Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 25
Filter
Add more filters










Publication year range
1.
Nat Commun ; 14(1): 1896, 2023 04 05.
Article in English | MEDLINE | ID: mdl-37019888

ABSTRACT

Transcriptional changes in Rett syndrome (RTT) are assumed to directly correlate with steady-state mRNA levels, but limited evidence in mice suggests that changes in transcription can be compensated by post-transcriptional regulation. We measure transcription rate and mRNA half-life changes in RTT patient neurons using RATEseq, and re-interpret nuclear and whole-cell RNAseq from Mecp2 mice. Genes are dysregulated by changing transcription rate or half-life and are buffered when both change. We utilized classifier models to predict the direction of transcription rate changes and find that combined frequencies of three dinucleotides are better predictors than CA and CG. MicroRNA and RNA-binding Protein (RBP) motifs are enriched in 3'UTRs of genes with half-life changes. Nuclear RBP motifs are enriched on buffered genes with increased transcription rate. We identify post-transcriptional mechanisms in humans and mice that alter half-life or buffer transcription rate changes when a transcriptional modulator gene is mutated in a neurodevelopmental disorder.


Subject(s)
Rett Syndrome , Humans , Mice , Animals , Rett Syndrome/genetics , RNA, Messenger , Half-Life , Methyl-CpG-Binding Protein 2/metabolism , Gene Expression Regulation
2.
bioRxiv ; 2023 Mar 01.
Article in English | MEDLINE | ID: mdl-36909614

ABSTRACT

The contribution of mRNA half-life is commonly overlooked when examining changes in mRNA abundance during development. mRNA levels of some genes are regulated by transcription rate only, but others may be regulated by mRNA half-life only shifts. Furthermore, transcriptional buffering is predicted when changes in transcription rates have compensating shifts in mRNA half-life resulting in no change to steady-state levels. Likewise, transcriptional boosting should result when changes in transcription rate are accompanied by amplifying half-life shifts. During neurodevelopment there is widespread 3'UTR lengthening that could be shaped by differential shifts in the stability of existing short or long 3'UTR transcript isoforms. We measured transcription rate and mRNA half-life changes during induced human Pluripotent Stem Cell (iPSC)-derived neuronal development using RATE-seq. During transitions to progenitor and neuron stages, transcriptional buffering occurred in up to 50%, and transcriptional boosting in up to 15%, of genes with changed transcription rates. The remaining changes occurred by transcription rate only or mRNA half-life only shifts. Average mRNA half-life decreased two-fold in neurons relative to iPSCs. Short gene isoforms were more destabilized in neurons and thereby increased the average 3'UTR length. Small RNA sequencing captured an increase in microRNA copy number per cell during neurodevelopment. We propose that mRNA destabilization and 3'UTR lengthening are driven in part by an increase in microRNA load in neurons. Our findings identify mRNA stability mechanisms in human neurodevelopment that regulate gene and isoform level abundance and provide a precedent for similar post-transcriptional regulatory events as other tissues develop.

3.
Cell Rep ; 30(12): 4179-4196.e11, 2020 03 24.
Article in English | MEDLINE | ID: mdl-32209477

ABSTRACT

Regulation of translation during human development is poorly understood, and its dysregulation is associated with Rett syndrome (RTT). To discover shifts in mRNA ribosomal engagement (RE) during human neurodevelopment, we use parallel translating ribosome affinity purification sequencing (TRAP-seq) and RNA sequencing (RNA-seq) on control and RTT human induced pluripotent stem cells, neural progenitor cells, and cortical neurons. We find that 30% of transcribed genes are translationally regulated, including key gene sets (neurodevelopment, transcription and translation factors, and glycolysis). Approximately 35% of abundant intergenic long noncoding RNAs (lncRNAs) are ribosome engaged. Neurons translate mRNAs more efficiently and have longer 3' UTRs, and RE correlates with elements for RNA-binding proteins. RTT neurons have reduced global translation and compromised mTOR signaling, and >2,100 genes are translationally dysregulated. NEDD4L E3-ubiquitin ligase is translationally impaired, ubiquitinated protein levels are reduced, and protein targets accumulate in RTT neurons. Overall, the dynamic translatome in neurodevelopment is disturbed in RTT and provides insight into altered ubiquitination that may have therapeutic implications.


