Pitx3 KNOCKOUT MICE ENTRAIN to SCHEDULED FEEDING DESPITE

Total Page:16

File Type:pdf, Size:1020Kb

Pitx3 KNOCKOUT MICE ENTRAIN to SCHEDULED FEEDING DESPITE Pitx3 KNOCKOUT MICE ENTRAIN TO SCHEDULED FEEDING DESPITE FREE-RUNNING LIGHT ENTRAINED RHYTHMS A Thesis Presented to the Faculty of California State Polytechnic University, Pomona In Partial Fulfillment Of the Requirements for the Degree Master of Science In Biological Sciences By Lori L. Scarpa 2019 SIGNATURE PAGE THESIS: Pitx3 KNOCKOUT MICE ENTRAIN TO SCHEDULED FEEINDING DESPITE FREE-RUNNING LIGHT ENTRAINED RHYTHMS AUTHOR: Lori L. Scarpa DATE SUBMITTED: Fall 2019 Department of Biological Sciences Dr. Andrew D. Steele Thesis Committee Chair Biological Sciences Dr. Juanita Jellyman Biological Sciences Dr. Robert Talmadge Biological Sciences ii ACKNOWLEDGEMENTS Funding for this project was provided by the Whitehall Foundation and California State Polytechnic University of Pomona, California. I would like to thank the many people who assisted in the daily labors required to complete this project: Raymundo Miranda, Michael Sidikpramana, Jeffrey Falkenstein, Michael Williams, Jaskaran Dhanoa, and many other members of the Steele lab. I would like to specially recognize fellow graduate students in the Steele lab, Damien Wolfe and Andrew Villa, for their unconditional encouragement and support. I would like to thank Dr. Juanita Jellyman for her kindness and nurturing spirit. Dr. Jellyman never wavered her belief in me and it was this that kept me working harder towards my goals. Lastly, I would like thank Dr. Andrew Steele for accepting me into his lab, for being my personal mentor, and for pushing me towards success. Through him, I have grown to become a better scientist and found greater inspiration in my pursuit to further study neuroscience. Thank you, Dr. Steele, for your support and patience, and for playing a leading role in my success as a graduate student in your lab. iii ABSTRACT It is well known that many of our biological processes are regulated by circadian rhythms. Photic input is the best characterized stimulus for entraining circadian rhythms. In mammals, light input is conveyed to a brain structure called the suprachiasmatic nucleus (SCN), which serves as a central clock, controlling the timing of sleep-wake, hormone secretion, and many other physiological functions. Feeding is another modulator of circadian rhythms, acting in an SCN-independent manner, as rodents with lesions to the SCN show normal anticipation of scheduled meals. Recent studies implicate dopamine signaling as a key mediator of entraining circadian rhythms to scheduled feeding. Mice with impairments in dopamine production or in dopamine receptors show decreased food anticipatory activity (FAA), a measure of food entrainment. Here, we are studying the influence of food on circadian rhythms in paired- like homeodomain 3 (Pitx3) knockout mice, which are severely depleted for dopamine neurons of the substantia nigra. These knockout mice also lack retinal ganglion cells and exhibit free-running behavioral rhythms that are not linked to the light:dark cycle, making it difficult to determine whether their activity entrains to scheduled feeding. By making continuous measurements of their activity, our home cage behavioral data suggests that the Pitx3 knockouts exhibit FAA. In addition, we demonstrated that metabolic rhythms in these mice entrain to scheduled feeding rapidly. To study the contribution of Pitx3-expressing dopamine neurons to FAA in a cleaner system, we attempted to perform a follow-up experiment where we deleted the dopamine producing enzyme, tyrosine hydroxylase, from Pitx3 expressing neurons. However, these mice die about 2 weeks after birth, making it impossible for us to study food entrainment in this iv conditional deletion model. Future studies to further verify our findings that a small population of dopamine neurons in the midbrain are sufficient for FAA are warranted. v TABLE OF CONTENTS SIGNATURE PAGE ..........................................................................................................ii ACKNOWLEDGEMENTS ..............................................................................................iii ABSTRACT ......................................................................................................................iv LIST OF