Dual Functions for Insulinoma-Associated 1 in Retinal Development

Total Page:16

File Type:pdf, Size:1020Kb

Dual Functions for Insulinoma-Associated 1 in Retinal Development University of Kentucky UKnowledge Theses and Dissertations--Biology Biology 2015 DUAL FUNCTIONS FOR INSULINOMA-ASSOCIATED 1 IN RETINAL DEVELOPMENT Marie A. Forbes-Osborne University of Kentucky, [email protected] Right click to open a feedback form in a new tab to let us know how this document benefits ou.y Recommended Citation Forbes-Osborne, Marie A., "DUAL FUNCTIONS FOR INSULINOMA-ASSOCIATED 1 IN RETINAL DEVELOPMENT" (2015). Theses and Dissertations--Biology. 31. https://uknowledge.uky.edu/biology_etds/31 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. I agree that the document mentioned above may be made available immediately for worldwide access unless an embargo applies. I retain all other ownership rights to the copyright of my work. I also retain the right to use in future works (such as articles or books) all or part of my work. I understand that I am free to register the copyright to my work. REVIEW, APPROVAL AND ACCEPTANCE The document mentioned above has been reviewed and accepted by the student’s advisor, on behalf of the advisory committee, and by the Director of Graduate Studies (DGS), on behalf of the program; we verify that this is the final, approved version of the student’s thesis including all changes required by the advisory committee. The undersigned agree to abide by the statements above. Marie A. Forbes-Osborne, Student Dr. Ann C. Morris, Major Professor Dr. David F. Westneat, Director of Graduate Studies DUAL FUNCTIONS FOR INSULINOMA-ASSOCIATED 1 IN RETINAL DEVELOPMENT __________________________ DISSERTATION __________________________ A dissertation submitted in partial fulfillment of the requirements for the degree of Doctor of Philosophy in the College of Arts and Sciences at the University of Kentucky By Marie A. Forbes-Osborne Lexington, Kentucky Director: Ann C. Morris, Associate Professor of Biology Lexington, Kentucky 2015 Copyright © Marie A. Forbes-Osborne 2015 ABSTRACT OF DISSERTATION DUAL FUNCTIONS FOR INSULINOMA-ASSOCIATED 1 IN RETINAL DEVELOPMENT Proper visual system function requires tightly controlled proliferation of a pool of relatively homogeneous retinal progenitor cells, followed by the stepwise specification and differentiation of multiple distinct cell types. These retinal cells, both neuronal and glial, must be generated in the correct numbers, and the correct laminar location to permit the formation of synaptic connections between individual cell types. After synapses are made, constant signaling is required as part of normal retinal function, and to maintain cellular identity and connectivity. These processes rely on both extrinsic and intrinsic signaling, with regulation of gene expression by cascades of transcription factors having a key role. While considerable work has been done to identify key regulators of retinal development, maturation, and homeostasis, many factors remain unidentified or poorly characterized, either at large or within the retina. One such factor is Insulinoma-associated 1 (Insm1). Known to function in endocrine cell, sympathetic and monoaminergic neuron, and olfactory epithelial cell differentiation and maturation, Insm1 is also a regulator of cell cycle progression in the adrenal system and cerebral cortex. Although Insm1 was previously considered a transcriptional regulator, recently, non- nuclear functions have also been identified. However, the retinal function of Insm1 remained a mystery. To determine the role of Insm1 in retinal development, I characterized the retinal expression pattern of Insm1, as well as the effect of perturbation of Insm1 expression levels at both the cellular and molecular level. Chapter 1 of this dissertation provides an overview of the retina and its development, vision and retinal degenerative diseases, and a review of Insm1 expression and function in tissues outside the retina. Chapter 2 presents data generated from the knockdown of the retinal-expressed zebrafish co-ortholog of Insm1, insm1a, which demonstrated a requirement for insm1a in proper differentiation of photoreceptor cells. Additionally, these experiments showed a cell cycle regulatory function for insm1a in retinal development. Characterization of a zebrafish insm1a mutant and functional examination of insm1a truncation variants is discussed in Chapter 3. Chapter 4 presents data from an RNA-Seq analysis of wild-type and Insm1 knockout mouse retinas at two developmental time points, and