FARE2021WINNERS Sorted by Institute

Total Page:16

File Type:pdf, Size:1020Kb

FARE2021WINNERS Sorted by Institute FARE2021WINNERS Sorted By Institute Swati Shah Postdoctoral Fellow CC Radiology/Imaging/PET and Neuroimaging Characterization of CNS involvement in Ebola-Infected Macaques using Magnetic Resonance Imaging, 18F-FDG PET and Immunohistology The Ebola (EBOV) virus outbreak in Western Africa resulted in residual neurologic abnormalities in survivors. Many case studies detected EBOV in the CSF, suggesting that the neurologic sequelae in survivors is related to viral presence. In the periphery, EBOV infects endothelial cells and triggers a “cytokine stormâ€. However, it is unclear whether a similar process occurs in the brain, with secondary neuroinflammation, neuronal loss and blood-brain barrier (BBB) compromise, eventually leading to lasting neurological damage. We have used in vivo imaging and post-necropsy immunostaining to elucidate the CNS pathophysiology in Rhesus macaques infected with EBOV (Makona). Whole brain MRI with T1 relaxometry (pre- and post-contrast) and FDG-PET were performed to monitor the progression of disease in two cohorts of EBOV infected macaques from baseline to terminal endpoint (day 5-6). Post-necropsy, multiplex fluorescence immunohistochemical (MF-IHC) staining for various cellular markers in the thalamus and brainstem was performed. Serial blood and CSF samples were collected to assess disease progression. The linear mixed effect model was used for statistical analysis. Post-infection, we first detected EBOV in the serum (day 3) and CSF (day 4) with dramatic increases until euthanasia. The standard uptake values of FDG-PET relative to whole brain uptake (SUVr) in the midbrain, pons, and thalamus increased significantly over time (p<0.01) and positively correlated with blood viremia (p≤0.01). On MF-IHC, EBOV antigen (VP40) co-localized with endothelial cells as expected, but also with neurons (NeuN), which is consistent with direct neuronal infection and has not been shown before. Many of the infected neurons were apoptotic (cleaved caspase-3/PARP1 staining co-localizing with NeuN and VP40) and showed increased expression of glucose transporters (mainly GLUT-3). Elevated GLUT-3 levels could explain the increased FDG uptake in these regions. On MRI, significant increases in pre-contrast T1-relaxation times with decreased post contrast T1 values suggested BBB disruption and leakage. This was confirmed by MF-IHC showing extravascular albumin staining. Using imaging and histopathology, we showed that acute EBOV infection in macaques leads to BBB disruption, neuronal infection, apoptosis and increased GLUT3 expression. Our findings shed light on the pathophysiology of CNS involvement in EBOV that could explain residual neurologic manifestations in survivors. __________________________________________________________________________________ Yen-Ting Tung Postdoctoral Fellow NCATS Biophysics and Biomedical Engineering A Patient-derived Glioblastoma Model in a Three Dimensional Bioprinted Neurovascular Unit tissue: Moving towards in Tissue Personalized Medicine Glioblastoma (GBM) is a fast-growing, aggressive brain cancer with high heterogeneity of subpopulations. The prognosis for patients with high-grade GBM remains extremely poor, thus, there is a tremendous medical need for more effective treatments. Using three dimensional (3D) biofabrication technologies, including tissue bioprinting, we have successfully established a 3D neurovascular unit (NVU) tissue in a multiwell plate format. The biofabricated NVU includes human GFP-tagged brain endothelial cells, brain pericytes, and astrocytes. A ring-shaped bioprinted structure of vasculature with an inner circular-shape drop of mCherry expressing GBMCs was created in each well to facilitate the quantification of vascular morphology and GBM invasion and growth, respectively. Time dependent vascular angiogenesis in the presence or absence of GBMCs was monitored using a high content fluorescence imaging system. In the tissues without the inclusion of the GBMCs, we confirmed vasculogenic and angiogenic activities occurring for two weeks after bioprinting. The total length of angiogenetic vessel gradually increased with time, along with deposition of brain relevant extra cellular matrix components, like collagen IV, laminin, and fibronectin, demonstrating formation of a 3D neurovascular unit. When introducing the GBMCs into this NVU model, we observed cancer cell infiltration into the outer ring vascular area through the branched vessels. Moreover, the microvascular structure in contact with the GBMCs area collapsed, even though the endothelial cells remained viable and proliferated inside the GBM area. The GBM-microvessel interaction observed is similar to that described in human GBM