Long Non‑Coding RNA Plasmacytoma Variant Translocation 1 Gene Promotes the Development of Cervical Cancer Via the NF‑Κb Pathway

Total Page:16

File Type:pdf, Size:1020Kb

Long Non‑Coding RNA Plasmacytoma Variant Translocation 1 Gene Promotes the Development of Cervical Cancer Via the NF‑Κb Pathway MOLECULAR MEDICINE REPORTS 20: 2433-2440, 2019 Long non‑coding RNA plasmacytoma variant translocation 1 gene promotes the development of cervical cancer via the NF‑κB pathway CHANG WANG1, HAO ZOU2, HONGJUAN YANG1, LEI WANG1, HUIJUN CHU1, JINWEN JIAO1, YANKUI WANG1 and AIPING CHEN1 1Department of Gynecology, The Affiliated Hospital of Qingdao University, Qingdao, Shandong 266590; 2Department of Hepatobiliary and Pancreatic Surgery, The Affiliated Hospital of Qingdao University, Qingdao, Shandong 266003, P.R. China Received November 16, 2018; Accepted May 20, 2019 DOI: 10.3892/mmr.2019.10479 Abstract. The long noncoding RNA plasmacytoma variant of cervical cancer cells, potentially via the NF‑κB pathway. translocation 1 gene (LncRNA PVT1) has an important role in The data from the present study will contribute to the current tumor occurrence and development, yet the role and underlying knowledge surrounding the theoretical basis of cervical cancer molecular mechanisms of this RNA in cervical cancer have and provide a new perspective for the treatment of cervical not yet been elucidated. In the present study, three cervical cancer. cancer cell lines (HeLa, Ca Ski and SiHa) were used to verify how LncRNA PVT1 mediates cervical cancer development, Introduction and the H8 cell line was used as a control. A LncRNA PVT1 overexpression vector or small interfering RNAs targeting There were ~500,000 people with cervical cancer worldwide LncRNA PVT1 were transfected into cervical cancer cells to in 2012, and in women, cervical cancer ranks second in cancer generate LncRNA PVT1 overexpression and silencing in these occurrence and 15th in cancer mortality (1-3). Currently, the cells. LncRNA PVT1 overexpression accelerated the growth of pathogenesis of cervical cancer is not fully clear, but human cervical cancer cells by advancing the cell cycle and inhibiting papillomavirus (HPV) infection is established to be the most cellular apoptosis; increases in Cyclin D1 (CCND1) mRNA closely associated risk factor for cervical cancer occurrence. and activated Bcl-2 protein expression levels also supported However, high-risk HPV infection alone is not enough to this finding. Furthermore, NF‑κB activation and expression induce tumor progression (4). was increased by LncRNA PVT1 overexpression. In addition, Previous studies have revealed that HPV activates the NF‑κB activation or inhibition induced changes in cell viability, NF‑κB pathway to promote expression of different anti-apop- accompanied by changes in CCND1 and Bcl-2 expression. tosis genes that induce cervical cancer development through Increases or decreases in microRNA‑16 (miR‑16) expres- inhibition of cellular apoptosis (5‑8). NF‑κB an essential sion (using miR mimics and inhibitors) also corresponded nuclear transcription factor composed of RelA/p65, p50, p52 to changes in LncRNA PVT1 expression, in vitro. miR‑16 and NF‑κB family members (9). NF‑κB pathway activation is mimics and inhibitor had opposite effects to those of NF‑κB associated with the development of various tumors and can activity, and miR‑16 was demonstrated to directly interact with promote tumor cell growth, proliferation and anti-apoptotic the NF‑κB gene as measured using the dual-luciferase assay. effects to promote malignant transformation and tumor cell In summary, LncRNA PVT1 inhibits the effect of miR‑16, metastases (10,11). NF‑κB pathway activation is a common promoting the cell cycle and inhibiting cellular apoptosis mechanism used to promote tumor survival; the activated NF‑κB induces expression of various anti-apoptotic genes, such as the cellular inhibitor of apoptosis protein (12), the X-linked inhibitor of apoptosis protein (13) and Bcl-2 family proteins (14). In addition, NF‑κB activation was shown to be Correspondence to: Professor Yankui Wang, Department of present in cervical cancer (15), suggesting that NF‑κB may Gynecology, The Affiliated Hospital of Qingdao University, 16 Jiangsu Road, Shinan, Qingdao, Shandong 266590, P.R. China have a role in the development and progression of cervical E-mail: [email protected] cancer. MicroRNAs (miRs) serve as markers of cervical cancer Key words: cervical cancer cell, long non-coding RNA, plasmacytoma progression (16), and it was previously reported that miR‑16 has variant translocation 1 gene, microRNA‑16, NF‑κB, cell proliferation an important role in the regulation of cell apoptosis, proliferation, and apoptosis migration and invasion of cervical cancer (17-19). In addition, miR‑16 targets inhibitor of nuclear factor κB kinase subunit β (Iκ-Bα) in hepatocellular carcinoma (20), and decreases NF‑κB 2434 WANG et al: REGULATION OF CERVICAL CANCER gene expression; thus, reducing the levels of activated NF‑κB (21). gene were obtained via artificial nucleotide synthesis at Beijing Therefore, miR‑16 has the important role of regulating the Tianyi Huiyuan Bioscience & Technology Inc. and the expres- NF‑κB signaling pathway. On the other hand, long non-coding sion plasmid constructed. The siRNA (5'-GAG​CUG​CGA​GCA​ RNAs (lncRNAs) are involved in different tumor processes (22) AAG​AUG​U‑3') and control siRNA (si‑NC; 5'-GAU​CGU​ACU​ through regulation of target gene expression at the transcriptional, AUA​GCU​UGU​A‑3') of LncRNA PVT1 were purchased from post-transcriptional and epigenetic levels (23-25). LncRNAs Guangzhou RiboBio Co., Ltd. also at serve as precursors of small interfering RNAs (siRNAs), miRs and piwi-interacting RNAs (26,27), and can compete with Cell transfection. Fugene6 transfection reagent (Roche miR as competing endogenous RNAs (ceRNAs) to achieve Diagnostics) was used according to the manufacturer's inter‑communication and regulation (28‑33). Furthermore, the protocol. The overexpression vector (final concentration lncRNA‑plasmacytoma variant translocation 1 (LncRNA PVT1) 1 µg/ml) or siRNA of LncRNA PVT1 (final concentration gene was reported to be a biomarker of tumorigenesis and 50 nm) were transfected into HeLa, Ca Ski and SiHa cells associated with the development of cervical cancer (34-37) by in 96-well plates or 100 mm dishes with 1x105 cells/ml and competitively inhibiting miRs (38,39). cultured for 48 or 60 h, respectively, and the pEGFP‑N3 empty Although no evidence currently exists to support the vector or si‑NC were also transfected with the corresponding hypothesis that the interaction between LncRNA PVT1 and internal controls. Additionally, HeLa cells were respectively miR‑16 induces cervical cancer development, it is speculated treated with 5 µg/ml lipopolysaccharides (LPS; Beyotime that the LncRNA PVT1 gene promotes cervical cancer devel- Institute of Biotechnology) plus the lncRNA PVT1 siRNA, opment by inhibiting miR expression and activating the NF‑κB with pyrrolidinedithiocarbamate (PDTC; 50 µmol/l; Beyotime pathway. In the present study, three known cervical cancer Institute of Biotechnology) alone, or with PDTC (50 µmol/l) cell lines (HeLa, Ca Ski and SiHa cells) and the H8 cell line on the basis of the lncRNA PVT1 overexpression treatment for (as an external control of three cervical cancer cell lines) were 8 h to activate or inhibit the NF‑κB pathway, and then the cells selected as the experimental models for in vitro experiments to were continuously cultured for 60 h. In addition, using the investigate the effect and molecular mechanisms of LncRNA Fugene6 transfection reagent, HeLa cells with 60% confluency PVT1 on cervical cancer development. were transiently transfected with the miR‑16 mimics (final concentration 100 nm) or miR‑16 inhibitor (final concentration Materials and methods 50 nm). All cells were collected for the experiments below following 60 h treatment. Cell culture. As it involved the use of human materials, the present study was approved by the Ethics Committee of the MTT assay. Following culture of the cells from the four cell Affiliated Hospital of Qingdao University (Qingdao, China) lines in 96-well plates with different treatments or no treatment according to the experimental requirements of the school. for 60 h, cell numbers were determined using the MTT assay. HeLa, Ca Ski, SiHa and H8 cells were purchased from the The assay was conducted by adding 20 µl MTT (5 mg/ml) to National Infrastructure of Cell Line Resources of China. H8 each well of the plates and incubating the plates for 4 h at 37˚C. cell line (the human cervical epithelial immortalized cell Following removal of the supernatant, formazan crystals were line) was used as the external control of the three cervical dissolved in 200 µl dimethyl sulfoxide, and the absorbance cancer cell lines, HeLa, Ca Ski and SiHa. Each cell line was was measured at 490 nm by microplate reader (Thermo Fisher cultured in DMEM (Gibco; Thermo Fisher Scientific, Inc.) Scientific, Inc.). There were eight replicate wells for each group containing 10% FBS (Gibco; Thermo Fisher Scientific, Inc.) in to ensure the accuracy of the experiments. 100 mm culture dishes (Corning Inc.) and incubated in a 5% CO2. incubator at 37˚C. The media was changed every 3 days, Flow cytometry (FCM). Following treatment with or without and the cells were passaged once they reached 80% conflu- the LncRNA‑PVT1 siRNA for 48 h, HeLa cells were ence. Additionally, 293T cells (purchased from the National collected to use for cell cycle or cell apoptosis analyses using Infrastructure of Cell Line Resources of China) were used in a FACSCanto II flow cytometer (Becton, Dickinson and co-transfection experiment and cultured with the same afore- Company). For the cell cycle analysis, cells were centrifuged mentioned treatment. To ensure cell activity, the cells from at 300 x g for 5 min at 4˚C and washed with ice‑cold PBS passage 2-4 were used for further experiments. following digestion with trypsin. The cells were then mixed with 0.25% Triton X-100 and 5 µl propidium iodide (PI) and Construction of LncRNA PVT1 and 3'‑untranslated region incubated for 30 min at room temperature in the dark. Cells (3'UTR) of NF‑κB overexpression vectors and siRNA of LncRNA were resuspended in 0.5 ml PBS and analyzed immediately.
Recommended publications
  • Investigating Cone Photoreceptor Development Using Patient-Derived NRL Null Retinal Organoids
    ARTICLE https://doi.org/10.1038/s42003-020-0808-5 OPEN Investigating cone photoreceptor development using patient-derived NRL null retinal organoids Alyssa Kallman1,11, Elizabeth E. Capowski 2,11, Jie Wang 3, Aniruddha M. Kaushik4, Alex D. Jansen2, Kimberly L. Edwards2, Liben Chen4, Cynthia A. Berlinicke3, M. Joseph Phillips2,5, Eric A. Pierce6, Jiang Qian3, ✉ ✉ Tza-Huei Wang4,7, David M. Gamm2,5,8 & Donald J. Zack 1,3,9,10 1234567890():,; Photoreceptor loss is a leading cause of blindness, but mechanisms underlying photoreceptor degeneration are not well understood. Treatment strategies would benefit from improved understanding of gene-expression patterns directing photoreceptor development, as many genes are implicated in both development and degeneration. Neural retina leucine zipper (NRL) is critical for rod photoreceptor genesis and degeneration, with NRL mutations known to cause enhanced S-cone syndrome and retinitis pigmentosa. While murine Nrl loss has been characterized, studies of human NRL can identify important insights for human retinal development and disease. We utilized iPSC organoid models of retinal development to molecularly define developmental alterations in a human model of NRL loss. Consistent with the function of NRL in rod fate specification, human retinal organoids lacking NRL develop S- opsin dominant photoreceptor populations. We report generation of two distinct S-opsin expressing populations in NRL null retinal organoids and identify MEF2C as a candidate regulator of cone development. 1 Institute of Genetic Medicine, Johns Hopkins University School of Medicine, Baltimore, USA. 2 Waisman Center, University of Wisconsin-Madison, Madison, USA. 3 Department of Ophthalmology, Wilmer Eye Institute, Johns Hopkins University School of Medicine, Baltimore, USA.
    [Show full text]
  • Supplemental Materials ZNF281 Enhances Cardiac Reprogramming
    Supplemental Materials ZNF281 enhances cardiac reprogramming by modulating cardiac and inflammatory gene expression Huanyu Zhou, Maria Gabriela Morales, Hisayuki Hashimoto, Matthew E. Dickson, Kunhua Song, Wenduo Ye, Min S. Kim, Hanspeter Niederstrasser, Zhaoning Wang, Beibei Chen, Bruce A. Posner, Rhonda Bassel-Duby and Eric N. Olson Supplemental Table 1; related to Figure 1. Supplemental Table 2; related to Figure 1. Supplemental Table 3; related to the “quantitative mRNA measurement” in Materials and Methods section. Supplemental Table 4; related to the “ChIP-seq, gene ontology and pathway analysis” and “RNA-seq” and gene ontology analysis” in Materials and Methods section. Supplemental Figure S1; related to Figure 1. Supplemental Figure S2; related to Figure 2. Supplemental Figure S3; related to Figure 3. Supplemental Figure S4; related to Figure 4. Supplemental Figure S5; related to Figure 6. Supplemental Table S1. Genes included in human retroviral ORF cDNA library. Gene Gene Gene Gene Gene Gene Gene Gene Symbol Symbol Symbol Symbol Symbol Symbol Symbol Symbol AATF BMP8A CEBPE CTNNB1 ESR2 GDF3 HOXA5 IL17D ADIPOQ BRPF1 CEBPG CUX1 ESRRA GDF6 HOXA6 IL17F ADNP BRPF3 CERS1 CX3CL1 ETS1 GIN1 HOXA7 IL18 AEBP1 BUD31 CERS2 CXCL10 ETS2 GLIS3 HOXB1 IL19 AFF4 C17ORF77 CERS4 CXCL11 ETV3 GMEB1 HOXB13 IL1A AHR C1QTNF4 CFL2 CXCL12 ETV7 GPBP1 HOXB5 IL1B AIMP1 C21ORF66 CHIA CXCL13 FAM3B GPER HOXB6 IL1F3 ALS2CR8 CBFA2T2 CIR1 CXCL14 FAM3D GPI HOXB7 IL1F5 ALX1 CBFA2T3 CITED1 CXCL16 FASLG GREM1 HOXB9 IL1F6 ARGFX CBFB CITED2 CXCL3 FBLN1 GREM2 HOXC4 IL1F7
    [Show full text]
  • Perkinelmer Genomics to Request the Saliva Swab Collection Kit for Patients That Cannot Provide a Blood Sample As Whole Blood Is the Preferred Sample
    Microphthalmia/ Anophthalmia/ Coloboma Spectrum Panel Test Code D4300 Test Summary This test analyzes 48 genes that have been associated with microphthalmia, anophthalmia, coloboma and/or disorders associated with these ocular findings Turn-Around-Time (TAT)* 3 - 5 weeks Acceptable Sample Types Whole Blood (EDTA) (Preferred sample type) DNA, Isolated Dried Blood Spots Saliva Acceptable Billing Types Self (patient) Payment Institutional Billing Commercial Insurance Indications for Testing Individual born with small eyes, missing a piece of their eye or missing their entire eye Individual suspected of having a syndrome associated with microphtalmia, anophthalmia, and/or colobomas Family planning purposes for individuals with a personal and/or family history of microphthalmia, anophthalmia, and/or coloboma Test Description This panel analyzes 48 genes that have been associated with microphthalmia, anophthalmia, coloboma and/or disorders associated with these ocular findings. Both sequencing and deletion/duplication (CNV) analysis will be performed on the coding regions of all genes included (unless otherwise marked). All analysis is performed utilizing Next Generation Sequencing (NGS) technology. CNV analysis is designed to detect the majority of deletions and duplications of three exons or greater in size. Smaller CNV events may also be detected and reported, but additional follow-up testing is recommended if a smaller CNV is suspected. All variants are classified according to ACMG guidelines. Condition Description Microphthalmia is an eye disease where an individual is born with one or both eyes being abnormally small. Anophthalmia is an eye disease where an individual is born without one or both eyes. A coloboma is an eye disease where an individual is born with a piece of their eye missing.
    [Show full text]
  • Congenital Eye Disorders Gene Panel
    Congenital eye disorders gene panel Contact details Introduction Regional Genetics Service Ocular conditions are highly heterogeneous and show considerable phenotypic overlap. 1 in Levels 4-6, Barclay House 2,500 children in the UK are diagnosed as blind or severely visually impaired by the time they 37 Queen Square reach one year old. As many as half of these cases are likely to be inherited and remain undiagnosed due to the vast number of genes involved in these conditions. Many congenital London, WC1N 3BH eye disorders causing visual impairment or blindness at birth or progressive visual impairment T +44 (0) 20 7762 6888 also include syndromic conditions involving additional metabolic, developmental, physical or F +44 (0) 20 7813 8578 sensory abnormalities. Gene panels offer the enhanced probability of diagnosis as a very large number of genes can be interrogated. Samples required Ocular birth defects include all inheritance modalities. Autosomal dominant and recessive 5ml venous blood in plastic EDTA diseases as well as X-linked dominant and recessive diseases are seen. These conditions can bottles (>1ml from neonates) also be caused by de novo variants. Prenatal testing must be arranged Referrals in advance, through a Clinical Genetics department if possible. Patients presenting with a phenotype appropriate for the requested sub-panel Amniotic fluid or CV samples Referrals will be accepted from clinical geneticists and consultants in ophthalmology. should be sent to Cytogenetics for Prenatal testing dissecting and culturing, with instructions to forward the sample Prenatal diagnosis may be offered as appropriate where pathogenic variants have been to the Regional Molecular Genetics identified in accordance with expected inheritance pattern and where appropriate parental laboratory for analysis testing and counselling has been conducted.
    [Show full text]
  • Molecular Correlations Between Breast Cancer and Diabetes Mellitus Type 2 Examined by in Silico Analyses and Drug Inhibitory Effects
    bioRxiv preprint doi: https://doi.org/10.1101/568634; this version posted March 5, 2019. The copyright holder for this preprint (which was not certified by peer review) is the author/funder. All rights reserved. No reuse allowed without permission. 1 Molecular Correlations between Breast Cancer and Diabetes Mellitus Type 2 Examined by In Silico analyses and Drug Inhibitory Effects Farzaneh Afzali1, Zahra Nayeri1, Zarrin Minuchehr2, Mossa Gardaneh1* 1Department of Medical Biotechnology, 2Department of Systems Biotechnology, National Institute of Genetic Engineering and Biotechnology (NIGEB), Tehran, Iran. *Corresponding author: Mossa Gardaneh National Institute of Genetic Engineering and Biotechnology, Pazhoohesh Blvd, Tehran-Karaj HWY Kilometer 15, PO BOX 14965/161, Tehran-Iran, Tel: (+98) 21 44787 344 Fax: (+98 21) 4458-0395 Email: [email protected], [email protected] bioRxiv preprint doi: https://doi.org/10.1101/568634; this version posted March 5, 2019. The copyright holder for this preprint (which was not certified by peer review) is the author/funder. All rights reserved. No reuse allowed without permission. 2 ABSTRACT Background: Nearly 16% of people with breast cancer (BC) have Diabetes Mellitus type 2 (DM2) and are at a higher risk of death worldwide. However, their common regulatory factors and functional mechanisms have not been profoundly understood. In this study, we used in silico approaches to study their co-regulated differentially expressed genes (DEGs) in order to explore and introduce novel therapeutic targets. Results: Microarray analysis identified 196 co-regulated DEGs (156 upregulated and 40 downregulated) between BC and DM2. The most significant pathway was predicted to be Fc gamma R-mediated phagocytosis through pathway analysis.
    [Show full text]
  • Spatial and Temporal Specification of Neural Fates by Transcription Factor Codes François Guillemot
    REVIEW 3771 Development 134, 3771-3780 (2007) doi:10.1242/dev.006379 Spatial and temporal specification of neural fates by transcription factor codes François Guillemot The vertebrate central nervous system contains a great diversity Box 1. Neurons and glial cells of neurons and glial cells, which are generated in the embryonic neural tube at specific times and positions. Several classes of transcription factors have been shown to control various steps in the differentiation of progenitor cells in the neural tube and to determine the identity of the cells produced. Recent evidence indicates that combinations of transcription factors of the homeodomain and basic helix-loop-helix families establish molecular codes that determine both where and when the different kinds of neurons and glial cells are generated. Introduction Neuron Oligodendrocyte Astrocyte A multitude of neurons of different types, as well as oligodendrocytes and astrocytes (see Box 1), are generated as the vertebrate central The vertebrate central nervous system comprises three primary cell nervous system develops. These different neural cells are generated types, including neurons and two types of glial cells. Neurons are at defined times and positions by multipotent progenitors located in electrically excitable cells that process and transmit information via the walls of the embryonic neural tube. Progenitors located in the the release of neurotransmitters at synapses. Different subtypes of ventral neural tube at spinal cord level first produce motor neurons, neurons can be distinguished by the morphology of their cell body which innervate skeletal muscles and later produce oligodendrocytes and dendritic tree, the type of cells they connect with via their axon, the type of neurotransmitter used, etc.
    [Show full text]
  • GATA Transcription Factors and Their Co-Regulators Guide the Development Of
    GATA transcription factors and their co-regulators guide the development of GABAergic and serotonergic neurons in the anterior brainstem Laura Tikker Molecular and Integrative Biosciences Research Programme Faculty of Biological and Environmental Sciences Doctoral Programme Integrative Life Science University of Helsinki ACADEMIC DISSERTATION Doctoral thesis, to be presented for public examination, with the permission of the Faculty of Biological and Environmental Sciences of the University of Helsinki, in Raisio Hall (LS B2) in Forest Sciences building, Latokartanonkaari 7, Helsinki, on the 3rd of April, 2020 at 12 noon. Supervisor Professor Juha Partanen University of Helsinki (Finland) Thesis Committee members Docent Mikko Airavaara University of Helsinki (Finland) Professor Timo Otonkoski University of Helsinki (Finland) Pre-examinators Docent Satu Kuure University of Helsinki (Finland) Research Scientist Siew-Lan Ang, PhD The Francis Crick Institute (United Kingdom) Opponent Research Scientist Johan Holmberg, PhD Karolinska Institutet (Sweden) Custos Professor Juha Partanen University of Helsinki (Finland) The Faculty of Biological and Environmental Sciences, University of Helsinki, uses the Urkund system for plagiarism recognition to examine all doctoral dissertations. ISBN: 978-951-51-5930-4 (paperback) ISBN: 978-951-51-5931-1 (PDF) ISSN: 2342-3161 (paperback) ISSN: 2342-317X (PDF) Printing house: Painosalama Oy Printing location: Turku, Finland Printed on: 03.2020 Cover artwork: Serotonergic neurons in adult dorsal raphe (mouse).
    [Show full text]
  • Transcriptional Code and Disease Map for Adult Retinal Cell Types
    RE so UR C E Transcriptional code and disease map for adult retinal cell types Sandra Siegert1,7, Erik Cabuy1,7, Brigitte Gross Scherf1, Hubertus Kohler1, Satchidananda Panda2, Yun-Zheng Le3,4, Hans Jörg Fehling5, Dimos Gaidatzis1,6, Michael B Stadler1,6 & Botond Roska1 Brain circuits are assembled from a large variety of morphologically and functionally diverse cell types. It is not known how the intermingled cell types of an individual adult brain region differ in their expressed genomes. Here we describe an atlas of cell type transcriptomes in one brain region, the mouse retina. We found that each adult cell type expressed a specific set of genes, including a unique set of transcription factors, forming a ‘barcode’ for cell identity. Cell type transcriptomes carried enough information to categorize cells into morphological classes and types. Several genes that were specifically expressed in particular retinal circuit elements, such as inhibitory neuron types, are associated with eye diseases. The resource described here allows gene expression to be compared across adult retinal cell types, experimenting with specific transcription factors to differentiate stem or somatic cells to retinal cell types, and predicting cellular targets of newly discovered disease-associated genes. The brain is composed of many neuronal cell types that are deter- across the retina with a mosaic-like distribution (Supplementary Text mined during development by a dynamic transcriptional program1–5. and Supplementary Fig. 1). The cellular architecture of the retina is In adults, neurons sampled from different brain areas such as the highly conserved among mammals14–17. cortex, cerebellum and hippocampus maintain differences in their By constructing and analyzing a transcriptome atlas for retinal cell expressed genomes6,7.