details transcriptional changes during retinal development in the absence of Insm1. Finally, Chapter 5 discusses the conclusions from the data generated for this dissertation, additional studies identified as the result of this work, and the implications of these results on our understanding of retinal development. KEYWORDS: Retina, Photoreceptor, Insm1, Development, Zebrafish, Mouse Marie A. Forbes-Osborne Student’s Signature December 10, 2015 Date DUAL FUNCTIONS FOR INSULINOMA-ASSOCIATED 1 IN RETINAL DEVELOPMENT By Marie A. Forbes-Osborne Ann C. Morris Director of Dissertation David F. Westneat Director of Graduate Studies December 10, 2015 Date For MB Really, Really. ACKOWLEDGEMENTS “Hard work spotlights the character of people: some turn up their sleeves, some turn up their noses, and some don't turn up at all.” –Sam Ewing Throughout life (and especially during graduate school) this quote has inspired me, and I have committed to always being the type person who turns up their sleeves. However, hard work seldom occurs in a vacuum, and good company can make difficult tasks far easier. Though this dissertation bears only my name, it is built on the hard work and knowledge of innumerable others. First, I owe more than I can ever express to my mother, Antoinette Forbes. She taught me the value of hard work and dedication from childhood. She made me a voracious learner, and more importantly, a limitless questioner. She never stifled my curiosity and showed me the value and importance of questions without answers. For this and so much more, I am forever grateful. Second, I would like to thank my doctoral advisor, Dr. Ann Morris. Far beyond simply giving knowledge, Ann gave me a toolbox to solve nearly any problem that presented itself. She also gave me the freedom to explore, experiment and make (not mention learn from) mistakes, all while being ready to swoop in and save the day if I couldn’t figure it out on my own. This made me a better scientist, and gave me the opportunity to learn to be independent, without fear of failing. She also showed me that excellent science must be accompanied by excellent writing and engaging presentation skills. Her continued support has been essential to my growth and success in the lab and in life. I would also like to express my gratitude to the members of my committee. Dr. Pete Mirabito, who offered me unbelievable support and guidance from the lab to the classroom. His emphasis on understanding why and how have forever changed the way that I look at and solve problems. Dr. Randal Voss, whose questions and observations often forced me outside my comfort zone. Without a doubt, this made me a more thorough researcher and a better student of science and the world. Dr. Liz Debski, who always held me to exceptionally high standards, and showed me the importance in being certain of the underlying facts and the best ways to present them. Lastly, Dr. Jayakrishna Ambati, who helped me understand the importance of making research work for the betterment of society, and the value of using basic science and simple techniques to answer real world difficult questions. Their assistance has been invaluable to me throughout. My initial research experiences came from Dr. Rebecca Kellum. Without her support and assistance I would never have found the passion for research. I will be forever grateful to her for all that she has done for me. Day to day, I have been fortunate to have amazing lab mates and department colleagues, who made the daily grind more enjoyable. Dr. Lakshmi Pillai-Kastoori, my batch- mate and confidant. I am forever thankful for her support and friendship. Wen Wen, who attacks every obstacle with unending enthusiasm. Every task is possible when untaken so positively. Stephen Wilson, who showed me the value of taking every opportunity to enjoy the high points, while never letting the low points destroy you. Cagney (CC) Coomer, who has brought a new vibrancy to the lab in the last year, and has reminded me how important it is to pass on what we have learned. Sara Perkins, our fabulous technician, who kept things together for all of us. Whether supporting us at the bench or keeping us in line, none of this work would be possible without her. She has also become one of my best friends during this time, and has made my life better in more ways than I can ever express. Michelle Geidt, who has been a great friend, always ready to shore up flagging energy any way possible. Melissa Keinath and Stephanie Bryant for a listening ear and copious help when the science got ahead of my abilities. Thank you all.
