This monoclonal antibody specifically against human KRT5 protein was produced using hybridoma technology. First, mice were immunized with a synthesized peptide derived from human KRT5, and then B cells were isolated from their spleen. The B cells were fused with myeloma cells to form hybridomas, from which those continuously produce the KRT5 antibody was screened and selected. The KRT5 antibody-secreting hybridomas were cultured in the mouse abdominal cavity. The purified KRT5 monoclonal antibody was obtained from mouse ascites using a specific immunogen. This KRT5 monoclonal antibody has been validated in ELISA and IHC applications.
KRT5 is a type II intermediate filament protein that is primarily expressed in the basal cells of stratified epithelia, including the epidermis of the skin, hair follicles, and nails. KRT5 functions to provide structural support and integrity to these tissues by forming a cytoskeletal network of filaments. It also plays a role in cell signaling pathways and gene regulation. Mutations in the KRT5 gene have been associated with several skin disorders, including epidermolysis bullosa simplex and Dowling-Degos disease.
This monoclonal antibody, specifically targeting human KRT5 protein, was developed using hybridoma technology. Mice were initially immunized with a synthetic peptide derived from human KRT5. Subsequently, B cells were isolated from their spleens and fused with myeloma cells to form hybridomas. From these hybridomas, those continuously producing the KRT5 antibody were screened and selected. The KRT5 antibody-secreting hybridomas were then cultured in the mouse abdominal cavity. The purified KRT5 monoclonal antibody was obtained from mouse ascites using a specific immunogen. This KRT5 monoclonal antibody has been validated in ELISA and IHC applications.
KRT5 is a type II intermediate filament protein primarily expressed in the basal cells of stratified epithelia. These include the epidermis of the skin, hair follicles, and nails. KRT5 functions to provide structural support and integrity to these tissues by forming a cytoskeletal network of filaments. Additionally, it plays a role in cell signaling pathways and gene regulation. Mutations in the KRT5 gene have been associated with several skin disorders, including epidermolysis bullosa simplex and Dowling-Degos disease.
Keratin 5 (KRT5) is a type II cytoskeletal protein belonging to the subfamily of intermediate filament proteins expressed in epithelial tissues. It has a predicted molecular weight of approximately 62.3 kDa and shows remarkable biochemical diversity. KRT5 is one of at least 20 different cytokeratin polypeptides expressed in human epithelial tissues, with molecular weights ranging between 40-68 kDa and isoelectric pH between 4.9-7.8 . KRT5 is particularly significant in research because its expression patterns are characteristic of specific epithelial types and reflect the degree of maturation or differentiation within an epithelium. This makes it an invaluable marker for studying epithelial biology, tissue development, and pathological conditions including cancer and inflammatory diseases .
KRT5 monoclonal antibodies are versatile tools employed across multiple research applications:
Immunohistochemistry (IHC): Primary application for tissue section analysis and cancer classification
Immunofluorescence (IF): For co-localization studies with other markers
Western Blotting (WB): For protein expression quantification
Flow Cytometry (FACS): For cell sorting and quantitative analysis
Enzyme-Linked Immunosorbent Assay (ELISA): For protein detection in solution
Immunocytochemistry (ICC): For cellular localization studies
Different antibody clones may show varying performance across these applications. For example, clone MA5-17057 has demonstrated effectiveness in ELISA, FACS, ICC, IHC, IF, and WB applications with reactivity to human, mouse, and non-human primate samples . Clone MA5-12596 (XM26) is particularly optimized for IHC in paraffin-embedded tissues and shows specific reactivity with human samples .
Normal KRT5 expression is primarily observed in basal cells of stratified epithelia, including the epidermis and specific regions of respiratory, digestive, and urogenital tracts. In pathological conditions, aberrant expression patterns emerge that can serve as diagnostic indicators:
Normal expression: Confined to basal layer of stratified epithelia, paired with KRT14
Pathological expression:
When interpreting KRT5 staining, researchers should carefully consider both the intensity and pattern of expression, as well as co-expression with other markers to accurately distinguish between normal and pathological states. The expansion of KRT5+ cells in viral pneumonia, for instance, represents a distinct pathological response that differs from other injury models like bleomycin-induced damage .
Research comparing four major KRT5 antibody clones has revealed significant differences in analytical performance:
| Antibody Clone | Type | Analytical Sensitivity | Analytical Specificity | Cross-Reactivity Issues | Recommended Applications |
|---|---|---|---|---|---|
| SP27 | Rabbit monoclonal | Highest | Lower | Potential cross-reactivity with adenocarcinomas (25% false positives) | Research requiring high sensitivity |
| D5/16 B4 | Mouse monoclonal | Moderate | Higher | Mouse Ascites Golgi (MAG) reaction in some tissues | General purpose, careful interpretation needed |
| XM26 | Mouse monoclonal | Moderate | Higher | Minimal reported | Standard IHC, high specificity applications |
| EP1601Y | Rabbit monoclonal | Moderate | Higher | Minimal reported | General purpose with good specificity |
The SP27 clone demonstrated significantly higher analytical sensitivity than the other clones but showed a distinct positive reaction in 25% of adenocarcinomas (AC) cases where other clones showed no staining, suggesting lower specificity. Clone D5/16 B4 displayed granular staining in 14 AC cases, likely representing Mouse Ascites Golgi-reaction, a potential source of false positives . These findings highlight the importance of clone selection based on the specific research question and the need for appropriate controls.
