Investigation of the Role of HMGB2 in Endocrine Resistant Breast Cancer

Total Page:16

File Type:pdf, Size:1020Kb

Investigation of the Role of HMGB2 in Endocrine Resistant Breast Cancer Investigation of the role of HMGB2 in endocrine resistant breast cancer Katie O’ Brien B.Sc., M.Sc. Waterford Institute of Technology Supervisors: Dr. Leonie Young (RCSI), Dr. Orla O’Donovan (W.I.T.) Thesis submitted to Waterford Institute of Technology for the award of M.Sc. June 2012 I declare that this thesis, which I submit to RCSI and W.I.T for consideration of the award of M.Sc. is my own personal effort. Where any of the content presented is the result of input from any other source other than my own work this is duly acknowledged in the text. Furthermore, I took reasonable care to ensure that the work is original, and, to the best of my knowledge, does not breach copyright law, and has not been taken from other sources except where such work has been cited and acknowledged within the text. Signed Student Number: W20042831 Date: 24 June 2012 Acknowledgements There are many people to whom I am incredibly grateful to for their guidance, support and their friendship during the course of this research M.Sc. This project would never have been possible without the funding provided by the Council of Directors, under the Strand III Technological Sector Research Funding, which is gratefully acknowledged. I would also like to thank Dr. Leonie Young in the Royal College of Surgeons, Ireland. Dr. Young has shared with me her extensive scientific knowledge and has provided me with constant mentorship throughout the course of this research. I must sincerely thank Dr. Aisling Redmond who has patiently given her time in assisting me with the research, making a significant contribution to the bulk of this work. Her patience in explaining experimental techniques and her corrections for this thesis are very much appreciated. Dr. Damian McCartan also made a significant contribution to this research and is acknowledged in the results chapters accordingly. I was fortunate to be surrounded by an amazing research team in the Royal College of Surgeons (endocrine oncology lab). I am grateful to Fiona Bane for her support and her friendship during my time in RCSI. Damir Varešlija and Sinead Cocchiglia have both been a great support and good friends to me in the lab. Also Eamon Hughes has to be mentioned for making me laugh every single day; which is often needed during research. I am grateful to Paul Tibbitts for assisting me with the immunhistochemistry. Dr. Christopher Byrne was a constant source of scientific knowledge and provided me with many entertaining stories. I also have to thank Dr. Marie McIlroy and Dr. Jean McBryan for their advice throughout the research project; two women I admire very much. Finally, I am sincerely thankful for the support and encouragement of my parents, Marie and Michael O’ Brien and Martin Collier. Table of Contents Table of Contents i Table of Figures iv List of Tables v Abbreviations vi Abstract ix 1. General Introduction 1 1.1 Introduction to breast cancer 1.1.1 Breast cancer epidemiology 2 1.1.2 Genetic susceptibility to breast cancer 3 1.1.3 Molecular classification of breast cancer 4 1.2 Hormone signalling in breast cancer 1.2.1 The Estrogen Receptor α 6 1.2.2 Endocrine therapies for breast cancer 10 1.2.3 Resistance to endocrine therapies 13 1.2.4 SRC1 role in endocrine resistance 14 1.3 High Mobility Group Box 2 protein 16 1.4 Breast Cancer Amplified Sequence 3 gene 17 1.5 Hypothesis 19 1.6 Aims 20 2. Materials and Methods 21 2.1 Cell Culture 2.1.1 Breast cancer cell lines 22 2.1.2 Cell culture environment 23 2.1.3 Passaging of cells 23 2.1.4 Culturing of cells from cryo-storage ` 23 2.1.5 Cell treatments 23 2.1.6 Cell counting 24 2.2 Protein Biochemistry 2.2.1 Protein extraction 24 2.2.2 Protein quantification 25 2.2.3 Coimmunoprecipitation 25 2.2.4 SDS-PAGE and western blotting 26 i 2.2.5 Mass Spectrometry 29 2.2.6 Chromatin Immunoprecipitation 30 2.3 Nucleic Acid