The Extensive Proliferation of Human Cancer Cells with Ever-Shorter

Total Page:16

File Type:pdf, Size:1020Kb

The Extensive Proliferation of Human Cancer Cells with Ever-Shorter THE EXTENSIVE PROLIFERATION OF HUMAN CANCER CELLS WITH EVER-SHORTER TELOMERES Rebecca Dagg A thesis submitted in fulfilment of the requirements for the degree of Doctor of Philosophy Faculty of Medicine The University of Sydney 2017 1 Statement of Originality This is to certify that to the best of my knowledge, the content of this thesis is my own work. This thesis has not been submitted for any degree or other purposes. I certify that the intellectual content of this thesis is the product of my own work and that all the assistance received in preparing this thesis and sources have been acknowledged. Rebecca Dagg i Abstract Cellular immortalisation is currently regarded as an essential step in malignant transformation and is consequently considered a hallmark of cancer. Acquisition of replicative immortality is achieved by activation of a telomere lengthening mechanism (TLM), either telomerase or the alternative lengthening of telomeres (ALT), to counter normal telomere attrition. However, a substantial proportion of glioblastomas, liposarcomas, retinoblastomas, and osteosarcomas are reported to be TLM-negative. The lack of serial untreated malignant human tumour samples over time has made it impossible to examine telomere length over time and therefore determine whether they are truly TLM-deficient, or whether this is the result of false-negative assays. Here we describe a subset (11%) of high-risk neuroblastomas (NB; MYCN-amplified or metastatic disease) that lack evidence of any significant TLM activity despite a 51% 5-year mortality rate. NB cell lines (COG-N-291 and LA-N-6) derived from such tumours proliferated for 500 population doublings (PDs) with ever-shorter telomeres (EST). COG-N-291 and LA-N-6 cells had exceptionally long and heterogeneous telomere lengths as measured by terminal restriction fragment (TRF) analysis (mean TRF of 31 and 37.8 kb, respectively) and telomere fluorescence in situ hybridisation. Both cell lines were telomerase negative during culturing and did not have elevated markers of ALT or associated gene mutations. The telomeres of these cells shortened by 80 and 55 bases/PD, consistent with telomere attrition due to normal cell division, but did not reach senescence after 500 PDs in culture. This is conclusive evidence that cells from highly malignant, lethal tumours are able to undergo continuous proliferation in spite of an EST phenotype. The EST phenotype was rescued by activation of telomerase (via transduction with hTERT expression constructs) or ALT (spontaneous occurrence of a nonsense TP53 mutation, followed by spontaneous activation of ALT after 100 PDs). We also found that NB EST cells are very sensitive to topoisomerase I inhibitors indicating the potential to target the EST phenotype with topoisomerase I inhibitors in high-risk NB. ii Acknowledgements The first person that I need to thank is my supervisor Dr Loretta Lau. I cannot express how grateful I am that you took a chance and employed me eight years ago. I am the scientist that I am today because of you. Thank you for your guidance and support and putting up with short deadlines with my writing. You have been a great teacher, friend and mentor and I couldn’t have asked for a better supervisor. I would also like to thank my co-supervisor Prof. Roger Reddel. I am constantly astounded by your replies to emails at all times of the day and I am so grateful for your guidance and insights. Thank you for always having an open door, because of you I have learnt to be a better scientist and critical thinker. The success of my PhD is in large part due to the support that I have received from both of my supervisors. To all of the members of the CCRU past and present you have made the long hours in the lab far more enjoyable and become some of my dearest friends. I would especially like to thank Vanita for always answering my questions (no matter how many times I had to ask about the same form), Belinda for always offering me advice with a smile, and lunches and dinners spent laughing with Radikha, Jess, Cuc, Kylie, Michael, Greg, Nick, Amanda, Oksana, Natalie, Namratha, Camilla, and Yu Wooi. Yuyan your support has helped me more than I can express and I will miss being able to chat with you at the bench. To Peta, Sarah, Naz, Kaitlin and Amy I can’t believe we decided to start Stress whilst doing our PhD but I’m so proud of what we achieved! Ladies you have become some of my dearest