Modeling of Neural Differentiation by Using Embryonic Stem Cells to Detect Developmental Toxicants

Total Page:16

File Type:pdf, Size:1020Kb

Modeling of Neural Differentiation by Using Embryonic Stem Cells to Detect Developmental Toxicants Modeling of neural differentiation by using embryonic stem cells to detect developmental toxicants Dissertation zur Erlangung des akademischen Grades eines Doktors der Naturwissenschaften (Dr. rer. nat.) vorgelegt von Bastian Zimmer an der Mathematisch-Naturwissenschaftliche Sektion Fachbereich Biologie Konstanz 2011 Tag der mündlichen Prüfung: 02.12.2012 1. Referent: Prof. Dr. Marcel Leist 2. Referent: PD Dr. Gerrit Begemann 3. Referent: Prof. Dr. Marcus Groettrup 4. Referent: Prof. Dr. Gabsang Lee List of publications Publications, integrated in this thesis Chapter C Zimmer B, Kuegler PB, Baudis B, Genewsky A, Tanavde V, Koh W, Tan B, Waldmann T, Kadereit S, Leist M. Coordinated waves of gene expression during neuronal differentiation of embryonic stem cells as basis for novel approaches to developmental neurotoxicity testing. Cell Death Differ. 2011 Mar;18(3):383-95. Epub 2010 Sep 24. Chapter D Zimmer B, Schildknecht S, Kuegler PB, Tanavde V, Kadereit S, Leist M. Sensitivity of dopaminergic neuron differentiation from stem cells to chronic low-dose methylmercury exposure. Toxicol Sci. 2011 Jun;121(2):357-67. Epub 2011 Mar 7. Chapter E Zimmer B, Lee G, Meganathan K, Sacchinidis A, Studer L, Leist M. Genuine human neural crest cells as test system for developmental toxicity and potential rescue strategies. under review Some text passages in the general introduction were taken from: Kadereit S*, Zimmer B*, van Thriel C, Hengstler J.G., Leist M. Compound selection for modeling developmental neurotoxicity (DNT) and prenatal neurotoxicity with embryonic stem cells. accepted for publication in Frontiers in Bioscience *: both authors contributed equally Publications, not integrated in this thesis Kuegler PB, Zimmer B, Waldmann T, Baudis B, Ilmjärv S, Hescheler J, Gaughwin P, Brundin P, Mundy W, Bal-Price AK, Schrattenholz A, Krause KH, van Thriel C, Rao MS, Kadereit S, Leist M. Markers of murine embryonic and neural stem cells, neurons and astrocytes: reference points for developmental neurotoxicity testing. ALTEX. 2010;27(1):17- 42. Kuegler PB, Baumann BA, Zimmer B, Kadereit S, Leist M, GFAP-independent inflammatory competence and trophic functions of astrocytes generated from murine embryonic stem cells, GLIA, in press Waldmann, T., Weng, M., Zimmer, B., Pöltl, D., Scholz, D., Broeg, M., Kadereit, S.,Wuellner, U., Leist, M. Extensive transcriptional regulation of chromatin modifiers during human neurodevelopment. submitted Table of contents A. Summary .............................................................................................................................. 6 Zusammenfassung.................................................................................................................... 7 B. General introduction ........................................................................................................... 9 1. (Neuro)development in general...................................................................................... 9 1.1 Proliferation.......................................................................................................... 11 1.2 Differentiation/Patterning..................................................................................... 12 1.3 Migration.............................................................................................................. 14 1.4 Neurite outgrowth................................................................................................ 15 1.5 Synaptogenesis / Neurotransmitter household ..................................................... 16 2. Environmental chemicals and (neuro)development..................................................... 17 2.1 Barker Hypothesis................................................................................................ 18 2.2 Time of insult vs. time of phenotype onset .......................................................... 18 2.3 Susceptibility of the developing brain to chemicals............................................. 19 2.4 Environmental chemicals and developmental disabilities ................................... 20 2.5 Phenotype vs. biological process ......................................................................... 22 3 Development of in vitro test systems in the 21st century ............................................. 23 4. Embryonic stem cells (ESC) as source for in-vitro testing .......................................... 24 Aims of the thesis.................................................................................................................... 27 Chapter C.................................................................................................................................... Coordinated waves of gene expression during neuronal differentiation of embryonic stem cells as basis for novel approaches to developmental neurotoxicity testing............. 29 Abstract ................................................................................................................................ 31 Introduction .......................................................................................................................... 32 Results .................................................................................................................................. 34 Discussion ............................................................................................................................ 45 Materials and Methods......................................................................................................... 49 Supplements ......................................................................................................................... 56 Chapter D.................................................................................................................................... Sensitivity of dopaminergic neuron differentiation from stem cells to chronic low-dose methylmercury exposure ....................................................................................................... 73 Abstract ................................................................................................................................ 75 Introduction .......................................................................................................................... 76 Materials and Methods......................................................................................................... 78 Results .................................................................................................................................. 82 Discussion ............................................................................................................................ 91 Supplements ......................................................................................................................... 95 Chapter E.................................................................................................................................... Genuine human neural crest cells as test system for developmental toxicity and potential rescue strategies.................................................................................................................... 101 Abstract .............................................................................................................................. 102 Introduction ........................................................................................................................ 103 Materials and Methods....................................................................................................... 105 Results ................................................................................................................................ 107 Discussion .......................................................................................................................... 114 Supplements ....................................................................................................................... 118 F. Concluding discussion ..................................................................................................... 131 G. Bibliography .................................................................................................................... 142 Record of contribution ....................................................................................................... 169 Summary A. Summary Developmental disabilities and congenital malformations associated with neural development are increasing problems in western countries. More and more evidence emerges from human epidemiological studies that environmental chemicals as well as drug and food constituents contribute to such an increase. Unfortunately, developmental neurotoxicity is currently the least examined form of developmental toxicity. Less then 200 compounds worldwide, mostly pesticides, have been tested in vivo according to the OECD test guideline TG 426. This guideline requires expensive and labor intensive animal experiments which often lack human relevance. As embryonic stem cells are able to differentiate into every cell type of an organism and have been shown to recapitulate in vivo development in the culture dish, they are considered a powerful alternative to whole animal experiments. Since also human embryonic stem cells have been generated, it is possible to mimic effects of chemicals on human neural development in vitro today. In the framework of
Recommended publications
  • Case Report Clinical Report of a 17Q12 Microdeletion with Additionally Unreported Clinical Features
    Hindawi Publishing Corporation Case Reports in Genetics Volume 2014, Article ID 264947, 6 pages http://dx.doi.org/10.1155/2014/264947 Case Report Clinical Report of a 17q12 Microdeletion with Additionally Unreported Clinical Features Jennifer L. Roberts,1 Stephanie K. Gandomi,2 Melissa Parra,2 Ira Lu,2 Chia-Ling Gau,2 Majed Dasouki,1,3 and Merlin G. Butler1 1 The University of Kansas Medical Center, Kansas City, KS 66160, USA 2 Ambry Genetics, Aliso Viejo, CA 92656, USA 3 King Faisal Specialist Hospital and Research Center, Riyadh 12713, Saudi Arabia Correspondence should be addressed to Stephanie K. Gandomi; [email protected] Received 18 March 2014; Revised 13 May 2014; Accepted 14 May 2014; Published 2 June 2014 Academic Editor: Philip D. Cotter Copyright © 2014 Jennifer L. Roberts et al. This is an open access article distributed under the Creative Commons Attribution License, which permits unrestricted use, distribution, and reproduction in any medium, provided the original work is properly cited. Copy number variations involving the 17q12 region have been associated with developmental and speech delay, autism, aggression, self-injury, biting and hitting, oppositional defiance, inappropriate language, and auditory hallucinations. We present a tall- appearing 17-year-old boy with marfanoid habitus, hypermobile joints, mild scoliosis, pectus deformity, widely spaced nipples, pes cavus, autism spectrum disorder, intellectual disability, and psychiatric manifestations including physical and verbal aggression, obsessive-compulsive behaviors, and oppositional defiance. An echocardiogram showed borderline increased aortic root size. An abdominal ultrasound revealed a small pancreas, mild splenomegaly with a 1.3 cm accessory splenule, and normal kidneys and liver.