To enhance the reliability of KRT5 detection and minimize false results, researchers should implement these methodological strategies:
Multi-method validation: Combine protein detection (IHC) with mRNA detection (in situ hybridization) for confirmation. Studies have shown that weak, scattered expression of KRT5 mRNA can be detected in 71% of adenocarcinomas, even when protein is not detected by most antibody clones .
Scoring system implementation: Utilize quantitative scoring methods like H-score (0-300) to standardize interpretation across samples and studies .
Positive and negative controls: Include known positive tissues (squamous epithelium) and negative tissues in each experiment.
Clone-specific optimization:
Optimize antigen retrieval methods for each clone
Adjust antibody concentration based on preliminary titration
Determine optimal incubation times and temperatures
Complementary markers: Use p40 immunohistochemistry alongside KRT5 for more accurate cell type identification .
Implementing these approaches minimizes technical variability and increases confidence in experimental results, particularly in challenging diagnostic scenarios like distinguishing between squamous cell carcinomas and adenocarcinomas.
The behavior of KRT5+ cells differs significantly between viral infection and other lung injury models:
In viral pneumonia, such as influenza A virus (IAV) infection, KRT5+ basal-like cells undergo rapid expansion in small airways and alveoli, forming distinctive scar-like structures. This expansion is significantly more extensive than in bleomycin-induced injury models. IAV-infected mice demonstrate substantially larger KRT5+ lung areas and higher Krt5 mRNA levels compared to bleomycin-challenged mice . Additionally, IAV-infected lungs develop goblet cell hyperplasia in both airways and alveoli, whereas bleomycin-injured lungs show virtually no goblet cells .
The expansion of KRT5+ cells in viral infection appears to be regulated by specific inflammatory signals, particularly interferon-gamma (IFN-γ). In IAV infection, IFN-γ expression peaks around day 7, coinciding with the first appearance of dysplastic KRT5+ cells in the distal airway, and remains elevated through day 10 when these cells begin forming pod-like structures . This temporal correlation suggests a mechanistic link between IFN-γ signaling and KRT5+ cell expansion.
Experimental evidence supports this relationship, as mice with epithelial-specific knockout of interferon-gamma receptor 1 (Ifngr1) show reduced KRT5+ lung area and decreased Krt5 mRNA expression following IAV infection . This reduction in KRT5+ cells is not due to changes in viral clearance efficiency, indicating a direct role for IFN-γ signaling in KRT5+ cell formation. Importantly, this IFN-γ dependence appears specific to viral injury, as bleomycin-induced KRT5+ cell formation remains unaffected in Ifngr1 knockout mice .
The JAK/STAT signaling pathway plays a crucial role in mediating IFN-γ-induced KRT5+ cell formation during viral lung injury:
In vitro evidence: Treatment of cultured intrapulmonary p63+ progenitor cells with IFN-γ promotes their transdifferentiation into KRT5+ cells. This effect is significantly diminished when JAK1/JAK2 inhibitors (baricitinib or fedratinib) are added to the culture medium .
In vivo confirmation: Administration of baricitinib to IAV-infected mice from day 7 to day 11 post-infection results in reduced KRT5+ alveolar area, confirming the pathway's importance in the living organism .
Molecular mechanism: Single-cell RNA sequencing of lung epithelial cells from IAV-infected mice reveals that dysplastic KRT5+ cells, proliferating cells, and CLDN4+ intermediate cells are enriched in genes related to the type I cytokine signaling pathway .
This signaling cascade appears to be specifically important for viral-induced KRT5+ cell formation but not for other injury models, suggesting distinct molecular mechanisms regulate epithelial responses to different types of lung injury. Researchers investigating epithelial regeneration should consider these pathway-specific effects when designing experiments and interpreting results.
While KRT5 is a valuable marker in cancer research, several challenges exist:
Antibody clone variability: Different antibody clones show varying sensitivity and specificity profiles. For example, clone SP27 demonstrates higher analytical sensitivity but may produce false positives in adenocarcinomas .
Expression heterogeneity: KRT5 expression in tumors can be heterogeneous, with weak and scattered expression patterns that are difficult to interpret. Studies have shown scattered KRT5 mRNA expression in 71% of adenocarcinomas despite negative or minimal protein detection by most antibody clones .
Protein versus mRNA discrepancies: Research has identified discrepancies between KRT5 protein detection by immunohistochemistry and mRNA detection by in situ hybridization. Some tumors with high H-scores for protein expression show low ISH scores for mRNA, and vice versa .
Non-specific reactions: Technical artifacts can complicate interpretation, such as the Mouse Ascites Golgi reaction observed with the D5/16 B4 clone .
Context-dependent significance: The diagnostic and prognostic value of KRT5 expression varies across cancer types and subtypes, requiring context-specific interpretation.
Addressing these challenges requires careful methodological approaches, including the use of multiple detection methods, standardized scoring systems, and consideration of KRT5 expression in the context of other biomarkers.
Single-cell RNA sequencing (scRNA-Seq) has emerged as a powerful tool for characterizing KRT5+ cells and their role in disease:
Recent scRNA-Seq analysis of lung epithelial cells from influenza virus-infected mice has revealed complex heterogeneity among KRT5+ cells. This approach identified multiple distinct epithelial cell clusters, including dysplastic KRT5+ cells, proliferating cells, secretory cells, ciliated cells, AT1 cells, AT2 cells, and CLDN4+ intermediate cells (also known as damage-associated transient progenitors or prealveolar type-1 transitional cell state) .
This single-cell approach provides several advantages:
Resolution of cellular heterogeneity: Distinguishes subpopulations within KRT5+ cells that may have different functions or developmental trajectories.
Identification of gene signatures: Reveals that dysplastic KRT5+ cells, proliferating cells, and CLDN4+ intermediate cells are enriched in genes related to type I cytokine signaling pathways .
Lineage relationships: Helps establish developmental relationships between different cell populations during injury and repair.
Temporal dynamics: When applied across different time points, can reveal the dynamic changes in cell populations during disease progression and resolution.
Therapeutic target identification: Identifies potential molecular targets by revealing the specific pathways active in disease-associated KRT5+ cells.
Researchers can leverage this approach to move beyond simple marker analysis to a comprehensive understanding of KRT5+ cell biology in various disease contexts.
Effective co-staining experiments with KRT5 require careful planning:
Antibody compatibility:
Choose primary antibodies raised in different host species to avoid cross-reactivity
Verify that secondary antibodies do not cross-react with inappropriate primary antibodies
Consider using directly conjugated antibodies when possible to reduce background
Technical parameters:
Optimize antigen retrieval methods that work for all target proteins
Determine the optimal order of antibody application if sequential staining is needed
Test for potential epitope masking when multiple antibodies are used
Biologically relevant co-markers:
For basal cell characterization: p63, KRT14, integrin α6/β4
For differentiation studies: KRT10, involucrin, loricrin
For cancer research: p40, TTF-1, Napsin A
For regenerative studies: proliferation markers (Ki67, PCNA)
Controls:
Single stain controls to assess specificity
Absorption controls with recombinant proteins
Isotype controls to evaluate non-specific binding
Imaging considerations:
Select fluorophores with minimal spectral overlap
Include proper compensation controls if using flow cytometry
Establish consistent exposure settings for quantitative comparisons
When properly designed, co-staining experiments can provide valuable insights into the biological context of KRT5 expression, cellular identity, and functional state.
Non-specific staining is a common challenge when working with KRT5 antibodies. The following methodological approaches can help minimize these issues:
Clone-specific considerations:
Protocol optimization:
Titrate antibody concentration to determine optimal dilution
Extend blocking steps using species-appropriate serum or protein blockers
Increase washing duration and frequency between steps
Reduce secondary antibody concentration if background is high
Sample preparation improvements:
Ensure proper fixation duration (overfixation can increase background)
Optimize antigen retrieval method and duration
Use fresh tissue samples when possible to minimize autofluorescence
Validation approaches:
Compare results with mRNA detection via in situ hybridization
Use multiple antibody clones targeting different epitopes
Include isotype control antibodies at the same concentration
Include known positive and negative tissue controls
When non-specific staining persists despite these measures, researchers should consider alternative detection methods or antibody clones with higher specificity profiles, even if sensitivity is somewhat compromised.
Accurate quantification of KRT5 expression requires standardized approaches:
Immunohistochemistry quantification:
Implement the H-score system (0-300) which incorporates both staining intensity and percentage of positive cells
Use digital image analysis software for objective assessment
Establish clear criteria for positive staining (membrane, cytoplasmic, or both)
Score multiple fields (at least 5-10) to account for heterogeneity
Western blot analysis:
Include recombinant KRT5 standards for calibration
Normalize to appropriate loading controls
Use densitometry software for quantitative analysis
Validate antibodies for specificity using knockout or knockdown controls
mRNA quantification:
For in situ hybridization, implement standardized scoring systems
With RT-qPCR, use validated reference genes for normalization
Conduct melt curve analysis to confirm specificity
Consider digital droplet PCR for absolute quantification
Flow cytometry considerations:
Establish clear gating strategies based on appropriate controls
Use median fluorescence intensity (MFI) rather than mean
Consider using standardized beads for day-to-day calibration
Include viability dye to exclude dead cells from analysis
Regardless of the method, researchers should report detailed protocols, quantification methods, and statistical approaches to enable reproducibility. Comparative studies should maintain consistent methodologies across all samples to ensure valid comparisons.