Domains: Central α-helical rod domain flanked by non-helical N-terminal (head) and C-terminal (tail) regions, characteristic of intermediate filament proteins .
Post-Translational Modifications: Phosphorylation regulates filament reorganization and interactions with signaling molecules .
Mechanical Support: Stabilizes cell structure and protects against stress .
Apoptosis Regulation: Caspase-cleaved KRT18 fragments (M30, M65) serve as biomarkers for cell death .
Drug Response: Modulates chemosensitivity in cancers (e.g., lung, breast) .
Liver Diseases:
Cancer:
Alternative Splicing in Gastric Cancer (Frontiers in Genetics, 2021) :
Experimental Model: KRT18 knockdown in AGS gastric cancer cells.
Findings:
Reduced proliferation and increased apoptosis via dysregulation of AS in genes like PTBP1 and HNRNPK.
RNA-seq revealed 1,278 KRT18-coexpressed genes enriched in RNA splicing pathways.
Mechanism: KRT18 modulates splicing factors (SFs) at transcriptional/post-transcriptional levels, altering AS profiles in apoptosis-related pathways.
TCGA Data Analysis (450 Stomach Adenocarcinomas) :
KRT18 expression significantly higher in tumor tissues vs. normal.
High KRT18 correlates with poor prognosis and drug resistance.
Therapeutic Implications:
Protein Interactions: Binds thrombin-antithrombin complexes (liver) , collagen XVII (skin) , and TRADD (apoptosis signaling) .
Pathways:
KRT18 (Keratin 18) is a type I intermediate filament protein that partners predominantly with the type II keratin KRT8 to form stable heterodimers. Together, they establish a complex cytoskeletal network that provides structural integrity and resilience to epithelial cells . This keratin-based network is essential for maintaining cell shape and stability . As an intermediate filament protein, KRT18 contributes to the mechanical strength of cells and helps them resist physical stress that could otherwise lead to cell rupture or damage.
In skeletal muscle, research indicates that KRT18 serves as an integral component of the intermediate filament network, working complementarily with KRT19 in both assembling keratin-based filaments and transducing contractile forces .
KRT18 participates in multiple cellular processes beyond cytoskeletal support:
Uptake of thrombin-antithrombin complexes by hepatic cells (by similarity)
Delivery of mutated CFTR (Cystic Fibrosis Transmembrane Conductance Regulator) to the plasma membrane
Interleukin-6 (IL-6)-mediated barrier protection (in partnership with KRT8)
Pathologically, KRT18 appears to function as an oncogene in several cancers, particularly colorectal cancer, where its expression correlates with advanced clinical parameters and poor prognosis .
Several methodologies have proven effective for KRT18 detection, depending on the specific research question:
Optimal for visualizing KRT18 distribution in formalin-fixed, paraffin-embedded tissues
Typically employs mouse monoclonal antibodies (e.g., ab233913) at concentrations around 1 μg/ml
Effective for quantitative protein expression analysis
Protocol typically includes:
Sample preparation by centrifugation (10,000 × g)
Protein concentration determination (Modified Lowry Protein Assay Kit)
SDS-PAGE (4-12% gradient gel)
Transfer to nitrocellulose membranes
Blocking in 3% milk in PBS with 0.1% Tween-20
Incubation with KRT18 antibody (e.g., DC-10 from Santa Cruz Biotechnology)
For KRT18 mRNA quantification using primers:
KRT18 deficiency in mouse models provides important insights into human non-alcoholic fatty liver disease pathogenesis. Studies with aged (17-20-months-old) Krt18−/− mice show they spontaneously develop:
Progressive pathology beginning with steatosis in younger animals
Features of steatohepatitis (SH) in older animals
Histopathological findings in Krt18−/− mice include:
Hepatocyte anisocytosis and anisokaryosis
Cleared-out cytoplasm in hepatocytes
Focal hemorrhagic parenchymal necrosis with neutrophil infiltration
Mallory-Denk bodies (MDBs) appearing as eosinophilic cytoplasmic clumps in ballooned hepatocytes
Interestingly, heterozygous Krt18+/− mice exhibited more pronounced pathological alterations than homozygous Krt18−/− mice, with higher frequency of liver tumors, particularly in males:
Genotype | Male Tumor Frequency | Female Tumor Frequency |
---|---|---|
Wild-type | ~30% | ~25% |
Krt18−/− | ~73% | ~22% |
Krt18+/− | ~80% | ~35% |
These findings suggest KRT18 deficiency creates an environment conducive to steatohepatitis and hepatocellular neoplasia, with potential implications for understanding human NAFLD progression and the gender disparity observed in clinical settings .
Multiple lines of evidence support KRT18's oncogenic role, particularly in colorectal cancer (CRC):
KRT18 protein expression is significantly elevated in CRC tissues (57.4% high expression) compared to normal colorectal tissues (27.8% high expression, p=0.002)
All tested human CRC cell lines showed KRT18 overexpression compared to normal colonic epithelial cells
Analysis of TCGA and GTEx databases confirmed dramatically increased KRT18 expression in both colon and rectal cancers compared to normal tissues (p<0.001)
Clinical Correlations in CRC:
KRT18 overexpression significantly correlates with:
Clinical Parameter | Statistical Significance |
---|---|
Advanced clinical stage | p=0.003 |
Deep tumor invasion | p=0.009 |
Lymph node metastasis | p=0.008 |
Distant metastasis | p=0.001 |
Poor differentiation | p=0.005 |
These findings collectively suggest that KRT18 serves as an oncogenic factor in CRC progression and represents a potential therapeutic target for improving patient prognosis .
Based on the available research, a comprehensive experimental approach should include:
siRNA-mediated knockdown using validated oligonucleotides (e.g., proprietary mixture from Santa Cruz Biotechnology at 3 nM concentration)
CRISPR/Cas9 gene editing for complete knockout models
Transgenic mouse models with tissue-specific KRT18 deficiency
Co-immunoprecipitation to assess KRT18 interactions with KRT8, KRT19, and other cytoskeletal proteins
Proximity ligation assays to visualize protein-protein interactions in situ
FRET-based approaches to evaluate dynamic interactions
Immunofluorescence microscopy with co-staining for KRT18 and KRT19 to evaluate filament organization
Super-resolution microscopy to visualize fine structural details
Electron microscopy to examine ultrastructural changes in filament architecture
Atomic force microscopy to measure cellular stiffness in KRT18-deficient vs. normal muscle cells
Traction force microscopy to quantify contractile forces
Ex vivo muscle contractility measurements in KRT18-deficient models
Site-directed mutagenesis of key phosphorylation sites in KRT18
Phospho-specific antibodies to monitor phosphorylation status during contraction
Kinase inhibition experiments to identify regulatory pathways
Such a multifaceted approach would help elucidate KRT18's complementary role to KRT19 in both the assembly of keratin-based filaments and the transduction of contractile forces in skeletal muscle .
Researchers should consider several critical methodological aspects when employing KRT18 as a cancer biomarker:
Ensure consistent tissue sampling procedures (biopsy location, size, preservation)
Standardize fixation protocols for IHC (typically formalin-fixed, paraffin-embedded)
Include matched normal-tumor pairs whenever possible
Consider tissue microarrays for high-throughput screening
For IHC: Determine optimal antibody concentration (typically 1 μg/ml for anti-KRT18)
For Western blot: Standardize protein extraction protocols and loading controls
For RT-PCR: Validate primer specificity and efficiency
For all methods: Include appropriate positive and negative controls
Establish clear scoring criteria for KRT18 expression (e.g., percentage of positive cells, staining intensity)
Use digital image analysis where possible to reduce subjective interpretation
Correlate expression with standardized clinical parameters (TNM staging, differentiation grade)
Determine appropriate cutoff values for "high" versus "low" expression
Use multivariate analyses to control for confounding variables
Employ Kaplan-Meier survival analysis with log-rank test for prognostic assessment
Perform Cox proportional hazards regression to identify independent predictors of survival
Validate findings in independent cohorts
Compare results across multiple detection methodologies
Correlate tissue expression with potential circulating biomarkers
These considerations are essential for establishing KRT18 as a reliable biomarker with clinical utility across different cancer types, particularly given its demonstrated value in colorectal cancer prognostication .
