Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 16 de 16
Filter
1.
Development ; 149(22)2022 11 15.
Article in English | MEDLINE | ID: mdl-36355065

ABSTRACT

Female mouse embryonic stem cells (mESCs) present differently from male mESCs in several fundamental ways; however, complications with their in vitro culture have resulted in an under-representation of female mESCs in the literature. Recent studies show that the second X chromosome in female, and more specifically the transcriptional activity from both of these chromosomes due to absent X chromosome inactivation, sets female and male mESCs apart. To avoid this undesirable state, female mESCs in culture preferentially adopt an XO karyotype, with this adaption leading to loss of their unique properties in favour of a state that is near indistinguishable from male mESCs. If female pluripotency is to be studied effectively in this system, it is crucial that high-quality cultures of XX mESCs are available. Here, we report a method for better maintaining XX female mESCs in culture that also stabilises the male karyotype and makes study of female-specific pluripotency more feasible.


Subject(s)
Mouse Embryonic Stem Cells , X Chromosome Inactivation , Male , Animals , Female , Mice , Cell Differentiation/physiology , X Chromosome Inactivation/genetics , Karyotype
2.
Nucleic Acids Res ; 47(8): e46, 2019 05 07.
Article in English | MEDLINE | ID: mdl-30793194

ABSTRACT

Systematic variation in the methylation of cytosines at CpG sites plays a critical role in early development of humans and other mammals. Of particular interest are regions of differential methylation between parental alleles, as these often dictate monoallelic gene expression, resulting in parent of origin specific control of the embryonic transcriptome and subsequent development, in a phenomenon known as genomic imprinting. Using long-read nanopore sequencing we show that, with an average genomic coverage of ∼10, it is possible to determine both the level of methylation of CpG sites and the haplotype from which each read arises. The long-read property is exploited to characterize, using novel methods, both methylation and haplotype for reads that have reduced basecalling precision compared to Sanger sequencing. We validate the analysis both through comparison of nanopore-derived methylation patterns with those from Reduced Representation Bisulfite Sequencing data and through comparison with previously reported data. Our analysis successfully identifies known imprinting control regions (ICRs) as well as some novel differentially methylated regions which, due to their proximity to hitherto unknown monoallelically expressed genes, may represent new ICRs.


Subject(s)
Genome , Genomic Imprinting , Genotyping Techniques , Haplotypes , Sequence Analysis, DNA/statistics & numerical data , Alleles , Animals , Chromosome Mapping , CpG Islands , DNA Methylation , Embryo, Mammalian/chemistry , Embryo, Mammalian/metabolism , Female , High-Throughput Nucleotide Sequencing , Male , Mice , Placenta/chemistry , Placenta/metabolism , Pregnancy
3.
Breast Cancer Res ; 17: 85, 2015 Jun 18.
Article in English | MEDLINE | ID: mdl-26080807

ABSTRACT

INTRODUCTION: MicroRNAs (miRNAs) have been implicated in governing lineage specification and differentiation in multiple organs; however, little is known about their specific roles in mammopoiesis. We have determined the global miRNA expression profiles of functionally distinct epithelial subpopulations in mouse and human mammary tissue, and compared these to their cognate transcriptomes and epigenomes. Finally, the human miRNA signatures were used to interrogate the different subtypes of breast cancer, with a view to determining miRNA networks deregulated during oncogenesis. METHODS: RNA from sorted mouse and human mammary cell subpopulations was subjected to miRNA expression analysis using the TaqMan MicroRNA Array. Differentially expressed (DE) miRNAs were correlated with gene expression and histone methylation profiles. Analysis of miRNA signatures of the intrinsic subtypes of breast cancer in The Cancer Genome Atlas (TCGA) database versus those of normal human epithelial subpopulations was performed. RESULTS: Unique miRNA signatures characterized each subset (mammary stem cell (MaSC)/basal, luminal progenitor, mature luminal, stromal), with a high degree of conservation across species. Comparison of miRNA and transcriptome profiles for the epithelial subtypes revealed an inverse relationship and pinpointed key developmental genes. Interestingly, expression of the primate-specific miRNA cluster (19q13.4) was found to be restricted to the MaSC/basal subset. Comparative analysis of miRNA signatures with H3 lysine modification maps of the different epithelial subsets revealed a tight correlation between active or repressive marks for the top DE miRNAs, including derepression of miRNAs in Ezh2-deficient cellular subsets. Interrogation of TCGA-identified miRNA profiles with the miRNA signatures of different human subsets revealed specific relationships. CONCLUSIONS: The derivation of global miRNA expression profiles for the different mammary subpopulations provides a comprehensive resource for understanding the interplay between miRNA networks and target gene expression. These data have highlighted lineage-specific miRNAs and potential miRNA-mRNA networks, some of which are disrupted in neoplasia. Furthermore, our findings suggest that key developmental miRNAs are regulated by global changes in histone modification, thus linking the mammary epigenome with genome-wide changes in the expression of genes and miRNAs. Comparative miRNA signature analyses between normal breast epithelial cells and breast tumors confirmed an important linkage between luminal progenitor cells and basal-like tumors.


Subject(s)
Breast/metabolism , Epigenesis, Genetic , Epithelial Cells/metabolism , Gene Expression Regulation , MicroRNAs/genetics , Transcriptome , Animals , Breast Neoplasms/genetics , Breast Neoplasms/pathology , Cell Lineage/genetics , Cluster Analysis , Female , Gene Expression Profiling , Genetic Loci , Humans , Mammary Glands, Animal/metabolism , Mice , RNA Interference , RNA, Messenger/genetics
4.
BMC Cancer ; 15: 221, 2015 Apr 03.
Article in English | MEDLINE | ID: mdl-25879659

ABSTRACT

BACKGROUND: The molecular regulators that orchestrate stem cell renewal, proliferation and differentiation along the mammary epithelial hierarchy remain poorly understood. Here we have performed a large-scale pooled RNAi screen in primary mouse mammary stem cell (MaSC)-enriched basal cells using 1295 shRNAs against genes principally involved in transcriptional regulation. METHODS: MaSC-enriched basal cells transduced with lentivirus pools carrying shRNAs were maintained as non-adherent mammospheres, a system known to support stem and progenitor cells. Integrated shRNAs that altered culture kinetics were identified by next generation sequencing as relative frequency changes over time. RNA-seq-based expression profiling coupled with in vitro progenitor and in vivo transplantation assays was used to confirm a role for candidate genes in mammary stem and/or progenitor cells. RESULTS: Utilizing a mammosphere-based assay, the screen identified several candidate regulators. Although some genes had been previously implicated in mammary gland development, the vast majority of genes uncovered have no known function within the mammary gland. RNA-seq analysis of freshly purified primary mammary epithelial populations and short-term cultured mammospheres was used to confirm the expression of candidate regulators. Two genes, Asap1 and Prox1, respectively implicated in breast cancer metastasis and progenitor cell function in other systems, were selected for further analysis as their roles in the normal mammary gland were unknown. Both Prox1 and Asap1 were shown to act as negative regulators of progenitor activity in vitro, and Asap1 knock-down led to a marked increase in repopulating activity in vivo, implying a role in stem cell activity. CONCLUSIONS: This study has revealed a number of novel genes that influence the activity or survival of mammary stem and/or progenitor cells. Amongst these, we demonstrate that Prox1 and Asap1 behave as negative regulators of mammary stem/progenitor function. Both of these genes have also been implicated in oncogenesis. Our findings provide proof of principle for the use of short-term cultured primary MaSC/basal cells in functional RNAi screens.


Subject(s)
Adaptor Proteins, Signal Transducing/genetics , Homeodomain Proteins/genetics , Mammary Glands, Animal/metabolism , RNA, Small Interfering/genetics , Stem Cells/metabolism , Tumor Suppressor Proteins/genetics , Animals , Cell Count , Cell Differentiation/genetics , Epithelial Cells/metabolism , Female , Flow Cytometry , Gene Expression Profiling , Gene Expression Regulation , High-Throughput Nucleotide Sequencing , Immunophenotyping , Mice , Reproducibility of Results
5.
Nat Commun ; 14(1): 5466, 2023 09 25.
Article in English | MEDLINE | ID: mdl-37749075

