Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 57
Filter
Add more filters










Publication year range
1.
Stem Cell Rev Rep ; 19(8): 2980-2990, 2023 11.
Article in English | MEDLINE | ID: mdl-37702917

ABSTRACT

Embryonic development is a continuum in vivo. Transcriptional analysis can separate established human embryonic stem cells (hESC) into at least four distinct developmental pluripotent stages, two naïve and two primed, early and late relative to the intact epiblast. In this study we primarily show that exposure of frozen human blastocysts to an inhibitor of checkpoint kinase 1 (CHK1) upon thaw greatly enhances establishment of karyotypically normal late naïve hESC cultures. These late naïve cells are plastic and can be toggled back to early naïve and forward to early primed pluripotent stages. The early primed cells are transcriptionally equivalent to the post inner cell mass intermediate (PICMI) stage seen one day following transfer of human blastocysts into in vitro culture and are stable at an earlier stage than conventional primed hESC.


Subject(s)
Cell Culture Techniques , Checkpoint Kinase 1 , Human Embryonic Stem Cells , Human Embryonic Stem Cells/cytology , Human Embryonic Stem Cells/drug effects , Humans , Checkpoint Kinase 1/antagonists & inhibitors , Protein Kinase Inhibitors/pharmacology , Blastocyst/cytology , Pluripotent Stem Cells/cytology
2.
PLoS Comput Biol ; 16(9): e1008173, 2020 09.
Article in English | MEDLINE | ID: mdl-32946435

ABSTRACT

Single-cell Hi-C (scHi-C) interrogates genome-wide chromatin interaction in individual cells, allowing us to gain insights into 3D genome organization. However, the extremely sparse nature of scHi-C data poses a significant barrier to analysis, limiting our ability to tease out hidden biological information. In this work, we approach this problem by applying topic modeling to scHi-C data. Topic modeling is well-suited for discovering latent topics in a collection of discrete data. For our analysis, we generate nine different single-cell combinatorial indexed Hi-C (sci-Hi-C) libraries from five human cell lines (GM12878, H1Esc, HFF, IMR90, and HAP1), consisting over 19,000 cells. We demonstrate that topic modeling is able to successfully capture cell type differences from sci-Hi-C data in the form of "chromatin topics." We further show enrichment of particular compartment structures associated with locus pairs in these topics.


Subject(s)
Chromatin , Computational Biology/methods , High-Throughput Nucleotide Sequencing/methods , Single-Cell Analysis/methods , Cell Line , Chromatin/chemistry , Chromatin/genetics , Cluster Analysis , Gene Library , Humans , Natural Language Processing
3.
Dev Cell ; 52(2): 236-250.e7, 2020 Jan 27.
Article in English | MEDLINE | ID: mdl-31991105

ABSTRACT

Regulation of embryonic diapause, dormancy that interrupts the tight connection between developmental stage and time, is still poorly understood. Here, we characterize the transcriptional and metabolite profiles of mouse diapause embryos and identify unique gene expression and metabolic signatures with activated lipolysis, glycolysis, and metabolic pathways regulated by AMPK. Lipolysis is increased due to mTORC2 repression, increasing fatty acids to support cell survival. We further show that starvation in pre-implantation ICM-derived mouse ESCs induces a reversible dormant state, transcriptionally mimicking the in vivo diapause stage. During starvation, Lkb1, an upstream kinase of AMPK, represses mTOR, which induces a reversible glycolytic and epigenetically H4K16Ac-negative, diapause-like state. Diapause furthermore activates expression of glutamine transporters SLC38A1/2. We show by genetic and small molecule inhibitors that glutamine transporters are essential for the H4K16Ac-negative, diapause state. These data suggest that mTORC1/2 inhibition, regulated by amino acid levels, is causal for diapause metabolism and epigenetic state.


