Prediction of Future Metastasis and Molecular Characterization of Head and Neck Squamous-Cell Carcinoma Based on Transcriptome and Genome Analysis Bymicroarrays

Total Page:16

File Type:pdf, Size:1020Kb

Prediction of Future Metastasis and Molecular Characterization of Head and Neck Squamous-Cell Carcinoma Based on Transcriptome and Genome Analysis Bymicroarrays Oncogene (2008) 27, 6607–6622 & 2008 Macmillan Publishers Limited All rights reserved 0950-9232/08 $32.00 www.nature.com/onc ONCOGENOMICS Prediction of future metastasis and molecular characterization of head and neck squamous-cell carcinoma based on transcriptome and genome analysis bymicroarrays DS Rickman1,4, R Millon2, A De Reynies1, E Thomas1, C Wasylyk3, D Muller2, J Abecassis2 and BWasylyk 3 1Ligue Nationale Contre le Cancer, Paris, France; 2Centre Re´gional de Lutte Contre le Cancer Paul Strauss, 3 rue Porte de l’Ho¨pital, Strasbourg, France and 3IGBMC CNRS/INSERM/ULP, Illkirch, France Propensityfor subsequent distant metastasis in head and Introduction neck squamous-cell carcinoma (HNSCC) was analysed using 186 primarytumours from patients initiallytreated There are various aetiological factors for head and neck bysurgerythat developed (M) or did not develop (NM) squamous-cell carcinoma (HNSCC), including alcohol metastases as the first recurrent event. Transcriptome consumption with smoking and human papilloma- (Affymetrix HGU133_Plus2, QRT–PCR) and array- virus (HPV) infection, for our patient population comparative genomic hybridization data were collected. (Applebaum et al., 2007; Hashibe et al., 2007). Survival Non-supervised hierarchical clustering based on Affyme- is still poor (5-year survival 30–50%), mainly due to trix data distinguished tumours differing in pathological relapse, metastasis or second cancer (Forastiere et al., differentiation, and identified associated functional 2001; Le Tourneau et al., 2005). The anatomic location changes. Propensityfor metastasis was not associated and TNM staging guide treatment selection, but patients with these subgroups. Using QRT–PCR data we identified with similar tumour characteristics differ in their clinical a four-gene model (PSMD10, HSD17B12, FLOT2 and outcome. Our aim is to identify, in primary tumours, KRT17) that predicts M/NM status with 77% success in molecular signatures that predict the subsequent devel- a separate 79-sample validation group of HNSCC opment of distant metastases in patients treated with samples. This prediction is independent of clinical criteria complete surgical resection and adjuvant therapy. (age, lymph node status, stage, differentiation and Furthermore, we want to study the still poorly under- localization). The most significantlyaltered transcripts stood biological processes that predispose human in M versus NM were significantlyassociated to tumours to the development of metastases, and to metastasis-related functions, including adhesion, mobility define targets for therapy. and cell survival. Several genomic modifications were Our previous study, using large-scale validated significantlyassociated with M/NM status (most notably differential display, identified 820 transcripts that were gains at 4q11–22 and Xq12–28; losses at 11q14–24 and differentially expressed between tumours and normal 17q11 losses) and partlylinked to transcription modifica- tissue, about 10% of which differed between tumours tions. This work yields a basis for the development of that did (M) or did not (NM) develop metastasis as the prognostic molecular signatures, markers and therapeutic first recurrence event (Carles et al., 2006). A subsequent targets for HNSCC metastasis. study, with 28 tumours and Affymetrix U95A arrays Oncogene (2008) 27, 6607–6622; doi:10.1038/onc.2008.251; (Affymetrix, Santa Clara, CA, USA) detected 164 published online 4 August 2008 transcripts whose levels differed significantly between N and NM tumours, but we did not find changes that Keywords: HNSCC; distant metastasis; prognosis; could be used to predict N/NM status in independent intrinsic groups; differentiation samples (Cromer et al., 2004). Signatures of poor ONCOGENOMICS prognosis and metastasis have been identified in other larger scale studies (review: Nguyen and Massague, 2007). We now report a larger study, using more samples (186) and RNA variables (Affymetrix HG- U133 plus 2.0 GeneChips), and we have included an Correspondence: Dr BWasylyk, IGBMC,CNRS/INSERM/ULP, analysis of genomic changes (IntegraChip 4.4K bacterial 1 rue Laurent Fries, BP 10142, Illkirch 67404, France. artificial chromosomes (BAC) comparative genomic E-mail: [email protected] hybridisation (CGH) arrays, array-CGH (aCGH)). 