pathological condition, which showed an abnormal and collapsed microvascular morphology. The single cell RNA sequencing and anti-cancer drug treatments are now being used to study changes in cell populations in the tissue as well as finding potential druggable targets driving cancer cell growth in these tissue models. For the future work, neuronal cells will be included in the tissue model to achieve a better physiological relevance of the neurovascular tissue. This GBM in-a-tissue model will ultimately provide extensive opportunities to investigate patient-specific pathology and to find a personalized drugs for patients. __________________________________________________________________________________ Vishal Babu Siramshetty Postdoctoral Fellow NCATS Chemistry Machine Learning versus Artificial Intelligence Methods for Predicting Cardiotoxicity in the ‘Big Data’ Era Cardiotoxicity is a leading concern behind the recall of marketed drugs and failure of clinical candidates. Inhibition of the potassium ion channel, whose alpha subunit is encoded by human Ether-à-go-go- Related Gene, leads to prolonged QT interval in the cardiac action potential. Fatal cases of cardiac arrhythmia have been reported due to drug-induced hERG channel blockade. Several computational methods, including machine learning, have been employed in modeling this endpoint. Reports based on artificial intelligence methods have only recently surfaced in the literature. To this end, we performed a comprehensive comparison of classification models developed using the classical machine learning and modern deep learning methods by employing different molecular descriptors. The training set (~9000 compounds) was constructed by combining hERG bioactivity data from ChEMBL database with data generated in an in-house high-throughput ion flux assay. Prospective validation of the models was performed on ~1700 in-house compounds screened for hERG activity in the same flux assay. We noticed that the deep learning models performed on par with the baseline models (Random Forests and Gradient Boosting). Latent descriptors derived directly from the deep learning architectures provided nearly the same performance as traditional molecular descriptors. We compared our models with other neural network architectures and state-of-the-art models from literature. Our models outperformed these models on the validation data. The study highlights the application of artificial intelligence methods on chemistry big data, readily available from public domain compound repositories, for generation of novel descriptors that are promising for molecular property prediction. __________________________________________________________________________________ Travis Kinder Postdoctoral Fellow NCATS Immunology - Autoimmune Quantitative High Throughput Screening to Repurpose Drugs to Treat Myositis Idiopathic inflammatory myopathies, collectively referred to as myositis, are a group of rare autoimmune diseases characterized by skeletal muscle inflammation, weakness, and often pathologies of the skin, lungs, or vasculature. Current treatment with steroids and immunosuppressive agents are not curative and have serious side-effects. We initiated a drug repurposing program to identify more targeted, less toxic therapeutics to treat myositis. Repurposing is the quickest path for drug development because it involves identifying approved medications currently used to treat other conditions. Although the cause of myositis is not fully understood, muscle from patients have an overabundance of the inflammatory cytokine type 1 interferon (IFN) and a down-stream product major histocompatibility complex (MHC) class I, which targets T cells for cytotoxic attack of host tissue. Genome-wide association studies in myositis have confirmed that alleles of MHC class I are the strongest risk factor, the protein is up-regulated in patient muscles, and forced over-expression in mouse skeletal muscle leads to many features of myositis. We conducted quantitative high throughput screening of >15,000 compound titrations, including all approved drugs, through a series of cell-based assays to identify those that inhibit the IFN-beta stimulated expression of MHC class I in muscle precursor cells (myoblasts). The primary screen utilized CRISPR/Cas9 genome-engineered human myoblasts that contained a pro-luminescent reporter HiBit fused to the C-terminus of endogenous MHC class I. Active compounds were counter-screened for cytotoxicity and validated by MCH class I immunofluorescence, Western blot, and RT-qPCR. All assays were optimized to have Z’-factors >0.5, a statistical measure of assay robustness. Sixty-seven active compounds were identified falling into two major pharmacological mechanisms, Janus kinase (JAK) inhibitors and epigenetic/transcription factor modulators.
