New Approach to Capture and Characterize Synaptic Proteome

Total Page:16

File Type:pdf, Size:1020Kb

New Approach to Capture and Characterize Synaptic Proteome New approach to capture and characterize synaptic proteome Xin-An Liua, Beena Kadakkuzhaa,1, Bruce Pascalb,1, Caitlin Stecklerc, Komolitdin Akhmedova, Long Yand, Michael Chalmersc, and Sathyanarayanan V. Puthanveettila,2 aDepartment of Neuroscience, bInformatics Core, and cProteomics Core Facility, The Scripps Research Institute, Scripps Florida, Jupiter, FL 33458; and dOptical Workshop and Light Microscopy Facility, Max Planck Florida Institute for Neuroscience, Jupiter, FL 33458 Edited* by Eric R. Kandel, Columbia University, New York, NY, and approved October 8, 2014 (received for review January 23, 2014) Little is known regarding the identity of the population of proteins abundant in mouse hippocampus. We studied the expression of 31 that are transported and localized to synapses. Here we describe different kinesins in mouse hippocampus using specific primers (SI a new approach that involves the isolation and systematic proteomic Appendix, Dataset S1). Quantitative real-time PCR (qRT-PCR) characterization of molecular motor kinesins to identify the popula- analysis of RNAs isolated from mouse hippocampus suggests that tions of proteins transported to synapses. We used this approach to Kif5C is highly abundant in hippocampus (normalized expression identify and compare proteins transported to synapses by kinesin level, 5.3 ± 0.14; SI Appendix,Fig.S1A). We then confirmed the (Kif) complexes Kif5C and Kif3A in the mouse hippocampus and pre- expression of Kif5C by RNA in situ hybridization using digoxigenin- frontal cortex. Approximately 40–50% of the protein cargos identified labeled sense and antisense riboprobes (SI Appendix,Fig.S1B)and in our proteomics analysis of kinesin complexes are known synaptic by immunohistochemical and Western blot analyses using an anti- proteins. We also found that the identity of kinesins and where they Kif5C antibody (SI Appendix,Fig.S1C and D and Dataset S1). are expressed determine what proteins they transport. Our results Kif5C transport complexes were then immunoprecipitated reveal a previously unappreciated role of kinesins in regulating the from the hippocampus (Fig. 1A) following a previously published composition of synaptic proteome. protocol (13) and analyzed by silver stain and Western blot analysis (Fig. 1B). Immunoprecipitated beads alone and immu- kinesin | protein transport | synapse | proteome | hippocampus noprecipitated anti-CREB antibody (SI Appendix, Dataset S1) served as specificity controls for the immunoprecipitation (IP). NEUROSCIENCE number of researchers have reported specific roles of pro- The specific protein band corresponding to Kif5C was present Ateins that are localized to synapses, termed the “synaptic only in IP of Kif5C, and not in the beads alone or in antibody proteome,” in determining synaptic function and plasticity (1–6). controls (Fig. 1B). We analyzed the protein bands corresponding Identifying the composition and dynamics of the synaptic pro- to Kif5C by mass spectrometry, which identified ∼80% of the teome is key to understanding the remodeling of existing syn- predicted tryptic peptides of Kif5C (SI Appendix, Fig. S1E). apses and growth of new synapses, as well as identifying novel To identify the population of proteins transported by Kif5C, therapeutic targets for synaptopathies (7, 8). The composition of we performed a systematic proteomic analysis of Kif5C com- the synaptic proteome is determined by proteins transported plexes from hippocampus. We performed four biological repli- from the cell body and local synthesis of proteins at the synapses. cations of Kif5C IPs and four beads-alone control IPs. Scaffold Several studies have shown the identification and localization of software identified 72 proteins present only in the Kif5C IPs and RNAs at the synapse (9–12). Comprehensive knowledge of the not in the control IPs. proteins that are actively targeted to synapses will help elucidate We considered two factors affecting the abundance of proteins the dynamics of signal transduction at the synapse, as well as the associated with kinesin in individual IP biological replications: (i) molecules that are critical for the remodeling of synapses and the IP efficiency of samples and the stability of specific proteins formation of new synapses that accompany long-term memory interacting with kinesin and (ii) the physiological state of the storage. Despite this perceived significance, however, there are neurons. Thus, to identify the maximum number of possible no currently available methodologies for studying populations of cargos associated with Kif5C in the hippocampus, we analyzed proteins targeted to synapses. the proteomics dataset (SI Appendix, Dataset S1) to identify To identify actively transported proteins, we focused on the protein cargos that were 1.5-fold enriched in kinesin IPs com- protein complexes associated with kinesins, molecular motor pared with the control IPs. We believed that this analysis would proteins that move from the cell body to distal neuronal pro- cesses and mediate the transport of various gene products (9, Significance 13). We considered that proteins associated with kinesins include both synaptic proteins and proteins localized to dendrites and Here we report a strategy for isolating and characterizing axons. Our methodology involved isolating specific kinesin populations of proteins targeted to synapses. Using this ap- complexes by coimmunoprecipitation (co-IP) from different proach, we isolated and characterized multiple protein trans- regions of the mouse brain and systematically identifying the port complexes from mouse brain, providing novel insights A protein cargos by mass spectrometry (Fig. 1 ). Using this strat- into the synaptic targeting of proteins and composition of egy, we have successfully isolated and characterized the Kif5C synaptic proteome. kinesin complex from hippocampus and prefrontal cortex (PFC) and the Kif3A kinesin complex from hippocampus. The cargos Author contributions: S.V.P. designed research; X.-A.L., B.K., C.S., K.A., and S.V.P. per- include organelles and proteins involved in translation, signaling, formed research; L.Y. contributed new reagents/analytic tools; X.-A.L., B.P., M.C., and and ion channels. Several of these proteins are implicated in S.V.P. analyzed data; and X.-A.L., M.C., and S.V.P. wrote the paper. neuropsychiatric disorders (14). The authors declare no conflict of interest. *This Direct Submission article had a prearranged editor. Results 1B.K. and B.P. contributed equally to this work. Isolation and Characterization of Protein Complexes of Molecular Motor 2To whom correspondence should be addressed. Email: [email protected]. Kif5C in the Mouse Hippocampus. To identify the population of syn- This article contains supporting information online at www.pnas.org/lookup/suppl/doi:10. aptically targeted proteins, we first searched for kinesins that are 1073/pnas.1401483111/-/DCSupplemental. www.pnas.org/cgi/doi/10.1073/pnas.1401483111 PNAS Early Edition | 1of6 Downloaded by guest on September 26, 2021 Fig. 1C, we found that these proteins are indeed specifically asso- ciated with the Kif5C complex and are not present in the beads- alone control IPs. Examination of the protein expression and dis- tribution of Kif5C (SI Appendix,Fig.S1C) and the selected cargos in hippocampus by immunohistochemical staining (SI Appendix,Fig. S2) revealed that they are expressed in hippocampal neurons as well. We studied the expression levels of Kif5C in primary neuron cultures by qRT-PCR and found that, consistent with our data on Kif5C levels in dissected hippocampus, Kif5C is abundantly expressed in primary hippocampal neurons (SI Appendix,Fig.S1F). Using specific siRNAs, we next knocked down the expression of Kif5C in primary hippocampal neurons, and evaluated the knockdown by Western blot and immunohistochemistry analyses. We found that Kif5C siRNA, but not the scrambled siRNA control, was effective in knocking down Kif5C protein levels in hippocampal neurons. Analysis of Western blot data by two-way ANOVA followed by the Tukey post hoc test indicated statisti- cally significant Kif5C knockdown (Kif5C SiRNA, scrambled and no siRNA control, F(2, 63) = 16.866, P < 0.0001) at 72 h (0, 48, and 72 h, F(2, 63) = 12.622, P < 0.0001) after the siRNA transfection (SI Appendix, Fig. S3 A and B; 37.22 ± 4.69% knockdown, Kif5C siRNA vs. scrambled siRNA and control without siRNA transfection; n = 8), which was further supported by Kif5C immunohistochemistry data (SI Appendix, Fig. S3 C and D; F(2, 30) = 81.035, ***P < 0.001, one-way ANOVA). We then examined the distributions of PURα, DIC1, and GluR1 in primary neurons (Fig. 2). As shown in SI Appendix, Fig. S3 A–D, we observed an ∼40% decrease in Kif5C expression Fig. 1. Identification of the Kif5C complex in mouse hippocampus. (A) after siRNA transfection at 72 h. We measured the intensity Strategy used to analyze Kif5C-specific protein cargos in the mouse hippo- level of these cargos on random spots in the neurites that were at campus. The schematic diagram shows the workflow for profiling Kif5C- least 10 μm away from the soma, and found that the mean transported proteins by proteomics. The processes include brain dissection, fluorescence intensities of the three cargos of Kif5C in neurites hippocampal tissue extraction and homogenization, Kif5C IP, complex sep- were significantly decreased after Kif5C knockdown compared aration by SDS/PAGE, isolation of specific bands followed by in-gel tryptic with those in the neurons without siRNA transfection
Recommended publications
  • Hares, K. M., Miners, S., Scolding, N
    Hares, K. M. , Miners, S., Scolding, N., Love, S., & Wilkins, A. (2019). KIF5A and KLC1 expression in Alzheimer’s disease: relationship and genetic influences. AMRC Open Research, 1, [1]. https://doi.org/10.12688/amrcopenres.12861.1 Publisher's PDF, also known as Version of record License (if available): CC BY Link to published version (if available): 10.12688/amrcopenres.12861.1 Link to publication record in Explore Bristol Research PDF-document This is the final published version of the article (version of record). It first appeared online via F1000 Research at https://amrcopenresearch.org/articles/1-1/v1 . Please refer to any applicable terms of use of the publisher. University of Bristol - Explore Bristol Research General rights This document is made available in accordance with publisher policies. Please cite only the published version using the reference above. Full terms of use are available: http://www.bristol.ac.uk/red/research-policy/pure/user-guides/ebr-terms/ AMRC Open Research AMRC Open Research 2019, 1:1 Last updated: 11 APR 2019 RESEARCH ARTICLE KIF5A and KLC1 expression in Alzheimer’s disease: relationship and genetic influences [version 1; peer review: 1 approved, 1 approved with reservations, 1 not approved] Kelly Hares 1, Scott Miners2, Neil Scolding1, Seth Love2, Alastair Wilkins1 1Bristol Medical School: Translational Health Sciences, MS and Stem Cell Group, University of Bristol, Bristol, BS10 5NB, UK 2Bristol Medical School: Translational Health Sciences, Dementia Research Group, University of Bristol, Bristol, BS10 5NB, UK First published: 19 Feb 2019, 1:1 (https://doi.org/10.12688/amrcopenres.12861.1 Open Peer Review v1 ) Latest published: 19 Feb 2019, 1:1 ( https://doi.org/10.12688/amrcopenres.12861.1) Referee Status: Abstract Invited Referees Background: Early disturbances in axonal transport, before the onset of gross 1 2 3 neuropathology, occur in a spectrum of neurodegenerative diseases including Alzheimer’s disease.
    [Show full text]
  • A Comparative Analysis of Transcribed Genes in the Mouse Hypothalamus and Neocortex Reveals Chromosomal Clustering
    A comparative analysis of transcribed genes in the mouse hypothalamus and neocortex reveals chromosomal clustering Wee-Ming Boon*, Tim Beissbarth†, Lavinia Hyde†, Gordon Smyth†, Jenny Gunnersen*, Derek A. Denton*‡, Hamish Scott†, and Seong-Seng Tan* *Howard Florey Institute, University of Melbourne, Parkville 3052, Australia; and †Genetics and Bioinfomatics Division, Walter and Eliza Hall Institute of Medical Research, Royal Parade, Parkville 3050, Australia Contributed by Derek A. Denton, August 26, 2004 The hypothalamus and neocortex are subdivisions of the mamma- representing all of the genes that are expressed (qualitative and lian forebrain, and yet, they have vastly different evolutionary quantitative) in the hypothalamus and neocortex under standard histories, cytoarchitecture, and biological functions. In an attempt conditions. to define these attributes in terms of their genetic activity, we have In the current study, we describe the use of the Serial Analysis compared their genetic repertoires by using the Serial Analysis of of Gene Expression (SAGE) database, which allows simulta- Gene Expression database. From a comparison of 78,784 hypothal- neous detection of the expression levels of the entire genome amus tags with 125,296 neocortical tags, we demonstrate that each without a priori knowledge of gene sequences (13). SAGE takes structure possesses a different transcriptional profile in terms of advantage of the fact that a small sequence tag taken from a gene ontological characteristics and expression levels. Despite its defined position within the transcript is sufficient to identify the more recent evolutionary history, the neocortex has a more com- gene (from known cDNA or EST sequences), and up to 40 tags plex pattern of gene activity.
    [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]
  • Drp1 Overexpression Induces Desmin Disassembling and Drives Kinesin-1 Activation Promoting Mitochondrial Trafficking in Skeletal Muscle
    Cell Death & Differentiation (2020) 27:2383–2401 https://doi.org/10.1038/s41418-020-0510-7 ARTICLE Drp1 overexpression induces desmin disassembling and drives kinesin-1 activation promoting mitochondrial trafficking in skeletal muscle 1 1 2 2 2 3 Matteo Giovarelli ● Silvia Zecchini ● Emanuele Martini ● Massimiliano Garrè ● Sara Barozzi ● Michela Ripolone ● 3 1 4 1 5 Laura Napoli ● Marco Coazzoli ● Chiara Vantaggiato ● Paulina Roux-Biejat ● Davide Cervia ● 1 1 2 1,4 6 Claudia Moscheni ● Cristiana Perrotta ● Dario Parazzoli ● Emilio Clementi ● Clara De Palma Received: 1 August 2019 / Revised: 13 December 2019 / Accepted: 23 January 2020 / Published online: 10 February 2020 © The Author(s) 2020. This article is published with open access Abstract Mitochondria change distribution across cells following a variety of pathophysiological stimuli. The mechanisms presiding over this redistribution are yet undefined. In a murine model overexpressing Drp1 specifically in skeletal muscle, we find marked mitochondria repositioning in muscle fibres and we demonstrate that Drp1 is involved in this process. Drp1 binds KLC1 and enhances microtubule-dependent transport of mitochondria. Drp1-KLC1 coupling triggers the displacement of KIF5B from 1234567890();,: 1234567890();,: kinesin-1 complex increasing its binding to microtubule tracks and mitochondrial transport. High levels of Drp1 exacerbate this mechanism leading to the repositioning of mitochondria closer to nuclei. The reduction of Drp1 levels decreases kinesin-1 activation and induces the partial recovery of mitochondrial distribution. Drp1 overexpression is also associated with higher cyclin-dependent kinase-1 (Cdk-1) activation that promotes the persistent phosphorylation of desmin at Ser-31 and its disassembling. Fission inhibition has a positive effect on desmin Ser-31 phosphorylation, regardless of Cdk-1 activation, suggesting that induction of both fission and Cdk-1 are required for desmin collapse.
    [Show full text]
  • Understanding the Role of the Chromosome 15Q25.1 in COPD Through Epigenetics and Transcriptomics
    European Journal of Human Genetics (2018) 26:709–722 https://doi.org/10.1038/s41431-017-0089-8 ARTICLE Understanding the role of the chromosome 15q25.1 in COPD through epigenetics and transcriptomics 1 1,2 1,3,4 5,6 5,6 Ivana Nedeljkovic ● Elena Carnero-Montoro ● Lies Lahousse ● Diana A. van der Plaat ● Kim de Jong ● 5,6 5,7 6 8 9 10 Judith M. Vonk ● Cleo C. van Diemen ● Alen Faiz ● Maarten van den Berge ● Ma’en Obeidat ● Yohan Bossé ● 11 1,12 12 1 David C. Nickle ● BIOS Consortium ● Andre G. Uitterlinden ● Joyce J. B. van Meurs ● Bruno C. H. Stricker ● 1,4,13 6,8 5,6 1 1 Guy G. Brusselle ● Dirkje S. Postma ● H. Marike Boezen ● Cornelia M. van Duijn ● Najaf Amin Received: 14 March 2017 / Revised: 6 November 2017 / Accepted: 19 December 2017 / Published online: 8 February 2018 © The Author(s) 2018. This article is published with open access Abstract Chronic obstructive pulmonary disease (COPD) is a major health burden in adults and cigarette smoking is considered the most important environmental risk factor of COPD. Chromosome 15q25.1 locus is associated with both COPD and smoking. Our study aims at understanding the mechanism underlying the association of chromosome 15q25.1 with COPD through epigenetic and transcriptional variation in a population-based setting. To assess if COPD-associated variants in 1234567890();,: 15q25.1 are methylation quantitative trait loci, epigenome-wide association analysis of four genetic variants, previously associated with COPD (P < 5 × 10−8) in the 15q25.1 locus (rs12914385:C>T-CHRNA3, rs8034191:T>C-HYKK, rs13180: C>T-IREB2 and rs8042238:C>T-IREB2), was performed in the Rotterdam study (n = 1489).
    [Show full text]
  • Proteomic Analysis Uncovers Measles Virus Protein C Interaction with P65
    bioRxiv preprint doi: https://doi.org/10.1101/2020.05.08.084418; this version posted May 9, 2020. 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. Proteomic Analysis Uncovers Measles Virus Protein C Interaction with p65/iASPP/p53 Protein Complex Alice Meignié1,2*, Chantal Combredet1*, Marc Santolini 3,4, István A. Kovács4,5,6, Thibaut Douché7, Quentin Giai Gianetto 7,8, Hyeju Eun9, Mariette Matondo7, Yves Jacob10, Regis Grailhe9, Frédéric Tangy1**, and Anastassia V. Komarova1, 10** 1 Viral Genomics and Vaccination Unit, Department of Virology, Institut Pasteur, CNRS UMR-3569, 75015 Paris, France 2 Université Paris Diderot, Sorbonne Paris Cité, Paris, France 3 Center for Research and Interdisciplinarity (CRI), Université de Paris, INSERM U1284 4 Network Science Institute and Department of Physics, Northeastern University, Boston, MA 02115, USA 5 Department of Physics and Astronomy, Northwestern University, Evanston, IL 60208-3109, USA 6 Department of Network and Data Science, Central European University, Budapest, H-1051, Hungary 7 Proteomics platform, Mass Spectrometry for Biology Unit (MSBio), Institut Pasteur, CNRS USR 2000, Paris, France. 8 Bioinformatics and Biostatistics Hub, Computational Biology Department, Institut Pasteur, CNRS USR3756, Paris, France 9 Technology Development Platform, Institut Pasteur Korea, Seongnam-si, Republic of Korea 10 Laboratory of Molecular Genetics of RNA Viruses, Institut Pasteur, CNRS UMR-3569,
    [Show full text]
  • 1 Supporting Information for a Microrna Network Regulates
    Supporting Information for A microRNA Network Regulates Expression and Biosynthesis of CFTR and CFTR-ΔF508 Shyam Ramachandrana,b, Philip H. Karpc, Peng Jiangc, Lynda S. Ostedgaardc, Amy E. Walza, John T. Fishere, Shaf Keshavjeeh, Kim A. Lennoxi, Ashley M. Jacobii, Scott D. Rosei, Mark A. Behlkei, Michael J. Welshb,c,d,g, Yi Xingb,c,f, Paul B. McCray Jr.a,b,c Author Affiliations: Department of Pediatricsa, Interdisciplinary Program in Geneticsb, Departments of Internal Medicinec, Molecular Physiology and Biophysicsd, Anatomy and Cell Biologye, Biomedical Engineeringf, Howard Hughes Medical Instituteg, Carver College of Medicine, University of Iowa, Iowa City, IA-52242 Division of Thoracic Surgeryh, Toronto General Hospital, University Health Network, University of Toronto, Toronto, Canada-M5G 2C4 Integrated DNA Technologiesi, Coralville, IA-52241 To whom correspondence should be addressed: Email: [email protected] (M.J.W.); yi- [email protected] (Y.X.); Email: [email protected] (P.B.M.) This PDF file includes: Materials and Methods References Fig. S1. miR-138 regulates SIN3A in a dose-dependent and site-specific manner. Fig. S2. miR-138 regulates endogenous SIN3A protein expression. Fig. S3. miR-138 regulates endogenous CFTR protein expression in Calu-3 cells. Fig. S4. miR-138 regulates endogenous CFTR protein expression in primary human airway epithelia. Fig. S5. miR-138 regulates CFTR expression in HeLa cells. Fig. S6. miR-138 regulates CFTR expression in HEK293T cells. Fig. S7. HeLa cells exhibit CFTR channel activity. Fig. S8. miR-138 improves CFTR processing. Fig. S9. miR-138 improves CFTR-ΔF508 processing. Fig. S10. SIN3A inhibition yields partial rescue of Cl- transport in CF epithelia.
    [Show full text]
  • Actin Nucleator Spire 1 Is a Regulator of Ectoplasmic Specialization in the Testis Qing Wen1,Nanli1,Xiangxiao 1,2,Wing-Yeelui3, Darren S
    Wen et al. Cell Death and Disease (2018) 9:208 DOI 10.1038/s41419-017-0201-6 Cell Death & Disease ARTICLE Open Access Actin nucleator Spire 1 is a regulator of ectoplasmic specialization in the testis Qing Wen1,NanLi1,XiangXiao 1,2,Wing-yeeLui3, Darren S. Chu1, Chris K. C. Wong4, Qingquan Lian5,RenshanGe5, Will M. Lee3, Bruno Silvestrini6 and C. Yan Cheng 1 Abstract Germ cell differentiation during the epithelial cycle of spermatogenesis is accompanied by extensive remodeling at the Sertoli cell–cell and Sertoli cell–spermatid interface to accommodate the transport of preleptotene spermatocytes and developing spermatids across the blood–testis barrier (BTB) and the adluminal compartment of the seminiferous epithelium, respectively. The unique cell junction in the testis is the actin-rich ectoplasmic specialization (ES) designated basal ES at the Sertoli cell–cell interface, and the apical ES at the Sertoli–spermatid interface. Since ES dynamics (i.e., disassembly, reassembly and stabilization) are supported by actin microfilaments, which rapidly converts between their bundled and unbundled/branched configuration to confer plasticity to the ES, it is logical to speculate that actin nucleation proteins play a crucial role to ES dynamics. Herein, we reported findings that Spire 1, an actin nucleator known to polymerize actins into long stretches of linear microfilaments in cells, is an important regulator of ES dynamics. Its knockdown by RNAi in Sertoli cells cultured in vitro was found to impede the Sertoli cell tight junction (TJ)-permeability barrier through changes in the organization of F-actin across Sertoli cell cytosol. Unexpectedly, Spire 1 knockdown also perturbed microtubule (MT) organization in Sertoli cells cultured in vitro.
    [Show full text]
  • Applying Expression Profile Similarity for Discovery of Patient-Specific
    bioRxiv preprint doi: https://doi.org/10.1101/172015; this version posted September 17, 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 4.0 International license. Applying expression profile similarity for discovery of patient-specific functional mutations Guofeng Meng Partner Institute of Computational Biology, Yueyang 333, Shanghai, China email: [email protected] Abstract The progress of cancer genome sequencing projects yields unprecedented information of mutations for numerous patients. However, the complexity of mutation profiles of patients hinders the further understanding of mechanisms of oncogenesis. One basic question is how to uncover mutations with functional impacts. In this work, we introduce a computational method to predict functional somatic mutations for each of patient by integrating mutation recurrence with similarity of expression profiles of patients. With this method, the functional mutations are determined by checking the mutation enrichment among a group of patients with similar expression profiles. We applied this method to three cancer types and identified the functional mutations. Comparison of the predictions for three cancer types suggested that most of the functional mutations were cancer-type-specific with one exception to p53. By checking prediction results, we found that our method effectively filtered non-functional mutations resulting from large protein sizes. In addition, this methods can also perform functional annotation to each patient to describe their association with signalling pathways or biological processes. In breast cancer, we predicted "cell adhesion" and other mutated gene associated terms to be significantly enriched among patients.
    [Show full text]
  • Cldn19 Clic2 Clmp Cln3
    NewbornDx™ Advanced Sequencing Evaluation When time to diagnosis matters, the NewbornDx™ Advanced Sequencing Evaluation from Athena Diagnostics delivers rapid, 5- to 7-day results on a targeted 1,722-genes. A2ML1 ALAD ATM CAV1 CLDN19 CTNS DOCK7 ETFB FOXC2 GLUL HOXC13 JAK3 AAAS ALAS2 ATP1A2 CBL CLIC2 CTRC DOCK8 ETFDH FOXE1 GLYCTK HOXD13 JUP AARS2 ALDH18A1 ATP1A3 CBS CLMP CTSA DOK7 ETHE1 FOXE3 GM2A HPD KANK1 AASS ALDH1A2 ATP2B3 CC2D2A CLN3 CTSD DOLK EVC FOXF1 GMPPA HPGD K ANSL1 ABAT ALDH3A2 ATP5A1 CCDC103 CLN5 CTSK DPAGT1 EVC2 FOXG1 GMPPB HPRT1 KAT6B ABCA12 ALDH4A1 ATP5E CCDC114 CLN6 CUBN DPM1 EXOC4 FOXH1 GNA11 HPSE2 KCNA2 ABCA3 ALDH5A1 ATP6AP2 CCDC151 CLN8 CUL4B DPM2 EXOSC3 FOXI1 GNAI3 HRAS KCNB1 ABCA4 ALDH7A1 ATP6V0A2 CCDC22 CLP1 CUL7 DPM3 EXPH5 FOXL2 GNAO1 HSD17B10 KCND2 ABCB11 ALDOA ATP6V1B1 CCDC39 CLPB CXCR4 DPP6 EYA1 FOXP1 GNAS HSD17B4 KCNE1 ABCB4 ALDOB ATP7A CCDC40 CLPP CYB5R3 DPYD EZH2 FOXP2 GNE HSD3B2 KCNE2 ABCB6 ALG1 ATP8A2 CCDC65 CNNM2 CYC1 DPYS F10 FOXP3 GNMT HSD3B7 KCNH2 ABCB7 ALG11 ATP8B1 CCDC78 CNTN1 CYP11B1 DRC1 F11 FOXRED1 GNPAT HSPD1 KCNH5 ABCC2 ALG12 ATPAF2 CCDC8 CNTNAP1 CYP11B2 DSC2 F13A1 FRAS1 GNPTAB HSPG2 KCNJ10 ABCC8 ALG13 ATR CCDC88C CNTNAP2 CYP17A1 DSG1 F13B FREM1 GNPTG HUWE1 KCNJ11 ABCC9 ALG14 ATRX CCND2 COA5 CYP1B1 DSP F2 FREM2 GNS HYDIN KCNJ13 ABCD3 ALG2 AUH CCNO COG1 CYP24A1 DST F5 FRMD7 GORAB HYLS1 KCNJ2 ABCD4 ALG3 B3GALNT2 CCS COG4 CYP26C1 DSTYK F7 FTCD GP1BA IBA57 KCNJ5 ABHD5 ALG6 B3GAT3 CCT5 COG5 CYP27A1 DTNA F8 FTO GP1BB ICK KCNJ8 ACAD8 ALG8 B3GLCT CD151 COG6 CYP27B1 DUOX2 F9 FUCA1 GP6 ICOS KCNK3 ACAD9 ALG9
    [Show full text]
  • Viewed and Published Immediately Upon Acceptance Cited in Pubmed and Archived on Pubmed Central Yours — You Keep the Copyright
    BMC Genomics BioMed Central Research article Open Access Differential gene expression in ADAM10 and mutant ADAM10 transgenic mice Claudia Prinzen1, Dietrich Trümbach2, Wolfgang Wurst2, Kristina Endres1, Rolf Postina1 and Falk Fahrenholz*1 Address: 1Johannes Gutenberg-University, Institute of Biochemistry, Mainz, Johann-Joachim-Becherweg 30, 55128 Mainz, Germany and 2Helmholtz Zentrum München – German Research Center for Environmental Health, Institute for Developmental Genetics, Ingolstädter Landstraße 1, 85764 Neuherberg, Germany Email: Claudia Prinzen - [email protected]; Dietrich Trümbach - [email protected]; Wolfgang Wurst - [email protected]; Kristina Endres - [email protected]; Rolf Postina - [email protected]; Falk Fahrenholz* - [email protected] * Corresponding author Published: 5 February 2009 Received: 19 June 2008 Accepted: 5 February 2009 BMC Genomics 2009, 10:66 doi:10.1186/1471-2164-10-66 This article is available from: http://www.biomedcentral.com/1471-2164/10/66 © 2009 Prinzen et al; licensee BioMed Central Ltd. This is an Open Access article distributed under the terms of the Creative Commons Attribution License (http://creativecommons.org/licenses/by/2.0), which permits unrestricted use, distribution, and reproduction in any medium, provided the original work is properly cited. Abstract Background: In a transgenic mouse model of Alzheimer disease (AD), cleavage of the amyloid precursor protein (APP) by the α-secretase ADAM10 prevented amyloid plaque formation, and alleviated cognitive deficits. Furthermore, ADAM10 overexpression increased the cortical synaptogenesis. These results suggest that upregulation of ADAM10 in the brain has beneficial effects on AD pathology. Results: To assess the influence of ADAM10 on the gene expression profile in the brain, we performed a microarray analysis using RNA isolated from brains of five months old mice overexpressing either the α-secretase ADAM10, or a dominant-negative mutant (dn) of this enzyme.
    [Show full text]
  • Structural Basis for Isoform-Specific Kinesin-1 Recognition of Y-Acidic
    bioRxiv preprint doi: https://doi.org/10.1101/322057; this version posted May 14, 2018. 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. Structural basis for isoform-specific kinesin-1 recognition of Y-acidic cargo adaptors Stefano Pernigo1, Magda Chegkazi1, Yan Y. Yip1, Conor Treacy1, Giulia Glorani1, Kjetil Hansen2, Argyris Politis2, Mark P. Dodding1,3,*, Roberto A. Steiner1,* 1Randall Centre of Cell and Molecular Biophysics, Faculty of Life Sciences and Medicine, King’s College London, London SE1 1UL, UK 2Department of Chemistry, King’s College London Britannia House 7 Trinity Street, London, SE1 1DB (UK) 3current address School of Biochemistry, Faculty of Biomedical Sciences, University of Bristol, University Walk, Bristol BS8 1TD *Correspondence email: [email protected] or [email protected] 1 bioRxiv preprint doi: https://doi.org/10.1101/322057; this version posted May 14, 2018. 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. The light chains (KLCs) of the heterotetrameric microtubule motor kinesin-1, that bind to cargo adaptor proteins and regulate its activity, have a capacity to recognize short peptides via their tetratricopeptide repeat domains (KLCTPR). Here, using X-ray crystallography, we show how kinesin-1 recognizes a novel class of adaptor motifs that we call ‘Y-acidic’ (tyrosine flanked by acidic residues), in a KLC-isoform specific manner. Binding specificities of Y-acidic motifs (present in JIP1 and in TorsinA) to KLC1TPR are distinct from those utilized for the recognition of W-acidic motifs found in adaptors that are KLC- isoform non-selective.
    [Show full text]