4Current address: Department of Pathology and Laboratory Medicine, Using unsupervised analysis, we defined intrinsic groups Weill Cornell Medical Center, 1300 York Avenue, Room C-458D, New York, NY 10021, USA. that correspond to pathological differentiation. Using Received 6 February 2008; revised 21 May 2008; accepted 27 June 2008; QRT–PCR-validated RNA levels and a training group, published online 4 August 2008 we have defined a four-gene model that predicts future HNSCC, distant metastasis signatures and classifiers DS Rickman et al 6608 distant metastasis with 77% success in an independent associated GO terms and pathways. For example, validation group. We have identified genomic and poorly differentiated tumours (C3) are characterized transcriptomic changes that are significantly different by upregulation in gene cluster a, which is significantly in primary tumours with dissimilar M/NM status. These associated with cell motility. Sample group C4 is findings are useful for the development of prospective associated with upregulation of genes encoding for signatures of metastasis, and for the understanding of proteins related to muscle structure and function biological processes that predispose to the development (approximately half of the 59 significant GO terms that of metastases in patients with HNSCC. are enriched in the gene cluster f, Figure 1; see the GO analysis Supplementary Table 2 sheet 2, and the genes in cluster f in Supplementary Table 2 sheet 1). This is a clue to the origin of cluster C4. Results To further analyse the biological functions associated with pathologically defined differentiation (as opposed Classification of tumours to the intrinsic groups defined by unsupervised classifi- The global differences between the transcriptomes of the cation) we performed a supervised analysis between the HNSCC tumours may result from metastatic propen- tumours classified according to pathological differentia- sity, or other clinical or biological features. To detect tion (Table 1; Supplementary Table 3). Using 1-way these ‘intrinsic’ features, we performed unsupervised ANOVA (Po0.01) and pairwise Wilcoxon tests hierarchical classification as already described (Boyault (Po0.001) we established three lists of genes (a total et al., 2007). We studied extensively the robustness of of 835 genes) that were significantly differentially the topologies (series of dendrograms) obtained under expressed between at least two of the three clinically different conditions: three distinct agglomerative clus- defined groups (Supplementary Table 4). As expected, a tering methods, various thresholds for variance based number of the genes (50) involved in the unsupervised unsupervised gene selection, resampling and addition of clustering (Figure 1) were among the 835 significant Gaussian noise to the data. We found that the sample genes obtained from the supervised analysis. QRT–PCR partitions that yielded between two to four sample validation of seven genes confirmed that there is a groups were very reproducible (Supplementary Table 1) gradient of expression between poorly to well-differ- and identified a consensus partition of four groups entiated tumours (Figure 2; in general there was a very (Supplementary Figure 1). For illustrative purposes, high correlation between the Affymetrix and QRT– we selected a representative sample dendrogram that PCR, data not shown). These genes were chosen as was most related to the consensus partition (Figure 1). representative genes that are differentially expressed in Using Fisher’s exact tests we found that these four tumours according to their pathologically defined major clusters (C1–C4) were not significantly associated differentiation status, and due to the potential impor- with stage, localization, metastasis-free survival tance of the encoded proteins for cell differentiation (see (Figure 1), or any of the other characteristics of the Supplementary Table 5, column H, for a brief descrip- tumours analysed (data not shown). However, C1–C3 tion). Further analysis of the genes that are differentially were significantly associated with degree of differentia- expressed between the tumours could provide insights tion, corresponding to well (C1), moderately (C2) and into the molecular events that are related to pathological poorly (C3) differentiated tumours. C4 was not associa- differentiation of HNSCC. tion with any of the characteristics analysed. These results suggest that pathological differentiation has the biggest effect on the overall differences in transcription Four-gene model predicting future metastasis in HNSCC of the tumours. that is independent of clinical variables To get a better understanding of the molecular Given that metastasis was not associated with any of the determinants of the intrinsic groups, we analysed the intrinsic HNSCC subgroups, we set out to identify a 449-gene list (548 probe sets) that was used to generate molecular signature that is predictive of developing the four clusters in the representative dendrogram future metastasis irrespective of differentiation status shown in Figure 1. Unsupervised cluster analysis was and/or other clinical variables, and that could be used to segregate the genes into six gene groups (a–f; employed in a clinical
