Single-Cell Multi-omics: An Engine for New Quantitative Models of Gene Regulation.

Cells in a multicellular organism fulfill specific functions by enacting cell-type-specific programs of gene regulation. Single-cell RNA sequencing technologies have provided a transformative view of cell-type-specific gene expression, the output of cell-type-specific gene regulatory programs. This review discusses new single-cell genomic technologies that complement single-cell RNA sequencing by providing additional readouts of cellular state beyond the transcriptome. We highlight regression models as a simple yet powerful approach to relate gene expression to other aspects of cellular state, and in doing so, gain insights into the biochemical mechanisms that are necessary to produce a given gene expression output.

[1]  Myong-Hee Sung,et al.  Transcription factor AP1 potentiates chromatin accessibility and glucocorticoid receptor binding. , 2011, Molecular cell.

[2]  Philip A. Ewels,et al.  Mapping long-range promoter contacts in human cells with high-resolution capture Hi-C , 2015, Nature Genetics.

[3]  S. Tapscott,et al.  A conserved motif N-terminal to the DNA-binding domains of myogenic bHLH transcription factors mediates cooperative DNA binding with pbx-Meis1/Prep1. , 1999, Nucleic acids research.

[4]  G. Sanguinetti,et al.  scNMT-seq enables joint profiling of chromatin accessibility DNA methylation and transcription in single cells , 2018, Nature Communications.

[5]  Simon Zhongyuan Tian,et al.  Long-read ChIA-PET for base-pair-resolution mapping of haplotype-specific chromatin interactions , 2017, Nature Protocols.

[6]  H. Binder,et al.  Multilineage communication regulates human liver bud development from pluripotency , 2017, Nature.

[7]  L. Cai,et al.  In Situ Transcription Profiling of Single Cells Reveals Spatial Organization of Cells in the Mouse Hippocampus , 2016, Neuron.

[8]  Hannah A. Pliner,et al.  Reversed graph embedding resolves complex single-cell trajectories , 2017, Nature Methods.

[9]  Michael Boutros,et al.  The art and design of genetic screens: RNA interference , 2008, Nature Reviews Genetics.

[10]  Li Qian,et al.  SLICER: inferring branched, nonlinear cellular trajectories from single cell RNA-seq data , 2016, Genome Biology.

[11]  Evan Z. Macosko,et al.  Comprehensive Classification of Retinal Bipolar Neurons by Single-Cell Transcriptomics , 2016, Cell.

[12]  Hannah A. Pliner,et al.  The cis-regulatory dynamics of embryonic development at single cell resolution , 2017, Nature.

[13]  David J. Jörg,et al.  Defining murine organogenesis at single cell resolution reveals a role for the leukotriene pathway in regulating blood progenitor formation , 2018, Nature Cell Biology.

[14]  Justin P Sandoval,et al.  Single-cell methylomes identify neuronal subtypes and regulatory elements in mammalian cortex , 2017, Science.

[15]  André F. Rendeiro,et al.  Pooled CRISPR screening with single-cell transcriptome read-out , 2016, bioRxiv.

[16]  Aaron N. Chang,et al.  Combinatorial CRISPR-Cas9 screens for de novo mapping of genetic interactions , 2017, Nature Methods.

[17]  C. Vinson,et al.  C/EBP maintains chromatin accessibility in liver and facilitates glucocorticoid receptor recruitment to steroid response elements , 2013, The EMBO journal.

[18]  I. Goldstein,et al.  Transcription factor assisted loading and enhancer dynamics dictate the hepatic fasting response. , 2017, Genome research.

[19]  Shane J. Neph,et al.  Foxp3 Exploits a Pre-Existent Enhancer Landscape for Regulatory T Cell Lineage Specification , 2012, Cell.

[20]  Madeline A. Lancaster,et al.  Human cerebral organoids recapitulate gene expression programs of fetal neocortex development , 2015, Proceedings of the National Academy of Sciences.

[21]  Sean C. Bendall,et al.  Wishbone identifies bifurcating developmental trajectories from single-cell data , 2016, Nature Biotechnology.

[22]  William Stafford Noble,et al.  Massively multiplex single-cell Hi-C , 2016, Nature Methods.

[23]  Howard Y. Chang,et al.  Enhancer connectome in primary human cells identifies target genes of disease-associated DNA elements , 2017, Nature Genetics.

[24]  Salah Ayoub,et al.  The Drosophila Embryo at Single Cell Transcriptome Resolution , 2017, bioRxiv.

[25]  I. Amit,et al.  Dissecting Immune Circuits by Linking CRISPR-Pooled Screens with Single-Cell RNA-Seq , 2016, Cell.

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

[27]  Cole Trapnell,et al.  On the design of CRISPR-based single cell molecular screens , 2018, Nature Methods.

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

[29]  Fabian J Theis,et al.  Diffusion pseudotime robustly reconstructs lineage branching , 2016, Nature Methods.

[30]  C. Glass,et al.  Simple combinations of lineage-determining transcription factors prime cis-regulatory elements required for macrophage and B cell identities. , 2010, Molecular cell.

[31]  I. Amit,et al.  Single-cell spatial reconstruction reveals global division of labor in the mammalian liver , 2016, Nature.

[32]  Timothy K Lu,et al.  Multiplexed barcoded CRISPR-Cas9 screening enabled by CombiGEM , 2016, Proceedings of the National Academy of Sciences.

[33]  Andrew C. Adey,et al.  Single-Cell Transcriptional Profiling of a Multicellular Organism , 2017 .

[34]  Nicola K. Wilson,et al.  A Two-Step, PU.1-Dependent Mechanism for Developmentally Regulated Chromatin Remodeling and Transcription of the c-fms Gene , 2006, Molecular and Cellular Biology.

[35]  Omri Wurtzel,et al.  Cell type transcriptome atlas for the planarian Schmidtea mediterranea , 2018, Science.

[36]  Morteza Mohammad Noori,et al.  Enhanced Regulatory Sequence Prediction Using Gapped k-mer Features , 2014, PLoS Comput. Biol..

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

[38]  P. Kharchenko,et al.  Integrative single-cell analysis of transcriptional and epigenetic states in the human adult brain , 2017, Nature Biotechnology.

[39]  Nikolaus Rajewsky,et al.  The Drosophila embryo at single-cell transcriptome resolution , 2017, Science.

[40]  O. Troyanskaya,et al.  Predicting effects of noncoding variants with deep learning–based sequence model , 2015, Nature Methods.

[41]  Sharon R Grossman,et al.  Systematic mapping of functional enhancer–promoter connections with CRISPR interference , 2016, Science.

[42]  Ilya M. Flyamer,et al.  Single-nucleus Hi-C reveals unique chromatin reorganization at oocyte-to-zygote transition , 2017, Nature.

[43]  Vanessa M. Peterson,et al.  Multiplexed quantification of proteins and transcripts in single cells , 2017, Nature Biotechnology.

[44]  Erik Sundström,et al.  RNA velocity of single cells , 2018, Nature.

[45]  Peter Wernet,et al.  Small molecules enable highly efficient neuronal conversion of human fibroblasts , 2012, Nature Methods.

[46]  Allon M. Klein,et al.  The dynamics of gene expression in vertebrate embryogenesis at single-cell resolution , 2018, Science.

[47]  S. Tapscott,et al.  MyoD Targets Chromatin Remodeling Complexes to the Myogenin Locus Prior to Forming a Stable DNA-Bound Complex , 2005, Molecular and Cellular Biology.

[48]  Martin J. Aryee,et al.  Integrated Single-Cell Analysis Maps the Continuous Regulatory Landscape of Human Hematopoietic Differentiation , 2018, Cell.

[49]  Jonathan M. Cairns,et al.  Lineage-Specific Genome Architecture Links Enhancers and Non-coding Disease Variants to Target Gene Promoters , 2016, Cell.

[50]  Rainer Spang,et al.  Non-transcriptional pathway features reconstructed from secondary effects of RNA interference , 2005, Bioinform..

[51]  W. Sung,et al.  Chromatin connectivity maps reveal dynamic promoter–enhancer long-range associations , 2013, Nature.

[52]  Michael T. McManus,et al.  Dual gene activation and knockout screen reveals directional dependencies in genetic networks , 2018, Nature Biotechnology.

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

[54]  X. Zhuang,et al.  Spatially resolved, highly multiplexed RNA profiling in single cells , 2015, Science.

[55]  Samuel L. Wolock,et al.  A Single-Cell Transcriptomic Map of the Human and Mouse Pancreas Reveals Inter- and Intra-cell Population Structure. , 2016, Cell systems.

[56]  Andrew C. Adey,et al.  Highly scalable generation of DNA methylation profiles in single cells , 2018, Nature Biotechnology.

[57]  Evan Z. Macosko,et al.  A Molecular Census of Arcuate Hypothalamus and Median Eminence Cell Types , 2017, Nature Neuroscience.

[58]  Kendall R. Sanson,et al.  Orthologous CRISPR-Cas9 enzymes for Combinatorial Genetic Screens , 2017, Nature Biotechnology.

[59]  Max A. Horlbeck,et al.  Compact and highly active next-generation libraries for CRISPR-mediated gene repression and activation , 2016, eLife.

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

[61]  S. Quake,et al.  Transcriptomic characterization of 20 organs and tissues from mouse at single cell resolution creates a Tabula Muris , 2017, bioRxiv.

[62]  A. McKenna,et al.  CRISPR/Cas9-Mediated Scanning for Regulatory Elements Required for HPRT1 Expression via Thousands of Large, Programmed Genomic Deletions. , 2017, American journal of human genetics.

[63]  Patrick Flick,et al.  GOATOOLS: Tools for Gene Ontology , 2015 .

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

[65]  André F. Rendeiro,et al.  Pooled CRISPR screening with single-cell transcriptome read-out , 2017, Nature Methods.

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

[67]  Benjamin J. Strober,et al.  A method to predict the impact of regulatory variants from DNA sequence , 2015, Nature Genetics.

[68]  G. Crawford,et al.  DNase-seq: a high-resolution technique for mapping active gene regulatory elements across the genome from mammalian cells. , 2010, Cold Spring Harbor protocols.

[69]  Pang Wei Koh,et al.  Mapping the Pairwise Choices Leading from Pluripotency to Human Bone, Heart, and Other Mesoderm Cell Types , 2016, Cell.

[70]  Hongkui Deng,et al.  Direct lineage reprogramming: strategies, mechanisms, and applications. , 2015, Cell stem cell.

[71]  A. Regev,et al.  Single-cell reconstruction of developmental trajectories during zebrafish embryogenesis , 2018, Science.

[72]  Thomas Lengauer,et al.  DNA Methylation Dynamics of Human Hematopoietic Stem Cell Differentiation , 2016, Cell stem cell.

[73]  David R. Kelley,et al.  Basset: learning the regulatory code of the accessible genome with deep convolutional neural networks , 2015, bioRxiv.

[74]  Kun Zhang,et al.  Characterization of chromatin accessibility with a transposome hypersensitive sites sequencing (THS-seq) assay , 2016, Genome Biology.

[75]  Daniel S. Kim,et al.  Lineage-specific dynamic and pre-established enhancer–promoter contacts cooperate in terminal differentiation , 2017, Nature Genetics.

[76]  W. Reik,et al.  Genome-wide base-resolution mapping of DNA methylation in single cells using single-cell bisulfite sequencing (scBS-seq) , 2017, Nature Protocols.

[77]  Sean C. Bendall,et al.  Single-Cell Trajectory Detection Uncovers Progression and Regulatory Coordination in Human B Cell Development , 2014, Cell.

[78]  Hans Clevers,et al.  Modeling Development and Disease with Organoids , 2016, Cell.

[79]  Joshua C. Chang,et al.  Small Molecules Enable Neurogenin 2 to Efficiently Convert Human Fibroblasts to Cholinergic Neurons , 2013, Nature Communications.

[80]  O. Stegle,et al.  Single-Cell Genome-Wide Bisulfite Sequencing for Assessing Epigenetic Heterogeneity , 2014, Nature Methods.

[81]  I. Goldstein,et al.  Steroid Receptors Reprogram FoxA1 Occupancy through Dynamic Chromatin Transitions , 2016, Cell.

[82]  Mingyao Li,et al.  Single-cell transcriptomics of the mouse kidney reveals potential cellular targets of kidney disease , 2018, Science.

[83]  Yaniv Lubling,et al.  Single-cell Hi-C for genome-wide detection of chromatin interactions that occur simultaneously in a single cell , 2015, Nature Protocols.

[84]  G. Nolan,et al.  Mass Cytometry: Single Cells, Many Features , 2016, Cell.

[85]  G. Hon,et al.  Multiplexed Engineering and Analysis of Combinatorial Enhancer Activity in Single Cells. , 2017, Molecular cell.

[86]  R. Agami,et al.  Applying CRISPR–Cas9 tools to identify and characterize transcriptional enhancers , 2016, Nature Reviews Molecular Cell Biology.

[87]  Yutaka Suzuki,et al.  Long-term expansion of alveolar stem cells derived from human iPS cells in organoids , 2017, Nature Methods.

[88]  P. Rigollet,et al.  Reconstruction of developmental landscapes by optimal-transport analysis of single-cell gene expression sheds light on cellular reprogramming , 2017, bioRxiv.

[89]  Yarden Katz,et al.  A single-cell survey of the small intestinal epithelium , 2017, Nature.

[90]  Thomas M. Norman,et al.  Perturb-Seq: Dissecting Molecular Circuits with Scalable Single-Cell RNA Profiling of Pooled Genetic Screens , 2016, Cell.

[91]  J. Carroll,et al.  Pioneer transcription factors: establishing competence for gene expression. , 2011, Genes & development.

[92]  C. A. Albers,et al.  Immuno-Detection by sequencing (ID-seq) enables large-scale high-dimensional phenotyping in cells , 2017, bioRxiv.

[93]  Michael B. Elowitz,et al.  Combinatorial Signal Perception in the BMP Pathway , 2017, Cell.

[94]  Thomas M. Norman,et al.  A Multiplexed Single-Cell CRISPR Screening Platform Enables Systematic Dissection of the Unfolded Protein Response , 2016, Cell.

[95]  S. Orkin,et al.  Mapping the Mouse Cell Atlas by Microwell-Seq , 2018, Cell.

[96]  Sean C. Bendall,et al.  Conditional density-based analysis of T cell signaling in single-cell data , 2014, Science.

[97]  Eliah G. Overbey,et al.  Aligning single-cell developmental and reprogramming trajectories identifies molecular determinants of reprogramming outcome , 2017, bioRxiv.

[98]  Madeline A. Lancaster,et al.  Dishing out mini-brains: Current progress and future prospects in brain organoid research , 2016, Developmental biology.

[99]  J. Stamatoyannopoulos,et al.  Chromatin accessibility pre-determines glucocorticoid receptor binding patterns , 2011, Nature Genetics.

[100]  S. Henikoff,et al.  An efficient targeted nuclease strategy for high-resolution mapping of DNA binding sites , 2016, bioRxiv.

[101]  B. Frey,et al.  Predicting the sequence specificities of DNA- and RNA-binding proteins by deep learning , 2015, Nature Biotechnology.

[102]  Evan Z. Macosko,et al.  Highly Parallel Genome-wide Expression Profiling of Individual Cells Using Nanoliter Droplets , 2015, Cell.

[103]  Neville E. Sanjana,et al.  Genome-Scale CRISPR-Cas9 Knockout Screening in Human Cells , 2014, Science.

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

[105]  Ty C. Voss,et al.  Dynamic Exchange at Regulatory Elements during Chromatin Remodeling Underlies Assisted Loading Mechanism , 2011, Cell.

[106]  Dongwon Lee,et al.  LS-GKM: a new gkm-SVM for large-scale datasets , 2016, Bioinform..

[107]  Raymond K. Auerbach,et al.  Extensive Promoter-Centered Chromatin Interactions Provide a Topological Basis for Transcription Regulation , 2012, Cell.

[108]  Cole Trapnell,et al.  The dynamics and regulators of cell fate decisions are revealed by pseudotemporal ordering of single cells , 2014, Nature Biotechnology.

[109]  Hans Clevers,et al.  Single-cell messenger RNA sequencing reveals rare intestinal cell types , 2015, Nature.

[110]  Eric Haugen,et al.  Large-scale identification of sequence variants impacting human transcription factor occupancy in vivo , 2015, Nature Genetics.

[111]  A. Regev,et al.  Spatial reconstruction of single-cell gene expression , 2015, Nature Biotechnology.

[112]  Maria Kasper,et al.  Single-Cell Transcriptomics Reveals that Differentiation and Spatial Signatures Shape Epidermal and Hair Follicle Heterogeneity , 2016, Cell systems.

[113]  Max A. Horlbeck,et al.  Genome-Scale CRISPR-Mediated Control of Gene Repression and Activation , 2014, Cell.

[114]  Andre J. Faure,et al.  3D structure of individual mammalian genomes studied by single cell Hi-C , 2017, Nature.

[115]  M. Busslinger,et al.  Stepwise activation of enhancer and promoter regions of the B cell commitment gene Pax5 in early lymphopoiesis. , 2009, Immunity.

[116]  Howard Y. Chang,et al.  Joint single-cell DNA accessibility and protein epitope profiling reveals environmental regulation of epigenomic heterogeneity , 2018, Nature Communications.

[117]  Allon M. Klein,et al.  Single-cell mapping of gene expression landscapes and lineage in the zebrafish embryo , 2018, Science.

[118]  Smita Krishnaswamy,et al.  Learning Edge Rewiring in EMT From Single Cell Data , 2017, bioRxiv.

[119]  A. Tanay,et al.  Single cell Hi-C reveals cell-to-cell variability in chromosome structure , 2013, Nature.

[120]  Elena K. Kandror,et al.  Single-cell topological RNA-Seq analysis reveals insights into cellular differentiation and development , 2017, Nature Biotechnology.

[121]  David A. Orlando,et al.  Master Transcription Factors Determine Cell-Type-Specific Responses to TGF-β Signaling , 2011, Cell.

[122]  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.

[123]  S. Mandrup,et al.  Extensive chromatin remodelling and establishment of transcription factor ‘hotspots’ during early adipogenesis , 2011, The EMBO journal.

[124]  Lars E. Borm,et al.  Molecular Architecture of the Mouse Nervous System , 2018, Cell.

[125]  Aviv Regev,et al.  Massively-parallel single nucleus RNA-seq with DroNc-seq , 2017, Nature Methods.

[126]  S. Tapscott,et al.  Pbx marks genes for activation by MyoD indicating a role for a homeodomain protein in establishing myogenic potential. , 2004, Molecular cell.