Recessive Retinopathy Consequent on Mutant G-Protein Β Subunit 3 (GNB3)

Total Page:16

File Type:pdf, Size:1020Kb

Recessive Retinopathy Consequent on Mutant G-Protein Β Subunit 3 (GNB3) Supplementary Online Content Arno G, Holder GE, Chakarova C, et al; UK Inherited Retinal Disease Consortium. Recessive retinopathy consequent on mutant G-protein β subunit 3 (GNB3). JAMA Ophthalmol. Published online June 9, 2016. doi:10.1001/jamaophthalmol.2016.1543. eAppendix 1. Methods eAppendix 2. Results: Variant Data eTable 1. Exome Sequencing Homozygous and Double Heterozygous Variants eTable 2. Variants Identified by Direct Sanger Sequencing This supplementary material has been provided by the authors to give readers additional information about their work. © 2016 American Medical Association. All rights reserved. Downloaded From: https://jamanetwork.com/ on 09/25/2021 eAppendix 1. Methods Gene panel screening Prior genetic screening comprised direct Sanger sequencing of all coding exons of NR2E3 and a Next Generation Sequencing (NGS) panel of 105 retinal dystrophy genes tested at the Manchester Centre for Genomic Medicine, UK (Gene list below). No Pathogenic mutations were identified. ABHD12 CDH3 GPR179 LRIT3 PCYT1A TRPM1 ACBD5 CEP164 GRM6 LZTFL1 PEX1 TSPAN12 ADAMTS18 CIB2 HARS MFRP PEX2 TUB AHI1 CLN3 HMX1 MVK PEX7 VCAN ARL2BP CNNM4 IFT140 NEK2 PHYH VPS13B BB1P1 CSPP1 IMPG1 NMNAT1 PLA2G5 WDR19 C21orf2 CYP4V2 INPP5E NPHP1 PRPF4 ZNF423 C2ORF86 DTHD1 INVS NPHP3 RAB28 C8ORF37 EMC1 IQCB1 NPHP4 RBP4 CABP4 FLVCR1 ITM2B NYX RPGRIP1L CACNA1F GNAT1 KCNJ13 OAT SDCCAG8 CAPN5 GNPTG KIAA1549 OFD1 SLC24A1 CC2D2A GPR125 KIF11 PANK2 TMEM237 Whole exome sequencing methodology 200ng of genomic DNA were processed according to the Agilent SureSelect XT Library Prep protocol (Agilent Technologies, CA, US) with exons being captured using the SureSelect Exome V5 Capture library. After hybridization and indexing, six samples were pooled and 100bp paired end sequencing was performed (Illumina HiSeq 2500 sequencer) at the University of Leeds Next Generation Sequencing Facility, UK. Reads were aligned to the hg19 human reference sequence (build GRCh37) using Novoalign version 2.08. Duplicate reads were marked using Picard tools MarkDuplicates. Calling was performed using GATK, creating gVCF formatted files for each sample. The individual gVCF files for the exomes discussed in this study, in combination with ~ 3,000 clinical exomes (UCL-exomes consortium), were combined into merged VCF files for each chromosome containing on average 100 samples each. The final variant calling was performed using the GATK Genotype gVCFs module jointly for all samples (cases and controls). Variant quality scores were then re-calibrated according to GATK best practices separately for indels and SNPs. Resulting variants were annotated using ANNOVAR based on Ensembl gene and transcript definitions. Candidate variants were filtered based on function (non- synonymous, presumed loss-of-function or splicing) and MAF (< 0.001). © 2016 American Medical Association. All rights reserved. Downloaded From: https://jamanetwork.com/ on 09/25/2021 eAppendix 2. Results: Variant Data WES analysis revealed 25,877 exonic ±5bp variants that passed quality filter; 1080 of which had MAF ≤0.001 in NHLBI GO Exome Sequencing Project (ESP, available at http://evs.gs.washington.edu/EVS/EVS) controls. No likely pathogenic variants were noted in any retinal dystrophy