Biochemistry 2.3.1 PCR 32 2.3.2 Real-Time PCR 34 2.4 Transfections 2.4.1 Gene silencing 36 2.4.2 Protein over-expression 37 2.5 Immunohistochemistry 2.5.1 Biological samples 38 2.5.2 Staining protocol 38 2.5.3 Immunohistochemistry scoring system 40 2.6 3D Mammosphere Assay 40 3. Investigation of HMGB2 in the ER/SRC1 complex in endocrine resistant breast cancer 42 3.1 Introduction 43 3.2 Results 3.2.1 ER α knockdown in MCF7 and LY2 cells prior to 3D culture assay 45 3.2.2 3D Culture Assay in MCF7 and LY2 cells with siRNA ER α 46 3.2.3 Mass Spectrometry 47 3.2.4 HMGB2 expression in the MCF7 and LY2 cell lines 49 3.2.5 Tamoxifen driven HMGB2 regulates the expression of SRC1 in LY2 cells 50 3.2.6 ER α/ SRC1 interactions following HMGB2 knock down in LY2 cells 51 3.2.7 HMGB2 Immunohistochemistry 53 3.3 Discussion 55 4. HMGB2 regulates the expression of estrogen responsive genes in endocrine resistant breast cancer 57 4.1 Introduction 58 4.2 Results 4.2.1 HMGB2 regulates the expression of cMyc in LY2 cell line 60 4.2.2 SRC1 ChIP-Sequencing 61 4.2.3 SRC1 regulates the expression of BCAS3 in LY2 cell line 62 ii 4.2.4 HMGB2 regulates the expression of BCAS3 in LY2 cell line 63 4.2.5 ER α regulates the expression of BCAS3 in LY2 cell line 64 4.2.6 HMGB2 recruitment to the BCAS3 promoter via PCR in the LY2 cell line 65 4.2.7 HMGB2 recruitment to the BCAS3 promoter via Real-Time PCR in the LY2 cell line 66 4.2.8 BCAS3 regulates the expression of ER α in LY2 cell line 67 4.3 Discussion 68 5. GENERAL DISCUSSION 71 5.1 HMGB2 acts as a facilitator in ER α/SRC1 interactions 73 5.2 HMGB2 regulates protein expression of BCAS3 75 5.3 Conclusion 78 6. REFERENCES 79 7. APPENDIX 89 iii Table of Figures 1.1 The average number of newly diagnosed breast cancer cases from 2004-2007 3 1.2 Location of luminal epithelial cells on the inner lining of the mammary duct 5 1.3 Basic structure of ER α 7 1.4 Classical and non-classical regulation of estrogen responsive genes 9 1.5 Endocrine therapies for breast cancer 12 1.6 Cruciform structures in chromosomal region of 17q23 18 2.1 Schematic representation of the method of 3D cell culture on Matrigel 41 3.1 ER α knock-down in MCF7 and LY2 cell lines 45 3.2 3D culture assay following ER α knock-down in MCF7 and LY2 cell lines 46 3.3 SDS Page gel from which proteins were selected for mass spectrometry 48 3.4 Expression of HMGB2 in MCF7 and LY2 cell lines 49 3.5 Expression of SRC1 in LY2 cell line following HMGB2 knock-down 50 3.6 ER α/SRC1 interaction following HMGB2 knock-down in LY2 cell line 52 3.7 HMGB2 immunohistochemistry and Kaplan-Meier survival curve 53 4.1 c-Myc expression following HMGB2 knock-down in LY2 cell line 60 4.2 SRC1 ChIP sequencing in LY2 cell line 61 4.3 BCAS3 expression following SRC1 knock-down and over-expression in LY2 62 cell line 4.4 BCAS3 expression following HMGB2 knock-down in LY2 cell line 63 4.5 BCAS3 expression following ER α knock-down in LY2 cell line 64 4.6 HMGB2 recruitment to the BCAS3 promoter in the LY2 cell line 65 4.7 Q-PCR of HMGB2 ChIP to BCAS3 promoter in the LY2 cell line 66 4.8 ER α expression following BCAS3 knock-down in the LY2 cell line 67 iv List of Tables 2.1 Gel preparation for SDS-PAGE 27 2.2 Conditions used for western blotting for proteins studied 28 2.3 PCR reaction reagents and composition 32 2.4 Primers used 33 2.5 Cycling conditions for PCR reactions used 34 2.6 Calculation of fold enrichment of ChIP samples on qPCR 35 2.7 Run conditions used for qPCR reactions 36 2.8 Conditions used for immunohistochemistry 39 v Abbreviations ABC Avidin-biotin complex AD Activation domain AF-1 and AF-2 Activation function domains AI Aromatase inhibitor AIB1 Amplified-in-breast cancer 1 AP-1 Activating protein BCA Bichinchonic Assay bFGF Basic fibroblast growth factor bp Base pairs BCAS3 Breast Cancer Amplified Sequence 3 gene BRCA Breast cancer susceptibility gene BSA Bovine serum albumin CARM1 Coactivator-associated arginine methyltransferase 1 CBP CREB-binding protein Cdk1 Cyclin-dependent kinase 1 cDNA Complementary DNA