Subject(s)
Nervous System/growth & development , Nervous System/pathology , Rett Syndrome/genetics , Ribosomes/metabolism , Ubiquitination , 3' Untranslated Regions/genetics , Animals , Base Sequence , Female , Gene Expression Regulation, Developmental , Glycolysis/genetics , Induced Pluripotent Stem Cells/metabolism , Methyl-CpG-Binding Protein 2/metabolism , Mice , Nedd4 Ubiquitin Protein Ligases/metabolism , Neurons/metabolism , Protein Binding , Protein Biosynthesis , RNA, Untranslated/genetics , RNA, Untranslated/metabolism , RNA-Binding Proteins/metabolism , Ribosomal Proteins/genetics , Ribosomal Proteins/metabolism , Transcription Factors/metabolism , Ubiquitination/genetics
4.
Biol Psychiatry ; 87(2): 139-149, 2020 01 15.
Article in English | MEDLINE | ID: mdl-31540669

ABSTRACT

BACKGROUND: The Xp22.11 locus that encompasses PTCHD1, DDX53, and the long noncoding RNA PTCHD1-AS is frequently disrupted in male subjects with autism spectrum disorder (ASD), but the functional consequences of these genetic risk factors for ASD are unknown. METHODS: To evaluate the functional consequences of PTCHD1 locus deletions, we generated induced pluripotent stem cells (iPSCs) from unaffected control subjects and 3 subjects with ASD with microdeletions affecting PTCHD1-AS/PTCHD1, PTCHD1-AS/DDX53, or PTCHD1-AS alone. Function of iPSC-derived cortical neurons was assessed using molecular approaches and electrophysiology. We also compiled novel and known genetic variants of the PTCHD1 locus to explore the roles of PTCHD1 and PTCHD1-AS in genetic risk for ASD and other neurodevelopmental disorders. Finally, genome editing was used to explore the functional consequences of deleting a single conserved exon of PTCHD1-AS. RESULTS: iPSC-derived neurons from subjects with ASD exhibited reduced miniature excitatory postsynaptic current frequency and N-methyl-D-aspartate receptor hypofunction. We found that 35 ASD-associated deletions mapping to the PTCHD1 locus disrupted exons of PTCHD1-AS. We also found a novel ASD-associated deletion of PTCHD1-AS exon 3 and showed that exon 3 loss altered PTCHD1-AS splicing without affecting expression of the neighboring PTCHD1 coding gene. Finally, targeted disruption of PTCHD1-AS exon 3 recapitulated diminished miniature excitatory postsynaptic current frequency, supporting a role for the long noncoding RNA in the etiology of ASD. CONCLUSIONS: Our genetic findings provide strong evidence that PTCHD1-AS deletions are risk factors for ASD, and human iPSC-derived neurons implicate these deletions in the neurophysiology of excitatory synapses and in ASD-associated synaptic impairment.


Subject(s)
Autism Spectrum Disorder , Autistic Disorder , Induced Pluripotent Stem Cells , Autism Spectrum Disorder/genetics , Autistic Disorder/genetics , Humans , Male , Membrane Proteins , Neurons , Synapses
5.
Stem Cell Reports ; 13(6): 1126-1141, 2019 12 10.
Article in English | MEDLINE | ID: mdl-31813827

ABSTRACT

Induced pluripotent stem cells (iPSC) derived from healthy individuals are important controls for disease-modeling studies. Here we apply precision health to create a high-quality resource of control iPSCs. Footprint-free lines were reprogrammed from four volunteers of the Personal Genome Project Canada (PGPC). Multilineage-directed differentiation efficiently produced functional cortical neurons, cardiomyocytes and hepatocytes. Pilot users demonstrated versatility by generating kidney organoids, T lymphocytes, and sensory neurons. A frameshift knockout was introduced into MYBPC3 and these cardiomyocytes exhibited the expected hypertrophic phenotype. Whole-genome sequencing-based annotation of PGPC lines revealed on average 20 coding variants. Importantly, nearly all annotated PGPC and HipSci lines harbored at least one pre-existing or acquired variant with cardiac, neurological, or other disease associations. Overall, PGPC lines were efficiently differentiated by multiple users into cells from six tissues for disease modeling, and variant-preferred healthy control lines were identified for specific disease settings.


Subject(s)
Cell Differentiation , Cell Lineage , Induced Pluripotent Stem Cells/cytology , Induced Pluripotent Stem Cells/metabolism , CRISPR-Cas Systems , Cell Self Renewal , Cell Separation , Ectoderm/cytology , Ectoderm/metabolism , Gene Editing , Humans , Myocytes, Cardiac/cytology , Myocytes, Cardiac/metabolism , Neurons/cytology , Neurons/metabolism , Organoids , Phenotype , T-Lymphocytes/metabolism , Whole Genome Sequencing
6.
Nat Neurosci ; 22(4): 556-564, 2019 04.
Article in English | MEDLINE | ID: mdl-30911184

