The KRT6A antibody is utilized across diverse experimental platforms:
Tissue Targets: Detects KRT6A in stratified epithelia (e.g., epidermis), hair follicles, and head/neck squamous cell carcinomas .
IF: Identifies KRT6A in cultured epithelial cells (1:50–1:200 dilution) .
WB: Validates KRT6A expression in lysates from squamous cell carcinomas .
Multiple clones and conjugation options are available, tailored to experimental needs:
Conjugation Options (Mouse Antibody KRT6A/2368) :
Conjugate | Ex/Em (nm) | Detection Channel | Laser Line | Dye Features |
---|---|---|---|---|
CF®488A | 490/515 | GFP, FITC | 488 | High brightness, photostability |
CF®647 | 650/665 | Cy®5 | 633–640 | Minimal non-specific binding |
Biotin | N/A | Streptavidin systems | N/A | Versatile labeling |
KRT6A is implicated in:
Pachyonychia Congenita: Mutations in KRT6A cause thickened nails and skin lesions .
Squamous Cell Carcinomas: Overexpressed in ~75% of head/neck SCCs, aiding in tumor stratification .
Wound Healing: Activated in keratinocytes post-injury, promoting epithelial repair .
Antimicrobial Activity: C-terminal peptides of KRT6A exhibit antimicrobial properties against bacterial pathogens .
Disease | KRT6A Role | References |
---|---|---|
Pachyonychia Congenita | Structural defects in keratin filaments | |
Squamous Cell Carcinoma | Tumor progression, hyper-proliferation | |
Wound Healing | Epithelial migration regulation |
Biotium. Cytokeratin 6A Monoclonal Mouse Antibody (KRT6A/2368). Link
Thermo Fisher. Cytokeratin 6A (KRT6A) (Basal Cell Marker) Recombinant Rabbit Monoclonal Antibody. Link
Abcam. Anti-Cytokeratin 6a antibody [KRT6A/2368]. Link
Cusabio. KRT6A Recombinant Monoclonal Antibody. Link
Antibodies.com. Anti-Cytokeratin 6A Antibody [KRT6A/2368]. Link
Creative Diagnostics. Rabbit Anti-Human KRT6A monoclonal antibody. Link
GeneCards. KRT6A Gene. Link
MyBioSource. Krt6a antibody. Link
The KRT6A recombinant monoclonal antibody is produced using in vitro expression systems. DNA sequences encoding KRT6A antibodies from immunoreactive rabbits are cloned. The immunogen employed is a synthesized peptide derived from the human KRT6A protein. The genes encoding the KRT6A antibodies are then inserted into plasmid vectors, which are subsequently transfected into host cells to enable antibody expression. Following expression, the KRT6A recombinant monoclonal antibody undergoes affinity-chromatography purification. Its functionality is rigorously tested in ELISA, IF, and FC applications, confirming reactivity with the human KRT6A protein.
KRT6A is a keratin protein that primarily provides structural support to epithelial tissues, including the skin, hair, and nails. Its functions are crucial for maintaining the structural integrity of these tissues and for protecting the body from external factors. Dysregulation of KRT6A and other keratins can lead to various skin and nail disorders.
KRT6A is an epidermis-specific type I keratin involved in wound healing. It plays a role in the activation of follicular keratinocytes after wounding, but does not significantly contribute to keratinocyte proliferation or migration. KRT6A participates in the regulation of epithelial migration by inhibiting the activity of SRC during wound repair.
KRT6A (Keratin 6A) is a 56 kDa protein that belongs to the type II keratin family. It provides structural support to epithelial tissues, including skin, hair, and nails, playing an essential role in maintaining tissue integrity and protection against external factors. In humans, multiple isoforms of Cytokeratin 6 (6A-6F) exist, each encoded by highly homologous genes with distinct tissue expression patterns, with KRT6A being the dominant form in epithelial tissue .
Research interest in KRT6A stems from its critical role in normal epithelial biology and pathological conditions. KRT6A is particularly valuable as a marker of hyperproliferative keratinocytes and is upregulated during wound healing processes. The protein is also strongly expressed in approximately 75% of head and neck squamous cell carcinomas, making it significant for cancer research applications .
Polyclonal KRT6A antibodies, such as rabbit polyclonal antibodies to keratin 6A, are derived from multiple B-cell lineages and recognize different epitopes on the KRT6A protein. These are typically produced by immunizing animals with recombinant proteins or synthetic peptides derived from the KRT6A sequence .
