HSPA5, also known as BiP (Binding Immunoglobulin Protein), GRP78 (78 kDa Glucose-Regulated Protein), or Heat Shock Protein Family A Member 5, is a 78 kDa molecular chaperone localized primarily in the endoplasmic reticulum (ER). It belongs to the HSP70 family and plays a critical role in ER homeostasis, protein folding, and stress response. HSPA5 is ubiquitously expressed in human tissues (Table 1) and is highly upregulated under conditions of ER stress, such as hypoxia, glucose deprivation, or viral infections .
HSPA5 operates through an ATPase cycle:
ATP-bound state: SBDα lid opens, allowing substrate release.
ADP-bound state: SBDα lid closes, trapping substrates for folding or degradation .
Co-chaperones like PDI (Protein Disulfide Isomerase) and J proteins enhance its activity .
HSPA5 is expressed in all tissues, with notable abundance in:
Tissue | Expression Level | Source |
---|---|---|
Liver | High | |
Lung | High | |
Pancreas | High | |
Kidney | Moderate | |
Normal vs. Cancer Tissues | Upregulated in 14/31 cancer types (e.g., bladder, pancreatic, lung) . |
HSPA5 is a prognostic marker in multiple cancers:
Bladder cancer: Promotes proliferation, metastasis, and VEGFA/VEGFR2 pathway activation; linked to poor prognosis .
Pancreatic adenocarcinoma: Associated with reduced survival in neoadjuvant therapy patients .
Mechanism: Enhances cell cycle progression (S-phase accumulation), inhibits apoptosis, and regulates ferroptosis .
HSPA5 acts as a potential co-receptor for SARS-CoV-2:
Expression: HSPA5 mRNA levels are 253-fold higher than ACE2 in lung cancer tissues, suggesting enhanced viral entry .
Clinical Impact: Elevated HSPA5 correlates with severe COVID-19 outcomes, particularly in cancer patients .
HSPA5 modulates lipid metabolism in nonalcoholic fatty liver disease (NAFLD):
RNA binding: Interacts with mRNAs involved in lipid synthesis and AMPK signaling, influencing alternative splicing and gene expression .
Target | Therapeutic Strategy | Application |
---|---|---|
ER stress modulation | Small-molecule inhibitors (e.g., KIRA6) | Cancer, COVID-19 |
HSPA5 knockdown | siRNA/shRNA, CRISPR-Cas9 | Preclinical cancer models |
cs-HSPA5 antibodies | Neutralizing antibodies | Viral entry blockade |
RNA interactions: HSPA5 binds lncRNAs and coding RNAs, regulating splicing and lipid metabolism in NAFLD .
Senescence: Inhibits cisplatin-induced senescence in ovarian cancer, suggesting a dual role in chemoresistance .
Species | Conservation Status | Cysteine Residues |
---|---|---|
Human | Yes | 2 |
Mouse | Yes | 2 |
Rat | Yes | 3 |
Cow | Yes | 2 |
Drosophila melanogaster | Yes (Hsc70-3) | N/A |
Saccharomyces cerevisiae | Yes (SSA1) | N/A |
HSPA5 is a 78 kDa member of the heat shock protein 70 family. The human HSPA5 protein consists of 654 amino acids, including an 18 amino acid signal sequence and a 636 amino acid mature region containing a hydantoinase A region (amino acids 145-245) . The protein contains a C-terminal KDEL motif present on intracellular HSPA5 but absent on secreted forms .
HSPA5 functions include:
Acting as an ER chaperone for protein folding
Participating in the unfolded protein response
Maintaining cellular homeostasis under stress conditions
When secreted extracellularly, inducing IL-10 production from T cells and interacting with Cripto to block TGF-beta signaling
Multiple validated methods exist for detecting HSPA5 in research settings:
Western Blot Analysis:
Human-specific detection: Use MAB4846 at 1 μg/mL with HRP-conjugated Anti-Mouse IgG Secondary Antibody
Multi-species detection (human/mouse/rat): Use AF4846 at 0.5 μg/mL with HRP-conjugated Anti-Goat IgG Secondary Antibody
Simple Western™ Analysis:
Human samples: HSPA5 detected at approximately 72-74 kDa using 5-10 μg/mL antibody concentration
Use 12-230 kDa separation system
Note: Non-specific interaction with the 230 kDa standard may occur with some antibodies
Immunohistochemistry:
Effective for subcellular localization in tissue sections, particularly retinal tissues
Can distinguish between extracellular and intracellular forms of the protein
Expression analysis from multiple databases (GTEx, HPA, FANTOM5) reveals:
Bone marrow exhibits the highest HSPA5 expression among normal tissues
HSPA5 shows relatively low RNA tissue specificity across normal tissues
In single-cell RNA analysis, HSPA5 exhibits limited cell-type specificity
HSPA5 demonstrates weak cancer specificity in RNA cancer classification analyses
This limited tissue specificity suggests HSPA5 performs crucial functions across diverse cell types, consistent with its role in fundamental cellular processes like protein folding.
Based on comprehensive pan-cancer analyses, researchers should consider:
Multi-omics Integration Strategy:
Transcriptomic data: Utilize TCGA datasets to analyze mRNA expression
Proteomic verification: Incorporate CPTAC data to confirm translational changes
Differential expression analysis: Compare HSPA5 levels between tumor and normal tissues using tools like TIMER2
Validation in specific cancer types: Focus on cancers like BRCA, LIHC, and COAD for detailed investigation
Survival Analysis Protocol:
Determine optimal cutoff values specific to each cancer type
Generate Kaplan-Meier curves and survival heatmaps
Analyze multiple survival metrics:
Use R packages "survminer" and "survival" for statistical analysis
Functional Network Analysis:
Construct protein-protein interaction networks using STRING
Identify top 100 HSPA5-correlated genes using GEPIA2
Perform KEGG pathway and GO enrichment analyses on combined datasets
Conduct Gene Set Enrichment Analysis (GSEA) comparing high vs. low HSPA5 expression groups
HSPA5 shows significant correlations with immune parameters requiring specific experimental approaches:
Immune Checkpoint Correlation Studies:
Comprehensive Immune Infiltration Analysis:
Validation Experimental Design:
Select representative cancer types spanning different tissue origins
Perform immunohistochemistry to visualize HSPA5 and immune cell markers simultaneously
Conduct flow cytometry analysis of tumor-infiltrating lymphocytes in relation to HSPA5 expression
Research focusing on retinal HSPA5 illustrates effective systems genetic methods applicable across tissues:
Quantitative Trait Locus (QTL) Mapping:
Utilize genetic reference panels (e.g., BXD family from C57BL/6J × DBA/2J mice)
Apply stepwise refinement through:
Integration with Phenotypic Data:
Connect molecular findings with disease-relevant phenotypes
Incorporate datasets from repositories like GeneNetwork
Model cellular responses and disease outcomes using high-throughput quantitative data
Validation Methodology:
Confirm subcellular localization patterns through immunohistochemistry
Compare expression between healthy and diseased tissues
Assess functional consequences of candidate gene perturbation
The UPR represents a complex signaling network requiring sophisticated experimental approaches:
Gene Ontology Enrichment Focus Areas:
Response to ER stress
Golgi vesicle transport
Response to topologically incorrect proteins
ER to Golgi vesicle-mediated transport
Experimental Design Recommendations:
Analyze HSPA5 interactions with other UPR components:
Assess protein quality control mechanisms:
Utilize stress induction protocols:
Chemical inducers (tunicamycin, thapsigargin)
Physiological stressors (glucose deprivation, hypoxia)
Monitor temporal dynamics of HSPA5 response
Based on validated antibody data:
Storage and Handling:
Use a manual defrost freezer and avoid repeated freeze-thaw cycles
Store for 12 months from receipt at -20 to -70°C as supplied
After reconstitution, store for 1 month at 2 to 8°C or 6 months at -20 to -70°C under sterile conditions
Western Blot Optimization:
Use reducing conditions consistently
Determine optimal antibody dilutions empirically for each application
Be aware that antibody MAB4846 is human-specific, while AF4846 detects human, mouse, and rat HSPA5
HSPA5 can appear at different molecular weights depending on methodology:
Detection Method | Observed Molecular Weight | Notes |
---|---|---|
Western Blot | 78-80 kDa | Standard reducing conditions |
Simple Western™ | 72-74 kDa | Capillary-based immunoassay |
Theoretical | 78 kDa | Based on amino acid sequence |
Researchers should consider that:
Post-translational modifications may alter migration patterns
Different detection systems can yield varying apparent molecular weights
Alternative splicing in the signal sequence (aa 1-10) and multiple single amino acid substitutions can affect observed size
When investigating HSPA5 in pathological conditions:
Expression Control Panel:
Subcellular Fractionation Controls:
Functional Validation Approaches:
To delineate HSPA5's multiple roles:
Domain-Specific Analysis:
Localization-Based Functional Studies:
Context-Dependent Protein Interaction Mapping:
HSPA5 is a multifunctional protein that primarily resides in the endoplasmic reticulum (ER). It is involved in:
HSPA5 has been implicated in various physiological and pathological processes:
Recombinant HSPA5 is produced using genetic engineering techniques, where the HSPA5 gene is cloned and expressed in a suitable host system, such as bacteria or yeast. This allows for the production of large quantities of the protein for research and therapeutic purposes.