TABLES............................................................................................................ vii LIST OF FIGURES .........................................................................................................viii INTRODUCTION ..............................................................................................................1 METHODS AND MATERIALS .......................................................................................11 RESULTS ..........................................................................................................................17 DISCUSSION ....................................................................................................................50 REFERENCES .................................................................................................................55 vi LIST OF TABLES Table 1. Primer sequences for amplification of Pitx3....................................................... 14 vii LIST OF FIGURES Figure 1: Transcriptional-translational feedback loop controlling circadian rhythms........ 3 Figure 2: Average daily food intake measurements of Pitx3 control and Pitx3 KO mice.22 Figure 3: Survival plot of Pitx3 mice on 60% CR. ........................................................... 22 Figure 4: Average weekly body weights of Pitx3 control and Pitx3 KO mice on 60% CR. ........................................................................................................................................... 23 Figure 5: Average weekly activity measurements from video recording in Pitx3 control and Pitx3 KO mice on 60% CR. ....................................................................................... 23 Figure 6: Weekly activity waveforms on 60% CR. .......................................................... 24 Figure 7: Activity monitoring of individual Pitx3 mice on 60% CR via photobeam breaks. ........................................................................................................................................... 25 Figure 8: Survival plot of Pitx3 mice on 80% CR. ........................................................... 29 Figure 9: Average weekly body weights of Pitx3 control and Pitx3 KO mice on 80% CR. ........................................................................................................................................... 29 Figure 10: Average weekly activity measurements from video recording in Pitx3 control and Pitx3 KO mice on 80% CR. ....................................................................................... 30 Figure 11: Weekly activity waveforms on 80% CR. ........................................................ 31 Figure 12: Activity monitoring of individual Pitx3 mice on 80% CR via photobeam breaks. ............................................................................................................................... 35 Figure 13: Activity monitoring of individual Pitx3 control mice on 60% CR via photobeam breaks. ............................................................................................................ 36 Figure 14: Activity monitoring of individual Pitx3 KO mice on 80% CR via photobeam breaks. ............................................................................................................................... 37 Figure 15: Averaged activity waveforms on 80% CR. ..................................................... 40 Figure 16: Total activity and FAA ratios on AL and 80% CR. ........................................ 39 Figure 17: Average body composition measurements in Pitx3 control and Pitx3 KO mice on AL. ............................................................................................................................... 42 Figure 18: Average waveforms of total activity in Pitx3 mice on AL and 80% CR. ....... 43 Figure 19: Gas exchange of Pitx3 mice on AL and 80% CR. .......................................... 43 Figure 20: Identification of midbrain dopaminergic neurons via IF staining for TH and DAPI at 10x magnification in Pitx3 control brain tissue. ................................................. 45 Figure 21: Identification of midbrain dopaminergic neurons via IF staining for TH and DAPI at 10x magnification in Pitx3 KO brain tissue. ...................................................... 46 Figure 22: In situ Hybridization staining for TH and DAPI at 20x magnification. .......... 