CDKN1A Human

Cyclin-Dependent Kinase Inhibitor 1A Human Recombinant
Shipped with Ice Packs
In Stock

Description

Molecular Structure and Function

CDKN1A/p21 is a 164-amino acid protein that operates through three primary mechanisms:

Cyclin-Dependent Kinase (CDK) Inhibition

  • Binds to CDK complexes (CDK2, CDK1, CDK4/6) via N-terminal domains, blocking cell cycle progression at G₁ and S phases .

  • Exhibits bistable behavior in single-cell studies: high p21 levels drive quiescence, while low levels permit proliferation .

PCNA Interaction

  • Competes with DNA polymerase δ for PCNA binding, inhibiting DNA synthesis while allowing nucleotide excision repair .

  • Structural analysis reveals preferential binding to PCNA's PIP-box region under DNA damage conditions .

Apoptosis Modulation

  • Caspase-3 cleavage activates CDK2, promoting apoptosis execution .

  • Paradoxically, p21 overexpression can suppress apoptosis in replication stress .

p53-Dependent Pathway

CDKN1A is a major transcriptional target of TP53 (p53), linking DNA damage to G₁ arrest. Key findings include:

MechanismOutcomeSource
p53 activation by γ-irradiationInduces p21-mediated CDK2 inhibition
Single-cell p53 pulsesGenerate oscillatory p21 expression, phase-dependent arrest
p21 knockout miceDevelop spontaneous tumors and defective G₁ checkpoints

Post-Transcriptional Control

  • UVB exposure triggers eIF2α phosphorylation, enhancing CDKN1A mRNA translation .

  • Alternative splicing produces isoforms with distinct 5′-UTRs, enabling stress-specific regulation .

Cancer

A 2024 pan-cancer analysis of 33 cancer types revealed:

Cancer TypeCDKN1A ExpressionPrognostic AssociationImmune Correlation
GBMUpregulatedPoor OS (HR=1.2)Macrophage infiltration↑
BLCADownregulatedReduced DFS (p<0.01)CD8+ T cells↓
KIRCUpregulatedImproved DSS (p=0.03)Neutrophils↑

Data derived from TCGA and GEO datasets .

  • Glioblastoma (GBM): High CDKN1A correlates with temozolomide resistance via AKT pathway activation .

  • Gastric Cancer: Low p21 expression predicts recurrence (HR=2.1, p<0.001) and shorter OS (25 vs. 68 months) .

Aging and Senescence

  • Induces stress-induced premature senescence in human fibroblasts .

  • Rare CDKN1A alleles associate with reduced longevity in Italian centenarians (OR=0.32, p=0.008) .

Cardiovascular Disease

  • Mendelian randomization implicates CDKN1A in heart failure (OR=1.2, FDR<0.0001) through cardiomyocyte senescence .

  • Methylation at cg03714916 and cg08179530 loci modifies HF risk .

Key Studies (2015–2024)

Study FocusMajor FindingSource
Chemoresistancep21↑ in TMZ-resistant glioma cells promotes AKT activation
Pan-cancer analysisCDKN1A inversely correlates with TMB/MSI in 12 cancers
Cardiac agingCDKN1A methylation initiates senescence in HF cardiomyocytes

Therapeutic Targets

  • Oncology: Dual CDK2/PCNA inhibitors under development exploit p21’s bistable dynamics .

  • Cardiology: Repurposing CDKN1A-targeted senolytics (e.g., navitoclax) shows preclinical promise for HF .

Product Specs

Introduction
As a crucial regulator of the cell cycle, CDKN1A plays a significant role in the cellular response to DNA damage. Its overexpression leads to cell cycle arrest. CDKN1A interacts with CDK2 complexes to inhibit kinase activity, thus preventing progression through the G1/S phase. However, CDKN1A can also enhance the assembly and activity of complexes involving CDK4 or CDK6 and cyclin D. It acts as a key mediator for p53/TP53 to exert its function as a suppressor of cell proliferation in response to DNA damage.
Description
Recombinant human CDKN1A, produced in E. coli, is a single polypeptide chain comprising 184 amino acids (residues 1-164) with a molecular weight of 20.2 kDa. A 20 amino acid His-tag is fused to the N-terminus of CDKN1A, and the protein is purified using proprietary chromatographic methods.
Physical Appearance
Clear, colorless solution, sterile-filtered.
Formulation
The CDKN1A solution is formulated in 20mM Tris-HCl buffer (pH 8.0), 2M Urea, 0.1M NaCl, and 10% glycerol.
Stability
For short-term storage (2-4 weeks), keep at 4°C. For extended storage, freeze at -20°C. Adding a carrier protein (0.1% HSA or BSA) is recommended for long-term storage. Avoid repeated freeze-thaw cycles.
Purity
Purity is determined to be greater than 85.0% by SDS-PAGE analysis.
Synonyms
Cyclin-Dependent Kinase Inhibitor 1A (P21, Cip1), Melanoma Differentiation Associated Protein 6, Wild-Type P53-Activated Fragment 1, CDK-Interaction Protein 1, DNA Synthesis Inhibitor, CDK-Interacting Protein 1, CDKN1, P21Cip1/Waf1, MDA6, P21CIP1, CAP20, WAF1, CIP1, PIC1, P21, SDI1.
Source
Escherichia Coli.
Amino Acid Sequence
MGSSHHHHHH SSGLVPRGSH MSEPAGDVRQ NPCGSKACRR LFGPVDSEQL SRDCDALMAG CIQEARERWN FDFVTETPLE GDFAWERVRG LGLPKLYLPT GPRRGRDELG GGRRPGTSPA LLQGTAEEDH VDLSLSCTLV PRSGEQAEGS PGGPGDSQGR KRRQTSMTDF YHSKRRLIFS KRKP.

Q&A

What is the molecular function of CDKN1A and how does it regulate the cell cycle?

CDKN1A encodes the p21 protein, which functions as a cyclin-dependent kinase inhibitor primarily targeting CDK2 and CDK4 complexes. The primary mechanism of action involves regulating cell cycle progression at the G1 phase via the retinoblastoma protein (RB1) pathway . When activated, p21 binds to and inhibits the activity of cyclin/CDK complexes, which ultimately blocks cell progression from G1 to S phase . This cell cycle arrest mechanism is critical for allowing DNA repair processes to occur before DNA replication.

The molecular function of p21 extends beyond simple CDK inhibition. Research has demonstrated that p21 participates in:

  • Direct inhibition of PCNA (proliferating cell nuclear antigen), preventing DNA replication

  • Regulation of transcription factor activity

  • Modulation of apoptotic responses

  • Facilitation of DNA repair mechanisms

Experimental approaches to study CDKN1A's cell cycle regulation typically include flow cytometry for cell cycle distribution analysis, co-immunoprecipitation for protein-protein interaction studies, and genetic manipulation models with p21 overexpression or knockdown.

What are the known CDKN1A transcript variants and their functional significance?

Human CDKN1A produces multiple transcript variants through alternative splicing, with distinct functional characteristics:

Known CDKN1A Transcript Variants:

VariantDistinguishing FeaturesFunctional SignificanceResponse to Stress
Variant 1Well-studied canonical formImmediate response to stressRapidly upregulated by genotoxic stress
Variant 2Distinct 5'-leader sequenceAssociated with aging and senescenceGradual increase during senescence
Variant 4Alternative 5'-leaderPreferentially translated during stressEnhanced by eIF2α phosphorylation

Research has demonstrated that variant 4 is preferentially translated following stress-induced eIF2α phosphorylation through a mechanism mediated by upstream open reading frames (uORFs) in its 5'-leader sequence . This selective translation represents an important regulatory mechanism during cellular stress responses.

In mouse models, Cdkn1a variant 2 (not the better-studied variant 1) is selectively elevated during natural aging across multiple tissues. Moreover, variant 2 exhibits different temporal dynamics in response to genotoxic stress compared to variant 1: variant 1 responds almost immediately, while variant 2 increases much more slowly as cells acquire senescent characteristics . These findings suggest specialized roles for different transcript variants in aging and stress responses.

How does CDKN1A contribute to cellular senescence and aging?

CDKN1A plays a central role in cellular senescence, which has significant implications for the aging process:

The induction of p21 is a critical mediator of cell cycle arrest during senescence establishment. In human cells, increased CDKN1A mRNA levels are observed during stress-induced premature senescence . The stringent cell growth arrest associated with cellular senescence is determined in part by p21 activity, alongside other mechanisms like p16INK4a expression .

Specifically regarding aging:

  • Transcript variant 2 of CDKN1A appears to be a more sensitive biomarker of aging than variant 1 or total p21 protein for assessing senescent cell burden in mice

  • When treating aged mice with the senolytic drug ABT-263, variant 2 levels were more sensitive to treatment than variant 1

  • In studies of human longevity, certain CDKN1A alleles were found to be very rare in Italian centenarians, suggesting potential detrimental effects on longevity

Interestingly, deletion of p21 in mice with dysfunctional telomeres actually prolonged lifespan. These mice exhibited improved hematopoiesis and intestinal epithelial maintenance, along with enhanced proliferation of intestinal progenitor cells and improved self-renewal of hematopoietic stem cells . This suggests that p21 may play different roles depending on the genetic and physiological context.

What methodologies are most effective for studying CDKN1A expression patterns?

Several complementary methodologies are employed to comprehensively analyze CDKN1A expression:

Transcript-level Analysis:

  • RT-qPCR with variant-specific primers to distinguish between splice variants

  • Northern blotting for total mRNA quantification

  • RNA-seq for genome-wide expression profiling and splice variant detection

  • Single-cell RNA sequencing (scRNA-seq) to capture expression heterogeneity at the cellular level

Protein-level Analysis:

  • Western blotting for total p21 protein quantification

  • Immunohistochemistry (IHC) to analyze spatial distribution in tissues

  • Immunofluorescence microscopy for subcellular localization

  • Flow cytometry for quantitative analysis in cell populations

A comprehensive study of CDKN1A should incorporate multiple techniques. For example, immunohistochemistry can reveal important subcellular localization patterns - research has shown that in normal gastric tissues, CDKN1A protein is primarily nuclear, whereas in gastric adenocarcinoma tissues, it shows both nuclear and cytoplasmic expression .

How does CDKN1A expression correlate with prognosis across different cancer types?

CDKN1A expression exhibits variable patterns across cancer types with significant prognostic implications:

Cancer Types with Low CDKN1A Expression Compared to Normal Tissue:

  • Bladder carcinoma (BLCA)

  • Breast cancer (BRCA)

  • Colon adenocarcinoma (COAD)

  • Kidney chromophobe (KICH)

  • Lung adenocarcinoma (LUAD)

  • Lung squamous cell carcinoma (LUSC)

  • Prostate adenocarcinoma (PRAD)

  • Rectum adenocarcinoma (READ)

  • Stomach adenocarcinoma (STAD)

Cancer Types with High CDKN1A Expression Compared to Normal Tissue:

  • Cholangiocarcinoma (CHOL)

  • Head and neck squamous cell carcinoma (HNSC)

  • Kidney renal clear cell carcinoma (KIRC)

  • Kidney renal papillary cell carcinoma (KIRP)

  • Thyroid carcinoma (THCA)

The prognostic significance of CDKN1A varies by cancer type. In resected gastric adenocarcinoma (RGA), low CDKN1A expression is significantly associated with lymph node metastasis, increased recurrence risk, and shorter survival time . Multiple statistical analyses have confirmed that low CDKN1A expression in RGA tissues represents an independent prognostic factor for poor outcomes.

Methodologically, researchers investigating CDKN1A as a prognostic marker should employ multivariate survival analyses, controlling for established clinicopathological factors to demonstrate independent prognostic value.

What are the mechanisms underlying differential regulation of CDKN1A in response to cellular stress?

The differential regulation of CDKN1A under various stress conditions involves multiple layers of control:

Transcriptional Regulation:

  • p53-dependent activation following DNA damage

  • p53-independent pathways through other transcription factors (Sp1, Sp3, AP2)

  • Epigenetic mechanisms including promoter methylation

Post-transcriptional Regulation:

  • Modulation of mRNA stability

  • Alternative splicing generating distinct variants

  • Long non-coding RNA (lncRNA) involvement in regulation

Translational Control:

  • Selective translation of specific splice variants during stress

  • Regulation via upstream open reading frames (uORFs)

  • eIF2α phosphorylation mediating preferential translation

Research has demonstrated that UVB irradiation triggers the integrated stress response, leading to eIF2α phosphorylation and subsequent preferential translation of CDKN1A splice variant 4 . This mechanism is cytoprotective, facilitating G1 arrest and subsequent DNA repair. Notably, loss of eIF2α phosphorylation diminishes UVB-induced G1 arrest, reduces DNA repair rates, weakens cellular senescence induction, and increases apoptosis .

When investigating these mechanisms, researchers should consider employing polysome profiling to study translational regulation, chromatin immunoprecipitation (ChIP) to analyze transcription factor binding, and reporter gene assays to study promoter activity under various conditions.

How does CDKN1A influence the tumor microenvironment and immune response?

Recent research has revealed that CDKN1A plays significant roles in shaping the tumor microenvironment (TME) and modulating anti-tumor immunity:

CDKN1A expression is significantly associated with immune cell infiltration in various cancer types. Analysis using TIMER and other computational tools has demonstrated correlations between CDKN1A expression and infiltration of:

  • CD4+ T cells

  • CD8+ T cells

  • Neutrophils

  • Macrophages

  • Myeloid dendritic cells

These associations vary across cancer types, suggesting context-dependent roles of CDKN1A in immune regulation. The mechanisms by which CDKN1A influences the TME may involve:

  • Modulation of cytokine/chemokine production

  • Regulation of cancer cell immunogenicity

  • Effects on immune checkpoint expression

  • Influence on immunogenic cell death

Methodological approaches to investigate these relationships include:

  • Multiplex immunohistochemistry to assess spatial relationships between p21-expressing cells and immune infiltrates

  • Single-cell RNA sequencing to characterize cell-type-specific expression patterns

  • Co-culture experiments to study direct interactions between p21-manipulated cancer cells and immune cell populations

  • Mouse models with cell-type-specific p21 knockouts to evaluate in vivo immune responses

Researchers should consider these immune-related functions when designing experiments to study CDKN1A in cancer contexts, as they may represent important mechanisms underlying its tumor-suppressive effects beyond cell cycle regulation.

What therapeutic strategies target CDKN1A in cancer treatment?

Several therapeutic approaches leverage CDKN1A's functions for cancer treatment:

Strategies to Restore CDKN1A Expression:

  • Demethylating agents to reverse epigenetic silencing of CDKN1A

  • HDAC inhibitors that can enhance p21 expression

  • miRNA inhibitors targeting p21-suppressing microRNAs

  • Small molecules that stabilize p21 protein

Strategies Based on CDKN1A Status:

  • Synthetic lethal approaches exploiting CDKN1A deficiency

  • Cell cycle checkpoint inhibitors in combination with p21-inducing agents

  • Senolytic drugs targeting senescent cells (p21-positive) in the tumor microenvironment

Experimental evidence has shown that p21 overexpression leads to significant reduction in proliferative capacity, facilitates cell apoptosis, and promotes senescence in multiple cancer cell lines . Conversely, p21 silencing facilitates cell growth and wound closure while preventing cell senescence . These findings suggest that modulating p21 levels could be therapeutically valuable depending on cancer context.

The senolytic drug ABT-263 has shown effectiveness in reducing levels of CDKN1A variant 2 in aged mice , suggesting potential applications in targeting senescent cells in aging-related pathologies and possibly in cancer contexts where senescent cells contribute to disease progression.

Researchers developing CDKN1A-targeted therapies should consider:

  • Cancer-specific expression patterns and prognostic implications

  • Differential roles of splice variants

  • Combination approaches with standard therapies

  • Potential for synthetic lethality in specific genetic backgrounds

How do CDKN1A splice variants contribute to differential cellular responses?

The differential expression and regulation of CDKN1A splice variants contribute significantly to varied cellular responses:

Temporal Dynamics in Stress Response:
Different CDKN1A variants show distinct kinetics following stress exposure. While variant 1 responds almost immediately to genotoxic stress, variant 2 increases much more gradually as cells acquire senescent characteristics . This temporal separation may allow cells to orchestrate immediate versus long-term responses to damage.