Recommended publications
  • Polyphosphate-Dependent Synthesis of ATP and ADP by the Family-2 Polyphosphate Kinases in Bacteria
    Polyphosphate-dependent synthesis of ATP and ADP by the family-2 polyphosphate kinases in bacteria Boguslaw Noceka, Samvel Kochinyanb, Michael Proudfootb, Greg Brownb, Elena Evdokimovaa,b, Jerzy Osipiuka, Aled M. Edwardsa,b, Alexei Savchenkoa,b, Andrzej Joachimiaka,c,1, and Alexander F. Yakuninb,1 aMidwest Center for Structural Genomics and Structural Biology Center, Department of Biosciences, Argonne National Laboratory, 9700 South Cass Avenue, Building 202, Argonne, IL 60439; bBanting and Best Department of Medical Research, University of Toronto, Toronto, ON, Canada M5G 1L6; and cDepartment of Biochemistry and Molecular Biology, University of Chicago, 920 East 58th Street, Chicago, IL 60637 Edited by Robert Haselkorn, University of Chicago, Chicago, IL, and approved September 30, 2008 (received for review August 1, 2008) Inorganic polyphosphate (polyP) is a linear polymer of tens or hun- exopolysaccharide essential for the virulence of P. aeruginosa (12). dreds of phosphate residues linked by high-energy bonds. It is found In the social slime mold Dictyostelium discoideum, a different PPK in all organisms and has been proposed to serve as an energy source was found (DdPPK2) with sequence and properties similar to that in a pre-ATP world. This ubiquitous and abundant biopolymer plays of actin-related proteins (Arps) (14). DdPPK2 is a complex of 3 numerous and vital roles in metabolism and regulation in prokaryotes Arps (Arp1, Arp2, and Arpx) and resembles the actin family in and eukaryotes, but the underlying molecular mechanisms for most molecular weight, sequence, and filamentous structure. Remark- activities of polyP remain unknown. In prokaryotes, the synthesis and ably, DdPPK2 can polymerize into an actin-like filament concurrent utilization of polyP are catalyzed by 2 families of polyP kinases, PPK1 with the synthesis of polyP (14).
    [Show full text]
  • Genomic Correlates of Relationship QTL Involved in Fore- Versus Hind Limb Divergence in Mice
    Loyola University Chicago Loyola eCommons Biology: Faculty Publications and Other Works Faculty Publications 2013 Genomic Correlates of Relationship QTL Involved in Fore- Versus Hind Limb Divergence in Mice Mihaela Palicev Gunter P. Wagner James P. Noonan Benedikt Hallgrimsson James M. Cheverud Loyola University Chicago, [email protected] Follow this and additional works at: https://ecommons.luc.edu/biology_facpubs Part of the Biology Commons Recommended Citation Palicev, M, GP Wagner, JP Noonan, B Hallgrimsson, and JM Cheverud. "Genomic Correlates of Relationship QTL Involved in Fore- Versus Hind Limb Divergence in Mice." Genome Biology and Evolution 5(10), 2013. This Article is brought to you for free and open access by the Faculty Publications at Loyola eCommons. It has been accepted for inclusion in Biology: Faculty Publications and Other Works by an authorized administrator of Loyola eCommons. For more information, please contact [email protected]. This work is licensed under a Creative Commons Attribution-Noncommercial-No Derivative Works 3.0 License. © Palicev et al., 2013. GBE Genomic Correlates of Relationship QTL Involved in Fore- versus Hind Limb Divergence in Mice Mihaela Pavlicev1,2,*, Gu¨ nter P. Wagner3, James P. Noonan4, Benedikt Hallgrı´msson5,and James M. Cheverud6 1Konrad Lorenz Institute for Evolution and Cognition Research, Altenberg, Austria 2Department of Pediatrics, Cincinnati Children‘s Hospital Medical Center, Cincinnati, Ohio 3Yale Systems Biology Institute and Department of Ecology and Evolutionary Biology, Yale University 4Department of Genetics, Yale University School of Medicine 5Department of Cell Biology and Anatomy, The McCaig Institute for Bone and Joint Health and the Alberta Children’s Hospital Research Institute for Child and Maternal Health, University of Calgary, Calgary, Canada 6Department of Anatomy and Neurobiology, Washington University *Corresponding author: E-mail: [email protected].
    [Show full text]
  • Analysis of Gene Expression Data for Gene Ontology
    ANALYSIS OF GENE EXPRESSION DATA FOR GENE ONTOLOGY BASED PROTEIN FUNCTION PREDICTION A Thesis Presented to The Graduate Faculty of The University of Akron In Partial Fulfillment of the Requirements for the Degree Master of Science Robert Daniel Macholan May 2011 ANALYSIS OF GENE EXPRESSION DATA FOR GENE ONTOLOGY BASED PROTEIN FUNCTION PREDICTION Robert Daniel Macholan Thesis Approved: Accepted: _______________________________ _______________________________ Advisor Department Chair Dr. Zhong-Hui Duan Dr. Chien-Chung Chan _______________________________ _______________________________ Committee Member Dean of the College Dr. Chien-Chung Chan Dr. Chand K. Midha _______________________________ _______________________________ Committee Member Dean of the Graduate School Dr. Yingcai Xiao Dr. George R. Newkome _______________________________ Date ii ABSTRACT A tremendous increase in genomic data has encouraged biologists to turn to bioinformatics in order to assist in its interpretation and processing. One of the present challenges that need to be overcome in order to understand this data more completely is the development of a reliable method to accurately predict the function of a protein from its genomic information. This study focuses on developing an effective algorithm for protein function prediction. The algorithm is based on proteins that have similar expression patterns. The similarity of the expression data is determined using a novel measure, the slope matrix. The slope matrix introduces a normalized method for the comparison of expression levels throughout a proteome. The algorithm is tested using real microarray gene expression data. Their functions are characterized using gene ontology annotations. The results of the case study indicate the protein function prediction algorithm developed is comparable to the prediction algorithms that are based on the annotations of homologous proteins.
    [Show full text]
  • Annexina7 and CAP1 Are Associated with Regulating Tumor Cell Adhesion Molecule Expression and Biological Behavior
    AnnexinA7 and CAP1 are associated with regulating tumor cell adhesion molecule expression and biological behavior Type Research paper Keywords hepatocellular carcinoma, adhesion molecule, AnnexinA7, CAP1, Biological behaviors Abstract Introduction To find out the correlations between the effects of down-regulating AnnexinA7 and CAP1 gene on cell adhesion factors and the biological behavior of Hca-p cells cells, and finally infer the relationship between the two genes. Material and methods Western blot ,qRT-PCR,immunocytochemistry, CCK8 cell proliferation, flow cytometry, lymph node adhesion and transwell chamber assay testing . Results AnnexinA7 and CAP1 were consistent with the regulation of the expression of the adhesion molecules such as FAK, Src, Paxillin and E-cadherin. At the mRNA and protein levels, the expression of FAK, Src and Paxillin were increased with down-regulated AnnexinA7 and CAP1 genes, while E-cadherin was down-regulated in the change of these two genes. And the low expression of AnnexinA7 could affect the expression of CAP1 in mRNA and protein levels. otherwise, the localization of AnnexinA7 and CAP1 in hepatocellular carcinoma cells was also the same. After down-regulating the expression of CAP1, the functions of proliferation, lymph node adhesion and invasion were increased and earlyPreprint apoptotic ability was decreased in Hca-P cells. Conclusions AnnexinA7 and CAP1 could control the expression of these adhesion molecules in the same trend. We speculated they may be co-localization. And AnnexinA7 gene may be related to the molecular mechanism of CAP1 gene, which is likely to have a consistent effect on cell adhesion molecules and be closely related to the biological behaviors of Hca-P cells.AnnexinA7 and CAP1 may play an inhibitory role in lymph node metastasis to provide a reliable basis for the early identification of lymphatic metastasis in hepatocellular carcinoma.
    [Show full text]
  • Protein Interaction Network of Alternatively Spliced Isoforms from Brain Links Genetic Risk Factors for Autism
    ARTICLE Received 24 Aug 2013 | Accepted 14 Mar 2014 | Published 11 Apr 2014 DOI: 10.1038/ncomms4650 OPEN Protein interaction network of alternatively spliced isoforms from brain links genetic risk factors for autism Roser Corominas1,*, Xinping Yang2,3,*, Guan Ning Lin1,*, Shuli Kang1,*, Yun Shen2,3, Lila Ghamsari2,3,w, Martin Broly2,3, Maria Rodriguez2,3, Stanley Tam2,3, Shelly A. Trigg2,3,w, Changyu Fan2,3, Song Yi2,3, Murat Tasan4, Irma Lemmens5, Xingyan Kuang6, Nan Zhao6, Dheeraj Malhotra7, Jacob J. Michaelson7,w, Vladimir Vacic8, Michael A. Calderwood2,3, Frederick P. Roth2,3,4, Jan Tavernier5, Steve Horvath9, Kourosh Salehi-Ashtiani2,3,w, Dmitry Korkin6, Jonathan Sebat7, David E. Hill2,3, Tong Hao2,3, Marc Vidal2,3 & Lilia M. Iakoucheva1 Increased risk for autism spectrum disorders (ASD) is attributed to hundreds of genetic loci. The convergence of ASD variants have been investigated using various approaches, including protein interactions extracted from the published literature. However, these datasets are frequently incomplete, carry biases and are limited to interactions of a single splicing isoform, which may not be expressed in the disease-relevant tissue. Here we introduce a new interactome mapping approach by experimentally identifying interactions between brain-expressed alternatively spliced variants of ASD risk factors. The Autism Spliceform Interaction Network reveals that almost half of the detected interactions and about 30% of the newly identified interacting partners represent contribution from splicing variants, emphasizing the importance of isoform networks. Isoform interactions greatly contribute to establishing direct physical connections between proteins from the de novo autism CNVs. Our findings demonstrate the critical role of spliceform networks for translating genetic knowledge into a better understanding of human diseases.
    [Show full text]
  • Seq2pathway Vignette
    seq2pathway Vignette Bin Wang, Xinan Holly Yang, Arjun Kinstlick May 19, 2021 Contents 1 Abstract 1 2 Package Installation 2 3 runseq2pathway 2 4 Two main functions 3 4.1 seq2gene . .3 4.1.1 seq2gene flowchart . .3 4.1.2 runseq2gene inputs/parameters . .5 4.1.3 runseq2gene outputs . .8 4.2 gene2pathway . 10 4.2.1 gene2pathway flowchart . 11 4.2.2 gene2pathway test inputs/parameters . 11 4.2.3 gene2pathway test outputs . 12 5 Examples 13 5.1 ChIP-seq data analysis . 13 5.1.1 Map ChIP-seq enriched peaks to genes using runseq2gene .................... 13 5.1.2 Discover enriched GO terms using gene2pathway_test with gene scores . 15 5.1.3 Discover enriched GO terms using Fisher's Exact test without gene scores . 17 5.1.4 Add description for genes . 20 5.2 RNA-seq data analysis . 20 6 R environment session 23 1 Abstract Seq2pathway is a novel computational tool to analyze functional gene-sets (including signaling pathways) using variable next-generation sequencing data[1]. Integral to this tool are the \seq2gene" and \gene2pathway" components in series that infer a quantitative pathway-level profile for each sample. The seq2gene function assigns phenotype-associated significance of genomic regions to gene-level scores, where the significance could be p-values of SNPs or point mutations, protein-binding affinity, or transcriptional expression level. The seq2gene function has the feasibility to assign non-exon regions to a range of neighboring genes besides the nearest one, thus facilitating the study of functional non-coding elements[2]. Then the gene2pathway summarizes gene-level measurements to pathway-level scores, comparing the quantity of significance for gene members within a pathway with those outside a pathway.
    [Show full text]
  • Gene Expression Studies: from Case-Control to Multiple-Population-Based Studies
    From the Institute of Human Genetics, Helmholtz Zentrum Munchen,¨ Deutsches Forschungszentrum fur¨ Gesundheit und Umwelt (GmbH) Head: Prof. Dr. Thomas Meitinger Gene expression studies: From case-control to multiple-population-based studies Thesis Submitted for a Doctoral Degree in Natural Sciences at the Faculty of Medicine, Ludwig-Maximilians-Universitat¨ Munchen¨ Katharina Schramm Dachau, Germany 2016 With approval of the Faculty of Medicine Ludwig-Maximilians-Universit¨atM ¨unchen Supervisor/Examiner: Prof. Dr. Thomas Illig Co-Examiners: Prof. Dr. Roland Kappler Dean: Prof. Dr. med. dent. Reinhard Hickel Date of oral examination: 22.12.2016 II Dedicated to my family. III Abstract Recent technological developments allow genome-wide scans of gene expression levels. The reduction of costs and increasing parallelization of processing enable the quantification of 47,000 transcripts in up to twelve samples on a single microarray. Thereby the data collec- tion of large population-based studies was improved. During my PhD, I first developed a workflow for the statistical analyses of case-control stu- dies of up to 50 samples. With large population-based data sets generated I established a pipeline for quality control, data preprocessing and correction for confounders, which re- sulted in substantially improved data. In total, I processed more than 3,000 genome-wide expression profiles using the generated pipeline. With 993 whole blood samples from the population-based KORA (Cooperative Health Research in the Region of Augsburg) study we established one of the largest population-based resource. Using this data set we contributed to a number of transcriptome-wide association studies within national (MetaXpress) and international (CHARGE) consortia.
    [Show full text]
  • Dual Proteome-Scale Networks Reveal Cell-Specific Remodeling of the Human Interactome
    bioRxiv preprint doi: https://doi.org/10.1101/2020.01.19.905109; this version posted January 19, 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. Dual Proteome-scale Networks Reveal Cell-specific Remodeling of the Human Interactome Edward L. Huttlin1*, Raphael J. Bruckner1,3, Jose Navarrete-Perea1, Joe R. Cannon1,4, Kurt Baltier1,5, Fana Gebreab1, Melanie P. Gygi1, Alexandra Thornock1, Gabriela Zarraga1,6, Stanley Tam1,7, John Szpyt1, Alexandra Panov1, Hannah Parzen1,8, Sipei Fu1, Arvene Golbazi1, Eila Maenpaa1, Keegan Stricker1, Sanjukta Guha Thakurta1, Ramin Rad1, Joshua Pan2, David P. Nusinow1, Joao A. Paulo1, Devin K. Schweppe1, Laura Pontano Vaites1, J. Wade Harper1*, Steven P. Gygi1*# 1Department of Cell Biology, Harvard Medical School, Boston, MA, 02115, USA. 2Broad Institute, Cambridge, MA, 02142, USA. 3Present address: ICCB-Longwood Screening Facility, Harvard Medical School, Boston, MA, 02115, USA. 4Present address: Merck, West Point, PA, 19486, USA. 5Present address: IQ Proteomics, Cambridge, MA, 02139, USA. 6Present address: Vor Biopharma, Cambridge, MA, 02142, USA. 7Present address: Rubius Therapeutics, Cambridge, MA, 02139, USA. 8Present address: RPS North America, South Kingstown, RI, 02879, USA. *Correspondence: [email protected] (E.L.H.), [email protected] (J.W.H.), [email protected] (S.P.G.) #Lead Contact: [email protected] bioRxiv preprint doi: https://doi.org/10.1101/2020.01.19.905109; this version posted January 19, 2020. The copyright holder for this preprint (which was not certified by peer review) is the author/funder.
