Fact-checked by Grok 2 weeks ago

MNase-seq

MNase-seq, short for micrococcal nuclease sequencing, is a high-throughput genomic technique designed to map nucleosome occupancy and positioning across eukaryotic genomes at high resolution. The method relies on the enzymatic activity of micrococcal nuclease (MNase), an endonuclease derived from Staphylococcus aureus that preferentially cleaves accessible linker DNA between nucleosomes while protecting the approximately 147 base pairs of DNA wrapped around histone octamers. These protected mononucleosomal DNA fragments, typically 100–200 base pairs in length, are isolated, size-selected, and subjected to paired-end sequencing, with nucleosome positions inferred from the midpoints of sequencing reads or through advanced computational modeling. This approach provides a genome-wide snapshot of chromatin architecture, enabling the study of nucleosome spacing, phasing, and dynamic changes in response to cellular processes. The core principle of MNase-seq stems from the nuclease's sensitivity to chromatin accessibility, where well-positioned nucleosomes shield DNA from digestion, resulting in distinct fragment length distributions that correlate with nucleosome density. Early applications focused on yeast and model organisms to delineate periodic nucleosome arrays at promoters and enhancers, but refinements such as quantitative MNase-seq (qMNase-seq) have addressed limitations like enzymatic sequence bias—where MNase digests A/T-rich linkers up to 30 times faster than G/C-rich ones—and over-digestion artifacts through kinetic modeling and spike-in normalization with foreign DNA. These improvements allow for precise quantification of nucleosome occupancy levels, distinguishing stable from transient nucleosomes and revealing subtle variations in chromatin accessibility. MNase-seq has become a cornerstone for investigating epigenetic regulation, as nucleosome positioning influences transcription factor binding, DNA accessibility, and higher-order chromatin structures like topologically associating domains (TADs). Key applications include profiling nucleosome barriers at active promoters in Drosophila and mapping chromatin remodeling during development or disease states, such as cancer. Despite its prevalence, the technique's reliance on limited digestion conditions can introduce noise, necessitating computational tools like iNPS or NucHMM for bias correction and fuzzy nucleosome detection. Overall, MNase-seq complements other chromatin profiling methods, such as ATAC-seq or chemical mapping, by offering detailed insights into the foundational role of nucleosomes in genome function.

Background and History

Discovery of Micrococcal Nuclease

Micrococcal (MNase), also known as staphylococcal , was first discovered in 1956 as an extracellular secreted by the bacterium . This was initially isolated and partially for its ability to hydrolyze nucleic acids, marking it as a novel with both endonucleolytic and exonucleolytic activities. In 1966, MNase was successfully crystallized in a phosphatase-free form, enabling further structural studies and purification advancements that improved its usability in biochemical assays. A comprehensive enzymatic followed in 1967, revealing MNase's specificity for cleaving phosphodiester bonds in DNA and RNA to produce 3'-mononucleotides and oligonucleotides, with a requirement for calcium ions as a cofactor. MNase exhibits dual endo- and exonucleolytic properties, preferentially digesting single-stranded nucleic acids over double-stranded forms, though it can cleave both; in chromatin contexts, it selectively targets regions between nucleosomes. Its optimal activity occurs at 37°C and 8–9, where it demonstrates and broad substrate tolerance, making it suitable for controlled experiments. During the 1970s, MNase became a key tool for probing structure, with early applications demonstrating its utility in limited digestion assays that revealed the modular of eukaryotic . Early studies noted MNase's preference for A/T-rich linkers, influencing patterns. In seminal experiments, partial digestion of followed by produced characteristic "laddering" patterns of DNA fragments at multiples of approximately 200 base pairs, reflecting the periodic spacing of s. These findings, notably from Noll in 1974 and collaborative work by Noll and Kornberg in 1977, confirmed the nucleosome core particle as a stable unit comprising about 146 base pairs of DNA wrapped around an octamer of proteins, with susceptible to MNase . Such biochemical insights laid the groundwork for later adaptations of MNase in genome-wide studies during the .

Evolution to Genome-Wide Sequencing

The transition from classical digestion experiments using micrococcal nuclease (MNase) to genome-wide sequencing approaches marked a pivotal shift in studying architecture, enabling high-throughput analysis of positioning and occupancy across entire genomes. Building on the foundational role of MNase in early studies from the 1970s, researchers began adapting the enzyme for large-scale mapping by combining it with emerging genomic technologies. The first genome-wide application of MNase-based nucleosome mapping occurred in 2006, when Johnson et al. utilized high-density tiling arrays to profile nucleosome core landscapes in Caenorhabditis elegans. By hybridizing MNase-protected DNA fragments to arrays covering the worm genome, they identified over 1.4 million nucleosome positions, revealing patterns of flexibility in nucleosome placement influenced by local sequence features and revealing periodicities in AT-rich linkers. This study demonstrated the feasibility of genome-scale profiling, highlighting translational constraints at promoters and exons while showing greater positional variability in introns. In 2007, efforts to incorporate sequencing for higher resolution advanced the field, with Albert et al. applying Roche 454 pyrosequencing to short MNase-protected fragments (~150 bp) in to map H2A.Z variant s. This approach sequenced mononucleosomal DNA directly, achieving base-pair resolution for ~67,000 s and demonstrating the practicality of next-generation sequencing for capturing protected fragments without reliance on arrays, thus paving the way for unbiased, high-throughput nucleosome profiling. Concurrently, Lee et al. used tiling arrays in to generate a comprehensive atlas of over 70,000 nucleosome positions, further validating genome-wide occupancy patterns and their correlation with . The method extended to mammalian systems in 2008, when Schones et al. applied deep sequencing to MNase-digested from human CD4+ T cells, producing the first comprehensive occupancy maps for the . Using Illumina sequencing, they aligned millions of reads to identify positioned , uncovering dynamic changes in positioning upon T-cell activation, such as depletion at transcription start sites and periodic arrays downstream. This work highlighted the role of nucleosome remodeling in immune response and established sequencing as superior for detecting cell-type-specific variations. Around this time, the term "MNase-seq" was coined by et al. in 2009, who developed a high-resolution for analyzing Illumina-sequenced MNase fragments in , standardizing nomenclature for the technique and emphasizing its utility in resolving fuzzy versus well-positioned nucleosomes at promoters. These advancements were enabled by next-generation sequencing platforms, particularly Illumina's high-throughput short-read technology introduced in the mid-2000s, which allowed cost-effective mapping of ~150 bp nucleosomal fragments at single-base resolution across billions of reads. This technological leap shifted MNase-based assays from array-limited snapshots to scalable, quantitative genome-wide surveys, influencing subsequent epigenomic studies.

Principles and Methodology

Biochemical Mechanism

Micrococcal nuclease (MNase), an extracellular enzyme from , preferentially cleaves the phosphodiester bonds in regions between nucleosomes, thereby protecting the approximately 147 (bp) DNA core wrapped around octamers. This selective occurs because nucleosome-bound DNA is shielded from the enzyme's due to its tight association with histones, while exposed remains accessible. Under limited conditions, MNase generates a ladder of protected fragments, with mononucleosomes at ~150 bp and dinucleosomes at ~300 bp, reflecting periodic protection patterns that allow inference of nucleosome positioning. Over-digestion, however, can trim nucleosomal DNA from the edges, producing sub-nucleosomal fragments of 100–140 bp, which reveal finer structural details but may introduce biases if not controlled. The biochemical mechanism of MNase involves a calcium-dependent that facilitates the of phosphodiester bonds. MNase requires two Ca²⁺ ions for activity; one Ca²⁺ polarizes the scissile through ionic with a non-bridging oxygen, while the other coordinates with residues like Asp-21, Asp-40, and Glu-43 to position a nucleophilic . This , activated by Glu-43 acting as a general , attacks the phosphorus atom, cleaving the 5'-P-O bond and yielding a 5'-hydroxyl group and a 3'- monoester product. The enzyme exhibits both endo- and activities, with endonucleolytic cleavage preferred in double-stranded DNA contexts like . In chromatin, MNase digestion kinetics follow a pseudo-first-order model, where the rate of linker DNA cleavage (k₁) exceeds that of nucleosomal DNA decay (k₂), leading to sequential release of nucleosomes from bulk chromatin. This can be described by the differential equations for the reaction chain (bound chromatin B → free nucleosome N → degraded fragments Ø): \frac{d[B]}{dt} = -k_1 [E][B], \quad \frac{d[N]}{dt} = k_1 [E][B] - k_2 [E][N] The solution for nucleosome concentration is N(t) = C_0 \frac{k_1}{k_2 - k_1} (e^{-k_1 [E] t} - e^{-k_2 [E] t}), highlighting exponential decay of linker DNA and slower degradation of protected cores. In euchromatic regions, which are more open and A/T-rich, cleavage proceeds faster due to enhanced accessibility, resulting in quicker nucleosome release; conversely, compact heterochromatin resists digestion, producing stronger occupancy signals in sequencing reads. MNase's specificity contrasts with other endonucleases like DNase I, as it primarily maps occupied, protected nucleosomal regions rather than hypersensitive, accessible sites, enabling orthogonal profiling of chromatin structure. This protection-based approach underscores MNase's utility in revealing nucleosome occupancy without directly targeting regulatory elements.

Standard Experimental Protocol

The standard experimental protocol for bulk MNase-seq involves isolating chromatin from eukaryotic cells, digesting it with micrococcal nuclease (MNase) to generate nucleosomal DNA fragments, purifying mononucleosomal DNA, and preparing libraries for next-generation sequencing (NGS). This approach relies on MNase's preferential cleavage of linker DNA between nucleosomes, producing a characteristic laddering pattern of DNA fragments corresponding to mono-, di-, and higher-order nucleosomes. Sample preparation begins with harvesting 1–10 million cells, typically cultured mammalian cell lines such as GM12878 or K562, grown to log phase and cross-linked with 1% for 10 minutes at to stabilize and prevent displacement during handling. Cells are then pelleted by , washed in , and resuspended in hypotonic (e.g., 10 mM Tris-HCl pH 7.4, 10 mM NaCl, 3 mM MgCl₂, 0.5% ) to swell and rupture the plasma membrane, releasing cytoplasmic contents while preserving nuclear integrity. Nuclei are isolated by low-speed (500–1,000 × g, 5 minutes at 4°C), resuspended in a storage buffer (e.g., 50 mM Tris-HCl pH 7.5, 40 mM NaCl, 100 mM , 5 mM MgCl₂, 1 mM CaCl₂, 0.1 mM ), and quantified by DNA content using a spectrophotometer or , aiming for 5–50 μg total DNA. This step yields intact nuclei suitable for digestion, with yields scalable using low-input modifications for precious samples down to 0.5 million cells. MNase digestion is performed on isolated nuclei to fragment chromatin selectively at linker regions. Nuclei are equilibrated in digestion buffer (e.g., 50 mM Tris-HCl pH 7.9, 5 mM CaCl₂) and titrated with MNase (typically 0.05–0.5 U per μg DNA, ranging from 10–250 total units depending on cell type) for 5–15 minutes at 37°C to achieve primarily mononucleosomal fragments. Digestion is stopped by adding EDTA to 10 mM and chilling on ice, followed by reversal of formaldehyde cross-links at 65°C for 4–6 hours in the presence of proteinase K (0.2 mg/mL). Limited digestion (lower enzyme units, shorter time) preserves di- and trinucleosomes for assessing higher-order chromatin structure, while extensive digestion (higher units) enriches mononucleosomes (~147 bp protected DNA) for precise positioning maps, with cell-type optimizations such as adjusted Ca²⁺ concentrations for tissues like liver. Purification of mononucleosomal DNA follows digestion to isolate fragments of 100–200 bp for NGS. Chromatin is treated with RNase A (0.1 mg/mL, 37°C, 30 minutes) to remove , then DNA is extracted using phenol-chloroform-isoamyl alcohol (25:24:1) followed by , or alternatively via spin columns (e.g., Zymo Research DNA Clean & Concentrator) for higher throughput. The ~150 bp mononucleosomal band is size-selected using (1.5% ) or automated systems like BluePippin, yielding 100–500 ng DNA. End-repair is performed with T4 DNA polymerase and , followed by A-tailing with Klenow exo-minus, and ligation of Illumina-compatible adapters using T4 , per standard NGS kits (e.g., NEBNext Ultra II). Libraries are amplified by (12–15 cycles) and quantified by qPCR. Quality control ensures optimal digestion and library integrity. Post-digestion DNA is assessed on a Bioanalyzer or TapeStation for laddering, targeting ~80% mononucleosomal DNA with minimal sub- or higher-order fragments to confirm even cleavage. Yield and purity are verified by NanoDrop (A260/A280 > 1.8), and library fragment distribution post-adapter ligation should peak at ~250–300 bp (including adapters). Over-digestion (excess sub-nucleosomal DNA <100 bp) or under-digestion (prominent higher ladders) prompts titration adjustments. Sequencing typically requires 20–50 million reads per sample for genome-wide coverage.

Data Processing and Analysis

The processing of MNase-seq data begins with and preprocessing of raw sequencing reads, typically in . Initial assessment involves tools like FastQC to evaluate read quality metrics, including per-base sequence quality, GC content, and adapter contamination, ensuring high-quality input for downstream analysis. Trimming of low-quality bases and adapters is performed using software such as Trim Galore or Cutadapt if necessary, followed by alignment to a using aligners like Bowtie2 for efficient handling of short reads or for splice-aware mapping in eukaryotic contexts. Aligned reads in BAM format are then sorted and indexed with , and duplicates—arising from amplification—are marked and removed using MarkDuplicates to mitigate bias in signal estimation. Post-alignment, fragment size filtering isolates mononucleosome-protected DNA, typically retaining paired-end reads with insert sizes between 140 and 180 base pairs, which correspond to the core length plus , while excluding shorter subnucleosomal or longer polynucleosomal fragments. Normalization accounts for sequencing depth and library size differences, commonly using reads per million (RPM) to scale counts, enabling comparable profiles across samples; for quantitative comparisons, spike-in controls may be incorporated to adjust for global changes. Specialized tools then perform peak calling and scoring: , an R/ package, identifies positions via non-parametric scanning of smoothed coverage, distinguishing well-positioned from fuzzy nucleosomes based on signal . Similarly, DANPOS2 quantifies by calculating as protection scores and fuzziness as the variance in dyad positions across replicates, with higher fuzzy scores indicating dynamic or delocalized nucleosomes. Visualization and interpretation leverage genome browsers like Integrative Genomics Viewer (IGV) to display aligned reads or normalized tracks, revealing periodic arrays, while heatmaps generated with tools such as deepTools highlight phasing patterns around transcription start sites (TSS) or promoters. Processed data are often converted to bigWig format for efficient storage and integration, facilitating overlay with other epigenomic tracks. Quantitative metrics include , computed as the ratio of protected reads in defined windows (e.g., 147 bp) to total genomic coverage, providing insights into density; in advanced setups, this is refined using spike-in for values. Comprehensive pipelines, such as nf-core/mnaseseq, automate these steps from reads to maps, incorporating quality filtering and reproducibility checks. Custom /Bioconductor workflows with nucleR or DANPOS2 are widely adopted for flexibility, and batch effects in multi-sample datasets can be modeled using DESeq2 to for technical variation while preserving biological signals in counts. ENCODE-inspired standards emphasize uniform alignment parameters and metadata annotation to ensure interoperability across studies.

