Fact-checked by Grok 2 weeks ago

ChIP-on-chip

ChIP-on-chip, also known as ChIP-chip, is a technique that integrates (ChIP) with hybridization to map the genome-wide binding sites of DNA-associated proteins, such as transcription factors and modifications, by identifying enriched DNA fragments relative to total input DNA. Developed in 2000 by Bing Ren and colleagues at the Whitehead Institute, the method was first applied to the yeast to locate the binding sites of transcription activators like Gal4 and Ste12, revealing their roles in regulating specific metabolic and signaling pathways. The core workflow begins with formaldehyde cross-linking of protein-DNA interactions , followed by chromatin fragmentation via , selective using antibodies specific to the target protein, reversal of cross-links to release bound DNA, amplification and fluorescent labeling (typically with Cy3 or Cy5 dyes), and hybridization to high-density tiling microarrays that cover intergenic or promoter regions of the genome. Enrichment is then quantified by comparing fluorescence intensities between immunoprecipitated samples and reference input DNA, allowing identification of binding peaks through statistical analysis. This technique revolutionized the study of gene regulation by enabling high-throughput, unbiased profiling of protein-DNA interactions across entire genomes, which previously relied on low-throughput methods like reporter assays or electrophoretic mobility shift assays. Early applications demonstrated its utility in uncovering novel regulatory networks, such as Gal4's control of carbon metabolism genes and Ste12's involvement in pheromone response pathways, while also integrating binding data with gene expression profiles to distinguish direct from indirect targets. Over time, ChIP-on-chip was extended to more complex organisms like Drosophila melanogaster and humans, facilitating studies of histone modifications, nucleosome positioning, and epigenetic landscapes through projects like modENCODE. Despite its advantages in reproducibility (with intra-platform correlations often exceeding 0.85) and accessibility via commercial microarrays from platforms like Agilent, ChIP-on-chip has notable limitations, including probe-specific biases that limit genomic coverage to about 70%, broader peak resolutions (typically 300–500 bp), and higher susceptibility to artifacts from cross-link reversion or non-specific binding. These issues can lead to false positives, particularly in regions with repetitive sequences, necessitating careful controls like input DNA normalization and RNase treatment. In the current era, ChIP-on-chip has been largely superseded by ChIP-seq (chromatin immunoprecipitation followed by high-throughput sequencing), which offers superior resolution (down to single-base pair), fuller genome coverage, and reduced biases due to unbiased sequencing of all fragments, especially as sequencing costs have declined since the mid-2000s. Nonetheless, ChIP-on-chip remains valuable in resource-limited settings or for validating sequencing data, and its foundational principles continue to inform modern research.

Background and Principles

Definition and Objectives

ChIP-on-chip, also known as ChIP-chip, is a high-throughput genomic technique that integrates chromatin immunoprecipitation (ChIP) with DNA microarray hybridization to identify and map the binding sites of DNA-associated proteins across the genome. In this method, proteins such as transcription factors or histone modifications are cross-linked to DNA in vivo, the chromatin is fragmented, and specific protein-DNA complexes are enriched using antibodies; the resulting DNA fragments are then amplified, labeled, and hybridized to microarrays containing genomic probes to detect enriched regions indicative of binding sites. The primary objectives of ChIP-on-chip are to enable genome-wide profiling of protein-DNA interactions, thereby facilitating the study of , epigenetic modifications, and architecture in a systematic manner. By revealing where regulatory proteins bind, the technique helps elucidate mechanisms of control, identify targets of transcription factors involved in cellular processes like and , and map modifications that influence accessibility and epigenetic states. For instance, it has been used to uncover how oncogenes like c-Myc regulate gene networks in human cells, providing insights into cancer biology. Originally developed for the eukaryotic , ChIP-on-chip was first applied to map the binding locations of yeast transcription factors such as Gal4 and Ste12 across all intergenic regions, demonstrating its utility in simple genomes. The approach has since been extended to more complex mammalian systems, including human and mouse cells, where it has profiled binding of factors like and marks in cultured cell lines. Early implementations were constrained to predefined genomic regions, such as promoter arrays, limiting coverage to known elements; however, advancements with high-density tiled arrays have expanded its scope to near-complete genome-wide analysis with resolutions typically ranging from 300–500 base pairs, limited by fragment size and array design, enhancing its applicability for comprehensive eukaryotic studies.

Core Components: ChIP and Microarray Integration

ChIP, or chromatin immunoprecipitation, begins with the covalent crosslinking of proteins to DNA in living cells, typically using formaldehyde, which forms reversible bonds between nearby amino groups and creates short-range links (approximately 2 Å) to preserve in vivo interactions. Following crosslinking, cells are lysed using a detergent-based buffer, such as SDS, to release chromatin while maintaining the crosslinks. The chromatin is then sheared into small fragments, usually 200-1000 base pairs in length, through mechanical methods like sonication, which disrupts DNA while keeping protein-DNA complexes intact; sonication parameters, including probe depth and duration, are optimized to achieve this fragment size for efficient immunoprecipitation. Immunoprecipitation follows, where specific antibodies against the target protein capture the associated DNA fragments, often bound to protein A/G agarose or magnetic beads during overnight incubation at 4°C; antibody specificity is critical, requiring validation on crosslinked chromatin to ensure recognition of the native epitope without cross-reactivity, as non-specific binding can lead to artifacts in downstream analyses. Crosslinks are subsequently reversed by heating (e.g., at 65-68°C for 4-6 hours) in the presence of high salt, EDTA, and proteinase K to digest proteins, followed by DNA purification via phenol-chloroform extraction and ethanol precipitation to yield enriched DNA fragments. Microarray technology in this context relies on high-density arrays of immobilized DNA probes, typically synthetic oligonucleotides representing genomic regions, spotted or synthesized on a solid substrate like a glass slide. Target DNA is fluorescently labeled—commonly with cyanine dyes such as Cy3 (green) or Cy5 (red)—after denaturation and fragmentation, allowing it to hybridize to complementary probes under controlled conditions (e.g., 60-65°C in a hybridization buffer). Non-hybridized DNA is washed away, and the array is scanned with a laser to detect fluorescence intensity at each probe spot, where signal strength correlates with the amount of bound target DNA; in ChIP-on-chip, genome-tiling microarrays are used, featuring overlapping probes spaced every 200-500 bp to provide comprehensive coverage of intergenic and promoter regions. The integration of ChIP and microarray in ChIP-on-chip occurs post-purification, where the enriched ChIP DNA is amplified (e.g., via ligation-mediated PCR) and differentially labeled with Cy5, while a reference sample—such as total input DNA or mock-immunoprecipitated DNA—is labeled with Cy3 to serve as a control for background and normalization. These labeled samples are combined and hybridized to the microarray, enabling competitive binding where enrichment appears as a higher Cy5/Cy3 ratio at protein-bound loci; this ratio-based detection identifies genome-wide binding sites by comparing signal intensities, with mock or input controls accounting for non-specific hybridization or copy number variations. This hybrid approach leverages ChIP's specificity for protein-DNA capture with microarray's high-throughput readout, assuming prior knowledge of DNA-protein interaction dynamics but emphasizing validated antibodies to minimize off-target effects.

Experimental Workflow

Technological Platforms

ChIP-on-chip experiments rely on platforms designed to hybridize enriched fragments, with array types evolving from targeted to comprehensive coverage. Promoter arrays focus on regulatory regions, typically interrogating 1-2 kb upstream of annotated genes to detect binding and other protein-DNA interactions in promoter elements. In contrast, tiling arrays provide whole-genome coverage by incorporating overlapping probes spaced 200-500 apart, enabling detection of binding events across intergenic and intronic regions without prior assumptions about functional elements. These tiling designs accommodate the typical 200-500 size of ChIP-generated fragments, resulting in an effective of approximately 200 , limited by fragment length rather than probe spacing alone. Major commercial platforms dominate ChIP-on-chip applications, including those from , Agilent Technologies, and NimbleGen (acquired by in 2007). Affymetrix arrays utilize short 25-mer probes at high density, with up to 6 million probes per array spaced every 35 , offering superior for detailed genomic . Agilent and NimbleGen platforms employ longer 60-mer probes, with NimbleGen providing customizable high-density options that support up to 2.1 million features per for flexible species-specific designs. Both custom arrays, fabricated for unique genomic targets, and commercial off-the-shelf arrays are available, though commercial options from these vendors are preferred for their reproducibility and validated performance in genome-wide assays. These platforms typically incorporate two-channel detection, labeling immunoprecipitated DNA with one (e.g., Cy5) and total input DNA with another (e.g., Cy3), borrowing principles from to normalize signals and enhance signal-to-noise ratios. Advancements in the mid-2000s shifted ChIP-on-chip toward higher-density arrays, enabling practical whole-genome tiling for complex mammalian genomes like and , which previously required multiple lower-density arrays. This evolution, driven by improved photolithographic and fabrication techniques, increased probe counts and reduced spacing, allowing studies of large eukaryotic genomes with resolutions approaching single kilobase pairs. However, array designs are susceptible to biases, such as variations that influence hybridization stability and probe affinity, potentially skewing enrichment signals in AT- or GC-rich regions. While costs have declined with these improvements, early genome-wide experiments historically required substantial investment due to the need for multiple arrays and specialized scanning equipment.

Wet-Lab Procedures

The wet-lab procedures for ChIP-on-chip begin with to capture protein-DNA interactions . Cells or tissues are first fixed using 1% for 10-15 minutes at to proteins to DNA, preserving their associations; this concentration and duration are optimized for mammalian cells to balance fixation efficiency and accessibility. The reaction is quenched with to stop , followed by cell lysis in a containing detergents like and , often with protease inhibitors to prevent degradation. is then isolated by and fragmented, typically via , to generate DNA fragments of 200-500 bp, which is suitable for high-resolution mapping on microarrays; enzymatic digestion with micrococcal nuclease serves as an alternative for gentler shearing in sensitive samples. Immunoprecipitation follows to enrich for target protein-bound DNA. The sheared chromatin is incubated overnight at 4°C with a specific , such as anti-H3K9me2 for histone modifications, at concentrations of 1-5 µg per immunoprecipitation; antibody validation via is essential to confirm specificity and avoid non-specific binding. - or G-conjugated beads are added to capture the antibody-chromatin complexes, with incubation for 1-2 hours, followed by extensive washing to remove unbound material. Controls include input DNA (unimmunoprecipitated chromatin) and non-specific IgG to assess background enrichment. The complexes are then eluted in a high-pH or SDS-containing buffer and decrosslinked by heating at 65°C in the presence of NaCl, often overnight, to reverse links. DNA recovery involves treating the decrosslinked samples with RNase A and Proteinase K to digest RNA and proteins, respectively, yielding purified DNA. This is followed by extraction using phenol-chloroform or spin-column purification kits to isolate the DNA, with yields typically in the nanogram range suitable for downstream amplification and labeling prior to hybridization. Quantification is performed using , such as NanoDrop, measuring at 260 nm to ensure sufficient material (e.g., 5-50 ng/µL). Quality controls are critical throughout to ensure protocol success. Fragment size is verified by electrophoresis on a 1% agarose gel or Bioanalyzer, confirming a smear centered at 200-500 bp; deviations indicate issues like incomplete shearing. Antibody efficiency is checked pre-IP via or , while post-IP enrichment is preliminarily assessed by qPCR at known target loci. Common pitfalls include over-fixation, which hinders fragmentation and reduces recovery (mitigated by precise timing), or inconsistent due to overheating, addressed by using cooled baths or optimized cycles. The entire wet-lab process, from fixation to DNA recovery, typically spans 2-3 days: day 1 for fixation, lysis, and initial fragmentation; day 2 for and washes; and day 3 for , decrosslinking, and purification.

