Fact-checked by Grok 2 weeks ago

CDKN1B

CDKN1B is a protein-coding located on chromosome 12p13.1 that encodes inhibitor 1B, commonly known as p27<sup>Kip1</sup>, a key regulator of the that inhibits the activity of (CDK) complexes to prevent progression from the to the . This 198-amino-acid protein functions primarily by binding to and inhibiting E-CDK2 and D-CDK4/6 complexes, thereby maintaining cellular quiescence and responding to antiproliferative signals such as damage or cues. Beyond its core inhibitory role, p27<sup>Kip1</sup> acts as a , facilitating the assembly of certain CDK complexes and modulating non-cell cycle processes including , , and through interactions with proteins like RhoA and c-Jun. The regulation of CDKN1B expression and p27<sup>Kip1</sup> activity is tightly controlled at multiple levels to ensure precise control. Transcriptionally, the is responsive to growth factors and signals, while post-translationally, the protein undergoes (e.g., at Thr187 by CDK2 or Tyr88 by kinases) leading to ubiquitination and proteasomal degradation, which is essential for . Nuclear export via at Ser10 allows cytosolic localization, where p27<sup>Kip1</sup> influences dynamics and cell motility. As an intrinsically disordered protein, p27<sup>Kip1</sup> exhibits high conformational flexibility, enabling it to adopt structured conformations upon binding partners, which underlies its multifaceted roles. Dysregulation of CDKN1B is implicated in various pathologies, particularly as a tumor suppressor. heterozygous mutations, such as variants (e.g., W76X) or frameshifts, cause type 4 (MEN4), characterized by tumors in the pituitary, parathyroid, and other endocrine tissues. Somatic alterations, including low expression or inactivating mutations, are associated with aggressive cancers like , , colorectal, and neuroendocrine tumors, correlating with poor due to unchecked . Additionally, p27<sup>Kip1</sup> promotes tumorigenesis in mouse models, highlighting its role in suppressing and maintaining tissue homeostasis.

Gene and Protein Overview

Gene Structure and Expression

The CDKN1B gene is situated on the short arm of human chromosome 12 at locus 12p13.1, spanning approximately 37 kilobases of genomic DNA from position 12,685,498 to 12,722,373 on the forward strand (GRCh38 assembly). This genomic region encompasses three exons, with the coding sequence distributed across exons 2 and 3, while exon 1 is primarily non-coding and includes part of the 5' untranslated region. The gene's compact structure facilitates tight transcriptional control, essential for its role in cell cycle regulation. The promoter of CDKN1B lacks a , characteristic of many and cell cycle-related genes, relying instead on initiator elements and proximal binding sites for basal transcription. Key regulatory features include consensus binding sites for Forkhead box O (FoxO) transcription factors, such as FoxO1, located upstream of the transcription start site. These sites enable FoxO-mediated activation in response to signaling or stress conditions, promoting CDKN1B transcription in nutrient-deprived or growth-arrested states. CDKN1B demonstrates ubiquitous basal expression across human tissues, as detected by and analyses, reflecting its fundamental role in cellular . According to GTEx data (as of 2023), expression levels vary, with the highest median TPM observed in (~35-40 TPM), high levels in heart and (~25 TPM), moderate levels in liver (~15 TPM) and colon (~12 TPM), where it supports maintenance of the G0/G1 phase in non-proliferating cells; in contrast, lower expression occurs in rapidly dividing tissues like and testis (~5-10 TPM). These patterns align with CDKN1B's upregulation in response to antiproliferative signals, contributing to restraint in post-mitotic environments. Older studies reported highest levels in , but provides a more comprehensive view. Alternative splicing of CDKN1B pre-mRNA generates at least 10 transcript variants, though the majority are lowly expressed and predicted to produce non-coding or truncated products. The canonical isoform, encoded by transcript ENST00000228872 (NM_004064.5), translates to the full-length p27Kip1 protein of 198 , which predominates in most tissues. Rare isoforms, such as those retaining intronic sequences or altering the 3' , may modulate mRNA stability or localization but lack well-characterized functional impacts.

Protein Structure and Localization

The p27<sup>Kip1</sup> protein, encoded by the CDKN1B gene, consists of 198 with a calculated molecular weight of approximately 22 kDa, though post-translational modifications such as can increase its apparent size to around 27 kDa on SDS-PAGE gels. The protein features an intrinsically disordered structure, particularly in its N- and C-terminal regions, which enables flexible interactions with binding partners. The N-terminal region contains the cyclin-binding domain spanning residues 1-65, which facilitates association with subunits in cyclin-CDK complexes. Adjacent to this is the central CDK-inhibitory domain (residues 28-106), also known as the kinase inhibitory domain (), which directly blocks the of cyclin-dependent kinases (CDKs) to inhibit their activity. The C-terminal region harbors a nuclear localization signal (NLS) at residues 153-187, promoting import into the , as well as a nuclear export signal () that mediates shuttling out of the . Key sites, such as 187 (Thr187), are targeted by CDK2, altering the protein's conformation and influencing both its and subcellular by facilitating for ubiquitin-mediated processes. Other sites, including serine 10 (Ser10) and 157 (Thr157), modulate localization through interactions with export machinery or retention factors like 14-3-3 proteins. p27<sup>Kip1</sup> is predominantly localized in the during the G0 and G1 phases of the , where it exerts its inhibitory effects on progression. In response to mitogenic signals, events trigger its export to the via the CRM1-dependent pathway, utilizing the , which sequesters the protein away from nuclear CDK targets and potentially enables cytoplasmic functions.

Molecular Function

Cell Cycle Inhibition Mechanism

The protein product of CDKN1B, known as p27<sup>Kip1</sup>, primarily enforces arrest at the G1/S checkpoint by binding to and inhibiting cyclin E-CDK2 and cyclin A-CDK2 complexes. These inhibitory interactions prevent the phosphorylation of the (Rb), thereby maintaining Rb in its hypophosphorylated state and repressing the transcriptional activity of transcription factors essential for S-phase entry. In this manner, elevated p27<sup>Kip1</sup> levels ensure that cells remain in G1 until growth-promoting signals are sufficient to titrate away the inhibitor. The inhibitory action of p27<sup>Kip1</sup> follows a stoichiometric binding model, wherein a single p27<sup>Kip1</sup> molecule associates with one CDK subunit within the cyclin-CDK complex. reveals that the of p27<sup>Kip1</sup> inserts into the catalytic cleft of CDK2, directly occluding the ATP-binding and distorting the kinase's active conformation to abolish . This precise, one-to-one inhibition contrasts with non-stoichiometric models and underscores p27<sup>Kip1</sup>'s efficiency as a tight-binding CDK regulator. In maintaining G0 quiescence, p27<sup>Kip1</sup> additionally sequesters D-CDK4/6 complexes in an inactive state at high p27<sup>Kip1</sup> concentrations typical of non-proliferating states, while at lower levels it promotes their assembly and localization. This mode involves p27<sup>Kip1</sup>, which initially promotes assembly of D-CDK4/6 but at excess levels binds and renders the holoenzymes inactive, reinforcing the quiescent phenotype by limiting early G1 progression. A key amplifying p27<sup>Kip1</sup>-mediated involves its induction via the transforming factor-β (TGF-β) signaling pathway, which transcriptionally upregulates CDKN1B expression to enforce G1 in response to antiproliferative cues. This pathway links extracellular inhibitors to intracellular CDK suppression, ensuring rapid and sustained blockade.

