HSPA8 antibodies target the constitutively expressed HSPA8 protein (also termed HSC70), which functions as a molecular chaperone involved in protein folding, autophagy, and immune regulation . These antibodies are widely used in basic research and clinical diagnostics to investigate HSPA8's role in diseases such as cancer, autoimmune disorders, and neurodegenerative conditions .
HSPA8 regulates key cellular processes:
Protein Quality Control: Facilitates folding and refolding of misfolded proteins via ATP-dependent mechanisms .
Chaperone-Mediated Autophagy (CMA): Acts as a gatekeeper by identifying substrates for lysosomal degradation .
Immune Regulation: Modulates antigen presentation via MHC class II molecules and influences T-cell activation .
Cancer Progression: Overexpressed in malignancies like triple-negative breast cancer (TNBC), correlating with tumor growth and drug resistance .
HSPA8 antibodies are utilized in diverse experimental workflows:
Cancer Biomarker: HSPA8 overexpression in TNBC correlates with advanced TNM stage and poor prognosis .
Therapeutic Target: Pharmacologic inhibitors (e.g., VER-155008) block HSPA8’s amyloidase activity, enhancing chemotherapy-induced necroptosis in cancer cells .
Autoimmune Diseases: HSPA8 dysregulation is linked to systemic lupus erythematosus (SLE), with antibodies aiding in mechanistic studies .
| Cancer Type | HSPA8 Role | Therapeutic Insight |
|---|---|---|
| Triple-Negative Breast Cancer | Overexpression linked to poor prognosis | Predicts response to necroptosis-inducing therapies |
| Fibrosarcoma | HSPA8 inhibition enhances MTA efficacy | Reduces required drug doses, minimizing toxicity |
Current research focuses on:
HSPA8 (Heat Shock Protein Family A Member 8), also known as HSC70 or HSP73, is a constitutively expressed molecular chaperone that plays critical roles in multiple cellular processes. Unlike inducible heat shock proteins, HSPA8 is expressed under normal conditions and is crucial for:
Protein quality control, ensuring proper folding and refolding of selected proteins
Regulation of chaperone-mediated autophagy (CMA) as a detector of substrates
Anti-bacterial autophagy through interaction with proteins like RHOB and BECN1
Prevention of necroptosis by acting as an amyloidase that inhibits RHIM-amyloid formation
The importance of HSPA8 in research stems from its involvement in fundamental cellular processes and its dysregulation in various disease states, including autoimmune disorders and bacterial infections.
Several types of HSPA8 antibodies are available for research applications, varying in their target regions, host species, and recommended applications:
When selecting an HSPA8 antibody, researchers should consider:
Target region of interest (N-terminal vs. C-terminal epitopes)
Required applications (some antibodies perform better in specific applications)
Species cross-reactivity (particularly important for comparative studies)
For optimal Western blot results with HSPA8 antibodies, follow these methodological guidelines:
Sample Preparation:
Use appropriate lysis buffers containing protease inhibitors
Load 25-35 μg of total protein per lane for cell lines (as demonstrated in validated protocols)
Running Conditions:
Use reducing conditions with standard SDS-PAGE (typically 10-12% gels)
Include positive control samples (validated cell lines include HeLa, A431, NIH/3T3)
Antibody Dilutions:
Primary antibody: 1:1000 dilution for most commercial HSPA8 antibodies
Secondary antibody: typically 1:5000 dilution (HRP-conjugated anti-rabbit or anti-mouse IgG)
Detection:
Allow sufficient exposure time (HSPA8 is generally abundant but may vary by cell type)
For reproducible results, standardize protein loading using housekeeping protein controls
Note that HSPA8 is highly conserved across species, allowing most antibodies to detect human, mouse, and rat orthologs with similar efficiency .
For optimal immunofluorescence results when detecting HSPA8:
Cell Preparation:
Use appropriate fixation: 4% paraformaldehyde (10-15 minutes) works well for most applications
Permeabilization: 0.1-0.2% Triton X-100 for 5-10 minutes at room temperature
Staining Protocol:
Blocking: 1-5% BSA or normal serum from secondary antibody host species (1 hour at room temperature)
Primary antibody concentration: 10 μg/mL for the mouse monoclonal anti-HSPA8/HSC71 antibody
Incubation time: 3 hours at room temperature or overnight at 4°C
Secondary antibody: fluorophore-conjugated anti-mouse or anti-rabbit IgG (depending on primary)
Expected Localization Pattern:
Primarily cytoplasmic distribution
Nuclear localization, especially in nucleoli
Potential punctate structures under stress conditions or specific experimental treatments
Co-staining Recommendations:
DAPI for nuclear counterstaining
Phalloidin for actin filament visualization to provide cellular context
For autophagy studies, consider co-staining with LC3 or LAMP2A
Validated examples show clear nuclear and nucleolar localization in HeLa cells when using mouse anti-HSPA8/HSC71 at 10 μg/mL with NorthernLights 557-conjugated secondary antibody .
