Sleep Architecture in Mice Is Shaped by the Transcription Factor AP-2Β

Total Page:16

File Type:pdf, Size:1020Kb

Sleep Architecture in Mice Is Shaped by the Transcription Factor AP-2Β Genetics: Early Online, published on September 2, 2020 as 10.1534/genetics.120.303435 Title: Sleep architecture in mice is shaped by the transcription factor AP-2β Authors: Ayaka Nakai1,2, Tomoyuki Fujiyama1, Nanae Nagata1, Mitsuaki Kashiwagi1, Aya Ikkyu1, Marina Takagi1, Chika Tatsuzawa1, Kaeko Tanaka1, Miyo Kakizaki1, Mika Kanuka1, Taizo Kawano1, Seiya Mizuno3, Fumihiro Sugiyama3, Satoru Takahashi3, Hiromasa Funato1,4, Takeshi Sakurai1,5,6, Masashi Yanagisawa1,6,7,8, Yu Hayashi1,9* Affiliations: 1) International Institute for Integrative Sleep Medicine (WPI-IIIS), University of Tsukuba, Tsukuba, 305-8575, Japan 2) PhD Program in Neuroscience, Comprehensive Human Sciences, Graduate School of Comprehensive Human Sciences, University of Tsukuba, Tsukuba, 305-8575, Japan 3) Laboratory Animal Resource Center, University of Tsukuba, Tsukuba, 305-8575, Japan 4) Department of Anatomy, Faculty of Medicine, Toho University, Tokyo, 143-8540, Japan 5) Faculty of Medicine, University of Tsukuba, Tsukuba, 305-8575, Japan 6) Life Science Center for Survival Dynamics, Tsukuba Advanced Research Alliance (TARA), University of Tsukuba, Tsukuba, 305-8575, Japan 7) Department of Molecular Genetics, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA 8) R&D Center for Frontiers of MIRAI in Policy and Technology, University of Tsukuba, Tsukuba, 305-8575, Japan 9) Department of Human Health Sciences, Graduate School of Medicine, Kyoto University, Kyoto, 606-8507, Japan *Correspondence to: [email protected] 1 Copyright 2020. Abstract The molecular mechanism regulating sleep largely remains to be elucidated. In humans, families that carry mutations in TFAP2B, which encodes the transcription factor AP-2β, self-reported sleep abnormalities such as short-sleep and parasomnia. Notably, AP- 2 transcription factors play essential roles in sleep regulation in the nematode Caenorhabditis elegans and the fruit fly Drosophila melanogaster. Thus, AP-2 transcription factors might have a conserved role in sleep regulation across the animal phyla. However, direct evidence supporting the involvement of TFAP2B in mammalian sleep was lacking. In this study, by using the CRISPR/Cas9 technology, we generated two Tfap2b mutant mouse strains, Tfap2bK144 and Tfap2bK145, each harboring a single-nucleotide mutation within the introns of Tfap2b mimicking the mutations in two human kindreds that self-reported sleep abnormalities. The effects of these mutations were compared with those of a Tfap2b knockout allele (Tfap2b-). The protein expression level of TFAP2B in the embryonic brain was reduced to about half in Tfap2b+/- mice and was further reduced in Tfap2b-/- mice. By contrast, the protein expression level was normal in Tfap2bK145/+ mice but was reduced in Tfap2bK145/K145 mice to a similar extent as Tfap2b-/- mice. Tfap2bK144/+ and Tfap2bK144/K144 showed normal protein expression levels. Tfap2b+/- female mice showed increased wakefulness time and decreased non-rapid eye movement sleep (NREMS) time. By contrast, Tfap2bK145/+ female mice showed an apparently normal amount of sleep but instead exhibited fragmented NREMS, whereas Tfap2bK144/+ male mice showed reduced NREMS time specifically in the dark phase. Finally, in the adult brain, Tfap2b-LacZ expression was detected in the superior colliculus, locus coeruleus, cerebellum, and the nucleus of solitary tract. These findings provide direct evidence that TFAP2B influences NREMS amounts in mice and also show that different mutations in Tfap2b can lead to diverse effects on the sleep architecture. Introduction Sleep is a physiological state widely observed in animals. However, the molecular