The PAT2B11AT clone is a mouse anti-human monoclonal antibody (IgG2b/k) generated by immunizing BALB/c mice with a recombinant human KCTD15 protein fragment (amino acids 1–159) . It is designed for research applications including ELISA, Western blot (WB), flow cytometry, and immunocytochemistry/immunofluorescence (ICC/IF) . KCTD15 is implicated in neural crest development, obesity-linked pathways, and cancer biology, making this antibody a critical tool for studying these processes .
Colorectal Cancer (CRC): KCTD15 acts as a tumor suppressor by stabilizing p53 via HDAC1 inhibition. Overexpression reduces CRC cell viability and induces apoptosis .
Acute Myeloid Leukemia (AML): KCTD15 is upregulated in AML cell lines (e.g., HL-60, NB4) compared to normal peripheral blood cells, suggesting diagnostic potential .
NF-κB Signaling: KCTD15 enhances IKK-β activity, promoting NF-κB pathway activation, which is critical in leukemia progression .
KCTD15 regulates neural crest induction by suppressing Wnt/β-catenin signaling during embryogenesis .
Biomarker Potential: Differential KCTD15 expression in CRC and AML highlights its utility in disease monitoring .
Mechanistic Studies: The antibody enables exploration of KCTD15’s role in HDAC1-p53 interactions and NF-κB activation .
Obesity Research: KCTD15 is linked to BMI regulation, with SNPs near its locus associated with obesity .
PAT2B11AT.
Anti-human KCTD15 mAb, is derived from hybridization of mouse F0 myeloma cells with spleen cells from BALB/c mice immunized with a recombinant human KCTD15 protein 1-159 amino acids purified from E. coli.
Mouse IgG2b heavy chain and k light chain.
KCTD15 (Potassium Channel Tetramerization Domain Containing 15) is a protein encoded by the KCTD15 gene in humans. Also known as BTB/POZ domain-containing protein KCTD15, it serves several important biological functions:
During embryonic development, it is involved in neural crest formation
It inhibits AP2 transcriptional activity by interacting with its activation domain
It has been identified as a genetic locus linked to higher than normal BMI in humans, along with genes such as GNPDA2, MTCH2, FTO, and TMEM18
Recent research has revealed its significant overexpression in certain breast cancer subtypes, particularly in HER2+ breast cancer patients
The PAT2B11AT antibody specifically recognizes an epitope within amino acids 1-159 of the human KCTD15 protein. This monoclonal antibody was generated by immunizing BALB/c mice with a recombinant human KCTD15 protein fragment (amino acids 1-159) purified from E. coli . The antibody was derived from the hybridization of mouse F0 myeloma cells with spleen cells from the immunized mice, resulting in a mouse IgG2b heavy chain and kappa light chain antibody that specifically targets the N-terminal region of KCTD15 .
The KCTD15 PAT2B11AT antibody has been validated for multiple research applications, making it versatile for KCTD15 studies:
Researchers should note that while the antibody has been tested for specificity and reactivity in these applications, optimal conditions may vary between laboratories. Each investigation should include appropriate titration of the reagent to obtain optimal results for the specific experimental system being used .
To maintain optimal efficacy of the KCTD15 PAT2B11AT antibody, researchers should follow these storage and handling guidelines:
Avoid freeze-thaw cycles, which can degrade antibody quality and functionality
The antibody has a shelf life of approximately 12 months at -20°C and 1 month at 4°C
The antibody is formulated at 1mg/ml in PBS (pH 7.4) with 10% Glycerol and 0.02% Sodium Azide
Proper storage conditions are critical for maintaining antibody performance across multiple experiments and ensuring reproducible results.
To validate the specificity of the KCTD15 PAT2B11AT antibody in your experimental system, consider implementing these methodological approaches:
Positive control testing: Utilize cell lines known to express high levels of KCTD15, such as SKBR3 breast cancer cells
Negative control testing: Include cells with low KCTD15 expression (e.g., normal breast cell lines like MCF10A) for comparison
Western blot validation: Confirm detection of a single band at the expected molecular weight for KCTD15
Knockdown validation: Test the antibody on wild-type vs. KCTD15-silenced cells using siRNA or CRISPR methods
Blocking experiments: Pre-incubate the antibody with the immunizing peptide (amino acids 1-159) to confirm specific binding
Cross-reactivity assessment: Test against closely related proteins to ensure specificity to KCTD15
Recent research has revealed critical insights into KCTD15's role in breast cancer, particularly in HER2+ subtypes:
Studies demonstrate significant KCTD15 overexpression in Luminal A, Luminal B, and especially HER2+ breast cancer patients compared to healthy controls
The SKBR3 cell line (a model system for HER2+ breast cancer) shows remarkably higher KCTD15 expression compared to other breast cancer cell lines and normal breast epithelial cells (MCF10A)
KCTD15 silencing in SKBR3 cells using CRISPR/Cas9 technology significantly attenuates cell proliferation and cell cycle progression
KCTD15 silencing also sensitizes HER2+ breast cancer cells to the cytotoxic agent doxorubicin, suggesting a potential role in resistance mechanisms
These findings collectively indicate that KCTD15 may play an active role in HER2+ breast cancer carcinogenesis, suggesting its potential as both a biomarker and therapeutic target for this aggressive breast cancer subtype .
To investigate KCTD15's impact on cell proliferation, particularly in cancer models, researchers should consider these methodological approaches:
CRISPR/Cas9 gene editing: This approach has been successfully used to silence KCTD15 in breast cancer cell lines, allowing for precise study of its impact on proliferation
Cell proliferation assays: Methods such as MTT/XTT, BrdU incorporation, or real-time cell analysis can quantify changes in proliferation after KCTD15 manipulation
Cell cycle analysis: Flow cytometry-based analysis of cell cycle distribution following KCTD15 silencing can reveal its impact on cell cycle progression
Colony formation assays: These assess the long-term effects of KCTD15 modulation on cell growth and survival
Protein interaction studies: Co-immunoprecipitation to identify KCTD15 binding partners involved in proliferation pathways
Transcriptional profiling: RNA-seq analysis of cells with modulated KCTD15 expression to identify affected pathways
In experimental design, it's essential to include appropriate controls and consider cell line-specific contexts, as KCTD15 expression varies significantly across different breast cancer subtypes .
Optimizing KCTD15 detection across various breast cancer cell lines requires consideration of several methodological factors:
Expression level variation: Be aware that KCTD15 expression varies significantly across breast cancer subtypes:
Antibody titration: The recommended starting dilution for PAT2B11AT antibody is 1:250-500 for most applications , but optimal concentration should be determined for each cell line
Detection method selection:
Signal amplification: Consider signal enhancement techniques for low-expressing cell lines
Quantification approach: Use digital image analysis for immunofluorescence or densitometry for Western blots to enable objective comparison across cell lines
When conducting KCTD15 expression studies in breast cancer research, implement these essential controls:
Positive Controls:
SKBR3 cells (HER2+ breast cancer cell line) which consistently show high KCTD15 expression
Luminal A and B breast cancer cell lines that demonstrate significant KCTD15 expression
Brain or hypothalamus tissue samples, where KCTD15 is naturally highly expressed
HER2+ breast cancer tissue sections for immunohistochemistry studies
Negative Controls:
MCF10A normal breast epithelial cells (showing very low KCTD15 expression)
KCTD15 knockout/knockdown cell lines generated via CRISPR-Cas9 or siRNA technology
Isotype control antibodies matched to the primary antibody class and species
Secondary antibody-only controls to assess background staining
Internal tissue controls with mild KCTD15 expression (normal ductal-lining epithelium) for IHC studies
Methodological Controls:
Include gradient expression controls when possible (cell lines with known low, medium, and high expression)
For quantitative analyses, include calibration standards of known KCTD15 concentrations
Technical replicates to assess method reproducibility
Biological replicates to account for natural variation
Based on published research, several techniques have proven effective for KCTD15 gene silencing in breast cancer research:
CRISPR/Cas9 Gene Editing:
Most effective for complete and stable KCTD15 knockout
Successfully employed in SKBR3 cells (HER2+ breast cancer model)
Enables generation of stable cell lines for long-term studies
Provides precise genomic modifications with minimal off-target effects when designed properly
RNA Interference (RNAi):
siRNA: Effective for transient KCTD15 knockdown (3-7 days)
shRNA: Suitable for longer-term knockdown when delivered via lentiviral vectors
Provides flexibility in targeting different regions of KCTD15 mRNA
Methodological Considerations:
Include scrambled or non-targeting controls with similar GC content
Validate knockdown efficiency using both qRT-PCR (mRNA level) and Western blot (protein level)
For breast cancer studies, optimize transfection/transduction protocols specific to the cell line being used
Consider rescue experiments with KCTD15 overexpression to confirm specificity of observed effects
The choice of silencing technique should be based on experimental duration, desired knockdown efficiency, and specific research questions.
KCTD15 has been identified as an inhibitor of AP2 transcriptional activity through interaction with its activation domain . To study this interaction and its functional consequences, researchers can employ these methodological approaches:
Molecular Interaction Studies:
Co-immunoprecipitation (Co-IP): Use anti-KCTD15 antibodies to pull down protein complexes and probe for AP2
Proximity ligation assay (PLA): Visualize and quantify KCTD15-AP2 interactions in situ
Yeast two-hybrid or mammalian two-hybrid assays: Map specific interaction domains
GST pull-down assays with recombinant proteins: Determine if interaction is direct or requires cofactors
Functional Analysis:
Luciferase reporter assays: Measure AP2 transcriptional activity in the presence/absence of KCTD15
ChIP-seq: Analyze AP2 binding to target genes with and without KCTD15 expression
RNA-seq: Identify genes differentially regulated by AP2 when KCTD15 is modulated
KCTD15 domain mutation analysis: Identify which domains are critical for AP2 inhibition
Physiological Relevance:
Correlate KCTD15-AP2 interaction with cancer phenotypes (proliferation, migration, etc.)
Investigate how this interaction may contribute to breast cancer development and progression
This interaction may be particularly relevant in HER2+ breast cancer, where KCTD15 is highly expressed , and understanding it could reveal potential therapeutic targets in the AP2 signaling pathway.
Recent research has revealed an important relationship between KCTD15 expression and chemotherapy sensitivity in breast cancer:
KCTD15 silencing in SKBR3 cells (HER2+ breast cancer model) sensitizes them to the cytotoxic agent doxorubicin, suggesting KCTD15 may contribute to chemoresistance mechanisms
This relationship appears particularly significant in HER2+ breast cancers, where KCTD15 is highly overexpressed
Methodological Approaches to Study This Relationship:
Drug sensitivity assays: Compare IC50 values of chemotherapeutic agents in KCTD15-expressing vs. KCTD15-silenced cells
Apoptosis assays: Measure changes in apoptotic response to chemotherapy following KCTD15 manipulation
Mechanistic investigations: Explore whether KCTD15 affects drug efflux pumps, DNA damage repair pathways, or anti-apoptotic mechanisms
Patient-derived xenografts: Assess correlation between KCTD15 expression and treatment response in clinically relevant models
Clinical sample analysis: Retrospectively analyze KCTD15 expression in relation to treatment outcomes in breast cancer patients
Understanding this relationship could lead to the development of KCTD15 as a predictive biomarker for chemotherapy response or as a target for combination therapies aimed at overcoming resistance in HER2+ breast cancer .
Several quantitative methods can be employed to assess KCTD15 expression in clinical samples, each with specific advantages for research applications:
Immunohistochemistry (IHC):
Allows visualization of KCTD15 expression in tissue context
Can be performed on formalin-fixed paraffin-embedded (FFPE) tissues from archives
Recommended scoring system: Assess staining intensity (0=negative, 1=weak, 2=moderate, 3=strong) and percentage of positive cells
Compare with normal ductal-lining epithelium as an internal control
Advantages: Preserves tissue architecture, enables retrospective studies
Limitations: Semi-quantitative, subject to interpreter variability
Quantitative Real-Time PCR (qRT-PCR):
Measures KCTD15 mRNA expression levels
Use validated reference genes appropriate for breast tissue
Calculate relative expression using the 2^-ΔΔCt method
Advantages: Highly sensitive, specific, good dynamic range
Limitations: Requires fresh or properly preserved tissue, does not assess protein expression
Western Blot:
Quantifies KCTD15 protein expression
Normalize to appropriate loading controls
Advantages: Confirms protein size, semi-quantitative
Limitations: Requires significant amount of fresh/frozen tissue
Flow Cytometry:
Allows single-cell quantification of KCTD15 expression
Recommended for analysis of disaggregated tumor samples
Advantages: Provides detailed expression distribution, can be combined with other markers
Limitations: Requires viable cells, labor-intensive sample preparation
For clinical applications, IHC is most commonly used due to practicality with FFPE samples, while research applications may benefit from combining multiple approaches for comprehensive analysis.
Researchers may encounter several technical challenges when using KCTD15 antibodies for immunohistochemistry:
Epitope Accessibility:
Formalin fixation can mask the KCTD15 epitope
Solution: Optimize antigen retrieval methods (heat-induced epitope retrieval with appropriate buffers)
Test different retrieval times and temperatures to determine optimal conditions
Expression Heterogeneity:
KCTD15 expression varies significantly across breast cancer subtypes
Solution: Include known positive controls (HER2+ tissues) and negative controls with each staining batch
Use hyperplastic areas with mild positivity in normal ductal-lining epithelium as internal controls
Background Staining:
Non-specific binding can complicate interpretation
Solution: Optimize blocking procedures (5% normal serum, 1% BSA)
Titrate primary antibody concentration (starting at 1:250 dilution)
Consider mouse-on-mouse blocking if using mouse antibodies on mouse tissues
Signal Optimization:
Weak signal in low-expressing samples
Solution: Consider signal amplification systems
Optimize incubation times and temperatures
Use fresher tissue sections when possible
Reproducibility:
Batch-to-batch variability in staining intensity
Solution: Include standard reference tissues in each run
Standardize all protocol steps (fixation time, section thickness, staining conditions)
Quantification Standardization:
Consistent scoring across different samples and observers
Solution: Establish clear scoring criteria based on comparison with internal controls
Consider digital image analysis for more objective quantification
Research has revealed significant variations in KCTD15 expression across breast cancer molecular subtypes, with important biological and clinical implications:
Expression Pattern Across Subtypes:
HER2+ subtype: Shows the highest level of KCTD15 overexpression among breast cancer subtypes
Luminal A and Luminal B subtypes: Demonstrate significant but generally lower KCTD15 overexpression compared to HER2+
Normal breast tissue: Exhibits very mild positivity in normal ductal-lining epithelium
Research and Clinical Implications:
Diagnostic potential:
Therapeutic relevance:
Biological insights:
Methodological considerations:
When studying KCTD15, researchers should select appropriate cell line models based on the subtype of interest
Subtype-specific expression patterns should be considered when designing experiments and interpreting results
Validation across multiple cell lines representing different subtypes is recommended
Clone PAT2B11AT is a mouse monoclonal antibody specifically designed to target the KCTD15 protein in humans . Monoclonal antibodies are laboratory-produced molecules engineered to serve as substitute antibodies that can restore, enhance, or mimic the immune system’s attack on cells.
The PAT2B11AT clone is widely used in various research applications, including:
Mouse anti-human antibodies are antibodies produced in mice that are specific to human antigens. These antibodies are commonly used in research and diagnostic applications due to their high specificity and affinity for human proteins . They are essential tools in various immunological assays and have significantly contributed to advancements in biomedical research.
One potential issue with using mouse anti-human antibodies is the Human Anti-Mouse Antibody (HAMA) response. This response occurs when the human immune system recognizes the mouse antibodies as foreign and mounts an immune response against them. The HAMA response can range from mild reactions, such as rashes, to severe and life-threatening conditions like kidney failure .