“The Interplay of Central and Peripheral Circadian Clocks in White Adipose Function and Metabolic Homeostasis”

Total Page:16

File Type:pdf, Size:1020Kb

“The Interplay of Central and Peripheral Circadian Clocks in White Adipose Function and Metabolic Homeostasis” From the Institute of Neurobiology of the University of Lübeck Director: Prof. Dr. rer. nat. H. Oster “The interplay of central and peripheral circadian clocks in white adipose function and metabolic homeostasis” Dissertation for Fulfillment of Requirements for the Doctoral Degree of the University of Lübeck from the Department of Natural Sciences Submitted by Isa Kolbe from Geesthacht, Germany Lübeck 2017 1 First referee: Prof. Dr. rer. nat. Henrik Oster Second referee: Prof. Dr. rer. nat. Stefan Anemüller Date of oral examination: 13. Oktober 2017 Approved for printing. Lübeck, 24. Oktober 2017 2 Declaration Herewith, I confirm that I have written the present PhD thesis independently and with no other sources and aids than quoted. Lübeck, Mai 2017 Isa Kolbe 3 “Zeit. Es gibt Kalender und Uhren, um sie zu messen, aber das will wenig besagen, denn jeder weiß, dass einem eine einzige Stunde wie eine Ewigkeit vorkommen kann, mitunter kann sie aber auch wie in einem Augenblick vergehen – je nachdem, was man in dieser Stunde erlebt. Denn Zeit ist Leben” Michael Ende 4 Contents Summary ........................................................................................................................................ 8 Zusammenfassung ..................................................................................................................... 10 Abbreviations ............................................................................................................................... 12 1 Introduction ............................................................................................................................... 15 1.1 Biological Rhythms .......................................................................................................... 15 1.2 Properties of circadian clocks ........................................................................................ 16 1.3 Molecular mammalian circadian timing system ........................................................... 18 1.4 The master clock in the suprachiasmatic nucleus (SCN) .......................................... 20 1.4.1 SCN anatomy ............................................................................................................ 23 1.4.2 SCN output ................................................................................................................ 25 1.5 Peripheral clocks .............................................................................................................. 27 1.6 Circadian clock hierarchy ................................................................................................ 27 1.6.1 Peripheral clock synchronization ............................................................................ 28 1.7 Circadian clocks and metabolism .................................................................................. 30 1.7.1 Metabolic phenotypes of circadian clock gene mutant mice .............................. 31 1.7.1.1 Bmal1 .................................................................................................................. 32 1.7.1.2 Clock .................................................................................................................... 32 1.7.1.3 Period .................................................................................................................. 32 1.7.1.4 Cryptochrome ..................................................................................................... 33 1.7.1.5 Nuclear receptors .............................................................................................. 33 1.7.2 SCN lesion and metabolism .................................................................................... 33 1.8 Peripheral clocks as metabolic regulators .................................................................... 34 1.9 Adipose tissue .................................................................................................................. 35 1.9.1 WAT innervation........................................................................................................ 35 1.9.2 Fatty acid metabolism .............................................................................................. 36 1.9.2.1 Lipogenesis ........................................................................................................ 36 1.9.2.2 Lipolysis .............................................................................................................. 37 1.9.4. Endocrine function of adipose tissue .................................................................... 38 1.9.4.1 Leptin ................................................................................................................... 39 1.9.5. The adipose clock .................................................................................................... 40 1.9.5.1 AT specific clock disruption and metabolism ................................................ 41 5 1.9.5.2 Regulation of AT by clock regulated hormones ............................................ 42 1.9.5.3 Glucocorticoids .................................................................................................. 43 1.9.5.4 Insulin .................................................................................................................. 43 1.10 Aims of this work ............................................................................................................ 45 2 Material and Methods ............................................................................................................. 46 2.1 Animal experiments ......................................................................................................... 