KCTD15 Antibody

Potassium channel tetramerisation domain containing 15, Mouse Anti Human
Shipped with Ice Packs
In Stock

Product Specs

Introduction
KCTD15 protein is encoded in humans by the KCTD15 gene. It is highly expressed in the brain, particularly the hypothalamus. Research has identified the KCTD15 potassium channel as a genetic locus associated with a higher body mass index (BMI) in humans. This association is observed alongside other genes like GNPDA2, MTCH2, FTO, and TMEM18. Studies examining single nucleotide polymorphisms (SNPs) in individuals with and without diabetes revealed a strong link between FTO and obesity. Notably, MTCH2 and GNPDA2 were also significantly associated with elevated BMI levels.
Formulation
The antibody is supplied at a concentration of 1mg/ml in a solution of phosphate-buffered saline (PBS) at pH 7.4 with 0.1% sodium azide as a preservative.
Storage Procedures
For short-term storage (up to 1 month), keep at 4°C. For extended storage, store at -20°C. Avoid repeated freeze-thaw cycles.
Stability / Shelf Life
The antibody has a shelf life of 12 months when stored at -20°C and 1 month when stored at 4°C.
Applications
The KCTD15 antibody has undergone rigorous testing using ELISA, Western blot, and Immunofluorescence analysis to ensure its specificity and reactivity. However, optimal working dilutions should be determined empirically for each application. For Western blot and Immunofluorescence, a dilution range of 1:250 to 1:500 is recommended, with a starting dilution of 1:250.
Synonyms
BTB/POZ domain-containing protein KCTD15, Potassium channel tetramerisation domain containing 15, KCTD15, MGC2628, MGC25497.
Purification Method
KCTD15 antibody was purified from mouse ascitic fluids by protein-G affinity chromatography.
Type
Mouse Anti Human Monoclonal.
Clone
PAT4C3AT.
Immunogen
Anti-human KCTD15 mAb, is derived from hybridization of mouse F0 myeloma cells with spleen cells from BALB/c mice immunized with recombinant human KCTD15 amino acids 1-234 purified from E. coli.
Ig Subclass
Mouse IgG3 heavy chain and L light chain.

Q&A

What is KCTD15 and what are its primary biological functions?

KCTD15 (Potassium Channel Tetramerization Domain Containing 15) is a member of the KCTD family of proteins involved in various biological processes. It functions primarily during embryonic development by regulating neural crest formation . At the molecular level, KCTD15 inhibits AP2 transcriptional activity by interacting with its activation domain . The protein is expressed at particularly high levels in the brain and hypothalamus .

KCTD15 has also been identified as a genetic locus associated with higher than normal body mass index (BMI) in humans, alongside genes such as GNPDA2, MTCH2, FTO, and TMEM18 . Recent research has expanded our understanding of KCTD15's role in disease contexts, particularly in cancer biology where it may function as an anti-tumor factor in colorectal cancer (CRC) .

When conducting Western blot analysis, researchers should anticipate potential variations in band size depending on the cell or tissue type being studied. For instance, KCTD15 has been successfully detected in various samples including mouse brain tissue, C6 cells, HEK-293 cells, mouse lung tissue, HeLa cells, and human leukemia cell lines .

How can KCTD15 protein expression be accurately measured in peripheral blood samples for leukemia research?

Flow cytometry represents an optimal approach for measuring KCTD15 expression in peripheral blood samples, particularly in leukemia research. A validated protocol involves:

  • Sample preparation: Use the PerFix Expose kit for intracellular detection of KCTD15 .

  • Multidimensional staining strategy: Co-stain samples with:

    • CD45 (pan-leukocyte marker)

    • CD14 (monocyte-specific marker)

    • Anti-KCTD15 antibody (intracellular staining)

  • Gating strategy:

    • First, select single-cell events on an FSC-H versus FSC-A dot plot

    • Identify fixed single live cells on an FSC-A versus SSC-A dot plot

    • Use CD45 versus SSC dot plot to identify lymphocytes, monocytes, and granulocytes based on differential CD45 expression and light SSC properties

    • Confirm monocyte populations using CD14 versus SSC dot plot

This approach enables quantitative assessment of KCTD15 expression across leukocyte populations. Research has demonstrated that KCTD15 is expressed at significantly higher levels in the myeloid compartment compared to lymphoid cells, with granulocytes exhibiting the highest expression levels .

