KRT15 Antibody

Shipped with Ice Packs
In Stock

Description

Molecular and Functional Characteristics of KRT15

KRT15 is a 49–52 kDa protein critical for maintaining epithelial integrity and stem cell populations, particularly in stratified epithelia like the skin and breast . It forms intermediate filaments with type II keratins, contributing to cellular structure and mechanical resilience. Key roles include:

  • Stem cell maintenance: KRT15 is a marker for hair follicle bulge stem cells and basal keratinocytes .

  • Tumor suppression: Downregulation correlates with aggressive cancer phenotypes, including breast invasive carcinoma (BRCA) .

Cancer Prognosis and Mechanisms

KRT15 expression is inversely linked to tumor aggressiveness. Key studies demonstrate:

Diagnostic Utility

  • Histopathological Discrimination: KRT15 antibodies help differentiate basal cell carcinoma from benign trichoblastoma, as the latter retains KRT15 expression .

  • Tissue Staining: Immunohistochemistry (IHC) on BRCA tissues revealed significantly lower KRT15 positivity (37.5%) compared to normal tissues (80%) .

Table 1: Association Between KRT15 Expression and Clinical Outcomes in BRCA

ParameterLow KRT15 vs. High KRT15P-value
5-Year OS68% vs. 85%< 0.001
TNM StageAdvanced (T3/T4, N2/N3) more common in low-KRT15 group0.021 (T stage)
ER/PR/HER2 StatusNegative receptor status prevalent in low-KRT15 group< 0.001

Table 2: Functional Enrichment of KRT15-Associated Genes

CategoryEnriched Pathways/Processes
Biological ProcessesCornification, antimicrobial response, keratinization
Molecular FunctionsReceptor ligand activity, ion channel regulation
Cellular ComponentsTransmembrane transporter complex, cell surface

Validation and Limitations

  • Technical Validation: Antibodies like ab268078 (Abcam) show high specificity in IHC and flow cytometry, with validation in HeLa and A431 cells .

  • Limitations: Cross-reactivity with non-human species is variable, and staining protocols require antigen retrieval (e.g., citrate buffer heating) .

Future Directions

  • Therapeutic Targeting: KRT15’s role in immune modulation highlights its potential as a biomarker for immunotherapy response.

  • Mechanistic Studies: Further research is needed to clarify KRT15’s interplay with tumor-suppressive pathways like Wnt/β-catenin .

Product Specs

Buffer
The antibody is provided in PBS with 0.1% Sodium Azide, 50% Glycerol, pH 7.3. Store at -20°C. Avoid repeated freeze-thaw cycles.
Lead Time
We typically ship products within 1-3 business days of receiving your order. Delivery times may vary depending on your location and the chosen shipping method. For specific delivery times, please consult your local distributor.
Synonyms
AI528832 antibody; CK 15 antibody; CK-15 antibody; CK15 antibody; Cytokeratin-15 antibody; Cytokeratin15 antibody; K15 antibody; K1C15_HUMAN antibody; K1CO antibody; Ka15 antibody; Keratin 15 antibody; Keratin 15 basic antibody; Keratin 15 beta antibody; Keratin antibody; Keratin complex 1 acidic gene 15 antibody; Keratin type I cytoskeletal 15 antibody; Keratin-15 antibody; Keratin15 antibody; KRT 15 antibody; Krt1-15 antibody; KRT15 antibody; KRTB antibody; KRTL15 antibody; Type I cytoskeletal 15 antibody; Type I keratin Ka15 antibody
Target Names
Uniprot No.

Q&A

What is Keratin 15 and why is it important in research?

Keratin 15 (KRT15) is a type I keratin protein with 456 amino acids and a molecular weight of 49.2 kDa. It functions as a structural constituent of the cytoskeleton and participates in scaffold protein binding . KRT15 is uniquely expressed in basal keratinocytes of stratified epithelia and, notably, does not appear to have a natural type II keratin expression partner, distinguishing it from most other keratins . Its importance in research stems from its role as a specific marker of stem cells in the hair-follicle bulge, making it valuable for studying epithelial stem cell biology, tissue regeneration, and certain skin disorders . Additionally, KRT15 is downregulated in activated keratinocytes, providing a useful indicator of cellular activation states in experimental systems .

How do I choose between monoclonal and polyclonal KRT15 antibodies for my research?

The choice between monoclonal and polyclonal KRT15 antibodies should be based on your specific experimental requirements:

Monoclonal KRT15 antibodies (e.g., mouse monoclonal KRT15-2554, KRT15-2958, or rabbit monoclonal KRT15-2103R) offer:

  • High specificity for a single epitope

  • Consistent lot-to-lot reproducibility

  • Lower background in applications like IHC and flow cytometry

  • Ideal for quantitative analyses or when detecting specific KRT15 domains

Polyclonal KRT15 antibodies provide:

  • Recognition of multiple epitopes on the KRT15 protein

  • Often higher sensitivity for low-abundance targets

  • Better tolerance to protein denaturation (advantageous for Western blot)

  • Useful when protein conformation may be altered

For critical diagnostic applications such as distinguishing between basal cell carcinoma and trichoepithelioma, well-characterized monoclonal antibodies with validated specificity are generally recommended to ensure consistent results across experiments .

What applications are KRT15 antibodies validated for in research settings?

KRT15 antibodies have been validated for multiple research applications with specific methodological considerations for each:

ApplicationValidated ConditionsKey Considerations
Immunohistochemistry (IHC)1-2 μg/mL for 30 min at RT on formalin-fixed tissuesRequires antigen retrieval (boiling)
Flow Cytometry1-2 μg/million cellsOptimal for analyzing KRT15+ cell populations
Western Blot (WB)Detects 49.2 kDa bandValidated with human, rat, and mouse samples
Immunofluorescence (IF)Works on both fixed and frozen samplesOften paired with stem cell markers
ELISAUsed in sandwich ELISA (paired format)Utilized as capture or detection antibody in paired systems
Immunocytochemistry (ICC)Effective for cultured epithelial cellsHeLa and A431 cells serve as positive controls

The versatility across multiple platforms allows for comprehensive investigation of KRT15 expression in diverse experimental contexts .

How should I optimize KRT15 antibody concentration for immunohistochemistry of skin specimens?

Optimizing KRT15 antibody concentration for immunohistochemistry requires a systematic approach:

  • Initial titration: Begin with the manufacturer's recommended range (typically 1-2 μg/mL for KRT15 antibodies) and test 3-4 concentrations in a dilution series .

  • Tissue preparation considerations:

    • Formalin-fixed tissues require appropriate antigen retrieval

    • Boiling is often necessary for optimal KRT15 epitope exposure

    • Include positive control tissues (skin, colon, or prostate)

  • Incubation parameters:

    • Standard protocol: 30 minutes at room temperature

    • For weaker signals: Consider extending to overnight at 4°C with lower antibody concentration

    • Secondary antibody selection should match the primary antibody host species (typically mouse or rabbit for KRT15)

  • Signal-to-noise evaluation: Assess specific staining of basal keratinocytes versus background at each concentration. The optimal concentration provides strong specific signal in the basal layer of stratified epithelia with minimal non-specific background .

  • Validation with controls:

    • Positive controls: HeLa or A431 cell lines, normal skin tissue

    • Negative controls: Primary antibody omission and tissues known to lack KRT15 expression

The optimized protocol should yield clear visualization of KRT15 in basal keratinocytes and hair follicle bulge regions with minimal background staining in other tissue components .

What are the critical factors for successful KRT15 detection in flow cytometry experiments?

Successful KRT15 detection in flow cytometry requires attention to several critical factors:

  • Sample preparation protocol:

    • PFA fixation (typically 2-4%) preserves cellular architecture

    • Gentle permeabilization is necessary as KRT15 is an intracellular target

    • Use of detergent concentration that maintains epitope integrity while allowing antibody access

  • Antibody concentration optimization:

    • Start with 1-2 μg per million cells as recommended

    • Include appropriate isotype controls to establish gating parameters

    • Consider using bright fluorophores like CF®488A for optimal signal separation

  • Gating strategy development:

    • Use forward/side scatter to eliminate debris and select intact cells

    • Apply dead cell exclusion dye to remove non-viable cells

    • Design hierarchical gating to identify KRT15+ populations within relevant cell subsets

  • Controls and validation:

    • HeLa cells serve as positive controls for human KRT15 expression

    • Include fluorescence minus one (FMO) controls to set accurate gates

    • Run parallel samples with isotype-matched control antibodies at equivalent concentrations

  • Multicolor panel considerations:

    • When including KRT15 in multicolor panels, select fluorophores with minimal spectral overlap

    • Consider the relative abundance of KRT15 versus other markers when selecting brightness of conjugates

    • Perform compensation using single-stained controls for each fluorophore

Flow cytometric analysis using anti-KRT15 antibodies has been successfully demonstrated in PFA-fixed human cell lines with detection through secondary antibody systems (e.g., goat anti-Mouse IgG-CF488) .

How do I select appropriate positive and negative controls for KRT15 antibody validation?

Rigorous validation of KRT15 antibodies requires thoughtfully selected controls:

Positive controls for KRT15 antibody validation:

  • Cell lines:

    • HeLa and A431 cells: Established expression of KRT15

    • Include as positive controls for Western blot, ICC, and flow cytometry

  • Tissue samples:

    • Normal skin: Focus on basal layer of epidermis and hair follicle bulge regions

    • Prostate tissue: Shows specific KRT15 staining patterns

    • Colon tissue: Contains KRT15-positive epithelial components

  • Recombinant protein:

    • Full-length human KRT15 recombinant protein: Particularly useful for Western blot or ELISA validation

    • Matches the immunogen used in many commercial antibodies

Negative controls for KRT15 antibody validation:

  • Methodological controls:

    • Isotype-matched irrelevant antibodies: Same host species and isotype (e.g., Mouse IgG2b for many KRT15 clones)

    • Primary antibody omission: Verifies secondary antibody specificity

    • Absorption controls: Pre-incubation with immunizing peptide should abolish specific staining

  • Biological controls:

    • Tissues known to lack KRT15 expression

    • Cell lines with confirmed absence of KRT15

    • Regions within positive tissues that should be negative (e.g., suprabasal layers of epidermis)

  • Genetic controls (for advanced validation):

    • KRT15 knockout or knockdown systems

    • Comparison of expression levels across tissues with confirmed differential expression

The integration of these controls allows for comprehensive validation of antibody specificity and establishes confidence in experimental results across different applications .

What are common pitfalls when using KRT15 antibodies in immunohistochemistry and how can they be resolved?

Several common pitfalls can affect KRT15 immunohistochemistry results:

  • False negative staining:

    • Problem: Complete absence of expected KRT15 signal in basal epithelial layers

    • Potential causes: Inadequate antigen retrieval, over-fixation, inappropriate primary antibody dilution

    • Solution: Optimize antigen retrieval through boiling and testing multiple retrieval buffers; ensure tissue fixation time is standardized (excessive formalin fixation can mask epitopes); titrate antibody concentration (1-2 μg/mL is typically optimal)

  • High background staining:

    • Problem: Non-specific signal throughout tissue sections

    • Potential causes: Insufficient blocking, excessive antibody concentration, endogenous peroxidase activity

    • Solution: Increase blocking time and concentration (5-10% normal serum from secondary antibody host species); reduce primary antibody concentration; implement more stringent washing; include hydrogen peroxide treatment for peroxidase-based detection systems