46 2.1.1 Animal housing and breeding ................................................................................. 46 2.1.3 SCN mutant (SCN-KO) mouse breeding .............................................................. 46 2.1.4 Wheel-running analysis ............................................................................................ 47 2.1.5 Energy expenditure .................................................................................................. 48 2.1.6 Temperature measurement ..................................................................................... 48 2.1.7 Tissue collection and blood collection ................................................................... 49 2.1.9 Restricted feeding ..................................................................................................... 49 2.2 Immunobiological experiments ...................................................................................... 49 2.2.1 Leptin enzyme-linked immunosorbent assay (ELISA) ........................................ 49 2.3 Molecular biological experiments .................................................................................. 50 2.3.1 DNA extraction from mouse ear biopsies ............................................................. 50 2.3.2 Genotyping PCRs ..................................................................................................... 51 2.3.2.1 Bmal1-flox genotyping ...................................................................................... 51 2.3.2.2 Synaptotagmin10-Cre genotyping .................................................................. 51 2.3.3 mRNA isolation and cDNA synthesis..................................................................... 53 2.3.4 Quantitative real-time PCR (qPCR) ....................................................................... 54 2.4 Metabolic experiments .................................................................................................... 55 2.4.1 Glucose, insulin and pyruvate tolerance tests ...................................................... 55 2.5 Microarray hybridizations ................................................................................................ 56 2.5.1 Circadian rhythm analysis ....................................................................................... 57 2.5.2 Gene enrichment analysis ....................................................................................... 57 2.6 Magnetic resonance imaging ......................................................................................... 57 2.7 Statistical analysis ............................................................................................................ 58 3 Results ...................................................................................................................................... 59 3.1 Behavior and metabolic phenotype of SCN-KO mice ................................................ 59 3.2 White adipose tissue microarray analysis .................................................................... 66 6 3.3 Influence of SCN and peripheral clocks on body weight and glucose homeostasis ................................................................................................................................................... 77 4 Discussion ................................................................................................................................ 83 4.1 Metabolic phenotype of SCN-KO mice ......................................................................... 84 4.2 Transcriptional alteration in eWAT in the absence of the master pacemaker SCN86 4.3 Body weight regulation and glucose homeostasis in synchronized and unsynchronized conditions ...................................................................................................
Recommended publications
  • Osr1 Maintains Renal Progenitors and Regulates Podocyte Development by Promoting Wnt2ba Through Antagonism of Hand2
    bioRxiv preprint doi: https://doi.org/10.1101/2020.12.21.423845; this version posted December 22, 2020. 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-NC-ND 4.0 International license. osr1 maintains renal progenitors and regulates podocyte development by promoting wnt2ba through antagonism of hand2 Bridgette E. Drummond1, Brooke E. Chambers1, Hannah M. Wesselman1, Marisa N. Ulrich1, Gary F. Gerlach1, Paul T. Kroeger1, Ignaty Leshchiner2, Wolfram Goessling2, and Rebecca A. Wingert1* 1Department of Biological Sciences, Center for Stem Cells and Regenerative Medicine, Center for Zebrafish Research, University of Notre Dame, Notre Dame, 46556, USA 2Brigham and Women's Hospital, Genetics and Gastroenterology Division, Harvard Medical School, Harvard Stem Cell Institute, Boston, MA 02215, USA *Corresponding author Keywords: zebrafish, kidney, nephron, podocyte, segmentation, osr1, wnt2ba, hand2 Abbreviations: chronic kidney disease (CKD), N-ethyl-N-nitrosourea (ENU), days post fertilization (dpf), hours post fertilization (hpf), whole mount in situ hybridization (WISH), flourescent in situ hybridization (FISH), immunofluorescence (IF), immunocytochemistry (ICC), intermediate mesoderm (IM), congenital anomalies of the kidney and urinary tract (CAKUT), amino acid (aa), Zebrafish International Research Center (ZIRC), capped RNA (cRNA), somite stage (ss), wild-type (WT), LIM homeobox 1a (lhx1a), paired box 2a (pax2a), wilms tumor 1a (wt1a), wilms tumor 1b (wt1b), nephrosis 1, congenital Finnish type (nephrin) (nphs1), nephrosis 2, idiopathic, steroid-resistant (podocin)(nphs2), odd-skipped related transcription factor 1 (osr1), cadherin 17 (cdh17), odd-skipped related transcription factor 2 (osr2), wingless-type MMTV integration site family, member 2Ba (wnt2ba) and wingless-type MMTV integration site family, member 2Bb (wnt2bb), heart and neural crest derivatives expressed 2 (hand2), myosin light chain (myl7).
