GPSM1 antibodies are immunological reagents designed to detect and quantify GPSM1, a 72 kDa protein encoded by the GPSM1 gene (UniProt ID: Q86YR5). This protein regulates G-protein signaling by maintaining Gα subunits in their GDP-bound state, uncoupling them from GPCR pathways . GPSM1 is associated with metabolic disorders (e.g., type 2 diabetes), immune dysregulation, and cancers such as B-cell acute lymphoblastic leukemia (B-ALL) .
GPSM1 antibodies enable detection of myeloid GPSM1 overexpression in adipose tissue, linking it to insulin resistance and hepatic steatosis in obesity .
Mechanistic studies reveal GPSM1 suppresses TNFAIP3 transcription via the Gαi3/cAMP/PKA/CREB axis, exacerbating NF-κB-driven inflammation .
In B-ALL, GPSM1 knockdown reduces proliferation (48–60% suppression in BALL-1/Reh cells) and induces apoptosis via ADCY6-RAPGEF3-JNK pathway inhibition .
High GPSM1 expression correlates with poor survival in leukemia patients (HR = 1.7, p < 0.05) .
GPSM1 antibodies identify its role in macrophage polarization and adipose tissue inflammation, critical for metabolic syndrome .
GPSM1, also known as Activator of G protein signaling 3 (AGS3), functions as a guanine nucleotide dissociation inhibitor (GDI) that modulates heterotrimeric G-protein signaling. It keeps G(i/o) alpha subunits in their GDP-bound form, thus uncoupling heterotrimeric G-proteins signaling from G protein-coupled receptors. GPSM1 plays crucial roles in:
Controlling spindle orientation and asymmetric cell fate of cerebral cortical progenitors
Regulating macroautophagy in intestinal cells
Modulating drug addiction pathways
Influencing inflammatory responses via macrophage polarization
GPSM1 exhibits strong genetic association with Type 2 diabetes and Body Mass Index in population studies, implicating its role in metabolic regulation .
GPSM1 antibodies can be utilized in multiple experimental applications:
Western Blotting (WB): Typically at dilutions of 1:500-1:2000
Immunohistochemistry (IHC): Optimally at dilutions of 1:50-1:300
ELISA: Generally used at dilutions of 1:5000-1:10000
The selection of technique should be guided by your specific research question and experimental model.
When selecting a GPSM1 antibody, researchers should consider:
Target epitope: Antibodies targeting different regions (e.g., AA 1-166, AA 60-300, AA 158-457) may yield different results based on protein conformation and interactions
Clonality: Both monoclonal and polyclonal antibodies are available; monoclonals offer higher specificity while polyclonals may provide stronger signals
Host species: Available in rabbit, mouse, and other hosts, which is important for co-localization studies
Validated applications: Ensure the antibody has been validated for your specific application
Reactivity: Confirm cross-reactivity with your species of interest (human, mouse, rat)
Based on recent research, effective experimental designs should include:
Cell-based approaches:
Loss-of-function studies: Use shRNA or siRNA to knockdown GPSM1 in cancer cell lines (e.g., BALL-1 and Reh cells for B-ALL studies)
Gain-of-function studies: Adenovirus-mediated GPSM1 overexpression to examine effects on proliferation and apoptosis
Cell viability assays: CCK-8 assays at multiple time points (48h, 72h, 96h) following GPSM1 manipulation
Apoptosis assessment: Annexin V-PE/7-AAD dual staining followed by flow cytometry
In vivo approaches:
Orthotopic xenograft models using GPSM1-manipulated cancer cells
Analysis of tumor growth, invasion, and metastasis
Immunohistochemical analysis of tumor sections for GPSM1 expression
For colorectal cancer research specifically, consider the relationship between GPSM1 expression and immune checkpoint blockade resistance .
Proper experimental controls are critical for GPSM1 functional studies:
Negative control siRNA/shRNA (non-targeting sequence)
Empty vector control for overexpression studies
Untransfected/untreated cells (blank control)
Quantification of knockdown efficiency at both mRNA level (by RT-qPCR) and protein level (by Western blot)
Inclusion of housekeeping genes/proteins (e.g., GAPDH) for normalization
Time-course analysis to account for temporal changes in expression
In one study, siRNA-mediated knockdown of GPSM1 achieved 48% and 42% reduction at the mRNA level in BALL-1 and Reh cells respectively, with protein interference efficiency of 45% and 60% .
To analyze GPSM1's effects on downstream signaling:
For G-protein signaling studies:
For cancer signaling pathways:
For inflammatory pathways:
When facing discrepancies in GPSM1 expression data:
Consider methodological differences:
Antibody epitope targets may affect detection efficiency
Fixation protocols can influence antibody accessibility
Detection methods (colorimetric vs. fluorescent) vary in sensitivity
Account for biological variables:
Validation strategies:
Variability in GPSM1 detection can result from:
To improve GPSM1 detection in Western blotting:
Sample preparation optimization:
Ensure complete lysis using appropriate buffers (containing protease inhibitors)
Optimize protein loading (30-50 μg total protein typically works well)
Use fresh samples when possible
Technical adjustments:
Try different antibody dilutions (start with manufacturer's recommendation, then 1:500-1:2000)
Extend primary antibody incubation time (overnight at 4°C)
Test alternative blocking agents (5% BSA often performs better than milk for phospho-epitopes)
Increase sensitivity using enhanced chemiluminescence reagents
Control experiments:
Recent research has revealed GPSM1's important role in tumor microenvironment regulation, particularly in:
Macrophage polarization studies:
Use multiplex immunofluorescence with GPSM1 antibodies alongside macrophage markers (CD68, CD163)
Analyze co-localization of GPSM1 with M1 vs. M2 macrophage markers
Quantify GPSM1 expression in tumor-associated macrophages vs. cancer cells
Assess correlation between GPSM1 levels and macrophage infiltration
Immunosuppressive mechanisms:
In vitro co-culture systems:
To investigate GPSM1's interactions with G-protein signaling:
Protein-protein interaction studies:
Co-immunoprecipitation of GPSM1 with Gα subunits (particularly Gαi/o)
Proximity ligation assays to visualize interactions in situ
FRET/BRET approaches for real-time interaction monitoring
Functional analyses:
Structural considerations:
To investigate GPSM1's role in metabolic regulation:
Animal models:
Mechanistic investigations:
Translational approaches:
To investigate GPSM1 regulation:
Post-translational modification studies:
Transcriptional regulation:
Protein-protein interaction network:
These methodologies have revealed that GPSM1 stability is regulated by deubiquitination via USP9X, and that stabilized GPSM1 can promote MEIS3 nuclear translocation, activating macrophage colony-stimulating factor expression .
Emerging research suggests several approaches for therapeutic targeting of GPSM1:
In cancer therapy:
For metabolic disorders:
Target validation approaches:
Genetic validation using conditional knockout models
Pharmacological inhibition with structure-based designed compounds
PROTAC-based approaches for targeted GPSM1 degradation
For accurate quantification of GPSM1:
Expression analysis:
Activity assessment:
Clinical correlation: