Early During Myelomagenesis Alterations in DNA Methylation That Occur Myeloma Is Characterized by Stage-Specific

Total Page:16

File Type:pdf, Size:1020Kb

Early During Myelomagenesis Alterations in DNA Methylation That Occur Myeloma Is Characterized by Stage-Specific The Journal of Immunology Myeloma Is Characterized by Stage-Specific Alterations in DNA Methylation That Occur Early during Myelomagenesis Christoph J. Heuck,* Jayesh Mehta,† Tushar Bhagat,* Krishna Gundabolu,* Yiting Yu,* Shahper Khan,‡ Grigoris Chrysofakis,* Carolina Schinke,* Joseph Tariman,† Eric Vickrey,† Natalie Pulliam,† Sangeeta Nischal,* Li Zhou,* Sanchari Bhattacharyya,* Richard Meagher,† Caroline Hu,* Shahina Maqbool,* Masako Suzuki,* Samir Parekh,* Frederic Reu,‡ Ulrich Steidl,* John Greally,* Amit Verma,* and Seema B. Singhal† Epigenetic changes play important roles in carcinogenesis and influence initial steps in neoplastic transformation by altering ge- nome stability and regulating gene expression. To characterize epigenomic changes during the transformation of normal plasma cells to myeloma, we modified the HpaII tiny fragment enrichment by ligation–mediated PCR assay to work with small numbers of purified primary marrow plasma cells. The nano-HpaII tiny fragment enrichment by ligation–mediated PCR assay was used to analyze the methylome of CD138+ cells from 56 subjects representing premalignant (monoclonal gammopathy of uncertain significance), early, and advanced stages of myeloma, as well as healthy controls. Plasma cells from premalignant and early stages of myeloma were characterized by striking, widespread hypomethylation. Gene-specific hypermethylation was seen to occur in the advanced stages, and cell lines representative of relapsed cases were found to be sensitive to decitabine. Aberrant demethylation in monoclonal gammopathy of uncertain significance occurred primarily in CpG islands, whereas differentially methylated loci in cases of myeloma occurred predominantly outside of CpG islands and affected distinct sets of gene pathways, demonstrating qualitative epigenetic differences between premalignant and malignant stages. Examination of the methylation machinery revealed that the methyltransferase, DNMT3A, was aberrantly hypermethylated and underexpressed, but not mutated in mye- loma. DNMT3A underexpression was also associated with adverse overall survival in a large cohort of patients, providing insights into genesis of hypomethylation in myeloma. These results demonstrate widespread, stage-specific epigenetic changes during myelomagenesis and suggest that early demethylation can be a potential contributor to genome instability seen in myeloma. We also identify DNMT3A expression as a novel prognostic biomarker and suggest that relapsed cases can be therapeutically targeted by hypomethylating agents. The Journal of Immunology, 2013, 190: 2966–2975. espite therapeutic advances, multiple myeloma (MM) re- the development of a risk model that remains robust even in the age mains incurable and needs newer insights into the path- of newer therapies (1). The use of gene expression profiling by D ogenic mechanisms that cause this disease. Gene expres- different groups has also led to the identification of distinct mo- sion profiling has been used extensively in myeloma and has led to lecular subgroups that show defined clinical features (2, 3). How- ever, pathogenic mechanisms driving these differences in gene expression have not been well described, especially for early stages *Albert Einstein College of Medicine, Bronx, NY 10461; †Northwestern University, in myelomagenesis (4). Feinberg School of Medicine, Chicago, IL 60611; and ‡Cleveland Clinic, Cleveland, Recent studies of the epigenome and in particular the methylome OH 44195 have shown that cancer is characterized by widespread epigenetic Received for publication September 7, 2012. Accepted for publication January 9, changes. These changes lead to altered gene expression that can 2013. result in activation of oncogenic pathways. Furthermore, methyl- This work was supported by National Institutes of Health Immunooncology Training ome profiling has shown greater prognostic ability than gene ex- Program Grant T32 CA009173, Gabrielle’s Angel Foundation, the Leukemia and Lymphoma Society, the American Cancer Society, the Department of Defense, and pression profiling in acute myeloid leukemia (AML) and has led to a Partnership for Cures grant. identification of newer molecular subgroups with differences in The methylation data presented in this article have been deposited in the National overall survival (5). We have shown that methylome profiling is Institutes of Health Gene Expression Omnibus (National Center for Biotechnology able to identify changes that occur early during carcinogenesis in Information, http://www.ncbi.nlm.nih.gov/geo/) under accession number GSE 43860. solid tumors such as esophageal cancer (6). These studies have Address correspondence and reprint requests to Dr. Amit Verma, Dr. Christoph J. Heuck, and Dr. Seema B. Singhal, Albert Einstein College of Medicine, Bronx, NY been conducted with the use of the HpaII tiny fragment enrich- 10461 (A.V. and C.J.H.) and Feinberg School of Medicine, Northwestern University, ment by ligation–mediated PCR (HELP) assay, which is a ge- Chicago, IL 60611 (S.B.S.). E-mail addresses: [email protected] (A.V.), nome-wide assay that provides a reproducible analysis of the [email protected] (C.J.H.), and [email protected] (S.B.S.) methylome that is not biased toward CpG islands (7). Analyzing The online version of this article contains supplemental material. the methylome of myeloma can help identify changes that can Abbreviations used in this article: AML, acute myeloid leukemia; HAF, HpaII-amplifi- able fragment; HELP, HpaII tiny fragment enrichment by ligation–mediated PCR; MM, define disease subsets and lead to identification of newer thera- multiple myeloma; MGUS, monoclonal gammopathy of uncertain significance; peutic targets. Recent sequencing studies in myeloma have also NEWMM, newly diagnosed myeloma; NL, normal; REL, relapsed myeloma; REM, revealed mutations in enzymes that are involved in epigenetic complete remission; SMM, smoldering myeloma; TF, transcription factor. machinery, again reinforcing the need to study the epigenetic Copyright Ó 2013 by The American Association of Immunologists, Inc. 0022-1767/13/$16.00 alterations in this disease (8). We have conducted a genome-wide www.jimmunol.org/cgi/doi/10.4049/jimmunol.1202493 The Journal of Immunology 2967 analysis of changes in DNA methylation in monoclonal gamm- tative for methylation and analyzed as a continuous variable. For most loci, opathy of uncertain significance (MGUS), newly diagnosed MM, each fragment was categorized as either methylated, if the centered log , as well as relapsed MM, and compared them with normal plasma HpaII/MspI ratio was generally 0, or hypomethylated, if, in contrast, the log ratio was .0. cell controls. We used a modification of the HELP assay (nano- HELP) that was developed to work with low amounts of DNA to Quantitative DNA methylation analysis by MassArray interrogate the methylome of sorted CD138+ cells from patient Epityping samples. We report that widespread alterations in DNA methyla- Validation of the findings of the HELP assay was carried out by MALDI- tion are seen in myeloma and have the power to discriminate TOF mass spectrometry using EpiTyper by MassArray (Sequenom, CA) on between MGUS and new/relapsed cases. We report that hypo- bisulfite-converted DNA, as previously described (6). Validation was done methylation is the predominant early change during myeloma- on all samples with sufficient available DNA. MassARRAY primers were designed to cover the flanking HpaII sites for a given HAF, as well as for genesis that is gradually transformed to hypermethylation in any other HpaII sites found up to 2000 bp upstream of the downstream site relapsed cases, thus providing the epigenetic basis for the differ- and up to 2000 bp downstream of the upstream site, to cover all possible entiation between these different stages of myeloma. alternative sites of digestion within our range of PCR amplification. Microarray data analysis Materials and Methods Patient samples Unsupervised clustering of HELP data by hierarchical clustering was performed using the statistical software R version 2.6.2. A two-sample t test Bone marrow aspirates were obtained with signed informed consent from was used for each gene to summarize methylation differences between 11 patients with MGUS, 4 patients with smoldering myeloma (SMM), groups. Genes were ranked on the basis of this test statistic, and a set of top 13 patients with newly diagnosed myeloma (NEWMM), 16 patients with differentially methylated genes with an observed log fold change of .1 relapsed myeloma (REL), including 2 patients with serial samples, and 2 between group means was identified. Genes were further grouped patients in clinical complete remission (REM) who are followed at the according to the direction of the methylation change (hypomethylated myeloma clinic at the Robert H. Lurie Cancer Center at Northwestern versus hypermethylated), and the relative frequencies of these changes University. All specimens underwent CD138+ microbead selection (Mil- were computed among the top candidates to explore global methylation tenyi Biotec). Purity was confirmed by flow cytometry staining for CD38 patterns. Validation with MassArray showed good correlation with the data and CD45. Samples with a purity ,90% by flow cytometry were not used generated by the HELP assay. MassArray analysis validated significant for this study. Eight samples from normal donors without known malig- quantitative differences in methylation for differentially methylated genes nancies were obtained commercially
Recommended publications
  • 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]
  • Calcium-Induced Calcium Release in Noradrenergic Neurons of the Locus Coeruleus
    bioRxiv preprint doi: https://doi.org/10.1101/853283; this version posted November 23, 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-NC-ND 4.0 International license. Calcium-induced calcium release in noradrenergic neurons of the locus coeruleus Hiroyuki Kawano1, Sara B. Mitchell1, Jin-Young Koh1,2,3, Kirsty M. Goodman1,4, and N. Charles Harata1,* 1 Department of Molecular Physiology and Biophysics, University of Iowa Carver College of Medicine, Iowa City, IA, USA 2 Molecular Otolaryngology and Renal Research Laboratories, Department of Otolaryngology-Head and Neck Surgery, University of Iowa Carver College of Medicine, Iowa City, IA, USA 3 Department of Biomedical Engineering, University of Iowa College of Engineering, Iowa City, IA, USA 4 Department of Biology & Biochemistry, University of Bath, Bath, UK * Correspondence to: N. Charles Harata, MD, PhD Department of Molecular Physiology & Biophysics University of Iowa Carver College of Medicine 51 Newton Road, Iowa City, IA 52242, USA Phone: 1-319-335-7820 Fax: 1-319-335-7330 E-mail: [email protected] Number of words: 8620; Number of figures: 12. 1 bioRxiv preprint doi: https://doi.org/10.1101/853283; this version posted November 23, 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-NC-ND 4.0 International license.
    [Show full text]
  • Tlx3 Exerts Direct Control in Specifying Excitatory Over Inhibitory Neurons in the Dorsal Spinal Cord
    fcell-09-642697 April 24, 2021 Time: 18:18 # 1 ORIGINAL RESEARCH published: 29 April 2021 doi: 10.3389/fcell.2021.642697 Tlx3 Exerts Direct Control in Specifying Excitatory Over Inhibitory Neurons in the Dorsal Spinal Cord Filipe A. Monteiro1,2,3*, Rafael M. Miranda1,2,3, Marta C. Samina1,2,3, Ana F. Dias2,3,4, Alexandre A. S. F. Raposo5,6, Patrícia Oliveira3,7, Carlos Reguenga1,2,3, Diogo S. Castro3,5,8† and Deolinda Lima1,2,3† 1 Unidade de Biologia Experimental, Departamento de Biomedicina, Faculdade de Medicina da Universidade do Porto, Porto, Portugal, 2 Pain Research Group, Instituto de Biologia Molecular e Celular, Porto, Portugal, 3 Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Porto, Portugal, 4 Instituto de Ciências Biomédicas Abel Salazar, Universidade do Porto, Porto, Portugal, 5 Molecular Neurobiology Group, Instituto Gulbenkian de Ciência, Oeiras, Portugal, 6 Instituto de Medicina Molecular João Lobo Antunes, Faculdade de Medicina da Universidade de Lisboa, Lisboa, Portugal, 7 Diagnostics, Institute of Molecular Pathology and Immunology, University of Porto, Porto, Portugal, 8 Stem Cells & Neurogenesis Group, Instituto de Biologia Molecular e Celular, Porto, Portugal The spinal cord dorsal horn is a major station for integration and relay of somatosensory information and comprises both excitatory and inhibitory neuronal populations. The homeobox gene Tlx3 acts as a selector gene to control the development of late- Edited by: Jiri Novotny, born excitatory (dILB) neurons by specifying glutamatergic transmitter fate in dorsal Charles University, Czechia spinal cord. However, since Tlx3 direct transcriptional targets remain largely unknown, Reviewed by: it remains to be uncovered how Tlx3 functions to promote excitatory cell fate.
