The HSPA1A Antibody, Biotin conjugated is a specialized immunological reagent designed for detecting the human heat shock protein 70 family member A (HSPA1A), also known as Hsp72. This antibody is covalently linked to biotin, a small molecule that facilitates signal amplification in immunoassays via streptavidin-biotin interactions. Its applications span molecular biology, cancer research, and stress response studies, with a focus on identifying HSPA1A in diverse cellular contexts.
HSPA1A is a 70 kDa molecular chaperone ubiquitously expressed in cells, primarily in the cytoplasm but also localized to the plasma membrane under stress or pathological conditions . It stabilizes nascent proteins, prevents aggregation, and regulates proteasomal degradation .
The biotin-conjugated HSPA1A antibody is typically polyclonal (rabbit) or monoclonal (mouse), with reactivity across human, mouse, rat, and occasionally other species (e.g., bovine, sheep) .
Biotin is attached via NHS-ester or similar chemistry to lysine residues on the antibody, enabling binding to streptavidin-alkaline phosphatase or horseradish peroxidase complexes. This enhances sensitivity in techniques like Western blot (WB), immunohistochemistry (IHC), and immunofluorescence (IF) .
BosterBio
Bio-Techne
Abbexa
Supplier | Host | Reactivity | Applications | Price (USD) |
---|---|---|---|---|
BosterBio | Rabbit | Hu/Ms/Rt | WB, FCM, IF, IHC | $370 |
Bio-Techne | Rabbit | Hu/Rt | WB, IHC, FCM | $409 |
Abbexa | Rabbit | Cow | WB, IHC, IF | $166–$235 |
HSPA1A’s surface embedding is regulated by phosphoinositides (PI(4)P, PI(3)P), as shown in cell surface biotinylation assays . Biotin-conjugated antibodies (e.g., Abbexa’s rabbit polyclonal) are critical for detecting this phenomenon .
Biotin treatment downregulates heat shock transcription factor 4b (HSF4b) activity, indirectly modulating HSPA1A expression . This highlights the role of biotinylated antibodies in studying stress pathways.
HSPA1A is overexpressed in tumors, where its membrane localization correlates with metastasis and drug resistance . Biotin-conjugated antibodies enable precise quantification of this biomarker in clinical samples.
HSPA1A is a gene encoding Heat Shock Protein 70 (HSP70), a molecular chaperone implicated in aging, viral replication, lupus, and tumor progression. It has been utilized in anti-tumor vaccine development and plays critical roles in cellular stress response pathways . The protein is primarily localized in the cytoplasm, specifically in cytoplasmic mRNP granules containing untranslated mRNAs .
HSPA1A Antibody, Biotin conjugated can be employed across multiple research applications including:
Western Blot (WB) for protein expression analysis
Immunohistochemistry (IHC) for tissue localization studies
Immunocytochemistry (ICC) for cellular distribution analysis
Flow Cytometry for quantitative cellular expression evaluation
Electron Microscopy for ultrastructural localization
The biotin conjugation provides significant signal amplification through avidin-biotin detection systems, making this antibody particularly valuable for detecting low abundance proteins or for studies requiring enhanced sensitivity in complex biological samples.
HSPA1A Antibody, Biotin conjugated demonstrates compatibility with diverse biological sample types including:
Multiple cell lines including:
Cancer tissues including:
Neural tissues including:
When working with these samples, researchers should optimize preparation protocols specific to their tissue or cell type. For example, different fixation methods may be required for optimal epitope preservation in neural tissues compared to cancer samples. Additionally, expression levels of HSPA1A vary significantly between sample types, which may necessitate adjustments in antibody concentration and detection methods to achieve optimal signal-to-noise ratios.
Optimal working dilutions for HSPA1A Antibody, Biotin conjugated vary by application technique:
For applications without specific recommendations, researchers should perform titration experiments to determine optimal concentration. This typically involves testing serial dilutions (e.g., 1:250, 1:500, 1:1000, 1:2000) against positive control samples. The ideal dilution provides maximum specific signal with minimal background.
Several factors influence optimal dilution, including:
Target protein abundance in the sample
Sample preparation method (fixation can reduce epitope accessibility)
Detection system sensitivity (HRP, fluorescent, or colorimetric)
Incubation conditions (time and temperature)
A systematic optimization approach should be documented to ensure reproducibility across experiments and sample types.
