This antibody is validated for enzyme-linked immunosorbent assay (ELISA) applications . While other HSPA1L antibodies have demonstrated utility in western blotting (WB), immunohistochemistry (IHC), and immunofluorescence (IF) , the FITC-conjugated variant is specifically optimized for fluorescence-based detection in ELISA workflows.
HSPA1L plays critical roles in:
Cancer stem cell regulation: Enhances epithelial-mesenchymal transition (EMT) and radiation resistance in non-small cell lung cancer by activating IGF1Rβ/γβ-catenin pathways
Immune modulation: Interacts with TLR4 to activate dendritic cells and induce Th1 responses
Mitochondrial quality control: Participates in mitophagy regulation through the HSPA1L-Parkin pathway
The antibody has been manufactured and validated under strict quality parameters :
| Parameter | Specification |
|---|---|
| Immunogen | Recombinant human HSPA1L protein |
| Cross-reactivity | Confirmed for human samples |
| Stability | Maintains activity for ≥12 months at -20°C |
Titration required: Optimal dilution varies between experimental systems
Sample compatibility: Validated for human cell lysates and recombinant proteins
Negative controls: Essential to confirm signal specificity in fluorescence-based assays
While this FITC-conjugated version specializes in ELISA, other formats offer broader application compatibility:
This reagent enables quantitative analysis of HSPA1L expression dynamics in:
It plays a pivotal role in the protein quality control system, ensuring proper protein folding, refolding of misfolded proteins, and controlling protein targeting for subsequent degradation. This is accomplished through cycles of ATP binding, ATP hydrolysis, and ADP release, mediated by co-chaperones. The affinity for polypeptides is regulated by its nucleotide-bound state. In the ATP-bound form, it has low affinity for substrate proteins. However, upon ATP hydrolysis to ADP, it undergoes a conformational change that increases its affinity for substrate proteins. This process involves repeated cycles of ATP hydrolysis and nucleotide exchange, allowing for cycles of substrate binding and release. HSPA1L is also a positive regulator of PRKN translocation to damaged mitochondria.
Research has shown that HSPA1L exerts its functions through two primary mechanisms. First, it forms a triple complex with IGF1Rβ (Insulin-like Growth Factor 1 Receptor β) and integrin αV, leading to activation of IGF1Rβ. This activation intensifies EMT-associated cancer stemness and radiation resistance through downstream AKT/NF-κB or AKT/GSK3β/β-catenin pathways. Second, HSPA1L can translocate to the nucleus where it directly binds to the promoter region of β-catenin, functioning as a transcription activator of this important signaling protein that characterizes cancer stem cells by regulating ALDH1 expression .
FITC (Fluorescein Isothiocyanate) conjugation to HSPA1L antibodies provides significant advantages for protein detection and localization studies. While unconjugated antibodies require secondary detection systems, FITC-conjugated antibodies allow direct visualization of the target protein through fluorescence microscopy or flow cytometry.
For HSPA1L research, FITC-conjugated antibodies enable researchers to:
Directly visualize subcellular localization (cytoplasmic versus nuclear) of HSPA1L
Perform co-localization studies with other proteins of interest (especially IGF1Rβ and integrin αV)
Quantify expression levels through flow cytometry
Track dynamic changes in HSPA1L expression or localization in response to experimental conditions
The excitation maximum of FITC at approximately 495nm and emission maximum around 519nm provides a bright green fluorescence that is compatible with most standard fluorescence detection systems, making it an ideal conjugate for immunofluorescence studies of HSPA1L in cancer stem cell research.
Based on comprehensive validation studies, HSPA1L antibodies have demonstrated reliable detection across multiple sample types. The reactivity pattern provides researchers with confidence when selecting appropriate experimental models.
When working with FITC-conjugated HSPA1L antibodies, researchers should consider that validation may vary from unconjugated versions. It is recommended to perform preliminary validation experiments with positive controls known to express HSPA1L, such as HepG2 cells or cancer tissues with documented HSPA1L expression .
HSPA1L expression demonstrates a strong positive correlation with cancer stem cell properties, particularly in non-small cell lung cancer. Research has revealed that HSPA1L is significantly upregulated in ALDH1high cells (a recognized cancer stem cell population) compared to ALDH1low cells .
To effectively investigate this relationship, researchers should employ a multi-methodological approach:
Flow cytometry sorting and analysis: Using the ALDEFLUOR assay to separate ALDH1high and ALDH1low populations, followed by HSPA1L detection using FITC-conjugated antibodies. This allows for quantitative correlation between ALDH1 activity and HSPA1L expression .
Sphere formation assays: Comparing sphere-forming capacity between cells with different HSPA1L expression levels (wild-type, knockdown, and overexpression). Research has demonstrated that HSPA1L suppression significantly decreases sphere formation, while overexpression enhances it .