Dry-Lab Procedures

Following the purification of ChIP-enriched and input DNA from the wet-lab procedures, the dry-lab phase begins with amplification to generate sufficient material for analysis. Typically, 1-10 ng of purified DNA serves as starting material, which is amplified using ligation-mediated (LM-PCR) or random priming methods to yield microgram quantities while preserving representation across the . In LM-PCR, blunt-ended DNA fragments are ligated to synthetic linkers, followed by PCR amplification with linker-specific primers for 35-45 cycles to produce amplicons of 200-1,000 ; this approach, introduced in early ChIP-on-chip studies, ensures broad coverage but can introduce minor biases if cycle numbers exceed optimal ranges. Random priming, an alternative enzymatic method, uses Klenow with random hexamer primers to linearly amplify DNA, often preferred for its reduced bias in later protocols. Amplified yields are quantified, aiming for >500 ng to support downstream labeling and multiple replicates. Labeling incorporates fluorescent dyes to enable differential detection of enriched versus control DNA on the array. Dual-color labeling is standard, with Cy3 (green fluorescence) typically assigned to input/control DNA and Cy5 (red fluorescence) to immunoprecipitated (IP) DNA, or vice versa in dye-swap replicates. The process involves denaturing 500-1,000 ng of amplified DNA at 95°C, followed by incubation with dye-conjugated random primers or dNTPs and (e.g., exo-Klenow) at 37°C for 2 hours, yielding labeled fragments of 100-500 suitable for hybridization. Specific activity is assessed post-labeling, targeting >15 pmol/μg for Cy3 and >18 pmol/μg for Cy5 to ensure robust signal detection; yields of >3 μg labeled DNA per sample are ideal for array application. This step integrates the amplified products directly into the detection workflow. Hybridization applies the labeled samples to the microarray slide for binding to immobilized probes. The Cy3- and Cy5-labeled DNAs (typically 5-15 μg total per array) are combined with blocking agents like Cot-1 DNA (1-5 μg) and hybridization buffer, denatured at 95°C, then applied to the array under a coverslip. occurs in a humidified chamber at 42-65°C for 16-40 hours with rotation (10-20 rpm) to promote even contact; temperatures vary by array type, with higher settings (e.g., 65°C) used for arrays to enhance specificity. Post-hybridization, slides are washed sequentially in low-stringency buffer (e.g., 0.6× with 0.005% at room temperature for 5-10 min) followed by high-stringency buffer (e.g., 0.06× at 37°C for 5 min) to remove unbound and non-specifically bound fragments, minimizing background. Ozone levels are monitored, as they can degrade Cy5; protective measures like acetonitrile washes are applied if >5 ppb. Scanning captures the hybridized signals using a dedicated microarray scanner. Laser excitation at 532 nm (Cy3) and 635 nm (Cy5) induces fluorescence, detected by photomultiplier tubes (PMTs) at resolutions of 5-10 μm; settings include 100% PMT gain and 16-bit depth for dynamic range. Raw output consists of TIFF images representing pixel intensities for each spot, from which software extracts median foreground and background signals per probe, computing log2 ratios (Cy5/Cy3) as initial enrichment metrics. Signal uniformity is visualized in pseudo-color overlays, with saturated or absent spots flagged. Typical scan times are 10-30 minutes per slide, producing files of 100-500 MB. Quality metrics ensure reliable data acquisition throughout these steps. Pre-hybridization, DNA yield (>500 ng amplified) and labeling efficiency (e.g., A260/A555 >1.0 for Cy3, A260/A650 >1.0 for Cy5) are verified via NanoDrop to confirm sufficient material and minimal degradation. Post-scan, assessments include signal-to-noise ratios (>5:1), spot uniformity (CV <20% across array), and background noise levels (<10% of foreground); grids are aligned with >95% success in feature extraction software. Low yields or uneven signals prompt repetition of or labeling. These checks establish baseline before advanced processing. Dye bias, where one fluorophore yields systematically higher or lower signals due to sequence-specific effects, is a common artifact addressed through experimental design. Performing dye-swap replicates—switching Cy3 and Cy5 assignments between IP and input in parallel arrays—allows averaging of ratios to correct bias, improving reproducibility by 20-30%. Without swaps, normalization alone may insufficiently mitigate this, leading to false positives in enrichment calls. This practice is essential for quantitative accuracy in dual-color setups.

Data Analysis and Interpretation

Computational Tools and Software

The analysis of ChIP-on-chip data begins with processing raw scanning outputs from images, where tools like GenePix Pro software quantify spot intensities by extracting fluorescence signals from scanned files generated by array scanners. This step is followed by background subtraction to remove non-specific signals and loess normalization to correct for dye bias in two-color arrays, often implemented via / packages such as limma, which applies locally weighted scatterplot smoothing to balance Cy3 and Cy5 channel intensities across probes. For peak detection and initial processing, several specialized open-source tools have been developed to handle tiling array data. The Model-based Analysis of Tiling-arrays () algorithm processes Affymetrix .CEL files by modeling probe-specific biases and identifying enriched regions using a sliding-window approach with trimmed means, enabling reliable detection even from single-color arrays without replicates. Similarly, TileMap employs a hierarchical empirical Bayes framework to model spatial correlations and errors in tiling arrays, supporting both and NimbleGen formats for identifying binding sites or modifications. Additional open-source options include MA2C (Model-based Analysis of 2-Color arrays), which normalizes NimbleGen or Agilent two-color data by accounting for and probe effects before scoring enrichments, and cisGenome, an integrated system that supports visualization, , and peak calling for both ChIP-on-chip and ChIP-seq data across multiple platforms. Commercial software like ArrayStar from DNASTAR provides a graphical interface for importing data, performing , and visualizing enrichments, with support for legacy ChIP-on-chip formats alongside modern sequencing workflows. Custom pipelines are often built using scripting languages such as Perl or Python to integrate these tools, for instance, parsing NimbleGen .pair files or Affymetrix outputs for automated batch processing. As of 2025, while ChIP-on-chip tools remain available, their use has declined in favor of ChIP-seq, with legacy support integrated into platforms like Galaxy workflows for historical data reanalysis.

Statistical Methods and Validation

Normalization of ChIP-on-chip data typically involves quantile or median normalization across multiple arrays to account for technical variations in hybridization and scanning, followed by the computation of log2(IP/input) ratios to generate enrichment scores for each probe. These ratios quantify the relative abundance of immunoprecipitated DNA compared to input DNA, highlighting regions of protein-DNA interaction while mitigating biases from global copy number or GC content. This approach, introduced in early ChIP-on-chip studies, enables the identification of enriched genomic loci by transforming raw fluorescence intensities into a comparable scale. Peak calling in ChIP-on-chip analysis often employs sliding window methods, such as evaluating 500 bp windows across the genome to detect contiguous regions of elevated enrichment scores exceeding predefined thresholds, typically corresponding to p-values less than 0.001. To address the challenge of multiple testing inherent in genome-wide scans, error models incorporate false discovery rate (FDR) corrections, commonly using the Benjamini-Hochberg procedure, which adjusts p-values to control the expected proportion of false positives among significant peaks. The enrichment score is described as log2(observed/expected), where the expected signal derives from control experiments like input DNA, providing a measure of deviation from background levels. Validation of identified peaks relies on quantitative PCR (qPCR) confirmation, where primers targeting candidate regions amplify DNA from both IP and input samples to verify enrichment independently of microarray artifacts. Additional rigor comes from comparing results across biological replicates, assessing reproducibility through metrics such as overlap rates or correlation coefficients, and evaluating overall performance via false discovery rate estimates alongside sensitivity and specificity measures. These steps ensure that statistically significant peaks correspond to true biological signals rather than noise. Key challenges in ChIP-on-chip statistical analysis include handling spatial in data, where signals from adjacent probes are correlated due to structure and probe proximity, potentially inflating significance estimates. Bayesian models address this by incorporating probe-specific variances and dependencies, such as through hierarchical frameworks that model latent binding states while accounting for neighboring probe correlations to improve accuracy in detection. These advanced approaches enhance robustness against uneven probe coverage and experimental variability.