pathways involved in regulating sleep and to what the extent they are conserved among vertebrates and invertebrates remain largely unknown. In humans, natural short sleepers exhibit a reduced amount of time in sleep without reporting daytime fatigue (reviewed in (Shi et al. 2017)). In cases where the natural short sleep trait is hereditary, it is expected that the identification of the causal gene might lead to the understanding of the molecules that control sleepiness. In addition, recent studies using invertebrate animal models suggest that sleep-regulatory mechanisms are at least partly conserved at the molecular 2 level between vertebrates and invertebrates (reviewed in (Miyazaki et al. 2017)). For example, a gain-of-function mutation in the salt-inducible kinase 3 leads to a drastic increase in the amount of non-rapid eye movement sleep (NREMS) in mice, whereas its orthologue in the nematode Caenorhabditis elegans promotes developmentally-timed sleep and the orthologue in the fruit fly Drosophila melanogaster promotes total sleep (Funato et al. 2016). Thus, studies using invertebrate animal models are also expected to lead to the identification of key molecules that contribute to sleep regulation across the animal phyla. In this study, we focused on the transcription factor AP-2β (TFAP2B). TFAP2B is a member of the AP-2 transcription factor family, which is involved in diverse processes including cell proliferation, differentiation, and apoptosis at both developmental and mature stages (reviewed in (Eckert et al. 2005)). In mice, Tfap2b knockout pups die shortly after birth due to renal failure(Moser et al. 1997, 2003). Tfap2b is also crucial for limb formation and differentiation of several neuronal cells (Seok et al. 2008; Jin et al. 2015; Seki et al. 2015). In humans, mutations in TFAP2B lead to Char syndrome (MIM 169100) (Satoda et al. 2000). Char syndrome is a congenital disease characterized by patent ductus arteriosus, facial dysmorphism, and hand anomalies (Davidson 1993). Notably, two human kindreds with Char syndrome, kindred 144 and 145, self-reported abnormalities in sleep, namely parasomnia (sleep-walking) and short-sleep, respectively (Mani et al. 2005). The two kindreds harbor a mutation in the introns of TFAP2B, thus implying the involvement of TFAP2B in sleep regulation. Moreover, in the nematode Caenorhabditis elegans, the orthologue APTF-1 is crucial for specification of the sleep-promoting neuron RIS, and aptf-1 mutants exhibit severely reduced quiescence during both developmentally-timed sleep and stress-induced sleep (Turek et al. 2013, 2016; Konietzka et al. 2020). In addition, in the fruit fly Drosophila melanogaster, knockdown of neuronal TFAP-2 results in reduced night time sleep (Kucherenko et al. 2016). These findings, in combination with recent studies supporting that the molecular mechanisms of sleep are conserved between mammals and invertebrates, suggest that mammalian TFAP2B is involved in sleep regulation. However, in case of mammals, conclusive evidence was lacking because the direct effect of mutating this gene was unknown. In this study, we tested whether sleep is affected in mice deficient for Tfap2b. Furthermore, considering that the human kindreds 144 and 145 self-reported totally different sleep abnormalities, we generated two mutant mouse strains, each carrying a mutation in Tfap2b that mimicked the mutation in the human kindreds 144 and 145 by utilizing CRISPR/Cas9 genome editing, and examined their effects on the sleep architecture. 