In contrast, recombinant monoclonal KRT6A antibodies are produced using in vitro expression systems. The process involves cloning DNA sequences of KRT6A antibodies from immunoreactive animals (often rabbits), inserting these genes into plasmid vectors, and transfecting host cells for expression. The resulting antibodies recognize a single epitope with high specificity .
The key methodological advantage of recombinant monoclonal antibodies is their consistent specificity and reproducibility across different batches, which is particularly valuable for longitudinal studies where antibody performance consistency is critical .
KRT6A recombinant monoclonal antibodies serve multiple research purposes across different experimental techniques:
These applications are particularly valuable in epithelial biology research, wound healing studies, and cancer research focusing on squamous cell carcinomas and other epithelial-derived malignancies .
Optimization of KRT6A antibody concentration requires a systematic approach that varies by application:
For Western blotting (WB), begin with mid-range dilutions (e.g., 1:1000) and adjust based on signal strength. Remember that over-concentrated antibody can lead to high background and non-specific binding, while over-diluted antibody may result in weak or undetectable signals .
When switching between applications (e.g., from IF to WB), do not assume optimal dilutions will transfer directly. Each application requires separate optimization due to differences in sample preparation, protein conformation, and detection systems.
For tissue-specific studies, consider that KRT6A expression varies significantly between tissue types. Tissues with high baseline expression (hair follicles, stratified epithelia) may require higher antibody dilutions than tissues with lower or inducible expression .
Robust experimental design with appropriate controls is essential for reliable interpretation of results when using KRT6A antibodies:
Positive controls:
Cell lines or tissues with known KRT6A expression (e.g., HaCaT keratinocytes, squamous cell carcinoma lines)
Tissues with well-documented KRT6A expression patterns such as hair follicles and suprabasal cells of stratified epithelia
Negative controls:
Tissues or cells known to lack KRT6A expression
Primary antibody omission controls to assess secondary antibody specificity
Isotype controls matching the KRT6A antibody's host species and immunoglobulin class
Specificity controls:
Peptide competition/blocking experiments to confirm binding specificity
Validation in KRT6A knockout or knockdown systems when available
Comparison with alternative KRT6A antibody clones recognizing different epitopes
These controls help distinguish true KRT6A signal from technical artifacts and enable confident interpretation of experimental outcomes, particularly in tissues with complex keratin expression profiles where cross-reactivity is a concern.
Sample preparation significantly impacts KRT6A detection quality across different experimental approaches:
For immunohistochemistry and immunofluorescence:
Fixation with 4% paraformaldehyde preserves epitope accessibility for many KRT6A antibodies
Antigen retrieval is usually necessary—heat-induced epitope retrieval (HIER) using citrate buffer (pH 6.0) or EDTA buffer (pH 9.0) is often effective
Over-fixation with formalin can mask KRT6A epitopes; optimize fixation time for your specific tissue
For skin and other keratinized tissues, deparaffinization must be complete to prevent non-specific binding
For flow cytometry:
Permeabilization is essential as KRT6A is an intracellular protein; saponin or methanol-based permeabilization protocols are typically effective
Gentle fixation (0.5-2% paraformaldehyde) often provides better results than harsher methods
Single-cell suspensions from epithelial tissues require careful enzymatic digestion to preserve epitopes while achieving adequate dissociation
For Western blotting:
Standard RIPA or NP-40 based lysis buffers with protease inhibitors are generally suitable
Heat denaturation (95-100°C for 5 minutes) in reducing sample buffer is typically required
For highly keratinized tissues, consider including chaotropic agents like urea in your lysis buffer to improve solubilization
Validation of KRT6A antibody specificity requires multiple complementary approaches:
Molecular validation:
Western blot analysis should show a single band at the expected molecular weight of 56 kDa for KRT6A
Mass spectrometry confirmation of immunoprecipitated proteins can provide definitive identification
RNA interference (siRNA/shRNA) knockdown of KRT6A should result in corresponding reduction of antibody signal
Cross-reactivity assessment:
Testing against similar keratins, particularly other type II keratins and other KRT6 isoforms (KRT6B-F), is essential due to high sequence homology
Systematic epitope mapping to confirm the specific region of KRT6A recognized by the antibody
Expression correlation between protein detection (antibody-based) and mRNA levels (qPCR or RNA-seq)
Comparison across techniques:
Concordant results across multiple detection methods (IHC, IF, WB, FC) increase confidence in specificity
Comparison with alternative antibody clones targeting different KRT6A epitopes
Consistent results with both N-terminal and C-terminal targeting antibodies provide strong validation
Documentation of these validation steps is critical for research reproducibility and should be included in publications using KRT6A antibodies.
Ensuring consistency between antibody batches is essential for research continuity and reproducibility. Key parameters to evaluate include:
Binding characteristics:
Affinity measurements using surface plasmon resonance (SPR) or bio-layer interferometry (BLI) to determine KD values
Epitope binning to confirm consistent recognition of the same KRT6A region
Titration curves in validated positive control samples to assess binding consistency
Physicochemical properties:
Size exclusion chromatography to assess aggregation state
Isoelectric focusing to confirm charge profile consistency
Glycosylation analysis, particularly important for Fc-mediated functions
Thermal stability assessment via differential scanning calorimetry
Functional performance:
Side-by-side comparison of sensitivity and specificity in application-specific contexts
Signal-to-noise ratio in identical positive control samples
Background levels in negative control samples
When significant manufacturing changes occur, more extensive comparability assessment may be required. According to accepted comparability principles for recombinant monoclonal antibodies, changes in post-translational modifications, like glycosylation patterns, can impact antibody functionality and must be carefully evaluated .
Integration of KRT6A antibodies with single-cell technologies enables powerful insights into heterogeneous epithelial populations:
Single-cell mass cytometry (CyTOF): Metal-conjugated KRT6A antibodies can be incorporated into CyTOF panels allowing simultaneous detection of KRT6A alongside numerous other proteins with minimal spectral overlap. This approach is particularly valuable for differentiating epithelial subpopulations in complex tissues like epidermis or tumor microenvironments.
Single-cell RNA-seq validation: KRT6A antibodies serve as important validation tools for single-cell transcriptomics findings. Protein-level confirmation using index-sorted flow cytometry with KRT6A antibodies can validate mRNA expression patterns identified by scRNA-seq, addressing potential discrepancies between transcript and protein abundance.
Spatial proteomics: Combining KRT6A immunofluorescence with multiplexed imaging technologies (e.g., cyclic immunofluorescence, CODEX, or Imaging Mass Cytometry) enables spatial mapping of KRT6A-expressing cells in relation to their microenvironment. This is particularly valuable for studying wound healing dynamics or tumor invasion patterns.
Live-cell imaging: For dynamic studies, non-perturbing KRT6A antibody fragments can be used to track changes in keratin organization during cellular processes like migration or division. This requires careful validation to ensure that antibody binding doesn't interfere with KRT6A function.
When designing these advanced applications, consider that KRT6A is part of an expression program often involving KRT16 and other markers of activated keratinocytes, which provides opportunities for creating informative multi-parameter panels .
Studying KRT6A in disease contexts requires specific methodological considerations:
Wound healing models:
Temporal analysis is critical as KRT6A expression is dynamically regulated during wound healing
Standardized wounding protocols are essential for reproducible results
Consider co-staining with proliferation markers (Ki67, EdU) and other activation keratins (KRT16) to contextualize KRT6A expression
In vivo wound models require consistent sampling methods relative to wound margins
Cancer research applications:
In squamous cell carcinomas, KRT6A expression varies by differentiation status and may correlate with invasive potential
Quantitative analysis should include both expression intensity and percentage of positive cells
Consider tumor heterogeneity—compare KRT6A expression at invasive fronts versus tumor centers
Patient-derived xenograft models may better preserve KRT6A expression patterns than cell line xenografts
Inflammatory skin conditions:
Baseline KRT6A expression may vary between anatomical sites; use site-matched controls
Standardize biopsy processing timelines as KRT6A can be rapidly induced ex vivo
Co-staining with inflammatory markers helps contextualize KRT6A upregulation
Consider epidermal thickness and architecture changes when quantifying KRT6A expression
For all disease models, remember that KRT6A expression represents a stress response program rather than a specific disease marker, so contextual interpretation alongside other biomarkers is essential .
