The Role of Transcription Factors Sox4 and Sox11 in Mouse Heart Development

Total Page:16

File Type:pdf, Size:1020Kb

The Role of Transcription Factors Sox4 and Sox11 in Mouse Heart Development The role of transcription factors Sox4 and Sox11 in mouse heart development Die Rolle der Transkriptionsfaktoren Sox4 und Sox11 während der Herzentwicklung der Maus Der Naturwissenschaftlichen Fakultät der Friedrich‐Alexander‐Universität Erlangen‐Nürnberg zur Erlangung des Doktorgrades Dr. rer. nat. vorgelegt von Mandy Paul aus Bautzen Als Dissertation genehmigt von der Naturwissenschaftlichen Fakultät der Friedrich‐Alexander Universität Erlangen‐Nürnberg. Tag der mündlichen Prüfung: 09.10.2014 Vorsitzende/r des Promotionsorgans: Prof. Dr. Johannes Barth Gutachter/in: Prof. Dr. Michael Wegner PD Dr. Dieter Engelkamp “The scientist is not a person who gives the right answers, he is one who asks the right questions” Claude Lévi‐Strauss Meiner Familie gewidmet I. Table of content SUMMARY ............................................................................................. I ZUSAMMENFASSUNG.......................................................................... III 1. INTRODUCTION ................................................................................ 1 1.1 CARDIAC DEVELOPMENT ..................................................................................................... 1 1.1.1 MORPHOLOGICAL DEVELOPMENT OF THE HEART ‐ AN OVERVIEW ................................................ 1 1.1.2 CARDIAC SEPTATION .......................................................................................................... 4 1.1.2.1 ATRIOVENTRICULAR CANAL AND ATRIAL SEPTATION ................................................................. 4 1.1.2.2 VENTRICULAR GROWTH AND SEPTATION ................................................................................ 5 1.1.3 OUTFLOW TRACT DEVELOPMENT ........................................................................................... 6 1.1.3.1 MORPHOLOGICAL DEVELOPMENT AND SEPTATION ................................................................... 6 1.1.3.2 CONTRIBUTION OF NEURAL CREST CELLS TO OUTFLOW TRACT MORPHOLOGY ............................... 8 1.1.4 OUTFLOW TRACT MALFORMATIONS ...................................................................................... 9 1.2 SOX PROTEIN FAMILY ........................................................................................................ 12 1.2.1 SOXC PROTEINS ............................................................................................................... 13 1.2.1.1 SOX4 ............................................................................................................................ 14 1.2.1.2 SOX11 .......................................................................................................................... 15 1.2.1.3 SOX12 .......................................................................................................................... 17 2. AIM OF THE STUDY ......................................................................... 19 3. RESULTS .......................................................................................... 21 3.1 SOX4 AND SOX11 FUNCTION IN CARDIAC OUTFLOW TRACT DEVELOPMENT .................................. 21 3.1.1 SOX4 AND SOX11 FUNCTION IN CARDIAC NEURAL CREST CELLS ................................................. 21 3.1.1.1 MACROSCOPIC ANALYSIS OF OUTFLOW TRACT DEVELOPMENT IN MICE WITH SOX4 AND SOX11 ABLATION IN CARDIAC NEURAL CREST CELLS ........................................................................... 21 3.1.1.2 DETAILED HISTOLOGICAL ANALYSIS OF OUTFLOW TRACT DEVELOPMENT IN MICE WITH SOX4 AND SOX11 ABLATION IN CARDIAC NEURAL CREST CELLS ................................................................ 24 3.1.1.3 DETAILED HISTOLOGICAL ANALYSIS OF VENTRICULAR SEPTUM DEVELOPMENT IN MICE WITH SOX4 AND SOX11 ABLATION IN CARDIAC NEURAL CREST CELLS ................................................................ 