Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 57
Filter
1.
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
2.
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
3.
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
4.
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
5.
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
6.
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
7.
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
8.
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
9.
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
10.
EMBO J ; 31(9): 2103-16, 2012 May 02.
Article in English | MEDLINE | ID: mdl-22446391

ABSTRACT

The function of metabolic state in stemness is poorly understood. Mouse embryonic stem cells (ESC) and epiblast stem cells (EpiSC) are at distinct pluripotent states representing the inner cell mass (ICM) and epiblast embryos. Human embryonic stem cells (hESC) are similar to EpiSC stage. We now show a dramatic metabolic difference between these two stages. EpiSC/hESC are highly glycolytic, while ESC are bivalent in their energy production, dynamically switching from glycolysis to mitochondrial respiration on demand. Despite having a more developed and expanding mitochondrial content, EpiSC/hESC have low mitochondrial respiratory capacity due to low cytochrome c oxidase (COX) expression. Similarly, in vivo epiblasts suppress COX levels. These data reveal EpiSC/hESC functional similarity to the glycolytic phenotype in cancer (Warburg effect). We further show that hypoxia-inducible factor 1α (HIF1α) is sufficient to drive ESC to a glycolytic Activin/Nodal-dependent EpiSC-like stage. This metabolic switch during early stem-cell development may be deterministic.


Subject(s)
Cell Differentiation/physiology , Embryonic Stem Cells/cytology , Embryonic Stem Cells/physiology , Glycolysis , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Activins/metabolism , Adenosine Triphosphate/metabolism , Animals , Cells, Cultured , DNA, Mitochondrial/analysis , Female , Humans , Membrane Potential, Mitochondrial , Mice , Mice, Inbred C57BL , Prostaglandin-Endoperoxide Synthases/metabolism
11.
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
12.
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
13.
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
14.
PLoS Genet ; 6(10): e1001181, 2010 Oct 28.
Article in English | MEDLINE | ID: mdl-21060811

ABSTRACT

Each unit of the D4Z4 macrosatellite repeat contains a retrotransposed gene encoding the DUX4 double-homeobox transcription factor. Facioscapulohumeral dystrophy (FSHD) is caused by deletion of a subset of the D4Z4 units in the subtelomeric region of chromosome 4. Although it has been reported that the deletion of D4Z4 units induces the pathological expression of DUX4 mRNA, the association of DUX4 mRNA expression with FSHD has not been rigorously investigated, nor has any human tissue been identified that normally expresses DUX4 mRNA or protein. We show that FSHD muscle expresses a different splice form of DUX4 mRNA compared to control muscle. Control muscle produces low amounts of a splice form of DUX4 encoding only the amino-terminal portion of DUX4. FSHD muscle produces low amounts of a DUX4 mRNA that encodes the full-length DUX4 protein. The low abundance of full-length DUX4 mRNA in FSHD muscle cells represents a small subset of nuclei producing a relatively high abundance of DUX4 mRNA and protein. In contrast to control skeletal muscle and most other somatic tissues, full-length DUX4 transcript and protein is expressed at relatively abundant levels in human testis, most likely in the germ-line cells. Induced pluripotent (iPS) cells also express full-length DUX4 and differentiation of control iPS cells to embryoid bodies suppresses expression of full-length DUX4, whereas expression of full-length DUX4 persists in differentiated FSHD iPS cells. Together, these findings indicate that full-length DUX4 is normally expressed at specific developmental stages and is suppressed in most somatic tissues. The contraction of the D4Z4 repeat in FSHD results in a less efficient suppression of the full-length DUX4 mRNA in skeletal muscle cells. Therefore, FSHD represents the first human disease to be associated with the incomplete developmental silencing of a retrogene array normally expressed early in development.


