Key specifications from commercial sources ( ):
| Property | Details |
|---|---|
| Target | RAB22A (UniProt ID: Q9UL26) |
| Reactivity | Human, mouse, canine, hamster |
| Applications | WB, IHC, IF, IP, ELISA |
| Host/Isotype | Rabbit IgG |
| Immunogen | RAB22A fusion protein (residues 170–185: DANLPSGGKGFKLRRQ) |
| Molecular Weight | 22 kDa (calculated: 194 aa) |
| Storage | -20°C in PBS with 0.02% sodium azide and 50% glycerol |
Developed using a carboxy-terminal peptide antigen, this antibody localizes to recycling endosomal tubules and regulates membrane protein trafficking .
RAB22A governs clathrin-independent endocytic pathways, particularly for proteins like major histocompatibility complex class I (MHCI):
Mechanism: Rab22a associates with MHCI-containing tubular intermediates and controls their formation through GTPase activity. Dominant-negative Rab22a mutants inhibit tubule formation and MHCI recycling .
Coordination: Works with Rab11a to regulate distinct steps: Rab22a initiates tubule formation, while Rab11a facilitates final fusion with the plasma membrane .
RAB22A modulates immune cell function and infiltration, particularly in hepatocellular carcinoma (HCC):
| Immune Cell Type | Correlation | Key Markers Affected | p-value |
|---|---|---|---|
| M2 Macrophages | Positive | CD163, VSIG4, MS4A4A | <0.001 |
| Cytotoxic T cells | Negative | CD8A, CD3D, GZMB | <0.001 |
| T helper cells | Positive | GATA3, STAT5A | <0.001 |
| Dendritic cells | Negative | CD1C, ITGAX | <0.001 |
| T-cell exhaustion | Negative | PD-1 (PDCD1), TIM-3 (HAVCR2) | <0.001 |
Reduces cytotoxic T-cell infiltration, impairing antitumor immunity .
Associated with T-cell exhaustion markers, suggesting a role in immune evasion .
RAB22A drives tumor progression via:
Standardized methods for RAB22A antibody use include:
RAB22A is a membrane-bound GTPase that cycles between GTP-bound active and GDP-bound inactive forms . It plays essential roles in endocytosis and intracellular protein transport, particularly in trafficking between early endosomes and recycling endosomes . RAB22A mediates the trafficking of transferrin from early endosomes to recycling endosomes and is required for NGF-mediated endocytosis of NTRK1, which supports neurite outgrowth . In epithelial cells, RAB22A contributes to the establishment of cell polarity and localizes to the cell-cell interface of polarizing cell pairs .
Validating RAB22A antibody specificity requires multiple approaches:
Western blot analysis to confirm the predicted 22 kDa band size, as demonstrated with antibodies like EPR9486 and EPR9487
Positive controls using cell lines with known RAB22A expression (MCF7, BxPC-3, HeLa)
Negative controls using RAB22A knockdown models
Cross-validation across multiple applications (WB, ICC/IF, IP) to ensure consistent results
Peptide competition assays using the immunizing peptide sequence (e.g., residues 170-185: DANLPSGGKGFKLRRQ)
Comparative analysis with different antibody clones targeting distinct epitopes
When performing immunofluorescence with RAB22A antibodies, expect the following patterns:
Partial localization to recycling endosomes
Overexpression of wild-type RAB22A can induce formation of abnormally large vacuole-like structures containing EEA1 but not Rab11 (recycling endosome marker) or LAMP-1 (late endosome/lysosomal marker)
In polarizing epithelial cells, localization to the cell-cell interface
Experimental design for studying hypoxia-induced RAB22A regulation should include:
Exposure of cells to controlled hypoxic conditions (typically 1-2% O₂) compared to normoxia (21% O₂)
RT-qPCR analysis to measure RAB22A mRNA induction under hypoxia, as demonstrated in MCF-7, MDA-231, and MDA-435 cell lines
HIF dependency assessment using HIF-1α and HIF-2α knockdown cell models (shRNA or CRISPR-Cas9)
Chromatin immunoprecipitation (ChIP) assays to analyze HIF binding to the RAB22A promoter, focusing on the HIF binding site in the 5'-untranslated region of exon 1
Western blot analysis to confirm protein-level changes correlate with transcript changes
Functional rescue experiments using ectopic RAB22A expression in HIF-deficient cells
The data indicate that knockdown of either HIF-1α or HIF-2α blocks hypoxia-induced RAB22A expression, and both HIF-1α and HIF-1β bind to a specific site in the RAB22A gene under hypoxic conditions .
To study RAB22A's role in microvesicle (MV) formation:
Nanoparticle tracking analysis
Collect conditioned media from cells with manipulated RAB22A expression
Ultracentrifuge to isolate MVs
Analyze particle size distribution and concentration
Compare MV production between normoxic and hypoxic conditions
Colocalization studies
Use immunofluorescence to visualize RAB22A at budding MVs
Employ high-resolution microscopy techniques (TIRF, super-resolution)
Analyze colocalization with membrane markers
Functional studies
Generate stable RAB22A knockdown cell lines
Compare effects of wild-type RAB22A versus GTPase-deficient mutants (e.g., Q64L) on MV production
Transfer labeled MVs to recipient cells and assess functional effects on invasion and metastasis
Analyze MV cargo composition through proteomics and RNA-seq
Research shows that hypoxia-induced MV shedding requires RAB22A, and RAB22A knockdown completely eliminates increased MV production under hypoxic conditions .
