KRT8 Human

Cytokeratin 8 Human Recombinant
Shipped with Ice Packs
In Stock

Description

Cancer Metastasis and EMT

  • Lung Adenocarcinoma (LUAD): High KRT8 expression promotes epithelial-mesenchymal transition (EMT), enhancing cell migration and invasion. Knockdown reduces tumor growth in vivo and downregulates mesenchymal markers (N-cadherin, Vimentin) .

  • Clear Cell Renal Cell Carcinoma (ccRCC): KRT8 upregulation drives metastasis via IL-11/STAT3 signaling .

  • Gastric Cancer: KRT8 stabilizes FAS ligand, enabling immune evasion .

Prognostic Biomarker Potential

Cancer TypeAssociation with KRT8Clinical Impact
LUADCorrelates with advanced T/N stage (p < 0.05); poor 5-year survival (HR = 1.526, p = 0.004) Independent predictor of OS and DSS
ccRCCOverexpressed in venous tumor thrombi; linked to metastasis (p < 0.001) Predictive of poor prognosis
Breast CancerCAM 5.2 antibody (KRT8/KRT18) identifies lobular carcinoma and Paget’s disease Diagnostic utility in immunohistochemistry

4. Research Tools and Reagents
KRT8 is widely studied using targeted assays and recombinant proteins:

ProductHostApplicationKey FeaturesSource
Recombinant KRT8 (TP309570)HEK293TCell culture, protein interactionC-Myc/DDK tag; >80% purityOriGene
KRT8 ELISA Kit (EH147RB)-Quantification in serum/plasmaSensitivity: 0.41 pg/mL; detects natural/KRT8Thermo Fisher
Recombinant KRT8-HisE. coliStructural studies, antibody productionHis-tagged; 56 kDa; >90% purityProSpec

Mechanistic Insights from Experimental Studies

  • In LUAD Models:

    • KRT8 knockdown via shRNA reduced proliferation (CCK-8 assay, p < 0.001) and induced apoptosis (Annexin V assay, p < 0.01) .

    • In vivo metastasis assays showed 50% fewer lung nodules in KRT8-silenced mice (p < 0.05) .

  • NF-κB Pathway Modulation: KRT8 regulates TNF-α–induced nuclear translocation of p65, influencing pro-survival signaling .

6. Commercial and Research Applications
KRT8 reagents are pivotal for:

  1. Diagnostics: CAM 5.2 antibody for histopathological differentiation of carcinomas .

  2. Therapeutic Research: Targeting KRT8-IL-11 axis in ccRCC or EMT pathways in LUAD .

  3. Biomarker Development: Plasma KRT8 levels correlate with tumor burden in epithelial cancers .

Product Specs

Introduction
Keratin 8 and 18 (K8/18) are the primary intermediate filament (IF) proteins found in simple epithelia, which are composed of a single layer of cells.
Description
Recombinant human Cytokeratin 8, expressed in E. coli, is a non-glycosylated polypeptide chain with a molecular weight of 53,532 Daltons. The purification of KRT8 is achieved through proprietary chromatographic techniques.
Physical Appearance
Sterile white powder obtained by lyophilization (freeze-drying).
Formulation
The protein solution (1 mg/ml) was lyophilized from a sterile solution containing 30 mM Tris-HCl (pH 8), 9.5 M urea, 2 mM DTT, 2 mM EDTA, and 10 mM methylammonium chloride.
Solubility
To reconstitute the lyophilized KRT8, it is recommended to dissolve it in sterile 18 MΩ·cm H2O at a concentration of at least 100 µg/ml. This solution can then be further diluted with other aqueous solutions as needed.
Stability
Lyophilized KRT8 remains stable at room temperature for 3 weeks; however, it is recommended to store it at 2-8°C. After reconstitution, KRT8 should be stored at 4°C for 2-7 days. For long-term storage, it is advised to store it below -18°C. The addition of a carrier protein (0.1% HSA or BSA) is recommended for long-term storage. Avoid repeated freeze-thaw cycles.
Purity
The purity is determined to be greater than 95.0% using the following methods: (a) RP-HPLC analysis, (b) SDS-PAGE analysis.
Reconstitution To Filaments
To form filaments, mix equal molar amounts of type I and type II cytokeratins at a concentration of approximately 0.5 mg/ml, both dissolved in a 9.5 M urea buffer (see above). Protofilaments and filament complexes can be obtained by stepwise dialysis of the polypeptide solution against a 4 M urea solution and then against a low salt buffer (50 mM NaCl, 2 mM dithiothreitol, 10 mM Tris-HCl, pH 7.4). For immunization purposes, the solution can be further dialyzed against PBS (e.g., Dulbecco's PBS).
Synonyms
Keratin type II cytoskeletal 8, Cytokeratin-8, CK-8, Keratin-8, K8, KRT8, CYK8, KO, CK8, K2C8, CARD2.
Source
Escherichia Coli.

Q&A

What is KRT8 and what is its fundamental role in human cells?

