Regulation of Hematopoietic Progenitor Formation in a Shwachman Diamond Syndrome Induced Pluripotent Stem Cell Disease Model

Total Page:16

File Type:pdf, Size:1020Kb

Regulation of Hematopoietic Progenitor Formation in a Shwachman Diamond Syndrome Induced Pluripotent Stem Cell Disease Model Regulation of Hematopoietic Progenitor Formation in a Shwachman Diamond Syndrome Induced Pluripotent Stem Cell Disease Model by Alice Maria Luca A thesis submitted in conformity with the requirements for the degree of Masters of Science Institute of Medical Science University of Toronto © Copyright by Alice Maria Luca 2015 Regulation of Hematopoietic Progenitor Formation in a Shwachman-Diamond Syndrome Induced Pluripotent Stem Cell Disease Model Alice Maria Luca Master of Science Institute of Medical Sciences University of Toronto 2015 Abstract Shwachman-Diamond syndrome (SDS) is an inherited bone marrow failure disease, with 90% of SDS patients carrying a mutation in the SBDS gene. Due to limited efficacy or toxicity of current treatments available, and the otherwise reduced life expectancy of SDS patients, novel therapeutic strategies are needed. Since the main morbidity and mortality are related to the blood dyscrasia, studying hematopoiesis will help characterize the hematological phenotype. We hypothesized that the definitive wave of hematopoiesis is markedly impaired. We generated SDS iPSCs that recapitulated the human SDS disease, specifically, the reduced blood cell formation. The SDS iPSCs showed a defect in definitive hematopoiesis, with a marked reduction in the hemogenic endothelium population. We did not observe a defect in primitive hematopoiesis. Our study sheds light on the onset and progression of the SDS hematopoietic phenotype, and provides a platform for the development of novel, potential therapeutic targets to improve patient care. ii Acknowledgments My graduate journey has been a wonderful learning experience, and this work would not have been possible without the guidance, support and encouragement I have received from a number of influential people. I would like to take a moment to extend my deepest gratitude to all that were involved in this process. First of all, I would like to thank my supervisor, Dr.Yigal Dror, without whom this project would not have been possible. His guidance, critical-thinking and support were instrumental to my journey. I am grateful for all the time and attention that he always put into ensuring that I was successful every step of the way. Thank you for always pushing me to think critically, and for your unwavering support, even when I doubted myself. I would also like to thank Mathura Sabanayagam, for the ribosome profile experiments, and her willingness to spend her time performing them in a 4°C room. A special thank you to Bozana Zlateska for her instrumental role in the generation of the patient-specific iPSCs. Thank you to Hongbing and other lab members for always providing valuable input, and for your continuous encouragement. To Santhosh Dhanraj, thank you for your kind words and friendship. I am thankful for all the valuable discussion, constructive-criticism and encouragement that Dr.Gordon Keller has provided. I am also very grateful for all the time and patience that Marion Kennedy offered me, as well as her expertise, guidance and support. To iii Dr.Gordon Keller, Marion Kennedy, and the rest of the Keller lab, thank you for always keeping me laughing, and always making me feel welcomed. I would also like to thank my committee members, Dr.Michael Glogauer and Walter Kahr for all of their critical insight to my work. Last but not least, I would like to thank my personal cheerleaders, my parents, my sister, Delia, NV and Ryan for their constant words of encouragement every step of the way. iv Contributions I would like to thank Dr. Yigal Dror for his instrumental role in designing the research, critically interpreting the data and editing the thesis. Mathura Sabanayagam performed the ribosome profile experiments and analyzed the acquired