Subject(s)
Gene Expression Profiling , Homeodomain Proteins/genetics , Muscular Dystrophy, Facioscapulohumeral/genetics , RNA Splicing , Adult , Animals , Blotting, Western , Cell Line , Chromosomes, Human, Pair 4/genetics , Female , Fluorescent Antibody Technique , Gene Expression Regulation , HCT116 Cells , Humans , Induced Pluripotent Stem Cells/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, SCID , Molecular Sequence Data , Muscle Cells/metabolism , Muscle, Skeletal/metabolism , Muscle, Skeletal/pathology , Muscular Dystrophy, Facioscapulohumeral/pathology , Repetitive Sequences, Nucleic Acid/genetics , Retroelements/genetics , Reverse Transcriptase Polymerase Chain Reaction
15.
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
16.
Prostate ; 72(13): 1453-63, 2012 Sep 15.
Article in English | MEDLINE | ID: mdl-22314551

ABSTRACT

BACKGROUND: CD90(+) prostate cancer-associated (CP) stromal cells represent a diseased cell type found only in tumor tissue. They differ from their normal counterpart in gene expression and inductive signaling. Genetic reprogramming by induced pluripotent stem (iPS) cell technology can effectively change adult cells into stem-like cells through wholesale alteration of the gene expression program. This technology might be used to 'erase' the abnormal gene expression of diseased cells. The resultant iPS cells would no longer express the disease phenotype, and behave like stem cells. METHODS: CP stromal cells, isolated from tumor tissue of a surgically resected prostate by anti-CD90-mediated sorting and cultured in vitro, were transfected with in vitro packaged lentiviral expression vectors containing stem cell transcription factor genes POU5F1, LIN28, NANOG, and SOX2. RESULTS: Alkaline phosphatase-positive iPS cells were obtained in about 3 weeks post-transfection at a frequency of 10(-4) . Their colony morphology was indistinguishable from that of human embryonic stem (ES) cells. Transcriptome analysis showed a virtually complete match in gene expression between the iPS and ES cells. CONCLUSIONS: Genes of CP stromal cells could be fully inactivated by genetic reprogramming. As a consequence, the disease phenotype was 'cured'.


Subject(s)
Embryonic Stem Cells/cytology , Induced Pluripotent Stem Cells/cytology , Prostatic Neoplasms/pathology , Stromal Cells/pathology , Embryonic Stem Cells/metabolism , Gene Expression Regulation, Neoplastic , Homeodomain Proteins/genetics , Homeodomain Proteins/metabolism , Humans , Induced Pluripotent Stem Cells/metabolism , Male , Nanog Homeobox Protein , Octamer Transcription Factor-3/genetics , Octamer Transcription Factor-3/metabolism , Prostatic Neoplasms/genetics , Prostatic Neoplasms/metabolism , RNA-Binding Proteins/genetics , RNA-Binding Proteins/metabolism , SOXB1 Transcription Factors/genetics , SOXB1 Transcription Factors/metabolism , Stromal Cells/metabolism , Transcriptome
17.
Mol Ther ; 18(6): 1192-9, 2010 Jun.
Article in English | MEDLINE | ID: mdl-20407427

ABSTRACT

Precise genetic manipulation of human pluripotent stem cells will be required to realize their scientific and therapeutic potential. Here, we show that adeno-associated virus (AAV) gene targeting vectors can be used to genetically engineer human embryonic stem cells (ESCs) and induced pluripotent stem cells (iPSCs). Different types of sequence-specific changes, including the creation and correction of mutations, were introduced into the human HPRT1 and HMGA1 genes (HPRT1 mutations being responsible for Lesch-Nyhan syndrome). Gene targeting occurred at high frequencies in both ESCs and iPSCs, with over 1% of all colony-forming units (CFUs) undergoing targeting in some experiments. AAV vectors could also be used to target genes in human fibroblasts that were subsequently used to derive iPSCs. Accurate and efficient targeting took place with minimal or no cytotoxicity, and most of the gene-targeted stem cells produced were euploid and pluripotent.


