Profiling chromatin regulatory landscape: insights into the development of ChIP-seq and ATAC-seq

Chromatin regulatory landscape plays a critical role in many disease processes and embryo development. Epigenome sequencing technologies such as chromatin immunoprecipitation sequencing (ChIP-seq) and assay for transposase-accessible chromatin with high-throughput sequencing (ATAC-seq) have enabled us to dissect the pan-genomic regulatory landscape of cells and tissues in both time and space dimensions by detecting specific chromatin state and its corresponding transcription factors. Pioneered by the advancement of chromatin immunoprecipitation-chip (ChIP-chip) technology, abundant epigenome profiling technologies have become available such as ChIP-seq, DNase I hypersensitive site sequencing (DNase-seq), ATAC-seq and so on. The advent of single-cell sequencing has revolutionized the next-generation sequencing, applications in single-cell epigenetics are enriched rapidly. Epigenome sequencing technologies have evolved from low-throughput to high-throughput and from bulk sample to the single-cell scope, which unprecedentedly benefits scientists to interpret life from different angles. In this review, after briefly introducing the background knowledge of epigenome biology, we discuss the development of epigenome sequencing technologies, especially ChIP-seq & ATAC-seq and their current applications in scientific research. Finally, we provide insights into future applications and challenges.

[1]  D. Ward,et al.  Immunological method for mapping genes on Drosophila polytene chromosomes. , 1982, Proceedings of the National Academy of Sciences of the United States of America.

[2]  W. Reznikoff,et al.  Tn5 in Vitro Transposition* , 1998, The Journal of Biological Chemistry.

[3]  C. Allis,et al.  Translating the Histone Code , 2001, Science.

[4]  Hilde van der Togt,et al.  Publisher's Note , 2003, J. Netw. Comput. Appl..

[5]  B. Turner,et al.  Immunoprecipitation of native chromatin: NChIP. , 2003, Methods.

[6]  W. Reznikoff Tn5 as a model for understanding DNA transposition , 2003, Molecular microbiology.

[7]  R. Schreiber,et al.  The three Es of cancer immunoediting. , 2004, Annual review of immunology.

[8]  N. Friedman,et al.  Single-Nucleosome Mapping of Histone Modifications in S. cerevisiae , 2005, PLoS biology.

[9]  G. Blobel,et al.  Histone H3 lysine 9 methylation and HP1gamma are associated with transcription elongation through mammalian chromatin. , 2005, Molecular cell.

[10]  James A. Cuff,et al.  A Bivalent Chromatin Structure Marks Key Developmental Genes in Embryonic Stem Cells , 2006, Cell.

[11]  T. Kouzarides Chromatin Modifications and Their Function , 2007, Cell.

[12]  E. Lander,et al.  The Mammalian Epigenome , 2007, Cell.

[13]  Allen D. Delaney,et al.  Genome-wide profiles of STAT1 DNA association using chromatin immunoprecipitation and massively parallel sequencing , 2007, Nature Methods.

[14]  S. Berger The complex language of chromatin regulation during transcription , 2007, Nature.

[15]  Dustin E. Schones,et al.  High-Resolution Profiling of Histone Methylations in the Human Genome , 2007, Cell.

[16]  A. Mortazavi,et al.  Genome-Wide Mapping of in Vivo Protein-DNA Interactions , 2007, Science.

[17]  Dustin E. Schones,et al.  Dynamic Regulation of Nucleosome Positioning in the Human Genome , 2008, Cell.

[18]  Catalin C. Barbacioru,et al.  mRNA-Seq whole-transcriptome analysis of a single cell , 2009, Nature Methods.

[19]  W. Reik,et al.  Epigenetic dynamics of stem cells and cell lineage commitment: digging Waddington's canal , 2009, Nature Reviews Molecular Cell Biology.

[20]  P. Park ChIP–seq: advantages and challenges of a maturing technology , 2009, Nature Reviews Genetics.

[21]  A. Peters,et al.  Mechanisms of transcriptional repression by histone lysine methylation. , 2009, The International journal of developmental biology.

[22]  R. Young,et al.  Histone H3K27ac separates active from poised enhancers and predicts developmental state , 2010, Proceedings of the National Academy of Sciences.

[23]  Timothy J. Durham,et al.  "Systematic" , 1966, Comput. J..

[24]  K. Zhao,et al.  ChIP-Seq: technical considerations for obtaining high-quality data , 2011, Nature Immunology.

[25]  Timothy J. Durham,et al.  Systematic analysis of chromatin state dynamics in nine human cell types , 2011, Nature.

[26]  E. Birney,et al.  High-resolution genome-wide in vivo footprinting of diverse transcription factors in human cells. , 2011, Genome research.

[27]  Marc D. Perry,et al.  ChIP-seq guidelines and practices of the ENCODE and modENCODE consortia , 2012, Genome research.

[28]  B. Maher ENCODE: The human encyclopaedia , 2012, Nature.

[29]  Daniel J. Gaffney,et al.  Controls of Nucleosome Positioning in the Human Genome , 2012, PLoS genetics.

[30]  Jennifer R. Harris,et al.  Limitations and possibilities of low cell number ChIP-seq , 2012, BMC Genomics.

[31]  B. Ren,et al.  Mapping Human Epigenomes , 2013, Cell.

[32]  Michael Q. Zhang,et al.  Epigenomic Analysis of Multilineage Differentiation of Human Embryonic Stem Cells , 2013, Cell.

[33]  Howard Y. Chang,et al.  Transposition of native chromatin for fast and sensitive epigenomic profiling of open chromatin, DNA-binding proteins and nucleosome position , 2013, Nature Methods.

[34]  Clifford A. Meyer,et al.  Identifying and mitigating bias in next-generation sequencing methods for chromatin biology , 2014, Nature Reviews Genetics.

[35]  High-resolution mapping of transcription factor binding sites on native chromatin , 2013, Nature Methods.

[36]  Åsa K. Björklund,et al.  Tn5 transposase and tagmentation procedures for massively scaled sequencing projects , 2014, Genome research.

[37]  G. Beatty,et al.  Immune Escape Mechanisms as a Guide for Cancer Immunotherapy , 2014, Clinical Cancer Research.

[38]  Mohammad M. Karimi,et al.  An ultra-low-input native ChIP-seq protocol for genome-wide profiling of rare cell populations , 2015, Nature Communications.

[39]  Michael Q. Zhang,et al.  Integrative analysis of haplotype-resolved epigenomes across human tissues , 2015, Nature.

[40]  C. Glass,et al.  Epigenomics: Roadmap for regulation , 2015, Nature.

[41]  Andrew C. Adey,et al.  Multiplex single-cell profiling of chromatin accessibility by combinatorial cellular indexing , 2015, Science.

[42]  Howard Y. Chang,et al.  Single-cell chromatin accessibility reveals principles of regulatory variation , 2015, Nature.

[43]  D. Weitz,et al.  Single-cell ChIP-seq reveals cell subpopulations defined by chromatin state , 2015, Nature Biotechnology.

[44]  Nathan C. Sheffield,et al.  ChIPmentation: fast, robust, low-input ChIP-seq for histones and transcription factors , 2015, Nature Methods.

[45]  Chang Lu,et al.  A microfluidic device for epigenomic profiling using 100 cells , 2015, Nature Methods.

[46]  Single-cell ATAC-seq: strength in numbers , 2015, Genome Biology.

[47]  Michael Q. Zhang,et al.  Integrative analysis of 111 reference human epigenomes , 2015, Nature.

[48]  Wei Xie,et al.  Resetting Epigenetic Memory by Reprogramming of Histone Modifications in Mammals. , 2016, Molecular cell.

[49]  Y. Zhang,et al.  Allelic reprogramming of the histone modification H3K4me3 in early mammalian development , 2016, Nature.

[50]  Howard Y. Chang,et al.  HiChIP: efficient and sensitive analysis of protein-directed genome architecture , 2016, Nature Methods.

[51]  Patrik L. Ståhl,et al.  Visualization and analysis of gene expression in tissue sections by spatial transcriptomics , 2016, Science.

[52]  Howard Y. Chang,et al.  Lineage-specific and single cell chromatin accessibility charts human hematopoiesis and leukemia evolution , 2016, Nature Genetics.

[53]  Christopher D. Scharer,et al.  ATAC-seq on biobanked specimens defines a unique chromatin accessibility structure in naïve SLE B cells , 2016, Scientific Reports.

[54]  J. Wysocka,et al.  Ever-Changing Landscapes: Transcriptional Enhancers in Development and Evolution , 2016, Cell.

[55]  Jun S. Song,et al.  Intratumoral Heterogeneity of the Epigenome. , 2016, Cancer cell.

[56]  Howard Y. Chang,et al.  HiChIP: efficient and sensitive analysis of protein-directed genome architecture , 2016, Nature Methods.

[57]  C. Allis,et al.  The molecular hallmarks of epigenetic control , 2016, Nature Reviews Genetics.

[58]  Wei Xie,et al.  The landscape of accessible chromatin in mammalian preimplantation embryos , 2016, Nature.

[59]  Howard Y. Chang,et al.  ATAC-see reveals the accessible genome by transposase-mediated imaging and sequencing , 2016, Nature Methods.

[60]  E. Fraenkel,et al.  Cell freezing protocol suitable for ATAC-Seq on motor neurons derived from human induced pluripotent stem cells , 2016, Scientific Reports.

[61]  S. Mango,et al.  Pioneer transcription factors, chromatin dynamics, and cell fate control. , 2016, Current opinion in genetics & development.

[62]  W. Reik,et al.  Single-cell epigenomics: powerful new methods for understanding gene regulation and cell identity , 2016, Genome Biology.

[63]  S. Picelli Single-cell RNA-sequencing: The future of genome biology is now , 2017, RNA biology.

[64]  Andrew C. Adey,et al.  Chromatin accessibility dynamics of myogenesis at single cell resolution , 2017, bioRxiv.

[65]  William J. Greenleaf,et al.  chromVAR: Inferring transcription factor-associated accessibility from single-cell epigenomic data , 2017, Nature Methods.

[66]  Jessica M. Rusert,et al.  Extrachromosomal oncogene amplification drives tumor evolution and genetic heterogeneity , 2017, Nature.

[67]  X. Xie,et al.  Single-cell whole-genome analyses by Linear Amplification via Transposon Insertion (LIANTI) , 2017, Science.

[68]  A. Lusis,et al.  Considerations for the design of omics studies , 2017 .

[69]  Nicholas A. Sinnott-Armstrong,et al.  An improved ATAC-seq protocol reduces background and enables interrogation of frozen tissues , 2017, Nature Methods.

[70]  Andrew C. Adey,et al.  Cicero Predicts cis-Regulatory DNA Interactions from Single-Cell Chromatin Accessibility Data. , 2018, Molecular cell.

[71]  S. Teichmann,et al.  Single cell RNA-seq and ATAC-seq analysis of cardiac progenitor cell transition states and lineage settlement , 2018, Nature Communications.

[72]  S. Teichmann,et al.  Exponential scaling of single-cell RNA-seq in the past decade , 2017, Nature Protocols.

[73]  Kyle P. Eagen Principles of Chromosome Architecture Revealed by Hi-C. , 2018, Trends in biochemical sciences.

[74]  D. Dickel,et al.  Single-nucleus analysis of accessible chromatin in developing mouse forebrain reveals cell-type-specific transcriptional regulation , 2018, Nature Neuroscience.

[75]  M. Gerstein,et al.  Transcriptome and epigenome landscape of human cortical development modeled in organoids , 2018, Science.

[76]  Andrew C. Adey,et al.  Joint profiling of chromatin accessibility and gene expression in thousands of single cells , 2018, Science.

[77]  S. Henikoff,et al.  Targeted in situ genome-wide profiling with high efficiency for low cell numbers , 2018, Nature Protocols.

[78]  Xuepeng Wang,et al.  Chromatin analysis in human early development reveals epigenetic transition during ZGA , 2018, Nature.

[79]  Sandy L. Klemm,et al.  High-throughput chromatin accessibility profiling at single-cell resolution , 2018, Nature Communications.

[80]  Hatice S. Kaya-Okur,et al.  CUT&Tag for efficient epigenomic profiling of small samples and single cells , 2019, Nature Communications.

[81]  Howard Y. Chang,et al.  Circular ecDNA promotes accessible chromatin and high oncogene expression , 2019, Nature.

[82]  Aibin He,et al.  CoBATCH for High-Throughput Single-Cell Epigenomic Profiling. , 2019, Molecular cell.

[83]  A Bayesian approach for correcting Tn5 transposition bias in ATAC-seq footprinting , 2019 .

[84]  J. Hadfield,et al.  RNA sequencing: the teenage years , 2019, Nature Reviews Genetics.

[85]  N. Zhang,et al.  Resetting histone modifications during human parental-to-zygotic transition , 2019, Science.

[86]  J. Mallm,et al.  Single-nucleus chromatin accessibility reveals intratumoral epigenetic heterogeneity in IDH1 mutant gliomas , 2019, Acta Neuropathologica Communications.

[87]  Kecheng Zhang,et al.  Epigenomic analysis of gastrulation identifies a unique chromatin state for primed pluripotency , 2019, Nature Genetics.

[88]  Aibin He,et al.  Profiling chromatin states using single-cell itChIP-seq , 2019, Nature Cell Biology.

[89]  Stein Aerts,et al.  cisTopic: cis-regulatory topic modeling on single-cell ATAC-seq data , 2019, Nature Methods.

[90]  T. Takebe,et al.  Organoids by design , 2019, Science.

[91]  Kun Zhang,et al.  High-throughput sequencing of the transcriptome and chromatin accessibility in the same cell , 2019, Nature Biotechnology.

[92]  Albert J. Keung,et al.  Engineering Epigenetic Regulation Using Synthetic Read-Write Modules , 2019, Cell.

[93]  Howard Y. Chang,et al.  Massively parallel single-cell chromatin landscapes of human immune cell development and intratumoral T cell exhaustion , 2019, Nature Biotechnology.

[94]  O. Rando,et al.  Profiling of Pluripotency Factors in Single Cells and Early Embryos , 2019, Cell.

[95]  G. Wahl,et al.  Single-Cell Chromatin Analysis of Mammary Gland Development Reveals Cell-State Transcriptional Regulators and Lineage Relationships , 2019, Cell reports.

[96]  C. Vallot,et al.  High-throughput single-cell ChIP-seq identifies heterogeneity of chromatin states in breast cancer , 2019, Nature Genetics.

[97]  Aibin He,et al.  CoBATCH for high-throughput single-cell epigenomic profiling , 2019, bioRxiv.

[98]  B. Ren,et al.  An ultra high-throughput method for single-cell joint analysis of open chromatin and transcriptome , 2019, Nature Structural & Molecular Biology.

[99]  S. Ramón y. Cajal,et al.  Clinical implications of intratumor heterogeneity: challenges and opportunities , 2020, Journal of Molecular Medicine.

[100]  F. Tang,et al.  Decoding the development of the human hippocampus , 2020, Nature.

[101]  K. Qu,et al.  APEC: an accesson-based method for single-cell chromatin accessibility analysis , 2020, Genome biology.

[102]  Xiaoqun Wang,et al.  Chromatin accessibility analysis reveals regulatory dynamics of developing human retina and hiPSC-derived retinal organoids , 2020, Science Advances.

[103]  Howard Y. Chang,et al.  Chromatin accessibility dynamics in a model of human forebrain development , 2020, Science.

[104]  S. Preissl,et al.  Single-cell multimodal omics: the power of many , 2020, Nature Methods.

[105]  C. Martinez-Jimenez,et al.  Advances of single-cell genomics and epigenomics in human disease: where are we now? , 2020, Mammalian Genome.

[106]  Xiaoyu Li,et al.  Transposase assisted tagmentation of RNA/DNA hybrid duplexes , 2020, bioRxiv.