Review Article
2 August 2024
Open access

Emerging toolkits for decoding the co-occurrence of modified histones and chromatin proteins

This article has been corrected.
VIEW CORRECTION
EMBO Rep
(2024)
25: 3202 - 3220

Abstract

In eukaryotes, DNA is packaged into chromatin with the help of highly conserved histone proteins. Together with DNA-binding proteins, posttranslational modifications (PTMs) on these histones play crucial roles in regulating genome function, cell fate determination, inheritance of acquired traits, cellular states, and diseases. While most studies have focused on individual DNA-binding proteins, chromatin proteins, or histone PTMs in bulk cell populations, such chromatin features co-occur and potentially act cooperatively to accomplish specific functions in a given cell. This review discusses state-of-the-art techniques for the simultaneous profiling of multiple chromatin features in low-input samples and single cells, focusing on histone PTMs, DNA-binding, and chromatin proteins. We cover the origins of the currently available toolkits, compare and contrast their characteristic features, and discuss challenges and perspectives for future applications. Studying the co-occurrence of histone PTMs, DNA-binding proteins, and chromatin proteins in single cells will be central for a better understanding of the biological relevance of combinatorial chromatin features, their impact on genomic output, and cellular heterogeneity.

Glossary

ACT-seq
Antibody-guided chromatin tagmentation
ATAC
Assay for transposase accessible chromatin
cDNA
Complementary DNA
ChDIP
Chromatin double immunoprecipitation
ChEC
Chromatin endogenous cleavage
ChEC-seq
Chromatin endogenous cleavage sequencing
ChIC
Chromatin immunocleavage
ChIL
Chromatin integration labeling
ChIP
Chromatin immunoprecipitation
ChIP-DIP
Chromatin immunoprecipitation done in parallel
CNV
Copy number variant
Co-ChIP
Sequential chromatin immunoprecipitation
CoTECH
Combined assay of transcriptome and enriched chromatin binding
CUTAC
Cleavage under targeted accessible chromatin
CUT&RUN
Cleavage under targets and release using nuclease
CUT&Tag
Cleavage under targets and tagmentation
Dam
DNA adenine methytransferase
DamID
DNA adenine methytransferase identification
Double ChIP
Double chromatin immunoprecipitation
ESC
Embryonic stem cell
FACS
Fluorescence-activated cell sorting
FRiP
Fraction of reads in peak
IgG
Immunoglobulin-G
LMP
Ligation-mediated PCR
MAbID
Multiplexing antibodies by barcode identification
MNase
Micrococcal nuclease
MulTI-Tag
Multiple target identification by tagmentation
mRNA
Messenger RNA
Nano-CT
Nanobody-based single-cell cleavage under targets and tagmentation
NTT-seq
Nanobody-tethered transposition followed by sequencing
pA
Protein A
pA-MNase
Protein A and micrococcal nuclease fusion protein
pA-Tn5
Protein A and Tn5 transposase fusion protein
PCR
Polymerase chain reaction
PIN*POINT
Protein position identification with nuclease tail
PIP-seq
Particle-templated instant partition sequencing
POI
Protein of interest
Pol2S5p
RNA polymerase II with phosphoserine-5
PTM
Posttranslational modification
reChIP
Sequential chromatin immunoprecipitation
scChIC-seq
Single-cell chromatin immunocleavage followed by sequencing
scChIP
Single-cell chromatin immunoprecipitation
scChIX-seq
Single-cell chromatin immunocleavage and unmixing sequencing
scCUT&Tag
Single-cell cleavage under targets and tagmentation
scCUT&Tag2for1
Single-cell cleavage under targets and tagmentation two for one
scCUT&Tag-pro
Single-cell cleavage under targets and tagmentation with cell surface proteins
scDam&T-seq
Single-cell DNA adenine methyltransferase and transcriptome sequencing
scGET-seq
Single-cell genome and epigenome by transposases sequencing
scMulti-CUT&Tag
Single-cell multi cleavage under targets and tagmentation
scPCOR-seq
Single-cell profiling of chromatin occupancy and RNAs sequencing
scRNA-seq
Single-cell RNA sequencing
seq-ChIP
Sequential chromatin immunoprecipitation
SET-seq
Same cell epigenome and transcriptome sequencing
SHARE-seq
Simultaneous high-throughput ATAC and RNA expression with sequencing
SNV
Single-nucleotide variant
sortChIC
Sort-assisted single-cell chromatin immunocleavage
SpDamID
Split DNA adenine methyltransferase identification
TAM-ChIP
Transposase-assisted chromatin multiplex immunoprecipitation
TnH
Tn5 hybrid transposase
TIRF
Total internal reflection
TSCs
Trophoblast stem cells
uCoTargetX
Ultrahigh-throughput combined tagmenting enrichment for multiple epigenetic proteins in the same cells and transcriptome

Introduction

Regulation of gene expression involves the complex and coordinated integration of numerous factors including chromatin architecture, regulatory elements, histone, RNA and DNA modifications, as well as proteins interacting with DNA and/or chromatin. Eukaryotic DNA is packaged into nucleosomes whereby 147 bases of DNA are wrapped around a histone protein octamer containing two copies each of histone H2A, H2B, H3, and H4 (Luger et al, 1997). Histones carry many posttranslational modifications (PTMs), both within their core and also on the tails protruding from the nucleosome (Turner, 1993, 2000; Strahl and Allis, 2000; Jenuwein and Allis, 2001; Zhao and Garcia, 2015). Advances in proteomic technologies and chromatin research have enabled the identification of novel histone PTMs, with currently a growing catalog of more than 25 different types of PTMs and over a hundred distinct possible sites for modifications, considerably extending the so-called “histone code” (Turner, 1993, 2000; Strahl and Allis, 2000; Jenuwein and Allis, 2001; Zhao and Garcia, 2015; Millán-Zambrano et al, 2022). These modifications can affect chromatin structure directly or determine the binding of effector proteins (readers) that can facilitate DNA-templated processes (Millán-Zambrano et al, 2022; Lukauskas et al, 2024; Policarpi et al, 2024). Importantly, mass spectrometry-based methods and sequential chromatin immunoprecipitation (ChIP) experiments have identified combinatorial occurrence (or co-occurrence) of histone PTMs, giving rise to a vast space for combinatorial modification states that could alter, for example, binding affinities for reader proteins (Bernstein et al, 2006; Sidoli et al, 2012; Voigt et al, 2012; Schwämmle et al, 2014; Bai et al, 2018; Janssen et al, 2019; Lu et al, 2021). Of note, histone PTMs are only one example out of many chromatin components that can display co-occurrence, as also DNA- and chromatin-binding proteins, as well as modifications of DNA, can co-occur and potentially act in concert.
In general, the concept of genomic co-occurrence as we use it here, involves the presence of multiple chromatin components. The co-occurrence of histone PTMs, DNA-binding proteins, and/or chromatin-binding proteins is the focus of this review, referred to as “chromatin features”, and is thought to provide functional synergy, and enable the fine-tuning of transcriptional outputs (Goudarzi et al, 2016; Gao et al, 2021; Policarpi et al, 2024). Importantly, the level of genomic co-occurrence that can be detected by specific approaches is dependent on their resolution, and determines which biological questions can be addressed (see Fig. 1A). Specifically, we define spatial co-occurrence as co-occurring histone PTMs or co-occupying proteins at the same genomic coordinates, irrespective of whether this occurs within the same cell. In contrast, cellular co-occurrence indicates co-occurrence within the same cell. Cellular co-occurrence may take place on two different alleles, or within the same allele (allelic co-occurrence). Co-occurrence can also occur within a single nucleosome (nucleosomal co-occurrence). Lastly, multiple histone PTMs may be present within a single nucleosome either on two different copies of a histone protein, or on the same histone protein (histone co-occurrence). Overall, these different levels of genomic co-occurrence can reflect important information on the functional cooperativity of chromatin features that may result in varying transcriptional outputs within a complex biological context. However, current challenges to study the different levels of co-occurrence high-throughput or genome-wide limit our understanding of the functional relevances of the co-occurrences of chromatin features (Voigt et al, 2012; Sidoli and Garcia, 2015).
Figure 1. Genomic co-occurrence of chromatin features.
(A) Different levels of genomic co-occurrence. Spatial co-occurrence refers to the combinatorial presence of multiple chromatin features at the same genomic coordinates irrespective of whether this occurs within the same cell. Co-occurrence can also take place within the same cell (cellular co-occurence), on different alleles or on the same allele (allelic co-occurrence). Co-occurrence can occur within a single nucleosome (nucleosomal co-occurrence), and for histone PTMs either on different histone proteins in the same nucleosome or on the same histone protein (histone co-occurrence). (B) Spatial versus cellular co-occurrence and in silico inference from bulk profiling. Schematic representation of spatial co-occurrence where different chromatin features are found at the same genomic sites but in different cells (upper left panel), versus cellular co-occurrence where multiple chromatin features are found at a specific locus within the same single cell (upper right panel, with sites of cellular co-occurrence highlighted in dark blue). Note the corresponding enrichment signals derived from bulk experiments (lower panel).
Methods to map the genome-wide distribution of DNA-binding proteins, chromatin proteins, or histone PTMs have provided valuable insights into gene regulatory networks. Thus far, such approaches have largely focused on profiling individual targets in bulk samples. Integration of multiple datasets from bulk genome-wide mapping studies can provide hints on potential sites of spatial co-occurrence by identifying genomic regions where chromatin features are enriched in distinct datasets (Fig. 1B). Importantly, collaborative efforts such as the ENCODE project have built a useful resource to obtain datasets of an increasing number of targets, from cell lines and tissues, from different source organisms, generated with standardized procedures, that can serve for in silico analysis of genomic co-occurrence of chromatin factors (Dunham et al, 2012; Akbarian et al, 2015; Abascal et al, 2020; Partridge et al, 2020). However, the co-occurrence inferred from such in silico comparative analysis cannot distinguish between overlapping chromatin features that are due to alternative binding events in different population of cells (spatial co-occurrence) rather than “true” co-occurrence within the same cell (cellular co-occurrence; Fig. 1B).
To obtain a better understanding of the cooperative function of chromatin features, methods that can simultaneously profile the cellular co-occurrence of multiple histone PTMs and nonhistone proteins are required. Generating precise chromatin state maps with single-cell resolution will allow to unambiguously study the multifactorial nature of gene expression regulation. The past decade has been marked by a growing toolkit of newly developed single-cell and spatial multi-omics methodologies, and some of these methods have been recently reviewed in (Vandereyken et al, 2023). It is only within the last 3 years that ground-breaking single-cell -omics advances now permit to profile the cellular co-occurrence of multiple chromatin features. In this context, we cover the underlying methodological principles and provide perspectives on advances to allow for the simultaneous multi-mapping of chromatin features in low-input samples and in particular in single cells. We compare and contrast the toolkits that are based on ChIP, as well as those involving enzyme-tethering approaches: targeted DNA adenine methylation tagging, targeted chromatin cleavage, and antibody-tethered tagmentation. We further elaborate on the limitations and challenges associated with these different approaches for profiling the co-occurrence of chromatin features, and the considerations to account for based on the characteristic features of the currently established toolkits. Finally, we provide perspectives on the challenges and limitations that still have to be overcome moving forward. This review provides background on the underlying principles of such approaches and can help researchers in the planning of future experiments to determine the most appropriate method to pursue for multifactorial profiling in a complex biological context.

Section 1: ChIP-based methods and related approaches

Early methodological milestones attempting to map DNA-protein contacts date 40 years back, with UV crosslinking-immunoprecipitation methods, formaldehyde-pronase techniques, and formaldehyde-antibody approaches (Fig. 2A-1) (Gilmour and Lis, 1984, 1985, 1986; Solomon and Varshavsky, 1985; Hebbes et al, 1988; Solomon et al, 1988). Since then, ChIP has been widely used to map the binding profiles of DNA-binding proteins, chromatin proteins and histone PTMs in vivo and has been for many years the gold standard method for this purpose. ChIP can be performed with crosslinking to preserve protein–DNA interactions (X-ChIP) or alternatively without a crosslinking step under native conditions (N-ChIP). Subsequently, the chromatin is fragmented by sonication or enzymatic digestion (Skene and Henikoff, 2015). The sheared chromatin is then incubated with an antibody specific to a protein or histone PTM of interest to immunoprecipitate and enrich for the targeted proteins together with the DNA fragments they are binding to. Next, crosslinks are reversed, and the bound DNA fragments are purified. To determine the sequences enriched with a specific protein following the ChIP, hybridization assays (Hebbes et al, 1988; Dedon et al, 1991; Orlando et al, 1997), polymerase chain reaction (PCR) with primers specific to genomic regions (ChIP-qPCR), and microarray hybridization as a global mapping strategy (ChIP-chip, (Ren et al, 2000; Boyer et al, 2005; Kim et al, 2005a, 2005b; Lee et al, 2006), can be used. ChIP combined with next-generation sequencing (NGS; ChIP-seq) allows now to assay the enrichment of proteins genome-wide with base-pair resolution (Barski et al, 2007; Mikkelsen et al, 2007; Park, 2009; Furey, 2012).
Figure 2. ChIP-based methods and enzyme-tethering approaches.
Overview of ChIP-based methods (A), targeted DNA adenine methylation-based approaches (B), and targeted DNA cleavage-based techniques (C), to profile genomic sites of co-occurring chromatin features. For further details on the methods, see main text. (A) ChIP-based methods. (1) Formaldehyde-antibody method (Hebbes et al, 1988; Solomon et al, 1988). Chromatin fragments are subjected to immunoprecipitation using antibodies specific for a protein of interest. (2) Sequential ChIP and variations (Chaya et al, 2001). Chromatin fragments are subjected to a first immunoprecipitation using antibodies specific to a chromatin feature of interest, followed by a second immunoprecipitation step targeting another feature to enrich for fragments with both features. (3) Single-molecule combinatorial nucleosome profiling (Shema et al, 2016). Chromatin is digested by MNase and mononucleosomes are ligated to fluorescent adapters and captured on slides. Total internal reflection (TIRF) microscopy is used to image nucleosome positions. Following the release of the fluorophores, fluorescently labeled antibodies targeting histone PTMs are applied to the slides and antibody binding events are recorded. DNA sequence is determined by NGS. (4) ChIP-DIP (preprint: Perez et al, 2023). Streptavidin beads are bound with biotinylated antibody-specific barcoded adapters, and with biotinylated protein G beads bound with antibodies, to create a bead pool. Following crosslinking and cell lysis, the bead pool is applied to perform ChIP. A total of five rounds of split-pool indexing can assign the genomic DNA sequences to their associated antibodies. (B) DamID-based methods. (1) DamID (Steensel and Henikoff 2000). Dam is tethered by its fusion partner to the genomic binding sites of a protein of interest (P1) and methylates adenines within nearby GATC motifs. (2) scDam&T (Rooijers et al, 2019). An additional reverse transcription step allows to jointly measure gene expression and profile a chromatin protein in single cells. (3) SpDamID (Hass et al, 2015). Two fusion proteins are generated, consisting of two nonhistone chromatin proteins (P1 and P2) and two halves of Dam (D + AM). Upon the reconstitution of two halves of Dam, Dam methylates adenines in GATC motifs in the vicinity of the binding sites. (4) EpiDamID (Rang et al, 2022). A fusion protein consisting of Dam and a chromatin reader domain is used to tether Dam near genomic sites associated with a histone PTM of interest. (C) Chromatin cleavage-based methods. (1) PIN*POINT (Lee et al, 1998). A fusion protein consisting of a protein of interest (POI) and a nuclease is used to tether the cleavage of DNA at the genomic target sites of the POI. (2) ChEC (Schmid et al, 2004). A fusion protein composed of a POI and a micrococcal nuclease (MNase) is generated to direct DNA cleavage at the POI binding sites in the genome. (3) ChIC, CUT&RUN and uliCUT&RUN (Schmid et al, 2004; Skene and Henikoff, 2017; Hainer et al, 2019). Samples are successively treated with antibodies targeting a chromatin protein of interest and a protein A and MNase (pA-MNase) fusion that will cleave and release DNA fragments bound by the targeted protein. (4) scChIC-seq and sortChIC (Ku et al, 2019; Zeller et al, 2023). Cells are treated with antibodies and pA-MNase and then single cells are distributed on a plate using fluorescence-activated cell sorting (FACS). ChIC is performed in each individual well. (5) scPCOR-seq (Pan et al, 2022). In addition to the scChIC-seq/sortChIC workflow, a reverse transcription step captures simultaneously mRNAs. (6) scChIX-seq (Yeung et al, 2023). Three separate sortChIC maps are generated by using two distinct antibodies separately and by using both antibodies simultaneously. The two first maps are used to assign the fragments to their respective target.
Several variations of ChIP have been developed to assess whether two proteins (or histone PTMs) associate with the same genomic binding site in vivo, such as sequential ChIP (seq-ChIP), reChIP, co-ChIP, double ChIP, chromatin double immunoprecipitation (ChDIP), and others (Fig. 2A-2) (Chaya et al, 2001; Geisberg and Struhl, 2004; Henry et al, 2003; IJpenberg et al, 2004; Métivier et al, 2003; Proft and Struhl, 2002; Scully et al, 2000; Soutoglou and Talianidis, 2002). Using mononucleosomes as input, these methods involve two sequential rounds of immunoprecipitation: a first immunoprecipitation of protein–DNA complexes with a first antibody, followed by a second one with an antibody against a different target, thereby enriching for DNA bound by both targets. These different sequential ChIP variations allow to determine allelic or nucleosomal co-occurrence genome-wide (Kinkley et al, 2016; Weiner et al, 2016). While ChIP-seq-based assays are amenable to ultra-low-input samples and single-cell profiling (Brind’Amour et al, 2015; Rotem et al, 2015; Grosselin et al, 2019), and reChIP-seq has been recently adapted for low-input samples (Seneviratne et al, 2024), to our knowledge, further optimizations would have to be achieved to apply seq-ChIP, reChIP-seq, or co-ChIP-seq, in single cells.
Sequential ChIP has led to the important discovery that activating and repressing histone PTMs, namely histone H3 lysine 4 and lysine 27 tri-methylation (H3K4me3 and H3K27me3, respectively), display nucleosomal co-occurrence at transcription start sites (TSSs) of key developmental transcription factor genes in mouse embryonic stem cells (mESCs) and primary human CD4+ memory T cells (Bernstein et al, 2006; Kinkley et al, 2016). These large H3K27me3-enriched regions harboring smaller H3K4me3 peaks were termed bivalent domains (Azuara et al, 2006; Voigt et al, 2012; Harikumar and Meshorer, 2015; Blanco et al, 2020; Kumar et al, 2021). Importantly, it was shown that ~15% of TSSs showing co-occurring H3K4me3 and H3K27me3 as detected by in silico dataset comparison were not detected by reChIP-seq and are thus regions of potential spatial co-occurrence rather than cellular co-occurrence (Kinkley et al, 2016). The presence of such bivalent domains was further confirmed using single-molecule combinatorial profiling of modified mononucleosomes (Fig. 2A-3) (Shema et al, 2016). This method elegantly makes use of fluorescently labeled antibodies, total internal reflection (TIRF) microscopy, and sequencing reactions to record the modification states, position, and sequence of mononucleosomes (Shema et al, 2016). In brief, the chromatin is first digested with micrococcal nuclease (MNase). Mononucleosomes are then ligated with fluorescent and biotinylated adapters, isolated, and captured on slides. Next, TIRF microscopy is used to record the nucleosome positions on the slide, as well as binding and dissociation events upon incubation with fluorescently labeled antibodies specific to histone PTMs. Finally, to determine the genomic positions of the imaged modified nucleosomes, primers are hybridized onto the biotinylated adapters and sequencing-by-synthesis is performed at each position on the slide. Applying this method to isolated histone proteins from ESCs allowed to determine the proportion of bivalent nucleosomes that bear the two modifications of interest on the same histone (Shema et al, 2016). As such, a unique feature of this approach is that it can be used to detect not only nucleosomal, but also histone co-occurrence of PTMs. However, the workflow is complex and requires specialized equipment. Further optimizations will be needed to profile low-input samples or single cells in high-throughput approaches.
Finally, a novel method based on ChIP involves a highly scalable and multiplexed platform that enables the genome-wide mapping of hundreds of proteins simultaneously (Fig. 2A-4) (preprint: Perez et al, 2023). Named chromatin immunoprecipitation done in parallel (ChIP-DIP), this technique relies on a bead labeling workflow whereby streptavidin beads are conjugated with (1) antibody-assigned barcoded and biotinylated adapters and (2) biotinylated protein G beads. In a next step, antibodies are coupled to the corresponding barcoded streptavidin–protein G bead complex by binding to protein G. A bead complex pool is generated with various antibodies and their corresponding barcoded adapters, and incubated together with fixed cells for immunoprecipitation in a single tube. Subsequently, multiple rounds of split-and-pool tagging are performed to generate a unique combinatorial index for each streptavidin bead (Quinodoz et al, 2018, 2021, 2022; preprint: Perez et al, 2023). This split-pool barcode is shared between genomic DNA and antibody-specific oligonucleotide bound to the same bead, and serves to assign the epitope barcode to its associated genomic regions. With this technique, the authors were able to generate what they qualify as “consortium level” datasets, by multiplexing over 225 antibodies specific for 160 distinct proteins in a single experiment, with target proteins encompassing histone PTMs, chromatin regulators (readers, writers, and erasers), transcription factors, and RNA polymerases (I, II including different PTM forms, and III) (preprint: Perez et al, 2023). This highly multiplexed approach shows the potential to identify regulatory features based on the combinatorial occurrence of histone PTMs. As ChIP-DIP is performed in bulk, and mapping the targets involves the matching of combinatorial barcodes from a single antibody and a genomic DNA sequence, it allows to interrogate spatial co-occurrence only.

Section 2: Targeted DNA adenine methylation-based methods

An alternative suite of methods to map DNA-protein interactions is based on the DNA adenine methyltransferase (Dam), an E. coli enzyme that endogenously methylates adenine in a GATC context (Steensel and Henikoff 2000; Greil et al, 2006). Because adenine methylation (m6A) in DNA is absent in most eukaryotic cells, Dam can be used to mark regions in the genome with m6A and probe the genomic occupancy of proteins in vivo. This approach is referred to as Dam identification (DamID, Fig. 2B-1) (Steensel and Henikoff 2000; Greil et al, 2006). For this purpose, Dam is fused with a protein of interest (POI), and expressed in cells, resulting in adenine methylation on the DNA surrounding the binding sites of the POI. To determine the sites of methylation, the genomic DNA is digested by either restriction enzymes DpnI or DpnII, which specifically cleave methylated or unmethylated adenine sites, respectively, at the 5’ end of a GATC motif. The digested genomic DNA is then subjected to adapter ligation followed by adapter-mediated PCR (LMP), where DNA fragments can be amplified by PCR proportionally to the methylation frequency (Dai et al, 2000). The resulting libraries can either be used for targeted interrogation of cooperative binding at specific genomic regions with PCR or with NGS for genome-wide mapping.
DamID was later on adapted for single-cell profiling (scDamID), by sorting and distributing single cells on a 96-well plate for cell lysis and adding well-specific indexes for single-cell DamID multiplexing (Kind et al, 2015). To assess how protein-genome interactions influence gene expression in the same cell, a DamID-based method was introduced for the joint profiling of protein occupancy and messenger RNA (mRNA) expression, termed single-cell DamID with messenger RNA sequencing (scDam&T-seq, Fig. 2B-2) (Rooijers et al, 2019). Similarly to the scDamID approach, single cells sorted with fluorescence-activated cell sorting (FACS) are distributed to 384-well plates for greater throughput. To capture RNA in the same cell, polyadenylated mRNA molecules are subjected to reverse transcription, and both genomic DNA and resulting complementary DNA (cDNA) products are amplified by in vitro transcription (Rooijers et al, 2019). scDam&T was, for example, successfully applied to simultaneously profile in single cells the transcriptome and the occupancy of the PRC1 subunit RING1B. Correlation analysis of these two modalities revealed allelic bias reflecting X chromosome inactivation (Rooijers et al, 2019).
To leverage DamID and study combinatorial protein binding to chromatin, Split DamID (SpDamID, Fig. 2B-3) was developed as a protein complementation version of DamID (Hass et al, 2015). In brief, complementary halves of Dam are fused to two potentially interacting or juxtapositioned DNA-binding or chromatin proteins. If both proteins are in close proximity, a functional Dam will be reconstituted, resulting in the methylation of nearby adenines within GATC sequences. SpDamID was successfully used on as little as 100 cells but has yet to be adapted for single-cell interrogation. SpDamID has, for example, allowed to identify genomic sites of allelic co-occurrence of Notch1 and its binding partners, as well as the co-occurrence of non-interacting proteins (for example, Notch1 and Runx1 both binding to the same enhancer) (Hass et al, 2015).
A general limitation of the described DamID-based methods is that they are not directly suitable to map modified histones. To address this, EpiDamID extends the suite of DamID-based protocols (Fig. 2B-4), allowing to profile histone PTMs along with the transcriptome, in low-input samples and in single cells (Gopalan and Fazzio, 2022b; Rang et al, 2022). For this, Dam was fused to chromatin-binding molecules that can detect various histone PTMs. Three different Dam-fusion types were generated with distinct targeting domains, including (1) full-length nonhistone proteins, (2) histone modification binding domains, or (3) single-chain variable fragments, a recombinant antibody construct comprised of the variable regions of the heavy and light immunoglobulin chains connected via a peptide linker (Bird et al, 1988; Ahmad et al, 2012; Xenaki et al, 2017; Rang et al, 2022). This approach was applied for various histone PTMs, encompassing active, heterochromatin, and Polycomb marks. While the authors state that EpiDamID can be applied to any existing DamID protocol, it has yet to be adapted to study co-occurring histone PTMs (Hass et al, 2015; Rang et al, 2022). Recently, an alternative method based on the deposition of methylation marks on adenines was developed, named directed methylation with long-read sequencing (DiMeLo-seq) (Altemose et al, 2022). DiMeLo-seq makes use of a protein A (pA)–Hia5 fusion (pA-Hia5) and tethers pA-Hia5 to a POI or histone PTM via specific antibodies. Hia5 is a nonspecific deoxyadenosine methyltransferase, thus marking binding sites of the targeted POI. This approach has also yet to be adapted to study the co-occurrence of histone PTMs, DNA-binding proteins, or chromatin proteins.