Subject(s)
Amino Acid Transport System A/metabolism , Blastocyst/metabolism , Embryo, Mammalian/cytology , Mechanistic Target of Rapamycin Complex 2/metabolism , Protein Serine-Threonine Kinases/metabolism , AMP-Activated Protein Kinases , Animals , Cell Proliferation/genetics , Cell Proliferation/physiology , Embryonic Stem Cells/cytology , Gene Knockout Techniques , Mice
4.
iScience ; 19: 450-460, 2019 Sep 27.
Article in English | MEDLINE | ID: mdl-31430690

ABSTRACT

Efficient stem cell differentiation into pancreatic islet cells is of critical importance for the development of cell replacement therapies for diabetes. Here, we identify the expression pattern of connexin 43 (Cx43), a gap junction (GJ) channel protein, in human embryonic stem cell (hESC)-derived definitive endoderm (DE) and primitive gut tube cells, representing early lineages for posterior foregut (PF), pancreatic progenitors (PP), pancreatic endocrine progenitors (PE), and islet cells. As the function of GJ channels is dependent on their gating status, we tested the impact of supplementing hESC-derived PP cell cultures with AAP10, a peptide that promotes Cx43 GJ channel opening. We found that this treatment promotes the expression of DE markers FoxA2 and Sox17, leads to a more efficient derivation of DE, and improves the yield of PF, PP, and PE cells. These results demonstrate a functional involvement of GJ channels in the differentiation of embryonic stem cells into pancreatic cell lineages.

5.
Stem Cell Reports ; 12(5): 1129-1144, 2019 05 14.
Article in English | MEDLINE | ID: mdl-31056477

ABSTRACT

During mammalian embryogenesis, changes in morphology and gene expression are concurrent with epigenomic reprogramming. Using human embryonic stem cells representing the preimplantation blastocyst (naive) and postimplantation epiblast (primed), our data in 2iL/I/F naive cells demonstrate that a substantial portion of known human enhancers are premarked by H3K4me1, providing an enhanced open chromatin state in naive pluripotency. The 2iL/I/F enhancer repertoire occupies 9% of the genome, three times that of primed cells, and can exist in broad chromatin domains over 50 kb. Enhancer chromatin states are largely poised. Seventy-seven percent of 2iL/I/F enhancers are decommissioned in a stepwise manner as cells become primed. While primed topologically associating domains are largely unaltered upon differentiation, naive 2iL/I/F domains expand across primed boundaries, affecting three-dimensional genome architecture. Differential topologically associating domain edges coincide with 2iL/I/F H3K4me1 enrichment. Our results suggest that naive-derived 2iL/I/F cells have a unique chromatin landscape, which may reflect early embryogenesis.


Subject(s)
Blastocyst/metabolism , Chromatin/genetics , Enhancer Elements, Genetic/genetics , Germ Layers/metabolism , Human Embryonic Stem Cells/metabolism , Animals , Blastocyst/cytology , Cell Differentiation/genetics , Embryo, Mammalian/cytology , Embryo, Mammalian/embryology , Embryo, Mammalian/metabolism , Embryonic Development/genetics , Gene Expression Regulation, Developmental , Germ Layers/cytology , Human Embryonic Stem Cells/cytology , Humans
6.
Nucleic Acids Res ; 47(12): e69, 2019 07 09.
Article in English | MEDLINE | ID: mdl-30927002

ABSTRACT

Chromatin immunoprecipitation (ChIP) is the most widely used approach for identification of genome-associated proteins and their modifications. We have previously introduced a microplate-based ChIP platform, Matrix ChIP, where the entire ChIP procedure is done on the same plate without sample transfers. Compared to conventional ChIP protocols, the Matrix ChIP assay is faster and has increased throughput. However, even with microplate ChIP assays, sample preparation and chromatin fragmentation (which is required to map genomic locations) remains a major bottleneck. We have developed a novel technology (termed 'PIXUL') utilizing an array of ultrasound transducers for simultaneous shearing of samples in standard 96-well microplates. We integrated PIXUL with Matrix ChIP ('PIXUL-ChIP'), that allows for fast, reproducible, low-cost and high-throughput sample preparation and ChIP analysis of 96 samples (cell culture or tissues) in one day. Further, we demonstrated that chromatin prepared using PIXUL can be used in an existing ChIP-seq workflow. Thus, the high-throughput capacity of PIXUL-ChIP provides the means to carry out ChIP-qPCR or ChIP-seq experiments involving dozens of samples. Given the complexity of epigenetic processes, the use of PIXUL-ChIP will advance our understanding of these processes in health and disease, as well as facilitate screening of epigenetic drugs.