Recommended publications
  • Large-Scale Analysis of Genome and Transcriptome Alterations in Multiple Tumors Unveils Novel Cancer-Relevant Splicing Networks
    Downloaded from genome.cshlp.org on September 28, 2021 - Published by Cold Spring Harbor Laboratory Press Research Large-scale analysis of genome and transcriptome alterations in multiple tumors unveils novel cancer-relevant splicing networks Endre Sebestyén,1,5 Babita Singh,1,5 Belén Miñana,1,2 Amadís Pagès,1 Francesca Mateo,3 Miguel Angel Pujana,3 Juan Valcárcel,1,2,4 and Eduardo Eyras1,4 1Universitat Pompeu Fabra, E08003 Barcelona, Spain; 2Centre for Genomic Regulation, E08003 Barcelona, Spain; 3Program Against Cancer Therapeutic Resistance (ProCURE), Catalan Institute of Oncology (ICO), Bellvitge Institute for Biomedical Research (IDIBELL), E08908 L’Hospitalet del Llobregat, Spain; 4Catalan Institution for Research and Advanced Studies, E08010 Barcelona, Spain Alternative splicing is regulated by multiple RNA-binding proteins and influences the expression of most eukaryotic genes. However, the role of this process in human disease, and particularly in cancer, is only starting to be unveiled. We system- atically analyzed mutation, copy number, and gene expression patterns of 1348 RNA-binding protein (RBP) genes in 11 solid tumor types, together with alternative splicing changes in these tumors and the enrichment of binding motifs in the alter- natively spliced sequences. Our comprehensive study reveals widespread alterations in the expression of RBP genes, as well as novel mutations and copy number variations in association with multiple alternative splicing changes in cancer drivers and oncogenic pathways. Remarkably, the altered splicing patterns in several tumor types recapitulate those of undifferen- tiated cells. These patterns are predicted to be mainly controlled by MBNL1 and involve multiple cancer drivers, including the mitotic gene NUMA1. We show that NUMA1 alternative splicing induces enhanced cell proliferation and centrosome am- plification in nontumorigenic mammary epithelial cells.
    [Show full text]
  • Compaction and Condensation of DNA Mediated by the C-Terminal Domain Of
    i “CTR-NAR” — 2017/5/3 — 11:51 — page 1 — #1 i i i Published online 03/05/2017 Nucleic Acids Research, 2017, Vol. 00, No. 00 1–10 doi:10.1093/nar/gkn000 Compaction and condensation of DNA mediated by the C-terminal domain of Hfq 1, 2, 1,3 4 Antoine Malabirade †, Kai Jiang †, Krzysztof Kubiak , Alvaro Diaz-Mendoza , Fan Liu 2, Jeroen A. van Kan 2, Jean-François Berret 4, 1,4, 2, Véronique Arluison ú, and Johan R.C. van der Maarel ú 1Laboratoire Léon Brillouin, CEA, CNRS, Université Paris Saclay, 91191 Gif-sur-Yvette, France; 2Department of Physics, National University of Singapore, 2 Science Drive 3, Singapore 117542; 3Department of Molecular Biology, University of Gdansk, Wita Stwosza 59, 80-308 Gdansk, Poland; 4Université Paris Diderot, Sorbonne Paris Cité, 75013 Paris, France. ABSTRACT it is usually needed to mediate post-transcriptional stress-response with small noncoding RNA (sRNA). This Hfq is a bacterial protein that is involved in several regulatory mechanism is based on the hybridisation of aspects of nucleic acids metabolism. It has been sRNA to its target mRNA. Accordingly, the majority described as one of the nucleoid associated proteins of known sRNA interacts nearby the ribosome binding shaping the bacterial chromosome, although it is site to prevent initiation of translation and thus favours better known to influence translation and turnover mRNA decay (4, 5, 6). Few studies have shed light of cellular RNAs. Here, we explore the role of on the importance of Hfq in DNA metabolism. It has Escherichia coli Hfq’s C-terminal domain in the been suggested that Hfq affects plasmid replication and compaction of double stranded DNA.
