43 results on '"PRC2"'
Search Results
2. Nucleation and spreading maintain Polycomb domains every cell cycle
- Author
-
Veronezi, Giovana M.B. and Ramachandran, Srinivas
- Published
- 2024
- Full Text
- View/download PDF
3. A conserved switch to less catalytically active Polycomb repressive complexes in non-dividing cells
- Author
-
McCole, Rachel, Nolan, James, Reck, David M., Monger, Craig, Rustichelli, Samantha, Conway, Eric, Brien, Gerard L., Wang, Cheng, Deevy, Orla, Neikes, Hannah K., Bashore, Frances M., Mooney, Aoibhinn, Flavin, Richard, Vandenberghe, Elisabeth, Flanigan, Sarena F., Pasini, Diego, Davidovich, Chen, Vermeulen, Michiel, James, Lindsey I., Healy, Evan, and Bracken, Adrian P.
- Published
- 2025
- Full Text
- View/download PDF
4. Gene silencing by EZH2 suppresses TGF-β activity within the decidua to avert pregnancy-adverse wound healing at the maternal-fetal interface
- Author
-
Osokine, Ivan, Siewiera, Johan, Rideaux, Damon, Ma, Stephany, Tsukui, Tatsuya, and Erlebacher, Adrian
- Subjects
Biological Sciences ,Contraception/Reproduction ,Genetics ,Underpinning research ,1.1 Normal biological development and functioning ,Reproductive health and childbirth ,Animals ,Decidua ,Embryo Implantation ,Embryo ,Mammalian ,Enhancer of Zeste Homolog 2 Protein ,Female ,Gene Expression ,Gene Silencing ,Histones ,Humans ,Mice ,Inbred C57BL ,Placenta ,Pregnancy ,Stromal Cells ,Transforming Growth Factor beta ,Wound Healing ,EZH2 ,PRC2 ,TGF-beta ,decidualization ,epigenetics ,fibrosis ,pregnancy ,wound healing ,Biochemistry and Cell Biology ,Medical Physiology ,Biological sciences - Abstract
A little-appreciated feature of early pregnancy is that embryo implantation and placental outgrowth do not evoke wound-healing responses in the decidua, the specialized endometrial tissue that surrounds the conceptus. Here, we provide evidence that this phenomenon is partly due to an active program of gene silencing mediated by EZH2, a histone methyltransferase that generates repressive histone 3 lysine 27 trimethyl (H3K27me3) histone marks. We find that pregnancies in mice with EZH2-deficient decidual stromal cells frequently fail by mid-gestation, with the decidua showing ectopic myofibroblast formation, peri-embryonic collagen deposition, and gene expression profiles associated with transforming growth factor β (TGF-β)-driven fibroblast activation and fibrogenic extracellular matrix (ECM) remodeling. Analogous responses are observed when the mutant decidua is surgically wounded, while blockade of TGF-β receptor signaling inhibits the defects and improves reproductive outcomes. Together, these results highlight a critical feature of reproductive success and have implications for the context-specific control of TGF-β-mediated wound-healing responses elsewhere in the body.
- Published
- 2022
5. The apparent loss of PRC2 chromatin occupancy as an artifact of RNA depletion.
- Author
-
Healy, Evan, Zhang, Qi, Gail, Emma H., Agius, Samuel C., Sun, Guizhi, Bullen, Michael, Pandey, Varun, Das, Partha Pratim, Polo, Jose M., and Davidovich, Chen
- Abstract
RNA has been implicated in the recruitment of chromatin modifiers, and previous studies have provided evidence in favor and against this idea. RNase treatment of chromatin is commonly used to study RNA-mediated regulation of chromatin modifiers, but the limitations of this approach remain unclear. RNase A treatment during chromatin immunoprecipitation (ChIP) reduces chromatin occupancy of the H3K27me3 methyltransferase Polycomb repressive complex 2 (PRC2). This led to suggestions of an "RNA bridge" between PRC2 and chromatin. Here, we show that RNase A treatment during ChIP causes the apparent loss of all facultative heterochromatin, including both PRC2 and H3K27me3 genome-wide. We track this observation to a gain of DNA from non-targeted chromatin, sequenced at the expense of DNA from facultative heterochromatin, which reduces ChIP signals. Our results emphasize substantial limitations in using RNase A treatment for mapping RNA-dependent chromatin occupancy and invalidate conclusions that were previously established for PRC2 based on this assay. [Display omitted] • RNA degradation during ChIP-seq is insufficient to displace PRC2 from chromatin • RNA degradation leads to the artificial depletion of ChIP-seq signals in multiple cell lines • Artificially reduced ChIP-seq signals are explained by a gain of non-targeted DNA • RNA is critical in maintaining the solubility of chromatin during experimentation Healy et al. demonstrate that RNase A treatment of chromatin during immunoprecipitation leads to the artificial loss of all facultative heterochromatin. This observation can be explained by a gain in non-targeted chromatin that is sequenced at the expense of DNA from facultative heterochromatin, which reduces ChIP signals. [ABSTRACT FROM AUTHOR]
- Published
- 2024
- Full Text
- View/download PDF
6. Allele-Specific DNA Methylation and Its Interplay with Repressive Histone Marks at Promoter-Mutant TERT Genes
- Author
-
Josh Lewis Stern, Richard D. Paucek, Franklin W. Huang, Mahmoud Ghandi, Ronald Nwumeh, James C. Costello, and Thomas R. Cech
- Subjects
telomerase ,TERT promoter ,Polycomb repressive complex 2 ,PRC2 ,5-methylcytosine ,allele-specific ,monoallelic ,CpG island ,cancer ,Biology (General) ,QH301-705.5 - Abstract
A mutation in the promoter of the Telomerase Reverse Transcriptase (TERT) gene is the most frequent noncoding mutation in cancer. The mutation drives unusual monoallelic expression of TERT, allowing immortalization. Here, we find that DNA methylation of the TERT CpG island (CGI) is also allele-specific in multiple cancers. The expressed allele is hypomethylated, which is opposite to cancers without TERT promoter mutations. The continued presence of Polycomb repressive complex 2 (PRC2) on the inactive allele suggests that histone marks of repressed chromatin may be causally linked to high DNA methylation. Consistent with this hypothesis, TERT promoter DNA containing 5-methyl-CpG has much increased affinity for PRC2 in vitro. Thus, CpG methylation and histone marks appear to collaborate to maintain the two TERT alleles in different epigenetic states in TERT promoter mutant cancers. Finally, in several cancers, DNA methylation levels at the TERT CGI correlate with altered patient survival.
- Published
- 2017
- Full Text
- View/download PDF
7. UTX/KDM6A Loss Enhances the Malignant Phenotype of Multiple Myeloma and Sensitizes Cells to EZH2 inhibition
- Author
-
Teresa Ezponda, Daphné Dupéré-Richer, Christine M. Will, Eliza C. Small, Nobish Varghese, Tej Patel, Behnam Nabet, Relja Popovic, Jon Oyer, Marinka Bulic, Yupeng Zheng, Xiaoxiao Huang, Mrinal Y. Shah, Sayantan Maji, Alberto Riva, Manuela Occhionorelli, Giovanni Tonon, Neil Kelleher, Jonathan Keats, and Jonathan D. Licht
- Subjects
multiple myeloma ,UTX ,KDM6A ,EZH2 inhibitors ,IRF4 ,BCL6 ,PRC2 ,H3K27me3 ,epigenetic regulator ,Biology (General) ,QH301-705.5 - Abstract
Loss or inactivation of the histone H3K27 demethylase UTX occurs in several malignancies, including multiple myeloma (MM). Using an isogenic cell system, we found that loss of UTX leads to deactivation of gene expression ultimately promoting the proliferation, clonogenicity, adhesion, and tumorigenicity of MM cells. Moreover, UTX mutant cells showed increased in vitro and in vivo sensitivity to inhibition of EZH2, a histone methyltransferase that generates H3K27me3. Such sensitivity was related to a decrease in the levels of IRF4 and c-MYC and an activation of repressors of IRF4 characteristic of germinal center B cells such as BCL6 and IRF1. Rebalance of H3K27me3 levels at specific genes through EZH2 inhibitors may be a therapeutic strategy in MM cases harboring UTX mutations.
- Published
- 2017
- Full Text
- View/download PDF
8. The Histone Methyltransferase Ezh2 Controls Mechanisms of Adaptive Resistance to Tumor Immunotherapy
- Author
-
Daniel Zingg, Natalia Arenas-Ramirez, Dilara Sahin, Rodney A. Rosalia, Ana T. Antunes, Jessica Haeusel, Lukas Sommer, and Onur Boyman
- Subjects
immunotherapy ,IL-2 complexes ,anti-CTLA-4 ,anti-PD-1 ,tumor resistance ,tumor immune escape ,melanoma ,epigenetics ,EZH2 ,PRC2 ,Biology (General) ,QH301-705.5 - Abstract
Immunotherapy and particularly immune checkpoint inhibitors have resulted in remarkable clinical responses in patients with immunogenic tumors, although most cancers develop resistance to immunotherapy. The molecular mechanisms of tumor resistance to immunotherapy remain poorly understood. We now show that induction of the histone methyltransferase Ezh2 controls several tumor cell-intrinsic and extrinsic resistance mechanisms. Notably, T cell infiltration selectively correlated with high EZH2-PRC2 complex activity in human skin cutaneous melanoma. During anti-CTLA-4 or IL-2 immunotherapy in mice, intratumoral tumor necrosis factor-α (TNF-α) production and T cell accumulation resulted in increased Ezh2 expression in melanoma cells, which in turn silenced their own immunogenicity and antigen presentation. Ezh2 inactivation reversed this resistance and synergized with anti-CTLA-4 and IL-2 immunotherapy to suppress melanoma growth. These anti-tumor effects depended on intratumorally accumulating interferon-γ (IFN-γ)-producing PD-1low CD8+ T cells and PD-L1 downregulation on melanoma cells. Hence, Ezh2 serves as a molecular switch controlling melanoma escape during T cell-targeting immunotherapies.
- Published
- 2017
- Full Text
- View/download PDF
9. Trimethylation and Acetylation of β-Catenin at Lysine 49 Represent Key Elements in ESC Pluripotency
- Author
-
Katrin Hoffmeyer, Dirk Junghans, Benoit Kanzler, and Rolf Kemler
- Subjects
Wnt signaling ,mouse embryonic stem cells ,β-catenin ,methylation ,PRC2 ,gene repression ,Biology (General) ,QH301-705.5 - Abstract
Wnt/β-catenin signaling is required for embryonic stem cell (ESC) pluripotency by inducing mesodermal differentiation and inhibiting neuronal differentiation; however, how β-catenin counter-regulates these differentiation pathways is unknown. Here, we show that lysine 49 (K49) of β-catenin is trimethylated (β-catMe3) by Ezh2 or acetylated (β-catAc) by Cbp. Significantly, β-catMe3 acts as a transcriptional co-repressor of the neuronal differentiation genes sox1 and sox3, whereas β-catAc acts as a transcriptional co-activator of the key mesodermal differentiation gene t-brachyury (t-bra). Furthermore, β-catMe3 and β-catAc are alternatively enriched on repressed or activated genes, respectively, during ESC and adult stem cell differentiation into neuronal or mesodermal progenitor cell lineages. Importantly, expression of a β-catenin K49A mutant results in major defects in ESC differentiation. We conclude that β-catenin K49 trimethylation and acetylation are key elements in regulating ESC pluripotency and differentiation potential.
