Selective Induction of Apoptosis in HIV-Infected Macrophages

Total Page:16

File Type:pdf, Size:1020Kb

Selective Induction of Apoptosis in HIV-Infected Macrophages Selective Induction of Apoptosis in HIV-infected Macrophages by Simon Xin Min Dong A thesis submitted in partial fulfillment of the requirements for the Doctorate in Philosophy degree in Microbiology and Immunology Department of Biochemistry, Microbiology, and Immunology Faculty of Medicine University of Ottawa © Simon Xin Min Dong, Ottawa, Canada, 2020 Abstract The eradication of Human Immunodeficiency Virus (HIV) from infected patients is one of the major medical problems of our time, primarily due to HIV reservoir formation. Macrophages play important roles in HIV reservoir formation: once infected, they shield HIV against host anti-viral immune responses and anti-retroviral therapies, help viral spread and establish infection in anatomically protected sites. Thus, it is imperative to selectively induce the apoptosis of HIV-infected macrophages for a complete cure of this disease. I hypothesize that HIV infection dysregulates the expression of some specific genes, which is essential to the survival of infected host cells, and these genes can be targeted to selectively induce the apoptosis of HIV-infected macrophages. My objective is to identify the genes that can be targeted to eradicate HIV reservoir in macrophages, and to briefly elucidate the mechanism of cell death induced by targeting one of the identified genes. A four-step strategy was proposed to reach the goal. First, 90k shRNA lentivirus pool technology and microarray analysis were employed in a genome-wide screen of genes and 28 promising genes were found. Second, siRNA silencing was applied to validate these genes with 2 different HIV-1 viruses; as a result, 4 genes, Cox7a2, Znf484, Cdk2, and Cstf2t, were identified to be novel gene targets. Third, the 4 validated genes were characterized by literature review. Lastly, I briefly elucidated the mechanism of apoptosis induced by targeting one of the identified gene, Cox7a2. The results of this study can help to understand the mechanisms of HIV reservoir formation in macrophages and propose promising therapeutic targets for the eradication of HIV-infected macrophages from patients. II Acknowledgements First of all, I would like to thank my supervisor, Dr. Ashok Kumar, for his mentorship of my PhD studies. It was his diligence, intelligence, and full support that made me step forward day by day. This study was also under the instructions of Dr. Franco Vizeacoumar from the University of Saskatchewan; his timely and patient guidance made applying a novel technology to this research possible. Dr. Angela Crawley, Dr. Tommy Alain, and Dr. Jonathan Angel, who were my Thesis Advisory Committee members, also gave me pertinent advice during this study; their encouragement allowed me to grow as an independent medical researcher. In addition, Xiao Xiang, Niranjala Gajanayaka, Ramon Caballero, and Hamza Ali provided me everyday assistance. Duale Ahmed, David L. F. Roy, and Faria Ahmed also helped for some experiments and data analysis. Dr. Sandra Côte, Stephanie B. Schinkel, and Teslin Sandstrom at OHRI provided me assistance whenever I needed. Dr. Jason Fernandes, and Dr. Hapsa Mamady were my role models to be a medical researcher. Both Jasmine Huang and Ana Radar worked as part-time summer students for this project. Finally, I would also like to say many thanks to Lynn Kelly, Martine St-Jean, Stephen Baird, Keith Wilson, and many other staff at the Apoptosis Research Center of Children’s Hospital of Eastern Ontario. It was their timely assistance that made my work highly efficient so that I could progress rapidly. The selfless contributions from Katelynn J. Rowe (OHRI) were also indispensable to complete this research project. Here I would like to express my sincere gratitude and respects to all of them. III Table of Contents Abstract .................................................................................................................... II Acknowledgements ................................................................................................ III Table of Contents ................................................................................................... IV List of Abbreviations ............................................................................................ VIII List of Figures ........................................................................................................ XI List of Tables ........................................................................................................ XIII 1. General Introduction ........................................................................................... 1 1.1 A brief history on HIV/AIDS ........................................................................... 1 1.2 General information about HIV/AIDS ............................................................ 2 1.3 Transmission, infection, integration, and life cycle of HIV ........................ 6 1.4 Immune dysregulation due to massive depletion of CD4+ T cells ............ 9 1.5 The natural course of HIV infection without treatment ............................. 14 1.6 Anti-retroviral treatments after HIV infection ............................................ 16 1.7 Genetic diversity of HIV viruses ................................................................. 18 1.8 HIV reservoir formation after infection ...................................................... 19 1.9 The interplay between HIV and macrophages ........................................... 27 1.10 HIV infection dysregulates the gene expression of macrophages ........ 28 1.11 Roles of HIV proteins in apoptosis ........................................................... 30 1.12 Mitochondria in apoptosis of HIV-infected macrophages ...................... 