Engineered Stem Cells in Hematologic Diseases

Total Page:16

File Type:pdf, Size:1020Kb

Engineered Stem Cells in Hematologic Diseases Next Generation Transplantation: Engineered Stem Cells in Hematologic Diseases George Q. Daley Boston Children’s Hospital/ Harvard Medical School/ HHMI Tandem Meetings 2016 Disclosures: Scientific Advisory Boards (consulting fees and equity) MPM Capital, Solasia, Epizyme, Ocata, Raze, True North, Verastem, AthenaHealth Research Funding: Megakaryon Boston Children’s/ Dana Farber Pediatric Stem Cell Transplant 2015: 114 Transplants Leslie Lehmann Christy Duncan Nick Haining Clinical Director Assoc. Clinical Dir. Assoc. Scientific Dir. Alessandra Biffi Director Gene Therapy Steve Margossian Sung-Yun Pai Suneet Agarwal Michelle Lee Christy Ullirch Jen Whangbo Susanne Baumeister Esther Obang Leo Wang Birgit Knoechel Inga Hoffman Autologous Cell Therapy from iPS Cells NSG Transplant Skin or Blood Cell Primary Immune Deficiency Hematopoietic Bone Marrow Failure stem cells Hemoglobinopathy Reprogramming (Oct4/Sox2/Klf4/Myc) ? Gene Repair Diseased iPS cells Healthy iPS cells ES/ iPS Mammalian Embryoidblood development:Bodies Primitive and Definitive= Yolk Sac waves Morphogens? Drugs? Transcription Factors? Strategies for deriving HSCs from HUMAN pluripotent stem cells (ESC/iPSCs) HSC-specific TFs ?? Respecification CD34+43- ESC/iPSC Hemogenic HSC? CD34+ Endothelium CD45+ TFs Myeloid CFUs Specification Morphogens Respecifying blood progenitors from iPSCs hESC/iPSC EB differentiation - BMP4/cytokines CD34+45+ HPCs D0 D4-7 D11+ D14 HSC TFs 7+ days 9 HSC TFs: Transplant HLF ERG Control MYB SOX4 CD34+45+ KLF4 HPCs RORA Microarray HOXA5 HOXA9 HOXA10 6 6 Respecified progenitors mediate short-term engraftment in NSG mice 5 Dox-inducible factors: ERG, HOXA9, RORA, SOX4, MYB (CD34-5F) Cord blood Erythro-myeloid Erythroid only GlyA CD45 CD45++ gate CD45++ gate GLYA+ gate CD19 CD71 CD33 mTer119 7 7 Engrafted erythroid cells undergo globin switching Erythroid engraftment in vivo 125 CD71 t e p i r 100 c g s n a r 75 b t n i b 50 o l G 25 % 0 B F B 1 2 3 4 5 1 2 3 5 E C m m m m m m m m F F F F F B B B 5 5 5 5 5 C C C GlyA SYTO60 BAND3 8 8 RED BLOOD CELL DIFFERENTIATION FROM iPS PROGENITORS IN VITRO ERG, HOXA9, RORA, SOX4, MYB Respecification Stage I Stage II Stage III 14d -DOX CD34+CD45+ DBA 21 days Stage I Stage II Stage IIIa Stage IIIb D9 D14 D18 D18 E M Lee et al. Nature 2015 9 9 Human IPS RBCs circulate after transfusion Bone Marrow BM + Clodronate Peripheral blood 24h (Enucleation) SYTO60 GLA Human RBCs Colony screen of an expanded set of 28 HSC transcription factors GEMM/BFU-E CFU-GM - HSC- specific + HOXA9 100 TFs ERG 95 RORA 68 68 47 47 EB 34+ EAR 13F 42 37 32 26 ERG TCF7L2 16 HOXA9 ETS1 16 RORA SSBP2 SOX4 PRDM16 11 MYB DACH1 5 HOXA5 PBX1 5 KLF5 HIF3A 5 CIITA HDAC9 5 GATA3 NFIA MEIS1 NRIP1 5 THRB EVI1 5 SKAP1 SMARCA1 0 FOXO1 WWTR1 0 NKX2-3 HOXB4 0 11 11 A subset of TFs is sufficient to induce T- and B-lymphoid potential 28/13F EB CD34+ CD34+CD38- Stromal culture +DOX -DOX OP9-DL1: CD4+CD8+ T cells MS-5: CD19+CD20+ B cells 2wks 5wks 28F 13F 13F CD8 T cells CD3 CD4 TCRab TCRgd 28F 13F 13F B cells CD19 CD19 CD11b CD20 CD10 13F-1 further reduction to 7F A screen for epigenetic modifiers that restrict definitive potential Onder et al. Nature (2012). ERG HOXA9 shRNA RORA SOX4 Measure MYB %CD4+CD8+ T cells CD34+ CD45+ OP9-DL1 shRNA Screen identifies H3K9 HMTs as repressors of T cell potential 5F + shLUC 5F+sh1 5F+sh2 5F+sh3 CD8 5F+sh4 5F+sh5 5F+sh6 5F+sh7 CD4 Repression of top candidate enhances B/T potential of respecified iPSC-derived blood lineages 5F + shLUC 5F + sh1-1 5F + sh1-2 CD8 CELLS T CD4 5F + shLUC 5F + sh1-1 5F + sh1-2 CD19 B CELLS CD56 Generating multipotential progenitors from iPSCs CD34+CD45+ IPSCs GMP-like Patient HOXA9/5 “Core” transcription ERG factors RORA SOX4 NFIA and epigenetic MYB DACH1 regulators CMP-like CLP-like Congenital anemias Immunodeficiencies eg SCA, DBA eg RD, SCID Engineered T cells 16 16 Definitive HSCs from Hemogenic Endothelium Yokomizo T & Dzierzak E, Development, 2010; Wang LD & Wagers AJ, Nature Rev Mol Cell Biol, 2011 Bertrand et al., Nature, 2010 Hematopoietic differentiation thru hemogenic endothelium EB differentiation Endothelial-hematopoietic transition (EHT) 0 1 2 3 4 5 6 7 8 +1 2 3 4 5 6 7 bFGF MACS (CD34+) BMP4 EPO VEGF BMP4 BMP4 BMP4 bFGF TPO IL-6 bFGF bFGF bFGF VEGF FLT-3L IL-11 SB VEGF IGF-1 IL-6 IL-3 CHIR SCF IL-11 SHH EPO IGF-1 Ang II SCF Losartan plate on Matrigel human iPS Protocol adapted from: Ditadi et al., Nature Cell Biol, 2015 Missing signals to confer HSC fate on ES/iPS-HE? HE cells Human Embryo Intrafemoral injection of HEHE can engraft sub-lethally irradiated NSG No engraftment… Selecting Transcription Factors for Screening 26 HSC-TFs (including Rafii, Rossi and Daley) SSBP2, ZKSCAN1, DACH1, TGIF2, GATA2, HLF, BCL11A, NRIP1, IRF2, HOXA5, HOXA9, HOXA10, SIX5, EVI1, MAFF, KLF4, LCOR, RUNX1, GFI1, SPI1, FOSB, MEISS1, ERG, RORA, SOX4, MYB HE dHE FL-HSC Gateway Destination 5’LTR 3’LTR PsiPsi RRE TRE2 Cm ccdB HA hEF1α rtTA3 IRES eGFP GFP+ GFP+ Clone factors into Dox-inducible lenti- vector w/o infection Infected HE cells Screening for factors that convert HE to HSCs Library of 26 TFs Engraftment (peripheral blood) 4w 6w 8w 10w 14w At 10 weeks sort out human- derived BM cells iPS-Hemogenic NSG mice Endothelium Weeks HSC, T, B, Myeloid, Erythroid Flow on engrafted cells Multi-lineage marrow engraftment • Transgene detection in engrafted cells hiPSCs hESCs 7 Defined TFs: HE HSC hiPSC (vivo) SPI1, ERG, hESC (vivo) RUNX1, LCOR, ERG, SPI1 HOX SSBP2, Multiple Hox genes ZKSCAN1 MAFF RUNX1, LCOR MYB, RORA, BCL11A, GFI1 CFU Multilineage engraftment in primary recipients Multilineage engraftment in secondary recipients Globin switching in engrafted red blood cells HBG HBB HBE Engraftment with T and B cells DP T cells in Thymus PMA IFNg Serum ELISA detects Human IgM and IgG Deriving HSCs from human PSCs: a combinatorial approach Developmental Biology: Synthetic Biology: mimicking normal ectopic expression of embryonic development transcription factors (TFs) Embryoid body TFs Cytokines HSC- like cells Evo-devo Synbio HE cells In vivo specification Next Generation Transplantation… Transplant Skin or RBCs Blood Cell PLTs Hematopoietic Combined Gene and Cell Therapy stem cells Transfusable blood products http://www.sciencephoto.com/ …clinical trials in 2017 Koji Eto, Harvey Lodish Reprogramming (Oct4/Sox2/Klf4/Myc) Gene Repair Diseased iPS cells Healthy iPS cells Acknowledgements Sergei Doulatov Linda Thuy Vo Rio Sugimura Thorsten Schlaeger Patricia Sousa Len Zon Marcella Cesana Michael YiFen Lu Hu Li, Jim Collins Chen Vanessa Lundin Melissa Kinney Funding: NHLBI UO1 PCBC, NIDDK R24, DORIS DUKE, HHMI .