genes. Further variant filtering using an in-house control dataset of approximately 3000 exomes revealed 48 genes containing ≥2 rare (≤0.001 MAF) variants, of which 4 genes were predicted to be probable recessive null assuming double heterozygous null variants were in trans (WDR86, GNB3, TCHH, AVL9, see eTable 1). Homozygosity analysis of all variants revealed 2 ≥10mb stretches of unbroken homozygous SNPs (chr12:235,022-11,214,212 [encompassing the GNB3 locus] and chr19:9,213,396-19,230,868) suggestive of parental relatedness. Analysis of NGS read depth data using ExomeDepthe1 and close scrutiny of reads and coverage of the GNB3 locus using the Integrated Genome Viewer (IGV)e2,e3 was not suggestive of any genomic rearrangements resulting in loss of one allele. Uniparental isodisomy could not be ruled out due to unavailability of parental DNA samples. e1. Plagnol V, Curtis J, Epstein M, et al. A robust model for read count data in exome sequencing experiments and implications for copy number variant calling. Bioinformatics. 2012;28(21):2747‐54. doi:10.1093/bioinformatics/bts526. e2. Robinson JT, Thorvaldsdóttir H, Winckler W, et al. Integrative genomics viewer. Nat Biotechnol. 2011;29(1):24‐6. doi:10.1038/nbt.1754. e3. Thorvaldsdóttir H, Robinson JT, Mesirov JP. Integrative Genomics Viewer (IGV): high‐ performance genomics data visualization and exploration. Brief Bioinform. 2013;14(2):178‐92. doi:10.1093/bib/bbs017. © 2016 American Medical Association. All rights reserved. Downloaded From: https://jamanetwork.com/ on 09/25/2021 eTable 1. Exome Sequencing Homozygous and Double Heterozygous Variants Homozygous Functio Exonic signat n Function HUGO Description AA Change ure IQ motif containing 1_156 nonsynonymo GTPase activating protein 50097 exonic us SNV IQGAP3 3 ENSG00000183856:ENST00000491900:exon32:c.C4036G:p.L1346V,ENSG00000183856:ENST00000361170:exon33:c.C4165G:p.L1389V 8_G_C 1_200 nonsynonymo DEAD (Asp‐Glu‐Ala‐Asp) ENSG00000118197:ENST00000331314:exon2:c.A536G:p.E179G,ENSG00000118197:ENST00000367348:exon2:c.A536G:p.E179G,ENSG00 63533 exonic us SNV DDX59 box polypeptide 59 000118197:ENST00000447706:exon2:c.A536G:p.E179G 3_T_C 7_168 nonsynonymo 72905 exonic us SNV AGR2 anterior gradient 2 ENSG00000106541:ENST00000419572:exon1:c.A28G:p.S10G _T_C 7_931 25288 splicing CALCR calcitonin receptor _T_C 7_151 10653 exonic stopgain SNV WDR86 WD repeat domain 86 ENSG00000187260:ENST00000334493:exon1:c.C142T:p.Q48X,ENSG00000187260:ENST00000469830:exon1:c.C142T:p.Q48X 4_G_A 9_148 nonsynonymo FRAS1 related ENSG00000164946:ENST00000380880:exon4:c.C583T:p.P195S,ENSG00000164946:ENST00000380875:exon5:c.C583T:p.P195S,ENSG0000 59229 exonic us SNV FREM1 extracellular matrix 1 0164946:ENST00000380881:exon5:c.C583T:p.P195S,ENSG00000164946:ENST00000422223:exon5:c.C583T:p.P195S _G_A 9_131 nonsynonymo cysteine conjugate‐beta ENSG00000171097:ENST00000320665:exon11:c.A1004T:p.Y335F,ENSG00000171097:ENST00000302586:exon12:c.A1154T:p.Y385F,ENSG 59607 exonic us SNV CCBL1 lyase, cytoplasmic 00000171097:ENST00000436267:exon14:c.A1436T:p.Y479F 4_T_A © 2016 American Medical Association. All rights reserved. Downloaded From: https://jamanetwork.com/ on 09/25/2021 ENSG00000066468:ENST00000346997:exon1:c.G34A:p.V12M,ENSG00000066468:ENST00000369056:exon1:c.G34A:p.V12M,ENSG00000 066468:ENST00000369061:exon1:c.G34A:p.V12M,ENSG00000066468:ENST00000336553:exon2:c.G34A:p.V12M,ENSG00000066468:ENS T00000351936:exon2:c.G34A:p.V12M,ENSG00000066468:ENST00000356226:exon2:c.G34A:p.V12M,ENSG00000066468:ENST000003575 55:exon2:c.G34A:p.V12M,ENSG00000066468:ENST00000358487:exon2:c.G34A:p.V12M,ENSG00000066468:ENST00000359354:exon2:c. 10_12 G34A:p.V12M,ENSG00000066468:ENST00000360144:exon2:c.G34A:p.V12M,ENSG00000066468:ENST00000369058:exon2:c.G34A:p.V12 33532 nonsynonymo fibroblast growth factor M,ENSG00000066468:ENST00000369059:exon2:c.G34A:p.V12M,ENSG00000066468:ENST00000369060:exon2:c.G34A:p.V12M,ENSG000 98_C_ exonic us SNV FGFR2 receptor 2 00066468:ENST00000457416:exon2:c.G34A:p.V12M T olfactory receptor, family 11_57 nonsynonymo 52, subfamily N, member 76368 exonic us SNV OR52N4 4 (gene/pseudogene) ENSG00000181074:ENST00000317254:exon1:c.G398A:p.R133H _G_A 11_65 nonsynonymo dynein heavy chain ENSG00000179532:ENST00000527990:exon1:c.G17A:p.R6K,ENSG00000179532:ENST00000354685:exon2:c.G17A:p.R6K,ENSG000001795 19462 exonic us SNV DNHD1 domain 1 32:ENST00000254579:exon3:c.G17A:p.R6K _G_A guanine nucleotide binding protein (G ENSG00000111664:ENST00000537035:exon3:c.C124T:p.R42X,ENSG00000111664:ENST00000435982:exon4:c.C124T:p.R42X,ENSG00000 12_69 protein), beta polypeptide 111664:ENST00000541257:exon4:c.C124T:p.R42X,ENSG00000111664:ENST00000541978:exon4:c.C124T:p.R42X,ENSG00000111664:ENS 52161 exonic stopgain SNV GNB3 3 T00000229264:exon5:c.C124T:p.R42X _C_T 12_11 13369 nonsynonymo coiled‐coil domain ENSG00000173093:ENST00000552694:exon8:c.C1094T:p.P365L,ENSG00000173093:ENST00000545036:exon9:c.C1211T:p.P404L,ENSG00 18_C_ exonic us SNV CCDC63 containing 63 000173093:ENST00000308208:exon10:c.C1331T:p.P444L T leucine rich repeat 19_79 nonsynonymo containing 8 family, 64234 exonic us SNV LRRC8E member E ENSG00000171017:ENST00000306708:exon3:c.A827G:p.Y276C _A_G 19_81 nonsynonymo ENSG00000142449:ENST00000270509:exon54:c.C6826T:p.R2276W,ENSG00000142449:ENST00000600128:exon55:c.C6826T:p.R2276W, 51138 exonic us SNV FBN3 fibrillin 3 ENSG00000142449:ENST00000601739:exon55:c.C6826T:p.R2276W _G_A 19_11 nonsynonymo SWIM‐type zinc finger 7 48619 exonic us SNV SWSAP1 associated protein 1 ENSG00000173928:ENST00000312423:exon2:c.G194A:p.R65H 6_G_A © 2016 American Medical Association. All rights reserved. Downloaded From: https://jamanetwork.com/ on 09/25/2021 19_13 nonsynonymo synaptonemal complex 01088 exonic us SNV SYCE2 central element protein 2 ENSG00000161860:ENST00000293695:exon5:c.C545T:p.P182L 5_G_A C3 and PZP‐like, alpha‐2‐ 19_17 nonsynonymo macroglobulin domain 08689 exonic us SNV CPAMD8 containing 8 ENSG00000160111:ENST00000443236:exon16:c.C1964T:p.A655V 7_G_A ENSG00000130479:ENST00000324096:exon1:c.G23T:p.G8V,ENSG00000130479:ENST00000594212:exon1:c.G23T:p.G8V,ENSG000001304 19_17 nonsynonymo microtubule‐associated 79:ENST00000597735:exon1:c.G23T:p.G8V,ENSG00000130479:ENST00000600186:exon1:c.G23T:p.G8V,ENSG00000130479:ENST000006 83033 exonic us SNV MAP1S protein 1S 00608:exon1:c.G23T:p.G8V
Recommended publications
  • RBP-J Signaling − Cells Through Notch Novel IRF8-Controlled
    Sca-1+Lin−CD117− Mesenchymal Stem/Stromal Cells Induce the Generation of Novel IRF8-Controlled Regulatory Dendritic Cells through Notch −RBP-J Signaling This information is current as of September 25, 2021. Xingxia Liu, Shaoda Ren, Chaozhuo Ge, Kai Cheng, Martin Zenke, Armand Keating and Robert C. H. Zhao J Immunol 2015; 194:4298-4308; Prepublished online 30 March 2015; doi: 10.4049/jimmunol.1402641 Downloaded from http://www.jimmunol.org/content/194/9/4298 Supplementary http://www.jimmunol.org/content/suppl/2015/03/28/jimmunol.140264 http://www.jimmunol.org/ Material 1.DCSupplemental References This article cites 59 articles, 19 of which you can access for free at: http://www.jimmunol.org/content/194/9/4298.full#ref-list-1 Why The JI? Submit online. • Rapid Reviews! 30 days* from submission to initial decision by guest on September 25, 2021 • No Triage! Every submission reviewed by practicing scientists • Fast Publication! 4 weeks from acceptance to publication *average Subscription Information about subscribing to The Journal of Immunology is online at: http://jimmunol.org/subscription Permissions Submit copyright permission requests at: http://www.aai.org/About/Publications/JI/copyright.html Email Alerts Receive free email-alerts when new articles cite this article. Sign up at: http://jimmunol.org/alerts The Journal of Immunology is published twice each month by The American Association of Immunologists, Inc., 1451 Rockville Pike, Suite 650, Rockville, MD 20852 Copyright © 2015 by The American Association of Immunologists, Inc. All rights reserved. Print ISSN: 0022-1767 Online ISSN: 1550-6606. The Journal of Immunology Sca-1+Lin2CD1172 Mesenchymal Stem/Stromal Cells Induce the Generation of Novel IRF8-Controlled Regulatory Dendritic Cells through Notch–RBP-J Signaling Xingxia Liu,*,1 Shaoda Ren,*,1 Chaozhuo Ge,* Kai Cheng,* Martin Zenke,† Armand Keating,‡,x and Robert C.
    [Show full text]
  • Microglia Emerge from Erythromyeloid Precursors Via Pu.1- and Irf8-Dependent Pathways
    ART ic LE S Microglia emerge from erythromyeloid precursors via Pu.1- and Irf8-dependent pathways Katrin Kierdorf1,2, Daniel Erny1, Tobias Goldmann1, Victor Sander1, Christian Schulz3,4, Elisa Gomez Perdiguero3,4, Peter Wieghofer1,2, Annette Heinrich5, Pia Riemke6, Christoph Hölscher7,8, Dominik N Müller9, Bruno Luckow10, Thomas Brocker11, Katharina Debowski12, Günter Fritz1, Ghislain Opdenakker13, Andreas Diefenbach14, Knut Biber5,15, Mathias Heikenwalder16, Frederic Geissmann3,4, Frank Rosenbauer6 & Marco Prinz1,17 Microglia are crucial for immune responses in the brain. Although their origin from the yolk sac has been recognized for some time, their precise precursors and the transcription program that is used are not known. We found that mouse microglia were derived from primitive c-kit+ erythromyeloid precursors that were detected in the yolk sac as early as 8 d post conception. + lo − + − + These precursors developed into CD45 c-kit CX3CR1 immature (A1) cells and matured into CD45 c-kit CX3CR1 (A2) cells, as evidenced by the downregulation of CD31 and concomitant upregulation of F4/80 and macrophage colony stimulating factor receptor (MCSF-R). Proliferating A2 cells became microglia and invaded the developing brain using specific matrix metalloproteinases. Notably, microgliogenesis was not only dependent on the transcription factor Pu.1 (also known as Sfpi), but also required Irf8, which was vital for the development of the A2 population, whereas Myb, Id2, Batf3 and Klf4 were not required. Our data provide cellular and molecular insights into the origin and development of microglia. Microglia are the tissue macrophages of the brain and scavenge dying have the ability to give rise to microglia and macrophages in vitro cells, pathogens and molecules using pattern recognition receptors and in vivo under defined conditions.
    [Show full text]
  • Roles and Regulation of Transcription Factor Mafa in Islet Β-Cells
    Endocr. J./ S. ARAMATA et al.: INSULIN TRANSCRIPTION AND MafA doi: 10.1507/endocrj.KR-101 REVIEW Roles and Regulation of Transcription Factor MafA in Islet β-cells * SHINSAKU ARAMATA, SONG-IEE HAN AND KOHSUKE KATAOKA Graduate School of Biological Science, Nara Institute of Science and Technology, 8916-5 Takayama-cho, Ikoma, Nara 630-0192, Japan *Graduate School of Life and Environmental Sciences, University of Tsukuba, Tsukuba Science City, Ibaraki 305-8572, Japan. Released online August 30, 2007 Correspondence to: Kohsuke KATAOKA, Graduate School of Biological Science, Nara Institute of Science and Technology, 8916-5 Takayama-cho, Ikoma, Nara 630-0192, Japan Abstract. Insulin is a critical hormone in the regulation of blood glucose levels. It is produced exclusively by pancreatic islet β-cells. β-cell-enriched transcription factors, such as Pdx1 and Beta2, have dual roles in the activation of the insulin gene promoter establishing β-cell-specific insulin expression, and in the regulation of β-cell differentiation. It was shown that MafA, a β-cell-specific member of the Maf family of transcription factors, binds to the conserved C1/RIPE3b element of the insulin promoter. The Maf family proteins regulate tissue-specific gene expression and cell differentiation in a wide variety of tissues. MafA acts synergistically with Pdx1 and Beta2 to activate the insulin gene promoter, and mice with a targeted deletion of mafA develop age-dependent diabetes. MafA also regulates genes involved in β-cell function such as Glucose transporter 2, Glucagons-like peptide 1 receptor, and Prohormone convertase 1/3. The abundance of MafA in β-cells is regulated at both the transcriptional and post-translational levels by glucose and oxidative stress.
    [Show full text]
  • Differential Expression of Vitamin D Binding Protein in Thyroid Cancer Health Disparities
    www.oncotarget.com Oncotarget, 2021, Vol. 12, (No. 7), pp: 596-607 Research Paper Differential expression of Vitamin D binding protein in thyroid cancer health disparities Brittany Mull1, Ryan Davis2,3, Iqbal Munir4, Mia C. Perez5, Alfred A. Simental6 and Salma Khan2,3,6,7 1Harbor UCLA Medical Center, Torrance, CA 90502, USA 2Division of Biochemistry, Loma Linda, CA 92350, USA 3Center for Health Disparities & Molecular Medicine, Loma Linda, CA 92350, USA 4Riverside University Health System, Moreno Valley, CA 92555, USA 5Department of Pathology & Human Anatomy, Loma Linda University School of Medicine, Loma Linda, CA 92354, USA 6Department of Otolaryngology, Loma Linda University School of Medicine, Loma Linda, CA 92354, USA 7Department of Internal Medicine, Loma Linda University School of Medicine, Loma Linda, CA 92354, USA Correspondence to: Salma Khan, email: [email protected] Keywords: DBP; thyroid cancer; health disparities Received: November 16, 2020 Accepted: March 05, 2021 Published: March 30, 2021 Copyright: © 2021 Mull et al. This is an open access article distributed under the terms of the Creative Commons Attribution License (CC BY 3.0), which permits unrestricted use, distribution, and reproduction in any medium, provided the original author and source are credited. ABSTRACT Thyroid cancer incidence, recurrence, and death rates are higher among Filipino Americans than European Americans. We propose that vitamin D binding protein (DBP) with multifunctionality with ethnic variability plays a key role within different ethnicities. In this study, we determined the correlation between differential DBP expression in tumor tissues and cancer staging in Filipino Americans versus European Americans. We assayed DBP expression by immunohistochemistry and analyzed the data with confocal microscopy on 200 thyroid cancer archival tissue samples obtained from both ethnicities.
    [Show full text]
  • Prox1regulates the Subtype-Specific Development of Caudal Ganglionic
    The Journal of Neuroscience, September 16, 2015 • 35(37):12869–12889 • 12869 Development/Plasticity/Repair Prox1 Regulates the Subtype-Specific Development of Caudal Ganglionic Eminence-Derived GABAergic Cortical Interneurons X Goichi Miyoshi,1 Allison Young,1 Timothy Petros,1 Theofanis Karayannis,1 Melissa McKenzie Chang,1 Alfonso Lavado,2 Tomohiko Iwano,3 Miho Nakajima,4 Hiroki Taniguchi,5 Z. Josh Huang,5 XNathaniel Heintz,4 Guillermo Oliver,2 Fumio Matsuzaki,3 Robert P. Machold,1 and Gord Fishell1 1Department of Neuroscience and Physiology, NYU Neuroscience Institute, Smilow Research Center, New York University School of Medicine, New York, New York 10016, 2Department of Genetics & Tumor Cell Biology, St. Jude Children’s Research Hospital, Memphis, Tennessee 38105, 3Laboratory for Cell Asymmetry, RIKEN Center for Developmental Biology, Kobe 650-0047, Japan, 4Laboratory of Molecular Biology, Howard Hughes Medical Institute, GENSAT Project, The Rockefeller University, New York, New York 10065, and 5Cold Spring Harbor Laboratory, Cold Spring Harbor, New York 11724 Neurogliaform (RELNϩ) and bipolar (VIPϩ) GABAergic interneurons of the mammalian cerebral cortex provide critical inhibition locally within the superficial layers. While these subtypes are known to originate from the embryonic caudal ganglionic eminence (CGE), the specific genetic programs that direct their positioning, maturation, and integration into the cortical network have not been eluci- dated. Here, we report that in mice expression of the transcription factor Prox1 is selectively maintained in postmitotic CGE-derived cortical interneuron precursors and that loss of Prox1 impairs the integration of these cells into superficial layers. Moreover, Prox1 differentially regulates the postnatal maturation of each specific subtype originating from the CGE (RELN, Calb2/VIP, and VIP).
    [Show full text]
  • BRI2 (Itm2b) Inhibits A␤ Deposition in Vivo
    6030 • The Journal of Neuroscience, June 4, 2008 • 28(23):6030–6036 Neurobiology of Disease BRI2 (ITM2b) Inhibits A␤ Deposition In Vivo Jungsu Kim,1 Victor M. Miller,1 Yona Levites,1 Karen Jansen West,1 Craig W. Zwizinski,1 Brenda D. Moore,1 Fredrick J. Troendle,1 Maralyssa Bann,1 Christophe Verbeeck,1 Robert W. Price,1 Lisa Smithson,1 Leilani Sonoda,1 Kayleigh Wagg,1 Vijayaraghavan Rangachari,1 Fanggeng Zou,1 Steven G. Younkin,1 Neill Graff-Radford,2 Dennis Dickson,1 Terrone Rosenberry,1 and Todd E. Golde1 Departments of 1Neuroscience and 2Neurology, Mayo Clinic College of Medicine, Mayo Clinic Jacksonville, Jacksonville, Florida 32224 AnalysesofthebiologiceffectsofmutationsintheBRI2(ITM2b)andtheamyloid␤precursorprotein(APP)genessupportthehypothesis that cerebral accumulation of amyloidogenic peptides in familial British and familial Danish dementias and Alzheimer’s disease (AD) is associatedwithneurodegeneration.WehaveusedsomaticbraintransgenictechnologytoexpresstheBRI2andBRI2-A␤1–40transgenes in APP mouse models. Expression of BRI2-A␤1–40 mimics the suppressive effect previously observed using conventional transgenic methods, further validating the somatic brain transgenic methodology. Unexpectedly, we also find that expression of wild-type human BRI2 reduces cerebral A␤ deposition in an AD mouse model. Additional data indicate that the 23 aa peptide, Bri23, released from BRI2 by normal processing, is present in human CSF, inhibits A␤ aggregation in vitro and mediates its anti-amyloidogenic effect in vivo. These studies demonstrate that BRI2 is a novel mediator of A␤ deposition in vivo. Key words: BRI2; ITM2b; amyloid ␤ protein; Alzheimer’s disease; somatic brain transgenesis; adeno-associated virus Introduction pathological and clinical similarities between FBD, FDD, and Familial British and Danish dementias (FBD and FDD, respec- Alzheimer’s disease (AD).
    [Show full text]
  • Cerebral Amyloidosis, Amyloid Angiopathy, and Their Relationship to Stroke and Dementia
    65 Cerebral amyloidosis, amyloid angiopathy, and their relationship to stroke and dementia ∗ Jorge Ghiso and Blas Frangione β-pleated sheet structure, the conformation responsi- Department of Pathology, New York University School ble for their physicochemical properties and tinctoreal of Medicine, New York, NY, USA characteristics. So far, 20 different proteins have been identified as subunits of amyloid fibrils [56,57,60 (for review and nomenclature)]. Although collectively they Cerebral amyloid angiopathy (CAA) is the common term are products of normal genes, several amyloid precur- used to define the deposition of amyloid in the walls of sors contain abnormal amino acid substitutions that can medium- and small-size leptomeningeal and cortical arteries, arterioles and, less frequently, capillaries and veins. CAA impose an unusual potential for self-aggregation. In- is an important cause of cerebral hemorrhages although it creased levels of amyloid precursors, either in the cir- may also lead to ischemic infarction and dementia. It is a culation or locally at sites of deposition, are usually the feature commonly associated with normal aging, Alzheimer result of overexpression, defective clearance, or both. disease (AD), Down syndrome (DS), and Sporadic Cerebral Of all the amyloid proteins identified, less than half are Amyloid Angiopathy. Familial conditions in which amyloid known to cause amyloid deposition in the central ner- is chiefly deposited as CAA include hereditary cerebral hem- vous system (CNS), which in turn results in cognitive orrhage with amyloidosis of Icelandic type (HCHWA-I), fa- decline, dementia, stroke, cerebellar and extrapyrami- milial CAA related to Aβ variants, including hereditary cere- dal signs, or a combination of them.