friends and I can’t imagine having survived the last few years without you. I also need to thank the past and present members of the cancer research and telomere length regulation units at CMRI for all of the advice and assistance I’ve received which have made this a better thesis. During my PhD I have spent more time in the lab than anywhere else and I would like to thank all of my friends and family for being so understanding about the events that I’ve missed, left early or been late to, or had to reschedule around experiments. You have all been so understanding and I iii cannot say how much I love you and appreciate your support. I would especially like to thank Chris and my grandparents for being an unending source of support, you have helped me become who I am today and I love you more than I can say. To Mum and Katherine, I don’t think that there are words to say what you mean to me, but to start with I love you. You have seen me at my best and worst during the PhD and done everything you can to make sure that I’m still sane at the end of it. Thank you for always listening, being there when I need you, and doing everything from eating cake with me to getting posters printed when I was on the other side of the world. I never doubted that I would get to the end of this because of your unending love, support and encouragement. In many ways, I think of this as something that we have achieved together, so thank you for getting onto this rollercoaster with me and helping me become the person that I am today. iv Publications Dagg RA, Pickett HA, Neumann AA, Napier CE, Henson JD, Teber ET, Arthur JW, Reynolds CP, Murray J, Haber M, Lau L MS, Reddel RR (2017). Extensive proliferation of human cancer cells with ever-shorter telomeres. Cell Reports. Vol 20;19(12): 2544-2556. Hayward NK, Wilmott JS, Waddell N, Johansson PA, Field MA, Nones K, Patch A, Kakavand H, Alexandrov LB, Burke H, Jakrot V, Kazakoff S, Holmes O, Leonard C, Sabarinathan R, Mularoni L, Wood S, Xu Q, Waddell N, Tembe V, Pupo GM, Paoli-Iseppi RD, Vilain RE, Shang P, Lau L MS, Dagg RA, Schramm S, Pritchard A, Dutton-Regester K, Newll F, Fitzgerald A, Shang CA, Grimmond SM, Pickett HA, Jean Y. Yang JY, Stretch JR, Behren A, Kefford RF, Hersey P, Long GV, Cebon J, Shackleton M, Spillane AJ, Saw R PM, Lopez-Bigas N, Pearson JV, Thompson JF, Scolyer RA, Mann GJ (2017). Whole genome landscapes of major melanoma subtypes. Nature. Vol 11;545(7653): 175-180. Bradbury P, Turner K, Mitchell C, Griffin K, Lau L, Dagg RA, Taran E, Cooper-White J, Fabry B, O’Neill GM (2017). The focal adhesion targeting (FAT) domain of p130 Crk associated substrate (p130Cas) confers mechanosensing function. Journal of Cell Science. Vol 1;130(7): 1263-1273. Scarpa A, Chang DK, Nones k, Corbo V, Patch A, Bailey P, Lawlor RT, Johns AL, Miller DK, Mafficini A, Rusev B, Scardoni M, Antonello D, Barbi S, Sikora KO, Cingarlini S, Vicentini C, Simpson S, Quinn MCJ, Bruxner TJC, Christ AN, Harliwong I, Idrisoglu S, Manning S, Nourse C, Nourbakhsh E, Wilson PJ, Anderson MJ, Fink JL, Newell F, Waddell N, Holmes O, Kazakoff SH, Leonard C, Wood S, Xu Q, Nagaraj SH, Amato E, Dalai I, Bersani S, Cataldo I, Capelli P, Davì MV, Landoni L, Malpaga A, Miotto M, Whitehall V, Leggett B, Khanna KK, Harris J, Jones MD, Humphris J, Chantrill LA, Chin V, Nagril A, Pajic M, Scarlett v CJ, Pinho A, Rooman I, Toon C, Wu J, Pinese M, Cowley M, Giry-Laterriere M, Mawson A, Humphrey ES, Colvin EK, Chou A, Lovell JA, Jamieson NB, Duthie F, Gingras M, Muzny D, Dagg RA, Lau LMS, Lee M, Pickett HA, Reddel RR, Samra JS, Kench JG, Merrett ND, Epari K, Nguyen NQ, Zeps N, Falconi M, Simbolo M, Butturini G, Fassan M, Australian Pancreatic Cancer Genome Initiative, Gill AJ, Wheeler DA, Gibbs RA, Musgrove EA, Bassi C, Tortora G, Pederzoli P, Pearson JV, Waddell N, Biankin AV and Grimmond SM (2017). Whole-genome landscape of pancreatic neuroendocrine tumours. Nature. Vol 543: 65–71. Henson JD, Lau L MS, Koch S, Martin La Rotta N, Dagg RA, Reddel RR (2016). The C-circle assay for Alternative-Lengthening-of-Telomeres activity. Methods. http://dx.doi.org/10.1016/j.ymeth.2016. 08.01 Farooqi AS, Dagg RA, Choi LM R, Shay JW, Reynolds P, Lau L MS (2014). Alternative Lengthening of Telomeres in neuroblastoma cell lines is associated with a lack of MYCN genomic amplification and with p53 pathway aberrations. Journal of Neuro-Oncology. Vol 119(1): DOI:10.1007/s11060-014- 1456-8. Lee M, Hills M, Conomos D, Stutz M, Dagg RA, Lau L MS, Reddel RR, Pickett HA (2014). Telomere extension by telomerase and ALT generates variant repeats by mechanistically distinct processes.