  • Inconsistent staining across tissue sections:

    • Problem: Variable KRT15 signal intensity between sections

    • Potential causes: Uneven fixation, inconsistent antigen retrieval, section thickness variation

    • Solution: Standardize tissue processing protocols; ensure uniform heating during antigen retrieval; maintain consistent section thickness (typically 4-5 μm); process all comparative samples in the same experimental batch

  • Cross-reactivity with other keratins:

    • Problem: Unexpected staining patterns suggesting detection of non-KRT15 proteins

    • Potential causes: Antibody cross-reactivity, particularly with other type I keratins

    • Solution: Select monoclonal antibodies with validated specificity (clones KRT15-2554, KRT15-2958, or KRT15-2103R have demonstrated specificity); confirm staining patterns align with known KRT15 distribution; consider parallel detection with alternative KRT15 antibody clones

  • Poor signal in paraffin-embedded tissues:

    • Problem: Weak or absent signal despite proper controls

    • Potential causes: Epitope masking during processing, paraffin interference

    • Solution: Implement enhanced antigen retrieval protocols (extended boiling time in citrate buffer pH 6.0); consider using alternative detection systems with signal amplification; evaluate frozen sections in parallel to confirm target accessibility

Implementing these targeted solutions within a systematic troubleshooting approach will significantly improve the reliability and interpretability of KRT15 immunohistochemistry results .

How do I interpret discrepancies in KRT15 detection between different application methods?

Discrepancies in KRT15 detection across different applications (e.g., IHC, WB, flow cytometry) should be systematically analyzed:

  • Epitope accessibility differences:

    • Nature of discrepancy: Positive by IHC but negative by Western blot (or vice versa)

    • Analysis approach: Different applications expose different protein epitopes; IHC primarily detects native conformations while WB detects denatured proteins

    • Resolution strategy: Select antibodies validated for multiple applications or use application-specific antibodies; consider clone KRT15-2958 which is validated for both WB and IHC

  • Sensitivity threshold variations:

    • Nature of discrepancy: Detection in one method but not another despite similar samples

    • Analysis approach: Compare detection limits across methods; flow cytometry can detect lower expression levels in individual cells compared to whole-tissue WB

    • Resolution strategy: For low-abundance detection, optimize more sensitive methods (e.g., enhanced chemiluminescence for WB, signal amplification for IHC); concentrate protein samples for WB when expression is low

  • Reagent compatibility issues:

    • Nature of discrepancy: Antibody performs well in one buffer system but poorly in another

    • Analysis approach: Evaluate buffer compatibility; some KRT15 antibodies are provided BSA-free and azide-free for specific applications

    • Resolution strategy: Select antibody formulations appropriate for intended applications; for conjugation chemistry, use antibodies specifically designated as conjugation-ready (e.g., those in PBS only buffer)

  • Fixation and processing effects:

    • Nature of discrepancy: Different results between fresh-frozen versus formalin-fixed samples

    • Analysis approach: Determine if epitope is fixation-sensitive; some KRT15 epitopes are altered by chemical crosslinking

    • Resolution strategy: For fixation-sensitive epitopes, consider clones specifically validated for FFPE tissues; use recombinant rabbit monoclonal antibodies (e.g., KRT15-2103R) which often show better performance in fixed tissues

  • Interpretive framework for resolving discrepancies:

Application ComparisonCommon DiscrepancyAnalytical Approach
IHC vs. WBPositive IHC, negative WBConfirm molecular weight (49.2 kDa); optimize protein extraction from relevant cell types
Flow vs. IHCPositive flow, negative IHCEvaluate permeabilization conditions; compare single-cell vs. tissue-level detection sensitivity
WB vs. ELISAPositive WB, negative ELISAConsider epitope masking in sandwich format; test alternative antibody pairs

Understanding these application-specific considerations helps establish a coherent interpretation framework when facing seemingly contradictory results across methods .