3 Methods Animals. All animal experiments were approved by the institutional animal care and use committee of the University of Tsukuba. All animals were maintained according to the institutional guidelines of the animal facilities of the Laboratory of Animal Resource Center, University of Tsukuba. Mice were maintained and bred in International Institute for Integrative Sleep Medicine under 12-hour light/dark cycle and free access to food and water. Male and female mice of 10-13 weeks of age were used for sleep analyses. To obtain Tfap2b+/- mice, Tfap2btm1a(EUCOMM)Wtsi/+ mice from KOMP (Knockout Mouse Project) (Skarnes et al. 2011) were crossed with mice carrying the Ayu1-cre allele (Niwa et al. 1993) to remove a genomic DNA fragment containing Tfap2b exon3 and a neomycin resistant cassette flanked by loxP. The Tfap2btm1a(EUCOMM)Wtsi allele contains an IRES:lacZ trapping cassette with Engrailed (En2) splice acceptor sequences immediately after Tfap2b exon2 and is followed by a poly-A transcription termination signals. F1 offspring were then maintained by crossing with C57BL/6J mice. Generation of Tfap2bK144 and Tfap2bK145 mice by CRISPR/Cas9 technology The Tfap2bK144 point mutation (c.601+5G>A) and the Tfap2bK145 point mutation (c.822-1G>C) were introduced to the mouse genome by CRISPR/Cas9 technology as previously reported with some modifications (Sato et al. 2018). The guide RNA (gRNA) was synthesized and purified using the GeneArt Precision gRNA Synthesis Kit (ThermoFisher). The sequences of the gRNA were 5‘-GTCAGTCATTAAAAAAGGTA-3’ for Tfap2bK144 and 5‘-TTTTCGATTTGGCTCTGCAG-3’ for Tfap2bK145. The donor oligonucleotides for Tfap2bK144 and Tfap2bK145 were both 101 bases (Integrated DNA Technologies), in which the desired point mutations were introduced. Pregnant mare serum gonadotropin and human chorionic gonadotropin were intraperitoneally injected into female C57BL/6J mice (Charles River Laboratories), and unfertilized oocytes were collected from their oviducts. We then performed in vitro fertilization with these oocytes and sperm from male C57BL/6J mice. The gRNA, donor oligonucleotide, and GeneArt Platinum Cas9 Nuclease (ThermoFisher) were electroporated into zygotes using a NEPA 21 electroporator (NEPAGNENE). After electroporation, embryos developed to the two-cell stage in KSOM- AA medium. The next day, the electroporated
Recommended publications
  • Watsonjn2018.Pdf (1.780Mb)
    UNIVERSITY OF CENTRAL OKLAHOMA Edmond, Oklahoma Department of Biology Investigating Differential Gene Expression in vivo of Cardiac Birth Defects in an Avian Model of Maternal Phenylketonuria A THESIS SUBMITTED TO THE GRADUATE FACULTY In partial fulfillment of the requirements For the degree of MASTER OF SCIENCE IN BIOLOGY By Jamie N. Watson Edmond, OK June 5, 2018 J. Watson/Dr. Nikki Seagraves ii J. Watson/Dr. Nikki Seagraves Acknowledgements It is difficult to articulate the amount of gratitude I have for the support and encouragement I have received throughout my master’s thesis. Many people have added value and support to my life during this time. I am thankful for the education, experience, and friendships I have gained at the University of Central Oklahoma. First, I would like to thank Dr. Nikki Seagraves for her mentorship and friendship. I lucked out when I met her. I have enjoyed working on this project and I am very thankful for her support. I would like thank Thomas Crane for his support and patience throughout my master’s degree. I would like to thank Dr. Shannon Conley for her continued mentorship and support. I would like to thank Liz Bullen and Dr. Eric Howard for their training and help on this project. I would like to thank Kristy Meyer for her friendship and help throughout graduate school. I would like to thank my committee members Dr. Robert Brennan and Dr. Lilian Chooback for their advisement on this project. Also, I would like to thank the biology faculty and staff. I would like to thank the Seagraves lab members: Jailene Canales, Kayley Pate, Mckayla Muse, Grace Thetford, Kody Harvey, Jordan Guffey, and Kayle Patatanian for their hard work and support.
