN mRNA, frataxin protein, and chromatin modification in blood cells. RESULTS: In the neuronal cell model, HDACi 109/RG2833 increases FXN mRNA levels and frataxin protein, with concomitant changes in the epigenetic state of the gene. Chromatin signatures indicate that histone H3 lysine 9 is a key residue for gene silencing through methylation and reactivation through acetylation, mediated by the HDACi. Drug treatment in FRDA patients demonstrated increased FXN mRNA and H3 lysine 9 acetylation in peripheral blood mononuclear cells. No safety issues were encountered. INTERPRETATION: Drug exposure inducing epigenetic changes in neurons in vitro is comparable to the exposure required in patients to see epigenetic changes in circulating lymphoid cells and increases in gene expression. These findings provide a proof of concept for the development of an epigenetic therapy for this fatal neurological disease.


Subject(s)
Friedreich Ataxia/drug therapy , Friedreich Ataxia/genetics , Gene Expression Regulation/drug effects , Histone Deacetylase Inhibitors/therapeutic use , Iron-Binding Proteins/genetics , Administration, Oral , Adolescent , Adult , Aminocaproates/pharmacology , Aminocaproates/therapeutic use , Area Under Curve , Benzamides/pharmacology , Benzamides/therapeutic use , Cell Differentiation/drug effects , Cell Differentiation/genetics , Cell Line, Transformed , Chromatin Immunoprecipitation , Cohort Studies , Cross-Sectional Studies , DNA Methylation/drug effects , DNA Methylation/genetics , Dose-Response Relationship, Drug , Double-Blind Method , Female , Friedreich Ataxia/pathology , Gene Expression Regulation/genetics , Humans , Leukocytes, Mononuclear/drug effects , Leukocytes, Mononuclear/metabolism , Male , Membrane Potentials/drug effects , Membrane Potentials/physiology , Middle Aged , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/metabolism , Neurons/drug effects , Pluripotent Stem Cells , Trinucleotide Repeat Expansion/genetics , Young Adult , Frataxin
3.
Hum Mol Genet ; 22(25): 5276-87, 2013 Dec 20.
Article in English | MEDLINE | ID: mdl-23933738

ABSTRACT

Myotonic dystrophy type 1 (DM1) is an inherited dominant muscular dystrophy caused by expanded CTG·CAG triplet repeats in the 3' untranslated region of the DMPK1 gene, which produces a toxic gain-of-function CUG RNA. It has been shown that the severity of disease symptoms, age of onset and progression are related to the length of the triplet repeats. However, the mechanism(s) of CTG·CAG triplet-repeat instability is not fully understood. Herein, induced pluripotent stem cells (iPSCs) were generated from DM1 and Huntington's disease patient fibroblasts. We isolated 41 iPSC clones from DM1 fibroblasts, all showing different CTG·CAG repeat lengths, thus demonstrating somatic instability within the initial fibroblast population. During propagation of the iPSCs, the repeats expanded in a manner analogous to the expansion seen in somatic cells from DM1 patients. The correlation between repeat length and expansion rate identified the interval between 57 and 126 repeats as being an important length threshold where expansion rates dramatically increased. Moreover, longer repeats showed faster triplet-repeat expansion. However, the overall tendency of triplet repeats to expand ceased on differentiation into differentiated embryoid body or neurospheres. The mismatch repair components MSH2, MSH3 and MSH6 were highly expressed in iPSCs compared with fibroblasts, and only occupied the DMPK1 gene harboring longer CTG·CAG triplet repeats. In addition, shRNA silencing of MSH2 impeded CTG·CAG triplet-repeat expansion. The information gained from these studies provides new insight into a general mechanism of triplet-repeat expansion in iPSCs.


Subject(s)
Myotonic Dystrophy/genetics , Pluripotent Stem Cells/metabolism , Protein Serine-Threonine Kinases/genetics , Trinucleotide Repeat Expansion/genetics , 3' Untranslated Regions/genetics , Cell Culture Techniques , DNA-Binding Proteins/biosynthesis , DNA-Binding Proteins/genetics , Fibroblasts/metabolism , Fibroblasts/pathology , Gene Expression Regulation , Humans , Huntington Disease/genetics , Huntington Disease/pathology , MutS Homolog 2 Protein/biosynthesis , MutS Homolog 2 Protein/genetics , MutS Homolog 3 Protein , Myotonic Dystrophy/pathology , Myotonin-Protein Kinase , Pluripotent Stem Cells/pathology
4.
J Biol Chem ; 287(35): 29861-72, 2012 Aug 24.
Article in English | MEDLINE | ID: mdl-22798143

ABSTRACT

The genetic mutation in Friedreich ataxia (FRDA) is a hyperexpansion of the triplet-repeat sequence GAA·TTC within the first intron of the FXN gene. Although yeast and reporter construct models for GAA·TTC triplet-repeat expansion have been reported, studies on FRDA pathogenesis and therapeutic development are limited by the availability of an appropriate cell model in which to study the mechanism of instability of the GAA·TTC triplet repeats in the human genome. Herein, induced pluripotent stem cells (iPSCs) were generated from FRDA patient fibroblasts after transduction with the four transcription factors Oct4, Sox2, Klf4, and c-Myc. These cells were differentiated into neurospheres and neuronal precursors in vitro, providing a valuable cell model for FRDA. During propagation of the iPSCs, GAA·TTC triplet repeats expanded at a rate of about two GAA·TTC triplet repeats/replication. However, GAA·TTC triplet repeats were stable in FRDA fibroblasts and neuronal stem cells. The mismatch repair enzymes MSH2, MSH3, and MSH6, implicated in repeat instability in other triplet-repeat diseases, were highly expressed in pluripotent stem cells compared with fibroblasts and neuronal stem cells and occupied FXN intron 1. In addition, shRNA silencing of MSH2 and MSH6 impeded GAA·TTC triplet-repeat expansion. A specific pyrrole-imidazole polyamide targeting GAA·TTC triplet-repeat DNA partially blocked repeat expansion by displacing MSH2 from FXN intron 1 in FRDA iPSCs. These studies suggest that in FRDA, GAA·TTC triplet-repeat instability occurs in embryonic cells and involves the highly active mismatch repair system.


Subject(s)
DNA Mismatch Repair , Friedreich Ataxia/metabolism , Genome, Human , Induced Pluripotent Stem Cells/metabolism , Iron-Binding Proteins/metabolism , Models, Biological , MutS Homolog 2 Protein/metabolism , Trinucleotide Repeat Expansion , Animals , Cell Differentiation/genetics , Cell Line , Fibroblasts/metabolism , Fibroblasts/pathology , Friedreich Ataxia/genetics , Friedreich Ataxia/pathology , Humans , Induced Pluripotent Stem Cells/pathology , Introns/genetics , Iron-Binding Proteins/genetics , Kruppel-Like Factor 4 , Mice , MutS Homolog 2 Protein/genetics , Neural Stem Cells/metabolism , Neural Stem Cells/pathology , Spheroids, Cellular/metabolism , Spheroids, Cellular/pathology , Transcription Factors/genetics , Transcription Factors/metabolism , Frataxin
5.
Cell Stem Cell ; 7(5): 631-7, 2010 Nov 05.
Article in English | MEDLINE | ID: mdl-21040903

ABSTRACT

The inherited neurodegenerative disease Friedreich's ataxia (FRDA) is caused by GAA⋅TTC triplet repeat hyperexpansions within the first intron of the FXN gene, encoding the mitochondrial protein frataxin. Long GAA⋅TTC repeats cause heterochromatin-mediated gene silencing and loss of frataxin in affected individuals. We report the derivation of induced pluripotent stem cells (iPSCs) from FRDA patient fibroblasts by transcription factor reprogramming. FXN gene repression is maintained in the iPSCs, as are the global gene expression signatures reflecting the human disease. GAA⋅TTC repeats uniquely in FXN in the iPSCs exhibit repeat instability similar to patient families, where they expand and/or contract with discrete changes in length between generations. The mismatch repair enzyme MSH2, implicated in repeat instability in other triplet repeat diseases, is highly expressed in pluripotent cells and occupies FXN intron 1, and shRNA silencing of MSH2 impedes repeat expansion, providing a possible molecular explanation for repeat expansion in FRDA.


Subject(s)
Friedreich Ataxia/genetics , Induced Pluripotent Stem Cells , Trinucleotide Repeat Expansion/genetics , Cells, Cultured , DNA Repeat Expansion , Humans , Iron-Binding Proteins/genetics , Microsatellite Instability , Frataxin
SELECTION OF CITATIONS
SEARCH DETAIL
...