DNAJC10 (DnaJ homolog subfamily C member 10), also termed ERDJ5, is an endoplasmic reticulum (ER)-resident protein critical for protein folding and quality control . Key features of the DNAJC10 antibody include:
DNAJC10 is implicated in tumor progression and therapy resistance:
Leukemia:
Maintains leukemia stem cell (LSC) survival by suppressing ER stress-induced apoptosis via PERK-EIF2α-ATF4 pathway inhibition .
Knockdown reduces viability of AML cells (THP-1, U937) and sensitizes them to daunorubicin/cytarabine (IC50 decreased 5–8.5-fold) .
High DNAJC10 correlates with poor prognosis in AML patients .
Glioma:
ER Stress Regulation:
DNAJC10 deficiency triggers PERK-EIF2α-CHOP activation, leading to apoptosis under ER stress conditions .
Therapeutic Targeting:
Blocking DNAJC10 enhances chemotherapy efficacy by promoting pro-apoptotic UPR signaling .
While specific protocols for the FITC-conjugated variant are not detailed in the reviewed sources, standard workflows for FITC-based assays include:
Immunofluorescence (IF):
Fix cells with 4% paraformaldehyde.
Permeabilize with 0.1% Triton X-100.
Incubate with DNAJC10-FITC antibody (1:100–1:500 dilution).
Image using fluorescence microscopy (excitation: 494 nm, emission: 518 nm).
Flow Cytometry:
Stain live or fixed cells with antibody (1:50–1:200).
Analyze using a FITC-compatible channel (e.g., FL1).
Current studies focus on unconjugated DNAJC10 antibodies . The FITC-conjugated variant’s performance in multiplex assays or in vivo models remains unexplored. Further research should address:
Cross-reactivity with non-target proteins in FITC-conjugated formats.
Quantitative comparison with other conjugates (e.g., Alexa Fluor®).
DNAJC10, also known as ERdj5 or JPDI, functions as a crucial endoplasmic reticulum co-chaperone that plays a significant role in protein folding and translocation across the endoplasmic reticulum membrane. It's particularly important because it assists in proper folding of nascent polypeptides, maintaining cellular homeostasis and preventing accumulation of misfolded proteins that can lead to cellular stress and various diseases. DNAJC10 interacts with HSP70 chaperone machinery, enhancing activity and ensuring proteins are correctly folded before proceeding to functional destinations . Recent research has shown DNAJC10 is frequently up-regulated in various types of acute myeloid leukemia (AML) and in leukemia stem cell (LSC)-enriched cells, suggesting its importance in cancer research .
DNAJC10-FITC conjugated antibodies have been validated for multiple detection methods including:
Flow cytometry - Optimal for quantifying expression levels in cell populations
Immunofluorescence microscopy - For visualizing subcellular localization in the endoplasmic reticulum lumen
Confocal microscopy - For high-resolution co-localization studies with other ER markers
High-content imaging - For automated screening applications
When designing experiments, researchers should use positive controls (cells known to express DNAJC10) and negative controls (secondary antibody only) to validate staining specificity and optimize detection parameters .
FITC conjugation provides direct visualization capability but researchers should consider these methodological factors:
FITC has excitation/emission peaks of approximately 495/519 nm, making it compatible with standard fluorescence filter sets
The conjugation may slightly reduce antibody avidity compared to unconjugated versions
FITC conjugated antibodies are more susceptible to photobleaching than some other fluorophores
The conjugation ratio (FITC molecules per antibody) affects signal intensity and should be considered when comparing experimental results
pH sensitivity of FITC may impact results in certain buffer conditions
To account for these factors, researchers should use consistent imaging parameters, incorporate anti-fade mounting media, and validate results with alternative detection methods when possible .
For effective DNAJC10-FITC antibody labeling, sample preparation should follow these methodological guidelines:
Cell fixation: 4% paraformaldehyde for 15-20 minutes maintains ER structure while preserving epitope accessibility
Permeabilization: 0.1-0.2% Triton X-100 for 10 minutes provides optimal access to endoplasmic reticulum lumen antigens
Blocking: 5% normal serum (from the species of secondary antibody if using indirect methods) with 1% BSA for 30-60 minutes minimizes non-specific binding
Antibody dilution: Initial testing at 1:500-1:2000 dilution range is recommended based on validation data, with optimization for specific applications
Incubation conditions: Overnight incubation at 4°C generally produces better signal-to-noise ratio than shorter incubations at room temperature
These parameters should be systematically optimized for each cell type and experimental condition to ensure reproducible results.
To effectively study UPR in leukemia models using DNAJC10-FITC antibodies, researchers should implement this methodological framework:
Baseline expression analysis: Establish DNAJC10 expression levels in normal vs. leukemic cells using flow cytometry with FITC-conjugated antibodies
UPR induction verification: Confirm UPR activation using complementary markers (GRP78, CHOP) alongside DNAJC10
Time-course analysis: Monitor DNAJC10 expression changes during UPR activation using:
Chemical inducers (tunicamycin, thapsigargin)
Physiological stressors (glucose deprivation, hypoxia)
Chemotherapeutic agents (daunorubicin, cytarabine)
Co-localization studies: Use confocal microscopy to examine DNAJC10 interaction with PERK pathway components during stress
Functional correlation: Integrate DNAJC10 expression data with survival analysis, as AML patients with high DNAJC10 expression show shorter disease-free survival after chemotherapy
This approach allows researchers to comprehensively characterize how DNAJC10 functions as an oncogene in MLL-AF9-induced AML via regulation of the PERK branch of the UPR .
A rigorous experimental design with DNAJC10-FITC antibodies requires these methodological controls:
Positive control: Cells known to express DNAJC10 (such as THP-1 or U937 leukemia cell lines)
Negative control: DNAJC10 knockout cells generated via CRISPR/Cas9 to confirm antibody specificity
Isotype control: FITC-conjugated non-specific IgG1 (for monoclonal antibodies) or normal IgG (for polyclonal antibodies) at the same concentration
Autofluorescence control: Unstained cells to establish baseline fluorescence
Absorption control: Pre-incubation of antibody with recombinant DNAJC10 protein to demonstrate binding specificity
Secondary antibody-only control: For indirect immunofluorescence protocols
Wild-type vs. stressed cells: Comparing normal conditions with ER stress inducers to demonstrate functional regulation
These controls enable proper interpretation of results and troubleshooting of technical issues that may arise during experiments.
For multi-color flow cytometry with DNAJC10-FITC antibodies, researchers should implement these methodological steps:
Panel design considerations:
FITC emits in the 520nm range, so avoid fluorophores with significant spectral overlap (PE, GFP)
Pair with fluorophores like APC, PE-Cy7, or BV421 for clear separation
Reserve FITC for moderately expressed proteins; use brighter fluorophores for low-abundance targets
Compensation controls:
Prepare single-color controls for each fluorophore
Include DNAJC10-FITC single-stained cells
Use compensation beads for channels where cellular expression is variable
Staining protocol optimization:
Titrate antibody concentrations to determine optimal signal-to-noise ratio
Sequence antibody addition based on epitope accessibility
For intracellular DNAJC10 detection, use permeabilization reagents compatible with surface marker detection
Data analysis approach:
This comprehensive approach enables accurate assessment of DNAJC10 expression in heterogeneous cell populations.
When utilizing DNAJC10 antibodies in patient-derived xenograft (PDX) models, researchers should address these methodological considerations:
Species cross-reactivity verification:
Confirm antibody reactivity with both human DNAJC10 (from patient-derived cells) and potential mouse DNAJC10 (from microenvironment)
Use species-specific blocking reagents if necessary
Sample processing protocol:
Fresh tissue: Process within 1-2 hours of collection for optimal epitope preservation
Fixed tissue: Optimize antigen retrieval methods for FFPE samples
Bone marrow samples: Use gentle RBC lysis protocols to preserve DNAJC10 epitopes
Experimental design framework:
Compare DNAJC10 expression between control and experimental groups
Track DNAJC10 expression during disease progression and treatment response
Correlate with clinical parameters and treatment outcomes
Validation approaches:
Use multiple detection methods (flow cytometry, immunohistochemistry)
Confirm expression changes with protein quantification (Western blot)
Correlate with functional assays measuring UPR activation
Research has shown that mice transplanted with DNAJC10-KO cells displayed significantly decreased leukemia burden compared with wild-type controls, making this approach valuable for studying DNAJC10's role in disease progression .