    [Show full text]
  • Table 2. Significant
    Table 2. Significant (Q < 0.05 and |d | > 0.5) transcripts from the meta-analysis Gene Chr Mb Gene Name Affy ProbeSet cDNA_IDs d HAP/LAP d HAP/LAP d d IS Average d Ztest P values Q-value Symbol ID (study #5) 1 2 STS B2m 2 122 beta-2 microglobulin 1452428_a_at AI848245 1.75334941 4 3.2 4 3.2316485 1.07398E-09 5.69E-08 Man2b1 8 84.4 mannosidase 2, alpha B1 1416340_a_at H4049B01 3.75722111 3.87309653 2.1 1.6 2.84852656 5.32443E-07 1.58E-05 1110032A03Rik 9 50.9 RIKEN cDNA 1110032A03 gene 1417211_a_at H4035E05 4 1.66015788 4 1.7 2.82772795 2.94266E-05 0.000527 NA 9 48.5 --- 1456111_at 3.43701477 1.85785922 4 2 2.8237185 9.97969E-08 3.48E-06 Scn4b 9 45.3 Sodium channel, type IV, beta 1434008_at AI844796 3.79536664 1.63774235 3.3 2.3 2.75319499 1.48057E-08 6.21E-07 polypeptide Gadd45gip1 8 84.1 RIKEN cDNA 2310040G17 gene 1417619_at 4 3.38875643 1.4 2 2.69163229 8.84279E-06 0.0001904 BC056474 15 12.1 Mus musculus cDNA clone 1424117_at H3030A06 3.95752801 2.42838452 1.9 2.2 2.62132809 1.3344E-08 5.66E-07 MGC:67360 IMAGE:6823629, complete cds NA 4 153 guanine nucleotide binding protein, 1454696_at -3.46081884 -4 -1.3 -1.6 -2.6026947 8.58458E-05 0.0012617 beta 1 Gnb1 4 153 guanine nucleotide binding protein, 1417432_a_at H3094D02 -3.13334396 -4 -1.6 -1.7 -2.5946297 1.04542E-05 0.0002202 beta 1 Gadd45gip1 8 84.1 RAD23a homolog (S.
    [Show full text]
  • Edinburgh Research Explorer
    Edinburgh Research Explorer International Union of Basic and Clinical Pharmacology. LXXXVIII. G protein-coupled receptor list Citation for published version: Davenport, AP, Alexander, SPH, Sharman, JL, Pawson, AJ, Benson, HE, Monaghan, AE, Liew, WC, Mpamhanga, CP, Bonner, TI, Neubig, RR, Pin, JP, Spedding, M & Harmar, AJ 2013, 'International Union of Basic and Clinical Pharmacology. LXXXVIII. G protein-coupled receptor list: recommendations for new pairings with cognate ligands', Pharmacological reviews, vol. 65, no. 3, pp. 967-86. https://doi.org/10.1124/pr.112.007179 Digital Object Identifier (DOI): 10.1124/pr.112.007179 Link: Link to publication record in Edinburgh Research Explorer Document Version: Publisher's PDF, also known as Version of record Published In: Pharmacological reviews Publisher Rights Statement: U.S. Government work not protected by U.S. copyright General rights Copyright for the publications made accessible via the Edinburgh Research Explorer is retained by the author(s) and / or other copyright owners and it is a condition of accessing these publications that users recognise and abide by the legal requirements associated with these rights. Take down policy The University of Edinburgh has made every reasonable effort to ensure that Edinburgh Research Explorer content complies with UK legislation. If you believe that the public display of this file breaches copyright please contact [email protected] providing details, and we will remove access to the work immediately and investigate your claim. Download date: 02. Oct. 2021 1521-0081/65/3/967–986$25.00 http://dx.doi.org/10.1124/pr.112.007179 PHARMACOLOGICAL REVIEWS Pharmacol Rev 65:967–986, July 2013 U.S.
    [Show full text]
  • Keratins and Plakin Family Cytolinker Proteins Control the Length Of
    RESEARCH ARTICLE Keratins and plakin family cytolinker proteins control the length of epithelial microridge protrusions Yasuko Inaba*, Vasudha Chauhan, Aaron Paul van Loon, Lamia Saiyara Choudhury, Alvaro Sagasti* Molecular, Cell and Developmental Biology Department and Molecular Biology Institute, University of California, Los Angeles, Los Angeles, United States Abstract Actin filaments and microtubules create diverse cellular protrusions, but intermediate filaments, the strongest and most stable cytoskeletal elements, are not known to directly participate in the formation of protrusions. Here we show that keratin intermediate filaments directly regulate the morphogenesis of microridges, elongated protrusions arranged in elaborate maze-like patterns on the surface of mucosal epithelial cells. We found that microridges on zebrafish skin cells contained both actin and keratin filaments. Keratin filaments stabilized microridges, and overexpressing keratins lengthened them. Envoplakin and periplakin, plakin family cytolinkers that bind F-actin and keratins, localized to microridges, and were required for their morphogenesis. Strikingly, plakin protein levels directly dictate microridge length. An actin-binding domain of periplakin was required to initiate microridge morphogenesis, whereas periplakin-keratin binding was required to elongate microridges. These findings separate microridge morphogenesis into distinct steps, expand our understanding of intermediate filament functions, and identify microridges as protrusions that integrate actin and intermediate filaments. *For correspondence: [email protected] (YI); Introduction [email protected] (AS) Cytoskeletal filaments are scaffolds for membrane protrusions that create a vast diversity of cell shapes. The three major classes of cytoskeletal elements—microtubules, actin filaments, and inter- Competing interests: The mediate filaments (IFs)—each have distinct mechanical and biochemical properties and associate authors declare that no with different regulatory proteins, suiting them to different functions.