    [Show full text]
  • Molecular Profile of Tumor-Specific CD8+ T Cell Hypofunction in a Transplantable Murine Cancer Model
    Downloaded from http://www.jimmunol.org/ by guest on September 25, 2021 T + is online at: average * The Journal of Immunology , 34 of which you can access for free at: 2016; 197:1477-1488; Prepublished online 1 July from submission to initial decision 4 weeks from acceptance to publication 2016; doi: 10.4049/jimmunol.1600589 http://www.jimmunol.org/content/197/4/1477 Molecular Profile of Tumor-Specific CD8 Cell Hypofunction in a Transplantable Murine Cancer Model Katherine A. Waugh, Sonia M. Leach, Brandon L. Moore, Tullia C. Bruno, Jonathan D. Buhrman and Jill E. Slansky J Immunol cites 95 articles Submit online. Every submission reviewed by practicing scientists ? is published twice each month by Receive free email-alerts when new articles cite this article. Sign up at: http://jimmunol.org/alerts http://jimmunol.org/subscription Submit copyright permission requests at: http://www.aai.org/About/Publications/JI/copyright.html http://www.jimmunol.org/content/suppl/2016/07/01/jimmunol.160058 9.DCSupplemental This article http://www.jimmunol.org/content/197/4/1477.full#ref-list-1 Information about subscribing to The JI No Triage! Fast Publication! Rapid Reviews! 30 days* Why • • • Material References Permissions Email Alerts Subscription Supplementary The Journal of Immunology The American Association of Immunologists, Inc., 1451 Rockville Pike, Suite 650, Rockville, MD 20852 Copyright © 2016 by The American Association of Immunologists, Inc. All rights reserved. Print ISSN: 0022-1767 Online ISSN: 1550-6606. This information is current as of September 25, 2021. The Journal of Immunology Molecular Profile of Tumor-Specific CD8+ T Cell Hypofunction in a Transplantable Murine Cancer Model Katherine A.
    [Show full text]
  • A Computational Approach for Defining a Signature of Β-Cell Golgi Stress in Diabetes Mellitus
    Page 1 of 781 Diabetes A Computational Approach for Defining a Signature of β-Cell Golgi Stress in Diabetes Mellitus Robert N. Bone1,6,7, Olufunmilola Oyebamiji2, Sayali Talware2, Sharmila Selvaraj2, Preethi Krishnan3,6, Farooq Syed1,6,7, Huanmei Wu2, Carmella Evans-Molina 1,3,4,5,6,7,8* Departments of 1Pediatrics, 3Medicine, 4Anatomy, Cell Biology & Physiology, 5Biochemistry & Molecular Biology, the 6Center for Diabetes & Metabolic Diseases, and the 7Herman B. Wells Center for Pediatric Research, Indiana University School of Medicine, Indianapolis, IN 46202; 2Department of BioHealth Informatics, Indiana University-Purdue University Indianapolis, Indianapolis, IN, 46202; 8Roudebush VA Medical Center, Indianapolis, IN 46202. *Corresponding Author(s): Carmella Evans-Molina, MD, PhD ([email protected]) Indiana University School of Medicine, 635 Barnhill Drive, MS 2031A, Indianapolis, IN 46202, Telephone: (317) 274-4145, Fax (317) 274-4107 Running Title: Golgi Stress Response in Diabetes Word Count: 4358 Number of Figures: 6 Keywords: Golgi apparatus stress, Islets, β cell, Type 1 diabetes, Type 2 diabetes 1 Diabetes Publish Ahead of Print, published online August 20, 2020 Diabetes Page 2 of 781 ABSTRACT The Golgi apparatus (GA) is an important site of insulin processing and granule maturation, but whether GA organelle dysfunction and GA stress are present in the diabetic β-cell has not been tested. We utilized an informatics-based approach to develop a transcriptional signature of β-cell GA stress using existing RNA sequencing and microarray datasets generated using human islets from donors with diabetes and islets where type 1(T1D) and type 2 diabetes (T2D) had been modeled ex vivo. To narrow our results to GA-specific genes, we applied a filter set of 1,030 genes accepted as GA associated.
    [Show full text]
  • 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]
  • UNIVERSITY of CALIFORNIA, IRVINE Combinatorial Regulation By
    UNIVERSITY OF CALIFORNIA, IRVINE Combinatorial regulation by maternal transcription factors during activation of the endoderm gene regulatory network DISSERTATION submitted in partial satisfaction of the requirements for the degree of DOCTOR OF PHILOSOPHY in Biological Sciences by Kitt D. Paraiso Dissertation Committee: Professor Ken W.Y. Cho, Chair Associate Professor Olivier Cinquin Professor Thomas Schilling 2018 Chapter 4 © 2017 Elsevier Ltd. © 2018 Kitt D. Paraiso DEDICATION To the incredibly intelligent and talented people, who in one way or another, helped complete this thesis. ii TABLE OF CONTENTS Page LIST OF FIGURES vii LIST OF TABLES ix LIST OF ABBREVIATIONS X ACKNOWLEDGEMENTS xi CURRICULUM VITAE xii ABSTRACT OF THE DISSERTATION xiv CHAPTER 1: Maternal transcription factors during early endoderm formation in 1 Xenopus Transcription factors co-regulate in a cell type-specific manner 2 Otx1 is expressed in a variety of cell lineages 4 Maternal otx1 in the endodermal conteXt 5 Establishment of enhancers by maternal transcription factors 9 Uncovering the endodermal gene regulatory network 12 Zygotic genome activation and temporal control of gene eXpression 14 The role of maternal transcription factors in early development 18 References 19 CHAPTER 2: Assembly of maternal transcription factors initiates the emergence 26 of tissue-specific zygotic cis-regulatory regions Introduction 28 Identification of maternal vegetally-localized transcription factors 31 Vegt and OtX1 combinatorially regulate the endodermal 33 transcriptome iii
    [Show full text]
  • Genome-Wide DNA Methylation Analysis of KRAS Mutant Cell Lines Ben Yi Tew1,5, Joel K
    www.nature.com/scientificreports OPEN Genome-wide DNA methylation analysis of KRAS mutant cell lines Ben Yi Tew1,5, Joel K. Durand2,5, Kirsten L. Bryant2, Tikvah K. Hayes2, Sen Peng3, Nhan L. Tran4, Gerald C. Gooden1, David N. Buckley1, Channing J. Der2, Albert S. Baldwin2 ✉ & Bodour Salhia1 ✉ Oncogenic RAS mutations are associated with DNA methylation changes that alter gene expression to drive cancer. Recent studies suggest that DNA methylation changes may be stochastic in nature, while other groups propose distinct signaling pathways responsible for aberrant methylation. Better understanding of DNA methylation events associated with oncogenic KRAS expression could enhance therapeutic approaches. Here we analyzed the basal CpG methylation of 11 KRAS-mutant and dependent pancreatic cancer cell lines and observed strikingly similar methylation patterns. KRAS knockdown resulted in unique methylation changes with limited overlap between each cell line. In KRAS-mutant Pa16C pancreatic cancer cells, while KRAS knockdown resulted in over 8,000 diferentially methylated (DM) CpGs, treatment with the ERK1/2-selective inhibitor SCH772984 showed less than 40 DM CpGs, suggesting that ERK is not a broadly active driver of KRAS-associated DNA methylation. KRAS G12V overexpression in an isogenic lung model reveals >50,600 DM CpGs compared to non-transformed controls. In lung and pancreatic cells, gene ontology analyses of DM promoters show an enrichment for genes involved in diferentiation and development. Taken all together, KRAS-mediated DNA methylation are stochastic and independent of canonical downstream efector signaling. These epigenetically altered genes associated with KRAS expression could represent potential therapeutic targets in KRAS-driven cancer. Activating KRAS mutations can be found in nearly 25 percent of all cancers1.
    [Show full text]
  • Investigation of the Underlying Hub Genes and Molexular Pathogensis in Gastric Cancer by Integrated Bioinformatic Analyses
    bioRxiv preprint doi: https://doi.org/10.1101/2020.12.20.423656; this version posted December 22, 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. Investigation of the underlying hub genes and molexular pathogensis in gastric cancer by integrated bioinformatic analyses Basavaraj Vastrad1, Chanabasayya Vastrad*2 1. Department of Biochemistry, Basaveshwar College of Pharmacy, Gadag, Karnataka 582103, India. 2. Biostatistics and Bioinformatics, Chanabasava Nilaya, Bharthinagar, Dharwad 580001, Karanataka, India. * Chanabasayya Vastrad [email protected] Ph: +919480073398 Chanabasava Nilaya, Bharthinagar, Dharwad 580001 , Karanataka, India bioRxiv preprint doi: https://doi.org/10.1101/2020.12.20.423656; this version posted December 22, 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. Abstract The high mortality rate of gastric cancer (GC) is in part due to the absence of initial disclosure of its biomarkers. The recognition of important genes associated in GC is therefore recommended to advance clinical prognosis, diagnosis and and treatment outcomes. The current investigation used the microarray dataset GSE113255 RNA seq data from the Gene Expression Omnibus database to diagnose differentially expressed genes (DEGs). Pathway and gene ontology enrichment analyses were performed, and a proteinprotein interaction network, modules, target genes - miRNA regulatory network and target genes - TF regulatory network were constructed and analyzed. Finally, validation of hub genes was performed. The 1008 DEGs identified consisted of 505 up regulated genes and 503 down regulated genes.
