Article Reference

Total Page:16

File Type:pdf, Size:1020Kb

Article Reference Article Comprehensive evaluation of coding region point mutations in microsatellite‐unstable colorectal cancer KONDELIN, Johanna, et al. Reference KONDELIN, Johanna, et al. Comprehensive evaluation of coding region point mutations in microsatellite‐unstable colorectal cancer. EMBO Molecular Medicine, 2018, vol. 10, no. 9, p. e8552 PMID : 30108113 DOI : 10.15252/emmm.201708552 Available at: http://archive-ouverte.unige.ch/unige:120174 Disclaimer: layout of this document may differ from the published version. 1 / 1 Published online: August 14, 2018 Report Comprehensive evaluation of coding region point mutations in microsatellite-unstable colorectal cancer Johanna Kondelin1,2, Kari Salokas3,4, Lilli Saarinen2, Kristian Ovaska2, Heli Rauanheimo1,2, Roosa-Maria Plaketti1,2, Jiri Hamberg1,2, Xiaonan Liu3,4 , Leena Yadav3,4, Alexandra E Gylfe1,2, Tatiana Cajuso1,2, Ulrika A Hänninen1,2, Kimmo Palin1,2, Heikki Ristolainen1,2, Riku Katainen1,2, Eevi Kaasinen1,2, Tomas Tanskanen1,2, Mervi Aavikko1,2, Minna Taipale5, Jussi Taipale1,2,6,7 , Laura Renkonen-Sinisalo8, Anna Lepistö8, Selja Koskensalo9, Jan Böhm10, Jukka-Pekka Mecklin11,12, Halit Ongen13,14,15, Emmanouil T Dermitzakis13,14,15, Outi Kilpivaara1,2, Pia Vahteristo1,2, Mikko Turunen2, Sampsa Hautaniemi2, Sari Tuupanen1,2, Auli Karhu1,2, Niko Välimäki1,2, Markku Varjosalo3,4, Esa Pitkänen1,2 & Lauri A Aaltonen1,2,* Abstract not been linked to CRC before. Two genes, SMARCB1 and STK38L, were also functionally scrutinized, providing evidence of Microsatellite instability (MSI) leads to accumulation of an a tumorigenic role, for SMARCB1 mutations in particular. excessive number of mutations in the genome, mostly small insertions and deletions. MSI colorectal cancers (CRCs), however, Keywords cancer genetics; colorectal cancer; microsatellite instability also contain more point mutations than microsatellite-stable Subject Categories Cancer; Chromatin, Epigenetics, Genomics & Functional (MSS) tumors, yet they have not been as comprehensively stud- Genomics; Systems Medicine ied. To identify candidate driver genes affected by point muta- DOI 10.15252/emmm.201708552 | Received 5 October 2017 | Revised 6 July tions in MSI CRC, we ranked genes based on mutation 2018 | Accepted 9 July 2018 | Published online 14 August 2018 significance while correcting for replication timing and gene EMBO Mol Med (2018) 10:e8552 expression utilizing an algorithm, MutSigCV. Somatic point mutation data from the exome kit-targeted area from 24 exome-sequenced sporadic MSI CRCs and respective normals, Introduction and 12 whole-genome-sequenced sporadic MSI CRCs and respective normals were utilized. The top 73 genes were vali- Colorectal cancer (CRC) is one of the most fatal cancers in Western dated in 93 additional MSI CRCs. The MutSigCV ranking identi- countries leading to death in nearly 50% of the cases (Jemal et al, fied several well-established MSI CRC driver genes and provided 2011). Approximately 15% of CRCs exhibit microsatellite instability additional evidence for previously proposed CRC candidate (MSI), which results from defective DNA mismatch repair (MMR) genes as well as shortlisted genes that have to our knowledge machinery (Boland & Goel, 2010). This is most often the result of 1 Medicum/Department of Medical and Clinical Genetics, University of Helsinki, Helsinki, Finland 2 Genome-Scale Biology Research Program, Research Programs Unit, University of Helsinki, Helsinki, Finland 3 Institute of Biotechnology, University of Helsinki, Helsinki, Finland 4 Helsinki Institute of Life Science, University of Helsinki, Helsinki, Finland 5 Division of Functional Genomics, Department of Medical Biochemistry and Biophysics (MBB), Karolinska Institutet, Stockholm, Sweden 6 Department of Biosciences and Nutrition, Karolinska Institutet, Huddinge, Sweden 7 Science for Life Center, Huddinge, Sweden 8 Department of Surgery, Helsinki University Central Hospital, Hospital District of Helsinki and Uusimaa, Helsinki, Finland 9 The HUCH Gastrointestinal Clinic, Helsinki University Central Hospital, Helsinki, Finland 10 Department of Pathology, Jyväskylä Central Hospital, Jyväskylä, Finland 11 Department of Surgery, Jyväskylä Central Hospital, University of Eastern Finland, Jyväskylä, Finland 12 Department Sport and Health Sciences, University of Jyväskylä, Jyväskylä, Finland 13 Department of Genetic Medicine and Development, University of Geneva Medical School, Geneva, Switzerland 14 Institute for Genetics and Genomics in Geneva (iGE3), University of Geneva, Geneva, Switzerland 15 Swiss Institute of Bioinformatics, Geneva, Switzerland *Corresponding author. Tel: +358 2941 25595; E-mail: [email protected] ª 2018 The Authors. Published under the terms of the CC BY 4.0 license EMBO Molecular Medicine 10:e8552 | 2018 1 of 20 Published online: August 14, 2018 EMBO Molecular Medicine Candidate genes based on point mutations Johanna Kondelin et al hypermethylation of the promoter of MLH1, one of the central genes To continue on our previous studies where candidate MSI CRC involved in MMR. oncogenes were identified based on mutation hot spots in a smaller It is estimated that approximately 90% of CRCs are sporadic, dataset (Gylfe et al, 2013; Tuupanen et al, 2014), we repeated the whereas the remaining 10% arise due to inherited predisposition hot spot analysis in this dataset of 36 exome- or whole-genome- (Bogaert & Prenen, 2014). The most common form of inherited sequenced sporadic MSI CRCs and corresponding normals. Hence, predisposition is Lynch syndrome, where the individual inherits a genes containing mutation hot spots in these somatic point mutation germline mutation in one of the MMR genes (MLH1, MSH2, MSH6, data were detected (Figs 1 and 2). From this set of hot spots, the 90 PMS2) and is therefore highly predisposed to CRC and endometrial novel hot spots as well as seven previously studied hot spots were cancer (Boland & Goel, 2010). In addition to CRC, MSI is also re-sequenced in the validation set of 93 additional MSI CRCs. From observed in approximately 15% of sporadic endometrial and gastric this effort, seven new candidate oncogenes emerged (CORIN, cancers (Hamelin et al, 2008). MSI CRCs arise through a distinct KLHL6, PCDHB16, PLEKHG1, PROS1, SPP2, and TROAP). To our genetic pathway as compared to microsatellite-stable (MSS) CRCs knowledge, this study represents the first effort to uncover driver (Boland & Goel, 2010). The defective MMR machinery results in the point mutations in MSI CRC utilizing deep sequencing of a large set accumulation of an excessive number of mutations in the genome. of tumors for validation. Most of the mutations are small insertions and deletions (indels) that target short nucleotide repeats, microsatellites. Genes that provide growth advantage to cells via loss-of-function mutations in Results microsatellites, or MSI target genes (Duval & Hamelin, 2002), have been extensively studied and numerous genes have been published In order to identify new candidates for driver genes affected by point as candidate targets and thus putative tumor suppressors (Alhopuro mutations in MSI CRC, we analyzed sequencing data from a discov- et al, 2012; Kondelin et al, 2017). MSI tumors also contain an order ery set of 36 exome- or whole-genome-sequenced MSI CRCs and of magnitude more point mutations than MSS tumors (Boland & respective normals. MutSigCV analysis was performed on the Goel, 2010), yet