data. Members of the Sickkids-University Health Network Flow Cytometry Facility provided their assistance for the cell sorting experiments. Members of The Centre for Applied Genomics (TCAG) at Sickkids Hospital provided their expertise in sequencing the SBDS mutations. The Centre for Commercialization of Regenerative Medicine (CCRM) generated the Sendai- derived iPSCs, performed reverse transcription quantitative real time polymerase chain reaction and immunofluorescence staining of stem cell pluripotency markers. Gordon Keller helped in designing the research, as well as critically interpreting the data. Marion Kennedy helped design the research, critically interpreted the data, and also trained me in induced pluripotent stem cells culturing and hematopoietic development. The rest of the members of the Dror and Keller labs provided their input regarding the design of the project. Dr. Michael Glogauer and Dr. Walter Kahr helped in designing the research. I performed the DNA isolation, polymerase chain reaction amplification, hematopoietic differentiation, clonogenic assay, flow cytometry and microscopy experiments, and analyzed the data. I was also responsible for designing the research and writing the thesis. This work would not have been possible without funding from the Institute of Medical Sciences, the Butterfly Guild and Nicola’s Triathlon for Kids. v Table of Contents Abstract…………………………………………………………………………………. ii Acknowledgments……………………………………………………………………… iii Contributions…………………………………………………………………………… v Table of Contents………………………………………………………………………. vi List of Tables and Figures……………………………………………………………… ix List of Abbreviations…………………………………………………………………… x Chapter I General Introduction 1.1 Shwachman Diamond Syndrome 1 1.1.1 Hematological abnormalities 2 1.1.2 Non-hematological abnormalities 6 1.1.3 Treatment 8 1.1.4 Characterization of Shwachman-Bodian Diamond Syndrome gene 9 1.1.5 Current disease models 14 1.2 Hematopoietic development and hematopoietic differentiation models 16 1.2.1 Primitive hematopoiesis 21 1.2.2 Definitive hematopoiesis 22 1.2.3 Hematopoietic differentiation model using control PSCs 28 1.3 Disease-specific hiPSCs 32 1.4 Objective of Study 35 1.4.1 Rationale 35 1.4.2 Hypothesis 39 1.4.3 Specific aims 39 Chapter II Materials and Methods 2.1 Cell culture 2.1.1 Cell lines 40 2.1.1.1 Mouse embryonic fibroblasts 40 2.1.1.2 OP9-DL1 40 2.1.1.3 hiPSCs 41 2.1.2 Feeder depletion 42 2.1.3 Establishing embryoid bodies for hematopoietic differentiation 42 2.1.3.1 Primitive hematopoiesis 43 2.1.3.2 Definitive hematopoiesis 45 2.1.4 Preparation of single-cell suspension from embryoid bodies 48 2.1.5 Preparation of single-cell suspensions from OP9-DL1 co-cultures 48 2.1.6 Clonogenic assay 48 2.1.7 Morphological analysis by Wright-Giemsa staining 49 2.2 DNA isolation 49 2.3 PCR amplification and sequencing 50 2.4 Flow cytometry and cell sorting 50 2.4.1 Antibodies 50 2.4.2. Fluorescence-activated cell sorting and flow cytometry analysis 50 vi 2.5 Sucrose density gradient ultracentrifugation 51 2.6 Statistics 52 Chapter III Results 3.1 SDS and control iPCSs can be generated from SDS patient and control fibroblasts 53 3.1.1 Generation efficiency 53 3.1.2 SDS iPSCs carry the original patient mutations 55 3.1.3 Pluripotency markers 56 3.1.4 Germ layer differentiation 58 3.2 SDS iPSCs are characterized by an abnormal ribosome profile 60 3.2.1 SDS iPSCs show reduced levels of 80S ribosome subunit 60 3.3 SDS iPSCs can form definitive hematopoietic progenitors 62 3.4 SDS iPSCs can form terminally differentiated blood cells characteristic of 64 definitive hematopoiesis 3.4.1 Terminally differentiated blood cells have a morphology characteristic of macrophages, 64 erythrocytes and granulocytes ……------------------------------------…………………….. 