The E3 Ligase RING1 Targets P53 for Degradation and Promotes Cancer

Total Page:16

File Type:pdf, Size:1020Kb

The E3 Ligase RING1 Targets P53 for Degradation and Promotes Cancer Published OnlineFirst November 29, 2017; DOI: 10.1158/0008-5472.CAN-17-1805 Cancer Molecular Cell Biology Research The E3 Ligase RING1 Targets p53 for Degradation and Promotes Cancer Cell Proliferation and Survival Jiajia Shen1, Pengyu Li2, Xuejing Shao3, Yang Yang4, Xiujun Liu1, Min Feng5, Qiang Yu5, Ronggui Hu4, and Zhen Wang1 Abstract As a component of the transcriptional repression complex 1 p53-deficient cells. Its growth inhibitory effect was partially res- (PRC1), the ring finger protein RING1 participates in the epige- cued by p53 silencing, suggesting an important role for the netic regulation in cancer. However, the contributions of RING1 RING1–p53 complex in human cancer. In clinical specimens of to cancer etiology or development are unknown. In this study, we hepatocellular carcinoma, RING1 upregulation was evident in report that RING1 is a critical negative regulator of p53 homeo- association with poor clinical outcomes. Collectively, our results stasis in human hepatocellular and colorectal carcinomas. RING1 elucidate a novel PRC1-independent function of RING1 and acts as an E3 ubiquitin (Ub) ligase to directly interact with and provide a mechanistic rationale for its candidacy as a new prog- ubiquitinate p53, resulting in its proteasome-dependent degra- nostic marker and/or therapeutic target in human cancer. dation. The RING domain of RING1 was required for its E3 Ub Significance: These results elucidate a novel PRC1-independ- ligase activity. RING1 depletion inhibited the proliferation and ent function of RING1 and provide a mechanistic rationale for its survival of the p53 wild-type cancer cells by inducing cell-cycle candidacy as a new prognostic marker and/or therapeutic target in arrest, apoptosis, and senescence, with only modest effects on human cancer. Cancer Res; 78(2); 1–13. Ó2017 AACR. Introduction quitinate histone H2A at lysine 119 (H2AK119Ub1; refs. 8, 9). Different from the well-studied roles of RNF2 and BMI1 in In mammals, polycomb group (PcG) proteins consist of two carcinogenesis (10–12), limited evidence has been provided major polycomb repressive complexes (PRC): PRC1 and PRC2 with regard to the role of RING1 in cancer. An earlier report (1, 2). The PRC1 core complex includes RING1/RING1A, suggested overexpression of RING1 contributed to cellular RING1B/RNF2, and BMI1, whereas the PRC2 core complex transformation by upregulating the expression of proto- includes EED, EZH2, and SUZ12. PRC1 and PRC2 are known to oncogenes such as c-jun and c-fos (13). However, homeostatic play an important role as transcriptional repressors in embryonic levels of RING1 were found to vary significantly among development, stem cell self-renewal, and cell proliferation different cancer types, including lung cancer, prostate through epigenetic modifications of target genes (3, 4). Deregu- cancer, lymphoma, and kidney cancer (14–20), suggesting a lation of PcG genes is frequently found to be associated with yet unclear role of RING1 in cancer. developmental defects and cancer (3, 5–7). In this study, we have uncovered tumor suppressor p53 protein RING1 is a crucial component of the PRC1 and, along with as a novel bona fide substrate of RING1. RING1 directly interacts RNF2 and BMI1, acts as an E3 ubiquitin ligase to monoubi- with, and ubiquitinates p53, which finally leads to its proteasome- dependent degradation. As a result, RING1 depletion results in p53 stabilization, leading to cell-cycle arrest, apoptosis, senes- 1 Department of Biochemistry, Institute of Medicinal Biotechnology, Chinese cence, and migration inhibition in p53-proficient cells. Moreover, Academy of Medical Sciences and Peking Union Medical College, Beijing, China. we found that RING1 is highly expressed in hepatocellular car- 2Qilu Hospital of Shandong University, Jinan, China. 3Zhejiang Province Key Laboratory of Anti-Cancer Drug Research, Institute of Pharmacology and cinoma (HCC) tissues and its expression is associated with poor Toxicology, School of Pharmaceutical Sciences, Zhejiang University, Hangzhou, outcomes. China. 4State Key Laboratory of Molecular Biology, Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences, Shanghai, China. 5Cancer Thera- peutics and Stratified Oncology, Genome Institute of Singapore, Agency for Materials and Methods à Science, Technology, and Research (A STAR), Biopolis, Singapore. Plasmids Note: Supplementary data for this article are available at Cancer Research The pCMV-HA-p53, pcDNA3.0-RING1-Flag and deletion or Online (http://cancerres.aacrjournals.org/). point mutants, pGEX-4T-1-GST-p53, pSJ8-MBP-RING1-6His, Corresponding Authors: Zhen Wang, Institute of Medicinal Biotechnology and pGL4.17-p21-luc plasmids were constructed in our lab. Chinese Academy of Medical Sciences, 1# Tian Tan Xi Li, Beijing 100050, The siRNAs duplexes were synthesized by RiboBio Co., Ltd. China. Phone/Fax: 8610-6316-5289; E-mail: [email protected]; and si-RING1-1: 50-GTGGGAACTGAGTCTGTATGA-30,si-RING1-2: Ronggui Hu, E-mail: [email protected] 50-CAGATCAGACCACAACGAT-30. sh-RING1 and sh-p53 (with doi: 10.1158/0008-5472.CAN-17-1805 asequenceof50-GACTCCAGTGGTAATCTAC-30)werecloned Ó2017 American Association for Cancer Research. into pLKO.1 vector. www.aacrjournals.org OF1 Downloaded from cancerres.aacrjournals.org on September 28, 2021. © 2017 American Association for Cancer Research. Published OnlineFirst November 29, 2017; DOI: 10.1158/0008-5472.CAN-17-1805 Shen et al. Antibodies, reagents, and cell lines Etoposide treatment The anti-RING1 (D2P4D) (#13069), anti-p21 (12D1; #2947), HepG2 cells were seeded (5  105) into 6-well plates. Seventy- anti-p53 (1C12; #2524), anti-ubiquitin (P4D1; #3936) antibo- two hours after transfection with pcDNA3.0-RING1-Flag or dies were purchased from Cell Signaling Technology. The anti- si-RING1-1, cells were treated with 50 mmol/L etoposide for p53 (DO-1) (#SAB5100001), anti-Flag (#F1804), anti-HA indicated time before harvest and subsequent immunoblotting (#H6908), anti-GAPDH (#G8795), anti-b-actin (#A1978) anti- analyses, using described antibodies. bodies, anti-Flag M2 Affinity Gel (#A2220), and propidium iodide (PI, #P4170) were purchased from Sigma-Aldrich. Annexin Cell growth and colony formation assay V-PI Staining Kit was from Beijing 4A Biotech Co, Ltd. The anti-HA Following plasmid transfection or lentivirus infections, cell Affinity Gel (#B23302) was purchased from Biotool. The Gluta- growth was assessed as described previously (21). For colony thione Sepharose 4B (#17075601) was purchased from GE formation assay, the cells stably expressing sh-RING1 or sh-p53 Healthcare. Etoposide (ETO, #S1225) was purchased from Sell- were seeded (5  103/well) onto 6-well plates. After 3 weeks, the eckchem. HepG2 and HCT116 cell lines were obtained from colonies were stained with hematoxylin and counted. À À China Infrastructure of Cell Line Resources. HCT116 p53 / cells were gifted from Prof. Bert Volgestein at Johns Hopkins University Cell cycle and apoptosis analysis School of Medicine, Baltimore, MD. HEK293T cell lines were The cells were infected with sh-RING1 for 72 hours, and cell- from ATCC in the United States. All of the cell lines were authen- cycle distribution was performed as described previously (21). ticated by STR profiling at December 2016. Apoptosis was determined by Annexin V/PI Apoptosis Detection Kit. In drug treatment groups, the cells were infected with Cell transfection and infection sh-RING1 for 24 hours, followed by etoposide (50 mmol/L) Cell transfection was carried out using Lipofectamine 2000 treatment for another 48 hours. following the manufacturer's instruction. For lentiviral packaging, psPAX2, pMD2.0G, and pLKO.1-sh-p53, pLKO.1-sh-RING1, or the Senescence-associated b-galactosidase staining pLKO.1 scramble vectors were cotransfected into HEK293T cells, Senescence-associated b-galactosidase (SA-b-gal) staining was and the supernatants harvested at 48 or 96 hours after transfection. performed and quantified as described before (21). Lentivirus infection was transient or followed by screening with appropriate antibiotics to establish cell lines stably expressing the Migration assay indicated shRNAs. Specifically, for stable knockdown of RING1 or Migration capacity was measured by transwell and wound fi p53, the cells were infected with lentivirus expressing RING1 or p53 healing assay. The transwell lter (8-mm pores; Millicell Cell shRNA (sh-RING1 or sh-p53) in the presence of 8 mg/mL polybrene Culture Inserts) was used according to the instructions. Hema- (#107689; Sigma). Stable cell lines were selected and maintained at toxylin was used to stain the cells that migrated to the lower side of 5 mg/mL puromycin (#P8833; Sigma). the top chamber. For wound-healing assay, the cells infected with sh-RING1 were seeded into 6-well plates, and confluent cell Protein array analyses monolayers were wounded by manually scraping the cells, PathScan Cancer Phenotype Antibody Array Kit (#14821, Cell washed with PBS, and further cultured in medium without FBS Signaling Technology) was used to examine the level of cancer for the indicated time. Migration ability was represented by the cell-associated proteins in RING1 knockdown cells. The cells were percentage of the wound-healing area after normalization to infected with shRNA for 72 hours. Cell extracts were prepared and control using ImageJ. analyzed following the manufacturer's instruction. Generation of RNF2 KO cells using CRISPR/Cas9 system RNF2 fi Dual-luciferase assay A CRISPR/Cas9 system was used to establish -de cient The cells infected with sh-RING1 for 24 hours were seeded in HEK293T cells. Single guide RNA (sgRNA) was generated by 96-well plates at 1  104 cells per well for 24 hours before online CRISPR Design tool (http://crispr.mit.edu/). The sgRNA transfection with the pGL4.17-p21-luc plasmid (100 ng per well) sequences were cloned into pX260-U6-Chimeric-BB-CBh- and Renilla (5 ng per well) in triplicate. After 48 hours, the hSpCas9 plasmid and transfected into HEK293T cells. After being luciferase activity was measured as described (21). selected with 5 mg/mL puromycin, the cells were seeded into 96-well plates for monoclonalization.
Recommended publications
  • RNF2/Ring1b Negatively Regulates P53 Expression in Selective Cancer Cell Types to Promote Tumor Development
    RNF2/Ring1b negatively regulates p53 expression in selective cancer cell types to promote tumor development Wen-jing Sua,b, Jun-shun Fanga, Feng Chenga,b, Chao Liua, Fang Zhoua, and Jian Zhanga,1 aState Key Laboratory of Molecular Developmental Biology, Institute of Genetics and Developmental Biology and bGraduate School, Chinese Academy of Sciences, Beijing 100101, China Edited by Carol Prives, Columbia University, New York, NY, and approved December 19, 2012 (received for review July 15, 2012) Large numbers of studies have focused on the posttranslational evolution in some invertebrates, such as Caenorhabditis elegans regulation of p53 activity. One of the best-known negative regu- and Drosophila melanogaster, suggesting that other factors (in- lators for p53 is MDM2, an E3 ubiquitin ligase that promotes p53 cluding other E3 ligases) might be involved in controlling p53 degradation through proteasome degradation pathways. Additional activities in these animals. For example, Bonus is a homolog of E3 ligases have also been reported to negatively regulate p53. How- TRIM 24, a p53 ligase in vertebrates. The results from loss-of- ever, whether these E3 ligases have distinct/overlapping roles in function studies indicated that Bonus is critical for maintaining the regulation of p53 is largely unknown. In this study, we identify p53 activity in Drosophila, suggesting that Bonus might be an RNF2 (ring finger protein 2) as an E3 ligase that targets p53 for evolutionarily conserved E3 ligase for p53 (14). The existence of multiple E3 ligases for p53 strongly suggests that specialization degradation. The E3 ligase activity of RNF2 requires Bmi1 protein, among them must be required for controlling p53 at multiple a component of the polycomb group (PcG) complex.
    [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]
  • RNF2 Antibody Order 021-34695924 [email protected] Support 400-6123-828 50Ul [email protected] 100 Ul √ √ Web
    TD7403 RNF2 Antibody Order 021-34695924 [email protected] Support 400-6123-828 50ul [email protected] 100 uL √ √ Web www.ab-mart.com.cn Description: E3 ubiquitin-protein ligase that mediates monoubiquitination of 'Lys-119' of histone H2A (H2AK119Ub), thereby playing a central role in histone code and gene regulation. H2AK119Ub gives a specific tag for epigenetic transcriptional repression and participates in X chromosome inactivation of female mammals. May be involved in the initiation of both imprinted and random X inactivation (By similarity). Essential component of a Polycomb group (PcG) multiprotein PRC1-like complex, a complex class required to maintain the transcriptionally repressive state of many genes, including Hox genes, throughout development. PcG PRC1 complex acts via chromatin remodeling and modification of histones, rendering chromatin heritably changed in its expressibility. E3 ubiquitin-protein ligase activity is enhanced by BMI1/PCGF4. Acts as the main E3 ubiquitin ligase on histone H2A of the PRC1 complex, while RING1 may rather act as a modulator of RNF2/RING2 activity (Probable). Association with the chromosomal DNA is cell-cycle dependent. In resting B- and T-lymphocytes, interaction with AURKB leads to block its activity, thereby maintaining transcription in resting lymphocytes (By similarity). Uniprot:Q99496 Alternative Names: BAP 1; BAP1; DING; DinG protein; E3 ubiquitin protein ligase RING 2; E3 ubiquitin protein ligase RING2; E3 ubiquitin-protein ligase RING2; HIP2 interacting protein 3; HIP2- interacting protein 3; HIPI 3; HIPI3; Huntingtin interacting protein 2 interacting protein 3; Huntingtin-interacting protein 2-interacting protein 3; OTTHUMP00000060668; Polycomb M33 interacting protein Ring 1B; Polycomb M33 interacting protein Ring1B; Protein DinG; RING 1B; RING 2; RING finger protein 1B; RING finger protein 2; RING finger protein BAP 1; RING finger protein BAP-1; RING finger protein BAP1; RING1b; RING2_HUMAN; RNF 2; Rnf2; Specificity: RNF2 Antibody detects endogenous levels of total RNF2.
    [Show full text]
  • RING1B/RNF2 Rabbit Pab
    Leader in Biomolecular Solutions for Life Science [KO Validated] RING1B/RNF2 Rabbit pAb Catalog No.: A18076 KO Validated Basic Information Background Catalog No. Polycomb group (PcG) of proteins form the multiprotein complexes that are important A18076 for the transcription repression of various genes involved in development and cell proliferation. The protein encoded by this gene is one of the PcG proteins. It has been Observed MW shown to interact with, and suppress the activity of, transcription factor CP2 38kDa (TFCP2/CP2). Studies of the mouse counterpart suggested the involvement of this gene in the specification of anterior-posterior axis, as well as in cell proliferation in early Calculated MW development. This protein was also found to interact with huntingtin interacting protein 29kDa/37kDa 2 (HIP2), an ubiquitin-conjugating enzyme, and possess ubiquitin ligase activity. Category Primary antibody Applications WB, IHC, IF, IP, ChIP Cross-Reactivity Human, Mouse, Rat Recommended Dilutions Immunogen Information WB 1:500 - 1:2000 Gene ID Swiss Prot 6045 Q99496 IHC 1:50 - 1:200 Immunogen 1:50 - 1:200 IF Recombinant fusion protein containing a sequence corresponding to amino acids 1-336 of human RING1B/RING1B/RNF2 (NP_009143.1). IP 1:50 - 1:200 Synonyms ChIP 1:50 - 1:200 RNF2;BAP-1;BAP1;DING;HIPI3;RING1B;RING2 Contact Product Information www.abclonal.com Source Isotype Purification Rabbit IgG Affinity purification Storage Store at -20℃. Avoid freeze / thaw cycles. Buffer: PBS with 0.02% sodium azide,50% glycerol,pH7.3. Validation Data Western blot analysis of extracts from normal (control) and RING1B/RNF2 knockout (KO) HeLa cells, using RING1B/RNF2 antibody (A18076) at 1:1000 dilution.
    [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]
  • Chromatin Condensation of Xist Genomic Loci During Oogenesis In
    © 2015. Published by The Company of Biologists Ltd | Development (2015) 142, 4049-4055 doi:10.1242/dev.127308 RESEARCH REPORT Chromatin condensation of Xist genomic loci during oogenesis in mice Atsushi Fukuda1, Atsushi Mitani1,2, Toshiyuki Miyashita2, Akihiro Umezawa1 and Hidenori Akutsu1,3,* ABSTRACT (Augui et al., 2011; Sado and Sakaguchi, 2013; Nesterova et al., Repression of maternal Xist (Xm-Xist) during preimplantation in 2001). This expression pattern leads to the establishment of mouse embryos is essential for establishing imprinted X chromosome imprinted XCI in extra-embryonic tissues (Takagi and Sasaki, Xist Xist inactivation. Nuclear transplantation (NT) studies using nuclei derived 1975). Paternal (Xp- ) expression is driven by the deposition – from non-growing (ng) and full-grown (fg) oocytes have indicated that of maternal Rnf12 (also known as Rlim Mouse Genome maternal-specific repressive modifications are imposed on Xm-Xist Informatics) (Shin et al., 2010; Jonkers et al., 2009). However, Xist during oogenesis, as well as on autosomal imprinted genes. Recent the locus on the maternal X chromosome (Xm) is tightly studies have revealed that histone H3 lysine 9 trimethylation protected by epigenetic factors. Using parthenogenetic embryos, (H3K9me3) enrichments on Xm-Xist promoter regions are involved which are composed of two maternal genomes, we previously in silencing at the preimplantation stages. However, whether demonstrated that histone 3 lysine 9 trimethylation (H3K9me3) is Xist H3K9me3 is imposed on Xm-Xist during oogenesis is not known. essential for Xm- repression during early preimplantation Here, we dissected the chromatin states in ng and fg oocytes and phases (Fukuda et al., 2014). early preimplantation stage embryos.
    [Show full text]
  • Oncogenic Potential of BMI1: Race-Based Evidence in Prostate Cancer
    Editorial Page 1 of 5 Oncogenic potential of BMI1: race-based evidence in prostate cancer Eswar Shankar1,2, Shiv Verma1, Sanjay Gupta1,2,3,4,5 1Department of Urology, School of Medicine, Case Western Reserve University, Cleveland, OH 44106, USA; 2The Urology Institute, University Hospitals Cleveland Medical Center, Cleveland, OH 44106, USA; 3Department of Urology, Louis Stokes Cleveland Veterans Affairs Medical Center, Cleveland, OH 44106, USA; 4Department of Nutrition, Case Western Reserve University, Cleveland, OH 44106, USA; 5Division of General Medical Sciences, Case Comprehensive Cancer Center, Cleveland, OH 44106, USA Correspondence to: Sanjay Gupta, PhD. Department of Urology, Case Western Reserve University, 10900 Euclid Avenue, Cleveland, OH 44106, USA. Email: [email protected]. Provenance: This is an invited Editorial commissioned by Section Editor Xiao Li (Department of Urologic Surgery, The Affiliated Cancer Hospital of Jiangsu Province of Nanjing Medical University, Nanjing, China). Comment on: Ganaie AA, Beigh FH, Astone M, et al. BMI1 Drives Metastasis of Prostate Cancer in Caucasian and African-American Men and Is A Potential Therapeutic Target: Hypothesis Tested in Race-specific Models. Clin Cancer Res 2018. [Epub ahead of print]. Received: 12 October 2018; Accepted: 02 November 2018; Published: 09 November 2018. doi: 10.21037/amj.2018.11.01 View this article at: http://dx.doi.org/10.21037/amj.2018.11.01 A recent publication by Ganaie et al. (1) in Clinical Cancer sequence between various species (4). The human BMI1 Research (doi: 10.1158/1078-0432.CCR-18-1394, 2018) gene is localized on chromosome 10 (10p11.23) (Figure 1A). demonstrate BMI1 as a potential driver of metastasis in The BMI1 protein comprises of 326 amino acids having prostate cancer.
