Fact-checked by Grok 2 weeks ago

Regulation of gene expression

Regulation of gene expression is the multifaceted process by which cells control the production of functional gene products, such as proteins or non-coding RNAs, in response to developmental, environmental, and physiological signals, ensuring precise spatiotemporal coordination of genetic activity. This regulation occurs at multiple levels, from and transcription initiation to mRNA processing, , and protein stability, allowing organisms to adapt efficiently while conserving cellular resources. In eukaryotes, where the contains approximately 20,000 protein-coding genes but only 1.5% of DNA directly encodes them, such control mechanisms generate vast functional diversity through processes like . At the core of transcriptional regulation lie cis-regulatory elements and factors that orchestrate the assembly of the transcription machinery. Promoters, typically 35–40 base pairs around the transcription start site, include motifs like the or initiator (Inr) sequences that recruit and general transcription factors for basal transcription. Enhancers, distal DNA segments often spanning over 400,000 sites in the and covering more than 10% of it, enhance transcription by looping to interact with promoters, marked by modifications such as H3K4me1 and H3K27ac. Insulators, meanwhile, prevent unwanted enhancer-promoter or spreading through proteins like and cohesin-mediated looping. Central to these interactions are transcription factors (TFs), sequence-specific DNA-binding proteins that serve as master regulators of by activating or repressing transcription through recruitment of coactivators, corepressors, or chromatin-modifying complexes. The encodes over 1,600 such TFs, classified into families based on DNA-binding domains, including C2H2-zinc finger (the largest with 747 members), homeodomain, and basic helix-loop-helix proteins. Epigenetic mechanisms further fine-tune this process; for instance, silences genes by inhibiting TF binding, while histone acetylation opens for accessibility. Beyond transcription, ensures mRNA quality and abundance, involving capping, splicing, , and degradation pathways, often modulated by microRNAs that target specific transcripts for silencing. Translational control, occurring in the via recruitment and initiation factors, and post-translational modifications like ubiquitination for protein degradation, provide rapid response layers. Gene expression dynamics exhibit stochastic bursting and cellular heterogeneity, driven by factors such as abundance and states, which enable adaptation to stimuli like or . This intricate regulatory network is fundamental to multicellular organization, immune responses, and disease ; dysregulation contributes to conditions like and cancer by altering or timing.

Fundamental Concepts

Levels of Gene Regulation

Gene expression encompasses the multistep process by which genetic information encoded in DNA is transcribed into (mRNA) and subsequently translated into functional proteins that perform cellular activities. This regulation is essential for cellular efficiency, as uncontrolled expression would lead to wasteful energy expenditure on synthesizing unneeded proteins, potentially compromising survival and function. The foundational understanding of multi-level gene regulation emerged from François Jacob and Jacques Monod's 1961 operon model, which illustrated how bacterial genes could be coordinately controlled at the transcriptional level in response to environmental signals, inspiring the recognition of regulatory opportunities across the entire gene expression pathway.80072-7) Subsequent research expanded this to identify six primary levels of control: transcriptional initiation, where factors determine the rate and specificity of mRNA synthesis from DNA templates; RNA processing, involving modifications like capping, splicing, and polyadenylation that prepare mature mRNA; RNA transport and stability, which govern the export of mRNA from the nucleus (in eukaryotes) and its degradation rate to fine-tune available transcripts; translational initiation, regulating the recruitment of ribosomes to mRNA for protein synthesis; post-translational modifications, such as phosphorylation or ubiquitination, that alter protein activity, localization, or interactions; and protein degradation, mediated by pathways like the proteasome, which rapidly removes proteins to adjust their levels dynamically. These levels allow cells to respond precisely to internal and external cues, with each stage offering independent yet interconnected points of intervention.80072-7) A key feature of this hierarchical regulation is combinatorial , where inputs from multiple regulatory levels are integrated to achieve precise timing, , and of gene products, enabling complex cellular responses that a single level could not support alone. For instance, signals affecting transcription may be modulated by post-transcriptional controls, ensuring robust and adaptable expression patterns. This multilayered integration enhances specificity and efficiency, allowing organisms to coordinate thousands of genes with minimal genomic redundancy.

Prokaryotic versus Eukaryotic Differences

Prokaryotes and eukaryotes exhibit distinct strategies for regulating , largely shaped by their cellular structures and lifestyles. In prokaryotes, the absence of a nuclear membrane allows transcription and to occur simultaneously in the , enabling rapid and efficient responses to environmental changes through primarily transcriptional control. This coupling means that regulatory mechanisms can immediately influence protein synthesis without intermediate processing steps, optimizing resource use in single-celled organisms that must quickly adapt to availability or . For instance, genes involved in metabolic pathways are often organized into operons, allowing coordinated expression of multiple genes from a single promoter in response to specific signals, as exemplified by the in responding to presence. In contrast, eukaryotes possess a that physically separates transcription in the from translation in the , introducing opportunities for at multiple stages beyond transcription. This compartmentalization supports the complexity of multicellular organisms, where must be finely tuned for , , and tissue-specific functions. As a result, eukaryotic emphasizes post-transcriptional modifications, such as mRNA stability and localization, as well as translational and post-translational controls, in addition to transcriptional mechanisms. The nuclear barrier also necessitates to access DNA, adding an extra layer of control that is absent in prokaryotes. Quantitatively, prokaryotic gene regulation occurs almost entirely at the transcriptional level, with estimates indicating that the vast majority—often described as the primary mode—of happens here to conserve in resource-limited environments. Eukaryotes, however, distribute across levels, with transcriptional remaining significant but complemented by substantial post-transcriptional and epigenetic contributions, reflecting the need for precise, long-term modulation in complex genomes. This distribution arises from barriers that restrict DNA accessibility, requiring additional regulatory steps. From an evolutionary perspective, the simplicity of prokaryotic systems facilitates inducible operon-based regulation for immediate environmental adaptation, aligning with their unicellular, fast-replicating nature. Eukaryotic complexity, evolved through endosymbiosis and multicellularity, demands stable epigenetic memory to maintain cell identities across generations, such as through heritable modifications that ensure developmental programs persist. This shift underscores how prokaryotic "default on" logic contrasts with eukaryotic "default off" states, prioritizing repression in larger genomes to prevent aberrant expression.

Transcriptional Regulation

Mechanisms in Prokaryotes

In prokaryotes, is predominantly regulated at the transcriptional level to enable rapid adaptation to environmental changes, given their unicellular nature and lack of nuclear compartmentalization. This regulation often involves simple, direct mechanisms that control the initiation of transcription by , allowing coordinated expression of functionally related genes. A key feature of prokaryotic transcriptional regulation is the operon, a cluster of contiguous structural genes transcribed as a single polycistronic mRNA from a shared promoter region upstream. The operon structure facilitates coordinate regulation, where environmental signals modulate access to the promoter via regulatory DNA sequences called operators, typically located between the promoter and the first structural gene. For instance, in the lac operon of , the operator serves as a for the protein, which blocks progression in the absence of , thereby preventing transcription of genes encoding lactose-metabolizing enzymes. Transcription initiation in prokaryotes requires the holoenzyme, which includes a core enzyme and a (σ) factor that confers promoter specificity by recognizing conserved promoter sequences, such as the -10 and -35 boxes. The σ70 factor directs transcription of most genes under standard conditions, while alternative factors, like σS during , redirect the holoenzyme to distinct promoters for adaptive responses. and activators further fine-tune this process; LacI exemplifies a that binds the with high affinity in its apo form, dissociating upon binding to allow transcription. Conversely, activators such as the (CAP) in the enhance binding to the promoter when complexed with cyclic AMP (cAMP) during glucose scarcity, illustrating relief. In the arabinose operon, the AraC protein acts dually as a repressor in the absence of arabinose and an activator upon binding, recruiting to the promoter. An additional layer of transcriptional control in prokaryotes is , a that terminates transcription prematurely based on mRNA secondary structure formation in the leader region. In the () operon of E. coli, low levels cause the to stall at tandem Trp codons in the leader coding sequence due to scarce charged tRNA-Trp, favoring an antiterminator that allows transcription to proceed into the structural genes; high levels enable rapid , allowing a to form and halt transcription before structural genes are reached. This couples transcription to and availability, providing sensitive . Global regulation extends beyond individual operons through mechanisms like , where bacteria monitor population density via diffusible autoinducers. In Vibrio fischeri, the autoinducer N-acyl homoserine lactone accumulates at high cell densities, binding the LuxR receptor to activate transcription of genes, coordinating population-level behaviors such as light emission in symbiotic hosts. Mathematical models often describe these binding events using the Hill equation to capture regulation, particularly in repression. The fractional occupancy θ of a on its is given by \theta = \frac{[L]^n}{K_d + [L]^n}, where [L] is the (repressor) concentration, n is the Hill reflecting (n > 1 for positive cooperativity), and K_d is the indicating affinity. This equation derives from the applied to cooperative , assuming rapid and multiple binding sites that enhance affinity nonlinearly; for n=1, it reduces to the Michaelis-Menten form for non-cooperative . In prokaryotic contexts, such as the , this models how repressor tetramers achieve ultrasensitive switching.

Mechanisms in Eukaryotes

In eukaryotes, is achieved through intricate, combinatorial mechanisms that integrate signals from distant genomic elements and cellular pathways to control (Pol II) activity at promoters. Unlike simpler prokaryotic systems, eukaryotic regulation involves multi-subunit general transcription factors (GTFs) that assemble the preinitiation complex (PIC) and specific transcription factors (TFs) that modulate initiation rates in response to developmental cues or environmental stimuli. This complexity enables precise spatiotemporal control of , often spanning kilobases of DNA and involving architecture. Core promoter elements, such as the TATA box and initiator (Inr), serve as docking sites for GTFs to position Pol II accurately for transcription initiation. The TATA box, located approximately 25-35 base pairs upstream of the transcription start site, is recognized by the TATA-binding protein (TBP) subunit of TFIID, which bends DNA to facilitate subsequent recruitment of TFIIA and TFIIB. The Inr element, encompassing the start site, interacts with TFIID's TAF subunits to stabilize the complex, particularly in TATA-less promoters prevalent in higher eukaryotes. Together, these elements recruit the remaining GTFs (TFIIE, TFIIF, TFIIH) and Pol II, forming the holoenzyme that unwinds DNA via TFIIH's helicase activity to initiate synthesis. Specific TFs, such as activators, enhance transcription by binding upstream or downstream sites and recruiting coactivators through modular domains. Activators like contain a (DBD) that recognizes specific sequences and an activation domain (AD) rich in acidic residues that interacts with coactivators to bridge the PIC. The Mediator complex, a large multiprotein coactivator, serves as a central hub, with its head, middle, and tail modules contacting TF ADs and transmitting signals to Pol II's C-terminal domain (CTD) for and promoter clearance. This modular architecture allows combinatorial control, where multiple TFs synergize to amplify initiation rates. Enhancer-promoter looping enables long-range regulation by bringing distal enhancers into proximity with promoters, often mediated by the architectural proteins and . CTCF binds to convergent DNA motifs at enhancer and promoter boundaries, while cohesin extrudes chromatin loops until stalled at CTCF sites, stabilizing interactions that facilitate TF and recruitment. A classic example is the beta-globin locus control region (LCR), where multiple CTCF-bound hypersensitive sites loop to the promoter in erythroid cells, coordinating high-level expression during development. This looping is dynamic and cell-type specific, insulating genes from inappropriate activation. Signal-responsive regulation integrates extracellular cues via kinase cascades that post-translationally modify TFs to trigger rapid gene activation. The (MAPK) pathway exemplifies this, where growth factors activate a cascade (Raf-MEK-ERK) that phosphorylates the ETS-domain TF Elk-1 at serine residues in its . Phosphorylated Elk-1 binds the serum response element in immediate-early gene promoters like FOS, recruiting to boost Pol II pausing release and elongation. This mechanism underlies fast responses in processes such as neuronal plasticity. Recent discoveries highlight liquid-liquid phase separation (LLPS) as a mechanism concentrating TFs and coactivators into condensates at active promoters and enhancers. IDR-rich ADs of TFs like those in the Mediator complex drive LLPS, forming membraneless hubs that sequester Pol II and increase local reaction rates by orders of magnitude, particularly at super-enhancers. This phase-separated state, observed in the , enhances transcriptional bursting and fidelity by compartmentalizing machinery away from nonspecific interactions. TF binding kinetics to promoters can be modeled using Michaelis-Menten equations, adapted to describe transcription initiation rates as a saturable process dependent on TF concentration. The rate v of initiation follows: v = \frac{V_{\max} [\text{TF}]}{K_m + [\text{TF}]} where V_{\max} is the maximum rate at saturating TF levels, [\text{TF}] is the free TF concentration, and K_m (the ) reflects binding affinity. This framework predicts ultrasensitive responses at low TF levels and plateauing at high occupancy, aligning with observed dose-dependent activation in eukaryotic systems.

Post-Transcriptional Regulation

RNA Processing and Stability

In eukaryotic cells, RNA processing and stability represent critical post-transcriptional mechanisms that refine primary transcripts into mature mRNAs suitable for , while also controlling their lifespan to fine-tune . These processes occur co- and post-transcriptionally, involving modifications that enhance from the , protect against degradation, and modulate decay rates based on cellular needs. Dysregulation of these steps can lead to imbalances in , contributing to diseases such as cancer. The 5' capping of pre-mRNA involves the addition of a 7-methylguanosine (m⁷G) cap structure shortly after transcription initiation, typically after about 20-30 nucleotides are synthesized. This cap is formed by three enzymatic steps: RNA 5'-triphosphatase removes the gamma phosphate, guanylyltransferase adds GMP, and methyltransferase adds a methyl group to form m⁷GpppN. The cap protects the mRNA 5' end from exonucleolytic degradation by 5'→3' exoribonucleases such as Xrn1, thereby enhancing stability, and also facilitates nuclear export via binding to the nuclear cap-binding complex (CBC) and promotes translation initiation by interacting with eIF4E. This modification was first identified in eukaryotic mRNAs, including viral transcripts, underscoring its conserved role in mRNA function. At the 3' end, polyadenylation entails cleavage of the pre-mRNA downstream of a polyadenylation signal (AAUAAA) by the cleavage and polyadenylation specificity factor (CPSF) complex, followed by addition of a (typically 200-250 adenines) by poly(A) polymerase. The , bound by poly(A)-binding proteins (PABPs), stabilizes the mRNA by preventing 3'→5' exonucleolytic attack and aids in circularization for efficient . In certain transcripts, such as those encoding cytokines like tumor necrosis factor-alpha (TNF-α), AU-rich elements (AREs) in the 3' (UTR) promote rapid deadenylation and turnover, ensuring during immune responses; for instance, AREs trigger within hours via recruitment of decay factors like TTP. Alternative splicing further diversifies mRNA isoforms from a single pre-mRNA, regulated by splicing factors including serine/arginine-rich (SR) proteins and heterogeneous nuclear ribonucleoproteins (hnRNPs). SR proteins, such as SRSF1, bind exonic splicing enhancers to promote exon inclusion by recruiting the spliceosome, while hnRNPs like hnRNP A1 bind silencers to repress exon usage, often through steric hindrance or looping out of exons. This antagonism enables tissue- or condition-specific isoform production; a classic example is the Sex-lethal (Sxl) gene in Drosophila melanogaster, where female-specific Sxl protein blocks a male-specific 3' splice site via hnRNP-like binding, leading to functional Sxl isoforms that drive sex determination. Nonsense-mediated decay (NMD) serves as a pathway to degrade mRNAs harboring premature termination codons (PTCs), preventing production of truncated proteins. Triggered during pioneer translation, NMD relies on the up-frameshift (UPF) proteins (UPF1, UPF2, UPF3), where UPF1 activity is enhanced upon PTC recognition more than 50-55 upstream of an exon-exon (the "50-nt rule" or up-frameshift rule, derived from frameshift mutants). This leads to recruitment of endonucleases or deadenylases, resulting in rapid decay and quality control of ~5-10% of transcripts, including natural regulatory mRNAs. Epitranscriptomic modifications provide another layer of regulation, with N6-methyladenosine (m6A) being the most abundant internal modification in eukaryotic mRNAs. m6A is dynamically installed by writer complexes (e.g., METTL3-METTL14) near stop codons and 3' UTRs, recognized by reader proteins such as YTHDF2 to promote decay via deadenylation or recruitment to NMD, while other readers like YTHDF1 enhance translation. Erasers like FTO and ALKBH5 remove m6A, allowing reversible control. Dysregulated m6A contributes to cancer, immunity, and , with recent studies (as of 2025) elucidating its roles in mRNA , splicing , and stress responses. RNA-binding proteins (RBPs) dynamically influence mRNA stability by binding specific sequences, often in 3' UTRs. For example, HuR (ELAVL1) stabilizes proto-oncogene mRNAs such as those encoding cyclins A and B1 by binding AU-rich or U-rich elements, counteracting decay factors and extending half-lives during ; HuR shuttles from to to exert this effect. Other RBPs may destabilize transcripts, integrating signals like or growth factors. MicroRNAs can briefly interact with these elements to repress stability, but detailed mechanisms overlap with translational control. mRNA stability is quantitatively modeled using exponential decay kinetics, where the concentration of mRNA at time t follows [ \text{mRNA}(t) ] = [ \text{mRNA} ]_0 e^{-kt} , with k as the degradation rate constant and half-life t_{1/2} = \frac{\ln 2}{k}. This model, fitted to transcription-inhibited time courses, reveals half-lives ranging from minutes (e.g., for immediate-early genes) to days (e.g., for housekeeping genes), highlighting how processing modifications and RBPs tune decay rates for precise temporal control.

Translational Control

Translational control regulates the rate and fidelity of protein synthesis from mRNA, allowing cells to rapidly adjust protein levels in response to environmental cues without altering transcription or mRNA stability. This layer of gene expression fine-tunes the by modulating initiation, elongation, and termination, often through of initiation factors or RNA-binding proteins that influence recruitment. In eukaryotes, such mechanisms are particularly prominent during stress, where global is suppressed to conserve while selective mRNAs are preferentially translated. A key mechanism involves the of at serine 51 by stress-activated , which inhibits guanine nucleotide exchange and reduces ternary complex formation, thereby halting cap-dependent translation initiation for most mRNAs. The PERK (PKR-like ER ) is activated during endoplasmic reticulum () stress, such as unfolded protein accumulation, leading to eIF2α phosphorylation that globally represses translation while sparing mRNAs with upstream open reading frames (uORFs) or internal ribosome entry sites (IRES), like ATF4, which promote adaptive responses. This selective translation ensures cell survival under stress by prioritizing proteins involved in defense and . Internal ribosome entry sites (IRES) enable cap-independent by directly recruiting to structured regions in the (UTR) of mRNAs, bypassing the need for and eIF4G under conditions where cap-dependent initiation is impaired, such as or viral infection. In cellular responses, IRES elements in mRNAs like that of hypoxia-inducible factor-1α (HIF-1α) facilitate translation during oxygen deprivation, allowing HIF-1α protein accumulation to activate genes for and despite reduced global translation. Viral IRES, such as those in or , similarly hijack host ribosomes for efficient replication, highlighting the evolutionary conservation of this mechanism. MicroRNAs (miRNAs) exert translational repression by base-pairing with target sites in the 3' UTR of mRNAs via (AGO) proteins within the (RISC), which recruits factors to block initiation or elongation and promotes mRNA deadenylation and decay. In development, the miRNA lin-4 binds multiple sites in the 3' UTR of lin-14 mRNA through imperfect complementarity, reducing LIN-14 protein levels to time transitions from larval to adult stages without fully degrading the transcript. This discovery of miRNAs and their regulatory role was recognized with the 2024 in or awarded to Victor Ambros and Gary Ruvkun. This AGO-mediated inhibition often combines translational silencing with gradual mRNA turnover, providing precise spatiotemporal control over gene expression. Beyond translation, post-translational modifications such as ubiquitination rapidly degrade nascent or excess proteins to maintain , with ubiquitin ligases targeting specific substrates for proteasomal . In the , SCF (Skp1-Cullin-F-box) and APC/C (anaphase-promoting complex/cyclosome) ligases ubiquitinate —such as during or in G1—for timely degradation, ensuring progression through checkpoints and preventing uncontrolled proliferation. Dysregulation of these ligases, as seen in cancers, leads to cyclin stabilization and aberrant . Ribosome stalling during elongation triggers no-go decay (NGD) pathways, where collided ribosomes are recognized by factors like Dom34 (Pelota in eukaryotes) and Hbs1, leading to mRNA cleavage upstream of the stall site and ribosome rescue to prevent toxic aggregates. Stalls often arise from stable secondary structures, rare codons, or nascent chain misfolding, activating endonucleases like SMG6 for mRNA fragmentation and subsequent exonucleolytic degradation. This quality control mechanism, conserved from yeast to mammals, safeguards the proteome by eliminating defective transcripts and rescuing stalled ribosomes for reuse. Translation efficiency (TE), defined as the ratio of protein output to steady-state mRNA levels, quantifies how sequence features modulate ribosomal output, where TE = \frac{\text{protein output}}{\text{mRNA level}}. Upstream ORFs (uORFs) in the 5' UTR often reduce TE by sequestering ribosomes and promoting leaky scanning, as seen in stress-response genes like , while mRNA secondary structures impede initiation by hindering eIF4A activity, lowering TE by up to 10-fold in structured leaders. These elements allow nuanced control, with uORFs and folds integrating signals from prior regulatory layers like RNA stability to dictate final protein yields.

Epigenetic and Chromatin Regulation

DNA Modifications

DNA modifications refer to chemical alterations of DNA bases that do not change the underlying sequence but influence accessibility and , acting as heritable epigenetic marks primarily in eukaryotes. The most prominent of these is cytosine , where a is added to the 5-position of residues, predominantly at CpG dinucleotides. This modification is catalyzed by (DNMT) enzymes, including for maintenance and DNMT3A/3B for , and typically correlates with transcriptional repression by recruiting repressive protein complexes or inhibiting binding at promoters and enhancers. In mammals, plays a critical role in , where parent-of-origin-specific patterns silence one of certain genes, such as the (Igf2) locus, ensuring proper embryonic development. Other notable DNA modifications include N6-methyladenine (6mA), an emerging epigenetic mark in eukaryotes such as thermophila. 6mA is deposited de novo by methyltransferases like AMT2 and AMT5 and maintained semi-conservatively by AMT1 during , enabling heritability. It influences by modulating transcription, organization, replication, and stress responses; for example, its deletion alters developmental gene patterns and reduces cell viability. As of 2025, 6mA detection and functional studies continue to expand understanding of its regulatory roles beyond prokaryotes. Demethylation counteracts to dynamically regulate , particularly during . This process is mediated by ten-eleven translocation () enzymes (TET1, TET2, TET3), which oxidize (5mC) to (5hmC) and further to 5-formylcytosine (5fC) and 5-carboxylcytosine (5caC), facilitating and passive dilution during replication. In early mammalian embryos, TET3-driven oxidation is essential for paternal demethylation in the , enabling zygotic activation by removing repressive marks and allowing expression of developmental genes. Unlike 5mC's repressive function, 5hmC often marks active regulatory regions; in postmitotic neurons, elevated 5hmC levels at gene bodies and enhancers correlate with transcriptional activation, as seen in hippocampal and cerebellar neurons where it promotes neuronal differentiation and . In prokaryotes, analogous DNA modifications occur through restriction-modification (RM) systems, where methyltransferases protect host DNA from restriction endonucleases that cleave unmethylated foreign DNA, such as from phages. These systems, first described in the 1960s, provided early insights into methylation as a heritable barrier to gene expression and inspired studies on eukaryotic epigenetics, highlighting conserved roles in genome defense and stability despite mechanistic differences. Aberrant global DNA hypomethylation is a hallmark of many cancers, often resulting from reduced DNMT activity or TET dysregulation, leading to oncogene activation; for instance, hypomethylation of retrotransposons and cancer-germline genes in colorectal and breast tumors drives genomic instability and aberrant expression. To map these modifications, remains the gold standard assay, treating DNA with to convert unmethylated cytosines to uracils while leaving 5mC (and 5hmC, with adjustments) intact, followed by amplification and next-generation sequencing for single-base resolution profiling of states across genomes. This technique, originally developed in the early 1990s, has enabled comprehensive atlases of patterns in development and disease, though it requires validation for distinguishing 5hmC from 5mC using oxidative bisulfite methods.

Histone and Chromatin Remodeling

Histone modifications and are essential epigenetic mechanisms that regulate gene accessibility by altering structure and compaction. , the core proteins around which DNA winds to form nucleosomes, undergo covalent modifications on their N-terminal tails, such as and , which influence the recruitment of transcription factors and other regulatory proteins. These changes, along with ATP-dependent remodeling complexes, dynamically reposition or evict nucleosomes to facilitate or repress transcription. In eukaryotes, this layer of regulation integrates with DNA modifications to fine-tune during development and in response to environmental cues. Histone acetylation involves the addition of acetyl groups to residues primarily by acetyltransferases (s), such as p300, which neutralizes the positive charge on lysines, reducing the affinity between histones and negatively charged DNA. This charge neutralization loosens structure, promoting an open conformation that enhances access for transcriptional machinery. For instance, acetylation of at 27 (H3K27ac) is particularly enriched at active enhancers and promoters, correlating with increased . Conversely, histone deacetylases (HDACs) remove these acetyl groups, restoring positive charges and facilitating chromatin compaction to repress transcription. The balance between HAT and HDAC activities maintains dynamic equilibrium, with dysregulation implicated in various cellular processes. Emerging modifications include lactylation, where lactate-derived lactyl groups are added to residues, often mediated by M2 () and p300 in high-glycolysis contexts. This mark promotes activation by enhancing accessibility and facilitating enhancer-promoter looping, as observed in where it upregulates steroidogenesis genes like CYP17A1. As of 2025, lactylation represents a metabolic-epigenetic link in disease pathogenesis. Histone methylation presents diverse outcomes depending on the specific residue and degree of methylation (mono-, di-, or tri-). Trimethylation of at 4 () is a hallmark of active promoters, where it recruits readers like TFIID to initiate transcription. This mark is deposited by methyltransferases such as SET1 and is prevalent at the transcription start sites of expressed genes. In contrast, trimethylation of H3 at 27 (), catalyzed by the Polycomb repressive complex 2 (PRC2), mediates gene repression, particularly at developmental loci like , where it maintains silencing during embryogenesis. Polycomb group proteins enforce this repression by compacting and preventing activator binding, ensuring proper body patterning. ATP-dependent complexes, such as the family, use the energy from to slide, eject, or restructure nucleosomes, thereby exposing or occluding DNA regulatory elements. The mammalian complex, containing the BRG1 subunit, is crucial for enhancer activation, where it repositions nucleosomes to allow binding and looping between enhancers and promoters. For example, BRG1 facilitates the activation of mesoderm-specific enhancers during differentiation by increasing accessibility at key loci. These remodelers often cooperate with modifications to propagate open states. Histone variants and higher-order chromatin structures further diversify regulation. The variant H2A.Z, incorporated into nucleosomes at promoters and enhancers by the SWR1 complex, destabilizes nucleosomes to promote transcriptional activation while also poising genes for repression in certain contexts. Super-enhancers, clusters of enhancers densely occupied by transcriptional machinery and marked by high H3K27ac levels, drive robust expression of cell identity genes; their discovery highlighted phase-separated domains that concentrate factors for amplified signaling. These structures, often spanning large genomic regions, integrate multiple signals to sustain high-level transcription.00392-9) Bivalent domains, characterized by the coexistence of activating and repressive marks at promoters, are prominent in embryonic stem cells and maintain developmental genes in a poised state—repressed yet primed for rapid activation upon differentiation cues. This bivalency ensures lineage flexibility, with resolution of marks directing cell fate decisions, such as in clusters. Genome-wide mapping of these modifications relies on followed by sequencing (ChIP-seq), which cross-links proteins to DNA, immunoprecipitates with specific antibodies, and sequences enriched fragments to identify modification landscapes at base-pair resolution. ChIP-seq has revolutionized the field by revealing modification patterns across entire genomes, enabling the annotation of regulatory elements.

Regulatory Elements and Networks

Promoters, Enhancers, and Silencers

Promoters are cis-regulatory DNA sequences located upstream of the transcription start site (TSS) that serve as binding platforms for the transcription initiation complex, including and general transcription factors. The core promoter, typically spanning -40 to +40 pairs relative to the TSS, contains essential motifs such as the (consensus TATAAA, located ~ -25 to -35 bp upstream) and the Initiator (Inr, consensus YYANWYY, where Y is , N any , W A or T, centered at the TSS), which direct precise transcription initiation. These core recruit the TFIID complex, with TBP binding the and TAFs recognizing the Inr, enabling basal transcription in eukaryotes. Proximal promoter , extending from ~ -40 to -200 bp upstream, include motifs like the TFIIB recognition element (BRE) and downstream promoter element (DPE, consensus RGWYVT, located +28 to +32 bp downstream of TSS), which fine-tune initiation efficiency and interact with specific general transcription factors. Promoters vary in architecture based on gene function: housekeeping promoters, which drive constitutive expression of essential genes like those for basic metabolism, often feature CpG islands—GC-rich regions lacking TATA boxes but rich in proximal elements for broad, stable transcription across cell types. In contrast, tissue-specific promoters, regulating genes like those involved in developmental or specialized functions, typically contain boxes and fewer CpG islands, enabling inducible or cell-type-restricted activation through interactions with lineage-specific transcription factors. For instance, the β-globin promoter in erythroid cells relies on a -driven core with proximal CCAAT and motifs for high-level, stage-specific expression. Enhancers are distal cis-regulatory elements, often located thousands of base pairs away from their target promoters (up to megabases), that boost transcription by looping to contact promoters and recruiting co-activators. Unlike promoters, enhancers function in an orientation-independent manner, meaning their activity persists regardless of whether they are upstream, downstream, or inverted relative to the gene, due to folding that brings them into proximity. They exhibit strong specificity, with clusters of binding sites tailored to types; for example, the (Igh) enhancers in B , such as the intronic enhancer (Eμ) and 3' regulatory region, drive high-level, B-cell-specific expression of rearranged genes during development. This specificity arises from combinatorial binding of factors like Pax5 and E2A, which stabilize enhancer-promoter loops in mature B . Silencers are repressive cis-elements that inhibit transcription, either by binding repressor proteins that block activator access or by insulating promoters from nearby enhancers. These elements can be proximal or distal and often overlap with enhancers in bidirectional regulatory landscapes, allowing context-dependent switching between activation and repression. A prominent example involves -bound insulators, such as those in the β-globin locus control region, where CTCF binding at boundary sites prevents inappropriate enhancer-promoter contacts, thereby repressing ectopic activation in non-erythroid cells. -mediated silencing relies on its zinc-finger domains forming loops that physically separate regulatory domains, maintaining spatial organization to enforce cell-type-specific repression. The higher-order architecture of these elements is organized into topologically associating domains (TADs), self-interacting regions averaging 1 Mb in size that confine enhancers, silencers, and promoters to limit promiscuous interactions. TADs are delimited by and cohesin-bound boundaries, creating regulatory landscapes where intra-TAD enhancer-promoter looping drives coordinated expression, while inter-TAD contacts are restricted. Disruptions to TAD structure, such as deletions or inversions at boundaries, can lead to pathological misregulation; for instance, TAD boundary alterations in the or HOXD loci cause limb malformations or synpolydactyly by allowing ectopic enhancer access, highlighting their role in developmental diseases. In cancer, TAD disruptions similarly promote activation, as seen in enhancer hijacking events. Large-scale discovery of promoters and enhancers has been advanced by the Encyclopedia of DNA Elements (ENCODE) project, initiated in 2003, which has mapped over 100,000 putative enhancers in the human genome through integrative analyses of chromatin accessibility (DNase-seq), histone marks (e.g., H3K27ac enrichment at active enhancers), and transcription factor occupancy across hundreds of cell types. ENCODE data reveal that enhancers comprise ~8-10% of the non-coding genome, with many forming "super-enhancers" at key lineage genes, providing a comprehensive atlas for understanding regulatory element distribution. Prediction of transcription factor binding to these elements relies on position weight matrices (PWMs), probabilistic models representing binding site motifs as 4xL matrices (L = motif length) where each position scores nucleotide preferences. The binding score for a sequence is calculated as the sum over positions j of \log_2 \left( \frac{p_{b_j}}{f_b} \right), where p_{b_j} is the frequency of base b at position j in aligned binding sites, and f_b is the background frequency (e.g., 0.25 for uniform DNA). \text{Score} = \sum_{j=1}^{L} \log_2 \left( \frac{p_{b_j}}{f_b} \right) Higher scores indicate stronger predicted binding affinity, enabling genome-wide scans to annotate motifs in promoters and enhancers.

Gene Regulatory Circuits

Gene regulatory circuits integrate multiple regulatory elements and transcription factors to form dynamic networks that process environmental signals and ensure precise, robust patterns of gene expression. These circuits often rely on recurring structural motifs, or network motifs, that perform specific computational functions such as signal filtering, response acceleration, or stable state maintenance. By combining positive and negative interactions, these networks enable cells to respond adaptively to stimuli while minimizing noise and variability in expression levels. Feed-forward loops (FFLs) are among the most prevalent motifs in bacterial regulation, consisting of a regulatory X that controls both a target Z directly and an intermediary Y, which in turn regulates Z. In coherent FFLs, the direct and indirect paths from X to Z have the same regulatory sign (both or both repression), enabling functions like sign-sensitive that out brief, noisy signals while allowing sustained inputs to propagate; for instance, in the (ara) utilization system of , the AraC and global regulator CRP form a coherent type-1 FFL that araBAD until arabinose levels persist, reducing premature expression in fluctuating environments. Incoherent FFLs, where the paths have opposite signs, accelerate responses and can sharpen pulses or generate ; the (gal) system in E. coli exemplifies this, with GalS and CRP forming an incoherent FFL that rapidly induces gal genes upon exposure while adapting to sustained signals. These motifs enhance circuit robustness by processing inputs in a nonlinear manner, as demonstrated in comparative analyses of E. coli regulatory networks. Negative feedback loops contribute to circuit stability by having a suppress its own expression, thereby buffering fluctuations and maintaining steady-state levels. In autosuppression, a binds its own promoter to limit overproduction; the (LacI) in E. coli exemplifies this, where LacI autoregulates the lacI promoter via operator O3, ensuring consistent concentrations despite variations in growth conditions and reducing cell-to-cell variability in induction. This motif not only stabilizes protein levels but also speeds response times compared to simple regulation, as the rapid dilution of excess product accelerates adaptation to new signals. Gene regulatory circuits often distinguish between up-regulation () and down-regulation (repression) to fine-tune responses to availability or . Inducible systems activate upon detecting an environmental signal, such as in the where σ32 up-regulates chaperones like DnaK in E. coli upon temperature elevation, enabling rapid protein refolding under . Repressible systems, conversely, maintain basal expression that is attenuated when end products accumulate; the tryptophan () operon in E. coli is repressed when binds the , forming a trp-TrpR complex that blocks trp promoter activity and conserves resources during abundance. These opposing strategies allow circuits to efficiently allocate cellular resources based on metabolic needs. Analyses of regulatory networks in the 2000s revealed the prevalence of specific motifs shaped by evolutionary pressures for functionality. Uri Alon's work identified FFLs and feedback loops as overrepresented in E. coli and transcription networks, comprising a significant fraction of three-node subgraphs due to their roles in dynamic control. Toggle switches, mutual repression motifs between two genes, enable —two stable expression states that toggle based on inputs—facilitating cell fate decisions; the synthetic toggle switch constructed in E. coli using lacI and genes demonstrated this, maintaining either state until perturbed by inducers like IPTG, with applications in modeling developmental switches. Stochastic models capture the inherent noise in arising from low molecule numbers, using algorithms like Gillespie's to track probabilistic transitions. In these birth-death processes, is modeled as a where "birth" events (transcription) occur at rate proportional to promoter activity, and "death" events (degradation) at a basal rate, with the P(n, t) of having n molecules evolving via the chemical master equation: \frac{dP(n,t)}{dt} = b(n-1)P(n-1,t) - b(n)P(n,t) + d(n+1)P(n+1,t) - d(n)P(n,t) Here, b(n) and d(n) are birth and death rates, respectively, allowing of in circuits like loops to predict variability in expression levels. Synthetic biology has engineered circuits to validate and extend natural motifs, demonstrating their . The repressilator, a ring of three repressors (lacI, , ) in E. coli, produces sustained oscillations in protein levels with periods of about 40 minutes, driven by delayed that mimics circadian rhythms and highlights how simple motifs can generate temporal patterns when interconnected.

Examples in Biology and Disease

Developmental Gene Regulation

Developmental gene regulation orchestrates the precise temporal and spatial activation of genes during development, ensuring proper patterning and differentiation in model systems like and vertebrates. In these processes, is controlled through intricate networks that integrate signaling pathways, epigenetic modifications, and cis-regulatory elements to generate diverse cell fates from a uniform . This regulation is exemplified by conserved mechanisms such as clusters, which establish body axes, and gradients that interpret positional information along embryonic axes. Hox gene clusters exhibit expression, where genes are activated sequentially along the anterior-posterior axis in a manner mirroring their , a phenomenon conserved from to s. This is mediated by dynamic looping that brings distant enhancers into proximity with promoters, facilitating coordinated activation. The balance between Polycomb group (PcG) proteins, which maintain repressive marks like to silence genes prematurely, and Trithorax group (TrxG) proteins, which promote active marks such as for sustained expression, ensures the timed onset of Hox transcription. In , PcG complexes compact the cluster early in embryogenesis, while TrxG factors progressively open domains from the anterior end; similar mechanisms operate in vertebrate Hox clusters, where looping events detected via highlight regulatory hubs. Segment polarity in Drosophila embryogenesis relies on Hedgehog (Hh) signaling gradients to refine parasegment boundaries, where Hh secreted from engrailed-expressing posterior cells diffuses anteriorly to induce wingless (wg) expression in adjacent cells. This creates a feedback loop: Wg, a Wnt family ligand, maintains engrailed in posterior cells while restricting its own domain, establishing alternating stripes of gene expression that polarize each segment. The Hh gradient's asymmetric range—shorter posteriorly due to Engrailed-mediated repression—ensures sharp boundaries, with threshold concentrations activating target genes like decapentaplegic (dpp) in broader domains. Mutations disrupting this pathway, such as in hh or en, lead to segment fusion, underscoring its role in patterning the ventral epidermis.90175-9) The maternal-to-zygotic transition (MZT) marks a critical phase in early mammalian embryos, involving epigenetic reprogramming that clears parental imprints through global DNA demethylation to activate the zygotic genome. In mice, paternal DNA undergoes active demethylation by TET3-mediated oxidation shortly after fertilization, while maternal DNA experiences passive loss over cleavage divisions, achieving near-total demethylation by the blastocyst stage. This reprogramming, coupled with histone modifications like H3K4me3 enrichment at promoters, enables zygotic transcription around the 2- to 8-cell stage, replacing maternal factors for lineage specification. Disruptions in this process, such as in TET3 knockouts, impair development, highlighting its essentiality for totipotency establishment. Morphogen gradients, such as Bicoid (Bcd) in Drosophila, provide positional cues for anterior-posterior patterning by forming concentration gradients that elicit threshold-dependent responses in target genes. Bcd protein, translated from maternally deposited anterior mRNA, diffuses posteriorly, creating an exponential gradient where high anterior levels activate genes like buttonhead for head structures, while lower posterior thresholds induce hunchback for thoracic segments. Target enhancers interpret these levels via cooperative binding sites, with affinity determining response sharpness; for instance, high-affinity sites in orthodenticle respond at lower Bcd concentrations than low-affinity ones in giant. This French flag model of interpretation ensures robust patterning despite gradient variability.00353-4) Evolutionary conservation of developmental regulation is evident in gene regulatory networks (GRNs) governing endomesoderm specification in sea urchins, as pioneered by Davidson's models in the 2000s. These GRNs integrate transcription factors like beta-catenin and Blimp1/ with cis-regulatory modules to drive sequential gene activation from fertilization through , forming a predictive framework for spatial . The provisional endomesoderm GRN, comprising over 40 genes wired by double-repression and activation motifs, reveals how ancient bilaterian circuitry persists, with rewiring in related species like sea stars altering outputs while preserving core logic. This approach underscores GRNs' utility in dissecting conserved developmental modules across phyla.

Dysregulation in Cancer and Neurological Disorders

Dysregulation of is a hallmark of cancer, where epigenetic modifications frequently silence tumor suppressor genes or aberrantly activate , driving uncontrolled proliferation and tumor progression. In , hypermethylation of the CDKN2A () promoter region serves as a key mechanism to transcriptionally repress this inhibitor, which normally halts progression; this silencing occurs in a significant proportion of cases and correlates with advanced disease stages and poor . Similarly, amplification of the in multiple cancer types, including lymphomas and solid tumors, often involves the hijacking of super-enhancers—clusters of densely occupied enhancers that amplify transcription. This phenomenon, identified in , repositions potent regulatory elements near , leading to its overexpression and the promotion of hallmarks such as sustained proliferation and evasion of apoptosis.00393-0) In neurological disorders, failures in gene regulatory mechanisms contribute to synaptic dysfunction, neurodegeneration, and behavioral pathologies. exemplifies transcriptional dysregulation in the dopamine reward pathway, where repeated drug exposure induces stable accumulation of the ΔFosB in dynorphin-expressing medium spiny neurons of the . This persistence arises from prolonged mRNA stabilization and reduced degradation, resulting in sustained activation of target genes that heighten reward sensitivity and reinforce compulsive behaviors long after drug cessation. Disruptions in learning and further highlight epigenetic vulnerabilities; in the , CREB orchestrates (LTP) by recruiting coactivators like CBP to increase histone at promoters of plasticity-related genes, such as BDNF. Pathological hypoacetylation impairs this process, weakening synaptic strengthening and , whereas administration of HDAC inhibitors elevates acetylation levels, enhances CREB-dependent transcription, and ameliorates deficits in rodent models of . Inherited and sporadic neurological conditions often stem from targeted regulatory defects. arises from expansion of CGG trinucleotide repeats (>200) in the of the gene, triggering CpG island hypermethylation and formation that silences expression; the resulting absence of fragile X mental retardation protein (FMRP), which regulates mRNA translation in dendrites, leads to and autism-like features. In (ALS), TDP-43 pathology—characterized by its nuclear depletion and cytoplasmic aggregation—disrupts splicing fidelity, promoting the inclusion of cryptic exons in transcripts of maintenance genes like STMN2 and UNC13A, thereby reducing functional protein levels and accelerating neurodegeneration. Emerging therapeutics leverage precise to restore proper . Since 2016, CRISPR-based epigenetic tools, such as dCas9 fused to TET demethylases, have enabled locus-specific removal of aberrant from silenced promoters, reactivating genes like tumor suppressors in cancer cells or FMR1 in Fragile X neurons without sequence alterations. These approaches demonstrate durable reactivation in preclinical models, including demethylation of hypermethylated CDKN2A in lines and FMR1 in patient-derived cells, paving the way for targeted interventions in both oncological and neurological contexts.

Methods for Studying Gene Regulation

Experimental Techniques

Experimental techniques for studying gene regulation encompass a range of methods designed to perturb, detect, and quantify regulatory processes at the molecular level. These approaches allow researchers to manipulate regulatory elements, measure (TF) binding, assess abundance, and evaluate the functional impact of genetic perturbations. From classical assays to modern genome-editing tools, these methods provide direct evidence of how genes are controlled in cellular contexts. Reporter assays are widely used to quantify the activity of regulatory elements such as promoters and enhancers by linking them to a , typically encoding , whose bioluminescent output is measured to reflect transcriptional activation or repression. In transient experiments, cells are introduced with constructs containing the driving expression, allowing rapid assessment of cis-regulatory function in response to stimuli or TFs; for instance, levels can increase up to 100-fold upon activation by specific enhancers. This technique was pioneered with the cloning and expression of the gene in mammalian cells, enabling sensitive, non-radioactive detection of changes. Knockout and knockdown strategies enable targeted disruption of genes involved in regulation, such as or components of regulatory networks, to observe downstream effects on . RNA interference (RNAi) uses double-stranded to silence specific genes post-transcriptionally by triggering mRNA degradation, as demonstrated in the discovery of potent interference in Caenorhabditis elegans where dsRNA reduced target gene activity by over 90% compared to single-stranded . For precise genomic edits, -Cas9 has revolutionized the field since 2012, allowing site-specific cleavage and modification of enhancers or TF loci via guide RNA-directed Cas9 nuclease, achieving editing efficiencies of 20-80% in mammalian cells and revealing regulatory roles, such as enhancer deletions altering target gene expression by 50-90%. These methods complement each other, with RNAi offering transient knockdown and providing stable, heritable changes. The (EMSA) detects direct interactions between TFs and motifs by observing the slower migration of protein- complexes in non-denaturing gels compared to free . In the assay, labeled probes containing putative binding sites are incubated with nuclear extracts, and shifts in electrophoretic mobility indicate binding, often confirmed by competition with unlabeled or supershifts with antibodies; binding affinities can be quantified via , revealing dissociation constants in the nanomolar range for specific TF- pairs. This technique, originally developed for quantifying lactose operon regulator binding in E. coli, remains a cornerstone for validating in vitro TF specificity before in vivo studies. To measure RNA levels as a proxy for , Northern blotting and reverse transcription quantitative (RT-qPCR) provide complementary approaches for assessing steady-state mRNA abundance pre- and post-regulatory events. Northern blotting involves size-fractionating total RNA on gels, transferring to membranes, and hybridizing with labeled probes to detect specific transcripts, offering size information and relative quantification; it was foundational for early studies, detecting mRNA differences across tissues with sensitivities down to 1-5 pg of target RNA. RT-qPCR, an advancement for precise quantification, reverses transcribes RNA to cDNA followed by real-time monitoring of amplification via fluorescent probes, enabling absolute or relative expression analysis with dynamic ranges exceeding 10^5-fold and efficiencies near 100%; introduced through kinetic monitoring of , it is now standard for validating regulatory changes, such as TF-induced fold increases in target mRNAs. Chromatin immunoprecipitation (ChIP) isolates protein-DNA complexes to map occupancy or modifications at regulatory sites, using antibodies to pull down crosslinked followed by or sequencing of associated DNA. The method crosslinks proteins to DNA with , shears , immunoprecipitates targets, and reverses crosslinks to recover DNA, enriching bound sequences 10-100-fold over input; it originated from studies showing H4 retention on active genes in , establishing 's utility for capturing dynamic interactions. A variant, ChIP-exo, enhances precision by trimming post-immunoprecipitation, defining binding sites to single-nucleotide resolution with near-zero background and improved detection of binding events compared to standard ChIP, as applied to genome-wide mapping in . These techniques integrate with computational analyses for broader regulatory insights but focus here on the core wet-lab workflows. A more recent advancement, Under Targets and Tagmentation (CUT&Tag), developed in 2019, enables efficient epigenomic profiling of modifications and TF binding using antibody-tethered transposases for targeted tagmentation, requiring as few as 1,000 cells and producing low-bias libraries with higher signal-to-noise than traditional . This method has become widely adopted by 2025 for its simplicity, cost-effectiveness, and compatibility with single-cell applications in gene regulation studies.

Computational and Genomic Approaches

Genomic approaches, particularly high-throughput sequencing techniques, have enabled systematic mapping of regulatory elements and their interactions across entire genomes, providing empirical data essential for understanding gene regulation. followed by sequencing (ChIP-seq) identifies protein-DNA interactions, such as binding sites and modifications, by immunoprecipitating fragments bound by specific proteins and sequencing the associated DNA. Developed in 2007, ChIP-seq has been widely adopted for genome-wide profiling, revealing regulatory landscapes in diverse cell types and conditions. Similarly, Assay for Transposase-Accessible using sequencing (), introduced in 2013, detects open regions by leveraging hyperactive Tn5 transposase to insert sequencing adapters into accessible DNA, requiring minimal cell input (as few as 500 cells) and facilitating the identification of promoters, enhancers, and insulators. These methods generate large datasets that highlight dynamic states influencing . Complementary to accessibility assays, RNA sequencing () quantifies transcript abundance to link regulatory features with expression outcomes, while variants like single-cell RNA-seq (scRNA-seq) resolve heterogeneity in regulatory responses across cell populations. RNA-seq, established in 2008, maps and measures mammalian transcriptomes with high sensitivity, enabling differential expression analysis and correlation with epigenetic marks from ChIP-seq or data. To capture spatial aspects of regulation, techniques such as map three-dimensional chromatin interactions, identifying topologically associating domains (TADs) and enhancer-promoter loops that constrain regulatory influences. The original method, developed in 2009, uses proximity to quantify pairwise chromatin contacts genome-wide, demonstrating how 3D folding modulates by bringing distant elements into proximity. Computational approaches process these genomic datasets to infer regulatory mechanisms and predict interactions. Sequence-based motif discovery tools, such as (Multiple Em for Motif Elicitation), scan non-coding regions for enriched DNA patterns indicative of binding sites, aiding in the annotation of potential regulatory elements. First described in 1994, employs expectation-maximization to fit mixture models, identifying s from unaligned sequences and remaining a cornerstone for cis-regulatory analysis. Genome-wide association of motifs with functional data from ChIP-seq further refines predictions of active binding events. For reconstructing gene regulatory networks (GRNs), algorithms infer causal relationships from expression profiles by modeling dependencies between genes. ARACNE (Algorithm for the Reconstruction of Accurate Cellular Networks), proposed in 2006, applies to estimate direct regulatory interactions from data, pruning indirect edges using the principle to scale to mammalian network complexity. Building on ensemble methods, GENIE3 (GEne Network Inference Engine), introduced in 2010, uses to rank potential regulators based on feature importance scores derived from expression predictors, outperforming competitors in the DREAM4 challenge for multifactorial network inference. These tree-based approaches handle nonlinear relationships and noisy data effectively, providing sparse, interpretable GRNs. Advances in machine learning have integrated multi-omics data for more predictive models of regulation. Deep learning architectures, such as convolutional neural networks in Basenji (2018) and transformers in Enformer (2021), forecast gene expression directly from DNA sequences by capturing local motifs and long-range dependencies up to 100 kb away. Enformer, in particular, achieves superior accuracy in held-out tissues and developmental stages by modeling chromatin context through dilated convolutions and attention mechanisms. As of 2025, further progress includes models like scGPT (2023), which leverages large language models for single-cell multi-omics integration to infer regulatory networks from heterogeneous data, enhancing predictions of cell-type-specific gene expression. Such models not only interpret variant effects on regulation but also simulate perturbations, advancing personalized genomics. Integration of genomic and computational tools, often via pipelines like those in or Epigenomics projects, reveals context-specific regulatory grammars, though challenges persist in handling single-cell resolution and causal validation. These approaches collectively bridge sequence to function, illuminating how and environmental cues orchestrate .

References

  1. [1]
    Gene expression and regulation - Autoimmunity - NCBI Bookshelf
    This chapter focuses on gene expression and its regulation including transcription and translation processes.
  2. [2]
    REGULATION OF GENE EXPRESSION IN THE GENOMIC CONTEXT
    In this review we summarize the basic features and functions of the crucial regulatory elements promoters, enhancers, and insulators.Mini Review Article · Genomic Context · Acknowledgments
  3. [3]
    Review The Human Transcription Factors - ScienceDirect.com
    Feb 8, 2018 · Transcription factors (TFs) directly interpret the genome, performing the first step in decoding the DNA sequence. Many function as “master ...
  4. [4]
    Capturing and Understanding the Dynamics and Heterogeneity of ...
    Nov 5, 2020 · In this review, we highlight the dynamic nature of transient gene expression changes to better understand cell physiology and development in general.
  5. [5]
    Gene Regulation and Cellular Metabolism: An Essential Partnership
    Like other biochemical processes, gene expression adjusts with the energy budget. If energy expenditure for other biochemical processes goes up, then ...Missing: unnecessary | Show results with:unnecessary
  6. [6]
    An Overview of Gene Control - Molecular Biology of the Cell - NCBI
    Six steps at which eucaryotic gene expression can be controlled. Controls that operate at steps 1 through 5 are discussed in this chapter. Step 6, the ...<|control11|><|separator|>
  7. [7]
    Combinatorial Control of Gene Expression - PMC - PubMed Central
    The principle of combinatorial control of gene expression that is addressed in this paper sheds light on the mechanisms used by the biological systems to ...
  8. [8]
    A Network of Multiple Regulatory Layers Shapes Gene Expression ...
    Apr 12, 2007 · Gene expression is controlled at multiple layers, and cells may integrate different regulatory steps for coherent production of proper protein ...
  9. [9]
    16.2: Regulation of Gene Expression - Prokaryotic versus Eukaryotic ...
    Nov 22, 2024 · Prokaryotes regulate gene expression by controlling the amount of transcription, whereas eukaryotic control is much more complex.
  10. [10]
    22.2: Gene Expression- Prokaryotes vs. Eukaryotes
    ### Key Differences in Gene Regulation: Prokaryotes vs. Eukaryotes
  11. [11]
    Regulation of Transcription and Gene Expression in Eukaryotes
    Gene expression is controlled on two levels. First, transcription is controlled by limiting the amount of mRNA that is produced from a particular gene.
  12. [12]
    How Genes Are Regulated – Introductory Biology
    Prokaryotic versus Eukaryotic Gene Expression ; RNA transcription and protein translation occur almost simultaneously. RNA transcription occurs prior to protein ...
  13. [13]
    Fundamentally Different Logic of Gene Regulation in Eukaryotes ...
    ... gene regulation in prokaryotes and eukaryotes is fundamentally different. This difference in logic reflects important differences in transcriptional regulatory ...
  14. [14]
    Mechanisms of Evolutionary Innovation Point to Genetic Control ...
    We tentatively identify the difference as a difference in control logic, that prokaryotic genes are by default 'on' and eukaryotic genes are by default 'off.' ...
  15. [15]
    Operons - PMC - NIH
    Operons (clusters of co-regulated genes with related functions) are a well-known feature of prokaryotic genomes.
  16. [16]
    Bacterial Sigma Factors and Anti-Sigma Factors: Structure, Function ...
    Jun 26, 2015 · Sigma factors are multi-domain subunits of bacterial RNA polymerase (RNAP) that play critical roles in transcription initiation.
  17. [17]
    Regulation of Bacterial Gene Expression by Transcription Attenuation
    Jul 3, 2019 · Pioneering studies by Charles Yanofsky and colleagues in the 1970s led to the discovery of transcription attenuation control of the trpEDCBA (or ...Tryptophan Operon Of E. Coli · Pyrg Operon Of B. Subtilis · Ribosome-Mediated...
  18. [18]
    [PDF] The input functions of genes: Michaelis-Menten and Hill equations
    So far we discussed how the repressor binds the promoter and inhibits transcription. To turn the gene system ON, a signal must cause X to unbind from the DNA.
  19. [19]
    A method for estimating Hill function-based dynamic models of gene ...
    Feb 21, 2018 · 2.1 Hill function-based ordinary differential equation model of gene regulatory networks. ODEs belong to the category of continuous ...Missing: prokaryotes | Show results with:prokaryotes
  20. [20]
    Yeast Gal4: a transcriptional paradigm revisited - PMC - NIH
    Gal4 is a yeast protein used as a model for studying transcriptional activation, controlling galactose-mediated gene expression and recruiting transcriptional ...
  21. [21]
    Analysis of Gal4-directed transcription activation using Tra1 ... - PNAS
    Activators are modular proteins that contain a DNA-binding domain (DBD) and an activation domain (AD). Activator-mediated stimulation of PIC assembly is ...
  22. [22]
    The mediator coactivator complex: functional and physical roles in ...
    Sep 15, 2003 · The three activators (GCN4, VP16 and GAL4) are shown binding to their DNA sites and recruiting yeast mediator to the promoter via a physical ...
  23. [23]
    Building regulatory landscapes reveals that an enhancer can recruit ...
    Jun 16, 2022 · The enhancer recruits cohesin to stimulate domain formation and engage flanking CTCF sites in loop formation. It requires cohesin exclusively ...
  24. [24]
    Multiple CTCF sites cooperate with each other to maintain a TAD for ...
    Jul 10, 2021 · Our results show that multiple CTCF sites surrounding the β-globin locus cooperate with each other to maintain a TAD.Missing: sources | Show results with:sources
  25. [25]
    The MAPK/ERK Cascade Targets Both Elk-1 and cAMP Response ...
    Jun 15, 2000 · In conclusion, we suggest that Elk-1 forms an important link in the MAP kinase pathway to transduce signals from the cell surface to the nucleus ...
  26. [26]
    Elk-1 a Transcription Factor with Multiple Facets in the Brain - Frontiers
    Elk-1 is a transcription factor that regulates immediate early gene (IEG) expression via the serum response element (SRE) DNA consensus site.
  27. [27]
    Coactivator condensation at super-enhancers links phase ... - Science
    These results support the idea that coactivators form phase-separated condensates at SEs that compartmentalize and concentrate the transcription apparatus, ...
  28. [28]
    Variance-corrected Michaelis-Menten equation predicts transient ...
    Dec 4, 2015 · The operator bound TF changes the DNA conformation or interacts with RNA polymerase (RNAP) directly, which changes the transcription rate of the ...
  29. [29]
    Nutrient dose-responsive transcriptome changes driven by ... - PNAS
    May 15, 2020 · We found that rate changes of genome-wide transcript levels in response to N-dose could be explained by a simple kinetic principle: the Michaelis–Menten (MM) ...Missing: initiation | Show results with:initiation
  30. [30]
    Emerging Roles of RNA 3′-end Cleavage and Polyadenylation in ...
    A crucial feature of gene expression involves RNA processing to produce 3′ ends through a process termed 3′ end cleavage and polyadenylation (CPA).
  31. [31]
    Discovery of m7G-cap in eukaryotic mRNAs - PMC - NIH
    Caps increase mRNA stability by protecting against 5′→3′ exonucleolytic degradation. Splicing accuracy and efficiency both increase by the presence of 5′- ...Missing: seminal | Show results with:seminal
  32. [32]
    mRNA capping: biological functions and applications
    Abstract. The 5′ m7G cap is an evolutionarily conserved modification of eukaryotic mRNA. Decades of research have established that the m7G cap serves as a.Missing: seminal | Show results with:seminal
  33. [33]
    Alternative polyadenylation: methods, mechanism, function, and role ...
    Feb 1, 2021 · The core pre-mRNA 3'end processing complex contains four subcomplexes, namely cleavage and polyadenylation factor (CPSF), cleavage stimulation ...
  34. [34]
    3′-End Processing of Eukaryotic mRNA: Machinery, Regulation ...
    Jun 20, 2023 · RBBP6 isoforms regulate the human polyadenylation machinery and modulate expression of mRNAs with AU-rich 3′ UTRs. Genes Dev. 28:2248–60.
  35. [35]
    Ubiquitin-dependent mechanism regulates rapid turnover of AU-rich ...
    An AU rich element (ARE) in the 3′ noncoding region promotes the rapid degradation of mammalian cytokine and proto-oncogene mRNAs, such as tumor necrosis ...Missing: seminal | Show results with:seminal
  36. [36]
    Global analysis of positive and negative pre-mRNA splicing ... - NIH
    Traditionally, SR proteins and hnRNP proteins have been viewed as antagonistic partners, regulating in opposite directions, many of the same alternative ...
  37. [37]
    The search for alternative splicing regulators: new approaches offer ...
    These come in two groups: SR proteins, which, when bound to exons, tend to promote exon inclusion, and hnRNP proteins, which usually have the opposite effect ( ...
  38. [38]
    Distinct regulatory programs establish widespread sex-specific ...
    In Drosophila melanogaster, female-specific expression of Sex-lethal (SXL) and Transformer (TRA) proteins controls sex-specific alternative splicing and/or ...
  39. [39]
    Functions of the Nonsense-Mediated mRNA Decay Pathway in ...
    Nonsense-mediated mRNA decay (NMD) is a cellular surveillance mechanism that degrades transcripts containing premature translation termination codons, ...
  40. [40]
    Nonsense-Mediated mRNA Decay: Mechanisms and Recent ...
    Aug 19, 2025 · Key players in this pathway include up-frameshift proteins (UPFs), nonsense-mediated mRNA decay associated with p13K-related kinases (SMGs) ...
  41. [41]
    The function and regulatory mechanism of RNA-binding proteins in ...
    HuR binds to the 3'UTR of many proto-oncogenes and unstable AREs to regulate the stability and enhance the translation of target mRNAs, and it is also a key ...<|separator|>
  42. [42]
    HuR regulates cyclin A and cyclin B1 mRNA stability during cell ...
    Our results indicate that HuR may play a critical role in cell proliferation, at least in part by mediating cell cycle‐dependent stabilization of mRNAs ...
  43. [43]
    Precision and functional specificity in mRNA decay - PNAS
    A nonlinear least squares model was fit to determine the decay rate constant (k) and half-life (t1/2) of each mRNA. The decay rate constant, k, is the value ...Missing: kinetics | Show results with:kinetics
  44. [44]
    Molecular mechanisms of translational control - Nature
    Oct 1, 2004 · Translational control is widely used to regulate gene expression. This mode of regulation is especially relevant in situations where transcription is silent.
  45. [45]
    Perk Is Essential for Translational Regulation and Cell Survival ...
    PERK abolishes the phosphorylation of eIF2α in response to accumulation of malfolded proteins in the ER resulting in abnormally elevated protein synthesis and ...
  46. [46]
    Integrated stress response of vertebrates is regulated by four eIF2α ...
    Sep 16, 2016 · There are several phosphorylation sites in eIF2α, but serine 51 is critical for translational control. eIF2α phosphorylation causes transient ...
  47. [47]
    Hypoxia-inducible Factor-1α mRNA Contains an Internal Ribosome ...
    These data indicate that the presence of an IRES in the HIF-1α 5′UTR allows translation to be maintained under conditions that are inhibitory to cap-dependent ...
  48. [48]
    Control of translation and mRNA degradation by miRNAs and siRNAs
    Fifth, the interaction of miRNA and Argonaute with the mRNA may be influenced by other sequence-specific RNA-binding proteins, thus providing an additional ...
  49. [49]
    Regulation by let-7 and lin-4 miRNAs Results in Target mRNA ...
    Here, we report that in C. elegans, regulation by the let-7 miRNA results in degradation of its lin-41 target mRNA, despite the fact that its 3′UTR regulatory ...
  50. [50]
    Ubiquitin signaling in cell cycle control and tumorigenesis - Nature
    Oct 31, 2020 · Ubiquitin-mediated proteolysis of CDK regulators, via E3 ligases, ensures cell cycle progression. Dysregulation leads to uncontrolled cell ...
  51. [51]
    Regulation of the cell cycle by SCF-type ubiquitin ligases
    SCF ligases regulate cell cycle via protein degradation, targeting G1 cyclins and CKIs, and play roles in most cell cycle phases.
  52. [52]
    Translation drives mRNA quality control - PMC - PubMed Central
    No-go decay (NGD) and non-stop decay (NSD) both involve the recognition of stalled ribosome complexes. These stalls can arise through multiple mechanisms. (a) ...Ribosome Recognition By Key... · Figure 3. Mrna Surveillance... · Mrna Decay
  53. [53]
    Ribosome collision is critical for quality control during no-go decay
    Under normal circumstances, cycloheximide stalls every translating ribosome and as a result collisions are avoided.
  54. [54]
    Translational regulation by uORFs and start codon selection ...
    Jul 11, 2023 · Insertion mutagenesis to increase secondary structure within the 5′ noncoding region of a eukaryotic mRNA reduces translational efficiency.
  55. [55]
    Secondary structures that regulate mRNA translation provide ...
    uORFs are known to regulate the expression of proteins that play important roles in diverse biological processes such as cell differentiation or catabolic ...
  56. [56]
    DNA Methylation in Mammals - PMC - NIH
    Molecular and genetic studies in mammals have shown that DNA cytosine methylation (abbreviated to 5mC, for 5-methyl cytosine) is associated with gene silencing.Missing: seminal | Show results with:seminal
  57. [57]
    Conversion of 5-Methylcytosine to 5-Hydroxymethylcytosine in ...
    2009年4月16日 · Thus, TET proteins have potential roles in epigenetic regulation through modification of 5mC to hmC. 5-methylcytosine (5mC) is a minor base in ...
  58. [58]
    5-Hydroxymethylcytosine in the mammalian zygote is linked ... - Nature
    Mar 15, 2011 · The Tet knockdown effects in the zygote directly link the rapid active loss/demethylation of 5mC to its conversion into 5hmC. Third, the ...
  59. [59]
    DNA hypomethylation in cancer cells - PMC - NIH
    Gene region hypomethylation could contribute to oncogenesis by affecting classical oncogenes, but evidence suggests a greater involvement in the activation of ...Missing: seminal | Show results with:seminal
  60. [60]
    Regulation of chromatin by histone modifications | Cell Research
    Feb 15, 2011 · Our review has a transcriptional focus, simply reflecting the fact that most studies involving histone modifications have also had this focus.
  61. [61]
    Histone acetylation and transcriptional regulatory mechanisms
    This review will focus on molecular mechanisms by which histone acetylation affects transcriptional activity in living cells.
  62. [62]
    Regulating histone acetyltransferases and deacetylases - EMBO Press
    Oct 1, 2003 · Histone acetylation is catalysed by histone acetyl transferases (HATs), whereas the reverse reaction is performed by histone deacetylases (HDACs) ...Introduction · Regulating Enzyme Activity · PerspectivesMissing: seminal papers
  63. [63]
    Stimulation of GAL4 Derivative Binding to Nucleosomal DNA by the ...
    The SWI/SNF protein complex is required for the enhancement of transcription by many transcriptional activators in yeast. Here it is shown that the purified ...
  64. [64]
    The histone variant H2A.Z in gene regulation
    Jun 14, 2019 · The histone variant H2A.Z is involved in several processes such as transcriptional control, DNA repair, regulation of centromeric heterochromatin and, not ...Missing: super- | Show results with:super-
  65. [65]
    The RNA polymerase II core promoter - Genes & Development
    The core promoter is the ultimate target of action of all of the factors that are involved in the regulation of transcription by RNA polymerase II.The Tata Box · The Tfiib Recognition... · Other Core Promoter...
  66. [66]
    Core Promoters in Transcription: Old Problem, New Insights - NIH
    The two most common core promoter elements associated with protein-coding genes are the TATAA box and the Initiator (Inr), which occur either together or ...The Core Promoter: A... · The Transcriptional... · Core Promoters Of Non-Coding...
  67. [67]
    Housekeeping and tissue-specific cis-regulatory elements - NIH
    Cell type-specific and housekeeping enhancers and promoters collectively control the transcriptional output of mammalian cells.
  68. [68]
    Housekeeping genes tend to show reduced upstream sequence ...
    Jul 13, 2007 · We describe that genes with housekeeping expression contain more divergent promoters than genes with a more restricted tissue expression.Cpg Island Content And... · Discussion · Sequence Retrieval And...
  69. [69]
    The unexpected traits associated with core promoter elements
    Having a TATA-box in the core promoter seems beneficial for expression of short genes, while its advantage diminishes with longer genes. We have therefore ...Core Promoter Elements · Proximal Promoter Elements... · The Core Promoter And The...
  70. [70]
    Mechanisms of enhancer action: the known and the unknown
    Apr 15, 2021 · Enhancers are non-coding sequences in the genome that activate the expression of target genes transcribed by the RNA polymerase II (RNAPII).
  71. [71]
    Immunoglobulin Enhancers & B Cell Organization
    Jun 24, 2022 · In this review, we examined multiple data showing the critical interest of studying Ig gene regulation at the whole nucleus scale.
  72. [72]
    An Igh distal enhancer modulates antigen receptor diversity ... - Nature
    Mar 3, 2023 · Each mature B cell has a unique Ig receptor, created via V(D)J recombination through an ordered set of rearrangements. One of the ~100 ...
  73. [73]
    Enhancers and silencers: an integrated and simple model for their ...
    Regulatory DNA elements such as enhancers, silencers and insulators are embedded in metazoan genomes, and they control gene expression during development.
  74. [74]
    Insulators: many functions, many mechanisms - Genes & Development
    ... insulators must interfere with a signal between the enhancer and the promoter. That signal could be a direct contact between the enhancer and the promoter ...
  75. [75]
    CTCF is conserved from Drosophila to humans and confers ...
    Feb 1, 2005 · Expression of the eukaryotic genome is controlled by enhancer and silencer elements, both of which can mediate their function from a distance.<|separator|>
  76. [76]
    Topologically Associating Domains and Regulatory Landscapes in ...
    Jul 6, 2021 · In this review, we discuss the connections of the 3D genome with gene expression, the relationship between TADs and RLs, and their dynamics in ...
  77. [77]
    Disruptions of Topological Chromatin Domains Cause Pathogenic ...
    May 7, 2015 · Disruptions of TADs cause ectopic promoter interactions, altered gene expression, and misexpression, leading to pathogenic phenotypes and ...<|separator|>
  78. [78]
    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 ...
  79. [79]
    The Encyclopedia of DNA Elements (ENCODE)
    Sep 17, 2023 · The ENCODE Project started in 2003 with the ENCODE Pilot Project, which focused on 1% of the human genome and subsequently completed two ...
  80. [80]
    Position Weight Matrix, Gibbs Sampler, and the Associated ... - NIH
    Position weight matrix (PWM) is a key bioinformatic tool used to characterize and predict motifs in nucleotide or amino acid sequences.
  81. [81]
    Feedback regulation of Lac repressor expression in Escherichia coli
    Negative feedback regulation, mediated through repressor binding site O3, which overlaps the lacI gene, could explain the robustness of the weak expression ...
  82. [82]
    Hox genes in development and beyond
    Jan 16, 2023 · Hox genes encode evolutionarily conserved transcription factors that are essential for the proper development of bilaterian organisms.
  83. [83]
    Chromatin organization and global regulation of Hox gene clusters
    The epigenetic regulation of Hox gene clusters seems to rely, mostly, on the activities of protein complexes encoded by Polycomb (PcG) and Trithorax (TrxG) ...
  84. [84]
    Hox genes regulation in vertebrates - Soshnikova - 2014
    Jul 5, 2013 · Collinear patterns of Hox genes expression are controlled by multiple proximal regulatory elements scattered throughout the clusters (Spitz et ...
  85. [85]
    Two Tier Hox Collinearity Mediates Vertebrate Axial Patterning
    Sep 3, 2018 · A two tier mechanism mediates Hox collinearity. Besides the familiar collinear chromatin modification within each Hox cluster ...
  86. [86]
    Engrailed and Hedgehog Make the Range of Wingless Asymmetric ...
    We found that Wingless acts at a different range in the anterior and posterior directions. We show that this asymmetry follows in part from differential ...
  87. [87]
    hedgehog and engrailed: pattern formation and polarity in the ...
    Jun 1, 1999 · First, we show that engrailed acts in the A compartment: Hedgehog leaves the P cells and crosses the A/P boundary where it induces engrailed in ...
  88. [88]
    The Maternal to Zygotic Transition in Mammals - PMC - NIH
    This transition occurs at species-specific times after one or several rounds of blastomere cleavage and is essential for normal development.
  89. [89]
    Epigenetic reprogramming during the maternal‐to‐zygotic transition
    The epigenetic reprogramming plays crucial roles in regulating the process of MZT and supervising the normal development of early development of embryos.
  90. [90]
    The maternal-to-zygotic transition revisited | Development
    Jun 12, 2019 · The MZT describes a coordinated series of molecular events, starting with the degradation of maternally deposited transcripts and ending with global activation ...
  91. [91]
    Seeing Is Believing: The Bicoid Morphogen Gradient Matures
    Jan 23, 2004 · These experiments suggested a very simple model in which the thresholds for the response to Bicoid are determined by the affinity of the Bicoid ...
  92. [92]
    ERIC DAVIDSON: STEPS TO A GENE REGULATORY NETWORK ...
    The next decade, Eric's lab spent a major effort clarifying, refining, correcting, and recasting details of the endomesoderm network and extending it to other ...
  93. [93]
    The endoderm gene regulatory network in sea urchin embryos up to ...
    Apr 15, 2010 · We have analyzed the GRN driving endoderm specification up to the onset of gastrulation and present in this paper the mechanisms which determine this process ...The Endoderm Gene Regulatory... · Results · Appendix A
  94. [94]
    The prognostic value of CDKN2A hypermethylation in colorectal ...
    May 23, 2013 · CDKN2A promoter methylation is a frequent epigenetic event and an important mechanism leading to silencing and dysfunction of CDKN2A gene, which ...
  95. [95]
    ΔFosB: A sustained molecular switch for addiction - PNAS
    Increasing evidence suggests that the transcription factor ΔFosB represents one mechanism by which drugs of abuse produce relatively stable changes in the brain ...Missing: stabilization | Show results with:stabilization
  96. [96]
    Differential epigenetic modifications in the FMR1 gene of the fragile ...
    Mar 2, 2005 · The fragile X syndrome is caused by a >200 CGG repeat expansion within the FMR1 gene promoter, with consequent DNA hypermethylation and ...
  97. [97]
    ALS-linked TDP-43 mutations produce aberrant RNA splicing and ...
    Mutations in the RNA binding protein TDP-43 cause amyotrophic lateral sclerosis and frontotemporal dementia.
  98. [98]
  99. [99]
    Epigenetic editing for autosomal dominant neurological disorders
    Mar 6, 2024 · Epigenetic modification can be used to target and treat disorders through the modification of endogenous gene expression.
  100. [100]
    Firefly luciferase gene: structure and expression in mammalian cells
    The nucleotide sequence of the luciferase gene from the firefly Photinus pyralis was determined from the analysis of cDNA and genomic clones.
  101. [101]
    Potent and specific genetic interference by double-stranded RNA in ...
    Feb 19, 1998 · We found that double-stranded RNA was substantially more effective at producing interference than was either strand individually.Missing: discovery | Show results with:discovery
  102. [102]
    A Programmable Dual-RNA–Guided DNA Endonuclease ... - Science
    Jun 28, 2012 · Our study reveals a family of endonucleases that use dual-RNAs for site-specific DNA cleavage and highlights the potential to exploit the system for RNA- ...
  103. [103]
    Kinetic PCR Analysis: Real-time Monitoring of DNA Amplification ...
    Sep 1, 1993 · We describe a simple, quantitative assay for any amplifiable DNA sequence that uses a video camera to monitor multiple polymerase chain reactions (PCRs) ...Author Information · About This Article · Cite This Article
  104. [104]
    ARACNE: An Algorithm for the Reconstruction of Gene Regulatory ...
    Mar 20, 2006 · ARACNE, a novel algorithm, using microarray expression profiles, specifically designed to scale up to the complexity of regulatory networks in mammalian cells.
  105. [105]
    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.
  106. [106]
    Effective gene expression prediction from sequence by integrating ...
    Oct 4, 2021 · We report substantially improved gene expression prediction accuracy from DNA sequences through the use of a deep learning architecture, called Enformer.