Fact-checked by Grok 2 weeks ago

Microarray

A microarray is a high-throughput laboratory technique in molecular biology that enables the simultaneous analysis of thousands to millions of biomolecules, such as DNA, RNA, proteins, or antibodies, by immobilizing them as probes on a solid substrate like a glass slide or silicon chip and detecting interactions with labeled target samples through hybridization or binding assays. Developed in the mid-1990s, microarray technology revolutionized genomics by providing a cost-effective alternative to full DNA sequencing for large-scale studies, allowing researchers to measure gene expression levels, identify genetic variations such as single nucleotide polymorphisms (SNPs), and detect mutations associated with diseases. The process typically involves spotting or synthesizing probes in a grid pattern on the substrate, hybridizing fluorescently labeled sample nucleic acids or proteins to complementary probes, and scanning the resulting fluorescence intensity to quantify binding, which reveals relative abundance or presence of targets. There are several types of microarrays, broadly categorized by the biomolecules involved. DNA microarrays, the most common, include spotted arrays where pre-synthesized DNA fragments are deposited robotically on coated glass slides, in situ synthesized arrays using photolithography or inkjet printing to build oligonucleotides directly on the surface, and bead-based arrays with DNA-attached microspheres in etched wells. Protein microarrays, on the other hand, immobilize proteins, peptides, antibodies, or glycans to study protein interactions, biomarker discovery, or immune responses, often employing functional, analytical, or reverse-phase formats for applications in diagnostics and drug development. Key applications of microarray technology span research and clinical settings. In genomics, DNA microarrays facilitate genome-wide association studies (GWAS) to link genetic variants with diseases like breast cancer or diabetes, measure differential gene expression between healthy and diseased tissues, and support prenatal diagnosis of chromosomal abnormalities via chromosomal microarray analysis (CMA). Protein and antibody microarrays aid in pathogen detection, such as identifying SARS-CoV-2 variants or multiple swine viruses with high sensitivity (e.g., down to 10² copies/µL). Additionally, they contribute to personalized medicine by enabling genotyping for pharmacogenomics and monitoring immune responses in vaccine development. Despite their versatility, microarrays have limitations, including reliance on prior knowledge of probe sequences, potential cross-hybridization leading to false positives, and lower resolution compared to next-generation sequencing (NGS). As of 2025, while DNA microarrays for genotyping and chromosomal analysis remain widely used in clinical diagnostics due to their speed and affordability, gene expression profiling has increasingly shifted to RNA sequencing for its unbiased detection and higher dynamic range, though microarrays continue to evolve with improvements in fabrication, such as 3D structures and label-free detection methods like surface plasmon resonance imaging (SPRi), alongside recent advancements including AI-enhanced analysis and integrated genotyping devices.

Overview

Definition and Principles

A microarray is a high-throughput analytical platform consisting of a solid support with an ordered array of microscopic spots, each containing immobilized biomolecular probes such as DNA oligonucleotides, cDNA fragments, or proteins, arranged in a grid-like format to enable the simultaneous detection and quantification of multiple target analytes through specific binding interactions. These probes are designed to capture complementary targets from a sample, such as nucleic acids or proteins, facilitating parallel analysis of thousands to millions of interactions in a single experiment. This technology extends beyond nucleic acids to include protein microarrays, where capture molecules like antibodies or antigens are spotted to study protein-protein, protein-DNA, or enzyme-substrate interactions. The fundamental principles of microarray operation rely on the immobilization of probes onto a substrate, followed by the application of a labeled target sample, selective binding (e.g., hybridization for nucleic acids or affinity interactions for proteins), and detection of bound targets via signal intensity proportional to their abundance. Substrates commonly include glass slides, silicon wafers, or plastic for their optical clarity and chemical stability, allowing precise spotting and scanning. Binding specificity ensures that only complementary targets hybridize or associate with probes under controlled conditions like temperature and salt concentration, minimizing non-specific interactions, while washing steps remove unbound material to enhance signal-to-noise ratios. Quantitative measurement typically involves fluorescence detection, where the intensity correlates with target concentration, enabling relative or absolute quantification. Key components include probes, which are short, sequence-specific molecules (e.g., 20-100 nucleotides for DNA or antibodies for proteins) that serve as capture agents; targets, the analyte molecules (e.g., mRNA-derived cDNA or cellular proteins) from the biological sample; solid substrates for probe attachment; and labeling strategies, such as fluorescent dyes like Cy3 (green) and Cy5 (red) for dual-color detection in comparative assays. These dyes are covalently linked to targets during preparation, allowing differential labeling of samples (e.g., control vs. experimental) for ratio-based analysis without needing identical probe amounts across arrays. The general workflow begins with probe array creation through spotting or synthesis on the substrate, followed by target sample preparation involving extraction, amplification if needed, and fluorescent labeling (e.g., reverse transcription of RNA to cDNA for nucleic acid arrays). Labeled targets are then incubated with the array under hybridization conditions to allow binding, excess targets are washed away, and the array is scanned using a laser confocal microscope to capture fluorescence signals from each spot, generating data for downstream analysis of target abundance. This streamlined process supports high-density arrays, such as those monitoring expression of over 45 genes in early implementations, scalable to genome-wide levels.

Historical Development

The concept of microarrays was first introduced in 1983 by Tse Wen Chang for antibody microarrays, as described in a scientific publication and US Patent 4,376,110, laying the groundwork for multiplexed biomolecular analysis. High-throughput DNA microarray technology originated in the early 1990s at Stanford University, where researchers Patrick O. Brown and Ronald W. Davis pioneered spotted complementary DNA (cDNA) arrays to study gene expression patterns on a genomic scale. These early arrays involved robotic spotting of DNA probes onto glass slides, enabling the simultaneous monitoring of thousands of genes through hybridization with labeled targets. The foundational work was detailed in a 1995 publication by Mark Schena, Dari Shalon, Davis, and Brown, which demonstrated quantitative gene expression analysis using fluorescent detection for 45 Arabidopsis thaliana genes, marking a significant advance over previous low-throughput methods like Northern blotting. A key milestone occurred in 1995 when Affymetrix introduced high-density oligonucleotide arrays fabricated via photolithography, allowing for the in situ synthesis of up to hundreds of thousands of short DNA probes on a single chip. This light-directed approach, building on earlier concepts from Stephen Fodor's team, enabled precise spatial control and massively parallel analysis, particularly for genotyping and expression profiling. The technology's first commercial application was an HIV genotyping GeneChip in 1994. Affymetrix expanded to eukaryotic gene expression arrays in the mid-1990s, with early products like the yeast genome array released around 1997, solidifying the platform's impact. During the late 1990s and 2000s, microarray technology expanded rapidly through commercialization by major companies, transitioning from academic prototypes to standardized platforms. Affymetrix dominated early oligo-based markets, while Agilent Technologies launched inkjet in situ synthesis arrays in 2000, offering customizable probe lengths and higher flexibility. Illumina introduced bead-based arrays in 2003, utilizing silica microbeads for genotyping and expression studies, which improved throughput and reduced costs. Protein microarrays also advanced during this period, with formats for antibody and antigen arrays emerging for proteomics applications in the early 2000s. This era also saw a shift from two-color microarray formats—common in spotted cDNA arrays for comparative hybridization—to single-color formats in commercial oligo and bead systems, enhancing data consistency and simplifying analysis. In the 2010s, advancements integrated microarrays with next-generation sequencing (NGS) for hybrid workflows, such as microarray-based target capture to enrich specific genomic regions before sequencing, improving efficiency for clinical diagnostics and large-scale studies. Bead-based systems from Illumina further evolved, supporting multiplexed assays for epigenetics and copy number variation. By the early 2020s, innovations like nanomaterial-enhanced detection, including silver island films for metal-enhanced fluorescence, boosted sensitivity for low-abundance targets. The global microarrays market reached approximately USD 6.5 billion in 2024, fueled by post-COVID demand for diagnostic applications in infectious disease monitoring and personalized medicine.

Types of Microarrays

DNA Microarrays

DNA microarrays are specialized arrays designed for the analysis of nucleic acids, featuring probes that are typically short oligonucleotides of 25-70 bases or longer cDNA fragments, each representing specific genes, exons, or genomic regions. These probes are immobilized on a solid substrate, such as glass or silicon, allowing for high-density arrangements with up to millions of features per array, enabling parallel interrogation of thousands to entire genomes. Common formats include spotted arrays where pre-synthesized probes are deposited, in situ synthesized arrays built directly on the surface, and bead-based arrays using DNA-attached microspheres in etched wells for flexible genotyping applications. The primary applications of DNA microarrays include gene expression profiling, where they measure mRNA levels to assess transcriptional activity across samples; comparative genomic hybridization (CGH), which detects copy number variations by comparing test and reference DNA; and single nucleotide polymorphism (SNP) genotyping, which identifies genetic variants for association studies. For instance, array CGH, pioneered in high-resolution formats, uses differentially labeled genomic DNA hybridized to arrays to reveal chromosomal gains or losses with precision down to kilobase scales, as demonstrated in early applications to cancer genomes. Similarly, SNP genotyping via microarrays, with early assays targeting over 1,400 loci, has scaled to genome-wide coverage for population genetics and disease mapping. DNA microarrays operate in two main formats: two-color systems, which involve competitive hybridization of two samples labeled with distinct fluorophores like Cy3 (green) and Cy5 (red) on the same array for direct ratio-based comparisons, and one-color systems, such as the Affymetrix GeneChip, where each sample is hybridized separately with a single label and data normalized across arrays. Whole-genome arrays cover the entire genome, while focused arrays target specific pathways or regions for cost-effective analysis. A defining feature of DNA microarrays is their reliance on sequence-specific hybridization, governed by Watson-Crick base pairing, where target DNA or RNA binds complementarily to probes, though challenges like cross-hybridization—non-specific binding due to sequence similarity—can introduce errors, particularly in complex eukaryotic genomes, necessitating careful probe design and validation. This principle builds on fundamental nucleic acid hybridization, allowing quantitative readout of binding affinity through fluorescence intensity.

Protein Microarrays

Protein microarrays, also known as protein chips, are high-throughput platforms where probes consisting of purified proteins, antibodies, or peptides are immobilized on a solid surface, such as glass slides or nitrocellulose membranes, to enable the simultaneous analysis of multiple protein interactions or activities. Unlike nucleic acid-based arrays, these platforms rely on affinity-based capture rather than hybridization, with immobilization techniques ensuring the probes retain their native conformation for functional assays. They are broadly classified into analytical arrays, which focus on detecting and quantifying protein analytes from complex samples, and functional arrays, which assess enzymatic activities or binding events, such as kinase-substrate interactions where thousands of substrates are screened against a kinase to map phosphorylation sites. For instance, functional arrays have identified over 280 kinase substrates in human proteomes, highlighting their utility in signaling pathway elucidation. Primary applications of protein microarrays include studying protein-protein interactions, profiling antibody specificities, and discovering biomarkers in serum or tissue samples. In protein-protein interaction studies, arrays featuring the entire yeast proteome (over 5,800 proteins) have detected interactions like those involving calmodulin with 30 targets, providing insights into cellular networks. Antibody profiling uses arrays to evaluate monoclonal antibody binding to immobilized antigens, ensuring specificity and reducing off-target effects in therapeutic development. For biomarker discovery, arrays screen serum for autoantibodies, such as identifying three specific markers for autoimmune hepatitis diagnosis with high sensitivity. Protein microarrays operate in two main formats: forward-phase and reverse-phase, each suited to different analytical needs.
FormatDescriptionKey Features and Examples
Forward-PhasePurified probes (e.g., antibodies or proteins) are immobilized on the surface; complex samples (e.g., serum) are flowed over the array for capture.High-throughput analyte detection; used for simultaneous profiling of multiple proteins in one sample, such as cytokine arrays for immune response analysis.
Reverse-PhaseComplex samples (e.g., cell lysates or tissue extracts) are spotted onto the surface; specific probes (e.g., antibodies) are applied to detect targets.Quantitative pathway analysis with minimal sample; applied to microdissected tumors to measure phosphorylated proteins in signaling cascades.
Challenges in protein microarray design include maintaining protein stability during immobilization and storage, as denaturation can impair activity, and ensuring proper orientation to expose functional domains, often addressed through oriented capture via tags like His or biotin. Non-specific binding, a common issue leading to false positives, is mitigated by pre-treating arrays with blocking agents such as bovine serum albumin (BSA) or non-fat milk to occupy unbound sites on the surface. Detection in protein microarrays typically employs ELISA-like sandwich assays, where a secondary antibody with an enzyme conjugate amplifies the signal, or direct fluorescence labeling of probes for multiplexed readout via laser scanning. These methods achieve sensitivities down to the femtogram level, particularly with tyramide signal amplification in reverse-phase formats, enabling detection of low-abundance proteins without extensive sample processing.

Other Specialized Microarrays

Tissue microarrays (TMAs) consist of small cylindrical cores, typically 0.6 to 2 mm in diameter, extracted from paraffin-embedded donor tissue blocks and arrayed into a single recipient paraffin block for parallel analysis. This format enables high-throughput immunohistochemical (IHC) staining and molecular pathology assessments across hundreds of tissue samples simultaneously, preserving the spatial architecture and histological context of the original tissues to facilitate comparative studies of protein expression patterns. In oncology, TMAs have been instrumental for profiling cancer subtypes, such as identifying differential biomarker expression in breast and prostate tumors, allowing validation of therapeutic targets in large cohorts with minimal tissue consumption. The technique's ability to maintain spatial relationships between cellular components, like tumor-stroma interactions, distinguishes it from dissociated sample analyses and supports prognostic evaluations in pathology. Cell microarrays involve the precise spotting or printing of live or fixed cells onto substrates, often in defined patterns, to enable high-throughput functional assays with reduced reagent volumes and cell numbers compared to traditional well-plate formats. These arrays are particularly suited for drug screening and toxicity testing, where individual spots containing as few as 10-100 cells can be exposed to compounds and monitored for responses like proliferation, apoptosis, or morphological changes via fluorescence imaging. For instance, they have been used to assess chemotherapeutic sensitivity in cancer cell lines and evaluate environmental toxins on primary hepatocytes, accelerating lead compound identification. Three-dimensional (3D) cell microarrays extend this capability by incorporating cells within hydrogel matrices, such as collagen or alginate, to mimic extracellular environments and sustain long-term viability—often exceeding 90% cell survival over weeks—while enabling assays for invasion, differentiation, and spheroid formation in contexts like stem cell research. This hydrogel encapsulation preserves cellular viability and three-dimensional organization, contrasting with two-dimensional arrays that may alter cell behavior due to substrate rigidity. Beyond tissue and cell variants, other specialized microarrays address distinct biomolecular interactions. Carbohydrate microarrays, or glycan arrays, display diverse oligosaccharides and polysaccharides immobilized on surfaces to probe glycan-binding proteins (GBPs) and lectins, advancing glycomics by revealing roles in cell adhesion, immune recognition, and pathogen-host interactions. These arrays have decoded specificity in mammalian siglec proteins and microbial adhesins, supporting vaccine design against bacterial infections. Small molecule microarrays (SMMs) facilitate screening of compound libraries for binding to drug targets, such as enzymes or receptors, by covalently attaching diverse small molecules to slides for fluorescence-based detection of interactions, which has identified inhibitors for protein kinases and RNA structures in high-throughput campaigns. Emerging nanoarrays leverage nanoscale patterning, often via DNA origami or nanopores, to achieve single-molecule detection sensitivity, enabling real-time observation of biomolecular dynamics like protein unfolding or nucleic acid hybridization without ensemble averaging. These nano-scale platforms, with spot sizes below 100 nm, enhance resolution for rare event analysis in diagnostics and biophysics. As of 2025, advances in suspension microarrays, including bead-based formats with integrated fluorescence signal amplification, have improved multiplexing for biosensing applications.

Fabrication Techniques

Spotting Methods

Spotting methods encompass physical deposition techniques used to fabricate microarrays by transferring pre-synthesized probes, such as DNA or proteins, onto a solid substrate in a defined pattern. These approaches enable the creation of high-density arrays for applications in genomics and beyond, with spot diameters typically ranging from 50 to 600 μm depending on the method. Unlike in situ synthesis, spotting relies on robotic systems to apply probe solutions, allowing flexibility in probe selection and customization. Contact printing involves direct mechanical transfer of probe solutions using pins or microstamps that physically contact the substrate. Quill pins, which load sample via capillary action within a split tip, and solid pins, which retain a small droplet on a flat or pointed surface, are widely used variants. These produce spot sizes of 100–200 μm and densities up to 10,000 spots/cm², suitable for low- to medium-throughput arrays. The seminal development of contact printing was demonstrated by Schena et al. in 1995, who used high-speed robotic printing of complementary DNAs on glass slides to monitor yeast gene expression patterns quantitatively. Robotic arrayers employing quill or solid pins, such as those based on TeleChem's Stealth technology, facilitate reproducible deposition across multiple slides. Non-contact printing dispenses probe solutions through ejection mechanisms without substrate contact, minimizing mechanical damage and contamination. Piezoelectric inkjet systems generate pressure waves to eject nanoliter droplets (0.1–1 nL), while bubble-jet (thermal) methods use heat to vaporize solvent for droplet formation. These techniques achieve spot sizes of 100–150 μm and slightly higher densities than contact methods, up to 10,000–30,000 spots per slide, with advantages in precision and adaptability for custom probe layouts. Glass slides coated with amino-silane, such as aminopropylsilane (APS), serve as the primary substrates, providing a positively charged surface for electrostatic immobilization of negatively charged probes like DNA oligonucleotides. Post-spotting treatments, including UV cross-linking at 65 mJ of 254 nm irradiation, covalently attach probes to the surface, improving array stability and signal intensity. Quality control in spotting methods emphasizes spot uniformity, density, and reproducibility to ensure reliable hybridization and data accuracy. Uniformity is optimized by adjusting solution viscosity, pin geometry, and substrate hydrophobicity, resulting in consistent spot morphology with minimal satellite droplets or bleeding. Densities of 400–10,000 spots/cm² are achievable, scaling with pin design and printing speed. Reproducibility metrics include positional accuracy of ±3 μm and coefficient of variation below 10% in spot intensity, as validated in controlled robotic systems. The MicroArray Quality Control (MAQC) project confirmed high reproducibility across spotting platforms when fabrication parameters are standardized, with inter-array correlation coefficients exceeding 0.99 for gene expression measurements.

In Situ Synthesis Methods

In situ synthesis methods involve the on-array construction of oligonucleotide probes through sequential chemical reactions, enabling the creation of high-density microarrays without pre-synthesizing and depositing probes. These techniques build probes base by base directly on the substrate, typically using solid-phase synthesis chemistry adapted for parallel, spatially controlled addition of nucleotides. This approach contrasts with mechanical deposition by allowing precise control over probe sequences and positions at the molecular level. Photolithography represents a foundational in situ method, pioneered for DNA microarrays by Affymetrix in their GeneChip platform. It employs light-directed synthesis where photolabile protecting groups on nucleotides are selectively removed using ultraviolet (UV) light through patterned masks or projectors, followed by coupling of the next protected nucleotide. This stepwise deprotection and extension cycle is repeated for each base position across the array, synthesizing probes up to 25 nucleotides long, which facilitates single-nucleotide mismatch discrimination in hybridization assays. The process utilizes chrome masks to define illuminated regions, achieving feature sizes as small as 5 micrometers and enabling arrays with densities exceeding 500,000 probes per square centimeter. Maskless array synthesis builds on photolithography by eliminating physical masks, using digital micromirror devices (DMDs) to project programmable light patterns onto the synthesis surface. Developed as an advancement for flexible probe design, this method generates virtual masks via computer software, which control the orientation of thousands of micromirrors to direct UV light selectively. It supports custom array configurations without mask fabrication costs and times, producing microarrays with over 76,000 features in a single synthesis run. DMD-based systems maintain high resolution, with feature densities up to 1 million per square centimeter, and are particularly suited for rapid prototyping of diverse probe sets. Emerging electrochemical and enzymatic methods offer alternatives for in situ synthesis, addressing limitations in yield and length for longer probes. Electrochemical approaches use electrode arrays to generate localized pH changes or redox reactions that deprotect nucleotides, enabling site-specific base addition with conventional phosphoramidite chemistry. This technique has demonstrated synthesis of oligonucleotides up to 50 bases with improved uniformity on microarrays. Enzymatic methods, such as those employing terminal deoxynucleotidyl transferase (TdT) or template-directed polymerases, incorporate nucleotides via biocatalyzed extension, providing higher fidelity and error rates below 1% for probes exceeding 100 bases. These methods support integration of modified bases and are gaining traction for applications requiring longer or chemically diverse sequences. Overall, in situ synthesis methods excel in producing ultra-high-density microarrays, with capabilities up to 1 million features per square centimeter, far surpassing spotting techniques in spatial resolution and probe uniformity. The 25-mer probes commonly used enhance specificity for detecting sequence variations, making these arrays ideal for genome-wide analyses.

Operation and Detection

Sample Preparation and Hybridization

Sample preparation for microarray experiments begins with the extraction of nucleic acids from biological samples, such as total RNA or genomic DNA, using methods like phenol-chloroform extraction or column-based purification kits to ensure high purity and integrity. Quality assessment, often via spectrophotometry or bioanalyzer, confirms RNA integrity number (RIN) above 7-8 to minimize degradation effects. Amplification is typically required for low-abundance samples; linear methods like T7-based in vitro transcription, pioneered by Van Gelder et al. in 1990, generate amplified RNA (aRNA) from cDNA primed with oligo-dT-T7 adapters, yielding 10^3 to 10^5-fold increase while preserving relative transcript abundances with correlations of 0.8-0.84. Alternatively, PCR amplification provides higher yields (up to 10^9-fold or more) but introduces biases toward shorter transcripts and non-linear representation. Labeling integrates during amplification: fluorophores like Cy3 or Cy5 are incorporated via nucleotide analogs in reverse transcription or transcription steps, or biotin for indirect detection, targeting specific activities of at least 8 pmol/μg for optimal signal. Hybridization involves incubating the labeled target (e.g., fragmented cRNA at 50-100 nt) with the immobilized probes on the microarray surface under controlled conditions to promote specific binding. Typical protocols use temperatures of 42-65°C for 16-17 hours in buffers like 2× SSC (sodium citrate-saline) with 0.005-0.1% detergents to stabilize duplexes and reduce non-specific interactions. The binding kinetics follow the Langmuir isotherm model, where surface coverage θ approaches equilibrium as θ = (K P) / (1 + K P), with K as the equilibrium constant and P as target concentration, describing reversible adsorption without multilayer formation. Post-hybridization washing removes unbound or weakly bound targets through stringent conditions, such as sequential rinses in 1× to 0.1× SSC with 0.1% SDS at room temperature or 37°C for 1-5 minutes each, often incorporating 20-50% formamide to increase stringency by lowering the effective melting temperature and minimizing mismatches. These steps ensure high specificity, with formamide reducing non-specific binding by disrupting partial hybrids. Efficiency of hybridization depends on probe-target complementarity, where perfect matches yield stronger signals than mismatches differing by 1-3 bases, and on the melting temperature (Tm), roughly estimated for short oligos as Tm = 4(G+C) + 2(A+T) °C to guide condition optimization and avoid off-target binding. GC content and secondary structures further influence binding stability, with higher GC pairs favoring duplex formation under standard salt conditions.

Signal Detection and Scanning

Signal detection in microarrays primarily relies on fluorescence-based methods, where fluorescently labeled target molecules bound to the array probes emit light upon excitation, allowing for the quantification of hybridization events. In fluorescence scanning, a laser excites the fluorophores (such as Cy3 at 532 nm or Cy5 at 635 nm), and the resulting emitted light is captured by photomultiplier tubes (PMTs) to convert photons into electrical signals for imaging. Scanning hardware typically employs confocal laser scanners to achieve high-resolution imaging of the array surface, focusing light to minimize out-of-focus signals and enable precise spot mapping. Commercial systems like the Agilent SureScan and the Axon GenePix series use dual-laser setups for multi-color detection, with resolutions commonly set at 5-10 μm per pixel to balance detail and scan speed for typical spot sizes of 100-200 μm. Quantification begins with converting raw pixel intensities from the scanned image into signal values representing bound target abundance, often using software to segment spots and compute mean or median foreground intensities. Background noise, arising from unbound labels or slide autofluorescence, is subtracted via local methods, such as median filtering of pixels surrounding each spot to estimate and deduct non-specific signal. In multi-channel setups, such as two-color DNA microarrays, separate images are acquired for each fluorophore, enabling ratio-based analysis for comparative expression; for instance, differential gene expression is derived from the log base 2 ratio of Cy5 to Cy3 intensities, log₂(Cy5/Cy3), after background correction. Alternative detection mechanisms include chemiluminescence, where enzymatic reactions produce light without external excitation, suitable for antibody-based protein microarrays to avoid fluorescence quenching issues, and mass spectrometry, which directly measures molecular weights of bound analytes in proteomics arrays for label-free quantification.

Applications

Genomics and Transcriptomics

Microarrays have revolutionized genomics and transcriptomics by enabling the simultaneous measurement of mRNA levels for thousands of genes, providing insights into gene expression patterns under various conditions. In gene expression analysis, DNA microarrays hybridize labeled cDNA derived from cellular mRNA to probes on the array, allowing quantification of transcript abundance across the genome. This approach has been instrumental in identifying differentially expressed genes in diseases such as cancer; for instance, microarray profiling of breast tumors revealed overexpression of the HER2 gene in a distinct subtype characterized by aggressive proliferation and poor prognosis. Such analyses typically involve comparing expression profiles between diseased and normal tissues to pinpoint biomarkers, with studies demonstrating that HER2-positive breast cancers exhibit elevated expression of genes involved in cell cycle progression and growth signaling. Beyond transcriptomics, microarrays facilitate genomic applications like comparative genomic hybridization (CGH), which detects chromosomal copy number variations such as aneuploidies by comparing test and reference DNA samples labeled with different fluorophores. Array CGH, an advancement over traditional metaphase CGH, uses densely arrayed genomic probes to achieve higher resolution, enabling the identification of submicroscopic deletions and duplications associated with genetic disorders and cancers. For example, it has been widely applied in prenatal diagnostics to detect aneuploidies like trisomy 21 with sensitivity comparable to karyotyping but faster turnaround. Another key application is ChIP-on-chip, which combines chromatin immunoprecipitation with microarray hybridization to map transcription factor binding sites genome-wide. This technique has elucidated regulatory networks by identifying where factors like RNA polymerase II bind during stress responses in yeast, revealing dynamic gene activation patterns. Case studies highlight microarrays' role in large-scale genomic projects. During the Human Genome Project era, microarray technology, pioneered in the mid-1990s, supported functional annotation by monitoring expression of thousands of genes in parallel, aiding the transition from sequence data to understanding gene functions. In model organisms, transcriptomic profiling via microarrays in yeast (Saccharomyces cerevisiae) provided early demonstrations of genome-wide expression dynamics, such as during the diauxic shift from glucose fermentation to respiration, where over 1,000 genes showed coordinated temporal changes. These efforts established microarrays as a cornerstone for systems biology. The impact of microarray applications in genomics and transcriptomics is evident in the discovery of prognostic gene signatures. A notable example is the 70-gene signature derived from breast cancer microarray data, which stratifies node-negative patients into low- and high-risk groups for recurrence, outperforming traditional clinical factors and guiding adjuvant therapy decisions. Validated in prospective trials, this signature has influenced clinical practice by identifying patients who may avoid chemotherapy, demonstrating microarrays' potential to personalize medicine through nucleic acid-level insights.

Proteomics and Diagnostics

Protein microarrays enable the high-throughput analysis of protein interactions and post-translational modifications (PTMs), facilitating comprehensive proteomics studies by immobilizing thousands of proteins or peptides on a solid surface for simultaneous interrogation with complex samples. These arrays are particularly valuable for mapping protein-protein interactions, where bait proteins are arrayed and probed with cellular extracts to identify binding partners, revealing functional networks in signaling pathways. For PTM analysis, arrays functionalized with modified peptides allow detection of kinase substrates, phosphorylation sites, and ubiquitination patterns, aiding in the elucidation of regulatory mechanisms underlying cellular processes. In autoimmune disease profiling, autoantigen arrays display a diverse panel of self-antigens, recombinant proteins, and peptides to capture and characterize autoantibody repertoires from patient sera, enabling the identification of disease-specific signatures. These arrays have been instrumental in studying systemic lupus erythematosus (SLE), where multiplex profiling reveals epitope spreading and autoantibody diversity, correlating with disease activity and progression. By quantifying reactivity against hundreds of autoantigens in a single assay, such platforms support early diagnosis and monitoring of autoimmune conditions like rheumatoid arthritis and multiple sclerosis. For diagnostics, protein microarrays serve as biomarker panels to detect disease-associated proteins in clinical samples, with cytokine arrays exemplifying their role in inflammation assessment by simultaneously measuring multiple pro- and anti-inflammatory cytokines such as IL-6, TNF-α, and IL-10. These arrays provide quantitative profiles that distinguish inflammatory states in conditions like sepsis or chronic inflammatory diseases, offering higher sensitivity and specificity than single-analyte tests. Adaptations for point-of-care use in infectious diseases integrate protein microarrays with microfluidic amplification, enabling rapid detection of pathogen-specific antibodies or antigens in resource-limited settings, as demonstrated in assays for HIV and tuberculosis biomarkers. Case studies highlight protein microarrays' utility in drug target validation, where functional arrays screen small molecules or antibodies against immobilized kinases and receptors to confirm selectivity and off-target effects, accelerating lead optimization in oncology drug development. In personalized medicine, these arrays integrate with pharmacoproteomics to profile patient-specific protein responses to therapies, such as monitoring drug metabolism enzymes or therapeutic antibodies, thereby guiding dosing and predicting adverse reactions beyond genomic pharmacogenomics alone. Regulatory advancements underscore the clinical translation of protein microarrays, with several products cleared by the U.S. Food and Drug Administration (FDA) for diagnostic applications, such as the Ig_Plex Celiac DGP Panel for detecting autoantibodies associated with celiac disease. These approvals ensure standardized performance and analytical validation, facilitating their incorporation into routine clinical workflows for biomarker-driven diagnostics.

Data Analysis Methods

Preprocessing and Normalization

Preprocessing and normalization are essential initial steps in microarray data analysis, transforming raw scanned images into reliable expression measures by addressing technical artifacts, noise, and systematic biases. These processes begin with image processing to accurately delineate spots and flag irregularities, followed by background correction to isolate true signals, normalization to ensure comparability across arrays, and quality assessment to validate the results. Proper execution minimizes non-biological variation, enabling downstream statistical inference. Image processing starts with grid alignment, which positions a virtual grid over the scanned microarray image to define spot locations, often using techniques like the Radon transform for rotation correction and maximum margin classifiers to optimize line placement between spot rows and columns, achieving high accuracy even in noisy images. Spot segmentation then isolates individual spots, employing methods such as fixed-circle algorithms for uniform spots or adaptive approaches like unsupervised clustering with partial differential equations to handle irregular contours, inner holes, or varying sizes, thereby extracting foreground intensities more precisely. Artifacts, including scratches or dust, are flagged through outlier detection in size distributions or clustering anomalies, excluding affected spots from further analysis to prevent signal distortion. Background correction subtracts non-specific fluorescence noise, such as from unbound probes or slide imperfections, using local methods that estimate and deduct intensity from surrounding regions for each spot in two-color arrays. For single-channel oligonucleotide arrays, the Robust Multi-array Average (RMA) method models perfect match probe intensities as signal plus background, assuming a log-normal background distribution and applying a transformation to recover expected signal values, effectively reducing noise while preserving low-expression data. These corrections enhance signal-to-noise ratios without introducing bias. Normalization standardizes intensities across arrays to account for technical variations like dye biases or hybridization efficiencies. In two-color microarrays, dye-swap designs—where fluorophores are reversed in replicate hybridizations—correct for channel-specific effects by averaging log-ratios from paired slides, balancing differential labeling responses. Global approaches, such as quantile normalization, equalize probe intensity distributions by ranking values across arrays and replacing them with rank averages, ensuring identical empirical distributions and reducing between-array variance in high-density oligonucleotide data. Quality metrics evaluate preprocessing efficacy, with MA plots visualizing log-ratio (M) versus average intensity (A) to detect remaining biases post-normalization; well-normalized data show scatter around zero without trends. Spike-in controls, predefined RNA transcripts added at known concentrations, serve as calibration standards, confirming accurate signal recovery and dynamic range across experiments.

Statistical Interpretation

Statistical interpretation of microarray data involves applying statistical methods to identify biologically significant patterns and differences in gene expression levels from normalized datasets. These methods build on preprocessed data to test hypotheses, control for errors, and uncover functional insights. Differential expression analysis is a core approach to detect genes whose expression levels differ significantly between experimental conditions, such as treated versus control samples. For two-group comparisons, the t-test is commonly used to assess whether the mean expression differs, often combined with a fold-change metric to quantify the magnitude of change; a typical threshold is a fold-change greater than 2 with a p-value less than 0.05 to balance biological relevance and statistical significance. For multi-group designs, analysis of variance (ANOVA) extends this by evaluating overall differences across groups, followed by post-hoc tests for pairwise comparisons. Given the high dimensionality of microarray data, with thousands of genes tested simultaneously, multiple testing correction is essential to control the false discovery rate (FDR), which estimates the proportion of false positives among significant results. The Benjamini-Hochberg procedure, a widely adopted step-up method, adjusts p-values to maintain FDR at a desired level, such as 5%, thereby reducing spurious findings in large-scale analyses. To discover patterns and relationships, clustering techniques group genes or samples with similar expression profiles, while classification methods predict sample categories. Hierarchical clustering, often using average linkage and Euclidean distance, visualizes expression patterns as dendrograms to reveal co-regulated genes or sample subtypes. Principal component analysis (PCA) reduces dimensionality by projecting data onto principal components that capture maximum variance, aiding in the identification of underlying structure and outlier detection. For classification, support vector machines (SVM) have been effectively applied to distinguish disease states based on gene expression signatures, leveraging kernel functions to handle non-linear separations. Pathway analysis integrates differentially expressed genes with prior biological knowledge to infer affected cellular processes. Tools like DAVID perform gene ontology (GO) enrichment to identify overrepresented functional categories among selected genes, using hypergeometric tests corrected for multiple comparisons. Gene Set Enrichment Analysis (GSEA) evaluates whether predefined gene sets, such as those from KEGG pathways, show coordinated up- or down-regulation, computing an enrichment score based on ranked gene lists to detect subtle pathway perturbations. Integration with databases like KEGG maps these genes onto metabolic or signaling pathways, highlighting dysregulated networks.

Advantages, Limitations, and Future Directions

Strengths and Challenges

Microarray technology offers significant strengths in high-throughput analysis, enabling the simultaneous interrogation of thousands of genes or proteins in a single experiment, which facilitates rapid screening of complex biological samples. This capability supports applications in genomics and proteomics by allowing multiplexing of hundreds to thousands of probes on a single chip, reducing the need for multiple individual assays. Additionally, microarrays are cost-effective for targeted studies, with per-sample costs typically ranging from $100 to $500 in academic and core facility settings as of 2025, making them more affordable than comprehensive next-generation sequencing (NGS) for predefined gene sets. For validated probes, reproducibility is high, particularly when sequence-verified designs are used, achieving correlation coefficients above 0.95 in replicate experiments and enabling reliable differential expression detection across platforms. Despite these advantages, microarrays face notable challenges, including static probe sets that are limited to predefined, known sequences, restricting discovery of novel variants or transcripts not included in the array design. Cross-hybridization errors arise from non-specific binding between similar sequences, which can reduce specificity and introduce noise, especially in samples with homologous genes like ribosomal RNAs. The dynamic range of detection is another limitation, typically spanning 3 orders of magnitude due to background noise at low expression levels and signal saturation at high levels, compared to over 5 orders of magnitude in NGS approaches. Technical issues further complicate microarray use, as experiments require high-quality RNA—typically 100 ng to 1 µg of total RNA per sample, though amplification enables lower inputs from limited or degraded clinical specimens—to ensure sufficient material for labeling and hybridization. Sensitivity for low-abundance targets is also constrained, with detection limits potentially missing transcripts below 1% of total RNA, necessitating amplification steps that may introduce bias. Environmental factors, such as variations in hybridization temperature and humidity, can affect probe stability and signal uniformity, leading to inconsistent results if not tightly controlled. In comparison to alternatives like NGS, microarrays provide faster turnaround times—often hours to days for targeted analysis—versus the more time-intensive sequencing workflows, though they offer less comprehensive coverage of the genome or proteome. For specific microarray types, such as protein arrays, additional challenges include the instability of immobilized proteins, which can denature or lose activity over time, reducing assay reliability compared to more stable nucleic acid-based formats. Recent advancements in microarray technology emphasize the incorporation of nanomaterials to boost detection sensitivity, particularly in nucleic acid assays. Gold nanoparticles, for instance, have been integrated into microarray platforms to amplify fluorescence signals through surface plasmon resonance effects, enabling detection limits as low as single-molecule levels in biosensing applications. This enhancement addresses limitations in traditional microarrays by improving signal-to-noise ratios and reducing background interference, as demonstrated in recent developments of gold nanochip-based systems for high-throughput biomarker analysis. Market trends indicate robust growth for microarray technologies, with the global market valued at USD 6.22 billion in 2024 and projected to reach USD 8.46 billion by 2030, reflecting a compound annual growth rate (CAGR) of 5.48%. This expansion is propelled by rising applications in personalized medicine and diagnostics, alongside a post-2020 pandemic shift toward multiplexed kits that allow simultaneous detection of multiple pathogens. For example, microarray-based assays like CovidArray have emerged for rapid SARS-CoV-2 screening from nasopharyngeal swabs, achieving limits of detection around 1 copy/μL with full workflow completion in under 2 hours, thereby supporting scalable infectious disease monitoring. Hybrid approaches combining microarrays with next-generation sequencing (NGS) are gaining traction for enhanced validation and accuracy in genomic profiling. Recent studies have compared hybrid SNP microarrays with nanopore NGS for copy number variation (CNV) calling, showing concordant results at low coverage depths and improved resolution for structural variants in human cell lines. Similarly, single-cell microarrays integrated with microfluidics, such as microwell array chips, facilitate high-throughput isolation and analysis of individual cells, enabling detailed transcriptomic insights without bulk averaging effects. These combinations mitigate microarray limitations like resolution constraints by leveraging NGS depth for confirmatory sequencing. Looking ahead, AI-driven methods are optimizing microarray probe design by predicting hybridization efficiency and minimizing cross-reactivity, improving specificity. Machine learning algorithms analyze thermodynamic models and sequence features to refine probe sets, streamlining array fabrication for diverse applications. Additionally, portable microarray systems are advancing field diagnostics in global health contexts; a compact LED-based fluorescence reader, for instance, supports multiplexed biomarker detection for lupus nephritis using minimal sample volumes, correlating highly (r > 0.96) with laboratory scanners and enabling point-of-care testing in underserved areas. As of 2025, regulatory advancements include FDA clearance for novel microarray diagnostics, and hybrid systems integrating CRISPR with microarrays are emerging for enhanced pathogen detection. These innovations promise broader accessibility and integration with AI for real-time data interpretation in resource-limited settings.

References

  1. [1]
    Microarray Technology - National Human Genome Research Institute
    Microarray technology is a general laboratory approach that involves binding an array of thousands to millions of known nucleic acid fragments to a solid ...
  2. [2]
    Recent Progress in Development and Application of DNA, Protein ...
    Mar 27, 2023 · The aim of this review is to explore the development of biomolecule-based microarray applications since 2018 onwards.
  3. [3]
    DNA Microarray Technology Fact Sheet
    Aug 15, 2020 · A DNA microarray is a tool used to determine whether the DNA from a particular individual contains a mutation in genes.
  4. [4]
    DNA microarrays: Types, Applications and their future - PMC - NIH
    Microarrays are a technology in which 1000's of nucleic acids are bound to a surface and are used to measure the relative concentration of nucleic acid ...
  5. [5]
    DNA Microarray - Learn Genetics Utah
    DNA microarray analysis is a technique that scientists use to determine whether genes are on or off. Scientists know a gene is on in a cell if its mRNA is ...
  6. [6]
    Advances in chromosomal microarray analysis - PubMed Central - NIH
    Jan 25, 2025 · CMA is a molecular technique that is increasingly used in the prenatal diagnosis of chromosomal abnormalities, which can include abnormalities ...
  7. [7]
    An updated comparison of microarray and RNA-seq for ... - NIH
    Apr 23, 2025 · In the current study, we provide an updated comparison between microarray and RNA-seq with the most up-to-date technologies for both platforms.
  8. [8]
    A perspective on microarrays: current applications, pitfalls, and ...
    A microarray is typically defined as a collection of microscopic spots arranged in an array or grid-like format and attached to a solid surface or membrane, ...
  9. [9]
  10. [10]
    Basic Concepts of Microarrays and Potential Applications in Clinical ...
    This review highlights uses of microarray technology that impact diagnostic microbiology, including the detection and identification of pathogens.
  11. [11]
    Quantitative Monitoring of Gene Expression Patterns with ... - Science
    Mark Schena et al. ,. Quantitative Monitoring of Gene Expression Patterns with a Complementary DNA Microarray.Science270,467-470(1995).DOI:10.1126/science ...
  12. [12]
    The emergence and diffusion of DNA microarray technology - PMC
    Ron Davis had contributed to those early methods for identifying genes, and the same technique was used to fix DNA to slides as solid support for his later ...
  13. [13]
  14. [14]
    [PDF] On Limits of Performance of DNA Microarrays - Babak Hassibi
    DNA capturing probe has a length of 25-70 bases, depend- ing on the exact ... Typically, probe design is based on minimizing the amount of cross ...
  15. [15]
  16. [16]
    One-Color vs Two-Color Microarray- How to Choose the Right One?
    Jun 14, 2023 · The name itself defines that in a one-color microarray, the fragments are labeled with a single dye while in a two-color microarray, two ...One-color DNA microarray · Advantages · Two-color DNA MicroarrayMissing: formats | Show results with:formats
  17. [17]
    Protein Microarrays: Novel Developments and Applications - NIH
    Protein microarray technology possesses some of the greatest potential for providing direct information on protein function and potential drug targets.
  18. [18]
  19. [19]
    Reverse phase protein microarrays advance to use in clinical trials
    In a forward phase, or antibody array, multiple antibodies are immobilized on a surface to capture proteins from a sample.
  20. [20]
    Demystified … Tissue microarray technology - PMC - PubMed Central
    Tissue microarray (TMA) technology is one of these new tools. It is based on the idea of applying miniaturisation and a high throughput approach to the analysis ...
  21. [21]
    Tissue microarrays for high-throughput molecular pathology - NIH
    TMAs can be made not only from paraffin embedded tissues, but also from frozen tissue samples (Figure 3)., Frozen TMAs are well suited for RNA-ISH detection of ...
  22. [22]
    Tissue Microarrays in Clinical Oncology - PMC - PubMed Central
    TMAs are similarly useful in characterizing the immunohistochemical profile of cancer subtypes. For example, two studies have applied a large panel of ...
  23. [23]
    Advances in cancer tissue microarray technology - NIH
    Tissue microarray (TMA) is a powerful new technology designed to assess the expression of proteins or genes across large sets of tissue specimens assembled on a ...
  24. [24]
    Living Cell Microarrays: An Overview of Concepts - PMC - NIH
    Living cell microarrays are a highly efficient cellular screening system. Due to the low number of cells required per spot, cell microarrays enable the use ...
  25. [25]
    Influence of a Three-Dimensional, Microarray Environment on ... - NIH
    Sep 19, 2012 · We have used a modified 3D cellular microarray platform for the high-throughput analysis of growth, cytotoxicity, and protein expression ...
  26. [26]
    Cell Microarray Technologies for High-Throughput Cell-Based ...
    This review discusses the applications of the cell microarray including toxicology, drug discovery and detection of toxic agents.Cell Microarray Technologies... · 3. Cell Microarrays In A... · 3.1. Hydrogel-Based 3d Cell...
  27. [27]
    An adaptable hydrogel array format for 3-dimensional cell culture ...
    The adaptable, array-based format developed in this study may be useful as an enhanced throughput platform for 3-D culture of a variety of cell types.
  28. [28]
    Three-Dimensional Cell Culture Microarray for High-Throughput ...
    In this article we present a high-throughput, 3D cell-based microarray platform designed to allow the expansion and differentiation of embryonic stem (ES) cells ...
  29. [29]
    Glycan microarrays for decoding the glycome - PMC - NIH
    This review covers the development of glycan microarrays and applications that have provided insights into the roles of mammalian and microbial glycan binding ...Missing: seminal papers
  30. [30]
    Carbohydrate microarrays - PMC
    Nowadays, carbohydrate microarrays have become the leading edge tools for functional studies of glycans and GBPs because the microarray-based technology has the ...Missing: seminal papers
  31. [31]
    Small-molecule microarrays as tools in ligand discovery - PMC - NIH
    Small molecules that bind and modulate specific protein targets are increasingly used as tools to decipher protein function in a cellular context.Missing: impact | Show results with:impact
  32. [32]
    Sensitive single-molecule protein quantification and protein complex ...
    Here we demonstrate a single-molecule protein assay in a microarray format enabled by an ultra-low background surface and single-molecule imaging.
  33. [33]
    Bench-Top Fabrication of Single-Molecule Nanoarrays by DNA ...
    We introduce a cleanroom-free, $1 benchtop technique to create meso-to-macro-scale DNA origami nanoarrays using self-assembled colloidal nanoparticles.Missing: emerging | Show results with:emerging
  34. [34]
    [PDF] Bio-Microarray Fabrication Techniques—A Review
    A microarray is a two-dimensional arrangement of specific biological probes (e.g., DNA or protein molecules) deposited in an addressable fashion on a glass ...<|control11|><|separator|>
  35. [35]
  36. [36]
    Glass slides to DNA microarrays - ScienceDirect.com
    After DNA probes are immobilized on an aminosilane-coated surface by ionic attraction, ultraviolet (UV) cross-linking is generally used to achieve covalent ...
  37. [37]
    None
    ### Summary of Amino-Silane Coated Slides and Post-Spotting Treatments
  38. [38]
    The MicroArray Quality Control (MAQC) project shows inter
    The MicroArray Quality Control (MAQC) project shows inter- and intraplatform reproducibility of gene expression measurements. MAQC Consortium*. Author ...
  39. [39]
    Light-Directed, Spatially Addressable Parallel Chemical Synthesis
    Solid-phase chemistry, photolabile protecting groups, and photolithography have been combined to achieve light-directed, spatially addressable parallel ...
  40. [40]
    Photolithographic synthesis of high-density DNA probe arrays
    Dec 11, 2007 · This process has the ability to fabricate arrays with 1 micron feature pitch and smaller, providing an unprecedented sequence density of and greater.INTRODUCTION · DNA synthesis · Change in paradigm... · Synthesis efficiency
  41. [41]
    Efficiency, error and yield in light-directed maskless synthesis of ...
    Dec 8, 2011 · Maskless array synthesis (MAS) uses a digital micromirror device (DMD) in place of photomasks to deliver patterned ultraviolet light. The ...
  42. [42]
    Electrochemically directed synthesis of oligonucleotides for DNA ...
    We demonstrate a new method for making oligonucleotide microarrays by synthesis in situ. The method uses conventional DNA synthesis chemistry with an ...Hybridization To Synthesized... · Results · Discussion<|control11|><|separator|>
  43. [43]
    In situ enzymatic template replication on DNA microarrays
    Here, we describe an enzymatic approach to the synthesis of high-density DNA microarrays that can also contain chemical modifications.
  44. [44]
    Photolithographic synthesis of high-density DNA probe arrays
    Aug 6, 2025 · Light directed in situ synthesis can currently produce high density DNA microarrays with features up to 4 × 10 6 cm −2 [12] . The light ...
  45. [45]
    DNA Microarrays: Sample Quality Control, Array Hybridization ... - NIH
    Mar 15, 2011 · To prepare the hybridization sample, mix the following in a 1.5 ml tube: 825 ng Cy3-cRNA (this is usually your "treated" sample) · To load the ...Missing: seminal papers review
  46. [46]
    Microarray experiments and factors which affect their reliability
    Sep 3, 2015 · In this article we describe the individual steps of a microarray experiment, highlighting important elements and factors that may affect the processes involved.Missing: seminal | Show results with:seminal
  47. [47]
  48. [48]
    Robust and efficient synthetic method for forming DNA microarrays
    After hybridization, the array was washed at room temperature in 1× SSC, 0.1% SDS for 5 min, 0.1× SSC, 0.1% SDS for 5 min and 0.1× SSC for 5 min. The array ...
  49. [49]
  50. [50]
    [PDF] GenePix 4000B Microarray Scanner | Molecular Devices
    The GenePix 4000B Scanner acquires data at user-selectable resolutions between. 5–100 microns, allowing optimization of image resolution and file size for each ...
  51. [51]
    Microarray Scanners | Biocompare
    Resolution: 0.3 to 40 µm/pixel · Sample Type: Standard Microscope Slide · Lasers: 532 nm and 635 nm · Detector: Two PMT confocal detection.
  52. [52]
  53. [53]
    [PDF] GenePix® Pro - Microarray Acquisition and Analysis Software
    The local median intensity is the default background subtraction method. Local methods, where the background value for a feature is calculated from a region ...
  54. [54]
    comparison of background correction methods for two-colour ...
    Two-colour microarray experiments quantify the relative gene expression between experimental samples for thousands of probes simultaneously. The pixel ...Missing: color<|separator|>
  55. [55]
    Exploratory differential gene expression analysis in microarray ...
    Mar 1, 2004 · We discuss a model for such data in which the log2(Cy5) and log2(Cy3) values are linearly related and are samples drawn from a bivariate normal ...
  56. [56]
    Microarray methods for protein biomarker detection - PMC - NIH
    The most widely used techniques for the discovery and simultaneous profiling of multiple protein biomarkers are mass spectrometry,16-18 2-D western blotting,19 ...
  57. [57]
    Multiplexed chemiluminescence imaging assay of protein ...
    Nov 22, 2018 · A multiplexed chemiluminescence (CL) imaging assay was designed for sensitive screen detection of a panel of protein biomarkers by integrating DNA microarray
  58. [58]
    Differential Gene Expression Patterns in HER2/neu-Positive and
    The aim of this study was to identify genes associated with the aggressive phenotype of HER2/neu-positive breast cancer cells using cDNA microarrays.Missing: seminal | Show results with:seminal
  59. [59]
    A Gene-Expression Signature as a Predictor of Survival in Breast ...
    Using microarray analysis to evaluate our previously established 70-gene prognosis profile, we classified a series of 295 consecutive patients with primary ...<|control11|><|separator|>
  60. [60]
    Use of 70-gene signature to predict prognosis of patients with node ...
    A microarray-based 70-gene prognosis signature might improve the selection of patients with node-negative breast cancer for adjuvant systemic treatment.
  61. [61]
    Autoantigen microarrays for multiplex characterization of ... - PubMed
    Autoantigen microarrays represent a powerful tool to study the specificity and pathogenesis of autoantibody responses, and to identify and define relevant ...
  62. [62]
    Autoantigen Microarray for High-Throughput Autoantibody Profiling ...
    Autoantigen microarrays provide a unique tool to profile autoantibodies in systemic autoimmune diseases. SLE is an extremely-heterogeneous autoimmune disorder.
  63. [63]
    [PDF] Review Protein arrays for autoantibody profiling and fine-specificity ...
    As will be discussed below, protein arrays provide a powerful approach to profile autoantibody responses in autoimmune disease for autoantigen discovery.
  64. [64]
    Detection and Quantification of Cytokines and Other Biomarkers - PMC
    Accurate measurement of cytokine concentrations is a powerful and essential approach to the study of inflammation. The enzyme-linked immunosorbent assay (ELISA) ...
  65. [65]
    Integrated Amplification Microarrays for Infectious Disease Diagnostics
    This overview describes microarray-based tests that combine solution-phase amplification chemistry and microarray hybridization within a single microfluidic ...1. Introduction · 3. Integrated Biochemistry... · 3.1. One-Step Rt-Pcr...<|control11|><|separator|>
  66. [66]
    Protein Microarrays for Personalized Medicine - PMC - NIH
    This review focuses on protein microarray applications for biomarker discovery and validation, disease diagnosis, and use within the area of personalized ...
  67. [67]
    Precision medicine: from pharmacogenomics to pharmacoproteomics
    Sep 26, 2016 · In this article, we discuss the current status of pharmacogenomics in precision medicine and highlight the needs for concordant analysis at the proteome and ...Pharmacogenomics · Pharmacoproteomics · Integration Of...
  68. [68]
  69. [69]
    Analysis of microarray experiments of gene expression profiling - PMC
    This article describes the types of studies that can be conducted with microarray experiments (class comparison, class prediction, class discovery).Data Preprocessing · Class Comparison Studies · Gene Selection Methods
  70. [70]
    Statistical tests for differential expression in cDNA microarray ...
    Here, we review some widely used methods for testing differential expression among conditions. For these purposes, we assume that the data to be used are of ...
  71. [71]
    Fold change and p-value cutoffs significantly alter microarray ...
    Mar 13, 2012 · Arbitrary fold change (FC) cut-offs of >2 and significance p-values of <0.02 lead data collection to look only at genes which vary wildly amongst other genes.
  72. [72]
    Microarrays and Fees | Iowa Institute of Human Genetics
    (Prices effective March 1, 2025) ; $412. $512 (low quality or FFPE). 8, >935K, - ; $284, 12, >285K, -.
  73. [73]
    Increased measurement accuracy for sequence-verified microarray ...
    The consistently significant increase in the correlations derived from verified probe sets indicated that probe accuracy affects data reproducibility. Open ...Microarray Data Sets And... · Replicate Precision Of... · Accuracy Of Verified Probe...
  74. [74]
    RNA-Seq vs Microarrays | Compare technologies - Illumina
    Wider dynamic range: With array hybridization technology, gene expression measurement is limited by background at the low end and signal saturation at the high ...
  75. [75]
    Microarray & NGS: RNA QA Guidelines - University at Albany
    For each mRNA sample, please send at least 5 µg at a concentration of 0.5 - 2 µg/µL. For each total RNA sample, send 12 µg at a concentration 1 µg/µL. Users ...
  76. [76]
    Facing Current Quantification Challenges in Protein Microarrays - NIH
    Peptide microarrays have the advantage of being more stable than protein microarrays. The content can be adapted to any scientific question. Additionally ...2. Different Formats Of... · 3. Validation Of... · 4. Blocking
  77. [77]
    Recent development of gold nanochips in biosensing and ...
    Jan 1, 2025 · This paper summarized the sensitivity enhancement strategies of optical biosensing techniques based on the three main optical properties of gold nanomaterials.
  78. [78]
    Rapid signal enhancement method for nanoprobe-based biosensing
    Jul 28, 2017 · The novel method presented here is shown to enhance the signal of gold nanoparticles enabling visual detection of a spot containing <10 nanoparticles.Methods · Aunps And Agnps Synthesis · Immunocap Isac Sige 112 Gold...<|separator|>
  79. [79]
    Microarray Market Size And Share | Industry Report, 2030
    The global microarray market size was estimated at USD 6,221.2 million in 2024 and is projected to reach USD 8,463.7 million by 2030, growing at a CAGR of 5.48 ...Missing: 8.5 credible
  80. [80]
    CovidArray: A Microarray-Based Assay with High Sensitivity for ... - NIH
    We propose an approach to detect SARS-CoV-2 endowed with high sensitivity, accuracy, and multiplexing capability based on microarray technology, named ...
  81. [81]
    Comparative analysis of hybrid-SNP microarray and nanopore ... - NIH
    Jul 23, 2025 · In this work, we used nanopore sequencing technology to sequence 2 human cell lines at low depth of coverage to call copy number variations (CNV) ...Missing: papers | Show results with:papers
  82. [82]
    Microwell array chip-based single-cell analysis - RSC Publishing
    Recent advances of single-cell analysis based on microwell array chips are summarized. We first introduce the design and preparation of different microwell ...Missing: advancements | Show results with:advancements
  83. [83]
    Optimization of signal-to-noise ratio for efficient microarray probe ...
    Oct 24, 2025 · Optimization of signal-to-noise ratio for efficient microarray probe design ... Artificial intelligence, trained via machine learning or ...Missing: driven | Show results with:driven
  84. [84]
    Portable Fluorescence Microarray Reader-Enabled Biomarker ...
    This study introduces an LED-based fluorescence reader designed for POC applications, enabling multiplex detection of lupus nephritis (LN) biomarkers using a ...2. Materials And Methods · 3. Results · 4. Discussion