KIF25, also known as Kinesin Family Member 25 or KNSL3, belongs to the kinesin-like protein family. These proteins function as microtubule-dependent molecular motors responsible for transporting organelles within cells and moving chromosomes during cell division . KIF25 specifically has been identified as a tetrameric kinesin that plays a crucial role in cellular processes related to centrosome dynamics and mitotic spindle formation .
KIF25 antibodies are immunological reagents designed to detect and bind specifically to the KIF25 protein in various experimental applications. These antibodies are available in multiple formats, including monoclonal and polyclonal variants, with different host species and targeting various epitopes of the KIF25 protein structure . The development of highly specific KIF25 antibodies has enabled researchers to investigate the expression, localization, and function of this important motor protein in cellular contexts.
The availability of diverse KIF25 antibodies has facilitated significant advances in understanding the protein's role in normal cellular function and potential implications in disease processes. These antibodies serve as essential tools for exploring the mechanistic details of KIF25's role in centrosome dynamics and spindle formation during cell division .
KIF25 antibodies are available in various formats to accommodate different experimental needs. The two primary types include:
Monoclonal antibodies: Derived from a single B-cell clone, these antibodies recognize a single epitope on the KIF25 protein, offering high specificity. For example, mouse monoclonal antibody clone 2C9 is available for KIF25 detection .
Polyclonal antibodies: Generated from multiple B-cell lineages, these antibodies bind to different epitopes on the KIF25 protein, potentially providing stronger signals through multiple binding sites. Rabbit polyclonal antibodies are commonly used for KIF25 detection .
The conjugation status of KIF25 antibodies varies according to experimental requirements:
Unconjugated antibodies: The most common format, suitable for western blotting, immunohistochemistry, and immunofluorescence applications .
HRP-conjugated antibodies: Directly linked to horseradish peroxidase enzyme for enhanced detection in western blotting and ELISA applications without requiring secondary antibodies .
FITC-conjugated antibodies: Labeled with fluorescein isothiocyanate for direct fluorescence detection in imaging applications .
KIF25 antibodies are designed to target different regions of the KIF25 protein:
N-terminal region antibodies: These target the amino-terminal portion of KIF25, such as antibodies recognizing the sequence TWTSGQLQREKQARPGSGAVLAFPDDKDLRVYGPAESQSAVFGDVCPLLT .
C-terminal region antibodies: These recognize the carboxy-terminal region, with some targeting the sequence VLGALLEHRGHAPYRNSRLTHLLQDCLGGDAKLLVILCISPSQRHLAQTL .
Mid-region antibodies: Some antibodies target specific amino acid sequences in the internal regions of KIF25, such as AA 57-106 or AA 302-351 .
The specificity of KIF25 antibodies varies, with some demonstrating cross-reactivity with KIF25 proteins from multiple species:
Some antibodies show cross-reactivity with KIF25 from species such as guinea pig, horse, dog, rat, and monkey, based on sequence homology .
Western blotting represents one of the most common applications for KIF25 antibodies, allowing researchers to detect and quantify KIF25 protein in cell or tissue lysates. The typical recommended dilution range for KIF25 antibodies in western blotting applications is 1:2000 to 2.5 µg/mL . This technique has been used to confirm the expression of KIF25 in various cell lines, including HeLa cells, as demonstrated in research examining KIF25's role in centrosome separation .
Western blotting with KIF25 antibodies has enabled researchers to validate siRNA knockdown experiments, confirming successful depletion of endogenous KIF25 protein . This application is critical for functional studies examining the consequences of KIF25 loss in cellular processes.
Immunohistochemistry (IHC) using KIF25 antibodies allows researchers to examine the tissue distribution and localization of KIF25 protein in fixed tissue sections. KIF25 antibodies are typically used at dilutions ranging from 1:150 to 1:500 for IHC applications .
Both paraffin-embedded sections (IHC-P) and frozen sections can be analyzed using KIF25 antibodies, with specific protocols optimized for each preparation method . These applications have contributed to understanding the expression patterns of KIF25 across different tissues and cell types.
Immunofluorescence (IF) using KIF25 antibodies has provided crucial insights into the subcellular localization of KIF25 protein. Dilutions of approximately 1:100 are typically recommended for IF applications . Advanced microscopy techniques including structured illumination microscopy (SIM) and super-resolution expansion microscopy have been employed with KIF25 antibodies to precisely localize the protein within cellular structures .
Research utilizing IF with KIF25 antibodies has revealed that KIF25 localizes to centrosomes during all stages of the cell cycle, forming a ring structure with radial projections . These findings have been instrumental in understanding KIF25's role in centrosome dynamics.
Enzyme-linked immunosorbent assay (ELISA) represents another application for KIF25 antibodies, particularly for quantitative analysis of KIF25 protein levels. Some KIF25 antibodies have been specifically validated for ELISA applications .
Additional applications of KIF25 antibodies include:
Immunoprecipitation for protein-protein interaction studies
Flow cytometry for analyzing KIF25 expression in cell populations
Chromatin immunoprecipitation for studying potential DNA interactions
Research using KIF25 antibodies has revealed that KIF25 functions as a bipolar, minus-end directed kinesin that suppresses centrosome separation . This represents one of the most significant discoveries regarding KIF25's cellular role.
Studies have demonstrated that KIF25 is required to prevent premature centrosome separation during interphase. When KIF25 is depleted using siRNA, the average distance between duplicated centrosomes in fixed cells increases significantly in both interphase (from 2.44 ± 0.21 μm to 3.54 ± 0.25 μm) and prophase (from 8.57 ± 0.76 μm to 11.39 ± 0.69 μm) . This phenotype can be rescued by expressing EGFP-KIF25, confirming the specificity of the effect.
The mechanism by which KIF25 prevents centrosome separation appears to involve its minus-end directed motor activity, which likely antagonizes the plus-end directed motor Eg5 that drives centrosome separation . This opposing force relationship helps maintain proper centrosome positioning during the cell cycle.
KIF25 also plays a role in mitotic spindle formation and orientation. Overexpression of KIF25 significantly delays bipolar spindle formation, while knockdown of KIF25 increases mitotic spindle length . These findings suggest that KIF25 contributes to the regulation of spindle dynamics during mitosis.
The connection between premature centrosome separation and spindle orientation defects suggests that KIF25-mediated centrosome positioning during interphase is crucial for subsequent proper spindle orientation during mitosis.
Immunofluorescence studies using KIF25 antibodies have revealed that expressed EGFP-KIF25 localizes to centrosomes during all stages of the cell cycle . Super-resolution expansion microscopy has shown that KIF25 forms a ring at the centrosome with radial projections.
The diameter of the EGFP-KIF25 ring was found to be greater than RFP-pericentrin in co-localized images collected with structured illumination microscopy . Additionally, microtubules were detected passing between duplicated interphase centrosomes that co-localized with EGFP-KIF25 rings, suggesting that KIF25 may directly mediate centrosomal linkage via crosslinked microtubules.
These structural insights provide important clues about how KIF25 functions at the molecular level to regulate centrosome dynamics and spindle formation.
Commercial KIF25 antibodies undergo various validation procedures to ensure specificity and reliability. These validation methods include:
Western blotting validation: Using cell lysates as positive controls to confirm the antibody detects a protein of the expected molecular weight (approximately 35 kDa for KIF25) .
Immunohistochemistry validation: Testing on tissue arrays containing multiple normal human tissues and cancer tissues to verify specific staining patterns .
Cross-reactivity testing: Evaluation against protein arrays containing human recombinant protein fragments to assess potential cross-reactivity with other proteins .
Peptide competition assays: Using specific blocking peptides to confirm antibody specificity .
Enhanced validation: Some suppliers offer additional validation approaches, such as recombinant expression validation, to further ensure antibody specificity .
The discovery that KIF25 plays a crucial role in centrosome dynamics and spindle orientation suggests potential therapeutic applications, particularly in cancer research. Since proper centrosome function and spindle orientation are essential for normal cell division, targeting KIF25 might represent a novel approach for treating cancers characterized by centrosomal abnormalities.
Future research could explore the development of small molecule inhibitors or other therapeutic approaches targeting KIF25, particularly in cancer types where centrosome amplification is prevalent. KIF25 antibodies will be essential tools in this research, both for studying the effects of potential therapeutic agents and for diagnostic applications.
Several aspects of KIF25 biology remain to be fully elucidated and represent important areas for future research:
The regulation of KIF25 expression and activity during the cell cycle
The complete interactome of KIF25, including binding partners that mediate its function
The role of KIF25 in development and differentiation processes
Potential involvement of KIF25 in diseases beyond cancer
The evolutionary conservation of KIF25 function across species
Addressing these questions will require continued improvement and diversification of KIF25 antibodies, including the development of antibodies against post-translationally modified forms of KIF25 and antibodies suitable for emerging technologies such as proximity labeling approaches.
Advances in antibody technology will likely enhance KIF25 research in several ways:
Super-resolution microscopy: Further refinement of super-resolution microscopy techniques coupled with highly specific KIF25 antibodies will allow even more detailed analysis of KIF25 localization and dynamics at centrosomes.
Single-cell proteomics: As single-cell proteomics technologies mature, KIF25 antibodies will play a role in examining the expression and function of KIF25 at the single-cell level, potentially revealing cell-to-cell variability in KIF25 function.
In vivo imaging: Development of antibody fragments or alternative binding proteins against KIF25 may enable in vivo imaging of KIF25 dynamics in model organisms.
Combinatorial approaches: Combining KIF25 antibodies with other molecular tools such as CRISPR-Cas9 gene editing will provide powerful approaches for dissecting KIF25 function in diverse cellular contexts.
KIF25 (Kinesin Family Member 25) is a tetrameric kinesin protein that functions as a bipolar, minus-end directed motor protein. Its primary biological role is to suppress centrosome separation during interphase by tethering centrosomes together through microtubule-dependent mechanisms . This function is critical for maintaining proper nuclear positioning and ensuring stable spindle orientation at the onset of mitosis.
KIF25 antibodies are valuable research tools because they enable:
Visualization of centrosome dynamics during cell cycle progression
Investigation of microtubule-dependent nuclear positioning
Analysis of spindle orientation mechanisms
Study of cell division aberrations in disease models
The importance of KIF25 can be understood through its knockdown effects: when KIF25 is depleted, cells exhibit premature centrosome separation during interphase (increasing from 2.44 ± 0.21 μm in control cells to 3.54 ± 0.25 μm in KIF25 knockdown cells) . This leads to microtubule-dependent nuclear translocation and disruption of cortical spindle positioning machinery.
Several types of KIF25 antibodies have been developed for research applications:
| Antibody Type | Host Species | Clonality | Target Region | Applications | Reactivity |
|---|---|---|---|---|---|
| Anti-KIF25 (C-Term) | Rabbit | Polyclonal | C-Terminal region | WB, IHC | Human, Dog, Guinea Pig, Horse, Rat |
| Anti-KIF25 (N-Term) | Rabbit | Polyclonal | N-Terminal region | WB, IHC, ELISA | Human |
| Anti-KIF25 (AA 1-262) | Rabbit | Polyclonal | Amino acids 1-262 | IHC, ELISA | Human |
| Anti-KIF25 (Full length) | Mouse | Monoclonal (2C9) | Full recombinant protein | WB, IHC, IF, IHC(p) | Human |
These antibodies offer researchers flexibility in experimental design based on specific requirements such as species cross-reactivity, epitope accessibility, and application compatibility .
Validating antibody specificity is critical for ensuring reliable research outcomes. For KIF25 antibodies, consider these methodological approaches:
Genetic knockout/knockdown controls:
Recombinant protein expression validation:
Express KIF25 with epitope tags in cells lacking endogenous KIF25
Verify co-localization of antibody signal with the tagged protein
Western blot analysis:
Confirm single band detection at the expected molecular weight (~220 kDa)
Compare multiple antibodies targeting different epitopes
Immunoprecipitation followed by mass spectrometry:
Verify that KIF25 is the primary protein pulled down by the antibody
Cross-reactivity assessment:
Test against closely related kinesin family proteins to ensure specificity
Example validation protocol from published research: "We found that the anti-Kif26b antibodies recognized protein bands at the predicted size of Kif26b (~220 kD) in wild-type MEF lysates but not in the Kif26b knockout or knockdown MEF lysates, confirming that our antibodies specifically recognize endogenous Kif26b" .
Optimizing KIF25 antibody protocols for high-quality immunofluorescence imaging of centrosome dynamics requires attention to several methodological details:
Fixation and Permeabilization:
For standard resolution: Fix cells in 1% PFA in -20°C methanol for 10 minutes, followed by blocking with 20% donkey serum for 1 hour
For super-resolution imaging: Fix cells in 37°C PBS containing 3.2% paraformaldehyde and 0.1% glutaraldehyde for 10 minutes, then reduce with 0.1% Sodium Borohydride before blocking with full-strength FBS
Antibody Dilutions and Incubation:
Primary antibody: Use at 1:100-1:200 dilution for monoclonal antibodies like 2C9
For IHC applications: Sigma's HPA046153 antibody performs optimally at 1:200-1:500 dilution
Extend primary antibody incubation to overnight at 4°C for improved signal-to-noise ratio
Co-labeling Strategy:
Pair KIF25 antibody with pericentrin/γ-tubulin antibody for precise centrosome localization
Include α-tubulin labeling to visualize microtubule networks
Image Acquisition Parameters:
Use confocal microscopy with z-stacks (0.2-0.3 μm steps) for accurate centrosome distance measurements
For live-cell imaging applications, consider using recombinant single-chain variable fragments derived from KIF25 antibodies
Quantification Method:
Measure inter-centrosomal distance using line scan analysis
Normal interphase centrosome separation typically ranges from 2.44 ± 0.21 μm in control cells
KIF25 antibodies can be valuable tools for studying disease-associated protein dynamics, particularly in conditions affecting cellular organization and division. For optimal results:
Disease-specific considerations:
Sensitivity enhancement strategies:
Signal amplification: Consider tyramide signal amplification (TSA) to detect low-abundance KIF25 in patient samples
Antibody combinations: Use multiple antibodies targeting different epitopes to enhance detection reliability
Quantitative analysis approaches:
Develop automated image analysis workflows for unbiased quantification
Establish clear thresholds for abnormal KIF25 distribution based on control samples
Experimental controls for disease studies:
Include age-matched controls when studying age-related conditions
Use isotype controls to distinguish specific binding from background
Incorporate genetic models with known KIF25 alterations as reference points
Tissue-specific protocol adaptations:
Recent advances in computational biology offer powerful approaches to analyze and predict antibody-antigen interactions. For KIF25 antibody research:
Binding mode analysis techniques:
Structural analysis of paratope-epitope interactions:
Machine learning applications:
Validation approach:
Experimental validation through site-directed mutagenesis of predicted key residues
Affinity measurements before and after computational optimization
Robust experimental controls are essential when using KIF25 antibodies in knockdown studies to ensure reliable interpretation of results:
Genetic controls:
Negative control siRNA: Use non-targeting control siRNA (e.g., Ambion negative control siRNA1) processed identically to KIF25 siRNA samples
