Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 15 de 15
Filter
1.
Cell ; 151(7): 1431-42, 2012 Dec 21.
Article in English | MEDLINE | ID: mdl-23260136

ABSTRACT

De novo mutation plays an important role in autism spectrum disorders (ASDs). Notably, pathogenic copy number variants (CNVs) are characterized by high mutation rates. We hypothesize that hypermutability is a property of ASD genes and may also include nucleotide-substitution hot spots. We investigated global patterns of germline mutation by whole-genome sequencing of monozygotic twins concordant for ASD and their parents. Mutation rates varied widely throughout the genome (by 100-fold) and could be explained by intrinsic characteristics of DNA sequence and chromatin structure. Dense clusters of mutations within individual genomes were attributable to compound mutation or gene conversion. Hypermutability was a characteristic of genes involved in ASD and other diseases. In addition, genes impacted by mutations in this study were associated with ASD in independent exome-sequencing data sets. Our findings suggest that regional hypermutation is a significant factor shaping patterns of genetic variation and disease risk in humans.


Subject(s)
Autistic Disorder/genetics , Genome-Wide Association Study , Germ-Line Mutation , Mutation Rate , Animals , Cell Line , Exons , Female , Humans , Male , Maternal Age , Pan troglodytes/genetics , Paternal Age , Sequence Analysis, DNA , Twins, Monozygotic
2.
Nature ; 478(7367): 70-5, 2011 Oct 05.
Article in English | MEDLINE | ID: mdl-21979046

ABSTRACT

The exchange of the oocyte's genome with the genome of a somatic cell, followed by the derivation of pluripotent stem cells, could enable the generation of specific cells affected in degenerative human diseases. Such cells, carrying the patient's genome, might be useful for cell replacement. Here we report that the development of human oocytes after genome exchange arrests at late cleavage stages in association with transcriptional abnormalities. In contrast, if the oocyte genome is not removed and the somatic cell genome is merely added, the resultant triploid cells develop to the blastocyst stage. Stem cell lines derived from these blastocysts differentiate into cell types of all three germ layers, and a pluripotent gene expression program is established on the genome derived from the somatic cell. This result demonstrates the feasibility of reprogramming human cells using oocytes and identifies removal of the oocyte genome as the primary cause of developmental failure after genome exchange.


Subject(s)
Cellular Reprogramming , Induced Pluripotent Stem Cells/cytology , Induced Pluripotent Stem Cells/metabolism , Oocytes/cytology , Oocytes/physiology , Adult , Blastocyst/cytology , Blastocyst/metabolism , Cell Differentiation , DNA Methylation , Epigenesis, Genetic , Female , Gene Expression Profiling , Gene Expression Regulation, Developmental , Genome, Human/genetics , Germ Layers/cytology , Germ Layers/embryology , Germ Layers/metabolism , Humans , Oocyte Donation , Oocytes/growth & development , Primary Cell Culture , Transcription, Genetic , Triploidy , Young Adult
3.
Nature ; 471(7336): 63-7, 2011 Mar 03.
Article in English | MEDLINE | ID: mdl-21368825

ABSTRACT

Defined transcription factors can induce epigenetic reprogramming of adult mammalian cells into induced pluripotent stem cells. Although DNA factors are integrated during some reprogramming methods, it is unknown whether the genome remains unchanged at the single nucleotide level. Here we show that 22 human induced pluripotent stem (hiPS) cell lines reprogrammed using five different methods each contained an average of five protein-coding point mutations in the regions sampled (an estimated six protein-coding point mutations per exome). The majority of these mutations were non-synonymous, nonsense or splice variants, and were enriched in genes mutated or having causative effects in cancers. At least half of these reprogramming-associated mutations pre-existed in fibroblast progenitors at low frequencies, whereas the rest occurred during or after reprogramming. Thus, hiPS cells acquire genetic modifications in addition to epigenetic modifications. Extensive genetic screening should become a standard procedure to ensure hiPS cell safety before clinical use.


Subject(s)
Cellular Reprogramming/genetics , Induced Pluripotent Stem Cells/metabolism , Mutagenesis/genetics , Point Mutation/genetics , Cells, Cultured , DNA Mutational Analysis , Epistasis, Genetic/genetics , Fibroblasts/cytology , Fibroblasts/metabolism , Humans , Induced Pluripotent Stem Cells/cytology , Male , Middle Aged , Models, Genetic , Open Reading Frames/genetics
4.
Nat Methods ; 9(3): 270-2, 2012 Feb 05.
Article in English | MEDLINE | ID: mdl-22306810

ABSTRACT

Targeted quantification of DNA methylation allows for interrogation of the most informative loci across many samples quickly and cost-effectively. Here we report improved bisulfite padlock probes (BSPPs) with a design algorithm to generate efficient padlock probes, a library-free protocol that dramatically reduces sample-preparation cost and time and is compatible with automation, and an efficient bioinformatics pipeline to accurately obtain both methylation levels and genotypes from sequencing of bisulfite-converted DNA.


Subject(s)
DNA Probes/chemistry , DNA Probes/genetics , DNA/chemistry , DNA/genetics , Polymerase Chain Reaction/methods , Sequence Analysis, DNA/methods , Sulfites/chemistry , Base Sequence , Gene Library , Molecular Sequence Data
5.
Proc Natl Acad Sci U S A ; 109(40): 16196-201, 2012 Oct 02.
Article in English | MEDLINE | ID: mdl-22991473

ABSTRACT

Generation of human induced pluripotent stem cells (hiPSCs) by the expression of specific transcription factors depends on successful epigenetic reprogramming to a pluripotent state. Although hiPSCs and human embryonic stem cells (hESCs) display a similar epigenome, recent reports demonstrated the persistence of specific epigenetic marks from the somatic cell type of origin and aberrant methylation patterns in hiPSCs. However, it remains unknown whether the use of different somatic cell sources, encompassing variable levels of selection pressure during reprogramming, influences the level of epigenetic aberrations in hiPSCs. In this work, we characterized the epigenomic integrity of 17 hiPSC lines derived from six different cell types with varied reprogramming efficiencies. We demonstrate that epigenetic aberrations are a general feature of the hiPSC state and are independent of the somatic cell source. Interestingly, we observe that the reprogramming efficiency of somatic cell lines inversely correlates with the amount of methylation change needed to acquire pluripotency. Additionally, we determine that both shared and line-specific epigenetic aberrations in hiPSCs can directly translate into changes in gene expression in both the pluripotent and differentiated states. Significantly, our analysis of different hiPSC lines from multiple cell types of origin allow us to identify a reprogramming-specific epigenetic signature comprised of nine aberrantly methylated genes that is able to segregate hESC and hiPSC lines regardless of the somatic cell source or differentiation state.


Subject(s)
Cellular Reprogramming/physiology , DNA Methylation/genetics , Epigenesis, Genetic/physiology , Induced Pluripotent Stem Cells/physiology , Cell Line , Cellular Reprogramming/genetics , CpG Islands/genetics , Epigenesis, Genetic/genetics , Epigenomics , Fluorescent Antibody Technique , Gene Library , Humans , Microarray Analysis , Real-Time Polymerase Chain Reaction , Sequence Analysis, DNA
6.
Proc Natl Acad Sci U S A ; 109(30): 11920-7, 2012 Jul 24.
Article in English | MEDLINE | ID: mdl-22797899

ABSTRACT

Rapid advances in DNA sequencing promise to enable new diagnostics and individualized therapies. Achieving personalized medicine, however, will require extensive research on highly reidentifiable, integrated datasets of genomic and health information. To assist with this, participants in the Personal Genome Project choose to forgo privacy via our institutional review board- approved "open consent" process. The contribution of public data and samples facilitates both scientific discovery and standardization of methods. We present our findings after enrollment of more than 1,800 participants, including whole-genome sequencing of 10 pilot participant genomes (the PGP-10). We introduce the Genome-Environment-Trait Evidence (GET-Evidence) system. This tool automatically processes genomes and prioritizes both published and novel variants for interpretation. In the process of reviewing the presumed healthy PGP-10 genomes, we find numerous literature references implying serious disease. Although it is sometimes impossible to rule out a late-onset effect, stringent evidence requirements can address the high rate of incidental findings. To that end we develop a peer production system for recording and organizing variant evaluations according to standard evidence guidelines, creating a public forum for reaching consensus on interpretation of clinically relevant variants. Genome analysis becomes a two-step process: using a prioritized list to record variant evaluations, then automatically sorting reviewed variants using these annotations. Genome data, health and trait information, participant samples, and variant interpretations are all shared in the public domain-we invite others to review our results using our participant samples and contribute to our interpretations. We offer our public resource and methods to further personalized medical research.


Subject(s)
Databases, Genetic , Genetic Variation , Genome, Human/genetics , Phenotype , Precision Medicine/methods , Software , Cell Line , Data Collection , Humans , Precision Medicine/trends , Sequence Analysis, DNA
7.
Proc Natl Acad Sci U S A ; 108(16): 6537-42, 2011 Apr 19.
Article in English | MEDLINE | ID: mdl-21464322

ABSTRACT

Gene-corrected patient-specific induced pluripotent stem (iPS) cells offer a unique approach to gene therapy. Here, we begin to assess whether the mutational load acquired during gene correction of iPS cells is compatible with use in the treatment of genetic causes of retinal degenerative disease. We isolated iPS cells free of transgene sequences from a patient with gyrate atrophy caused by a point mutation in the gene encoding ornithine-δ-aminotransferase (OAT) and used homologous recombination to correct the genetic defect. Cytogenetic analysis, array comparative genomic hybridization (aCGH), and exome sequencing were performed to assess the genomic integrity of an iPS cell line after three sequential clonal events: initial reprogramming, gene targeting, and subsequent removal of a selection cassette. No abnormalities were detected after standard G-band metaphase analysis. However, aCGH and exome sequencing identified two deletions, one amplification, and nine mutations in protein coding regions in the initial iPS cell clone. Except for the targeted correction of the single nucleotide in the OAT locus and a single synonymous base-pair change, no additional mutations or copy number variation were identified in iPS cells after the two subsequent clonal events. These findings confirm that iPS cells themselves may carry a significant mutational load at initial isolation, but that the clonal events and prolonged cultured required for correction of a genetic defect can be accomplished without a substantial increase in mutational burden.


Subject(s)
Gyrate Atrophy/enzymology , Gyrate Atrophy/genetics , Ornithine-Oxo-Acid Transaminase/genetics , Ornithine-Oxo-Acid Transaminase/metabolism , Pluripotent Stem Cells/enzymology , Cells, Cultured , Gene Targeting/methods , Genome-Wide Association Study , Genomic Instability/genetics , Gyrate Atrophy/pathology , Gyrate Atrophy/therapy , Humans , Pluripotent Stem Cells/pathology , Recombination, Genetic
8.
Hum Mutat ; 32(12): 1450-9, 2011 Dec.
Article in English | MEDLINE | ID: mdl-21901789

ABSTRACT

It has been well documented that mutations in the same retinal disease gene can result in different clinical phenotypes due to difference in the mutant allele and/or genetic background. To evaluate this, a set of consanguineous patient families with Leber congenital amaurosis (LCA) that do not carry mutations in known LCA disease genes was characterized through homozygosity mapping followed by targeted exon/whole-exome sequencing to identify genetic variations. Among these families, a total of five putative disease-causing mutations, including four novel alleles, were found for six families. These five mutations are located in four genes, ALMS1, IQCB1, CNGA3, and MYO7A. Therefore, in our LCA collection from Saudi Arabia, three of the 37 unassigned families carry mutations in retinal disease genes ALMS1, CNGA3, and MYO7A, which have not been previously associated with LCA, and 3 of the 37 carry novel mutations in IQCB1, which has been recently associated with LCA. Together with other reports, our results emphasize that the molecular heterogeneity underlying LCA, and likely other retinal diseases, may be highly complex. Thus, to obtain accurate diagnosis and gain a complete picture of the disease, it is essential to sequence a larger set of retinal disease genes and combine the clinical phenotype with molecular diagnosis.


Subject(s)
Calmodulin-Binding Proteins/genetics , Cyclic Nucleotide-Gated Cation Channels/genetics , Exome/genetics , Leber Congenital Amaurosis/genetics , Mutation , Myosins/genetics , Proteins/genetics , Cell Cycle Proteins , Child, Preschool , Chromosome Mapping , Consanguinity , DNA Mutational Analysis , Family , Homozygote , Humans , Leber Congenital Amaurosis/pathology , Myosin VIIa , Pedigree , Saudi Arabia , Sequence Analysis, DNA
9.
Nat Commun ; 11(1): 3475, 2020 07 21.
Article in English | MEDLINE | ID: mdl-32694610

ABSTRACT

Early detection has the potential to reduce cancer mortality, but an effective screening test must demonstrate asymptomatic cancer detection years before conventional diagnosis in a longitudinal study. In the Taizhou Longitudinal Study (TZL), 123,115 healthy subjects provided plasma samples for long-term storage and were then monitored for cancer occurrence. Here we report the preliminary results of PanSeer, a noninvasive blood test based on circulating tumor DNA methylation, on TZL plasma samples from 605 asymptomatic individuals, 191 of whom were later diagnosed with stomach, esophageal, colorectal, lung or liver cancer within four years of blood draw. We also assay plasma samples from an additional 223 cancer patients, plus 200 primary tumor and normal tissues. We show that PanSeer detects five common types of cancer in 88% (95% CI: 80-93%) of post-diagnosis patients with a specificity of 96% (95% CI: 93-98%), We also demonstrate that PanSeer detects cancer in 95% (95% CI: 89-98%) of asymptomatic individuals who were later diagnosed, though future longitudinal studies are required to confirm this result. These results demonstrate that cancer can be non-invasively detected up to four years before current standard of care.


Subject(s)
Circulating Tumor DNA/blood , Early Detection of Cancer/methods , Neoplasms/blood , Neoplasms/diagnosis , Adult , Aged , Aged, 80 and over , Biomarkers, Tumor/blood , China , DNA Methylation , Epigenomics , Female , Genetic Markers , Healthy Volunteers , High-Throughput Nucleotide Sequencing , Humans , Longitudinal Studies , Male , Middle Aged , Reproducibility of Results , Sensitivity and Specificity , Young Adult
10.
Cell Stem Cell ; 15(5): 634-42, 2014 Nov 06.
Article in English | MEDLINE | ID: mdl-25517467

ABSTRACT

The recent finding that reprogrammed human pluripotent stem cells can be derived by nuclear transfer into human oocytes as well as by induced expression of defined factors has revitalized the debate on whether one approach might be advantageous over the other. Here we compare the genetic and epigenetic integrity of human nuclear-transfer embryonic stem cell (NT-ESC) lines and isogenic induced pluripotent stem cell (iPSC) lines, derived from the same somatic cell cultures of fetal, neonatal, and adult origin. The two cell types showed similar genome-wide gene expression and DNA methylation profiles. Importantly, NT-ESCs and iPSCs had comparable numbers of de novo coding mutations, but significantly more than parthenogenetic ESCs. As iPSCs, NT-ESCs displayed clone- and gene-specific aberrations in DNA methylation and allele-specific expression of imprinted genes. The occurrence of these genetic and epigenetic defects in both NT-ESCs and iPSCs suggests that they are inherent to reprogramming, regardless of derivation approach.


Subject(s)
Genomic Imprinting/genetics , Mutation Rate , Nuclear Transfer Techniques , Open Reading Frames/genetics , Pluripotent Stem Cells/cytology , Adult , Base Sequence , Clone Cells , DNA Methylation/genetics , Gene Expression Regulation , Genetic Loci , Humans , Induced Pluripotent Stem Cells/cytology , Induced Pluripotent Stem Cells/metabolism , Male , Molecular Sequence Data , Pluripotent Stem Cells/metabolism
11.
Nat Biotechnol ; 31(12): 1126-32, 2013 Dec.
Article in English | MEDLINE | ID: mdl-24213699

ABSTRACT

Genome sequencing of single cells has a variety of applications, including characterizing difficult-to-culture microorganisms and identifying somatic mutations in single cells from mammalian tissues. A major hurdle in this process is the bias in amplifying the genetic material from a single cell, a procedure known as polymerase cloning. Here we describe the microwell displacement amplification system (MIDAS), a massively parallel polymerase cloning method in which single cells are randomly distributed into hundreds to thousands of nanoliter wells and their genetic material is simultaneously amplified for shotgun sequencing. MIDAS reduces amplification bias because polymerase cloning occurs in physically separated, nanoliter-scale reactors, facilitating the de novo assembly of near-complete microbial genomes from single Escherichia coli cells. In addition, MIDAS allowed us to detect single-copy number changes in primary human adult neurons at 1- to 2-Mb resolution. MIDAS can potentially further the characterization of genomic diversity in many heterogeneous cell populations.


Subject(s)
Cell Separation/instrumentation , Chromosome Mapping/instrumentation , Cloning, Molecular/methods , DNA-Directed DNA Polymerase/genetics , DNA/genetics , High-Throughput Nucleotide Sequencing/instrumentation , Nanotechnology/instrumentation , Equipment Design , Equipment Failure Analysis , High-Throughput Nucleotide Sequencing/methods , Nanotechnology/methods
12.
Cell Rep ; 5(4): 974-85, 2013 Nov 27.
Article in English | MEDLINE | ID: mdl-24239350

ABSTRACT

Presenilin 1 (PS1) is the catalytic core of γ-secretase, which cleaves type 1 transmembrane proteins, including the amyloid precursor protein (APP). PS1 also has γ-secretase-independent functions, and dominant PS1 missense mutations are the most common cause of familial Alzheimer's disease (FAD). Whether PS1 FAD mutations are gain- or loss-of-function remains controversial, primarily because most studies have relied on overexpression in mouse and/or nonneuronal systems. We used isogenic euploid human induced pluripotent stem cell lines to generate and study an allelic series of PS1 mutations, including heterozygous null mutations and homozygous and heterozygous FAD PS1 mutations. Rigorous analysis of this allelic series in differentiated, purified neurons allowed us to resolve this controversy and to conclude that FAD PS1 mutations, expressed at normal levels in the appropriate cell type, impair γ-secretase activity but do not disrupt γ-secretase-independent functions of PS1. Thus, FAD PS1 mutations do not act as simple loss of PS1 function but instead dominantly gain an activity toxic to some, but not all, PS1 functions.


Subject(s)
Amyloid Precursor Protein Secretases/genetics , Neurogenesis/genetics , Pluripotent Stem Cells/enzymology , Presenilin-1/genetics , Alzheimer Disease/genetics , Amyloid Precursor Protein Secretases/antagonists & inhibitors , Amyloid Precursor Protein Secretases/metabolism , Amyloid beta-Peptides/biosynthesis , Amyloid beta-Peptides/genetics , Amyloid beta-Protein Precursor/metabolism , Base Sequence , Cell Line , Cells, Cultured , Green Fluorescent Proteins/genetics , Humans , Mutation , Neurons/cytology , Peptide Fragments/biosynthesis , Peptide Fragments/genetics , Pluripotent Stem Cells/cytology , Protein Processing, Post-Translational , Sequence Analysis, DNA
13.
Nat Commun ; 4: 1382, 2013.
Article in English | MEDLINE | ID: mdl-23340422

ABSTRACT

Recent studies indicate that human-induced pluripotent stem cells contain genomic structural variations and point mutations in coding regions. However, these studies have focused on fibroblast-derived human induced pluripotent stem cells, and it is currently unknown whether the use of alternative somatic cell sources with varying reprogramming efficiencies would result in different levels of genetic alterations. Here we characterize the genomic integrity of eight human induced pluripotent stem cell lines derived from five different non-fibroblast somatic cell types. We show that protein-coding mutations are a general feature of the human induced pluripotent stem cell state and are independent of somatic cell source. Furthermore, we analyse a total of 17 point mutations found in human induced pluripotent stem cells and demonstrate that they do not generally facilitate the acquisition of pluripotency and thus are not likely to provide a selective advantage for reprogramming.


Subject(s)
Cellular Reprogramming/genetics , Induced Pluripotent Stem Cells/metabolism , Mutation/genetics , Open Reading Frames/genetics , Alleles , Base Sequence , Cell Line , Fibroblasts/cytology , Gene Silencing , Human Umbilical Vein Endothelial Cells , Humans , Molecular Sequence Data , Point Mutation/genetics , Retroviridae , Sequence Analysis, RNA
15.
Nat Biotechnol ; 27(4): 353-60, 2009 Apr.
Article in English | MEDLINE | ID: mdl-19330000

ABSTRACT

Current DNA methylation assays are limited in the flexibility and efficiency of characterizing a large number of genomic targets. We report a method to specifically capture an arbitrary subset of genomic targets for single-molecule bisulfite sequencing for digital quantification of DNA methylation at single-nucleotide resolution. A set of ~30,000 padlock probes was designed to assess methylation of ~66,000 CpG sites within 2,020 CpG islands on human chromosome 12, chromosome 20, and 34 selected regions. To investigate epigenetic differences associated with dedifferentiation, we compared methylation in three human fibroblast lines and eight human pluripotent stem cell lines. Chromosome-wide methylation patterns were similar among all lines studied, but cytosine methylation was slightly more prevalent in the pluripotent cells than in the fibroblasts. Induced pluripotent stem (iPS) cells appeared to display more methylation than embryonic stem cells. We found 288 regions methylated differently in fibroblasts and pluripotent cells. This targeted approach should be particularly useful for analyzing DNA methylation in large genomes.


Subject(s)
Cell Nucleus/genetics , DNA Methylation/genetics , DNA/chemistry , DNA/genetics , Gene Targeting/methods , Metabolome/genetics , Sequence Analysis, DNA/methods , Base Sequence , Humans , Molecular Sequence Data , Sulfites
SELECTION OF CITATIONS
SEARCH DETAIL