- Published
- 2017
- Full Text
- View/download PDF
10. Inactivation of Ezh2 Upregulates Gfi1 and Drives Aggressive Myc-Driven Group 3 Medulloblastoma
- Author
-
BaoHan T. Vo, Chunliang Li, Marc A. Morgan, Ilan Theurillat, David Finkelstein, Shaela Wright, Judith Hyle, Stephanie M.C. Smith, Yiping Fan, Yong-Dong Wang, Gang Wu, Brent A. Orr, Paul A. Northcott, Ali Shilatifard, Charles J. Sherr, and Martine F. Roussel
- Subjects
group 3 medulloblastoma ,MYC ,polycomb-repressive complex 2 ,PRC2 ,enhancer of zeste homology 2 ,EZH2 ,suppressor of zeste 12 homolog ,SUZ12 ,growth factor independent 1 ,GFI1 ,histone H3 modification ,Hox genes ,epigenetic repression ,Biology (General) ,QH301-705.5 - Abstract
The most aggressive of four medulloblastoma (MB) subgroups are cMyc-driven group 3 (G3) tumors, some of which overexpress EZH2, the histone H3K27 mono-, di-, and trimethylase of polycomb-repressive complex 2. Ezh2 has a context-dependent role in different cancers as an oncogene or tumor suppressor and retards tumor progression in a mouse model of G3 MB. Engineered deletions of Ezh2 in G3 MBs by gene editing nucleases accelerated tumorigenesis, whereas Ezh2 re-expression reversed attendant histone modifications and slowed tumor progression. Candidate oncogenic drivers suppressed by Ezh2 included Gfi1, a proto-oncogene frequently activated in human G3 MBs. Gfi1 disruption antagonized the tumor-promoting effects of Ezh2 loss; conversely, Gfi1 overexpression collaborated with Myc to bypass effects of Trp53 inactivation in driving MB progression in primary cerebellar neuronal progenitors. Although negative regulation of Gfi1 by Ezh2 may restrain MB development, Gfi1 activation can bypass these effects.
- Published
- 2017
- Full Text
- View/download PDF
11. UTX/KDM6A Loss Enhances the Malignant Phenotype of Multiple Myeloma and Sensitizes Cells to EZH2 inhibition.
- Author
-
Ezponda, Teresa, Dupéré-Richer, Daphné, Will, Christine M., Small, Eliza C., Varghese, Nobish, Patel, Tej, Nabet, Behnam, Popovic, Relja, Oyer, Jon, Bulic, Marinka, Zheng, Yupeng, Huang, Xiaoxiao, Shah, Mrinal Y., Maji, Sayantan, Riva, Alberto, Occhionorelli, Manuela, Tonon, Giovanni, Kelleher, Neil, Keats, Jonathan, and Licht, Jonathan D.
- Abstract
Summary Loss or inactivation of the histone H3K27 demethylase UTX occurs in several malignancies, including multiple myeloma (MM). Using an isogenic cell system, we found that loss of UTX leads to deactivation of gene expression ultimately promoting the proliferation, clonogenicity, adhesion, and tumorigenicity of MM cells. Moreover, UTX mutant cells showed increased in vitro and in vivo sensitivity to inhibition of EZH2, a histone methyltransferase that generates H3K27me3. Such sensitivity was related to a decrease in the levels of IRF4 and c-MYC and an activation of repressors of IRF4 characteristic of germinal center B cells such as BCL6 and IRF1. Rebalance of H3K27me3 levels at specific genes through EZH2 inhibitors may be a therapeutic strategy in MM cases harboring UTX mutations. [ABSTRACT FROM AUTHOR]
- Published
- 2017
- Full Text
- View/download PDF
12. The Histone Methyltransferase Ezh2 Controls Mechanisms of Adaptive Resistance to Tumor Immunotherapy.
- Author
-
Zingg, Daniel, Arenas-Ramirez, Natalia, Sahin, Dilara, Rosalia, Rodney A., Antunes, Ana T., Haeusel, Jessica, Sommer, Lukas, and Boyman, Onur
- Abstract
Summary Immunotherapy and particularly immune checkpoint inhibitors have resulted in remarkable clinical responses in patients with immunogenic tumors, although most cancers develop resistance to immunotherapy. The molecular mechanisms of tumor resistance to immunotherapy remain poorly understood. We now show that induction of the histone methyltransferase Ezh2 controls several tumor cell-intrinsic and extrinsic resistance mechanisms. Notably, T cell infiltration selectively correlated with high EZH2-PRC2 complex activity in human skin cutaneous melanoma. During anti-CTLA-4 or IL-2 immunotherapy in mice, intratumoral tumor necrosis factor-α (TNF-α) production and T cell accumulation resulted in increased Ezh2 expression in melanoma cells, which in turn silenced their own immunogenicity and antigen presentation. Ezh2 inactivation reversed this resistance and synergized with anti-CTLA-4 and IL-2 immunotherapy to suppress melanoma growth. These anti-tumor effects depended on intratumorally accumulating interferon-γ (IFN-γ)-producing PD-1 low CD8 + T cells and PD-L1 downregulation on melanoma cells. Hence, Ezh2 serves as a molecular switch controlling melanoma escape during T cell-targeting immunotherapies. [ABSTRACT FROM AUTHOR]
- Published
- 2017
- Full Text
- View/download PDF
13. Trimethylation and Acetylation of β-Catenin at Lysine 49 Represent Key Elements in ESC Pluripotency.
- Author
-
Hoffmeyer, Katrin, Junghans, Dirk, Kanzler, Benoit, and Kemler, Rolf
- Abstract
Summary Wnt/β-catenin signaling is required for embryonic stem cell (ESC) pluripotency by inducing mesodermal differentiation and inhibiting neuronal differentiation; however, how β-catenin counter-regulates these differentiation pathways is unknown. Here, we show that lysine 49 (K49) of β-catenin is trimethylated (β-catMe3) by Ezh2 or acetylated (β-catAc) by Cbp. Significantly, β-catMe3 acts as a transcriptional co-repressor of the neuronal differentiation genes sox1 and sox3 , whereas β-catAc acts as a transcriptional co-activator of the key mesodermal differentiation gene t-brachyury ( t-bra ). Furthermore, β-catMe3 and β-catAc are alternatively enriched on repressed or activated genes, respectively, during ESC and adult stem cell differentiation into neuronal or mesodermal progenitor cell lineages. Importantly, expression of a β-catenin K49A mutant results in major defects in ESC differentiation. We conclude that β-catenin K49 trimethylation and acetylation are key elements in regulating ESC pluripotency and differentiation potential. [ABSTRACT FROM AUTHOR]
- Published
- 2017
- Full Text
- View/download PDF
14. Inactivation of Ezh2 Upregulates Gfi1 and Drives Aggressive Myc-Driven Group 3 Medulloblastoma.
- Author
-
Vo, BaoHan T., Li, Chunliang, Morgan, Marc A., Theurillat, Ilan, Finkelstein, David, Wright, Shaela, Hyle, Judith, Smith, Stephanie M.C., Fan, Yiping, Wang, Yong-Dong, Wu, Gang, Orr, Brent A., Northcott, Paul A., Shilatifard, Ali, Sherr, Charles J., and Roussel, Martine F.
- Abstract
Summary The most aggressive of four medulloblastoma (MB) subgroups are cMyc-driven group 3 (G3) tumors, some of which overexpress EZH2, the histone H3K27 mono-, di-, and trimethylase of polycomb-repressive complex 2. Ezh2 has a context-dependent role in different cancers as an oncogene or tumor suppressor and retards tumor progression in a mouse model of G3 MB. Engineered deletions of Ezh2 in G3 MBs by gene editing nucleases accelerated tumorigenesis, whereas Ezh2 re-expression reversed attendant histone modifications and slowed tumor progression. Candidate oncogenic drivers suppressed by Ezh2 included Gfi1, a proto-oncogene frequently activated in human G3 MBs. Gfi1 disruption antagonized the tumor-promoting effects of Ezh2 loss; conversely, Gfi1 overexpression collaborated with Myc to bypass effects of Trp5 3 inactivation in driving MB progression in primary cerebellar neuronal progenitors. Although negative regulation of Gfi1 by Ezh2 may restrain MB development, Gfi1 activation can bypass these effects. [ABSTRACT FROM AUTHOR]
- Published
- 2017
- Full Text
- View/download PDF
15. Loss of Ezh2 function remodels the DNA replication initiation landscape.
- Author
-
Prorok, Paulina, Forouzanfar, Faezeh, Murugarren, Nerea, Peiffer, Isabelle, Charton, Romain, Akerman, Ildem, and Méchali, Marcel
- Abstract
In metazoan cells, DNA replication initiates from thousands of genomic loci scattered throughout the genome called DNA replication origins. Origins are strongly associated with euchromatin, particularly open genomic regions such as promoters and enhancers. However, over a third of transcriptionally silent genes are associated with DNA replication initiation. Most of these genes are bound and repressed by the Polycomb repressive complex-2 (PRC2) through the repressive H3K27me3 mark. This is the strongest overlap observed for a chromatin regulator with replication origin activity. Here, we asked whether Polycomb-mediated gene repression is functionally involved in recruiting DNA replication origins to transcriptionally silent genes. We show that the absence of EZH2, the catalytic subunit of PRC2, results in increased DNA replication initiation, specifically in the vicinity of EZH2 binding sites. The increase in DNA replication initiation does not correlate with transcriptional de-repression or the acquisition of activating histone marks but does correlate with loss of H3K27me3 from bivalent promoters. [Display omitted] • Polycomb-repressed promoters are hotspots for DNA replication initiation • EZH2
−/− cells display a substantial increase in replication initiation at repressed promoters • Increased replication initiation does not correlate with de novo H3K27ac or transcription • Instead, replication initiates at sites of H3K27me3 loss and increased open chromatin Most promoters repressed by the Polycomb group of proteins also harbor DNA replication initiation in pluripotent stem cells. Prorok et al. find that EZH2 loss of function leads to marked increase in DNA replication initiation at these repressed promoters with loss of H3K27me3, irrespective of the activating histone H3K27ac mark or transcription. [ABSTRACT FROM AUTHOR]- Published
- 2023
- Full Text
- View/download PDF
16. Jarid2 promotes temporal progression of retinal progenitors via repression of Foxp1.
- Author
-
Zhang, Jianmin, Roberts, Jacqueline M., Chang, Fei, Schwakopf, Joon, and Vetter, Monica L.
