KRT14/KRT16/KRT5/KRT6A/KRT8 Monoclonal Antibody

Shipped with Ice Packs
In Stock

Description

Immunohistochemistry (IHC)

  • Identifies basal (KRT5/KRT14-positive) vs. differentiated (KRT1/KRT10-positive) epithelial cells .

  • Detects aberrant keratin expression in tumors, aiding cancer subtyping .

ELISA

  • Quantifies cytokeratin levels in serum or tissue lysates for disease biomarker studies .

Disease Mechanisms

  • Psoriasis: KRT6A/KRT16 overexpression correlates with keratinocyte hyperproliferation and immune activation .

  • Epidermolysis Bullosa: KRT5/KRT14 mutations disrupt epidermal-dermal adhesion, causing blistering .

Research Findings

  1. Co-Expression in Disease:

    • In hidradenitis suppurativa (HS), KRT6A and KRT17 show 5–10× higher expression compared to healthy skin .

    • Psoriatic keratinocytes exhibit strong KRT16-KRT6A pairing (r2=0.47r^2 = 0.47), driving abnormal migration .

  2. Diagnostic Utility:

    • This antibody distinguishes basal-like breast cancers (KRT5+/KRT14+) from luminal subtypes (KRT8+/KRT18+) .

  3. Functional Insights:

    • KRT14-KRT5 maintains epidermal proliferation via AKT phosphorylation, while KRT1-KRT10 promotes differentiation via ERK1/2 suppression .

Significance in Epithelial Biology

The KRT14/KRT16/KRT5/KRT6A/KRT8 monoclonal antibody is pivotal for unraveling epithelial plasticity in health and disease. Its ability to simultaneously target keratins across differentiation states makes it indispensable for studying wound healing, cancer progression, and genetic skin disorders .

Product Specs

Buffer
The antibody is provided as a liquid solution in phosphate-buffered saline (PBS) containing 50% glycerol, 0.5% bovine serum albumin (BSA), and 0.02% sodium azide as a preservative.
Form
Liquid
Lead Time
Our standard lead time for dispatching this antibody is 1-3 business days after receiving your order. Delivery timelines may vary depending on the chosen shipping method and destination. We recommend contacting your local distributor for specific delivery estimates.

Q&A

What are keratin proteins and what is their significance in cancer research?

Keratins (KRTs) are intermediate filament-forming proteins primarily expressed in epithelial cells. They form an integral part of the cytoskeleton and provide structural integrity to cells. In cancer research, KRTs serve as extensively used diagnostic biomarkers and are associated with tumorigenesis and metastasis across multiple cancer types. KRTs play significant roles in various cellular processes including epidermis development, intermediate filament cytoskeleton organization, keratinization, and keratinocyte migration. Dysregulation of KRT expression patterns has been implicated in cancer progression, making them valuable research targets . KRT expression analysis enables researchers to differentiate between normal and malignant tissues, as well as between different cancer stages, contributing to both diagnostic and prognostic assessments in clinical oncology.

How do KRT5, KRT6, KRT8, KRT14, and KRT16 differ functionally in normal and cancer tissues?

These keratin proteins belong to different subgroups with distinct expression patterns and functions:

  • KRT5 and KRT14: Typically expressed as pairs in basal epithelial cells, providing structural support. In cancer, their aberrant expression often indicates basal-like carcinomas.

  • KRT6 (including isoforms KRT6A, KRT6B, and KRT6C): Normally found in specialized stratified epithelia and activated in wound healing. KRT6A shows significantly higher concentration in head and neck squamous cell carcinoma (HNSCC) tumor samples compared to surgical margins, while KRT6B and KRT6C demonstrate inverse relationships .

  • KRT8: Primarily expressed in simple epithelial cells. Often upregulated in adenocarcinomas, with unique expression patterns showing higher levels in metastatic melanoma compared to primary melanoma .

  • KRT16: Associated with hyperproliferative conditions and wound healing. Its expression is significantly altered between primary and metastatic melanoma, correlating with cancer progression .

These differential expression patterns serve as valuable markers for distinguishing various cancer types and stages.

What molecular pathways interact with KRT proteins in cancer progression?

