Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 11 de 11
Filter
Add more filters










Publication year range
1.
Nat Commun ; 9(1): 4384, 2018 10 22.
Article in English | MEDLINE | ID: mdl-30349051

ABSTRACT

Self-renewal and pluripotency in human embryonic stem cells (hESCs) depends upon the function of a remarkably small number of master transcription factors (TFs) that include OCT4, SOX2, and NANOG. Endogenous factors that regulate and maintain the expression of master TFs in hESCs remain largely unknown and/or uncharacterized. Here, we use a genome-wide, proteomics approach to identify proteins associated with the OCT4 enhancer. We identify known OCT4 regulators, plus a subset of potential regulators including a zinc finger protein, ZNF207, that plays diverse roles during development. In hESCs, ZNF207 partners with master pluripotency TFs to govern self-renewal and pluripotency while simultaneously controlling commitment of cells towards ectoderm through direct regulation of neuronal TFs, including OTX2. The distinct roles of ZNF207 during differentiation occur via isoform switching. Thus, a distinct isoform of ZNF207 functions in hESCs at the nexus that balances pluripotency and differentiation to ectoderm.


Subject(s)
Embryonic Stem Cells/cytology , Embryonic Stem Cells/metabolism , Human Embryonic Stem Cells/cytology , Human Embryonic Stem Cells/metabolism , Microtubule-Associated Proteins/metabolism , Protein Isoforms/metabolism , Blotting, Western , Cell Differentiation/genetics , Cell Differentiation/physiology , Cell Line , Chromatin Immunoprecipitation , Humans , Immunoprecipitation , Mass Spectrometry , Microtubule-Associated Proteins/genetics , Nanog Homeobox Protein/genetics , Nanog Homeobox Protein/metabolism , Octamer Transcription Factor-3/metabolism , Pluripotent Stem Cells/cytology , Pluripotent Stem Cells/metabolism , Protein Isoforms/genetics , RNA, Small Interfering/genetics , SOXB1 Transcription Factors/genetics , SOXB1 Transcription Factors/metabolism
2.
Heliyon ; 4(6): e00667, 2018 Jun.
Article in English | MEDLINE | ID: mdl-30003161

ABSTRACT

Chromosome instability and aneuploidies occur very frequently in human embryos, impairing proper embryogenesis and leading to cell cycle arrest, loss of cell viability, and developmental failures in 50-80% of cleavage-stage embryos. This high frequency of cellular extinction events represents a significant experimental obstacle challenging analyses of individual cells isolated from human preimplantation embryos. We carried out single cell expression profiling of 241 individual cells recovered from 32 human embryos during the early and late stages of viable human blastocyst (VHB) differentiation. Classification of embryonic cells was performed solely based on expression patterns of human pluripotency-associated transcripts (HPAT), which represent a family of primate-specific transposable element-derived lincRNAs highly expressed in human embryonic stem cells and regulating nuclear reprogramming and pluripotency induction. We then validated our findings by analyzing transcriptomes of 1,708 individual cells recovered from more than 100 human embryos and 259 mouse cells from more than 40 mouse embryos at different stages of preimplantation embryogenesis. HPAT's expression-guided spatiotemporal reconstruction of human embryonic development inferred from single-cell expression analysis of VHB differentiation enabled identification of telomerase-positive embryonic cells co-expressing key pluripotency regulatory genes and genetic markers of three major lineages. Follow-up validation analyses confirmed the emergence in human embryos prior to lineage segregation of telomerase-positive cells co-expressing genetic markers of multiple lineages. Observations reported in this contribution support the hypothesis of a developmental pathway of creation embryonic lineages and extraembryonic tissues from telomerase-positive pre-lineage cells manifesting multi-lineage precursor phenotype.

3.
Dev Cell ; 38(1): 100-15, 2016 07 11.
Article in English | MEDLINE | ID: mdl-27404362

ABSTRACT

Human preimplantation embryo development involves complex cellular and molecular events that lead to the establishment of three cell lineages in the blastocyst: trophectoderm, primitive endoderm, and epiblast. Owing to limited resources of biological specimens, our understanding of how the earliest lineage commitments are regulated remains narrow. Here, we examined gene expression in 241 individual cells from early and late human blastocysts to delineate dynamic gene-expression changes. We distinguished all three lineages and further developed a 3D model of the inner cell mass and trophectoderm in which individual cells were mapped into distinct expression domains. We identified in silico precursors of the epiblast and primitive endoderm lineages and revealed a role for MCRS1, TET1, and THAP11 in epiblast formation and their ability to induce naive pluripotency in vitro. Our results highlight the potential of single-cell gene-expression analysis in human preimplantation development to instruct human stem cell biology.


Subject(s)
Blastocyst/cytology , Cell Lineage/genetics , Endoderm/cytology , Gene Expression Profiling , Germ Layers/cytology , Pluripotent Stem Cells/cytology , Single-Cell Analysis/methods , Biomarkers/metabolism , Blastocyst/metabolism , Cell Differentiation , Embryonic Development , Endoderm/metabolism , Gene Expression Regulation, Developmental , Genes, Developmental , Germ Layers/metabolism , Humans , Mixed Function Oxygenases/genetics , Nuclear Proteins/genetics , Pluripotent Stem Cells/metabolism , Proto-Oncogene Proteins/genetics , RNA-Binding Proteins/genetics , Repressor Proteins/genetics
4.
Cell Rep ; 14(10): 2301-12, 2016 Mar 15.
Article in English | MEDLINE | ID: mdl-26947063

ABSTRACT

The human naive pluripotent stem cell (PSC) state, corresponding to a pre-implantation stage of development, has been difficult to capture and sustain in vitro. We report that the Hippo pathway effector YAP is nuclearly localized in the inner cell mass of human blastocysts. Overexpression of YAP in human embryonic stem cells (ESCs) and induced PSCs (iPSCs) promotes the generation of naive PSCs. Lysophosphatidic acid (LPA) can partially substitute for YAP to generate transgene-free human naive PSCs. YAP- or LPA-induced naive PSCs have a rapid clonal growth rate, a normal karyotype, the ability to form teratomas, transcriptional similarities to human pre-implantation embryos, reduced heterochromatin levels, and other hallmarks of the naive state. YAP/LPA act in part by suppressing differentiation-inducing effects of GSK3 inhibition. CRISPR/Cas9-generated YAP(-/-) cells have an impaired ability to form colonies in naive but not primed conditions. These results uncover an unexpected role for YAP in the human naive state, with implications for early human embryology.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , Phosphoproteins/metabolism , Pluripotent Stem Cells/metabolism , Adaptor Proteins, Signal Transducing/antagonists & inhibitors , Adaptor Proteins, Signal Transducing/genetics , Benzamides/pharmacology , Blastocyst/cytology , Blastocyst/metabolism , Blastocyst/pathology , CRISPR-Cas Systems/genetics , Cell Differentiation/drug effects , Cells, Cultured , Diphenylamine/analogs & derivatives , Diphenylamine/pharmacology , Embryonic Stem Cells/cytology , Embryonic Stem Cells/drug effects , Embryonic Stem Cells/metabolism , Female , Gene Expression Regulation/drug effects , Glycogen Synthase Kinase 3/antagonists & inhibitors , Glycogen Synthase Kinase 3/metabolism , Heterochromatin/metabolism , Humans , Induced Pluripotent Stem Cells/cytology , Induced Pluripotent Stem Cells/metabolism , Karyotype , Lysophospholipids/pharmacology , Male , Phosphoproteins/antagonists & inhibitors , Phosphoproteins/genetics , Pluripotent Stem Cells/cytology , Pyridines/pharmacology , Pyrimidines/pharmacology , RNA Interference , Teratoma/metabolism , Teratoma/pathology , Transcription Factors/genetics , Transcription Factors/metabolism , Wnt Signaling Pathway/drug effects , YAP-Signaling Proteins
5.
Nat Genet ; 48(1): 44-52, 2016 Jan.
Article in English | MEDLINE | ID: mdl-26595768

ABSTRACT

Long intergenic noncoding RNAs (lincRNAs) are derived from thousands of loci in mammalian genomes and are frequently enriched in transposable elements (TEs). Although families of TE-derived lincRNAs have recently been implicated in the regulation of pluripotency, little is known of the specific functions of individual family members. Here we characterize three new individual TE-derived human lincRNAs, human pluripotency-associated transcripts 2, 3 and 5 (HPAT2, HPAT3 and HPAT5). Loss-of-function experiments indicate that HPAT2, HPAT3 and HPAT5 function in preimplantation embryo development to modulate the acquisition of pluripotency and the formation of the inner cell mass. CRISPR-mediated disruption of the genes for these lincRNAs in pluripotent stem cells, followed by whole-transcriptome analysis, identifies HPAT5 as a key component of the pluripotency network. Protein binding and reporter-based assays further demonstrate that HPAT5 interacts with the let-7 microRNA family. Our results indicate that unique individual members of large primate-specific lincRNA families modulate gene expression during development and differentiation to reinforce cell fate.


Subject(s)
Blastocyst/physiology , Gene Expression Regulation, Developmental , Pluripotent Stem Cells/physiology , Primates/genetics , RNA, Long Noncoding/genetics , Animals , Blastocyst/cytology , Cell Differentiation/genetics , Embryonic Development/genetics , Gene Knockdown Techniques , Humans , MicroRNAs/genetics , Pluripotent Stem Cells/cytology , RNA, Long Noncoding/metabolism , Single-Cell Analysis
6.
Sci Rep ; 5: 15041, 2015 Oct 12.
Article in English | MEDLINE | ID: mdl-26456624

ABSTRACT

Deletions of the AZFa region (AZoospermia Factor-a) region of the human Y chromosome cause irreversible spermatogenic failure that presents clinically in men as Sertoli-cell only (SCO) pathology of the testis. Deletions of the AZFa region typically encompass two genes: DDX3Y and USP9Y. However, human genetic evidence indicates that SCO is most tightly linked to deletion of DDX3Y and that deletions/mutations of USP9Y can be transmitted from one generation to the next. Here, we generated stable iPSC lines with AZFa deletions, tested complementation via introduction of DDX3Y, and assessed ability to form germ cells in vivo in a xenotransplantation model. We observed a quantifiable improvement in formation of germ cell like cells (GCLCs) from complemented donor iPSCs. Moreover, expression of UTF1, a prospermatogonial protein, was restored in cells complemented by introduction of DDX3Y on the AZFa background. Whole-genome RNA sequencing of purified GCLCs revealed an enrichment of genes involved in translational suppression and transcriptional control in DDX3Y-rescued GCLCs over mutant GCLCs, which maintained a molecular phenotype more similar to undifferentiated iPSCs. This study demonstrates the ability to probe fundamental genetics of human germ cell formation by complementation and indicates that DDX3Y functions in the earliest stages of human germ cell development.


Subject(s)
Chromosomes, Human, Y/metabolism , DEAD-box RNA Helicases/genetics , Induced Pluripotent Stem Cells/cytology , Spermatogenesis/genetics , Spermatozoa/metabolism , Transcription, Genetic , Animals , Busulfan/pharmacology , Cell Differentiation , Chromosomes, Human, Y/chemistry , DEAD-box RNA Helicases/metabolism , Fibroblasts/cytology , Fibroblasts/metabolism , Gene Expression Regulation, Developmental , Genes, Reporter , Genetic Complementation Test , Genetic Vectors , Humans , Induced Pluripotent Stem Cells/metabolism , Luminescent Proteins/genetics , Luminescent Proteins/metabolism , Male , Mice, Nude , Minor Histocompatibility Antigens , Nuclear Proteins/genetics , Nuclear Proteins/metabolism , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Skin/cytology , Skin/metabolism , Spermatozoa/cytology , Testis/cytology , Testis/drug effects , Testis/metabolism , Trans-Activators/genetics , Trans-Activators/metabolism , Transplantation, Heterologous , Red Fluorescent Protein
7.
Cell Rep ; 7(4): 1284-97, 2014 May 22.
Article in English | MEDLINE | ID: mdl-24794432

ABSTRACT

Historically, spontaneous deletions and insertions have provided means to probe germline developmental genetics in Drosophila, mouse and other species. Here, induced pluripotent stem cell (iPSC) lines were derived from infertile men with deletions that encompass three Y chromosome azoospermia factor (AZF) regions and are associated with production of few or no sperm but normal somatic development. AZF-deleted iPSC lines were compromised in germ cell development in vitro. Undifferentiated iPSCs transplanted directly into murine seminiferous tubules differentiated extensively to germ-cell-like cells (GCLCs) that localized near the basement membrane, demonstrated morphology indistinguishable from fetal germ cells, and expressed germ-cell-specific proteins diagnostic of primordial germ cells. Alternatively, all iPSCs that exited tubules formed primitive tumors. iPSCs with AZF deletions produced significantly fewer GCLCs in vivo with distinct defects in gene expression. Findings indicate that xenotransplantation of human iPSCs directs germ cell differentiation in a manner dependent on donor genetic status.


Subject(s)
Azoospermia/pathology , Fertility/physiology , Induced Pluripotent Stem Cells/cytology , Seminiferous Tubules/cytology , Animals , Cell Differentiation/physiology , Cell Line , Humans , Induced Pluripotent Stem Cells/metabolism , Male , Mice , Mice, Nude , Skin/cytology , Transplantation, Heterologous/methods
8.
PLoS One ; 9(4): e94231, 2014.
Article in English | MEDLINE | ID: mdl-24718618

ABSTRACT

Data suggest that clinical applications of human induced pluripotent stem cells (hiPSCs) will be realized. Nonetheless, clinical applications will require hiPSCs that are free of exogenous DNA and that can be manufactured through Good Manufacturing Practice (GMP). Optimally, derivation of hiPSCs should be rapid and efficient in order to minimize manipulations, reduce potential for accumulation of mutations and minimize financial costs. Previous studies reported the use of modified synthetic mRNAs to reprogram fibroblasts to a pluripotent state. Here, we provide an optimized, fully chemically defined and feeder-free protocol for the derivation of hiPSCs using synthetic mRNAs. The protocol results in derivation of fully reprogrammed hiPSC lines from adult dermal fibroblasts in less than two weeks. The hiPSC lines were successfully tested for their identity, purity, stability and safety at a GMP facility and cryopreserved. To our knowledge, as a proof of principle, these are the first integration-free iPSCs lines that were reproducibly generated through synthetic mRNA reprogramming that could be putatively used for clinical purposes.


Subject(s)
Cell Culture Techniques/methods , Cellular Reprogramming , Induced Pluripotent Stem Cells/cytology , RNA, Messenger/pharmacology , Adult , Animals , Cell Culture Techniques/standards , Cell Differentiation , Cell Line , Cellular Reprogramming/drug effects , Embryoid Bodies , Female , Fibroblasts/cytology , Fibroblasts/drug effects , Gene Expression Profiling , Germ Layers/cytology , Green Fluorescent Proteins/genetics , Humans , Induced Pluripotent Stem Cells/drug effects , Induced Pluripotent Stem Cells/transplantation , Infant, Newborn , Kruppel-Like Factor 4 , Kruppel-Like Transcription Factors/genetics , Male , Mice , Mice, SCID , Middle Aged , Octamer Transcription Factor-3/genetics , Primary Cell Culture , Proto-Oncogene Proteins c-myc/genetics , RNA, Messenger/chemical synthesis , RNA, Messenger/isolation & purification , RNA-Binding Proteins/genetics , SOXB1 Transcription Factors/genetics , Skin/cytology , Teratoma/etiology , Teratoma/pathology , Transfection
9.
Hum Mol Genet ; 23(12): 3071-84, 2014 Jun 15.
Article in English | MEDLINE | ID: mdl-24449759

ABSTRACT

Studies of human germ cell development are limited in large part by inaccessibility of germ cells during development. Moreover, although several studies have reported differentiation of mouse and human germ cells from pluripotent stem cells (PSCs) in vitro, differentiation of human germ cells from PSCs in vivo has not been reported. Here, we tested whether mRNA reprogramming in combination with xeno-transplantation may provide a viable system to probe the genetics of human germ cell development via use of induced pluripotent stem cells (iPSCs). For this purpose, we derived integration-free iPSCs via mRNA-based reprogramming with OCT3/4, SOX2, KLF4 and cMYC alone (OSKM) or in combination with the germ cell-specific mRNA, VASA (OSKMV). All iPSC lines met classic criteria of pluripotency. Moreover, global gene expression profiling did not distinguish large differences between undifferentiated OSKM and OSKMV iPSCs; however, some differences were observed in expression of pluripotency factors and germ cell-specific genes, and in epigenetic profiles and in vitro differentiation studies. In contrast, transplantation of undifferentiated iPSCs directly into the seminiferous tubules of germ cell-depleted immunodeficient mice revealed divergent fates of iPSCs produced with different factors. Transplantation resulted in morphologically and immunohistochemically recognizable germ cells in vivo, particularly in the case of OSKMV cells. Significantly, OSKMV cells also did not form tumors while OSKM cells that remained outside the seminiferous tubule proliferated extensively and formed tumors. Results indicate that mRNA reprogramming in combination with transplantation may contribute to tools for genetic analysis of human germ cell development.


Subject(s)
Cellular Reprogramming , Induced Pluripotent Stem Cells/metabolism , Induced Pluripotent Stem Cells/transplantation , Seminiferous Tubules/metabolism , Spermatozoa/physiology , Animals , Cell Differentiation , Cell Line , Epigenesis, Genetic , Gene Expression Profiling , Gene Expression Regulation , Humans , Kruppel-Like Factor 4 , Male , Mice , Mice, Nude , Spermatozoa/cytology , Transplantation, Heterologous/methods
10.
Cell Stem Cell ; 14(2): 237-52, 2014 Feb 06.
Article in English | MEDLINE | ID: mdl-24412311

ABSTRACT

Human pluripotent stem cell (hPSC) differentiation typically yields heterogeneous populations. Knowledge of signals controlling embryonic lineage bifurcations could efficiently yield desired cell types through exclusion of alternate fates. Therefore, we revisited signals driving induction and anterior-posterior patterning of definitive endoderm to generate a coherent roadmap for endoderm differentiation. With striking temporal dynamics, BMP and Wnt initially specified anterior primitive streak (progenitor to endoderm), yet, 24 hr later, suppressed endoderm and induced mesoderm. At lineage bifurcations, cross-repressive signals separated mutually exclusive fates; TGF-ß and BMP/MAPK respectively induced pancreas versus liver from endoderm by suppressing the alternate lineage. We systematically blockaded alternate fates throughout multiple consecutive bifurcations, thereby efficiently differentiating multiple hPSC lines exclusively into endoderm and its derivatives. Comprehensive transcriptional and chromatin mapping of highly pure endodermal populations revealed that endodermal enhancers existed in a surprising diversity of "pre-enhancer" states before activation, reflecting the establishment of a permissive chromatin landscape as a prelude to differentiation.


Subject(s)
Cell Lineage , Endoderm/embryology , Pluripotent Stem Cells/cytology , Signal Transduction , Animals , Base Sequence , Body Patterning/drug effects , Bone Morphogenetic Proteins/metabolism , Cell Lineage/drug effects , Chromatin/metabolism , Culture Media, Serum-Free/pharmacology , Digestive System/embryology , Embryonic Stem Cells/cytology , Embryonic Stem Cells/metabolism , Endoderm/cytology , Endoderm/drug effects , Enhancer Elements, Genetic/genetics , Epigenesis, Genetic/drug effects , Fibroblast Growth Factors/metabolism , Humans , Liver/embryology , MAP Kinase Signaling System/drug effects , Mice , Molecular Sequence Data , Pancreas/embryology , Pluripotent Stem Cells/drug effects , Primitive Streak/cytology , Primitive Streak/embryology , Protein Binding/drug effects , Signal Transduction/drug effects , Transcription, Genetic/drug effects , Transforming Growth Factor beta/metabolism , Wnt Proteins/metabolism
11.
Stem Cell Res Ther ; 4(4): 87, 2013 Jul 26.
Article in English | MEDLINE | ID: mdl-23890092

ABSTRACT

INTRODUCTION: The reprogramming of a patient's somatic cells back into induced pluripotent stem cells (iPSCs) holds significant promise for future autologous cellular therapeutics. The continued presence of potentially oncogenic transgenic elements following reprogramming, however, represents a safety concern that should be addressed prior to clinical applications. The polycistronic stem cell cassette (STEMCCA), an excisable lentiviral reprogramming vector, provides, in our hands, the most consistent reprogramming approach that addresses this safety concern. Nevertheless, most viral integrations occur in genes, and exactly how the integration, epigenetic reprogramming, and excision of the STEMCCA reprogramming vector influences those genes and whether these cells still have clinical potential are not yet known. METHODS: In this study, we used both microarray and sensitive real-time PCR to investigate gene expression changes following both intron-based reprogramming and excision of the STEMCCA cassette during the generation of human iPSCs from adult human dermal fibroblasts. Integration site analysis was conducted using nonrestrictive linear amplification PCR. Transgene-free iPSCs were fully characterized via immunocytochemistry, karyotyping and teratoma formation, and current protocols were implemented for guided differentiation. We also utilized current good manufacturing practice guidelines and manufacturing facilities for conversion of our iPSCs into putative clinical grade conditions. RESULTS: We found that a STEMCCA-derived iPSC line that contains a single integration, found to be located in an intronic location in an actively transcribed gene, PRPF39, displays significantly increased expression when compared with post-excised stem cells. STEMCCA excision via Cre recombinase returned basal expression levels of PRPF39. These cells were also shown to have proper splicing patterns and PRPF39 gene sequences. We also fully characterized the post-excision iPSCs, differentiated them into multiple clinically relevant cell types (including oligodendrocytes, hepatocytes, and cardiomyocytes), and converted them to putative clinical-grade conditions using the same approach previously approved by the US Food and Drug Administration for the conversion of human embryonic stem cells from research-grade to clinical-grade status. CONCLUSION: For the first time, these studies provide a proof-of-principle for the generation of fully characterized transgene-free human iPSCs and, in light of the limited availability of current good manufacturing practice cellular manufacturing facilities, highlight an attractive potential mechanism for converting research-grade cell lines into putatively clinical-grade biologics for personalized cellular therapeutics.


Subject(s)
Induced Pluripotent Stem Cells/metabolism , Animals , Cell Differentiation , Cellular Reprogramming , Gene Expression , Genomics , Humans , Mice , Transgenes
SELECTION OF CITATIONS
SEARCH DETAIL
...