Péter Antal Ádám Arany Bence Bolgár András Gézsi Gergely Hajós Gábor Hullám Péter Marx András Millinghoffer László Poppe Péter Sárközy BIOINFORMATICS

Similar documents
Bioinformatics. Created by XMLmind XSL-FO Converter.

This place covers: Methods or systems for genetic or protein-related data processing in computational molecular biology.

Capabilities & Services

Data Mining and Applications in Genomics

Biology 644: Bioinformatics

Computational methods in bioinformatics: Lecture 1

Transcriptomics analysis with RNA seq: an overview Frederik Coppens

GREG GIBSON SPENCER V. MUSE

Pioneering Clinical Omics

Introduction to RNA-Seq. David Wood Winter School in Mathematics and Computational Biology July 1, 2013

Pharmacogenetics: A SNPshot of the Future. Ani Khondkaryan Genomics, Bioinformatics, and Medicine Spring 2001

Welcome to the NGS webinar series

Microbial Metabolism Systems Microbiology

CS273B: Deep Learning in Genomics and Biomedicine. Recitation 1 30/9/2016

Introductie en Toepassingen van Next-Generation Sequencing in de Klinische Virologie. Sander van Boheemen Medical Microbiology

AGILENT S BIOINFORMATICS ANALYSIS SOFTWARE

Sequencing technologies. Jose Blanca COMAV institute bioinf.comav.upv.es

Understanding the science and technology of whole genome sequencing

Genetics and Bioinformatics

resequencing storage SNP ncrna metagenomics private trio de novo exome ncrna RNA DNA bioinformatics RNA-seq comparative genomics

TECHNOLOGIES, PRODUCTS & SERVICES for MOLECULAR DIAGNOSTICS, MDx ABA 298

Types of Databases - By Scope

Marker types. Potato Association of America Frederiction August 9, Allen Van Deynze

Contact us for more information and a quotation

Sequencing technologies. Jose Blanca COMAV institute bioinf.comav.upv.es

Motivation From Protein to Gene

RNA-SEQUENCING ANALYSIS

SolCAP. Executive Commitee : David Douches Walter De Jong Robin Buell David Francis Alexandra Stone Lukas Mueller AllenVan Deynze

ALGORITHMS IN BIO INFORMATICS. Chapman & Hall/CRC Mathematical and Computational Biology Series A PRACTICAL INTRODUCTION. CRC Press WING-KIN SUNG

Sequencing technologies. Jose Blanca COMAV institute bioinf.comav.upv.es

Era with Computational Biology/Toxicology

Prostate Cancer Genetics: Today and tomorrow

Outline General NGS background and terms 11/14/2016 CONFLICT OF INTEREST. HLA region targeted enrichment. NGS library preparation methodologies

The application of hidden markov model in building genetic regulatory network

The Expanded Illumina Sequencing Portfolio New Sample Prep Solutions and Workflow

Lecture #1. Introduction to microarray technology

Informatic Issues in Genomics

Phenotype analysis: biological-biochemical analysis. Genotype analysis: molecular and physical analysis

Chapter 6 - Molecular Genetic Techniques

Single Nucleotide Polymorphisms: Methods And Protocols (Methods In Molecular Biology)

Frumkin, 2e Part 1: Methods and Paradigms. Chapter 6: Genetics and Environmental Health

Introduction to Bioinformatics and Gene Expression Technologies

Introduction to Bioinformatics and Gene Expression Technologies

VALLIAMMAI ENGINEERING COLLEGE

From Bench to Bedside: Role of Informatics. Nagasuma Chandra Indian Institute of Science Bangalore

2/23/16. Protein-Protein Interactions. Protein Interactions. Protein-Protein Interactions: The Interactome

Smart India Hackathon

BIOINFORMATICS AND SYSTEM BIOLOGY (INTERNATIONAL PROGRAM)

Cover Page. The handle holds various files of this Leiden University dissertation.

