RNA Dysregulation in Models of Frontotemporal Dementia and Amyotrophic Lateral Sclerosis

Total Page:16

File Type:pdf, Size:1020Kb

RNA Dysregulation in Models of Frontotemporal Dementia and Amyotrophic Lateral Sclerosis University College London RNA dysregulation in models of frontotemporal dementia and amyotrophic lateral sclerosis Doctoral Thesis Author: Jack Humphrey Supervisors: Prof Adrian Isaacs Dr Vincent Plagnol Dr Pietro Fratta UCL Institute of Neurology UCL Genetics Institute I confirm that the work presented in this thesis is my own. Where information has been derived from other sources, I confirm that this has been indicated in the thesis. Jack Humphrey 3 Abstract Amyotrophic Lateral Sclerosis (ALS) and Frontotemporal dementia (FTD) are two rare but devastating neurodegenerative diseases that share pathological features and genetic factors. A central question in both diseases is the role of the RNA-binding proteins transactive response DNA-binding protein 43kDa (TDP-43) and fused in sarcoma (FUS). These proteins play a vital role in RNA regulation in all cells but in diseased neurons they alter their cellular localisation to form potentially pathogenic aggregates. This process can be linked to rare genetic mutations in the TARDBP and FUS genes, although most cases of ALS and FTD have no known genetic cause. My work uses the revolutionary technology of RNA sequencing to measure and compare gene expression and RNA splicing in different cellular and animal models of sporadic and genetic disease. Here I present the results of four studies that investigate the biology of TDP-43 and FUS, assessing both their normal cellular roles and the impact of rare disease-causing mutations. In these projects I analyse RNA sequencing data to discover novel gene expression and RNA splicing phenomena. This includes the repression of cryptic splicing by TDP-43 but not FUS, the progressive downregulation of mitochondrial and ribosomal transcripts in a mouse model of FUS ALS, a gain of splicing function by TDP-43 mutations affecting constitutive exon splicing, and widespread changes in intron retention caused by FUS knockout or aggressive FUS mutations. I also discover a novel mechanism for how FUS might regulate its own translation. This work expands on what is currently known about the roles in RNA regulation for TDP- 43 and FUS and provides new avenues for understanding both the causes and progression of ALS and FTD. 5 Impact Statement This thesis comprises four studies on TDP-43 and FUS, two proteins linked to the devas- tating neurodegenerative diseases Amyotrophic Lateral Sclerosis and Frontotemporal De- mentia. I analysed RNA sequencing data to develop insights in to the functions of these two proteins in different models of disease. This work has uncovered new roles for TDP-43 and FUS in specific types of RNA regulation. Additionally, I have discovered new mech- anisms to explain how disease-associated mutations in the two proteins affect these roles. Conclusions from my work are applicable to the RNA biology field as a whole as well as efforts to understand and treat these diseases. Insights into RNA-binding proteins and RNA regulation from this work can be transferred to many non-neurological diseases including muscular diseases, retinal diseases and certain cancers (Scotti and Swanson, 2015). Several of my chapters focus on developing new methods or adapting existing methods to analyse RNA splicing, a key mechanism of RNA regulation. RNA-sequencing is becoming a stan- dard experiment across all fields of biology and the use of sequencing data in analysis of RNA splicing is an area of extreme interest and method development. Three peer-reviewed papers have been published from this thesis. As of November 2018, “Quantitative analysis of cryptic splicing associated with TDP-43 depletion” (Humphrey et al., 2017) has been cited 11 times, “Humanized mutant FUS drives progressive motor neuron degeneration without aggregation in FUS Delta14 knockin mice” (Devoy et al., 2017) has been cited 11 times and “Mice with endogenous TDP-43 mutations exhibit gain of splicing function and characteristics of amyotrophic lateral sclerosis” (Fratta et al., 2018) has been cited 9 times and was the subject of a “News and Views” article in The EMBO Journal (Rouaux et al., 2018). These outcomes illustrate the importance of this work to the neurodegenerative disease and RNA biology fields. Three new mouse models of ALS/FTD have been generated for projects carried out in this thesis: the FUS ∆14 mouse, the TDP-43 RRM2mut mouse, and the TDP-43 LCDmut mouse. All three mice are available for labs around the world to use in further experiments. RNA-seq data created from these mice have been made publicly available and can be down- loaded from the Sequence Read Archive1. Several of my chapters rely on the re-analysis of published RNA sequencing data. I am pleased to see that sequencing data analysed in this thesis (chapter 5) has been reused in a further paper on TDP-43 (Sivakumar et al., 2018). I have tried to make all software written during this thesis publicly available where possible. This can be downloaded from the GitHub platform. This includes the RNA-seq analysis pipeline used in all chapters2, software developed to find and classify cryptic splicing (chapter 3)3, and software written to classify splicing events found in the TDP-43 LCDmut and RRM2mut mice (chapter 5)4. 1https://www.ncbi.nlm.nih.gov/sra 2https://github.com/plagnollab/RNASeq_pipeline 3https://github.com/jackhump/CryptEx 4https://github.com/jackhump/Two_TDP-43_Mutant_Mice 7 Contents 1 Introduction 15 1.1 Amyotrophic Lateral Sclerosis and Frontotemporal Dementia comprise a spec- trum of disease . 15 1.2 RNA splicing is a key step for RNA regulation . 16 1.3 RNA-sequencing is a revolutionary technology to quantify gene expression and splicing . 20 1.4 RNA-binding proteins implicated in ALS/FTD . 20 1.5 Aims of my PhD . 25 1.6 Overview of chapters . 25 2 Methods 27 2.1 Library preparation and sequencing . 27 2.2 The Plagnol lab RNA-seq pipeline . 30 2.3 Differential splicing . 32 2.4 Functional analyses . 35 2.5 Conclusions . 38 3 Cryptic splicing occurs in published TDP-43 but not FUS depletion data 39 3.1 Overview . 39 3.2 Methods . 39 3.3 Results . 44 3.4 Discussion . 54 3.5 Summary . 57 4 FUS mutant mice show progressive changes in mitochondrial and riboso- mal transcripts 58 4.1 Overview . 58 4.2 Contributions . 58 4.3 Background . 58 4.4 Methods . 59 4.5 Results . 61 4.6 Discussion . 63 5 Loss and gain of TDP-43 splicing function in two mutant mouse lines 65 5.1 Overview . 65 5.2 Contributions . 65 5.3 Methods . 66 5.4 Results . 69 5.5 Discussion . 82 9 6 ALS-causative FUS mutations impair FUS autoregulation through intron retention 85 6.1 Overview . 85 6.2 Contributions . 85 6.3 Background . 86 6.4 Methods . 88 6.5 Results . 93 6.6 Discussion . 104 7 Conclusions 109 7.1 Issues arising . 110 7.2 Future directions . 113 8 Appendices 143 8.1 Appendices to chapter 3 . 144 8.2 Appendices to chapter 4 . 149 8.3 Appendices to chapter 5 . 151 8.4 Appendices to chapter 6 . 155 List of Figures 1.1 Splicing of a U2-dependent intron . 17 1.2 Protein domains of TDP-43 and FUS . 20 2.1 Pipeline for stranded RNA-seq preparation and analysis . 29 3.1 Schematic of the Cryptex pipeline . 41 3.2 Cryptic splicing discovered by the CryptEx pipeline . 45 3.3 Evidence of TDP-43 binding cryptic exons . 47 3.4 Cryptic exons in transposable elements . 