BMI1–UBR5 Axis Regulates Transcriptional Repression at Damaged Chromatin

Total Page:16

File Type:pdf, Size:1020Kb

BMI1–UBR5 Axis Regulates Transcriptional Repression at Damaged Chromatin BMI1–UBR5 axis regulates transcriptional repression at damaged chromatin Anthony Sancheza, Angelo De Vivoa, Nadima Upretyb, Jonghwan Kimb, Stanley M. Stevens Jr.a, and Younghoon Keea,1 aDepartment of Cell Biology, Microbiology, and Molecular Biology, College of Arts and Sciences, University of South Florida, Tampa, FL 33620; and bDepartment of Molecular Biosciences, University of Texas at Austin, Austin, TX 78712 Edited by Terry Magnuson, University of North Carolina at Chapel Hill, Chapel Hill, NC, and accepted by Editorial Board Member James E. Cleaver August 15, 2016 (received for review July 6, 2016) BMI1 is a component of the Polycomb Repressive Complex 1 (PRC1), at damaged chromatin (11). How BMI1 or BMI1-induced H2AK119- which plays a key role in maintaining epigenetic silencing during de- Ub modulates transcriptional output upon DNA damage remains velopment. BMI1 also participates in gene silencing during DNA dam- incompletely understood. age response, but the precise downstream function of BMI1 in gene Here we found that the chromatin localization of the HECT E3 silencing is unclear. Here we identified the UBR5 E3 ligase as a down- ubiquitin ligase UBR5 is largely dependent on the PRC1 com- stream factor of BMI1. We found that UBR5 forms damage-inducible ponents BMI1, RNF1, and RNF2. Similar to BMI1 and PRC1 nuclear foci in a manner dependent on the PRC1 components BMI1, components, UBR5-depleted cells fail to repress transcription at RNF1 (RING1a), and RNF2 (RING1b). Whereas transcription is re- damaged chromatin. We further show that BMI1- and UBR5- pressed at UV-induced lesions on chromatin, depletion of the PRC1 mediated transcription repression involves the FACT histone members or UBR5 alone derepressed transcription elongation at chaperon complex. Our findings altogether suggest that UBR5 is a these sites, suggesting that UBR5 functions in a linear pathway with downstream effector of the PRC1 components in transcription PRC1 in inducing gene silencing at lesions. Mass spectrometry (MS) silencing at damaged chromatin. analysis revealed that UBR5 associates with BMI1 as well as FACT components SPT16 and SSRP1. We found that UBR5 localizes to the Results UV-induced lesions along with SPT16. We show that UBR5 ubiquiti- UBR5 Chromatin Localization Is Dependent on BMI1, RNF1, and RNF2. nates SPT16, and depletion of UBR5 or BMI1 leads to an enlargement During the course of our studies, we found that endogenous UBR5 of SPT16 foci size at UV lesions, suggesting that UBR5 and BMI1 re- proteins form distinct foci in the nucleus, which can be enhanced press SPT16 enrichment at the damaged sites. Consistently, depletion upon various DNA damaging agent treatments (Fig. 1A). Treat- of the FACT components effectively reversed the transcriptional de- ment of two independent siRNAs against UBR5 effectively elimi- repression incurred in the UBR5 and BMI1 KO cells. Finally, UBR5 and nated the foci, indicating that the foci represent the bona fide BMI1 KO cells are hypersensitive to UV, which supports the notion UBR5 proteins (knockdown efficiency is shown in Fig. 1B). The that faulty RNA synthesis at damaged sites is harmful to the cell foci partially overlap with ubiquitin aggregate (FK2 antibody) and fitness. Altogether, these results suggest that BMI1 and UBR5 repress γH2AX, markers of DNA damage (Fig. 1C); however, a significant the polymerase II (Pol II)-mediated transcription at damaged sites, by portion of the foci do not overlap, suggesting that UBR5 in chro- negatively regulating the FACT-dependent Pol II elongation. matin may have both DNA damage-dependent and -independent functions. The Pearson’s overlap coefficient is quantified using Polycomb | BMI1 | UBR5 | FACT | transcription ImageJ software and plotted (Fig. 1D), where the 53BP1–γH2AX overlap is used as a positive control (1 indicates a complete positive erturbation of chromatin structures can cause inappropriate relationship), and the MED12–γH2AX overlap as a negative Pgene expression and loss of genome integrity. Polycomb pro- control (−1 indicates a complete negative relationship). Consistent teins are recognized in all metazoans for their conserved tran- with the immunofluorescence (IF) data, a biochemical fractionation scriptional repressive function. The canonical Polycomb Repressive Complex 1 (PRC1) contains BMI1, RNF1 (RING1a), RNF2 Significance (RING1b), core components (PC), Polyhomeotic (PH), and CBX proteins (1, 2). BMI1 serves as a key regulatory component of the The BMI1-containing Polycomb Repressive Complex is an impor- PRC1complex,whichisrequiredtomaintainthetranscriptionally tant gene silencer during development, stem cell maintenance, and repressed state of many genes throughout development via chro- cancer progression. BMI1 is also involved in DNA damage response, matin remodeling and histone modification (1). The only known but its downstream mechanism is unclear. Here we identified UBR5 enzymatic activity of the BMI1-containing PRC1 complex is to as a downstream factor whose chromatin recruitment is regulated monoubiquitinate histone H2A at Lys-119 (K119) residue, which is by BMI1. We showed that BMI1 and UBR5 repress the RNA poly- associated with transcriptional repression (1, 3, 4). However, the merase II (Pol II) elongation and nascent RNA synthesis at UV- E3 ubiquitin ligase activity of RNF2 or the H2AK119-Ub is dis- induced DNA lesions. UBR5 associates with and regulates the pensable for repression of canonical PRC1 target genes during abundance of the histone chaperon complex FACT at damaged mouse or Drosophila embryonic development, respectively (5, 6), chromatin and regulates elongation of Pol II at the lesions. Thus, suggesting that the PRC1 complex may also induce gene silencing our studies identify a pathway that regulates the damage-induced through other mechanisms (7). A series of studies has suggested transcriptional repression that is governed by BMI1. that multiple distinct forms of the PRC1 complex with varying Author contributions: Y.K. designed research; A.S., A.D.V., and N.U. performed research; J.K. components could exist, and each of these may have distinct modes and S.M.S. contributed new reagents/analytic tools; A.S., A.D.V., J.K., S.M.S., and Y.K. ana- of regulation and functions (reviewed in ref. 2). lyzed data; and Y.K. wrote the paper. In addition to its well-known role as an oncogene, recent evi- The authors declare no conflict of interest. dence suggests that BMI1 participates in the DNA damage re- This article is a PNAS Direct Submission. T.M. is a Guest Editor invited by the Editorial sponse and genome integrity maintenance. BMI1 is known to Board. localize to DNA double-strand break (DSB) sites and facilitates 1To whom correspondence should be addressed. Email: [email protected]. – CELL BIOLOGY DNA repair (8 10). Additionally, consistent with its role in gene This article contains supporting information online at www.pnas.org/lookup/suppl/doi:10. silencing, BMI1 represses local elongation of RNA polymerase II 1073/pnas.1610735113/-/DCSupplemental. www.pnas.org/cgi/doi/10.1073/pnas.1610735113 PNAS | October 4, 2016 | vol. 113 | no. 40 | 11243–11248 Downloaded by guest on September 27, 2021 Etoposide UBR5 in the UBR5-knockdown cells (siUBR5). Fig. 3A, Right show A 80 Bsi: c #1 #2 NT UV - siUBR5 60 measurements of relative fluorescent intensity (RFI) along the UV 40 UBR5 250 spots, which highlight that there is no repression of Pol II elon- 20 50 Tubulin gation at the γH2AX spots in the knockdown cells. The same % cells with >5 UBR5 foci 0 - 1 2 3 NT UV MMC phenotypes were observed in BMI1 and UBR5 KO HeLa cells siUBR5#1siUBR5#2 (Fig. 3B; shown as Pearson’s correlation). A chemical probe, C D Etoposide E UBR5 FK2 Merge DAPI NT UV 5-ethynyl uridine (5-EU), is frequently used for detecting global S P S P 1 nascent RNA synthesis status using the Click-IT labeling technol- UBR5 0.5 250 ogy; whereas the 5-EU stain is excluded from the γH2AX spots, 0 SPT16 the 5-EU stain becomes overlapped in both BMI1 and UBR5 γ 100 UBR5 H2AX Merge DAPI -0.5 C D X knockdown cells (Fig. 3 ), or the KO cells (Fig. 3 ). These results Pearson’s correlation Pearson’s A AX H2A Ab pair: H2AX H2 15 suggest that RNA synthesis at damaged sites has indeed occurred γ ANCD2 UBR5/FK2 1 2 3 4 when UBR5 or BMI1 is depleted. 