Skip to the content.

CUT&RUN

A simple method for chromatin profiling

Cleavage Under Targets and Release Using Nuclease (CUT&RUN) is an innovative chromatin mapping strategy that is rapidly gaining traction in the field. The protocol requires less than a day to go from from cells to DNA, and can be done entirely on the benchtop using standard equipment that is already present in most molecular biology laboratories.

How does it work?

  1. Cells/nuclei are bound to concanavalin A–coated magnetic beads.
    • In the original Henikoff lab paper they isolate nuclei. Using purified nuclei allows for maximal binding of antibodies to nuclear factors and will result in cleaner CUT&RUN signal compared to protocol using whole cells.
    • In the more recent paper, also from the Henikoff lab, whole cells are harvested. They introduce the use of a strong detergent to permeabilize cells rather than relying on the extraction of nuclei.
  2. Cell membranes (or nuclear membrances) are permeabilized with digitonin to allow the antibody access to its target (1h to overnight)
  3. The Protein A fused MNase is then added. Protein A binds the Immunoglobulin G (IgG) on the primary antibody (or mock IgG) thus targeting the MNase to antibody bound proteins.
  4. The nuclease is briefly activated to digest the DNA around the target protein. This targeted digestion is controlled by the release of (previously chelated) calcium, which MNase requires for its nuclease activity. The nuclease reaction is performed on ice, and only for a short period of time, thus precisely controlling the amount of cutting and thereby mitigating noise generated by off target digestion.
  5. At this point mononucleosomal-sized DNA fragments from a different organism is added (spike-in DNA).
  6. Fragments are released from nuclei by a short incubation at 37 °C.
  7. These short DNA fragments are then purified for subsequent library preparation and high-throughput sequencing.

Image source: “AddGene Blog”

What about CUT&TAG?

For the Cleavage Under Targets and Tagmentation assay, pAG is fused to a hyperactive Tn5 transposase (pAG-Tn5) pre-loaded with sequencing adaptors, and is activated by magnesium to simultaneously fragment and “tag” antibody-labelled chromatin with adaptors. This bypasses traditional library prep steps and accelerates sample processing. However, it only works well with nuclei.

Both assays were developed in the laboratories of Dr. Steven Henikoff (Fred Hutchinson Cancer Research Center, Seattle, WA, USA) and Dr. Ulrich Laemmli (University of Geneva, Switzerland).

CUT&RUN versus ChIP-seq

ChIP-seq is a notoriously challenging approach, in which target fragments are enriched from excess pools of sheared chromatin. Despite rigorous optimization and washing, the method is subject to high background. The resulting low signal to noise ratio makes it difficult to identify true binding sites.

Advantages of CUT&RUN

Limitations of CUT&RUN

Image source: Epicypher Blog

Controls for CUT&RUN

Negative (background) control

Similar to ChIP-seq, only a fraction of the DNA in at CUT&RUN sample will correspond to actual signal amidst background noise. However, CUT&RUN is thought to produce a sharper stronger signal, so do we need an input control?

Rather than having an input DNA control, the use of a nonspecific rabbit IgG antibody is recommended by the Henikoff lab. It will randomly coat the chromatin at low efficiency without sequence bias. While a no-antibody input DNA sample will generate a more diverse DNA library, the lack of tethering increases the possibility that slight carryover of pA-MN will result in preferential fragmentation of hyperaccessible DNA.

Positive control

To assess protocol efficacy, it is recommended to run, in parallel, a control CUT&RUN with an antibody against a histone mark. How does it help? After quantifying purified DNA, run it on the Bioanalyzer system. Transcription factor CUT&RUN DNA are typically around 50–150 bp and may not show up on the bioanalyzer electropherogram if run prior to amplification. This is due to the low concentration of DNA present. However, with the control histone mark CUT&RUN, you should see mono-, di-, and tri-nucleosomes in the Bioanalyzer traces. In the figure below, H3K4me3 and H3K27me3 libraries are predominantly enriched for mononucleosomes as indicated by the peak at 275 bp (~150 bp mononucleosomes + 125 bp sequence adapters). Bioanalyzer traces are the best indicator of success prior to sequencing.

Typical Bioanalyzer traces for IgG negative control and H3K4me3 / H3K27me3 positive control CUT&RUN sequencing libraries. Image source: Epicypher CUT&RUN Protocol v1.5.4

Analysis of CUT&RUN

Quality Control

A number of metrics are used to evaluate the quality of a CUT&RUN dataset, including:

Alignment

CUT&RUN libraries are sequenced using paired-end reads which are are aligned by Bowtie2. There are a few additional parameters that are used by various groups, which are not typically used with ChIP-seq data. They are listed and described below:

CUT&RUN sequencing workflow. Image source: Adapted from CUT&RUNTools: a flexible pipeline for CUT&RUN processing and footprint analysis

It is not neccessary to include all or any of these options! We just want to bring to your attention that some combination of these options have been used by other groups. We encourage you to read through th literature and decide what is best for your data.

To trim or not to trim?

Trimming is optional.

Peak calling

For the most part, MACS2 remains the peak caller of choice for CUT&RUN data. After alignment, fragments can be divided into ≤ 120-bp and > 120-bp fractions. For transcription factors or proteins with an expected punctate binding profile, you can use the ≤ 120-bp fraction which is likely to contain binding sites. The range can be increased depending on the protein of interest, and alternatively BAM files without filtering can also be used. The default narrow peak setting will suffice.

Another recent approach developed by the Henikoff Lab is a tool called SEACR. Peak calling by Sparse Enrichment Analysis for CUT&RUN sequencing data, provides an analysis strategy that uses the global distribution of background signal to calibrate a simple threshold for peak calling.

ChIP-seq experiments are typically sequenced deeply and thus feature high background, thus most peak calling algorithms designed for this type of data. By contrast, CUT&RUN data features exceedingly low background and low sequence depth. The sparseness of the background can increase false positives, resulting in any spurious background read being called as a peak. Thus, rather than requiring highly sensitive methods to distinguish signal from background noise, peak calling from CUT&RUN data requires high specificity for true positive peaks.

Image source: “Peak calling by Sparse Enrichment Analysis for CUT&RUN chromatin profiling”

Features of SEACR include:


This lesson has been developed by members of the teaching team at the Harvard Chan Bioinformatics Core (HBC). These are open access materials distributed under the terms of the Creative Commons Attribution license (CC BY 4.0), which permits unrestricted use, distribution, and reproduction in any medium, provided the original author and source are credited.