Fact-checked by Grok 2 weeks ago

Proliferating cell nuclear antigen

Proliferating cell nuclear antigen (PCNA) is a highly conserved eukaryotic protein that functions as a sliding clamp, forming a homotrimeric ring-shaped structure to encircle double-stranded DNA and enhance the processivity of DNA polymerases δ and ε during replication. This doughnut-like architecture, with pseudo-six-fold symmetry and an internal diameter of approximately 3.4 nm, allows PCNA to tether polymerases to the DNA template, enabling efficient and continuous nucleotide incorporation while minimizing dissociation events. Originally identified in 1978 as an autoantigen in patients with systemic lupus erythematosus, PCNA was later recognized for its association with cell proliferation and DNA synthesis, earning its name due to elevated expression in cycling cells. Beyond replication, PCNA plays multifaceted roles in maintaining genome integrity by participating in various DNA repair pathways, including base excision repair (BER), nucleotide excision repair (NER), and mismatch repair (MMR), where it coordinates the recruitment of repair enzymes through specific protein-protein interactions. These interactions are primarily mediated by the inter-domain connecting loop (IDCL) on PCNA's surface and motifs such as the PCNA-interacting protein (PIP) box (consensus sequence Q-x-x-ψ-x-x-ϑ-ϑ, where ψ is hydrophobic and ϑ is aromatic) on partner proteins like DNA ligase I, flap endonuclease 1 (FEN1), and replication factor C (RFC). Additionally, PCNA regulates cell cycle progression by binding to cyclin-dependent kinase inhibitors such as p21^WAF1/CIP1, which inhibits DNA synthesis and promotes G1/S checkpoint control to prevent replication in damaged cells. Post-translational modifications, including monoubiquitination at lysine 164 for error-prone translesion synthesis and SUMOylation for suppressing inappropriate recombination, further modulate PCNA's functions in response to genotoxic stress. In clinical contexts, PCNA serves as a reliable biomarker for cellular proliferation and is overexpressed in numerous cancers, correlating with poor prognosis, advanced tumor stages, and resistance to therapies due to its central role in rapid DNA replication and repair in malignant cells. Mutations or dysregulation of PCNA have been linked to genomic instability and hereditary disorders, such as a rare DNA repair deficiency syndrome, underscoring its importance in human health. Ongoing research explores PCNA as a therapeutic target, with small-molecule inhibitors disrupting its interactions to selectively impair cancer cell proliferation while sparing normal cells.

Structure and Properties

Molecular Architecture

Proliferating cell nuclear antigen (PCNA) is a homotrimeric protein that assembles into a toroidal ring structure, enabling it to encircle double-stranded DNA and facilitate processive enzymatic activities. Each monomer consists of 261 amino acids and adopts a globular fold resembling a right-handed α/β horseshoe, with the three identical subunits arranged in a head-to-tail manner to form the closed ring. The overall structure exhibits pseudo-sixfold rotational symmetry due to the similar topologies of the domains within each monomer, contributing to the ring's stability and uniform interaction surfaces. The central cavity of the PCNA ring has an inner diameter of approximately 3.5 nm, sufficient to accommodate double-stranded DNA, while the outer diameter measures about 10-11 nm, providing a broad platform for protein binding. Each monomer is divided into an N-terminal domain (residues 1-118), a C-terminal domain (residues 164-256), and an interdomain connecting loop (IDCL, residues 119-132) that links the two domains. The N- and C-terminal domains share structural homology, each featuring a six-stranded β-sheet flanked by α-helices, which together generate the pseudo-sixfold symmetry in the trimer. PCNA exhibits high biophysical stability as a trimer, with a dissociation constant in the nanomolar range, ensuring it remains intact during cellular processes. Once loaded onto DNA, the ring slides along the double helix in an ATPase-independent manner, driven by thermal motion and constrained by electrostatic interactions with the DNA backbone. Loading of the topologically closed ring onto DNA requires the pentameric replication factor C (RFC) clamp loader, which uses ATP hydrolysis to open the ring at an intersubunit interface and assemble it around primer-template junctions. PCNA is evolutionarily conserved across eukaryotes, reflecting its essential role in DNA metabolism, with sequence identity exceeding 30% between human and yeast orthologs. In humans, the PCNA gene is located on chromosome 20p12.2 and encodes the 28.6 kDa protein. Structural insights derive primarily from X-ray crystallography, including high-resolution structures of human PCNA at 2.3 Å (PDB: 2ZVM), which reveal atomic details of the intersubunit interfaces. Recent cryo-EM studies, including a 2020 structure of yeast PCNA in complex with DNA and polymerase δ at 3.2 Å resolution (PDB: 7KC0), have provided dynamic views of DNA-bound conformations, highlighting subtle ring distortions upon DNA engagement.