5-EU was added to dividing cells 53BP1/γUBR5/ UBR5/FMED12/γH2 immediately following UV irradiation, to specifically monitor the Fig. 1. UBR5 chromatin recruitment is enhanced upon DNA damage. (A) UBR5 nascent RNA synthesis that occurred after the damaging event. forms distinct nuclear foci in HeLa (shown), U2OS, and HCT116 cells in SI Ap- Similar phenotypes were observed when the WT, UBR5 KO, and pendix,Fig.S2following UV (30 J/m2, 1-h recovery), etoposide treatment (2 μM, BMI1 KO cells were globally irradiated with UV; the γH2AX and 12 h), or mitomycin C (MMC) (2 μM, 12 h). n = 80. (B) Confirmation of UBR5 P-Ser2 overlap is shown in SI Appendix,Fig.S5,andtheγH2AX knockdown. (C) The foci partially colocalize with ubiquitin aggregates (FK2) and 5-EU overlap is shown in SI Appendix, Fig. S6. Knockdown of and γH2AX. (D)Pearson’s correlation for overlap from each Ab pair is measured RNF2 also led to a near-identical phenotype (Fig. 3E), suggesting = + (n 100). (E) UBR5 becomes enriched in P fraction after UV (S, cyto nucleo- that Pol II elongation at damaged sites is regulated by the PRC1 plasm; P, chromatin enriched). complex activity. Knockdown of RNF20 (which induces H2B ubiquitination) or RNF8 (which induces H1 and H2A ubiquitina- assay shows that a fraction of UBR5 is present in the chromatin- tion) (13) did not show noticeable effects (Fig. 3E). The Pol II enriched fraction (P), which is increased upon UV damage (Fig. 1E). To identify the upstream regulator(s) for the foci formation, we screened siRNAs targeting known regulators of chromatin me- 100 BMI1 siC A B siBMI1#1 tabolism and DNA damage response (Fig.
Recommended publications
  • The Cross-Talk Between Methylation and Phosphorylation in Lymphoid-Specific Helicase Drives Cancer Stem-Like Properties
    Signal Transduction and Targeted Therapy www.nature.com/sigtrans ARTICLE OPEN The cross-talk between methylation and phosphorylation in lymphoid-specific helicase drives cancer stem-like properties Na Liu1,2,3, Rui Yang1,2, Ying Shi1,2, Ling Chen1,2, Yating Liu1,2, Zuli Wang1,2, Shouping Liu1,2, Lianlian Ouyang4, Haiyan Wang1,2, Weiwei Lai1,2, Chao Mao1,2, Min Wang1,2, Yan Cheng5, Shuang Liu4, Xiang Wang6, Hu Zhou7, Ya Cao1,2, Desheng Xiao1 and Yongguang Tao1,2,6 Posttranslational modifications (PTMs) of proteins, including chromatin modifiers, play crucial roles in the dynamic alteration of various protein properties and functions including stem-cell properties. However, the roles of Lymphoid-specific helicase (LSH), a DNA methylation modifier, in modulating stem-like properties in cancer are still not clearly clarified. Therefore, exploring PTMs modulation of LSH activity will be of great significance to further understand the function and activity of LSH. Here, we demonstrate that LSH is capable to undergo PTMs, including methylation and phosphorylation. The arginine methyltransferase PRMT5 can methylate LSH at R309 residue, meanwhile, LSH could as well be phosphorylated by MAPK1 kinase at S503 residue. We further show that the accumulation of phosphorylation of LSH at S503 site exhibits downregulation of LSH methylation at R309 residue, which eventually promoting stem-like properties in lung cancer. Whereas, phosphorylation-deficient LSH S503A mutant promotes the accumulation of LSH methylation at R309 residue and attenuates stem-like properties, indicating the critical roles of LSH PTMs in modulating stem-like properties. Thus, our study highlights the importance of the crosstalk between LSH PTMs in determining its activity and function in lung cancer stem-cell maintenance.
    [Show full text]
  • A Family of Mammalian E3 Ubiquitin Ligases That Contain the UBR Box Motif and Recognize N-Degrons Takafumi Tasaki,1 Lubbertus C
    MOLECULAR AND CELLULAR BIOLOGY, Aug. 2005, p. 7120–7136 Vol. 25, No. 16 0270-7306/05/$08.00ϩ0 doi:10.1128/MCB.25.16.7120–7136.2005 Copyright © 2005, American Society for Microbiology. All Rights Reserved. A Family of Mammalian E3 Ubiquitin Ligases That Contain the UBR Box Motif and Recognize N-Degrons Takafumi Tasaki,1 Lubbertus C. F. Mulder,2 Akihiro Iwamatsu,3 Min Jae Lee,1 Ilia V. Davydov,4† Alexander Varshavsky,4 Mark Muesing,2 and Yong Tae Kwon1* Center for Pharmacogenetics and Department of Pharmaceutical Sciences, School of Pharmacy, University of Pittsburgh, Pittsburgh, Pennsylvania 152611; Aaron Diamond AIDS Research Center, The Rockefeller University, New York, New York 100162; Protein Research Network, Inc., Yokohama, Kanagawa 236-0004, Japan3; and Division of Biology, California Institute of Technology, Pasadena, California 911254 Received 15 March 2005/Returned for modification 27 April 2005/Accepted 13 May 2005 A subset of proteins targeted by the N-end rule pathway bear degradation signals called N-degrons, whose determinants include destabilizing N-terminal residues. Our previous work identified mouse UBR1 and UBR2 as E3 ubiquitin ligases that recognize N-degrons. Such E3s are called N-recognins. We report here that while double-mutant UBR1؊/؊ UBR2؊/؊ mice die as early embryos, the rescued UBR1؊/؊ UBR2؊/؊ fibroblasts still retain the N-end rule pathway, albeit of lower activity than that of wild-type fibroblasts. An affinity assay for proteins that bind to destabilizing N-terminal residues has identified, in addition to UBR1 and UBR2, a huge (570 kDa) mouse protein, termed UBR4, and also the 300-kDa UBR5, a previously characterized mammalian E3 known as EDD/hHYD.
    [Show full text]
  • A Computational Approach for Defining a Signature of Β-Cell Golgi Stress in Diabetes Mellitus
    Page 1 of 781 Diabetes A Computational Approach for Defining a Signature of β-Cell Golgi Stress in Diabetes Mellitus Robert N. Bone1,6,7, Olufunmilola Oyebamiji2, Sayali Talware2, Sharmila Selvaraj2, Preethi Krishnan3,6, Farooq Syed1,6,7, Huanmei Wu2, Carmella Evans-Molina 1,3,4,5,6,7,8* Departments of 1Pediatrics, 3Medicine, 4Anatomy, Cell Biology & Physiology, 5Biochemistry & Molecular Biology, the 6Center for Diabetes & Metabolic Diseases, and the 7Herman B. Wells Center for Pediatric Research, Indiana University School of Medicine, Indianapolis, IN 46202; 2Department of BioHealth Informatics, Indiana University-Purdue University Indianapolis, Indianapolis, IN, 46202; 8Roudebush VA Medical Center, Indianapolis, IN 46202. *Corresponding Author(s): Carmella Evans-Molina, MD, PhD ([email protected]) Indiana University School of Medicine, 635 Barnhill Drive, MS 2031A, Indianapolis, IN 46202, Telephone: (317) 274-4145, Fax (317) 274-4107 Running Title: Golgi Stress Response in Diabetes Word Count: 4358 Number of Figures: 6 Keywords: Golgi apparatus stress, Islets, β cell, Type 1 diabetes, Type 2 diabetes 1 Diabetes Publish Ahead of Print, published online August 20, 2020 Diabetes Page 2 of 781 ABSTRACT The Golgi apparatus (GA) is an important site of insulin processing and granule maturation, but whether GA organelle dysfunction and GA stress are present in the diabetic β-cell has not been tested. We utilized an informatics-based approach to develop a transcriptional signature of β-cell GA stress using existing RNA sequencing and microarray datasets generated using human islets from donors with diabetes and islets where type 1(T1D) and type 2 diabetes (T2D) had been modeled ex vivo. To narrow our results to GA-specific genes, we applied a filter set of 1,030 genes accepted as GA associated.