33 1.13 Vpr-induced apoptosis of macrophages ................................................. 34 1.14 Selective induction of apoptosis in HIV-infected macrophages ............ 36 1.15 Targeting IAPs may induce apoptosis of HIV-infected macrophages ... 37 1.16 90k shRNA lentivirus pool for a genome-wide screen of target genes . 38 1.17 Approaches of targeting genes in primary human macrophages ......... 42 1.18 HIV viruses for in vitro infection of primary human macrophages ......... 44 2. Rationale ............................................................................................................ 49 3. Hypothesis ......................................................................................................... 49 4. Objectives .......................................................................................................... 49 5. Materials and Methods ...................................................................................... 50 5.1 Cell culture and differentiation of U937, THP-1, and U1 cell lines ........... 50 IV 5.2 Preparation of primary human MDMs and M1 polarization ...................... 50 5.3 HIV-1 production, in vitro infection of MDMs and HIV-p24 ELISA ............ 51 5.4 Preparation of 90k shRNA lentivirus pool ................................................. 52 5.5 Infecting myeloid lineage cells with 90k shRNA lentivirus pool .............. 52 5.6 Preparation of HIV-eGFP and HIV-HSA viruses from plasmid DNA ........ 53 5.7 Infection of primary MDMs with HIV-eGFP and HIV-HSA ......................... 54 5.8 siRNA transfection for transfection reagent selection ............................. 55 5.9 Detection of siRNA transfection rates ....................................................... 56 5.10 Monitoring cell viability and GFP expression.......................................... 56 5.11 Localization of transfected siRNA in primary human MDMs ................. 57 5.12 Flow cytometry analysis of PI staining and GFP expression................. 57 5.13 siRNA transfection of primary human MDMs for validation ................... 58 5.14 siRNA transfection of respirasome complex genes ............................... 58 5.15 Apoptosis of HIV-infected MDMs induced by siRNA silencing .............. 59 5.16 Propidium Iodide (PI) Staining to evaluate cell death ............................. 59 5.17 Isolation of HIV-HSA-infected MDMs ........................................................ 60 5.18 Analysis of caspase activation ................................................................. 60 5.19 TNF-α ELISA and cytokine ELISA array ................................................... 60 5.20 SDS-PAGE and Western blotting analyses.............................................. 61 5.21 Apoptosis of HIV-infected MDMs induced by chemical inhibitors ........ 62 5.22 ROS production detection ........................................................................ 63 5.23 Microscopy ................................................................................................. 64 5.24 Statistical analysis ..................................................................................... 64 5.25 Ethics statement ........................................................................................ 64 6. Results ............................................................................................................... 65 Chapter 6.1: Optimizing siRNA transfection into primary human macrophages by reversing Resveratrol-induced apoptosis ..................................................... 65 1. Introduction ................................................................................................. 65 2. Results .......................................................................................................
Recommended publications
  • Multi-Targeted Mechanisms Underlying the Endothelial Protective Effects of the Diabetic-Safe Sweetener Erythritol
    Multi-Targeted Mechanisms Underlying the Endothelial Protective Effects of the Diabetic-Safe Sweetener Erythritol Danie¨lle M. P. H. J. Boesten1*., Alvin Berger2.¤, Peter de Cock3, Hua Dong4, Bruce D. Hammock4, Gertjan J. M. den Hartog1, Aalt Bast1 1 Department of Toxicology, Maastricht University, Maastricht, The Netherlands, 2 Global Food Research, Cargill, Wayzata, Minnesota, United States of America, 3 Cargill RandD Center Europe, Vilvoorde, Belgium, 4 Department of Entomology and UCD Comprehensive Cancer Center, University of California Davis, Davis, California, United States of America Abstract Diabetes is characterized by hyperglycemia and development of vascular pathology. Endothelial cell dysfunction is a starting point for pathogenesis of vascular complications in diabetes. We previously showed the polyol erythritol to be a hydroxyl radical scavenger preventing endothelial cell dysfunction onset in diabetic rats. To unravel mechanisms, other than scavenging of radicals, by which erythritol mediates this protective effect, we evaluated effects of erythritol in endothelial cells exposed to normal (7 mM) and high glucose (30 mM) or diabetic stressors (e.g. SIN-1) using targeted and transcriptomic approaches. This study demonstrates that erythritol (i.e. under non-diabetic conditions) has minimal effects on endothelial cells. However, under hyperglycemic conditions erythritol protected endothelial cells against cell death induced by diabetic stressors (i.e. high glucose and peroxynitrite). Also a number of harmful effects caused by high glucose, e.g. increased nitric oxide release, are reversed. Additionally, total transcriptome analysis indicated that biological processes which are differentially regulated due to high glucose are corrected by erythritol. We conclude that erythritol protects endothelial cells during high glucose conditions via effects on multiple targets.
