Recurrent Integration of Human Papillomavirus Genomes at Transcriptional

Total Page:16

File Type:pdf, Size:1020Kb

Recurrent Integration of Human Papillomavirus Genomes at Transcriptional bioRxiv preprint doi: https://doi.org/10.1101/2021.08.30.457540; this version posted August 31, 2021. The copyright holder for this preprint (which was not certified by peer review) is the author/funder. This article is a US Government work. It is not subject to copyright under 17 USC 105 and is also made available for use under a CC0 license. 1 Recurrent Integration of Human Papillomavirus Genomes at Transcriptional 2 Regulatory Hubs 3 Alix Warburton1, Tovah E. Markowitz2-3, Joshua P. Katz4, James M. Pipas4 and Alison 4 A. McBride1 * 5 6 1Laboratory of Viral Diseases, National Institute of Allergy and Infectious Diseases, 33 North Drive, 7 MSC3209, National Institutes of Health, Bethesda, Maryland 20892, USA 8 2NIAID Collaborative Bioinformatics Resource (NCBR), National Institute of Allergy and Infectious 9 Diseases, National Institutes of Health, Bethesda, MD, USA 10 3Advanced Biomedical Computational Science, Frederick National Laboratory for Cancer Research, 11 Frederick, MD, USA 12 4Department of Biological Sciences, University of Pittsburgh, Pittsburgh, Pennsylvania, USA 13 14 *Corresponding author 15 E-mail: [email protected] 16 Short Title: Integration of HPV genomes in human cancers 17 18 19 20 21 22 1 bioRxiv preprint doi: https://doi.org/10.1101/2021.08.30.457540; this version posted August 31, 2021. The copyright holder for this preprint (which was not certified by peer review) is the author/funder. This article is a US Government work. It is not subject to copyright under 17 USC 105 and is also made available for use under a CC0 license. 23 ABSTRACT 24 Oncogenic human papillomavirus (HPV) genomes are often integrated into host chromosomes in HPV- 25 associated cancers. HPV genomes are integrated either as a single copy, or as tandem repeats of viral 26 DNA interspersed with, or without, host DNA. Integration occurs frequently in common fragile sites 27 susceptible to tandem repeat formation, and the flanking or interspersed host DNA often contains 28 transcriptional enhancer elements. When co-amplified with the viral genome, these enhancers can form 29 super-enhancer-like elements that drive high viral oncogene expression. Here, we compiled highly 30 curated datasets of HPV integration sites in cervical (CESC) and head and neck squamous cell carcinoma 31 (HNSCC) cancers and assessed the number of breakpoints, viral transcriptional activity, and host genome 32 copy number at each insertion site. Tumors frequently contained multiple distinct HPV integration sites, 33 but often only one “driver” site that expressed viral RNA. Since common fragile sites and active 34 enhancer elements are cell-type specific, we mapped these regions in cervical cell lines using FANCD2 35 and Brd4/H3K27ac ChIP-seq, respectively. Large enhancer clusters, or super-enhancers, were also 36 defined using the Brd4/H3K27ac ChIP-seq dataset. HPV integration breakpoints were enriched at both 37 FANCD2-associated fragile sites, and enhancer-rich regions, and frequently showed adjacent focal DNA 38 amplification in CESC samples. We identified recurrent integration “hotspots” that were enriched for 39 super-enhancers, some of which function as regulatory hubs for cell-identity genes. We propose that 40 during persistent infection, extrachromosomal HPV minichromosomes associate with these 41 transcriptional epicenters, and accidental integration could promote viral oncogene expression and 42 carcinogenesis. 2 bioRxiv preprint doi: https://doi.org/10.1101/2021.08.30.457540; this version posted August 31, 2021. The copyright holder for this preprint (which was not certified by peer review) is the author/funder. This article is a US Government work. It is not subject to copyright under 17 USC 105 and is also made available for use under a CC0 license. 