KIF13A antibodies are immunoreagents targeting the KIF13A protein, a member of the kinesin-3 family. These antibodies are typically raised in rabbits (polyclonal) and validated for specificity across applications like Western blot (WB), immunohistochemistry (IHC), and immunoprecipitation (IP) . Key features include:
KIF13A antibodies are pivotal in studying:
Intracellular Transport: Tracking KIF13A’s role in recycling endosome dynamics and cargo delivery (e.g., mannose-6-phosphate receptors) .
Cell Division: Detecting KIF13A’s localization during cytokinesis .
Disease Mechanisms: Investigating overexpression in cancers (e.g., retinoblastoma, lung adenocarcinoma) and viral exploitation (e.g., influenza A) .
Western Blot: Detects KIF13A at ~202–238 kDa in HeLa, HepG2, and neuronal lysates .
Immunohistochemistry: Localizes KIF13A to Golgi, cytoplasm, and midbodies in human tissues .
Functional Studies: Knockdown experiments in hippocampal slices abolished long-term potentiation (LTP), linking KIF13A to synaptic plasticity .
RhoB Recycling: KIF13A antibodies revealed its role in recycling RhoB via Rab11-positive endosomes, promoting plasma membrane blebbing and amoeboid migration in cancer cells .
Oncogenic Fusion: A KIF13A-RET fusion protein was identified in lung adenocarcinoma, driving tumor progression .
Synaptic Plasticity: KIF13A depletion in hippocampal neurons abolished LTP, demonstrating its necessity for AMPA receptor trafficking .
Serotonin Receptors: Antibodies linked KIF13A dysfunction to anxiety phenotypes via serotonin receptor mislocalization .
Viral Transport: KIF13A facilitates influenza A ribonucleoprotein transport to the cell surface, enhancing viral replication .
Phosphorylation: MARK2 phosphorylates KIF13A at Ser1371, enhancing 14-3-3 binding and regulating polarity .
Dimerization: KIF13A forms homodimers or heterodimers with KIF13B, modulating recycling endosome biogenesis .
KIF13A (Kinesin Family Member 13A) is a plus end-directed microtubule-dependent motor protein involved in intracellular transport processes. In humans, the canonical protein has 1805 amino acid residues with a molecular mass of 202.3 kDa . KIF13A plays critical roles in mediating the transport of mannose-6-phosphate receptor (M6PR)-containing vesicles from the trans-Golgi network to the plasma membrane through direct interaction with the AP-1 complex . Additionally, it regulates endosomal sorting during melanosome biogenesis and participates in cytokinesis . The protein is widely expressed throughout the body, with particularly high expression levels in heart, brain, and skeletal muscle tissues .
KIF13A is predominantly localized in the Golgi apparatus and cytoplasm . The protein exists in up to five different isoforms, allowing for functional diversity across different cellular contexts . KIF13A is a member of the kinesin protein family, which consists of motor proteins that move along microtubule filaments using energy derived from ATP hydrolysis. The protein has several alternative names, including homolog of mouse KIF13A mannose-6-phosphate receptor transporter, kinesin-like protein RBKIN, and kinesin-like protein KIF13A . Evolutionarily, KIF13A is highly conserved, with orthologs identified in multiple species including mouse, rat, bovine, frog, zebrafish, chimpanzee, and chicken .
Several immunodetection methods are routinely employed for KIF13A research. The most frequently used applications include:
| Application | Typical Dilution | Common Sample Types | Detection Specificity |
|---|---|---|---|
| Western Blot (WB) | 1:500-1:1000 | Cell lysates, tissue extracts | Protein observed at 202 kDa |
| Immunohistochemistry (IHC) | 1:50-1:500 | Fixed tissue sections | Cellular and subcellular localization |
| Immunofluorescence (IF) | Variable | Fixed cells | Subcellular distribution |
| Immunoprecipitation (IP) | Variable | Cell lysates | Protein-protein interactions |
| ELISA | Variable | Purified proteins, serum | Quantitative detection |
For optimal immunohistochemistry results with KIF13A antibodies, antigen retrieval with TE buffer pH 9.0 is typically recommended, although citrate buffer pH 6.0 may alternatively be used . When performing Western blot analysis, HepG2 cells have been confirmed as a positive control for KIF13A expression .
When selecting a KIF13A antibody, researchers should consider multiple factors beyond basic reactivity. First, determine the specific application requirements (WB, IHC, IF, IP, ELISA) as antibodies often perform differently across applications. For instance, some KIF13A antibodies are optimized for multiple applications like WB and IP, while others may be specific to a single application like ELISA .
Second, consider the species reactivity required for your experiment. Available KIF13A antibodies demonstrate varying reactivity profiles, with some specific to human samples only, and others showing cross-reactivity with mouse and rat . For cross-species studies, antibodies with documented reactivity across multiple species are essential.
Third, evaluate antibody validation data through published literature and manufacturer validation. Antibodies with citations in peer-reviewed publications provide increased confidence in performance. For example, some KIF13A antibodies have associated publications with validation figures demonstrating specificity and reproducibility .
Proper validation of KIF13A antibodies is critical for reliable experimental results. A comprehensive validation process should include:
Positive and negative controls: Use cell lines with known KIF13A expression (e.g., HepG2) as positive controls . For negative controls, consider using KIF13A knockout/knockdown samples or primary antibody omission.
Antibody titration: Test a range of antibody dilutions to determine optimal concentration. For KIF13A antibodies, typical Western blot dilutions range from 1:500-1:1000, while IHC applications may require 1:50-1:500 dilutions .
Specificity verification: Confirm a single band of appropriate molecular weight (202 kDa) in Western blot applications . Multiple bands might indicate degradation products, isoforms, or non-specific binding.
Cross-reactivity assessment: If working with non-human samples, verify cross-reactivity with your species of interest, as KIF13A antibodies vary in their species reactivity profiles .
Reproducibility testing: Perform replicate experiments to ensure consistent results across different lots and conditions.
Subcellular localization concordance: In immunofluorescence or IHC applications, verify that the localization pattern matches expected Golgi and cytoplasmic distribution .
For optimal Western blot detection of KIF13A, researchers should follow these methodological guidelines:
Sample preparation: Prepare cell or tissue lysates using RIPA buffer supplemented with protease inhibitors. Given KIF13A's high molecular weight (202 kDa), complete protein extraction and denaturation are critical.
Gel electrophoresis: Use low percentage (6-8%) SDS-PAGE gels to effectively resolve high molecular weight proteins like KIF13A. Consider gradient gels (4-15%) for better separation.
Transfer conditions: For large proteins like KIF13A, employ wet transfer methods with extended transfer times (overnight at 30V, 4°C) to ensure complete transfer of high molecular weight proteins.
Antibody incubation:
Block membranes with 5% non-fat milk or BSA in TBST for 1 hour at room temperature
Incubate with primary KIF13A antibody at 1:500-1:1000 dilution overnight at 4°C
Wash extensively with TBST (at least 3×10 minutes)
Incubate with appropriate secondary antibody (typically 1:5000-1:10000) for 1 hour at room temperature
Detection: Use enhanced chemiluminescence detection systems with extended exposure times, as high molecular weight proteins may require longer exposures for optimal visualization.
Controls: Include positive control samples (HepG2 cell lysate) and molecular weight markers spanning the 200 kDa range .
For effective KIF13A immunohistochemical detection in tissue samples, consider these methodological recommendations:
Tissue fixation: Use 4% paraformaldehyde or 10% neutral buffered formalin with fixation time optimized for tissue thickness (typically 24-48 hours for standard biopsies).
Antigen retrieval: KIF13A detection specifically requires heat-induced epitope retrieval using TE buffer pH 9.0 for optimal results, though citrate buffer pH 6.0 may serve as an alternative . Perform retrieval by:
Heating sections in retrieval buffer to 95-100°C for 15-20 minutes
Allowing gradual cooling to room temperature for 20 minutes
Washing thoroughly with PBS before blocking
Blocking and antibody application:
Detection system: Use polymer-based detection systems rather than avidin-biotin methods for enhanced sensitivity and lower background when detecting KIF13A.
Controls: Include positive control tissues (human liver cancer tissue has been validated) and negative controls (primary antibody omission and isotype controls).
To investigate KIF13A's protein-protein interactions, particularly its well-documented interaction with the AP-1 complex , researchers can employ multiple complementary approaches:
Co-immunoprecipitation (Co-IP):
Proximity ligation assay (PLA):
This technique allows visualization of protein interactions in situ
Use primary antibodies against KIF13A and suspected interaction partners
Follow with PLA-specific secondary antibodies and amplification
Visualization reveals punctate signals where proteins are in close proximity (<40 nm)
FRET/BRET analysis:
Create fluorescent protein fusions with KIF13A and potential partners
Measure energy transfer between fluorophores as indicator of protein proximity
Particularly useful for studying dynamic interactions in live cells
Yeast two-hybrid screening:
For discovery of novel KIF13A interaction partners
Use KIF13A domains as bait to screen for interacting proteins
Validate findings with co-IP or other methods listed above
Mass spectrometry-based interactomics:
Perform IP with KIF13A antibodies followed by mass spectrometry
Compare results to control IPs to identify specific interactors
Classify interactions based on abundance and specificity scores
Live-cell imaging of KIF13A-mediated transport requires specialized approaches that preserve protein functionality while allowing visualization:
Antibody fragments for live-cell applications:
Convert conventional KIF13A antibodies into Fab fragments
Conjugate with cell-permeable peptides and fluorophores
Microinject or otherwise introduce into living cells
Monitor dynamics using spinning disk or lattice light-sheet microscopy
Complementary fluorescent protein approaches:
Generate KIF13A-FP fusion constructs (ensuring functionality is preserved)
Transfect into cells of interest at low expression levels
Pair with labeled cargo markers (e.g., fluorescently-tagged M6PR)
Perform dual-color time-lapse imaging to track co-transport
Quantitative analysis parameters:
Measure velocity (typically 0.5-1 μm/sec for kinesin-mediated transport)
Determine run length (distance traveled before dissociation)
Calculate pause frequency and duration
Assess directionality and persistence of movement
Perturbation approaches:
Combine imaging with acute inhibition (e.g., photoinactivation)
Apply microtubule-disrupting drugs to confirm microtubule dependence
Use ATP depletion to verify energy requirements
Implement optogenetic tools for spatiotemporal control of KIF13A activity
Despite its widespread expression, KIF13A can be difficult to detect in some tissues due to low abundance. Advanced strategies to overcome this limitation include:
Signal amplification techniques:
Tyramide signal amplification (TSA) can enhance immunohistochemical detection by 10-100 fold
For Western blot, consider enhanced chemiluminescence substrates with extended sensitivity
Use biotin-streptavidin amplification systems with multiple layers
Sample enrichment approaches:
Perform subcellular fractionation to concentrate Golgi and cytoplasmic fractions where KIF13A localizes
Implement immunoprecipitation before detection to concentrate the target protein
Consider proximity labeling methods (BioID, APEX) to identify KIF13A-associated proteomes even when KIF13A itself is difficult to detect directly
Alternative detection technologies:
Digital droplet PCR for extremely sensitive mRNA quantification
Single-molecule detection methods
Mass spectrometry with targeted multiple reaction monitoring (MRM)
Tissue-specific optimization table:
| Tissue Type | Recommended Fixation | Optimal Antigen Retrieval | Suggested Antibody Dilution | Signal Amplification Method |
|---|---|---|---|---|
| Brain | 4% PFA, 24h | TE buffer pH 9.0, 30 min | 1:50 | TSA |
| Heart | 10% NBF, 48h | Pressure cooker, citrate pH 6.0 | 1:100 | Polymer detection |
| Skeletal muscle | Zinc fixative | EDTA pH 8.0, 40 min | 1:50 | Biotin-streptavidin |
| Low-expressing tissues | PAXgene fixation | Combined heat and enzyme digestion | 1:25 | Multiple layer amplification |
Distinguishing between the five reported isoforms of KIF13A presents a significant challenge requiring specialized approaches:
Isoform-specific antibody selection:
Carefully evaluate epitope information for available antibodies
Select antibodies targeting regions with isoform-specific sequences
Validate specificity using overexpression systems with individual isoforms
PCR-based isoform detection:
Design primer pairs spanning exon junctions specific to each isoform
Perform RT-PCR or qPCR with isoform-specific primers
Validate amplicon identity through sequencing
Consider digital PCR for absolute quantification of isoform ratios
Mass spectrometry approaches:
Analyze tryptic digests for isoform-specific peptides
Implement parallel reaction monitoring for targeted detection
