Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 38
Filter
Add more filters










Publication year range
1.
J Vis Exp ; (159)2020 05 10.
Article in English | MEDLINE | ID: mdl-32449724

ABSTRACT

Custom designed endonucleases, such as RNA-guided Clustered Regularly Interspaced Short Palindromic Repeats (CRISPR)-Cas9, enable efficient genome editing in mammalian cells. Here we describe detailed procedures to seamlessly genome edit the hepatocyte nuclear factor 4 alpha (HNF4α) locus as an example in human pluripotent stem cells. Combining a piggyBac-based donor plasmid and the CRISPR-Cas9 nickase mutant in a two-step genetic selection, we demonstrate correct and efficient targeting of the HNF4α locus.


Subject(s)
Gene Editing/methods , Pluripotent Stem Cells/metabolism , Point Mutation/genetics , Base Sequence , CRISPR-Cas Systems/genetics , DNA Transposable Elements/genetics , Genetic Vectors/genetics , Genome, Human , Humans
2.
iScience ; 16: 206-217, 2019 Jun 28.
Article in English | MEDLINE | ID: mdl-31185456

ABSTRACT

During mammalian development, liver differentiation is driven by signals that converge on multiple transcription factor networks. The hepatocyte nuclear factor signaling network is known to be essential for hepatocyte specification and maintenance. In this study, we have generated deletion and point mutants of hepatocyte nuclear factor-4alpha (HNF4α) to precisely evaluate the function of protein domains during hepatocyte specification from human pluripotent stem cells. We demonstrate that nuclear HNF4α is essential for hepatic progenitor specification, and the introduction of point mutations in HNF4α's Small Ubiquitin-like Modifier (SUMO) consensus motif leads to disrupted hepatocyte differentiation. Taking a multiomics approach, we identified key deficiencies in cell biology, which included dysfunctional metabolism, substrate adhesion, tricarboxylic acid cycle flux, microRNA transport, and mRNA processing. In summary, the combination of genome editing and multiomics analyses has provided valuable insight into the diverse functions of HNF4α during pluripotent stem cell entry into the hepatic lineage and during hepatocellular differentiation.

3.
Stem Cell Reports ; 10(6): 1895-1907, 2018 06 05.
Article in English | MEDLINE | ID: mdl-29779895

ABSTRACT

Human embryonic stem cells (hESCs) display substantial heterogeneity in gene expression, implying the existence of discrete substates within the stem cell compartment. To determine whether these substates impact fate decisions of hESCs we used a GFP reporter line to investigate the properties of fractions of putative undifferentiated cells defined by their differential expression of the endoderm transcription factor, GATA6, together with the hESC surface marker, SSEA3. By single-cell cloning, we confirmed that substates characterized by expression of GATA6 and SSEA3 include pluripotent stem cells capable of long-term self-renewal. When clonal stem cell colonies were formed from GATA6-positive and GATA6-negative cells, more of those derived from GATA6-positive cells contained spontaneously differentiated endoderm cells than similar colonies derived from the GATA6-negative cells. We characterized these discrete cellular states using single-cell transcriptomic analysis, identifying a potential role for SOX17 in the establishment of the endoderm-biased stem cell state.


Subject(s)
Cell Self Renewal , Endoderm/cytology , Human Embryonic Stem Cells/cytology , Human Embryonic Stem Cells/metabolism , Biomarkers , Cell Differentiation/genetics , GATA6 Transcription Factor/genetics , GATA6 Transcription Factor/metabolism , Gene Expression Profiling , Genes, Reporter , Humans , Immunophenotyping , Single-Cell Analysis/methods
4.
Dev Cell ; 44(3): 362-377.e7, 2018 02 05.
Article in English | MEDLINE | ID: mdl-29290585

ABSTRACT

ETV6-RUNX1 is associated with childhood acute B-lymphoblastic leukemia (cALL) functioning as a first-hit mutation that initiates a clinically silent pre-leukemia in utero. Because lineage commitment hierarchies differ between embryo and adult, and the impact of oncogenes is cell-context dependent, we hypothesized that the childhood affiliation of ETV6-RUNX1 cALL reflects its origins in a progenitor unique to embryonic life. We characterize the first emerging B cells in first-trimester human embryos, identifying a developmentally restricted CD19-IL-7R+ progenitor compartment, which transitions from a myeloid to lymphoid program during ontogeny. This developmental series is recapitulated in differentiating human pluripotent stem cells (hPSCs), thereby providing a model for the initiation of cALL. Genome-engineered hPSCs expressing ETV6-RUNX1 from the endogenous ETV6 locus show expansion of the CD19-IL-7R+ compartment, show a partial block in B lineage commitment, and produce proB cells with aberrant myeloid gene expression signatures and potential: features (collectively) consistent with a pre-leukemic state.


Subject(s)
B-Lymphocytes/pathology , Core Binding Factor Alpha 2 Subunit/metabolism , Embryonic Development , Gene Expression Regulation, Leukemic , Induced Pluripotent Stem Cells/pathology , Myeloid Cells/pathology , Oncogene Proteins, Fusion/metabolism , Precursor Cell Lymphoblastic Leukemia-Lymphoma/pathology , Acute Disease , B-Lymphocytes/metabolism , Core Binding Factor Alpha 2 Subunit/genetics , Female , Humans , Induced Pluripotent Stem Cells/metabolism , Models, Biological , Myeloid Cells/metabolism , Oncogene Proteins, Fusion/genetics , Precursor Cell Lymphoblastic Leukemia-Lymphoma/metabolism , Pregnancy , Pregnancy Trimester, First , Receptors, Interleukin-7 , Transcriptome
5.
Proc Natl Acad Sci U S A ; 114(36): E7526-E7535, 2017 09 05.
Article in English | MEDLINE | ID: mdl-28827334

ABSTRACT

The human genome contains ∼30,000 CpG islands (CGIs). While CGIs associated with promoters nearly always remain unmethylated, many of the ∼9,000 CGIs lying within gene bodies become methylated during development and differentiation. Both promoter and intragenic CGIs may also become abnormally methylated as a result of genome rearrangements and in malignancy. The epigenetic mechanisms by which some CGIs become methylated but others, in the same cell, remain unmethylated in these situations are poorly understood. Analyzing specific loci and using a genome-wide analysis, we show that transcription running across CGIs, associated with specific chromatin modifications, is required for DNA methyltransferase 3B (DNMT3B)-mediated DNA methylation of many naturally occurring intragenic CGIs. Importantly, we also show that a subgroup of intragenic CGIs is not sensitive to this process of transcription-mediated methylation and that this correlates with their individual intrinsic capacity to initiate transcription in vivo. We propose a general model of how transcription could act as a primary determinant of the patterns of CGI methylation in normal development and differentiation, and in human disease.


Subject(s)
Cell Differentiation/genetics , CpG Islands/genetics , DNA Methylation/genetics , Transcription, Genetic/genetics , Animals , Cell Line , Epigenesis, Genetic/genetics , Genome, Human/genetics , Humans , Mice , Promoter Regions, Genetic/genetics , Sequence Analysis, DNA/methods
6.
Nat Genet ; 48(8): 895-903, 2016 08.
Article in English | MEDLINE | ID: mdl-27376235

ABSTRACT

Many genes determining cell identity are regulated by clusters of Mediator-bound enhancer elements collectively referred to as super-enhancers. These super-enhancers have been proposed to manifest higher-order properties important in development and disease. Here we report a comprehensive functional dissection of one of the strongest putative super-enhancers in erythroid cells. By generating a series of mouse models, deleting each of the five regulatory elements of the α-globin super-enhancer individually and in informative combinations, we demonstrate that each constituent enhancer seems to act independently and in an additive fashion with respect to hematological phenotype, gene expression, chromatin structure and chromosome conformation, without clear evidence of synergistic or higher-order effects. Our study highlights the importance of functional genetic analyses for the identification of new concepts in transcriptional regulation.


Subject(s)
Enhancer Elements, Genetic/genetics , Erythroid Cells/metabolism , Gene Expression Regulation , Transcription Factors/metabolism , Transcription, Genetic/genetics , alpha-Globins/genetics , Animals , Chromatin/genetics , Embryo, Mammalian/cytology , Embryo, Mammalian/metabolism , Mice , Mice, Knockout
7.
BMC Dev Biol ; 15: 35, 2015 Oct 09.
Article in English | MEDLINE | ID: mdl-26453549

ABSTRACT

BACKGROUND: Pluripotent cells are present in early embryos until the levels of the pluripotency regulator Oct4 drop at the beginning of somitogenesis. Elevating Oct4 levels in explanted post-pluripotent cells in vitro restores their pluripotency. Cultured pluripotent cells can participate in normal development when introduced into host embryos up to the end of gastrulation. In contrast, pluripotent cells efficiently seed malignant teratocarcinomas in adult animals. In humans, extragonadal teratomas and teratocarcinomas are most frequently found in the sacrococcygeal region of neonates, suggesting that these tumours originate from cells in the posterior of the embryo that either reactivate or fail to switch off their pluripotent status. However, experimental models for the persistence or reactivation of pluripotency during embryonic development are lacking. METHODS: We manually injected embryonic stem cells into conceptuses at E9.5 to test whether the presence of pluripotent cells at this stage correlates with teratocarcinoma formation. We then examined the effects of reactivating embryonic Oct4 expression ubiquitously or in combination with Nanog within the primitive streak (PS)/tail bud (TB) using a transgenic mouse line and embryo chimeras carrying a PS/TB-specific heterologous gene expression cassette respectively. RESULTS: Here, we show that pluripotent cells seed teratomas in post-gastrulation embryos. However, at these stages, induced ubiquitous expression of Oct4 does not lead to restoration of pluripotency (indicated by Nanog expression) and tumour formation in utero, but instead causes a severe phenotype in the extending anteroposterior axis. Use of a more restricted T(Bra) promoter transgenic system enabling inducible ectopic expression of Oct4 and Nanog specifically in the posteriorly-located primitive streak (PS) and tail bud (TB) led to similar axial malformations to those induced by Oct4 alone. These cells underwent induction of pluripotency marker expression in Epiblast Stem Cell (EpiSC) explants derived from somitogenesis-stage embryos, but no teratocarcinoma formation was observed in vivo. CONCLUSIONS: Our findings show that although pluripotent cells with teratocarcinogenic potential can be produced in vitro by the overexpression of pluripotency regulators in explanted somitogenesis-stage somatic cells, the in vivo induction of these genes does not yield tumours. This suggests a restrictive regulatory role of the embryonic microenvironment in the induction of pluripotency.


Subject(s)
Embryo, Mammalian/metabolism , Embryonic Stem Cells/metabolism , Teratoma/metabolism , Teratoma/pathology , Animals , Embryo, Mammalian/pathology , Fetal Proteins/metabolism , Homeodomain Proteins/genetics , Humans , Mice , Nanog Homeobox Protein , Octamer Transcription Factor-3/genetics , Octamer Transcription Factor-3/metabolism , Pluripotent Stem Cells/metabolism , T-Box Domain Proteins/metabolism , Tail/embryology
8.
Stem Cells ; 31(8): 1511-22, 2013 Aug.
Article in English | MEDLINE | ID: mdl-23649667

ABSTRACT

Robust development of the early embryo may benefit from mechanisms that ensure that not all pluripotent cells differentiate at exactly the same time: such mechanisms would build flexibility into the process of lineage allocation. This idea is supported by the observation that pluripotent stem cells differentiate at different rates in vitro. We use a clonal commitment assay to confirm that pluripotent cells commit to differentiate asynchronously even under uniform differentiation conditions. Stochastic variability in expression of the Notch target gene Hes1 has previously been reported to influence neural versus mesodermal differentiation through modulation of Notch activity. Here we report that Hes1 also has an earlier role to delay exit from the pluripotent state into all lineages. The early function of Hes1 to delay differentiation can be explained by an ability of Hes1 to amplify STAT3 responsiveness in a cell-autonomous manner. Variability in Hes1 expression therefore helps to explain why STAT3 responsiveness varies between individual ES cells, and this in turn helps to explain why pluripotent cells commit to differentiate asynchronously.