3.5 SDS iPSCs have a reduced capacity to form hematopoietic progenitors characteristic of definitive hematopoiesis 65 3.6 SDS iPSCs have a developmental defect in definitive hematopoiesis 67 3.6.1 Mesoderm induction is not affected in SDS iPSCs 67 3.6.2 Induction of cells with hemogenic endothelium potential is reduced in SDS iPSCs 70 3.6.3 Early hematopoietic progenitor induction is reduced in SDS iPSCs 73 3.6.4 Myeloid cell induction is reduced in SDS iPSCs 76 3.6.5 Granulocyte/Monocyte induction is reduced in SDS iPSCs 78 3.7 SDS iPSCs manifest a delay in the development of primitive 80 hematopoiesis, without a quantitative defect 3.7.1 Mesoderm/Hemangioblast induction is intact in SDS iPSCs 80 3.7.2 Induction of cells with hemogenic endothelium potential is not impaired in SDS iPSCs 82 3.7.3 Early hematopoietic progenitor induction is not affected in SDS iPSCs 85 3.7.4 Mature blood cell induction is not impaired in SDS iPSCs 88 3.7.5 SDS iPSCs showcase a delay in their ability to form hematopoietic progenitors characteristic of primitive hematopoiesis 92 Chapter IV Discussion 4.1 Discussion of results 94 4.1.1 SDS iPSCs can be generated using integrative and non-integrative transgenes 96 4.1.2 Recapitulation of SDS using SDS iPSCs ………………………………... 98 4.1.3 Mesoderm development is intact when SDS iPSCs are induced to undergo definitive hematopoiesis 102 4.1.4 The SDS definitive hematopoietic defect was first noticed at the hemogenic 103 endothelium induction phase 4.1.5 During primitive hematopoiesis, clonogenic potential is delayed, but not impaired 106 4.2 Limitations of study 107 4.3 Significance 109 vii Chapter V Conclusion 112 Chapter VI 114 Future Directions References 116 viii List of Tables and Figures Table 1. Human induced pluripotent stem cells generation efficiency 54 Table 2. Human induced pluripotent stem cells germ layer differentiation 59 Figure 1. Human SBDS gene structure 13 Figure 2. Human SBDS protein structure 13 Figure 3. Sites of human hematopoietic development during fetal life and early infancy 20 Figure 4. Current model of human hematopoietic hierarchy 27 Figure 5. Model of primitive and definitive hematopoiesis using pluripotent stem cells 31 Figure 6. Primitive hematopoiesis differentiation scheme for human pluripotent stem cells 44 Figure 7. Definitive hematopoiesis differentiation scheme for human pluripotent stem cells 47 Figure 8. Sequencing of SBDS in SDS induced pluripotent stem cells 55 Figure 9.
Recommended publications
  • Core Transcriptional Regulatory Circuitries in Cancer
    Oncogene (2020) 39:6633–6646 https://doi.org/10.1038/s41388-020-01459-w REVIEW ARTICLE Core transcriptional regulatory circuitries in cancer 1 1,2,3 1 2 1,4,5 Ye Chen ● Liang Xu ● Ruby Yu-Tong Lin ● Markus Müschen ● H. Phillip Koeffler Received: 14 June 2020 / Revised: 30 August 2020 / Accepted: 4 September 2020 / Published online: 17 September 2020 © The Author(s) 2020. This article is published with open access Abstract Transcription factors (TFs) coordinate the on-and-off states of gene expression typically in a combinatorial fashion. Studies from embryonic stem cells and other cell types have revealed that a clique of self-regulated core TFs control cell identity and cell state. These core TFs form interconnected feed-forward transcriptional loops to establish and reinforce the cell-type- specific gene-expression program; the ensemble of core TFs and their regulatory loops constitutes core transcriptional regulatory circuitry (CRC). Here, we summarize recent progress in computational reconstitution and biologic exploration of CRCs across various human malignancies, and consolidate the strategy and methodology for CRC discovery. We also discuss the genetic basis and therapeutic vulnerability of CRC, and highlight new frontiers and future efforts for the study of CRC in cancer. Knowledge of CRC in cancer is fundamental to understanding cancer-specific transcriptional addiction, and should provide important insight to both pathobiology and therapeutics. 1234567890();,: 1234567890();,: Introduction genes. Till now, one critical goal in biology remains to understand the composition and hierarchy of transcriptional Transcriptional regulation is one of the fundamental mole- regulatory network in each specified cell type/lineage.