to date the point mutations in MSI CRCs have been somatic single-nucleotide variation (SNV) data to identify the genes mostly overlooked. Only few genes with causative point mutations most likely to display an excess of point mutations due to selection, have been identified in this tumor type. Most of these have been and the resulting top 73 genes were further validated by MiSeq flagged by missense mutation hot spots (e.g., BRAF, KRAS, CTNNB1, sequencing in a validation set of 93 additional MSI CRCs. Next, a and PIK3CA), a mutation pattern typical of oncogenes (Fearon, new algorithm, OncodriveFML, had become available during the 2011). study and was utilized on the somatic SNV data from the MiSeq In our past efforts, we have identified candidate oncogenes sequencing to identify the most likely candidates for previously with missense mutation hot spots based on next-generation unknown CRC-driving genes. Of these, SMARCB1 and STK38L were sequencing (NGS) data from a small discovery set of MSI CRCs further validated in functional experiments. The analysis workflow (Gylfe et al, 2013; Tuupanen et al, 2014). To our knowledge, is summarized in Fig 1. however, only few studies have attempted to systematically char- In addition, to continue on our previous efforts (Gylfe et al, acterize the full landscape of coding point mutations in MSI CRC 2013; Tuupanen et al, 2014), we performed an analysis on genes in order to identify new driver genes (Cancer Genome Atlas containing somatic mutation hot spots—mutations residing in either Network 2012; Seshagiri et al, 2012; Kim et al, 2013; Cortes- the same or two adjacent codons, or two bases flanking an exon– Ciriano et al, 2017). intron boundary—in at least two samples. Ninety-seven hot spots In the past few years, NGS has been largely accepted into both from 94 genes were selected for further validation in the set of 93 research and clinical use, and numerous mutations—both somatic additional MSI CRCs. The analysis workflow is summarized in and germline—have been reported to contribute to disease. There Fig 2. is, however, debate on which of the genes and
Recommended publications
  • Profiling Data
    Compound Name DiscoveRx Gene Symbol Entrez Gene Percent Compound Symbol Control Concentration (nM) JNK-IN-8 AAK1 AAK1 69 1000 JNK-IN-8 ABL1(E255K)-phosphorylated ABL1 100 1000 JNK-IN-8 ABL1(F317I)-nonphosphorylated ABL1 87 1000 JNK-IN-8 ABL1(F317I)-phosphorylated ABL1 100 1000 JNK-IN-8 ABL1(F317L)-nonphosphorylated ABL1 65 1000 JNK-IN-8 ABL1(F317L)-phosphorylated ABL1 61 1000 JNK-IN-8 ABL1(H396P)-nonphosphorylated ABL1 42 1000 JNK-IN-8 ABL1(H396P)-phosphorylated ABL1 60 1000 JNK-IN-8 ABL1(M351T)-phosphorylated ABL1 81 1000 JNK-IN-8 ABL1(Q252H)-nonphosphorylated ABL1 100 1000 JNK-IN-8 ABL1(Q252H)-phosphorylated ABL1 56 1000 JNK-IN-8 ABL1(T315I)-nonphosphorylated ABL1 100 1000 JNK-IN-8 ABL1(T315I)-phosphorylated ABL1 92 1000 JNK-IN-8 ABL1(Y253F)-phosphorylated ABL1 71 1000 JNK-IN-8 ABL1-nonphosphorylated ABL1 97 1000 JNK-IN-8 ABL1-phosphorylated ABL1 100 1000 JNK-IN-8 ABL2 ABL2 97 1000 JNK-IN-8 ACVR1 ACVR1 100 1000 JNK-IN-8 ACVR1B ACVR1B 88 1000 JNK-IN-8 ACVR2A ACVR2A 100 1000 JNK-IN-8 ACVR2B ACVR2B 100 1000 JNK-IN-8 ACVRL1 ACVRL1 96 1000 JNK-IN-8 ADCK3 CABC1 100 1000 JNK-IN-8 ADCK4 ADCK4 93 1000 JNK-IN-8 AKT1 AKT1 100 1000 JNK-IN-8 AKT2 AKT2 100 1000 JNK-IN-8 AKT3 AKT3 100 1000 JNK-IN-8 ALK ALK 85 1000 JNK-IN-8 AMPK-alpha1 PRKAA1 100 1000 JNK-IN-8 AMPK-alpha2 PRKAA2 84 1000 JNK-IN-8 ANKK1 ANKK1 75 1000 JNK-IN-8 ARK5 NUAK1 100 1000 JNK-IN-8 ASK1 MAP3K5 100 1000 JNK-IN-8 ASK2 MAP3K6 93 1000 JNK-IN-8 AURKA AURKA 100 1000 JNK-IN-8 AURKA AURKA 84 1000 JNK-IN-8 AURKB AURKB 83 1000 JNK-IN-8 AURKB AURKB 96 1000 JNK-IN-8 AURKC AURKC 95 1000 JNK-IN-8
    [Show full text]
  • Application of a MYC Degradation
    SCIENCE SIGNALING | RESEARCH ARTICLE CANCER Copyright © 2019 The Authors, some rights reserved; Application of a MYC degradation screen identifies exclusive licensee American Association sensitivity to CDK9 inhibitors in KRAS-mutant for the Advancement of Science. No claim pancreatic cancer to original U.S. Devon R. Blake1, Angelina V. Vaseva2, Richard G. Hodge2, McKenzie P. Kline3, Thomas S. K. Gilbert1,4, Government Works Vikas Tyagi5, Daowei Huang5, Gabrielle C. Whiten5, Jacob E. Larson5, Xiaodong Wang2,5, Kenneth H. Pearce5, Laura E. Herring1,4, Lee M. Graves1,2,4, Stephen V. Frye2,5, Michael J. Emanuele1,2, Adrienne D. Cox1,2,6, Channing J. Der1,2* Stabilization of the MYC oncoprotein by KRAS signaling critically promotes the growth of pancreatic ductal adeno- carcinoma (PDAC). Thus, understanding how MYC protein stability is regulated may lead to effective therapies. Here, we used a previously developed, flow cytometry–based assay that screened a library of >800 protein kinase inhibitors and identified compounds that promoted either the stability or degradation of MYC in a KRAS-mutant PDAC cell line. We validated compounds that stabilized or destabilized MYC and then focused on one compound, Downloaded from UNC10112785, that induced the substantial loss of MYC protein in both two-dimensional (2D) and 3D cell cultures. We determined that this compound is a potent CDK9 inhibitor with a previously uncharacterized scaffold, caused MYC loss through both transcriptional and posttranslational mechanisms, and suppresses PDAC anchorage- dependent and anchorage-independent growth. We discovered that CDK9 enhanced MYC protein stability 62 through a previously unknown, KRAS-independent mechanism involving direct phosphorylation of MYC at Ser .
    [Show full text]
  • Genome-Wide DNA Methylation Analysis of KRAS Mutant Cell Lines Ben Yi Tew1,5, Joel K
    www.nature.com/scientificreports OPEN Genome-wide DNA methylation analysis of KRAS mutant cell lines Ben Yi Tew1,5, Joel K. Durand2,5, Kirsten L. Bryant2, Tikvah K. Hayes2, Sen Peng3, Nhan L. Tran4, Gerald C. Gooden1, David N. Buckley1, Channing J. Der2, Albert S. Baldwin2 ✉ & Bodour Salhia1 ✉ Oncogenic RAS mutations are associated with DNA methylation changes that alter gene expression to drive cancer. Recent studies suggest that DNA methylation changes may be stochastic in nature, while other groups propose distinct signaling pathways responsible for aberrant methylation. Better understanding of DNA methylation events associated with oncogenic KRAS expression could enhance therapeutic approaches. Here we analyzed the basal CpG methylation of 11 KRAS-mutant and dependent pancreatic cancer cell lines and observed strikingly similar methylation patterns. KRAS knockdown resulted in unique methylation changes with limited overlap between each cell line. In KRAS-mutant Pa16C pancreatic cancer cells, while KRAS knockdown resulted in over 8,000 diferentially methylated (DM) CpGs, treatment with the ERK1/2-selective inhibitor SCH772984 showed less than 40 DM CpGs, suggesting that ERK is not a broadly active driver of KRAS-associated DNA methylation. KRAS G12V overexpression in an isogenic lung model reveals >50,600 DM CpGs compared to non-transformed controls. In lung and pancreatic cells, gene ontology analyses of DM promoters show an enrichment for genes involved in diferentiation and development. Taken all together, KRAS-mediated DNA methylation are stochastic and independent of canonical downstream efector signaling. These epigenetically altered genes associated with KRAS expression could represent potential therapeutic targets in KRAS-driven cancer. Activating KRAS mutations can be found in nearly 25 percent of all cancers1.
    [Show full text]
  • Detection of Genetic Variations of Five MMAF-Related Genes and Their
    Detection of genetic variations of ve MMAF-related genes and their associations with litter size in goat Zhen Wang Northwest Agriculture and Forestry University Yun Pan Northwest Agriculture and Forestry University Libang He Northwest Agriculture and Forestry University Hong Chen Northwest Agriculture and Forestry University Chuanying Pan Northwest Agriculture and Forestry University Lei Qu Yulin University Xiaoyue Song Yulin University Xianyong Lan ( [email protected] ) https://orcid.org/0000-0003-2254-5805 Research article Keywords: goats; insertion/deletion (indel); MMAF; DNAH1; litter sizes; correlation analysis Posted Date: September 12th, 2019 DOI: https://doi.org/10.21203/rs.2.14419/v1 License: This work is licensed under a Creative Commons Attribution 4.0 International License. Read Full License Page 1/13 Abstract Background: Multiple morphological abnormalities of the sperm agella (MMAF) makes an assignable contribution to male infertility, including QRICH2 , CFAP43 , CFAP44 , CFAP69 , CCDC39 , AKAP4 and DNAH1 gene. This work studied 28 putative indel mutations of MMAF related genes including QRICH2 , CFAP69 , CFAP43 , CCDC39 and DNAH1 gene and their correlation with the rst-born litter sizes of 769 Shaanbei white cashmere (SBWC) goats. Results: Electrophoresis and DNA sequencing analysis showed the 11-bp indel within QRICH2 ( QRICH2 -P4), the three indel variations in CFAP69 ( CFAP69 -P4, CFAP69 - P6 and CFAP69 -P7) and the 27-bp indel of DNAH1 ( DNAH1 -P1) were found to be polymorphic. The 27- bp indel variation within DNAH1 was not in consistent with HWE and the other four indel of QRICH2 and CFAP69 were in consistent with HWE. The linkage disequilibrium (LD) analysis showed the 8-bp indel ( CFAP69 -P4) and the 6-bp indel ( CFAP69- P6) within CFAP69 were in complete LD with each other (D'=0.99, r 2 =1.00).
    [Show full text]
  • HEY1 Functions Are Regulated by Its Phosphorylation at Ser-68
    Biosci. Rep. (2016) / 36 / art:e00343 / doi 10.1042/BSR20160123 HEY1 functions are regulated by its phosphorylation at Ser-68 Irene Lopez-Mateo*,´ Amaia Arruabarrena-Aristorena†, Cristina Artaza-Irigaray‡, Juan A. Lopez´ §, Enrique Calvo§ and Borja Belandia*1 *Department of Cancer Biology, Instituto de Investigaciones Biomedicas´ Alberto Sols, CSIC-UAM, Arturo Duperier 4, 28029 Madrid, Spain †Proteomics Unit, CIC bioGUNE, Bizkaia Technology Park, 48160 Derio, Spain ‡Division´ de Inmunolog´ıa, Centro de Investigacion´ Biomedica´ de Occidente – IMSS, Sierra Mojada 800, 44340 Guadalajara, Jalisco, Mexico §Unidad de Proteomica, Centro Nacional de Investigaciones Cardiovasculares, CNIC, 28029 Madrid, Spain Synopsis HEY1 (hairy/enhancer-of-split related with YRPW motif 1) is a member of the basic helix–loop–helix-orange (bHLH-O) family of transcription repressors that mediate Notch signalling. HEY1 acts as a positive regulator of the tumour suppressor p53 via still unknown mechanisms. A MALDI-TOF/TOF MS analysis has uncovered a novel HEY1 regulatory phosphorylation event at Ser-68. Strikingly, this single phosphorylation event controls HEY1 stability and function: simulation of HEY1 Ser-68 phosphorylation increases HEY1 protein stability but inhibits its ability to enhance p53 transcriptional activity. Unlike wild-type HEY1, expression of the phosphomimetic mutant HEY1-S68D failed to induce p53-dependent cell cycle arrest and it did not sensitize U2OS cells to p53-activating chemotherapeutic drugs. We have identified two related kinases, STK38 (serine/threonine kinase 38) and STK38L (serine/threonine kinase 38 like), which interact with and phosphorylate HEY1 at Ser-68. HEY1 is phosphorylated at Ser-68 during mitosis and it accumulates in the centrosomes of mitotic cells, suggesting a possible integration of HEY1-dependent signalling in centrosome function.
    [Show full text]
  • Photoreceptor Proliferation and Dysregulation of Cell Cycle Genes in Early Onset Inherited Retinal Degenerations
    University of Pennsylvania ScholarlyCommons Departmental Papers (Vet) School of Veterinary Medicine 3-11-2016 Photoreceptor Proliferation and Dysregulation of Cell Cycle Genes in Early Onset Inherited Retinal Degenerations Kristin L. Gardiner University of Pennsylvania Louise Downs University of Pennsylvania Agnes I. Berta-Antalics University of Pennsylvania Evelyn Santana University of Pennsylvania Gustavo D. Aguirre University of Pennsylvania, [email protected] See next page for additional authors Follow this and additional works at: https://repository.upenn.edu/vet_papers Part of the Veterinary Medicine Commons Recommended Citation Gardiner, K. L., Downs, L., Berta-Antalics, A. I., Santana, E., Aguirre, G. D., & Genini, S. (2016). Photoreceptor Proliferation and Dysregulation of Cell Cycle Genes in Early Onset Inherited Retinal Degenerations. BMC Genomics, 17 (221), http://dx.doi.org/10.1186/s12864-016-2477-9 This paper is posted at ScholarlyCommons. https://repository.upenn.edu/vet_papers/152 For more information, please contact [email protected]. Photoreceptor Proliferation and Dysregulation of Cell Cycle Genes in Early Onset Inherited Retinal Degenerations Abstract Background Mitotic terminally differentiated photoreceptors (PRs) are observed in early retinal degeneration (erd), an inherited canine retinal disease driven by mutations in the NDR kinase STK38L (NDR2). Results We demonstrate that a similar proliferative response, but of lower magnitude, occurs in two other early onset disease models, X-linked progressive retinal atrophy 2 (xlpra2) and rod cone dysplasia 1 (rcd1). Proliferating cells are rod PRs, and not microglia or Müller cells. Expression of the cell cycle related genes RB1 and E2F1 as well as CDK2,4,6 was up-regulated, but changes were mutation-specific.
    [Show full text]
  • Insights Into MYC Biology Through Investigation of Synthetic Lethal Interactions with MYC Deregulation
    Insights into MYC biology through investigation of synthetic lethal interactions with MYC deregulation Mai Sato Submitted in partial fulfillment of the requirements for the degree of Doctor of Philosophy under the Executive Committee of the Graduate School of Arts and Sciences COLUMBIA UNIVERSITY 2014 © 2014 Mai Sato All Rights Reserved ABSTRACT Insights into MYC biology through investigation of synthetic lethal interactions with MYC deregulation Mai Sato MYC (or c-myc) is a bona fide “cancer driver” oncogene that is deregulated in up to 70% of human tumors. In addition to its well-characterized role as a transcription factor that can directly promote tumorigenic growth and proliferation, MYC has transcription-independent functions in vital cellular processes including DNA replication and protein synthesis, contributing to its complex biology. MYC expression, activity, and stability are highly regulated through multiple mechanisms. MYC deregulation triggers genome instability and oncogene-induced DNA replication stress, which are thought to be critical in promoting cancer via mechanisms that are still unclear. Because regulated MYC activity is essential for normal cell viability and MYC is a difficult protein to target pharmacologically, targeting genes or pathways that are essential to survive MYC deregulation offer an attractive alternative as a means to combat tumor cells with MYC deregulation. To this end, we conducted a genome-wide synthetic lethal shRNA screen in MCF10A breast epithelial cells stably expressing an inducible MYCER transgene. We identified and validated FBXW7 as a high-confidence synthetic lethal (MYC-SL) candidate gene. FBXW7 is a component of an E3 ubiquitin ligase complex that degrades MYC. FBXW7 knockdown in MCF10A cells selectively induced cell death in MYC-deregulated cells compared to control.
    [Show full text]
  • A Yeast Bifc-Seq Method for Genome-Wide Interactome Mapping
    bioRxiv preprint doi: https://doi.org/10.1101/2020.06.16.154146; this version posted June 17, 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. 1 A yeast BiFC-seq method for genome-wide interactome mapping 2 3 Limin Shang1,a, Yuehui Zhang1,b, Yuchen Liu1,c, Chaozhi Jin1,d, Yanzhi Yuan1,e, 4 Chunyan Tian1,f, Ming Ni2,g, Xiaochen Bo2,h, Li Zhang3,i, Dong Li1,j, Fuchu He1,*,k & 5 Jian Wang1,*,l 6 1State Key Laboratory of Proteomics, Beijing Proteome Research Center, National 7 Center for Protein Sciences (Beijing), Beijing Institute of Lifeomics, Beijing 102206, 8 China; 2Department of Biotechnology, Beijing Institute of Radiation Medicine, 9 Beijing 100850, China; 3Department of Rehabilitation Medicine, Nan Lou; 10 Department of Key Laboratory of Wound Repair and Regeneration of PLA, College 11 of Life Sciences, Chinese PLA General Hospital, Beijing 100853, China; 12 Correspondence should be addressed to F.H. ([email protected]) and J.W. 13 ([email protected]). 14 * Corresponding authors. 15 E-mail:[email protected] (Fuchu He),[email protected] (Jian Wang) 16 a ORCID: 0000-0002-6371-1956. 17 b ORCID: 0000-0001-5257-1671 18 c ORCID: 0000-0003-4691-4951 19 d ORCID: 0000-0002-1477-0255 20 e ORCID: 0000-0002-6576-8112 21 f ORCID: 0000-0003-1589-293X 22 g ORCID: 0000-0001-9465-2787 23 h ORCID: 0000-0003-3490-5812 24 i ORCID: 0000-0002-3477-8860 25 j ORCID: 0000-0002-8680-0468 26 k ORCID: 0000-0002-8571-2351 27 l ORCID: 0000-0002-8116-7691 28 Total word counts:4398 (from “Introduction” to “Conclusions”) 29 Total Keywords: 5 bioRxiv preprint doi: https://doi.org/10.1101/2020.06.16.154146; this version posted June 17, 2020.