    [Show full text]
  • Supplementary Table S1. Upregulated Genes Differentially
    Supplementary Table S1. Upregulated genes differentially expressed in athletes (p < 0.05 and 1.3-fold change) Gene Symbol p Value Fold Change 221051_s_at NMRK2 0.01 2.38 236518_at CCDC183 0.00 2.05 218804_at ANO1 0.00 2.05 234675_x_at 0.01 2.02 207076_s_at ASS1 0.00 1.85 209135_at ASPH 0.02 1.81 228434_at BTNL9 0.03 1.81 229985_at BTNL9 0.01 1.79 215795_at MYH7B 0.01 1.78 217979_at TSPAN13 0.01 1.77 230992_at BTNL9 0.01 1.75 226884_at LRRN1 0.03 1.74 220039_s_at CDKAL1 0.01 1.73 236520_at 0.02 1.72 219895_at TMEM255A 0.04 1.72 201030_x_at LDHB 0.00 1.69 233824_at 0.00 1.69 232257_s_at 0.05 1.67 236359_at SCN4B 0.04 1.64 242868_at 0.00 1.63 1557286_at 0.01 1.63 202780_at OXCT1 0.01 1.63 1556542_a_at 0.04 1.63 209992_at PFKFB2 0.04 1.63 205247_at NOTCH4 0.01 1.62 1554182_at TRIM73///TRIM74 0.00 1.61 232892_at MIR1-1HG 0.02 1.61 204726_at CDH13 0.01 1.6 1561167_at 0.01 1.6 1565821_at 0.01 1.6 210169_at SEC14L5 0.01 1.6 236963_at 0.02 1.6 1552880_at SEC16B 0.02 1.6 235228_at CCDC85A 0.02 1.6 1568623_a_at SLC35E4 0.00 1.59 204844_at ENPEP 0.00 1.59 1552256_a_at SCARB1 0.02 1.59 1557283_a_at ZNF519 0.02 1.59 1557293_at LINC00969 0.03 1.59 231644_at 0.01 1.58 228115_at GAREM1 0.01 1.58 223687_s_at LY6K 0.02 1.58 231779_at IRAK2 0.03 1.58 243332_at LOC105379610 0.04 1.58 232118_at 0.01 1.57 203423_at RBP1 0.02 1.57 AMY1A///AMY1B///AMY1C///AMY2A///AMY2B// 208498_s_at 0.03 1.57 /AMYP1 237154_at LOC101930114 0.00 1.56 1559691_at 0.01 1.56 243481_at RHOJ 0.03 1.56 238834_at MYLK3 0.01 1.55 213438_at NFASC 0.02 1.55 242290_at TACC1 0.04 1.55 ANKRD20A1///ANKRD20A12P///ANKRD20A2///
    [Show full text]
  • The Ciliogenic Transcription Factor RFX3 Regulates Early Midline Distribution of Guidepost Neurons Required for Corpus Callosum Development
    The Ciliogenic Transcription Factor RFX3 Regulates Early Midline Distribution of Guidepost Neurons Required for Corpus Callosum Development Carine Benadiba1,2, Dario Magnani3, Mathieu Niquille1, Laurette Morle´ 2, Delphine Valloton1, Homaira Nawabi2, Aouatef Ait-Lounis4, Belkacem Otsmane1, Walter Reith4, Thomas Theil3, Jean- Pierre Hornung1,Ce´cile Lebrand1,5.*, Be´ne´dicte Durand2.* 1 De´partement de Biologie Cellulaire et de Morphologie, University of Lausanne, Lausanne, Switzerland, 2 Centre de Ge´ne´tique et de Physiologie Mole´culaire et Cellulaire, CNRS UMR 5534, Universite´ Claude Bernard Lyon 1, Lyon, France, 3 Centre for Integrative Physiology, University of Edinburgh, Edinburgh, United Kingdom, 4 Department of Pathology and Immunology, Faculty of Medicine, University of Geneva, Centre Me´dical Universitaire, Geneva, Switzerland, 5 National Center of Competence in Research Robotics, Ecole Polytechnique Fe´de´rale, Lausanne, Switzerland Abstract The corpus callosum (CC) is the major commissure that bridges the cerebral hemispheres. Agenesis of the CC is associated with human ciliopathies, but the origin of this default is unclear. Regulatory Factor X3 (RFX3) is a transcription factor involved in the control of ciliogenesis, and Rfx3–deficient mice show several hallmarks of ciliopathies including left–right asymmetry defects and hydrocephalus. Here we show that Rfx3–deficient mice suffer from CC agenesis associated with a marked disorganisation of guidepost neurons required for axon pathfinding across the midline. Using transplantation assays, we demonstrate that abnormalities of the mutant midline region are primarily responsible for the CC malformation. Conditional genetic inactivation shows that RFX3 is not required in guidepost cells for proper CC formation, but is required before E12.5 for proper patterning of the cortical septal boundary and hence accurate distribution of guidepost neurons at later stages.
