The most common technologies and tools for functional genome analysis

Since the sequence of the human genome is complete, the main issue is how to understand the information written in the DNA sequence. Despite numerous genome-wide studies that have already been performed, the challenge to determine the function of genes, gene products, and also their interaction is still open. As changes in the human genome are highly likely to cause pathological conditions, functional analysis is vitally important for human health. For many years there have been a variety of technologies and tools used in functional genome analysis. However, only in the past decade there has been rapid revolutionizing progress and improvement in high-throughput methods, which are ranging from traditional real-time polymerase chain reaction to more complex systems, such as next-generation sequencing or mass spectrometry. Furthermore, not only laboratory investigation, but also accurate bioinformatic analysis is required for reliable scientific results. These methods give an opportunity for accurate and comprehensive functional analysis that involves various fields of studies: genomics, epigenomics, proteomics, and interactomics. This is essential for filling the gaps in the knowledge about dynamic biological processes at both cellular and organismal level. However, each method has both advantages and limitations that should be taken into account before choosing the right method for particular research in order to ensure successful study. For this reason, the present review paper aims to describe the most frequent and widely-used methods for the comprehensive functional analysis.

[1]  P. Laird Principles and challenges of genome-wide DNA methylation analysis , 2010, Nature Reviews Genetics.

[2]  Uwe Schlattner,et al.  Yeast Two-Hybrid, a Powerful Tool for Systems Biology , 2009, International journal of molecular sciences.

[3]  C. McHale,et al.  Functional genomic screening approaches in mechanistic toxicology and potential future applications of CRISPR-Cas9. , 2015, Mutation research. Reviews in mutation research.

[4]  E. Bakker,et al.  Is the DNA sequence the gold standard in genetic testing? Quality of molecular genetic tests assessed. , 2006, Clinical chemistry.

[5]  Xuelin Huang,et al.  An improvement of the 2ˆ(-delta delta CT) method for quantitative real-time polymerase chain reaction data analysis. , 2013, Biostatistics, bioinformatics and biomathematics.

[6]  G. Loots Genomic identification of regulatory elements by evolutionary sequence comparison and functional analysis. , 2008, Advances in genetics.

[7]  M. Schlaak,et al.  Comparison of high resolution melting analysis, pyrosequencing, next generation sequencing and immunohistochemistry to conventional Sanger sequencing for the detection of p.V600E and non-p.V600E BRAF mutations , 2014, BMC Cancer.

[8]  E. van Duijn Current limitations in native mass spectrometry based structural biology. , 2010, Journal of the American Society for Mass Spectrometry.

[9]  Yong-shu He,et al.  [Structural variation in the human genome]. , 2009, Yi chuan = Hereditas.

[10]  Ryan Bishop,et al.  Applications of fluorescence in situ hybridization (FISH) in detecting genetic aberrations of medical significance , 2010 .

[11]  M. Bullock,et al.  DNA Methylation Analysis: Choosing the Right Method , 2016, Biology.

[12]  D. Altshuler,et al.  A map of human genome variation from population-scale sequencing , 2010, Nature.

[13]  Thierry Rabilloud,et al.  Two-dimensional gel electrophoresis in proteomics: a tutorial. , 2011, Journal of proteomics.

[14]  R. Stöger,et al.  Errors in the bisulfite conversion of DNA: modulating inappropriate- and failed-conversion frequencies , 2008, Nucleic acids research.

[15]  Alexander G Williams,et al.  RNA‐seq Data: Challenges in and Recommendations for Experimental Design and Analysis , 2014, Current protocols in human genetics.

[16]  M. Gerstein,et al.  RNA-Seq: a revolutionary tool for transcriptomics , 2009, Nature Reviews Genetics.

[17]  Michael R. Green,et al.  Transcriptional regulatory elements in the human genome. , 2006, Annual review of genomics and human genetics.

[18]  John R Yates,et al.  Proteomics by mass spectrometry: approaches, advances, and applications. , 2009, Annual review of biomedical engineering.

[19]  James E. DiCarlo,et al.  RNA-Guided Human Genome Engineering via Cas9 , 2013, Science.

[20]  M. Wagner,et al.  Advantages and limitations of quantitative PCR (Q-PCR)-based approaches in microbial ecology. , 2009, FEMS microbiology ecology.

[21]  Martin Kircher,et al.  High‐throughput DNA sequencing – concepts and limitations , 2010, BioEssays : news and reviews in molecular, cellular and developmental biology.

[22]  Erez Lieberman Aiden,et al.  The expanding scope of DNA sequencing , 2012, Nature Biotechnology.

[23]  M. Marra,et al.  Applications of next-generation sequencing technologies in functional genomics. , 2008, Genomics.

[24]  K. Bunai,et al.  Effectiveness and limitation of two-dimensional gel electrophoresis in bacterial membrane protein proteomics and perspectives. , 2005, Journal of chromatography. B, Analytical technologies in the biomedical and life sciences.

[25]  C. Ponting,et al.  Finishing the euchromatic sequence of the human genome , 2004 .

[26]  R. Barrangou,et al.  Cas9–crRNA ribonucleoprotein complex mediates specific DNA cleavage for adaptive immunity in bacteria , 2012, Proceedings of the National Academy of Sciences.

[27]  Steven Henikoff,et al.  Genome-wide analysis of DNA methylation patterns , 2007, Development.

[28]  Y. Ko,et al.  Epigenomics: the science of no-longer-junk DNA. Why study it in chronic kidney disease? , 2013, Seminars in nephrology.

[29]  E. Bunnik,et al.  An Introduction to Functional Genomics and Systems Biology. , 2013, Advances in wound care.

[30]  J. Guénet Chemical mutagenesis of the mouse genome: an overview , 2004, Genetica.

[31]  Hanlee P. Ji,et al.  Next-generation DNA sequencing , 2008, Nature Biotechnology.

[32]  Gregory D. Schuler,et al.  Database resources of the National Center for Biotechnology Information: update , 2004, Nucleic acids research.

[33]  Michael Krawczak,et al.  Where genotype is not predictive of phenotype: towards an understanding of the molecular basis of reduced penetrance in human inherited disease , 2013, Human Genetics.

[34]  L. Hood,et al.  Complementary Profiling of Gene Expression at the Transcriptome and Proteome Levels in Saccharomyces cerevisiae*S , 2002, Molecular & Cellular Proteomics.

[35]  Takeru Nakazato,et al.  Experimental Design-Based Functional Mining and Characterization of High-Throughput Sequencing Data in the Sequence Read Archive , 2013, PloS one.

[36]  M. Ascano,et al.  Multi-disciplinary methods to define RNA-protein interactions and regulatory networks. , 2013, Current opinion in genetics & development.

[37]  Magnus Lundgren,et al.  The CRISPR-Cas immune system: biology, mechanisms and applications. , 2015, Biochimie.

[38]  Stefan Rödiger,et al.  A survey of tools for the analysis of quantitative PCR (qPCR) data , 2014, Biomolecular detection and quantification.

[39]  F. Stanczyk,et al.  Advantages and challenges of mass spectrometry assays for steroid hormones , 2010, The Journal of Steroid Biochemistry and Molecular Biology.

[40]  Andrew J. Bannister,et al.  Regulation of chromatin by histone modifications , 2011, Cell Research.

[41]  J. Oost,et al.  Unravelling the structural and mechanistic basis of CRISPR–Cas systems , 2014, Nature Reviews Microbiology.

[42]  K. Tarte,et al.  Risk of tumorigenicity in mesenchymal stromal cell-based therapies--bridging scientific observations and regulatory viewpoints. , 2013, Cytotherapy.

[43]  Bassem A Bejjani,et al.  Application of array-based comparative genomic hybridization to clinical diagnostics. , 2006, The Journal of molecular diagnostics : JMD.

[44]  D. Botstein,et al.  Comparing whole genomes using DNA microarrays , 2008, Nature Reviews Genetics.

[45]  K. Polyak,et al.  Serial analysis of gene expression , 2006, Nature Protocols.

[46]  B. Oliver,et al.  Microarrays, deep sequencing and the true measure of the transcriptome , 2011, BMC Biology.

[47]  Jörg D. Hoheisel,et al.  Analysis of DNA–protein interactions: from nitrocellulose filter binding assays to microarray studies , 2010, Analytical and bioanalytical chemistry.

[48]  Hiroshi Kimura,et al.  Histone modifications for human epigenome analysis , 2013, Journal of Human Genetics.

[49]  M. Stephens,et al.  RNA-seq: an assessment of technical reproducibility and comparison with gene expression arrays. , 2008, Genome research.

[50]  R. Nelson,et al.  Mass spectrometric immunoassay. , 1995, Analytical chemistry.

[51]  Matthew E Hurles,et al.  The population genetics of structural variation , 2007, Nature Genetics.

[52]  T. Glisovic,et al.  RNA‐binding proteins and post‐transcriptional gene regulation , 2008, FEBS letters.

[53]  S. Gygi,et al.  Correlation between Protein and mRNA Abundance in Yeast , 1999, Molecular and Cellular Biology.

[54]  S. Schuster Next-generation sequencing transforms today's biology , 2008, Nature Methods.

[55]  Steven J. M. Jones,et al.  Genome-wide identification of DNA-protein interactions using chromatin immunoprecipitation coupled with flow cell sequencing. , 2009, The Journal of endocrinology.

[56]  Robert J. Brooker,et al.  Genetics: Analysis and Principles , 1998 .

[57]  P. Collas The Current State of Chromatin Immunoprecipitation , 2010, Molecular biotechnology.

[58]  Data production leads,et al.  An integrated encyclopedia of DNA elements in the human genome , 2012 .

[59]  D. G. MacArthur,et al.  Guidelines for investigating causality of sequence variants in human disease , 2014, Nature.

[60]  A. Hamilton,et al.  Improved northern blot method for enhanced detection of small RNA , 2008, Nature Protocols.

[61]  S. Walker,et al.  Quantitative RT-PCR : Pitfalls and Potential , 1999 .