Characterizing Novel Interactions of Transcriptional Repressor Proteins BCL6 & BCL6B

Total Page:16

File Type:pdf, Size:1020Kb

Characterizing Novel Interactions of Transcriptional Repressor Proteins BCL6 & BCL6B Characterizing Novel Interactions of Transcriptional Repressor Proteins BCL6 & BCL6B by Geoffrey Graham Lundell-Smith A thesis submitted in conformity with the requirements for the degree of Master of Science Department of Biochemistry University of Toronto © Copyright by Geoffrey Lundell-Smith, 2017 Characterizing Novel Interactions of Transcriptional Repression Proteins BCL6 and BCL6B Geoffrey Graham Lundell-Smith Masters of Science Department of Biochemistry University of Toronto 2016 Abstract B-cell Lymphoma 6 (BCL6) and its close homolog BCL6B encode proteins that are members of the BTB-Zinc Finger family of transcription factors. BCL6 plays an important role in regulating the differentiation and proliferation of B-cells during the adaptive immune response, and is also involved in T cell development and inflammation. BCL6 acts by repressing genes involved in DNA damage response during the affinity maturation of immunoglobulins, and the mis- expression of BCL6 can lead to diffuse large B-cell lymphoma. Although BCL6B shares high sequence similarity with BCL6, the functions of BCL6B are not well-characterized. I used BioID, an in vivo proximity-dependent labeling method, to identify novel BCL6 and BCL6B protein interactors and validated a number of these interactions with co-purification experiments. I also examined the evolutionary relationship between BCL6 and BCL6B and identified conserved residues in an important interaction interface that mediates corepressor binding and gene repression. ii Acknowledgments Thank you to my supervisor, Gil Privé for his mentorship, guidance, and advice, and for giving me the opportunity to work in his lab. Thanks to my committee members, Dr. John Rubinstein and Dr. Jeff Lee for their ideas, thoughts, and feedback during my Masters. Thank you to Dr. Etienne Coyaud for performing the mass spec experiments and providing help with data analysis, advice, and troubleshooting. An incredible amount of thanks to Neil Pomroy without whom much of this work would have been so much more difficult, if not nigh-impossible. Thanks also to Dr. Doug Kuntz, Dr. Wesley Errington, and Dr. Yong Wei, your guidance and advice has been invaluable. A big thank you to my fellow grad students in the lab, Andrew, Alan, Darren, Xiong, Yaseen, and the new guy, for the advice, support, thoughtful discussion, and allowing me free reign over the lab’s music playlist. I’d also like to thank the staff of the Department of Biochemistry, especially Carrie Harber, who works administrative wonders. Special thanks to my friends, you guys helped me through all the ups and downs, and all of the craziness. T follicular helper cells, right? Most importantly, to my mother and father, my sister Kierstin and my brother Connor, your support has meant the world to me. Thank you so much for being there. iii Table of Contents Contents Acknowledgments .......................................................................................................................... iii Table of Contents ........................................................................................................................... iv List of Abbreviations .................................................................................................................... vii List of Tables ................................................................................................................................. ix List of Figures ................................................................................................................................. x List of Appendices ........................................................................................................................ xii 1 Introduction ................................................................................................................................ 1 1.1 BTB Domains ..................................................................................................................... 1 1.2 BTB-Zinc finger proteins .................................................................................................... 1 1.3 General Mechanism of Transcriptional Repression ............................................................ 4 1.4 BCL6 ................................................................................................................................... 6 1.4.1 Introduction ............................................................................................................. 6 1.4.2 BCL6 Biological Roles ........................................................................................... 