Fact-checked by Grok 2 weeks ago

Transcriptome

The transcriptome is the complete set of all RNA transcripts, including messenger RNA (mRNA), non-coding RNA, and other RNA molecules, produced by the genome of a cell, tissue, or organism at a specific point in time. It represents a dynamic snapshot of gene expression, capturing which genes are actively transcribed and to what extent, in contrast to the static DNA sequence of the genome. Studying the transcriptome, known as transcriptomics, provides critical insights into biological processes by revealing patterns of gene activity across different types, developmental stages, environmental conditions, and states. For instance, variations in transcript levels can explain functional differences between healthy and diseased tissues, such as elevated expression of certain s in cancer cells that promote uncontrolled growth. This field has advanced our understanding of regulation, including how external factors influence RNA production and how non-coding RNAs contribute to cellular functions beyond protein synthesis. Key technologies for transcriptome analysis have evolved significantly since the early 1990s. Microarrays, introduced in the mid-1990s, allow simultaneous measurement of thousands of predefined RNA sequences through hybridization, enabling gene expression profiling in specific contexts. More recently, RNA sequencing (RNA-seq), leveraging next-generation sequencing since the 2000s, offers a comprehensive, unbiased view by capturing the full range of transcripts, including low-abundance and novel ones, with higher sensitivity and dynamic range. These methods have facilitated large-scale projects, such as the Genotype-Tissue Expression (GTEx) initiative and the Encyclopedia of DNA Elements (ENCODE), which map transcriptome variations across human tissues to link genes to functions. Historically, the concept of the transcriptome emerged from early efforts to catalog mRNA sequences, with the first comprehensive transcriptome study in analyzing 609 sequences, expanding to over 16,000 genes by 2008. Today, transcriptomics plays a pivotal role in biomedical research, aiding in discovery, , and elucidating complex regulatory networks that drive development and response to therapies.

Definition and Fundamentals

Definition and Scope

The transcriptome refers to the complete set of RNA transcripts produced by the genome of a cell, tissue, or organism under specific conditions, encompassing messenger RNAs (mRNAs), non-coding RNAs (such as ribosomal RNAs, transfer RNAs, and long non-coding RNAs), and their splice variants or isoforms. This collection represents the expressed portion of the genome, capturing the diversity of RNA molecules generated through transcription at a given moment. Unlike the static genome, which consists of the entire DNA sequence, the transcriptome is limited to those genomic regions actively transcribed into RNA, excluding untranscribed DNA elements. The scope of the transcriptome extends to all transcribed RNAs within defined biological contexts, such as a single , a sample, or an entire , and is profoundly influenced by factors including developmental stage, environmental stimuli, and physiological perturbations. For instance, it distinguishes itself from the —the full array of proteins translated from those RNAs—by focusing solely on the intermediate RNA products rather than post-transcriptional modifications or outcomes. This boundary underscores the transcriptome's role as a bridge between genetic and functional outputs, highlighting regulatory layers like that generate transcript variants without altering the underlying DNA. As a dynamic entity, the transcriptome provides a temporal of regulation, reflecting real-time responses to cellular needs and external cues, with its composition fluctuating across conditions. Quantitative assessment of transcript abundance often employs metrics such as transcripts per million (TPM), which normalizes read counts by transcript length and total sequencing depth to enable comparable expression levels across samples. The term "transcriptome" originated in the amid early post-genome sequencing efforts to catalog expressed sequences.

Etymology and Historical Development

The term "transcriptome" is a portmanteau derived from "transcript," referring to a copy of genetic , and the "-ome," denoting a complete set or body, analogous to "" and "." It was first proposed by Charles Auffray in 1996 and first used in a scientific in 1997 by E. Velculescu and colleagues in their analysis of in , where they described the transcriptome as the full set of expressed genes and their expression levels in a defined of cells. This emerged during the rapid expansion of in the late , building on the conceptual framework established by earlier "-ome" terms to encapsulate the dynamic output of the . The historical roots of transcriptome research trace back to the early 1990s, when efforts to catalog expressed genes laid the groundwork for comprehensive profiling. A pivotal precursor was the development of expressed sequence tags (ESTs) by Mark D. Adams and colleagues in 1991, who sequenced short complementary DNA fragments from human brain mRNA to identify and map expressed genes efficiently, generating over 600 novel sequences as part of the Human Genome Project's initial phases. This approach shifted focus from genomic DNA to RNA transcripts, enabling the first large-scale glimpses into gene expression patterns. By 1995, Victor E. Velculescu's team introduced serial analysis of gene expression (SAGE), a method that concatenated short tags from transcripts for high-throughput quantification, formalizing the study of transcriptomes in eukaryotic cells. Key technological milestones accelerated transcriptome exploration in the mid-1990s and beyond. In 1995, Mark Schena, working with Patrick O. Brown at Stanford University, pioneered DNA microarray technology, which allowed simultaneous measurement of thousands of gene expression levels by hybridizing labeled RNA to immobilized DNA probes on glass slides, revolutionizing parallel transcript analysis. The completion of the Human Genome Project in 2003 provided a reference sequence that propelled transcriptome studies, enabling more precise mapping and comparison of expressed genes across conditions. The advent of RNA sequencing (RNA-seq) in 2008, demonstrated by Ali Mortazavi and colleagues, marked a transformative leap by directly sequencing cDNA libraries to quantify transcript abundance without prior knowledge of sequences, offering unprecedented depth and accuracy in mammalian transcriptomes. In the 2010s and 2020s, advances in further refined the field's resolution, with pioneering demonstrations by Fuchou Tang and colleagues in 2009 enabling the profiling of individual cells to reveal cellular heterogeneity, building on earlier bulk methods to uncover dynamic expression landscapes in and . Influential figures like , whose innovations democratized expression profiling, and early pioneers such as Barbara Wold, have shaped the trajectory toward integrative, high-resolution transcriptome analysis.

Biological Processes

Transcription Mechanism

Transcription is the process by which genetic information encoded in DNA is copied into RNA molecules, primarily through the action of RNA polymerases, generating the foundational transcripts that comprise the transcriptome. In eukaryotes, RNA polymerase II (Pol II) is responsible for synthesizing messenger RNA (mRNA) precursors from protein-coding genes, while RNA polymerases I and III handle ribosomal and transfer RNAs, respectively. The core mechanism involves three main stages: initiation, elongation, and termination, each regulated to ensure precise gene expression. In prokaryotes, a single RNA polymerase, aided by sigma factors for promoter recognition, performs all transcription, differing from eukaryotes by lacking a nucleus and involving simpler initiation without compartmentalization. Initiation in eukaryotes begins with the assembly of the pre-initiation complex () at the promoter, where general transcription factors such as TFIID bind the and recruit Pol II along with TFIIB, TFIIE, TFIIF, and TFIIH. TFIIH's activity unwinds DNA to form the transcription bubble, allowing Pol II to start RNA synthesis from the +1 site. Regulatory elements like enhancers and silencers, often located distal to promoters, modulate this process by binding specific transcription factors that loop DNA to interact with the , while modifications such as acetylation by coactivators open nucleosomes for access. follows, with Pol II processively synthesizing nascent at rates of 20–60 per second, during which early capping, splicing, and signals are recognized for co-transcriptional processing. Termination in eukaryotes for Pol II-transcribed genes occurs upon recognition of polyadenylation signals (e.g., AAUAAA) in the nascent , triggering cleavage and addition of a , followed by the torpedo mechanism where Rat1 degrades the downstream , releasing Pol II. In prokaryotes, termination relies on rho-dependent or intrinsic mechanisms involving structures, without , and factors dissociate post-initiation for reuse. Transcription frequency can be modeled as the number of transcripts produced per per , with essential genes maintaining a minimum of one transcript per cycle to ensure viability, highlighting the balance between initiation efficiency and regulatory control. A key source of transcript diversity arises during or shortly after transcription through , where introns are removed and exons joined in varying combinations by the , potentially generating multiple isoforms from a single pre-mRNA. This process, coupled to elongation, allows fine-tuned regulation but is distinct from the core synthesis mechanism.

