Bioinformatic Identification of Disease Driver Networks Using Functional Profiling Data

Total Page:16

File Type:pdf, Size:1020Kb

Bioinformatic Identification of Disease Driver Networks Using Functional Profiling Data Institute for Molecular Medicine Finland, FIMM University of Helsinki, Helsinki, Finland Doctoral Programme in Biomedicine (DPBM) BIOINFORMATIC IDENTIFICATION OF DISEASE DRIVER NETWORKS USING FUNCTIONAL PROFILING DATA Agnieszka Szwajda ACADEMIC DISSERTATION To be presented, with the permission of the Faculty of Medicine, University of Helsinki, for public examination in lecture hall 3, Biomedicum Helsinki, Haartmaninkatu 8, on Friday 2nd of February 2018, at 12 noon. Helsinki, 2018 Supervised by: Professor Tero Aittokallio, Ph.D. Institute for Molecular Medicine Finland, FIMM University of Helsinki Helsinki, Finland Krister Wennerberg, Ph.D. Institute for Molecular Medicine Finland, FIMM University of Helsinki Helsinki, Finland Reviewed by: Associate Professor Harri Lähdesmäki, D.Sc. Department of Computer Science Aalto University School of Science Espoo, Finland Associate Professor Henri Xhaard, Ph.D. Division of Pharmaceutical Chemistry and Technology University of Helsinki Helsinki, Finland Opponent: Professor Matti Nykter, Ph.D. Institute of Biosciences and Medical Technology University of Tampere Tampere, Finland Custos: Professor Samuli Ripatti, Ph.D. Institute for Molecular Medicine Finland, FIMM University of Helsinki Helsinki, Finland © Agnieszka Szwajda Cover layout by Anita Tienhaara ISBN 978-951-51-3959-7 (paperback) ISBN 978-951-51-3960-3 (PDF) ISSN 2342-3161 (print) ISSN 2342-317X (online) Unigrafia Oy, Helsinki 2018 2 Table of contents ABBREVIATIONS ....................................................................................................................................... 5 LIST OF ORIGINAL PUBLICATIONS ............................................................................................................ 6 AUTHOR’S CONTRIBUTIONS .................................................................................................................... 7 ABSTRACT ................................................................................................................................................ 8 1. INTRODUCTION ................................................................................................................................. 10 2. REVIEW OF LITERATURE .................................................................................................................... 12 2.1. Methods for the identification of disease drivers (II, III) ............................................................ 12 2.2. Oncogene addiction and other molecular vulnerabilities in cancer (II) ..................................... 13 2.3. Functional proteomics using mass spectrometry (III, IV) ........................................................... 14 2.3.1. Quantitative MS ................................................................................................................... 15 2.3.2. Affinity Purification (AP) coupled with MS .......................................................................... 15 2.3.3. Limitation of MS-approaches .............................................................................................. 15 2.4. Protein-protein interaction networks (III, IV) ............................................................................. 16 2.5. Protein phosphorylation and nitrosylation (II, IV) ...................................................................... 17 2.6. Chemical and genetic perturbation screening (II) ...................................................................... 18 2.7. Quantitative scoring of single and combinatorial drug responses (II) ........................................ 20 2.8. Compound-target selectivity profiling (I, II) ............................................................................... 21 2.9. Background of diseases used in the studies (II, IV) .................................................................... 23 2.9.1. Breast cancer ....................................................................................................................... 23 2.9.2. Alzheimer’s disease ............................................................................................................. 24 3. AIMS OF THE STUDY .......................................................................................................................... 25 4. MATERIALS AND METHODS ............................................................................................................... 26 4.1. Comparison of kinase inhibitor profiling datasets (I) ................................................................. 26 4.2. Evaluation of reproducibility and reliability of kinase inhibitor profiling datasets (I) ................ 26 4.3. Integration of the target bioactivity measurements (I) .............................................................. 27 4.4. Kinase inhibition sensitivity score (KISS) for prediction of molecular cancer addictions (II) ..... 27 4.5. Combinatorial KISS for prediction of co-essential target pairs and synergistic drug combinations (II) ...................................................................................................................................................... 27 4.6. Compound sensitivity testing (II) ................................................................................................ 28 4.7. Compound-target mappings (II) ................................................................................................. 28 4.8. Comparison of driver de-convolution methods (II) .................................................................... 29 4.9. Independent compound and siRNA testing to validate KISS predictions (II).............................. 