    [Show full text]
  • A Computational Approach for Defining a Signature of Β-Cell Golgi Stress in Diabetes Mellitus
    Page 1 of 781 Diabetes A Computational Approach for Defining a Signature of β-Cell Golgi Stress in Diabetes Mellitus Robert N. Bone1,6,7, Olufunmilola Oyebamiji2, Sayali Talware2, Sharmila Selvaraj2, Preethi Krishnan3,6, Farooq Syed1,6,7, Huanmei Wu2, Carmella Evans-Molina 1,3,4,5,6,7,8* Departments of 1Pediatrics, 3Medicine, 4Anatomy, Cell Biology & Physiology, 5Biochemistry & Molecular Biology, the 6Center for Diabetes & Metabolic Diseases, and the 7Herman B. Wells Center for Pediatric Research, Indiana University School of Medicine, Indianapolis, IN 46202; 2Department of BioHealth Informatics, Indiana University-Purdue University Indianapolis, Indianapolis, IN, 46202; 8Roudebush VA Medical Center, Indianapolis, IN 46202. *Corresponding Author(s): Carmella Evans-Molina, MD, PhD ([email protected]) Indiana University School of Medicine, 635 Barnhill Drive, MS 2031A, Indianapolis, IN 46202, Telephone: (317) 274-4145, Fax (317) 274-4107 Running Title: Golgi Stress Response in Diabetes Word Count: 4358 Number of Figures: 6 Keywords: Golgi apparatus stress, Islets, β cell, Type 1 diabetes, Type 2 diabetes 1 Diabetes Publish Ahead of Print, published online August 20, 2020 Diabetes Page 2 of 781 ABSTRACT The Golgi apparatus (GA) is an important site of insulin processing and granule maturation, but whether GA organelle dysfunction and GA stress are present in the diabetic β-cell has not been tested. We utilized an informatics-based approach to develop a transcriptional signature of β-cell GA stress using existing RNA sequencing and microarray datasets generated using human islets from donors with diabetes and islets where type 1(T1D) and type 2 diabetes (T2D) had been modeled ex vivo. To narrow our results to GA-specific genes, we applied a filter set of 1,030 genes accepted as GA associated.
    [Show full text]
  • 4-6 Weeks Old Female C57BL/6 Mice Obtained from Jackson Labs Were Used for Cell Isolation
    Methods Mice: 4-6 weeks old female C57BL/6 mice obtained from Jackson labs were used for cell isolation. Female Foxp3-IRES-GFP reporter mice (1), backcrossed to B6/C57 background for 10 generations, were used for the isolation of naïve CD4 and naïve CD8 cells for the RNAseq experiments. The mice were housed in pathogen-free animal facility in the La Jolla Institute for Allergy and Immunology and were used according to protocols approved by the Institutional Animal Care and use Committee. Preparation of cells: Subsets of thymocytes were isolated by cell sorting as previously described (2), after cell surface staining using CD4 (GK1.5), CD8 (53-6.7), CD3ε (145- 2C11), CD24 (M1/69) (all from Biolegend). DP cells: CD4+CD8 int/hi; CD4 SP cells: CD4CD3 hi, CD24 int/lo; CD8 SP cells: CD8 int/hi CD4 CD3 hi, CD24 int/lo (Fig S2). Peripheral subsets were isolated after pooling spleen and lymph nodes. T cells were enriched by negative isolation using Dynabeads (Dynabeads untouched mouse T cells, 11413D, Invitrogen). After surface staining for CD4 (GK1.5), CD8 (53-6.7), CD62L (MEL-14), CD25 (PC61) and CD44 (IM7), naïve CD4+CD62L hiCD25-CD44lo and naïve CD8+CD62L hiCD25-CD44lo were obtained by sorting (BD FACS Aria). Additionally, for the RNAseq experiments, CD4 and CD8 naïve cells were isolated by sorting T cells from the Foxp3- IRES-GFP mice: CD4+CD62LhiCD25–CD44lo GFP(FOXP3)– and CD8+CD62LhiCD25– CD44lo GFP(FOXP3)– (antibodies were from Biolegend). In some cases, naïve CD4 cells were cultured in vitro under Th1 or Th2 polarizing conditions (3, 4).