Multiple siRNA constructs: Validate phenotypes with at least two independent siRNAs targeting different regions of the KIF25 gene to rule out off-target effects
Published example: "For Kif25 depletion, two siRNAs targeted to distinct regions of the Kif25 gene were used independently to assess Kif25 KD phenotypes (targeting sequences (1) 5′-AGUGGAAGUUUACAAUAAU-3′ and (2) 5′-CAGAGUGACUUAGGAAUUA-3′)"
Expression rescue controls:
Antibody validation in knockdown context:
Confirm reduction of KIF25 signal via:
Western blot (quantitative band intensity reduction)
Immunofluorescence (reduced staining intensity)
Include signal quantification relative to loading control or housekeeping protein
Timing considerations:
Combinatorial knockdowns:
Recombinant antibody technology offers opportunities to customize KIF25 antibodies for specialized applications:
Single-chain variable fragment (scFv) development:
Convert conventional KIF25 antibodies to scFv format for applications requiring smaller size
Methodology: Determine VH and VL sequences, connect with flexible linker, express in appropriate system
Similar approaches with kinesin antibodies have shown that scFvs can specifically recognize targets in cells
Species diversification:
Functional modifications:
Create blocking antibodies by targeting functional domains of KIF25
Develop conformation-specific antibodies to distinguish active vs. inactive states
Fusion protein strategies:
Generate fluorescent protein fusions for live-cell imaging
Create intrabody versions for tracking KIF25 in living cells
Engineering challenge: "While we have not yet succeeded in doing this with the H2 mAb, the scFv can be used as a genetically engineered probe to label endogenous antigens in the cell. This intrabody technique can be used to inhibit the function of antigens in the cell"
Expression system considerations:
HEK293/293FT cells provide cost-effective expression systems for recombinant antibodies
For some applications, purification may not be required: "The experiments described in this report were performed using the cultured medium because we found that purification was not required for western blotting or immunofluorescence microscopy"
| Problem | Possible Causes | Solutions |
|---|---|---|
| High background in immunofluorescence | Insufficient blocking, antibody concentration too high | Extend blocking time to 2+ hours, titrate antibody concentration, include 0.1-0.3% Triton X-100 in antibody diluent |
| Weak or no signal in Western blot | Protein degradation, inefficient transfer, low antibody sensitivity | Add protease inhibitors during lysis, optimize transfer conditions, increase antibody concentration or incubation time |
| Inconsistent staining patterns | Fixation variations, cell cycle differences | Standardize fixation protocol, synchronize cells, co-stain with cell cycle markers |
| Non-specific bands in Western blot | Cross-reactivity with related kinesins | Use KIF25 knockout controls, adjust antibody concentration, increase washing stringency |
| Poor reproducibility | Antibody batch variation, sample preparation inconsistencies | Use monoclonal antibodies when possible, standardize all protocol steps, include positive controls |
For KIF25-specific considerations:
If detecting centrosome separation dynamics, remember that nocodazole treatment promotes interphase centrosome separation that cannot be rescued by KIF25 overexpression
For enhanced specificity in Western blots, recommended antibody dilutions range from 1:2000 for monoclonal antibodies to 0.04-0.4 μg/mL for polyclonal antibodies
For Western Blotting:
Protein extraction: Use RIPA buffer with protease inhibitors
Protein loading: 20-40 μg total protein per lane
Antibody dilutions: 1:2000 for monoclonal (ABIN1499039) , 0.04-0.4 μg/mL for polyclonal (HPA046153)
Signal detection: Enhanced chemiluminescence with exposure time optimization
For Immunohistochemistry:
Antigen retrieval: Heat-induced epitope retrieval in citrate buffer (pH 6.0)
Antibody dilutions: 1:200-1:500 for HPA046153 , 1:150 for IHC-P applications of ABIN1499039
Detection system: DAB chromogen with hematoxylin counterstain
Controls: Include isotype control and known positive tissue sections
For Immunofluorescence:
Fixation: 1% PFA in -20°C methanol for standard applications
Specialized fixation for super-resolution: 3.2% paraformaldehyde with 0.1% glutaraldehyde
Antibody dilution: 1:100 for IF with monoclonal antibodies like 2C9
Nuclear counterstain: DAPI at 1:10,000 dilution
For Co-immunoprecipitation:
Lysis buffer: 50 mM Tris-HCl pH 7.5, 150 mM NaCl, 1% NP-40, 0.5% sodium deoxycholate with protease inhibitors
Antibody amount: 2-5 μg per 500 μg of total protein
Pre-clearing: Incubate lysate with protein A/G beads before adding antibody
Controls: Include IgG control and input sample
Recent advances in KIF25 antibody applications have expanded capabilities for studying centrosome dynamics:
Super-resolution imaging techniques:
Sample preparation: Specialized fixation with 3.2% paraformaldehyde and 0.1% glutaraldehyde
Co-labeling strategy: GFP-Kif25 and RFP-Pericentrin for protein localization combined with YL1/2 antibody against detyrosinated tubulin
This approach enables nanoscale resolution of KIF25 localization relative to centrosome structures
Live-cell imaging approaches:
Quantitative analysis methods:
Automated tracking of centrosome separation using machine learning algorithms
Statistical approaches to correlate centrosome separation and spindle orientation:
"In control cells the majority of the population display normal centrosome separation and form a properly oriented parallel spindle (grey bars, 66% of cells)"
Genetic modification strategies:
Computational modeling integration:
By combining these methodological advances, researchers can gain unprecedented insights into the role of KIF25 in regulating centrosome dynamics and nuclear positioning.
The role of KIF25 in maintaining proper nuclear positioning and spindle orientation suggests potential implications in disease contexts where these processes are dysregulated:
Cancer research applications:
Investigate KIF25 expression and localization in cancer cells with aberrant cell division
Develop KIF25 antibodies as potential biomarkers for tumors with centrosome amplification
Research question: Does KIF25 dysregulation contribute to chromosomal instability in cancer?
Neurodevelopmental disorders:
Methodological advances needed:
Development of phospho-specific KIF25 antibodies to monitor regulation
Creation of conformation-specific antibodies to distinguish active vs. inactive states
Integration with emerging spatial transcriptomics technologies
Therapeutic implications:
The field of antibody technology is rapidly evolving, offering exciting possibilities for KIF25 research:
Nanobody and single-domain antibody applications:
Development of camelid-derived nanobodies against KIF25 for super-resolution microscopy
Advantages: smaller size (~15 kDa) allows better tissue penetration and epitope access
Research findings show that single-domain antibodies employ different mechanisms to compensate for their smaller size when compared to conventional antibodies
Bispecific antibody development:
Create bispecific antibodies targeting KIF25 and other centrosome components
Applications: co-localization studies, protein-protein interaction analysis
Antibody engineering through computational approaches:
Apply machine learning models to predict improved KIF25 antibody variants
Use structural data from antibody-antigen complexes to inform design strategies
"Predicting a small, manageable set (~10) of high-likelihood protein variants from a single antibody sequence using the predictive capabilities of protein language models"
Intrabody approaches:
Develop genetically encoded antibody fragments for tracking endogenous KIF25 in living cells
Challenge: "The scFv is often not soluble in cytosol. Fusion with superfolder GFP (sfGFP) or the ultra-stable cytoplasmic antibody (STAND) could be used to increase the folding and solubility of scFv in the cytoplasm"
Integration with emerging technologies:
CRISPR-based proximity labeling combined with KIF25 antibodies
Spatial transcriptomics to correlate KIF25 protein localization with gene expression patterns
By leveraging these emerging antibody technologies, researchers can gain unprecedented insights into KIF25 biology and its role in cellular processes.