- Abstract
Transitions in competence underlie the ability of CNS progenitors to generate a diversity of neurons and glia. Retinal progenitor cells in mouse generate early-born cell types embryonically and late-born cell types largely postnatally. We find that the transition from early to late progenitor competence is regulated by Jarid2. Loss of Jarid2 results in extended production of early cell types and extended expression of early progenitor genes. Jarid2 can regulate histone modifications, and we find reduction of repressive mark H3K27me3 on a subset of early progenitor genes with loss of Jarid2 , most notably Foxp1. We show that Foxp1 regulates the competence to generate early-born retinal cell types, promotes early and represses late progenitor gene expression, and is required for extending early retinal cell production after loss of Jarid2. We conclude that Jarid2 facilitates progression of retinal progenitor temporal identity by repressing Foxp1 , which is a primary regulator of early temporal patterning. [Display omitted] • Loss of Jarid2 delays retinal progenitor early- to late-identity transition • Jarid2 mediates H3K27me3 deposition and repression of early genes including Foxp1 • Foxp1 regulates the timing of early retinal cell production • Foxp1 is an effector of Jarid2 -mediated temporal patterning Temporal patterning of neural progenitors is essential to generate neuronal diversity. Zhang et al. find that Jarid2 facilitates retinal progenitor temporal identity transition via H3K27me3 deposition and repression of early genes, most notably Foxp1. They find that Foxp1 is an effector of Jarid2 in regulating timing of retinal neurogenesis. [ABSTRACT FROM AUTHOR]
- Published
- 2023
- Full Text
- View/download PDF
17. A Functional Link between Nuclear RNA Decay and Transcriptional Control Mediated by the Polycomb Repressive Complex 2
- Author
-
Kristoffer Vitting-Seerup, Mengjun Wu, Torben Heick Jensen, Aliaksandra Radzisheuskaya, Leonor Rib, Albin Sandelin, Kristian Helin, Itys Comet, William A. Garland, and Marta Lloret-Llinares
- Subjects
CHROMATIN ,0301 basic medicine ,PAXT ,Transcription, Genetic ,Exosome complex ,RNA Stability ,RNA exosome ,H3K27 methylation ,PROTEIN EED ,Mice ,chemistry.chemical_compound ,0302 clinical medicine ,Transcriptional regulation ,DNA METHYLATION ,lcsh:QH301-705.5 ,GENE-EXPRESSION ,Epigenomics ,Mice, Knockout ,biology ,Chromatin/genetics ,Polycomb Repressive Complex 2 ,Mouse Embryonic Stem Cells ,nuclear RNA decay ,PRC2 ,Chromatin ,READ ALIGNMENT ,Cell biology ,stem cell regulation ,RNA, Nuclear/genetics ,Polycomb Repressive Complex 2/genetics ,Transcription Factors/genetics ,macromolecular substances ,EXOSOME ,PLURIPOTENCY ,Article ,General Biochemistry, Genetics and Molecular Biology ,03 medical and health sciences ,Downregulation and upregulation ,Animals ,Mouse Embryonic Stem Cells/cytology ,RNA, Nuclear ,DEVELOPMENTAL REGULATORS ,RNA ,030104 developmental biology ,chemistry ,lcsh:Biology (General) ,DIFFERENTIAL EXPRESSION ANALYSIS ,biology.protein ,030217 neurology & neurosurgery ,DNA ,Transcription Factors - Abstract
Summary Pluripotent embryonic stem cells (ESCs) constitute an essential cellular niche sustained by epigenomic and transcriptional regulation. Any role of post-transcriptional processes remains less explored. Here, we identify a link between nuclear RNA levels, regulated by the poly(A) RNA exosome targeting (PAXT) connection, and transcriptional control by the polycomb repressive complex 2 (PRC2). Knockout of the PAXT component ZFC3H1 impairs mouse ESC differentiation. In addition to the upregulation of bona fide PAXT substrates, Zfc3h1−/− cells abnormally express developmental genes usually repressed by PRC2. Such de-repression is paralleled by decreased PRC2 binding to chromatin and low PRC2-directed H3K27 methylation. PRC2 complex stability is compromised in Zfc3h1−/− cells with elevated levels of unspecific RNA bound to PRC2 components. We propose that excess RNA hampers PRC2 function through its sequestration from DNA. Our results highlight the importance of balancing nuclear RNA levels and demonstrate the capacity of bulk RNA to regulate chromatin-associated proteins., Graphical Abstract, Highlights • Depletion of ZFC3H1 in mouse ESCs results in differentiation defects • PRC2 target genes are deregulated in Zfc3h1−/− cells • Chromatin binding of PRC2 and H3K27me3 is reduced in Zfc3h1−/− cells • Increased binding of RNA impairs PRC2 complex stability, ZFC3H1 targets pA+ RNA for decay by the nuclear RNA exosome. Garland et al. report a disruptive relationship between excess RNA and PRC2 upon depletion of ZFC3H1 in mouse ESCs. In such conditions, RNA is bound by PRC2 components, which show reduced binding to chromatin and fellow PRC2 proteins.
- Published
- 2019
- Full Text
- View/download PDF
18. Human telomerase is directly regulated by non-telomeric TRF2-G-quadruplex interaction
- Author
-
Shantanu Chowdhury, Antara Sengupta, Silje Lier, Meenakshi Verma, Manish Kumar, Shalu Sharma, Deo Prakash Pandey, Shuvra Shekhar Roy, Ananda Kishore Mukherjee, Gaute Nesse, and Sulochana Bagri
- Subjects
0301 basic medicine ,Telomerase ,Somatic cell ,QH301-705.5 ,cells ,G-quadruplex ,telomerase ,General Biochemistry, Genetics and Molecular Biology ,Article ,03 medical and health sciences ,0302 clinical medicine ,Humans ,cancer ,Neoplastic transformation ,Telomerase reverse transcriptase ,Biology (General) ,neoplasms ,telomere repeat binding factor ,G-quadruplexe ,biology ,EZH2 ,Telomere ,G-Quadruplexes ,shelterin proteins ,enzymes and coenzymes (carbohydrates) ,030104 developmental biology ,hTERT promoter mutations ,embryonic structures ,biology.protein ,Cancer research ,biological phenomena, cell phenomena, and immunity ,PRC2 ,030217 neurology & neurosurgery - Abstract
Summary Human telomerase reverse transcriptase (hTERT) remains suppressed in most normal somatic cells. Resulting erosion of telomeres leads eventually to replicative senescence. Reactivation of hTERT maintains telomeres and triggers progression of >90% of cancers. However, any direct causal link between telomeres and telomerase regulation remains unclear. Here, we show that the telomere-repeat-binding-factor 2 (TRF2) binds hTERT promoter G-quadruplexes and recruits the polycomb-repressor EZH2/PRC2 complex. This is causal for H3K27 trimethylation at the hTERT promoter and represses hTERT in cancer as well as normal cells. Two highly recurrent hTERT promoter mutations found in many cancers, including ~83% glioblastoma multiforme, that are known to destabilize hTERT promoter G-quadruplexes, showed loss of TRF2 binding in patient-derived primary glioblastoma multiforme cells. Ligand-induced G-quadruplex stabilization restored TRF2 binding, H3K27-trimethylation, and hTERT re-suppression. These results uncover a mechanism of hTERT regulation through a telomeric factor, implicating telomere-telomerase molecular links important in neoplastic transformation, aging, and regenerative therapy., Graphic abstractSchematic representation of sample collection and analysis. The figure was created using BioRender.com
- Published
- 2021
19. Single-cell sequencing reveals activation of core transcription factors in PRC2-deficient malignant peripheral nerve sheath tumor.
- Author
-
Zhang, Xiyuan, Lou, Hannah E., Gopalan, Vishaka, Liu, Zhihui, Jafarah, Hilda M., Lei, Haiyan, Jones, Paige, Sayers, Carly M., Yohe, Marielle E., Chittiboina, Prashant, Widemann, Brigitte C., Thiele, Carol J., Kelly, Michael C., Hannenhalli, Sridhar, and Shern, Jack F.
- Abstract
Loss-of-function mutations in the polycomb repressive complex 2 (PRC2) occur frequently in malignant peripheral nerve sheath tumor, an aggressive sarcoma that arises from NF1 -deficient Schwann cells. To define the oncogenic mechanisms underlying PRC2 loss, we use engineered cells that dynamically reassemble a competent PRC2 coupled with single-cell sequencing from clinical samples. We discover a two-pronged oncogenic process: first, PRC2 loss leads to remodeling of the bivalent chromatin and enhancer landscape, causing the upregulation of developmentally regulated transcription factors that enforce a transcriptional circuit serving as the cell's core vulnerability. Second, PRC2 loss reduces type I interferon signaling and antigen presentation as downstream consequences of hyperactivated Ras and its cross talk with STAT/IRF transcription factors. Mapping of the transcriptional program of these PRC2-deficient tumor cells onto a constructed developmental trajectory of normal Schwann cells reveals that changes induced by PRC2 loss enforce a cellular profile characteristic of a primitive mesenchymal neural crest stem cell. [Display omitted] • PRC2 loss activates bivalent genes via transcriptional recruitment of active enhancers • PRC2 loss activates core TFs required to sustain the MPNST oncogenic program • PRC2-null MPNSTs mimic Schwann cell progenitors and reduce antigen presentation • scRNA-seq reveals heterogeneity within the MPNST microenvironment Zhang et al. provide evidence that PRC2 loss activates cell-fate-determining transcription factors by recruiting active enhancers and dampens type I interferon signaling and antigen presentation through transcriptional cross talk with the hyperactivated Ras. These observations are supported by integrative analysis of single-cell sequencing of patient MPNST samples. [ABSTRACT FROM AUTHOR]
- Published
- 2022
- Full Text
- View/download PDF
20. A TET1-PSPC1-Neat1 molecular axis modulates PRC2 functions in controlling stem cell bivalency.
- Author
-
Huang, Xin, Bashkenova, Nazym, Hong, Yantao, Lyu, Cong, Guallar, Diana, Hu, Zhe, Malik, Vikas, Li, Dan, Wang, Hailin, Shen, Xiaohua, Zhou, Hongwei, and Wang, Jianlong
- Abstract
TET1 maintains hypomethylation at bivalent promoters through its catalytic activity in embryonic stem cells (ESCs). However, TET1 catalytic activity-independent function in regulating bivalent genes is not well understood. Using a proteomics approach, we map the TET1 interactome in ESCs and identify PSPC1 as a TET1 partner. Genome-wide location analysis reveals that PSPC1 functionally associates with TET1 and Polycomb repressive complex-2 (PRC2). We establish that PSPC1 and TET1 repress, and the lncRNA Neat1 activates, bivalent gene expression. In ESCs, Neat1 is preferentially bound to PSPC1 alongside its PRC2 association at bivalent promoters. During the ESC-to-epiblast-like stem cell (EpiLC) transition, PSPC1 and TET1 maintain PRC2 chromatin occupancy at bivalent gene promoters, while Neat1 facilitates the activation of certain bivalent genes by promoting PRC2 binding to their mRNAs. Our study demonstrates a TET1-PSPC1- Neat1 molecular axis that modulates PRC2-binding affinity to chromatin and bivalent gene transcripts in controlling stem cell bivalency. [Display omitted] • The TET1 interactome identifies PSPC1 as a partner of TET1 in ESCs • PSPC1 interacts with TET1 and PRC2 for bivalency control in formative pluripotency • TET1 and PSPC1 repress bivalent genes by promoting PRC2 chromatin occupancy • Neat1 facilitates bivalent gene activation by promoting PRC2 binding to their mRNAs Huang et al. use proteomics and genetic approaches to show that catalytic activity-independent functions of TET1, coordinated with the paraspeckle components PSPC1 and its cognate lncRNA Neat1 , dynamically regulate stem cell bivalency by modulating PRC2 binding to chromatin and bivalent gene transcripts in the naive-to-formative pluripotent state transition. [ABSTRACT FROM AUTHOR]
- Published
- 2022
- Full Text
- View/download PDF
21. dCas9 fusion to computer-designed PRC2 inhibitor reveals functional TATA box in distal promoter region.
- Author
-
Levy, Shiri, Somasundaram, Logeshwaran, Raj, Infencia Xavier, Ic-Mex, Diego, Phal, Ashish, Schmidt, Sven, Ng, Weng I., Mar, Daniel, Decarreau, Justin, Moss, Nicholas, Alghadeer, Ammar, Honkanen, Henrik, Sarthy, Jay, Vitanza, Nicholas, Hawkins, R. David, Mathieu, Julie, Wang, Yuliang, Baker, David, Bomsztyk, Karol, and Ruohola-Baker, Hannele
- Abstract
Bifurcation of cellular fates, a critical process in development, requires histone 3 lysine 27 methylation (H3K27me3) marks propagated by the polycomb repressive complex 2 (PRC2). However, precise chromatin loci of functional H3K27me3 marks are not yet known. Here, we identify critical PRC2 functional sites at high resolution. We fused a computationally designed protein, EED binder (EB), which competes with EZH2 and thereby inhibits PRC2 function, to dCas9 (EBdCas9) to allow for PRC2 inhibition at a precise locus using gRNA. Targeting EBdCas9 to four different genes (TBX18 , p16 , CDX2, and GATA3) results in precise H3K27me3 and EZH2 reduction, gene activation, and functional outcomes in the cell cycle (p16) or trophoblast transdifferentiation (CDX2 and GATA3). In the case of TBX18, we identify a PRC2-controlled, functional TATA box >500 bp upstream of the TBX18 transcription start site (TSS) using EBdCas9. Deletion of this TATA box eliminates EBdCas9-dependent TATA binding protein (TBP) recruitment and transcriptional activation. EBdCas9 technology may provide a broadly applicable tool for epigenomic control of gene regulation. [Display omitted] • EBdCas9 inhibits PRC2 function in precise genomic locations • EBdCas9 upregulates TBX18 , p16 , CDX2 , and GATA3 by de-repression • EBdCas9 identifies PRC2-controlled, active TATA box >500 bp upstream of TBX18 TSS • EBdCas9 is sufficient to induce transdifferentiation and repress cancer cell cycle Levy et al. fused a computationally designed protein, EED binder (EB), which competes with EZH2 and thereby inhibits PRC2 function, to dCas9 (EBdCas9). EBdCas9 represses PRC2 action in precise loci, remodels epigenomic marks, exposes transcriptional elements, and induces transdifferentiation. [ABSTRACT FROM AUTHOR]
- Published
- 2022
- Full Text
- View/download PDF
22. Reprogramming of bivalent chromatin states in NRAS mutant melanoma suggests PRC2 inhibition as a therapeutic strategy
- Author
-
Khalida Wani, Jonathan Schulz, Scott E. Woodman, Alexander J. Lazar, Junna Oba, Kaifu Chen, Sharmistha Sarkar, Samir B. Amin, Praveen Barrodia, Ayush T. Raman, Katarzyna Tomczak, Chang-Jiun Wu, Anand K Singh, Caitlin Creasy, Kunal Rai, Rossana Lazcano, Samia Khan, Dongyu Zhao, Elias Orouji, Ming Tang, Lauren E. Haydu, Wei-Lien Wang, Christopher Terranova, Chantale Bernatchez, Mayinuer Maitituoheti, and Archit K. Ghosh
- Subjects
Neuroblastoma RAS viral oncogene homolog ,Transcription, Genetic ,Mice, Nude ,Article ,General Biochemistry, Genetics and Molecular Biology ,GTP Phosphohydrolases ,Histones ,Mesoderm ,Histone H3 ,Cell Line, Tumor ,Animals ,Humans ,Enhancer of Zeste Homolog 2 Protein ,Epigenetics ,Neoplasm Metastasis ,Melanoma ,Cell Proliferation ,Mitogen-Activated Protein Kinase Kinases ,biology ,EZH2 ,Polycomb Repressive Complex 2 ,Membrane Proteins ,Chromatin ,Tumor Burden ,Mutation ,Cancer research ,biology.protein ,Melanocytes ,Female ,PRC2 ,Reprogramming ,Bivalent chromatin - Abstract
SUMMARY The dynamic evolution of chromatin state patterns during metastasis, their relationship with bona fide genetic drivers, and their therapeutic vulnerabilities are not completely understood. Combinatorial chromatin state profiling of 46 melanoma samples reveals an association of NRAS mutants with bivalent histone H3 lysine 27 trimethylation (H3K27me3) and Polycomb repressive complex 2. Reprogramming of bivalent domains during metastasis occurs on master transcription factors of a mesenchymal phenotype, including ZEB1, TWIST1, and CDH1. Resolution of bivalency using pharmacological inhibition of EZH2 decreases invasive capacity of melanoma cells and markedly reduces tumor burden in vivo, specifically in NRAS mutants. Coincident with bivalent reprogramming, the increased expression of pro-metastatic and melanocyte-specific cell-identity genes is associated with exceptionally wide H3K4me3 domains, suggesting a role for this epigenetic element. Overall, we demonstrate that reprogramming of bivalent and broad domains represents key epigenetic alterations in metastatic melanoma and that EZH2 plus MEK inhibition may provide a promising therapeutic strategy for NRAS mutant melanoma patients., Graphical abstract, In brief Terranova et al. provide a comprehensive epigenome resource for melanoma encompassing 284 chromatin maps. They find key regulatory roles for bivalent and broad domains in expression of pro-metastatic genes and identify EZH2 plus MEK inhibition as a therapeutic strategy for NRAS mutant melanomas.
- Published
- 2021
- Full Text
- View/download PDF
23. H3K27M in Gliomas Causes a One-Step Decrease in H3K27 Methylation and Reduced Spreading within the Constraints of H3K36 Methylation
- Author
-
Hamid Nikbakht, Tianyuan Lu, Dylan M. Marchione, Brian Krug, Mariel Coradin, Benjamin A. Garcia, Nada Jabado, Ashot S. Harutyunyan, Caterina Russo, Eric Bareke, Jacek Majewski, Cynthia Horth, and Haifen Chen
- Subjects
computational modeling ,Epigenomics ,0301 basic medicine ,Gene Expression ,Biology ,medicine.disease_cause ,Methylation ,Article ,General Biochemistry, Genetics and Molecular Biology ,Histones ,03 medical and health sciences ,Methionine ,0302 clinical medicine ,Cell Line, Tumor ,Glioma ,Histone methylation ,medicine ,Humans ,Gene silencing ,histone methylation ,lcsh:QH301-705.5 ,Mutation ,H3.3K27M ,Lysine ,fungi ,Polycomb Repressive Complex 2 ,Epigenome ,DNA Methylation ,medicine.disease ,Chromatin ,Gene Expression Regulation, Neoplastic ,030104 developmental biology ,lcsh:Biology (General) ,pediatric high-grade glioma ,Cancer research ,biology.protein ,PRC2 ,Protein Processing, Post-Translational ,030217 neurology & neurosurgery - Abstract
SUMMARY The discovery of H3K27M mutations in pediatric gliomas marked a new chapter in cancer epigenomics. Numerous studies have investigated the effect of this mutation on H3K27 trimethylation, but only recently have we started to realize its additional effects on the epigenome. Here, we use isogenic glioma H3K27M+/− cell lines to investigate H3K27 methylation and its interaction with H3K36 and H3K9 modifications. We describe a “step down” effect of H3K27M on the distribution of H3K27 methylation: me3 is reduced to me2, me2 is reduced to me1, whereas H3K36me2/3 delineates the boundaries for the spread of H3K27me marks. We also observe a replacement of H3K27me2/3 silencing by H3K9me3. Using a computational simulation, we explain our observations by reduced effectiveness of PRC2 and constraints imposed on the deposition of H3K27me by antagonistic H3K36 modifications. Our work further elucidates the effects of H3K27M in gliomas as well as the general principles of deposition in H3K27 methylation., Graphical Abstract, In Brief Harutyunyan et al. use isogenic glioma H3K27M+/− cell lines to demonstrate the rewiring of the epigenome, specifically H3K27me1/2/3, H3K36me2/3, and H3K9me3. The dynamic deposition of histone marks is simulated by a stochastic model. This work further advances the understanding of the deposition of H3K27 methylation in H3K27M mutant gliomas.
- Published
- 2020
- Full Text
- View/download PDF
24. PRC2 Acts as a Critical Timer That Drives Oligodendrocyte Fate over Astrocyte Identity by Repressing the Notch Pathway
- Author
-
Younjung Park, Hyeyoung Cho, Seung-Hee Lee, Wenxian Wang, Michael McCane, Yungki Park, Sangsoo Kim, Sue A. Aicher, Dongkyeong Kim, Ben Emery, Jae Woon Lee, Soo Kyung Lee, Sung Min Yoon, Ji Hwan Moon, and Su Jeong Lim
- Subjects
0301 basic medicine ,Lineage (genetic) ,Notch signaling pathway ,macromolecular substances ,Methylation ,Article ,General Biochemistry, Genetics and Molecular Biology ,Histones ,Mice ,03 medical and health sciences ,Histone H3 ,0302 clinical medicine ,medicine ,Animals ,Cell Lineage ,Enhancer of Zeste Homolog 2 Protein ,Wnt Signaling Pathway ,Psychological repression ,Myelin Sheath ,Receptors, Notch ,biology ,Lysine ,Stem Cells ,Polycomb Repressive Complex 2 ,Wnt signaling pathway ,Cell Differentiation ,Oligodendrocyte ,Cell biology ,NFI Transcription Factors ,Oligodendroglia ,030104 developmental biology ,medicine.anatomical_structure ,Spinal Cord ,Astrocytes ,biology.protein ,PRC2 ,Chickens ,030217 neurology & neurosurgery ,Signal Transduction ,Astrocyte - Abstract
SUMMARY PRC2 creates the repressive mark histone H3 Lys27 trimethylation. Although PRC2 is involved in various biological processes, its role in glial development remains ambiguous. Here, we show that PRC2 is required for oligodendrocyte (OL) differentiation and myelination, but not for OL precursor formation. PRC2-deficient OL lineage cells differentiate into OL precursors, but they fail to trigger the molecular program for myelination, highlighting that PRC2 is essential for directing the differentiation timing of OL precursors. PRC2 null OL lineage cells aberrantly induce Notch pathway genes and acquire astrocytic features. The repression of the Notch pathway restores the myelination program and inhibits abnormal astrocytic differentiation in the PRC2-deficient OL lineage, indicating that Notch is a major target of PRC2. Altogether, our studies propose a specific action of PRC2 as a novel gatekeeper that determines the glial fate choice and the timing of OL lineage progression and myelination by impinging on the Notch pathway., In Brief Wang et al. show that the polycomb repressive complex PRC2 is required for the differentiation of oligodendrocyte precursors to myelinating oligodendrocytes. They further show that PRC2 promotes oligodendrocyte differentiation and inhibits erroneous astrocytic fate by repressing the Notch pathway., Graphical Abstract
- Published
- 2020
- Full Text
- View/download PDF
25. Allele-Specific DNA Methylation and Its Interplay with Repressive Histone Marks at Promoter-Mutant TERT Genes
- Author
-
James C. Costello, Franklin W. Huang, Thomas R. Cech, Mahmoud Ghandi, Richard D. Paucek, Ronald Nwumeh, and Josh Lewis Stern
- Subjects
0301 basic medicine ,Transcription, Genetic ,medicine.disease_cause ,telomerase ,Article ,General Biochemistry, Genetics and Molecular Biology ,03 medical and health sciences ,Cell Line, Tumor ,Neoplasms ,medicine ,monoallelic ,Humans ,cancer ,Telomerase reverse transcriptase ,Enhancer of Zeste Homolog 2 Protein ,Epigenetics ,Promoter Regions, Genetic ,5-methylcytosine ,lcsh:QH301-705.5 ,Alleles ,Mutation ,biology ,Base Sequence ,allele-specific ,Polycomb repressive complex 2 ,DNA ,DNA Methylation ,Molecular biology ,Survival Analysis ,PRC2 ,Chromatin ,Histone Code ,030104 developmental biology ,Histone ,CpG site ,lcsh:Biology (General) ,DNA methylation ,biology.protein ,CpG island ,TERT promoter ,CpG Islands ,Protein Binding - Abstract
Summary A mutation in the promoter of the Telomerase Reverse Transcriptase (TERT) gene is the most frequent noncoding mutation in cancer. The mutation drives unusual monoallelic expression of TERT , allowing immortalization. Here, we find that DNA methylation of the TERT CpG island (CGI) is also allele-specific in multiple cancers. The expressed allele is hypomethylated, which is opposite to cancers without TERT promoter mutations. The continued presence of Polycomb repressive complex 2 (PRC2) on the inactive allele suggests that histone marks of repressed chromatin may be causally linked to high DNA methylation. Consistent with this hypothesis, TERT promoter DNA containing 5-methyl-CpG has much increased affinity for PRC2 in vitro . Thus, CpG methylation and histone marks appear to collaborate to maintain the two TERT alleles in different epigenetic states in TERT promoter mutant cancers. Finally, in several cancers, DNA methylation levels at the TERT CGI correlate with altered patient survival.
- Published
- 2020
- Full Text
- View/download PDF
26. Compartmentalization-aided interaction screening reveals extensive high-order complexes within the SARS-CoV-2 proteome.
- Author
-
Xu, Weifan, Pei, Gaofeng, Liu, Hongrui, Ju, Xiaohui, Wang, Jing, Ding, Qiang, and Li, Pilong
- Abstract
Bearing a relatively large single-stranded RNA genome in nature, severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) utilizes sophisticated replication/transcription complexes (RTCs), mainly composed of a network of nonstructural proteins and nucleocapsid protein, to establish efficient infection. In this study, we develop an innovative interaction screening strategy based on phase separation in cellulo , namely compartmentalization of protein-protein interactions in cells (CoPIC). Utilizing CoPIC screening, we map the interaction network among RTC-related viral proteins. We identify a total of 47 binary interactions among 14 proteins governing replication, discontinuous transcription, and translation of coronaviruses. Further exploration via CoPIC leads to the discovery of extensive ternary complexes composed of these components, which infer potential higher-order complexes. Taken together, our results present an efficient and robust interaction screening strategy, and they indicate the existence of a complex interaction network among RTC-related factors, thus opening up opportunities to understand SARS-CoV-2 biology and develop therapeutic interventions for COVID-19. [Display omitted] • CoPIC emerges as a simple and efficient method to investigate PPI in cellulo • CoPIC screening reveals extensive high-order complexes within the SARS-CoV-2 RTC • Profiling intraviral interactions provides targets for therapeutic intervention The unprecedented COVID-19 pandemic is caused by infection with severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2). Xu et al. establish an innovative interaction screening strategy based on phase separation in cellulo and reveal comprehensive high-order complexes within the SARS-CoV-2 proteome, thus providing more effective therapeutic opportunities. [ABSTRACT FROM AUTHOR]
- Published
- 2021
- Full Text
- View/download PDF
27. Non-canonical PRC1.1 Targets Active Genes Independent of H3K27me3 and Is Essential for Leukemogenesis
- Author
-
Marjan Geugien, Jan Jacob Schuringa, Anton C.M. Martens, Annet Z. Brouwers-Vos, Richard W.J. Groen, Aida Rodríguez López, Hendrik G. Stunnenberg, Jennifer Jaques, Joost H.A. Martens, Huipin Yuan, Edo Vellenga, Fabrizia Fusetti, Ana M. Sotoca, Vincent van den Boom, Henny Maat, Hematology laboratory, CCA - Cancer biology, Enzymology, Guided Treatment in Optimal Selected Cancer Patients (GUTS), and Stem Cell Aging Leukemia and Lymphoma (SALL)
- Subjects
0301 basic medicine ,Jumonji Domain-Containing Histone Demethylases ,Cellular differentiation ,Cell ,macromolecular substances ,Biology ,General Biochemistry, Genetics and Molecular Biology ,03 medical and health sciences ,Cancer stem cell ,medicine ,Humans ,H2A UBIQUITYLATION ,GeneralLiterature_REFERENCE(e.g.,dictionaries,encyclopedias,glossaries) ,lcsh:QH301-705.5 ,HISTONE H3 ,Molecular Biology ,PRC2 RECRUITMENT ,Cell Proliferation ,Polycomb Repressive Complex 1 ,Transcriptionally active chromatin ,Genetics ,HEMATOPOIETIC STEM/PROGENITOR CELLS ,Cell growth ,DEVELOPMENTAL REGULATORS ,Cell Differentiation ,Cell Biology ,Embryonic stem cell ,HUMAN CD34(+) CELLS ,Cell biology ,SELF-RENEWAL ,FAMILY-MEMBERS ,030104 developmental biology ,medicine.anatomical_structure ,lcsh:Biology (General) ,biology.protein ,POLYCOMB REPRESSIVE COMPLEX ,Stem cell ,PRC2 ,EMBRYONIC STEM-CELLS - Abstract
Polycomb proteins are classical regulators of stem cell self-renewal and cell lineage commitment and are frequently deregulated in cancer. Here, we find that the non-canonical PRC1.1 complex, as identified by mass-spectrometry-based proteomics, is critically important for human leukemic stem cells. Downmodulation of PRC1.1 complex members, like the DNA-binding subunit KDM2B, strongly reduces cell proliferation in vitro and delays or even abrogates leukemogenesis in vivo in humanized xenograft models. PRC1.1 components are significantly overexpressed in primary AML CD34(+) cells. Besides a set of genes that is targeted by PRC1 and PRC2, ChIP-seq studies show that PRC1.1 also binds a distinct set of genes that are devoid of H3K27me3, suggesting a gene-regulatory role independent of PRC2. This set encompasses genes involved in metabolism, which have transcriptionally active chromatin profiles. These data indicate that PRC1.1 controls specific genes involved in unique cell biological processes required for leukemic cell viability.
- Published
- 2016
- Full Text
- View/download PDF
28. Human telomerase is directly regulated by non-telomeric TRF2-G-quadruplex interaction.
- Author
-
Sharma, Shalu, Mukherjee, Ananda Kishore, Roy, Shuvra Shekhar, Bagri, Sulochana, Lier, Silje, Verma, Meenakshi, Sengupta, Antara, Kumar, Manish, Nesse, Gaute, Pandey, Deo Prakash, and Chowdhury, Shantanu
- Abstract
Human telomerase reverse transcriptase (hTERT) remains suppressed in most normal somatic cells. Resulting erosion of telomeres leads eventually to replicative senescence. Reactivation of hTERT maintains telomeres and triggers progression of >90% of cancers. However, any direct causal link between telomeres and telomerase regulation remains unclear. Here, we show that the telomere-repeat-binding-factor 2 (TRF2) binds hTERT promoter G-quadruplexes and recruits the polycomb-repressor EZH2/PRC2 complex. This is causal for H3K27 trimethylation at the hTERT promoter and represses hTERT in cancer as well as normal cells. Two highly recurrent hTERT promoter mutations found in many cancers, including ∼83% glioblastoma multiforme, that are known to destabilize hTERT promoter G-quadruplexes, showed loss of TRF2 binding in patient-derived primary glioblastoma multiforme cells. Ligand-induced G-quadruplex stabilization restored TRF2 binding, H3K27-trimethylation, and hTERT re-suppression. These results uncover a mechanism of hTERT regulation through a telomeric factor, implicating telomere-telomerase molecular links important in neoplastic transformation, aging, and regenerative therapy. [Display omitted] • Non-telomeric TRF2 suppresses re-activated human telomerase in glioblastoma cells • PRC2 recruitment depends on the TRF2 hTERT-G-quadruplex interaction • Clinically deleterious hTERT promoter mutations disrupt G4-TRF2 association • G4 stabilization reinstates TRF2-induced hTERT repression in patient glioblastoma Sharma et al. show transcriptional repression of hTERT by non-telomeric TRF2. TRF2 binds hTERT promoter G-quadruplex and recruits the REST/PRC2-complex. High-frequency clinical mutations associated with cancer destabilize hTERT promoter G-quadruplex, compromise TRF2 binding, and result in hTERT re-activation. Stabilization of G-quadruplex reinstates TRF2 binding and hTERT re-suppression in patient-derived glioblastoma cells. [ABSTRACT FROM AUTHOR]
- Published
- 2021
- Full Text
- View/download PDF
29. Polycomb- and Methylation-Independent Roles of EZH2 as a Transcription Activator
- Author
-
Qi Cao, Ka Wing Fong, Jindan Yu, Xiaodong Lu, Jonathan D. Licht, Bing Song, Jung Kim, Jonathan C. Zhao, and Yongik Lee
- Subjects
0301 basic medicine ,Male ,Transcription, Genetic ,Carcinogenesis ,macromolecular substances ,Methylation ,General Biochemistry, Genetics and Molecular Biology ,Article ,03 medical and health sciences ,Histone H3 ,Mice, Inbred NOD ,Cell Line, Tumor ,Gene silencing ,Animals ,Humans ,Enhancer of Zeste Homolog 2 Protein ,RNA, Messenger ,Enhancer ,lcsh:QH301-705.5 ,Cell Proliferation ,Regulation of gene expression ,biology ,Base Sequence ,Chemistry ,EZH2 ,Polycomb Repressive Complex 2 ,Prostatic Neoplasms ,Promoter ,3. Good health ,Cell biology ,Androgen receptor ,Gene Expression Regulation, Neoplastic ,030104 developmental biology ,HEK293 Cells ,lcsh:Biology (General) ,Receptors, Androgen ,biology.protein ,Androgens ,Histone Methyltransferases ,Trans-Activators ,PRC2 ,Protein Binding ,Signal Transduction - Abstract
SUMMARY Enhancer of Zeste 2 (EZH2) is the enzymatic subunit of Polycomb Repressive Complex 2 (PRC2), which catalyzes histone H3 lysine 27 trimethylation (H3K27me3) at target promoters for gene silencing. Here, we report that EZH2 activates androgen receptor (AR) gene transcription through direct occupancy at its promoter. Importantly, this activating role of EZH2 is independent of PRC2 and its methyltransferase activities. Genome-wide assays revealed extensive EZH2 occupancy at promoters marked by either H3K27ac or H3K27me3, leading to gene activation or repression, respectively. Last, we demonstrate enhanced efficacy of enzymatic EZH2 inhibitors when used in combination with AR antagonists in blocking the dual roles of EZH2 and suppressing prostate cancer progression in vitro and in vivo. Taken together, our study reports EZH2 as a transcriptional activator, a key target of which is AR, and suggests a drug-combinatory approach to treat advanced prostate cancer., Graphical Abstract, In Brief Kim et al. report EZH2 as a transcriptional activator that directly induces AR gene expression in a Polycomb- and methylation-independent manner, providing a mechanism to escape enzymatic EZH2 inhibitors. Combination of inhibitors with AR-targeted therapies showed a strong synergy in blocking the EZH2 downstream pathways and suppressing prostate cancer progression.
- Published
- 2018
30. PRC2 Acts as a Critical Timer That Drives Oligodendrocyte Fate over Astrocyte Identity by Repressing the Notch Pathway.
- Author
-
Wang, Wenxian, Cho, Hyeyoung, Kim, Dongkyeong, Park, Younjung, Moon, Ji Hwan, Lim, Su Jeong, Yoon, Sung Min, McCane, Michael, Aicher, Sue A., Kim, Sangsoo, Emery, Ben, Lee, Jae W., Lee, Seunghee, Park, Yungki, and Lee, Soo-Kyung
- Abstract
PRC2 creates the repressive mark histone H3 Lys27 trimethylation. Although PRC2 is involved in various biological processes, its role in glial development remains ambiguous. Here, we show that PRC2 is required for oligodendrocyte (OL) differentiation and myelination, but not for OL precursor formation. PRC2-deficient OL lineage cells differentiate into OL precursors, but they fail to trigger the molecular program for myelination, highlighting that PRC2 is essential for directing the differentiation timing of OL precursors. PRC2 null OL lineage cells aberrantly induce Notch pathway genes and acquire astrocytic features. The repression of the Notch pathway restores the myelination program and inhibits abnormal astrocytic differentiation in the PRC2-deficient OL lineage, indicating that Notch is a major target of PRC2. Altogether, our studies propose a specific action of PRC2 as a novel gatekeeper that determines the glial fate choice and the timing of OL lineage progression and myelination by impinging on the Notch pathway. • PRC2 null oligodendrocyte (OL) lineage cells fail to differentiate to OL • PRC2 null OL lineage cells aberrantly upregulate Notch and Wnt pathway genes • The inhibition of Notch and Wnt restores OL differentiation in PRC2 null OL cells • The Notch inhibition blocks aberrant astrocytic gene induction in PRC2 null OL cells Wang et al. show that the polycomb repressive complex PRC2 is required for the differentiation of oligodendrocyte precursors to myelinating oligodendrocytes. They further show that PRC2 promotes oligodendrocyte differentiation and inhibits erroneous astrocytic fate by repressing the Notch pathway. [ABSTRACT FROM AUTHOR]
- Published
- 2020
- Full Text
- View/download PDF
31. DEAD-Box Helicase 18 Counteracts PRC2 to Safeguard Ribosomal DNA in Pluripotency Regulation.
- Author
-
Zhang, Hui, Wu, Zhongyang, Lu, J. Yuyang, Huang, Bo, Zhou, Hongwei, Xie, Wei, Wang, Jianlong, and Shen, Xiaohua
- Abstract
Embryonic stem cells (ESCs) exhibit high levels of ribosomal RNA (rRNA) transcription and ribosome biogenesis. Here, we reveal an unexpected role for an essential DEAD-box helicase, DDX18, in antagonizing the polycomb repressive complex 2 (PRC2) to prevent deposition of the repressive H3K27me3 mark onto rDNA in pluripotent cells. DDX18 binds and sequesters PRC2 in the outer layer of the nucleolus and counteracts PRC2 complex formation in vivo and in vitro. DDX18 knockdown leads to increased occupancy of PRC2 and H3K27me3 at rDNA loci, accompanied by drastically decreased rRNA transcription and reduced ribosomal protein expression and translation. Auxin-induced rapid degradation of DDX18 enhances PRC2 binding at rDNA. The inhibition of PRC2 partially rescues the effects of DDX18 depletion on rRNA transcription and ESC self-renewal. These results demonstrate a critical role for DDX18 in safeguarding the chromatin and transcriptional integrity of rDNA by counteracting the epigenetic silencing machinery to promote pluripotency. • DDX18 is essential for ESC self-renewal and embryonic development • DDX18 directly binds PRC2 and modulates PRC2 complex formation • DDX18 prevents PRC2 from accessing rDNA in the nucleolus • DDX18 promotes open chromatin and hyperactive transcription at rDNA Zhang et al. report an interplay between the DEAD-box helicase DDX18 and the polycomb repressive complex 2 (PRC2) in governing the hyperactive state of ribosomal DNA in pluripotency regulation. DDX18 counteracts PRC2 complex formation and prevents PRC2 binding and aberrant histone methylation at rDNA loci. [ABSTRACT FROM AUTHOR]
- Published
- 2020
- Full Text
- View/download PDF
32. A Functional Link between Nuclear RNA Decay and Transcriptional Control Mediated by the Polycomb Repressive Complex 2.
- Author
-
Garland, William, Comet, Itys, Wu, Mengjun, Radzisheuskaya, Aliaksandra, Rib, Leonor, Vitting-Seerup, Kristoffer, Lloret-Llinares, Marta, Sandelin, Albin, Helin, Kristian, and Jensen, Torben Heick
- Abstract
Pluripotent embryonic stem cells (ESCs) constitute an essential cellular niche sustained by epigenomic and transcriptional regulation. Any role of post-transcriptional processes remains less explored. Here, we identify a link between nuclear RNA levels, regulated by the poly(A) RNA exosome targeting (PAXT) connection, and transcriptional control by the polycomb repressive complex 2 (PRC2). Knockout of the PAXT component ZFC3H1 impairs mouse ESC differentiation. In addition to the upregulation of bona fide PAXT substrates, Zfc3h1
−/− cells abnormally express developmental genes usually repressed by PRC2. Such de-repression is paralleled by decreased PRC2 binding to chromatin and low PRC2-directed H3K27 methylation. PRC2 complex stability is compromised in Zfc3h1−/− cells with elevated levels of unspecific RNA bound to PRC2 components. We propose that excess RNA hampers PRC2 function through its sequestration from DNA. Our results highlight the importance of balancing nuclear RNA levels and demonstrate the capacity of bulk RNA to regulate chromatin-associated proteins. • Depletion of ZFC3H1 in mouse ESCs results in differentiation defects • PRC2 target genes are deregulated in Zfc3h1−/− cells • Chromatin binding of PRC2 and H3K27me3 is reduced in Zfc3h1−/− cells • Increased binding of RNA impairs PRC2 complex stability ZFC3H1 targets pA+ RNA for decay by the nuclear RNA exosome. Garland et al. report a disruptive relationship between excess RNA and PRC2 upon depletion of ZFC3H1 in mouse ESCs. In such conditions, RNA is bound by PRC2 components, which show reduced binding to chromatin and fellow PRC2 proteins. [ABSTRACT FROM AUTHOR]- Published
- 2019
- Full Text
- View/download PDF
33. Reduction of Global H3K27me3 Enhances HER2/ErbB2 Targeted Therapy.
- Author
-
Hirukawa, Alison, Singh, Salendra, Wang, Jarey, Rennhack, Jonathan P., Swiatnicki, Matthew, Sanguin-Gendreau, Virginie, Zuo, Dongmei, Daldoul, Kamilia, Lavoie, Cynthia, Park, Morag, Andrechek, Eran R., Westbrook, Thomas F., Harris, Lyndsay N., Varadan, Vinay, Smith, Harvey W., and Muller, William J.
- Abstract
Monoclonal antibodies (mAbs) targeting the oncogenic receptor tyrosine kinase ERBB2/HER2, such as Trastuzumab, are the standard of care therapy for breast cancers driven by ERBB2 overexpression and activation. However, a substantial proportion of patients exhibit de novo resistance. Here, by comparing matched Trastuzumab-naive and post-treatment patient samples from a neoadjuvant trial, we link resistance with elevation of H3K27me
3 , a repressive histone modification catalyzed by polycomb repressor complex 2 (PRC2). In ErbB2+ breast cancer models, PRC2 silences endogenous retroviruses (ERVs) to suppress anti-tumor type-I interferon (IFN) responses. In patients, elevated H3K27me3 in tumor cells following Trastuzumab treatment correlates with suppression of interferon-driven viral defense gene expression signatures and poor response. Using an immunocompetent model, we provide evidence that EZH2 inhibitors promote interferon-driven immune responses that enhance the efficacy of anti-ErbB2 mAbs, suggesting the potential clinical benefit of epigenomic reprogramming by H3K27me3 depletion in Trastuzumab-resistant disease. • EZH2 activity correlates with Trastuzumab resistance and antiviral gene silencing • EZH2 silences retrotransposons in ErbB2+ breast cancer • EZH2 inhibition induces retrotransposon expression and a type-I interferon response • Inhibiting EZH2 in vivo boosts immune infiltration and response to anti-ErbB2 mAbs Hirukawa et al. link Trastuzumab resistance in ErbB2+ breast cancers with activity of the methyltransferase EZH2, a key epigenetic regulator. By silencing retrotransposons, EZH2 suppresses type-I interferon signaling to limit immune surveillance. Retrotransposon de-repression following EZH2 inhibition triggers interferon responses and sensitizes immunocompetent in vivo models to ErbB2 antibody therapy. [ABSTRACT FROM AUTHOR]- Published
- 2019
- Full Text
- View/download PDF
34. The TGFB2-AS1 lncRNA Regulates TGF-β Signaling by Modulating Corepressor Activity.
- Author
-
Papoutsoglou, Panagiotis, Tsubakihara, Yutaro, Caja, Laia, Morén, Anita, Pallis, Paris, Ameur, Adam, Heldin, Carl-Henrik, and Moustakas, Aristidis
- Abstract
Molecular processes involving lncRNAs regulate cell function. By applying transcriptomics, we identify lncRNAs whose expression is regulated by transforming growth factor β (TGF-β). Upon silencing individual lncRNAs, we identify several that regulate TGF-β signaling. Among these lncRNAs, TGFB2-antisense RNA1 (TGFB2-AS1) is induced by TGF-β through Smad and protein kinase pathways and resides in the nucleus. Depleting TGFB2-AS1 enhances TGF-β/Smad-mediated transcription and expression of hallmark TGF-β-target genes. Increased dose of TGFB2-AS1 reduces expression of these genes, attenuates TGF-β-induced cell growth arrest, and alters BMP and Wnt pathway gene profiles. Mechanistically, TGFB2-AS1 , mainly via its 3′ terminal region, binds to the EED adaptor of the Polycomb repressor complex 2 (PRC2), promoting repressive histone H3K27me
3 modifications at TGF-β-target gene promoters. Silencing EED or inhibiting PRC2 methylation activity partially rescues TGFB2-AS1 -mediated gene repression. Thus, the TGF-β-induced TGFB2-AS1 lncRNA exerts inhibitory functions on TGF-β/BMP signaling output, supporting auto-regulatory negative feedback that balances TGF-β/BMP-mediated responses. • TGF-β signaling transcriptionally regulates lncRNAs that regulate TGF-β signaling • TGFB2-AS1 is induced by TGF-β to negatively regulate Smad transcriptional output • TGFB2-AS1 associates with EED, the Polycomb repressor complex 2 adaptor • TGFB2-AS1 promotes repressive histone modifications at TGF-β-target genes Papoutsoglou et al. show that TGFB2-antisense RNA1 (TGFB2-AS1) is induced by TGF-β, interacts with the EED adaptor of the Polycomb repressor complex 2, and limits the response of target genes to TGF-β signaling. [ABSTRACT FROM AUTHOR]- Published
- 2019
- Full Text
- View/download PDF
35. Reduction of Global H3K27me 3 Enhances HER2/ErbB2 Targeted Therapy.
- Author
-
Hirukawa A, Singh S, Wang J, Rennhack JP, Swiatnicki M, Sanguin-Gendreau V, Zuo D, Daldoul K, Lavoie C, Park M, Andrechek ER, Westbrook TF, Harris LN, Varadan V, Smith HW, and Muller WJ
- Subjects
- Adult, Animals, Breast Neoplasms drug therapy, Cell Line, Tumor, Drug Resistance, Neoplasm, Enhancer of Zeste Homolog 2 Protein metabolism, Female, Humans, Interferon Type I metabolism, Methylation, Mice, Models, Biological, Polycomb Repressive Complex 2 metabolism, Retroelements genetics, Trastuzumab therapeutic use, Up-Regulation, Histones metabolism, Lysine metabolism, Molecular Targeted Therapy, Receptor, ErbB-2 metabolism
- Abstract
Monoclonal antibodies (mAbs) targeting the oncogenic receptor tyrosine kinase ERBB2/HER2, such as Trastuzumab, are the standard of care therapy for breast cancers driven by ERBB2 overexpression and activation. However, a substantial proportion of patients exhibit de novo resistance. Here, by comparing matched Trastuzumab-naive and post-treatment patient samples from a neoadjuvant trial, we link resistance with elevation of H3K27me
3 , a repressive histone modification catalyzed by polycomb repressor complex 2 (PRC2). In ErbB2+ breast cancer models, PRC2 silences endogenous retroviruses (ERVs) to suppress anti-tumor type-I interferon (IFN) responses. In patients, elevated H3K27me3 in tumor cells following Trastuzumab treatment correlates with suppression of interferon-driven viral defense gene expression signatures and poor response. Using an immunocompetent model, we provide evidence that EZH2 inhibitors promote interferon-driven immune responses that enhance the efficacy of anti-ErbB2 mAbs, suggesting the potential clinical benefit of epigenomic reprogramming by H3K27me3 depletion in Trastuzumab-resistant disease., (Copyright © 2019 The Authors. Published by Elsevier Inc. All rights reserved.)- Published
- 2019
- Full Text
- View/download PDF
36. Meg3 Non-coding RNA Expression Controls Imprinting by Preventing Transcriptional Upregulation in cis.
- Author
-
Sanli I, Lalevée S, Cammisa M, Perrin A, Rage F, Llères D, Riccio A, Bertrand E, and Feil R
- Subjects
- Alleles, Animals, CRISPR-Cas Systems genetics, Calcium-Binding Proteins, Cell Differentiation, Cell Line, DNA Methylation, Embryonic Stem Cells, Enhancer of Zeste Homolog 2 Protein metabolism, Histones metabolism, Intercellular Signaling Peptides and Proteins genetics, Intercellular Signaling Peptides and Proteins metabolism, Mice, Mice, Inbred C57BL, Neurons cytology, Neurons metabolism, Polycomb-Group Proteins metabolism, Promoter Regions, Genetic, RNA, Long Noncoding genetics, Transcriptional Activation, Up-Regulation, Genomic Imprinting, RNA, Long Noncoding metabolism
- Abstract
Although many long non-coding RNAs (lncRNAs) are imprinted, their roles often remain unknown. The Dlk1-Dio3 domain expresses the lncRNA Meg3 and multiple microRNAs and small nucleolar RNAs (snoRNAs) on the maternal chromosome and constitutes an epigenetic model for development. The domain's Dlk1 (Delta-like-1) gene encodes a ligand that inhibits Notch1 signaling and regulates diverse developmental processes. Using a hybrid embryonic stem cell (ESC) system, we find that Dlk1 becomes imprinted during neural differentiation and that this involves transcriptional upregulation on the paternal chromosome. The maternal Dlk1 gene remains poised. Its protection against activation is controlled in cis by Meg3 expression and also requires the H3-Lys-27 methyltransferase Ezh2. Maternal Meg3 expression additionally protects against de novo DNA methylation at its promoter. We find that Meg3 lncRNA is partially retained in cis and overlaps the maternal Dlk1 in embryonic cells. Combined, our data evoke an imprinting model in which allelic lncRNA expression prevents gene activation in cis., (Copyright © 2018 The Author(s). Published by Elsevier Inc. All rights reserved.)
- Published
- 2018
- Full Text
- View/download PDF
37. Jarid2 Coordinates Nanog Expression and PCP/Wnt Signaling Required for Efficient ESC Differentiation and Early Embryo Development
- Author
-
David Landeira, Neil Brockdorff, Elodie Ndjetehe, Irene Cantone, Hakan Bagci, Jorge Soza-Ried, Amelie Feytout, Karen E. Brown, Andrzej R. Malinowski, Amanda G. Fisher, Thomas L. Carroll, Matthias Merkenschlager, Zoe Webster, Helena G Asenjo, Landeira, D., Bagci, H., Malinowski, A. R., Brown, K. E., Soza-Ried, J., Feytout, A., Webster, Z., Ndjetehe, E., Cantone, I., Asenjo, H. G., Brockdorff, N., Carroll, T., Merkenschlager, M., Fisher, A. G., and Commission of the European Communities
- Subjects
Frizzled ,Cell self-renewal ,Cellular differentiation ,MOUSE ,BETA-CATENIN ,Mice ,lcsh:QH301-705.5 ,Wnt Signaling Pathway ,Cells, Cultured ,beta Catenin ,reproductive and urinary physiology ,GENE-EXPRESSION ,Regulation of gene expression ,WNT/BETA-CATENIN ,Wnt signaling pathway ,Polycomb Repressive Complex 2 ,Gene Expression Regulation, Developmental ,Cell Differentiation ,Nanog Homeobox Protein ,LIM Domain Proteins ,PRC2 ,Cell biology ,embryonic structures ,biological phenomena, cell phenomena, and immunity ,Life Sciences & Biomedicine ,Homeobox protein NANOG ,Pluripotency ,Rex1 ,EPIBLAST STEM-CELLS ,Biology ,PLURIPOTENCY ,General Biochemistry, Genetics and Molecular Biology ,Article ,Animals ,JUMONJI ,Embryonic Stem Cells ,POLYCOMB REPRESSION ,Adaptor Proteins, Signal Transducing ,Homeodomain Proteins ,Science & Technology ,Polycom repression ,urogenital system ,Cell Biology ,Molecular biology ,Embryonic stem cell ,Frizzled Receptors ,Wnt Proteins ,Epiblast stem-cells ,Blastocyst ,lcsh:Biology (General) ,Beta-catenin ,Gene expression ,CELL SELF-RENEWAL - Abstract
Summary Jarid2 is part of the Polycomb Repressor complex 2 (PRC2) responsible for genome-wide H3K27me3 deposition. Unlike other PRC2-deficient embryonic stem cells (ESCs), however, Jarid2-deficient ESCs show a severe differentiation block, altered colony morphology, and distinctive patterns of deregulated gene expression. Here, we show that Jarid2−/− ESCs express constitutively high levels of Nanog but reduced PCP signaling components Wnt9a, Prickle1, and Fzd2 and lowered β-catenin activity. Depletion of Wnt9a/Prickle1/Fzd2 from wild-type ESCs or overexpression of Nanog largely phenocopies these cellular defects. Co-culture of Jarid2−/− with wild-type ESCs restores variable Nanog expression and β-catenin activity and can partially rescue the differentiation block of mutant cells. In addition, we show that ESCs lacking Jarid2 or Wnt9a/Prickle1/Fzd2 or overexpressing Nanog induce multiple ICM formation when injected into normal E3.5 blastocysts. These data describe a previously unrecognized role for Jarid2 in regulating a core pluripotency and Wnt/PCP signaling circuit that is important for ESC differentiation and for pre-implantation development., Graphical Abstract, Highlights • ESCs lacking Jarid2 show constitutive Nanog expression • ESCs lacking Jarid2 have reduced PCP/Wnt signaling • Co-culture of Jarid2-null and WT ESCs restores differentiation capability • Jarid2-null ESCs form more than one ICM upon injection to E3.5 mouse blastocysts, Landeira et al. show that Jarid2-null ESCs have reduced Wnt9a/Prickle1/Fzd2 and low β-catenin activity, resulting in altered adhesion, constitutive expression of Nanog, and failure to differentiate. Their experiments identify a non-canonical function for Jarid2 in regulating the balance between ESC self-renewal and differentiation.