Proper storage and handling of HSPA1A Antibody, Biotin conjugated is critical for maintaining its integrity and performance across experiments. Follow these evidence-based guidelines:
Store lyophilized antibody at -20°C for up to one year from receipt date
After reconstitution, store at 4°C if using within one month
For longer storage after reconstitution, prepare small aliquots and store at -20°C for up to six months
Strictly avoid repeated freeze-thaw cycles which can damage antibody structure and reduce activity
When preparing working dilutions, use fresh, sterile buffers with appropriate preservatives
Centrifuge vials briefly before opening to ensure collection of all material
Verify the concentration on the vial label as this may vary between lots
Consider adding carrier proteins (e.g., BSA) to diluted antibodies to prevent adsorption to tube walls
Biotin conjugation introduces additional stability considerations. The biotin-streptavidin interaction remains one of the strongest non-covalent biological interactions, but exposure to extreme pH, organic solvents, or prolonged elevated temperatures should be avoided to maintain the integrity of both the antibody and the conjugated biotin.
HSPA1A Antibody, Biotin conjugated exhibits documented cross-reactivity with multiple species due to evolutionary conservation of the heat shock protein family:
This cross-reactivity is explained by sequence homology in the immunogen region. For example, some HSPA1A antibodies are generated against a synthetic peptide corresponding to a sequence at the C-terminus of human Hsp70 (577-596aa VISWLDANTLAEKDEFEHKR), which differs from the related mouse sequence by four amino acids and from the related rat sequence by three amino acids .
When working with species not explicitly listed, researchers can:
Compare sequence homology in the immunogen region across species of interest
Perform preliminary validation experiments with positive control samples
Include appropriate negative controls to verify specificity
While cross-reactivity enables comparative studies across species, researchers should note that signal intensity may vary due to species-specific sequence variations, and validation in each species is recommended for quantitative studies.
Rigorous validation of antibody specificity is essential for generating reliable data, particularly in complex systems where multiple HSP70 family members may be present. Implement this comprehensive validation strategy:
Sequence homology assessment:
Multi-level controls:
Positive controls: Include tissues/cells with confirmed high HSPA1A expression
Negative controls: Utilize HSPA1A-knockout models or siRNA-treated samples
Peptide competition: Pre-incubate antibody with immunogen peptide to confirm binding specificity
Isotype controls: Use biotin-conjugated non-specific antibodies of the same isotype
Orthogonal technique verification:
Application-specific validation:
For Western blot: Confirm appropriate molecular weight (70 kDa) and band pattern
For IHC/ICC: Compare staining patterns with published literature
For flow cytometry: Validate with positive/negative cell populations
Documentation and transparency:
Record complete validation protocols
Document lot-to-lot variation
Maintain validation data for reproducibility assessment
This systematic approach ensures that experimental findings truly reflect HSPA1A biology rather than technical artifacts or cross-reactivity with related proteins.
Non-specific binding can significantly compromise experimental results when working with biotin-conjugated antibodies. Address this methodically:
Biotin-specific interference management:
Block endogenous biotin using commercial avidin/biotin blocking kits
Note tissues with naturally high biotin content (liver, kidney, brain)
Implement longer blocking steps (60+ minutes) before primary antibody application
Consider using streptavidin instead of avidin (less non-specific binding)
Buffer optimization strategy:
Test increasing salt concentrations (150-500 mM NaCl) to reduce ionic interactions
Add non-ionic detergents (0.1-0.3% Triton X-100) to reduce hydrophobic binding
Incorporate carrier proteins (1-5% BSA or non-fat milk) to compete for non-specific interactions
Adjust pH slightly (±0.5 units) to modify charge-based interactions
Antibody dilution refinement:
Detection system modifications:
Test alternative streptavidin conjugates (different manufacturers)
Reduce streptavidin-conjugate concentration
Extend wash steps (4-5 washes, 5+ minutes each)
Apply gradient elution with increasing salt concentration
Sample-specific adaptations:
For fixed tissues: Optimize fixation time and antigen retrieval methods
For cell lysates: Adjust detergent concentration in lysis buffers
For flow cytometry: Include viability dyes to exclude dead cells
Systematic elimination approach:
Compare secondary-only and isotype controls to identify source of background
Test each component individually (blockers, buffers, detection reagents)
Document successful modifications for future reference
Implementing these strategies systematically will identify the source of non-specific binding and establish reliable protocols for specific experimental systems.