CDS-FCS Charcoal dextran-stripped fetal calf serum ChIP Chromatin Immunoprecipitation Co-IP Co-Immunoprecipitation DAB 3,3-diaminobenzidine tetrahydrochloride DAPI 4’,6-diamidino-2-phenylindole dihydrochloride DBD DNA binding domain DCIS Ductal carcinoma in situ dH 2O Distilled water DFS Disease-free survival DMEM Dulbecco’s modified Eagle’s medium DMSO Dimethyl sulfoxide DNA Deoxyribonucleic acid dNTP Deoxyribonucleotide triphosphate E2 17 β-estradiol ECL Enhanced chemiluminescent reagent vi EGF Epidermal growth factor EGFR Epidermal growth factor receptor ER Estrogen receptor Erb Erythroblastic Leukemia Viral Oncogene ERE Estrogen response elements ERK Extracellular signal-regulated kinase HAT Histone acetyltransferases HDAC Histone deacetylases HER-2 Human epidermal growth factor receptor 2 HMGB2 High Mobility Group Box 2 gene HRP Horseradish peroxidase IGF-1 Insulin-like growth factor 1 IMS Industrial methylated spirits JNK C-Jun NH2-terminal kinase LiCl Lithium chloride MAPK Mitogen-activated protein kinase MEM Minimal essential medium MgCl 2 Magnesium chloride MMP Matrix metalloproteinases MTA1 Metastasis associated protein 1 NCoA Nuclear co-activator NCoR Nuclear co-repressor PCR Polymerase Chain Reaction PELP1 Proline glutamic acid leucine rich protein 1 PI3K Phosphatidylinositol-3 kinase PR Progesterone receptor qPCR Quantitative PCR RIPA Radioimmunoprecipitation RNA Ribonucleic acid RT-PCR Reverse transcriptase polymerase chain reaction SDS-PAGE Sodium dodecyl sulphate-polyacrylamide gel electrophoresis Ser Serine SERM Selective estrogen receptor modulator SMRT Silencing mediator of retinoic acid and thyroid hormone receptor vii SNP Single nucleotide polymorphisms SRC Steroid receptor coactivators TAE Tris acetate-EDTA Taq Taq polymerase (Thermophilus aquaticus ) TBS Tris buffered saline TBS-T Tris buffered saline plus 1% Tween TMA Tissue microarray TNF α Tumour necrosis factor-α UV Ultraviolet v/v Volume/volume w/v Weight/volume viii Investigation of the role of HMGB2 in endocrine resistant breast cancer Abstract The high mobility group box 2 protein (HMGB2) is an abundant chromatin remodelling protein with an affinity for unusual DNA structures.
Recommended publications
  • Molecular Profile of Tumor-Specific CD8+ T Cell Hypofunction in a Transplantable Murine Cancer Model
    Downloaded from http://www.jimmunol.org/ by guest on September 25, 2021 T + is online at: average * The Journal of Immunology , 34 of which you can access for free at: 2016; 197:1477-1488; Prepublished online 1 July from submission to initial decision 4 weeks from acceptance to publication 2016; doi: 10.4049/jimmunol.1600589 http://www.jimmunol.org/content/197/4/1477 Molecular Profile of Tumor-Specific CD8 Cell Hypofunction in a Transplantable Murine Cancer Model Katherine A. Waugh, Sonia M. Leach, Brandon L. Moore, Tullia C. Bruno, Jonathan D. Buhrman and Jill E. Slansky J Immunol cites 95 articles Submit online. Every submission reviewed by practicing scientists ? is published twice each month by Receive free email-alerts when new articles cite this article. Sign up at: http://jimmunol.org/alerts http://jimmunol.org/subscription Submit copyright permission requests at: http://www.aai.org/About/Publications/JI/copyright.html http://www.jimmunol.org/content/suppl/2016/07/01/jimmunol.160058 9.DCSupplemental This article http://www.jimmunol.org/content/197/4/1477.full#ref-list-1 Information about subscribing to The JI No Triage! Fast Publication! Rapid Reviews! 30 days* Why • • • Material References Permissions Email Alerts Subscription Supplementary The Journal of Immunology The American Association of Immunologists, Inc., 1451 Rockville Pike, Suite 650, Rockville, MD 20852 Copyright © 2016 by The American Association of Immunologists, Inc. All rights reserved. Print ISSN: 0022-1767 Online ISSN: 1550-6606. This information is current as of September 25, 2021. The Journal of Immunology Molecular Profile of Tumor-Specific CD8+ T Cell Hypofunction in a Transplantable Murine Cancer Model Katherine A.