7 1.4.3 BCL6 and the Germinal Centre Reaction ............................................................... 8 1.4.4 BCL6 as Master Regulator of the Germinal Centre Reaction .............................. 10 1.4.5 BCL6 Regulation and Lymphomagenesis ............................................................ 11 1.5 BCL6B is a paralog of BCL6 ............................................................................................ 12 1.6 The Structure of the BCL6 BTB domain .......................................................................... 14 1.7 The BCL6 Interactome ..................................................................................................... 20 1.8 Protein-Protein Interactions .............................................................................................. 22 1.9 BioID ................................................................................................................................. 24 1.10 Thesis Objectives .............................................................................................................. 27 2 Materials and Methods ............................................................................................................. 28 iv 2.1 Cloning and Generation of Constructs .............................................................................. 28 2.2 SDS-PAGE and Western Blots ......................................................................................... 28 2.3 Mammalian Tissue Culture ............................................................................................... 29 2.4 Analysis of BioID Results ................................................................................................ 30 2.5 Transient Transfection and Co-Immunoprecipitation ....................................................... 30 2.6 Expression and Purification of BCL6BBTB ....................................................................... 31 2.7 Circular Dichroism of BCL6B BTB in Sarcosyl .............................................................. 32 2.8 Microscale Thermophoresis .............................................................................................. 32 2.9 Crystallization of BCL6BBTB ............................................................................................ 33 2.10 Phylogenetic Analysis and Sequence Conservation ......................................................... 33 3 Results ...................................................................................................................................... 35 3.1 BioID identifies known and novel interactors of BCL6 and BCL6B ............................... 35 3.2 Validation of Identified Interactions ................................................................................. 45 3.3 Purification of the BCL6B BTB domain .......................................................................... 46 3.3.1 Constructs and Purification Strategy .................................................................... 46 3.3.2 Evaluation of stability, solubility, and folding of BCL6BBTB in the presence of Sarcosyl ................................................................................................................. 49 3.3.3 Crystallization Trials ............................................................................................. 51 3.4 Biophysical Characterization of the interaction between BCL6B and SMRT ................. 52 3.5 Bioinformatic Analyses .................................................................................................... 55 3.5.1 Phylogenetic analysis of BCL6/B ......................................................................... 55 3.5.2 Conservation of Corepressor BBDs ...................................................................... 60 3.5.3 Conservation of the BCL6 and BCL6B lateral grooves ....................................... 60 3.5.4 Co-evolving Residues in the BTB Domain .......................................................... 64 4 Discussion ................................................................................................................................ 67 4.1 Experimental Rationale ..................................................................................................... 67 v 4.1.1 Choice of Technique ............................................................................................. 67 4.1.2 Identification of Statistically Significant Interactors ............................................ 70 4.1.3 Validation of Target Interactions: ......................................................................... 70 4.2 Interactions Identified by BioID ....................................................................................... 71 4.2.1 The BCL6 BTB domain interacts with TLE1 and TLE3 ...................................... 75 4.2.2
Recommended publications
  • Product Data Sheet
    For research purposes only, not for human use Product Data Sheet HIST1H4E siRNA (Human) Catalog # Source Reactivity Applications CRH5487 Synthetic H RNAi Description siRNA to inhibit HIST1H4E expression using RNA interference Specificity HIST1H4E siRNA (Human) is a target-specific 19-23 nt siRNA oligo duplexes designed to knock down gene expression. Form Lyophilized powder Gene Symbol HIST1H4E Alternative Names H4/E; H4FE; Histone H4 Entrez Gene 8367 (Human) SwissProt P62805 (Human) Purity > 97% Quality Control Oligonucleotide synthesis is monitored base by base through trityl analysis to ensure appropriate coupling efficiency. The oligo is subsequently purified by affinity-solid phase extraction. The annealed RNA duplex is further analyzed by mass spectrometry to verify the exact composition of the duplex. Each lot is compared to the previous lot by mass spectrometry to ensure maximum lot-to-lot consistency. Components We offers pre-designed sets of 3 different target-specific siRNA oligo duplexes of human HIST1H4E gene. Each vial contains 5 nmol of lyophilized siRNA. The duplexes can be transfected individually or pooled together to achieve knockdown of the target gene, which is most commonly assessed by qPCR or western blot. Our siRNA oligos are also chemically modified (2’-OMe) at no extra charge for increased stability and enhanced knockdown in vitro and in vivo. Directions for Use We recommends transfection with 100 nM siRNA 48 to 72 hours prior to cell lysis. Application key: E- ELISA, WB- Western blot, IH- Immunohistochemistry,
    [Show full text]
  • Supplementary Materials
    DEPs in osteosarcoma cells comparing to osteoblastic cells Biological Process Protein Percentage of Hits metabolic process (GO:0008152) 29.3 29.3% cellular process (GO:0009987) 20.2 20.2% localization (GO:0051179) 9.4 9.4% biological regulation (GO:0065007) 8 8.0% developmental process (GO:0032502) 7.8 7.8% response to stimulus (GO:0050896) 5.6 5.6% cellular component organization (GO:0071840) 5.6 5.6% multicellular organismal process (GO:0032501) 4.4 4.4% immune system process (GO:0002376) 4.2 4.2% biological adhesion (GO:0022610) 2.7 2.7% apoptotic process (GO:0006915) 1.6 1.6% reproduction (GO:0000003) 0.8 0.8% locomotion (GO:0040011) 0.4 0.4% cell killing (GO:0001906) 0.1 0.1% 100.1% Genes 2179Hits 3870 biological adhesion apoptotic process … reproduction (GO:0000003) , 0.8% (GO:0022610) , 2.7% locomotion (GO:0040011) ,… immune system process cell killing (GO:0001906) , 0.1% (GO:0002376) , 4.2% multicellular organismal process (GO:0032501) , metabolic process 4.4% (GO:0008152) , 29.3% cellular component organization (GO:0071840) , 5.6% response to stimulus (GO:0050896), 5.6% developmental process (GO:0032502) , 7.8% biological regulation (GO:0065007) , 8.0% cellular process (GO:0009987) , 20.2% localization (GO:0051179) , 9.
    [Show full text]
  • HIST1H3D: a Promising Therapeutic Target for Lung Cancer
    INTERNATIONAL JOURNAL OF ONCOLOGY 50: 815-822, 2017 HIST1H3D: A promising therapeutic target for lung cancer YAN RUI1*, WEN-JIA PENG2*, MING WANG1*, QIAN WANG1*, ZI-LI LIU1, YU-QING CHEN1 and LI-NIAN HUANG1 1Department of Respiration and Critical Care Medicine, The First Affiliated Hospital of Bengbu Medical College, Lung Cancer Diagnosis and Treatment Center of Anhui Province, Anhui Provincial Key Laboratory of Clinical Basic Research on Respiratory Disease, Bengbu, Anhui 233004; 2Department of Epidemiology and Health Statistics, Bengbu Medical College, Bengbu, Anhui 233000, P.R. China Received October 6, 2016; Accepted December 1, 2016 DOI: 10.3892/ijo.2017.3856 Abstract. HIST1H3D gene encodes histone H3.1 and is involved CDKN1 and CCNE2 genes. In conclusion, our results suggest in gene-silencing and heterochromatin formation. HIST1H3D that HIST1H3D is highly expressed in lung cancer cell lines expression is upregulated in primary gastric cancer tissue. In and tissues. Furthermore, HIST1H3D may be important in this study, we explored the effects of HIST1H3D expression cell proliferation, apoptosis and cell cycle progression, and is on lung cancer, and its mechanisms. HIST1H3D expression implicated as a potential therapeutic target for lung cancer. was measured by immunohistochemistry and RT-PCR in lung cancer tissues and human lung cancer cell lines. Cell prolif- Introduction eration was assessed by MTT assay. Flow cytometric analysis was used to determine cell cycle distribution and apoptosis. Lung cancer is one of the most common cancers and the Levels of related proteins were detected by western blotting. major cause of cancer deaths worldwide, with 1.6 million new Bioinformatics analysis was performed to investigate related lung cancer cases and 1.4 million lung cancer deaths each signaling pathways.