Recommended publications
  • How Shelterin Protects Mammalian Telomeres 303 ANRV361-GE42-15 ARI 3 October 2008 10:10 (See 3' 5' TRF2 S Mplex
    ANRV361-GE42-15 ARI 3 October 2008 10:10 ANNUAL How Shelterin Protects REVIEWS Further Click here for quick links to Annual Reviews content online, Mammalian Telomeres including: • Other articles in this volume 1 • Top cited articles Wilhelm Palm and Titia de Lange • Top downloaded articles • Our comprehensive search Laboratory for Cell Biology and Genetics, The Rockefeller University, New York, NY 10021; email: [email protected] 1Current address: Max Planck Institute of Molecular Cell Biology and Genetics, 01307 Dresden, Germany Annu. Rev. Genet. 2008. 42:301–34 Key Words First published online as a Review in Advance on ATM, ATR, cancer, NHEJ, HR August 4, 2008 by Rockefeller University on 10/13/09. For personal use only. The Annual Review of Genetics is online at Abstract genet.annualreviews.org The genomes of prokaryotes and eukaryotic organelles are usually cir- This article’s doi: cular as are most plasmids and viral genomes. In contrast, the nuclear Annu. Rev. Genet. 2008.42:301-334. Downloaded from arjournals.annualreviews.org 10.1146/annurev.genet.41.110306.130350 genomes of eukaryotes are organized on linear chromosomes, which Copyright c 2008 by Annual Reviews. require mechanisms to protect and replicate DNA ends. Eukaryotes All rights reserved navigate these problemswith the advent of telomeres, protective nucle- 0066-4197/08/1201-0301$20.00 oprotein complexes at the ends of linear chromosomes, and telomerase, the enzyme that maintains the DNA in these structures. Mammalian telomeres contain a specific protein complex, shelterin, that functions to protect chromosome ends from all aspects of the DNA damage re- sponse and regulates telomere maintenance by telomerase.
    [Show full text]
  • Pan-Cancer Analysis of Alternative Lengthening of Telomere Activity
    cancers Article Pan-Cancer Analysis of Alternative Lengthening of Telomere Activity Ji-Yong Sung 1,2, Hee-Woong Lim 3, Je-Gun Joung 1,* and Woong-Yang Park 1,2,4,* 1 Samsung Genome Institute, Samsung Medical Center, Seoul 06351, Korea; [email protected] 2 Department of Health Science and Technology, Samsung Advanced Institute of Health Science and Technology, Sungkyunkwan University, Seoul 06351, Korea 3 Division of Biomedical Informatics, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH 45229, USA; [email protected] 4 Department of Molecular Cell Biology, School of Medicine, Sungkyunkwan University, Seoul 06351, Korea * Correspondence: [email protected] (J.-G.J.); [email protected] (W.-Y.P.); Tel.: +82-2-3410-1706 (J.-G.J.); +82-2-3410-6128 (W.-Y.P.); Fax: +82-2-2148-9819 (W.-Y.P.) Received: 30 June 2020; Accepted: 5 August 2020; Published: 7 August 2020 Abstract: Alternative lengthening of telomeres (ALT) is a telomerase-independent mechanism that extends telomeres in cancer cells. It influences tumorigenesis and patient survival. Despite the clinical significance of ALT in tumors, the manner in which ALT is activated and influences prognostic outcomes in distinct cancer types is unclear. In this work, we profiled distinct telomere maintenance mechanisms (TMMs) using 8953 transcriptomes of 31 different cancer types from The Cancer Genome Atlas (TCGA). Our results demonstrated that approximately 29% of cancer types display high ALT activity with low telomerase activity in the telomere-lengthening group. Among the distinct ALT mechanisms, homologous recombination was frequently observed in sarcoma, adrenocortical carcinoma, and kidney chromophobe. Five cancer types showed a significant difference in survival in the presence of high ALT activity.