    [Show full text]
  • Characterization and Expression Profile of CMTM3/CKLFSF3
    Journal of Biochemistry and Molecular Biology, Vol. 39, No. 5, September 2006, pp. 537-545 Characterization and Expression Profile of CMTM3/CKLFSF3 Ji Zhong, Yu Wang, Xiaoyan Qiu, Xiaoning Mo, Yanan Liu, Ting Li, Quansheng Song, Dalong Ma and Wenling Han* Peking University Center for Human Disease Genomics, 38 Xueyuan Road, Beijing 100083, China Received 22 February 2006, Accepted 10 May 2006 CMTM/CKLFSF is a novel family of proteins linking Introduction chemokines and TM4SF. In humans, these proteins are encoded by nine genes, CKLF and CMTM1-8/CKLFSF1-8. CKLFSF is a novel family of proteins linking chemokines and Here we report the characteristics and expression profile Transmembrane 4 Super Family (TM4SF), which are encoded of CMTM3/CKLFSF3. Human CMTM3/CKLFSF3 has a by nine genes, CKLF and CKLFSF1-8, in humans. CKLF, high sequence identity among various species and similar CKLFSF1, and CKLFSF2 are active during evolution, and characteristics as its mouse and rat homologues. Established CKLFSF2 has two orthologues in rat and mouse designated by results both of RT-PCR and Quantitative Real-time Cklfsf2a and Cklfsf2b. CKLFSF3-8 are evolutionarily conserved. PCR, the gene is highly transcribed in testis, leukocytes CKLFSF members form two gene clusters: CKLF and and spleen. For further verification, we generated a CKLFSF1-4 at chromosome 16q22.1, and CKLFSF6-8 at polyclonal antibody against human CMTM3/CKLFSF3 chromosome 3p23. Most CKLFSFs have different spliced and found that the protein is highly expressed in the testis forms, and the protein product of at least one spliced form of and some cells of PBMCs.
    [Show full text]
  • RNA-Seq Transcriptome Analysis for the Study of Prostate Cancer Development and Evolution
    MASTER THESIS RNA-Seq Transcriptome Analysis for the Study of Prostate Cancer Development and Evolution Esther Sauras Colón Tutors: María Jesús Álvarez Cubero, Eduardo Andrés León September 2019 MASTER IN TRANSLATIONAL RESEARCH AND PERSONALIZED MEDICINE Master Thesis RNA-Seq Transcriptome Analysis for the Study of Prostate Cancer Development and Evolution Esther Sauras Colón 1*, María Jesús Álvarez Cubero 2,3* and Eduardo Andrés León 4* 1 Master in Translational Research and Personalized Medicine. Faculty of Medicine, University of Granada. Granada, Spain. 2 GENYO. Centre for Genomics and Oncological Research. Pfizer / University of Granada / Andalusian Regional Government. Granada, Spain. 3 Department of Biochemistry and Molecular Biology III. Faculty of Medicine, University of Granada. Granada, Spain. 4 Bioinformatics Unit, Institute of Parasitology and Biomedicine “López-Neyra” (IPBLN). Spanish National Research Council (CSIC). Granada, Spain. * Correspondence: Esther Sauras Colón: [email protected]. María Jesús Álvarez Cubero: [email protected]. Eduardo Andrés León: [email protected]. September 2019 Abstract: Prostate cancer (PCa) is one of the most common cancers worldwide. Even though prostate specific antigen (PSA) test is the non-invasive routine blood test for the detection of asymptomatic disease, it can result in problems of diagnostic accuracy and overdiagnosis. Thus, it is necessary to continue investigating new efficient biomarkers for the prevention, diagnosis and prognosis of PCa. Here, we analyse the transcriptome of seven individuals by the use of next-generation sequencing (NGS) technique to identify differentially expressed genes, which can help to better understand PCa aggressiveness. Present analysis show that there are two upregulated genes in PCa regarding to controls: HP (Haptoglobin) and HLA-G (Human Leukocyte Antigen-G).