    [Show full text]
  • 4-6 Weeks Old Female C57BL/6 Mice Obtained from Jackson Labs Were Used for Cell Isolation
    Methods Mice: 4-6 weeks old female C57BL/6 mice obtained from Jackson labs were used for cell isolation. Female Foxp3-IRES-GFP reporter mice (1), backcrossed to B6/C57 background for 10 generations, were used for the isolation of naïve CD4 and naïve CD8 cells for the RNAseq experiments. The mice were housed in pathogen-free animal facility in the La Jolla Institute for Allergy and Immunology and were used according to protocols approved by the Institutional Animal Care and use Committee. Preparation of cells: Subsets of thymocytes were isolated by cell sorting as previously described (2), after cell surface staining using CD4 (GK1.5), CD8 (53-6.7), CD3ε (145- 2C11), CD24 (M1/69) (all from Biolegend). DP cells: CD4+CD8 int/hi; CD4 SP cells: CD4CD3 hi, CD24 int/lo; CD8 SP cells: CD8 int/hi CD4 CD3 hi, CD24 int/lo (Fig S2). Peripheral subsets were isolated after pooling spleen and lymph nodes. T cells were enriched by negative isolation using Dynabeads (Dynabeads untouched mouse T cells, 11413D, Invitrogen). After surface staining for CD4 (GK1.5), CD8 (53-6.7), CD62L (MEL-14), CD25 (PC61) and CD44 (IM7), naïve CD4+CD62L hiCD25-CD44lo and naïve CD8+CD62L hiCD25-CD44lo were obtained by sorting (BD FACS Aria). Additionally, for the RNAseq experiments, CD4 and CD8 naïve cells were isolated by sorting T cells from the Foxp3- IRES-GFP mice: CD4+CD62LhiCD25–CD44lo GFP(FOXP3)– and CD8+CD62LhiCD25– CD44lo GFP(FOXP3)– (antibodies were from Biolegend). In some cases, naïve CD4 cells were cultured in vitro under Th1 or Th2 polarizing conditions (3, 4).
    [Show full text]
  • Functional Diversification of the Nleg Effector Family in Enterohemorrhagic Escherichia Coli
    Functional diversification of the NleG effector family in enterohemorrhagic Escherichia coli Dylan Valleaua, Dustin J. Littleb, Dominika Borekc,d, Tatiana Skarinaa, Andrew T. Quailea, Rosa Di Leoa, Scott Houlistone,f, Alexander Lemake,f, Cheryl H. Arrowsmithe,f, Brian K. Coombesb, and Alexei Savchenkoa,g,1 aDepartment of Chemical Engineering and Applied Chemistry, University of Toronto, Toronto, ON M5S 3E5, Canada; bDepartment of Biochemistry and Biomedical Sciences, Michael G. DeGroote Institute for Infectious Disease Research, McMaster University, Hamilton, ON L8S 4K1, Canada; cDepartment of Biophysics, University of Texas Southwestern Medical Center, Dallas, TX 75390; dDepartment of Biochemistry, University of Texas Southwestern Medical Center, Dallas, TX 75390; ePrincess Margaret Cancer Centre, University of Toronto, Toronto, ON M5G 2M9, Canada; fDepartment of Medical Biophysics, University of Toronto, Toronto, ON M5G 1L7, Canada; and gDepartment of Microbiology, Immunology and Infectious Diseases, University of Calgary, Calgary, AB T2N 4N1, Canada Edited by Ralph R. Isberg, Howard Hughes Medical Institute and Tufts University School of Medicine, Boston, MA, and approved August 15, 2018 (receivedfor review November 6, 2017) The pathogenic strategy of Escherichia coli and many other gram- chains. Depending on the length and nature of the polyubiquitin negative pathogens relies on the translocation of a specific set of chain, it posttranslationally regulates the target protein’s locali- proteins, called effectors, into the eukaryotic host cell during in- zation, activation, or degradation. Ubiquitination is a multistep fection. These effectors act in concert to modulate host cell pro- process which begins with the ubiquitin-activating enzyme (E1) cesses in favor of the invading pathogen. Injected by the type III using ATP to “charge” ubiquitin, covalently binding the ubiquitin secretion system (T3SS), the effector arsenal of enterohemorrhagic C terminus by a thioester linkage.
