Molecular Chaperone: HSP90B1 (GRP94/gp96) is an endoplasmic reticulum (ER)-resident HSP90 family member critical for folding, stabilizing, and activating secreted/membrane proteins like integrins, TLRs, and LDLR .
Stress Response: Upregulated during ER stress (e.g., hypoxia, calcium imbalance) but not cytosolic stressors like heat shock .
Autophagy Regulation: Modulates the PI3K/AKT/mTOR pathway to inhibit autophagy in cancers like head and neck squamous cell carcinoma (HNSC) .
Cancer Progression: Overexpressed in HNSC, gastric cancer (GC), and colorectal cancer (CRC), correlating with advanced tumor stages, metastasis, and poor prognosis .
Therapeutic Target: Cell surface HSP90B1 promotes tumor angiogenesis and growth, making it a focus for antibody-based therapies .
Western Blot Validation:
Immunofluorescence:
Cancer Mechanism Studies:
Antibody Internalization: The AT94B9 clone induces time-dependent internalization of cell surface HSP90B1, downregulating oncogenic signaling .
Combination Therapy: Synergizes with chemotherapeutics (e.g., 5-FU) in xenograft models .
HSP90B1, also known as Grp94, is a molecular chaperone belonging to the heat shock protein 90 family. It plays a crucial role in cellular processes by facilitating the degradation and removal of misfolded proteins and impaired organelles through autophagy mechanisms. Its significance in cancer research stems from its overexpression in various cancers, including head and neck squamous cell carcinoma (HNSC), where it correlates with advanced disease stages and poor prognosis . HSP90B1 interacts with over 100 different client proteins and contributes to tumor adaptation to microenvironmental stressors such as glucose deficiency, hypoxia, acidosis, and immune stimulation .
HSP90B1 antibodies are versatile tools applicable across multiple experimental techniques. Based on verified samples and recommended protocols, HSP90B1 antibodies demonstrate high utility in Western blot (WB) analyses at dilutions of 1:500-1:2000, immunohistochemistry (IHC) at 1:50-1:100, and immunofluorescence (IF) at 1:50-1:100 . These antibodies have been successfully validated in various cell lines (A375, HeLa, L-O2, NIH/3T3) and tissue samples (mouse liver, rat testis, human lung cancer, mouse kidney), confirming their reliability across human, mouse, and rat samples . For protein expression quantification, Western blot represents the gold standard, while IHC provides valuable insights into tissue localization patterns.
Methodological approach to antibody validation should include:
Positive and negative controls: Use cell lines with known HSP90B1 expression levels as verified in previous studies, such as TU686 and Fadu (high expression) compared to normal human oral keratinocytes (HOK, lower expression) .
Knockdown verification: Generate HSP90B1 knockdown models (using siRNA or shRNA approaches as described in research) to confirm antibody specificity by demonstrating reduced signal in Western blot or IHC .
Cross-reactivity testing: When working with non-human models, confirm reactivity across species using appropriate positive controls in mouse and rat samples .
Multiple detection methods: Validate expression using complementary techniques such as qRT-PCR to correlate protein detection with mRNA expression levels .
For optimal performance and longevity of HSP90B1 antibodies, researchers should adhere to the following methodological practices:
Store antibodies at -20°C for long-term storage, avoiding repeated freeze-thaw cycles
For working solutions, store at 4°C for up to one month
Prepare aliquots of concentrated antibody to minimize freeze-thaw cycles
Always centrifuge briefly before opening vials to collect solution at the bottom
For dilution, use appropriate buffers as recommended in specific protocols (typically PBS with 0.1% BSA)
When performing Western blot, IHC, or IF, optimize blocking conditions to minimize background signal
Investigating HSP90B1's role in autophagy requires sophisticated methodological approaches:
Dual immunofluorescence staining: Co-localize HSP90B1 with autophagy markers such as LC3B to visualize interaction points. Use HSP90B1 antibodies at 1:50-1:100 dilution for immunofluorescence .
Autophagy flux assessment: Combine HSP90B1 detection with autophagy inhibitors (chloroquine or bafilomycin A1) to distinguish between autophagy induction and blockade.
Protein interaction studies: Perform co-immunoprecipitation with HSP90B1 antibodies to identify binding partners within the PI3K/AKT/mTOR signaling pathway .
Functional validation: Use HSP90B1 knockdown and overexpression models to measure autophagy markers (LC3-I/LC3-II conversion, p62/SQSTM1 levels) by Western blot to establish causality .
Research has demonstrated that HSP90B1 obstructs autophagy and promotes HNSC progression through the PI3K/Akt/mTOR pathway, suggesting that targeting this interaction could present therapeutic opportunities .
When designing experiments to evaluate HSP90B1's influence on cancer progression, researchers should consider:
Resolving data inconsistencies requires methodological rigor:
Standardization of detection methods:
Cross-platform validation: Confirm expression patterns using complementary techniques:
Statistical approaches: Apply appropriate statistical methods to handle variability:
For smaller sample sizes: Non-parametric tests
For larger datasets: Consider data transformation when assumptions of normality are violated
Reference databases: Validate findings against public databases:
Use GEPIA database (http://gepia.cancer-pku.cn/) to compare expression in cancerous versus normal tissues
Correlate with survival data from Kaplan–Meier plotter database
Developing prognostic panels requires sophisticated methodological integration:
Multiplex immunohistochemistry:
Multivariate analysis frameworks:
Prognostic algorithm development:
Validation cohorts:
Test prognostic panels across diverse patient populations
Validate using both retrospective and prospective approaches
Common technical challenges in IHC with HSP90B1 antibodies include:
Background staining issues:
Antigen retrieval optimization:
Test both heat-induced epitope retrieval (HIER) and enzymatic retrieval methods
Adjust pH conditions (citrate buffer pH 6.0 vs. EDTA buffer pH 9.0)
Optimize retrieval duration based on tissue fixation conditions
Antibody concentration optimization:
Signal amplification for low-expressing samples:
Methodological approach to data interpretation should include:
Standardized scoring systems:
Statistical analysis framework:
Correlation with clinical parameters:
Presentation of results:
Rigorous quality control requires:
Reference standard inclusion:
Batch normalization strategies:
Include internal reference samples in each experimental batch
Apply appropriate normalization factors to adjust for batch effects
Consider replicate testing across batches for high-priority samples
Antibody validation for each new lot:
Perform Western blot validation with known positive samples
Compare staining patterns across antibody lots
Document lot-specific optimal working dilutions
Standardized protocols:
Maintain strict adherence to established protocols
Document any deviations or modifications
Use automated systems where possible to reduce technical variability
Future research methodologies should explore:
Proximity ligation assays (PLA):
Combine HSP90B1 antibodies with antibodies against key interacting proteins
Visualize direct protein-protein interactions in situ
Map interaction networks in different cellular compartments
CRISPR-based functional genomics:
Generate HSP90B1 domain-specific mutations to identify functional regions
Create cellular models with modified HSP90B1 phosphorylation sites
Correlate structural modifications with changes in autophagy markers
Single-cell analysis platforms:
Apply HSP90B1 antibodies in single-cell protein profiling
Identify cell subpopulations with distinct HSP90B1 expression patterns
Correlate with single-cell transcriptomics data
Live-cell imaging technologies:
Develop fluorescently tagged HSP90B1 constructs
Monitor real-time dynamics of HSP90B1 during autophagy induction
Correlate with autophagosome formation and clearance
Therapeutic development methodologies include:
Companion diagnostic development:
Standardize HSP90B1 IHC protocols for patient stratification
Establish clinically relevant expression thresholds
Correlate expression patterns with response to specific therapies
Drug screening platforms:
Combination therapy rational design:
Target HSP90B1 in conjunction with PI3K/AKT/mTOR pathway inhibitors
Monitor pathway modulation using phospho-specific antibodies
Assess synergistic effects on autophagy and apoptosis markers
Antibody-drug conjugate development:
Explore HSP90B1 as a potential target for antibody-drug conjugates
Assess internalization dynamics
Optimize linker chemistry and payload selection
Heat Shock Protein 90kDa Beta (GRP94) Member 1, also known as HSP90B1, is a molecular chaperone that plays a crucial role in the folding and stabilization of other proteins. It is a member of the heat shock protein 90 (HSP90) family and is predominantly found in the endoplasmic reticulum (ER). This protein is also referred to as endoplasmin, gp96, or ERp99 .
HSP90B1 is an ATP-metabolizing molecular chaperone that assists in the proper folding of secreted and membrane-bound proteins. It has ATPase activity, which is essential for its chaperone function. The protein is involved in the endoplasmic reticulum-associated degradation (ERAD) pathway, where it helps in the degradation of misfolded proteins .
HSP90B1 is also known to participate in the unfolding of cytosolic leaderless cargos, such as interleukin 1 (IL-1), to facilitate their translocation into the ER-Golgi intermediate compartment (ERGIC) and subsequent secretion. This translocation process is mediated by the cargo receptor TMED10 .
HSP90B1 plays a critical role in various biological processes, including:
The expression of HSP90B1 is associated with various pathogenic states, including tumor formation. It has been implicated in cancer progression and is considered a potential target for cancer therapy. Additionally, mutations in the HSP90B1 gene have been linked to diseases such as Pelizaeus-Merzbacher-Like Disease and Nemaline Myopathy 9 .
Mouse anti-human HSP90B1 antibodies are commonly used in research to study the protein’s function and its role in various diseases. These antibodies are generated by immunizing mice with human HSP90B1 protein, leading to the production of antibodies that specifically recognize and bind to the human version of the protein. These antibodies are valuable tools for detecting HSP90B1 in various experimental settings, including Western blotting, immunohistochemistry, and immunoprecipitation .