STAMBP (STAM-binding protein), also known as AMSH, is a JAMM metalloprotease in the deubiquitinase (DUB) family. It functions primarily as an endosome-associated DUB by interacting with the SH3 domain of STAM, a component in the endosomal sorting complexes required for transport (ESCRT) machinery . STAMBP plays critical roles in:
Regulating endosomal sorting and recycling
Maintaining cellular homeostasis
Modulating inflammasome activation
Cancer progression in several malignancies
Research significance has increased as STAMBP dysregulation has been linked to various diseases, including cancer and neurodegenerative disorders . Notably, elevated STAMBP expression correlates with poor clinical prognosis in multiple cancers, such as lung adenocarcinoma, breast cancer, triple-negative breast cancer, and pancreatic cancer .
Based on validated antibodies in the search results, STAMBP antibodies can be reliably used for:
When selecting an antibody, researchers should consider the specific reactivity (human, mouse, rat) and whether the experimental design requires polyclonal or monoclonal antibodies .
For proper validation of STAMBP antibody specificity, implement the following methodological approach:
Positive controls: Use cell lines known to express STAMBP, such as HT-29 human colon adenocarcinoma cells, which have been validated to express detectable levels of STAMBP by western blot .
Knockdown validation: Compare antibody reactivity between normal cells and those with STAMBP knockdown via siRNA or CRISPR/Cas9. The search results show that shSTAMBP plasmid effectively decreased STAMBP expression as validated by western blotting and qRT-PCR analyses .
Cross-reactivity testing: Verify that STAMBP antibodies don't cross-react with other JAMM deubiquitinase family members. Western blot analysis has confirmed that altering STAMBP does not affect other proteins in the Zn+-dependent JAMM deubiquitinases family .
Multiple detection methods: Confirm findings using more than one technique (e.g., WB, IHC, and IF) to ensure consistent results across platforms .
STAMBP plays a significant role in chemoresistance, particularly in pancreatic cancer through the STAMBP-E2F1-PDK1 axis:
PDK1-mediated metabolic reprogramming: STAMBP enhances aerobic glycolysis and suppresses mitochondrial respiration to increase gemcitabine resistance in pancreatic cancer cells. This occurs through stabilization of PDK1, an essential regulator of the aerobic glycolytic process .
E2F1 stabilization mechanism: STAMBP promotes PDK1-mediated Warburg effect and chemotherapy resistance by modulating E2F1. It directly binds to E2F1 and suppresses its degradation and ubiquitination .
Experimental evidence: Knockdown of STAMBP significantly increased gemcitabine sensitivity in pancreatic cancer cells both in vitro and in vivo. In TCGA cohort analysis, patients with low STAMBP expression showed better response to gemcitabine treatment, while approximately 80.6% of high STAMBP-expressing patients exhibited progressive disease after treatment .
Therapeutic potential: Entrectinib, an FDA-approved drug, was identified as a STAMBP inhibitor that enhances gemcitabine effectiveness in a patient-derived xenograft model, suggesting a potential approach to overcome chemoresistance .
STAMBP modulates inflammasome activation and cytokine secretion through multiple mechanisms:
NLRP3 deubiquitination: STAMBP regulates NLRP3 inflammasome activity by controlling its K63-linked polyubiquitination. In monocytes, STAMBP knockout increased NLRP3 K63 chain polyubiquitination resulting in increased NLRP3 inflammasome activation .
NALP7 protein stabilization: STAMBP stabilizes NALP7 by preventing its lysosomal degradation through deubiquitination. Using a cell-free DUB assay, researchers demonstrated that recombinant STAMBP directly deubiquitinates NALP7 in a time-dependent manner .
IL-1β regulation: STAMBP knockout in monocytes increased expression of numerous cytokines and chemokines in response to TLR agonists or LPS. This exaggerated inflammatory response was dependent on IL-1β signaling, as STAMBP knockout directly increased IL-1β release with TLR ligation .
Pharmacological targeting: Small-molecule inhibitor BC-1471 decreases NALP7 protein levels and suppresses IL-1β release after TLR agonism, suggesting potential therapeutic applications for inflammatory conditions .
For investigating STAMBP interactions with partner proteins, researchers should consider:
Co-Immunoprecipitation: Use anti-STAMBP antibodies (0.5μg-4μg) with 200-400μg extracts of whole cells for IP experiments . This approach was successfully used to demonstrate STAMBP's interaction with E2F1 in pancreatic cancer research .
Immunoprecipitation-Mass Spectrometry: This combined approach identified the interaction between STAMBP and the actin-binding protein RAI14 in triple-negative breast cancer cells .
Cell-free deubiquitination assays: Develop assays using recombinant STAMBP and immunopurified ubiquitinated substrates to directly test STAMBP's deubiquitination activity, as demonstrated with Ub-NALP7 .
Proximity ligation assays: For detecting in situ protein-protein interactions within cells to confirm STAMBP binding partners identified through other methods.
Functional validation: Confirm the significance of identified interactions through genetic manipulation (knockdown/overexpression) of STAMBP and assessment of target protein stability, as demonstrated with RAI14 in TNBC .
To ensure reliable STAMBP detection in different experimental contexts:
For Western blotting:
For Immunohistochemistry:
For Immunofluorescence/ICC:
Optimize fixation method (paraformaldehyde is commonly used)
Include permeabilization step to access intracellular STAMBP
Use appropriate blocking solutions to minimize background
The search results demonstrate several validated approaches:
RNA interference:
CRISPR/Cas9 gene editing:
Overexpression systems:
In vivo models:
Based on published research methodologies, include these critical controls:
For gene expression studies:
For protein stability experiments:
For ubiquitination studies:
For in vivo studies:
When encountering inconsistent results with STAMBP antibodies:
Antibody validation:
Verify antibody specificity using positive controls (e.g., HT-29 human colon adenocarcinoma cell line)
Test multiple antibodies targeting different epitopes of STAMBP
Consider the immunogen used to generate the antibody - some use recombinant fusion protein containing sequences corresponding to amino acids 100-270 of human STAMBP
Sample preparation issues:
Ensure complete protein denaturation for western blotting
Optimize fixation protocols for immunohistochemistry and immunofluorescence
Include appropriate protease inhibitors in lysis buffers
Technical considerations:
Cell type variations:
To comprehensively analyze STAMBP's role in diseases like cancer:
Multi-omics integration:
Pathway analysis:
Clinical correlation:
Drug sensitivity testing:
To differentiate between direct and indirect STAMBP effects:
In vitro deubiquitination assays:
Rescue experiments:
Restore wild-type STAMBP in knockout models to confirm phenotype reversibility
Use catalytically inactive STAMBP mutants to determine if enzymatic activity is required for observed effects
Domain-specific mutations:
Generate STAMBP constructs with mutations in specific functional domains
Test which domains are essential for specific protein interactions or cellular functions
Temporal analysis:
Use inducible expression/knockdown systems to track immediate versus delayed effects of STAMBP manipulation
Combine with time-course analyses of downstream events to establish causality
Direct binding confirmation:
By implementing these methodological approaches, researchers can more confidently distinguish STAMBP's direct enzymatic and binding functions from secondary pathway effects.
Multiple studies have established significant correlations between STAMBP expression and clinical outcomes:
These clinical correlations suggest STAMBP could serve as both a prognostic biomarker and potential therapeutic target in multiple cancer types.
To evaluate STAMBP as a potential therapeutic target:
High-throughput screening approaches:
Preclinical model testing:
Combination therapy assessment:
Biomarker development: