FKBP10 facilitates post-translational modifications of collagen types I and III through its peptidyl-prolyl cis-trans isomerase (PPIase) activity, directly influencing extracellular matrix (ECM) stability . Its interaction with HSP47 and BiP forms a collagen-specific chaperone complex critical for proper fibril assembly .
Cancer Biomarker Detection: Quantifies FKBP10 overexpression in clear cell renal cell carcinoma (ccRCC), gastric cancer (GC), and melanoma (IHC, WB)
Therapeutic Development: Evaluates FKBP10 inhibition efficacy in:
PI3K-AKT Modulation: FKBP10 knockdown reduces p-AKT (S473) by 62% in GC cells, impairing adhesion
Metabolic Reprogramming: In ccRCC, FKBP10-LDHA interaction increases lactate production 2.3-fold (p<0.01)
Therapeutic Resistance: FKBP10high tumors show 48% reduced response to HIF2α inhibitors (p=0.014)
FKBP10, also known as FKBP65 (65 kDa FK506-binding protein), is an endoplasmic reticulum (ER) resident protein with four tandem peptidyl-prolyl cis/trans isomerase (PPIase) domains. It functions as a molecular chaperone that modulates the folding and trafficking of secretory proteins . FKBP10 plays a particularly important role in collagen folding and secretion, with mutations in this protein being associated with osteogenesis imperfecta . The protein is typically expressed in developing tissues and re-expressed in adult tissues following injury . Beyond its structural roles, FKBP10 has been implicated in cell adhesion processes and may function through the integrin/AKT signaling pathway .
FKBP10 antibodies are validated for multiple research applications including:
Western Blotting (WB): Typically used at dilutions of 1:1000-1:16000 depending on the specific antibody
Immunohistochemistry (IHC): Used at dilutions of 1:50-1:500, often with TE buffer pH 9.0 for antigen retrieval
Immunoprecipitation (IP): Approximately 0.5-4.0 μg antibody per 1.0-3.0 mg of total protein lysate
Immunofluorescence (IF): Validated in multiple published studies
These applications allow researchers to detect endogenous FKBP10 in various cell lines including HEK-293, HeLa, A375, and NIH/3T3 cells, as well as in human tissues such as brain, placenta, kidney, and ovarian cancer samples .
FKBP10 antibodies should be stored at -20°C for long-term stability. Most commercial preparations come in PBS buffer with 0.02% sodium azide and 50% glycerol at pH 7.3 . Under these conditions, the antibodies remain stable for at least one year after shipment. For the 20 μl size that contains 0.1% BSA, aliquoting is unnecessary for -20°C storage . It's important to avoid repeated freeze-thaw cycles to maintain antibody activity and specificity. Do not aliquot certain commercial antibody preparations as specified by the manufacturer .
For optimal FKBP10 immunohistochemistry staining, two antigen retrieval methods have shown consistent results:
For sodium citrate buffer method, tissue sections should be boiled at 100-120°C for 5 minutes . Following antigen retrieval, endogenous peroxidase activity should be blocked using 3% hydrogen peroxide at room temperature for 10 minutes . The sections should then be incubated with the primary FKBP10 antibody (dilutions ranging from 1:50 to 1:700 depending on the specific antibody) overnight at 4°C, followed by incubation with an appropriate conjugated secondary antibody at room temperature for 30 minutes . Visualization can be performed using 3',3'-diaminobenzidene staining at room temperature for approximately 5 minutes .
When evaluating FKBP10 expression in immunohistochemistry, a systematic scoring approach is recommended:
Select five random fields under a light microscope (magnification ×200)
Calculate an immunoreaction score (IRS) based on:
This standardized scoring system allows for consistent quantification of FKBP10 expression across different samples and studies, facilitating reliable comparison of results. For prognostic studies, correlation of these scores with clinical outcomes can help establish the value of FKBP10 as a biomarker.
For rigorous Western blot experiments with FKBP10 antibodies, researchers should include:
Positive controls: HEK-293 cells, HeLa cells, and A375 cells have been validated to express detectable levels of endogenous FKBP10
Negative controls: Include a lane with the secondary antibody only (no primary antibody)
Knockdown/knockout controls: siRNA-mediated knockdown of FKBP10 (as described in several publications) provides a critical specificity control
Loading controls: β-actin is commonly used as a loading control in FKBP10 studies
The expected molecular weight for FKBP10 is approximately 64-70 kDa , although the calculated molecular weight based on amino acid sequence is 64 kDa . This slight discrepancy may be due to post-translational modifications. Researchers should ensure their gel system can resolve proteins in this range and adjust antibody dilutions based on expression levels in their specific samples.