Types of RNA Transcripts

The transcriptome encompasses a diverse array of molecules produced by transcription, broadly classified into protein-coding messenger RNAs (mRNAs) and non-coding RNAs (ncRNAs), which together regulate cellular functions from protein synthesis to control. mRNAs constitute approximately 1-5% of total cellular and serve as templates for protein , while ncRNAs, comprising the majority, include structural and regulatory species such as ribosomal RNAs (rRNAs), transfer RNAs (tRNAs), small nuclear RNAs (snRNAs), microRNAs (miRNAs), small interfering RNAs (siRNAs), long non-coding RNAs (lncRNAs), and circular RNAs (circRNAs). This classification highlights the transcriptome's complexity, where ncRNAs often outnumber mRNAs and exert multifaceted regulatory roles. Messenger RNAs are the primary protein-coding transcripts, generated from protein-coding genes and processed through capping, splicing, and to ensure stability and export from the . Capping involves the addition of a 7-methylguanosine cap at the 5' end shortly after transcription , protecting the mRNA from and facilitating ribosome binding; splicing removes introns via the , which includes snRNAs; and polyadenylation adds a poly-A tail at the 3' end, enhancing mRNA export and efficiency. These mRNAs are translated by into proteins, representing the core of . In humans, of pre-mRNAs generates over 100,000 distinct mRNA isoforms from approximately 20,000 genes, enabling proteomic diversity through inclusion or exclusion of exons. Among ncRNAs, rRNAs form the structural backbone of ribosomes, accounting for about 80-90% of total cellular RNA and facilitating protein synthesis by catalyzing formation. tRNAs, comprising roughly 10-15% of RNA, function as adaptors in translation, delivering to the based on anticodon-mRNA codon matching. snRNAs, part of the , mediate removal during mRNA processing, ensuring accurate splicing. Regulatory ncRNAs include small species like miRNAs and siRNAs, which typically span 20-25 and post-transcriptionally repress . miRNAs are processed from primary transcripts (pri-miRNAs) in the by the Drosha-DGCR8 to form precursor miRNAs (pre-miRNAs), which are exported to the and cleaved by into mature miRNAs; these then bind to the 3' (UTR) of target mRNAs, promoting degradation or translational inhibition. siRNAs, often derived from exogenous sources or endogenous duplexes, similarly induce mRNA silencing via the (RISC).01206-6) LncRNAs, defined as transcripts longer than 200 without protein-coding potential, are transcribed by similarly to mRNAs but often retained in the due to inefficient splicing, repeat elements, or interactions with chromatin-modifying complexes, enabling roles in epigenetic such as X-chromosome inactivation or enhancer . CircRNAs arise from back-splicing events where exons are joined in a covalent loop, bypassing linear 5' and 3' ends, resulting in highly stable molecules that act as miRNA sponges or transcriptional regulators. Together, these types underscore the transcriptome's regulatory depth, with ncRNAs modulating mRNA abundance and function to fine-tune cellular responses.

Methods of Transcriptome Profiling

DNA Microarrays

DNA microarrays, also known as arrays, operate on the principle of , where short DNA probes are immobilized on a , such as a glass slide or chip, in a high-density grid format. These probes are designed to be complementary to specific mRNA sequences from the transcriptome of interest. Total RNA or mRNA is extracted from a sample, reverse transcribed into (cDNA), and labeled with fluorescent dyes. The labeled cDNA hybridizes to matching probes on the array, and the resulting fluorescence intensity, detected via , is proportional to the abundance of the corresponding transcript in the original sample, thereby quantifying levels. The typical workflow for DNA microarray-based transcriptome profiling begins with RNA extraction from cells or tissues, followed by reverse transcription of the RNA into cDNA using reverse transcriptase enzymes. The cDNA is then chemically labeled, often with fluorescent dyes such as Cy3 (green) or Cy5 (red) in two-color formats, where two samples are hybridized simultaneously to enable direct comparison of expression levels via the ratio of signals. In one-color formats, such as those used by platforms, a single sample is labeled (e.g., with for indirect fluorescence detection) and hybridized separately. After hybridization, the is washed to remove unbound targets, and a measures fluorescence at each probe spot to generate raw intensity data for downstream analysis. DNA microarrays are categorized into several types based on probe design and fabrication. cDNA microarrays use longer PCR-amplified DNA fragments (typically 200–500 base pairs) spotted onto the array surface via robotic printing, allowing for custom arrays targeting specific gene sets. microarrays, in contrast, employ shorter synthetic probes (20–60 ), which offer higher specificity and are produced either by synthesis methods, such as on GeneChips using 25-mer probes, or by as in Agilent or NimbleGen platforms. These can be designed for whole-genome coverage or focused subsets, with GeneChips being a seminal example that enabled comprehensive transcriptome profiling starting in the late 1990s. DNA microarrays provided high-throughput profiling of thousands of known genes at relatively low cost, making them a cornerstone of transcriptomics research during their historical peak in the 2000s, including their use in the pilot project in 2007 to map transcribed regions across 1% of the via arrays. However, they are limited to detecting only predefined transcripts, missing novel or low-abundance ones, and suffer from issues like cross-hybridization between similar sequences, which reduces specificity. Additionally, their for quantifying expression levels is constrained to approximately 3–4 orders of magnitude, potentially under- or overestimating extreme expression differences compared to more sensitive methods.

Bulk RNA Sequencing

Bulk RNA sequencing, commonly referred to as , is a high-throughput method that involves the deep sequencing of (cDNA) libraries derived from samples to digitally quantify the abundance of all RNA species through read counts. This approach enables comprehensive transcriptome profiling at the population level by capturing the average expression across a bulk sample of cells, providing a snapshot of gene activity without reliance on predefined probes. The workflow for bulk begins with isolation from the sample, followed by depletion of (rRNA), which constitutes the majority of total , to enrich for informative transcripts. Common rRNA depletion strategies include poly-A selection, which captures mRNA via oligo-dT primers targeting polyadenylated tails, or methods like Ribo-Zero, an enzymatic approach that removes both cytoplasmic and mitochondrial rRNA using targeted probes. The is then fragmented, reverse-transcribed into cDNA, and ligated with adapters for sequencing compatibility. amplification increases library yield, after which the library undergoes sequencing, typically using short-read platforms like Illumina, generating millions of reads that are subsequently aligned to a for quantification. Emerging long-read using platforms like Oxford Nanopore or PacBio offers improved isoform resolution, with recent benchmarks highlighting its advantages as of 2025. Several variants of bulk RNA-seq exist to address specific research needs. Total RNA-seq profiles the entire population, including non-coding RNAs, after rRNA depletion, whereas mRNA-enriched protocols focus on polyadenylated transcripts for targeted analysis. Libraries can also be prepared as stranded, preserving information on the originating DNA strand to distinguish overlapping genes, or unstranded, which simplifies preparation but loses this detail. For eukaryotic samples, sequencing depth typically ranges from 20 to 50 million reads per sample to achieve sufficient coverage for accurate quantification. Bulk offers key advantages over earlier methods like microarrays, including unbiased detection of novel transcripts, events, and gene fusions, as well as a wide exceeding 10^5-fold for expression levels and single-nucleotide resolution for variant identification. However, limitations include challenges in aligning short reads to resolve complex isoforms or repetitive regions, and historically high costs that restricted accessibility before the 2010s. To normalize for length and sequencing depth, expression levels are often quantified using metrics such as reads per kilobase million (RPKM) or fragments per kilobase million (FPKM). The RPKM formula is given by: \text{RPKM} = \frac{\text{reads mapped to [gene](/page/Gene)}}{(\text{[gene](/page/Gene) length in kb}) \times (\text{total million reads})} \times 10^9 This measure allows comparable expression estimates across and samples. FPKM extends this to paired-end data by accounting for fragments rather than individual reads.

Single-Cell and Spatial Transcriptomics

Single-cell RNA sequencing (scRNA-seq) enables the profiling of transcriptomes from individual cells, revealing cellular heterogeneity that bulk methods obscure. This approach emerged in the early 2010s, with foundational protocols like Smart-seq, a plate-based method that captures full-length transcripts through reverse transcription and template switching, allowing sensitive detection in low-input samples. Droplet-based techniques, such as Drop-seq and inDrop, revolutionized scalability by encapsulating single cells with barcoded beads in microfluidic droplets, enabling parallel processing of thousands to millions of cells. Commercial platforms like , introduced in 2016, built on these principles to achieve high-throughput droplet encapsulation for 3' or 5' end profiling. The typical scRNA-seq workflow begins with cell isolation, often using fluorescence-activated (FACS) to select viable cells based on surface markers or viability dyes. Cells are then lysed to release , which is captured on barcoded primers; poly-A selection targets mRNA, followed by reverse transcription, amplification, and library preparation for next-generation sequencing. Post-sequencing, involves demultiplexing by cell barcodes and collapsing duplicates using unique molecular identifiers (UMIs), which tag individual transcripts to mitigate bias. This resolves distinct cell types through unsupervised clustering of profiles, typically detecting 1,000 to 10,000 genes per cell depending on capture efficiency and cell type. Spatial transcriptomics extends scRNA-seq by preserving tissue architecture, mapping transcripts to their positional context. Early methods like the Spatial Transcriptomics platform used arrayed barcodes on slides to capture mRNA from permeabilized tissue sections, enabling genome-wide profiling at ~100 μm resolution. The Visium platform, launched by 10x Genomics in 2019 following their acquisition of Spatial Transcriptomics, refines this with higher-density arrays (~55 μm spots) for unbiased whole-transcriptome analysis in fresh-frozen or FFPE samples. More recently, the Visium HD platform, launched in 2024, provides higher resolution spatial profiling at approximately 2 μm pixel size, enabling near-single-cell analysis. In situ hybridization techniques achieve single-cell or subcellular resolution without dissociation; multiplexed error-robust FISH (MERFISH) images hundreds to thousands of genes using combinatorial barcoding and error-correcting codes, while sequential FISH (seqFISH) employs iterative hybridizations for scalable multiplexing up to ~10,000 genes. Recent advances, particularly from the early 2020s, include long-read scRNA-seq using PacBio or platforms to resolve full-length isoforms and complex splicing patterns at single-cell resolution, as demonstrated in studies profiling parasite transcriptomes in 2022. Multi-modal approaches like integrate profiling with surface protein detection via oligo-tagged antibodies, providing complementary phenotypic data in the same droplet-based . Key concepts such as UMIs address amplification biases by tags per ; for a given , the corrected expression is the number of distinct UMIs, normalized as: \text{Expression} = \frac{\text{Unique UMIs per gene}}{\text{Total UMIs per cell}} \times 10^4 This UMI-based normalization enhances accuracy over raw read counts. Despite these innovations, scRNA-seq and spatial methods face challenges including high dropout rates—where low-abundance transcripts are missed due to inefficient capture—leading to sparse matrices with up to 90% zeros. Sparsity complicates downstream clustering and imputation, while computational scaling demands efficient algorithms for datasets exceeding millions of cells. These techniques have previewed applications in dissecting tumor heterogeneity, such as identifying therapy-resistant subclones in melanoma via scRNA-seq clustering. In development, they trace cellular trajectories, revealing dynamic gene programs across lineages.

Data Analysis

Bioinformatics Pipelines

Bioinformatics pipelines for transcriptome analysis transform raw sequencing data, typically from experiments, into interpretable formats through a series of computational steps focused on , , and quantification. These workflows ensure and accuracy in handling the complexities of transcriptomic data, such as variable read lengths and splicing events. The initial step involves quality control and preprocessing to assess and improve raw read quality. Tools like FastQC evaluate sequence quality metrics, including per-base quality scores and adapter , identifying issues such as low-quality bases or overrepresented sequences. Following assessment, adapter removal and trimming are performed using programs like Trimmomatic, which employs sliding window algorithms to clip low-quality regions and remove Illumina adapters, thereby reducing artifacts that could bias downstream analyses. Additional preprocessing addresses transcriptome-specific challenges, such as rRNA via tools like SortMeRNA or poly-A handling through targeted trimming to focus on mRNA. Read alignment maps processed reads to a or transcriptome, accounting for splicing. Spliced aligners like use a suffix array-based approach with seed-and-extend matching to rapidly align reads across boundaries, achieving high sensitivity for novel isoforms. Similarly, HISAT2 employs a graph-based indexing strategy with Burrows-Wheeler transform for efficient alignment of spliced reads, particularly suited for large genomes. For non-model organisms lacking a reference, de novo assembly precedes alignment; , for instance, constructs transcripts via de Bruijn graph-based of overlapping k-mers, enabling discovery of unannotated genes despite challenges like chimeric assemblies from repetitive regions. Quantification follows alignment to estimate transcript or gene expression levels. featureCounts assigns aligned reads to genomic features using efficient interval tree data structures, producing raw count matrices for statistical analysis. For faster pseudo-alignment, Salmon employs lightweight quasi-mapping and probabilistic inference to quantify abundances without full base-pair alignment, mitigating biases from multi-mapping reads. Workflow management systems enhance pipeline reproducibility and scalability. Open-source platforms like provide web-based interfaces for integrating tools into visual workflows, supporting RNA-seq analysis from raw to outputs without local installation. Nextflow facilitates portable, containerized pipelines using for parallel execution across clusters or clouds, addressing variability in computational environments. Scalability for large datasets often leverages , such as AWS Batch, to distribute alignment and quantification tasks, handling terabyte-scale from bulk or single-cell experiments. Key challenges in these pipelines include batch effects from technical variations across sequencing runs, which can confound biological signals; correction methods like apply empirical Bayes modeling to adjust expression data while preserving true differences. Reference bias arises when reads align preferentially to the , underrepresenting variants in diverse samples; this is exacerbated in non-model organisms and can be partially alleviated by de novo approaches or personalized references. Spliced algorithms, such as those in , incorporate dynamic programming for exon chaining to score potential splice junctions, balancing speed and accuracy without exhaustive global . Outputs from these pipelines include aligned reads in BAM or formats for and further processing, alongside gene-level count matrices that serve as input for expression .

Differential Expression and Functional

Differential expression identifies genes or transcripts whose expression levels differ significantly between conditions, such as healthy versus diseased tissues, using statistical models tailored to the overdispersed count data from sequencing. Widely adopted tools like DESeq2 and edgeR model read counts with a to account for both Poisson-like sampling variance and additional biological variability, enabling robust estimation of dispersion and mean expression. In DESeq2, shrinkage estimation stabilizes variance and calculations, improving detection power especially for low-count genes. The fold change is typically computed as the log2 ratio of normalized mean counts between conditions: \text{Fold change} = \log_2 \left( \frac{\text{mean normalized counts}_{\text{condition 1}}}{\text{mean normalized counts}_{\text{condition 2}}} \right), with statistical significance assessed via Wald or likelihood ratio tests, followed by multiple testing correction using the false discovery rate (FDR), where adjusted p-values below 0.05 indicate significant differential expression. edgeR employs an empirical Bayes approach to moderate dispersion estimates across genes, enhancing reliability in experimental designs with limited replicates. Isoform-level analysis extends differential expression to alternative splicing and transcript variants, crucial for understanding regulatory complexity beyond gene-level summaries. Tools like StringTie assemble transcripts de novo from aligned reads using network flow algorithms, producing accurate abundance estimates via maximum flow minimization. Cufflinks, an earlier reference-based assembler, quantifies isoform expression by modeling read compatibility with transcript structures and normalizing for library size and biases. For detecting splicing differences, MAJIQ quantifies local splicing variations (LSVs) and computes percent spliced-in (Ψ) values, identifying complex events like mutually exclusive exons with condition-specific changes. Functional enrichment analysis interprets lists of differentially expressed genes by assessing overrepresentation in predefined biological categories, revealing impacted processes or pathways. Gene Ontology (GO) enrichment, performed via tools like or g:Profiler, categorizes genes into biological processes, molecular functions, and cellular components, using statistical tests to identify significant terms. integrates multiple annotations and applies modified Fisher's exact tests for clustering and visualization, while g:Profiler supports ordered queries and custom backgrounds for nuanced interpretations. Pathway analysis similarly evaluates enrichment in curated databases like and Reactome, which map genes to metabolic, signaling, and regulatory pathways. The hypergeometric test computes the probability of observing k or more overlapping differentially expressed genes in a pathway of size g, from n total differentially expressed genes out of N annotated genes: p = 1 - \sum_{i=0}^{k-1} \frac{\binom{g}{i} \binom{N-g}{n-i}}{\binom{N}{n}}. This test assumes random sampling and is adjusted for multiple comparisons to highlight biologically relevant pathways. Visualization techniques aid in exploring differential expression results, with volcano plots plotting log fold changes against -log10 FDR to highlight significant genes, heatmaps displaying clustered expression patterns across samples, and principal component analysis (PCA) revealing sample grouping and outliers based on variance. These methods facilitate intuitive assessment of data structure and biological signals. Advanced approaches, such as weighted gene co-expression network analysis (WGCNA), integrate machine learning to construct networks from expression correlations, identifying modules of co-expressed genes associated with traits or conditions for deeper functional insights. By 2025, WGCNA remains a cornerstone for modular analysis, often combined with enrichment to link network topology to biological function.

Applications

In Human Health and Disease

The transcriptome plays a pivotal role in elucidating human and by revealing dynamic patterns that underpin cellular responses to health and disease states. In , large-scale initiatives like (TCGA), spanning 2006 to 2018, profiled transcriptomes from over 20,000 primary cancer and matched normal samples across 33 cancer types, enabling the identification of recurrent gene fusions such as fusions that drive tumorigenesis. For instance, pan-cancer transcriptome analyses uncovered fusion transcripts like EML4-ALK in lung adenocarcinoma, highlighting actionable oncogenic drivers validated in nearly 7,000 tumor samples. Similarly, during the , early 2020 transcriptomic studies revealed dysregulated immune responses, including pro-inflammatory profiles and shifts in immune cell subsets, as seen in single-cell analyses of peripheral blood from infected patients. In , transcriptome profiling via sequencing has advanced by identifying expression signatures predictive of drug responses, allowing tailored therapeutic strategies to minimize adverse effects and optimize efficacy. For example, expands pharmacogenomic analysis beyond static variants to capture how genetic differences influence broader transcriptional networks involved in and resistance. Biomarker discovery has benefited from circulating tumor RNA (ctRNA) in liquid biopsies, which detects tumor-derived transcripts non-invasively; a 2025 multicenter study demonstrated that combining ctRNA with increased actionable diagnostic yield by 36.7% in advanced cancers. Transcriptomic approaches also illuminate developmental and neurological processes, such as differentiation trajectories inferred from single-cell sequencing, which map transitional transcriptional states guiding lineage commitment in human pluripotent s. In , single-cell transcriptomic atlases of the , including multi-region dissections from 2023 onward, have profiled over 1.3 million cells to uncover cell-type-specific vulnerabilities, such as reactive signatures and neuronal resilience factors across cortical regions. Recent advances as of 2025 emphasize to dissect (TME) heterogeneity, revealing spatially resolved interactions between cancer cells and immune infiltrates that contribute to therapy resistance; for instance, heterogeneous learning on spatial has quantified TME compartmentalization in solid tumors. Multi-omics integration, combining with and , has identified novel targets, such as tumor cell-derived (MIF) in , which potentiates anti-PD-1 efficacy when inhibited. A prominent is subtyping using the PAM50 gene set, derived from and , which classifies tumors into luminal A, luminal B, HER2-enriched, basal-like, and normal-like subtypes based on expression of 50 genes, informing and decisions with high concordance across platforms.

In Plant and Agricultural Sciences

In plant and agricultural sciences, transcriptome profiling has revolutionized the understanding of dynamics in crops, enabling improvements in yield, resilience, and quality under varying environmental conditions. By capturing the complete set of transcripts, these analyses reveal how adapt to abiotic stresses, developmental cues, and genetic variations, informing breeding strategies for . Seminal sequencing studies in model like have identified key regulatory networks that underpin agronomic traits, facilitating targeted interventions to enhance . Transcriptome studies have been pivotal in elucidating responses to abiotic stresses such as and , identifying genes that confer for crop improvement. In , analyses under conditions revealed upregulated pathways involving , , and long non-coding RNAs, highlighting adaptive mechanisms like stomatal regulation and synthesis. Similarly, dehydration-responsive element-binding (DREB) transcription factors, such as DREB1A and DREB2A, emerge as central regulators in response to and across species; for instance, overexpression of DREB genes in and enhances by modulating downstream targets in signaling and scavenging. In , recent transcriptomic profiling under stress (2025) showed alternative splicing events in hormone-related genes, providing insights into temporal regulatory shifts that could be harnessed for resilient varieties. In , transcriptome data integrated with quantitative trait loci (QTL) mapping, particularly expression QTL (eQTL), has accelerated the identification of genetic variants influencing agronomic performance. eQTL analyses in have linked non-additive patterns to hybrid vigor (), revealing trans-eQTLs that dominate dominance effects and contribute to yield enhancements in hybrids. In , genome-wide transcriptome profiling of super hybrids identified differentially expressed genes in and stress response pathways, explaining up to 20-30% of for grain weight and plant height. These approaches enable , as demonstrated in QTL mapping studies that correlate eQTL hotspots with like flowering time and nutrient efficiency, streamlining breeding for elite cultivars. Transcriptome profiling has illuminated developmental processes critical to crop productivity, such as flowering time regulation and fruit . In and , has delineated networks involving the FLOWERING LOCUS T () gene family, where FT-like genes (e.g., Hd3a and RFT1 in ) integrate photoperiod and temperature signals to trigger floral transition; temporal analyses show phased expression waves that fine-tune heading date for optimal yield. For fruit , spatiotemporal transcriptome mapping in revealed dynamic shifts in -responsive genes and cell wall modifiers across fruit layers, with over 5,000 differentially expressed transcripts linking metabolic pathways to flavor and texture development. Similar studies in identified and signaling hubs that orchestrate climacteric , aiding post-harvest . Recent advances as of 2025 have expanded transcriptome applications through -based editing and spatial technologies for precise trait engineering. /Cas9-mediated edits targeting transcriptome regulators, such as DREB or FT homologs, have produced drought-tolerant and lines with verified expression changes enhancing yield under stress, bypassing traditional breeding timelines. Spatial in has uncovered zone-specific transcript gradients for nutrient transporters (e.g., NRT1 family for uptake), revealing how developmental stages influence and acquisition efficiency during grain filling. These tools enable pan-transcriptomic designs that capture varietal for climate-adaptive crops. Case studies underscore the practical impact of transcriptomics in . The 2012 tomato incorporated transcriptome data to annotate over 34,000 , identifying flavor-related loci like those in and phenylpropanoid pathways, which informed for improved taste in commercial varieties. In , pan-transcriptome analyses across cultivars (2025) have revealed structural variants enriching agronomic traits, such as QTLs with expression biases in hexaploid lines, supporting genomics-assisted to boost global yields by targeting adaptive networks.

In Microbial and Environmental Studies

In microbial transcriptomics, RNA sequencing has been instrumental in elucidating regulation and the discovery of small regulatory RNAs (sRNAs) in model organisms like . Strand-specific applied to E. coli K-12 during steady-state exponential growth revealed an unprecedented high-resolution view of the bacterial architecture, identifying 2,566 transcription units and highlighting the prevalence of overlapping and nested operons that challenge traditional models of polycistronic transcription. In the early 2000s, analyses uncovered novel sRNAs, such as those encoded in intergenic regions, which modulate under diverse physiological conditions like stress responses. In the , analyses expanded this to uncover additional novel sRNAs. These sRNAs often act by base-pairing with target mRNAs to influence operon-level regulation, as demonstrated in large-scale compendia that identified 92 independently regulated transcription units across over 250 E. coli datasets. Transcriptomic studies have also illuminated antibiotic resistance mechanisms in , revealing dynamic changes that confer survival advantages. Comparative of E. coli exposed to nine antibiotic classes showed class-specific transcriptomic rewiring, including upregulation of efflux pumps and stress response pathways like the regulon, which enable rapid adaptation to sublethal doses. In multidrug-resistant strains, transcriptomics has pinpointed stereotypic rewiring of core metabolic and ribosomal genes, with plasticity in response to antibiotics like beta-lactams driving evolutionary resistance trajectories. Metatranscriptomics extends these insights to microbial communities by profiling community-wide , often from complex microbiomes like the gut or , where mRNA enrichment is crucial to separate eukaryotic rRNA and focus on prokaryotic activity. In the human gut, metatranscriptomic sequencing of total from healthy volunteers identified active pathways in uncultured species, such as dominated by and , revealing functional stratification without prior cultivation. For microbiomes, mRNA enrichment followed by has enabled functional profiling of uncultured and fungi, uncovering nutrient cycling genes like those for that are expressed under varying edaphic conditions. This approach highlights the metabolic contributions of rare or unculturable taxa, providing a snapshot of ecosystem-level . In environmental contexts, transcriptomics has decoded microbial responses to stressors, including pathogen-host interactions and climate-driven changes. During viral infections, dual RNA-seq of host-pathogen pairs has captured simultaneous transcriptomes, showing how bacterial pathogens like Staphylococcus aureus upregulate virulence factors in response to host immune signals, while viral transcripts reveal replication dynamics in microbial infections. Metatranscriptomic profiling of nasopharyngeal swabs in respiratory infections further demonstrated pathogen detection alongside host responses, identifying microbial shifts that exacerbate viral persistence. For climate change effects, meta-transcriptomics of algal blooms has shown upregulated toxin biosynthesis and photosynthesis genes in dinoflagellates like Alexandrium, correlating with warming temperatures that extend bloom durations. In Prorocentrum species, RNA-seq revealed adaptive shifts in stress response transcripts, linking ocean acidification to increased bloom toxicity. Recent advances as of 2025 include long-read , which improves of full-length transcripts in communities, and for . Long-read combined with has reconstructed metabolic networks in soil microbiomes, resolving isoform diversity and structures that short reads miss, thus enhancing functional of uncultured taxa. Tools like Fungen further enable error-corrected clustering of long-read data, achieving near-complete transcript recovery in diverse bacterial assemblages. In , bacterial single-cell methods such as BaSSSh-seq have profiled heterogeneity in S. aureus, identifying subpopulation-specific expression of and quorum-sensing genes that drive community persistence. Improved rRNA depletion protocols have boosted sensitivity, allowing transcriptome-wide mapping of translational rates in single cells. Case studies underscore these applications, such as the Project's integration of since 2012, which profiled gut community dynamics and linked microbial transcripts to host in and states. In the Integrative HMP, of body-site microbiomes revealed site-specific functional profiles, like in the gut. For adaptations, transcriptomic analysis of relic microbial mats in dry valleys showed upregulated cold-shock proteins and genes in and , enabling survival in subzero temperatures and low water availability. These mats exhibit nutrient-scavenging transcriptomes, reflecting evolutionary tweaks to extreme oligotrophy amid climate variability.

Integration with Other Omics

Relation to Proteomics

The transcriptome serves as a critical intermediary in the , bridging the and the by encoding (mRNA) transcripts that are translated into proteins. However, mRNA levels do not perfectly predict protein abundance due to extensive , with studies showing only moderate correlation, such as a Spearman's rank coefficient of approximately 0.46 in human cell lines. This imperfect correspondence arises because protein levels are influenced by factors beyond transcription, including efficiency and protein rates, which can account for the remaining variation in abundance. Key discrepancies between transcriptome and profiles stem from regulatory mechanisms like variable efficiency, where mRNA transcripts are translated at different rates based on sequence features such as (UTR) elements, and protein stability, governed by degradation pathways. In contrast, genes, essential for basal cellular functions, often display relatively stable mRNA and protein levels across conditions due to balanced transcription, , and turnover, ensuring consistent expression of proteins like GAPDH. To bridge these gaps, parallel technologies have emerged for direct measurement: (Ribo-seq) captures ribosome-protected mRNA fragments to quantify efficiency genome-wide, revealing active translation sites and regulatory elements like upstream open reading frames (uORFs). Complementarily, -based , such as liquid chromatography-tandem (LC-MS/MS), provides quantitative snapshots by ionizing and fragmenting peptides to identify and measure protein abundance and modifications. Integration of these datasets often employs correlation analyses, like Spearman's rank, to assess concordance, or advanced models such as frameworks (e.g., TransPro) that predict proteome profiles from transcriptomic inputs by learning regulatory patterns. Seminal efforts like the Human Proteome Project (HPP), initiated in 2010, have highlighted how expands diversity beyond transcriptomic predictions, with analyses of nearly 20,000 protein-coding genes revealing thousands of splice isoforms that contribute to tissue-specific protein variants and functional complexity. These studies underscore the transcriptome's role in informing but not fully determining the , emphasizing the need for joint analyses to uncover regulatory layers.

Relation to Genomics and Multiomics

The transcriptome represents the dynamic expression of the , revealing which portions of the genetic material are actively transcribed into under specific conditions, including pervasive transcription that extends beyond protein-coding genes to produce thousands of long non-coding RNAs and other transcripts from intergenic and intronic regions. This phenomenon highlights the genome's complexity, where much of the serves regulatory roles rather than direct protein synthesis, as evidenced by comprehensive RNA sequencing studies from the project showing that approximately 62% of the is transcribed at some level in at least one . Furthermore, (eQTLs) provide a direct link between genomic variants, such as single nucleotide polymorphisms (SNPs), and transcriptome regulation; these loci identify how genetic variations influence levels, bridging genome-wide association studies (GWAS) to molecular mechanisms of traits and diseases. For instance, cis-eQTLs often localize near target genes, modulating transcription initiation, while trans-eQTLs exert distal effects, underscoring the genome's role in shaping transcriptomic landscapes. In approaches, the transcriptome integrates with other layers to elucidate regulatory networks from to , such as combining transcriptomics with via ChIP-seq to map (TF) binding sites and modifications that drive . This integration reveals how epigenetic marks, like H3K27ac at enhancers, correlate with transcriptional activity, enabling the reconstruction of TF-gene interactions. Similarly, transcriptomics paired with uncovers downstream metabolic pathways influenced by , as seen in studies integrating with to trace how transcriptional changes in biosynthetic genes alter profiles in response to environmental stressors. Tools like Multi-Omics Factor Analysis (MOFA) facilitate this by performing across layers, identifying shared latent factors that explain variance in transcriptomic, epigenomic, and metabolomic data simultaneously. MOFA+ extends this to single-cell resolutions, handling sparse multi-modal datasets to infer cell-type-specific regulations. Key challenges in transcriptome-genomics and multiomics integration arise from data heterogeneity—due to varying technologies, resolutions, and scales—and high dimensionality, which complicates joint modeling and interpretation. Dimensionality reduction techniques, such as tensor decomposition, address this by representing multiomics data as higher-order tensors for joint analysis, decomposing them into low-rank factors that capture interactions across genomic, transcriptomic, and other layers while mitigating noise and missing values. For example, non-negative tensor factorization methods like MONTI select biologically relevant features from multiomics tensors, improving inference accuracy in cancer subtyping. Despite these advances, aligning datasets from different omics remains computationally intensive, often requiring normalization to handle batch effects and sparsity. Recent advances, particularly by 2025, leverage for integration, with models like scGPT enabling generative predictions across single-cell transcriptomes and other by pretraining on vast datasets to align modalities and infer regulatory dynamics. This facilitates applications in , such as simulating perturbation effects on networks. Conceptual frameworks for regulatory networks from to transcriptome rely on algorithms like GENIE3, which uses to predict TF-target interactions from expression data, outperforming earlier methods in benchmark challenges by prioritizing feature importance as regulatory strengths. These tools collectively advance holistic views of biological systems, linking static genomic blueprints to dynamic transcriptomic responses.

Databases and Resources

Major Transcriptome Databases

The (GEO), maintained by the (NCBI), serves as a primary public repository for high-throughput data, including and datasets, archiving raw and processed data since its inception in 2000. It hosts over 6.5 million samples (as of 2023) from over 200,000 diverse experiments, enabling researchers to access profiles across species, conditions, and platforms, with tools for querying by gene symbol, organism, or experimental factor. The (ENCODE) project, launched in 2003, provides comprehensive functional genomic annotations for the and genomes, incorporating transcriptome data such as profiles to map transcriptional units and regulatory elements. ENCODE datasets include processed expression values for protein-coding genes and support comparative analyses across species like , , and fly, with utilities for visualizing transcription in specific tissues or cell types. The Genotype-Tissue Expression (GTEx) project, initiated in 2015, offers a resource of genotype and transcriptome data from 948 postmortem donors, encompassing 19,788 samples across 54 non-diseased tissue sites to study tissue-specific (as of version 8, 2020). Its version 8 release in 2020 includes (eQTLs) for thousands of s, facilitating analyses of genetic variants' impact on expression in tissues like , adipose, and muscle. For organism-specific resources, the Arabidopsis Information Resource (TAIR) curates genomic and expression data for the model plant , including and profiles linked to gene functions, phenotypes, and metabolic pathways. Phytozome, a comparative platform from the Joint Genome Institute, aggregates transcriptome assemblies and expression data for 186 plant species, supporting evolutionary studies through analyses and co-expression networks. The database integrates co-expression networks derived from transcriptomic data across thousands of organisms, predicting functional associations between proteins based on similar expression patterns in tissues or conditions. These databases store diverse content types, such as raw FASTQ sequencing files, normalized expression counts, and rich detailing experimental conditions, sample sources, and sequencing platforms, with search functionalities allowing retrieval by identifier, type, or . By 2025, repositories like and collectively archive billions of sequencing reads from global experiments, underscoring their scale in supporting large-scale meta-analyses. Annotation resources complement these databases by providing standardized transcript models; Ensembl generates automated gene annotations, including splice variants and promoter regions, for over 4,800 eukaryotic and more than 31,300 prokaryotic genomes using aligned transcriptomic evidence (as of 2025). Similarly, NCBI's offers a curated, non-redundant collection of transcript sequences with functional annotations, ensuring consistency in mapping expression data to genomic coordinates.

Data Standards and Accessibility

Standardization efforts in transcriptome research began with the Minimum Information About a Experiment (MIAME) guidelines, proposed in 2001 to ensure the minimum data required for unambiguous interpretation and of microarray-based experiments. These guidelines were later extended to next-generation sequencing through the Minimum Information about a Next-generation Sequencing Experiment (MINSEQE), introduced in 2008, which specifies essential details such as experimental design, sample characteristics, and raw data processing for high-throughput sequencing data, including . Common file formats for raw transcriptome data include FASTQ for storing nucleotide sequences and quality scores from sequencing reads, and (Browser Extensible Data) for representing genomic intervals and annotations. The Sequence Read Archive () serves as a primary repository for archiving these raw sequencing files, enabling long-term preservation and public access to petabyte-scale datasets. Processed transcriptome data often utilizes count matrices to represent levels, commonly stored in efficient formats like HDF5 for handling large sparse matrices in single-cell analyses. Metadata accompanying these datasets follows schemas such as ISA-Tab, a tab-delimited framework for capturing experimental context across studies, including investigations, studies, and assays. Ontologies like EDAM further support interoperability by standardizing descriptions of bioinformatics operations, data types, and formats, facilitating tool integration and workflow automation in transcriptome analysis. Accessibility initiatives emphasize principles, notably the (Findable, Accessible, Interoperable, Reusable) guidelines established in 2016, which promote machine-readable and standardized protocols to enhance data reuse in life sciences, including transcriptomics. Public funding agencies have reinforced these through mandates like the NIH Public Access Policy of 2008, requiring deposition of peer-reviewed articles and associated data into public repositories within 12 months of publication to broaden access to federally funded research outputs. As of 2025, challenges in transcriptome data management include the exponential growth to petabyte-scale storage needs, with the NCBI SRA exceeding 47 petabytes as of late 2024, straining computational resources and requiring advanced compression and cloud-based solutions. Privacy concerns are particularly acute for single-cell transcriptome data, which can inadvertently reveal donor identities, necessitating compliance with regulations like the EU's GDPR through techniques such as data anonymization and federated learning frameworks. Tools like the Federated European Genome-phenome Archive (EGA) enable secure, distributed access without centralizing sensitive data, supporting collaborative analysis while upholding privacy. Looking ahead, emerging technologies such as are being explored to provide immutable tracking for transcriptome datasets, ensuring traceability of data origins and modifications in shared genomic platforms. Infrastructure like ELIXIR's integration will further promote seamless data exchange across nodes, standardizing access to multi-omics resources and fostering in future transcriptome studies.