ABSTRACT

The interplay between 3D chromatin architecture and gene silencing is incompletely understood. Here, we report a novel point mutation in the non-canonical SMC protein SMCHD1 that enhances its silencing capacity at endogenous developmental targets. Moreover, it also results in enhanced silencing at the facioscapulohumeral muscular dystrophy associated macrosatellite-array, D4Z4, resulting in enhanced repression of DUX4 encoded by this repeat. Heightened SMCHD1 silencing perturbs developmental Hox gene activation, causing a homeotic transformation in mice. Paradoxically, the mutant SMCHD1 appears to enhance insulation against other epigenetic regulators, including PRC2 and CTCF, while depleting long range chromatin interactions akin to what is observed in the absence of SMCHD1. These data suggest that SMCHD1's role in long range chromatin interactions is not directly linked to gene silencing or insulating the chromatin, refining the model for how the different levels of SMCHD1-mediated chromatin regulation interact to bring about gene silencing in normal development and disease.


Subject(s)
Chromatin , Chromosomal Proteins, Non-Histone , Muscular Dystrophy, Facioscapulohumeral , Animals , Mice , Chromatin/genetics , Epigenomics , Gene Silencing , Genes, Homeobox , Muscular Dystrophy, Facioscapulohumeral/genetics , Chromosomal Proteins, Non-Histone/genetics
6.
Nat Commun ; 13(1): 4295, 2022 07 25.
Article in English | MEDLINE | ID: mdl-35879318

ABSTRACT

Parents transmit genetic and epigenetic information to their offspring. Maternal effect genes regulate the offspring epigenome to ensure normal development. Here we report that the epigenetic regulator SMCHD1 has a maternal effect on Hox gene expression and skeletal patterning. Maternal SMCHD1, present in the oocyte and preimplantation embryo, prevents precocious activation of Hox genes post-implantation. Without maternal SMCHD1, highly penetrant posterior homeotic transformations occur in the embryo. Hox genes are decorated with Polycomb marks H2AK119ub and H3K27me3 from the oocyte throughout early embryonic development; however, loss of maternal SMCHD1 does not deplete these marks. Therefore, we propose maternal SMCHD1 acts downstream of Polycomb marks to establish a chromatin state necessary for persistent epigenetic silencing and appropriate Hox gene expression later in the developing embryo. This is a striking role for maternal SMCHD1 in long-lived epigenetic effects impacting offspring phenotype.


Subject(s)
Gene Expression Regulation, Developmental , Genes, Homeobox , Animals , Chromatin/genetics , Chromosomal Proteins, Non-Histone/genetics , Chromosomal Proteins, Non-Histone/metabolism , Embryo, Mammalian/metabolism , Female , Gene Expression , Mice , Polycomb-Group Proteins/genetics , Polycomb-Group Proteins/metabolism , Pregnancy
7.
iScience ; 25(7): 104684, 2022 Jul 15.
Article in English | MEDLINE | ID: mdl-35856023

ABSTRACT

SMCHD1 (structural maintenance of chromosomes hinge domain containing 1) is a noncanonical SMC protein that mediates long-range repressive chromatin structures. SMCHD1 is required for X chromosome inactivation in female cells and repression of imprinted and clustered autosomal genes, with SMCHD1 mutations linked to human diseases facioscapulohumeral muscular dystrophy (FSHD) and bosma arhinia and micropthalmia syndrome (BAMS). We used a conditional mouse model to investigate SMCHD1 in hematopoiesis. Smchd1-deleted mice maintained steady-state hematopoiesis despite showing an impaired reconstitution capacity in competitive bone marrow transplantations and age-related hematopoietic stem cell (HSC) loss. This phenotype was more pronounced in Smchd1-deleted females, which showed a loss of quiescent HSCs and fewer B cells. Gene expression profiling of Smchd1-deficient HSCs and B cells revealed known and cell-type-specific SMCHD1-sensitive genes and significant disruption to X-linked gene expression in female cells. These data show SMCHD1 is a regulator of HSCs whose effects are more profound in females.

8.
Epigenetics Chromatin ; 15(1): 26, 2022 07 18.
Article in English | MEDLINE | ID: mdl-35843975

ABSTRACT

Embryonic development is dependent on the maternal supply of proteins through the oocyte, including factors setting up the adequate epigenetic patterning of the zygotic genome. We previously reported that one such factor is the epigenetic repressor SMCHD1, whose maternal supply controls autosomal imprinted expression in mouse preimplantation embryos and mid-gestation placenta. In mouse preimplantation embryos, X chromosome inactivation is also an imprinted process. Combining genomics and imaging, we show that maternal SMCHD1 is required not only for the imprinted expression of Xist in preimplantation embryos, but also for the efficient silencing of the inactive X in both the preimplantation embryo and mid-gestation placenta. These results expand the role of SMCHD1 in enforcing the silencing of Polycomb targets. The inability of zygotic SMCHD1 to fully restore imprinted X inactivation further points to maternal SMCHD1's role in setting up the appropriate chromatin environment during preimplantation development, a critical window of epigenetic remodelling.


Subject(s)
Chromosomal Proteins, Non-Histone , RNA, Long Noncoding , X Chromosome Inactivation , Animals , Blastocyst/physiology , Chromatin/genetics , Chromatin/metabolism , Chromosomal Proteins, Non-Histone/metabolism , Embryonic Development , Genomic Imprinting , Mice , RNA, Long Noncoding/biosynthesis , RNA, Long Noncoding/genetics , RNA, Long Noncoding/metabolism , X Chromosome
9.
Nat Commun ; 13(1): 1658, 2022 03 29.
Article in English | MEDLINE | ID: mdl-35351876

ABSTRACT

The process of epigenetic silencing, while fundamentally important, is not yet completely understood. Here we report a replenishable female mouse embryonic stem cell (mESC) system, Xmas, that allows rapid assessment of X chromosome inactivation (XCI), the epigenetic silencing mechanism of one of the two X chromosomes that enables dosage compensation in female mammals. Through a targeted genetic screen in differentiating Xmas mESCs, we reveal that the BAF complex is required to create nucleosome-depleted regions at promoters on the inactive X chromosome during the earliest stages of establishment of XCI. Without this action gene silencing fails. Xmas mESCs provide a tractable model for screen-based approaches that enable the discovery of unknown facets of the female-specific process of XCI and epigenetic silencing more broadly.


Subject(s)
RNA, Long Noncoding , X Chromosome Inactivation , Animals , Chromatin/genetics , Dosage Compensation, Genetic , Epigenesis, Genetic , Female , Mice , RNA, Long Noncoding/genetics , X Chromosome/genetics , X Chromosome Inactivation/genetics
10.
Elife ; 92020 11 13.
Article in English | MEDLINE | ID: mdl-33186096

ABSTRACT

Genomic imprinting establishes parental allele-biased expression of a suite of mammalian genes based on parent-of-origin specific epigenetic marks. These marks are under the control of maternal effect proteins supplied in the oocyte. Here we report epigenetic repressor Smchd1 as a novel maternal effect gene that regulates the imprinted expression of ten genes in mice. We also found zygotic SMCHD1 had a dose-dependent effect on the imprinted expression of seven genes. Together, zygotic and maternal SMCHD1 regulate three classic imprinted clusters and eight other genes, including non-canonical imprinted genes. Interestingly, the loss of maternal SMCHD1 does not alter germline DNA methylation imprints pre-implantation or later in gestation. Instead, what appears to unite most imprinted genes sensitive to SMCHD1 is their reliance on polycomb-mediated methylation as germline or secondary imprints, therefore we propose that SMCHD1 acts downstream of polycomb imprints to mediate its function.


Subject(s)
Chromosomal Proteins, Non-Histone/metabolism , Genomic Imprinting/genetics , Animals , Blastocyst , Chromosomal Proteins, Non-Histone/genetics , DNA Methylation , Embryo, Mammalian/metabolism , Female , Gene Expression Regulation, Developmental/physiology , Genotype , Green Fluorescent Proteins , Male , Mice , Neural Stem Cells
11.
Nat Commun ; 11(1): 2420, 2020 05 15.
Article in English | MEDLINE | ID: mdl-32415101

ABSTRACT

Archetypal human pluripotent stem cells (hPSC) are widely considered to be equivalent in developmental status to mouse epiblast stem cells, which correspond to pluripotent cells at a late post-implantation stage of embryogenesis. Heterogeneity within hPSC cultures complicates this interspecies comparison. Here we show that a subpopulation of archetypal hPSC enriched for high self-renewal capacity (ESR) has distinct properties relative to the bulk of the population, including a cell cycle with a very low G1 fraction and a metabolomic profile that reflects a combination of oxidative phosphorylation and glycolysis. ESR cells are pluripotent and capable of differentiation into primordial germ cell-like cells. Global DNA methylation levels in the ESR subpopulation are lower than those in mouse epiblast stem cells. Chromatin accessibility analysis revealed a unique set of open chromatin sites in ESR cells. RNA-seq at the subpopulation and single cell levels shows that, unlike mouse epiblast stem cells, the ESR subset of hPSC displays no lineage priming, and that it can be clearly distinguished from gastrulating and extraembryonic cell populations in the primate embryo. ESR hPSC correspond to an earlier stage of post-implantation development than mouse epiblast stem cells.


Subject(s)
Embryonic Stem Cells/cytology , Germ Layers/cytology , Pluripotent Stem Cells/cytology , Animals , Cell Differentiation , Chromatin/metabolism , DNA Methylation , Epigenome , Flow Cytometry , Fluorescent Antibody Technique, Indirect , G1 Phase , Germ Layers/metabolism , Glycolysis , Humans , MAP Kinase Signaling System , Metabolomics , Mice , Mitochondria/metabolism , Oxidative Phosphorylation , RNA-Seq , Signal Transduction
12.
Cell Rep ; 27(2): 442-454.e5, 2019 04 09.
Article in English | MEDLINE | ID: mdl-30970248

ABSTRACT

Neural tube defects (NTDs) are common birth defects in humans and show an unexplained female bias. Female mice lacking the tumor suppressor p53 display NTDs with incomplete penetrance. We found that the combined loss of pro-apoptotic BIM and p53 caused 100% penetrant, female-exclusive NTDs, which allowed us to investigate the female-specific functions of p53. We report that female p53-/- embryonic neural tube samples show fewer cells with inactive X chromosome markers Xist and H3K27me3 and a concomitant increase in biallelic expression of the X-linked genes, Huwe1 and Usp9x. Decreased Xist and increased X-linked gene expression was confirmed by RNA sequencing. Moreover, we found that p53 directly bound response elements in the X chromosome inactivation center (XIC). Together, these findings suggest p53 directly activates XIC genes, without which there is stochastic failure in X chromosome inactivation, and that X chromosome inactivation failure may underlie the female bias in neural tube closure defects.


Subject(s)
Neural Tube Defects/genetics , Tumor Suppressor Protein p53/deficiency , Animals , Embryonic Stem Cells/pathology , Female , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Neural Tube Defects/pathology , Pregnancy , Stochastic Processes , Tumor Suppressor Protein p53/genetics , X Chromosome Inactivation
13.
Nat Struct Mol Biol ; 25(9): 766-777, 2018 09.
Article in English | MEDLINE | ID: mdl-30127357

ABSTRACT

The regulation of higher-order chromatin structure is complex and dynamic, and a full understanding of the suite of mechanisms governing this architecture is lacking. Here, we reveal the noncanonical SMC protein Smchd1 to be a novel regulator of long-range chromatin interactions in mice, and we add Smchd1 to the canon of epigenetic proteins required for Hox-gene regulation. The effect of losing Smchd1-dependent chromatin interactions has varying outcomes that depend on chromatin context. At autosomal targets transcriptionally sensitive to Smchd1 deletion, we found increased short-range interactions and ectopic enhancer activation. In contrast, the inactive X chromosome was transcriptionally refractive to Smchd1 ablation, despite chromosome-wide increases in short-range interactions. In the inactive X, we observed spreading of trimethylated histone H3 K27 (H3K27me3) domains into regions not normally decorated by this mark. Together, these data suggest that Smchd1 is able to insulate chromatin, thereby limiting access to other chromatin-modifying proteins.


