Fact-checked by Grok 2 weeks ago

p21

p21, also known as cyclin-dependent kinase inhibitor 1A (CDKN1A) or WAF1/CIP1, is a protein encoded by the CDKN1A gene located on chromosome 6p21.2 in humans. This 164-amino-acid protein functions primarily as a potent of (CDK) activity, binding to and suppressing cyclin-CDK complexes such as CDK2, CDK1, CDK4, and CDK6 to prevent of key substrates and thereby block progression, especially at the G1/S and G2/M checkpoints. Through these mechanisms, p21 mediates cellular responses to stress, including DNA damage, by inducing cell cycle arrest, , and . The expression of p21 is tightly regulated, most notably as a downstream target of the tumor suppressor , where it links DNA damage detection to arrest to allow for repair or . Beyond p53-dependent pathways, p21 can be activated by various stressors through p53-independent mechanisms, such as transforming growth factor-beta (TGF-β) signaling or MAPK pathways, highlighting its versatile role in maintaining genomic stability. Additionally, p21 interacts with (PCNA) to modulate and repair during the , further underscoring its multifaceted involvement in cellular . In the context of cancer, predominantly acts as a tumor suppressor by inhibiting uncontrolled in response to oncogenic , with its or dysregulation—often concomitant with mutations—promoting tumorigenesis in various malignancies, including , , and colon cancers. However, paradoxical oncogenic functions have been observed, particularly in p53-mutant backgrounds, where cytoplasmic localization of p21 can enhance cell motility, survival, and chemoresistance, contributing to cancer progression and in tumors like and endometrial carcinomas. These dual roles emphasize p21's context-dependent effects, making it a key and therapeutic target in research.

Molecular Biology

Gene and Expression

The CDKN1A gene, which encodes the p21 protein, is located on the short arm of human chromosome 6 at position 6p21.2 and spans approximately 10.8 kb of genomic DNA. The canonical transcript consists of three exons, producing a mature mRNA that translates into the p21 protein. Alternative splicing generates multiple transcripts, though the predominant isoform is the full-length p21^Cip1/Waf1. The protein product of CDKN1A is a 164-amino-acid polypeptide with a calculated molecular weight of approximately 18 kDa, consistent with its common designation as p21. It is also known by the aliases CIP1, WAF1, and SDI1, reflecting its identification in early studies as a inhibitor, a wild-type p53-activated fragment, and a senescence-associated protein, respectively. Rare splice variants of CDKN1A mRNA have been reported, such as a variant that influences translation efficiency in response to , but these are not the primary forms expressed in most cellular contexts. CDKN1A exhibits basal expression across a variety of tissues, including epithelial cells, fibroblasts, and hematopoietic cells, where low levels support normal cellular such as maintenance. Expression is inducible under stress conditions, leading to elevated p21 levels that contribute to adaptive responses like temporary arrest, though specific induction mechanisms are multifaceted. The CDKN1A gene and its p21 product demonstrate strong evolutionary conservation among mammals, with particularly high in the N-terminal cyclin- domain, which is essential for its inhibitory function. This conservation extends to vertebrates, underscoring the domain's critical role in core cellular processes.

Protein

The p21 protein is characterized by its intrinsically nature, lacking a in isolation and instead adopting extended, flexible conformations that enable dynamic interactions with partners. This is particularly pronounced in the linker regions between functional domains, allowing p21 to undergo induced folding upon complex formation and facilitating its role as a versatile regulator. Structural studies, including () , confirm that p21 remains largely unstructured in solution, with transient secondary elements that provide conformational flexibility essential for multi-site . The N-terminal domain of p21, encompassing residues 1-56, forms the primary -binding region critical for interaction with cyclin-dependent kinases (CDKs), such as CDK2. This domain includes conserved motifs like the Cy (RXL) sequence that mediate high-affinity binding to , positioning p21 to sterically hinder CDK substrate access in complexes. Crystal structures of CDK2- in complex with p21-derived peptides highlight how this N-terminal segment inserts into the groove, stabilizing the inhibitory conformation without resolving the full-length disordered protein. In the C-terminal region, p21 features a PCNA-binding motif spanning residues 141-160, which includes the conserved QLSLT hydrophobic that docks into a pocket on the interdomain connecting loop of (PCNA). The of this complexed with human PCNA (PDB: 1AXC) reveals a β-sheet augmentation mechanism, where the p21 peptide extends the PCNA β-sheet, enhancing specificity and affinity for and repair factors. p21 harbors multiple post-translational modification sites that influence its structural dynamics, including lysine residues such as within the PCNA-binding , which is targeted for ubiquitination, and various threonine and serine residues (e.g., , ) prone to . These sites are embedded in the disordered regions, allowing solvent accessibility for enzymatic modification without disrupting core interactions. Recent studies have revealed that p21's cysteines and serve as switches, forming bonds in response to (ROS) to fine-tune its interactions and cellular responses, a mechanism conserved in the CIP/KIP family. Under standard physiological conditions, p21 adopts a monomeric structure without stable intramolecular bonds. However, its two cysteine residues ( and ) can form transient bonds in response to , enabling sensing and modulation of its inhibitory function. The protein's stability is modulated by environmental factors, including and ; at neutral and moderate salt concentrations, p21 maintains its disordered state, but extremes can induce aggregation or partial folding. Compared to its CIP/KIP family members p27 and p57, p21 shares the N-terminal CDK-inhibitory domain but stands out with its robust C-terminal PCNA-binding motif, which is conserved in p57 but absent or weaker in p27, enabling p21's unique dual inhibition of cell cycle progression and DNA synthesis.

