Doctorat De L'université De Toulouse

Total Page:16

File Type:pdf, Size:1020Kb

Doctorat De L'université De Toulouse En vue de l'obtention du DOCTORAT DE L'UNIVERSITÉ DE TOULOUSE Délivré par : Institut National Polytechnique de Toulouse (Toulouse INP) Discipline ou spécialité : Pathologie, Toxicologie, Génétique et Nutrition Présentée et soutenue par : Mme MARIA EUGENIA MARTI MARIMON le vendredi 9 novembre 2018 Titre : 3D genome conformation and gene expression in fetal pig muscle at late gestation Ecole doctorale : Sciences Ecologiques, Vétérinaires, Agronomiques et Bioingénieries (SEVAB) Unité de recherche : Génétique, Physiologie et Systèmes d'Elevage (GenPhySE) Directeur(s) de Thèse : MME MARTINE YERLE M. SYLVAIN FOISSAC Rapporteurs : M. FREDERIC BANTIGNIES, CNRS Mme CAROLE CHARLIER, UNIVERSITE DE LIEGE Membre(s) du jury : M. OLIVIER GADAL, CNRS TOULOUSE, Président Mme MARTINE YERLE, INRA TOULOUSE, Membre M. SYLVAIN FOISSAC, INRA TOULOUSE, Membre Acknowledgments I would like in first place to express my gratitude to the members of the jury for the time they spent evaluating this work. Thanks to Dr. Carole Charlier and Dr. Frédéric Bantignies for agreeing to be the reviewers of this thesis and for their kind and helpful feedback on my thesis. I would like to thank also to Dr. Olivier Gadal and Dr. Roderic Guigo for accepting to be part of the thesis committee. Je voudrais remercier tout spécialement mes trois encadrants. Merci Martine d’avoir accepté d’encadrer cette thèse et de t’être rendue disponible quand j’en avais besoin malgré toutes tes responsabilités. Merci aussi d’avoir veillé à ce que cette thèse ait bien été menée à terme. Sylvain, un grand merci pour toute ton aide, tes conseils et ta patience. Oui, il a fallu l’être avec cette thésarde débutante en bioinfo dont tu as hérité. Mais surtout, Sylvain, merci pour ton optimisme et pour avoir su me remonter le moral dans les moments difficiles. Enfin, Yvette, je te remercie beaucoup pour tous ces échanges dans nos bureaux qui ont fait naitre des questions et des idées qui ont permis d’améliorer mon travail, ainsi que pour ces petits coups de pouce pour les manips (j’ai adoré travailler avec toi au labo). D’autres personnes ont énormément contribué à mon travail et je leur en serai toujours reconnaissante. Tout particulièrement Nathalie V., merci à toi pour ton aide inestimable dans toutes les analyses biostatisiques, sans lesquelles ce travail n’aurait pas été possible. Malgré ta charge de travail, tu as toujours su trouver le temps pour m’aider. Je voudrais aussi remercier Pierre Neuvial, responsable du projet SCALES qui a servi à financer une bonne partie du séquençage permettant d’améliorer la qualité de mes données. Merci également Laurence L. pour nos échanges et pour ton implication dans mon premier article, j’ai vraiment apprécié de collaborer avec toi. Sarah, merci pour tous ces petits coups de main et pour ta gentillesse. Hervé, tu as été présent dans la plupart des moments clés de mon parcours à l’INRA depuis mon arrivée. Tu as été la toute première personne rencontrée, et qui de plus parlait l’espagnol!). C’est en partie grâce à toi que je me suis inscrite au master, que j’ai prolongé mon CDD avec Alain V. (merci à toi aussi), et que ce projet a vu le jour (je me souviens encore de cette réunion improvisée au patio avec Sylvain où nous avons établi les bases de cette thèse). Sans votre aide, je ne serais pas là aujourd’hui en train d’écrire ces lignes. Merci bien sûr à tous les membres de mon équipe Cytogène pour votre accueil chaleureux et votre soutien. Enfin, merci à Florence M., David R., Matthias Z., Diane E., Noémie T., Lisa B., Katia F., Nicolas M. et tous ceux que j’ai pu oublier pour leurs petites mais précieuses contributions. La vie au labo n’aurait pas été la même sans « mes filles adorées », Manu, Lisa, Laure, Nathalie M. et Sonia, ainsi que sans toi Valentin (une petite pensée pour toi, même si tu as quitté le labo il y a longtemps). Yvette et Sylvain, je reviens à vous car plus qu’encadrants, vous êtes devenus de chers amis sur le plan personnel. Merci à tous pour tous ces moments partagés non seulement au travail mais aussi en soirées, vacances, etc., je vous porterai toujours dans mon cœur. Sachez que ces petites pauses- cigarette au patio avec vous les filles ainsi qu’avec Pitou, Yann, David, Sylvain, Juliette… vont énormément me manquer. Enfin un grand merci en général à tous les membres du ex-LGC qui depuis mon arrivée m’ont fait me sentir comme chez moi et qui, pour une étrangère comme moi, sont devenus une grande famille. A tous mes amis, spécialement aux « Toulousanos », et à ma famille un très grand merci; aux premiers pour me donner l’envie de rester en France, et pour me distraire et/ou me supporter quand la thèse devenait difficile; et à la deuxième, pour son aide inconditionnelle. ”Gràcies mamà i papà, per tota la vostra ajuda i per creure sempre en mi: Si és que tinc la sort de tindre els millors pares del món! Ana i Dani gràcies també a vosaltres per estar sempre a l’escolta encara que la distància no ens permeta parlar més sovint”. Enfin, en dernier, mais le plus important, Stéphane, je ne pourrais pas trouver tous les mots nécessaires pour te remercier mais je vais essayer... Depuis le début tu as été toujours à mes côtés, pour les bons mais aussi pour les mauvais moments. Tu as souffert pour moi plus que moi-même dans mes plus grandes périodes de stress et tu t’es réjoui et senti fier de moi à chaque petit succès que j’ai pu avoir. Sans jamais lâcher et toujours prêt à m’aider et à me faire sourire, je ne peux pas imaginer un meilleur compagnon dans ma vie. Table of contents 1 Abstract ................................................................................................................ 11 2 General introduction ........................................................................................... 13 3 Bibliographic review ........................................................................................... 17 3.1 Chapter 1. Breeding context ..................................................................................... 17 3.1.1 Early mortality: a major breeding issue in pig farming ....................................... 17 3.1.1.1 Background ................................................................................................. 17 3.1.1.2 Selection towards prolificacy ...................................................................... 17 3.1.1.3 Critical factors of piglets mortality ............................................................. 19 3.1.2 Maturity and survival ........................................................................................... 19 3.1.2.1 Critical factors for piglets survival ............................................................. 19 3.1.2.2 Piglet’s maturity .......................................................................................... 21 3.1.3 The role of muscle maturity in survival at birth ................................................... 21 3.1.3.1 Myogenesis: the fetal skeletal muscle development ................................... 21 3.1.3.2 Peculiarities of pig skeletal myogenesis and muscle metabolism .............. 25 3.1.3.3 Muscle and maturity ................................................................................... 27 3.2 Chapter 2. Muscle transcriptome studies ................................................................. 29 3.2.1 Functional Annotation of porcine genome ........................................................... 29 3.2.1.1 Main efforts in pig genome sequencing and annotation ............................. 29 3.2.2 Transcriptome technologies and approaches ........................................................ 31 3.2.2.1 DNA microarray and RNA-seq .................................................................. 31 3.2.2.2 Co-expression networks .............................................................................. 33 3.2.3 Muscle transcriptome studies in pigs ................................................................... 35 3.3 Chapter 3. Nuclear architecture ................................................................................ 37 3.3.1 Higher order genome organization ....................................................................... 37 3.3.1.1 Generalities ................................................................................................. 37 3.3.1.2 Chromosome territories .............................................................................. 39 3.3.1.3 NPCs, LADs, NADs, TFs and PcG domains .............................................. 41 3.3.1.4 A and B compartments ................................................................................ 43 3.3.1.5 Topologically associated domains .............................................................. 45 3.3.2 Chromatin loops and gene-gene interactions ....................................................... 49 3.3.2.1 CTCF and cohesin functions ....................................................................... 49 3.3.2.2 Insulated neighborhoods (CTCF/cohesin-mediated loops) ........................ 49 3.3.2.3 Gene-gene interactions ................................................................................ 51 3.3.3 Dynamic organization of the genome .................................................................. 55 3.3.4 Single cell genome organization .......................................................................... 57 3.3.5 3D genome architecture and disease .................................................................... 57 3 3.3.6 3D genome architecture
Recommended publications
  • Spatial Chromatin Architecture Alteration By
    Sadowski et al. Genome Biology (2019) 20:148 https://doi.org/10.1186/s13059-019-1728-x RESEARCH Open Access Spatial chromatin architecture alteration by structural variations in human genomes at the population scale Michal Sadowski1,2, Agnieszka Kraft1,3, Przemyslaw Szalaj1,4,5, Michal Wlasnowolski1,3, Zhonghui Tang6, Yijun Ruan7* and Dariusz Plewczynski1,3* Abstract Background: The number of reported examples of chromatin architecture alterations involved in the regulation of gene transcription and in disease is increasing. However, no genome-wide testing has been performed to assess the abundance of these events and their importance relative to other factors affecting genome regulation. This is particularly interesting given that a vast majority of genetic variations identified in association studies are located outside coding sequences. This study attempts to address this lack by analyzing the impact on chromatin spatial organization of genetic variants identified in individuals from 26 human populations and in genome- wide association studies. Results: We assess the tendency of structural variants to accumulate in spatially interacting genomic segments and design an algorithm to model chromatin conformational changes caused by structural variations. We show that differential gene transcription is closely linked to the variation in chromatin interaction networks mediated by RNA polymerase II. We also demonstrate that CTCF-mediated interactions are well conserved across populations, but enriched with disease-associated SNPs. Moreover, we find boundaries of topological domains as relatively frequent targets of duplications, which suggest that these duplications can be an important evolutionary mechanism of genome spatial organization. Conclusions: This study assesses the critical impact of genetic variants on the higher-order organization of chromatin folding and provides insight into the mechanisms regulating gene transcription at the population scale, of which local arrangement of chromatin loops seems to be the most significant.
    [Show full text]
  • Epigenetics Bladder Cancer
    Published OnlineFirst March 3, 2016; DOI: 10.1158/2159-8290.CD-RW2016-040 RESEARCH WATCH Bladder Cancer Major finding: CDH1 mutations are Approach: Plasmacytoid variant tumors Impact: Plasmacytoid variant tumors specific to, and occur in the majority of, were molecularly characterized using are distinguishable from nonvariant plasmacytoid variant bladder cancers. whole-exome and targeted sequencing. bladder cancers by CDH1 mutations. PLASMACYTOID VARIANT BLADDER CANCERS HARBOR CDH1 MUTATIONS Bladder cancers have a large range of variant mor- was similar to lobular breast carcinoma and diffuse phologies, and tumors with wholly variant histolo- gastric carcinoma, both of which also have frequent gies exhibit worse survival rates. Variant histology CDH1 mutations. Patients with plasmacytoid vari- bladder cancers were not included in The Cancer ant cancer had a poorer survival rate, increased inci- Genome Atlas (TCGA), and, thus, the molecular dence of recurrence, and an increased incidence of events underlying these tumors remain poorly peritoneal spread compared to patients with other understood. Al-Ahmadie, Iyer, Lee, and colleagues types of bladder cancer. To determine if loss of used whole-exome and targeted sequencing to char- E-cadherin contributed to the aggressive phenotype, acterize plasmacytoid bladder cancer, an aggressive histologic CDH1 was deleted by CRISPR/Cas9 in two urothelial carci- variant cancer. Whole-exome sequencing of six plasmacytoid noma cell lines. CDH1 loss resulted in increased migration, variant bladder tumors revealed truncating somatic muta- suggesting that loss of E-cadherin may result in the increased tions in cadherin 1 (CDH1), which encodes E-cadherin, in all invasiveness of plasmacytoid variant tumors. These fi ndings of the tumors, whereas there were no CDH1-truncating muta- indicate that plasmacytoid variant bladder cancers are charac- tions in the 127 bladder cancers in TCGA.
    [Show full text]
  • Functional Analysis of Structural Variation in the 2D and 3D Human Genome
    FUNCTIONAL ANALYSIS OF STRUCTURAL VARIATION IN THE 2D AND 3D HUMAN GENOME by Conor Mitchell Liam Nodzak A dissertation submitted to the faculty of The University of North Carolina at Charlotte in partial fulfillment of the requirements for the degree of Doctor of Philosophy in Bioinformatics and Computational Biology Charlotte 2019 Approved by: Dr. Xinghua Mindy Shi Dr. Rebekah Rogers Dr. Jun-tao Guo Dr. Adam Reitzel ii c 2019 Conor Mitchell Liam Nodzak ALL RIGHTS RESERVED iii ABSTRACT CONOR MITCHELL LIAM NODZAK. Functional analysis of structural variation in the 2D and 3D human genome. (Under the direction of DR. XINGHUA MINDY SHI) The human genome consists of over 3 billion nucleotides that have an average distance of 3.4 Angstroms between each base, which equates to over two meters of DNA contained within the 125 µm3 volume diploid cell nuclei. The dense compaction of chromatin by the supercoiling of DNA forms distinct architectural modules called topologically associated domains (TADs), which keep protein-coding genes, noncoding RNAs and epigenetic regulatory elements in close nuclear space. It has recently been shown that these conserved chromatin structures may contribute to tissue-specific gene expression through the encapsulation of genes and cis-regulatory elements, and mutations that affect TADs can lead to developmental disorders and some forms of cancer. At the population-level, genomic structural variation contributes more to cumulative genetic difference than any other class of mutation, yet much remains to be studied as to how structural variation affects TADs. Here, we study the func- tional effects of structural variants (SVs) through the analysis of chromatin topology and gene activity for three trio families sampled from genetically diverse popula- tions from the Human Genome Structural Variation Consortium.
    [Show full text]
  • Dissecting Super-Enhancer Hierarchy Based on Chromatin Interactions
    ARTICLE DOI: 10.1038/s41467-018-03279-9 OPEN Dissecting super-enhancer hierarchy based on chromatin interactions Jialiang Huang1,2, Kailong Li3, Wenqing Cai2, Xin Liu3, Yuannyu Zhang3, Stuart H. Orkin2,4, Jian Xu3 & Guo-Cheng Yuan 1 Recent studies have highlighted super-enhancers (SEs) as important regulatory elements for gene expression, but their intrinsic properties remain incompletely characterized. Through an 1234567890():,; integrative analysis of Hi-C and ChIP-seq data, here we find that a significant fraction of SEs are hierarchically organized, containing both hub and non-hub enhancers. Hub enhancers share similar histone marks with non-hub enhancers, but are distinctly associated with cohesin and CTCF binding sites and disease-associated genetic variants. Genetic ablation of hub enhancers results in profound defects in gene activation and local chromatin landscape. As such, hub enhancers are the major constituents responsible for SE functional and struc- tural organization. 1 Department of Biostatistics and Computational Biology, Dana-Farber Cancer Institute and Harvard T.H. Chan School of Public Health, Boston, MA 02215, USA. 2 Division of Hematology/Oncology, Boston Childrens Hospital and Department of Pediatric Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA 02215, USA. 3 Department of Pediatrics, Childrens Medical Center Research Institute, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA. 4 Howard Hughes Medical Institute, Boston, MA 02215, USA. These authors contributed equally: Jialiang Huang, Kailong Li. Correspondence and requests for materials should be addressed to J.X. (email: [email protected]) or to G.-C.Y. (email: [email protected]) NATURE COMMUNICATIONS | (2018) 9:943 | DOI: 10.1038/s41467-018-03279-9 | www.nature.com/naturecommunications 1 ARTICLE NATURE COMMUNICATIONS | DOI: 10.1038/s41467-018-03279-9 nhancers are cis-acting DNA sequences that control cell- chromatin organization and gene activation.
    [Show full text]
  • Spatial Chromatin Architecture Alteration by Structural
    bioRxiv preprint doi: https://doi.org/10.1101/266981; this version posted September 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. Spatial Chromatin Architecture Alteration by Structural Variations in Human Genomes at Population Scale Michal Sadowski1,3, Agnieszka Kraft1,6, Przemyslaw Szalaj1,4,5, Michal Wlasnowolski1,7, Zhonghui Tang2, Yijun Ruan2*, Dariusz Plewczynski1,2,7,* 1Centre of New Technologies, University of Warsaw, Banacha 2c, 02-097 Warsaw, Poland, 2Jackson Laboratory for Genomic Medicine, Farmington, CT, USA, 3Faculty of Physics, University of Warsaw, Pasteura 5, 02-093 Warsaw, Poland, 4Centre for Innovative Research, Medical University of Bialystok, Bialystok, Poland, 5I-BioStat, Hasselt University, Belgium, 6Faculty of Mathematics, Informatics and Mechanics, University of Warsaw, Banacha 2, 02-097 Warsaw, Poland, 7Faculty of Mathematics and Information Science, Warsaw University of Technology, Warsaw, Poland. *Correspondence: [email protected]; [email protected] 1 bioRxiv preprint doi: https://doi.org/10.1101/266981; this version posted September 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. Abstract This genome-wide study is focused on the impact of structural variants identified in individuals from 26 human populations onto three-dimensional structures of their genomes. We assess the tendency of structural variants to accumulate in spatially interacting genomic segments and design a high-resolution computational algorithm to model the 3D conformational changes resulted by structural variations. We show that differential gene transcription is closely linked to variation in chromatin interaction networks mediated by RNA polymerase II.
    [Show full text]
  • Editing DNA Methylation in the Mammalian Genome
    Resource Editing DNA Methylation in the Mammalian Genome Graphical Abstract Authors X. Shawn Liu, Hao Wu, Xiong Ji, ..., Daniel Dadon, Richard A. Young, Rudolf Jaenisch Correspondence [email protected] In Brief DNA methylation patterns can be specifically altered in mammalian cells using CRISPR/Cas9-based approaches. Highlights d dCas9-Tet1 and -Dnmt3a enable precise editing of CpG methylation in vitro and in vivo d Targeted demethylation of BDNF promoter IV activates BDNF in neurons d Targeted enhancer demethylation facilitates MyoD-induced muscle cell reprogramming d Targeted de novo methylation of CTCF motifs alters CTCF- mediated gene loops Liu et al., 2016, Cell 167, 233–247 September 22, 2016 ª 2016 Elsevier Inc. http://dx.doi.org/10.1016/j.cell.2016.08.056 Resource Editing DNA Methylation in the Mammalian Genome X. Shawn Liu,1,4 Hao Wu,1,4 Xiong Ji,1,5 Yonatan Stelzer,1 Xuebing Wu,1 Szymon Czauderna,1,3 Jian Shu,1 Daniel Dadon,1,2 Richard A. Young,1,2 and Rudolf Jaenisch1,2,6,* 1Whitehead Institute for Biomedical Research, Cambridge, MA 02142, USA 2Department of Biology, Massachusetts Institute of Technology, Cambridge, MA 02142, USA 3Department of Medical Biotechnology, Faculty of Biochemistry, Biophysics, and Biotechnology, Jagiellonian University, 31-007 Krako´ w, Poland 4Co-first author 5Present address: School of Life Sciences, Peking-Tsinghua Center for Life Sciences, Peking University, Beijing 100871, China 6Lead Contact *Correspondence: [email protected] http://dx.doi.org/10.1016/j.cell.2016.08.056 SUMMARY resolution during different stages of normal development as well as disease (De Jager et al., 2014; Landau et al., 2014).
    [Show full text]
  • Noncoding RNA World: from Mechanism to Therapy 2021 21–23 JULY 2021 | ONLINE
    ncRNA Noncoding RNA World: 2021 From Mechanism to Therapy 21–23 JULY 2021 | ONLINE Program Booklet Organizer Gold Sponsors Award & Bronze Sponsor ncRNA Noncoding RNA World: From Mechanism to Therapy 2021 21–23 JULY 2021 | ONLINE Noncoding RNA World: From Mechanism to Therapy Virtual 21-23 July 2021 MDPI • Basel • Beijing • Wuhan • Barcelona • Belgrade • Manchester • Tianjin • Tokyo • Cluj • Toronto • Bucharest • Novi Sad • Krakow • Singapore • Nanjing Organizing Committees Conference Chairs Prof. Dr. Rory Johnson Prof. Dr. Ling-Ling Chen Scientific Committee Prof. Dr. Xichen Bao Prof. Dr. Assaf Bester Dr. Valerio Costa Dr. Phillip Grote Dr. Chung Chau Hon Dr. Reini Luco Prof. Dr. Lina Ma Prof. Dr. Eulàlia Martí Prof. Dr. Pieter Mestdagh Dr. Samir Ounzain Dr. Yue Wan Prof. Dr. Timofei Zatsepin Dr. Qiangfeng Cliff Zhang Organized by Conference Secretariat Ms. Charlotte Gardini Mr. Pablo Velázquez Email: [email protected] Welcome from the Chairs Dear Colleagues, It is a great pleasure to announce the upcoming conference “Noncoding RNA World: from Mechanism to Therapy”, to take place virtually (originally planned in Basel (Switzerland)) on 21-23 July 2021. Vast numbers of new and mysterious ncRNAs continue to be discovered across the tree of life. Although many key questions remain to be answered, nevertheless there are grounds for great optimism for the relevance of ncRNAs to biology and medicine. Powerful new tools are being refined to detect and manipulate ncRNAs with confidence and accuracy, including chemical structure probing, spatially-resolved sequencing, and of course CRISPR-based technologies. At the same time, rapid strides are being made in translating these findings to the clinic, not least thanks to oligonucleotide drugs.
    [Show full text]
  • Mitochondrial Nucleoid in Cardiac Homeostasis: Bidirectional Signaling of Mitochondria and Nucleus in Cardiac Diseases
    Basic Research in Cardiology (2021) 116:49 https://doi.org/10.1007/s00395-021-00889-1 REVIEW Mitochondrial nucleoid in cardiac homeostasis: bidirectional signaling of mitochondria and nucleus in cardiac diseases Yuliang Feng1 · Wei Huang2 · Christian Paul2 · Xingguo Liu3,4,5 · Sakthivel Sadayappan6 · Yigang Wang2 · Siim Pauklin1 Received: 10 June 2021 / Accepted: 20 July 2021 © The Author(s) 2021 Abstract Metabolic function and energy production in eukaryotic cells are regulated by mitochondria, which have been recognized as the intracellular ‘powerhouses’ of eukaryotic cells for their regulation of cellular homeostasis. Mitochondrial function is important not only in normal developmental and physiological processes, but also in a variety of human pathologies, includ- ing cardiac diseases. An emerging topic in the feld of cardiovascular medicine is the implication of mitochondrial nucleoid for metabolic reprogramming. This review describes the linear/3D architecture of the mitochondrial nucleoid (e.g., highly organized protein-DNA structure of nucleoid) and how it is regulated by a variety of factors, such as noncoding RNA and its associated R-loop, for metabolic reprogramming in cardiac diseases. In addition, we highlight many of the presently unsolved questions regarding cardiac metabolism in terms of bidirectional signaling of mitochondrial nucleoid and 3D chromatin structure in the nucleus. In particular, we explore novel techniques to dissect the 3D structure of mitochondrial nucleoid and propose new insights into the mitochondrial
    [Show full text]
  • Epigenetics Bladder Cancer
    Published OnlineFirst March 10, 2016; DOI: 10.1158/2159-8290.CD-RW2016-045 RESEARCH WATCH Bladder Cancer Major finding: CDH1 mutations are Approach: Plasmacytoid variant tumors Impact: Plasmacytoid variant tumors specific to, and occur in the majority of, were molecularly characterized using are distinguishable from nonvariant plasmacytoid variant bladder cancers. whole-exome and targeted sequencing. bladder cancers by CDH1 mutations. PLASMACYTOID VARIANT BLADDER CANCERS HARBOR CDH1 MUTATIONS Bladder cancers have a large range of variant mor- was similar to lobular breast carcinoma and diffuse phologies, and tumors with wholly variant histolo- gastric carcinoma, both of which also have frequent gies exhibit worse survival rates. Variant histology CDH1 mutations. Patients with plasmacytoid vari- bladder cancers were not included in The Cancer ant cancer had a poorer survival rate, increased inci- Genome Atlas (TCGA), and, thus, the molecular dence of recurrence, and an increased incidence of events underlying these tumors remain poorly peritoneal spread compared to patients with other understood. Al-Ahmadie, Iyer, Lee, and colleagues types of bladder cancer. To determine if loss of used whole-exome and targeted sequencing to char- E-cadherin contributed to the aggressive phenotype, acterize plasmacytoid bladder cancer, an aggressive histologic CDH1 was deleted by CRISPR/Cas9 in two urothelial carci- variant cancer. Whole-exome sequencing of six plasmacytoid noma cell lines. CDH1 loss resulted in increased migration, variant bladder tumors revealed truncating somatic muta- suggesting that loss of E-cadherin may result in the increased tions in cadherin 1 (CDH1), which encodes E-cadherin, in all invasiveness of plasmacytoid variant tumors. These fi ndings of the tumors, whereas there were no CDH1-truncating muta- indicate that plasmacytoid variant bladder cancers are charac- tions in the 127 bladder cancers in TCGA.
    [Show full text]
  • Spatial Genome Organization As a Framework for Somatic Alterations in Human Cancer
    bioRxiv preprint doi: https://doi.org/10.1101/179176; this version posted August 22, 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. Spatial Genome Organization as a Framework for Somatic Alterations in Human Cancer Kadir C. Akdemir1, Yilong Li2, Roel G. Verhaak1,3, Rameen Beroukhim4, Peter Campbell2, Lynda Chin5 and P. Andrew Futreal1* on behalf of the PCAWG-Structural Variation Working Group 1 Genomic Medicine Department, MD Anderson Cancer Center, Houston, TX 77054. USA 2 Wellcome Trust Sanger Institute, Hinxton CB10 1SA, Cambridge, UK 3 Division of Computational Biology, The Jackson Laboratory for Genomic Medicine, Farmington, CT 06031, USA 4 Departments of Medical Oncology and Cancer Biology, Dana-Farber Cancer Institute, Boston, MA 02115, USA, and Broad Institute, Cambridge, MA 02142, USA 5 Institute for Health Transformation, University of Texas, Houston, TX 77021, USA * Corresponding author ([email protected]) Genomic material within the nucleus is folded into successive layers in order to package and organize the long string of linear DNA. This hierarchical level of folding is closely associated with transcriptional regulation and DNA replication. Microscopic studies concluded that genome organization inside the nucleus is not random and chromosomes within the nucleus create territories1. More recent chromosome conformation studies have revealed that mammalian chromosomes are structured into tissue-invariant topologically associating domains (TADs) where the DNA within a given domain interacts more frequently together than with regions in other domains2,3.
    [Show full text]
  • Cis-Regulatory Mechanisms of Acute Phase Gene Expression
    Cis-regulatory Mechanisms of Acute Phase Gene Expression by Minggao Liang A thesis submitted in conformity with the requirements for the degree of Doctor of Philosophy The Department of Molecular Genetics University of Toronto © Copyright by Minggao Liang 2021 Cis-regulatory Mechanisms of Acute Phase Gene Expression Minggao Liang Doctor of Philosophy The Department of Molecular Genetics University of Toronto 2021 Abstract The accurate control of when and where genes are expressed is central to our understanding of development and disease. Elucidating the mechanisms that control this complex, multifaceted process is a major endeavor at the forefront of genetics and genome biology. The acute phase response (APR) is an evolutionarily conserved systemic response to inflammation that triggers rapid gene expression changes in the liver and other tissues. Genes induced during the APR are regulated via unique mechanisms that facilitate their rapid expression upon stimulation while maintaining their silencing under basal conditions. To dissect the mechanisms controlling APR gene expression, I employ a combination of epigenetic profiling, comparative genomics, and chromatin conformation capture to characterize the cis-regulation of a representative APR gene, PLAU. I investigate how PLAU is dysregulated in Quebec platelet disorder, a bleeding disorder in which a single tandem-duplication of PLAU causes its ectopic >100-fold overexpression in platelets and megakaryocytes. I demonstrate that the silencing of PLAU is normally facilitated by local 3-dimensional (3D) chromatin architecture which isolates it from the activity of nearby tissue-specific enhancers, while disruption of this architecture in QPD results in ectopic activation of PLAU by a conserved megakaryocyte enhancer.
    [Show full text]
  • CRISPR-Mediated Deletion of Prostate Cancer Risk-Associated CTCF Loop Anchors Identifies Repressive Chromatin Loops Yu Guo1, Andrew A
    Guo et al. Genome Biology (2018) 19:160 https://doi.org/10.1186/s13059-018-1531-0 RESEARCH Open Access CRISPR-mediated deletion of prostate cancer risk-associated CTCF loop anchors identifies repressive chromatin loops Yu Guo1, Andrew A. Perez1, Dennis J. Hazelett2, Gerhard A. Coetzee3, Suhn Kyong Rhie1* and Peggy J. Farnham1,4* Abstract Background: Recent genome-wide association studies (GWAS) have identified more than 100 loci associated with increased risk of prostate cancer, most of which are in non-coding regions of the genome. Understanding the function of these non-coding risk loci is critical to elucidate the genetic susceptibility to prostate cancer. Results: We generate genome-wide regulatory element maps and performed genome-wide chromosome confirmation capture assays (in situ Hi-C) in normal and tumorigenic prostate cells. Using this information, we annotate the regulatory potential of 2,181 fine-mapped prostate cancer risk-associated SNPs and predict a set of target genes that are regulated by prostate cancer risk-related H3K27Ac-mediated loops. We next identify prostate cancer risk-associated CTCF sites involved in long-range chromatin loops. We use CRISPR-mediated deletion to remove prostate cancer risk-associated CTCF anchor regions and the CTCF anchor regions looped to the prostate cancer risk-associated CTCF sites, and we observe up to 100-fold increases in expression of genes within the loops when the prostate cancer risk-associated CTCF anchor regions are deleted. Conclusions: We identify GWAS risk loci involved in long-range loops that function to repress gene expression within chromatin loops. Our studies provide new insights into the genetic susceptibility to prostate cancer.
    [Show full text]