Core Applications

Nucleosome Positioning and Occupancy

MNase-seq enables the genome-wide mapping of phased arrays, particularly evident around transcription start sites (TSS), where a characteristic depletion occurs at the promoter followed by regularly spaced downstream. The +1 is typically positioned with its dyad approximately 40–100 base pairs downstream of the TSS, depending on the and promoter activity, with subsequent (+2, +3, etc.) forming periodic arrays at roughly 10-base pair resolution, reflecting the helical structure of DNA wrapped around the . This phasing is quantified through oscillatory patterns in read density from MNase-protected fragments, allowing precise delineation of boundaries and regions. Nucleosome occupancy profiles derived from MNase-seq reveal high coverage in gene bodies, where transcription-coupled positioning maintains stable arrays that facilitate processive RNA polymerase II elongation. In contrast, enhancers exhibit lower occupancy, enabling access for transcription factors and correlating positively with active histone modifications such as H3K27ac, which further destabilizes nucleosomes in these regulatory elements. Quantitative analysis of occupancy, often normalized by spike-in controls, distinguishes well-positioned nucleosomes (low fuzziness) from dynamic or depleted regions, providing insights into chromatin organization. Nucleosome positioning and occupancy are highly dynamic, varying across cell types; for instance, embryonic stem cells display more diffuse positioning at developmental genes compared to differentiated lineages, where sharpened arrays reinforce lineage-specific expression. In response to stimuli, such as T cell or hormone treatment, rapid repositioning occurs, with nucleosome eviction at inducible promoters facilitating transcriptional . These changes highlight MNase-seq's utility in capturing context-dependent . The sub-nucleosome resolution of MNase-seq, achieving approximately 10 precision, is particularly advantageous for identifying barrier nucleosomes at insulators, where positioned nucleosomes prevent ectopic interactions and maintain domain boundaries.

Chromatin Accessibility Profiling

MNase-seq indirectly profiles accessibility by exploiting the enzyme's preferential cleavage of in open regions, where nucleosomes provide less steric hindrance compared to compacted domains. Regions exhibiting rapid MNase digestion, characterized by an abundance of short fragments or reduced occupancy signals, signify accessible , while slower digestion rates in densely packed areas indicate protection. This approach reveals dynamic states, with accessible sites often corresponding to regulatory elements susceptible to enzymatic attack even at low MNase concentrations. Sub-nucleosomal fragments, typically 50-100 bp in length, arise from over-digestion in highly accessible loci and serve as hallmarks of open chromatin. These fragments, generated when MNase further trims protected nucleosomal DNA in vulnerable regions, preferentially mark active promoters and enhancers, where nucleosome instability facilitates transcription factor binding. For instance, protocols like MNase-SSP enhance detection of these short reads, improving resolution of sub-nucleosomal signals over standard MNase-seq. MNase-seq accessibility profiles integrate well with gene activity measures, such as , to link openness to transcriptional output. Nucleosome-free regions (NFRs) immediately upstream of transcription start sites (TSS) correlate with higher levels, as these depleted zones enable promoter access for . In active genes, NFRs and flanking positioned nucleosomes predict expression variance, with broader accessibility at TSS associating with elevated transcript abundance across cell types. Quantitative assessment of accessibility in MNase-seq often employs metrics like the MNase accessibility (MACC) score, derived as the slope of on fragment frequencies across digestion titrations, or the nucMACC score, which quantifies changes in mono- protection via log-transformed ratios of cleavage rates relative to nucleosome stability. These scores, validated against (eQTLs), highlight how accessibility variants influence regulatory function, with higher scores in open regions aligning with eQTL hotspots. In contexts, MNase-seq uncovers altered at regulatory elements, aiding of epigenetic drivers in pathologies like cancer. For example, comprehensive mapping in epithelial cells versus cancer lines reveals gain or loss of at tumor-specific enhancers, correlating with oncogenic gene dysregulation. Such profiles, using pipelines like nucMACC, detect fragile at cancer-associated motifs, such as binding sites, informing therapeutic targeting of epigenomic reprogramming.

Advanced Techniques and Variants

Antibody-Targeted Methods (CUT&RUN and CUT&Tag)

Antibody-targeted methods represent an evolution of MNase-based profiling, leveraging specific antibodies to direct enzymatic to protein-DNA interactions of interest, thereby enhancing precision and reducing off-target effects compared to unbiased approaches. These techniques build on the core principle of MNase digestion by tethering the enzyme or a related activity to antibody-bound targets within intact nuclei, allowing fragmentation that captures nearby DNA sequences for sequencing. CUT&RUN, introduced in , employs -MNase (pA-MNase), a fusion of and micrococcal , to target proteins. In this method, permeabilized nuclei are immobilized on magnetic beads and incubated with a primary specific to the target protein, such as a or modification. Secondary binding of pA-MNase tethers the near the target, and calcium activation triggers controlled cleavage, releasing ~120-150 bp fragments corresponding to protected nucleosomal DNA. The process occurs , avoiding the need for extraction or , and the released fragments are collected from the supernatant for direct library preparation and sequencing. CUT&Tag, developed in as a streamlined variant, replaces MNase with a protein A-Tn5 (pA-Tn5) to combine targeting with tagmentation. Following binding in permeabilized s or nuclei bound to Concanavalin A beads, pA-Tn5 is added and activated by magnesium, simultaneously cleaving DNA and appending sequencing adapters, which simplifies library preparation into a single-tube . This yields high-resolution profiles with fragment sizes around 80 bp for footprints, enabling efficient mapping from low inputs as few as 1,000 cells while minimizing background noise. Both methods offer key advantages over traditional ChIP-seq, including higher due to the absence of sonication-induced , substantially lower background signal from non-specific cleavage, and a exceeding 100-fold for detecting enrichment levels across genomic regions. CUT&RUN and CUT&Tag require fewer cells (typically 10,000-100,000) and less sequencing depth—often 10-fold lower—while providing base-pair precision for binding sites. details include a brief 10-minute calcium-activated for CUT&RUN on ice and direct tagmentation for CUT&Tag, followed by amplification; analysis pipelines like CUT&RUNTools facilitate , peak calling, and from the resulting short-read data. These techniques excel in applications such as mapping precise binding sites and modifications, revealing sub-nucleosomal details like protection patterns around motifs. For instance, CUT&RUN has delineated occupancy with sharp boundaries, while CUT&Tag profiles broad marks like across cell types. From 2020 to 2025, advancements include multiplexing strategies like Multi-CUT&Tag for simultaneous of multiple epitopes in the same cells and integrations with single-cell via methods such as Paired-Tag, enabling multi-omic correlations of epigenetic states with at cellular resolution.00753-X)

Single-Cell MNase-seq Adaptations

Single-cell adaptations of MNase-seq address the limitations of bulk methods by enabling the resolution of heterogeneity across individual cells, revealing variations in positioning and accessibility that are masked in population averages. The foundational technique, single-cell MNase-seq (scMNase-seq), was introduced to profile genome-wide occupancy and open regions simultaneously in single mammalian cells. Developed in 2018, this method applies MNase digestion to isolate nucleosome-protected fragments and accessible DNA subfragments from individual cells, providing insights into cell-to-cell differences in architecture. The experimental workflow for scMNase-seq begins with the isolation of single cells or nuclei via fluorescence-activated cell sorting (FACS) into multi-well plates, typically 96- or 384-well formats, to ensure high viability and purity. Following sorting, cells are lysed directly in the wells, and MNase is added for controlled digestion, generating subnucleosome-sized fragments (≤80 ) from accessible and mononucleosome-sized fragments (140–180 ) from core-protected regions. DNA is then purified using column-based kits, end-repaired, and ligated to Y-shaped adapters compatible with Illumina sequencing. Linear or low-cycle amplification with cell-specific indexing primers barcodes the libraries, minimizing amplification bias, and size selection recovers the relevant fragments for paired-end sequencing on platforms like HiSeq, yielding 0.5–1 million unique mapped reads per cell. This plate-based approach, detailed in a 2019 protocol, supports processing up to hundreds of cells per run and has been applied to diverse cell types including mouse embryonic stem cells and naive CD4+ T cells. Low-input adaptations of scMNase-seq are inherent to its single-cell design, requiring only 1–100 cells for library preparation, making it suitable for precious samples like sorted rare populations or dissociated tissues. While have revolutionized single-cell and , scMNase-seq primarily relies on nanowell or multi-well systems for precise control over digestion conditions, though hybrid integrations with droplet platforms for initial cell encapsulation have been explored to enhance throughput. These adaptations maintain the method's to detect phasing and accessibility with minimal material loss, outperforming bulk MNase-seq in resolving heterogeneous states within mixed populations. Analysis of scMNase-seq data presents challenges due to the sparsity and high dimensionality of single-cell profiles, where low read coverage per cell (often <1% genome coverage) leads to dropout events in low-occupancy regions. Computational pipelines align reads to reference genomes, separate sub- and peaks using size-based filtering, and quantify occupancy via fragmentation patterns. To address sparsity, imputation algorithms such as Markov affinity-based graph imputation of cells (), adapted from scRNA-seq, denoise data by propagating signals across similar cells in a diffusion framework. Dimensionality reduction with uniform manifold approximation and projection (UMAP) facilitates clustering of landscapes, enabling identification of cell subtypes based on shared motifs. These tools reveal principles like uniform spacing in versus variable positioning at active enhancers across cells. Applications of single-cell MNase-seq emphasize dissecting variability in and , such as detecting primed subpopulations in undifferentiated mouse embryonic stem cells with reduced nucleosome occupancy at de novo enhancers. In tissues, it profiles cell-type-specific nucleosome positioning, contributing to atlases of heterogeneity by highlighting accessibility differences in neuronal versus glial lineages. For , the method identifies rare tumor subpopulations with altered nucleosome arrays indicative of epigenetic dysregulation, aiding in the mapping of intratumor diversity. These insights underscore scMNase-seq's role in linking chromatin states to cellular identity and function. Recent advances from 2020 to 2025 have scaled scMNase-seq through combinatorial indexing strategies, inspired by sci-ATAC-seq, to profile over 10,000 cells in a single experiment by splitting and barcoding pools iteratively, reducing per-cell costs to approximately $0.1. Optimized analysis tools like scNucMap (2025) enhance -free region calling from sparse data, improving resolution for heterogeneity studies. These developments expand scMNase-seq's utility for large-scale epigenomic atlases while preserving its unbiased profiling of dynamics.

Comparisons to Other Assays

DNase-seq and ATAC-seq

DNase-seq, a genome-wide method adapted from earlier DNase I hypersensitivity assays developed in the 1970s–1980s, employs the nuclease DNase I to preferentially cleave DNA at hypersensitive sites (DHSs), generating fragments typically ranging from 10 to 500 bp that mark regions of open chromatin. This method requires approximately 10 million cells for library preparation and effectively identifies accessible genomic regions associated with regulatory elements, though it provides limited insight into nucleosome-level details due to its focus on cleavage rather than protection. In contrast, , introduced by Buenrostro et al. in 2013, utilizes hyperactive Tn5 for tagmentation, simultaneously fragmenting accessible DNA and inserting sequencing adapters, enabling rapid library preparation in just a few hours from as few as 500 to 50,000 cells. While highly efficient for low-input samples, is susceptible to biases, including overrepresentation of reads and artifacts in footprinting due to the transposase's insertion preferences. A fundamental distinction lies in their approaches to structure: MNase-seq maps -protected DNA regions, revealing occupancy and precise positioning, whereas DNase-seq and target exposed, accessible DNA, highlighting regions free of . This makes MNase-seq particularly orthogonal and advantageous for delineating arrays and barriers, offering higher specificity in compact contexts compared to the broader accessibility signals from DNase-seq and . excels in speed and minimal cell requirements but exhibits lower signal-to-noise ratios, especially in densely packed , where MNase-seq provides superior resolution for dynamics. Despite these differences, all three assays overlap in detecting regulatory elements such as promoters and enhancers, with MNase-seq complementing DNase-seq and by identifying nucleosomal barriers that constrain accessibility in those regions.

FAIRE-seq and NOMe-seq

FAIRE-seq, or Formaldehyde-Assisted Isolation of Regulatory Elements, is a chemical-based method developed in to isolate nucleosome-depleted DNA regions associated with regulatory activity. The protocol involves crosslinking with 1% to stabilize protein-DNA interactions, followed by to shear the into fragments of approximately 0.5–1 kb. The sheared is then subjected to phenol-chloroform , where nucleosome-bound DNA partitions into the organic phase due to its association with crosslinked proteins, while open, nucleosome-free DNA enriches in the aqueous phase for recovery and sequencing. This approach typically requires at least 100,000 cells and provides enrichment for accessible regions, though with a resolution limited to around 1 kb owing to the shearing process. NOMe-seq, or Nucleosome Occupancy and Methylome sequencing, introduced in 2012, employs a GpC-specific methyltransferase (M.CviPI) to probe accessibility while simultaneously mapping . In the procedure, isolated nuclei from intact cells are treated with M.CviPI in the presence of , which methylates accessible GpC dinucleotides in open ; nucleosome-protected regions remain unmethylated, creating accessibility footprints. The DNA is then extracted, fragmented to ~200 bp, bisulfite-converted to distinguish endogenous CpG from GpC marks, and sequenced to generate dual maps of occupancy and at single-molecule . NOMe-seq requires fewer than 1 million intact cells and excels at revealing fine-scale footprints, such as nucleosome-depleted regions at promoters, but excludes certain trinucleotides like GCG due to sequencing ambiguities. In contrast to MNase-seq's enzymatic digestion with micrococcal nuclease, which is tunable for ladder resolution, FAIRE-seq relies on chemical crosslinking that can introduce artifacts from non-specific protein associations, while NOMe-seq uses enzymatic without digestion, providing context absent in standard MNase-seq. MNase-seq offers precise positioning but may bias toward AT-rich sequences, whereas FAIRE-seq avoids enzymatic biases through physical separation, though it suffers from higher noise and lower signal-to-noise ratios due to crosslinking variability. NOMe-seq adds value by integrating with endogenous but demands intact cells and can be affected by off-target methyltransferase activity on CpGs. FAIRE-seq's strengths include its simplicity, lack of sequence bias, and ability to capture lipid-associated open regions unbiased by nucleases, making it suitable for diverse cell types; however, it is noisier and less precise for ultra-short open elements compared to MNase-seq's -level accuracy. NOMe-seq provides high-resolution footprints and dual epigenomic layers but is more complex due to processing, while MNase-seq remains simpler for pure occupancy profiling without readout. Studies in the 2010s and 2020s have explored integrating MNase-seq with NOMe-seq in multi-omics frameworks to construct comprehensive epigenomic maps, such as combining positioning data with and accessibility profiles for studying promoter states in lines. These approaches leverage bioinformatics to correlate datasets, enhancing insights into dynamics beyond what either method achieves alone.

References

  1. [1]
    Quantitative MNase-seq accurately maps nucleosome occupancy ...
    Sep 13, 2019 · By considering the kinetics of nucleosome release from chromatin by MNase, we have developed a rigorous method for analyzing MNase digestion ...
  2. [2]
    MAINE-Seq/Mnase-Seq/Nucleo-Seq - Illumina
    In MNase-Seq, gDNA is treated with MNase. Sequences bound by chromatin proteins are protected from MNase digestion. Next, the DNA from the DNA-protein complexes ...
  3. [3]
    Genome-wide mapping of the nucleosome landscape by ... - PMC
    First, MNase-seq is a nuclease protection assay that can determine nucleosome positions, but its accuracy is limited by sequence bias and over-digestion of ...
  4. [4]
    Mapping nucleosome and chromatin architectures: A survey of ...
    1). MNase-seq, a profiling method for mapping nucleosome landscape, is the most prevalent technique to study nucleosome organization.
  5. [5]
    Nuclease, Micrococcal - Worthington Enzyme Manual
    Micrococcal nuclease (MN) catalyzes cleavage of both DNA and RNA to yield 3'-nucleotides (Alexander et al. 1961). MN is the extracellular nuclease of ...
  6. [6]
    Flexibility and constraint in the nucleosome core landscape of ... - NIH
    Flexibility and constraint in the nucleosome core landscape of Caenorhabditis elegans chromatin. Steven M Johnson. Steven M Johnson. 1Department of Pathology ...
  7. [7]
    Volatile accretion history of the terrestrial planets and dynamic implications - Nature
    No readable text found in the HTML.<|control11|><|separator|>
  8. [8]
    The Effect of Micrococcal Nuclease Digestion on Nucleosome ...
    A common experimental approach to identify nucleosome positions involves the use of Micrococcal nuclease (MNase). MNase preferentially cuts linker DNA ...
  9. [9]
    Micrococcal Nuclease Does Not Substantially Bias Nucleosome ...
    The study found that biases in nucleosome positioning data collected using micrococcal nuclease (MNase) are not significant under the conditions tested.Missing: biochemical | Show results with:biochemical
  10. [10]
  11. [11]
    Micrococcal nuclease - M-CSA Mechanism and Catalytic Site Atlas
    Staphyloccal nuclease is a Ca2+ activated phosphodiesterase which catalyses the hydrolysis of both DND and RNA at the 5' position of the phophodiester bond.Missing: dependent phosphodiester
  12. [12]
    Whole-genome methods to define DNA and histone accessibility ...
    MNase-seq: MNase is used to digest away linker DNA regions and nucleosomal DNA is sequenced. D. ATAC-seq: A hyperactive Tn5 transposase simultaneously ...Missing: decay | Show results with:decay
  13. [13]
  14. [14]
    Standardized collection of MNase-seq experiments enables ...
    May 6, 2012 · Our standardized approach to MNase-seq experiments was designed to enable the collection of similarly sampled chromatin populations which ...
  15. [15]
    MNase titration reveals differences between nucleosome occupancy ...
    May 6, 2016 · The data on histone marks and DNaseI hypersensitivity (DHS) were taken from Kharchenko et al. ... a single map of nucleosome occupancy.
  16. [16]
    Standardized collection of MNase-seq experiments enables ...
    May 6, 2012 · We present a standardized chromatin preparation which controls for technical variance between MNase-based chromatin preparations and enables the collection of ...
  17. [17]
    An efficient targeted nuclease strategy for high-resolution mapping ...
    An MNase-seq profile is shown in grey. Ticks mark the location of ... Nuclei were isolated by hypotonic lysis in 1 ml NE1 (20 mM HEPES-KOH pH 7.9; 10 ...
  18. [18]
    Rapid and inexpensive preparation of genome-wide nucleosome ...
    Jun 18, 2021 · This method for rapidly producing sequencing-ready nucleosome footprints from several organisms makes MNase-seq faster and easier, with less chemical waste.Protocol · Make Spheroplasts And Digest... · Expected OutcomesMissing: paper | Show results with:paper
  19. [19]
    GSM1694835 - GEO Accession viewer - NIH
    May 15, 2019 · Micrococcal nuclease (MNase) digestion of chromatin was performed by the addition of 10 (MNase-LOW) to 250 (MNase-HIGH) total units of MNase ( ...
  20. [20]
    Protocol for the genomic analysis of salt-fractionated chromatin from ...
    May 2, 2024 · Nuclei isolation and MNase digestion. Timing: day 2, 1 h. This section describes the procedures for isolating intact nuclei from frozen liver ...
  21. [21]
    [PDF] High Sensitivity Profiling of Chromatin Structure by MNase-SSP
    Feb 26, 2019 · In Brief. Ramani et al. describe MNase-SSP, a single-stranded DNA sequencing library preparation method for profiling chromatin structure.
  22. [22]
    None
    ### MNase-seq Protocol Summary
  23. [23]
    FastQC A Quality Control tool for High Throughput Sequence Data
    FastQC aims to provide a simple way to do some quality control checks on raw sequence data coming from high throughput sequencing pipelines.Bad Illumina Data · Index of /projects/fastqc/Help · Good Illumina · Publications
  24. [24]
  25. [25]
    nucleR: a package for non-parametric nucleosome positioning
    nucleR is an R/Bioconductor package for a flexible and fast recognition of nucleosome positioning from next generation sequencing and tiling arrays experiments.
  26. [26]
    nf-core/mnaseseq: MNase-seq analysis pipeline using ... - GitHub
    nfcore/mnaseseq is a bioinformatics analysis pipeline used for DNA sequencing data obtained via micrococcal nuclease digestion. The pipeline is built using ...Missing: processing | Show results with:processing
  27. [27]
    Data Processing Pipelines - ENCODE
    DNA accessibility assays such as DNase-seq, ATAC-seq, FAIRE-seq, and MNase-seq are common assays that support the goals of the ENCODE project. DNase-seq ...
  28. [28]
    Dynamic Regulation of Nucleosome Positioning in the Human ...
    Mar 7, 2008 · We have generated genome-wide maps of nucleosome positions in both resting and activated human CD4 + T cells by direct sequencing of nucleosome ends.
  29. [29]
    A computational approach to map nucleosome positions and ... - eLife
    Sep 13, 2016 · Here we develop a computational approach leveraging digestion variability to determine nucleosome positions at a base-pair resolution from MNase-seq data.<|control11|><|separator|>
  30. [30]
    Nucleosomal occupancy changes locally over key regulatory ...
    Aug 27, 2014 · Using normalized MNase-seq data, we initially examined nucleosome occupancy of promoters in relation to both the transcriptional status of the ...Missing: history original
  31. [31]
    Nucleosome positioning changes during human embryonic stem ...
    Nucleosome positioning (NP) plays a key role in transcriptional regulation and other biological processes. To better understand NP we used MNase-seq to ...
  32. [32]
    Controls of Nucleosome Positioning in the Human Genome
    Ernst J, Kheradpour P, Mikkelsen TS, Shoresh N, Ward LD, et al. (2011) Mapping and analysis of chromatin state dynamics in nine human cell types. Nature 473 ...
  33. [33]
    High Sensitivity Profiling of Chromatin Structure by MNase-SSP
    Feb 26, 2019 · Here we introduce MNase-SSP, a single-stranded sequencing library preparation method for nuclease-digested chromatin that enables simultaneous mapping of TF ...
  34. [34]
    Chromatin interaction maps reveal genetic regulation for quantitative ...
    Jun 14, 2019 · Bottom, histone modifications, RNAPII occupancy, chromatin accessibility (MNase-Seq) ... These genes were referred to as eQTL interacting genes ( ...
  35. [35]
    Comprehensive nucleosome mapping of the human genome in ...
    Mar 22, 2016 · Altered chromatin structure is a hallmark of cancer, and inappropriate regulation of chromatin structure may represent the origin of ...Comprehensive Nucleosome... · Results · Mtss-Seq Identifies Specific...
  36. [36]
    nucMACC: An MNase-seq pipeline to identify structurally altered ...
    Jul 3, 2024 · Micrococcal nuclease sequencing is the state-of-the-art method for determining chromatin structure and nucleosome positioning.
  37. [37]
    An efficient targeted nuclease strategy for high-resolution mapping ...
    Jan 12, 2017 · A new method, called CUT&RUN (which is short for “Cleavage Under Targets & Release Using Nuclease”) to map specific interactions between protein and DNA.
  38. [38]
    An efficient targeted nuclease strategy for high-resolution mapping ...
    We describe Cleavage Under Targets and Release Using Nuclease (CUT&RUN), a chromatin profiling strategy in which antibody-targeted controlled cleavage by ...
  39. [39]
    CUT&Tag for efficient epigenomic profiling of small samples ... - Nature
    Apr 29, 2019 · Skene, P. J. & Henikoff, S. An efficient targeted nuclease strategy for high-resolution mapping of DNA binding sites. Elife 6, e21856 (2017).<|control11|><|separator|>
  40. [40]
    CUT&Tag for efficient epigenomic profiling of small samples and ...
    CUT&Tag for efficient epigenomic profiling of small samples and single cells ... Nat Commun. 2019 Apr 29;10(1):1930. doi: 10.1038/s41467-019-09982-5.
  41. [41]
    CUT&RUNTools 2.0: a pipeline for single-cell and bulk-level ...
    Here, we have developed a flexible pipeline for analysis and visualization of single-cell CUT&Tag and CUT&RUN ... Skene and Henikoff, 2017). As a result, it has ...
  42. [42]
  43. [43]
  44. [44]
    Chromatin accessibility: a window into the genome - PubMed Central
    Nov 20, 2014 · MNase-seq has been implemented in a number of organisms, ranging from yeast to humans, for the mapping of chromatin structure [47–49]. In ...Missing: coining | Show results with:coining
  45. [45]
    From reads to insight: a hitchhiker's guide to ATAC-seq data analysis
    Feb 3, 2020 · In conclusion, integrating ChIP-seq and ATAC-seq helps to understand TF and histone facilitated chromatin accessibility changes. We foresee ...
  46. [46]
    Genomic methods in profiling DNA accessibility and factor localization
    Nov 27, 2019 · While MNase-seq has traditionally ... seq typically requires millions of input cells, particularly to examine transcription factor binding.
  47. [47]
    FAIRE (Formaldehyde-Assisted Isolation of Regulatory Elements ...
    Here we demonstrate a simple procedure for the isolation of nucleosome-depleted DNA from human chromatin, termed FAIRE (Formaldehyde-Assisted Isolation of ...Missing: original paper
  48. [48]
    Using FAIRE (Formaldehyde-Assisted Isolation of Regulatory ...
    Sep 26, 2013 · FAIRE has been used to create catalogs of regulatory elements in normal or diseased cells, narrow the search space for causal sequence variants ...Missing: original paper
  49. [49]
    Genome-wide mapping of nucleosome positioning and DNA ...
    Sep 7, 2012 · NOMe-seq maps nucleosome positioning and DNA methylation using a methyltransferase and next-gen sequencing, showing an anti-correlation at CTCF ...Missing: paper 2013
  50. [50]
    Genome-wide mapping of nucleosome positioning and DNA ...
    Using next generation sequencing, we describe a genome-wide nucleosome footprinting method termed NOMe-seq (nucleosome occupancy and methylome sequencing), ...
  51. [51]
    Chromatin accessibility: a window into the genome
    Nov 20, 2014 · MNase digestion simulation experiments have provided evidence that nucleosome configurations with or near long linkers are sampled easier ...Missing: euchromatin | Show results with:euchromatin
  52. [52]
    Links between DNA methylation and nucleosome occupancy in the ...
    Apr 11, 2017 · The results demonstrated that increasing methylated CpG density is correlated with nucleosome occupancy in the total genome and within nearly all subgenomic ...Genome Silencing By Dna... · Dna Methylation... · MethodsMissing: NOR | Show results with:NOR
  53. [53]
    Nucleosome-Omics: A Perspective on the Epigenetic Code and 3D ...
    Jun 22, 2022 · Micrococcal nuclease sequencing (MNase-seq) is a sequencing technology first developed to study nucleosome localization. This technology ...