Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 20
Filter
1.
Proc Natl Acad Sci U S A ; 119(49): e2207824119, 2022 12 06.
Article in English | MEDLINE | ID: mdl-36454756

ABSTRACT

Revealing the molecular events associated with reprogramming different somatic cell types to pluripotency is critical for understanding the characteristics of induced pluripotent stem cell (iPSC) therapeutic derivatives. Inducible reprogramming factor transgenic cells or animals-designated as secondary (2Ā°) reprogramming systems-not only provide excellent experimental tools for such studies but also offer a strategy to study the variances in cellular reprogramming outcomes due to different in vitro and in vivo environments. To make such studies less cumbersome, it is desirable to have a variety of efficient reprogrammable mouse systems to induce successful mass reprogramming in somatic cell types. Here, we report the development of two transgenic mouse lines from which 2Ā°Ā cells reprogram with unprecedented efficiency. These systems were derived by exposing primary reprogramming cells containing doxycycline-inducible Yamanaka factor expression to a transient interruption in transgene expression, resulting in selection for a subset of clones with robust transgene response. These systems also include reporter genes enabling easy readout of endogenous Oct4 activation (GFP), indicative of pluripotency, and reprogramming transgene expression (mCherry). Notably, somatic cells derived from various fetal and adult tissues from these 2Ā°Ā mouse lines gave rise to highly efficient and rapid reprogramming, with transgene-independent iPSC colonies emerging as early as 1 wk after induction. These mouse lines serve as a powerful tool to explore sources of variability in reprogramming and the mechanistic underpinnings of efficient reprogramming systems.


Subject(s)
Cellular Reprogramming , Doxycycline , Animals , Mice , Mice, Transgenic , Cellular Reprogramming/genetics , Transgenes , Clone Cells , Doxycycline/pharmacology
2.
Nature ; 516(7530): 192-7, 2014 Dec 11.
Article in English | MEDLINE | ID: mdl-25503232

ABSTRACT

Pluripotency is defined by the ability of a cell to differentiate to the derivatives of all the three embryonic germ layers: ectoderm, mesoderm and endoderm. Pluripotent cells can be captured via the archetypal derivation of embryonic stem cells or via somatic cell reprogramming. Somatic cells are induced to acquire a pluripotent stem cell (iPSC) state through the forced expression of key transcription factors, and in the mouse these cells can fulfil the strictest of all developmental assays for pluripotent cells by generating completely iPSC-derived embryos and mice. However, it is not known whether there are additional classes of pluripotent cells, or what the spectrum of reprogrammed phenotypes encompasses. Here we explore alternative outcomes of somatic reprogramming by fully characterizing reprogrammed cells independent of preconceived definitions of iPSC states. We demonstrate that by maintaining elevated reprogramming factor expression levels, mouse embryonic fibroblasts go through unique epigenetic modifications to arrive at a stable, Nanog-positive, alternative pluripotent state. In doing so, we prove that the pluripotent spectrum can encompass multiple, unique cell states.


Subject(s)
Cellular Reprogramming/genetics , Cellular Reprogramming/physiology , Epigenesis, Genetic , Induced Pluripotent Stem Cells/cytology , Induced Pluripotent Stem Cells/metabolism , Animals , Embryonic Stem Cells/cytology , Embryonic Stem Cells/metabolism , Female , Fibroblasts/classification , Fibroblasts/cytology , Fibroblasts/metabolism , Histone Deacetylases/metabolism , Induced Pluripotent Stem Cells/classification , Mice , Mice, Nude , Transcription Factors/genetics , Transcription Factors/metabolism , Transgenes/genetics
3.
Nature ; 516(7530): 198-206, 2014 Dec 11.
Article in English | MEDLINE | ID: mdl-25503233

ABSTRACT

Somatic cell reprogramming to a pluripotent state continues to challenge many of our assumptions about cellular specification, and despite major efforts, we lack a complete molecular characterization of the reprograming process. To address this gap in knowledge, we generated extensive transcriptomic, epigenomic and proteomic data sets describing the reprogramming routes leading from mouse embryonic fibroblasts to induced pluripotency. Through integrative analysis, we reveal that cells transition through distinct gene expression and epigenetic signatures and bifurcate towards reprogramming transgene-dependent and -independent stable pluripotent states. Early transcriptional events, driven by high levels of reprogramming transcription factor expression, are associated with widespread loss of histone H3 lysine 27 (H3K27me3) trimethylation, representing a general opening of the chromatin state. Maintenance of high transgene levels leads to re-acquisition of H3K27me3 and a stable pluripotent state that is alternative to the embryonic stem cell (ESC)-like fate. Lowering transgene levels at an intermediate phase, however, guides the process to the acquisition of ESC-like chromatin and DNA methylation signature. Our data provide a comprehensive molecular description of the reprogramming routes and is accessible through the Project Grandiose portal at http://www.stemformatics.org.


Subject(s)
Cellular Reprogramming/genetics , Genome/genetics , Induced Pluripotent Stem Cells/cytology , Induced Pluripotent Stem Cells/metabolism , Animals , Chromatin/chemistry , Chromatin/genetics , Chromatin/metabolism , Chromatin Assembly and Disassembly , DNA Methylation , Embryonic Stem Cells/cytology , Embryonic Stem Cells/metabolism , Epistasis, Genetic/genetics , Fibroblasts/cytology , Fibroblasts/metabolism , Histones/chemistry , Histones/metabolism , Internet , Mice , Proteome/genetics , Proteomics , RNA, Long Noncoding/genetics , Transcription Factors/genetics , Transcription Factors/metabolism , Transcription, Genetic/genetics , Transcriptome/genetics , Transgenes/genetics
6.
Proteomics ; 15(18): 3219-31, 2015 Sep.
Article in English | MEDLINE | ID: mdl-26080932

ABSTRACT

Cellular reprogramming remodels the gene expression program by re-setting the epigenome of somatic cells into an embryonic-like pluripotent state. Post-translational modifications of histones play an important role in this process. Previously, we found by ChIP-seq widespread changes of specific histone H3 marks in two divergent reprogramming routes leading to alternative pluripotent sates . Here, using an unbiased middle-down proteomics approach we have identified 72 unique isoforms of histone H4 and quantified 56 of them in the same set of samples. We found substantial differences between somatic and late-phase reprogramming cells. Also, ESCs and iPSCs displayed higher levels of H4 acetylation and tri-methylation concomitantly with lower levels of mono- and di-methylation when compared to cells undergoing reprogramming. Our data shows that the epigenetic remodeling induced by the reprogramming process goes beyond histone H3 and reveals the importance of H4 modifications as well. The presented data is a valuable resource to study the epigenetic mechanisms involved in the acquisition of induced pluripotency. All MS data have been deposited in the ProteomeXchange with identifier PXD002062 (http://proteomecentral.proteomexchange.org/dataset/PXD002062).


Subject(s)
Cellular Reprogramming/physiology , Epigenesis, Genetic/physiology , Histones/chemistry , Histones/metabolism , Pluripotent Stem Cells/metabolism , Proteomics/methods , Animals , Cells, Cultured , Cluster Analysis , Mice , Protein Isoforms/chemistry , Protein Isoforms/metabolism , Proteome/analysis , Proteome/chemistry , Proteome/metabolism
7.
Proc Natl Acad Sci U S A ; 109(18): 6993-8, 2012 May 01.
Article in English | MEDLINE | ID: mdl-22509029

ABSTRACT

Vascular smooth muscle cells (VSMC) have been suggested to arise from various developmental sources during embryogenesis, depending on the vascular bed. However, evidence also points to a common subpopulation of vascular progenitor cells predisposed to VSMC fate in the embryo. In the present study, we use binary transgenic reporter mice to identify a Tie1(+)CD31(dim)vascular endothelial (VE)-cadherin(-)CD45(-) precursor that gives rise to VSMC in vivo in all vascular beds examined. This precursor does not represent a mature endothelial cell, because a VE-cadherin promoter-driven reporter shows no expression in VSMC during murine development. Blockade of Notch signaling in the Tie1(+) precursor cell, but not the VE-cadherin(+) endothelial cell, decreases VSMC investment of developing arteries, leading to localized hemorrhage in the embryo at the time of vascular maturation. However, Notch signaling is not required in the Tie1(+) precursor after establishment of a stable artery. Thus, Notch activity is required in the differentiation of a Tie1(+) local precursor to VSMC in a spatiotemporal fashion across all vascular beds.


Subject(s)
Cell Differentiation/physiology , Myoblasts, Smooth Muscle/cytology , Myoblasts, Smooth Muscle/metabolism , Myocytes, Smooth Muscle/cytology , Myocytes, Smooth Muscle/metabolism , Neovascularization, Physiologic , Receptors, Notch/metabolism , Animals , Antigens, CD/genetics , Arteries/embryology , Arteries/growth & development , Arteries/metabolism , Base Sequence , Cadherins/deficiency , Cadherins/genetics , Cell Differentiation/genetics , DNA Primers/genetics , Female , Leukocyte Common Antigens/deficiency , Leukocyte Common Antigens/genetics , Mice , Mice, Transgenic , Neovascularization, Physiologic/genetics , Platelet Endothelial Cell Adhesion Molecule-1/metabolism , Pregnancy , Receptor, TIE-1/metabolism , Receptors, Notch/antagonists & inhibitors , Signal Transduction
8.
Stem Cells ; 30(1): 10-4, 2012 Jan.
Article in English | MEDLINE | ID: mdl-22102565

ABSTRACT

Extraordinary advances in pluripotent stem cell research have initiated an era of hope for regenerative strategies to treat human disease. Besides embryonic stem cells, the discovery of induced pluripotent stem cells widened the possibility of patient-specific cell therapy, drug discovery, and disease modeling. Although similar, it has become clear that these two pluripotent cell types display significant differences. In this review, we explore current knowledge of the molecular and functional similarities and differences between these two cell types to emphasize the necessity for thorough characterization of their properties as well as their differentiation capabilities in the pluripotent state. Such comparative studies will be crucial for determining the more suitable cell type for future stem cell-based therapies for human degenerative diseases.


Subject(s)
Cell Differentiation/physiology , Embryonic Stem Cells/physiology , Induced Pluripotent Stem Cells/physiology , Pluripotent Stem Cells/physiology , Drug Discovery , Embryonic Stem Cells/metabolism , Humans , Induced Pluripotent Stem Cells/metabolism , Stem Cell Transplantation
9.
Dev Biol ; 335(1): 66-77, 2009 Nov 01.
Article in English | MEDLINE | ID: mdl-19703439

ABSTRACT

Vascular patterning depends on precisely coordinated timing of endothelial cell differentiation and onset of cardiac function. Endoglin is a transmembrane receptor for members of the TGF-beta superfamily that is expressed on endothelial cells from early embryonic gestation to adult life. Heterozygous loss of function mutations in human ENDOGLIN cause Hereditary Hemorrhagic Telangiectasia Type 1, a vascular disorder characterized by arteriovenous malformations that lead to hemorrhage and stroke. Endoglin null mice die in embryogenesis with numerous lesions in the cardiovascular tree including incomplete yolk sac vessel branching and remodeling, vessel dilation, hemorrhage and abnormal cardiac morphogenesis. Since defects in multiple cardiovascular tissues confound interpretations of these observations, we performed in vivo chimeric rescue analysis using Endoglin null embryonic stem cells. We demonstrate that Endoglin is required cell autonomously for endocardial to mesenchymal transition during formation of the endocardial cushions. Endoglin null cells contribute widely to endothelium in chimeric embryos rescued from cardiac development defects, indicating that Endoglin is dispensable for angiogenesis and vascular remodeling in the midgestation embryo, but is required for early patterning of the heart.


Subject(s)
Embryo, Mammalian , Endocardium , Heart , Intracellular Signaling Peptides and Proteins/metabolism , Neovascularization, Physiologic , Telangiectasia, Hereditary Hemorrhagic/genetics , Animals , Body Patterning/physiology , Cell Differentiation/physiology , Chimera/anatomy & histology , Chimera/physiology , Embryo, Mammalian/anatomy & histology , Embryo, Mammalian/physiology , Endocardium/cytology , Endocardium/embryology , Endoglin , Endothelial Cells/cytology , Endothelial Cells/physiology , Gene Expression Regulation, Developmental , Heart/anatomy & histology , Heart/embryology , Humans , Intracellular Signaling Peptides and Proteins/genetics , Mice , Mice, Knockout , Neovascularization, Physiologic/genetics , Neovascularization, Physiologic/physiology
10.
Science ; 364(6438)2019 04 26.
Article in English | MEDLINE | ID: mdl-30898844

ABSTRACT

The ability to generate induced pluripotent stem cells from differentiated cell types has enabled researchers to engineer cell states. Although studies have identified molecular networks that reprogram cells to pluripotency, the cellular dynamics of these processes remain poorly understood. Here, by combining cellular barcoding, mathematical modeling, and lineage tracing approaches, we demonstrate that reprogramming dynamics in heterogeneous populations are driven by dominant "elite" clones. Clones arise a priori from a population of poised mouse embryonic fibroblasts derived from Wnt1-expressing cells that may represent a neural crest-derived population. This work highlights the importance of cellular dynamics in fate programming outcomes and uncovers cell competition as a mechanism by which cells with eliteness emerge to occupy and dominate the reprogramming niche.


Subject(s)
Cellular Reprogramming/physiology , Clonal Evolution , Induced Pluripotent Stem Cells/cytology , Animals , Cellular Reprogramming/genetics , Cellular Reprogramming Techniques , Clone Cells/cytology , DNA/genetics , Fibroblasts/cytology , Mice , Models, Theoretical
11.
Mol Cell Biol ; 25(11): 4693-702, 2005 Jun.
Article in English | MEDLINE | ID: mdl-15899871

ABSTRACT

The development of the cardiovascular system and the development of the early hematopoietic systems are closely related, and both require signaling through the Tie2 receptor tyrosine kinase. Although endothelial cells and hematopoietic cells as well as their precursors share common gene expression patterns during development, it remains completely unknown how Tie2 signaling coordinately regulates cardiovascular development and early hematopoiesis in vivo. We show here that mice with a targeted mutation in tyrosine residue 1100 in the carboxyl-terminal tail of Tie2 display defective cardiac development and impaired hematopoietic and endothelial cell development in the paraaortic splanchnopleural mesoderm similar to that seen in Tie2-null mutant mice. Surprisingly, however, unlike Tie2-null mutant mice, mice deficient in signaling through this tyrosine residue show a normal association of perivascular cells with nascent blood vessels. These studies are the first to demonstrate the physiological importance of a single tyrosine residue in Tie2, and they suggest that multiple tyrosine residues in the receptor may coordinate cardiovascular development and early hematopoietic development.


Subject(s)
Endothelium, Vascular/embryology , Heart/embryology , Hematopoiesis , Receptor, TIE-2/chemistry , Receptor, TIE-2/physiology , Tyrosine/genetics , Animals , Embryo, Mammalian/cytology , Endothelium, Vascular/abnormalities , Heart Defects, Congenital/embryology , Homozygote , Mice , Mice, Mutant Strains , Point Mutation , Receptor, TIE-2/genetics
12.
J Vis Exp ; (97)2015 Mar 04.
Article in English | MEDLINE | ID: mdl-25867243

ABSTRACT

Ultrasound contrast-enhanced imaging can convey essential quantitative information regarding tissue vascularity and perfusion and, in targeted applications, facilitate the detection and measure of vascular biomarkers at the molecular level. Within the mouse embryo, this noninvasive technique may be used to uncover basic mechanisms underlying vascular development in the early mouse circulatory system and in genetic models of cardiovascular disease. The mouse embryo also presents as an excellent model for studying the adhesion of microbubbles to angiogenic targets (including vascular endothelial growth factor receptor 2 (VEGFR2) or αvƟ3) and for assessing the quantitative nature of molecular ultrasound. We therefore developed a method to introduce ultrasound contrast agents into the vasculature of living, isolated embryos. This allows freedom in terms of injection control and positioning, reproducibility of the imaging plane without obstruction and motion, and simplified image analysis and quantification. Late gestational stage (embryonic day (E)16.6 and E17.5) murine embryos were isolated from the uterus, gently exteriorized from the yolk sac and microbubble contrast agents were injected into veins accessible on the chorionic surface of the placental disc. Nonlinear contrast ultrasound imaging was then employed to collect a number of basic perfusion parameters (peak enhancement, wash-in rate and time to peak) and quantify targeted microbubble binding in an endoglin mouse model. We show the successful circulation of microbubbles within living embryos and the utility of this approach in characterizing embryonic vasculature and microbubble behavior.


Subject(s)
Contrast Media/administration & dosage , Embryo, Mammalian/diagnostic imaging , Mice/embryology , Microbubbles , Ultrasonics/methods , Animals , Contrast Media/chemistry , Contrast Media/pharmacokinetics , Embryo, Mammalian/blood supply , Endoglin , Female , Image Enhancement/methods , Intracellular Signaling Peptides and Proteins/genetics , Intracellular Signaling Peptides and Proteins/metabolism , Kinetics , Male , Mice, Transgenic , Reproducibility of Results , Ultrasonography , Vascular Endothelial Growth Factor Receptor-2
13.
Mol Cell Biol ; 35(9): 1573-87, 2015 May.
Article in English | MEDLINE | ID: mdl-25691664

ABSTRACT

Development of the cardiovascular system is critically dependent on the ability of endothelial cells (ECs) to reorganize their intracellular actin architecture to facilitate migration, adhesion, and morphogenesis. Nck family cytoskeletal adaptors function as key mediators of actin dynamics in numerous cell types, though their role in EC biology remains largely unexplored. Here, we demonstrate an essential requirement for Nck within ECs. Mouse embryos lacking endothelial Nck1/2 expression develop extensive angiogenic defects that result in lethality at about embryonic day 10. Mutant embryos show immature vascular networks, with decreased vessel branching, aberrant perivascular cell recruitment, and reduced cardiac trabeculation. Strikingly, embryos deficient in endothelial Nck also fail to undergo the endothelial-to-mesenchymal transition (EnMT) required for cardiac valve morphogenesis, with loss of Nck disrupting expression of major EnMT markers, as well as suppressing mesenchymal outgrowth. Furthermore, we show that Nck-null ECs are unable to migrate downstream of vascular endothelial growth factor and angiopoietin-1, and they exhibit profound perturbations in cytoskeletal patterning, with disorganized cellular projections, impaired focal adhesion turnover, and disrupted actin-based signaling. Our collective findings thereby reveal a crucial role for Nck as a master regulator within the endothelium to control actin cytoskeleton organization, vascular network remodeling, and EnMT during cardiovascular development.


Subject(s)
Adaptor Proteins, Signal Transducing/genetics , Cardiovascular Abnormalities/embryology , Cardiovascular System/embryology , Endothelial Cells/metabolism , Epithelial-Mesenchymal Transition , Oncogene Proteins/genetics , Adaptor Proteins, Signal Transducing/metabolism , Animals , Cardiovascular Abnormalities/genetics , Cardiovascular System/metabolism , Cell Movement , Endothelial Cells/cytology , Gene Deletion , Mice , Mice, Transgenic , Oncogene Proteins/metabolism
14.
Nat Commun ; 6: 7329, 2015 Jun 16.
Article in English | MEDLINE | ID: mdl-26076835

ABSTRACT

Reprogramming is a dynamic process that can result in multiple pluripotent cell types emerging from divergent paths. Cell surface protein expression is a particularly desirable tool to categorize reprogramming and pluripotency as it enables robust quantification and enrichment of live cells. Here we use cell surface proteomics to interrogate mouse cell reprogramming dynamics and discover CD24 as a marker that tracks the emergence of reprogramming-responsive cells, while enabling the analysis and enrichment of transgene-dependent (F-class) and -independent (traditional) induced pluripotent stem cells (iPSCs) at later stages. Furthermore, CD24 can be used to delineate epiblast stem cells (EpiSCs) from embryonic stem cells (ESCs) in mouse pluripotent culture. Importantly, regulated CD24 expression is conserved in human pluripotent stem cells (PSCs), tracking the conversion of human ESCs to more naive-like PSC states. Thus, CD24 is a conserved marker for tracking divergent states in both reprogramming and standard pluripotent culture.


Subject(s)
CD24 Antigen/metabolism , Cellular Reprogramming , Human Embryonic Stem Cells/metabolism , Induced Pluripotent Stem Cells/metabolism , Mouse Embryonic Stem Cells/metabolism , Animals , Germ Layers/cytology , Human Embryonic Stem Cells/cytology , Humans , Induced Pluripotent Stem Cells/cytology , Mice , Mouse Embryonic Stem Cells/cytology , Stem Cells/cytology , Stem Cells/metabolism
15.
Ultrasound Med Biol ; 40(2): 389-99, 2014 Feb.
Article in English | MEDLINE | ID: mdl-24342913

ABSTRACT

As a tumor surrogate, the mouse embryo presents as an excellent alternative for examining the binding of angiogenesis-targeting microbubbles and assessing the quantitative nature of molecular ultrasound. We establish the validity of this model by developing a robust method to study microbubble kinetic behavior and investigate the reproducibility of targeted binding in the murine embryo. Vascular endothelial growth factor receptor 2Ā (VEGFR2)-targeted (MBV), rat immunoglobulin G2 (IgG2) control antibody-targeted (MBC) and untargeted (MBU) microbubbles were introduced into vasculature of living mouse embryos. Non-linear contrast-specific and B-mode ultrasound imaging, performed at 21 MHz with a Vevo-2100 scanner, was used to collect basic perfusion parameters and contrast mean power ratios for all bubble types. We observed a twofold increase (p < 0.001) in contrast mean power ratios for MBV (4.14Ā Ā± 1.78) compared with those for MBC (1.95Ā Ā± 0.78) and MBU (1.79Ā Ā± 0.45). Targeted imaging of endogenous endothelial cell surface markers in mouse embryos is possible with labeled microbubbles. The mouse embryo thus presents as a versatile model for testing the performance of ultrasound molecular targeting, where further development of quantitative imaging techniques may enable rapid evaluations of biomarker expression in studies of vascular development, disease and angiogenesis.


Subject(s)
Antibodies, Monoclonal/pharmacokinetics , Endothelium, Vascular/diagnostic imaging , Endothelium, Vascular/metabolism , Molecular Imaging/methods , Ultrasonography, Prenatal/methods , Vascular Endothelial Growth Factor Receptor-2/metabolism , Animals , Contrast Media/pharmacokinetics , Endothelium, Vascular/embryology , Female , Male , Mice , Microbubbles , Molecular Diagnostic Techniques/methods , Neoplasms/diagnostic imaging , Neoplasms/embryology , Neoplasms/metabolism , Reproducibility of Results , Sensitivity and Specificity
16.
Nat Commun ; 5: 5613, 2014 Dec 10.
Article in English | MEDLINE | ID: mdl-25494451

ABSTRACT

The ectopic expression of Oct4, Klf4, c-Myc and Sox2 (OKMS) transcription factors allows reprogramming of somatic cells into induced pluripotent stem cells (iPSCs). The reprogramming process, which involves a complex network of molecular events, is not yet fully characterized. Here we perform a quantitative mass spectrometry-based analysis to probe in-depth dynamic proteome changes during somatic cell reprogramming. Our data reveal defined waves of proteome resetting, with the first wave occurring 48 h after the activation of the reprogramming transgenes and involving specific biological processes linked to the c-Myc transcriptional network. A second wave of proteome reorganization occurs in a later stage of reprogramming, where we characterize the proteome of two distinct pluripotent cellular populations. In addition, the overlay of our proteome resource with parallel generated -omics data is explored to identify post-transcriptionally regulated proteins involved in key steps during reprogramming.

17.
Nat Commun ; 5: 5619, 2014 Dec 10.
Article in English | MEDLINE | ID: mdl-25493341

ABSTRACT

Reprogramming of somatic cells to induced pluripotent stem cells involves a dynamic rearrangement of the epigenetic landscape. To characterize this epigenomic roadmap, we have performed MethylC-seq, ChIP-seq (H3K4/K27/K36me3) and RNA-Seq on samples taken at several time points during murine secondary reprogramming as part of Project Grandiose. We find that DNA methylation gain during reprogramming occurs gradually, while loss is achieved only at the ESC-like state. Binding sites of activated factors exhibit focal demethylation during reprogramming, while ESC-like pluripotent cells are distinguished by extension of demethylation to the wider neighbourhood. We observed that genes with CpG-rich promoters demonstrate stable low methylation and strong engagement of histone marks, whereas genes with CpG-poor promoters are safeguarded by methylation. Such DNA methylation-driven control is the key to the regulation of ESC-pluripotency genes, including Dppa4, Dppa5a and Esrrb. These results reveal the crucial role that DNA methylation plays as an epigenetic switch driving somatic cells to pluripotency.

18.
Nat Commun ; 5: 5522, 2014 Dec 10.
Article in English | MEDLINE | ID: mdl-25494340

ABSTRACT

MicroRNAs (miRNAs) are critical to somatic cell reprogramming into induced pluripotent stem cells (iPSCs), however, exactly how miRNA expression changes support the transition to pluripotency requires further investigation. Here we use a murine secondary reprogramming system to sample cellular trajectories towards iPSCs or a novel pluripotent 'F-class' state and perform small RNA sequencing. We detect sweeping changes in an early and a late wave, revealing that distinct miRNA milieus characterize alternate states of pluripotency. miRNA isoform expression is common but surprisingly varies little between cell states. Referencing other omic data sets generated in parallel, we find that miRNA expression is changed through transcriptional and post-transcriptional mechanisms. miRNA transcription is commonly regulated by dynamic histone modification, while DNA methylation/demethylation consolidates these changes at multiple loci. Importantly, our results suggest that a novel subset of distinctly expressed miRNAs supports pluripotency in the F-class state, substituting for miRNAs that serve such roles in iPSCs.

19.
Proc Natl Acad Sci U S A ; 100(22): 12753-8, 2003 Oct 28.
Article in English | MEDLINE | ID: mdl-14530387

ABSTRACT

In mammals, the continuous production of hematopoietic cells (HCs) is sustained by a small number of hematopoietic stem cells (HSCs) residing in the bone marrow. Early HSC activity arises in the aorta-gonad mesonephros region, within cells localized to the ventral floor of the major blood vessels, suggesting that the first HSCs may be derived from cells capable of giving rise to the hematopoietic system and to the endothelial cells of the vasculature. TIE1 (TIE) and TIE2 (TEK) are related receptor tyrosine kinases with an embryonic expression pattern in endothelial cells, their precursors, and HCs, suggestive of a role in the divergence and function of both lineages. Indeed, gene targeting approaches have shown that TIE1, TIE2, and ligands for TIE2, the angiopoietins, are essential for vascular development and maintenance. To explore possible roles for these receptors in HCs, we have examined the ability of embryonic cells lacking both TIE1 and TIE2 to contribute to developmental and adult hematopoiesis by generating chimeric animals between normal embryonic cells and cells lacking these receptors. We show here that TIE receptors are not required for differentiation and proliferation of definitive hematopoietic lineages in the embryo and fetus; surprisingly, however, these receptors are specifically required during postnatal bone marrow hematopoiesis.


Subject(s)
Bone Marrow Cells/cytology , Gene Expression Regulation, Developmental/physiology , Hematopoiesis/physiology , Hematopoietic Stem Cells/physiology , Liver/embryology , Receptor, TIE-1/genetics , Receptor, TIE-2/genetics , Aging/physiology , Animals , Cells, Cultured , Chimera , Colony-Forming Units Assay , Hematopoietic Stem Cells/cytology , Ligands , Liver/cytology , Mice , Mice, Knockout , Receptor, TIE-1/deficiency , Receptor, TIE-2/deficiency
20.
Exp Lung Res ; 29(2): 113-22, 2003 Mar.
Article in English | MEDLINE | ID: mdl-12554357

ABSTRACT

The molecular mechanisms responsible for organ-specific differences in vascular development are not well established. Animals lacking the receptor tyrosine kinase Tie1 die of hemorrhage and pulmonary edema. Furthermore, cells lacking Tie1 are excluded from blood vessels of the mature lung. These findings suggest the importance of Tie1 in the pulmonary vasculature. We quantified the organ-specific expression of Tie1 during embryonic and postnatal murine development using both quantitative real-time polymerase chain reaction (PCR) and chemiluminescence employing a tie1.lacZ reporter. In the lung, Tie1 expression increases markedly immediately prior to birth and rises further in the newborn animal, a pattern not found in other organs. Furthermore, expression of Tie1 in the lung is also unique by its persistent increase in the adult animal. This unique pattern of Tie1 gene expression in the embryonic and mature lung supports a distinct role for Tie1 in the development and function of the pulmonary vasculature.


Subject(s)
Gene Expression Regulation, Developmental , Lung/metabolism , Receptor Protein-Tyrosine Kinases/genetics , Receptors, Cell Surface/genetics , Animals , Animals, Newborn , DNA Primers/chemistry , Endothelium, Vascular/embryology , Endothelium, Vascular/metabolism , Female , Genes, Reporter , Heterozygote , Lac Operon/genetics , Lung/blood supply , Lung/embryology , Male , Mice , Mice, Inbred Strains , Mice, Transgenic , Oligonucleotide Probes/chemistry , Pregnancy , RNA, Messenger/metabolism , Receptor Protein-Tyrosine Kinases/metabolism , Receptor, TIE-1 , Receptors, Cell Surface/metabolism , Receptors, TIE , Reverse Transcriptase Polymerase Chain Reaction
SELECTION OF CITATIONS
SEARCH DETAIL