    [Show full text]
  • Transcriptional Control of Tissue-Resident Memory T Cell Generation
    Transcriptional control of tissue-resident memory T cell generation Filip Cvetkovski Submitted in partial fulfillment of the requirements for the degree of Doctor of Philosophy in the Graduate School of Arts and Sciences COLUMBIA UNIVERSITY 2019 © 2019 Filip Cvetkovski All rights reserved ABSTRACT Transcriptional control of tissue-resident memory T cell generation Filip Cvetkovski Tissue-resident memory T cells (TRM) are a non-circulating subset of memory that are maintained at sites of pathogen entry and mediate optimal protection against reinfection. Lung TRM can be generated in response to respiratory infection or vaccination, however, the molecular pathways involved in CD4+TRM establishment have not been defined. Here, we performed transcriptional profiling of influenza-specific lung CD4+TRM following influenza infection to identify pathways implicated in CD4+TRM generation and homeostasis. Lung CD4+TRM displayed a unique transcriptional profile distinct from spleen memory, including up-regulation of a gene network induced by the transcription factor IRF4, a known regulator of effector T cell differentiation. In addition, the gene expression profile of lung CD4+TRM was enriched in gene sets previously described in tissue-resident regulatory T cells. Up-regulation of immunomodulatory molecules such as CTLA-4, PD-1, and ICOS, suggested a potential regulatory role for CD4+TRM in tissues. Using loss-of-function genetic experiments in mice, we demonstrate that IRF4 is required for the generation of lung-localized pathogen-specific effector CD4+T cells during acute influenza infection. Influenza-specific IRF4−/− T cells failed to fully express CD44, and maintained high levels of CD62L compared to wild type, suggesting a defect in complete differentiation into lung-tropic effector T cells.
    [Show full text]
  • Interplay Between Human Nucleolar GNL1 and RPS20 Is Critical To
    www.nature.com/scientificreports OPEN Interplay between human nucleolar GNL1 and RPS20 is critical to modulate cell proliferation Received: 19 February 2018 Rehna Krishnan, Neelima Boddapati & Sundarasamy Mahalingam Accepted: 13 July 2018 Human Guanine nucleotide binding protein like 1 (GNL1) belongs to HSR1_MMR1 subfamily of nucleolar Published: xx xx xxxx GTPases. Here, we report for the frst time that GNL1 promotes cell cycle and proliferation by inducing hyperphosphorylation of retinoblastoma protein. Using yeast two-hybrid screening, Ribosomal protein S20 (RPS20) was identifed as a functional interacting partner of GNL1. Results from GST pull-down and co-immunoprecipitation assays confrmed that interaction between GNL1 and RPS20 was specifc. Further, GNL1 induced cell proliferation was altered upon knockdown of RPS20 suggesting its critical role in GNL1 function. Interestingly, cell proliferation was signifcantly impaired upon expression of RPS20 interaction defcient GNL1 mutant suggest that GNL1 interaction with RPS20 is critical for cell growth. Finally, the inverse correlation of GNL1 and RPS20 expression in primary colon and gastric cancers with patient survival strengthen their critical importance during tumorigenesis. Collectively, our data provided evidence that cross-talk between GNL1 and RPS20 is critical to promote cell proliferation. Te YawG/YIqF/HSR1_MMR1 GTP-binding protein subfamily of GTPases is evolutionarily conserved across from prokaryotes to mammals. Te members of this family have shown to be involved in ribosomal assembly and ribosomal RNA processing and are characterized by the presence of circular permutation of guanine nucleotide binding motifs1. Te guanine nucleotide motifs G1-G5 of YawG/YIqF GTPases are arranged in G5-G4-G1-G2-G3 order whereas G1-G2-G3-G4-G5 order in classical GTPases2.
    [Show full text]
  • Increased Expression of RNA Binding Protein GNL3 Has Potential
    Increased expression of RNA binding protein GNL3 has potential influence on osteoarthritis by up-regulating IL24 and PTN Zhen Zhu1, Jun Xie1, Yawen Bian1, Upasana Manandhar1, Xiaomin Yao1, and Bo Zhang1 1Affiliation not available June 3, 2020 Abstract Osteoarthritis (OA) is a degenerative joint disorder which affects about 80% of the population above 65 years revealing ra- diographic evidence of OA. Recently, more and more researches have been conducted on the molecular mechanism of OA to find target treatments. RNA binding proteins (RBPs) play a key role in genome regulation. Nucleolar GTP-binding Protein 3 (GNL3) is abundantly expressed in bone marrow mesenchymal stem cells and correlates with chondrocytes differentiation. Here we report the transcriptome study of GNL3, which regulates transcription in osteoarthritis (OA). In this study, RNA sequencing (RNA-seq) was used to analyze the global transcription level in HeLa cells. The results showed that the expression of IL24 and PTN was down-regulated when GNL3 was knocked down. Likewise, in the lesions of osteoarthritis, the expres- sion level of GNL3, IL24 and PTN gene were significantly up-regulated compared with the control group. IL24 contributes to the progression of osteoarthritis by inducing bone cells apoptosis at the joint, and PTN contributes to the progression of osteoarthritis by promoting angiogenesis. This study aimed to assess the association between GNL3 and both of these two downstream genes as a potential biomarker for investigating the development of OA. 1 Introduction Osteoarthritis (OA) is a degenerative joint disorder characterized by progressive destruction of articular cartilage, synovial hyperplasia and subchondral osteosclerosis [1]. It is one of the main causes of disability in middle-aged and elderly people.
    [Show full text]
  • Supplementary File 2A Revised
    Supplementary file 2A. Differentially expressed genes in aldosteronomas compared to all other samples, ranked according to statistical significance. Missing values were not allowed in aldosteronomas, but to a maximum of five in the other samples. Acc UGCluster Name Symbol log Fold Change P - Value Adj. P-Value B R99527 Hs.8162 Hypothetical protein MGC39372 MGC39372 2,17 6,3E-09 5,1E-05 10,2 AA398335 Hs.10414 Kelch domain containing 8A KLHDC8A 2,26 1,2E-08 5,1E-05 9,56 AA441933 Hs.519075 Leiomodin 1 (smooth muscle) LMOD1 2,33 1,3E-08 5,1E-05 9,54 AA630120 Hs.78781 Vascular endothelial growth factor B VEGFB 1,24 1,1E-07 2,9E-04 7,59 R07846 Data not found 3,71 1,2E-07 2,9E-04 7,49 W92795 Hs.434386 Hypothetical protein LOC201229 LOC201229 1,55 2,0E-07 4,0E-04 7,03 AA454564 Hs.323396 Family with sequence similarity 54, member B FAM54B 1,25 3,0E-07 5,2E-04 6,65 AA775249 Hs.513633 G protein-coupled receptor 56 GPR56 -1,63 4,3E-07 6,4E-04 6,33 AA012822 Hs.713814 Oxysterol bining protein OSBP 1,35 5,3E-07 7,1E-04 6,14 R45592 Hs.655271 Regulating synaptic membrane exocytosis 2 RIMS2 2,51 5,9E-07 7,1E-04 6,04 AA282936 Hs.240 M-phase phosphoprotein 1 MPHOSPH -1,40 8,1E-07 8,9E-04 5,74 N34945 Hs.234898 Acetyl-Coenzyme A carboxylase beta ACACB 0,87 9,7E-07 9,8E-04 5,58 R07322 Hs.464137 Acyl-Coenzyme A oxidase 1, palmitoyl ACOX1 0,82 1,3E-06 1,2E-03 5,35 R77144 Hs.488835 Transmembrane protein 120A TMEM120A 1,55 1,7E-06 1,4E-03 5,07 H68542 Hs.420009 Transcribed locus 1,07 1,7E-06 1,4E-03 5,06 AA410184 Hs.696454 PBX/knotted 1 homeobox 2 PKNOX2 1,78 2,0E-06
    [Show full text]
  • Supplementary Table S4. FGA Co-Expressed Gene List in LUAD
    Supplementary Table S4. FGA co-expressed gene list in LUAD tumors Symbol R Locus Description FGG 0.919 4q28 fibrinogen gamma chain FGL1 0.635 8p22 fibrinogen-like 1 SLC7A2 0.536 8p22 solute carrier family 7 (cationic amino acid transporter, y+ system), member 2 DUSP4 0.521 8p12-p11 dual specificity phosphatase 4 HAL 0.51 12q22-q24.1histidine ammonia-lyase PDE4D 0.499 5q12 phosphodiesterase 4D, cAMP-specific FURIN 0.497 15q26.1 furin (paired basic amino acid cleaving enzyme) CPS1 0.49 2q35 carbamoyl-phosphate synthase 1, mitochondrial TESC 0.478 12q24.22 tescalcin INHA 0.465 2q35 inhibin, alpha S100P 0.461 4p16 S100 calcium binding protein P VPS37A 0.447 8p22 vacuolar protein sorting 37 homolog A (S. cerevisiae) SLC16A14 0.447 2q36.3 solute carrier family 16, member 14 PPARGC1A 0.443 4p15.1 peroxisome proliferator-activated receptor gamma, coactivator 1 alpha SIK1 0.435 21q22.3 salt-inducible kinase 1 IRS2 0.434 13q34 insulin receptor substrate 2 RND1 0.433 12q12 Rho family GTPase 1 HGD 0.433 3q13.33 homogentisate 1,2-dioxygenase PTP4A1 0.432 6q12 protein tyrosine phosphatase type IVA, member 1 C8orf4 0.428 8p11.2 chromosome 8 open reading frame 4 DDC 0.427 7p12.2 dopa decarboxylase (aromatic L-amino acid decarboxylase) TACC2 0.427 10q26 transforming, acidic coiled-coil containing protein 2 MUC13 0.422 3q21.2 mucin 13, cell surface associated C5 0.412 9q33-q34 complement component 5 NR4A2 0.412 2q22-q23 nuclear receptor subfamily 4, group A, member 2 EYS 0.411 6q12 eyes shut homolog (Drosophila) GPX2 0.406 14q24.1 glutathione peroxidase
    [Show full text]
  • Identification of Differentially Expressed Genes in Human Bladder Cancer Through Genome-Wide Gene Expression Profiling