    [Show full text]
  • Genomic Interaction Between ER and HMGB2 Identifies DDX18 As A
    Oncogene (2015) 34, 3871–3880 © 2015 Macmillan Publishers Limited All rights reserved 0950-9232/15 www.nature.com/onc ORIGINAL ARTICLE Genomic interaction between ER and HMGB2 identifies DDX18 as a novel driver of endocrine resistance in breast cancer cells AM Redmond1,2, C Byrne1, FT Bane1, GD Brown2, P Tibbitts1,KO’Brien1, ADK Hill1, JS Carroll2 and LS Young1 Breast cancer resistance to endocrine therapies such as tamoxifen and aromatase inhibitors is a significant clinical problem. Steroid receptor coactivator-1 (SRC-1), a coregulatory protein of the oestrogen receptor (ER), has previously been shown to have a significant role in the progression of breast cancer. The chromatin protein high mobility group box 2 (HMGB2) was identified as an SRC-1 interacting protein in the endocrine-resistant setting. We investigated the expression of HMGB2 in a cohort of 1068 breast cancer patients and found an association with increased disease-free survival time in patients treated with endocrine therapy. However, it was also verified that HMGB2 expression could be switched on in endocrine-resistant tumours from breast cancer patients. To explore the function of this poorly characterized protein, we performed HMGB2 ChIPseq and found distinct binding patterns between the two contexts. In the resistant setting, the HMGB2, SRC-1 and ER complex are enriched at promoter regions of target genes, with bioinformatic analysis indicating a switch in binding partners between the sensitive and resistant phenotypes. Integration of binding and gene expression data reveals a concise set of target genes of this complex including the RNA helicase DDX18. Modulation of DDX18 directly affects growth of tamoxifen-resistant cells, suggesting that it may be a critical downstream effector of the HMGB2:ER complex.
    [Show full text]
  • Itraq‑Based Proteomic Analysis of Endotoxin Tolerance Induced by Lipopolysaccharide
    584 MOLECULAR MEDICINE REPORTS 20: 584-592, 2019 iTRAQ‑based proteomic analysis of endotoxin tolerance induced by lipopolysaccharide QIAN ZHANG1, YINGCHUN HU2, JING ZHANG1 and CUNLIANG DENG1 Departments of 1Infectious Diseases and 2Emergency, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan 646000, P.R. China Received August 25, 2018; Accepted February 15, 2019 DOI: 10.3892/mmr.2019.10264 Abstract. The purpose of the present study was to investigate Introduction the differentially expressed proteins between endotoxin toler- ance and sepsis. Cell models of an endotoxin tolerance group Sepsis is a serious healthcare concern due to its high costs (ET group) and sepsis group [lipopolysaccharide (LPS) group] and mortality rate (1). It is triggered mainly by Gram-negative were established using LPS and evaluated using ELISA and bacteria (2) and lipopolysaccharides (LPS) are the main flow cytometry methods. Differentially expressed proteins component of the outer membrane of Gram-negative bacteria. between the ET and the LPS groups were identified using Previous studies have demonstrated that LPS serve an important isobaric tags for relative and absolute quantitation (iTRAQ) role in sepsis, and can stimulate the mononuclear phagocytic analysis and evaluated by bioinformatics analysis. The expres- system to produce a large number of inflammatory factors sion of core proteins was detected by western blotting. It was and cause systemic inflammatory response, sepsis, infectious identified that the expression of tumor necrosis factor‑α and shock and multiple organ dysfunctions (3). It has been reported interleukin-6 was significantly decreased in the ET group that following low-dose LPS stimulation, the host can survive a compared with the LPS group.