    [Show full text]
  • Temporal Mapping of CEBPA and CEBPB Binding
    Downloaded from genome.cshlp.org on September 26, 2021 - Published by Cold Spring Harbor Laboratory Press Temporal mapping of CEBPA and CEBPB binding during liver regeneration reveals dynamic occupancy and specific regulatory codes for homeostatic and cell cycle gene batteries Janus Schou Jakobsen1,2,3,*, Johannes Waage1,2,3,4, Nicolas Rapin1,2,3,4, Hanne Cathrine Bisgaard5, Fin Stolze Larsen6, Bo Torben Porse1,2,3,* 1 The Finsen Laboratory, Rigshospitalet, Faculty of Health Sciences, University of Copenhagen, DK2200 Copenhagen, Denmark; 2 Biotech Research and Innovation Centre (BRIC), University of Copenhagen, DK-2200 Copenhagen, Denmark; 3 The Danish Stem Cell Centre (DanStem) Faculty of Health Sciences, University of Copenhagen, DK2200 Copenhagen Denmark; 4 The Bioinformatics Centre, University of Copenhagen, DK2200, Copenhagen, Denmark; 5 Department of Cellular and Molecular Medicine, Faculty of Health Sciences, University of Copenhagen, DK- 2100 Copenhagen, Denmark; 6 Department of Hepatology, Rigshospitalet, DK2200 Copenhagen, Denmark. JW: [email protected]; NR: [email protected]; HCB: [email protected]; FSL: [email protected] * Corresponding authors: BTP: [email protected]; JSJ: [email protected], The Finsen Laboratory, Ole Maaløesvej 5, Copenhagen Biocenter, University of Copenhagen, DK2200 Copenhagen, Denmark. Telephone: +45 3545 6023 Running title: Regulation of liver regeneration Keywords: Temporal ChIP-seq, dynamic binding, liver regeneration, C/EBPalpha, C/EBPbeta, transcriptional networks 1 Downloaded from genome.cshlp.org on September 26, 2021 - Published by Cold Spring Harbor Laboratory Press Abstract Dynamic shifts in transcription factor binding are central to the regulation of biological processes by allowing rapid changes in gene transcription.
    [Show full text]
  • Unbiased RNA-Seq-Driven Identification and Validation Of
    Dai et al. BMC Genomics (2021) 22:27 https://doi.org/10.1186/s12864-020-07318-y RESEARCH ARTICLE Open Access Unbiased RNA-Seq-driven identification and validation of reference genes for quantitative RT-PCR analyses of pooled cancer exosomes Yao Dai1, Yumeng Cao1, Jens Köhler2, Aiping Lu1, Shaohua Xu1* and Haiyun Wang1* Abstract Background: Exosomes are extracellular vesicles (EVs) derived from endocytic compartments of eukaryotic cells which contain various biomolecules like mRNAs or miRNAs. Exosomes influence the biologic behaviour and progression of malignancies and are promising candidates as non-invasive diagnostic biomarkers or as targets for therapeutic interventions. Usually, quantitative real-time polymerase chain reaction (qRT-PCR) is used to assess gene expression in cancer exosomes, however, the ideal reference genes for normalization yet remain to be identified. Results: In this study, we performed an unbiased analysis of high-throughput mRNA and miRNA-sequencing data from exosomes of patients with various cancer types and identify candidate reference genes and miRNAs in cancer exosomes. The expression stability of these candidate reference genes was evaluated by the coefficient of variation “CV” and the average expression stability value “M”. We subsequently validated these candidate reference genes in exosomes from an independent cohort of ovarian cancer patients and healthy control individuals by qRT-PCR. Conclusions: Our study identifies OAZ1 and hsa-miR-6835-3p as the most reliable individual reference genes for mRNA and miRNA quantification, respectively. For superior accuracy, we recommend the use of a combination of reference genes - OAZ1/SERF2/MPP1 for mRNA and hsa-miR-6835-3p/hsa-miR-4468-3p for miRNA analyses.
    [Show full text]
  • Transcriptional Control of Microglia Phenotypes in Health and Disease
    REVIEW SERIES: GLIA AND NEURODEGENERATION The Journal of Clinical Investigation Series Editors: Marco Colonna and David Holtzmann Transcriptional control of microglia phenotypes in health and disease Inge R. Holtman,1,2 Dylan Skola,1 and Christopher K. Glass1,3 1Department of Cellular and Molecular Medicine, UCSD, San Diego, California, USA. 2Department of Medical Physiology, University of Groningen, University Medical Center Groningen, Groningen, Netherlands. 3Department of Medicine, UCSD, San Diego, California, USA. Microglia are the main resident macrophage population of the CNS and perform numerous functions required for CNS development, homeostasis, immunity, and repair. Many lines of evidence also indicate that dysregulation of microglia contributes to the pathogenesis of neurodegenerative and behavioral diseases. These observations provide a compelling argument to more clearly define the mechanisms that control microglia identity and function in health and disease. In this Review, we present a conceptual framework for how different classes of transcription factors interact to select and activate regulatory elements that control microglia development and their responses to internal and external signals. We then describe functions of specific transcription factors in normal and pathological contexts and conclude with a consideration of open questions to be addressed in the future. Introduction regulatory control necessary to generate cell type–specific programs Microglia are tissue-resident macrophages that perform CNS- of gene expression. This additional information is provided by distal specific functions (1). They derive from a unique lineage of eryth- regulatory elements called enhancers (12). Enhancers represent the romyeloid precursors (EMPs) in the yolk sac and fetal liver (2). most numerous binding sites for LDTFs and signal-dependent tran- EMPs infiltrate the brain during early development, differentiate scription factors (SDTFs), and are major sites for the integration of into microglia, and maintain their population by self-renewal (3).
    [Show full text]
  • Human Induced Pluripotent Stem Cell–Derived Podocytes Mature Into Vascularized Glomeruli Upon Experimental Transplantation
    BASIC RESEARCH www.jasn.org Human Induced Pluripotent Stem Cell–Derived Podocytes Mature into Vascularized Glomeruli upon Experimental Transplantation † Sazia Sharmin,* Atsuhiro Taguchi,* Yusuke Kaku,* Yasuhiro Yoshimura,* Tomoko Ohmori,* ‡ † ‡ Tetsushi Sakuma, Masashi Mukoyama, Takashi Yamamoto, Hidetake Kurihara,§ and | Ryuichi Nishinakamura* *Department of Kidney Development, Institute of Molecular Embryology and Genetics, and †Department of Nephrology, Faculty of Life Sciences, Kumamoto University, Kumamoto, Japan; ‡Department of Mathematical and Life Sciences, Graduate School of Science, Hiroshima University, Hiroshima, Japan; §Division of Anatomy, Juntendo University School of Medicine, Tokyo, Japan; and |Japan Science and Technology Agency, CREST, Kumamoto, Japan ABSTRACT Glomerular podocytes express proteins, such as nephrin, that constitute the slit diaphragm, thereby contributing to the filtration process in the kidney. Glomerular development has been analyzed mainly in mice, whereas analysis of human kidney development has been minimal because of limited access to embryonic kidneys. We previously reported the induction of three-dimensional primordial glomeruli from human induced pluripotent stem (iPS) cells. Here, using transcription activator–like effector nuclease-mediated homologous recombination, we generated human iPS cell lines that express green fluorescent protein (GFP) in the NPHS1 locus, which encodes nephrin, and we show that GFP expression facilitated accurate visualization of nephrin-positive podocyte formation in
    [Show full text]
  • Calculating the Statistical Significance of Rare Variants Causal for Mendelian and Complex Disorders
    bioRxiv preprint doi: https://doi.org/10.1101/103218; this version posted January 25, 2017. 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. Calculating the statistical significance of rare variants causal for Mendelian and complex disorders Aliz R Rao1*, Stanley F Nelson1,2,3† *Correspondence: [email protected] †Email: [email protected] 1Department of Human Genetics, University of California, Los Angeles, Los Angeles, California, USA. 2Department of Psychiatry and Biobehavioral Sciences at the David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, California, USA. 3Department of Pathology and Laboratory Medicine, University of California, Los Angeles, Los Angeles, California, USA. 1 bioRxiv preprint doi: https://doi.org/10.1101/103218; this version posted January 25, 2017. 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. Abstract With the expanding use of next-gen sequencing (NGS) to diagnose the thousands of rare Mendelian genetic diseases, it is critical to be able to interpret individual DNA variation. We developed a general method to better interpret the likelihood that a rare variant is disease causing if observed in a given gene or genic region mapping to a described protein domain, using genome-wide information from a large control sample.
    [Show full text]