Post-Translational Modifications

Proliferating cell nuclear antigen (PCNA) undergoes a variety of post-translational modifications (PTMs) that dynamically regulate its function as a during and maintenance. These modifications, including ubiquitination, SUMOylation, , , and , occur primarily on specific and serine residues, altering PCNA's stability, localization, and interactions without disrupting its core homotrimeric ring structure. Such PTMs enable PCNA to adapt to cellular stresses, ensuring efficient processivity along DNA strands. A key site for modification is lysine 164 (K164), located on the interdomain connecting loop (IDCL) of PCNA, which serves as a hub for ubiquitin-based signaling. Monoubiquitination at K164 is catalyzed by the E2-E3 complex RAD6-RAD18 in response to DNA damage, adding a single ubiquitin moiety that enhances PCNA's role in damage tolerance. Polyubiquitination at the same site, involving K63-linked chains extended by the Ubc13-Mms2 heterodimer and the E3 ligase Rad5 (or mammalian homologs HLTF/SHPRH), further diversifies PCNA's regulatory landscape. These ubiquitination events are reversible, mediated by deubiquitinases such as USP1, USP7, and USP10, allowing PCNA to return to its unmodified state after stress resolution. SUMOylation also targets K164, primarily through the E3 ligase MMS21 (a subunit of the SMC5/6 complex), which conjugates small ubiquitin-like modifier (SUMO) proteins to PCNA, promoting its association with chromatin. This modification exhibits competitive crosstalk with ubiquitination, as both utilize the same lysine residue, enabling pathway-specific choices in cellular responses; it is reversible via SUMO-specific proteases. A minor SUMOylation site exists at K127, but K164 predominates in regulatory functions. SUMOylated PCNA influences sliding clamp dynamics by modulating its loading and unloading efficiency on DNA. Acetylation occurs at multiple lysine residues, including K13, K14, K77, and K80 (catalyzed by CBP/p300 in humans) and K20 (by Eco1 in yeast), often on the inner or sliding surfaces of the PCNA ring. These acetyl groups neutralize positive charges, affecting PCNA's electrostatic interactions with DNA and promoting its degradation or stability as needed for cell cycle progression. Acetylation is reversible through histone deacetylases (HDACs), contributing to the fine-tuned control of PCNA turnover. Phosphorylation at serine 261 (S261) introduces a negative charge that enhances PCNA's sliding clamp efficiency by facilitating its interaction with the replication machinery and improving loading. This modification is reversible by protein phosphatases, allowing temporal regulation aligned with dynamics. Methylation patterns include monomethylation at K248 by SETD8 and at K110 by (in humans, absent in ), which stabilize the PCNA trimer and enhance its structural integrity for processivity. These methyl marks are generally stable but can influence subsequent PTMs, impacting overall clamp function. The reversibility of these PTMs—through dedicated enzymes like deubiquitinases, desumoylases, deacetylases, and phosphatases—ensures PCNA's adaptability, preventing prolonged activation that could lead to genomic instability. Collectively, they modulate sliding clamp efficiency by altering charge distribution and conformational flexibility, optimizing PCNA's translocation along DNA without compromising its core encircling mechanism. Experimental evidence from , particularly K164R variants, demonstrates that ablating these modifications impairs PCNA's damage-responsive functions while preserving basal replication fidelity, as shown in and mammalian cell models.

Biological Functions

Role in DNA Replication

Proliferating cell nuclear antigen (PCNA) functions as a sliding clamp during eukaryotic DNA replication, encircling double-stranded DNA and tethering replicative DNA polymerases δ (pol δ) and ε (pol ε) to the template strand. This association enables the polymerases to remain bound to DNA for extended periods, increasing their processivity from approximately 10 nucleotides without PCNA to over 5,000 nucleotides with the clamp present. The ring-shaped architecture of PCNA allows it to slide freely along the DNA helix without rotating around the double helix, facilitating continuous synthesis.90014-0) The assembly of PCNA onto DNA occurs at primer-template junctions through an ATP-dependent process mediated by replication factor C (RFC), a heteropentameric clamp loader complex composed of five subunits (RFC1–5). RFC recognizes the 3' primer terminus, hydrolyzes ATP to transiently open the PCNA trimer, and loads the open clamp onto the DNA, which then closes upon ATP hydrolysis to encircle the duplex. Once loaded, PCNA coordinates with the pol δ holoenzyme—comprising the polymerase, accessory subunits, and PCNA—for both leading and lagging strand synthesis, with pol δ primarily extending on the lagging strand and pol ε synthesizing the leading strand continuously. On the lagging strand, PCNA plays a key role in Okazaki fragment processing by recruiting flap endonuclease 1 (Fen1) via PCNA-interacting protein (PIP) motifs to cleave RNA-DNA flaps generated during strand displacement by pol δ, and by subsequently binding DNA ligase I to seal the resulting nicks and join fragments. This coordinated handoff ensures efficient maturation of discontinuous fragments into continuous DNA. PCNA expression and nuclear accumulation are tightly regulated, peaking during S-phase when replication is active, localizing to replication foci within the nucleus. In vitro replication assays using reconstituted systems, such as those with DNA or primed templates, have shown that omitting or depleting PCNA severely impairs the elongation phase, resulting in short DNA products and halted synthesis, while addition of PCNA restores processive replication. These experiments confirm PCNA's indispensable role in maintaining replication fidelity and efficiency.

Role in DNA Repair

Proliferating cell nuclear antigen (PCNA) participates in (NER) by serving as a scaffold that interacts with key proteins such as XPA, activates the endonuclease XPF, targets XPG for degradation, and recruits δ for resynthesis. In (BER), particularly the long-patch subpathway, PCNA enhances processivity of δ/ε and facilitates flap endonuclease 1 (FEN-1)-mediated excision and I sealing of the repair patch. Similarly, in mismatch repair (MMR), PCNA directs strand-specific repair by binding MSH2-MSH6 and MLH1-PMS2 complexes to recognize mismatches and coordinates excision and resynthesis steps preceding DNA gap filling. A central role of PCNA in involves translesion synthesis (TLS), where monoubiquitination at 164 enables recruitment of Y-family DNA polymerases, such as polymerase η (pol η), to bypass replication-blocking lesions while maintaining fork progression. This modification, briefly referenced in post-translational contexts, switches PCNA from high-fidelity replication to error-prone bypass, allowing specialized polymerases like pol η and Rev1 to insert nucleotides opposite damaged bases. In post-replication repair, ubiquitinated PCNA further supports gap-filling downstream of stalled forks by facilitating TLS or template switching to resolve single-stranded DNA gaps left behind during replication of damaged templates. PCNA provides scaffold support in double-strand break (DSB) repair pathways, notably (HR), where it is loaded onto structures to promote invasion and extension of the 3' invading strand by DNA polymerase δ. This loading ensures efficient recombination-associated DNA synthesis, contrasting with its more limited involvement in (NHEJ), where PCNA may indirectly stabilize repair complexes at breaks. Evidence from model systems underscores these distinct functions: in , pol30 mutants (e.g., pol30-79) exhibit proficient but defective UV-induced repair, highlighting PCNA's specialized repair interactions. Human hypomorphic PCNA mutations similarly cause disorders with preserved replication capacity, manifesting as premature aging and genomic instability. PCNA is integrated into interstrand crosslink () repair through interactions with the () pathway, where ubiquitinated PCNA facilitates FANCD2 monoubiquitination and recruitment to stalled forks, enabling TLS-mediated bypass and error-free resolution. For instance, RAD18-mediated PCNA ubiquitination activates the FA-BRCA pathway, coordinating incisions and access during unhooking. Additionally, PCNA K164 ubiquitination supports FANCD2-dependent mitotic () to fill under-replicated regions, preventing chromosomal instability in FA-deficient cells.

Additional Cellular Roles

PCNA plays a pivotal role in by recruiting the assembly factor 1 (CAF-1) to facilitate assembly on newly synthesized DNA strands during replication. This interaction occurs through direct binding between the PCNA sliding clamp and the p150 subunit of CAF-1, ensuring the faithful deposition of histones H3 and H4 to maintain structure post-replication. Such is essential for restoring epigenetic marks and preventing genomic instability, as disruptions in this process lead to defects in chromatin organization. Beyond replication, PCNA contributes to transcriptional processes by interacting with (RNAPII), modulating conflicts between transcription and replication machinery. Specifically, PCNA associates with the RPB1 subunit of RNAPII, which can influence the progression of transcription complexes and resolve transcription-replication collisions that arise during S phase.00221-0) This interaction helps in dissociating stalled RNAPII from under stress conditions, thereby preventing fork stalling. In epigenetic regulation, SUMOylated PCNA promotes modifications that contribute to . SUMO2 conjugation on PCNA enhances CAF-1-mediated deposition at common fragile sites, leading to increased levels of repressive marks such as and H3K27me3. This modification facilitates compaction and heritable of heterochromatic regions, linking replication-coupled events to long-term epigenetic memory.00540-0) PCNA also participates in cell cycle checkpoint activation by sensing replication stress through the ATR signaling pathway. Upon replication fork stalling, monoubiquitinated PCNA serves as a platform for ATR-mediated phosphorylation events, which activate downstream effectors like CHK1 to halt cell cycle progression and allow fork restart. This mechanism ensures genome stability by coordinating DNA repair with checkpoint enforcement during genotoxic insults. In non-proliferative contexts, PCNA is expressed and functional in quiescent cells to support maintenance . In growth-arrested fibroblasts, PCNA forms complexes at sites of oxidative damage, facilitating without stimulating proliferation. This role underscores PCNA's versatility in preserving genomic integrity during periods of cellular dormancy. Recent advances have revealed PCNA's involvement in mitochondrial DNA maintenance and cellular senescence. In 2024 studies, mitochondrial-localized PCNA was shown to protect yeast mtDNA from UV-induced mutagenesis by aiding in damage tolerance pathways, independent of nuclear replication functions. Additionally, reduced PCNA levels in senescent cells serve as a marker of irreversible growth arrest, with replication stress-induced PCNA modifications contributing to senescence onset through persistent DNA damage signaling.00640-8)

Regulation and Expression

Cell Cycle Dynamics

Proliferating cell nuclear antigen (PCNA) exhibits dynamic localization and expression patterns tightly synchronized with the , ensuring its availability for primarily during . In quiescent or G0/G1 cells, PCNA is predominantly cytoplasmic or diffusely distributed in the at low levels. As cells approach the , PCNA undergoes nuclear accumulation facilitated by nonclassical nuclear import mechanisms, including direct interaction with importin-β or piggybacking on partners containing nuclear localization signals (NLS), such as p21. This relocation prepares PCNA for loading onto at replication origins by the replication factor C () clamp loader. During S phase, PCNA expression and chromatin association reach their peak, forming punctate nuclear foci visible via immunofluorescence microscopy that correspond to active replication sites. These foci reflect PCNA's role as a sliding clamp encircling DNA, enhancing polymerase processivity. Protein levels increase modestly due to elevated synthesis rates peaking in early S phase, while the protein itself remains relatively stable with a constant low degradation rate. Synchronization experiments using aphidicolin, which inhibits DNA polymerase α and arrests cells in early S phase, demonstrate that PCNA loading onto chromatin is blocked, preventing focus formation and confirming S-phase dependency. As cells progress beyond S phase, PCNA levels decline following the cessation of synthesis, with chromatin unloading mediated by factors like Elg1/ATAD5 to prevent aberrant retention. In , breakdown leads to PCNA exclusion from the , dispersing it into the ; re-import occurs upon in the subsequent . This cyclic exclusion ensures PCNA is not inappropriately engaged during segregation. Dysregulation of these dynamics, particularly PCNA overexpression, is associated with uncontrolled in cancer, where elevated levels sustain replication even outside , disrupting growth control and promoting tumorigenesis. Transcription of PCNA is briefly upregulated by factors at the G1/S boundary to support this timely accumulation.

Transcriptional and Epigenetic Control

The promoter of the PCNA gene contains multiple -binding sites that are critical for its transcriptional activation during the G1/ transition of the . These sites allow transcription factors to bind and recruit the transcriptional machinery, ensuring elevated PCNA expression coincides with the onset of . In contrast, under cellular stress conditions, the tumor suppressor represses PCNA transcription by binding to specific elements in the promoter, thereby limiting proliferation in response to DNA damage or developmental cues. This repression helps maintain cellular quiescence or induce arrest, preventing inappropriate replication. Enhancers and silencers within the PCNA promoter region further modulate its activity through interactions with transcription factors such as AP-1 and Sp1. AP-1 binding sites confer serum responsiveness, enabling rapid upregulation of PCNA in response to growth signals by facilitating the recruitment of co-activators. Similarly, Sp1 sites in the proximal promoter act as positive regulatory elements, promoting basal and induced transcription by interacting with the general transcription apparatus and stabilizing the pre-initiation complex. These factors collectively fine-tune PCNA expression to match proliferative demands, with mutations or dysregulation often observed in proliferative disorders. Epigenetic modifications provide an additional layer of control over PCNA . acetylation at the promoter, including marks like H3K9ac, correlates with transcriptional activation by opening structure and facilitating access to and other activators during active . In quiescent cells, at CpG sites within the PCNA promoter contributes to , maintaining low expression levels until growth stimuli trigger demethylation and reactivation. This pattern is particularly evident in non-proliferating states, such as serum-starved hepatocytes, where hypomethylation accompanies re-entry into the . PCNA itself participates in feedback loops that influence its regulatory landscape via chromatin assembly. Through direct interaction with chromatin assembly factor 1 (CAF-1), PCNA deposits histones onto newly replicated DNA, ensuring faithful propagation of epigenetic marks that can affect the expression of its own upstream regulators, such as E2F or p53 target genes. Disruptions in this process, as seen in PCNA mutants, lead to altered histone modifications and derepression of silenced loci, highlighting PCNA's role in epigenetic homeostasis. These mechanisms are evolutionarily conserved, with the homolog POL30 exhibiting similar transcriptional control. The POL30 promoter is bound by stress-responsive factors like Sfp1p and Msn2p, as well as regulators Fkh1p and Fkh2p, mirroring the - and stress-mediated in mammals. Recent studies from 2023 onward have elucidated additional of PCNA in cancer, including miRNA-mediated control.

Protein Interactions

Binding Mechanisms

Proliferating cell nuclear antigen (PCNA) engages partner proteins through specific molecular interfaces on its homotrimeric structure, enabling coordinated DNA transactions. These interactions primarily occur via short linear motifs on partner proteins that insert into hydrophobic pockets on PCNA's surface, with the interdomain connector loop (IDCL) serving as a key docking site for many ligands. The canonical PCNA-interacting protein (PIP) box, characterized by the QxLxΦΦ (where x denotes any residue and Φ is a hydrophobic or aromatic residue), is a primary for replication-associated proteins. This binds to a hydrophobic pocket on the IDCL of PCNA, forming a 3₁₀ that inserts into the pocket, stabilized by bonds and van der Waals interactions involving and residues of the PIP box with PCNA residues such as Ile128 and Met129. Structural studies, including structures (e.g., PDB: 1AXC for PCNA-p21 ), reveal that the PIP box buries approximately 800–1000 Ų of surface area, facilitating high-affinity engagement. In contrast, the AlkB homologue 2 PCNA-interacting motif (APIM), with the consensus sequence (K/R)-(F/Y/W)-(L/I/V/A)-(L/I/V/A)-(K/R), predominates in DNA repair and stress response proteins. APIM binds to the same hydrophobic pocket as the PIP box but with distinct orientation, leveraging charged residues for additional electrostatic interactions that enhance specificity in non-replicative contexts. Crystal structures confirm this overlap, showing APIM's central hydrophobic di-leucine inserting similarly to the PIP box's ΦΦ, yet with broader tolerance. Beyond these motifs, PCNA utilizes alternative interfaces for specific partners. For flap endonuclease 1 (FEN1), binding involves a C-terminal extension forming a βA-αA-βB motif, where residues 326–337 of FEN1 adopt a short α-helix (αA, 339RLDDFF344) that docks into PCNA's interdomain helical loop pocket, complemented by β-sheet interactions between FEN1's βA strand and PCNA's C-terminus (Ala252–Ile255). This interface, resolved in crystal structures (PDB: 1UL1), buries ~2500 Ų and supports dual binding modes modulated by DNA presence. For replication factor C (RFC), interactions occur via PCNA's outer surface acidic patches, which engage basic regions of RFC subunits, as inferred from clamp-loading complexes where electrostatic complementarity facilitates ring opening. Binding affinities for these interfaces typically range from 10–100 nM, reflecting physiological relevance; for instance, the PIP box of p21WAF1 yields a K_d of ~80 nM, while FEN1's interaction is weaker at ~60 μM in solution but enhanced to sub-micromolar levels on DNA. DNA binding to PCNA induces allosteric changes, such as IDCL repositioning, that modulate pocket accessibility and affinity for motifs. Cryo-EM structures of PCNA-polymerase complexes (e.g., resolutions 3.1–4.0 Å) and NMR studies of solution dynamics illustrate motif insertion into the pocket, with DNA occupancy rigidifying the ring for stable ligation. These binding pockets exhibit remarkable evolutionary conservation, with hydrophobic residues (e.g., Ile128, Met129 in humans) preserved across eukaryotes from to mammals, underscoring PCNA's ancient role in genome maintenance. Comparative analyses reveal minimal in pocket architecture, enabling motif compatibility across species.

Major Interacting Partners

Proliferating cell nuclear antigen (PCNA) serves as a central hub for protein interactions, with over 500 known binding partners documented in databases such as BioGRID, enabling its diverse roles in DNA metabolism. These interactions primarily occur through the interdomain connecting loop (IDCL) of PCNA, often mediated by PCNA-interacting protein () or AlkB homolog 2 PCNA-interacting (APIM) sequences in partner proteins, allowing dynamic exchange of partners to switch cellular functions. Evidence for these associations has been established through methods like co-immunoprecipitation (co-IP) and , which have identified both direct and context-dependent bindings. In DNA replication, PCNA interacts with key components of the replisome to enhance processivity and fidelity. DNA polymerase δ (Pol δ) and DNA polymerase ε (Pol ε) bind PCNA via PIP boxes on their accessory subunits, enabling the synthesis of leading and lagging strands, respectively, as demonstrated in biochemical assays and structural studies. Replication factor C (RFC) loads PCNA onto DNA as the clamp loader, with direct binding confirmed through peptide mapping and co-IP experiments showing regions of PCNA (amino acids 36-55 and 196-215) critical for this interaction. Replication protein A (RPA) associates with PCNA at replication forks, stabilizing single-stranded DNA and facilitating primer-template junctions, as observed in ensemble assays monitoring RPA-PCNA dynamics during replication stress. For DNA repair pathways, PCNA recruits specialized enzymes to sites of damage. η (Pol η), a translesion synthesis , binds PCNA to bypass UV-induced lesions, with this interaction enhanced by post-translational modifications like ubiquitination, as shown in functional reconstitution experiments. Flap endonuclease 1 (FEN1) interacts with PCNA to process and repair intermediates, inhibiting excessive in coordinated assays. I (LIG1) binds PCNA to seal nicks post-repair, with co-IP confirming this association and revealing mutual inhibition to regulate ligation timing. In , PCNA engages Rad51 recombinase on intermediates, promoting extension while Rad51 can inhibit PCNA loading to prevent premature replication. PCNA also coordinates with chromatin factors for epigenetic maintenance. Chromatin assembly factor 1 (CAF-1) binds PCNA via a PIP box on its p150 subunit, depositing histones H3-H4 onto newly synthesized DNA during replication-coupled assembly, as evidenced by in vitro binding and cellular colocalization studies. Histone deacetylase 1 (HDAC1) associates with PCNA to modulate chromatin remodeling and gene silencing, with co-IP and colocalization in nuclear foci demonstrating their interaction in human cells. Regulatory proteins fine-tune PCNA activity across the . The inhibitor p21 binds PCNA with high affinity (Kd ≈ 0.08 μM) via its C-terminal PIP box, inhibiting replication and repair to enforce , as structurally resolved in the p21-PCNA complex. , particularly cyclin A, form ternary complexes with PCNA and CDK2, facilitating phosphorylation of replication factors and linking replication to progression, confirmed through co-IP of endogenous proteins. This partner diversity allows PCNA to orchestrate functional switches, such as from replication to repair, through competitive binding and post-translational regulation.

Clinical and Research Applications

Diagnostic Applications

Proliferating cell nuclear antigen (PCNA) is widely utilized as a biomarker for assessing cell proliferation in diagnostic pathology, primarily through immunohistochemistry (IHC) employing monoclonal antibodies such as PC10 or 19A2 to quantify the proliferation index in tumor specimens. The PCNA labeling index (LI), calculated as the percentage of nuclei exhibiting positive staining relative to total tumor cells, provides a measure of proliferative activity and correlates strongly with Ki-67 expression, another established proliferation marker, enabling evaluation of tumor growth rates and aggressiveness. In clinical practice, this approach helps distinguish malignant from benign lesions, as malignant tumors consistently show higher PCNA LIs; for example, poorly differentiated prostate carcinomas exhibit mean LIs of 7.6%, compared to 1.2% in benign prostatic hyperplasia. In specific applications, PCNA IHC aids tumor grading and staging, particularly in (CNS) neoplasms like astrocytomas, where LI values escalate with histological grade—for instance, averaging 4.1% in (WHO) grade II, 8.1% in grade III, and 26.1% in grade IV tumors—supporting prognostic assessments and recurrence risk evaluation alongside molecular markers in WHO classifications. Similarly, in , elevated PCNA LIs are linked to adverse outcomes, with studies demonstrating its utility in stratifying minimal and invasive carcinomas for prognostic purposes, often integrated into multiparametric panels with hormone receptors. These findings underscore PCNA's role in refining tumor grading, though it complements rather than replaces emphasized in the 2021 WHO CNS tumor classification. Beyond tissue-based IHC, enables quantitative detection of PCNA in dissociated tumor cells, offering a complementary method to assess S-phase fractions and , particularly in hematological malignancies or fresh solid tumor samples. For non-invasive monitoring, emerging applications include analysis of PCNA in liquid biopsies; recent studies have identified exosomal PCNA mRNA in as a prognostic for early detection, with models combining it with genes like CDK1 and FEN1 achieving high diagnostic accuracy. While serum PCNA protein levels via have been investigated for therapy response tracking in select cancers, their clinical adoption remains limited due to variability in circulating forms. A key limitation of PCNA IHC is specificity, as cytoplasmic localization can occur, potentially overestimating if not distinguished; is thus prioritized for accurate interpretation of active cell cycling. Despite these challenges, PCNA's integration into routine diagnostics enhances precision in -based staging, with ongoing refinements in 2023–2024 studies validating its prognostic value in colorectal and esophageal cancers through combined panels.

Therapeutic Potential

Proliferating cell nuclear antigen (PCNA) has emerged as a promising therapeutic target in cancer due to its overexpression in many tumor types, which sustains aberrant and mechanisms essential for cancer survival. Inhibiting PCNA disrupts and repair processes, leading to accumulation of DNA damage and selective death, as normal cells with lower PCNA dependency are less affected. This rationale is supported by preclinical evidence showing that PCNA inhibition induces in cancers reliant on high replication stress, such as those with defective . Key PCNA inhibitors include AOH1996, a small-molecule agent that binds to the interdomain connecting loop (IDCL) of PCNA, preventing its loading onto DNA and blocking interactions with replication and repair proteins. AOH1996 has demonstrated antitumor activity in preclinical models. It entered phase I clinical trials for advanced solid tumors in 2023, with ongoing studies as of 2025 reporting tolerability and early efficacy signals, such as tumor reduction in a pancreatic cancer patient. Another inhibitor, T2AA, disrupts PCNA's interactions with PIP-box-containing proteins like DNA polymerase η, impairing translesion synthesis and increasing double-strand breaks without affecting PCNA monoubiquitination. T2AA has shown promise in preclinical settings by sensitizing cells to DNA-damaging agents. Combination therapies exploit PCNA's role in repair to enhance the efficacy of standard treatments, achieving . For instance, PCNA inhibitors like PCNA-I1S augment the of (e.g., ) and by preventing repair of induced DNA lesions, as demonstrated in models. Similarly, ATX-101, a targeting PCNA, potentiates radiotherapy in xenografts by increasing DNA fragmentation and . However, a major challenge is achieving specificity to cancer cells, as PCNA is essential for normal proliferating tissues, potentially causing toxicity; strategies focus on cancer-specific PCNA conformations to mitigate this. Preclinical studies in 2024 have highlighted efficacy in specific cancers: in prostate cancer, PCNA inhibitors enhanced ionizing radiation sensitivity by impairing DNA repair, reducing tumor growth in xenografts. For lung cancer, AOH1996 suppressed proliferation and metastasis in cell lines and mouse models, outperforming controls. Emerging developments in 2025 include PCNA inhibitors in immunotherapy combinations; AOH1996 synergizes with anti-PD-1 antibodies in squamous cell carcinoma by eliminating cancer stem cells and boosting immune infiltration. Additionally, research on the Srs2-PCNA axis in checkpoint modulation suggests potential for targeting PCNA to downregulate DNA damage responses, enhancing therapy in replication-stressed tumors, though translation from yeast models to human applications remains investigational.