Detecting HSPA1A in challenging tissues requires specialized protocols to overcome inherent difficulties such as high background, variable target expression, and complex tissue architecture:
For Brain Tissue:
Fixation optimization:
Enhanced antigen retrieval:
Heat-induced epitope retrieval in citrate buffer (pH 6.0) for 30-40 minutes
Allow gradual cooling in retrieval solution (20-30 minutes)
For difficult samples, test pH 9.0 EDTA buffer as an alternative
Background reduction for neural tissue:
Pre-treat sections with Sudan Black B (0.1%) to reduce lipofuscin autofluorescence
Extend blocking time to 2+ hours with 10% normal serum + 0.3% Triton X-100
Include 0.1% Tween-20 in all antibody diluents
For Tumor Samples:
Sample selection strategy:
Include tumor core and margin for comparative analysis
Process samples consistently (fixation time, processing protocol)
Consider tissue microarrays for high-throughput analysis
Signal enhancement approaches:
Implement tyramide signal amplification for low-expressing tumors
Use polymer-based detection systems rather than ABC method
Consider multi-step amplification for poorly differentiated tumors
Cancer-specific controls:
Include adjacent normal tissue as internal control
Use known HSPA1A-expressing cancer cell lines as positive controls
Add tissue-matched negative controls (antibody omission, isotype control)
These specialized protocols address the documented challenges in detecting HSPA1A in neural tissues (including Cajal-Retzius cells and fetal brain cortex ) and various cancer types (including colon carcinoma and cervix carcinoma ).
Multiplex immunoassays provide comprehensive insights into cellular contexts. Incorporate HSPA1A Antibody, Biotin conjugated effectively using these strategic approaches:
Panel design principles:
Combine with antibodies raised in different host species
Ensure targets have distinct cellular distributions or expression levels
Consider size differences for multiplex Western blot applications
Test for potential cross-reactivity between primary antibodies
Detection system optimization:
For immunofluorescence:
Pair with streptavidin conjugated to fluorophores with minimal spectral overlap
Consider far-red fluorophores to minimize tissue autofluorescence
Implement sequential scanning for confocal microscopy
For chromogenic detection:
Use enzyme systems with distinct substrates (HRP vs. AP)
Apply spectral unmixing algorithms for quantification
Consider sequential development with intermediate fixation
Sequential labeling methodology:
First round: Apply HSPA1A Antibody, Biotin conjugated → Detect with streptavidin conjugate → Fix
Between rounds: Apply microwave treatment or glycine elution to remove primary antibodies
Subsequent rounds: Apply additional antibodies → Detect with non-biotin systems
Technical optimizations:
Increase wash duration and volume between steps
Apply higher blocking reagent concentrations
Optimize each antibody individually before combining
Consider antibody order of application (start with lowest abundance target)
Analysis considerations:
Implement proper image registration for sequential multiplexing
Apply spectral unmixing for overlapping signals
Include single-stain controls for each fluorophore
Use software algorithms for co-localization analysis
This structured approach allows researchers to leverage biotin-streptavidin signal amplification while addressing the specific challenges of multiplex experimental design in both imaging and quantitative applications.
Fixation critically impacts epitope preservation and accessibility. For HSPA1A detection with biotin-conjugated antibodies:
Recommended fixatives:
Paraformaldehyde (PFA) is preferred over formalin due to superior tissue penetration
Prepare fresh 4% PFA solution to avoid formaldehyde formation during storage
Avoid methanol-based fixatives which can disrupt protein conformation
Consider zinc-based fixatives for specialized applications requiring enhanced antigen preservation
Fixation parameters:
Duration optimization:
Cell cultures: 10-15 minutes at room temperature
Tissue sections (≤5mm): 24-48 hours at 4°C
Perfusion fixation: 4% PFA followed by 24-hour post-fixation
Critical conditions:
Maintain pH 7.2-7.4 during fixation
Process tissues at 4°C to minimize protein degradation
Use phosphate-buffered fixatives for optimal pH stability
Tissue-specific adaptations:
For brain tissue: Add 4% sucrose to PFA to improve morphology
For tumor tissue: Limit fixation time to 24 hours to reduce epitope masking
For highly fibrous tissues: Consider gentle pre-digestion with proteinase K
Post-fixation processing:
Transfer to 70% ethanol rather than extended storage in fixative
For paraffin embedding, maintain temperatures ≤60°C to preserve epitopes
Store cut sections at 4°C and use within one month for optimal results
Antigen retrieval optimization:
Test multiple antigen retrieval methods:
Heat-induced (citrate buffer pH 6.0 and EDTA buffer pH 9.0)
Enzymatic (proteinase K for certain applications)
Optimize duration (10-30 minutes) and temperature
Allow gradual cooling in retrieval solution
These fixation recommendations address the critical balance between structural preservation and epitope accessibility for optimal HSPA1A detection, based on documented successful applications in various tissue types .
HSPA1A expression exhibits cancer type-specific patterns that necessitate tailored detection approaches:
Expression patterns in major cancer types:
Cervical carcinoma: Shows elevated expression with predominantly cytoplasmic localization
Colon carcinoma: Demonstrates increased expression particularly in advanced stages
Pancreatic cancer: Exhibits strong cytoplasmic staining with significant heterogeneity
Brain tumors: Displays variable expression depending on grade and histological type
Cancer-specific detection strategies:
For high-expressing cancers:
For heterogeneous tumors:
Sample multiple regions within tumor
Consider single-cell resolution techniques
Implement digital pathology with cellular segmentation
Subcellular localization considerations:
Technical adaptations:
Optimize fixation time based on tissue density
Adjust antigen retrieval conditions for specific tumor types
Implement tiered dilution strategy for comparative studies
Consider laser microdissection for region-specific analysis
Biomarker application considerations:
Establish threshold values for overexpression in each cancer type
Compare with matched normal tissue controls
Correlate with clinical parameters
Evaluate prognostic/predictive potential
Understanding these cancer-specific expression patterns enables researchers to develop optimized detection protocols and correctly interpret HSPA1A staining patterns in the context of tumor biology, particularly important given its role in tumor progression .
Robust quantitative analysis requires comprehensive controls to ensure accuracy and validity:
Essential experimental controls:
Positive controls:
Negative controls:
Isotype-matched, biotin-conjugated non-specific antibody
Primary antibody omission
Competitive blocking with immunogen peptide
HSPA1A-knockdown or knockout samples (when available)
Technical controls for biotin-specific considerations:
Endogenous biotin blocking validation:
Compare staining with and without avidin-biotin blocking
Include known biotin-rich tissues as internal controls
Signal amplification calibration:
Titrate streptavidin-detection reagent
Include standard curves for absolute quantification
Determine linear dynamic range of detection system
Quantitation-specific controls:
Normalization standards:
Housekeeping proteins (β-actin, GAPDH) for Western blot
Internal reference tissues for IHC/IF
Spike-in standards for absolute quantification
Technical replication:
Intra-assay replicates (minimum triplicate measurements)
Inter-assay validation across multiple experiments
Multi-operator verification for subjective scoring methods
Application-specific controls:
For Western blot:
Complete molecular weight marker documentation
Loading controls with similar abundance to target
Serial dilutions to establish linearity
For IHC/IF:
Multiple field quantification (minimum 5-10 fields)
Background subtraction validation
Digital vs. manual scoring comparison
Implementing this comprehensive control strategy ensures that quantitative data generated using HSPA1A Antibody, Biotin conjugated is reliable, reproducible, and scientifically valid across different experimental platforms and conditions.
