HSPA5, also known as GRP78, BiP, or endoplasmic reticulum chaperone BiP, is a 72.3 kilodalton heat shock protein encoded by the HSPA5 gene in humans. It belongs to the heat shock protein 70 (HSP70) family and functions primarily as a molecular chaperone in the endoplasmic reticulum (ER). HSPA5 is critical for protein folding, assembly, and quality control in the ER, making it a central player in the unfolded protein response (UPR) and ER stress pathways . Recent research has also revealed its essential role in embryonic development, particularly in pronephros formation through mediation of retinoic acid signaling . The protein's involvement in cellular stress responses, development, and multiple disease states makes it a high-value research target across various biological disciplines.
Selecting the optimal HSPA5 antibody requires consideration of multiple experimental factors:
Application compatibility: Determine which applications (WB, IHC, ICC, IF, etc.) the antibody has been validated for. Many HSPA5 antibodies are validated for Western Blot, while fewer may be validated for specialized applications like ChIP .
Species reactivity: Confirm the antibody's reactivity with your experimental model. Many HSPA5 antibodies react with human, mouse, and rat samples, but species-specific variability exists .
Specific epitope recognition: Determine which domain or region of HSPA5 the antibody targets. Some antibodies target the middle region while others target specific amino acid sequences (e.g., 505-570 aa) .
Antibody format: Consider whether you need an unconjugated antibody or one conjugated to a detection tag (biotin, Cy3, DyLight488) based on your experimental design .
Citations and validation: Review published research using the antibody to assess its performance in contexts similar to your planned experiments.
The choice between monoclonal and polyclonal HSPA5 antibodies depends on your specific research requirements:
Monoclonal HSPA5 Antibodies:
Recognize a single epitope on the HSPA5 protein
Provide high specificity and consistent lot-to-lot reproducibility
Often preferred for applications requiring precise epitope targeting
Typically demonstrate lower background signal in applications like IHC
Examples include BiP (C50B12) Rabbit mAb from Cell Signaling Technology
Polyclonal HSPA5 Antibodies:
Recognize multiple epitopes on the HSPA5 protein
Offer potentially higher sensitivity due to multiple binding sites
Can be advantageous when protein conformation might mask certain epitopes
Typically less expensive to produce
Examples include numerous options from suppliers like Novus Biologicals and R&D Systems
For critical quantitative experiments where reproducibility is paramount, monoclonal antibodies typically provide more consistent results. For applications like IHC where signal amplification might be beneficial, polyclonal antibodies often perform well.
Optimizing Western blot protocols for HSPA5 detection requires attention to several technical factors:
Sample preparation: HSPA5 is predominantly located in the ER lumen, so proper cell lysis with detergents capable of solubilizing membrane proteins (e.g., RIPA buffer with 1% NP-40 or Triton X-100) is essential.
Protein loading: Load 20-40 μg of total protein per lane for cell lysates; HSPA5 is typically abundant in most cell types, especially under conditions of ER stress.
Gel percentage: Use 10% SDS-PAGE gels for optimal resolution of the 72.3 kDa HSPA5 protein .
Transfer conditions: Wet transfer at 100V for 60-90 minutes or overnight at 30V at 4°C is generally effective for complete transfer of HSPA5.
Blocking: 5% non-fat dry milk in TBST for 1 hour at room temperature works well for most HSPA5 antibodies.
Primary antibody dilution: Typically 1:1000 to 1:2000 for most commercial HSPA5 antibodies, but always refer to the manufacturer's recommended dilution .
Secondary antibody: Use HRP-conjugated secondary antibodies at 1:5000 to 1:10,000 dilution.
Expected band size: HSPA5 will appear at approximately 72-78 kDa depending on the gel system used .
Successful IHC detection of HSPA5 requires tissue-specific optimization:
Fixation: 10% neutral buffered formalin fixation for 24-48 hours is suitable for most tissues; overfixation can mask the HSPA5 epitope.
Antigen retrieval: Heat-induced epitope retrieval using citrate buffer (pH 6.0) for 15-20 minutes is effective for most HSPA5 antibodies.
Permeabilization: For paraffin sections, standard deparaffinization and rehydration is typically sufficient; for frozen sections, 0.1-0.3% Triton X-100 for 10 minutes may enhance antibody penetration.
Blocking: 5-10% normal serum from the same species as the secondary antibody for 1 hour at room temperature.
Primary antibody incubation:
Detection system: For low expression tissues, amplification systems like tyramide signal amplification may improve sensitivity.