30 4.10. Construction of addiction networks (II) .................................................................................... 30 4.11. Generation of PME-1 interactome using MS/AP (III) ................................................................ 31 4.12. Relevance rank platform (RRP) for predicting functional similarity (III) ................................... 31 3 4.13. Identification of nitrosylated proteins (IV) ............................................................................... 33 4.14. Functional analysis of the proteomic data (IV) ......................................................................... 33 5. RESULTS ............................................................................................................................................. 34 5.1. Comparative evaluation and integration of target selectivity profiles (I) ................................... 34 5.1.1. Comparative analysis of the compound bioactivity studies ................................................ 34 5.1.2. Evaluation of the reliability of the compound bioactivity studies ....................................... 34 5.1.3. Integration of kinase inhibitor bioactivity data ................................................................... 35 5.2. KISS for prediction of single and combinatorial cancer drivers (II)............................................. 35 5.2.1. Comparison of KISS with other driver deconvolution approaches...................................... 36 5.2.2. Comparison of KISS and transcriptomic profiles ................................................................. 37 5.2.3. KISS-predicted kinase addictions showed consistency between independent drug collections ...................................................................................................................................... 37 5.2.4. KISS-based prediction of pharmacologically actionable drivers in breast cancer cell lines 37 5.2.5. Predicted signal addictions form cell line specific driver networks .................................... 38 5.2.6. Combinatorial KISS prediction of co-essential target pairs and synergistic drug combinations ................................................................................................................................. 39 5.3. RRP analysis of PIN1 and PME-1 interactome (III) ...................................................................... 39 5.3.1. Functional similarity ranking of PIN1 interactome .............................................................. 39 5.3.2. Functional similarity ranking of AP-MS-derived PME-1 interactome .................................. 41 5.4. Functional analysis of S-nitrosylated proteins (IV) ..................................................................... 42 6. DISCUSSION AND CONCLUSIONS....................................................................................................... 44 7. ACKNOWLEDGEMENTS...................................................................................................................... 49 8. REFERENCES....................................................................................................................................... 51 4 ABBREVIATIONS AD – Alzheimer’s disease AP – affinity purification AP-MS – affinity purification coupled with mass spectrometry APP-amyloid precursor protein CML- chronic myeloid leukemia DSS – drug sensitivity score EC50 - half-maximal effective concentration ER – estrogen receptor GSEA – gene set enrichment analysis HSA - highest singe agent IC50 – half-maximal inhibitory concentration Kd – dissociation constant Ki – inhibition constant KIBA - kinase inhibitor bioactivity KISS - kinase inhibition sensitivity score KS – Kolmogorov-Smirnoff MS-mass spectrometry NGS – next generation sequencing NO – nitrogen oxide PPI- protein-protein interaction
Recommended publications
  • Depleting PTOV1 Sensitizes Non-Small Cell Lung Cancer Cells to Chemotherapy Through Attenuating Cancer Stem Cell Traits
    Wu et al. Journal of Experimental & Clinical Cancer Research (2019) 38:341 https://doi.org/10.1186/s13046-019-1349-y RESEARCH Open Access Depleting PTOV1 sensitizes non-small cell lung cancer cells to chemotherapy through attenuating cancer stem cell traits Zhiqiang Wu1*† , Zhuang Liu1†, Xiangli Jiang2†, Zeyun Mi3, Maobin Meng1, Hui Wang1, Jinlin Zhao1, Boyu Zheng1 and Zhiyong Yuan1* Abstract Background: Prostate tumor over expressed gene 1 (PTOV1) has been reported as an oncogene in several human cancers. However, the clinical significance and biological role of PTOV1 remain elusive in non-small cell lung cancer (NSCLC). Methods: The Cancer Genome Atlas (TCGA) data and NCBI/GEO data mining, western blotting analysis and immunohistochemistry were employed to characterize the expression of PTOV1 in NSCLC cell lines and tissues. The clinical significance of PTOV1 in NSCLC was studied by immunohistochemistry statistical analysis and Kaplan–Meier Plotter database mining. A series of in-vivo and in-vitro assays, including colony formation, CCK-8 assays, flow cytometry, wound healing, trans-well assay, tumor sphere formation, quantitative PCR, gene set enrichment analysis (GSEA), immunostaining and xenografts tumor model, were performed to demonstrate the effects of PTOV1 on chemosensitivity of NSCLC cells and the underlying mechanisms. Results: PTOV1 is overexpressed in NSCLC cell lines and tissues. High PTOV1 level indicates a short survival time and poor response to chemotherapy of NSCLC patients. Depleting PTOV1 increased sensitivity to chemotherapy drugs cisplatin and docetaxel by increasing cell apoptosis, inhibiting cell migration and invasion. Our study verified that depleting PTOV1 attenuated cancer stem cell traits through impairing DKK1/β-catenin signaling to enhance chemosensitivity of NSCLC cells.
