Investigation of Activated Tyrosine Kinases in Myeloproliferative Neoplasms

Total Page:16

File Type:pdf, Size:1020Kb

Investigation of Activated Tyrosine Kinases in Myeloproliferative Neoplasms Investigation of activated tyrosine kinases in myeloproliferative neoplasms by Michael Ross Marit A thesis submitted in conformity with the requirements for the degree of Doctor of Philosophy Graduate Department of Medical Biophysics University of Toronto Copyright © 2012 by Michael Ross Marit Abstract Investigation of activated tyrosine kinases in myeloproliferative neoplasms Michael Ross Marit Doctor of Philosophy Graduate Department of Medical Biophysics University of Toronto 2012 Myeloproliferative neoplasms (MPNs) are a group of disorders characterized by an excess production of a specific, fully functional blood cell type. Many cases involve deregulation of a protein tyrosine kinase. JAK2 is one such kinase, involved in a subset of MPNs. JAK2-selective inhibitors are currently being evaluated in clinical trials. In order to identify inhibitor-resistant JAK2 mutations before they appear in the clinic, we utilized TEL-JAK2 to conduct an in vitro random mutagenesis screen for JAK2 alleles resistant to JAK Inhibitor-I. Isolated mutations were evaluated for their ability to sustain cellular growth, stimulate downstream signalling pathways, and phosphorylate a novel JAK2 substrate in the presence of inhibitor. When testing the panel of mutations in the context of the Jak2 V617F allele, we observed that a subset of mutations conferred resistance to inhibitor. These results demonstrate that small-molecule inhibitors select for JAK2 inhibitor-resistant alleles. Chronic myeloid leukemia is an MPN characterized by the presence of the BCR-ABL fusion gene. We determined that a specific cohort bearing deletions near the ABL gene, which is associated with poor prognosis, do not suffer from genomic instability. We also examined the role of a putative tumour suppressor gene EXOSC2 as an explanation for the reduced survival time, and suggest it may have a role in disease progression. ii Contents Abstract ii List of Tables v List of Figures vi List of Abbreviations viii 1 Introduction 1 1.1 Hematopoiesis . .1 1.1.1 Epo signalling and erythropoiesis . .3 1.1.2 The tyrosine kinase domain and JAK2 . .3 1.1.3 JAK-STAT signalling pathway . .4 1.1.3.1 Stat activation . .5 1.1.3.2 Mitogen-activated protein kinase activation . .5 1.1.3.3 Phosphatidylinositol 3 kinase activation . .6 1.1.4 Tpo signalling and thrombopoiesis . .6 1.2 Myeloproliferative neoplasms . .8 1.3 Ph+ myeloproliferative neoplasm { CML . .9 1.3.1 CML history and the BCR-ABL translocation . 10 1.3.2 Functions of wild-type BCR and ABL . 12 1.3.3 Intracellular signalling downstream of BCR-ABL . 13 iii 1.3.4 The mouse bone marrow transplant model . 15 1.3.5 Deletions at chromosome 9q34 in CML . 18 1.3.5.1 The exosome and function of EXOSC2 . 19 1.3.5.2 Function of PRDM12 . 24 1.3.6 Historical therapies for CML . 24 1.3.7 Development and efficacy of imatinib mesylate . 