Binding to Cyclin-CDK Complexes

p27<sup>Kip1</sup>, encoded by the CDKN1B gene, primarily exerts its inhibitory effects on progression by to (CDK) complexes, with a particular emphasis on interactions involving G1/S-phase regulators. The process is sequential, beginning with the recognition of the subunit, which positions the inhibitory domain of p27<sup>Kip1</sup> for subsequent engagement with the CDK. This ensures specific and high-affinity with active cyclin-CDK holoenzymes, preventing access and ATP at the catalytic site. A key feature of this involves hydrophobic contacts within the cyclin-binding groove. The N-terminal of p27<sup>Kip1</sup> docks into a hydrophobic pocket on the surface, exemplified by interactions with residues in the α-helices of E, such as Leu258 and Phe256, which stabilize the complex through van der Waals forces and contribute to specificity for cyclins over others. This initial docking induces a conformational rearrangement in p27<sup>Kip1</sup>, transitioning its intrinsically disordered inhibitory domain into a structured form. Notably, a short 3<sub>10</sub>- in the second subdomain of p27<sup>Kip1</sup> (residues approximately 28–36) inserts directly into the ATP-binding cleft of CDK2, occluding the and mimicking the ring of ATP to block binding and phosphate transfer. This insertion distorts the CDK2 catalytic loop, rendering the inactive without altering its overall fold. The affinity of p27<sup>Kip1</sup> for these complexes varies, reflecting functional nuances in control. It exhibits high affinity for E- and A-CDK2 complexes, enabling potent suppression of S-phase entry. In contrast, binding to D-CDK4/6 is weaker, allowing partial activity in early G1 while still permitting inhibition at elevated p27<sup>Kip1</sup> levels. This differential affinity arises from subtle structural differences in the cyclin grooves and CDK activation loops, with E/A forming tighter interfaces via conserved motifs like the MRAIL in p27<sup>Kip1</sup>. p27<sup>Kip1</sup> displays a biphasic role in modulating D-CDK4 assembly, dependent on its concentration. At low levels, p27<sup>Kip1</sup> acts as a scaffold, promoting the formation and nuclear localization of active D-CDK4 complexes by stabilizing subunit interactions and facilitating T-loop , as evidenced in p27/p21 double-knockout fibroblasts where complex assembly is severely impaired. Recent structural studies have revealed that p27<sup>Kip1</sup> allosterically activates CDK4 by rotating its T-loop, facilitating and activity in the D-CDK4-p27 ternary complex at low p27 levels. However, at higher concentrations, p27<sup>Kip1</sup> shifts to a dominant inhibitory mode, fully blocking CDK4 activity by occupying the catalytic site and preventing , thus enforcing G1 under or quiescence signals. This dual functionality fine-tunes G1 progression without overlapping the broader inhibitory effects on CDK2 complexes.

Regulation of Expression and Activity

Transcriptional Control

The transcriptional regulation of the CDKN1B gene, which encodes the inhibitor p27^Kip1, is governed by a of transcription factors that respond to mitogenic and stress signals, as well as epigenetic modifications that modulate promoter accessibility. Key activators include members of the Forkhead box O (FoxO) family, such as FoxO3a, which bind to specific sites in the CDKN1B promoter to drive its expression. Inhibition of the PI3K/AKT pathway, often triggered by deprivation or therapeutic agents, dephosphorylates FoxO3a, allowing its nuclear translocation and subsequent enhancement of CDKN1B transcription, thereby promoting arrest. In contrast, repressors like c-Myc suppress CDKN1B transcription during proliferative states by binding to elements in the promoter or indirectly inhibiting FoxO3a activity, facilitating progression. Similarly, E2F1 can contribute to repression in certain contexts by interacting with promoter elements, often in coordination with other factors to downregulate CDKN1B during S-phase entry. These opposing regulatory mechanisms ensure tight control of p27^Kip1 levels in response to extracellular cues like serum availability. Epigenetic alterations further fine-tune CDKN1B expression. Histone acetylation, particularly at H3K9ac marks on the promoter, correlates with an open state and active transcription, often observed in quiescent or differentiated cells. Conversely, hypermethylation of CpG islands in the CDKN1B promoter leads to in various cancers, reducing p27^Kip1 levels and promoting uncontrolled proliferation. These modifications are dynamically influenced by environmental signals, such as contact inhibition, which can upregulate CDKN1B through .

Post-Translational Modifications and Degradation

The stability and activity of the p27<sup>Kip1</sup> protein, encoded by CDKN1B, are primarily regulated through post-translational modifications that influence its degradation, localization, and interactions with (CDK) complexes. In proliferating cells, p27<sup>Kip1</sup> exhibits a short of approximately 2-6 hours, which is extended in quiescent cells to maintain arrest. A key regulatory mechanism involves ubiquitination and proteasomal degradation mediated by the SCF<sup>Skp2</sup> ubiquitin ligase complex, which targets p27<sup>Kip1</sup> following at threonine 187 (Thr187). This , often catalyzed by E-CDK2, creates a binding site for the adaptor protein Skp2, facilitating polyubiquitination and subsequent degradation by the 26S , thereby allowing G1/S progression. at Thr187 is essential for this process, as mutants lacking this site resist SCF<sup>Skp2</sup>-dependent degradation. Phosphorylation at specific sites modulates p27<sup>Kip1</sup> activity and localization. 88 (Tyr88) phosphorylation by kinases such as or JAK2 disrupts p27<sup>Kip1</sup> binding to CDK2, impairing its inhibitory function and promoting progression; this modification also enhances ubiquitination and degradation. Similarly, serine 10 (Ser10) phosphorylation by AKT kinase promotes binding to 14-3-3 proteins, leading to cytoplasmic retention of p27<sup>Kip1</sup> and sequestration from nuclear CDKs. Other modifications further fine-tune p27<sup>Kip1</sup> stability and localization. of residues, such as at position 100, prevents ubiquitination and stabilizes the protein, counteracting pathways and sustaining its tumor-suppressive role. Sumoylation, mediated by UBE2I/ machinery, enhances nuclear retention by inhibiting CRM1-dependent export, thereby preserving p27<sup>Kip1</sup>'s nuclear CDK-inhibitory activity in response to signals like TGF-β. These modifications collectively ensure precise control of p27<sup>Kip1</sup> levels and function across phases.

MicroRNA and Other Non-Coding RNA Regulation

MicroRNAs (miRNAs) play a critical role in post-transcriptional regulation of CDKN1B, primarily by binding to its 3' untranslated region (3'UTR) to suppress translation or induce mRNA degradation, thereby reducing p27^Kip1 protein levels and promoting cell cycle progression in various cancers. Among these, the miR-221/222 cluster is a well-established oncogenic regulator that directly targets the CDKN1B 3'UTR, leading to decreased p27^Kip1 expression. This mechanism contributes to uncontrolled proliferation in breast cancer, where miR-221/222 overexpression correlates with estrogen receptor signaling and enhanced tumor growth, as well as in thyroid papillary carcinomas, where elevated levels of these miRNAs are associated with aggressive disease phenotypes. Other miRNAs, such as miR-16 and miR-106b, further contribute to CDKN1B suppression by inhibiting its translation, often in a cancer-specific context. MiR-16, frequently downregulated in cells under certain conditions, targets cell cycle regulators including components that indirectly stabilize p27^Kip1, but its enforced expression can suppress proliferation by modulating pathways. MiR-106b, part of the miR-106b9325 cluster, directly binds the CDKN1B transcript to repress p27^Kip1 synthesis, facilitating in gastric cancer cells where cluster overexpression is common. In contrast, members of the let-7 family exhibit tumor-suppressive effects by enhancing CDKN1B mRNA stability through indirect mechanisms, such as repressing factors that promote p27^Kip1 degradation, thereby sustaining cell cycle arrest in and other epithelial cancers. Long non-coding RNAs (lncRNAs) modulate CDKN1B expression through competitive endogenous RNA (ceRNA) networks, often by sponging miRNAs that target p27^Kip1. lncRNA GAS5 sequesters miR-222, relieving repression on CDKN1B and elevating p27^Kip1 to suppress hepatic fibrogenesis and tumor growth. These interactions highlight lncRNAs as key mediators of miRNA availability for CDKN1B regulation. Circular RNAs (circRNAs), a class of stable non-coding RNAs, are emerging as regulators of CDKN1B via miRNA . CircRNAs such as circ-YAP1 sponge miR-367-5p to derepress CDKN1B , reducing gastric by increasing p27^Kip1 abundance. In , circCRKL similarly acts as a miR-196a-5p/b-5p decoy, enhancing p27^Kip1-mediated . These underscore the potential of circRNAs as therapeutic targets for restoring p27^Kip1 function in .

