Computational Analyses of Small Molecules Activity from Phenotypic Screens

Total Page:16

File Type:pdf, Size:1020Kb

Computational Analyses of Small Molecules Activity from Phenotypic Screens Computational analyses of small molecules activity from phenotypic screens Azedine Zoufir Hughes Hall This dissertation is submitted for the degree of Doctor of Philosophy July 2018 Declaration This thesis is submitted as the result of my own work and includes nothing which is the outcome of work done in collaboration except where specifically indicated in the text. It is not substantially the same as any that I have submitted, or, is being concurrently submitted for a degree or diploma or other qualification at the University of Cambridge or any other University or similar institution except as declared in the preface and specified in the text. I further state that no substantial part of my dissertation has already been submitted, or, is being concurrently submitted for any such degree, diploma or other qualification at the University of Cambridge or any other University or similar institution except as declared in the Preface and specified in the text. This dissertation does not exceed the word limit of 60,000 words. Azedine Zoufir July 2018 Summary Title: Computational analyses of small molecules activity from phenotypic screens Author: Azedine Zoufir Drug discovery is no longer relying on the one gene-one disease paradigm nor on target-based screening alone to discover new drugs. Phenotypic-based screening is regaining momentum to discover new compounds since those assays provide an environment closer to the physiological state of the disease and allow to better anticipate off-target effects and other factors that can limit the efficacy of the drugs. However, uncovering the mechanism of action of the compounds active in those assays relies on in vitro techniques that are expensive and time- consuming. In silico approaches are therefore beneficial to prioritise mechanism of action hypotheses to be tested in such systems. In this thesis, the use of machine learning algorithms for in silico ligand-target prediction for target deconvolution in phenotypic screening datasets was investigated. A computational workflow is presented in Chapter 2, that allows to improve the coverage of mechanism of action hypotheses obtained by combining two conceptually different target prediction algorithms. These models rely on the principle that two structurally similar compounds are likely to have the same target. In Chapter 3 of this thesis, it was shown that structural similarity and the similarity in phenotypic activity are correlated, and the fraction of phenotypically similar compounds that can be expected for an increase in structural similarity was subsequently quantified. Morgan fingerprints were also found to be less sensitive to the dataset employed in these analyses than two other commonly used molecular descriptors. In Chapter 4, the mechanism of action hypotheses obtained through target prediction was compared to those obtained by extracting experimental bioactivity data of compounds active in phenotypic assays. It was then showed that the mechanism of action hypotheses generated from these two types of approach agreed where a large number of compounds were active in the phenotypic assay. When there were fewer compounds active in the phenotypic assay, target prediction complemented the use of experimental bioactivity data and allowed to uncover alternative mechanisms of action for compounds active in these assays. Finally, the in silico target prediction workflow described in Chapter 2 was applied in Chapter 5 to deconvolute the activity of compounds in a kidney cyst growth reduction assay, aimed at discovering novel therapeutic opportunities for polycystic kidney disease. A metric was developed to rank predicted targets according to the activity of the compounds driving their prediction. Gene expression data and occurrences in the literature were combined with the target predictions to further narrow down the most probable mechanisms of action of cyst growth reducing compounds in the screen. Two target predictions were proposed as a potential mechanism for the reduction of kidney cyst growth, one of which agreed with docking studies. Acknowledgements I would like to thank my supervisor Dr Andreas Bender for allowing me to be a part of his lab. I thank him for his continuous guidance and patience through all the revisions of my work. I also thank my collaborators from the University of Leiden, Dr Tijmen Booij and Dr Leo Price for providing the kidney cyst screening data, and Dr Dorien Peters and Tareq Malas for providing their gene expression dataset. I am grateful to Dr Xitong Li and Dr Ellen Berg for providing the BioMAP dataset. I thank the European Research Council for funding my research. Next, I thank Dr Fredrik Svensson, Dr Krishna Bulusu, Dr Avid Afzal and Dr Deszo Modos, for providing excellent scientific advice and very constructive feedback on my work. I am grateful to the whole of the Bender group for being supportive colleagues and always friendly. Rich and Lewis are thanked for their technical help in using our computing server. Also, my time in this group would not have been the same without Sefer, Siti, Fatima, Nitin, Avid, Leen, Nadia, Fredrik, Deszo and Krishna, who all have been very supportive and helpful with me, and I am very grateful to all of them. I really enjoyed working among such a diverse and talented group of people. I also thank Susan Begg without whom the lab would not be running so smoothly. Last but not least, I thank my family and particularly my parents for their encouragements throughout my studies. My deepest gratitude goes to my friends Ain, Charles, Ben and Cristian for being there in those times when friends are needed, and for keeping me away of my thesis when I would become too preoccupied about it. Table of Contents TABLE OF CONTENTS ........................................................................................................ I LIST OF FIGURES .............................................................................................................. VI LIST OF TABLES ............................................................................................................. VIII ABBREVIATIONS ................................................................................................................. X CHAPTER 1 INTRODUCTION ........................................................................................... 1 1.1 FROM TARGET-BASED TO PHENOTYPIC-BASED DRUG DISCOVERY ........ 2 1.1.1 TARGET-BASED SCREENING AND LIMITATIONS ...................................................................... 2 1.1.2 PHENOTYPIC-BASED SCREENING COMPENSATES FOR THE LIMITATIONS OF TARGET- BASED SCREENING ........................................................................................................................................ 3 1.1.3 ASSAYS USED IN PHENOTYPIC-BASED SCREENING ................................................................. 4 1.1.4 IN VITRO DECONVOLUTION IN PHENOTYPIC SCREENS AND LIMITATIONS .......................... 5 1.2 MOLECULAR AND BIOLOGICAL SIMILARITY .................................................. 7 1.2.1 REPRESENTATION OF CHEMICALS .............................................................................................. 7 1.2.2 MOLECULAR SIMILARITY PRINCIPLE IN VIRTUAL SCREENING AND NEIGHBOURHOOD PROPERTY ..................................................................................................................................................... 10 1.3 IN SILICO DECONVOLUTION METHODS OF COMPOUND ACTIVITY IN PHENOTYPIC SCREENS ................................................................................................... 14 1.3.1 DATA-DRIVEN DECONVOLUTION .............................................................................................. 14 1.3.2 DECONVOLUTION METHODS BASED ON IN SILICO LIGAND-TARGET PREDICTIONS ........ 16 1.3.2.1 Bioactivity datasets and limitations relevant to target prediction ................... 16 i 1.3.2.2 Current target prediction methods ................................................................... 19 1.3.2.3 Applications to deconvolution of compounds active in phenotypic screens .... 26 1.4 CONCLUSIONS AND AIMS OF THE THESIS ........................................................ 28 CHAPTER 2 COMPUTATIONAL METHODS ................................................................ 30 2.1 WORKFLOW OVERVIEW ........................................................................................ 30 2.2 MOLECULAR FINGERPRINTS ................................................................................ 32 2.2.1 ECFP4 FINGERPRINTS ................................................................................................................. 32 2.2.2 MACCS KEYS AND PUBCHEM FINGERPRINTS ...................................................................... 35 2.3 SIMILARITY SCORING ............................................................................................. 35 2.3.1 STRUCTURAL SIMILARITY SCORING ......................................................................................... 35 2.3.2 BIOLOGICAL SIMILARITY SCORING .......................................................................................... 36 2.4 LIGAND-TARGET PREDICTION MODELS .......................................................... 37 2.4.1 CHEMBL TARGET PREDICTION MODEL .................................................................................. 38 2.4.1.1 Laplacian-corrected multinomial Naïve Bayes machine learning model ........ 38 2.4.1.2 Multinomial Naïve Bayes target prediction
Recommended publications
  • Large-Scale Analysis of Genome and Transcriptome Alterations in Multiple Tumors Unveils Novel Cancer-Relevant Splicing Networks
    Downloaded from genome.cshlp.org on September 28, 2021 - Published by Cold Spring Harbor Laboratory Press Research Large-scale analysis of genome and transcriptome alterations in multiple tumors unveils novel cancer-relevant splicing networks Endre Sebestyén,1,5 Babita Singh,1,5 Belén Miñana,1,2 Amadís Pagès,1 Francesca Mateo,3 Miguel Angel Pujana,3 Juan Valcárcel,1,2,4 and Eduardo Eyras1,4 1Universitat Pompeu Fabra, E08003 Barcelona, Spain; 2Centre for Genomic Regulation, E08003 Barcelona, Spain; 3Program Against Cancer Therapeutic Resistance (ProCURE), Catalan Institute of Oncology (ICO), Bellvitge Institute for Biomedical Research (IDIBELL), E08908 L’Hospitalet del Llobregat, Spain; 4Catalan Institution for Research and Advanced Studies, E08010 Barcelona, Spain Alternative splicing is regulated by multiple RNA-binding proteins and influences the expression of most eukaryotic genes. However, the role of this process in human disease, and particularly in cancer, is only starting to be unveiled. We system- atically analyzed mutation, copy number, and gene expression patterns of 1348 RNA-binding protein (RBP) genes in 11 solid tumor types, together with alternative splicing changes in these tumors and the enrichment of binding motifs in the alter- natively spliced sequences. Our comprehensive study reveals widespread alterations in the expression of RBP genes, as well as novel mutations and copy number variations in association with multiple alternative splicing changes in cancer drivers and oncogenic pathways. Remarkably, the altered splicing patterns in several tumor types recapitulate those of undifferen- tiated cells. These patterns are predicted to be mainly controlled by MBNL1 and involve multiple cancer drivers, including the mitotic gene NUMA1. We show that NUMA1 alternative splicing induces enhanced cell proliferation and centrosome am- plification in nontumorigenic mammary epithelial cells.
    [Show full text]
  • 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]
  • Remodeling Adipose Tissue Through in Silico Modulation of Fat Storage For
    Chénard et al. BMC Systems Biology (2017) 11:60 DOI 10.1186/s12918-017-0438-9 RESEARCHARTICLE Open Access Remodeling adipose tissue through in silico modulation of fat storage for the prevention of type 2 diabetes Thierry Chénard2, Frédéric Guénard3, Marie-Claude Vohl3,4, André Carpentier5, André Tchernof4,6 and Rafael J. Najmanovich1* Abstract Background: Type 2 diabetes is one of the leading non-infectious diseases worldwide and closely relates to excess adipose tissue accumulation as seen in obesity. Specifically, hypertrophic expansion of adipose tissues is related to increased cardiometabolic risk leading to type 2 diabetes. Studying mechanisms underlying adipocyte hypertrophy could lead to the identification of potential targets for the treatment of these conditions. Results: We present iTC1390adip, a highly curated metabolic network of the human adipocyte presenting various improvements over the previously published iAdipocytes1809. iTC1390adip contains 1390 genes, 4519 reactions and 3664 metabolites. We validated the network obtaining 92.6% accuracy by comparing experimental gene essentiality in various cell lines to our predictions of biomass production. Using flux balance analysis under various test conditions, we predict the effect of gene deletion on both lipid droplet and biomass production, resulting in the identification of 27 genes that could reduce adipocyte hypertrophy. We also used expression data from visceral and subcutaneous adipose tissues to compare the effect of single gene deletions between adipocytes from each
    [Show full text]
  • RBM11 Sirna (H): Sc-91387
    SANTA CRUZ BIOTECHNOLOGY, INC. RBM11 siRNA (h): sc-91387 BACKGROUND PRODUCT Proteins containing RNA recognition motifs, including various hnRNP proteins, RBM11 siRNA (h) is a pool of 3 target-specific 19-25 nt siRNAs designed are implicated in the regulation of alternative splicing and protein components to knock down gene expression. Each vial contains 3.3 nmol of lyophilized of snRNPs. The RBM (RNA-binding motif) gene family encodes proteins with siRNA, sufficient for a 10 µM solution once resuspended using protocol an RNA binding motif that have been suggested to play a role in the modu- below. Suitable for 50-100 transfections. Also see RBM11 shRNA Plasmid (h): lation of apoptosis. RBM11 (RNA-binding protein 11) is a 281 amino acid sc-91387-SH and RBM11 shRNA (h) Lentiviral Particles: sc-91387-V as alter- nuclear protein that contains one RNA recognition motif. RBM11 exists as nate gene silencing products. two isoforms produced by alternative splicing and is expressed in testis, kid- For independent verification of RBM11 (h) gene silencing results, we also ney, spleen, brain, spinal cord and mammary gland. The gene that encodes provide the individual siRNA duplex components. Each is available as 3.3 nmol RBM11 maps to chromosome 21, the smallest of the human chromosomes. of lyophilized siRNA. These include: sc-91387A, sc-91387B and sc-91387C. Down syndrome, also known as trisomy 21, is the disease most commonly associated with chromosome 21. Alzheimer’s disease, Jervell and Lange- STORAGE AND RESUSPENSION Nielsen syndrome and amyotrophic lateral sclerosis are also associated with chromosome 21. Store lyophilized siRNA duplex at -20° C with desiccant.
    [Show full text]
  • Recombinant Rabbit Anti-Human RAB8A Monoclonal Antibody, Clone NKG-S33-80-4 (CABT- Z616R) This Product Is for Research Use Only and Is Not Intended for Diagnostic Use
    Recombinant Rabbit Anti-Human RAB8A Monoclonal Antibody, clone NKG-S33-80-4 (CABT- Z616R) This product is for research use only and is not intended for diagnostic use. PRODUCT INFORMATION Immunogen Recombinant full length protein within Human RAB8A aa 1 to the C-terminus. Isotype IgG Source/Host Rabbit Species Reactivity Human Clone NKG-S33-80-4 Purification Protein A purified Conjugate Unconjugated Applications WB, IHC-P, ICC/IF, FC, IP Recommended dilution: WB: 1:1000 IHC-P: 1:1000 ICC/IF: 1:500 FC: 1:600 IP: 1:30 Positive Control WB: HEK-293T, HCT116 and HeLa whole cell lysates. IHC-P: Human bladder cancer and breast tissues. ICC/IF: A549 and HeLa cells. Flow Cyt: A549 cells. IP: A549 whole cell lysate. Format Liquid Concentration Lot specific Size 100 μl Buffer PBS, 40% Glycerol (glycerin, glycerine), 0.05% BSA, pH 7.2. Preservative 0.01% Sodium azide Storage Store at +4℃ short term (1-2 weeks). Upon delivery aliquot. Store at -20℃. Stable for 12 months at -20℃. 45-1 Ramsey Road, Shirley, NY 11967, USA Email: [email protected] Tel: 1-631-624-4882 Fax: 1-631-938-8221 1 © Creative Diagnostics All Rights Reserved Ship Wet ice Warnings This product is for research use only and is not intended for diagnostic use. BACKGROUND Introduction RAB8A may be involved in vesicular trafficking and neurotransmitter release. Together with RAB11A, RAB3IP, the exocyst complex, PARD3, PRKCI, ANXA2, CDC42 and DNMBP promotes transcytosis of PODXL to the apical membrane initiation sites (AMIS), apical surface formation and lumenogenesis. Together with MYO5B and RAB11A participates in epithelial cell polarization.
    [Show full text]
  • Mutation-Specific and Common Phosphotyrosine Signatures of KRAS G12D and G13D Alleles Anticipated Graduation August 1St, 2018
    MUTATION-SPECIFIC AND COMMON PHOSPHOTYROSINE SIGNATURES OF KRAS G12D AND G13D ALLELES by Raiha Tahir A dissertation submitted to The Johns Hopkins University in conformity with the requirement of the degree of Doctor of Philosophy Baltimore, MD August 2018 © 2018 Raiha Tahir All Rights Reserved ABSTRACT KRAS is one of the most frequently mutated genes across all cancer subtypes. Two of the most frequent oncogenic KRAS mutations observed in patients result in glycine to aspartic acid substitution at either codon 12 (G12D) or 13 (G13D). Although the biochemical differences between these two predominant mutations are not fully understood, distinct clinical features of the resulting tumors suggest involvement of disparate signaling mechanisms. When we compared the global phosphotyrosine proteomic profiles of isogenic colorectal cancer cell lines bearing either G12D or G13D KRAS mutations, we observed both shared as well as unique signaling events induced by the two KRAS mutations. Remarkably, while the G12D mutation led to an increase in membrane proximal and adherens junction signaling, the G13D mutation led to activation of signaling molecules such as non-receptor tyrosine kinases, MAPK kinases and regulators of metabolic processes. The importance of one of the cell surface molecules, MPZL1, which found to be hyperphosphorylated in G12D cells, was confirmed by cellular assays as its knockdown led to a decrease in proliferation of G12D but not G13D expressing cells. Overall, our study reveals important signaling differences across two common KRAS mutations and highlights the utility of our approach to systematically dissect the subtle differences between related oncogenic mutants and potentially lead to individualized treatments.
    [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]
  • Coordinate Regulation of Long Non-Coding Rnas and Protein-Coding Genes in Germ- Free Mice Joseph Dempsey, Angela Zhang and Julia Yue Cui*
    Dempsey et al. BMC Genomics (2018) 19:834 https://doi.org/10.1186/s12864-018-5235-3 RESEARCHARTICLE Open Access Coordinate regulation of long non-coding RNAs and protein-coding genes in germ- free mice Joseph Dempsey, Angela Zhang and Julia Yue Cui* Abstract Background: Long non-coding RNAs (lncRNAs) are increasingly recognized as regulators of tissue-specific cellular functions and have been shown to regulate transcriptional and translational processes, acting as signals, decoys, guides, and scaffolds. It has been suggested that some lncRNAs act in cis to regulate the expression of neighboring protein-coding genes (PCGs) in a mechanism that fine-tunes gene expression. Gut microbiome is increasingly recognized as a regulator of development, inflammation, host metabolic processes, and xenobiotic metabolism. However, there is little known regarding whether the gut microbiome modulates lncRNA gene expression in various host metabolic organs. The goals of this study were to 1) characterize the tissue-specific expression of lncRNAs and 2) identify and annotate lncRNAs differentially regulated in the absence of gut microbiome. Results: Total RNA was isolated from various tissues (liver, duodenum, jejunum, ileum, colon, brown adipose tissue, white adipose tissue, and skeletal muscle) from adult male conventional and germ-free mice (n = 3 per group). RNA-Seq was conducted and reads were mapped to the mouse reference genome (mm10) using HISAT. Transcript abundance and differential expression was determined with Cufflinks using the reference databases NONCODE 2016 for lncRNAs and UCSC mm10 for PCGs. Although the constitutive expression of lncRNAs was ubiquitous within the enterohepatic (liver and intestine) and the peripheral metabolic tissues (fat and muscle) in conventional mice, differential expression of lncRNAs by lack of gut microbiota was highly tissue specific.
    [Show full text]
  • 1 Supporting Information for a Microrna Network Regulates
    Supporting Information for A microRNA Network Regulates Expression and Biosynthesis of CFTR and CFTR-ΔF508 Shyam Ramachandrana,b, Philip H. Karpc, Peng Jiangc, Lynda S. Ostedgaardc, Amy E. Walza, John T. Fishere, Shaf Keshavjeeh, Kim A. Lennoxi, Ashley M. Jacobii, Scott D. Rosei, Mark A. Behlkei, Michael J. Welshb,c,d,g, Yi Xingb,c,f, Paul B. McCray Jr.a,b,c Author Affiliations: Department of Pediatricsa, Interdisciplinary Program in Geneticsb, Departments of Internal Medicinec, Molecular Physiology and Biophysicsd, Anatomy and Cell Biologye, Biomedical Engineeringf, Howard Hughes Medical Instituteg, Carver College of Medicine, University of Iowa, Iowa City, IA-52242 Division of Thoracic Surgeryh, Toronto General Hospital, University Health Network, University of Toronto, Toronto, Canada-M5G 2C4 Integrated DNA Technologiesi, Coralville, IA-52241 To whom correspondence should be addressed: Email: [email protected] (M.J.W.); yi- [email protected] (Y.X.); Email: [email protected] (P.B.M.) This PDF file includes: Materials and Methods References Fig. S1. miR-138 regulates SIN3A in a dose-dependent and site-specific manner. Fig. S2. miR-138 regulates endogenous SIN3A protein expression. Fig. S3. miR-138 regulates endogenous CFTR protein expression in Calu-3 cells. Fig. S4. miR-138 regulates endogenous CFTR protein expression in primary human airway epithelia. Fig. S5. miR-138 regulates CFTR expression in HeLa cells. Fig. S6. miR-138 regulates CFTR expression in HEK293T cells. Fig. S7. HeLa cells exhibit CFTR channel activity. Fig. S8. miR-138 improves CFTR processing. Fig. S9. miR-138 improves CFTR-ΔF508 processing. Fig. S10. SIN3A inhibition yields partial rescue of Cl- transport in CF epithelia.
    [Show full text]
  • Anti-Inflammatory Role of Curcumin in LPS Treated A549 Cells at Global Proteome Level and on Mycobacterial Infection
    Anti-inflammatory Role of Curcumin in LPS Treated A549 cells at Global Proteome level and on Mycobacterial infection. Suchita Singh1,+, Rakesh Arya2,3,+, Rhishikesh R Bargaje1, Mrinal Kumar Das2,4, Subia Akram2, Hossain Md. Faruquee2,5, Rajendra Kumar Behera3, Ranjan Kumar Nanda2,*, Anurag Agrawal1 1Center of Excellence for Translational Research in Asthma and Lung Disease, CSIR- Institute of Genomics and Integrative Biology, New Delhi, 110025, India. 2Translational Health Group, International Centre for Genetic Engineering and Biotechnology, New Delhi, 110067, India. 3School of Life Sciences, Sambalpur University, Jyoti Vihar, Sambalpur, Orissa, 768019, India. 4Department of Respiratory Sciences, #211, Maurice Shock Building, University of Leicester, LE1 9HN 5Department of Biotechnology and Genetic Engineering, Islamic University, Kushtia- 7003, Bangladesh. +Contributed equally for this work. S-1 70 G1 S 60 G2/M 50 40 30 % of cells 20 10 0 CURI LPSI LPSCUR Figure S1: Effect of curcumin and/or LPS treatment on A549 cell viability A549 cells were treated with curcumin (10 µM) and/or LPS or 1 µg/ml for the indicated times and after fixation were stained with propidium iodide and Annexin V-FITC. The DNA contents were determined by flow cytometry to calculate percentage of cells present in each phase of the cell cycle (G1, S and G2/M) using Flowing analysis software. S-2 Figure S2: Total proteins identified in all the three experiments and their distribution betwee curcumin and/or LPS treated conditions. The proteins showing differential expressions (log2 fold change≥2) in these experiments were presented in the venn diagram and certain number of proteins are common in all three experiments.
    [Show full text]
  • Gene Symbol Category ACAN ECM ADAM10 ECM Remodeling-Related ADAM11 ECM Remodeling-Related ADAM12 ECM Remodeling-Related ADAM15 E
    Supplementary Material (ESI) for Integrative Biology This journal is (c) The Royal Society of Chemistry 2010 Gene symbol Category ACAN ECM ADAM10 ECM remodeling-related ADAM11 ECM remodeling-related ADAM12 ECM remodeling-related ADAM15 ECM remodeling-related ADAM17 ECM remodeling-related ADAM18 ECM remodeling-related ADAM19 ECM remodeling-related ADAM2 ECM remodeling-related ADAM20 ECM remodeling-related ADAM21 ECM remodeling-related ADAM22 ECM remodeling-related ADAM23 ECM remodeling-related ADAM28 ECM remodeling-related ADAM29 ECM remodeling-related ADAM3 ECM remodeling-related ADAM30 ECM remodeling-related ADAM5 ECM remodeling-related ADAM7 ECM remodeling-related ADAM8 ECM remodeling-related ADAM9 ECM remodeling-related ADAMTS1 ECM remodeling-related ADAMTS10 ECM remodeling-related ADAMTS12 ECM remodeling-related ADAMTS13 ECM remodeling-related ADAMTS14 ECM remodeling-related ADAMTS15 ECM remodeling-related ADAMTS16 ECM remodeling-related ADAMTS17 ECM remodeling-related ADAMTS18 ECM remodeling-related ADAMTS19 ECM remodeling-related ADAMTS2 ECM remodeling-related ADAMTS20 ECM remodeling-related ADAMTS3 ECM remodeling-related ADAMTS4 ECM remodeling-related ADAMTS5 ECM remodeling-related ADAMTS6 ECM remodeling-related ADAMTS7 ECM remodeling-related ADAMTS8 ECM remodeling-related ADAMTS9 ECM remodeling-related ADAMTSL1 ECM remodeling-related ADAMTSL2 ECM remodeling-related ADAMTSL3 ECM remodeling-related ADAMTSL4 ECM remodeling-related ADAMTSL5 ECM remodeling-related AGRIN ECM ALCAM Cell-cell adhesion ANGPT1 Soluble factors and receptors
    [Show full text]
  • Noelia Díaz Blanco
    Effects of environmental factors on the gonadal transcriptome of European sea bass (Dicentrarchus labrax), juvenile growth and sex ratios Noelia Díaz Blanco Ph.D. thesis 2014 Submitted in partial fulfillment of the requirements for the Ph.D. degree from the Universitat Pompeu Fabra (UPF). This work has been carried out at the Group of Biology of Reproduction (GBR), at the Department of Renewable Marine Resources of the Institute of Marine Sciences (ICM-CSIC). Thesis supervisor: Dr. Francesc Piferrer Professor d’Investigació Institut de Ciències del Mar (ICM-CSIC) i ii A mis padres A Xavi iii iv Acknowledgements This thesis has been made possible by the support of many people who in one way or another, many times unknowingly, gave me the strength to overcome this "long and winding road". First of all, I would like to thank my supervisor, Dr. Francesc Piferrer, for his patience, guidance and wise advice throughout all this Ph.D. experience. But above all, for the trust he placed on me almost seven years ago when he offered me the opportunity to be part of his team. Thanks also for teaching me how to question always everything, for sharing with me your enthusiasm for science and for giving me the opportunity of learning from you by participating in many projects, collaborations and scientific meetings. I am also thankful to my colleagues (former and present Group of Biology of Reproduction members) for your support and encouragement throughout this journey. To the “exGBRs”, thanks for helping me with my first steps into this world. Working as an undergrad with you Dr.
    [Show full text]