48 3.5 Conservation and premature termination codon analysis . 49 3.6 Differential expression of cryptic exon genes . 50 3.7 Scoring cryptic splice sites against canonical splice sites . 51 3.8 Mining of ENCODE eCLIP data in K562 and HepG2 cells . 53 3.9 Cryptic exons found in different ENCODE shRNA knockdowns . 56 4.1 The FUS ∆14 model . 59 4.2 Differential gene expression analysis on the ∆14 mouse . 61 4.3 Z-score heatmap of the 12 month spinal cord dataset . 62 4.4 Gene ontology categories in the 12 month spinal cord samples . 63 5.1 The two TARDBP mutations and their location within the TDP-43 protein 69 10 5.2 Opposite effects on splicing CFTR exon 9 minigene . 70 5.3 Comparing exon usage between the two mutations and a TDP-43 knockdown 71 5.4 Direction of cassette exon splicing in the two mutant lines . 71 5.5 RNA maps visualise positional enrichment of iCLIP peaks and sequence motifs 72 5.6 RRM2mut leads to cryptic exon splicing . 73 5.7 RRM2mut gene expression has bias for long gene downregulation . 74 5.8 LCDmut leads to skiptic exon splicing . 75 5.9 Permuting sample order shows clear splicing difference in LCDmut mice . 75 5.10 RNA maps of skiptic and cryptic exons show direct binding by TDP-43 . 76 5.11 Functional analyses of the skiptic exons . 77 5.12 LCDmut and TDP-43 autoregulation . 79 5.13 Skiptic exon splicing in TARDBP ALS . 80 6.1 The structure of the FUS protein and known ALS mutations . 86 6.2 Joint differential expression increases power and adjusts effect sizes . 94 6.3 Overlapping the joint differential expression models shows that FUS muta- tions affect gene expression in generally the same direction . 95 6.4 Xlr genes are upregulated in both FUS KO and FUS MUT . 96 6.5 Overlapping genes are enriched in neuronal and RNA terms . 97 6.6 Overlapping genes are enriched in neuronal and RNA terms . 98 6.7 Splicing changes strongly overlap between KO and MUT joint models . 100 6.8 Intron retention events are highly conserved and occur in RNA binding proteins101 6.9 FUS intron retention is an NMD-insensitive autoregulation mechanism .
Recommended publications
  • Gene Symbol Gene Description ACVR1B Activin a Receptor, Type IB
    Table S1. Kinase clones included in human kinase cDNA library for yeast two-hybrid screening Gene Symbol Gene Description ACVR1B activin A receptor, type IB ADCK2 aarF domain containing kinase 2 ADCK4 aarF domain containing kinase 4 AGK multiple substrate lipid kinase;MULK AK1 adenylate kinase 1 AK3 adenylate kinase 3 like 1 AK3L1 adenylate kinase 3 ALDH18A1 aldehyde dehydrogenase 18 family, member A1;ALDH18A1 ALK anaplastic lymphoma kinase (Ki-1) ALPK1 alpha-kinase 1 ALPK2 alpha-kinase 2 AMHR2 anti-Mullerian hormone receptor, type II ARAF v-raf murine sarcoma 3611 viral oncogene homolog 1 ARSG arylsulfatase G;ARSG AURKB aurora kinase B AURKC aurora kinase C BCKDK branched chain alpha-ketoacid dehydrogenase kinase BMPR1A bone morphogenetic protein receptor, type IA BMPR2 bone morphogenetic protein receptor, type II (serine/threonine kinase) BRAF v-raf murine sarcoma viral oncogene homolog B1 BRD3 bromodomain containing 3 BRD4 bromodomain containing 4 BTK Bruton agammaglobulinemia tyrosine kinase BUB1 BUB1 budding uninhibited by benzimidazoles 1 homolog (yeast) BUB1B BUB1 budding uninhibited by benzimidazoles 1 homolog beta (yeast) C9orf98 chromosome 9 open reading frame 98;C9orf98 CABC1 chaperone, ABC1 activity of bc1 complex like (S. pombe) CALM1 calmodulin 1 (phosphorylase kinase, delta) CALM2 calmodulin 2 (phosphorylase kinase, delta) CALM3 calmodulin 3 (phosphorylase kinase, delta) CAMK1 calcium/calmodulin-dependent protein kinase I CAMK2A calcium/calmodulin-dependent protein kinase (CaM kinase) II alpha CAMK2B calcium/calmodulin-dependent
    [Show full text]
  • A Genome-Wide Association Study of Bisphosphonate-Associated
    Calcifed Tissue International (2019) 105:51–67 https://doi.org/10.1007/s00223-019-00546-9 ORIGINAL RESEARCH A Genome‑Wide Association Study of Bisphosphonate‑Associated Atypical Femoral Fracture Mohammad Kharazmi1 · Karl Michaëlsson1 · Jörg Schilcher2 · Niclas Eriksson3,4 · Håkan Melhus3 · Mia Wadelius3 · Pär Hallberg3 Received: 8 January 2019 / Accepted: 8 April 2019 / Published online: 20 April 2019 © The Author(s) 2019 Abstract Atypical femoral fracture is a well-documented adverse reaction to bisphosphonates. It is strongly related to duration of bisphosphonate use, and the risk declines rapidly after drug withdrawal. The mechanism behind bisphosphonate-associated atypical femoral fracture is unclear, but a genetic predisposition has been suggested. With the aim to identify common genetic variants that could be used for preemptive genetic testing, we performed a genome-wide association study. Cases were recruited mainly through reports of adverse drug reactions sent to the Swedish Medical Products Agency on a nation- wide basis. We compared atypical femoral fracture cases (n = 51) with population-based controls (n = 4891), and to reduce the possibility of confounding by indication, we also compared with bisphosphonate-treated controls without a current diagnosis of cancer (n = 324). The total number of single-nucleotide polymorphisms after imputation was 7,585,874. A genome-wide signifcance threshold of p < 5 × 10−8 was used to correct for multiple testing. In addition, we performed candidate gene analyses for a panel of 29 genes previously implicated in atypical femoral fractures (signifcance threshold of p < 5.7 × 10−6). Compared with population controls, bisphosphonate-associated atypical femoral fracture was associated with four isolated, uncommon single-nucleotide polymorphisms.
    [Show full text]
  • HOLOHAN-DISSERTATION-2015.Pdf (6.419Mb)
    IDENTIFYING, CHARACTERIZING AND INHIBITING THE TELOMERASE REGULATORY NETWORK APPROVED BY SUPERVISORY COMMITTEE Jerry Shay, PhD Woodring Wright, MD, PhD David Corey, PhD Rolf Brekken, PhD Melanie Cobb, PhD DEDICATION Dedicated to my parents, Mark and Tracy Holohan, as well as my siblings, Kelly Nudleman and Kyle Holohan for their support and encouragement. IDENTIFYING, CHARACTERIZING AND INHIBITING THE TELOMERASE REGULATORY NETWORK by BRODY CHRISTOPHER HOLOHAN DISSERTATION / THESIS Presented to the Faculty of the Graduate School of Biomedical Sciences The University of Texas Southwestern Medical Center at Dallas In Partial Fulfillment of the Requirements For the Degree of DOCTOR OF PHILOSOPHY The University of Texas Southwestern Medical Center at Dallas Dallas, Texas May, 2015 Copyright by Brody Christopher Holohan, 2015 All Rights Reserved ACKNOWLEDGEMENTS I would like the thank my mentors, Jerry Shay and Woodring Wright for their continuous support, advice and encouragement, as well as allowing me time and resources to pursue my ideas. They are a fantastic team, and I could not have asked for better mentors. I would also like to thank my lab-mates, particularly Dr. Andrew Ludlow, Dr. Yong Zhou, Dr. Tsung-Po Lai, Dr. Oliver Delgado, Dr. Wanil Kim, Dr. Guido Stadler, Kimberly Batten, Ryan Laranger and Neal Jones. I would also like to thank Kevin Kennon for administrative support. Without my collaborators much of this would have been impossible, so I would like to thank Dr. Tim De Meyer, Dr. Abraham Aviv, Dr. Fowzan Aklayura, Dr. Steve Hunt, Dr. Tim Spector, Dr. Massimo Mangino, Dr. Daphne Friedman and Dr. Daniel Eisenberg for their help, intellectual discussions and willingness to pursue challenenging ideas.
