KIF15 antibodies are specialized immunological tools designed to detect and study Kinesin Family Member 15 (KIF15), a microtubule-associated motor protein critical for mitotic spindle assembly, chromosome segregation, and intracellular transport. These antibodies enable researchers to investigate KIF15's roles in cellular processes and its dysregulation in diseases such as cancer, making them indispensable for molecular biology and oncology research .
Glioma:
KIF15 is overexpressed in glioma tissues and correlates with advanced tumor stage, recurrence, and poor prognosis. Knockdown (KD) of KIF15 in U87MG and T98G glioblastoma cells suppresses proliferation, induces G0/G1 cell cycle arrest, and reduces metastasis by downregulating Erk/Akt pathways and EMT markers (e.g., N-cadherin, Snail) .
Breast Cancer:
KIF15 inhibition via Kif15-IN-1 reduces viability in MDA-MB231 (TNBC) and MCF7 (ER+) cells by 40–60%, induces apoptosis, and suppresses migration. Normal fibroblasts remain unaffected, highlighting its therapeutic specificity .
Pancreatic Cancer:
KIF15 interacts with PI3K-C2α to promote focal adhesion turnover and metastasis by recycling integrin β1 via Rab11A endosomes. Silencing KIF15 inhibits FAK/SRC phosphorylation and reduces tumor growth in xenograft models .
KIF15 drives cell cycle progression by upregulating Cyclin A2, Cyclin D1, and CDK2 while suppressing p21 and p18 .
In nasopharyngeal carcinoma (NPC), KIF15 overexpression correlates with poor survival (HR = 2.1, p < 0.001) and promotes proliferation via MEK/ERK signaling .
KIF15 is a plus-end directed kinesin-like motor enzyme involved in mitotic spindle dynamics and chromosome alignment, making it a key player in cell division processes . It functions as a molecular motor that travels along microtubules during cell division to help establish and maintain the bipolar spindle structure necessary for proper chromosome segregation . KIF15 has also been identified as having partial functional redundancy with KIF11, another mitotic kinesin, as KIF15 can replace essential functions of KIF11 in the creation of the bipolar spindle . This functional redundancy has significant implications for research targeting either protein, as cells may compensate for the loss of one through upregulation of the other.
KIF15 antibodies are valuable tools for multiple research applications including:
Western blot analysis to detect and quantify KIF15 protein expression
Immunohistochemistry to visualize KIF15 in tissue sections, as demonstrated in studies examining KIF15 expression in glioblastoma and prostate cancer tissues
Immunofluorescence for subcellular localization of KIF15, as shown in HeLa cells using specific antibodies like PACO57476
ELISA for quantitative detection of KIF15 in various sample types
These applications enable researchers to investigate KIF15 expression patterns, subcellular localization, and potential involvement in normal cellular processes and disease states.
To ensure the reliability of KIF15 antibody-based experiments, researchers should validate antibody specificity through multiple approaches:
Genetic controls: Using KIF15 knockdown or knockout cells as negative controls. For example, creating a KIF15 KO cell line via CRISPR/Cas9 technique as described in studies with S462 cells provides an excellent specificity control .
Peptide competition assays: Pre-incubating the antibody with the immunogen peptide to confirm signal elimination.
Multiple antibody validation: Using different antibodies targeting distinct KIF15 epitopes to confirm consistent staining patterns.
Western blot analysis: Confirming a single band of appropriate molecular weight (~160 kDa for human KIF15).
Cross-reactivity testing: Evaluating the antibody in multiple species to confirm expected reactivity patterns, as illustrated by the PACO57476 antibody which specifically detects human KIF15 .
Thorough validation is essential for obtaining reliable research data, especially when characterizing KIF15's role in complex biological processes like cancer development.
Optimizing KIF15 immunohistochemistry protocols requires consideration of tissue-specific factors:
For paraffin-embedded tissues, the following protocol has been successfully employed:
Deparaffinization of tissue sections
Antigen retrieval using citrate buffer
Blocking with 3% H₂O₂ for 10 minutes at room temperature
Overnight incubation with primary KIF15 antibody at 4°C (typically at 1:50 dilution for commercial antibodies)
Incubation with HRP-conjugated secondary antibody (e.g., goat anti-rabbit IgG H&L at 1:400) at 37°C for 1 hour
Visualization using appropriate detection systems and counterstaining
For cultured cells, such as in immunofluorescence applications, protocols typically involve:
Fixation in 4% formaldehyde
Permeabilization with 0.2% Triton X-100
Blocking in 10% normal goat serum
Overnight incubation with primary antibody at 4°C (recommended dilutions for PACO57476: 1:200-1:500)
Incubation with fluorophore-conjugated secondary antibody (e.g., Alexa Fluor 488-conjugated anti-rabbit IgG)
Optimization requires testing different antibody concentrations, incubation times, and antigen retrieval methods for each specific tissue type and fixation method.
Distinguishing between KIF15 expression and its functional activity requires complementary approaches:
For expression analysis:
Western blotting and immunohistochemistry quantify protein levels
RT-qPCR measures mRNA expression (shown to achieve ~80% reduction in KIF15 expression following siRNA treatment in cell lines like ST88-14)
Flow cytometry can analyze expression at the single-cell level
For activity assessment:
Functional assays evaluating mitotic spindle formation and integrity
Cell viability and proliferation assays following KIF15 modulation (as demonstrated in MPNST cell lines)
Cell cycle analysis by flow cytometry to detect G2/M arrest following KIF15 inhibition
Colony formation assays to assess long-term effects of KIF15 activity modulation
In vivo tumor growth experiments using xenograft models with modified KIF15 expression or activity
Combining these approaches provides a comprehensive understanding of both KIF15 expression and its functional significance in experimental systems.
KIF15 knockdown has demonstrated significant effects on cancer cell behavior across multiple studies:
In glioma cells, silencing of KIF15 inhibited cell proliferation and stemness, arrested cells in the G2 phase, and induced cell apoptosis . The inhibitory effect was also verified in vivo using xenograft models .
In MPNST cells, RNAi-mediated suppression of KIF15 showed cell line-dependent effects:
In ST88-14 cells, KIF15 knockdown significantly inhibited cell proliferation
In S462 cells, with less efficient KIF15 depletion (~60% reduction vs. ~80% in ST88-14), the effects were less pronounced
Methods for effective KIF15 knockdown include:
siRNA transfection: Using siRNA pools targeting KIF15, achieving 60-80% expression reduction in different cell lines
CRISPR/Cas9 gene editing: Creating complete KIF15 knockout cell lines for more definitive functional studies
Chemical inhibitors: Compounds like KIF15-IN-1 have been reported to suppress cancer cell growth
The choice of knockdown method depends on the research question, with transient siRNA approaches suitable for initial screening and CRISPR/Cas9 knockout preferred for in-depth mechanistic studies requiring complete protein elimination.
The molecular mechanisms underlying KIF15's role in cancer progression involve multiple pathways:
Cell cycle regulation: KIF15 knockdown arrests cancer cells in the G2/M phase, indicating its critical role in mitotic progression . This is likely due to disruption of mitotic spindle assembly and function.
Apoptosis regulation: KIF15 knockdown induces apoptosis in cancer cells, with Human Apoptosis Antibody Array showing upregulation of pro-apoptotic factors including CD40L, cytoC, DR6, and p21, while anti-apoptotic factors like IGF-I and Survivin were downregulated .
Cell stemness: KIF15 appears to regulate cancer stem cell-like properties, as its knockdown reduces stemness in glioma cells .
Compensatory mechanisms: KIF15 can functionally compensate for KIF11 inhibition in mitotic spindle formation, which has implications for therapeutic strategies targeting either kinesin .
Cell viability pathways: KIF15 deficiency sensitizes cancer cells to other therapeutic agents, as demonstrated in S462 cells where KIF15 knockout enhanced sensitivity to KIF11 inhibitors and combined KIF11/BRD4 inhibition .
These mechanisms collectively contribute to KIF15's role as a tumor promoter, making it an attractive therapeutic target in multiple cancer types.
Research indicates that targeting KIF15 in combination with other mitotic kinesins and cancer-related proteins offers promising therapeutic strategies:
KIF11 and KIF15 dual targeting: Studies have demonstrated that KIF15-deficient cells show increased sensitivity to KIF11 inhibitors like ispinesib and ARRY-520 . This approach exploits the partial functional redundancy between these kinesins, where KIF15 loss prevents compensatory mechanisms that might otherwise confer resistance to KIF11 inhibition.
Triple inhibition approaches: The combination of KIF11, KIF15, and BRD4 inhibition has shown synergistic antitumoral effects in MPNST cell lines . ARRY-520 (KIF11 inhibitor) combined with JQ1 (BRD4 inhibitor) was particularly effective in KIF15-deficient cells, where it:
Sequential targeting: Researchers might consider sequential inhibition approaches, first depleting KIF15 (via genetic approaches or specific inhibitors) before administering KIF11 inhibitors to maximize efficacy.
Cell-line specific optimization: Research indicates variable dependence on KIF15 across cancer cell lines, necessitating personalized approaches. For example, ST88-14 cells showed greater sensitivity to KIF15 knockdown than S462 cells .