Distinguishing between constitutive (HSC70/HSPA8) and inducible (HSP70/HSPA1A) forms requires careful experimental design:
Antibody selection strategy:
Verify immunogen specificity to HSPA1A (inducible form)
The immunogen for many HSPA1A antibodies targets the C-terminal region (residues 600-641 or 577-596 ) which differs from constitutive HSP70 forms
Consider parallel detection with antibodies specific to constitutive forms
Validate antibody specificity using recombinant proteins or knockout samples
Experimental induction protocols:
Stress induction methods:
Heat shock (42°C for 1-2 hours)
Heavy metals (cadmium chloride, arsenite)
Proteasome inhibitors (MG132, bortezomib)
Oxidative stress inducers (hydrogen peroxide, paraquat)
Time-course analysis:
Baseline (pre-stress)
Early induction (2-6 hours post-stress)
Peak expression (6-12 hours)
Resolution phase (24-48 hours)
Detection and discrimination techniques:
Western blot optimization:
Use high-resolution SDS-PAGE (8-10% gels) for isoform separation
Employ gradient gels for enhanced discrimination
Quantify relative induction compared to constitutive forms
Immunofluorescence approaches:
Dual-labeling with isoform-specific antibodies
Analyze subcellular distribution patterns
Quantify intensity changes in stressed vs. unstressed cells
Molecular validation methods:
Correlate protein detection with mRNA expression (RT-qPCR)
Employ isoform-specific siRNA knockdown
Use mass spectrometry for peptide-level confirmation
Functional correlation studies:
Link expression patterns to chaperone activity assays
Correlate with stress granule formation
Monitor association with client proteins
This multifaceted approach enables researchers to reliably distinguish between constitutive and inducible HSP70 forms, critical for understanding stress response mechanisms and therapeutic targeting strategies in various pathological conditions.
Dual labeling experiments with HSPA1A Antibody, Biotin conjugated require methodical planning to ensure valid co-localization data:
Detection system compatibility planning:
Avidin/Streptavidin conjugate selection:
Choose fluorophores with minimal spectral overlap with second target
Consider far-red fluorophores to minimize tissue autofluorescence
For chromogenic detection, pair HRP-streptavidin with AP-conjugated secondary
Secondary antibody selection for co-target:
Protocol optimization:
Sequential vs. simultaneous approach:
Sequential recommended for biotin-conjugated antibodies
Complete HSPA1A detection before introducing second primary
Consider intermediate fixation step (2% PFA for 10 minutes)
Enhanced blocking strategy:
Implement avidin-biotin blocking before primary antibody
Extend blocking time (2+ hours) with 5-10% normal serum
Include detergent (0.1-0.3% Triton X-100) for membrane permeabilization
Signal balancing methods:
Adjust antibody concentrations to achieve comparable signal intensities
Optimize exposure/gain settings for each channel
Consider signal amplification for weaker target
Establish detection thresholds before quantitative analysis
Advanced imaging considerations:
Acquisition parameters:
Use sequential scanning to prevent bleed-through
Match optical section thickness between channels
Implement Nyquist sampling criteria for co-localization studies
Analysis methodologies:
Apply appropriate co-localization metrics (Pearson's, Manders' coefficients)
Utilize threshold-based co-localization
Implement object-based rather than pixel-based analysis for discrete structures
Validation approaches:
Include single-labeled controls for each target
Perform antibody reversal experiments (switch application order)
Test multiple fixation and permeabilization protocols
Compare results with proximity ligation assays for protein interaction studies
These methodological considerations ensure that dual labeling experiments with HSPA1A Antibody, Biotin conjugated yield reliable and meaningful co-localization data while minimizing technical artifacts common in multiplex immunodetection studies.
HSPA1A Antibody, Biotin conjugated offers versatile applications for investigating functional aspects of stress response pathways:
Dynamic expression analysis:
Temporal profiling protocol:
Apply stressor (heat, oxidative, chemical)
Collect samples at multiple timepoints (0, 2, 6, 12, 24, 48 hours)
Process matched samples for protein and mRNA analysis
Correlate protein levels with functional readouts
Subcellular translocation tracking:
Protein interaction network mapping:
Proximity ligation methodology:
Combine HSPA1A Antibody, Biotin conjugated with antibodies against potential interactors
Visualize protein-protein interactions (<40nm proximity)
Quantify interaction dynamics under various stress conditions
Co-immunoprecipitation protocol:
Utilize streptavidin-coated magnetic beads for pull-down
Identify stress-specific binding partners
Validate interactions with reverse co-immunoprecipitation
Stress response pathway interrogation:
Inhibition studies:
Client protein folding analysis:
Track association with misfolded proteins
Monitor aggregate clearance efficiency
Correlate with cell survival metrics
Translational research applications:
Cancer therapeutic response studies:
Stress adaptation mechanisms:
Compare acute vs. chronic stress responses
Analyze hormetic conditioning effects
Investigate cross-tolerance between stressors
This comprehensive functional research approach leverages the specificity and signal amplification capabilities of HSPA1A Antibody, Biotin conjugated to gain mechanistic insights into stress response pathways across diverse physiological and pathological contexts.