Role in Cancer Pathogenesis

Suppression of Cell Proliferation

CDKN1B, encoding the inhibitor p27<sup>Kip1</sup>, exerts a tumor-suppressive effect by restraining , primarily through regulation of the G1/S transition. In p27<sup>Kip1</sup> models, ablation of the gene results in uncontrolled progression from G1 to , leading to multi-organ , including enlarged pituitary glands, adrenal medullas, and gonads, as well as increased body size due to enhanced cellular proliferation. This phenotype underscores p27<sup>Kip1</sup>'s essential role in maintaining proliferative , with heterozygous mice also showing predisposition to pituitary adenomas, further highlighting dosage-dependent suppression of aberrant growth. An inverse correlation exists between p27<sup>Kip1</sup> expression levels and tumor aggressiveness across various malignancies. High p27<sup>Kip1</sup> expression is typically observed in benign tumors, such as nevi in , where it effectively curbs proliferation, whereas low or heterogeneous expression predominates in aggressive malignant tumors, correlating with poorer and increased proliferative activity. Beyond its canonical inhibition of cyclin-CDK complexes, p27<sup>Kip1</sup> contributes to suppression of cell proliferation by promoting cellular senescence via the retinoblastoma (Rb) pathway. Accumulation of p27<sup>Kip1</sup> is necessary for Rb-mediated induction of senescence, as its depletion abrogates Rb's ability to enforce cell cycle arrest and maintain a senescent state, thereby linking CDK-independent functions to long-term proliferative control. A pan-cancer demonstrated CDKN1B downregulation in multiple tumor types, with protein expression reduced in approximately 60% of cancers, fostering signatures and adverse clinical outcomes.

Influence on Tumor Metastasis

The localization of p27<sup>Kip1</sup> (encoded by CDKN1B) within cancer cells critically influences tumor cell motility and invasion, with distinct roles for its cytoplasmic and nuclear forms. In the , p27<sup>Kip1</sup> promotes independent of its canonical (CDK) inhibitory function by binding directly to RhoA, a that regulates dynamics. This interaction interferes with guanine nucleotide exchange factors (GEFs), preventing RhoA activation and GDP/GTP cycling, which paradoxically enhances migratory dynamics in tumor cells such as fibroblasts and lines. Studies in p27<sup>Kip1</sup>-null models demonstrate reduced motility upon loss of this cytoplasmic function, underscoring its pro-invasive role in tumor progression. Conversely, nuclear p27<sup>Kip1</sup> acts as a transcriptional repressor that suppresses epithelial-mesenchymal transition (EMT), a key process enabling metastasis, by directly associating with the promoters of EMT-inducing transcription factors. Specifically, p27<sup>Kip1</sup> binds to the Twist1 promoter in a p130/E2F4-dependent manner, repressing its transcription and preventing the downregulation of epithelial markers like E-cadherin. This mechanism also extends to inhibiting Snail expression through similar repressive complexes, thereby maintaining epithelial integrity and limiting invasive potential in contexts like embryonic stem cell differentiation and early tumor stages. Loss of nuclear p27<sup>Kip1</sup> leads to Twist1 upregulation and EMT-like morphological changes, facilitating dissemination. Clinically, reduced nuclear p27<sup>Kip1</sup> expression correlates with increased metastatic risk, particularly in , where low levels are inversely associated with involvement and poorer . Meta-analyses of patient cohorts confirm that diminished nuclear p27<sup>Kip1</sup> independently predicts higher rates of nodal and reduced overall survival, highlighting its utility as a for aggressive disease. Recent investigations into circulating tumor cells (CTCs) reveal that p27<sup>Kip1</sup> modulates their invasive potential by promoting a drug-tolerant persister state. In breast cancer CTC cultures exposed to mitotic inhibitors, elevated p27<sup>Kip1</sup> restricts and endomitosis via signaling, enabling reversible quiescence that enhances survival and regrowth capacity post-therapy. This adaptation increases CTC and metastatic potential, as evidenced by 2024–2025 studies showing p27<sup>Kip1</sup>-dependent in ≤4N states.

Dysregulation in Specific Cancer Types

In , particularly the luminal subtypes, CDKN1B mutations and reduced p27 expression contribute to disease progression. The V109G polymorphism has been identified in luminal breast tumors, where it impairs p27 protein stability and correlates with aggressive phenotypes. A 2025 analysis of luminal-type cohorts demonstrated that low CDKN1B expression is significantly associated with reduced metastasis-free survival, highlighting its role in sustaining uncontrolled proliferation in these hormone receptor-positive tumors. In , dysregulation of CDKN1B often involves post-translational mechanisms rather than frequent genetic alterations. Overexpression of the ubiquitin ligase Skp2 promotes ubiquitin-mediated degradation of p27, leading to diminished CDKN1B activity and enhanced progression in androgen-dependent and castration-resistant tumors. Promoter hypermethylation of CDKN1B has been observed infrequently, contributing to epigenetic in a subset of cases, though this mechanism is less prevalent compared to Skp2-driven proteolysis. Germline mutations in CDKN1B define multiple endocrine neoplasia type 4 (MEN4), a hereditary characterized by endocrine tumors including pituitary adenomas. Nonsense variants, which result in truncated p27 proteins lacking functional domains, disrupt cell cycle inhibition and predispose carriers to pituitary tumorigenesis. A 2025 characterization of CDKN1B variants in MEN4 patients revealed that these truncating mutations abolish p27's ability to bind cyclin-CDK complexes, thereby accelerating pituitary and tumor formation. Emerging evidence points to CDKN1B alterations in other malignancies, including lung cancer (SCLC). In SCLC, where RB1 and TP53 losses already compromise the G1/S checkpoint, concurrent CDKN1B dysregulation—often through reduced expression—further exacerbates defects, promoting rapid tumor growth. Pan-cancer analyses indicate that CDKN1B hypermutation rates are generally low (typically below 3-4%), with higher frequencies observed in uterine and neuroendocrine tumors, where frameshift and missense variants predominate.

Clinical and Therapeutic Implications

Prognostic and Diagnostic Value

CDKN1B, encoding the inhibitor p27<sup>Kip1</sup>, serves as a valuable for assessing cancer through immunohistochemical evaluation of protein expression. Low nuclear p27<sup>Kip1</sup> expression is associated with poor in , correlating with aggressive disease and reduced survival. Similarly, in , low nuclear p27<sup>Kip1</sup> expression correlates with shorter recurrence-free survival and increased risk of progression. These findings underscore the utility of standardized IHC scoring systems, where low nuclear expression often signals heightened tumor aggressiveness and poorer outcomes. At the transcriptional level, reduced CDKN1B mRNA expression in tumor tissues predicts adverse prognosis across multiple cancers, as evidenced by analyses of (TCGA) datasets. For instance, in cohorts from TCGA, low CDKN1B mRNA levels are linked to shorter overall survival, with hazard ratios (HR) typically ranging from 1.5 to 2.0 in multivariate models adjusting for age, stage, and subtype. This pattern holds in other solid tumors, where diminished CDKN1B transcripts associate with increased tumor burden and metastatic potential, highlighting its potential as a non-invasive prognostic indicator via RNA-based assays. In the context of hereditary syndromes, germline variants in CDKN1B are diagnostic hallmarks of type 4 (MEN4), a condition predisposing individuals to endocrine tumors such as pituitary adenomas and parathyroid carcinomas. Molecular for CDKN1B mutations, including sequencing of coding exons, is recommended for screening in patients with familial or sporadic endocrine neoplasms suggestive of MEN syndromes, enabling early detection and surveillance. Identification of pathogenic variants, such as frameshift or missense alterations, confirms MEN4 diagnosis and guides clinical management, distinguishing it from MEN1. Recent pan-cancer analyses using TCGA data have reinforced CDKN1B's role as an independent prognostic factor. A 2023 study across 40 cancer types found variable CDKN1B expression, with high levels correlating to favorable outcomes in entities like kidney renal clear cell carcinoma (KIRC) and cholangiocarcinoma (CHOL), emphasizing its broad applicability in risk stratification.

Association with Treatment Response

In hormone receptor-positive (HR+) , high levels of phosphorylated (specifically at tyrosine-88) correlate with enhanced responsiveness to CDK4/6 inhibitors such as . This phosphorylation status reduces CDK4 activity and stratifies sensitive tumors in explant cultures, where pY88-positive samples exhibit significant decreases in Ki-67-positive cells following treatment, unlike pY88-negative counterparts. Conversely, decreased total p27<sup>Kip1</sup> levels in resistant cell lines, like palbociclib-resistant variants, are associated with upregulated CDK2 activity, underscoring p27<sup>Kip1</sup>'s role in maintaining sensitivity through inhibition. Regarding mitotic inhibitors like , used in , low p27<sup>Kip1</sup> expression promotes (≥8N DNA content) as an escape mechanism from mitotic arrest, potentially contributing to by allowing through endomitosis, though this often leads to without regrowth in circulating tumor cells (CTCs). In contrast, high p27<sup>Kip1</sup> stabilizes via AKT-mediated serine-10 , restricting and enabling a reversible drug-tolerant persister (DTP) state limited to ≤4N ploidy, which supports CTC regrowth post-treatment. Suppression of p27<sup>Kip1</sup> in persister-proficient CTCs shifts them toward and abrogates recovery, highlighting its dual role in modulating mitotic escape pathways. In , restoration of p27<sup>Kip1</sup> enhances the efficacy of (ADT) by mediating therapy-induced through the p27<sup>Kip1</sup>/CDK/pRb pathway, promoting G1/S arrest and inhibiting proliferation. ADT upregulates p27<sup>Kip1</sup> expression, leading to in androgen-dependent cells like and LAPC-4, with over 80% exhibiting senescent markers after 10 days of treatment; high p27<sup>Kip1</sup> levels, coupled with Skp2 downregulation, amplify this response and counteract progression in xenografts. This mechanism positions p27<sup>Kip1</sup> restoration as a sensitizer to ADT, improving outcomes by enforcing permanent growth arrest. Recent studies (2024–2025) further elucidate CDKN1B's involvement in drug-tolerant persister states following mitotic drug exposure, particularly in CTCs. Stabilized p27<sup>Kip1</sup> facilitates DTP emergence by preventing excessive , allowing reversible tolerance to without permanent resistance; this was observed in 12 of 18 patient-derived CTC cultures, where p27<sup>Kip1</sup> knockdown increased lethality via unchecked endomitosis. These findings suggest targeting p27<sup>Kip1</sup> stabilization could disrupt persister formation and improve outcomes.

Strategies for Therapeutic Targeting

One key strategy for therapeutic targeting of CDKN1B (encoding p27<sup>Kip1</sup>) involves stabilizing the protein to prevent its degradation, thereby enhancing its inhibitory function in s. Skp2, a component of the SCF<sup>Skp2</sup> complex, promotes p27 ubiquitination and proteasomal degradation, and its inhibition has shown promise in preclinical models. For instance, the small-molecule inhibitor SZL-P1-41 disrupts the Skp2-Skp1 interaction, selectively suppressing SCF<sup>Skp2</sup> activity without affecting other cullin-RING ligases, leading to p27 accumulation and reduced proliferation in various lines. This approach has demonstrated anti-tumor effects in xenograft models, including decreased tumor growth in by elevating p27 levels and inhibiting downstream activity. Gene therapy approaches using viral vectors to deliver CDKN1B have been explored in preclinical cancer models to restore p27 expression and suppress tumor progression. Adenoviral vectors expressing p27<sup>Kip1</sup> (Ad-p27) have been shown to inhibit proliferation in cell lines by inducing G1 arrest and , with no significant toxicity to normal cells. In vivo studies using these vectors in subcutaneous tumor models further confirmed reduced tumor volume and increased p27 protein levels, highlighting the potential for targeted delivery to overcome p27 downregulation in aggressive malignancies. Modulating microRNA regulation of CDKN1B represents another targeted intervention, particularly through antagomirs that inhibit oncogenic miRNAs. In glioblastoma, miR-221/222 directly suppress p27 translation, contributing to uncontrolled proliferation; antagomirs against these miRNAs restore p27 expression, inducing and sensitizing cells to chemotherapy in preclinical models. This strategy has shown efficacy in reducing tumor growth in orthotopic glioblastoma xenografts by elevating intratumoral p27 levels and disrupting progression. Recent innovations as of 2025 focus on upstream pathway modulation to leverage CDKN1B in specific cancers. In small cell lung cancer (SCLC), which often exhibits G1-S checkpoint defects including low p27 activity due to high signaling, dual inhibitors of A and RxL (e.g., CIRc series) selectively induce lethality in SCLC cells by disrupting A-1 interactions and other RxL bindings, leading to replication stress and in tumors with RB1/TP53 alterations. As of November 2025, a Phase 1 (NCT06577987) is evaluating CID-078, a related RxL , in advanced solid tumors including SCLC. For (CTC) persisters, which survive through p27 stabilization via AKT at serine 10, modulation strategies aim to disrupt this process; preclinical data indicate that enhancing p27 function via stabilizers can influence persister , though suppression reduces CTC regrowth in patient-derived models post-mitotic exposure.

Role in Tissue Regeneration

Mechanisms in Cellular Reprogramming

p27<sup>Kip1</sup>, the protein encoded by CDKN1B, plays a critical role in regulating cellular reprogramming by inhibiting (CDK2) activity, which prevents premature entry into the in progenitor cells. This inhibition maintains quiescence and ensures proper timing of progression during developmental and regenerative processes, such as in retinal histogenesis where p27<sup>Kip1</sup> constrains excessive of retinal progenitors to support ordered . In the context of reprogramming, this mechanism helps preserve progenitor identity by blocking unscheduled that could disrupt cell fate decisions. In retinal repair, p27<sup>Kip1</sup> contributes to the quiescence of , the primary glial cells in the , and works alongside p21<sup>Cip1</sup> (encoded by CDKN1A) as part of the Cip/Kip family of CDK inhibitors to limit and proliferation following injury. In mammalian models, such as those involving N-methyl-N-nitrosourea (MNU)-induced photoreceptor loss, upregulation of p21<sup>Cip1</sup> modulates proteins to facilitate limited , while in sodium iodate (NaIO<sub>3</sub>)-induced damage, downregulation of p27<sup>Kip1</sup> via signaling promotes re-entry into the and neuronal differentiation. This combined regulatory action underscores their shared function in balancing quiescence against regenerative potential in mammals, where robust reprogramming is restrained compared to lower vertebrates like . During liver regeneration, downregulation of p27<sup>Kip1</sup> is essential for enabling proliferation after partial . Following , p27<sup>Kip1</sup> levels decrease rapidly, allowing CDK activation and re-entry in quiescent hepatocytes to restore tissue mass. This transient suppression is mediated by factors such as microRNA-221, which inhibits p27<sup>Kip1</sup> translation, thereby promoting S-phase progression and regenerative expansion without leading to uncontrolled growth. A recent study demonstrated that combining overexpression with p27<sup>Kip1</sup> knockdown via (AAV) vectors induces robust proliferation of in uninjured mammalian retinas, mimicking regenerative observed in non-mammals. This approach resulted in approximately 45% of re-entering the , with self-limiting divisions and evidence of neurogenic potential in daughter cells, highlighting p27<sup>Kip1</sup> as a key barrier to mammalian retinal regeneration.

Applications in Regenerative Medicine

In vitro studies on neonatal cardiomyocytes demonstrate that siRNA-mediated knockdown of p27<sup>Kip1</sup>, in combination with other CDK inhibitors like p21<sup>Cip1</sup>, induces re-entry and proliferation, suggesting potential for enhancing cardiac repair in models of myocardial without long-term adverse effects. This approach promotes the generation of new cardiomyocytes from resident cell populations, as demonstrated by enhanced BrdU incorporation and mitotic indices. Similarly, targeting p27<sup>Kip1</sup> in adult cardiac stem cells via genetic knockdown augments their self-renewal and differentiation potential, contributing to better functional recovery in ischemic models. In neural regeneration, inhibition of p27<sup>Kip1</sup> facilitates the of progenitor cells (OPCs), which is crucial for remyelination and repair in demyelinating injuries. Studies in lesion models reveal that reducing p27<sup>Kip1</sup> levels through or genetic manipulation increases OPC recruitment to the injury site, boosting their and subsequent into mature . This modulation enhances the number of myelinating cells, as evidenced by greater NG2-positive OPC density and improved axonal remyelination in p27<sup>Kip1</sup>-deficient conditions compared to controls. Such strategies hold potential for treating conditions like , where OPC exhaustion limits endogenous repair. Restoring p27<sup>Kip1</sup> expression in senescent tissues offers a means to rebalance quiescence in compartments affected by age-related diseases. In aged s, activation of the AMPK/p27<sup>Kip1</sup> pathway via pharmacological or genetic means promotes while suppressing and markers, such as SA-β-gal activity and <sup>INK4a</sup> levels. This restoration maintains stem cell quiescence, preventing exhaustion and supporting tissue in models of . Overexpression of p27<sup>Kip1</sup> in these contexts similarly shifts aged cells toward a reversible quiescent state, enhancing their regenerative potential without inducing permanent arrest. In auditory regeneration, inhibition of p27<sup>Kip1</sup> promotes the and regeneration of cochlear in mammalian models. Auditory -specific deletion of p27<sup>Kip1</sup> in postnatal mice enables supporting cells to re-enter the , generating new and preserving normal hearing function. This approach highlights p27<sup>Kip1</sup> as a barrier to inner ear repair, with potential therapeutic applications for caused by damage.

Protein Interactions

Core Interacting Partners

CDKN1B, also known as p27<sup>Kip1</sup>, primarily interacts with (CDK) complexes through its N-terminal (KID), which encompasses distinct motifs for and CDKs. This includes an RxL (D1) for , a hydrophobic α-helix (LH) for initial docking, and a β-hairpin/3<sub>10</sub>-helix (D2) that inserts into the CDK to inhibit ATP and activity. Specifically, p27<sup>Kip1</sup> binds E-CDK2 and A-CDK2 with high , forming inhibitory complexes that block G1/S progression; the of p27<sup>Kip1</sup>/ A/CDK2 reveals p27<sup>Kip1</sup> as an extended polypeptide spanning both subunits, with residues like Arg194 forming hydrogen bonds to Glu42 on CDK2 and Pro272 on A. In contrast, to (D1, D2, or D3)-CDK4/6 complexes can promote their assembly at low p27<sup>Kip1</sup> levels while inhibiting at higher concentrations, with at Tyr74 reducing and facilitating activation. p27<sup>Kip1</sup> degradation is mediated by E3 ubiquitin ligases, including Skp2 (part of the SCF<sup>Skp2</sup> complex) and the KPC complex (KPC1/KPC2). Skp2 recognizes p27<sup>Kip1</sup> phosphorylated at Thr187 (primarily by E-CDK2), recruiting it for polyubiquitination and proteasomal degradation during ; this interaction requires the adaptor Cks1 and occurs in the . The KPC complex targets cytoplasmic p27<sup>Kip1</sup> in early following nuclear export, ubiquitinating it independently of Thr187 phosphorylation but dependent on the cullin-RING architecture. Signaling kinases such as AKT and GSK3β regulate p27<sup>Kip1</sup> stability and localization through at specific sites. AKT directly p27<sup>Kip1</sup> at Thr157 (and to a lesser extent Thr198), impairing its nuclear import by promoting 14-3-3 binding and cytosolic sequestration, thereby reducing CDK inhibition. GSK3β p27<sup>Kip1</sup> at sites including Ser160 and Ser161 (in a pathway often primed by prior AKT inhibition) and contributes to its inactivation or degradation in response to signaling. Additional core partners include JAB1 (CSN5), which binds the C-terminal domain of p27<sup>Kip1</sup> to facilitate its CRM1-dependent nuclear export, exposing it to cytoplasmic degradation pathways, and stathmin (STMN1), which interacts with the cyclin-binding domain of p27<sup>Kip1</sup> to inhibit stathmin's microtubule-depolymerizing activity, thereby stabilizing and modulating in non-proliferative contexts. Beyond CDK-related interactions, p27<sup>Kip1</sup> engages with non-cell cycle proteins to influence migration and transcription. It binds RhoA via its N-terminal domain, sequestering it from its effectors and inhibiting actomyosin contractility to suppress cell motility. Additionally, p27<sup>Kip1</sup> interacts with the transcription factor c-Jun in the nucleus, repressing AP-1 activity and thereby modulating related to and .