How can KRT15 antibodies be utilized to identify and isolate epithelial stem cell populations?

KRT15 antibodies offer powerful tools for epithelial stem cell identification and isolation through several methodological approaches:

  • Multiparameter flow cytometry for stem cell isolation:

    • Implement a KRT15 antibody (1-2 μg/million cells) in conjunction with established stem cell markers (CD34, CD200, Lgr5)

    • Develop a sequential gating strategy first selecting viable epithelial cells, then identifying KRT15+ subpopulations

    • For intracellular KRT15 staining, optimize fixation (2-4% PFA) and permeabilization conditions that preserve stem cell surface markers

    • Consider using bright fluorophores like CF®488A for KRT15 detection to enable clear separation of positive populations

  • Lineage tracing and fate-mapping studies:

    • Use KRT15 immunostaining to validate genetic lineage tracing models (e.g., KRT15-CrePR systems)

    • Apply serial immunohistochemistry of KRT15 alongside differentiation markers to monitor stem cell activation and differentiation

    • Implement co-localization studies with KRT15 antibodies and other hair follicle bulge markers to confirm stem cell identity

    • Correlate KRT15 expression patterns with functional stem cell assays such as colony-forming efficiency tests

  • Laser capture microdissection of KRT15+ stem cells:

    • Apply immunohistochemistry protocols optimized for rapid staining (30 min at RT) to maintain RNA/DNA integrity

    • Identify and isolate KRT15+ regions from hair follicle bulge or basal epidermal layers

    • Perform subsequent molecular analysis (transcriptomics, proteomics) of isolated KRT15+ populations

    • Compare molecular signatures between KRT15+ and KRT15- epithelial populations to identify novel stem cell regulators

  • 3D organoid culture systems:

    • Use FACS-sorted KRT15+ cells as founding populations for epithelial organoids

    • Apply immunofluorescence with KRT15 antibodies to monitor stem cell maintenance in long-term cultures

    • Develop time-course analyses of KRT15 expression during organoid development and manipulation

    • Correlate KRT15 expression with functional organoid characteristics such as self-renewal capacity and differentiation potential

These methodological approaches enable researchers to leverage KRT15 as a specific marker for identifying, isolating, and characterizing epithelial stem cell populations across diverse experimental systems .

What are the methodological considerations when using KRT15 antibodies for differential diagnosis between basal cell carcinoma and trichoepithelioma?

Implementing KRT15 antibodies for differential diagnosis between basal cell carcinoma (BCC) and trichoepithelioma requires careful methodological considerations:

  • Standardized immunohistochemistry protocol development:

    • Optimize antibody concentration (1-2 μg/mL) and incubation conditions (30 min at RT) specifically for dermatopathology specimens

    • Implement consistent antigen retrieval through boiling in appropriate buffer systems

    • Establish standardized counterstaining procedures to facilitate pattern recognition

    • Develop validated positive controls using normal skin samples with known KRT15 expression patterns

  • Staining pattern interpretation methodology:

    • BCC typically shows reduced or absent KRT15 staining compared to trichoepithelioma

    • Evaluate both the percentage of positive cells and staining intensity

    • Assess staining patterns at tumor-stroma interface versus central tumor regions

    • Implement semi-quantitative scoring systems to standardize interpretation:

ScoreKRT15 Staining PatternTypical Association
0Absent or <5% positive cellsFavors BCC diagnosis
1Patchy/focal (5-25% positive)Indeterminate
2Moderate (26-50% positive)Suggestive of trichoepithelioma
3Diffuse (>50% positive)Strongly favors trichoepithelioma
  • Complementary marker integration:

    • Combine KRT15 staining with cytokeratin 19 assessment on sequential sections

    • Incorporate immunostaining for CD10, bcl-2, and berEP4 in a comprehensive panel