47 Figure 23: Visualization of dopaminergic projections via IF staining for TH and DAPI. 47 Figure 24: Visualization of dopaminergic positive areas in the forebrain of Pitx3 mice. 48 Figure 25: Survival plot of Pitx3-cre mice during first 30 days of life…………………..52 viii INTRODUCTION Biological Rhythms In 1729, Jacques d’Ortous de Mairan demonstrated that the leaves of the mimosa plant continued to open and close with the same daily rhythm when kept in constant darkness (de Marian, 1729). This experiment was the first evidence of an autonomous clock regulating biological rhythms even. Biological rhythms are natural cycles of change in a body’s function. In order to qualify as a biological rhythm, the cycle must persist in the absence of a time-cue, often referred to as a zeitgeber (Aschoff, 1965). There are three main types of biological rhythms: circadian, infradian,
Recommended publications
  • Detailed Review Paper on Retinoid Pathway Signalling
    1 1 Detailed Review Paper on Retinoid Pathway Signalling 2 December 2020 3 2 4 Foreword 5 1. Project 4.97 to develop a Detailed Review Paper (DRP) on the Retinoid System 6 was added to the Test Guidelines Programme work plan in 2015. The project was 7 originally proposed by Sweden and the European Commission later joined the project as 8 a co-lead. In 2019, the OECD Secretariat was added to coordinate input from expert 9 consultants. The initial objectives of the project were to: 10 draft a review of the biology of retinoid signalling pathway, 11 describe retinoid-mediated effects on various organ systems, 12 identify relevant retinoid in vitro and ex vivo assays that measure mechanistic 13 effects of chemicals for development, and 14 Identify in vivo endpoints that could be added to existing test guidelines to 15 identify chemical effects on retinoid pathway signalling. 16 2. This DRP is intended to expand the recommendations for the retinoid pathway 17 included in the OECD Detailed Review Paper on the State of the Science on Novel In 18 vitro and In vivo Screening and Testing Methods and Endpoints for Evaluating 19 Endocrine Disruptors (DRP No 178). The retinoid signalling pathway was one of seven 20 endocrine pathways considered to be susceptible to environmental endocrine disruption 21 and for which relevant endpoints could be measured in new or existing OECD Test 22 Guidelines for evaluating endocrine disruption. Due to the complexity of retinoid 23 signalling across multiple organ systems, this effort was foreseen as a multi-step process.
    [Show full text]
  • Noninvasive Sleep Monitoring in Large-Scale Screening of Knock-Out Mice
    bioRxiv preprint doi: https://doi.org/10.1101/517680; this version posted January 11, 2019. The copyright holder for this preprint (which was not certified by peer review) is the author/funder, who has granted bioRxiv a license to display the preprint in perpetuity. It is made available under aCC-BY-ND 4.0 International license. Noninvasive sleep monitoring in large-scale screening of knock-out mice reveals novel sleep-related genes Shreyas S. Joshi1*, Mansi Sethi1*, Martin Striz1, Neil Cole2, James M. Denegre2, Jennifer Ryan2, Michael E. Lhamon3, Anuj Agarwal3, Steve Murray2, Robert E. Braun2, David W. Fardo4, Vivek Kumar2, Kevin D. Donohue3,5, Sridhar Sunderam6, Elissa J. Chesler2, Karen L. Svenson2, Bruce F. O'Hara1,3 1Dept. of Biology, University of Kentucky, Lexington, KY 40506, USA, 2The Jackson Laboratory, Bar Harbor, ME 04609, USA, 3Signal solutions, LLC, Lexington, KY 40503, USA, 4Dept. of Biostatistics, University of Kentucky, Lexington, KY 40536, USA, 5Dept. of Electrical and Computer Engineering, University of Kentucky, Lexington, KY 40506, USA. 6Dept. of Biomedical Engineering, University of Kentucky, Lexington, KY 40506, USA. *These authors contributed equally Address for correspondence and proofs: Shreyas S. Joshi, Ph.D. Dept. of Biology University of Kentucky 675 Rose Street 101 Morgan Building Lexington, KY 40506 U.S.A. Phone: (859) 257-2805 FAX: (859) 257-1717 Email: [email protected] Running title: Sleep changes in knockout mice bioRxiv preprint doi: https://doi.org/10.1101/517680; this version posted January 11, 2019. The copyright holder for this preprint (which was not certified by peer review) is the author/funder, who has granted bioRxiv a license to display the preprint in perpetuity.
    [Show full text]
  • Identification of Novel Sleep Related Genes from Large Scale Phenotyping Experiments in Mice
    University of Kentucky UKnowledge Theses and Dissertations--Biology Biology 2017 IDENTIFICATION OF NOVEL SLEEP RELATED GENES FROM LARGE SCALE PHENOTYPING EXPERIMENTS IN MICE Shreyas Joshi University of Kentucky, [email protected] Digital Object Identifier: https://doi.org/10.13023/ETD.2017.159 Right click to open a feedback form in a new tab to let us know how this document benefits ou.y Recommended Citation Joshi, Shreyas, "IDENTIFICATION OF NOVEL SLEEP RELATED GENES FROM LARGE SCALE PHENOTYPING EXPERIMENTS IN MICE" (2017). Theses and Dissertations--Biology. 42. https://uknowledge.uky.edu/biology_etds/42 This Doctoral Dissertation is brought to you for free and open access by the Biology at UKnowledge. It has been accepted for inclusion in Theses and Dissertations--Biology by an authorized administrator of UKnowledge. For more information, please contact [email protected]. STUDENT AGREEMENT: I represent that my thesis or dissertation and abstract are my original work. Proper attribution has been given to all outside sources. I understand that I am solely responsible for obtaining any needed copyright permissions. I have obtained needed written permission statement(s) from the owner(s) of each third-party copyrighted matter to be included in my work, allowing electronic distribution (if such use is not permitted by the fair use doctrine) which will be submitted to UKnowledge as Additional File. I hereby grant to The University of Kentucky and its agents the irrevocable, non-exclusive, and royalty-free license to archive and make accessible my work in whole or in part in all forms of media, now or hereafter known.
    [Show full text]
  • SUPPLEMENTARY MATERIAL Bone Morphogenetic Protein 4 Promotes
    www.intjdevbiol.com doi: 10.1387/ijdb.160040mk SUPPLEMENTARY MATERIAL corresponding to: Bone morphogenetic protein 4 promotes craniofacial neural crest induction from human pluripotent stem cells SUMIYO MIMURA, MIKA SUGA, KAORI OKADA, MASAKI KINEHARA, HIROKI NIKAWA and MIHO K. FURUE* *Address correspondence to: Miho Kusuda Furue. Laboratory of Stem Cell Cultures, National Institutes of Biomedical Innovation, Health and Nutrition, 7-6-8, Saito-Asagi, Ibaraki, Osaka 567-0085, Japan. Tel: 81-72-641-9819. Fax: 81-72-641-9812. E-mail: [email protected] Full text for this paper is available at: http://dx.doi.org/10.1387/ijdb.160040mk TABLE S1 PRIMER LIST FOR QRT-PCR Gene forward reverse AP2α AATTTCTCAACCGACAACATT ATCTGTTTTGTAGCCAGGAGC CDX2 CTGGAGCTGGAGAAGGAGTTTC ATTTTAACCTGCCTCTCAGAGAGC DLX1 AGTTTGCAGTTGCAGGCTTT CCCTGCTTCATCAGCTTCTT FOXD3 CAGCGGTTCGGCGGGAGG TGAGTGAGAGGTTGTGGCGGATG GAPDH CAAAGTTGTCATGGATGACC CCATGGAGAAGGCTGGGG MSX1 GGATCAGACTTCGGAGAGTGAACT GCCTTCCCTTTAACCCTCACA NANOG TGAACCTCAGCTACAAACAG TGGTGGTAGGAAGAGTAAAG OCT4 GACAGGGGGAGGGGAGGAGCTAGG CTTCCCTCCAACCAGTTGCCCCAAA PAX3 TTGCAATGGCCTCTCAC AGGGGAGAGCGCGTAATC PAX6 GTCCATCTTTGCTTGGGAAA TAGCCAGGTTGCGAAGAACT p75 TCATCCCTGTCTATTGCTCCA TGTTCTGCTTGCAGCTGTTC SOX9 AATGGAGCAGCGAAATCAAC CAGAGAGATTTAGCACACTGATC SOX10 GACCAGTACCCGCACCTG CGCTTGTCACTTTCGTTCAG Suppl. Fig. S1. Comparison of the gene expression profiles of the ES cells and the cells induced by NC and NC-B condition. Scatter plots compares the normalized expression of every gene on the array (refer to Table S3). The central line
    [Show full text]
  • Investigating the Genetic Basis of Altered Activity Profiles in the Blind
    Investigating the genetic basis of altered activity profiles in the blind Mexican cavefish, Astyanax mexicanus A dissertation submitted to the Graduate School of the University of Cincinnati in partial fulfillment of the requirements for the degree of Doctor of Philosophy in the Department of Biological Sciences of the McMicken College of Arts and Sciences by by Brian M. Carlson B.S. Biology, Xavier University, May 2010 Committee Chair: Dr. Joshua B. Gross June 2015 ABSTRACT Organisms that have evolved to exploit extreme ecological niches may alter or abandon survival strategies that no longer provide a benefit, or may even impose a cost, in the environment to which they have adapted. Cave environments are characterized by perpetual darkness, isolation and relatively constant temperature and humidity. Accordingly, cave-adapted species tend to converge on a suite of regressive and constructive morphological, physiological and behavioral alterations, including loss or reduction of eyes and pigmentation, increased locomotor activity and reduction or alteration of behavioral rhythmicity. The cave environment and the associated changes in locomotor behavior make species of cavefish prime natural models in which to examine the complex genetic architecture underlying these behavioral phenotypes. The principal goal of this dissertation was to investigate the genetic basis of altered locomotor activity patterns in the blind Mexican tetra, Astyanax mexicanus. Initially, a custom locomotor assay rig and experimental protocols were developed to assess, characterize and compare activity patterns in surface and Pachón cavefish. The results of these assays clarified differences between the morphotypes, provided evidence that Pachón cavefish retain a weakly-entrainable circadian oscillator with limited capacity to self-sustain entrained rhythms and suggested that patterns in spatial “tank usage” data may be the result of a positive masking effect in response to light stimulus in both morphotypes.
    [Show full text]
  • A Chromosome Level Genome of Astyanax Mexicanus Surface Fish for Comparing Population
    bioRxiv preprint doi: https://doi.org/10.1101/2020.07.06.189654; this version posted July 6, 2020. 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 Title 2 A chromosome level genome of Astyanax mexicanus surface fish for comparing population- 3 specific genetic differences contributing to trait evolution. 4 5 Authors 6 Wesley C. Warren1, Tyler E. Boggs2, Richard Borowsky3, Brian M. Carlson4, Estephany 7 Ferrufino5, Joshua B. Gross2, LaDeana Hillier6, Zhilian Hu7, Alex C. Keene8, Alexander Kenzior9, 8 Johanna E. Kowalko5, Chad Tomlinson10, Milinn Kremitzki10, Madeleine E. Lemieux11, Tina 9 Graves-Lindsay10, Suzanne E. McGaugh12, Jeff T. Miller12, Mathilda Mommersteeg7, Rachel L. 10 Moran12, Robert Peuß9, Edward Rice1, Misty R. Riddle13, Itzel Sifuentes-Romero5, Bethany A. 11 Stanhope5,8, Clifford J. Tabin13, Sunishka Thakur5, Yamamoto Yoshiyuki14, Nicolas Rohner9,15 12 13 Authors for correspondence: Wesley C. Warren ([email protected]), Nicolas Rohner 14 ([email protected]) 15 16 Affiliation 17 1Department of Animal Sciences, Department of Surgery, Institute for Data Science and 18 Informatics, University of Missouri, Bond Life Sciences Center, Columbia, MO 19 2 Department of Biological Sciences, University of Cincinnati, Cincinnati, OH 20 3 Department of Biology, New York University, New York, NY 21 4 Department of Biology, The College of Wooster, Wooster, OH 22 5 Harriet L. Wilkes Honors College, Florida Atlantic University, Jupiter FL 23 6 Department of Genome Sciences, University of Washington, Seattle, WA 1 bioRxiv preprint doi: https://doi.org/10.1101/2020.07.06.189654; this version posted July 6, 2020.
    [Show full text]
  • Fig1-13Tab1-5.Pdf
    Supplementary Information Promoter hypomethylation of EpCAM-regulated bone morphogenetic protein genes in advanced endometrial cancer Ya-Ting Hsu, Fei Gu, Yi-Wen Huang, Joseph Liu, Jianhua Ruan, Rui-Lan Huang, Chiou-Miin Wang, Chun-Liang Chen, Rohit R. Jadhav, Hung-Cheng Lai, David G. Mutch, Paul J. Goodfellow, Ian M. Thompson, Nameer B. Kirma, and Tim Hui-Ming Huang Tables of contents Page Table of contents 2 Supplementary Methods 4 Supplementary Figure S1. Summarized sequencing reads and coverage of MBDCap-seq 8 Supplementary Figure S2. Reproducibility test of MBDCap-seq 10 Supplementary Figure S3. Validation of MBDCap-seq by MassARRAY analysis 11 Supplementary Figure S4. Distribution of differentially methylated regions (DMRs) in endometrial tumors relative to normal control 12 Supplementary Figure S5. Network analysis of differential methylation loci by using Steiner-tree analysis 13 Supplementary Figure S6. DNA methylation distribution in early and late stage of the TCGA endometrial cancer cohort 14 Supplementary Figure S7. Relative expression of BMP genes with EGF treatment in the presence or absence of PI3K/AKT and Raf (MAPK) inhibitors in endometrial cancer cells 15 Supplementary Figure S8. Induction of invasion by EGF in AN3CA and HEC1A cell lines 16 Supplementary Figure S9. Knockdown expression of BMP4 and BMP7 in RL95-2 cells 17 Supplementary Figure S10. Relative expression of BMPs and BMPRs in normal endometrial cell and endometrial cancer cell lines 18 Supplementary Figure S11. Microfluidics-based PCR analysis of EMT gene panel in RL95-2 cells with or without EGF treatment 19 Supplementary Figure S12. Knockdown expression of EpCAM by different shRNA sequences in RL95-2 cells 20 Supplementary Figure S13.
    [Show full text]
  • A Novel Mutation in the OAR Domain of PITX3 Associated with Congenital
    Fan et al. BMC Medical Genetics (2019) 20:42 https://doi.org/10.1186/s12881-019-0782-2 RESEARCH ARTICLE Open Access A novel mutation in the OAR domain of PITX3 associated with congenital posterior subcapsular cataract Qi Fan1,2,3,4,5, Dan Li1,2,3,4,5, Lei Cai1,2,3,4,5, Xiaodi Qiu1,2,3,4,5, Zhennan Zhao1,2,3,4,5, Jihong Wu1,2,3,4,5, Jin Yang1,2,3,4,5*† and Yi Lu1,2,3,4,5*† Abstract Background: Congenital cataract is the most common cause of blindness among children worldwide. The aim of this study was to identify causative mutations in a Chinese family with isolated autosomal dominant posterior subcapsular cataract. Methods: The proband and her parents underwent full ophthalmological examinations. DNA was extracted from the participants’ peripheral venous blood. The mutation was identified via panel-based next-generation sequencing (NGS) and was validated via Sanger sequencing. Results: Posterior subcapsular lenticular opacity was observed in both of the proband’s eyes. The novel deletion mutation c.797_814del, p.Ser266_Ala271del in the PITX3 gene was identified in the proband and her father. This mutation is located within the otp/aristaless/rax (OAR) domain at the COOH-terminus of the protein, which functions in DNA binding and transactivation. This mutation would result in a deletion of 6 amino acid residues at the C terminal of the protein. Conclusions: The mutation c.797_814del, p.Ser266_Ala271del is a novel mutation in the conserved DNA-binding OAR domain of PITX3 that causes congenital cataract. Keywords: Congenital cataract, PITX3, The OAR domain Background 100 causative genes for congenital cataract, including Congenital cataract is defined as lens opacity that occurs at many with minor additional features (e.g., ABCA3 [7], birth or during the first decade of life.