Subject(s)
Chromatin/metabolism , Chromosomal Proteins, Non-Histone/physiology , Genes, Homeobox , Multigene Family , X Chromosome , Animals , Chromosomal Proteins, Non-Histone/genetics , Enhancer Elements, Genetic , Gene Deletion , Gene Silencing , Mice , Mice, Inbred C57BL , Mice, Knockout
14.
Nat Genet ; 49(2): 249-255, 2017 Feb.
Article in English | MEDLINE | ID: mdl-28067911

ABSTRACT

Bosma arhinia microphthalmia syndrome (BAMS) is an extremely rare and striking condition characterized by complete absence of the nose with or without ocular defects. We report here that missense mutations in the epigenetic regulator SMCHD1 mapping to the extended ATPase domain of the encoded protein cause BAMS in all 14 cases studied. All mutations were de novo where parental DNA was available. Biochemical tests and in vivo assays in Xenopus laevis embryos suggest that these mutations may behave as gain-of-function alleles. This finding is in contrast to the loss-of-function mutations in SMCHD1 that have been associated with facioscapulohumeral muscular dystrophy (FSHD) type 2. Our results establish SMCHD1 as a key player in nasal development and provide biochemical insight into its enzymatic function that may be exploited for development of therapeutics for FSHD.


Subject(s)
Choanal Atresia/genetics , Chromosomal Proteins, Non-Histone/genetics , Microphthalmos/genetics , Mutation, Missense/genetics , Nose/abnormalities , Animals , Cell Line , Child, Preschool , Epigenesis, Genetic/genetics , Female , Genetic Predisposition to Disease/genetics , Humans , Male , Mice , Mice, Inbred C57BL , Muscular Dystrophy, Facioscapulohumeral/genetics , Xenopus laevis/genetics
15.
Genom Data ; 10: 97-100, 2016 Dec.
Article in English | MEDLINE | ID: mdl-27766205

ABSTRACT

Reduced representation bisulfite sequencing (RRBS) provides an efficient method for measuring DNA methylation at single base resolution in regions of high CpG density. This technique has been extensively tested on the HiSeq2500, which uses a 4-colour detection method, however it is unclear if the method will also work on the NextSeq500 platform, which employs a 2-colour detection system. We created an RRBS library and sequenced it on both the HiSeq2500 and NextSeq500, and found no significant difference in the base composition of reads derived from either machine. Moreover, the methylation calls made from the data of each instrument were highly concordant, with methylation patterns across the genome appearing as expected. Therefore, RRBS can be sequenced on the Nextseq500 with comparable quality to that of the HiSeq2500. All sequencing data are deposited in the GEO database under accession number GSE87097.

16.
Nat Cell Biol ; 17(4): 365-75, 2015 Apr.
Article in English | MEDLINE | ID: mdl-25730472

ABSTRACT

Expansion and remodelling of the mammary epithelium requires a tight balance between cellular proliferation, differentiation and death. To explore cell survival versus cell death decisions in this organ, we deleted the pro-survival gene Mcl-1 in the mammary epithelium. Mcl-1 was found to be essential at multiple developmental stages including morphogenesis in puberty and alveologenesis in pregnancy. Moreover, Mcl-1-deficient basal cells were virtually devoid of repopulating activity, suggesting that this gene is required for stem cell function. Profound upregulation of the Mcl-1 protein was evident in alveolar cells at the switch to lactation, and Mcl-1 deficiency impaired lactation. Interestingly, EGF was identified as one of the most highly upregulated genes on lactogenesis and inhibition of EGF or mTOR signalling markedly impaired lactation, with concomitant decreases in Mcl-1 and phosphorylated ribosomal protein S6. These data demonstrate that Mcl-1 is essential for mammopoiesis and identify EGF as a critical trigger of Mcl-1 translation to ensure survival of milk-producing alveolar cells.


Subject(s)
Epidermal Growth Factor/biosynthesis , Lactation/genetics , Lactation/metabolism , Mammary Glands, Animal/metabolism , Myeloid Cell Leukemia Sequence 1 Protein/genetics , Animals , Apoptosis/genetics , Base Sequence , Cell Differentiation/genetics , Cell Line , Cell Proliferation/genetics , Cell Survival , Epidermal Growth Factor/antagonists & inhibitors , Epidermal Growth Factor/metabolism , Female , Gene Knockout Techniques , Mice , Mice, Inbred C57BL , Myeloid Cell Leukemia Sequence 1 Protein/biosynthesis , Phosphorylation , Pregnancy , Ribosomal Protein S6/metabolism , Sequence Analysis, RNA , Stem Cells/cytology , TOR Serine-Threonine Kinases/antagonists & inhibitors , Up-Regulation
SELECTION OF CITATIONS
SEARCH DETAIL