KIFC3 antibodies are immunoreagents designed to detect and study the KIFC3 protein, a minus end–directed microtubule motor implicated in apical vesicle transport in epithelial cells and oncogenic processes . These antibodies enable researchers to investigate KIFC3's localization, expression levels, and functional roles in diseases like colorectal cancer (CRC) and non-small cell lung cancer (NSCLC).
KIFC3 antibodies are utilized in diverse methodologies:
Western Blot (WB): Detects KIFC3 at ~80–85 kDa in lysates from CRC and NSCLC cell lines.
Immunohistochemistry (IHC): Highlights KIFC3 overexpression in tumor tissues, correlating with poor prognosis in CRC and NSCLC .
Immunofluorescence (IF): Localizes KIFC3 to cytoplasmic vesicles and cell junctions in polarized epithelial cells .
Co-Immunoprecipitation (CoIP): Identifies interactions between KIFC3 and partners like PI3Kp85α or USP47 .
Colorectal Cancer:
Non-Small Cell Lung Cancer:
KIFC3 binds USP47 at cell junctions, stabilizing E-cadherin by suppressing its ubiquitination .
Co-IP studies confirm direct interaction with PI3Kp85α, linking KIFC3 to oncogenic signaling .
Specificity: Variability in observed molecular weights (e.g., 80–100 kDa) suggests post-translational modifications or isoform detection .
Species Reactivity: Proteintech’s antibody detects multiple species, whereas Sigma’s is human-specific .
Validation: Functional assays (e.g., siRNA knockdown) are critical to confirm antibody specificity in experimental systems .
KIFC3 is a C-terminal motor domain-type kinesin superfamily protein that functions as a minus end-directed microtubule motor. In polarized epithelial cells, KIFC3 localizes on membrane organelles immediately beneath the apical plasma membrane of renal tubular epithelial cells in vivo and polarized MDCK II cells in vitro . Its primary function involves the transport of membrane vesicles along microtubules, particularly in apically targeted transport pathways within polarized cells . Additionally, KIFC3 plays crucial roles in Golgi positioning and integration, as disruption of the kifC3 gene can induce fragmentation of the Golgi apparatus under conditions such as cholesterol depletion . Recent research has also highlighted KIFC3's involvement in cancer progression, particularly in non-small cell lung cancer (NSCLC) .
KIFC3 antibodies serve multiple research applications across various technical platforms. According to validated protocols, KIFC3 antibodies have demonstrated effectiveness in:
Western Blot (WB): Detects KIFC3 protein at 80-85 kDa and 95-100 kDa molecular weights with recommended dilutions of 1:500-1:1000 .
Immunohistochemistry (IHC): Effectively labels KIFC3 in tissue sections at dilutions of 1:50-1:500, with positive detection in human intrahepatic cholangiocarcinoma tissue .
Immunofluorescence (IF): Visualizes KIFC3 localization in cellular compartments .
Immunoprecipitation (IP): Successfully pulls down KIFC3 using 0.5-4.0 μg of antibody for 1.0-3.0 mg of total protein lysate .
Co-immunoprecipitation (CoIP): Detects KIFC3 interactions with other proteins, which has been particularly valuable in studying KIFC3's association with transport vesicles and signaling pathway components .
These applications have been validated with human and mouse samples, making KIFC3 antibodies versatile tools for cross-species research .
Validating KIFC3 antibody specificity requires a multi-faceted approach to ensure experimental reliability:
Knockout/Knockdown Controls: Utilize KIFC3 knockout or knockdown models as negative controls. Published research demonstrates that KIFC3 antibodies show no reactivity in KIFC3 knockout mice, confirming specificity . The antibody recognized multiple protein bands in wild-type kidney samples, but these bands were absent in KIFC3 knockout mice .
Western Blot Validation: Verify the antibody detects bands at the expected molecular weights (80-85 kDa and 95-100 kDa) . Multiple bands may represent post-translational modifications or NH₂-terminally truncated proteins, as suggested by research showing only the highest band was recognized by anti-KIFC3 NH₂-terminus antibody .
Cross-reactivity Assessment: Test antibody performance across multiple species (human, mouse) and tissue types to confirm consistent recognition patterns .
Positive Control Tissues: Include known KIFC3-expressing tissues such as mouse lung, mouse kidney, and human intrahepatic cholangiocarcinoma as positive controls in validation experiments .
Immunofluorescence Correlation: Compare KIFC3 staining patterns with known interacting partners or subcellular structures (e.g., apical membrane in polarized cells, Golgi apparatus in adrenocortical cells) to confirm expected localization patterns .
Investigating KIFC3's role in cancer progression requires sophisticated experimental designs that address multiple aspects of cancer biology:
Studying KIFC3's role in cellular transport requires specialized methodologies that capture the dynamic nature of these processes:
Live Cell Imaging:
Subcellular Fractionation and Vesicle Isolation:
Isolate membrane organelles using flotation assays coupled with detergent extraction to identify KIFC3 association with specific vesicle populations.
Research has shown that KIFC3 associates with Triton X-100-insoluble membrane organelles that overlap with apically transported TGN-derived vesicles .
Transport Assays with Perturbation Approaches:
Use low-temperature blocking (18.5°C incubation) to partially inhibit vesicular exit from the TGN and observe KIFC3 redistribution.
Apply brefeldin A (BFA) treatment (2 μg/ml) to selectively inhibit TGN to apical surface transport.
Quantify changes in apical localization using quantitative confocal microscopy after immunofluorescent staining .
Protein-Protein Interaction Studies:
Conduct co-immunoprecipitation assays to confirm interactions between KIFC3 and transport vesicle components.
Perform GST pulldown experiments to identify specific interactions, such as those demonstrated between KIFC3 and annexin XIIIb, a membrane protein characteristic of apically transported vesicles .
Functional Motor Assays:
Distinguishing between KIFC3-dependent and dynein-dependent transport pathways requires careful experimental design:
Molecular Composition Analysis:
Perform immunoisolation of transport vesicles followed by proteomic analysis to define the complete composition of KIFC3-associated vesicles versus dynein-associated vesicles.
Research has demonstrated the absence of the dynein-dynactin complex on KIFC3-associated vesicles, suggesting at least two independent apically targeted transport pathways .
Selective Inhibition Approaches:
Use dominant-negative constructs of KIFC3 to specifically inhibit KIFC3-dependent transport without affecting dynein function.
Apply dynein-specific inhibitors or dominant-negative constructs to selectively block dynein-dependent transport.
Compare the effects of these selective inhibitions on different cargo molecules to map pathway-specific transport routes.
Cargo-Specific Transport Assays:
Track the transport of specific cargoes known to utilize different motor proteins.
Identify distinguishing characteristics of vesicles transported by each pathway, such as size, density, or membrane composition.
Double Knockdown/Knockout Experiments:
Create single and double knockdowns/knockouts of KIFC3 and dynein components.
Compare phenotypes to identify synergistic or independent effects that would indicate separate or overlapping pathways.
Designing effective KIFC3 knockdown experiments requires careful consideration of multiple factors:
Knockdown Method Selection:
Small interfering RNA (siRNA): Provides transient knockdown suitable for short-term experiments. This approach has been successfully used to demonstrate that KIFC3 knockdown suppresses proliferation, invasion, and migration in NSCLC cells .
Short hairpin RNA (shRNA): Offers more stable, long-term knockdown for extended studies or in vivo experiments.
CRISPR-Cas9: Provides complete gene knockout when assessing null phenotypes is preferred.
Validation of Knockdown Efficiency:
Quantify KIFC3 mRNA levels using RT-PCR to confirm transcriptional suppression.
Assess protein levels via western blot to verify translational inhibition, using validated KIFC3 antibodies at recommended dilutions (1:500-1:1000) .
Include appropriate controls: scrambled siRNA/shRNA sequences or non-targeting CRISPR guides.
Phenotypic Analysis:
Proliferation: Employ MTT and colony formation assays to assess effects on cell growth .
Migration: Use wound healing assays to measure migratory capability .
Invasion: Conduct Matrigel invasion assays to evaluate invasive potential .
Signaling pathway analysis: Measure protein levels of PI3K/Akt pathway components via western blot to determine mechanistic effects .
Rescue Experiments:
Express siRNA-resistant KIFC3 constructs to confirm phenotype specificity.
Use mutant KIFC3 constructs (e.g., motor-dead variants) to identify which domains are essential for observed functions.
Timing Considerations:
Account for KIFC3 protein half-life when designing experimental timelines.
Allow sufficient time after knockdown before phenotypic assessment, particularly for processes that may have delayed effects.
Investigating KIFC3's interaction with the PI3K/Akt pathway requires multifaceted experimental approaches:
Protein-Protein Interaction Analysis:
Perform co-immunoprecipitation assays to confirm direct interaction between KIFC3 and PI3K components. Research has confirmed interaction between endogenous/exogenous KIFC3 and PI3Kp85α .
Use GST pulldown assays to map the specific binding domains mediating these interactions.
Apply proximity ligation assays to visualize interactions in intact cells.
Pathway Activation Assessment:
Pharmacological Intervention:
Functional Rescue Experiments:
Attempt to rescue KIFC3 knockdown phenotypes through constitutive activation of PI3K/Akt pathway components.
Design KIFC3 mutants with altered ability to interact with PI3K components to define functional domains.
In Vivo Validation:
Optimizing KIFC3 antibody use in immunohistochemistry requires careful attention to several technical parameters:
Antigen Retrieval Method:
Antibody Dilution Range:
Positive Control Selection:
Detection System Considerations:
Use amplification systems for low-abundance expression.
Consider chromogen selection based on colocalization needs with other markers.
Counterstaining Parameters:
Adjust hematoxylin timing to ensure nuclear detail without obscuring cytoplasmic KIFC3 staining.
Monitor counterstain pH to prevent interference with antibody binding.
When encountering conflicting data about KIFC3 expression or function, researchers should implement a systematic reconciliation approach:
Antibody Validation Comparison:
Isoform Analysis:
Investigate potential splice variants or post-translational modifications that might explain discrepancies. Research has noted multiple bands in western blots that may represent NH₂-terminally truncated proteins produced by posttranslational modification or specific degradation pathways .
Perform RT-PCR with primers targeting different regions to identify possible transcript variants.
Context-Dependent Expression:
Evaluate cell type-specific expression patterns across different tissues and cell lines.
Assess whether expression changes under different physiological or experimental conditions.
Methodological Standardization:
Standardize sample preparation, fixation methods, and antibody dilutions across experiments.
Document and control variables that might influence detection, such as tissue handling, fixation duration, and antibody lot variations.
Quantification Approaches:
Implement multiple quantification methods (e.g., western blot, qPCR, immunofluorescence intensity) to cross-validate findings.
Use appropriate statistical analyses to determine the significance of observed differences.
Optimizing co-immunoprecipitation (Co-IP) experiments with KIFC3 antibodies requires attention to several critical factors:
Antibody Amount Optimization:
Lysis Buffer Selection:
For membrane-associated complexes: Use buffers containing mild detergents (e.g., 1% NP-40 or 0.5% Triton X-100) to preserve protein-protein interactions while solubilizing membrane components.
For cytoskeletal interactions: Consider specialized buffers that maintain microtubule integrity during extraction.
Pre-clearing Strategy:
Implement thorough pre-clearing with protein A/G beads to reduce non-specific binding.
Include appropriate IgG controls matched to the host species of the KIFC3 antibody.
Interaction Preservation Techniques:
Cross-linking: Consider mild cross-linking (0.5-1% formaldehyde) for transient interactions.
Temperature control: Maintain samples at 4°C throughout the procedure to preserve complex integrity.
Elution and Detection Optimization:
Mild elution: Use non-denaturing conditions for subsequent enzymatic assays.
Complete elution: Apply reducing SDS buffer for comprehensive western blot analysis.
Enhanced detection: Consider silver staining or highly sensitive western blot systems for low-abundance interacting partners.
KIFC3's role in NSCLC progression suggests several therapeutic development approaches:
Advanced techniques for investigating KIFC3's microtubule functions include:
Super-Resolution Microscopy:
Structured illumination microscopy (SIM) can resolve KIFC3 localization along microtubules with ~100 nm resolution.
Single-molecule localization microscopy techniques (STORM/PALM) can achieve even higher resolution (~20 nm) to precisely map KIFC3 distribution relative to microtubule structures.
Live-Cell Force Measurement:
Optical tweezers can measure the force generation capabilities of single KIFC3 motor proteins.
Magnetic tweezers allow long-duration force measurements to characterize KIFC3's processivity and load-bearing properties.
Microtubule Dynamics Visualization:
EB1-GFP tracking can reveal how KIFC3 influences microtubule plus-end dynamics.
Photoactivatable tubulin can help track microtubule turnover rates in the presence or absence of KIFC3.
In Vitro Reconstitution Systems:
Purified component systems using recombinant KIFC3 and fluorescently labeled microtubules can directly measure motor activity and microtubule bundling capabilities.
Microfluidic chambers allow controlled introduction of regulatory factors to assess their effects on KIFC3-microtubule interactions.
Cryo-Electron Microscopy:
High-resolution structural analysis of KIFC3-microtubule complexes can reveal the molecular basis of motor function and regulation.
Comparative analysis with other kinesin family members can highlight unique structural features of KIFC3.