HSPA8 plays a crucial role in anti-bacterial autophagy through its interactions with RHOB and BECN1. To effectively study this function:
Experimental Design:
Bacterial Infection Models:
HSPA8 Manipulation Approaches:
siRNA knockdown of HSPA8 (validated in both cell culture and mouse models)
Overexpression of wild-type HSPA8 or domain-specific mutants (particularly IDR domain mutants)
Pharmacological modulators of HSPA8 function
Readout Assays:
Quantification of intracellular bacteria (CFU assays, immunofluorescence)
LC3-decoration of bacteria (co-localization studies)
Autophagy marker analysis (LC3-II, BECN1, SQSTM1 levels by Western blot)
Methodological Example:
Research has shown that HSPA8 knockdown significantly impairs bacterial clearance. This can be assessed by:
Transfecting cells with HSPA8-specific siRNAs
Infecting with bacteria (e.g., S. typhi LT2 or UPEC CFT073)
Measuring bacterial load at different time points
Additionally, you can study the mechanism by examining:
HSPA8-RHOB-BECN1 complex formation using co-immunoprecipitation
Domain-specific interactions using truncated constructs
LLPS (liquid-liquid phase separation) properties using fluorescently-tagged proteins
Research has demonstrated that HSPA8, through its IDR domain (residues 243-283), promotes LLPS to concentrate RHOB and BECN1, enhancing their interaction and stabilization .
Studying HSPA8's role in CMA requires specific methodological approaches:
Experimental System Selection:
Cell lines with high CMA activity (e.g., fibroblasts, hepatocytes)
Tissues with notable CMA dependency (liver, brain, immune cells)
CMA Substrate Tracking:
Use established CMA substrates containing KFERQ-like motifs (e.g., GAPDH, RNase A)
Generate fluorescently-tagged CMA substrate reporters
Employ pulse-chase experiments to track substrate degradation
HSPA8-LAMP2A Interaction Studies:
Co-immunoprecipitation assays to assess HSPA8-LAMP2A binding
Proximity ligation assays for in situ visualization of interactions
FRET/BRET approaches for real-time interaction monitoring
CMA Activity Assessment:
Lysosomal isolation and in vitro uptake assays
Selective degradation of known CMA substrates
LAMP2A multimerization analysis
Specific Methodology Example:
To determine if a protein is degraded via CMA involving HSPA8:
Generate cells with HSPA8 knockdown or overexpression
Measure half-life of candidate CMA substrates in presence/absence of lysosomal inhibitors
Perform competition assays with known KFERQ-containing peptides
Assess direct binding of the substrate to HSPA8 using purified proteins
Evaluate LAMP2A dependency through selective LAMP2A knockdown
Remember that HSPA8 recognizes and delivers KFERQ-motif containing proteins with the help of co-chaperones for degradation via macroautophagy, CMA, or endosomal microautophagy (eMI) pathways .
Inconsistent HSPA8 staining patterns across different cell types can result from several factors:
Biological Factors:
Differential Expression Levels:
Subcellular Localization Differences:
HSPA8 shuttles between cellular compartments depending on cell state
Stress conditions can alter localization patterns
Interaction with different partners affects distribution
Post-translational Modifications:
Phosphorylation state affects localization and function
Ubiquitination can alter stability and detection
Technical Considerations:
Fixation Method Impact:
Paraformaldehyde vs. methanol fixation reveals different epitopes
Overfixation can mask epitopes, particularly affecting N-terminal antibodies
Antibody Specificity:
N-terminal antibodies (aa 82-110) may detect different conformations than C-terminal antibodies (aa 534-615)
Some antibodies may recognize specific functional states
Protocol Optimization Requirements:
Cell-type specific permeabilization conditions
Blocking protocol adjustments
Primary antibody concentration titration
Recommendations for Standardization:
Include positive control cell lines (HeLa cells show consistent nuclear/nucleolar staining)
Optimize fixation time for each cell type
Consider dual-antibody approaches (N-terminal and C-terminal antibodies) for complete picture
When comparing cell types, process and image them under identical conditions
Distinguishing HSPA8 from other HSP70 family members is crucial for accurate data interpretation due to their high sequence homology. Here's a methodological approach:
Antibody Selection Strategy:
Epitope Targeting:
Select antibodies raised against unique regions (typically C-terminal domains)
Verify epitope specificity against sequence alignments of HSP70 family members
Examine published validation data showing specificity testing
Validation Using Knockout/Knockdown Controls:
Include HSPA8 knockout or knockdown samples as negative controls
Test antibody reactivity against recombinant HSPA8 and other HSP70 proteins
Experimental Approaches:
Western Blot Discrimination:
Use gradient gels (8-12%) to resolve subtle size differences
Include positive controls for other HSP70 family members
Consider 2D gel electrophoresis to separate based on both MW and pI
Immunoprecipitation Specificity:
Perform IP-Mass Spectrometry to confirm identity
Use stringent washing conditions to minimize cross-reactivity
Expression Pattern Analysis:
HSPA8 is constitutively expressed, while HSPA1A/B are stress-inducible
Examine expression under normal vs. stress conditions
Mass Spectrometry Approach:
For definitive identification, use MS-based methods:
Immunoprecipitate with HSPA8 antibody
Digest and analyze by LC-MS/MS
Identify unique peptides specific to HSPA8 vs. other family members
Documented Distinction Methods:
Research has demonstrated effective discrimination through:
Using antibodies targeting the C-terminal region (aa 534-615)
Conducting parallel knockdown experiments for verification
Employing isoform-specific PCR to correlate protein with mRNA expression
LLPS is an emerging mechanism in cellular biology, and HSPA8 has recently been identified as a key player in LLPS-mediated autophagy regulation. Here's how to investigate this phenomenon:
Experimental Approaches:
In Vitro Phase Separation Assays:
Purify recombinant HSPA8 protein (full-length and domain variants)
Monitor droplet formation using microscopy under various conditions (protein concentration, salt, pH)
Use fluorescently-labeled HSPA8 to visualize droplet dynamics
Cellular LLPS Visualization:
Express fluorescently-tagged HSPA8 constructs (GFP-HSPA8)
Employ live-cell imaging to monitor droplet formation
Test HSPA8 mutants (particularly IDR domain mutants) for LLPS capability
HSPA8-Mediated Concentration of Autophagy Factors:
Co-express tagged RHOB and BECN1 with HSPA8
Quantify co-localization in liquid-like condensates
Assess functional consequences through autophagy assays
Methodological Details:
Research has demonstrated that HSPA8 contains predicted intrinsically disordered regions (IDRs) and drives LLPS to concentrate RHOB and BECN1 into HSPA8-formed liquid-phase droplets . To study this:
LLPS Demonstration:
Functional Impact Assessment:
Generate HSPA8 constructs lacking the IDR domain (HSPA8Δ243-283)
Compare wild-type and mutant HSPA8 for:
Ability to form condensates
Capacity to concentrate RHOB and BECN1
Effect on bacterial clearance
Client Protein Concentration:
Quantify BECN1 and RHOB recruitment to HSPA8 droplets
Measure interaction strength using FRET or biochemical assays
Correlate condensate formation with autophagy induction
Research has shown that HSPA8 lacking the IDR domain failed to form puncta and was unable to effectively concentrate RHOB and BECN1, resulting in impaired bacterial clearance .
HSPA8 has been implicated in various immune disorders, particularly autoimmune diseases like systemic lupus erythematosus. Here are methodological approaches using HSPA8 antibodies:
Cellular Distribution Analysis:
Immune Cell Phenotyping:
Use flow cytometry with HSPA8 antibodies to quantify expression across immune cell subsets
Compare HSPA8 levels between healthy controls and disease samples
Correlate with disease activity markers
Tissue-Specific Expression:
Perform IHC on tissue sections from affected organs
Quantify HSPA8 expression in specific cell types within the tissue
Co-stain with cell type markers and disease-associated proteins
Functional Studies:
Antigen Presentation Role:
Assess HSPA8 involvement in MHC class II presentation pathway
Study co-localization with processed antigens in antigen-presenting cells
Analyze effect of HSPA8 modulation on T cell activation
Autophagy Regulation in Immune Cells:
Measure CMA activity in different immune cell populations
Correlate with HSPA8 expression and localization
Determine impact on immune cell function and survival
Disease-Specific Applications:
Research has shown that in lupus-prone mice, HSPA8 expression is elevated in multiple immune cell types . To investigate this:
Comparative Expression Analysis:
Use flow cytometry with HSPA8 antibodies to quantify surface and intracellular expression
Compare B cells, T cells, activated T cells, and CD11b+Gr-1+ granulocytes/macrophages
Correlate with mRNA expression levels
Mechanistic Investigation:
Assess HSPA8's role in autoantigen presentation
Determine if HSPA8 modulation affects autoantibody production
Study HSPA8-client protein interactions in disease states
Therapeutic Target Validation:
Test HSPA8-targeting compounds in disease models
Monitor immune parameters and disease progression
Identify specific pathways affected by HSPA8 modulation
These approaches can provide valuable insights into HSPA8's role in immune disorders and potentially identify new therapeutic strategies targeting this chaperone protein .
This FAQ collection represents the current state of knowledge regarding HSPA8 antibodies and their applications in research. As scientific understanding continues to evolve, researchers are encouraged to:
Validate antibodies in their specific experimental systems
Consider the growing role of HSPA8 in cellular phase transitions and compartmentalization
Explore the therapeutic potential of targeting HSPA8 in infectious and autoimmune diseases
Employ complementary approaches (genetic, biochemical, imaging) for comprehensive insights into HSPA8 function