Translation Regulation During Stress:
Human CDKN1A splice variant 4 is preferentially translated during the integrated stress response. This selective translation is mediated by upstream open reading frames (uORFs) in the 5'-leader sequence and is dependent on eIF2α phosphorylation . This mechanism ensures that specific p21 isoforms are produced under stress conditions.

Aging and Senescence Biomarkers:
Cdkn1a transcript variant 2 (not variant 1) is selectively elevated during natural aging across multiple mouse tissues, making it a more sensitive biomarker for aging and cellular senescence . When mice are treated systemically with doxorubicin to induce widespread cellular senescence, variant 2 increases to a greater extent than variant 1 .

For researchers studying these differential responses, recommended methodological approaches include:

  • Variant-specific RT-qPCR with carefully designed primers

  • Translational efficiency assays using luciferase reporters with variant-specific 5'-leaders

  • Polysome profiling combined with variant-specific RT-qPCR to assess translational status

  • CRISPR-based approaches to selectively manipulate individual splice variants

  • Time-course experiments to capture temporal dynamics of variant expression

Understanding these variant-specific roles has important implications for biomarker development and therapeutic targeting strategies in both cancer and aging-related conditions.

What are the best experimental models for studying CDKN1A function?

Selecting appropriate experimental models is crucial for investigating CDKN1A's diverse functions:

Cellular Models:

  • Primary human cells versus established cell lines (important considerations for senescence studies)

  • Cell type-specific differences in CDKN1A regulation and function

  • Isogenic cell lines with CDKN1A knockout/knockin modifications

  • 3D organoid cultures that better recapitulate tissue architecture

Animal Models:

  • CDKN1A knockout mice exhibit normal development but defective G1 checkpoint control

  • Tissue-specific or inducible CDKN1A knockout models

  • Models with dysfunctional telomeres combined with p21 status manipulation

  • Humanized mouse models for studying human-specific splice variants

Experimental Approaches:

  • CRISPR/Cas9 genome editing for precise manipulation of CDKN1A locus

  • Inducible expression systems to study acute versus chronic effects

  • Single-cell approaches to capture heterogeneity in CDKN1A expression and function

  • Patient-derived xenografts to study CDKN1A in human tumor contexts

When designing experiments, researchers should consider that CDKN1A functions may be highly context-dependent. For example, p21 deletion in mice with dysfunctional telomeres actually prolonged lifespan despite its canonical role as a tumor suppressor . This underscores the importance of choosing models that appropriate reflect the biological context of interest.

How can contradictory findings on CDKN1A functions be reconciled in research?

The scientific literature contains seemingly contradictory findings regarding CDKN1A functions. These can be addressed through several methodological considerations:

Context-Dependent Functions:
CDKN1A exhibits different, sometimes opposing roles depending on cellular context. In gastric cancer, low CDKN1A expression is associated with poor prognosis , while in other cancers, high expression correlates with aggressive features . These conflicting observations may reflect genuine biological differences rather than experimental artifacts.

Subcellular Localization Differences:
CDKN1A protein can localize to different cellular compartments, with distinct functions. In normal gastric tissues, it primarily shows nuclear localization, whereas in gastric adenocarcinoma tissues, it exhibits both nuclear and cytoplasmic expression . Researchers should employ methods that distinguish between nuclear and cytoplasmic p21 pools.

Temporal Considerations:
The timing of CDKN1A induction can determine functional outcomes. Early versus late induction may have different consequences for cell fate decisions . Time-course experiments are essential to capture these dynamic effects.

To reconcile conflicting findings, researchers should:

  • Clearly specify the cellular context and experimental conditions

  • Distinguish between different splice variants

  • Analyze subcellular localization

  • Consider temporal dynamics

  • Employ multiple complementary techniques

  • Validate findings across different model systems

This comprehensive approach will help clarify the nuanced and context-dependent functions of CDKN1A.

What bioinformatic approaches are useful for analyzing CDKN1A in multi-omics datasets?

Multi-omics analysis of CDKN1A requires sophisticated bioinformatic approaches:

Transcriptomic Analysis:

  • RNA-seq analysis with splice-aware alignment to distinguish variants

  • Differential expression analysis across conditions and tissues

  • Co-expression network analysis to identify functional modules

  • Alternative splicing analysis to characterize splice variant usage

Genomic Analysis:

  • Analysis of CDKN1A promoter variants and genetic polymorphisms

  • Identification of cis-regulatory elements affecting expression

  • Assessment of copy number variations in cancer contexts

  • Integration with genotype-phenotype correlation studies

Epigenomic Analysis:

  • DNA methylation profiling of CDKN1A promoter regions

  • Chromatin accessibility (ATAC-seq) analysis

  • Histone modification ChIP-seq to assess chromatin state

  • Analysis of long-range chromatin interactions affecting regulation

Integrative Multi-omics:

  • Integration of transcriptomic, genomic, and epigenomic data

  • Correlation with proteomic data to assess translation efficiency

  • Pathway enrichment analysis to contextualize CDKN1A function

  • Machine learning approaches to identify predictive signatures

Useful bioinformatic resources and tools include:

  • cBioPortal for accessing and analyzing cancer genomics datasets

  • TIMER for analyzing immune infiltration correlations

  • UCSCXenaShiny for interactive multi-omics data visualization

  • Kaplan-Meier analysis tools for survival correlations

Single-cell RNA sequencing analysis is particularly valuable for understanding CDKN1A expression heterogeneity within complex tissues and for characterizing cell type-specific regulatory patterns .

Product Science Overview

Introduction

Cyclin-Dependent Kinase Inhibitor 1A (CDKN1A), also known as p21, WAF1, or CIP1, is a crucial protein in the regulation of the cell cycle. It is encoded by the CDKN1A gene and plays a significant role in cell cycle control, DNA damage response, and apoptosis. The recombinant form of this protein is used in various research and therapeutic applications.

Gene and Protein Structure

The CDKN1A gene is located on chromosome 6p21.2 and encodes a protein of 164 amino acids. The protein structure includes several functional domains that allow it to interact with cyclins, cyclin-dependent kinases (CDKs), and proliferating cell nuclear antigen (PCNA). These interactions are essential for its role in cell cycle regulation.

Function and Mechanism

CDKN1A functions primarily as a cyclin-dependent kinase inhibitor. It binds to and inhibits the activity of cyclin-CDK2 or cyclin-CDK4 complexes, thereby regulating cell cycle progression at the G1 phase . This inhibition is crucial for the maintenance of the G1 checkpoint, preventing cells with damaged DNA from entering the S phase and undergoing replication.

The expression of CDKN1A is tightly controlled by the tumor suppressor protein p53. In response to DNA damage, p53 induces the transcription of CDKN1A, leading to cell cycle arrest at the G1 checkpoint . This mechanism allows the cell to repair DNA damage before proceeding with division, thereby preventing the propagation of mutations.

Role in DNA Damage Response

CDKN1A is a key player in the cellular response to DNA damage. Upon DNA damage, p53 is activated and subsequently induces the expression of CDKN1A. The increased levels of CDKN1A inhibit CDK activity, leading to cell cycle arrest. This process allows the cell to repair the DNA damage before resuming the cell cycle .

In addition to its role in cell cycle arrest, CDKN1A also interacts with PCNA, a protein involved in DNA replication and repair. By binding to PCNA, CDKN1A can inhibit DNA synthesis and regulate DNA repair processes .

Clinical Significance

The dysregulation of CDKN1A has been implicated in various cancers. Overexpression of CDKN1A can lead to cell cycle arrest and apoptosis, making it a potential target for cancer therapy. Conversely, the loss of CDKN1A function can result in uncontrolled cell proliferation and tumor development .

Recombinant CDKN1A is used in research to study its role in cell cycle regulation and its potential as a therapeutic target. It is also used in drug screening assays to identify compounds that can modulate its activity.

Quick Inquiry

Personal Email Detected
Please use an institutional or corporate email address for inquiries. Personal email accounts ( such as Gmail, Yahoo, and Outlook) are not accepted. *
© Copyright 2024 Thebiotek. All Rights Reserved.