Recommended publications
  • Microglia Emerge from Erythromyeloid Precursors Via Pu.1- and Irf8-Dependent Pathways
    ART ic LE S Microglia emerge from erythromyeloid precursors via Pu.1- and Irf8-dependent pathways Katrin Kierdorf1,2, Daniel Erny1, Tobias Goldmann1, Victor Sander1, Christian Schulz3,4, Elisa Gomez Perdiguero3,4, Peter Wieghofer1,2, Annette Heinrich5, Pia Riemke6, Christoph Hölscher7,8, Dominik N Müller9, Bruno Luckow10, Thomas Brocker11, Katharina Debowski12, Günter Fritz1, Ghislain Opdenakker13, Andreas Diefenbach14, Knut Biber5,15, Mathias Heikenwalder16, Frederic Geissmann3,4, Frank Rosenbauer6 & Marco Prinz1,17 Microglia are crucial for immune responses in the brain. Although their origin from the yolk sac has been recognized for some time, their precise precursors and the transcription program that is used are not known. We found that mouse microglia were derived from primitive c-kit+ erythromyeloid precursors that were detected in the yolk sac as early as 8 d post conception. + lo − + − + These precursors developed into CD45 c-kit CX3CR1 immature (A1) cells and matured into CD45 c-kit CX3CR1 (A2) cells, as evidenced by the downregulation of CD31 and concomitant upregulation of F4/80 and macrophage colony stimulating factor receptor (MCSF-R). Proliferating A2 cells became microglia and invaded the developing brain using specific matrix metalloproteinases. Notably, microgliogenesis was not only dependent on the transcription factor Pu.1 (also known as Sfpi), but also required Irf8, which was vital for the development of the A2 population, whereas Myb, Id2, Batf3 and Klf4 were not required. Our data provide cellular and molecular insights into the origin and development of microglia. Microglia are the tissue macrophages of the brain and scavenge dying have the ability to give rise to microglia and macrophages in vitro cells, pathogens and molecules using pattern recognition receptors and in vivo under defined conditions.
    [Show full text]
  • I LITERATURE-BASED DISCOVERY of KNOWN and POTENTIAL NEW
    LITERATURE-BASED DISCOVERY OF KNOWN AND POTENTIAL NEW MECHANISMS FOR RELATING THE STATUS OF CHOLESTEROL TO THE PROGRESSION OF BREAST CANCER BY YU WANG THESIS Submitted in partial fulfillment of the requirements for the degree of Master of Science in Bioinformatics with a concentration in Library and Information Science in the Graduate College of the University of Illinois at Urbana-Champaign, 2019 Urbana, Illinois Adviser: Professor Vetle I. Torvik Professor Erik Russell Nelson i ABSTRACT Breast cancer has been studied for a long period of time and from a variety of perspectives in order to understand its pathogeny. The pathogeny of breast cancer can be classified into two groups: hereditary and spontaneous. Although cancer in general is considered a genetic disease, spontaneous factors are responsible for most of the pathogeny of breast cancer. In other words, breast cancer is more likely to be caused and deteriorated by the dysfunction of a physical molecule than be caused by germline mutation directly. Interestingly, cholesterol, as one of those molecules, has been discovered to correlate with breast cancer risk. However, the mechanisms of how cholesterol helps breast cancer progression are not thoroughly understood. As a result, this study aims to study known and discover potential new mechanisms regarding to the correlation of cholesterol and breast cancer progression using literature review and literature-based discovery. The known mechanisms are further classified into four groups: cholesterol membrane content, transport of cholesterol, cholesterol metabolites, and other. The potential mechanisms, which are intended to provide potential new treatments, have been identified and checked for feasibility by an expert.