    [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]
  • The Snakeskin-Mesh Complex of Smooth Septate Junction Restricts Yorkie to Regulate Intestinal Homeostasis in Drosophila
    University of Massachusetts Medical School eScholarship@UMMS GSBS Dissertations and Theses Graduate School of Biomedical Sciences 2020-01-15 The Snakeskin-Mesh Complex of Smooth Septate Junction Restricts Yorkie to Regulate Intestinal Homeostasis in Drosophila Hsi-Ju Chen University of Massachusetts Medical School Let us know how access to this document benefits ou.y Follow this and additional works at: https://escholarship.umassmed.edu/gsbs_diss Part of the Cell Biology Commons, and the Genetics Commons Repository Citation Chen H. (2020). The Snakeskin-Mesh Complex of Smooth Septate Junction Restricts Yorkie to Regulate Intestinal Homeostasis in Drosophila. GSBS Dissertations and Theses. https://doi.org/10.13028/ 0r15-ze63. Retrieved from https://escholarship.umassmed.edu/gsbs_diss/1059 This material is brought to you by eScholarship@UMMS. It has been accepted for inclusion in GSBS Dissertations and Theses by an authorized administrator of eScholarship@UMMS. For more information, please contact [email protected]. The Snakeskin-Mesh Complex of Smooth Septate Junction Restricts Yorkie to Regulate Intestinal Homeostasis in Drosophila A Dissertation Presented by Hsi-Ju Chen Submitted to the Faculty of the University of Massachusetts Graduate School of Biomedical Sciences, Worcester In partial fulfilment of the requirement for the requirements for the Degree of Doctor of Philosophy November 19, 2019 TABLE OF CONTENTS ABSTRACT CHAPTER I.....................................................................................................
    [Show full text]
  • 4-6 Weeks Old Female C57BL/6 Mice Obtained from Jackson Labs Were Used for Cell Isolation
    Methods Mice: 4-6 weeks old female C57BL/6 mice obtained from Jackson labs were used for cell isolation. Female Foxp3-IRES-GFP reporter mice (1), backcrossed to B6/C57 background for 10 generations, were used for the isolation of naïve CD4 and naïve CD8 cells for the RNAseq experiments. The mice were housed in pathogen-free animal facility in the La Jolla Institute for Allergy and Immunology and were used according to protocols approved by the Institutional Animal Care and use Committee. Preparation of cells: Subsets of thymocytes were isolated by cell sorting as previously described (2), after cell surface staining using CD4 (GK1.5), CD8 (53-6.7), CD3ε (145- 2C11), CD24 (M1/69) (all from Biolegend). DP cells: CD4+CD8 int/hi; CD4 SP cells: CD4CD3 hi, CD24 int/lo; CD8 SP cells: CD8 int/hi CD4 CD3 hi, CD24 int/lo (Fig S2). Peripheral subsets were isolated after pooling spleen and lymph nodes. T cells were enriched by negative isolation using Dynabeads (Dynabeads untouched mouse T cells, 11413D, Invitrogen). After surface staining for CD4 (GK1.5), CD8 (53-6.7), CD62L (MEL-14), CD25 (PC61) and CD44 (IM7), naïve CD4+CD62L hiCD25-CD44lo and naïve CD8+CD62L hiCD25-CD44lo were obtained by sorting (BD FACS Aria). Additionally, for the RNAseq experiments, CD4 and CD8 naïve cells were isolated by sorting T cells from the Foxp3- IRES-GFP mice: CD4+CD62LhiCD25–CD44lo GFP(FOXP3)– and CD8+CD62LhiCD25– CD44lo GFP(FOXP3)– (antibodies were from Biolegend). In some cases, naïve CD4 cells were cultured in vitro under Th1 or Th2 polarizing conditions (3, 4).
    [Show full text]