Quantify isoform-specific peptides for relative abundance determination
Functional differentiation strategies:
Generate isoform-specific knockdowns using siRNAs targeting unique regions
Perform rescue experiments with individual isoforms
Assess functional readouts (e.g., M6PR transport efficiency, melanosome distribution)
Combined detection workflow:
Initial screening with pan-KIF13A antibodies by Western blot
High-resolution gel electrophoresis to separate closely related isoforms
Confirmation using isoform-specific detection methods
Correlation with functional assays to determine isoform-specific roles
Non-specific binding presents a common challenge when working with KIF13A antibodies. Systematic troubleshooting approaches include:
Blocking optimization:
Test different blocking agents (BSA, normal serum, commercial blockers)
Extend blocking time (2-3 hours or overnight)
Include detergents (0.1-0.3% Triton X-100 or Tween-20) to reduce hydrophobic interactions
Consider adding 1-5% non-fat dry milk to both blocking and antibody diluent solutions
Antibody dilution optimization:
Washing protocol enhancement:
Increase number of washes (minimum 5-6 washes)
Extend wash duration (15-20 minutes per wash)
Use gentle agitation during washing
Consider adding 0.5M NaCl to wash buffer to disrupt low-affinity interactions
Pre-adsorption controls:
Pre-incubate antibody with immunizing peptide (if available)
Compare staining patterns with and without pre-adsorption
Specific signals should disappear after pre-adsorption
Validation across multiple detection methods:
Confirm findings using at least two independent detection techniques
Compare patterns between fresh and fixed samples where possible
Correlate protein detection with mRNA expression data
The high molecular weight of KIF13A (202 kDa) presents specific technical challenges in Western blot applications. To effectively detect the full-length protein:
Sample preparation optimization:
Use strong lysis buffers (RIPA with 0.5% sodium deoxycholate)
Add protease inhibitor cocktails immediately after lysis
Minimize freeze-thaw cycles to prevent degradation
Consider adding phosphatase inhibitors to preserve post-translational modifications
Sonicate briefly to shear DNA without damaging proteins
Gel electrophoresis adjustments:
Use gradient gels (4-15%) or low percentage (6%) gels
Reduce voltage during electrophoresis (60-80V)
Extend running time to improve resolution of high molecular weight proteins
Include molecular weight markers that extend beyond 200 kDa
Pre-chill running buffer to minimize heat-induced protein degradation
Transfer optimization for high-molecular-weight proteins:
Implement wet transfer systems rather than semi-dry
Add 0.05-0.1% SDS to transfer buffer to improve elution of large proteins
Reduce methanol concentration to 10% to prevent gel hardening
Extend transfer time (overnight at 30V, 4°C)
Verify transfer efficiency using reversible staining of membranes
Detection system considerations:
Select high-sensitivity chemiluminescent substrates
Extend exposure times (may require 5-15 minutes versus standard 1-5 minutes)
Consider fluorescent Western blot systems for better linear dynamic range
Use fresh antibody dilutions to maximize binding efficiency
Positive controls and validation:
The kinesin superfamily contains multiple related proteins with structural similarities that can lead to antibody cross-reactivity. To ensure KIF13A-specific detection:
Antibody selection criteria:
Choose antibodies raised against unique regions of KIF13A rather than conserved motor domains
Evaluate manufacturer cross-reactivity testing data
Select antibodies validated in knockout/knockdown systems
Consider monoclonal antibodies for increased specificity
Validation through genetic approaches:
Perform siRNA/shRNA knockdown of KIF13A and verify signal reduction
Implement CRISPR/Cas9 knockout systems as definitive controls
Conduct overexpression studies with tagged KIF13A constructs
Compare detection patterns between these systems
Bioinformatic cross-reactivity analysis:
Compare epitope sequences against related kinesin family members
Predict potential cross-reactive proteins through sequence homology
Design experiments to specifically rule out detected cross-reactivity
Comparative detection strategies:
Use multiple antibodies targeting different KIF13A epitopes
Compare detection patterns for consistency
Implement peptide competition assays with KIF13A-specific peptides
Include parallel detection of closely related kinesins (e.g., KIF13B) for comparison
Functional validation approaches:
Given KIF13A's high expression in brain tissue and its crucial role in intracellular transport, it presents a significant target for neurodegenerative disease research:
Pathological sample analysis:
Compare KIF13A expression and localization patterns between healthy and diseased brain tissues
Evaluate KIF13A function in various neurodegenerative conditions including Alzheimer's, Parkinson's, and ALS
Assess potential co-localization with disease-specific aggregates or markers
Determine if KIF13A transport is impaired in conditions with known axonal transport defects
Mechanistic investigation approaches:
Examine KIF13A-mediated transport of specific neuronal cargoes
Assess potential interactions between KIF13A and disease-associated proteins
Evaluate the impact of disease-relevant stressors on KIF13A function
Implement live-cell imaging in primary neurons to visualize transport dynamics
Therapeutic targeting strategies:
Screen for compounds that modulate KIF13A activity or expression
Evaluate the potential of KIF13A as a biomarker for disease progression
Assess whether restoration of compromised KIF13A function improves neuronal health
Develop targeted delivery systems utilizing KIF13A transport mechanisms
Model system applications:
Implement KIF13A antibodies in iPSC-derived neurons from patients
Utilize animal models with KIF13A mutations or manipulations
Correlate findings across species to establish evolutionary conservation of mechanisms
Develop high-throughput screening systems for KIF13A functional assessment
Post-translational modifications (PTMs) likely play critical roles in regulating KIF13A function, but studying these modifications presents unique challenges:
PTM-specific detection approaches:
Develop and validate phospho-specific antibodies for key regulatory sites
Implement enrichment strategies (e.g., phosphopeptide enrichment) before analysis
Use mass spectrometry-based approaches for unbiased PTM mapping
Apply proximity labeling methods to identify enzymes regulating KIF13A modifications
Functional correlation strategies:
Generate phosphomimetic and phospho-deficient mutants of key sites
Assess impact on KIF13A localization, motor activity, and cargo binding
Implement live-cell imaging with biosensors to monitor dynamic modifications
Correlate modifications with specific cellular states or stimuli
Technical challenges and solutions table:
| Challenge | Solution Approach | Methodological Considerations |
|---|---|---|
| Low abundance of modified KIF13A | Affinity purification before analysis | May require large sample input |
| Transient nature of some PTMs | Chemical crosslinking or rapid lysis | Time-sensitive protocols |
| Multiple modification sites | Targeted mass spectrometry | Requires preliminary site identification |
| Functional redundancy | Combinatorial mutation analysis | Complex experimental design |
| Context-dependent modifications | Cell-type specific analysis | Requires multiple model systems |
Integrative analysis workflow:
Initial PTM identification through mass spectrometry
Validation with site-specific antibodies when available
Functional characterization through mutagenesis
Correlation with cellular contexts and stimuli
Computational modeling of modification impacts on protein structure and function
Advanced multiplexed imaging allows simultaneous visualization of KIF13A alongside multiple markers to understand its contextual relationships in tissues:
Multiplexed immunofluorescence strategies:
Sequential immunostaining with antibody stripping between rounds
Spectral unmixing approaches for simultaneous multi-color imaging
Implement tyramide signal amplification for detecting low-abundance targets
Use DNA-barcoded antibodies for highly multiplexed detection (CODEX, Imaging Mass Cytometry)
Antibody panel design considerations:
Include KIF13A antibodies validated for tissue applications
Combine with markers for subcellular compartments (Golgi, endosomes, plasma membrane)
Add antibodies for known interaction partners (AP-1 complex components)
Include cell-type specific markers for contextual analysis
Image analysis pipeline optimization:
Implement machine learning approaches for signal segmentation
Develop quantitative colocalization metrics
Use spatial statistics to analyze distribution patterns
Apply trajectory analysis for transport studies in tissue contexts
Validation controls for multiplexed approaches:
Single-stain controls for spectral unmixing
Isotype controls matched to each primary antibody
Absorption controls to verify signal specificity
Replication across multiple samples and conditions
Advanced tissue preparation considerations:
Optimize fixation to preserve KIF13A epitopes while enabling multiplexed detection
Consider clearing techniques (CLARITY, iDISCO) for thick tissue sections
Implement expanded microscopy for improved resolution of transport complexes
Use precision-cut tissue slices for live imaging applications in near-native environments