Downloaded 13/6/12) (Kanehisa Et Al, 2010)

Total Page:16

File Type:pdf, Size:1020Kb

Downloaded 13/6/12) (Kanehisa Et Al, 2010) bioRxiv preprint doi: https://doi.org/10.1101/455709; this version posted October 30, 2018. The copyright holder for this preprint (which was not certified by peer review) is the author/funder, who has granted bioRxiv a license to display the preprint in perpetuity. It is made available under aCC-BY-NC-ND 4.0 International license. Coherent Transcription Factor Target Networks Illuminate Epithelial Remodelling and Oncogenic Notch Ian M. Overton1,2,3,4*, Andrew H. Sims1, Jeremy A. Owen2,5, Bret S. E. Heale1,6, Matthew J. Ford1,7, Alexander L. R. Lubbock1,8, Erola Pairo-Castineira1, Abdelkader Essafi1,9 1. MRC Institute of Genetics and Molecular Medicine, University of Edinburgh, Edinburgh EH4 2XU, UK 2. Department of Systems Biology, Harvard University, Boston MA 02115, USA 3. Centre for Synthetic and Systems Biology (SynthSys), University of Edinburgh, Edinburgh EH9 3BF, UK 4. Centre for Cancer Research and Cell Biology, Queen’s University Belfast, Belfast BT9 7AE, UK 5. Department of Physics, Massachusetts Institute of Technology, Cambridge MA 02139, USA 6. Current address: Intermountain Healthcare, 3930 River Walk, West Valley City UT 84120, USA 7. Current address: Rosalind & Morris Goodman Cancer Research Centre, McGill University, Montreal H3A 0G4, Canada 8. Current address: Department of Biochemistry, Vanderbilt University, Nashville TN 37232, USA 9. Current address: School of Cellular and Molecular Medicine, University of Bristol, Bristol BS8 1TD, UK *Corresponding author: [email protected] Running title: Mapping Transcription Factor Networks Subject categories: Network biology; Chromatin, Epigenetics, Genomics & Functional Genomics Manuscript character count: 168,671 1 bioRxiv preprint doi: https://doi.org/10.1101/455709; this version posted October 30, 2018. The copyright holder for this preprint (which was not certified by peer review) is the author/funder, who has granted bioRxiv a license to display the preprint in perpetuity. It is made available under aCC-BY-NC-ND 4.0 International license. Abstract Cell identity is governed by gene expression, regulated by Transcription Factor (TF) binding at cis- regulatory modules. Decoding the relationship between patterns of TF binding and the regulation of cognate target genes is nontrivial, remaining a fundamental limitation in understanding cell decision-making mechanisms. Identification of TF physical binding that is biologically ‘neutral’ is a current challenge. We present the ‘NetNC’ software for discovery of functionally coherent TF targets, applied to study gene regulation in early embryogenesis. Predicted neutral binding accounted for 50% to ≥80% of candidate target genes assigned from significant binding peaks. Novel gene functions and network modules were identified, including regulation of chromatin organisation and crosstalk with notch signalling. Orthologues of predicted TF targets discriminated breast cancer molecular subtypes and our analysis evidenced new tumour biology; for example, predicting networks that reshape Waddington’s landscape during EMT-like phenotype switching. Predicted invasion roles for SNX29, ATG3, UNK and IRX4 were validated using a tractable cell model. This work illuminates conserved molecular networks that regulate epithelial remodelling in development and disease, with potential implications for precision medicine. 2 bioRxiv preprint doi: https://doi.org/10.1101/455709; this version posted October 30, 2018. The copyright holder for this preprint (which was not certified by peer review) is the author/funder, who has granted bioRxiv a license to display the preprint in perpetuity. It is made available under aCC-BY-NC-ND 4.0 International license. 1 Introduction Transcriptional regulatory factors (TFs) govern gene expression, which is a crucial determinant of phenotype. Therefore, mapping transcriptional regulatory networks is an attractive approach to gain understanding of the molecular mechanisms underpinning both normal biology and disease (Shlyueva et al, 2014; Stampfel et al, 2015; Rhee et al, 2014). TF action is controlled in multiple ways; including protein-protein interactions, DNA sequence affinity, 3D chromatin conformation, post-translational modifications and the processes required for TF delivery to the nucleus (Zabidi & Stark, 2016; Rhee et al, 2014; Khoueiry et al, 2017). The interplay of mechanisms influencing TF specificity across different biological contexts encompasses considerable complexity and genome- scale assignment of TFs to individual genes is challenging (Shlyueva et al, 2014; Wilczynski & Furlong, 2010; Khoueiry et al, 2017). Indeed, much remains to be learned about the regulation of gene expression. For example, the relationship between enhancer sequences and the transcriptional activity of cognate promoters is only beginning to be understood (Khoueiry et al, 2017; Zabidi & Stark, 2016). Prediction of TF occupancy from DNA sequence composition alone has had only limited success, likely because protein interactions influence TF binding specificity (Jolma et al, 2015; Khoueiry et al, 2017). TF binding sites may be determined experimentally using chromatin immunoprecipitation followed by sequencing (ChIP-seq) or microarray (ChIP-chip). These and related methods (e.g. ChIP-exo, DamID) have revealed a substantial proportion of statistically significant ‘neutral’ TF binding, that has apparently no effect on transcription from the promoters of assigned target genes (Shlyueva et al, 2014; Li et al, 2008; Ozdemir et al, 2011; Biggin, 2011). Evidence suggests that neutral binding can arise from TF association with euchromatin; for example, the binding of randomly-selected TFs and genome-wide transcription levels are correlated (Cheng et al, 2012; Consortium, 2012; Brown & Celniker, 2015). Genomic regions that bind large numbers of TFs have been termed Highly Occupied Target (HOT) regions (Roy et al, 2010). HOT regions are enriched for disease SNPs and can function as developmental enhancers (Kvon et al, 2012; Li et al, 2015). However, a considerable proportion of individual TF binding events at HOT regions may have little effect on gene expression and association with chromatin accessibility suggests non-canonical regulatory function such as sequestration of TFs or in 3D genome organisation (Moorman et al, 2006; Montavon et al, 2011) as well as possible technical artefacts (Teytelman et al, 2013). A proportion of apparently neutral binding sites may also have more subtle functions; for example in combinatorial context-specific regulation and in buffering transcriptional noise (Cannavò et al, 3 bioRxiv preprint doi: https://doi.org/10.1101/455709; this version posted October 30, 2018. The copyright holder for this preprint (which was not certified by peer review) is the author/funder, who has granted bioRxiv a license to display the preprint in perpetuity. It is made available under aCC-BY-NC-ND 4.0 International license. 2016; Stampfel et al, 2015). Furthermore, enhancers may control the expression of genes that are sequence-distant but spatially close due to the 3D chromatin conformation (Moorman et al, 2006; Montavon et al, 2011). Current approaches to match bound TFs to candidate target genes may miss these distant regulatory relationships. Identification of bona fide, functional TF target genes remains a major obstacle in understanding the regulatory networks that control cell behaviour (Biggin, 2011; Stampfel et al, 2015; Keung et al, 2014; Brown & Celniker, 2015; Khoueiry et al, 2017). The set of genes regulated by an individual TF typically have overlapping expression patterns and coherent biological function (Igual et al, 1996; Karczewski et al, 2014; MacArthur et al, 2009). Indeed, gene regulatory networks are organised in a hierarchical, modular structure and TFs frequently act upon multiple nodes of a given module (Hartwell et al, 1999; Hooper et al, 2007). Therefore, we hypothesised that functional TF targets collectively share network properties that may differentiate them from neutrally bound sites. Graph theoretic analysis can reveal biologically meaningful gene modules, including cross-talk between canonical pathways (Ideker et al, 2002; Vidal et al, 2011; Jaeger et al, 2017) and conversely may enable elimination of neutrally bound candidate TF targets derived from statistically significant ChIP-seq or ChIP-chip peaks. For this purpose, we have developed a novel algorithm (NetNC) that may be applied to discover functional TF targets and so help to illuminate mechanisms controlling cell phenotype, for example to inform causality in regulatory network inference (Shlyueva et al, 2014; Wilczynski & Furlong, 2010). NetNC analyses the connectivity between candidate TF target genes in the context of a functional gene network (FGN), in order to discover biologically coherent TF targets. Network approaches afford significant advantages for handling biological complexity, enable genome-scale analysis of gene function (Hu et al, 2016; Greene et al, 2015), and are not restricted to predefined gene groupings used by standard functional annotation tools (e.g. GSEA, DAVID) (Ideker et al, 2002; Subramanian et al, 2005; Huang et al, 2009). FGNs seek to comprehensively represent gene function and provide a useful framework for analysis of noisy real-world data (Marcotte et al, 1999; Pe’er & Hacohen, 2011). Clustering is frequently applied to a FGN in order to define a fixed network decomposition, as basis for identification of biological modules (Enright et al, 2002; Wang et al, 2011). Modules with a high proportion
Recommended publications
  • Table S1. List of Proteins in the BAHD1 Interactome
    Table S1. List of proteins in the BAHD1 interactome BAHD1 nuclear partners found in this work yeast two-hybrid screen Name Description Function Reference (a) Chromatin adapters HP1α (CBX5) chromobox homolog 5 (HP1 alpha) Binds histone H3 methylated on lysine 9 and chromatin-associated proteins (20-23) HP1β (CBX1) chromobox homolog 1 (HP1 beta) Binds histone H3 methylated on lysine 9 and chromatin-associated proteins HP1γ (CBX3) chromobox homolog 3 (HP1 gamma) Binds histone H3 methylated on lysine 9 and chromatin-associated proteins MBD1 methyl-CpG binding domain protein 1 Binds methylated CpG dinucleotide and chromatin-associated proteins (22, 24-26) Chromatin modification enzymes CHD1 chromodomain helicase DNA binding protein 1 ATP-dependent chromatin remodeling activity (27-28) HDAC5 histone deacetylase 5 Histone deacetylase activity (23,29,30) SETDB1 (ESET;KMT1E) SET domain, bifurcated 1 Histone-lysine N-methyltransferase activity (31-34) Transcription factors GTF3C2 general transcription factor IIIC, polypeptide 2, beta 110kDa Required for RNA polymerase III-mediated transcription HEYL (Hey3) hairy/enhancer-of-split related with YRPW motif-like DNA-binding transcription factor with basic helix-loop-helix domain (35) KLF10 (TIEG1) Kruppel-like factor 10 DNA-binding transcription factor with C2H2 zinc finger domain (36) NR2F1 (COUP-TFI) nuclear receptor subfamily 2, group F, member 1 DNA-binding transcription factor with C4 type zinc finger domain (ligand-regulated) (36) PEG3 paternally expressed 3 DNA-binding transcription factor with
    [Show full text]
  • Hepatitis C Virus As a Unique Human Model Disease to Define
    viruses Review Hepatitis C Virus as a Unique Human Model Disease to Define Differences in the Transcriptional Landscape of T Cells in Acute versus Chronic Infection David Wolski and Georg M. Lauer * Liver Center at the Gastrointestinal Unit, Department of Medicine, Massachusetts General Hospital and Harvard Medical School, Boston, MA 02114, USA * Correspondence: [email protected]; Tel.: +1-617-724-7515 Received: 27 June 2019; Accepted: 23 July 2019; Published: 26 July 2019 Abstract: The hepatitis C virus is unique among chronic viral infections in that an acute outcome with complete viral elimination is observed in a minority of infected patients. This unique feature allows direct comparison of successful immune responses with those that fail in the setting of the same human infection. Here we review how this scenario can be used to achieve better understanding of transcriptional regulation of T-cell differentiation. Specifically, we discuss results from a study comparing transcriptional profiles of hepatitis C virus (HCV)-specific CD8 T-cells during early HCV infection between patients that do and do not control and eliminate HCV. Identification of early gene expression differences in key T-cell differentiation molecules as well as clearly distinct transcriptional networks related to cell metabolism and nucleosomal regulation reveal novel insights into the development of exhausted and memory T-cells. With additional transcriptional studies of HCV-specific CD4 and CD8 T-cells in different stages of infection currently underway, we expect HCV infection to become a valuable model disease to study human immunity to viruses. Keywords: viral hepatitis; hepatitis C virus; T cells; transcriptional regulation; transcription factors; metabolism; nucleosome 1.
    [Show full text]
  • Development and Validation of a Novel Immune-Related Prognostic Model
    Wang et al. J Transl Med (2020) 18:67 https://doi.org/10.1186/s12967-020-02255-6 Journal of Translational Medicine RESEARCH Open Access Development and validation of a novel immune-related prognostic model in hepatocellular carcinoma Zheng Wang1, Jie Zhu1, Yongjuan Liu3, Changhong Liu2, Wenqi Wang2, Fengzhe Chen1* and Lixian Ma1* Abstract Background: Growing evidence has suggested that immune-related genes play crucial roles in the development and progression of hepatocellular carcinoma (HCC). Nevertheless, the utility of immune-related genes for evaluating the prognosis of HCC patients are still lacking. The study aimed to explore gene signatures and prognostic values of immune-related genes in HCC. Methods: We comprehensively integrated gene expression data acquired from 374 HCC and 50 normal tissues in The Cancer Genome Atlas (TCGA). Diferentially expressed genes (DEGs) analysis and univariate Cox regression analysis were performed to identify DEGs that related to overall survival. An immune prognostic model was constructed using the Lasso and multivariate Cox regression analyses. Furthermore, Cox regression analysis was applied to identify independent prognostic factors in HCC. The correlation analysis between immune-related signature and immune cells infltration were also investigated. Finally, the signature was validated in an external independent dataset. Results: A total of 329 diferentially expressed immune‐related genes were detected. 64 immune‐related genes were identifed to be markedly related to overall survival in HCC patients using univariate Cox regression analysis. Then we established a TF-mediated network for exploring the regulatory mechanisms of these genes. Lasso and multivariate Cox regression analyses were applied to construct the immune-based prognostic model, which consisted of nine immune‐related genes.
    [Show full text]
  • 1714 Gene Comprehensive Cancer Panel Enriched for Clinically Actionable Genes with Additional Biologically Relevant Genes 400-500X Average Coverage on Tumor
    xO GENE PANEL 1714 gene comprehensive cancer panel enriched for clinically actionable genes with additional biologically relevant genes 400-500x average coverage on tumor Genes A-C Genes D-F Genes G-I Genes J-L AATK ATAD2B BTG1 CDH7 CREM DACH1 EPHA1 FES G6PC3 HGF IL18RAP JADE1 LMO1 ABCA1 ATF1 BTG2 CDK1 CRHR1 DACH2 EPHA2 FEV G6PD HIF1A IL1R1 JAK1 LMO2 ABCB1 ATM BTG3 CDK10 CRK DAXX EPHA3 FGF1 GAB1 HIF1AN IL1R2 JAK2 LMO7 ABCB11 ATR BTK CDK11A CRKL DBH EPHA4 FGF10 GAB2 HIST1H1E IL1RAP JAK3 LMTK2 ABCB4 ATRX BTRC CDK11B CRLF2 DCC EPHA5 FGF11 GABPA HIST1H3B IL20RA JARID2 LMTK3 ABCC1 AURKA BUB1 CDK12 CRTC1 DCUN1D1 EPHA6 FGF12 GALNT12 HIST1H4E IL20RB JAZF1 LPHN2 ABCC2 AURKB BUB1B CDK13 CRTC2 DCUN1D2 EPHA7 FGF13 GATA1 HLA-A IL21R JMJD1C LPHN3 ABCG1 AURKC BUB3 CDK14 CRTC3 DDB2 EPHA8 FGF14 GATA2 HLA-B IL22RA1 JMJD4 LPP ABCG2 AXIN1 C11orf30 CDK15 CSF1 DDIT3 EPHB1 FGF16 GATA3 HLF IL22RA2 JMJD6 LRP1B ABI1 AXIN2 CACNA1C CDK16 CSF1R DDR1 EPHB2 FGF17 GATA5 HLTF IL23R JMJD7 LRP5 ABL1 AXL CACNA1S CDK17 CSF2RA DDR2 EPHB3 FGF18 GATA6 HMGA1 IL2RA JMJD8 LRP6 ABL2 B2M CACNB2 CDK18 CSF2RB DDX3X EPHB4 FGF19 GDNF HMGA2 IL2RB JUN LRRK2 ACE BABAM1 CADM2 CDK19 CSF3R DDX5 EPHB6 FGF2 GFI1 HMGCR IL2RG JUNB LSM1 ACSL6 BACH1 CALR CDK2 CSK DDX6 EPOR FGF20 GFI1B HNF1A IL3 JUND LTK ACTA2 BACH2 CAMTA1 CDK20 CSNK1D DEK ERBB2 FGF21 GFRA4 HNF1B IL3RA JUP LYL1 ACTC1 BAG4 CAPRIN2 CDK3 CSNK1E DHFR ERBB3 FGF22 GGCX HNRNPA3 IL4R KAT2A LYN ACVR1 BAI3 CARD10 CDK4 CTCF DHH ERBB4 FGF23 GHR HOXA10 IL5RA KAT2B LZTR1 ACVR1B BAP1 CARD11 CDK5 CTCFL DIAPH1 ERCC1 FGF3 GID4 HOXA11 IL6R KAT5 ACVR2A
    [Show full text]
  • Xo GENE PANEL
    xO GENE PANEL Targeted panel of 1714 genes | Tumor DNA Coverage: 500x | RNA reads: 50 million Onco-seq panel includes clinically relevant genes and a wide array of biologically relevant genes Genes A-C Genes D-F Genes G-I Genes J-L AATK ATAD2B BTG1 CDH7 CREM DACH1 EPHA1 FES G6PC3 HGF IL18RAP JADE1 LMO1 ABCA1 ATF1 BTG2 CDK1 CRHR1 DACH2 EPHA2 FEV G6PD HIF1A IL1R1 JAK1 LMO2 ABCB1 ATM BTG3 CDK10 CRK DAXX EPHA3 FGF1 GAB1 HIF1AN IL1R2 JAK2 LMO7 ABCB11 ATR BTK CDK11A CRKL DBH EPHA4 FGF10 GAB2 HIST1H1E IL1RAP JAK3 LMTK2 ABCB4 ATRX BTRC CDK11B CRLF2 DCC EPHA5 FGF11 GABPA HIST1H3B IL20RA JARID2 LMTK3 ABCC1 AURKA BUB1 CDK12 CRTC1 DCUN1D1 EPHA6 FGF12 GALNT12 HIST1H4E IL20RB JAZF1 LPHN2 ABCC2 AURKB BUB1B CDK13 CRTC2 DCUN1D2 EPHA7 FGF13 GATA1 HLA-A IL21R JMJD1C LPHN3 ABCG1 AURKC BUB3 CDK14 CRTC3 DDB2 EPHA8 FGF14 GATA2 HLA-B IL22RA1 JMJD4 LPP ABCG2 AXIN1 C11orf30 CDK15 CSF1 DDIT3 EPHB1 FGF16 GATA3 HLF IL22RA2 JMJD6 LRP1B ABI1 AXIN2 CACNA1C CDK16 CSF1R DDR1 EPHB2 FGF17 GATA5 HLTF IL23R JMJD7 LRP5 ABL1 AXL CACNA1S CDK17 CSF2RA DDR2 EPHB3 FGF18 GATA6 HMGA1 IL2RA JMJD8 LRP6 ABL2 B2M CACNB2 CDK18 CSF2RB DDX3X EPHB4 FGF19 GDNF HMGA2 IL2RB JUN LRRK2 ACE BABAM1 CADM2 CDK19 CSF3R DDX5 EPHB6 FGF2 GFI1 HMGCR IL2RG JUNB LSM1 ACSL6 BACH1 CALR CDK2 CSK DDX6 EPOR FGF20 GFI1B HNF1A IL3 JUND LTK ACTA2 BACH2 CAMTA1 CDK20 CSNK1D DEK ERBB2 FGF21 GFRA4 HNF1B IL3RA JUP LYL1 ACTC1 BAG4 CAPRIN2 CDK3 CSNK1E DHFR ERBB3 FGF22 GGCX HNRNPA3 IL4R KAT2A LYN ACVR1 BAI3 CARD10 CDK4 CTCF DHH ERBB4 FGF23 GHR HOXA10 IL5RA KAT2B LZTR1 ACVR1B BAP1 CARD11 CDK5 CTCFL DIAPH1 ERCC1 FGF3 GID4
    [Show full text]
  • Cbx3 Maintains Lineage Specificity During Neural Differentiation
    Downloaded from genesdev.cshlp.org on September 27, 2021 - Published by Cold Spring Harbor Laboratory Press RESEARCH COMMUNICATION et al. 2013). However, Cbx3 is also enriched at promoters Cbx3 maintains lineage of mouse embryonic fibroblast (MEF)-derived pre-iPSCs specificity during neural (preinduced pluripotent stem cells), where it does not cor- relate with H3K9 methylation (Sridharan et al. 2013). differentiation RNAi knockdown of Cbx3 promotes reprogramming of fi- broblasts to iPSCs (Sridharan et al. 2013). Chengyang Huang,1,2 Trent Su,1 Yong Xue,1 1 3 1 Cbx3 plays important roles in developmental processes Chen Cheng, Fides D. Lay, Robin A. McKee, (Morikawa et al. 2013). In model systems, Cbx3 promotes Meiyang Li,2 Ajay Vashisht,1 James Wohlschlegel,1 neuronal maturation, kidney development, differentia- Bennett G. Novitch,4 Kathrin Plath,1 tion of ESCs to smooth muscle in culture, and embryonic Siavash K. Kurdistani,1 and Michael Carey1 arteriogenesis (Xiao et al. 2011; Dihazi et al. 2015; Oshiro et al. 2015). Little is known of how Cbx3 functions in dif- 1Department of Biological Chemistry, Eli and Edythe Broad ferentiation, but its promoter localization suggested that Center for Regenerative Medicine and Stem Cell Research, David it might directly affect transcription through the preinitia- Geffen School of Medicine, University of California at Los tion complex (PIC) (Grunberg and Hahn 2013; Allen and Angeles, Los Angeles California 90095, USA; 2Department of Taatjes 2015). The PIC is assembled in response to activa- Neurobiology, Shantou University Medical College, Shantou tors and requires the Mediator coactivator complex to re- 515041, China; 3Department of Molecular, Cell, and cruit the general transcription factors and Pol II (Chen Developmental Biology, University of California at Los Angeles, et al.
    [Show full text]
  • Targeting Cbx3/Hp1γ Induces LEF-1 and IL-21R to Promote Tumor-InLtrating CD8 T-Cell Persistence
    Targeting Cbx3/HP1γ Induces LEF-1 and IL-21R to Promote Tumor-Inltrating CD8 T-Cell Persistence To-Ha Thai ( [email protected] ) Beth Israel Deaconess Medical Center, Harvard Medical School Phuong Le Beth Israel Deaconess Medical Center, Harvard Medical School Ngoc Ha Drexel University Ngan Tran Beth Israel Deaconess Medical Center, Harvard Medical School Andrew Newman Institut für Zell- und Neurobiologie Charité - Universitätsmedizin Berlin Katharine Esselen Beth Israel Deaconess Medical Center, Harvard Medical School John Dalrymple Beth Israel Deaconess Medical Center Harvard Medical School Eva Schmelz Virginia Tech Avinash Bhandoola National Cancer Institute https://orcid.org/0000-0002-4657-8372 Hai-Hui Xue Hackensack University Medical Center https://orcid.org/0000-0002-9163-7669 Prim Singh Nazarbayev University School of Medicine Article Keywords: Cbx3/HP1g, LEF-1, IL-21R or IL-21 receptor, tumor-inltrating CD8+ T-cell persistence, adoptive T-cell therapy (ACT), effector memory/progenitor memory CD8+ T cells, ovarian cancer, neuroblastoma, melanoma. Posted Date: March 29th, 2021 DOI: https://doi.org/10.21203/rs.3.rs-343458/v1 License: This work is licensed under a Creative Commons Attribution 4.0 International License. Read Full License Targeting Cbx3/HP1g Induces LEF-1 and IL-21R to Promote Tumor-Infiltrating CD8 T- Cell Persistence Phuong T. Le1, Ngoc Ha1,3, Ngan K. Tran1,4, Andrew G. Newman5, Katharine M. Esselen6, John L. Dalrymple6, Eva M. Schmelz7, Avinash Bhandoola8,11, Hai-Hui Xue9,11, Prim B. Singh10,11, and To-Ha
    [Show full text]
  • CBX3 Monoclonal Antibody, Clone ACGB-3
    CBX3 monoclonal antibody, clone Entrez GeneID: 11335 ACGB-3 Gene Symbol: CBX3 Catalog Number: MAB22120 Gene Alias: HECH, HP1-GAMMA, HP1Hs-gamma Regulatory Status: For research use only (RUO) Gene Summary: At the nuclear envelope, the nuclear lamina and heterochromatin are adjacent to the inner Product Description: Rabbit monoclonal antibody nuclear membrane. The protein encoded by this gene raised against synthetic peptide of human CBX3. binds DNA and is a component of heterochromatin. This protein also can bind lamin B receptor, an integral Clone Name: ACGB-3 membrane protein found in the inner nuclear membrane. Immunogen: A synthetic peptide corresponding to The dual binding functions of the encoded protein may human CBX3. explain the association of heterochromatin with the inner nuclear membrane. Two transcript variants encoding the Host: Rabbit same protein but differing in the 5' UTR, have been found for this gene. [provided by RefSeq] Reactivity: Human,Mouse Applications: Flow Cyt, IHC, IP, WB-Ce (See our web site product page for detailed applications information) Protocols: See our web site at http://www.abnova.com/support/protocols.asp or product page for detailed protocols Specificity: The antibody reacts with human, mouse CBX3, in native form and recombinant. Superfamily members of CBX3 are not reactive to this antibody. Form: Liquid Purification: Affinity purification Isotype: IgG Recommend Usage: Flow Cytometry (1:100) Immunohistochemistry (1:50-1:200) Immunoprecipitation (1:50) Western Blot (1:500-1:2000) The optimal working dilution should be determined by the end user. Storage Buffer: In PBS, 150 mM NaCl, pH 7.4 (50% glycerol, 0.02% sodium azide).
    [Show full text]
  • Identifying the Risky SNP of Osteoporosis with ID3-PEP Decision Tree Algorithm
    Hindawi Complexity Volume 2017, Article ID 9194801, 8 pages https://doi.org/10.1155/2017/9194801 Research Article Identifying the Risky SNP of Osteoporosis with ID3-PEP Decision Tree Algorithm Jincai Yang,1 Huichao Gu,1 Xingpeng Jiang,1 Qingyang Huang,2 Xiaohua Hu,1 and Xianjun Shen1 1 School of Computer Science, Central China Normal University, Wuhan 430079, China 2School of Life Science, Central China Normal University, Wuhan 430079, China Correspondence should be addressed to Jincai Yang; [email protected] Received 31 March 2017; Revised 26 May 2017; Accepted 8 June 2017; Published 7 August 2017 Academic Editor: Fang-Xiang Wu Copyright © 2017 Jincai Yang et al. This is an open access article distributed under the Creative Commons Attribution License, which permits unrestricted use, distribution, and reproduction in any medium, provided the original work is properly cited. In the past 20 years, much progress has been made on the genetic analysis of osteoporosis. A number of genes and SNPs associated with osteoporosis have been found through GWAS method. In this paper, we intend to identify the suspected risky SNPs of osteoporosis with computational methods based on the known osteoporosis GWAS-associated SNPs. The process includes two steps. Firstly, we decided whether the genes associated with the suspected risky SNPs are associated with osteoporosis by using random walk algorithm on the PPI network of osteoporosis GWAS-associated genes and the genes associated with the suspected risky SNPs. In order to solve the overfitting problem in ID3 decision tree algorithm, we then classified the SNPs with positive results based on their features of position and function through a simplified classification decision tree which was constructed by ID3 decision tree algorithm with PEP (Pessimistic-Error Pruning).
    [Show full text]
  • Datasheet: VPA00776 Product Details
    Datasheet: VPA00776 Description: RABBIT ANTI CBX3 Specificity: CBX3 Format: Purified Product Type: PrecisionAb™ Polyclonal Isotype: Polyclonal IgG Quantity: 100 µl Product Details Applications This product has been reported to work in the following applications. This information is derived from testing within our laboratories, peer-reviewed publications or personal communications from the originators. Please refer to references indicated for further information. For general protocol recommendations, please visit www.bio-rad-antibodies.com/protocols. Yes No Not Determined Suggested Dilution Western Blotting 1/1000 PrecisionAb antibodies have been extensively validated for the western blot application. The antibody has been validated at the suggested dilution. Where this product has not been tested for use in a particular technique this does not necessarily exclude its use in such procedures. Further optimization may be required dependant on sample type. Target Species Human Species Cross Reacts with: Rat Reactivity N.B. Antibody reactivity and working conditions may vary between species. Product Form Purified IgG - liquid Preparation Rabbit polyclonal antibody purified by affinity chromatography on immunogen Buffer Solution Phosphate buffered saline Preservative 0.09% Sodium Azide (NaN3) Stabilisers 2% Sucrose Immunogen Synthetic peptide directed towards the middle region of human CBX3 External Database Links UniProt: Q13185 Related reagents Entrez Gene: 11335 CBX3 Related reagents Specificity Rabbi anti Human CBX3 antibody recognizes chromobox protein homolog 3, also known as HP1 gamma homolog. Page 1 of 2 At the nuclear envelope, the nuclear lamina and heterochromatin are adjacent to the inner nuclear membrane. The protein encoded by CBX3 binds DNA and is a component of heterochromatin. This protein also can bind lamin B receptor, an integral membrane protein found in the inner nuclear membrane.
    [Show full text]
  • Supplemental Information Jmjd1c Is Required for MLL-AF9 and HOXA9
    Supplemental Information Jmjd1c is Required for MLL-AF9 and HOXA9 Mediated AML Stem Cell Self- Renewal Nan Zhu, Mo Chen, Rowena Eng, Joshua DeJong, Amit U. Sinha, Noushin F. Rahnamay, Richard Koche, Fatima Al-Shahrour, Janna C. Minehart, Chun-Wei Chen, Aniruddha J. Deshpande, Haiming Xu, S. Haihua Chu, Benjamin L. Ebert, Robert G. Roeder, Scott A. Armstrong Inventory of Supplemental Information Supplemental Figure Legends Supplemental Figures Figure S1. Related to Figure 1. Figure S2. Related to Figure 2&4. Figure S3. Related to Figure 3&5. Figure S4. Related to Figure 6. Figure S5. Related to Figure 7. Figure S6. Related to Figure 7. Supplemental Tables Table S1. Related to Figure 1. Table S2. Related to Figure 1. Table S3. Related to Figure S2 Table S4. Related to Figure 3. Table S5. Related to Figure 5. Table S6. Related to Figure 7. Figure S1 A B Number of Illumina reads C Log2(Median Fold Change Bone Marrow vs. T0) Spleen vs. T0) Spleen vs. (Median Fold Change (Median Fold 2 Relative R2=0.89 Log Row min Row max D HOXA9 HOXA10 MLL t (8;21) t (15;17) Adult Inv 16 M7 other T_MLL JMJD1C MEIS1 SENP6 TSC22D2 HIPK3 HOXA7 CDK6 RDX WNK1 PHIP CHD2 PDE3B -2.5 +3 RUNX2 Hoxa9 probe 1 Hoxa9 probe 2 Row relative Figure S1. In vivo shRNA screen of MLL-AF9 targets. Related to Figure 1. (A) Number of reads obtained from Illumina sequencing of eight experimental replicates. (B) Unsupervised hierarchical clustering of normalized reads of all hairpins in eight replicates. Hairpins are ranked by their depletion level in the bone marrow using log2 median fold- change.
    [Show full text]
  • Three-Dimensional Regulation of HOXA Cluster Genes by a Cis-Element in Hematopoietic Stem Cell and Leukemia
    bioRxiv preprint doi: https://doi.org/10.1101/2020.04.16.017533; this version posted April 18, 2020. The copyright holder for this preprint (which was not certified by peer review) is the author/funder, who has granted bioRxiv a license to display the preprint in perpetuity. It is made available under aCC-BY 4.0 International license. Three-dimensional regulation of HOXA cluster genes by a cis-element in hematopoietic stem cell and leukemia. Xue Qing David Wang1, Haley Gore1, Pamela Himadewi1, Fan Feng2, Lu Yang3, Wanding Zhou1,5, Yushuai Liu1, Xinyu Wang4, Chun-wei Chen3, Jianzhong Su4, Jie Liu2, Gerd Pfeifer1,*, Xiaotian Zhang1,* 1. Center for Epigenetics, Van Andel Research Institute, Grand Rapids, Michigan 2. Department of Computational Medicine and Bioinformatics, University of Michigan, Ann Arbor, Michigan. 3. Department of System Biology, City of Hope Cancer Center, California 4. Institute of Biomedical Big Data, Wenzhou Medical University, Wenzhou, China 5. Current address: Children Hospital of Philadelphia, University of Pennsylvania *. These authors jointly supervise this project Correspondence should be addressed to Xiaotian Zhang: [email protected] bioRxiv preprint doi: https://doi.org/10.1101/2020.04.16.017533; this version posted April 18, 2020. The copyright holder for this preprint (which was not certified by peer review) is the author/funder, who has granted bioRxiv a license to display the preprint in perpetuity. It is made available under aCC-BY 4.0 International license. Abstract: Proper gene regulation is crucial for cellular differentiation, and dysregulation of key genes can lead to diseased states such as cancer. The HOX transcription factors play such a role during hematopoiesis, and aberrant expression of certain HOXA genes is found in certain acute myeloid leukemias (AMLs).
    [Show full text]