Subject(s)
Dependovirus/genetics , Gene Targeting , Genetic Engineering , Pluripotent Stem Cells , Humans , Hypoxanthine Phosphoribosyltransferase/genetics
18.
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
19.
Stem Cells ; 26(10): 2496-505, 2008 Oct.
Article in English | MEDLINE | ID: mdl-18583537

ABSTRACT

We used massively parallel pyrosequencing to discover and characterize microRNAs (miRNAs) expressed in human embryonic stem cells (hESC). Sequencing of small RNA cDNA libraries derived from undifferentiated hESC and from isogenic differentiating cultures yielded a total of 425,505 high-quality sequence reads. A custom data analysis pipeline delineated expression profiles for 191 previously annotated miRNAs, 13 novel miRNAs, and 56 candidate miRNAs. Further characterization of a subset of the novel miRNAs in Dicer-knockdown hESC demonstrated Dicer-dependent expression, providing additional validation of our results. A set of 14 miRNAs (9 known and 5 novel) was noted to be expressed in undifferentiated hESC and then strongly downregulated with differentiation. Functional annotation analysis of predicted targets of these miRNAs and comparison with a null model using non-hESC-expressed miRNAs identified statistically enriched functional categories, including chromatin remodeling and lineage-specific differentiation annotations. Finally, integration of our data with genome-wide chromatin immunoprecipitation data on OCT4, SOX2, and NANOG binding sites implicates these transcription factors in the regulation of nine of the novel/candidate miRNAs identified here. Comparison of our results with those of recent deep sequencing studies in mouse and human ESC shows that most of the novel/candidate miRNAs found here were not identified in the other studies. The data indicate that hESC express a larger complement of miRNAs than previously appreciated, and they provide a resource for additional studies of miRNA regulation of hESC physiology. Disclosure of potential conflicts of interest is found at the end of this article.


Subject(s)
Embryonic Stem Cells/metabolism , Gene Expression Profiling , Gene Library , MicroRNAs/genetics , Sequence Analysis, RNA , Base Sequence , Cell Differentiation , Cell Line , Databases, Genetic , Embryonic Stem Cells/cytology , Embryonic Stem Cells/enzymology , Expressed Sequence Tags , Gene Expression Regulation, Developmental , Humans , MicroRNAs/chemistry , Molecular Sequence Data , Multipotent Stem Cells/cytology , Multipotent Stem Cells/metabolism , Nucleic Acid Conformation , Pluripotent Stem Cells/cytology , Pluripotent Stem Cells/metabolism , RNA, Small Interfering/chemistry , RNA, Small Interfering/genetics , Reproducibility of Results , Reverse Transcriptase Polymerase Chain Reaction , Ribonuclease III/metabolism , Transcription Factors/metabolism
20.
Exp Cell Res ; 314(16): 2930-40, 2008 Oct 01.
Article in English | MEDLINE | ID: mdl-18692044

ABSTRACT

Despite a growing body of literature concerning the hematopoietic differentiation of human embryonic stem cells (hESCs), the full hematopoietic potential of the majority of existing hESC lines remains unknown. In this study, the hematopoietic response of five NIH-approved hESC lines (H1, hSF6, BG01, BG02, and BG03) was compared. Our data show that despite expressing similar hESC markers under self-renewing conditions and initiating mesodermal differentiation under spontaneous differentiation conditions, marked differences in subsequent hematopoietic differentiation potential among these lines existed. A high degree of hematopoietic differentiation was attained only by H1 and BG02, whereas this process appeared to be abortive in nature for hSF6, BG01, and BG03. This difference in hematopoietic differentiation predisposition was readily apparent during spontaneous differentiation, and further augmented under hematopoietic-inducing conditions. This predisposition appeared to be intrinsic to the specific hESC line and independent of passage number or gender karyotype. Interestingly, H1 and BG02 displayed remarkable similarities in their kinetics of hematopoietic marker expression, hematopoietic colony formation, erythroid differentiation, and globin expression, suggesting that a similar, predetermined differentiation sequence is followed. The identification of intrinsic and extrinsic factors governing the hematopoietic differentiation potential of hESCs will be of great importance for the putative clinical utility of hESC lines.


Subject(s)
Cell Differentiation/physiology , Embryonic Stem Cells/physiology , Hematopoiesis/physiology , Hematopoietic Stem Cells/physiology , Animals , Biomarkers/metabolism , Cell Line , Cell Lineage , Coculture Techniques , Embryonic Stem Cells/cytology , GATA1 Transcription Factor/genetics , GATA1 Transcription Factor/metabolism , Gene Expression Profiling , Hematopoietic Stem Cells/cytology , Humans , Mice , Phenotype
SELECTION OF CITATIONS
SEARCH DETAIL