Section 3: Targeted DNA cleavage-based approaches

An early approach to direct a nuclease to cleave DNA-binding sites of a POI was named Protein Position Identification with Nuclease Tail (PIN*POINT, Fig. 2C-1) (Lee et al, 1998). With PIN*POINT, a fusion protein comprised of a non-sequence-specific nuclease domain and a POI is expressed in vivo. The nuclease is recruited by the fusion partner to chromatin and digests the DNA specifically near the POI binding sites (Dai et al, 2000). Subsequently, two methods making use of MNase were simultaneously introduced using different tethering approaches to direct DNA cleavage, namely chromatin immunocleavage (ChIC) and chromatin endogenous cleavage (ChEC) (Schmid et al, 2004).
With ChEC (Fig. 2C-2), a POI is fused with MNase and (over)expressed, directing the MNase-induced DNA cleavage near the protein binding sites. ChEC was developed for both fixed cells (termed in vitro ChEC) and for permeabilized cells in native conditions (termed in vivo ChEC). ChIC (Fig. 2C-3) is based on the fusion of immunoglobulin binding domains from the staphylococcal protein A (pA) to MNase, which is used to tether the nuclease to antibodies (Schmid et al, 2004). Specifically, fixed cells were first treated with primary antibodies specific for an epitope of interest, optionally followed by secondary antibodies, and ultimately with the pA-MNase fusion protein (with pA recognizing the primary or secondary antibody) to induce double-strand DNA cleavage in the vicinity of the target protein binding sites. Later, these methods were combined with NGS to allow for genome-wide mapping of protein binding sites, giving rise to the CUT&RUN technique (for Cleavage under targets and release under nuclease, Fig. 2C-3) and ChEC-seq, respectively (Zentner et al, 2015; Skene and Henikoff, 2017).
CUT&RUN rapidly became adopted by the research community as an efficient alternative to ChIP-based methods given its improved applicability for low-input samples (Skene and Henikoff, 2017). CUT&RUN is similar to ChIC (Fig. 2C-3): it involves immobilization of nuclei on magnetic beads and antibody-directed pA-MNase target cleavage. Chromatin fragments bound to the antibody target are cut and diffuse out of the nucleus. The released DNA can be extracted for subsequent library preparation and sequencing. It was later on further adapted to profile as little as 10 cells per experiment with ultra-low-input CUT&RUN (uliCUT&RUN, Fig. 2C-3) (Hainer et al, 2019). To achieve single-cell profiling with single-cell ChIC-seq and sort-assisted single-cell ChIC (scChIC-seq and sortChIC, respectively, Fig. 2C-4), single cells are sorted onto 384-well plates by FACS to perform the DNA cleavage reaction in individual wells, followed by fragment indexing with cell-specific barcodes, before pooling for amplification of the libraries (Ku et al, 2019; Zeller et al, 2023). Furthermore, the addition of an in situ reverse transcription step has provided the possibility to capture mRNAs from their poly-A tail in the same cell to jointly measure gene expression, named single-cell profiling of chromatin occupancy and RNA sequencing (scPCOR-seq, Fig. 2C-5) (Pan et al, 2022).
To allow for the multiplexing of target histone PTMs based on the ChIC principle, a strategy has been developed that involves an experimental and bioinformatics framework named single-cell chromatin immunocleavage and unmixing sequencing (scChIX-seq, Fig. 2C-6) (Yeung et al, 2023). In scChIX-seq, three sortChIC maps are generated: two maps from two distinct histone PTMs resulting from incubating each antibody separately, and one map resulting from double-antibody incubation targeting both histone PTMs simultaneous (Yeung et al, 2023; Zeller et al, 2023). The individual histone PTM profiles are used as training sets to deconvolve the multiplexed profile dataset. Consequently, the modeling allows to infer histone PTM profiles in single cells for both mutually exclusive sites of enrichment as well as sites of co-occurrence (Yeung et al, 2023). In addition, because the scChIX-seq workflow includes a FACS step, it is possible to purify samples based on cell types (Ku et al, 2019; Yeung et al, 2023; Zeller et al, 2023). By studying different pairs of either mutually exclusive or co-occurring PTMs, the scChIX-seq framework allowed to define cell-specific histone PTM landscapes (H3K27me3–H3K9me3), that distinguish cell types with more confidence than based on mRNA abundances alone (H3K4me1–H3K27me3), quantify overlapping modifications (H3K36me3–H3K9me3) across cell types during organogenesis, and apply pseudotime analysis to infer chromatin velocity throughout an in vitro macrophage differentiation timecourse (H3K36me3–H3K4me1) (Yeung et al, 2023). While scChIX-seq makes it possible to deconvolve multiplexed histone PTM profiles, it has a trade-off of a limited spatial resolution and thus on the applicability to study cellular co-occurrence.

Section 4: Targeted tagmentation-based methods and related approaches

During the past few years, methods making use of the Tn5 transposase for epigenomic applications have rapidly evolved, particularly for single-cell profiling. These toolkits are largely based on chromatin under targets and tagmentation (CUT&Tag, Fig. 3-1), antibody-guided chromatin tagmentation (ACT-seq), and combinatorial barcoding and targeted chromatin release (CoBATCH) (Carter et al, 2019; Kaya-Okur et al, 2019; Wang et al, 2019). CUT&Tag was initially developed for low-input and single-cell approaches, and showed utility for profiling histone PTMs, RNA Polymerase II, and transcription factors (Kaya-Okur et al, 2019). The basic principle involves the fusion of protein A with an hyperactive Tn5 transposase (pA-Tn5) that is loaded with sequencing adapters, termed transposome, and tethered with antibodies (that bind to pA) against a target of interest (Reznikoff, 2003; Picelli et al, 2014). The Tn5-catalyzed targeted tagmentation reaction is activated with the addition of magnesium ions which causes the integration of adapters into the DNA at antibody-bound regions, generating sequencing-ready libraries within a day of wet lab work (Kaya-Okur et al, 2019). The protocol was originally implemented to profile from 100,000 cells and down to 60 cells in bulk. Using a nano-dispensing system to distribute cells on a 5184-nanowell chip, and indexed primers, epigenomic profiling with CUT&Tag was also achieved at single-cell resolution (Kaya-Okur et al, 2019). With variations in the tagmentation reaction conditions, the transposome can be redirected to generate chromatin accessibility profiles in a single tube and even on a home workbench, referred to as Cleavage Under Targeted Accessible Chromatin (CUTAC) (Henikoff et al, 2020, 2021). Single-cell CUT&Tag (scCUT&Tag) was later on adapted in combination with the droplet-based 10x Genomics platform with optimization for complex tissues (Bartosovic et al, 2021). In addition, scCUT&Tag was recently combined with cell surface protein abundance measurement (scCUT&Tag-pro) (Zhang et al, 2022). In the same article, a computational framework termed single-cell ChromHMM (scChromHMM) was also introduced, allowing to integrate scCUT&Tag-pro datasets of six distinct histone PTMs, and predict chromatin state annotations at single-cell resolution (Zhang et al, 2022). This approach further highlights the added benefit of integrating multiple datasets for chromatin state inference, but still lacks direct detection of chromatin feature co-occurrence.
Figure 3. Tagmentation-based methods and related approaches.
Overview of tagmentation-based approaches and related methods to study co-occurrence of chromatin features. For further details on the methods, see main text. (1) scCUT&Tag (Kaya-Okur et al, 2019). An antibody specific for a target of interest plus a secondary antibody tether and enrich the genomic region bound with pA-Tn5 transposomes that insert sequencing adapters upon activation. (2) scMulti-CUT&Tag (Gopalan et al, 2021). Antibodies coupled with pA-Tn5 complexes preloaded with sequencing adapters containing antibody-specific barcodes are tethered to the targeted epitopes. Antibody-specific barcoded adapters are inserted at the targeted genomic sites. (3) scGET-seq (Tedesco et al, 2022). Tn5 transposase and a protein fusion (TnH) composed of Tn5 with the chromodomain (CD) of HP1α are both preloaded with specific barcoded adapters. Both Tn5 and TnH are applied to cells in order to simultaneously profile open and closed chromatin. (4) scCUT&Tag2for1 (Janssens et al, 2022). Antibodies specific for RNA polymerase II with phosphoserine-5 (RNApol2S5p) and for H3K27me3 are applied to samples to tether pA-Tn5 to antibody-bound genomic regions. The tagmentation densities and fragment sizes are used to assign reads to either target. (5) nano-CT and NTT-seq (Bartosovic & Castelo-Branco, 2022; Stuart et al, 2022). Fusion proteins composed of Tn5 and a single-chain secondary nanobody that specifically recognize primary antibodies raised in different species are used to tether Tn5 to primary antibodies bound to targets of interest and insert antibody-specific barcoded adapters. (6) MulTI-Tag (Meers et al, 2023). Antibody-specific barcoded adapters are covalently conjugated to antibodies and loaded to pA-Tn5 directing the transposome to integrate barcodes at genomic binding sites of targeted proteins. (7) MAbID (Lochs et al, 2023). Antibodies specific to chromatin proteins of interest are covalently conjugated to barcoded adapters bearing motifs recognized by different sets of restriction enzymes to cleave both adapters and nearby genomic sites followed by ligation to integrate antibody-specific barcodes. (8) uCoTargetX (Xiong et al, 2024). Primary antibodies specific for different chromatin targets are coupled with pA-Tn5 that are preloaded with antibody-specific barcodes and are used to introduce barcodes and forward sequencing adapters. Secondary antibodies coupled with pA-Tn5 preloaded with reverse sequencing adapters are applied to further tagment genomic regions.
The first method that was described to directly profile multiple targets based on the CUT&Tag procedure was multi-CUT&Tag (Fig. 3-2) (Gopalan et al, 2021; Gopalan and Fazzio, 2022a). This method uses a histidine-tagged version of the pA-Tn5 fusion that allows to purify the transposome complexes pre-coupled with antibodies specific for the epitopes of interest. To incorporate antibody-specific barcodes during tagmentation, the pA-Tn5 fusion proteins pre-coupled with antibodies are also preloaded with antibody-specific barcodes flanked by transposase-compatible adapters and sequences suitable for NGS for the production of sequencing-ready libraries. Sequencing reads can then be demultiplexed based on the antibody-assigned barcodes to determine binding profiles of each target, while simultaneously defining sites of nucleosomal co-occurrence. This approach has also been adapted for examination in single cells with the 10x Genomics platform (scMulti-CUT&Tag) (Gopalan et al, 2021; Gopalan and Fazzio, 2022a). Unbiased clustering performed with the simultaneously profiled H3K27me3 and H3K27ac marks enhanced the identification of distinct cell types from a mixture of ESCs and trophoblast stem cells (TSCs) (Gopalan et al, 2021).
To jointly probe open and closed chromatin states in the same cell, single-cell genome and epigenome by transposases sequencing (scGET-seq, Fig. 3-3) was developed (Tedesco et al, 2022). This tagmentation-based method makes use of a hybrid transposase (TnH) where Tn5 is fused with the chromodomain (CD) of the heterochromatin protein-1alpha (HP1α), a reader for H3K9me3. By sequentially treating permeabilized cells with Tn5 and TnH loaded with two different sets of barcoded indexes, the authors were able to probe accessible and compacted chromatin (Tedesco et al, 2022). This approach was adapted to achieve single-cell resolution by combining with the 10x Genomics droplet-based protocol, and has the ability to also call copy number variant (CNV) events as well as single-nucleotide variants (SNVs). Another alternative to jointly profile open and closed chromatin is single-cell CUT&Tag2for1 (scCUT&Tag2for1, Fig. 3-4), which extends from CUTAC (Henikoff et al, 2020, 2021; Janssens et al, 2022). Here, the Tn5 transposase is directed to antibodies specific for RNA polymerase II with phosphoserine-5 (Pol2S5p) and a “repressive” histone PTM (H3K27me3) with mutually exclusive binding profiles. Sequencing reads are then deconvoluted computationally to generate single-cell maps of both active and repressed chromatin, by accounting for tagmentation densities and fragment length differences. Single-cell profiles are generated with the use of a microfluidic chip to add cell-specific barcodes and amplify libraries. Both scGET-seq and scCUT&Tag2for1 can serve to simultaneously profile open and closed chromatin states and are currently not directly applicable to study the co-occurrence of chromatin features (Janssens et al, 2022; Tedesco et al, 2022).
Two novel and highly similar approaches now employ nanobody-tethered transposases to profile up to three modalities in bulk or single cells with starting material ranging from 25 K to 1 M cells, namely the nanobody-CUT&Tag (nano-CT, Fig. 3-5) and the nanobody-tethered transposition followed by sequencing (NTT-seq, Fig. 3-5) (Bartosovic and Castelo-Branco, 2022; Stuart et al, 2022). These methods make use of several sets of recombinant fusion proteins that are composed of the Tn5 transposase and nanobodies (nano-Tn5). Nanobodies are small single polypeptide chain antibodies that can strongly and stably bind to their target over a range of temperature and pH conditions. The engineered nano-Tn5 are made specific for a set of immunoglobulins from different species or for different IgG subtypes directed against the epitope of interest. Similarly to scMulti-CUT&Tag and scGET-seq, the transposases are loaded with different sets of barcoded adapters for the corresponding antibody/species and therefore insertion events originating from the different nanobody-Tn5-antibody complexes can be demultiplexed based on these barcoded sequences. Assaying combinations of chromatin features enabled improved identification of cell types (Stuart et al, 2022). In addition, chromatin remodeling events were captured across the differentiation progression of human bone marrow cells (Stuart et al, 2022) and mouse brain oligodendrocyte cells (Bartosovic and Castelo-Branco, 2022) through profiling of H3K27ac and H3K27me3. The multimodal feature of these approaches allows to identify the trajectory of differentiating cells with their corresponding chromatin states, as well as additional chromatin state dynamics. Specifically, combining multimodal chromatin profiling by nano-CT and chromatin velocity modeling from differentiating cells in the mouse brain (also including ATAC signal along with H3K27ac and H3K27me3) was used to infer differentiation kinetics, and to identify genes that regulate the chromatin landscape (Bartosovic and Castelo-Branco, 2022).
To circumvent potential cross-contamination of antibody-specific adapters, an alternative approach made use of the principle of chromatin integration labeling (ChIL), where pA-Tn5 is coupled with antibodies that are covalently conjugated with antibody-specific adapters (termed ChIL probe). This approach combined immunostaining, transposase tagging, followed by linear amplification, for profiling up to three targets simultaneously (Schmidl et al, 2015; Harada et al, 2019; Handa et al, 2020; Meers et al, 2023). With Multiple Target Identification by Tagmentation (MulTI-Tag, Fig. 3-6), permeabilized cells are immunostained and tagmented with different ChIL probes for each target sequentially, and a final tagmentation step is carried out with secondary antibodies conjugated with sequences complementary to reverse primers to increase fragments per cell (Meers et al, 2023). To achieve single-cell profiling, samples are first processed in bulk as described above, and the final tagmentation step is performed on a 96-well plate with each well containing transposomes with a unique barcode. Finally, nuclei are pooled back together and then distributed on a microfluidic chip for combinatorial indexing and amplification, generating cell-specific barcode combinations. Analysis of MulTI-Tag maps in H1 human ESCs at different differentiation stages revealed that the integration of data from the multifactorial chromatin states improved developmental trajectories inference (Meers et al, 2023). The authors also investigated the allelic and nucleosomal co-occurrence of different targets, and intriguingly detected sites co-enriched with H3K27me3-H3K36me3 on different nucleosomes but within the same gene (Meers et al, 2023).
Recent advances in single-cell multi-omics technologies have pushed the limits for profiling multiple targets, with the possibility to jointly map up to six epitopes in a single experiment. First, for Multiplexing Antibodies by barcode Identification (MAbID, Fig. 3-7), nuclei are isolated and lightly fixed, treated with primary antibodies with different host origin, incubated with secondary antibodies that are covalently conjugated with barcoded adapters and specific for each primary antibody, and sorted with FACS (Lochs et al, 2023). In lieu of exploiting the Tn5 transposase to tagment the targeted genomic regions, the genome as well as the antibody-assigned adapters are digested with restriction enzymes, generating overhang templates that are compatible for subsequent ligation. Including additional sets of restriction enzymes allowed to increase the combinatorial space for antibody multiplexing. To further increase the number of multiplexed targets in a single experiment and overcome the dependency on antibodies of varied host origins, the authors applied the antibody–adapter conjugation directly to the primary antibodies (Lochs et al, 2023). While this lowered the signal-to-noise ratio, the multiplexing capacity is greatly enhanced, and the simultaneous mapping of six epitopes encompassing all major chromatin types was validated. MAbID was also adapted for the investigation of single cells (scMAbID) by integrating the use of 384-well plates and liquid-handling robotics, and of primary tissues. Similar to the other multimodal methods described here, scMAbID was shown to improve the identification of different cell types with multiplexed measurements in comparison with single profiles. scMAbID was also applied to study allele-specific chromatin dynamics associated with X chromosome inactivation during development (Lochs et al, 2023).
Finally, recent technological advancements allow for the profiling of up to five histone PTMs at single-cell level by two related methods: (ultra)high-throughput Combined TAgmenting enRichment for multiple epiGEneTic proteins in the same cell (uCoTarget), and uCoTargetX, which allows for the simultaneous profiling of the transcriptome in the same cell (Fig. 3-8) (Xiong et al, 2024). This strategy involves the sequential treatment of lightly fixed and permeabilized cells first with pA-Tn5 complexes coupled to different antibodies and preloaded with antibody-assigned barcodes containing a first set of indexed reverse adapter sequences. Cells are next treated with a second set of transposomes coupled to secondary antibodies and preloaded with the second set of indexed forward adapter sequences to facilitate further tagmentation at genomic regions enriched for targeted chromatin proteins and therefore increase the number of fragments generated per cell. For single-cell assays, the authors introduced a split-pool combinatorial indexing strategy into the protocol to generate cell-specific barcoding that vastly expands the scalability of the assay (Rosenberg et al, 2018; Ma et al, 2020). Integrating both epigenomic and transcriptomic modalities improved accuracy for cell type identification.

Section 5: Selecting the method of choice: some pros and cons

The following section compares and contrasts approaches that can simultaneously profile multiple chromatin features genome-wide. We discuss considerations regarding the applicability of the methods, the ease of their implementation, the interpretation of the generated data, and possible additional features. Table EV1 contains a summary of the characteristic features of the relevant methods, along with selected advantages and limitations.

Application

Based on the research question, the methods described here can serve to either interrogate (1) solely sites of co-occurrence, such as with sequential ChIP-based approaches or with SpDamID (Hass et al, 2015; Kinkley et al, 2016; Weiner et al, 2016; Seneviratne et al, 2024), or alternatively (2) obtain the genome-wide occurrence of multiple chromatin features in a given experiment, including the localization of individual features and their colocalization (Gopalan et al, 2021). Most of the mapping approaches described here can map multiple chromatin features in parallel in one experiment, whereas a subset of the methods is based on inferring co-occurrence, either by using available datasets of singly assayed targets (in silico inference), or via unmixing (deconvolution) of reads such as with scChIX-seq (see Table EV1: Deconvolution of target profile) (Yeung et al, 2023). Indeed, in silico inference can be useful to identify candidate proteins or genomic sites, but subsequent validation of cellular co-occurrence is required (Fig. 1B). Likewise, deconvolution of reads originating from multiplexed targets may be more challenging when the assayed targets are not mutually exclusive or show highly similar enrichment profiles on the genome.
The approaches vary in terms of the possible targets studied (see Table EV1: Types of targets assayed). Some methods were, for example, designed to specifically target simultaneously open and closed chromatin such as with scGET-seq and scCUT&Tag2for1 (Janssens et al, 2022; Tedesco et al, 2022). Other approaches may be more suitable to study histone PTMs, such as with single-molecule combinatorial nucleosome profiling and scChIX-seq (Shema et al, 2016; Yeung et al, 2023), whereas certain methods can only profile nonhistone proteins as with SpDamID (Hass et al, 2015). Most ChIP-based and tagmentation-based methods are suitable for assaying both histone PTMs and nonhistone proteins simultaneously.
The multiplexing capacity to assay multiple chromatin features in a single experiment depends on the underlying principle of the approach used (see Table EV1: Number of targets assayed). For example, sequential ChIP-based approaches may not be suitable for more than two targeted chromatin features as increasing the number of immunoprecipitation steps will further reduce the yield and these approaches already require relatively high amounts of starting material (Kinkley et al, 2016; Weiner et al, 2016; Seneviratne et al, 2024). Some methods were specifically designed to assay two targets only, such as those for open and closed chromatin (scGET-seq and scCUT&Tag2for1) (Janssens et al, 2022; Tedesco et al, 2022), or SpDamID that can only profile two nonhistone proteins (Hass et al, 2015). With scChIX-seq, three sortChIC maps are already required to assay two histone PTMs simultaneously (one map from two multiplexed targets, and two separate maps of individual targets for deconvolution) making it experimentally and computationally complex to increase the number of targets assayed (Yeung et al, 2023). Nanobody-tethered tagmentation approaches (nano-CT and NTT-seq) are currently limited to two (plus open chromatin profiling with nano-CT) or three targets assayed in a single experiment, respectively, because the antibodies used need to be raised in different host species (Bartosovic and Castelo-Branco, 2022; Stuart et al, 2022). In general, methods that insert target-specific barcodes are applicable to an increased number of assayed targets, such as multi-CUT&Tag and MulTI-Tag (Gopalan et al, 2021; Gopalan and Fazzio, 2022a; Meers et al, 2023). Currently, the methods that have proven the greatest multiplexing capacity are uCoTarget with 5 targets, MAbID with 6 targets, and ChIP-DIP with hundreds of targets (>225) (Lochs et al, 2023; preprint: Perez et al, 2023; Xiong et al, 2024).
Based on the research question one needs to also consider the input sample requirements and the data output generated. Sample conditions range from native, to light or strong fixation, which can be tailored based on the sample origin and the targets assayed (for more details, see Table EV1: Sample conditions). Although some methods have only been tested in cultured cells, optimized sample preparation and tissue dissociation procedures, along with optimized fixation conditions, should make many methods applicable to tissues or clinical samples (see Table EV1: Sample type tested). Of note, methods that require expression of a fusion protein such as with SpDamID are generally not suitable for processing clinical samples or most non-cultured sample types (Hass et al, 2015).
Recent advances with ChIP-reChIP have allowed to reduce the number of cells required as starting material (from tens of million to two million cells) (Seneviratne et al, 2024), but most ChIP-based methods still require a higher number of cells than enzyme-tethering approaches (see Table EV1: Input; Cells loaded; Cells recovered). Indeed, most of the enzyme-tethering approaches presented here have been adapted for single-cell or ultra-low-input profiling. Several optimizations particularly with tagmentation-based methods have improved the tagmentation efficiency and thus the number of fragments per cell (see Table EV1: Fragments per cell; Fragments per cell per target), for example by performing the tagmentation reactions sequentially (rather than simultaneously), beginning with the least abundant target (see Table EV1: Multi-targeting approach) (Stuart et al, 2022; Meers et al, 2023; Xiong et al, 2024). Additionally, using a second set of transposomes tethered by a secondary antibody increases the number of enzyme complexes recruited thereby facilitating further tagmentation (see Table EV1: Antibody targeting) (Meers et al, 2023; Xiong et al, 2024). Another elegant strategy is exploited in the nano-CT protocol: it involves linear amplification for library construction, which obviates the need for two nearby and properly oriented tagmentation events for fragment amplification (Bartosovic and Castelo-Branco, 2022). While this strategy results in a greater number of fragments per cell allowing for a lower amount of starting material to be used, a trade-off can be a slightly higher tagmentation background and therefore a lower fraction of reads in peaks (FRiP) (Bartosovic and Castelo-Branco, 2022).

Accessibility

The implementation of these toolkits in a laboratory may come with high costs as well as certain challenges related to specialized equipment, compatibility with commonly used platforms, intricate experimental steps, or complex analysis workflows. In particular, different methods for single-cell profiling often involve the use of costly and specialized platforms (see Table EV1: Single-cell approach). These specific single-cell profiling approaches as well as more accessible and scalable alternatives are described in Box 1.
It is important to note that a subset of methods is currently not compatible with standard illumina protocols and require custom sequencing primers, which often prevents a pooling of samples from different experiments (see Table EV1: Compatibility with illumina standard protocols). These include multi-CUT&Tag, scGET-seq, nano-CT and NTT-seq (Gopalan et al, 2021; Bartosovic and Castelo-Branco, 2022; Gopalan and Fazzio, 2022a; Stuart et al, 2022; Tedesco et al, 2022). However, as performed with uCoTarget, adaptations of adapter sequences used are possible in order to obtain libraries compatible with standard Iillumina protocols (Xiong et al, 2024).
Several workflows require additional reagent preparation steps or specialized equipment (see Table EV1: Extra reagent preparation and specialized equipment required). An example is the multi-CUT&Tag protocol, which makes use of a specific histidine-tagged pA-Tn5 fusion that has to be expressed and purified (Gopalan et al, 2021; Gopalan and Fazzio, 2022a). Along the same lines, ChIP-DIP requires the preparation of labeled beads for each antibody used (preprint: Perez et al, 2023). Similarly, MulTI-Tag and MAbID make use of a ChIL probe (Harada et al, 2019; Handa et al, 2020) that requires covalent conjugation of antibodies to barcoded adapters (Lochs et al, 2023; Meers et al, 2023). This step was introduced in order to reduce off-target signals, however Xiong et al, have recently suggested that these antibody conjugates may actually hinder epitope recognition, particularly with histone PTMs (Xiong et al, 2024). Methods that involve the expression of a fusion protein such as with SpDamID require testing and optimization for each fusion protein as well as for each new experimental system (Gopalan and Fazzio, 2022b; Hass et al, 2015). Lastly, several methods require additional specialized equipment, such as TIRF microscopy (Shema et al, 2016), FACS devices, or microfluidics instruments (Gopalan et al, 2021; Bartosovic and Castelo-Branco, 2022; Gopalan and Fazzio, 2022a; Janssens et al, 2022; Stuart et al, 2022; Tedesco et al, 2022; Lochs et al, 2023; Meers et al, 2023; Yeung et al, 2023).

Box 1 Approaches to achieve single-cell -omics profiling and alternatives with microfluidics-free methods

Since the establishment of the first approach for cell-specific barcoding (Islam et al, 2011), the field of single-cell -omics profiling has rapidly evolved and been adapted for various modalities (Vandereyken et al, 2023). The novel methods used employ different strategies (plate-based, droplet-based, or combinatorial indexing) to barcode individual cells for subsequent pooling and amplification of the libraries ready for high-throughput next-generation sequencing.
Early approaches to achieve single-cell barcoding involved picking individual cells, distributing them on a 96-well PCR plate preloaded with buffers for processing and barcoding (Islam et al, 2011). Nowadays, plate-based methods (A) take advantage of automated strategies such as FACS or multi-sample nano-dispensing platforms to distribute individual cells into wells, where the experimental reactions take place, greatly reducing labor and improving throughput (Fan et al, 2015; Goldstein et al, 2017; Mereu et al, 2020). In addition, a FACS step can enrich specific cells, reducing sequencing costs. Similarly, a built-in imaging software with nano-dispensing instruments also allows to visualize wells of the microchip, and thus providing control over the selection of wells to use for downstream processing. Of note, these approaches have limited scalability and require robotics for high-throughput liquid-handling and processing of samples (Mereu et al, 2020). Nevertheless, they have been adopted in several workflows presented in this review (Janssens et al, 2022; Lochs et al, 2023; Meers et al, 2023; Yeung et al, 2023).
Droplet-based single-cell profiling methods (B) make use of a microfluidics system that enables the formation of droplets through water-in-oil emulsion (Salomon et al, 2019). These droplets serve as a reaction chamber where individual cells or nuclei are encapsulated with barcoding beads. Following barcoding, amplification, and library preparation, fragments bearing cell-specific barcodes are sequenced and bioinformatically demultiplexed for single-cell interrogation. Important advantages with this approach include the automation of the procedure, the increased throughput while simultaneously improving sensitivity and minimize reagent use (Salomon et al, 2019). This principle led to the establishment and wide adoption of technologies such as Drop-seq, inDrop, and Chromium 10X/GemCode (Klein et al, 2015; Macosko et al, 2015; Zheng et al, 2017). Single-cell profiling was initially developed for single-cell RNA sequencing (scRNA-seq) and has since then been adapted for other modalities where cells/nuclei are used as the reaction chamber for experimental processing (for example tagmentation), as with most methods presented in this review (Gopalan et al, 2021; Bartosovic and Castelo-Branco, 2022; Stuart et al, 2022; Tedesco et al, 2022). Commercial systems have streamlined the workflow thereby enabling the wide adoption of these approaches, but with limited flexibility for optimizations or adapting protocols to broaden the applicability and often high costs. More recently, a microfluidics-free droplet-based method has been established as an alternative, where droplets are generated simply with a vortexer, named particle-templated instant partition sequencing (PIP-seq) (Clark et al, 2023).
Combinatorial indexing (C) involves the sequential addition of barcodes to RNA or DNA molecules, generating cell-specific barcode combinations (Rosenberg et al, 2018; Cao et al, 2017). In brief, processed samples (native or fixed, cells or nuclei) are distributed into a 96-well plate where each well contains a unique barcoded adapter, to ligate the barcoded oligonucleotides to chromatin or cDNA fragments (Rosenberg et al, 2018; Ma et al, 2020). Samples are then pooled back together, and these split-and-pool steps are repeated three to four times, generating an exponential number of cell-specific barcode combinations. This approach comes with great advantages over microfluidics-based methods and plate-based methods given it obviates the need for partitioning individual cells into droplets or microwells. Combinatorial barcoding therefore offers greater flexibility in terms of scalability potential and adapter compatibility, and it does not require any specialized equipment. Split-pool combinatorial indexing was initially developed for scRNA-seq and has now been adopted to simultaneously profile multiple chromatin features along with the transcriptome in the same single cells (Zhu et al, 2019; Ma et al, 2020; Xiong et al, 2024), providing a scalable and flexible alternative over expensive kits and specialized equipment.

Interpretation

Several factors need to be considered regarding the interpretation of the data generated, such as the specificity of the targeting approach, the intrinsic biases inherent to the technique used, the potential background, and the level of co-occurrence detection (see Table EV1: Data interpretation, Signal resolution, and Co-occurrence level detected).
The specificity of the targeting strategy depends heavily on the antibodies used as commercial antibodies vary in their specificity and often even from batch to batch. In addition, covalent labeling with a barcoded oligonucleotide (ChIL probe) (Harada et al, 2019; Handa et al, 2020; Lochs et al, 2023; Meers et al, 2023) may result in epitope masking (Xiong et al, 2024). Many antibodies were originally selected for specificity in ChIP and their specificity in other assays still has to be validated. Several approaches such as DamID-based methods or scGET-seq circumvent the use of antibodies (Hass et al, 2015; Tedesco et al, 2022). However, testing is required to ensure the tethered Dam does not interfere with the activity of the target proteins (Greil et al, 2006). In addition, Dam is known to have a bias toward accessible chromatin, and Dam expression for extended periods of time can result in off-target adenine methylation (Greil et al, 2006). This can be controlled for by comparing the enrichment signal with that of an unfused Dam (Greil et al, 2006). Similarly, tagmentation-based approaches are known to have inherent accessibility bias (Steiniger et al, 2006; Buenrostro et al, 2013; Kaya-Okur et al, 2019). Higher salt conditions during tagmentation steps have been performed to mitigate this bias, but it still remains unclear whether these buffer conditions are sufficient to completely overcome open chromatin biases (preprint: Meng and Yi 2021).
Background signal can for example arise from residual activity of untethered enzymes. ChIP-based methods are known to show higher background signals with potential for epitope masking resulting from crosslinking conditions (Park et al, 2013; Teytelman et al, 2013; Meyer and Liu, 2014; Jain et al, 2015; Baranello et al, 2016). N-ChIP can improve sensitivity, specificity and overall efficiency (Brind’Amour et al, 2015). Anti-IgG controls can be used to account for signal background in ChIP-based methods, but such negative controls perform poorly with tagmentation-based experiments (Gopalan and Fazzio, 2022a). With DamID approaches, the in vivo expression of Dam can result in spurious adenine methylation with levels that gradually accumulate in accessible chromatin, and the background level greatly differs between different Dam fusions (Greil et al, 2006; Rang et al, 2022).
The signal resolution and co-occurrence detection both depend on the fragmentation strategy, the specific motif recognition of the enzyme used, the “tagging” approach, and the binning strategy during analysis (see Table EV1: Signal resolution and co-occurrence level detected).
ChIP-based methods that make use of sonication for chromatin shearing will give rise to broader fragment sizes (and therefore lower resolution) (Kinkley et al, 2016; Weiner et al, 2016), in comparison with MNase-digested chromatin which can be easily adapted to obtain predominantly mononucleosome-sized fragments (Seneviratne et al, 2024; Shema et al, 2016). For this reason, the level of genomic co-occurrence detected can range from allelic to nucleosomal level based on the fragment size with reChIP-seq, co-ChIP-seq, and ChIP-reChIP (Kinkley et al, 2016; Weiner et al, 2016; Seneviratne et al, 2024). Because single-molecule combinatorial nucleosome profiling allows to profile mononucleosomes and individual histone proteins, its detection resolution is at the histone co-occurrence level (Shema et al, 2016). In contrast, although the fragment sizes range from 150–700 bp, ChIP-DIP can only study spatial-level co-occurrence given the tagging strategy is performed in bulk (preprint: Perez et al, 2023).
With SpDamID, as Dam specifically methylates adenine in the GATC motif, the resolution is limited to 1–2 kb, depending on the frequency distribution of GATC motifs in the genome, allowing in most cases to measure co-occurrence at the allelic level (Greil et al, 2006; Hass et al, 2015; Gopalan and Fazzio, 2022b; Rang et al, 2022). Similarly, MAbID can detect allelic co-occurrence, and its resolution is limited by the target motifs of the restriction enzymes used to insert antibody-specific barcodes and was determined to be 1–2 kb broader than with ChIP-seq (3–4 kb for H3K4me3, 7–8 kb for H3K27me3) (Lochs et al, 2023).
With scChIX-seq, even though the MNase-digested chromatin fragments predominantly reflect mononucleosomes in size, the signal can only be at locus-resolution (Yeung et al, 2023). This is because even though a single nucleosome bears multiple modifications, it will only be cut once. As this approach does not involve any “tagging” (as for example in tagmentation-introduced barcodes), the read fragments need to be merged into windows of 5–50 kb in size. Furthermore, because with scChIX-seq the profiles are deconvoluted, the genomic co-occurrence can only be inferred rather than directly measured (Yeung et al, 2023).
To allow the detection of co-occurrence on a single fragment with scMulti-CUT&Tag, Gopalan and al. have designed a technique that inserts antibody-specific barcoded adapters, where co-existing targets will result in fragments flanked by two different barcodes (mixed insertion events) whereas fragments flanked with the same barcode would arise from DNA bound to an individual assayed target (single insertion event). While the genomic resolution of the co-occurrence detected has not been specifically tested, the size distribution of the reads arising from mixed insertion events were representative of nucleosomal and sub-nucleosomal fragments (Gopalan and Fazzio, 2022a; Gopalan et al, 2021). For this reason, both scMulti-CUT&Tag, MulTI-Tag, and uCoTargetX can detect allelic or nucleosomal co-occurrence (Gopalan et al, 2021; Gopalan and Fazzio, 2022a; Meers et al, 2023; Xiong et al, 2024). Importantly, most methods are limited by two targets that can be detected per fragment (for example with methods that insert antibody-specific barcodes on each end of a fragment). Binning strategies allow to mitigate this drawback but result in a decreased resolution as a trade-off.