Subject(s)
Chromatin Immunoprecipitation/methods , Epigenesis, Genetic , Animals , Cell Line , Chromatin/radiation effects , DNA/radiation effects , Embryonic Stem Cells/metabolism , Female , Humans , Male , Mice, Inbred C57BL , RNA Polymerase II/analysis , Ultrasonic Waves
7.
Cell Cycle ; 17(5): 535-549, 2018.
Article in English | MEDLINE | ID: mdl-29466914

ABSTRACT

To easily edit the genome of naïve human embryonic stem cells (hESC), we introduced a dual cassette encoding an inducible Cas9 into the AAVS1 site of naïve hESC (iCas9). The iCas9 line retained karyotypic stability, expression of pluripotency markers, differentiation potential, and stability in 5iLA and EPS pluripotency conditions. The iCas9 line induced efficient homology-directed repair (HDR) and non-homologous end joining (NHEJ) based mutations through CRISPR-Cas9 system. We utilized the iCas9 line to study the epigenetic regulator, PRC2 in early human pluripotency. The PRC2 requirement distinguishes between early pluripotency stages, however, what regulates PRC2 activity in these stages is not understood. We show reduced H3K27me3 and pluripotency markers in JARID2 2iL-I-F hESC mutants, indicating JARID2 requirement in maintenance of hESC 2iL-I-F state. These data suggest that JARID2 regulates PRC2 in 2iL-I-F state and the lack of PRC2 function in 5iLA state may be due to lack of sufficient JARID2 protein.


Subject(s)
CRISPR-Cas Systems/genetics , Gene Editing , Polycomb Repressive Complex 2/metabolism , Blastocyst/cytology , Blastocyst/metabolism , Cell Self Renewal , DNA End-Joining Repair , Genetic Loci , Histones/metabolism , Human Embryonic Stem Cells , Humans , INDEL Mutation , Microscopy, Confocal , Phenotype , Polycomb Repressive Complex 2/chemistry , Polycomb Repressive Complex 2/deficiency , Polycomb Repressive Complex 2/genetics , Presenilin-2/genetics , Presenilin-2/metabolism , Protein Domains
8.
Methods ; 142: 59-73, 2018 06 01.
Article in English | MEDLINE | ID: mdl-29382556

ABSTRACT

The folding and three-dimensional (3D) organization of chromatin in the nucleus critically impacts genome function. The past decade has witnessed rapid advances in genomic tools for delineating 3D genome architecture. Among them, chromosome conformation capture (3C)-based methods such as Hi-C are the most widely used techniques for mapping chromatin interactions. However, traditional Hi-C protocols rely on restriction enzymes (REs) to fragment chromatin and are therefore limited in resolution. We recently developed DNase Hi-C for mapping 3D genome organization, which uses DNase I for chromatin fragmentation. DNase Hi-C overcomes RE-related limitations associated with traditional Hi-C methods, leading to improved methodological resolution. Furthermore, combining this method with DNA capture technology provides a high-throughput approach (targeted DNase Hi-C) that allows for mapping fine-scale chromatin architecture at exceptionally high resolution. Hence, targeted DNase Hi-C will be valuable for delineating the physical landscapes of cis-regulatory networks that control gene expression and for characterizing phenotype-associated chromatin 3D signatures. Here, we provide a detailed description of method design and step-by-step working protocols for these two methods.


Subject(s)
Chromosome Mapping/methods , Deoxyribonuclease I/metabolism , High-Throughput Nucleotide Sequencing/methods , Imaging, Three-Dimensional/methods , Molecular Imaging/methods , Cell Culture Techniques/instrumentation , Cell Culture Techniques/methods , Cell Nucleus/genetics , Cell Nucleus/metabolism , Chromatin/chemistry , Chromatin/genetics , Chromosome Mapping/instrumentation , Cross-Linking Reagents/chemistry , DNA Restriction Enzymes/chemistry , DNA Restriction Enzymes/metabolism , Deoxyribonuclease I/chemistry , Formaldehyde/chemistry , Gene Library , High-Throughput Nucleotide Sequencing/instrumentation , Imaging, Three-Dimensional/instrumentation , Molecular Imaging/instrumentation , Tissue Culture Techniques/instrumentation , Tissue Culture Techniques/methods , Whole Genome Sequencing/instrumentation , Whole Genome Sequencing/methods
9.
PLoS Genet ; 13(11): e1007060, 2017 Nov.
Article in English | MEDLINE | ID: mdl-29107996

ABSTRACT

In storing and transmitting epigenetic information, organisms must balance the need to maintain information about past conditions with the capacity to respond to information in their current and future environments. Some of this information is encoded by DNA methylation, which can be transmitted with variable fidelity from parent to daughter strand. High fidelity confers strong pattern matching between the strands of individual DNA molecules and thus pattern stability over rounds of DNA replication; lower fidelity confers reduced pattern matching, and thus greater flexibility. Here, we present a new conceptual framework, Ratio of Concordance Preference (RCP), that uses double-stranded methylation data to quantify the flexibility and stability of the system that gave rise to a given set of patterns. We find that differentiated mammalian cells operate with high DNA methylation stability, consistent with earlier reports. Stem cells in culture and in embryos, in contrast, operate with reduced, albeit significant, methylation stability. We conclude that preference for concordant DNA methylation is a consistent mode of information transfer, and thus provides epigenetic stability across cell divisions, even in stem cells and those undergoing developmental transitions. Broader application of our RCP framework will permit comparison of epigenetic-information systems across cells, developmental stages, and organisms whose methylation machineries differ substantially or are not yet well understood.


Subject(s)
Cell Differentiation , DNA Methylation , Epigenesis, Genetic , Animals , CCAAT-Enhancer-Binding Proteins , Cells, Cultured , DNA (Cytosine-5-)-Methyltransferase 1/genetics , DNA (Cytosine-5-)-Methyltransferase 1/metabolism , DNA Replication , Embryonic Stem Cells/cytology , Female , Fibroblasts/cytology , Genetic Loci , Humans , Male , Mice , Nuclear Proteins/genetics , Nuclear Proteins/metabolism , Repressor Proteins/genetics , Repressor Proteins/metabolism , Ubiquitin-Protein Ligases
10.
Stem Cells ; 35(1): 35-41, 2017 01.
Article in English | MEDLINE | ID: mdl-27663171

ABSTRACT

The naïve state of pluripotency is actively being explored by a number of labs. There is some controversy in the field as to the true identity of naïve human pluripotent cells as they are not exact mirrors of the mouse. The various reports published, although in basic agreement, present discrepancies in the characterization of the various lines, which likely reflect the etiology of these lines. The primary lesson learned from these contributions is that a human naïve state reflecting the preimplantation human is likely to exist. The essential factors that will universally maintain the naïve state in human cells in vitro are not yet fully understood. These first need to be identified in order to describe the definitive characteristics of this state. Comparisons of naïve and primed human pluripotent cells have also highlighted consistencies between states and broadened our understanding of embryonic metabolism, epigenetic change required for development, embryonic DNA repair strategies and embryonic expression dynamics. Stem Cells 2017;35:35-41.