Subject(s)
Basic Helix-Loop-Helix Transcription Factors/metabolism , Homeodomain Proteins/metabolism , Pluripotent Stem Cells/cytology , Pluripotent Stem Cells/metabolism , Receptors, Notch/metabolism , STAT3 Transcription Factor/metabolism , Animals , Cell Differentiation/physiology , Down-Regulation , Humans , Mice , Nanog Homeobox Protein , Signal Transduction , Transcription Factor HES-1 , Transfection
9.
Cell Rep ; 3(2): 472-84, 2013 Feb 21.
Article in English | MEDLINE | ID: mdl-23395635

ABSTRACT

The events that prime pluripotent cells for differentiation are not well understood. Inhibitor of DNA binding/differentiation (Id) proteins, which are inhibitors of basic helix-loop-helix (bHLH) transcription factor activity, contribute to pluripotency by blocking sequential transitions toward differentiation. Using yeast-two-hybrid screens, we have identified Id-regulated transcription factors that are expressed in embryonic stem cells (ESCs). One of these, Tcf15, is also expressed in the embryonic day 4.5 embryo and is specifically associated with a novel subpopulation of primed ESCs. An Id-resistant form of Tcf15 rapidly downregulates Nanog and accelerates somatic lineage commitment. We propose that because Tcf15 can be held in an inactive state through Id activity, it may prime pluripotent cells for entry to somatic lineages upon downregulation of Id. We also find that Tcf15 expression is dependent on fibroblast growth factor (FGF) signaling, providing an explanation for how FGF can prime for differentiation without driving cells out of the pluripotent state.


Subject(s)
Basic Helix-Loop-Helix Transcription Factors/metabolism , Embryonic Stem Cells/metabolism , Animals , Cell Differentiation , Cells, Cultured , Down-Regulation , Embryo, Mammalian/metabolism , Embryonic Stem Cells/cytology , Endoderm/cytology , Endoderm/metabolism , Fibroblast Growth Factors/metabolism , Gene Expression Regulation, Developmental , Homeodomain Proteins/metabolism , Mice , Nanog Homeobox Protein , Otx Transcription Factors/metabolism , Signal Transduction
10.
Nucleic Acids Res ; 40(19): e150, 2012 Oct.
Article in English | MEDLINE | ID: mdl-22753106

ABSTRACT

Transgenesis is a cornerstone of molecular biology. The ability to integrate a specifically engineered piece of DNA into the genome of a living system is fundamental to our efforts to understand life and exploit its implications for medicine, nanotechnology and bioprospecting. However, transgenesis has been hampered by position effects and multi-copy integration problems, which are mainly due to the use of small, plasmid-based transgenes. Large transgenes based on native genomic regions cloned into bacterial artificial chromosomes (BACs) circumvent these problems but are prone to fragmentation. Herein, we report that contrary to widely held notions, large BAC-sized constructs do not prohibit transposition. We also report the first reliable method for BAC transgenesis in human embryonic stem cells (hESCs). The PiggyBac or Sleeping Beauty transposon inverted repeats were integrated into BAC vectors by recombineering, followed by co-lipofection with the corresponding transposase in hESCs to generate robust fluorescent protein reporter lines for OCT4, NANOG, GATA4 and PAX6. BAC transposition delivers several advantages, including increased frequencies of single-copy, full-length integration, which will be useful in all transgenic systems but especially in difficult venues like hESCs.


Subject(s)
Chromosomes, Artificial, Bacterial , DNA Transposable Elements , Gene Transfer Techniques , Transgenes , Animals , Cell Line , Embryonic Stem Cells , Gene Dosage , Genes, Reporter , Homeodomain Proteins/genetics , Humans , Luminescent Proteins/genetics , Mice , Nanog Homeobox Protein , Octamer Transcription Factor-3/genetics
11.
EMBO J ; 31(2): 317-29, 2012 Jan 18.
Article in English | MEDLINE | ID: mdl-22056776

ABSTRACT

The role of DNA sequence in determining chromatin state is incompletely understood. We have previously demonstrated that large chromosomal segments from human cells recapitulate their native chromatin state in mouse cells, but the relative contribution of local sequences versus their genomic context remains unknown. In this study, we compare orthologous chromosomal regions for which the human locus establishes prominent sites of Polycomb complex recruitment in pluripotent stem cells, whereas the corresponding mouse locus does not. Using recombination-mediated cassette exchange at the mouse locus, we establish the primacy of local sequences in the encoding of chromatin state. We show that the signal for chromatin bivalency is redundantly encoded across a bivalent domain and that this reflects competition between Polycomb complex recruitment and transcriptional activation. Furthermore, our results suggest that a high density of unmethylated CpG dinucleotides is sufficient for vertebrate Polycomb recruitment. This model is supported by analysis of DNA methyltransferase-deficient embryonic stem cells.


