Associations Between Genetic Polymorphisms in Dna Bypass Polymerases and Base Excision Repair Genes with the Risk of Breast Cancer

Total Page:16

File Type:pdf, Size:1020Kb

Associations Between Genetic Polymorphisms in Dna Bypass Polymerases and Base Excision Repair Genes with the Risk of Breast Cancer ASSOCIATIONS BETWEEN GENETIC POLYMORPHISMS IN DNA BYPASS POLYMERASES AND BASE EXCISION REPAIR GENES WITH THE RISK OF BREAST CANCER Leila Family A dissertation submitted to the faculty of the University of North Carolina at Chapel Hill in partial fulfillment of the requirements for the degree of Doctor of Philosophy in the Department of Epidemiology. Chapel Hill 2014 Approved by: Andrew F. Olshan Jeanette T. Bensen Melissa A. Troester Michael C. Wu Carey K. Anders ©2014 Leila Family ALL RIGHTS RESERVED ii ABSTRACT LEILA FAMILY: Associations between genetic polymorphisms in DNA bypass polymerases and base excision repair genes with the risk of breast cancer (Under the direction of Andrew F. Olshan) Mutations in BRCA1, a DNA repair gene, have been associated with a lifetime increased risk of breast cancer (1). Therefore, researchers hypothesized there may be other DNA repair genes associated with breast cancer risk. However thus far, studies of common low-penetrant DNA repair SNPs have not yielded consistent results. In this proposed study, we hypothesized one or more of the following mechanisms may explain the lack of main SNP effects: combined SNP effects, modification by race or breast cancer subtype, and functional redundancy. To evaluate these hypotheses, we used genotype data from the Carolina Breast Cancer Study (1,972 cases and 1,776 controls) to investigate race-specific, subtype-specific, and combined SNP associations using unconditional logistic regression in two DNA damage pathways, base excision repair (BER) and translesion synthesis (TLS). For BER, we evaluated the association between 31 single-nucleotide polymorphisms (SNPs) in 15 genes and breast cancer risk. SKAT, a pathway-based analytic method, was used to evaluate the combined SNP effects within the BER pathway. Among Whites, our results showed a significant positive association for NEIL2 rs1534862 and a significant inverse association for PCNA rs17352. Among African Americans, we found a suggestive positive association for UNG rs3219275 and an inverse association for NEIL2 rs8191613. Tumor subtype analysis showed that NEIL2 rs1534862 was associated with luminal and HER2+/ER- subtypes. SKAT analysis showed no significant combined effects between SNPs. For DNA bypass polymerases, we evaluated the association between 22 single- iii nucleotide polymorphisms (SNPs) in 7 bypass polymerase genes and breast cancer risk. We found similar increased odds ratios for breast cancer with three POLQ SNPs (rs487848, rs532411, rs3218634), which were also in high LD in both races. Furthermore, analysis by specific tumor subtypes showed all three SNPs were associated with increased risk of luminal breast cancer. These significant findings need to be replicated independently in other studies. Overall, our results did not indicate associations with breast cancer, which may concur with the theory that our cells possess an intricate system of functionally redundant DNA repair mechanisms in order to avoid the catastrophic effects of genomic instability. iv To Dr. Robert C. Millikan Who always believed in me “The thing always happens that you really believe in, and the belief in a thing makes it happen.” -Frank Lloyd Wright v ACKNOWLEDGEMENTS First, I would like to thank God for giving the strength and endurance to continue and finish my dissertation work. Also, I would like to thank my dissertation committee members, Dr. Andrew Olshan, Dr. Jeannette Bensen, Dr. Melissa Troester, Dr. Michael Wu, and Dr. Carey Anders, for providing me with their expertise and valuable feedback. I would like to extend a special thanks to my chair, Dr. Andrew Olshan, for his continuous guidance and support and for navigating me through the dissertation process and dealing with unforeseen circumstances. I would also like to thank everyone who has contributed their time and energy to the Carolina Breast Cancer Study. Without their hard work and dedication, this work would not be possible. I would especially like to thank Mary Beth Bell, CBCS Project Manager, for her strong commitment to the study. Of course, I would also like to acknowledge the selfless contributions of all the women that participated in the Carolina Breast Cancer Study. I would also like to thank my vast circle of support, including the helpful staff in the Epidemiology Department. I especially want to thank Nancy Colvin for her unwavering support and dedication to my success. I would also like to thank my awesome friends and family. In particular, I would like to give special thanks to Katie O’Brien and Lauren McCullough. These ladies went out of their way to make sure I was successful, including volunteering countless hours of their time to provide advice and feedback. I would also like to take a moment to acknowledge and honor the work of Dr. Robert Millikan and Dr. Keith Amos, whose impact in the field of breast cancer will be felt for years to come. vi Last, but certainly not least, I would like to thank my amazing parents, Gity and Siamak Family, for their unwavering support and unconditional love throughout my whole life, and the many selfless sacrifices they have made for me. I am truly blessed and honored to know such wonderful and genuinely humble people. I would also like to acknowledge the sources of financial support that enabled me to complete this research: the University Cancer Research Fund of North Carolina, the National Cancer Institute Specialized Program of Research Excellence (SPORE) in Breast Cancer (NIH/ NCI P50-CA58223) and NRSA Pre Doctoral Training Grant (2007-2009), University of North Carolina at Chapel Hill Grant # 2-T32-CA09330. vii TABLE OF CONTENTS LIST OF FIGURES .................................................................................................................. xvii LIST OF ABBREVIATIONS ................................................................................................. xviii CHAPTER 1. REVIEW OF THE LITERATURE .................................................................... 1 1.1 Introduction ........................................................................................................................... 1 1.2 Definition of breast cancer .................................................................................................... 1 1.3 Epidemiology of breast cancer.............................................................................................. 2 1.3.1 Breast cancer incidence .................................................................................................. 2 1.3.2 Breast cancer mortality .................................................................................................. 3 1.3.3 Non-genetic risk factors of breast cancer ....................................................................... 3 1.3.3.1 Non-genetic risk factors of breast cancer by race ................................................... 4 1.3.4 Genetic risk factors of breast cancer .............................................................................. 5 1.3.4.1 Genetic risk factors of breast cancer by race .......................................................... 7 1.4 Heterogeneity of breast cancer.............................................................................................. 7 1.4.1 Non-genetic risk factors of breast cancer by subtype .................................................... 9 1.4.2 Genetic risk factors of breast cancer by subtype.......................................................... 10 1.4.3 Summary of breast cancer risk factors ......................................................................... 12 1.5 Variation in DNA repair capacity ....................................................................................... 12 viii 1.5.1 Low-penetrant common DNA repair variation in breast cancer .................................. 13 1.6 DNA damage responses ...................................................................................................... 13 1.6.1 Overview of DNA repair.............................................................................................. 14 1.6.2 Overview of base excision repair (BER) ..................................................................... 14 1.6.2.1 Base excision repair and breast cancer ................................................................. 16 1.6.2.1.1 UNG ............................................................................................................................. 16 1.6.2.1.2 SMUG1 ....................................................................................................................... 16 1.6.2.1.3 MBD ............................................................................................................................. 17 1.6.2.1.4 MPG ............................................................................................................................. 17 1.6.2.1.5 MYH/MUYTH .............................................................................................................. 17 1.6.2.1.6 TDG .............................................................................................................................. 18 1.6.2.1.7 OGG1 ........................................................................................................................... 18 1.6.2.1.8 NEIL1 ........................................................................................................................... 19 1.6.2.1.9 NEIL2 ..........................................................................................................................
Recommended publications
  • Zhou Et Al POLQ Inhibitor.Docx
    bioRxiv preprint doi: https://doi.org/10.1101/2020.05.23.111658; this version posted May 26, 2020. The copyright holder for this preprint (which was not certified by peer review) is the author/funder, who has granted bioRxiv a license to display the preprint in perpetuity. It is made available under aCC-BY-NC-ND 4.0 International license. Polymerase Theta Inhibition Kills Homologous Recombination Deficient Tumors Jia Zhou1, Camille Gelot2, Constantia Pantelidou3, Adam Li1, Hatice Yücel2, Rachel E. Davis4, Anniina Farkkila1, Bose Kochupurakkal1, Aleem Syed5, Geoffrey I. Shapiro3,6, John A. Tainer5, Brian S. J. Blagg4, Raphael Ceccaldi2,7* and Alan D. D’Andrea1,6,7* 1Department of Radiation Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA 02215, USA. 2Inserm U830, PSL Research University, Institut Curie, 75005, Paris, France. 3Department of Medical Oncology, Dana-Farber Cancer Institute and Department of Medicine, Harvard Medical School, Boston, Massachusetts, USA. 4Department of Chemistry and Biochemistry, University of Notre Dame, Notre Dame, IN 46556, USA. 5Departments of Cancer Biology and of Molecular and Cellular Oncology, University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA. 6Center for DNA Damage and Repair, Dana-Farber Cancer Institute, Boston, MA, USA. 7Co-senior authors. * Co-corresponding authors. * Corresponding authors: Alan D. D’Andrea, M.D. Director, Susan F. Smith Center for Women’s Cancers (SFSCWC) Director, Center for DNA Damage and Repair Dana-Farber Cancer Institute The Fuller-American Cancer Society Professor Harvard Medical School Phone: 617-632-2080 Email: [email protected] Raphael Ceccaldi Institut Curie, 75005, Paris, France Phone: +33 (0)1 56 24 69 49 Email: [email protected] Key Words: Novobiocin, Polymerase theta (POLθ), Homologous Recombination, PARP inhibitor resistance.
    [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]
  • Investigation of the Underlying Hub Genes and Molexular Pathogensis in Gastric Cancer by Integrated Bioinformatic Analyses
    bioRxiv preprint doi: https://doi.org/10.1101/2020.12.20.423656; this version posted December 22, 2020. The copyright holder for this preprint (which was not certified by peer review) is the author/funder. All rights reserved. No reuse allowed without permission. Investigation of the underlying hub genes and molexular pathogensis in gastric cancer by integrated bioinformatic analyses Basavaraj Vastrad1, Chanabasayya Vastrad*2 1. Department of Biochemistry, Basaveshwar College of Pharmacy, Gadag, Karnataka 582103, India. 2. Biostatistics and Bioinformatics, Chanabasava Nilaya, Bharthinagar, Dharwad 580001, Karanataka, India. * Chanabasayya Vastrad [email protected] Ph: +919480073398 Chanabasava Nilaya, Bharthinagar, Dharwad 580001 , Karanataka, India bioRxiv preprint doi: https://doi.org/10.1101/2020.12.20.423656; this version posted December 22, 2020. The copyright holder for this preprint (which was not certified by peer review) is the author/funder. All rights reserved. No reuse allowed without permission. Abstract The high mortality rate of gastric cancer (GC) is in part due to the absence of initial disclosure of its biomarkers. The recognition of important genes associated in GC is therefore recommended to advance clinical prognosis, diagnosis and and treatment outcomes. The current investigation used the microarray dataset GSE113255 RNA seq data from the Gene Expression Omnibus database to diagnose differentially expressed genes (DEGs). Pathway and gene ontology enrichment analyses were performed, and a proteinprotein interaction network, modules, target genes - miRNA regulatory network and target genes - TF regulatory network were constructed and analyzed. Finally, validation of hub genes was performed. The 1008 DEGs identified consisted of 505 up regulated genes and 503 down regulated genes.
    [Show full text]
  • Supplementary Table S4. FGA Co-Expressed Gene List in LUAD
    Supplementary Table S4. FGA co-expressed gene list in LUAD tumors Symbol R Locus Description FGG 0.919 4q28 fibrinogen gamma chain FGL1 0.635 8p22 fibrinogen-like 1 SLC7A2 0.536 8p22 solute carrier family 7 (cationic amino acid transporter, y+ system), member 2 DUSP4 0.521 8p12-p11 dual specificity phosphatase 4 HAL 0.51 12q22-q24.1histidine ammonia-lyase PDE4D 0.499 5q12 phosphodiesterase 4D, cAMP-specific FURIN 0.497 15q26.1 furin (paired basic amino acid cleaving enzyme) CPS1 0.49 2q35 carbamoyl-phosphate synthase 1, mitochondrial TESC 0.478 12q24.22 tescalcin INHA 0.465 2q35 inhibin, alpha S100P 0.461 4p16 S100 calcium binding protein P VPS37A 0.447 8p22 vacuolar protein sorting 37 homolog A (S. cerevisiae) SLC16A14 0.447 2q36.3 solute carrier family 16, member 14 PPARGC1A 0.443 4p15.1 peroxisome proliferator-activated receptor gamma, coactivator 1 alpha SIK1 0.435 21q22.3 salt-inducible kinase 1 IRS2 0.434 13q34 insulin receptor substrate 2 RND1 0.433 12q12 Rho family GTPase 1 HGD 0.433 3q13.33 homogentisate 1,2-dioxygenase PTP4A1 0.432 6q12 protein tyrosine phosphatase type IVA, member 1 C8orf4 0.428 8p11.2 chromosome 8 open reading frame 4 DDC 0.427 7p12.2 dopa decarboxylase (aromatic L-amino acid decarboxylase) TACC2 0.427 10q26 transforming, acidic coiled-coil containing protein 2 MUC13 0.422 3q21.2 mucin 13, cell surface associated C5 0.412 9q33-q34 complement component 5 NR4A2 0.412 2q22-q23 nuclear receptor subfamily 4, group A, member 2 EYS 0.411 6q12 eyes shut homolog (Drosophila) GPX2 0.406 14q24.1 glutathione peroxidase
    [Show full text]
  • Error-Prone DNA Repair As Cancer's Achilles' Heel
    cancers Review Alternative Non-Homologous End-Joining: Error-Prone DNA Repair as Cancer’s Achilles’ Heel Daniele Caracciolo, Caterina Riillo , Maria Teresa Di Martino , Pierosandro Tagliaferri and Pierfrancesco Tassone * Department of Experimental and Clinical Medicine, Magna Græcia University, Campus Salvatore Venuta, 88100 Catanzaro, Italy; [email protected] (D.C.); [email protected] (C.R.); [email protected] (M.T.D.M.); [email protected] (P.T.) * Correspondence: [email protected] Simple Summary: Cancer onset and progression lead to a high rate of DNA damage, due to replicative and metabolic stress. To survive in this dangerous condition, cancer cells switch the DNA repair machinery from faithful systems to error-prone pathways, strongly increasing the mutational rate that, in turn, supports the disease progression and drug resistance. Although DNA repair de-regulation boosts genomic instability, it represents, at the same time, a critical cancer vulnerability that can be exploited for synthetic lethality-based therapeutic intervention. We here discuss the role of the error-prone DNA repair, named Alternative Non-Homologous End Joining (Alt-NHEJ), as inducer of genomic instability and as a potential therapeutic target. We portray different strategies to drug Alt-NHEJ and discuss future challenges for selecting patients who could benefit from Alt-NHEJ inhibition, with the aim of precision oncology. Abstract: Error-prone DNA repair pathways promote genomic instability which leads to the onset of cancer hallmarks by progressive genetic aberrations in tumor cells. The molecular mechanisms which Citation: Caracciolo, D.; Riillo, C.; Di foster this process remain mostly undefined, and breakthrough advancements are eagerly awaited. Martino, M.T.; Tagliaferri, P.; Tassone, In this context, the alternative non-homologous end joining (Alt-NHEJ) pathway is considered P.
    [Show full text]
  • Lorena Novoa-Aponte
    A Strain: fl/fl ∆hep Strain: fl/fl ∆hep AnalysisAAV: ofLuc the ironLuc chaperonePCBP1-WT PCBP1PCBP1-∆Fe interactome:PCBP1-∆RNA PCBP1 variant intersection of DNA repair and iron traffickingAAV: Luc Luc WT ∆Fe ∆RNA PCBP1 −40 Lorena Novoa-Aponte a*, Sarju J. Patel a, Olga Protchenko a, James Wohlschlegel b, Caroline C. Philpott a −40 PCBP1, IHC PCBP1, GAPDH - a Genetics and Metabolism Section,a NIDDK, NIH, Bethesda, MD, USA. b Department of Biological Chemistry, UCLA, Los Angeles, CA, USA C.C. Philpott, et al. *[email protected] 80 B P = 0.0262 60 40 1. Iron is essential but toxic 2. Increased DNA damage in cells lacking PCBP1 ns H&E ? + 20 C.C. Philpott, et al. Iron is used as an essential cofactor by several enzymes involved in DNA ⦿ Increased TUNEL in cells and tissue livers from mice lacking PCBP1. ? replication and repair. However, unchaperoned iron promotes redox ⦿ PCBP1 binds both, iron and single-stranded nucleicTG, nmol/mg protein 0 acids. stress that may affect DNA stability. Strain: Δhep Δhep Δhep ? ⦿ The iron binding activity of PCBP1 controls suppressionAAV: of DNA damage C WT ∆Fe ∆RNA PCBP1 variant ? Iron storage HEK293 cells Mouse Liver ? A 8 siNT+P1var 100 ? A 8 var siPCBP1 2 ? Mammals use the iron siNT+P1 ns var Fe-S cluster assembly 6 siPCBP1+P1siPCBP1 80 ADI1 TUNEL chaperone PCBP1 to var ? 6 siPCBP1+P1 metalate several iron- 60 = 0.0468 = 0.0354 = 0.0465 cell / mm population P P P population + dependent enzymes + 4 + 4 ? 40 Degradation of HIF1α Fig. 3. Iron chaperone-mediated handling of cytosolic labile iron pool.
    [Show full text]
  • Human Induced Pluripotent Stem Cell–Derived Podocytes Mature Into Vascularized Glomeruli Upon Experimental Transplantation
    BASIC RESEARCH www.jasn.org Human Induced Pluripotent Stem Cell–Derived Podocytes Mature into Vascularized Glomeruli upon Experimental Transplantation † Sazia Sharmin,* Atsuhiro Taguchi,* Yusuke Kaku,* Yasuhiro Yoshimura,* Tomoko Ohmori,* ‡ † ‡ Tetsushi Sakuma, Masashi Mukoyama, Takashi Yamamoto, Hidetake Kurihara,§ and | Ryuichi Nishinakamura* *Department of Kidney Development, Institute of Molecular Embryology and Genetics, and †Department of Nephrology, Faculty of Life Sciences, Kumamoto University, Kumamoto, Japan; ‡Department of Mathematical and Life Sciences, Graduate School of Science, Hiroshima University, Hiroshima, Japan; §Division of Anatomy, Juntendo University School of Medicine, Tokyo, Japan; and |Japan Science and Technology Agency, CREST, Kumamoto, Japan ABSTRACT Glomerular podocytes express proteins, such as nephrin, that constitute the slit diaphragm, thereby contributing to the filtration process in the kidney. Glomerular development has been analyzed mainly in mice, whereas analysis of human kidney development has been minimal because of limited access to embryonic kidneys. We previously reported the induction of three-dimensional primordial glomeruli from human induced pluripotent stem (iPS) cells. Here, using transcription activator–like effector nuclease-mediated homologous recombination, we generated human iPS cell lines that express green fluorescent protein (GFP) in the NPHS1 locus, which encodes nephrin, and we show that GFP expression facilitated accurate visualization of nephrin-positive podocyte formation in
    [Show full text]
  • Biomarkers in Triple-Negative Breast Cancer: State-Of-The-Art and Future Perspectives
    International Journal of Molecular Sciences Review Biomarkers in Triple-Negative Breast Cancer: State-of-the-Art and Future Perspectives Stefania Cocco , Michela Piezzo , Alessandra Calabrese, Daniela Cianniello, Roberta Caputo, Vincenzo Di Lauro, Giuseppina Fusco, Germira di Gioia, Marina Licenziato and Michelino de Laurentiis * Istituto Nazionale Tumori IRCCS “Fondazione G. Pascale”, Via Mariano Semmola, 53, 80131 Napoli NA, Italy; [email protected] (S.C.); [email protected] (M.P.); [email protected] (A.C.); [email protected] (D.C.); [email protected] (R.C.); [email protected] (V.D.L.); [email protected] (G.F.); [email protected] (G.d.G.); [email protected] (M.L.) * Correspondence: [email protected]; Tel.: +39-081-5903-535 Received: 31 May 2020; Accepted: 25 June 2020; Published: 27 June 2020 Abstract: Triple-negative breast cancer (TNBC) is a heterogeneous group of tumors characterized by aggressive behavior, high risk of distant recurrence, and poor survival. Chemotherapy is still the main therapeutic approach for this subgroup of patients, therefore, progress in the treatment of TNBC remains an important challenge. Data derived from molecular technologies have identified TNBCs with different gene expression and mutation profiles that may help developing targeted therapies. So far, however, only a few of these have shown to improve the prognosis and outcomes of TNBC patients. Robust predictive biomarkers to accelerate clinical progress are needed. Herein, we review prognostic and predictive biomarkers in TNBC, discuss the current evidence supporting their use, and look at the future of this research field. Keywords: TNBC; BRCA1/2; HRR; PDL1; TILs; PI3KCA; PTEN; CTCs; CSC 1.
    [Show full text]
  • Candidate Synthetic Lethality Partners to PARP Inhibitors in the Treatment of Ovarian Clear Cell Cancer (Review)
    BIOMEDICAL REPORTS 7: 391-399, 2017 Candidate synthetic lethality partners to PARP inhibitors in the treatment of ovarian clear cell cancer (Review) NAOKI KAWAHARA, KENJI OGAWA, MIKA NAGAYASU, MAI KIMURA, YOSHIKAZU SASAKI and HIROSHI KOBAYASHI Department of Obstetrics and Gynecology, Nara Medical University, Nara 634-8522, Japan Received August 18, 2017; Accepted September 14, 2017 DOI: 10.3892/br.2017.990 Abstract. Inhibitors of poly(ADP-ribose) polymerase Contents (PARP) are new types of personalized treatment of relapsed platinum-sensitive ovarian cancer harboring BRCA1/2 1. Introduction mutations. Ovarian clear cell cancer (CCC), a subset of 2. Systematic review of the literature using electronic ovarian cancer, often appears as low-stage disease with search in the PubMed/Medline databases a higher incidence among Japanese. Advanced CCC is 3. Future opportunities in the use of PARP inhibition in CCC highly aggressive with poor patient outcome. The aim of 4. Candidate mutated genes for enhancing the therapeutic the present study was to determine the potential synthetic ratio achieved by PARP inhibitors in CCC (Table IA). lethality gene pairs for PARP inhibitions in patients with 5. Upregulated genes enhancing synthetic lethality of CCC through virtual and biological screenings as well as PARP inhibitors in CCC (Table IB) clinical studies. We conducted a literature review for puta- 6. Synthetic lethal gene partners based on tive PARP sensitivity genes that are associated with the chemo resi stance-related genes in CCC (Table IC) CCC pathophysiology. Previous studies identified a variety 7. Discussion of putative target genes from several pathways associated with DNA damage repair, chromatin remodeling complex, PI3K-AKT-mTOR signaling, Notch signaling, cell cycle 1.
    [Show full text]
  • Polymerase Is a Robust Terminal Transferase That Oscillates Between
    RESEARCH ARTICLE Polymerase is a robust terminal transferase that oscillates between three different mechanisms during end-joining Tatiana Kent1,2, Pedro A Mateos-Gomez3,4, Agnel Sfeir3,4, Richard T Pomerantz1,2* 1Fels Institute for Cancer Research, Temple University Lewis Katz School of Medicine, Philadelphia, United States; 2Department of Medical Genetics and Molecular Biochemistry, Temple University Lewis Katz School of Medicine, Philadelphia, United States; 3Skirball Institute of Biomolecular Medicine, New York University School of Medicine, New York, United States; 4Department of Cell Biology, New York University School of Medicine, New York, United States Abstract DNA polymerase q (Polq) promotes insertion mutations during alternative end-joining (alt-EJ) by an unknown mechanism. Here, we discover that mammalian Polq transfers nucleotides to the 3’ terminus of DNA during alt-EJ in vitro and in vivo by oscillating between three different modes of terminal transferase activity: non-templated extension, templated extension in cis, and templated extension in trans. This switching mechanism requires manganese as a co-factor for Polq template-independent activity and allows for random combinations of templated and non- templated nucleotide insertions. We further find that Polq terminal transferase activity is most efficient on DNA containing 3’ overhangs, is facilitated by an insertion loop and conserved residues that hold the 3’ primer terminus, and is surprisingly more proficient than terminal deoxynucleotidyl transferase. In summary, this report identifies an unprecedented switching mechanism used by Polq to generate genetic diversity during alt-EJ and characterizes Polq as among the most proficient terminal transferases known. DOI: 10.7554/eLife.13740.001 *For correspondence: richard.