    [Show full text]
  • Analysis of Molecular Mechanisms in Intestinal Stem Cells and Colorectal
    JENNY HÖGSTRÖM-STAKEM Analysis of Molecular Pathways in Intestinal Stem Cells and Colorectal Cancer Progression JENNY HÖGSTRÖM-STAKEM Analysis of Molecular Pathways in Recent Publications in this Series 67/2018 Lasse Karhu Computational Analysis of Orexin Receptors and Their Interactions with Natural and Synthetic Ligands 68/2018 Hanna Pitkänen Alterations and Impact of Thrombin Generation and Clot Formation in Solvent/Detergent Plasma, FXIII Deficiency and Lysinuric Protein Intolerance DISSERTATIONES SCHOLAE DOCTORALIS AD SANITATEM INVESTIGANDAM 69/2018 Riikka Havunen UNIVERSITATIS HELSINKIENSIS 87/2018 Enhancing Adoptive Cell Therapy of Solid Tumours with Armed Oncolytic Adenoviruses 70/2018 Patrick Penttilä Novel Biomarkers in Metastatic Renal Cell Carcinoma 71/2018 Jaakko Keinänen Metabolic Changes, Inflammation and Mortality in Psychotic Disorders 72/2018 Heidi Marjonen JENNY HÖGSTRÖM-STAKEM Effects of Prenatal Alcohol Exposure on the Epigenome, Gene Expression and Development 73/2018 Dyah Listyarifah The Role of the Oral Spirochete Treponema denticola in Periodontitis and Orodigestive Analysis of Molecular Pathways in Intestinal Stem Carcinogenesis Cells and Colorectal Cancer Progression 74/2018 Teija-Kaisa Aholaakko Intraoperative Aseptic Practices and Surgical Site Infections in Breast Surgery 75/2018 Cecilia Anna Brunello Tau Pathology: Secretion and Internalization as the Key for Understanding Protein Propagation 76/2018 Niko M. Perttilä Exercise and Falls among Frail Older People – Special Focus on People with Dementia 77/2018
    [Show full text]
  • Supplementary Table S4. FGA Co-Expressed Gene List in LUAD
    Supplementary Table S4. FGA co-expressed gene list in LUAD tumors Symbol R Locus Description FGG 0.919 4q28 fibrinogen gamma chain FGL1 0.635 8p22 fibrinogen-like 1 SLC7A2 0.536 8p22 solute carrier family 7 (cationic amino acid transporter, y+ system), member 2 DUSP4 0.521 8p12-p11 dual specificity phosphatase 4 HAL 0.51 12q22-q24.1histidine ammonia-lyase PDE4D 0.499 5q12 phosphodiesterase 4D, cAMP-specific FURIN 0.497 15q26.1 furin (paired basic amino acid cleaving enzyme) CPS1 0.49 2q35 carbamoyl-phosphate synthase 1, mitochondrial TESC 0.478 12q24.22 tescalcin INHA 0.465 2q35 inhibin, alpha S100P 0.461 4p16 S100 calcium binding protein P VPS37A 0.447 8p22 vacuolar protein sorting 37 homolog A (S. cerevisiae) SLC16A14 0.447 2q36.3 solute carrier family 16, member 14 PPARGC1A 0.443 4p15.1 peroxisome proliferator-activated receptor gamma, coactivator 1 alpha SIK1 0.435 21q22.3 salt-inducible kinase 1 IRS2 0.434 13q34 insulin receptor substrate 2 RND1 0.433 12q12 Rho family GTPase 1 HGD 0.433 3q13.33 homogentisate 1,2-dioxygenase PTP4A1 0.432 6q12 protein tyrosine phosphatase type IVA, member 1 C8orf4 0.428 8p11.2 chromosome 8 open reading frame 4 DDC 0.427 7p12.2 dopa decarboxylase (aromatic L-amino acid decarboxylase) TACC2 0.427 10q26 transforming, acidic coiled-coil containing protein 2 MUC13 0.422 3q21.2 mucin 13, cell surface associated C5 0.412 9q33-q34 complement component 5 NR4A2 0.412 2q22-q23 nuclear receptor subfamily 4, group A, member 2 EYS 0.411 6q12 eyes shut homolog (Drosophila) GPX2 0.406 14q24.1 glutathione peroxidase
    [Show full text]
  • 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]
  • Histone Deacetylases 1 and 2 Regulate the Transcriptional Programs Of
    © 2018. Published by The Company of Biologists Ltd | Development (2018) 145, dev153619. doi:10.1242/dev.153619 STEM CELLS AND REGENERATION RESEARCH ARTICLE Histone deacetylases 1 and 2 regulate the transcriptional programs of nephron progenitors and renal vesicles Hongbing Liu*, Shaowei Chen, Xiao Yao, Yuwen Li, Chao-Hui Chen, Jiao Liu, Zubaida Saifudeen and Samir S. El-Dahr ABSTRACT Six2, a homeodomain transcription factor, is a key factor within Nephron progenitor cells (NPCs) are Six2-positive metanephric the kidney metanephric mesenchyme that maintains the NPC pool mesenchyme cells, which undergo self-renewal and differentiation (Kobayashi et al., 2008; Self et al., 2006). In Six2 null mice, ectopic to give rise to nephrons until the end of nephrogenesis. Histone renal vesicles (the earliest epithelialized forms of nascent nephrons) deacetylases (HDACs) are a group of epigenetic regulators that develop at the onset of nephrogenesis and the progenitor pool is – control cell fate, but their role in balancing NPC renewal and rapidly lost (Self et al., 2006). Many transcriptional regulators differentiation is unknown. Here, we report that NPC-specific such as Osr1, WT1 and Sall1/Mi-2b (Chd4)/nucleosome deletion of Hdac1 and Hdac2 genes in mice results in early remodeling and deacetylase (NuRD), which function to maintain – postnatal lethality owing to renal hypodysplasia and loss of NPCs. the NPC pool display genetic interactions with Six2 (Basta et al., HDAC1/2 interact with the NPC renewal regulators Six2, Osr1 and 2014; Denner and Rauchman, 2013; Hartwig et al., 2010; Kanda Sall1, and are co-bound along with Six2 on the Six2 enhancer. et al., 2014; Xu et al., 2014).
    [Show full text]
  • Downloaded Is Properly from Bioscientifica.Com Cited
    291 Effects of FSH on testicular mRNA transcript levels in the hypogonadal mouse M H Abel, D Baban, S Lee, H M Charlton and P J O’Shaughnessy1 Department of Physiology, Anatomy and Genetics, University of Oxford, Le Gros Clarke Building, Oxford OX1 3QX, UK 1Institute of Comparative Medicine, University of Glasgow Veterinary School, Bearsden Road, Glasgow G61 1QH, UK (Correspondence should be addressed to P J O’Shaughnessy; Email: [email protected]) (D Baban is now at Genomics Group, The Wellcome Trust Centre for Human Genetics, University of Oxford, Roosevelt Drive, Oxford OX3 7BN, UK) Abstract FSH acts through the Sertoli cell to ensure normal testicular development and function. To identify transcriptional mechanisms through which FSH acts in the testis, we have treated gonadotrophin-deficient hypogonadal (hpg) mice with recombinant FSH and measured changes in testicular transcript levels using microarrays and real-time PCR 12, 24 and 72 h after the start of treatment. Approximately 400 transcripts were significantly altered at each time point by FSH treatment. At 12 h, there was a clear increase in the levels of a number of known Sertoli cell transcripts (e.g. Fabp5, Lgals1, Tesc, Scara5, Aqp5). Additionally, levels of Leydig cell transcripts were also markedly increased (e.g. Ren1, Cyp17a1, Akr1b7, Star, Nr4a1). This was associated with a small but significant rise in testosterone at 24 and 72 h. At 24 h, androgen-dependent Sertoli cell transcripts were up-regulated (e.g. Rhox5, Drd4, Spinlw1, Tubb3 and Tsx) and this trend continued up to 72 h. By contrast with the somatic cells, only five germ cell transcripts (Dkkl1, Hdc, Pou5f1, Zfp541 and 1700021K02Rik) were altered by FSH within the time-course of the experiment.
    [Show full text]