    [Show full text]
  • Chronic Exposure of Humans to High Level Natural Background Radiation Leads to Robust Expression of Protective Stress Response Proteins S
    www.nature.com/scientificreports OPEN Chronic exposure of humans to high level natural background radiation leads to robust expression of protective stress response proteins S. Nishad1,2, Pankaj Kumar Chauhan3, R. Sowdhamini3 & Anu Ghosh1,2* Understanding exposures to low doses of ionizing radiation are relevant since most environmental, diagnostic radiology and occupational exposures lie in this region. However, the molecular mechanisms that drive cellular responses at these doses, and the subsequent health outcomes, remain unclear. A local monazite-rich high level natural radiation area (HLNRA) in the state of Kerala on the south-west coast of Indian subcontinent show radiation doses extending from ≤ 1 to ≥ 45 mGy/y and thus, serve as a model resource to understand low dose mechanisms directly on healthy humans. We performed quantitative discovery proteomics based on multiplexed isobaric tags (iTRAQ) coupled with LC–MS/MS on human peripheral blood mononuclear cells from HLNRA individuals. Several proteins involved in diverse biological processes such as DNA repair, RNA processing, chromatin modifcations and cytoskeletal organization showed distinct expression in HLNRA individuals, suggestive of both recovery and adaptation to low dose radiation. In protein–protein interaction (PPI) networks, YWHAZ (14-3-3ζ) emerged as the top-most hub protein that may direct phosphorylation driven pro- survival cellular processes against radiation stress. PPI networks also identifed an integral role for the cytoskeletal protein ACTB, signaling protein PRKACA; and the molecular chaperone HSPA8. The data will allow better integration of radiation biology and epidemiology for risk assessment [Data are available via ProteomeXchange with identifer PXD022380]. Te basic principles of low linear energy transfer (LET) ionizing radiation (IR) induced efects on mammalian systems have been broadly explored and there exists comprehensive knowledge on the health efects of high doses of IR delivered at high dose rates.
    [Show full text]
  • Visualization of Human Karyopherin Beta-1/Importin Beta-1 Interactions
    www.nature.com/scientificreports OPEN Visualization of human karyopherin beta-1/importin beta-1 interactions with protein partners in mitotic Received: 28 April 2017 Accepted: 29 December 2017 cells by co-immunoprecipitation Published: xx xx xxxx and proximity ligation assays Laura Di Francesco1,4, Annalisa Verrico2, Italia Anna Asteriti2, Paola Rovella2, Pietro Cirigliano3, Giulia Guarguaglini2, Maria Eugenia Schinin1 & Patrizia Lavia 2 Karyopherin beta-1/Importin beta-1 is a conserved nuclear transport receptor, acting in protein nuclear import in interphase and as a global regulator of mitosis. These pleiotropic functions refect its ability to interact with, and regulate, diferent pathways during the cell cycle, operating as a major efector of the GTPase RAN. Importin beta-1 is overexpressed in cancers characterized by high genetic instability, an observation that highlights the importance of identifying its partners in mitosis. Here we present the frst comprehensive profle of importin beta-1 interactors from human mitotic cells. By combining co-immunoprecipitation and proteome-wide mass spectrometry analysis of synchronized cell extracts, we identifed expected (e.g., RAN and SUMO pathway factors) and novel mitotic interactors of importin beta-1, many with RNA-binding ability, that had not been previously associated with importin beta-1. These data complement interactomic studies of interphase transport pathways. We further developed automated proximity ligation assay (PLA) protocols to validate selected interactors. We succeeded in obtaining spatial and temporal resolution of genuine importin beta-1 interactions, which were visualized and localized in situ in intact mitotic cells. Further developments of PLA protocols will be helpful to dissect importin beta-1-orchestrated pathways during mitosis.