    [Show full text]
  • The Genetic Program of Pancreatic Beta-Cell Replication in Vivo
    Page 1 of 65 Diabetes The genetic program of pancreatic beta-cell replication in vivo Agnes Klochendler1, Inbal Caspi2, Noa Corem1, Maya Moran3, Oriel Friedlich1, Sharona Elgavish4, Yuval Nevo4, Aharon Helman1, Benjamin Glaser5, Amir Eden3, Shalev Itzkovitz2, Yuval Dor1,* 1Department of Developmental Biology and Cancer Research, The Institute for Medical Research Israel-Canada, The Hebrew University-Hadassah Medical School, Jerusalem 91120, Israel 2Department of Molecular Cell Biology, Weizmann Institute of Science, Rehovot, Israel. 3Department of Cell and Developmental Biology, The Silberman Institute of Life Sciences, The Hebrew University of Jerusalem, Jerusalem 91904, Israel 4Info-CORE, Bioinformatics Unit of the I-CORE Computation Center, The Hebrew University and Hadassah, The Institute for Medical Research Israel- Canada, The Hebrew University-Hadassah Medical School, Jerusalem 91120, Israel 5Endocrinology and Metabolism Service, Department of Internal Medicine, Hadassah-Hebrew University Medical Center, Jerusalem 91120, Israel *Correspondence: [email protected] Running title: The genetic program of pancreatic β-cell replication 1 Diabetes Publish Ahead of Print, published online March 18, 2016 Diabetes Page 2 of 65 Abstract The molecular program underlying infrequent replication of pancreatic beta- cells remains largely inaccessible. Using transgenic mice expressing GFP in cycling cells we sorted live, replicating beta-cells and determined their transcriptome. Replicating beta-cells upregulate hundreds of proliferation- related genes, along with many novel putative cell cycle components. Strikingly, genes involved in beta-cell functions, namely glucose sensing and insulin secretion were repressed. Further studies using single molecule RNA in situ hybridization revealed that in fact, replicating beta-cells double the amount of RNA for most genes, but this upregulation excludes genes involved in beta-cell function.
    [Show full text]
  • Genomic Interrogation of Melanoma for Identification of Driver Oncogenic Factors
    UC Merced UC Merced Electronic Theses and Dissertations Title Genomic Interrogation of Melanoma for Identification of Driver Oncogenic Factors Permalink https://escholarship.org/uc/item/1fb0q0s2 Author Gupta, Rohit Publication Date 2019 License https://creativecommons.org/licenses/by/4.0/ 4.0 Peer reviewed|Thesis/dissertation eScholarship.org Powered by the California Digital Library University of California UNIVERSITY OF CALIFORNIA, MERCED Genomic Interrogation of Melanoma for Identification of Driver Oncogenic Factors by Rohit Gupta A dissertation submitted in partial fulfillment for the degree of Doctor of Philosophy in the Program for Quantitative & Systems Biology School of Natural Sciences August 2019 Committee in charge: Professor Miriam Barlow, Advisor Professor Michael Colvin, Chair Professor Suzanne Sindi Professor Mark Sistrom Copyright Rohit Gupta, 2019 All rights reserved The Dissertation of Rohit Gupta is approved, and it is acceptable in quality and form for publication on microfilm and electronically: Professor Miriam Barlow, Advisor Professor Michael Colvin, Chair Professor Suzanne Sindi Professor Mark Sistrom University of California, Merced 2019 \To reach a port we must set sail { Sail, not tie at anchor Sail, not drift." Franklin D. Roosevelt UNIVERSITY OF CALIFORNIA, MERCED Abstract Program for Quantitative & Systems Biology School of Natural Sciences Doctor of Philosophy by Rohit Gupta Underpinning genomic principles are defining feature in every physiologic and pathologic process. Through advances in metabolomics, systems biologists can now track the dynamic interactions of the metabolome with the epigenome, genome, transcriptome and proteome. Understanding of cross talk between genomic, epige- nomic and structural changes at biophysical scale on cellular metabolism is still in its infancy. Using Next-Gen Sequencing data in tandem with biophysical approaches, metabolism was found to play an important role in cellular prolif- eration, differentiation, metastasis.
    [Show full text]