Additional features

Finally, several workflows allow to assay additional features/modalities and thus broaden the method’s applicability (see Table EV1: Additional features). Along with profiling DNA-binding proteins or histone PTMs, tagmentation-based approaches have the possibility to assay transposase accessible chromatin (ATAC) in parallel, as exemplified with scGET-seq and nano-CT (Buenrostro et al, 2015; Bartosovic and Castelo-Branco, 2022; Tedesco et al, 2022). In addition, several toolkits have integrated a reverse transcription step in order to jointly capture mRNAs along with chromatin features in single cells, including scDam&T, SHARE-seq (simultaneous high-throughput ATAC and RNA expression with sequencing), Paired-tag, CoTECH (combined assay of transcriptome and enriched chromatin binding), SET-seq (same cell epigenome and transcriptome sequencing), scPCOR-seq, and more (Rooijers et al, 2019; Ma et al, 2020; Sun et al, 2021; Xiong et al, 2021; Zhu et al, 2021; Pan et al, 2022). More recently, joint gene expression measurement with multiple chromatin features has been described with uCoTargetX (Xiong et al, 2024). Such approaches have the potential to unambiguously link chromatin features with transcriptional output. Other methods that have a FACS step included in their framework such as NTT-seq, scChIX-seq, and MAbID, can simultaneously profile cell surface protein expression and thereby enrich for specific cell types (Stuart et al, 2022; Lochs et al, 2023; Yeung et al, 2023). Lastly, because Dam-induced adenine methylation deposition is stable and cumulative, the DamID-based toolkits offer the possibility to track epigenetic histories over varying time windows (Kind et al, 2013, 2015; Park et al, 2019; Rooijers et al, 2019; Gopalan and Fazzio, 2022b; Rang et al, 2022).

Conclusions and perspectives

Over two decades ago, the “histone code” hypothesis proposed that the co-occurrence of multiple histone PTMs could give rise to unique biological outcomes (Turner, 1993, 2000; Strahl and Allis, 2000; Jenuwein and Allis, 2001). Since then, it has become evident that multiple chromatin features, not only histone PTMs, but also DNA-binding proteins, chromatin proteins, DNA modifications, or chromatin accessibility, co-occur and act cooperatively to dictate specific biological outcomes.
The number of approaches to profile various epigenomic modalities in single cells has rapidly evolved in the recent years and has revealed the ever-growing complexity of biological systems and heterogeneity of cellular states (Vandereyken et al, 2023). More recently, advances in methodological toolkits as described in this review allow now to profile hundreds of multiplexed chromatin proteins in bulk samples (preprint: Perez et al, 2023), and multiple chromatin features in single cells (Gopalan et al, 2021; Bartosovic and Castelo-Branco, 2022; Janssens et al, 2022; Stuart et al, 2022; Tedesco et al, 2022; Lochs et al, 2023; Meers et al, 2023; Yeung et al, 2023; Xiong et al, 2024).
The currently available toolkits described in this review have so far mainly been applied to improve the identification of cell types, or to define cellular states, trajectories, and chromatin velocity, by leveraging the multimodal features of the approaches. Moving forward, the potential of these toolkits in profiling varying levels of chromatin feature co-occurrence will be used to address further biological questions. In this context, major challenges in studying the combinatorial occurrence of chromatin features involve the applicability of the toolkits to study low-abundant targets and rare cell populations (see also Box 2 for more information). Specifically, optimizations will be required to overcome issues with data sparsity, limited coverage, and enhance the throughput of single-cell profiling, as well as to increase the number of chromatin targets simultaneously multiplexed per experiment. In the long run, these advances will allow to unambiguously link combinatorial chromatin feature occurrences to transcriptomic states and the systematic decoding of chromatin features in health and disease.

Box 2 In need of answers

1.
How does the co-occurrence of chromatin features fine-tune gene expression?
Simultaneously profiling the transcriptome along with various chromatin features in the same cell will allow to unambiguously correlate chromatin states with gene expression outputs.
2.
What is the functional relevance of the combinatorial occurrence of histone PTMs, DNA-binding proteins, and/or chromatin proteins?
Genetic, epigenetic, pharmacological, or environmental/metabolic manipulations will be useful to dissect the role of different combinations of chromatin features on cellular function and gene expression in the context of a wide range of biological processes such as development, stemness, differentiation, metabolism, in health and disease.
3.
How do the distinct levels of co-occurrence (spatial, cellular, allelic, nucleosomal, and histone) contribute to the regulation of biological processes?
Combinations of different approaches, their future refinements, and novel techniques, will allow greater resolution, the profiling of an increasing number of targets, in lower cell numbers and thus providing new insights into the biological relevance of chromatin feature co-occurrence.
4.
What aspects of the currently available toolkits require improvements?
Combining cellular co-occurrence profiling along with measurement of additional features such as of the nascent transcriptome, 3D-chromatin architecture, or temporal information, will allow to directly correlate combinatorial occurrence with different genomic outputs. The described toolkits have already shown that multimodal profiling allows to more accurately define cellular states and chromatin landscape, and that multifactorial data integration enhances the study of chromatin velocity and trajectories along developmental and differentiation progression. Increasing the number of targets or genomic features simultaneously assayed within each cell or the temporal resolution will allow to further interrogate the heterogeneity of cellular types and states and provide increasingly complex maps of chromatin features. Improving the sensitivity of the methods in order to obtain a higher number of reads per cell will also be beneficial to mitigate the data sparsity inherent to single-cell profiling approaches.
5.
What are the current major challenges?
Experimental challenges involve studying rare cell populations as well as the profiling of low-abundant targets. Furthermore, the wider adoption of rather complex workflows may require optimizations and possibilities of cost-efficient and fast sequencing turn around. Computational challenges involve data analysis and visualization due to the increasing complexity of the datasets, as well as the implementation and utilization of computational pipelines in non-expert research groups. Importantly, the increasing number of datasets generated will require the establishment of novel data integration approaches as well as the benchmarking of such computational toolkits. Additionally, standardized and open-access pipelines will be needed in order to ease the process of data mining, pre-processing, and analysing such complex datasets.

Author contributions

Anne-Sophie Pepin: Visualization; Writing—original draft; Writing—review and editing. Robert Schneider: Resources; Supervision; Writing—original draft; Writing—review and editing.

Disclosure and competing interests statement

The authors declare no competing interests.

Acknowledgements

The authors would like to thank Dr. Till Bartke for useful discussions. This work was supported by the Deutsche Forschungsgemeinschaft (DFG, German Research Foundation) through SFB 1064 (Project-ID 213249687), SFB 1309 (Project-ID 325871075), and the Helmholtz Gemeinschaft.

Supporting Information

References

Abascal F, Acosta R, Addleman NJ, Adrian J, Afzal V, Ai R, Aken B, Akiyama JA, Jammal OA, Amrhein H, et al. Expanded encyclopaedias of DNA elements in the human and mouse genomes Nature 2020 583 699-710
Ahmad ZA, Yeap SK, Ali AM, Ho WY, Alitheen NBM, and Hamid M scFv antibody: principles and clinical application Clin Dev Immunol 2012 2012 980250
Akbarian S, Liu C, Knowles JA, Vaccarino FM, Farnham PJ, Crawford GE, Jaffe AE, Pinto D, Dracheva S, Geschwind DH, et al. The PsychENCODE project Nat Neurosci 2015 18 1707-1712
Altemose N, Maslan A, Smith OK, Sundararajan K, Brown RR, Mishra R, Detweiler AM, Neff N, Miga KH, Straight AF, et al. DiMeLo-seq: a long-read, single-molecule method for mapping protein–DNA interactions genome wide Nat Methods 2022 19 711-723
Azuara V, Perry P, Sauer S, Spivakov M, Jørgensen HF, John RM, Gouti M, Casanova M, Warnes G, Merkenschlager M, et al. Chromatin signatures of pluripotent cell lines Nat Cell Biol 2006 8 532-538
Bai X, Bi W, Dong H, Chen P, Tian S, Zhai G, and Zhang K An integrated approach based on a DNA self-assembly technique for characterization of crosstalk among combinatorial histone modifications Anal Chem 2018 90 3692-3696
Baranello L, Kouzine F, Sanford S, and Levens D ChIP bias as a function of cross-linking time Chromosome Res 2016 24 175-181
Barski A, Cuddapah S, Cui K, Roh T-Y, Schones DE, Wang Z, Wei G, Chepelev I, and Zhao K High-resolution profiling of histone methylations in the human genome Cell 2007 129 823-837
Bartosovic M and Castelo-Branco G Multimodal chromatin profiling using nanobody-based single-cell CUT&Tag Nat Biotechnol 2022 1 12
Bartosovic M, Kabbe M, and Castelo-Branco G Single-cell CUT&Tag profiles histone modifications and transcription factors in complex tissues Nat Biotechnol 2021 39 825-835
Bernstein BE, Mikkelsen TS, Xie X, Kamal M, Huebert DJ, Cuff J, Fry B, Meissner A, Wernig M, Plath K, et al. A bivalent chromatin structure marks key developmental genes in embryonic stem cells Cell 2006 125 315-326
Bird RE, Hardman KD, Jacobson JW, Johnson S, Kaufman BM, Lee SM, Lee T, Pope SH, Riordan GS, and Whitlow M Single-chain antigen-binding proteins Science 1988 242 423-426
Blanco E, González-Ramírez M, Alcaine-Colet A, Aranda S, and Di Croce L The bivalent genome: characterization, structure, and regulation Trends Genet TIG 2020 36 118-131
Boyer LA, Lee TI, Cole MF, Johnstone SE, Levine SS, Zucker JP, Guenther MG, Kumar RM, Murray HL, Jenner RG, et al. Core transcriptional regulatory circuitry in human embryonic stem cells Cell 2005 122 947-956
Brind’Amour J, Liu S, Hudson M, Chen C, Karimi MM, and Lorincz MC An ultra-low-input native ChIP-seq protocol for genome-wide profiling of rare cell populations Nat Commun 2015 6 6033
Buenrostro JD, Giresi PG, Zaba LC, Chang HY, and Greenleaf WJ Transposition of native chromatin for fast and sensitive epigenomic profiling of open chromatin, DNA-binding proteins and nucleosome position Nat Methods 2013 10 1213-1218
Buenrostro JD, Wu B, Chang HY, and Greenleaf WJ ATAC-seq: a method for assaying chromatin accessibility genome-wide Curr Protoc Mol Biol 2015 109 21.29.1-21.29.9
Cao J, Packer JS, Ramani V, Cusanovich DA, Huynh C, Daza R, Qiu X, Lee C, Furlan SN, Steemers FJ, et al. Comprehensive single-cell transcriptional profiling of a multicellular organism Science 2017 357 661-667
Carter B, Ku WL, Kang JY, Hu G, Perrie J, Tang Q, and Zhao K Mapping histone modifications in low cell number and single cells using antibody-guided chromatin tagmentation (ACT-seq) Nat Commun 2019 10 3747
Chaya D, Hayamizu T, Bustin M, and Zaret KS Transcription factor FoxA (HNF3) on a nucleosome at an enhancer complex in liver chromatin J Biol Chem 2001 276 44385-44389
Clark IC, Fontanez KM, Meltzer RH, Xue Y, Hayford C, May-Zhang A, D’Amato C, Osman A, Zhang JQ, Hettige P, et al. Microfluidics-free single-cell genomics with templated emulsification Nat Biotechnol 2023 41 1557-1566
Dai S-M, Chen H-H, Chang C, Riggs AD, and Flanagan SD Ligation-mediated PCR for quantitative in vivo footprinting Nat Biotechnol 2000 18 1108-1111
Dedon PC, Soults JA, Allis CD, and Gorovsky MA Formaldehyde cross-linking and immunoprecipitation demonstrate developmental changes in H1 association with transcriptionally active genes Mol Cell Biol 1991 11 1729-1733
Dunham I, Kundaje A, Aldred SF, Collins PJ, Davis CA, Doyle F, Epstein CB, Frietze S, Harrow J, Kaul R, et al. An integrated encyclopedia of DNA elements in the human genome Nature 2012 489 57-74
Fan HC, Fu GK, and Fodor SPA Combinatorial labeling of single cells for gene expression cytometry Science 2015 347 1258367
Furey TS ChIP–seq and beyond: new and improved methodologies to detect and characterize protein–DNA interactions Nat Rev Genet 2012 13 840-852
Gao M, Wang J, Rousseaux S, Tan M, Pan L, Peng L, Wang S, Xu W, Ren J, Liu Y, et al. Metabolically controlled histone H4K5 acylation/acetylation ratio drives BRD4 genomic distribution Cell Rep 2021 36 109460
Geisberg JV and Struhl K Cellular stress alters the transcriptional properties of promoter-bound Mot1-TBP complexes Mol Cell 2004 14 479-489
Gilmour DS and Lis JT Detecting protein-DNA interactions in vivo: distribution of RNA polymerase on specific bacterial genes Proc Natl Acad Sci USA 1984 81 4275-4279
Gilmour DS and Lis JT In vivo interactions of RNA polymerase II with genes of Drosophila melanogaster Mol Cell Biol 1985 5 2009-2018
Gilmour DS and Lis JT RNA polymerase II interacts with the promoter region of the noninduced hsp70 gene in Drosophila melanogaster cells Mol Cell Biol 1986 6 3984-3989
Goldstein LD, Chen Y-JJ, Dunne J, Mir A, Hubschle H, Guillory J, Yuan W, Zhang J, Stinson J, Jaiswal B, et al. Massively parallel nanowell-based single-cell gene expression profiling BMC Genomics 2017 18 519
Gopalan S and Fazzio TG Multi-CUT&Tag to simultaneously profile multiple chromatin factors STAR Protoc 2022 3 101100
Gopalan S and Fazzio TG Multiomic chromatin and transcription profiling with EpiDamID Cell Rep Methods 2022 2 100219
Gopalan S, Wang Y, Harper NW, Garber M, and Fazzio TG Simultaneous profiling of multiple chromatin proteins in the same cells Mol Cell 2021 81 4736-4746.e5
Goudarzi A, Zhang D, Huang H, Barral S, Kwon OK, Qi S, Tang Z, Buchou T, Vitte A-L, He T, et al. Dynamic competing histone H4 K5K8 acetylation and butyrylation are hallmarks of highly active gene promoters Mol Cell 2016 62 169-180
Greil F, Moorman C, van Steensel B (2006) [16] DamID: mapping of in vivo protein–genome interactions using tethered DNA adenine methyltransferase. In: Methods in enzymology. (eds Kimmel A, Oliver B) Academic Press, pp 342–359
Grosselin K, Durand A, Marsolier J, Poitou A, Marangoni E, Nemati F, Dahmani A, Lameiras S, Reyal F, Frenoy O, et al. High-throughput single-cell ChIP-seq identifies heterogeneity of chromatin states in breast cancer Nat Genet 2019 51 1060-1066
Hainer SJ, Bošković A, McCannell KN, Rando OJ, and Fazzio TG Profiling of pluripotency factors in single cells and early embryos Cell 2019 177 1319-1329.e11
Handa T, Harada A, Maehara K, Sato S, Nakao M, Goto N, Kurumizaka H, Ohkawa Y, and Kimura H Chromatin integration labeling for mapping DNA-binding proteins and modifications with low input Nat Protoc 2020 15 3334-3360
Harada A, Maehara K, Handa T, Arimura Y, Nogami J, Hayashi-Takanaka Y, Shirahige K, Kurumizaka H, Kimura H, and Ohkawa Y A chromatin integration labelling method enables epigenomic profiling with lower input Nat Cell Biol 2019 21 287-296
Harikumar A and Meshorer E Chromatin remodeling and bivalent histone modifications in embryonic stem cells EMBO Rep 2015 16 1609-1619
Hass MR, Liow H-H, Chen X, Sharma A, Inoue YU, Inoue T, Reeb A, Martens A, Fulbright M, Raju S, et al. SpDamID: marking DNA bound by protein complexes identifies notch-dimer responsive enhancers Mol Cell 2015 59 685-697
Hebbes TR, Thorne AW, and Crane-Robinson C A direct link between core histone acetylation and transcriptionally active chromatin EMBO J 1988 7 1395-1402
Henikoff S, Henikoff JG, and Ahmad K Simplified epigenome profiling using antibody-tethered tagmentation Bio-Protoc 2021 11 e4043
Henikoff S, Henikoff JG, Kaya-Okur HS, and Ahmad K Efficient chromatin accessibility mapping in situ by nucleosome-tethered tagmentation eLife 2020 9 e63274
Henry KW, Wyce A, Lo W-S, Duggan LJ, Emre NCT, Kao C-F, Pillus L, Shilatifard A, Osley MA, and Berger SL Transcriptional activation via sequential histone H2B ubiquitylation and deubiquitylation, mediated by SAGA-associated Ubp8 Genes Dev 2003 17 2648-2663
IJpenberg A, Tan NS, Gelman L, Kersten S, Seydoux J, Xu J, Metzger D, Canaple L, Chambon P, Wahli W, et al. In vivo activation of PPAR target genes by RXR homodimers EMBO J 2004 23 2083-2091
Islam S, Kjällquist U, Moliner A, Zajac P, Fan J-B, Lönnerberg P, and Linnarsson S Characterization of the single-cell transcriptional landscape by highly multiplex RNA-seq Genome Res 2011 21 1160-1167
Jain D, Baldi S, Zabel A, Straub T, and Becker PB Active promoters give rise to false positive ‘Phantom Peaks’ in ChIP-seq experiments Nucleic Acids Res 2015 43 6959-6968
Janssen KA, Coradin M, Lu C, Sidoli S, and Garcia BA Quantitation of single and combinatorial histone modifications by integrated chromatography of bottom-up peptides and middle-down polypeptide tails J Am Soc Mass Spectrom 2019 30 2449-2459
Janssens DH, Otto DJ, Meers MP, Setty M, Ahmad K, and Henikoff S CUT&Tag2for1: a modified method for simultaneous profiling of the accessible and silenced regulome in single cells Genome Biol 2022 23 81
Jenuwein T and Allis CD Translating the histone code Science 2001 293 1074-1080
Kaya-Okur HS, Wu SJ, Codomo CA, Pledger ES, Bryson TD, Henikoff JG, Ahmad K, and Henikoff S CUT&Tag for efficient epigenomic profiling of small samples and single cells Nat Commun 2019 10 1930
Kim TH, Barrera LO, Qu C, Van Calcar S, Trinklein ND, Cooper SJ, Luna RM, Glass CK, Rosenfeld MG, Myers RM, et al. Direct isolation and identification of promoters in the human genome Genome Res 2005 15 830-839
Kim TH, Barrera LO, Zheng M, Qu C, Singer MA, Richmond TA, Wu Y, Green RD, and Ren B A high-resolution map of active promoters in the human genome Nature 2005 436 876-880
Kind J, Pagie L, de Vries SS, Nahidiazar L, Dey SS, Bienko M, Zhan Y, Lajoie B, de Graaf CA, Amendola M, et al. Genome-wide maps of nuclear lamina interactions in single human cells Cell 2015 163 134-147
Kind J, Pagie L, Ortabozkoyun H, Boyle S, de Vries SS, Janssen H, Amendola M, Nolen LD, Bickmore WA, and van Steensel B Single-cell dynamics of genome-nuclear lamina interactions Cell 2013 153 178-192
Kinkley S, Helmuth J, Polansky JK, Dunkel I, Gasparoni G, Fröhler S, Chen W, Walter J, Hamann A, and Chung H-R reChIP-seq reveals widespread bivalency of H3K4me3 and H3K27me3 in CD4+ memory T cells Nat Commun 2016 7 12514
Klein AM, Mazutis L, Akartuna I, Tallapragada N, Veres A, Li V, Peshkin L, Weitz DA, and Kirschner MW Droplet barcoding for single-cell transcriptomics applied to embryonic stem cells Cell 2015 161 1187-1201
Ku WL, Nakamura K, Gao W, Cui K, Hu G, Tang Q, Ni B, and Zhao K Single-cell chromatin immunocleavage sequencing (scChIC-seq) to profile histone modification Nat Methods 2019 16 323-325
Kumar D, Cinghu S, Oldfield AJ, Yang P, and Jothi R Decoding the function of bivalent chromatin in development and cancer Genome Res 2021 31 2170-2184
Lee J-S, Lee C-H, and Chung JH Studying the recruitment of Sp1 to the β-globin promoter with an in vivo method: Protein position identification with nuclease tail (PIN*POINT) Proc Natl Acad Sci USA 1998 95 969-974
Lee TI, Jenner RG, Boyer LA, Guenther MG, Levine SS, Kumar RM, Chevalier B, Johnstone SE, Cole MF, Isono K, et al. Control of developmental regulators by polycomb in human embryonic stem cells Cell 2006 125 301-313
Lochs SJA, van der Weide RH, de Luca KL, Korthout T, van Beek RE, Kimura H, and Kind J Combinatorial single-cell profiling of major chromatin types with MAbID Nat Methods 2023 1 11
Lu C, Coradin M, Janssen KA, Sidoli S, and Garcia BA Combinatorial histone H3 modifications are dynamically altered in distinct cell cycle phases J Am Soc Mass Spectrom 2021 32 1300-1311
Luger K, Mäder AW, Richmond RK, Sargent DF, and Richmond TJ Crystal structure of the nucleosome core particle at 2.8 Å resolution Nature 1997 389 251-260
Lukauskas S, Tvardovskiy A, Nguyen NV, Stadler M, Faull P, Ravnsborg T, Özdemir Aygenli B, Dornauer S, Flynn H, Lindeboom RGH, et al. Decoding chromatin states by proteomic profiling of nucleosome readers Nature 2024 627 671-679
Ma S, Zhang B, LaFave LM, Earl AS, Chiang Z, Hu Y, Ding J, Brack A, Kartha VK, Tay T, et al. Chromatin potential identified by shared single-cell profiling of RNA and chromatin Cell 2020 183 1103-1116.e20
Macosko EZ, Basu A, Satija R, Nemesh J, Shekhar K, Goldman M, Tirosh I, Bialas AR, Kamitaki N, Martersteck EM, et al. Highly parallel genome-wide expression profiling of individual cells using nanoliter droplets Cell 2015 161 1202-1214
Meers MP, Llagas G, Janssens DH, Codomo CA, and Henikoff S Multifactorial profiling of epigenetic landscapes at single-cell resolution using MulTI-Tag Nat Biotechnol 2023 41 708-716
Meng W, Yi Z (2021) Tn5 transposase-based epigenomic profiling methods are prone to open chromatin bias. Preprint at BioRxiv
Mereu, Lafzi E, Moutinho A, Ziegenhain C, McCarthy C, Álvarez-Varela DJ, Batlle A, Sagar E, Grün D, Lau JK, et al. Benchmarking single-cell RNA-sequencing protocols for cell atlas projects Nat Biotechnol 2020 38 747-755
Métivier R, Penot G, Hübner MR, Reid G, Brand H, Kos M, and Gannon F Estrogen receptor-alpha directs ordered, cyclical, and combinatorial recruitment of cofactors on a natural target promoter Cell 2003 115 751-763
Meyer CA and Liu XS Identifying and mitigating bias in next-generation sequencing methods for chromatin biology Nat Rev Genet 2014 15 709-721
Mikkelsen TS, Ku M, Jaffe DB, Issac B, Lieberman E, Giannoukos G, Alvarez P, Brockman W, Kim T-K, Koche RP, et al. Genome-wide maps of chromatin state in pluripotent and lineage-committed cells Nature 2007 448 553-560
Millán-Zambrano G, Burton A, Bannister AJ, and Schneider R Histone post-translational modifications—cause and consequence of genome function Nat Rev Genet 2022 23 563-580
Orlando V, Strutt H, and Paro R Analysis of chromatin structure by in vivo formaldehyde cross-linking Methods 1997 11 205-214
Pan L, Ku WL, Tang Q, Cao Y, and Zhao K scPCOR-seq enables co-profiling of chromatin occupancy and RNAs in single cells Commun Biol 2022 5 1-9
Park D, Lee Y, Bhupindersingh G, and Iyer VR Widespread misinterpretable ChIP-seq bias in yeast PLoS ONE 2013 8 e83506
Park M, Patel N, Keung AJ, and Khalil AS Engineering epigenetic regulation using synthetic read-write modules Cell 2019 176 227-238.e20
Park PJ ChIP–seq: advantages and challenges of a maturing technology Nat Rev Genet 2009 10 669-680
Partridge EC, Chhetri SB, Prokop JW, Ramaker RC, Jansen CS, Goh S-T, Mackiewicz M, Newberry KM, Brandsmeier LA, Meadows SK, et al. Occupancy maps of 208 chromatin-associated proteins in one human cell type Nature 2020 583 720-728
Perez AA, Goronzy IN, Blanco MR, Guo JK, Guttman M (2023) ChIP-DIP: a multiplexed method for mapping hundreds of proteins to DNA uncovers diverse regulatory elements controlling gene expression. Preprint at BioRxiv
Picelli S, Björklund AK, Reinius B, Sagasser S, Winberg G, and Sandberg R Tn5 transposase and tagmentation procedures for massively scaled sequencing projects Genome Res 2014 24 2033-2040
Policarpi C, Munafò M, Tsagkris S, Carlini V, and Hackett JA Systematic epigenome editing captures the context-dependent instructive function of chromatin modifications Nat Genet 2024 56 1168-1180
Proft M and Struhl K Hog1 kinase converts the Sko1-Cyc8-Tup1 repressor complex into an activator that recruits SAGA and SWI/SNF in response to osmotic stress Mol Cell 2002 9 1307-1317
Quinodoz SA, Bhat P, Chovanec P, Jachowicz JW, Ollikainen N, Detmar E, Soehalim E, and Guttman M SPRITE: a genome-wide method for mapping higher-order 3D interactions in the nucleus using combinatorial split-and-pool barcoding Nat Protoc 2022 17 36-75
Quinodoz SA, Jachowicz JW, Bhat P, Ollikainen N, Banerjee AK, Goronzy IN, Blanco MR, Chovanec P, Chow A, Markaki Y, et al. RNA promotes the formation of spatial compartments in the nucleus Cell 2021 184 5775-5790.e30
Quinodoz SA, Ollikainen N, Tabak B, Palla A, Schmidt JM, Detmar E, Lai MM, Shishkin AA, Bhat P, Takei Y, et al. Higher-order inter-chromosomal hubs shape 3D genome organization in the nucleus Cell 2018 174 744-757.e24
Rang FJ, de Luca KL, de Vries SS, Valdes-Quezada C, Boele E, Nguyen PD, Guerreiro I, Sato Y, Kimura H, Bakkers J, et al. Single-cell profiling of transcriptome and histone modifications with EpiDamID Mol Cell 2022 82 1956-1970.e14
Ren B, Robert F, Wyrick JJ, Aparicio O, Jennings EG, Simon I, Zeitlinger J, Schreiber J, Hannett N, Kanin E, et al. Genome-wide location and function of DNA binding proteins Science 2000 290 2306-2309
Reznikoff WS Tn5 as a model for understanding DNA transposition Mol Microbiol 2003 47 1199-1206
Rooijers K, Markodimitraki CM, Rang FJ, de Vries SS, Chialastri A, de Luca KL, Mooijman D, Dey SS, and Kind J Simultaneous quantification of protein–DNA contacts and transcriptomes in single cells Nat Biotechnol 2019 37 766-772
Rosenberg AB, Roco CM, Muscat RA, Kuchina A, Sample P, Yao Z, Graybuck LT, Peeler DJ, Mukherjee S, Chen W, et al. Single-cell profiling of the developing mouse brain and spinal cord with split-pool barcoding Science 2018 360 176-182
Rotem A, Ram O, Shoresh N, Sperling RA, Goren A, Weitz DA, and Bernstein BE Single-cell ChIP-seq reveals cell subpopulations defined by chromatin state Nat Biotechnol 2015 33 1165-1172
Salomon R, Kaczorowski D, Valdes-Mora F, Nordon RE, Neild A, Farbehi N, Bartonicek N, and Gallego-Ortega D Droplet-based single cell RNAseq tools: a practical guide Lab Chip 2019 19 1706-1727
Schmid M, Durussel T, and Laemmli UK ChIC and ChEC: genomic mapping of chromatin proteins Mol Cell 2004 16 147-157
Schmidl C, Rendeiro AF, Sheffield NC, and Bock C ChIPmentation: fast, robust, low-input ChIP-seq for histones and transcription factors Nat Methods 2015 12 963-965
Schwämmle V, Aspalter C-M, Sidoli S, and Jensen ON Large scale analysis of co-existing post-translational modifications in histone tails reveals global fine structure of cross-talk Mol Cell Proteomics MCP 2014 13 1855-1865
Scully KM, Jacobson EM, Jepsen K, Lunyak V, Viadiu H, Carrière C, Rose DW, Hooshmand F, Aggarwal AK, and Rosenfeld MG Allosteric effects of Pit-1 DNA sites on long-term repression in cell type specification Science 2000 290 1127-1131
Seneviratne JA, Ho WWH, Glancy E, and Eckersley-Maslin MA A low-input high resolution sequential chromatin immunoprecipitation method captures genome-wide dynamics of bivalent chromatin Epigenet Chromatin 2024 17 3
Shema E, Jones D, Shoresh N, Donohue L, Ram O, and Bernstein BE Single-molecule decoding of combinatorially modified nucleosomes Science 2016 352 717-721
Sidoli S, Cheng L, and Jensen ON Proteomics in chromatin biology and epigenetics: elucidation of post-translational modifications of histone proteins by mass spectrometry J Proteomics 2012 75 3419-3433
Sidoli S and Garcia BA Properly reading the histone code by MS-based proteomics Proteomics 2015 15 2901-2902
Skene PJ and Henikoff S A simple method for generating high-resolution maps of genome-wide protein binding eLife 2015 4 e09225
Skene PJ and Henikoff S An efficient targeted nuclease strategy for high-resolution mapping of DNA binding sites eLife 2017 6 e21856
Solomon MJ, Larsen PL, and Varshavsky A Mapping proteinDNA interactions in vivo with formaldehyde: evidence that histone H4 is retained on a highly transcribed gene Cell 1988 53 937-947
Solomon MJ and Varshavsky A Formaldehyde-mediated DNA-protein crosslinking: a probe for in vivo chromatin structures Proc Natl Acad Sci USA 1985 82 6470-6474
Soutoglou E and Talianidis I Coordination of PIC assembly and chromatin remodeling during differentiation-induced gene activation Science 2002 295 1901-1904
Steiniger M, Adams CD, Marko JF, and Reznikoff WS Defining characteristics of Tn5 Transposase non-specific DNA binding Nucleic Acids Res 2006 34 2820-2832
Strahl BD and Allis CD The language of covalent histone modifications Nature 2000 403 41-45
Stuart T, Hao S, Zhang B, Mekerishvili L, Landau DA, Maniatis S, Satija R, Raimondi I (2022) Nanobody-tethered transposition enables multifactorial chromatin profiling at single-cell resolution. Nat Biotechnol 41:806–812
Sun Z, Tang Y, Zhang Y, Fang Y, Jia J, Zeng W, and Fang D Joint single-cell multiomic analysis in Wnt3a induced asymmetric stem cell division Nat Commun 2021 12 5941
Tedesco M, Giannese F, Lazarević D, Giansanti V, Rosano D, Monzani S, Catalano I, Grassi E, Zanella ER, Botrugno OA, et al. Chromatin velocity reveals epigenetic dynamics by single-cell profiling of heterochromatin and euchromatin Nat Biotechnol 2022 40 235-244
Teytelman L, Thurtle DM, Rine J, and van Oudenaarden A Highly expressed loci are vulnerable to misleading ChIP localization of multiple unrelated proteins Proc Natl Acad Sci USA 2013 110 18602-18607
Turner BM Decoding the nucleosome Cell 1993 75 5-8
Turner BM Histone acetylation and an epigenetic code BioEssays News Rev Mol Cell Dev Biol 2000 22 836-845
van Steensel B and Henikoff S Identification of in vivo DNA targets of chromatin proteins using tethered Dam methyltransferase Nat Biotechnol 2000 18 424-428
Vandereyken K, Sifrim A, Thienpont B, and Voet T Methods and applications for single-cell and spatial multi-omics Nat Rev Genet 2023 1 22
Voigt P, LeRoy G, Drury WJ, Zee BM, Son J, Beck DB, Young NL, Garcia BA, and Reinberg D Asymmetrically modified nucleosomes Cell 2012 151 181-193
Wang Q, Xiong H, Ai S, Yu X, Liu Y, Zhang J, and He A CoBATCH for high-throughput single-cell epigenomic profiling Mol Cell 2019 76 206-216.e7
Weiner A, Lara-Astiaso D, Krupalnik V, Gafni O, David E, Winter DR, Hanna JH, and Amit I Co-ChIP enables genome-wide mapping of histone mark co-occurrence at single-molecule resolution Nat Biotechnol 2016 34 953-961
Xenaki KT, Oliveira S, van Bergen en Henegouwen PMP (2017) Antibody or antibody fragments: implications for molecular imaging and targeted therapy of solid tumors. Front Immunol 8:1287
Xiong H, Luo Y, Wang Q, Yu X, and He A Single-cell joint detection of chromatin occupancy and transcriptome enables higher-dimensional epigenomic reconstructions Nat Methods 2021 18 652-660
Xiong H, Wang Q, Li CC, and He A Single-cell joint profiling of multiple epigenetic proteins and gene transcription Sci Adv 2024 10 eadi3664
Yeung J, Florescu M, Zeller P, de Barbanson BA, Wellenstein MD, and van Oudenaarden A scChIX-seq infers dynamic relationships between histone modifications in single cells Nat Biotechnol 2023 41 813-823
Zeller P, Yeung J, Viñas Gaza H, de Barbanson BA, Bhardwaj V, Florescu M, van der Linden R, and van Oudenaarden A Single-cell sortChIC identifies hierarchical chromatin dynamics during hematopoiesis Nat Genet 2023 55 333-345
Zentner GE, Kasinathan S, Xin B, Rohs R, and Henikoff S ChEC-seq kinetics discriminates transcription factor binding sites by DNA sequence and shape in vivo Nat Commun 2015 6 8733
Zhang B, Srivastava A, Mimitou E, Stuart T, Raimondi I, Hao Y, Smibert P, and Satija R Characterizing cellular heterogeneity in chromatin state with scCUT&Tag-pro Nat Biotechnol 2022 40 1220-1230
Zhao Y and Garcia BA Comprehensive catalog of currently documented histone modifications Cold Spring Harb Perspect Biol 2015 7 a025064
Zheng GXY, Terry JM, Belgrader P, Ryvkin P, Bent ZW, Wilson R, Ziraldo SB, Wheeler TD, McDermott GP, Zhu J, et al. Massively parallel digital transcriptional profiling of single cells Nat Commun 2017 8 14049
Zhu C, Yu M, Huang H, Juric I, Abnousi A, Hu R, Lucero J, Behrens MM, Hu M, and Ren B An ultra high-throughput method for single-cell joint analysis of open chromatin and transcriptome Nat Struct Mol Biol 2019 26 1063-1070
Zhu C, Zhang Y, Li YE, Lucero J, Behrens MM, and Ren B Joint profiling of histone modifications and transcriptome in single cells from mouse brain Nat Methods 2021 18 283-292

Information & Authors

Information

Published In

EMBO reports cover image
Read More
EMBO Reports
Vol. 25 | No. 8
9 August 2024
Table of contents
Pages: 3202 - 3220

Submission history

Received: 28 February 2024
Revision received: 10 May 2024
Accepted: 10 June 2024
Published online: 2 August 2024
Published in issue: 9 August 2024

Permissions

Request permissions for this article.

Keywords

  1. Single-cell Omics
  2. Histone Posttranslational Modifications
  3. Nonhistone Chromatin Proteins
  4. Combinatorial Occurrence
  5. Chromatin States

Notes

9 October 2024: A Correction to this paper has been published: https://doi.org/10.1038/s44319-024-00269-5
A correction to this article is available online at https://doi.org/10.1038/s44319-024-00269-5.

Authors

Affiliations

Institute of Functional Epigenetics (IFE)Helmholtz Zentrum München Neuherberg Germany
Institute of Functional Epigenetics (IFE)Helmholtz Zentrum München Neuherberg Germany
Faculty of BiologyLudwig-Maximilians-Universität München Planegg-Martinsried Germany

Research Funding

Deutsche Forschungsgemeinschaft (DFG): SFB 1064 (Project-ID 213249687), SFB 1309 (Project-ID 325871075)
Helmholtz Gemeinschaft

Metrics & Citations

Metrics

Citations

Download Citations

If you have the appropriate software installed, you can download article citation data to the citation manager of your choice. Select your manager software from the list below and click Download.

View Options

View options

PDF

View PDF

Figures

Tables

Media

Share

Share

Copy the content Link

Share on social media