Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 21
Filter
1.
Nature ; 582(7810): 109-114, 2020 06.
Article in English | MEDLINE | ID: mdl-32494068

ABSTRACT

Advances in genetics and sequencing have identified a plethora of disease-associated and disease-causing genetic alterations. To determine causality between genetics and disease, accurate models for molecular dissection are required; however, the rapid expansion of transcriptional populations identified through single-cell analyses presents a major challenge for accurate comparisons between mutant and wild-type cells. Here we generate mouse models of human severe congenital neutropenia (SCN) using patient-derived mutations in the GFI1 transcription factor. To determine the effects of SCN mutations, we generated single-cell references for granulopoietic genomic states with linked epitopes1, aligned mutant cells to their wild-type equivalents and identified differentially expressed genes and epigenetic loci. We find that GFI1-target genes are altered sequentially, as cells go through successive states of differentiation. These insights facilitated the genetic rescue of granulocytic specification but not post-commitment defects in innate immune effector function, and underscore the importance of evaluating the effects of mutations and therapy within each relevant cell state.


Subject(s)
Disease Models, Animal , Granulocyte Precursor Cells/pathology , Mutation , Neutropenia/genetics , Neutropenia/pathology , Neutrophils/pathology , Animals , Candida albicans/immunology , Candida albicans/pathogenicity , Cell Lineage , DNA-Binding Proteins/genetics , Female , Humans , Immunity, Innate , Male , Mice , Mice, Transgenic , Neutropenia/congenital , Neutropenia/immunology , Neutrophils/immunology , Transcription Factors/genetics
2.
Nat Methods ; 18(3): 272-282, 2021 03.
Article in English | MEDLINE | ID: mdl-33589839

ABSTRACT

The paired measurement of RNA and surface proteins in single cells with cellular indexing of transcriptomes and epitopes by sequencing (CITE-seq) is a promising approach to connect transcriptional variation with cell phenotypes and functions. However, combining these paired views into a unified representation of cell state is made challenging by the unique technical characteristics of each measurement. Here we present Total Variational Inference (totalVI; https://scvi-tools.org ), a framework for end-to-end joint analysis of CITE-seq data that probabilistically represents the data as a composite of biological and technical factors, including protein background and batch effects. To evaluate totalVI's performance, we profiled immune cells from murine spleen and lymph nodes with CITE-seq, measuring over 100 surface proteins. We demonstrate that totalVI provides a cohesive solution for common analysis tasks such as dimensionality reduction, the integration of datasets with different measured proteins, estimation of correlations between molecules and differential expression testing.


Subject(s)
Lymph Nodes/metabolism , Proteins/analysis , Single-Cell Analysis/methods , Spleen/metabolism , Transcriptome/genetics , Animals , Cells, Cultured , Data Analysis , Female , High-Throughput Screening Assays/methods , Lymph Nodes/cytology , Mice , Mice, Inbred C57BL , RNA/analysis , RNA/genetics , Spleen/cytology
3.
Nat Methods ; 18(8): 903-911, 2021 08.
Article in English | MEDLINE | ID: mdl-34354295

ABSTRACT

The development of DNA-barcoded antibodies to tag cell surface molecules has enabled the use of droplet-based single-cell sequencing (dsc-seq) to profile protein abundances from thousands of cells simultaneously. As compared to flow and mass cytometry, the high per cell cost of current dsc-seq-based workflows precludes their use in clinical applications and large-scale pooled screens. Here, we introduce SCITO-seq, a workflow that uses splint oligonucleotides (oligos) to enable combinatorially indexed dsc-seq of DNA-barcoded antibodies from over 105 cells per reaction using commercial microfluidics. By encoding sample barcodes into splint oligos, we demonstrate that multiplexed SCITO-seq produces reproducible estimates of cellular composition and surface protein expression comparable to those from mass cytometry. We further demonstrate two modified splint oligo designs that extend SCITO-seq to achieve compatibility with commercial DNA-barcoded antibodies and simultaneous expression profiling of the transcriptome and surface proteins from the same cell. These results demonstrate SCITO-seq as a flexible and ultra-high-throughput platform for sequencing-based single-cell protein and multimodal profiling.


Subject(s)
Flow Cytometry/methods , High-Throughput Nucleotide Sequencing/methods , Microfluidics/methods , Sequence Analysis, RNA/methods , Single-Cell Analysis/methods , Transcriptome , Case-Control Studies , Gene Expression Profiling , Humans
4.
Proc Natl Acad Sci U S A ; 118(15)2021 04 13.
Article in English | MEDLINE | ID: mdl-33833060

ABSTRACT

Parkinson's disease is characterized by accumulation of α-synuclein (αSyn). Release of oligomeric/fibrillar αSyn from damaged neurons may potentiate neuronal death in part via microglial activation. Heretofore, it remained unknown if oligomeric/fibrillar αSyn could activate the nucleotide-binding oligomerization domain (NOD)-like receptor (NLR) family pyrin domain-containing 3 (NLRP3) inflammasome in human microglia and whether anti-αSyn antibodies could prevent this effect. Here, we show that αSyn activates the NLRP3 inflammasome in human induced pluripotent stem cell (hiPSC)-derived microglia (hiMG) via dual stimulation involving Toll-like receptor 2 (TLR2) engagement and mitochondrial damage. In vitro, hiMG can be activated by mutant (A53T) αSyn secreted from hiPSC-derived A9-dopaminergic neurons. Surprisingly, αSyn-antibody complexes enhanced rather than suppressed inflammasome-mediated interleukin-1ß (IL-1ß) secretion, indicating these complexes are neuroinflammatory in a human context. A further increase in inflammation was observed with addition of oligomerized amyloid-ß peptide (Aß) and its cognate antibody. In vivo, engraftment of hiMG with αSyn in humanized mouse brain resulted in caspase-1 activation and neurotoxicity, which was exacerbated by αSyn antibody. These findings may have important implications for antibody therapies aimed at depleting misfolded/aggregated proteins from the human brain, as they may paradoxically trigger inflammation in human microglia.


Subject(s)
Inflammasomes/metabolism , Microglia/immunology , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , Parkinson Disease/immunology , alpha-Synuclein/immunology , Amyloid beta-Peptides/immunology , Antibodies/immunology , Cell Differentiation , Cells, Cultured , Humans , Induced Pluripotent Stem Cells/cytology , Microglia/cytology , Toll-Like Receptor 2/metabolism , alpha-Synuclein/genetics
5.
Hum Mol Genet ; 25(R2): R182-R189, 2016 Oct 01.
Article in English | MEDLINE | ID: mdl-27439388

ABSTRACT

Next generation sequencing (NGS) has ignited an unprecedented pace of discovery in the biomedical sciences that is fundamentally transforming the way that we understand, diagnose and treat disease, and has motivated the belief that true precision medicine - medicine that is tailored to an individual's genetic, biochemical and exposure profile - will be a reality in the near term. With minimal sample requirement, NGS can enable the concurrent genome-wide study of genetic variations, transcriptomes, and certain epigenetic modifications. However, interrogating proteins as efficiently as DNA and RNA can be interrogated with NGS is lacking and this hampers more comprehensive views of molecular physiology and limits advances in biomedical science and precision medicine. The fact is that innovations in proteomic technologies pale in comparison to the advances in NGS, with current methodologies suffering from issues related to reproducibility, sensitivity, sample requirements, and limited multiplexing capacity. The development of proteomic technologies to overcome these limitations would fill the void in systems biology research, catalyze clinical innovations, and expedite the realization of precision medicine.

6.
Development ; 142(23): 4010-25, 2015 Dec 01.
Article in English | MEDLINE | ID: mdl-26483210

ABSTRACT

Mechanisms of initial cell fate decisions differ among species. To gain insights into lineage allocation in humans, we derived ten human embryonic stem cell lines (designated UCSFB1-10) from single blastomeres of four 8-cell embryos and one 12-cell embryo from a single couple. Compared with numerous conventional lines from blastocysts, they had unique gene expression and DNA methylation patterns that were, in part, indicative of trophoblast competence. At a transcriptional level, UCSFB lines from different embryos were often more closely related than those from the same embryo. As predicted by the transcriptomic data, immunolocalization of EOMES, T brachyury, GDF15 and active ß-catenin revealed differential expression among blastomeres of 8- to 10-cell human embryos. The UCSFB lines formed derivatives of the three germ layers and CDX2-positive progeny, from which we derived the first human trophoblast stem cell line. Our data suggest heterogeneity among early-stage blastomeres and that the UCSFB lines have unique properties, indicative of a more immature state than conventional lines.