References

  1. [1]
    Transcriptome Fact Sheet
    Aug 17, 2020 · A transcriptome is a collection of all the gene readouts present in a cell. What is a transcriptome? The human genome is made up of DNA ...
  2. [2]
    transcriptome | Learn Science at Scitable - Nature
    A transcriptome is the full range of messenger RNA, or mRNA, molecules expressed by an organism. The term transcriptome can also be used to describe the array ...
  3. [3]
    Transcriptomics technologies - PMC - NIH
    A transcriptome captures a snapshot in time of the total transcripts present in a cell. The first attempts to study the whole transcriptome began in the early ...
  4. [4]
    Transcriptomes and Proteomes - Genomes - NCBI Bookshelf - NIH
    The initial product of genome expression is the transcriptome, a collection of RNA molecules derived from those protein-coding genes whose biological ...
  5. [5]
    Transcriptome: Connecting the Genome to Gene Function - Nature
    Studying the transcriptome, RNA expressed from the genome, reveals a more complex picture of the gene expression behind it all.
  6. [6]
    The Human Transcriptome: An Unfinished Story - PMC
    Jun 29, 2012 · The transcriptome of a cell is the collection of all the RNA molecules, or transcripts, present in that cell. To generate the transcriptome, the ...<|control11|><|separator|>
  7. [7]
    Transcriptomics - Latest research and news - Nature
    Transcriptomics is the study of the transcriptome—the complete set of RNA transcripts that are produced by the genome, under specific circumstances or in a ...
  8. [8]
    Omics-Based Clinical Discovery: Science, Technology, and ... - NCBI
    The transcriptome is the complete set of RNA transcripts from DNA in a cell or tissue. The transcriptome includes ribosomal RNA (rRNA), messenger RNA (mRNA) ...
  9. [9]
    Understanding the Molecular Mechanisms of Asthma through ...
    The transcriptome represents the complete set of RNA transcripts that are produced by the genome under a specific circumstance or in a specific cell.
  10. [10]
    TPM, FPKM, or Normalized Counts? A Comparative Study of ... - NIH
    Jun 22, 2021 · TPM stands for transcript per million, and the sum of all TPM values is the same in all samples, such that a TPM value represents a relative ...
  11. [11]
    transcriptome, n. meanings, etymology and more
    The earliest known use of the noun transcriptome is in the 1990s. OED's earliest evidence for transcriptome is from 1997, in a text by V. E. Velculescu et al ...
  12. [12]
    Expressed Sequence Tags and Human Genome Project - Science
    Complementary DNA Sequencing: Expressed Sequence Tags and Human Genome Project. Mark D. Adams, Jenny M.
  13. [13]
    Serial Analysis of Gene Expression - Science
    A method was developed, called serial analysis of gene expression (SAGE), that allows the quantitative and simultaneous analysis of a large number of ...
  14. [14]
    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 ...
  15. [15]
    Mapping and quantifying mammalian transcriptomes by RNA-Seq
    May 30, 2008 · Mortazavi, A., Williams, B., McCue, K. et al. Mapping and quantifying mammalian transcriptomes by RNA-Seq. Nat Methods 5, 621–628 (2008).
  16. [16]
    Patrick O. Brown - Stanford Profiles
    Patrick O. Brown is part of Stanford Profiles ... We have developed and tested a method for printing protein microarrays and using these microarrays ...
  17. [17]
    Structure and mechanism of the RNA polymerase II transcription ...
    Apr 1, 2020 · In this review, we detail the structure and mechanism of over a dozen factors that govern Pol II initiation (eg, TFIID, TFIIH, and Mediator), pausing, and ...
  18. [18]
    RNA Transcription by RNA Polymerase: Prokaryotes vs Eukaryotes
    Transcription in Bacteria​​ Sigma factors are thus discriminatory, as each binds a distinct set of promoter sequences. A striking example of the specialization ...
  19. [19]
    Modification of enhancer chromatin: what, how and why? - PMC
    Histone acetylation at enhancers. The ability of TFs to activate transcription is dependent on the recruitment of coactivator proteins, many of which have ...
  20. [20]
    Genomic Analyses of Transcription Factor Binding, Histone ... - NIH
    The binding of signal-regulated transcription factors to regulatory sites (i.e., enhancers and silencers) across the genome ultimately regulates the ...
  21. [21]
    Knowing when to stop: Transcription termination on protein-coding ...
    Transcription can be divided into three phases: initiation, elongation, and termination (Figure 1). During transcription initiation, the preinitiation complex ...
  22. [22]
    A conserved minimum transcription level for essential genes - PMC
    We conclude that virtually all essential genes are transcribed at a rate of at least once per cell cycle. This analysis strongly supports the hypothesis that ...
  23. [23]
    Mechanism of alternative splicing and its regulation - PMC
    Alternative splicing of precursor mRNA is an essential mechanism to increase the complexity of gene expression, and it plays an important role in cellular ...
  24. [24]
    Long non-coding RNAs: definitions, functions, challenges ... - Nature
    Jan 3, 2023 · Many lncRNAs associate with chromatin-modifying complexes, are transcribed from enhancers and nucleate phase separation of nuclear condensates ...
  25. [25]
    From DNA to RNA - Molecular Biology of the Cell - NCBI Bookshelf
    Overall, RNA makes up a few percent of a cell's dry weight. Most of the RNA in cells is rRNA; mRNA comprises only 3–5% of the total RNA in a typical mammalian ...
  26. [26]
    Decoding the Non-coding: Tools and Databases Unveiling the ...
    There are different kinds of ncRNAs, such as rRNA, snRNA, tRNA, microRNA, lncRNA, circRNA, and piwiRNA. A plethora of novel transcripts determined by advanced ...
  27. [27]
    Integrating mRNA Processing with Transcription - ScienceDirect.com
    The messenger RNA processing reactions of capping, splicing, and polyadenylation occur cotranscriptionally. They not only influence one another's efficiency ...
  28. [28]
    Predicting the structural impact of human alternative splicing
    Sep 17, 2025 · In humans, although there are approximately 20,000 genes, the number of alternative splicing events can reach up to 100,000 [1, 2, 3].
  29. [29]
    Ribogenomics: the Science and Knowledge of RNA - ScienceDirect
    In a typical mammalian cell, mRNA takes ∼4% of the total RNA mass and aside from 80% ribosomal RNA (rRNA), other operational RNAs make up the rest. If we take ...
  30. [30]
    A brief review of noncoding RNA
    Sep 2, 2024 · Micro RNA (miRNA), small nuclear RNA (snRNA) and small nucleolar RNA (snoRNA) are other examples of this category of small ncRNA. Functions ...
  31. [31]
    Overview of MicroRNA Biogenesis, Mechanisms of Actions, and ...
    In general, the non-canonical miRNA biogenesis can be grouped into Drosha/DGCR8-independent and Dicer-independent pathways. Pre-miRNAs produced by the Drosha/ ...
  32. [32]
    The biogenesis, biology and characterization of circular RNAs - Nature
    Aug 8, 2019 · Circular RNAs (circRNAs) are covalently closed, endogenous biomolecules in eukaryotes with tissue-specific and cell-specific expression patterns.
  33. [33]
    DNA microarrays: Types, Applications and their future - PMC - NIH
    This chapter provides an overview of DNA microarrays. Microarrays are a technology in which 1000's of nucleic acids are bound to a surface and are used to ...Missing: workflow | Show results with:workflow
  34. [34]
    Microarrays | Functional genomics II - EMBL-EBI
    The hybridisation data are reported as a ratio of the Cy5/Cy3 fluorescent signals at each probe. By contrast, in one colour microarrays, each sample is labelled ...Missing: workflow | Show results with:workflow
  35. [35]
  36. [36]
    Strengths and limitations of laboratory procedures for microRNA ...
    First, the larger dynamic range of stem-loop qRT-PCR (7 logs vs. 3–4 logs for microarray) may provide greater sensitivity (27). qRT-PCR may also have higher ...
  37. [37]
    RNA-Seq: a revolutionary tool for transcriptomics - Nature
    RNA-Seq is a recently developed approach to transcriptome profiling that uses deep-sequencing technologies. Studies using this method have already altered ...Missing: workflow | Show results with:workflow
  38. [38]
    Comparison of RNA-Seq by poly (A) capture, ribosomal RNA ...
    Jun 2, 2014 · Our results demonstrate that compared to mRNA-Seq and microarrays, Ribo-Zero-Seq provides equivalent rRNA removal efficiency, coverage uniformity, genome-based ...
  39. [39]
    A survey of best practices for RNA-seq data analysis | Genome Biology
    Jan 26, 2016 · Mortazavi A, Williams BA, McCue K, Schaeffer L, Wold B. Mapping and quantifying mammalian transcriptomes by RNA-Seq. Nat Methods. 2008;5:1–8.
  40. [40]
    Full-length RNA-seq from single cells using Smart-seq2 - Nature
    Jan 2, 2014 · Here we present a detailed protocol for Smart-seq2 that allows the generation of full-length cDNA and sequencing libraries by using standard reagents.
  41. [41]
    The neXt generation of single cell RNA-seq: An introduction to GEM ...
    Mar 11, 2024 · Explore how Chromium GEM-X single cell RNA-sequencing can level up your research in developmental biology, cancer, neuroscience, ...Missing: seminal | Show results with:seminal
  42. [42]
    Embracing the dropouts in single-cell RNA-seq analysis - Nature
    Mar 3, 2020 · As a result of the dropouts, the scRNA-seq data is often highly sparse. The excessive zero counts cause the data to be zero-inflated, only ...
  43. [43]
    Visium Spatial Platform | 10x Genomics
    The Visium platform delivers unbiased, whole transcriptome spatial gene expression analysis at single cell scale with unmatched spatial data quality.Missing: 2019 | Show results with:2019
  44. [44]
    Spatially resolved, highly multiplexed RNA profiling in single cells
    We report multiplexed error-robust FISH (MERFISH), a single-molecule imaging method that allows thousands of RNA species to be imaged in single cells.
  45. [45]
    Single-cell in situ RNA profiling by sequential hybridization - Nature
    Mar 28, 2014 · In our previous paper, Lubeck and Cai, we used super-resolution microscopy to resolve a large number of mRNAs in single cells. In this ...
  46. [46]
    Long read single cell RNA sequencing reveals the isoform diversity ...
    Dec 16, 2022 · Here, we describe the sequencing of scRNA-seq libraries using Pacific Biosciences (PacBio) chemistry to characterize full-length Plasmodium ...
  47. [47]
    Simultaneous epitope and transcriptome measurement in single cells
    Jul 31, 2017 · Here, we describe cellular indexing of transcriptomes and epitopes by sequencing (CITE-seq), a method in which oligonucleotide-labeled ...
  48. [48]
    Eleven grand challenges in single-cell data science | Genome Biology
    Feb 7, 2020 · Challenge I: Handling sparsity in single-cell RNA sequencing. A comprehensive characterization of the transcriptional status of individual cells ...
  49. [49]
    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 ...
  50. [50]
    StringTie enables improved reconstruction of a transcriptome from ...
    Feb 18, 2015 · Grabherr, M.G. et al. Full-length transcriptome assembly from RNA-Seq data without a reference genome. Nat. Biotechnol. 29, 644–652 (2011).Missing: original | Show results with:original
  51. [51]
    A new view of transcriptome complexity and regulation through the ...
    Feb 1, 2016 · Unlike many tools, MAJIQ supplements annotated ... (2013) AVISPA: a web tool for the prediction and analysis of alternative splicing.
  52. [52]
    A machine learning approach for multimodal data fusion for survival ...
    May 6, 2025 · The TCGA project (2006–2018) molecularly characterized more than 20,000 primary cancer samples and matched normal samples, spanning 33 cancer ...
  53. [53]
    The landscape of kinase fusions in cancer | Nature Communications
    Sep 10, 2014 · We describe here a pan-cancer analysis of the transcriptomes of nearly 7,000 tumours from TCGA that is specifically focused on kinase gene ...
  54. [54]
    Single-cell multi-omics analysis of the immune response in COVID-19
    Apr 20, 2021 · We performed single-cell transcriptome, surface proteome and T and B lymphocyte antigen receptor analyses of over 780,000 peripheral blood ...
  55. [55]
    RNA Sequencing in Drug Discovery and Development - Biostate.ai
    Jul 28, 2025 · RNA sequencing expands pharmacogenomic analysis by measuring how genetic differences affect entire transcriptional networks involved in drug ...
  56. [56]
    Clinical utility of circulating tumor RNA (ctRNA) in a combined ...
    May 28, 2025 · This is the first large study showing that adding ctRNA to ctDNA liquid biopsy increases total actionable diagnostic yield by 36.7%.
  57. [57]
    Inference of differentiation trajectories by transfer learning across ...
    Dec 20, 2023 · Stem cells differentiate into distinct fates by transitioning through a series of transcriptional states. Current computational approaches ...
  58. [58]
    Single-cell multiregion dissection of Alzheimer's disease - Nature
    Jul 24, 2024 · Here we report a single-cell transcriptomic atlas of six different brain regions in the aged human brain, covering 1.3 million cells from 283 post-mortem human ...
  59. [59]
    Dissecting tumor microenvironment from spatially resolved ... - Nature
    Jun 13, 2024 · Published: 13 June 2024. Dissecting tumor microenvironment from spatially resolved transcriptomics data by heterogeneous graph learning.
  60. [60]
    Multiomics integration analysis identifies tumor cell-derived MIF as a ...
    Aug 22, 2025 · Multiomics integration analysis identifies tumor cell-derived MIF as a therapeutic target and potentiates anti-PD-1 therapy in osteosarcoma.
  61. [61]
    Breast cancer PAM50 signature: correlation and concordance ...
    Jun 3, 2019 · PAM50 was performed on both digital multiplexed gene expression and RNA-Seq platforms. Subtype assignment was based on the nearest centroid ...
  62. [62]
    Unprecedented High-Resolution View of Bacterial Operon ... - NIH
    Jul 8, 2014 · We analyzed the transcriptome of Escherichia coli K-12 by strand-specific RNA sequencing at single-nucleotide resolution during steady-state ...Missing: seminal papers
  63. [63]
    Novel small RNA-encoding genes in the intergenic regions of ...
    Here we report on the discovery of 14 genes encoding novel small RNAs in E. coli and their expression patterns under a variety of physiological conditions.Missing: seminal papers
  64. [64]
    The Escherichia coli transcriptome mostly consists of independently ...
    Dec 4, 2019 · Here, we apply unsupervised machine learning to a diverse compendium of over 250 high-quality Escherichia coli RNA-seq datasets to identify 92 ...
  65. [65]
    Comparative Analysis of Transcriptomic Response of Escherichia ...
    Feb 28, 2023 · In this work, a comprehensive comparative transcriptomic analysis on how Escherichia coli responds to nine representative classes of antibiotics
  66. [66]
    Plasticity and Stereotypic Rewiring of the Transcriptome Upon ...
    Jan 31, 2023 · Excessive use of antibiotics promotes the rapid evolution of resistant bacteria that eventually limit the clinical use of existing antibiotics.
  67. [67]
    Metatranscriptomic Approach to Analyze the Functional Human Gut ...
    Mar 8, 2011 · We performed a metatranscriptomic study on ten healthy volunteers to elucidate the active members of the gut microbiome and their functionality ...
  68. [68]
    (PDF) Soil microbial ecology through the lens of metatranscriptomics
    Oct 21, 2025 · Metatranscriptomics is a cutting-edge technology for exploring the gene expression by, and functional activities of, the microbial community ...
  69. [69]
    Functional Profiling of Unfamiliar Microbial Communities Using ... - NIH
    Jan 12, 2016 · Metatranscriptomic landscapes can provide insights in functional relationships within natural microbial communities.Missing: uncultured | Show results with:uncultured
  70. [70]
    Spatial Transcriptomics to Study Virus-Host Interactions
    Sep 25, 2025 · Understanding how viruses cause disease and identifying the viral and host factors that determine the outcome of infection are essential to ...
  71. [71]
    Metatranscriptomic profiling reveals pathogen and host response ...
    Mar 17, 2025 · Through RNA-seq analysis of NP swab samples, we performed metatranscriptomic pathogen detection and assessed its ability to reproduce culture ...
  72. [72]
    Species specific gene expression dynamics during harmful algal ...
    Apr 10, 2020 · During three consecutive years, the meta-transcriptome of micro-eukaryote communities was sequenced during blooms of the toxic dinoflagellate ...
  73. [73]
    Transcriptome analysis of two bloom-forming Prorocentrum species ...
    Harmful algal blooms (HABs) have become more prevalent worldwide due to climate change, threatening ecosystem balance and public health (Anderson et al ...
  74. [74]
    Metatranscriptomics-guided genome-scale metabolic reconstruction ...
    Jul 5, 2024 · In this study, we combined long-read sequencing and metatranscriptomics-guided metabolic reconstruction to provide a genome-wide perspective of carbon ...
  75. [75]
    Fungen: clustering and correcting long-read metatranscriptomic data ...
    Jun 19, 2025 · We present Fungen, a reference-free tool that constructs accurate transcripts from long-read metatranscriptomic data through read clustering and error ...<|separator|>
  76. [76]
    Bacterial single-cell RNA sequencing captures biofilm ... - Nature
    Nov 24, 2024 · Here, we present an optimized bacterial single-cell RNA sequencing method, BaSSSh-seq, to study Staphylococcus aureus diversity during biofilm ...
  77. [77]
    An improved bacterial single-cell RNA-seq reveals biofilm ... - eLife
    Dec 17, 2024 · This work introduces an important new method for depleting ribosomal RNA from bacterial single-cell RNA sequencing libraries, demonstrating its applicability ...
  78. [78]
    Metatranscriptomics for the Human Microbiome and Microbial ...
    Jul 20, 2021 · Shotgun metatranscriptomics (MTX) is an increasingly practical way to survey microbial community gene function and regulation at scale.
  79. [79]
    The Integrative Human Microbiome Project - Nature
    May 29, 2019 · The HMP1 focused on the characterization of microbial communities from numerous body sites (oral, nasal, vaginal, gut, and skin) in a baseline ...
  80. [80]
    Antarctic Relic Microbial Mat Community Revealed by ... - Frontiers
    Jan 22, 2019 · Our transcriptome data suggests that the organisms in the community are adept at taking up nutrients from their environment. In addition to ...
  81. [81]
    Comprehensive insights on environmental adaptation strategies in ...
    The review illustrates the different adaptation strategies of Antarctic microbes to changing climate factors at the structural, physiological and molecular ...
  82. [82]
    Sequence signatures and mRNA concentration can explain two ...
    Aug 24, 2010 · We observe a significant positive correlation between mRNA and protein concentrations (Figure 2; Spearman's rank correlation Rs=0.46 (P-value<2E ...
  83. [83]
    Evaluation of Protein Expression in Housekeeping Genes across ...
    In protein expression studies, it is important to identify housekeeping genes that show comparatively stable expression patterns across body tissues. Expression ...
  84. [84]
    Plasma proteomics, the Human Proteome Project, and cancer ...
    Nov 8, 2013 · Among the isoforms of proteins, splice variants have the special feature of greatly enlarging protein diversity without enlarging the genome; ...
  85. [85]
    The Reality of Pervasive Transcription - PMC - NIH
    Jul 12, 2011 · Abstract. Despite recent controversies, the evidence that the majority of the human genome is transcribed into RNA remains strong.Missing: seminal | Show results with:seminal
  86. [86]
    Pervasive transcription constitutes a new level of eukaryotic genome ...
    Pervasive transcription is widespread and, far from being a futile process, has a crucial role in controlling gene expression and genomic plasticity.Missing: beyond seminal
  87. [87]
    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 ...
  88. [88]
    Single-cell eQTL mapping identifies cell type–specific genetic ...
    Apr 8, 2022 · These studies examined both proximal (cis) and distal (trans) genetic variants affecting gene expression in 14 different immune cell types.
  89. [89]
    Role of ChIP-seq in the discovery of transcription factor binding sites ...
    This review addresses the important applications of ChIP-seq with an emphasis on its role in genome-wide mapping of transcription factor binding sites.Missing: epigenomics transcriptome
  90. [90]
    Quantitative integration of epigenomic variation and transcription ...
    Jul 10, 2018 · It can automatically perform quantitative comparison between ChIP-seq samples of the same protein but from different cell types, and identify ...Missing: epigenomics | Show results with:epigenomics
  91. [91]
    Metabolomics and transcriptomics based multi-omics integration ...
    Sep 9, 2023 · The integrated-omics approach will allow simultaneous analysis of mRNAs and metabolites providing whole transcriptome and metabolic changes ...
  92. [92]
    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 ...
  93. [93]
    MOFA+: a statistical framework for comprehensive integration of ...
    May 11, 2020 · We present Multi-Omics Factor Analysis v2 (MOFA+), a statistical framework for the comprehensive and scalable integration of single-cell multi-modal data.
  94. [94]
    Navigating Challenges and Opportunities in Multi-Omics Integration ...
    This review evaluates these challenges while spotlighting pivotal milestones: the development of targeted sampling methods, the use of artificial intelligence.
  95. [95]
    Gene-set integrative analysis of multi-omics data using tensor-based ...
    We introduce a tensor-based framework for variable-wise inference in multi-omics analysis. By accounting for the matrix structure of an individual's multi- ...2.3 Simulation Studies · 3 Results · 3.1 Simulation Studies
  96. [96]
    MONTI: A Multi-Omics Non-negative Tensor Decomposition ...
    Sep 9, 2021 · We propose a multi-omics analysis method called MONTI (Multi-Omics Non-negative Tensor decomposition for Integrative analysis), which goal is to select multi- ...Abstract · Introduction · Materials and Methods · Results
  97. [97]
    A technical review of multi-omics data integration methods
    Aug 1, 2025 · However, multi-omics data integration remains challenging due to the high-dimensionality, heterogeneity, and frequency of missing values across ...
  98. [98]
    Inferring Regulatory Networks from Expression Data Using Tree ...
    In this article, we present GENIE3, a new algorithm for the inference of GRNs that was best performer in the DREAM4 In Silico Multifactorial challenge.
  99. [99]
    Gene Expression Omnibus - NCBI - NIH
    Gene Expression Omnibus (GEO) is a database repository of high throughput gene expression data and hybridization arrays, chips, microarrays.Submitting data · Login to Submit · Download GEO data · About GEO DataSets
  100. [100]
    GEO Overview - NCBI
    Jul 16, 2024 · Gene Expression Omnibus (GEO) is a database repository of high throughput gene expression data and hybridization arrays, chips, microarrays.
  101. [101]
    ENCODE project
    Experiment search · Experiment matrix · ChIP-seq matrix · Human and mouse body maps · Functional genomics series · Single-cell experiments.Getting Started · Data release policy · Project Overview · Citing ENCODEMissing: 2007 | Show results with:2007
  102. [102]
    Transcriptome - ENCODE
    These tables provide a summary of all annotations with processed expression values associated to protein coding genes in human, worm, and fly.
  103. [103]
    Genotype-Tissue Expression (GTEx) Portal
    Oct 18, 2017 · The Adult GTEx project is a comprehensive resource of WGS, RNA-Seq, and QTL data from samples collected from 54 non-diseased tissue sites across ...
  104. [104]
  105. [105]
    TAIR - Home
    The Arabidopsis Information Resource (TAIR) maintains a database of genetic and molecular biology data for the model higher plant Arabidopsis thaliana.TAIR Login Page · Sequence Viewer · GO Term Enrichment · Subscription
  106. [106]
    Phytozome
    Search and visualization tools let users quickly find and analyze genes or genomic regions of interest. Query-based data access is provided by Phytozome's ...BioMart · Glycine max Wm82.a6.v1 · Glycine max Wm82.a2.v1 · O.sativa v7.0
  107. [107]
    STRING: functional protein association networks
    STRING. Protein-Protein Interaction Networks, Functional Enrichment Analysis. Featuring detailed information on 12,535 organisms. Includes 59.3 million ...
  108. [108]
    NCBI GEO: archive for gene expression and epigenomics data sets
    Nov 2, 2023 · The Gene Expression Omnibus (GEO) is an international public repository that archives gene expression and epigenomics data sets generated by ...
  109. [109]
    Gene annotation in Ensembl
    Gene annotation provided by Ensembl includes automatic annotation, ie genome-wide determination of transcripts. For selected species (ie human, mouse, zebrafish ...Ensembl Canonical transcriptManual gene annotation by ...
  110. [110]
    RefSeq: NCBI Reference Sequence Database - NIH
    A comprehensive, integrated, non-redundant, well-annotated set of reference sequences including genomic, transcript, and protein.About RefSeq · RefSeqGene · Prokaryotic RefSeq Genomes · RefSeq Select
  111. [111]
    Minimum information about a microarray experiment (MIAME) - Nature
    Dec 1, 2001 · Here we present a proposal, the Minimum Information About a Microarray Experiment (MIAME), that describes the minimum information required to ensure that ...
  112. [112]
    MINSEQE - FGED Society
    MINSEQE describes the Minimum Information about a high-throughput nucleotide SEQuencing Experiment that is needed to enable the unambiguous interpretation.Missing: original | Show results with:original
  113. [113]
    ISA software suite: supporting standards-compliant experimental ...
    ISA software suite: supporting standards-compliant experimental annotation and enabling curation at the community level Open Access. Philippe Rocca-Serra,.
  114. [114]
    The FAIR Guiding Principles for scientific data management ... - Nature
    Mar 15, 2016 · The FAIR Principles put specific emphasis on enhancing the ability of machines to automatically find and use the data, in addition to supporting its reuse by ...
  115. [115]
    NOT-OD-08-033: Revised Policy on Enhancing Public Access to ...
    Jan 11, 2008 · The NIH Public Access Policy applies to all peer-reviewed articles that arise, in whole or in part, from direct costs funded by NIH, or from NIH ...Missing: original | Show results with:original
  116. [116]
    [PDF] Council of Councils Working Group on Sequence Read Archive Data
    The SRA continues to experience exponential growth in submission rates, and the normalized format data is projected to grow to 57.5 petabytes by 2025; at this ...Missing: volume | Show results with:volume<|control11|><|separator|>
  117. [117]
    FedscGen: privacy-preserving federated batch effect correction of ...
    Jul 22, 2025 · Single-cell RNA-seq data from clinical samples often suffer from batch effects, but data sharing is limited due to genomic privacy concerns.
  118. [118]
    Federated EGA - European Genome-Phenome Archive
    Overview. Federated EGA is a global network of repositories enabling secure discovery and access to sensitive human data.Missing: EDA | Show results with:EDA<|control11|><|separator|>
  119. [119]
    Storing and analyzing a genome on a blockchain
    Jun 29, 2022 · In this study, we present the first open-source, proof-of-concept private blockchain network, which allows efficient storage and retrieval of ...