    [Show full text]
  • Seq2pathway Vignette
    seq2pathway Vignette Bin Wang, Xinan Holly Yang, Arjun Kinstlick May 19, 2021 Contents 1 Abstract 1 2 Package Installation 2 3 runseq2pathway 2 4 Two main functions 3 4.1 seq2gene . .3 4.1.1 seq2gene flowchart . .3 4.1.2 runseq2gene inputs/parameters . .5 4.1.3 runseq2gene outputs . .8 4.2 gene2pathway . 10 4.2.1 gene2pathway flowchart . 11 4.2.2 gene2pathway test inputs/parameters . 11 4.2.3 gene2pathway test outputs . 12 5 Examples 13 5.1 ChIP-seq data analysis . 13 5.1.1 Map ChIP-seq enriched peaks to genes using runseq2gene .................... 13 5.1.2 Discover enriched GO terms using gene2pathway_test with gene scores . 15 5.1.3 Discover enriched GO terms using Fisher's Exact test without gene scores . 17 5.1.4 Add description for genes . 20 5.2 RNA-seq data analysis . 20 6 R environment session 23 1 Abstract Seq2pathway is a novel computational tool to analyze functional gene-sets (including signaling pathways) using variable next-generation sequencing data[1]. Integral to this tool are the \seq2gene" and \gene2pathway" components in series that infer a quantitative pathway-level profile for each sample. The seq2gene function assigns phenotype-associated significance of genomic regions to gene-level scores, where the significance could be p-values of SNPs or point mutations, protein-binding affinity, or transcriptional expression level. The seq2gene function has the feasibility to assign non-exon regions to a range of neighboring genes besides the nearest one, thus facilitating the study of functional non-coding elements[2]. Then the gene2pathway summarizes gene-level measurements to pathway-level scores, comparing the quantity of significance for gene members within a pathway with those outside a pathway.
    [Show full text]
  • Autism Multiplex Family with 16P11.2P12.2 Microduplication Syndrome in Monozygotic Twins and Distal 16P11.2 Deletion in Their Brother
    European Journal of Human Genetics (2012) 20, 540–546 & 2012 Macmillan Publishers Limited All rights reserved 1018-4813/12 www.nature.com/ejhg ARTICLE Autism multiplex family with 16p11.2p12.2 microduplication syndrome in monozygotic twins and distal 16p11.2 deletion in their brother Anne-Claude Tabet1,2,3,4, Marion Pilorge2,3,4, Richard Delorme5,6,Fre´de´rique Amsellem5,6, Jean-Marc Pinard7, Marion Leboyer6,8,9, Alain Verloes10, Brigitte Benzacken1,11,12 and Catalina Betancur*,2,3,4 The pericentromeric region of chromosome 16p is rich in segmental duplications that predispose to rearrangements through non-allelic homologous recombination. Several recurrent copy number variations have been described recently in chromosome 16p. 16p11.2 rearrangements (29.5–30.1 Mb) are associated with autism, intellectual disability (ID) and other neurodevelopmental disorders. Another recognizable but less common microdeletion syndrome in 16p11.2p12.2 (21.4 to 28.5–30.1 Mb) has been described in six individuals with ID, whereas apparently reciprocal duplications, studied by standard cytogenetic and fluorescence in situ hybridization techniques, have been reported in three patients with autism spectrum disorders. Here, we report a multiplex family with three boys affected with autism, including two monozygotic twins carrying a de novo 16p11.2p12.2 duplication of 8.95 Mb (21.28–30.23 Mb) characterized by single-nucleotide polymorphism array, encompassing both the 16p11.2 and 16p11.2p12.2 regions. The twins exhibited autism, severe ID, and dysmorphic features, including a triangular face, deep-set eyes, large and prominent nasal bridge, and tall, slender build. The eldest brother presented with autism, mild ID, early-onset obesity and normal craniofacial features, and carried a smaller, overlapping 16p11.2 microdeletion of 847 kb (28.40–29.25 Mb), inherited from his apparently healthy father.
