Supplemental Information DYRK1A Regulates the Recruitment of 53BP1 to the Sites of DNA Damage in Part Through Interaction with R

Total Page:16

File Type:pdf, Size:1020Kb

Supplemental Information DYRK1A Regulates the Recruitment of 53BP1 to the Sites of DNA Damage in Part Through Interaction with R Supplemental Information DYRK1A regulates the recruitment of 53BP1 to the sites of DNA damage in part through interaction with RNF169. Vijay R. Menon, Varsha Ananthapadmanabhan, Selene Swanson, Siddharth Saini, Fatmata Sesay, Vasily Yakovlev, Laurence Florens, James A. DeCaprio, Michael P. Washburn, Mikhail Dozmorov, Larisa Litovchick Supplemental Figure Legends Figure S1. Validation the DYRK1A interactions detected by MudPIT. A. Comparison of DYRK1A interacting proteins detected in HEK293T cells (Varjosalo et al., 2013) and T98G cells (this paper). Asterisk indicates the proteins known to interact with adenovirus E1A according to the UniProt database (http://www.uniprot.org/uniprot/P03255#interaction). DYRK1A binds E1A indirectly through DCAF7 (Glenewinkel et al., 2016). B. T98G cells stably expressing Flag-HA tagged DCAF7, LZTS2, TROAP and FAM117B (C-terminal fragment 245-589 a.a.) or GFP (control) were used for endogenous DYRK1A IP followed by WB with anti-HA antibody. C. Reciprocal IP/WB assays confirming the interaction between DYRK1A and the indicated candidate interacting proteins at the endogenous levels in T98G cells. Normal rabbit IgG serves as negative control. Figure S2. Induced expression of DYRK1A inhibits 53BP1 accumulation at DSB sites. A. Representative images (63x) of the indicated inducible U-2 OS cell lines treated with doxycycline for 16h, γ-irradiated (5Gy) and incubated for 3h before staining with anti-53BP1 antibody and DAPI. Figure S3. Characterization of the DYRK1A phospho-site mutants of RNF169. A. Representative images show that RNF169-DD mutant are recruited to IRIFs similar to the wild type RNF169. U-2 OS stably expressing RNF169-DD were γ-irradiated (5Gy) and incubated for 3h before staining with anti-HA antibody to detect RNF169. B-D. Quantitation of the HA-RNF169-DD and 53BP1 IRIF formation in U-2 OS cells shown in A. Graph shows average ± stdev of 3 independent experiments. E. Ubiquitin binding assay reveals no differences between the wild type and the phosphosite-mutant RNF169 proteins. HA-tagged RNF169 constructs were transiently expressed in HEK293T cells, extracted using RIPA buffer, immunoprecipitated with anti-HA antibodies and incubated with K63 poly-ubiquitin chains (Boston Biochem, Cat# UC-330) for 2 h at 4°C. After washing the beads, bound ubiquitin as well as RNF169 proteins in the samples were detected by WB. F. Co- immunoprecipitation assay shown that disruption of the DYRK1A phosphorylation sites in RNF169 does not affect its interaction with USP7. Figure S4. shRNA depletion of DYRK1A inhibits recruitment of RNF169 and 53BP1. A. DYRK1A depletion causes a modest decrease of HA-RNF169 IRIF formation. HA-RNF169 was transiently expressed in control and DYRK1A-depleted U-2 OS cells. Graph shows quantification (average ± stdev, N=3) of HA- RNF169 foci in the control and DYRK1A-depleted cells 3 h after γ-radiation (5 Gy). B. Depletion of DYRK1A using shRNA results in decreased 53BP1 IRIF formation. U-2 OS cells were treated and analyzed as in panel A. Western blots above the graphs confirm the efficiency of shRNA-mediated depletion of DYRK1A. Figure S5. Generation of cell lines devoid of DYRK1A expression using CRISR-Cas9 approach. A. DNA fluorescence in situ hybridization (FISH) assay using two different probes specific to DYRK1A labeled with Cy3 (red) and two control probes labeled with FITC (green) detects a single allele of DYRK1A gene in U-2 OS cell line. Schematic of human ch21 regions spanned by the FISH probes specific to DYRK1A gene (shown in red) and control regions (shown in green), as well as representative cell images are shown. The DNA probes were obtained from the BACPAC Resource Center, the Children's Hospital Oakland Research Institute (Oakland, CA), labeled and used for FISH assay as described in (Zheng et al., 2010). B. Location of DYRK1A-specific guiding RNA sequences relative to genomic sequences of human and mouse DYRK1A. C. Human DYRK1A- KO U-2 OS clones. Sequencing analysis of the DYRK1A gene region targeted by CRISPR/Cas9 mutagenesis confirms presence of an extended insertion/deletion lesion in the KO clone #1, and a single nucleotide deletion in the KO clone #2. Genomic DNA region of interest was amplified by PCR, sequenced and aligned to genomic DYRK1A reference sequence using SnapGene software. WB analysis using two different DYRK1A antibodies confirms the loss of the full-length protein. D. Same as in panel C, only with mouse Dyrk1a-KO NIH3T3 clones. Figure S6. Characterization of the DYRK1A-KO U-2 OS cells. A. WB analysis of the indicated DNA damage signaling factors in the control (C) and DYRK1A-KO cells (clone #1) at different times after γ-irradiation (5 Gy). Vinculin and β-actin serve as loading controls for high and low molecular weight proteins, respectively. Note elevated expression of 53BP1 and BRCA1 in DYRK1A-KO cells both before and after irradiation. B. Propidium iodide (PI) stained DNA FACS analysis of the control and DYRK1A-KO U-2 OS cells reveals similar cell cycle changes in response to DNA damage. Figure S7. Characterization of the DYRK1A-KO U-2 OS cells (continued). A. DYRK1A-KO U-2 OS cells (clone #1) display normal patterns of the γH2AX and ubiquitylated protein accumulation at the sites of DNA damage. Cells were processed for staining with anti-γH2AX and anti-ubiquitylated protein antibodies (FK2) at 3h post-γ-irradiation (5 Gy). B. Recruitment of BRCA1 to the DSB sites appears normal in DYRK1A-KO U-2 OS cells (clone #1). The cells were treated and processed as in panel A. Figure S8. A. WB detection of adeno-SceI (HA-tagged) expression in the control and DYRK1A-KO U-2 OS cells. DYRK1A and vinculin are shown as controls. B. Representative FACS data show higher fraction of the GFP-positive cells in Ad-SceI-infected DYRK1A-KO U-2 OS cells compared to controls. Supplemental References Glenewinkel, F., Cohen, M.J., King, C.R., Kaspar, S., Bamberg-Lemper, S., Mymryk, J.S., and Becker, W. (2016). The adaptor protein DCAF7 mediates the interaction of the adenovirus E1A oncoprotein with the protein kinases DYRK1A and HIPK2. Scientific reports 6, 28241. Pierce, A.J., Johnson, R.D., Thompson, L.H., and Jasin, M. (1999). XRCC3 promotes homology-directed repair of DNA damage in mammalian cells. Genes Dev 13, 2633-2638. Poulsen, M., Lukas, C., Lukas, J., Bekker-Jensen, S., and Mailand, N. (2012). Human RNF169 is a negative regulator of the ubiquitin-dependent response to DNA double-strand breaks. J Cell Biol 197, 189-199. Varjosalo, M., Keskitalo, S., Van Drogen, A., Nurkkala, H., Vichalkovski, A., Aebersold, R., and Gstaiger, M. (2013). The protein interaction landscape of the human CMGC kinase group. Cell reports 3, 1306-1320. Yabut, O., Domogauer, J., and D'Arcangelo, G. (2010). Dyrk1A overexpression inhibits proliferation and induces premature neuronal differentiation of neural progenitor cells. J Neurosci 30, 4004-4014. Yakovlev, V.A. (2013). Nitric oxide-dependent downregulation of BRCA1 expression promotes genetic instability. Cancer Res 73, 706-715. Zheng, H., Ying, H., Wiedemeyer, R., Yan, H., Quayle, S.N., Ivanova, E.V., Paik, J.H., Zhang, H., Xiao, Y., Perry, S.R., et al. (2010). PLAGL2 regulates Wnt signaling to impede differentiation in neural stem cells and gliomas. Cancer Cell 17, 497-509. Menon et al., Supplemental Figure 1 (related to Fig. 1) Varjosalo only Overlap A (10) (14) ABCD3 DCAF7* CCDC8 DYNLL2 CCNA2 FAM117A CREBBP FAM117B DYNLL1 FAM53C EP300* FNTA FNTB GLCCI1 RB1* KIAA0232 RBL1* LZTS2 RBL2* PPP1R3F PRKAR1A RNF169 SIPA1L1 TROAP B IP: IP: IP: IP: Input DYRK1A Input DYRK1A Input DYRK1A Input DYRK1A FAM117B-CTAP FAM117B-CTAP FAM117B-CTAP DCAF7-CTAP DCAF7-CTAP DCAF7-CTAP GFP-CTAP GFP-CTAP GFP-CTAP GFP-CTAP GFP-CTAP GFP-CTAP LZTS2-CTAP LZTS2-CTAP LZTS2-CTAP GFP-CTAP GFP-CTAP GFP-CTAP NTAP-TROAP NTAP-TROAP NTAP-TROAP GFP-CTAP GFP-CTAP GFP-CTAP HA DYRK1A C IP: FAM117B FAM117B IP: Input IP: IgG Input IP: IgG IP: LZTS2 Input TROAP IP: IP: IgG IP: IgG Input IP: IgG IP: IgG IP: DYRK1A DYRK1A IP: DYRK1A DYRK1A DYRK1A DYRK1A DCAF7 FAM117B LZTS2 TROAP Menon et al., Supplemental Figure 2 (related to Fig. 2) Vector DYRK1A-WT DYRK1A-KR 53BP1 DAPI 53BP1+ Menon et al., Supplemental Figure 3 (related to Fig. 5) A WT S368D/S403D (DD) HA-RNF169 B C 90 n.s. 100 14 75 12 n.s. 80 10 60 * 60 8 45 6 40 30 4 HA Foci/Nucleus HA 15 20 2 Foci % Cells>10 HA % Cells>10 53BP1 Foci 0 0 0 WT DD WT DD Mock WT DD D E IP:HA Input IP: HA UIR 15 Gy, 3h UIR 15 Gy, 3h HA-RNF169 - WT AA DD Input K63 HA-RNF169 - WT AA DD - WT AA DD - WT AA DD - WT AA DD Ubiquitin USP7 HA β-actin HA HA (Input) Menon et al., Supplemental Figure 4 Related to Fig. 5) A DYRK1A HA-RNF169 β-actin 25 75 * 60 20 * 45 15 30 10 5 RNF169 Foci 15 %Cells with >10 HA- 0 0 HA-RNF169 Foci/Nucleus B DYRK1A β-actin 100 25 * 20 75 * 15 50 10 25 5 0 53BP1 Foci/Nucleus 0 Cells with >10 53BP1 Foci Menon et al., Supplemental Figure 5 (related to Fig. 6) A B RP11- 98O13 ! RP11-61A21 ! Genomic DNA! gRNA binding! RP11- 24M21 ! RP11- 105O24 ! hDYRK1A gRNA! 37486469 – RP11- 98O13 ! Homo sapiens chromosome 21, 37486488" RP11-61A21 ! GRCh38.p2 Primary Assembly; " RP11- 24M21 ! RP11- 105O24 ! DYRK1A; NC_000021.9 " " " mDYRK1A gRNA! 94665960 –" Mus musculus strain C57BL/6J 94665979" chromosome 16, GRCm38.p4 C57BL/ " 6J; DYRK1A; NC_000082.6" " " " CST D30C10 Bethyl 303-801A (around N523) (N-terminus) kDa KO #1 KO #2 Parental Control KO #1 #2 KO Parental Control C 100 DCAF7 Kinase HIS DYRK1A 75 93-104 159-479 599-619 50 37 25 1 NLS 763 20 117-134 15 10 Actin KO #2 KO #1 +112 bp del8 bp Sigma 7D10 CST D30C10 (C-Terminus) (around N523) KO #1 #2 KO Control KO #1 KO KO #2 kDa Control 100 DCAF7 Kinase HIS DYRK1A 75 93-104 159-479 599-619 D 50 37 1 NLS 763 25 117-134 20 15 Actin KO #1 KO #2 Menon et al., Supplemental Figure 6 (related to Fig.
Recommended publications
  • Clinical Utility Gene Card For: 3-M Syndrome – Update 2013
    European Journal of Human Genetics (2014) 22, doi:10.1038/ejhg.2013.156 & 2014 Macmillan Publishers Limited All rights reserved 1018-4813/14 www.nature.com/ejhg CLINICAL UTILITY GENE CARD UPDATE Clinical utility gene card for: 3-M syndrome – Update 2013 Muriel Holder-Espinasse*,1, Melita Irving1 and Vale´rie Cormier-Daire2 European Journal of Human Genetics (2014) 22, doi:10.1038/ejhg.2013.156; published online 31 July 2013 Update to: European Journal of Human Genetics (2011) 19, doi:10.1038/ejhg.2011.32; published online 2 March 2011 1. DISEASE CHARACTERISTICS nonsense and missense mutations c.4333C4T (p.Arg1445*) and 1.1 Name of the disease (synonyms) c.4391A4C (p.His1464Pro), respectively, render CUL7 deficient 3-M syndrome (gloomy face syndrome, dolichospondylic dysplasia). in recruiting ROC1, leading to impaired ubiquitination. OBSL1: microsatellites analysis of the locus (2q35-36.1) in con- 1.2 OMIM# of the disease sanguineous families. OBSL1: microsatellites analysis of the locus 273750. (2q35-36.1) in consanguineous families. Mutations induce non- sense mediated decay. Knockdown of OBSL1 in HEK293 cells 1.3 Name of the analysed genes or DNA/chromosome segments shows the role of this gene in the maintenance of normal levels of CUL7, OBSL1 and CCDC8.1–5 CUL7. Abnormal IGFBP2 andIGFBP5 mRNA levels in two patients with OBSL1 mutations, suggesting that OBSL1 modulates the 1.4 OMIM# of the gene(s) expression of IGFBP proteins. CCDC8: microsatellites analysis 609577 (CUL7), 610991 (OBSL1) and 614145 (CCDC8). at the locus (19q13.2-q13.32). CCDC8, 1-BP DUP, 612G and CCDC8, 1-BP.
    [Show full text]
  • Identification and Validation of Methylation-Driven Genes Prognostic Signature for Recurrence of Laryngeal Squamous Cell Carcino
    Cui et al. Cancer Cell Int (2020) 20:472 https://doi.org/10.1186/s12935-020-01567-3 Cancer Cell International PRIMARY RESEARCH Open Access Identifcation and validation of methylation-driven genes prognostic signature for recurrence of laryngeal squamous cell carcinoma by integrated bioinformatics analysis Jie Cui3†, Liping Wang2†, Waisheng Zhong4†, Zhen Chen5, Jie Chen4*, Hong Yang3* and Genglong Liu1* Abstract Background: Recurrence remains a major obstacle to long-term survival of laryngeal squamous cell carcinoma (LSCC). We conducted a genome-wide integrated analysis of methylation and the transcriptome to establish methyla- tion-driven genes prognostic signature (MDGPS) to precisely predict recurrence probability and optimize therapeutic strategies for LSCC. Methods: LSCC DNA methylation datasets and RNA sequencing (RNA-seq) dataset were acquired from the Cancer Genome Atlas (TCGA). MethylMix was applied to detect DNA methylation-driven genes (MDGs). By univariate and multivariate Cox regression analyses, fve genes of DNA MDGs was developed a recurrence-free survival (RFS)-related MDGPS. The predictive accuracy and clinical value of the MDGPS were evaluated by receiver operating characteristic (ROC) and decision curve analysis (DCA), and compared with TNM stage system. Additionally, prognostic value of MDGPS was validated by external Gene Expression Omnibus (GEO) database. According to 5 MDGs, the candidate small molecules for LSCC were screen out by the CMap database. To strengthen the bioinformatics analysis results, 30 pairs of clinical samples were evaluated by digoxigenin-labeled chromogenic in situ hybridization (CISH). Results: A total of 88 DNA MDGs were identifed, and fve RFS-related MDGs (LINC01354, CCDC8, PHYHD1, MAGEB2 and ZNF732) were chosen to construct a MDGPS.
    [Show full text]
  • 1 Supporting Information for a Microrna Network Regulates
    Supporting Information for A microRNA Network Regulates Expression and Biosynthesis of CFTR and CFTR-ΔF508 Shyam Ramachandrana,b, Philip H. Karpc, Peng Jiangc, Lynda S. Ostedgaardc, Amy E. Walza, John T. Fishere, Shaf Keshavjeeh, Kim A. Lennoxi, Ashley M. Jacobii, Scott D. Rosei, Mark A. Behlkei, Michael J. Welshb,c,d,g, Yi Xingb,c,f, Paul B. McCray Jr.a,b,c Author Affiliations: Department of Pediatricsa, Interdisciplinary Program in Geneticsb, Departments of Internal Medicinec, Molecular Physiology and Biophysicsd, Anatomy and Cell Biologye, Biomedical Engineeringf, Howard Hughes Medical Instituteg, Carver College of Medicine, University of Iowa, Iowa City, IA-52242 Division of Thoracic Surgeryh, Toronto General Hospital, University Health Network, University of Toronto, Toronto, Canada-M5G 2C4 Integrated DNA Technologiesi, Coralville, IA-52241 To whom correspondence should be addressed: Email: [email protected] (M.J.W.); yi- [email protected] (Y.X.); Email: [email protected] (P.B.M.) This PDF file includes: Materials and Methods References Fig. S1. miR-138 regulates SIN3A in a dose-dependent and site-specific manner. Fig. S2. miR-138 regulates endogenous SIN3A protein expression. Fig. S3. miR-138 regulates endogenous CFTR protein expression in Calu-3 cells. Fig. S4. miR-138 regulates endogenous CFTR protein expression in primary human airway epithelia. Fig. S5. miR-138 regulates CFTR expression in HeLa cells. Fig. S6. miR-138 regulates CFTR expression in HEK293T cells. Fig. S7. HeLa cells exhibit CFTR channel activity. Fig. S8. miR-138 improves CFTR processing. Fig. S9. miR-138 improves CFTR-ΔF508 processing. Fig. S10. SIN3A inhibition yields partial rescue of Cl- transport in CF epithelia.