Subject(s)
Pluripotent Stem Cells/cytology , Cell Line , Epigenesis, Genetic , Humans , Pluripotent Stem Cells/metabolism
11.
J Cell Physiol ; 231(9): 2040-7, 2016 09.
Article in English | MEDLINE | ID: mdl-26773436

ABSTRACT

The lineage relationship between prostate adenocarcinoma and small cell carcinoma was studied by using the LuCaP family of xenografts established from primary neoplasm to metastasis. Expression of four stem cell transcription factor (TF) genes, LIN28A, NANOG, POU5F1, SOX2, were analyzed in the LuCaP lines. These genes, when force expressed in differentiated cells, can reprogram the recipients into stem-like induced pluripotent stem (iPS) cells. Most LuCaP lines expressed POU5F1, while LuCaP 145.1, representative of small cell carcinoma, expressed all four. Through transcriptome database query, many small cell carcinoma genes were also found in stem cells. To test the hypothesis that prostate cancer progression from "differentiated" adenocarcinoma to "undifferentiated" small cell carcinoma could involve re-expression of stem cell genes, the four TF genes were transduced via lentiviral vectors into five adenocarcinoma LuCaP lines-70CR, 73CR, 86.2, 92, 105CR-as done in iPS cell reprogramming. The resultant cells from these five transductions displayed a morphology of small size and dark appearing unlike the parentals. Transcriptome analysis of LuCaP 70CR* ("*" to denote transfected progeny) revealed a unique gene expression close to that of LuCaP 145.1. In a prostate principal components analysis space based on cell-type transcriptomes, the different LuCaP transcriptome datapoints were aligned to suggest a possible ordered sequence of expression changes from the differentiated luminal-like adenocarcinoma cell types to the less differentiated, more stem-like small cell carcinoma types, and LuCaP 70CR*. Prostate cancer progression can thus be molecularly characterized by loss of differentiation with re-expression of stem cell genes. J. Cell. Physiol. 231: 2040-2047, 2016. © 2016 Wiley Periodicals, Inc.


Subject(s)
Adenocarcinoma/metabolism , Carcinoma, Small Cell/metabolism , Cell Differentiation/genetics , Induced Pluripotent Stem Cells/cytology , Prostate/metabolism , Prostatic Neoplasms/metabolism , Cell Differentiation/physiology , Cell Line, Tumor , Cellular Reprogramming , Gene Expression Profiling/methods , Genes, Homeobox/genetics , Humans , Male , Prostate/pathology , Prostatic Neoplasms/pathology , Xenograft Model Antitumor Assays/methods
12.
Nat Cell Biol ; 17(12): 1523-35, 2015 Dec.
Article in English | MEDLINE | ID: mdl-26571212

ABSTRACT

For nearly a century developmental biologists have recognized that cells from embryos can differ in their potential to differentiate into distinct cell types. Recently, it has been recognized that embryonic stem cells derived from both mice and humans exhibit two stable yet epigenetically distinct states of pluripotency: naive and primed. We now show that nicotinamide N-methyltransferase (NNMT) and the metabolic state regulate pluripotency in human embryonic stem cells (hESCs).  Specifically, in naive hESCs, NNMT and its enzymatic product 1-methylnicotinamide are highly upregulated, and NNMT is required for low S-adenosyl methionine (SAM) levels and the H3K27me3 repressive state. NNMT consumes SAM in naive cells, making it unavailable for histone methylation that represses Wnt and activates the HIF pathway in primed hESCs. These data support the hypothesis that the metabolome regulates the epigenetic landscape of the earliest steps in human development.


Subject(s)
Cell Differentiation , Epigenesis, Genetic/genetics , Human Embryonic Stem Cells/metabolism , Metabolome , Animals , Blotting, Western , Cells, Cultured , Embryonic Stem Cells/metabolism , Gas Chromatography-Mass Spectrometry , Gene Expression Profiling/methods , Gene Knockdown Techniques , Histones/metabolism , Humans , Lysine/metabolism , Mass Spectrometry , Metabolomics/methods , Methylation , Mice , Niacinamide/analogs & derivatives , Niacinamide/metabolism , Nicotinamide N-Methyltransferase/genetics , Nicotinamide N-Methyltransferase/metabolism , Proteomics/methods , Reverse Transcriptase Polymerase Chain Reaction , S-Adenosylmethionine/metabolism , Signal Transduction
13.
Stem Cells ; 33(9): 2674-85, 2015 Sep.
Article in English | MEDLINE | ID: mdl-26013465

