Functional Loss of ATRX and TERC Activates Alternative Lengthening of Telomeres (ALT) in LAPC4 Prostate Cancer Cells Mindy K

Total Page:16

File Type:pdf, Size:1020Kb

Functional Loss of ATRX and TERC Activates Alternative Lengthening of Telomeres (ALT) in LAPC4 Prostate Cancer Cells Mindy K Published OnlineFirst October 14, 2019; DOI: 10.1158/1541-7786.MCR-19-0654 Genome Maintenance Molecular Cancer Research Functional Loss of ATRX and TERC Activates Alternative Lengthening of Telomeres (ALT) in LAPC4 Prostate Cancer Cells Mindy K. Graham1, Jiyoung Kim1, Joseph Da1, Jacqueline A. Brosnan-Cashman1, Anthony Rizzo1, Javier A. Baena Del Valle1, Lionel Chia1, Michael Rubenstein2, Christine Davis1, Qizhi Zheng1, Leslie Cope3, Michael Considine3, Michael C. Haffner1, Angelo M. De Marzo1,3,4, Alan K. Meeker1,3,4, and Christopher M. Heaphy1,3 Abstract A key hallmark of cancer, unlimited replication, requires LAPC-4, but not CWR22Rv1, abolishing ATRX was sufficient cancer cells to evade both replicative senescence and potentially to induce multiple ALT-associated hallmarks, including the lethal chromosomal instability induced by telomere dysfunc- presence of ALT-associated promyelocytic leukemia bodies tion. The majority of cancers overcome these critical barriers by (APB), extrachromosomal telomere C-circles, and dramatic upregulating telomerase, a telomere-specific reverse transcrip- telomere length heterogeneity. However, telomerase activity tase. However, a subset of cancers maintains telomere lengths was still present in these ATRXKO cells. Telomerase activity was by the telomerase-independent Alternative Lengthening of subsequently crippled in these LAPC-4 ATRXKO cells by intro- Telomeres (ALT) pathway. The presence of ALT is strongly ducing mutations in the TERC locus, the essential RNA com- associated with recurrent cancer-specific somatic inactivating ponent of telomerase. These LAPC-4 ATRXKO TERCmut cells mutations in the ATRX-DAXX chromatin-remodeling complex. continued to proliferate long-term and retained ALT-associated Here, we generate an ALT-positive adenocarcinoma cell line hallmarks, thereby demonstrating their reliance on the ALT following functional inactivation of ATRX and telomerase in a mechanism for telomere maintenance. telomerase-positive adenocarcinoma cell line. Inactivating mutations in ATRX were introduced using CRISPR-cas9 nickase Implications: These prostate cancer cell line models provide a into two prostate cancer cell lines, LAPC-4 (derived from a unique system to explore the distinct molecular alterations that lymph node metastasis) and CWR22Rv1 (sourced from a occur upon induction of ALT, and may be useful tools to screen xenograft established from a primary prostate cancer). In for ALT-specific therapies. Introduction incomplete DNA replication in the lagging strand (2). Once telo- meres reach a critical length, normal cells undergo p53-dependent Telomeres are nucleoprotein complexes that consist of a repet- replicative senescence or apoptosis (3). While there are rare excep- itive hexameric DNA sequence, 50-TTAGGG/30-AATCCC, that are tions without a known telomere maintenance mechanism (4, 5), bound by the shelterin protein complex. Located at the ends of telomere maintenance is required for most cancers to maintain eukaryotic chromosomes, telomeres maintain genomic stability unlimited replication while evading replicative senescence or apo- and integrity by preventing exonucleolytic degradation and recog- ptosis (6). The vast majority of cancers upregulate telomerase, a nition of the chromosomal ends as double stranded breaks by DNA telomere-specific reverse transcriptase (7); however, 10%–15% of damage response (DDR) proteins (1). Following each round of cell cancers lack telomerase activity (7, 8). division, telomeres progressively shorten as a direct result of A subset of telomerase-negative cancers maintains their telo- mere lengths by a telomere-specific mechanism of homology- directed repair called Alternative Lengthening of Telomeres (ALT; 1 Department of Pathology, Johns Hopkins University School of Medicine, Balti- refs. 9–11). The mechanism underlying ALT generates unique 2 more, Maryland. Department of Biological Sciences, University of Maryland molecular characteristics that readily distinguish ALT-positive and Baltimore County, Baltimore, Maryland. 3Department of Oncology Johns Hop- kins University School of Medicine, Baltimore, Maryland. 4Department of Urol- ALT-negative cancers. For example, ALT-positive cancers display ogy, Johns Hopkins University School of Medicine, Baltimore, Maryland. dramatic telomere length heterogeneity, manifesting both as cell- to-cell variation and intracellular heterogeneity (9). ALT-positive Note: Supplementary data for this article are available at Molecular Cancer Research Online (http://mcr.aacrjournals.org/). cancers also have ALT-associated promyelocytic leukemia (PML) bodies, called APBs. These unique nuclear structures contain Corresponding Author: Christopher M. Heaphy Johns Hopkins University School donut-shaped PML protein bodies that colocalize with telomeric of Medicine, 411 N. Caroline St., Bond St. Bldg B301, Baltimore, MD 21231. Phone: 443-287-4730; Fax. 410-592-5158; E-mail: [email protected] DNA, the shelterin components TRF1 and TRF2, and proteins involved in DNA synthesis and recombination (12, 13). In Mol Cancer Res 2019;17:1–12 addition, ALT-positive cancers are enriched for C-circles, extra- doi: 10.1158/1541-7786.MCR-19-0654 chromosomal circular DNA comprised of single-stranded C-rich Ó2019 American Association for Cancer Research. telomere DNA sequence partially overlapped by linear www.aacrjournals.org OF1 Downloaded from mcr.aacrjournals.org on September 25, 2021. © 2019 American Association for Cancer Research. Published OnlineFirst October 14, 2019; DOI: 10.1158/1541-7786.MCR-19-0654 Graham et al. complementary G-rich telomere DNA (14). Sequencing studies of (Sigma), 1% of mixture of penicillin (10,000 U/mL) and strep- molecularly characterized ALT-positive cancers from our group, tomycin (10,000 mg/mL; Quality Biological), and 1 nmol/L of and others, identified a robust association between ALT and R1881. CWR22Rv1 and PC3 were cultured in RPMI1640 (Gibco) mutations in a-thalassemia/mental retardation X-linked protein supplemented with 10% FBS and 1% penicillin/streptomycin. (ATRX) and death domain–associated protein (DAXX) in multi- U2OS was cultured in DMEM, high glucose (DMEM, Gibco) ple tumor types, including pancreatic neuroendocrine tumors supplemented with 10% FBS and 1% penicillin/streptomycin. (PanNET), sarcomas, and tumors of the central nervous All cell lines were submitted to the Genetic Resources Core Facility system (15–17). As a complex, ATRX/DAXX recognizes H3K9me3 at Johns Hopkins for Mycoplasma detection and cell line authen- in chromatin and deposits histone variant H3.3 at repetitive tication by short tandem repeat (STR) profiling using the Gene- sequences. This chromatin-remodeling function of ATRX/DAXX Print 10 kit (Promega, June 2018). is important for heterochromatin maintenance at repetitive sequences, including telomeres (18, 19). More recently, inactivat- ATRX CRISPR genome editing ing mutations in the ATP-dependent annealing helicase, SMAR- As described previously, two CRISPR Cas9 nickase guide RNAs CAL1, were also found to be tightly associated with ALT in a subset (Supplementary Table S1) were designed to target exon 9 of ATRX of ATRX and DAXX wild-type glioblastomas (20). using CRISPR Design (crispr.mit.edu, Supplementary Fig. S2). The majority of established ALT-positive cancer cell lines have The gRNAs were cloned into the GFP-expressing Cas9n plasmid, inactivated ATRX, although there are some recent excep- PX461, a gift from Feng Zhang (Addgene #48140; ref. 33). Lipo- tions (20–23). Previous attempts to induce ALT through knock- fectamine 3000 (Thermo Fisher Scientific) was used to transfect down or knockout of ATRX have largely been unsuccess- either empty vector PX461 or cotransfect both ATRX gRNA1- ful (21, 23–25). However, in a context-dependent manner, genet- PX461 and ATRX gRNA 2-PX461 into LAPC-4 and CWR22Rv1 ic knockout of ATRX or SMARCAL1 in some telomerase-positive cells. GFP-positive cells were sorted by FACS after 48 hours and glioma cell lines has induced multiple hallmarks of ALT (20, 26). 1,000 cells were plated in 150-mm dishes. Cell colonies were Thus, a constellation of genetic and epigenetic changes may be isolated using cloning cylinders (Sigma) and screened prelim- gatekeepers for permitting ALT. Interestingly, the combination of inarily for ATRX protein by immunostaining. Promising clones knocking down ATRX, knocking out DAXX, and introducing a were subsequently validated by Western blotting and Sanger mutation in TPP1 (that suppresses telomerase activity and sequencing. induces telomere-specific DNA damage) was sufficient to activate ALT in the telomerase-positive fibrosarcoma cell line HTC75 (27). CRISPR genome editing of the TERC locus Strategies to cripple telomerase via knocking out TERC in telo- A CRISPR plasmid cloned with the TERC gRNA-1 (Supplemen- merase-positive cell lines have been successful in activating ALT at tary Table S1) cloned into plasmid PX458 was a kind gift extremely low frequency in the spontaneously immortalized from Jaewon Min and colleagues (Department of Cell Biology, human lung fibroblast cell line SW39 and the lung carcinoma University of Texas Southwestern Medical Center; ref. 34). The cell line H1299 (28). original vector was a gift from Feng Zhang (Addgene #48138; We previously identified a prostate cancer case where a chro- ref. 35). Lipofectamine 3000 (Thermo Fisher Scientific) was used mosomal inversion disrupting ATRX was found in multiple to transfect TERC1-gRNA-PX458 in LAPC-4 ATRXKO cells. GFP- distant metastases, but not in
Recommended publications
  • 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]
  • 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]
  • 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]
  • Gene Expression Profiling Analysis Contributes to Understanding the Association Between Non-Syndromic Cleft Lip and Palate, and Cancer
    2110 MOLECULAR MEDICINE REPORTS 13: 2110-2116, 2016 Gene expression profiling analysis contributes to understanding the association between non-syndromic cleft lip and palate, and cancer HONGYI WANG, TAO QIU, JIE SHI, JIULONG LIANG, YANG WANG, LIANGLIANG QUAN, YU ZHANG, QIAN ZHANG and KAI TAO Department of Plastic Surgery, General Hospital of Shenyang Military Area Command, PLA, Shenyang, Liaoning 110016, P.R. China Received March 10, 2015; Accepted December 18, 2015 DOI: 10.3892/mmr.2016.4802 Abstract. The present study aimed to investigate the for NSCL/P were implicated predominantly in the TGF-β molecular mechanisms underlying non-syndromic cleft lip, signaling pathway, the cell cycle and in viral carcinogenesis. with or without cleft palate (NSCL/P), and the association The TP53, CDK1, SMAD3, PIK3R1 and CASP3 genes were between this disease and cancer. The GSE42589 data set found to be associated, not only with NSCL/P, but also with was downloaded from the Gene Expression Omnibus data- cancer. These results may contribute to a better understanding base, and contained seven dental pulp stem cell samples of the molecular mechanisms of NSCL/P. from children with NSCL/P in the exfoliation period, and six controls. Differentially expressed genes (DEGs) were Introduction screened using the RankProd method, and their potential functions were revealed by pathway enrichment analysis and Non-syndromic cleft lip, with or without cleft palate (NSCL/P) construction of a pathway interaction network. Subsequently, is one of the most common types of congenital defect and cancer genes were obtained from six cancer databases, and affects 3.4-22.9/10,000 individuals worldwide (1).
    [Show full text]
  • Polymerase Δ Deficiency Causes Syndromic Immunodeficiency with Replicative Stress
    Polymerase δ deficiency causes syndromic immunodeficiency with replicative stress Cecilia Domínguez Conde, … , Mirjam van der Burg, Kaan Boztug J Clin Invest. 2019. https://doi.org/10.1172/JCI128903. Research Article Genetics Immunology Graphical abstract Find the latest version: https://jci.me/128903/pdf The Journal of Clinical Investigation RESEARCH ARTICLE Polymerase δ deficiency causes syndromic immunodeficiency with replicative stress Cecilia Domínguez Conde,1,2 Özlem Yüce Petronczki,1,2,3 Safa Baris,4,5 Katharina L. Willmann,1,2 Enrico Girardi,2 Elisabeth Salzer,1,2,3,6 Stefan Weitzer,7 Rico Chandra Ardy,1,2,3 Ana Krolo,1,2,3 Hanna Ijspeert,8 Ayca Kiykim,4,5 Elif Karakoc-Aydiner,4,5 Elisabeth Förster-Waldl,9 Leo Kager,6 Winfried F. Pickl,10 Giulio Superti-Furga,2,11 Javier Martínez,7 Joanna I. Loizou,2 Ahmet Ozen,4,5 Mirjam van der Burg,8 and Kaan Boztug1,2,3,6 1Ludwig Boltzmann Institute for Rare and Undiagnosed Diseases, 2CeMM Research Center for Molecular Medicine of the Austrian Academy of Sciences, and 3St. Anna Children’s Cancer Research Institute (CCRI), Vienna, Austria. 4Pediatric Allergy and Immunology, Marmara University, Faculty of Medicine, Istanbul, Turkey. 5Jeffrey Modell Diagnostic Center for Primary Immunodeficiency Diseases, Marmara University, Istanbul, Turkey. 6St. Anna Children’s Hospital, Department of Pediatrics and Adolescent Medicine, Vienna, Austria. 7Center for Medical Biochemistry, Medical University of Vienna, Vienna, Austria. 8Department of Pediatrics, Laboratory for Immunology, Leiden University Medical Centre, Leiden, Netherlands. 9Department of Neonatology, Pediatric Intensive Care and Neuropediatrics, Department of Pediatrics and Adolescent Medicine, 10Institute of Immunology, Center for Pathophysiology, Infectiology and Immunology, and 11Center for Physiology and Pharmacology, Medical University of Vienna, Vienna, Austria.