    [Show full text]
  • Genome-Wide Expression Analysis Ofptf1a- and Ascl1-Deficient Mice
    The Journal of Neuroscience, April 24, 2013 • 33(17):7299–7307 • 7299 Development/Plasticity/Repair Genome-Wide Expression Analysis of Ptf1a- and Ascl1-Deficient Mice Reveals New Markers for Distinct Dorsal Horn Interneuron Populations Contributing to Nociceptive Reflex Plasticity Hendrik Wildner,1 Rebecca Das Gupta,2 Dominique Bro¨hl,3 Paul A. Heppenstall,4 Hanns Ulrich Zeilhofer,1,2* and Carmen Birchmeier3* 1Institute of Pharmacology and Toxicology, University of Zurich, CH-8057 Zurich, Switzerland, 2Institute of Pharmaceutical Sciences, Swiss Federal Institute of Technology (ETH) Zurich, CH-8093 Zurich, Switzerland, 3Department of Neuroscience, Max Delbru¨ck Center for Molecular Medicine, D-13125 Berlin, Germany, and 4Mouse Biology Unit, European Molecular Biology Laboratory, I-00015 Monterotondo, Italy Inhibitoryinterneuronsofthespinaldorsalhornplaycriticalrolesintheprocessingofnoxiousandinnocuoussensoryinformation.They form a family of morphologically and functionally diverse neurons that likely fall into distinct subtypes. Traditional classifications rely mainly on differences in dendritic tree morphology and firing patterns. Although useful, these markers are not comprehensive and cannot be used to drive specific genetic manipulations targeted at defined subsets of neurons. Here, we have used genome-wide expres- sion profiling of spinal dorsal horns of wild-type mice and of two strains of transcription factor-deficient mice (Ptf1aϪ/Ϫ and Ascl1/ Mash1Ϫ/Ϫ mice) to identify new genetic markers for specific subsets of dorsal horn inhibitory interneurons. Ptf1aϪ/Ϫ mice lack all inhibitory interneurons in the dorsal horn, whereas only the late-born inhibitory interneurons are missing in Ascl1Ϫ/Ϫ mice. We found 30 genes that were significantly downregulated in the dorsal horn of Ptf1aϪ/Ϫ mice. Twenty-one of those also showed reduced expression in Ascl1Ϫ/Ϫ mice.
    [Show full text]
  • Single Cell Regulatory Landscape of the Mouse Kidney Highlights Cellular Differentiation Programs and Disease Targets
    ARTICLE https://doi.org/10.1038/s41467-021-22266-1 OPEN Single cell regulatory landscape of the mouse kidney highlights cellular differentiation programs and disease targets Zhen Miao 1,2,3,8, Michael S. Balzer 1,2,8, Ziyuan Ma 1,2,8, Hongbo Liu1,2, Junnan Wu 1,2, Rojesh Shrestha 1,2, Tamas Aranyi1,2, Amy Kwan4, Ayano Kondo 4, Marco Pontoglio 5, Junhyong Kim6, ✉ Mingyao Li 7, Klaus H. Kaestner2,4 & Katalin Susztak 1,2,4 1234567890():,; Determining the epigenetic program that generates unique cell types in the kidney is critical for understanding cell-type heterogeneity during tissue homeostasis and injury response. Here, we profile open chromatin and gene expression in developing and adult mouse kidneys at single cell resolution. We show critical reliance of gene expression on distal regulatory elements (enhancers). We reveal key cell type-specific transcription factors and major gene- regulatory circuits for kidney cells. Dynamic chromatin and expression changes during nephron progenitor differentiation demonstrates that podocyte commitment occurs early and is associated with sustained Foxl1 expression. Renal tubule cells follow a more complex differentiation, where Hfn4a is associated with proximal and Tfap2b with distal fate. Mapping single nucleotide variants associated with human kidney disease implicates critical cell types, developmental stages, genes, and regulatory mechanisms. The single cell multi-omics atlas reveals key chromatin remodeling events and gene expression dynamics associated with kidney development. 1 Renal, Electrolyte, and Hypertension Division, Department of Medicine, University of Pennsylvania, Perelman School of Medicine, Philadelphia, PA, USA. 2 Institute for Diabetes, Obesity, and Metabolism, University of Pennsylvania, Perelman School of Medicine, Philadelphia, PA, USA.
