Defining Diversity, Specialization, and Gene Specificity in Transcriptomes Through Information Theory

Total Page:16

File Type:pdf, Size:1020Kb

Defining Diversity, Specialization, and Gene Specificity in Transcriptomes Through Information Theory Defining diversity, specialization, and gene specificity in transcriptomes through information theory Octavio Marti´nez*† and M. Humberto Reyes-Valde´ s‡ *Laboratorio Nacional de Geno´mica para la Biodiversidad (Langebio), Cinvestav, Campus Guanajuato, Apartado Postal 629, C.P. 36500 Irapuato, Guanajuato, Mexico; and ‡Department of Plant Breeding, Universidad Auto´noma Agraria Antonio Narro, Buenavista, C.P. 25315 Saltillo, Coahuila, Mexico Communicated by Luis Herrera-Estrella, Center for Research and Advanced Studies, Guanajuato, Mexico, April 10, 2008 (received for review March 10, 2008) The transcriptome is a set of genes transcribed in a given tissue applied to many scientific fields (8). In particular, it has been under specific conditions and can be characterized by a list of genes repeatedly applied to genetics in distinct contexts (9–12). Our with their corresponding frequencies of transcription. Transcrip- approach consists of considering as symbols, in the sense of tome changes can be measured by counting gene tags from mRNA information theory, the distinct transcripts found in a tissue and libraries or by measuring light signals in DNA microarrays. In any counting their abundance to calculate information parameters. case, it is difficult to completely comprehend the global changes that occur in the transcriptome, given that thousands of gene Results and Discussion expression measurements are involved. We propose an approach Theoretical Framework. Consider the division of an organism in to define and estimate the diversity and specialization of tran- tissues; the transcriptomes of each tissue can then be simply scriptomes and gene specificity. We define transcriptome diversity described as the set of relative frequencies, pij, for the ith gene as the Shannon entropy of its frequency distribution. Gene spec- (i ϭ 1, 2, ...,g)inthejth tissue (j ϭ 1, 2, ...,t). Then the ificity is defined as the mutual information between the tissues and diversity of the transcriptome of each tissue can be quantified by the corresponding transcript, allowing detection of either house- an adaptation of Shannon’s entropy formula, keeping or highly specific genes and clarifying the meaning of these concepts in the literature. Tissue specialization is measured g ϭ Ϫ͸ ͑ ͒ by average gene specificity. We introduce the formulae using a Hj pijlog2 pij . [1] simple example and show their application in two datasets of gene iϭ1 expression in human tissues. Visualization of the positions of transcriptomes in a system of diversity and specialization coordi- Hj will vary from zero when only one gene is transcribed up to nates makes it possible to understand at a glance their interrela- log2(g), where all g genes are transcribed at the same frequency: tions, summarizing in a powerful way which transcriptomes are 1/g. If we consider the average frequency of the ith gene among richer in diversity of expressed genes, or which are relatively more tissues, say, specialized. The framework presented enlightens the relation among transcriptomes, allowing a better understanding of their 1 t changes through the development of the organism or in response p ϭ ͸ p , [2] i t ij to environmental stimuli. jϭ1 biological complexity ͉ gene expression ͉ microarrays ͉ serial analysis of and define gene specificity as the information that its expression gene expression (SAGE) ͉ Shannon entropy provides about the identity of the source tissue as t he transcriptome is highly dynamic; the relative transcription 1 pij pij S ϭ ͩ͸ log ͪ . [3] i 2 Tfrequencies of the genes change in response to environmen- t pi pi ϭ tal and internal stimuli redirecting the functional and structural j 1 landscape of living organisms. Currently, we can measure tran- Si will give a value of zero if the gene is transcribed at the same scriptome changes by counting gene tags with technologies as frequency in all tissues and a maximum value of log2(t)ifthe serial analysis of gene expression (SAGE) (1), massively parallel gene is exclusively expressed in a single tissue. To quantify signature sequencing (MPSS) (2), pyrosequencing of cDNA the tissue specialization we can obtain for each jth tissue, the libraries obtained from mRNA (3), or alternatively by measuring average of the gene specificities, say, light signals in DNA microarrays (4). In any case, it is difficult to completely understand the global changes that occur in the g transcriptome, given that thousands of gene frequency measure- ␦ ϭ ͸ p S . [4] ments are involved. Here, we present a set of indexes that allow j ij i iϭ1 the calculation of transcriptome diversity and context special- ization and the degree of gene specificity. These indexes are ␦j varies from zero if all genes expressed in the tissue are based on the adaptation of Shannon’s information theory (5) to completely unspecific (Si ϭ 0 for all i) up to a maximum of the transcriptome framework. Our approach is exemplified by log2(t), when all genes expressed in the tissue are not expressed the analysis of a dataset of the transcriptome of 32 human tissues, from which Ͼ32 million gene tags were obtained (6) and a comparable dataset for the expression of human genes in 36 Author contributions: O.M. and M.H.R.-V. designed research, performed research, contrib- human tissues using the Affymetrix GeneChip for the human uted new reagents/analytic tools, analyzed data, and wrote the paper. genome (7). The main conclusion of our study is that this The authors declare no conflict of interest. conceptualization allows elucidation of aspects of the transcrip- Freely available online through the PNAS open access option. tome previously uncharacterized due to the quantity and com- †To whom correspondence should be addressed. E-mail: [email protected]. plexity of the data. This article contains supporting information online at www.pnas.org/cgi/content/full/ GENETICS Information theory was pioneered by Claude E. Shannon in a 0803479105/DCSupplemental. seminal paper in 1948 (5), and it has been generalized and © 2008 by The National Academy of Sciences of the USA www.pnas.org͞cgi͞doi͞10.1073͞pnas.0803479105 PNAS ͉ July 15, 2008 ͉ vol. 105 ͉ no. 28 ͉ 9709–9714 Downloaded by guest on September 26, 2021 1.0 Gene 1; S = 2.0000 a Gene 2; S = 0.7341 2.0 Gene 3; S = 0.4151 Gene 4; S = 1.0501 0.8 1.5 0.6 transcription (pij) ation) 0.4 j (Specializ δ δ H / mean 1.0 0.2 b Relative frequency of d c 0.0 abcd 0.0 0.5 1.0 1.5 2.0 Tissue Hj (Diversity) Fig. 1. Example of values of information parameters as functions of the relative frequencies of transcription of genes in a system with four tissues and four genes. (A) Bar plot of the relative frequencies of transcription of each gene in the four tissues. Values of gene specificities, Si, are presented. (B) Scatter plot of Hj (diversity) vs. ␦j (specialization, given by the average of the gene specificities) for each tissue. The value of H in the whole system and the mean of ␦j is plotted as a diamond. anywhere else. If we substitute the values of pij by pi in Eq. 1 and an approximate intermediate diversity, whereas tissue b, tran- ignore the subindex j, we obtain a measure, say H, of the diversity scribing three genes, one with relatively high specificity (S4 ϭ of the whole system. 1.05), is the second most diverse and specialized. The diversity To define a measure of divergence with respect to the whole of the whole system, with H ϭ 1.9965, almost reaches the ϭ average transcriptome, let us define the average log2 of the global maximum diversity for a system with four genes, log2(4) 2, and transcript frequencies in a given tissue, say the mean average diversity of the tissues, mean (␦j) ϭ 1.0424, is almost in the center of the range of possible diversities, 0 to log2 g (4) ϭ 2 (diamond in Fig. 1B). In this example, the properties of ϭ Ϫ͸ ͑ ͒ HRj pijlog2 pi . [5] the transcriptome can be easily understood by inspection of the iϭ1 transcription frequencies of the four genes, but in any real case, thousands of genes are involved, and the appreciation of the HRj will be equal to or larger than the corresponding Hj, reaching transcriptome properties becomes impossible without the tools equality if and only if pi ϭ pij for all values of i. Now we can define described here. the Kullback–Leibler divergence of the tissue j as Analysis of Human Data: The Tissue Perspective. To exemplify our ϭ Ϫ Dj HRj Hj. [6] approach with real cases, we analyzed two comparable datasets. The first consists of Ͼ31 millions MPSS tags for 22,935 genes Dj measures how much a given tissue j departs from the measured in 32 human tissues (6), and the second is a microarray corresponding transcriptome distribution of the whole system. expression profiling of 36 human tissues (7). These two datasets Notice that Hj, the measure of diversity, depends only on the share 28 human tissues and thus present the possibility of relative transcription frequencies of the tissue j; thus, it is comparing the results of our approach with two highly dissimilar independent of the context. However, the measures of tissue methodologies. specialization and divergence, ␦ and D , respectively, depend not j j Fig. 2 shows a scatter plot of the values of diversity, Hj vs. the only on these frequencies but also on those of the remaining values of specialization given by the average gene specificity of tissues; thus, these parameters are sensitive to the context where the tissues, ␦j. they are measured [see supporting information (SI) Text]. From the results of the MPSS dataset (Fig.
Recommended publications
  • A Minimal Set of Internal Control Genes for Gene Expression Studies in Head
    bioRxiv preprint doi: https://doi.org/10.1101/108381; this version posted February 14, 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. 1 A minimal set of internal control genes for gene expression studies in head 2 and neck squamous cell carcinoma. 1 1 1 2 3 Vinayak Palve , Manisha Pareek , Neeraja M Krishnan , Gangotri Siddappa , 2 2 1,3* 4 Amritha Suresh , Moni Abraham Kuriakose and Binay Panda 5 1 6 Ganit Labs, Bio-IT Centre, Institute of Bioinformatics and Applied Biotechnology, 7 Bangalore 560100 2 8 Mazumdar Shaw Cancer Centre and Mazumdar Shaw Centre for Translational 9 Research, Bangalore 560096 3 10 Strand Life Sciences, Bangalore 560024 11 * Corresponding author: [email protected] 12 13 Abstract: 14 Background: Selection of the right reference gene(s) is crucial in the analysis and 15 interpretation of gene expression data. In head and neck cancer, studies evaluating 16 the efficacy of internal reference genes are rare. Here, we present data for a minimal 17 set of candidates as internal control genes for gene expression studies in head and 18 neck cancer. 19 Methods: We analyzed data from multiple sources (in house whole-genome gene 20 expression microarrays, n=21; TCGA RNA-seq, n=42, and published gene 21 expression studies in head and neck tumors from literature) to come up with a set of 22 genes (discovery set) for their stable expression across tumor and normal tissues.
    [Show full text]
  • Anti-Inflammatory Role of Curcumin in LPS Treated A549 Cells at Global Proteome Level and on Mycobacterial Infection
    Anti-inflammatory Role of Curcumin in LPS Treated A549 cells at Global Proteome level and on Mycobacterial infection. Suchita Singh1,+, Rakesh Arya2,3,+, Rhishikesh R Bargaje1, Mrinal Kumar Das2,4, Subia Akram2, Hossain Md. Faruquee2,5, Rajendra Kumar Behera3, Ranjan Kumar Nanda2,*, Anurag Agrawal1 1Center of Excellence for Translational Research in Asthma and Lung Disease, CSIR- Institute of Genomics and Integrative Biology, New Delhi, 110025, India. 2Translational Health Group, International Centre for Genetic Engineering and Biotechnology, New Delhi, 110067, India. 3School of Life Sciences, Sambalpur University, Jyoti Vihar, Sambalpur, Orissa, 768019, India. 4Department of Respiratory Sciences, #211, Maurice Shock Building, University of Leicester, LE1 9HN 5Department of Biotechnology and Genetic Engineering, Islamic University, Kushtia- 7003, Bangladesh. +Contributed equally for this work. S-1 70 G1 S 60 G2/M 50 40 30 % of cells 20 10 0 CURI LPSI LPSCUR Figure S1: Effect of curcumin and/or LPS treatment on A549 cell viability A549 cells were treated with curcumin (10 µM) and/or LPS or 1 µg/ml for the indicated times and after fixation were stained with propidium iodide and Annexin V-FITC. The DNA contents were determined by flow cytometry to calculate percentage of cells present in each phase of the cell cycle (G1, S and G2/M) using Flowing analysis software. S-2 Figure S2: Total proteins identified in all the three experiments and their distribution betwee curcumin and/or LPS treated conditions. The proteins showing differential expressions (log2 fold change≥2) in these experiments were presented in the venn diagram and certain number of proteins are common in all three experiments.
    [Show full text]
  • Nuclear Factor-Erythroid-2 Related Transcription Factor-1 (Nrf1) Is Regulated MARK by O-Glcnac Transferase
    Free Radical Biology and Medicine 110 (2017) 196–205 Contents lists available at ScienceDirect Free Radical Biology and Medicine journal homepage: www.elsevier.com/locate/freeradbiomed Original article Nuclear factor-erythroid-2 related transcription factor-1 (Nrf1) is regulated MARK by O-GlcNAc transferase Jeong Woo Hana,1, Joshua L. Valdeza,1, Daniel V. Hoa, Candy S. Leea, Hyun Min Kima, ⁎ Xiaorong Wangb, Lan Huangb,Jefferson Y. Chana, a Department of Laboratory Medicine and Pathology, University of California, Irvine, D440 Medical Sciences, Irvine, CA 92697, USA b Departments of Physiology and Biophysics, University of California, Irvine, D440 Medical Sciences, Irvine, CA 92697, USA ARTICLE INFO ABSTRACT Keywords: The Nrf1 (Nuclear factor E2-related factor 1) transcription factor performs a critical role in regulating cellular Oxidative stress homeostasis. Using a proteomic approach, we identified Host Cell Factor-1 (HCF1), a co-regulator of tran- O-GlcNAcylation scription, and O-GlcNAc transferase (OGT), the enzyme that mediates protein O-GlcNAcylation, as cellular OGT partners of Nrf1a, an isoform of Nrf1. Nrf1a directly interacts with HCF1 through the HCF1 binding motif Transcription factor (HBM), while interaction with OGT is mediated through HCF1. Overexpression of HCF1 and OGT leads to Protein stability increased Nrf1a protein stability. Addition of O-GlcNAc decreases ubiquitination and degradation of Nrf1a. Transcriptional activation by Nrf1a is increased by OGT overexpression and treatment with PUGNAc. Together, these data suggest that OGT can act as a regulator of Nrf1a. 1. Introduction recognized as an essential co-factor for various transcription factors, and it is involved in regulating a plethora of cellular processes including O-GlcNAc transferase (OGT) is a highly conserved enzyme that cell-cycle progression and gene expression [18–21].
    [Show full text]
  • Supplementary Table S1. Correlation Between the Mutant P53-Interacting Partners and PTTG3P, PTTG1 and PTTG2, Based on Data from Starbase V3.0 Database
    Supplementary Table S1. Correlation between the mutant p53-interacting partners and PTTG3P, PTTG1 and PTTG2, based on data from StarBase v3.0 database. PTTG3P PTTG1 PTTG2 Gene ID Coefficient-R p-value Coefficient-R p-value Coefficient-R p-value NF-YA ENSG00000001167 −0.077 8.59e-2 −0.210 2.09e-6 −0.122 6.23e-3 NF-YB ENSG00000120837 0.176 7.12e-5 0.227 2.82e-7 0.094 3.59e-2 NF-YC ENSG00000066136 0.124 5.45e-3 0.124 5.40e-3 0.051 2.51e-1 Sp1 ENSG00000185591 −0.014 7.50e-1 −0.201 5.82e-6 −0.072 1.07e-1 Ets-1 ENSG00000134954 −0.096 3.14e-2 −0.257 4.83e-9 0.034 4.46e-1 VDR ENSG00000111424 −0.091 4.10e-2 −0.216 1.03e-6 0.014 7.48e-1 SREBP-2 ENSG00000198911 −0.064 1.53e-1 −0.147 9.27e-4 −0.073 1.01e-1 TopBP1 ENSG00000163781 0.067 1.36e-1 0.051 2.57e-1 −0.020 6.57e-1 Pin1 ENSG00000127445 0.250 1.40e-8 0.571 9.56e-45 0.187 2.52e-5 MRE11 ENSG00000020922 0.063 1.56e-1 −0.007 8.81e-1 −0.024 5.93e-1 PML ENSG00000140464 0.072 1.05e-1 0.217 9.36e-7 0.166 1.85e-4 p63 ENSG00000073282 −0.120 7.04e-3 −0.283 1.08e-10 −0.198 7.71e-6 p73 ENSG00000078900 0.104 2.03e-2 0.258 4.67e-9 0.097 3.02e-2 Supplementary Table S2.
    [Show full text]
  • Produktinformation
    Produktinformation Diagnostik & molekulare Diagnostik Laborgeräte & Service Zellkultur & Verbrauchsmaterial Forschungsprodukte & Biochemikalien Weitere Information auf den folgenden Seiten! See the following pages for more information! Lieferung & Zahlungsart Lieferung: frei Haus Bestellung auf Rechnung SZABO-SCANDIC Lieferung: € 10,- HandelsgmbH & Co KG Erstbestellung Vorauskassa Quellenstraße 110, A-1100 Wien T. +43(0)1 489 3961-0 Zuschläge F. +43(0)1 489 3961-7 [email protected] • Mindermengenzuschlag www.szabo-scandic.com • Trockeneiszuschlag • Gefahrgutzuschlag linkedin.com/company/szaboscandic • Expressversand facebook.com/szaboscandic PSMD10 Antibody, Biotin conjugated Product Code CSB-PA018899LD01HU Abbreviation 26S proteasome non-ATPase regulatory subunit 10 Storage Upon receipt, store at -20°C or -80°C. Avoid repeated freeze. Uniprot No. O75832 Immunogen Recombinant Human 26S proteasome non-ATPase regulatory subunit 10 protein (1-226AA) Raised In Rabbit Species Reactivity Human Tested Applications ELISA Relevance Acts as a chaperone during the assembly of the 26S proteasome, specifically of the PA700/19S regulatory complex (RC). In the initial step of the base subcomplex assembly is part of an intermediate PSMD10:PSMC4:PSMC5:PAAF1 module which probably assembles with a PSMD5:PSMC2:PSMC1:PSMD2 module. Independently of the proteasome, regulates EGF-induced AKT activation through inhibition of the RHOA/ROCK/PTEN pahway, leading to prolonged AKT activation. Plays an important role in RAS-induced tumorigenesis. Acts as an proto-oncoprotein by being involved in negative regulation of tumor suppressors RB1 and p53/TP53. Overexpression is leading to phosphorylation of RB1 and proteasomal degradation of RB1. Regulates CDK4-mediated phosphorylation of RB1 by competing with CDKN2A for binding with CDK4. Facilitates binding of MDM2 to p53/TP53 and the mono- and polyubiquitination of p53/TP53 by MDM2 suggesting a function in targeting the TP53:MDM2 complex to the 26S proteasome.
    [Show full text]
  • Cancer Vulnerabilities Unveiled by Genomic Loss
    Cancer Vulnerabilities Unveiled by Genomic Loss The MIT Faculty has made this article openly available. Please share how this access benefits you. Your story matters. Citation Nijhawan, Deepak, Travis I. Zack, Yin Ren, Matthew R. Strickland, Rebecca Lamothe, Steven E. Schumacher, Aviad Tsherniak, et al. “Cancer Vulnerabilities Unveiled by Genomic Loss.” Cell 150, no. 4 (August 2012): 842–854. © 2012 Elsevier Inc. As Published http://dx.doi.org/10.1016/j.cell.2012.07.023 Publisher Elsevier B.V. Version Final published version Citable link http://hdl.handle.net/1721.1/91899 Terms of Use Article is made available in accordance with the publisher's policy and may be subject to US copyright law. Please refer to the publisher's site for terms of use. Cancer Vulnerabilities Unveiled by Genomic Loss Deepak Nijhawan,1,2,7,9,10 Travis I. Zack,1,2,3,9 Yin Ren,5 Matthew R. Strickland,1 Rebecca Lamothe,1 Steven E. Schumacher,1,2 Aviad Tsherniak,2 Henrike C. Besche,4 Joseph Rosenbluh,1,2,7 Shyemaa Shehata,1 Glenn S. Cowley,2 Barbara A. Weir,2 Alfred L. Goldberg,4 Jill P. Mesirov,2 David E. Root,2 Sangeeta N. Bhatia,2,5,6,7,8 Rameen Beroukhim,1,2,7,* and William C. Hahn1,2,7,* 1Departments of Cancer Biology and Medical Oncology, Dana Farber Cancer Institute, Boston, MA 02215, USA 2Broad Institute of Harvard and MIT, Cambridge, MA 02142, USA 3Biophysics Program, Harvard University, Boston, MA 02115, USA 4Department of Cell Biology, Harvard Medical School, 240 Longwood Avenue, Boston, MA 02115, USA 5Harvard-MIT Division of Health Sciences and Technology 6David H.
    [Show full text]
  • PSMD10 Antibody (Center) Purified Rabbit Polyclonal Antibody (Pab) Catalog # AW5126
    10320 Camino Santa Fe, Suite G San Diego, CA 92121 Tel: 858.875.1900 Fax: 858.622.0609 PSMD10 Antibody (Center) Purified Rabbit Polyclonal Antibody (Pab) Catalog # AW5126 Specification PSMD10 Antibody (Center) - Product Information Application IF, WB, IHC-P,E Primary Accession O75832 Other Accession Q9Z2X2 Reactivity Human Predicted Mouse Host Rabbit Clonality Polyclonal Calculated MW H=24,16;M=25;Ra t=25 KDa Isotype Rabbit Ig Antigen Source HUMAN PSMD10 Antibody (Center) - Additional Information Gene ID 5716 Fluorescent image of Hela cells stained with PSMD10 Antibody (Center)(Cat#AW5126). Antigen Region AW5126 was diluted at 1:25 dilution. An 43-76 Alexa Fluor 488-conjugated goat anti-rabbit lgG at 1:400 dilution was used as the Other Names secondary antibody (green). Cytoplasmic 26S proteasome non-ATPase regulatory subunit 10, 26S proteasome regulatory actin was counterstained with Alexa Fluor® subunit p28, Gankyrin, p28(GANK), PSMD10 555 conjugated with Phalloidin (red). Dilution IF~~1:25 WB~~ 1:1000 IHC-P~~1:25 Target/Specificity This PSMD10 antibody is generated from a rabbit immunized with a KLH conjugated synthetic peptide between 43-76 amino acids from the Central region of human PSMD10. Format Purified polyclonal antibody supplied in PBS with 0.09% (W/V) sodium azide. This antibody is purified through a protein A column, followed by peptide affinity purification. Western blot analysis of lysates from MCF-7, PC-3, K562 cell line (from left to right), using Storage PSMD10 Antibody (Center)(Cat. #AW5126). Page 1/3 10320 Camino Santa Fe, Suite G San Diego, CA 92121 Tel: 858.875.1900 Fax: 858.622.0609 Maintain refrigerated at 2-8°C for up to 2 AW5126 was diluted at 1:1000 at each lane.
    [Show full text]
  • The Viral Oncoproteins Tax and HBZ Reprogram the Cellular Mrna Splicing Landscape
    bioRxiv preprint doi: https://doi.org/10.1101/2021.01.18.427104; this version posted January 18, 2021. 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 viral oncoproteins Tax and HBZ reprogram the cellular mRNA splicing landscape Charlotte Vandermeulen1,2,3, Tina O’Grady3, Bartimee Galvan3, Majid Cherkaoui1, Alice Desbuleux1,2,4,5, Georges Coppin1,2,4,5, Julien Olivet1,2,4,5, Lamya Ben Ameur6, Keisuke Kataoka7, Seishi Ogawa7, Marc Thiry8, Franck Mortreux6, Michael A. Calderwood2,4,5, David E. Hill2,4,5, Johan Van Weyenbergh9, Benoit Charloteaux2,4,5,10, Marc Vidal2,4*, Franck Dequiedt3*, and Jean-Claude Twizere1,2,11* 1Laboratory of Viral Interactomes, GIGA Institute, University of Liege, Liege, Belgium.2Center for Cancer Systems Biology (CCSB), Dana-Farber Cancer Institute, Boston, MA, USA.3Laboratory of Gene Expression and Cancer, GIGA Institute, University of Liege, Liege, Belgium.4Department of Genetics, Blavatnik Institute, Harvard Medical School, Boston, MA, USA. 5Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, MA, USA.6Laboratory of Biology and Modeling of the Cell, CNRS UMR 5239, INSERM U1210, University of Lyon, Lyon, France.7Department of Pathology and Tumor Biology, Kyoto University, Japan.8Unit of Cell and Tissue Biology, GIGA Institute, University of Liege, Liege, Belgium.9Laboratory of Clinical and Epidemiological Virology, Rega Institute for Medical Research, Department of Microbiology, Immunology and Transplantation, Catholic University of Leuven, Leuven, Belgium.10Department of Human Genetics, CHU of Liege, University of Liege, Liege, Belgium.11Lead Contact. *Correspondence: [email protected]; [email protected]; [email protected] bioRxiv preprint doi: https://doi.org/10.1101/2021.01.18.427104; this version posted January 18, 2021.
    [Show full text]
  • Protein Homeostasis Networks and the Use of Yeast to Guide Interventions in Alzheimer’S Disease
    International Journal of Molecular Sciences Review Protein Homeostasis Networks and the Use of Yeast to Guide Interventions in Alzheimer’s Disease Sudip Dhakal and Ian Macreadie * School of Science, RMIT University, Bundoora, Victoria 3083, Australia; [email protected] * Correspondence: [email protected]; Tel.: +61-3-9925-6627 Received: 29 September 2020; Accepted: 26 October 2020; Published: 28 October 2020 Abstract: Alzheimer’s Disease (AD) is a progressive multifactorial age-related neurodegenerative disorder that causes the majority of deaths due to dementia in the elderly. Although various risk factors have been found to be associated with AD progression, the cause of the disease is still unresolved. The loss of proteostasis is one of the major causes of AD: it is evident by aggregation of misfolded proteins, lipid homeostasis disruption, accumulation of autophagic vesicles, and oxidative damage during the disease progression. Different models have been developed to study AD, one of which is a yeast model. Yeasts are simple unicellular eukaryotic cells that have provided great insights into human cell biology. Various yeast models, including unmodified and genetically modified yeasts, have been established for studying AD and have provided significant amount of information on AD pathology and potential interventions. The conservation of various human biological processes, including signal transduction, energy metabolism, protein homeostasis, stress responses, oxidative phosphorylation, vesicle trafficking, apoptosis, endocytosis, and ageing, renders yeast a fascinating, powerful model for AD. In addition, the easy manipulation of the yeast genome and availability of methods to evaluate yeast cells rapidly in high throughput technological platforms strengthen the rationale of using yeast as a model.
    [Show full text]
  • Proteasome Dysfunction Induces Muscle Growth Defects and Protein Aggregation
    ß 2014. Published by The Company of Biologists Ltd | Journal of Cell Science (2014) 127, 5204–5217 doi:10.1242/jcs.150961 RESEARCH ARTICLE Proteasome dysfunction induces muscle growth defects and protein aggregation Yasuo Kitajima1,2,§, Yoshitaka Tashiro3,`,§, Naoki Suzuki1,*,§,", Hitoshi Warita1, Masaaki Kato1, Maki Tateyama1, Risa Ando1, Rumiko Izumi1, Maya Yamazaki4, Manabu Abe4, Kenji Sakimura4, Hidefumi Ito5, Makoto Urushitani3, Ryoichi Nagatomi2, Ryosuke Takahashi3 and Masashi Aoki1," ABSTRACT rate-limiting enzymes (Glass, 2010; Jagoe and Goldberg, 2001). Proteasomal proteolysis is important in several organs; for The ubiquitin–proteasome and autophagy–lysosome pathways are example, proteasome inhibition using MG-132 leads to the the two major routes of protein and organelle clearance. The role of cytoplasmic aggregation of TAR DNA-binding protein 43 (TDP- the proteasome pathway in mammalian muscle has not been 43) in cultured hippocampal and cortical neurons and in examined in vivo. In this study, we report that the muscle-specific immortalized motor neurons (van Eersel et al., 2011). Similarly, deletion of a crucial proteasomal gene, Rpt3 (also known as the depletion of the 26S proteasome in mouse brain neurons Psmc4), resulted in profound muscle growth defects and a causes neurodegeneration (Bedford et al., 2008). decrease in force production in mice. Specifically, developing The loss of skeletal muscle mass in humans at an older age, muscles in conditional Rpt3-knockout animals showed which is called sarcopenia, is a rapidly growing health issue dysregulated proteasomal activity. The autophagy pathway was worldwide (Vellas et al., 2013). The regulation of skeletal muscle upregulated, but the process of autophagosome formation was mass largely depends on protein synthesis and degradation impaired.
    [Show full text]
  • Alterations of the Pro-Survival Bcl-2 Protein Interactome in Breast Cancer
    bioRxiv preprint doi: https://doi.org/10.1101/695379; this version posted July 12, 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 Alterations of the pro-survival Bcl-2 protein interactome in 2 breast cancer at the transcriptional, mutational and 3 structural level 4 5 Simon Mathis Kønig1, Vendela Rissler1, Thilde Terkelsen1, Matteo Lambrughi1, Elena 6 Papaleo1,2 * 7 1Computational Biology Laboratory, Danish Cancer Society Research Center, 8 Strandboulevarden 49, 2100, Copenhagen 9 10 2Translational Disease Systems Biology, Faculty of Health and Medical Sciences, Novo 11 Nordisk Foundation Center for Protein Research University of Copenhagen, Copenhagen, 12 Denmark 13 14 Abstract 15 16 Apoptosis is an essential defensive mechanism against tumorigenesis. Proteins of the B-cell 17 lymphoma-2 (Bcl-2) family regulates programmed cell death by the mitochondrial apoptosis 18 pathway. In response to intracellular stresses, the apoptotic balance is governed by interactions 19 of three distinct subgroups of proteins; the activator/sensitizer BH3 (Bcl-2 homology 3)-only 20 proteins, the pro-survival, and the pro-apoptotic executioner proteins. Changes in expression 21 levels, stability, and functional impairment of pro-survival proteins can lead to an imbalance 22 in tissue homeostasis. Their overexpression or hyperactivation can result in oncogenic effects. 23 Pro-survival Bcl-2 family members carry out their function by binding the BH3 short linear 24 motif of pro-apoptotic proteins in a modular way, creating a complex network of protein- 25 protein interactions.
    [Show full text]
  • Supplementary Table 4: the Association of the 26S Proteasome
    Supplementary Material (ESI) for Molecular BioSystems This journal is (c) The Royal Society of Chemistry, 2009 Supplementary Table 4: The association of the 26S proteasome and tumor progression/metastasis Note: the associateion between cancer and the 26S proteasome genes has been manually checked in PubMed a) GSE2514 (Lung cancer, 20 tumor and 19 normal samples; 25 out of 43 26S proteasome genes were mapped on the microarray platform. FWER p-value: 0.02) Entrez GeneID Gene Symbol RANK METRIC SCORE* Genes have been reported in cancer 10213 PSMD14 0.288528293 5710 PSMD4 0.165639699 Kim et al., Mol Cancer Res., 6:426, 2008 5713 PSMD7 0.147187442 5721 PSME2 0.130215749 5717 PSMD11 0.128598183 Deng et al., Breast Cancer Research and Treatment, 104:1067, 2007 5704 PSMC4 0.123157509 5706 PSMC6 0.115970835 5716 PSMD10 0.112173758 Mayer et al., Biochem Society Transaction, 34:746, 2006 5700 PSMC1 0.0898761 Kim et al., Mol Cancer Res., 6:426, 2008 5701 PSMC2 0.081513479 Cui et al., Proteomics, 6:498, 2005 5709 PSMD3 0.071682706 5719 PSMD13 0.071118504 7415 VCP 0.060464829 9861 PSMD6 0.055711303 Ren et al., Oncogene, 19:1419, 2000 5720 PSME1 0.052469168 5714 PSMD8 0.047414459 Deng et al., Breast Cancer Research and Treatment, 104:1067, 2007 5702 PSMC3 0.046327863 Pollice et al., JBC, 279:6345, 2003 6184 RPN1 0.043426223 55559 UCHL5IP 0.041885283 5705 PSMC5 0.041615516 5715 PSMD9 0.033147983 5711 PSMD5 0.030562362 Deng et al., Breast Cancer Research and Treatment, 104:1067, 2007 10197 PSME3 0.015149679 Roessler et al., Molecular & Cellular Proteomics 5:2092, 2006 5718 PSMD12 -0.00983229 Cui et al., Proteomics, 6:498, 2005 9491 PSMF1 -0.069156095 *Positive rank metric score represent that a gene is highly expressed in tumors.
    [Show full text]