Recommended publications
  • The Function of the Zinc Finger Transcription Factor Insm1 in Neuronal Progenitors Madeleine Julie Larrosa
    The function of the zinc finger transcription factor Insm1 in neuronal progenitors Inaugural-Dissertation To obtain the academic degree Doctor rerum naturalium (Dr. rer. nat.) Submitted to the Department of Biology, Chemistry and Pharmacy of Freie Universität Berlin By Madeleine Julie Larrosa From Paris 2019 This work was carried out at the Max Delbrück Centrum for Molecular Medicine in Berlin from November 2015 to October 2019 under the supervision of Prof. Dr. Carmen Birchmeier and Prof. Dr. Holger Gerhardt. 1st Reviewer: Prof. Dr. Holger Gerhardt 2nd Reviewer: Prof. Dr. Stephan Sigrist Date of PhD Defense: This doctoral thesis is dedicated to my mother Juliette Ban, who offered unconditional love and endless devotion, and taught me to work hard for the things I aspire to achieve. I also dedicate this work to Mohammed Marine, who has been a constant source of support and encouragement, and without whom I would not have completed this doctoral thesis. In loving memory of my friend Lorelei Arbeille, who taught me to never give up through her incredible strength and perseverance. Statement of contribution I confirm that the work presented in this doctoral thesis is my own and that all information derived from other sources is indicated. Madlen Sohn, technician in the group of Prof. Dr. Wei Chen at the MDC, performed the sequencing of the immunoprecipitated chromatin and the transcriptome for the ChIP-seq and RNA-seq experiments, respectively. Dr. Mahmoud Ibrahim and Dr. Scott Lacadie, bioinformaticians in the group of Prof. Dr. Uwe Ohler at the MDC, analyzed the ChIP-seq data and Dr.
    [Show full text]
  • Table S1. List of Proteins in the BAHD1 Interactome
    Table S1. List of proteins in the BAHD1 interactome BAHD1 nuclear partners found in this work yeast two-hybrid screen Name Description Function Reference (a) Chromatin adapters HP1α (CBX5) chromobox homolog 5 (HP1 alpha) Binds histone H3 methylated on lysine 9 and chromatin-associated proteins (20-23) HP1β (CBX1) chromobox homolog 1 (HP1 beta) Binds histone H3 methylated on lysine 9 and chromatin-associated proteins HP1γ (CBX3) chromobox homolog 3 (HP1 gamma) Binds histone H3 methylated on lysine 9 and chromatin-associated proteins MBD1 methyl-CpG binding domain protein 1 Binds methylated CpG dinucleotide and chromatin-associated proteins (22, 24-26) Chromatin modification enzymes CHD1 chromodomain helicase DNA binding protein 1 ATP-dependent chromatin remodeling activity (27-28) HDAC5 histone deacetylase 5 Histone deacetylase activity (23,29,30) SETDB1 (ESET;KMT1E) SET domain, bifurcated 1 Histone-lysine N-methyltransferase activity (31-34) Transcription factors GTF3C2 general transcription factor IIIC, polypeptide 2, beta 110kDa Required for RNA polymerase III-mediated transcription HEYL (Hey3) hairy/enhancer-of-split related with YRPW motif-like DNA-binding transcription factor with basic helix-loop-helix domain (35) KLF10 (TIEG1) Kruppel-like factor 10 DNA-binding transcription factor with C2H2 zinc finger domain (36) NR2F1 (COUP-TFI) nuclear receptor subfamily 2, group F, member 1 DNA-binding transcription factor with C4 type zinc finger domain (ligand-regulated) (36) PEG3 paternally expressed 3 DNA-binding transcription factor with
    [Show full text]
  • The Mineralocorticoid Receptor Leads to Increased Expression of EGFR
