The recombinant protein is synthesized in E. coli and purified via nickel-affinity chromatography leveraging the His-tag. Proprietary techniques ensure high yield and purity ( ). Key steps include:
Expression: Optimized in E. coli for non-glycosylated, monomeric production.
Purification: His-tag enables selective binding to Ni²⁺ columns, followed by buffer exchange to remove denaturing agents like urea.
Formulation: Lyophilized or in solution with stabilizing agents (e.g., glycerol, Tris-HCl) to prevent aggregation ( ).
Head and Neck Squamous Cell Carcinoma (HNSCC): High KRT20 expression correlates with lymphatic metastasis and poor prognosis. Overexpression in HNSCC cell lines (Tu686, FD-LSC-1) enhances migration and invasion ( ).
Colorectal Cancer: Elevated KRT20 levels in tumors versus normal mucosa predict recurrence and reduced survival ( ).
CDX1 Dependence: The KRT20 promoter contains CDX1/2 binding sites. CDX1 overexpression upregulates KRT20 by 9.3-fold in colorectal cells, linking it to intestinal differentiation ( ).
Targeting Metabolic Pathways: KRT20-associated glycerophospholipid metabolism and glutathione synthesis pathways are potential targets for inhibiting cancer progression ( ).
KRT20 (Keratin 20) is a protein encoded by the KRT20 gene in humans. The canonical form consists of 424 amino acid residues with a molecular mass of approximately 48.5 kDa. As a member of the Intermediate filament protein family, KRT20 is primarily localized in the cytoplasm and plays a crucial role in maintaining keratin filament organization in intestinal epithelia. It undergoes several post-translational modifications, including protein cleavage and phosphorylation, which may affect its function and localization .
The protein is known by several alternative names in the scientific literature, including CK-20, CK20, K20, KRT21, keratin type I cytoskeletal 20, cytokeratin 20, and keratin 20 type I. Gene orthologs have been identified across multiple species including mouse, rat, bovine, and chimpanzee, indicating evolutionary conservation of this protein .
KRT20 demonstrates a highly specific expression pattern, predominantly in intestinal epithelium. It serves as a valuable marker for characterizing several specialized cell types, including:
Large Intestine Epithelial Fibroblasts
Large Intestine Crypt Goblet Cells
Large Intestine Transit Amplifying Cells
Intestinal Tuft Cells
This specific expression pattern makes KRT20 a useful marker in both normal tissue identification and pathological diagnosis, particularly in determining the origin of metastatic carcinomas from gastrointestinal sources.
KRT20 can be detected through various immunological techniques, with Western Blot and Immunohistochemistry being the most widely employed methods. Over 140 citations in scientific literature describe the use of Keratin 20 antibodies in research applications . Typical detection approaches include:
Western Blot: Provides quantitative analysis of KRT20 protein expression levels
Immunohistochemistry (IHC): Offers spatial localization information in tissue sections
Immunofluorescence: Allows for co-localization studies with other proteins
Flow Cytometry: Enables analysis in cell suspensions
ELISA: Provides quantitative measurement in solution
The choice of detection method should align with specific research questions, with IHC being particularly valuable for examining tissue distribution patterns and Western Blot for quantifying expression levels.
Stable overexpression of KRT20 in cell lines can be achieved through lentiviral transfection systems. A detailed protocol based on published methodology includes:
Prepare KRT20 overexpression lentivirus (such as PGMLV-CMV-KRT20-eGFP-3Flag-PGK, Puromycin Lentivirus)
Mix the lentivirus with serum-free medium containing 1 μg/mL polybrene
Incubate cells with the mixture for 48 hours
Add puromycin (starting at 5 μg/mL and gradually increasing to 10 μg/mL) for selection
Maintain selection pressure for approximately one week to obtain stably transfected cells
This approach has been successfully implemented in head and neck squamous cell carcinoma (HNSCC) cell lines (Tu686 and FD-LSC-1) to study the effects of KRT20 overexpression on cellular migration and invasion capabilities .