    [Show full text]
  • HMGB2 Polyclonal Antibody Catalog Number:14597-1-AP Featured Product 8 Publications
    For Research Use Only HMGB2 Polyclonal antibody Catalog Number:14597-1-AP Featured Product 8 Publications www.ptglab.com Catalog Number: GenBank Accession Number: Purification Method: Basic Information 14597-1-AP BC001063 Antigen affinity purification Size: GeneID (NCBI): Recommended Dilutions: 150ul , Concentration: 900 μg/ml by 3148 WB 1:500-1:3000 Nanodrop and 400 μg/ml by Bradford Full Name: IP 0.5-4.0 ug for IP and 1:500-1:1000 method using BSA as the standard; high-mobility group box 2 for WB Source: IHC 1:50-1:500 Calculated MW: IF 1:50-1:500 Rabbit 24 kDa Isotype: Observed MW: IgG 33-35 kDa Immunogen Catalog Number: AG6135 Applications Tested Applications: Positive Controls: IF, IHC, IP, WB,ELISA WB : Jurkat cells, HEK-293 cells, HL-60 cells, human Cited Applications: cerebellum tissue, K-562 cells WB IP : HEK-293 cells, Species Specificity: IHC : mouse brain tissue, human, mouse, rat Cited Species: IF : HepG2 cells, human, mouse, rat Note-IHC: suggested antigen retrieval with TE buffer pH 9.0; (*) Alternatively, antigen retrieval may be performed with citrate buffer pH 6.0 High mobility group protein B2 (HMGB2) belongs to a family of highly conserved proteins that contain HMG box Background Information domains (11246022,14871457). All three family members (HMGB1, HMGB2, and HMGB3) contain two HMG box domains and a C-terminal acidic domain. HMGB1 is a widely expressed and highly abundant protein (14871457). HMGB2 is widely expressed during embryonic development, but it is restricted to lymphoid organs and testis in adult animals (11262228). HMGB3 is only expressed during embryogenesis (9598312).
    [Show full text]
  • The Genetic Program of Pancreatic Beta-Cell Replication in Vivo
    Page 1 of 65 Diabetes The genetic program of pancreatic beta-cell replication in vivo Agnes Klochendler1, Inbal Caspi2, Noa Corem1, Maya Moran3, Oriel Friedlich1, Sharona Elgavish4, Yuval Nevo4, Aharon Helman1, Benjamin Glaser5, Amir Eden3, Shalev Itzkovitz2, Yuval Dor1,* 1Department of Developmental Biology and Cancer Research, The Institute for Medical Research Israel-Canada, The Hebrew University-Hadassah Medical School, Jerusalem 91120, Israel 2Department of Molecular Cell Biology, Weizmann Institute of Science, Rehovot, Israel. 3Department of Cell and Developmental Biology, The Silberman Institute of Life Sciences, The Hebrew University of Jerusalem, Jerusalem 91904, Israel 4Info-CORE, Bioinformatics Unit of the I-CORE Computation Center, The Hebrew University and Hadassah, The Institute for Medical Research Israel- Canada, The Hebrew University-Hadassah Medical School, Jerusalem 91120, Israel 5Endocrinology and Metabolism Service, Department of Internal Medicine, Hadassah-Hebrew University Medical Center, Jerusalem 91120, Israel *Correspondence: [email protected] Running title: The genetic program of pancreatic β-cell replication 1 Diabetes Publish Ahead of Print, published online March 18, 2016 Diabetes Page 2 of 65 Abstract The molecular program underlying infrequent replication of pancreatic beta- cells remains largely inaccessible. Using transgenic mice expressing GFP in cycling cells we sorted live, replicating beta-cells and determined their transcriptome. Replicating beta-cells upregulate hundreds of proliferation- related genes, along with many novel putative cell cycle components. Strikingly, genes involved in beta-cell functions, namely glucose sensing and insulin secretion were repressed. Further studies using single molecule RNA in situ hybridization revealed that in fact, replicating beta-cells double the amount of RNA for most genes, but this upregulation excludes genes involved in beta-cell function.
    [Show full text]
  • Full Text (PDF)
    Published OnlineFirst March 6, 2018; DOI: 10.1158/2159-8290.CD-16-0861 RESEARCH ARTICLE Cross-Cohort Analysis Identifi es a TEAD4–MYCN Positive Feedback Loop as the Core Regulatory Element of High-Risk Neuroblastoma Presha Rajbhandari 1 , 2 , Gonzalo Lopez 1 , 3 , Claudia Capdevila 1 , Beatrice Salvatori 1 , Jiyang Yu 1 , 4 , Ruth Rodriguez-Barrueco5 , 6 , Daniel Martinez 3 , Mark Yarmarkovich 3 , Nina Weichert-Leahey 7 , Brian J. Abraham 8 , Mariano J. Alvarez 1 , Archana Iyer 1 , Jo Lynne Harenza 3 , Derek Oldridge 3 , Katleen De Preter9 , Jan Koster 10 , Shahab Asgharzadeh 11 , 12 , Robert C. Seeger 11 , 12 , Jun S. Wei 13 , Javed Khan13 , Jo Vandesompele 9 , Pieter Mestdagh 9 , Rogier Versteeg 10 , A. Thomas Look 7 , Richard A. Young8 , 14 , Antonio Iavarone 15 , Anna Lasorella 16 , Jose M. Silva 5 , John M. Maris 3 , 17 , 18 , and Andrea Califano1 , 19 , 20 , 21 ABSTRACT High-risk neuroblastomas show a paucity of recurrent somatic mutations at diag- nosis. As a result, the molecular basis for this aggressive phenotype remains elu- sive. Recent progress in regulatory network analysis helped us elucidate disease-driving mechanisms downstream of genomic alterations, including recurrent chromosomal alterations. Our analysis identi- fi ed three molecular subtypes of high-risk neuroblastomas, consistent with chromosomal alterations, and identifi ed subtype-specifi c master regulator proteins that were conserved across independent cohorts. A 10-protein transcriptional module—centered around a TEAD4–MYCN positive feedback loop—emerged as the regulatory driver of the high-risk subtype associated with MYCN amplifi cation. Silencing of either gene collapsed MYCN -amplifi ed MYCN( Amp ) neuroblastoma transcriptional hall- marks and abrogated viability in vitro and in vivo .
    [Show full text]
  • Identification of MAZ As a Novel Transcription Factor Regulating Erythropoiesis
    Identification of MAZ as a novel transcription factor regulating erythropoiesis Darya Deen1, Falk Butter2, Michelle L. Holland3, Vasiliki Samara4, Jacqueline A. Sloane-Stanley4, Helena Ayyub4, Matthias Mann5, David Garrick4,6,7 and Douglas Vernimmen1,7 1 The Roslin Institute and Royal (Dick) School of Veterinary Studies, University of Edinburgh, Easter Bush, Midlothian EH25 9RG, United Kingdom. 2 Institute of Molecular Biology (IMB), 55128 Mainz, Germany 3 Department of Medical and Molecular Genetics, School of Basic and Medical Biosciences, King's College London, London SE1 9RT, UK 4 MRC Molecular Haematology Unit, Weatherall Institute for Molecular Medicine, University of Oxford, John Radcliffe Hospital, Oxford OX3 9DS, United Kingdom. 5 Department of Proteomics and Signal Transduction, Max Planck Institute of Biochemistry, 82152 Martinsried, Germany. 6 Current address : INSERM UMRS-976, Institut de Recherche Saint Louis, Université de Paris, 75010 Paris, France. 7 These authors contributed equally Correspondence: [email protected], [email protected] ADDITIONAL MATERIAL Suppl Fig 1. Erythroid-specific hypersensitivity regions DNAse-Seq. (A) Structure of the α-globin locus. Chromosomal position (hg38 genome build) is shown above the genes. The locus consists of the embryonic ζ gene (HBZ), the duplicated foetal/adult α genes (HBA2 and HBA1) together with two flanking pseudogenes (HBM and HBQ1). The upstream, widely-expressed gene, NPRL3 is transcribed from the opposite strand to that of the HBA genes and contains the remote regulatory elements of the α-globin locus (MCS -R1 to -R3) (indicated by red dots). Below is shown the ATAC-seq signal in human erythroblasts (taken from 8) indicating open chromatin structure at promoter and distal regulatory elements (shaded regions).
    [Show full text]
  • Single-Cell RNA Landscape of Intratumoral Heterogeneity and Immunosuppressive Microenvironment in Advanced Osteosarcoma
    ARTICLE https://doi.org/10.1038/s41467-020-20059-6 OPEN Single-cell RNA landscape of intratumoral heterogeneity and immunosuppressive microenvironment in advanced osteosarcoma Yan Zhou1,11, Dong Yang2,11, Qingcheng Yang2,11, Xiaobin Lv 3,11, Wentao Huang4,11, Zhenhua Zhou5, Yaling Wang 1, Zhichang Zhang2, Ting Yuan2, Xiaomin Ding1, Lina Tang 1, Jianjun Zhang1, Junyi Yin1, Yujing Huang1, Wenxi Yu1, Yonggang Wang1, Chenliang Zhou1, Yang Su1, Aina He1, Yuanjue Sun1, Zan Shen1, ✉ ✉ ✉ ✉ Binzhi Qian 6, Wei Meng7,8, Jia Fei9, Yang Yao1 , Xinghua Pan 7,8 , Peizhan Chen 10 & Haiyan Hu1 1234567890():,; Osteosarcoma is the most frequent primary bone tumor with poor prognosis. Through RNA- sequencing of 100,987 individual cells from 7 primary, 2 recurrent, and 2 lung metastatic osteosarcoma lesions, 11 major cell clusters are identified based on unbiased clustering of gene expression profiles and canonical markers. The transcriptomic properties, regulators and dynamics of osteosarcoma malignant cells together with their tumor microenvironment particularly stromal and immune cells are characterized. The transdifferentiation of malignant osteoblastic cells from malignant chondroblastic cells is revealed by analyses of inferred copy-number variation and trajectory. A proinflammatory FABP4+ macrophages infiltration is noticed in lung metastatic osteosarcoma lesions. Lower osteoclasts infiltration is observed in chondroblastic, recurrent and lung metastatic osteosarcoma lesions compared to primary osteoblastic osteosarcoma lesions. Importantly, TIGIT blockade enhances the cytotoxicity effects of the primary CD3+ T cells with high proportion of TIGIT+ cells against osteo- sarcoma. These results present a single-cell atlas, explore intratumor heterogeneity, and provide potential therapeutic targets for osteosarcoma. 1 Oncology Department of Shanghai Jiao Tong University Affiliated Sixth People’s Hospital, Shanghai 200233, China.
    [Show full text]
  • Manuscript S4.0N Supplementary Material
    Integrative genomic and epigenomic analyses identified IRAK1 as a novel target for chronic inflammation-driven prostate tumorigenesis Saheed Oluwasina Oseni1,*, Olayinka Adebayo2, Adeyinka Adebayo3, Alexander Kwakye4, Mirjana Pavlovic5, Waseem Asghar5, James Hartmann1, Gregg B. Fields6, and James Kumi-Diaka1 Affiliations: 1 Department of Biological Sciences, Florida Atlantic University, Florida, USA 2 Morehouse School of Medicine, Atlanta, Georgia, USA 3 Georgia Institute of Technology, Atlanta, Georgia, USA 4 College of Medicine, Florida Atlantic University, Florida, USA 5 Department of Computer and Electrical Engineering, Florida Atlantic University, Florida, USA 6 Department of Chemistry & Biochemistry and I-HEALTH, Florida Atlantic University, Florida, USA Corresponding Author: [email protected] (S.O.O) List of Supplementary Materials Supplementary Method S1: Preparing genomic and clinical data for biological network analysis Supplementary Method S2: WGCNA R packages for biological network analysis Supplementary Table S1: Enrichr gene enrichment analysis of the 10 inflammatory genes in the magenta module based on a significant p-value. Supplementary Table S2: Functional impact ranking rubrics for IRAK1 gene mutations in PRAD samples using algorithms from different tools. Supplementary Figure S1: Batch effect correction plots for the PRAD dataset. Supplementary Figure S2: Hierarchical sample cluster heatmap of PRAD eigengene network. Supplementary Figure S3: Hierarchical gene expression cluster heatmap of PRAD eigengene network. Supplementary Figure S4: Kaplan-Meier DFS survival analysis for high-expressing vs low-expressing PRAD samples. Supplementary Figure S5: Receiver Operating Characteristic (ROC) analysis for IRAK in predicting cancer relapse. Supplementary Figure S6: Heatmap of the expression profile of IRAK family genes in indolent PRAD patients (n = 472) showing the overexpression of IRAK1 compared to other IRAKs.
    [Show full text]
  • NATURE Immunology
    RESOURCE Identification of transcriptional regulators in the mouse immune system Vladimir Jojic1,7, Tal Shay2,7, Katelyn Sylvia3, Or Zuk2, Xin Sun4, Joonsoo Kang3, Aviv Regev2,5,8, Daphne Koller1,8 & the Immunological Genome Project Consortium6 The differentiation of hematopoietic stem cells into cells of the immune system has been studied extensively in mammals, but the transcriptional circuitry that controls it is still only partially understood. Here, the Immunological Genome Project gene-expression profiles across mouse immune lineages allowed us to systematically analyze these circuits. To analyze this data set we developed Ontogenet, an algorithm for reconstructing lineage-specific regulation from gene-expression profiles across lineages. Using Ontogenet, we found differentiation stage–specific regulators of mouse hematopoiesis and identified many known hematopoietic regulators and 175 previously unknown candidate regulators, as well as their target genes and the cell types in which they act. Among the previously unknown regulators, we emphasize the role of ETV5 in the differentiation of gd T cells. As the transcriptional programs of human and mouse cells are highly conserved, it is likely that many lessons learned from the mouse model apply to humans. The Immunological Genome Project (ImmGen) is a consortium of ­programs of human and mouse cells are highly conserved4, many immunologists and computational biologists who aim, through the lessons learned from the mouse model will probably be applicable use of shared and rigorously controlled data-generation pipelines, to humans. Two key approaches for the identification of regulatory to exhaustively chart gene-expression profiles and their underlying networks5 are physical models based on the association of a transcrip- regulatory networks in the mouse immune system1.
