Fact-checked by Grok 2 weeks ago

Degron

A degron is a short linear , , or within a protein that acts as a recognition signal targeting the protein for , primarily through the ubiquitin-proteasome system () where E3 ubiquitin ligases bind to initiate and subsequent proteasomal breakdown. These degrons confer metabolic instability to proteins, enabling precise control over their half-life and abundance in response to cellular needs. Degrons are classified into several types based on their location and activation mechanism, including N-degrons at the protein's amino terminus (governed by the pathway, where specific N-terminal residues like or destabilize the protein), C-degrons at the carboxyl terminus (such as those forming cyclic imides recognized by specific ligases, e.g., ), and internal degrons embedded within the protein sequence (often conditional motifs like phosphodegrons activated by ). -independent degrons also exist, allowing direct proteasomal targeting without ubiquitination, as seen in proteins like . The encodes approximately 600 ligases that recognize diverse degrons, with recent high-throughput identifying thousands of such motifs across the . In cellular biology, degrons are essential for maintaining protein (proteostasis), quality control of misfolded or damaged proteins, and dynamic signaling pathways, such as cell cycle regulation (e.g., cyclin degradation via the destruction box degron) and stress responses (e.g., hypoxia-inducible factor targeting by VHL ligase). Dysregulation of degron-mediated degradation contributes to diseases, including cancer (where mutations stabilize oncoproteins like β-catenin via phosphodegron disruption) and neurodegeneration (e.g., accumulation of tau fragments due to impaired N-degron recognition). Therapeutically, degrons inspire targeted protein degradation strategies using bifunctional molecules like PROTACs, which hijack E3 ligases to eliminate disease-related proteins. The concept of degrons originated in the , with seminal work by Bachmair et al. demonstrating that N-terminal residues dictate protein stability in , laying the foundation for understanding ubiquitin-dependent turnover. Ongoing continues to refine degron "rules," revealing their roles in evolutionary adaptation and potential as biomarkers for proteostasis disorders.

Definition and Overview

Definition

A degron is a short sequence or within a protein that serves as a recognition signal for targeted , marking the protein as unstable and directing it toward proteolytic destruction. These motifs typically consist of 2 to 15 residues, though their length can vary depending on the type and location within the protein. Unlike other protein motifs that may facilitate localization, enzymatic activation, or protein-protein interactions, degrons are specifically evolved to promote degradation rather than other regulatory functions. The primary role of degrons is to mediate by recruiting components of cellular degradation machinery, most notably the -proteasome system (), where they enable the attachment of ubiquitin chains that tag proteins for proteasomal breakdown. This process is essential for maintaining , responding to cellular signals, and eliminating misfolded or damaged proteins. Representative examples of degrons include single N-terminal destabilizing residues, such as , which initiate under the pathway, as demonstrated in early studies on protein regulation. Another classic instance is the internal PEST motif, characterized by regions enriched in (P), (E), serine (S), and (T), which correlates with rapid in short-lived regulatory proteins.

Historical Discovery

The concept of degrons, short for degradation signals that determine protein stability, was first introduced in the 1980s by Alexander Varshavsky, who linked the N-terminal residues of proteins to their through the foundational . The term "degron" was coined by Varshavsky in 1991. This rule posits that the identity of a protein's N-terminal influences its rate by the ubiquitin-proteasome system. A pivotal demonstration came in 1986, when Varshavsky and colleagues published findings showing that specific N-terminal exert destabilizing effects on proteins in , using the ubiquitin fusion technique to engineer reporter proteins. In these experiments, chimeric genes encoding fused to β-galactosidase variants with altered N-termini were expressed in , revealing that residues like conferred short half-lives (approximately 2 minutes), while others like led to stability (over 20 hours). This work established N-degrons as the first characterized class of degrons and highlighted their role in selective . In the 1990s and 2000s, research expanded to reveal additional degron types beyond N-terminal ones, driven by investigations into cell cycle regulators and transcription factors. Studies on mitotic cyclins identified internal degrons, such as the destruction box (D-box) motif, essential for their timely degradation during anaphase. For instance, fusion of the cyclin B D-box to stable reporter proteins accelerated their ubiquitination and proteasomal degradation. Similarly, analyses of transcription factors, including those involved in stress responses, uncovered internal degrons that modulate their activity through rapid turnover, often tested via domain swaps in fusion constructs. These advancements, building on Varshavsky's fusion protein approaches, underscored degrons' versatility in regulating diverse cellular processes.

Types of Degrons

N-Degrons

N-degrons are degradation signals located at the of proteins, which become exposed following the co-translational or post-translational cleavage of the initial N-terminal by methionine aminopeptidases. These signals determine the of the protein through targeted ubiquitination and subsequent , primarily via the ubiquitin-proteasome system (UPS). The pathway encompasses these N-degrons, establishing a hierarchical classification of N-terminal residues that dictate protein stability, where the identity of the residue directly correlates with the rate of . In eukaryotes, the N-end rule pathway categorizes N-terminal residues into destabilizing and stabilizing groups. Primary destabilizing residues include the basic amino acids arginine (Arg), lysine (Lys), and histidine (His), which confer short half-lives (typically 2-30 minutes in yeast and mammalian cells) by serving as direct ligands for recognition components. In contrast, residues such as alanine (Ala), serine (Ser), and threonine (Thr) are stabilizing, resulting in longer half-lives (over 10 hours) unless modified. Secondary destabilizing residues, such as the large hydrophobic amino acids phenylalanine (Phe), leucine (Leu), isoleucine (Ile), tyrosine (Tyr), and tryptophan (Trp), require prior conjugation with Arg or other basic residues via arginyltransferase (ATE1) or leucyl/phenylalanyl-tRNA-protein transferase (ATE2) to become functional N-degrons. Tertiary destabilizing residues like asparagine (Asn), glutamine (Gln), aspartic acid (Asp), and glutamic acid (Glu) are converted to primary forms through deamidation or transamidation before recognition. Recognition of N-degrons occurs through ubiquitin ligases known as N-recognins, such as UBR1 in mammals and , which bind the destabilizing N-terminal residue via a conserved UBR box and internal residues on the substrate, facilitating K48-linked polyubiquitination for proteasomal degradation. UBR1 exhibits specificity for type 1 (, Lys, His) and type 2 (Phe, Leu, etc.) residues, with the adjoining sequence context influencing binding affinity. A prominent example is the hypoxia-inducible factor 1α (HIF1α), where oxygen-dependent prolyl by prolyl hydroxylase enzymes exposes the N-terminus to arginylation by ATE1, generating an N-Arg degron that targets HIF1α for UBR-mediated degradation under normoxic conditions, thereby enabling oxygen sensing independent of the canonical von Hippel-Lindau pathway. Variants of N-degrons include extensions beyond the first residue, such as secondary destabilizing motifs or unstructured regions that enhance accessibility to N-recognins, thereby modulating ; for instance, the presence of proximal lysines serves as auxiliary ubiquitination sites. Additionally, N-terminal can convert stabilizing residues into Ac/N-degrons recognized by distinct ligases like Doa10/TEB4, further diversifying the pathway. These elements ensure fine-tuned regulation of in response to cellular cues.

C-Degrons

C-degrons are degradation signals located at the of proteins, where the primary determinants of recognition are the C-terminal residues themselves, often in the form of short motifs such as glycine-rich sequences (e.g., -, -) or dipeptides that mediate ubiquitin-dependent proteasomal degradation. Unlike more structured rules, C-degrons exhibit contextual flexibility, with stability influenced by surrounding sequences, , and post-translational modifications like , which can either expose or mask the motif. These motifs are recognized by specific substrate receptors within cullin-RING ligases (CRLs), including members of the CRL2 and CRL1 families, which facilitate polyubiquitination and subsequent targeting to the . For instance, glycine-ended C-degrons, such as the diglycine (-GG) motif, are bound by the KLHDC2 receptor in the CRL2 complex, enabling precise recognition of the terminal residues through a conserved pocket that accommodates the penultimate . Similarly, phosphorylated C-terminal degrons, like the Cdc4-phosphodegron (CPD) centered on 380 in human cyclin E, are targeted by the Fbw7 receptor in the SCF^{Fbw7} (CRL1) complex, where dual by GSK-3 and CDK2 creates a high-affinity involving the and a +4 residue. In biological contexts, C-degrons play key roles in regulating during cellular processes such as the . A prominent example is the C-terminal CPD in cyclin E, which ensures timely degradation after S-phase entry; phosphorylation at T380 promotes Fbw7 binding, reducing cyclin E to approximately 5 hours and preventing aberrant accumulation that could disrupt . Mutations ablating this phosphorylation, such as T380A, extend to over 20 hours, leading to hyperproliferation in hematopoietic and epithelial cells. Other C-degrons, like the glycine-ended motif in the tumor suppressor p14ARF, contribute to and stoichiometric balance in protein complexes, indirectly supporting fidelity by eliminating aberrant isoforms. The stability conferred by C-degrons lacks the rigid hierarchy of the pathway, where degradation rates strictly follow N-terminal residue identity; instead, C-degron efficiency varies with exposure and modifications, allowing transient shielding by protein interactions or folding to modulate half-lives dynamically. often amplifies this by enhancing ligase affinity, as seen in cyclin E, where it shifts the degron from inert to highly destabilizing, thereby fine-tuning regulatory responses without a fixed scale. This adaptability enables C-degrons to integrate environmental cues, such as activity during transitions, into protein pathways. C-degrons can also mediate ubiquitin-independent degradation, where the C-terminal sequence directly engages the without prior ubiquitination. For example, (ODC) features a C-terminal that binds the 26S , leading to its rapid turnover independent of the system. Additionally, C-terminal residues capable of forming cyclic imides, such as aspartimide or glutarimide, serve as degrons recognized by the core, facilitating the clearance of damaged or aged proteins. These mechanisms expand the role of C-degrons in beyond E3 ligase-dependent pathways.