ABSTRACT

Heterozygous loss-of-function mutations in SHANK2 are associated with autism spectrum disorder (ASD). We generated cortical neurons from induced pluripotent stem cells derived from neurotypic and ASD-affected donors. We developed sparse coculture for connectivity assays where SHANK2 and control neurons were differentially labeled and sparsely seeded together on a lawn of unlabeled control neurons. We observed increases in dendrite length, dendrite complexity, synapse number, and frequency of spontaneous excitatory postsynaptic currents. These findings were phenocopied in gene-edited homozygous SHANK2 knockout cells and rescued by gene correction of an ASD SHANK2 mutation. Dendrite length increases were exacerbated by IGF1, TG003, or BDNF, and suppressed by DHPG treatment. The transcriptome in isogenic SHANK2 neurons was perturbed in synapse, plasticity, and neuronal morphogenesis gene sets and ASD gene modules, and activity-dependent dendrite extension was impaired. Our findings provide evidence for hyperconnectivity and altered transcriptome in SHANK2 neurons derived from ASD subjects.


Subject(s)
Autism Spectrum Disorder/genetics , Autism Spectrum Disorder/pathology , Dendrites/pathology , Nerve Tissue Proteins/genetics , Neurons/pathology , Autism Spectrum Disorder/metabolism , Coculture Techniques , Dendrites/metabolism , Excitatory Postsynaptic Potentials , Gene Knockout Techniques , Haploinsufficiency , Humans , Induced Pluripotent Stem Cells , Male , Neuronal Plasticity , Neurons/metabolism , Transcriptome
7.
Elife ; 82019 02 12.
Article in English | MEDLINE | ID: mdl-30747104

ABSTRACT

Induced pluripotent stem cell (iPSC)-derived neurons are increasingly used to model Autism Spectrum Disorder (ASD), which is clinically and genetically heterogeneous. To study the complex relationship of penetrant and weaker polygenic risk variants to ASD, 'isogenic' iPSC-derived neurons are critical. We developed a set of procedures to control for heterogeneity in reprogramming and differentiation, and generated 53 different iPSC-derived glutamatergic neuronal lines from 25 participants from 12 unrelated families with ASD. Heterozygous de novo and rare-inherited presumed-damaging variants were characterized in ASD risk genes/loci. Combinations of putative etiologic variants (GLI3/KIF21A or EHMT2/UBE2I) in separate families were modeled. We used a multi-electrode array, with patch-clamp recordings, to determine a reproducible synaptic phenotype in 25% of the individuals with ASD (other relevant data on the remaining lines was collected). Our most compelling new results revealed a consistent spontaneous network hyperactivity in neurons deficient for CNTN5 or EHMT2. The biobank of iPSC-derived neurons and accompanying genomic data are available to accelerate ASD research. Editorial note: This article has been through an editorial process in which authors decide how to respond to the issues raised during peer review. The Reviewing Editor's assessment is that all the issues have been addressed (see decision letter).


Subject(s)
Autistic Disorder/physiopathology , Contactins/metabolism , Histocompatibility Antigens/metabolism , Histone-Lysine N-Methyltransferase/metabolism , Induced Pluripotent Stem Cells/physiology , Nerve Net/physiology , Neurons/physiology , Adolescent , Adult , Cells, Cultured , Child , Contactins/deficiency , Contactins/genetics , Electrophysiological Phenomena , Female , Heterozygote , Histocompatibility Antigens/genetics , Histone-Lysine N-Methyltransferase/deficiency , Histone-Lysine N-Methyltransferase/genetics , Humans , Kinesins/genetics , Kinesins/metabolism , Male , Middle Aged , Models, Theoretical , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/metabolism , Patch-Clamp Techniques , Ubiquitin-Conjugating Enzymes/genetics , Ubiquitin-Conjugating Enzymes/metabolism , Young Adult , Zinc Finger Protein Gli3/genetics , Zinc Finger Protein Gli3/metabolism
9.
Stem Cell Reports ; 11(5): 1211-1225, 2018 11 13.
Article in English | MEDLINE | ID: mdl-30392976

ABSTRACT

Autism spectrum disorder (ASD) is phenotypically and genetically heterogeneous. We present a CRISPR gene editing strategy to insert a protein tag and premature termination sites creating an induced pluripotent stem cell (iPSC) knockout resource for functional studies of ten ASD-relevant genes (AFF2/FMR2, ANOS1, ASTN2, ATRX, CACNA1C, CHD8, DLGAP2, KCNQ2, SCN2A, TENM1). Neurogenin 2 (NGN2)-directed induction of iPSCs allowed production of excitatory neurons, and mutant proteins were not detectable. RNA sequencing revealed convergence of several neuronal networks. Using both patch-clamp and multi-electrode array approaches, the electrophysiological deficits measured were distinct for different mutations. However, they culminated in a consistent reduction in synaptic activity, including reduced spontaneous excitatory postsynaptic current frequencies in AFF2/FMR2-, ASTN2-, ATRX-, KCNQ2-, and SCN2A-null neurons. Despite ASD susceptibility genes belonging to different gene ontologies, isogenic stem cell resources can reveal common functional phenotypes, such as reduced functional connectivity.