26 3.1.1.4 APPEARANCE OF ANIMALS WITH A SPECIFIC ABLATION OF SOX4 AND SOX11 IN NEURAL CREST CELLS 27 3.1.1.5 ANALYSIS OF EARLY OUTFLOW TRACT DEVELOPMENT IN MICE WITH SOX4 ABLATION IN CARDIAC NEURAL CREST CELLS VIA AP2::CRE ................................................................................... 28 3.1.1.6 EFFICIENT DELETION OF SOX4 AND SOX11 IN NEURAL CREST CELLS ............................................ 28 3.1.2 SOX4 AND SOX11 FUNCTION IN MESODERMAL CELLS .............................................................. 30 3.1.2.1 ANALYSIS OF EARLY OUTFLOW TRACT DEVELOPMENT IN MICE WITH SOX4 AND SOX11 ABLATION IN MESODERMAL CELLS ......................................................................................................... 30 3.1.2.2 ANALYSIS OF LATE OUTFLOW TRACT DEVELOPMENT IN MICE WITH SOX4 AND SOX11 ABLATION IN MESODERMAL CELLS ......................................................................................................... 31 3.1.3 ANALYSIS OF OUTFLOW TRACT DEVELOPMENT IN MICE WITH SOX4 AND SOX11 ABLATION IN CARDIAC NEURAL CREST AND MESODERMAL CELLS .............................................................................. 33 3.1.4 ANALYSIS OF EARLY OUTFLOW TRACT DEVELOPMENT IN MICE WITH SOX4 AND SOX11 ABLATION IN ENDOTHELIAL CELLS .......................................................................................................... 34 3.1.5 PROPERTIES OF CARDIAC NEURAL CREST CELLS IN THE ABSENCE OF SOX4 AND SOX11 .................... 35 3.1.6 ABLATION OF SOX4 AND SOX11 LEADS TO MORPHOLOGICAL CHANGES IN CARDIAC NEURAL CREST CELLS ............................................................................................................................ 38 3.1.7. SOX12 EXPRESSION IN THE OUTFLOW TRACT ......................................................................... 39 3.1.8 ABLATION OF SOX4 AND SOX11 LEADS TO ALTERED MARKER GENE EXPRESSION IN CARDIAC NEURAL CREST CELLS .................................................................................................................... 39 3.1.8.1 IMMUNOHISTOCHEMICAL ANALYSIS OF MARKER GENE EXPRESSION OF CARDIAC NEURAL CREST CELLS IN MICE WITH SOX4 AND SOX11 ABLATION IN NEURAL CREST CELLS ............................................... 39 3.1.8.2 QUANTITATIVE RT‐PCR ANALYSIS OF MARKER GENE EXPRESSION OF CARDIAC NEURAL CREST CELLS IN MICE WITH SOX4 AND SOX11 ABLATION IN NEURAL CREST CELLS ............................................... 42 3.1.9 PUTATIVE SOX4 AND SOX11 BINDING SITES IN ADAM19 AND E‐CADHERIN PROMOTER REGIONS ..... 43 3.1.10 DIRECT BINDING OF SOX4 AND SOX11 TO NEWLY IDENTIFIED SOX BINDING SITES IN ADAM19 AND E‐ CADHERIN PROMOTER REGIONS .......................................................................................... 44 3.1.11 SOX11 ASSOCIATES WITH ENDOGENOUS ADAM19 AND E‐CADHERIN PROMOTER REGION .............. 46 3.1.12 SOX4 AND SOX11 ACTIVATE ADAM19 AND E‐CADHERIN PROMOTERS ....................................... 49 3.2 POSTTRANSLATIONAL MODIFICATION OF SOX4 AND SOX11 ..................................................... 51 3.2.1 PUTATIVE PHOSPHORYLATION SITES OF SOX4 AND SOX11 ....................................................... 51 3.2.2 PHOSPHORYLATION ANALYSIS OF SOX4 AND SOX11 ............................................................... 52 3.2.3 ANALYSIS OF ACTIVATION POTENTIAL OF PUTATIVE SOXC PHOSPHORYLATION MUTANTS ................ 54 3.2.4 ANALYSIS OF BINDING ABILITY OF PUTATIVE SOXC PHOSPHORYLATION MUTANTS ......................... 55 4. DISCUSSION ............................................................................... 57 4.1 THE ROLE OF SOXC PROTEINS IN OUTFLOW TRACT DEVELOPMENT .............................................. 57 4.1.1 THE ROLE OF SOX4 IN OUTFLOW TRACT DEVELOPMENT ........................................................... 57 4.1.2 THE ROLE OF SOX11 IN OUTFLOW TRACT DEVELOPMENT ......................................................... 58 4.1.3 REDUNDANT FUNCTION OF SOX4 AND SOX11 IN OUTFLOW TRACT DEVELOPMENT ........................ 59 4.1.4 THE ROLE OF SOX12 IN OUTFLOW TRACT DEVELOPMENT ......................................................... 60 4.2 PARTICIPATION OF OTHER SOX PROTEINS IN HEART DEVELOPMENT ............................................. 61 4.3 THE ROLE OF SOXC PROTEINS IN OTHER TISSUES ..................................................................... 62 4.4 PROLIFERATION, MIGRATION AND APOPTOSIS OF NEURAL CREST CELLS ARE NOT AFFECTED BY SOXC DELETION ....................................................................................................................... 63 4.5 MORPHOLOGICAL AND CYTOSKELETAL ALTERATIONS IN SOXC DEFICIENT CARDIAC NEURAL CREST CELLS .................................................................................................................................... 63 4.6 ALTERED TRANSCRIPTION FACTOR EXPRESSION IN OUTFLOW TRACT REGIONS CONTAINING SOXC DEFICIENT CARDIAC NEURAL CREST CELLS .............................................................................. 65 4.7 ADAM19: A DIRECT TARGET OF SOXC PROTEINS .................................................................... 66 4.8 ALTERED EMT AND MET MARKER GENE EXPRESSION IN OUTFLOW TRACT REGIONS CONTAINING SOXC DEFICIENT CARDIAC NEURAL CREST CELLS .............................................................................. 67 4.9 E‐CADHERIN: A DIRECT TARGET OF SOXC PROTEINS ................................................................ 68 4.10 RELEVANCE IN DISEASE ..................................................................................................... 69 4.11 OUTLOOK .....................................................................................................................
Recommended publications
  • 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]
  • SOX12 Contributes to the Activation of the JAK2/STAT3 Pathway and Malignant Transformation of Esophageal Squamous Cell Carcinoma
    ONCOLOGY REPORTS 45: 129-138, 2021 SOX12 contributes to the activation of the JAK2/STAT3 pathway and malignant transformation of esophageal squamous cell carcinoma CHUNGUANG LI1*, MAOLING ZHU2*, JI ZHU1, QIJUE LU1, BOWEN SHI1, BIN SUN3 and HEZHONG CHEN1 1Department of Thoracic Surgery, Changhai Hospital, Second Military Medical University, Shanghai 200438; 2Department of Gastroenterology, Yangpu Hospital, Tongji University School of Medicine, Shanghai 200090; 3Molecular Oncology Laboratory, Eastern Hepatobiliary Surgery Hospital, Second Military Medical University, Shanghai 200438, P.R. China Received May 15, 2020; Accepted October 6, 2020 DOI: 10.3892/or.2020.7863 Abstract. As a crucial transcription factor, sex-determining cells in vitro. Recombinant protein of SOX12 could restore the region Y box 12 (SOX12) is closely related with tumorigenesis aggressive phenotype of ESCC cells. Furthermore, knockdown and malignant transformation in various malignant tumor of the expression of SOX12 inhibited the activation of the Janus types. To date, the specific function of SOX12 in esophageal kinase 2 (JAK2)/signal transducer and activator of transcrip- squamous cell carcinoma (ESCC) has remained largely tion 3 (STAT3) signaling pathway by decreasing the expression elusive and requires further investigation. The present study of the JAK2/STAT3 signaling pathway. Recombinant protein aimed to determine whether aberrant expression of SOX12 is of SOX12 could recover the activation of the JAK2/STAT3 associated with malignant development of ESCC. The expres- signaling pathway. Analysis of the clinical data revealed that sion level of SOX12 in ESCC cells and tissues was analyzed overexpression of SOX12 indicated shorter overall survival by RT-qPCR and western blotting. Short hairpin RNA time (OS; P=0.0341) and disease-free survival time (DFS; (shRNA) targeting SOX12 was transfected into ESCC cells to P=0.04).