Functional Networks and Pathways

The inhibitor p27<sup>Kip1</sup>, encoded by CDKN1B, integrates into several key signaling networks that regulate progression, growth arrest, and stress responses. In the PI3K/AKT pathway, stimulation activates PI3K, leading to AKT-mediated of p27<sup>Kip1</sup> at 157 (T157) and 198 (T198), which promotes its cytoplasmic sequestration and reduces its nuclear inhibitory activity on cyclin E-CDK2 complexes, thereby facilitating G1/S transition and . This mechanism links mitogenic signals to entry, with dysregulation often observed in oncogenic contexts where hyperactive AKT impairs p27<sup>Kip1</sup>'s tumor-suppressive role. In the TGF-β signaling pathway, TGF-β ligands induce receptor activation, recruiting Smad2/3 proteins that form a complex with Smad4 to translocate to the nucleus and upregulate CDKN1B transcription, thereby elevating p27<sup>Kip1</sup> levels and enforcing G1 arrest by inhibiting CDK2 activity. Additionally, TGF-β stabilizes p27<sup>Kip1</sup> protein through Smad-dependent inhibition of proteasomal degradation, enhancing its association with cyclin-CDK complexes to mediate cytostatic responses in epithelial and mesenchymal cells. This network underscores p27<sup>Kip1</sup>'s role in TGF-β-induced growth suppression. The DNA damage response pathway positions p27<sup>Kip1</sup> downstream of the axis, where DNA lesions activate /ATR kinases that stabilize , indirectly promoting p27<sup>Kip1</sup> accumulation to reinforce G1 checkpoint arrest after initial p21-mediated inhibition. directly phosphorylates p27<sup>Kip1</sup> at serine 140, enhancing its stability and CDK-inhibitory function, which is essential for sustained blockade in p53-proficient cells and prevents progression of damaged genomes. Recent investigations (2023–2025) have revealed p27<sup>Kip1</sup>'s integration into networks controlling in drug-tolerant persister cells, where AKT1-mediated at serine 10 stabilizes p27<sup>Kip1</sup>, restricting endomitosis and limiting ploidy to ≤4N following mitotic inhibitors like , thereby enabling reversible in circulating tumor cells. In the context of type 4 (MEN4), germline CDKN1B mutations disrupt p27<sup>Kip1</sup>'s function within endocrine signaling networks, impairing its inhibition of cyclin-CDK complexes in pituitary and parathyroid cells and leading to through dysregulated G1/S progression linked to menin-dependent transcriptional control.