Subject(s)
Blastomeres/cytology , Embryo Culture Techniques , Embryonic Stem Cells/cytology , Trophoblasts/cytology , Blastocyst/cytology , Cell Differentiation , Cell Line , Cell Lineage , DNA Methylation , Endoderm/metabolism , Fibroblasts/metabolism , Gene Expression Profiling , Gene Expression Regulation, Developmental , Growth Differentiation Factor 15/metabolism , Humans , Myocytes, Cardiac/cytology , Myocytes, Cardiac/metabolism , Neural Stem Cells/cytology , Oligonucleotide Array Sequence Analysis , Transcription, Genetic , Transcriptome , beta Catenin/metabolism
7.
Brain ; 140(3): 582-598, 2017 03 01.
Article in English | MEDLINE | ID: mdl-28137726

ABSTRACT

New research suggests that common pathways are altered in many neurodevelopmental disorders including autism spectrum disorder; however, little is known about early molecular events that contribute to the pathology of these diseases. The study of monogenic, neurodevelopmental disorders with a high incidence of autistic behaviours, such as fragile X syndrome, has the potential to identify genes and pathways that are dysregulated in autism spectrum disorder as well as fragile X syndrome. In vitro generation of human disease-relevant cell types provides the ability to investigate aspects of disease that are impossible to study in patients or animal models. Differentiation of human pluripotent stem cells recapitulates development of the neocortex, an area affected in both fragile X syndrome and autism spectrum disorder. We have generated induced human pluripotent stem cells from several individuals clinically diagnosed with fragile X syndrome and autism spectrum disorder. When differentiated to dorsal forebrain cell fates, our fragile X syndrome human pluripotent stem cell lines exhibited reproducible aberrant neurogenic phenotypes. Using global gene expression and DNA methylation profiling, we have analysed the early stages of neurogenesis in fragile X syndrome human pluripotent stem cells. We discovered aberrant DNA methylation patterns at specific genomic regions in fragile X syndrome cells, and identified dysregulated gene- and network-level correlates of fragile X syndrome that are associated with developmental signalling, cell migration, and neuronal maturation. Integration of our gene expression and epigenetic analysis identified altered epigenetic-mediated transcriptional regulation of a distinct set of genes in fragile X syndrome. These fragile X syndrome-aberrant networks are significantly enriched for genes associated with autism spectrum disorder, giving support to the idea that underlying similarities exist among these neurodevelopmental diseases.


Subject(s)
Cell Differentiation/genetics , Fragile X Mental Retardation Protein/genetics , Fragile X Syndrome/pathology , Gene Expression Regulation/genetics , Models, Biological , Pluripotent Stem Cells/physiology , Animals , Autism Spectrum Disorder/genetics , Autism Spectrum Disorder/pathology , Cell Movement/genetics , Cells, Cultured , DNA Methylation/genetics , Fetus , Fragile X Syndrome/genetics , Gene Regulatory Networks , Humans , Male , Mice , Neurogenesis , Transfection , Trinucleotide Repeats/genetics
8.
Nature ; 482(7384): 216-20, 2012 Jan 25.
Article in English | MEDLINE | ID: mdl-22278060