Following text taken from Suresh Kumar. Bioinformatics Web - Comprehensive educational resource on Bioinformatics. 6th May.2005

Chapter 17 Lecture. Concepts of Genetics. Tenth Edition. Regulation of Gene Expression in Eukaryotes

SNPs - GWAS - eqtls. Sebastian Schmeier

Bi 8 Lecture 4. Ellen Rothenberg 14 January Reading: from Alberts Ch. 8

Phenotype analysis: biological-biochemical analysis. Genotype analysis: molecular and physical analysis

Introduction to Bioinformatics and Gene Expression Technology

MATH 5610, Computational Biology

Agilent GeneSpring GX 10: Beyond. Pam Tangvoranuntakul Product Manager, GeneSpring October 1, 2008

Applicazioni biotecnologiche

Bioinformatics. Dick de Ridder. Tuinbouw Digitaal, 12/11/15

Axiom Biobank Genotyping Solution

ROAD TO STATISTICAL BIOINFORMATICS CHALLENGE 1: MULTIPLE-COMPARISONS ISSUE

GENETICS - CLUTCH CH.15 GENOMES AND GENOMICS.

Introduction to human genomics and genome informatics

Just the Facts: A Basic Introduction to the Science Underlying NCBI Resources

THE HEALTH AND RETIREMENT STUDY: GENETIC DATA UPDATE

The University of California, Santa Cruz (UCSC) Genome Browser

Chapter 1. from genomics to proteomics Ⅱ

02 Agenda Item 03 Agenda Item

Syllabus for BIOS 101, SPRING 2013

Sequence Variations. Baxevanis and Ouellette, Chapter 7 - Sequence Polymorphisms. NCBI SNP Primer:

Unit 6: Molecular Genetics & DNA Technology Guided Reading Questions (100 pts total)

Genome-Wide Association Studies (GWAS): Computational Them

Chapter 10 Genetic Engineering: A Revolution in Molecular Biology

Quality assurance in NGS (diagnostics)

Bioinformatics Advice on Experimental Design

Chapter 7. DNA Microarrays

Human Genetic Variation. Ricardo Lebrón Dpto. Genética UGR

Introduction to Bioinformatics

QIAGEN s NGS Solutions for Biomarkers NGS & Bioinformatics team QIAGEN (Suzhou) Translational Medicine Co.,Ltd

Single-cell RNA-sequencing

BIOINF/BENG/BIMM/CHEM/CSE 184: Computational Molecular Biology. Lecture 2: Microarray analysis

Aaron Liston, Oregon State University Botany 2012 Intro to Next Generation Sequencing Workshop

Structure, Measurement & Analysis of Genetic Variation

Genomic resources. for non-model systems

Get to Know Your DNA. Every Single Fragment.

From Genotype to Phenotype

BIMM 143: Introduction to Bioinformatics (Winter 2018)

CS262 Lecture 12 Notes Single Cell Sequencing Jan. 11, 2016

Perspectives on the Priorities for Bioinformatics Education in the 21 st Century

Functional Genomics Overview RORY STARK PRINCIPAL BIOINFORMATICS ANALYST CRUK CAMBRIDGE INSTITUTE 18 SEPTEMBER 2017

Matthew Tinning Australian Genome Research Facility. July 2012

Introduction to some aspects of molecular genetics

The Biotechnology Toolbox

Microarray Gene Expression Analysis at CNIO

From Proteomics to Systems Biology. Integration of omics - information

Chapter 15 Gene Technologies and Human Applications

Data representation for clinical data and metadata

Genomic region (ENCODE) Gene definitions

Redefine what s possible with the Axiom Genotyping Solution

Human Genomics. Higher Human Biology

Transcription:

Péter Antal Ádám Arany Bence Bolgár András Gézsi Gergely Hajós Gábor Hullám Péter Marx András Millinghoffer László Poppe Péter Sárközy BIOINFORMATICS The Bioinformatics book covers new topics in the rapidly expanding field of bioinformatics, from next-generation sequencing to drug discovery and metagenomics. The first two chapters overviews genetic measurement methods. The next four chapters discuss topics related to the effect of genetic variants from protein modeling to gene regulatory networks. Standard statistical analysis in association studies are discussed in the next two chapters. The systems biology approach is illustrated by discussing a systems-based biomarker analysis method, the graph-based network science, the dynamical systems based approaches and a Bayesian causal inference method in subsequent chapters. The next chapter discusses text-mining methods in biomedicine, especially their application in interpretation and translation. The decision theoretic approach to study design, especially multi-stage, sequential study design is discussed in the next chapter, introducing the concepts of value of information and the expected value of an experiment. Next, the heterogeneity of biomedical big data sources is overviewed, together with data and knowledge fusion methods, and with the discussion of semantic publishing, which can lead to a new unification of biomedicine. Subsequently, bioinformatic workflow methods are summarized. At last, drug discovery methods are overviewed with an outlook for personalized medicine and the final chapter presents the main steps and workflows in metagenomics. Keywords: genotyping, next-generation sequencing methods, protein modeling, gene regulatory networks, omic networks, study design, data and knowledge fusion, worklfow systems, association study, biomarker analysis, medical decision support systems, semantic publishing, similarity based drug discovery, metagenomics. Budapest University of Technology & Economics and Semmelweis University Typotex Kiadó 2014

COPYRIGHT: 2014 2019, Péter Antal, Ádám Arany, Bence Bolgár, András Gézsi, Gergely Hajós, Gábor Hullám, Péter Marx, András Millinghoffer, László Poppe, Péter Sárközy, Budapest University of Technology and Economics, Semmelweis University Creative Commons NonCommercial-NoDerivs 3.0 (CC BY-NC-ND 3.0) Terms of use of : This work can be reproduced, circulated, published and performed for non-commercial purposes without restriction by indicating the author s name, but it cannot be modified. Scientific lectors: Viktor Molnár, András Antos ISBN 978 963 279 179 1 Prepared under the editorship of Typotex Kiadó Responsible manager: Zsuzsa Votisky Prepared within the framework of the project Konzorcium a biotechnológia aktív tanulásáért ( Consortium for the Active Studying of Biotechnology ) Grant No. TÁMOP- 4.1.2/A/1-11/1-2011-0079.

Contents 1 DNA recombinant measurement technology, noise and error models 11 1.1 Historic overview................................ 11 1.1.1 Clinical aspects of genome sequencing................. 12 1.1.2 Partial Genetic Association Studies.................. 12 1.1.3 Genome Wide Association Studies................... 12 1.2 First generation automated Sanger sequencing................ 12 1.3 Next generation sequencing technologies................... 14 1.3.1 Pyrosequencing and ph based sequencing............... 15 1.3.2 Reversible terminator based sequencing................ 16 1.3.3 Nanopore based sequencing...................... 16 1.4 Error characteristics of Next Generation Sequencing............. 16 1.4.1 Carry forward/incomplete extension.................. 18 1.4.2 Homopolymer errors.......................... 18 1.5 Capture technologies.............................. 19 1.5.1 PCR capture.............................. 19 1.6 Emulsion PCR................................. 21 1.7 Bridge amplification.............................. 24 1.8 Targeted resequencing............................. 24 1.9 De-novo sequencing............................... 25 1.10 Next generation sequencing workflows..................... 25 1.10.1 Filtering................................. 25 1.10.2 Mapping................................. 25 1.10.3 Assembly................................ 26 1.10.4 Variant calling............................. 26 1.10.5 Paired end sequencing......................... 26 1.11 Multiplexing samples.............................. 26 2 The post-processing, haplotype reconstruction, and imputation... 28 2.1 Genome..................................... 28 2.2 Genotype.................................... 28 2.2.1 Single nucleotide polymorphisms................... 29 2.2.2 Types of point mutation........................ 30 2.3 Haplotypes and recombination......................... 30

Contents 4 2.4 Linkage Disequilibrium............................. 31 2.5 Haplotype reconstruction............................ 32 2.6 Imputation................................... 33 2.7 Genotyping platforms.............................. 33 2.7.1 Sample preparation........................... 34 2.7.2 Regions of interest........................... 34 2.7.3 Primer Design.............................. 35 2.7.4 PCR................................... 35 2.7.5 Probe-tag based genotyping...................... 37 2.7.6 Sanger sequencing............................ 37 2.7.7 Real-time qualitative polymerase chain reaction........... 37 2.7.8 SNP arrays............................... 38 2.8 Genotyping vs. gene expression........................ 38 2.8.1 Call rate and accuracy......................... 39 3 Comparative protein modeling and molecular docking 40 3.1 Introduction................................... 40 3.1.1 The protein structure gap....................... 40 3.1.2 Methods of protein modeling...................... 41 3.2 Comparative protein modeling......................... 43 3.2.1 Steps of homology modeling...................... 43 3.2.2 Tools for homology modeling...................... 48 3.3 Molecular docking................................ 50 3.3.1 Protein-ligand interaction predictions................. 51 3.3.2 Protein-biomacromolecule interaction predictions.......... 51 4 Methods of determining structure of proteins and protein structure databases 56 4.1 Introduction................................... 56 4.1.1 Protein identification tools....................... 56 4.1.2 Simple protein analyses......................... 57 4.1.3 Levels and problems of protein structure predictions......... 57 4.2 Experimental methods to determine the secondary structure of proteins.. 58 4.2.1 Protein circular dichroism (CD).................... 59 4.2.2 Synchrotron radiation circular dichroism (SRCD).......... 60 4.3 Experimental methods to determining atomic structures of proteins.... 60 4.3.1 Protein X-ray crystallography..................... 61 4.3.2 Protein NMR spectroscopy....................... 63 4.3.3 Protein electron microscopy, electron diffraction and electron crystallography............................... 66 4.3.4 Protein neutron crystallography.................... 67

Contents 5 5 Quantitative models of the functional effects of genetic variants 70 5.1 Introduction................................... 70 5.2 Variants..................................... 70 5.2.1 SNP, indel................................ 71 5.2.2 Alternative splicing........................... 71 5.3 Levels of regulation............................... 72 5.4 Different regulatory elements.......................... 72 5.5 microrna.................................... 72 5.5.1 mirna development.......................... 73 5.5.2 mirna regulatory methods...................... 73 5.6 Transcription factors.............................. 73 5.7 Epigenetics................................... 74 5.7.1 Methylation............................... 74 5.7.2 Histone modifications.......................... 74 6 Mathematical models of gene regulatory networks 78 6.1 Introduction................................... 78 6.2 Learning networks............................... 78 6.2.1 Representation............................. 78 6.2.2 Types of network learning algorithms................. 78 6.3 TF, mirna, mrna regulatory networks................... 81 7 Standard analysis of genetic association studies 84 7.1 Introduction................................... 84 7.2 Genetic data transformation.......................... 85 7.2.1 Filtering................................. 85 7.2.2 Standard test for Hardy Weinberg equilibrium............ 85 7.3 Phenotype data transformation........................ 86 7.3.1 Transformation............................. 86 7.3.2 Discretization.............................. 87 7.4 Univariate analysis methods.......................... 87 7.4.1 Standard association tests....................... 87 7.4.2 Cochran Armitage test for trend................... 89 7.4.3 Odds ratios............................... 90 7.4.4 Univariate Bayesian methods..................... 92 7.5 Multivariate analysis methods......................... 92 7.5.1 Logistic regression........................... 92 7.5.2 Haplotype association......................... 93 7.5.3 Analysis of statistical power...................... 97 8 Analyzing gene expression studies 101 8.1 Introduction................................... 101 8.2 Pre-procession.................................. 102

Contents 6 8.2.1 Background correction......................... 102 8.2.2 Normalization.............................. 102 8.2.3 Summarization............................. 103 8.2.4 Filtering................................. 104 8.3 Data analysis.................................. 104 8.3.1 Clustering................................ 104 8.3.2 Differential expression......................... 108 8.3.3 Biological interpretation of results................... 110 9 Biomarker analysis 115 Notation........................................ 115 9.1 Introduction................................... 118 9.2 Background................................... 118 9.3 Bayesian multilevel analysis of relevance................... 120 9.4 Multivariate scalability: k-mbs and k-mbg features............ 121 9.5 A knowledge-rich aggregation of input features................ 122 9.6 Interaction, redundancy based on posterior decomposition......... 122 9.7 Relevance for multiple targets......................... 123 9.8 Conditional and contextual relevance..................... 124 9.9 Posteriors for the predictive power of input features............. 125 9.10 Algorithmic aspects and applications..................... 125 9.11 Summary.................................... 125 10 Network biology 129 10.1 Introduction................................... 129 10.2 Biological networks............................... 130 10.3 Basics of graph theory............................. 131 10.4 Network analysis................................ 132 10.4.1 Network topology............................ 132 10.4.2 Network models and dynamics..................... 133 10.4.3 Assortativity, degree distribution and scale-free networks...... 134 10.4.4 Tasks and challenges.......................... 135 10.5 An application to drug discovery....................... 137 11 Dynamic modeling in cell biology 141 11.1 Biochemical concepts and their computational representations....... 141 11.2 Modeling with ordinary differential equations................. 144 11.3 Stochastic modeling............................... 145 11.4 Hybrid methods................................. 146 11.5 Reaction diffusion systems........................... 147 11.6 Model fitting.................................. 148 11.7 Whole-cell simulation.............................. 149 11.8 Overview..................................... 149

Contents 7 12 Causal inference in biomedicine 152 Notation..................................... 152 12.1 Introduction................................... 155 12.2 Representing independence and causal relations by Bayesian networks... 155 12.3 Constraint based inference of causal relations and models.......... 159 12.4 Learning complete causal domain models................... 160 12.5 Bayesian inference of causal features..................... 161 12.5.1 Edges: direct pairwise dependencies.................. 161 12.5.2 Pairwise causal relations........................ 162 12.5.3 MBG subnetworks........................... 162 12.5.4 Ordering of the variables........................ 162 12.5.5 Effect modifiers............................. 163 13 Text mining methods in bioinformatics 168 13.1 Introduction................................... 168 13.2 Biomedical text mining............................. 168 13.2.1 Constructing the corpus........................ 169 13.2.2 Constructing the vocabulary...................... 171 13.2.3 Text mining tasks............................ 172 13.3 Basic techniques................................. 173 13.3.1 Pattern matching............................ 173 13.3.2 Document representation........................ 173 13.3.3 Methods for named entity recognition................. 175 13.3.4 Methods for relation extraction.................... 175 13.3.5 Lexicalized probabilistic context-free grammars........... 176 13.3.6 Difficulties in biomedical text mining................. 178 13.4 Text mining and knowledge management................... 179 14 Experimental design: from the basics to active learning extensions 182 14.1 Introduction................................... 182 14.2 The elements of experimental design..................... 182 14.2.1 Phases of biomedical DOE....................... 183 14.2.2 Types of biological experiments.................... 183 14.3 A decision theoretic approach to DoE..................... 185 14.3.1 Expected value of an experiment................... 185 14.3.2 Adaptive designs and budgeted learning............... 186 14.3.3 A Bayesian treatment of sequential decision processes........ 188 14.4 Approaches to target variable selection.................... 189 14.4.1 Gene Prioritization........................... 189 14.4.2 Active learning............................. 191 14.4.3 Other practical tasks relying on bioinformatics............ 191

Contents 8 15 Big data in biomedicine 194 15.1 Introduction................................... 194 15.2 The first wave of biomedical big data..................... 196 15.3 Post-genomic big data: the second wave................... 196 15.4 The common big data............................. 197 15.5 The health-related common big data in biomedicine............. 199 15.6 Bioinformatic challenges of common big data................. 201 16 Analysis of heterogeneous biomedical data through information fusion 206 16.1 Introduction................................... 206 16.2 Information fusion and data fusion...................... 208 16.3 Types of data fusion.............................. 209 16.3.1 Early fusion............................... 210 16.3.2 Intermediate fusion........................... 211 16.3.3 Late fusion............................... 211 16.4 Similarity-based data fusion.......................... 212 17 The Bayesian Encyclopedia 217 17.1 Introduction................................... 217 17.2 The three worlds of data, knowledge and computation............ 220 17.3 From fragmentation problems to workflow for unification.......... 221 17.4 Data repositories with semantic technologies................. 224 17.5 Semantic publishing for the literature world................. 224 17.6 Causal Bayesian network-based data analytic knowledge bases....... 227 17.7 Examples for links between worlds....................... 228 17.8 Prospects for the Bayesian Encyclopedia................... 228 18 Bioinformatical workflow systems case study 235 18.1 Overview of tasks................................ 235 18.2 Data model and representation........................ 236 18.3 Use cases and architecture........................... 237 18.4 Implementation details of the server...................... 238 18.5 Postprocessing steps.............................. 240 19 Computational aspects of pharmaceutical research 241 19.1 Overview of the process............................ 241 19.2 Chemoinformatical background........................ 242 19.3 Screening criteria................................ 244 19.4 Method..................................... 247 19.5 Fragment-based design............................. 249 19.6 Drug repositioning............................... 250

Contents 9 20 Metagenomics 253 20.1 Introduction................................... 253 20.2 Metagenome analysis.............................. 254 20.2.1 Community profiling.......................... 254 20.2.2 Functional metagenomics........................ 255 20.3 Metagenomics step by step........................... 255 20.3.1 Sampling................................ 255 20.3.2 Sequencing............................... 257 20.3.3 Assembly................................ 257 20.3.4 Binning................................. 258 20.3.5 Gene calling and functional inference................. 259