    [Show full text]
  • RNF2 Polyclonal Antibody Catalog No: Tcba9469
    Web: www.taiclone.com Tel: +886-2-2735-9682 Email: [email protected] RNF2 Polyclonal Antibody Catalog No: tcba9469 Available Sizes Size: 50ul Size: 100ul Size: 200ul Specifications Application: WB,IHC,IP,ChIP Research Area: ,Epigenetics, Species Reactivity: Human,Mouse,Rat Host Species: Rabbit Isotype: IgG Form: Liquid Storage Buffer: Buffer: PBS with 0.02% sodium azide, 50% glycerol, pH7.3. Recommended Dilution: WB 1:500 - 1:2000 IHC 1:50 - 1:200 IP 1:50 - 1:200 ChIP 1:50 - 1:200 Copyright 2021 Taiclone Biotech Corp. Web: www.taiclone.com Tel: +886-2-2735-9682 Email: [email protected] Storage Instruction: Store at -20℃. Avoid freeze / thaw cycles. Alternative Names: BAP-1;BAP1;DING;HIPI3;RING1B;RING2 SwissProt: Q99496 Gene ID: 6045 (human); Calculated Molecular Weight: 29kDa/37kDa Purification: Affinity purification Cellular Location: Chromosome,Nucleus, Product Description Polycomb group (PcG) of proteins form the multiprotein complexes that are important for the transcription repression of various genes involved in development and cell proliferation. The protein encoded by this gene is one of the PcG proteins. It has been shown to interact with, and suppress the activity of, transcription factor CP2 (TFCP2/CP2). Studies of the mouse counterpart suggested the involvement of this gene in the specification of anterior-posterior axis, as well as in cell proliferation in early development. This protein was also found to interact with huntingtin interacting protein 2 (HIP2), an ubiquitin-conjugating enzyme, and possess ubiquitin ligase activity. Copyright 2021 Taiclone Biotech Corp. Web: www.taiclone.com Tel: +886-2-2735-9682 Email: [email protected] Western blot analysis of extracts of various cell lines, using RNF2 antibody at 1:1000 Immunohistochemistry of paraffin-embedded rat lung using RNF2 antibody at dilution.
    [Show full text]
  • Radiosensitization of Esophageal Carcinoma Cells by Knockdown of RNF2 Expression
    INTERNATIONAL JOURNAL OF ONCOLOGY 48: 1985-1996, 2016 Radiosensitization of esophageal carcinoma cells by knockdown of RNF2 expression XING-XIAO YANG1, MING MA2, MEI-XIANG SANG3, XUE-XIAO WANG4, HENG SoNG1, ZHI-KUN LIU1 and SHu-CHAI ZHu1 Departments of 1Radiation Oncology, 2Clinical Laboratory, 3Research Centre, Department of Biotherapy, and 4Division of Cancer Biotherapy, The Fourth Hospital of Hebei Medical university, Shijiazhuang, Hebei 050011, P.R. China Received December 4, 2015; Accepted January 26, 2016 DoI: 10.3892/ijo.2016.3404 Abstract. Radiotherapy has been widely used for the treat- RNF2. Expression of the short-hairpin RNA is also correlated ment of cancer patients, especially for esophageal cancer with the upregulation of p16 and Bax, and the downregula- patients. Ring finger protein 2 (RNF2) plays an important tion of cyclin D2, cyclin-dependent kinase (CDK)-4, H2AX role in promoting the growth of cancer cells after expo- and Bcl-2. RNF2 gene knockdown induces radiosensitivity sure to irradiation. The present study aims to characterize of esophageal cancer cells in vitro and significantly inhibits the proliferative effects of RNF2 on cancer cells, and its the growth of tumor cells. The mechanisms include inducing mechanisms on the growth of esophageal cancer cells. the cell cycle arrest at G0/G1 phase and promoting apoptosis. We demonstrate that expression of RNF2 was markedly upregulated in esophageal cancer cell lines and surgically Introduction resected cancer specimens. In addition, RNF2 expression level is positively correlated with the presence of tumor Esophageal carcinoma is one of alimentary canal malignan- size, lymph node metastases and negatively correlated with cies with high incidence of approximately 0.3104 million new patient survival rates, suggesting that it plays an important malignancies worldwide each year.
    [Show full text]