Subject(s)
CpG Islands/physiology , Gene Expression Regulation/genetics , Repressor Proteins/metabolism , alpha-Globins/genetics , Animals , Cells, Cultured/metabolism , Chromatin/genetics , Chromosome Mapping , Chromosomes, Human, Pair 16 , DNA Methylation , DNA, Recombinant/genetics , Embryonic Stem Cells/metabolism , Humans , Mice , Mice, Transgenic , Pluripotent Stem Cells/metabolism , Polycomb-Group Proteins , Recombination, Genetic , Regulatory Sequences, Nucleic Acid , Species Specificity , Transcription, Genetic
12.
Nat Rev Genet ; 13(1): 14-20, 2011 Dec 16.
Article in English | MEDLINE | ID: mdl-22179716

ABSTRACT

Mouse models have become an invaluable tool for understanding human health and disease owing to our ability to manipulate the mouse genome exquisitely. Recent progress in genomic analysis has led to an increase in the number and type of disease-causing mutations detected and has also highlighted the importance of non-coding regions. As a result, there is increasing interest in creating 'genomically' humanized mouse models, in which entire human genomic loci are transferred into the mouse genome. The technical challenges towards achieving this aim are large but are starting to be tackled with success.


Subject(s)
Chromosomes, Artificial, Mammalian/genetics , Gene Targeting/methods , Gene Transfer Techniques , Mice, Transgenic/genetics , Transgenes/genetics , Animals , Disease Models, Animal , Genetic Diseases, Inborn/genetics , Genome-Wide Association Study/methods , Humans , Mice , Promoter Regions, Genetic , Recombination, Genetic , Regulatory Elements, Transcriptional
13.
PLoS Genet ; 7(11): e1002348, 2011 Nov.
Article in English | MEDLINE | ID: mdl-22072979

ABSTRACT

The forkhead transcription factor Foxn1 is indispensable for thymus development, but the mechanisms by which it mediates thymic epithelial cell (TEC) development are poorly understood. To examine the cellular and molecular basis of Foxn1 function, we generated a novel and revertible hypomorphic allele of Foxn1. By varying levels of its expression, we identified a number of features of the Foxn1 system. Here we show that Foxn1 is a powerful regulator of TEC differentiation that is required at multiple intermediate stages of TE lineage development in the fetal and adult thymus. We find no evidence for a role for Foxn1 in TEC fate-choice. Rather, we show it is required for stable entry into both the cortical and medullary TEC differentiation programmes and subsequently is needed at increasing dosage for progression through successive differentiation states in both cortical and medullary TEC. We further demonstrate regulation by Foxn1 of a suite of genes with diverse roles in thymus development and/or function, suggesting it acts as a master regulator of the core thymic epithelial programme rather than regulating a particular aspect of TEC biology. Overall, our data establish a genetics-based model of cellular hierarchies in the TE lineage and provide mechanistic insight relating titration of a single transcription factor to control of lineage progression. Our novel revertible hypomorph system may be similarly applied to analyzing other regulators of development.


Subject(s)
Adrenal Medulla/cytology , Cell Differentiation/genetics , Cell Lineage/genetics , Embryonic Development/genetics , Forkhead Transcription Factors/metabolism , Thymus Gland/growth & development , Adrenal Medulla/metabolism , Alleles , Animals , Epithelial Cells/metabolism , Epithelial Cells/physiology , Forkhead Transcription Factors/genetics , Gene Expression Regulation, Developmental , Integrases/chemistry , Integrases/genetics , Mice , Mice, Inbred C57BL , Mice, Transgenic , Tamoxifen/chemistry
14.
Genes Dev ; 25(15): 1583-8, 2011 Aug 01.
Article in English | MEDLINE | ID: mdl-21828268

ABSTRACT

Remote distal enhancers may be located tens or thousands of kilobases away from their promoters. How they control gene expression is still poorly understood. Here, we analyze the influence of a remote enhancer on the balance between repression (Polycomb-PcG) and activation (Trithorax-TrxG) of a developmentally regulated gene associated with a CpG island. We reveal its essential, nonredundant role in clearing the PcG complex and H3K27me3 from the CpG island. In the absence of the enhancer, the H3K27me3 demethylase (JMJD3) is not recruited to the CpG island. We propose a new role of long-range regulatory elements in removing repressive PcG complexes.


Subject(s)
Enhancer Elements, Genetic/physiology , Repressor Proteins/metabolism , Animals , Cell Line , Cells, Cultured , Chromatin/metabolism , Chromatin Immunoprecipitation , CpG Islands , Erythropoiesis/physiology , Gene Expression Regulation , Humans , Polycomb-Group Proteins , Protein Binding , Protein Methyltransferases/metabolism
15.
Cell Metab ; 10(5): 343-54, 2009 Nov.
Article in English | MEDLINE | ID: mdl-19883613

ABSTRACT

PI3K signaling is thought to mediate leptin and insulin action in hypothalamic pro-opiomelanocortin (POMC) and agouti-related protein (AgRP) neurons, key regulators of energy homeostasis, through largely unknown mechanisms. We inactivated either p110alpha or p110beta PI3K catalytic subunits in these neurons and demonstrate a dominant role for the latter in energy homeostasis regulation. In POMC neurons, p110beta inactivation prevented insulin- and leptin-stimulated electrophysiological responses. POMCp110beta null mice exhibited central leptin resistance, increased adiposity, and diet-induced obesity. In contrast, the response to leptin was not blocked in p110alpha-deficient POMC neurons. Accordingly, POMCp110alpha null mice displayed minimal energy homeostasis abnormalities. Similarly, in AgRP neurons, p110beta had a more important role than p110alpha. AgRPp110alpha null mice displayed normal energy homeostasis regulation, whereas AgRPp110beta null mice were lean, with increased leptin sensitivity and resistance to diet-induced obesity. These results demonstrate distinct metabolic roles for the p110alpha and p110beta isoforms of PI3K in hypothalamic energy regulation.


Subject(s)
Agouti-Related Protein/metabolism , Energy Metabolism/physiology , Isoenzymes/metabolism , Phosphatidylinositol 3-Kinases/metabolism , Pro-Opiomelanocortin/metabolism , Adiposity/genetics , Animals , Class I Phosphatidylinositol 3-Kinases , Diet , Electrophysiological Phenomena , Hypothalamus/metabolism , Insulin/metabolism , Isoenzymes/antagonists & inhibitors , Isoenzymes/genetics , Leptin/metabolism , Mice , Mice, Knockout , Neuroendocrine Cells/enzymology , Obesity/genetics , Obesity/metabolism , Phosphatidylinositol 3-Kinases/genetics , Phosphoinositide-3 Kinase Inhibitors , Signal Transduction
16.
Blood ; 114(19): 4253-60, 2009 Nov 05.
Article in English | MEDLINE | ID: mdl-19696202

ABSTRACT

Previous studies in the mouse have shown that high levels of alpha-globin gene expression in late erythropoiesis depend on long-range, physical interactions between remote upstream regulatory elements and the globin promoters. Using quantitative chromosome conformation capture (q3C), we have now analyzed all interactions between 4 such elements lying 10 to 50 kb upstream of the human alpha cluster and their interactions with the alpha-globin promoter. All of these elements interact with the alpha-globin gene in an erythroid-specific manner. These results were confirmed in a mouse model of human alpha globin expression in which the human cluster replaces the mouse cluster in situ (humanized mouse). We have also shown that expression and all of the long-range interactions depend largely on just one of these elements; removal of the previously characterized major regulatory element (called HS -40) results in loss of all the interactions and alpha-globin expression. Reinsertion of this element at an ectopic location restores both expression and the intralocus interactions. In contrast to other more complex systems involving multiple upstream elements and promoters, analysis of the human alpha-globin cluster during erythropoiesis provides a simple and tractable model to understand the mechanisms underlying long-range gene regulation.