Transwell migration and invasion assays have been effectively employed to evaluate the impact of KRT20 expression on cellular behavior. The protocols can be implemented as follows:
Transwell Migration Assay:
Seed 2 × 10⁴ cells in the upper chamber of 8 μm transwell inserts with 100 μL serum-free medium
Fill the lower chamber with medium containing 10% bovine serum albumin
Incubate for 36 hours
Remove non-migrated cells from the upper chamber
Fix migrated cells with paraformaldehyde (15 minutes)
Stain with 1% crystal violet (20 minutes)
Invasion Assay:
Coat the upper chamber with 50 μL Matrigel (diluted 1:8 with serum-free medium)
Seed 2 × 10⁵ cells in 100 μL serum-free medium
Research utilizing these methods has demonstrated that KRT20 overexpression significantly enhances both migration and invasion capabilities in HNSCC cell lines, suggesting a potential role in tumor progression and metastasis .
Several bioinformatic methods have proven valuable for investigating KRT20's involvement in cancer:
Differential Expression Analysis: Using RNA-seq data from repositories such as The Cancer Genome Atlas (TCGA) to identify differences in KRT20 expression between tumor and normal tissues or between cases with and without lymphatic metastasis
Random Forest Modeling: This machine learning approach can identify hub genes from differentially expressed genes (DEGs), helping to establish KRT20 as a key factor in processes like lymphatic metastasis
Protein-Protein Interaction Networks: Tools such as the Search Tool for the Retrieval of Interacting Genes, Cytoscape, and cytoHubba can help identify functional protein networks involving KRT20
Gene Set Enrichment Analysis (GSEA): This technique reveals biological pathways associated with KRT20 expression levels. GSEA analysis has shown that high KRT20 expression correlates with pathways including drug metabolism cytochrome P450, glycerophospholipid metabolism, glutathione metabolism, retinol metabolism, lysine degradation, valine leucine and isoleucine degradation, and steroid hormone biosynthesis
Survival Analysis: Correlating KRT20 expression with patient outcomes to determine prognostic significance
These approaches collectively provide comprehensive insights into KRT20's functional roles and clinical significance in cancer development.
Evidence suggests that KRT20 expression patterns have significant prognostic implications across multiple cancer types:
Interestingly, contradictory findings exist regarding KRT20's role in tumor progression. Some research indicates that tumors with complete absence of KRT20 expression are poorly differentiated with high Ki67+ cell percentages, suggesting a complex relationship between KRT20 expression and tumor biology that may vary by cancer type .
When employing KRT20 as a diagnostic marker, researchers should consider several methodological factors:
Antibody Selection: Choose validated antibodies with proven specificity for KRT20. Both monoclonal and polyclonal antibodies have been used successfully, but monoclonal antibodies typically offer higher specificity .
Tissue Processing: Standard formalin-fixed paraffin-embedded (FFPE) tissues work well for KRT20 detection, but optimization of antigen retrieval methods is crucial for maximizing signal.
Expression Pattern Analysis: KRT20 localization varies by tissue type. In intestinal cells, it's primarily cytoplasmic, while in some cancer cells, its distribution may extend to the intercellular matrix . Proper interpretation requires understanding of these pattern variations.
Quantification Methods:
For IHC: Consider both staining intensity and percentage of positive cells
For Western Blot: Normalize to appropriate housekeeping proteins
For qPCR: Select stable reference genes for accurate relative quantification
Context-Specific Interpretation: KRT20 expression patterns should be interpreted in the context of other markers and clinical information, as its significance can vary by cancer type and stage.
Researchers face contradictory findings regarding KRT20's role in cancer progression and prognosis. To address these contradictions methodologically:
Meta-analysis Approaches: Systematically review existing literature to identify patterns in contradictory findings, potentially revealing context-dependent effects.
Integrated Multi-omics Analysis: Combine transcriptomic, proteomic, and clinical data to develop a more comprehensive understanding of KRT20's role in specific contexts.
Explainable AI Methods: Utilize advanced machine learning approaches with explanatory components to detect and analyze contradictions in biomedical literature . These systems can retrieve relevant research articles, extract key research statements, and predict whether pairs of statements on the same topic contradict each other.
Context-Specific Studies: Design experiments that specifically test KRT20's function in different cancer types, stages, and cellular contexts to determine whether contradictions reflect genuine biological differences rather than methodological variations.
Standardized Reporting: Implement detailed reporting of experimental conditions, antibody specifications, and quantification methods to facilitate comparison across studies.
By applying these approaches, researchers can develop a more nuanced understanding of KRT20's context-dependent roles in cancer biology and resolve apparent contradictions in the literature.
Optimizing KRT20 detection requires consideration of several technical factors depending on the experimental approach:
For Western Blot:
Protein Extraction: Use RIPA buffer with protease inhibitors to prevent degradation
Sample Preparation: Denature samples at 95°C for 5 minutes in reducing conditions
Gel Selection: 10-12% polyacrylamide gels typically provide optimal separation
Transfer Conditions: Semi-dry transfer at 15V for 60 minutes or wet transfer at 100V for 60 minutes
Blocking: 5% non-fat milk in TBST for 1 hour at room temperature
Primary Antibody: Dilutions typically range from 1:500 to 1:2000 depending on antibody source
Detection: Both chemiluminescence and fluorescence-based systems can be effective
For Immunohistochemistry:
Fixation: 10% neutral buffered formalin is standard
Antigen Retrieval: Heat-induced epitope retrieval in citrate buffer (pH 6.0) or EDTA buffer (pH 9.0)
Blocking: 3-5% normal serum or BSA for 30-60 minutes
Primary Antibody Incubation: 1-2 hours at room temperature or overnight at 4°C
Detection System: Polymer-based detection systems often provide optimal signal-to-noise ratio
For Cell Culture Studies:
When overexpressing KRT20 in cell lines, maintaining consistent expression levels is critical. Regularly validate expression using Western blot or immunofluorescence to ensure stable transfection throughout experiments .
Purification of His-tagged KRT20 requires specialized protocols due to its intermediate filament properties:
Expression System Selection:
Bacterial systems (E. coli): Simpler but may require refolding steps
Eukaryotic systems (insect cells, mammalian cells): More likely to produce properly folded protein with authentic post-translational modifications
Lysis Conditions:
Buffer: 50 mM Tris-HCl pH 8.0, 300 mM NaCl, 10 mM imidazole, 0.1% Triton X-100
Include protease inhibitors to prevent degradation
Consider inclusion of reducing agents like 1-5 mM DTT to maintain protein stability
Purification Strategy:
Immobilized Metal Affinity Chromatography (IMAC) using Ni-NTA resin
Initial binding in presence of 10-20 mM imidazole to reduce non-specific binding
Step gradient or linear gradient of imidazole (50-300 mM) for elution
Follow with size exclusion chromatography to achieve higher purity
Quality Control:
SDS-PAGE with Coomassie staining to assess purity
Western blot with anti-His and anti-KRT20 antibodies to confirm identity
Mass spectrometry to verify protein integrity and identify any post-translational modifications
Storage Conditions:
Store in buffer containing 20 mM Tris-HCl pH 7.5, 150 mM NaCl, 10% glycerol
Aliquot and flash-freeze in liquid nitrogen
Store at -80°C for long-term stability
KRT20 undergoes several post-translational modifications (PTMs) that may influence its function. Effective experimental design for studying these modifications should include:
PTM Identification Strategies:
Mass spectrometry approaches (LC-MS/MS) with enrichment for specific modifications
Phosphorylation-specific protocols such as titanium dioxide enrichment
Targeted analysis using modification-specific antibodies
Site-Directed Mutagenesis:
Generate mutants where potential modification sites are altered
For phosphorylation: Ser/Thr/Tyr → Ala (phospho-null) or Asp/Glu (phospho-mimetic)
Express in appropriate cell systems and assess functional consequences
PTM Dynamics Analysis:
Pulse-chase experiments to track modification kinetics
Treatment with pathway inhibitors to identify regulatory mechanisms
Stress conditions to determine context-dependent modification patterns
Functional Impact Assessment:
Compare wild-type and mutant proteins in cellular assays
Assess changes in protein-protein interactions
Evaluate effects on subcellular localization using fluorescence microscopy
Determine impact on filament formation and stability
PTM Crosstalk Evaluation:
Investigate potential interactions between different types of modifications
Determine whether one modification influences the occurrence of others
Map modification patterns across different cellular contexts
Recent research has begun exploring KRT20's potential as a therapeutic target, particularly in cancers where it shows aberrant expression:
Targeted Antibody Approaches:
Development of KRT20-specific antibodies conjugated to cytotoxic agents
Investigation of KRT20 as a tumor-associated antigen for immunotherapy strategies
Gene Expression Modulation:
siRNA or shRNA strategies to downregulate KRT20 in cancers where high expression correlates with poor outcomes
CRISPR/Cas9 approaches for more permanent genetic modification
Signaling Pathway Intervention:
Biomarker Development:
Utilization of KRT20 expression patterns for patient stratification in clinical trials
Development of companion diagnostics for therapies targeting KRT20-associated pathways
While still in early stages, these approaches represent promising directions for translating basic KRT20 research into clinical applications.
Despite extensive study, several significant knowledge gaps remain in our understanding of KRT20:
Regulatory Mechanisms: The precise transcriptional and post-transcriptional mechanisms controlling KRT20 expression remain incompletely understood, particularly in pathological contexts.
Protein Interaction Network: While KRT20 is known to participate in intermediate filament formation, its complete interactome and how these interactions change in disease states requires further elucidation.
Context-Dependent Functions: The apparently contradictory roles of KRT20 in different cancer types suggest context-dependent functions that are not fully characterized .
Mechanistic Understanding: Despite correlations between KRT20 expression and cancer progression, the molecular mechanisms by which KRT20 influences cellular processes like migration and invasion remain to be fully determined.
Post-Translational Regulation: While post-translational modifications of KRT20 have been documented , their functional significance and regulatory mechanisms require further investigation.
Therapeutic Potential: The feasibility and efficacy of targeting KRT20 or its associated pathways for therapeutic purposes remains largely unexplored.
Addressing these knowledge gaps represents priority areas for future KRT20 research.
Cytokeratin 20 (CK20) is a type I cytokeratin, which is a member of the keratin family of proteins. These proteins are essential for the structural integrity of epithelial cells. Cytokeratins are subdivided into two types: type I (acidic) and type II (basic to neutral). CK20 is specifically expressed in the gastric and intestinal mucosa, making it a significant marker in the study of epithelial cell biology.
CK20 is a major cellular protein of mature enterocytes and goblet cells. The protein encoded by the CK20 gene consists of 424 amino acids and has a molecular weight of approximately 50.9 kDa . The recombinant form of CK20, fused with a His-tag at the N-terminus, is expressed in Escherichia coli (E. coli) and purified using chromatographic techniques .
CK20 plays a crucial role in maintaining the structural integrity of epithelial cells. It forms intermediate filaments that provide mechanical support and stability to the cells. The expression of CK20 is highly specific to certain types of epithelial cells, making it a valuable marker in diagnostic pathology. It is often used to identify and differentiate between various types of carcinomas, particularly those originating in the gastrointestinal tract.
The recombinant form of CK20, tagged with a His-tag, is widely used in research. The His-tag facilitates the purification and detection of the protein. The recombinant CK20 is produced in E. coli and is available in a purified form. It is typically stored in a buffer containing Tris-HCl, urea, and glycerol to maintain its stability .
Recombinant CK20 is used in various research applications, including: