Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 27
Filter
1.
Nat Cell Biol ; 24(7): 1114-1128, 2022 07.
Article in English | MEDLINE | ID: mdl-35817961

ABSTRACT

The mammalian heart arises from various populations of Mesp1-expressing cardiovascular progenitors (CPs) that are specified during the early stages of gastrulation. Mesp1 is a transcription factor that acts as a master regulator of CP specification and differentiation. However, how Mesp1 regulates the chromatin landscape of nascent mesodermal cells to define the temporal and spatial patterning of the distinct populations of CPs remains unknown. Here, by combining ChIP-seq, RNA-seq and ATAC-seq during mouse pluripotent stem cell differentiation, we defined the dynamic remodelling of the chromatin landscape mediated by Mesp1. We identified different enhancers that are temporally regulated to erase the pluripotent state and specify the pools of CPs that mediate heart development. We identified Zic2 and Zic3 as essential cofactors that act with Mesp1 to regulate its transcription-factor activity at key mesodermal enhancers, thereby regulating the chromatin remodelling and gene expression associated with the specification of the different populations of CPs in vivo. Our study identifies the dynamics of the chromatin landscape and enhancer remodelling associated with temporal patterning of early mesodermal cells into the distinct populations of CPs that mediate heart development.


Subject(s)
Basic Helix-Loop-Helix Transcription Factors , Chromatin , Animals , Basic Helix-Loop-Helix Transcription Factors/metabolism , Cell Differentiation/genetics , Chromatin/genetics , Chromatin/metabolism , Enhancer Elements, Genetic/genetics , Gene Expression Regulation, Developmental , Heart , Homeodomain Proteins/metabolism , Mammals/metabolism , Mesoderm , Mice , Transcription Factors/genetics , Transcription Factors/metabolism
3.
Nat Commun ; 12(1): 6132, 2021 10 21.
Article in English | MEDLINE | ID: mdl-34675200

ABSTRACT

Studies of mechanical signalling are typically performed by comparing cells cultured on soft and stiff hydrogel-based substrates. However, it is challenging to independently and robustly control both substrate stiffness and extracellular matrix tethering to substrates, making matrix tethering a potentially confounding variable in mechanical signalling investigations. Moreover, unstable matrix tethering can lead to poor cell attachment and weak engagement of cell adhesions. To address this, we developed StemBond hydrogels, a hydrogel in which matrix tethering is robust and can be varied independently of stiffness. We validate StemBond hydrogels by showing that they provide an optimal system for culturing mouse and human pluripotent stem cells. We further show how soft StemBond hydrogels modulate stem cell function, partly through stiffness-sensitive ERK signalling. Our findings underline how substrate mechanics impact mechanosensitive signalling pathways regulating self-renewal and differentiation, indicating that optimising the complete mechanical microenvironment will offer greater control over stem cell fate specification.


Subject(s)
Cell Culture Techniques/instrumentation , Extracellular Matrix/chemistry , Hydrogels/chemistry , Pluripotent Stem Cells/cytology , Animals , Biomechanical Phenomena , Cell Adhesion , Cell Differentiation , Cells, Cultured , Extracellular Matrix/metabolism , Humans , Mechanotransduction, Cellular , Mice , Pluripotent Stem Cells/chemistry , Pluripotent Stem Cells/metabolism
4.
Proc Natl Acad Sci U S A ; 118(38)2021 09 21.
Article in English | MEDLINE | ID: mdl-34518230

ABSTRACT

Genome remethylation is essential for mammalian development but specific reasons are unclear. Here we examined embryonic stem (ES) cell fate in the absence of de novo DNA methyltransferases. We observed that ES cells deficient for both Dnmt3a and Dnmt3b are rapidly eliminated from chimeras. On further investigation we found that in vivo and in vitro the formative pluripotency transition is derailed toward production of trophoblast. This aberrant trajectory is associated with failure to suppress activation of Ascl2Ascl2 encodes a bHLH transcription factor expressed in the placenta. Misexpression of Ascl2 in ES cells provokes transdifferentiation to trophoblast-like cells. Conversely, Ascl2 deletion rescues formative transition of Dnmt3a/b mutants and improves contribution to chimeric epiblast. Thus, de novo DNA methylation safeguards against ectopic activation of Ascl2 However, Dnmt3a/b-deficient cells remain defective in ongoing embryogenesis. We surmise that multiple developmental transitions may be secured by DNA methylation silencing potentially disruptive genes.


Subject(s)
DNA Methylation/genetics , Embryonic Stem Cells/physiology , Animals , Basic Helix-Loop-Helix Transcription Factors/genetics , Cell Differentiation/genetics , Cells, Cultured , DNA (Cytosine-5-)-Methyltransferases/genetics , Embryonic Development/genetics , Mice , Trophoblasts/physiology , DNA Methyltransferase 3B
5.
Stem Cell Reports ; 16(5): 1182-1196, 2021 05 11.
Article in English | MEDLINE | ID: mdl-33035465

ABSTRACT

Mammalian embryogenesis is a paradigm of regulative development as mouse embryos show plasticity in the regulation of cell fate, cell number, and tissue morphogenesis. However, the mechanisms behind embryo plasticity remain largely unknown. Here, we determine how mouse embryos respond to an increase in cell numbers to regulate the timing and mechanism of embryonic morphogenesis, leading to the formation of the pro-amniotic cavity. Using embryos and embryonic stem cell aggregates of different size, we show that while pro-amniotic cavity formation in normal-sized embryos is achieved through basement membrane-induced polarization and exocytosis, cavity formation of increased-size embryos is delayed and achieved through apoptosis of cells that lack contact with the basement membrane. Importantly, blocking apoptosis, both genetically and pharmacologically, alters pro-amniotic cavity formation but does not affect size regulation in enlarged embryos. We conclude that the regulation of embryonic size and morphogenesis, albeit concomitant, have distinct molecular underpinnings.


Subject(s)
Embryo, Mammalian/anatomy & histology , Morphogenesis , Amnion/embryology , Animals , Apoptosis , Cell Aggregation , Embryo, Mammalian/cytology , Embryonic Stem Cells/cytology , Embryonic Stem Cells/metabolism , Female , Mice, Inbred C57BL , Mice, Inbred CBA , Organ Size , Time Factors
6.
Cell Stem Cell ; 28(3): 453-471.e8, 2021 03 04.
Article in English | MEDLINE | ID: mdl-33271069

ABSTRACT

Pluripotent cells emerge as a naive founder population in the blastocyst, acquire capacity for germline and soma formation, and then undergo lineage priming. Mouse embryonic stem cells (ESCs) and epiblast-derived stem cells (EpiSCs) represent the initial naive and final primed phases of pluripotency, respectively. Here, we investigate the intermediate formative stage. Using minimal exposure to specification cues, we derive stem cells from formative mouse epiblast. Unlike ESCs or EpiSCs, formative stem (FS) cells respond directly to germ cell induction. They colonize somatic tissues and germline in chimeras. Whole-transcriptome analyses show similarity to pre-gastrulation formative epiblast. Signal responsiveness and chromatin accessibility features reflect lineage capacitation. Furthermore, FS cells show distinct transcription factor dependencies, relying critically on Otx2. Finally, FS cell culture conditions applied to human naive cells or embryos support expansion of similar stem cells, consistent with a conserved staging post on the trajectory of mammalian pluripotency.


Subject(s)
Pluripotent Stem Cells , Animals , Blastocyst , Cell Differentiation , Embryonic Stem Cells , Germ Layers , Humans , Mice
7.
Cell Stem Cell ; 25(3): 388-406.e8, 2019 09 05.
Article in English | MEDLINE | ID: mdl-31422912

ABSTRACT

Understanding how cell identity transitions occur and whether there are multiple paths between the same beginning and end states are questions of wide interest. Here we show that acquisition of naive pluripotency can follow transcriptionally and mechanistically distinct routes. Starting from post-implantation epiblast stem cells (EpiSCs), one route advances through a mesodermal state prior to naive pluripotency induction, whereas another transiently resembles the early inner cell mass and correspondingly gains greater developmental potency. These routes utilize distinct signaling networks and transcription factors but subsequently converge on the same naive endpoint, showing surprising flexibility in mechanisms underlying identity transitions and suggesting that naive pluripotency is a multidimensional attractor state. These route differences are reconciled by precise expression of Oct4 as a unifying, essential, and sufficient feature. We propose that fine-tuned regulation of this "transition factor" underpins multidimensional access to naive pluripotency, offering a conceptual framework for understanding cell identity transitions.


Subject(s)
Blastocyst Inner Cell Mass/physiology , Germ Layers/physiology , Octamer Transcription Factor-3/metabolism , Pluripotent Stem Cells/physiology , Animals , Cell Differentiation , Cell Line , Cell Plasticity , Cellular Reprogramming , Female , Gene Expression Regulation, Developmental , Gene Regulatory Networks , Mice , Mice, Inbred C57BL , Octamer Transcription Factor-3/genetics , Signal Transduction
8.
Genes Dev ; 31(9): 876-888, 2017 05 01.
Article in English | MEDLINE | ID: mdl-28546514

ABSTRACT

The nuclear matrix protein Cip1-interacting zinc finger protein 1 (CIZ1) promotes DNA replication in association with cyclins and has been linked to adult and pediatric cancers. Here we show that CIZ1 is highly enriched on the inactive X chromosome (Xi) in mouse and human female cells and is retained by interaction with the RNA-dependent nuclear matrix. CIZ1 is recruited to Xi in response to expression of X inactive-specific transcript (Xist) RNA during the earliest stages of X inactivation in embryonic stem cells and is dependent on the C-terminal nuclear matrix anchor domain of CIZ1 and the E repeats of Xist CIZ1-null mice, although viable, display fully penetrant female-specific lymphoproliferative disorder. Interestingly, in mouse embryonic fibroblast cells derived from CIZ1-null embryos, Xist RNA localization is disrupted, being highly dispersed through the nucleoplasm rather than focal. Focal localization is reinstated following re-expression of CIZ1. Focal localization of Xist RNA is also disrupted in activated B and T cells isolated from CIZ1-null animals, suggesting a possible explanation for female-specific lymphoproliferative disorder. Together, these findings suggest that CIZ1 has an essential role in anchoring Xist to the nuclear matrix in specific somatic lineages.


Subject(s)
Gene Expression Regulation , Lymphoproliferative Disorders/pathology , Nuclear Proteins/physiology , RNA, Long Noncoding/metabolism , X Chromosome Inactivation , X Chromosome/metabolism , Animals , Cell Differentiation , Cells, Cultured , Embryo, Mammalian/metabolism , Embryo, Mammalian/pathology , Embryonic Stem Cells/metabolism , Embryonic Stem Cells/pathology , Female , Fibroblasts/metabolism , Fibroblasts/pathology , Humans , Lymphoproliferative Disorders/genetics , Lymphoproliferative Disorders/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Inbred CBA , Mice, Knockout , RNA, Long Noncoding/genetics , Sex Characteristics , X Chromosome/genetics
9.
Biol Open ; 6(1): 92-99, 2017 Jan 15.
Article in English | MEDLINE | ID: mdl-27934662

ABSTRACT

Myocardial injury in mammals leads to heart failure through pathological cardiac remodelling that includes hypertrophy, fibrosis and ventricular dilatation. Central to this is inability of the mammalian cardiomyocyte to self-renew due to entering a quiescent state after birth. Modulation of the cardiomyocyte cell-cycle after injury is therefore a target mechanism to limit damage and potentiate repair and regeneration. Here, we show that cardiomyocyte-specific over-expression of the nuclear-matrix--associated DNA replication protein, CIZ1, extends their window of proliferation during cardiac development, delaying onset of terminal differentiation without compromising function. CIZ1-expressing hearts are enlarged, but the cardiomyocytes are smaller with an overall increase in number, correlating with increased DNA replication after birth and retention of an increased proportion of mono-nucleated cardiomyocytes into adulthood. Furthermore, these CIZ1 induced changes in the heart reduce the impact of myocardial injury, identifying CIZ1 as a putative therapeutic target for cardiac repair.