    [Show full text]
  • Supplemental Materials ZNF281 Enhances Cardiac Reprogramming
    Supplemental Materials ZNF281 enhances cardiac reprogramming by modulating cardiac and inflammatory gene expression Huanyu Zhou, Maria Gabriela Morales, Hisayuki Hashimoto, Matthew E. Dickson, Kunhua Song, Wenduo Ye, Min S. Kim, Hanspeter Niederstrasser, Zhaoning Wang, Beibei Chen, Bruce A. Posner, Rhonda Bassel-Duby and Eric N. Olson Supplemental Table 1; related to Figure 1. Supplemental Table 2; related to Figure 1. Supplemental Table 3; related to the “quantitative mRNA measurement” in Materials and Methods section. Supplemental Table 4; related to the “ChIP-seq, gene ontology and pathway analysis” and “RNA-seq” and gene ontology analysis” in Materials and Methods section. Supplemental Figure S1; related to Figure 1. Supplemental Figure S2; related to Figure 2. Supplemental Figure S3; related to Figure 3. Supplemental Figure S4; related to Figure 4. Supplemental Figure S5; related to Figure 6. Supplemental Table S1. Genes included in human retroviral ORF cDNA library. Gene Gene Gene Gene Gene Gene Gene Gene Symbol Symbol Symbol Symbol Symbol Symbol Symbol Symbol AATF BMP8A CEBPE CTNNB1 ESR2 GDF3 HOXA5 IL17D ADIPOQ BRPF1 CEBPG CUX1 ESRRA GDF6 HOXA6 IL17F ADNP BRPF3 CERS1 CX3CL1 ETS1 GIN1 HOXA7 IL18 AEBP1 BUD31 CERS2 CXCL10 ETS2 GLIS3 HOXB1 IL19 AFF4 C17ORF77 CERS4 CXCL11 ETV3 GMEB1 HOXB13 IL1A AHR C1QTNF4 CFL2 CXCL12 ETV7 GPBP1 HOXB5 IL1B AIMP1 C21ORF66 CHIA CXCL13 FAM3B GPER HOXB6 IL1F3 ALS2CR8 CBFA2T2 CIR1 CXCL14 FAM3D GPI HOXB7 IL1F5 ALX1 CBFA2T3 CITED1 CXCL16 FASLG GREM1 HOXB9 IL1F6 ARGFX CBFB CITED2 CXCL3 FBLN1 GREM2 HOXC4 IL1F7
    [Show full text]
  • Genome-Wide DNA Methylation Analysis of KRAS Mutant Cell Lines Ben Yi Tew1,5, Joel K
    www.nature.com/scientificreports OPEN Genome-wide DNA methylation analysis of KRAS mutant cell lines Ben Yi Tew1,5, Joel K. Durand2,5, Kirsten L. Bryant2, Tikvah K. Hayes2, Sen Peng3, Nhan L. Tran4, Gerald C. Gooden1, David N. Buckley1, Channing J. Der2, Albert S. Baldwin2 ✉ & Bodour Salhia1 ✉ Oncogenic RAS mutations are associated with DNA methylation changes that alter gene expression to drive cancer. Recent studies suggest that DNA methylation changes may be stochastic in nature, while other groups propose distinct signaling pathways responsible for aberrant methylation. Better understanding of DNA methylation events associated with oncogenic KRAS expression could enhance therapeutic approaches. Here we analyzed the basal CpG methylation of 11 KRAS-mutant and dependent pancreatic cancer cell lines and observed strikingly similar methylation patterns. KRAS knockdown resulted in unique methylation changes with limited overlap between each cell line. In KRAS-mutant Pa16C pancreatic cancer cells, while KRAS knockdown resulted in over 8,000 diferentially methylated (DM) CpGs, treatment with the ERK1/2-selective inhibitor SCH772984 showed less than 40 DM CpGs, suggesting that ERK is not a broadly active driver of KRAS-associated DNA methylation. KRAS G12V overexpression in an isogenic lung model reveals >50,600 DM CpGs compared to non-transformed controls. In lung and pancreatic cells, gene ontology analyses of DM promoters show an enrichment for genes involved in diferentiation and development. Taken all together, KRAS-mediated DNA methylation are stochastic and independent of canonical downstream efector signaling. These epigenetically altered genes associated with KRAS expression could represent potential therapeutic targets in KRAS-driven cancer. Activating KRAS mutations can be found in nearly 25 percent of all cancers1.
    [Show full text]
  • 140503 IPF Signatures Supplement Withfigs Thorax
    Supplementary material for Heterogeneous gene expression signatures correspond to distinct lung pathologies and biomarkers of disease severity in idiopathic pulmonary fibrosis Daryle J. DePianto1*, Sanjay Chandriani1⌘*, Alexander R. Abbas1, Guiquan Jia1, Elsa N. N’Diaye1, Patrick Caplazi1, Steven E. Kauder1, Sabyasachi Biswas1, Satyajit K. Karnik1#, Connie Ha1, Zora Modrusan1, Michael A. Matthay2, Jasleen Kukreja3, Harold R. Collard2, Jackson G. Egen1, Paul J. Wolters2§, and Joseph R. Arron1§ 1Genentech Research and Early Development, South San Francisco, CA 2Department of Medicine, University of California, San Francisco, CA 3Department of Surgery, University of California, San Francisco, CA ⌘Current address: Novartis Institutes for Biomedical Research, Emeryville, CA. #Current address: Gilead Sciences, Foster City, CA. *DJD and SC contributed equally to this manuscript §PJW and JRA co-directed this project Address correspondence to Paul J. Wolters, MD University of California, San Francisco Department of Medicine Box 0111 San Francisco, CA 94143-0111 [email protected] or Joseph R. Arron, MD, PhD Genentech, Inc. MS 231C 1 DNA Way South San Francisco, CA 94080 [email protected] 1 METHODS Human lung tissue samples Tissues were obtained at UCSF from clinical samples from IPF patients at the time of biopsy or lung transplantation. All patients were seen at UCSF and the diagnosis of IPF was established through multidisciplinary review of clinical, radiological, and pathological data according to criteria established by the consensus classification of the American Thoracic Society (ATS) and European Respiratory Society (ERS), Japanese Respiratory Society (JRS), and the Latin American Thoracic Association (ALAT) (ref. 5 in main text). Non-diseased normal lung tissues were procured from lungs not used by the Northern California Transplant Donor Network.
    [Show full text]
  • Whole Exome Sequencing in Families at High Risk for Hodgkin Lymphoma: Identification of a Predisposing Mutation in the KDR Gene
    Hodgkin Lymphoma SUPPLEMENTARY APPENDIX Whole exome sequencing in families at high risk for Hodgkin lymphoma: identification of a predisposing mutation in the KDR gene Melissa Rotunno, 1 Mary L. McMaster, 1 Joseph Boland, 2 Sara Bass, 2 Xijun Zhang, 2 Laurie Burdett, 2 Belynda Hicks, 2 Sarangan Ravichandran, 3 Brian T. Luke, 3 Meredith Yeager, 2 Laura Fontaine, 4 Paula L. Hyland, 1 Alisa M. Goldstein, 1 NCI DCEG Cancer Sequencing Working Group, NCI DCEG Cancer Genomics Research Laboratory, Stephen J. Chanock, 5 Neil E. Caporaso, 1 Margaret A. Tucker, 6 and Lynn R. Goldin 1 1Genetic Epidemiology Branch, Division of Cancer Epidemiology and Genetics, National Cancer Institute, NIH, Bethesda, MD; 2Cancer Genomics Research Laboratory, Division of Cancer Epidemiology and Genetics, National Cancer Institute, NIH, Bethesda, MD; 3Ad - vanced Biomedical Computing Center, Leidos Biomedical Research Inc.; Frederick National Laboratory for Cancer Research, Frederick, MD; 4Westat, Inc., Rockville MD; 5Division of Cancer Epidemiology and Genetics, National Cancer Institute, NIH, Bethesda, MD; and 6Human Genetics Program, Division of Cancer Epidemiology and Genetics, National Cancer Institute, NIH, Bethesda, MD, USA ©2016 Ferrata Storti Foundation. This is an open-access paper. doi:10.3324/haematol.2015.135475 Received: August 19, 2015. Accepted: January 7, 2016. Pre-published: June 13, 2016. Correspondence: [email protected] Supplemental Author Information: NCI DCEG Cancer Sequencing Working Group: Mark H. Greene, Allan Hildesheim, Nan Hu, Maria Theresa Landi, Jennifer Loud, Phuong Mai, Lisa Mirabello, Lindsay Morton, Dilys Parry, Anand Pathak, Douglas R. Stewart, Philip R. Taylor, Geoffrey S. Tobias, Xiaohong R. Yang, Guoqin Yu NCI DCEG Cancer Genomics Research Laboratory: Salma Chowdhury, Michael Cullen, Casey Dagnall, Herbert Higson, Amy A.