25 1.3.8 Development of imatinib mesylate-insensitive relapse . 26 1.3.9 Next-generation ABL kinase inhibitors . 27 1.4 Ph{ myeloproliferative neoplasms { PV, ET, and MF . 29 1.4.1 MPN disease phenotypes . 30 1.4.2 Discovery of JAK2 V617F . 31 1.4.3 Genetic diversity among Ph{ MPN disease . 34 1.4.3.1 JAK2 exon 12 mutations . 34 1.4.3.2 MPL (Tpo-R) mutations . 35 1.4.3.3 LNK mutations . 35 1.4.3.4 TET2 mutations . 36 1.4.3.5 ASXL1 mutations . 37 1.4.3.6 EZH2 mutations . 37 1.4.3.7 JAK2 haplotype as a disease predictor . 38 1.4.3.8 Clonal diversity within disease and individual patients . 38 1.4.4 Inhibitors of JAK2 in the treatment of MPNs . 39 1.4.4.1 Ruxolitinib (INCB018424) . 40 1.4.4.2 SAR302503 (TG101348) . 41 1.4.4.3 Lestaurtinib (CEP-701) . 41 1.4.4.4 Momelotinib (CYT387) . 42 1.4.4.5 Pacritinib (SB1518) . 43 1.4.5 Mouse models for development and treatment of Ph{ MPNs . 43 iv 1.4.5.1 BMT models . 44 1.4.5.2 Transgenic models . 45 1.5 Rationale and hypothesis . 48 1.6 Thesis statement and study aims . 49 2 Random mutagenesis reveals residues of JAK2 critical in evading inhi- bition by a tyrosine kinase inhibitor 51 2.1 Abstract . 52 2.2 Introduction . 53 2.3 Materials and methods . 55 2.4 Results . 60 2.5 Discussion . 77 3 Examining Chromosome 9q34 Deletion as a Marker for Genomic Insta- bility in Chronic Myeloid Leukemia 85 3.1 Abstract . 86 3.2 Introduction . 87 3.3 Materials and methods . 89 3.4 Results . 96 3.5 Discussion . 117 4 Discussion 122 5 Concluding Remarks 136 References 138 v List of Tables 1.1 Exosome cross-kingdom protein sequence conservation . 22 2.1 Structural diagrams of selected JAK2 inhibitors . 62 2.2 JAK2 kinase domain mutations identified in an inhibitor-resistance screen 63 3.1 EXOSC2 and PRDM12 exon sequencing primers . 97 3.2 Quantitative PCR raw data from CML patient samples . 100 3.3 Quantitative PCR relative expression from CML patient samples . 101 3.4 9q34 and 22q11 status in CML samples as called by aCGH . 103 3.5 A genomic map of the 9q34 and 22q11 loci in CML patient samples . 106 3.6 CML patient copy number gains from related publications . 109 3.7 CML patient copy number losses from related publications . 110 3.8 EXOSC2 and PRDM12 exon sequencing . 116 vi List of Figures 1.1 Hematopoietic differentiation . .2 1.2 Epo signalling and JAK2 domain structure . .7 1.3 BCR-ABL structure and signalling . 16 1.4 The mammalian exosome . 23 2.1 Location of the putative JAK2 inhibitor-resistant mutations . 64 2.2 JAK2 mutations display resistance to JAK inhibitor-I . 65 2.3 TEL-JAK2 mutants are not resistant to TG101348 or CEP-701 . 66 2.4 TEL-JAK2 inhibitor-resistant mutants display enhanced phosphorylation of Stat5, Akt and Erk1/2 - I . 68 2.5 TEL-JAK2 inhibitor-resistant mutants display enhanced phosphorylation of Stat5, Akt and Erk1/2 - II . 69 2.6 TEL-JAK2 and Jak2 V617F phosphorylate JAK2 substrate activation loop sequences . 70 2.7 TEL-JAK2 mutants G935R and R975G display a strong degree of inhibitor resistance . 72 2.8 Jak2 V617F G935R is resistant to JAK Inhibitor-I . 74 2.9 Jak2 V617F G935R is not resistant to TG101348 or CEP-701 . 75 2.10 Jak2 V617F G935R displays enhanced Stat5 and Erk1/2 phosphorylation 76 2.11 Jak2 V617F G935R displays a strong degree of inhibitor resistance . 78 vii 2.12 JAK2 inhibitor-resistant residues mapped to the crystal structure bound to JAK Inhibitor-I . 82 2.13 Inhibitor-resistant mutations identified in mJak1 and hJAK2 mutagenesis screens . 84 3.1 EXOSC2 and PRDM12 quantitative PCR standard curves . 99 3.2 Nexus software visualization of the 9q34 locus in CML patient samples . 104 3.3 Nexus software visualization of the 22q11 locus in CML patient samples . 105 3.4 Nexus-generated virtual karyotype from CML patient samples . 108 3.5 Genomic loss at 12q13.32 in the RB1 tumour suppressor gene . 112 3.6 EXOSC2 antibody validation . 114 3.7 Exosc2 and L32 expression in hematopoietic progenitors . 115 viii List of Abbreviations 5-FU 5-fluorouracil BMT Bone marrow transplant 5mC 5-methylcytosine c-abl ABL1 tyrosine kinase, human orthologue ABL Abelson murine leukemia viral oncogene homo- c-mpl Thrombopoietin recep- logue tor, human orthologue CC Coiled coil aCGH Array comparative ge- nomic hybridization cDNA Complementary DNA AML Acute myeloid leukemia CFU-E Colony-forming unit, ery- throid ARE AU-rich element CLP Common lymphoid pro- ARED AU-rich element genitor database CML Chronic myeloid leu- ATP Adenosine tri-phosphate kemia CMML Chronic monomyelocytic aUPD Acquired uniparental dis- leukemia omy CMP Common myeloid pro- B-ALL B-cell acute lymphoblas- genitor tic leukemia der(9) Derivative chromosome 9 BAC Bacterial artificial chro- mosome DH DBL homology DNA Deoxyribonucleic acid BCR Breakpoint cluster region dNTP Dinucleotide triphos- BCR-ABL Fusion gene created with phate BCR and ABL1 ENU N-ethyl-N-nitrosourea BFU-E Burst-forming unit, ery- throid EPO Erythropoietin Epo Erythropoietin BID Bis in die (Latin: twice a day) EPO-R Erythropoietin receptor ix ET Essential thrombo- HSC Hematopoietic stem cell cythemia IFN-α Interferon alpha EXOSC Exosome sub-component IFN-γ Interferon gamma FCS Fetal calf serum IgG Immunoglobulin G FDA Food and drug adminis- IL-3 Interleukin 3 tration IL-3-R Interleukin 3 receptor FERM 4.1 / ezrin / radixin / moesin IL-6 Interleukin 6 FF1 Flip-flop1 IM Imatinib mesylate FISH Fluorescent in situ hy- ITD Internal tandem duplica- bridization tion G-CSF Granulocyte colony stim- JAK Janus kinase ulating factor JH1 JAK kinase domain GAS Gamma activated se- quence JH2 JAK dual-specificity kin- ase domain GDP Guanine diphosphate JI1 JAK Inhibitor-I GM-CSF Granulocyte macrophage colony stimulating factor LOH Loss of heterozygosity GMP Granulocyte macrophage LP Lymphoid progenitor progenitor LSK Lineage{, sca-1+, c-kit+ GST Glutathione-S-transferase MAP Mitogen activated pro- tein GST-J2s Fusion of glutathione-S- transferase and the 11 MAPK Mitogen activated pro- amino acid JAK2 sub- tein kinase strate Mast Mast cell GTP Guanine triphosphate MDR Minimal deleted