KRT8 (Keratin 8) is a type II intermediate filament protein that forms an essential component of the cytoskeleton in epithelial cells. Under physiological conditions, KRT8 pairs with KRT18 (Keratin 18) in a 1:1 ratio to form heterodimeric intermediate filaments . These structures provide mechanical stability to cells, protect against various stressors, and participate in cellular signaling networks. Beyond structural support, KRT8 plays critical roles in protecting cells from apoptosis and participates in various cell signaling cascades. Its expression is particularly important in simple epithelia such as those found in the liver, gastrointestinal tract, and lungs.

How is KRT8 expression distributed across different human tissues?

KRT8 shows a tissue-specific expression pattern predominantly in simple epithelial cells . In normal human tissues, significant KRT8 expression is observed in:

  • Digestive system: liver, gallbladder, pancreas, stomach, small intestine, colon, rectum

  • Reproductive system: prostate, testis, epididymis, seminal vesicle, fallopian tube, endometrium, cervix

  • Respiratory system: primarily in bronchial epithelium, not typically in alveolar spaces under normal conditions

  • Endocrine glands: thyroid, parathyroid, adrenal glands

  • Others: kidney, bladder, salivary glands

In contrast, tissues like brain, heart muscle, skeletal muscle, and skin show minimal or absent KRT8 expression under normal conditions . This tissue-specific distribution serves as an important baseline for understanding pathological alterations in KRT8 expression.

What is the significance of the KRT8/KRT18 ratio in normal and diseased tissues?

The KRT8/KRT18 ratio is critical for proper intermediate filament function. In normal epithelial tissues, KRT8 and KRT18 are expressed in a balanced 1:1 ratio . This balance is essential for:

  • Formation of proper heterodimeric intermediate filaments

  • Normal cytoskeletal architecture and function

  • Protection against mechanical and non-mechanical stress

  • Regulated cell signaling

Disruption of this balance has significant pathological implications. Elevated KRT8/KRT18 ratios have been specifically associated with:

  • Formation of Mallory-Denk bodies (MDBs) in steatohepatitis

  • Enhanced lipogenesis and altered lipid metabolism in liver diseases

  • Increased aggressiveness of hepatocellular carcinoma (HCC)

  • Poorer prognosis in several cancer types

Studies in mouse models have demonstrated that Krt18−/− mice (which have excess KRT8 without KRT18) develop steatohepatitis and are prone to HCC that resembles the S1 subclass of human HCC . The KRT8/KRT18 ratio has emerged as a potential biomarker for cancer aggressiveness and treatment response.

What experimental approaches are most effective for detecting KRT8 in human samples?

Several complementary approaches can be used for reliable KRT8 detection:

Protein-level detection:

  • Immunohistochemistry (IHC): Provides spatial information about KRT8 distribution in tissues. Particularly effective with monoclonal antibodies specific to KRT8.

  • Immunofluorescence: Allows co-localization studies with other markers. Double immunofluorescence for KRT8 and KRT18 has been effectively used to study their relationship .

  • Western blotting: Provides quantitative information about total KRT8 protein levels.

  • Flow cytometry: Enables quantification of KRT8-expressing cells in complex mixtures.

  • Mass spectrometry: Offers unbiased detection and absolute quantification of KRT8 peptides .

mRNA-level detection:

  • qRT-PCR: For quantitative analysis of KRT8 transcript levels.

  • RNA in situ hybridization: Provides spatial information about KRT8 mRNA expression.

  • RNA sequencing: Offers comprehensive transcriptomic profiling including KRT8.

  • Single-cell RNA sequencing: Provides cell-specific resolution of KRT8 expression, valuable for heterogeneous tissues .

How does KRT8 expression change during cellular stress or injury?

KRT8 expression undergoes dynamic changes during cellular stress and injury, particularly in the context of tissue regeneration. Research reveals distinct temporal patterns:

In lung injury models, KRT8 shows a characteristic expression pattern:

  • Initial induction: KRT8 expression begins ~3 days post-injury

  • Peak expression: Reaches maximum levels during the fibrogenic phase (days 10-14)

  • Resolution: Diminishes as regeneration completes

This pattern represents an injury-specific progenitor cell state, as KRT8+ cells were found to cover alveolar surfaces during repair. Importantly, this phenomenon is conserved between mouse models and human patients with acute lung injury and fibrosis .

In liver injury, changes in KRT8 expression relative to KRT18 appear particularly significant:

  • Increased KRT8/KRT18 ratio correlates with development of Mallory-Denk bodies (MDBs)

  • This altered ratio is associated with steatohepatitis and hepatocellular carcinoma development

These dynamic changes in KRT8 expression appear to be fundamental to cellular stress responses and tissue regeneration mechanisms rather than simply reflecting damage.

What signaling pathways interact with KRT8 in human cells?

KRT8 participates in multiple signaling networks beyond its structural role:

NF-κB pathway: Knockdown studies in lung adenocarcinoma have demonstrated that KRT8 regulates NF-κB signaling . This interaction may mediate KRT8's effects on cell proliferation, migration, and epithelial-to-mesenchymal transition.

WNT signaling: In hepatocellular carcinoma, altered KRT8/KRT18 ratios correlate with aberrant activation of the WNT pathway . This relationship appears particularly relevant in the S1 subclass of HCC.

Growth factor networks: KRT8+ progenitor cells express growth factors including amphiregulin (AREG) and heparin-binding EGF-like growth factor (HBEGF) . These factors mediate communication with surrounding cell types during tissue regeneration.

Cell death regulation: KRT8/KRT18 intermediate filaments regulate susceptibility to FAS-mediated apoptosis but not TNFα-mediated cell death . This selective modulation of death receptor signaling may contribute to tissue-specific outcomes of injury.

Integrin signaling: KRT8+ cells during lung regeneration show increased expression of integrin β6 (ITGB6) , suggesting involvement in cell-matrix interactions during repair processes.

These interactions position KRT8 as an active participant in cellular signaling networks rather than merely a structural component or passive biomarker.

How do alterations in KRT8 expression contribute to cancer progression?

KRT8 expression alterations significantly impact cancer development and progression through multiple mechanisms:

In lung adenocarcinoma (LUAD):

In hepatocellular carcinoma (HCC):

  • Elevated KRT8/KRT18 ratio predicts aggressive cancer phenotypes

  • KRT8 overexpression associates with enhanced lipogenesis and altered lipid metabolism

  • HCC with high KRT8/KRT18 ratios resembles the S1 molecular subclass, characterized by WNT pathway activation

  • Early recurrence (within 2 years post-surgery) is more common in patients with high KRT8/KRT18 ratios

These findings suggest that KRT8 may represent more than a biomarker—it appears to actively participate in oncogenic signaling and metabolic reprogramming that drives cancer progression. The mechanisms appear to be cancer-type specific but converge on promoting aggressive disease behavior.

What role does KRT8 play in epithelial differentiation and regeneration?

KRT8 serves critical functions in epithelial differentiation and regeneration, particularly following tissue injury:

In lung regeneration:

  • Single-cell RNA sequencing has identified a distinct KRT8+ progenitor cell state that appears transiently during repair

  • These cells feature a squamous morphology, covering damaged alveolar surfaces

  • RNA velocity analysis suggests KRT8+ cells derive from activated alveolar type 2 (AT2) cells and subsequently differentiate into alveolar type 1 (AT1) cells

  • Both airway stem cells (club cells) and AT2 cells can converge transcriptionally onto the same KRT8+ progenitor state, demonstrating plasticity in regeneration pathways

Developmental implications:

  • KRT8+ progenitor cells express genes associated with epithelial differentiation, including SPRR1A (cornifin-A), suggesting a role in squamous differentiation

  • The transient nature of KRT8 expression during regeneration parallels developmental processes where intermediate states are often marked by distinct keratin expression patterns

Conservation across species:

  • KRT8+ cells are observed in both mouse models of injury and human patients with acute respiratory distress syndrome (ARDS) and interstitial lung disease

  • This conservation suggests fundamental importance in epithelial regeneration mechanisms

These findings position KRT8 as a marker of a critical intermediate progenitor state during epithelial regeneration, with potential implications for therapeutic approaches to enhance tissue repair.

How can the KRT8/KRT18 ratio serve as a predictive biomarker in hepatocellular carcinoma?

The KRT8/KRT18 ratio has emerged as a sophisticated biomarker for hepatocellular carcinoma (HCC) with significant prognostic implications:

Molecular classification correlation:
Tumors with elevated KRT8/KRT18 ratios correspond to the S1 subclass of HCC according to established molecular classifications . This subclass is characterized by:

  • Aberrant WNT pathway activation

  • Generally more aggressive disease behavior

  • Distinct transcriptional programs

Prognostic value:
Analysis of mRNA expression profiles from HCC patient cohorts demonstrates:

  • High KRT8/KRT18 ratios (>1 standard deviation above mean) correlate with early HCC recurrence within 2 years post-surgery

  • This early recurrence is typically attributed to dissemination of primary tumor cells rather than de novo tumor formation

  • The prognostic value appears independent of traditional clinical parameters

Mechanistic basis:
The prognostic significance has biological underpinnings:

  • Altered KRT8/KRT18 ratios associate with changes in hepatic lipid metabolism that promote steatohepatitis

  • These metabolic alterations create a pro-oncogenic microenvironment

  • Experimental models show that Krt18−/− mice (with excess KRT8) develop HCCs that resemble human S1 subclass tumors

This biomarker represents an intersection between cytoskeletal dynamics, metabolic reprogramming, and oncogenic signaling, highlighting the multifaceted role of keratins beyond structural functions. Implementation in clinical settings would require standardization of measurement techniques and validation in prospective trials.

What mechanisms explain the emergence of KRT8+ progenitor cells during tissue regeneration?