    521-531 24/7/06 18:28 Page 521 ONCOLOGY REPORTS 16: 521-531, 2006 521 Identification of differentially expressed genes in human bladder cancer through genome-wide gene expression profiling KAZUMORI KAWAKAMI1,3, HIDEKI ENOKIDA1, TOKUSHI TACHIWADA1, TAKENARI GOTANDA1, KENGO TSUNEYOSHI1, HIROYUKI KUBO1, KENRYU NISHIYAMA1, MASAKI TAKIGUCHI2, MASAYUKI NAKAGAWA1 and NAOHIKO SEKI3 1Department of Urology, Graduate School of Medical and Dental Sciences, Kagoshima University, 8-35-1 Sakuragaoka, Kagoshima 890-8520; Departments of 2Biochemistry and Genetics, and 3Functional Genomics, Graduate School of Medicine, Chiba University, 1-8-1 Inohana, Chuo-ku, Chiba 260-8670, Japan Received February 15, 2006; Accepted April 27, 2006 Abstract. Large-scale gene expression profiling is an effective CKS2 gene not only as a potential biomarker for diagnosing, strategy for understanding the progression of bladder cancer but also for staging human BC. This is the first report (BC). The aim of this study was to identify genes that are demonstrating that CKS2 expression is strongly correlated expressed differently in the course of BC progression and to with the progression of human BC. establish new biomarkers for BC. Specimens from 21 patients with pathologically confirmed superficial (n=10) or Introduction invasive (n=11) BC and 4 normal bladder samples were studied; samples from 14 of the 21 BC samples were subjected Bladder cancer (BC) is among the 5 most common to microarray analysis. The validity of the microarray results malignancies worldwide, and the 2nd most common tumor of was verified by real-time RT-PCR. Of the 136 up-regulated the genitourinary tract and the 2nd most common cause of genes we detected, 21 were present in all 14 BCs examined death in patients with cancer of the urinary tract (1-7).
    [Show full text]
  • Both Sense and Antisense Strands of the LTR of the Schistosoma Mansoni Pao-Like Retrotransposon Sinbad Drive Luciferase Expression
    Mol Genet Genomics DOI 10.1007/s00438-006-0181-1 ORIGINAL PAPER Both sense and antisense strands of the LTR of the Schistosoma mansoni Pao-like retrotransposon Sinbad drive luciferase expression Claudia S. Copeland · Victoria H. Mann · Paul J. Brindley Received: 20 August 2006 / Accepted: 4 October 2006 © Springer-Verlag 2006 Abstract Long terminal repeat (LTR) retrotranspo- activity in HeLa cells. The ability of the Sinbad LTR to sons, mobile genetic elements comprising substantial transcribe in both its forward and inverted orientation proportions of many eukaryotic genomes, are so represents one of few documented examples of bidirec- named for the presence of LTRs, direct repeats about tional promotor function. 250–600 bp in length Xanking the open reading frames that encode the retrotransposon enzymes and struc- Keywords Schistosome · Boudicca · Promotor · tural proteins. LTRs include promotor functions as Promoter · Bidirectional · Inverse well as other roles in retrotransposition. LTR retro- transposons, including the Gypsy-like Boudicca and the Pao/BEL-like Sinbad elements, comprise a sub- Introduction stantial proportion of the genome of the human blood Xuke, Schistosoma mansoni. In order to deduce the Eukaryotic genomes generally contain substantial capability of speciWc copies of Boudicca and Sinbad amounts of repetitive sequences, many of which repre- LTRs to function as promotors, these LTRs were sent mobile genetic elements (e.g., Lander et al. 2001; investigated analytically and experimentally. Sequence Holt et al. 2002). These mobile elements have played analysis revealed the presence of TATA boxes, canoni- fundamental roles in host genome evolution (Charles- cal polyadenylation signals, and direct inverted repeats worth et al.
    [Show full text]
  • Antibodies Products
    Chapter 2 : Gentaur Products List • Human Signal peptidase complex catalytic subunit • Human Sjoegren syndrome nuclear autoantigen 1 SSNA1 • Human Small proline rich protein 2A SPRR2A ELISA kit SEC11A SEC11A ELISA kit SpeciesHuman ELISA kit SpeciesHuman SpeciesHuman • Human Signal peptidase complex catalytic subunit • Human Sjoegren syndrome scleroderma autoantigen 1 • Human Small proline rich protein 2B SPRR2B ELISA kit SEC11C SEC11C ELISA kit SpeciesHuman SSSCA1 ELISA kit SpeciesHuman SpeciesHuman • Human Signal peptidase complex subunit 1 SPCS1 ELISA • Human Ski oncogene SKI ELISA kit SpeciesHuman • Human Small proline rich protein 2D SPRR2D ELISA kit kit SpeciesHuman • Human Ski like protein SKIL ELISA kit SpeciesHuman SpeciesHuman • Human Signal peptidase complex subunit 2 SPCS2 ELISA • Human Skin specific protein 32 C1orf68 ELISA kit • Human Small proline rich protein 2E SPRR2E ELISA kit kit SpeciesHuman SpeciesHuman SpeciesHuman • Human Signal peptidase complex subunit 3 SPCS3 ELISA • Human SLAIN motif containing protein 1 SLAIN1 ELISA kit • Human Small proline rich protein 2F SPRR2F ELISA kit kit SpeciesHuman SpeciesHuman SpeciesHuman • Human Signal peptide CUB and EGF like domain • Human SLAIN motif containing protein 2 SLAIN2 ELISA kit • Human Small proline rich protein 2G SPRR2G ELISA kit containing protein 2 SCUBE2 ELISA kit SpeciesHuman SpeciesHuman SpeciesHuman • Human Signal peptide CUB and EGF like domain • Human SLAM family member 5 CD84 ELISA kit • Human Small proline rich protein 3 SPRR3 ELISA kit containing protein
    [Show full text]
  • Characterisation of Isomirs in Stem Cells
    Characterisation of isomiRs in stem cells Geok Chin Tan Institute of Reproductive and Developmental Biology Department of Surgery and Cancer Faculty of Medicine Imperial College London Thesis submitted to Imperial College London for the degree of Doctor of Philosophy 1 Statement of Originality All experiments included in this thesis were performed by me unless otherwise stated in the text. 2 Copyright Statement ‘The copyright of this thesis rests with the author and is made available under a Creative Commons Attribution Non-Commercial No Derivatives licence. Researchers are free to copy, distribute or transmit the thesis on the condition that they attribute it, that they do not use it for commercial purposes and that they do not alter, transform or build upon it. For any reuse or redistribution, researchers must make clear to others the licence terms of this work’ 3 Acknowledgements I would like to thank my supervisor Dr Nicholas Dibb for giving me the opportunity to work in his lab and for all of his guidance and support throughout my PhD, without which this project would not have been possible. I am also very grateful to Dr Wei Cui for teaching me the technique of stem cell culture, her comments on my project related to stem cells and as a wonderful co-supervisor. I would like to also thank Professor Malcolm Parker for his supports and advise on academic and non-academic related subjects. Many thanks to Elcie Chan for the generation of all the stem cell libraries which forms the platform for my project. My sincere thanks also to Gunter Meister for supplying the Argonaute antibodies, Leandro Castellano for the help in the design of RNA sponges, Laki Buluwela for the pTRIPz lentiviral vector and last but not least Alywn Dart from Charlotte Bevan group for the prostate cancer cell lines.
    [Show full text]