10.
Diabetes ; 65(1): 120-8, 2016 Jan.
Article in English | MEDLINE | ID: mdl-26450994

ABSTRACT

CTLA-4 is a critical "checkpoint" regulator in autoimmunity. Variation in CTLA-4 isoform expression has been linked to type 1 diabetes development in human and NOD mouse studies. In the NOD mouse, a causative link between increased expression of the minor isoform ligand-independent CTLA-4 and a reduction in diabetes has become widely accepted. Altered splicing of CTLA-4 has been attributed to a single nucleotide polymorphism (SNP) in Ctla4 exon2 (e2_77A/G). To investigate this link, we have used NOD embryonic stem (ES) cells to generate a novel NOD transgenic line with the 77A/G SNP. This strain phenocopies the increase in splicing toward the liCTLA4 isoform seen in B10 Idd5.1 mice. Crucially, the SNP does not alter the spontaneous incidence of diabetes, the incidence of cyclophosphamide-induced diabetes, or the activation of diabetogenic T-cell receptor transgenic CD4(+) T cells after adoptive transfer. Our results show that one or more of the many other linked genetic variants between the B10 and NOD genome are required for the diabetes protection conferred by Idd5.1. With the NOD mouse model closely mimicking the human disease, our data demonstrate that knock-in transgenic mice on the NOD background can test causative mutations relevant in human diabetes.


Subject(s)
Autoimmunity/genetics , CD4-Positive T-Lymphocytes/immunology , CTLA-4 Antigen/genetics , Diabetes Mellitus, Type 1/genetics , RNA Splicing/genetics , RNA, Messenger/metabolism , Adoptive Transfer , Animals , Autoimmunity/immunology , CTLA-4 Antigen/immunology , Diabetes Mellitus, Type 1/immunology , Gene Knock-In Techniques , Genetic Predisposition to Disease , Mice , Mice, Inbred NOD , Mice, Transgenic , Mouse Embryonic Stem Cells , Polymorphism, Single Nucleotide , Protein Isoforms
12.
Cell ; 158(6): 1254-1269, 2014 Sep 11.
Article in English | MEDLINE | ID: mdl-25215486

ABSTRACT

Current human pluripotent stem cells lack the transcription factor circuitry that governs the ground state of mouse embryonic stem cells (ESC). Here, we report that short-term expression of two components, NANOG and KLF2, is sufficient to ignite other elements of the network and reset the human pluripotent state. Inhibition of ERK and protein kinase C sustains a transgene-independent rewired state. Reset cells self-renew continuously without ERK signaling, are phenotypically stable, and are karyotypically intact. They differentiate in vitro and form teratomas in vivo. Metabolism is reprogrammed with activation of mitochondrial respiration as in ESC. DNA methylation is dramatically reduced and transcriptome state is globally realigned across multiple cell lines. Depletion of ground-state transcription factors, TFCP2L1 or KLF4, has marginal impact on conventional human pluripotent stem cells but collapses the reset state. These findings demonstrate feasibility of installing and propagating functional control circuitry for ground-state pluripotency in human cells.


