KRT14 antibodies are highly specific probes designed to target keratin 14 in various experimental and clinical contexts. Key features include:
LL002 (Mouse Monoclonal): Widely cited (255+ publications), validated in KRT14 KO cells, and used for IHC, WB, and ICC .
SP53 (Rabbit Monoclonal): Detects KRT14 in human, mouse, and rat samples, validated via KO cell lines and confocal microscopy .
MSVA-614R (Rabbit Monoclonal): Targets cytoplasmic KRT14 in squamous epithelia and basal cells; validated against RNA/protein databases .
KRT14 antibodies have been pivotal in studying epithelial pathophysiology and cancer biology:
Skin Integrity: KRT14 forms heterodimers with keratin 5 (KRT5) to create intermediate filaments, critical for epidermal resilience. Antibodies detect structural defects in epidermolysis bullosa simplex .
Bladder Regeneration: KRT14+ basal cells contribute to bladder repair and neoplastic transformation, as shown in lineage-tracing studies .
Antibody validation is critical for reproducibility:
Knockout Validation: SP53 and LL002 clones show loss of signal in KRT14 KO A431 cells, confirming specificity .
Cross-Reactivity Checks: MSVA-614R excludes non-epithelial tissues (e.g., liver, lung) and avoids false positives in mesenchymal cells .
Protein Atlas Alignment: Reactivity mirrors RNA/protein expression in squamous epithelia, basal cells, and placental tissues .
Skin Disorders: Detects KRT14 mutations in epidermolysis bullosa simplex and other blistering diseases .
Cancer Prognostics: KRT14 expression correlates with metastatic potential in TNBC and ovarian cancer, aiding biomarker discovery .
KRT14 (Keratin 14) is a type I intermediate filament protein of approximately 51.6 kilodaltons that forms part of the cytoskeletal structure in epithelial cells. It is primarily expressed in the basal cells of stratified epithelia, including the skin, and in basal-like subtypes of certain cancers . KRT14 partners with Keratin 5 (KRT5) to form heterodimers that are essential for maintaining cellular structural integrity.
The biological significance of KRT14 includes:
Providing mechanical support and resilience to epithelial cells
Playing crucial roles in cell signaling and differentiation processes
Serving as a diagnostic marker for specific epithelial cell types and cancers
Mutations in KRT14 are associated with epidermolysis bullosa simplex, a skin fragility disorder
KRT14 expression follows a specific pattern in normal tissues:
Positive KRT14 Expression:
Basal cells of stratified epithelia
Squamous epithelia (skin, esophagus, cervix)
Hair follicles
Salivary glands
Basal cells of prostate
Basal cells of respiratory epithelium
Negative KRT14 Expression:
Lung parenchyma
Liver
Pancreas
Epididymis
Testis
Urothelium
Gastrointestinal epithelial cells
Brunner glands
Fallopian tube
Trophoblastic cells of the placenta
Adrenal and parathyroid glands
Brain tissues
Hematopoietic cells
This expression pattern is supported by both immunohistochemical findings and RNA expression data from the Human Protein Atlas, FANTOM5, and GTEx projects .
KRT14 antibodies perform reliably in the following applications:
For optimal results, validation in your specific experimental system is recommended, as antibody performance can vary between different clones and suppliers .
The choice between monoclonal and polyclonal KRT14 antibodies depends on your specific research needs:
Monoclonal Antibodies (e.g., RCK107, LL002):
Advantages: High specificity, consistent lot-to-lot performance, reduced background
Best for: Clinical diagnostics, quantitative studies, scenarios requiring high reproducibility
Polyclonal Antibodies:
Advantages: Recognize multiple epitopes, higher sensitivity, better for detecting denatured proteins
Best for: Western blotting, detecting low-abundance targets, applications with potentially altered epitopes
Recommended for: Initial protein characterization studies, WB applications
When selecting an antibody, also consider:
Species reactivity needed (human, mouse, rat, etc.)
Required applications (different antibodies may perform better in WB vs. IHC)
Epitope location (N-terminal, C-terminal, or internal regions)
Proper validation ensures reliable results with KRT14 antibodies:
Positive and negative tissue controls:
Knockout/knockdown validation:
Western blot confirmation:
Antibody comparison:
Test multiple antibodies against the same samples
Look for consistent staining patterns across different antibody clones
RNA correlation:
KRT14 has been identified as a marker for "leader cells" in ovarian cancer invasion, representing a significant application in cancer research:
Methodology for leader cell identification:
Isolation of migratory cells:
KRT14 expression analysis:
Functional validation:
Co-culture experiments:
This approach has demonstrated that KRT14-positive cells are crucial for tumor invasion processes, suggesting their potential as therapeutic targets or prognostic markers .
KRT14 engages in important protein interactions, such as with RIPK4, which may regulate keratin turnover during keratinocyte differentiation . Here's an optimized co-immunoprecipitation (co-IP) protocol:
Materials needed:
Anti-KRT14 antibody (preferably monoclonal for specificity)
Protein A/G beads
Cell lysis buffer (non-denaturing)
Wash buffers
SDS-PAGE materials
Protocol steps:
Cell preparation:
Cell lysis:
Lyse cells in non-denaturing buffer containing:
50 mM Tris-HCl pH 7.5
150 mM NaCl
1% NP-40 or 0.5% Triton X-100
Protease and phosphatase inhibitors
Perform lysis on ice for 30 minutes with gentle agitation
Centrifuge at 14,000 × g for 15 minutes at 4°C
Pre-clearing (reduces non-specific binding):
Incubate lysate with Protein A/G beads for 1 hour at 4°C
Remove beads by centrifugation
Immunoprecipitation:
Add 2-5 μg of anti-KRT14 antibody to pre-cleared lysate
Incubate overnight at 4°C with gentle rotation
Add fresh Protein A/G beads and incubate for 2-4 hours at 4°C
Collect beads by centrifugation and wash 4-5 times with wash buffer
Elution and analysis:
For KRT14-RIPK4 interaction specifically, both proteins can be cloned into appropriate expression vectors (e.g., GST-KRT14 and FLAG-RIPK4) to facilitate detection of the interaction .
Dual immunofluorescence with KRT14 antibodies allows visualization of its relationship with other proteins. This is particularly valuable for studying epithelial architecture and cellular interactions.
Optimized protocol:
Sample preparation:
Blocking:
Primary antibody incubation:
Secondary antibody incubation:
Nuclear counterstaining and mounting:
Counterstain nuclei with DAPI (1:1000) for 5 minutes
Mount with anti-fade mounting medium
Tips for successful dual staining:
Validate antibodies individually before performing dual staining
Include appropriate controls (single stains, no primary antibody controls)
Select fluorophores with minimal spectral overlap
When studying KRT14 with other keratins, be particularly careful about cross-reactivity
Quantitative analysis of KRT14 expression in heterogeneous tissues requires specialized approaches:
1. Digital Pathology and Image Analysis:
Scan immunostained slides using digital slide scanners
Use image analysis software to:
Identify KRT14-positive cells based on staining intensity
Quantify percentage of positive cells
Measure staining intensity (low, medium, high)
Assess distribution patterns within the tissue
2. Tissue Microdissection and Molecular Analysis:
Use laser capture microdissection to isolate KRT14-positive regions
Extract RNA/protein from microdissected samples
Quantify KRT14 expression by qRT-PCR or Western blotting
Compare with adjacent KRT14-negative regions
3. Single-cell Analysis Approaches:
Prepare single-cell suspensions from heterogeneous tissues
Perform intracellular staining for KRT14
Analyze by flow cytometry to quantify:
Percentage of KRT14+ cells
Expression level (Mean Fluorescence Intensity)
Co-expression with other markers
4. Multiplexed Immunofluorescence:
Perform multiplexed staining (KRT14 plus 3-5 additional markers)
Image using multispectral imaging systems
Apply computational analysis to:
Identify cell types
Quantify marker expression
Map spatial relationships
5. Data Representation and Analysis:
Analysis Method | Advantages | Limitations | Best For |
---|---|---|---|
H-score (0-300) | Simple, widely accepted | Subjective component | Clinical studies |
Percentage positivity | Clear, reproducible | Loses intensity information | Population analysis |
Mean fluorescence intensity | Quantitative, sensitive | Requires calibration | Expression level comparisons |
Spatial analysis | Maps tissue architecture | Technically complex | Tumor microenvironment studies |
Post-translational modifications (PTMs) of KRT14 can significantly impact antibody binding and experimental results:
Common PTMs affecting KRT14:
Phosphorylation (especially during stress responses)
Glycosylation
Ubiquitination (during degradation)
Acetylation
SUMOylation
Effects on antibody binding:
Epitope masking:
PTMs can physically block antibody access to epitopes
Phosphorylation of sites within or adjacent to the antibody epitope may prevent recognition
Solution: Use antibodies raised against different epitopes or specific PTM-state antibodies
Conformational changes:
PTMs can alter KRT14 protein folding and structure
This may expose or hide epitopes recognized by certain antibodies
Solution: Use denaturing conditions for applications like Western blotting
Altered migration patterns:
Phosphorylation typically causes slower migration on SDS-PAGE
May result in band shifts from the expected 50 kDa
Solution: Treat samples with phosphatases before SDS-PAGE if phosphorylation is suspected
Recommendations for experiments:
Selection of appropriate antibodies:
For general KRT14 detection: Use antibodies targeting conserved regions less likely to be modified
For PTM studies: Use modification-specific antibodies (e.g., phospho-KRT14)
Sample preparation considerations:
Include phosphatase inhibitors in lysis buffers to preserve phosphorylation state
Consider native vs. denaturing conditions based on experimental goals
Control experiments:
Treat samples with enzymes that remove specific PTMs
Compare antibody reactivity before and after treatment
Include both negative and positive controls with known PTM status
Understanding the impact of PTMs on KRT14 is particularly important when studying cellular stress responses, as keratin phosphorylation often increases during stress conditions.
Identifying and resolving false results is crucial for reliable KRT14 detection:
False-positive causes and solutions:
Cross-reactivity with other keratins:
Excessive antibody concentration:
Inadequate blocking:
Endogenous peroxidase activity:
Problem: Can cause background in IHC using HRP-based detection
Solution: Block endogenous peroxidase with H₂O₂ treatment before antibody incubation
False-negative causes and solutions:
Epitope masking during fixation:
Problem: Overfixation can cross-link proteins and hide epitopes
Solution: Optimize fixation time; perform antigen retrieval (heat-induced or enzymatic)
Inappropriate antigen retrieval:
Problem: Insufficient retrieval of epitopes in FFPE tissues
Solution: Test different retrieval methods (citrate, EDTA, or Tris buffers at various pH)
Degraded antibody:
Sample-specific issues:
Problem: Heterogeneous expression or low abundance in certain samples
Solution: Use amplification systems (tyramide signal amplification) for low-expressing samples
Validation checklist:
Test antibody on positive and negative control tissues
Compare staining pattern with literature and Human Protein Atlas data
Confirm specificity using knockout/knockdown controls when possible
Detection of KRT14 in FFPE tissues requires optimized protocols for successful immunohistochemistry:
Standard protocol for FFPE tissues:
Deparaffinization and rehydration:
Heat slides at 60°C for 1 hour
Deparaffinize in xylene (3 × 5 minutes)
Rehydrate through graded alcohols to water
Antigen retrieval:
Heat-induced epitope retrieval (HIER) using:
Citrate buffer (pH 6.0) or EDTA buffer (pH 9.0)
Pressure cooker: 125°C for 30-40 seconds or
Microwave: 20 minutes at sub-boiling temperature
Cool slides to room temperature (20 minutes)
Blocking steps:
Endogenous peroxidase block: 3% H₂O₂ for 10 minutes
Protein block: 10% normal goat serum for 30 minutes
Primary antibody incubation:
Detection system:
Polymer-based detection systems generally work well
For chromogenic detection: DAB substrate (brown)
For fluorescent detection: Use fluorophore-conjugated secondary antibodies
Counterstain with hematoxylin (for chromogenic) or DAPI (for fluorescent)
Key optimization considerations:
Antigen retrieval method: Test both citrate and EDTA-based buffers to determine optimal retrieval
Antibody dilution: Titrate antibody to find optimal concentration for your specific samples
Incubation time and temperature: Adjust based on staining intensity and background
Detection system sensitivity: Consider amplification systems for weak expression
Expected results:
KRT14-positive cells show cytoplasmic/membrane staining
Positive control (skin) should show strong basal cell staining
Negative controls (liver, lung) should show no specific staining
Flow cytometry with KRT14 antibodies requires special considerations due to its intracellular location:
Optimized protocol for flow cytometry:
Cell preparation:
Harvest cells (enzymatic or mechanical dissociation)
Wash in cold PBS with 2% FBS
Adjust to 1 × 10⁶ cells per sample
Fixation and permeabilization:
Blocking:
Block with 10% serum (from secondary antibody species) for 30 minutes
This reduces non-specific binding
Antibody staining:
Analysis considerations:
Use appropriate compensation controls if multiplexing
Include FMO (Fluorescence Minus One) controls
Use isotype controls to assess background staining
Troubleshooting flow cytometry issues:
Problem | Possible Cause | Solution |
---|---|---|
Low signal | Insufficient permeabilization | Optimize permeabilization time/reagent |
Antibody concentration too low | Increase antibody concentration | |
High background | Non-specific binding | Increase blocking time/concentration |
Antibody concentration too high | Titrate antibody to optimal concentration | |
Poor separation | Suboptimal fluorophore choice | Select brighter fluorophores |
Insufficient compensation | Adjust compensation settings |
Dual staining strategies:
For co-expression analysis, consider these marker combinations:
KRT14 + KRT5 (basal cell identification)
KRT14 + Proliferation markers (Ki67)
KRT14 + Differentiation markers (Involucrin)
KRT14 + Cancer stem cell markers (CD44, ALDH)
Certain sample types present challenges for KRT14 detection, requiring specialized approaches:
1. Highly keratinized tissues (e.g., skin):
Challenge: High background due to endogenous keratins
Solutions:
Extended blocking (2-3 hours) with 10% serum + 1% BSA
Use monoclonal antibodies with high specificity
Include highly stringent washing steps (0.1% Tween-20 in PBS)
Pre-absorb antibodies with keratin-rich extracts
2. Archival FFPE tissues:
Challenge: Epitope degradation or masking
Solutions:
Extended antigen retrieval (30-40 minutes)
Test multiple antibody clones targeting different epitopes
Use signal amplification systems (tyramide signal amplification)
Consider dual antibody approach (cocktail of two KRT14 antibodies)
3. Frozen tissue sections:
Challenge: Compromised morphology, high background
Solutions:
Optimal cutting temperature (8-10 μm sections)
Post-fixation with acetone (10 minutes at -20°C)
Pre-treatment with 0.3% Triton X-100 (5 minutes)
Extended washing steps to remove OCT compound
4. Cell cultures with low KRT14 expression:
Challenge: Weak signal, difficult detection
Solutions:
Use high-sensitivity detection systems
Longer primary antibody incubation (overnight at 4°C)
Consider more sensitive antibody clones
Use signal amplification techniques
5. Tissue microarrays (TMAs):
Challenge: Heterogeneous expression, small sample size
Solutions:
Optimize protocols on whole sections before TMA analysis
Include positive control cores
Consider duplicate/triplicate cores for each sample
Adjust scoring systems to account for heterogeneity
For all challenging samples, pilot studies with multiple antibody clones, dilutions, and detection systems are recommended to determine optimal conditions.
KRT14 antibodies have diverse applications in cancer research, particularly for studying tumor progression and classification:
1. Tumor classification and subtyping:
Methodology: IHC staining of tissue microarrays or whole sections
Applications:
2. Identification of cancer stem cells and leader cells:
Methodology: Dual immunofluorescence, flow cytometry, or single-cell sequencing
Applications:
3. Epithelial-mesenchymal transition (EMT) studies:
Methodology: Sequential staining of tumor sections or time-course analysis
Applications:
Track changes in KRT14 expression during EMT
Correlate with expression of EMT markers (E-cadherin, Vimentin)
Study partial EMT states in collective invasion
4. Therapeutic response prediction:
Methodology: Pre- and post-treatment biopsy analysis
Applications:
Assess changes in KRT14+ cell populations after therapy
Correlate KRT14 expression with resistance to specific treatments
Identify KRT14+ residual disease after treatment
5. Metastasis research:
Methodology: Comparative analysis of primary tumors and metastatic sites
Applications:
Determine if KRT14+ cells are enriched in metastatic lesions
Study if KRT14 expression changes during metastatic colonization
Evaluate the role of KRT14+ cells in metastasis initiation
3D spheroid invasion assay:
Mesothelial clearance assay:
Understanding KRT14's interactions with other proteins provides insights into its functions beyond structural support:
1. Proximity Ligation Assay (PLA):
Principle: Detects protein interactions in situ with single-molecule sensitivity
Methodology:
Incubate samples with primary antibodies against KRT14 and partner protein
Add PLA probes (secondary antibodies with oligonucleotide tags)
If proteins are in proximity (<40 nm), oligonucleotides can be ligated
Amplify signal by rolling circle amplification
Detect fluorescent spots indicating interaction sites
2. FRET (Förster Resonance Energy Transfer):
Principle: Energy transfer between fluorophores when proteins are in close proximity
Methodology:
Label KRT14 and interaction partner with donor/acceptor fluorophores
Measure energy transfer using specialized microscopy
Calculate FRET efficiency to quantify interaction strength
Perform acceptor photobleaching to confirm specificity
3. Co-Immunoprecipitation with specific controls:
Principle: Physical isolation of protein complexes
Methodology:
4. Yeast Two-Hybrid (Y2H) screening:
Principle: Identifies direct protein-protein interactions
Methodology:
5. Bimolecular Fluorescence Complementation (BiFC):
Principle: Protein interaction brings together fragments of fluorescent protein
Methodology:
Fuse KRT14 and partner protein with complementary fragments of fluorescent protein
Co-express in relevant cell types
Interaction brings fragments together, reconstituting fluorescence
Visualize by fluorescence microscopy
Clone KRT14 into GST-tagged expression vectors and RIPK4 into FLAG-tagged vectors
Express in HeLa or HaCaT cells
Perform co-immunoprecipitation with anti-FLAG antibodies
Detect pulled-down KRT14 using anti-GST antibodies
This approach revealed that RIPK4 may regulate keratin turnover required for keratinocyte differentiation
Integrating KRT14 antibodies into single-cell technologies enables unprecedented insights into cellular heterogeneity:
1. Single-cell proteomics with antibody-based approaches:
CITE-seq (Cellular Indexing of Transcriptomes and Epitopes by Sequencing):
Label cells with oligo-tagged KRT14 antibodies
Perform single-cell RNA sequencing
Simultaneously measure KRT14 protein and transcriptome
Reveals potential post-transcriptional regulation
CyTOF (Mass cytometry):
Label with metal-conjugated KRT14 antibodies
Allows simultaneous detection of 40+ proteins
No fluorescence overlap issues
Ideal for deep phenotyping of KRT14+ subpopulations
2. Spatial transcriptomics with protein detection:
Visium with immunofluorescence:
Perform KRT14 immunostaining on tissue sections
Capture spatial transcriptomics data
Correlate KRT14 protein expression with local transcriptome
Reveals tissue microenvironments around KRT14+ cells
CODEX (CO-Detection by indEXing):
Multiplex KRT14 with 40+ other antibodies
Preserves spatial context
Reveals complex cellular neighborhoods
Ideal for tumor microenvironment studies
3. Live-cell imaging approaches:
Antibody fragments for live imaging:
Use fluorescently labeled Fab fragments of KRT14 antibodies
Track dynamics in living cells
Minimal interference with protein function
Enables study of KRT14 dynamics during migration
4. In situ sequencing with protein detection:
MERFISH with antibody detection:
Combine in situ RNA detection with KRT14 antibody staining
Visualize both transcripts and protein in the same cells
Assess correlation between mRNA and protein levels
Reveals post-transcriptional regulation mechanisms
Practical considerations for single-cell applications:
Antibody validation is critical for single-cell applications
Titrate antibodies to minimize background
Include appropriate controls (isotype, FMO)
Consider clone-specific performance in different applications
Validate staining on known positive and negative populations
1. Validation of knockout/knockdown efficiency:
Protein level validation:
Western blotting with at least two different KRT14 antibodies targeting different epitopes
Immunofluorescence to assess cellular distribution and expression level
Flow cytometry for quantitative assessment of population-level changes
Transcript level validation:
qRT-PCR with primers spanning different exons
RNA-seq to confirm complete absence of KRT14 transcripts
Assessment of potential compensatory changes in other keratins
2. Essential experimental controls:
Genetic controls:
Technical controls:
Multiple independent knockout/knockdown clones to avoid clone-specific effects
Mixed population analysis to assess heterogeneous responses
Time-course analysis to distinguish acute vs. adaptative responses
3. Phenotypic assessment controls:
Functional assays:
Molecular profiling:
Assessment of other keratin expression (potential compensation)
Analysis of keratin-binding proteins
Evaluation of cell-type-specific markers to confirm cell identity
4. Specialized controls for specific applications:
For in vivo studies:
Conditional knockout models to avoid developmental effects
Tissue-specific promoters to target specific cell populations
Littermate controls with matched genetic background
Assessment of immune cell infiltration as potential confounder
For therapeutic targeting:
Dose-response studies to establish specificity
Off-target effect assessment
Rescue experiments with modified KRT14 constructs
Comparison with other cytoskeletal disruption approaches
Example from research: In a study of KRT14's role in ovarian cancer invasion, researchers generated both KRT14 knockout and overexpression cell lines, comparing them to wild-type and non-targeting CRISPR control cells. The phenotypic differences were assessed through spheroid formation assays, which revealed that KRT14 knockout cells showed impaired spheroid formation initially but formed comparable spheroids after extended incubation. Conversely, KRT14 overexpression lines formed dense spheroids rapidly with visible outgrowth .