    [Show full text]
  • Transcription Factor Gene Expression Profiling and Analysis of SOX Gene Family Transcription Factors in Human Limbal Epithelial
    Transcription factor gene expression profiling and analysis of SOX gene family transcription factors in human limbal epithelial progenitor cells Der Naturwissenschaftlichen Fakultät der Friedrich-Alexander-Universität Erlangen-Nürnberg zur Erlangung des Doktorgrades Dr. rer. nat. vorgelegt von Dr. med. Johannes Menzel-Severing aus Bonn Als Dissertation genehmigt von der Naturwissenschaftlichen Fakultät der Friedrich-Alexander-Universität Erlangen-Nürnberg Tag der mündlichen Prüfung: 7. Februar 2018 Vorsitzender des Promotionsorgans: Prof. Dr. Georg Kreimer Gutachter: Prof. Dr. Andreas Feigenspan Prof. Dr. Ursula Schlötzer-Schrehardt 1 INDEX 1. ABSTRACTS Page 1.1. Abstract in English 4 1.2. Zusammenfassung auf Deutsch 7 2. INTRODUCTION 2.1. Anatomy and histology of the cornea and the corneal surface 11 2.2. Homeostasis of corneal epithelium and the limbal stem cell paradigm 13 2.3. The limbal stem cell niche 15 2.4. Cell therapeutic strategies in ocular surface disease 17 2.5. Alternative cell sources for transplantation to the corneal surface 18 2.6. Transcription factors in cell differentiation and reprogramming 21 2.7. Transcription factors in limbal epithelial cells 22 2.8. Research question 25 3. MATERIALS AND METHODS 3.1. Human donor corneas 27 3.2. Laser Capture Microdissection (LCM) 28 3.3. RNA amplification and RT2 profiler PCR arrays 29 3.4. Real-time PCR analysis 33 3.5. Immunohistochemistry 34 3.6. Limbal epithelial cell culture 38 3.7. Transcription-factor knockdown/overexpression in vitro 39 3.8. Proliferation assay 40 3.9. Western blot 40 3.10. Statistical analysis 41 2 4. RESULTS 4.1. Quality control of LCM-isolated and amplified RNA 42 4.2.
    [Show full text]
  • Identification of Transcriptional Mechanisms Downstream of Nf1 Gene Defeciency in Malignant Peripheral Nerve Sheath Tumors Daochun Sun Wayne State University
    Wayne State University DigitalCommons@WayneState Wayne State University Dissertations 1-1-2012 Identification of transcriptional mechanisms downstream of nf1 gene defeciency in malignant peripheral nerve sheath tumors Daochun Sun Wayne State University, Follow this and additional works at: http://digitalcommons.wayne.edu/oa_dissertations Recommended Citation Sun, Daochun, "Identification of transcriptional mechanisms downstream of nf1 gene defeciency in malignant peripheral nerve sheath tumors" (2012). Wayne State University Dissertations. Paper 558. This Open Access Dissertation is brought to you for free and open access by DigitalCommons@WayneState. It has been accepted for inclusion in Wayne State University Dissertations by an authorized administrator of DigitalCommons@WayneState. IDENTIFICATION OF TRANSCRIPTIONAL MECHANISMS DOWNSTREAM OF NF1 GENE DEFECIENCY IN MALIGNANT PERIPHERAL NERVE SHEATH TUMORS by DAOCHUN SUN DISSERTATION Submitted to the Graduate School of Wayne State University, Detroit, Michigan in partial fulfillment of the requirements for the degree of DOCTOR OF PHILOSOPHY 2012 MAJOR: MOLECULAR BIOLOGY AND GENETICS Approved by: _______________________________________ Advisor Date _______________________________________ _______________________________________ _______________________________________ © COPYRIGHT BY DAOCHUN SUN 2012 All Rights Reserved DEDICATION This work is dedicated to my parents and my wife Ze Zheng for their continuous support and understanding during the years of my education. I could not achieve my goal without them. ii ACKNOWLEDGMENTS I would like to express tremendous appreciation to my mentor, Dr. Michael Tainsky. His guidance and encouragement throughout this project made this dissertation come true. I would also like to thank my committee members, Dr. Raymond Mattingly and Dr. John Reiners Jr. for their sustained attention to this project during the monthly NF1 group meetings and committee meetings, Dr.