    [Show full text]
  • Aneuploidy: Using Genetic Instability to Preserve a Haploid Genome?
    Health Science Campus FINAL APPROVAL OF DISSERTATION Doctor of Philosophy in Biomedical Science (Cancer Biology) Aneuploidy: Using genetic instability to preserve a haploid genome? Submitted by: Ramona Ramdath In partial fulfillment of the requirements for the degree of Doctor of Philosophy in Biomedical Science Examination Committee Signature/Date Major Advisor: David Allison, M.D., Ph.D. Academic James Trempe, Ph.D. Advisory Committee: David Giovanucci, Ph.D. Randall Ruch, Ph.D. Ronald Mellgren, Ph.D. Senior Associate Dean College of Graduate Studies Michael S. Bisesi, Ph.D. Date of Defense: April 10, 2009 Aneuploidy: Using genetic instability to preserve a haploid genome? Ramona Ramdath University of Toledo, Health Science Campus 2009 Dedication I dedicate this dissertation to my grandfather who died of lung cancer two years ago, but who always instilled in us the value and importance of education. And to my mom and sister, both of whom have been pillars of support and stimulating conversations. To my sister, Rehanna, especially- I hope this inspires you to achieve all that you want to in life, academically and otherwise. ii Acknowledgements As we go through these academic journeys, there are so many along the way that make an impact not only on our work, but on our lives as well, and I would like to say a heartfelt thank you to all of those people: My Committee members- Dr. James Trempe, Dr. David Giovanucchi, Dr. Ronald Mellgren and Dr. Randall Ruch for their guidance, suggestions, support and confidence in me. My major advisor- Dr. David Allison, for his constructive criticism and positive reinforcement.
    [Show full text]
  • Binding Specificities of Human RNA Binding Proteins Towards Structured
    bioRxiv preprint doi: https://doi.org/10.1101/317909; this version posted March 1, 2019. 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. 1 Binding specificities of human RNA binding proteins towards structured and linear 2 RNA sequences 3 4 Arttu Jolma1,#, Jilin Zhang1,#, Estefania Mondragón4,#, Teemu Kivioja2, Yimeng Yin1, 5 Fangjie Zhu1, Quaid Morris5,6,7,8, Timothy R. Hughes5,6, Louis James Maher III4 and Jussi 6 Taipale1,2,3,* 7 8 9 AUTHOR AFFILIATIONS 10 11 1Department of Medical Biochemistry and Biophysics, Karolinska Institutet, Solna, Sweden 12 2Genome-Scale Biology Program, University of Helsinki, Helsinki, Finland 13 3Department of Biochemistry, University of Cambridge, Cambridge, United Kingdom 14 4Department of Biochemistry and Molecular Biology and Mayo Clinic Graduate School of 15 Biomedical Sciences, Mayo Clinic College of Medicine and Science, Rochester, USA 16 5Department of Molecular Genetics, University of Toronto, Toronto, Canada 17 6Donnelly Centre, University of Toronto, Toronto, Canada 18 7Edward S Rogers Sr Department of Electrical and Computer Engineering, University of 19 Toronto, Toronto, Canada 20 8Department of Computer Science, University of Toronto, Toronto, Canada 21 #Authors contributed equally 22 *Correspondence: [email protected] 23 24 25 SUMMARY 26 27 Sequence specific RNA-binding proteins (RBPs) control many important 28 processes affecting gene expression. They regulate RNA metabolism at multiple 29 levels, by affecting splicing of nascent transcripts, RNA folding, base modification, 30 transport, localization, translation and stability. Despite their central role in most 31 aspects of RNA metabolism and function, most RBP binding specificities remain 32 unknown or incompletely defined.
    [Show full text]
  • Mitoxplorer, a Visual Data Mining Platform To
    mitoXplorer, a visual data mining platform to systematically analyze and visualize mitochondrial expression dynamics and mutations Annie Yim, Prasanna Koti, Adrien Bonnard, Fabio Marchiano, Milena Dürrbaum, Cecilia Garcia-Perez, José Villaveces, Salma Gamal, Giovanni Cardone, Fabiana Perocchi, et al. To cite this version: Annie Yim, Prasanna Koti, Adrien Bonnard, Fabio Marchiano, Milena Dürrbaum, et al.. mitoXplorer, a visual data mining platform to systematically analyze and visualize mitochondrial expression dy- namics and mutations. Nucleic Acids Research, Oxford University Press, 2020, 10.1093/nar/gkz1128. hal-02394433 HAL Id: hal-02394433 https://hal-amu.archives-ouvertes.fr/hal-02394433 Submitted on 4 Dec 2019 HAL is a multi-disciplinary open access L’archive ouverte pluridisciplinaire HAL, est archive for the deposit and dissemination of sci- destinée au dépôt et à la diffusion de documents entific research documents, whether they are pub- scientifiques de niveau recherche, publiés ou non, lished or not. The documents may come from émanant des établissements d’enseignement et de teaching and research institutions in France or recherche français ou étrangers, des laboratoires abroad, or from public or private research centers. publics ou privés. Distributed under a Creative Commons Attribution| 4.0 International License Nucleic Acids Research, 2019 1 doi: 10.1093/nar/gkz1128 Downloaded from https://academic.oup.com/nar/advance-article-abstract/doi/10.1093/nar/gkz1128/5651332 by Bibliothèque de l'université la Méditerranée user on 04 December 2019 mitoXplorer, a visual data mining platform to systematically analyze and visualize mitochondrial expression dynamics and mutations Annie Yim1,†, Prasanna Koti1,†, Adrien Bonnard2, Fabio Marchiano3, Milena Durrbaum¨ 1, Cecilia Garcia-Perez4, Jose Villaveces1, Salma Gamal1, Giovanni Cardone1, Fabiana Perocchi4, Zuzana Storchova1,5 and Bianca H.
    [Show full text]
  • Mouse Cox7a2 Knockout Project (CRISPR/Cas9)
    https://www.alphaknockout.com Mouse Cox7a2 Knockout Project (CRISPR/Cas9) Objective: To create a Cox7a2 knockout Mouse model (C57BL/6J) by CRISPR/Cas-mediated genome engineering. Strategy summary: The Cox7a2 gene (NCBI Reference Sequence: NM_009945 ; Ensembl: ENSMUSG00000032330 ) is located on Mouse chromosome 9. 4 exons are identified, with the ATG start codon in exon 1 and the TAA stop codon in exon 4 (Transcript: ENSMUST00000034881). Exon 1~4 will be selected as target site. Cas9 and gRNA will be co-injected into fertilized eggs for KO Mouse production. The pups will be genotyped by PCR followed by sequencing analysis. Note: Exon 1 starts from about 0.4% of the coding region. Exon 1~4 covers 100.0% of the coding region. The size of effective KO region: ~4106 bp. The KO region does not have any other known gene. Page 1 of 8 https://www.alphaknockout.com Overview of the Targeting Strategy Wildtype allele 5' gRNA region gRNA region 3' 1 2 3 4 Legends Exon of mouse Cox7a2 Knockout region Page 2 of 8 https://www.alphaknockout.com Overview of the Dot Plot (up) Window size: 15 bp Forward Reverse Complement Sequence 12 Note: The 2000 bp section upstream of start codon is aligned with itself to determine if there are tandem repeats. Tandem repeats are found in the dot plot matrix. The gRNA site is selected outside of these tandem repeats. Overview of the Dot Plot (down) Window size: 15 bp Forward Reverse Complement Sequence 12 Note: The 2000 bp section downstream of stop codon is aligned with itself to determine if there are tandem repeats.
    [Show full text]
  • Rcf2 Revealed in Cryo-EM Structures of Hypoxic Isoforms of Mature Mitochondrial III-IV Supercomplexes
    Rcf2 revealed in cryo-EM structures of hypoxic isoforms of mature mitochondrial III-IV supercomplexes Andrew M. Hartleya, Brigitte Meunierb, Nikos Pinotsisa, and Amandine Maréchala,c,1 aInstitute of Structural and Molecular Biology, Birkbeck College, WC1E 7HX London, United Kingdom; bInstitute for Integrative Biology of the Cell (I2BC), CNRS, Commissariat à l’Énergie Atomique et aux Énergies Alternatives, Université Paris-Saclay, 91198 Gif-sur-Yvette, France; and cInstitute of Structural and Molecular Biology, University College London, WC1E 6BT London, United Kingdom Edited by Harry B. Gray, California Institute of Technology, Pasadena, CA, and approved March 12, 2020 (received for review November 25, 2019) The organization of the mitochondrial electron transport chain formed by CI, III, and IV in varying stoichiometries (11–14) and I– proteins into supercomplexes (SCs) is now undisputed; however, III2 SCs (11, 14). Overall, they reveal a conserved arrangement of their assembly process, or the role of differential expression CI and CIII across species, including strong protein–protein inter- isoforms, remain to be determined. In Saccharomyces cerevisiae, actions. Slightly different orientationsofCIIIrelativetoCIwithin cytochrome c oxidase (CIV) forms SCs of varying stoichiometry the respirasome have been reported that have been linked to in- bc with cytochrome 1 (CIII). Recent studies have revealed, in stability of the SC during the purification procedure, although the normoxic growth conditions, an interface made exclusively by possibility of a subpopulation of respirasomes in which CI is in the Cox5A, the only yeast respiratory protein that exists as one of deactivated state could not be excluded (9, 14). However, when two isoforms depending on oxygen levels.
    [Show full text]
  • Role of Cytochrome C Oxidase Nuclear-Encoded Subunits in Health and Disease
    Physiol. Res. 69: 947-965, 2020 https://doi.org/10.33549/physiolres.934446 REVIEW Role of Cytochrome c Oxidase Nuclear-Encoded Subunits in Health and Disease Kristýna ČUNÁTOVÁ1, David PAJUELO REGUERA1, Josef HOUŠTĚK1, Tomáš MRÁČEK1, Petr PECINA1 1Department of Bioenergetics, Institute of Physiology, Czech Academy of Sciences, Prague, Czech Republic Received February 2, 2020 Accepted September 13, 2020 Epub Ahead of Print November 2, 2020 Summary [email protected] and Tomáš Mráček, Department of Cytochrome c oxidase (COX), the terminal enzyme of Bioenergetics, Institute of Physiology CAS, Vídeňská 1083, 142 mitochondrial electron transport chain, couples electron transport 20 Prague 4, Czech Republic. E-mail: [email protected] to oxygen with generation of proton gradient indispensable for the production of vast majority of ATP molecules in mammalian Cytochrome c oxidase cells. The review summarizes current knowledge of COX structure and function of nuclear-encoded COX subunits, which may Energy demands of mammalian cells are mainly modulate enzyme activity according to various conditions. covered by ATP synthesis carried out by oxidative Moreover, some nuclear-encoded subunits possess tissue-specific phosphorylation apparatus (OXPHOS) located in the and development-specific isoforms, possibly enabling fine-tuning central bioenergetic organelle, mitochondria. OXPHOS is of COX function in individual tissues. The importance of nuclear- composed of five multi-subunit complexes embedded in encoded subunits is emphasized by recently discovered the inner mitochondrial membrane (IMM). Electron pathogenic mutations in patients with severe mitopathies. In transport from reduced substrates of complexes I and II to addition, proteins substoichiometrically associated with COX were cytochrome c oxidase (COX, complex IV, CIV) is found to contribute to COX activity regulation and stabilization of achieved by increasing redox potential of individual the respiratory supercomplexes.
    [Show full text]
  • Human Primitive Brain Displays Negative Mitochondrial-Nuclear Expression Correlation of Respiratory Genes
    Downloaded from genome.cshlp.org on October 7, 2021 - Published by Cold Spring Harbor Laboratory Press Research Human primitive brain displays negative mitochondrial-nuclear expression correlation of respiratory genes Gilad Barshad, Amit Blumberg, Tal Cohen, and Dan Mishmar Department of Life Sciences, Ben-Gurion University of the Negev, Beer Sheva 8410501, Israel Oxidative phosphorylation (OXPHOS), a fundamental energy source in all human tissues, requires interactions between mitochondrial (mtDNA)- and nuclear (nDNA)-encoded protein subunits. Although such interactions are fundamental to OXPHOS, bi-genomic coregulation is poorly understood. To address this question, we analyzed ∼8500 RNA-seq exper- iments from 48 human body sites. Despite well-known variation in mitochondrial activity, quantity, and morphology, we found overall positive mtDNA-nDNA OXPHOS genes’ co-expression across human tissues. Nevertheless, negative mtDNA- nDNA gene expression correlation was identified in the hypothalamus, basal ganglia, and amygdala (subcortical brain re- gions, collectively termed the “primitive” brain). Single-cell RNA-seq analysis of mouse and human brains revealed that this phenomenon is evolutionarily conserved, and both are influenced by brain cell types (involving excitatory/inhibitory neu- rons and nonneuronal cells) and by their spatial brain location. As the “primitive” brain is highly oxidative, we hypothesized that such negative mtDNA-nDNA co-expression likely controls for the high mtDNA transcript levels, which enforce tight OXPHOS regulation, rather than rewiring toward glycolysis. Accordingly, we found “primitive” brain-specific up-regula- tion of lactate dehydrogenase B (LDHB), which associates with high OXPHOS activity, at the expense of LDHA, which pro- motes glycolysis. Analyses of co-expression, DNase-seq, and ChIP-seq experiments revealed candidate RNA-binding proteins and CEBPB as the best regulatory candidates to explain these phenomena.
    [Show full text]
  • Oxidative-Phosphorylation Genes from the Mitochondrial and Nuclear
    bioRxiv preprint doi: https://doi.org/10.1101/136457; this version posted May 11, 2017. The copyright holder for this preprint (which was not certified by peer review) is the author/funder, who has granted bioRxiv a license to display the preprint in perpetuity. It is made available under aCC-BY-NC-ND 4.0 International license. Oxidative-phosphorylation genes from the mitochondrial and nuclear genomes co-express in all human tissues except for the ancient brain Gilad Barshad1, Amit Blumberg1 and Dan Mishmar1* Department of Life Sciences, Ben-Gurion University of the Negev, Beer Sheva 8410501, Israel *Corresponding author Address: Dan Mishmar, PhD Department of Life Sciences Ben-Gurion University of the Negev Beer Sheva 8410501, Israel Tel: +972-86461355 FAX: 972-86461356 Email: [email protected] 1 bioRxiv preprint doi: https://doi.org/10.1101/136457; this version posted May 11, 2017. The copyright holder for this preprint (which was not certified by peer review) is the author/funder, who has granted bioRxiv a license to display the preprint in perpetuity. It is made available under aCC-BY-NC-ND 4.0 International license. Abstract In humans, oxidative phosphorylation (OXPHOS), the cellular energy producer, harbors ~90 nuclear DNA (nDNA)- and mitochondrial DNA (mtDNA)-encoded subunits. Although nDNA- and mtDNA-encoded OXPHOS proteins physically interact, their transcriptional regulation profoundly diverges, thus questioning their co-regulation. To address mtDNA-nDNA gene co- expression, we analyzed ~8,500 RNA-seq Gene-Tissue-Expression (GTEx) experiments encompassing 48 human tissues. We found overall positive cross-tissue mtDNA-nDNA OXPHOS gene co-expression.
    [Show full text]
  • Supplemental Figure 1. Protein-Protein Interaction Network with Increased Expression in Fteb During the Luteal Phase
    Supplemental Figure 1. Protein-protein interaction network with increased expression in FTEb during the luteal phase. Supplemental Figure 2. Protein-protein interaction network with decreased expression in FTEb during luteal phase. LEGENDS TO SUPPLEMENTAL FIGURES Supplemental Figure 1. Protein-protein interaction network with increased expression in FTEb during the luteal phase. Submission of probe sets differentially expressed in the FTEb specimens that clustered with SerCa as well as those specifically altered in FTEb luteal samples to the online I2D database revealed overlapping networks of proteins with increased expression in the four FTEb samples and/or FTEb luteal samples overall. Proteins are represented by nodes, and known and predicted first-degree interactions are represented by solid lines. Genes encoding proteins shown as large ovals highlighted in blue were exclusively found in the first comparison (Manuscript Figure 2), whereas those highlighted in red were only found in the second comparison (Manuscript Figure 3). Genes encoding proteins shown as large ovals highlighted in black were found in both comparisons. The color of each node indicates the ontology of the corresponding protein as determined by the Online Predicted Human Interaction Database (OPHID) link with the NAViGaTOR software. Supplemental Figure 2. Protein-protein interaction network with decreased expression in FTEb during the luteal phase. Submission of probe sets differentially expressed in the FTEb specimens that clustered with SerCa as well as those specifically altered in FTEb luteal samples to the online I2D database revealed overlapping networks of proteins with decreased expression in the four FTEb samples and/or FTEb luteal samples overall. Proteins are represented by nodes, and known and predicted first-degree interactions are represented by solid lines.
    [Show full text]
  • Downloaded from Genome Reference Consortium (94)
    bioRxiv preprint doi: https://doi.org/10.1101/2020.05.05.076927; this version posted January 20, 2021. 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. Conserved long-range base pairings are associated with pre-mRNA processing of human genes Svetlana Kalmykova1,2, Marina Kalinina1, Stepan Denisov1, Alexey Mironov1, Dmitry Skvortsov3, Roderic Guigo´4, and Dmitri Pervouchine1,* 1Skolkovo Institute of Science and Technology, Moscow 143026, Russia 2Institute of Pathology, Charite´ Universitatsmedizin¨ Berlin, 10117, Berlin, Germany 3Faculty of Chemistry, Moscow State University, Moscow 119234, Russia 4Center for Genomic Regulation and UPF, Barcelona 08003, Spain *e-mail: [email protected] Abstract The ability of nucleic acids to form double-stranded structures is essential for all liv- ing systems on Earth. While DNA employs it for genome replication, RNA molecules fold into complicated secondary and tertiary structures. Current knowledge on functional RNA structures in human protein-coding genes is focused on locally-occurring base pairs. However, chemical crosslinking and proximity ligation experiments have demonstrated that long-range RNA structures are highly abundant. Here, we present the most complete to- date catalog of conserved long-range RNA structures in the human transcriptome, which consists of 916,360 pairs of conserved complementary regions (PCCRs). PCCRs tend to occur within introns proximally to splice sites, suppress intervening exons, circumscribe circular RNAs, and exert an obstructive effect on cryptic and inactive splice sites.
    [Show full text]
  • Oxidative-Phosphorylation Genes from the Mitochondrial and Nuclear
    bioRxiv preprint doi: https://doi.org/10.1101/136457; this version posted May 11, 2017. The copyright holder for this preprint (which was not certified by peer review) is the author/funder, who has granted bioRxiv a license to display the preprint in perpetuity. It is made available under aCC-BY-NC-ND 4.0 International license. Oxidative-phosphorylation genes from the mitochondrial and nuclear genomes co-express in all human tissues except for the ancient brain Gilad Barshad1, Amit Blumberg1 and Dan Mishmar1* Department of Life Sciences, Ben-Gurion University of the Negev, Beer Sheva 8410501, Israel *Corresponding author Address: Dan Mishmar, PhD Department of Life Sciences Ben-Gurion University of the Negev Beer Sheva 8410501, Israel Tel: +972-86461355 FAX: 972-86461356 Email: [email protected] 1 bioRxiv preprint doi: https://doi.org/10.1101/136457; this version posted May 11, 2017. The copyright holder for this preprint (which was not certified by peer review) is the author/funder, who has granted bioRxiv a license to display the preprint in perpetuity. It is made available under aCC-BY-NC-ND 4.0 International license. Abstract In humans, oxidative phosphorylation (OXPHOS), the cellular energy producer, harbors ~90 nuclear DNA (nDNA)- and mitochondrial DNA (mtDNA)-encoded subunits. Although nDNA- and mtDNA-encoded OXPHOS proteins physically interact, their transcriptional regulation profoundly diverges, thus questioning their co-regulation. To address mtDNA-nDNA gene co- expression, we analyzed ~8,500 RNA-seq Gene-Tissue-Expression (GTEx) experiments encompassing 48 human tissues. We found overall positive cross-tissue mtDNA-nDNA OXPHOS gene co-expression.
    [Show full text]