Researchers should incorporate appropriate controls, including non-tumor cells (like human fibroblasts), to assess potential differential sensitivity between normal and cancer cells to these combination approaches .
Based on published protocols and manufacturer specifications, the following dilutions and conditions are recommended for KIF15 antibodies:
For immunohistochemistry:
Incubation: Overnight at 4°C
Secondary antibody: Goat anti-rabbit IgG H&L HRP-conjugated at 1:400 dilution
Incubation: 37°C for 1 hour
For immunofluorescence:
Primary KIF15 antibody (e.g., PACO57476): 1:200-1:500 dilution
Cell preparation: Fixation in 4% formaldehyde, permeabilization with 0.2% Triton X-100, blocking in 10% normal goat serum
Incubation: Overnight at 4°C
Secondary antibody: Alexa Fluor 488-conjugated anti-rabbit IgG
For ELISA:
These recommendations serve as starting points, and researchers should optimize conditions for their specific experimental systems, antibody lots, and sample types through dilution series and positive/negative controls.
Designing and interpreting KIF15 knockdown experiments requires careful consideration of several factors:
Experimental design recommendations:
Include multiple knockdown approaches:
Validate knockdown efficiency:
Include comprehensive functional readouts:
Essential controls:
Interpretation considerations:
Cell line-dependent effects: Different cell lines may show variable sensitivity to KIF15 depletion (e.g., ST88-14 vs. S462)
Partial vs. complete knockdown: The degree of knockdown achieved may significantly impact phenotypic outcomes
Compensatory mechanisms: Consider potential upregulation of functionally redundant proteins (e.g., KIF11)
Time-dependent effects: Distinguish between immediate and long-term consequences of KIF15 depletion
By following these design principles and interpretation guidelines, researchers can generate more reliable and translatable data on KIF15 function in cancer and other contexts.
To investigate the functional relationship between KIF15 and other mitotic kinesins, particularly KIF11, researchers can employ several effective techniques:
Sequential and simultaneous knockdown/inhibition:
Sensitivity assays in genetically modified backgrounds:
Rescue experiments:
Determine if KIF15 overexpression can rescue phenotypes caused by KIF11 inhibition
Identify the minimal domains of KIF15 required for functional compensation
Proximity-based interaction studies:
Proximity ligation assays or FRET to detect potential physical interactions
Co-immunoprecipitation to identify binding partners shared between KIF15 and other kinesins
Imaging approaches:
Live-cell imaging to visualize mitotic spindle dynamics in cells with various kinesin modifications
Super-resolution microscopy to examine co-localization patterns
Protein expression analysis:
Assess whether inhibition of one kinesin leads to compensatory upregulation of others
Study the relationship between expression levels and functional outcomes
These approaches have revealed important findings, such as KIF15's ability to replace essential functions of KIF11 in bipolar spindle formation and the enhanced sensitivity of KIF15-deficient cells to KIF11 inhibitors , highlighting the complex interplay between these motor proteins in mitotic regulation.
Quantification and interpretation of KIF15 expression in patient samples requires standardized approaches:
For immunohistochemistry quantification:
Score system development: Researchers commonly use semi-quantitative scoring methods that incorporate both staining intensity and percentage of positive cells.
Cut-off determination: The median of half-quantified expression scores from all samples can serve as a cut-off point to distinguish high versus low KIF15 expression cases .
Blinding procedures: Pathologists should evaluate slides in a blinded manner to minimize bias.
Statistical analysis: Apply appropriate statistical tests to compare expression levels between normal and tumor tissues or across different disease stages.
For clinical correlation analysis:
Use Mann-Whitney U-analysis to evaluate associations between KIF15 expression and clinicopathological parameters like tumor invasion depth .
Apply Spearman rank correlation analysis to confirm significant correlations .
Perform survival analysis using Kaplan-Meier curves and log-rank tests to assess relationships between KIF15 expression and patient outcomes .
Utilize multivariate analyses to determine whether KIF15 expression is an independent prognostic factor.
When interpreting results:
Consider tissue heterogeneity and the need for multiple sampling within tumors.
Acknowledge potential differences between protein and mRNA expression patterns.
Compare findings with publicly available datasets (e.g., TCGA) to validate observations across larger cohorts .
Interpret expression data in the context of other molecular markers and clinical parameters.
These approaches have revealed significant correlations between high KIF15 expression and advanced pathological features in multiple cancer types, supporting its potential value as a prognostic biomarker .
When evaluating the effects of KIF15 inhibition, several complementary functional assays provide comprehensive insights:
These assays collectively provide a comprehensive understanding of how KIF15 inhibition affects cancer cell biology across multiple functional domains.