    [Show full text]
  • Regulation and Function of Ptf1a in the Developing Nervous System
    REGULATION AND FUNCTION OF PTF1A IN THE DEVELOPING NERVOUS SYSTEM APPROVED BY SUPERVISORY COMMITTEE Jane E. Johnson, Ph.D. Raymond J. MacDonald, Ph.D. Ondine Cleaver, Ph.D. Steven L. McKnight, Ph.D. DEDICATION Work of this nature never occurs in a vacuum and would be utterly impossible without the contributions of a great many. I would like to thank my fellow lab members and collaborators, both past and present, for all of their help over the past several years, my thesis committee for their guidance and encouragement, and of course Jane for giving me every opportunity to succeed. To my friends, family, and my dear wife Maria, your love and support have made all of this possible. REGULATION AND FUNCTION OF PTF1A IN THE DEVELOPING NERVOUS SYSTEM by DAVID MILES MEREDITH DISSERTATION Presented to the Faculty of the Graduate School of Biomedical Sciences The University of Texas Southwestern Medical Center at Dallas In Partial Fulfillment of the Requirements For the Degree of DOCTOR OF PHILOSOPHY The University of Texas Southwestern Medical Center at Dallas Dallas, Texas May, 2012 REGULATION AND FUNCTION OF PTF1A IN THE DEVELOPING NERVOUS SYSTEM DAVID MILES MEREDITH The University of Texas Southwestern Medical Center at Dallas, 2012 JANE E. JOHNSON, Ph.D. Basic helix-loop-helix transcription factors serve many roles in development, including regulation of neurogenesis. Many of these factors are activated in naïve neural progenitors and function to promote neuronal differentiation and cell-type specification. Ptf1a is a basic helix-loop-helix protein that is required for proper inhibitory neuron formation in several regions of the developing nervous system, including the spinal cord, cerebellum, retina, and hypothalamus.
    [Show full text]
  • A Massively Parallel Reporter Assay Reveals Context-Dependent Activity of Homeodomain Binding Sites in Vivo
    Downloaded from genome.cshlp.org on October 6, 2021 - Published by Cold Spring Harbor Laboratory Press A MASSIVELY PARALLEL REPORTER ASSAY REVEALS CONTEXT-DEPENDENT ACTIVITY OF HOMEODOMAIN BINDING SITES IN VIVO Andrew E. O. Hughes1, Connie A. Myers1, and Joseph C. Corbo1* 1Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, Missouri 63110, USA *To whom correspondence should be addressed. Tel: +1 314 362 6254; Fax: +1 314 362 4096; Email: [email protected] Running title: Context-dependent activity of CRX binding sites Key words: homeodomain, cis-regulatory element, massively parallel reporter assay, transcription factor binding site, retina 1 Downloaded from genome.cshlp.org on October 6, 2021 - Published by Cold Spring Harbor Laboratory Press ABSTRACT Cone-rod homeobox (CRX) is a paired-like homeodomain transcription factor (TF) and a master regulator of photoreceptor development in vertebrates. The in vitro DNA binding preferences of CRX have been described in detail, but the degree to which in vitro binding affinity is correlated with in vivo enhancer activity is not known. In addition, paired-class homeodomain TFs can bind DNA cooperatively as both homodimers and heterodimers at inverted TAAT half-sites separated by two or three nucleotides. This dimeric configuration is thought to mediate target specificity, but whether monomeric and dimeric sites encode distinct levels of activity is not known. Here, we used a massively parallel reporter assay to determine how local sequence context shapes the regulatory activity of CRX binding sites in mouse photoreceptors. We assayed inactivating mutations in >1,700 TF binding sites and found that dimeric CRX binding sites act as stronger enhancers than monomeric CRX binding sites.
    [Show full text]
  • Preservation of Cone Photoreceptors After a Rapid Yet Transient Degeneration and Remodeling in Cone-Only Nrlϫ/Ϫ Mouse Retina
    528 • The Journal of Neuroscience, January 11, 2012 • 32(2):528–541 Development/Plasticity/Repair Preservation of Cone Photoreceptors after a Rapid yet Transient Degeneration and Remodeling in Cone-Only NrlϪ/Ϫ Mouse Retina Jerome E. Roger,1 Keerthi Ranganath,1 Lian Zhao,2 Radu I. Cojocaru,1 Matthew Brooks,1 Norimoto Gotoh,1 Shobi Veleri,1 Avinash Hiriyanna,1 Rivka A. Rachel,1 Maria Mercedes Campos,3 Robert N. Fariss,3 Wai T. Wong,2 and Anand Swaroop1 1Neurobiology-Neurodegeneration and Repair Laboratory, 2Unit on Neuron–Glia Interactions, 3Biological Imaging Core, National Eye Institute, National Institutes of Health, Bethesda, Maryland 20892 Cone photoreceptors are the primary initiator of visual transduction in the human retina. Dysfunction or death of rod photoreceptors precedes cone loss in many retinal and macular degenerative diseases, suggesting a rod-dependent trophic support for cone survival. Rod differentiation and homeostasis are dependent on the basic motif leucine zipper transcription factor neural retina leucine zipper (NRL). The loss of Nrl (Nrl Ϫ/Ϫ) in mice results in a retina with predominantly S-opsin-containing cones that exhibit molecular and functional characteristics of wild-type cones. Here, we report that Nrl Ϫ/Ϫ retina undergoes a rapid but transient period of degeneration in early adulthood, with cone apoptosis, retinal detachment, alterations in retinal vessel structure, and activation and translocation of retinal microglia. However, cone degeneration stabilizes by 4 months of age, resulting in a thinner but intact outer nuclear layer with residual cones expressing S- and M-opsins and a preserved photopic electroretinogram. At this stage, microglia translocate back to the inner retina and reacquire a quiescent morphology.
    [Show full text]
  • BMC Biology Biomed Central
    BMC Biology BioMed Central Research article Open Access Classification and nomenclature of all human homeobox genes PeterWHHolland*†1, H Anne F Booth†1 and Elspeth A Bruford2 Address: 1Department of Zoology, University of Oxford, South Parks Road, Oxford, OX1 3PS, UK and 2HUGO Gene Nomenclature Committee, European Bioinformatics Institute (EMBL-EBI), Wellcome Trust Genome Campus, Hinxton, Cambridgeshire, CB10 1SA, UK Email: Peter WH Holland* - [email protected]; H Anne F Booth - [email protected]; Elspeth A Bruford - [email protected] * Corresponding author †Equal contributors Published: 26 October 2007 Received: 30 March 2007 Accepted: 26 October 2007 BMC Biology 2007, 5:47 doi:10.1186/1741-7007-5-47 This article is available from: http://www.biomedcentral.com/1741-7007/5/47 © 2007 Holland et al; licensee BioMed Central Ltd. This is an Open Access article distributed under the terms of the Creative Commons Attribution License (http://creativecommons.org/licenses/by/2.0), which permits unrestricted use, distribution, and reproduction in any medium, provided the original work is properly cited. Abstract Background: The homeobox genes are a large and diverse group of genes, many of which play important roles in the embryonic development of animals. Increasingly, homeobox genes are being compared between genomes in an attempt to understand the evolution of animal development. Despite their importance, the full diversity of human homeobox genes has not previously been described. Results: We have identified all homeobox genes and pseudogenes in the euchromatic regions of the human genome, finding many unannotated, incorrectly annotated, unnamed, misnamed or misclassified genes and pseudogenes.
    [Show full text]