ABSTRACT

During early patterning of the neural plate, a single region of the embryonic forebrain, the eye field, becomes competent for eye development. The hallmark of eye field specification is the expression of the eye field transcription factors (EFTFs). Experiments in fish, amphibians, birds, and mammals have demonstrated largely conserved roles for the EFTFs. Although some of the key signaling events that direct the synchronized expression of these factors to the eye field have been elucidated in fish and frogs, it has been more difficult to study these mechanisms in mammalian embryos. In this study, we have used two different methods for directed differentiation of mouse embryonic stem cells (mESCs) to generate eye field cells and retina in vitro to test for a role of the PDZ domain-containing protein GIPC1 in the specification of the mammalian eye primordia. We find that the overexpression of a dominant-negative form of GIPC1 (dnGIPC1), as well as the downregulation of endogenous GIPC1, is sufficient to inhibit the development of eye field cells from mESCs. GIPC1 interacts directly with IGFR and participates in Akt1 activation, and pharmacological inhibition of Akt1 phosphorylation mimics the dnGIPC1 phenotype. Our data, together with previous studies in Xenopus, support the hypothesis that the GIPC1-PI3K-Akt1 pathway plays a key role in eye field specification in vertebrates.


Subject(s)
Adaptor Proteins, Signal Transducing/biosynthesis , Eye Proteins/biosynthesis , Mouse Embryonic Stem Cells/metabolism , Proto-Oncogene Proteins c-akt/biosynthesis , Retina/metabolism , Signal Transduction/physiology , Animals , Cell Differentiation/physiology , HEK293 Cells , Humans , Mice , Retina/cytology , Xenopus laevis
14.
Nature ; 522(7555): 221-5, 2015 Jun 11.
Article in English | MEDLINE | ID: mdl-25896322

ABSTRACT

Endogenous retroviruses (ERVs) are remnants of ancient retroviral infections, and comprise nearly 8% of the human genome. The most recently acquired human ERV is HERVK(HML-2), which repeatedly infected the primate lineage both before and after the divergence of the human and chimpanzee common ancestor. Unlike most other human ERVs, HERVK retained multiple copies of intact open reading frames encoding retroviral proteins. However, HERVK is transcriptionally silenced by the host, with the exception of in certain pathological contexts such as germ-cell tumours, melanoma or human immunodeficiency virus (HIV) infection. Here we demonstrate that DNA hypomethylation at long terminal repeat elements representing the most recent genomic integrations, together with transactivation by OCT4 (also known as POU5F1), synergistically facilitate HERVK expression. Consequently, HERVK is transcribed during normal human embryogenesis, beginning with embryonic genome activation at the eight-cell stage, continuing through the emergence of epiblast cells in preimplantation blastocysts, and ceasing during human embryonic stem cell derivation from blastocyst outgrowths. Remarkably, we detected HERVK viral-like particles and Gag proteins in human blastocysts, indicating that early human development proceeds in the presence of retroviral products. We further show that overexpression of one such product, the HERVK accessory protein Rec, in a pluripotent cell line is sufficient to increase IFITM1 levels on the cell surface and inhibit viral infection, suggesting at least one mechanism through which HERVK can induce viral restriction pathways in early embryonic cells. Moreover, Rec directly binds a subset of cellular RNAs and modulates their ribosome occupancy, indicating that complex interactions between retroviral proteins and host factors can fine-tune pathways of early human development.


Subject(s)
Blastocyst/virology , Endogenous Retroviruses/metabolism , Pluripotent Stem Cells/virology , Virus Activation , Antigens, Differentiation/metabolism , Blastocyst/cytology , Blastocyst/metabolism , Cell Line , DNA Methylation , Endogenous Retroviruses/genetics , Female , Gene Products, gag/metabolism , Humans , Male , Octamer Transcription Factor-3/metabolism , Open Reading Frames/genetics , Pluripotent Stem Cells/cytology , Pluripotent Stem Cells/metabolism , RNA, Messenger/genetics , RNA, Messenger/metabolism , Ribosomes/genetics , Ribosomes/metabolism , Terminal Repeat Sequences/genetics , Transcription, Genetic/genetics , Transcriptional Activation , Viral Envelope Proteins/genetics , Viral Envelope Proteins/metabolism
15.
Curr Urol Rep ; 16(1): 468, 2015 Jan.
Article in English | MEDLINE | ID: mdl-25404182