    [Show full text]
  • Functional Loss of ATRX and TERC Activates Alternative Lengthening of Telomeres (ALT) in LAPC4 Prostate Cancer Cells
    Author Manuscript Published OnlineFirst on October 14, 2019; DOI: 10.1158/1541-7786.MCR-19-0654 Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited. Functional loss of ATRX and TERC activates Alternative Lengthening of Telomeres (ALT) in LAPC4 prostate cancer cells Mindy K. Graham1, Jiyoung Kim1, Joseph Da1, Jacqueline A. Brosnan-Cashman1, Anthony Rizzo1, Javier A. Baena Del Valle1, Lionel Chia1, Michael Rubenstein4, Christine Davis1, Qizhi Zheng1, Leslie Cope2, Michael Considine2, Michael C. Haffner1, Angelo M. De Marzo1,2,3, Alan K. Meeker1,2,3, and Christopher M. Heaphy1,2* 1 Department of Pathology, 2 Department of Oncology, 3 Department of Urology, Johns Hopkins University School of Medicine, Baltimore, MD 21231, USA 4 Department of Biological Sciences, University of Maryland Baltimore County, Baltimore, MD 21205, USA Running Title: Effects of ATRX and hTR loss in prostate cancer *To whom correspondence should be addressed: Christopher M. Heaphy: Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, MD 21231; [email protected] Tel. (443) 287-4730; Fax. (410) 592-5158. Keywords: ATRX, TERC, Telomeres, Alternative lengthening of telomeres, Cancer, Prostate cancer, telomerase, gene knockout, CRISPR/Cas Conflicts of Interest: None 1 Downloaded from mcr.aacrjournals.org on September 30, 2021. © 2019 American Association for Cancer Research. Author Manuscript Published OnlineFirst on October 14, 2019; DOI: 10.1158/1541-7786.MCR-19-0654 Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited. Effects of ATRX and hTR loss in prostate cancer ABSTRACT A key hallmark of cancer, unlimited replication, requires cancer cells to evade both replicative senescence and potentially lethal chromosomal instability induced by telomere dysfunction.
    [Show full text]
  • Family a and B DNA Polymerases in Cancer: Opportunities for Therapeutic Interventions
    biology Review Family A and B DNA Polymerases in Cancer: Opportunities for Therapeutic Interventions Vinit Shanbhag 1,2, Shrikesh Sachdev 2,3, Jacqueline A. Flores 2,3, Mukund J. Modak 4 and Kamalendra Singh 2,3,4,5,* 1 Department of Biochemistry, University of Missouri, Columbia, MO 65211, USA; [email protected] 2 The Christopher S. Bond Life Science Center, University of Missouri, Columbia, MO 65211, USA; [email protected] (S.S.); [email protected] (J.A.F.) 3 Molecular Microbiology and Immunology, University of Missouri, Columbia, MO 65211, USA 4 Department of Microbiology, Biochemistry and Molecular Genetics 225 Warren Street, NJ 07103, USA; [email protected] 5 Department of Laboratory Medicine, Karolinska Institutet, Stockholm 141 86, Sweden * Correspondence: [email protected]; Tel.: +1-573-882-9024 Received: 13 November 2017; Accepted: 29 December 2017; Published: 2 January 2018 Abstract: DNA polymerases are essential for genome replication, DNA repair and translesion DNA synthesis (TLS). Broadly, these enzymes belong to two groups: replicative and non-replicative DNA polymerases. A considerable body of data suggests that both groups of DNA polymerases are associated with cancer. Many mutations in cancer cells are either the result of error-prone DNA synthesis by non-replicative polymerases, or the inability of replicative DNA polymerases to proofread mismatched nucleotides due to mutations in 30-50 exonuclease activity. Moreover, non-replicative, TLS-capable DNA polymerases can negatively impact cancer treatment by synthesizing DNA past lesions generated from treatments such as cisplatin, oxaliplatin, etoposide, bleomycin, and radiotherapy. Hence, the inhibition of DNA polymerases in tumor cells has the potential to enhance treatment outcomes.
    [Show full text]
  • Microrna Modulate Alveolar Epithelial Response to Cyclic Stretch
    University of Pennsylvania ScholarlyCommons Departmental Papers (BE) Department of Bioengineering 2012 MicroRNA Modulate Alveolar Epithelial Response to Cyclic Stretch Nadir Yehya University of Pennsylvania, [email protected] Adi Yerrapureddy University of Pennsylvania John Tobias University of Pennsylvania, [email protected] Susan S. Margulies University of Pennsylvania, [email protected] Follow this and additional works at: https://repository.upenn.edu/be_papers Part of the Biomedical Engineering and Bioengineering Commons Recommended Citation Yehya, N., Yerrapureddy, A., Tobias, J., & Margulies, S. S. (2012). MicroRNA Modulate Alveolar Epithelial Response to Cyclic Stretch. BMC Genomics, 13 (154), http://dx.doi.org/10.1186/1471-2164-13-154 This paper is posted at ScholarlyCommons. https://repository.upenn.edu/be_papers/205 For more information, please contact [email protected]. MicroRNA Modulate Alveolar Epithelial Response to Cyclic Stretch Abstract Background MicroRNAs (miRNAs) are post-transcriptional regulators of gene expression implicated in multiple cellular processes. Cyclic stretch of alveoli is characteristic of mechanical ventilation, and is postulated to be partly responsible for the lung injury and inflammation in entilatv or-induced lung injury. We propose that miRNAs may regulate some of the stretch response, and therefore hypothesized that miRNAs would be differentially expressed between cyclically stretched and unstretched rat alveolar epithelial cells (RAECs). Results RAECs were isolated and cultured to express type I epithelial characteristics. They were then equibiaxially stretched to 25% change in surface area at 15 cycles/minute for 1 hour or 6 hours, or served as unstretched controls, and miRNAs were extracted. Expression profiling of the miRNAs with at least 1.5-fold change over controls revealed 42 miRNAs were regulated (34 up and 8 down) with stretch.
    [Show full text]
  • Upregulation of Pole2 Promotes Clear Cell Renal Cell Carcinoma Progression Via AKT/Mtor Pathway and Predicts a Poor Prognosis
    Upregulation of Pole2 Promotes Clear Cell Renal Cell Carcinoma Progression via AKT/mTOR Pathway and Predicts a Poor Prognosis Yajuan Su Tumor Hospital of Harbin Medical University Changfu Li Tumor Hospital of Harbin Medical University Kun Liu Tumor Hospital of Harbin Medical University Liangjun Wei Tumor Hospital of Harbin Medical University Dechao Li Tumor Hospital of Harbin Medical University Wentao Wang Tumor Hospital of Harbin Medical University Yongpeng Xu Tumor Hospital of Harbin Medical University Hongxin Pan Tumor Hospital of Harbin Medical University Lichen Teng ( [email protected] ) Tumor Hospital of Harbin Medical University https://orcid.org/0000-0002-5840-0723 Primary research Keywords: renal cell carcinoma, pole2, survival, cell cycle, apoptosis Posted Date: June 22nd, 2020 DOI: https://doi.org/10.21203/rs.3.rs-35733/v1 License: This work is licensed under a Creative Commons Attribution 4.0 International License. Read Full License Page 1/18 Abstract Background: Pole2 gene is a subunit of DNA polymerases localized in the nucleus, which commonly present in DNA repair. The effect of pole2 in renal cell carcinoma (RCC) still remain unclear. Here we investigate its clinical signicance, function in RCC cells and possible mechanism of effect. Methods: Using TCGA database, we identied that up-regulation of pole2 is associated with poor prognosis in ccRCC. We analyzed association between pole2 expression and T stage or Fuhrman grade. Thus, we investigate the effects of pole2 down-regulation on proliferation, cell cycles, apoptosis and possible mechanism in cells using lentivirus vector with shPole2. Results: Our study showed overexpression of pole2 in ccRCC samples, compared with normal kidney tissues, moreover, high expression of it related to high Fuhrman grade, also may predict poor prognosis in patients with ccRCC (p < 0.05).