    [Show full text]
  • Homeobox Gene Expression Profile in Human Hematopoietic Multipotent
    Leukemia (2003) 17, 1157–1163 & 2003 Nature Publishing Group All rights reserved 0887-6924/03 $25.00 www.nature.com/leu Homeobox gene expression profile in human hematopoietic multipotent stem cells and T-cell progenitors: implications for human T-cell development T Taghon1, K Thys1, M De Smedt1, F Weerkamp2, FJT Staal2, J Plum1 and G Leclercq1 1Department of Clinical Chemistry, Microbiology and Immunology, Ghent University Hospital, Ghent, Belgium; and 2Department of Immunology, Erasmus Medical Center, Rotterdam, The Netherlands Class I homeobox (HOX) genes comprise a large family of implicated in this transformation proces.14 The HOX-C locus transcription factors that have been implicated in normal and has been primarily implicated in lymphomas.15 malignant hematopoiesis. However, data on their expression or function during T-cell development is limited. Using degener- Hematopoietic cells are derived from stem cells that reside in ated RT-PCR and Affymetrix microarray analysis, we analyzed fetal liver (FL) in the embryo and in the adult bone marrow the expression pattern of this gene family in human multipotent (ABM), which have the unique ability to self-renew and thereby stem cells from fetal liver (FL) and adult bone marrow (ABM), provide a life-long supply of blood cells. T lymphocytes are a and in T-cell progenitors from child thymus. We show that FL specific type of hematopoietic cells that play a major role in the and ABM stem cells are similar in terms of HOX gene immune system. They develop through a well-defined order of expression, but significant differences were observed between differentiation steps in the thymus.16 Several transcription these two cell types and child thymocytes.
    [Show full text]
  • ERG Dependence Distinguishes Developmental Control of Hematopoietic Stem Cell Maintenance from Hematopoietic Specification
    Downloaded from genesdev.cshlp.org on September 28, 2021 - Published by Cold Spring Harbor Laboratory Press ERG dependence distinguishes developmental control of hematopoietic stem cell maintenance from hematopoietic specification Samir Taoudi,1,2,6 Thomas Bee,3 Adrienne Hilton,1 Kathy Knezevic,3 Julie Scott,4 Tracy A. Willson,1,2 Caitlin Collin,1 Tim Thomas,1,2 Anne K. Voss,1,2 Benjamin T. Kile,1,2 Warren S. Alexander,2,5 John E. Pimanda,3 and Douglas J. Hilton1,2 1Molecular Medicine Division, The Walter and Eliza Institute of Medical Research, Melbourne, Parkville, Victoria 3052, Australia; 2Department of Medical Biology, The University of Melbourne, Melbourne, Parkville, Victoria 3010, Australia; 3Lowy Cancer Research Centre, The Prince of Wales Clinical School, University of New South Wales, Sydney 2052, Australia; 4Microinjection Services, The Walter and Eliza Institute of Medical Research, Melbourne, Parkville, Victoria 3052, Australia; 5Cancer and Haematology Division, The Walter and Eliza Institute of Medical Research, Melbourne, Parkville, Victoria 3052, Australia Although many genes are known to be critical for early hematopoiesis in the embryo, it remains unclear whether distinct regulatory pathways exist to control hematopoietic specification versus hematopoietic stem cell (HSC) emergence and function. Due to their interaction with key regulators of hematopoietic commitment, particular interest has focused on the role of the ETS family of transcription factors; of these, ERG is predicted to play an important role in the initiation of hematopoiesis, yet we do not know if or when ERG is required. Using in vitro and in vivo models of hematopoiesis and HSC development, we provide strong evidence that ERG is at the center of a distinct regulatory program that is not required for hematopoietic specification or differentiation but is critical for HSC maintenance during embryonic development.
    [Show full text]
  • Ubiquitin-Mediated Control of ETS Transcription Factors: Roles in Cancer and Development
    International Journal of Molecular Sciences Review Ubiquitin-Mediated Control of ETS Transcription Factors: Roles in Cancer and Development Charles Ducker * and Peter E. Shaw * Queen’s Medical Centre, School of Life Sciences, University of Nottingham, Nottingham NG7 2UH, UK * Correspondence: [email protected] (C.D.); [email protected] (P.E.S.) Abstract: Genome expansion, whole genome and gene duplication events during metazoan evolution produced an extensive family of ETS genes whose members express transcription factors with a conserved winged helix-turn-helix DNA-binding domain. Unravelling their biological roles has proved challenging with functional redundancy manifest in overlapping expression patterns, a common consensus DNA-binding motif and responsiveness to mitogen-activated protein kinase signalling. Key determinants of the cellular repertoire of ETS proteins are their stability and turnover, controlled largely by the actions of selective E3 ubiquitin ligases and deubiquitinases. Here we discuss the known relationships between ETS proteins and enzymes that determine their ubiquitin status, their integration with other developmental signal transduction pathways and how suppression of ETS protein ubiquitination contributes to the malignant cell phenotype in multiple cancers. Keywords: E3 ligase complex; deubiquitinase; gene fusions; mitogens; phosphorylation; DNA damage 1. Introduction Citation: Ducker, C.; Shaw, P.E. Cell growth, proliferation and differentiation are complex, concerted processes that Ubiquitin-Mediated Control of ETS Transcription Factors: Roles in Cancer rely on careful regulation of gene expression. Control over gene expression is maintained and Development. Int. J. Mol. Sci. through signalling pathways that respond to external cellular stimuli, such as growth 2021, 22, 5119. https://doi.org/ factors, cytokines and chemokines, that invoke expression profiles commensurate with 10.3390/ijms22105119 diverse cellular outcomes.
    [Show full text]
  • Inducible Transgene Expression in PDX Models
    Liu et al. Biomarker Research (2020) 8:46 https://doi.org/10.1186/s40364-020-00226-z RESEARCH Open Access Inducible transgene expression in PDX models in vivo identifies KLF4 as a therapeutic target for B-ALL Wen-Hsin Liu1, Paulina Mrozek-Gorska2, Anna-Katharina Wirth1, Tobias Herold1,3, Larissa Schwarzkopf1, Dagmar Pich2, Kerstin Völse1, M. Camila Melo-Narváez2, Michela Carlet1, Wolfgang Hammerschmidt2,4 and Irmela Jeremias1,5,6* Abstract Background: Clinically relevant methods are not available that prioritize and validate potential therapeutic targets for individual tumors, from the vast amount of tumor descriptive expression data. Methods: We established inducible transgene expression in clinically relevant patient-derived xenograft (PDX) models in vivo to fill this gap. Results: With this technique at hand, we analyzed the role of the transcription factor Krüppel-like factor 4 (KLF4) in B-cell acute lymphoblastic leukemia (B-ALL) PDX models at different disease stages. In competitive preclinical in vivo trials, we found that re-expression of wild type KLF4 reduced the leukemia load in PDX models of B-ALL, with the strongest effects being observed after conventional chemotherapy in minimal residual disease (MRD). A nonfunctional KLF4 mutant had no effect on this model. The re-expression of KLF4 sensitized tumor cells in the PDX model towards systemic chemotherapy in vivo. It is of major translational relevance that azacitidine upregulated KLF4 levels in the PDX model and a KLF4 knockout reduced azacitidine-induced cell death, suggesting that azacitidine can regulate KLF4 re-expression. These results support the application of azacitidine in patients with B-ALL as a therapeutic option to regulate KLF4.