    [Show full text]
  • Ailanthone Inhibits Non-Small Cell Lung Cancer Cell Growth Through Repressing DNA Replication Via Downregulating RPA1
    FULL PAPER British Journal of Cancer (2017) 117, 1621–1630 | doi: 10.1038/bjc.2017.319 Keywords: ailanthone; non-small cell lung cancer; DNA replication; RPA1; Chinese medicine Ailanthone inhibits non-small cell lung cancer cell growth through repressing DNA replication via downregulating RPA1 Zhongya Ni1, Chao Yao1, Xiaowen Zhu1, Chenyuan Gong1, Zihang Xu2, Lixin Wang3, Suyun Li4, Chunpu Zou2 and Shiguo Zhu*,1,3 1Laboratory of Integrative Medicine, School of Basic Medical Sciences, Shanghai University of Traditional Chinese Medicine, 1200 Cai Lun Rd, Shanghai 201203, PR China; 2Department of Internal Classic of Medicine, School of Basic Medical Sciences, Shanghai University of Traditional Chinese Medicine, 1200 Cai Lun Rd, Shanghai 201203, PR China; 3Department of Immunology and Pathogenic Biology, School of Basic Medical Sciences, Shanghai University of Traditional Chinese Medicine, 1200 Cai Lun Rd, Shanghai 201203, PR China and 4Department of Pathology, School of Basic Medical Sciences, Shanghai University of Traditional Chinese Medicine, 1200 Cai Lun Rd, Shanghai 201203, PR China Background: The identification of bioactive compounds from Chinese medicine plays a crucial role in the development of novel reagents against non-small cell lung cancer (NSCLC). Methods: High throughput screening assay and analyses of cell growth, cell cycle, apoptosis, cDNA microarray, BrdU incorporation and gene expression were performed. Results: Ailanthone (Aila) suppressed NSCLC cell growth and colony formation in vitro and inhibited NSCLC tumour growth in subcutaneously xenografted and orthotopic lung tumour models, leading to prolonged survival of tumour-bearing mice. Moreover, Aila induced cell cycle arrest in a dose-independent manner but did not induce apoptosis in all NSCLC cells.
    [Show full text]
  • Iron Depletion Reduces Abce1 Transcripts While Inducing The
    Preprints (www.preprints.org) | NOT PEER-REVIEWED | Posted: 22 October 2019 doi:10.20944/preprints201910.0252.v1 1 Research Article 2 Iron depletion Reduces Abce1 Transcripts While 3 Inducing the Mitophagy Factors Pink1 and Parkin 4 Jana Key 1,2, Nesli Ece Sen 1, Aleksandar Arsovic 1, Stella Krämer 1, Robert Hülse 1, Suzana 5 Gispert-Sanchez 1 and Georg Auburger 1,* 6 1 Experimental Neurology, Goethe University Medical School, 60590 Frankfurt am Main; 7 2 Faculty of Biosciences, Goethe-University Frankfurt am Main, Germany 8 * Correspondence: [email protected] 9 10 Abstract: Lifespan extension was recently achieved in Caenorhabditis elegans nematodes by 11 mitochondrial stress and mitophagy, triggered via iron depletion. Conversely in man, deficient 12 mitophagy due to Pink1/Parkin mutations triggers iron accumulation in patient brain and limits 13 survival. We now aimed to identify murine fibroblast factors, which adapt their mRNA expression 14 to acute iron manipulation, relate to mitochondrial dysfunction and may influence survival. After 15 iron depletion, expression of the plasma membrane receptor Tfrc with its activator Ireb2, the 16 mitochondrial membrane transporter Abcb10, the heme-release factor Pgrmc1, the heme- 17 degradation enzyme Hmox1, the heme-binding cholesterol metabolizer Cyp46a1, as well as the 18 mitophagy regulators Pink1 and Parkin showed a negative correlation to iron levels. After iron 19 overload, these factors did not change expression. Conversely, a positive correlation of mRNA levels 20 with both conditions of iron availability was observed for the endosomal factors Slc11a2 and Steap2, 21 as well as for the iron-sulfur-cluster (ISC)-containing factors Ppat, Bdh2 and Nthl1.