Subject(s)
Autistic Disorder/genetics , Autistic Disorder/physiopathology , Gene Editing , Genetic Predisposition to Disease , Neurons/metabolism , Neurons/pathology , Cell Line , Electrodes , Gene Knockout Techniques , HEK293 Cells , Humans , Induced Pluripotent Stem Cells/metabolism , Mutagenesis, Insertional/genetics , Phenotype
10.
CMAJ ; 190(5): E126-E136, 2018 02 05.
Article in English | MEDLINE | ID: mdl-29431110

ABSTRACT

BACKGROUND: The Personal Genome Project Canada is a comprehensive public data resource that integrates whole genome sequencing data and health information. We describe genomic variation identified in the initial recruitment cohort of 56 volunteers. METHODS: Volunteers were screened for eligibility and provided informed consent for open data sharing. Using blood DNA, we performed whole genome sequencing and identified all possible classes of DNA variants. A genetic counsellor explained the implication of the results to each participant. RESULTS: Whole genome sequencing of the first 56 participants identified 207 662 805 sequence variants and 27 494 copy number variations. We analyzed a prioritized disease-associated data set (n = 1606 variants) according to standardized guidelines, and interpreted 19 variants in 14 participants (25%) as having obvious health implications. Six of these variants (e.g., in BRCA1 or mosaic loss of an X chromosome) were pathogenic or likely pathogenic. Seven were risk factors for cancer, cardiovascular or neurobehavioural conditions. Four other variants - associated with cancer, cardiac or neurodegenerative phenotypes - remained of uncertain significance because of discrepancies among databases. We also identified a large structural chromosome aberration and a likely pathogenic mitochondrial variant. There were 172 recessive disease alleles (e.g., 5 individuals carried mutations for cystic fibrosis). Pharmacogenomics analyses revealed another 3.9 potentially relevant genotypes per individual. INTERPRETATION: Our analyses identified a spectrum of genetic variants with potential health impact in 25% of participants. When also considering recessive alleles and variants with potential pharmacologic relevance, all 56 participants had medically relevant findings. Although access is mostly limited to research, whole genome sequencing can provide specific and novel information with the potential of major impact for health care.


Subject(s)
Genetic Variation/genetics , Genome, Human/genetics , Sequence Analysis, DNA/methods , Whole Genome Sequencing/methods , Canada , Female , Genes, Recessive/genetics , Genetic Predisposition to Disease/genetics , Humans , Male
11.
Neurobiol Dis ; 76: 37-45, 2015 Apr.
Article in English | MEDLINE | ID: mdl-25644311

ABSTRACT

MECP2 mutations cause the X-linked neurodevelopmental disorder Rett Syndrome (RTT) by consistently altering the protein encoded by the MECP2e1 alternative transcript. While mutations that simultaneously affect both MECP2e1 and MECP2e2 isoforms have been widely studied, the consequence of MECP2e1 deficiency on human neurons remains unknown. Here we report the first isoform-specific patient induced pluripotent stem cell (iPSC) model of RTT. RTTe1 patient iPS cell-derived neurons retain an inactive X-chromosome and express only the mutant allele. Single-cell mRNA analysis demonstrated they have a molecular signature of cortical neurons. Mutant neurons exhibited a decrease in soma size, reduced dendritic complexity and decreased cell capacitance, consistent with impaired neuronal maturation. The soma size phenotype was rescued cell-autonomously by MECP2e1 transduction in a level-dependent manner but not by MECP2e2 gene transfer. Importantly, MECP2e1 mutant neurons showed a dysfunction in action potential generation, voltage-gated Na(+) currents, and miniature excitatory synaptic current frequency and amplitude. We conclude that MECP2e1 mutation affects soma size, information encoding properties and synaptic connectivity in human neurons that are defective in RTT.


Subject(s)
Induced Pluripotent Stem Cells/pathology , Induced Pluripotent Stem Cells/physiology , Methyl-CpG-Binding Protein 2/genetics , Neurons/pathology , Neurons/physiology , Rett Syndrome/genetics , Action Potentials , Humans , Mutation , Neurons/metabolism , Protein Isoforms , Rett Syndrome/pathology , Rett Syndrome/physiopathology
12.
Mol Ther ; 21(8): 1536-50, 2013 Aug.
Article in English | MEDLINE | ID: mdl-23752310

ABSTRACT

Retroviral vectors are silenced in embryonic stem (ES) cells by epigenetic mechanisms whose kinetics are poorly understood. We show here that a 3'D4Z4 insulator directs retroviral expression with persistent but variable expression for up to 5 months. Combining an internal 3'D4Z4 with HS4 insulators in the long terminal repeats (LTRs) shows that these elements cooperate, and defines the first retroviral vector that fully escapes long-term silencing. Using FLP recombinase to induce deletion of 3'D4Z4 from the provirus in ES cell clones, we established retroviral silencing at many but not all integration sites. This finding shows that 3'D4Z4 does not target retrovirus integration into favorable epigenomic domains but rather protects the transgene from silencing. Chromatin analyses demonstrate that 3'D4Z4 blocks the spread of heterochromatin marks including DNA methylation and repressive histone modifications such as H3K9 methylation. In addition, our deletion system reveals three distinct kinetic classes of silencing (rapid, gradual or not silenced), in which multiple epigenetic pathways participate in silencing at different integration sites. We conclude that vectors with both 3'D4Z4 and HS4 insulator elements fully block silencing, and may have unprecedented utility for gene transfer applications that require long-term gene expression in pluripotent stem (PS) cells.


Subject(s)
Embryonic Stem Cells/metabolism , Epigenesis, Genetic , Gene Silencing , Genetic Vectors/genetics , Insulator Elements , Retroviridae/genetics , Sequence Deletion , Animals , Chromatin/metabolism , DNA Methylation , Gene Expression Regulation , Gene Order , Histones/metabolism , Homologous Recombination , Kinetics , Methylation , Mice , Proviruses/genetics , Terminal Repeat Sequences , Transgenes
13.
Stem Cells Transl Med ; 2(1): 2-15, 2013 Jan.
Article in English | MEDLINE | ID: mdl-23283491

ABSTRACT

Elastin haploinsufficiency in Williams-Beuren syndrome (WBS) leads to increased vascular smooth muscle cell (SMC) proliferation and stenoses. Our objective was to generate a human induced pluripotent stem (hiPS) cell model for in vitro assessment of the WBS phenotype and to test the ability of candidate agents to rescue the phenotype. hiPS cells were reprogrammed from skin fibroblasts of a WBS patient with aortic and pulmonary stenosis and healthy control BJ fibroblasts using four-factor retrovirus reprogramming and were differentiated into SMCs. Differentiated SMCs were treated with synthetic elastin-binding protein ligand 2 (EBPL2) (20 µg/ml) or the antiproliferative drug rapamycin (100 nM) for 5 days. We generated four WBS induced pluripotent stem (iPS) cell lines that expressed pluripotency genes and differentiated into all three germ layers. Directed differentiation of BJ iPS cells yielded an 85%-92% pure SMC population that expressed differentiated SMC markers, were functionally contractile, and formed tube-like structures on three-dimensional gel assay. Unlike BJ iPS cells, WBS iPS cells generated immature SMCs that were highly proliferative, showed lower expression of differentiated SMC markers, reduced response to the vasoactive agonists, carbachol and endothelin-1, impaired vascular tube formation, and reduced calcium flux. EBPL2 partially rescued and rapamycin fully rescued the abnormal SMC phenotype by decreasing the smooth muscle proliferation rate and enhancing differentiation and tube formation. WBS iPS cell-derived SMCs demonstrate an immature proliferative phenotype with reduced functional and contractile properties, thereby recapitulating the human disease phenotype. The ability of rapamycin to rescue the phenotype provides an attractive therapeutic candidate for patients with WBS and vascular stenoses.


Subject(s)
Induced Pluripotent Stem Cells/physiology , Muscle, Smooth, Vascular/pathology , Myocytes, Smooth Muscle/metabolism , Williams Syndrome/pathology , Antigens, Differentiation/genetics , Antigens, Differentiation/metabolism , Calcium Signaling , Cell Differentiation/drug effects , Cell Proliferation/drug effects , Cells, Cultured , Fibroblasts/metabolism , Fibroblasts/physiology , Hemizygote , Human Umbilical Vein Endothelial Cells , Humans , Induced Pluripotent Stem Cells/metabolism , Kruppel-Like Factor 4 , Kruppel-Like Transcription Factors/biosynthesis , Male , Muscle Contraction , Myocytes, Smooth Muscle/drug effects , Myocytes, Smooth Muscle/physiology , Octamer Transcription Factor-3/biosynthesis , Peptide Fragments/pharmacology , Phenotype , Proto-Oncogene Proteins c-myc/biosynthesis , Recombinant Proteins/biosynthesis , SOXB1 Transcription Factors/biosynthesis , Sequence Analysis, DNA , Sirolimus/pharmacology , TOR Serine-Threonine Kinases/antagonists & inhibitors , Transcriptome/drug effects , Williams Syndrome/genetics
14.
Nat Biotechnol ; 30(9): 876-82, 2012 Sep.
Article in English | MEDLINE | ID: mdl-22922672

ABSTRACT

Cystic fibrosis (CF) is a fatal genetic disease caused by mutations in the CFTR (cystic fibrosis transmembrane conductance regulator) gene, which regulates chloride and water transport across all epithelia and affects multiple organs, including the lungs. Here we report an in vitro directed differentiation protocol for generating functional CFTR-expressing airway epithelia from human embryonic stem cells. Carefully timed treatment by exogenous growth factors that mimic endoderm developmental pathways in vivo followed by air-liquid interface culture results in maturation of patches of tight junction­coupled differentiated airway epithelial cells that demonstrate active CFTR transport function. As a proof of concept, treatment of CF patient induced pluripotent stem cell­derived epithelial cells with a small-molecule compound to correct for the common CF processing mutation resulted in enhanced plasma membrane localization of mature CFTR protein. Our study provides a method for generating patient-specific airway epithelial cells for disease modeling and in vitro drug testing.


Subject(s)
Cell Differentiation/drug effects , Cystic Fibrosis Transmembrane Conductance Regulator/biosynthesis , Epithelial Cells/cytology , Pluripotent Stem Cells/cytology , Animals , Cell Culture Techniques , Cell Differentiation/physiology , Cystic Fibrosis Transmembrane Conductance Regulator/genetics , Down-Regulation/drug effects , Epithelial Cells/drug effects , Epithelial Cells/metabolism , Gene Expression Profiling , Humans , Intercellular Signaling Peptides and Proteins/pharmacology , Mice , Pluripotent Stem Cells/drug effects , Pluripotent Stem Cells/metabolism , Respiratory Mucosa/cytology , Up-Regulation/drug effects
15.
PLoS One ; 7(5): e37130, 2012.
Article in English | MEDLINE | ID: mdl-22606340

ABSTRACT

Single cell imaging studies suggest that transcription is not continuous and occurs as discrete pulses of gene activity. To study mechanisms by which retroviral transgenes can transcribe to high levels, we used the MS2 system to visualize transcriptional dynamics of high expressing proviral integration sites in embryonic stem (ES) cells. We established two ES cell lines each bearing a single copy, self-inactivating retroviral vector with a strong ubiquitous human EF1α gene promoter directing expression of mRFP fused to an MS2-stem-loop array. Transfection of MS2-EGFP generated EGFP focal dots bound to the mRFP-MS2 stem loop mRNA. These transcription foci colocalized with the transgene integration site detected by immunoFISH. Live tracking of single cells for 20 minutes detected EGFP focal dots that displayed frequent and rapid fluctuations in transcription over periods as short as 25 seconds. Similarly rapid fluctuations were detected from focal doublet signals that colocalized with replicated proviral integration sites by immunoFISH, consistent with transcriptional pulses from sister chromatids. We concluded that retroviral transgenes experience rapid transcriptional pulses in clonal ES cell lines that exhibit high level expression. These events are directed by a constitutive housekeeping gene promoter and may provide precedence for rapid transcriptional pulsing at endogenous genes in mammalian stem cells.


Subject(s)
Embryonic Stem Cells/metabolism , Embryonic Stem Cells/virology , Transgenes , Animals , Base Sequence , Cell Line , Chromatids/genetics , Gene Expression , Genes, Viral , Green Fluorescent Proteins/genetics , Humans , In Situ Hybridization, Fluorescence , Levivirus/genetics , Luminescent Proteins/genetics , Mice , Plasmids/genetics , Promoter Regions, Genetic , Retroviridae/genetics , Transcription, Genetic , Transfection , Virus Integration/genetics , Red Fluorescent Protein
16.
Hum Mol Genet ; 20(11): 2103-15, 2011 Jun 01.
Article in English | MEDLINE | ID: mdl-21372149

ABSTRACT

Rett syndrome (RTT) is a neurodevelopmental autism spectrum disorder that affects girls due primarily to mutations in the gene encoding methyl-CpG binding protein 2 (MECP2). The majority of RTT patients carry missense and nonsense mutations leading to a hypomorphic MECP2, while null mutations leading to the complete absence of a functional protein are rare. MECP2 is an X-linked gene subject to random X-chromosome inactivation resulting in mosaic expression of mutant MECP2. The lack of human brain tissue motivates the need for alternative human cellular models to study RTT. Here we report the characterization of a MECP2 mutation in a classic female RTT patient involving rearrangements that remove exons 3 and 4 creating a functionally null mutation. To generate human neuron models of RTT, we isolated human induced pluripotent stem (hiPS) cells from RTT patient fibroblasts. RTT-hiPS cells retained the MECP2 mutation, are pluripotent and fully reprogrammed, and retained an inactive X-chromosome in a nonrandom pattern. Taking advantage of the latter characteristic, we obtained a pair of isogenic wild-type and mutant MECP2 expressing RTT-hiPS cell lines that retained this MECP2 expression pattern upon differentiation into neurons. Phenotypic analysis of mutant RTT-hiPS cell-derived neurons demonstrated a reduction in soma size compared with the isogenic control RTT-hiPS cell-derived neurons from the same RTT patient. Analysis of isogenic control and mutant hiPS cell-derived neurons represents a promising source for understanding the pathogenesis of RTT and the role of MECP2 in human neurons.