ABSTRACT

Our understanding of Alzheimer's disease pathogenesis is currently limited by difficulties in obtaining live neurons from patients and the inability to model the sporadic form of the disease. It may be possible to overcome these challenges by reprogramming primary cells from patients into induced pluripotent stem cells (iPSCs). Here we reprogrammed primary fibroblasts from two patients with familial Alzheimer's disease, both caused by a duplication of the amyloid-ß precursor protein gene (APP; termed APP(Dp)), two with sporadic Alzheimer's disease (termed sAD1, sAD2) and two non-demented control individuals into iPSC lines. Neurons from differentiated cultures were purified with fluorescence-activated cell sorting and characterized. Purified cultures contained more than 90% neurons, clustered with fetal brain messenger RNA samples by microarray criteria, and could form functional synaptic contacts. Virtually all cells exhibited normal electrophysiological activity. Relative to controls, iPSC-derived, purified neurons from the two APP(Dp) patients and patient sAD2 exhibited significantly higher levels of the pathological markers amyloid-ß(1-40), phospho-tau(Thr 231) and active glycogen synthase kinase-3ß (aGSK-3ß). Neurons from APP(Dp) and sAD2 patients also accumulated large RAB5-positive early endosomes compared to controls. Treatment of purified neurons with ß-secretase inhibitors, but not γ-secretase inhibitors, caused significant reductions in phospho-Tau(Thr 231) and aGSK-3ß levels. These results suggest a direct relationship between APP proteolytic processing, but not amyloid-ß, in GSK-3ß activation and tau phosphorylation in human neurons. Additionally, we observed that neurons with the genome of one sAD patient exhibited the phenotypes seen in familial Alzheimer's disease samples. More generally, we demonstrate that iPSC technology can be used to observe phenotypes relevant to Alzheimer's disease, even though it can take decades for overt disease to manifest in patients.


Subject(s)
Alzheimer Disease/metabolism , Alzheimer Disease/pathology , Induced Pluripotent Stem Cells/metabolism , Induced Pluripotent Stem Cells/pathology , Neurons/metabolism , Aged, 80 and over , Amyloid Precursor Protein Secretases/antagonists & inhibitors , Amyloid Precursor Protein Secretases/metabolism , Amyloid beta-Peptides/metabolism , Amyloid beta-Protein Precursor/genetics , Amyloid beta-Protein Precursor/metabolism , Astrocytes/cytology , Biomarkers/metabolism , Cells, Cultured , Cellular Reprogramming , Coculture Techniques , Endosomes/metabolism , Enzyme Activation , Female , Fibroblasts/cytology , Fibroblasts/metabolism , Glycogen Synthase Kinase 3/metabolism , Humans , Male , Middle Aged , Models, Biological , Neurons/drug effects , Neurons/pathology , Peptide Fragments/metabolism , Phosphoproteins/metabolism , Phosphorylation/drug effects , Protease Inhibitors/pharmacology , Proteolysis , Synapsins/metabolism , tau Proteins/metabolism
10.
Circ Res ; 115(2): 311-24, 2014 Jul 07.
Article in English | MEDLINE | ID: mdl-24989490

ABSTRACT

The precise, temporal order of gene expression during development is critical to ensure proper lineage commitment, cell fate determination, and ultimately, organogenesis. Epigenetic regulation of chromatin structure is fundamental to the activation or repression of genes during embryonic development. In recent years, there has been an explosion of research relating to various modes of epigenetic regulation, such as DNA methylation, post-translational histone tail modifications, noncoding RNA control of chromatin structure, and nucleosome remodeling. Technological advances in genome-wide epigenetic profiling and pluripotent stem cell differentiation have been primary drivers for elucidating the epigenetic control of cellular identity during development and nuclear reprogramming. Not only do epigenetic mechanisms regulate transcriptional states in a cell-type-specific manner but also they establish higher order genomic topology and nuclear architecture. Here, we review the epigenetic control of pluripotency and changes associated with pluripotent stem cell differentiation. We focus on DNA methylation, DNA demethylation, and common histone tail modifications. Finally, we briefly discuss epigenetic heterogeneity among pluripotent stem cell lines and the influence of epigenetic patterns on genome topology.


Subject(s)
Epigenesis, Genetic/physiology , Gene Expression Regulation, Developmental/physiology , Pluripotent Stem Cells/cytology , Acetylation , Animals , Cell Differentiation , CpG Islands , DNA (Cytosine-5-)-Methyltransferases/metabolism , DNA Methylation , Dioxygenases/metabolism , Enhancer Elements, Genetic/genetics , Enhancer Elements, Genetic/physiology , Gene Expression Regulation, Developmental/genetics , Histones/metabolism , Humans , Mice , Models, Biological , Myocytes, Cardiac/cytology , Myocytes, Cardiac/metabolism , Phosphorylation , Promoter Regions, Genetic/genetics , Promoter Regions, Genetic/physiology , Protein Processing, Post-Translational , X Chromosome Inactivation/genetics , X Chromosome Inactivation/physiology
11.
J Cell Sci ; 126(Pt 17): 3848-61, 2013 Sep 01.
Article in English | MEDLINE | ID: mdl-23813959

ABSTRACT

The differentiation of human pluripotent stem cells (hPSCs) to insulin-expressing beta islet-like cells is a promising in vitro model system for studying the molecular signaling pathways underlying beta cell differentiation, as well as a potential source of cells for the treatment of type 1 diabetes. MicroRNAs (miRNAs) are a class of small non-coding RNAs that regulate many biological processes, including cellular differentiation. We studied the miRNA and mRNA expression profiles of hPSCs at five stages of in vitro differentiation along the pancreatic beta cell lineage (definitive endoderm, primitive gut tube, posterior foregut, pancreatic progenitor and hormone-expressing endocrine cells) in the context of samples of primary human fetal pancreas and purified adult islet cells using microarray analysis. Bioinformatic analysis of the resulting data identified a unique miRNA signature in differentiated beta islet cells, and predicted the effects of key miRNAs on mRNA expression. Many of the predicted miRNA-mRNA interactions involved mRNAs known to play key roles in the epithelial-mesenchymal transition process and pancreatic differentiation. We validated a subset of the predictions using qRT-PCR, luciferase reporter assays and western blotting, including the known interaction between miR-200 and ZEB2 (involved in epithelial-mesenchymal transition) and the novel interaction between miR-200 and SOX17 (a key transcription factor in specification of definitive endoderm). In addition, we found that miR-30d and let-7e, two miRNAs induced during differentiation, regulated the expression of RFX6, a transcription factor that directs pancreatic islet formation. These findings suggest that precise control of target mRNA expression by miRNAs ensures proper lineage specification during pancreatic development.


Subject(s)
Homeodomain Proteins/metabolism , Insulin-Secreting Cells/metabolism , MicroRNAs/metabolism , Pluripotent Stem Cells/metabolism , Repressor Proteins/metabolism , SOXF Transcription Factors/metabolism , Cell Differentiation/genetics , Cell Differentiation/physiology , Cell Lineage , Cell- and Tissue-Based Therapy , Cells, Cultured , DNA-Binding Proteins/biosynthesis , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Diabetes Mellitus, Type 1/therapy , Epithelial-Mesenchymal Transition/physiology , Gene Expression Profiling , Gene Expression Regulation, Developmental , Homeodomain Proteins/biosynthesis , Homeodomain Proteins/genetics , Humans , MicroRNAs/genetics , Pancreas/embryology , RNA, Messenger/genetics , Regulatory Factor X Transcription Factors , Repressor Proteins/biosynthesis , Repressor Proteins/genetics , SOXF Transcription Factors/biosynthesis , SOXF Transcription Factors/genetics , Transcription Factors/biosynthesis , Transcription Factors/genetics , Transcription Factors/metabolism , Zinc Finger E-box Binding Homeobox 2
12.
Nature ; 461(7260): 91-4, 2009 Sep 03.
Article in English | MEDLINE | ID: mdl-19672243

ABSTRACT

Recent landmark experiments have shown that transient overexpression of a small number of transcription factors can reprogram differentiated cells into induced pluripotent stem (iPS) cells that resemble embryonic stem (ES) cells. These iPS cells hold great promise for medicine because they have the potential to generate patient-specific cell types for cell replacement therapy and produce in vitro models of disease, without requiring embryonic tissues or oocytes. Although current iPS cell lines resemble ES cells, they have not passed the most stringent test of pluripotency by generating full-term or adult mice in tetraploid complementation assays, raising questions as to whether they are sufficiently potent to generate all of the cell types in an organism. Whether this difference between iPS and ES cells reflects intrinsic limitations of direct reprogramming is not known. Here we report fertile adult mice derived entirely from iPS cells that we generated by inducible genetic reprogramming of mouse embryonic fibroblasts. Producing adult mice derived entirely from a reprogrammed fibroblast shows that all features of a differentiated cell can be restored to an embryonic level of pluripotency without exposure to unknown ooplasmic factors. Comparing these fully pluripotent iPS cell lines to less developmentally potent lines may reveal molecular markers of different pluripotent states. Furthermore, mice derived entirely from iPS cells will provide a new resource to assess the functional and genomic stability of cells and tissues derived from iPS cells, which is important to validate their utility in cell replacement therapy and research applications.


Subject(s)
Aging , Pluripotent Stem Cells/physiology , Reproductive Techniques , Animals , Cell Dedifferentiation , Cell Differentiation , Cell Line , Cell Lineage , Embryo, Mammalian/cytology , Embryo, Mammalian/embryology , Embryo, Mammalian/metabolism , Female , Fibroblasts/cytology , Male , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Pluripotent Stem Cells/cytology , Pregnancy , Survival Rate
13.
Genomics ; 104(5): 358-67, 2014 Nov.
Article in English | MEDLINE | ID: mdl-25179373

ABSTRACT

5-hydroxymethylcytosine (5hmC), an oxidized derivative of 5-methylcytosine (5mC), has been implicated as an important epigenetic regulator of mammalian development. Current procedures use DNA sequencing methods to discriminate 5hmC from 5mC, limiting their accessibility to the scientific community. Here we report a method that combines TET-assisted bisulfite conversion with Illumina 450K DNA methylation arrays for a low-cost high-throughput approach that distinguishes 5hmC and 5mC signals at base resolution. Implementing this approach, termed "TAB-array", we assessed DNA methylation dynamics in the differentiation of human pluripotent stem cells into cardiovascular progenitors and neural precursor cells. With the ability to discriminate 5mC and 5hmC, we identified a large number of novel dynamically methylated genomic regions that are implicated in the development of these lineages. The increased resolution and accuracy afforded by this approach provides a powerful means to investigate the distinct contributions of 5mC and 5hmC in human development and disease.


Subject(s)
5-Methylcytosine/metabolism , Cytosine/analogs & derivatives , Pluripotent Stem Cells/metabolism , Sequence Analysis, DNA/methods , Cell Differentiation , Cells, Cultured , Cytosine/metabolism , DNA Methylation , Epigenesis, Genetic , Humans , Molecular Sequence Data , Myoblasts, Cardiac/metabolism , Neural Stem Cells
14.
Nat Biotechnol ; 39(10): 1246-1258, 2021 10.
Article in English | MEDLINE | ID: mdl-34083792

ABSTRACT

Recent technological advances have enabled massively parallel chromatin profiling with scATAC-seq (single-cell assay for transposase accessible chromatin by sequencing). Here we present ATAC with select antigen profiling by sequencing (ASAP-seq), a tool to simultaneously profile accessible chromatin and protein levels. Our approach pairs sparse scATAC-seq data with robust detection of hundreds of cell surface and intracellular protein markers and optional capture of mitochondrial DNA for clonal tracking, capturing three distinct modalities in single cells. ASAP-seq uses a bridging approach that repurposes antibody:oligonucleotide conjugates designed for existing technologies that pair protein measurements with single-cell RNA sequencing. Together with DOGMA-seq, an adaptation of CITE-seq (cellular indexing of transcriptomes and epitopes by sequencing) for measuring gene activity across the central dogma of gene regulation, we demonstrate the utility of systematic multi-omic profiling by revealing coordinated and distinct changes in chromatin, RNA and surface proteins during native hematopoietic differentiation and peripheral blood mononuclear cell stimulation and as a combinatorial decoder and reporter of multiplexed perturbations in primary T cells.