region HCT Hematocrit MEP Megakaryocyte erythro- hmC 5-hydroxymethylcytosine cyte progenitor hopTum-1 HopscotchTumorous- MF Myelofibrosis lethal mJak Murine Jak orthologue HR Homologous repair MMEJ Microhomology-mediated HRP Horseradish peroxidase end joining x MPL Thrombopoietin receptor PRC2 Polycomb repressive complex 2 MPN Myeloproliferative neo- plasm / myeloprolifera- PRDM Positive regulatory do- tive disease main member NHEJ Non-homologous end PTK Protein tyrosine kinase joining PV Polycythemia vera NK Natural killer cell qPCR Quantitative real-time PCR p190 The 190 kDa isoform of BCR-ABL qRT-PCR Quantitative real-time PCR p210 The 210 kDa isoform of BCR-ABL RB1 Retinoblastoma
Recommended publications
  • 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]
  • Viewed in [2, 3])
    Yildiz et al. Neural Development (2019) 14:5 https://doi.org/10.1186/s13064-019-0129-x RESEARCH ARTICLE Open Access Zebrafish prdm12b acts independently of nkx6.1 repression to promote eng1b expression in the neural tube p1 domain Ozge Yildiz1, Gerald B. Downes2 and Charles G. Sagerström1* Abstract Background: Functioning of the adult nervous system depends on the establishment of neural circuits during embryogenesis. In vertebrates, neurons that make up motor circuits form in distinct domains along the dorsoventral axis of the neural tube. Each domain is characterized by a unique combination of transcription factors (TFs) that promote a specific fate, while repressing fates of adjacent domains. The prdm12 TF is required for the expression of eng1b and the generation of V1 interneurons in the p1 domain, but the details of its function remain unclear. Methods: We used CRISPR/Cas9 to generate the first germline mutants for prdm12 and employed this resource, together with classical luciferase reporter assays and co-immunoprecipitation experiments, to study prdm12b function in zebrafish. We also generated germline mutants for bhlhe22 and nkx6.1 to examine how these TFs act with prdm12b to control p1 formation. Results: We find that prdm12b mutants lack eng1b expression in the p1 domain and also possess an abnormal touch-evoked escape response. Using luciferase reporter assays, we demonstrate that Prdm12b acts as a transcriptional repressor. We also show that the Bhlhe22 TF binds via the Prdm12b zinc finger domain to form a complex. However, bhlhe22 mutants display normal eng1b expression in the p1 domain. While prdm12 has been proposed to promote p1 fates by repressing expression of the nkx6.1 TF, we do not observe an expansion of the nkx6.1 domain upon loss of prdm12b function, nor is eng1b expression restored upon simultaneous loss of prdm12b and nkx6.1.
    [Show full text]
  • NICU Gene List Generator.Xlsx