In acute myeloid leukemia (AML) studies, researchers have successfully labeled HL-60 cells with Violet-Cell Tracer dye and mixed them into normal whole blood to simulate AML samples, allowing simultaneous measurement of KCTD15 expression in both leukemic and normal cells .

What methodological approaches should be used to investigate KCTD15's role in cancer pathophysiology?

Investigating KCTD15's role in cancer pathophysiology requires multiple complementary approaches:

  • Expression analysis in clinical samples:

    • Compare KCTD15 expression between paired tumor and normal adjacent tissues using qRT-PCR and Western blot

    • Perform immunohistochemistry (IHC) staining on tissue microarrays to assess protein levels

    • Correlate expression with clinicopathological parameters (see table below)

Clinical ParameterAssociation with KCTD15 ExpressionStatistical Significance
AgeNo significant impactNot significant
GenderNo significant impactNot significant
Tumor sizeNo significant impactNot significant
Tumor invasionNo significant impactNot significant
DifferentiationNo significant impactNot significant
CRC stagesLower in stages I-IISignificant
  • Functional studies using gene modulation:

    • Employ tetracycline-inducible expression vectors to control KCTD15 expression

    • Verify altered expression using qRT-PCR and Western blot

    • Measure effects on cellular phenotypes using:

      • MTT assay for cell viability

      • Flow cytometry with Annexin V-FITC/PI double staining for apoptosis assessment

      • Western blot for apoptosis-related proteins (cleaved caspase 3, cleaved caspase 9, p53)

  • In vivo studies:

    • Generate xenograft models using KCTD15-modulated cell lines

    • Assess tumor growth dynamics

    • Perform TUNEL staining to detect apoptosis in tumor tissues

    • Use IHC to evaluate p53 expression changes

Research using these approaches has revealed that KCTD15 acts as an anti-tumor factor in colorectal cancer, with overexpression reducing cell viability and inducing apoptosis through regulation of p53 .

How should researchers address contradictory data regarding KCTD15 expression patterns across different cancer types?

Research on KCTD15 expression across different cancer types has yielded contradictory results that require careful methodological consideration:

  • Standardize detection methods:

    • When comparing KCTD15 expression across cancer types, consistent antibody selection is crucial. Use antibodies validated for the specific application and tissue type .

    • Employ multiple detection methods (qRT-PCR, Western blot, IHC) to confirm expression patterns.

    • Include appropriate positive and negative controls in each experiment.

  • Consider tissue-specific context:

    • KCTD15 appears downregulated in colorectal cancer tissues compared to para-cancerous tissues .

    • In contrast, it is upregulated in acute myeloid leukemia cell lines (K562, NB4, HL-60) compared to normal peripheral blood mononuclear cells .

    • These contradictions suggest tissue-specific roles that should be explicitly acknowledged.

  • Perform systematic bioinformatic analyses:

    • Utilize publicly available transcriptomic datasets like the Microarray Innovations in Leukemia (MILE) dataset to validate experimental findings .

    • Compare expression data across multiple cancer databases to identify tissue-specific patterns.

    • Consider single-cell RNA sequencing to address cellular heterogeneity within tumor samples.

  • Validate functional implications:

    • Beyond expression levels, assess functional consequences through:

      • Cell viability assays following KCTD15 overexpression or knockdown

      • Apoptosis measurements

      • Analysis of pathway-specific markers (e.g., p53, caspases)

These contradictory findings suggest that KCTD15 may have context-dependent functions, acting as a tumor suppressor in some tissues while potentially promoting malignancy in others. The differences highlight the importance of tissue-specific analysis rather than generalizing findings across cancer types.

What are the optimal conditions for KCTD15 antibody use in immunohistochemistry?

Optimizing KCTD15 antibody use in immunohistochemistry (IHC) requires attention to several critical parameters:

  • Sample preparation:

    • For paraffin-embedded tissues, optimal antigen retrieval is essential

    • Use TE buffer pH 9.0 for antigen retrieval, though citrate buffer pH 6.0 may serve as an alternative

    • For human smooth muscle tissue, successful staining has been achieved with paraffin-embedded samples

  • Antibody selection and dilution:

    • For rabbit polyclonal antibodies:

      • ab254929: Use at 1/500 dilution for paraffin-embedded human smooth muscle tissue

      • 20128-1-AP: Use at 1:50-1:500 dilution, with optimal dilution determined through titration

    • For mouse monoclonal antibodies:

      • AT4C3 (ATGA0178): Follow manufacturer recommendations for specific tissue types

  • Detection system:

    • Secondary antibody selection should match the host species of the primary antibody

    • For low abundance proteins, consider signal amplification systems

    • Include appropriate positive controls (mouse spleen tissue has been validated)

  • Troubleshooting high background:

    • Increase blocking time or concentration

    • Optimize antibody dilution through titration

    • Reduce incubation time of the primary antibody

    • Thoroughly wash between steps

A representative IHC protocol would include: deparaffinization, antigen retrieval with TE buffer pH 9.0, blocking with serum-free protein block, primary KCTD15 antibody incubation at optimized dilution (1:50-1:500) overnight at 4°C, appropriate detection system application, and counterstaining.

How can researchers optimize Western blot protocols for detecting endogenous KCTD15 in different cell types?

Optimizing Western blot protocols for KCTD15 detection requires careful consideration of several factors:

  • Sample preparation:

    • For cell lines (such as RT4, U-251MG, MCF7, HEK-293, C6):

      • Use RIPA buffer supplemented with protease inhibitors

      • Sonicate briefly to ensure complete lysis and DNA shearing

      • Centrifuge at 12,000g for 15 minutes at 4°C to remove cellular debris

    • For tissue samples (brain, lung, spleen):

      • Homogenize in RIPA buffer with protease inhibitors

      • Process similarly to cell lysates

  • Protein loading and separation:

    • Load appropriate amounts (30-50 μg) of total protein per lane

    • Use 10-12% SDS-PAGE gels for optimal separation

    • Include molecular weight markers to identify the expected 26-32 kDa band

  • Transfer and blocking:

    • Transfer to PVDF or nitrocellulose membranes

    • Block with 5% non-fat milk or BSA in TBST for 1 hour at room temperature

  • Antibody incubation:

    • Primary antibody dilutions:

      • ab254929: 0.4 μg/ml for human cell lines

      • 20128-1-AP: 1:500-1:1000 for mouse tissues and various cell lines

    • Incubate overnight at 4°C

    • Wash thoroughly with TBST (3-5 times, 5 minutes each)

    • Use HRP-conjugated secondary antibodies appropriate for the primary antibody host species

  • Detection and troubleshooting:

    • Use enhanced chemiluminescence (ECL) detection systems

    • If signal is weak:

      • Increase protein loading

      • Increase primary antibody concentration

      • Extend primary antibody incubation time

      • Consider using more sensitive ECL substrates

    • If multiple bands appear:

      • Optimize blocking conditions

      • Increase wash times and volumes

      • Consider using KCTD15 knockout/knockdown controls to identify specific bands

Different cell types show varying KCTD15 expression levels, with notable expression in RT4 (human urinary bladder cancer), U-251MG, HEK-293, C6, and mouse brain and lung tissues .

What strategies can be employed to validate KCTD15 antibody specificity for research applications?

Validating KCTD15 antibody specificity is crucial for generating reliable research data. Several complementary approaches should be employed:

  • Genetic validation approaches:

    • KCTD15 knockdown/knockout controls:

      • Use siRNA or CRISPR-Cas9 to generate KCTD15-depleted cells

      • Compare antibody signals between wild-type and KCTD15-depleted samples

      • Absence or significant reduction of signal in knockout/knockdown samples confirms specificity

    • Overexpression controls:

      • Use tetracycline-inducible expression vectors for controlled KCTD15 overexpression

      • Verify increased signal intensity corresponding with overexpression levels

  • Immunological validation approaches:

    • Peptide competition assays:

      • Pre-incubate antibody with excess immunizing peptide

      • Specific signals should be blocked by the competing peptide

    • Multiple antibody validation:

      • Test several antibodies targeting different KCTD15 epitopes

      • Consistent detection patterns across antibodies support specificity

      • Compare results from different antibodies like ab254929 (targeting aa 1-50) , 20128-1-AP (targeting recombinant full protein) , and ATGA0178 (targeting recombinant human KCTD15 aa 1-234)

  • Application-specific validation:

    • For Western blot:

      • Verify that observed molecular weight matches predicted size (approximately 26-32 kDa)

      • Include positive control lysates from tissues with known KCTD15 expression (brain, spleen)

    • For IHC/ICC:

      • Compare staining patterns with known KCTD15 localization data

      • Include negative control sections (primary antibody omitted)

    • For flow cytometry:

      • Use fluorescence minus one (FMO) controls to establish gating boundaries

      • Validate with spike-in experiments using cells with known KCTD15 expression levels

  • Mass spectrometry validation:

    • For immunoprecipitation experiments, confirm pulled-down proteins by mass spectrometry

    • Identify KCTD15 peptides in immunoprecipitated samples as ultimate confirmation of specificity

Implementing these validation strategies establishes confidence in antibody specificity and ensures experimental reproducibility across different research applications of KCTD15 antibodies.

How can KCTD15 expression patterns be utilized as potential biomarkers in cancer diagnostics?

KCTD15 shows promising potential as a biomarker in cancer diagnostics, with distinct expression patterns observed across different malignancies:

  • Colorectal cancer (CRC):

    • KCTD15 is significantly downregulated in CRC tissues compared to adjacent normal tissues

    • Lower KCTD15 expression correlates with early CRC stages (I-II)

    • IHC staining protocols can reliably detect these differences in clinical specimens

    Potential diagnostic application: KCTD15 downregulation could serve as an early-stage CRC biomarker when used in combination with established markers.

  • Acute myeloid leukemia (AML):

    • KCTD15 is upregulated in myeloid leukemia cell lines (K562, NB4, HL-60)

    • Flow cytometry analysis can detect increased KCTD15 expression in AML samples

    • Microarray Innovations in Leukemia (MILE) dataset confirms upregulation at the mRNA level

    Potential diagnostic application: Flow cytometric detection of elevated KCTD15 could complement existing AML diagnostic panels.

  • Implementation of KCTD15 in diagnostic protocols:

    • For solid tumors:

      • IHC staining of KCTD15 in biopsy samples using optimized protocols (1:50-1:500 dilution, TE buffer pH 9.0 for antigen retrieval)

      • Scoring based on staining intensity and percentage of positive cells

    • For hematological malignancies:

      • Multiparametric flow cytometry with CD45, CD14, and intracellular KCTD15 staining

      • Gating strategy to identify leukemic populations based on KCTD15 expression levels

  • Advantages and limitations:

    • Advantages:

      • Tissue-specific expression patterns provide context-relevant information

      • Compatible with standard clinical laboratory techniques (IHC, flow cytometry)

      • Can be integrated with existing biomarker panels

    • Limitations:

      • Contradictory expression patterns across cancer types necessitate tissue-specific interpretation

      • Further validation in larger patient cohorts required

      • Optimal cut-off values for diagnostic applications not yet established

The diagnostic utility of KCTD15 is strengthened by its differential expression between normal and malignant tissues and its correlation with certain clinical parameters, making it a promising candidate for integration into cancer diagnostic panels.

What is the relationship between KCTD15 and apoptotic pathways in cancer cells?

Research has revealed important connections between KCTD15 and apoptotic pathways in cancer cells, particularly in colorectal cancer:

  • Direct evidence of KCTD15's pro-apoptotic function:

    • Flow cytometry with Annexin V-FITC/PI double staining demonstrates that KCTD15 overexpression significantly increases the percentage of apoptotic cells in HCT116 and LoVo colorectal cancer cell lines

    • TUNEL staining of tumor tissues confirms that KCTD15 overexpression induces apoptosis in vivo

  • Molecular mechanisms linking KCTD15 to apoptotic pathways:

    • KCTD15 overexpression increases expression of key apoptosis-related biomarkers:

      • Cleaved caspase 3: Executioner caspase in apoptotic cascade

      • Cleaved caspase 9: Initiator caspase in intrinsic apoptotic pathway

      • p53: Master regulator of apoptosis and cell cycle arrest

    • IHC staining confirms increased p53 expression in KCTD15-overexpressing tumor tissues

  • Experimental approaches to investigate KCTD15-mediated apoptosis:

    • Gene modulation:

      • Use tetracycline-inducible expression systems for controlled KCTD15 overexpression

      • Verify expression changes by qRT-PCR and Western blot

    • Apoptosis detection methods:

      • Flow cytometry with Annexin V-FITC/PI staining

      • Immunoblotting for cleaved caspases and other apoptotic markers

      • TUNEL assay for in situ detection of apoptotic cells in tissue sections

    • Pathway analysis:

      • Investigate relationships with intrinsic vs. extrinsic apoptotic pathways

      • Examine mitochondrial membrane potential changes

      • Evaluate Bcl-2 family protein balance

  • Therapeutic implications:

    • KCTD15's pro-apoptotic function suggests potential as a therapeutic target

    • Strategies to upregulate or restore KCTD15 expression might sensitize colorectal cancer cells to apoptosis

    • Combination with established apoptosis-inducing agents could yield synergistic effects

The relationship between KCTD15 and apoptotic pathways appears to be mediated through p53 and the intrinsic apoptotic pathway, as evidenced by increased cleaved caspase 9 levels. This mechanism explains why KCTD15 overexpression reduces cancer cell viability and suggests potential therapeutic applications targeting this pathway.

How do biological contexts influence KCTD15 protein expression and function across different tissue types?

KCTD15 exhibits context-dependent expression and function across different biological systems, highlighting the importance of tissue-specific analysis:

  • Tissue-specific expression patterns:

    • Neural tissues:

      • High expression in brain and hypothalamus

      • Involved in neural crest formation during embryonic development

    • Hematopoietic system:

      • Differential expression across leukocyte populations

      • Highest expression in granulocytes, followed by monocytes, with lowest levels in lymphocytes

      • Upregulated in myeloid leukemia cell lines (K562, NB4, HL-60)

    • Gastrointestinal tissues:

      • Downregulated in colorectal cancer compared to adjacent normal tissues

  • Developmental context influences:

    • During embryogenesis:

      • Regulates neural crest domain formation

      • Inhibits neural crest induction by repressing Wnt/β-catenin signaling pathway

      • Interacts with AP2 transcriptional activity

    • In adult tissues:

      • Associated with metabolic regulation

      • Genetic loci linked to body mass index in genome-wide association studies

  • Disease-specific functional variations:

    • In colorectal cancer:

      • Acts as an anti-tumor factor

      • Overexpression decreases cell viability

      • Induces apoptosis through p53 pathway activation

    • In acute myeloid leukemia:

      • Upregulated compared to normal peripheral blood cells

      • Potential role in leukemic transformation of myeloid cells

  • Methodological approaches to study context-dependent functions:

    • Tissue-specific gene modulation:

      • Conditional knockout/knockin models

      • Tissue-specific promoters for expression control

    • Multi-omics integration:

      • Correlate protein expression with transcriptomic and proteomic data

      • Identify tissue-specific interaction partners through co-immunoprecipitation followed by mass spectrometry

    • Functional assays tailored to tissue context:

      • Neural differentiation assays for developmental studies

      • Metabolic assays for hypothalamic function

      • Cancer-specific assays (proliferation, migration, apoptosis) for oncology research

The context-dependent nature of KCTD15 function underscores the importance of studying this protein within appropriate tissue and disease models rather than generalizing findings across biological systems. This variability may explain the seemingly contradictory roles in different cancers and highlights the complexity of KCTD15 biology.

What are the common challenges in detecting KCTD15 by Western blot and how can they be overcome?

Researchers frequently encounter several challenges when detecting KCTD15 by Western blot. Here are the common issues and evidence-based solutions:

  • Discrepancy between predicted and observed molecular weight:

    • Challenge: KCTD15's calculated molecular weight is 31-32 kDa, but it often appears at approximately 26 kDa on Western blots .

    • Solutions:

      • Use positive control lysates from validated sources (mouse brain tissue, C6 cells, HEK-293 cells)

      • Run gradient gels (4-20%) to better resolve proteins in the 20-35 kDa range

      • Include ladder markers with close spacing in the 20-35 kDa region

      • Validate specific band using KCTD15 knockdown/knockout controls

  • Weak or absent signal:

    • Challenge: KCTD15 may be expressed at low levels in some cell types.

    • Solutions:

      • Optimize protein loading (35-50 μg recommended for cell lines such as RT4, U-251MG)

      • Increase antibody concentration (try 0.4-1.0 μg/ml for ab254929)

      • Extend primary antibody incubation to overnight at 4°C

      • Use high-sensitivity ECL detection systems

      • Consider concentrating proteins through immunoprecipitation before Western blot

  • Multiple or non-specific bands:

    • Challenge: Some antibodies may detect non-specific proteins.

    • Solutions:

      • Compare results using different antibodies targeting distinct epitopes

      • Optimize blocking conditions (try 5% BSA instead of milk for phospho-specific detection)

      • Increase washing duration and volume (5 washes x 5 minutes with TBST)

      • Use freshly prepared lysates to minimize protein degradation

      • Include KCTD15 knockdown controls to identify specific bands

  • Tissue-specific detection variations:

    • Challenge: KCTD15 expression and detection vary across tissue types.

    • Solutions:

      • Select antibodies validated for your specific tissue type

      • Optimize extraction buffers based on tissue type (add deoxycholate for membrane-rich tissues)

      • Consider tissue-specific positive controls:

        • For brain studies: mouse brain tissue

        • For cancer studies: validated cell lines (RT4, U-251MG)

        • For leukocyte studies: isolated peripheral blood mononuclear cells

  • Quantification challenges:

    • Challenge: Accurately quantifying KCTD15 expression differences.

    • Solutions:

      • Use appropriate housekeeping controls (β-actin, GAPDH)

      • Run technical triplicates

      • Use fluorescence-based Western blot systems for wider dynamic range

      • Perform densitometry using software that can correct for background

A systematically optimized Western blot protocol for KCTD15 detection should include: complete cell lysis with RIPA buffer containing protease inhibitors, 35-50 μg protein loading, 10-12% SDS-PAGE gels, overnight primary antibody incubation at 1:500-1:1000 dilution, extensive washing, and appropriate controls to validate specific bands.

How can researchers accurately compare KCTD15 expression across different experimental conditions using flow cytometry?

Flow cytometry offers powerful quantitative analysis of KCTD15 expression, but requires careful methodology to ensure accurate comparisons across experimental conditions:

  • Standardized sample preparation:

    • Use the PerFix Expose kit for consistent intracellular detection of KCTD15

    • Process all experimental conditions in parallel to minimize batch effects

    • Maintain consistent cell numbers across samples (typically 1×10^6 cells per sample)

    • Use fixation/permeabilization buffers from the same lot

  • Optimized staining protocol:

    • Multiparameter panel design:

      • Include CD45 (pan-leukocyte marker) and CD14 (monocyte marker) for leukocyte subpopulation identification

      • Select fluorochromes to minimize spectral overlap

      • Include a viability dye to exclude dead cells

    • Controls for accurate quantification:

      • Unstained controls for autofluorescence assessment

      • Single-color controls for compensation setup

      • Fluorescence Minus One (FMO) controls to set positive/negative boundaries

      • Isotype controls to assess non-specific binding

      • Biological controls: known high and low KCTD15 expressors (e.g., granulocytes vs. lymphocytes)

  • Standardized data acquisition:

    • Daily instrument quality control with calibration beads

    • Consistent PMT voltages across experiments

    • Collect sufficient events (minimum 10,000 events per population of interest)

    • Run samples in the same order relative to controls

  • Robust gating strategy:

    • Implement the validated sequential gating approach:

      • Single-cell selection (FSC-H vs. FSC-A)

      • Live cell identification (FSC-A vs. SSC-A)

      • Leukocyte subpopulation gating (CD45 vs. SSC-A: lymphocytes, monocytes, granulocytes)

      • Confirmation of monocyte population (CD14 vs. SSC-A)

  • Quantification methods:

    • For comparison across conditions, use:

      • Median Fluorescence Intensity (MFI) rather than mean (less sensitive to outliers)

      • Calculate fold change relative to control samples

      • Consider normalized MFI (sample MFI divided by FMO control MFI)

    • For experimental models such as AML detection:

      • Label cells of interest with tracking dyes (e.g., Violet-Cell Tracer for HL-60 cells)

      • Mix labeled cells with control samples to enable direct side-by-side comparison

  • Data visualization and statistical analysis:

    • Present data in standardized formats:

      • Overlay histograms for visual comparison

      • Box plots for population statistics

      • Bar graphs with error bars showing fold change

    • Apply appropriate statistical tests:

      • Paired t-tests for matched samples

      • ANOVA with post-hoc tests for multiple condition comparisons

      • Non-parametric tests if normality assumptions are violated

What are the most promising avenues for exploring KCTD15's potential as a therapeutic target in cancer?

Given the emerging evidence of KCTD15's role in cancer biology, several promising research avenues warrant exploration:

  • Targeting KCTD15 in colorectal cancer therapy:

    • Restore KCTD15 expression in CRC:

      • Develop small molecules that upregulate KCTD15 expression

      • Explore epigenetic modifiers that may reverse KCTD15 silencing

      • Design gene therapy approaches to reintroduce KCTD15 in tumors

    • Exploit KCTD15-induced apoptotic pathways:

      • Identify the precise mechanism linking KCTD15 to p53, caspase 3, and caspase 9 activation

      • Screen for compounds that mimic KCTD15's pro-apoptotic effects

      • Develop combination strategies with established chemotherapeutics

  • Understanding tissue-specific effects in hematological malignancies:

    • Clarify KCTD15's role in acute myeloid leukemia:

      • Determine whether KCTD15 upregulation in AML is a driver or passenger event

      • Investigate if KCTD15 inhibition affects leukemic cell survival or differentiation

      • Explore KCTD15 as a potential diagnostic biomarker for AML using flow cytometry

    • Investigate KCTD15 in other hematological malignancies:

      • Extend studies to other leukemia subtypes and lymphomas

      • Correlate KCTD15 expression with clinical outcomes and treatment response

  • Exploiting KCTD15's molecular interactions:

    • Target KCTD15's interaction with AP2 transcriptional activity:

      • Characterize the structural basis of KCTD15-AP2 interaction

      • Screen for compounds that modulate this interaction

      • Determine downstream transcriptional effects relevant to cancer biology

    • Explore KCTD15's role in Wnt/β-catenin signaling:

      • Investigate how KCTD15 represses Wnt/β-catenin signaling in development

      • Determine if this mechanism is relevant in cancer contexts

      • Develop strategies to modulate this pathway for therapeutic benefit

  • Addressing the apparent contradictions in KCTD15's role:

    • Conduct systematic multi-cancer analyses:

      • Compare KCTD15 expression and function across diverse cancer types

      • Identify tissue-specific interaction partners

      • Define molecular contexts that determine whether KCTD15 acts as a tumor suppressor or oncogene

    • Develop conditional animal models:

      • Generate tissue-specific KCTD15 knockout/knockin mice

      • Study cancer development and progression in these models

      • Test therapeutic strategies in physiologically relevant in vivo systems

The most promising approach appears to be pursuing KCTD15 as a potential tumor suppressor in colorectal cancer, where restoring its expression or mimicking its pro-apoptotic effects could yield therapeutic benefits. Meanwhile, understanding its apparently contradictory role in hematological malignancies may reveal alternative therapeutic opportunities.

How might multi-omics approaches enhance our understanding of KCTD15's biological functions?

Integrating multi-omics approaches can provide comprehensive insights into KCTD15's biological functions across different contexts:

  • Genomic approaches to understand KCTD15 regulation:

    • Chromatin immunoprecipitation sequencing (ChIP-seq):

      • Identify transcription factors binding to KCTD15 promoter regions

      • Map epigenetic modifications affecting KCTD15 expression

      • Compare regulatory landscapes across tissue types to explain tissue-specific expression

    • Genome-wide association studies (GWAS):

      • Expand on existing associations with body mass index

      • Explore genetic variations affecting KCTD15 expression or function

      • Correlate with disease susceptibility beyond metabolic disorders

  • Transcriptomic profiling to identify KCTD15-dependent gene networks:

    • RNA sequencing after KCTD15 modulation:

      • Compare gene expression changes following KCTD15 overexpression or knockdown

      • Identify tissue-specific transcriptional responses

      • Apply pathway analysis to map KCTD15's influence on cellular processes

    • Single-cell RNA sequencing:

      • Characterize cell-type-specific expression patterns

      • Identify rare cell populations with unique KCTD15 expression

      • Track expression dynamics during development or disease progression

  • Proteomic approaches to map KCTD15 interaction networks:

    • Immunoprecipitation-mass spectrometry (IP-MS):

      • Identify direct KCTD15 binding partners

      • Compare interactomes across different cell types

      • Validate key interactions with co-immunoprecipitation

    • Proximity labeling approaches:

      • Use BioID or APEX2 fused to KCTD15 to identify proximal proteins

      • Map spatial organization of KCTD15 complexes

      • Discover transient or weak interactions missed by traditional IP

  • Metabolomic integration to elucidate functional consequences:

    • Metabolite profiling after KCTD15 modulation:

      • Identify metabolic pathways influenced by KCTD15

      • Connect to known associations with body mass index and obesity

      • Discover potential metabolic vulnerabilities in cancer contexts

  • Integrative data analysis approaches:

    • Network-based data integration:

      • Construct multi-layer networks connecting genomic, transcriptomic, and proteomic data

      • Identify key nodes and pathways regulated by KCTD15

      • Develop predictive models of KCTD15 function

    • Machine learning applications:

      • Train algorithms to recognize KCTD15-associated patterns across omics datasets

      • Predict functional outcomes of KCTD15 modulation

      • Identify potential therapeutic targets within KCTD15-associated networks

This multi-omics strategy would be particularly valuable for resolving the apparent contradictions in KCTD15's function across different tissues and disease states, potentially revealing context-specific interaction networks that explain how the same protein can act as a tumor suppressor in colorectal cancer while being upregulated in leukemia.

Product Science Overview

Gene and Protein Information
  • Gene Name: KCTD15
  • Aliases: BTB/POZ domain-containing protein KCTD15, MGC25497
  • Protein Coding: Yes
  • Location: The KCTD15 gene is located on chromosome 19 in humans .
Function and Expression

KCTD15 is involved in several critical functions:

  • Embryonic Development: During embryonic development, KCTD15 interferes with neural crest formation .
  • Transcriptional Activity: It inhibits AP2 transcriptional activity by interacting with its activation domain .
  • Expression: KCTD15 is highly expressed in the brain and hypothalamus .
Clinical Significance

KCTD15 has been linked to various health conditions:

  • Obesity: Genetic loci associated with KCTD15 have been linked to higher than normal BMI in humans .
  • Diseases: Diseases associated with KCTD15 include Brachydactyly, Type D, and Coronary Artery Anomaly .
Mouse Anti Human KCTD15 Antibody

The Mouse Anti Human KCTD15 antibody is a monoclonal antibody used in research to study the KCTD15 protein. Here are some key details:

  • Type: Mouse Anti Human Monoclonal .
  • Immunogen: Derived from hybridization of mouse F0 myeloma cells with spleen cells from BALB/c mice immunized with recombinant human KCTD15 amino acids 1-234 purified from E. coli .
  • Applications: This antibody has been tested by ELISA, Western blot, and Immunofluorescence analysis to ensure specificity and reactivity .
Storage and Stability
  • Storage: For periods up to 1 month, store at 4°C. For longer periods, store at -20°C. Prevent freeze-thaw cycles .
  • Shelf Life: 12 months at -20°C, 1 month at 4°C .

Quick Inquiry

Personal Email Detected
Please use an institutional or corporate email address for inquiries. Personal email accounts ( such as Gmail, Yahoo, and Outlook) are not accepted. *
© Copyright 2024 Thebiotek. All Rights Reserved.