Skip to main content
Erschienen in: Clinical and Translational Medicine 1/2017

Open Access 01.12.2017 | Review

Using single-cell multiple omics approaches to resolve tumor heterogeneity

verfasst von: Michael A. Ortega, Olivier Poirion, Xun Zhu, Sijia Huang, Thomas K. Wolfgruber, Robert Sebra, Lana X. Garmire

Erschienen in: Clinical and Translational Medicine | Ausgabe 1/2017

Abstract

It has become increasingly clear that both normal and cancer tissues are composed of heterogeneous populations. Genetic variation can be attributed to the downstream effects of inherited mutations, environmental factors, or inaccurately resolved errors in transcription and replication. When lesions occur in regions that confer a proliferative advantage, it can support clonal expansion, subclonal variation, and neoplastic progression. In this manner, the complex heterogeneous microenvironment of a tumour promotes the likelihood of angiogenesis and metastasis. Recent advances in next-generation sequencing and computational biology have utilized single-cell applications to build deep profiles of individual cells that are otherwise masked in bulk profiling. In addition, the development of new techniques for combining single-cell multi-omic strategies is providing a more precise understanding of factors contributing to cellular identity, function, and growth. Continuing advancements in single-cell technology and computational deconvolution of data will be critical for reconstructing patient specific intra-tumour features and developing more personalized cancer treatments.
Abkürzungen
3C
chromosome conformation capture
BAT-seq
barcoded 3′-end specific sequencing
BWA
Burrows-Wheeler Aligner
ccfDNA
circulating cell-free DNA
CD
cluster of differentiation
CEL-Seq
cell expression by linear amplification and sequencing
CIViC
clinical interpretation of variants in cancer
CNV
copy number variation
CNVkit
copy number variation kit
CTC
circulating tumor cells
ddSeq
droplet digital sequencing
DOP-PCR
degenerate oligonucleotide primed PCR
ECdetect
extrachromosomal DNA
EwS
Ewing sarcoma
GATK
genome analysis tool kit
GUI
graphical user interface
IDH
isocitrate dehydrogenase
iEVORA
epigenetic variable outliers for cancer risk prediction analysis
IFC
integrated microfluidic circuit
InferCNV
infer copy number variation
INTEGRATE-Neo
integrate neoantigens
LIANTI
linear amplification via transposon insertion
MALBAC
multiple annealing and looping based amplification cycles
MARS-seq
massively parallel single-cell RNA-sequencing
MDA
multiple displacement amplification
MS
mass spectrometry
mTORki
mTOR kinase inhibitor
MutSigCV 2.0
mutation significance copy variation
NTRK1
neurotrophic receptor tyrosine kinase 1
OncoNEM
oncogenetic nested effects model
scATAC-seq
single-cell assay for transposase accessible chromatin sequencing
scBS
single-cell bisulfite sequencing
scCGI-seq
CpG island methylation sequencing for single-cell
sciHi-C
high-resolution chromatin conformation capture (3C) assay
SCITE
single-cell inference of tumor evolution
scLVM
single-cell latent variable models
scMAB-seq
single-cell methylase assisted bisulfite sequencing
scMT-seq
single-cell methylome and transcriptome sequencing
SCoPE-MS
single-cell ProtEomics by mass spectrometry
scRNA-seq
single-cell RNA sequencing
scRNASeqDB
single-cell RNA sequencing database
scRRBS
single-cell reduced representation bisulfite sequencing
scWES
single-cell whole exome sequencing
scWGBS-seq
single-cell whole genome bisulfite sequencing
scWGS
single-cell whole genome sequencing
SESN2
sestrin 2
SMART-seq
switching mechanism at the 5′ end of the RNA transcript sequencing
SNP
single-nucleotide polymorphism
SPADE
spanning-tree progression analysis of density-normalized events
SPLiT-Seq
split pool ligation-based transcriptome sequencing
SSrGE
sparse SNV inference to reflect gene expression
STRT-Seq
single-cell tagged reverse transcription
SUPeR-seq
single-cell universal poly(A)-independent RNA sequencing
TAD
topologically associated domains
TCGA
The Cancer Genome Atlas
UMI-seq
unique molecular identifier sequencing
WGA
whole genome amplification

Introduction

DNA serves as the source code for specific mechanisms that regulate cellular identity, function, and growth. The genome is generally replicated with high-fidelity. However, stochastic somatic alterations can occur at an average rate of 3 mutations per cell division in normal cells [1, 2]. These genetic changes can be the effect of inherited mutations, environmental factors, or inaccurately resolved errors in transcription or replication. Mutations typically occur in non-coding regions of the genome and have no immediately apparent effect on the phenotype of the cell [25]. However, as mutations accumulate over time, they increase genetic variations and the likelihood of developing a neoplasm. Communities of mutations, or alterations to driver genes, can lead to increases in proliferation, a higher frequency of errors in transcription and replication, and/or the enabling of apoptotic evasion [6, 7]. Finally, recent studies indicate that metastases may also derive from early disseminated cancer cells [8]. These features are hallmarks of cancer that subsequently facilitate neoplastic progression (Fig. 1) [9].
To better interpret cellular heterogeneity, researchers have developed various high-throughput applications to generate a more comprehensive cellular atlas of the human body. Tang et al. [10] initially reported a single-cell RNA-seq experiment, where only one cell was sequenced in a single run. This cell was manually separated under the microscope. Since then, the technology has improved several times, each time providing a higher cell count and/or expression sensitivity in a single run. Notably, published in 2012, SMART-seq allowed for greater sensitivity and capturing of full-length transcripts, however cells had to be manually picked in that experiment limiting practical cell capture counts. The Fluidigm C1 capture method introduced microfluidic chips for more automated larger scale cell capture that could be paired with effective library preparation technologies. Starting from 2014, a number of emulsion-based protocols including that by 10× Genomics increased this number by another one to two orders of magnitude (Table 1).
Table 1
Notable advancements in single-cell techniques
Year introduced
Notable technology advancements
Method cell rangea
2009
Tang et al. [10]
1b
2011
STRT-seq [23]
< 100
2012
SMART-seq [24]
< 100
2012
CEL-Seq [25]
< 100
2013
Fluidigm C1 (IFC) [26]
< 800
2013
Smart-seq 2 [27]
< 1000
2014
MARS-seq [28]
10,000 s
2015
Drop-seq [29]
10,000 s
2015
inDrop [30]
10,000 s
2016
Chromium (10× Genomics) [31]
10,000 s
2017
ddSeq (Bio-Rad) [32]
10,000 s
2017
SPLiT-seq [33]
10,000 s
2017
Seq-well [34]
10,000 s
This is a non-comprehensive list of peer-reviewed studies that advanced single-cell isolation and preparation techniques
aThe “range” lists the largest relative population that can or has been studied using this technique
bThis method involves mechanical separation and isolation of individual blastomeres into single wells
Catching up with the advances in the technology, methods to investigate complex populations are only now coming to fruition with single-cell precision. For example, bulk high-throughput sequencing has been previously used to reveal that intra-tumour genetic and epigenetic heterogeneity progress through sub-clonal branched evolution rather than through linear expansion (Fig. 2) [11, 12]. However, for similar studies, single-cell tools for phylogenetic reconstruction of clonal evolution are more complicated due to lower coverage than bulk samples [1316]. Characterizing the branched sub-clonal evolution of a neoplasm is critical for identifying key sub-population driver mutations promoting diversification, expansion, invasion, and eventually colonization to other parts of the body. In addition, the aggregated effect of tumour heterogeneity is important to resolve because resistance in one or more clonal subsets of a global tumour cellular population can impact chemotherapeutic efficacy (Fig. 2) [17]. In fact, chemotherapies have a modest overall median survival benefit of 2.1 months while costing around $100,000/year in the U.S. [18, 19]. One option to mitigate this inefficiency is to remodel patient specific intra-tumour heterogeneity computationally using single-cell genomics data and determine functional pathways at a high resolution [2022]. While the circulating tumor cells provide an opportunity to directly profile the difference when comparing to the primary tumor samples retrospectively, investigating different tumor subgroups allows one to reconstruct the evolution of single tumor cells relative to each other in a more continuous fashion. To assemble a comprehensive cellular map of the body, accurate and reproducible experimental protocols, and computational analysis pipelines will be critical to extract and interpret heterogeneous information. Here, we provide a review of current single-cell genomics strategies developed for investigating cellular heterogeneity.

Single-cell partitioning

The standard workflow of single-cell investigations includes dissociating a bulk-cell sample into individual cells, isolating those cells, preparing them for the desired application, acquiring data, and analyzing data. Today, methods for separating cells still include mechanical perturbation or enzymatic digestion to separate bulk samples into single cells. However, the downstream methods for isolation, preparation, data generation, and analysis have made rapid advancements. Partitioning bulk samples and isolating individual cells can be technically challenging and necessitates optimization, often on a tissue-specific level. This initial step for investigating heterogeneity through single-cell applications can be complicated by the inefficient separation, which results in higher doublet capture rates in fluidics and droplet-based technologies. Caution should also be taken not to induce unnecessary mechanical or chemical stress on the cell during this process. Performing a clean isolation will also avoid unnecessary molecular debris that can impact the ability to assign individual unique molecular identifiers (UMIs) to single cells during the process of demultiplexing on the cellular and transcript/allele levels.
After separating and suspending the population of tumour cells from biopsy tissue, single cells can either be processed in bulk or sorted and enriched to select specific sub-populations. Most commonly, single cells can be isolated by flow-cytometry, laser capture microdissection, serial dilution, using antibody-coated magnetic beads, or microfluidic-based techniques. Droplet-based technologies such as Drop-seq, inDrop, Chromium, and ddSeq can produce tens to hundreds of thousands of uniquely barcoded cells (Table 1) [28, 35, 36]. The droplet-based approach for isolating and preparing single cells involves using bead-based surface chemistry to facilitate molecular sample preparation methods while encapsulating the cell in an emulsion or aqueous microfluidic partition (Fig. 3a). Each bead contains DNA fragments with unique barcode sequences that are incorporated with cell material during encapsulation. While encapsulated, RNA is also reverse transcribed. The emulsions are then broken prior to pooled amplification and sequencing. Integrated microfluidic circuit (IFC) chips offer an alternative approach to isolate and process cells individually by capturing them in small chambers [37]. IFC protocols have a natural quality-control step whereby doublets can be recorded by microscopy visualization before preparing downstream applications (Fig. 3b).
Costs should be considered when deciding the satisfying number of cells to run relevant to the depth of heterogeneity in the tissue being interrogated. Isolating cell populations through a chip limits the number of cells that can be captured in a single run. It can be more expensive than droplet-based approaches (~ $3.5 versus $0.10 per cell) [38]. In addition to financial differences, these recent approaches and their derivatives each have unique technical concerns and limitations that have been reported in Svensson et al. [39]. After isolating the single-cell population of interest, the subsequent preparation steps vary widely depending on the desired application. Below, we elaborate on exemplary applications for studying single-cell heterogeneity.

Single-cell whole genome and whole exome sequencing

Fundamental technical concerns exist with generating and analyzing single-cell genetic, epigenetic, and expression data, because of the low amount of starting material. Single cells have ~ 6 pg of genomic DNA, 10–30 pg of total RNA that must be amplified before sequencing, and roughly 250–300 pg of protein that can be analyzed. A battery of computational models have been developed to address false-positives due to nonlinear amplification, false-negative allelic drop-out due to amplification bias, non-uniform coverage, and noise that arises during single-cell genome or transcript amplification (Table 2) [40]. For this reason, bioinformatics and computational biology applications are critical for analyzing NGS output files and accurately identifying genetic variation. Single-cell whole-genome and whole-exome sequencing (scWGS and scWES, respectively) provide amplified genomic DNA variant datasets that can then be used to reconstruct clonal evolution or to measure genetic heterogeneity.
Table 2
Tools for investigating heterogeneity
Name
Description
Link
Input
Specific to single-cell
References
Accessibility
Databases
 scRNASeqDB
A database for gene expression profiling in human single cell by RNA-seq
N/A
Yes
Cao et al. [62]
***
 The Human Protein Atlas
Spatial distribution of protein expressions
Protein name
No
Uhlén et al. [108, 109]  
*****
 Enrichr
Very complete meta-database
List of genes/proteins—BED file
No
Kuleshov et al. [110]
*****
 CIViC
Clinical interpretation of cancer variant
Gene or variant ID
No
Griffith et al. [111]
****
 MyGene2
A portal for sharing health and genetic information
Genetic information
No
Xin et al. [112]
****
Genome sequencing
 SCITE
Pseudo-temporal clonal tree construction
Presence/absence/unknown mutation matrix
Yes
Jhan et al. [113]
*
 oncoNEM
Pseudo-temporal clonal tree construction
Binary matrix + estimation of FPR and FNR for each SNVS
Yes
Ross and Markowetz [114]
*
 BWA
DNA reads aligner
fastq file + reference genome
No
Li [115]
*
Methylation
 Bismark
Aligner for bisulfite treated sequencing reads
fastq files
No
Krueger and Andrews [83]
**
RNA-seq
 Granatum
Graphical pipeline for scRNA-seq analysis
Expression matrix and sample metadata
Yes
Zhu et al. [56]
*****
 Monocle2
Pseudo-time construction using DDRTree
Expression matrix and sample metadata
Yes
Trapnell et al. [57]
*
 scLVM
Subpopulation detection
Expression matrix
Yes
Buettner et al. [58]
*
 PseudoGP
Probabilistic pseudotime for single-cell RNA-seq data
Expression matrix
Yes
Campbell et al. [116]
*
 SPADE
Cell hierarchy inference
Expression matrix
Yes
Anchang et al. [60]
*
 STAR
RNA-seq reads aligner
Fast
No
Dobin et al. [117]
**
CNV
 InferCNV
Average gene expression on large genomic regions
Gene expression matrix
Yes
Patel et al. [63]
**
 ECdetect
Detection of extrachromosomal DNA
Bam file
Yes
Turner et al. [118]
*
 CNVkit
Detection of CNV from DNA sequencing
BAM file + target regions (BED files)
No
Talevich et al. [119]
**
 SynthEx
Detection of copy number alteration and tumour heterogeneity profiling for whole genome and exome sequencing
Count data (bed files) + optional vcf files for tumor samples
No
Silva et al. [120]
**
 Ginkgo
Web platform for visualization and clustering
Bed files
Yes
 
*****
 MutSigCV 2.0
Eliminate false positive mutations in large datasets
Mutations for each sample + sequencing coverage
No
Lawrence et al. [121]
**
 HotNet2
Identify mutated subnetworks across pathways and protein complexes
Mutation data + protein–protein interaction network
No
Leiserson et al. [122]
*
Proteomics
 Wishbone
Reconstructing bifurcating developmental trajectories of single-cells
tsv expression files
Yes
Setty et al. [123]
***
Multi-omics
 DeepCpG
Infer missing methylation states and expressive DNA motifs linked to methylation
Methylation position file + ref genomes + fastq files
Yes
Angermueller et al. [87]
*
 SSrGE
Link gene expression with SNVs. Provide a pipeline to extract SNVs from scRNA-seq
Expression matrix + binary matrix; fastq files
Yes
Poirion et al. [106]
*
Genetic architecture
 combinatorialHiC
Processing single cell combinatorial indexed Hi-C
fastq files + barcodes
Yes
Ramani et al. [124]
*
Others
 Integrate-neo
Gene fusion neoantigen discovering tool
fastq files (or tsv) + bedpe files + reference genomics
No
Zhang et al. [125]
*
 awesome-single-cell
Exhaustive community-driven list of single-cell analytical tools
N/A
Yes
N/A
*****
Software, computational packages, and databases mentioned in the paper
The accessibility of a tool is our evaluation of its user-friendliness towards bench scientists who are not necessarily computationally trained
The accessibility ranges from “*” (least accessible) to “*****” (most accessible)

Experimental methods

To date, three primary techniques are used for whole genome amplification (WGA) which include: (1) PCR-based methods such as multiple annealing and looping-based amplification cycles (MALBAC), (2) degenerate oligonucleotide-primed polymerase chain reaction (DOP-PCR); and (3) non-PCR based method using random hexamers or non-specific priming like multiple displacement amplification (MDA) [4143]. Each of the three primary techniques becomes unreliable when there is less than 1.5 pg of genomic material. The smaller starting volumes are susceptible to environmental contamination and require optimal sterile working conditions to avoid the creation of false positives [44]. Each method used for single-cell genomic DNA amplification has biases to consider and can be affected by poor coverage or uneven sequencing depth, which will result in noisy and inaccurate read counts.
MALBAC tends to have GC bias, but reports find that MALBAC libraries are more reproducible than those generated with MDA [45]. Some analytic tools, such as the Ginkgo web platform (http://​qb.​cshl.​edu/​ginkgo), take measures to reduce or eliminate these amplification biases. According to the authors of Ginkgo, of the three genetic amplification techniques used, MDA has non-uniform coverage and worse GC bias than either DOP-PCR or MALBAC. Reports suggest that despite moderate physical coverage DOP-PCR is the most reliable method [42, 46]. While this Review was being prepared, an approach that uses linear amplification via transposon insertion (LIANTI) was reported that may prove to have less amplification bias and high (97%) genome coverage [47].

Exemplary applications

With new isolation, amplification, and computational modelling techniques becoming available there has been a general progression toward increased populations of cells or higher quality coverage in rare populations. In 2012, scWGA by MDA of 58 cells was used to demonstrate mutational contributions of SESN2 and NTRK1 in neoplasm progression. In this study, more than 90% of the single-cell genomic data mapped back to the reference genome and they had an 11% allele dropout rate [48]. In another early example, Xu et al. performed scWES-seq of 25 single cells by MDA amplification which was used to reconstruct clonal mutations occurring within PBRM1 and VHL in kidney cancer. Here, the MDA approach yielded a false positive rate of 2.67 × 10−5 and an allele dropout rate of 16.43% [49]. A more recent study by Gao et al. used DOP-PCR to amplify genomic content from 1000 cells and reported that the majority of subclonal CNV occurs shortly after the onset of the primary driver mutation in breast cancer. While the false discovery rate was not mentioned in this article, their data suggests that 20–40 single cells were required for detecting subpopulations with 95% power [50]. It is clear that the number of cells being reported in a single study has changed rapidly. However, while the available number of cells per analysis has been used to reach up to nearly 1500 cells (using MDA), the average number of reads per cell decreases as the number of cells increase. Thus, much like scRNA-seq, there is a tradeoff between the individual cell quality and the total number of cells analyzed within a batch [39]. The shortcoming of large batches emphasizes the need for computational tools that can correct such bias (Table 2).

Single-cell RNA sequencing

The most broadly developed method for single-cell analysis is single-cell RNA sequencing (scRNA-seq). High-throughput RNA-seq of bulk samples from scores of patients have provided novel insights into many cancer types [5153]. However, deploying RNA-seq analysis at single-cell resolution can provide an even deeper level of understanding the heterogeneous composition of tumour samples by identifying constituents otherwise masked in bulk RNA-seq [54, 55]. The first scRNA-seq was performed in 2009 using single mouse embryo blastomeres [10]. Since then, there has been an increasing surge of sc-RNA-seq publications. Compared to bulk level RNA-seq, scRNA-seq has at least two advantages. First, a more accurate and sensitive presentation of the cell-to-cell variability can be discerned. Second, these data can be reorganized into pseudo-temporal arrangements that can more accurately reconstruct clonal evolution. Beyond experimental advantages, monitoring the dynamics of sub-clonal populations across the course of treatment also has the potential to inform and allow more precise adjustment of therapies.

Experimental and computational methods

There are several platforms available for isolating and preparing RNA from single cells. One of the greatest technical concerns is in amplifying these low concentrations without introducing significant bias. Data generated by droplet-based approaches permit 10,000 s of cells to be counted, while other platforms that use chip-based systems process only 100 s of cells at a time but are more sensitive to calling the number of genes per cell [39]. After successful isolation of single cells, there are a wide number of molecular approaches to creating scRNA-seq libraries such as SMART-seq, SUPeR-seq, BAT-seq, CEL-seq, and STRT-seq amongst others. The SMART-seq approach can generate full-length cDNA, whereas approaches like STRT-seq (targeting 5′ end of mRNA) and CEL-seq (targeting 3′ end of mRNA) can be used for multiplexing samples [23, 25, 27]. SMART-seq employs a special reverse-transcriptase that anchors both ends of cDNA with distinct nucleotides. The absence of one of these ends eliminates incompletely reverse-transcribed RNA molecules after a subsequent cloning step. It is important to note that sensitivity and accuracy become concerns when the experiments scale up to larger numbers. Here, sensitivity is defined as the smallest quantity of input RNA molecules required for a gene to be confidently called. Accuracy is defined here as the closeness between the estimated and the actual abundance levels of input molecules. Whereas studies have shown that the droplet-based microfluidic approaches permit greater numbers of cells to be counted, chip-based systems appear to be more sensitive [39].
Computationally, many tools have been tailored to take advantage of the high-resolution of scRNA-seq data and deconvolute noise [56]. For example, Monocle2 is an unsupervised algorithm designed to analyze the heterogeneity among cells and reconstruct the micro-evolution timeline from scRNA-seq data [57]. Other tools such as scLVM [58], PseudoGP [59], and SPADE [60] have provided various solutions to analyze heterogeneity with scRNA-seq data computationally. With the scRNA-seq analysis toolbox expanding rapidly, graphical user interface (GUI) pipelines such as Granatum (http://​garmiregroup.​org/​granatum/​app) have recently been developed to ensure that accessing the latest development in computational methods is amenable for clinical and non-informatics researchers [61]. In addition, with datasets accumulating at an astonishing speed, there have been efforts like the RIKEN Single-Cell Project (http://​singlecell.​riken.​jp/​en/​) to consolidate, index, and organize publically available datasets [62].

Exemplary applications

scRNA-seq has been used broadly to provide data on genetic expression and has now been widely applied to a variety of cancer types. Since there are many more techniques developed and reported for scRNA-seq, we will only highlight a few applications as examples of how scRNA-seq is impacting the discussion on heterogeneity. In one example, the SMART-seq protocol was used to profile full-length mRNA from 430 primary glioblastomas to reveal an intratumor spectrum of differentiation states [63]. The SMART-seq protocol was later improved to increase mRNA yield, coverage, sensitivity, accuracy and reintroduced as Smart-seq 2 [27]. Smart-seq 2 is now a widely used approach in scRNA-seq. In one case, it was used to profile 4347 cells from six oligodendrogliomas and revealed subgroups of undifferentiated cells with a stem-cell-like expression that may be the source of oligodendrogliomas [54]. These data highlight that one benefit of performing expression analysis at single-cell resolution is it can reveal subpopulations otherwise masked in bulk data. In addition, enhancing sensitivity for clonal-level therapies alongside offers the potential for discovering novel, previously undetectable biomarkers on an individual level.
Currently, one of the most interesting shifts in research to recognize is that scRNA-seq is progressing to include a broader range of samples in addition to a deeper pool from a single source. Thus, as the cost of performing scRNA-seq continues to drop, it will facilitate simultaneous inter- and intra-tumour investigations. For example, a recent report using 9879 cells from 10 IDH-A tumours and 4347 cells from six IDH-O tumours were compared with 165 TCGA bulk samples to identify a common progenitor for IDH mutant gliomas [64]. This report is representative of the growing trend to combine available bulk data with single-cell data cohorts for broader and deeper data mining potential. In addition to profiling tissue samples, scRNA-seq is also used to investigate circulating tumour cells (CTC), which is particularly valuable for prospective monitoring [65]. The SMART-seq method was initially reported in an investigation on CTCs and was proposed as a method for identifying candidate cancer biomarkers [24]. Isolating and analyzing expression in CTCs alongside monitoring circulating cell-free DNA (ccfDNA) burden has high direct translational potential for identifying personalized biomarker panels to guide treatment in real-time.

Single-cell chromosome conformation capture

Developments in studying single-cell genomic architecture have become increasingly deployed to understand the relationship between topology and phenotype. Topology is interesting because single nucleotide variations (SNV), point mutations, and insertions or deletions (indels) can indirectly impact the expression of a distant gene by rearranging the genetic architecture [66]. Since the expression of some genes is affected by long-distance interactors, another level of heterogeneity to consider is the arrangement and proximity of chromosome territories within the nucleus [67]. Genomic architecture is described as being organized in A/B compartments, topologically associated domains (TADs), and loops. Perturbation at any level of these structures can have a significant role in determining domain accessibility which can either improve or inhibit activity in that region. These physical genomic folding structures have been revealed using long-range genomic interaction maps derived from high-throughput sequencing data [68]. When integrated with other techniques such as scWES or single-cell RNA-seq, Hi-C provides an informative tool for identifying the relationship between the 3D architecture of the genome and gene expression [69].

Experimental methods

To analyze single-cell nuclear DNA structure, high-throughput sequencing is coupled with a high-resolution chromatin conformation capture (3C) assay (sciHi-C) [70]. The chromatin architecture of single cells is reconstructed by generating short and long-range interaction maps. Briefly, interaction is inferred by fixing the DNA, followed by enzymatic restriction digestion, adaptor ligation, and proximity ligation. This sequence of steps allows interacting loops and TADs to be ligated together and will thereby yield a higher number of reads due to more frequent interaction [71]. Pipelines for analyzing multiplexed scHi-C data have recently become open-sourced [71]. On a more local scale, single-cell ATAC-seq (scATAC-seq) is used to profile open (transcription-permissive) chromatin. During scATAC-seq, isolated nuclei are processed by Tn5 tagmentation, which inserts adapters into nucleosome-free regions [72, 73].

Exemplary applications

scHi-C methods give detailed information about the state of the chromatin accessibility and long-range interactions. These methods are currently being adopted to define how architecture evolves throughout the cell-cycle at single-cell resolution [74]. A recent publication used scHi-C to reveal the architecture during pronuclear formation of G1 zygotes. In the zygote, maternal and paternal pronuclei have different levels of organization with maternal DNA lacking A/B compartments. This suggests that organization of paternal compartmentalization is likely inherited from the sperm [75]. The organization of loops, TADs, and compartments is significant because it infers which regions are more active and perhaps more prone to mutations [7]. On a more localized level, a separate study used a modified sciATAC-SEQ approach called SCI-seq was demonstrated on 16,000 single cells from different cancer types. SCI-seq uses a lithium-assisted nucleosome depletions strategy to remove histones followed by cross-linking than by the scATAC-seq protocol [76]. Together, these techniques provide information on the organization of DNA in the nucleus.

Single-cell epigenetics

Epigenetic diversity involves heritable changes that affect genomic expression but that do not affect the DNA information. This includes direct modification of nucleic acids (i.e. 5mC, 6mA, m6A, and pseudouridine), and post-translational modification of histones (e.g. methylation, and acetylation) [77, 78]. DNA methylation adds another layer of complexity to our understanding of how heterogeneity affects cellular identity and function. Hypermethylation of DNA is associated with transcriptional repression, while the reverse is true for hypomethylation. Single-cell epigenetics studies have advanced more rapidly than those that deal with proteins. Elucidating epigenetic heterogeneity at the single-cell level add a deeper understanding of how methylation patterns are maintained on a clonal level across cell populations and across individuals. However, it should be noted that the most significant roadblock to generating single-cell resolution methylation data is that current techniques are harsh and result in massive loss of DNA template. Also, these techniques often include amplification methods that remove the ability to detect epigenetic modifications.

Experimental methods and applications

Amongst the experimental approaches inferring cytosine modification, single-cell bisulfite sequencing (scBS) is the most widely used technique. scBS-seq preferentially de-aminates unmethylated cytosine thereby converting unmethylated cytosines to thymines. However, during this bisulfite treatment step, nicks and fragmentation in the DNA occur that reduce the quality and quantity of the input. This is followed by primary and secondary adapter ligation and PCR [7981]. Single-cell reduced representation bisulfite sequencing (scRRBS) has lower coverage of total CpG sites but higher coverage of CpG islands [82]. Aligned reads generated from this technique require special tools such as Bismark for read mapping and methylation calling [83]. A third technique known as single-cell whole-genome bisulfite sequencing (scWGBS-seq) was developed that does not include the pre-amplification step that takes place in scBS-seq but this approach has lower coverage complexity [84]. Comparatively, the scRRBS method only covers 1% CpG sites across the genome, in contrast to 48.4% of CpG sites by scWGBS. Single-cell methylase assisted bisulfite sequencing (scMAB-seq) and CpG island methylation sequencing for single-cell (scCGI-seq) have also recently been proposed [85, 86]. Future comparative studies of these various methods will assist in determining the differences in mapping and cost efficiency.
Cytosine methylation studies on cancer samples at the single-cell level currently lag behind other -omics approaches. The technical difficulties of bisulfite treatment yield poor coverage, thus these methods might not be able to unveil untargeted features of different cell populations. For this reason, Farlik et al. inferred cell line drug response in developing the scWGBS approach [84]. Finally, recent single-cell studies proposed different analytical tools to correct the input features, such as the use of a deep neural network to reconstruct noisy and missing CpG data [87]. Using computational methods to fill in sparse data will not only rescue poorly resolved data but may also be applied to identify biomarkers, project clonal evolution, or rank potential drug responses.

Single-cell proteomics

There are many layers to deciphering cell-to-cell heterogeneity. Since there is not a direct 1:1 turnover of mRNA occurrence to protein translation, adopting single-cell proteomic studies provides information on the final layer of inter- and intra-tumour heterogeneity. Unlike genetic or expression analysis, proteomic investigations at the single-cell level lack a way to amplify the starting material. Therefore, single-cell proteomic studies have the technical challenge of developing more sensitive methods for detection.

Experimental and computational methods

Quantifying proteins at single-cell resolution is a developing technology complicated by the transient nature of functional proteins. Much like other -omics sections covered earlier, investigating the proteome at single-cell resolution requires accounting for low input levels [88]. Single-cell time-of-flight mass cytometry (CyTOF) is one method used to address this issue. CyTOF targets epitopes of interest utilizing antibodies coupled with transient metal element isotopes. Single-cell droplets are introduced to inductively coupled argon plasma where the cell is vaporized, and the atomic constituents are ionized before time-of-flight sampling [89]. Since CyTOF is limited to around 50 parameters, this approach yields a much lower throughput than scRNA-seq. However, it is more affordable than scRNA-seq and can help to determine if an enrichment (e.g. FACS) step is necessary before transcriptome analysis. Another mass spectrometry approach recently proposed is a method known as single-cell proteomics by mass spectrometry (SCoPE-MS). The SCoPE-MS workflow was designed to isolate protein from single-cells and prepare each cell for MS. SCoPE-MS attempts to resolve the issue of protein loss during transfer and low starting material by manually separating and lysing cells. This method includes tandem mass tags for reporter ion relative abundance quantification [90]. Alternatively, non-MS approaches for single-cell proteomic studies can utilize chip-based isolation. In the single-cell barcoded chip (SCBC) method, cells are isolated into wells and probed with antibody arrays which are analyzed by a microarray scanner [91]. Antibody arrays are also utilized in single-cell western blotting (scWB) for which isolated cells are lysed, and SDS-PAGE is applied to each well. Relative to the MS approaches, scWB is limited to probing the sample with a smaller panel of antibodies.
There are a variety of statistical tools available for inferring subpopulations and subpopulation specific markers from single-cell protein data including SPADE, Phenograph, and Wishbone [92]. For example, histoCAT is a new powerful integrative method used to integrate single-cell CyTOF measurement with image-based spatial information to detect spatial and phenotype interaction at the cellular level [93]. Also, new studies propose different methods to construct dynamic protein signalling networks using single-cell protein measurement [94, 95]. In particular, one new approach created a dynamic regulation network from CyTOF measurements to model the drug perturbation of the epithelial-to-mesenchymal transition [96]. This type of approach can then facilitate the discovery of critical events correlated with a cell state transition.

Exemplary applications

One example of the SCBC method for single-cell chip-based proteomic investigation first heavily dilutes a FACS enriched population before loading them into microchambers. Wei et al. used a phosphoproteomic antibody array to profile mTORki resistant glioblastomas [97]. This SCBC study, cells were lysed after isolation, and the protein contents were captured using custom antibody barcodes. Chip-based methods can assay a relatively larger number of proteins (n > 40) than other techniques currently available, but still fewer the MS approaches [88]. For this reason, the continuing development of single-cell mass spectrometry strategies such as CyTOF will be the technology that unlocks single-cell proteomic scalability. As a recent example, a panel of more than 30 antibodies was used in CyTOF to analyze tumour cells, adjacent normal tissue, and blood from 28 patients with lung adenocarcinoma [98]. The CyTOF data reported here revealed that tumour-infiltrating myeloid cells likely shape the composition of anti-tumour T-cells. Thus, the ability to profile large population of single-cell surface markers has powerful implication in immunology.

Single-cell multi-omics

Single-cell multi-omic strategies capture the most accurate state of factors that contribute to the cellular phenotype. Ultimately, the integration of several layers of data will be necessary for deconvoluting the relationship between expression, function, and identity. This is because bulk level analysis can only describe the general trends in a population that can mask cellular subtypes [99]. Multi-omic studies are complicated by the technical requirement of separating and preserving different molecular layers from the same cell. Bioinformatics and computational biology provide critical support for reconstructing features that may become noisy as a result of sample loss during multi-omic sample preparation.

Experimental methods

The most cutting-edge research calls for investigators to combine all of the techniques discussed to reconstruct multi–omic single-cell profiles (Fig. 4). Achieving this level of resolution will provide the most comprehensive profile of cell-to-cell diversity in normal and tumour tissue and inform researchers on the impact of single-cell genetic and epigenetic heterogeneity. Bock et al. proposed that molecules collected from the same cell can be assayed by one of several approaches depending on the desired downstream application [100]. For example, two methods described involve either separating the molecular layers (e.g. DNA and RNA) into their equivalents or splitting the sample itself into different fractions and proceeding with the desired isolation within the given fraction. Alternatively, multi-omic methods can be combined into a single workflow [100]. Taken together, the cluster of data generated by multi-omic approaches can infer the underlying triggers of cellular identity and function. As these techniques in cell isolation and amplification continue to improve, multiple layers of heterogeneity reconstruction can be used to identify neoplastic predisposition markers and provide a refined map for precise drug treatment regimen.

Exemplary applications

Since multi-omic strategies are at the frontier of single-cell research, the body of literature remains relatively nascent. The majority of current available single-cell multi-omics technologies are focused on the link between epigenetic and transcriptional variations. Macaulay et al. established scG&T-seq to simultaneously measure genetic variation and gene expression via separation of gDNA from polyA RNA using a biotinylated oligo-dT primer [101]. scG&T-seq equips Smart-seq 2 for whole transcriptome analysis and offers various methods for DNA amplification depending on the downstream application (MDA, PicoPlex etc.). In contrast to conventional scRNASeq sequencing methods, scG&T-seq utilizes ERCC-spike-ins to assess the number of genes expressed and transcript coverage lengths. Angermueller et al. developed another method called scM&T-seq to evaluate the relationship between methylation and transcription variations in heterogeneous cell populations through scRNASeq and scBS-seq techniques [102]. scM&T-seq was applied in discriminating 61 mouse serum ESCs (embryonic stem cells) and 16 ESCs grown in 2i media. The results showed that the connection strength between methylome and transcriptome varies from cell to cell. Another similar method called scMT-seq applied Smart-seq 2 and scRRBS for single-cell transcriptome sequencing and methylome sequencing, respectively [103]. Compared to scM&T-seq, scMT-seq provides similar CpG islands overlap in a more cost-effective way. This study helped to reveal the relationship of gene expression and DNA methylation in gene body and promoter regions in neuron single cells.
Regarding simultaneous measurement of gene expression with another omics data generated from the same cell, DR-Seq is an example of pioneering work on single-cell DNA and RNA parallel sequencing [104]. Without manually separating the nucleus and cytosolic mRNA, DR-Seq applies a quasi-linear amplification method with predefined adaptors to quantify gDNA and mRNA. Additionally, the comparison of DR-Seq and CEL-Seq showed that the additional steps for amplification of gDNA would not affect the mRNA results. However, this single-pot strategy requires in silico masking of the coding sequences (exonic region) of the genome to determine copy number variation, which leads to incomplete transcripts from the cell. Another recently published work by Stoeckius et al. developed CITE-seq to integrate cellular protein markers and transcriptome in single cells through oligonucleotide-labeled antibodies [105]. CITE-Seq not only enabled to differentiate cellular subgroups based on surface protein expression, but also achieved a consistent output of protein detection with currently standardized flow cytometry. Compared to scRNA-seq alone, CITE-Seq demonstrates both the highly consistent protein and RNA profiles with literature and also an enhancement of characterization of cell phenotypes based on immune cell experiments.
From the analytical perspective, certain additional layers can be reconstructed given one layer is provided. For example, SNVs can be directly extracted from RNA-seq reads and then correlated to gene expression [106]. In addition, by using the input fastq files it is now possible to highlight predictive DNA motifs linked to methylation profiles [87]. However, most of the high-throughput scRNA-seq pipelines are designed specifically for mRNA expression counting. Since the library products are quite short, there is fairly limited capability to do this without modifying more commercial protocols.
Three-omics single-cell assays also became possible. Recently, Hou et al. established scTrio-seq to simultaneously sequence and analyze single cell’s genomic copy number variations, DNA methylation and transcriptomic gene expression together [107]. scTrio-seq demonstrated its ability to efficiently measure DNA methylome, transcriptome and genome copy number compared to scRRBS, bulk cell RNA-seq and bulk cell RRBS and bulk cell WGBS. The integration of triple omics information via scTrio-seq on 25 HCC cancer cells identifies two heterogeneous subpopulations with different malignancy and metastasis potential.

Conclusions

High-throughput sequencing techniques provide clinicians with a more comprehensive understanding of the genetic and epigenetic heterogeneity in normal and cancer cells. Moreover, future personalized treatments might integrate as a routine single-cell strategy to unveil intra-tumour heterogeneity and thus provide a more accurate therapy regimen. Multi-omics approaches that detail inter- and intra-tumour heterogeneity within individual patients will continue to evolve and provide critical insight to informing more accurate treatment regimens based on prognosticated drug response. In addition, these emerging molecular techniques when combined with computational analysis tools will advance research in other areas such as developmental biology, biotechnology, pathology and more. As larger amounts of single-cell data become publicly available, there will be increased opportunities to identify subclonal-specific biomarkers at a personalized level. User-friendly data portals for single-cell analysis, such as Granatum, will become increasingly integral in the bench-to-bedside transition [61]. The comprehensive annotation and analysis of single-cell datasets will be the foundation of understanding how cell-to-cell variability in normal and cancer cells influence cellular identity and function in the human body.

Authors' contributions

LXG conceptualized the project. MAO wrote the manuscript, with assistance from OP, XZ, SH, TKW, RS and LXG. All authors read and approved the final manuscript.

Acknowledgements

We would like to thank all the members of the Garmire Lab for their helpful discussions and manuscript review.

Competing interests

The authors declare that they have no competing interests.

Availability of data and materials

Not applicable.
Not applicable.

Funding

This research was supported by grants K01ES025434 awarded by NIEHS through funds provided by the trans-NIH Big Data to Knowledge (BD2 K) initiative (http://​datascience.​nih.​gov/​bd2k), P20 COBRE GM103457 awarded by NIH/NIGMS, R01 LM012373 awarded by NLM, and R01 HD084633 awarded by NICHD to LX Garmire.

Publisher’s Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.
Open AccessThis article is distributed under the terms of the Creative Commons Attribution 4.0 International License (http://​creativecommons.​org/​licenses/​by/​4.​0/​), which permits unrestricted use, distribution, and reproduction in any medium, provided you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons license, and indicate if changes were made.
Literatur
1.
Zurück zum Zitat Araten DJ, Golde DW, Zhang RH, Thaler HT, Gargiulo L, Notaro R et al (2005) A quantitative measurement of the human somatic mutation rate. Cancer Res 65(18):8111–8117CrossRefPubMed Araten DJ, Golde DW, Zhang RH, Thaler HT, Gargiulo L, Notaro R et al (2005) A quantitative measurement of the human somatic mutation rate. Cancer Res 65(18):8111–8117CrossRefPubMed
2.
Zurück zum Zitat Tomasetti C, Vogelstein B, Parmigiani G (2013) Half or more of the somatic mutations in cancers of self-renewing tissues originate prior to tumor initiation. Proc Natl Acad Sci 110(6):1999–2004CrossRefPubMedPubMedCentral Tomasetti C, Vogelstein B, Parmigiani G (2013) Half or more of the somatic mutations in cancers of self-renewing tissues originate prior to tumor initiation. Proc Natl Acad Sci 110(6):1999–2004CrossRefPubMedPubMedCentral
3.
Zurück zum Zitat Blokzijl F, de Ligt J, Jager M, Sasselli V, Roerink S, Sasaki N et al (2016) Tissue-specific mutation accumulation in human adult stem cells during life. Nature 538(7624):260–264CrossRefPubMedPubMedCentral Blokzijl F, de Ligt J, Jager M, Sasselli V, Roerink S, Sasaki N et al (2016) Tissue-specific mutation accumulation in human adult stem cells during life. Nature 538(7624):260–264CrossRefPubMedPubMedCentral
4.
Zurück zum Zitat Tomasetti C, Li L, Vogelstein B (2017) Stem cell divisions, somatic mutations, cancer etiology, and cancer prevention. Science 355(6331):1330CrossRefPubMed Tomasetti C, Li L, Vogelstein B (2017) Stem cell divisions, somatic mutations, cancer etiology, and cancer prevention. Science 355(6331):1330CrossRefPubMed
5.
Zurück zum Zitat Tak YG, Farnham PJ (2015) Making sense of GWAS: using epigenomics and genome engineering to understand the functional relevance of SNPs in non-coding regions of the human genome. Epigenetics Chromatin 8(1):57CrossRefPubMedPubMedCentral Tak YG, Farnham PJ (2015) Making sense of GWAS: using epigenomics and genome engineering to understand the functional relevance of SNPs in non-coding regions of the human genome. Epigenetics Chromatin 8(1):57CrossRefPubMedPubMedCentral
8.
Zurück zum Zitat Harper KL, Sosa MS, Entenberg D, Hosseini H, Cheung JF, Nobre R et al (2016) Mechanism of early dissemination and metastasis in Her2+ mammary cancer. Nature 540:588CrossRef Harper KL, Sosa MS, Entenberg D, Hosseini H, Cheung JF, Nobre R et al (2016) Mechanism of early dissemination and metastasis in Her2+ mammary cancer. Nature 540:588CrossRef
9.
Zurück zum Zitat Robinson DR, Wu Y-M, Lonigro RJ, Vats P, Cobain E, Everett J et al (2017) Integrative clinical genomics of metastatic cancer. Nature 548(7667):297–303CrossRefPubMed Robinson DR, Wu Y-M, Lonigro RJ, Vats P, Cobain E, Everett J et al (2017) Integrative clinical genomics of metastatic cancer. Nature 548(7667):297–303CrossRefPubMed
10.
Zurück zum Zitat Tang F, Barbacioru C, Wang Y, Nordman E, Lee C, Xu N et al (2009) mRNA-Seq whole-transcriptome analysis of a single cell. Nat Methods 6(5):377–382CrossRefPubMed Tang F, Barbacioru C, Wang Y, Nordman E, Lee C, Xu N et al (2009) mRNA-Seq whole-transcriptome analysis of a single cell. Nat Methods 6(5):377–382CrossRefPubMed
11.
Zurück zum Zitat Gerlinger M, Rowan AJ, Horswell S, Larkin J, Endesfelder D, Gronroos E (2012) Intratumor heterogeneity and branched evolution revealed by multiregion sequencing. N Engl J Med 366:883–892CrossRefPubMedPubMedCentral Gerlinger M, Rowan AJ, Horswell S, Larkin J, Endesfelder D, Gronroos E (2012) Intratumor heterogeneity and branched evolution revealed by multiregion sequencing. N Engl J Med 366:883–892CrossRefPubMedPubMedCentral
12.
Zurück zum Zitat Cheow LF, Courtois ET, Tan Y, Viswanathan R, Xing Q, Tan RZ et al (2016) Single-cell multimodal profiling reveals cellular epigenetic heterogeneity. Nat Methods 13(10):833–836CrossRefPubMed Cheow LF, Courtois ET, Tan Y, Viswanathan R, Xing Q, Tan RZ et al (2016) Single-cell multimodal profiling reveals cellular epigenetic heterogeneity. Nat Methods 13(10):833–836CrossRefPubMed
14.
Zurück zum Zitat Navin N, Kendall J, Troge J, Andrews P, Rodgers L, McIndoo J et al (2011) Tumour evolution inferred by single-cell sequencing. Nature 472(7341):90–94CrossRefPubMedPubMedCentral Navin N, Kendall J, Troge J, Andrews P, Rodgers L, McIndoo J et al (2011) Tumour evolution inferred by single-cell sequencing. Nature 472(7341):90–94CrossRefPubMedPubMedCentral
15.
Zurück zum Zitat Smith MA, Nielsen CB, Chan FC, McPherson A, Roth A, Farahani H et al (2017) E-scape: interactive visualization of single-cell phylogenetics and cancer evolution. Nat Methods 14(6):549–550CrossRefPubMed Smith MA, Nielsen CB, Chan FC, McPherson A, Roth A, Farahani H et al (2017) E-scape: interactive visualization of single-cell phylogenetics and cancer evolution. Nat Methods 14(6):549–550CrossRefPubMed
16.
Zurück zum Zitat Salehi S, Steif A, Roth A, Aparicio S, Bouchard-Côté A, Shah SP (2017) ddClone: joint statistical inference of clonal populations from single cell and bulk tumour sequencing data. Genome Biol 18(1):44CrossRefPubMedPubMedCentral Salehi S, Steif A, Roth A, Aparicio S, Bouchard-Côté A, Shah SP (2017) ddClone: joint statistical inference of clonal populations from single cell and bulk tumour sequencing data. Genome Biol 18(1):44CrossRefPubMedPubMedCentral
17.
18.
Zurück zum Zitat Prasad V, De Jesus K, Mailankody S (2017) The high price of anticancer drugs: origins, implications, barriers, solutions. Nat Rev Clin Oncol 14(6):381–390 (advance online publication) CrossRefPubMed Prasad V, De Jesus K, Mailankody S (2017) The high price of anticancer drugs: origins, implications, barriers, solutions. Nat Rev Clin Oncol 14(6):381–390 (advance online publication) CrossRefPubMed
19.
Zurück zum Zitat Fojo T, Mailankody S, Lo A (2014) Unintended consequences of expensive cancer therapeutics—the pursuit of marginal indications and a me-too mentality that stifles innovation and creativity: the John Conley lecture. JAMA Otolaryngol Head Neck Surg 140(12):1225–1236CrossRefPubMed Fojo T, Mailankody S, Lo A (2014) Unintended consequences of expensive cancer therapeutics—the pursuit of marginal indications and a me-too mentality that stifles innovation and creativity: the John Conley lecture. JAMA Otolaryngol Head Neck Surg 140(12):1225–1236CrossRefPubMed
20.
Zurück zum Zitat Gerlinger M, Horswell S, Larkin J, Rowan AJ, Salm MP, Varela I et al (2014) Genomic architecture and evolution of clear cell renal cell carcinomas defined by multiregion sequencing. Nat Genet 46(3):225–233CrossRefPubMedPubMedCentral Gerlinger M, Horswell S, Larkin J, Rowan AJ, Salm MP, Varela I et al (2014) Genomic architecture and evolution of clear cell renal cell carcinomas defined by multiregion sequencing. Nat Genet 46(3):225–233CrossRefPubMedPubMedCentral
21.
Zurück zum Zitat Ding L, Ley TJ, Larson DE, Miller CA, Koboldt DC, Welch JS et al (2012) Clonal evolution in relapsed acute myeloid leukaemia revealed by whole-genome sequencing. Nature 481(7382):506–510CrossRefPubMedPubMedCentral Ding L, Ley TJ, Larson DE, Miller CA, Koboldt DC, Welch JS et al (2012) Clonal evolution in relapsed acute myeloid leukaemia revealed by whole-genome sequencing. Nature 481(7382):506–510CrossRefPubMedPubMedCentral
22.
Zurück zum Zitat Schwartz R, Schaffer AA (2017) The evolution of tumour phylogenetics: principles and practice. Nat Rev Genet 18(4):213–229CrossRefPubMed Schwartz R, Schaffer AA (2017) The evolution of tumour phylogenetics: principles and practice. Nat Rev Genet 18(4):213–229CrossRefPubMed
23.
Zurück zum Zitat Islam S, Kjällquist U, Moliner A, Zajac P, Fan J-B, Lönnerberg P et al (2011) Characterization of the single-cell transcriptional landscape by highly multiplex RNA-seq. Genome Res 21(7):1160–1167CrossRefPubMedPubMedCentral Islam S, Kjällquist U, Moliner A, Zajac P, Fan J-B, Lönnerberg P et al (2011) Characterization of the single-cell transcriptional landscape by highly multiplex RNA-seq. Genome Res 21(7):1160–1167CrossRefPubMedPubMedCentral
24.
Zurück zum Zitat Ramskold D, Luo S, Wang YC, Li R, Deng Q, Faridani OR (2012) Full-length mRNA-seq from single-cell levels of RNA and individual circulating tumor cells. Nat Biotechnol 30:777–782CrossRefPubMedPubMedCentral Ramskold D, Luo S, Wang YC, Li R, Deng Q, Faridani OR (2012) Full-length mRNA-seq from single-cell levels of RNA and individual circulating tumor cells. Nat Biotechnol 30:777–782CrossRefPubMedPubMedCentral
25.
Zurück zum Zitat Hashimshony T, Wagner F, Sher N, Yanai I (2012) CEL-Seq: single-cell RNA-seq by multiplexed linear amplification. Cell Reports 2(3):666–673CrossRefPubMed Hashimshony T, Wagner F, Sher N, Yanai I (2012) CEL-Seq: single-cell RNA-seq by multiplexed linear amplification. Cell Reports 2(3):666–673CrossRefPubMed
26.
Zurück zum Zitat Islam S, Zeisel A, Joost S, La Manno G, Zajac P, Kasper M et al (2014) Quantitative single-cell RNA-seq with unique molecular identifiers. Nat Methods 11(2):163–166CrossRefPubMed Islam S, Zeisel A, Joost S, La Manno G, Zajac P, Kasper M et al (2014) Quantitative single-cell RNA-seq with unique molecular identifiers. Nat Methods 11(2):163–166CrossRefPubMed
27.
Zurück zum Zitat Picelli S, Bjorklund AK, Faridani OR, Sagasser S, Winberg G, Sandberg R (2013) Smart-seq2 for sensitive full-length transcriptome profiling in single cells. Nat Methods 10(11):1096–1098CrossRefPubMed Picelli S, Bjorklund AK, Faridani OR, Sagasser S, Winberg G, Sandberg R (2013) Smart-seq2 for sensitive full-length transcriptome profiling in single cells. Nat Methods 10(11):1096–1098CrossRefPubMed
28.
Zurück zum Zitat Zheng GXY, Terry JM, Belgrader P, Ryvkin P, Bent ZW, Wilson R et al (2017) Massively parallel digital transcriptional profiling of single cells. Nat Commun 8:14049CrossRefPubMedPubMedCentral Zheng GXY, Terry JM, Belgrader P, Ryvkin P, Bent ZW, Wilson R et al (2017) Massively parallel digital transcriptional profiling of single cells. Nat Commun 8:14049CrossRefPubMedPubMedCentral
29.
Zurück zum Zitat Macosko EZ, Basu A, Satija R, Nemesh J, Shekhar K, Goldman M et al (2015) Highly parallel genome-wide expression profiling of individual cells using nanoliter droplets. Cell 161(5):1202–1214CrossRefPubMedPubMedCentral Macosko EZ, Basu A, Satija R, Nemesh J, Shekhar K, Goldman M et al (2015) Highly parallel genome-wide expression profiling of individual cells using nanoliter droplets. Cell 161(5):1202–1214CrossRefPubMedPubMedCentral
30.
Zurück zum Zitat Klein AM, Mazutis L, Akartuna I, Tallapragada N, Veres A, Li V et al (2015) Droplet barcoding for single-cell transcriptomics applied to embryonic stem cells. Cell 161(5):1187–1201CrossRefPubMedPubMedCentral Klein AM, Mazutis L, Akartuna I, Tallapragada N, Veres A, Li V et al (2015) Droplet barcoding for single-cell transcriptomics applied to embryonic stem cells. Cell 161(5):1187–1201CrossRefPubMedPubMedCentral
31.
Zurück zum Zitat Zheng GX, Lau BT, Schnall-Levin M, Jarosz M, Bell JM, Hindson CM et al (2016) Haplotyping germline and cancer genomes with high-throughput linked-read sequencing. Nat Biotechnol 34(3):303–311CrossRefPubMedPubMedCentral Zheng GX, Lau BT, Schnall-Levin M, Jarosz M, Bell JM, Hindson CM et al (2016) Haplotyping germline and cancer genomes with high-throughput linked-read sequencing. Nat Biotechnol 34(3):303–311CrossRefPubMedPubMedCentral
34.
Zurück zum Zitat Gierahn TM, Wadsworth MH II, Hughes TK, Bryson BD, Butler A, Satija R et al (2017) Seq-Well: portable, low-cost RNA sequencing of single cells at high throughput. Nat Methods 14(4):395–398CrossRefPubMedPubMedCentral Gierahn TM, Wadsworth MH II, Hughes TK, Bryson BD, Butler A, Satija R et al (2017) Seq-Well: portable, low-cost RNA sequencing of single cells at high throughput. Nat Methods 14(4):395–398CrossRefPubMedPubMedCentral
35.
Zurück zum Zitat Macosko EZ, Basu A, Satija R, Nemesh J, Shekhar K, Goldman M et al (2015) Highly parallel genome-wide expression profiling of individual CELLS using nanoliter droplets. Cell 161(5):1202–1214CrossRefPubMedPubMedCentral Macosko EZ, Basu A, Satija R, Nemesh J, Shekhar K, Goldman M et al (2015) Highly parallel genome-wide expression profiling of individual CELLS using nanoliter droplets. Cell 161(5):1202–1214CrossRefPubMedPubMedCentral
36.
Zurück zum Zitat Zilionis R, Nainys J, Veres A, Savova V, Zemmour D, Klein AM et al (2017) Single-cell barcoding and sequencing using droplet microfluidics. Nat Protoc 12(1):44–73CrossRefPubMed Zilionis R, Nainys J, Veres A, Savova V, Zemmour D, Klein AM et al (2017) Single-cell barcoding and sequencing using droplet microfluidics. Nat Protoc 12(1):44–73CrossRefPubMed
37.
Zurück zum Zitat Unger MA, Chou H-P, Thorsen T, Scherer A, Quake SR (2000) Monolithic microfabricated valves and pumps by multilayer soft lithography. Science 288(5463):113CrossRefPubMed Unger MA, Chou H-P, Thorsen T, Scherer A, Quake SR (2000) Monolithic microfabricated valves and pumps by multilayer soft lithography. Science 288(5463):113CrossRefPubMed
39.
Zurück zum Zitat Svensson V, Natarajan KN, Ly L-H, Miragaia RJ, Labalette C, Macaulay IC et al (2017) Power analysis of single-cell RNA-sequencing experiments. Nat Methods 14(4):381–387CrossRefPubMedPubMedCentral Svensson V, Natarajan KN, Ly L-H, Miragaia RJ, Labalette C, Macaulay IC et al (2017) Power analysis of single-cell RNA-sequencing experiments. Nat Methods 14(4):381–387CrossRefPubMedPubMedCentral
40.
Zurück zum Zitat Huang L, Ma F, Chapman A, Lu S, Xie XS (2015) Single-cell whole-genome amplification and sequencing: methodology and applications. Annu Rev Genomics Hum Genet 16:79–102CrossRefPubMed Huang L, Ma F, Chapman A, Lu S, Xie XS (2015) Single-cell whole-genome amplification and sequencing: methodology and applications. Annu Rev Genomics Hum Genet 16:79–102CrossRefPubMed
41.
42.
Zurück zum Zitat Garvin T, Aboukhalil R, Kendall J, Baslan T, Atwal GS, Hicks J et al (2015) Interactive analysis and assessment of single-cell copy-number variations. Nat Methods 12(11):1058–1060CrossRefPubMedPubMedCentral Garvin T, Aboukhalil R, Kendall J, Baslan T, Atwal GS, Hicks J et al (2015) Interactive analysis and assessment of single-cell copy-number variations. Nat Methods 12(11):1058–1060CrossRefPubMedPubMedCentral
43.
Zurück zum Zitat Telenius H, Carter NP, Bebb CE, Nordenskjold M, Ponder BA, Tunnacliffe A (1992) Degenerate oligonucleotide-primed PCR: general amplification of target DNA by a single degenerate primer. Genomics 13(3):718–725CrossRefPubMed Telenius H, Carter NP, Bebb CE, Nordenskjold M, Ponder BA, Tunnacliffe A (1992) Degenerate oligonucleotide-primed PCR: general amplification of target DNA by a single degenerate primer. Genomics 13(3):718–725CrossRefPubMed
44.
Zurück zum Zitat de Bourcy CFA, De Vlaminck I, Kanbar JN, Wang J, Gawad C, Quake SR (2014) A quantitative comparison of single-cell whole genome amplification methods. PLoS ONE 9(8):e105585CrossRefPubMedPubMedCentral de Bourcy CFA, De Vlaminck I, Kanbar JN, Wang J, Gawad C, Quake SR (2014) A quantitative comparison of single-cell whole genome amplification methods. PLoS ONE 9(8):e105585CrossRefPubMedPubMedCentral
45.
Zurück zum Zitat Ning L, Li Z, Wang G, Hu W, Hou Q, Tong Y et al (2015) Quantitative assessment of single-cell whole genome amplification methods for detecting copy number variation using hippocampal neurons. Sci Rep 5:11415CrossRefPubMedPubMedCentral Ning L, Li Z, Wang G, Hu W, Hou Q, Tong Y et al (2015) Quantitative assessment of single-cell whole genome amplification methods for detecting copy number variation using hippocampal neurons. Sci Rep 5:11415CrossRefPubMedPubMedCentral
47.
Zurück zum Zitat Chen C, Xing D, Tan L, Li H, Zhou G, Huang L et al (2017) Single-cell whole-genome analyses by linear amplification via transposon insertion (LIANTI). Science 356(6334):189–194CrossRefPubMedPubMedCentral Chen C, Xing D, Tan L, Li H, Zhou G, Huang L et al (2017) Single-cell whole-genome analyses by linear amplification via transposon insertion (LIANTI). Science 356(6334):189–194CrossRefPubMedPubMedCentral
48.
Zurück zum Zitat Hou Y, Song L, Zhu P, Zhang B, Tao Y, Xu X (2012) Single-cell exome sequencing and monoclonal evolution of a JAK2-negative myeloproliferative neoplasm. Cell 148:873–885CrossRefPubMed Hou Y, Song L, Zhu P, Zhang B, Tao Y, Xu X (2012) Single-cell exome sequencing and monoclonal evolution of a JAK2-negative myeloproliferative neoplasm. Cell 148:873–885CrossRefPubMed
49.
Zurück zum Zitat Xu X, Hou Y, Yin X, Bao L, Tang A, Song L (2012) Single-cell exome sequencing reveals single-nucleotide mutation characteristics of a kidney tumor. Cell 148:373–385 Xu X, Hou Y, Yin X, Bao L, Tang A, Song L (2012) Single-cell exome sequencing reveals single-nucleotide mutation characteristics of a kidney tumor. Cell 148:373–385
50.
Zurück zum Zitat Gao R, Davis A, McDonald TO, Sei E, Shi X, Wang Y et al (2016) Punctuated copy number evolution and clonal stasis in triple-negative breast cancer. Nat Genet 48(10):1119–1130CrossRefPubMedPubMedCentral Gao R, Davis A, McDonald TO, Sei E, Shi X, Wang Y et al (2016) Punctuated copy number evolution and clonal stasis in triple-negative breast cancer. Nat Genet 48(10):1119–1130CrossRefPubMedPubMedCentral
51.
Zurück zum Zitat The Cancer Genome Atlas Research Network (2017) Integrated genomic and molecular characterization of cervical cancer. Nature 543(7645):378–384CrossRef The Cancer Genome Atlas Research Network (2017) Integrated genomic and molecular characterization of cervical cancer. Nature 543(7645):378–384CrossRef
52.
Zurück zum Zitat The Cancer Genome Atlas Research Network (2017) Integrated genomic characterization of oesophageal carcinoma. Nature 541(7636):169–175CrossRef The Cancer Genome Atlas Research Network (2017) Integrated genomic characterization of oesophageal carcinoma. Nature 541(7636):169–175CrossRef
53.
Zurück zum Zitat Robinson DR, Wu YM, Lonigro RJ, Vats P, Cobain E, Everett J et al (2017) Integrative clinical genomics of metastatic cancer. Nature 548(7667):297–303CrossRefPubMed Robinson DR, Wu YM, Lonigro RJ, Vats P, Cobain E, Everett J et al (2017) Integrative clinical genomics of metastatic cancer. Nature 548(7667):297–303CrossRefPubMed
54.
Zurück zum Zitat Tirosh I, Venteicher AS, Hebert C, Escalante LE, Patel AP, Yizhak K et al (2016) Single-cell RNA-seq supports a developmental hierarchy in human oligodendroglioma. Nature 539(7628):309–313CrossRefPubMedPubMedCentral Tirosh I, Venteicher AS, Hebert C, Escalante LE, Patel AP, Yizhak K et al (2016) Single-cell RNA-seq supports a developmental hierarchy in human oligodendroglioma. Nature 539(7628):309–313CrossRefPubMedPubMedCentral
55.
Zurück zum Zitat Yang J, Tanaka Y, Seay M, Li Z, Jin J, Garmire LX et al (2017) Single cell transcriptomics reveals unanticipated features of early hematopoietic precursors. Nucleic Acids Res 45(3):1281–1296PubMed Yang J, Tanaka Y, Seay M, Li Z, Jin J, Garmire LX et al (2017) Single cell transcriptomics reveals unanticipated features of early hematopoietic precursors. Nucleic Acids Res 45(3):1281–1296PubMed
56.
Zurück zum Zitat Zhu X, Ching T, Pan X, Weissman SM, Garmire L (2017) Detecting heterogeneity in single-cell RNA-Seq data by non-negative matrix factorization. PeerJ 5:e2888CrossRefPubMedPubMedCentral Zhu X, Ching T, Pan X, Weissman SM, Garmire L (2017) Detecting heterogeneity in single-cell RNA-Seq data by non-negative matrix factorization. PeerJ 5:e2888CrossRefPubMedPubMedCentral
57.
Zurück zum Zitat Trapnell C, Cacchiarelli D, Grimsby J, Pokharel P, Li S, Morse M et al (2014) The dynamics and regulators of cell fate decisions are revealed by pseudotemporal ordering of single cells. Nat Biotechnol 32(4):381–386CrossRefPubMedPubMedCentral Trapnell C, Cacchiarelli D, Grimsby J, Pokharel P, Li S, Morse M et al (2014) The dynamics and regulators of cell fate decisions are revealed by pseudotemporal ordering of single cells. Nat Biotechnol 32(4):381–386CrossRefPubMedPubMedCentral
58.
Zurück zum Zitat Buettner F, Natarajan KN, Casale FP, Proserpio V, Scialdone A, Theis FJ et al (2015) Computational analysis of cell-to-cell heterogeneity in single-cell RNA-sequencing data reveals hidden subpopulations of cells. Nat Biotechol 33(2):155–160CrossRef Buettner F, Natarajan KN, Casale FP, Proserpio V, Scialdone A, Theis FJ et al (2015) Computational analysis of cell-to-cell heterogeneity in single-cell RNA-sequencing data reveals hidden subpopulations of cells. Nat Biotechol 33(2):155–160CrossRef
59.
Zurück zum Zitat Campbell KR, Yau C (2016) Order under uncertainty: robust differential expression analysis using probabilistic models for pseudotime inference. PLoS Comput Biol 12(11):e1005212CrossRefPubMedPubMedCentral Campbell KR, Yau C (2016) Order under uncertainty: robust differential expression analysis using probabilistic models for pseudotime inference. PLoS Comput Biol 12(11):e1005212CrossRefPubMedPubMedCentral
60.
Zurück zum Zitat Anchang B, Hart TD, Bendall SC, Qiu P, Bjornson Z, Linderman M et al (2016) Visualization and cellular hierarchy inference of single-cell data using SPADE. Nat Protoc 11(7):1264–1279CrossRefPubMed Anchang B, Hart TD, Bendall SC, Qiu P, Bjornson Z, Linderman M et al (2016) Visualization and cellular hierarchy inference of single-cell data using SPADE. Nat Protoc 11(7):1264–1279CrossRefPubMed
61.
Zurück zum Zitat Zhu X, Wolfgruber TK, Tasato A, Arisdakessian C, Garmire DG, Garmire LX (2017) Granatum: a graphical single-cell RNA-Seq analysis pipeline for genomics scientists. Genome Med 9(1):108 Zhu X, Wolfgruber TK, Tasato A, Arisdakessian C, Garmire DG, Garmire LX (2017) Granatum: a graphical single-cell RNA-Seq analysis pipeline for genomics scientists. Genome Med 9(1):108
63.
Zurück zum Zitat Patel AP, Tirosh I, Trombetta JJ, Shalek AK, Gillespie SM, Wakimoto H (2014) Single-cell RNA-seq highlights intratumoral heterogeneity in primary glioblastoma. Science 344(6190):1396–1401CrossRefPubMedPubMedCentral Patel AP, Tirosh I, Trombetta JJ, Shalek AK, Gillespie SM, Wakimoto H (2014) Single-cell RNA-seq highlights intratumoral heterogeneity in primary glioblastoma. Science 344(6190):1396–1401CrossRefPubMedPubMedCentral
65.
Zurück zum Zitat Jordan NV, Bardia A, Wittner BS, Benes C, Ligorio M, Zheng Y et al (2016) HER2 expression identifies dynamic functional states within circulating breast cancer cells. Nature 537(7618):102–106CrossRefPubMedPubMedCentral Jordan NV, Bardia A, Wittner BS, Benes C, Ligorio M, Zheng Y et al (2016) HER2 expression identifies dynamic functional states within circulating breast cancer cells. Nature 537(7618):102–106CrossRefPubMedPubMedCentral
66.
Zurück zum Zitat Lupianez DG, Spielmann M, Mundlos S (2016) Breaking TADs: how alterations of chromatin domains result in disease. Trends Genet 32(4):225–237CrossRefPubMed Lupianez DG, Spielmann M, Mundlos S (2016) Breaking TADs: how alterations of chromatin domains result in disease. Trends Genet 32(4):225–237CrossRefPubMed
67.
Zurück zum Zitat Cremer T, Cremer C (2001) Chromosome territories, nuclear architecture and gene regulation in mammalian cells. Nat Rev Genet 2(4):292–301CrossRefPubMed Cremer T, Cremer C (2001) Chromosome territories, nuclear architecture and gene regulation in mammalian cells. Nat Rev Genet 2(4):292–301CrossRefPubMed
68.
Zurück zum Zitat Lieberman-Aiden E, van Berkum NL, Williams L, Imakaev M, Ragoczy T, Telling A et al (2009) Comprehensive mapping of long range interactions reveals folding principles of the human genome. Science 326(5950):289–293CrossRefPubMedPubMedCentral Lieberman-Aiden E, van Berkum NL, Williams L, Imakaev M, Ragoczy T, Telling A et al (2009) Comprehensive mapping of long range interactions reveals folding principles of the human genome. Science 326(5950):289–293CrossRefPubMedPubMedCentral
69.
Zurück zum Zitat Seaman L, Chen H, Brown M, Wangsa D, Patterson G, Camps J et al (2017) Nucleome analysis reveals structure-function relationships for colon cancer. Mol Can Res 15(7):821–830CrossRef Seaman L, Chen H, Brown M, Wangsa D, Patterson G, Camps J et al (2017) Nucleome analysis reveals structure-function relationships for colon cancer. Mol Can Res 15(7):821–830CrossRef
70.
Zurück zum Zitat Nagano T, Lubling Y, Stevens TJ, Schoenfelder S, Yaffe E, Dean W et al (2013) Single-cell Hi-C reveals cell-to-cell variability in chromosome structure. Nature 502(7469):59–64CrossRefPubMed Nagano T, Lubling Y, Stevens TJ, Schoenfelder S, Yaffe E, Dean W et al (2013) Single-cell Hi-C reveals cell-to-cell variability in chromosome structure. Nature 502(7469):59–64CrossRefPubMed
71.
72.
Zurück zum Zitat Cusanovich DA, Daza R, Adey A, Pliner H, Christiansen L, Gunderson KL et al (2015) Multiplex single cell profiling of chromatin accessibility by combinatorial cellular indexing. Science 348(6237):910–914CrossRefPubMedPubMedCentral Cusanovich DA, Daza R, Adey A, Pliner H, Christiansen L, Gunderson KL et al (2015) Multiplex single cell profiling of chromatin accessibility by combinatorial cellular indexing. Science 348(6237):910–914CrossRefPubMedPubMedCentral
73.
Zurück zum Zitat Buenrostro JD, Giresi PG, Zaba LC, Chang HY, Greenleaf WJ (2013) Transposition of native chromatin for fast and sensitive epigenomic profiling of open chromatin, DNA-binding proteins and nucleosome position. Nat Methods 10(12):1213–1218CrossRefPubMedPubMedCentral Buenrostro JD, Giresi PG, Zaba LC, Chang HY, Greenleaf WJ (2013) Transposition of native chromatin for fast and sensitive epigenomic profiling of open chromatin, DNA-binding proteins and nucleosome position. Nat Methods 10(12):1213–1218CrossRefPubMedPubMedCentral
74.
Zurück zum Zitat Nagano T, Lubling Y, Várnai C, Dudley C, Leung W, Baran Y et al (2017) Cell-cycle dynamics of chromosomal organization at single-cell resolution. Nature 547(7661):61-67 Nagano T, Lubling Y, Várnai C, Dudley C, Leung W, Baran Y et al (2017) Cell-cycle dynamics of chromosomal organization at single-cell resolution. Nature 547(7661):61-67
75.
Zurück zum Zitat Flyamer IM, Gassler J, Imakaev M, Brandão HB, Ulianov SV, Abdennur N et al (2017) Single-nucleus Hi-C reveals unique chromatin reorganization at oocyte-to-zygote transition. Nature 544(7648):110–114CrossRefPubMedPubMedCentral Flyamer IM, Gassler J, Imakaev M, Brandão HB, Ulianov SV, Abdennur N et al (2017) Single-nucleus Hi-C reveals unique chromatin reorganization at oocyte-to-zygote transition. Nature 544(7648):110–114CrossRefPubMedPubMedCentral
76.
Zurück zum Zitat Vitak SA, Torkenczy KA, Rosenkrantz JL, Fields AJ, Christiansen L, Wong MH et al (2017) Sequencing thousands of single-cell genomes with combinatorial indexing. Nat Methods 14(3):302–308CrossRefPubMed Vitak SA, Torkenczy KA, Rosenkrantz JL, Fields AJ, Christiansen L, Wong MH et al (2017) Sequencing thousands of single-cell genomes with combinatorial indexing. Nat Methods 14(3):302–308CrossRefPubMed
79.
Zurück zum Zitat Miura F, Ito T (2015) Highly sensitive targeted methylome sequencing by post-bisulfite adaptor tagging. DNA Res 22(1):13–18CrossRefPubMed Miura F, Ito T (2015) Highly sensitive targeted methylome sequencing by post-bisulfite adaptor tagging. DNA Res 22(1):13–18CrossRefPubMed
80.
Zurück zum Zitat Smallwood SA, Lee HJ, Angermueller C, Krueger F, Saadeh H, Peat J (2014) Single-cell genome-wide bisulfite sequencing for assessing epigenetic heterogeneity. Nat Methods 11:817–820CrossRefPubMedPubMedCentral Smallwood SA, Lee HJ, Angermueller C, Krueger F, Saadeh H, Peat J (2014) Single-cell genome-wide bisulfite sequencing for assessing epigenetic heterogeneity. Nat Methods 11:817–820CrossRefPubMedPubMedCentral
81.
Zurück zum Zitat Clark SJ, Smallwood SA, Lee HJ, Krueger F, Reik W, Kelsey G (2017) Genome-wide base-resolution mapping of DNA methylation in single cells using single-cell bisulfite sequencing (scBS-seq). Nat Protoc 12(3):534–547CrossRefPubMed Clark SJ, Smallwood SA, Lee HJ, Krueger F, Reik W, Kelsey G (2017) Genome-wide base-resolution mapping of DNA methylation in single cells using single-cell bisulfite sequencing (scBS-seq). Nat Protoc 12(3):534–547CrossRefPubMed
82.
Zurück zum Zitat Guo H, Zhu P, Guo F, Li X, Wu X, Fan X (2015) Profiling DNA methylome landscapes of mammalian cells with single-cell reduced-representation bisulfite sequencing. Nat Protoc 10:645–659CrossRefPubMed Guo H, Zhu P, Guo F, Li X, Wu X, Fan X (2015) Profiling DNA methylome landscapes of mammalian cells with single-cell reduced-representation bisulfite sequencing. Nat Protoc 10:645–659CrossRefPubMed
83.
Zurück zum Zitat Krueger F, Andrews SR (2011) Bismark: a flexible aligner and methylation caller for bisulfite-Seq applications. Bioinformatics 27(11):1571–1572CrossRefPubMedPubMedCentral Krueger F, Andrews SR (2011) Bismark: a flexible aligner and methylation caller for bisulfite-Seq applications. Bioinformatics 27(11):1571–1572CrossRefPubMedPubMedCentral
84.
Zurück zum Zitat Farlik M, Sheffield NC, Nuzzo A, Datlinger P, Schonegger A, Klughammer J et al (2015) Single-cell DNA methylome sequencing and bioinformatic inference of epigenomic cell-state dynamics. Cell Rep 10(8):1386–1397CrossRefPubMedPubMedCentral Farlik M, Sheffield NC, Nuzzo A, Datlinger P, Schonegger A, Klughammer J et al (2015) Single-cell DNA methylome sequencing and bioinformatic inference of epigenomic cell-state dynamics. Cell Rep 10(8):1386–1397CrossRefPubMedPubMedCentral
85.
Zurück zum Zitat Wu X, Inoue A, Suzuki T, Zhang Y (2017) Simultaneous mapping of active DNA demethylation and sister chromatid exchange in single cells. Genes Dev 31(5):511–523CrossRefPubMedPubMedCentral Wu X, Inoue A, Suzuki T, Zhang Y (2017) Simultaneous mapping of active DNA demethylation and sister chromatid exchange in single cells. Genes Dev 31(5):511–523CrossRefPubMedPubMedCentral
86.
Zurück zum Zitat Han L, Wu HJ, Zhu H, Kim KY, Marjani SL, Riester M et al (2017) Bisulfite-independent analysis of CpG island methylation enables genome-scale stratification of single cells. Nucleic Acids Res 5:e77 Han L, Wu HJ, Zhu H, Kim KY, Marjani SL, Riester M et al (2017) Bisulfite-independent analysis of CpG island methylation enables genome-scale stratification of single cells. Nucleic Acids Res 5:e77
87.
Zurück zum Zitat Angermueller C, Lee HJ, Reik W, Stegle O (2017) DeepCpG: accurate prediction of single-cell DNA methylation states using deep learning. Genome Biol 18(1):67CrossRefPubMedPubMedCentral Angermueller C, Lee HJ, Reik W, Stegle O (2017) DeepCpG: accurate prediction of single-cell DNA methylation states using deep learning. Genome Biol 18(1):67CrossRefPubMedPubMedCentral
88.
Zurück zum Zitat Heath JR, Ribas A, Mischel PS (2016) Single cell analytic tools for drug discovery and development. Nat Rev Drug Discov 15(3):204–216CrossRefPubMed Heath JR, Ribas A, Mischel PS (2016) Single cell analytic tools for drug discovery and development. Nat Rev Drug Discov 15(3):204–216CrossRefPubMed
89.
Zurück zum Zitat Bendall SC, Simonds EF, Qiu P, el Amir AD, Krutzik PO, Finck R et al (2011) Single-cell mass cytometry of differential immune and drug responses across a human hematopoietic continuum. Science 332(6030):687–696CrossRefPubMedPubMedCentral Bendall SC, Simonds EF, Qiu P, el Amir AD, Krutzik PO, Finck R et al (2011) Single-cell mass cytometry of differential immune and drug responses across a human hematopoietic continuum. Science 332(6030):687–696CrossRefPubMedPubMedCentral
91.
Zurück zum Zitat Shi Q, Qin L, Wei W, Geng F, Fan R, Shin YS et al (2012) Single-cell proteomic chip for profiling intracellular signaling pathways in single tumor cells. Proc Natl Acad Sci USA 109(2):419–424CrossRefPubMed Shi Q, Qin L, Wei W, Geng F, Fan R, Shin YS et al (2012) Single-cell proteomic chip for profiling intracellular signaling pathways in single tumor cells. Proc Natl Acad Sci USA 109(2):419–424CrossRefPubMed
93.
Zurück zum Zitat Schapiro D, Jackson HW, Raghuraman S, Fischer JR, Zanotelli VRT, Schulz D et al (2017) histoCAT: analysis of cell phenotypes and interactions in multiplex image cytometry data. Nat Methods 14(9):873-876 Schapiro D, Jackson HW, Raghuraman S, Fischer JR, Zanotelli VRT, Schulz D et al (2017) histoCAT: analysis of cell phenotypes and interactions in multiplex image cytometry data. Nat Methods 14(9):873-876
94.
Zurück zum Zitat Marr C, Zhou JX, Huang S (2016) Single-cell gene expression profiling and cell state dynamics: collecting data, correlating data points and connecting the dots. Curr Opin Biotechnol 39:207–214CrossRefPubMedPubMedCentral Marr C, Zhou JX, Huang S (2016) Single-cell gene expression profiling and cell state dynamics: collecting data, correlating data points and connecting the dots. Curr Opin Biotechnol 39:207–214CrossRefPubMedPubMedCentral
95.
Zurück zum Zitat Mukherjee S, Stewart D, Stewart W, Lanier LL, Das J (2017) Connecting the dots across time: reconstruction of single-cell signalling trajectories using time-stamped data. R Soc Open Sci 4(8):170811CrossRefPubMedPubMedCentral Mukherjee S, Stewart D, Stewart W, Lanier LL, Das J (2017) Connecting the dots across time: reconstruction of single-cell signalling trajectories using time-stamped data. R Soc Open Sci 4(8):170811CrossRefPubMedPubMedCentral
97.
Zurück zum Zitat Wei W, Shin Young S, Xue M, Matsutani T, Masui K, Yang H et al (2016) Single-cell phosphoproteomics resolves adaptive signaling dynamics and informs targeted combination therapy in glioblastoma. Cancer Cell 29(4):563–573CrossRefPubMedPubMedCentral Wei W, Shin Young S, Xue M, Matsutani T, Masui K, Yang H et al (2016) Single-cell phosphoproteomics resolves adaptive signaling dynamics and informs targeted combination therapy in glioblastoma. Cancer Cell 29(4):563–573CrossRefPubMedPubMedCentral
98.
Zurück zum Zitat Lavin Y, Kobayashi S, Leader A, Amir ED, Elefant N, Bigenwald C et al (2017) Innate immune landscape in early lung adenocarcinoma by paired single-cell analyses. Cell 169(4):750.e17–765.e17CrossRef Lavin Y, Kobayashi S, Leader A, Amir ED, Elefant N, Bigenwald C et al (2017) Innate immune landscape in early lung adenocarcinoma by paired single-cell analyses. Cell 169(4):750.e17–765.e17CrossRef
99.
Zurück zum Zitat Li H, Courtois ET, Sengupta D, Tan Y, Chen KH, Goh JJL et al (2017) Reference component analysis of single-cell transcriptomes elucidates cellular heterogeneity in human colorectal tumors. Nat Genet 49(5):708–718 (advance online publication) CrossRefPubMed Li H, Courtois ET, Sengupta D, Tan Y, Chen KH, Goh JJL et al (2017) Reference component analysis of single-cell transcriptomes elucidates cellular heterogeneity in human colorectal tumors. Nat Genet 49(5):708–718 (advance online publication) CrossRefPubMed
100.
Zurück zum Zitat Bock C, Farlik M, Sheffield NC (2017) Multi-omics of single cells: strategies and applications. Trends Biotechnol 34(8):605–608CrossRef Bock C, Farlik M, Sheffield NC (2017) Multi-omics of single cells: strategies and applications. Trends Biotechnol 34(8):605–608CrossRef
101.
Zurück zum Zitat Macaulay IC, Haerty W, Kumar P, Li YI, Hu TX, Teng MJ et al (2015) G&T-seq: parallel sequencing of single-cell genomes and transcriptomes. Nat Methods 12(6):519–522CrossRefPubMed Macaulay IC, Haerty W, Kumar P, Li YI, Hu TX, Teng MJ et al (2015) G&T-seq: parallel sequencing of single-cell genomes and transcriptomes. Nat Methods 12(6):519–522CrossRefPubMed
102.
Zurück zum Zitat Angermueller C, Clark SJ, Lee HJ, Macaulay IC, Teng MJ, Hu TX et al (2016) Parallel single-cell sequencing links transcriptional and epigenetic heterogeneity. Nat Methods 13(3):229–232CrossRefPubMedPubMedCentral Angermueller C, Clark SJ, Lee HJ, Macaulay IC, Teng MJ, Hu TX et al (2016) Parallel single-cell sequencing links transcriptional and epigenetic heterogeneity. Nat Methods 13(3):229–232CrossRefPubMedPubMedCentral
103.
Zurück zum Zitat Hu Y, Huang K, An Q, Du G, Hu G, Xue J et al (2016) Simultaneous profiling of transcriptome and DNA methylome from a single cell. Genome Biol 17(1):88CrossRefPubMedPubMedCentral Hu Y, Huang K, An Q, Du G, Hu G, Xue J et al (2016) Simultaneous profiling of transcriptome and DNA methylome from a single cell. Genome Biol 17(1):88CrossRefPubMedPubMedCentral
104.
Zurück zum Zitat Dey SS, Kester L, Spanjaard B, Bienko M, van Oudenaarden A (2015) Integrated genome and transcriptome sequencing of the same cell. Nat Biotechnol 33(3):285–289CrossRefPubMedPubMedCentral Dey SS, Kester L, Spanjaard B, Bienko M, van Oudenaarden A (2015) Integrated genome and transcriptome sequencing of the same cell. Nat Biotechnol 33(3):285–289CrossRefPubMedPubMedCentral
105.
Zurück zum Zitat Stoeckius M, Hafemeister C, Stephenson W, Houck-Loomis B, Chattopadhyay PK, Swerdlow H et al (2017) Simultaneous epitope and transcriptome measurement in single cells. Nat Methods 14(9):865–868 (advance online publication) CrossRefPubMed Stoeckius M, Hafemeister C, Stephenson W, Houck-Loomis B, Chattopadhyay PK, Swerdlow H et al (2017) Simultaneous epitope and transcriptome measurement in single cells. Nat Methods 14(9):865–868 (advance online publication) CrossRefPubMed
107.
Zurück zum Zitat Hou Y, Guo H, Cao C, Li X, Hu B, Zhu P et al (2016) Single-cell triple omics sequencing reveals genetic, epigenetic, and transcriptomic heterogeneity in hepatocellular carcinomas. Cell Res 26(3):304–319CrossRefPubMedPubMedCentral Hou Y, Guo H, Cao C, Li X, Hu B, Zhu P et al (2016) Single-cell triple omics sequencing reveals genetic, epigenetic, and transcriptomic heterogeneity in hepatocellular carcinomas. Cell Res 26(3):304–319CrossRefPubMedPubMedCentral
108.
Zurück zum Zitat Uhlén M, Fagerberg L, Hallström BM, Lindskog C, Oksvold P, Mardinoglu A et al (2015) Tissue-based map of the human proteome. Science 347(6220):1260419–1260419 Uhlén M, Fagerberg L, Hallström BM, Lindskog C, Oksvold P, Mardinoglu A et al (2015) Tissue-based map of the human proteome. Science 347(6220):1260419–1260419
109.
Zurück zum Zitat Thul PJ, Åkesson L, Wiking M, Mahdessian D, Geladaki A, Ait Blal H et al (2017) A subcellular map of the human proteome. Science 356(6340):eaal3321 Thul PJ, Åkesson L, Wiking M, Mahdessian D, Geladaki A, Ait Blal H et al (2017) A subcellular map of the human proteome. Science 356(6340):eaal3321
110.
Zurück zum Zitat Kuleshov MV, Jones MR, Rouillard AD, Fernandez NF, Duan Q, Wang Z et al (2016) Enrichr: a comprehensive gene set enrichment analysis web server 2016 update. Nucleic Acids Res 44(W1):W90–W97 Kuleshov MV, Jones MR, Rouillard AD, Fernandez NF, Duan Q, Wang Z et al (2016) Enrichr: a comprehensive gene set enrichment analysis web server 2016 update. Nucleic Acids Res 44(W1):W90–W97
111.
Zurück zum Zitat Griffith M, Spies NC, Krysiak K, McMichael JF, Coffman AC, Danos AM et al (2017) CIViC is a community knowledgebase for expert crowdsourcing the clinical interpretation of variants in cancer. Nat Genet 49(2):170–174 Griffith M, Spies NC, Krysiak K, McMichael JF, Coffman AC, Danos AM et al (2017) CIViC is a community knowledgebase for expert crowdsourcing the clinical interpretation of variants in cancer. Nat Genet 49(2):170–174
112.
Zurück zum Zitat Xin J, Mark A, Afrasiabi C, Tsueng G, Juchler M, Gopal N et al (2016) High-performance web services for querying gene and variant annotation. Genome Biol 17(1):91 Xin J, Mark A, Afrasiabi C, Tsueng G, Juchler M, Gopal N et al (2016) High-performance web services for querying gene and variant annotation. Genome Biol 17(1):91
113.
Zurück zum Zitat Jahn K, Kuipers J, Beerenwinkel N (2016) Tree inference for single-cell data. Genome Biol 17(1):86 Jahn K, Kuipers J, Beerenwinkel N (2016) Tree inference for single-cell data. Genome Biol 17(1):86
114.
Zurück zum Zitat Ross EM, Markowetz F (2016) OncoNEM: inferring tumor evolution from single-cell sequencing data. Genome Biol 17(1):69 Ross EM, Markowetz F (2016) OncoNEM: inferring tumor evolution from single-cell sequencing data. Genome Biol 17(1):69
115.
Zurück zum Zitat Li H (2012) Exploring single-sample SNP and INDEL calling with whole-genome de novo assembly. Bioinformatics 28(14):1838–1844 Li H (2012) Exploring single-sample SNP and INDEL calling with whole-genome de novo assembly. Bioinformatics 28(14):1838–1844
116.
Zurück zum Zitat Campbell KR, Yau C, Rattray M (2016) Order Under Uncertainty: Robust Differential Expression Analysis Using Probabilistic Models for Pseudotime Inference. PLoS Comput Biol 12(11):e1005212 Campbell KR, Yau C, Rattray M (2016) Order Under Uncertainty: Robust Differential Expression Analysis Using Probabilistic Models for Pseudotime Inference. PLoS Comput Biol 12(11):e1005212
117.
Zurück zum Zitat Dobin A, Davis CA, Schlesinger F, Drenkow J, Zaleski C, Jha S et al (2013) STAR: ultrafast universal RNA-seq aligner. Bioinformatics 29(1):15–21 Dobin A, Davis CA, Schlesinger F, Drenkow J, Zaleski C, Jha S et al (2013) STAR: ultrafast universal RNA-seq aligner. Bioinformatics 29(1):15–21
118.
Zurück zum Zitat Turner KM, Deshpande V, Beyter D, Koga T, Rusert J, Lee C et al (2017) Extrachromosomal oncogene amplification drives tumour evolution and genetic heterogeneity. Nature 543(7643):122–125 Turner KM, Deshpande V, Beyter D, Koga T, Rusert J, Lee C et al (2017) Extrachromosomal oncogene amplification drives tumour evolution and genetic heterogeneity. Nature 543(7643):122–125
119.
Zurück zum Zitat Talevich E, Shain AH, Botton T, Bastian BC (2016) CNVkit: genome-wide copy number detection and visualization from targeted DNA sequencing. PLoS Comput Biol 12(4):e1004873 Talevich E, Shain AH, Botton T, Bastian BC (2016) CNVkit: genome-wide copy number detection and visualization from targeted DNA sequencing. PLoS Comput Biol 12(4):e1004873
120.
Zurück zum Zitat Silva GO, Siegel MB, Mose LE, Parker JS, Sun W, Perou CM et al (2017) SynthEx: a synthetic-normal-based DNA sequencing tool for copy number alteration detection and tumor heterogeneity profiling. Genome Biol 18(1):66 Silva GO, Siegel MB, Mose LE, Parker JS, Sun W, Perou CM et al (2017) SynthEx: a synthetic-normal-based DNA sequencing tool for copy number alteration detection and tumor heterogeneity profiling. Genome Biol 18(1):66
121.
Zurück zum Zitat Lawrence MS, Stojanov P, Polak P, Kryukov GV, Cibulskis K, Sivachenko A et al (2013) Mutational heterogeneity in cancer and the search for new cancer-associated genes. Nature 499(7457):214–218 Lawrence MS, Stojanov P, Polak P, Kryukov GV, Cibulskis K, Sivachenko A et al (2013) Mutational heterogeneity in cancer and the search for new cancer-associated genes. Nature 499(7457):214–218
122.
Zurück zum Zitat Leiserson MDM, Vandin F, Wu HT, Dobson JR, Eldridge JV, Thomas JL et al (2014) Pan-cancer network analysis identifies combinations of rare somatic mutations across pathways and protein complexes. Nat Genet 47(2):106–114 Leiserson MDM, Vandin F, Wu HT, Dobson JR, Eldridge JV, Thomas JL et al (2014) Pan-cancer network analysis identifies combinations of rare somatic mutations across pathways and protein complexes. Nat Genet 47(2):106–114
123.
Zurück zum Zitat Setty M, Tadmor MD, Reich-Zeliger S, Angel O, Salame TM, Kathail P et al (2016) Wishbone identifies bifurcating developmental trajectories from single-cell data. Nat Biotechnol 34(6):637–645 Setty M, Tadmor MD, Reich-Zeliger S, Angel O, Salame TM, Kathail P et al (2016) Wishbone identifies bifurcating developmental trajectories from single-cell data. Nat Biotechnol 34(6):637–645
124.
Zurück zum Zitat Ramani V, Deng X, Qiu R, Gunderson KL, Steemers FJ, Disteche CM et al (2017) Massively multiplex single-cell Hi-C. Nat Methods 14(3):263–266 Ramani V, Deng X, Qiu R, Gunderson KL, Steemers FJ, Disteche CM et al (2017) Massively multiplex single-cell Hi-C. Nat Methods 14(3):263–266
125.
Zurück zum Zitat Zhang J, Mardis ER, Maher CA (2017) INTEGRATE-neo: a pipeline for personalized gene fusion neoantigen discovery. Bioinformatics 33(4):555 Zhang J, Mardis ER, Maher CA (2017) INTEGRATE-neo: a pipeline for personalized gene fusion neoantigen discovery. Bioinformatics 33(4):555
Metadaten
Titel
Using single-cell multiple omics approaches to resolve tumor heterogeneity
verfasst von
Michael A. Ortega
Olivier Poirion
Xun Zhu
Sijia Huang
Thomas K. Wolfgruber
Robert Sebra
Lana X. Garmire
Publikationsdatum
01.12.2017
Verlag
Springer Berlin Heidelberg
Erschienen in
Clinical and Translational Medicine / Ausgabe 1/2017
Elektronische ISSN: 2001-1326
DOI
https://doi.org/10.1186/s40169-017-0177-y

Weitere Artikel der Ausgabe 1/2017

Clinical and Translational Medicine 1/2017 Zur Ausgabe