    [Show full text]
  • Single Cell Transcriptomics Reveal Temporal Dynamics of Critical Regulators of Germ Cell Fate During Mouse Sex Determination
    bioRxiv preprint doi: https://doi.org/10.1101/747279; this version posted November 2, 2020. The copyright holder for this preprint (which was not certified by peer review) is the author/funder, who has granted bioRxiv a license to display the preprint in perpetuity. It is made available under aCC-BY-NC-ND 4.0 International license. 1 Single cell transcriptomics reveal temporal dynamics of critical regulators of germ 2 cell fate during mouse sex determination 3 Authors: Chloé Mayère1,2, Yasmine Neirijnck1,3, Pauline Sararols1, Chris M Rands1, 4 Isabelle Stévant1,2, Françoise Kühne1, Anne-Amandine Chassot3, Marie-Christine 5 Chaboissier3, Emmanouil T. Dermitzakis1,2, Serge Nef1,2,*. 6 Affiliations: 7 1Department of Genetic Medicine and Development, University of Geneva, 1211 Geneva, 8 Switzerland; 9 2iGE3, Institute of Genetics and Genomics of Geneva, University of Geneva, 1211 10 Geneva, Switzerland; 11 3Université Côte d'Azur, CNRS, Inserm, iBV, France; 12 Lead Contact: 13 *Corresponding Author: Serge Nef, 1 rue Michel-Servet CH-1211 Genève 4, 14 [email protected]. + 41 (0)22 379 51 93 15 Running Title: Single cell transcriptomics of germ cells 1 bioRxiv preprint doi: https://doi.org/10.1101/747279; this version posted November 2, 2020. The copyright holder for this preprint (which was not certified by peer review) is the author/funder, who has granted bioRxiv a license to display the preprint in perpetuity. It is made available under aCC-BY-NC-ND 4.0 International license. 16 Abbreviations; 17 AGC: Adrenal Germ Cell 18 GC: Germ cell 19 OGC: Ovarian Germ Cell 20 TGC: Testicular Germ Cell 21 scRNA-seq: Single-cell RNA-Sequencing 22 DEG: Differentially Expressed Gene 23 24 25 Keywords: 26 Single-cell RNA-Sequencing (scRNA-seq), sex determination, ovary, testis, gonocytes, 27 oocytes, prospermatogonia, meiosis, gene regulatory network, germ cells, development, 28 RNA splicing 29 2 bioRxiv preprint doi: https://doi.org/10.1101/747279; this version posted November 2, 2020.
    [Show full text]
  • Multifactorial Erβ and NOTCH1 Control of Squamous Differentiation and Cancer
    Multifactorial ERβ and NOTCH1 control of squamous differentiation and cancer Yang Sui Brooks, … , Karine Lefort, G. Paolo Dotto J Clin Invest. 2014;124(5):2260-2276. https://doi.org/10.1172/JCI72718. Research Article Oncology Downmodulation or loss-of-function mutations of the gene encoding NOTCH1 are associated with dysfunctional squamous cell differentiation and development of squamous cell carcinoma (SCC) in skin and internal organs. While NOTCH1 receptor activation has been well characterized, little is known about how NOTCH1 gene transcription is regulated. Using bioinformatics and functional screening approaches, we identified several regulators of the NOTCH1 gene in keratinocytes, with the transcription factors DLX5 and EGR3 and estrogen receptor β (ERβ) directly controlling its expression in differentiation. DLX5 and ERG3 are required for RNA polymerase II (PolII) recruitment to the NOTCH1 locus, while ERβ controls NOTCH1 transcription through RNA PolII pause release. Expression of several identified NOTCH1 regulators, including ERβ, is frequently compromised in skin, head and neck, and lung SCCs and SCC-derived cell lines. Furthermore, a keratinocyte ERβ–dependent program of gene expression is subverted in SCCs from various body sites, and there are consistent differences in mutation and gene-expression signatures of head and neck and lung SCCs in female versus male patients. Experimentally increased ERβ expression or treatment with ERβ agonists inhibited proliferation of SCC cells and promoted NOTCH1 expression and squamous differentiation both in vitro and in mouse xenotransplants. Our data identify a link between transcriptional control of NOTCH1 expression and the estrogen response in keratinocytes, with implications for differentiation therapy of squamous cancer. Find the latest version: https://jci.me/72718/pdf Research article Multifactorial ERβ and NOTCH1 control of squamous differentiation and cancer Yang Sui Brooks,1,2 Paola Ostano,3 Seung-Hee Jo,1,2 Jun Dai,1,2 Spiro Getsios,4 Piotr Dziunycz,5 Günther F.L.
    [Show full text]
  • Table S1 the Four Gene Sets Derived from Gene Expression Profiles of Escs and Differentiated Cells
    Table S1 The four gene sets derived from gene expression profiles of ESCs and differentiated cells Uniform High Uniform Low ES Up ES Down EntrezID GeneSymbol EntrezID GeneSymbol EntrezID GeneSymbol EntrezID GeneSymbol 269261 Rpl12 11354 Abpa 68239 Krt42 15132 Hbb-bh1 67891 Rpl4 11537 Cfd 26380 Esrrb 15126 Hba-x 55949 Eef1b2 11698 Ambn 73703 Dppa2 15111 Hand2 18148 Npm1 11730 Ang3 67374 Jam2 65255 Asb4 67427 Rps20 11731 Ang2 22702 Zfp42 17292 Mesp1 15481 Hspa8 11807 Apoa2 58865 Tdh 19737 Rgs5 100041686 LOC100041686 11814 Apoc3 26388 Ifi202b 225518 Prdm6 11983 Atpif1 11945 Atp4b 11614 Nr0b1 20378 Frzb 19241 Tmsb4x 12007 Azgp1 76815 Calcoco2 12767 Cxcr4 20116 Rps8 12044 Bcl2a1a 219132 D14Ertd668e 103889 Hoxb2 20103 Rps5 12047 Bcl2a1d 381411 Gm1967 17701 Msx1 14694 Gnb2l1 12049 Bcl2l10 20899 Stra8 23796 Aplnr 19941 Rpl26 12096 Bglap1 78625 1700061G19Rik 12627 Cfc1 12070 Ngfrap1 12097 Bglap2 21816 Tgm1 12622 Cer1 19989 Rpl7 12267 C3ar1 67405 Nts 21385 Tbx2 19896 Rpl10a 12279 C9 435337 EG435337 56720 Tdo2 20044 Rps14 12391 Cav3 545913 Zscan4d 16869 Lhx1 19175 Psmb6 12409 Cbr2 244448 Triml1 22253 Unc5c 22627 Ywhae 12477 Ctla4 69134 2200001I15Rik 14174 Fgf3 19951 Rpl32 12523 Cd84 66065 Hsd17b14 16542 Kdr 66152 1110020P15Rik 12524 Cd86 81879 Tcfcp2l1 15122 Hba-a1 66489 Rpl35 12640 Cga 17907 Mylpf 15414 Hoxb6 15519 Hsp90aa1 12642 Ch25h 26424 Nr5a2 210530 Leprel1 66483 Rpl36al 12655 Chi3l3 83560 Tex14 12338 Capn6 27370 Rps26 12796 Camp 17450 Morc1 20671 Sox17 66576 Uqcrh 12869 Cox8b 79455 Pdcl2 20613 Snai1 22154 Tubb5 12959 Cryba4 231821 Centa1 17897
    [Show full text]
  • Low Basal Expression and Slow Induction of IFITM3 Puts Immune
    bioRxiv preprint doi: https://doi.org/10.1101/2019.12.20.885590; this version posted December 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. 1 TITLE: Low basal expression and slow induction of IFITM3 puts immune 2 cells at risk of influenza A infection 3 4 Running head (40 characters): 5 Cells at risk of Influenza A infection 6 7 Authors: 8 Dannielle Wellington1,2,*, , Zixi Yin1,2, Liwei Zhang1, Jessica Forbester1,3, Kerry 9 Kite1, Henry Laurenson-Schafer1, Shokouh Makvandi-Nejad1 , Boquan Jin4, 10 Emma Bowes5, Krishnageetha Manoharan5, David Maldonado-Perez5, Clare 11 Verill5, Ian Humphreys3 & Tao Dong1,2,* 12 1. MRC Human Immunology Unit, MRC Weatherall Institute of Molecular Medicine, Radcliffe 13 Department of Medicine, Oxford University, Oxford OX3 9DS, UK 14 2. Chinese Academy of Medical Sciences (CAMS) Oxford Institute, Nuffield Department of 15 Medicine, Oxford University, OX3 7BN, UK 16 3. Division of Infection and Immunity/Systems Immunity University Research Institute, Cardiff 17 University, Cardiff, CF14 4XN, UK 18 4. Fourth Military Medical University, Xian, China 19 5. Oxford Radcliffe BioBank, Nuffield Department of Surgical Sciences, Oxford University, 20 Oxford OX3 9DU, UK 21 *Corresponding to Dannielle Wellington [email protected] or Tao Dong, 22 [email protected] 23 24 Acknowledgements: 25 The authors would like to thank the following facilities and individuals within 26 the MRC Weatherall Institute (Oxford) for their invaluaBle help in the following 27 experiments: Mass Cytometry Facility and Alain Townsend for kindly providing 28 virus stocks.
    [Show full text]
  • I LITERATURE-BASED DISCOVERY of KNOWN and POTENTIAL NEW
    LITERATURE-BASED DISCOVERY OF KNOWN AND POTENTIAL NEW MECHANISMS FOR RELATING THE STATUS OF CHOLESTEROL TO THE PROGRESSION OF BREAST CANCER BY YU WANG THESIS Submitted in partial fulfillment of the requirements for the degree of Master of Science in Bioinformatics with a concentration in Library and Information Science in the Graduate College of the University of Illinois at Urbana-Champaign, 2019 Urbana, Illinois Adviser: Professor Vetle I. Torvik Professor Erik Russell Nelson i ABSTRACT Breast cancer has been studied for a long period of time and from a variety of perspectives in order to understand its pathogeny. The pathogeny of breast cancer can be classified into two groups: hereditary and spontaneous. Although cancer in general is considered a genetic disease, spontaneous factors are responsible for most of the pathogeny of breast cancer. In other words, breast cancer is more likely to be caused and deteriorated by the dysfunction of a physical molecule than be caused by germline mutation directly. Interestingly, cholesterol, as one of those molecules, has been discovered to correlate with breast cancer risk. However, the mechanisms of how cholesterol helps breast cancer progression are not thoroughly understood. As a result, this study aims to study known and discover potential new mechanisms regarding to the correlation of cholesterol and breast cancer progression using literature review and literature-based discovery. The known mechanisms are further classified into four groups: cholesterol membrane content, transport of cholesterol, cholesterol metabolites, and other. The potential mechanisms, which are intended to provide potential new treatments, have been identified and checked for feasibility by an expert.
    [Show full text]
  • Producing Hfq/Sm Proteins and Srnas for Structural and Biophysical Studies of Ribonucleoprotein Assembly
    bioRxiv preprint doi: https://doi.org/10.1101/150672; this version posted June 16, 2017. The copyright holder for this preprint (which was not certified by peer review) is the author/funder. All rights reserved. No reuse allowed without permission. Producing Hfq/Sm Proteins and sRNAs for Structural and Biophysical Studies of Ribonucleoprotein Assembly Kimberly A. Stanek* and Cameron Mura* Author affiliations & correspondence: Department of Chemistry; University of Virginia; Charlottesville, VA 22904 USA *Correspondence can be addressed to KS ([email protected]) or CM ([email protected]). Document information: Last modified: 15 June 2017 Running title: Producing Hfq•RNA Complexes for Structural Studies Abbreviations: 3D, three-dimensional; AU, asymmetric unit; CV, column volume; DEPC, diethyl pyrocarbonate; HDV, hepatitis δ virus; IMAC, immobilized metal affinity chroma- tography; MW, molecular weight; MWCO, molecular weight cut-off; nt, nucleotide; PDB, Protein Data Bank; RNP, ribonucleoprotein; RT, room temperature Additional notes: The main text is accompanied by 2 figures and 1 table. Journal format: Methods in Molecular Biology (Springer Protocols series); this volume is entitled "Bacterial Regulatory RNA: Methods and Protocols"; an author guide is linked at http://www.springer.com/series/7651 bioRxiv preprint doi: https://doi.org/10.1101/150672; this version posted June 16, 2017. The copyright holder for this preprint (which was not certified by peer review) is the author/funder. All rights reserved. No reuse allowed without permission. Abstract Hfq is a bacterial RNA-binding protein that plays key roles in the post–transcriptional regulation of gene expression. Like other Sm proteins, Hfq assembles into toroidal discs that bind RNAs with varying affin- ities and degrees of sequence specificity.