Discrepancies between KRT6A protein (detected by antibodies) and mRNA expression are common and can arise from several mechanisms:
Methodological considerations:
Antibody specificity: Confirm whether the antibody differentiates between KRT6A and other KRT6 isoforms (KRT6B-F), as antibody cross-reactivity might detect proteins encoded by different transcripts
Primer specificity: Similarly, RT-PCR primers may not distinguish between highly homologous KRT6 isoforms unless carefully designed
Detection sensitivity: Protein and RNA detection methods have different sensitivity thresholds; calibrate expectations accordingly
Biological explanations:
Post-transcriptional regulation: KRT6A mRNA may be subject to miRNA-mediated repression or altered stability in certain contexts
Translation efficiency: Changes in translation machinery can affect protein synthesis rates without altering transcript levels
Protein stability: KRT6A protein has different turnover kinetics than its mRNA; stress conditions may stabilize the protein
Temporal dynamics: In wounding or other rapid response scenarios, protein levels may lag behind mRNA induction
Resolution approach:
Validate findings using multiple antibody clones and RNA detection methods
Perform time-course experiments to capture potential temporal discrepancies
Investigate post-transcriptional mechanisms through targeted experiments (e.g., miRNA inhibition, protein stability assays)
Consider isoform-specific analyses to distinguish between different KRT6 variants
Use absolute quantification methods rather than relative comparisons when possible
Careful documentation of these discrepancies can yield novel insights into KRT6A regulation in your experimental system.
Accurate interpretation of KRT6A expression patterns requires consideration of several biological and technical factors:
Tissue-specific baseline expression:
KRT6A shows constitutive expression in certain tissues (hair follicles, oral mucosa, esophageal epithelium) but is induced by stress in others (epidermis)
Constitutive vs. inducible expression may involve different regulatory mechanisms
Reference unexposed/unstressed tissue sections from the same anatomical site for proper baseline comparison
Cellular localization considerations:
Normal KRT6A distribution is cytoplasmic with filamentous patterns
Altered patterns (aggregation, cortical redistribution) may indicate cellular stress or pathology
Subcellular localization should be documented alongside expression levels
Pattern analysis beyond intensity:
Spatial distribution within tissue (e.g., confined to suprabasal layers vs. pan-epithelial)
Homogeneity vs. heterogeneity of expression within a given cell population
Relationship to tissue architecture (e.g., expression relative to basement membrane)
Co-expression context:
KRT6A typically pairs with KRT16 during hyperproliferation responses
Co-expression with other stress markers (e.g., SPRR proteins, involucrin) provides context
Relationship to proliferation markers (Ki67, PCNA) helps distinguish hyperproliferation from other stress responses
Technical considerations:
Section orientation affects observed patterns in stratified epithelia
Edge effects in tissue sections can show artifactual KRT6A induction
Autofluorescence from keratin-rich structures requires appropriate controls
These considerations help distinguish pathological KRT6A expression from normal variation and technical artifacts .
Understanding potential sources of error helps ensure reliable KRT6A detection:
Common causes of false positives:
Cross-reactivity with other keratin family members, particularly KRT6B-F isoforms which share high sequence homology with KRT6A
Edge artifacts in tissue sections, as mechanical stress during sectioning can induce KRT6A expression
Endogenous peroxidase activity in tissues causing non-specific signal in HRP-based detection systems
Inadequate blocking, particularly in keratinized tissues which can bind antibodies non-specifically
Secondary antibody cross-reactivity with endogenous immunoglobulins in the sample
Common causes of false negatives:
Epitope masking due to overfixation, particularly with formalin-fixed tissues
Inadequate antigen retrieval for immunohistochemistry applications
Protein degradation during sample preparation, especially in post-mortem tissues
Sub-optimal permeabilization when detecting intracellular KRT6A
Competition between endogenous KRT6A-binding proteins and antibodies for the same epitope
Methodological solutions:
Use multiple antibody clones targeting different KRT6A epitopes to confirm findings
Include comprehensive positive and negative controls in every experiment
Optimize fixation and antigen retrieval protocols specifically for KRT6A detection
Implement isotype controls and peptide competition controls to verify specificity
Consider alternative detection methods (e.g., RNA in situ hybridization) for validation
Careful troubleshooting and appropriate controls minimize both false positive and negative results, increasing data reliability.
Contradictory results between different antibody clones require systematic investigation:
Identify the exact epitopes recognized by each antibody clone when possible
Determine if the antibodies are isoform-specific or recognize multiple KRT6 isoforms
Review validation data for each clone, including western blot profiles and knockout controls
Test whether discrepancies are application-specific (e.g., one antibody works in IHC but not WB)
Evaluate sensitivity differences through dilution series in positive control samples
Assess reproducibility by repeating experiments in multiple laboratories if possible
Consider epitope masking due to protein-protein interactions in specific cellular contexts
Investigate post-translational modifications that might affect epitope recognition
Evaluate isoform expression through isoform-specific PCR to complement protein data
Use orthogonal methods (mass spectrometry, RNA-seq) to validate protein identity and abundance
Consider recombinant expression of KRT6A constructs as defined positive controls
Implement genetic approaches (CRISPR/Cas9 modification of KRT6A) for definitive validation
Document discrepancies transparently in publications rather than selecting only confirming data
Consider reporting ranges of expression based on multiple antibodies rather than absolute values
Discuss potential biological significance of epitope-specific detection differences
This structured approach transforms contradictory results into opportunities for deeper biological insights about KRT6A structure and function.
KRT6A antibodies serve several important functions in therapeutic research contexts:
Biomarker development:
KRT6A overexpression in squamous cell carcinomas makes it a potential prognostic or predictive biomarker
Development of standardized immunohistochemical assays using recombinant monoclonal antibodies ensures reproducible patient stratification
Correlation of KRT6A expression patterns with response to specific treatments can guide personalized medicine approaches
Target validation:
While KRT6A itself is primarily an intracellular protein not directly targetable by conventional antibody therapeutics, it serves as a marker for validating other therapeutic approaches
KRT6A antibodies can monitor treatment effects on keratinocyte activation states in preclinical models
Detection of circulating KRT6A (released from damaged epithelial cells) might serve as a pharmacodynamic biomarker
Therapeutic antibody development:
Lessons from KRT6A recombinant antibody development inform broader therapeutic antibody engineering
Analytical comparability studies on KRT6A antibodies provide methodological frameworks applicable to therapeutic antibody manufacturing changes
Post-translational modification analysis techniques developed for KRT6A antibodies apply to therapeutic antibody characterization
Drug delivery research:
KRT6A expression patterns can identify target cell populations for epithelial-directed therapeutics
Antibody-drug conjugate research may utilize targeting of cell surface proteins co-expressed with KRT6A
Understanding KRT6A dynamics in response to therapeutic intervention provides mechanistic insights into drug action
These research applications bridge basic KRT6A biology with translational medicine, particularly in dermatological conditions and epithelial malignancies.
Multiplex imaging with KRT6A antibodies requires specific optimization strategies:
Panel design considerations:
KRT6A is often co-expressed with KRT16 and other stress-induced keratins; plan panels to capture this biological relationship
Include differentiation markers (e.g., KRT1, KRT10, involucrin) to contextualize KRT6A expression within epithelial maturation states
When studying wound healing or cancer, incorporate proliferation markers (Ki67) and inflammatory signals (cytokines, immune cell markers)
Technical optimization:
Signal strength balancing: KRT6A often gives strong signals that may overwhelm other markers; titrate antibody concentration carefully
Sequence planning: In cyclic immunofluorescence protocols, test whether KRT6A detection is affected by preceding staining/stripping cycles
Spectral considerations: Select fluorophores that minimize spectral overlap with tissue autofluorescence, particularly in keratinized tissues
Validation requirements:
Perform single-stain controls to establish baseline signal and crosstalk parameters
Include serial sections stained with individual antibodies to validate multiplex findings
Implement computational approaches to address spectral overlap and autofluorescence
Analysis approaches:
Develop quantitative algorithms that capture both expression levels and subcellular distribution patterns
Consider spatial relationships between KRT6A-positive cells and other cell populations
Integrate multiplex imaging data with transcriptomic or proteomic datasets for comprehensive analysis
Emerging technologies:
Adapter-based multiplexing systems allow simultaneous use of multiple mouse monoclonal antibodies, expanding panel design options
Mass cytometry imaging (IMC) and Multiplexed Ion Beam Imaging (MIBI) overcome fluorescence limitations for highly multiparametric analysis
Digital spatial profiling technologies combine KRT6A antibody detection with region-specific molecular profiling
These considerations enable integration of KRT6A analysis into complex tissue microenvironment studies.