    [Show full text]
  • Nonhistone Proteins HMGB1 and HMGB2 Differentially Modulate The
    biomolecules Article Nonhistone Proteins HMGB1 and HMGB2 Differentially Modulate the Response of Human Embryonic Stem Cells and the Progenitor Cells to the Anticancer Drug Etoposide Alireza Jian Bagherpoor y, Martin Kuˇcírek, Radek Fedr, Soodabeh Abbasi Sani and Michal Štros * Institute of Biophysics of the Czech Academy of Sciences, Královopolská 135, 612 00 Brno, Czech Republic; [email protected] (A.J.B.); [email protected] (M.K.); [email protected] (R.F.); [email protected] (S.A.S.) * Correspondence: [email protected]; Tel.: +420-541517260 Current affiliation: Centre of New Technologies, University of Warsaw, Banacha 2c, 02-097 Warsaw, Poland. y Received: 14 September 2020; Accepted: 9 October 2020; Published: 15 October 2020 Abstract: HMGB1 and HMGB2 proteins are abundantly expressed in human embryonic stem cells (hESCs) and hESC-derived progenitor cells (neuroectodermal cells, hNECs), though their functional roles in pluripotency and the mechanisms underlying their differentiation in response to the anticancer drug etoposide remain to be elucidated. Here, we show that HMGB1 and/or HMGB2 knockdown (KD) by shRNA in hESCs did not affect the cell stemness/pluripotency regardless of etoposide treatments, while in hESC-derived neuroectodermal cells, treatment resulted in differential effects on cell survival and the generation of rosette structures. The objective of this work was to determine whether HMGB1/2 proteins could modulate the sensitivity of hESCs and hESC-derived progenitor cells (hNECs) to etoposide. We observed that HMGB1 KD knockdown (KD) and, to a lesser extent, HMGB2 KD enhanced the sensitivity of hESCs to etoposide. Enhanced accumulation of 53BP1 on telomeres was detected by confocal microscopy in both untreated and etoposide-treated HMGB1 KD hESCs and hNECs, indicating that the loss of HMGB1 could destabilize telomeres.
    [Show full text]
  • Cell Cycle Arrest Through Indirect Transcriptional Repression by P53: I Have a DREAM
    Cell Death and Differentiation (2018) 25, 114–132 Official journal of the Cell Death Differentiation Association OPEN www.nature.com/cdd Review Cell cycle arrest through indirect transcriptional repression by p53: I have a DREAM Kurt Engeland1 Activation of the p53 tumor suppressor can lead to cell cycle arrest. The key mechanism of p53-mediated arrest is transcriptional downregulation of many cell cycle genes. In recent years it has become evident that p53-dependent repression is controlled by the p53–p21–DREAM–E2F/CHR pathway (p53–DREAM pathway). DREAM is a transcriptional repressor that binds to E2F or CHR promoter sites. Gene regulation and deregulation by DREAM shares many mechanistic characteristics with the retinoblastoma pRB tumor suppressor that acts through E2F elements. However, because of its binding to E2F and CHR elements, DREAM regulates a larger set of target genes leading to regulatory functions distinct from pRB/E2F. The p53–DREAM pathway controls more than 250 mostly cell cycle-associated genes. The functional spectrum of these pathway targets spans from the G1 phase to the end of mitosis. Consequently, through downregulating the expression of gene products which are essential for progression through the cell cycle, the p53–DREAM pathway participates in the control of all checkpoints from DNA synthesis to cytokinesis including G1/S, G2/M and spindle assembly checkpoints. Therefore, defects in the p53–DREAM pathway contribute to a general loss of checkpoint control. Furthermore, deregulation of DREAM target genes promotes chromosomal instability and aneuploidy of cancer cells. Also, DREAM regulation is abrogated by the human papilloma virus HPV E7 protein linking the p53–DREAM pathway to carcinogenesis by HPV.Another feature of the pathway is that it downregulates many genes involved in DNA repair and telomere maintenance as well as Fanconi anemia.
    [Show full text]