    [Show full text]
  • Exploring Prostate Cancer Genome Reveals Simultaneous Losses of PTEN, FAS and PAPSS2 in Patients with PSA Recurrence After Radical Prostatectomy
    Int. J. Mol. Sci. 2015, 16, 3856-3869; doi:10.3390/ijms16023856 OPEN ACCESS International Journal of Molecular Sciences ISSN 1422-0067 www.mdpi.com/journal/ijms Article Exploring Prostate Cancer Genome Reveals Simultaneous Losses of PTEN, FAS and PAPSS2 in Patients with PSA Recurrence after Radical Prostatectomy Chinyere Ibeawuchi 1, Hartmut Schmidt 2, Reinhard Voss 3, Ulf Titze 4, Mahmoud Abbas 5, Joerg Neumann 6, Elke Eltze 7, Agnes Marije Hoogland 8, Guido Jenster 9, Burkhard Brandt 10 and Axel Semjonow 1,* 1 Prostate Center, Department of Urology, University Hospital Muenster, Albert-Schweitzer-Campus 1, Gebaeude 1A, Muenster D-48149, Germany; E-Mail: [email protected] 2 Center for Laboratory Medicine, University Hospital Muenster, Albert-Schweitzer-Campus 1, Gebaeude 1A, Muenster D-48149, Germany; E-Mail: [email protected] 3 Interdisciplinary Center for Clinical Research, University of Muenster, Albert-Schweitzer-Campus 1, Gebaeude D3, Domagkstrasse 3, Muenster D-48149, Germany; E-Mail: [email protected] 4 Pathology, Lippe Hospital Detmold, Röntgenstrasse 18, Detmold D-32756, Germany; E-Mail: [email protected] 5 Institute of Pathology, Mathias-Spital-Rheine, Frankenburg Street 31, Rheine D-48431, Germany; E-Mail: [email protected] 6 Institute of Pathology, Klinikum Osnabrueck, Am Finkenhuegel 1, Osnabrueck D-49076, Germany; E-Mail: [email protected] 7 Institute of Pathology, Saarbrücken-Rastpfuhl, Rheinstrasse 2, Saarbrücken D-66113, Germany; E-Mail: [email protected] 8 Department
    [Show full text]
  • ADAMTSL3 As a Candidate Gene for Schizophrenia: Gene Sequencing and Ultra-High Density Association Analysis by Imputation
    Schizophrenia Research 127 (2011) 28–34 Contents lists available at ScienceDirect Schizophrenia Research journal homepage: www.elsevier.com/locate/schres ADAMTSL3 as a candidate gene for schizophrenia: Gene sequencing and ultra-high density association analysis by imputation David J. Dow a, Julie Huxley-Jones b, Jamie M. Hall a, Clyde Francks b, Peter R. Maycox a, James N.C. Kew a, Israel S. Gloger a, Nalini A.L. Mehta a, Fiona M. Kelly a, Pierandrea Muglia b, Gerome Breen c, Sarah Jugurnauth c, Inti Pederoso c, David St.Clair d, Dan Rujescu e, Michael R. Barnes b,c,⁎ a Molecular Discovery Research, GlaxoSmithKline Pharmaceuticals, New Frontiers Science Park (North), Third Avenue, Harlow, CM19 5AW, UK b Computational Biology and Genetics, Quantitative Sciences, Drug Discovery, GlaxoSmithKline Pharmaceuticals, Gunnels Wood Road, Stevenage, SG1 2NY, UK c Division of Psychological Medicine and Social, Genetic and Developmental Psychiatry Centre, Institute of Psychiatry, King's College, London, UK d Department of Mental Health, University of Aberdeen, Aberdeen, United Kingdom e Division of Molecular and Clinical Neurobiology, Department of Psychiatry, Ludwig-Maximilians-University, Munich, Germany article info abstract Article history: We previously reported an association with a putative functional variant in the ADAMTSL3 gene, just below Received 27 July 2010 genome-wide significance in a genome-wide association study of schizophrenia. As variants impacting the Received in revised form 29 November 2010 function of ADAMTSL3 (a disintegrin-like and metalloprotease domain with thrombospondin type I motifs- Accepted 11 December 2010 like-3) could illuminate a novel disease mechanism and a potentially specific target, we have used Available online 15 January 2011 complementary approaches to further evaluate the association.
    [Show full text]
  • CRAVAT 4: Cancer-Related Analysis of Variants Toolkit David L
    Cancer Focus on Computer Resources Research CRAVAT 4: Cancer-Related Analysis of Variants Toolkit David L. Masica1,2, Christopher Douville1,2, Collin Tokheim1,2, Rohit Bhattacharya2,3, RyangGuk Kim4, Kyle Moad4, Michael C. Ryan4, and Rachel Karchin1,2,5 Abstract Cancer sequencing studies are increasingly comprehensive and level interpretation, including joint prioritization of all nonsilent well powered, returning long lists of somatic mutations that can be mutation consequence types, and structural and mechanistic visu- difficult to sort and interpret. Diligent analysis and quality control alization. Results from CRAVAT submissions are explored in an can require multiple computational tools of distinct utility and interactive, user-friendly web environment with dynamic filtering producing disparate output, creating additional challenges for the and sorting designed to highlight the most informative mutations, investigator. The Cancer-Related Analysis of Variants Toolkit (CRA- even in the context of very large studies. CRAVAT can be run on a VAT) is an evolving suite of informatics tools for mutation inter- public web portal, in the cloud, or downloaded for local use, and is pretation that includes mutation mapping and quality control, easily integrated with other methods for cancer omics analysis. impact prediction and extensive annotation, gene- and mutation- Cancer Res; 77(21); e35–38. Ó2017 AACR. Background quickly returning mutation interpretations in an interactive and user-friendly web environment for sorting, visualizing, and An investigator's work is far from over when results are returned inferring mechanism. CRAVAT (see Supplementary Video) is from the sequencing center. Depending on the service, genetic suitable for large studies (e.g., full-exome and large cohorts) mutation calls can require additional mapping to include all and small studies (e.g., gene panel or single patient), performs relevant RNA transcripts or correct protein sequences.
    [Show full text]
  • Essential Genes and Their Role in Autism Spectrum Disorder
    University of Pennsylvania ScholarlyCommons Publicly Accessible Penn Dissertations 2017 Essential Genes And Their Role In Autism Spectrum Disorder Xiao Ji University of Pennsylvania, [email protected] Follow this and additional works at: https://repository.upenn.edu/edissertations Part of the Bioinformatics Commons, and the Genetics Commons Recommended Citation Ji, Xiao, "Essential Genes And Their Role In Autism Spectrum Disorder" (2017). Publicly Accessible Penn Dissertations. 2369. https://repository.upenn.edu/edissertations/2369 This paper is posted at ScholarlyCommons. https://repository.upenn.edu/edissertations/2369 For more information, please contact [email protected]. Essential Genes And Their Role In Autism Spectrum Disorder Abstract Essential genes (EGs) play central roles in fundamental cellular processes and are required for the survival of an organism. EGs are enriched for human disease genes and are under strong purifying selection. This intolerance to deleterious mutations, commonly observed haploinsufficiency and the importance of EGs in pre- and postnatal development suggests a possible cumulative effect of deleterious variants in EGs on complex neurodevelopmental disorders. Autism spectrum disorder (ASD) is a heterogeneous, highly heritable neurodevelopmental syndrome characterized by impaired social interaction, communication and repetitive behavior. More and more genetic evidence points to a polygenic model of ASD and it is estimated that hundreds of genes contribute to ASD. The central question addressed in this dissertation is whether genes with a strong effect on survival and fitness (i.e. EGs) play a specific oler in ASD risk. I compiled a comprehensive catalog of 3,915 mammalian EGs by combining human orthologs of lethal genes in knockout mice and genes responsible for cell-based essentiality.
    [Show full text]