Fact-checked by Grok 2 weeks ago

Gene expression profiling

Gene expression profiling is a technique that simultaneously measures the expression levels of thousands of genes in a given biological sample, primarily by quantifying the abundance of (mRNA) transcripts. This method generates a comprehensive snapshot of the —the complete set of molecules—enabling the identification of patterns associated with specific cellular states, developmental stages, environmental responses, or pathological conditions. The primary technologies for expression profiling have evolved significantly since the mid-1990s. Early approaches relied on DNA microarrays, which use immobilized or cDNA probes on a to hybridize with labeled targets, allowing quantification through signal intensity measurements limited to known gene sequences. More recently, RNA sequencing (RNA-seq) has become the dominant method, involving the conversion of to (cDNA), fragmentation, and high-throughput sequencing to count RNA-derived reads, offering advantages in detecting novel transcripts, , and low-abundance genes without prior sequence knowledge. Other techniques, such as digital molecular barcoding (e.g., NanoString nCounter), provide targeted quantification but are less comprehensive. typically involves to account for technical variations, followed by statistical methods to identify differentially expressed genes and cluster patterns. In research and medicine, gene expression profiling has transformative applications across diverse fields. In , it enables tumor classification, prognosis prediction, and identification of therapeutic targets, such as distinguishing subtypes of (AML) or (JIA). In , it supports tissue-specific target validation—revealing, for instance, epididymis-enriched genes in mice—and toxicogenomics to predict side effects by comparing profiles against databases like DrugMatrix, which catalogs responses to over 600 compounds. applications further personalize treatments by linking expression variations, such as those in enzymes like , to drug efficacy and adverse reactions. Despite its power, gene expression profiling faces challenges that impact reliability and interpretation. Batch effects from experimental variations can confound results, while assumptions of uniform extraction efficiency may overlook transcriptional amplification in certain cells, necessitating spike-in controls for accurate quantification. Reproducibility issues and the high cost of , particularly for large-scale studies, remain barriers, though public repositories like NCBI's ()—housing over 6.5 million samples (as of 2024)—facilitate data sharing and validation. Ongoing advancements aim to integrate profiling with multi-omics data for deeper biological insights.

Fundamentals

Definition and Principles

Gene expression profiling is the simultaneous measurement of the expression levels of multiple or all genes within a biological sample, typically achieved by quantifying the abundance of (mRNA) transcripts, to produce a comprehensive profile representing the under defined conditions. This technique captures the dynamic activity of genes, allowing researchers to assess how cellular states, environmental stimuli, or processes alter transcriptional output across the genome. The resulting profile provides a snapshot of gene activity, highlighting patterns that reflect biological function and regulation. The foundational principles of gene expression profiling stem from the central dogma of molecular biology, which outlines the unidirectional flow of genetic information from DNA to RNA via transcription, followed by translation into proteins. Transcription serves as the primary regulatory checkpoint, where external signals modulate the initiation and rate of mRNA synthesis, making it a focal point for profiling efforts. Quantitatively, profiling measures expression as relative or absolute mRNA levels, often expressed in terms of fold changes, to distinguish qualitative differences in gene activation or repression between samples. Central to this approach are key concepts such as the , which comprises the complete set of molecules transcribed from the at a specific time, and differential expression, referring to statistically significant variations in activity across conditions or types. Normalization of data typically relies on housekeeping genes—constitutively expressed genes like GAPDH or ACTB that maintain levels—to correct for technical biases in measurement. Although mRNA abundance approximates protein production by indicating transcriptional output, this correlation is imperfect due to post-transcriptional controls, including mRNA stability and translational efficiency, which can decouple transcript levels from final protein amounts. As an illustrative example, gene expression profiling of immune cells during bacterial infection often detects upregulation of genes encoding cytokines and , such as those in the pathway, thereby revealing the molecular basis of the host's defensive response.

Historical Development

The foundations of gene expression profiling trace back to low-throughput techniques developed in the late 1970s, such as Northern blotting, which enabled the detection and quantification of specific transcripts by hybridizing labeled probes to electrophoretically separated samples transferred to a membrane. This method, introduced by Alwine et al. in 1977, laid the groundwork for measuring mRNA abundance but was limited to analyzing one or a few genes per experiment due to its labor-intensive nature. By the mid-1990s, advancements like (SAGE), developed by Velculescu et al., marked a shift toward higher-throughput profiling by generating short sequence tags from expressed genes, allowing simultaneous analysis of thousands of transcripts via . The microarray era began in 1995 with the invention of complementary DNA (cDNA) microarrays by Schena, Shalon, and colleagues under Patrick Brown at Stanford University, enabling parallel hybridization-based measurement of thousands of gene expressions on glass slides printed with DNA probes. Commercialization accelerated in 1996 when Affymetrix released the GeneChip platform, featuring high-density oligonucleotide arrays for genome-wide expression monitoring, as demonstrated in early applications like Lockhart et al.'s work on hybridization to arrays. Microarrays gained widespread adoption during the 2000s, playing a key role in the Human Genome Project's functional annotation efforts and enabling large-scale studies, such as Golub et al.'s 1999 demonstration of cancer subclassification using gene expression patterns from acute leukemias. The advent of next-generation sequencing (NGS) around 2005, exemplified by the 454 platform, revolutionized profiling by shifting from hybridization to direct sequencing of cDNA fragments, drastically increasing throughput and reducing biases. emerged as a cornerstone in 2008 with Mortazavi et al.'s method for mapping and quantifying mammalian transcriptomes through deep sequencing, providing unbiased detection of novel transcripts and precise abundance measurements. By the , NGS costs plummeted—from millions per genome in the early 2000s to approximately $50–$200 per sample for as of 2024, trending under $100 by 2025—driving a transition to sequencing-based methods over microarrays for most applications. In the 2010s, single-cell (scRNA-Seq) advanced to individual cells, with early protocols like et al. in 2009 evolving into scalable droplet-based systems such as Drop-seq in 2015 by Macosko et al., enabling profiling of thousands of cells to uncover cellular heterogeneity. further integrated positional data, highlighted by ' Visium platform launched in 2019, which captures on sections at near-single-cell . Into the , integration of has enhanced pattern detection in expression data, as seen in models like GET (2025) that simulate and predict dynamics from sequencing inputs to identify disease-associated regulatory networks.

Techniques

Microarray-Based Methods

Microarray-based methods for gene expression profiling rely on the hybridization of labeled nucleic acids to immobilized DNA probes on a solid substrate, enabling the simultaneous measurement of expression levels for thousands of genes. In this approach, short DNA sequences known as probes, complementary to target genes of interest, are fixed to a chip or slide. Total RNA or mRNA from the sample is reverse-transcribed into complementary DNA (cDNA), labeled with fluorescent dyes, and allowed to hybridize to the probes. The intensity of fluorescence at each probe location, detected via laser scanning, quantifies the abundance of corresponding transcripts, providing a snapshot of gene expression patterns. Two primary types of microarrays are used: cDNA microarrays and microarrays. cDNA microarrays typically employ longer probes (500–1,000 base pairs) derived from cloned cDNA fragments, which are spotted onto the array surface using robotic printing; these often operate in a two-color format, where samples from two conditions (e.g., control and treatment) are labeled with distinct dyes like Cy3 (green) and Cy5 (red) and hybridized to the same array for direct ratio-based comparisons. In contrast, microarrays use shorter synthetic probes (25–60 mers), either spotted or synthesized ; prominent examples include the GeneChip, which features photolithographic synthesis of one-color arrays with multiple probes per gene for mismatch controls to enhance specificity, and Illumina BeadChips, which attach oligonucleotides to microbeads in wells for high-density, one-color detection. NimbleGen arrays represent a variant of microarrays using maskless for flexible, high-density probe synthesis, supporting both one- and two-color formats. Spotted arrays (common for cDNA) offer flexibility in custom probe selection but may suffer from variability in spotting, while synthesized arrays provide uniformity and higher probe densities, up to 1.4 million probe sets (comprising over 5 million probes) on platforms like the Exon 1.0 ST array. The standard workflow begins with RNA extraction from cells or tissues, followed by isolation of mRNA and reverse transcription to generate first-strand cDNA. This cDNA is then labeled—using Cy3 and Cy5 for two-color arrays or a single dye like for one-color systems—and hybridized to the overnight under controlled temperature and stringency conditions to allow specific binding. Post-hybridization, unbound material is washed away, and the array is scanned with a to measure intensities at each probe spot, yielding raw data as pixel intensity values that reflect transcript abundance. These methods achieved peak adoption in the for high-throughput profiling of known s, offering cost-effective analysis for targeted gene panels, but have become niche with the rise of sequencing technologies due to limitations like probe cross-hybridization, which can lead to false positives from non-specific binding, and an inability to detect novel or low-abundance transcripts beyond the fixed probe set. Compared to sequencing, microarrays exhibit lower , typically spanning 3–4 orders of magnitude in detection sensitivity. Invented in , this technology revolutionized expression analysis by enabling genome-scale studies.

Sequencing-Based Methods

Sequencing-based methods for gene expression profiling primarily rely on RNA sequencing (RNA-Seq), which enables comprehensive, unbiased measurement of the transcriptome by directly sequencing RNA molecules or their complementary DNA derivatives. Introduced as a transformative approach in the late 2000s, RNA-Seq has become the gold standard for transcriptomics since the 2010s, surpassing microarray techniques due to its ability to detect novel transcripts without prior knowledge of gene sequences. The core mechanism begins with RNA extraction from cells or tissues, followed by fragmentation to generate shorter pieces suitable for sequencing. These fragments are then reverse-transcribed into complementary DNA (cDNA), which undergoes library preparation involving end repair, adapter ligation, and amplification to create a sequencing-ready library. Next-generation sequencing (NGS) platforms, such as Illumina's short-read systems, are commonly used to sequence these libraries, producing millions of reads that represent the original population. The resulting , typically output as FASTQ files containing raw sequence reads, require alignment to a using tools like or HISAT2 to map reads accurately, accounting for splicing events. Quantification occurs by counting aligned reads per or transcript, often via featureCounts or , yielding digital expression measures in the form of read counts. A key step in library preparation is mRNA enrichment, either through poly-A selection for eukaryotic polyadenylated transcripts or (rRNA) depletion to capture non-coding and prokaryotic RNAs, ensuring comprehensive coverage. Sequencing depth for samples generally ranges from to million reads per sample to achieve robust detection of expressed genes, with higher depths for low-input or complex analyses. Bulk RNA-Seq represents the standard variant, aggregating expression from millions of cells to provide an average profile suitable for population-level studies. Single-cell RNA-Seq (scRNA-Seq) extends this to individual cells, enabling dissection of cellular heterogeneity; droplet-based methods like those from , commercialized around 2016, fueled an explosion in scRNA-Seq applications post-2016 by allowing high-throughput profiling of thousands to tens of thousands of cells per run. Long-read sequencing technologies, such as PacBio's Iso-Seq, offer full-length transcript coverage, excelling in isoform resolution and detection without the need for computational assembly of short reads. Spatial RNA-Seq variants, including ' Visium platform introduced in 2020 and building on earlier from 2016, preserve tissue architecture by capturing transcripts on spatially barcoded arrays, mapping expression to specific locations within samples. These methods provide key advantages, including the discovery of novel transcripts, precise quantification of , and sensitive detection of low-abundance genes, which microarrays cannot achieve due to reliance on predefined probes. RNA-Seq exhibits a dynamic range exceeding 10^5-fold, far surpassing the ~10^3-fold of arrays, allowing accurate measurement across expression levels from rare transcripts to highly abundant ones. By 2025, costs for bulk have declined to under $200 per sample, including library preparation and sequencing, driven by advances in and platform efficiency. In precision medicine, RNA-Seq variants like scRNA-Seq are increasingly applied to resolve tumor heterogeneity, informing personalized therapies by revealing subclonal variations and therapeutic responses as of 2025.

Other Techniques

Other methods for gene expression profiling include digital molecular barcoding approaches, such as the NanoString nCounter system, which uses color-coded barcoded probes to directly hybridize with target molecules without amplification or sequencing. This technique enables targeted quantification of up to 1,000 genes per sample with high precision and reproducibility, particularly useful for clinical diagnostics and validation studies due to its low technical variability and ability to handle degraded . Unlike microarrays, NanoString provides counts rather than analog signals, reducing , though it is limited to predefined gene panels and less comprehensive than .

Data Acquisition and Preprocessing

Experimental Design

The experimental design phase of gene expression profiling begins with clearly defining the biological question to guide all subsequent decisions, such as investigating the effect of a treatment on in specific cell types or tissues. This involves specifying the , such as detecting differential expression due to a or disease state, to ensure the experiment addresses targeted objectives rather than exploratory aims. For instance, questions focused on treatment effects might prioritize controlled perturbations like siRNA knockdown or pharmacological interventions, while those involving disease modeling could use patient-derived samples. Adhering to established guidelines, such as the Minimum Information About a Experiment (MIAME) introduced in 2001, ensures comprehensive documentation of experimental parameters for reproducibility, with updates extending to sequencing-based methods via MINSEQE. Sample selection and preparation are critical, encompassing choices like cell lines for studies, animal tissues for preclinical models, or biopsies for clinical relevance. Biological replicates, derived from independent sources (e.g., different animals or patients), are essential to capture variability, with a minimum of three recommended per group to enable , though six or more enhance power for detecting subtle changes. Technical replicates, which assess consistency, should supplement but not replace biological ones. To mitigate systematic biases, of sample processing order is employed to avoid batch effects, where unintended variations from equipment or timing confound results. Controls include reference samples (e.g., untreated baselines) and exogenous spike-ins like ERCC controls for , which provide standardized benchmarks for normalization and sensitivity assessment across experiments. Sample size determination relies on power analysis to detect desired fold changes (e.g., 1.5- to 2-fold) with adequate statistical power (typically 80-90%), factoring in expected variability and sequencing depth; tools like RNAseqPS facilitate this by simulating or negative binomial distributions for data. For experiments, similar calculations apply but emphasize probe hybridization efficiency. Platform selection weighs for cost-effective, targeted profiling of known genes against for unbiased, comprehensive coverage, including low-abundance transcripts and isoforms, though incurs higher costs and requires deeper sequencing for rare events. High-throughput formats, such as 96-well plates for single-cell , support scaled designs but demand careful optimization. When human samples are involved, ethical oversight via (IRB) approval is mandatory to ensure , privacy protection, and minimal risk. Best practices from projects like in the 2010s emphasize these elements for robust, reproducible designs.

Normalization and Quality Control

Normalization and quality control are essential initial steps in processing data to ensure reliability and comparability across samples. addresses technical variations such as differences in starting amounts, library preparation efficiencies, and sequencing depths, while identifies and mitigates artifacts like low-quality reads or outliers that could skew downstream analyses. These processes aim to remove systematic biases without altering biological signals, enabling accurate quantification of levels. For microarray-based methods, is a widely adopted technique that adjusts probe intensities so that the of values across arrays matches a reference , typically the empirical of all samples. This assumes that most genes are not differentially expressed and equalizes the rank-order statistics between arrays, effectively correcting for global shifts and scaling differences. Introduced by Bolstad et al., has become standard in tools like the limma package for preprocessing and other oligonucleotide arrays. In sequencing-based methods like RNA-seq, normalization accounts for both sequencing depth (library size) and gene length biases to produce comparable expression estimates. Common metrics include reads per kilobase of transcript per million mapped reads (RPKM), fragments per kilobase of transcript per million mapped reads (FPKM) for paired-end data, and transcripts per million (TPM), which scales RPKM to sum to 1 million across genes for better cross-sample comparability. TPM is calculated as: \text{TPM}_{i} = \frac{ \text{reads mapped to gene } i / \text{gene length in kb} }{ \sum_{j} ( \text{reads mapped to gene } j / \text{gene length in kb} ) } \times 1,000,000 This formulation ensures length- and depth-normalized values that are additive across transcripts. For count-based differential analysis, methods like the median-of-ratios approach in DESeq2 estimate size factors by dividing each gene's counts by its across samples, then taking the of these ratios as the factor. Quality control begins with assessing RNA integrity using the RNA Integrity Number (RIN), an automated metric derived from analysis that scores total RNA from 1 (degraded) to 10 (intact), with values above 7 generally recommended for reliable gene expression profiling. For sequencing data, tools like FastQC evaluate raw reads for per-base quality scores, adapter contamination, overrepresented sequences, and GC content bias, flagging issues that necessitate trimming or filtering. Post-alignment, (PCA) plots visualize sample clustering to detect outliers, while saturation curves assess sequencing depth adequacy by plotting unique reads against total reads. Low-quality reads are typically removed using thresholds such as Phred scores below 20. Batch effects, arising from technical variables like different experimental runs or reagent lots, can confound biological interpretations and are detected via or surrogate variable analysis showing non-biological clustering. The method corrects these using an empirical Bayes framework that adjusts expression values while preserving biological variance, modeling batch as a covariate in a parametric or non-parametric manner. Spike-in controls, such as External RNA Controls (ERCC) mixes added at known concentrations, facilitate absolute quantification and validation of by providing an independent scale for technical performance assessment. Common pitfalls include ignoring 3' bias in poly-A selected , where reverse transcription from oligo-dT primers favors reads near the poly-A tail, leading to uneven coverage and distorted expression estimates for genes with varying 3' UTR lengths. Replicates from experimental design aid in robust by allowing variance estimation during detection. Software packages like edgeR (using trimmed mean of M-values, TMM, ) and limma (with voom for count data) integrate these preprocessing steps seamlessly before differential expression analysis.

Analysis Methods

Differential Expression Analysis

Differential expression analysis identifies genes whose expression levels differ significantly between experimental conditions, such as treated versus control samples or disease versus healthy states, by comparing normalized expression values across groups. The core metric is the log2 fold change (log2FC), which quantifies the magnitude of change on a , where a log2FC of 1 indicates a twofold upregulation and -1 a twofold downregulation. is assessed using tests to compute p-values, which are then adjusted for multiple testing across thousands of genes to control the (FDR) via methods like the Benjamini-Hochberg procedure. This approach assumes that expression data have been preprocessed through to account for technical variations. For microarray data, which produce continuous intensity values, standard parametric tests like the t-test are commonly applied, though moderated versions improve reliability by borrowing information across genes. The limma package implements linear models with empirical Bayes moderation of t-statistics, enhancing power for detecting differences in small sample sizes. In contrast, RNA-Seq data consist of discrete read counts following a to model biological variability and , where variance exceeds the mean. Tools like DESeq2 fit generalized linear models assuming variance = mean + α × mean², with shrinkage estimation for dispersions and fold changes to stabilize estimates for low-count genes. Similarly, edgeR employs to estimate common and tagwise dispersions, enabling robust testing even with limited replicates. Genes are typically selected as differentially expressed using thresholds such as |log2FC| > 1 and FDR < 0.05, balancing biological relevance and statistical confidence. Results are often visualized in , scatter plots of against -log10(p-value), where points above significance cutoffs highlight differentially expressed genes. For , zero counts—common due to low expression or technical dropout—are handled by adding small pseudocounts (e.g., 1) before log transformation for fold change calculation, preventing undefined values, though testing models like avoid pseudocounts in likelihood-based inference. Power to detect changes depends on sample size, sequencing depth, and effect size; for instance, detecting a twofold change ( = 1) at 80% power and FDR < 0.05 often requires at least three replicates per group for moderately expressed genes. In practice, this analysis has revealed upregulated genes in cancer tissues compared to normal, such as proliferation markers like in breast tumors, aiding molecular classification. Early microarray studies on acute leukemias identified sets of upregulated oncogenes distinguishing subtypes, demonstrating the method's utility in biomarker discovery.

Statistical and Computational Tools

Gene expression profiling generates high-dimensional datasets where the number of genes often exceeds the number of samples, necessitating advanced statistical and computational tools to uncover patterns and make predictions. Unsupervised learning methods, such as clustering and dimensionality reduction, are fundamental for exploring inherent structures in these data without predefined labels. Clustering algorithms group genes or samples based on similarity in expression profiles; for instance, hierarchical clustering builds a tree-like structure to reveal nested relationships, while k-means partitioning assigns data points to a fixed number of clusters by minimizing intra-cluster variance. These techniques have been pivotal since the late 1990s, enabling the identification of co-expressed gene modules and sample subtypes in microarray experiments. Dimensionality reduction complements clustering by projecting high-dimensional data into lower-dimensional spaces to mitigate noise and enhance visualization. Principal Component Analysis (PCA) achieves this linearly by identifying directions of maximum variance, commonly used as a preprocessing step in gene expression workflows to retain the top principal components that capture most variability. Nonlinear methods like t-distributed Stochastic Neighbor Embedding (t-SNE) preserve local structures for visualizing clusters in two or three dimensions, particularly effective for single-cell RNA-seq data where it reveals cell type separations. Uniform Manifold Approximation and Projection (UMAP) offers a faster alternative to t-SNE, balancing local and global data structures while scaling better to large datasets, as demonstrated in comparative evaluations of transcriptomic analyses. Supervised learning methods leverage labeled data to train models for classification or regression tasks in gene expression profiling. Support Vector Machines (SVMs) construct hyperplanes to separate classes with maximum margins, proving robust for phenotype prediction from expression profiles, such as distinguishing cancer subtypes, through efficient handling of high-dimensional inputs via kernel tricks. Random Forests, an ensemble of decision trees, aggregate predictions to reduce overfitting and provide variable importance rankings, widely applied in genomic classification for tasks like tumor identification with high accuracy on microarray data. Regression variants, including ridge or lasso, predict continuous traits like drug response by penalizing coefficients to address multicollinearity in expression matrices. Advanced techniques extend these approaches to infer complex relationships and enhance interpretability. Weighted Gene Co-expression Network Analysis (WGCNA) constructs scale-free networks from pairwise gene correlations, using soft thresholding to identify modules of co-expressed genes that correlate with traits, as formalized in its foundational framework for microarray and RNA-seq data. For machine learning models in the 2020s, SHapley Additive exPlanations (SHAP) quantifies feature contributions to predictions, aiding interpretability in genomic applications like variant effect scoring by attributing importance to specific genes or interactions. Software ecosystems facilitate implementation of these tools. In R, Bioconductor packages like support clustering and downstream exploration of gene groups, integrating statistical tests for profile comparisons. Python's toolkit streamlines single-cell RNA-seq analysis, incorporating UMAP, Leiden clustering, and batch correction for scalable processing of millions of cells. High-dimensionality poses the "curse of dimensionality," where sparse data leads to overfitting and unreliable distances; mitigation strategies include feature selection to retain informative genes and embedding into lower dimensions via PCA or autoencoders before modeling. Recent advancements in gene pair methods, focusing on ratios or differences between expression levels of gene pairs, have improved biomarker discovery by reducing dimensionality while preserving relational information, as demonstrated in a 2025 review of gene pair methods in clinical research advancing precision medicine and in 2025 studies applying them to cancer subtyping. These approaches yield robust signatures with fewer features than single-gene models, enhancing predictive power in heterogeneous datasets. As of 2025, integration of deep learning models, such as graph neural networks for co-expression analysis, has further advanced pattern detection in large-scale transcriptomic data.

Functional Annotation and Pathway Analysis

Functional annotation involves mapping differentially expressed genes to known biological functions, processes, and components using standardized ontologies and databases. The Gene Ontology (GO) consortium provides a structured vocabulary for annotating genes across three domains: molecular function, biological process, and cellular component, enabling systematic classification of gene products. Databases such as integrate GO terms with protein sequence and functional data, while and offer comprehensive gene annotations derived from experimental evidence, computational predictions, and literature curation. In RNA-Seq profiling, handling transcript isoforms is crucial, as multiple isoforms per gene can contribute to expression variability; tools often aggregate isoform-level counts to gene-level summaries or use isoform-specific quantification to avoid underestimating functional diversity. Tools like DAVID and g:Profiler facilitate ontology assignment by integrating multiple annotation sources for high-throughput analysis of gene lists. DAVID clusters functionally related genes and terms into biological modules, supporting GO enrichment alongside other annotations from over 40 databases. g:Profiler performs functional profiling by mapping genes to GO terms, pathways, and regulatory motifs, with support for over 500 organisms and regular updates from Ensembl. These tools assign annotations based on evidence codes, prioritizing experimentally validated terms to ensure reliability in interpreting expression profiles. Pathway analysis extends annotation by identifying coordinated changes in biological pathways, using enrichment tests to detect over-representation of profiled genes in predefined sets. Common databases include , which maps genes to metabolic and signaling pathways, and , focusing on detailed reaction networks. Over-representation analysis (ORA) applies to lists of differentially expressed genes, employing the hypergeometric test (equivalent to ) to compute significance:
p = \sum_{i = x}^{n} \frac{\binom{n}{i} \binom{N - n}{M - i}}{\binom{N}{M}}
where N is the total number of genes, n the number in the pathway, M the number of differentially expressed genes, and x the observed overlap. This test assesses whether pathway genes are enriched beyond chance, with multiple-testing corrections like to control false positives.
Gene Set Enrichment Analysis (GSEA) complements ORA by evaluating ranked gene lists from full expression profiles, detecting subtle shifts in pathway activity without arbitrary significance cutoffs. GSEA uses a Kolmogorov-Smirnov-like statistic to measure enrichment at the top or bottom of the ranking, weighted by gene metric, and permutes phenotypes to estimate empirical p-values. For example, in cancer studies, GSEA has revealed upregulation of the , linking altered expression of genes like and to tumor proliferation and survival. Regulated genes are often categorized by function to highlight regulatory mechanisms, such as grouping into transcription factors (e.g., E2F family regulating cell cycle and apoptosis genes) or apoptosis-related sets (e.g., BCL2 family modulators). This categorization integrates annotations to infer upstream regulators and downstream effects, aiding in the interpretation of co-regulated patterns in expression profiles. As of 2025, advancements in pathway analysis include AI-driven tools for dynamic pathway modeling, enhancing predictions of pathway perturbations in disease contexts.

Applications

Basic Research and Hypothesis Testing

Gene expression profiling serves as a cornerstone in basic research by enabling the systematic analysis of genome-wide transcription patterns to uncover the molecular underpinnings of biological processes. In functional genomics, it has been instrumental in identifying genes responsive to environmental cues or developmental stages, such as the profiling of abscisic acid-regulated genes in Arabidopsis thaliana, which revealed key regulators of stress responses. Similarly, in developmental biology, microarray analysis of Drosophila melanogaster during metamorphosis highlighted temporal gene expression waves coordinating tissue remodeling, providing insights into conserved regulatory mechanisms across species. These applications allow researchers to map co-expression networks, as demonstrated by early clustering methods that grouped functionally related genes in yeast, facilitating the discovery of operon-like structures in eukaryotes. In hypothesis testing, gene expression profiling supports both generation and validation of biological hypotheses by quantifying differential expression under controlled perturbations. For instance, significance analysis of microarrays (SAM) has been widely adopted to test hypotheses about cellular responses to stressors, such as in human fibroblasts, where it identified reproducible gene signatures for DNA damage pathways with controlled false discovery rates. This approach extends to model organisms, where profiling mouse brain tissues across genetic strains tested hypotheses on polygenic traits, revealing pleiotropic networks modulating nervous system function and behavior. By integrating expression data with phenotypic variation, such studies prioritize candidate genes for follow-up experiments, enhancing the efficiency of hypothesis-driven research in complex traits like addiction or . Beyond individual experiments, profiling aids in constructing gene co-expression networks to test hypotheses about regulatory interactions in basic research. Weighted gene co-expression network analysis (WGCNA), for example, has been applied to dissect modules perturbed in schizophrenia, hypothesizing synaptic dysfunction as a core mechanism based on hub gene disruptions in postmortem brain samples. In ethanol response studies, network topology analysis in mouse prefrontal cortex validated hypotheses on neuroadaptive pathways, linking expression modules to behavioral tolerance. These methods emphasize scale-free network properties, where highly connected genes often represent key regulators, guiding targeted validations like knockdown experiments to confirm causal roles. Overall, such profiling strategies have transformed basic research by bridging transcriptomics with systems biology, prioritizing high-impact discoveries over exhaustive listings.

Clinical and Diagnostic Uses

Gene expression profiling plays a pivotal role in clinical diagnostics by facilitating the molecular subtyping of diseases, allowing for more precise disease classification and personalized therapeutic strategies. In breast cancer, the , introduced in the late 2000s, analyzes the expression of 50 genes to categorize tumors into intrinsic subtypes—Luminal A, Luminal B, HER2-enriched, and basal-like—which informs prognosis and treatment selection beyond traditional histopathology. Similarly, the evaluates a 21-gene panel to generate a recurrence score for early-stage, hormone receptor-positive, HER2-negative breast cancer, helping clinicians decide on the necessity of adjuvant chemotherapy. These biomarker panels derived from gene expression profiles have become integral to diagnostic workflows, reducing overtreatment while identifying high-risk patients. In prognostics, gene expression signatures enable risk stratification and pharmacogenomic predictions to guide treatment outcomes. For acute lymphoblastic leukemia (ALL), multigene signatures, such as those involving , , and others, have been identified to predict relapse risk and overall survival, allowing for intensified therapy in high-risk subgroups. In pharmacogenomics, expression profiles predict chemotherapy responses; for example, models integrating gene expression data have shown utility in forecasting sensitivity to agents like doxorubicin in breast cancer, supporting personalized dosing and combination regimens. The , FDA-cleared in 2007 as the first gene expression-based prognostic test for breast cancer, uses a 70-gene signature to assess distant metastasis risk in early-stage node-negative patients, influencing decisions on systemic therapy. Therapeutic applications include companion diagnostics and treatment monitoring. HER2 gene expression levels, often assessed via profiling, serve as a companion diagnostic for targeted therapies like trastuzumab in HER2-positive breast cancer, with overexpression indicating eligibility for antibody-drug conjugates. Single-cell RNA sequencing (scRNA-seq) has advanced monitoring of minimal residual disease (MRD), detecting low-level cancer cells post-treatment in leukemias and solid tumors to guide relapse prevention strategies. By 2025, advancements in liquid biopsy-based RNA profiling, such as nanopore sequencing of circulating tumor RNA, have enhanced non-invasive diagnostics for early detection and monitoring in cancers like lung and colorectal, improving accessibility over tissue biopsies. During the COVID-19 pandemic in the 2020s, gene expression profiling elucidated host immune responses, identifying signatures of interferon-stimulated genes and cytokine dysregulation that correlated with disease severity and guided immunomodulatory therapies. Despite these successes, challenges persist in clinical translation, including reproducibility across cohorts due to variability in sample processing and platform differences, necessitating standardized protocols for robust multi-center validation.

Comparisons with Other Approaches

Relation to Proteomics

Gene expression profiling (GEP) measures mRNA transcript levels, providing insights into transcriptional activity, but exhibits poor correlation with actual protein abundances, typically quantified by Spearman's rank correlation coefficients ranging from 0.4 to 0.6 across large-scale studies. This discrepancy arises primarily from extensive post-transcriptional regulation, including -mediated repression of translation and mRNA degradation, which can suppress protein synthesis despite elevated transcript levels. Seminal work by Vogel et al. (2010) in a human cell line demonstrated that mRNA concentration alone explains approximately 25-30% of protein abundance variation (Spearman's rho = 0.46), with sequence features and post-transcriptional factors accounting for much of the remainder, highlighting concordance below 50% for direct mRNA-protein mapping. A combined model incorporating mRNA levels and sequence signatures explains about two-thirds of the variation. GEP and proteomics serve complementary roles in biological research, with GEP enabling rapid, high-throughput screening of thousands of transcripts to identify potential regulatory changes, while proteomics, often via , directly assesses protein levels and modifications as functional endpoints of gene expression. For instance, in cellular stress responses such as or heat shock, translation often dominates over transcription, where proteomics reveals rapid protein remodeling and post-translational adjustments that GEP overlooks, such as selective translation of stress-protective factors under inhibited global cap-dependent translation. Integration of GEP with proteomics in multi-omics studies enhances understanding by correlating transcript profiles with protein data, revealing regulatory layers like translation efficiency and degradation rates that mediate cellular phenotypes. GEP offers advantages in cost-effectiveness and scalability, allowing genome-wide analysis at lower expense than proteomics, which provides superior resolution for direct measures of protein activity, localization, and interactions but requires more complex sample preparation. Tools like reverse-phase protein arrays (RPPA) in the 2020s serve as a bridge, offering targeted, high-throughput protein quantification that aligns more closely with transcriptomic data for validation in cancer and signaling studies. A unique limitation of GEP is its focus on protein-coding mRNAs, which misses the regulatory effects of non-coding RNAs () on protein levels, such as long non-coding RNAs () that modulate translation or stability of target proteins without altering transcript abundance. This oversight can lead to incomplete models of protein regulation, underscoring the need for proteomics to capture ncRNA-driven post-transcriptional influences.

Integration with Multi-Omics

Gene expression profiling (GEP) is increasingly integrated with other omics layers, such as , , and , to provide a more comprehensive understanding of biological systems by capturing interactions across molecular levels. This multi-omics integration addresses limitations of GEP alone, such as its inability to fully explain phenotypic outcomes, by incorporating regulatory and downstream effects; for instance, combining with has been shown to enhance predictive accuracy for disease states by resolving discrepancies between mRNA levels and protein function. Such approaches enable the identification of holistic pathways and biomarkers that single-omics analyses might overlook. Key integration strategies include data fusion methods like iCluster, which performs joint clustering of multi-omics datasets using a Gaussian latent variable model to identify coherent sample or feature groups across layers such as genomics and transcriptomics. Another approach involves correlating layers through expression quantitative trait loci (eQTLs), which link single nucleotide polymorphisms (SNPs) from genomic data to variations in gene expression, thereby revealing regulatory mechanisms underlying traits. In genomics integration, eQTL analysis validates genome-wide association study (GWAS) hits by associating SNPs with expression changes, for example, the GTEx project identified over 4 million eQTLs regulating more than 23,000 genes across 49 human tissues. For epigenomics, GEP is combined with DNA methylation profiles to elucidate how epigenetic modifications influence transcription; integrative analyses have shown that methylation patterns at promoter regions correlate with gene expression levels, aiding in the discovery of disease-associated regulatory networks. In metabolomics integration, transcriptomic data complements metabolite profiles to complete pathway reconstructions, where expression changes in enzymes are mapped to metabolic flux alterations, enhancing insights into cellular responses. Prominent tools for multi-omics integration include Multi-Omics Factor Analysis (MOFA), a probabilistic factor model that decomposes variation across datasets like transcriptomics and epigenomics into shared latent factors for unsupervised discovery of principal sources of heterogeneity. Network-based methods further facilitate integration by overlaying GEP with protein-protein interaction (PPI) networks; for example, iOmicsPASS combines mRNA expression and protein data over PPI and transcription factor networks to prioritize disease-relevant pathways. The Cancer Genome Atlas (TCGA) project, launched in the 2010s, exemplifies large-scale multi-omics integration in cancer research, profiling over 11,000 primary tumor samples across genomic, transcriptomic, epigenomic, and proteomic layers to uncover molecular subtypes and therapeutic targets. As of 2025, emerging trends emphasize spatial multi-omics, combining RNA expression with protein imaging to map cellular interactions in tissue context; technologies like MultiGATE enable regulatory inference from spatially resolved transcriptomic and proteomic data, revealing tumor microenvironment dynamics.

Limitations and Challenges

Technical Limitations

Gene expression profiling techniques, such as and (), are susceptible to various technical artifacts that compromise data accuracy and reliability. These limitations arise from inherent methodological constraints, including biases in signal detection and quantification, which can lead to systematic errors in measuring transcript abundance. In microarray-based profiling, probe design introduces significant bias, as sequence-specific hybridization efficiencies vary, leading to inconsistent signal intensities for similar expression levels across genes. Additionally, saturation occurs at high expression levels due to the finite dynamic range of fluorescent signals, which compresses measurements of highly abundant transcripts and reduces sensitivity for fold-change detection beyond approximately 10^3-fold. RNA-Seq mitigates some of these issues by providing a broader dynamic range exceeding 10^5-fold, yet it still falls short of capturing extreme expression differences greater than 10^6-fold, particularly in low-abundance transcripts overwhelmed by sequencing noise. RNA-Seq introduces its own artifacts, notably PCR amplification bias during library preparation, where shorter or GC-rich fragments are preferentially amplified, skewing quantification of transcript abundances. Mapping errors further exacerbate inaccuracies, especially for paralogous genes with high sequence similarity, as short reads often align ambiguously, resulting in multi-mapped reads that are discarded or misassigned, underestimating expression in gene families. Batch effects represent a pervasive general limitation across both methods, manifesting as systematic variations from technical factors like reagent lots or processing dates, which can mimic biological signals and inflate false discovery rates in differential expression analyses. Low-input samples pose additional challenges, as RNA degradation in limited material—common in clinical or archival tissues—alters transcript profiles by preferentially losing 5' ends, biasing toward 3' sequences and reducing overall mappability. Quantification of complex transcript features is also hindered; short-read RNA-Seq under-detects alternative splicing events, accurately recapitulating only about 50% of isoforms identified by long-read methods due to insufficient read length spanning splice junctions. Standard poly(A)-selection protocols miss non-polyadenylated RNAs, such as certain long non-coding and regulatory transcripts, unless rRNA depletion is employed, which increases complexity and potential off-target biases. These technical flaws contribute to error rates in differential expression calling, with false positive rates often ranging from 1-5% even under controlled conditions, depending on method and dataset size. In single-cell RNA-Seq (), cost barriers remain substantial, with per-cell expenses estimated at $0.01–$0.50 as of November 2025 for high-depth profiling, limiting scalability for large cohorts. Mitigation strategies include experimental designs incorporating spike-in controls to calibrate batch effects and unique molecular identifiers to correct PCR biases, though these add preparatory complexity without fully eliminating artifacts.

Interpretative Challenges

One major interpretative challenge in gene expression profiling arises from the difficulty in distinguishing correlation from causation. Differentially expressed genes (DEGs) identified in profiling studies often reflect downstream effects of a disease or perturbation rather than direct causal drivers, as observational data cannot isolate mechanistic relationships from mere associations. To confirm causality, perturbation experiments—such as CRISPR-based knockouts or single-cell RNA-seq with genetic manipulations—are essential, as they enable direct testing of how altering a gene's expression impacts downstream profiles and phenotypes. Without such interventions, interpretations risk overattributing regulatory roles to correlated changes, leading to misguided hypotheses about disease mechanisms. Gene expression is highly context-dependent, varying significantly across cell types, environmental conditions, and developmental stages, which complicates the extrapolation of profiles from one setting to another. In heterogeneous samples like tumors, where diverse cell populations coexist, bulk profiling averages signals and masks subtype-specific patterns, potentially leading to incomplete or misleading insights into tumor behavior. For instance, intratumor heterogeneity can result in variable expression signatures that reflect spatial or clonal differences rather than uniform disease states, underscoring the need for single-cell or spatially resolved profiling to resolve these ambiguities. This variability emphasizes that expression profiles are not absolute but contingent on biological context, challenging the generalizability of findings across studies or patient cohorts. A critical gap in gene expression profiling lies in its focus on transcriptional levels, overlooking post-transcriptional regulation such as mRNA translation efficiency and degradation, which can profoundly alter protein output. RNA-seq and microarray data capture steady-state mRNA abundance but ignore how factors like microRNAs, RNA-binding proteins, or codon usage influence translation and mRNA stability, providing an incomplete view of regulatory networks. For example, extensive buffering at post-transcriptional steps can decouple mRNA levels from protein expression, meaning profiled changes may not translate to functional outcomes. This limitation highlights the necessity of integrating profiling with proteomics or ribosome profiling to bridge the transcript-to-protein divide and avoid erroneous assumptions about gene function. Stochastic noise inherent in gene expression further hinders accurate interpretation, as it introduces variability unrelated to deterministic biological signals. Bursty transcription—episodic bursts of mRNA production interspersed with inactive periods—generates cell-to-cell heterogeneity even in genetically identical populations, amplifying noise in profiled data and obscuring subtle regulatory effects. This intrinsic stochasticity can lead to overinterpretation of expression signatures, with reproducibility across independent studies often below 70% due to such noise confounding differential analyses. Balancing noise reduction through deeper sequencing with recognition of its biological relevance is crucial to prevent artifactual conclusions. Ethical considerations add another layer of interpretative complexity, particularly in clinical applications of gene expression profiling. Privacy risks are heightened when profiles contain identifiable genetic information, necessitating robust data anonymization to protect patient confidentiality in shared datasets or AI-driven analyses. For instance, studies as of 2024 have shown that single-cell RNA-seq datasets are vulnerable to linking attacks that can re-identify donors with high accuracy. Additionally, biases in training data for machine learning models interpreting profiles—such as underrepresentation of diverse populations—can perpetuate health inequities by producing skewed predictions that favor certain demographics. Addressing these issues requires transparent methodologies and inclusive data practices to ensure equitable and trustworthy interpretations.

Validation Strategies

Experimental Validation

Experimental validation of gene expression profiling results typically involves orthogonal, low-throughput laboratory techniques to confirm transcript abundance and functional relevance observed in high-throughput assays like microarrays or RNA sequencing. These methods provide direct molecular evidence, often targeting a subset of candidate genes identified from profiling data, and are essential for establishing reliability before clinical or biological interpretation. Common approaches include nucleic acid-based assays for RNA levels and protein-based methods for downstream effects, with functional perturbations to assess causality. Quantitative reverse transcription polymerase chain reaction (qRT-PCR) serves as the gold standard for validating gene expression changes due to its high sensitivity, specificity, and quantitative accuracy. This technique amplifies and detects specific cDNA sequences derived from RNA, using either SYBR-Green dye for non-specific fluorescence or TaqMan probes for target-specific detection during real-time monitoring. Relative quantification is commonly performed via the delta-delta Ct (ΔΔCt) method, where the fold change in expression is calculated as $2^{-\Delta\Delta C_t}, with ΔCt representing the difference in cycle threshold (Ct) values between the target gene and a reference gene, and ΔΔCt the difference between experimental and control samples normalized to the reference. Adherence to the MIQE (Minimum Information for Publication of Quantitative Real-Time PCR Experiments) guidelines, established in 2009, ensures standardized reporting of experimental design, data analysis, and quality controls to enhance reproducibility. Concordance rates between qRT-PCR and microarray results typically range from 70% to 90%, reflecting strong but not perfect agreement, particularly for genes with moderate to high expression changes. Northern blotting offers an additional RNA validation method by separating RNA by size via electrophoresis, transferring it to a membrane, and hybridizing with labeled probes to confirm transcript size and abundance. This technique, though labor-intensive, provides a direct visualization of RNA integrity and is particularly useful for validating alternative splicing or polyadenylation variants detected in profiling. In situ hybridization (ISH) extends validation to spatial contexts, using labeled nucleic acid probes to localize gene expression within tissues or cells, thereby confirming cell-type-specific patterns from bulk profiling data. At the protein level, Western blotting detects translated products by separating proteins via electrophoresis and probing with antibodies, validating whether observed transcript changes correlate with protein abundance. Immunofluorescence complements this by enabling visualization of protein localization and expression in fixed cells or tissues, often using fluorescently tagged antibodies for high-resolution imaging. These methods bridge the gap between mRNA profiling and functional outcomes, as post-transcriptional regulation can decouple transcript and protein levels. Functional assays further test causality by perturbing gene expression and observing phenotypic effects. Reporter gene constructs, where a promoter of interest drives a detectable reporter like luciferase, quantify transcriptional activity in response to stimuli. Knockdown using small interfering RNA (siRNA) or overexpression via plasmids reduces or increases target levels, respectively, while CRISPR-based editing (e.g., CRISPR interference or knockout) provides precise, stable perturbations to assess regulatory roles. For instance, in cancer research, qRT-PCR has validated microarray-identified biomarkers such as EGFR and HER2 in non-small cell lung cancer tissues, confirming their prognostic value through correlation with clinical outcomes.

Computational Validation

Computational validation of gene expression profiling involves in silico techniques to evaluate the robustness, stability, and reproducibility of results using existing datasets, without requiring additional biological experiments. These methods assess model performance, detect biases, and confirm findings across independent sources, ensuring that identified gene signatures or differential expression patterns are reliable for downstream applications like biomarker discovery. Key approaches include cross-validation for internal consistency, meta-analysis for cross-dataset comparability, and simulation for sensitivity testing, often leveraging public repositories such as the and . Cross-validation techniques, such as k-fold, leave-one-out, and bootstrap resampling, are widely used to gauge the stability of classifiers or gene signatures derived from expression data. For instance, leave-one-out cross-validation partitions the dataset by iteratively excluding one sample for testing, providing an unbiased estimate of prediction error for diagnostic models based on gene expression profiles. Bootstrap methods resample the data with replacement to quantify variability in feature selection, helping identify stable gene lists less prone to overfitting. Performance is typically evaluated using receiver operating characteristic (ROC) curves, where area under the curve (AUC) values above 0.8 indicate robust signature discrimination, as demonstrated in validations of melanoma gene expression classifiers. These approaches reveal that many initial signatures overfit training data, with cross-validated error rates often 10-20% higher than naive estimates. Reproducibility is assessed by comparing results across multiple datasets from repositories like GEO and ArrayExpress, which together host millions of gene expression studies, many compliant with minimum information standards. Meta-analysis integrates these via fixed- or random-effects models to pool effect sizes, such as log fold changes, increasing statistical power and reducing false positives; for example, random-effects models account for heterogeneity between studies, yielding more conservative yet reproducible differentially expressed gene lists in cancer transcriptomics. The intraclass correlation coefficient (ICC) quantifies reliability, with values >0.8 signifying high consistency in expression measurements across replicates or cohorts, as applied in benchmarking. Adherence to the FAIR principles (findable, accessible, interoperable, and reusable), established in 2016, is a key goal for these repositories, with ongoing enhancements as of to facilitate automated data retrieval and integration through standardized metadata. Simulation generates synthetic datasets to test method sensitivity, such as detecting fold changes under varying noise levels or dropout rates in single-cell . Tools like scDesign2 create realistic count preserving correlations and zero-inflation, enabling evaluation of expression algorithms; for instance, simulations have shown that tools like DESeq2 maintain >90% power for detecting 2-fold changes at low expression levels. Validating differentially expressed lists often involves applying the same pipeline to independent cohorts from , where overlap >50% (e.g., via ) confirms generalizability, as seen in cross-cohort verifications of inflammatory response signatures. These computational strategies complement experimental validation by providing rapid, cost-effective assessments of result trustworthiness.

References

  1. [1]
    Gene Expression Profiling - an overview | ScienceDirect Topics
    Gene expression profiling is defined as a technique that identifies RNA expression patterns and clusters them into subgroups based on parameters such as ...Regulated Cell Death Part B · 2 Gene Expression Profiling · Applications
  2. [2]
    Revisiting Global Gene Expression Analysis - PMC - NIH
    Gene expression analysis is a widely used and powerful method for investigating the transcriptional behavior of biological systems, for classifying cell states ...Global Gene Expression... · Assumptions And... · Rna-Seq Sample Preparation...
  3. [3]
    Gene Expression Profiling and its Practice in Drug Development - NIH
    In this review, the authors highlight some of the applications of expression profiling using microarray analysis in drug development. TISSUE EXPRESSION ...Tissue Expression Profiling · Side Effect Profiling · Pharmacogenomics
  4. [4]
    Gene Expression Profiling - an overview | ScienceDirect Topics
    Gene expression profiling refers to the quantification of thousands of mRNAs simultaneously to create a picture of global gene expression in the cell ...Molecular Diagnostics · Regulated Cell Death Part B · 2 Gene Expression Profiling
  5. [5]
    transcriptome | Learn Science at Scitable - Nature
    A transcriptome is the full range of messenger RNA, or mRNA, molecules expressed by an organism.
  6. [6]
    Biochemistry, Replication and Transcription - StatPearls - NCBI - NIH
    The flow of genetic information in biological systems from DNA>RNA>Protein is the central dogma in molecular biology. This explains how the genetic information ...
  7. [7]
    Central dogma at the single-molecule level in living cells - PMC
    The central dogma of molecular biology states that genetic information encoded in DNA is transcribed to mRNA by RNA polymerase, and mRNA is translated to ...
  8. [8]
    Differential gene expression analysis pipelines and bioinformatic ...
    Differential gene expression (DGE). DGE analysis is a technique used in molecular biology to compare gene expression levels between two or more sample groups, ...
  9. [9]
    Differential Gene Expression - Developmental Biology - NCBI - NIH
    Differential gene expression was shown to be the way a single genome derived from the fertilized egg could generate the hundreds of different cell types in the ...
  10. [10]
    Selection of housekeeping genes for gene expression studies in the ...
    Candidate housekeeping genes were normalised by dividing the relative expression value by the geometric mean of the expression levels of the two selected ...
  11. [11]
    Comparing protein abundance and mRNA expression levels on a ...
    We review the results of attempts to correlate protein abundance with mRNA expression levels, focusing on yeast.
  12. [12]
    Using protein turnover to expand the applications of transcriptomics
    While RNAseq is a powerful tool, it has one major caveat, in that RNA expression does not necessarily match protein abundance. RNA may be present in a cell, but ...
  13. [13]
    Distinct Gene Expression Profiling after Infection of Immature Human ...
    The expression profiles of transcription factors such as NF-κB/Rel and STAT were regulated similarly by both viruses; in contrast, OASL, MDA5, and IRIG-I ...
  14. [14]
    Serial Analysis of Gene Expression - Science
    The characteristics of an organism are determined by the genes expressed within it. A method was developed, called serial analysis of gene expression (SAGE) ...
  15. [15]
    Quantitative Monitoring of Gene Expression Patterns with ... - Science
    Microarrays prepared by high-speed robotic printing of complementary DNAs on glass were used for quantitative expression measurements of the corresponding genes ...
  16. [16]
    Molecular Classification of Cancer: Class Discovery and ... - Science
    The results demonstrate the feasibility of cancer classification based solely on gene expression monitoring and suggest a general strategy for discovering and ...
  17. [17]
    Mapping and quantifying mammalian transcriptomes by RNA-Seq
    May 30, 2008 · We have mapped and quantified mouse transcriptomes by deeply sequencing them and recording how frequently each gene is represented in the sequence sample (RNA- ...
  18. [18]
    Next-Generation Sequencing Costs: The Sub $100 Genome
    Sep 16, 2024 · This new instrument could produce a whopping 50,000 WGS per year for under $100 per 30x human genome. If Illumina had won the race to the $1,000 ...
  19. [19]
    From bulk, single-cell to spatial RNA sequencing - PMC - NIH
    Nov 15, 2021 · The first 10X visium spatial transcriptomics platform was launched in late 2019. This technology utilizes the power of classic microarrays ...Bulk Rnaseq · Single Cell Rna Sequencing... · Spatial Rna Sequencing...
  20. [20]
    Using AI To Predict Gene Expression
    Jan 16, 2025 · GET simulates gene expression, predicting where and when genes express under normal or disease conditions, using sequencing and DNA ...<|separator|>
  21. [21]
  22. [22]
  23. [23]
    Illumina Microarray Technology
    How Do Illumina Microarrays Work? As DNA fragments pass over the BeadChip, each probe binds to a complementary sequence in the sample DNA, stopping one base ...
  24. [24]
    Reproducible probe-level analysis of the Affymetrix Exon 1.0 ST ...
    To interrogate each potential exon with at least one probeset, the exon array contains about 5.6 million probes grouped into >1.4 million probesets (most ...
  25. [25]
    Microarrays, deep sequencing and the true measure of the ...
    May 31, 2011 · We conclude that microarrays remain useful and accurate tools for measuring expression levels, and RNA-Seq complements and extends microarray measurements.
  26. [26]
    Performance of a scalable RNA extraction-free transcriptome ...
    Sep 30, 2021 · The typical RNA-seq workflow starts with RNA extraction from samples, followed by cDNA synthesis, adaptor ligation, size selection and high ...Results · Pathway Analysis Reflects... · Methods<|separator|>
  27. [27]
    What is a good sequencing depth for bulk RNA-Seq?
    For that reason, many published human RNA-Seq experiments have been sequenced with a sequencing depth between 20 M - 50 M reads per sample. This gives a ...
  28. [28]
    High-throughput single-cell gene-expression profiling with ... - PNAS
    Sep 13, 2016 · With these improvements, we performed RNA profiling in more than 100,000 human cells, with as many as 40,000 cells measured in a single 18-h ...
  29. [29]
    Augmenting precision medicine via targeted RNA-Seq detection of ...
    Jun 13, 2025 · In this study, we conducted targeted RNA-seq on a reference sample set for expressed variant detection to explore its potential capability to complement DNA ...
  30. [30]
    Minimum information about a microarray experiment (MIAME)
    The ultimate goal of this work is to establish a standard for recording and reporting microarray-based gene expression data, which will in turn facilitate the ...
  31. [31]
    MIAME and MINSEQE guidelines - GEO - NCBI
    Jul 16, 2024 · The MIAME (Minimum Information About a Microarray Experiment) and MINSEQE (Minimum Information About a Next-generation Sequencing Experiment) guidelines
  32. [32]
    How many biological replicates are needed in an RNA-seq ... - NIH
    At least 12 replicates per condition for experiments where identifying the majority of all DE genes is important. For experiments with <12 replicates per ...Missing: profiling | Show results with:profiling
  33. [33]
    Synthetic spike-in standards for RNA-seq experiments - PMC
    The ERCC is working to develop and disseminate a standard set of exogenous RNA controls for use in gene expression assays. These controls, and methods that ...
  34. [34]
    Power analysis and sample size estimation for RNA-Seq differential ...
    Increasing sample size or sequencing depth increases power; however, increasing sample size is more potent than sequencing depth to increase power, especially ...
  35. [35]
    RNAseqPS: A Web Tool for Estimating Sample Size and Power for ...
    Oct 13, 2014 · We present RNAseqPS, an advanced online RNAseq power and sample size calculation tool based on the Poisson and negative binomial distributions.
  36. [36]
    Advantages of RNA-seq Compared to RNA Microarrays for ... - NIH
    RNA-seq has considerable advantages for examining transcriptome profile structure such as the detection of novel transcripts and splice junctions, although it ...
  37. [37]
    Bulk RNA-seq Data Standards and Processing Pipeline - ENCODE
    Best practices for ENCODE2 RNA-seq experiments have been outlined here. Replicate concordance: the gene level quantification should have a Spearman correlation ...Pipeline Overview · Outputs · Genomic References
  38. [38]
    Social, Legal, and Ethical Implications of Genetic Testing - NCBI - NIH
    The individuals should have a right to consent or to object to particular uses of the sample or information. Subsequent anonymous use of samples for research is ...
  39. [39]
    RNA-seq analysis is easy as 1-2-3 with limma, Glimma and edgeR
    Jun 17, 2016 · In this workflow article, we analyse RNA-sequencing data from the mouse mammary gland, demonstrating use of the popular edgeR package to import, ...
  40. [40]
    [PDF] A Comparison of Normalization Methods for High Density ...
    The paper compares three complete data methods (Cyclic loess, Contrast, Quantiles) and two baseline methods (scaling, non-linear) for normalization.
  41. [41]
    limma powers differential expression analyses for RNA-sequencing ...
    The article outlines limma's functionality at each of the main steps in a gene expression analysis, from data import, pre-processing, quality assessment and ...
  42. [42]
    Measurement of mRNA abundance using RNA-Seq data
    Aug 10, 2025 · We propose a slight modification of RPKM that eliminates this inconsistency and call it TPM for transcripts per million. TPM respects the ...<|separator|>
  43. [43]
    Moderated estimation of fold change and dispersion for RNA-seq ...
    Dec 5, 2014 · We present DESeq2, a method for differential analysis of count data, using shrinkage estimation for dispersions and fold changes to improve stability and ...
  44. [44]
    The RIN: an RNA integrity number for assigning integrity values to ...
    Jan 31, 2006 · A user-independent, automated and reliable procedure for standardization of RNA quality control that allows the calculation of an RNA integrity number (RIN).
  45. [45]
    FastQC A Quality Control tool for High Throughput Sequence Data
    FastQC aims to provide a simple way to do some quality control checks on raw sequence data coming from high throughput sequencing pipelines.Bad Illumina Data · Index of /projects/fastqc/Help · Good Illumina · Publications
  46. [46]
    Adjusting batch effects in microarray expression data using ...
    We propose parametric and non-parametric empirical Bayes frameworks for adjusting data for batch effects that is robust to outliers in small sample sizes.Abstract · INTRODUCTION · EXISTING METHODS FOR... · EB METHOD FOR...
  47. [47]
    [PDF] ERCC RNA Spike-In Control Mixes User Guide (PN 4455352D)
    The Ambion® ERCC RNA Spike-In Control Mixes provide a set of external RNA controls that enable performance assessment of a variety of technology platforms used.
  48. [48]
    Bias in RNA-seq Library Preparation: Current Challenges and ... - NIH
    Apr 19, 2021 · The workflow of RNA-seq is extremely complicated and it is easy to produce bias. This may damage the quality of RNA-seq dataset and lead to an incorrect ...
  49. [49]
    edgeR: a Bioconductor package for differential expression analysis ...
    edgeR is a Bioconductor package for examining differential expression of replicated count data, using an overdispersed Poisson model.
  50. [50]
    DESeq2 vignettes - Bioconductor
    No information is available for this page. · Learn why
  51. [51]
    Cluster analysis and display of genome-wide expression patterns
    A system of cluster analysis for genome-wide expression data from DNA microarray hybridization is described that uses standard statistical algorithms.
  52. [52]
    Dimensionality reduction by UMAP reinforces sample heterogeneity ...
    Jul 27, 2021 · We compare four major dimensionality reduction methods (PCA, multidimensional scaling [MDS], t-SNE, and UMAP) in analyzing 71 large bulk transcriptomic ...
  53. [53]
    Gene selection and classification of microarray data using random ...
    Jan 6, 2006 · We investigate the use of random forest for classification of microarray data (including multi-class problems) and propose a new method of gene selection.Estimation Of Error Rates · Gene Selection Using Random... · Competing MethodsMissing: key | Show results with:key
  54. [54]
    Cell and tumor classification using gene expression data - PNAS
    The purpose of this paper is to describe a new forest construction strategy and illustrate its use for cancer classification based on gene expression levels in ...Methods · Data Structure And The... · Deterministic ForestMissing: supervised | Show results with:supervised<|separator|>
  55. [55]
    A general framework for weighted gene co-expression network ...
    We describe a general framework for soft thresholding that assigns a connection weight to each gene pair. This leads us to define the notion of a weighted gene ...
  56. [56]
    Genomic examples — SHAP latest documentation
    These examples explain machine learning models applied to genomic data. They are all generated from Jupyter notebooks available on GitHub. DeepExplainer ...
  57. [57]
    clusterProfiler: an R Package for Comparing Biological Themes ...
    We present an R package called clusterProfiler for statistical analysis of GO and KEGG, allowing biological theme comparison among gene clusters.
  58. [58]
    SCANPY: large-scale single-cell gene expression data analysis
    Feb 6, 2018 · Scanpy is a scalable toolkit for analyzing single-cell gene expression data. It includes methods for preprocessing, visualization, clustering, pseudotime and ...
  59. [59]
    Gene Expression-Based Cancer Classification for Handling ... - MDPI
    Feb 9, 2024 · In this paper, we address the problem of classifying cancer based on gene expression for handling the class imbalance problem and the curse of dimensionality.
  60. [60]
    Applications of gene pair methods in clinical research
    Apr 9, 2025 · Gene pair analysis as a transformative paradigm for mining high-dimensional omics data, with direct implications for precision biomarker discovery.
  61. [61]
    GO enrichment analysis - Gene Ontology
    An enrichment analysis will find which GO terms are over-represented (or under-represented) using annotations for that gene set.Enrichment Analysis Tool · Using The Go Enrichment... · Interpreting The Results...
  62. [62]
    Gene Ontology (GO) | UniProt help
    Oct 16, 2025 · In UniProt, GO annotations from all three aspects are displayed in the Function section of protein entry pages, and annotated GO terms from ...Missing: expression | Show results with:expression
  63. [63]
  64. [64]
    Home - Gene - NCBI - NIH
    A portal to gene-specific content based on NCBI's RefSeq project, information from model organism databases, and links to other resources.<br />Advanced Search Builder · Help · RefSeqGene · TP53 tumor protein p53
  65. [65]
    A model for isoform-level differential expression analysis using RNA ...
    May 16, 2022 · We developed an isoform-free (without need to pre-specify isoform structures) splicing-graph based negative binomial (SGNB) model for differential expression ...
  66. [66]
    The DAVID Gene Functional Classification Tool - Genome Biology
    Sep 4, 2007 · It is a powerful method to group functionally related genes and terms into a manageable number of biological modules for efficient interpretation of gene lists.
  67. [67]
    g:Profiler—interoperable web service for functional enrichment ...
    May 5, 2023 · g:Profiler is a reliable and up-to-date functional enrichment analysis tool that supports various evidence types, identifier types and organisms.INTRODUCTION · THE CORE OF g:PROFILER · NEW DEVELOPMENTS IN g...
  68. [68]
    Pathway enrichment analysis and visualization of omics data using ...
    A common statistical test used for pathway enrichment analysis of a gene list is a Fisher's exact test based on the hypergeometric distribution. It ...
  69. [69]
    Gene set enrichment analysis: A knowledge-based approach for ...
    We describe a powerful analytical method called Gene Set Enrichment Analysis (GSEA) for interpreting gene expression data.
  70. [70]
    Pathway-Specific Analysis of Gene Expression Data Identifies ... - NIH
    Activation of the PI3K/Akt pathway is associated with incomplete metabolic response in cervical cancer. Targeted inhibition of PI3K/Akt may improve response to ...
  71. [71]
    E2Fs regulate the expression of genes involved in differentiation ...
    The E2F transcription factors are best known for their involvement in the regulation of cell cycle and apoptosis. A considerable number of genes involved in ...Results · E2f-Regulated Genes Are... · Cell Cycle Control And Dna...
  72. [72]
  73. [73]
  74. [74]
  75. [75]
  76. [76]
  77. [77]
    Novel gene signature reveals prognostic model in acute ... - NIH
    In this study, we identified six genes (BAALC, HGF, CPXM1, CCL4, ZBTB10, and B3GNT2) associated with ALL prognosis. In Multi-index ROC analysis of the training ...
  78. [78]
    Improving drug response prediction via integrating gene ...
    Apr 9, 2024 · Pharmacogenomics aims to study how genomic alterations and transcriptomic programming affect drug response, allowing for personalized drug ...
  79. [79]
    K070675 - 510(k) Premarket Notification - FDA
    Classifier, Prognostic, Recurrence Risk Assessment, Rna Gene Expression, Breast Cancer22. 510(k) Number, K070675. Device Name, MAMMAPRINT. Applicant. AGENDIA BV.
  80. [80]
    List of Cleared or Approved Companion Diagnostic Devices - FDA
    Mar 5, 2025 · List of Cleared or Approved Companion Diagnostic Devices (In Vitro and Imaging Tools) ; Bond Oracle HER2 IHC System (Leica Biosystems), Breast ...
  81. [81]
    RNA liquid biopsy via nanopore sequencing for novel biomarker ...
    Jul 5, 2025 · Our findings highlight the utility of our RNA liquid biopsy platform technology for discovering and targeting early stages of disease with molecular precision.
  82. [82]
    EXPECTATIONS, VALIDITY, AND REALITY IN GENE EXPRESSION ...
    To provide a critical overview of gene expression profiling methodology and discuss areas of future development. Gene expression profiling has been used ...
  83. [83]
    Experimental reproducibility limits the correlation between mRNA ...
    Sep 8, 2022 · Within each study, we calculated the median Spearman correlation between mRNA and protein for all proteins that were measured in at least 80% of ...Missing: rho | Show results with:rho
  84. [84]
    Correlation of mRNA and protein in complex biological samples
    Oct 20, 2009 · Their reference data-set containing 2044 proteins showed a good correlation between mRNA and protein levels (r = 0.66).
  85. [85]
    Relationship between differentially expressed mRNA and ... - Nature
    Jun 8, 2015 · We found that differentially expressed mRNAs correlate significantly better with their protein product than non-differentially expressed mRNAs.
  86. [86]
    Introduction to Gene Expression Profiling - Thermo Fisher Scientific
    Gene expression profiling is often used in hypothesis generation. If very little is known about when and why a gene will be expressed, expression profiling ...
  87. [87]
    Perspectives for mass spectrometry and functional proteomics - 2001
    Feb 12, 2001 · A perhaps decisive advantage of proteomics is, however, that changes in gene expression and other cellular processes at the purely protein ...
  88. [88]
    Regulation of transcriptome, translation, and proteome in response ...
    Apr 18, 2012 · In the oxidative stress condition analyzed, transcription dominates translation to control protein abundance.
  89. [89]
    Translational reprogramming in stress response - PMC
    Indeed, cap-independent translation dominates only when the general cap-dependent translation is inhibited by cellular stress.
  90. [90]
    Multi-omics Integration Is the Key to Understanding Biological Systems
    Aug 9, 2019 · The first group of articles describes new methods and tools for studying associations between proteomics data and other types of omics data, ...
  91. [91]
    High-throughput proteomics: a methodological mini-review - Nature
    Aug 3, 2022 · Here, we summarize scientific research and clinical practice of existing and emerging high-throughput proteomics approaches, including mass ...
  92. [92]
    Integration of pan-cancer transcriptomics with RPPA proteomics ...
    Jan 2, 2018 · In this study, we integrate transcriptomics and RPPA data from multiple cancer cell lines to study pan-cancer cellular states associated with ...Missing: bridge | Show results with:bridge
  93. [93]
    Long Non-Coding RNAs in the Regulation of Gene Expression
    Long non-coding RNA can regulates neighbor protein-coding genes expression and thus contribute to the mRNA and protein content in the cell [32,35].Missing: limitations | Show results with:limitations
  94. [94]
    Gene regulation by long non-coding RNAs and its biological functions
    Dec 22, 2020 · Evidence accumulated over the past decade shows that long non-coding RNAs (lncRNAs) are widely expressed and have key roles in gene regulation.
  95. [95]
    Advances and challenges in studying noncoding RNA regulation of ...
    Sep 10, 2019 · Since 1) ncRNAs may influence the levels of proteins responsible for drug metabolism and drug toxicity and 2) drugs can affect the expression of ...2.1. Ncrnas Influence The... · 4. Roles Of Ncrnas In The... · 5. Translating Ncrna Biology...
  96. [96]
    Multi-omics Data Integration, Interpretation, and Its Application - PMC
    They help in assessing the flow of information from one omics level to the other and thus help in bridging the gap from genotype to phenotype. Integrative ...
  97. [97]
    More Is Better: Recent Progress in Multi-Omics Data Integration ...
    For the purpose of precision medicine, additional benefits may be obtained by integrating omics data with other data types, such as imaging and electronic ...
  98. [98]
    Integrated multi-omics is more than the sum of its parts - Nature
    One of the ways multi-omics can better characterize the causes of disease is by identifying genetic variants that affect gene expression. Many different ...
  99. [99]
    Multiview clustering of multi-omics data integration by using a ...
    Jul 21, 2022 · iCluster is an integrative clustering method based on a Gaussian latent variable model with lasso-type penalty terms to induce sparsity in ...
  100. [100]
    eQTL analysis: A bridge from genome to mechanism - ScienceDirect
    Sep 17, 2025 · In this regard, the eQTL analysis can detect the regulatory relationship between SNPs and gene expression and explain the regulation route from ...
  101. [101]
    eQTLs play critical roles in regulating gene expression and ...
    Genome‐wide association study identified 44 354 expression quantitative trait loci (eQTLs), which regulate the expression of 13 201 genes.
  102. [102]
    Multi-omics: Integration Analysis of Epigenomics and Transcriptomics
    Epigenomics is often jointly analyzed with transcriptomics to obtain target genes or identify downstream genes regulated by transcription factors.
  103. [103]
    Multi-omics integration in biomedical research – A metabolomics ...
    By mapping omics, such as transcriptomics, proteomics or epigenomics, back to the gene-level, multiple omics types can be integrated alongside metabolomics ...
  104. [104]
    Multi‐Omics Factor Analysis—a framework for unsupervised ...
    We present Multi‐Omics Factor Analysis (MOFA), a computational method for discovering the principal sources of variation in multi‐omics data sets. MOFA infers a ...Results · Multi‐omics Factor... · Gene Set Enrichment Analysis
  105. [105]
    iOmicsPASS: network-based integration of multiomics data for ...
    Jul 9, 2019 · Our current implementation focuses on the integration of mRNA and protein data over TF regulatory networks and PPI networks (with or without DNA ...
  106. [106]
    The Cancer Genome Atlas Program (TCGA) - NCI
    The Cancer Genome Atlas (TCGA) is a landmark cancer genomics program that sequenced and molecularly characterized over 11000 cases of primary cancer samples ...
  107. [107]
    MultiGATE: integrative analysis and regulatory inference in spatial ...
    Oct 24, 2025 · Spatial multi-omics analysis has emerged as a powerful approach for understanding the interplay between genes, chromatin accessibility, protein, ...
  108. [108]
    Comparison of RNA-Seq and Microarray Gene Expression Platforms ...
    Jan 21, 2019 · The main difference between RNA-Seq and microarrays is that the former allows for full sequencing of the whole transcriptome while the latter ...
  109. [109]
    Microarray experiments and factors which affect their reliability - PMC
    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: paper | Show results with:paper
  110. [110]
    Comparison of RNA-Seq and Microarray in Transcriptome Profiling ...
    RNA-Seq was superior in detecting low abundance transcripts, differentiating biologically critical isoforms, and allowing the identification of genetic ...
  111. [111]
    Handling multi-mapped reads in RNA-seq - ScienceDirect.com
    Such repeated sequences complicate gene/transcript quantification during RNA-seq analysis due to reads mapping to more than one locus, sometimes involving genes ...
  112. [112]
    Assessing and mitigating batch effects in large-scale omics studies
    Oct 3, 2024 · In this review, we highlight the profound negative impact of batch effects and the urgent need to address this challenging problem in large-scale omics studies.
  113. [113]
    RNA-seq: impact of RNA degradation on transcript quantification
    Different transcripts are degraded at different rates. We sought to understand better the nature of transcript degradation in the RNA samples of lower quality.Missing: issues paper
  114. [114]
    Exploring differential exon usage via short- and long-read RNA ...
    Sep 28, 2022 · A recent study has demonstrated that standard RNA-seq is able to robustly recapitulate only about 50% of isoforms detected by long-read Iso-Seq ...
  115. [115]
    Single-cell full-length total RNA sequencing uncovers dynamics of ...
    Feb 12, 2018 · Ideal single-cell total RNA-seq would have high sensitivity, especially to non-poly(A) RNAs, to fully capture transcriptome in single cells ...
  116. [116]
    Benchmarking of a Bayesian single cell RNAseq differential gene ...
    ANOVA, scBT, KW, limma-trend and LRT linear had false positive rates (FPRs) below 3% indicating better performance compared to two group tests. After filtering ...
  117. [117]
    Single-Cell RNA-Seq Data Analysis Guidelines - Biostate.ai
    Sep 13, 2025 · A: The cost of scRNA-seq depends on sample size, sequencing depth, and analysis complexity. Library preparation can cost $50-200 per sample, ...
  118. [118]
    Differentially expressed genes reflect disease-induced rather than ...
    Sep 24, 2021 · However, DEG analyses are unable to distinguish between causes, consequences, or mere correlations between gene expression and phenotypes. To ...
  119. [119]
    Exploiting fluctuations in gene expression to detect causal ... - eLife
    Jan 25, 2024 · Perturbation experiments are a conceptually simple solution to avoid this problem and directly infer causal relationships in gene regulatory ...
  120. [120]
    Gene co-expression in the interactome: moving from correlation ...
    Jan 21, 2021 · The basic premise of this exercise is that, even though correlation is not causation, co-expressed genes are functionally coordinated in ...
  121. [121]
    The challenge of gene expression profiling in heterogeneous ...
    This chapter will (1) give a brief overview on how heterogeneity may influence gene expression profiling data and (2) describe the methods that are currently ...
  122. [122]
    An algorithm to quantify intratumor heterogeneity based on ... - Nature
    Sep 11, 2020 · Because alterations in genome and epigenome profiles often lead to heterogeneous gene expression profiles in tumors, defining ITH based on gene ...Missing: context | Show results with:context
  123. [123]
    Dynamic Cancer Cell Heterogeneity: Diagnostic and Therapeutic ...
    Jan 7, 2022 · Cancer heterogeneity refers to subpopulations of cells with distinct genotypes and phenotypes within a tumor, arising from genetic, epigenetic, ...
  124. [124]
    Complementary Post Transcriptional Regulatory Information is ...
    Feb 22, 2016 · Due to translation regulation (and other post transcriptional regulatory steps such as degradation of mRNAs and proteins) the protein levels ...
  125. [125]
    Extensive post-transcriptional buffering of gene expression in the ...
    Jul 29, 2019 · Here we perform RNA-Seq, ribosome profiling and proteomics analyses in baker's yeast cells grown in rich media and oxidative stress conditions ...
  126. [126]
    Transcriptional bursting dynamics in gene expression - PMC - NIH
    Transcriptional bursting represents a type of molecular dynamics that manifests as the heterogeneous expression of identical genes across different cells. The ...
  127. [127]
    The balance of reproducibility, sensitivity, and specificity of lists of ...
    Reproducibility is a fundamental requirement in scientific experiments. Some recent publications have claimed that microarrays are unreliable because lists ...
  128. [128]
    Addressing bias in big data and AI for health care - ScienceDirect.com
    Oct 8, 2021 · Here, we describe the challenges in rendering AI algorithms fairer, and we propose concrete steps for addressing bias using tools from the field of open ...
  129. [129]
    Ethical Machine Learning in Healthcare - Annual Reviews
    Jul 20, 2021 · The use of machine learning (ML) in healthcare raises numerous ethical concerns, especially as models can amplify existing health inequities.
  130. [130]
    ArrayExpress update – from bulk to single-cell expression data
    Oct 24, 2018 · ArrayExpress is an archive of functional genomics data that includes a range of experiment types, such as gene expression, methylation profiling ...
  131. [131]
    A closer look at cross-validation for assessing the accuracy of gene ...
    Apr 26, 2018 · Cross-validation (CV) is a technique to assess the generalizability of a model to unseen data. This technique relies on assumptions that may not ...
  132. [132]
    Review and Cross-Validation of Gene Expression Signatures and ...
    This successful cross-validation indicates that gene expression analysis-based signatures are becoming translationally relevant to care of melanoma patients, ...
  133. [133]
    Diagnostic and prognostic prediction using gene expression profiles ...
    The cross-validated prediction error is an estimate of the prediction error associated with the algorithm for model building used. It is not an estimate ...Estimating Accuracy Of A... · Figure 1 · Class Prediction Algorithms
  134. [134]
    Methods to increase reproducibility in differential gene expression ...
    One of the ways to improve reproducibility is integrating multiple microarray datasets via gene expression meta-analysis, which has proven useful in practice ...
  135. [135]
    GEO Datasets for Transcriptomics Meta-Analysis in Research
    Sep 14, 2023 · By combining multiple datasets, researchers can identify gene expression patterns that are more robust and reproducible across different ...
  136. [136]
    Meta Analysis of Gene Expression Data within and Across Species
    Public databases, such as GEO [26] or ArrayExpress [27] offer a central repository of MIAME-compliant microarray data. Although these databases are an extremely ...
  137. [137]
    scDesign2: a transparent simulator that generates high-fidelity ...
    May 25, 2021 · We propose scDesign2, a transparent simulator that achieves all three goals and generates high-fidelity synthetic data for multiple single-cell gene expression ...
  138. [138]
    A benchmark study of simulation methods for single-cell RNA ...
    Nov 25, 2021 · scDesign2: a transparent simulator that generates high-fidelity single-cell gene expression count data with gene correlations captured.
  139. [139]
    FAIRification of an RNAseq dataset - Galaxy Training!
    Mar 27, 2024 · This lesson will take you through a publicly available RNAseq dataset in ArrayExpress and show you how it meets FAIR principles.