To investigate KRT18 post-translational modifications (PTMs) and their functional implications, researchers should implement a comprehensive strategy:
Mass spectrometry-based proteomics to map phosphorylation, glycosylation, acetylation sites
Phospho-specific antibodies to monitor phosphorylated KRT18 in various cellular contexts
Special focus on phosphorylation, which is known to regulate filament reorganization
Generate phosphomimetic mutants (Ser/Thr to Asp/Glu) and phospho-deficient mutants (Ser/Thr to Ala)
Introduce the mutants into KRT18-deficient cell lines
Assess functional consequences on filament organization, cell morphology, and disease-relevant phenotypes
Kinase inhibitor screens to identify enzymes regulating KRT18 phosphorylation
Co-immunoprecipitation to detect physical interactions with modifying enzymes
siRNA knockdown of candidate kinases/phosphatases to confirm their role in KRT18 regulation
Monitor changes in KRT18 PTMs during disease progression (e.g., steatosis to steatohepatitis)
Compare PTM patterns between normal and cancer tissues
Correlate specific PTMs with clinical parameters and outcomes
Assess impact of PTMs on KRT18-KRT8 heterodimer formation
Evaluate changes in filament solubility, organization, and mechanical properties
Determine effects on interactions with other cellular components
Develop compounds that specifically inhibit or enhance disease-relevant KRT18 modifications
Test their effects in cellular and animal models of disease
Evaluate potential for clinical application
This approach would provide crucial insights into how phosphorylation and other PTMs regulate KRT18 function in both normal physiology and disease states, potentially identifying novel therapeutic targets.
Based on research findings showing KRT18's correlation with invasion and metastasis , an optimal experimental design would include:
2D Migration Assays:
Wound healing/scratch assays with KRT18-knockdown vs. control cells
Transwell migration assays to quantify directional cell movement
3D Invasion Assays:
Matrigel invasion chambers to assess ECM penetration capacity
Spheroid invasion assays in collagen matrices for more physiological assessment
Cell Line Selection:
Utilize paired cell lines with different metastatic potential (e.g., SW480/SW620 for CRC)
Generate isogenic lines differing only in KRT18 expression levels
Adhesion Dynamics:
Focal adhesion turnover assays in KRT18-modulated cells
Analysis of integrin expression and activation patterns
Cytoskeletal Interactions:
Co-immunoprecipitation of KRT18 with actin, microtubules, and associated proteins
Live-cell imaging of cytoskeletal dynamics during migration
Signaling Pathway Analysis:
Phosphoproteomics to identify KRT18-dependent signaling events
Targeted inhibition of candidate pathways to establish causality
Orthotopic Xenografts:
Implantation of KRT18-modulated cancer cells into relevant organs
Real-time tracking of tumor cell dissemination using bioluminescence
Metastasis Assays:
Tail vein injection to assess extravasation and colonization capacity
Quantification of circulating tumor cells in KRT18-high vs. KRT18-low models
Tissue Microarray Analysis:
KRT18 staining in primary tumors vs. matched metastases
Correlation with invasion markers (MMPs, EMT markers)
Liquid Biopsy Integration:
Analysis of circulating KRT18 levels in relation to metastatic burden
Correlation with CTCs and disease progression
This comprehensive approach would provide mechanistic insights into how KRT18 overexpression promotes invasion and metastasis in cancers, building on existing correlative clinical data .
When facing contradictory results between different KRT18 knockout models, researchers should implement a systematic approach to resolve these discrepancies:
Compare mouse strains used in different studies (the search results mention 129P2/OlaHsd background)
Generate knockouts on multiple genetic backgrounds to assess strain-dependent effects
Consider the presence of genetic modifiers that might influence phenotype penetrance
Compare homozygous (Krt18−/−) vs. heterozygous (Krt18+/−) phenotypes
Note that search results showed more pronounced pathology in Krt18+/− mice than in Krt18−/− mice
Investigate potential compensatory mechanisms in complete knockout models
Employ tissue-specific conditional knockouts to isolate effects
Use inducible systems to distinguish developmental from adult-onset phenotypes
Consider age-dependent effects (search results highlight phenotype progression with age)
Analyze male and female animals separately
Note significant sexual dimorphism in tumor development (males showed higher tumor frequency)
Investigate hormonal influences on phenotype expression
Standardize housing conditions, diet, and microbiome
Consider environmental stressors that might trigger or exacerbate phenotypes
Design challenge experiments to reveal latent phenotypes
Profile expression of other keratins that might compensate for KRT18 loss
Investigate the relationship between KRT18 and KRT19 in different tissues
Generate double or triple knockouts to overcome functional redundancy
Harmonize phenotyping protocols between laboratories
Establish consistent criteria for pathological assessment
Use quantitative rather than qualitative measures where possible
By systematically addressing these factors, researchers can better understand context-dependent KRT18 functions and resolve apparent contradictions between experimental models.
When manipulating KRT18 expression for functional studies, the following controls are essential for rigorous experimental design:
For siRNA Studies:
Non-targeting siRNA with similar GC content
Multiple siRNA sequences targeting different regions of KRT18 to rule out off-target effects
Rescue experiments with siRNA-resistant KRT18 constructs
For CRISPR/Cas9 Knockout:
Non-targeting gRNA controls
Single cell-derived clones with sequence verification
Multiple independent knockout clones to account for clonal variation
Rescue experiments with exogenous KRT18 expression
mRNA Level:
Protein Level:
Western blot with validated antibodies
Multiple loading controls
Immunofluorescence to verify subcellular localization changes
Assessment of potential compensatory changes in other keratins (especially KRT8 and KRT19)
Evaluation of heterodimer partner KRT8 expression and localization
Monitoring of other intermediate filament proteins
For Viability Assays:
Multiple timepoints to distinguish acute vs. chronic effects
Different methodologies (e.g., MTT, ATP-based assays, cell counting)
For Migration/Invasion Assays:
Proliferation controls to distinguish migration from proliferation effects
Both 2D and 3D assay formats to confirm consistent phenotypes
Experiments in multiple cell lines to ensure generalizability
Include both normal and cancer cell lines when studying oncogenic functions
Controls for cell density and confluency effects
Time course experiments to capture dynamic effects
Inducible systems to control onset of KRT18 manipulation
Inhibitors or genetic manipulation of proposed downstream pathways
Structure-function studies with mutant KRT18 variants
These comprehensive controls ensure that observed phenotypes are specifically attributable to KRT18 manipulation rather than experimental artifacts or off-target effects.
Interpreting differences in KRT18 expression between primary tumors and metastatic lesions requires a nuanced analytical approach:
Determine whether KRT18 upregulation is a driver or consequence of metastasis
Consider whether expression changes reflect selection of pre-existing subclones or adaptive responses
Evaluate if expression differences correlate with specific metastatic routes or target organs
Consistent Upregulation in Metastases:
Consistent Downregulation in Metastases:
Could suggest KRT18 inhibits later stages of metastatic cascade
May indicate different cytoskeletal requirements at secondary sites
Warrants investigation of epithelial-mesenchymal transition dynamics
Heterogeneous Expression:
Might reflect tumor heterogeneity and polyclonal metastatic origin
Could indicate context-dependent roles in different microenvironments
Should trigger single-cell analysis approaches
Correlate expression patterns with specific metastatic sites (liver, lung, lymph nodes)
Assess whether expression differences predict organ-specific metastatic tropism
Determine if metastasis-specific patterns correlate with treatment response
For upregulated KRT18 in metastases, focus on:
Anoikis resistance mechanisms
Cell-ECM interactions at secondary sites
Adaptation to new microenvironments
For downregulated KRT18 in metastases, investigate:
EMT-related cytoskeletal remodeling
Phenotypic plasticity mechanisms
Alternative structural proteins compensating for KRT18
High KRT18 in metastases might indicate utility as a therapeutic target for advanced disease
Expression differences could inform stage-specific treatment strategies
Changes might predict sensitivity to cytoskeleton-targeting drugs
The fact that KRT18 expression correlates with lymph node metastasis (p=0.008) and distant metastasis (p=0.001) in CRC provides a foundation for such comparative studies between primary and metastatic lesions.
The phosphorylation of KRT18 plays a critical role in filament reorganization , with significant implications for cellular stress responses:
Phosphorylation alters the charge distribution on KRT18 molecules
This modification disrupts inter-filament interactions, promoting filament solubility
Specific serine residues (likely including those in the head and tail domains) serve as phosphorylation sites
Phosphorylation enables rapid and reversible reorganization of the keratin network
Mechanical Stress Protection:
Phosphorylation-induced reorganization allows cytoskeletal adaptation to mechanical forces
Provides structural plasticity while maintaining cellular integrity
May protect against cell rupture during tissue deformation
Metabolic Stress Response:
Oxidative Stress Handling:
Phosphorylated KRT18 may interact differently with redox-sensitive proteins
Could affect cellular antioxidant responses
May influence susceptibility to oxidative damage in steatohepatitis
KRT18 phosphorylation likely integrates signals from multiple stress-activated kinases
Serves as both a sensor and effector in stress response pathways
May coordinate cytoskeletal responses with other cellular stress adaptations
In Liver Disease:
In Cancer:
Targeting kinases that phosphorylate KRT18 could modify filament dynamics
Modulating KRT18 phosphorylation might enhance cellular stress resistance
Could represent a strategy to normalize cytoskeletal function in disease states
The significant role of phosphorylated KRT18 in filament reorganization provides a foundation for understanding how post-translational modifications of this protein contribute to cellular stress adaptations in both physiological and pathological contexts.
Given KRT18's established role as an oncogenic factor in multiple cancers , several innovative therapeutic approaches warrant investigation:
Aptamer-Based Approaches:
Develop RNA/DNA aptamers that specifically bind KRT18
Conjugate with cytotoxic agents for targeted delivery to KRT18-overexpressing cells
Engineer aptamers that disrupt KRT18-KRT8 heterodimer formation
Proteolysis-Targeting Chimeras (PROTACs):
Design bifunctional molecules linking KRT18-binding ligands to E3 ubiquitin ligase recruiters
Induce selective proteasomal degradation of KRT18 in cancer cells
Overcome limitations of traditional inhibition approaches
Kinase Inhibition:
Phosphatase Activation:
Activate phosphatases that dephosphorylate KRT18
Stabilize keratin filaments to restrict cancer cell plasticity and motility
Epigenetic Modulation:
Target epigenetic modifiers controlling KRT18 expression
Develop selective enhancer-blocking compounds
Explore CRISPR-based epigenome editing of KRT18 regulatory regions
Antisense Oligonucleotides/siRNA Therapeutics:
Develop delivery systems for KRT18-targeting oligonucleotides
Leverage nanoparticle technology for tumor-specific delivery
Design modified siRNAs with enhanced stability and specificity
CAR-T Cell Therapy:
Generate CAR-T cells recognizing KRT18-derived peptides presented on cancer cells
Explore dual-targeting approaches to enhance specificity
Investigate potential for targeting circulating tumor cells with high KRT18 expression
Antibody-Drug Conjugates:
Develop antibodies targeting cancer-specific KRT18 conformations or modifications
Conjugate with cytotoxic payloads for selective delivery
Optimize internalization and drug release properties
Identify Genetic Dependencies:
Screen for genes synthetically lethal with high KRT18 expression
Target partner proteins essential for KRT18's oncogenic functions
Explore dependencies on specific cytoskeletal regulators in KRT18-high cancers
Sensitization to Standard Therapies:
Investigate KRT18 inhibition as a means to overcome chemoresistance
Explore synergies with cytoskeleton-targeting agents
Develop rational combinations based on KRT18-dependent pathways
The strong correlation between KRT18 expression and poor prognosis in colorectal cancer provides a compelling rationale for pursuing these therapeutic approaches, potentially offering new strategies for patients with KRT18-overexpressing tumors.
Understanding KRT18's tissue-specific functions in normal physiology represents an important research frontier, with several promising directions:
Differential Interactome Mapping:
Apply proximity labeling techniques (BioID, APEX) to identify tissue-specific KRT18 interacting partners
Compare KRT18 protein complexes across hepatocytes, intestinal epithelium, and other expressing tissues
Identify tissue-specific adaptors and regulatory proteins
Transcriptomic Co-expression Networks:
Analyze tissue-specific co-expression patterns to identify functional networks
Compare KRT18 regulatory mechanisms across tissues
Identify tissue-specific transcription factors controlling KRT18 expression
Cryo-Electron Microscopy:
Determine high-resolution structures of tissue-specific KRT18-KRT8 filaments
Investigate structural differences in filament organization across tissues
Analyze how tissue-specific post-translational modifications alter filament structure
In-Cell NMR:
Characterize dynamic aspects of KRT18 in living cells from different tissues
Monitor conformational changes during cellular processes
Identify tissue-specific structural adaptations
Biomechanical Characterization:
Mechanotransduction Studies:
Analyze KRT18's role in converting mechanical stimuli to biochemical signals
Compare mechanosensitive pathways across tissues
Investigate interactions with tissue-specific mechanosensors
Inducible Tissue-Specific Knockout Systems:
Generate conditional KRT18 knockout models for specific tissues
Analyze acute vs. chronic effects of KRT18 loss
Investigate compensatory mechanisms in different cellular contexts
Tissue-Specific Phospho-Mutant Models:
Lineage-Tracing Studies:
Track KRT18 expression during tissue development and specification
Investigate potential roles in epithelial differentiation programs
Analyze interactions with developmental signaling pathways
Single-Cell Multi-Omics:
Profile KRT18 expression, modifications, and interactions at single-cell resolution
Map changes during tissue development and homeostasis
Identify cell-state-specific functions
Signaling Scaffold Roles:
Investigate KRT18's function in organizing signaling complexes
Compare signaling roles across tissues
Analyze tissue-specific consequences of KRT18-mediated signaling
Cell Death Regulation:
Explore KRT18's role in tissue-specific apoptotic pathways
Investigate caspase-mediated cleavage in different contexts
Analyze fragment-specific functions across tissues
These research directions would significantly advance our understanding of KRT18's diverse roles beyond its well-established structural functions, potentially revealing new therapeutic opportunities for tissue-specific pathologies.
CK18 is predominantly expressed in single-layer epithelial tissues throughout the body, including the gastrointestinal tract, respiratory tract, urogenital tract, as well as endocrine and exocrine tissues . It plays a crucial role in maintaining the structural stability of these cells and is involved in various cellular processes such as cell signaling, apoptosis, and stress response .
Mutations in the CK18 gene have been linked to cryptogenic cirrhosis, a liver disease of unknown origin . Additionally, CK18 is a valuable biomarker in the diagnosis and prognosis of various cancers. It is present in a majority of adenocarcinomas and ductal carcinomas but is not typically found in squamous cell carcinomas . Hepatocellular carcinomas, a type of liver cancer, express only CK8 and CK18 .
Recombinant human CK18 is produced using an expression system, typically in Escherichia coli (E. coli), and is purified to high levels of purity for research purposes . This recombinant protein is often tagged with a hexahistidine (His) tag to facilitate purification and detection in various assays . It is used as a positive control in immunological assays and is valuable in studying the protein’s function and interactions .
CK18 is widely used in research to study epithelial cell biology, cancer, and liver diseases. Its expression patterns are utilized in distinguishing different types of epithelial malignancies through immunohistochemistry, cytopathology, and flow cytometric assays . The recombinant form of CK18 allows researchers to investigate its role in cellular processes and its interactions with other proteins under controlled conditions .