    [Show full text]
  • Polycomb Group Protein Expression During Differentiation of Human
    Pethe et al. BMC Cell Biology 2014, 15:18 http://www.biomedcentral.com/1471-2121/15/18 RESEARCH ARTICLE Open Access Polycomb group protein expression during differentiation of human embryonic stem cells into pancreatic lineage in vitro Prasad Pethe, Punam Nagvenkar and Deepa Bhartiya* Abstract Background: Polycomb Group (PcG) proteins are chromatin modifiers involved in early embryonic development as well as in proliferation of adult stem cells and cancer cells. PcG proteins form large repressive complexes termed Polycomb Repressive Complexes (PRCs) of which PRC1 and PRC2 are well studied. Differentiation of human Embryonic Stem (hES) cells into insulin producing cells has been achieved to limited extent, but several aspects of differentiation remain unexplored. The PcG protein dynamics in human embryonic stem (hES) cells during differentiation into pancreatic lineage has not yet been reported. In the present study, the expression of RING1A, RING1B, BMI1, CBX2, SUZ12, EZH2, EED and JARID2 during differentiation of hES cells towards pancreatic lineage was examined. Results: In-house derived hES cell line KIND1 was used to study expression of PcG protein upon spontaneous and directed differentiation towards pancreatic lineage. qRT-PCR analysis showed expression of gene transcripts for various lineages in spontaneously differentiated KIND1 cells, but no differentiation into pancreatic lineage was observed. Directed differentiation induced KIND1 cells grown under feeder-free conditions to transition from definitive endoderm (Day 4), primitive gut tube stage (Day 8) and pancreatic progenitors (Day 12-Day 16) as evident from expression of SOX17, PDX1 and SOX9 by qRT-PCR and Western blotting. In spontaneously differentiating KIND1 cells, RING1A and SUZ12 were upregulated at day 15, while other PcG transcripts were downregulated.
    [Show full text]
  • Polycomb Repressor Complex 1 Promotes Gene Silencing Through H2AK119 Mono-Ubiquitination in Acinar-To-Ductal Metaplasia and Pancreatic Cancer Cells
    www.impactjournals.com/oncotarget/ Oncotarget, Vol. 7, No. 10 Polycomb repressor complex 1 promotes gene silencing through H2AK119 mono-ubiquitination in acinar-to-ductal metaplasia and pancreatic cancer cells Simone Benitz1,*, Ivonne Regel1,3,*, Tobias Reinhard1, Anna Popp1, Isabell Schäffer1, Susanne Raulefs1, Bo Kong1, Irene Esposito3, Christoph W. Michalski2,*, Jörg Kleeff1,4,5,* 1Department of Surgery, Technische Universität München, Munich, Germany 2Department of Surgery, University of Heidelberg, Heidelberg, Germany 3Institute of Pathology, Heinrich-Heine University, Duesseldorf, Germany 4The Royal Liverpool and Broadgreen University Hospitals, Liverpool, United Kingdom 5Department of Surgery, Heinrich-Heine University, Duesseldorf, Germany *These authors contribute equally to the manuscript. Correspondence to: Ivonne Regel, e-mail: [email protected] Keywords: polycomb repressor complex, histone mono-ubiquitination, pancreatic cancer, differentiation gene silencing Abbreviations: ADM (acinar-to-ductal metaplasia), PDAC (pancreatic ductal adenocarcinoma), PRC (polycomb repressor complex), PTF (pancreas specific transcription factor) Received: July 29, 2015 Accepted: November 16, 2015 Published: December 22, 2015 ABSTRACT Acinar-to-ductal metaplasia (ADM) occurring in cerulein-mediated pancreatitis or in oncogenic Kras-driven pancreatic cancer development is accompanied by extensive changes in the transcriptional program. In this process, acinar cells shut down the expression of acinar specific differentiation genes
    [Show full text]
  • Ezh2, the Histone Methyltransferase of PRC2, Regulates the Balance Between Self-Renewal and Differentiation in the Cerebral Cortex
    Ezh2, the histone methyltransferase of PRC2, regulates the balance between self-renewal and differentiation in the cerebral cortex João D. Pereiraa, Stephen N. Sansoma, James Smitha, Marc-Werner Dobeneckerb, Alexander Tarakhovskyb, and Frederick J. Liveseya,1 aGurdon Institute and Department of Biochemistry, University of Cambridge, Cambridge CB2 1QN, United Kingdom; and bLaboratory of Lymphocyte Signaling, The Rockefeller University, New York, NY 10021 Edited by Gerald R. Crabtree, Howard Hughes Medical Institute, Stanford, CA, and approved August 4, 2010 (received for review February 26, 2010) Multipotent progenitor cells of the cerebral cortex balance self- Knockout of the Bmi1 subunit of PRC1 has little effect on renewal and differentiation to produce complex neural lineages in progenitor cell self-renewal during development but is essential for a fixed temporal order in a cell-autonomous manner. We studied neural stem cell maintenance in the adult CNS (19). However, the role of the polycomb epigenetic system, a chromatin-based acute deletion of Bmi1 by RNAi does compromise cortical pro- repressive mechanism, in controlling cortical progenitor cell self- genitor cell self-renewal (20). Furthermore, removal of Ring1B, an renewal and differentiation. We found that the histone methyl- ubiquitin ligase component of PRC1, from the developing cortex transferase of polycomb repressive complex 2 (PCR2), enhancer of during neurogenesis lengthens the period of neurogenesis and Zeste homolog 2 (Ezh2), is essential for controlling the rate at
    [Show full text]
  • SOX9 Promotes Tumor Progression Through the Axis BMI1-P21cip
    www.nature.com/scientificreports OPEN SOX9 promotes tumor progression through the axis BMI1-p21CIP Paula Aldaz1,7, Maddalen Otaegi-Ugartemendia1,7, Ander Saenz-Antoñanzas1, Mikel Garcia- Puga1, Manuel Moreno-Valladares1,2, Juana M. Flores3, Daniela Gerovska4, Marcos J. Arauzo- Bravo 4,5,6, Nicolas Samprón1,2,5, Ander Matheu1,5,6* & Estefania Carrasco-Garcia1* The developmental regulator SOX9 is linked to cancer progression mainly as a result of its role in the regulation of cancer stem cells (CSCs). However, its activity in the diferentiated cells that constitute the heterogeneous tumor bulk has not been extensively studied. In this work, we addressed this aspect in gastric cancer, glioblastoma and pancreatic adenocarcinoma. SOX9 silencing studies revealed that SOX9 is required for cancer cell survival, proliferation and evasion of senescence in vitro and tumor growth in vivo. Gain of-SOX9 function showed that high levels of SOX9 promote tumor cell proliferation in vitro and in vivo. Mechanistically, the modulation of SOX9 changed the expression of the transcriptional repressor BMI1 in the same direction in the three types of cancer, and the expression of the tumor suppressor p21CIP in the opposite direction. In agreement with this, SOX9 expression positively correlated with BMI1 levels and inversely with p21CIP in clinical samples of the diferent cancers. Moreover, BMI1 re-establishment in SOX9-silenced tumor cells restored cell viability and proliferation as well as decreased p21CIP in vitro and tumor growth in vivo. These results indicate that BMI1 is a critical efector of the pro-tumoral activity of SOX9 in tumor bulk cells through the repression of p21CIP.
    [Show full text]
  • UBR-Box Containing Protein, UBR5, Is Over-Expressed in Human Lung 2 Adenocarcinoma and Is a Potential Therapeutic Target 3 Authors: Kumar Saurabh1, Parag P
    bioRxiv preprint doi: https://doi.org/10.1101/2020.06.03.131912; this version posted June 4, 2020. The copyright holder for this preprint (which was not certified by peer review) is the author/funder, who has granted bioRxiv a license to display the preprint in perpetuity. It is made available under aCC-BY-NC-ND 4.0 International license. 1 UBR-Box containing protein, UBR5, is over-expressed in human lung 2 adenocarcinoma and is a potential therapeutic target 3 Authors: Kumar Saurabh1, Parag P. Shah1, Mark A. Doll1,2, Leah J. Siskind1,2 and Levi J. 4 Beverly1,2,3,# 5 Affiliations: 6 1James Graham Brown Cancer Center, School of Medicine, University of Louisville, Louisville, 7 KY USA. 8 2Department of Pharmacology and Toxicology, University of Louisville, Louisville, KY, USA. 9 3Division of Hematology and Oncology, School of Medicine, University of Louisville, Louisville, 10 KY, USA. 11 #Corresponding author (Email: [email protected]) 12 13 Abstract 14 Background: N-end rule ubiquitination pathway is known to be disrupted in many diseases, 15 including cancer. UBR5, an E3 ubiquitin ligase, is mutated and/or overexpressed in human lung 16 cancer cells suggesting its pathological role in cancer. 17 Methods: We determined expression of UBR5 protein in multiple lung cancer cell lines and 18 human patient samples. Using immunoprecipitation followed by mass spectrometry we 19 determined the UBR5 interacting proteins. The impact of loss of UBR5 for lung adenocarcinoma 20 cell lines was analyzed using cell viability, clonogenic assays and in vivo xenograft models in 21 nude mice. Additional Western blot analysis was performed to assess the loss of UBR5 on 22 downstream signaling.
    [Show full text]