    [Show full text]
  • A Molecular and Genetic Analysis of Otosclerosis
    A molecular and genetic analysis of otosclerosis Joanna Lauren Ziff Submitted for the degree of PhD University College London January 2014 1 Declaration I, Joanna Ziff, 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. Where work has been conducted by other members of our laboratory, this has been indicated by an appropriate reference. 2 Abstract Otosclerosis is a common form of conductive hearing loss. It is characterised by abnormal bone remodelling within the otic capsule, leading to formation of sclerotic lesions of the temporal bone. Encroachment of these lesions on to the footplate of the stapes in the middle ear leads to stapes fixation and subsequent conductive hearing loss. The hereditary nature of otosclerosis has long been recognised due to its recurrence within families, but its genetic aetiology is yet to be characterised. Although many familial linkage studies and candidate gene association studies to investigate the genetic nature of otosclerosis have been performed in recent years, progress in identifying disease causing genes has been slow. This is largely due to the highly heterogeneous nature of this condition. The research presented in this thesis examines the molecular and genetic basis of otosclerosis using two next generation sequencing technologies; RNA-sequencing and Whole Exome Sequencing. RNA–sequencing has provided human stapes transcriptomes for healthy and diseased stapes, and in combination with pathway analysis has helped identify genes and molecular processes dysregulated in otosclerotic tissue. Whole Exome Sequencing has been employed to investigate rare variants that segregate with otosclerosis in affected families, and has been followed by a variant filtering strategy, which has prioritised genes found to be dysregulated during RNA-sequencing.
    [Show full text]
  • 1213508110.Full.Pdf
    Staufen2 functions in Staufen1-mediated mRNA decay INAUGURAL ARTICLE by binding to itself and its paralog and promoting UPF1 helicase but not ATPase activity Eonyoung Parka,b, Michael L. Gleghorna,b, and Lynne E. Maquata,b,1 aDepartment of Biochemistry and Biophysics, School of Medicine and Dentistry, and bCenter for RNA Biology, University of Rochester, Rochester, NY 14642 This contribution is part of the special series of Inaugural Articles by members of the National Academy of Sciences elected in 2011. Edited by Michael R. Botchan, University of California, Berkeley, CA, and approved November 16, 2012 (received for review August 3, 2012) Staufen (STAU)1-mediated mRNA decay (SMD) is a posttranscrip- harbor a STAU1-binding site (SBS) downstream of their normal tional regulatory mechanism in mammals that degrades mRNAs termination codon in a pathway called STAU1-mediated mRNA harboring a STAU1-binding site (SBS) in their 3′-untranslated regions decay or SMD (13, 14), and work published by others indicates (3′ UTRs). We show that SMD involves not only STAU1 but also its that SMD does not involve STAU2 (3, 15). paralog STAU2. STAU2, like STAU1, is a double-stranded RNA-binding According to our current model for SMD, when translation protein that interacts directly with the ATP-dependent RNA helicase terminates upstream of an SBS, recruitment of the nonsense-me- diated mRNA decay (NMD) factor UPF1 to SBS-bound STAU1 up-frameshift 1 (UPF1) to reduce the half-life of SMD targets that form fl an SBS by either intramolecular or intermolecular base-pairing. Com- triggers mRNA decay. SMD in uences a number of cellular pro- pared with STAU1, STAU2 binds ∼10-foldmoreUPF1and∼two- to cesses, including the differentiation of mouse C2C12 myoblasts to myotubes (16), the motility of human HaCaT keratinocytes (17), fivefold more of those SBS-containing mRNAs that were tested, and it and the differentiation of mouse 3T3-L1 preadipocytes to adipo- comparably promotes UPF1 helicase activity, which is critical for SMD.
    [Show full text]
  • Supplemental Materials ZNF281 Enhances Cardiac Reprogramming
    Supplemental Materials ZNF281 enhances cardiac reprogramming by modulating cardiac and inflammatory gene expression Huanyu Zhou, Maria Gabriela Morales, Hisayuki Hashimoto, Matthew E. Dickson, Kunhua Song, Wenduo Ye, Min S. Kim, Hanspeter Niederstrasser, Zhaoning Wang, Beibei Chen, Bruce A. Posner, Rhonda Bassel-Duby and Eric N. Olson Supplemental Table 1; related to Figure 1. Supplemental Table 2; related to Figure 1. Supplemental Table 3; related to the “quantitative mRNA measurement” in Materials and Methods section. Supplemental Table 4; related to the “ChIP-seq, gene ontology and pathway analysis” and “RNA-seq” and gene ontology analysis” in Materials and Methods section. Supplemental Figure S1; related to Figure 1. Supplemental Figure S2; related to Figure 2. Supplemental Figure S3; related to Figure 3. Supplemental Figure S4; related to Figure 4. Supplemental Figure S5; related to Figure 6. Supplemental Table S1. Genes included in human retroviral ORF cDNA library. Gene Gene Gene Gene Gene Gene Gene Gene Symbol Symbol Symbol Symbol Symbol Symbol Symbol Symbol AATF BMP8A CEBPE CTNNB1 ESR2 GDF3 HOXA5 IL17D ADIPOQ BRPF1 CEBPG CUX1 ESRRA GDF6 HOXA6 IL17F ADNP BRPF3 CERS1 CX3CL1 ETS1 GIN1 HOXA7 IL18 AEBP1 BUD31 CERS2 CXCL10 ETS2 GLIS3 HOXB1 IL19 AFF4 C17ORF77 CERS4 CXCL11 ETV3 GMEB1 HOXB13 IL1A AHR C1QTNF4 CFL2 CXCL12 ETV7 GPBP1 HOXB5 IL1B AIMP1 C21ORF66 CHIA CXCL13 FAM3B GPER HOXB6 IL1F3 ALS2CR8 CBFA2T2 CIR1 CXCL14 FAM3D GPI HOXB7 IL1F5 ALX1 CBFA2T3 CITED1 CXCL16 FASLG GREM1 HOXB9 IL1F6 ARGFX CBFB CITED2 CXCL3 FBLN1 GREM2 HOXC4 IL1F7
    [Show full text]
  • Hras Intracellular Trafficking and Signal Transduction Jodi Ho-Jung Mckay Iowa State University
    Iowa State University Capstones, Theses and Retrospective Theses and Dissertations Dissertations 2007 HRas intracellular trafficking and signal transduction Jodi Ho-Jung McKay Iowa State University Follow this and additional works at: https://lib.dr.iastate.edu/rtd Part of the Biological Phenomena, Cell Phenomena, and Immunity Commons, Cancer Biology Commons, Cell Biology Commons, Genetics and Genomics Commons, and the Medical Cell Biology Commons Recommended Citation McKay, Jodi Ho-Jung, "HRas intracellular trafficking and signal transduction" (2007). Retrospective Theses and Dissertations. 13946. https://lib.dr.iastate.edu/rtd/13946 This Dissertation is brought to you for free and open access by the Iowa State University Capstones, Theses and Dissertations at Iowa State University Digital Repository. It has been accepted for inclusion in Retrospective Theses and Dissertations by an authorized administrator of Iowa State University Digital Repository. For more information, please contact [email protected]. HRas intracellular trafficking and signal transduction by Jodi Ho-Jung McKay A dissertation submitted to the graduate faculty in partial fulfillment of the requirements for the degree of DOCTOR OF PHILOSOPHY Major: Genetics Program of Study Committee: Janice E. Buss, Co-major Professor Linda Ambrosio, Co-major Professor Diane Bassham Drena Dobbs Ted Huiatt Iowa State University Ames, Iowa 2007 Copyright © Jodi Ho-Jung McKay, 2007. All rights reserved. UMI Number: 3274881 Copyright 2007 by McKay, Jodi Ho-Jung All rights reserved. UMI Microform 3274881 Copyright 2008 by ProQuest Information and Learning Company. All rights reserved. This microform edition is protected against unauthorized copying under Title 17, United States Code. ProQuest Information and Learning Company 300 North Zeeb Road P.O.
    [Show full text]