ABSTRACT

Prostate cancer progression is characterized by tumor dedifferentiation. Cancer cells of less differentiated tumors have a gene expression/transcriptome more similar to that of stem cells. In dedifferentiation, cancer cells may follow a specific program of gene expression changes to a stem-like state. In order to treat cancer effectively, the stem-like cancer cells and cancer differentiation pathway need to be identified and studied. Due to the very low abundance of stem-like cancer cells, their isolation from fresh human tumors is technically challenging. Induced pluripotent stem cell technology can reprogram differentiated cells into stem-like, and this may be a tool to generate sufficient stem-like cancer cells.


Subject(s)
Cell Dedifferentiation , Cellular Reprogramming , Induced Pluripotent Stem Cells , Neoplastic Stem Cells/cytology , Prostate/cytology , Prostatic Neoplasms , Cell Line, Tumor , Humans , Male , Principal Component Analysis
16.
Nat Methods ; 12(1): 71-8, 2015 Jan.
Article in English | MEDLINE | ID: mdl-25437436

ABSTRACT

High-throughput methods based on chromosome conformation capture have greatly advanced our understanding of the three-dimensional (3D) organization of genomes but are limited in resolution by their reliance on restriction enzymes. Here we describe a method called DNase Hi-C for comprehensively mapping global chromatin contacts. DNase Hi-C uses DNase I for chromatin fragmentation, leading to greatly improved efficiency and resolution over that of Hi-C. Coupling this method with DNA-capture technology provides a high-throughput approach for targeted mapping of fine-scale chromatin architecture. We applied targeted DNase Hi-C to characterize the 3D organization of 998 large intergenic noncoding RNA (lincRNA) promoters in two human cell lines. Our results revealed that expression of lincRNAs is tightly controlled by complex mechanisms involving both super-enhancers and the Polycomb repressive complex. Our results provide the first glimpse of the cell type-specific 3D organization of lincRNA genes.


Subject(s)
Chromatin/physiology , RNA, Untranslated/genetics , Chromatin/chemistry , Chromatin/ultrastructure , Chromosome Mapping , Deoxyribonuclease I/metabolism , Genome , Humans , K562 Cells , Protein Conformation , Regulatory Elements, Transcriptional/genetics
18.
Proc Natl Acad Sci U S A ; 111(12): 4484-9, 2014 Mar 25.
Article in English | MEDLINE | ID: mdl-24623855

ABSTRACT

The naïve pluripotent state has been shown in mice to lead to broad and more robust developmental potential relative to primed mouse epiblast cells. The human naïve ES cell state has eluded derivation without the use of transgenes, and forced expression of OCT4, KLF4, and KLF2 allows maintenance of human cells in a naïve state [Hanna J, et al. (2010) Proc Natl Acad Sci USA 107(20):9222-9227]. We describe two routes to generate nontransgenic naïve human ES cells (hESCs). The first is by reverse toggling of preexisting primed hESC lines by preculture in the histone deacetylase inhibitors butyrate and suberoylanilide hydroxamic acid, followed by culture in MEK/ERK and GSK3 inhibitors (2i) with FGF2. The second route is by direct derivation from a human embryo in 2i with FGF2. We show that human naïve cells meet mouse criteria for the naïve state by growth characteristics, antibody labeling profile, gene expression, X-inactivation profile, mitochondrial morphology, microRNA profile and development in the context of teratomas. hESCs can exist in a naïve state without the need for transgenes. Direct derivation is an elusive, but attainable, process, leading to cells at the earliest stage of in vitro pluripotency described for humans. Reverse toggling of primed cells to naïve is efficient and reproducible.