    [Show full text]
  • Human Social Genomics in the Multi-Ethnic Study of Atherosclerosis
    Getting “Under the Skin”: Human Social Genomics in the Multi-Ethnic Study of Atherosclerosis by Kristen Monét Brown A dissertation submitted in partial fulfillment of the requirements for the degree of Doctor of Philosophy (Epidemiological Science) in the University of Michigan 2017 Doctoral Committee: Professor Ana V. Diez-Roux, Co-Chair, Drexel University Professor Sharon R. Kardia, Co-Chair Professor Bhramar Mukherjee Assistant Professor Belinda Needham Assistant Professor Jennifer A. Smith © Kristen Monét Brown, 2017 [email protected] ORCID iD: 0000-0002-9955-0568 Dedication I dedicate this dissertation to my grandmother, Gertrude Delores Hampton. Nanny, no one wanted to see me become “Dr. Brown” more than you. I know that you are standing over the bannister of heaven smiling and beaming with pride. I love you more than my words could ever fully express. ii Acknowledgements First, I give honor to God, who is the head of my life. Truly, without Him, none of this would be possible. Countless times throughout this doctoral journey I have relied my favorite scripture, “And we know that all things work together for good, to them that love God, to them who are called according to His purpose (Romans 8:28).” Secondly, I acknowledge my parents, James and Marilyn Brown. From an early age, you two instilled in me the value of education and have been my biggest cheerleaders throughout my entire life. I thank you for your unconditional love, encouragement, sacrifices, and support. I would not be here today without you. I truly thank God that out of the all of the people in the world that He could have chosen to be my parents, that He chose the two of you.
    [Show full text]
  • PRODUCTS and SERVICES Target List
    PRODUCTS AND SERVICES Target list Kinase Products P.1-11 Kinase Products Biochemical Assays P.12 "QuickScout Screening Assist™ Kits" Kinase Protein Assay Kits P.13 "QuickScout Custom Profiling & Panel Profiling Series" Targets P.14 "QuickScout Custom Profiling Series" Preincubation Targets Cell-Based Assays P.15 NanoBRET™ TE Intracellular Kinase Cell-Based Assay Service Targets P.16 Tyrosine Kinase Ba/F3 Cell-Based Assay Service Targets P.17 Kinase HEK293 Cell-Based Assay Service ~ClariCELL™ ~ Targets P.18 Detection of Protein-Protein Interactions ~ProbeX™~ Stable Cell Lines Crystallization Services P.19 FastLane™ Structures ~Premium~ P.20-21 FastLane™ Structures ~Standard~ Kinase Products For details of products, please see "PRODUCTS AND SERVICES" on page 1~3. Tyrosine Kinases Note: Please contact us for availability or further information. Information may be changed without notice. Expression Protein Kinase Tag Carna Product Name Catalog No. Construct Sequence Accession Number Tag Location System HIS ABL(ABL1) 08-001 Full-length 2-1130 NP_005148.2 N-terminal His Insect (sf21) ABL(ABL1) BTN BTN-ABL(ABL1) 08-401-20N Full-length 2-1130 NP_005148.2 N-terminal DYKDDDDK Insect (sf21) ABL(ABL1) [E255K] HIS ABL(ABL1)[E255K] 08-094 Full-length 2-1130 NP_005148.2 N-terminal His Insect (sf21) HIS ABL(ABL1)[T315I] 08-093 Full-length 2-1130 NP_005148.2 N-terminal His Insect (sf21) ABL(ABL1) [T315I] BTN BTN-ABL(ABL1)[T315I] 08-493-20N Full-length 2-1130 NP_005148.2 N-terminal DYKDDDDK Insect (sf21) ACK(TNK2) GST ACK(TNK2) 08-196 Catalytic domain
    [Show full text]