Subject(s)
RNA-Seq/methods , Single-Cell Analysis/methods , Cell Differentiation , Cell Lineage , Chromatin/genetics , Chromatin/metabolism , DNA, Mitochondrial/genetics , Epigenomics , Gene Expression Profiling , Gene Expression Regulation , Hematopoiesis , Humans , Leukocytes, Mononuclear/cytology , Leukocytes, Mononuclear/metabolism , Proteins/genetics , Proteins/metabolism , T-Lymphocytes/cytology , T-Lymphocytes/metabolism
16.
Cell Rep ; 26(10): 2566-2579.e10, 2019 03 05.
Article in English | MEDLINE | ID: mdl-30840882

ABSTRACT

In this report we describe a human pluripotent stem cell-derived vascular progenitor (MesoT) cell of the mesothelium lineage. MesoT cells are multipotent and generate smooth muscle cells, endothelial cells, and pericytes and self-assemble into vessel-like networks in vitro. MesoT cells transplanted into mechanically damaged neonatal mouse heart migrate into the injured tissue and contribute to nascent coronary vessels in the repair zone. When seeded onto decellularized vascular scaffolds, MesoT cells differentiate into the major vascular lineages and self-assemble into vasculature capable of supporting peripheral blood flow following transplantation. These findings demonstrate in vivo functionality and the potential utility of MesoT cells in vascular engineering applications.


Subject(s)
Epithelium/metabolism , Induced Pluripotent Stem Cells/metabolism , Regenerative Medicine/methods , Tissue Engineering/methods , Cell Lineage , Humans
17.
Nat Commun ; 7: 10536, 2016 Feb 19.
Article in English | MEDLINE | ID: mdl-26892726

ABSTRACT

There is concern that the stresses of inducing pluripotency may lead to deleterious DNA mutations in induced pluripotent stem cell (iPSC) lines, which would compromise their use for cell therapies. Here we report comparative genomic analysis of nine isogenic iPSC lines generated using three reprogramming methods: integrating retroviral vectors, non-integrating Sendai virus and synthetic mRNAs. We used whole-genome sequencing and de novo genome mapping to identify single-nucleotide variants, insertions and deletions, and structural variants. Our results show a moderate number of variants in the iPSCs that were not evident in the parental fibroblasts, which may result from reprogramming. There were only small differences in the total numbers and types of variants among different reprogramming methods. Most importantly, a thorough genomic analysis showed that the variants were generally benign. We conclude that the process of reprogramming is unlikely to introduce variants that would make the cells inappropriate for therapy.


Subject(s)
DNA Mutational Analysis/methods , Fibroblasts/cytology , Genome , Genomics/methods , Induced Pluripotent Stem Cells/cytology , Mutation , Cell Differentiation , Fibroblasts/chemistry , Humans , Induced Pluripotent Stem Cells/chemistry
18.
Epigenomics ; 7(7): 1137-53, 2015 Oct.
Article in English | MEDLINE | ID: mdl-26067621

ABSTRACT

AIM: To define the DNA methylation landscape of neuroblastoma and its clinicopathological impact. MATERIALS & METHODS: Microarray DNA methylation data were analyzed and associated with functional/regulatory genome annotation data, transcriptional profiles and clinicobiological parameters. RESULTS: DNA methylation changes in neuroblastoma affect not only promoters but also intragenic and intergenic regions at cytosine-phosphate-guanine (CpG) and non-CpG sites, and target functional chromatin domains of development and cancer-related genes such as CCND1. Tumors with diverse clinical risk showed differences affecting CpG and, remarkably, non-CpG sites. Non-CpG methylation observed essentially in clinically favorable cases was associated with the differentiation status of neuroblastoma and expression of key genes such as ALK. CONCLUSION: This epigenetic fingerprint of neuroblastoma provides new insights into the pathogenesis and clinical behavior of this pediatric tumor.


Subject(s)
Brain Neoplasms/genetics , Cyclin D1/genetics , Epigenesis, Genetic , Gene Expression Regulation, Neoplastic , Neuroblastoma/genetics , Receptor Protein-Tyrosine Kinases/genetics , Anaplastic Lymphoma Kinase , Brain Neoplasms/diagnosis , Brain Neoplasms/metabolism , Brain Neoplasms/mortality , Cell Line, Tumor , Child , Child, Preschool , Chromatin/chemistry , Chromatin/metabolism , CpG Islands , Cyclin D1/metabolism , DNA Fingerprinting , DNA Methylation , DNA, Intergenic , Female , Gene Expression Profiling , Genome, Human , Humans , Infant , Male , Neuroblastoma/diagnosis , Neuroblastoma/metabolism , Neuroblastoma/mortality , Oligonucleotide Array Sequence Analysis , Prognosis , Promoter Regions, Genetic , Receptor Protein-Tyrosine Kinases/metabolism , Survival Analysis
19.
J Vis Exp ; (69): e4003, 2012 Nov 29.
Article in English | MEDLINE | ID: mdl-23222420

ABSTRACT

The production of induced pluripotent stem cells (iPSCs) from somatic cells provides a means to create valuable tools for basic research and may also produce a source of patient-matched cells for regenerative therapies. iPSCs may be generated using multiple protocols and derived from multiple cell sources. Once generated, iPSCs are tested using a variety of assays including immunostaining for pluripotency markers, generation of three germ layers in embryoid bodies and teratomas, comparisons of gene expression with embryonic stem cells (ESCs) and production of chimeric mice with or without germline contribution(2). Importantly, iPSC lines that pass these tests still vary in their capacity to produce different differentiated cell types(2). This has made it difficult to establish which iPSC derivation protocols, donor cell sources or selection methods are most useful for different applications. The most stringent test of whether a stem cell line has sufficient developmental potential to generate all tissues required for survival of an organism (termed full pluripotency) is tetraploid embryo complementation (TEC)(3-5). Technically, TEC involves electrofusion of two-cell embryos to generate tetraploid (4n) one-cell embryos that can be cultured in vitro to the blastocyst stage(6). Diploid (2n) pluripotent stem cells (e.g. ESCs or iPSCs) are then injected into the blastocoel cavity of the tetraploid blastocyst and transferred to a recipient female for gestation (see Figure 1). The tetraploid component of the complemented embryo contributes almost exclusively to the extraembryonic tissues (placenta, yolk sac), whereas the diploid cells constitute the embryo proper, resulting in a fetus derived entirely from the injected stem cell line. Recently, we reported the derivation of iPSC lines that reproducibly generate adult mice via TEC(1). These iPSC lines give rise to viable pups with efficiencies of 5-13%, which is comparable to ESCs(3,4,7) and higher than that reported for most other iPSC lines(8-12). These reports show that direct reprogramming can produce fully pluripotent iPSCs that match ESCs in their developmental potential and efficiency of generating pups in TEC tests. At present, it is not clear what distinguishes between fully pluripotent iPSCs and less potent lines(13-15). Nor is it clear which reprogramming methods will produce these lines with the highest efficiency. Here we describe one method that produces fully pluripotent iPSCs and "all- iPSC" mice, which may be helpful for investigators wishing to compare the pluripotency of iPSC lines or establish the equivalence of different reprogramming methods.


Subject(s)
Pluripotent Stem Cells/cytology , Animals , Cytological Techniques/methods , Female , Mice , Pregnancy , Stem Cell Research
20.
Cell Stem Cell ; 10(5): 620-34, 2012 May 04.
Article in English | MEDLINE | ID: mdl-22560082

ABSTRACT

Human pluripotent stem cells (hPSCs) are potential sources of cells for modeling disease and development, drug discovery, and regenerative medicine. However, it is important to identify factors that may impact the utility of hPSCs for these applications. In an unbiased analysis of 205 hPSC and 130 somatic samples, we identified hPSC-specific epigenetic and transcriptional aberrations in genes subject to X chromosome inactivation (XCI) and genomic imprinting, which were not corrected during directed differentiation. We also found that specific tissue types were distinguished by unique patterns of DNA hypomethylation, which were recapitulated by DNA demethylation during in vitro directed differentiation. Our results suggest that verification of baseline epigenetic status is critical for hPSC-based disease models in which the observed phenotype depends on proper XCI or imprinting and that tissue-specific DNA methylation patterns can be accurately modeled during directed differentiation of hPSCs, even in the presence of variations in XCI or imprinting.


Subject(s)
Genetic Variation , Pluripotent Stem Cells/physiology , Cell Differentiation , Cells, Cultured , Chromosome Aberrations , Chromosomes, Human, X , DNA Methylation , Genomic Imprinting , Humans , Organ Specificity , Recurrence , Stem Cell Niche , X Chromosome Inactivation
SELECTION OF CITATIONS
SEARCH DETAIL