Subject(s)
Homeodomain Proteins/metabolism , Kruppel-Like Transcription Factors/metabolism , Pluripotent Stem Cells/metabolism , Animals , Cytological Techniques , Embryonic Stem Cells/cytology , Embryonic Stem Cells/metabolism , Epigenesis, Genetic , Humans , Kruppel-Like Factor 4 , Mice , Mitochondria/metabolism , Nanog Homeobox Protein , Pluripotent Stem Cells/cytology , Transcription Factors/metabolism , Transcriptome
13.
Development ; 141(5): 1001-10, 2014 Mar.
Article in English | MEDLINE | ID: mdl-24504341

ABSTRACT

The transcription factor Oct4 is required in vitro for establishment and maintenance of embryonic stem cells and for reprogramming somatic cells to pluripotency. In vivo, it prevents the ectopic differentiation of early embryos into trophoblast. Here, we further explore the role of Oct4 in blastocyst formation and specification of epiblast versus primitive endoderm lineages using conditional genetic deletion. Experiments involving mouse embryos deficient for both maternal and zygotic Oct4 suggest that it is dispensable for zygote formation, early cleavage and activation of Nanog expression. Nanog protein is significantly elevated in the presumptive inner cell mass of Oct4 null embryos, suggesting an unexpected role for Oct4 in attenuating the level of Nanog, which might be significant for priming differentiation during epiblast maturation. Induced deletion of Oct4 during the morula to blastocyst transition disrupts the ability of inner cell mass cells to adopt lineage-specific identity and acquire the molecular profile characteristic of either epiblast or primitive endoderm. Sox17, a marker of primitive endoderm, is not detected following prolonged culture of such embryos, but can be rescued by provision of exogenous FGF4. Interestingly, functional primitive endoderm can be rescued in Oct4-deficient embryos in embryonic stem cell complementation assays, but only if the host embryos are at the pre-blastocyst stage. We conclude that cell fate decisions within the inner cell mass are dependent upon Oct4 and that Oct4 is not cell-autonomously required for the differentiation of primitive endoderm derivatives, as long as an appropriate developmental environment is established.


Subject(s)
Blastocyst/metabolism , Octamer Transcription Factor-3/metabolism , Animals , Blastocyst/cytology , Embryo, Mammalian/cytology , Embryo, Mammalian/metabolism , Endoderm/cytology , Endoderm/metabolism , Female , Gene Expression Regulation, Developmental , HMGB Proteins/genetics , HMGB Proteins/metabolism , Homeodomain Proteins/genetics , Homeodomain Proteins/metabolism , Male , Mice , Morula/cytology , Morula/metabolism , Nanog Homeobox Protein , Octamer Transcription Factor-3/genetics , Oocytes/cytology , Oocytes/metabolism , Pregnancy , SOXF Transcription Factors/genetics , SOXF Transcription Factors/metabolism , Zygote/cytology , Zygote/metabolism
14.
Nat Struct Mol Biol ; 20(3): 311-6, 2013 Mar.
Article in English | MEDLINE | ID: mdl-23416945

ABSTRACT

Naive pluripotent embryonic stem cells (ESCs) and embryonic germ cells (EGCs) are derived from the preimplantation epiblast and primordial germ cells (PGCs), respectively. We investigated whether differences exist between ESCs and EGCs, in view of their distinct developmental origins. PGCs are programmed to undergo global DNA demethylation; however, we find that EGCs and ESCs exhibit equivalent global DNA methylation levels. Inhibition of MEK and Gsk3b by 2i conditions leads to pronounced reduction in DNA methylation in both cell types. This is driven by Prdm14 and is associated with downregulation of Dnmt3a and Dnmt3b. However, genomic imprints are maintained in 2i, and we report derivation of EGCs with intact genomic imprints. Collectively, our findings establish that culture in 2i instills a naive pluripotent state with a distinctive epigenetic configuration that parallels molecular features observed in both the preimplantation epiblast and nascent PGCs.


Subject(s)
DNA Methylation , Embryonic Stem Cells/physiology , Germ Cells/cytology , Pluripotent Stem Cells/physiology , Animals , Benzamides/pharmacology , Cell Differentiation , Cells, Cultured/drug effects , DNA (Cytosine-5-)-Methyltransferases/metabolism , DNA Methyltransferase 3A , DNA-Binding Proteins , Diphenylamine/analogs & derivatives , Diphenylamine/pharmacology , Enzyme Inhibitors/pharmacology , Epigenesis, Genetic , Gene Expression Profiling , Genomic Imprinting , Germ Cells/physiology , Germ Layers/cytology , Glycogen Synthase Kinase 3/antagonists & inhibitors , Glycogen Synthase Kinase 3/metabolism , Glycogen Synthase Kinase 3 beta , MAP Kinase Kinase Kinases/antagonists & inhibitors , MAP Kinase Kinase Kinases/metabolism , Mice , Mice, Inbred C57BL , Pluripotent Stem Cells/cytology , Pyridines/pharmacology , Pyrimidines/pharmacology , RNA-Binding Proteins , Transcription Factors/genetics , Transcription Factors/metabolism , Transcriptome , DNA Methyltransferase 3B
15.
Environ Sci Technol ; 47(13): 6995-7001, 2013 Jul 02.
Article in English | MEDLINE | ID: mdl-23383991

ABSTRACT

Neither amplification nor attenuation of antibiotic resistance genes (ARG) in the environment are well understood processes. Here, we report on continuous culture and batch experiments to determine how tetracycline (TC), aerobic vs anaerobic conditions, bacterial growth rate, and medium richness affect the maintenance of plasmid-borne TC resistance (Tet(R)) genes. The response of E. coli (a model resistant strain excreted by farm animals) versus Pseudomonas aeruginosa (a model bacterium that could serve as a reservoir for ARGs in the environment) were compared to gain insight into response variability. Complete loss of the Tet(R) RP1 plasmid (56 kb) occurred for P. aeruginosa in the absence of TC, and faster loss was observed in continuous culture at higher growth rates. In contrast, E. coli retained its smaller pSC101 plasmid (9.3 kb) after 500 generations without TC (albeit at lower levels, with ratios of resistance to 16S rDNA genes decreasing by about 2-fold). A higher rate of ARG loss was observed in P. aeruginosa when grown in minimal growth medium (M9) than in richer Luria broth. Faster ARG loss occurred in E. coli under anaerobic (fermentative) conditions than under aerobic conditions. Thus, in these two model strains it was observed that conditions that ease the metabolic burden of plasmid reproduction (e.g., higher substrate and O2 availability) enhanced resistance plasmid maintenance; such conditions (in the presence of residual antibiotics) may be conducive to the establishment and preservation of ARG reservoirs in the environment. These results underscore the need to consider antibiotic concentrations, redox conditions, and substrate availability in efforts to evaluate ARG propagation and natural attenuation.


Subject(s)
Anti-Bacterial Agents/pharmacology , Escherichia coli/drug effects , Pseudomonas aeruginosa/drug effects , Tetracycline Resistance/genetics , Tetracycline/pharmacology , Antiporters/genetics , Bacterial Proteins/genetics , DNA, Bacterial/genetics , Escherichia coli/physiology , Oxygen/metabolism , Plasmids , Pseudomonas aeruginosa/physiology , Repressor Proteins/genetics
16.
Biol Open ; 1(1): 58-65, 2012 Jan 15.
Article in English | MEDLINE | ID: mdl-23213369

ABSTRACT

The ability of cultured pluripotent cells to contribute to the germline of chimaeric animals is essential to their utility for genetic manipulation. In the three years since rat embryonic stem (ES) cells were first reported the anticipated proliferation of genetically modified rat models from this new resource has not been realised. Culture instability, karyotypic anomalies, and strain variation are postulated to contribute to poor germline colonisation capacity. The resolution of these issues is essential to bring pluripotent cell-based genetic manipulation technology in the rat to the level of efficiency achieved in the mouse. Recent reports have described various alternative methods to maintain rat ES cells that include provision of additional small molecules and selective passaging methods. In contrast, we report that euploid, germline competent rat ES and embryonic germ (EG) cell lines can be maintained by simple adherent culture methods in defined medium supplemented with the original two inhibitors (2i) of the mitogen-activated protein kinase (ERK1/2) cascade and of glycogen synthase kinase 3, in combination with the cytokine leukaemia inhibitory factor (LIF). We demonstrate genetic modification, clonal expansion and transmission through the germline of rat ES and EG cell lines. We also describe a marked preference for full-term chimaera contribution when SD strain blastocysts are used as recipients for either DA or SD pluripotent stem cells.

17.
ScientificWorldJournal ; 2012: 474582, 2012.
Article in English | MEDLINE | ID: mdl-22566770

ABSTRACT

PURPOSE: We sought to determine the prevalence of clinically significant non-cardiac abnormalities found in pediatric and adult patients undergoing cardiac magnetic resonance imaging (CMRI), and understand the impact of age on it's occurrence. METHODS: We retrospectively reviewed all patients undergoing CMRI between May 2004 and July 2007. Findings were considered significant if they required radiographic or clinical follow-up. RESULTS: A total of 408 patients underwent CMRI during the study period. Twenty two (16%) pediatric patients (age < 19 years, n = 135) were found to have a total of 22 non- cardiac abnormalities, 3 of which were clinically significant. Sixty four (23%) adult patients (age > 19 years, n = 273) were found to have a total of 77 non-cardiac abnormalities, 33 of which were clinically significant. The prevalence of clinically significant non-cardiac abnormalities was 2% in the pediatric cohort and 11% in the adult cohort (P = 0.05). Within the adult population, the prevalence of significant non-cardiac abnormalities increased with advancing age (P = 0.05). CONCLUSIONS: In a population of unselected patients undergoing CMRI, unanticipated noncardiac abnormalities were frequently seen. A small number of these were significant, with the prevalence increasing with age.


Subject(s)
Magnetic Resonance Imaging , Mediastinal Diseases/diagnosis , Pleural Effusion/diagnosis , Syringomyelia/diagnosis , Adolescent , Adult , Age Factors , Aged , Aged, 80 and over , Aortic Diseases/diagnosis , Child , Child, Preschool , Female , Heart Defects, Congenital/diagnosis , Humans , Infant , Male , Mediastinal Diseases/epidemiology , Middle Aged , Pleural Effusion/epidemiology , Prevalence , Retrospective Studies , Syringomyelia/epidemiology , Young Adult
18.
Cell Stem Cell ; 10(5): 583-94, 2012 May 04.
Article in English | MEDLINE | ID: mdl-22560079

ABSTRACT

Transcriptional heterogeneity within embryonic stem cell (ESC) populations has been suggested as a mechanism by which a seemingly homogeneous cell population can initiate differentiation into an array of different cell types. Chromatin remodeling proteins have been shown to control transcriptional variability in yeast and to be important for mammalian ESC lineage commitment. Here we show that the Nucleosome Remodeling and Deacetylation (NuRD) complex, which is required for ESC lineage commitment, modulates both transcriptional heterogeneity and the dynamic range of a set of pluripotency genes in ESCs. In self-renewing conditions, the influence of NuRD at these genes is balanced by the opposing action of self-renewal factors. Upon loss of self-renewal factors, the action of NuRD is sufficient to silence transcription of these pluripotency genes, allowing cells to exit self-renewal. We propose that modulation of transcription levels by NuRD is key to maintaining the differentiation responsiveness of pluripotent cells.


Subject(s)
Embryonic Stem Cells/physiology , Gene Expression Regulation, Developmental , Mi-2 Nucleosome Remodeling and Deacetylase Complex/metabolism , Pluripotent Stem Cells/physiology , Animals , Cell Differentiation/genetics , Cell Lineage/genetics , Cells, Cultured , DNA-Binding Proteins/genetics , Genetic Heterogeneity , Mi-2 Nucleosome Remodeling and Deacetylase Complex/genetics , Mice , Mice, Knockout , Transcription Factors/genetics
19.
Dev Biol ; 366(2): 290-7, 2012 Jun 15.
Article in English | MEDLINE | ID: mdl-22537489

ABSTRACT

Hair follicle development and growth are regulated by Wnt signalling and depend on interactions between epidermal cells and a population of fibroblasts at the base of the follicle, known as the dermal papilla (DP). DP cells have a distinct gene expression signature from non-DP dermal fibroblasts. However, their origins are largely unknown. By generating chimeric mice and performing skin reconstitution assays we show that, irrespective of whether DP form during development, are induced by epidermal Wnt activation in adult skin or assemble from disaggregated cells, they are polyclonal in origin. While fibroblast proliferation is necessary for hair follicle formation in skin reconstitution assays, mitotically inhibited cells readily contribute to DP. Although new hair follicles do not usually develop in adult skin, adult dermal fibroblasts are competent to contribute to DP during hair follicle neogenesis, irrespective of whether they originate from skin in the resting or growth phase of the hair cycle or skin with ß-catenin-induced ectopic follicles. We propose that during skin reconstitution fibroblasts may be induced to become DP cells by interactions with hair follicle epidermal cells, rather than being derived from a distinct subpopulation of cells.


Subject(s)
Cell Communication , Cell Differentiation , Dermis/cytology , Hair Follicle/cytology , Hair/growth & development , Animals , Epidermal Cells , Fibroblasts/cytology , Hair/cytology , Mice , Wnt Signaling Pathway
20.
Stem Cells Transl Med ; 1(12): 855-65, 2012 Dec.
Article in English | MEDLINE | ID: mdl-23283547

ABSTRACT

Induced pluripotent stem cells (iPSCs) have the potential to generate patient-specific tissues for disease modeling and regenerative medicine applications. However, before iPSC technology can progress to the translational phase, several obstacles must be overcome. These include uncertainty regarding the ideal somatic cell type for reprogramming, the low kinetics and efficiency of reprogramming, and karyotype discrepancies between iPSCs and their somatic precursors. Here we describe the use of late-outgrowth endothelial progenitor cells (L-EPCs), which possess several favorable characteristics, as a cellular substrate for the generation of iPSCs. We have developed a protocol that allows the reliable isolation of L-EPCs from peripheral blood mononuclear cell preparations, including frozen samples. As a proof-of-principle for clinical applications we generated EPC-iPSCs from both healthy individuals and patients with heritable and idiopathic forms of pulmonary arterial hypertension. L-EPCs grew clonally; were highly proliferative, passageable, and bankable; and displayed higher reprogramming kinetics and efficiencies compared with dermal fibroblasts. Unlike fibroblasts, the high efficiency of L-EPC reprogramming allowed for the reliable generation of iPSCs in a 96-well format, which is compatible with high-throughput platforms. Array comparative genome hybridization analysis of L-EPCs versus donor-matched circulating monocytes demonstrated that L-EPCs have normal karyotypes compared with their subject's reference genome. In addition, >80% of EPC-iPSC lines tested did not acquire any copy number variations during reprogramming compared with their parent L-EPC line. This work identifies L-EPCs as a practical and efficient cellular substrate for iPSC generation, with the potential to address many of the factors currently limiting the translation of this technology.


Subject(s)
Adult Stem Cells/cytology , Cell Culture Techniques/methods , Endothelium, Vascular/cytology , Hematopoietic Stem Cells/cytology , Induced Pluripotent Stem Cells/cytology , Adult , Adult Stem Cells/physiology , Animals , Cell Differentiation/physiology , Cell Lineage/physiology , Cellular Reprogramming/physiology , Hematopoietic Stem Cells/physiology , Humans , Induced Pluripotent Stem Cells/physiology , Karyotyping , Leukocytes, Mononuclear/cytology , Leukocytes, Mononuclear/physiology , Mice , Mice, SCID , Neoplasm Transplantation , Regenerative Medicine/methods , Stem Cell Transplantation/methods , Teratoma/pathology
SELECTION OF CITATIONS
SEARCH DETAIL