KRT6A Monoclonal Antibody is a mouse-derived immunoglobulin targeting cytokeratin 6A (KRT6A), a type-II cytoskeletal protein critical for epithelial cell integrity and stress response. It recognizes a 56 kDa protein isoform encoded by the KRT6A gene, which is part of a family of keratins (6A-6F) with distinct tissue-specific expression patterns .
Target: Cytokeratin 6A (56 kDa)
Host: Mouse
Isotype: IgG2a (common clone KRT6A/2368) or IgG1 (other clones)
Reactivity: Human (cross-reactivity with mouse may vary)
Applications: Immunohistochemistry (IHC-P), Flow Cytometry, Immunofluorescence (IF), and Western Blotting
The antibody is widely used to study epithelial cell biology, skin disorders, and cancer progression. Below are its primary applications:
KRT6A is strongly expressed in hyper-proliferative epithelial cells, including hair follicles, suprabasal epidermis, and squamous cell carcinomas . In lung cancer, high KRT6A expression correlates with:
Radioresistance: Enhanced survival of cancer cells under radiation .
Invasion and Metastasis: Promotion of epithelial-mesenchymal transition (EMT) via downregulation of E-cadherin .
Proliferation: Increased cell growth rates in vitro (e.g., A549 and H1299 lung adenocarcinoma cells) .
KRT6A regulates cancer progression through:
c-MYC/MYCN Axis: Upregulation of oncogenes to enhance glucose metabolism (e.g., G6PD expression) .
LSD1 Inhibition: LSD1 histone demethylase inhibitors suppress KRT6A expression, reducing tumor growth .
KRT6A is emerging as a predictive biomarker for:
Lung Cancer: High expression predicts poor prognosis and radioresistance .
Squamous Cell Carcinomas: Detected in ~75% of head/neck SCCs .
Pachyonychia Congenita: Mutations in KRT6A cause skin/nail disorders .
Antigen Retrieval: Boil sections in 10 mM citrate buffer (pH 6.0) for 10–20 minutes .
Blocking: Use 1% BSA or commercial blocking buffers.
Primary Antibody: Incubate at 1–2 µg/ml for 30–60 minutes at RT .
This KRT6A monoclonal antibody is produced using hybridoma technology. Splenic B cells from a mouse immunized with a synthetic human KRT6A-derived peptide were fused with myeloma cells to generate hybridoma cell lines. Antibody-secreting hybridomas were selected and subsequently injected into the peritoneal cavity of mice for ascites production. The KRT6A monoclonal antibody was purified from the mouse ascites fluid via affinity chromatography using the immunizing peptide, ensuring high purity. This antibody is suitable for ELISA and immunohistochemistry (IHC) applications and has been validated for its specificity to human KRT14 protein.
KRT6A is specifically expressed in hair follicles, nails, and other keratinized epithelial tissues. It plays a crucial role in hair shaft formation and maintenance, contributing to hair growth and differentiation. Furthermore, KRT6A is implicated in wound healing and the cellular response to various stressors, including heat shock and oxidative stress. Mutations in the KRT6A gene have been linked to hair disorders such as pachyonychia congenita.
KRT6A is an epidermis-specific type I keratin involved in wound healing. It participates in the activation of follicular keratinocytes following wounding, though its role in keratinocyte proliferation and migration is less significant. KRT6A regulates epithelial migration by inhibiting SRC kinase activity during wound repair.
KRT6A Function and Associated Genetic Studies:
KRT6A (Keratin 6A) is a type II keratin protein of approximately 56kDa that plays important roles in epithelial tissue structure and function. In normal tissues, KRT6A is predominantly expressed in several locations:
Hair follicles
Suprabasal cells of internal stratified epithelia
Epidermal tissue undergoing rapid turnover
KRT6A works in conjunction with KRT16 and is classified as a hyperproliferation-related keratin. Its expression is significantly upregulated during wound healing processes, where epidermal injury activates keratinocytes to express both KRT6 and KRT16 . This expression pattern makes KRT6A an important marker for tissue regeneration and hyperproliferative states.
In humans, multiple isoforms of Cytokeratin 6 (6A-6F) exist, encoded by several highly homologous genes, with KRT6A being the dominant form in epithelial tissue . This specificity must be considered when designing research studies targeting specific keratin isoforms.
KRT6A monoclonal antibodies are versatile research tools with several validated applications:
Flow Cytometry: Recommended dilution of 1-2 μg per million cells. Particularly useful for quantifying KRT6A-expressing cell populations and sorting cells based on KRT6A expression levels .
Immunohistochemistry on Paraffin-embedded Sections (IHC-P): Optimal at 1-2 μg/ml dilution. This application is particularly valuable for examining KRT6A expression patterns in tissue architecture and for comparative studies between normal and pathological samples .
Lineage Tracing Studies: When combined with transgenic approaches, KRT6A antibodies can be used to identify and track bipotential progenitor cells, as demonstrated in mammary tissue research .
Cancer Biomarker Identification: Particularly useful in head and neck squamous cell carcinomas where KRT6A is strongly expressed in approximately 75% of cases .
For optimal results, validation using both positive and negative control tissues is essential, as expression patterns can vary significantly between tissue types and pathological states.
Validating KRT6A monoclonal antibody specificity requires a multi-faceted approach:
Western Blotting Confirmation: Verify single-band detection at 56kDa, which corresponds to the KRT6A protein. Multiple bands may indicate cross-reactivity with other keratin family members due to high sequence homology.
Positive Control Selection: Use tissues known to express high levels of KRT6A, such as hyperproliferative epidermis or certain squamous cell carcinomas, particularly those from head and neck regions where expression reaches approximately 75% .
Negative Controls: Include tissues known to have minimal KRT6A expression and perform antibody tests on KRT6A-knockdown cell lines, which can be generated using siRNA approaches as demonstrated in lung cancer studies .
Immunohistochemical Pattern Analysis: Confirm the expected subcellular localization and tissue distribution pattern. KRT6A should primarily show cytoplasmic staining in epithelial cells within expected tissue compartments.
Cross-Validation with Multiple Detection Methods: Compare results between IHC-P, flow cytometry, and molecular techniques like qPCR to ensure consistency in the expression patterns detected .
The Human Protein Atlas database provides additional reference immunohistochemical data that can be used to benchmark expected staining patterns in normal and cancerous tissues .
KRT6A has been identified as a significant factor in multiple cancer types with distinct contributions to cancer progression:
Methodological approaches for studying KRT6A in cancer include:
siRNA Knockdown Experiments: As demonstrated in lung cancer studies, where two independent siRNAs were used to downregulate KRT6A expression, resulting in significantly reduced proliferation rates (p<0.05) .
Colony Formation Assays: KRT6A-knockdown A549 cells showed reduced colony formation (55±9) compared to control cells (157±14), providing a quantitative measure of proliferative capacity .
Migration Assays: Transwell migration studies revealed KRT6A-knockdown cells exhibited reduced migration (521±8 cells) compared to control (533±17 cells) .
In Vivo Models: Transgenic mouse models expressing tva receptor under the K6a promoter can be used to study tumor development from K6a+ cells using RCAS viral vectors carrying oncogenes .
The integrated use of these methods provides a comprehensive picture of KRT6A's role in cancer progression.
KRT6A has been identified as a potential mediator of radioresistance in lung cancer, with several experimental approaches available to investigate this relationship:
Differential Expression Analysis: GEO datasets (GSE73095 and GSE197236) have been used to compare mRNA expression between radioresistant and radiosensitive lung cancer tissues, identifying KRT6A as differentially expressed .
Gene Knockdown Before Irradiation: siRNA-mediated knockdown of KRT6A followed by radiation treatment can help evaluate changes in cell survival, DNA damage repair efficiency, and apoptotic response.
Clonogenic Survival Assays: The gold standard for measuring radioresistance, where KRT6A-knockdown and control cells are irradiated at various doses and their colony-forming ability is quantified over 14 days. Studies have shown significant differences in colony numbers between KRT6A-expressing (157±14) and KRT6A-knockdown (55±9) cells .
Cell Viability Assessment: CCK-8 assays following irradiation demonstrate that KRT6A knockdown significantly reduces cell viability post-radiation, with measurable differences at 12, 24, 48, and 72 hours .
Mechanistic Pathway Analysis: GO and KEGG enrichment analyses can identify pathways through which KRT6A mediates radioresistance, potentially involving DNA repair mechanisms, cell cycle regulation, or anti-apoptotic pathways .
When designing radioresistance studies, researchers should consider both intrinsic radioresistance (baseline resistance) and acquired radioresistance (resistance developing after radiation exposure), as KRT6A may play different roles in each scenario.
KRT6A expression correlates with clinical outcomes across multiple cancer types, with important implications for patient stratification and prognosis:
For researchers studying clinical correlations, optimal approaches include:
Tissue microarray analysis with quantitative scoring of KRT6A immunohistochemistry
Correlation of KRT6A expression with patient survival using Kaplan-Meier methods
Multivariate Cox regression analysis to determine independent prognostic value
Integration of KRT6A expression with other molecular markers for comprehensive prognostic models
To effectively study KRT6A function, researchers need reliable methods to modulate its expression:
siRNA-Mediated Knockdown:
shRNA Stable Knockdown:
Provides longer-term KRT6A suppression for extended studies or in vivo xenograft models.
Allows selection of stable cell lines with consistent KRT6A downregulation.
CRISPR-Cas9 Gene Editing:
Enables complete knockout of KRT6A for studying loss-of-function effects.
Particularly valuable for distinguishing between the roles of highly homologous keratin family members.
Transgenic Mouse Models:
Overexpression Systems:
Lentiviral or plasmid-based overexpression of KRT6A in low-expressing cell lines.
Important for gain-of-function studies and rescue experiments.
For optimal experimental design, researchers should:
Confirm knockdown or overexpression efficiency by both qPCR and Western blot
Include appropriate controls (scrambled siRNA, empty vector)
Consider potential compensatory upregulation of other keratin family members
Validate phenotypic changes with multiple functional assays (proliferation, migration, apoptosis)
Understanding KRT6A regulation requires investigating multiple levels of control:
Transcriptional Regulation:
Promoter analysis using luciferase reporter assays to identify key transcription factor binding sites.
ChIP-seq to identify transcription factors directly binding to the KRT6A promoter region.
Analysis of epigenetic modifications (DNA methylation, histone modifications) at the KRT6A locus using bisulfite sequencing and ChIP.
Post-transcriptional Regulation:
microRNA targeting of KRT6A has been identified, with miR-31-5p shown to regulate KRT6A expression in bladder cancer .
RNA-binding protein analysis to identify factors affecting KRT6A mRNA stability or translation.
Alternative splicing analysis to detect potential isoform switching in disease states.
Post-translational Modifications:
Phosphorylation, glycosylation, and other modifications may affect KRT6A function and stability.
Mass spectrometry-based proteomics can identify specific modifications and their sites.
Site-directed mutagenesis to evaluate the functional impact of specific modifications.
Stress-Induced Regulation:
Pathway Analysis:
A comprehensive understanding of KRT6A regulation requires integration of these approaches, with attention to tissue-specific regulatory mechanisms that may differ between epithelial compartments.
Researchers working with KRT6A monoclonal antibodies may encounter several technical challenges:
Cross-reactivity with Other Keratins:
Challenge: Due to high homology among keratin family members, antibodies may cross-react with other keratins, particularly other type II keratins.
Solution: Validate antibody specificity using Western blotting against recombinant keratins and include KRT6A-knockout controls. Select antibodies raised against unique epitopes of KRT6A rather than conserved domains.
Epitope Masking in Fixed Tissues:
Challenge: Formalin fixation can mask KRT6A epitopes, reducing antibody binding.
Solution: Optimize antigen retrieval methods, testing both heat-induced (citrate or EDTA buffer) and enzymatic retrieval. For the KRT6A/2368 clone, a standardized protocol with 1-2 μg/ml antibody concentration has been validated for IHC-P .
Variability in Expression Levels:
Challenge: KRT6A expression varies widely across tissues and in response to stress or disease.
Solution: Include positive controls (hyperproliferative epithelium or head and neck squamous cell carcinomas) and negative controls. Use quantitative image analysis rather than subjective scoring when possible.
Flow Cytometry Optimization:
Challenge: As an intracellular protein, KRT6A requires cell permeabilization for detection by flow cytometry.
Solution: Use optimized permeabilization protocols (typically 0.1% saponin or commercial permeabilization buffers) and titrate antibody concentration (recommended 1-2 μg per million cells) .
Background Staining in IHC:
Challenge: High background can obscure specific KRT6A staining.
Solution: Include a blocking step with serum from the same species as the secondary antibody. Use specialized blocking reagents for endogenous peroxidase, biotin, and avidin when applicable.
Thorough optimization and standardization of protocols are essential for reliable KRT6A detection across different experimental platforms.
KRT6A serves as a valuable marker for identifying specific cell populations within complex tissues:
Single-cell Analysis Techniques:
Flow cytometry combining KRT6A with other markers can isolate specific subpopulations. Use 1-2 μg of antibody per million cells for optimal results .
Single-cell RNA sequencing to identify KRT6A-expressing cells and their transcriptional profiles within heterogeneous populations.
Mass cytometry (CyTOF) for high-dimensional analysis of KRT6A alongside numerous other markers.
Lineage Tracing in Developmental and Cancer Studies:
Spatial Analysis in Tissue Sections:
Multiplex immunofluorescence to co-localize KRT6A with other markers.
In situ hybridization for KRT6A mRNA can complement protein detection.
Digital spatial profiling for quantitative assessment of KRT6A in defined tissue regions.
Enrichment Strategies:
Laser capture microdissection of KRT6A+ regions followed by molecular analysis.
Magnetic or fluorescence-activated cell sorting using KRT6A antibodies for downstream applications.
Validation in Patient-derived Samples:
These approaches enable researchers to leverage KRT6A as a marker for identifying and characterizing specific cell populations with potential roles in tissue homeostasis, regeneration, and disease.
The involvement of KRT6A in multiple cancer types suggests several potential therapeutic strategies:
Targeting Radioresistance Mechanisms:
Cell-Type Specific Therapy:
Immunotherapeutic Approaches:
Development of KRT6A-targeted antibody-drug conjugates to deliver cytotoxic agents specifically to KRT6A-expressing cancer cells.
CAR-T cell therapy directed against surface-exposed KRT6A epitopes in cancer cells.
Evaluation of KRT6A as a tumor-associated antigen for cancer vaccine development.
Combination Strategies:
Considerations for Clinical Translation:
Stratification of patients based on KRT6A expression levels for precision medicine approaches.
Development of companion diagnostics to identify patients most likely to benefit from KRT6A-targeted therapies.
Monitoring of KRT6A expression as a biomarker of treatment response.
Future research should focus on understanding the mechanistic basis of KRT6A's role in cancer progression to identify the most promising therapeutic strategies and potential combination approaches.
Integrative multi-omics strategies offer powerful approaches to comprehensively understand KRT6A biology:
Genomic and Transcriptomic Integration:
Whole genome or exome sequencing to identify mutations or copy number variations affecting KRT6A in cancer.
RNA-seq to characterize expression patterns of KRT6A and co-regulated genes across disease states.
Analysis of alternative splicing events affecting KRT6A function using long-read sequencing technologies.
Proteomic Approaches:
Mass spectrometry-based interactome analysis to identify KRT6A binding partners in normal versus disease contexts.
Phosphoproteomics to map post-translational modifications regulating KRT6A function.
Spatial proteomics to visualize KRT6A distribution and interactions at subcellular resolution.
Epigenomic Profiling:
ATAC-seq to identify changes in chromatin accessibility at the KRT6A locus in disease states.
DNA methylation analysis using bisulfite sequencing to characterize epigenetic regulation.
ChIP-seq to map histone modifications and transcription factor binding at the KRT6A promoter.
Single-cell Multi-omics:
Single-cell RNA-seq combined with protein analysis to characterize heterogeneity in KRT6A-expressing populations.
Spatial transcriptomics to map KRT6A expression within tissue architecture.
Lineage tracing using genetic barcoding to track the fate of KRT6A+ cells in development and disease.
Network Biology Approaches:
Construction of gene regulatory networks centered on KRT6A using datasets like those analyzed in GEO (GSE73095 and GSE197236) .
Pathway enrichment analyses using GO and KEGG to place KRT6A in functional contexts .
Systems biology modeling to predict the impact of KRT6A perturbation on cellular phenotypes.
These multi-omics approaches can reveal the complex biological contexts in which KRT6A functions, potentially identifying novel therapeutic targets and biomarkers associated with KRT6A-expressing cells in disease.
To effectively leverage KRT6A in translational cancer research, researchers should consider these best practices:
Standardized Detection Methods:
Use validated antibody clones with known specificity, such as KRT6A/2368, at optimized concentrations (1-2 μg/ml for IHC-P, 1-2 μg per million cells for flow cytometry) .
Implement quantitative scoring systems for KRT6A immunohistochemistry to enable consistent cross-study comparisons.
Include appropriate positive and negative controls in all experiments.
Comprehensive Patient Sample Analysis:
Correlate KRT6A expression with clinicopathological features, including TNM staging and treatment outcomes .
Perform multivariate analysis to determine KRT6A's independent prognostic value, as demonstrated in lung cancer studies (HR: 1.751, 95% CI: 1.190–1.915, p=0.001) .
Consider tissue microarray approaches for high-throughput analysis across large patient cohorts.
Functional Validation:
Combine observational studies with functional experiments using knockdown and overexpression models.
Employ multiple methodologies to assess key phenotypes (proliferation, migration, invasion, radioresistance) as demonstrated in lung cancer studies .
Use both in vitro and in vivo models to validate findings, such as the K6a-tva transgenic mouse model .
Context-Specific Interpretation:
Translational Pipeline Development:
Establish standardized workflows from KRT6A detection in patient samples to functional validation.
Develop and validate clinical assays for KRT6A that meet regulatory standards.
Create integrated databases linking KRT6A expression with clinical outcomes and molecular profiles.