Genome Folding in Evolution and Disease

Total Page:16

File Type:pdf, Size:1020Kb

Genome Folding in Evolution and Disease Johannes Gutenberg University Mainz Faculty of Biology Institute of Organismic and Molecular Evolution Computational Biology and Data Mining Group Dissertation Genome folding in evolution and disease Jonas Ibn-Salem February, 2018 Jonas Ibn-Salem Genome folding in evolution and disease Dissertation, February, 2018 Johannes Gutenberg University Mainz Computational Biology and Data Mining Group Institute of Organismic and Molecular Evolution Faculty of Biology Ackermannweg 4 55128 and Mainz Contents Abstract 7 1 Introduction 9 1.1 Regulation of gene expression ...................... 9 1.2 Distal regulation by enhancers ...................... 10 1.3 Methods to probe the 3D chromatin architecture . 11 1.3.1 Microscopy-based techniques to visualize the genome in 3D . 11 1.3.2 Proximity-ligation based method to quantify chromatin inter- actions ............................... 11 1.4 Hierarchy of chromatin 3D structure . 13 1.4.1 Chromosomal territories and inter-chromosomal contacts . 13 1.4.2 A/B compartments ........................ 15 1.4.3 Topologically associating domains (TADs) . 15 1.4.4 Hierarchy of domain structures across genomic length scales . 17 1.4.5 Chromatin looping interactions . 17 1.4.6 TAD and loop formation by architectural proteins . 18 1.5 Dynamics of chromatin structure .................... 20 1.5.1 Dynamics across the cell cycle . 20 1.5.2 Dynamics across cell types and differntiation . 20 1.6 Evolution of chromatin organization . 21 1.7 Disruption of chromatin architecture in disease . 23 1.8 Aims of this thesis ............................ 24 1.9 Structure of this thesis .......................... 27 2 Paralog genes in the 3D genome architecture 29 Preamble ................................. 29 Abstract .................................. 29 2.1 Introduction ................................ 30 2.2 Materials and methods .......................... 31 2.2.1 Selection of pairs of paralog genes . 31 2.2.2 Enhancers to gene association . 32 2.2.3 Topological associating domains . 32 2.2.4 Hi-C interaction maps ...................... 32 2.2.5 Randomization .......................... 32 1 2.2.6 Statistical tests .......................... 34 2.3 Results ................................... 34 2.3.1 Distribution of paralog genes in the human genome . 34 2.3.2 Co-expression of paralog gene pairs across tissues . 35 2.3.3 Paralog genes share enhancers . 37 2.3.4 Co-localization of paralogs in TADs . 38 2.3.5 Distal paralog pairs are enriched for long-range chromatin contacts .............................. 40 2.3.6 Close paralogs have fewer contacts than expected . 40 2.3.7 Paralogs in mouse and dog genome . 41 2.3.8 Orthologs of human paralogs show conserved co-localization 43 2.4 Discussions ................................ 45 2.5 Conclusion ................................ 47 Acknowledgements ............................... 47 3 Stability of TADs in evolution 49 Preamble ................................. 49 Abstract .................................. 49 3.1 Introduction ................................ 50 3.2 Results ................................... 51 3.2.1 Identification of evolutionary rearrangement breakpoints from whole-genome alignments . 51 3.2.2 Rearrangement breakpoints are enriched at TAD boundaries . 53 3.2.3 Clusters of conserved non-coding elements are depleted for rearrangement breakpoints ................... 55 3.2.4 Rearranged TADs are associated with divergent gene expres- sion between species ....................... 55 3.3 Discussion ................................. 58 3.4 Conclusion ................................ 61 3.5 Methods .................................. 61 3.5.1 Rearrangement breakpoints from whole-genome alignments . 61 3.5.2 Topologically associating domains and contact domains . 62 3.5.3 Breakpoint distributions at TADs . 62 3.5.4 Quantification of breakpoint enrichment . 63 3.5.5 Expression data for mouse and human orthologs . 63 3.5.6 Classification of TADs and genes according to rearrangements and GRBs ............................. 64 3.5.7 Source code and implementation details . 64 Declarations ................................... 64 Availability of data and material ..................... 64 Competing interests ........................... 65 Authors‘ contributions .......................... 65 2 Contents Acknowledgments ............................ 65 4 Position effects of rearrangements in disease genomes 67 Preamble ................................. 67 Abstract .................................. 67 4.1 Introduction ................................ 68 4.2 Materials and Methods .......................... 69 4.2.1 Selection of subjects with apparently balanced chromosome abnormalities ........................... 70 4.2.2 Clinical descriptions of DGAP cases . 70 4.2.3 Analysis of genes bordering the rearrangement breakpoints . 71 4.2.4 Assessment of disrupted functional elements and chromatin interactions bordering rearrangement breakpoints . 72 4.2.5 Ontological analysis of genes neighboring breakpoints . 73 4.2.6 Quantitative real-time PCR ................... 73 4.2.7 Assessment of DGAP breakpoints overlapping with non- coding structural variants in public databases . 73 4.3 Results ................................... 74 4.3.1 Genomic characterization of non-coding breakpoints . 74 4.3.2 Identification of genes with potential position effects . 76 4.3.3 Identification of subjects with shared non-coding chromo- some alterations and phenotypes . 78 4.4 Discussion ................................. 80 Declarations ................................... 84 Acknowledgements ............................ 84 Web Resources .............................. 84 5 Prediction of chromatin looping interactions 87 Preamble ................................. 87 Abstract .................................. 87 5.1 Introduction ................................ 88 5.2 Results ................................... 89 5.2.1 CTCF motif pairs as candidate chromatin loop anchors . 89 5.2.2 Similarity of ChIP-seq signals at looping CTCF motifs . 91 5.2.3 Genomic sequence features of CTCF motif pairs are associ- ated with looping ......................... 91 5.2.4 Chromatin loop prediction using 7C . 93 5.2.5 Prediction performance evaluation . 93 5.2.6 Prediction performance of 7C with sequence features and sin- gle TF ChIP-seq data sets ..................... 94 5.2.7 Comparison of transcription factors by prediction performance 96 5.2.8 Prediction performance in other cell types and for different TFs 96 Contents 3 5.2.9 The high resolution of ChIP-nexus improves prediction perfor- mance ............................... 98 5.3 Discussion ................................. 99 5.4 Conclusion ................................101 5.5 Methods ..................................102 5.5.1 CTCF motifs in the human genome . 102 5.5.2 Loop interaction data for training and validation . 102 5.5.3 ChIP-seq datasets in GM12878 cells . 103 5.5.4 ChIP-seq data types . 103 5.5.5 ChIP-nexus data processing for RAD21 and SCM3 . 103 5.5.6 Similarity of ChIP-seq profiles as correlation of coverage around motifs . 104 5.5.7 Genomic sequence features of chromatin loops . 104 5.5.8 Chromatin Loop prediction model . 104 5.5.9 Training and validation of prediction model . 105 5.5.10 Analysis of prediction performance . 105 5.5.11 Implementation of 7C and compatibility to other tools . 105 Declarations ...................................106 Availability of data and material . 106 Competing interests . 106 Authors‘ contributions . 106 Acknowledgments ............................106 6 Discussion 107 6.1 Co-regulation of functionally related genes in TADs . 107 6.2 Evolution by gene duplications and altered regulatory environments 109 6.3 TADs are stable across large evolutionary time-scales . 111 6.4 Gene expression changes by altered TADs in disease . 112 6.5 Towards predicting regulatory pathomechanisms of structural variants115 6.6 Towards studying genome folding in specific conditions . 117 6.6.1 Interpretation of non-coding variants by interacting genes . 117 6.6.2 Cell type-specific regulatory interactions between enhancers and genes .............................118 6.6.3 Variability of long-range interactions across individuals . 118 6.6.4 Genome folding in single cells . 119 6.6.5 Constantly improving targeted experimental methods. 119 6.7 Molecular mechanisms driving genome folding . 120 6.8 Conclusions ................................121 A Supporting Information: Co-regulation of paralog genes 123 B Supplementary Data: Stability of TADs in evolution 135 B.1 Supplementary Tables . 135 4 Contents B.2 Supplementary Figures . 135 C Supplemental Data: Position effects of rearrangements in disease genomes 139 C.1 Supplemental Note ............................139 C.1.1 Case Reports . 139 C.1.2 Nucleotide Level Nomenclature for DGAP karyotypes . 143 C.2 Supplemental Figure . 145 C.3 Supplemental Table Legends . 146 D Supplemental Information: Prediction of chromatin looping interac- tions 151 D.1 Supplementary Tables . 151 D.2 Supplementary Figures . 151 E Contribution to individual publications 153 Zusammenfassung 155 Bibliography 157 Contents 5 Abstract The human genome is hierarchically folded in the three-dimensional nucleus. Pair- wise chromatin contacts cluster in discrete chromosomal regions, termed topolog- ically associating domains (TADs). Whether TADs play an essential role in gene expression regulation in evolution and genetic diseases, is analyzed in this thesis by computationally integrating genome-wide contact maps with various
Recommended publications
  • PARSANA-DISSERTATION-2020.Pdf
    DECIPHERING TRANSCRIPTIONAL PATTERNS OF GENE REGULATION: A COMPUTATIONAL APPROACH by Princy Parsana A dissertation submitted to The Johns Hopkins University in conformity with the requirements for the degree of Doctor of Philosophy Baltimore, Maryland July, 2020 © 2020 Princy Parsana All rights reserved Abstract With rapid advancements in sequencing technology, we now have the ability to sequence the entire human genome, and to quantify expression of tens of thousands of genes from hundreds of individuals. This provides an extraordinary opportunity to learn phenotype relevant genomic patterns that can improve our understanding of molecular and cellular processes underlying a trait. The high dimensional nature of genomic data presents a range of computational and statistical challenges. This dissertation presents a compilation of projects that were driven by the motivation to efficiently capture gene regulatory patterns in the human transcriptome, while addressing statistical and computational challenges that accompany this data. We attempt to address two major difficulties in this domain: a) artifacts and noise in transcriptomic data, andb) limited statistical power. First, we present our work on investigating the effect of artifactual variation in gene expression data and its impact on trans-eQTL discovery. Here we performed an in-depth analysis of diverse pre-recorded covariates and latent confounders to understand their contribution to heterogeneity in gene expression measurements. Next, we discovered 673 trans-eQTLs across 16 human tissues using v6 data from the Genotype Tissue Expression (GTEx) project. Finally, we characterized two trait-associated trans-eQTLs; one in Skeletal Muscle and another in Thyroid. Second, we present a principal component based residualization method to correct gene expression measurements prior to reconstruction of co-expression networks.
    [Show full text]
  • CHD4/Nurd Complex Regulates Complement Gene Expression And
    Shao et al. Clinical Epigenetics (2020) 12:31 https://doi.org/10.1186/s13148-020-00827-3 RESEARCH Open Access CHD4/NuRD complex regulates complement gene expression and correlates with CD8 T cell infiltration in human hepatocellular carcinoma Simin Shao1†, Haowei Cao1†, Zhongkun Wang1, Dongmei Zhou2, Chaoshen Wu1, Shu Wang1, Dian Xia1 and Daoyong Zhang1* Abstract Backgrounds: The NuRD (Nucleosome Remodeling and Deacetylation) complex is a repressive complex in gene transcription by modulating chromatin accessibility of target genes to transcription factors and RNA polymerase II. Although individual subunits of the complex have been implicated in many other cancer types, the complex’s role in human hepatocellular carcinoma (HCC) is not fully understood. More importantly, the NuRD complex has not yet been investigated as a whole in cancers. Methods: We analyzed the expression of the NuRD complex in HCC and evaluated the prognostic value of NuRD complex expression in HCC using the RNA-seq data obtained from the TCGA project. We examined the effect of CHD4 knockdown on HCC cell proliferation, apoptosis, migration, invasion, epithelial-mesenchymal transition, colony-forming ability, and on complement gene expression. We also performed bioinformatic analyses to investigate the correlation between the NuRD complex expression and immune infiltration. Results: We found that nine subunits, out of 14 subunits of the NuRD complex examined, were significantly overexpressed in HCC, and their expression levels were positively correlated with cancer progression. More importantly, our data also demonstrated that these subunits tended to be overexpressed as a whole in HCC. Subsequent studies demonstrated that knockdown of CHD4 in HCC cells inhibits cell proliferation, migration, invasion, and colony-forming ability and promotes apoptosis of HCC cells, indicating that the CHD4/NuRD complex plays oncogenic roles in HCC.
    [Show full text]
  • Cyclin D1 Is a Direct Transcriptional Target of GATA3 in Neuroblastoma Tumor Cells
    Oncogene (2010) 29, 2739–2745 & 2010 Macmillan Publishers Limited All rights reserved 0950-9232/10 $32.00 www.nature.com/onc SHORT COMMUNICATION Cyclin D1 is a direct transcriptional target of GATA3 in neuroblastoma tumor cells JJ Molenaar1,2, ME Ebus1, J Koster1, E Santo1, D Geerts1, R Versteeg1 and HN Caron2 1Department of Human Genetics, Academic Medical Center, University of Amsterdam, Amsterdam, The Netherlands and 2Department of Pediatric Oncology, Emma Kinderziekenhuis, Academic Medical Center, University of Amsterdam, Amsterdam, The Netherlands Almost all neuroblastoma tumors express excess levels of 2000). Several checkpoints normally prevent premature Cyclin D1 (CCND1) compared to normal tissues and cell-cycle progression and cell division. The crucial G1 other tumor types. Only a small percentage of these entry point is controlled by the D-type Cyclins that can neuroblastoma tumors have high-level amplification of the activate CDK4/6 that in turn phosphorylate the pRb Cyclin D1 gene. The other neuroblastoma tumors have protein. This results in a release of the E2F transcription equally high Cyclin D1 expression without amplification. factor that causes transcriptional upregulation of Silencing of Cyclin D1 expression was previously found to numerous genes involved in further progression of the trigger differentiation of neuroblastoma cells. Over- cell cycle (Sherr, 1996). expression of Cyclin D1 is therefore one of the most Neuroblastomas are embryonal tumors that originate frequent mechanisms with a postulated function in neuro- from precursor cells of the sympathetic nervous system. blastoma pathogenesis. The cause for the Cyclin D1 This tumor has a very poor prognosis and despite the overexpression is unknown.
    [Show full text]
  • A Computational Approach for Defining a Signature of Β-Cell Golgi Stress in Diabetes Mellitus
    Page 1 of 781 Diabetes A Computational Approach for Defining a Signature of β-Cell Golgi Stress in Diabetes Mellitus Robert N. Bone1,6,7, Olufunmilola Oyebamiji2, Sayali Talware2, Sharmila Selvaraj2, Preethi Krishnan3,6, Farooq Syed1,6,7, Huanmei Wu2, Carmella Evans-Molina 1,3,4,5,6,7,8* Departments of 1Pediatrics, 3Medicine, 4Anatomy, Cell Biology & Physiology, 5Biochemistry & Molecular Biology, the 6Center for Diabetes & Metabolic Diseases, and the 7Herman B. Wells Center for Pediatric Research, Indiana University School of Medicine, Indianapolis, IN 46202; 2Department of BioHealth Informatics, Indiana University-Purdue University Indianapolis, Indianapolis, IN, 46202; 8Roudebush VA Medical Center, Indianapolis, IN 46202. *Corresponding Author(s): Carmella Evans-Molina, MD, PhD ([email protected]) Indiana University School of Medicine, 635 Barnhill Drive, MS 2031A, Indianapolis, IN 46202, Telephone: (317) 274-4145, Fax (317) 274-4107 Running Title: Golgi Stress Response in Diabetes Word Count: 4358 Number of Figures: 6 Keywords: Golgi apparatus stress, Islets, β cell, Type 1 diabetes, Type 2 diabetes 1 Diabetes Publish Ahead of Print, published online August 20, 2020 Diabetes Page 2 of 781 ABSTRACT The Golgi apparatus (GA) is an important site of insulin processing and granule maturation, but whether GA organelle dysfunction and GA stress are present in the diabetic β-cell has not been tested. We utilized an informatics-based approach to develop a transcriptional signature of β-cell GA stress using existing RNA sequencing and microarray datasets generated using human islets from donors with diabetes and islets where type 1(T1D) and type 2 diabetes (T2D) had been modeled ex vivo. To narrow our results to GA-specific genes, we applied a filter set of 1,030 genes accepted as GA associated.
    [Show full text]
  • 1 UST College of Science Department of Biological Sciences
    UST College of Science Department of Biological Sciences 1 Pharmacogenomics of Myofascial Pain Syndrome An Undergraduate Thesis Submitted to the Department of Biological Sciences College of Science University of Santo Tomas In Partial Fulfillment of the Requirements for the Degree of Bachelor of Science in Biology Jose Marie V. Lazaga Marc Llandro C. Fernandez May 2021 UST College of Science Department of Biological Sciences 2 PANEL APPROVAL SHEET This undergraduate research manuscript entitled: Pharmacogenomics of Myofascial Pain Syndrome prepared and submitted by Jose Marie V. Lazaga and Marc Llandro C. Fernandez, was checked and has complied with the revisions and suggestions requested by panel members after thorough evaluation. This final version of the manuscript is hereby approved and accepted for submission in partial fulfillment of the requirements for the degree of Bachelor of Science in Biology. Noted by: Asst. Prof. Marilyn G. Rimando, PhD Research adviser, Bio/MicroSem 602-603 Approved by: Bio/MicroSem 603 panel member Bio/MicroSem 603 panel member Date: Date: UST College of Science Department of Biological Sciences 3 DECLARATION OF ORIGINALITY We hereby affirm that this submission is our own work and that, to the best of our knowledge and belief, it contains no material previously published or written by another person nor material to which a substantial extent has been accepted for award of any other degree or diploma of a university or other institute of higher learning, except where due acknowledgement is made in the text. We also declare that the intellectual content of this undergraduate research is the product of our work, even though we may have received assistance from others on style, presentation, and language expression.
    [Show full text]
  • Cochlin in the Eye: Functional Implications
    Cochlin in the eye: Functional implications Picciani, R., Desaia, K., Guduric-Fuchs, J., Cogliati, T., Morton, C. C., & Bhattacharya, S. K. (2007). Cochlin in the eye: Functional implications. Progress in Retinal and Eye Research, 26 (5)(5), 453-469. https://doi.org/10.1016/j.preteyeres.2007.06.002 Published in: Progress in Retinal and Eye Research Queen's University Belfast - Research Portal: Link to publication record in Queen's University Belfast Research Portal General rights Copyright for the publications made accessible via the Queen's University Belfast Research Portal is retained by the author(s) and / or other copyright owners and it is a condition of accessing these publications that users recognise and abide by the legal requirements associated with these rights. Take down policy The Research Portal is Queen's institutional repository that provides access to Queen's research output. Every effort has been made to ensure that content in the Research Portal does not infringe any person's rights, or applicable UK laws. If you discover content in the Research Portal that you believe breaches copyright or violates any law, please contact [email protected]. Download date:26. Sep. 2021 Author’s Accepted Manuscript Cochlin in the eye: Functional implications Renata Picciani, Kavita Desai, Jasenka Guduric- Fuchs,Tiziana Cogliati, Cynthia C. Morton, Sanjoy K. Bhattacharya PII: S1350-9462(07)00040-7 DOI: doi:10.1016/j.preteyeres.2007.06.002 Reference: JPRR 345 www.elsevier.com/locate/prer To appear in: Progress in Retinal and Eye Research Cite this article as: Renata Picciani, Kavita Desai, Jasenka Guduric-Fuchs, Tiziana Cogliati, Cynthia C.
    [Show full text]
  • Mouse Mid2 Conditional Knockout Project (CRISPR/Cas9)
    https://www.alphaknockout.com Mouse Mid2 Conditional Knockout Project (CRISPR/Cas9) Objective: To create a Mid2 conditional knockout Mouse model (C57BL/6J) by CRISPR/Cas-mediated genome engineering. Strategy summary: The Mid2 gene (NCBI Reference Sequence: NM_011845 ; Ensembl: ENSMUSG00000000266 ) is located on Mouse chromosome X. 9 exons are identified, with the ATG start codon in exon 1 and the TAA stop codon in exon 9 (Transcript: ENSMUST00000112993). Exon 5 will be selected as conditional knockout region (cKO region). Deletion of this region should result in the loss of function of the Mouse Mid2 gene. To engineer the targeting vector, homologous arms and cKO region will be generated by PCR using BAC clone RP23-340F6 as template. Cas9, gRNA and targeting vector will be co-injected into fertilized eggs for cKO Mouse production. The pups will be genotyped by PCR followed by sequencing analysis. Note: Exon 5 starts from about 49.34% of the coding region. The knockout of Exon 5 will result in frameshift of the gene. The size of intron 4 for 5'-loxP site insertion: 11103 bp, and the size of intron 5 for 3'-loxP site insertion: 1122 bp. The size of effective cKO region: ~628 bp. The cKO region does not have any other known gene. Page 1 of 7 https://www.alphaknockout.com Overview of the Targeting Strategy Wildtype allele gRNA region 5' gRNA region 3' 1 5 6 9 Targeting vector Targeted allele Constitutive KO allele (After Cre recombination) Legends Exon of mouse Mid2 Homology arm cKO region loxP site Page 2 of 7 https://www.alphaknockout.com Overview of the Dot Plot Window size: 10 bp Forward Reverse Complement Sequence 12 Note: The sequence of homologous arms and cKO region is aligned with itself to determine if there are tandem repeats.
    [Show full text]
  • Effects and Mechanisms of Eps8 on the Biological Behaviour of Malignant Tumours (Review)
    824 ONCOLOGY REPORTS 45: 824-834, 2021 Effects and mechanisms of Eps8 on the biological behaviour of malignant tumours (Review) KAILI LUO1, LEI ZHANG2, YUAN LIAO1, HONGYU ZHOU1, HONGYING YANG2, MIN LUO1 and CHEN QING1 1School of Pharmaceutical Sciences and Yunnan Key Laboratory of Pharmacology for Natural Products, Kunming Medical University, Kunming, Yunnan 650500; 2Department of Gynecology, Yunnan Tumor Hospital and The Third Affiliated Hospital of Kunming Medical University; Kunming, Yunnan 650118, P.R. China Received August 29, 2020; Accepted December 9, 2020 DOI: 10.3892/or.2021.7927 Abstract. Epidermal growth factor receptor pathway substrate 8 1. Introduction (Eps8) was initially identified as the substrate for the kinase activity of EGFR, improving the responsiveness of EGF, which Malignant tumours are uncontrolled cell proliferation diseases is involved in cell mitosis, differentiation and other physiological caused by oncogenes and ultimately lead to organ and body functions. Numerous studies over the last decade have demon- dysfunction (1). In recent decades, great progress has been strated that Eps8 is overexpressed in most ubiquitous malignant made in the study of genes and signalling pathways in tumours and subsequently binds with its receptor to activate tumorigenesis. Eps8 was identified by Fazioli et al in NIH-3T3 multiple signalling pathways. Eps8 not only participates in the murine fibroblasts via an approach that allows direct cloning regulation of malignant phenotypes, such as tumour proliferation, of intracellular substrates for receptor tyrosine kinases (RTKs) invasion, metastasis and drug resistance, but is also related to that was designed to study the EGFR signalling pathway. Eps8 the clinicopathological characteristics and prognosis of patients.
    [Show full text]
  • The Gene Repressor Complex Nurd Interacts with the Histone Variant H3.3
    Downloaded from genome.cshlp.org on October 3, 2021 - Published by Cold Spring Harbor Laboratory Press The gene repressor complex NuRD interacts with the histone variant H3.3 at promoters of active genes Daniel C. Kraushaar1,3,4, Zuozhou Chen2,4, Qingsong Tang1, Kairong Cui1, Junfang Zhang2,*, Keji Zhao1,* 1 Systems Biology Center, National Heart, Lung, and Blood Institute, NIH, MD 2 Key Laboratory of Exploration and Utilization of Aquatic Genetic Resources (Shanghai Ocean University), Ministry of Education; International Research Center for Marine Biosciences at Shanghai Ocean University, Ministry of Science and Technology; National Demonstration Center for Experimental Fisheries Science Education, Shanghai Ocean University, Shanghai 201306, China. 3 Current address: Genomic and RNA Profiling Core, Baylor College of Medicine, TX 4These authors contributed equally to this work *Correspondence: Junfang Zhang ([email protected]); Keji Zhao ([email protected]) Key words: histone variant H3.3; chromatin modification; epigenetics; ChIP-seq; protein interaction; NuRD; histone modification 1 Downloaded from genome.cshlp.org on October 3, 2021 - Published by Cold Spring Harbor Laboratory Press Abstract The histone variant H3.3 is deposited across active genes, regulatory regions and telomeres. It remains unclear how H3.3 interacts with chromatin modifying enzymes and thereby modulates gene activity. In this study, we performed a co- immunoprecipitation-mass spectrometry analysis of proteins associated with H3.3-containing nucleosomes and identified the Nucleosome Remodeling and Deacetylase complex (NuRD) as a major H3.3-interactor. We show that the H3.3- NuRD interaction is dependent on the H3.3 lysine 4 residue and that NuRD binding occurs when lysine 4 is in its unmodified state.
    [Show full text]
  • A Comprehensive Network and Pathway Analysis of Human Deafness Genes
    Otology & Neurotology 34:961Y970 Ó 2013, Otology & Neurotology, Inc. A Comprehensive Network and Pathway Analysis of Human Deafness Genes *Georgios A. Stamatiou and †Konstantina M. Stankovic *Department of Otolaryngology, Hippokration General Hospital, University of Athens, Athens, Greece; and ÞDepartment of Otology and Laryngology, Harvard Medical School and Department of Otolaryngology, Massachusetts Eye and Ear Infirmary, Boston, Massachusetts, U.S.A. Objective: To perform comprehensive network and pathway factor beta1 (TGFB1) for Group 1, MAPK3/MAPK1 MAP kinase analyses of the genes known to cause genetic hearing loss. (ERK 1/2) and the G protein coupled receptors (GPCR) for Study Design: In silico analysis of deafness genes using inge- Group 2, and TGFB1 and hepatocyte nuclear factor 4 alpha (HNF4A) nuity pathway analysis (IPA). for Group 3. The nodal molecules included not only those known Methods: Genes relevant for hearing and deafness were iden- to be associated with deafness (GPCR), or with predisposition to tified through PubMed literature searches and the Hereditary otosclerosis (TGFB1), but also novel genes that have not been Hearing Loss Homepage. The genes were assembled into 3 groups: described in the cochlea (HNF4A) and signaling kinases (ERK 1/2). 63 genes that cause nonsyndromic deafness, 107 genes that cause Conclusion: A number of molecules that are likely to be key nonsyndromic or syndromic sensorineural deafness, and 112 genes mediators of genetic hearing loss were identified through three associated with otic capsule development and malformations. Each different network and pathway analyses. The molecules included group of genes was analyzed using IPA to discover the most new candidate genes for deafness.
    [Show full text]
  • A Multi-Stage Genome-Wide Association Study of Uterine Fibroids in African Americans
    UCLA UCLA Previously Published Works Title A multi-stage genome-wide association study of uterine fibroids in African Americans. Permalink https://escholarship.org/uc/item/0mc5r0xh Journal Human genetics, 136(10) ISSN 0340-6717 Authors Hellwege, Jacklyn N Jeff, Janina M Wise, Lauren A et al. Publication Date 2017-10-01 DOI 10.1007/s00439-017-1836-1 Peer reviewed eScholarship.org Powered by the California Digital Library University of California Hum Genet (2017) 136:1363–1373 DOI 10.1007/s00439-017-1836-1 ORIGINAL INVESTIGATION A multi‑stage genome‑wide association study of uterine fbroids in African Americans Jacklyn N. Hellwege1,2,3 · Janina M. Jef4 · Lauren A. Wise5,6 · C. Scott Gallagher7 · Melissa Wellons8,9 · Katherine E. Hartmann3,9 · Sarah F. Jones1,3 · Eric S. Torstenson1,2 · Scott Dickinson10 · Edward A. Ruiz‑Narváez6 · Nadin Rohland7 · Alexander Allen7 · David Reich7,11,12 · Arti Tandon7 · Bogdan Pasaniuc13,14 · Nicholas Mancuso13 · Hae Kyung Im10 · David A. Hinds15 · Julie R. Palmer6 · Lynn Rosenberg6 · Joshua C. Denny16,17 · Dan M. Roden2,16,17,18 · Elizabeth A. Stewart19 · Cynthia C. Morton12,20,21,22 · Eimear E. Kenny4 · Todd L. Edwards1,2,3 · Digna R. Velez Edwards2,3,9 Received: 12 April 2017 / Accepted: 16 August 2017 / Published online: 23 August 2017 © Springer-Verlag GmbH Germany 2017 Abstract Uterine fbroids are benign tumors of the uterus imaging, genotyped and imputed to 1000 Genomes. Stage 2 afecting up to 77% of women by menopause. They are the used self-reported fbroid and GWAS data from 23andMe, leading indication for hysterectomy, and account for $34 bil- Inc.
    [Show full text]
  • Supplementary Materials
    Supplementary materials Supplementary Table S1: MGNC compound library Ingredien Molecule Caco- Mol ID MW AlogP OB (%) BBB DL FASA- HL t Name Name 2 shengdi MOL012254 campesterol 400.8 7.63 37.58 1.34 0.98 0.7 0.21 20.2 shengdi MOL000519 coniferin 314.4 3.16 31.11 0.42 -0.2 0.3 0.27 74.6 beta- shengdi MOL000359 414.8 8.08 36.91 1.32 0.99 0.8 0.23 20.2 sitosterol pachymic shengdi MOL000289 528.9 6.54 33.63 0.1 -0.6 0.8 0 9.27 acid Poricoic acid shengdi MOL000291 484.7 5.64 30.52 -0.08 -0.9 0.8 0 8.67 B Chrysanthem shengdi MOL004492 585 8.24 38.72 0.51 -1 0.6 0.3 17.5 axanthin 20- shengdi MOL011455 Hexadecano 418.6 1.91 32.7 -0.24 -0.4 0.7 0.29 104 ylingenol huanglian MOL001454 berberine 336.4 3.45 36.86 1.24 0.57 0.8 0.19 6.57 huanglian MOL013352 Obacunone 454.6 2.68 43.29 0.01 -0.4 0.8 0.31 -13 huanglian MOL002894 berberrubine 322.4 3.2 35.74 1.07 0.17 0.7 0.24 6.46 huanglian MOL002897 epiberberine 336.4 3.45 43.09 1.17 0.4 0.8 0.19 6.1 huanglian MOL002903 (R)-Canadine 339.4 3.4 55.37 1.04 0.57 0.8 0.2 6.41 huanglian MOL002904 Berlambine 351.4 2.49 36.68 0.97 0.17 0.8 0.28 7.33 Corchorosid huanglian MOL002907 404.6 1.34 105 -0.91 -1.3 0.8 0.29 6.68 e A_qt Magnogrand huanglian MOL000622 266.4 1.18 63.71 0.02 -0.2 0.2 0.3 3.17 iolide huanglian MOL000762 Palmidin A 510.5 4.52 35.36 -0.38 -1.5 0.7 0.39 33.2 huanglian MOL000785 palmatine 352.4 3.65 64.6 1.33 0.37 0.7 0.13 2.25 huanglian MOL000098 quercetin 302.3 1.5 46.43 0.05 -0.8 0.3 0.38 14.4 huanglian MOL001458 coptisine 320.3 3.25 30.67 1.21 0.32 0.9 0.26 9.33 huanglian MOL002668 Worenine
    [Show full text]