    [Show full text]
  • Single Cell Derived Clonal Analysis of Human Glioblastoma Links
    SUPPLEMENTARY INFORMATION: Single cell derived clonal analysis of human glioblastoma links functional and genomic heterogeneity ! Mona Meyer*, Jüri Reimand*, Xiaoyang Lan, Renee Head, Xueming Zhu, Michelle Kushida, Jane Bayani, Jessica C. Pressey, Anath Lionel, Ian D. Clarke, Michael Cusimano, Jeremy Squire, Stephen Scherer, Mark Bernstein, Melanie A. Woodin, Gary D. Bader**, and Peter B. Dirks**! ! * These authors contributed equally to this work.! ** Correspondence: [email protected] or [email protected]! ! Supplementary information - Meyer, Reimand et al. Supplementary methods" 4" Patient samples and fluorescence activated cell sorting (FACS)! 4! Differentiation! 4! Immunocytochemistry and EdU Imaging! 4! Proliferation! 5! Western blotting ! 5! Temozolomide treatment! 5! NCI drug library screen! 6! Orthotopic injections! 6! Immunohistochemistry on tumor sections! 6! Promoter methylation of MGMT! 6! Fluorescence in situ Hybridization (FISH)! 7! SNP6 microarray analysis and genome segmentation! 7! Calling copy number alterations! 8! Mapping altered genome segments to genes! 8! Recurrently altered genes with clonal variability! 9! Global analyses of copy number alterations! 9! Phylogenetic analysis of copy number alterations! 10! Microarray analysis! 10! Gene expression differences of TMZ resistant and sensitive clones of GBM-482! 10! Reverse transcription-PCR analyses! 11! Tumor subtype analysis of TMZ-sensitive and resistant clones! 11! Pathway analysis of gene expression in the TMZ-sensitive clone of GBM-482! 11! Supplementary figures and tables" 13" "2 Supplementary information - Meyer, Reimand et al. Table S1: Individual clones from all patient tumors are tumorigenic. ! 14! Fig. S1: clonal tumorigenicity.! 15! Fig. S2: clonal heterogeneity of EGFR and PTEN expression.! 20! Fig. S3: clonal heterogeneity of proliferation.! 21! Fig.
    [Show full text]
  • Xo PANEL DNA GENE LIST
    xO PANEL DNA GENE LIST ~1700 gene comprehensive cancer panel enriched for clinically actionable genes with additional biologically relevant genes (at 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
    [Show full text]
  • Factor Expression and Correlate with Specific Transcription in Early Human Precursor B Cell Subsets Ig Gene Rearrangement Steps
    The Journal of Immunology Ig Gene Rearrangement Steps Are Initiated in Early Human Precursor B Cell Subsets and Correlate with Specific Transcription Factor Expression1 Menno C. van Zelm,*† Mirjam van der Burg,* Dick de Ridder,*‡ Barbara H. Barendregt,*† Edwin F. E. de Haas,* Marcel J. T. Reinders,‡ Arjan C. Lankester,§ Tom Re´ve´sz,¶ Frank J. T. Staal,* and Jacques J. M. van Dongen2* The role of specific transcription factors in the initiation and regulation of Ig gene rearrangements has been studied extensively in mouse models, but data on normal human precursor B cell differentiation are limited. We purified five human precursor B cell subsets, and assessed and quantified their IGH, IGK, and IGL gene rearrangement patterns and gene expression profiles. Pro-B cells already massively initiate DH-JH rearrangements, which are completed with VH-DJH rearrangements in pre-B-I cells. Large cycling pre-B-II cells are selected for in-frame IGH gene rearrangements. The first IGK/IGL gene rearrangements were initiated in pre-B-I cells, but their frequency increased enormously in small pre-B-II cells, and in-frame selection was found in immature B cells. Transcripts of the RAG1 and RAG2 genes and earlier defined transcription factors, such as E2A, early B cell factor, E2-2, PAX5, and IRF4, were specifically up-regulated at stages undergoing Ig gene rearrangements. Based on the combined Ig gene rearrangement status and gene expression profiles of consecutive precursor B cell subsets, we identified 16 candidate genes involved in initiation and/or regulation of Ig gene rearrangements. These analyses provide new insights into early human pre- cursor B cell differentiation steps and represent an excellent template for studies on oncogenic transformation in precursor B acute lymphoblastic leukemia and B cell differentiation blocks in primary Ab deficiencies.
    [Show full text]
  • Function of Sox2 As a Transcriptional Repressor
    Function of Sox2 as a transcriptional repressor By Yu-Ru Liu, M. Sc Thesis submitted to the University of Nottingham for the degree of Doctor of Philosophy September 2011 1 Abstract Sox2 is one of the earliest known transcription factors to be expressed during development of the nervous system (Rex et al., 1997; Silvia Brunelli, 2003; Wang et al., 2006b; Dee et al., 2008). Ectodermal cells expressing Sox2 have the potential to differentiate into nerve cells. Cells expressing Sox2 are specified to a neural fate during neural induction. Sox2 belongs to the SoxB1 family, comprising Sox1, Sox2 and Sox3, which are generally considered to activate specific target genes, whereas, the SoxB2 group, Sox14 and Sox21, act as transcriptional repressors (Uchikawa, Kamachi, & Kondoh, 1999). However, Sox2 has also been demonstrated to act as a repressor (Kopp et al., 2008) which implies that Sox2 could have a dual-function in vivo . Previous studies indicated that the HMG box-containing protein, Tcf/Lef, interacts with the transcriptional co-repressor, Groucho (Helen Brantjes, 2001). We therefore set out to determine if interaction with the Groucho co-repressor could also explain the repressor ability of Sox2. In this study, we have examined the interaction between Sox2 and Groucho using nuclear translocation, yeast-two-hybrid and co-immunoprecipitation assays. The data suggest that Sox2 interacts with Groucho through a C-terminal, engrailed-like motif. The effect of Groucho on Sox2 function was measured using a luciferase reporter assay. The transcriptional activation activity of Sox2 was repressed after co-expressing with Groucho. To address the biological function of Sox2-Groucho interaction, a loss-of-repressor-function mutant of Sox2 was created by point mutating the essential engrailed-like motif.
    [Show full text]
  • Sox9 and Rbpj Differentially Regulate Endothelial to Mesenchymal Transition and Wound Scarring in Murine Endovascular Progenitors
    ARTICLE https://doi.org/10.1038/s41467-021-22717-9 OPEN Sox9 and Rbpj differentially regulate endothelial to mesenchymal transition and wound scarring in murine endovascular progenitors Jilai Zhao 1,6, Jatin Patel1,2,6, Simranpreet Kaur1, Seen-Ling Sim1, Ho Yi Wong1, Cassandra Styke 1, Isabella Hogan1, Sam Kahler 1, Hamish Hamilton1, Racheal Wadlow1, James Dight1, Ghazaleh Hashemi 1, ✉ Laura Sormani1, Edwige Roy 1, Mervin C. Yoder 3, Mathias Francois4,5 & Kiarash Khosrotehrani 1 1234567890():,; Endothelial to mesenchymal transition (EndMT) is a leading cause of fibrosis and disease, however its mechanism has yet to be elucidated. The endothelium possesses a profound regenerative capacity to adapt and reorganize that is attributed to a population of vessel- resident endovascular progenitors (EVP) governing an endothelial hierarchy. Here, using fate analysis, we show that two transcription factors SOX9 and RBPJ specifically affect the murine EVP numbers and regulate lineage specification. Conditional knock-out of Sox9 from the vasculature (Sox9fl/fl/Cdh5-CreER RosaYFP) depletes EVP while enhancing Rbpj expression and canonical Notch signalling. Additionally, skin wound analysis from Sox9 conditional knock-out mice demonstrates a significant reduction in pathological EndMT resulting in reduced scar area. The converse is observed with Rbpj conditionally knocked-out from the murine vas- culature (Rbpjfl/fl/Cdh5-CreER RosaYFP) or inhibition of Notch signaling in human endothelial colony forming cells, resulting in enhanced Sox9 and EndMT related gene (Snail, Slug, Twist1, Twist2, TGF-β) expression. Similarly, increased endothelial hedgehog signaling (Ptch1fl/fl/ Cdh5-CreER RosaYFP), that upregulates the expression of Sox9 in cells undergoing patholo- gical EndMT, also results in excess fibrosis.
    [Show full text]