KRT17 Human

Cytokeratin 17 Human Recombinant
Shipped with Ice Packs
In Stock

Description

KRT17 Human Recombinant produced in E.Coli is a single, non-glycosylated polypeptide chain containing 455 amino acids (1-432 a.a) and having a molecular mass of 50.5kDa.
KRT17 is fused to a 23 amino acid His-tag at N-terminus & purified by proprietary chromatographic techniques.

Product Specs

Introduction
Cytokeratin 17, also known as KRT17, is a type I cytokeratin found in nail beds, hair follicles, sebaceous glands, and other epidermal appendages. Mutations in the KRT17 gene can lead to disorders such as Jackson-Lawler type pachyonychia congenita and steatocystoma multiplex. KRT17 plays a crucial role in the development and maintenance of various skin appendages, especially in determining hair shape and orientation. It also influences the function of TNF-alpha during hair cycling. Furthermore, KRT17 regulates protein synthesis and epithelial cell growth by binding to the adapter protein SFN and activating the Akt/mTOR pathway.
Description
Recombinant human KRT17, expressed in E. coli, is a single, non-glycosylated polypeptide chain. This protein consists of 455 amino acids (with amino acids 1-432 derived from the KRT17 sequence) and has a molecular weight of 50.5 kDa. It includes a 23 amino acid His-tag at the N-terminus. Purification is achieved using proprietary chromatographic techniques.
Physical Appearance
Clear, sterile-filtered solution.
Formulation
The KRT17 protein solution is provided at a concentration of 0.5 mg/ml. The solution contains 20 mM Tris-HCl buffer (pH 8.0), 10% glycerol, and 0.4 M Urea.
Stability
For short-term storage (up to 2-4 weeks), the product can be stored at 4°C. For extended storage, it is recommended to store the protein at -20°C. To ensure long-term stability, consider adding a carrier protein (0.1% HSA or BSA). Avoid repeated freeze-thaw cycles.
Purity
The purity of the KRT17 protein is greater than 90% as determined by SDS-PAGE analysis.
Synonyms
Keratin 17, PCHC1, 39.1, CK-17, K17, PC2, PC, cytokeratin-17, Keratin 17 Epitope S1, Keratin 17 Epitope S2, Keratin 17 Epitope S4, Keratin, Type I Cytoskeletal 1, keratin-17, Cytokeratin-17, Keratin-17.
Source
Escherichia Coli.
Amino Acid Sequence
MGSSHHHHHH SSGLVPRGSH MGSMTTSIRQ FTSSSSIKGS SGLGGGSSRT SCRLSGGLGA GSCRLGSAGG LGSTLGGSSY SSCYSFGSGG GYGSSFGGVD GLLAGGEKAT MQNLNDRLAS YLDKVRALEE ANTELEVKIR DWYQRQAPGP ARDYSQYYRT IEELQNKILT ATVDNANILL QIDNARLAAD DFRTKFETEQ ALRLSVEADI NGLRRVLDEL TLARADLEMQ IENLKEELAY LKKNHEEEMN ALRGQVGGEI NVEMDAAPGV DLSRILNEMR DQYEKMAEKN RKDAEDWFFS KTEELNREVA TNSELVQSGK SEISELRRTM QALEIELQSQ LSMKASLEGN LAETENRYCV QLSQIQGLIG SVEEQLAQLR CEMEQQNQEY KILLDVKTRL EQEIATYRRL LEGEDAHLTQ YKKEPVTTRQ VRTIVEEVQD GKVISSREQV HQTTR

Q&A

What is KRT17 and what are its fundamental molecular characteristics?

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

What methodologies are most effective for detecting and quantifying KRT17 expression in tissue samples?

The gold standard for KRT17 detection in clinical and research settings is immunohistochemistry (IHC). A standardized protocol includes:

Table 1: Standardized Scoring System for KRT17 Immunohistochemistry

ParameterScoreCriteria
Staining Intensity0Negative
1Weak
2Moderate
3Strong
Percentage of Positive Cells0<5%
15-25%
226-50%
351-75%
4>75%
Final ScoreIntensity × Percentage
Expression Classification<6Low expression
≥6High 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

How does KRT17 expression differ between normal tissues and pathological conditions?

  • 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 .

What are the molecular mechanisms by which KRT17 influences cancer progression and metastasis?

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

How should researchers reconcile the contradictory prognostic significance of KRT17 across different cancer types?

The prognostic significance of KRT17 varies by cancer type and molecular context:

Table 2: Differential Prognostic Value of KRT17 Across Cancer Types

Cancer TypeKRT17 ExpressionPrognostic AssociationReference
NSCLCHighPoor prognosis
HER2-high breast cancerLowPoor prognosis
HER2-low breast cancerHighPoor prognosis
ER-high breast cancerLowPoor prognosis
ER-low breast cancerHighPoor prognosis

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.

What role does KRT17 play in the DNA damage response pathway?

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

What is the relationship between KRT17 and the tumor immune microenvironment?

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

What are the optimal experimental conditions for studying KRT17's effect on cell proliferation and migration?

Based on published research, the following experimental conditions yield reliable results:

Table 3: Optimized Conditions for KRT17 Functional Studies

Experimental ParameterRecommended ConditionsNotes
Recombinant KRT17 Concentration1 ng/mlShowed maximal effect on proliferation
Incubation Time24h, 48h, 72hFor time-course analysis
Cell Density (Proliferation)1×10^4 cells/well (96-well)For CCK-8 assay
Cell Density (Migration)90-95% confluenceFor scratch assay
Controls0 ng/ml KRT17Vehicle-only control
Quantification MethodCCK-8 assayFor proliferation
Scratch wound healingFor 2D migration
Transwell assayFor invasion
MicroscopyDaily recordingTo track morphological changes

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

What methodologies are most effective for manipulating KRT17 expression in research models?

Researchers have successfully employed several approaches to modulate KRT17 expression:

  • Protein supplementation:

    • Recombinant human KRT17 at 0.1-10 ng/ml (optimal: 1 ng/ml)

    • Direct addition to culture medium for immediate effects

  • Gene silencing:

    • siRNA for transient knockdown

    • siRNA emulsion mixtures for in vivo application (demonstrated in diabetic wound healing)

    • shRNA for stable knockdown

    • CRISPR-Cas9 for complete knockout

  • Overexpression systems:

    • Plasmid transfection with KRT17 cDNA

    • Viral vectors for stable integration

    • Inducible expression systems for temporal control

  • Animal models:

    • KRT17 knockout mice

    • Mice with attenuated KRT17 nuclear import

    • Tissue-specific conditional knockout 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

How should researchers design experiments to study KRT17's role in the context of high glucose and diabetic conditions?

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:

    • Human keratinocytes (HaCaT cells have been validated)

    • Primary cells from diabetic and non-diabetic donors

    • 3D skin equivalents to better mimic tissue architecture

  • In vivo models:

    • db/db diabetic mice (validated for KRT17 studies)

    • Streptozotocin-induced diabetes models

    • Wound healing assessment with standardized protocols

  • Molecular and functional readouts:

    • KRT17 expression levels (IHC, Western blot, qPCR)

    • Proliferation assays (CCK-8)

    • Migration assays (scratch wound, Transwell)

    • Transcriptome analysis (RNA-seq identified 493 differentially expressed genes)

  • GO analysis targets:

    • Transcriptional regulation

    • Cell adhesion

    • Inflammatory response

    • Metal ion binding

    • Calcium ion binding

    • Membrane components

What statistical approaches are recommended for correlating KRT17 expression with clinical outcomes?

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:

    • Chi-square or Fisher's exact test for categorical variables (e.g., correlation with lymph node metastasis, p=0.040)

    • Student's t-test or Mann-Whitney U test for continuous variables

    • One-way ANOVA with least significant difference (LSD) method for multiple group comparisons

  • Expression cutoff determination:

    • ROC curve analysis to identify optimal cutpoint

    • Standardized cutoff (score ≥6 for high expression in IHC)

    • Sensitivity analysis with different thresholds

  • Subgroup analysis:

    • Stratification by molecular subtypes (e.g., HER2-high/low, ER-high/low)

    • Forest plots to visualize effect size across subgroups

    • Interaction tests to identify effect modification

How can researchers address the challenge of KRT17 having opposing prognostic values in different tumor contexts?

To address the context-dependent roles of KRT17:

  • Comprehensive molecular profiling:

    • Integrate KRT17 expression with receptor status (HER2, ER)

    • Consider pathway activation status (Wnt, MAPK, PI3K)

    • Assess cytokine and inflammatory markers

  • 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

Table 4: Gene Ontology Enrichment of KRT17-Responsive Genes

GO CategoryEnriched TermsSignificance
Biological ProcessPositive regulation of transcriptionHigh
Cell adhesionHigh
Inflammatory responseHigh
Molecular FunctionMetal ion bindingHigh
Calcium ion bindingHigh
Carbohydrate bindingHigh
Cellular ComponentIntegral component of membraneHigh
Plasma membraneHigh
Extracellular regionHigh

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.

What are the most promising translational applications of KRT17 research?

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:

    • Inhibition of KRT17 nuclear localization to attenuate DNA damage response in cancer

    • Modulation of KRT17 to enhance immune cell infiltration

    • Targeting of KRT17-dependent pathways (Wnt signaling, EMT)

  • Wound healing applications:

    • KRT17 silencing therapies for diabetic wounds

    • Biomarkers for predicting wound healing complications

    • Personalized approaches based on KRT17 expression patterns

Product Science Overview

Introduction

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.

Structure and Expression

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.

Biological Function

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.

Clinical Significance

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.

Applications in Research

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.

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 2024 Thebiotek. All Rights Reserved.