For investigating FKBP10 function in cancer through knockdown experiments:
Cell line selection: Based on published research, gastric cancer cell lines HGC-27 and MKN-7 show high endogenous expression of FKBP10, making them suitable models . Other cancer cell lines with verified FKBP10 expression include HEK-293, HeLa, and A375 .
Knockdown strategy:
Functional assays:
Adhesion assay: FKBP10 knockdown significantly suppresses cell adhesion in gastric cancer cells
Integrin pathway analysis: Measure changes in expression of integrin family proteins, particularly integrin αV and α6, which are reduced following FKBP10 knockdown
PI3K-AKT signaling assessment: Examine phosphorylated AKT levels (both P-AKT 473 and P-AKT 308), which decrease after FKBP10 knockdown while total AKT remains unchanged
Metastasis-related phenotypes: Since FKBP10 expression correlates with lymph node metastasis in gastric cancer, assess cell migration and invasion capacities before and after knockdown .
This experimental design enables comprehensive evaluation of FKBP10's role in cancer progression, particularly in adhesion-mediated processes that may contribute to metastasis.
Several sophisticated bioinformatic approaches have successfully identified FKBP10-associated pathways in cancer:
Differential expression gene (DEG) screening:
Prognostic value assessment:
Gene Set Enrichment Analysis (GSEA):
Integrative multi-omics analysis:
Correlate FKBP10 expression with other molecular features (mutations, copy number alterations, methylation)
Map FKBP10 into protein-protein interaction networks to identify functional modules
These approaches provide a comprehensive understanding of FKBP10's role in cancer biology and help identify potential therapeutic implications.
FKBP10 antibodies can be instrumental in investigating osteogenesis imperfecta (OI) mechanisms through several specialized approaches:
Mutation-specific analyses:
Collagen interaction studies:
ER stress response evaluation:
Therapeutic targeting:
Screen for compounds that might stabilize mutant FKBP10 or enhance remaining PPIase activity
Evaluate the effects of chemical chaperones on restoring proper collagen folding in FKBP10-deficient cells
These approaches capitalize on the specificity of FKBP10 antibodies to illuminate the molecular mechanisms underlying OI and potentially identify therapeutic interventions.
When encountering non-specific binding with FKBP10 antibodies, researchers should implement the following strategies:
Antibody validation and selection:
Use recombinant antibodies (e.g., rabbit recombinant antibodies) which often show higher specificity than conventional polyclonal antibodies
Verify antibody specificity through FKBP10 knockdown/knockout controls
Consider antibodies purified by antigen affinity methods rather than protein A purification alone
Protocol optimization:
Adjust antibody concentration: Test a dilution series (e.g., 1:2000-1:16000 for Western blot, 1:50-1:500 for IHC)
Increase blocking stringency: Use 5% non-fat milk or BSA in TBST for Western blot
Add 0.1-0.3% Triton X-100 in blocking buffer for IHC to reduce background
Optimize incubation times and temperatures
Sample-specific considerations:
For IHC, compare both recommended antigen retrieval methods (TE buffer pH 9.0 and citrate buffer pH 6.0)
Use fresh tissue samples where possible, as fixation artifacts can increase non-specific binding
For cell lines, ensure endogenous expression levels are sufficient (HEK-293, HeLa, A375, and MKN-7 cells show strong expression)
Confirmatory approaches:
Implementing these approaches systematically will help resolve specificity issues and ensure reliable FKBP10 detection.
When quantifying FKBP10 expression in clinical samples, researchers should be aware of several common pitfalls:
Sample heterogeneity challenges:
Tumor samples often contain varying amounts of stromal cells and infiltrating immune cells
Solution: Use laser capture microdissection to isolate specific cell populations or employ dual staining with cell-type specific markers
Standardization issues:
Variability in fixation protocols and processing times between samples affects antigen preservation
Solution: Standardize pre-analytical variables and include standardized control samples in each batch
Scoring system limitations:
Reference gene selection for RT-PCR:
Inappropriate housekeeping genes may themselves be regulated in disease states
Solution: Validate multiple reference genes for stability in your specific tissue type before quantitative analysis
Prognostic interpretation challenges:
When encountering discrepancies between FKBP10 mRNA and protein expression data, researchers should consider several biological and technical factors:
Post-transcriptional regulation mechanisms:
miRNA-mediated regulation may target FKBP10 mRNA without affecting transcription
RNA-binding proteins may alter mRNA stability or translation efficiency
Systematically investigate potential regulatory elements in the FKBP10 3'UTR
Post-translational modifications and protein stability:
Technical considerations:
Experimental validation approaches:
Pulse-chase experiments to determine protein half-life
Use of proteasome inhibitors to assess degradation pathways
Analysis of polysome fractions to evaluate translation efficiency
Comparison of multiple independent detection methods
Biological significance assessment:
Understanding these factors will help researchers interpret discrepancies meaningfully and may lead to discoveries about FKBP10 regulation in normal and disease states.