    [Show full text]
  • Viral Integration Transforms Chromatin to Drive Oncogenesis
    bioRxiv preprint doi: https://doi.org/10.1101/2020.02.12.942755; this version posted February 13, 2020. 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. Viral integration transforms chromatin to drive oncogenesis Mehran Karimzadeh (ORCID: 0000-0002-7324-6074)1,2,3, Christopher Arlidge (ORCID: 0000-0001-9454-8541)2, Ariana Rostami (ORCID: 0000-0002-3423-8303)1,2, Mathieu Lupien (ORCID: 0000-0003-0929-9478)1,2,5, Scott V. Bratman (ORCID: 0000-0001-8610-4908)1,2,5, and Michael M. Hoffman (ORCID: 0000-0002-4517-1562)1,2,3,4,5 1Department of Medical Biophysics, University of Toronto, Toronto, ON, Canada 2Princess Margaret Cancer Centre, Toronto, ON, Canada 3Vector Institute, Toronto, ON, Canada 4Department of Computer Science, University of Toronto, Toronto, ON, Canada 5Lead contact: michael.hoff[email protected], [email protected], [email protected] February 12, 2020 Abstract Human papillomavirus (HPV) drives almost all cervical cancers and up to ∼70% of head and neck cancers. Frequent integration into the host genome occurs only for tumourigenic strains of HPV. We hypothesized that changes in the epigenome and transcriptome contribute to the tumourigenicity of HPV. We found that viral integration events often occurred along with changes in chromatin state and expression of genes near the integration site. We investigated whether introduction of new transcription factor binding sites due to HPV integration could invoke these changes. Some regions within the HPV genome, particularly the position of a conserved CTCF sequence motif, showed enriched chromatin accessibility signal.
    [Show full text]
  • Upregulation of 15 Antisense Long Non-Coding Rnas in Osteosarcoma
    G C A T T A C G G C A T genes Article Upregulation of 15 Antisense Long Non-Coding RNAs in Osteosarcoma Emel Rothzerg 1,2 , Xuan Dung Ho 3 , Jiake Xu 1 , David Wood 1, Aare Märtson 4 and Sulev Kõks 2,5,* 1 School of Biomedical Sciences, The University of Western Australia, Perth, WA 6009, Australia; [email protected] (E.R.); [email protected] (J.X.); [email protected] (D.W.) 2 Perron Institute for Neurological and Translational Science, QEII Medical Centre, Nedlands, WA 6009, Australia 3 Department of Oncology, College of Medicine and Pharmacy, Hue University, Hue 53000, Vietnam; [email protected] 4 Department of Traumatology and Orthopaedics, University of Tartu, Tartu University Hospital, 50411 Tartu, Estonia; [email protected] 5 Centre for Molecular Medicine and Innovative Therapeutics, Murdoch University, Murdoch, WA 6150, Australia * Correspondence: [email protected]; Tel.: +61-(0)-8-6457-0313 Abstract: The human genome encodes thousands of natural antisense long noncoding RNAs (lncR- NAs); they play the essential role in regulation of gene expression at multiple levels, including replication, transcription and translation. Dysregulation of antisense lncRNAs plays indispensable roles in numerous biological progress, such as tumour progression, metastasis and resistance to therapeutic agents. To date, there have been several studies analysing antisense lncRNAs expression profiles in cancer, but not enough to highlight the complexity of the disease. In this study, we investi- gated the expression patterns of antisense lncRNAs from osteosarcoma and healthy bone samples (24 tumour-16 bone samples) using RNA sequencing.