    Neonatal Crisis Sequencing Panel Gene List Genes: A2ML1 - B3GLCT A2ML1 ADAMTS9 ALG1 ARHGEF15 AAAS ADAMTSL2 ALG11 ARHGEF9 AARS1 ADAR ALG12 ARID1A AARS2 ADARB1 ALG13 ARID1B ABAT ADCY6 ALG14 ARID2 ABCA12 ADD3 ALG2 ARL13B ABCA3 ADGRG1 ALG3 ARL6 ABCA4 ADGRV1 ALG6 ARMC9 ABCB11 ADK ALG8 ARPC1B ABCB4 ADNP ALG9 ARSA ABCC6 ADPRS ALK ARSL ABCC8 ADSL ALMS1 ARX ABCC9 AEBP1 ALOX12B ASAH1 ABCD1 AFF3 ALOXE3 ASCC1 ABCD3 AFF4 ALPK3 ASH1L ABCD4 AFG3L2 ALPL ASL ABHD5 AGA ALS2 ASNS ACAD8 AGK ALX3 ASPA ACAD9 AGL ALX4 ASPM ACADM AGPS AMELX ASS1 ACADS AGRN AMER1 ASXL1 ACADSB AGT AMH ASXL3 ACADVL AGTPBP1 AMHR2 ATAD1 ACAN AGTR1 AMN ATL1 ACAT1 AGXT AMPD2 ATM ACE AHCY AMT ATP1A1 ACO2 AHDC1 ANK1 ATP1A2 ACOX1 AHI1 ANK2 ATP1A3 ACP5 AIFM1 ANKH ATP2A1 ACSF3 AIMP1 ANKLE2 ATP5F1A ACTA1 AIMP2 ANKRD11 ATP5F1D ACTA2 AIRE ANKRD26 ATP5F1E ACTB AKAP9 ANTXR2 ATP6V0A2 ACTC1 AKR1D1 AP1S2 ATP6V1B1 ACTG1 AKT2 AP2S1 ATP7A ACTG2 AKT3 AP3B1 ATP8A2 ACTL6B ALAS2 AP3B2 ATP8B1 ACTN1 ALB AP4B1 ATPAF2 ACTN2 ALDH18A1 AP4M1 ATR ACTN4 ALDH1A3 AP4S1 ATRX ACVR1 ALDH3A2 APC AUH ACVRL1 ALDH4A1 APTX AVPR2 ACY1 ALDH5A1 AR B3GALNT2 ADA ALDH6A1 ARFGEF2 B3GALT6 ADAMTS13 ALDH7A1 ARG1 B3GAT3 ADAMTS2 ALDOB ARHGAP31 B3GLCT Updated: 03/15/2021; v.3.6 1 Neonatal Crisis Sequencing Panel Gene List Genes: B4GALT1 - COL11A2 B4GALT1 C1QBP CD3G CHKB B4GALT7 C3 CD40LG CHMP1A B4GAT1 CA2 CD59 CHRNA1 B9D1 CA5A CD70 CHRNB1 B9D2 CACNA1A CD96 CHRND BAAT CACNA1C CDAN1 CHRNE BBIP1 CACNA1D CDC42 CHRNG BBS1 CACNA1E CDH1 CHST14 BBS10 CACNA1F CDH2 CHST3 BBS12 CACNA1G CDK10 CHUK BBS2 CACNA2D2 CDK13 CILK1 BBS4 CACNB2 CDK5RAP2
    [Show full text]
  • Supplementary Material and Methods
    Supplementary material and methods Generation of cultured human epidermal sheets Normal human epidermal keratinocytes were isolated from human breast skin. Keratinocytes were grown on a feeder layer of irradiated human fibroblasts pre-seeded at 4000 cells /cm² in keratinocyte culture medium (KCM) containing a mix of 3:1 DMEM and HAM’s F12 (Invitrogen, Carlsbad, USA), supplemented with 10% FCS, 10ng/ml epidermal growth factor (EGF; R&D systems, Minneapolis, MN, USA), 0.12 IU/ml insulin (Lilly, Saint- Cloud, France), 0.4 mg/ml hydrocortisone (UpJohn, St Quentin en Yvelelines, France) , 5 mg/ml triiodo-L- thyronine (Sigma, St Quentin Fallavier, France), 24.3 mg/ml adenine (Sigma), isoproterenol (Isuprel, Hospira France, Meudon, France) and antibiotics (20 mg/ml gentamicin (Phanpharma, Fougères, France), 100 IU/ml penicillin (Phanpharma), and 1 mg/ml amphotericin B (Phanpharma)). The medium was changed every two days. NHEK were then cultured over a period of 13 days according to the protocol currently used at the Bank of Tissues and Cells for the generation of clinical grade epidermal sheets used for the treatment of severe extended burns (Ref). When needed, cells were harvested with trypsin-EDTA 0.05% (Thermo Fisher Scientific, Waltham, MA, USA) and collected for analysis. Clonogenic assay Keratinocytes were seeded on a feeder layer of irradiated fibroblasts, at a clonal density of 10-20 cells/cm² and cultivated for 10 to 14 days. Three flasks per tested condition were fixed and colored in a single 30 mns step using rhodamine B (Sigma) diluted at 0.01 g/ml in 4% paraformaldehyde. In each tested condition, cells from 3 other flasks were numerated after detachment by trypsin treatment.
    [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]
  • Developmental Biology 399 (2015) 164–176
    Developmental Biology 399 (2015) 164–176 Contents lists available at ScienceDirect Developmental Biology journal homepage: www.elsevier.com/locate/developmentalbiology The requirement of histone modification by PRDM12 and Kdm4a for the development of pre-placodal ectoderm and neural crest in Xenopus Shinya Matsukawa a, Kyoko Miwata b, Makoto Asashima b, Tatsuo Michiue a,n a Department of Sciences (Biology), Graduate School of Arts and Sciences, University of Tokyo, 3-8-1 Komaba, Meguro-ku, Tokyo 153-8902, Japan b Research Center for Stem Cell Engineering National Institute of Advanced Industrial Science and Technology (AIST), Tsukuba City, Ibaraki, Japan article info abstract Article history: In vertebrates, pre-placodal ectoderm and neural crest development requires morphogen gradients and Received 6 September 2014 several transcriptional factors, while the involvement of histone modification remains unclear. Here, we Received in revised form report that histone-modifying factors play crucial roles in the development of pre-placodal ectoderm 21 November 2014 and neural crest in Xenopus. During the early neurula stage, PRDM12 was expressed in the lateral pre- Accepted 23 December 2014 placodal ectoderm and repressed the expression of neural crest specifier genes via methylation of Available online 6 January 2015 histone H3K9. ChIP-qPCR analyses indicated that PRDM12 promoted the occupancy of the trimethylated Keywords: histone H3K9 (H3K9me3) on the Foxd3, Slug, and Sox8 promoters. Injection of the PRDM12 MO inhibited fi Histone modi cation the expression of presumptive trigeminal placode markers and decreased the occupancy of H3K9me3 on PRDM12 the Foxd3 promoter. Histone demethylase Kdm4a also inhibited the expression of presumptive Kdm4a trigeminal placode markers in a similar manner to PRDM12 MO and could compensate for the effects Pre-placodal ectoderm Neural crest of PRDM12.
    [Show full text]
  • In Vitro Differentiation of Human Skin-Derived Cells Into Functional
    cells Article In Vitro Differentiation of Human Skin-Derived Cells into Functional Sensory Neurons-Like Adeline Bataille 1 , Raphael Leschiera 1, Killian L’Hérondelle 1, Jean-Pierre Pennec 2, Nelig Le Goux 3, Olivier Mignen 3, Mehdi Sakka 1, Emmanuelle Plée-Gautier 1,4 , Cecilia Brun 5 , Thierry Oddos 5, Jean-Luc Carré 1,4, Laurent Misery 1,6 and Nicolas Lebonvallet 1,* 1 EA4685 Laboratory of Interactions Neurons-Keratinocytes, Faculty of Medicine and Health Sciences, University of Western Brittany, F-29200 Brest, France; [email protected] (A.B.); [email protected] (R.L.); [email protected] (K.L.); [email protected] (M.S.); [email protected] (E.P.-G.); [email protected] (J.-L.C.); [email protected] (L.M.) 2 EA 4324 Optimization of Physiological Regulation, Faculty of Medicine and Health Sciences, University of Western Brittany, F-29200 Brest, France; [email protected] 3 INSERM UMR1227 B Lymphocytes and autoimmunity, University of Western Brittany, F-29200 Brest, France; [email protected] (N.L.G.); [email protected] (O.M.) 4 Department of Biochemistry and Pharmaco-Toxicology, University Hospital of Brest, 29609 Brest, France 5 Johnson & Johnson Santé Beauté France Upstream Innovation, F-27100 Val de Reuil, France; [email protected] (C.B.); [email protected] (T.O.) 6 Department of Dermatology, University Hospital of Brest, 29609 Brest, France * Correspondence: [email protected]; Tel.: +33-2-98-01-21-81 Received: 23 March 2020; Accepted: 14 April 2020; Published: 17 April 2020 Abstract: Skin-derived precursor cells (SKPs) are neural crest stem cells that persist in certain adult tissues, particularly in the skin.