FKBP10's emerging roles in multiple pathological processes suggest several promising therapeutic targeting strategies:
Cancer therapeutic approaches:
Targeted inhibition: Develop specific small molecule inhibitors against FKBP10's PPIase domains
Disruption of integrin interactions: As FKBP10 knockdown reduces integrin αV and α6 and impairs cell adhesion, targeting this interaction could inhibit metastatic potential
PI3K-AKT pathway modulation: FKBP10's involvement in AKT phosphorylation suggests combination therapies with existing PI3K/AKT inhibitors might be synergistic
Biomarker-guided therapy: High FKBP10 expression correlates with poor prognosis and lymph node metastasis in gastric cancer, potentially identifying patients who would benefit from more aggressive treatment regimens
Fibrotic disease interventions:
Anti-fibrotic applications: FKBP10 is considered a therapeutic target for idiopathic pulmonary fibrosis (IPF)
Collagen deposition modulation: Given FKBP10's role in collagen folding and secretion, inhibitors could potentially reduce excessive collagen deposition in fibrotic conditions
Conditional targeting: Develop strategies that selectively inhibit FKBP10 in activated fibroblasts while sparing normal tissue functions
Technical challenges and solutions:
Specificity concerns: Design inhibitors that distinguish between different FKBP family members
Delivery strategies: Develop targeted delivery systems to reach specific tissue microenvironments
Combination approaches: Identify synergistic combinations with existing therapies
Clinical development considerations:
Patient stratification: Use FKBP10 expression as a biomarker to select patients most likely to respond to targeted therapies
Monitoring efficacy: Employ FKBP10 antibodies in pharmacodynamic studies to assess target engagement
These multifaceted approaches highlight FKBP10's potential as both a biomarker and therapeutic target across multiple diseases.
Several innovative applications of FKBP10 antibodies are emerging in translational research:
Liquid biopsy development:
Theranostic applications:
Antibody-drug conjugates targeting FKBP10 in cancers with high expression
Imaging applications using labeled FKBP10 antibodies to visualize tumor margins or metastatic spread
Monitoring treatment response through serial measurement of FKBP10 levels
Tissue microenvironment analysis:
Disease heterogeneity characterization:
FKBP10 expression patterns across cancer subtypes to identify distinct biological behaviors
Correlation with treatment resistance phenotypes in various cancer types
Integration with genomic profiling to identify synthetic lethality opportunities
Organoid and 3D culture applications:
Using FKBP10 antibodies to assess protein expression and localization in patient-derived organoids
Functional studies in 3D cultures to better recapitulate in vivo collagen interactions
These emerging applications demonstrate how FKBP10 antibodies are expanding beyond basic research tools to become valuable assets in translational medicine and personalized healthcare approaches.
Advanced multi-omics integration strategies involving FKBP10 antibody-based proteomics include:
Integrated protein-transcriptome analysis:
Correlation of FKBP10 protein levels (detected via antibody-based methods) with mRNA expression
Single-cell proteogenomics to identify cell populations with discordant FKBP10 protein and mRNA levels
Analysis of alternative splicing events affecting FKBP10 function through combined RNA-seq and protein isoform detection
Epigenetic-proteomic integration:
Correlation of FKBP10 protein expression with promoter methylation status
Chromatin immunoprecipitation followed by sequencing (ChIP-seq) to identify transcription factors regulating FKBP10
Investigation of histone modifications at the FKBP10 locus and their correlation with protein expression
Protein interaction networks:
Immunoprecipitation with FKBP10 antibodies followed by mass spectrometry to identify protein interaction partners
Proximity-dependent biotin identification (BioID) to map the FKBP10 protein interaction neighborhood
Integration of these data with public protein-protein interaction databases to place FKBP10 in functional networks
Metabolomic correlations:
Association of FKBP10 expression levels with metabolic profiles
Investigation of collagen metabolism alterations in relation to FKBP10 expression
Integration with lipid profiles, given the potential role of FKBP10 in ER function
Clinical data integration for precision medicine:
Correlation of FKBP10 protein levels with treatment responses
Development of integrated biomarker panels combining FKBP10 with other molecular features
Machine learning approaches to identify patient subgroups based on integrated multi-omics profiles including FKBP10 expression
These integrative approaches leverage the specificity of FKBP10 antibodies while providing a more comprehensive understanding of FKBP10's role in complex biological systems and disease processes.