    [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]
  • Supplemental Information
    Supplemental information Dissection of the genomic structure of the miR-183/96/182 gene. Previously, we showed that the miR-183/96/182 cluster is an intergenic miRNA cluster, located in a ~60-kb interval between the genes encoding nuclear respiratory factor-1 (Nrf1) and ubiquitin-conjugating enzyme E2H (Ube2h) on mouse chr6qA3.3 (1). To start to uncover the genomic structure of the miR- 183/96/182 gene, we first studied genomic features around miR-183/96/182 in the UCSC genome browser (http://genome.UCSC.edu/), and identified two CpG islands 3.4-6.5 kb 5’ of pre-miR-183, the most 5’ miRNA of the cluster (Fig. 1A; Fig. S1 and Seq. S1). A cDNA clone, AK044220, located at 3.2-4.6 kb 5’ to pre-miR-183, encompasses the second CpG island (Fig. 1A; Fig. S1). We hypothesized that this cDNA clone was derived from 5’ exon(s) of the primary transcript of the miR-183/96/182 gene, as CpG islands are often associated with promoters (2). Supporting this hypothesis, multiple expressed sequences detected by gene-trap clones, including clone D016D06 (3, 4), were co-localized with the cDNA clone AK044220 (Fig. 1A; Fig. S1). Clone D016D06, deposited by the German GeneTrap Consortium (GGTC) (http://tikus.gsf.de) (3, 4), was derived from insertion of a retroviral construct, rFlpROSAβgeo in 129S2 ES cells (Fig. 1A and C). The rFlpROSAβgeo construct carries a promoterless reporter gene, the β−geo cassette - an in-frame fusion of the β-galactosidase and neomycin resistance (Neor) gene (5), with a splicing acceptor (SA) immediately upstream, and a polyA signal downstream of the β−geo cassette (Fig.
    [Show full text]
  • SUPPLEMENTAL FIGURE LEGENDS Supplemental Figure S1. RBPJ
    Xie et al. SUPPLEMENTAL FIGURE LEGENDS Supplemental Figure S1. RBPJ correlates with BTIC marker expression. A-D. The TCGA GBM dataset was downloaded and correlations analyzed by R. RBPJ mRNA levels were highly correlated with (A) Olig2, (B) Sox2, (C) CD133, and (D) Sox4 levels. E. RBPJ is preferentially expressed in proneural glioblastomas. The glioblastoma TCGA dataset was interrogated for RBPJ mRNA expression segregated by transcriptional profile. The proneural tumors were further divided into G-CIMP (glioma CpG-island methylator phenotype) or non-G-CIMP. **, p < 0.01. ****, p < 0.0001. *****, p < 0.00001. Supplemental Figure S2. Targeting RBPJ induces BTIC apoptosis. A. 3691 BTICs were transduced with shCONT, shRBPJ-1, or shRBPJ-2. Lysates were prepared and immunoblotted with the indicated antibodies. shRNA-mediated knockdown of RBPJ was associated with increased cleaved (activated) PARP. B. 3691 BTICs were transduced with shCONT, shRBPJ-1, or shRBPJ-2. Apoptosis measured by Annexin V staining. Data are presented as mean ± SEM (two- way ANOVA; **, p < 0.01; n = 3). Supplemental Figure S3. Targeting RBPJ does not affect non-BTIC proliferation. Non-BTICs (Top, 3691; Bottom, 4121) were transduced with shCONT, shRBPJ-1, or shRBPJ-2. Cell proliferation was measured by CellTiter-Glo. 42 Xie et al. Supplemental Figure S4. RBPJ induces transcriptional profiles in BTICs distinct from Notch activation. A. In parallel experiments, 3691 BTICs were either treated with DAPT (at either 5 μM or 10 μM) vs. vehicle control (DMSO) or transduced with shRBPJ vs. shCONT. RNA-Seq was performed and the results displayed as a heat map with normalization to the relevant control.