    [Show full text]
  • The Role of Prostate Tumor Overexpressed 1 in Cancer Progression
    www.impactjournals.com/oncotarget/ Oncotarget, 2017, Vol. 8, (No. 7), pp: 12451-12471 Review The role of prostate tumor overexpressed 1 in cancer progression Verónica Cánovas1,4, Matilde Lleonart2,4, Juan Morote1,3,4, Rosanna Paciucci1,4 1Biomedical Research Group of Urology, Vall d’Hebron Research Institute, Pg. Vall d’Hebron 119-129, 08035 Barcelona, Spain 2Biomedical Research in Cancer Stem Cells, Vall d’Hebron Research Institute, Pg. Vall d’Hebron 119-129, 08035 Barcelona, Spain 3Deparment of Urology, Vall d’Hebron Hospital, Pg. Vall d’Hebron 119-129, 08035 Barcelona, Spain 4Universitat Autònoma de Barcelona, Spain Correspondence to: Rosanna Paciucci, email: [email protected] Keywords: PTOV1, cancer progression, prostate cancer, cancer stem cells, transcription Received: August 26, 2016 Accepted: November 14, 2016 Published: December 22, 2016 SUMMARY Prostate-Tumor-Overexpressed-1 (PTOV1) is a conserved adaptor protein discovered as overexpressed in prostate cancer. Since its discovery, the number of binding partners and associated cellular functions has increased and helped to identify PTOV1 as regulator of gene expression at transcription and translation levels. Its overexpression is associated with increased tumor grade and proliferation in prostate cancer and other neoplasms, including breast, ovarian, nasopharyngeal, squamous laryngeal, hepatocellular and urothelial carcinomas. An important contribution to higher levels of PTOV1 in prostate tumors is given by the frequent rate of gene amplifications, also found in other tumor types. The recent resolution of the structure by NMR of the PTOV domain in PTOV2, also identified as Arc92/ACID1/MED25, has helped to shed light on the functions of PTOV1 as a transcription factor.
    [Show full text]
  • Integrative Computational Biology for Cancer Research
    Hum Genet (2011) 130:465–481 DOI 10.1007/s00439-011-0983-z REVIEW PAPER Integrative computational biology for cancer research Kristen Fortney · Igor Jurisica Received: 3 February 2011 / Accepted: 2 April 2011 / Published online: 22 April 2011 © The Author(s) 2011. This article is published with open access at Springerlink.com Abstract Over the past two decades, high-throughput Introduction (HTP) technologies such as microarrays and mass spec- trometry have fundamentally changed clinical cancer Since the commercialization of DNA microarray technol- research. They have revealed novel molecular markers of ogy in the late 1990s, high-throughput (HTP) data relevant cancer subtypes, metastasis, and drug sensitivity and resis- to cancer research have been accumulating at an ever- tance. Some have been translated into the clinic as tools for increasing rate. These data have led to crucial insights into early disease diagnosis, prognosis, and individualized treat- fundamental cancer biology, including the mechanisms of ment and response monitoring. Despite these successes, tumorigenesis, metastasis, and drug resistance (Rhodes and many challenges remain: HTP platforms are often noisy Chinnaiyan 2005). They have also had enormous clinical and suVer from false positives and false negatives; optimal impact, e.g., several cancers can now be fractionated into analysis and successful validation require complex work- therapeutic subsets with unique prognostic outcomes Xows; and great volumes of data are accumulating at a based on their molecular phenotypes (Buyse et al. 2006; rapid pace. Here we discuss these challenges, and show Dhanasekaran et al. 2001; Lowe et al. 2010; Pegram et al. how integrative computational biology can help diminish 1998; Slamon and Press 2009; Spentzos et al.