For accurate quantification of DNAJC10 expression using FITC-conjugated antibodies, implement this methodological framework:
Flow cytometry quantification:
Use calibration beads with known FITC molecules to establish standard curves
Report results as molecules of equivalent soluble fluorochrome (MESF)
Apply compensation to correct for spectral overlap in multi-color panels
Calculate median fluorescence intensity (MFI) and fold change relative to controls
Image-based quantification:
Capture images using consistent exposure settings
Apply background subtraction using non-specific regions
Use integrated density measurements (area × mean intensity)
Normalize to cell number or nuclear staining
Western blot correlation:
Statistical analysis:
Apply appropriate statistical tests based on data distribution
Use multiple biological replicates (n≥3) to account for variability
Consider hierarchical analysis for patient-derived samples
This systematic approach ensures reliable quantification of DNAJC10 expression across different experimental platforms .
When working with DNAJC10-FITC antibodies, researchers may encounter these background sources and should implement the corresponding methodological solutions:
Non-specific binding:
Problem: Antibody binding to Fc receptors or hydrophobic interactions
Solution: Include 5-10% serum from the host species of secondary antibody and 1% BSA in blocking buffer; use Fc receptor blocking reagents for flow cytometry
Autofluorescence:
Problem: Cellular components naturally fluoresce in the FITC channel
Solution: Include unstained controls; use Sudan Black B (0.1-0.3%) to quench autofluorescence; consider spectral unmixing during analysis
Fixation artifacts:
Problem: Excessive fixation can create aldehyde-induced fluorescence
Solution: Optimize fixation time; use freshly prepared paraformaldehyde; include 0.1M glycine wash step to quench free aldehydes
Insufficient washing:
Problem: Residual unbound antibody contributing to background
Solution: Increase number and volume of washing steps; use gentle agitation during washes
Photobleaching effects:
Problem: FITC signal fading during analysis
Solution: Use anti-fade mounting media; minimize exposure to light; acquire images systematically from control to experimental groups
Implementing these targeted solutions will significantly improve signal-to-noise ratio when working with DNAJC10-FITC antibodies .
When interpreting DNAJC10 expression differences between normal and cancer cells, researchers should consider these methodological frameworks:
Baseline expression contextualization:
Functional correlation approach:
Pathway integration framework:
Interpret DNAJC10 expression in context of UPR activation markers (GRP78, CHOP)
Correlate with PERK-EIF2α-ATF4 pathway activation status
Consider relationship with pro-apoptotic (BAX) and pro-survival UPR components
Therapeutic relevance analysis:
This interpretive framework places DNAJC10 expression changes within a clinically and biologically relevant context.
To distinguish specific DNAJC10-FITC signals from non-specific fluorescence, implement these methodological approaches:
Control implementation strategy:
Use DNAJC10 knockout or knockdown cells as negative controls
Include isotype-matched FITC-conjugated irrelevant antibodies
Compare with secondary antibody-only controls for indirect detection methods
Signal validation techniques:
Quantitative analysis approach:
Establish signal threshold based on negative controls
Apply consistent gating strategy across all samples
Use signal-to-noise ratio rather than absolute intensity for comparisons
Advanced verification methods:
Demonstrate signal increase in cells overexpressing DNAJC10
Show signal augmentation under conditions known to upregulate DNAJC10 (ER stress)
Perform co-localization analysis with other ER markers
This comprehensive strategy ensures that observed signals represent genuine DNAJC10 expression rather than technical artifacts .
DNAJC10-FITC antibodies can be effectively employed to study ER stress in AML using these methodological approaches:
Baseline expression profiling:
Stress induction monitoring:
Functional manipulation experiments:
Mechanistic pathway analysis:
Use multi-parameter flow cytometry to correlate DNAJC10 with:
PERK-EIF2α-ATF4 pathway activation
Pro-survival vs. pro-apoptotic UPR balance
Cell cycle distribution and apoptosis markers
Research has demonstrated that DNAJC10 deficiency leads to activation of the pro-apoptotic PERK branch of UPR and enhances chemosensitivity, providing a valuable framework for therapeutic investigations .