Counterstaining: Hematoxylin counterstaining can help visualize tissue architecture while maintaining HSPA5 signal visibility.
Controls: Include both positive controls (tissues known to express HSPA5, such as liver or pancreas) and negative controls (primary antibody omission).
Reducing non-specific binding in immunofluorescence requires methodical optimization:
Fixation optimization: Test multiple fixatives (4% PFA, methanol, acetone) as HSPA5 epitope accessibility can vary with fixation method.
Enhanced blocking: Use a combination of 5-10% normal serum with 1-3% BSA and 0.1-0.3% Triton X-100 in PBS for 1-2 hours at room temperature.
Antibody dilution optimization: Test a dilution series (1:100, 1:250, 1:500, 1:1000) to identify the optimal signal-to-noise ratio for your specific HSPA5 antibody .
Secondary antibody controls: Always include a secondary-only control to assess non-specific binding from the secondary antibody.
Cross-adsorbed secondary antibodies: Use highly cross-adsorbed secondary antibodies to minimize species cross-reactivity.
Buffer optimization: Adding 0.05% Tween-20 to wash buffers can reduce background without compromising specific signal.
Autofluorescence reduction: For tissues with high autofluorescence (liver, kidney), consider treatment with Sudan Black B (0.1-0.3% in 70% ethanol) for 20 minutes after secondary antibody incubation.
Sequential double staining: When performing double immunofluorescence, use sequential rather than simultaneous antibody incubations to minimize cross-reactivity.
Accurate identification of HSPA5-specific bands requires analytical rigor:
Molecular weight verification: The primary HSPA5 band should appear at approximately 72-78 kDa. Any significant deviation suggests potential non-specific binding or protein degradation .
Positive controls: Include lysates from cells known to express high levels of HSPA5 (e.g., HeLa cells treated with thapsigargin to induce ER stress).
Validation with multiple antibodies: Test at least two antibodies targeting different HSPA5 epitopes to confirm band identity.
Knockout/knockdown controls: If possible, include samples from HSPA5 knockdown or knockout models as negative controls. HSPA5 shRNA knockdown systems have been described in published literature .
Competition assays: Pre-incubation of the antibody with purified HSPA5 protein should eliminate specific bands.
Gradient gels: Using 4-15% gradient gels can improve resolution around the HSPA5 molecular weight range.
Alternative detection methods: For ambiguous results, consider immunoprecipitation followed by mass spectrometry for definitive identification.
Several artifacts commonly confound HSPA5 IHC interpretation:
Edge effects: Increased staining at tissue edges represents antibody trapping rather than specific HSPA5 expression. Mitigation: Ensure thorough washing and avoid tissue drying during the protocol.
Necrotic tissue staining: Non-specific antibody binding to necrotic areas. Mitigation: Exclude necrotic regions from analysis or use dual staining with necrotic markers.
Nuclear staining artifacts: While HSPA5 is primarily located in the ER, non-specific nuclear staining can occur. Mitigation: Validate with subcellular fractionation and Western blotting.
Melanin interference: In pigmented tissues, endogenous melanin can be mistaken for DAB signal. Mitigation: Use Vector VIP or AEC as alternative chromogens.
Overfixation masking: Excessive fixation can cross-link proteins and mask epitopes. Mitigation: Optimize fixation time and use appropriate antigen retrieval methods.
Endogenous peroxidase activity: Particularly in tissues rich in peroxidases (liver, kidney). Mitigation: Include a hydrogen peroxide quenching step (0.3-3% H₂O₂ in methanol for 10-30 minutes).
Endogenous biotin interference: When using biotin-based detection systems. Mitigation: Block endogenous biotin with avidin/biotin blocking kit or switch to polymer-based detection.
When facing discrepancies between techniques, systematic troubleshooting is essential:
Epitope accessibility differences: HSPA5 epitopes may be differentially accessible in native (IHC/IF) versus denatured (WB) states. Solution: Try antibodies targeting different HSPA5 epitopes or domains.
Fixation and processing effects: Formalin fixation modifies protein structure differently than sample preparation for Western blot. Solution: Test alternative fixatives or fresh frozen samples.
Sensitivity threshold variations: Western blot may detect lower HSPA5 levels than IHC. Solution: Use amplification systems for IHC or more sensitive Western blot detection methods.
Isoform-specific detection: Antibodies may detect specific HSPA5 isoforms or post-translational modifications. Solution: Review antibody documentation for isoform specificity and use isoform-specific positive controls.
Cross-reactivity with related proteins: HSPA5 belongs to the HSP70 family, which contains highly homologous members. Solution: Perform specificity tests using recombinant proteins from related family members.
Quantitative vs. qualitative assessment: Western blot provides semi-quantitative data while IHC is often qualitative. Solution: Use quantitative image analysis for IHC and validate with complementary techniques like qPCR.
Technical validation approach: When critical discrepancies persist, orthogonal methods like RNA in situ hybridization or mass spectrometry can provide technique-independent validation.
HSPA5 antibodies serve as powerful tools for dissecting UPR dynamics in disease:
UPR activation monitoring: HSPA5 upregulation is a reliable marker of UPR activation. Western blot analysis using HSPA5 antibodies can quantify this upregulation across disease models .
Temporal analysis: Time-course studies using HSPA5 antibodies can reveal the kinetics of UPR activation in acute versus chronic disease states.
Correlation with other UPR markers: Multiplex immunofluorescence combining HSPA5 antibodies with antibodies against XBP1s, ATF6, and phospho-PERK can provide comprehensive UPR pathway analysis.
Cell-type specific UPR analysis: Combined immunofluorescence using HSPA5 antibodies and cell-type specific markers can identify which cells activate the UPR in heterogeneous tissues.
Subcellular localization changes: Under certain stress conditions, HSPA5 can relocalize from the ER to other cellular compartments. High-resolution confocal microscopy with HSPA5 antibodies can track these dynamics.
HSPA5 client protein interactions: Co-immunoprecipitation with HSPA5 antibodies can identify client proteins being chaperoned during disease progression.
HSPA5 post-translational modifications: Phospho-specific or other modification-specific HSPA5 antibodies can reveal regulatory mechanisms in disease models.
Based on recent findings connecting HSPA5 to retinoic acid signaling , several methodological approaches can elucidate this relationship:
Co-localization studies: Dual immunofluorescence with HSPA5 antibodies and retinoic acid receptor (RAR/RXR) antibodies can reveal spatial relationships in developing tissues.
Functional assays: Luciferase reporter assays with RARE (retinoic acid response element) constructs can quantify retinoic acid signaling in the presence of normal or altered HSPA5 levels .
HSPA5 knockdown/knockout systems: Using shRNA (as described in ) or CRISPR/Cas9 to modulate HSPA5 expression, followed by analysis of retinoic acid target gene expression via qPCR or RNA-seq.
Developmental phenotyping: Immunohistochemistry with HSPA5 antibodies in developmental models (e.g., Xenopus) under normal conditions versus treatment with retinoic acid synthesis inhibitors or exogenous retinoic acid.
Protein-protein interaction studies: Proximity ligation assays using HSPA5 antibodies and antibodies against components of the retinoic acid signaling pathway can detect direct or indirect interactions.
ChIP-seq approaches: Chromatin immunoprecipitation using HSPA5 antibodies followed by sequencing can identify potential chromatin associations relevant to retinoic acid signaling.
In vivo rescue experiments: In HSPA5-deficient developmental models, test whether retinoic acid pathway modulators can rescue developmental defects.
Advanced screening applications for HSPA5-targeting compounds include:
High-content imaging platforms: Automated immunofluorescence using HSPA5 antibodies can quantify changes in HSPA5 expression, localization, and co-localization with other UPR markers across large compound libraries.
In-cell Western assays: Using infrared-labeled secondary antibodies against HSPA5 primary antibodies in 96- or 384-well formats for rapid quantification of HSPA5 expression changes.
ELISA-based screening: Developing sandwich ELISAs with captured and detector HSPA5 antibodies to quantify secreted or released HSPA5 in response to compound treatment.
Flow cytometry applications: For suspension cells, intracellular staining with HSPA5 antibodies coupled with high-throughput flow cytometry can assess thousands of treatment conditions.
Reporter cell lines: Generating cells with fluorescent tags on endogenous HSPA5 (via CRISPR knock-in) for live-cell screening, validated with HSPA5 antibodies.
AlphaLISA or HTRF assays: Developing homogeneous, no-wash assays using HSPA5 antibody pairs for ultra-high-throughput screening applications.
Bead-based multiplex assays: Incorporating HSPA5 antibodies into Luminex or similar platforms for simultaneous quantification of multiple UPR markers.
Neurodegenerative disease research can leverage HSPA5 antibodies through several methodological approaches:
Brain region-specific analysis: Immunohistochemistry with HSPA5 antibodies can map ER stress patterns across brain regions in neurodegenerative disease models.
Temporal profiling: Using HSPA5 antibodies to track ER stress activation throughout disease progression, from presymptomatic to late-stage neurodegeneration.
Cell type-specific ER stress: Multiple-label immunofluorescence combining HSPA5 antibodies with neuronal, astrocytic, microglial, and oligodendrocyte markers can identify which CNS cells experience ER stress.
Protein aggregation correlation: Dual labeling with HSPA5 antibodies and antibodies against disease-specific protein aggregates (Aβ, tau, α-synuclein, TDP-43) can reveal spatial and temporal relationships between aggregation and ER stress.
Post-mortem human tissue validation: Comparing HSPA5 expression patterns between animal models and human post-mortem tissues can validate disease relevance.
Therapeutic intervention assessment: HSPA5 antibody-based assays can evaluate whether experimental therapeutics successfully mitigate neuronal ER stress.
Blood-brain barrier studies: Investigating whether peripheral HSPA5 can cross the blood-brain barrier under pathological conditions using labeled HSPA5 antibodies for tracking.
Cancer research applications for HSPA5 antibodies include:
Tissue microarray analysis: High-throughput immunohistochemistry with HSPA5 antibodies across large patient cohorts can correlate HSPA5 expression with clinical outcomes.
Therapy response prediction: Comparing pre- and post-treatment HSPA5 levels in patient-derived xenografts using HSPA5 antibodies to predict therapy responsiveness.
Circulating HSPA5 detection: Developing sensitive ELISAs with HSPA5 antibodies to detect secreted or released HSPA5 as a potential liquid biopsy biomarker.
Cell surface HSPA5 quantification: Flow cytometry with non-permeabilized cells using HSPA5 antibodies targeting extracellular epitopes can measure cancer-associated cell surface HSPA5.
HSPA5-directed therapy monitoring: Using HSPA5 antibodies to track the efficacy of emerging therapies targeting HSPA5 directly (e.g., HSPA5-targeting peptides or antibody-drug conjugates).
Combination therapy optimization: Assessing HSPA5 modulation during various therapeutic combinations to identify synergistic approaches that overcome therapy resistance.
Tumor microenvironment analysis: Investigating HSPA5 expression in cancer-associated fibroblasts, tumor-associated macrophages, and other stromal components using multiplex immunofluorescence with HSPA5 antibodies.
Cutting-edge imaging methodologies can revolutionize HSPA5 research:
Super-resolution microscopy: STORM, PALM, or STED microscopy with HSPA5 antibodies can visualize ER stress responses at nanoscale resolution, revealing previously undetectable organizational changes.
Live-cell imaging combinations: Pairing fluorescently-tagged HSPA5 with fixed-cell validation using HSPA5 antibodies to confirm dynamic observations.
Correlative light-electron microscopy (CLEM): Combining HSPA5 immunofluorescence with electron microscopy to correlate protein expression with ultrastructural ER changes.
Fluorescence resonance energy transfer (FRET): Using HSPA5 antibody-based FRET pairs to detect conformational changes or protein-protein interactions during ER stress.
Expansion microscopy: Physical expansion of specimens labeled with HSPA5 antibodies can achieve super-resolution-like images on conventional microscopes.
Light sheet microscopy: Enabling whole-organ or whole-embryo HSPA5 visualization with minimal photobleaching for developmental studies.
Intravital microscopy: Real-time imaging of HSPA5 dynamics in living tissues using fluorescently labeled HSPA5 antibodies or antibody fragments for in vivo studies.
Mass cytometry (CyTOF): Metal-conjugated HSPA5 antibodies can enable highly multiplexed single-cell analysis of ER stress in complex tissues.
Heat Shock 70kDa Protein 5 (HSP70) is a member of the heat shock protein family, which plays a crucial role in protein folding, protection against stress, and cellular homeostasis. The HSP70 family is highly conserved across species, including humans and mice. The Mouse Anti-Human HSP70 antibody is a monoclonal antibody used in various research applications to detect and study the HSP70 protein in human tissues and cells.
HSP70 proteins are characterized by their molecular weight of approximately 70 kDa. They consist of two main domains: the N-terminal ATPase domain and the C-terminal substrate-binding domain. The ATPase domain is responsible for binding and hydrolyzing ATP, which provides the energy required for the protein’s chaperone activity. The substrate-binding domain interacts with unfolded or misfolded proteins, preventing their aggregation and assisting in their proper folding.
HSP70 proteins are involved in several cellular processes, including:
The Mouse Anti-Human HSP70 antibody is typically produced using hybridoma technology. This involves the following steps:
The Mouse Anti-Human HSP70 antibody is widely used in various research applications, including: