Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 31
Filter
Add more filters










Publication year range
1.
Nat Cell Biol ; 25(1): 134-144, 2023 Jan.
Article in English | MEDLINE | ID: mdl-36635505

ABSTRACT

In female mammals, one of the two X chromosomes becomes inactivated during development by X-chromosome inactivation (XCI). Although Polycomb repressive complex (PRC) 1 and PRC2 have both been implicated in gene silencing, their exact roles in XCI during in vivo development have remained elusive. To this end, we have studied mouse embryos lacking either PRC1 or PRC2. Here we demonstrate that the loss of either PRC has a substantial impact on maintenance of gene silencing on the inactive X chromosome (Xi) in extra-embryonic tissues, with overlapping yet different genes affected, indicating potentially independent roles of the two complexes. Importantly, a lack of PRC1 does not affect PRC2/H3K27me3 accumulation and a lack of PRC2 does not impact PRC1/H2AK119ub1 accumulation on the Xi. Thus PRC1 and PRC2 contribute independently to the maintenance of XCI in early post-implantation extra-embryonic lineages, revealing that both Polycomb complexes can be directly involved and differently deployed in XCI.


Subject(s)
Polycomb Repressive Complex 1 , X Chromosome Inactivation , Female , Mice , Animals , Polycomb Repressive Complex 1/genetics , Polycomb Repressive Complex 1/metabolism , X Chromosome Inactivation/genetics , Polycomb Repressive Complex 2/genetics , Polycomb Repressive Complex 2/metabolism , Polycomb-Group Proteins/genetics , X Chromosome/genetics , X Chromosome/metabolism , Mammals/metabolism
2.
Nat Commun ; 13(1): 7159, 2022 11 28.
Article in English | MEDLINE | ID: mdl-36443290

ABSTRACT

Polycomb group proteins (PcG), polycomb repressive complexes 1 and 2 (PRC1 and 2), repress lineage inappropriate genes during development to maintain proper cellular identities. It has been recognized that PRC1 localizes at the replication fork, however, the precise functions of PRC1 during DNA replication are elusive. Here, we reveal that a variant PRC1 containing PCGF1 (PCGF1-PRC1) prevents overloading of activators and chromatin remodeling factors on nascent DNA and thereby mediates proper deposition of nucleosomes and correct downstream chromatin configurations in hematopoietic stem and progenitor cells (HSPCs). This function of PCGF1-PRC1 in turn facilitates PRC2-mediated repression of target genes such as Hmga2 and restricts premature myeloid differentiation. PCGF1-PRC1, therefore, maintains the differentiation potential of HSPCs by linking proper nucleosome configuration at the replication fork with PcG-mediated gene silencing to ensure life-long hematopoiesis.


Subject(s)
Chromatin , DNA Replication , Chromatin/genetics , Cell Lineage/genetics , Nucleosomes/genetics , Polycomb-Group Proteins , Polycomb Repressive Complex 2
3.
Science ; 375(6580): 574-578, 2022 02 04.
Article in English | MEDLINE | ID: mdl-35113719

ABSTRACT

The mammalian blastocyst consists of three distinct cell types: epiblast, trophoblast (TB), and primitive endoderm (PrE). Although embryonic stem cells (ESCs) and trophoblast stem cells (TSCs) retain the functional properties of epiblast and TB, respectively, stem cells that fully recapitulate the developmental potential of PrE have not been established. Here, we report derivation of primitive endoderm stem cells (PrESCs) in mice. PrESCs recapitulate properties of embryonic day 4.5 founder PrE, are efficiently incorporated into PrE upon blastocyst injection, generate functionally competent PrE-derived tissues, and support fetal development of PrE-depleted blastocysts in chimeras. Furthermore, PrESCs can establish interactions with ESCs and TSCs and generate descendants with yolk sac-like structures in utero. Establishment of PrESCs will enable the elucidation of the mechanisms for PrE specification and subsequent pre- and postimplantation development.


Subject(s)
Embryonic Stem Cells/cytology , Embryonic Stem Cells/physiology , Endoderm/cytology , Endoderm/embryology , Animals , Blastocyst/cytology , Blastocyst/physiology , Cell Differentiation , Cell Line , Cell Lineage , Chimera , Embryonic Development , Endoderm/growth & development , Fetal Development , Germ Layers/cytology , Germ Layers/embryology , Mice , Mice, Inbred C57BL , Trophoblasts/cytology , Trophoblasts/physiology
4.
Nat Commun ; 12(1): 5341, 2021 09 09.
Article in English | MEDLINE | ID: mdl-34504070

ABSTRACT

Polycomb repressive complexes-1 and -2 (PRC1 and 2) silence developmental genes in a spatiotemporal manner during embryogenesis. How Polycomb group (PcG) proteins orchestrate down-regulation of target genes upon differentiation, however, remains elusive. Here, by differentiating embryonic stem cells into embryoid bodies, we reveal a crucial role for the PCGF1-containing variant PRC1 complex (PCGF1-PRC1) to mediate differentiation-associated down-regulation of a group of genes. Upon differentiation cues, transcription is down-regulated at these genes, in association with PCGF1-PRC1-mediated deposition of histone H2AK119 mono-ubiquitination (H2AK119ub1) and PRC2 recruitment. In the absence of PCGF1-PRC1, both H2AK119ub1 deposition and PRC2 recruitment are disrupted, leading to aberrant expression of target genes. PCGF1-PRC1 is, therefore, required for initiation and consolidation of PcG-mediated gene repression during differentiation.


Subject(s)
Embryoid Bodies/metabolism , Gene Expression Regulation, Developmental , Histones/genetics , Mouse Embryonic Stem Cells/metabolism , Polycomb Repressive Complex 1/genetics , Polycomb Repressive Complex 2/genetics , Animals , Cell Differentiation , Embryo, Mammalian , Embryoid Bodies/cytology , Histones/metabolism , Kruppel-Like Factor 4 , Kruppel-Like Transcription Factors/genetics , Kruppel-Like Transcription Factors/metabolism , Mice , Mice, Transgenic , Mouse Embryonic Stem Cells/cytology , Platelet-Derived Growth Factor/genetics , Platelet-Derived Growth Factor/metabolism , Polycomb Repressive Complex 1/metabolism , Polycomb Repressive Complex 2/metabolism , Primary Cell Culture , SOXC Transcription Factors/genetics , SOXC Transcription Factors/metabolism , T-Box Domain Proteins/genetics , T-Box Domain Proteins/metabolism , Transcription Factors/genetics , Transcription Factors/metabolism , Transcription, Genetic , Ubiquitination
5.
Development ; 148(10)2021 05 15.
Article in English | MEDLINE | ID: mdl-33998651

ABSTRACT

Heterochromatin-related epigenetic mechanisms, such as DNA methylation, facilitate pairing of homologous chromosomes during the meiotic prophase of mammalian spermatogenesis. In pro-spermatogonia, de novo DNA methylation plays a key role in completing meiotic prophase and initiating meiotic division. However, the role of maintenance DNA methylation in the regulation of meiosis, especially in the adult, is not well understood. Here, we reveal that NP95 (also known as UHRF1) and DNMT1 - two essential proteins for maintenance DNA methylation - are co-expressed in spermatogonia and are necessary for meiosis in male germ cells. We find that Np95- or Dnmt1-deficient spermatocytes exhibit spermatogenic defects characterized by synaptic failure during meiotic prophase. In addition, assembly of pericentric heterochromatin clusters in early meiotic prophase, a phenomenon that is required for subsequent pairing of homologous chromosomes, is disrupted in both mutants. Based on these observations, we propose that DNA methylation, established in pre-meiotic spermatogonia, regulates synapsis of homologous chromosomes and, in turn, quality control of male germ cells. Maintenance DNA methylation, therefore, plays a role in ensuring faithful transmission of both genetic and epigenetic information to offspring.


Subject(s)
CCAAT-Enhancer-Binding Proteins/genetics , Chromosome Pairing/genetics , DNA (Cytosine-5-)-Methyltransferase 1/genetics , DNA Methylation/genetics , Spermatocytes/growth & development , Spermatogenesis/genetics , Ubiquitin-Protein Ligases/genetics , Adult Germline Stem Cells/cytology , Animals , CCAAT-Enhancer-Binding Proteins/metabolism , DNA (Cytosine-5-)-Methyltransferase 1/metabolism , Epigenesis, Genetic/genetics , Heterochromatin/metabolism , Male , Mice , Mice, Knockout , Spermatocytes/physiology , Spermatogenesis/physiology , Ubiquitin-Protein Ligases/metabolism
6.
Mol Cell ; 76(3): 437-452.e6, 2019 11 07.
Article in English | MEDLINE | ID: mdl-31521505

ABSTRACT

Polycomb repressive complex 2 (PRC2) is composed of EED, SUZ12, and EZH1/2 and mediates mono-, di-, and trimethylation of histone H3 at lysine 27. At least two independent subcomplexes exist, defined by their specific accessory proteins: PRC2.1 (PCL1-3, EPOP, and PALI1/2) and PRC2.2 (AEBP2 and JARID2). We show that PRC2.1 and PRC2.2 share the majority of target genes in mouse embryonic stem cells. The loss of PCL1-3 is sufficient to evict PRC2.1 from Polycomb target genes but only leads to a partial reduction of PRC2.2 and H3K27me3. Conversely, disruption of PRC2.2 function through the loss of either JARID2 or RING1A/B is insufficient to completely disrupt targeting of SUZ12 by PCLs. Instead, the combined loss of both PRC2.1 and PRC2.2 is required, leading to the global mislocalization of SUZ12. This supports a model in which the specific accessory proteins within PRC2.1 and PRC2.2 cooperate to direct H3K27me3 via both synergistic and independent mechanisms.


Subject(s)
Chromatin/metabolism , Histones/metabolism , Mouse Embryonic Stem Cells/metabolism , Polycomb Repressive Complex 2/metabolism , Protein Processing, Post-Translational , Animals , Binding Sites , Cell Line, Tumor , Chromatin/genetics , Humans , Methylation , Mice , Polycomb Repressive Complex 1/genetics , Polycomb Repressive Complex 1/metabolism , Polycomb Repressive Complex 2/genetics , Protein Binding , Ubiquitin-Protein Ligases/genetics , Ubiquitin-Protein Ligases/metabolism
7.
Nucleic Acids Res ; 47(17): 9005-9023, 2019 09 26.
Article in English | MEDLINE | ID: mdl-31363749

ABSTRACT

CpG islands (CGIs) are associated with the majority of mammalian gene promoters and function to recruit chromatin modifying enzymes. It has therefore been proposed that CGIs regulate gene expression through chromatin-based mechanisms, however in most cases this has not been directly tested. Here, we reveal that the histone H3 lysine 36 (H3K36) demethylase activity of the CGI-binding KDM2 proteins contributes only modestly to the H3K36me2-depleted state at CGI-associated gene promoters and is dispensable for normal gene expression. Instead, we discover that KDM2 proteins play a widespread and demethylase-independent role in constraining gene expression from CGI-associated gene promoters. We further show that KDM2 proteins shape RNA Polymerase II occupancy but not chromatin accessibility at CGI-associated promoters. Together this reveals a demethylase-independent role for KDM2 proteins in transcriptional repression and uncovers a new function for CGIs in constraining gene expression.


Subject(s)
CpG Islands/genetics , F-Box Proteins/physiology , Histones/metabolism , Jumonji Domain-Containing Histone Demethylases/physiology , Promoter Regions, Genetic , Transcription, Genetic , Animals , Chromatin/enzymology , Chromatin/metabolism , DNA Methylation , F-Box Proteins/genetics , F-Box Proteins/metabolism , Gene Expression Regulation , HEK293 Cells , Humans , Jumonji Domain-Containing Histone Demethylases/genetics , Jumonji Domain-Containing Histone Demethylases/metabolism , Lysine/metabolism , Mice , Models, Genetic , Mouse Embryonic Stem Cells/enzymology , Mouse Embryonic Stem Cells/metabolism , RNA Polymerase II/metabolism
8.
Life Sci Alliance ; 2(2)2019 04.
Article in English | MEDLINE | ID: mdl-30936185

ABSTRACT

The immune system encompasses acquired and innate immunity that matures through interaction with microenvironmental components. Cytokines serve as environmental factors that foster functional maturation of immune cells. Although NOD/SCID/IL2rgKO (NSG) humanized mice support investigation of human immunity in vivo, a species barrier between human immune cells and the mouse microenvironment limits human acquired as well as innate immune function. To study the roles of human cytokines in human acquired and innate immune cell development, we created NSG mice expressing hIL-7 and hIL-15. Although hIL-7 alone was not sufficient for supporting human NK cell development in vivo, increased frequencies of human NK cells were confirmed in multiple organs of hIL-7 and hIL-15 double knockin (hIL-7xhIL-15 KI) NSG mice engrafted with human hematopoietic stem cells. hIL-7xhIL-15 KI NSG humanized mice provide a valuable in vivo model to investigate development and function of human NK cells.


Subject(s)
Cell Differentiation , Gene Knock-In Techniques , Interleukin-15/blood , Interleukin-15/genetics , Interleukin-7/blood , Interleukin-7/genetics , Killer Cells, Natural/physiology , Animals , CD56 Antigen/metabolism , Female , Fetal Blood/cytology , Humans , Male , Mice , Mice, Inbred C57BL , Mice, Inbred NOD , Mice, SCID , Mice, Transgenic , Models, Animal , Thymus Gland/cytology , Transcriptome , Transplantation, Heterologous
9.
Mol Cell ; 74(5): 1020-1036.e8, 2019 06 06.
Article in English | MEDLINE | ID: mdl-31029541

ABSTRACT

The Polycomb system modifies chromatin and plays an essential role in repressing gene expression to control normal mammalian development. However, the components and mechanisms that define how Polycomb protein complexes achieve this remain enigmatic. Here, we use combinatorial genetic perturbation coupled with quantitative genomics to discover the central determinants of Polycomb-mediated gene repression in mouse embryonic stem cells. We demonstrate that canonical Polycomb repressive complex 1 (PRC1), which mediates higher-order chromatin structures, contributes little to gene repression. Instead, we uncover an unexpectedly high degree of synergy between variant PRC1 complexes, which is fundamental to gene repression. We further demonstrate that variant PRC1 complexes are responsible for distinct pools of H2A monoubiquitylation that are associated with repression of Polycomb target genes and silencing during X chromosome inactivation. Together, these discoveries reveal a new variant PRC1-dependent logic for Polycomb-mediated gene repression.


Subject(s)
Chromatin/genetics , Genomics , Polycomb Repressive Complex 1/genetics , X Chromosome Inactivation/genetics , Animals , Histones/genetics , Mice , Mouse Embryonic Stem Cells/metabolism , RNA Interference , Ubiquitination/genetics
10.
Development ; 145(19)2018 10 11.
Article in English | MEDLINE | ID: mdl-30190278

ABSTRACT

Suppression of Meis genes in the distal limb bud is required for proximal-distal (PD) specification of the forelimb. Polycomb group (PcG) factors play a role in downregulation of retinoic acid (RA)-related signals in the distal forelimb bud, causing Meis repression. It is, however, not known whether downregulation of RA-related signals and PcG-mediated proximal gene repression are functionally linked. Here, we reveal that PcG factors and RA-related signals antagonize each other to polarize Meis2 expression along the PD axis in mouse. Supported by mathematical modeling and simulation, we propose that PcG factors are required to adjust the threshold for RA-related signaling to regulate Meis2 expression. Finally, we show that a variant Polycomb repressive complex 1 (PRC1), incorporating PCGF3 and PCGF5, represses Meis2 expression in the distal limb bud. Taken together, we reveal a previously unknown link between PcG proteins and downregulation of RA-related signals to mediate the phase transition of Meis2 transcriptional status during forelimb patterning.


Subject(s)
Forelimb/embryology , Homeodomain Proteins/metabolism , Limb Buds/metabolism , Polycomb Repressive Complex 1/metabolism , Tretinoin/metabolism , Animals , Forelimb/metabolism , Gene Expression Regulation, Developmental , Genetic Loci , Mice , Signal Transduction
11.
Elife ; 72018 05 29.
Article in English | MEDLINE | ID: mdl-29809150

ABSTRACT

CpG islands are gene regulatory elements associated with the majority of mammalian promoters, yet how they regulate gene expression remains poorly understood. Here, we identify FBXL19 as a CpG island-binding protein in mouse embryonic stem (ES) cells and show that it associates with the CDK-Mediator complex. We discover that FBXL19 recruits CDK-Mediator to CpG island-associated promoters of non-transcribed developmental genes to prime these genes for activation during cell lineage commitment. We further show that recognition of CpG islands by FBXL19 is essential for mouse development. Together this reveals a new CpG island-centric mechanism for CDK-Mediator recruitment to developmental gene promoters in ES cells and a requirement for CDK-Mediator in priming these developmental genes for activation during cell lineage commitment.


Subject(s)
Cell Lineage , CpG Islands , DNA-Binding Proteins/physiology , F-Box Proteins/physiology , Genes, Developmental , Mouse Embryonic Stem Cells/cytology , Animals , Cell Differentiation , Cells, Cultured , Chromatin/genetics , Chromatin/metabolism , Cyclin-Dependent Kinase 8/genetics , Cyclin-Dependent Kinase 8/metabolism , DNA Methylation , Mediator Complex/physiology , Mice , Mice, Inbred C57BL , Mice, Knockout , Mouse Embryonic Stem Cells/metabolism , Promoter Regions, Genetic , Protein Binding , Protein Domains
12.
Mol Cell ; 70(3): 408-421.e8, 2018 05 03.
Article in English | MEDLINE | ID: mdl-29628311

ABSTRACT

The polycomb repressive complex 2 (PRC2) consists of core subunits SUZ12, EED, RBBP4/7, and EZH1/2 and is responsible for mono-, di-, and tri-methylation of lysine 27 on histone H3. Whereas two distinct forms exist, PRC2.1 (containing one polycomb-like protein) and PRC2.2 (containing AEBP2 and JARID2), little is known about their differential functions. Here, we report the discovery of a family of vertebrate-specific PRC2.1 proteins, "PRC2 associated LCOR isoform 1" (PALI1) and PALI2, encoded by the LCOR and LCORL gene loci, respectively. PALI1 promotes PRC2 methyltransferase activity in vitro and in vivo and is essential for mouse development. Pali1 and Aebp2 define mutually exclusive, antagonistic PRC2 subtypes that exhibit divergent H3K27-tri-methylation activities. The balance of these PRC2.1/PRC2.2 activities is required for the appropriate regulation of polycomb target genes during differentiation. PALI1/2 potentially link polycombs with transcriptional co-repressors in the regulation of cellular identity during development and in cancer.


Subject(s)
Polycomb Repressive Complex 2/genetics , Repressor Proteins/genetics , Vertebrates/genetics , Amino Acid Sequence , Animals , Cell Differentiation/genetics , Cell Line , HEK293 Cells , Histones/genetics , Humans , Methylation , Methyltransferases/genetics , Mice , Neoplasms/genetics , Sequence Alignment
13.
Science ; 356(6342): 1081-1084, 2017 06 09.
Article in English | MEDLINE | ID: mdl-28596365

ABSTRACT

Recruitment of the Polycomb repressive complexes PRC1 and PRC2 by Xist RNA is an important paradigm for chromatin regulation by long noncoding RNAs. Here, we show that the noncanonical Polycomb group RING finger 3/5 (PCGF3/5)-PRC1 complex initiates recruitment of both PRC1 and PRC2 in response to Xist RNA expression. PCGF3/5-PRC1-mediated ubiquitylation of histone H2A signals recruitment of other noncanonical PRC1 complexes and of PRC2, the latter leading to deposition of histone H3 lysine 27 methylation chromosome-wide. Pcgf3/5 gene knockout results in female-specific embryo lethality and abrogates Xist-mediated gene repression, highlighting a key role for Polycomb in Xist-dependent chromosome silencing. Our findings overturn existing models for Polycomb recruitment by Xist RNA and establish precedence for H2AK119u1 in initiating Polycomb domain formation in a physiological context.


Subject(s)
Embryonic Stem Cells/metabolism , Polycomb Repressive Complex 1/metabolism , Polycomb-Group Proteins/metabolism , X Chromosome Inactivation , Animals , Female , Mice , Polycomb-Group Proteins/genetics , RNA, Long Noncoding/metabolism
14.
Genes Dev ; 30(22): 2475-2485, 2016 11 15.
Article in English | MEDLINE | ID: mdl-27913604

ABSTRACT

In general, cell fate is determined primarily by transcription factors, followed by epigenetic mechanisms fixing the status. While the importance of transcription factors controlling cell fate has been well characterized, epigenetic regulation of cell fate maintenance remains to be elucidated. Here we provide an obvious fate conversion case, in which the inactivation of polycomb-medicated epigenetic regulation results in conversion of T-lineage progenitors to the B-cell fate. In T-cell-specific Ring1A/B-deficient mice, T-cell development was severely blocked at an immature stage. We found that these developmentally arrested T-cell precursors gave rise to functional B cells upon transfer to immunodeficient mice. We further demonstrated that the arrest was almost completely canceled by additional deletion of Pax5 These results indicate that the maintenance of T-cell fate critically requires epigenetic suppression of the B-lineage gene program.


Subject(s)
B-Lymphocytes/cytology , Cell Transformation, Neoplastic/genetics , Epigenesis, Genetic/genetics , Gene Silencing , Polycomb-Group Proteins/metabolism , T-Lymphocytes/cytology , Animals , Cell Lineage , Gene Deletion , Gene Expression Regulation, Developmental , Immunoglobulin Heavy Chains/genetics , Mice, Inbred C57BL , PAX5 Transcription Factor/genetics , PAX5 Transcription Factor/metabolism , Polycomb Repressive Complex 1/genetics , Promoter Regions, Genetic/genetics , Ubiquitin-Protein Ligases/genetics
15.
Cell Stem Cell ; 19(1): 81-94, 2016 07 07.
Article in English | MEDLINE | ID: mdl-27151458

ABSTRACT

Repression of endogenous retroviruses (ERVs) in mammals involves several epigenetic mechanisms. Acute loss of the maintenance methyltransferase Dnmt1 induces widespread DNA demethylation and transcriptional activation of ERVs, including CpG-rich IAP (intracisternal A particle) proviruses. Here, we show that this effect is not due simply to a loss of DNA methylation. Conditional deletions reveal that both Dnmt1 and Np95 are essential for maintenance DNA methylation. However, while IAPs are derepressed in Dnmt1-ablated embryos and embryonic stem cells (ESCs), these ERVs remain silenced when Np95 is deleted alone or in combination with Dnmt1. This paradoxical phenotype results from an ectopic interaction between NP95 and the H3K9 methyltransferase SETDB1. Normally, SETDB1 maintains silencing of IAPs, but in the absence of DNMT1, prolonged binding of NP95 to hemimethylated DNA transiently disrupts SETDB1-dependent H3K9me3 deposition. Thus, our observations reveal an unexpected antagonistic interplay between two repressive pathways involved in retroviral silencing in mammalian cells.


Subject(s)
DNA (Cytosine-5-)-Methyltransferases/metabolism , DNA Methylation/genetics , DNA/metabolism , Endogenous Retroviruses/metabolism , Histone-Lysine N-Methyltransferase/metabolism , Mouse Embryonic Stem Cells/metabolism , Nuclear Proteins/metabolism , Virus Activation , Animals , CCAAT-Enhancer-Binding Proteins , Cell Differentiation , DNA (Cytosine-5-)-Methyltransferase 1 , Female , Gene Dosage , Gene Expression Regulation, Developmental , Gene Silencing , Genes, Intracisternal A-Particle , Genetic Loci , Histones/metabolism , Lysine/metabolism , Mice , Mice, Knockout , Models, Biological , Mutation/genetics , Nuclear Proteins/chemistry , Placenta/metabolism , Pregnancy , Protein Binding , Protein Domains , Trophoblasts/metabolism , Ubiquitin-Protein Ligases
16.
PLoS Genet ; 12(1): e1005776, 2016 Jan.
Article in English | MEDLINE | ID: mdl-26789017

ABSTRACT

Thymic medullary regions are formed in neonatal mice as islet-like structures, which increase in size over time and eventually fuse a few weeks after birth into a continuous structure. The development of medullary thymic epithelial cells (TEC) is dependent on NF-κB associated signaling though other signaling pathways may contribute. Here, we demonstrate that Stat3-mediated signals determine medullary TEC cellularity, architectural organization and hence the size of the medulla. Deleting Stat3 expression selectively in thymic epithelia precludes the postnatal enlargement of the medulla retaining a neonatal architecture of small separate medullary islets. In contrast, loss of Stat3 expression in cortical TEC neither affects the cellularity or organization of the epithelia. Activation of Stat3 is mainly positioned downstream of EGF-R as its ablation in TEC phenocopies the loss of Stat3 expression in these cells. These results indicate that Stat3 meditated signal via EGF-R is required for the postnatal development of thymic medullary regions.


Subject(s)
Cell Differentiation/genetics , Epithelial Cells , ErbB Receptors/genetics , STAT3 Transcription Factor/biosynthesis , Animals , Embryonic Development , ErbB Receptors/biosynthesis , Flow Cytometry , Gene Expression Regulation, Developmental , Mice , STAT3 Transcription Factor/genetics , Signal Transduction , T-Lymphocytes/metabolism , Thymocytes/metabolism , Thymus Gland/growth & development , Thymus Gland/metabolism
17.
Development ; 143(2): 276-85, 2016 Jan 15.
Article in English | MEDLINE | ID: mdl-26674308

ABSTRACT

Polycomb group (PcG) proteins play a pivotal role in silencing developmental genes and help to maintain various stem and precursor cells and regulate their differentiation. PcG factors also regulate dynamic and complex regional specification, particularly in mammals, but this activity is mechanistically not well understood. In this study, we focused on proximal-distal (PD) patterning of the mouse forelimb bud to elucidate how PcG factors contribute to a regional specification process that depends on developmental signals. Depletion of the RING1 proteins RING1A (RING1) and RING1B (RNF2), which are essential components of Polycomb repressive complex 1 (PRC1), led to severe defects in forelimb formation along the PD axis. We show that preferential defects in early distal specification in Ring1A/B-deficient forelimb buds accompany failures in the repression of proximal signal circuitry bound by RING1B, including Meis1/2, and the activation of distal signal circuitry in the prospective distal region. Additional deletion of Meis2 induced partial restoration of the distal gene expression and limb formation seen in the Ring1A/B-deficient mice, suggesting a crucial role for RING1-dependent repression of Meis2 and likely also Meis1 for distal specification. We suggest that the RING1-MEIS1/2 axis is regulated by early PD signals and contributes to the initiation or maintenance of the distal signal circuitry.


Subject(s)
Forelimb/embryology , Homeodomain Proteins/metabolism , Polycomb Repressive Complex 1/metabolism , Ubiquitin-Protein Ligases/metabolism , Animals , Chromatin Immunoprecipitation , Cytochrome P-450 Enzyme System/genetics , Cytochrome P-450 Enzyme System/metabolism , Female , Gene Expression Regulation, Developmental/genetics , Gene Expression Regulation, Developmental/physiology , Homeodomain Proteins/genetics , In Situ Hybridization , Male , Mice , Mice, Mutant Strains , Polycomb Repressive Complex 1/genetics , Pregnancy , Retinoic Acid 4-Hydroxylase , Tretinoin/pharmacology , Ubiquitin-Protein Ligases/genetics
18.
Cell ; 157(6): 1445-1459, 2014 Jun 05.
Article in English | MEDLINE | ID: mdl-24856970

ABSTRACT

Chromatin modifying activities inherent to polycomb repressive complexes PRC1 and PRC2 play an essential role in gene regulation, cellular differentiation, and development. However, the mechanisms by which these complexes recognize their target sites and function together to form repressive chromatin domains remain poorly understood. Recruitment of PRC1 to target sites has been proposed to occur through a hierarchical process, dependent on prior nucleation of PRC2 and placement of H3K27me3. Here, using a de novo targeting assay in mouse embryonic stem cells we unexpectedly discover that PRC1-dependent H2AK119ub1 leads to recruitment of PRC2 and H3K27me3 to effectively initiate a polycomb domain. This activity is restricted to variant PRC1 complexes, and genetic ablation experiments reveal that targeting of the variant PCGF1/PRC1 complex by KDM2B to CpG islands is required for normal polycomb domain formation and mouse development. These observations provide a surprising PRC1-dependent logic for PRC2 occupancy at target sites in vivo.


Subject(s)
Embryonic Stem Cells/metabolism , F-Box Proteins/metabolism , Histones/metabolism , Jumonji Domain-Containing Histone Demethylases/metabolism , Polycomb Repressive Complex 1/metabolism , Polycomb Repressive Complex 2/metabolism , Animals , Bone Development , CpG Islands , F-Box Proteins/chemistry , F-Box Proteins/genetics , Genes, Lethal , Genome-Wide Association Study , Jumonji Domain-Containing Histone Demethylases/chemistry , Jumonji Domain-Containing Histone Demethylases/genetics , Mice , Protein Structure, Tertiary
19.
PLoS Genet ; 9(11): e1003897, 2013 Nov.
Article in English | MEDLINE | ID: mdl-24244179