    [Show full text]
  • CHK1 Inhibition Is Synthetically Lethal with Loss of B-Family DNA Polymerase Function in Human Lung and Colorectal Cancer Cells
    Author Manuscript Published OnlineFirst on March 11, 2020; DOI: 10.1158/0008-5472.CAN-19-1372 Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited. 1 Title: CHK1 inhibition is synthetically lethal with loss of B-family DNA polymerase function in human lung and colorectal cancer cells. Author List: Rebecca F. Rogers1, Mike I. Walton1, Daniel L. Cherry2, Ian Collins1, Paul A. Clarke1, Michelle D. Garrett2* and Paul Workman1* Affiliations: 1Cancer Research UK Cancer Therapeutics Unit, The Institute of Cancer Research, London, SM2 5NG, UK 2 School of Biosciences, Stacey Building, University of Kent, Canterbury, Kent, CT2 7NJ, UK Running title: Synthetic lethality of CHK1 and DNA polymerase inhibition *Corresponding authors: Michelle D Garrett and Paul Workman Corresponding Author Information Michelle D Garrett School of Biosciences, Stacey Building, University of Kent, Canterbury, Kent, CT2 7NJ, UK. Email: [email protected] Phone: +44 (0)1227 816140 Paul Workman Cancer Research UK Cancer Therapeutics Unit, The Institute of Cancer Research, London, SM2 5NG, UK. Email: [email protected] Phone: +44 (0)20 7153 5209 Conflict of interest statement: IC, MDG, MIW, RFR, PC and PW are current or past employees of The Institute of Cancer Research, which has a commercial interest in the discovery and development of CHK1 inhibitors, including SRA737, and operates a rewards-to- inventors scheme. IC, MDG and MIW have been involved in a commercial collaboration on CHK1 inhibitors with Sareum Ltd and intellectual property arising from the program, including SRA737, was licensed to Sierra Oncology. IC is a consultant for Sierra Oncology, Adorx Ltd, Epidarex LLP and Enterprise Therapeutics Ltd and holds equity in Monte Rosa Therapeutics AG.
    [Show full text]
  • In Utero Exposure to Chlordecone Affects Histone Modifications And
    In utero exposure to chlordecone affects histone modifications and activates LINE-1 in cord blood Louis Legoff, Shereen Cynthia D’cruz, Katia Bouchekhchoukha, Christine Monfort, Christian Jaulin, Luc Multigner, Fatima Smagulova To cite this version: Louis Legoff, Shereen Cynthia D’cruz, Katia Bouchekhchoukha, Christine Monfort, Christian Jaulin, et al.. In utero exposure to chlordecone affects histone modifications and activates LINE- 1 in cord blood. Life Science Alliance, Life Science Alliance LLC, 2021, 4 (6), pp.e202000944. 10.26508/lsa.202000944. hal-03217292 HAL Id: hal-03217292 https://hal.archives-ouvertes.fr/hal-03217292 Submitted on 4 May 2021 HAL is a multi-disciplinary open access L’archive ouverte pluridisciplinaire HAL, est archive for the deposit and dissemination of sci- destinée au dépôt et à la diffusion de documents entific research documents, whether they are pub- scientifiques de niveau recherche, publiés ou non, lished or not. The documents may come from émanant des établissements d’enseignement et de teaching and research institutions in France or recherche français ou étrangers, des laboratoires abroad, or from public or private research centers. publics ou privés. Distributed under a Creative Commons Attribution| 4.0 International License Published Online: 9 April, 2021 | Supp Info: http://doi.org/10.26508/lsa.202000944 Downloaded from life-science-alliance.org on 4 May, 2021 Research Article In utero exposure to chlordecone affects histone modifications and activates LINE-1 in cord blood Louis Legoff1, Shereen Cynthia D’Cruz1, Katia Bouchekhchoukha1, Christine Monfort1, Christian Jaulin2, Luc Multigner1 , Fatima Smagulova1 Environmental factors can induce detrimental consequences into Epigenetic reprogramming during development is associated with a adulthood life.
    [Show full text]