43 INTRODUCTION 44 Persistent infection with high-oncogenic risk HPV types is responsible for almost all cervical and 45 ~70% oropharyngeal carcinomas 1. One factor that can contribute to oncogenic progression of HPV- 46 positive lesions is integration of the viral genome into host chromatin. Integration is associated with 47 increased genetic instability in high-grade cervical intraepithelial neoplasia (CIN) and cervical and 48 oropharyngeal carcinomas 2-9 due to dysregulated expression of the viral oncoproteins, E6 and E7. Many 49 studies have compared the human genomic regions associated with HPV integration sites to elucidate 50 the mechanisms that might promote integration and carcinogenesis. Here, we have curated datasets 51 from The Cancer Genome Atlas and other published sources to define a rigorous database of integration 52 breakpoints and correlated these with “in-house” datasets of common fragile sites and enhancer 53 elements defined in cervical carcinoma cells. 54 Integration of HPV DNA occurs in all human chromosomes; however, integration sites are often 55 found within or in close proximity to common fragile sites 10-13. Common fragile sites are regions of the 56 genome that have difficulty completing replication and, as such, are susceptible to chromosome 57 breakage in mitosis. They are prone to replication stress that can be due to a shortage of replication 58 origins or clashes between replication and transcriptional processes. Therefore, they vary in fragility 59 depending on cell type and disease state 14. Most previous studies that documented an association of 60 HPV integration sites with common fragile sites used the classical FRA-regions that were defined 61 cytogenetically in lymphocytes 9-13,15. As such, these fragile sites are not cell-type specific and are often 62 large, poorly defined regions that cover a large proportion of the human genome. FANCD2 is required 63 for resolution of these genetically unstable sites and, as such, is a marker of common fragile sites 16-18. 64 HPV integration sites occur frequently in amplified regions of the host genome, and focal 65 amplification of cellular flanking sequences at sites of viral integration are frequently observed in HPV- 66 positive tumors 6,7,9,15. Co-amplification of the viral genome and flanking cellular sequences can result 3 bioRxiv preprint doi: https://doi.org/10.1101/2021.08.30.457540; this version posted August 31, 2021. The copyright holder for this preprint (which was not certified by peer review) is the author/funder. This article is a US Government work. It is not subject to copyright under 17 USC 105 and is also made available for use under a CC0 license. 67 from unlicensed initiation of replication at the viral origin resulting in endoreduplication 19-21. 68 Subsequent recombination can result in amplified tandem repeats. Genome amplification can also occur 69 at common fragile sites by breakage-fusion-bridge cycles 22. 70 HPV integration is also enriched at transcriptionally active regions of the host genome 15,23. We 71 previously identified an HPV16 integration site in the W12 20861 cervical cell line that was adjacent to a 72 cell-type specific enhancer. Co-amplification of this regulatory element and the viral genome to 73 approximately 25 copies resulted in the formation of a super-enhancer-like element to drive high viral 74 oncogene expression 24,25. This ‘enhancer-hijacking’ is a novel mechanism by which HPV integration can 75 promote oncogenesis; however, it is unclear how common this mechanism is in HPV-associated cancers. 76 The aim of this study was to examine the association among HPV integration loci, common 77 fragile sites, and genome amplification to determine whether insertion of HPV genomes adjacent to 78 active cellular enhancers often resulted in viral ‘enhancer-hijacking’, and whether genetic instability 79 could result in co-amplification of viral-cellular regulatory repeats to drive oncogenic progression of 80 HPV-associated cancers. We have extended our previously published work 26 to generate a common 81 fragile site dataset in cervical carcinoma cell lines using higher resolution mapping of FANCD2 binding 82 and have mapped cellular enhancers and super-enhancers in an HPV16-positive cell line derived from a 83 cervical lesion27 using H3K27ac and Brd4 ChIP-seq (Figure 1). Here we compare HPV integration sites 84 with these cervical cell-type specific enhancer and fragile site datasets. 4 bioRxiv preprint doi: https://doi.org/10.1101/2021.08.30.457540; this version posted August 31, 2021. The copyright holder for this preprint (which was not certified by peer review) is the author/funder. This article is a US Government work. It is not subject to copyright under 17 USC 105 and is also made available for use under a CC0 license. 85 RESULTS 86 CESC and HNSCC tumors frequently contain multiple, clustered HPV integration breakpoints 87 A dataset of HPV integration breakpoints was assembled from various sources 5,6,8,9,28-35, as 88 outlined in Figure 1 and Supplementary Table S1. Integration breakpoints were defined as the junctions 89 between the viral and host chimeric reads within the human reference genome. A total of 1,299 90 integration breakpoints from 333 cervical carcinomas (CESC) and 119 integration breakpoints from 41 91 head and neck squamous cell carcinomas (HNSCC) were included in this study (Supplementary Figure S1 92 and Supplementary Table S2-S3). We found that many tumor samples contained multiple integration 93 breakpoints that could have resulted from either independent integration events at different 94 chromosomal loci or from amplification of a single integration site resulting in a cluster of multiple, 95 closely spaced breakpoints. To classify this, we defined an integration locus as either a single HPV 96 insertion breakpoint, or as multiple, closely spaced breakpoints (a cluster), Figure 2a. Clustered 97 breakpoints within the same chromosome that had a maximum distance of 3 Mb between the most 5’ 98 and 3’ breakpoints were classified as a single integration locus. Based on this classification, the total 99 number of integration loci analyzed in our study was 584 for CESC samples and 58 for HNSCC samples.
Recommended publications
  • A Novel Approach to Identify Driver Genes Involved in Androgen-Independent Prostate Cancer
    Schinke et al. Molecular Cancer 2014, 13:120 http://www.molecular-cancer.com/content/13/1/120 RESEARCH Open Access A novel approach to identify driver genes involved in androgen-independent prostate cancer Ellyn N Schinke1, Victor Bii1, Arun Nalla1, Dustin T Rae1, Laura Tedrick1, Gary G Meadows1 and Grant D Trobridge1,2* Abstract Background: Insertional mutagenesis screens have been used with great success to identify oncogenes and tumor suppressor genes. Typically, these screens use gammaretroviruses (γRV) or transposons as insertional mutagens. However, insertional mutations from replication-competent γRVs or transposons that occur later during oncogenesis can produce passenger mutations that do not drive cancer progression. Here, we utilized a replication-incompetent lentiviral vector (LV) to perform an insertional mutagenesis screen to identify genes in the progression to androgen-independent prostate cancer (AIPC). Methods: Prostate cancer cells were mutagenized with a LV to enrich for clones with a selective advantage in an androgen-deficient environment provided by a dysregulated gene(s) near the vector integration site. We performed our screen using an in vitro AIPC model and also an in vivo xenotransplant model for AIPC. Our approach identified proviral integration sites utilizing a shuttle vector that allows for rapid rescue of plasmids in E. coli that contain LV long terminal repeat (LTR)-chromosome junctions. This shuttle vector approach does not require PCR amplification and has several advantages over PCR-based techniques. Results: Proviral integrations were enriched near prostate cancer susceptibility loci in cells grown in androgen- deficient medium (p < 0.001), and five candidate genes that influence AIPC were identified; ATPAF1, GCOM1, MEX3D, PTRF, and TRPM4.
    [Show full text]
  • The Interactome of KRAB Zinc Finger Proteins Reveals the Evolutionary History of Their Functional Diversification
    Resource The interactome of KRAB zinc finger proteins reveals the evolutionary history of their functional diversification Pierre-Yves Helleboid1,†, Moritz Heusel2,†, Julien Duc1, Cécile Piot1, Christian W Thorball1, Andrea Coluccio1, Julien Pontis1, Michaël Imbeault1, Priscilla Turelli1, Ruedi Aebersold2,3,* & Didier Trono1,** Abstract years ago (MYA) (Imbeault et al, 2017). Their products harbor an N-terminal KRAB (Kru¨ppel-associated box) domain related to that of Krüppel-associated box (KRAB)-containing zinc finger proteins Meisetz (a.k.a. PRDM9), a protein that originated prior to the diver- (KZFPs) are encoded in the hundreds by the genomes of higher gence of chordates and echinoderms, and a C-terminal array of zinc vertebrates, and many act with the heterochromatin-inducing fingers (ZNF) with sequence-specific DNA-binding potential (Urru- KAP1 as repressors of transposable elements (TEs) during early tia, 2003; Birtle & Ponting, 2006; Imbeault et al, 2017). KZFP genes embryogenesis. Yet, their widespread expression in adult tissues multiplied by gene and segment duplication to count today more and enrichment at other genetic loci indicate additional roles. than 350 and 700 representatives in the human and mouse Here, we characterized the protein interactome of 101 of the ~350 genomes, respectively (Urrutia, 2003; Kauzlaric et al, 2017). A human KZFPs. Consistent with their targeting of TEs, most KZFPs majority of human KZFPs including all primate-restricted family conserved up to placental mammals essentially recruit KAP1 and members target sequences derived from TEs, that is, DNA trans- associated effectors. In contrast, a subset of more ancient KZFPs posons, ERVs (endogenous retroviruses), LINEs, SINEs (long and rather interacts with factors related to functions such as genome short interspersed nuclear elements, respectively), or SVAs (SINE- architecture or RNA processing.
    [Show full text]
  • Análise Integrativa De Perfis Transcricionais De Pacientes Com
    UNIVERSIDADE DE SÃO PAULO FACULDADE DE MEDICINA DE RIBEIRÃO PRETO PROGRAMA DE PÓS-GRADUAÇÃO EM GENÉTICA ADRIANE FEIJÓ EVANGELISTA Análise integrativa de perfis transcricionais de pacientes com diabetes mellitus tipo 1, tipo 2 e gestacional, comparando-os com manifestações demográficas, clínicas, laboratoriais, fisiopatológicas e terapêuticas Ribeirão Preto – 2012 ADRIANE FEIJÓ EVANGELISTA Análise integrativa de perfis transcricionais de pacientes com diabetes mellitus tipo 1, tipo 2 e gestacional, comparando-os com manifestações demográficas, clínicas, laboratoriais, fisiopatológicas e terapêuticas Tese apresentada à Faculdade de Medicina de Ribeirão Preto da Universidade de São Paulo para obtenção do título de Doutor em Ciências. Área de Concentração: Genética Orientador: Prof. Dr. Eduardo Antonio Donadi Co-orientador: Prof. Dr. Geraldo A. S. Passos Ribeirão Preto – 2012 AUTORIZO A REPRODUÇÃO E DIVULGAÇÃO TOTAL OU PARCIAL DESTE TRABALHO, POR QUALQUER MEIO CONVENCIONAL OU ELETRÔNICO, PARA FINS DE ESTUDO E PESQUISA, DESDE QUE CITADA A FONTE. FICHA CATALOGRÁFICA Evangelista, Adriane Feijó Análise integrativa de perfis transcricionais de pacientes com diabetes mellitus tipo 1, tipo 2 e gestacional, comparando-os com manifestações demográficas, clínicas, laboratoriais, fisiopatológicas e terapêuticas. Ribeirão Preto, 2012 192p. Tese de Doutorado apresentada à Faculdade de Medicina de Ribeirão Preto da Universidade de São Paulo. Área de Concentração: Genética. Orientador: Donadi, Eduardo Antonio Co-orientador: Passos, Geraldo A. 1. Expressão gênica – microarrays 2. Análise bioinformática por module maps 3. Diabetes mellitus tipo 1 4. Diabetes mellitus tipo 2 5. Diabetes mellitus gestacional FOLHA DE APROVAÇÃO ADRIANE FEIJÓ EVANGELISTA Análise integrativa de perfis transcricionais de pacientes com diabetes mellitus tipo 1, tipo 2 e gestacional, comparando-os com manifestações demográficas, clínicas, laboratoriais, fisiopatológicas e terapêuticas.
    [Show full text]
  • Association of Gene Ontology Categories with Decay Rate for Hepg2 Experiments These Tables Show Details for All Gene Ontology Categories
    Supplementary Table 1: Association of Gene Ontology Categories with Decay Rate for HepG2 Experiments These tables show details for all Gene Ontology categories. Inferences for manual classification scheme shown at the bottom. Those categories used in Figure 1A are highlighted in bold. Standard Deviations are shown in parentheses. P-values less than 1E-20 are indicated with a "0". Rate r (hour^-1) Half-life < 2hr. Decay % GO Number Category Name Probe Sets Group Non-Group Distribution p-value In-Group Non-Group Representation p-value GO:0006350 transcription 1523 0.221 (0.009) 0.127 (0.002) FASTER 0 13.1 (0.4) 4.5 (0.1) OVER 0 GO:0006351 transcription, DNA-dependent 1498 0.220 (0.009) 0.127 (0.002) FASTER 0 13.0 (0.4) 4.5 (0.1) OVER 0 GO:0006355 regulation of transcription, DNA-dependent 1163 0.230 (0.011) 0.128 (0.002) FASTER 5.00E-21 14.2 (0.5) 4.6 (0.1) OVER 0 GO:0006366 transcription from Pol II promoter 845 0.225 (0.012) 0.130 (0.002) FASTER 1.88E-14 13.0 (0.5) 4.8 (0.1) OVER 0 GO:0006139 nucleobase, nucleoside, nucleotide and nucleic acid metabolism3004 0.173 (0.006) 0.127 (0.002) FASTER 1.28E-12 8.4 (0.2) 4.5 (0.1) OVER 0 GO:0006357 regulation of transcription from Pol II promoter 487 0.231 (0.016) 0.132 (0.002) FASTER 6.05E-10 13.5 (0.6) 4.9 (0.1) OVER 0 GO:0008283 cell proliferation 625 0.189 (0.014) 0.132 (0.002) FASTER 1.95E-05 10.1 (0.6) 5.0 (0.1) OVER 1.50E-20 GO:0006513 monoubiquitination 36 0.305 (0.049) 0.134 (0.002) FASTER 2.69E-04 25.4 (4.4) 5.1 (0.1) OVER 2.04E-06 GO:0007050 cell cycle arrest 57 0.311 (0.054) 0.133 (0.002)
    [Show full text]
  • A CRISPR-Based Screen for Hedgehog Signaling Provides Insights Into Ciliary Function and Ciliopathies
    ARTICLES https://doi.org/10.1038/s41588-018-0054-7 A CRISPR-based screen for Hedgehog signaling provides insights into ciliary function and ciliopathies David K. Breslow 1,2,7*, Sascha Hoogendoorn 3,7, Adam R. Kopp2, David W. Morgens4, Brandon K. Vu2, Margaret C. Kennedy1, Kyuho Han4, Amy Li4, Gaelen T. Hess4, Michael C. Bassik4, James K. Chen 3,5* and Maxence V. Nachury 2,6* Primary cilia organize Hedgehog signaling and shape embryonic development, and their dysregulation is the unifying cause of ciliopathies. We conducted a functional genomic screen for Hedgehog signaling by engineering antibiotic-based selection of Hedgehog-responsive cells and applying genome-wide CRISPR-mediated gene disruption. The screen can robustly identify factors required for ciliary signaling with few false positives or false negatives. Characterization of hit genes uncovered novel components of several ciliary structures, including a protein complex that contains δ -tubulin and ε -tubulin and is required for centriole maintenance. The screen also provides an unbiased tool for classifying ciliopathies and showed that many congenital heart disorders are caused by loss of ciliary signaling. Collectively, our study enables a systematic analysis of ciliary function and of ciliopathies, and also defines a versatile platform for dissecting signaling pathways through CRISPR-based screening. he primary cilium is a surface-exposed microtubule-based approach. Indeed, most studies to date have searched for genes that compartment that serves as an organizing center for diverse either intrinsically affect cell growth or affect sensitivity to applied Tsignaling pathways1–3. Mutations affecting cilia cause ciliopa- perturbations16–23. thies, a group of developmental disorders including Joubert syn- Here, we engineered a Hh-pathway-sensitive reporter to enable drome, Meckel syndrome (MKS), nephronophthisis (NPHP), and an antibiotic-based selection platform.
    [Show full text]
  • The Complex SNP and CNV Genetic Architecture of the Increased Risk of Congenital Heart Defects in Down Syndrome
    Downloaded from genome.cshlp.org on September 24, 2021 - Published by Cold Spring Harbor Laboratory Press Research The complex SNP and CNV genetic architecture of the increased risk of congenital heart defects in Down syndrome M. Reza Sailani,1,2 Periklis Makrythanasis,1 Armand Valsesia,3,4,5 Federico A. Santoni,1 Samuel Deutsch,1 Konstantin Popadin,1 Christelle Borel,1 Eugenia Migliavacca,1 Andrew J. Sharp,1,20 Genevieve Duriaux Sail,1 Emilie Falconnet,1 Kelly Rabionet,6,7,8 Clara Serra-Juhe´,7,9 Stefano Vicari,10 Daniela Laux,11 Yann Grattau,12 Guy Dembour,13 Andre Megarbane,12,14 Renaud Touraine,15 Samantha Stora,12 Sofia Kitsiou,16 Helena Fryssira,16 Chariklia Chatzisevastou-Loukidou,16 Emmanouel Kanavakis,16 Giuseppe Merla,17 Damien Bonnet,11 Luis A. Pe´rez-Jurado,7,9 Xavier Estivill,6,7,8 Jean M. Delabar,18 and Stylianos E. Antonarakis1,2,19,21 1–19[Author affiliations appear at the end of the paper.] Congenital heart defect (CHD) occurs in 40% of Down syndrome (DS) cases. While carrying three copies of chromosome 21 increases the risk for CHD, trisomy 21 itself is not sufficient to cause CHD. Thus, additional genetic variation and/or environmental factors could contribute to the CHD risk. Here we report genomic variations that in concert with trisomy 21, determine the risk for CHD in DS. This case-control GWAS includes 187 DS with CHD (AVSD = 69, ASD = 53, VSD = 65) as cases, and 151 DS without CHD as controls. Chromosome 21–specific association studies revealed rs2832616 and rs1943950 as CHD risk alleles (adjusted genotypic P-values <0.05).
    [Show full text]
  • Role of the PTPRD Locus and Limited Pleiotropy with Other Neuropathologies
    CORE Metadata, citation and similar papers at core.ac.uk Provided by Harvard University - DASH Susceptibility to neurofibrillary tangles: role of the PTPRD locus and limited pleiotropy with other neuropathologies The Harvard community has made this article openly available. Please share how this access benefits you. Your story matters Citation Chibnik, L. B., C. C. White, S. Mukherjee, T. Raj, L. Yu, E. B. Larson, T. J. Montine, et al. 2017. “Susceptibility to neurofibrillary tangles: role of the PTPRD locus and limited pleiotropy with other neuropathologies.” Molecular psychiatry :10.1038/mp.2017.20. doi:10.1038/mp.2017.20. http://dx.doi.org/10.1038/mp.2017.20. Published Version doi:10.1038/mp.2017.20 Citable link http://nrs.harvard.edu/urn-3:HUL.InstRepos:34492045 Terms of Use This article was downloaded from Harvard University’s DASH repository, and is made available under the terms and conditions applicable to Other Posted Material, as set forth at http:// nrs.harvard.edu/urn-3:HUL.InstRepos:dash.current.terms-of- use#LAA HHS Public Access Author manuscript Author ManuscriptAuthor Manuscript Author Mol Psychiatry Manuscript Author . Author Manuscript Author manuscript; available in PMC 2017 September 22. Susceptibility to neurofibrillary tangles: role of the PTPRD locus and limited pleiotropy with other neuropathologies Lori B Chibnik, PhD, MPH1,2,3,4, Charles C White, PhD1,3, Shubhabrata Mukherjee, PhD5, Towfique Raj, PhD1,3, Lei Yu, PhD6, Eric B. Larson, MD, MPH7, Thomas J. Montine, MD, PhD8, C. Dirk Keene, MD, PhD8, Joshua Sonnen, MD9, Julie A. Schneider, MD6, Paul K. Crane, MD, MPH5, Joshua M.
    [Show full text]
  • Genetic Interactions Between ANLN and KDR Are Prognostic for Breast Cancer Survival
    ONCOLOGY REPORTS 42: 2255-2266, 2019 Genetic interactions between ANLN and KDR are prognostic for breast cancer survival XIAOFENG DAI1*, XIAO CHEN2*, OLIVIER HAKIZIMANA2 and YI MEI2 1Wuxi School of Medicine, 2School of Biotechnology, Jiangnan University, Wuxi, Jiangsu 214122, P.R. China Received April 3, 2019; Accepted August 7, 2019 DOI: 10.3892/or.2019.7332 Abstract. Single nucleotide polymorphisms (SNPs) are the of ~627,000 annually estimated in 2018 (2). Uncontrolled most common genetic variation in mammalian cells with proliferative growth and angiogenesis are two basic cancer prognostic potential. Anillin-actin binding protein (ANLN) hallmarks governing the critical transitions towards malig- has been identified as being involved in PI3K/PTEN signaling, nancy during carcinogenesis (3). PI3K/PTEN signaling, which is critical in cell life/death control, and kinase insert frequently altered in breast carcinoma (4), confers a survival domain receptor (KDR) encodes a key receptor mediating advantage to tumor cells (5). Anillin, encoded by anillin the cancer angiogenesis/metastasis switch. Knowledge of actin-binding protein (ANLN), is an actin-binding protein, the intrinsic connections between PI3K/PTEN and KDR which has been identified as being involved in the PI3K/PTEN signaling, which represent two critical transitions in carcino- pathway (6,7). It is an F‑actin binding protein, which maintains genesis, led the present study to investigate the effects of the podocyte cytoskeletal dynamics, cell motility and signaling potential synergy between ANLN and KDR on breast cancer through its interaction with CD2-associated protein, which outcome and identify relevant SNPs driving such a synergy stimulates the phosphorylation of AKT at serine 473 (6,8).
    [Show full text]
  • Research Article Clinic-Genomic Association Mining for Colorectal Cancer Using Publicly Available Datasets
    Hindawi Publishing Corporation BioMed Research International Volume 2014, Article ID 170289, 10 pages http://dx.doi.org/10.1155/2014/170289 Research Article Clinic-Genomic Association Mining for Colorectal Cancer Using Publicly Available Datasets Fang Liu,1 Yaning Feng,1 Zhenye Li,2 Chao Pan,1 Yuncong Su,1 Rui Yang,1 Liying Song,1 Huilong Duan,1 and Ning Deng1 1 Department of Biomedical Engineering, Key Laboratory for Biomedical Engineering of Ministry of Education, Zhejiang University, Hangzhou 310027, China 2 General Hospital of Ningxia Medical University, Yinchuan 750004, China Correspondence should be addressed to Ning Deng; [email protected] Received 30 March 2014; Accepted 12 May 2014; Published 2 June 2014 Academic Editor: Degui Zhi Copyright © 2014 Fang Liu et al. This is an open access article distributed under the Creative Commons Attribution License, which permits unrestricted use, distribution, and reproduction in any medium, provided the original work is properly cited. In recent years, a growing number of researchers began to focus on how to establish associations between clinical and genomic data. However, up to now, there is lack of research mining clinic-genomic associations by comprehensively analysing available gene expression data for a single disease. Colorectal cancer is one of the malignant tumours. A number of genetic syndromes have been proven to be associated with colorectal cancer. This paper presents our research on mining clinic-genomic associations for colorectal cancer under biomedical big data environment. The proposed method is engineered with multiple technologies, including extracting clinical concepts using the unified medical language system (UMLS), extracting genes through the literature mining, and mining clinic-genomic associations through statistical analysis.
    [Show full text]
  • Transcriptional Consequences of Genomic Structural Aberrations in Breast Cancer
    Downloaded from genome.cshlp.org on September 30, 2021 - Published by Cold Spring Harbor Laboratory Press Research Transcriptional consequences of genomic structural aberrations in breast cancer Koichiro Inaki,1,10 Axel M. Hillmer,2,10 Leena Ukil,1,10 Fei Yao,2,3 Xing Yi Woo,4 Leah A. Vardy,5 Kelson Folkvard Braaten Zawack,4 Charlie Wah Heng Lee,4 Pramila Nuwantha Ariyaratne,4 Yang Sun Chan,1 Kartiki Vasant Desai,1 Jonas Bergh,6 Per Hall,7 Thomas Choudary Putti,8 Wai Loon Ong,9 Atif Shahab,9 Valere Cacheux-Rataboul,1 Radha Krishna Murthy Karuturi,4 Wing-Kin Sung,4 Xiaoan Ruan,2 Guillaume Bourque,4 Yijun Ruan,2 and Edison T. Liu1,11 1Cancer Biology and Pharmacology, Genome Institute of Singapore, Genome, Singapore 138672, Singapore; 2Genome Technology and Biology, Genome Institute of Singapore, Genome, Singapore 138672, Singapore; 3Department of Epidemiology and Public Health, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117597, Singapore; 4Computational and Mathematical Biology, Genome Institute of Singapore, Genome, Singapore 138672, Singapore; 5Institute of Medical Biology, Immunos, Singapore 138648, Singapore; 6Department of Oncology–Pathology, Karolinska Institute, SE-17177 Stockholm, Sweden; 7Department of Medical Epidemiology and Biostatistics, Karolinska Institute, SE-17177 Stockholm, Sweden; 8Department of Pathology, National University of Singapore, Singapore 119077, Singapore; 9Research Computing, Genome Institute of Singapore, Genome, Singapore 138672, Singapore Using a long-span, paired-end deep sequencing strategy, we have comprehensively identified cancer genome rear- rangements in eight breast cancer genomes. Herein, we show that 40%–54% of these structural genomic rearrangements result in different forms of fusion transcripts and that 44% are potentially translated.
    [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]
  • Co-Expression Network of Neural-Differentiation Genes Shows
    Maschietto et al. BMC Medical Genomics (2015) 8:23 DOI 10.1186/s12920-015-0098-9 RESEARCH ARTICLE Open Access Co-expression network of neural-differentiation genes shows specific pattern in schizophrenia Mariana Maschietto1,2†, Ana C Tahira1,2†, Renato Puga3, Leandro Lima4, Daniel Mariani4, Bruna da Silveira Paulsen5, Paulo Belmonte-de-Abreu6, Henrique Vieira4, Ana CV Krepischi7, Dirce M Carraro8, Joana A Palha9,10, Stevens Rehen5,11 and Helena Brentani1,2,12,13* Abstract Background: Schizophrenia is a neurodevelopmental disorder with genetic and environmental factors contributing to its pathogenesis, although the mechanism is unknown due to the difficulties in accessing diseased tissue during human neurodevelopment. The aim of this study was to find neuronal differentiation genes disrupted in schizophrenia and to evaluate those genes in post-mortem brain tissues from schizophrenia cases and controls. Methods: We analyzed differentially expressed genes (DEG), copy number variation (CNV) and differential methylation in human induced pluripotent stem cells (hiPSC) derived from fibroblasts from one control and one schizophrenia patient and further differentiated into neuron (NPC). Expression of the DEG were analyzed with microarrays of post-mortem brain tissue (frontal cortex) cohort of 29 schizophrenia cases and 30 controls. A Weighted Gene Co-expression Network Analysis (WGCNA) using the DEG was used to detect clusters of co-expressed genes that werenon-conserved between adult cases and controls brain samples. Results: We identified methylation alterations potentially involved with neuronal differentiation in schizophrenia, which displayed an over-representation of genes related to chromatin remodeling complex (adjP = 0.04). We found 228 DEG associated with neuronal differentiation.
    [Show full text]