KLHDC8A Antibody

Shipped with Ice Packs
In Stock

Description

Definition and Overview of KLHDC8A Antibody

KLHDC8A Antibody is a laboratory reagent designed for immunodetection of the kelch domain-containing protein 8A (KLHDC8A), a 38.9 kDa protein implicated in various cancers, including glioma. Its canonical form includes 350 amino acids, and it is also referred to as S-delta-E1 or substitute for delta-EGFR expression 1. The antibody facilitates protein detection via techniques such as Western blot (WB), immunohistochemistry (IHC), immunofluorescence (IF), and ELISA .

Applications in Research and Clinical Studies

ApplicationMethodsKey Use Cases
Protein expressionWB, IHC, IFQuantifying KLHDC8A levels in glioma cell lines (e.g., U251, U87)
Diagnostic biomarkerROC curve analysisDistinguishing glioblastoma (GBM) from normal tissue (AUC = 0.920)
Mechanistic studiesKnockdown + WBAnalyzing Wnt/β-catenin pathway activation in glioma proliferation
Immune profilingTissue microarray + IHCAssessing macrophage infiltration and PD-L1 co-expression in glioma

Mechanistic Insights

  • Wnt/β-catenin Pathway: Knockdown of KLHDC8A in glioma cells (U251, U87) reduced β-catenin, CDK4, and CDK6 levels, impairing proliferation and colony formation .

  • Hedgehog Signaling: In GBM stem cells (GSCs), KLHDC8A promotes primary cilia assembly and Hedgehog pathway activation via CCT binding, enhancing self-renewal .

  • Immune Microenvironment: KLHDC8A expression correlates with M2 macrophage infiltration, neutrophils, and regulatory T cells, suggesting a role in immune evasion .

Metabolic Regulation

  • Lactate-Driven Upregulation: Lactate from glycolysis stimulates KLHDC8A expression, promoting tumorigenesis via ERK and P38 MAPK activation .

Biomarker Utility

Cancer TypeDiagnostic/Prognostic ValueSource
LGGAUC = 0.748 for diagnosis; independent OS/DSS predictor
GBMAUC = 0.920 for diagnosis; no survival correlation

Therapeutic Targeting

  • Combination Therapy: Targeting KLHDC8A alongside Aurora B/C kinase inhibitors enhances antitumor effects in GSCs by suppressing cilia formation and Hedgehog signaling .

  • Immune Modulation: Inhibiting KLHDC8A may reduce M2 macrophage infiltration, a strategy to counteract immune suppression in glioma .

Product Specs

Buffer
**Preservative:** 0.03% Proclin 300
**Constituents:** 50% Glycerol, 0.01M PBS, pH 7.4
Form
Liquid
Lead Time
Typically, we can ship your order within 1-3 business days of receipt. Delivery timelines may vary depending on the purchasing method or location. For specific delivery timeframes, please contact your local distributor.
Synonyms
KLHDC8A antibody; Kelch domain-containing protein 8A antibody; Substitute for delta-EGFR expression 1 antibody; S-delta-E1 antibody
Target Names
KLHDC8A
Uniprot No.

Q&A

What is KLHDC8A and what cellular functions does it regulate?

KLHDC8A (Kelch Domain Containing 8A) is a protein implicated in several cancers, particularly gliomas. This protein plays multiple critical roles in cellular processes:

  • It binds to chaperonin-containing TCP1 (CCT) to promote the assembly of primary cilia, thereby activating the Hedgehog signaling pathway .

  • It functions within the Wnt/β-catenin signaling pathway, as knockdown of KLHDC8A in glioma cell lines leads to downregulation of key proteins including β-catenin, CDK4, and CDK6 .

  • It regulates cell proliferation and growth, evidenced by functional studies where KLHDC8A silencing reduced cell proliferation and colony formation in glioma cell lines (U251 and U87) .

  • It may influence tumor microenvironment interactions, though this finding comes from a retracted study and requires further validation .

For researchers using KLHDC8A antibodies, understanding these diverse functions is essential for designing experiments that investigate its role in both normal physiology and pathological conditions.

How is KLHDC8A expression regulated in normal tissues versus cancer tissues?

KLHDC8A expression shows distinct regulatory patterns between normal and cancer tissues:

  • Pan-cancer analysis reveals significant dysregulation of KLHDC8A across various tumor types, with notably increased expression in certain cancers .

  • In gliomas specifically, KLHDC8A expression is significantly higher in tumor tissues compared to normal brain tissues .

  • Epigenetic regulation through superenhancers plays a crucial role in KLHDC8A expression in glioblastoma stem cells (GSCs) .

  • Transcription factor control circuitry analyses have identified SOX2 as a master regulator that stimulates KLHDC8A expression .

  • SOX2 and OLIG2 display increased binding within 500 bp of the KLHDC8A superenhancer in GSCs, suggesting that binding of these transcription factors drives KLHDC8A expression .

  • KLHDC8A expression positively correlates with SOX2 and OLIG2 expression in glioblastoma patients from The Cancer Genome Atlas (TCGA) and Chinese Glioma Genome Atlas (CGGA) databases .

These regulatory mechanisms provide important context for researchers interpreting KLHDC8A antibody staining patterns across different tissue types and experimental models.

What are the challenges in detecting KLHDC8A protein in experimental settings?

Detecting KLHDC8A protein presents several experimental challenges:

  • Expression level variations: KLHDC8A expression can vary significantly across different glioma cell lines, with U251 and U87 showing particularly high expression levels . This variability necessitates careful selection of positive controls.

  • Specificity concerns: Like many antibody-based detections, ensuring specificity for KLHDC8A requires rigorous validation using knockdown controls. Previous studies validated specificity by achieving >60% reduction in KLHDC8A expression through shRNA approaches .

  • Cell type-specific expression: KLHDC8A expression appears to be particularly important in glioblastoma stem cells compared to differentiated glioma cells , suggesting that detection sensitivity may vary with cell differentiation state.

  • Subcellular localization: Given KLHDC8A's role in primary cilia assembly and signaling pathway regulation, its subcellular distribution may change depending on cellular context, requiring optimization of fixation and permeabilization protocols.

Researchers should address these challenges through careful experimental design and appropriate controls when using KLHDC8A antibodies.

How can KLHDC8A antibodies be utilized for studying tumor microenvironment interactions?

KLHDC8A's potential role in tumor microenvironment interactions can be investigated using antibody-based approaches:

  • Multiplex immunohistochemistry: Tissue microarray-based multiple immunohistochemical staining can be employed to simultaneously detect KLHDC8A and markers of tumor-infiltrating immune cells, enabling spatial relationship analysis between KLHDC8A-expressing cells and immune components .

  • Co-localization studies: Confocal microscopy using fluorescently-labeled KLHDC8A antibodies combined with immune cell markers (e.g., CD68 for macrophages) can reveal potential interactions at the cellular level.

  • Flow cytometry: For dissociated tumor samples, antibodies against KLHDC8A combined with immune cell markers can quantify co-expression patterns and identify specific cell populations expressing KLHDC8A.

  • Cell sorting followed by functional assays: KLHDC8A antibodies can be used to isolate KLHDC8A-high versus KLHDC8A-low cell populations for subsequent functional characterization of their interactions with immune cells.

While some research has suggested associations between KLHDC8A and immune features such as macrophage infiltration, it's important to note that one key study on this topic was retracted , highlighting the need for independent validation of these findings.

What molecular pathways interact with KLHDC8A and how can antibodies help elucidate these connections?

KLHDC8A interacts with several critical molecular pathways in cancer, which can be investigated using antibody-based approaches:

  • Wnt/β-catenin pathway:

    • KLHDC8A knockdown leads to downregulation of key Wnt/β-catenin pathway proteins, including β-catenin, CDK4, and CDK6 .

    • Co-immunoprecipitation with KLHDC8A antibodies followed by Western blotting for Wnt pathway components can identify direct protein-protein interactions.

    • Proximity ligation assays using KLHDC8A antibodies paired with antibodies against Wnt pathway proteins can visualize and quantify these interactions in situ.

  • Hedgehog signaling pathway:

    • KLHDC8A binds to chaperonin-containing TCP1 (CCT) to promote primary cilia assembly, activating the Hedgehog pathway .

    • Immunofluorescence co-localization studies with KLHDC8A antibodies and ciliary markers can visualize this function.

    • ChIP-seq with KLHDC8A antibodies followed by pathway enrichment analysis can identify genomic regions and genes associated with Hedgehog signaling.

  • Aurora B/C Kinase pathway:

    • KLHDC8A expression correlates with Aurora B/C Kinase inhibitor activity, which induces primary cilia and hedgehog signaling .

    • Phospho-specific antibodies for Aurora kinase substrates combined with KLHDC8A detection can map the relationships between these pathways.

Understanding these pathway interactions is crucial for developing targeted therapies that exploit KLHDC8A's role in glioma progression.

How does KLHDC8A expression correlate with genomic alterations in glioma?

The relationship between KLHDC8A expression and genomic alterations in glioma can provide insights into its role in tumorigenesis:

  • IDH mutation status: While not explicitly stated in the provided research, the differential prognostic significance of KLHDC8A in LGG versus GBM suggests potential associations with molecular subtypes defined by IDH mutation status. Researchers can investigate this relationship using KLHDC8A antibodies in combination with IDH mutation-specific antibodies in multiparameter analyses.

  • Chromosome 1p/19q codeletion: As another key molecular classifier of gliomas, the relationship between KLHDC8A expression and 1p/19q codeletion status warrants investigation through integrated analysis of immunohistochemistry data with genomic profiling.

  • MGMT promoter methylation: The relationship between KLHDC8A expression and MGMT promoter methylation status could inform understanding of treatment response patterns. Researchers can correlate KLHDC8A immunohistochemistry results with MGMT methylation status determined by methylation-specific PCR.

  • Superenhancer landscapes: KLHDC8A has been identified as an epigenetically driven oncogene through superenhancer landscape analysis . Researchers can employ ChIP-seq using antibodies against histone marks associated with superenhancers (e.g., H3K27ac) in conjunction with KLHDC8A expression analysis.

Integrating KLHDC8A antibody-based detection methods with genomic and epigenomic analyses can provide a more comprehensive understanding of its role in glioma biology.

What controls should be included when using KLHDC8A antibodies for immunohistochemistry?

Rigorous controls are essential for reliable immunohistochemistry with KLHDC8A antibodies:

  • Positive tissue controls: Include tissues known to express KLHDC8A, such as glioma tissues (particularly LGG and GBM) based on previous studies . Multiple positive controls representing different expression levels are ideal.

  • Negative tissue controls: Include normal brain tissue, which typically shows lower KLHDC8A expression compared to glioma tissues . Also consider including tissues not expected to express KLHDC8A based on database expression profiles.

  • Cellular controls: If possible, include KLHDC8A-knockdown or knockout cell lines or tissues as specificity controls. Previous studies have used shRNA-mediated knockdown of KLHDC8A in glioma cell lines to validate antibody specificity .

  • Technical controls:

    • Antibody omission control: Perform the staining procedure without primary antibody to assess non-specific binding of the detection system.

    • Isotype control: Use a non-specific antibody of the same isotype and concentration as the KLHDC8A antibody to identify non-specific binding.

    • Peptide competition/blocking: Pre-incubate the KLHDC8A antibody with its immunizing peptide to demonstrate binding specificity.

  • Validation of staining patterns: Compare staining patterns with published data and correlate with mRNA expression data from the same or similar samples when possible.

  • Multi-antibody validation: When feasible, compare staining patterns using different antibodies targeting distinct epitopes of KLHDC8A.

These controls help ensure that observed staining patterns accurately reflect KLHDC8A expression in experimental and clinical samples.

What are the key considerations for optimizing Western blot protocols for KLHDC8A detection?

Optimizing Western blot protocols for KLHDC8A detection requires attention to several technical details:

  • Sample preparation:

    • For cell lines, previous studies successfully detected KLHDC8A in glioma cell lines (U251, U87) using standard lysis protocols .

    • Protein extraction from tissue samples may require optimization to ensure complete solubilization of KLHDC8A.

    • Consider both reducing and non-reducing conditions to determine optimal detection of the native protein structure.

  • Protein loading:

    • Based on expression levels, 20-50 μg of total protein is typically sufficient for detection in glioma cell lines with high KLHDC8A expression .

    • Include a concentration gradient to determine the linear detection range for quantitative analyses.

  • Gel percentage:

    • Human KLHDC8A has a molecular weight of approximately 38 kDa, making 10-12% SDS-PAGE gels suitable for optimal resolution.

  • Transfer conditions:

    • Standard semi-dry or wet transfer protocols are generally suitable for proteins in this molecular weight range.

    • Consider transfer time and voltage/current optimization to ensure complete transfer without protein loss.

  • Blocking:

    • 5% non-fat dry milk or 3-5% BSA in TBST is typically effective for reducing non-specific binding.

    • Optimization may be required based on the specific antibody's performance characteristics.

  • Antibody dilution and incubation:

    • Begin with manufacturer's recommended dilution and optimize as needed.

    • Overnight incubation at 4°C often yields better results for primary antibodies with moderate affinity.

  • Detection system:

    • For low abundance detection, consider enhanced chemiluminescence (ECL) systems or fluorescence-based detection for better quantification.

  • Signal verification:

    • Confirm signal specificity with KLHDC8A knockdown samples, which should show at least 60% reduction in band intensity based on previous studies .

These considerations help ensure specific and sensitive detection of KLHDC8A protein in Western blot applications.

How can KLHDC8A antibodies be effectively used in functional studies of glioma cells?

KLHDC8A antibodies can be powerful tools in functional studies investigating its role in glioma biology:

  • Neutralization studies:

    • If the antibody targets functional domains, it may be used to neutralize KLHDC8A activity in live cells.

    • Compare phenotypic effects with those observed in genetic knockdown studies, where KLHDC8A silencing reduced cell proliferation and colony formation .

  • Immunoprecipitation for protein complex analysis:

    • Use KLHDC8A antibodies to pull down protein complexes and identify interaction partners through mass spectrometry.

    • Focus on components of the Wnt/β-catenin pathway or chaperonin-containing TCP1 (CCT) based on previously identified interactions.

  • ChIP-seq applications:

    • If KLHDC8A functions in transcriptional regulation, ChIP-seq with validated antibodies can map its genomic binding sites.

    • Integrate with transcriptomic data to correlate binding with gene expression changes.

  • Live-cell imaging:

    • Use fluorescently labeled KLHDC8A antibody fragments or anti-tag antibodies with tagged KLHDC8A to track its dynamics in living cells.

    • Focus on cilia formation and cell division based on its identified functions .

  • Flow cytometry:

    • Develop protocols for intracellular staining of KLHDC8A to quantify expression levels across cell populations.

    • Combine with cell cycle markers to assess potential cell cycle-dependent expression patterns.

  • Functional rescue experiments:

    • After KLHDC8A knockdown, add back specific domains of KLHDC8A and use domain-specific antibodies to verify expression and localization.

    • This approach can help map the functional significance of different protein regions.

These approaches enable researchers to move beyond correlative observations to establish causative relationships between KLHDC8A and glioma cell behaviors.

How can KLHDC8A antibodies be used for patient stratification in clinical glioma management?

KLHDC8A expression has demonstrated significant prognostic value that could inform clinical decision-making:

By implementing standardized KLHDC8A antibody-based detection in clinical pathology workflows, more precise prognostic information could be provided to guide treatment decisions, particularly for LGG patients.

What is the potential of KLHDC8A as a therapeutic target in glioma?

KLHDC8A shows promise as a therapeutic target based on several lines of evidence:

  • Functional importance:

    • KLHDC8A knockdown experiments have demonstrated reduced glioma cell proliferation and colony formation .

    • Targeting KLHDC8A decreased GSC proliferation and self-renewal, induced apoptosis, and impaired in vivo tumor growth .

  • Pathway modulation:

    • KLHDC8A functions through multiple cancer-relevant pathways:

      • Wnt/β-catenin signaling pathway, with knockdown leading to downregulation of β-catenin, CDK4, and CDK6 .

      • Hedgehog signaling pathway, which KLHDC8A activates by promoting primary cilia assembly through CCT binding .

  • Combination strategies:

    • KLHDC8A expression correlates with Aurora B/C Kinase inhibitor activity .

    • Combinatorial targeting of Aurora B/C kinase and hedgehog signaling showed augmented benefit against GSC proliferation .

  • Selective impact:

    • KLHDC8A inhibition appears to have a more pronounced effect on glioblastoma stem cells compared to differentiated glioma cells , suggesting potential for targeting the tumor-initiating cell population.

  • Diagnostic utility:

    • KLHDC8A's strong diagnostic performance (AUC of 0.920 for GBM and 0.748 for LGG) suggests it could serve as both a therapeutic target and a companion diagnostic marker.

These findings highlight the potential for developing KLHDC8A-targeted therapeutics, with antibody-based approaches potentially serving both diagnostic and therapeutic functions in glioma management.

How do KLHDC8A expression patterns correlate with response to standard glioma treatments?

Understanding the relationship between KLHDC8A expression and treatment response can inform personalized therapeutic strategies:

  • Predictive biomarker potential:

    • While direct evidence linking KLHDC8A expression to response to standard glioma treatments (surgery, radiation, temozolomide) is limited in the current research, its prognostic significance suggests potential relevance to treatment outcomes .

    • The association between KLHDC8A and survival specifically in LGG but not GBM indicates that its predictive value may vary by glioma subtype and treatment context.

  • Pathway-based treatment implications:

    • KLHDC8A's involvement in the Wnt/β-catenin pathway suggests that its expression might predict response to Wnt pathway inhibitors, which are in development for various cancers.

    • Its role in Hedgehog pathway activation implies that high KLHDC8A expression might be associated with sensitivity to Hedgehog inhibitors, some of which are FDA-approved for other cancer types.

  • Resistance mechanisms:

    • The relationship between KLHDC8A and cancer stem cell properties suggests it might contribute to treatment resistance, as cancer stem cells are often implicated in therapy resistance and tumor recurrence.

  • Monitoring treatment response:

    • KLHDC8A antibody-based detection in liquid biopsies or serial tumor samples could potentially monitor treatment efficacy and disease progression.

Future research should directly investigate the predictive value of KLHDC8A expression for response to specific therapeutic regimens, potentially leading to KLHDC8A antibody-based companion diagnostics for treatment selection in glioma.

What approaches can be used to validate KLHDC8A antibody specificity for research applications?

Rigorous validation of KLHDC8A antibody specificity is crucial for reliable research results:

  • Genetic validation:

    • The gold standard approach involves using KLHDC8A knockout or knockdown models as negative controls. Previous studies achieved >60% reduction in KLHDC8A expression using shRNA approaches .

    • Complement knockdown validation with rescue experiments by expressing exogenous KLHDC8A not targeted by the shRNA, which should restore antibody binding .

  • Multiple antibody approach:

    • Compare staining patterns using antibodies targeting different epitopes of KLHDC8A.

    • Consistent results across different antibodies increase confidence in specificity.

  • Western blot validation:

    • Confirm detection of a single band at the expected molecular weight (approximately 38 kDa for human KLHDC8A).

    • Perform peptide competition assays where pre-incubation with the immunizing peptide should eliminate or significantly reduce the specific band.

  • Correlation with mRNA expression:

    • Compare protein detection patterns with mRNA expression data from the same samples.

    • Strong correlation between protein and mRNA levels supports antibody specificity.

  • Cross-species reactivity testing:

    • If the antibody is claimed to be cross-reactive with KLHDC8A from multiple species, validate specificity in each species.

    • Consider species-specific positive and negative controls.

  • Mass spectrometry validation:

    • For definitive validation, perform immunoprecipitation with the KLHDC8A antibody followed by mass spectrometry to confirm the identity of the captured protein.

These validation approaches should be documented and reported in publications to support the reliability of findings based on KLHDC8A antibody applications.

What are the common technical challenges when using KLHDC8A antibodies in different applications?

Researchers may encounter several technical challenges when working with KLHDC8A antibodies:

  • Western blotting challenges:

    • Non-specific bands: Optimize blocking conditions (consider 5% milk vs. 3-5% BSA) and antibody dilutions.

    • Weak signal: Consider longer exposure times, enhanced chemiluminescence substrates, or signal amplification systems.

    • High background: Increase washing duration/frequency and optimize secondary antibody dilution.

  • Immunohistochemistry challenges:

    • Inconsistent staining: Standardize fixation time and conditions; consider automated staining platforms for reproducibility.

    • Background staining: Optimize blocking steps and consider tissue-specific blocking agents.

    • Antigen masking: Test multiple antigen retrieval methods (heat-induced epitope retrieval with citrate buffer pH 6.0 vs. EDTA buffer pH 9.0).

  • Immunofluorescence challenges:

    • Autofluorescence: Use Sudan Black B or specialized quenching reagents to reduce tissue autofluorescence.

    • Co-localization studies: Ensure secondary antibodies do not cross-react when performing double or triple labeling.

    • Signal-to-noise ratio: Consider signal amplification systems like tyramide signal amplification for detecting low abundance targets.

  • Flow cytometry challenges:

    • Cell permeabilization: Since KLHDC8A is an intracellular target, optimize permeabilization conditions to balance epitope accessibility with cellular integrity.

    • Fixation artifacts: Different fixatives may affect epitope recognition; compare paraformaldehyde, methanol, and other fixatives.

  • ChIP challenges:

    • Epitope accessibility: The chromatin environment may mask the KLHDC8A epitope; optimize crosslinking and sonication conditions.

    • Non-specific binding: Implement stringent washing conditions and include appropriate controls.

Addressing these technical challenges through methodical optimization can improve the reliability and reproducibility of experiments using KLHDC8A antibodies.

How can KLHDC8A detection be optimized in different glioma models and patient samples?

Optimizing KLHDC8A detection across different experimental systems requires tailored approaches:

  • Cell line models:

    • Based on previous studies, U251 and U87 glioma cell lines show high KLHDC8A expression and can serve as positive controls for antibody optimization .

    • For low-expressing cell lines, consider signal enhancement methods or more sensitive detection systems.

    • Standardize cell culture conditions, as KLHDC8A expression may vary with cell density and growth phase.

  • Patient-derived xenografts (PDXs):

    • For optimal fixation of PDX tissue, use 10% neutral-buffered formalin for 24 hours followed by paraffin embedding.

    • Compare multiple antibody clones and dilutions on PDX sections to identify optimal staining conditions.

    • Validate antibody performance across PDXs derived from different glioma subtypes, as KLHDC8A expression varies between LGG and GBM .

  • Primary patient samples:

    • Develop standardized protocols for tissue collection, fixation, and processing to minimize pre-analytical variables.

    • Consider the impact of treatment history on KLHDC8A expression when analyzing patient samples.

    • For archived FFPE samples, adjust antigen retrieval methods to account for prolonged storage effects.

  • 3D culture models:

    • For spheroids or organoids, optimize penetration of antibodies by extending incubation times or using specific permeabilization protocols.

    • Consider clearing techniques (CLARITY, iDISCO) for thicker 3D cultures to improve imaging depth.

  • Quantification approaches:

    • Implement digital pathology tools for objective quantification of KLHDC8A immunostaining.

    • Develop scoring algorithms that account for both staining intensity and percentage of positive cells.

    • For research with clinical implications, establish scoring thresholds that optimally stratify patients based on outcome data .

These optimization strategies can enhance the sensitivity and specificity of KLHDC8A detection across diverse experimental and clinical glioma samples.

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 2025 TheBiotek. All Rights Reserved.