- Full Text
- View/download PDF
38. Roles of H3K27me2 and H3K27me3 Examined during Fate Specification of Embryonic Stem Cells
- Author
-
Kyung Dae Ko, Jordan Krebs, Vittorio Sartorelli, Xuesong Feng, Roger A. Pedersen, Rafael Casellas, Hossein Zare, Adam L. Knight, Karinna O. Vivanco, Aster H. Juan, Pei Fang Tsai, Gustavo Gutierrez-Cruz, Anthony M. Ascoli, Benjamin A. Garcia, Simone Sidoli, Jizhong Zou, Stan Wang, Massachusetts Institute of Technology. Department of Biological Engineering, and Vivanco, Karinna O.
- Subjects
0301 basic medicine ,Transcription, Genetic ,Cellular differentiation ,Cell ,H3K27 methylation ,Embryoid body ,macromolecular substances ,Regulatory Sequences, Nucleic Acid ,Methylation ,General Biochemistry, Genetics and Molecular Biology ,Article ,Histones ,Mice ,03 medical and health sciences ,Histone H3 ,Transcription Activator-Like Effector Nucleases ,medicine ,Animals ,Cell Lineage ,Enhancer of Zeste Homolog 2 Protein ,Psychological repression ,lcsh:QH301-705.5 ,Embryoid Bodies ,Neurons ,Regulation of gene expression ,Genetics ,Genome ,biology ,Lysine ,EZH2 ,fungi ,Cell Differentiation ,Mouse Embryonic Stem Cells ,embryonic stem cells ,Embryonic stem cell ,Cell biology ,3. Good health ,medicine.anatomical_structure ,030104 developmental biology ,polycomb proteins ,Gene Expression Regulation ,lcsh:Biology (General) ,biology.protein ,RNA Editing ,PRC2 - Abstract
The polycomb repressive complex 2 (PRC2) methylates lysine 27 of histone H3 (H3K27) through its catalytic subunit Ezh2. PRC2-mediated di- and tri-methylation (H3K27me2/H3K27me3) have been interchangeably associated with gene repression. However, it remains unclear whether these two degrees of H3K27 methylation have different functions. In this study, we have generated isogenic mouse embryonic stem cells (ESCs) with a modified H3K27me2/H3K27me3 ratio. Our findings document dynamic developmental control in the genomic distribution of H3K27me2 and H3K27me3 at regulatory regions in ESCs. They also reveal that modifying the ratio of H3K27me2 and H3K27me3 is sufficient for the acquisition and repression of defined cell lineage transcriptional programs and phenotypes and influences induction of the ESC ground state., National Institute of Arthritis and Musculoskeletal and Skin Diseases (U.S.). Intramural Research Program
- Full Text
- View/download PDF
39. PARTICLE, a Triplex-Forming Long ncRNA, Regulates Locus-Specific Methylation in Response to Low-Dose Irradiation
- Author
-
Maximilian Niyazi, Matt Trau, Natasa Anastasov, Vanja Radulovic, Valerie B. O'Leary, Laura G. Carrascosa, Michael J. Atkinson, Saak V. Ovsepian, Simone Moertl, and Fabian A. Buske
- Subjects
radiotherapy [Carcinoma, Squamous Cell] ,Chromatin Immunoprecipitation ,biosynthesis [Methionine Adenosyltransferase] ,Protein subunit ,Immunoblotting ,genetics [RNA, Long Noncoding] ,Electrophoretic Mobility Shift Assay ,genetics [DNA Methylation] ,General Biochemistry, Genetics and Molecular Biology ,Cell Line ,03 medical and health sciences ,MAT2A protein, human ,0302 clinical medicine ,Radiation, Ionizing ,Humans ,Electrophoretic mobility shift assay ,ddc:610 ,lcsh:QH301-705.5 ,In Situ Hybridization ,030304 developmental biology ,Oligonucleotide Array Sequence Analysis ,0303 health sciences ,biology ,radiation effects [Gene Expression Regulation] ,Squamous Cell Carcinoma of Head and Neck ,Intercellular transport ,radiotherapy [Head and Neck Neoplasms] ,genetics [Methionine Adenosyltransferase] ,RNA ,Methylation ,Methionine Adenosyltransferase ,DNA Methylation ,Surface Plasmon Resonance ,Molecular biology ,Cell biology ,ddc ,radiation effects [DNA Methylation] ,CpG site ,lcsh:Biology (General) ,Gene Expression Regulation ,Head and Neck Neoplasms ,030220 oncology & carcinogenesis ,biosynthesis [RNA, Long Noncoding] ,biology.protein ,Carcinoma, Squamous Cell ,RNA, Long Noncoding ,PRC2 ,Chromatin immunoprecipitation - Abstract
SummaryExposure to low-dose irradiation causes transiently elevated expression of the long ncRNA PARTICLE (gene PARTICLE, promoter of MAT2A-antisense radiation-induced circulating lncRNA). PARTICLE affords both a cytosolic scaffold for the tumor suppressor methionine adenosyltransferase (MAT2A) and a nuclear genetic platform for transcriptional repression. In situ hybridization discloses that PARTICLE and MAT2A associate together following irradiation. Bromouridine tracing and presence in exosomes indicate intercellular transport, and this is supported by ex vivo data from radiotherapy-treated patients. Surface plasmon resonance indicates that PARTICLE forms a DNA-lncRNA triplex upstream of a MAT2A promoter CpG island. We show that PARTICLE represses MAT2A via methylation and demonstrate that the radiation-induced PARTICLE interacts with the transcription-repressive complex proteins G9a and SUZ12 (subunit of PRC2). The interplay of PARTICLE with MAT2A implicates this lncRNA in intercellular communication and as a recruitment platform for gene-silencing machineries through triplex formation in response to irradiation.
- Full Text
- View/download PDF
40. [Untitled]
- Subjects
0301 basic medicine ,Methylation ,Biology ,Phenotype ,General Biochemistry, Genetics and Molecular Biology ,3. Good health ,03 medical and health sciences ,chemistry.chemical_compound ,030104 developmental biology ,0302 clinical medicine ,chemistry ,DNA methylation ,Cancer research ,biology.protein ,Gene silencing ,Epithelial–mesenchymal transition ,PRC2 ,Psychological repression ,030217 neurology & neurosurgery ,DNA - Abstract
Summary Loss-of-function mutations in the SDHB subunit of succinate dehydrogenase predispose patients to aggressive tumors characterized by pseudohypoxic and hypermethylator phenotypes. The mechanisms leading to DNA hypermethylation and its contribution to SDH-deficient cancers remain undemonstrated. We examine the genome-wide distribution of 5-methylcytosine and 5-hydroxymethylcytosine and their correlation with RNA expression in SDHB-deficient tumors and murine Sdhb−/− cells. We report that DNA hypermethylation results from TET inhibition. Although it preferentially affects PRC2 targets and known developmental genes, PRC2 activity does not contribute to the DNA hypermethylator phenotype. We also prove, in vitro and in vivo, that TET silencing, although recapitulating the methylation profile of Sdhb−/− cells, is not sufficient to drive their EMT-like phenotype, which requires additional HIF2α activation. Altogether, our findings reveal synergistic roles of TET repression and pseudohypoxia in the acquisition of metastatic traits, providing a rationale for targeting HIF2α and DNA methylation in SDH-associated malignancies.
41. [Untitled]
- Subjects
Regulation of gene expression ,Genetics ,biology ,fungi ,macromolecular substances ,General Biochemistry, Genetics and Molecular Biology ,Histone H3 ,Histone ,Histone phosphorylation ,Histone methylation ,biology.protein ,Nucleosome ,Homeotic gene ,PRC2 - Abstract
Trimethylation at histone H3K27 is central to the polycomb repression system. Juxtaposed to H3K27 is a widely conserved phosphorylatable serine residue (H3S28) whose function is unclear. To assess the importance of H3S28, we generated a Drosophila H3 histone mutant with a serine-to-alanine mutation at position 28. H3S28A mutant cells lack H3S28ph on mitotic chromosomes but support normal mitosis. Strikingly, all methylation states of H3K27 drop in H3S28A cells, leading to Hox gene derepression and to homeotic transformations in adult tissues. These defects are not caused by active H3K27 demethylation nor by the loss of H3S28ph. Biochemical assays show that H3S28A nucleosomes are a suboptimal substrate for PRC2, suggesting that the unphosphorylated state of serine 28 is important for assisting in the function of polycomb complexes. Collectively, our data indicate that the conserved H3S28 residue in metazoans has a role in supporting PRC2 catalysis.
42. [Untitled]
- Subjects
0301 basic medicine ,biology ,Chemistry ,EZH2 ,SMAD ,Transforming growth factor beta ,General Biochemistry, Genetics and Molecular Biology ,Cell biology ,Transcriptome ,03 medical and health sciences ,030104 developmental biology ,0302 clinical medicine ,biology.protein ,SUZ12 ,Signal transduction ,PRC2 ,Corepressor ,030217 neurology & neurosurgery - Abstract
Molecular processes involving lncRNAs regulate cell function. By applying transcriptomics, we identify lncRNAs whose expression is regulated by transforming growth factor beta (TGF-beta). Upon sile ...
43. H3-K27M-mutant nucleosomes interact with MLL1 to shape the glioma epigenetic landscape
- Author
-
Efrat Shema, Bareket Dassa, Michelle Monje, Lawryn H. Kasper, Suzanne J. Baker, Natasha Morris, Danielle Algranati, Vadim Fedyuk, Noa Furth, Olga Beresh, and Daniel S. Jones
- Subjects
medicine.disease_cause ,General Biochemistry, Genetics and Molecular Biology ,Epigenesis, Genetic ,Histones ,Histone H3 ,medicine ,Nucleosome ,Humans ,Epigenetics ,Child ,Mutation ,biology ,Brain Neoplasms ,Polycomb Repressive Complex 2 ,Glioma ,Histone-Lysine N-Methyltransferase ,Chromatin ,Cell biology ,Nucleosomes ,Histone ,biology.protein ,H3K4me3 ,PRC2 ,Myeloid-Lymphoid Leukemia Protein - Abstract
Cancer-associated mutations in genes encoding histones dramatically reshape chromatin and support tumorigenesis. Lysine to methionine substitution of residue 27 on histone H3 (K27M) is a driver mutation in high-grade pediatric gliomas, known to abrogate Polycomb Repressive Complex 2 (PRC2) activity. We applied single-molecule systems to image individual nucleosomes and delineate the combinatorial epigenetic patterns associated with H3-K27M expression. We found that chromatin marks on H3-K27M-mutant nucleosomes are dictated both by their incorporation preferences and by intrinsic properties of the mutation. Mutant nucleosomes not only preferentially bind PRC2, but also directly interact with MLL1, thus leading to genome-wide redistribution of H3K4me3. H3-K27M-mediated deregulation of both repressive and active chromatin marks leads to unbalanced ‘bivalent’ chromatin, which may support a poorly differentiated cellular state. This study provides evidence for a direct effect of H3-K27M oncohistone on the MLL1-H3K4me3 pathway and highlights the capability of single-molecule tools to reveal mechanisms of chromatin deregulation in cancer.
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.