KRT17 (Keratin 17) is a type I keratin intermediate filament protein with a molecular weight of 48 KDa. It belongs to the keratin family, of which there are 54 functional types in humans . In normal physiology, KRT17 is poorly expressed in mature epithelial tissues but selectively expressed in hyperplasia-active cells such as stem cells or reserve cells in the epithelium, as well as in glandular myoepithelium . Beyond its structural cytoskeletal function, KRT17 has emerged as a regulatory protein affecting cell movement, size, proliferation, apoptosis, and signal transduction pathways .
The methodology to study KRT17's basic characteristics typically involves:
Protein characterization through Western blotting and immunoprecipitation
Subcellular localization analysis using immunofluorescence microscopy
Expression profiling across tissue types using RNA-seq and proteomics
Structural analysis through electron microscopy of intermediate filament networks
The gold standard for KRT17 detection in clinical and research settings is immunohistochemistry (IHC). A standardized protocol includes:
Parameter | Score | Criteria |
---|---|---|
Staining Intensity | 0 | Negative |
1 | Weak | |
2 | Moderate | |
3 | Strong | |
Percentage of Positive Cells | 0 | <5% |
1 | 5-25% | |
2 | 26-50% | |
3 | 51-75% | |
4 | >75% | |
Final Score | Intensity × Percentage | |
Expression Classification | <6 | Low expression |
≥6 | High expression |
For accurate assessment, researchers should examine five views per slide and 100 cells per view at 400× magnification . Alternative methods include:
RT-qPCR for mRNA quantification
Western blotting for protein level analysis
Flow cytometry for single-cell expression analysis in suspension cells
Digital spatial profiling for spatial context in tissue microenvironment
Cancer tissues: KRT17 serves as a diagnostic marker in multiple cancer types, including breast cancer, skin squamous cell carcinoma, cervical cancer, and non-small cell lung cancer (NSCLC) .
NSCLC: Overexpression of KRT17 correlates with histological type (p<0.001), lymph node metastasis (p=0.040), differentiation (p=0.011), age (p=0.024), and gender (p<0.001) .
Breast cancer: Interestingly, decreased expression of KRT17 correlates with poor prognosis in HER2-high and ER-high breast cancer patients, demonstrating context-dependent roles .
Diabetic conditions: KRT17 is upregulated under high glucose conditions in keratinocytes, promoting proliferation and migration that may contribute to hyperkeratosis in diabetic skin .
DNA damage response: KRT17 expression is induced following DNA damage .
KRT17 employs multiple mechanisms to influence cancer progression:
Wnt signaling activation: In NSCLC, KRT17 promotes proliferation and invasiveness by activating the Wnt signaling pathway .
EMT promotion: KRT17 activates the epithelial-mesenchymal transition (EMT) process, which is crucial for cancer cell invasion and metastasis .
DNA damage response modulation: A nuclear pool of KRT17 regulates the immediate response to DNA damage, with associated impact on cell survival. KRT17 is required at an early stage of the double-stranded break (DSB) repair pathway, where it associates with key DDR effectors, including γ-H2A.X, 53BP1, and DNA-PKcs .
Cytoskeletal reorganization: KRT17 alters the cytoskeletal structure, affecting cell migration and invasion capabilities .
Immune microenvironment modulation: KRT17 expression affects the infiltration of immune cells, including activated NK cells in HER2-high breast cancer and naïve CD4 T cells in ER-high breast cancer .
Research methodologies to investigate these mechanisms include:
Chromatin immunoprecipitation (ChIP) to identify DNA-binding sites
Co-immunoprecipitation to detect protein-protein interactions
Transwell and wound healing assays to assess migration and invasion
Luciferase reporter assays to measure pathway activation
The prognostic significance of KRT17 varies by cancer type and molecular context:
To reconcile these contradictions, researchers should:
Evaluate molecular context: Examine KRT17 in combination with other biomarkers (HER2, ER) that may modify its function.
Assess subcellular localization: Determine whether KRT17 is primarily cytoplasmic or nuclear, as its location significantly impacts function .
Consider signaling pathway interactions: Analyze how KRT17 interacts with tissue-specific signaling networks.
Perform multi-omics integration: Combine transcriptomics, proteomics, and epigenomics to identify co-regulated genes and pathways.
Conduct mechanistic studies: Use isogenic cell lines with KRT17 manipulation to determine causality in different contexts.
KRT17 has emerged as an unexpected participant in the DNA damage response (DDR):
Induction following damage: KRT17 expression is elevated in response to DNA damage .
Nuclear localization: A pool of KRT17 localizes to the nucleus in human and mouse tumor keratinocytes, where it impacts chromatin architecture, gene expression, and cell proliferation .
Early DSB response: Nuclear KRT17 is required at an early stage of the double-stranded break (DSB) arm of the DNA damage and repair cascade .
Protein associations: KRT17 physically associates with key DDR effectors, including γ-H2A.X, 53BP1, and DNA-PKcs .
Impact on tumorigenesis: Mice lacking K17 or with attenuated K17 nuclear import showed curtailed initiation in a two-step skin carcinogenesis model, suggesting that KRT17's role in DDR contributes to tumor development .
Methodological approaches to study this function include:
Immunofluorescence colocalization with DDR markers after damage induction
Proximity ligation assays to confirm protein-protein interactions
Chromatin fractionation to isolate nuclear KRT17
CRISPR-mediated knockout or knockdown with rescue experiments
In vivo models with targeted KRT17 nuclear localization disruption
KRT17 expression significantly impacts immune cell infiltration and function in tumors:
NK cell modulation: In HER2-high breast cancer, the levels of infiltrating activated natural killer (NK) cells differ significantly between KRT17-high and KRT17-low groups .
T cell effects: In ER-high breast cancer, the proportion of infiltrating naïve CD4 T cells differs significantly between KRT17 expression groups .
Cytokine signaling: KRT17 expression is related to cytokine signaling pathways, indicating that it may affect the tumor immune response .
IL-17 connection: The HER2 signaling pathway may affect IL-17 levels, which in turn could regulate KRT17 expression, creating a potential feedback loop .
Research methodologies to investigate these relationships include:
Multiplex immunohistochemistry to visualize KRT17 and immune markers simultaneously
Single-cell RNA sequencing to correlate KRT17 expression with immune cell states
Cytokine profiling in KRT17-manipulated models
Co-culture experiments with tumor and immune cells
In vivo immune cell depletion studies in KRT17-modulated tumors
Based on published research, the following experimental conditions yield reliable results:
For migration studies specifically:
Use a 20μl pipette tip for consistent scratch width
Serum-starve cells for 24h before assay
Wash with PBS to remove detached cells
Image at 0h, 12h, and 24h timepoints from the same area
Quantify using standardized image analysis software
Researchers have successfully employed several approaches to modulate KRT17 expression:
Protein supplementation:
Gene silencing:
Overexpression systems:
Plasmid transfection with KRT17 cDNA
Viral vectors for stable integration
Inducible expression systems for temporal control
Animal models:
For each approach, appropriate controls must include:
Vehicle-only or scrambled siRNA controls
Empty vector transfection controls
Wild-type or heterozygous littermates for animal studies
Validation of expression changes at both mRNA and protein levels
To study KRT17 in diabetic conditions:
In vitro glucose conditions:
Normal glucose: 5.5 mM
High glucose: 25-30 mM
Include osmotic controls (mannitol) to distinguish glucose-specific effects
Consider fluctuating vs. constant glucose levels
Cellular models:
In vivo models:
Molecular and functional readouts:
GO analysis targets:
For robust statistical analysis of KRT17 in clinical studies:
Survival analysis:
Kaplan-Meier curves with log-rank tests to compare high vs. low KRT17 expression groups
Cox proportional hazards regression for multivariate analysis
Adjustment for established prognostic factors (stage, grade, age, treatment)
Clinicopathological correlations:
Expression cutoff determination:
Subgroup analysis:
To address the context-dependent roles of KRT17:
Comprehensive molecular profiling:
Multi-level data integration:
Combine transcriptomics, proteomics, and clinical data
Network analysis to identify context-specific interacting partners
Machine learning approaches to identify patterns across cancer types
Functional validation studies:
Test KRT17 manipulation in multiple cell line models representing different subtypes
Assess phenotypic outcomes in different genetic backgrounds
Identify context-dependent binding partners through differential interactome analysis
Data presentation strategies:
Forest plots showing hazard ratios across cancer types and subtypes
Interaction plots demonstrating how additional factors modify KRT17's effect
Decision tree models incorporating KRT17 with other biomarkers
GO Category | Enriched Terms | Significance |
---|---|---|
Biological Process | Positive regulation of transcription | High |
Cell adhesion | High | |
Inflammatory response | High | |
Molecular Function | Metal ion binding | High |
Calcium ion binding | High | |
Carbohydrate binding | High | |
Cellular Component | Integral component of membrane | High |
Plasma membrane | High | |
Extracellular region | High |
This GO analysis from RNA-seq of KRT17-exposed cells provides insight into the diverse pathways through which KRT17 may exert its context-dependent effects.
Based on current evidence, several translational directions emerge:
Diagnostic biomarker development:
KRT17 as part of multi-marker panels for cancer detection
Context-specific interpretation based on tumor type and receptor status
Refinement of prognostic models incorporating KRT17
Therapeutic targeting:
Wound healing applications:
Cytokeratin 17, also known as Keratin 17 or KRT17, is a type I intermediate filament protein that plays a crucial role in the structural integrity of epithelial cells. It is predominantly found in the nail beds, hair follicles, sebaceous glands, and other epidermal appendages . The recombinant form of this protein, referred to as Human Recombinant Cytokeratin 17, is produced using various expression systems for research and diagnostic purposes.
Cytokeratin 17 is composed of 432 amino acids and has a predicted molecular weight of approximately 73.26 kDa . The protein is expressed in wheat germ and is suitable for applications such as SDS-PAGE, ELISA, and Western Blotting . The sequence of Cytokeratin 17 includes several glycine-rich regions that contribute to its structural properties.
Cytokeratin 17 is involved in the formation and maintenance of various skin appendages. It plays a significant role in determining the shape and orientation of hair, as well as in the structural integrity of nail beds and sebaceous glands . The protein is also implicated in cellular processes such as proliferation, differentiation, and apoptosis.
Mutations in the KRT17 gene can lead to several genetic disorders, including Jackson-Lawler type pachyonychia congenita and steatocystoma multiplex . These conditions are characterized by abnormalities in the skin, nails, and hair, highlighting the importance of Cytokeratin 17 in maintaining epithelial tissue integrity.
Recombinant Human Cytokeratin 17 is widely used in research to study its role in epithelial biology and related diseases. It is utilized in various assays, including SDS-PAGE, ELISA, and Western Blotting, to investigate protein expression, function, and interactions . The recombinant protein is also valuable in the development of diagnostic tools and therapeutic strategies for skin-related disorders.