    [Show full text]
  • A Network Propagation Approach to Prioritize Long Tail Genes in Cancer
    bioRxiv preprint doi: https://doi.org/10.1101/2021.02.05.429983; this version posted February 8, 2021. 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. A Network Propagation Approach to Prioritize Long Tail Genes in Cancer Hussein Mohsen1,*, Vignesh Gunasekharan2, Tao Qing2, Sahand Negahban3, Zoltan Szallasi4, Lajos Pusztai2,*, Mark B. Gerstein1,5,6,3,* 1 Computational Biology & Bioinformatics Program, Yale University, New Haven, CT 06511, USA 2 Breast Medical Oncology, Yale School of Medicine, New Haven, CT 06511, USA 3 Department of Statistics & Data Science, Yale University, New Haven, CT 06511, USA 4 Children’s Hospital Informatics Program, Harvard-MIT Division of Health Sciences and Technology, Harvard Medical School, Boston, MA 02115, USA 5 Department of Molecular Biophysics and Biochemistry, Yale University, New Haven, CT 06511, USA 6 Department of Computer Science, Yale University, New Haven, CT 06511, USA * Corresponding author Abstract Introduction. The diversity of genomic alterations in cancer pose challenges to fully understanding the etiologies of the disease. Recent interest in infrequent mutations, in genes that reside in the “long tail” of the mutational distribution, uncovered new genes with significant implication in cancer development. The study of these genes often requires integrative approaches with multiple types of biological data. Network propagation methods have demonstrated high efficacy in uncovering genomic patterns underlying cancer using biological interaction networks. Yet, the majority of these analyses have focused their assessment on detecting known cancer genes or identifying altered subnetworks.
    [Show full text]
  • Contrasting Expression Patterns of Histone Mrna and Microrna 760 in Patients with Gastric Cancer
    Published OnlineFirst October 4, 2013; DOI: 10.1158/1078-0432.CCR-12-3186 Clinical Cancer Human Cancer Biology Research Contrasting Expression Patterns of Histone mRNA and microRNA 760 in Patients with Gastric Cancer Takeshi Iwaya1,2, Takeo Fukagawa3, Yutaka Suzuki4, Yusuke Takahashi1, Genta Sawada1, Masahisa Ishibashi1, Junji Kurashige1, Tomoya Sudo1, Fumiaki Tanaka1, Kohei Shibata1, Fumitaka Endo2, Hirokatsu Katagiri2, Kaoru Ishida2, Kohei Kume2, Satoshi Nishizuka2, Hisae Iinuma5, Go Wakabayashi2, Masaki Mori6, Mitsuru Sasako7, and Koshi Mimori1 Abstract Purpose: Recent studies revealed that both disseminated tumor cells and noncancerous cells contributed to cancer progression cooperatively in the bone marrow. Here, RNA-seq analysis of bone marrow from gastric cancer patients was performed to identify prognostic markers for gastric cancer. Experimental Design: Bone marrow samples from eight gastric cancer patients (stages I and IV: n ¼ 4 each) were used for RNA-seq analysis. Results were validated through quantitative real-time PCR (qRT-PCR) analysis of HIST1H3D expression in 175 bone marrow, 92 peripheral blood, and 115 primary tumor samples from gastric cancer patients. miR-760 expression was assayed using qRT-PCR in 105 bone marrow and 96 primary tumor samples. Luciferase reporter assays were performed to confirm whether histone mRNAs were direct targets of miR-760. miR-760 expression was also evaluated in noncancerous cells from gastric cancer patients. Results: RNA-seq analysis of bone marrow samples from gastric cancer patients revealed higher expression of multiple histone mRNAs in stage IV patients. HIST1H3D expression in the bone marrow, peripheral blood, and primary tumor of stage IV patients was higher than that in stage I patients (P ¼ 0.0284, 0.0243, and 0.0006, respectively).
    [Show full text]
  • 2007-05 TLE1 Synovial Sarcoma
    TLE1 Immunostains in the Diagnosis of Synovial Sarcoma May 2007 by Rodney T. Miller, M.D., Director of Immunohistochemistry The diagnosis of synovial sarcoma can be a chal- lenging task, particularly on small biopsy specimens, as the morphologic features of this tumor can be mimicked by a variety of other neoplasms. This month we call attention to a paper published in the February 2007 edition of the American Journal of Surgical Pathology describing the utility of immu- nostains for TLE1 in the diagnosis of this tumor. TLE immunostains are now available at ProPath. Synovial sarcoma occurs in three morphologic varie- ties: monophasic, biphasic, and poorly differentiated. It has been known for some time that synovial sar- coma is associated with a specific chromosomal H&E (left) and TLE1 immunostain (right) on a monophasic translocation, t(X;18), that results in the fusion of the synovial sarcoma. Note the numerous strongly positive nuclei SYT gene on chromosome 18 to either the SSX1 or on the TLE1 immunostain, a typical feature of this tumor. SSX2 gene on the X chromosome, resulting in the production of a SYT-SSX fusion protein. Identifica- In their study, the authors performed TLE1 immu- tion of this translocation in the appropriate setting is nostains on multiple tissue microarrays using two dif- regarded by many to be diagnostic of synovial sar- ferent antibodies (monoclonal and polyclonal), both coma. However, the methodologies used for this performing in a similar fashion. A total of 693 adult purpose (cytogenetics, fluorescent in situ hybridiza- soft tissue tumors were examined, including 94 cases tion, and reverse-transcriptase polymerase chain re- of synovial sarcoma that had documentation of the t action) are not readily available in many diagnostic (X;18) translocation.
    [Show full text]
  • Cancer TNT Ashwin Ram 12/5/2017 Background: Chromatin Writers, Readers, Erasers
    Cancer TNT Ashwin Ram 12/5/2017 Background: Chromatin Writers, Readers, Erasers writer effector eg. HAT, HMT reader eg. bromodomain eraser eg. HDAC, KDM The writer HAT1: A known H4 lysine 5,12 di-acetyltransferase writer siHAT1 siControl HAT1 H4 K12Ac H4 K5Ac actin Western blot for histone H4 modifications after control and HAT1 siRNA transfections. HAT1: EGF-stimulated immunoprecipitation specific specific - - HAT1 IgG HAT1 IgG - - R α non R α non EGF: + + + - - - WB: HAT1 Immunoprecipitation / WB to measure HAT1 levels +/- Heatmap of gene expression changes of all human histone EGF acetyltransferases +/- EGF and siRNA treatments shows HAT1 expression is EGF-dependent Working model of HAT1 : The oldest “new” histone acetyltransferase EGF EGFR plasma membrane HAT1 H4 H3 Rbap46/48 a nuclear membrane H4 H2A H3 H2B HAT1 S phase Surprise: HAT1 also binds (a few sites) on chromatin HAT1 ChipSeq signal sits on Hist1 locus on Chromosome 6 Read Depth HAT1 bound sites (zoom) HAT1 ChIP-seq peaks cluster at Read Depth histone H4 promoters. Hist1H2BE Hist1H4D Hist1H3D Hist1H4E Is HAT1 a transcription factor for its substrate (H4)? EGF EGFR plasma membrane HAT1 H4 H3 Rbap46/48 a nuclear membrane H4 H2A H3 H2B HAT1 S phase HAT1 is required for S-phase burst of histone H4 mRNA HAT1 Hist1H4B mRNA level 50 Rbap46 45 shCont-3 H4 40 shHAT1-A7 35 shHAT1-B6 30 25 B6 A7 - - 20 15 shHAT1 shHAT1 shControl 10 Gene actin)Expression (versus 5 HAT1 0 0 2 4 6 8 10 actin hours after release from double thymidine block G1 S G2/M HAT1 loss: Life with less histones EGF
    [Show full text]
  • Growth and Molecular Profile of Lung Cancer Cells Expressing Ectopic LKB1: Down-Regulation of the Phosphatidylinositol 3؅-Phosphate Kinase/PTEN Pathway1
    [CANCER RESEARCH 63, 1382–1388, March 15, 2003] Growth and Molecular Profile of Lung Cancer Cells Expressing Ectopic LKB1: Down-Regulation of the Phosphatidylinositol 3؅-Phosphate Kinase/PTEN Pathway1 Ana I. Jimenez, Paloma Fernandez, Orlando Dominguez, Ana Dopazo, and Montserrat Sanchez-Cespedes2 Molecular Pathology Program [A. I. J., P. F., M. S-C.], Genomics Unit [O. D.], and Microarray Analysis Unit [A. D.], Spanish National Cancer Center, 28029 Madrid, Spain ABSTRACT the cell cycle in G1 (8, 9). However, the intrinsic mechanism by which LKB1 activity is regulated in cells and how it leads to the suppression Germ-line mutations in LKB1 gene cause the Peutz-Jeghers syndrome of cell growth is still unknown. It has been proposed that growth (PJS), a genetic disease with increased risk of malignancies. Recently, suppression by LKB1 is mediated through p21 in a p53-dependent LKB1-inactivating mutations have been identified in one-third of sporadic lung adenocarcinomas, indicating that LKB1 gene inactivation is critical in mechanism (7). In addition, it has been observed that LKB1 binds to tumors other than those of the PJS syndrome. However, the in vivo brahma-related gene 1 protein (BRG1) and this interaction is required substrates of LKB1 and its role in cancer development have not been for BRG1-induced growth arrest (10). Similar to what happens in the completely elucidated. Here we show that overexpression of wild-type PJS, Lkb1 heterozygous knockout mice show gastrointestinal hamar- LKB1 protein in A549 lung adenocarcinomas cells leads to cell-growth tomatous polyposis and frequent hepatocellular carcinomas (11, 12). suppression. To examine changes in gene expression profiles subsequent to Interestingly, the hamartomas, but not the malignant tumors, arising in exogenous wild-type LKB1 in A549 cells, we used cDNA microarrays.
    [Show full text]
  • The Middle Temporal Gyrus Is Transcriptionally Altered in Patients with Alzheimer’S Disease
    1 The middle temporal gyrus is transcriptionally altered in patients with Alzheimer’s Disease. 2 1 3 Shahan Mamoor 1Thomas Jefferson School of Law 4 East Islip, NY 11730 [email protected] 5 6 We sought to understand, at the systems level and in an unbiased fashion, how gene 7 expression was most different in the brains of patients with Alzheimer’s Disease (AD) by mining published microarray datasets (1, 2). Comparing global gene expression profiles between 8 patient and control revealed that a set of 84 genes were expressed at significantly different levels in the middle temporal gyrus (MTG) of patients with Alzheimer’s Disease (1, 2). We used 9 computational analyses to classify these genes into known pathways and existing gene sets, 10 and to describe the major differences in the epigenetic marks at the genomic loci of these genes. While a portion of these genes is computationally cognizable as part of a set of genes 11 up-regulated in the brains of patients with AD (3), many other genes in the gene set identified here have not previously been studied in association with AD. Transcriptional repression, both 12 pre- and post-transcription appears to be affected; nearly 40% of these genes are transcriptional 13 targets of MicroRNA-19A/B (miR-19A/B), the zinc finger protein 10 (ZNF10), or of the AP-1 repressor jun dimerization protein 2 (JDP2). 14 15 16 17 18 19 20 21 22 23 24 25 26 Keywords: Alzheimer’s Disease, systems biology of Alzheimer’s Disease, differential gene 27 expression, middle temporal gyrus.
    [Show full text]
  • Triplet Repeat Length Bias and Variation in the Human Transcriptome
    Triplet repeat length bias and variation in the human transcriptome Michael Mollaa,1,2, Arthur Delcherb,1, Shamil Sunyaevc, Charles Cantora,d,2, and Simon Kasifa,e aDepartment of Biomedical Engineering and dCenter for Advanced Biotechnology, Boston University, Boston, MA 02215; bCenter for Bioinformatics and Computational Biology, University of Maryland, College Park, MD 20742; cDepartment of Medicine, Division of Genetics, Brigham and Women’s Hospital and Harvard Medical School, Boston, MA 02115; and eCenter for Advanced Genomic Technology, Boston University, Boston, MA 02215 Contributed by Charles Cantor, July 6, 2009 (sent for review May 4, 2009) Length variation in short tandem repeats (STRs) is an important family including Huntington’s disease (10) and hereditary ataxias (11, 12). of DNA polymorphisms with numerous applications in genetics, All Huntington’s patients exhibit an expanded number of copies in medicine, forensics, and evolutionary analysis. Several major diseases the CAG tandem repeat subsequence in the N terminus of the have been associated with length variation of trinucleotide (triplet) huntingtin gene. Moreover, an increase in the repeat length is repeats including Huntington’s disease, hereditary ataxias and spi- anti-correlated to the onset age of the disease (13). Multiple other nobulbar muscular atrophy. Using the reference human genome, we diseases have also been associated with copy number variation of have catalogued all triplet repeats in genic regions. This data revealed tandem repeats (8, 14). Researchers have hypothesized that inap- a bias in noncoding DNA repeat lengths. It also enabled a survey of propriate repeat variation in coding regions could result in toxicity, repeat-length polymorphisms (RLPs) in human genomes and a com- incorrect folding, or aggregation of a protein.
    [Show full text]
  • Identification of the Key Genes and Pathways in Prostate Cancer
    ONCOLOGY LETTERS 16: 6663-6669, 2018 Identification of the key genes and pathways in prostate cancer SHUTONG FAN1*, ZUMU LIANG1*, ZHIQIN GAO1, ZHIWEI PAN2, SHAOJIE HAN3, XIAOYING LIU1, CHUNLING ZHAO1, WEIWEI YANG1, ZHIFANG PAN1 and WEIGUO FENG1 1College of Bioscience and Technology, Weifang Medical University, Weifang, Shandong 261053; 2Department of Internal Medicine, Laizhou Development Zone Hospital, Yantai, Shandong 261400; 3Animal Epidemic Prevention and Epidemic Control Center, Changle County Bureau of Animal Health and Production, Weifang, Shandong 262400, P.R. China Received March 5, 2018; Accepted September 17, 2018 DOI: 10.3892/ol.2018.9491 Abstract. Prostate cancer (PCa) is one of the most common Introduction malignancies in men globally. The aim of the present study was to identify the key genes and pathways involved in the Prostate cancer (PCa) is one of the most common malignancies occurrence of PCa. Gene expression profile (GSE55945) in men globally and the second leading cause of cancer was downloaded from Gene Expression Omnibus, and associated mortality in developed countries (1,2). Like other the differentially expressed genes (DEGs) were identified. cancers, PCa is considered to be a disease which caused by Subsequently, Gene ontology analysis, KEGG pathway age, diet and gene aberrations (3). Accumulating evidences analysis and protein-protein interaction (PPI) analysis of have demonstrated that a series of genes and pathways involved DEGs were performed. Finally, the identified key genes were in the occurrence, progression and metastasis of PCa (4). At confirmed by immunohistochemistry. The GO analysis results present, the underlying mechanism of PCa occurrence is still showed that the DEGs were mainly participated in cell cycle, unclear, which limits the diagnosis and therapy.
    [Show full text]
  • Protein Purification Protein Localization in Vivo Fluorescent Imaging Protein Arrays Real Time Imaging Protein Interactions Protein Trafficking Protein Turnover
    Overcoming Challenges of Protein Analysis in Mammalian Systems Danette L. Daniels, Ph.D. Current Technologies for Protein Analysis Biochemical/ In Vivo Proteomic Cell Based Animal Analysis Analysis Models Fluorescent proteins Affinity tags Antibodies How about a system applicable to the all approaches that also addresses limitations of current methods? • Minimal interference with protein of interest • Efficient capture/isolation • Detection/real-time imaging • Differential labeling • High Signal/background HaloTag Platform Biochemical/ In Vivo Proteomic Cell Based Animal Analysis Analysis Models Protein purification Protein localization In vivo fluorescent imaging Protein arrays Real time imaging Protein interactions Protein trafficking Protein turnover HaloTag® HaloCHIP™ HaloLink™ HaloTag® Fluorescent Purification Protein:DNA Protein Arrays Pull-Down Ligands HaloTag is a Genetically Engineered Protein Fusion Tag O Functional Protein of Cl O Interest HT + group Protein of Functional HT O O Interest group . A monomeric , 34 kDa, modified bacterial dehalogenase genetically engineered to covalently bind specific, synthetic HaloTag® ligands . Irreversible, covalent attachment of chemical functionalities . Suitable as either N- or C- terminal fusion Mutagenized HaloTag® Protein Enables Covalent HaloTag®-Ligand Complex Hydrolase (DhaA) HaloTag® Catalytic process Facilitated bond formation T r p 1 0 7 T r p 1 0 7 HaloTag®: • 34kDa protein • Monomeric N N H N 4 1 H N A s n - H H • Single change: C l 4 1 A s n C l 2 1 His272Phe for covalent O R O O - C C bond. 3 R O 1 0 6 A s p 1 0 6 A s p O H O H Covalent bond: H H O O • Stable after N C G l u 1 3 0 C G l u 1 3 0 N - O denaturation.
    [Show full text]