Subject(s)
Embryonic Stem Cells/cytology , Animals , Cell Lineage , Cells, Cultured , Embryonic Stem Cells/metabolism , Gene Expression Profiling , Glycogen Synthase Kinase 3/antagonists & inhibitors , Histone Deacetylase Inhibitors/pharmacology , Humans , Kruppel-Like Factor 4 , Mice , Protein Kinase Inhibitors/pharmacology , Transgenes , X Chromosome Inactivation
19.
Stem Cells ; 31(9): 1737-48, 2013 Sep.
Article in English | MEDLINE | ID: mdl-23765801

ABSTRACT

Adult stem cells reside in hypoxic niches, and embryonic stem cells (ESCs) are derived from a low oxygen environment. However, it is not clear whether hypoxia is critical for stem cell fate since for example human ESCs (hESCs) are able to self-renew in atmospheric oxygen concentrations as well. We now show that hypoxia can govern cell fate decisions since hypoxia alone can revert hESC- or iPSC-derived differentiated cells back to a stem cell-like state, as evidenced by re-activation of an Oct4-promoter reporter. Hypoxia-induced "de-differentiated" cells also mimic hESCs in their morphology, long-term self-renewal capacity, genome-wide mRNA and miRNA profiles, Oct4 promoter methylation state, cell surface markers TRA1-60 and SSEA4 expression, and capacity to form teratomas. These data demonstrate that hypoxia can influence cell fate decisions and could elucidate hypoxic niche function.


Subject(s)
Cell Lineage , Pluripotent Stem Cells/cytology , Adult , Animals , Basic Helix-Loop-Helix Transcription Factors/metabolism , Biomarkers/metabolism , Cell Dedifferentiation/drug effects , Cell Hypoxia/drug effects , Cell Line , Cell Lineage/drug effects , Embryonic Stem Cells/cytology , Embryonic Stem Cells/drug effects , Embryonic Stem Cells/metabolism , Green Fluorescent Proteins/metabolism , Histone Deacetylases/metabolism , Humans , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Mice , Models, Biological , Octamer Transcription Factor-3/metabolism , Oxygen/pharmacology , Pluripotent Stem Cells/drug effects , Pluripotent Stem Cells/metabolism
20.
Mol Cell Biol ; 33(9): 1845-58, 2013 May.
Article in English | MEDLINE | ID: mdl-23459939

ABSTRACT

During development, a small but significant number of CpG islands (CGIs) become methylated. The timing of developmentally programmed CGI methylation and associated mechanisms of transcriptional regulation during cellular differentiation, however, remain poorly characterized. Here, we used genome-wide DNA methylation microarrays to identify epigenetic changes during human embryonic stem cell (hESC) differentiation. We discovered a group of CGIs associated with developmental genes that gain methylation after hESCs differentiate. Conversely, erasure of methylation was observed at the identified CGIs during subsequent reprogramming to induced pluripotent stem cells (iPSCs), further supporting a functional role for the CGI methylation. Both global gene expression profiling and quantitative reverse transcription-PCR (RT-PCR) validation indicated opposing effects of CGI methylation in transcriptional regulation during differentiation, with promoter CGI methylation repressing and 3' CGI methylation activating transcription. By studying diverse human tissues and mouse models, we further confirmed that developmentally programmed 3' CGI methylation confers tissue- and cell-type-specific gene activation in vivo. Importantly, luciferase reporter assays provided evidence that 3' CGI methylation regulates transcriptional activation via a CTCF-dependent enhancer-blocking mechanism. These findings expand the classic view of mammalian CGI methylation as a mechanism for transcriptional silencing and indicate a functional role for 3' CGI methylation in developmental gene regulation.


Subject(s)
CpG Islands , DNA Methylation , Embryonic Stem Cells/cytology , Embryonic Stem Cells/metabolism , Gene Expression Regulation, Developmental , Transcriptional Activation , Animals , CCCTC-Binding Factor , Cell Differentiation , Cell Line , DNA/genetics , Epigenesis, Genetic , Humans , Induced Pluripotent Stem Cells/cytology , Induced Pluripotent Stem Cells/metabolism , Methylation , Mice , Mice, Inbred C57BL , Oligonucleotide Array Sequence Analysis , Repressor Proteins/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...