    • Develop a weighted diagnostic algorithm incorporating multiple marker results

    • Establish concordance rates between KRT15 patterns and final diagnosis through retrospective validation studies

  • Complex case analysis approach:

    • In morphologically ambiguous cases, implement double immunofluorescence with KRT15 and proliferation markers

    • Correlate KRT15 expression with histomorphological features (peripheral palisading, clefting)

    • Consider laser microdissection of KRT15+ and KRT15- regions for molecular analysis

    • Develop consultation protocols for challenging cases with discordant KRT15 expression patterns

  • Quality assurance implementation:

    • Establish inter-observer concordance rates for KRT15 pattern interpretation

    • Implement regular proficiency testing using validated case sets

    • Develop laboratory-specific validation of diagnostic sensitivity and specificity

    • Maintain documentation of antibody lot testing and protocol optimization

These methodological approaches enhance the diagnostic utility of KRT15 immunohistochemistry in distinguishing between trichoepithelioma and basal cell carcinoma in dermatopathology practice .

What advanced conjugation and multiplexing strategies can be employed with KRT15 antibodies for spatial tissue analysis?

Advanced conjugation and multiplexing strategies with KRT15 antibodies enable sophisticated spatial tissue analysis:

  • Optimal conjugation methodologies for KRT15 antibodies:

    • Select conjugation-ready formats (BSA-free, azide-free) in PBS buffer at 1 mg/mL concentration

    • For fluorescent applications, consider brightness hierarchy: CF®488A and CF®568 provide superior signal-to-noise for KRT15 detection

    • For mass cytometry applications, utilize metal-conjugated KRT15 antibodies with rare earth elements

    • Implement site-specific conjugation methods to preserve antigen-binding capacity:

Conjugation MethodOptimal ApplicationTechnical Considerations
Direct fluorophore labelingMulticolor IF, flow cytometryUse CF® dyes for superior brightness and photostability
Biotin conjugationSignal amplification workflowsEnables multilayer detection with enhanced sensitivity
Metal isotope labelingMass cytometry (CyTOF)Allows high-parameter analysis without spectral overlap
Oligonucleotide taggingCODEX, CITE-seqFacilitates combined protein-transcriptome analysis
Enzyme conjugationChromogenic IHCProvides permanent staining for long-term archiving
  • Multiplexing strategy development:

    • Implement cyclic immunofluorescence with KRT15 as an anchor marker for epithelial territories

    • Develop tyramide signal amplification protocols for low-abundance targets in combination with KRT15

    • Utilize spectral unmixing algorithms to separate overlapping fluorophores in dense multiplexing panels

    • Implement sequential staining with gentle antibody elution preserving tissue architecture

  • Spatial analysis methodological approaches:

    • Apply computational tissue segmentation algorithms using KRT15 to define epithelial compartments

    • Develop nearest-neighbor analysis to quantify spatial relationships between KRT15+ cells and other cell types

    • Implement grid-based quantification of KRT15 expression gradients across tissue regions

    • Correlate KRT15 expression patterns with extracellular matrix components through simultaneous visualization

  • Technical optimization for multiparameter imaging:

    • Balance fluorophore selection to prevent bleed-through when KRT15 signal is strong

    • Optimize antibody concentration (typically lower than standard IHC) to maintain signal separation

    • Implement appropriate negative controls for each parameter in the multiplexed panel

    • Develop batch-correction algorithms for comparing KRT15 patterns across multiple specimens

These advanced strategies enable researchers to position KRT15 expression within complex tissue architectures and correlate its distribution with multiple additional parameters for comprehensive spatial tissue analysis .

How might emerging antibody technologies enhance KRT15-based research applications?