Functions

Cell Cycle Regulation

p21, encoded by the CDKN1A gene, serves as a critical cyclin-dependent kinase inhibitor (CKI) that enforces cell cycle checkpoints, primarily at the G1/S and G2/M transitions, by suppressing the activity of cyclin-CDK complexes. This inhibition prevents the phosphorylation of downstream targets essential for cell cycle progression, allowing time for damage assessment and repair. Specifically, p21 binds stoichiometrically to complexes such as cyclin E/A-CDK2 (key for G1/S), cyclin B-CDK1 (for G2/M), and cyclin D-CDK4/6 (for early G1), with dissociation constants (K_i) in the low nanomolar range (0.5–15 nM for CDK2, CDK4, and CDK6). By doing so, p21 maintains the retinoblastoma protein (Rb) in a hypophosphorylated state, which binds and represses E2F transcription factors, thereby blocking the expression of S-phase genes like CCNE1 and MCM family members. This mechanism enforces G1 arrest, as demonstrated by early overexpression experiments in human tumor cell lines (e.g., Saos-2 osteosarcoma cells), where p21 induction led to accumulation in G0/G1 and suppression of DNA synthesis. The regulatory impact of p21 on CDK activity is dose-dependent, exhibiting a biphasic effect that fine-tunes dynamics. At low stoichiometric levels (below one p21 molecule per CDK complex), p21 facilitates the formation and localization of active cyclin-CDK assemblies, particularly for D-CDK4/6, promoting initial G1 progression. However, at higher concentrations (exceeding one p21 per complex), p21 acts as a potent by occupying the cyclin-CDK , blocking access and ATP binding, which halts events. This threshold behavior underpins a bistable switch in fate decisions through double-negative loops involving p21 and CDK2: CDK2 phosphorylates p21 for its degradation, while p21 inhibits CDK2, creating ultrasensitive transitions between and states in response to signals. Such dynamics ensure robust checkpoint enforcement, as modeled in single-cell analyses where variable p21 levels dictate population heterogeneity in cycling versus quiescent s. In senescence-associated cell cycle arrest, p21 integrates with the Rb pathway to drive irreversible withdrawal from the . Sustained elevation of p21, often in response to oncogenic or replicative stress, persistently inhibits CDK4/6 and CDK2, leading to stable Rb hypophosphorylation and long-term E2F repression. This enforces a permanent G1-like state, distinct from transient quiescence, and is evident in models of oncogene-induced senescence where p21 knockout impairs Rb-mediated . The inhibitory kinetics of p21 on CDK activity can be conceptually represented by the equation [\text{CDK}_{\text{active}}] = \frac{[\text{CDK}_{\text{total}}]}{1 + \frac{[\text{p21}]}{K_i}} where K_i is the inhibition constant, illustrating how escalating p21 concentrations suppress active CDK pools to threshold levels required for arrest. This simple model aligns with biochemical assays showing near-complete CDK2 inhibition at p21 levels above 10 nM.

DNA Repair and Replication

p21 interacts directly with the (PCNA) trimer via its C-terminal PCNA-interacting protein (PIP) box motif, binding to the interdomain connector loop on the front face of PCNA. This interaction inhibits the activity of replicative DNA polymerases such as polymerase δ (pol δ) and polymerase ε (pol ε), which rely on PCNA for processive synthesis during S-phase replication, thereby halting unscheduled in response to . In contrast, p21 does not inhibit PCNA-dependent stimulation of repair polymerases like polymerase β (pol β), allowing short-patch to proceed without interference. Through this selective PCNA modulation, p21 facilitates (NER) and (BER) by enabling lesion bypass and gap-filling without disrupting overall repair fidelity. In NER, p21-PCNA binding permits the recruitment of repair factors to UV-induced lesions, supporting post-incision synthesis while preventing replication fork progression across damaged sites. Similarly, in BER, p21 allows pol β to perform accurate repair of oxidative or alkylated bases, maintaining genomic integrity during transient stress without inducing prolonged arrest. During replication stress, p21 suppresses excessive replication origin firing to avert replication catastrophe, coordinating with ATR and kinase pathways to enforce intra-S-phase checkpoints. Low levels of p21 in S phase limit origin activation by modulating PCNA availability for replicative complexes, thereby slowing fork progression and stabilizing stalled forks under genotoxic conditions. This mechanism integrates p21's PCNA inhibition with broader checkpoint signaling, distinct from its CDK inhibition role in enforcing G1/S arrest. Studies in p21-null (p21-/-) cells demonstrate hyper-replication, with increased origin firing and elevated replication stress markers leading to genomic instability, such as double-strand breaks and chromosomal aberrations. For instance, p21-deficient fibroblasts exhibit deregulated replication licensing and heightened sensitivity to DNA-damaging agents, underscoring p21's essential role in preventing catastrophe during unperturbed and stressed replication. The selective inhibition model relies on p21's high-affinity binding to PCNA (Kd ≈ 2.5 nM), which competitively displaces proliferation-specific partners like pol δ while preserving interactions with repair factors due to differential binding stoichiometries and site overlaps on the PCNA ring. This affinity ensures transient modulation of replication without compromising repair efficiency, highlighting p21's role as a tunable guardian of DNA transactions.

Apoptosis and Senescence

p21 exerts an anti-apoptotic function primarily in p53-wildtype cells by inhibiting caspase activation and pro-apoptotic proteins, often through suppression of (CDK) activity under cellular stress conditions such as DNA damage or irradiation. Cytoplasmic localization of p21 enables direct binding and inhibition of caspases like pro-, , and caspase-10, thereby blocking effector pathways of and promoting cell survival during repair processes. This protective role is evident in models of lung injury and , where p21 knockdown exacerbates , highlighting its contribution to tissue resilience. Recent studies further indicate that p21 acts as a dual regulator in DNA damage responses, promoting at high damage levels while preventing at low doses by maintaining checkpoint integrity. In contrast, sustained p21 expression drives , a permanent cell cycle arrest state characterized by the (SASP), which involves secretion of inflammatory cytokines and . p21 induces by inhibiting CDK complexes, preventing (Rb) hyperphosphorylation and maintaining Rb-E2F repression of proliferation genes, leading to irreversible G1/S arrest in response to persistent stressors like telomere attrition or oncogene activation. Through Rb-dependent , p21 promotes SASP components such as CXCL14, facilitating immune surveillance of senescent cells while contributing to tissue remodeling. Recent findings (as of 2024) also show p21 regulates expression of components during , influencing tissue stiffness and aging-related pathologies. This mechanism is central to developmentally programmed and replicative limits, as seen in primary fibroblasts. The role of p21 in apoptosis exhibits context-dependent duality: in p53-mutant backgrounds, prolonged p21-mediated arrest can lead to mitotic catastrophe, a form of cell death arising from aberrant mitosis due to unresolved DNA damage and failed checkpoint enforcement. Recent studies underscore p21's involvement in therapy-induced senescence (TIS), where chemotherapeutic agents trigger p21 upregulation to enforce dormancy in cancer cells, potentially delaying recurrence but risking therapy resistance. In breast cancer models, TIS via CDKN1A contributes to tumor dormancy hallmarks, balancing short-term growth suppression against long-term proliferative escape. Furthermore, p21 links senescence to organ regeneration; knockout of p21 in mice bypasses senescence barriers, enabling enhanced wound healing, such as full regeneration of ear punches, by sustaining stem cell proliferation without exhaustion.

Regulation

Transcriptional Regulation

The promoter of the CDKN1A gene, encoding , features a complex regulatory region spanning approximately 2.4 kb upstream of the transcription start site, containing multiple cis-acting elements that facilitate both basal and inducible transcription. Central to this structure are two p53 response elements, located between approximately -2.3 kb and -1.4 kb upstream, which enable -dependent activation in response to genotoxic stress. Additionally, six Sp1/Sp3 binding sites (GC boxes 1-6) within the proximal promoter region (-119 to -50 bp) support constitutive expression and cooperate with for enhanced activation. An AP-1 motif further contributes to basal activity and stress-induced responses, particularly under oxidative conditions. Transcriptional induction of p21 is prominently triggered by cellular stresses such as DNA damage, oxidative stress, and hypoxia, which activate diverse signaling pathways converging on the promoter. DNA damage primarily engages the p53 pathway but also involves ATM/ATR-mediated phosphorylation events that enhance transcription factor recruitment. Oxidative stress, via reactive oxygen species, activates MAPK cascades (e.g., JNK and p38) that phosphorylate and stabilize transcription factors binding to the proximal promoter, leading to rapid p21 upregulation. Hypoxia-inducible factor 1α (HIF-1α) similarly binds upstream elements under low oxygen, while TGF-β signaling recruits Smad complexes to Sp1 sites for sustained induction in epithelial cells. Beyond p53-dependent mechanisms, several non-canonical pathways regulate p21 transcription to ensure context-specific control. directly transactivates the p21 promoter through binding to specific motifs and recruitment of coactivators like CARM1 and p300, independent of , particularly in contexts. (PML) promotes p21 expression via p53-independent stabilization of transcription factors and in response to . MicroRNAs, such as miR-93, suppress p21 levels post-transcriptionally by targeting the 3' UTR, thereby indirectly modulating transcriptional feedback loops, while tissue-specific enhancers, including those involving lincRNA-p21 in cis, fine-tune expression in contexts like . Dynamic expression patterns of p21 are evident in its pulsatile response to DNA damage, driven by oscillatory p53 levels that generate rhythmic transcription pulses in single cells. During recovery from genotoxic stress, p53 pulses (with periods of ~5-6 hours) lead to counter-oscillatory p21 mRNA accumulation, allowing temporal coordination of cell cycle arrest and repair without permanent senescence. This behavior, observed in real-time imaging studies, ensures reversible inhibition of proliferation. Epigenetic modifications at the p21 promoter critically govern its activation or silencing. Histone , particularly H3K9ac, marks active and facilitates access; for instance, PCAF-mediated H3K9 acetylation at the proximal promoter enhances p21 expression in response to signals. Conversely, promoter hypermethylation, often coupled with repressive marks, silences p21 in various cancers, such as colorectal and tumors, promoting uncontrolled proliferation.

Post-Translational Modifications and Degradation

p21 undergoes multiple post-translational modifications that fine-tune its subcellular localization, stability, and inhibitory activity on cyclin-dependent kinases, independent of transcriptional control. at Thr145 by (Akt) facilitates binding to 14-3-3 proteins, promoting nuclear export and cytoplasmic retention of p21, which inactivates its role in nuclear regulation. This modification is part of the Akt/GSK3β signaling axis, where GSK3β further phosphorylates p21 to reinforce cytoplasmic sequestration and limit its tumor-suppressive functions during proliferation. In contrast, at Ser146 by checkpoint kinase 2 (Chk2) occurs in response to DNA damage and stabilizes p21 by blocking ubiquitin-mediated degradation, thereby sustaining G1 arrest. Ubiquitination is the primary mechanism for p21 turnover, with distinct ligases acting in a cell cycle-dependent manner to ensure transient p21 accumulation. During , the SCF^{Skp2} complex targets phosphorylated p21 (particularly at Thr145) for proteasomal degradation, allowing progression to ; this process is redundant with other ligases but essential for overriding p21-mediated inhibition in cycling cells. In , the CRL4^{Cdt2} complex, recruited via interaction with (PCNA) at replication sites, ubiquitinates p21 to prevent its interference with . These pathways result in a short for p21 in asynchronously cycling cells, typically 20-30 minutes, enabling rapid adjustments to proliferative signals. Acetylation of at Lys161 and Lys163 by the acetyltransferase Tip60 competes with attachment sites in the C-terminal domain, inhibiting proteasomal and prolonging p21's antiproliferative effects, such as during . The overall of p21 can be conceptually represented by the \frac{d[p21]}{dt} = -k_{\deg} [p21], where k_{\deg} denotes the phase-specific , influenced by the balance of phosphorylations, ubiquitinations, and to dynamically control p21 levels.

Role in Disease

Cancer

p21 (CDKN1A) exhibits a paradoxical role in cancer, functioning primarily as a tumor suppressor through cell cycle inhibition and DNA damage response, yet promoting oncogenesis and therapy resistance in certain contexts. As a downstream effector of p53, p21 induces G1/S arrest to prevent propagation of genomic instability, thereby suppressing tumorigenesis. However, its dysregulation—through loss or overexpression—contributes to cancer progression across various malignancies. In its tumor suppressor capacity, p21 loss is frequently observed in human cancers, often via promoter hypermethylation, which silences expression and impairs DNA repair mechanisms, leading to genomic instability. This epigenetic inactivation occurs in multiple tumor types, including colorectal and breast cancers, where it promotes uncontrolled proliferation. For instance, promoter methylation of the CDKN1A locus has been reported in colorectal cancer cases. Conversely, elevated p21 expression in p53 wild-type tumors can confer pro-survival advantages and chemoresistance. In non-small cell lung cancer (NSCLC), high p21 levels in TP53 wild-type cells enhance survival during proliferation and post-chemotherapy, associating with poorer patient prognosis. This oncogenic shift may facilitate escape from p21-mediated during tumor progression, allowing adaptation to therapeutic stress. High p21 expression serves as a prognostic marker in several cancers, often indicating adverse outcomes. In , elevated CDKN1A correlates with reduced survival, particularly in aggressive subtypes. Similarly, in , high p21 is linked to poorer prognosis, reflecting its dual influence on cell fate decisions. Single nucleotide polymorphisms (SNPs) in CDKN1A, such as rs1059234 in the 3' , are associated with increased cancer risk, including of the head and neck and . Recent studies highlight p21's dual role in (CRC) signaling, where it modulates and mitotic fidelity in response to DNA damage, such as , promoting at high doses but preventing mitotic errors at low doses. Additionally, CDKN1A genotypes, including rs1059234 and rs1801270 variants, influence therapy response and overall cancer susceptibility, with certain alleles linked to heightened risk in leukemias and solid tumors. Mutations in CDKN1A are rare, with coding changes infrequently reported; however, epigenetic via promoter predominates as the mechanism of inactivation in cancers.

Other Pathologies

p21 contributes to organismal aging through tissue-specific induction of , where its upregulation leads to arrest in proliferative compartments, thereby limiting tissue renewal and promoting age-related decline. In models of , such as those involving BubR1 hypomorphs, p21 exhibits dual roles: it attenuates in some progenitor cells to preserve function but drives accelerated aging in others, highlighting its context-dependent impact on . Similarly, in Werner syndrome mouse models, p21 deficiency paradoxically accelerates aging phenotypes, including shortened lifespan and tissue degeneration, underscoring its protective role against premature aging in certain genetic backgrounds. In viral infections, p21 acts as a host restriction factor by inhibiting through cell cycle blockade. For instance, in elite HIV-1 controllers, selective upregulation of p21 in CD4+ T cells, including stem cell-like populations, restricts infection by suppressing activity required for reverse transcription and integration, thereby enhancing viral resistance without compromising cell viability. Regarding , infection triggers DNA damage responses that elevate p21 levels, inducing in airway epithelial and alveolar cells to limit viral spread, though this also contributes to persistent inflammation via the (SASP). p21 ablation enhances mammalian regenerative capacity by removing barriers that otherwise constrain tissue repair. In a seminal 2010 study, p21-null mice demonstrated scarless and partial regrowth, such as punch regeneration, mimicking regenerative traits observed in species like MRL mice, through sustained progenitor and reduced . Extensions in the 2020s have shown that p21 loss similarly accelerates cartilage repair in injury models by promoting and matrix deposition, with implications for cell-based therapies in musculoskeletal regeneration. In neurodegeneration, particularly (AD), p21 exerts protective effects by inducing neuronal cell cycle arrest in response to DNA damage and amyloid-beta accumulation, preventing aberrant re-entry into the that could exacerbate tau pathology and cell death. However, persistent p21-mediated senescence in post-mitotic neurons also drives pro-inflammatory SASP, secreting cytokines like IL-6 and IL-8 that amplify and glial activation, contributing to disease progression in AD brains. In cardiovascular pathologies, p21 is upregulated in response to hypertrophic stimuli, such as angiotensin II, where it inhibits cardiomyocyte and to maintain cardiac and prevent maladaptive remodeling. Conversely, in , defects in p21 degradation—often due to stabilizing the protein—lead to its excessive accumulation, impairing autophagic flux and exacerbating contractile dysfunction under stress conditions like lipopolysaccharide exposure.

Therapeutic Implications

Targeting Strategies

Targeting p21, also known as CDKN1A or p21WAF1/CIP1, involves pharmacological and genetic strategies to either enhance its expression for tumor suppression or inhibit its function to overcome resistance in contexts where it promotes oncogenesis. Activators primarily leverage upstream pathways or epigenetic mechanisms to upregulate p21 levels. For instance, Nutlin-3, an inhibitor, indirectly boosts p21 expression by stabilizing , leading to transcriptional activation of the CDKN1A gene in p53-wild-type cancer cells. Similarly, (HDAC) inhibitors such as promote epigenetic upregulation of p21 through increased histone acetylation at the CDKN1A promoter, resulting in arrest in various malignancies. In contrast, inhibitors aim to disrupt p21's inhibitory functions, particularly in resistant tumors where cytoplasmic p21 exerts pro-survival effects. Experimental small molecules targeting the p21-PCNA interaction, such as those identified through , block p21's binding to (PCNA), thereby sensitizing cancer cells to by impairing and replication. Recent developments highlight novel epigenetic modulators and applications. derivatives, as reviewed in 2025, act as multi-target epigenetic agents that upregulate p21 via HDAC and inhibition, offering improved selectivity for cancer therapy. Furthermore, p21 serves as a in strategies for aging, where intermittent clearance of p21-high senescent cells using targeted agents extends lifespan and enhances tissue function in preclinical models. A key challenge in p21 targeting is its context-dependent roles—tumor-suppressive in some settings but oncogenic in others—necessitating tumor profiling to predict responses and avoid adverse effects like accelerated proliferation.

Clinical Trials and Outcomes

Clinical trials evaluating p21 (CDKN1A) as a have demonstrated its utility in predicting responses in (NSCLC). A 2024 study utilizing (IHC) on tumor samples from TP53 wild-type (TP53WT) NSCLC patients found that high p21 expression correlates with poor and pro-survival effects, indicating reduced responsiveness to platinum-based regimens. This association highlights p21's role in sustaining cell survival under genotoxic stress, particularly in TP53-intact tumors where it may counteract treatment-induced . Intervention trials incorporating inhibitors (HDACi) like have explored p21-mediated mechanisms in , often revealing contributions to therapeutic . In preclinical models extended to clinical contexts, combined with upregulated p21, leading to arrest but also fostering in hormone-resistant cells; phase II trials post-2020, such as those evaluating HDACi with endocrine therapy (e.g., NCT03924264 for entinostat analogs), reported modest benefits yet noted p21 induction as a factor in incomplete responses. These findings underscore the dual-edged nature of p21 in HDACi-chemo combinations, where initial sensitization gives way to adaptive . Meta-analyses and cohort studies on p21 expression in head and neck cancers consistently associate low p21 levels with improved survival outcomes. In a study of patients with , reduced p21 expression predicted better overall survival. Emerging evidence also points to p21's role in synergy, as enhanced p21 expression in models sensitizes tumors to inhibitors by promoting and T-cell infiltration. Despite these insights, clinical application of p21 as a faces limitations due to assay heterogeneity and the need for more dynamic monitoring tools. Variations in IHC protocols and cutoff thresholds across studies lead to inconsistent , complicating standardization for routine use. Liquid biopsies, such as circulating tumor DNA assays detecting CDKN1A or expression, offer promise for overcoming tumor heterogeneity but require validation in larger prospective trials to address challenges.