Colony formation assays: Assessing clonogenic potential in relation to HSPA1L expression, which reveals the self-renewal capacity of cancer cells.
Radiation resistance testing: Examining cell survival after γ-radiation exposure, as HSPA1L has been implicated in radiation resistance through the HSPA1L/IGF1Rβ/AKT pathway .
Immunofluorescence co-localization studies: Using FITC-conjugated HSPA1L antibodies alongside other stem cell markers (CD44, CD133) to visualize spatial relationships.
When analyzing results, it's critical to consider that HSPA1L influences CSC properties through multiple pathways, including direct transcriptional regulation of β-catenin and enhancement of IGF1Rβ activation .
Designing effective co-localization experiments with FITC-conjugated HSPA1L antibodies requires careful planning to ensure reliable and interpretable results:
Spectral compatibility: FITC emits green fluorescence (emission peak ~519nm), so partner fluorophores should have minimal spectral overlap. Recommended pairings include:
FITC (HSPA1L) + TRITC/Cy3 (red: IGF1Rβ)
FITC (HSPA1L) + Cy5/Alexa 647 (far-red: integrin αV)
Sequential staining protocol: For optimal results when investigating the HSPA1L/IGF1Rβ/integrin αV triple complex:
Fix cells with 4% paraformaldehyde (15 minutes, room temperature)
Permeabilize with 0.1% Triton X-100 (10 minutes)
Block with 3% BSA (1 hour)
Apply FITC-conjugated HSPA1L antibody (1:50-1:500 dilution, based on optimization)
Apply compatible conjugated antibodies for other targets
Counterstain nuclei with DAPI
Controls: Include single-stained samples for each antibody to confirm specificity and rule out cross-reactivity or bleed-through.
Quantitative analysis: Use co-localization coefficients (Pearson's, Manders') to quantify spatial relationships between HSPA1L and interacting partners.
Subcellular localization assessment: HSPA1L functions both in cytoplasm (for IGF1Rβ complex formation) and nucleus (for β-catenin transcriptional regulation) . Z-stack confocal imaging provides the most accurate representation of these distinct pools.
Confirming functional associations: Complement imaging with biochemical approaches like proximity ligation assay (PLA) or co-immunoprecipitation to validate physical interactions suggested by co-localization .
HSPA1L's dual functionality presents a unique research opportunity requiring specialized approaches to distinguish between its cytoplasmic role (IGF1Rβ activation) and nuclear role (β-catenin transcription):
Subcellular fractionation coupled with immunoblotting:
Separate nuclear and cytoplasmic fractions
Probe each fraction with HSPA1L antibody
Compare relative abundance between compartments
Correlate with IGF1Rβ phosphorylation status and β-catenin levels
Chromatin immunoprecipitation (ChIP) assay:
Immunofluorescence with co-localization analysis:
FITC-conjugated HSPA1L antibody
Complementary antibodies for IGF1Rβ, integrin αV (cytoplasmic function)
Antibodies for transcription machinery components (nuclear function)
Confocal microscopy with Z-stack analysis
Mutant constructs with compartment-specific targeting:
Nuclear export signal (NES) mutants to restrict HSPA1L to cytoplasm
Nuclear localization signal (NLS) mutants to enhance nuclear localization
Compare effects on IGF1Rβ phosphorylation versus β-catenin expression
Proximity ligation assay (PLA):
The detection of HSPA1L using FITC-conjugated antibodies requires careful optimization of fixation and permeabilization conditions to preserve both antigenicity and fluorescence signal:
Recommended fixation protocols based on sample type:
| Sample Type | Fixative | Duration | Temperature | Notes |
|---|---|---|---|---|
| Cell lines | 4% PFA | 15-20 min | Room temperature | Preserves structure well for subcellular localization studies |
| Cell lines (alternative) | Methanol | 10 min | -20°C | Sometimes yields better detection of nuclear HSPA1L |
| Tissue sections | 10% neutral buffered formalin | 24-48 hours | Room temperature | Follow with paraffin embedding |
| Frozen tissue | 4% PFA | 15-20 min | Room temperature | Prior to cryopreservation |
Permeabilization optimization:
For detecting the dual localization of HSPA1L (cytoplasmic and nuclear), permeabilization conditions must be carefully balanced:
For predominantly cytoplasmic HSPA1L detection (IGF1Rβ complex):
0.1% Triton X-100 in PBS, 10 minutes at room temperature
Alternative: 0.5% Saponin in PBS, 15 minutes at room temperature
For enhanced nuclear HSPA1L detection (β-catenin transcriptional role):
0.5% Triton X-100 in PBS, 15-20 minutes at room temperature
Add a nuclear antigen retrieval step: 10mM sodium citrate buffer (pH 6.0) for 10-20 minutes at 95°C
For dual detection experiments:
Sequential approach: Start with gentler permeabilization (0.1% Triton X-100, 10 minutes)
Optional nuclear antigen retrieval if nuclear signal is weak
Use higher antibody concentration (1:50-1:100 dilution) than typically used for single-location detection
Based on published protocols, the optimal antibody incubation for FITC-conjugated HSPA1L antibodies is overnight at 4°C in a humidity chamber protected from light to prevent photobleaching .