    [Show full text]
  • Notch Signaling Regulates Growth and Differentiation in the Mammalian Lens
    Developmental Biology 321 (2008) 111–122 Contents lists available at ScienceDirect Developmental Biology journal homepage: www.elsevier.com/developmentalbiology Notch signaling regulates growth and differentiation in the mammalian lens Sheldon Rowan a, Kevin W. Conley b, Tien T. Le b, Amy L. Donner a, Richard L. Maas a, Nadean L. Brown b,⁎ a Division of Genetics, Department of Medicine, Brigham and Women’s Hospital and Harvard Medical School, Boston, MA 02115, USA b Division of Developmental Biology, Children’s Hospital Research Foundation, Departments of Pediatrics and Ophthalmology University of Cincinnati College of Medicine, 3333 Burnet Avenue, Cincinnati, OH 45229-7007, USA article info abstract Article history: The Notch signal transduction pathway regulates the decision to proliferate versus differentiate. Although Received for publication 25 April 2008 there are a myriad of mouse models for the Notch pathway, surprisingly little is known about how these Revised 2 June 2008 genes regulate early eye development, particularly in the anterior lens. We employed both gain-of-function Accepted 3 June 2008 and loss-of-function approaches to determine the role of Notch signaling in lens development. Here we Available online 13 June 2008 analyzed mice containing conditional deletion of the Notch effector Rbpj or overexpression of the activated Notch1 intracellular domain during lens formation. We demonstrate distinct functions for Notch signaling in Keywords: fi Lens development progenitor cell growth, ber cell differentiation and maintenance of the transition zone. In particular, Notch Notch signaling signaling controls the timing of primary fiber cell differentiation and is essential for secondary fiber cell Rbpj differentiation. Either gain or loss of Notch signaling leads to formation of a dysgenic lens, which in loss-of- Activated Notch function mice undergoes a profound postnatal degeneration.
    [Show full text]
  • 1 Novel Expression Signatures Identified by Transcriptional Analysis
    ARD Online First, published on October 7, 2009 as 10.1136/ard.2009.108043 Ann Rheum Dis: first published as 10.1136/ard.2009.108043 on 7 October 2009. Downloaded from Novel expression signatures identified by transcriptional analysis of separated leukocyte subsets in SLE and vasculitis 1Paul A Lyons, 1Eoin F McKinney, 1Tim F Rayner, 1Alexander Hatton, 1Hayley B Woffendin, 1Maria Koukoulaki, 2Thomas C Freeman, 1David RW Jayne, 1Afzal N Chaudhry, and 1Kenneth GC Smith. 1Cambridge Institute for Medical Research and Department of Medicine, Addenbrooke’s Hospital, Hills Road, Cambridge, CB2 0XY, UK 2Roslin Institute, University of Edinburgh, Roslin, Midlothian, EH25 9PS, UK Correspondence should be addressed to Dr Paul Lyons or Prof Kenneth Smith, Department of Medicine, Cambridge Institute for Medical Research, Addenbrooke’s Hospital, Hills Road, Cambridge, CB2 0XY, UK. Telephone: +44 1223 762642, Fax: +44 1223 762640, E-mail: [email protected] or [email protected] Key words: Gene expression, autoimmune disease, SLE, vasculitis Word count: 2,906 The Corresponding Author has the right to grant on behalf of all authors and does grant on behalf of all authors, an exclusive licence (or non-exclusive for government employees) on a worldwide basis to the BMJ Publishing Group Ltd and its Licensees to permit this article (if accepted) to be published in Annals of the Rheumatic Diseases and any other BMJPGL products to exploit all subsidiary rights, as set out in their licence (http://ard.bmj.com/ifora/licence.pdf). http://ard.bmj.com/ on September 29, 2021 by guest. Protected copyright. 1 Copyright Article author (or their employer) 2009.
    [Show full text]
  • Molecular Regulation in Dopaminergic Neuron Development
    International Journal of Molecular Sciences Review Molecular Regulation in Dopaminergic Neuron Development. Cues to Unveil Molecular Pathogenesis and Pharmacological Targets of Neurodegeneration Floriana Volpicelli 1 , Carla Perrone-Capano 1,2 , Gian Carlo Bellenchi 2,3, Luca Colucci-D’Amato 4,* and Umberto di Porzio 2 1 Department of Pharmacy, University of Naples Federico II, 80131 Naples, Italy; fl[email protected] (F.V.); [email protected] (C.P.C.) 2 Institute of Genetics and Biophysics “Adriano Buzzati Traverso”, CNR, 80131 Rome, Italy; [email protected] (G.C.B.); [email protected] (U.d.P.) 3 Department of Systems Medicine, University of Rome Tor Vergata, 00133 Rome, Italy 4 Department of Environmental, Biological and Pharmaceutical Sciences and Technologies, University of Campania “Luigi Vanvitelli”, 81100 Caserta, Italy * Correspondence: [email protected]; Tel.: +39-0823-274577 Received: 28 April 2020; Accepted: 1 June 2020; Published: 3 June 2020 Abstract: The relatively few dopaminergic neurons in the mammalian brain are mostly located in the midbrain and regulate many important neural functions, including motor integration, cognition, emotive behaviors and reward. Therefore, alteration of their function or degeneration leads to severe neurological and neuropsychiatric diseases. Unraveling the mechanisms of midbrain dopaminergic (mDA) phenotype induction and maturation and elucidating the role of the gene network involved in the development and maintenance of these neurons is of pivotal importance to rescue or substitute these cells in order to restore dopaminergic functions. Recently, in addition to morphogens and transcription factors, microRNAs have been identified as critical players to confer mDA identity. The elucidation of the gene network involved in mDA neuron development and function will be crucial to identify early changes of mDA neurons that occur in pre-symptomatic pathological conditions, such as Parkinson’s disease.
    [Show full text]
  • Multiple Coregulatory Control of Tyrosine Hydroxylase Gene Transcription
    Multiple coregulatory control of tyrosine hydroxylase gene transcription Sirigiri Divijendra Natha Reddya,b,1, Suresh K. Rayalac,1,2, Kazufumi Ohshiroa,b,c,1, Suresh B. Pakalaa,b, Nobuhide Koborid, Pramod Dashe, Sung Yunf, Jun Qinf,3, Bert W. O’Malleyf,3, and Rakesh Kumara,b,c,f,3 aDepartment of Biochemistry and Molecular Biology and bInstitute of Coregulator Biology, George Washington University Medical Center, Washington, DC 20037; cDepartment of Molecular and Cellular Oncology, MD Anderson Cancer Center, Houston, TX 77030; Departments of dNeurosurgery and eNeurobiology and Anatomy, University of Texas Medical School, Houston, TX 77030; and fDepartment of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX 77030 Contributed by Bert W. O’Malley, January 27, 2011 (sent for review December 16, 2010) Despite ubiquitous expression and a high level of metastasis- future strategies and modalities to treat or slow progression of associated protein 1 (MTA1) coregulator, the physiological role of the diseases associated with TH regulation. Several previous the MTA1 coactivator remains unknown. We found that MTA1 is a studies have attempted to identify the factors important in TH gene bona fide coactivator and stimulator of tyrosine hydroxylase (TH) expression in human (8, 9), mouse (10), and rat (11) models. For transcription in neuronal cells and that MTA1-null mice had lower example, TH expression has been shown to be regulated positively TH expression in the striatum and substantial nigra. MTA1 physically by the DJ1 coregulator (12). More recently, epigenetic profiling of achieves these functions by interacting directly with DJ1 (Parkinson the human TH promoter has suggested that chromatin remodeling disease 7) and in turn recruits the DJ1/MTA1/RNA polymerase II could have a significant impact on conferring tissue-specific gene complex to the bicoid binding element (BBE) in the TH promoter.
    [Show full text]