    [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]
  • Cell Growth-Regulated Expression of Mammalian MCM5 and MCM6 Genes Mediated by the Transcription Factor E2F
    Oncogene (1999) 18, 2299 ± 2309 ã 1999 Stockton Press All rights reserved 0950 ± 9232/99 $12.00 http://www.stockton-press.co.uk/onc Cell growth-regulated expression of mammalian MCM5 and MCM6 genes mediated by the transcription factor E2F Kiyoshi Ohtani1, Ritsuko Iwanaga1, Masataka Nakamura*,1, Masa-aki Ikeda2, Norikazu Yabuta3, Hiromichi Tsuruga3 and Hiroshi Nojima3 1Human Gene Sciences Center, Tokyo Medical and Dental University, Tokyo 113-8510, Japan 2Department of Developmental Biology, Graduate School of Dentistry, Tokyo Medical and Dental University, Tokyo 113-8549, Japan; 3Department of Molecular Genetics, Research Institute for Microbial Diseases, Osaka University, Suita 565-0871, Japan Initiation of DNA replication requires the function of family (MCM2-7) that have been identi®ed in yeast, MCM gene products, which participate in ensuring that Xenopus, and human. Mcm proteins seem to regulate DNA replication occurs only once in the cell cycle. the initiation at the replication origin where the loading Expression of all mammalian genes of the MCM family of the proteins onto the origin recognition complex is induced by growth stimulation, unlike yeast, and the (ORC) is regulated by Cdc6 and cyclin-dependent mRNA levels peak at G1/S boundary. In this study, we kinases (Donovan et al., 1997; Tanaka et al., 1997). examined the transcriptional activities of isolated human However, the mechanism(s) by which Mcm proteins MCM gene promoters. Human MCM5 and MCM6 control the initiation of DNA replication remains promoters with mutation in the E2F sites failed in unclear. promoter regulation following serum stimulation and Xenopus Mcm proteins seem to be able to access exogenous E2F expression.
    [Show full text]
  • Supplementary Table S5. Differentially Expressed Gene Lists of PD-1High CD39+ CD8 Tils According to 4-1BB Expression Compared to PD-1+ CD39- CD8 Tils
    BMJ Publishing Group Limited (BMJ) disclaims all liability and responsibility arising from any reliance Supplemental material placed on this supplemental material which has been supplied by the author(s) J Immunother Cancer Supplementary Table S5. Differentially expressed gene lists of PD-1high CD39+ CD8 TILs according to 4-1BB expression compared to PD-1+ CD39- CD8 TILs Up- or down- regulated genes in Up- or down- regulated genes Up- or down- regulated genes only PD-1high CD39+ CD8 TILs only in 4-1BBneg PD-1high CD39+ in 4-1BBpos PD-1high CD39+ CD8 compared to PD-1+ CD39- CD8 CD8 TILs compared to PD-1+ TILs compared to PD-1+ CD39- TILs CD39- CD8 TILs CD8 TILs IL7R KLRG1 TNFSF4 ENTPD1 DHRS3 LEF1 ITGA5 MKI67 PZP KLF3 RYR2 SIK1B ANK3 LYST PPP1R3B ETV1 ADAM28 H2AC13 CCR7 GFOD1 RASGRP2 ITGAX MAST4 RAD51AP1 MYO1E CLCF1 NEBL S1PR5 VCL MPP7 MS4A6A PHLDB1 GFPT2 TNF RPL3 SPRY4 VCAM1 B4GALT5 TIPARP TNS3 PDCD1 POLQ AKAP5 IL6ST LY9 PLXND1 PLEKHA1 NEU1 DGKH SPRY2 PLEKHG3 IKZF4 MTX3 PARK7 ATP8B4 SYT11 PTGER4 SORL1 RAB11FIP5 BRCA1 MAP4K3 NCR1 CCR4 S1PR1 PDE8A IFIT2 EPHA4 ARHGEF12 PAICS PELI2 LAT2 GPRASP1 TTN RPLP0 IL4I1 AUTS2 RPS3 CDCA3 NHS LONRF2 CDC42EP3 SLCO3A1 RRM2 ADAMTSL4 INPP5F ARHGAP31 ESCO2 ADRB2 CSF1 WDHD1 GOLIM4 CDK5RAP1 CD69 GLUL HJURP SHC4 GNLY TTC9 HELLS DPP4 IL23A PITPNC1 TOX ARHGEF9 EXO1 SLC4A4 CKAP4 CARMIL3 NHSL2 DZIP3 GINS1 FUT8 UBASH3B CDCA5 PDE7B SOGA1 CDC45 NR3C2 TRIB1 KIF14 TRAF5 LIMS1 PPP1R2C TNFRSF9 KLRC2 POLA1 CD80 ATP10D CDCA8 SETD7 IER2 PATL2 CCDC141 CD84 HSPA6 CYB561 MPHOSPH9 CLSPN KLRC1 PTMS SCML4 ZBTB10 CCL3 CA5B PIP5K1B WNT9A CCNH GEM IL18RAP GGH SARDH B3GNT7 C13orf46 SBF2 IKZF3 ZMAT1 TCF7 NECTIN1 H3C7 FOS PAG1 HECA SLC4A10 SLC35G2 PER1 P2RY1 NFKBIA WDR76 PLAUR KDM1A H1-5 TSHZ2 FAM102B HMMR GPR132 CCRL2 PARP8 A2M ST8SIA1 NUF2 IL5RA RBPMS UBE2T USP53 EEF1A1 PLAC8 LGR6 TMEM123 NEK2 SNAP47 PTGIS SH2B3 P2RY8 S100PBP PLEKHA7 CLNK CRIM1 MGAT5 YBX3 TP53INP1 DTL CFH FEZ1 MYB FRMD4B TSPAN5 STIL ITGA2 GOLGA6L10 MYBL2 AHI1 CAND2 GZMB RBPJ PELI1 HSPA1B KCNK5 GOLGA6L9 TICRR TPRG1 UBE2C AURKA Leem G, et al.
    [Show full text]
  • POLE2 Knockdown Reduce Tumorigenesis in Esophageal Squamous Cells Yongjun Zhu, Gang Chen, Yang Song, Zhiming Chen* and Xiaofeng Chen*
    Zhu et al. Cancer Cell Int (2020) 20:388 https://doi.org/10.1186/s12935-020-01477-4 Cancer Cell International PRIMARY RESEARCH Open Access POLE2 knockdown reduce tumorigenesis in esophageal squamous cells Yongjun Zhu, Gang Chen, Yang Song, Zhiming Chen* and Xiaofeng Chen* Abstract Background: Esophageal squamous cell carcinoma (ESCC) is one of the most frequent malignant tumors originated from digestive system around the world and the treatment was limited by the unclear mechanism. DNA polymerase epsilon 2, accessory subunit (POLE2) is involved in DNA replication, repair, and cell cycle control, whose association with ESCC is still not clear. Methods: In this study, the expression level of POLE2 in ESCC tissues was detected by IHC. The POLE2 knockdown cell line was constructed, identifed by qPCR and western blot and used for detecting cellular functions and con- structing xenotransplantation mice model. MTT Assay, colony formation assay, fow cytometry, wound-healing assay and Transwell assay were used to detected cell proliferation, apoptosis and migration. Results: We frstly identifed that the expression of POLE2 was overexpressed in ESCC. Moreover, the high expres- sion of POLE2 can predict the tumor deterioration and poor prognosis of ESCC patients. Additionally, downregulation of POLE2 was involved in ESCC progression by promoting proliferation, migration, and inhibiting apoptosis in vitro. In vivo studies proved that POLE2 was positively correlated with ESCC tumor formation, which was consistent with the results in vitro. We also illuminated that POLE2 knockdown upregulated pro-apoptotic proteins (Bax, Caspase3, CD40L, FasL, IGFBP-5 and P21) and downregulated anti-apoptotic proteins (CLAP-2, IGF-I and sTNF-R2).
    [Show full text]
  • ORC3 (1D6): Sc-23888
    SANTA CRUZ BIOTECHNOLOGY, INC. ORC3 (1D6): sc-23888 BACKGROUND STORAGE The initiation of DNA replication is a multi-step process that depends on the Store at 4° C, **DO NOT FREEZE**. Stable for one year from the date of formation of pre-replication complexes, which trigger initiation. Among the shipment. Non-hazardous. No MSDS required. proteins required for establishing these complexes are the origin recognition complex (ORC) proteins. ORC proteins bind specifically to origins of replication DATA where they serve as scaffold for the assembly of additional initiation factors. A B Human ORC subunits 1-6 are expressed in the nucleus of proliferating cells AB C DE and tissues, such as the testis. ORC1 and ORC2 are both expressed at equiv- 113 K – 89 K – alent concentrations throughout the cell cycle; however, only ORC2 remains < ORC3 stably bound to chromatin. ORC4 and ORC6 are also expressed constantly throughout the cell cycle. ORC2, ORC3, ORC4 and ORC5 form a core complex 49 K – upon which ORC6 and ORC1 assemble. The formation of this core complex suggests that ORC proteins play a crucial role in the G1-S transition in mam- malian cells. ORC3 (1D6): sc-23888. Western blot analysis of ORC3 ORC3 (1D6): sc-23888. Immunoperoxidase staining of expression in HeLa (A), Raji (B), Jurkat (C), WI-38 (D) formalin fixed, paraffin-embedded human ovary tissue CHROMOSOMAL LOCATION and A-549 (E) whole cell lysates. showing nuclear localization (A,B). Genetic locus: ORC3L (human) mapping to 6q15. SELECT PRODUCT CITATIONS SOURCE 1. Braden, W.A., et al. 2006. Distinct action of the retinoblastoma pathway ORC3 (1D6) is a rat monoclonal antibody raised against partially purified on the DNA replication machinery defines specific roles for cyclin-depen- His-tagged bacterially expressed fusion protein corresponding to human ORC3.
    [Show full text]
  • Systemic Analysis of the DNA Replication Regulators Origin Recognition Complex in Lung Adenocarcinomas Identifes Prognostic and Expression Signifcance
    Systemic Analysis of the DNA Replication Regulators Origin Recognition Complex in Lung Adenocarcinomas Identies Prognostic and Expression Signicance Juan Chen University of South China Juan Zou University of South China Juan Zeng University of South China Tian Zeng University of South China Qi-hao Hu University of South China Jun-hui Bai University of South China Min Tang University of South China Yu-kun Li ( [email protected] ) University of South China https://orcid.org/0000-0002-8517-9075 Primary research Keywords: lung adenocarcinomas, ORC complex, public databases, prognostic value, comprehensive bioinformatics Posted Date: May 11th, 2021 DOI: https://doi.org/10.21203/rs.3.rs-487176/v1 License: This work is licensed under a Creative Commons Attribution 4.0 International License. Read Full License Page 1/24 Abstract Background: Origin recognition complex (ORC) 1, ORC2, ORC3, ORC4, ORC5 and ORC6, form a replication- initiator complex to mediate DNA replication, which play a key role in carcinogenesis, while their role in lung adenocarcinomas (LUAD) remains poorly understood. Methods: We conrmed the transcriptional and post-transcriptional levels, DNA alteration, DNA methylation, miRNA network, protein structure, PPI network, functional enrichment, immune inltration and prognostic value of ORCs in LUAD based on Oncomine, GEPIA, HPA, cBioportal, TCGA, GeneMANIA, Metascape, KM-plot, GENT2, and TIMER database. Results: ORC mRNA and protein were both enhanced obviously based on Oncomine, Ualcan, GEPIA, TCGA and HPA database. Furthermore, ORC1 and ORC6 have signicant prognostic values for LUAD patients based on GEPIA database. Protein structure, PPI network, functional enrichment and immune inltration analysis indicated that ORC complex cooperatively accelerate the LUAD development by promoting DNA replication, cellular senescence and metabolic process.