    www.nature.com/scientificreports OPEN The mineralocorticoid receptor leads to increased expression of EGFR and T‑type calcium channels that support HL‑1 cell hypertrophy Katharina Stroedecke1,2, Sandra Meinel1,2, Fritz Markwardt1, Udo Kloeckner1, Nicole Straetz1, Katja Quarch1, Barbara Schreier1, Michael Kopf1, Michael Gekle1 & Claudia Grossmann1* The EGF receptor (EGFR) has been extensively studied in tumor biology and recently a role in cardiovascular pathophysiology was suggested. The mineralocorticoid receptor (MR) is an important efector of the renin–angiotensin–aldosterone‑system and elicits pathophysiological efects in the cardiovascular system; however, the underlying molecular mechanisms are unclear. Our aim was to investigate the importance of EGFR for MR‑mediated cardiovascular pathophysiology because MR is known to induce EGFR expression. We identifed a SNP within the EGFR promoter that modulates MR‑induced EGFR expression. In RNA‑sequencing and qPCR experiments in heart tissue of EGFR KO and WT mice, changes in EGFR abundance led to diferential expression of cardiac ion channels, especially of the T‑type calcium channel CACNA1H. Accordingly, CACNA1H expression was increased in WT mice after in vivo MR activation by aldosterone but not in respective EGFR KO mice. Aldosterone‑ and EGF‑responsiveness of CACNA1H expression was confrmed in HL‑1 cells by Western blot and by measuring peak current density of T‑type calcium channels. Aldosterone‑induced CACNA1H protein expression could be abrogated by the EGFR inhibitor AG1478. Furthermore, inhibition of T‑type calcium channels with mibefradil or ML218 reduced diameter, volume and BNP levels in HL‑1 cells. In conclusion the MR regulates EGFR and CACNA1H expression, which has an efect on HL‑1 cell diameter, and the extent of this regulation seems to depend on the SNP‑216 (G/T) genotype.
    [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]
  • CCAAT/Enhancer Binding Protein Epsilon) Thomas Burmeister Charite, Med
    Atlas of Genetics and Cytogenetics in Oncology and Haematology OPEN ACCESS JOURNAL INIST-CNRS Gene Section Short Communication CEBPE (CCAAT/enhancer binding protein epsilon) Thomas Burmeister Charite, Med. Klinik fur Hamatologie, Onkologie und Tumorimmunologie, Hindenburgdamm 30, 12200 Berlin, Germany; [email protected] Published in Atlas Database: March 2017 Online updated version : http://AtlasGeneticsOncology.org/Genes/CEBPEID42984ch14q11.html Printable original version : http://documents.irevues.inist.fr/bitstream/handle/2042/69005/03-2017-CEBPEID42984ch14q11.pdf DOI: 10.4267/2042/69005 This work is licensed under a Creative Commons Attribution-Noncommercial-No Derivative Works 2.0 France Licence. © 2017 Atlas of Genetics and Cytogenetics in Oncology and Haematology alternative 3-exon-organization of the human Abstract CEBPE gene (Figure 1b). However, exon 1, as described by Yamanaka et al. contains a frameshift Review on CEBPE, with data on DNA, on the according to the GRCh38.p7 NCBI assembly. protein encoded, and where the gene is implicated. Transcription Keywords CEBPE; Transcription factor; Neutrophil specific Various transcripts have been reported, resulting in granule deficiency; Acute lymphoblastic leukemia; four protein isoforms (Lekstrom-Himes 2001, Translocation. Yamanaka 1997; Figure 1c). All transcripts share a common 3' end. Identity Protein Other names: CRP1 Description HGNC (Hugo): CEBPE CEBPE is a member of the CCAAT/enhancer- Location: 14q11.2 binding protein (C/EBP) family, which also Location (base pair) includes CEBPA, CEBPB, CEBPG, CEBPD and Starts at 23117306 and ends at 23119611 bp from CEBPZ (Ramji & Foka; 2002). A common pter (according to GRCh38.p7 Annotation Release structural feature of the C/EBP proteins is the 108, May 5 2016) presence of a highly conserved 55-65 amino acid sequence at the C-terminus which encodes a basic DNA/RNA leucine zipper motif (bZIP domain) that functions as a dimerization domain.
    [Show full text]
  • Beta-Arrestin-Mediated Signaling in the Heart
    SPECIAL ARTICLE Circ J 2008; 72: 1725–1729 Beta-Arrestin-Mediated Signaling in the Heart Priyesh A. Patel, BS; Douglas G. Tilley, PhD*; Howard A. Rockman, MD*,** Beta-arrestin is a multifunctional adapter protein well known for its role in G-protein-coupled receptor (GPCR) desensitization. Exciting new evidence indicates thatβ-arrestin is also a signaling molecule capable of initiating its own G-protein-independent signaling at GPCRs. One of the best-studiedβ-arrestin signaling pathways is the one involvingβ-arrestin-dependent activation of a mitogen-activated protein kinase cascade, the extracellular regulated kinase (ERK). ERK signaling, which is classically activated by agonist stimulation of the epidermal growth factor receptor (EGFR), can be activated by a number of GPCRs in aβ-arrestin-dependent manner. Recent work in animal models of heart failure suggests thatβ-arrestin-dependent activation of EGFR/ERK signaling by theβ-1-adrenergic receptor, and possibly the angiotensin II Type 1A receptor, are cardioprotective. Hence, a new model of signaling at cardiac GPCRs has emerged and implicates classical G-protein-mediated signaling with promoting harmful remodeling in heart failure, while concurrently linkingβ-arrestin-dependent, G-protein-inde- pendent signaling with cardioprotective effects. Based on this paradigm, a new class of drugs could be identified, termed “biased ligands”, which simultaneously block harmful G-protein signaling, while also promoting cardio- protectiveβ-arrestin-dependent signaling, leading to a potential breakthrough
    [Show full text]
  • BMC Evolutionary Biology Biomed Central
    BMC Evolutionary Biology BioMed Central Research article Open Access On the origins of arrestin and rhodopsin Carlos E Alvarez1,2,3 Address: 1Center for Molecular and Human Genetics, The Research Institute at Nationwide Children's Hospital, Columbus, OH, 43205, USA, 2Department of Pediatrics, The Ohio State University College of Medicine, Columbus, OH, 43210, USA and 3Novartis Institutes of BioMedical Research, CH-4002 Basel, Switzerland Email: Carlos E Alvarez - [email protected] Published: 29 July 2008 Received: 11 January 2008 Accepted: 29 July 2008 BMC Evolutionary Biology 2008, 8:222 doi:10.1186/1471-2148-8-222 This article is available from: http://www.biomedcentral.com/1471-2148/8/222 © 2008 Alvarez; licensee BioMed Central Ltd. This is an Open Access article distributed under the terms of the Creative Commons Attribution License (http://creativecommons.org/licenses/by/2.0), which permits unrestricted use, distribution, and reproduction in any medium, provided the original work is properly cited. Abstract Background: G protein coupled receptors (GPCRs) are the most numerous proteins in mammalian genomes, and the most common targets of clinical drugs. However, their evolution remains enigmatic. GPCRs are intimately associated with trimeric G proteins, G protein receptor kinases, and arrestins. We conducted phylogenetic studies to reconstruct the history of arrestins. Those findings, in turn, led us to investigate the origin of the photosensory GPCR rhodopsin. Results: We found that the arrestin clan is comprised of the Spo0M protein family in archaea and bacteria, and the arrestin and Vps26 families in eukaryotes. The previously known animal arrestins are members of the visual/beta subfamily, which branched from the founding "alpha" arrestins relatively recently.
    [Show full text]
  • Histone-Binding of DPF2 Mediates Its Repressive Role in Myeloid Differentiation
    Histone-binding of DPF2 mediates its repressive role in myeloid differentiation Ferdinand M. Hubera,1, Sarah M. Greenblattb,1, Andrew M. Davenporta,1, Concepcion Martinezb,YeXub,LyP.Vuc, Stephen D. Nimerb,2, and André Hoelza,2 aDivision of Chemistry and Chemical Engineering, California Institute of Technology, Pasadena, CA 91125; bSylvester Comprehensive Cancer Center, University of Miami Miller School of Medicine, Miami, FL 33136; and cMolecular Pharmacology Program, Memorial Sloan Kettering Cancer Center, New York, NY 10065 Edited by Douglas C. Rees, Howard Hughes Medical Institute, California Institute of Technology, Pasadena, CA, and approved April 26, 2017 (received for review January 6, 2017) Double plant homeodomain finger 2 (DPF2) is a highly evolution- RUNX1 form a methylation-dependent repressive complex in arily conserved member of the d4 protein family that is ubiqui- AML, although it remains unclear whether the two proteins bind tously expressed in human tissues and was recently shown to each other directly or act concertedly as part of a larger complex. inhibit the myeloid differentiation of hematopoietic stem/progen- Here, we present the crystal structure of the human DPF2 itor and acute myelogenous leukemia cells. Here, we present the tandem PHD finger domain at a 1.6-Å resolution. We demon- crystal structure of the tandem plant homeodomain finger domain strate that the DPF2 tandem PHD finger domain binds acetylated of human DPF2 at 1.6-Å resolution. We show that DPF2 interacts H3 and H4 histone tails, identify the primary determinants of with the acetylated tails of both histones 3 and 4 via bipartite histone recognition, and confirm these interactions in vivo.
    [Show full text]
  • Mediator of DNA Damage Checkpoint 1 (MDC1) Is a Novel Estrogen Receptor Co-Regulator in Invasive 6 Lobular Carcinoma of the Breast 7 8 Evelyn K
    bioRxiv preprint doi: https://doi.org/10.1101/2020.12.16.423142; this version posted December 16, 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 4.0 International license. 1 Running Title: MDC1 co-regulates ER in ILC 2 3 Research article 4 5 Mediator of DNA damage checkpoint 1 (MDC1) is a novel estrogen receptor co-regulator in invasive 6 lobular carcinoma of the breast 7 8 Evelyn K. Bordeaux1+, Joseph L. Sottnik1+, Sanjana Mehrotra1, Sarah E. Ferrara2, Andrew E. Goodspeed2,3, James 9 C. Costello2,3, Matthew J. Sikora1 10 11 +EKB and JLS contributed equally to this project. 12 13 Affiliations 14 1Dept. of Pathology, University of Colorado Anschutz Medical Campus 15 2Biostatistics and Bioinformatics Shared Resource, University of Colorado Comprehensive Cancer Center 16 3Dept. of Pharmacology, University of Colorado Anschutz Medical Campus 17 18 Corresponding author 19 Matthew J. Sikora, PhD.; Mail Stop 8104, Research Complex 1 South, Room 5117, 12801 E. 17th Ave.; Aurora, 20 CO 80045. Tel: (303)724-4301; Fax: (303)724-3712; email: [email protected]. Twitter: 21 @mjsikora 22 23 Authors' contributions 24 MJS conceived of the project. MJS, EKB, and JLS designed and performed experiments. JLS developed models 25 for the project. EKB, JLS, SM, and AEG contributed to data analysis and interpretation. SEF, AEG, and JCC 26 developed and performed informatics analyses. MJS wrote the draft manuscript; all authors read and revised the 27 manuscript and have read and approved of this version of the manuscript.