The emergence of KRT8+ progenitor cells during tissue regeneration involves sophisticated cellular and molecular mechanisms:

Cell origin and lineage dynamics:

  • Single-cell RNA sequencing combined with RNA velocity analysis reveals that alveolar KRT8+ cells derive from activated alveolar type 2 (AT2) cells

  • This transition involves substantial transcriptional reprogramming

  • Remarkably, both AT2 cells and airway club cells can converge onto the same KRT8+ progenitor state, suggesting this is a key nodal point in regeneration

Signaling networks:

  • KRT8+ progenitors express a unique repertoire of signaling molecules including:

    • Growth factors: AREG (amphiregulin) and HBEGF (heparin-binding EGF-like growth factor)

    • Cell adhesion molecules: ITGB6 (integrin β6)

    • Epithelial differentiation regulators: SPRR1A (cornifin-A)

  • These cells feature a distinct "connectome" of receptor-ligand interactions with endothelial cells, fibroblasts, and macrophages

  • This specialized communication network likely orchestrates the coordinated tissue response to injury

Temporal dynamics:

  • KRT8+ cells appear transiently, peaking during the fibrogenic phase of repair (days 10-14 post-injury)

  • This timing corresponds to a critical transition between the inflammatory and resolving phases of wound healing

  • Their appearance is likely triggered by specific damage-associated molecular patterns (DAMPs) or inflammatory cytokines

Evolutionary conservation:

  • The KRT8+ progenitor state is observed in both mouse models and human patients with lung injury

  • This conservation suggests fundamental importance in mammalian tissue repair mechanisms

Understanding these mechanisms could enable targeted therapeutic approaches to enhance regeneration in conditions characterized by impaired tissue repair.

How do single-cell transcriptomic approaches reveal KRT8 function in heterogeneous tissues?

Single-cell transcriptomics has revolutionized our understanding of KRT8 function in complex tissues by revealing cellular heterogeneity and dynamic processes previously masked in bulk analyses:

Resolution of distinct cell states:

  • Single-cell RNA sequencing (scRNA-seq) has identified previously unrecognized KRT8+ cell populations, particularly during tissue regeneration

  • These approaches have revealed that cells with similar morphology can have distinct molecular profiles and developmental trajectories

  • In lung injury models, scRNA-seq identified a KRT8+ progenitor population that was previously undetectable by conventional methods

Trajectory inference:

  • RNA velocity analysis, which uses the ratio of spliced to unspliced reads, has enabled prediction of cell state transitions involving KRT8+ cells

  • This has revealed the directional flow from AT2 cells to KRT8+ progenitors to AT1 cells during lung regeneration

  • Without single-cell resolution, these transient states and trajectories would remain obscured

Cell-cell interaction networks:

  • Integration of receptor-ligand databases with scRNA-seq data has uncovered how KRT8+ cells communicate with their microenvironment

  • This has identified unique "connectomes" between KRT8+ progenitors and other cell types including endothelial cells, fibroblasts, and macrophages

  • Such analyses reveal the context-dependent function of KRT8+ cells beyond their intrinsic properties

Temporal dynamics:

  • High-resolution longitudinal sampling (e.g., 18 timepoints for sorted epithelial cells) has captured the complete life cycle of KRT8+ populations

  • This temporal detail reveals the transient nature of certain KRT8+ states that would be missed in cross-sectional studies

Methodological innovations:

  • Computational integration of lineage tracing with transcriptomic profiling

  • Inference of clonal relationships based on somatic mitochondrial mutations

  • Combined spatial and single-cell approaches to map KRT8 expression patterns while preserving tissue architecture

These approaches have transformed our understanding of KRT8 from a static structural protein to a dynamic marker of cellular states with context-dependent functions across different tissues and disease processes.

What therapeutic strategies could target KRT8 or KRT8+ cells in disease contexts?

Emerging research suggests several promising therapeutic strategies targeting KRT8 or KRT8+ cells:

In cancer contexts:

  • Direct targeting: Small molecule inhibitors or antibody-drug conjugates specifically targeting KRT8 in tumors with aberrant expression

  • Ratio normalization: Approaches to restore balanced KRT8/KRT18 expression in hepatocellular carcinoma, potentially reversing aggressive phenotypes

  • Pathway intervention: Targeting downstream effectors like NF-κB in lung adenocarcinoma, where KRT8 drives tumor progression through this pathway

  • Combinatorial approaches: Pairing KRT8-targeted therapies with conventional chemotherapy or immunotherapy, particularly in tumors where high KRT8 predicts poor outcomes

In regenerative medicine:

  • Progenitor cell augmentation: Methods to enhance or prolong the KRT8+ progenitor state in settings of impaired tissue regeneration

  • Biomimetic approaches: Synthetic matrices incorporating signals produced by KRT8+ cells to promote repair

  • Cell therapy: Ex vivo expansion and delivery of KRT8+ progenitors to injury sites

  • Pharmaceutical induction: Small molecules that promote the generation of KRT8+ cells from resident stem cells

Delivery strategies:

  • Nanoparticle-based delivery systems targeting specific tissue environments

  • Inhalation-based approaches for pulmonary applications

  • Liver-targeted formulations for hepatocellular applications

Potential challenges:

  • Specificity: KRT8 expression in multiple epithelial tissues raises off-target effect concerns

  • Timing: The transient nature of KRT8+ progenitor states requires precise temporal targeting

  • Context-dependency: KRT8 functions differently across tissues and disease states, necessitating context-specific approaches

These therapeutic directions remain largely exploratory, with substantial preclinical validation required before clinical translation. The most promising near-term applications may be in diagnostic and prognostic implementations rather than direct therapeutic targeting.

What are the optimal approaches for studying KRT8/KRT18 interactions in tissue samples?

Studying KRT8/KRT18 interactions in tissue samples requires specialized techniques to preserve and visualize their native relationships:

Immunohistochemical approaches:

  • Dual immunofluorescence staining: Using differentially labeled antibodies against KRT8 and KRT18 to visualize their co-localization

  • Proximity ligation assay (PLA): Detects protein-protein interactions when KRT8 and KRT18 are within 40nm of each other

  • Sequential immunostaining: When antibodies are from the same species, sequential protocols with blocking steps can be employed

Sample preparation considerations:

  • Fixation: 4% paraformaldehyde is generally suitable, but comparative studies with frozen and paraffin-embedded tissues are recommended for optimization

  • Antigen retrieval: Heat-induced epitope retrieval in citrate buffer (pH 6.0) often works well for keratin detection

  • Permeabilization: Critical for cytoskeletal proteins; Triton X-100 (0.2-0.5%) is commonly used

Quantitative assessment:

  • Ratio quantification: Techniques to accurately measure KRT8/KRT18 ratios include:

    • Western blotting with densitometry

    • Quantitative immunofluorescence with fluorescence intensity measurement

    • Mass spectrometry-based proteomics for absolute quantification

  • Spatial heterogeneity analysis: Assess variations in KRT8/KRT18 ratios across different regions of the same tissue

Advanced imaging methods:

  • Super-resolution microscopy: Techniques like STORM or STED to visualize intermediate filament organization at nanoscale resolution

  • Live cell imaging: For dynamic studies of KRT8/KRT18 interactions in cell culture models

  • Correlative light and electron microscopy: To relate filament ultrastructure to protein composition

Controls and validation:

  • Tissue-matched controls with known KRT8/KRT18 expression

  • Genetic models: Tissues from Krt18−/− mice serve as controls for KRT8-only expression

  • Peptide competition assays to confirm antibody specificity

These methodological considerations are particularly important when studying pathological conditions where altered KRT8/KRT18 ratios have biological significance, such as in hepatocellular carcinoma .

How should researchers design lineage tracing experiments to track KRT8+ progenitor cells?

Designing effective lineage tracing experiments for KRT8+ progenitor cells requires careful consideration of temporal dynamics and technical approaches:

Genetic lineage tracing system design:

  • Driver selection:

    • KRT8-CreERT2 transgenic lines for temporal control of lineage labeling

    • Consider transient vs. stable KRT8 expression when designing promoter elements

    • Alternative drivers (SPC-CreERT2 for AT2 cells) can be used to trace cells that become KRT8+

  • Reporter selection:

    • Multicolor reporters (Confetti, Rainbow) to distinguish clonal relationships

    • Dual recombination systems to specifically label cells transitioning through the KRT8+ state

    • Consider sensitivity of reporters, as KRT8 expression may be transient or low in some contexts

Temporal considerations:

  • Induction timing:

    • In injury models, tamoxifen administration at day 7-10 post-injury would target peak KRT8+ progenitor populations

    • Multiple labeling pulses at different timepoints can capture the dynamic KRT8+ population

  • Chase period:

    • Long-term tracing (8+ weeks) to observe terminal differentiation outcomes

    • Multiple intermediate timepoints to capture transitional states

Validation approaches:

  • Dual RNA/protein detection:

    • RNA in situ hybridization for Cre recombinase combined with KRT8 immunostaining

    • Single-molecule FISH for lineage tracing the transcriptional landscape

  • Complementary approaches:

    • scRNA-seq of lineage-labeled cells to confirm molecular identity

    • Combine with RNA velocity analysis to validate predicted trajectories

    • Correlate with proliferation markers to distinguish expansion vs. differentiation

Controls:

  • Reporter-only and Cre-only controls to assess leakiness

  • Induction in uninjured tissue to establish baseline labeling

  • Parallel tracing with known progenitor populations (e.g., AT2 cells in lung) for comparison

This experimental design would be particularly valuable for validating the proposed trajectory from AT2 cells through KRT8+ progenitors to AT1 cells in lung regeneration as suggested by computational analysis of scRNA-seq data .

What protocols yield optimal results for KRT8 immunostaining in different tissue types?

KRT8 immunostaining requires tissue-specific optimization to achieve reliable and reproducible results:

General principles across tissues:

  • Fixation: 10% neutral buffered formalin (24-48 hours) for routine FFPE samples; 4% paraformaldehyde (10-15 minutes) for frozen sections

  • Antigen retrieval: Heat-induced epitope retrieval with citrate buffer (pH 6.0) is most universally effective for keratins

  • Blocking: 5-10% normal serum with 0.1-0.3% Triton X-100 for permeabilization

  • Primary antibody incubation: Overnight at 4°C generally yields the best signal-to-noise ratio

  • Detection systems: Polymer-based systems often provide better sensitivity than ABC methods

Liver-specific considerations:

  • Mild protease digestion (0.01% trypsin, 5 minutes) may enhance KRT8 detection in fibrotic liver

  • Combined KRT8/KRT18 staining particularly important for assessing ratio

  • Consider using h-HTAA (heptameric oligothiophene) to detect β-sheet conformations in Mallory-Denk bodies alongside KRT8 staining

  • Lipid preservation protocols if correlating KRT8 expression with steatosis

Lung-specific considerations:

  • Perfusion fixation provides superior morphology for alveolar regions

  • Careful attention to inflation status of lungs to maintain alveolar architecture

  • Autofluorescence reduction techniques (sodium borohydride treatment or commercial kits) critical for immunofluorescence studies

  • Short fixation times (2-4 hours) may preserve antigenicity better in lung tissue

Tumor tissue considerations:

  • Tumor heterogeneity necessitates multiple sampling regions

  • Inclusion of tumor-normal interface in sections

  • Correlation with proliferation markers (Ki-67) or EMT markers in lung adenocarcinoma samples

  • Assessment of both intensity and percentage of positive cells for proper quantification

Antibody selection guidance:

  • For human tissues: Monoclonal antibodies like CAM5.2 or TS1 have shown good specificity

  • For mouse tissues: TROMA-I monoclonal antibody is widely validated

  • Validation using Krt8−/− tissues as negative controls where available

These optimized protocols are essential for reliable analysis of KRT8 expression patterns, particularly in contexts where KRT8 serves as a diagnostic, prognostic, or functional marker.

What are best practices for KRT8 knockdown or overexpression experiments?

Manipulating KRT8 expression requires careful experimental design to ensure specific and interpretable results:

Knockdown approaches:

  • siRNA strategy:

    • Design multiple siRNAs targeting different KRT8 regions

    • Screen for sequences achieving >80% knockdown efficiency

    • Include non-targeting control with similar chemical modifications

    • Typical concentration: 20-50nM for most cell lines

    • Consider reverse transfection for difficult-to-transfect epithelial cells

  • shRNA considerations:

    • Use inducible systems (e.g., doxycycline-regulated) to study temporal effects

    • Select for stable lines with moderate copy numbers to avoid integration effects

    • Empty vector and scrambled sequence controls are essential

    • Validate knockdown stability over multiple passages

  • CRISPR-Cas9 knockout:

    • Design gRNAs targeting early exons to ensure complete functional disruption

    • Clone derivation from single cells essential for complete knockout

    • Consider CRISPR interference (CRISPRi) for reversible suppression

Overexpression strategies:

  • Vector selection:

    • Use epithelial-appropriate promoters (e.g., CAG, EF1α) for consistent expression

    • Consider tet-inducible systems to control expression levels

    • Include epitope tags that don't interfere with filament assembly (small C-terminal tags preferred)

  • Expression level considerations:

    • Titrate expression to achieve near-physiological levels

    • Excessive overexpression can cause aggregation artifacts

    • Co-express KRT18 when studying filament assembly to maintain proper ratios

Validation requirements:

  • Multiple validation methods:

    • qRT-PCR for mRNA levels

    • Western blot for protein levels

    • Immunofluorescence for subcellular distribution

    • Monitor KRT18 levels and filament organization

  • Functional validation:

    • Rescue experiments with siRNA-resistant constructs

    • Assess impact on filament organization using confocal microscopy

    • Monitor cell morphology and mechanical properties

Assay selection based on context:

  • For cancer studies:

    • Proliferation, migration, invasion assays

    • NF-κB pathway activity assessment

    • EMT marker analysis

  • For liver cell studies:

    • Lipid metabolism assays

    • Stress response and apoptosis resistance tests

  • For lung epithelial studies:

    • Differentiation capacity assays

    • Wound healing/repair models

These practices ensure that observed phenotypes can be reliably attributed to specific changes in KRT8 expression rather than off-target effects or technical artifacts.

How can researchers effectively analyze KRT8 expression in cancer genomics datasets?

Analyzing KRT8 expression in cancer genomics datasets requires strategic approaches to extract meaningful biological insights:

Dataset selection and integration:

  • Primary resources:

    • The Cancer Genome Atlas (TCGA): Contains RNA-seq data across 33 cancer types

    • International Cancer Genome Consortium (ICGC): Provides complementary international datasets

    • Cancer Cell Line Encyclopedia (CCLE): For in vitro correlation

    • GEO datasets: Often contain valuable cohorts with specific clinical annotations

  • Integration techniques:

    • Batch effect correction (ComBat, Harmon-y) when combining datasets

    • Cross-platform normalization for microarray/RNA-seq comparison

Expression analysis strategies:

  • Beyond simple expression:

    • Calculate KRT8/KRT18 ratios rather than absolute expression alone

    • Consider co-expression patterns with other keratins

    • Analyze splice variants where data permits

  • Subtype stratification:

    • Correlate KRT8 expression with established molecular subtypes

    • In HCC, relate to S1-S3 classification system

    • In lung cancer, separate adenocarcinoma from squamous cell carcinoma analyses

Clinical correlation approaches:

Pathway and network analysis:

  • Enrichment approaches:

    • Gene Set Enrichment Analysis (GSEA) with KRT8-correlated genes

    • Pathway analysis focusing on NF-κB (lung cancer) or WNT (liver cancer) connections

  • Protein-protein interaction networks:

    • Construct networks centered on KRT8/KRT18

    • Identify hub proteins that may mediate KRT8's effects

Visualization recommendations:

  • Complex heatmaps showing KRT8 expression across cancer types, clinical features, and molecular subtypes

  • Forest plots for hazard ratios across different cancers or patient subgroups

  • Correlation networks visualizing KRT8's relationship with other cancer-relevant genes

This analytical framework can help researchers move beyond descriptive findings to mechanistic insights about KRT8's role in cancer biology.

What approaches are recommended for analyzing single-cell data to identify and characterize KRT8+ populations?

Analyzing single-cell data to identify and characterize KRT8+ populations requires specialized bioinformatic approaches:

Preprocessing and quality control:

  • Cell filtering parameters:

    • Minimum gene count thresholds (typically 200-500 genes per cell)

    • Maximum mitochondrial percentage (typically <20%)

    • Doublet removal using specialized algorithms (DoubletFinder, Scrublet)

  • Normalization considerations:

    • SCTransform or log-normalization with scaled factor

    • Consider RNA velocity analysis normalization requirements if trajectory inference is planned

KRT8+ population identification:

  • Unbiased clustering:

    • Use graph-based clustering (Louvain or Leiden algorithms)

    • Test multiple resolution parameters to capture rare populations

    • Hierarchical clustering may better resolve closely related epithelial states

  • Feature-based approaches:

    • KRT8 expression projection onto dimensionality reduction plots (UMAP/t-SNE)

    • Consider both binary (KRT8+ vs KRT8-) and quantitative (KRT8-high/medium/low) categorizations

    • Examine co-expression with KRT18 for ratio analysis

Characterization of KRT8+ cells:

  • Differential expression analysis:

    • Compare KRT8+ cells to relevant reference populations

    • For lung studies, compare to AT1 and AT2 cells

    • For liver, compare to hepatocytes and bile duct cells

    • Use MAST or other zero-inflation-aware methods designed for scRNA-seq

  • Signature extraction:

    • Define gene signatures characteristic of KRT8+ populations

    • Validate signatures across independent datasets

    • Test signature enrichment in bulk RNA-seq datasets

Trajectory analysis:

  • RNA velocity approaches:

    • Use spliced/unspliced ratios to infer developmental directionality

    • scVelo for dynamical modeling of cell state transitions

    • Validation of computationally inferred trajectories with lineage tracing data

  • Pseudotime ordering:

    • Monocle3 or Slingshot for reconstructing differentiation paths

    • Root trajectories at established progenitor populations

Cell-cell interaction analysis:

  • Receptor-ligand pairing:

    • CellPhoneDB or similar tools to identify communication with other cell types

    • Focus on the KRT8+ cell "connectome" with immune, stromal, and other epithelial cells

  • Niche reconstruction:

    • Identify cell types frequently co-localized with KRT8+ cells

    • Infer regulatory relationships from correlation analyses

These approaches have been successfully applied to identify KRT8+ alveolar progenitor cells in lung injury models, revealing their origin from AT2 cells and potential differentiation into AT1 cells .

What statistical methods are most appropriate for analyzing KRT8 as a prognostic biomarker?

Robust statistical analysis of KRT8 as a prognostic biomarker requires methods that account for the complexities of clinical data:

Cutpoint determination:

  • Data-driven approaches:

    • Minimum p-value method with correction for multiple testing

    • X-tile algorithm for visualizing the optimal cutpoint

    • Receiver operating characteristic (ROC) analysis to balance sensitivity and specificity

  • Biologically informed approaches:

    • Standard deviation-based thresholding (e.g., >1 SD above mean)

    • Percentile-based cutoffs (quartiles or tertiles)

    • Ratio-based thresholds for KRT8/KRT18

Survival analysis fundamentals:

  • Kaplan-Meier analysis:

    • Log-rank test for comparing high vs. low KRT8 expression groups

    • Stratification by important clinical covariates

    • Consider multiple endpoints (OS, DSS, RFS, PFS)

  • Cox proportional hazards regression:

    • Univariate analysis to establish initial association

    • Multivariate modeling to adjust for confounders

    • Test the proportional hazards assumption and use appropriate alternatives if violated

Advanced survival modeling:

  • Competing risks analysis:

    • Particularly important when analyzing disease-specific outcomes

    • Fine and Gray subdistribution hazard model

  • Joint modeling:

    • For integrating longitudinal KRT8 measurements with survival outcomes

  • Machine learning approaches:

    • Random survival forests for handling non-linear relationships

    • Regularized Cox models (elastic net) when incorporating multiple molecular markers

Validation strategies:

  • Internal validation:

    • Bootstrap resampling to assess stability of findings

    • Cross-validation for predictive performance

  • External validation:

    • Independent cohort validation with predefined cutpoints

    • Meta-analysis when multiple studies are available

Reporting recommendations:

  • Hazard ratios with confidence intervals and p-values

  • Survival curves with number-at-risk tables

  • C-index or other discrimination metrics

  • Calibration assessment for prediction models

  • Clear definition of the KRT8 measurement approach and cutpoint determination

How can researchers integrate transcriptomic and proteomic data to study KRT8 function comprehensively?

Integrating transcriptomic and proteomic data provides a more complete understanding of KRT8 biology across multiple regulatory levels:

Multi-omics data generation:

  • Matched sample collection:

    • Obtain RNA and protein from the same specimens whenever possible

    • Consider microdissection for heterogeneous tissues

    • Include temporal sampling when studying dynamic processes

  • Complementary technologies:

    • RNA-seq or microarray for transcriptome

    • Mass spectrometry-based proteomics for protein abundance and post-translational modifications

    • Ribosome profiling to assess translational efficiency

    • ATAC-seq or ChIP-seq for regulatory mechanisms

Integration approaches:

  • Correlation analysis:

    • Direct correlation between KRT8 mRNA and protein levels

    • Account for expected temporal offset (mRNA changes typically precede protein)

    • Identify genes with concordant vs. discordant regulation

  • Multi-omics factor analysis:

    • MOFA or similar methods to identify latent factors driving variation across data types

    • Relate these factors to biological processes and clinical outcomes

  • Network-based integration:

    • Construct protein-protein interaction networks centered on KRT8

    • Overlay with transcriptional co-expression networks

    • Identify network modules with coordinated regulation

Specific KRT8 analyses:

  • KRT8/KRT18 ratio assessment:

    • Compare ratios at mRNA vs. protein level to identify post-transcriptional regulation

    • Correlate ratio across regulatory layers with phenotypic outcomes

  • Post-translational modification mapping:

    • Identify phosphorylation, glycosylation, or other modifications of KRT8

    • Correlate modifications with altered function or localization

  • Protein complex analysis:

    • Identify KRT8-containing protein complexes via co-immunoprecipitation or proximity labeling

    • Correlate complex formation with transcriptional changes

Visualization and interpretation:

  • Multi-omics heatmaps:

    • Display coordinated changes across regulatory layers

    • Cluster samples based on integrated profiles

  • Pathway visualization:

    • Map integrated results onto biological pathways (e.g., NF-κB signaling in lung cancer)

    • Identify points of transcriptional vs. post-transcriptional regulation

Validation strategies:

  • Targeted experiments:

    • Validate key findings with orthogonal methods (e.g., Western blot, qPCR)

    • Use genetic manipulation to test causal relationships

  • System perturbation:

    • Assess how KRT8 knockdown affects both transcriptome and proteome

    • Look for feedback mechanisms between regulatory layers

This integrative approach has been successfully applied in lung injury models where both transcriptomic data (scRNA-seq) and proteomic measurements confirmed the transient expression of KRT8 in regenerating alveolar epithelium .

Product Science Overview

Introduction

Cytokeratin 8 (CK8), also known as Keratin 8 (K8), is a type II intermediate filament protein that plays a crucial role in the structural integrity and function of epithelial cells. It is encoded by the KRT8 gene located on the long arm of chromosome 12 . CK8 is typically found in simple single-layered epithelial cells and is known to heteropolymerize with Keratin 18 (K18) to form intermediate filaments .

Structure and Properties

Recombinant Human Cytokeratin 8 (rHuCTK-8) is produced in Escherichia coli (E. coli) and is a single, non-glycosylated polypeptide chain with a molecular mass of approximately 53,532 Daltons . The recombinant protein is purified using proprietary chromatographic techniques to ensure high purity and quality . The amino acid sequence of CK8 includes several key regions that contribute to its structural and functional properties .

Biological Function

CK8, along with K18, forms a network of intermediate filaments that provide mechanical support to epithelial cells. This network is essential for maintaining cell shape, integrity, and resilience against mechanical stress. CK8 is also involved in various cellular processes, including apoptosis, cell signaling, and differentiation .

Applications

Recombinant CK8 is widely used in research to study the function and regulation of intermediate filaments in epithelial cells. It is also utilized in various assays, including ELISA, immunoaffinity purification, protein arrays, and Western blotting . The recombinant protein’s high purity and stability make it a valuable tool for investigating the molecular mechanisms underlying epithelial cell biology.

Storage and Handling

Recombinant CK8 should be stored at -80°C to maintain its stability and prevent degradation. It is recommended to avoid repeated freeze-thaw cycles to preserve the protein’s functionality . The protein is typically supplied in a buffer containing Tris-HCl and reduced glutathione, which helps maintain its structural integrity .

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.