    [Show full text]
  • Screening and Identification of Hub Genes in Bladder Cancer by Bioinformatics Analysis and KIF11 Is a Potential Prognostic Biomarker
    ONCOLOGY LETTERS 21: 205, 2021 Screening and identification of hub genes in bladder cancer by bioinformatics analysis and KIF11 is a potential prognostic biomarker XIAO‑CONG MO1,2*, ZI‑TONG ZHANG1,3*, MENG‑JIA SONG1,2, ZI‑QI ZHOU1,2, JIAN‑XIONG ZENG1,2, YU‑FEI DU1,2, FENG‑ZE SUN1,2, JIE‑YING YANG1,2, JUN‑YI HE1,2, YUE HUANG1,2, JIAN‑CHUAN XIA1,2 and DE‑SHENG WENG1,2 1State Key Laboratory of Oncology in South China, Collaborative Innovation Centre for Cancer Medicine; 2Department of Biotherapy, Sun Yat‑Sen University Cancer Center; 3Department of Radiation Oncology, Sun Yat‑Sen University Cancer Center, Guangzhou, Guangdong 510060, P.R. China Received July 31, 2020; Accepted December 18, 2020 DOI: 10.3892/ol.2021.12466 Abstract. Bladder cancer (BC) is the ninth most common immunohistochemistry and western blotting. In summary, lethal malignancy worldwide. Great efforts have been devoted KIF11 was significantly upregulated in BC and might act as to clarify the pathogenesis of BC, but the underlying molecular a potential prognostic biomarker. The present identification mechanisms remain unclear. To screen for the genes associated of DEGs and hub genes in BC may provide novel insight for with the progression and carcinogenesis of BC, three datasets investigating the molecular mechanisms of BC. were obtained from the Gene Expression Omnibus. A total of 37 tumor and 16 non‑cancerous samples were analyzed to Introduction identify differentially expressed genes (DEGs). Subsequently, 141 genes were identified, including 55 upregulated and Bladder cancer (BC) is the ninth most common malignancy 86 downregulated genes. The protein‑protein interaction worldwide with substantial morbidity and mortality.
    [Show full text]
  • Supplementary Data
    SUPPLEMENTARY DATA A cyclin D1-dependent transcriptional program predicts clinical outcome in mantle cell lymphoma Santiago Demajo et al. 1 SUPPLEMENTARY DATA INDEX Supplementary Methods p. 3 Supplementary References p. 8 Supplementary Tables (S1 to S5) p. 9 Supplementary Figures (S1 to S15) p. 17 2 SUPPLEMENTARY METHODS Western blot, immunoprecipitation, and qRT-PCR Western blot (WB) analysis was performed as previously described (1), using cyclin D1 (Santa Cruz Biotechnology, sc-753, RRID:AB_2070433) and tubulin (Sigma-Aldrich, T5168, RRID:AB_477579) antibodies. Co-immunoprecipitation assays were performed as described before (2), using cyclin D1 antibody (Santa Cruz Biotechnology, sc-8396, RRID:AB_627344) or control IgG (Santa Cruz Biotechnology, sc-2025, RRID:AB_737182) followed by protein G- magnetic beads (Invitrogen) incubation and elution with Glycine 100mM pH=2.5. Co-IP experiments were performed within five weeks after cell thawing. Cyclin D1 (Santa Cruz Biotechnology, sc-753), E2F4 (Bethyl, A302-134A, RRID:AB_1720353), FOXM1 (Santa Cruz Biotechnology, sc-502, RRID:AB_631523), and CBP (Santa Cruz Biotechnology, sc-7300, RRID:AB_626817) antibodies were used for WB detection. In figure 1A and supplementary figure S2A, the same blot was probed with cyclin D1 and tubulin antibodies by cutting the membrane. In figure 2H, cyclin D1 and CBP blots correspond to the same membrane while E2F4 and FOXM1 blots correspond to an independent membrane. Image acquisition was performed with ImageQuant LAS 4000 mini (GE Healthcare). Image processing and quantification were performed with Multi Gauge software (Fujifilm). For qRT-PCR analysis, cDNA was generated from 1 µg RNA with qScript cDNA Synthesis kit (Quantabio). qRT–PCR reaction was performed using SYBR green (Roche).
    [Show full text]
  • TROAP Switches DYRK1 Activity to Drive Hepatocellular Carcinoma
    Li et al. Cell Death and Disease (2021) 12:125 https://doi.org/10.1038/s41419-021-03422-3 Cell Death & Disease ARTICLE Open Access TROAP switches DYRK1 activity to drive hepatocellular carcinoma progression Lei Li1,2,3,Jia-RuWei4, Ye Song5,ShuoFang6,YanyuDu6,ZhuoLi1, Ting-Ting Zeng1,Ying-HuiZhu 1, Yan Li 1 and Xin-Yuan Guan 1,2,3 Abstract Hepatocellular carcinoma (HCC) is one of the common malignancy and lacks effective therapeutic targets. Here, we demonstrated that ectopic expression of trophinin-associated protein (TROAP) dramatically drove HCC cell growth assessed by foci formation in monolayer culture, colony formation in soft agar and orthotopic liver transplantation in nude mice. Inversely, silencing TROAP expression with short-hairpin RNA attenuated the malignant proliferation of HCC cells in vitro and in vivo. Next, mechanistic investigation revealed that TROAP directly bound to dual specificity tyrosine phosphorylation regulated kinase 1A/B (DYRK1A/B), resulting in the cytoplasmic retention of proteins DYRK1A/B and promoting cell cycle process via activation of Akt/GSK-3β signaling. Combination of cisplatin with an inhibitor of DYRK1 AZ191 effectively inhibited tumor growth in mouse model for HCC cells with high level of TROAP. Clinically, TROAP was significantly upregulated by miR-142-5p in HCC tissues, which predicted the poor survival of patients with HCC. Therefore, TROAP/DYRK1/Akt axis may be a promising therapeutic target and prognostic indicator for patients with HCC. 1234567890():,; 1234567890():,; 1234567890():,; 1234567890():,; Introduction 19952. It as a cytoplasmic protein is composed of 778 Hepatocellular carcinoma (HCC) is the most common amino acid residues and contains potential phosphoryla- pathological type of liver cancer, accounting for 75%–85% tion sites for protein kinases.
    [Show full text]
  • Genome-Wide Transcriptional Sequencing Identifies Novel Mutations in Metabolic Genes in Human Hepatocellular Carcinoma DAOUD M
    CANCER GENOMICS & PROTEOMICS 11 : 1-12 (2014) Genome-wide Transcriptional Sequencing Identifies Novel Mutations in Metabolic Genes in Human Hepatocellular Carcinoma DAOUD M. MEERZAMAN 1,2 , CHUNHUA YAN 1, QING-RONG CHEN 1, MICHAEL N. EDMONSON 1, CARL F. SCHAEFER 1, ROBERT J. CLIFFORD 2, BARBARA K. DUNN 3, LI DONG 2, RICHARD P. FINNEY 1, CONSTANCE M. CULTRARO 2, YING HU1, ZHIHUI YANG 2, CU V. NGUYEN 1, JENNY M. KELLEY 2, SHUANG CAI 2, HONGEN ZHANG 2, JINGHUI ZHANG 1,4 , REBECCA WILSON 2, LAUREN MESSMER 2, YOUNG-HWA CHUNG 5, JEONG A. KIM 5, NEUNG HWA PARK 6, MYUNG-SOO LYU 6, IL HAN SONG 7, GEORGE KOMATSOULIS 1 and KENNETH H. BUETOW 1,2 1Center for Bioinformatics and Information Technology, National Cancer Institute, Rockville, MD, U.S.A.; 2Laboratory of Population Genetics, National Cancer Institute, National Cancer Institute, Bethesda, MD, U.S.A.; 3Basic Prevention Science Research Group, Division of Cancer Prevention, National Cancer Institute, Bethesda, MD, U.S.A; 4Department of Biotechnology/Computational Biology, St. Jude Children’s Research Hospital, Memphis, TN, U.S.A.; 5Department of Internal Medicine, University of Ulsan College of Medicine, Asan Medical Center, Seoul, Korea; 6Department of Internal Medicine, University of Ulsan College of Medicine, Ulsan University Hospital, Ulsan, Korea; 7Department of Internal Medicine, College of Medicine, Dankook University, Cheon-An, Korea Abstract . We report on next-generation transcriptome Worldwide, liver cancer is the fifth most common cancer and sequencing results of three human hepatocellular carcinoma the third most common cause of cancer-related mortality (1). tumor/tumor-adjacent pairs.
    [Show full text]
  • NIH Public Access Author Manuscript Mol Cell
    NIH Public Access Author Manuscript Mol Cell. Author manuscript; available in PMC 2015 June 05. NIH-PA Author ManuscriptPublished NIH-PA Author Manuscript in final edited NIH-PA Author Manuscript form as: Mol Cell. 2014 June 5; 54(5): 791–804. doi:10.1016/j.molcel.2014.03.047. The 3M complex maintains microtubule and genome integrity Jun Yan1, Feng Yan1, Zhijun Li1, Becky Sinnott4, Kathryn M. Cappell4,7, Yanbao Yu2, Jinyao Mo5, Joseph A. Duncan1,5, Xian Chen2, Valerie Cormier-Daire6, Angelique W. Whitehurst4,8, and Yue Xiong1,2,3,* 1Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599-7295, USA. 2Department of Biochemistry and Biophysics, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599-7295, USA. 3Program in Molecular Biology and Biotechnology, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599-7295, USA. 4Department of Pharmacology, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599-7295, USA. 5Department of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599-7295, USA. 6University Paris Descartes, Department of Genetics and INSERM U781, Hospital Necker Enfants-Malades, Paris, France SUMMARY CUL7, OBSL1, and CCDC8 genes are mutated in a mutually exclusive manner in 3M and other growth retardation syndromes. The mechanism underlying the function of the three 3M genes in development is not known. We found that OBSL1 and CCDC8 form a complex with CUL7 and regulate the level and centrosomal localization of CUL7, respectively. CUL7 depletion results in altered microtubule dynamics, prometaphase arrest, tetraploidy and mitotic cell death.
    [Show full text]
  • Why It Is Difficult to Distinguish the Silver-Russell
    Why it is dicult to distinguish the Silver-Russell syndrome (SRS) and 3M syndrome in clinical practice Beibei Zhang Department of Endocrinology, Genetics, Metabolism, Beijing Children’s Hospital, Capital Medical University, National Center for Children’s Health, Beijing 100045, China Chunxiu Gong ( [email protected] ) Department of Endocrinology, Genetics, Metabolism, Beijing Children’s Hospital, Capital Medical University, National Center for Children’s Health, Beijing 100045, China Research Keywords: Silver-Russell syndrome, 3M syndrome, NH-CSS, Phenotype Posted Date: April 15th, 2020 DOI: https://doi.org/10.21203/rs.3.rs-22088/v1 License: This work is licensed under a Creative Commons Attribution 4.0 International License. Read Full License Page 1/10 Abstract Background: To analyze why 3M syndrome can be regarded as a SRS-like syndrome by examining the patients with 3M syndrome and research the connection between 3M syndrome and SRS. Methods: The term “3M syndrome” was retrieved by Web of Science and the 3M patients were screened by NH-CSS to determine whether it was consist with the diagnosis of clinical SRS, and to analyze the relationship between the two diseases by exploring literature. Results: Among patients with 3M syndrome, 60/70 (83%) were in accordance with clinical SRS, and the coincidence rates of CUL7, OBSL1, CCDC8 gene mutations were: 30 (42%), 17 (24%) and 4 (6%) respectively; The phenotypes of 3M syndrome conrmed as clinical SRS were SGA (90%), short stature (100%), forehead protruding (100%), relative macrocephaly (100%), feeding diculties/low BMI (33%), body asymmetry (0); Skeletal abnormalities and pathogenesis were previously considered as the key points of differentiation also were overlaps between two diseases; Symptomatic treatment and GH- treatment were carried out.
    [Show full text]
  • Mouse Troap Knockout Project (CRISPR/Cas9)
    https://www.alphaknockout.com Mouse Troap Knockout Project (CRISPR/Cas9) Objective: To create a Troap knockout Mouse model (C57BL/6J) by CRISPR/Cas-mediated genome engineering. Strategy summary: The Troap gene (NCBI Reference Sequence: NM_001162506 ; Ensembl: ENSMUSG00000032783 ) is located on Mouse chromosome 15. 14 exons are identified, with the ATG start codon in exon 2 and the TGA stop codon in exon 14 (Transcript: ENSMUST00000230054). Exon 4~10 will be selected as target site. Cas9 and gRNA will be co-injected into fertilized eggs for KO Mouse production. The pups will be genotyped by PCR followed by sequencing analysis. Note: Exon 4 starts from about 16.72% of the coding region. Exon 4~10 covers 35.73% of the coding region. The size of effective KO region: ~3963 bp. The KO region does not have any other known gene. Page 1 of 8 https://www.alphaknockout.com Overview of the Targeting Strategy Wildtype allele 5' gRNA region gRNA region 3' 1 4 5 6 7 8 9 10 14 Legends Exon of mouse Troap Knockout region Page 2 of 8 https://www.alphaknockout.com Overview of the Dot Plot (up) Window size: 15 bp Forward Reverse Complement Sequence 12 Note: The 1490 bp section upstream of Exon 4 is aligned with itself to determine if there are tandem repeats. No significant tandem repeat is found in the dot plot matrix. So this region is suitable for PCR screening or sequencing analysis. Overview of the Dot Plot (down) Window size: 15 bp Forward Reverse Complement Sequence 12 Note: The 584 bp section downstream of Exon 10 is aligned with itself to determine if there are tandem repeats.