    [Show full text]
  • New Insights Into the Interplay Between Non-Coding Rnas and RNA-Binding Protein Hnrnpk in Regulating Cellular Functions
    cells Review New Insights into the Interplay between Non-Coding RNAs and RNA-Binding Protein HnRNPK in Regulating Cellular Functions Yongjie Xu, Wei Wu, Qiu Han, Yaling Wang, Cencen Li, Pengpeng Zhang * and Haixia Xu * College of Life Science, Xinyang Normal University, Xinyang 464000, China; [email protected] (Y.X.); [email protected] (W.W.); [email protected] (Q.H.); [email protected] (Y.W.); [email protected] (C.L.) * Correspondence: [email protected] (P.Z.); [email protected] (H.X.); Tel.: +86-376-6391380 (H.X.) Received: 19 December 2018; Accepted: 15 January 2019; Published: 17 January 2019 Abstract: The emerging data indicates that non-coding RNAs (ncRNAs) epresent more than the “junk sequences” of the genome. Both miRNAs and long non-coding RNAs (lncRNAs) are involved in fundamental biological processes, and their deregulation may lead to oncogenesis and other diseases. As an important RNA-binding protein (RBP), heterogeneous nuclear ribonucleoprotein K (hnRNPK) is known to regulate gene expression through the RNA-binding domain involved in various pathways, such as transcription, splicing, and translation. HnRNPK is a highly conserved gene that is abundantly expressed in mammalian cells. The interaction of hnRNPK and ncRNAs defines the novel way through which ncRNAs affect the expression of protein-coding genes and form autoregulatory feedback loops. This review summarizes the interactions of hnRNPK and ncRNAs in regulating gene expression at transcriptional and post-transcriptional levels or by changing the genomic structure, highlighting their involvement in carcinogenesis, glucose metabolism, stem cell differentiation, virus infection and other cellular functions. Drawing connections between such discoveries might provide novel targets to control the biological outputs of cells in response to different stimuli.
    [Show full text]
  • The Clinical Significance of Small Copy Number Variants In
    JMG Online First, published on August 8, 2014 as 10.1136/jmedgenet-2014-102588 Genome-wide studies J Med Genet: first published as 10.1136/jmedgenet-2014-102588 on 8 August 2014. Downloaded from ORIGINAL ARTICLE The clinical significance of small copy number variants in neurodevelopmental disorders Reza Asadollahi,1 Beatrice Oneda,1 Pascal Joset,1 Silvia Azzarello-Burri,1 Deborah Bartholdi,1 Katharina Steindl,1 Marie Vincent,1 Joana Cobilanschi,1 Heinrich Sticht,2 Rosa Baldinger,1 Regina Reissmann,1 Irene Sudholt,1 Christian T Thiel,3 Arif B Ekici,3 André Reis,3 Emilia K Bijlsma,4 Joris Andrieux,5 Anne Dieux,6 David FitzPatrick,7 Susanne Ritter,8 Alessandra Baumer,1 Beatrice Latal,8 Barbara Plecko,9 Oskar G Jenni,8 Anita Rauch1 ▸ Additional material is ABSTRACT complex syndromes.23Due to the extensive aetio- published online only. To view Background Despite abundant evidence for logical heterogeneity of NDDs, the majority of please visit the journal online (http://dx.doi.org/10.1136/ pathogenicity of large copy number variants (CNVs) in patients remain without aetiological diagnosis, jmedgenet-2014-102588). neurodevelopmental disorders (NDDs), the individual which hampers disease management, genetic coun- significance of genome-wide rare CNVs <500 kb has not selling and in-depth studies of the underlying For numbered affiliations see end of article. been well elucidated in a clinical context. molecular mechanisms. With the advent of new Methods By high-resolution chromosomal microarray genomic technologies, however, diagnostic yield is Correspondence to analysis, we investigated the clinical significance of all steadily improving and a rapidly growing number Professor Anita Rauch, Institute rare non-polymorphic exonic CNVs sizing 1–500 kb in a of novel, aetiologically defined disorders are of Medical Genetics, University of Zurich, Wagistrasse 12, cohort of 714 patients with undiagnosed NDDs.
    [Show full text]
  • Genes Associated with Anhedonia
    Ren et al. Translational Psychiatry (2018) 8:150 DOI 10.1038/s41398-018-0198-3 Translational Psychiatry ARTICLE Open Access Genes associated with anhedonia: a new analysis in a large clinical trial (GENDEP) Hongyan Ren1, Chiara Fabbri2, Rudolf Uher3, Marcella Rietschel 4,OleMors5, Neven Henigsberg 6,JoannaHauser7, Astrid Zobel8, Wolfgang Maier8, Mojca Z. Dernovsek9,DanielSouery10, Annamaria Cattaneo11, Gerome Breen2, Ian W. Craig2,AnneE.Farmer2,PeterMcGuffin2, Cathryn M. Lewis 2 and Katherine J. Aitchison 1,2 Abstract A key feature of major depressive disorder (MDD) is anhedonia, which is a predictor of response to antidepressant treatment. In order to shed light on its genetic underpinnings, we conducted a genome-wide association study (GWAS) followed by investigation of biological pathway enrichment using an anhedonia dimension for 759 patients with MDD in the GENDEP study. The GWAS identified 18 SNPs associated at genome-wide significance with the top one being an intronic SNP (rs9392549) in PRPF4B (pre-mRNA processing factor 4B) located on chromosome 6 (P = 2.07 × 10−9) while gene-set enrichment analysis returned one gene ontology term, axon cargo transport (GO: 0008088) with a nominally significant P value (1.15 × 10−5). Furthermore, our exploratory analysis yielded some interesting, albeit not statistically significant genetic correlation with Parkinson’s Disease and nucleus accumbens gray matter. In addition, polygenic risk scores (PRSs) generated from our association analysis were found to be able to predict treatment efficacy of the antidepressants in this study. In conclusion, we found some markers significantly associated with anhedonia, and some suggestive findings of related pathways and biological functions, which could be further investigated in other studies.
    [Show full text]
  • Content Based Search in Gene Expression Databases and a Meta-Analysis of Host Responses to Infection
    Content Based Search in Gene Expression Databases and a Meta-analysis of Host Responses to Infection A Thesis Submitted to the Faculty of Drexel University by Francis X. Bell in partial fulfillment of the requirements for the degree of Doctor of Philosophy November 2015 c Copyright 2015 Francis X. Bell. All Rights Reserved. ii Acknowledgments I would like to acknowledge and thank my advisor, Dr. Ahmet Sacan. Without his advice, support, and patience I would not have been able to accomplish all that I have. I would also like to thank my committee members and the Biomed Faculty that have guided me. I would like to give a special thanks for the members of the bioinformatics lab, in particular the members of the Sacan lab: Rehman Qureshi, Daisy Heng Yang, April Chunyu Zhao, and Yiqian Zhou. Thank you for creating a pleasant and friendly environment in the lab. I give the members of my family my sincerest gratitude for all that they have done for me. I cannot begin to repay my parents for their sacrifices. I am eternally grateful for everything they have done. The support of my sisters and their encouragement gave me the strength to persevere to the end. iii Table of Contents LIST OF TABLES.......................................................................... vii LIST OF FIGURES ........................................................................ xiv ABSTRACT ................................................................................ xvii 1. A BRIEF INTRODUCTION TO GENE EXPRESSION............................. 1 1.1 Central Dogma of Molecular Biology........................................... 1 1.1.1 Basic Transfers .......................................................... 1 1.1.2 Uncommon Transfers ................................................... 3 1.2 Gene Expression ................................................................. 4 1.2.1 Estimating Gene Expression ............................................ 4 1.2.2 DNA Microarrays ......................................................
    [Show full text]
  • Maternal Low-Fat Diet Programs the Hepatic Epigenome Despite Exposure to an Obesogenic Postnatal Diet
    nutrients Article Maternal Low-Fat Diet Programs the Hepatic Epigenome despite Exposure to an Obesogenic Postnatal Diet Laura Moody 1, Justin Shao 2, Hong Chen 3 and Yuan-Xiang Pan 4,* 1 Division of Nutritional Sciences, University of Illinois at Urbana-Champaign, Urbana, IL 61801, USA 2 Department of Food Science and Human Nutrition, University of Illinois at Urbana-Champaign, Exeter High School, 1 Blue Hawk Drive, Exeter, NH 03833, USA 3 Department of Food Science and Human Nutrition, Division of Nutritional Sciences, University of Illinois at Urbana-Champaign, Urbana, IL 61801, USA 4 Department of Food Science and Human Nutrition, Division of Nutritional Sciences, and Illinois Informatics Institute, University of Illinois at Urbana-Champaign, Urbana, IL 61801, USA * Correspondence: [email protected]; Tel.: +1-217-333-3466 Received: 29 June 2019; Accepted: 27 August 2019; Published: 3 September 2019 Abstract: Obesity and metabolic disease present a danger to long-term health outcomes. It has been hypothesized that epigenetic marks established during early life might program individuals and have either beneficial or harmful consequences later in life. In the present study, we examined whether maternal diet alters DNA methylation and whether such modifications persist after an obesogenic postnatal dietary challenge. During gestation and lactation, male Sprague-Dawley rats were exposed to either a high-fat diet (HF; n = 10) or low-fat diet (LF; n = 10). After weaning, all animals were fed a HF diet for an additional nine weeks. There were no differences observed in food intake or body weight between groups. Hepatic DNA methylation was quantified using both methylated DNA immunoprecipitation sequencing (MeDIP-seq) and methylation-sensitive restriction enzyme sequencing (MRE-seq).
    [Show full text]
  • Methods and Models for the Analysis of Biological Signifïcance Based on High-Throughput Data
    Methods and Models for the Analysis of Biological Signifïcance Based on High-Throughput Data Jose Luis Mosquera Mayo Aquesta tesi doctoral està subjecta a la llicència Reconeixement- NoComercial – CompartirIgual 3.0. Espanya de Creative Commons. Esta tesis doctoral está sujeta a la licencia Reconocimiento - NoComercial – CompartirIgual 3.0. España de Creative Commons. This doctoral thesis is licensed under the Creative Commons Attribution-NonCommercial- ShareAlike 3.0. Spain License. Methods and Models for the Analysis of Biological Significance Based on High Throughput Data Jose Luis Mosquera Mayo 2 Methods and Models for the Analysis of Biological Significance Based on High Throughput Data M`etodes i Models per a l’An`aliside la Significaci´oBiol`ogicaBasada en Dades d'Alt Rendiment Mem`oriapresentada per en Jose Luis Mosquera Mayo per optar al grau de doctor per la Universitat de Barcelona SIGNATURES El Doctorand: Jose Luis Mosquera Mayo El Director de la Tesi: Dr. Alexandre Sanchez´ Pla El Tutor de la Tesi: Dr. Josep Mar´ıa Oller Sala Programa de doctorat en estad´ıstica Departament d'Estad´ıstica,Facultat de Biologia Universitat de Barcelona 4 Acknowledgements \A teacher affects eternity, he can never tell where his influence stops." Henry Brooks Adams \Mathematicians do not study objects, but relations between objects." Jules Henri Poincar´e It is truly an honor for me to address these words of gratitude to everyone who has contributed, to a greater or lesser extent by supporting me during this exciting program of studies, to help make my lifelong dream come true: to complete my doctoral dissertation. First and foremost, I would like (and feel compelled) to mention a special debt of gratitude to Prof.
    [Show full text]
  • Investigating the Effects of Chronic Perinatal Alcohol and Combined
    www.nature.com/scientificreports OPEN Investigating the efects of chronic perinatal alcohol and combined nicotine and alcohol exposure on dopaminergic and non‑dopaminergic neurons in the VTA Tina Kazemi, Shuyan Huang, Naze G. Avci, Yasemin M. Akay & Metin Akay* The ventral tegmental area (VTA) is the origin of dopaminergic neurons and the dopamine (DA) reward pathway. This pathway has been widely studied in addiction and drug reinforcement studies and is believed to be the central processing component of the reward circuit. In this study, we used a well‑ established rat model to expose mother dams to alcohol, nicotine‑alcohol, and saline perinatally. DA and non‑DA neurons collected from the VTA of the rat pups were used to study expression profles of miRNAs and mRNAs. miRNA pathway interactions, putative miRNA‑mRNA target pairs, and downstream modulated biological pathways were analyzed. In the DA neurons, 4607 genes were diferentially upregulated and 4682 were diferentially downregulated following nicotine‑alcohol exposure. However, in the non‑DA neurons, only 543 genes were diferentially upregulated and 506 were diferentially downregulated. Cell proliferation, diferentiation, and survival pathways were enriched after the treatments. Specifcally, in the PI3K/AKT signaling pathway, there were 41 miRNAs and 136 mRNAs diferentially expressed in the DA neurons while only 16 miRNAs and 20 mRNAs were diferentially expressed in the non‑DA neurons after the nicotine‑alcohol exposure. These results depicted that chronic nicotine and alcohol exposures during pregnancy diferentially afect both miRNA and gene expression profles more in DA than the non‑DA neurons in the VTA. Understanding how the expression signatures representing specifc neuronal subpopulations become enriched in the VTA after addictive substance administration helps us to identify how neuronal functions may be altered in the brain.
    [Show full text]