    [Show full text]
  • Spatial Distribution of Leading Pacemaker Sites in the Normal, Intact Rat Sinoa
    Supplementary Material Supplementary Figure 1: Spatial distribution of leading pacemaker sites in the normal, intact rat sinoatrial 5 nodes (SAN) plotted along a normalized y-axis between the superior vena cava (SVC) and inferior vena 6 cava (IVC) and a scaled x-axis in millimeters (n = 8). Colors correspond to treatment condition (black: 7 baseline, blue: 100 µM Acetylcholine (ACh), red: 500 nM Isoproterenol (ISO)). 1 Supplementary Figure 2: Spatial distribution of leading pacemaker sites before and after surgical 3 separation of the rat SAN (n = 5). Top: Intact SAN preparations with leading pacemaker sites plotted during 4 baseline conditions. Bottom: Surgically cut SAN preparations with leading pacemaker sites plotted during 5 baseline conditions (black) and exposure to pharmacological stimulation (blue: 100 µM ACh, red: 500 nM 6 ISO). 2 a &DUGLDFIoQChDQQHOV .FQM FOXVWHU &DFQDG &DFQDK *MD &DFQJ .FQLS .FQG .FQK .FQM &DFQDF &DFQE .FQM í $WSD .FQD .FQM í .FQN &DVT 5\U .FQM &DFQJ &DFQDG ,WSU 6FQD &DFQDG .FQQ &DFQDJ &DFQDG .FQD .FQT 6FQD 3OQ 6FQD +FQ *MD ,WSU 6FQE +FQ *MG .FQN .FQQ .FQN .FQD .FQE .FQQ +FQ &DFQDD &DFQE &DOP .FQM .FQD .FQN .FQG .FQN &DOP 6FQD .FQD 6FQE 6FQD 6FQD ,WSU +FQ 6FQD 5\U 6FQD 6FQE 6FQD .FQQ .FQH 6FQD &DFQE 6FQE .FQM FOXVWHU V6$1 L6$1 5$ /$ 3 b &DUGLDFReFHSWRUV $GUDF FOXVWHU $GUDD &DY &KUQE &KUP &KJD 0\O 3GHG &KUQD $GUE $GUDG &KUQE 5JV í 9LS $GUDE 7SP í 5JV 7QQF 3GHE 0\K $GUE *QDL $QN $GUDD $QN $QN &KUP $GUDE $NDS $WSE 5DPS &KUP 0\O &KUQD 6UF &KUQH $GUE &KUQD FOXVWHU V6$1 L6$1 5$ /$ 4 c 1HXURQDOPURWHLQV
    [Show full text]
  • Transcriptional and Post-Transcriptional Regulation of ATP-Binding Cassette Transporter Expression
    Transcriptional and Post-transcriptional Regulation of ATP-binding Cassette Transporter Expression by Aparna Chhibber DISSERTATION Submitted in partial satisfaction of the requirements for the degree of DOCTOR OF PHILOSOPHY in Pharmaceutical Sciences and Pbarmacogenomies in the Copyright 2014 by Aparna Chhibber ii Acknowledgements First and foremost, I would like to thank my advisor, Dr. Deanna Kroetz. More than just a research advisor, Deanna has clearly made it a priority to guide her students to become better scientists, and I am grateful for the countless hours she has spent editing papers, developing presentations, discussing research, and so much more. I would not have made it this far without her support and guidance. My thesis committee has provided valuable advice through the years. Dr. Nadav Ahituv in particular has been a source of support from my first year in the graduate program as my academic advisor, qualifying exam committee chair, and finally thesis committee member. Dr. Kathy Giacomini graciously stepped in as a member of my thesis committee in my 3rd year, and Dr. Steven Brenner provided valuable input as thesis committee member in my 2nd year. My labmates over the past five years have been incredible colleagues and friends. Dr. Svetlana Markova first welcomed me into the lab and taught me numerous laboratory techniques, and has always been willing to act as a sounding board. Michael Martin has been my partner-in-crime in the lab from the beginning, and has made my days in lab fly by. Dr. Yingmei Lui has made the lab run smoothly, and has always been willing to jump in to help me at a moment’s notice.
    [Show full text]
  • Amacrine, Horizontal, and Retinal Ganglion Cells
    Biochemistry and Molecular Biology Jmjd3 Plays Pivotal Roles in the Proper Development of Early-Born Retinal Lineages: Amacrine, Horizontal, and Retinal Ganglion Cells Toshiro Iwagawa,1 Hiroaki Honda,2 and Sumiko Watanabe1 1Division of Molecular and Developmental Biology, Institute of Medical Science, University of Tokyo, Tokyo, Japan 2Field of Human Disease Models, Major in Advanced Life Sciences and Medicine, Institute of Laboratory Animals, Tokyo Women’s Medical University, Tokyo, Japan Correspondence: Sumiko Watanabe, PURPOSE. Trimethylation of histone H3 at lysine 27 (H3K27me3) is a critical mediator Division of Molecular and of transcriptional gene repression, and Jmjd3 and Utx are the demethylases specific to Developmental Biology, Institute of H3K27me3. Using an in vitro retinal explant culture system, we previously revealed the Medical Science, The University of role of Jmjd3 in the development of rod bipolar cells; however, the roles of Jmjd3 in the Tokyo, 4-6-1 Shirokanedai, development of early-born retinal cells are unknown due to limitations concerning the Minato-ku, Tokyo 108-8639, Japan; [email protected]. use of retinal explant culture systems. In this study, we investigated the roles of Jmjd3 in the development of early-born retinal cells. Received: April 12, 2020 Accepted: September 14, 2020 METHODS. We examined retina-specific conditional Jmjd3 knockout (Jmjd3-cKO) mice Published: September 28, 2020 using immunohistochemistry and quantitative reverse transcription PCR and JMJD3 bind- ing to a target locus by chromatin immunoprecipitation analysis. Citation: Iwagawa T, Honda H, Watanabe S. Jmjd3 plays pivotal RESULTS. We observed reductions in amacrine cells (ACs) and horizontal cells (HCs), as roles in the proper development of well as lowered expression levels of several transcription factors involved in the devel- early-born retinal lineages: opment of ACs and HCs in the Jmjd3-cKO mouse retina.
    [Show full text]
  • TFAP2 Transcription Factors Are Regulators of Lipid Droplet Biogenesis
    bioRxiv preprint doi: https://doi.org/10.1101/282376; this version posted March 14, 2018. The copyright holder for this preprint (which was not certified by peer review) is the author/funder, who has granted bioRxiv a license to display the preprint in perpetuity. It is made available under aCC-BY 4.0 International license. 1 TFAP2 transcription factors are 2 regulators of lipid droplet biogenesis 3 4 5 Cameron C Scott1, Stefania Vossio1, Jacques Rougemont2, Jean Gruenberg1,3 6 7 8 9 10 11 12 1. Department of Biochemistry, University of Geneva, 30 quai Ernest Ansermet 13 1211 Geneva 4, Switzerland 14 2. Department of Theoretical Physics, University of Geneva, 24 quai Ernest Ansermet 15 1211 Geneva 4, Switzerland 16 17 3. To whom correspondence should be addressed: [email protected] 18 19 20 21 22 1 bioRxiv preprint doi: https://doi.org/10.1101/282376; this version posted March 14, 2018. The copyright holder for this preprint (which was not certified by peer review) is the author/funder, who has granted bioRxiv a license to display the preprint in perpetuity. It is made available under aCC-BY 4.0 International license. 1 Abstract 2 How trafficking pathways and organelle abundance adapt in response to metabolic 3 and physiological changes is still mysterious, although a few transcriptional regulators of 4 organellar biogenesis have been identified in recent years. We previously found that the Wnt 5 signaling directly controls lipid droplet formation, linking the cell storage capacity to the 6 established functions of Wnt in development and differentiation. In the present paper, we 7 report that Wnt-induced lipid droplet biogenesis does not depend on the canonical TCF/LEF 8 transcription factors.