Optimal dilutions and conditions for FITC-conjugated HSPA1L antibodies vary by application. While specific data for FITC-conjugated versions is limited, these recommendations are extrapolated from unconjugated antibody protocols with adjustments for fluorescence preservation:
| Application | Recommended Dilution | Incubation Time | Temperature | Buffer | Special Considerations |
|---|---|---|---|---|---|
| Immunofluorescence (cells) | 1:50-1:500 | Overnight | 4°C | PBS + 1% BSA | Light-protected, humidity chamber |
| Immunofluorescence (tissue) | 1:50-1:200 | Overnight | 4°C | PBS + 1% BSA | Light-protected, humidity chamber |
| Flow cytometry | 1:50-1:200 | 30-60 min | 4°C | PBS + 2% FBS | Keep samples on ice and protected from light |
| Live cell imaging | 1:20-1:100 | 30-60 min | 37°C | Cell culture medium | Minimize exposure time to reduce toxicity |
| Fluorescence microscopy | 1:100-1:500 | Overnight | 4°C | PBS + 1% BSA | Use antifade mounting medium |
Critical optimization notes:
Titration experiment: Always perform a titration experiment (1:20, 1:50, 1:100, 1:200, 1:500) with positive controls (e.g., HepG2 cells, which express high levels of HSPA1L) .
Signal-to-noise ratio: FITC can produce background autofluorescence, especially in tissue samples. Include blocking steps with 10% normal serum from the same species as the secondary antibody (if using indirect methods).
Photobleaching prevention: FITC is relatively prone to photobleaching. Use antifade mounting media containing anti-photobleaching agents like ProLong Gold or DABCO.
Storage of stained samples: Store at 4°C in the dark; FITC signal typically remains stable for 1-2 weeks under proper storage conditions.
Multiplexing considerations: When combining with other fluorophores, analyze samples promptly to minimize differential photobleaching rates between fluorophores .
HSPA1L's dual localization presents unique challenges for comprehensive detection. Research has shown that HSPA1L functions both in the cytoplasm (in complex with IGF1Rβ and integrin αV) and in the nucleus (binding to the β-catenin promoter) . Optimizing detection of both pools requires specific technical approaches:
Recommended optimization protocol:
Fixation optimization:
Test parallel samples with different fixatives:
4% paraformaldehyde (preserves cytoplasmic structures)
Methanol/acetone (often enhances nuclear antigen access)
Compare results to determine if one fixative favors a particular localization
Permeabilization gradient:
Prepare a series of cells/tissues with increasing permeabilization strength:
0.1% Triton X-100 (5 minutes) - Mild
0.3% Triton X-100 (10 minutes) - Moderate
0.5% Triton X-100 (15 minutes) - Strong
Evaluate which condition best reveals both localizations
Antigen retrieval for nuclear detection:
Heat-induced epitope retrieval using citrate buffer (pH 6.0)
Enzymatic retrieval with proteinase K (very mild conditions)
Compare with and without retrieval to determine impact on nuclear signal
Confocal microscopy with Z-stack acquisition:
Capture multiple focal planes (0.5-1μm steps)
Generate maximum intensity projections and orthogonal views
Use nuclear counterstain (DAPI) to clearly delineate nuclear boundaries
Quantitative analysis:
Measure nuclear-to-cytoplasmic ratio of HSPA1L signal
Use cell fractionation followed by Western blot as validation
Compare results under different experimental conditions
Validation approaches:
Use nuclear-targeted or cytoplasm-restricted HSPA1L mutants as controls
Perform parallel chromatin immunoprecipitation to confirm nuclear binding
Co-localize with known markers (IGF1Rβ for cytoplasmic, histones for nuclear) .
Researchers working with FITC-conjugated HSPA1L antibodies may encounter several technical challenges. Here are common issues and their solutions:
1. High background/non-specific staining:
Problem: Diffuse green fluorescence throughout samples without distinct HSPA1L pattern.
Solutions:
Increase blocking stringency (5-10% normal serum, 1-2 hours)
Add 0.1-0.3% Triton X-100 to blocking buffer
Include additional blocking with 10mg/ml BSA
Try different blocking agents (normal serum, BSA, casein, or commercial blockers)
Increase washing steps (5 washes, 5 minutes each)
2. Weak or absent nuclear HSPA1L signal:
Problem: Only cytoplasmic HSPA1L visible despite expecting nuclear localization.
Solutions:
Add heat-induced epitope retrieval step (citrate buffer, pH 6.0)
Increase permeabilization (0.5% Triton X-100, 20 minutes)
Extend primary antibody incubation (48 hours at 4°C)
Reduce fixation time to prevent overfixation
Optimize antibody concentration (try higher concentration)
3. Photobleaching during analysis:
Problem: FITC signal fades rapidly during microscopic examination.
Solutions:
Use anti-fade mounting medium (ProLong Gold, VECTASHIELD)
Minimize exposure to excitation light
Capture images immediately after preparation
Consider spectral imaging to collect signal with minimal exposure
Use newer generation anti-fade reagents containing anti-oxidants
4. Poor co-localization with interacting partners:
Problem: Expected co-localization of HSPA1L with IGF1Rβ or integrin αV not observed.
Solutions:
Optimize fixation to preserve protein complexes (try mild fixatives)
Use proximity ligation assay as alternative approach
Try sequential staining protocol
Validate antibody specificity independently
Consider using split-fluorescent protein approaches for live cell studies
5. Inconsistent staining pattern between experiments:
Problem: Variable HSPA1L staining patterns across experimental replicates.
Solutions:
Accurate interpretation of HSPA1L expression patterns is crucial for understanding its role in cancer stem cells. Based on published research findings, here is a framework for interpretation:
1. Establishing baseline expression patterns:
HSPA1L shows heterogeneous expression in cancer cell populations
Higher expression correlates with ALDH1high cells (established CSC marker)
Co-expression with stem cell transcription factors (Sox2, Oct4, Nanog)
2. Quantitative assessment approaches:
Use digital image analysis software to quantify signal intensity
Establish H-score or intensity thresholds based on control samples
Compare nuclear-to-cytoplasmic ratio across different cell populations
Correlate with other CSC markers using multiparameter flow cytometry
3. Functional correlation matrix:
| HSPA1L Pattern | Biological Interpretation | Validation Approach | Expected Outcome |
|---|---|---|---|
| High cytoplasmic + membrane-proximal | Active IGF1Rβ-integrin αV complex | Measure phospho-IGF1Rβ levels | Increased AKT/NF-κB signaling |
| High nuclear | Transcriptional regulation active | ChIP for β-catenin promoter binding | Increased β-catenin expression |
| High in scattered cells | CSC-like subpopulation | Sphere formation assay with sorted cells | Enhanced self-renewal capacity |
| Gradient across tumor tissue | Heterogeneity in CSC properties | Microdissection + expression analysis | Correlation with invasion front |
4. Context-dependent interpretation:
In epithelial cancers: co-localize with EMT markers (vimentin, N-cadherin)
In therapy-resistant populations: correlate with radiation/drug response
In invasion/metastasis: assess relationship to cell motility markers
In cell cycle phases: determine if expression is cell-cycle dependent
5. Experimental validation framework:
Manipulate HSPA1L levels (siRNA knockdown, overexpression)
Measure resulting changes in CSC properties (ALDH activity, sphere formation)
Assess therapeutic response before/after HSPA1L modification
Proper validation of FITC-conjugated HSPA1L antibody specificity is essential for generating reliable research data. A comprehensive validation strategy should include:
1. Essential negative controls:
Secondary antibody-only control (if using indirect detection method)
Isotype control antibody (matched to HSPA1L antibody host and isotype)
HSPA1L-knockdown cells (siRNA or CRISPR/Cas9)
Blocking peptide competition (pre-incubation with immunizing peptide)
Non-expressing tissue/cell line (based on RNA-seq or proteomics data)
2. Positive controls:
Heat-shocked cells (HSPA1L is stress-inducible)
HSPA1L-overexpressing cells (transfected with expression vector)
Tissues with documented expression (testis, certain cancer tissues)
3. Specificity validation experiments:
Western blot correlation: Confirm that IF pattern corresponds to WB band at expected MW (70 kDa)
Peptide competition: Pre-incubate antibody with immunizing peptide before staining
Cross-reactivity assessment: Test against related HSP70 family members
Multiple antibody validation: Compare staining pattern with alternative HSPA1L antibody clones
Orthogonal validation: Correlate protein expression with mRNA levels (RNA-FISH or RT-PCR)
4. Fluorophore-specific controls:
Autofluorescence control: Unstained sample to assess background
Spectral overlap control: When multiplexing with other fluorophores
Photobleaching rate determination: Serial imaging under standard conditions
pH sensitivity test: FITC fluorescence can be affected by environmental pH
5. Documentation requirements:
Antibody catalog number and lot number
Complete staining protocol with all parameters
Microscope settings (exposure time, gain, etc.)
Image processing steps
Quantification method
An exemplary validation approach would combine siRNA knockdown of HSPA1L with Western blot and immunofluorescence to demonstrate parallel reduction in signal across methods, confirming specificity of the FITC-conjugated antibody .