Protein Interactions

Key Binding Partners

p21, encoded by the CDKN1A gene, primarily functions as a potent inhibitor of (CDK) activity by directly binding to cyclin-CDK complexes, thereby halting progression. Its core binding partners include A, D, and E, as well as CDKs 1, 2, 4, and 6. These interactions occur through distinct N-terminal domains of p21 that recognize both the cyclin and CDK subunits, with binding affinities typically in the low nanomolar range, as determined by and in structural studies. Evidence for these associations comes from co-immunoprecipitation (co-IP) assays and mass spectrometry-based interactome analyses, which consistently detect these complexes in cell lysates under physiological conditions. Another key interactor is (PCNA), to which p21 binds via its C-terminal PCNA-interacting protein (PIP) motif, inhibiting and repair processes. This interaction exhibits high affinity, with a (K_d) of approximately 2.5 nM for the full-length p21-PCNA complex, as measured by analysis, and has been validated through co-IP, , and of the p21 C-terminal fragment bound to PCNA. While does not directly bind p21, it indirectly regulates p21 expression as its primary transcriptional activator, leading to elevated p21 levels in response to DNA damage. Additional direct binding partners include MDM2, which interacts with p21 to promote its ubiquitin-independent proteasomal degradation, as demonstrated by in vitro pull-down assays and co-IP in p53-deficient cells. For degradation pathways, p21 is targeted by the SCF^{Skp2} E3 ubiquitin ligase complex through direct recognition by Skp2, particularly when p21 is hypophosphorylated, with evidence from ubiquitylation assays and mass spectrometry identifying Skp2-p21 complexes in S-phase cells. Similarly, the CRL4^{Cdt2} complex binds p21 in a PCNA-dependent manner during S phase, facilitating its ubiquitylation, as shown by co-IP and in vivo degradation studies. p21 enforces G1 arrest by inhibiting CDKs, thereby maintaining the retinoblastoma protein (Rb) in its active, hypophosphorylated form to repress E2F transcription factors. Interactome databases such as STRING reveal approximately 50 high-confidence partners (interaction score ≥ 0.7) for p21, derived from experimental (e.g., co-IP, affinity capture-MS) and database-curated evidence, underscoring its role in diverse cellular processes beyond cell cycle control. Recent studies have implicated p21 in Hippo-Wnt pathway crosstalk through regulatory interactions influenced by YAP/TEAD activity, where YAP/TEAD knockdown elevates p21 levels, though direct binding remains context-dependent.

Pathway Integration

p21 serves as a central in the DNA damage response (DDR) pathway, linking the /ATR kinases to -dependent . Upon detection of double-strand breaks or replication stress, and ATR kinases phosphorylate and activate by inhibiting its negative regulators, such as , leading to stabilization and transcriptional activation of p21. The induced p21 then inhibits complexes (e.g., CDK2-cyclin E), enforcing G1/S arrest to allow ; failure to repair triggers sustained arrest, , or via this axis. This /ATR--p21 cascade also coordinates intra-S and G2/M checkpoints through CHK2/CHK1-mediated crosstalk, preventing propagation of genomic instability. In growth control networks, p21 modulates the balance between Wnt and Hippo pathways, influencing homeostasis and . p21 negatively regulates Wnt signaling by repressing transcription of Wnt ligands like Wnt4 in a cycle-independent manner, thereby limiting β-catenin-driven . Concurrently, p21 loss shifts the /Rb-E2F1 toward MMB/E2F1 , upregulating 1, which binds β-catenin to suppress Wnt while enhancing Notch outputs like Hes1; p21 overexpression restores this Wnt-Notch equilibrium. Through these interactions, p21 integrates Hippo effectors like /TAZ, which inversely regulate p21 to fine-tune nuclear YAP localization and prevent unchecked growth, as seen in YAP-dependent downregulation of p21 during oncogenic stress. p21 cooperates with the TGF-β/Smad pathway to mediate growth arrest during epithelial-mesenchymal transition (), balancing differentiation and motility in epithelial cells. TGF-β receptor activation phosphorylates Smad2/3, which translocate to the and induce p21 expression alongside CDK inhibitors like p15^INK4B, repressing c-Myc and enforcing G1 arrest to coordinate EMT timing. This Smad-p21 axis ensures transient arrest for cytoskeletal remodeling without permanent , as p21 knockdown disrupts TGF-β-induced hypophosphorylation of and delays EMT progression. In contexts like or , this integration prevents hyperproliferative responses during Smad-driven transcription of EMT factors like . Recent studies highlight p21's integration into (CRC) pathways, particularly through cytoplasmic localization activating to promote stemness. In CRC models, cytoplasmic p21 translocates to the nucleus to inhibit p65 acetylation, but its dysregulation enhances -driven transcription of stemness genes, linking p21 to Wnt//PI3K axes for tumor initiation. A 2024 review underscores p21's suppression by PI3K/AKT in CRC, where it intersects for proliferation control, while miR-125b-mediated p21 upregulation ties into p53-dependent amid Wnt dysregulation. In networks, acts as a hub restraining to maintain viability under persistent . knockdown in senescent cells amplifies activation, elevating TNF-α secretion and JNK/ signaling, which exacerbates DNA lesions and () via unchecked . This feedback loop integrates with (/) and inflammatory pathways, preventing hyperactivation; inhibition partially rescues -deficient senescence lethality, underscoring 's role in over five interconnected networks including TGF-β and Wnt for systemic .

References

  1. [1]
    CDKN1A cyclin dependent kinase inhibitor 1A [ (human)] - NCBI
    Sep 27, 2025 · Activation of p21 limits acute lung injury and induces early senescence after acid aspiration and mechanical ventilation. Guanine nucleotide ...
  2. [2]
    p21 in Cancer Research - PMC - NIH
    Aug 14, 2019 · p21 functions as a cell cycle inhibitor and anti-proliferative effector in normal cells, and is dysregulated in some cancers.
  3. [3]
    Biology of the cell cycle inhibitor p21(CDKN1A) - PubMed
    The main, well-known function of p21 is to arrest cell cycle progression by inhibiting the activity of cyclin-dependent kinases. In addition, p21 is involved in ...
  4. [4]
    CDKN1A Gene - Cyclin Dependent Kinase Inhibitor 1A - GeneCards
    Q9BUT4. Protein attributes for CDKN1A Gene. Size: 164 amino acids; Molecular mass: 18119 Da.
  5. [5]
  6. [6]
    Cyclin-dependent kinase inhibitor 1 - P38936 - UniProt
    Binds to and inhibits cyclin-dependent kinase activity, preventing phosphorylation of critical cyclin-dependent kinase substrates and blocking cell cycle ...
  7. [7]
    Translational control of a human CDKN1A mRNA splice variant ...
    Expression of the cyclin-dependent kinase (CDK) inhibitor CDKN1A that encodes the p21 (CIP1/WAF1) protein is enhanced in response to UVB. The resulting elevated ...Missing: isoforms | Show results with:isoforms<|separator|>
  8. [8]
    The p21 Protein in Cell Differentiation, Migration, and Cancer Therapy
    Overall, basal p21 expression is required for stem cell maintenance protecting the cells from exhaustion, whereas high levels of p21 trigger differentiation ...
  9. [9]
    Long non-coding RNA generated from CDKN1A gene by alternative ...
    We show that a DDR-activated APA event occurs in the first intron of CDKN1A, inducing an alternate last exon-containing lncRNA. We named this lncRNA SPUD.Missing: size | Show results with:size<|separator|>
  10. [10]
    The Role of the Cyclin Dependent Kinase Inhibitor p21 cip1/waf1 in ...
    p21 binds the cyclin subunit through a conserved cyclin-binding motif 1 (Cy1), which is present in the N-terminal region. Additionally, p21 binds through ...<|control11|><|separator|>
  11. [11]
    Cell cycle regulation by the intrinsically disordered proteins, p21 and ...
    In this review, we provide a general introduction to IDPs and discuss how the intrinsic disorder of two prototypical IDPs, p21 and p27, mediate cell cycle ...
  12. [12]
    p21 contains independent binding sites for cyclin and cdk2 - PubMed
    p21 has independent binding sites for cyclin and cdk2. Both sites are required to inhibit cdk2 kinase activity. Binding via only one site does not inhibit.Missing: structure intrinsically disordered regions crystal structures PDB 1H8G
  13. [13]
    Cell cycle regulation by the intrinsically disordered proteins p21 and ...
    Sep 19, 2012 · The unique C-terminal region of p21 directly associates with the DNA polymerase δ processivity factor PCNA (proliferating-cell nuclear ...Functions Of P21 And P27 · P21 And P27 As Complex... · P21 And P27 As Flexible...
  14. [14]
  15. [15]
    CRL4BDCAF11 E3 ligase targets p21 for degradation to control cell ...
    Apr 26, 2017 · K16, K154, K161 and K163 are required for p21 ubiquitination. The amino acid sequence of p21 contains six lysine (K) residues, K16, K75, K141, ...
  16. [16]
    Evolutionary Approach of Intrinsically Disordered CIP/KIP Proteins
    Feb 7, 2019 · Here, p21 and p57 have a proliferating nuclear antigen (PCNA)-binding motif, and p27 and p57 share, a structural motif that is likely to ...
  17. [17]
    Inhibition of cyclin-dependent kinases by p21.
    Oct 13, 2017 · We have explored the interaction of p21 with the currently known Cdks. p21 effectively inhibits Cdk2, Cdk3, Cdk4, and Cdk6 kinases (Ki 0.5-15 nM) ...
  18. [18]
    Cell cycle regulation: p53-p21-RB signaling - Nature
    Mar 31, 2022 · The release from repression can be prevented by the cyclin-dependent kinase inhibitor p21/CDKN1A. The CDKN1A gene is transcriptionally activated ...
  19. [19]
    Basal p21 controls population heterogeneity in cycling and ... - PNAS
    We postulated that the bistability in reversible cell states arose from double-negative feedback regulation involving CDK2 and p21 (Fig. 3 A and B). We created ...
  20. [20]
    Mechanisms of Cellular Senescence: Cell Cycle Arrest ... - Frontiers
    Cell cycle arrest in senescence is largely mediated via activation of either one or both p53/p21WAF1/CIP1 and p16INK4A/pRB tumor suppressor pathways (Rovillain ...
  21. [21]
    Inhibition of cyclin-dependent kinases by p21 - PMC - NIH
    We have explored the interaction of p21 with the currently known Cdks. p21 effectively inhibits Cdk2, Cdk3, Cdk4, and Cdk6 kinases (Ki 0.5-15 nM) but is much ...
  22. [22]
    The p21 inhibitor of cyclin-dependent kinases controls DNA ... - Nature
    Jun 16, 1994 · p21 directly inhibits PCNA-dependent DNA replication in the absence of a cyclin/CDK. Furthermore, p21 blocks the ability of PCNA to activate DNA polymerase δ.
  23. [23]
    The C-terminal Domain of p21 Inhibits Nucleotide Excision Repair In ...
    Oct 13, 2017 · Although p21/PCNA binding occurs in a 1:1 ratio, a 5- to 10-fold excess of p21 to PCNA is necessary to inhibit PCNA-dependent replication both ...Missing: epsilon | Show results with:epsilon
  24. [24]
    CDK-Independent and PCNA-Dependent Functions of p21 in DNA ...
    p21 is a 165 amino acid protein characterized by high intrinsic molecular flexibility and nondefined tertiary structure unless when bound to other proteins [4].Missing: PDB 1H8G
  25. [25]
    Cyclin Kinase-independent role of p21 CDKN1A in the promotion of ...
    Oct 14, 2016 · The DNA replication defects caused by p21-depletion caused accumulation of replication stress markers, such as γH2AX and 53BP1, instability of ...
  26. [26]
    The Multiple Battles Fought by Anti-Apoptotic p21 - PubMed
    Very recently, we have shown that p21 protects cells from irradiation-induced apoptosis by suppression of CDK activity that appears to be required downstream of ...
  27. [27]
    Cellular senescence and SASP in tumor progression and ...
    Aug 31, 2024 · Phosphorylated p53 subsequently activates the downstream protein p21 [7]. p21 inhibits the phosphorylation of retinoblastoma protein (RB) by ...Missing: hyperphosphorylation | Show results with:hyperphosphorylation
  28. [28]
    p21 produces a bioactive secretome that places stressed cells under ...
    Here, we report that the cell cycle inhibitor p21 places cells under immunosurveillance to establish a biological timer mechanism that controls cell fate. p21 ...
  29. [29]
  30. [30]
    Dual role of p21 in regulating apoptosis and mitotic integrity in ...
    Apr 2, 2025 · At high doses (HD), p21 is more critical than p53 in mediating apoptosis, whereas at low doses (LD), p21 is essential for preventing mitotic ...
  31. [31]
    p53 mediated senescence impairs the apoptotic response to ... - NIH
    Doxorubicin-treated p53-mutant tumors failed to arrest proliferation leading to abnormal mitoses and cell death, while p53 wild-type tumors arrested, avoiding ...
  32. [32]
    When therapy-induced senescence meets tumors: A double-edged ...
    Jun 13, 2025 · However, existing evidence suggests that TIS may contribute to treatment failure by inducing tumor dormancy and triggering recurrence.
  33. [33]
    Lack of p21 expression links cell cycle control and appendage ...
    We directly tested the hypothesis that p21 down-regulation could functionally induce a regenerative response in an appendage of an otherwise nonregenerating ...
  34. [34]
    The role of p21 in regulating mammalian regeneration
    Jun 29, 2011 · A functional role for p21 was confirmed when tissue injury in an adult p21 -/- mouse showed a healing phenotype that matched the MRL mouse.
  35. [35]
    (PDF) Mutant P53 induces MELK expression by release of wild-type ...
    ... p53 response elements. (RE1, RE2, RE3, and RE4) is critical for WT p53 ... p21 promoter (used as a positive control). We. analyzed several p53-ChIP-seq ...
  36. [36]
    A Novel Evolutionarily Conserved Element Is a General ...
    Sp1/Sp3 is a potent activator for the p21 promoter activity, and the 6 classic binding sites within the −119 to −50 bp on the p21 promoter have been found to be ...
  37. [37]
    H(2)O(2)-induced AP-1 activation and its effect on p21(WAF1/CIP1)
    May 17, 2002 · Taken together, these results revealed a novel AP-1 binding site in ... Substances. CDKN1A protein, human; Cyclin-Dependent Kinase ...
  38. [38]
    Transcriptional regulation of the cyclin-dependent kinase inhibitor ...
    The cyclin-dependent kinase inhibitor 1A (CDKN1A), also known as p21 (WAF1/CIP1) modulates cell cycle, apoptosis, senescence and differentiation via ...
  39. [39]
    RAS/MAPK signaling functions in oxidative stress, DNA damage ...
    Feb 27, 2019 · During DNA damage response, the RAS/MAPK pathway is regularly activated, which contributes to the appropriate activation of DDR checkpoints ...
  40. [40]
    Cell cycle progression in response to oxygen levels
    May 25, 2014 · Hypoxia can also result in the induction of p21 in many cell types leading to a G1 arrest [66, 68]. The transcriptional regulation of p21 by ...
  41. [41]
    Regulated recruitment of tumor suppressor BRCA1 to the p21 gene ...
    Jan 15, 2011 · CARM1 and p300 cooperate with BRCA1 and p53 to induce expression of the critical cell cycle and proliferation regulator p21(WAF1/CIP1) in response to DNA ...Missing: PML miR- 93
  42. [42]
    PML contributes to p53-independent p21 up-regulation in gamma ...
    Oct 2, 2011 · In the present study, we showed that the tumor suppressor, promyelocytic leukemia protein (PML) contributes to the up-regulation of p21 in a p53 ...
  43. [43]
    MicroRNA93 Regulates Proliferation and Differentiation of Normal ...
    Jun 7, 2012 · To determine the molecular mechanisms of mir-93 CSC regulation, we employed an unbiased approach assessing the effect of mir-93 expression ...Missing: PML | Show results with:PML
  44. [44]
    Stimulus‐dependent dynamics of p53 in single cells - EMBO Press
    May 10, 2011 · We quantified the dynamics of p53 in individual cells in response to UV and observed a single pulse that increases in amplitude and duration in proportion to ...
  45. [45]
    Epigenetic Regulation of p21 cip1/waf1 in Human Cancer - MDPI
    p21cip1/waf1 is a low-molecular-weight molecule (21 kDa) that inhibits cyclin-dependent kinases. It is a key downstream target of p53 upon DNA damage and ...Missing: aliases | Show results with:aliases
  46. [46]
    Epigenetic Regulation of p21cip1/waf1 in Human Cancer - PMC
    p21cip1/waf1 is a central regulator of cell cycle control and survival. While mutations are rare, it is commonly dysregulated in several human cancers due to ...Missing: H3K9ac | Show results with:H3K9ac
  47. [47]
    The role of p21 in cellular senescence and aging-related diseases
    The p21-regulated cell cycle pathway operates through both p53-dependent and p53-independent mechanisms. In the p53-dependent pathway, exposure to factors such ...
  48. [48]
    p21 Both Attenuates and Drives Senescence and Aging in BubR1 ...
    Apr 25, 2013 · Together, these data suggest that p21, through p53 activation, acts to protect progenitor cells from senescence due to BubR1 insufficiency in ...
  49. [49]
    The Distinct Function of p21Waf1/Cip1 With p16Ink4a in ... - Frontiers
    In summary, our data revealed that p21 deficiency accelerated the aging process in WS, shortened mouse lifespan, and accelerated the degeneration of bone, ...
  50. [50]
    CD4+ T cells from elite controllers resist HIV-1 infection by selective ...
    Mar 14, 2011 · This suggests that p21 acts as a barrier against HIV-1 infection in CD4+ T cells from elite controllers by inhibiting a cyclin-dependent kinase ...
  51. [51]
    SARS-CoV-2 Infection Induces DNA Damage & Senescence
    Mar 9, 2023 · SARS-CoV-2 proteins ORF6 and NSP13 cause degradation of the DNA damage response kinase CHK1 through proteasome and autophagy, respectively.
  52. [52]
    Deletion of p21 expression accelerates cartilage tissue repair via ...
    Mar 13, 2020 · The association between the regulation of cell cycle checkpoints and tissue regeneration has been previously investigated, and p21 was initially ...
  53. [53]
    Senescence and SASP Are Potential Therapeutic Targets for ... - MDPI
    Senescence was initially observed in the penumbra and later in the ischemic core, and elevated p21 levels may protect the neuron by arresting the cell cycle.
  54. [54]
    Oridonin protects against cardiac hypertrophy by promoting P21 ...
    May 24, 2019 · Our data show that oridonin promoted P21-related autophagic lysosomal degradation, hence attenuating oxidative injury and cardiac hypertrophy.
  55. [55]
    Sirt1-inducible deacetylation of p21 promotes cardiomyocyte ... - NIH
    In the present study, the acetylation of p21 may inhibit p21 degradation by the ubiquitin-proteasome pathway, exhibiting a high activity and abundance of p21 ...
  56. [56]
    Autophagy is involved in the protective effect of p21 on LPS-induced ...
    Jul 21, 2020 · These results revealed that p21 controls LPS-induced cardiac dysfunction by modulating inflammatory and oxidative stress, and it is partially ...
  57. [57]
    Pharmacological activation of the p53 pathway by nutlin-3 exerts ...