    [Show full text]
  • Integrated Transcriptomic, Phenotypic, and Functional Study Reveals Tissue-Specific Immune Properties of Mesenchymal Stromal
    Integrated Transcriptomic, Phenotypic, and Functional Study Reveals Tissue-Specific Immune Properties of Mesenchymal Stromal Cells Cédric Ménard, Joelle Dulong, David Roulois, Benjamin Hebraud, Léa Verdière, Céline Pangault, Vonick Sibut, Isabelle Bezier, Nadège Bescher, Céline Monvoisin, et al. To cite this version: Cédric Ménard, Joelle Dulong, David Roulois, Benjamin Hebraud, Léa Verdière, et al.. Inte- grated Transcriptomic, Phenotypic, and Functional Study Reveals Tissue-Specific Immune Prop- erties of Mesenchymal Stromal Cells. STEM CELLS, AlphaMed Press, 2020, 38 (1), pp.146-159. 10.1002/stem.3077. hal-02282131 HAL Id: hal-02282131 https://hal-univ-rennes1.archives-ouvertes.fr/hal-02282131 Submitted on 10 Sep 2019 HAL is a multi-disciplinary open access L’archive ouverte pluridisciplinaire HAL, est archive for the deposit and dissemination of sci- destinée au dépôt et à la diffusion de documents entific research documents, whether they are pub- scientifiques de niveau recherche, publiés ou non, lished or not. The documents may come from émanant des établissements d’enseignement et de teaching and research institutions in France or recherche français ou étrangers, des laboratoires abroad, or from public or private research centers. publics ou privés. Stem Cells Integrated transcriptomic, phenotypic, and functional study reveals tissue-specific immune properties of mesenchymal stromal cells Journal: Stem Cells Manuscript ID Draft Wiley - Manuscript Type: Original Research Date Submitted by Forthe Peer Review n/a Author: Complete
    [Show full text]
  • Transcriptional Regulator PRDM12 Is Essential for Human Pain
    LETTERS Transcriptional regulator PRDM12 is essential for human pain perception Ya-Chun Chen1,2,52, Michaela Auer-Grumbach3,52, Shinya Matsukawa4, Manuela Zitzelsberger5, Andreas C Themistocleous6,7, Tim M Strom8,9, Chrysanthi Samara10, Adrian W Moore11, Lily Ting-Yin Cho12, Gareth T Young12, Caecilia Weiss5, Maria Schabhüttl3, Rolf Stucka5, Annina B Schmid6,13, Yesim Parman14, Luitgard Graul-Neumann15, Wolfram Heinritz16,17, Eberhard Passarge17,18, Rosemarie M Watson19, Jens Michael Hertz20, Ute Moog21, Manuela Baumgartner22, Enza Maria Valente23, Diego Pereira24, Carlos M Restrepo25, Istvan Katona26, Marina Dusl5, Claudia Stendel5,27, Thomas Wieland8, Fay Stafford1,2, Frank Reimann28, Katja von Au29, Christian Finke30, Patrick J Willems31, Michael S Nahorski1,2, Samiha S Shaikh1,2, Ofélia P Carvalho1,2, Adeline K Nicholas2, Gulshan Karbani32, Maeve A McAleer19, Maria Roberta Cilio33,34, John C McHugh35, Sinead M Murphy36,37, Alan D Irvine19,38, Uffe Birk Jensen39, Reinhard Windhager3, Joachim Weis26, Carsten Bergmann40–42, Bernd Rautenstrauss5,43, Jonathan Baets44–46, Peter De Jonghe44–46, Mary M Reilly47, Regina Kropatsch48, Ingo Kurth49, Roman Chrast9,50,51, Tatsuo Michiue4, David L H Bennett6, C Geoffrey Woods1,2 & Jan Senderek5 Pain perception has evolved as a warning mechanism to alert exome sequencing on the index patient of family A and the unre- organisms to tissue damage and dangerous environments1,2. lated single CIP patient from family B. Although exome sequenc- In humans, however, undesirable, excessive or chronic pain ing of the subject from family A yielded no obvious pathogenic is a common and major societal burden for which available variant in genes located in the autozygous region on chromosome 9, medical treatments are currently suboptimal3,4.
    [Show full text]
  • A Single Cell Transcriptional Atlas of Early Synovial Joint Development Qin Bian1,2, Yu-Hao Cheng1,3, Jordan P Wilson1, Dong Wo
    bioRxiv preprint doi: https://doi.org/10.1101/2019.12.23.887208; this version posted December 23, 2019. The copyright holder for this preprint (which was not certified by peer review) is the author/funder, who has granted bioRxiv a license to display the preprint in perpetuity. It is made available under aCC-BY-NC-ND 4.0 International license. 1 2 3 A single cell transcriptional atlas of early synovial joint development 4 5 1,2 1,3 1 4 4 6 Qin Bian , Yu-Hao Cheng , Jordan P Wilson , Dong Won Kim , Hong Wang , 1,4 1,2,3 7 Seth Blackshaw , Patrick Cahan 8 9 10 11 12 13 14 1Institute for Cell Engineering, Johns Hopkins School of Medicine, Baltimore MD 21205 15 USA 16 17 2Department of Biomedical Engineering, Johns Hopkins School of Medicine, Baltimore 18 MD 21205 USA 19 20 3Department of Molecular Biology and Genetics, Johns Hopkins School of Medicine, 21 Baltimore MD 21205 USA 22 23 4Solomon H. Snyder Department of Neuroscience, Johns Hopkins School of Medicine, 24 Baltimore MD 21205 USA 25 26 27 28 29 30 Correspondence to: [email protected] 31 32 33 34 35 36 37 38 39 40 41 Keywords: synovial joint development; interzone; single cell RNA-Seq; articular cartilage; 42 ligament; synovium; chondrocyte; meniscus; Gdf5 43 1 bioRxiv preprint doi: https://doi.org/10.1101/2019.12.23.887208; this version posted December 23, 2019. The copyright holder for this preprint (which was not certified by peer review) is the author/funder, who has granted bioRxiv a license to display the preprint in perpetuity.
    [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]
  • Repurposing of KLF5 Activates a Cell Cycle Signature During The
    RESEARCH ARTICLE Repurposing of KLF5 activates a cell cycle signature during the progression from a precursor state to oesophageal adenocarcinoma Connor Rogerson1, Samuel Ogden1, Edward Britton1, The OCCAMS Consortium, Yeng Ang2,3*, Andrew D Sharrocks1* 1School of Biological Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Manchester, United Kingdom; 2School of Medical Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Manchester, United Kingdom; 3GI Science Centre, Salford Royal NHS FT, University of Manchester, Salford, United Kingdom Abstract Oesophageal adenocarcinoma (OAC) is one of the most common causes of cancer deaths. Barrett’s oesophagus (BO) is the only known precancerous precursor to OAC, but our understanding about the molecular events leading to OAC development is limited. Here, we have integrated gene expression and chromatin accessibility profiles of human biopsies and identified a strong cell cycle gene expression signature in OAC compared to BO. Through analysing associated chromatin accessibility changes, we have implicated the transcription factor KLF5 in the transition from BO to OAC. Importantly, we show that KLF5 expression is unchanged during this transition, but instead, KLF5 is redistributed across chromatin to directly regulate cell cycle genes specifically in OAC cells. This new KLF5 target gene programme has potential prognostic significance as high *For correspondence: levels correlate with poorer patient survival. Thus, the repurposing of KLF5 for novel regulatory [email protected] (YA); activity in OAC provides new insights into the mechanisms behind disease progression. andrew.d.sharrocks@manchester. ac.uk (ADS) Competing interests: The authors declare that no Introduction competing interests exist. Oesophageal cancer is the eighth most common cancer worldwide, and its 5-year survival rate of Funding: See page 18 15% makes it the sixth most-common cause of cancer-related death (Ferlay et al., 2015; Received: 24 March 2020 Pennathur et al., 2013).
    [Show full text]