    [Show full text]
  • Figure S1. DMD Module Network. the Network Is Formed by 260 Genes from Disgenet and 1101 Interactions from STRING. Red Nodes Are the Five Seed Candidate Genes
    Figure S1. DMD module network. The network is formed by 260 genes from DisGeNET and 1101 interactions from STRING. Red nodes are the five seed candidate genes. Figure S2. DMD module network is more connected than a random module of the same size. It is shown the distribution of the largest connected component of 10.000 random modules of the same size of the DMD module network. The green line (x=260) represents the DMD largest connected component, obtaining a z-score=8.9. Figure S3. Shared genes between BMD and DMD signature. A) A meta-analysis of three microarray datasets (GSE3307, GSE13608 and GSE109178) was performed for the identification of differentially expressed genes (DEGs) in BMD muscle biopsies as compared to normal muscle biopsies. Briefly, the GSE13608 dataset included 6 samples of skeletal muscle biopsy from healthy people and 5 samples from BMD patients. Biopsies were taken from either biceps brachii, triceps brachii or deltoid. The GSE3307 dataset included 17 samples of skeletal muscle biopsy from healthy people and 10 samples from BMD patients. The GSE109178 dataset included 14 samples of controls and 11 samples from BMD patients. For both GSE3307 and GSE10917 datasets, biopsies were taken at the time of diagnosis and from the vastus lateralis. For the meta-analysis of GSE13608, GSE3307 and GSE109178, a random effects model of effect size measure was used to integrate gene expression patterns from the two datasets. Genes with an adjusted p value (FDR) < 0.05 and an │effect size│>2 were identified as DEGs and selected for further analysis. A significant number of DEGs (p<0.001) were in common with the DMD signature genes (blue nodes), as determined by a hypergeometric test assessing the significance of the overlap between the BMD DEGs and the number of DMD signature genes B) MCODE analysis of the overlapping genes between BMD DEGs and DMD signature genes.
    [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]
  • Supplementary Table S1. Prioritization of Candidate FPC Susceptibility Genes by Private Heterozygous Ptvs
    Supplementary Table S1. Prioritization of candidate FPC susceptibility genes by private heterozygous PTVs Number of private Number of private Number FPC patient heterozygous PTVs in heterozygous PTVs in tumors with somatic FPC susceptibility Hereditary cancer Hereditary Gene FPC kindred BCCS samples mutation DNA repair gene Cancer driver gene gene gene pancreatitis gene ATM 19 1 - Yes Yes Yes Yes - SSPO 12 8 1 - - - - - DNAH14 10 3 - - - - - - CD36 9 3 - - - - - - TET2 9 1 - - Yes - - - MUC16 8 14 - - - - - - DNHD1 7 4 1 - - - - - DNMT3A 7 1 - - Yes - - - PKHD1L1 7 9 - - - - - - DNAH3 6 5 - - - - - - MYH7B 6 1 - - - - - - PKD1L2 6 6 - - - - - - POLN 6 2 - Yes - - - - POLQ 6 7 - Yes - - - - RP1L1 6 6 - - - - - - TTN 6 5 4 - - - - - WDR87 6 7 - - - - - - ABCA13 5 3 1 - - - - - ASXL1 5 1 - - Yes - - - BBS10 5 0 - - - - - - BRCA2 5 6 1 Yes Yes Yes Yes - CENPJ 5 1 - - - - - - CEP290 5 5 - - - - - - CYP3A5 5 2 - - - - - - DNAH12 5 6 - - - - - - DNAH6 5 1 1 - - - - - EPPK1 5 4 - - - - - - ESYT3 5 1 - - - - - - FRAS1 5 4 - - - - - - HGC6.3 5 0 - - - - - - IGFN1 5 5 - - - - - - KCP 5 4 - - - - - - LRRC43 5 0 - - - - - - MCTP2 5 1 - - - - - - MPO 5 1 - - - - - - MUC4 5 5 - - - - - - OBSCN 5 8 2 - - - - - PALB2 5 0 - Yes - Yes Yes - SLCO1B3 5 2 - - - - - - SYT15 5 3 - - - - - - XIRP2 5 3 1 - - - - - ZNF266 5 2 - - - - - - ZNF530 5 1 - - - - - - ACACB 4 1 1 - - - - - ALS2CL 4 2 - - - - - - AMER3 4 0 2 - - - - - ANKRD35 4 4 - - - - - - ATP10B 4 1 - - - - - - ATP8B3 4 6 - - - - - - C10orf95 4 0 - - - - - - C2orf88 4 0 - - - - - - C5orf42 4 2 - - - -
    [Show full text]
  • Transcriptome Sequencing and Genome-Wide Association Analyses Reveal Lysosomal Function and Actin Cytoskeleton Remodeling in Schizophrenia and Bipolar Disorder
    Molecular Psychiatry (2015) 20, 563–572 © 2015 Macmillan Publishers Limited All rights reserved 1359-4184/15 www.nature.com/mp ORIGINAL ARTICLE Transcriptome sequencing and genome-wide association analyses reveal lysosomal function and actin cytoskeleton remodeling in schizophrenia and bipolar disorder Z Zhao1,6,JXu2,6, J Chen3,6, S Kim4, M Reimers3, S-A Bacanu3,HYu1, C Liu5, J Sun1, Q Wang1, P Jia1,FXu2, Y Zhang2, KS Kendler3, Z Peng2 and X Chen3 Schizophrenia (SCZ) and bipolar disorder (BPD) are severe mental disorders with high heritability. Clinicians have long noticed the similarities of clinic symptoms between these disorders. In recent years, accumulating evidence indicates some shared genetic liabilities. However, what is shared remains elusive. In this study, we conducted whole transcriptome analysis of post-mortem brain tissues (cingulate cortex) from SCZ, BPD and control subjects, and identified differentially expressed genes in these disorders. We found 105 and 153 genes differentially expressed in SCZ and BPD, respectively. By comparing the t-test scores, we found that many of the genes differentially expressed in SCZ and BPD are concordant in their expression level (q ⩽ 0.01, 53 genes; q ⩽ 0.05, 213 genes; q ⩽ 0.1, 885 genes). Using genome-wide association data from the Psychiatric Genomics Consortium, we found that these differentially and concordantly expressed genes were enriched in association signals for both SCZ (Po10 − 7) and BPD (P = 0.029). To our knowledge, this is the first time that a substantially large number of genes show concordant expression and association for both SCZ and BPD. Pathway analyses of these genes indicated that they are involved in the lysosome, Fc gamma receptor-mediated phagocytosis, regulation of actin cytoskeleton pathways, along with several cancer pathways.
    [Show full text]
  • Supplemental Information
    Supplemental information Dissection of the genomic structure of the miR-183/96/182 gene. Previously, we showed that the miR-183/96/182 cluster is an intergenic miRNA cluster, located in a ~60-kb interval between the genes encoding nuclear respiratory factor-1 (Nrf1) and ubiquitin-conjugating enzyme E2H (Ube2h) on mouse chr6qA3.3 (1). To start to uncover the genomic structure of the miR- 183/96/182 gene, we first studied genomic features around miR-183/96/182 in the UCSC genome browser (http://genome.UCSC.edu/), and identified two CpG islands 3.4-6.5 kb 5’ of pre-miR-183, the most 5’ miRNA of the cluster (Fig. 1A; Fig. S1 and Seq. S1). A cDNA clone, AK044220, located at 3.2-4.6 kb 5’ to pre-miR-183, encompasses the second CpG island (Fig. 1A; Fig. S1). We hypothesized that this cDNA clone was derived from 5’ exon(s) of the primary transcript of the miR-183/96/182 gene, as CpG islands are often associated with promoters (2). Supporting this hypothesis, multiple expressed sequences detected by gene-trap clones, including clone D016D06 (3, 4), were co-localized with the cDNA clone AK044220 (Fig. 1A; Fig. S1). Clone D016D06, deposited by the German GeneTrap Consortium (GGTC) (http://tikus.gsf.de) (3, 4), was derived from insertion of a retroviral construct, rFlpROSAβgeo in 129S2 ES cells (Fig. 1A and C). The rFlpROSAβgeo construct carries a promoterless reporter gene, the β−geo cassette - an in-frame fusion of the β-galactosidase and neomycin resistance (Neor) gene (5), with a splicing acceptor (SA) immediately upstream, and a polyA signal downstream of the β−geo cassette (Fig.
    [Show full text]
  • Noelia Díaz Blanco
    Effects of environmental factors on the gonadal transcriptome of European sea bass (Dicentrarchus labrax), juvenile growth and sex ratios Noelia Díaz Blanco Ph.D. thesis 2014 Submitted in partial fulfillment of the requirements for the Ph.D. degree from the Universitat Pompeu Fabra (UPF). This work has been carried out at the Group of Biology of Reproduction (GBR), at the Department of Renewable Marine Resources of the Institute of Marine Sciences (ICM-CSIC). Thesis supervisor: Dr. Francesc Piferrer Professor d’Investigació Institut de Ciències del Mar (ICM-CSIC) i ii A mis padres A Xavi iii iv Acknowledgements This thesis has been made possible by the support of many people who in one way or another, many times unknowingly, gave me the strength to overcome this "long and winding road". First of all, I would like to thank my supervisor, Dr. Francesc Piferrer, for his patience, guidance and wise advice throughout all this Ph.D. experience. But above all, for the trust he placed on me almost seven years ago when he offered me the opportunity to be part of his team. Thanks also for teaching me how to question always everything, for sharing with me your enthusiasm for science and for giving me the opportunity of learning from you by participating in many projects, collaborations and scientific meetings. I am also thankful to my colleagues (former and present Group of Biology of Reproduction members) for your support and encouragement throughout this journey. To the “exGBRs”, thanks for helping me with my first steps into this world. Working as an undergrad with you Dr.
    [Show full text]
  • Telomeres, Aging and Exercise: Guilty by Association?
    View metadata, citation and similar papers at core.ac.uk brought to you by CORE provided by Federation ResearchOnline COPYRIGHT NOTICE FedUni ResearchOnline https://researchonline.federation.edu.au This is the published version of: Chilton, W., et.al. (2017) Telomeres, aging and exercise: Guilty by association. International Journal of Molecular Sciences, 18(12), p.1-32. Available online at https://doi.org/10.3390/ijms18122573 Copyright © 2017 Chilton, W., et.al. This is an open-access article distributed under the terms of the Creative Commons Attribution License (CC BY 4.0) (http://creativecommons.org/licenses/by/4.0/). The use, distribution or reproduction in other forums is permitted, provided the original author(s) or licensor are credited and that the original publication in this journal is cited, in accordance with accepted academic practice. No use, distribution or reproduction is permitted which does not comply with these terms. International Journal of Molecular Sciences Review Telomeres, Aging and Exercise: Guilty by Association? Warrick Chilton 1,*, Brendan O’Brien 1 and Fadi Charchar 1,2,3,* ID 1 Faculty of Health Sciences and Faculty of Science and Technology, Federation University Australia, Ballarat, VIC 3350, Australia; [email protected] 2 Department of Physiology, University of Melbourne, Melbourne, VIC 3010, Australia 3 Department of Cardiovascular Sciences, University of Leicester, Leicester LE1 7RH, UK * Correspondence: [email protected] (W.C.); [email protected] (F.C.); Tel.: +61-3-5327-6254 (W.C.); +61-3-5327-6098 (F.C.) Received: 7 September 2017; Accepted: 25 November 2017; Published: 29 November 2017 Abstract: Telomeres are repetitive tandem DNA sequences that cap chromosomal ends protecting genomic DNA from enzymatic degradation.
    [Show full text]