Subject(s)
Chromosomes, Human/genetics , alpha-Globins/genetics , Animals , Base Sequence , Cell Line , Cells, Cultured , DNA Probes/genetics , Erythropoiesis/genetics , Female , Gene Regulatory Networks , Humans , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Multigene Family , Promoter Regions, Genetic , Regulatory Elements, Transcriptional
17.
J Cell Biol ; 182(6): 1083-97, 2008 Sep 22.
Article in English | MEDLINE | ID: mdl-18809724

ABSTRACT

Genes on different chromosomes can be spatially associated in the nucleus in several transcriptional and regulatory situations; however, the functional significance of such associations remains unclear. Using human erythropoiesis as a model, we show that five cotranscribed genes, which are found on four different chromosomes, associate with each other at significant but variable frequencies. Those genes most frequently in association lie in decondensed stretches of chromatin. By replacing the mouse alpha-globin gene cluster in situ with its human counterpart, we demonstrate a direct effect of the regional chromatin environment on the frequency of association, whereas nascent transcription from the human alpha-globin gene appears unaffected. We see no evidence that cotranscribed erythroid genes associate at shared transcription foci, but we do see stochastic clustering of active genes around common nuclear SC35-enriched speckles (hence the apparent nonrandom association between genes). Thus, association between active genes may result from their location on decondensed chromatin that enables clustering around common nuclear speckles.


Subject(s)
Blood Cells/physiology , Chromatin/metabolism , Chromosomes/metabolism , Erythropoiesis/genetics , Intranuclear Inclusion Bodies/metabolism , Transcription, Genetic , Animals , Anion Exchange Protein 1, Erythrocyte/genetics , Anion Exchange Protein 1, Erythrocyte/metabolism , Cell Nucleus/metabolism , Gene Expression Regulation , Globins/genetics , Humans , In Situ Hybridization, Fluorescence , Mice , Multigene Family
18.
Proc Natl Acad Sci U S A ; 105(24): 8292-7, 2008 Jun 17.
Article in English | MEDLINE | ID: mdl-18544649

ABSTRACT

The p110 isoforms of phosphoinositide 3-kinase (PI3K) are acutely regulated by extracellular stimuli. The class IA PI3K catalytic subunits (p110alpha, p110beta, and p110delta) occur in complex with a Src homology 2 (SH2) domain-containing p85 regulatory subunit, which has been shown to link p110alpha and p110delta to Tyr kinase signaling pathways. The p84/p101 regulatory subunits of the p110gamma class IB PI3K lack SH2 domains and instead couple p110gamma to G protein-coupled receptors (GPCRs). Here, we show, using small-molecule inhibitors with selectivity for p110beta and cells derived from a p110beta-deficient mouse line, that p110beta is not a major effector of Tyr kinase signaling but couples to GPCRs. In macrophages, both p110beta and p110gamma contributed to Akt activation induced by the GPCR agonist complement 5a, but not by the Tyr kinase ligand colony-stimulating factor-1. In fibroblasts, which express p110beta but not p110gamma, p110beta mediated Akt activation by the GPCR ligands stromal cell-derived factor, sphingosine-1-phosphate, and lysophosphatidic acid but not by the Tyr kinase ligands PDGF, insulin, and insulin-like growth factor 1. Introduction of p110gamma in these cells reduced the contribution of p110beta to GPCR signaling. Taken together, these data show that p110beta and p110gamma can couple redundantly to the same GPCR agonists. p110beta, which shows a much broader tissue distribution than the leukocyte-restricted p110gamma, could thus provide a conduit for GPCR-linked PI3K signaling in the many cell types where p110gamma expression is low or absent.


Subject(s)
Phosphatidylinositol 3-Kinases/metabolism , Receptors, G-Protein-Coupled/metabolism , Animals , Class I Phosphatidylinositol 3-Kinases , Complement C5a/pharmacology , Fibroblasts/enzymology , Genetic Complementation Test , Isoenzymes/genetics , Isoenzymes/metabolism , Ligands , Macrophage Colony-Stimulating Factor/pharmacology , Macrophages/enzymology , Mice , Mice, Mutant Strains , Phosphatidylinositol 3-Kinases/genetics , Proto-Oncogene Proteins c-akt/metabolism , Receptors, G-Protein-Coupled/agonists , Signal Transduction
19.
Nature ; 453(7195): 662-6, 2008 May 29.
Article in English | MEDLINE | ID: mdl-18449193

ABSTRACT

Phosphoinositide 3-kinases (PI3Ks) signal downstream of multiple cell-surface receptor types. Class IA PI3K isoforms couple to tyrosine kinases and consist of a p110 catalytic subunit (p110alpha, p110beta or p110delta), constitutively bound to one of five distinct p85 regulatory subunits. PI3Ks have been implicated in angiogenesis, but little is known about potential selectivity among the PI3K isoforms and their mechanism of action in endothelial cells during angiogenesis in vivo. Here we show that only p110alpha activity is essential for vascular development. Ubiquitous or endothelial cell-specific inactivation of p110alpha led to embryonic lethality at mid-gestation because of severe defects in angiogenic sprouting and vascular remodelling. p110alpha exerts this critical endothelial cell-autonomous function by regulating endothelial cell migration through the small GTPase RhoA. p110alpha activity is particularly high in endothelial cells and preferentially induced by tyrosine kinase ligands (such as vascular endothelial growth factor (VEGF)-A). In contrast, p110beta in endothelial cells signals downstream of G-protein-coupled receptor (GPCR) ligands such as SDF-1alpha, whereas p110delta is expressed at low level and contributes only minimally to PI3K activity in endothelial cells. These results provide the first in vivo evidence for p110-isoform selectivity in endothelial PI3K signalling during angiogenesis.


Subject(s)
Cell Movement , Endothelial Cells/cytology , Endothelial Cells/enzymology , Neovascularization, Physiologic , Phosphatidylinositol 3-Kinases/metabolism , Animals , Cells, Cultured , Class I Phosphatidylinositol 3-Kinases , Female , Humans , Mice , Phosphatidylinositol 3-Kinases/genetics , RNA Interference , Rats , Signal Transduction/drug effects , Vascular Endothelial Growth Factor A/pharmacology , Wounds and Injuries , rho GTP-Binding Proteins/metabolism , rhoA GTP-Binding Protein
20.
PLoS One ; 2(9): e869, 2007 Sep 11.
Article in English | MEDLINE | ID: mdl-17846664

ABSTRACT

The expression and function of the 8 distinct catalytic isoforms of PI 3-kinase (PI3K) in the nervous system are unknown. Whereas most PI3Ks have a broad tissue distribution, the tyrosine kinase-linked p110delta isoform has previously been shown to be enriched in leukocytes. Here we report that p110delta is also highly expressed in the nervous system. Inactivation of p110delta in mice did not affect gross neuronal development but led to an increased vulnerability of dorsal root ganglia neurons to exhibit growth cone collapse and decreases in axonal extension. Loss of p110delta activity also dampened axonal regeneration following peripheral nerve injury in adult mice and impaired functional recovery of locomotion. p110delta inactivation resulted in reduced neuronal signaling through the Akt protein kinase, and increased activity of the small GTPase RhoA. Pharmacological inhibition of ROCK, a downstream effector of RhoA, restored axonal extension defects in neurons with inactive p110delta, suggesting a key role of RhoA in p110delta signaling in neurons. Our data identify p110delta as an important signaling component for efficient axonal elongation in the developing and regenerating nervous system.


Subject(s)
Axons/physiology , Neurons, Afferent/metabolism , Phosphatidylinositol 3-Kinases/metabolism , Animals , Axons/drug effects , Blotting, Western , Cells, Cultured , Chromones/pharmacology , Class I Phosphatidylinositol 3-Kinases , Electrophoresis, Polyacrylamide Gel , Immunohistochemistry , Male , Mice , Mice, Knockout , Morpholines/pharmacology , Nerve Regeneration , Neurons, Afferent/cytology , Neurons, Afferent/drug effects , PTEN Phosphohydrolase/metabolism , Phosphatidylinositol 3-Kinases/genetics , Phosphoinositide-3 Kinase Inhibitors , Proto-Oncogene Proteins c-akt/metabolism , Sciatic Nerve/injuries , Sciatic Nerve/metabolism , Sciatic Nerve/physiopathology , rhoA GTP-Binding Protein/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...