    [Show full text]
  • The Role of Inhibitor of DNA Binding 4 (Id4) in Mammary Gland Development and Breast Cancer
    The role of Inhibitor of DNA binding 4 (Id4) in mammary gland development and breast cancer Simon Junankar A thesis in fulfilment of the requirements for the degree of Doctor of Philosophy UNSW Garvan Institute of Medical Research St. Vincent’s Hospital Clinical School Faculty of Medicine November 2012 Copyright and Authenticity statement 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 the assistance from others in project’s design and conception or in style, presentation and linguistic expression is acknowledged.’ Signed: Date: 14/11/12 II Acknowledgements Firstly I would like to thank Alex Swarbrick for being a great supervisor, friend and mentor. You gave me the freedom to follow my own ideas and also gave direction when I needed it. I would also like to thank the other members of the Swarbrick group for their help, advice and friendship. In particular I would like to thank Radhika for conceptualising the Id4 project and for so much advice and help along the way.
    [Show full text]
  • © 2018 Alexandria Pinto All Rights Reserved
    © 2018 ALEXANDRIA PINTO ALL RIGHTS RESERVED ROLE OF Top2b IN PHOTORECEPTOR GENE REGULATORY NETWORK BY SINGLE-CELL TRANSCRIPTOME ANALYSIS BY ALEXANDRIA PINTO A thesis submitted to the School of Graduate Studies Rutgers, The State University of New Jersey In partial fulfillment of the requirements For the degree of Master of Science Graduate Program in Biomedical Engineering Written under the direction of Li Cai And approved by ___________________________________ ___________________________________ ___________________________________ ___________________________________ New Brunswick, New Jersey May, 2018 ABSTRACT OF THE THESIS ROLE OF Top2b IN PHOTORECEPTOR GENE REGULATORY NETWORK BY SINGLE-CELL TRANSCRIPTOME ANALYSIS By ALEXANDRIA PINTO Thesis Director: Dr. Li Cai TOP2B is an enzyme that allows for access to the DNA strand for gene transcription. During development, TOP2B is found in cells which have finished mitosis and proliferation, suggesting its function in cell differentiation. Previously, bulk RNA-seq analysis of the retina revealed TOP2B controls expression of genes in the photoreceptor gene-regulatory network. However, bulk RNA-seq does not allow for direct analysis of individual cells to identify the role of TOP2B in photoreceptor cell differentiation. The central hypothesis is that grouping cells based on the photoreceptor gene regulatory network and applying bioinformatics analysis to the data can show that TOP2B plays an essential role in proper photoreceptor differentiation. In this study, we preform bioinformatics analysis on publically available single-cell RNA-seq (scRNA-seq) dataset of postnatal day 14 mouse retina (GSE63473) to determine to role of TOP2B in the photoreceptor gene regulatory network and identify novel genes which contribute to this pathway. Analysis of photoreceptor scRNA-seq data reveals that TOP2B expression is ii correlated with the expression of photoreceptor marker genes, confirming its role in photoreceptor differentiation.
    [Show full text]
  • Robles JTO Supplemental Digital Content 1
    Supplementary Materials An Integrated Prognostic Classifier for Stage I Lung Adenocarcinoma based on mRNA, microRNA and DNA Methylation Biomarkers Ana I. Robles1, Eri Arai2, Ewy A. Mathé1, Hirokazu Okayama1, Aaron Schetter1, Derek Brown1, David Petersen3, Elise D. Bowman1, Rintaro Noro1, Judith A. Welsh1, Daniel C. Edelman3, Holly S. Stevenson3, Yonghong Wang3, Naoto Tsuchiya4, Takashi Kohno4, Vidar Skaug5, Steen Mollerup5, Aage Haugen5, Paul S. Meltzer3, Jun Yokota6, Yae Kanai2 and Curtis C. Harris1 Affiliations: 1Laboratory of Human Carcinogenesis, NCI-CCR, National Institutes of Health, Bethesda, MD 20892, USA. 2Division of Molecular Pathology, National Cancer Center Research Institute, Tokyo 104-0045, Japan. 3Genetics Branch, NCI-CCR, National Institutes of Health, Bethesda, MD 20892, USA. 4Division of Genome Biology, National Cancer Center Research Institute, Tokyo 104-0045, Japan. 5Department of Chemical and Biological Working Environment, National Institute of Occupational Health, NO-0033 Oslo, Norway. 6Genomics and Epigenomics of Cancer Prediction Program, Institute of Predictive and Personalized Medicine of Cancer (IMPPC), 08916 Badalona (Barcelona), Spain. List of Supplementary Materials Supplementary Materials and Methods Fig. S1. Hierarchical clustering of based on CpG sites differentially-methylated in Stage I ADC compared to non-tumor adjacent tissues. Fig. S2. Confirmatory pyrosequencing analysis of DNA methylation at the HOXA9 locus in Stage I ADC from a subset of the NCI microarray cohort. 1 Fig. S3. Methylation Beta-values for HOXA9 probe cg26521404 in Stage I ADC samples from Japan. Fig. S4. Kaplan-Meier analysis of HOXA9 promoter methylation in a published cohort of Stage I lung ADC (J Clin Oncol 2013;31(32):4140-7). Fig. S5. Kaplan-Meier analysis of a combined prognostic biomarker in Stage I lung ADC.
    [Show full text]
  • Downloadedfrommultiple from Poplar, and We Could Identify TF Clusters That Can Resources
    Nie et al. BMC Systems Biology 2011, 5:53 http://www.biomedcentral.com/1752-0509/5/53 METHODOLOGYARTICLE Open Access TF-Cluster: A pipeline for identifying functionally coordinated transcription factors via network decomposition of the shared coexpression connectivity matrix (SCCM) Jeff Nie1†, Ron Stewart1†, Hang Zhang6, James A Thomson1,2,3,9, Fang Ruan7, Xiaoqi Cui5 and Hairong Wei4,8* Abstract Background: Identifying the key transcription factors (TFs) controlling a biological process is the first step toward a better understanding of underpinning regulatory mechanisms. However, due to the involvement of a large number of genes and complex interactions in gene regulatory networks, identifying TFs involved in a biological process remains particularly difficult. The challenges include: (1) Most eukaryotic genomes encode thousands of TFs, which are organized in gene families of various sizes and in many cases with poor sequence conservation, making it difficult to recognize TFs for a biological process; (2) Transcription usually involves several hundred genes that generate a combination of intrinsic noise from upstream signaling networks and lead to fluctuations in transcription; (3) A TF can function in different cell types or developmental stages. Currently, the methods available for identifying TFs involved in biological processes are still very scarce, and the development of novel, more powerful methods is desperately needed. Results: We developed a computational pipeline called TF-Cluster for identifying functionally coordinated TFs
    [Show full text]
  • Reprogramming of Avian Neural Crest Axial Identity and Cell Fate
    HHS Public Access Author manuscript Author ManuscriptAuthor Manuscript Author Science Manuscript Author . Author manuscript; Manuscript Author available in PMC 2016 December 24. Published in final edited form as: Science. 2016 June 24; 352(6293): 1570–1573. doi:10.1126/science.aaf2729. Reprogramming of avian neural crest axial identity and cell fate Marcos Simoes-Costa1 and Marianne E. Bronner1 1Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, CA 91125, USA Abstract Neural crest populations along the embryonic body axis differ in developmental potential and fate, such that only cranial neural crest can contribute to craniofacial skeleton in vivo. Here, we explore the regulatory program that imbues the cranial crest with its unique features. Using axial-level specific enhancers to isolate and perform genome-wide profiling of cranial versus trunk neural crest in chick embryos, we identify and characterize regulatory relationships between a set of cranial-specific transcription factors. Introducing components of this circuit into neural crest cells of the trunk alters their identity and endows these cells with the ability to give rise to chondroblasts in vivo. Our results demonstrate that gene regulatory circuits that support formation of particular neural crest derivatives may be employed for reprogramming specific neural crest derived cell types. Neural crest cells are characterized by multipotency and migratory ability. During embryonic development, the neural crest differentiates into multiple cell types, including chondrocytes and osteocytes, melanocytes, and neurons and glia of the peripheral nervous system (1,2). Neural crest stem cells are retained postnatally in skin and peripheral nerves, providing a potential target for replacement therapy in regenerative medicine (3–4).
    [Show full text]