KRT proteins interact with several critical cancer-related pathways. Gene Set Enrichment Analysis (GSEA) has revealed that KRT1, KRT2, KRT5, KRT6A, KRT6B, KRT6C, KRT10, KRT14, KRT15, KRT16, and KRT17 are most significantly involved in the p53 pathway, KRAS signaling, and estrogen response pathways (both early and late) . For KRT8, the most involved hallmark pathways include inflammatory response alongside KRAS signaling and estrogen responses . The role of KRT6A has been particularly noted in non-small-cell lung cancer, where its overexpression affects the upregulation of G6PD (glucose-6-phosphate dehydrogenase), subsequently activating metabolic pathways that promote cancer cell invasion and growth . These molecular interactions highlight the complex role of KRT proteins beyond their structural functions, positioning them as both biomarkers and potential therapeutic targets.

What are the optimal methods for detecting and quantifying KRT protein expression in clinical samples?

For accurate detection and quantification of KRT proteins in clinical samples, researchers should consider multiple complementary approaches:

  • Enzyme-linked immunosorbent assay (ELISA): Provides precise quantitative measurement of protein concentrations, as demonstrated in studies measuring KRT6A, KRT6B, and KRT6C in tumor and margin samples . ELISA is particularly valuable when distinguishing between closely related isoforms.

  • Immunohistochemistry (IHC): Allows visualization of spatial distribution of KRT proteins within tissue sections, providing insights into heterogeneity of expression.

  • Western blotting: Useful for semi-quantitative analysis and verification of antibody specificity.

  • RT-qPCR: For transcriptional analysis, as utilized in studies examining KRT mRNA expression across cancer databases including TCGA, GEO, and ONCOMINE .

Each method has distinct advantages, and combining multiple techniques provides the most comprehensive analysis. For example, ELISA measurements from the study on HNSCC revealed significantly higher KRT6A protein concentration in tumor samples compared to margins (0.01976 (0.00962–0.02825) vs. 0.00933 (0.00488–0.01938); p = 0.0003), while KRT6B and KRT6C showed inverse patterns .

How should researchers optimize protocols for simultaneous detection of multiple KRT proteins?

Optimizing protocols for simultaneous detection of multiple KRT proteins requires careful consideration of several factors:

  • Antibody selection: Choose antibodies with minimal cross-reactivity between KRT isoforms. Monoclonal antibodies offer higher specificity compared to polyclonal alternatives, which is crucial when distinguishing between closely related isoforms like KRT6A, KRT6B, and KRT6C.

  • Multiplexing strategies: For immunofluorescence or flow cytometry, select fluorophores with minimal spectral overlap. For chromogenic IHC, sequential staining protocols may be necessary.

  • Validation controls: Include single-stained controls and isotype controls to verify specificity and rule out cross-reactivity.

  • Sample preparation: Standardize fixation and antigen retrieval methods, as different KRT epitopes may have varying sensitivity to fixation.

  • Quantification methods: Implement digital pathology tools for objective quantification of multiple markers.

When analyzing correlations between different KRT proteins, as seen in HNSCC studies where KRT6C protein from margin samples significantly correlated with all three KRT6 proteins in tumor samples, proper multiplexing optimization ensures reliable results .

What are the critical considerations for sample collection and processing to preserve KRT protein integrity?

Preserving KRT protein integrity during sample collection and processing requires strict adherence to several key principles:

  • Rapid fixation: Process tissue samples immediately after collection or snap-freeze in liquid nitrogen to prevent protein degradation. Delays beyond 30 minutes can lead to significant deterioration of epitopes.

  • Standardized fixation protocols: For formalin-fixed paraffin-embedded (FFPE) samples, consistent fixation times (typically 24-48 hours) in 10% neutral-buffered formalin are essential for reproducible results.

  • Controlled pH: Maintain pH between 7.2-7.4 during processing to preserve protein conformation and epitope accessibility.

  • Optimized antigen retrieval: Different KRT proteins may require specific antigen retrieval methods (heat-induced vs. enzymatic).

  • Storage conditions: Store frozen samples at -80°C and FFPE blocks at room temperature in low-humidity environments.

  • Consistency between comparative samples: When comparing tumor and margin samples, as in the HNSCC study measuring KRT6 protein concentrations, identical processing protocols must be applied to all samples to avoid technical artifacts .

These considerations are critical for generating reliable and reproducible data, particularly in studies examining subtle differences in KRT isoform expression.

How should researchers interpret differential expression patterns of KRT proteins between tumor and non-tumor tissues?

Interpretation of differential KRT expression patterns requires contextual understanding of tissue-specific expression and cancer biology:

  • Direction of change: Determine whether expression is increased or decreased compared to control tissues. For example, KRT6A shows significantly higher concentration in HNSCC tumor samples compared to surgical margins, while KRT6B and KRT6C show inverse relationships .

  • Isoform-specific patterns: Analyze each KRT isoform separately rather than grouping them. In HNSCC, the contrasting patterns between KRT6A vs. KRT6B/KRT6C highlight the need for isoform-specific analysis .

  • Cancer type specificity: KRT expression patterns may be cancer-type specific. In melanoma, KRT1, KRT2, KRT5, KRT6A, KRT6B, KRT6C, KRT10, KRT14, KRT15, KRT16, and KRT17 are all highly expressed in primary melanoma compared to metastatic melanoma .

  • Fold change magnitude: Consider the magnitude of expression differences. In melanoma vs. normal tissue, dramatic differences were observed: KRT14 (fold change: -234.928), KRT15 (fold change: -219.782), and KRT5 (fold change: -71.976) .

  • Biological pathways: Connect expression changes to relevant biological processes, such as cell migration, invasion, or differentiation.

These differential expression patterns often reflect fundamental changes in cellular phenotype during tumorigenesis and progression.

What statistical approaches are most appropriate for analyzing KRT protein expression data in clinical studies?

When analyzing KRT protein expression data from clinical samples, robust statistical approaches are essential:

How can researchers correlate KRT protein expression with specific clinical parameters and outcomes?

Researchers can implement several approaches to correlate KRT protein expression with clinical parameters:

These correlations can provide valuable insights into the biological significance of KRT expression in disease progression and potentially identify novel prognostic biomarkers.

How can researchers utilize KRT expression patterns for tumor classification and prognostication?

KRT expression patterns offer significant potential for refined tumor classification and prognostication:

By systematically analyzing KRT expression in large patient cohorts and correlating with clinical outcomes, researchers can develop more accurate prognostic models that improve patient stratification for clinical management.

What are the methodological approaches for studying the functional roles of KRT proteins in cancer progression?

Several methodological approaches can elucidate the functional roles of KRT proteins in cancer:

  • Gene knockdown/knockout: siRNA, shRNA, or CRISPR-Cas9 techniques to modulate KRT expression and analyze effects on cellular phenotypes such as proliferation, migration, and invasion.

  • Overexpression studies: Transfection with KRT expression vectors to assess gain-of-function phenotypes, particularly important for understanding isoform-specific effects.

  • Cell line models: Compare KRT expression across cell lines with different invasive and metastatic potentials to identify correlations with aggressive phenotypes.

  • 3D culture systems: Examine the role of KRTs in more physiologically relevant models that better recapitulate tumor architecture and cell-cell interactions.

  • Animal models: Develop transgenic mouse models with modified KRT expression to study in vivo effects on tumor development and progression.

  • Mechanistic studies: Investigate downstream signaling pathways affected by KRT expression, such as the observed relationship between KRT6A overexpression and G6PD upregulation in lung cancer, which activates metabolic pathways promoting invasion and growth .

These approaches provide complementary insights into how KRT proteins influence cancer cell behavior beyond their structural roles.

How does HPV status influence KRT protein expression patterns in head and neck cancers?

HPV status significantly impacts KRT protein expression in head and neck squamous cell carcinomas (HNSCC):

  • Differential expression patterns: HPV-positive patients demonstrate significantly higher median protein concentrations of KRT6B in tumor samples compared to HPV-negative patients (2.43711 (0.59399–4.02520) vs. 0.36460 (0.16355–0.60257); p = 0.0335) .

  • KRT6C upregulation: Similarly, HPV-positive patients exhibit elevated median levels of KRT6C in tumor samples (0.19478 (0.18870–0.26409) vs. 0.10846 (0.07466–0.16088); p = 0.0199) .

  • p16 status correlation: p16-positive tumors (a surrogate marker for HPV) also show higher KRT6B (2.43711 (0.59399–4.0252) vs. 0.31389 (0.17382–0.62325); p = 0.0327) and KRT6C (0.19478 (0.18870–0.26409) vs. 0.10560 (0.06749–0.17158); p = 0.0451) protein levels .

  • Clinical implications: These differential expression patterns may contribute to the distinct biological behavior and improved prognosis typically observed in HPV-positive HNSCC.

  • Research considerations: When studying KRT proteins in HNSCC, stratification by HPV status is essential to avoid confounding results due to these significant biological differences.

Understanding these HPV-related differences in KRT expression may provide insights into the mechanisms underlying the different clinical behaviors of HPV-positive and HPV-negative HNSCC.

What are common challenges in KRT isoform discrimination and how can they be addressed?

Discriminating between highly homologous KRT isoforms presents significant technical challenges:

  • Cross-reactivity issues: KRT isoforms share high sequence homology, making specific antibody generation difficult. For example, KRT6A, KRT6B, and KRT6C isoforms share substantial sequence identity. Researchers should validate antibody specificity using positive and negative controls, including recombinant proteins and knockout cell lines.

  • Epitope selection: Target unique regions when developing or selecting antibodies. The C-terminal domains of KRTs often contain more variable sequences suitable for isoform-specific detection.

  • Validation strategies: Employ multiple detection methods (western blot, immunoprecipitation, mass spectrometry) to confirm isoform specificity before proceeding with complex experiments.

  • Complementary DNA analysis: When protein-level discrimination is challenging, analyze mRNA expression using isoform-specific primers as a complementary approach.

  • Blocking peptides: Use specific blocking peptides in competitive binding assays to confirm antibody specificity.

  • Quantitative controls: Include concentration gradients of recombinant KRT proteins to establish detection limits and linear ranges for quantification.

These strategies can help overcome the inherent difficulties in distinguishing between closely related KRT isoforms, essential for accurate interpretation of their differential roles in cancer.

How can researchers optimize protocols for detecting KRT proteins in different sample types?

Protocol optimization for KRT protein detection varies by sample type:

  • FFPE tissues: Optimize antigen retrieval methods (citrate vs. EDTA buffers, pH conditions, heating times) for each KRT epitope. Extended retrieval times (20-40 minutes) often improve detection of KRT proteins embedded in cross-linked matrices.

  • Frozen tissues: Adjust fixation protocols (acetone, methanol, or paraformaldehyde) based on the specific KRT antibody requirements. Test multiple fixatives to identify optimal conditions.

  • Cell lines: For immunofluorescence, optimize permeabilization conditions (0.1-0.5% Triton X-100 or 0.05-0.2% Saponin) to maximize antibody access while preserving cytoskeletal structure.

  • Lysates for immunoblotting: Select appropriate lysis buffers containing adequate detergent concentrations (1-2% SDS or 1% Triton X-100) to solubilize intermediate filament proteins effectively.

  • Flow cytometry: Develop specialized fixation and permeabilization protocols to maintain cell integrity while allowing antibody access to intracellular KRT proteins.

Empirical testing with systematic variation of key parameters (fixation time, buffer composition, antibody concentration, incubation time/temperature) is essential for each new application or sample type.

What controls and validation steps are essential when implementing new KRT antibodies in research?

Implementing new KRT antibodies requires rigorous validation:

  • Positive and negative tissue controls: Test antibodies on tissues known to express or lack the target KRT. For example, when studying KRT14, use normal epidermis as a positive control and simple columnar epithelia as a negative control.

  • Recombinant protein standards: Include purified recombinant KRT proteins of known concentration for quantitative assessments and verification of antibody specificity.

  • Knockout/knockdown validation: Validate antibody specificity using cell lines with genetic knockdown or knockout of the target KRT.

  • Western blot analysis: Confirm antibody specificity by verifying correct molecular weight binding and absence of non-specific bands.

  • Peptide competition assays: Pre-incubate antibodies with blocking peptides to confirm binding specificity.

  • Cross-reactivity testing: Test new antibodies against closely related KRT isoforms to ensure specificity, particularly important for distinguishing between KRT6A, KRT6B, and KRT6C .

  • Reproducibility assessment: Validate consistency across different lots of the same antibody and between different detection methods.

These validation steps are essential for generating reliable and reproducible results when implementing new KRT antibodies in cancer research.

How might single-cell analysis technologies advance our understanding of KRT expression heterogeneity in tumors?

Single-cell technologies offer transformative potential for understanding KRT expression patterns in cancer:

  • Intratumoral heterogeneity: Single-cell RNA sequencing (scRNA-seq) can reveal distinct subpopulations with different KRT expression profiles within the same tumor, potentially identifying therapy-resistant or metastasis-prone cell clusters.

  • Spatial context: Spatial transcriptomics and multiplexed immunofluorescence can map KRT expression patterns in relation to the tumor microenvironment, providing insights into how stromal-epithelial interactions influence KRT expression.

  • Lineage tracing: Single-cell lineage tracing combined with KRT expression analysis can help determine if KRT-expressing cells represent distinct lineages or states within tumors.

  • Transition states: scRNA-seq can capture cells in transition states (e.g., undergoing EMT), revealing dynamic changes in KRT expression patterns during cancer progression.

  • Biomarker refinement: Single-cell analysis may identify rare KRT-expressing cell populations with prognostic significance that would be missed in bulk tissue analysis.

These approaches could substantially refine our understanding of the diverse roles of KRT proteins in tumor progression and therapy resistance, potentially identifying new diagnostic and therapeutic opportunities.

What are emerging applications of KRT proteins as therapeutic targets in cancer?

While traditionally viewed primarily as biomarkers, emerging research suggests potential therapeutic applications targeting KRT proteins:

  • Metastasis inhibition: Given the associations between specific KRT expression patterns and metastatic behavior, targeted disruption of KRT-dependent invasion mechanisms could inhibit metastatic spread.

  • Signaling pathway modulation: KRTs interact with multiple signaling pathways, including p53 and KRAS pathways . Targeting these interactions could modulate downstream signaling events promoting tumor growth.

  • Metabolic reprogramming: The link between KRT6A and G6PD regulation suggests potential for metabolic intervention strategies, particularly in cancers where KRT6A overexpression drives metabolic changes supporting cancer cell growth .

  • Combination therapies: KRT expression patterns could guide selection of combination therapies targeting multiple vulnerabilities in KRT-expressing tumor cells.

  • Antibody-drug conjugates: KRT-targeting antibodies conjugated to cytotoxic payloads could deliver targeted therapy to KRT-expressing cancer cells.

These therapeutic applications remain exploratory but represent promising directions for translating KRT research from diagnostic to therapeutic applications.

How can multi-omics approaches enhance the utility of KRT expression data in precision oncology?

Integrating KRT expression data with other omics datasets can significantly enhance their utility in precision oncology:

  • Proteogenomic integration: Combining KRT protein expression with genomic alterations can reveal connections between specific mutations and KRT expression patterns, potentially identifying druggable dependencies.

  • Epigenomic correlations: Integrating KRT expression with DNA methylation and histone modification data can reveal regulatory mechanisms governing KRT expression in different cancer contexts.

  • Metabolomic associations: Correlating KRT expression with metabolomic profiles may uncover metabolic vulnerabilities in tumors with specific KRT expression patterns, as suggested by the KRT6A-G6PD relationship .

  • Immune microenvironment interaction: Analyzing relationships between KRT expression and immune cell infiltration patterns could identify immunotherapy-responsive subsets among KRT-expressing tumors.

  • Clinical data integration: Correlating KRT expression with treatment responses can help develop predictive biomarkers for therapy selection, particularly important given the associations between KRT expression and clinical parameters like nodal status .

By implementing these multi-omics approaches, researchers can develop more comprehensive models of how KRT expression influences tumor biology and treatment response, advancing precision medicine approaches for cancer patients.

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.