    [Show full text]
  • Ten Commandments for a Good Scientist
    Unravelling the mechanism of differential biological responses induced by food-borne xeno- and phyto-estrogenic compounds Ana María Sotoca Covaleda Wageningen 2010 Thesis committee Thesis supervisors Prof. dr. ir. Ivonne M.C.M. Rietjens Professor of Toxicology Wageningen University Prof. dr. Albertinka J. Murk Personal chair at the sub-department of Toxicology Wageningen University Thesis co-supervisor Dr. ir. Jacques J.M. Vervoort Associate professor at the Laboratory of Biochemistry Wageningen University Other members Prof. dr. Michael R. Muller, Wageningen University Prof. dr. ir. Huub F.J. Savelkoul, Wageningen University Prof. dr. Everardus J. van Zoelen, Radboud University Nijmegen Dr. ir. Toine F.H. Bovee, RIKILT, Wageningen This research was conducted under the auspices of the Graduate School VLAG Unravelling the mechanism of differential biological responses induced by food-borne xeno- and phyto-estrogenic compounds Ana María Sotoca Covaleda Thesis submitted in fulfillment of the requirements for the degree of doctor at Wageningen University by the authority of the Rector Magnificus Prof. dr. M.J. Kropff, in the presence of the Thesis Committee appointed by the Academic Board to be defended in public on Tuesday 14 September 2010 at 4 p.m. in the Aula Unravelling the mechanism of differential biological responses induced by food-borne xeno- and phyto-estrogenic compounds. Ana María Sotoca Covaleda Thesis Wageningen University, Wageningen, The Netherlands, 2010, With references, and with summary in Dutch. ISBN: 978-90-8585-707-5 “Caminante no hay camino, se hace camino al andar. Al andar se hace camino, y al volver la vista atrás se ve la senda que nunca se ha de volver a pisar” - Antonio Machado – A mi madre.
    [Show full text]
  • A Dual Cis-Regulatory Code Links IRF8 to Constitutive and Inducible Gene Expression in Macrophages
    Downloaded from genesdev.cshlp.org on October 1, 2021 - Published by Cold Spring Harbor Laboratory Press A dual cis-regulatory code links IRF8 to constitutive and inducible gene expression in macrophages Alessandra Mancino,1,3 Alberto Termanini,1,3 Iros Barozzi,1 Serena Ghisletti,1 Renato Ostuni,1 Elena Prosperini,1 Keiko Ozato,2 and Gioacchino Natoli1 1Department of Experimental Oncology, European Institute of Oncology (IEO), 20139 Milan, Italy; 2Laboratory of Molecular Growth Regulation, Genomics of Differentiation Program, National Institute of Child Health and Human Development (NICHD), National Institutes of Health, Bethesda, Maryland 20892, USA The transcription factor (TF) interferon regulatory factor 8 (IRF8) controls both developmental and inflammatory stimulus-inducible genes in macrophages, but the mechanisms underlying these two different functions are largely unknown. One possibility is that these different roles are linked to the ability of IRF8 to bind alternative DNA sequences. We found that IRF8 is recruited to distinct sets of DNA consensus sequences before and after lipopolysaccharide (LPS) stimulation. In resting cells, IRF8 was mainly bound to composite sites together with the master regulator of myeloid development PU.1. Basal IRF8–PU.1 binding maintained the expression of a broad panel of genes essential for macrophage functions (such as microbial recognition and response to purines) and contributed to basal expression of many LPS-inducible genes. After LPS stimulation, increased expression of IRF8, other IRFs, and AP-1 family TFs enabled IRF8 binding to thousands of additional regions containing low-affinity multimerized IRF sites and composite IRF–AP-1 sites, which were not premarked by PU.1 and did not contribute to the basal IRF8 cistrome.
    [Show full text]
  • Geminin Deletion Increases the Number of Fetal Hematopoietic Stem Cells by Affecting the Expression of Key Transcription Factors Dimitris Karamitros1,*, Alexandra L
    © 2015. Published by The Company of Biologists Ltd | Development (2015) 142, 70-81 doi:10.1242/dev.109454 RESEARCH ARTICLE STEM CELLS AND REGENERATION Geminin deletion increases the number of fetal hematopoietic stem cells by affecting the expression of key transcription factors Dimitris Karamitros1,*, Alexandra L. Patmanidi1,*, Panoraia Kotantaki1, Alexandre J. Potocnik2, Tomi Bähr-Ivacevic3, Vladimir Benes3, Zoi Lygerou4, Dimitris Kioussis2,‡ and Stavros Taraviras1,‡ ABSTRACT hematological stress and challenges (Beerman et al., 2010; Balancing stem cell self-renewal and initiation of lineage specification Cheshier et al., 2007). The prevailing model of hematopoiesis programs is essential for the development and homeostasis of the supports the existence of long-term hematopoietic stem cells (LT- hematopoietic system. We have specifically ablated geminin in the HSCs), which can provide long-term multipotent reconstitution of developing murine hematopoietic system and observed profound the hematopoietic system, and of short-term hematopoietic stem defects in the generation of mature blood cells, leading to embryonic cells (ST-HSCs) or multipotent progenitors (MPPs), with the lethality. Hematopoietic stem cells (HSCs) accumulated in the fetal potential to generate all blood lineages but with reduced self- liver following geminin ablation, while committed progenitors were renewal capacity (Luc et al., 2007; Mebius et al., 2001; Morrison reduced. Genome-wide transcriptome analysis identified key HSC et al., 1995; Randall et al., 1996; Traver et al., 2001). transcription factors as being upregulated upon geminin deletion, Even though it remains unclear how the balance between self- revealing a gene network linked with geminin that controls fetal renewal of HSCs and fate commitment is controlled and how a hematopoiesis.
    [Show full text]
  • C/Ebpα Is an Essential Collaborator in Hoxa9/Meis1-Mediated Leukemogenesis
    C/EBPα is an essential collaborator in Hoxa9/Meis1-mediated leukemogenesis Cailin Collinsa, Jingya Wanga, Hongzhi Miaoa, Joel Bronsteina, Humaira Nawera, Tao Xua, Maria Figueroaa, Andrew G. Munteana, and Jay L. Hessa,b,1 aDepartment of Pathology, University of Michigan, Ann Arbor, MI 48109; and bIndiana University School of Medicine, Indianapolis, IN 46202 Edited* by Louis M. Staudt, National Institutes of Health, Bethesda, MD, and approved May 19, 2014 (received for review February 12, 2014) Homeobox A9 (HOXA9) is a homeodomain-containing transcrip- with Hoxa9. In addition, C/EBP recognition motifs are enriched tion factor that plays a key role in hematopoietic stem cell expan- at Hoxa9 binding sites. sion and is commonly deregulated in human acute leukemias. A C/EBPα is a basic leucine-zipper transcription factor that plays variety of upstream genetic alterations in acute myeloid leukemia a critical role in lineage commitment during hematopoietic dif- −/− (AML) lead to overexpression of HOXA9, almost always in associ- ferentiation (18). Whereas Cebpa mice show complete loss of ation with overexpression of its cofactor meis homeobox 1 (MEIS1). the granulocytic compartment, recent work shows that loss of α A wide range of data suggests that HOXA9 and MEIS1 play a syn- C/EBP in adult HSCs leads to both an increase in the number ergistic causative role in AML, although the molecular mechanisms of functional HSCs and an increase in their proliferative and leading to transformation by HOXA9 and MEIS1 remain elusive. In repopulating capacity (19, 20). Conversely, CEBPA overexpression can promote transdifferentiation of a variety of fibroblastic cells to this study, we identify CCAAT/enhancer binding protein alpha (C/ the myeloid lineage and can induce monocytic differentiation in EBPα) as a critical collaborator required for Hoxa9/Meis1-mediated α MLL-fusion protein-mediated leukemias (21, 22).
    [Show full text]
  • Appendix 2. Significantly Differentially Regulated Genes in Term Compared with Second Trimester Amniotic Fluid Supernatant
    Appendix 2. Significantly Differentially Regulated Genes in Term Compared With Second Trimester Amniotic Fluid Supernatant Fold Change in term vs second trimester Amniotic Affymetrix Duplicate Fluid Probe ID probes Symbol Entrez Gene Name 1019.9 217059_at D MUC7 mucin 7, secreted 424.5 211735_x_at D SFTPC surfactant protein C 416.2 206835_at STATH statherin 363.4 214387_x_at D SFTPC surfactant protein C 295.5 205982_x_at D SFTPC surfactant protein C 288.7 1553454_at RPTN repetin solute carrier family 34 (sodium 251.3 204124_at SLC34A2 phosphate), member 2 238.9 206786_at HTN3 histatin 3 161.5 220191_at GKN1 gastrokine 1 152.7 223678_s_at D SFTPA2 surfactant protein A2 130.9 207430_s_at D MSMB microseminoprotein, beta- 99.0 214199_at SFTPD surfactant protein D major histocompatibility complex, class II, 96.5 210982_s_at D HLA-DRA DR alpha 96.5 221133_s_at D CLDN18 claudin 18 94.4 238222_at GKN2 gastrokine 2 93.7 1557961_s_at D LOC100127983 uncharacterized LOC100127983 93.1 229584_at LRRK2 leucine-rich repeat kinase 2 HOXD cluster antisense RNA 1 (non- 88.6 242042_s_at D HOXD-AS1 protein coding) 86.0 205569_at LAMP3 lysosomal-associated membrane protein 3 85.4 232698_at BPIFB2 BPI fold containing family B, member 2 84.4 205979_at SCGB2A1 secretoglobin, family 2A, member 1 84.3 230469_at RTKN2 rhotekin 2 82.2 204130_at HSD11B2 hydroxysteroid (11-beta) dehydrogenase 2 81.9 222242_s_at KLK5 kallikrein-related peptidase 5 77.0 237281_at AKAP14 A kinase (PRKA) anchor protein 14 76.7 1553602_at MUCL1 mucin-like 1 76.3 216359_at D MUC7 mucin 7,
    [Show full text]
  • Supplementary Materials
    Supplementary Materials + - NUMB E2F2 PCBP2 CDKN1B MTOR AKT3 HOXA9 HNRNPA1 HNRNPA2B1 HNRNPA2B1 HNRNPK HNRNPA3 PCBP2 AICDA FLT3 SLAMF1 BIC CD34 TAL1 SPI1 GATA1 CD48 PIK3CG RUNX1 PIK3CD SLAMF1 CDKN2B CDKN2A CD34 RUNX1 E2F3 KMT2A RUNX1 T MIXL1 +++ +++ ++++ ++++ +++ 0 0 0 0 hematopoietic potential H1 H1 PB7 PB6 PB6 PB6.1 PB6.1 PB12.1 PB12.1 Figure S1. Unsupervised hierarchical clustering of hPSC-derived EBs according to the mRNA expression of hematopoietic lineage genes (microarray analysis). Hematopoietic-competent cells (H1, PB6.1, PB7) were separated from hematopoietic-deficient ones (PB6, PB12.1). In this experiment, all hPSCs were tested in duplicate, except PB7. Genes under-expressed or over-expressed in blood-deficient hPSCs are indicated in blue and red respectively (related to Table S1). 1 C) Mesoderm B) Endoderm + - KDR HAND1 GATA6 MEF2C DKK1 MSX1 GATA4 WNT3A GATA4 COL2A1 HNF1B ZFPM2 A) Ectoderm GATA4 GATA4 GSC GATA4 T ISL1 NCAM1 FOXH1 NCAM1 MESP1 CER1 WNT3A MIXL1 GATA4 PAX6 CDX2 T PAX6 SOX17 HBB NES GATA6 WT1 SOX1 FN1 ACTC1 ZIC1 FOXA2 MYF5 ZIC1 CXCR4 TBX5 PAX6 NCAM1 TBX20 PAX6 KRT18 DDX4 TUBB3 EPCAM TBX5 SOX2 KRT18 NKX2-5 NES AFP COL1A1 +++ +++ 0 0 0 0 ++++ +++ ++++ +++ +++ ++++ +++ ++++ 0 0 0 0 +++ +++ ++++ +++ ++++ 0 0 0 0 hematopoietic potential H1 H1 H1 H1 H1 H1 PB6 PB6 PB7 PB7 PB6 PB6 PB7 PB6 PB6 PB6.1 PB6.1 PB6.1 PB6.1 PB6.1 PB6.1 PB12.1 PB12.1 PB12.1 PB12.1 PB12.1 PB12.1 Figure S2. Unsupervised hierarchical clustering of hPSC-derived EBs according to the mRNA expression of germ layer differentiation genes (microarray analysis) Selected ectoderm (A), endoderm (B) and mesoderm (C) related genes differentially expressed between hematopoietic-competent (H1, PB6.1, PB7) and -deficient cells (PB6, PB12.1) are shown (related to Table S1).
    [Show full text]