To effectively investigate DNAJC10's role in chemotherapy resistance, researchers should implement these methodological approaches:
Expression correlation studies:
Analyze DNAJC10 expression in paired pre-treatment and relapsed patient samples
Correlate baseline expression with treatment response
Monitor expression changes during treatment and at relapse
Data shows AML patients with high DNAJC10 expression tend to have shorter disease-free survival after "DNR + Ara-C" treatment
In vitro sensitivity profiling:
Mechanism exploration design:
In vivo resistance models:
Establish PDX models with varying DNAJC10 expression levels
Monitor treatment response and resistance development
Analyze DNAJC10 expression in residual disease after treatment
Test combination strategies targeting DNAJC10 and standard chemotherapy
This comprehensive experimental framework addresses the finding that up-regulated DNAJC10 expression is closely related to daunorubicin and cytarabine resistance in AML patients .
To identify potential therapeutic targets in the UPR pathway using DNAJC10 antibodies, researchers should implement this methodological framework:
Interaction partner identification:
Pathway modulation screening:
Drug sensitivity profiling:
Combination therapy design:
Develop rational combinations targeting:
DNAJC10 expression or function
Downstream effectors in pro-survival UPR branches
Protein quality control mechanisms
Test combinations in patient-derived models with varying DNAJC10 expression
This systematic approach leverages the finding that deficiency of DNAJC10 activates the PERK-EIF2α-CHOP axis and pro-apoptotic factors, potentially enhancing chemotherapy efficacy .
For rigorous validation of DNAJC10-FITC antibodies in novel cell types, researchers should implement this methodological framework:
Expression verification strategy:
Dilution optimization protocol:
Specificity confirmation methods:
Generate transient knockdown controls (siRNA/shRNA)
Validate with genetic knockout references when available
Perform epitope-blocking experiments
Compare staining patterns across different detection platforms
Application-specific validation:
For flow cytometry: optimize voltage settings and compensation
For microscopy: determine ideal exposure settings and filter combinations
For multiplexed assays: verify absence of interference with other fluorophores
This systematic validation approach ensures reliable and reproducible results when extending DNAJC10-FITC antibody applications to previously untested cell types .
To characterize DNAJC10 correlation with other UPR markers during stress responses, researchers should implement this methodological framework:
Temporal expression profiling:
Monitor expression kinetics of DNAJC10 alongside other UPR markers:
GRP78/BiP (general UPR indicator)
PERK, phospho-PERK (PERK pathway)
XBP1s (IRE1 pathway)
ATF6 (cleaved fragments)
Track at multiple timepoints (2h, 6h, 12h, 24h) after stress induction
Research shows Ara-C or DNR treatment significantly induces DNAJC10 and GRP78 up-regulated expression in a dose-dependent manner
Pathway-specific correlation analysis:
Multi-parameter quantification approaches:
Use flow cytometry for single-cell correlation analysis
Implement immunofluorescence for spatial relationship assessment
Apply Western blotting for population-level protein quantification
Correlate with functional readouts (apoptosis, proliferation)
Stress-type specific patterns:
Compare patterns across different stressors:
ER stress inducers (tunicamycin, thapsigargin)
Chemotherapeutic agents (daunorubicin, cytarabine)
Physiological stressors (hypoxia, nutrient deprivation)
This comprehensive approach provides mechanistic insights into how DNAJC10 functions within the broader UPR network during cellular stress responses .
To effectively incorporate DNAJC10-FITC antibodies into high-throughput screening applications, researchers should implement this methodological framework:
Assay miniaturization protocol:
Adapt staining protocols for 96/384-well format
Optimize antibody concentrations for microvolume applications
Develop automated liquid handling procedures
Establish consistent cell seeding densities for reproducibility
Automated image acquisition parameters:
Define optimal exposure settings for FITC channel
Establish z-stack parameters for maximum signal capture
Program consistent field selection algorithms
Implement autofocus routines optimized for ER structures
Analysis pipeline development:
Create cell segmentation algorithms based on nuclear and cytoplasmic markers
Develop DNAJC10 signal quantification methods:
Mean fluorescence intensity
Subcellular distribution patterns
Colocalization with ER markers
Establish normalization procedures for plate-to-plate comparison
Application-specific optimization:
For drug screens: determine optimal treatment duration and concentration ranges
For genetic screens: establish positive/negative control thresholds
For patient sample analysis: develop standardization protocols
For all applications: implement quality control metrics and outlier detection