Subject(s)
Induced Pluripotent Stem Cells/metabolism , Methyl-CpG-Binding Protein 2/genetics , Neurons/pathology , Rett Syndrome/genetics , X Chromosome Inactivation , Brain/cytology , Brain/metabolism , Cell Differentiation , Cell Line , Chromosome Mapping , Chromosomes, Human, X/genetics , DNA Fingerprinting , Exons , Female , Gene Expression Regulation , Genes, X-Linked , Genotype , Humans , Immunohistochemistry , Karyotyping , Male , Methyl-CpG-Binding Protein 2/metabolism , Mutation , Neurons/cytology , Phenotype
17.
Nat Protoc ; 4(12): 1828-44, 2009.
Article in English | MEDLINE | ID: mdl-20010937

ABSTRACT

Generation of induced pluripotent stem (iPS) cells from patients has exciting applications for studying molecular mechanisms of diseases, screening drugs and ultimately for use in cell therapies. However, the low efficiency and heterogeneous nature of reprogramming is a major impediment to the generation of personalized iPS cell lines. We reported in Nature Methods (6, 370-376, 2009) the first selection system to enrich for reprogrammed human iPS cells. Using a lentiviral vector that specifically expresses the enhanced green fluorescence protein and puromycin resistance genes in pluripotent stem cells, it is now possible to mark and enrich for human iPS cell colonies expressing endogenous pluripotency markers. In this study, we describe a detailed protocol for the production of the pluripotent state-specific lentiviral vector and the selection system for the induction of healthy and disease-specific human iPS cells. Overall, preparation of the selection system takes 2 weeks, and the generation of human iPS cells takes approximately 2 months.


Subject(s)
Cell Culture Techniques , Genetic Vectors , Induced Pluripotent Stem Cells/cytology , Lentivirus/genetics , Animals , Biomarkers/metabolism , Drug Resistance/genetics , Fibroblasts/cytology , Fibroblasts/virology , Green Fluorescent Proteins/analysis , Green Fluorescent Proteins/metabolism , Humans , Induced Pluripotent Stem Cells/virology , Mice , Transfection/methods
18.
PLoS One ; 4(8): e6810, 2009 Aug 27.
Article in English | MEDLINE | ID: mdl-19710912

ABSTRACT

BACKGROUND: Rett Syndrome (RTT) is an Autism Spectrum Disorder and the leading cause of mental retardation in females. RTT is caused by mutations in the Methyl CpG-Binding Protein-2 (MECP2) gene and has no treatment. Our objective is to develop viral vectors for MECP2 gene transfer into Neural Stem Cells (NSC) and neurons suitable for gene therapy of Rett Syndrome. METHODOLOGY/PRINCIPAL FINDINGS: We generated self-inactivating (SIN) retroviral vectors with the ubiquitous EF1alpha promoter avoiding known silencer elements to escape stem-cell-specific viral silencing. High efficiency NSC infection resulted in long-term EGFP expression in transduced NSC and after differentiation into neurons. Infection with Myc-tagged MECP2-isoform-specific (E1 and E2) vectors directed MeCP2 to heterochromatin of transduced NSC and neurons. In contrast, vectors with an internal mouse Mecp2 promoter (MeP) directed restricted expression only in neurons and glia and not NSC, recapitulating the endogenous expression pattern required to avoid detrimental consequences of MECP2 ectopic expression. In differentiated NSC from adult heterozygous Mecp2(tm1.1Bird)+/- female mice, 48% of neurons expressed endogenous MeCP2 due to random inactivation of the X-linked Mecp2 gene. Retroviral MECP2 transduction with EF1alpha and MeP vectors rescued expression in 95-100% of neurons resulting in increased dendrite branching function in vitro. Insulated MECP2 isoform-specific lentiviral vectors show long-term expression in NSC and their differentiated neuronal progeny, and directly infect dissociated murine cortical neurons with high efficiency. CONCLUSIONS/SIGNIFICANCE: MeP vectors recapitulate the endogenous expression pattern of MeCP2 in neurons and glia. They have utility to study MeCP2 isoform-specific functions in vitro, and are effective gene therapy vectors for rescuing dendritic maturation of neurons in an ex vivo model of RTT.


Subject(s)
Gene Expression Regulation , Genetic Therapy , Genetic Vectors , Methyl-CpG-Binding Protein 2/genetics , Rett Syndrome/therapy , Animals , Female , Humans , Lentivirus/genetics , Mice , Promoter Regions, Genetic , Transduction, Genetic
19.
Nat Methods ; 6(5): 370-6, 2009 May.
Article in English | MEDLINE | ID: mdl-19404254

ABSTRACT

Induced pluripotent stem (iPS) cells may be of use in regenerative medicine. However, the low efficiency of reprogramming is a major impediment to the generation of patient-specific iPS cell lines. Here we report the first selection system for the isolation of human iPS cells. We developed the EOS (Early Transposon promoter and Oct-4 (Pou5f1) and Sox2 enhancers) lentiviral vector to specifically express in mouse and human embryonic stem cells but not in primary fibroblasts. The bicistronic EOS vector marked emerging mouse and human iPS cell colonies with EGFP, and we used puromycin selection to aid the isolation of iPS cell lines that expressed endogenous pluripotency markers. These lines differentiated into cell types from all three germ layers. Reporter expression was extinguished upon differentiation and therefore monitored the residual pluripotent cells that form teratomas. Finally, we used EOS selection to establish Rett syndrome-specific mouse and human iPS cell lines with known mutations in MECP2.


Subject(s)
Cell Dedifferentiation/genetics , Cell Separation/methods , Genes, Reporter/genetics , Genetic Vectors/genetics , Lentivirus/genetics , Pluripotent Stem Cells/cytology , Pluripotent Stem Cells/metabolism , Animals , Cell Differentiation/drug effects , DNA Transposable Elements/genetics , Embryo, Mammalian/cytology , Embryonic Stem Cells/cytology , Embryonic Stem Cells/metabolism , Enhancer Elements, Genetic/genetics , Fibroblasts/cytology , Fibroblasts/metabolism , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , Humans , Methyl-CpG-Binding Protein 2/genetics , Mice , Mice, Inbred NOD , Mice, Mutant Strains , Mice, SCID , Promoter Regions, Genetic/genetics , Rett Syndrome/genetics , Rett Syndrome/pathology , Teratoma/pathology
20.
PLoS Genet ; 4(4): e1000051, 2008 Apr 11.
Article in English | MEDLINE | ID: mdl-18404216

ABSTRACT

The Locus Control Region (LCR) requires intronic elements within beta-globin transgenes to direct high level expression at all ectopic integration sites. However, these essential intronic elements cannot be transmitted through retrovirus vectors and their deletion may compromise the therapeutic potential for gene therapy. Here, we systematically regenerate functional beta-globin intron 2 elements that rescue LCR activity directed by 5'HS3. Evaluation in transgenic mice demonstrates that an Oct-1 binding site and an enhancer in the intron cooperate to increase expression levels from LCR globin transgenes. Replacement of the intronic AT-rich region with the Igmu 3'MAR rescues LCR activity in single copy transgenic mice. Importantly, a combination of the Oct-1 site, Igmu 3'MAR and intronic enhancer in the BGT158 cassette directs more consistent levels of expression in transgenic mice. By introducing intron-modified transgenes into the same genomic integration site in erythroid cells, we show that BGT158 has the greatest transcriptional induction. 3D DNA FISH establishes that induction stimulates this small 5'HS3 containing transgene and the endogenous locus to spatially reorganize towards more central locations in erythroid nuclei. Electron Spectroscopic Imaging (ESI) of chromatin fibers demonstrates that ultrastructural heterochromatin is primarily perinuclear and does not reorganize. Finally, we transmit intron-modified globin transgenes through insulated self-inactivating (SIN) lentivirus vectors into erythroid cells. We show efficient transfer and robust mRNA and protein expression by the BGT158 vector, and virus titer improvements mediated by the modified intron 2 in the presence of an LCR cassette composed of 5'HS2-4. Our results have important implications for the mechanism of LCR activity at ectopic integration sites. The modified transgenes are the first to transfer intronic elements that potentiate LCR activity and are designed to facilitate correction of hemoglobinopathies using single copy vectors.


Subject(s)
Genetic Therapy/methods , Globins/genetics , Anemia, Sickle Cell/genetics , Anemia, Sickle Cell/therapy , Animals , Base Sequence , Binding Sites/genetics , Cell Line , DNA Primers/genetics , Enhancer Elements, Genetic , Female , Gene Expression , Genetic Complementation Test , Genetic Vectors , Introns , Locus Control Region , Male , Mice , Mice, Transgenic , Octamer Transcription Factor-1/metabolism , Pregnancy , Recombinant Proteins/genetics , Reverse Transcriptase Polymerase Chain Reaction
SELECTION OF CITATIONS
SEARCH DETAIL