Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 4 de 4
Filter
Add more filters











Database
Language
Publication year range
1.
PLoS Comput Biol ; 15(12): e1007543, 2019 12.
Article in English | MEDLINE | ID: mdl-31815944

ABSTRACT

Pluripotent stem cells retain the developmental timing of their species of origin in vitro, an observation that suggests the existence of a cell-intrinsic developmental clock, yet the nature and machinery of the clock remain a mystery. We hypothesize that one possible component may lie in species-specific differences in the kinetics of transcriptional responses to differentiation signals. Using a liquid-handling robot, mouse and human pluripotent stem cells were exposed to identical neural differentiation conditions and sampled for RNA-sequencing at high frequency, every 4 or 10 minutes, for the first 10 hours of differentiation to test for differences in transcriptomic response rates. The majority of initial transcriptional responses occurred within a rapid window in the first minutes of differentiation for both human and mouse stem cells. Despite similarly early onsets of gene expression changes, we observed shortened and condensed gene expression patterns in mouse pluripotent stem cells compared to protracted trends in human pluripotent stem cells. Moreover, the speed at which individual genes were upregulated, as measured by the slopes of gene expression changes over time, was significantly faster in mouse compared to human cells. These results suggest that downstream transcriptomic response kinetics to signaling cues are faster in mouse versus human cells, and may offer a partial account for the vast differences in developmental rates across species.


Subject(s)
Cell Differentiation/genetics , Pluripotent Stem Cells/cytology , Pluripotent Stem Cells/metabolism , RNA-Seq/statistics & numerical data , Animals , Cell Line , Computational Biology , Gene Expression Regulation, Developmental , Human Embryonic Stem Cells/cytology , Human Embryonic Stem Cells/metabolism , Humans , Kinetics , Mice , Mouse Embryonic Stem Cells/cytology , Mouse Embryonic Stem Cells/metabolism , Regenerative Medicine , Species Specificity
2.
Dev Biol ; 423(2): 101-110, 2017 03 15.
Article in English | MEDLINE | ID: mdl-28179190

ABSTRACT

How species-specific developmental timing is controlled is largely unknown. By following human embryonic stem (ES) cell and mouse epiblast stem (EpiS) cell differentiation through detailed RNA-sequencing time courses, here we show that pluripotent stem cells closely retain in vivo species-specific developmental timing in vitro. In identical neural differentiation conditions in vitro, gene expression profiles are accelerated in mouse EpiS cells compared to human ES cells with relative rates of differentiation closely reflecting the rates of progression through the Carnegie stages in utero. Dynamic Time Warping analysis identified 3389 genes that were regulated more quickly in mouse EpiS cells and identified none that were regulated more quickly in human ES cells. Interestingly, we also find that human ES cells differentiated in teratomas maintain the same rate of differentiation observed in vitro in spite of being grown in a mouse host. These results suggest the existence of a cell autonomous, species-specific developmental clock that pluripotent stem cells maintain even out of context of an intact embryo.


Subject(s)
Pluripotent Stem Cells/cytology , Pluripotent Stem Cells/metabolism , Animals , Cell Differentiation/genetics , Cell Line , Embryonic Stem Cells/cytology , Female , Gene Expression Profiling , Gene Expression Regulation , Humans , Mice, SCID , Neurons/cytology , Species Specificity , Teratoma/pathology , Time Factors
3.
Stem Cell Reports ; 5(6): 1109-1118, 2015 Dec 08.
Article in English | MEDLINE | ID: mdl-26584543

ABSTRACT

The derivation of genetically modified induced pluripotent stem (iPS) cells typically involves multiple steps, requiring lengthy cell culture periods, drug selection, and several clonal events. We report the generation of gene-targeted iPS cell lines following a single electroporation of patient-specific fibroblasts using episomal-based reprogramming vectors and the Cas9/CRISPR system. Simultaneous reprogramming and gene targeting was tested and achieved in two independent fibroblast lines with targeting efficiencies of up to 8% of the total iPS cell population. We have successfully targeted the DNMT3B and OCT4 genes with a fluorescent reporter and corrected the disease-causing mutation in both patient fibroblast lines: one derived from an adult with retinitis pigmentosa, the other from an infant with severe combined immunodeficiency. This procedure allows the generation of gene-targeted iPS cell lines with only a single clonal event in as little as 2 weeks and without the need for drug selection, thereby facilitating "seamless" single base-pair changes.


Subject(s)
CRISPR-Cas Systems , Cellular Reprogramming , Fibroblasts/metabolism , Gene Targeting/methods , Induced Pluripotent Stem Cells/metabolism , Adult , Base Sequence , Cell Line , Cells, Cultured , DNA (Cytosine-5-)-Methyltransferases/genetics , Electroporation/methods , Fibroblasts/cytology , Genetic Vectors/genetics , Humans , Induced Pluripotent Stem Cells/cytology , Molecular Sequence Data , Octamer Transcription Factor-3/genetics , DNA Methyltransferase 3B
4.
Stem Cell Reports ; 4(2): 171-80, 2015 Feb 10.
Article in English | MEDLINE | ID: mdl-25601207

ABSTRACT

In this study, we demonstrate a newly derived mouse model that supports engraftment of human hematopoietic stem cells (HSCs) in the absence of irradiation. We cross the NOD.Cg-Prkdc(scid)Il2rg(tm1Wjl)/SzJ (NSG) strain with the C57BL/6J-Kit(W-41J)/J (C57BL/6.Kit(W41)) strain and engraft, without irradiation, the resulting NBSGW strain with human cord blood CD34+ cells. At 12-weeks postengraftment in NBSGW mice, we observe human cell chimerism in marrow (97% ± 0.4%), peripheral blood (61% ± 2%), and spleen (94% ± 2%) at levels observed with irradiation in NSG mice. We also detected a significant number of glycophorin-A-positive expressing cells in the developing NBSGW marrow. Further, the observed levels of human hematopoietic chimerism mimic those reported for both irradiated NSG and NSG-transgenic strains. This mouse model permits HSC engraftment while avoiding the complicating hematopoietic, gastrointestinal, and neurological side effects associated with irradiation and allows investigators without access to radiation to pursue engraftment studies with human HSCs.


Subject(s)
Cell Differentiation , Graft Survival , Hematopoietic Stem Cell Transplantation , Hematopoietic Stem Cells/cytology , Hematopoietic Stem Cells/metabolism , Interleukin Receptor Common gamma Subunit/genetics , Proto-Oncogene Proteins c-kit/genetics , Animals , Cell Lineage , Genotype , Heterografts , Humans , Immunophenotyping , Male , Mice , Mice, Inbred C57BL , Mice, Inbred NOD , Mice, Knockout , Mice, SCID , Phenotype , Time Factors , Transplantation Chimera
SELECTION OF CITATIONS
SEARCH DETAIL