Emerging antibody technologies offer significant potential to advance KRT15-based research through several methodological innovations:

  • Recombinant antibody engineering for enhanced KRT15 detection:

    • Development of high-affinity recombinant KRT15 antibody fragments (Fab, scFv)

    • Engineering of bispecific antibodies targeting KRT15 and complementary epithelial markers

    • Creation of humanized antibody variants for reduced background in human tissue analysis

    • Implementation of site-specific conjugation strategies to improve signal-to-noise ratio

  • Single-cell analytical approaches with KRT15 antibodies:

    • Integration of KRT15 detection in spatial transcriptomics workflows

    • Development of CITE-seq compatible KRT15 antibodies for simultaneous protein and RNA analysis

    • Implementation of KRT15 antibodies in microfluidic single-cell capture systems

    • Correlation of KRT15 protein levels with single-cell RNA expression profiles

  • Live-cell imaging applications:

    • Development of non-disruptive KRT15 labeling strategies for live epithelial cells

    • Engineering of minimally invasive nanobody or aptamer-based KRT15 detection systems

    • Implementation of split-GFP complementation strategies for KRT15 visualization

    • Creation of KRT15-targeted fluorogenic probe systems for dynamic imaging

  • Computational integration methodologies:

    • Development of machine learning algorithms for automated KRT15 expression pattern analysis

    • Implementation of cloud-based image analysis platforms for standardized KRT15 quantification

    • Creation of publicly accessible KRT15 expression atlases across normal and pathological tissues

    • Development of integrated multi-omics approaches correlating KRT15 protein with genomic and metabolomic data

These emerging technologies promise to enhance the specificity, sensitivity, and analytical depth of KRT15-based research applications, enabling more sophisticated investigations of epithelial biology and pathology .

What methodological approaches can resolve contradictory findings about KRT15 expression in cancer research?

Resolving contradictory findings regarding KRT15 expression in cancer research requires systematic methodological approaches:

  • Standardized detection and quantification protocols:

    • Implement consensus antibody validation guidelines for KRT15 detection in cancer tissues

    • Develop quantitative threshold standards for defining "positive" versus "negative" KRT15 expression

    • Establish multicenter ring studies using identical protocols and antibody clones

    • Create standardized reporting formats for KRT15 expression pattern description:

ParameterStandardized AssessmentTechnical Implementation
Expression extentPercentage of positive cells (0-100%)Whole slide digital quantification
Expression intensityScaled score (0, 1+, 2+, 3+)Calibrated against reference standards
Expression patternFocal, diffuse, peripheral, gradientSpatial pattern recognition algorithms
Heterogeneity indexCoefficient of variation across fieldsMultiple region sampling approach
  • Multi-level assessment approach:

    • Correlate protein-level findings (IHC) with mRNA expression (ISH or qPCR)

    • Implement laser capture microdissection to resolve tumor heterogeneity

    • Develop single-cell approaches to disentangle mixed cell populations

    • Correlate KRT15 expression with clonal evolution markers in longitudinal samples

  • Biological context integration:

    • Stratify analyses based on tumor stage, grade, and molecular subtype

    • Correlate KRT15 expression with specific genetic alterations (p53 status, EGFR mutations)

    • Analyze KRT15 expression in the context of epithelial-mesenchymal transition markers

    • Implement multiparameter analysis correlating KRT15 with stem cell and differentiation markers

  • Methodological triangulation:

    • Compare findings across multiple antibody clones targeting different KRT15 epitopes

    • Validate results across different detection platforms (chromogenic IHC, IF, flow cytometry)

    • Implement orthogonal functional assays to correlate expression with biological behavior

    • Develop transgenic reporter systems for prospective KRT15 lineage studies in preclinical models

These systematic approaches can help resolve contradictions in the literature regarding KRT15 expression in cancer, leading to more consistent findings and clearer understanding of its biological significance .

Quick Inquiry

Personal Email Detected
Please use an institutional or corporate email address for inquiries. Personal email accounts ( such as Gmail, Yahoo, and Outlook) are not accepted. *
© Copyright 2025 TheBiotek. All Rights Reserved.