Internal Degrons

Internal degrons are short linear motifs embedded within the protein sequence, distinct from terminal positions, that function as degradation signals by being recognized by components of the ubiquitin-proteasome system or machinery. These motifs must be solvent-accessible to enable efficient substrate targeting, and they often reside in unstructured or flexible regions of the protein interior. A classic example of an internal degron is the sequence, characterized by regions enriched in (P), (E), serine (S), and (T), flanked by basic residues. In the c-Myc oncoprotein, the domain drives rapid turnover with a of about 20 minutes in non-transformed cells, ensuring tight regulation of its transcriptional activity. The destruction box (D-box), another key internal degron, features the RxxLxxxxN and is prevalent in mitotic cyclins such as . This is specifically recognized by the anaphase-promoting complex/cyclosome (APC/C) E3 , facilitating ubiquitination and proteasomal degradation to coordinate exit. In the context of , KFERQ-like pentapeptide s act as internal degrons for , where they are bound by the heat shock cognate 71 kDa protein (HSC70) chaperone to direct substrate translocation across the lysosomal membrane. Internal degrons display considerable diversity, including both constitutive forms that inherently promote degradation and conditional ones whose activity is modulated by post-translational modifications. , for example, can activate PEST-containing degrons by enhancing their affinity for ligases, thereby triggering ation.

Mechanisms of Action

Recognition by Cellular Machinery

Degrons serve as specific recognition signals that enable ubiquitin ligases to selectively bind target proteins, initiating the process of ubiquitination. In the case of N-degrons, the UBR family of ligases, such as UBR1 and UBR2, directly interact with destabilizing N-terminal residues through their UBR box domains, which contain binding pockets tailored to hydrophobic or basic side chains of the exposed . For C-degrons, ligases like those in the KLHDC family (e.g., KLHDC2 and KLHDC3) recognize terminal motifs preceded by specific penultimate residues, utilizing Kelch domains to engage these sequences with high specificity. Similarly, the von Hippel-Lindau (VHL) ligase complex identifies hydroxylated proline-containing motifs in substrates like hypoxia-inducible factor 1α (HIF1α), binding via a surface groove on the VHL protein that accommodates the modified degron. Upon recognition, the bound facilitates the , a sequential enzymatic process that tags the for . is first activated by an E1 enzyme through ATP-dependent adenylation, forming a bond, and then transferred to an E2 conjugating enzyme. The then promotes the transfer of from E2 to a residue on the (or to the previous ), assembling polyubiquitin chains predominantly linked at 48 (K48-linked), which serve as the signal for proteasomal targeting. This ensures specificity, as the not only recognizes the degron but also positions the optimally for efficient chain formation. The structural basis of degron-ligase interactions often involves complementary binding s on the E3 ligase that engage the degron's key residues. For instance, in UBR ligases, a hydrophobic within the UBR box accommodates bulky side chains of N-terminal leucines or phenylalanines, stabilized by bonds to the backbone. In KLHDC ligases, β-propeller structures feature arginine-serine-arginine (RSR) motifs that anchor the C-terminal , while adjacent hydrophobic or charged s select for the penultimate residue, enabling combinatorial specificity. These interactions are typically low-affinity but sufficient for transient binding during ubiquitination. Regulatory post-translational modifications can dynamically expose or conceal degrons, modulating recognition. near a degron site may create a negative charge that enhances binding to positively charged pockets in the E3 ligase, as seen in some internal degrons, or alternatively mask the by steric hindrance. Oxidation, particularly of N-terminal cysteines in the N-degron pathway, converts the residue to cysteic acid, exposing it for recognition by downstream components like ATE1 arginyltransferase, which further modifies it for UBR binding. These modifications allow cells to fine-tune protein stability in response to signals like oxygen levels or activity.

Protein Degradation Pathways

The primary pathway for degron-mediated protein degradation is the ubiquitin-proteasome system (UPS), where proteins tagged with polyubiquitin chains following degron recognition are targeted for breakdown by the 26S proteasome. The 26S proteasome, composed of a 20S catalytic core and 19S regulatory caps, binds the polyubiquitin chain via its regulatory particles, which employ ATP-dependent unfoldases (such as Rpt subunits) to thread the substrate into the core for proteolytic cleavage into short peptides of approximately 7-10 residues. This process ensures the complete dismantling of the protein, preventing accumulation of potentially harmful fragments. Certain degrons also enable ubiquitin-independent proteasomal degradation (UbInPD), in which the 26S directly recognizes and degrades unstructured substrates without ubiquitination. For example, (ODC) is targeted via a C-terminal α-helix degron, whose to the is enhanced by the cofactor antizyme (AZ). Recent have revealed UbInPD as more widespread, particularly for C-degrons recognized by CRL2^KLHDC2, contributing to control. After proteasomal degradation, deubiquitinating enzymes (DUBs) associated with the 19S regulatory particle disassemble the poly chain, recycling free monomers for further conjugation reactions and maintaining cellular pools. The resulting peptides are released into the , where a subset is transported into the by the complex for loading onto () class I molecules, facilitating to cytotoxic T cells. The rate of in the is closely tied to degron strength, which influences the of ubiquitination and proteasomal recognition; stronger degrons accelerate these steps, yielding protein half-lives from as short as a few minutes (e.g., for N-terminal arginine-bearing substrates) to several hours, depending on the specific residue or and cellular . pathways handle certain internal degrons outside the , notably lysosomal through . () targets proteins containing the KFERQ-like , an internal degron recognized by the cytosolic chaperone HSC70, which escorts the to the lysosomal receptor LAMP2A for unfolding, translocation into the , and by resident proteases. This pathway provides a selective, non-ubiquitin-dependent route for degrading soluble cytosolic proteins, particularly under stress or proteotoxic conditions.

Identification Methods

Experimental Techniques

Reporter assays are a cornerstone for experimentally validating degron function by assessing protein half-life. In these assays, a suspected degron sequence is fused to the N- or of a stable reporter protein, such as (GFP), and the resulting fusion protein's stability is monitored in cells. Protein synthesis is then inhibited using , a elongation inhibitor, allowing researchers to track the rate of the reporter via Western blotting or fluorescence quantification over time. This chase method directly measures the degron's impact on protein turnover, with unstable fusions indicating functional degrons. For instance, studies have used this approach to confirm ΦN peptides as degrons by observing their destabilization of GFP reporters in mammalian cells. Similarly, light-inducible systems incorporating degron reporters have quantified changes under controlled conditions, revealing degron sensitivity to environmental cues. Mutagenesis studies provide targeted evidence for degron motifs by altering specific residues and evaluating consequent changes in protein stability. is employed to introduce point mutations, deletions, or substitutions within the putative degron of a protein, often fused to a reporter like GFP or . Stability is then assessed through chase assays or pulse-chase experiments, where mutations that extend suggest critical residues for degron recognition. This technique has been instrumental in dissecting N-degron pathways, such as modifying N-terminal residues in bacterial proteins to map stability determinants. In eukaryotic systems, combinatorial of N-terminal sequences has identified degron variants that modulate recognition by ligases, confirming sequence-specific degradation signals. Proteomics approaches enable high-throughput identification of potential degrons through the mapping of ubiquitination sites, which often overlap with degron motifs. remnant involves tryptic digestion of cell lysates to generate diglycine (K-ε-GG) remnants on modified lysines, followed by enrichment using specific that recognize this . Immunoaffinity purification with anti-K-ε-GG , combined with , quantifies thousands of ubiquitination sites across the , highlighting lysines proximal to known or novel degrons. This method has been refined for routine detection of approximately 20,000 endogenous sites in a single experiment, providing context for degron validation by revealing ubiquitination patterns in pathways. Specialized toolkits extend this to N-terminal ubiquitination, aiding in the of non-canonical degrons. In vivo validation techniques confirm direct interactions between degrons and E3 ubiquitin ligases, essential for establishing functional relevance. Yeast two-hybrid (Y2H) assays screen for binding by fusing the degron-containing to a DNA-binding domain bait and the E3 ligase to an domain prey, with strength gauged by on selective media. This has identified degron-E3 pairs in the GID/CTLH , demonstrating specificity for N-degron motifs. Co-immunoprecipitation (co-IP) complements Y2H by pulling down endogenous or overexpressed E3 ligases with epitope-tagged degron substrates from cell lysates, followed by Western blotting to detect associations. For example, co-IP has verified C-degron recognition by TRIM7, where mutations disrupting the abolish ubiquitination. These orthogonal methods together substantiate degron-mediated targeting in cellular contexts.

Computational Prediction

Computational prediction of degrons involves bioinformatics tools and algorithms that identify potential degradation motifs in protein sequences and structures, enabling proteome-wide screening without experimental intervention. These methods typically leverage scanning, models trained on experimental datasets, and structural modeling to pinpoint degron candidates, particularly those recognized by ubiquitin ligases. Sequence-based tools facilitate the detection of known degron motifs, including those governed by the pathway, by aligning query sequences against curated databases of degradation signals. For instance, UbiBrowser 2.0 employs a computational framework integrating protein motifs, domains, annotations, and to predict ligase-substrate interactions, which encompass N-degron recognition across eukaryotic proteomes. Similarly, DEGRONOPEDIA scans over 1800 curated motifs for N-terminal, C-terminal, and internal degrons, incorporating evolutionary conservation and proteolysis simulations to contextualize predictions. Another example is Degpred, a deep learning tool that uses embeddings to scan sequences for degron-like patterns beyond rigid motifs, achieving an of 0.8807 on test sets derived from experimental degron annotations. Machine learning approaches enhance prediction accuracy by training on large-scale proteomics data, such as ubiquitination site annotations from databases like dbPTM, to forecast sites where degrons initiate proteasomal targeting. Support vector machines (SVMs), often combined with physicochemical properties and position-specific scoring matrices, have been applied in tools like UbiSitePred, yielding accuracies up to 98.33% on benchmark datasets. Deep learning models, including convolutional neural networks (CNNs) and long short-term memory (LSTM) networks, outperform traditional methods by capturing contextual sequence features; for example, DeepUbi uses CNNs trained on ubiquitinated protein data to predict sites with superior sensitivity over random forest baselines. These models typically process flanking residues (e.g., window sizes of 45–77 amino acids) to account for local context, with LSTM variants achieving macro-F1 scores around 0.574 on human datasets. Structural prediction integrates tools like to evaluate the accessibility and conformational features of candidate degrons, as solvent-exposed or disordered regions are more likely to interact with recognition machinery. DEGRONOPEDIA employs and RoseTTAFold models to predict intrinsic disorder (via pLDDT scores) and relative solvent accessibility, mapping degrons to nearby ubiquitination-prone residues for refined scoring. In quality control degron studies, structures have revealed hydrophobic, alpha-helical motifs in solvent-accessible positions, such as in the Pca1 protein, where pLDDT confidence scores (70–90) corroborated experimental degron potency. This structural layer helps prioritize predictions by assessing burial or exposure, which influences degron functionality . Despite advances, computational methods suffer from high false positive rates due to the short, degenerate nature of degron motifs and their strong context-dependency, such as reliance on post-translational modifications or cellular conditions that sequence-only models overlook. For example, Degpred's predictions carry a of approximately 0.512 at stringent cutoffs, partly because it cannot differentiate modification-dependent degrons without integrated data. Tools like DEGRONOPEDIA mitigate these issues by incorporating multi-omics features (e.g., secondary and ), but broader limitations persist in capturing dynamic protein interactions or species-specific variations, often necessitating experimental validation.

Biological Significance

Regulatory Roles

Degrons play a crucial role in maintaining by marking misfolded, damaged, or excess proteins for ubiquitin-mediated degradation, thereby preventing their accumulation and ensuring cellular . In the N-degron pathway, destabilizing N-terminal residues serve as primary degrons that are recognized by ligases, initiating the degradation of aberrant proteins through the ubiquitin-proteasome system. Similarly, at the protein function as degradation signals that promote the timely clearance of unnecessary or faulty polypeptides, contributing to overall cellular . These mechanisms are essential for balancing protein synthesis and turnover across diverse organisms. In , conditional degrons enable rapid responses to environmental stimuli, such as , by controlling the stability of key regulatory proteins. For instance, in the NF-κB pathway, of IκB inhibitors by exposes cryptic degrons, leading to their ubiquitination and proteasomal degradation, which liberates for nuclear translocation and activation of proinflammatory gene expression. This phosphorylation-dependent mechanism acts as a switch, allowing degrons to become functional only upon specific signals like cytokines or pathogen-associated molecules, thus fine-tuning inflammatory responses. Degrons are integral to control, where they ensure precise timing of protein degradation to facilitate progression through checkpoints. The destruction box (D-box) degron, characterized by the RXXLXXXXN, is recognized by the -promoting complex/cyclosome (/C) in complex with Cdc20 or Cdh1, targeting substrates like securin and cyclins for ubiquitination. Degradation of securin releases separase to cleave , enabling sister chromatid separation during , while degradation inactivates Cdk1 to exit , preventing premature advancement and maintaining genomic stability. During development, degrons regulate the spatiotemporal activity of to establish patterns. In , the Cubitus interruptus (), a homolog in the Hedgehog signaling pathway, contains multiple Ser/Thr-rich degrons that are phosphorylated by kinases like CK1 and GSK3, promoting its partial proteolysis into a form or full degradation by SCF Slimb . This degron-mediated processing restricts Ci activator function to Hedgehog-responsive regions, delineating anterior-posterior patterns in the wing and embryonic segments essential for proper organ formation.

Implications in Disease and Therapy

Mutations in degrons or their recognition sites can stabilize oncoproteins, contributing to cancer progression. For instance, in colorectal and other cancers, mutations in the β-catenin (CTNNB1) gene often disrupt its phospho-degron, which is activated by phosphorylation at residues S33, S37, T41, and S45, preventing ubiquitination by β-TrCP and subsequent proteasomal degradation. This stabilization leads to aberrant Wnt signaling, promoting cell proliferation and tumor growth. Similarly, truncating mutations in genes like GATA3 (in breast cancer) and PPM1D eliminate C-terminal degrons, enhancing protein stability and dysregulating pathways such as p53 signaling. Across 33 cancer types in TCGA data, over 19% of driver genes involve UPS alterations, with mutations enriched in degron motifs identified by deep learning models like DeepDegron. In neurodegenerative diseases, impaired recognition of degrons by the contributes to . In , mutant forms of α-synuclein (e.g., A30P, A53T) exhibit defective binding to receptors like LAMP-2A via their KFERQ motif, leading to UPS overload and formation. This accumulation disrupts , exacerbating neuronal toxicity, as soluble α-synuclein is normally ubiquitinated (e.g., via Lys48 linkages) and degraded by the . Such dysregulation extends the normal regulatory roles of degrons in , resulting in pathological aggregates. Therapeutic strategies exploit degrons through PROTACs (proteolysis-targeting chimeras), which recruit ligases to induce ubiquitination and of disease-related proteins. PROTACs consist of a target-binding , linker, and recruiter (e.g., or VHL ligands), hijacking the to tag proteins with polyubiquitin chains for proteasomal elimination, enabling of otherwise "undruggable" targets. This approach has shown promise in by degrading stabilized oncoproteins, such as degraders in . In the 2020s, PROTAC-based drugs have advanced to clinical trials for . Although ARV-110 targeting the achieved a 46% PSA50 response rate in a mutation-specific subset of phase II trials for metastatic , its development was discontinued in 2023 due to limited broader efficacy. Vepdegestrant (formerly ARV-471), an degrader, demonstrated a 40% clinical benefit rate in phase I for , progressed to phase III by 2024, and as of May 2025 showed 51% CBR in pretreated ESR1-mutant cases in the VERITAC-2 trial. BTK degraders like NX-2127 induced over 80% target degradation in patients in ongoing phase 1 trials as of 2025, following resumption after a 2024 FDA hold.

References

  1. [1]
    Degrons: defining the rules of protein degradation - Nature
    Jul 14, 2025 · Degrons are pivotal components of the ubiquitin–proteasome system, serving as the recognition determinants through which E3 ubiquitin ...
  2. [2]
    Tripartite degrons confer diversity and specificity on regulated ...
    Jan 6, 2016 · A degron has been defined as a protein element that confers metabolic instability. Although seemingly straightforward, its molecular correlates ...
  3. [3]
    Degrons in cancer - PubMed
    Mar 14, 2017 · Degrons are the elements that are used by E3 ubiquitin ligases to target proteins for degradation. Most degrons are short linear motifs ...
  4. [4]
    N-degron and C-degron pathways of protein degradation - PMC - NIH
    Jan 8, 2019 · N-degrons and C-degrons are degradation signals whose main determinants are, respectively, the N-terminal and C-terminal residues of cellular proteins.<|control11|><|separator|>
  5. [5]
    Tying up loose ends: the N-degron and C-degron pathways of ...
    Jul 6, 2020 · Regulated protein degradation by the ubiquitin-proteasome system (UPS) plays a critical role in essentially all major cellular processes.
  6. [6]
    In Vivo Half-Life of a Protein Is a Function of Its Amino-Terminal ...
    In Vivo Half-Life of a Protein Is a Function of Its Amino-Terminal Residue. Andreas Bachmair, Daniel Finley, and Alexander VarshavskyAuthors Info & Affiliations.
  7. [7]
    Cyclin is degraded by the ubiquitin pathway - Nature
    Jan 10, 1991 · Cyclin is degraded by ubiquitin-dependent proteolysis. Thus anaphase may be triggered by the recognition of cyclin by the ubiquitin-conjugating system.
  8. [8]
    N-degron and C-degron pathways of protein degradation - PNAS
    Jan 7, 2019 · N-degrons and C-degrons are degradation signals whose main determinants are, respectively, the N-terminal and C-terminal residues of cellular proteins.
  9. [9]
    The N-End Rule Pathway - PMC - NIH
    The N-end rule pathway is a proteolytic system in which N-terminal residues of short-lived proteins are recognized by recognition components (N-recognins)
  10. [10]
    The evolutionarily conserved arginyltransferase 1 mediates a pVHL ...
    We report that ATE1 centrally controls the hypoxic response and glycolysis in mammalian cells by preferentially arginylating HIF1α that is hydroxylated by PHD ...
  11. [11]
  12. [12]
    Cyclin E phosphorylation regulates cell proliferation in ...
    Phosphorylations within N- and C-terminal degrons independently control the binding of cyclin E to the SCFFbw7 and thus its ubiquitination and proteasomal ...
  13. [13]
    Elucidation of E3 Ubiquitin Ligase Specificity Through Proteome ...
    We took a two-pronged approach to identify sequence specific degrons. First, we predicted peptides whose stabilities were composition-dependent. Secondly, we ...
  14. [14]
    A modular degron library for synthetic circuits in mammalian cells
    May 1, 2019 · Other examples of degrons include short intrinsic amino acid sequences, such as the D-element, the PEST sequence, unstructured initiation sites, ...
  15. [15]
    MYC Degradation - PMC - NIH
    Finally, MYC protein is rapidly degraded following its synthesis (half-life of ∼20 min in non-transformed cells) (Hann and Eisenman 1984). One of the most ...
  16. [16]
    Substrate Recognition by the Cdh1 Destruction Box Receptor Is a ...
    The destruction box (D-box), originally identified in sea urchin cyclin B and subsequently found in many other APC/C substrates, has the consensus sequence ...Missing: internal | Show results with:internal
  17. [17]
    The coming of age of chaperone-mediated autophagy - PMC
    The KFERQ-like motif provided the 'bait' to identify HSC70 as the cytosolic chaperone that, upon binding that region, targeted proteins for lysosomal ...Missing: internal degron
  18. [18]
    Protein degradation on the global scale - ScienceDirect
    Apr 21, 2022 · In many instances, phosphorylation of PEST sequence regions precedes ubiquitylation and degradation, exemplifying a so-called phosphodegron.
  19. [19]
    Signaling Pathways Regulated by UBR Box-Containing E3 Ligases
    Aug 3, 2021 · UBR box E3 ligases, also called N-recognins, are integral components of the N-degron pathway. Representative N-recognins include UBR1, UBR2, ...
  20. [20]
    Structural basis for C-degron selectivity across KLHDCX family E3 ...
    Nov 15, 2024 · The data reveal combinatorial mechanisms determining specificity and plasticity of substrate recognition by KLDCX-family C-degron E3 ligases.
  21. [21]
    Targeted Protein Degradation: Principles and Applications of ... - NIH
    Jul 13, 2023 · The ubiquitin mediates the K48-linked ubiquitination of substrates through a cascade involving E1, E2, and E3 enzymes. Ubiquitin chains are ...
  22. [22]
    Control of protein stability by post-translational modifications - Nature
    Jan 13, 2023 · Degrons as multicomponent control modules. Degrons were originally defined as protein motifs causing “metabolic instability of some or all of ...
  23. [23]
    The Ubiquitin-Proteasome Proteolytic Pathway: Destruction for the ...
    A Comprehensive Review of the Ubiquitin-Proteasome System in Synapse Remodeling and Neurodegenerative Diseases. 13 July 2025. Proteostasis Disturbances and ...
  24. [24]
    Antigen Presentation and the Ubiquitin‐Proteasome System in Host ...
    Antigen presentation involves the conversion of protein antigens into peptide ligands that can bind to MHC products that are displayed at the cell surface for ...
  25. [25]
    Degradation signal diversity in the ubiquitin-proteasome system - PMC
    In this review, we will focus on how different physiological requirements for the degradation of specific proteins dictate the design of different degrons.Missing: paper | Show results with:paper
  26. [26]
    Chaperone‐mediated autophagy: Molecular mechanisms, biological ...
    Aug 30, 2023 · Target proteins that are degraded via CMA must contain the KFERQ motif, which enables the high selectivity of CMA among the three autophagy ...
  27. [27]
    DEGRONOPEDIA: a web server for proteome-wide inspection of ...
    Apr 3, 2024 · As degron sites may undergo various PTMs, with the primary role of phosphorylation, which can modulate their exposure (62), the server reports ...
  28. [28]
    Systematic prediction of degrons and E3 ubiquitin ligase binding via ...
    Jul 14, 2022 · Degpred provides an efficient tool to proteome-wide prediction of degrons and binding E3s singly from protein sequences.
  29. [29]
    Machine learning-based approaches for ubiquitination site ...
    Nov 28, 2023 · Our work highlights the potential of ML, particularly DL techniques, in predicting Ubi-sites and furthering our knowledge of protein regulation through ...
  30. [30]
    Conserved degronome features governing quality control ... - Nature
    Dec 8, 2022 · Ubiquitin-conjugation necessitates the recognition of degradation determinants, termed degrons, by their cognate E3 ubiquitin-protein ligases.
  31. [31]
    Conserved degronome features governing quality control ... - NIH
    Dec 8, 2022 · The authors identify conserved features of proteome-derived degradation signals, including amino acid and structural preferences, that trigger quality-control- ...
  32. [32]
    Ubiquitination and Degradation of the Inhibitors of NF-κB - PMC
    Ubiquitin-dependent degradation of IκB activates NF-κB. A single E3 ubiquitin ligase, β-TrCP, targets several IκB proteins for degradation. Retrospectively, one ...
  33. [33]
    The IκB kinase complex in NF-κB regulation and beyond - PMC
    IKK activation results in phosphorylation of IκB and p65. IκB phosphorylation leads to its ubiquitin-mediated proteasomal degradation, enabling NF-κB dimers ...
  34. [34]
    Structural analysis of human Cdc20 supports multisite degron ...
    For example, cyclin B1 contains a destruction (D) box with the consensus of RXXLX4–5N (X, any residue), whereas securin contains both a D box and a KEN box (4-6) ...<|control11|><|separator|>
  35. [35]
    Cubitus interruptus - Society for Developmental Biology
    Jul 25, 2022 · Hh stimulates Ci phosphorylation by CK1 at multiple Ser/Thr-rich degrons to inhibit its recognition by the Hh-induced MATH and BTB domain ...
  36. [36]
    Degradation of misfolded proteins in neurodegenerative diseases
    Mar 13, 2015 · The UCH-L1 gene encodes two opposing enzymatic activities that affect alpha-synuclein degradation and Parkinson's disease susceptibility.
  37. [37]
  38. [38]
    PROTAC targeted protein degraders: the past is prologue - Nature
    Jan 18, 2022 · In this Review, we summarize the first two decades of PROTAC discovery and assess the current landscape, with a focus on industry activity.
  39. [39]
    Targeted protein degradation: advances in drug discovery and ...
    Nov 6, 2024 · Among the leading innovations in TPD are PROTACs and MGs, which promote protein degradation via the UPS, a pathway that tags proteins for ...<|separator|>