Fact-checked by Grok 2 weeks ago

Transcription factor

A transcription factor (TF) is a protein that regulates the transcription of genetic information from DNA to messenger RNA by binding to specific DNA sequences adjacent to the genes they control, thereby modulating gene expression in response to cellular signals. These proteins are essential for coordinating the precise activation or repression of genes, enabling cells to adapt to developmental cues, environmental changes, and physiological needs across both prokaryotic and eukaryotic organisms. In structure, transcription factors typically consist of at least two functional domains: a DNA-binding domain (DBD) that recognizes and attaches to short DNA motifs, such as 6–10 base pair sequences in promoter or enhancer regions, and an effector domain (which may include activation or repression subdomains) that interacts with RNA polymerase, co-regulatory proteins, or chromatin to influence transcription initiation. Common DBD motifs include helix-turn-helix, zinc fingers, and basic helix-loop-helix structures, which provide sequence-specific affinity with up to a million-fold preference for target sites over non-specific DNA. These domains allow TFs to respond to signals like metabolites, hormones, or stress, often through allosteric changes that alter their binding or activity. Mechanistically, transcription factors exert control by either activating or repressing transcription: activators bind to promoter-proximal elements (e.g., CAAT or GC boxes) or distant enhancers to recruit RNA polymerase II and general transcription machinery, facilitating the assembly of a pre-initiation complex, while repressors bind to silencer sequences to block this assembly, deform DNA, or inhibit co-activators. In prokaryotes, regulation is often simpler, with a single TF modulating operons like the lac operon via the catabolite activator protein (CAP); in eukaryotes, it involves combinatorial interactions among multiple TFs, chromatin remodeling, and long-range DNA looping to achieve tissue-specific expression. For instance, the beta-globin gene is activated only in erythroblasts through specific TF binding that overrides default repressive chromatin states. The significance of transcription factors lies in their role as master regulators of cellular identity and function, with the human genome encoding approximately 1,600 TFs (roughly 8% of protein-coding genes), organized into families like homeobox or nuclear receptor groups that share conserved motifs. Dysregulation of TFs contributes to diseases such as cancer, developmental disorders, and metabolic imbalances, underscoring their therapeutic potential in biotechnology, including engineered TFs for gene therapy and synthetic biology applications.

Definition and Overview

Definition

Transcription factors (TFs) are proteins that regulate the rate of transcription from DNA to messenger RNA (mRNA) by binding to specific DNA sequences in proximity to genes, thereby activating or repressing gene expression. These molecules play a pivotal role in controlling which genes are expressed in a given cell type or under specific conditions, influencing cellular identity and response to environmental cues. Although primarily proteins, some non-coding RNAs function analogously in transcriptional regulation, such as by recruiting protein factors or modulating chromatin. TFs are broadly classified into general transcription factors (GTFs) and specific transcription factors. GTFs, such as those in the TFII family (e.g., TFIIA, TFIIB), are essential for assembling the basal transcription machinery at promoter regions and initiating transcription by RNA polymerase II in eukaryotes, independent of the specific gene involved. In contrast, specific transcription factors bind to regulatory DNA elements like enhancers or silencers to modulate the transcription of particular genes, often in response to developmental signals or stressors. The concept of transcription factors emerged from early studies on gene regulation, with the lac repressor in prokaryotes—identified by Jacob and Monod in 1961—serving as a foundational analog by demonstrating how a protein could bind DNA to repress transcription of the lac operon. In eukaryotes, TFs were first characterized in the 1960s and 1970s through investigations into multi-subunit RNA polymerases and their associated factors, marking the shift from prokaryotic models to understanding complex eukaryotic regulation. Unlike DNA regulatory elements such as enhancers or silencers, which are static sequences in the genome, TFs are the dynamic binding proteins that recognize and interact with these sites to exert control. Additionally, while RNA polymerase directly catalyzes RNA synthesis, TFs do not perform this enzymatic function but instead recruit or modify the polymerase and associated machinery to fine-tune transcriptional output.

Biological Importance

Transcription factors (TFs) play a central role in gene regulation by integrating diverse cellular signals to orchestrate precise control over gene expression, thereby determining cell fate, driving differentiation, and enabling responses to environmental stimuli. In eukaryotes, TFs achieve this by binding to specific DNA sequences and modulating the transcriptional machinery, ensuring that only appropriate genes are expressed at the right time and level during processes such as development and homeostasis. This regulatory function is indispensable, as TFs collectively influence the expression of a substantial portion of the genome, with individual TFs often controlling hundreds of target genes. As of 2018, the human genome encodes approximately 1,639 TFs, which represent about 8% of all protein-coding genes; more recent estimates (as of 2023) suggest around 1,485–1,600, or 7–8%, depending on classification criteria. In contrast, prokaryotes like Escherichia coli possess a more modest repertoire of around 300 TFs, reflecting their simpler cellular organization and regulatory needs. These TFs in humans not only maintain tissue-specific gene programs but also facilitate multicellular coordination, underscoring their broad influence on organismal complexity. The regulatory logic of TFs differs markedly between prokaryotes and eukaryotes: bacterial systems rely on relatively straightforward, often sigma factor-mediated activation or repression of operons, whereas eukaryotic TFs employ combinatorial control, where multiple factors cooperate to achieve specificity and fine-tuned expression amid chromatin barriers. This complexity in eukaryotes allows for layered regulation essential to multicellular life. Without TFs, gene expression would default to a constitutive, unregulated state driven solely by basal transcriptional machinery, resulting in widespread cellular dysfunction, loss of specificity, and inability to adapt to changing conditions. Thus, TFs are prerequisites for dynamic and context-appropriate gene control across all domains of life.

Molecular Structure

DNA-Binding Domains

DNA-binding domains (DBDs) are specialized protein motifs within transcription factors that enable sequence-specific recognition and binding to DNA, typically through interactions with the major groove of the double helix. These domains vary in structure but share the common function of conferring binding affinity and specificity to particular nucleotide sequences, allowing transcription factors to target regulatory elements such as promoters and enhancers. The diversity of DBDs reflects the evolutionary adaptation of gene regulation across organisms. Common DBDs include the helix-turn-helix (HTH), zinc finger (C2H2 type), leucine zipper (bZIP), helix-loop-helix (HLH), winged helix, and homeodomain motifs. The HTH motif consists of two alpha helices connected by a short turn, with the second "recognition" helix inserting into the DNA major groove to make direct contacts with bases. Zinc fingers of the C2H2 type feature a compact beta-beta-alpha fold stabilized by coordination of a Zn²⁺ ion via two cysteines and two histidines, allowing the alpha helix to probe the major groove for sequence-specific interactions. The bZIP domain combines a leucine-rich zipper for dimerization with a basic region that forms an alpha helix binding the DNA major groove adjacently. HLH motifs involve two amphipathic alpha helices separated by a loop, promoting dimerization and positioning basic residues to contact DNA bases. Winged helix domains, often variants of HTH, include beta-sheet "wings" that stabilize binding via backbone interactions, as seen in forkhead factors. Homeodomains, a specialized HTH subclass, comprise a 60-amino-acid helix-turn-helix structure with three alpha helices, where the third helix recognizes DNA via hydrogen bonds to specific bases. Specificity in DNA binding is primarily determined by base-specific hydrogen bonds and van der Waals interactions between amino acid side chains in the DBD and nucleotide bases in the major groove. These contacts are often complemented by interactions with the DNA phosphate backbone and minor groove, while binding affinity is modulated by the surrounding sequence context, including DNA shape features like groove width and propeller twist that influence fit. DBDs arose early in eukaryotic evolution, with many motifs tracing origins to prokaryotic ancestors; for instance, the HTH motif is conserved across bacteria, archaea, and eukaryotes, appearing in prokaryotic repressors and eukaryotic developmental regulators. In contrast, C2H2 zinc fingers are largely eukaryotic innovations, expanding in metazoans through gene duplication and diversification to enable complex regulatory networks. Homeodomains similarly predate the metazoan radiation, with duplications occurring before the divergence of animals, fungi, and plants.

Transactivation and Other Domains

Transcription factors often possess transactivation domains (TADs) that recruit coactivators to stimulate gene expression. These domains are typically short, modular regions enriched in specific amino acid residues, including acidic (rich in aspartic and glutamic acids), glutamine-rich, and proline-rich motifs. Acidic TADs, the most common and potent class, feature hydrophobic residues like aromatic amino acids (tryptophan, phenylalanine, tyrosine) and leucines that bind to hydrophobic grooves on coactivators such as the Mediator complex and histone acetyltransferases (HATs) like CREBBP/EP300. Glutamine-rich TADs, though less frequent, contribute to activation by interacting with similar coactivators, often overlapping with other motif types. Proline-rich TADs, characterized by high proline content (>15%), can be inducible and modulate transcriptional bursting by engaging Mediator to influence pause release or HATs to alter burst duration. These interactions promote RNA polymerase II recruitment and chromatin remodeling through histone acetylation, enhancing transcription initiation. In contrast, repression domains (RDs) within transcription factors mediate transcriptional silencing by recruiting corepressors that compact chromatin or remove activating marks. RDs are often intrinsically disordered regions that, like TADs, contribute to effector domains with median lengths around 91 amino acids, and exhibit lower acidity compared to TADs but share hydrophobic features. They contain conserved motifs such as PxDLS (recruits CtBP corepressor), AAxxL (recruits Sin3A), and PLKKR/HKKF (recruits Smrter complex), which facilitate binding to corepressors like SIN3A and histone deacetylases (HDACs) such as HDAC1 and HDAC3. These interactions lead to histone deacetylation and chromatin condensation, thereby inhibiting transcription. Dimerization and other protein-protein interaction domains enable transcription factors to form homo- or heterodimers, which are crucial for cooperative DNA binding and modulating transcriptional output. The leucine zipper motif, a coiled-coil structure of 4-5 heptads with leucines at every seventh position, mediates parallel dimerization in bZIP family factors like Fos, Jun, and GCN4, dictating specificity through electrostatic interactions at interhelical interfaces. This specificity controls which dimers form and their affinity for target sites, thereby regulating gene expression. Similarly, SH2 and SH3 domains in factors like STAT proteins facilitate dimerization via phosphotyrosine recognition, promoting rapid activation in response to signals.90357-3) Many of these domains, particularly TADs and RDs, comprise intrinsically disordered regions (IDRs) that lack stable secondary structure, conferring flexibility for promiscuous interactions with multiple partners. IDRs in over 80% of eukaryotic transcription factors enable dynamic, low-affinity binding modes, such as fuzzy interactions with Mediator or CBP/p300, which support signal integration and high-turnover complexes essential for precise gene regulation. This disorder allows TADs to adopt transient conformations, like helices, upon binding, enhancing adaptability without rigid specificity.

Mechanism of Action

DNA Binding and Recognition

Transcription factors (TFs) locate their target DNA sites through a process known as facilitated diffusion, which combines three-dimensional (3D) diffusion in the nucleoplasm with one-dimensional (1D) sliding along the DNA backbone. This mechanism allows TFs to efficiently search vast genomic landscapes, alternating between bulk solution diffusion to approach DNA and surface diffusion to scan local sequences. Electrostatic interactions between the positively charged DNA-binding domains of TFs and the negatively charged DNA phosphate backbone facilitate initial non-specific associations, enabling rapid translocation without complete dissociation. TFs exhibit distinct binding affinities for specific versus non-specific DNA sequences, with dissociation constants (Kd) typically ranging from 10^{-9} M for high-affinity, sequence-specific sites to 10^{-6} M for non-specific interactions. Specific binding involves precise recognition of nucleotide sequences, often mediated by hydrogen bonds and van der Waals contacts within the major or minor grooves, while non-specific binding relies primarily on electrostatic and hydrophobic forces. This affinity gradient ensures TFs spend sufficient time at cognate sites to initiate regulation while minimizing off-target effects. Target sites, or response elements, are short consensus DNA sequences located in promoters or enhancers that dictate TF specificity. For instance, the TATA box (consensus: TATAAA) serves as a binding site for the general transcription factor TATA-binding protein (TBP), positioning it near transcription start sites in many eukaryotic promoters. In contrast, specific TFs recognize motifs like the cAMP response element (CRE; consensus: TGACGTCA), which is bound by CREB to mediate cAMP-dependent gene activation in response to signaling pathways. Cooperative binding enhances the specificity and stability of TF-DNA interactions when multiple TFs occupy adjacent sites on the same DNA segment. This phenomenon arises from protein-protein interactions between neighboring TFs, which can increase overall binding affinity by 10- to 100-fold compared to independent binding, particularly at enhancer regions with clustered motifs spaced 50 base pairs apart. Such cooperativity is crucial for combinatorial control, allowing cells to integrate multiple signals for precise gene regulation. In the context of chromatin, TFs initially bind to nucleosome-free or accessible regions, such as open promoters or enhancers, where DNA is less occluded by histone octamers. Pioneer TFs, like FOXA, possess unique properties that enable them to engage compacted chromatin directly, displacing linker histones and maintaining nucleosome accessibility for subsequent TF recruitment. This initial chromatin opening is essential for establishing regulatory competence in developmental and environmental contexts.

Interaction with Transcriptional Machinery

Transcription factors (TFs) primarily exert their regulatory effects by interacting with components of the transcriptional machinery after binding to specific DNA sequences. These interactions facilitate the recruitment and assembly of the pre-initiation complex (PIC), which includes RNA polymerase II (Pol II) and general transcription factors (GTFs). A key mechanism involves TFs contacting the Mediator complex through their transactivation domains (TADs), which form dynamic, fuzzy interfaces with Mediator subunits such as MED1 or MED23, thereby bridging enhancers or promoters to the core machinery. Additionally, TFs can directly engage TFIID via interactions with TATA-binding protein (TBP), promoting stable PIC formation at core promoters, as demonstrated by cooperative assembly assays showing enhanced transcription when TFIID and Mediator are both present. TFs also interact with the C-terminal domain (CTD) of Pol II, often indirectly through coactivators like CRSP (a Mediator-related complex), which binds the unphosphorylated CTD to stabilize the PIC and facilitate promoter clearance upon phosphorylation by TFIIH-associated CDK7. These recruitment steps culminate in activation loops, where TAD-coactivator bridges, such as those involving Mediator, enable enhancer-promoter looping mediated by cohesin and CTCF, allowing distal TFs to influence proximal PIC assembly. In basal transcription, GTFs including TFIIA through TFIIH assemble sequentially with Pol II at core promoters to form the PIC without specific TFs, supporting minimal, unregulated initiation. Regulated transcription, however, relies on specific TFs to enhance this process; for instance, activator TFs recruit Mediator to enhancers, which then loops to the promoter to boost PIC stability and Pol II recruitment, resulting in higher transcriptional output compared to basal levels. TFs can also mediate repression by interfering with PIC assembly or post-initiation steps. For example, certain transcriptional regulators like BRCA1, functioning as an E3 ubiquitin ligase, ubiquitinate Pol II and TFIIE, leading to their dissociation from the PIC and blocking stable complex formation during initiation. Other repressive TFs promote Pol II pausing by recruiting NELF and DSIF shortly after initiation or induce premature elongation termination through interactions that hinder CTD phosphorylation progression. Combinatorial control arises when multiple TFs integrate signals to produce graded transcriptional responses, allowing fine-tuned gene expression proportional to input stimuli. In this paradigm, noncooperative binding of 2–3 TFs (e.g., NF-κB and IRF3 in immune responses) to clustered sites forms logic gates like AND/OR configurations, where response amplitude scales with TF occupancy and affinity, enabling cells to discern signal strengths without binary on/off switches.

Functions in Cellular Processes

Developmental Roles

Transcription factors play pivotal roles in embryonic development by regulating the precise spatiotemporal expression of genes that drive cell differentiation, tissue patterning, and organ formation. Through their ability to bind specific DNA sequences and recruit transcriptional machinery, these proteins establish gene expression patterns that define cellular identities along developmental axes. In particular, they interpret positional information from morphogen gradients and coordinate sequential activation cascades to ensure proper body plan formation. Hox genes, encoding homeodomain-containing transcription factors, are essential for anterior-posterior body patterning in bilaterian animals. Expressed in collinear domains along the embryo's axis, Hox proteins specify segmental identities by activating or repressing downstream targets that control organ placement and morphology. For instance, in vertebrates and insects, Hox clusters direct the formation of structures such as limbs and vertebrae through combinatorial codes of expression. Basic helix-loop-helix (bHLH) transcription factors like MyoD exemplify their role in cell lineage specification during myogenesis. MyoD initiates skeletal muscle differentiation by binding to E-box motifs in promoters of muscle-specific genes, converting multipotent progenitors into committed myoblasts and promoting myotube fusion. This process highlights how individual transcription factors can act as master regulators to enforce tissue-specific programs. Paired domain transcription factors such as Pax6 are critical for sensory organ development, particularly the eye. In Drosophila, the Pax6 homolog Eyeless induces ectopic eye formation when misexpressed, activating a downstream network that includes genes for retinal cell specification and morphogenesis. Similarly, in vertebrates, Pax6 orchestrates lens placode induction and neural retina differentiation, underscoring its conserved function as a master control gene for visual system assembly. Transcription factors also interpret morphogen gradients to pattern appendages, as seen with the Gli family responding to Sonic hedgehog (Shh) signaling in vertebrate limbs. Gli proteins act as both activators and repressors in a concentration-dependent manner, translating the Shh gradient from the zone of polarizing activity into anterior-posterior digit identities. High Shh levels promote Gli activators for posterior fates, while low levels allow Gli repressors to specify anterior structures, thus decoding positional cues into discrete developmental outcomes. In Drosophila embryogenesis, temporal-spatial control is achieved through hierarchical cascades of transcription factors in segmentation. Gap genes, such as Krüppel and hunchback, are activated first in broad domains to subdivide the embryo into regions, subsequently regulating pair-rule genes like even-skipped and fushi tarazu, which establish periodic stripes corresponding to every other segment. This sequential activation refines the body plan, ensuring metameric organization. For stem cell maintenance, the pluripotency network in embryonic stem cells relies on core transcription factors Oct4 and Sox2, which cooperatively bind enhancers to sustain self-renewal and prevent differentiation. Oct4-Sox2 dimers regulate a circuit including Nanog and other targets, maintaining an undifferentiated state poised for lineage commitment upon signaling cues. This network exemplifies how transcription factors integrate to preserve developmental potential in progenitor cells.

Response to Signals and Environment

Transcription factors play a pivotal role in transducing extracellular signals into intracellular gene expression changes, enabling cells to adapt to environmental cues such as hormones, stress, and nutrients. In signal transduction pathways, NF-κB exemplifies this by mediating inflammatory responses; upon stimulation by cytokines or pathogen-associated molecular patterns, the IκB kinase complex phosphorylates IκBα, leading to its ubiquitination and proteasomal degradation, which liberates NF-κB dimers for nuclear translocation and activation of pro-inflammatory genes like TNF-α and IL-6. Similarly, p53 responds to DNA damage by accumulating through post-translational modifications, such as phosphorylation by ATM/ATR kinases, allowing it to bind DNA response elements and transactivate genes involved in cell cycle arrest (e.g., p21) or apoptosis (e.g., PUMA, BAX), thereby preventing propagation of genomic instability. In hypoxic conditions, HIF-1 activation occurs via stabilization of its α subunit under low oxygen, where it dimerizes with ARNT, undergoes conformational changes for enhanced DNA binding, and induces genes like VEGF and EPO to promote angiogenesis and metabolic adaptation. Environmental stresses trigger specific transcription factors to restore homeostasis. Heat shock factor 1 (HSF1) activates during thermal stress when Hsp70 chaperones dissociate from HSF1 due to competition with unfolded proteins, enabling HSF1 trimerization, nuclear translocation, and phosphorylation; this drives transcription of chaperone genes such as HSP70 and HSP40, bolstering protein refolding and cytoprotection. Steroid hormone receptors, such as the glucocorticoid receptor (GR), respond to ligands like cortisol by binding at the ligand-binding domain, which displaces inhibitory chaperones (e.g., FKBP51), exposes nuclear localization signals, and facilitates microtubule-dependent nuclear import; once nuclear, GR binds glucocorticoid response elements to regulate anti-inflammatory genes like annexin-1. Intercellular signaling often involves transcription factors that relay cues from neighboring cells or distant sources. In cytokine pathways, STAT family members (e.g., STAT1–6) are phosphorylated by JAK kinases upon ligand binding to receptors like those for interferons or interleukins, leading to dimerization, nuclear entry, and activation of immune-related genes; for instance, STAT1 promotes antiviral responses via IFN-γ, while STAT6 drives Th2 differentiation through IL-4. The Wnt pathway employs β-catenin as a transcriptional co-activator; Wnt ligands inhibit the destruction complex (AXIN/APC/GSK3β), stabilizing β-catenin for nuclear accumulation, where it interacts with TCF/LEF factors to transcribe targets like c-MYC and cyclin D1, influencing cell proliferation and tissue patterning in response to paracrine signals. Transcription factors frequently integrate multiple inputs through crosstalk, amplifying or fine-tuning responses. The AP-1 complex, composed of Fos and Jun dimers, exemplifies this in immune contexts by cooperating with NF-κB; inflammatory stimuli induce both via shared upstream kinases (e.g., MAPK and IKK), enabling synergistic binding at composite promoter elements to boost cytokine expression like IL-2 during T-cell activation. This integration ensures context-specific outputs, such as enhanced inflammation under combined stress and cytokine exposure.

Regulation of Activity

Synthesis and Post-Translational Modifications

Transcription factors (TFs) are synthesized through the transcription of dedicated genes into messenger RNA (mRNA) followed by translation into proteins. These genes are typically regulated at the transcriptional level by upstream TFs, which bind to promoter or enhancer regions to initiate or enhance their expression. Autoregulation is a common mechanism, where TFs positively or negatively control their own gene transcription, observed in approximately 56% of studied human TFs, based on analysis of a regulatory network. For instance, the NF-κB family member NF-κB2 exhibits positive autoregulation through κB elements in its promoter, allowing rapid amplification of its expression in response to stimuli. Additionally, mRNA stability plays a critical role in controlling TF levels; microRNAs (miRNAs) often bind to the 3' untranslated regions of TF mRNAs, promoting their degradation and thereby fine-tuning protein abundance. miRNA-mediated destabilization accounts for the majority of repressive effects on TF mRNAs, with half-lives varying widely depending on cellular context. Post-translational modifications (PTMs) further regulate TF activity, stability, and localization immediately after synthesis. Phosphorylation, mediated by kinases such as mitogen-activated protein kinases (MAPKs), activates or inactivates TFs by altering their conformation or interactions. A representative example is the phosphorylation of the ETS-domain TF Elk-1 at serine residues by MAPKs, which enhances its transcriptional activation potential in response to mitogenic signals. Acetylation, catalyzed by coactivators like p300/CBP, typically promotes TF stability and DNA-binding affinity; for p53, acetylation at C-terminal lysines by p300 increases its sequence-specific DNA binding and transactivation of target genes. Ubiquitination targets TFs for proteasomal degradation, providing a key mechanism for rapid turnover; this modification is interconnected with other PTMs, such as phosphorylation, to fine-tune degradation signals. TF protein stability is tightly controlled, with half-lives ranging from minutes to hours, enabling dynamic responses to cellular needs. For example, the oncoprotein c-Myc has a short half-life of 20-30 minutes in proliferating cells, primarily due to ubiquitin-mediated proteasomal degradation, which prevents excessive accumulation. Feedback loops, including autoregulatory circuits and PTM-dependent degradation, maintain steady-state levels; in some cases, upstream TFs induce synthesis to sustain activity during prolonged signaling. These mechanisms collectively ensure that TF levels and initial activity states are precisely calibrated for cellular homeostasis.

Nuclear Localization and DNA Accessibility

Transcription factors (TFs) must be transported from the cytoplasm to the nucleus to access DNA targets, a process regulated by nuclear localization signals (NLS) and nuclear export signals (NES). The NLS, typically a short sequence of basic amino acids, is recognized by importin α/β heterodimers, which facilitate active transport through nuclear pores via Ran-GTP gradients. For example, in STAT1, tyrosine phosphorylation exposes the NLS in its coiled-coil domain, enabling importin-mediated nuclear entry and retention until dephosphorylation allows export. Conversely, NES sequences mediate nuclear export via the exportin CRM1 (also known as XPO1), as seen in TFEB where phosphorylation at specific serine residues activates the NES for CRM1 binding, promoting cytoplasmic relocation in nutrient-replete conditions. Many TFs, such as STAT family members, undergo continuous nucleocytoplasmic shuttling, balancing nuclear accumulation with export to fine-tune transcriptional responses. Post-translational modifications, like phosphorylation, can influence these localization signals to regulate TF nuclear entry. Once in the nucleus, TFs encounter chromatin barriers that restrict DNA accessibility, but certain pioneer TFs can bind closed chromatin to initiate remodeling. Pioneer factors, such as FOXA and PU.1, possess winged-helix or ETS domains that enable binding to nucleosomal DNA, displacing linker histone H1 and partially unwrapping nucleosomes to expose binding sites. FOXA, for instance, maintains an accessible nucleosome configuration at liver-specific enhancers by evicting H1 and facilitating subsequent TF binding. PU.1 similarly opens compacted chromatin arrays in a motif-specific manner and recruits the SWI/SNF chromatin remodeling complex via its N-terminal domain, leading to ATP-dependent nucleosome displacement and extended DNA accessibility. These actions allow non-pioneer TFs to access previously inaccessible regions, amplifying transcriptional activation. Epigenetic modifications further modulate TF access by altering chromatin structure. Histone modifications like H3K27me3 and H3K9me3 promote heterochromatin compaction, inhibiting TF binding, while activating marks such as H3K27ac and H3K4me loosen chromatin to enhance accessibility. DNA methylation at CpG islands, catalyzed by DNMTs, creates repressive barriers that block TF motifs, as hypermethylation reduces binding affinity in silenced genes. However, TFs can overcome these barriers; pioneer factors recruit histone-modifying enzymes to deposit activating marks or demethylases like TET proteins, inducing local chromatin opening and enabling cooperative binding by other factors. In differentiated cells, TF access is often restricted to cell-type-specific enhancers through priming mechanisms that establish poised chromatin states. During endodermal lineage progression, enhancers acquire H3K4me1 marks at the gut tube stage, priming them for activation without immediate transcription, as seen in pancreatic and hepatic progenitors. Pioneer TFs like FOXA1/2 bind these primed enhancers early, recognizing motifs in closed chromatin to confer developmental competence and facilitate signal-dependent recruitment of lineage-specific TFs such as PDX1. This priming ensures precise, cell-type-restricted gene expression, with stronger FOXA motifs correlating to earlier binding and broader organ fate potential in foregut derivatives.

Classification

Structural Classes

Transcription factors are classified into structural classes primarily based on the architecture of their DNA-binding domains, which determine how they recognize and interact with specific DNA sequences. This classification highlights the diversity of motifs evolved to achieve sequence-specific binding, with eukaryotic transcription factors exhibiting a broader array of complex domains compared to their prokaryotic counterparts. In humans, approximately 1,639 genes encode transcription factors, representing about 8% of the protein-coding genome, with the majority belonging to a few dominant structural families. The zinc finger class, particularly the C2H2 subtype, is the largest in eukaryotes, comprising proteins with tandemly arranged zinc-coordinated modules that grip DNA via alpha-helices inserting into the major groove. A classic example is transcription factor IIIA (TFIIIA), which binds the internal control region of 5S rRNA genes using nine zinc fingers. In the human genome, this class includes around 747 members, underscoring their prevalence in regulatory networks. Basic helix-loop-helix (bHLH) factors feature a bipartite domain where a basic region contacts DNA and an adjacent helix-loop-helix motif facilitates dimerization for cooperative binding. Prominent examples include Myc and Max, which form heterodimers to regulate cell proliferation genes. Humans possess about 108 bHLH transcription factors, often involved in developmental and physiological processes through dimerization-dependent specificity. Nuclear receptors represent a ligand-activated class with a DNA-binding domain containing two zinc fingers and a ligand-binding domain that modulates activity upon hormone or small molecule binding. The estrogen receptor (ER) exemplifies this, binding estrogen response elements to control reproductive gene expression. This class includes roughly 46 human members, highlighting their role in inducible regulation. Homeodomain proteins contain a 60-amino-acid helix-turn-helix motif that binds AT-rich sequences, often in combinatorial codes for spatial patterning. Engrailed, a Drosophila homeodomain factor conserved in vertebrates, regulates segmentation genes. In humans, this class encompasses approximately 196 genes, reflecting expansion in metazoan genomes for developmental complexity. Other notable eukaryotic motifs include the Rel homology domain in NF-κB family factors, which dimerizes to bind kappa-B sites; the ETS domain, a winged helix-turn-helix in about 27 human factors like ETS1 for immune responses; and the MADS-box domain in 5 human proteins, such as MEF2 for muscle differentiation. These structures often correlate with dimerization (e.g., Rel, ETS) or specific binding modes. In prokaryotes, structural classes are simpler and more conserved, with helix-turn-helix motifs dominating. The LysR-type regulators, featuring an N-terminal DNA-binding helix-turn-helix and C-terminal effector domain, control catabolic and virulence genes in bacteria like E. coli. Sigma factors (σ), integral subunits of RNA polymerase, use helix-turn-helix regions to recognize promoter -10 and -35 elements, with multiple paralogs enabling stress responses. Eukaryotic classes have expanded from these prokaryotic foundations through gene duplication and domain shuffling.

Mechanistic and Functional Classes

Transcription factors (TFs) can be classified mechanistically based on their modes of action in regulating gene expression. Activators enhance transcription by recruiting components of the transcriptional machinery to promoter regions. For instance, the viral protein VP16 acts as a potent activator by directly recruiting RNA polymerase II (Pol II) and associated factors through protein-protein interactions, thereby stimulating the assembly of the pre-initiation complex. Repressors, in contrast, inhibit transcription by interfering with activator function or promoting chromatin compaction. The repressor element-1 silencing transcription factor (REST) exemplifies this by recruiting histone deacetylase (HDAC) complexes, such as those containing HDAC1 and HDAC2, to deacetylate histones and condense chromatin, thereby silencing neuronal genes in non-neuronal cells. Co-regulators, including co-activators and co-repressors, do not bind DNA directly but modulate TF activity by bridging interactions or altering chromatin structure. The co-activator p300 serves as a scaffold, linking TFs to the basal transcriptional machinery and acetylating histones to promote an open chromatin state conducive to transcription. Functionally, TFs are categorized by their roles in cellular contexts, such as constitutive maintenance, signal-responsive activation, or developmental specification. Housekeeping TFs maintain basal expression of essential genes across cell types. The specificity protein 1 (Sp1) is a prototypical housekeeping TF that binds GC-rich promoters to drive constitutive expression of genes involved in fundamental cellular processes like metabolism and DNA repair. Inducible TFs respond to extracellular signals to rapidly alter gene expression in specific conditions, such as immune responses. Nuclear factor of activated T-cells (NFAT) proteins are inducible TFs activated by calcium signaling in T cells, where they translocate to the nucleus to promote cytokine genes like interleukin-2 during immune activation. Developmental TFs orchestrate lineage commitment and differentiation programs. GATA family members, particularly GATA1 and GATA2, regulate hematopoiesis by controlling erythroid and megakaryocytic differentiation through sequential binding and activation of lineage-specific genes. A specialized mechanistic subclass distinguishes pioneer TFs from settler TFs based on chromatin interaction dynamics. Pioneer TFs bind closed or inaccessible chromatin, initiating remodeling to expose binding sites for other factors; examples include factors from the Klf/Sp and ETS families that displace nucleosomes and increase local accessibility. Settler TFs, conversely, preferentially bind chromatin that has been pre-opened by pioneers or other remodelers, such as Myc/MAX or nuclear receptors, and stabilize regulatory complexes without initiating access. These categories often overlap, as many TFs exhibit context-dependent roles—acting as activators in one setting and repressors in another—or switch between pioneer and settler functions during dynamic processes like development.

Evolutionary Aspects

Conservation Across Species

Transcription factors exhibit remarkable evolutionary conservation, reflecting their fundamental role in gene regulation across all domains of life. Core components such as the TATA-binding protein (TBP) and TFIIB are ancient transcription initiation factors preserved from archaea to eukaryotes, underscoring a shared mechanistic heritage for basal transcription machinery. In bacteria, the σ70 family of sigma factors, which direct RNA polymerase to promoters, displays high sequence conservation in key regions (2 and 4), enabling specific promoter recognition and initiating transcription in prokaryotes, with homologs extending to plastids in plants. This conservation highlights the deep evolutionary roots of transcription factor function, predating the divergence of bacteria, archaea, and eukaryotes over 3 billion years ago. Eukaryotic transcription factor repertoires have expanded significantly through gene duplication events, leading to increased complexity and diversification. Whole-genome duplications and tandem duplications have amplified TF families, such as those with zinc-finger or homeodomain motifs, allowing for specialized regulatory roles in multicellular organisms. For instance, in plants and animals, these duplications account for over 90% of TF family expansions in certain lineages, enabling finer control of developmental and environmental responses. In contrast to prokaryotes, where TFs constitute approximately 6% of the genome (e.g., ~300 in Escherichia coli), eukaryotic genomes allocate a similar or slightly higher proportion: ~6% in yeast (~300 TFs in Saccharomyces cerevisiae) and ~8% in humans (~1,600 TFs), though eukaryotic systems rely more on combinatorial interactions among fewer TFs per target gene. DNA-binding domains (DBDs) of transcription factors show strong conservation across species, while transactivation domains (TADs) are more variable. Motifs like the helix-turn-helix (HTH), a prevalent structural class in prokaryotes, are retained in archaeal and eukaryotic TFs, facilitating sequence-specific DNA recognition in diverse contexts. TADs, often intrinsically disordered regions, exhibit low sequence similarity despite functional equivalence, allowing flexibility in co-factor recruitment across evolutionary distances. Functionally, orthologous TFs maintain conserved roles; for example, p53 family proteins regulate stress responses, including DNA damage-induced cell cycle arrest and apoptosis, from ancient metazoans like Trichoplax adhaerens to humans. This preservation ensures robust gene regulation amid genomic changes.

Role in Adaptation and Speciation

Variations in transcription factors (TFs) and their binding sites have played a pivotal role in evolutionary adaptation by enabling fine-tuned changes in gene expression without disrupting core developmental processes. Cis-regulatory mutations, particularly in enhancer regions, allow for modular evolution where specific TF binding sites evolve to alter spatial or temporal patterns of gene activation. A classic example is the even-skipped (eve) stripe 2 enhancer in Drosophila, where nucleotide substitutions in binding sites for TFs such as Bicoid, Hunchback, Krüppel, and Giant have accumulated over evolutionary time, leading to species-specific modifications in embryonic patterning while preserving overall enhancer function. These changes demonstrate how subtle cis-regulatory evolution can drive morphological diversification across Drosophila species. Gene duplications of TFs provide another mechanism for evolutionary innovation, allowing redundant copies to acquire novel functions through subfunctionalization or neofunctionalization. In vertebrates, the Hox gene clusters, which encode homeodomain TFs critical for body plan specification, underwent two rounds of whole-genome duplication early in vertebrate evolution, resulting in four clusters (HoxA-D) from an ancestral single cluster. This duplication event expanded the regulatory repertoire, enabling greater complexity in axial patterning and facilitating adaptations such as the diversification of vertebrate appendages and sensory structures. The retention of duplicated Hox clusters correlates with morphological innovations, underscoring how TF duplication contributes to adaptive radiation. Specific adaptations illustrate how TF-related changes respond to environmental pressures. In humans, lactase persistence—the ability to digest lactose into adulthood—evolved independently in pastoralist populations through mutations in an enhancer region upstream of the LCT gene, creating or enhancing binding sites for TFs like Oct-1 (encoded by POU2F1) and HNF1α. The -13910T>C variant, for instance, strengthens Oct-1 binding, boosting LCT transcription and conferring a selective advantage in dairy-consuming societies. Similarly, in Darwin's finches, variation in beak morphology, adapted to different food sources, arises from differences in Bmp4 expression levels in the developing facial mesenchyme, regulated by upstream TFs that modulate signaling intensity to influence beak depth and width. Experimental overexpression of Bmp4 in avian embryos recapitulates these deep, broad beak phenotypes, highlighting TF-mediated regulation as a driver of adaptive morphological evolution. In speciation, rewiring of TF networks can generate reproductive isolation by altering behavioral or developmental traits. The fruitless (fru) TF in Drosophila, which is sex-specifically spliced to direct male courtship circuitry, exhibits species-specific wiring; for example, in D. subobscura, fru-labeled neurons mediate unique food-gifting behaviors during courtship, distinct from the song-based rituals in D. melanogaster, contributing to behavioral divergence and prezygotic isolation. Hybrid incompatibilities further promote speciation when TF-binding site mismatches disrupt gene regulation in hybrids. Computational models of TF-DNA interactions show that divergent evolution of TF sequences and cis-sites can lead to misbinding in hybrids, causing dysregulated expression and inviability, as simulated in sequence-based bioenergetic frameworks where compensatory mutations in parental lineages create incompatibilities. Recent advances in evolutionary developmental biology (evo-devo) have revealed TF roles in climate adaptation. In corals, modular gene regulatory networks involving TFs exhibit developmental system drift. Transcriptomic studies of coral responses to thermal stress across life stages highlight how population origin and developmental stage modulate gene expression, potentially constraining or enabling adaptation to ocean warming. These 2020s studies emphasize TFs as key nodes in evo-devo networks for environmental adaptation.

Clinical and Applied Significance

Associated Diseases

Dysfunction of transcription factors (TFs), through genetic mutations or dysregulation, underlies a variety of human diseases by disrupting gene expression programs critical for development, homeostasis, and cellular responses. In genetic disorders, heterozygous loss-of-function mutations in TF-encoding genes often lead to haploinsufficiency, resulting in developmental anomalies. For instance, mutations in the FOXP2 gene, which encodes a forkhead box TF essential for neural circuit formation in speech-related brain regions, cause FOXP2-related speech and language disorder, characterized by childhood apraxia of speech and impairments in expressive and receptive language skills beginning in early childhood. Similarly, RUNX2 mutations, affecting a runt-related TF that regulates osteoblast differentiation and bone formation, are the primary cause of cleidocranial dysplasia, an autosomal dominant skeletal disorder featuring hypoplastic or absent clavicles, delayed fontanelle closure, and dental abnormalities due to impaired cranial bone development. PAX6 mutations, disrupting a paired box TF vital for eye and brain development, result in aniridia, a condition marked by iris hypoplasia, foveal hypoplasia, and increased glaucoma risk, often as part of the broader WAGR syndrome involving Wilms tumor predisposition. More recently, variants in TFAP2A, encoding an AP-2 alpha TF involved in craniofacial and ectodermal patterning, have been linked to branchio-oculo-facial syndrome (BOFS), presenting with branchial arch anomalies, ocular defects like coloboma, and facial clefts; a 2025 study identified a novel heterozygous TFAP2A variant in a familial case emphasizing predominant ocular features, confirming its role in atypical presentations. In cancer, aberrant TF activity drives oncogenesis by promoting uncontrolled proliferation, survival, and metastasis. Deregulation of the MYC proto-oncogene, encoding a basic helix-loop-helix TF that amplifies transcription of growth-related genes, occurs in over 50% of human cancers, including Burkitt lymphoma and breast cancer, where it enhances tumor aggression and poor prognosis through global transcriptional amplification. Mutations in TP53, which encodes the p53 tumor suppressor TF that activates DNA repair and apoptosis pathways, are found in approximately 50% of all human cancers, with high frequencies (up to 89% in small cell lung cancer) leading to loss of tumor suppression and genomic instability. Fusion TFs, such as PML-RARA resulting from t(15;17) translocation in acute promyelocytic leukemia (APL), act as dominant-negative regulators of retinoic acid signaling, blocking myeloid differentiation and promoting leukemic blast accumulation; this fusion is present in nearly all APL cases and drives the disease's hallmark coagulopathy and promyelocyte maturation arrest. Neurological disorders also arise from TF dysregulation, often exacerbating neurodegeneration. In Alzheimer's disease (AD), reduced levels of REST (RE1-silencing transcription factor), a repressor of neuronal genes in non-neuronal contexts, correlate with increased amyloid-beta pathology and tau hyperphosphorylation; postmortem studies show REST nuclear loss in AD brains, linking it to accelerated cognitive decline and stress vulnerability in aging neurons. In Huntington's disease (HD), mutant huntingtin sequesters and impairs MEF2 (myocyte enhancer factor 2) TFs, which normally promote neuronal survival and synaptic plasticity; this leads to reduced MEF2 activity in the hippocampus and striatum, contributing to cognitive deficits, muscle atrophy, and progressive motor symptoms in HD models and patients. Recent advances in CRISPR-based screens (2023–2025) have illuminated TF variants in rare diseases by systematically perturbing TF function to reveal causal links. For example, large-scale CRISPR knockout screens of all known TFs have identified regulatory variants affecting epidermal differentiation genes in skin disorders, while single-cell CRISPR editing has pinpointed noncoding variants disrupting TF binding in neurodevelopmental syndromes, expanding the genetic architecture of rare conditions beyond coding mutations. These approaches, including joint multiomic phenotyping, underscore how rare TF variants contribute to disease heterogeneity, as seen in refined BOFS models.

Therapeutic Targeting and Biotechnological Uses

Transcription factors (TFs) represent promising therapeutic targets due to their central role in regulating gene expression underlying diseases such as cancer and genetic disorders. Small-molecule inhibitors have been developed to modulate TF activity, with ibrutinib serving as a notable example in B-cell lymphomas. By inhibiting Bruton's tyrosine kinase (BTK), ibrutinib disrupts downstream STAT3 signaling, which promotes cell survival in diffuse large B-cell lymphoma (DLBCL), thereby enhancing the efficacy of chemotherapy regimens like R-CHOP in non-germinal center B-cell-like subtypes. For more direct TF degradation, proteolysis-targeting chimeras (PROTACs) offer a strategy to induce ubiquitin-proteasome-mediated breakdown. Vepdegestrant (ARV-471), an oral PROTAC, selectively degrades the estrogen receptor (ER), a nuclear TF driving hormone-dependent breast cancers, achieving over 90% ER protein reduction in preclinical models compared to 63% with fulvestrant, and demonstrating antitumor activity in endocrine-resistant xenografts. Gene editing technologies like CRISPR-Cas9 enable precise modulation of TF genes to treat monogenic diseases. In sickle cell disease, CRISPR editing of the BCL11A TF gene in hematopoietic stem cells disrupts its repression of fetal hemoglobin production, restoring functional hemoglobin levels and alleviating sickling. The therapy exagamglogene autotemcel (Casgevy), approved by the FDA in 2023, has shown durable clinical responses in phase 1/2 trials, with 29 of 31 patients achieving transfusion independence for at least 12 months post-infusion. In synthetic biology, engineered TF-based circuits provide programmable control over gene expression for therapeutic applications. Synthetic transcription factors (synTFs) have been designed to regulate transgene expression in cell therapies, such as CAR-T cells, by responding to exogenous inducers like small molecules, thereby improving safety and efficacy through inducible activation or repression of target genes. Biotechnological applications extend TF engineering to agriculture and biocontrol. In plants, overexpression of WRKY TFs enhances tolerance to abiotic stresses, as seen with TaWRKY10 in wheat, which improves drought and salt resistance in transgenic tobacco by accumulating osmolytes like proline and soluble sugars, without compromising growth. For microbial biocontrol, engineered TFs in bacteria can confer resistance to environmental stressors, including pesticides. Emerging strategies leverage artificial intelligence (AI) and nanotechnology for advanced TF targeting. AI-driven design has accelerated the discovery of TF inhibitors. Additionally, nanoparticle delivery systems facilitate TF modulation in cancer therapy.

Analysis and Resources

Experimental and Computational Methods

Experimental methods for studying transcription factors (TFs) primarily focus on identifying binding sites, measuring activity, and detecting interactions. Chromatin immunoprecipitation followed by sequencing (ChIP-seq) is a cornerstone technique for genome-wide mapping of TF binding sites in vivo, enabling the identification of direct targets by isolating protein-DNA complexes and sequencing the associated DNA fragments. Reporter assays, such as those using firefly luciferase fused to promoter regions, quantify TF transcriptional activity by measuring reporter gene expression levels in transfected cells, providing insights into regulatory strength and context-specific effects. The yeast one-hybrid system screens for TF-DNA interactions by fusing a TF to a transcriptional activation domain and testing binding to bait DNA sequences integrated into yeast reporter genes, facilitating high-throughput discovery of binding partners. Recent advancements include single-cell assay for transposase-accessible chromatin with sequencing (scATAC-seq), which profiles chromatin accessibility at single-cell resolution to infer TF activity through open chromatin regions enriched for motifs, revealing cell-type-specific regulatory landscapes. Biochemical approaches complement these by assessing binding affinity and complex composition. Electrophoretic mobility shift assay (EMSA) detects TF-DNA interactions in vitro by observing shifts in DNA mobility upon protein binding in native gel electrophoresis, allowing quantification of binding affinities through competition experiments. Mass spectrometry (MS) identifies post-translational modifications (PTMs) on TFs and their associated protein complexes by analyzing affinity-purified samples, uncovering regulatory modifications like phosphorylation that modulate activity and dynamic interactomes. Computational methods enable motif discovery, binding prediction, and network reconstruction from genomic data. The MEME suite employs expectation maximization to identify ungapped motifs in unaligned DNA or protein sequences, aiding de novo discovery of TF binding motifs from ChIP-seq peaks or co-expressed genes. Machine learning models like DeepTF integrate convolutional neural networks with long short-term memory layers to predict TF binding sites from sequence features, achieving high accuracy on diverse ChIP-seq datasets by capturing multi-scale contextual information. For inferring TF regulatory networks from single-cell RNA-seq (scRNA-seq) and multi-omics data, methods such as SCENIC and its 2023 extension SCENIC+ reconstruct gene regulatory networks by combining co-expression modules with motif enrichment and chromatin accessibility analysis, identifying TF regulons and enhancer-driven interactions that define cell states without requiring prior binding data. Recent advances leverage structural biology and artificial intelligence for deeper mechanistic insights. Cryo-electron microscopy (cryo-EM) has resolved high-resolution structures of TF-pre-initiation complexes (PICs), such as those involving TFIIIC on RNA polymerase III promoters, illuminating assembly dynamics and TF positioning at core promoters. AI-driven models like Enformer enhance variant effect prediction by modeling long-range chromatin interactions to forecast how noncoding variants disrupt TF binding and gene expression, improving interpretation of regulatory mutations.

Databases and Tools

Several major databases serve as foundational resources for transcription factor (TF) data, including motifs, annotations, and binding information. TRANSFAC is a comprehensive, manually curated database containing over 49,000 eukaryotic TFs, their DNA-binding sites, and binding profiles, enabling analysis of gene regulation mechanisms. JASPAR provides the largest open-access collection of non-redundant TF binding profiles, primarily in the form of position weight matrices (PWMs), covering profiles from vertebrates, plants, insects, nematodes, and other taxa to support motif-based predictions. The 2024 update (10th release) expanded the JASPAR CORE collection by 20%, adding 329 new profiles and upgrading 72 existing ones. AnimalTFDB offers extensive annotations and classifications of TFs, cofactors, and chromatin remodelers across 183 animal species, including ortholog mappings and expression data for comparative studies. The ENCODE project delivers genome-wide maps of TF binding in human cells, integrating experimental data from ChIP-seq assays with motif instances to reveal regulatory landscapes. Specialized databases focus on targeted aspects of TF organization and species-specific details. TFCat is a curated catalog of mouse and human TFs, emphasizing functional classifications derived from expert-reviewed literature to aid in identifying regulatory networks. FlyTF catalogs computationally predicted and experimentally verified site-specific TFs in Drosophila melanogaster, providing annotations on DNA-binding domains and expression patterns for model organism research. TFClass maintains a hierarchical structural classification of eukaryotic TFs based on DNA-binding domains. Recent tools like TFClassPredict (2024) incorporate machine learning using the TFClass hierarchy and TFBS data from UniBind to enhance predictions. Key software tools facilitate TF motif scanning, binding prediction, and structural analysis. PROMO is a web-based tool for identifying putative TF binding sites in DNA sequences by scanning against TRANSFAC matrices, accounting for species-specific variations and weight thresholds. TRAP (Transcription factor Affinity Prediction) employs a biophysical model to compute relative binding affinities of TFs to DNA sequences, useful for analyzing ChIP-seq data and regulatory variants. Many resources integrate with UniProt, which annotates TF domains, functions, and predicted structures, allowing seamless access to sequence and 3D model data for over 200 million proteins. Accessibility varies across these resources, with open-source options like JASPAR, AnimalTFDB, and ENCODE promoting broad use through free downloads and APIs, while proprietary databases such as TRANSFAC require subscriptions for full access via platforms like geneXplain. Recent updates, including post-2023 expansions of the AlphaFold Protein Structure Database, provide open-access predicted 3D models for numerous TFs, covering over 214 million entries to support structural studies of DNA-binding domains.

References

  1. [1]
    Regulation of Transcription and Gene Expression in Eukaryotes
    Transcription factors (TFs) are regulatory proteins whose function is to activate (or more rarely, to inhibit) transcription of DNA by binding to specific DNA ...
  2. [2]
    Mechanisms and biotechnological applications of transcription factors
    Aug 31, 2023 · Transcription factors (TFs) play a pivotal role in regulating the transcription of target genes by selectively recognizing and binding specific ...
  3. [3]
    Transcription Factors and Transcriptional Control | Learn Science at Scitable
    ### Summary of Transcription Factors from Nature Education Article
  4. [4]
    Non-coding RNAs: key regulators of mammalian transcription - PMC
    Multiple ncRNAs have been shown to control gene expression by regulating the activity of transcriptional activators, coregulators, and the general transcription ...
  5. [5]
    [PDF] Transcriptional Regulatory Elements in the Human Genome
    Long-range transcriptional regulatory elements, including enhancers, silencers, in- sulators, and LCRs, may function through a DNA-looping mechanism that brings.
  6. [6]
    A 50 year history of technologies that drove discovery in eukaryotic ...
    Mar 31, 2020 · This article highlights the progress that has been made in understanding eukaryotic transcriptional mechanisms using an array of disciplines.
  7. [7]
  8. [8]
    Systematic discovery of uncharacterized transcription factors in ...
    Of the estimated 304 candidate transcription factors (TFs) in Escherichia coli K-12 MG1655, 185 have been experimentally identified, but ChIP methods have been ...
  9. [9]
    many faces of the helix-turn-helix domain: Transcription regulation ...
    The helix-turn-helix (HTH) domain is a common denominator in basal and specific transcription factors from the three super-kingdoms of life.
  10. [10]
    Transcription factor structure and DNA binding - ScienceDirect.com
    The metal-binding domains insert into the major grooves on opposite faces of the DNA, and additional contacts with the backbone are mediated by residues in the ...
  11. [11]
    DNA-Binding Motifs in Gene Regulatory Proteins - NCBI - NIH
    The common motifs include the helix-turn-helix, the homeodomain, the leucine zipper, the helix-loop-helix, and zinc fingers of several types. The precise ...
  12. [12]
    A natural classification of the basic helix–loop–helix class of ... - PNAS
    Helix-loop-helix proteins are characterized by common possession of highly conserved bipartite domains for DNA binding and protein–protein interaction (1–2). A ...Abstract · Sign Up For Pnas Alerts · Lz-Containing Sequences
  13. [13]
    The many faces of the helix-turn-helix domain - PubMed - NIH
    The helix-turn-helix (HTH) domain is a common denominator in basal and specific transcription factors from the three super-kingdoms of life.
  14. [14]
    Early evolutionary origin of major homeodomain sequence classes
    Based on our results, homeodomain sequences can be classified into at least 30 distinguishable classes. Sequences belonging to the same class are highly ...
  15. [15]
    Determinants of p53 DNA binding, gene regulation, and cell fate ...
    Jun 29, 2024 · Transcription factors contain DNA binding domains that enable them to recognize and bind short DNA sequences. Sequence content and context in ...
  16. [16]
    Zinc-finger domains in metazoans: evolution gone wild
    Sep 6, 2017 · A new study uncovers a potential mechanism that may allow zinc-finger domains in metazoans to recognize and bind virtually any DNA sequence.
  17. [17]
    Did homeodomain proteins duplicate before the origin of ... - PNAS
    These results support the hypothesis that there was at least one duplication of homeobox genes before the origin of angiosperms, fungi, and metazoa.
  18. [18]
  19. [19]
  20. [20]
  21. [21]
  22. [22]
    Transcription-factor binding and sliding on DNA studied using micro
    Transcription factors search for specific operator sequences by alternating rounds of 3D diffusion with rounds of 1D diffusion (sliding) along the DNA.
  23. [23]
    Facilitated DNA Search by Multidomain Transcription Factors - NIH
    Electrostatic interactions play a key role in protein-DNA binding and are relevant to any DNA sequence. Consequently, they dictate nonspecific interactions ...
  24. [24]
    Quantification of transcription factor-DNA binding affinity in a living cell
    Dec 10, 2015 · The apparent dissociation constant (Kd) for specific binding of glucocorticoid receptor (GR) and androgen receptor (AR) to DNA was determined in ...
  25. [25]
    Cell-type-specific binding of the transcription factor CREB to ... - PNAS
    Sep 1, 2004 · CREB binds to a target sequence termed the cAMP-response element (CRE) found in many cellular and viral gene promoters.
  26. [26]
    The Mediator complex as a master regulator of transcription by RNA ...
    Jun 20, 2022 · In general, TFs activate transcription by recruiting Mediator and chromatin modifying factors such as CBP or chromatin remodelling complexes ...Missing: seminal | Show results with:seminal
  27. [27]
    TFIID and human mediator coactivator complexes assemble ...
    The present study was designed to address whether purified TFIID and hMed coactivator complexes can cooperatively assemble on promoter DNA in a functionally ...Missing: seminal | Show results with:seminal
  28. [28]
    Human CRSP interacts with RNA polymerase II CTD and adopts a ...
    Together, these findings establish that the activator-targeted human CRSP coactivator can interact with the CTD of RNA Pol II. By virtue of this interaction ...
  29. [29]
    The Mediator complex: a central integrator of transcription - PMC
    Jul 27, 2016 · Numerous labs have shown cooperative and even synergistic functions between Mediator and TFIID during PIC assembly and transcription activation; ...The Mediator Complex: A... · Mediator Composition And... · Mediator Structural Dynamics
  30. [30]
    A mechanism for transcriptional repression dependent on the ... - NIH
    Apr 17, 2007 · We find in these experiments that the E3 ubiquitin ligase activity of BRCA1 strongly inhibits transcription by blocking PIC assembly.
  31. [31]
    Hypoxia Actively Represses Transcription by Inducing Negative ...
    PIC assembly and RNA polymerase II C-terminal domain (CTD) phosphorylation were blocked by hypoxic induction and core promoter binding of negative cofactor 2 ...
  32. [32]
    Identifying the combinatorial control of signal-dependent ...
    Noncooperative interactions between transcription factors and clustered DNA binding sites enable graded transcriptional responses to environmental inputs.
  33. [33]
    Transcriptional Control in the Segmentation Gene Network of ... - NIH
    The segmentation gene network of Drosophila consists of maternal and zygotic factors that generate, by transcriptional (cross-) regulation, expression patterns.
  34. [34]
    Hox genes in development and beyond - PMC - PubMed Central
    ABSTRACT. Hox genes encode evolutionarily conserved transcription factors that are essential for the proper development of bilaterian organisms.Hox Genes In Development And... · Hox Gene Regulation · Mechanisms Of Hox...Missing: seminal | Show results with:seminal
  35. [35]
    Activation of muscle-specific genes in pigment, nerve, fat, liver, and ...
    Jul 15, 1989 · The ability of MyoD to activate muscle genes in a variety of differentiated cell lines suggests that no additional tissue-specific factors other ...
  36. [36]
    Induction of Ectopic Eyes by Targeted Expression of the eyeless ...
    Georg Halder et al. ,. Induction of Ectopic Eyes by Targeted Expression of the eyeless Gene in Drosophila.Science267,1788-1792(1995).DOI:10.1126/science.7892602 ...
  37. [37]
    NF-κB signaling in inflammation - Nature
    Jul 14, 2017 · The processing of p100 involves degradation of its C-terminal IκB-like structure, resulting in generation of mature NF-κB2 p52 and nuclear ...
  38. [38]
    The p53 network: Cellular and systemic DNA damage responses in ...
    The p53 network mediates cell responses to DNA damage, including cell cycle arrest, senescence, and apoptosis, and is a key player in tumor suppression and ...Missing: paper | Show results with:paper
  39. [39]
    Activation of hypoxia-inducible factor 1α - PNAS
    In support of this model we demonstrate here that HIF-1α protein levels are dramatically and rapidly elevated after induction of the hypoxic response. Clearly, ...
  40. [40]
    Dynamic control of Hsf1 during heat shock by a chaperone switch ...
    Nov 10, 2016 · Upon heat shock, the chaperones are titrated away by unfolded or misfolded proteins, leaving Hsf1 free to activate transcription of chaperone ...
  41. [41]
    Frontiers | New insights in glucocorticoid receptor signaling
    Feb 5, 2017 · Beyond the consensus that steroidal or analog binding induces GR nuclear translocation, several evidences have shown that GR can be activated in ...
  42. [42]
    The JAK/STAT signaling pathway: from bench to clinic - Nature
    Nov 26, 2021 · More than 50 cytokines and growth factors have been identified in the JAK/STAT signaling pathway, such as hormones, interferons (IFN), ...
  43. [43]
    Wnt/β-catenin signalling: function, biological mechanisms ... - Nature
    Jan 3, 2022 · The Wnt/β-catenin pathway comprises a family of proteins that play critical roles in embryonic development and adult tissue homeostasis.
  44. [44]
    Crosstalk in NF-κB signaling pathways | Nature Immunology
    Jul 19, 2011 · Stimulus-induced degradation of IκB proteins is initiated through phosphorylation by the IκB kinase (IKK) complex, which consists of two ...
  45. [45]
    Dynamical gene regulatory networks are tuned by transcriptional ...
    Jul 31, 2020 · TFs that autoregulate have more interactions with microRNAs than non-autoregulatory genes and 89% of autoregulatory TFs were found in dual ...
  46. [46]
    Transcriptional Regulation of NF-κB2: Evidence for κB-Mediated ...
    Similar to other NF-κB family members, NF-κB2 is positively autoregulated. In contrast to other family members, we find that κB elements in the NFKB2 promoter ...<|separator|>
  47. [47]
    Article mRNA Destabilization Is the Dominant Effect of Mammalian ...
    Oct 2, 2014 · Regardless of the miRNA, cell type, growth condition, or translational state, mRNA destabilization explains most (66%–>90%) miRNA-mediated repression.Article · Results · Discussion
  48. [48]
    Activation of ternary complex factor Elk-1 by MAP kinases - PubMed
    We demonstrate that recombinant Elk-1 is hyperphosphorylated in vivo upon joint overexpression of MAPKs and constitutively activated Raf-1 kinase.<|separator|>
  49. [49]
    Activation of p53 Sequence-Specific DNA Binding by Acetylation of ...
    The acetylation by p300 of these lysines in the C termi- p300/CBP has emerged as a transcriptional coactivator for a broad group of cellular DNA-binding ...
  50. [50]
    An inventory of crosstalk between ubiquitination and other post ...
    May 19, 2023 · We provide an extensive inventory of ubiquitination crosstalk with multiple PTMs including SUMOylation, phosphorylation, methylation, acetylation, ...
  51. [51]
    MYC (MYC proto-oncogene, bHLH transcription factor)
    Aug 1, 2017 · Both MYC mRNA and protein have very short half-lives in normal cells (20-30 minutes each). Without appropriate positive regulatory signals, MYC ...
  52. [52]
    Regulation of transcription factor activity by interconnected, post ...
    Post-translational modifications regulate every aspect of transcription factor function and coordinate access of RNA polymerases to promoter templates. Site- ...Missing: paper | Show results with:paper
  53. [53]
  54. [54]
    Activator-Mediated Recruitment of the RNA Polymerase II Machinery ...
    Eukaryotic transcriptional activators bind to enhancer elements and stimulate the RNA polymerase II (pol II) machinery via functionally autonomous activation ...
  55. [55]
    REST Acts through Multiple Deacetylase Complexes: Neuron
    HDACs catalyze the removal of acetyl-lysine groups from the histone N termini and have been found to be a pervasive mechanism of transcriptional repression in ...
  56. [56]
    p300/CBP proteins: HATs for transcriptional bridges and scaffolds
    p300/CBP transcriptional co-activator proteins play a central role in co-ordinating and integrating multiple signal-dependent events with the transcription ...
  57. [57]
    The Role of the Ubiquitously Expressed Transcription Factor Sp1 in ...
    Dec 23, 2016 · Sp1 belongs to the 26 member strong Sp/KLF family of transcription factors. It is a paradigm for a ubiquitously expressed transcription ...
  58. [58]
    Transcription Factor NFAT - an overview | ScienceDirect Topics
    Transcription factor NFAT is defined as an inducible transcription factor that plays a crucial role in regulating the immune response of T cells ...
  59. [59]
    GATA family transcriptional factors: emerging suspects in ...
    Oct 6, 2015 · The three important GATA transcription factors GATA1, GATA2 and GATA3 play essential roles in the development and maintenance of hematopoietic systems.
  60. [60]
    Discovery of non-directional and directional pioneer transcription ...
    PIQ identifies pioneer and settler transcription factors. We next used PIQ to explore why ChIP-seq experiments have consistently shown that transcription ...
  61. [61]
    Transcriptional co-activators: emerging roles in signaling pathways ...
    Nov 13, 2023 · Depending on their mechanism of action, transcriptional co-activators can be broadly categorized into four different classes. The first class of ...
  62. [62]
    Uncovering ancient transcription systems with a novel evolutionary ...
    Jun 16, 2016 · TBP and TFIIB are evolutionarily conserved transcription initiation factors in archaea and eukaryotes. Information about their ancestral ...Missing: across | Show results with:across
  63. [63]
    The σ 70 family of sigma factors - Genome Biology - BioMed Central
    Jan 3, 2003 · Members of the σ 70 family of sigma factors are components of the RNA polymerase holoenzyme that direct bacterial or plastid core RNA polymerase to specific ...
  64. [64]
    Transcription factor evolution in eukaryotes and the assembly of the ...
    These expansions were mainly due to gene duplications, and, in some lineages, to diversification in protein domain architecture. In contrast, other TF classes ...Transcription Factor... · Results · Phylogenetic Patterns Of Tf...
  65. [65]
    Numbers of DNA-binding transcription factors - Various - BNID 109202
    coli to over 3000 in humans. These constitute between 6% (in E. coli and yeast) and 8% (in human) of the encoded proteins in these organisms. van Nimwegen's ...
  66. [66]
    P2TF: a comprehensive resource for analysis of prokaryotic ...
    Nov 15, 2012 · TFs are characterized by their DNA-binding domains (DBDs), of which the helix-turn-helix (HTH) domain is the most prevalent in prokaryotic ...<|control11|><|separator|>
  67. [67]
    Commonly asked questions about transcriptional activation domains
    Dec 5, 2023 · DNA binding domains bind the genome by recognizing structurally related DNA sequences; they are structured, conserved, and predictable from ...
  68. [68]
    Functional characterization of p53 pathway components in the ...
    Sep 28, 2016 · In response to stress, p53-dependent transactivation of target genes can result in numerous outcomes ranging from cell cycle arrest to apoptosis ...Missing: across | Show results with:across
  69. [69]
    Functional analysis of eve stripe 2 enhancer evolution in Drosophila
    Mar 1, 1998 · Our analysis revealed a large number of nucleotide substitutions in regulatory protein binding sites for bicoid, hunchback, Kruppel and giant, ...
  70. [70]
    Hox cluster duplications and the opportunity for evolutionary novelties
    We propose that the constraints on vertebrate Hox cluster structure lead to an association between the retention of duplicated Hox clusters and adaptive ...
  71. [71]
    T −13910 DNA variant associated with lactase persistence interacts ...
    The data suggest that the binding of Oct-1 to the T −13 910 variant directs increased lactase promoter activity and this might provide an explanation for the ...
  72. [72]
    Optogenetic Activation of the fruitless-Labeled Circuitry in ...
    Nov 29, 2017 · We studied the neural circuitry for mating behavior in Drosophila subobscura, a species that displays unique courtship actions not shared by other members of ...
  73. [73]
    Hybrid Incompatibility Arises in a Sequence-Based Bioenergetic ...
    We explore the evolutionary conditions that promote and constrain hybrid incompatibility in regulatory networks using a bioenergetic model (combining ...
  74. [74]
    Developmental system drift and modular gene regulatory networks ...
    Aug 14, 2025 · We compared gene expression profiles during gastrulation of Acropora digitifera and Acropora tenuis, species that diverged ∼50 million years ago ...
  75. [75]
    Divergent transcriptional response to thermal stress among life ...
    Divergent transcriptional response to thermal stress among life stages could constrain coral adaptation to climate change ... The ability for adaptation to track ...Missing: devo | Show results with:devo<|separator|>
  76. [76]
    FOXP2-Related Speech and Language Disorder - GeneReviews
    Jun 23, 2016 · The core phenotype of FOXP2-SLD is childhood apraxia of speech (CAS), a disorder of speech motor programming or planning that affects the production, ...Diagnosis · Clinical Characteristics · Genetically Related (Allelic... · Management
  77. [77]
    RUNX2 mutations in cleidocranial dysplasia patients - PubMed
    Mutations in the RUNX2 gene, a master regulator of bone formation, have been identified in cleidocranial dysplasia (CCD) patients.
  78. [78]
    PAX6-Related Aniridia - GeneReviews® - NCBI Bookshelf - NIH
    May 20, 2003 · PAX6-related aniridia occurs either as an isolated ocular abnormality or as part of the Wilms tumor-aniridia-genital anomalies-retardation (WAGR) syndrome.
  79. [79]
    TFAP2A mutations result in branchio-oculo-facial syndrome - PubMed
    We conclude BOFS is caused by mutations involving TFAP2A. More patients need to be studied to determine possible genetic heterogeneity.
  80. [80]
    A novel variant of TFAP2A in a familial case of branchio-oculo-facial ...
    Apr 15, 2025 · A family with branchio-oculo-facial syndrome with primarily ocular involvement associated with mutation of the TFAP2A gene. Ophthalmic Genet ...
  81. [81]
    The MYC oncogene — the grand orchestrator of cancer growth ... - NIH
    The MYC proto-oncogenes encode a family of transcription factors that are among the most commonly activated oncoproteins in human neoplasias.
  82. [82]
    TP53 Mutations in Human Cancers: Origins, Consequences ... - NIH
    Somatic TP53 mutations occur in almost every type of cancer at rates from 38%–50% in ovarian, esophageal, colorectal, head and neck, larynx, and lung cancers to ...
  83. [83]
    Acute Promyelocytic Leukemia: A Constellation of Molecular Events ...
    How this mutational spectrum, including point mutations in the PML-RARA fusion gene, could contribute to the 10%–15% of relapsed or resistant APL patients is ...
  84. [84]
    REST and Stress Resistance in Aging and Alzheimer's Disease - PMC
    Here we show that REST is induced in the aging human brain and regulates a network of genes that mediate cell death, stress resistance and AD pathology. This ...
  85. [85]
    MEF2 impairment underlies skeletal muscle atrophy in ... - NIH
    Apr 18, 2020 · We find that impairment of MEF2 also occurs in skeletal muscle of R6/2 mice, a model of Huntington disease where muscle atrophy is prominent.
  86. [86]
    Disease-linked regulatory DNA variants and homeostatic ... - Nature
    Sep 25, 2025 · CRISPR knockout screens of all known TFs to examine their effects on epidermal progenitor and differentiation gene expression will help ...
  87. [87]
    Precisely defining disease variant effects in CRISPR-edited single ...
    Jul 23, 2025 · Large-scale single-cell CRISPR screening approaches in cell lines enable the identification of key regulatory elements and their genes.
  88. [88]
    Functional phenotyping of genomic variants using joint multiomic ...
    Sep 1, 2025 · Genetic variants (both coding and noncoding) can impact gene function and expression, driving disease mechanisms such as cancer progression.
  89. [89]
    Ibrutinib reverses IL-6-induced osimertinib resistance through ...
    Feb 23, 2022 · IL-6/IL-6R blockade or STAT3 inhibition alone was unable to reverse osimertinib resistance. To explore the potential mediator of IL-6 ...
  90. [90]
    Oral Estrogen Receptor PROTAC Vepdegestrant (ARV-471) Is ...
    Herein, the orally bioavailable PROTAC ER degrader vepdegestrant (ARV-471) has demonstrated greater in vivo ER degradation (≥90%) compared with fulvestrant (63% ...
  91. [91]
    Arvinas and Pfizer's Vepdegestrant (ARV-471) Receives FDA Fast ...
    Feb 6, 2024 · Vepdegestrant is an investigational PROteolysis Targeting Chimera (PROTAC ® ) protein degrader designed to target and degrade the estrogen receptor (ER) ...
  92. [92]
    CRISPR-Cas9 Gene Editing for Sickle Cell Disease and β ...
    Dec 7, 2020 · BCL11A is a transcription factor that represses γ-globin expression and fetal hemoglobin in erythroid cells. We performed electroporation of ...
  93. [93]
    FDA Approves First Gene Therapies to Treat Patients with Sickle ...
    Dec 8, 2023 · Casgevy is the first FDA-approved therapy utilizing CRISPR/Cas9, a type of genome editing technology. Patients' hematopoietic (blood) stem cells ...
  94. [94]
    A Wheat WRKY Transcription Factor TaWRKY10 Confers Tolerance ...
    TaWRKY10 could enhance drought and salt stress tolerances in transgenic tobacco plants. These functions are achieved by accumulating water, proline and soluble ...
  95. [95]
    Engineering a synthetic gene circuit for high-performance inducible ...
    Apr 17, 2024 · By using a quantitative approach based on synthetic gene circuits, this study has overcome some of the major challenges of inducible gene ...
  96. [96]
    AI-Designed Molecules in Drug Discovery, Structural Novelty ...
    Aug 18, 2025 · Our central question is, to what extent are current AI-driven drug discovery methods capable of discovering truly novel chemical scaffolds, and ...
  97. [97]
    Advances in transcription factor delivery: Target selection ...
    Aug 10, 2025 · These findings suggest that TF-based therapies can either suppress oncogenic transcriptional programs or enhance immune-mediated tumor ...
  98. [98]
    Lipid nanoparticle-assisted mRNA therapy for cancer treatment
    Sep 8, 2025 · The main goal here is to achieve precise and highly effective delivery directly to tumors.13 To ensure the successful delivery of therapeutic ...
  99. [99]
    Genome-Wide Mapping of in Vivo Protein-DNA Interactions - Science
    Jun 8, 2007 · To map these protein-DNA interactions comprehensively across entire mammalian genomes, we developed a large-scale chromatin immunoprecipitation assay (ChIPSeq)
  100. [100]
    Isolation of ORC6, a Component of the Yeast Origin Recognition ...
    ... Yeast Origin Recognition Complex by a One-Hybrid System. Joachim J. Li and Ira HerskowitzAuthors Info & Affiliations. Science. 17 Dec 1993. Vol 262, Issue 5141.
  101. [101]
    Single-cell chromatin accessibility reveals principles of regulatory ...
    Jun 17, 2015 · Here we reveal the landscape and principles of mammalian DNA regulatory variation by developing a robust method for mapping the accessible genome of individual ...
  102. [102]
    Fitting a mixture model by expectation maximization to discover ...
    The algorithm described in this paper discovers one or more motifs in a collection of DNA or protein sequences by using the technique of expectation ...
  103. [103]
    TFIIIC as assembly factor and barrier in RNA polymerase III ...
    Aug 5, 2025 · The cryo-EM structure of TFIIIA- and TFIIIC-dependent RNAPIII transcription initiation on the type I promoter provides new insights into the ...
  104. [104]
    Effective gene expression prediction from sequence by integrating ...
    Oct 4, 2021 · Enformer improves variant effect prediction on eQTL data. A central goal of this research is to predict the influence of genetic variants on ...
  105. [105]
    TRANSFAC database - GeneXplain GmbH
    TRANSFAC is a database of transcription factors and their binding sites, including over 49,000 factors, used for gene regulation studies. It contains TF ...
  106. [106]
    JASPAR - A database of transcription factor binding profiles
    JASPAR is the largest open-access database of curated and non-redundant transcription factor (TF) binding profiles from six different taxonomic groups.Tools · Search profile(s) · Download JASPAR data · Documentation
  107. [107]
    AnimalTFDB 4.0: a comprehensive animal transcription factor ...
    Oct 21, 2022 · AnimalTFDB 4.0 is a useful resource for studying TF and transcription regulation, which contains comprehensive annotation and classification of ...
  108. [108]
    TFCat: the curated catalog of mouse and human transcription factors
    TFCat is a catalog of mouse and human transcription factors based on a reliable core collection of annotations obtained by expert review of the scientific ...
  109. [109]
    FlyTF: improved annotation and enhanced functionality of the ... - NIH
    FlyTF (http://www.flytf.org) is a database of computationally predicted and/or experimentally verified site-specific transcription factors (TFs) in the fruit ...
  110. [110]
    Predicting transcription factor affinities to DNA from a biophysical ...
    In this paper, we adopt a physical binding model to predict the relative binding affinity of a transcription factor for a given sequence.
  111. [111]
    tools in UniProt help search
    Protein domains and families databases, sequence analysis tools, scientific literature and sequence similarity search tools.
  112. [112]
    AlphaFold Protein Structure Database
    AlphaFold DB provides open access to over 200 million protein structure predictions to accelerate scientific research.AlphaFold · Downloads · View protein · FAQs