    [Show full text]
  • Identification of DNA Methylation-Driven Genes in Esophageal Squamous Cell Carcinoma: a Study Based on the Cancer Genome Atlas
    Lu et al. Cancer Cell Int (2019) 19:52 https://doi.org/10.1186/s12935-019-0770-9 Cancer Cell International PRIMARY RESEARCH Open Access Identifcation of DNA methylation-driven genes in esophageal squamous cell carcinoma: a study based on The Cancer Genome Atlas Tong Lu1, Di Chen2, Yuanyong Wang1, Xiao Sun1, Shicheng Li1, Shuncheng Miao1, Yang Wo1, Yanting Dong1, Xiaoliang Leng1, Wenxing Du1 and Wenjie Jiao1* Abstract Background: Aberrant DNA methylations are signifcantly associated with esophageal squamous cell carcinoma (ESCC). In this study, we aimed to investigate the DNA methylation-driven genes in ESCC by integrative bioinformatics analysis. Methods: Data of DNA methylation and transcriptome profling were downloaded from TCGA database. DNA methylation-driven genes were obtained by methylmix R package. David database and ConsensusPathDB were used to perform gene ontology (GO) analysis and pathway analysis, respectively. Survival R package was used to analyze overall survival analysis of methylation-driven genes. Results: Totally 26 DNA methylation-driven genes were identifed by the methylmix, which were enriched in molecu- lar function of DNA binding and transcription factor activity. Then, ABCD1, SLC5A10, SPIN3, ZNF69, and ZNF608 were recognized as signifcant independent prognostic biomarkers from 26 methylation-driven genes. Additionally, a fur- ther integrative survival analysis, which combined methylation and gene expression data, was identifed that ABCD1, CCDC8, FBXO17 were signifcantly associated with patients’ survival. Also, multiple aberrant methylation sites were found to be correlated with gene expression. Conclusion: In summary, we studied the DNA methylation-driven genes in ESCC by bioinformatics analysis, ofering better understand of molecular mechanisms of ESCC and providing potential biomarkers precision treatment and prognosis detection.
    [Show full text]
  • The GALNT9, BNC1 and CCDC8 Genes Are Frequently Epigenetically Dysregulated in Breast Tumours That Metastasise to the Brain Rajendra P
    Pangeni et al. Clinical Epigenetics (2015) 7:57 DOI 10.1186/s13148-015-0089-x RESEARCH Open Access The GALNT9, BNC1 and CCDC8 genes are frequently epigenetically dysregulated in breast tumours that metastasise to the brain Rajendra P. Pangeni1, Prasanna Channathodiyil1, David S. Huen2, Lawrence W. Eagles1, Balraj K. Johal2, Dawar Pasha2, Natasa Hadjistephanou2, Oliver Nevell2, Claire L. Davies2, Ayobami I. Adewumi2, Hamida Khanom2, Ikroop S. Samra2, Vanessa C. Buzatto2, Preethi Chandrasekaran2, Thoraia Shinawi3, Timothy P. Dawson4, Katherine M. Ashton4, Charles Davis4, Andrew R. Brodbelt5, Michael D. Jenkinson5, Ivan Bièche6, Farida Latif3, John L. Darling1, Tracy J. Warr1 and Mark R. Morris1,2,3* Abstract Background: Tumour metastasis to the brain is a common and deadly development in certain cancers; 18–30 % of breast tumours metastasise to the brain. The contribution that gene silencing through epigenetic mechanisms plays in these metastatic tumours is not well understood. Results: We have carried out a bioinformatic screen of genome-wide breast tumour methylation data available at The Cancer Genome Atlas (TCGA) and a broad literature review to identify candidate genes that may contribute to breast to brain metastasis (BBM). This analysis identified 82 candidates. We investigated the methylation status of these genes using Combined Bisulfite and Restriction Analysis (CoBRA) and identified 21 genes frequently methylated in BBM. We have identified three genes, GALNT9, CCDC8 and BNC1, that were frequently methylated (55, 73 and 71 %, respectively) and silenced in BBM and infrequently methylated in primary breast tumours. CCDC8 was commonly methylated in brain metastases and their associated primary tumours whereas GALNT9 and BNC1 were methylated and silenced only in brain metastases, but not in the associated primary breast tumours from individual patients.
    [Show full text]