Advantages and Limitations

Strengths

ChIP-on-chip enables high-throughput analysis by simultaneously interrogating thousands of genomic loci across the , facilitating the discovery of novel protein-DNA binding sites in regulatory regions such as promoters and enhancers. This genome-wide approach was a significant advancement over earlier low-throughput methods, allowing researchers to map binding events for transcription factors and histone modifications on a large scale without prior knowledge of target sites. The technology is cost-effective, particularly prior to the widespread adoption of next-generation sequencing around 2007, as it required less expensive microarray hybridization compared to early sequencing alternatives, making it accessible to laboratories without specialized sequencing equipment. Commercial tiled arrays could be designed for reuse across multiple experiments, further reducing overall costs while providing scalable profiling for various organisms. Tiled arrays in ChIP-on-chip achieve an effective resolution of typically 300-500 bp, determined by the size of sonicated chromatin fragments and probe spacing, which is sufficient for precise mapping of binding sites in promoter and enhancer regions. This resolution supports detailed insights into regulatory elements without the need for single-nucleotide precision in many applications. Reproducibility is enhanced by standardized commercial arrays from platforms like Affymetrix, NimbleGen, and Agilent, which minimize technical variability through consistent probe design and manufacturing. The dual-color hybridization format, where immunoprecipitated DNA and input control are labeled with different fluorescent dyes (e.g., Cy5 and Cy3) and co-hybridized to the same array, enables direct sample comparison, reducing dye bias and improving signal reliability across replicates. ChIP-on-chip offers high accessibility, as it relies on established infrastructure and does not require expertise in high-throughput sequencing or complex bioinformatics pipelines for data generation. The overall , from preparation to array scanning, typically yields results within 1-2 weeks, allowing rapid iteration in experimental design for studies of protein-DNA interactions.

Weaknesses

ChIP-on-chip assays are constrained by the resolution of DNA fragmentation and microarray probe spacing, typically involving sheared DNA fragments of 200–1000 base pairs (), which limits the detection of fine-scale binding motifs or low-affinity protein-DNA interactions below this scale. Probe densities on arrays, often spaced at 200–500 bp intervals for promoter-focused designs, further reduce precision, potentially overlooking subtle enrichments in intergenic or distal regulatory elements. Several sources of bias and artifacts compromise the reliability of ChIP-on-chip data. Antibody cross-reactivity can lead to non-specific , capturing unintended genomic regions and introducing false enrichments, a challenge inherent to protocols. During sample preparation, amplification for labeling introduces biases favoring certain sequences, while microarray hybridization exhibits inefficiencies such as AT/GC content bias, where GC-rich probes hybridize more efficiently, skewing signal intensities and exacerbating noise. These issues contribute to higher false positive rates in peak calls. Coverage in ChIP-on-chip is inherently limited to predefined array regions, often focusing on promoters or non-repetitive sequences, which excludes repetitive or low-complexity DNA comprising up to 50% of mammalian genomes and misses potential binding sites in these areas. Custom whole-genome tiling arrays, necessary for broader interrogation, incur high costs due to the need for multiple high-density slides and specialized manufacturing. Additionally, the technique's data are noisy, necessitating 3–5 biological replicates per condition to achieve reproducibility, and it struggles to differentiate direct from indirect binding events without orthogonal validation. As of 2025, ChIP-on-chip has become largely obsolete for most primary research, supplanted by sequencing-based methods like ChIP-seq, which offer superior resolution, unbiased genome-wide coverage, and lower error rates. Nonetheless, it remains valuable in resource-limited settings or for validating sequencing data.

Historical Development

Origins and Early Experiments

The origins of ChIP-on-chip trace back to the late , when researchers sought to extend the () technique—originally developed in the 1980s for studying protein-DNA interactions —to enable higher-throughput analysis of genomic binding sites. The first published ChIP-on-chip experiment appeared in 1999, conducted by Megee et al., who mapped the distribution of the complex along the entire length of budding yeast III. This proof-of-concept study used crosslinking followed by of cohesin subunits, with the enriched DNA fragments hybridized to a custom-spotted of 133 PCR-amplified segments spanning the ~315 kb chromosome, revealing preferential binding sites in intergenic regions and differential regulation near centromeres. Although not fully genome-wide, this work demonstrated the potential of combining ChIP with microarray hybridization to visualize protein occupancy at multiple loci, overcoming the limitations of low-throughput PCR-based validation. Building on this foundation, the technique was rapidly adapted for genome-scale studies of transcription factors by key pioneers in the laboratories of (then in Richard Young's group at the Whitehead Institute), (Harvard Medical School), and Richard Young. Motivated by the need to dissect gene regulatory networks amid the yeast genome sequencing completion in 1996 and the rise of technology in the mid-1990s, et al. published the seminal 2000 study applying ChIP-on-chip to identify binding sites for the transcription activators Gal4 and Ste12 across the genome. In this experiment, cells were crosslinked, chromatin sheared, and protein-DNA complexes immunoprecipitated using epitope-tagged factors; the recovered DNA was labeled and co-hybridized with input genomic DNA to custom arrays covering ~6,400 intergenic regions, identifying 10 direct Gal4 targets involved in galactose metabolism and 29 Ste12 sites linked to mating response pathways. This integration of ChIP's specificity with microarrays' parallel detection marked a pivotal advance in . Early ChIP-on-chip implementations were constrained by the technology's nascent state, primarily focusing on promoter regions due to the availability of intergenic-focused arrays and the challenges of whole-genome . These studies employed spotted cDNA or microarrays with resolutions limited to approximately 1 , as probe spacing and fragment sizes restricted precise peak localization, and cross-hybridization or low signal-to-noise ratios could confound results in repetitive sequences. Despite these hurdles, the initial demonstrations established ChIP-on-chip as a feasible for genome-wide mapping of occupancy, revealing direct regulatory targets and coordinated modules that were previously inaccessible, thereby igniting widespread interest in systematic analyses of chromatin-associated proteins.

Key Milestones and Adoption

In 2004, significant advancements in ChIP-on-chip application were achieved through large-scale mapping efforts. The laboratory of Richard Young at the Whitehead Institute mapped the sites of 203 DNA- proteins (primarily transcription factors) in using epitope-tagged proteins and intergenic hybridization, revealing extensive regulatory networks and combinatorial patterns across the . Concurrently, Bing Ren and colleagues in Richard Young's laboratory extended the technique to mammalian systems by identifying transcription factor sites -wide in human cells, demonstrating the method's utility for promoter and uncovering novel targets beyond consensus sites. Between 2003 and 2005, ChIP-on-chip was adapted for studying modifications and marks, broadening its scope to . A key example was the genome-wide profiling of H3K9 in , which showed enrichment at active promoters and its correlation with transcription levels, establishing ChIP-on-chip as a tool for mapping epigenetic landscapes. During this period, commercial high-density oligonucleotide arrays became available, with companies like Agilent and NimbleGen offering whole-genome tiling arrays for the , facilitating broader accessibility and higher resolution for mammalian studies. From 2006 to 2010, the technique evolved with the widespread use of whole-genome tiling arrays, enabling comprehensive coverage including intragenic regions. This advancement supported integration into large-scale initiatives, such as contributions to the project, where ChIP-on-chip data mapped occupancy and modifications across multiple human cell types, informing regulatory element annotations. Adoption of ChIP-on-chip peaked in the mid-2000s, particularly in and mammalian model systems, due to its role in pioneering genome-wide protein-DNA interaction studies. Usage declined after 2007 with the emergence of ChIP-seq, which offered higher resolution and lower costs, though ChIP-on-chip persisted in resource-limited settings through the for its simplicity and lack of sequencing requirements. As of 2025, ChIP-on-chip maintains niche applications, primarily for targeted custom arrays in validation experiments or low-throughput analyses, with approximately 5,000 total publications compared to over 50,000 for ChIP-seq, reflecting its displacement by sequencing-based s.

Alternatives and Comparisons

ChIP-seq

ChIP-seq, or followed by next-generation sequencing (NGS), is a for mapping protein-DNA interactions across the entire genome in an unbiased manner by sequencing the DNA fragments enriched through . This approach enables the identification of binding sites for transcription factors, modifications, and other chromatin-associated proteins at a genome-wide scale without reliance on predefined array elements. The technique was first introduced in 2007, with Johnson et al. demonstrating its application in using the Illumina platform to generate millions of short reads (approximately 25–50 at the time) for mapping protein-DNA interactions. Concurrently, Mikkelsen et al. applied ChIP-seq to mammalian cells, profiling modifications in embryonic stem cells and lineage-committed cells via high-throughput sequencing on the Illumina Genome Analyzer, yielding reads of 30–50 . Early implementations utilized platforms like Illumina and , which produce short reads (typically 50–100 in modern iterations) to achieve deep coverage, often sequencing tens to hundreds of millions of reads per sample. ChIP-seq shares the initial chromatin immunoprecipitation steps with array-based methods but diverges by replacing microarray hybridization with DNA library preparation— involving end repair, adapter ligation, and amplification—followed by NGS to directly sequence the enriched fragments. Key advantages include single-base-pair resolution, comprehensive coverage of the entire without probe biases, and reduced compared to earlier techniques limited by array tiling intervals of 200–500 bp. However, it demands substantial computational resources for read alignment, peak calling, and bias correction, alongside higher per-sample costs of approximately $500–$1,000 in 2025, encompassing library preparation and sequencing. Since around 2010, ChIP-seq has emerged as the dominant standard for genome-wide studies, supplanting array-based approaches in the vast majority of new experiments due to its superior precision and scalability. DamID (DNA adenine methyltransferase identification) is an antibody-independent method that employs fusion proteins of DNA adenine methyltransferase () with a protein of interest to label DNA binding sites through site-specific , enabling detection without . Developed in 2000, DamID is particularly advantageous for live-cell studies as it avoids fixation and crosslinking artifacts associated with traditional protocols, allowing real-time profiling of dynamic protein-DNA interactions. The technique achieves a resolution of approximately 100 base pairs by analyzing patterns via digestion or sequencing, making it suitable for mapping interactions in organisms like and mammalian cells. ChIP-exo represents an enhanced variant of chromatin immunoprecipitation that incorporates lambda exonuclease digestion after antibody pull-down to trim DNA fragments precisely to the protein-binding site, yielding nucleotide-level resolution. Introduced in 2011, this method significantly reduces background noise from non-specific DNA fragments compared to standard ChIP approaches, as the exonuclease selectively degrades unprotected DNA ends, resulting in sharper peaks and higher specificity for transcription factor binding sites. ChIP-exo has been applied to map interactions in yeast and mammalian systems, providing near-single-nucleotide precision that surpasses the array-based limitations of ChIP-on-chip. CUT&RUN (Cleavage Under Targets and Release Using Nuclease) is a targeted enzymatic cleavage technique that tethers micrococcal to antibodies bound to , releasing protein-DNA fragments directly from intact nuclei without extensive or washes. First described in 2017, it requires far fewer cells—typically hundreds to thousands versus millions for conventional —while being faster (1-2 days) and more antibody-efficient due to minimized loss during purification steps. This approach excels in low-input scenarios, such as primary tissues or rare cell types, and generates high-resolution profiles with low background by cleaving specifically at binding sites. These techniques collectively address key limitations of ChIP-on-chip, including moderate (tens to hundreds of base pairs limited by probes), potential biases from hybridization, and reliance on antibodies that can introduce . DamID, ChIP-exo, and offer improved spatial precision and reduced artifacts—DamID notably bypasses fixation-induced distortions—though they often necessitate specialized enzymes or sequencing adaptations that may require additional equipment beyond standard setups. For instance, ChIP-exo and enhance signal-to-noise ratios over ChIP-on-chip's indirect detection, enabling more accurate motif identification and co-occupancy analysis. By 2025, DamID, ChIP-exo, and CUT&RUN have gained significant traction for single-cell and low-input applications, driven by adaptations like targeted DamID for histone marks and optimized CUT&RUN protocols for rare populations, complementing sequencing-based methods in resource-limited settings.

Applications

In Transcription Factor Binding Studies

ChIP-on-chip has been instrumental in mapping the occupancy of transcription factors (TFs) across genomes, particularly in promoters and enhancers, enabling the identification of regulatory elements that control . Early applications focused on model organisms like , where a comprehensive atlas of TF binding was generated by analyzing 203 TFs using ChIP-on-chip, revealing that many TFs bind to hundreds of sites under standard growth conditions and highlighting the prevalence of combinatorial binding patterns. In human cells, similar studies mapped binding sites for key TFs such as , identifying over 1,800 high-confidence sites enriched in response elements associated with arrest and , and NF-κB, which showed widespread distribution across with clusters near immune response genes. These mappings demonstrated that TFs like preferentially occupy enhancers in addition to promoters, providing insights into how sequence-specific interactions drive targeted gene regulation. Beyond static mapping, ChIP-on-chip has uncovered condition-specific TF binding dynamics, such as during stress responses, where factors like exhibit altered occupancy at sites linked to DNA damage repair genes upon exposure. Integration of ChIP-on-chip with profiles has facilitated network inference, allowing researchers to distinguish direct targets from indirect effects; for instance, combining for multiple TFs with mRNA levels in revealed regulatory modules where co- TFs synergistically activate stress-responsive pathways. In studies, ChIP-on-chip for family members (E2F1, E2F4, E2F6) in normal and tumor cells identified overlapping sites near genes, enabling the construction of networks that predict proliferation states based on differential expression correlations. The project's pilot phase in 2007 exemplified large-scale application, using ChIP-on-chip to profile binding for approximately 20 human TFs across 1% of the genome, which identified motifs for factors like and revealed that ~30% of binding sites occur in non-promoter regions, informing models of distal regulation. Post-peak calling from these datasets, motif discovery tools applied to enriched regions have routinely uncovered consensus sequences, such as the (PuPuPuC(A/T)(T/A)GPyPyPy), validating direct binding and facilitating annotation of novel sites. This approach advanced understanding of combinatorial regulation, as seen in where ~40% of genes are co-regulated by multiple TFs binding nearby, forming logic gates for precise transcriptional control. Despite its contributions, ChIP-on-chip faces challenges in distinguishing direct from indirect binding, as occupancy can reflect protein-protein tethering rather than DNA contact, necessitating orthogonal validation like motif enrichment or reporter assays. Nonetheless, due to lower costs compared to sequencing-based alternatives, ChIP-on-chip remains relevant in 2025 for targeted panels of TFs in resource-limited settings, such as custom arrays for disease-specific regulators in clinical research.

In Epigenetic Research

ChIP-on-chip has been instrumental in genome-wide mapping of histone modifications, enabling the identification of epigenetic marks associated with gene regulation. For instance, it has been used to profile trimethylation of at lysine 4 (), which marks active promoters, and trimethylation at lysine 27 (), indicative of transcriptional repression. In early applications, this technique revealed distinct enrichment patterns across large genomic regions, providing insights into how these modifications organize states. Pioneering studies in embryonic stem cells demonstrated ChIP-on-chip's utility in epigenetic research, such as a 2006 analysis that mapped and across ~30 Mb of the mouse genome, identifying bivalent domains—regions simultaneously marked by both modifications—at promoters of developmental genes. These domains maintain genes in a poised, low-expression state during pluripotency, resolving into active or repressive monovalent marks upon , thus highlighting ChIP-on-chip's role in elucidating epigenetic mechanisms of . Epigenetic insights from such mappings correlate modifications with patterns; for example, enrichment aligns with transcribed loci, while correlates with silencing. In disease models, ChIP-on-chip has profiled altered landscapes in cancer, such as in gastric tumors where H3K9ac, , and distributions identified novel regulatory targets linked to tumorigenesis. Large-scale epigenomic efforts in the , building on ChIP-on-chip methodologies, generated maps across numerous cell types to catalog modification landscapes, including the identification of bivalent domains in developmental contexts. Outputs from these analyses are often visualized as enrichment profiles in heatmaps, which display modification intensities across genomic features like promoters and enhancers, facilitating the correlation of epigenetic states with regulatory elements such as loci. ChIP-on-chip has linked marks to regulation, as seen in studies where modification changes at lncRNA promoters influence their expression and downstream epigenetic control. As of 2025, ChIP-on-chip remains valuable for hypothesis-driven studies using targeted arrays, which focus on specific genomic regions to assess modifications cost-effectively. It complements techniques like , which profiles open , by providing targeted epigenetic mark data to interpret accessibility in terms of specific regulatory states.

References

  1. [1]
    Genome-Wide Location and Function of DNA Binding Proteins
    Bing Ren et al. ,. Genome-Wide Location and Function of DNA Binding Proteins.Science290,2306-2309(2000).DOI:10.1126/science.290.5500.2306. Export citation.Missing: Ren 2000
  2. [2]
    ChIP-chip versus ChIP-seq: Lessons for experimental design and ...
    Feb 28, 2011 · ChIP-based histone modification data is commonly used to reconstruct average signal profiles, or "epigenetic signatures," of key genomic regions ...Missing: review | Show results with:review
  3. [3]
    Chip on chips | Nature Reviews Genetics
    Feb 1, 2001 · A microarray-based technique that can monitor the chromosomal locations at which protein–DNA interactions occur across the entire yeast genome.
  4. [4]
    ChIP on Chip: surprising results are often artifacts - PMC
    The method of chromatin immunoprecipitation combined with microarrays (ChIP-Chip) is a powerful tool for genome-wide analysis of protein binding.
  5. [5]
    Genome-wide location and function of DNA binding proteins - PubMed
    Genome-wide location and function of DNA binding proteins. Science. 2000 Dec 22;290(5500):2306-9. doi: 10.1126/science.290.5500.2306. Authors. B Ren , F ...
  6. [6]
    ChIP-chip: considerations for the design, analysis, and application of ...
    ChIP-chip: considerations for the design, analysis, and application of genome-wide chromatin immunoprecipitation experiments. Genomics. 2004 Mar;83(3):349-60 ...
  7. [7]
    ChIP-on-chip significance analysis reveals large-scale binding ... - NIH
    Jan 6, 2009 · ChIP-on-chip has emerged as a powerful tool to dissect the complex network of regulatory interactions between transcription factors and their ...
  8. [8]
    Chromatin Immunoprecipitation Assay - Taylor & Francis Online
    Jun 6, 2018 · This review addresses the critical parameters and the variants of ChIP as well as the different analytical tools that can be combined with ChIP ...
  9. [9]
    A ChIP on the shoulder? Chromatin immunoprecipitation and ...
    Jul 13, 2015 · In this review, we will highlight some of the concerns and challenges that arise in selecting and validating antibodies for ChIP.Chip Method Overview · Endogenous Protein Vs Tagged... · Standard Assays For Chip...<|control11|><|separator|>
  10. [10]
    DNA Microarray Technology Fact Sheet
    Aug 15, 2020 · The DNA microarray is a tool used to determine whether the DNA from a particular individual contains a mutation in genes like BRCA1 and BRCA2.
  11. [11]
    Tiling Array - an overview | ScienceDirect Topics
    This type of tiling array is commercially available through NimbleGen and Affymetrix, but may not be available for many model organisms. Since short probes ...<|separator|>
  12. [12]
    Tiling the Genome - Drug Discovery and Development
    Jun 5, 2008 · For the specific promoter tiling array, he “tiles 4-kb upstream and 4-kb downstream of almost all known human genes.” ChIP-chip method protocol
  13. [13]
    Systematic evaluation of variability in ChIP-chip experiments using ...
    The groups labeled and hybridized the mixtures to one of three different types of tiling arrays (NimbleGen, Affymetrix, or Agilent). Each of the tiling array ...
  14. [14]
    Getting Started in Tiling Microarray Analysis - Research journals
    Oct 26, 2007 · Affymetrix tiles 6 million 25-mer probes per array, which offers the lowest price per probe and the highest resolution (chromosomal distance ...
  15. [15]
    Starr: Simple Tiling ARRay analysis of Affymetrix ChIP-chip data
    Apr 17, 2010 · Data import from the microarray manufacturers Nimblegen and Agilent has already been implemented in Ringo, the Affymetrix array platform is ...Missing: types | Show results with:types
  16. [16]
    ChIP-Seq: advantages and challenges of a maturing technology - NIH
    Oct 12, 2011 · ChIP-Seq is a technique for genome-wide profiling of DNA-binding proteins, histone modifications, or nucleosomes.<|separator|>
  17. [17]
    Model-based analysis of tiling-arrays for ChIP-chip - PNAS
    Aug 15, 2006 · We propose a fast and powerful algorithm, MAT, for data analysis of ChIP-chip on Affymetrix tiling arrays. By using a simple linear model, MAT ...
  18. [18]
    Complete Guide to Sonication of Chromatin for ChIP Assays
    Jan 31, 2020 · Many standard N-ChIP protocols include a micrococcal nuclease (MNase) treatment to fragment the chromatin right after cells/tissue collection or ...
  19. [19]
    Simple ChIP Assay Protocol (Agarose Beads)
    ### Summary of ChIP Immunoprecipitation Steps
  20. [20]
    Studying epigenetics using ChIP - Abcam
    The ChIP procedure utilizes an antibody to immunoprecipitate a protein of interest, such as a transcription factor, along with its associated DNA. The ...
  21. [21]
    COMPARISON OF SAMPLE PREPARATION METHODS FOR CHIP ...
    A commonly used technique for amplifying the DNA obtained from ChIP assays is linker-mediated PCR (LMPCR). However, using this amplification method, we could ...Missing: LM- | Show results with:LM-
  22. [22]
    [PDF] Agilent ChIP-on-chip Analysis
    This chapter describes the steps to hybridize, wash and scan Agilent ChIP-on-chip microarrays and to extract data using the Agilent Feature Extraction Software ...
  23. [23]
    Adaptable gene-specific dye bias correction for two-channel DNA ...
    Apr 28, 2009 · For two-channel microarray experiments, gene-specific dye bias (GSDB) is easily observed in dye-swap replicates. It results in probes being ...
  24. [24]
    Characterizing dye bias in microarray experiments - Oxford Academic
    Dye bias that is not removed by array normalization can introduce bias into comparisons between samples of interest. But if the bias is consistent across ...Missing: ChIP- chip
  25. [25]
    GenePix Microarray Systems - Molecular Devices
    GenePix® Pro Microarray Analysis Software. Designed to work with GenePix systems as a complete, integrated platform. The seamless communication between scanner ...
  26. [26]
    Chromatin Immunoprecipitation (ChIP) on Chip Experiments ...
    Hybridized slides were scanned with the GenePix 4000A scanner (Axon), and the acquired images were analyzed with the software GenePix Pro, Version 3.0. A ...
  27. [27]
    Normalization and experimental design for ChIP-chip data - PMC
    The purpose of the mock control experiment is to correct dye-bias and other systematic errors in ChIP-chip experiments. ... For the two ChIP-chip replicates ...Missing: swap | Show results with:swap
  28. [28]
    Model-based analysis of tiling-arrays for ChIP-chip - PMC - NIH
    Aug 15, 2006 · In this work, we propose a fast and powerful analysis algorithm, titled Model-based Analysis of Tiling-arrays (MAT), to identify regions ...Results · Method Comparisons · MethodsMissing: limma MA2C
  29. [29]
    TileMap
    TileMap is a tool designed for tiling array data analysis. It can be used to identify genomic loci that show transcriptional activities and transcription factor ...
  30. [30]
    Model-based analysis of two-color arrays (MA2C) - PMC
    ChIP-chip technology has quickly become popular among biologists, and high-density tiling microarrays are increasingly being used in novel genomic research.
  31. [31]
    An integrated system CisGenome for analyzing ChIP-chip and ... - NIH
    CisGenome is a software system for analyzing genome-wide chromatin immunoprecipitation (ChIP) data. It is designed to meet all basic needs of ChIP data ...Missing: limma | Show results with:limma
  32. [32]
    ArrayStar | DNASTAR
    ArrayStar provides in-depth gene expression and variant analysis, following an assembly of genomic or transcriptomic data in SeqMan NGen.
  33. [33]
    ChIP-Chip: Algorithms for Calling Binding Sites - PMC - NIH
    We focus on the analysis of ChIP-chip data on two of the most commonly used tiling array platforms for ChIP-chip, Affymetrix and NimbleGen, emphasizing methods ...
  34. [34]
    An efficient pseudomedian filter for tiling microrrays - PubMed Central
    The most widely employed algorithm for tiling array analysis involves smoothing observed signals by computing pseudomedians within sliding windows.
  35. [35]
    ChIP-chip versus ChIP-seq: Lessons for experimental design and ...
    Feb 28, 2011 · The genomic coverage of ChIP-chip is limited by the microarray probe design, and the coverage of ChIP-seq is dependent on sequencing depth. The ...
  36. [36]
    [PDF] Comparison of sequence-specific transcription factor determinations ...
    Sep 9, 2011 · ChIP-qPCR results for 12 transcription factors and found an overall good agreement between ChIP-seq signal and ChIP-qPCR enrichment. These ...
  37. [37]
    Bayesian modeling of ChIP-chip data using latent variables - PMC
    In this paper, we propose a Bayesian latent model for the ChIP-chip data. The new model mainly differs from the existing Bayesian models, such as the joint ...Missing: autocorrelation | Show results with:autocorrelation
  38. [38]
    [PDF] A Flexible and Powerful Bayesian Hierarchical Model for ChIP–Chip ...
    However, we take into ac- count the spatial dependence between probes by allowing the weights of the mixture to be correlated for neighboring probes on a ...
  39. [39]
    Sensitive and accurate identification of protein–DNA binding events ...
    Nov 4, 2010 · ... cost-effective technology for discovering protein–DNA binding events at the genome scale ... ChIP-chip is a high-throughput technology, and to ...
  40. [40]
    Computation for ChIP-seq and RNA-seq studies - Nature Methods
    **Summary of ChIP-on-chip Advantages and Strengths:**
  41. [41]
    ChIP-on-chip - Wikipedia
    The goal of ChIP-on-chip is to locate protein binding sites that may help identify functional elements in the genome. For example, in the case of a ...Technological platforms · Workflow of a ChIP-on-chip... · Strengths and weaknessesMissing: objectives seminal
  42. [42]
    ChIP-seq Advantages and Limitations - News-Medical.net
    Feb 26, 2019 · Limitations of ChIP-seq​​ ChIP-chip techniques cost around 400-800 USD per array, in comparison to ChIP-seq costs 1,000-2,000 USD per lane (for ...
  43. [43]
    Chromatin immunoprecipitation and microarray-based analysis of ...
    Genome-wide location analysis, also known as ChIP-Chip, combines chromatin immunoprecipitation and DNA microarray analysis to identify protein-DNA interactions ...
  44. [44]
    ChIP'ing the mammalian genome: technical advances and insights ...
    As described in this paper, combination of the ChIP assay with robust readout methods is extremely powerful for a variety of whole-genome analyses in order ...
  45. [45]
    ChIP-on-chip significance analysis reveals large-scale binding and ...
    Jan 6, 2009 · ChIP-on-chip has emerged as a powerful tool to dissect the complex network of regulatory interactions between transcription factors and ...Missing: foundational | Show results with:foundational
  46. [46]
    Pricing » Microarray and Sequencing Resource | Boston University
    Sequencing Data Analysis ; 1 – 6, $500.00 ; 7-12, $800.00 ; 13-18, $1000.00 ; 19-24, $1200.00 ; 25-30, $1400.00.
  47. [47]
  48. [48]
  49. [49]
  50. [50]
    Human Genome Placed on Chip; Biotech Rivals Put It Up for Sale
    Oct 2, 2003 · NimbleGen Systems, a small company in Madison, Wis., announced a few days later that it had a genome on a chip that it was not selling but that ...Missing: commercial 2003-2005
  51. [51]
    5528 PDFs | Review articles in CHIP-CHIP - ResearchGate
    Technologies include array comparative genomic hybridization (CGH), copy number variation (CNV), DNA methylation, ChIP-Chip, RNA splice variants, and microRNA.
  52. [52]
    TruSeq ChIP Library Preparation Kit - Illumina
    The TruSeq ChIP kit is a simple, cost-effective solution for ChIP-Seq, compatible with Illumina sequencers, and has a streamlined workflow with low DNA input.
  53. [53]
    Pricing - Heflin Center for Genomic Sciences
    A typical mRNA-Seq experiment will cost, $650/sample. A typical microRNA-Seq experiment will cost, $400/sample. A typical ChIP-Seq experiment will cost, $550/ ...
  54. [54]
    Dam mutants provide improved sensitivity and spatial resolution for ...
    Jun 13, 2019 · ... 100 bp bins) and hence how fast the signal varies. This supports an increase in spatial resolution with the mutants, with wild-type Dam ...
  55. [55]
    ChIP-exo: A Method to Identify Genomic Location of DNA-binding ...
    Oct 30, 2013 · ChIP-exo allows us to identify a nearly complete set of binding locations of DNA-bound proteins at near single nucleotide resolution with almost no background.
  56. [56]
    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.
  57. [57]
    An efficient targeted nuclease strategy for high-resolution mapping ...
    A new method, called CUT&RUN (which is short for “Cleavage Under Targets & Release Using Nuclease”) to map specific interactions between protein and DNA.
  58. [58]
    DamID as a versatile tool for understanding gene regulation - NIH
    Mar 15, 2019 · Although ChIP has been the predominant chromatin profiling method for many years, DamID offers a range of advantages for some applications.Missing: exo | Show results with:exo
  59. [59]
    DamID as a versatile tool for understanding gene regulation
    Mar 15, 2019 · DamID is an increasingly versatile tool and has recently been adapted in inventive ways to elucidate the molecular mechanisms of gene regulation.
  60. [60]
    Optimized ChIP-exo for mammalian cells and patterned sequencing ...
    Aug 20, 2025 · We validate MO-ChIP-exo by comparing it to previously published ChIP-exo protocols and demonstrate its adaptability to both suspension (K562) ...Missing: DamID CUT&RUN gaining traction single- low- input
  61. [61]
  62. [62]
  63. [63]
    ChIP-seq vs. ATAC-seq - CD Genomics
    While ATAC-seq complements ChIP-seq by providing a broader chromatin openness perspective, ChIP-seq ensures the direct identification of specific DNA ...