References

  1. [1]
    CDKN1B cyclin dependent kinase inhibitor 1B [ (human)] - NCBI
    Aug 19, 2025 · CDKN1B encodes a cyclin-dependent kinase inhibitor that controls cell cycle progression at G1 by preventing the activation of cyclin E-CDK2 or ...
  2. [2]
    600778 - CYCLIN-DEPENDENT KINASE INHIBITOR 1B; CDKN1B
    CDKN1B, or p27(KIP1), is a cyclin-dependent kinase inhibitor that blocks the cell cycle in the G0/G1 phase upon differentiation signals or cellular insult.
  3. [3]
    p27Kip1, an Intrinsically Unstructured Protein with Scaffold Properties
    p27 Kip1 is a gatekeeper of G1/S transition. It also regulates G2/M progression and cytokinesis completion, via CDK-dependent or -independent mechanisms.
  4. [4]
    Gene: CDKN1B (ENSG00000111276) - Summary - Homo_sapiens
    Chromosome 12: 12,685,498-12,722,373 forward strand. This gene has 10 transcripts (splice variants), 219 orthologues, 2 paralogues and is associated with 97 ...Missing: size | Show results with:size
  5. [5]
    Role of p27Kip1 as a transcriptional regulator - PMC - NIH
    May 25, 2018 · (A) The N-terminal domain of p27 includes the kinase inhibitory domain (KID) that consists of three subdomains: the cyclin-binding subdomain (D1) ...P27 Associates With... · P27 Regulates Transcription... · Figure 5. Model Of The Role...
  6. [6]
    p27kip1 at the crossroad between actin and microtubule dynamics
    Apr 1, 2019 · With regard to p27, its subcellular localization is mediated by two different domains, namely the Nuclear Export Signal (NES) and the Nuclear ...
  7. [7]
    Crystal structure of the p27Kip1 cyclin-dependent-kinase inibitor ...
    Jul 25, 1996 · Crystal structure of the p27Kip1 cyclin-dependent-kinase inibitor bound to the cyclin A–Cdk2 complex. Alicia A. Russo,; Philip D. Jeffrey, ...
  8. [8]
    p27Kip1 Inhibits Cyclin D-Cyclin-Dependent Kinase 4 by Two ... - NIH
    The two modes by which p27 inhibits cyclin D-cdk4 are independent and may reinforce one another to inhibit kinase activity in contact-arrested cells.
  9. [9]
    Regulation of Exit from Quiescence by p27 and Cyclin D1-CDK4 - NIH
    Indeed, the accumulation of p27 is required for cells to efficiently exit from the cell cycle and enter a quiescent state (12, 72). Conversely, p21 and p27 may ...
  10. [10]
    p27 binds cyclin–CDK complexes through a sequential mechanism ...
    Mar 14, 2004 · p27 binds cyclin–CDK complexes through a sequential mechanism involving binding-induced protein folding ... DOI : https://doi.org/10.1038/nsmb746 ...
  11. [11]
    The p21 Cip1 and p27 Kip1 CDK 'inhibitors' are essential activators ...
    The Cip/Kip proteins interact with a variety of cyclin–CDK complexes through a conserved N‐terminal domain that contains both cyclin and CDK binding sites ( ...
  12. [12]
    c-Myc represses FOXO3a-mediated transcription of the gene ...
    Induction of p27 (CDKN1B) gene transcription by Forkhead box O proteins such as FOXO3a leads to growth arrest and apoptosis. Previously, we observed that B cell ...
  13. [13]
    MYC Oncogene Contributions to Release of Cell Cycle Brakes - PMC
    At least in some cells types, MYC inhibits the transcription of the p27 gene (CDKN1B) but also enhances p27's degradation through the upregulation of components ...
  14. [14]
    Multiplexed profiling of candidate genes for CpG island methylation ...
    DNA methylation in CpG islands is associated with transcriptional silencing ... CpG methylation is a gene-specific and nonrandom event in colon cancer.
  15. [15]
    Epigenetic silencing of tumor suppressor genes - ScienceDirect.com
    Epigenetic silencing of TSGs, an early cancer event, involves mechanisms like DNA methylation, and is a key factor in the oncogenic process.4. Loss Of Ctcf Binding... · 6. Non-Coding Rnas · 7.2. Ezh2 Inhibitors
  16. [16]
    p27kip1: A Multifunctional Cyclin-Dependent Kinase Inhibitor ... - NIH
    This explains how p27 and other Kip/Cip inhibitors can bind isolated subunits. 15,24 Binding of the p27 cyclin-CDK complex is significantly tighter than binding ...Missing: NLS | Show results with:NLS
  17. [17]
    CRM1/Ran-Mediated Nuclear Export of p27 Kip1 Involves a Nuclear ...
    SENP1 promotes p27kip1 nuclear export though enhanced SUMOylation in cholangiocarcinoma leading to increased cell proliferation and chemoresistance. 11 July ...
  18. [18]
    p27(Kip1) ubiquitination and degradation is regulated by the SCF ...
    Phosphorylation of p27 at Thr187 was essential for its degradation. Degradation was also dependent on SCF(Skp2), a protein complex implicated in targeting ...
  19. [19]
    p27Kip1 ubiquitination and degradation is regulated by the ...
    Phosphorylation of p27 at Thr187 was essential for its degradation. Degradation was also dependent on SCFSkp2, a protein complex implicated in targeting ...
  20. [20]
    Phosphorylation of p27Kip1 by JAK2 directly links cytokine receptor ...
    Aug 11, 2011 · JAK2 phosphorylates tyrosine residue 88 (Y88) of p27(Kip1). We previously reported that Y88 phosphorylation of p27(Kip1) by oncogenic tyrosine ...Missing: Tyr88 Src
  21. [21]
    Akt-dependent phosphorylation of p27Kip1 promotes binding to 14 ...
    Akt promotes cell-cycle progression through the mechanisms of phosphorylation-dependent 14-3-3 binding to p27(Kip1) and cytoplasmic localization.
  22. [22]
    SIRT6 delays cellular senescence by promoting p27Kip1 ubiquitin ...
    These results demonstrate that acetylation of p27 prevents its ubiquitination and stabilizes the p27 protein. Figure 6. Acetylated-p27 is more stable.
  23. [23]
    SUMOylation regulates p27Kip1 stability and localization in ...
    Oct 8, 2015 · We observed that SUMOylation regulated p27Kip1 binding to CDK2, thereby governing its nuclear proteasomal degradation through the ...
  24. [24]
    SUMOylation regulates p27Kip1 stability and localization in ...
    Oct 8, 2015 · These data indicate that SUMOylation is a novel regulatory mechanism that modulates p27(Kip1) function in response to TGFβ stimulation.
  25. [25]
    MicroRNA Cluster 221-222 and Estrogen Receptor α Interactions in ...
    These findings suggest that the negative regulatory loop involving miR-221-222 and ERα may confer proliferative advantage and migratory activity to breast ...Microrna Cluster 221-222 And... · Results · Erα Regulation Of...
  26. [26]
    221 and miR-222, both overexpressed in human thyroid papillary ...
    Sep 1, 2007 · Here, we report that the enforced expression of miR-221 and miR-222 was able to reduce p27Kip1 protein levels in thyroid carcinoma and HeLa ...
  27. [27]
    Thyroglobulin Increases Thyroid Cell Proliferation via the ... - NIH
    We identified 21 miRNAs whose expression was significantly suppressed by Tg in rat thyroid FRTL-5 cells. Using specific miRNA analogs, we determined that miR-16 ...
  28. [28]
    suppression of cell-cycle inhibitors by microRNA clusters in gastric ...
    Here we report that the miRNAs in two clusters (miR-106b approximately 93 approximately 25 and miR-222 approximately 221) suppress the Cip/Kip family members ...
  29. [29]
  30. [30]
    Article Mice Lacking p27 Kip1 Display Increased Body Size, Multiple ...
    Mice lacking p27Kip1 have been created by gene targeting in embryonic stem cells. These mice are larger than the control animals, with thymus, pituitary, ...
  31. [31]
    Mice Lacking p27Kip1 Display Increased Body Size, Multiple Organ ...
    Histological examination of the adrenal gland showed that testis and ovary were about 2-fold larger in p27J/J marked hyperplasia of the medulla in p27J/J mice,.
  32. [32]
    Protein Expression of the Cell-Cycle Inhibitor p27Kip1 in Malignant ...
    We found that p27Kip1 was strongly expressed by normal melanocytes and benign nevi, whereas in malignant melanoma, a heterogeneous expression pattern was ...
  33. [33]
    Requirement for p27KIP1 in Retinoblastoma Protein-Mediated ... - NIH
    p27KIP1 accumulation is critical for pRb-induced senescence, and its ablation abrogates cell cycle arrest. p27KIP1 is critical for maintaining cell cycle ...
  34. [34]
    Biomedical Reports - Spandidos Publications
    Oct 4, 2023 · CDKN1B protein expression is reduced in ~60% of human cancer cases which is indicative of poor prognosis and chemotherapy resistance (101,102).
  35. [35]
    A pan-cancer analysis for the oncogenic role of cyclin-dependent ...
    Jul 11, 2023 · The CDKN1B gene encodes the p27Kip1 protein, which can block the production cyclin-dependent kinase (CDK), which is obviously related to the ...
  36. [36]
  37. [37]
    Prognostic Significance of p27 kip-1 Expression in Colorectal ...
    p27 cell-cycle inhibitor is inversely correlated with lymph node metastases in right-sided colon cancer. ... Prognostic significance of p27Kip1 expression in ...
  38. [38]
    The V109G polymorphism of the p27 gene CDKN1B indicates a ...
    The V109G polymorphism of the p27 gene CDKN1B indicates a worse outcome in node-negative breast cancer patients. Tumour Biol. Sep-Dec 2004;25(5-6):306-12 ...Missing: dysregulation missense mutations stability
  39. [39]
    Characterization of CDKN1B Variants Identified in MEN4 and Breast ...
    Jan 26, 2025 · Several germline or somatic CDKN1B (the p27Kip1 encoding gene) mutations have been demonstrated to be associated with multiple endocrine ...Missing: alternative splicing isoforms v1
  40. [40]
    p27 Cell Cycle Inhibitor and Survival in Luminal-Type Breast Cancer
    In GSE11121 and GSE25065, low CDKN1B expression was significantly correlated with metastasis-free survival (p = 0.043 and p = 0.002, respectively) ( ...
  41. [41]
    Deregulated proteolysis by the F-box proteins SKP2 and β-TrCP
    Expression of SKP2 was observed to increase with androgen addition in human prostate cells, leading to an increase in p27 degradation and increased ...
  42. [42]
    The impact of Skp2 overexpression on recurrence-free survival ...
    In several human cancers, overexpression of Skp2 (S-phase kinase associated protein 2), which targets p27 for degradation, portends a poorer prognosis.
  43. [43]
    Epigenetic Changes in Prostate Cancer: Implication for Diagnosis ...
    ... CDKN1B by hypermethylation is rare in prostate cancer ... Silencing of CD44 expression in prostate cancer by hypermethylation of the CD44 promoter region.
  44. [44]
    Germline CDKN1B Loss-of-Function Variants Cause Pediatric ...
    Germline loss-of-function CDKN1B gene variants cause the autosomal dominant syndrome of multiple endocrine neoplasia type 4 (MEN4).
  45. [45]
    p27Kip1 and Tumors: Characterization of CDKN1B Variants ... - MDPI
    Here, we analyzed the effect of four CDKN1B missense and nonsense mutations found in patients affected by MEN4 or cancers.P27 And Tumors... · 3. Results · 3.4. Analysis Of P27...Missing: splicing v1
  46. [46]
    Targeting G1–S-checkpoint-compromised cancers with cyclin A/B ...
    Aug 20, 2025 · Dual inhibitors of cyclin A and cyclin B RxL motifs (cyclin A/Bi) selectively kill SCLC cells and other cancer cells with high E2F activity.
  47. [47]
    Association of p27Kip1 levels with recurrence and survival in ...
    Levels of nuclear p27 immunoreactivity in the primary tumor can be used to predict recurrence and survival among patients with localized prostate cancer.
  48. [48]
    Low CDKN1B Expression Associated with Reduced CD8+ T ...
    Dec 25, 2023 · The cyclin-dependent kinase inhibitor 1B (CDKN1B) gene, which encodes the p27Kip1 protein, is important in regulating the cell cycle process ...1. Introduction · 3. Results · Figure 5
  49. [49]
    Multiple Endocrine Neoplasia Type 4 - GeneReviews - NCBI - NIH
    Sep 21, 2023 · The diagnosis of MEN4 is established in a proband with a germline heterozygous pathogenic variant in CDKN1B identified by molecular genetic ...
  50. [50]
    The ongoing search for biomarkers of CDK4/6 inhibitor ... - NIH
    ... p27Kip1 associates with Palbociclib responsiveness in breast cancer. Cancer ... breast cancer strongly sensitizes tumors to erlotinib and palbociclib ...
  51. [51]
    CDKN1B (p27/kip1) enhances drug-tolerant persister CTCs ... - PNAS
    Hengst, Phosphorylation of p27Kip1 by JAK2 directly links cytokine receptor signaling to cell cycle control. Oncogene 30, 3502–3512 (2011). Crossref · PubMed.
  52. [52]
    Distinct mechanisms mediating therapy-induced cellular ...
    Dec 15, 2022 · Mechanistically, ADT-induced senescence is partially mediated by the cyclin-dependent kinase inhibitor p27Kip1, which might depend on Skp2 ( ...<|separator|>
  53. [53]
    Small-molecule compounds inhibiting S-phase kinase-associated ...
    Apr 5, 2023 · Compound SZL-P1-41, also known as #25, has been reported as a small structure-based inhibitor of Skp2. Nineteen residues within the Skp2 protein ...
  54. [54]
    F-Box Proteins and Cancer - MDPI
    Several inhibitors of SKP2 have been developed [53]. Compound 25 (also known as SZL-P1–41), for example, disrupts the interaction between SKP2 and SKP1 and ...
  55. [55]
    Adenoviral Expression of the Cyclin-Dependent Kinase Inhibitor p27 ...
    Therefore, using cyclindependent kinase inhibitors for gene therapy of cancer is intuitively attractive because it has the potential to inhibit tumor growth ( 3 ) ...
  56. [56]
    Role of microRNAs in glioblastoma - Oncotarget
    Aug 17, 2021 · Treatment with antagomirs against miR-221/222 has been found to induce apoptosis and sensitize human glioblastoma cells to TMZ and to ...Abstract · Introduction · Gbm-Related Micrornas
  57. [57]
    miR221/222 in Cancer: Their Role in Tumor Progression and ... - NIH
    Here we will review the role of miR-221/222 in cancer progression and their use as prognostic and therapeutic tools in cancer.Fig. (1). Microrna... · Mir-221/222 Role In Tumor... · Mir-221/222: Their Role To...
  58. [58]
    p27Kip1 and p57Kip2 Regulate Proliferation in Distinct Retinal ...
    We have found that the p27 Kip1 cyclin kinase inhibitor regulates progenitor cell proliferation throughout retinal histogenesis.Missing: repair dedifferentiation
  59. [59]
    Cellular Signaling in Müller Glia: Progenitor Cells for Regenerative ...
    Mar 19, 2019 · When the retina is damaged, Müller cells can dedifferentiate and proliferate, generated neuronal progenitor cells, migrate to the injured ...2.1. Glaucoma And Nmda · 2.3. 1. Müller Glia In... · 2.3. 2. Müller Glia In...<|separator|>
  60. [60]
    Partial hepatectomy in rats results in immediate down-regulation of ...
    Purpose: The cyclin-dependent kinase (Cdk) inhibitor p27Kip1 may be involved in regulating re-entry of residual hepatocytes into the cell cycle upon loss of ...
  61. [61]
    Dicer-dependent production of microRNA221 in hepatocytes inhibits ...
    In vitro, miR-221 inhibited p27 production in primary hepatocytes and increased hepatocyte proliferation. Specific reconstitution of miR-221 in hepatocyte- ...
  62. [62]
    Simultaneous cyclin D1 overexpression and p27 kip1 knockdown ...
    Apr 3, 2025 · Simultaneous cyclin D1 overexpression and p27kip1 knockdown enable robust Müller glia cell cycle reactivation in uninjured mouse retina.
  63. [63]
    Knockdown of Cyclin-dependent Kinase Inhibitors Induces ...
    p21 and p27 knockdown regulates cells division and induces neonatal cardiomyocyte proliferation 2 days after transfection. Neonatal cardiomyocytes were ...Missing: expansion | Show results with:expansion
  64. [64]
    Cardiac Regeneration and Stem Cells | Physiological Reviews
    Oct 1, 2015 · Targeting inhibitory cell cycle molecules in neonatal or adult ventricular myocytes by knocking down CDK inhibitors p21(Waf1), p27(Kip1) ...
  65. [65]
    Number of oligodendrocyte progenitors recruited to the lesioned ...
    We conclude that the cellular levels of the cell cycle inhibitor p27Kip-1 modulate the repair response of OLPs to injury in the adult spinal cord. Publication ...<|separator|>
  66. [66]
    Number of oligodendrocyte progenitors recruited to the lesioned ...
    Aug 6, 2025 · Recruitment of progenitors to a demyelinated lesion in spinal cord is negatively regulated by Cdk2 inhibitor p27 Kip1 , as more OPCs were found ...<|control11|><|separator|>
  67. [67]
    The AMPK/p27Kip1 Axis Regulates Autophagy/Apoptosis Decisions ...
    Jul 19, 2018 · Acting through AMPK/p27Kip1, we have identified a pathway regulating the balance between autophagy, apoptosis, and senescence in aged MuSCs.
  68. [68]
    The AMPK/p27Kip1 Pathway as a Novel Target to Promote ... - MDPI
    Jun 8, 2021 · Aging promotes nuclear localization of p27Kip1 and a predisposition to senescence or apoptosis. Here, we will review the role of p27Kip1 in ...
  69. [69]
    Müller glia cell cycle re-activation by simultaneous cyclin D1 ...
    Jul 16, 2024 · Müller glia cell cycle re-activation by simultaneous cyclin D1 overexpression and p27kip1 knockdown promotes retinal regeneration in mice.Missing: glaucoma | Show results with:glaucoma
  70. [70]
  71. [71]
    Cloning of p27Kip1, a cyclin-dependent kinase inhibitor ... - PubMed
    We cloned p27Kip1, a cyclin-dependent kinase inhibitor implicated in G1 phase arrest by TGF beta and cell-cell contact.
  72. [72]
  73. [73]
  74. [74]
  75. [75]
  76. [76]
  77. [77]
  78. [78]
  79. [79]
    p27(Kip1)-stathmin interaction influences sarcoma cell migration ...
    This study provides a functional link between proliferation and invasion of tumor cells based on diverse activities of p27(kip1) in different subcellular ...Missing: core CDKs Skp2 KPC AKT GSK3β JAB1 review
  80. [80]
    Cytoplasmic relocalization and inhibition of the cyclin-dependent ...
    Threonine 157 (T157), which maps within the nuclear localization signal of p27(kip1), is a predicted Akt-phosphorylation site. Akt-induced T157 phosphorylation ...Missing: sequestration | Show results with:sequestration<|separator|>
  81. [81]
    Complex Regulation of the Cyclin-Dependent Kinase Inhibitor ... - NIH
    Here we demonstrate that dysregulation of the PI3K/AKT pathway is important in thyroid carcinogenesis and that p27kip1 is a key target of the growth-regulatory ...Missing: CDKN1B | Show results with:CDKN1B
  82. [82]
    TGFbeta Prevents Proteasomal Degradation of the Cyclin ... - PubMed
    These data suggest that TGFbeta inhibits cellular proliferation by increasing p27 levels through Smad2/3 signaling in HEC-1A cells. We further show that TGFbeta ...
  83. [83]
    p27kip1 stabilization is essential for the maintenance of cell cycle ...
    Our results reveal that p27 plays an important role in the response to DNA damage, subsequent to that of the DDR-p53-p21 axis, and which is only activated after ...
  84. [84]
    p27 Kip1 Is Required to Mediate a G1 Cell Cycle Arrest Downstream ...
    The tumor suppressor p27Kip1 is a cyclin-CDK inhibitor that plays an important role in regulating quiescence in a variety of tissues. Several studies have ...
  85. [85]
    CDKN1B (p27/kip1) enhances drug-tolerant persister CTCs ... - PMC
    In these CTCs, efficient cell cycle exit generates a ≤4 N drug-tolerant state dependent on CDKN1B (p27/kip1). Exposure to DTX triggers stabilization of CDKN1B ...
  86. [86]
    MEN4 and CDKN1B mutations: The latest of the MEN syndromes - NIH
    MEN4 is caused by germline mutations in Cdkn1b in rats and CDKN1B in humans, coding for p27Kip1 (commonly referred to as p27 or KIP1, hereafter p27), a putative ...