    [Show full text]
  • Bioinformatics-Based Screening of Key Genes for Transformation of Liver
    Jiang et al. J Transl Med (2020) 18:40 https://doi.org/10.1186/s12967-020-02229-8 Journal of Translational Medicine RESEARCH Open Access Bioinformatics-based screening of key genes for transformation of liver cirrhosis to hepatocellular carcinoma Chen Hao Jiang1,2, Xin Yuan1,2, Jiang Fen Li1,2, Yu Fang Xie1,2, An Zhi Zhang1,2, Xue Li Wang1,2, Lan Yang1,2, Chun Xia Liu1,2, Wei Hua Liang1,2, Li Juan Pang1,2, Hong Zou1,2, Xiao Bin Cui1,2, Xi Hua Shen1,2, Yan Qi1,2, Jin Fang Jiang1,2, Wen Yi Gu4, Feng Li1,2,3 and Jian Ming Hu1,2* Abstract Background: Hepatocellular carcinoma (HCC) is the most common type of liver tumour, and is closely related to liver cirrhosis. Previous studies have focussed on the pathogenesis of liver cirrhosis developing into HCC, but the molecular mechanism remains unclear. The aims of the present study were to identify key genes related to the transformation of cirrhosis into HCC, and explore the associated molecular mechanisms. Methods: GSE89377, GSE17548, GSE63898 and GSE54236 mRNA microarray datasets from Gene Expression Omni- bus (GEO) were analysed to obtain diferentially expressed genes (DEGs) between HCC and liver cirrhosis tissues, and network analysis of protein–protein interactions (PPIs) was carried out. String and Cytoscape were used to analyse modules and identify hub genes, Kaplan–Meier Plotter and Oncomine databases were used to explore relationships between hub genes and disease occurrence, development and prognosis of HCC, and the molecular mechanism of the main hub gene was probed using Kyoto Encyclopedia of Genes and Genomes(KEGG) pathway analysis.
    [Show full text]
  • Supplementary Table S1. Correlation Between the Mutant P53-Interacting Partners and PTTG3P, PTTG1 and PTTG2, Based on Data from Starbase V3.0 Database
    Supplementary Table S1. Correlation between the mutant p53-interacting partners and PTTG3P, PTTG1 and PTTG2, based on data from StarBase v3.0 database. PTTG3P PTTG1 PTTG2 Gene ID Coefficient-R p-value Coefficient-R p-value Coefficient-R p-value NF-YA ENSG00000001167 −0.077 8.59e-2 −0.210 2.09e-6 −0.122 6.23e-3 NF-YB ENSG00000120837 0.176 7.12e-5 0.227 2.82e-7 0.094 3.59e-2 NF-YC ENSG00000066136 0.124 5.45e-3 0.124 5.40e-3 0.051 2.51e-1 Sp1 ENSG00000185591 −0.014 7.50e-1 −0.201 5.82e-6 −0.072 1.07e-1 Ets-1 ENSG00000134954 −0.096 3.14e-2 −0.257 4.83e-9 0.034 4.46e-1 VDR ENSG00000111424 −0.091 4.10e-2 −0.216 1.03e-6 0.014 7.48e-1 SREBP-2 ENSG00000198911 −0.064 1.53e-1 −0.147 9.27e-4 −0.073 1.01e-1 TopBP1 ENSG00000163781 0.067 1.36e-1 0.051 2.57e-1 −0.020 6.57e-1 Pin1 ENSG00000127445 0.250 1.40e-8 0.571 9.56e-45 0.187 2.52e-5 MRE11 ENSG00000020922 0.063 1.56e-1 −0.007 8.81e-1 −0.024 5.93e-1 PML ENSG00000140464 0.072 1.05e-1 0.217 9.36e-7 0.166 1.85e-4 p63 ENSG00000073282 −0.120 7.04e-3 −0.283 1.08e-10 −0.198 7.71e-6 p73 ENSG00000078900 0.104 2.03e-2 0.258 4.67e-9 0.097 3.02e-2 Supplementary Table S2.
    [Show full text]