Fact-checked by Grok 2 weeks ago

Functional genomics

Functional genomics is a branch of that employs genome-wide approaches to elucidate the functions and interactions of genes, proteins, and other genomic elements, bridging the gap between genome sequencing and phenotypic outcomes. Unlike structural genomics, which focuses on mapping and sequencing , functional genomics investigates how these elements operate within biological systems to influence processes such as , , and environmental responses. The field emerged prominently following the completion of the in 2001, which provided comprehensive sequence data for the and enabled high-throughput analyses across species. Key goals include identifying patterns, protein interactions, and regulatory networks to model cellular dynamics and predict phenotypic variations. For instance, it seeks to determine how genetic variations contribute to traits like virulence in pathogens or susceptibility to diseases in hosts. Central techniques in functional genomics encompass transcriptomics (e.g., RNA sequencing to measure ), (e.g., for protein identification and quantification), and (e.g., analysis of and modifications). Advances in next-generation sequencing (NGS) technologies, such as Illumina platforms, have dramatically reduced sequencing costs—to approximately $0.09 per megabase by 2012—and facilitated applications like ChIP-Seq for mapping protein-DNA interactions and single-cell sequencing for resolving cellular heterogeneity. Other methods include microarrays for genome-wide expression profiling and CRISPR-Cas9 editing to validate functional roles of specific . In , functional genomics drives precision medicine by linking genomic variants to clinical outcomes, such as early detection of cancer through RNA profiling or identification of drug targets in antibiotic resistance. It has revealed insights into complex diseases like systemic lupus erythematosus (SLE) and juvenile rheumatoid arthritis (JRA) via differential in affected tissues. Ongoing challenges include integrating multi-omics data for comprehensive network models and addressing ethnic diversity in studies to enhance applicability across populations. As sequencing costs have declined to approximately $600 per (NHGRI data as of 2023), with some providers approaching $200 as of 2025, functional genomics is poised to transform diagnostics, therapeutics, and personalized healthcare.

Introduction

Definition

Functional genomics is the systematic study of the genome's dynamic functions, encompassing , , interactions among genomic elements, and their effects on phenotypes, often through high-throughput experimental and computational methods. Unlike structural genomics, which focuses on determining DNA sequences and physical genome maps, functional genomics seeks to elucidate how these sequences operate in biological contexts to influence cellular processes and organismal traits. This field integrates diverse disciplines, including and bioinformatics, to analyze the collective behavior of genes and their products on a genome-wide scale. A key distinction of functional genomics lies in its emphasis on comprehensive, large-scale investigations rather than the targeted analysis of individual genes typical of . It addresses the functional consequences of genomic variations, such as how mutations or environmental factors alter gene activity across entire genomes. Central to this approach are concepts like functional elements—non-coding DNA regions such as enhancers and promoters that regulate —and subfields including transcriptomics (which profiles transcripts to reveal expression patterns), (which examines protein abundance, modifications, and interactions), and (which explores heritable changes like and modifications without altering the DNA ). These components collectively provide insights into the regulatory networks and molecular mechanisms underlying biological diversity and disease. The term "functional genomics" emerged in the late 1990s, shortly following the initiation of the , to describe the next phase of genomic research aimed at interpreting the functional implications of sequenced genomes rather than merely cataloging them. This shift was driven by the need to move beyond static sequence data toward understanding dynamic genomic processes, marking a pivotal transition in post-sequencing . Seminal early discussions, such as those by Hieter and Boguski, highlighted how bioinformatics and experimental tools would enable this genome-wide functional annotation.

Historical Development

The foundations of functional genomics trace back to the 1980s, when systematic genetic studies in model organisms like the yeast Saccharomyces cerevisiae began to elucidate gene functions on a genome-wide scale. Early efforts, such as the yeast genome sequencing project initiated in 1989 under the leadership of international consortia, integrated classical mutagenesis with mapping techniques to assign functions to thousands of genes, laying the groundwork for high-throughput functional analysis. These studies shifted research from individual gene characterization to coordinated, large-scale approaches, exemplified by the identification of essential genes through systematic knockouts in the 1990s. The term "functional genomics" was first formally introduced in 1997, marking the formal emergence of the field as a distinct discipline focused on decoding gene functions using genome-scale tools. This coincided with the invention of DNA microarrays by Patrick Brown and colleagues in 1995, which enabled the simultaneous measurement of thousands of gene expression levels, revolutionizing the study of transcriptional responses. The draft sequence of the Human Genome Project was announced in 2000, with full completion in 2003 led by figures like Eric Lander who directed major sequencing efforts at the Whitehead Institute, accelerating the field by providing complete reference genomes and prompting a transition from sequence generation to functional annotation. In the same year, the ENCODE (Encyclopedia of DNA Elements) project was launched by the National Human Genome Research Institute to systematically map functional elements across the human genome, emphasizing data-driven strategies over traditional hypothesis testing. The 2010s saw the rise of next-generation sequencing (NGS) technologies, which democratized genome-wide functional studies by enabling cost-effective RNA sequencing and epigenomic profiling, further integrating omics data layers. The 2012 discovery of the -Cas9 system by and provided a precise tool for , facilitating large-scale perturbation screens to link genotypes to phenotypes across cell types. In the , advances in single-cell and spatial functional assays, such as single-cell screens combined with multi-omics integration, have enabled the dissection of cellular heterogeneity and tissue-level functions, with notable progress in mapping gene regulatory networks using base editors. These developments, including AI-assisted multi-omics frameworks for predictive modeling, continue to propel the field toward comprehensive systems-level understanding as of 2025.

Goals and Applications

Primary Objectives

Functional genomics seeks to elucidate the functions of genes and their regulatory elements across the genome, bridging the gap between genomic sequence data and observable biological phenotypes. A core objective is to determine the precise roles of the approximately 19,433 protein-coding genes in the human genome, as annotated by the GENCODE project in 2025, by integrating high-throughput experimental data to assign biological functions to these loci. This involves systematically characterizing how genetic variants influence gene expression and protein activity, thereby linking genotypes to phenotypes such as disease susceptibility. Another primary aim is to map regulatory elements, which constitute critical non-coding components of the genome—estimated at 98% of the total sequence—to identify enhancers, promoters, and silencers that control gene regulation. The Encyclopedia of DNA Elements (ENCODE) project exemplifies this goal by aiming to delineate all functional elements encoded in the human genome through biochemical assays and computational integration. Understanding genetic interactions represents a foundational objective, focusing on epistatic effects where the function of one modifies the impact of another, often revealed through network analyses that uncover compensatory or synergistic relationships. This approach addresses the complexity of polygenic traits by constructing interaction maps, such as networks, to predict how mutations propagate through biological pathways. Additionally, functional genomics prioritizes predicting the effects of genetic variants on , particularly in non-coding regions where most variants reside, to interpret their regulatory consequences and inform precision . These efforts tackle longstanding challenges in translating raw sequence information into mechanistic insights, as the majority of genomic variation occurs outside protein-coding exons and requires context-specific functional assays to assess impact. Metrics of success in functional genomics include the completeness of gene annotations, measured by the proportion of loci with assigned functions via resources like (eQTLs), which quantify how genetic variants influence transcript levels across tissues. For instance, projects such as the Genotype-Tissue Expression (GTEx) initiative have mapped over 500,000 eQTLs to enhance regulatory annotation, providing benchmarks for evaluating how well functional data covers the 's ~20,000 genes and their interactions. networks further serve as indicators, with completeness gauged by the density of detected interactions relative to expected genomic complexity, ensuring comprehensive coverage of regulatory landscapes. These quantitative frameworks underscore progress in annotating the non-coding , where functional validation remains a 2025 priority to resolve ambiguities in variant pathogenicity.

Biomedical and Industrial Applications

Functional genomics plays a pivotal role in biomedical applications by facilitating the identification of disease-causing genes and mutations. In , functional screens have pinpointed driver mutations that promote tumor growth and resistance to therapies; for instance, genome-wide CRISPR-based screens in cell lines revealed vulnerabilities in key drivers like PIK3CA and TP53, enabling targeted therapeutic strategies. These approaches extend to , where high-throughput functional assays evaluate the impact of genetic variants of unknown significance (VUS) on protein function, aiding clinical decision-making for patient-specific treatments such as in hereditary cancers. A notable 2025 advancement involved integrating functional genomics with analysis to subtype , identifying prognostic biological categories based on immune cell interactions and patterns that predict patient outcomes and guide . In industrial contexts, functional genomics drives enhancements in and . For improvement, studies on have cloned and characterized genes regulating seed size, weight, and pod number, leading to varieties with up to 10-15% higher yields through marker-assisted breeding and gene editing. In for biofuels, functional genomic platforms have identified and optimized lignocellulose-degrading enzymes from microbial consortia, improving conversion efficiency in engineered strains for production by reducing enzymatic costs and increasing release yields. These applications underscore the translation of genotype-phenotype linkages into practical outcomes, such as resilient s adapted to environmental stresses. Emerging developments in functional genomics address complex challenges like non-coding variants and computational integration. Functional phenotyping assays have tested thousands of osteoarthritis-associated non-coding variants, revealing regulatory effects on cartilage genes like GDF5, where variants alter enhancer activity and contribute to disease risk through altered expression in joint tissues. Additionally, deep learning models applied to gene signatures have improved predictions of compound-target interactions in drug discovery. By 2025, these efforts have culminated in impactful metrics, including the approval of multiple CRISPR-edited therapies like CASGEVY for and ongoing trials for cardiovascular conditions, demonstrating clinical efficacy with sustained gene correction rates above 80%. In precision agriculture, functional genomics contributes to yield gains and resource optimization in major crops like .

Techniques by Molecular Level

DNA-Level Techniques

DNA-level techniques in functional genomics focus on probing the static structure and regulatory potential of the by analyzing sequence variations, protein associations, and chromatin accessibility. These methods reveal how DNA elements, such as promoters, enhancers, and insulators, contribute to without directly measuring transcription or . By mapping interactions and accessible regions, researchers infer functional roles of and genetic dependencies that underlie cellular phenotypes. Genetic interaction mapping elucidates how genes function within networks by assessing the combined effects of perturbations, particularly through epistasis networks constructed via double knockouts. Epistasis occurs when the phenotypic effect of mutating one gene depends on the mutation in another, deviating from expected additive outcomes and highlighting pathway redundancies or dependencies. Synthetic lethality, a specific form of negative epistasis, arises when simultaneous inactivation of two genes is lethal, while individual knockouts are viable; this concept has been foundational in yeast studies and extended to human cells to identify therapeutic targets in cancer, where tumor suppressors create vulnerabilities exploitable by drugs. Double knockout screens, often using CRISPR-Cas9 libraries targeting gene pairs, generate comprehensive maps; for instance, a systematic CRISPR screen across 27 cancer cell lines analyzed 472 predicted synthetic lethal gene pairs, identifying 117 such pairs, revealing conserved interactions across melanoma and pancreatic cancers that inform precision oncology. These networks prioritize highly connected genes in solute carrier families, underscoring their roles in cellular homeostasis. Chromatin immunoprecipitation followed by sequencing (ChIP-seq) is a cornerstone for mapping DNA-protein interactions, particularly transcription factor (TF) binding sites that dictate regulatory logic. The protocol involves crosslinking proteins to DNA, fragmenting chromatin via sonication, immunoprecipitating with TF-specific antibodies, and sequencing the enriched DNA fragments to identify binding peaks genome-wide. Seminal work in 2007 demonstrated ChIP-seq's superiority over arrays by profiling STAT1 binding in response to interferon, achieving single-base resolution and detecting over 40,000 sites with low background. Data interpretation relies on peak calling algorithms like MACS, which model enrichment against input controls to distinguish true bindings, followed by motif discovery using tools such as MEME to validate TF specificities and infer co-binding partners. In practice, ChIP-seq has mapped bindings for hundreds of TFs across ENCODE cell types, revealing cell-specific patterns; for example, in embryonic stem cells, it identified Oct4 and Nanog sites enriched at promoters and enhancers, linking them to pluripotency maintenance. Challenges include antibody quality and indirect bindings, addressed by orthogonal validation like reporter assays. DNA accessibility assays, such as DNase-seq and , identify open regions where regulatory elements like enhancers and insulators are exposed for TF access. DNase I hypersensitive sites (DHSs) mark these areas due to DNase I's preferential cleavage of unprotected DNA; the original high-resolution DNase-seq protocol used deep sequencing of digested fragments from primary CD4+ T cells to map over 100,000 DHSs, correlating them with active genes and distal elements. , introduced in 2013, offers a simpler, low-input alternative by employing hyperactive Tn5 to simultaneously fragment and tag accessible DNA in intact nuclei, enabling profiling from as few as 500 cells. Both methods detect enhancers as broad peaks in intergenic regions and insulators as boundary elements preventing ectopic interactions; for instance, in identified cis-regulatory modules at promoters and enhancers, facilitating trait-associated variant prioritization. Interpretation involves aligning reads, calling peaks with tools like , and integrating with epigenomic data to annotate functional elements, though biases from sequence preferences require normalization. Recent advances incorporate in vivo CRISPR screens and precise editing to perturb DNA for studying complex traits. In vivo CRISPR screens, adapted for whole organisms, map genetic contributions to polygenic phenotypes; a 2025 review highlights their use in mice to dissect neural circuits, identifying epistatic interactions in behavior via pooled libraries delivered by AAV vectors. Base editing and prime editing extend these by enabling single-nucleotide changes without double-strand breaks, ideal for modeling disease variants. Base editors fuse deaminases to Cas9 nickases for C-to-T or A-to-G transitions, while prime editors use reverse transcriptase for arbitrary insertions/deletions up to 44 bp; 2025 innovations reduced off-target errors up to 14-fold in prime editing, enhancing functional perturbation of enhancers in vivo for trait engineering. These tools address limitations of traditional knockouts by preserving regulatory contexts, as seen in screens linking prime-edited variants to metabolic traits in organoids.

RNA-Level Techniques

RNA-level techniques in functional genomics focus on quantifying and perturbing RNA transcripts to elucidate gene expression dynamics, regulatory mechanisms, and cellular responses. These methods enable the measurement of transcript abundance, alternative splicing, and isoform diversity, providing insights into how transcriptional outputs are modulated without directly assessing genomic DNA sequences. By capturing the transcribed products of genes, such approaches reveal post-transcriptional regulation and environmental influences on expression patterns. Early hybridization-based methods laid the foundation for high-throughput RNA analysis. DNA microarrays involve immobilizing thousands of gene-specific probes on a solid surface, followed by hybridization with labeled (cDNA) derived from cellular RNA, allowing relative quantification of transcript levels across samples. This technique, introduced in the mid-1990s, facilitated genome-wide expression profiling by detecting fluorescence intensity proportional to mRNA abundance, though it is limited to predefined sequences and prone to cross-hybridization artifacts. (SAGE) complements microarrays by generating short sequence tags from cDNA, concatenating them for efficient sequencing, and counting tag frequencies to infer transcript levels without relying on prior sequence knowledge. Developed concurrently, SAGE provides a digital measure of expression and is particularly useful for discovering novel transcripts, albeit requiring more complex library preparation. RNA sequencing (RNA-seq) has revolutionized transcriptomics through next-generation sequencing technologies, enabling unbiased, high-resolution profiling of the entire . In RNA-seq, is converted to cDNA, fragmented, and sequenced to count reads aligning to genes, yielding quantitative measures of transcript abundance via reads per kilobase million (RPKM) or fragments per kilobase million (FPKM) normalization. This method, first demonstrated in mammalian systems in 2008, surpasses microarrays by detecting low-abundance transcripts, quantifying isoforms through junction reads, and identifying novel genes or non-coding RNAs. Single-cell RNA-seq (scRNA-seq) extends this to individual cells, isolating polyadenylated via barcoding and amplification before sequencing, thus resolving heterogeneity in cell populations such as during or progression. Pioneered in 2009, scRNA-seq has scaled to thousands of cells per experiment, revealing rare subpopulations and dynamic expression states. Reporter assays at the RNA level test cis-regulatory elements by linking candidate sequences to reporter genes, whose transcribed output is measured to assess regulatory strength. reporter assays (MPRAs) transfect pools of barcoded reporter constructs into cells, followed by to quantify barcode abundance in transcripts relative to input DNA, enabling simultaneous evaluation of thousands of enhancers or variants for their impact on expression. This approach, refined in mammalian systems by the mid-2010s, has identified functional non-coding variants associated with traits like and levels. Self-transcribing active regulatory sequencing (STARR-seq) innovates by using the candidate enhancer itself as the transcribed element downstream of a minimal promoter, capturing enhancer-driven RNA via sequencing to directly measure activity. Introduced in 2013 for and adapted to mammals, STARR-seq distinguishes active enhancers from poised ones and scales to genome-wide libraries, revealing tissue-specific regulatory landscapes. Perturb-seq integrates RNA-level readout with genetic perturbations, combining CRISPR guides or RNAi with scRNA-seq to link individual perturbations to transcriptomic changes in single cells. By barcoding perturbations and cells, this dissects regulatory networks, identifying downstream targets and interactions in pooled screens, as demonstrated in immune cells where it uncovered T cell differentiation pathways. Originally developed in 2016, Perturb-seq has evolved into multimodal variants by 2025, incorporating chromatin accessibility or protein profiling alongside to capture joint multi-omics effects of perturbations, enhancing resolution of regulatory mechanisms in complex tissues.

Protein-Level Techniques

Protein-level techniques in functional genomics focus on elucidating the functions, interactions, and structures of proteins at a genome-wide scale, providing insights into how genetic information is translated into cellular phenotypes. These methods complement RNA-level analyses by directly probing the , revealing post-translational modifications, complex formations, and variant effects that transcripts alone cannot capture. Key approaches include interaction mapping, complex identification, and high-throughput variant assessment, enabling the construction of protein networks and landscapes essential for understanding biological processes and mechanisms. The yeast two-hybrid (Y2H) system is a foundational binary assay for detecting protein-protein interactions (PPIs) in vivo. Developed in 1989, it leverages the modular nature of transcription factors by fusing a "bait" protein to a DNA-binding domain and a "prey" protein to an activation domain; if the bait and prey interact, they reconstitute a functional transcription factor, activating reporter genes such as those encoding selectable markers or colorimetric outputs. This method has been scaled to genome-wide screens, identifying thousands of PPIs in yeast and adapted for mammalian systems, though it can suffer from false positives due to non-specific activation or false negatives from improper protein folding in the yeast nucleus. Y2H excels in mapping binary interactions but is less suited for detecting transient or multi-subunit complexes. Mass spectrometry (MS)-based methods, particularly affinity purification followed by MS (AP/MS), enable the identification and quantification of protein complexes and interaction networks with high sensitivity and throughput. In AP/MS, a bait protein tagged with an affinity handle (e.g., FLAG or HA epitope) is expressed in cells, purified along with its binding partners using immunoprecipitation, and analyzed by liquid chromatography-tandem MS (LC-MS/MS) to identify co-purified proteins. Quantitative variants, such as stable isotope labeling by amino acids in cell culture (SILAC), distinguish specific interactors from background contaminants by comparing bait versus control purifications, yielding interaction scores that inform network topology. AP/MS has mapped over 20,000 human PPIs, revealing dynamic complexes in signaling pathways and disease contexts, with recent advances in instrumentation achieving proteome coverage in minutes. Deep mutational scanning (DMS) provides a high-throughput framework for mapping protein fitness landscapes by generating comprehensive variant libraries and quantifying their functional impacts. Typically, DMS involves to create all possible single or multiple substitutions in a protein of interest, followed by expression in cells or , selection for (e.g., via or ), and deep sequencing to count variant abundances before and after selection. This yields per-residue tolerance scores, highlighting functionally critical sites like active centers or interfaces, as demonstrated in studies of enzymes like TEM-1 where DMS revealed epistatic interactions across the sequence space. DMS has been applied to hundreds of human proteins, informing variant pathogenicity in diseases such as cancer and guiding , with models now integrating DMS data to predict unseen mutations. Recent advances in 2025 have integrated biophysical mapping with cell maps to link protein structures to functions at subcellular resolution. These maps combine AP/MS-derived data with imaging and other modalities (e.g., ) to construct comprehensive atlases of protein localization and assembly in human cells, revealing 104 novel protein assemblies and their contextual dependencies. Such approaches address gaps in structural-functional by enabling predictive modeling of complex disruptions in pathologies like neurodegeneration, where integration outperforms single-omics methods in resolving dynamic interactomes.

Perturbation and Screening Methods

Genetic Knockouts and Mutagenesis

Genetic knockouts and mutagenesis are foundational techniques in functional genomics for disrupting function to elucidate roles through loss-of-function phenotypes. These methods enable researchers to create targeted or random alterations in the , allowing observation of resulting cellular, physiological, or developmental changes in model organisms. By systematically inactivating genes, scientists can infer their contributions to biological processes, pathways, and states. Gene knockouts, particularly through , involve the precise replacement or deletion of a target in the of model organisms such as mice, rats, , and plants. This technique relies on the cell's natural machinery to integrate a modified DNA sequence—often containing a like neomycin resistance—into the endogenous locus via arms flanking the modification. In embryonic stem (ES) cells of mice, for instance, homologous recombination achieves targeted insertions at efficiencies of approximately 1 in 10^6 cells, followed by selection and injection into blastocysts to generate chimeric founders. Phenotypic readouts from these knockouts include embryonic lethality, morphological defects, or behavioral alterations, providing direct evidence of gene essentiality or function; for example, knockout of the tumor suppressor in mice leads to increased cancer susceptibility, mirroring human Li-Fraumeni syndrome. This method has been pivotal in creating over 10,000 knockout mouse lines, cataloging functions across the mammalian . Site-directed mutagenesis extends knockout approaches by introducing specific changes, such as point mutations or small insertions/deletions, to study subtle functional impacts without complete ablation. A common PCR-based strategy involves amplifying the or genomic target with overlapping primers incorporating the desired codon alteration, followed by replacement via recombination or ligation-independent . This yields precise variants, like changing a catalytic residue in an to assess specificity. In functional genomics, such has been used to generate hypomorphic alleles in and , revealing dosage-sensitive interactions; for example, altering a single codon in the in human cell lines disrupts fidelity, linking it to predisposition. Efficiencies exceed 90% in optimized protocols, making it suitable for iterative engineering in non-model systems. Classical chemical mutagenesis induces random mutations across the using alkylating agents like (EMS) or N-ethyl-N-nitrosourea (), which primarily cause G/C-to-A/T transitions. In non-model organisms such as or , mutagenized populations (M1 generation) are screened for phenotypes in subsequent generations (M2), with mutations mapped via bulk segregant analysis or whole-genome sequencing to identify causal variants. This forward approach has historically uncovered thousands of loci; ENU mutagenesis in mice, for instance, generated over 300,000 mutants, identifying genes in olfaction, immunity, and reproduction pathways. Though labor-intensive, it remains valuable for discovering novel genes in species lacking advanced tools, with mutation rates tunable to 1-5 per genome. Key principles underlying these techniques distinguish null alleles, which completely eliminate gene product and often result in severe or lethal phenotypes, from hypomorphs that partially reduce function and produce milder, viable effects. Null knockouts are ideal for identifying essential genes—those whose inactivation causes lethality or sterility—comprising about 15-20% of the genome in model organisms like yeast and mice. Hypomorphic mutations, achievable via partial deletions or missense changes, allow study of gene dosage effects and redundancy; for example, a hypomorphic allele of the Drosophila Notch gene causes subtle wing vein defects rather than embryonic death. Distinguishing these requires complementation tests and expression analysis to confirm loss-of-function extent, ensuring accurate functional annotation. Modern enhancements, such as CRISPR-assisted recombination, have improved precision but build on these classical foundations.

RNA Interference Approaches

RNA interference (RNAi) is a post-transcriptional mechanism that utilizes molecules to target and degrade specific messenger RNAs (mRNAs), thereby inhibiting . This process was first demonstrated in the Caenorhabditis elegans through the introduction of double-stranded (dsRNA), which triggered potent and sequence-specific interference with endogenous gene function. In functional genomics, RNAi enables reversible loss-of-function studies by transiently or stably suppressing target genes without altering the DNA sequence, distinguishing it from permanent genetic modifications like knockouts. The core RNAi pathway involves the processing of dsRNA into small interfering RNAs (siRNAs) by the enzyme , an RNase III family endonuclease that cleaves long dsRNAs into 21-23 nucleotide duplexes with 2-nucleotide 3' overhangs. These siRNAs are then incorporated into the (RISC), where the protein (primarily Ago2 in mammals) unwinds the duplex and uses the guide strand to recognize complementary mRNA via base-pairing, leading to mRNA or translational repression.00293-4) For siRNA in mammalian systems, synthetic 21-nucleotide duplexes are chemically synthesized to mimic Dicer products, ensuring efficient RISC loading and specificity; optimal designs feature low secondary structure, moderate (30-52%), and avoidance of immune-stimulatory motifs. Short hairpin RNAs (shRNAs), expressed from DNA vectors under promoters like U6 or H1, form stem-loop structures that are processed by Dicer into siRNAs, allowing stable, long-term knockdown in dividing cells through genomic integration. In functional genomics applications, RNAi facilitates loss-of-function phenotyping in mammalian cells and tissues by systematically silencing genes to reveal their roles in cellular processes, such as , , and signaling pathways. High-throughput RNAi screens using shRNA libraries have identified key regulators in cancer and , with phenotypes observed via , viability assays, or transcriptomics. Off-target effects, where siRNAs or shRNAs unintentionally silence non-target transcripts due to partial sequence complementarity (especially in the seed region), can confound results; mitigation strategies include using multiple orthogonal siRNAs per target, pooling low-concentration siRNAs to dilute individual off-targets, and incorporating chemical modifications like 2'-O-methyl groups on the passenger strand to enhance specificity and reduce immune activation. Variants of RNAi, such as miRNA mimics, extend its utility to studying endogenous regulation by replicating the multifaceted action of natural microRNAs (miRNAs), which typically repress multiple targets through imperfect base-pairing and translational inhibition rather than cleavage. Synthetic miRNA mimics, double-stranded designed to match mature miRNA sequences, are transfected into cells to overexpress miRNA activity, enabling gain-of-function analysis of regulatory networks in contexts like oncogenesis or immune responses. Despite its versatility, RNAi has limitations, including incomplete knockdown (often 70-90% mRNA reduction, with variable protein-level effects due to mRNA and translational buffering), which may yield subtle or ambiguous phenotypes compared to full knockouts. Recent advances as of 2025 have improved targeting of long non-coding RNAs (lncRNAs), which were historically challenging due to their low expression and localization; optimized siRNAs with enhanced stability and delivery via nanoparticles have achieved >80% knockdown of lncRNAs like SMILR in vascular cells, revealing roles in and without significant off-targeting.

CRISPR-Based Perturbations and Screens

CRISPR-Cas9 enables precise by employing a single-guide (sgRNA) that directs the endonuclease to target DNA sequences adjacent to a (), most commonly the NGG sequence derived from . The sgRNA design involves a 20-nucleotide spacer complementary to the target locus, fused to a scaffold that recruits , forming a ribonucleoprotein complex that induces a double-strand break (DSB) three base pairs upstream of the . These DSBs are repaired via (NHEJ), often resulting in insertions or deletions (indels) that disrupt gene function for knockouts, or () for knock-ins using donor templates, though efficiency remains lower, typically 5-20% in mammalian cells without optimization. Knockout efficiencies can exceed 80% with well-designed sgRNAs targeting early exons, minimizing off-target effects through algorithms that predict specificity based on mismatch tolerance and accessibility. Derived from the foundational system, CRISPR variants expand perturbation capabilities beyond DSBs. CRISPR interference (CRISPRi) uses a catalytically dead (dCas9) fused to the Krüppel-associated box () repressor domain to block transcription initiation, achieving up to 90% gene repression without altering the DNA sequence.00208-5) Conversely, CRISPR activation (CRISPRa) employs dCas9 fused to domains like VP64 or p300 to enhance transcription, with efficiencies reaching 100-fold upregulation for certain promoters.00826-X) editing integrates a or deaminase with a nickase (nCas9) to enable C-to-T or A-to-G transitions without DSBs, offering 30-60% efficiency and reduced formation compared to standard . Prime further advances precision by pairing nCas9 with a and a prime guide RNA (pegRNA) that specifies the edit, allowing all 12 base substitutions, small insertions, or deletions with efficiencies up to 50% and minimal byproducts. CRISPR-based screens systematically assess function by perturbing thousands of loci in parallel. Pooled screens deliver a of sgRNAs via lentiviral into a population, followed by phenotypic selection; changes in sgRNA abundance are quantified by next-generation sequencing to identify enriched or depleted genes, as demonstrated in genome-scale knockouts revealing essentiality in human s. Arrayed screens, in contrast, individual sgRNAs in multi-well plates for high-content imaging or biochemical readouts, enabling multiparametric analysis but at higher cost and lower throughput. Common readouts include fluorescence-activated sorting (FACS) for surface marker-based enrichment or sequencing for proliferation phenotypes, with pooled formats excelling in identifying regulators of or viral infection. Advancements by 2025 have enhanced 's utility in complex systems, including highly functional editors with improved flexibility and reduced off-target activity for vertebrate models. In and models, perturbations now facilitate tissue-specific screens, such as identifying developmental regulators via or nanoparticle delivery, bridging findings with physiological contexts. In , screens and edits have targeted non-human applications, generating crop varieties like drought-tolerant and disease-resistant through multiplexed knockouts, yielding 15-25% improved performance under stress without transgenes. These developments underscore 's role in scalable functional annotation beyond mammalian systems.

Functional Gene Annotation

Genome-Wide Annotation Strategies

Genome-wide annotation strategies in functional genomics involve the systematic assignment of biological functions to and their products across entire genomes, leveraging both experimental assays and computational predictions to build comprehensive functional maps. These strategies aim to catalog gene roles in processes such as molecular function, biological processes, and cellular components, facilitating downstream analyses in disease modeling and . Central to this is the use of standardized ontologies and that ensure consistency and across datasets. Annotation pipelines primarily rely on frameworks like the for assigning terms that describe gene functions, with approximately 39,000 terms organized hierarchically to represent molecular activities and pathways as of October 2025. GO annotations are generated through manual curation from literature and high-throughput experiments, or computationally via sequence similarity and inference, resulting in millions of associations for human genes alone. Similarly, entries provide detailed protein annotations, including function, subcellular location, and interactions, curated from experimental data and integrated with GO terms for cross-referencing. A key feature of these pipelines is the inclusion of evidence codes to qualify the reliability of annotations, such as (Inferred from Direct Assay) in GO, which denotes experimental validation through techniques like assays or studies. employs Evidence and Conclusion Ontology () codes, where experimental evidence like ECO:0000269 (sequence variant evidence) supports claims derived from assays, while computational codes like ECO:0000256 indicate model-based predictions. These codes enable users to filter annotations by confidence, with experimental evidence comprising about 20-30% of total GO annotations for well-studied organisms. Integration of diverse types enhances annotation accuracy by combining profiles, protein-protein interaction networks, and phenotypic outcomes to infer functions contextually. For instance, from microarrays or can link co-expressed genes to shared pathways, while interaction data from yeast two-hybrid screens refines functional partnerships, and phenotype associations from model organisms validate roles in . approaches, such as guilt-by-association methods, propagate annotations by clustering genes based on these integrated features, improving coverage for understudied genes. Tools like Ensembl and GENCODE provide genome-wide annotations for the human genome, with GENCODE offering evidence-based transcript models that include approximately 63,000 protein-coding and non-coding genes (19,433 protein-coding, 35,899 long non-coding RNA, and 7,563 small non-coding RNA genes) in release 49 (February 2025), aligned to the GRCh38 assembly. Ensembl integrates GENCODE annotations with comparative genomics and variant data, enabling visualization and querying of functional elements via its browser interface. These resources update regularly, incorporating community feedback to refine gene structures and add functional labels. Challenges persist in annotating non-coding RNAs (ncRNAs), which constitute over 80% of transcribed genes but lack conserved protein-coding signatures, complicating detection and functional assignment. Current pipelines struggle with ncRNA delineation due to variable lengths and low sequence conservation, often relying on expression patterns or secondary structure predictions, yet only a fraction receive GO terms compared to protein-coding genes. As of 2025, efforts like the Atlas of Variant Effects Alliance focus on variant-effect predictions to annotate ncRNA regulatory roles, standardizing multiplexed assays and computational predictors to map impacts on phenotypes and improve diagnostic utility.

Comparative and Evolutionary Methods

Comparative and evolutionary methods in functional genomics leverage sequence similarities and evolutionary relationships across to infer functions, complementing direct experimental by providing indirect evidence of conserved roles. These approaches exploit the principle that orthologous genes—those derived from a common ancestor—often retain similar functions, while patterns of or co-evolution reveal interactions and pathways. By analyzing genomic sequences from diverse , researchers can predict functions for uncharacterized genes, particularly in non-model where experimental data is scarce. This evolutionary perspective has been instrumental in scaling up functional genome-wide, drawing on principles of basic annotation strategies such as identifying coding regions and regulatory elements. The approach identifies potential protein-protein interactions by detecting fusion events where two separate proteins in one are fused into a single polypeptide in another, suggesting that the unfused components likely interact as partners in the original organism. Proposed by Marcotte and colleagues in , this method scans databases of protein sequences across genomes to find such "" proteins, which serve as indicators of functional associations; for instance, in and eukaryotes, fusions between enzymes in metabolic pathways have predicted interactions later validated in . The approach has been applied to predict over 25,000 interactions in proteins by comparing with prokaryotic genomes, highlighting conserved modules like complexes. Limitations include false positives from domain shuffling unrelated to interactions, but refinements using structural data have improved accuracy to around 70% in studies. Orthology-based function transfer relies on identifying orthologous genes across and annotating uncharacterized ones based on the known of their counterparts, using tools like for sequence similarity searches or OrthoMCL for clustering ortholog groups via reciprocal best hits and Markov clustering. Developed in the early , OrthoMCL has grouped over 100,000 families from 55 , enabling function predictions with 80-90% accuracy for well-conserved genes in databases like ; for example, transferring metabolic enzyme functions from to human orthologs has aided drug target identification. This method underpins resources like the database, which integrates with functional terms from for probabilistic transfers. Challenges arise with paralogs or rapidly evolving genes, where phylogenetic trees help refine assignments. Phylogenetic profiling infers functional relationships by examining the co-occurrence or co-absence of s across multiple genomes, positing that genes involved in the same pathway evolve together due to selective pressures. Introduced by Pellegrini et al. in 1999, this technique constructs binary profiles of presence/absence in a set of genomes and correlates them using metrics like Pearson's coefficient, identifying co-occurring genes as likely partners; in prokaryotes, it has reconstructed over 1,000 operons and pathways, such as biosynthesis networks in . Applications extend to eukaryotes, where profiling across 20+ genomes has predicted interactions in disease genes with 60-75% precision. Advanced variants incorporate neighborhood and data for higher resolution.

Bioinformatics in Functional Genomics

Data Processing and Analysis Pipelines

Data processing and analysis pipelines in functional genomics transform raw high-throughput sequencing data into interpretable insights, addressing challenges like sequence quality, alignment accuracy, and statistical variability across experiments. These workflows typically begin with preprocessing to ensure data reliability, followed by specialized analyses tailored to the assay type, such as RNA sequencing (RNA-seq), DNA variant detection from perturbations, or chromatin accessibility profiling. Standardized pipelines enhance reproducibility and scalability, particularly as functional genomics datasets grow exponentially with multi-omics integration. Preprocessing forms the foundational step, involving (), , and to mitigate technical artifacts. Tools like FastQC provide a rapid assessment of raw FASTQ files, evaluating metrics such as per-base sequence quality, adapter contamination, and GC content bias to identify issues before downstream analysis. For data, to a is commonly performed using ultrafast spliced aligners like , which maps reads with high accuracy by detecting splice junctions and handling multimapping efficiently, achieving speeds over 50 times faster than contemporaries on datasets. then adjusts for library size, sequencing depth, and compositional biases; methods such as relative log expression (RLE) or trimmed mean of M-values (TMM) are widely applied to stabilize variance across samples, enabling fair comparisons in differential analyses. Following preprocessing, variant calling identifies genetic alterations from perturbation experiments, such as those induced by or . The Genome Analysis Toolkit (GATK) from the Broad Institute is a cornerstone for this, employing a Bayesian framework to detect single nucleotide variants (SNVs) and insertions/deletions (indels) in data, with best practices workflows incorporating base quality score recalibration and joint genotyping for improved precision. In RNA-focused studies, differential expression analysis quantifies changes in gene activity using count-based models; DESeq2, for instance, applies negative binomial generalized linear models with shrinkage estimation for dispersions and fold changes, reducing false positives in low-count genes and outperforming alternatives in stability for count data. For chromatin immunoprecipitation (ChIP-seq) and assay for transposase-accessible chromatin (ATAC-seq), peak calling delineates enriched regions of DNA-protein interactions or open chromatin. MACS2 employs a dynamic lambda model to scan aligned reads for significant enrichments over background, accommodating varying fragment sizes and input controls, and has been optimized for broader applications including ATAC-seq with parameters like --nomodel for nucleosome-free regions. These analyses output annotated peaks or variant lists, often visualized in genome browsers for initial validation. Scalability has advanced with cloud-based platforms to manage the petabyte-scale volumes of multi-omics data generated in 2025. , an open-source , orchestrates end-to-end workflows for functional genomics, integrating tools like , DESeq2, and MACS2 into reusable pipelines with built-in provenance tracking, supporting for analyses involving thousands of samples across , ChIP-seq, and perturbation datasets. Recent frameworks, such as Suite, further enable scalable processing of spatial multi-omics by modularizing alignment, normalization, and peak/variant detection in containerized environments, handling integrated datasets from diverse assays with minimal computational overhead.

Integration and Predictive Modeling

Integration in functional genomics involves combining diverse datasets, such as protein-protein interaction () networks derived from yeast two-hybrid (Y2H) and affinity purification-mass spectrometry (APMS) methods, with gene regulatory networks (GRNs) inferred from (eQTLs). Y2H screens, which detect binary interactions by reconstituting transcription factors in cells, have mapped large-scale PPI networks in model organisms like , providing foundational maps of protein complexes. APMS, by contrast, captures stable multiprotein assemblies through affinity tagging followed by , enabling the construction of context-dependent interaction networks in cells. These approaches yield high-confidence interactomes that serve as scaffolds for functional inference, with recent probabilistic models integrating Y2H and APMS data to resolve and reduce false positives. GRNs are constructed by leveraging eQTL data, which links genetic variants to levels, to infer regulatory relationships. cis-eQTLs, acting on nearby genes, facilitate the identification of direct transcriptional regulators, while trans-eQTLs reveal broader network effects. Structural equation models (SEMs) jointly map eQTL effects and GRN structures, using sparse regularization to predict causal edges from expression data across tissues. Incorporating prior biological knowledge, such as transcription factor binding motifs, enhances GRN accuracy from eQTLs, enabling tissue-specific reconstructions that highlight key drivers of phenotypic variation. Machine learning advances predictive modeling in functional genomics, with random forests applied to forecast variant effects on protein function and disease. These ensemble methods integrate genomic features like conservation scores and biochemical annotations to classify pathogenic variants, outperforming single classifiers in prioritizing non-coding mutations. architectures, including convolutional neural networks, predict CRISPR off-target effects by modeling guide RNA-DNA mismatches and epigenetic contexts, achieving over 90% accuracy in validating experimental edits. Multi-omics employs joint models to link genotypes to phenotypes, synthesizing , transcriptomics, and for holistic predictions. Techniques like multi-view and graph neural networks integrate heterogeneous data layers, revealing genotype-phenotype associations in . In 2025, explainable models such as MOGATFF enhance fusion for modeling, using mechanisms to interpret regulatory pathways. These approaches draw on processed multi-omics inputs to simulate functional phenotyping, improving resolution in variant-to-function mapping. Predictive applications extend to disease risk assessment via functional scores that quantify variant impacts on networks and expression. Integrating and GRN data into polygenic models refines risk stratification for conditions like , where functional annotations boost predictive power beyond sequence alone. AI-designed editors, generated from large-scale datasets using generative models, enable precise functional perturbations, as demonstrated by OpenCRISPR-1's high-fidelity editing in human genomes. These tools predict and mitigate off-target risks while optimizing therapeutic designs.

Major Consortium Projects

ENCODE Project

The () project, launched in 2003 by the (), aims to identify all functional elements in the and , including protein-coding , transcripts, and regulatory elements that control activity. Initially, the pilot phase analyzed approximately 1% of the across 44 regions to test methods for annotating functional components. Subsequent production phases— 2 (2007–2012), 3 (2012–2017), and 4 (2017–2022)—expanded to whole- analyses, incorporating data from over 400 and and types. and updates continue, with enhancements to released as of October 2025. ENCODE employs a suite of high-throughput assays to map functional elements, including chromatin immunoprecipitation sequencing (ChIP-seq) for transcription factor binding and histone modifications, RNA sequencing () for transcriptomes, DNase I hypersensitive sites sequencing (DNase-seq) for open chromatin regions, and DNA methylation profiling to capture epigenetic states. These methods, combined with and computational integration, enable the identification of promoters, enhancers, insulators, and non-coding RNAs across diverse biological contexts. For instance, DNase-seq highlights accessible regulatory DNA, while delineates enhancer landscapes by revealing cell-type-specific marks like H3K27ac. Key findings from ENCODE have illuminated the genome's functional architecture, revealing that approximately 80% of the exhibits biochemical activity, such as RNA production or protein-DNA interactions, in at least one , challenging earlier views of extensive ". The project has mapped over 399,000 enhancer-like regions and 70,000 promoter-like regions, demonstrating their cell-specific roles in and linking many -associated variants to non-coding functional elements. These insights underscore the prevalence of regulatory complexity in non-coding sequences, with ongoing analyses emphasizing dynamic states and their implications for and . All data, exceeding 106,000 datasets as of March 2024, are freely accessible through the ENCODE Portal, which supports visualization, download, and integration via tools like the . Regular updates focus on non-coding functions, including single-cell assays and 3D mapping, to enhance predictive models of gene regulation.

GTEx Project

The Genotype-Tissue Expression (GTEx) project, initiated in 2010 by the Common Fund, serves as a foundational resource in functional genomics by systematically linking genetic variants to patterns across diverse human s. This effort involves the collection of postmortem samples from 948 donors, yielding 19,788 sequencing () samples from up to 54 non-diseased sites per donor, with data generation completed by 2020 and ongoing analyses through 2025, including the initiation of the Developmental GTEx (dGTEx) project for prenatal s as of January 2025. s are harvested rapidly after death—typically within 6 to 24 hours—using standardized protocols to ensure high quality and minimize postmortem degradation effects. This postmortem sampling approach enables the study of gene regulation in a broad array of s, including regions, heart, liver, and , providing a comprehensive atlas of baseline human expression variability. Methodologically, GTEx employs whole-genome sequencing for and deep for transcriptomic profiling, generating median coverage of 82.6 million reads per sample to quantify levels. These data facilitate expression (eQTL) mapping, which identifies - and genetic variants influencing expression in 50 tissues, encompassing 19,466 samples from 943 donors in the core analysis set (v10, as of 2024). -eQTLs, typically acting within 1 of target genes, were detected for a substantial portion of genes, while , operating distally, were rarer but highlighted key regulatory networks; splicing QTLs (sQTLs) further revealed variant effects on . This multi-omic integration allows fine-mapping of causal variants, with over 80% of -eQTL signals credibly assigned to a median of six variants per locus. Key findings from GTEx underscore the prevalence of tissue-specific gene regulation, where eQTL effects vary markedly by tissue context, with trans-eQTLs exhibiting greater tissue specificity than cis-eQTLs. For instance, while many cis-eQTLs are shared across tissues, others are restricted to specific organs like the or , reflecting localized regulatory mechanisms. Notably, more than 77% of trans-eQTL effects appear indirect, mediated through cis-eQTLs on intermediate genes, emphasizing the complexity of genetic cascades in expression control. These insights reveal that nearly all protein-coding genes (94.7%) and a of long non-coding RNAs (67.3%) harbor detectable regulatory variants, establishing genetic effects as a primary driver of expression diversity. The project's applications extend to illuminating disease mechanisms, particularly by interpreting non-coding associated with like and cancer through with GWAS signals. The GTEx portal (gtexportal.org) offers interactive tools for eQTL , fine-mapping, and analyses, enabling researchers to prioritize causal and target genes for functional follow-up. This resource has informed hundreds of studies, enhancing precision medicine by bridging genotype to phenotype in a tissue-aware manner.

Alliance of Genome Resources

The Alliance of Genome Resources (AGR) is a consortium established in 2016 to integrate and harmonize genetic and genomic data from major model organism databases, enabling comparative analyses that inform human biology and disease research. Founding members include FlyBase (Drosophila), Mouse Genome Informatics (MGI), Rat Genome Database (RGD), Saccharomyces Genome Database (SGD), WormBase (Caenorhabditis elegans), Zebrafish Information Network (ZFIN), and the Gene Ontology (GO) Consortium, with Xenbase (Xenopus) joining in 2022. The initiative addresses challenges faced by individual databases, such as resource limitations and data silos, by creating a centralized infrastructure for sharing annotations on gene function, phenotypes, and variants. By 2025, the Alliance has expanded its integration to include human data through orthology mappings and GO terms, facilitating cross-species insights into conserved biological processes, with recent releases like version 8.2 in September 2025. AGR employs standardized methods for data harmonization, leveraging predictions to align genes across species and map functional annotations. Orthologs serve as the core framework for propagating (GO) terms, which describe molecular functions, biological processes, and cellular components, derived from experimental evidence including genetic perturbations. ontologies, such as the Unified Phenotype Ontology (UPHENO), enable consistent representation of observable traits and their associations with alleles or variants, allowing researchers to compare phenotypic outcomes from model organisms to human conditions. These approaches ensure interoperability, with data curated from primary literature and high-throughput studies, and are accessible via tools like AllianceMine for querying orthologous gene sets. Key findings from AGR analyses highlight the conservation of functions across , revealing that orthologous s often share GO annotations for essential pathways, such as and , with implications for understanding evolutionary divergence and human disease orthologs. For instance, comparative studies have identified conserved variant effects on protein function, aiding predictions of pathogenicity in human variants by extrapolating from data. These insights underscore the value of model organisms in elucidating non-human functional genomics, where gaps persist in less-studied . The Alliance's primary contributions include a unified web portal (www.alliancegenome.org) launched in 2019, which provides searchable access to over 1 million genes and millions of annotations, streamlining research workflows. This portal integrates downloads, genome browsers, and visualization tools, promoting data reuse and collaboration. By addressing fragmentation in non-human functional genomics, AGR has democratized access to comparative resources, fostering discoveries in areas like variant interpretation and phenotype-genotype mapping without relying solely on human-centric datasets.

References

  1. [1]
    An Introduction to Functional Genomics and Systems Biology - PMC
    The field of functional genomics attempts to describe the functions and interactions of genes and proteins by making use of genome-wide approaches, in contrast ...Missing: review | Show results with:review
  2. [2]
    Functional Genomics - an overview | ScienceDirect Topics
    Functional genomics is defined as the study of the functional elements within a genome, including genes and regulatory sequences, to identify genetic changes ...
  3. [3]
    Clinical Functional Genomics - PMC - NIH
    Sep 15, 2021 · Functional genomics is the study of how the genome and its products, including RNA and proteins, function and interact to affect different biological processes.
  4. [4]
    Functional genomics - Latest research and news - Nature
    Functional genomics uses genomic data to study gene and protein expression and function on a global scale (genome-wide or system-wide)
  5. [5]
    Functional genomics: it's all how you read it - PubMed
    Functional genomics: it's all how you read it. Science. 1997 Oct 24;278(5338):601-2. doi: 10.1126/science.278.5338.601. Authors. P Hieter , M Boguski ...
  6. [6]
    Functional genomics | Nature
    Jun 15, 2000 · Functional genomics has leapt from being a surrealistic, or at least futuristic, concept in the 1980s to an accepted (if not yet everyday) part of science in ...<|control11|><|separator|>
  7. [7]
    NHGRI History and Timeline of Events
    Our institute's history is inextricably intertwined with the Human Genome Project and the history of the field of genomics. Introduction. The National Human ...
  8. [8]
    A Programmable Dual-RNA–Guided DNA Endonuclease ... - Science
    Jun 28, 2012 · Bacteria and archaea protect themselves from invasive foreign nucleic acids through an RNA-mediated adaptive immune system called CRISPR ...
  9. [9]
    CRISPR/Cas9 therapeutics: progress and prospects - Nature
    Jan 16, 2023 · This paper reviews the current developments in three aspects, namely, gene-editing type, delivery vector, and disease characteristics.
  10. [10]
    GAUDI: interpretable multi-omics integration with UMAP ... - Nature
    Jul 1, 2025 · We present a novel, non-linear, and unsupervised method called GAUDI (Group Aggregation via UMAP Data Integration) that leverages independent UMAP embeddings.
  11. [11]
    GENCODE 2025: reference gene annotation for human and mouse
    Nov 20, 2024 · Over the last 20 years, the number of human protein-coding genes annotated by GENCODE has gradually reduced, with 19 433 in v47. This largely ...Abstract · Introduction to GENCODE · Conclusion · Data availability
  12. [12]
    Functional genomics bridges the gap between quantitative genetics ...
    Functional genomics bridges the gap between quantitative genetics and molecular biology by studying the functional effects of genetic variants, bringing these ...
  13. [13]
    A Statistical Framework to Predict Functional Non-Coding Regions ...
    It is estimated that approximately 98% of the human genome is non-protein-coding. Because of the apparent importance of coding regions, many computational tools ...Missing: percentage | Show results with:percentage
  14. [14]
    An Integrated Encyclopedia of DNA Elements in the Human Genome
    The Encyclopedia of DNA Elements (ENCODE) Project aims to delineate all functional elements encoded in the human genome. Operationally, we define a functional ...Missing: objectives | Show results with:objectives
  15. [15]
    Statistical Challenges in Functional Genomics - Project Euclid
    Feb 16, 2001 · Microarray technology makes it possible to simultaneously observe thousands of genes in action and to dissect the functions, the reg- ulatory ...
  16. [16]
    From GWAS to Function: Using Functional Genomics to Identify the ...
    May 12, 2020 · Statistical methods designed to tackle these challenges integrate GWAS results with functional genomics data such as gene expression or ...<|separator|>
  17. [17]
    Sequence determinants of human gene regulatory elements - Nature
    Feb 21, 2022 · DNA can determine where and when genes are expressed, but the full set of sequence determinants that control gene expression is unknown.
  18. [18]
    Expression quantitative trait locus analysis for translational medicine
    eQTLs suggest mechanisms by which polymorphisms may influence gene function as it relates to disease, particularly where they alter experimentally or ...
  19. [19]
    Functional regression method for whole genome eQTL epistasis ...
    May 18, 2017 · Studying the effect of epistasis on the gene expression could provide a better understanding of the genetic architecture and gene regulation.Missing: annotation | Show results with:annotation
  20. [20]
    Endogenous fine-mapping and prioritization of functional regulatory ...
    Aug 28, 2025 · Identifying fine-grained regulatory elements and causal variants underlying complex traits remains a major challenge in functional genomics and ...Missing: goals | Show results with:goals
  21. [21]
    Functional assays provide a robust tool for the clinical annotation of ...
    Mar 2, 2016 · Our results indicate that systematic functional assays can provide a robust tool to aid in clinical annotation of VUS.Missing: personalized | Show results with:personalized
  22. [22]
  23. [23]
    Progress in soybean functional genomics over the past decade - PMC
    Yield is one of the breeding priorities for soybean. Soybean yield is determined mainly by plant architecture, seed weight and size, and seed number per pod. In ...
  24. [24]
    Soybean2035: A decadal vision for soybean functional genomics ...
    Feb 3, 2025 · Reduced genetic load was detected during soybean domestication and improvement, with a 7.1% reduction in deleterious mutations in landraces than ...
  25. [25]
    Functional Genomics - jbei.org
    This work includes identification of novel biomass-degrading enzymes from compost samples for the Deconstruction division, characterization of the cytosolic and ...
  26. [26]
    Functional testing of thousands of osteoarthritis-associated variants ...
    Jun 4, 2019 · Most of the underlying signal is believed to derive from variation in non-coding regulatory sequences. However, because of linkage ...
  27. [27]
    Translational genomics of osteoarthritis in 1,962,069 individuals
    Apr 9, 2025 · Common non-coding sequence variants associated with osteoarthritis phenotypes present concordant directions of effect with gene-burden ...
  28. [28]
    Drug target prediction through deep learning functional ... - Nature
    Feb 29, 2024 · We develop an approach that represents gene signatures projected onto their biological functions, instead of their identities, similar to how the word2vec ...
  29. [29]
    CRISPR Clinical Trials: A 2025 Update - Innovative Genomics Institute
    Jul 9, 2025 · An update on the progress of CRISPR clinical trials with the latest data and a survey of the CRISPR landscape in 2025.Blood Disorders · Cancers · Diabetes
  30. [30]
    CRISPR Therapeutics: Home
    November 8, 2025. CRISPR Therapeutics ... First-Ever Approved CRISPR-Based Therapy. CASGEVY™ (exagamglogene autotemcel), a CRISPR/Cas9 gene-edited therapy ...Pipeline · Careers · Therapies · Crispr-x
  31. [31]
    Genomics in the Agri-Food Economy: Creating Value from Genetic...
    Jul 22, 2025 · Genomics is driving transformative value creation across Canada's agri-food sector, delivering advancements that enhance productivity, ...
  32. [32]
    Synthetic lethality: General principles, utility and detection using ...
    Jan 3, 2011 · Based on studies ranging from yeast to human cells, this review provides an overview of the general principles that underlie synthetic lethality ...
  33. [33]
    A compendium of synthetic lethal gene pairs defined by extensive ...
    Sep 18, 2025 · We generate a dual-guide CRISPR/Cas9 Library and analyse 472 predicted synthetic lethal pairs in 27 cancer cell Lines from melanoma, pancreatic ...
  34. [34]
    The genetic interaction map of the human solute carrier superfamily
    This full-scale genetic interaction map of human SLC transporters is the backbone for understanding the intricate functional network of SLCs in cellular ...
  35. [35]
    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: seminal | Show results with:seminal
  36. [36]
    Practical Guidelines for the Comprehensive Analysis of ChIP-seq Data
    Nov 14, 2013 · We address all the major steps in the analysis of ChIP-seq data: sequencing depth selection, quality checking, mapping, data normalization, assessment of ...Missing: paper | Show results with:paper
  37. [37]
    Transcription Factor ChIP-seq Data Standards and Processing ...
    ChIP-seq is a method used to analyze protein interactions with DNA. ChIP-seq combines chromatin immunoprecipitation with DNA sequencing to infer the possible ...Missing: seminal paper
  38. [38]
    High-Resolution Mapping and Characterization of Open Chromatin ...
    Jan 25, 2008 · We employed high-throughput sequencing and whole-genome tiled array strategies to identify DNase I HS sites within human primary CD4 + T cells.Missing: original | Show results with:original
  39. [39]
    Chromatin accessibility profiling by ATAC-seq - PubMed Central
    ATAC-seq provides a simple and scalable way to detect the unique chromatin landscape associated with a cell type and how it may be altered by perturbation or ...
  40. [40]
    Methods and applications of in vivo CRISPR screening - PubMed
    Jul 29, 2025 · In vivo CRISPR screening uses pooled CRISPR-Cas perturbation to generate genotype-phenotype maps, accelerating gene function discovery across ...Missing: complex traits 2024
  41. [41]
    Engineered prime editors with minimal genomic errors - Nature
    Sep 17, 2025 · Prime editors are advanced CRISPR tools that enable replacement of targeted DNA with programmed sequences. A prime editor comprises a Cas9 ...<|control11|><|separator|>
  42. [42]
    Emerging trends in prime editing for precision genome editing - Nature
    Jul 31, 2025 · Prime editing is an advanced genome editing technology that enables precise genetic modifications without inducing double-strand breaks or ...
  43. [43]
    RNA-Seq: a revolutionary tool for transcriptomics - PMC - NIH
    RNA-Seq is a recently developed approach to transcriptome profiling that uses deep-sequencing technologies. Studies using this method have already altered ...
  44. [44]
    Quantitative Monitoring of Gene Expression Patterns with ... - Science
    Quantitative Monitoring of Gene Expression Patterns with a Complementary DNA Microarray. Mark Schena, Dari Shalon, [...] , Ronald W. Davis, and Patrick O ...
  45. [45]
    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 ...
  46. [46]
    mRNA-Seq whole-transcriptome analysis of a single cell - Nature
    Apr 6, 2009 · These new techniques usually need microgram amounts of total RNA for analysis, which corresponds to hundreds of thousands of mammalian cells.Missing: original | Show results with:original
  47. [47]
    Multiome Perturb-seq unlocks scalable discovery of integrated ...
    Jan 15, 2025 · We introduce Multiome Perturb-seq, extending single-cell CRISPR screens to simultaneously measure perturbation-induced changes in gene expression and chromatin ...
  48. [48]
  49. [49]
    The Yeast Two-Hybrid System for Identifying Protein ... - PubMed
    The yeast two-hybrid assay is a system for identifying and analysing protein-protein interactions. Since the original description in 1989, the technique has ...
  50. [50]
    Quantitative affinity purification mass spectrometry - Frontiers
    Affinity purification combined with mass spectrometry (AP-MS) has emerged as a particularly attractive method for PPI mapping (Gingras et al., 2007). A major ...Abstract · Introduction · Quantitative Shotgun Proteomics · Specificity and Sensitivity
  51. [51]
    Mass spectrometry‐based protein–protein interaction networks for ...
    Jan 12, 2021 · Here, we review MS techniques that have been instrumental for the identification of protein–protein interactions at a system‐level.
  52. [52]
    Affinity Purification Mass Spectrometry on the Orbitrap–Astral Mass ...
    Mar 2, 2025 · We describe our methodology using the Orbitrap–Astral mass spectrometer with 7 min, high-flow separations to analyze 216 AP-MS samples in ∼29 h.
  53. [53]
    Deep mutational scanning: a new style of protein science - PMC
    Deep mutational scanning assesses many mutant protein versions using high-throughput DNA sequencing to reveal protein properties and behavior.Missing: original | Show results with:original
  54. [54]
    A statistical framework for analyzing deep mutational scanning data
    Aug 7, 2017 · Deep mutational scanning is a widely used method for multiplex measurement of functional consequences of protein variants.
  55. [55]
    Learning protein fitness landscapes with deep mutational scanning ...
    Aug 16, 2023 · We describe a multi-protein training scheme that leverages the existing deep mutational scanning data from diverse proteins to aid in understanding the fitness ...
  56. [56]
    Multimodal cell maps as a foundation for structural and functional ...
    Apr 9, 2025 · Here we construct a global map of human subcellular architecture through joint measurement of biophysical interactions and immunofluorescence images.
  57. [57]
    [PDF] Multimodal cell maps as a foundation for structural and functional ...
    biophysical interaction data are integrated into a multimodal cell map, which is explored across five biological use cases and in an interactive visualization.
  58. [58]
    Overview: Generation of Gene Knockout Mice - PMC - NIH
    Homologous recombination is a DNA repair mechanism that is employed in gene targeting to insert a designed mutation into the homologous genetic locus. Targeted ...
  59. [59]
    Generating gene knockout rats by homologous recombination ... - NIH
    Abstract. We describe here a detailed protocol for generating gene knockout rats by homologous recombination in embryonic stem (ES) cells.
  60. [60]
    Harnessing model organism genomics to underpin the machine ...
    Homologous recombination-knockout. Methods for the specific deletion or transformation of selected target genes through homologous recombination (Fig. 1D) ...
  61. [61]
    Cloning-free PCR-based allele replacement methods - PubMed - NIH
    Here we describe a cloning-free, PCR-based allele replacement method that simplifies allele transfer between yeast strains. The desired allele from one strain ...Missing: functional | Show results with:functional
  62. [62]
    REPLACR-mutagenesis, a one-step method for site ... - Nature
    Jan 11, 2016 · A single-step method, named REPLACR-mutagenesis (Recombineering of Ends of linearised PLAsmids after PCR), for creating mutations (deletions, substitutions and ...
  63. [63]
    An efficient one-step site-directed and site-saturation mutagenesis ...
    We have developed a new primer design method based on the QuickChange™ site-directed mutagenesis protocol, which significantly improves the PCR amplification ...
  64. [64]
    Mutagenesis as a Tool in Plant Genetics, Functional Genomics, and ...
    This paper provides a comprehensive overview of the various techniques and workflows available to researchers today in the field of molecular breeding.
  65. [65]
    Moving forward with chemical mutagenesis in the mouse - O'Brien
    Dec 10, 2003 · The study of genetic variation in mice offers a powerful experimental platform for understanding gene function.
  66. [66]
    Functional genomics the old-fashioned way: Chemical mutagenesis ...
    Aug 6, 2025 · Genetic studies using mutants have led to a greater understanding of the mechanisms underlying the physiology, biochemistry and development ...
  67. [67]
    Rapid generation of hypomorphic mutations | Nature Communications
    Jan 20, 2017 · One example of a hypomorphic condition is a 50% reduction in gene activity from heterozygosity for a null allele, which for some genes can ...
  68. [68]
    Genomic Identification and Functional Characterization of Essential ...
    In this study, we used genetic mapping data, WGS techniques, bioinformatics analyses, and experimental validation, to identify 60 essential genes from 104 ...
  69. [69]
    Genomic identification and functional analysis of essential genes in ...
    Dec 4, 2018 · We successfully identified 44 essential genes with 130 lethal mutations in genomic regions of C. elegans of around 7.3 Mb from Chromosome I.
  70. [70]
    Potent and specific genetic interference by double-stranded RNA in ...
    Feb 19, 1998 · Experimental introduction of RNA into cells can be used in certain biological systems to interfere with the function of an endogenous gene,.
  71. [71]
    From sequence to function: using RNAi to elucidate mechanisms of ...
    Jan 18, 2008 · In this review, we will evaluate the major advancements in the field of mammalian RNAi, specifically in terms of high-throughput assays. Crucial ...
  72. [72]
    Duplexes of 21-nucleotide RNAs mediate RNA interference ... - Nature
    May 24, 2001 · Here we show that 21-nucleotide siRNA duplexes specifically suppress expression of endogenous and heterologous genes in different mammalian cell lines.
  73. [73]
    RNAi Mechanisms and Strategies to Reduce Off-Target Effects - PMC
    Jan 28, 2021 · An elegant way of lowering concentrations of siRNAs is siRNA pooling. Individual siRNAs within such a pool are directed against the same on- ...
  74. [74]
    siRNA Versus miRNA as Therapeutics for Gene Silencing
    In the replacement approach, synthetic miRNAs (also known as miRNA mimics) are used to mimic the function of the endogenous miRNAs. It thus leads to mRNA ...
  75. [75]
    Review Choosing the Right Tool for the Job: RNAi, TALEN, or CRISPR
    May 21, 2015 · RNAi is currently the most extensively used reverse genetics approach to study gene function in mammalian cells. Its success can be attributed ...
  76. [76]
    Small Interfering RNA Therapy Targeting the Long Noncoding RNA ...
    We designed and synthesized 76 SMILR-targeting siRNAs (BHFn ) and a nontargeting control siRNA (siNTC), and transfected each into saphenous vein smooth muscle ...
  77. [77]
    CRISPR–Cas9-mediated genome editing and guide RNA design - NIH
    Here we briefly review this fast moving field, introduce the CRISPR–Cas9 system and its application to genome editing, with a focus on the basic considerations.Missing: seminal | Show results with:seminal
  78. [78]
    CRISPR–Cas9 gRNA efficiency prediction: an overview of predictive ...
    These tools can assist researchers in the guide RNA (gRNA) design process by predicting cleavage efficiency and specificity and excluding undesirable targets.
  79. [79]
    Pooled Versus Arrayed Screens: Considerations Before Choosing a ...
    Aug 12, 2024 · As described above, pooled screens are restricted to binary assays, while arrayed screens are compatible with binary and multiparametric assays.Missing: impact | Show results with:impact
  80. [80]
    Pooled CRISPR screening with single-cell transcriptome read-out
    Widely used pooled screens are restricted to simple readouts including cell proliferation and sortable marker proteins. Arrayed screens allow for comprehensive ...Missing: FACS impact
  81. [81]
    CRISPR-based functional genomics tools in vertebrate models
    Jul 31, 2025 · The advent of CRISPR–Cas technologies has revolutionized functional genomics by enabling precise genetic manipulations in various model ...
  82. [82]
    Revolutionizing Agriculture With CRISPR Technology: Applications ...
    Sep 11, 2025 · In summary, CRISPR enables precise genetic improvements in crops, spanning stress tolerance, yield, nutrition, and disease resistance. Successes ...
  83. [83]
    What CRISPR can do for agriculture and livestock production
    Jun 27, 2025 · CRISPR and gene editing offers powerful new tools for agriculture, allowing scientists to make precise changes to the DNA of crops and livestock.
  84. [84]
    Gene Ontology annotations: what they mean and where they come ...
    Gene Ontology annotations report connections between gene products and the biological types that are represented in the GO using GO evidence codes. The evidence ...Results · The Curator Perspective · Figure 1
  85. [85]
    Introduction to GO annotations - Gene Ontology
    A standard GO annotation is a statement that links a gene product and a GO term via a relation from the Relations Ontology (RO). In standard GO annotations, ...Standard Go Annotations · Semantics Of A Standard Go... · Go-Causal Activity ModelsMissing: pipelines IDA
  86. [86]
    UniProtKB | UniProt help
    Jun 11, 2025 · UniProtKB is a central hub for protein functional information, with two sections: manually annotated (Swiss-Prot) and computationally analyzed ...
  87. [87]
    Guide to GO evidence codes - Gene Ontology
    Each annotation includes an evidence code to indicate how the annotation to a particular term is supported. Evidence codes fall into six general categories:.Experimental Evidence Codes · Phylogenetically-Inferred... · Computational Analysis...Missing: pipelines | Show results with:pipelines
  88. [88]
    Evidence | UniProt help
    Feb 20, 2025 · UniProtKB evidence tags describe the source of information, using a type (ECO code) and source, like experimental evidence or database records.
  89. [89]
    Integration of biological networks and gene expression data using ...
    Commonly used expression analysis methods identify active biological processes from expression profiles by finding enriched gene annotation terms in the lists ...
  90. [90]
    Linking gene expression to phenotypes via pathway information
    Apr 11, 2015 · Establishing robust links among gene expression, pathways and phenotypes is critical for understanding diseases and developing treatments.
  91. [91]
  92. [92]
    Long non-coding RNAs: definitions, functions, challenges and ...
    Genes specifying long non-coding RNAs (lncRNAs) occupy a large fraction of the genomes of complex organisms. The term 'lncRNAs' encompasses RNA polymerase I ...
  93. [93]
    FindNonCoding: rapid and simple detection of non-coding RNAs in ...
    Oct 12, 2021 · Non-coding RNAs are often neglected during genome annotation due to their difficulty of detection relative to protein coding genes.
  94. [94]
    Guidelines for releasing a variant effect predictor - Genome Biology
    Apr 15, 2025 · Here we provide guidelines and recommendations that we believe should be considered when releasing a novel VEP, focusing primarily on tools that score ...
  95. [95]
    Galaxy platform for accessible, reproducible, and collaborative data ...
    May 20, 2024 · Galaxy provides analytical tools that can be used individually or linked into complex workflows with intermediate data outputs capable of triggering logic ...
  96. [96]
  97. [97]
    STAR: ultrafast universal RNA-seq aligner - PubMed - NIH
    STAR outperforms other aligners by a factor of >50 in mapping speed, aligning to the human genome 550 million 2 × 76 bp paired-end reads per hour.Missing: original | Show results with:original
  98. [98]
    VCF - Variant Call Format - GATK - Broad Institute
    VCF, or Variant Call Format, It is a standardized text file format used for representing SNP, indel, and structural variation calls.
  99. [99]
    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 ...
  100. [100]
    Giotto Suite: a multiscale and technology-agnostic spatial multiomics ...
    Oct 1, 2025 · Here we present Giotto Suite, a suite of modular packages that provides scalable and extensible end-to-end solutions for multiscale and ...
  101. [101]
    Building Protein-Protein Interaction Networks with Proteomics ... - NIH
    In this minireview, we survey the most common methods for the systematic identification of protein interactions and exemplify different strategies for the ...
  102. [102]
    Mass spectrometry‐based protein–protein interaction networks ... - NIH
    Jan 12, 2021 · This review discusses mass spectrometry techniques that have been instrumental for identifying protein‐protein interactions.
  103. [103]
    Joint eQTL mapping and inference of gene regulatory network ...
    In this article, we use the structure equation model (SEM) to model both GRN and effect of eQTLs on gene expression, and then develop a novel algorithm, named ...
  104. [104]
    Leveraging prior knowledge to infer gene regulatory networks from ...
    Feb 12, 2025 · This review explores one promising approach that has been proposed to address these challenges: integrating prior knowledge into the inference process.
  105. [105]
    Random Forests for Genomic Data Analysis - PMC - NIH
    Random forests (RF) are a tree-based tool for genomic data analysis, used for prediction, classification, variable selection, and accounting for feature ...
  106. [106]
    Prediction of driver variants in the cancer genome via machine ...
    Oct 22, 2020 · One dedicated tool is CHASM [29, 41], which ranks somatic driver variants for specific cancer types using a Random Forest classifier. CHASM ...
  107. [107]
    Off-target predictions in CRISPR-Cas9 gene editing using deep ...
    Sep 8, 2018 · We design and implement two algorithms using deep neural networks to predict off-target mutations in CRISPR-Cas9 gene editing.
  108. [108]
    A technical review of multi-omics data integration methods
    Aug 1, 2025 · CCA has proven particularly useful as a joint dimensionality reduction and information extraction method in genomic studies, where multiple ...
  109. [109]
    A review of multi-omics data integration through deep learning ...
    Jul 19, 2023 · In this review, we systematically evaluate the recent trends in multi-omics data analysis based on deep learning techniques and their application in disease ...
  110. [110]
    MOGATFF: An Explainable Multi-Omics Prediction Model with ...
    Aug 3, 2025 · MOGATFF: An Explainable Multi-Omics Prediction Model with Feature Enhancement for Genotype-Phenotype Association Analysis. August 2025. DOI: ...
  111. [111]
    Breaking binary in cardiovascular disease risk prediction - Nature
    Jan 13, 2025 · Polygenic risk scores (PRS) and composite ML-based algorithms help shift the paradigm away from binary predictions towards more comprehensive continuum models.
  112. [112]
    A review of disease risk prediction methods and applications in the ...
    Mar 24, 2024 · Leveraging functional annotations in genetic risk prediction for human complex diseases ... Polygenic risk scores and the prediction of common ...4 Epigenomics-Based Risk... · 6 Metabolomics-Based Risk... · 9 Risk Prediction In Target...
  113. [113]
    Design of highly functional genome editors by modelling CRISPR ...
    Jul 30, 2025 · CRISPR-based gene editors derived from microorganisms, although powerful, often show notable functional tradeoffs when ported into non-native ...
  114. [114]
    The Encyclopedia of DNA Elements (ENCODE)
    Sep 17, 2023 · ENCODE is a public research consortium aimed at identifying all functional elements in the human and mouse genomes.
  115. [115]
    Project Overview - ENCODE
    The goal of ENCODE is to build a comprehensive parts list of functional elements in the human genome, including elements that act at the protein and RNA levels.
  116. [116]
    ENCODE Pilot Project - National Human Genome Research Institute
    Oct 18, 2012 · The Encyclopedia of DNA Elements (ENCODE) Pilot Project launched in September 2003 to identify all functional elements in the human genome ...
  117. [117]
    Data navigation on the ENCODE portal | Nature Communications
    Oct 30, 2025 · Spanning two decades, the collaborative ENCODE project aims to identify all the functional elements within human and mouse genomes.
  118. [118]
    Data standards - ENCODE
    ENCODE uses standards for epigenomic assays, including DNA binding, accessibility, methylation, 3D structure, transcription, RNA, and single-cell assays.
  119. [119]
    An integrated encyclopedia of DNA elements in the human genome
    Sep 5, 2012 · The Encyclopedia of DNA Elements (ENCODE) project aims to delineate all functional elements encoded in the human genome. Operationally, we ...
  120. [120]
    ENCODE project
    Experiment search · Experiment matrix · ChIP-seq matrix · Human and mouse body maps · Functional genomics series · Single-cell experiments.Project Overview · Getting Started · Data release policy · Citing ENCODE
  121. [121]
    Genotype-Tissue Expression (GTEx) - NIH Common Fund
    The GTEx (Genotype-Tissue Expression) Project identified genetic variants that influence how genes are turned on and off in human tissues and organs. Genetic ...
  122. [122]
    Adult GTEx Project - GTEx Portal
    The Adult Genotype Tissue Expression (GTEx) Project is a comprehensive public resource to study human gene expression and regulation, and its relationship to ...
  123. [123]
    The GTEx Consortium atlas of genetic regulatory effects across ...
    The GTEx project was launched in 2010 with the aim of building a catalog of genetic effects on gene expression across a large number of human tissues to ...
  124. [124]
  125. [125]
    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 ...
  126. [126]
    Alliance of Genome Resources Portal: unified model organism ...
    Sep 25, 2019 · The Alliance web portal (www.alliancegenome.org) provides a single point of access to multiple types of genetic and genomic data from diverse model organisms.Missing: formation | Show results with:formation
  127. [127]
    The alliance of genome resources: transforming comparative ...
    The mission of the Alliance is to support comparative genomics as a means to investigate the genetic and genomic basis of human biology, health, and disease.
  128. [128]
    The Alliance of Genome Resources (Alliance) Consortium
    Jul 17, 2024 · Current Members and Principal Investigators · FlyBase. Brian Calvi, Ph. · Mouse Genome Database (MGI). *Carol Bult, Ph. · Rat Genome Database (RGD).
  129. [129]
    Harmonizing model organism data in the Alliance of Genome ...
    Feb 25, 2022 · The Alliance has harmonized cross-organism data to provide useful comparative views of gene function, gene expression, and human disease relevance.
  130. [130]
    Updates to the Alliance of Genome Resources central infrastructure
    Abstract. The Alliance of Genome Resources (Alliance) is an extensible coalition of knowledgebases focused on the genetics and genomics of intensively stud.
  131. [131]
    Alliance of Genome Resources: Home
    Access official announcements, ask questions, and view discussions with other members of the Alliance Community. Join today to stay up-to-date. Members. FlyBase.Categories · AllianceMine · Downloads · GOCMissing: formation 2019 MGI