    The tumor-promoting p53-MDM2 interaction can be inhibited by the small molecule, nutlin-3, restoring p53 function. Targeting the p53-MDM2 axis using nutlin-3 ...
  58. [58]
    Cancer biology: mechanism of antitumour action of vorinostat ...
    Dec 12, 2006 · Induction of gene expression for the cell kinase inhibitor p21WAF1 has been shown for several HDAC inhibitors including vorinostat and this ...
  59. [59]
    Identification of Small Molecule Proliferating Cell Nuclear Antigen ...
    Significance: Inhibition of the PCNA protein-protein interaction can be a new strategy to sensitize cancer cells to chemotherapy. Keywords: Anticancer Drug, ...
  60. [60]
    Proteolysis-targeting chimeras (PROTACs) in cancer therapy
    Apr 11, 2022 · Photocaged PROTACs irreversibly accomplish protein degradation by incorporating photolabile blocking groups (e.g. nitroveratryloxycarbonyl group ...
  61. [61]
    P21 expressing monocytes for cancer cell therapy - Google Patents
    They showed that overexpression of p21 can act as a macrophage ... adeno-associated virus for in vivo delivery thereof and application in cancer treatment.
  62. [62]
    Anticancer benzimidazole derivatives as inhibitors of epigenetic ...
    Jan 13, 2025 · This review provides a comprehensive overview of benzimidazole derivatives that inhibit different acetylation and methylation reader, writer and eraser ...
  63. [63]
    p21 in cancer: intricate networks and multiple activities - PMC
    p21 also inhibits CDK activity indirectly by interfering with the activating phosphorylation of CDK1 and CDK2 in the activation segment by an unidentified ...
  64. [64]
    Pro-survival roles for p21(Cip1/Waf1) in non-small cell lung cancer
    Dec 20, 2024 · In summary, our analyses show that high p21 expression correlates with poor NSCLC patient prognosis specifically in TP53WT tumours, which ...
  65. [65]
    Vorinostat (SAHA) and Breast Cancer: An Overview - MDPI
    The article presents the state of the knowledge on vorinostat (SAHA) in the therapy of various histological subtypes of BC, individually or in polytherapy.
  66. [66]
    Impact of Cyclin-dependent Kinase Inhibitor 1A Genotypes ... - In Vivo
    Taken together, these findings suggest that rs1801270 and rs1059234 variants in CDKN1A may contribute to cancer susceptibility, though their role appears to be ...
  67. [67]
    Prognostic significance of p21, p27 and survivin protein expression ...
    Jun 7, 2013 · The results of the current study revealed that the five-year survival rate was significantly lower in patients with high p21 expression. In ...
  68. [68]
    Enhanced expression of p21 promotes sensitivity of melanoma cells ...
    May 6, 2022 · We show that p53 activation increases BRAFi sensitivity in a synergistic manner exclusively in cells with a high expression of CDKN1A/p21. In a ...
  69. [69]
    Liquid biopsy and tumor heterogeneity in metastatic solid tumors
    May 27, 2020 · In this review, we summarize the evidences on the role of blood-based liquid biopsy as a potential tool to capture tumor heterogeneity in metastatic cancer ...
  70. [70]
    DNA Methylation-Based Testing in Liquid Biopsies as Detection and ...
    Firstly, and mainly, tissue samples might not fully represent tumor heterogeneity, constituting a limitation for accurate outcome prediction and treatment ...
  71. [71]
    Structural studies of p2lWa1CiPl/Sdil in the free and Cdk2-bound
    The p21 kinase inhibitory domain, therefore, undergoes a dramatic disorder-order transition upon binding to Cdk2. What is thebiological role for the disorder- ...
  72. [72]
    p21 Is a Critical CDK2 Regulator Essential for Proliferation Control ...
    In vitro, p21 has a very high affinity for complexes containing CDK2 and CDK4 (Harper et al., 1995). To examine the role of p21 in the regulation of these G1 ...
  73. [73]
    A 39 amino acid fragment of the cell cycle regulator p21 is sufficient ...
    The cell cycle regulator p21 interacts with and inhibits the DNA replication and repair factor proliferating cell nuclear antigen (PCNA).
  74. [74]
    Role of the SCFSkp2 ubiquitin ligase in the degradation of p21Cip1 ...
    Jul 11, 2003 · We show here that p21 is a good substrate for an SCF (Skp1-Cullin1-F-box protein) ubiquitin ligase complex, which contains the F-box protein Skp2 (S phase ...Missing: Cdt2 | Show results with:Cdt2
  75. [75]
    The CRL4Cdt2 ubiquitin ligase targets the degradation of p21Cip1 ...
    Human CRL4Cdt2 targets the degradation of p21 in S phase, with the in vivo ubiquitylation of p21 by CRL4Cdt2 dependent on p21 binding to PCNA. Inactivation of ...
  76. [76]
    CDKN1A protein (human) - STRING interaction network
    Minimum required interaction score: highest confidence (0.900), high confidence (0.700), medium confidence (0.400), low confidence (0.150), custom value. Max ...Missing: interactome | Show results with:interactome
  77. [77]
    In NSCLC cells, p21 is derepressed upon YAP/TAZ and TEAD1 ...
    In H1299 and H1975 cells, p21 protein and transcript were increased upon YAP, TAZ and TEAD siRNA-mediated knockdown as well as upon CER treatment.
  78. [78]
    DNA damage response revisited: the p53 family and its regulators ...
    Oct 7, 2022 · As discussed above, ATM-CHK2 and ATR-CHK1 are the central kinases that constantly phosphorylate p53 and its negative regulators (such as MDM2, ...
  79. [79]
    p21 WAF1/Cip1 is a negative transcriptional regulator of Wnt4 ...
    p21 is a negative regulator of Wnts transcription downstream of Notch1 activation, independently of effects on the cell cycle.
  80. [80]
    p21 Regulates Wnt-Notch balance via DREAM/MMB/Rb-E2F1 and ...
    Sep 28, 2024 · The overexpression of p21 rescued the DREAM pathway, as well as the imbalance of Wnt-Notch pathway. In summary, our data identify p21 as an ...Missing: review | Show results with:review
  81. [81]
    Non-Smad pathways in TGF-β signaling | Cell Research - Nature
    Dec 30, 2008 · In the growth arrest response, TGF-β may inhibit ... The PI3K/Akt pathway is another non-Smad pathway contributing to TGF-β-induced EMT.
  82. [82]
    Signaling pathways involved in colorectal cancer: pathogenesis and ...
    Oct 7, 2024 · Apart from the Ras pathway, the JNK signaling pathway displays a dual role in CRC progression. The proto-oncoprotein c-Jun, part of the AP-1 ...
  83. [83]
  84. [84]
    p21 maintains senescent cell viability under persistent DNA damage ...
    Here, we show that the CDK inhibitor p21 (CDKN1A) maintains the viability of DNA damage‐induced senescent cells. Upon p21 knockdown, senescent cells acquired ...
  85. [85]
    Control of the senescence-associated secretory phenotype by NF ...
    We find that NF-κB is a master regulator of the SASP and then use this to further probe the impact of SASP biology on the senescence program. Results.