    [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]
  • Integrating Single-Step GWAS and Bipartite Networks Reconstruction Provides Novel Insights Into Yearling Weight and Carcass Traits in Hanwoo Beef Cattle
    animals Article Integrating Single-Step GWAS and Bipartite Networks Reconstruction Provides Novel Insights into Yearling Weight and Carcass Traits in Hanwoo Beef Cattle Masoumeh Naserkheil 1 , Abolfazl Bahrami 1 , Deukhwan Lee 2,* and Hossein Mehrban 3 1 Department of Animal Science, University College of Agriculture and Natural Resources, University of Tehran, Karaj 77871-31587, Iran; [email protected] (M.N.); [email protected] (A.B.) 2 Department of Animal Life and Environment Sciences, Hankyong National University, Jungang-ro 327, Anseong-si, Gyeonggi-do 17579, Korea 3 Department of Animal Science, Shahrekord University, Shahrekord 88186-34141, Iran; [email protected] * Correspondence: [email protected]; Tel.: +82-31-670-5091 Received: 25 August 2020; Accepted: 6 October 2020; Published: 9 October 2020 Simple Summary: Hanwoo is an indigenous cattle breed in Korea and popular for meat production owing to its rapid growth and high-quality meat. Its yearling weight and carcass traits (backfat thickness, carcass weight, eye muscle area, and marbling score) are economically important for the selection of young and proven bulls. In recent decades, the advent of high throughput genotyping technologies has made it possible to perform genome-wide association studies (GWAS) for the detection of genomic regions associated with traits of economic interest in different species. In this study, we conducted a weighted single-step genome-wide association study which combines all genotypes, phenotypes and pedigree data in one step (ssGBLUP). It allows for the use of all SNPs simultaneously along with all phenotypes from genotyped and ungenotyped animals. Our results revealed 33 relevant genomic regions related to the traits of interest.
    [Show full text]
  • Yan Et Al. Supplementary Material
    SUPPLEMENTARY MATERIAL FOR: CELL ATLAS OF THE HUMAN FOVEA AND PERIPHERAL RETINA Wenjun Yan*, Yi-Rong Peng*, Tavé van Zyl*, Aviv Regev, Karthik Shekhar, Dejan Juric, and Joshua R, Sanes^ *Co-First authors ^Author for correspondence, [email protected] Figure S1 tSNE visualization showing contributions to cell types by batch for photoreceptors (a), horizontal cells (b), bipolar cells (c), amacrine cells (d), retinal ganglion cells (e) and non-neuronal cells (f). Each dot represents one cell. Colors distinguish retina samples. Source of each sample is shown in Table S1. Overall, batch eFFects were minimal. Figure S2 Violin and superimposed box plots showing expression of OPN4 in RGC clusters Figure S3 Heat maps showing expression patterns of disease genes by cell classes in the Fovea and periphery. Only genes expressed by more than 20% of cells in any individual class in either Fovea or peripheral cells are plotted. Table S1 Information on donors from whom retinal cells were obtained for scRNA-seq proFiling. Table S2 Publications reporting single cell or single nucleus profiling on cells from human retina. 1 Figure S1 a b c PR HC BP H1 H2F1 H2F2 H3 tSNE1 tSNE1 H4 tSNE1 H5 H9 H11 tSNE2 tSNE2 tSNE2 e AC f RGC g Non-neuronal tSNE1 tSNE1 tSNE1 tSNE2 tSNE2 tSNE2 Figure S2 OPN4 4 2 log Expression 0 MG-ON MG-OFF PG-OFF PG-ON hRGC5 hRGC6 hRGC7 hRGC8 hRGC9 hRGC10 hRGC11 hRGC12 Figure S3 Fovea Fovea Peripheral Peripheral Rods Cones BP HC AC RGC Muller Astro MicG Endo Rods Cones BP HC AC RGC Muller Astro MicG Endo ARL13B MAPK8IP3 EXOC6 LSM4 Expression
    [Show full text]