    [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]
  • SOX4 Is Activated by C-MYC in Prostate Cancer
    Medical Oncology (2019) 36:92 https://doi.org/10.1007/s12032-019-1317-6 ORIGINAL PAPER SOX4 is activated by C‑MYC in prostate cancer Hongyan Dong1,2 · Jing Hu1 · Lin Wang3 · Mei Qi1 · Ning Lu1 · Xiao Tan2 · Muyi Yang1 · Xinnuo Bai4 · Xuemei Zhan2 · Bo Han1,5 Received: 26 July 2019 / Accepted: 12 September 2019 / Published online: 27 September 2019 © Springer Science+Business Media, LLC, part of Springer Nature 2019 Abstract Although MYC proto-oncogene (C-MYC) amplifcation has been consistently reported to be a potential marker for prostate cancer (PCa) progression and prognosis, the clinicopathological and prognostic signifcance of C-MYC protein expression remains controversial. Overexpression of SOX4 has been shown to play important roles in multiple cancers including PCa. However, the link between these two critical genetic aberrations is unclear. In the current study, we showed that C-MYC was overexpressed in 16.2% (17/105) of Chinese patients with localized PCa. Overexpression of C-MYC was signifcantly associated with high Gleason scores (P = 0.012) and high Ki67 labeling index (P = 0.005). C-MYC overexpression was cor- related with cancer-related mortality and suggested to be an unfavorable prognostic factor in Chinese PCa patients (P = 0.018). Overexpression of C-MYC is associated with SOX4 overexpression in PCa tissues. Notably, SOX4 is a direct target gene of C-MYC; C-MYC activates SOX4 expression via binding to its promoter. In addition, Co-IP analysis demonstrated a physi- cal interaction between C-MYC and SOX4 protein in PCa cells. Clinically, C-MYC+/SOX4+ characterized poor prognosis in a subset of PCa patients.
    [Show full text]
  • Transcription Factor Gene Expression Profiling and Analysis of SOX Gene Family Transcription Factors in Human Limbal Epithelial
    Transcription factor gene expression profiling and analysis of SOX gene family transcription factors in human limbal epithelial progenitor cells Der Naturwissenschaftlichen Fakultät der Friedrich-Alexander-Universität Erlangen-Nürnberg zur Erlangung des Doktorgrades Dr. rer. nat. vorgelegt von Dr. med. Johannes Menzel-Severing aus Bonn Als Dissertation genehmigt von der Naturwissenschaftlichen Fakultät der Friedrich-Alexander-Universität Erlangen-Nürnberg Tag der mündlichen Prüfung: 7. Februar 2018 Vorsitzender des Promotionsorgans: Prof. Dr. Georg Kreimer Gutachter: Prof. Dr. Andreas Feigenspan Prof. Dr. Ursula Schlötzer-Schrehardt 1 INDEX 1. ABSTRACTS Page 1.1. Abstract in English 4 1.2. Zusammenfassung auf Deutsch 7 2. INTRODUCTION 2.1. Anatomy and histology of the cornea and the corneal surface 11 2.2. Homeostasis of corneal epithelium and the limbal stem cell paradigm 13 2.3. The limbal stem cell niche 15 2.4. Cell therapeutic strategies in ocular surface disease 17 2.5. Alternative cell sources for transplantation to the corneal surface 18 2.6. Transcription factors in cell differentiation and reprogramming 21 2.7. Transcription factors in limbal epithelial cells 22 2.8. Research question 25 3. MATERIALS AND METHODS 3.1. Human donor corneas 27 3.2. Laser Capture Microdissection (LCM) 28 3.3. RNA amplification and RT2 profiler PCR arrays 29 3.4. Real-time PCR analysis 33 3.5. Immunohistochemistry 34 3.6. Limbal epithelial cell culture 38 3.7. Transcription-factor knockdown/overexpression in vitro 39 3.8. Proliferation assay 40 3.9. Western blot 40 3.10. Statistical analysis 41 2 4. RESULTS 4.1. Quality control of LCM-isolated and amplified RNA 42 4.2.
    [Show full text]
  • Directing an Artificial Zinc Finger Protein to New Targets by Fusion to a Non-DNA Binding Domain
    Directing an artificial zinc finger protein to new targets by fusion to a non-DNA binding domain Wooi Fang (Catheryn) Lim A thesis in fulfilment of the requirements for the degree of Doctor of Philosophy School of Biotechnology and Biomolecular Sciences Faculty of Science March 2016 Page | 0 THESIS/ DISSERTATION SHEET Page | i ORIGINALITY STATEMENT ‘I hereby declare that this submission is my own work and to the best of my knowledge it contains no materials previously published or written by another person, or substantial proportions of material which have been accepted for the award of any other degree or diploma at UNSW or any other educational institution, except where due acknowledgement is made in the thesis. Any contribution made to the research by others, with whom I have worked at UNSW or elsewhere, is explicitly acknowledged in the thesis. I also declare that the intellectual content of this thesis is the product of my own work, except to the extent that assistance from others in the project's design and conception or in style, presentation and linguistic expression is acknowledged.’ WOOI FANG LIM Signed …………………………………………….............. 31-03-2016 Date …………………………………………….............. Page | i COPYRIGHT STATEMENT ‘I hereby grant the University of New South Wales or its agents the right to archive and to make available my thesis or dissertation in whole or part in the University libraries in all forms of media, now or here after known, subject to the provisions of the Copyright Act 1968. I retain all proprietary rights, such as patent rights. I also retain the right to use in future works (such as articles or books) all or part of this thesis or dissertation.
    [Show full text]
  • SOX4 Expression in Bladder Carcinoma: Clinical Aspects and in Vitro Functional Characterization
    Research Article SOX4 Expression in Bladder Carcinoma: Clinical Aspects and In vitro Functional Characterization Mads Aaboe,1 Karin Birkenkamp-Demtroder,1 Carsten Wiuf,1,3 Flemming Brandt Sørensen,4 Thomas Thykjaer,1 Guido Sauter,5 Klaus Møller-Ernst Jensen,2 Lars Dyrskjøt,1 and Torben Ørntoft1 1Molecular Diagnostic Laboratory, Department of Clinical Biochemistry, and 2Department of Urology, Aarhus University Hospital/Skejby Sygehus, Aarhus N, Denmark; 3Bioinformatics Research Center, University of Aarhus; 4Institute of Pathology, Aarhus University Hospital, Aarhus Sygehus, Aarhus C, Denmark; and 5Department of Pathology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany Abstract shown to be involved in a range of developmental processes. The SOX4À/À phenotype in mouse results in death during embryo- The human transcription factor SOX4 was 5-fold up-regulated in bladder tumors compared with normal tissue based on genesis due to lack of normal heart development, which results in whole-genome expression profiling of 166 clinical bladder circulatory failure (6, 7). Furthermore, other studies have shown tumor samples and 27 normal urothelium samples.Using a that SOX4 is expressed in hormone-responsive tissues of the SOX4-specific antibody, we found that the cancer cells reproductive system in mouse, as well as in brain and T- and B-cell expressed the SOX4 protein and, thus, did an evaluation of lymphocytes (8, 9), and may play a role in osteoblastic differenti- SOX4 protein expression in 2,360 bladder tumors using a ation (10). tissue microarray with clinical annotation.We found a SOX4 binds to the 7-bp DNA-motif AACAAAG through its high mobility group domain and thereby transcriptionally activates correlation (P < 0.05) between strong SOX4 expression and target genes (9, 11).
    [Show full text]
  • SOX2 and SOX12 Are Predictive of Prognosis in Patients with Clear Cell Renal Cell Carcinoma
    4564 ONCOLOGY LETTERS 15: 4564-4570, 2018 SOX2 and SOX12 are predictive of prognosis in patients with clear cell renal cell carcinoma WEIJIE GU1,2*, BEIHE WANG1,2*, FANGNING WAN1,2*, JUNLONG WU1,2, XIAOLIN LU1,2, HONGKAI WANG1,2, YAO ZHU1,2, HAILIANG ZHANG1,2, GUOHAI SHI1,2, BO DAI1,2 and DINGWEI YE1,2 1Department of Urology, Fudan University Shanghai Cancer Center; 2Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, P.R. China Received October 21, 2016; Accepted September 28, 2017 DOI: 10.3892/ol.2018.7828 Abstract. Sex-determining region Y-box protein (SOX) genes were associated with poor prognosis for OS (log-rank test, all serve an important role in cancer growth and metastasis. P<0.05). SOX2 and SOX12 were identified as independent The present study aimed to determine the predictive ability prognostic factors of OS in clear cell RCC. of SOX and associated genes identified through molecular network in clear cell renal cell carcinoma (RCC). A total of Introduction 505 patients with clear cell RCC from The Cancer Genome Atlas (TCGA) cohorts were collected in this study. The Renal cell carcinoma (RCC) accounts for ~2-3% of all malig- expression profile of SOX and associated genes were obtained nancies worldwide (1). Despite an increasing proportion of from the TCGA RNAseq database. Clinicopathological patients with early stage tumors at diagnosis and the develop- characteristics, including age, gender, tumor grade, stage, ment of novel treatment strategies, a quarter still present with laterality disease-free-survival and overall survival (OS) were locally advanced or metastatic disease, and eventually, one collected.
    [Show full text]