ABSTRACT

Molecular mechanisms for the establishment of transcriptional memory are poorly understood. 5,6-dichloro-1-D-ribofuranosyl-benzimidazole (DRB) is a P-TEFb kinase inhibitor that artificially induces the poised RNA polymerase II (RNAPII), thereby manifesting intermediate steps for the establishment of transcriptional activation. Here, using genetics and DRB, we show that mammalian Absent, small, or homeotic discs 1-like (Ash1l), a member of the trithorax group proteins, methylates Lys36 of histone H3 to promote the establishment of Hox gene expression by counteracting Polycomb silencing. Importantly, we found that Ash1l-dependent Lys36 di-, tri-methylation of histone H3 in a coding region and exclusion of Polycomb group proteins occur independently of transcriptional elongation in embryonic stem (ES) cells, although both were previously thought to be consequences of transcription. Genome-wide analyses of histone H3 Lys36 methylation under DRB treatment have suggested that binding of the retinoic acid receptor (RAR) to a certain genomic region promotes trimethylation in the RAR-associated gene independent of its ongoing transcription. Moreover, DRB treatment unveils a parallel response between Lys36 methylation of histone H3 and occupancy of either Tip60 or Mof in a region-dependent manner. We also found that Brg1 is another key player involved in the response. Our results uncover a novel regulatory cascade orchestrated by Ash1l with RAR and provide insights into mechanisms underlying the establishment of the transcriptional activation that counteracts Polycomb silencing.


Subject(s)
DNA-Binding Proteins/metabolism , Histones/metabolism , Polycomb-Group Proteins/genetics , Transcription Factors/metabolism , Transcription, Genetic , Transcriptional Elongation Factors/genetics , Animals , Chromatin/genetics , DNA-Binding Proteins/genetics , Dichlororibofuranosylbenzimidazole/pharmacology , Drosophila Proteins/metabolism , Drosophila melanogaster , Gene Silencing , Histone Acetyltransferases/metabolism , Histone-Lysine N-Methyltransferase , Histones/genetics , Humans , Lysine/genetics , Methylation , Nuclear Proteins/metabolism , Polycomb-Group Proteins/metabolism , Positive Transcriptional Elongation Factor B/antagonists & inhibitors , Positive Transcriptional Elongation Factor B/metabolism , RNA Polymerase II/genetics , Transcription Factors/genetics , Transcriptional Elongation Factors/metabolism
20.
Mol Cell Biol ; 31(2): 351-64, 2011 Jan.
Article in English | MEDLINE | ID: mdl-21059868

ABSTRACT

The Polycomb group of proteins forms at least two distinct complexes designated the Polycomb repressive complex-1 (PRC1) and PRC2. These complexes cooperate to mediate transcriptional repression of their target genes, including the Hox gene cluster and the Cdkn2a genes. Mammalian Polycomb-like gene Pcl2/Mtf2 is expressed as four different isoforms, and the longest one contains a Tudor domain and two plant homeodomain (PHD) fingers. Pcl2 forms a complex with PRC2 and binds to Hox genes in a PRC2-dependent manner. We show that Pcl2 is a functional component of PRC2 and is required for PRC2-mediated Hox repression. Pcl2, however, exhibits a profound synergistic effect on PRC1-mediated Hox repression, which is not accompanied by major alterations in the local trimethylation of histone H3 at lysine 27 (H3K27me3) or PRC1 deposition. Pcl2 therefore functions in collaboration with both PRC2 and PRC1 to repress Hox gene expression during axial development. Paradoxically, in embryonic fibroblasts, Pcl2 is shown to activate the expression of Cdkn2a and promote cellular senescence, presumably by suppressing the catalytic activity of PRC2 locally. Taken together, we show that Pcl2 differentially regulates Polycomb-mediated repression of Hox and Cdkn2a genes. We therefore propose a novel role for Pcl2 to modify functional engagement of PRC2 and PRC1, which could be modulated by sensing cellular circumstances.


Subject(s)
Genes, Homeobox , Genes, p16 , Histone-Lysine N-Methyltransferase/metabolism , Multigene Family , Protein Isoforms/metabolism , Repressor Proteins/metabolism , Animals , Cells, Cultured , Fibroblasts/cytology , Fibroblasts/physiology , Histone-Lysine N-Methyltransferase/genetics , Humans , Mice , Polycomb Repressive Complex 2 , Polycomb-Group Proteins , Promoter Regions, Genetic , Protein Isoforms/genetics , RNA, Small Interfering/genetics , RNA, Small Interfering/metabolism , Repressor Proteins/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...