To investigate RAB22A-EEA1 interactions:
Biochemical approaches
Perform pull-down assays using GST-tagged RAB22A loaded with GTPγS (active) or GDP (inactive)
Conduct co-immunoprecipitation experiments with antibodies against RAB22A and EEA1
Use the yeast two-hybrid system to map interaction domains
Microscopy techniques
Analyze colocalization of RAB22A and EEA1 using confocal microscopy
Implement FRET or BiFC assays to confirm direct protein interactions
Use live-cell imaging to track dynamic interactions
Functional studies
Generate RAB22A mutants affecting GTPase activity (constitutively active Q64L or inactive forms)
Analyze effects on early endosome morphology and function
Assess trafficking of cargo proteins like transferrin in cells with manipulated RAB22A-EEA1 interaction
Research demonstrates that the GTP-bound form of RAB22A interacts with the N-terminus of EEA1, and this interaction is implicated in controlling endosomal membrane trafficking .
For rigorous analysis of RAB22A as a prognostic marker:
Research shows that high RAB22A expression correlates with decreased survival in breast cancer and other malignancies .
To study RAB22A in the tumor immune microenvironment:
Immune cell infiltration analysis
Use single-sample Gene Set Enrichment Analysis (ssGSEA) to evaluate correlation between RAB22A expression and immune cell infiltration
Perform immunohistochemistry for immune cell markers in tissues with varying RAB22A expression
Analyze correlation between RAB22A and immune cell markers using appropriate statistical methods
Correlation with immune markers
Conduct comprehensive correlation analysis between RAB22A and immune cell markers
Focus on markers for T cells, B cells, macrophages, dendritic cells, and other immune populations
Functional validation
Manipulate RAB22A expression in tumor models and assess changes in immune infiltration
Perform co-culture experiments with tumor cells and immune cells
Evaluate effects on antigen presentation and T cell activation
Research indicates RAB22A expression positively correlates with T helper cells, Tcm cells, and Th2 cells, but negatively with cytotoxic cells, dendritic cells, and plasmacytoid dendritic cells . The table below shows correlation between RAB22A and various immune markers in hepatocellular carcinoma:
| Immune Cell Type | Marker | Correlation Coefficient | P-value |
|---|---|---|---|
| CD8+ T cell | CD8A | -0.463 | <0.001 |
| T cell (general) | CD3D | -0.446 | <0.001 |
| B cell | CD79A | -0.487 | <0.001 |
| M2 Macrophage | CD163 | -0.480 | <0.001 |
| Dendritic cell | BDCA-1 (CD1C) | -0.426 | <0.001 |
| T cell exhaustion | PD-1 (PDCD1) | -0.429 | <0.001 |
These correlations suggest RAB22A may influence the immunosuppressive tumor microenvironment .
To investigate RAB22A's role in PI3K/Akt/mTOR signaling:
Protein interaction studies
Signaling activation analysis
Assess phosphorylation levels of key pathway components (PI3K, Akt, mTOR, p70S6K, 4EBP1) in cells with altered RAB22A expression
Use Western blotting with phospho-specific antibodies
Implement kinase activity assays
Functional validation
Research demonstrates that RAB22A transfection in lung adenocarcinoma cells upregulates phosphorylation of core PI3K/Akt/mTOR pathway proteins, and rapamycin treatment significantly reduces the enhanced proliferation, migration, and invasion induced by RAB22A overexpression .
To investigate RAB22A's role in MHC-I trafficking:
Trafficking assays
Track fluorescently labeled MHC-I molecules in cells with manipulated RAB22A expression
Analyze internalization, recycling, and degradation rates of surface MHC-I
Compare trafficking in different immune cell types (dendritic cells, T cells)
Endosomal characterization
Perform subcellular fractionation to isolate endosomal compartments
Analyze co-localization of RAB22A with MHC-I in different endosomal subpopulations
Use immunoelectron microscopy for high-resolution localization
Functional consequences
Assess antigen presentation efficiency using T cell activation assays
Measure cell surface MHC-I levels using flow cytometry
Evaluate impacts on immune synapse formation
Research indicates that accurate intracellular transport of MHC-I molecules in dendritic cells and T lymphocytes depends on RAB22A function . RAB22A also regulates clathrin-independent endocytosis processes, including the internalization of MHC-I molecules in T lymphocytes .
To investigate RAB22A's differential effects on these pathways:
Pathway-specific markers
Track fluid-phase endocytic markers (e.g., dextran) versus phagocytic targets (e.g., latex beads)
Analyze recruitment of ER-derived proteins to phagosomes versus endosomes
Use pathway-specific inhibitors to distinguish between mechanisms
Compartment isolation
Perform magnetic isolation of phagosomes versus endosomes
Conduct proteomic analysis of isolated compartments
Compare RAB22A recruitment to different compartments
Functional assays
Assess antigen translocation to the cytosol from phagosomes versus endosomes
Evaluate cross-presentation efficiency of antigens delivered via different routes
Analyze maturation kinetics of each compartment type
Research shows that in RAB22A-deficient dendritic cells, the recruitment of ER-derived proteins is normal in phagosomes but diminished in endosomes labeled with fluid-phase markers . Additionally, early endosomal maturation is altered in RAB22A-deficient DCs, highlighting the importance of studying these pathways separately .
To differentiate RAB22A functions from related RABs:
Comparative expression analysis
Perform detailed phylogenetic analysis of the RAB family
Assess tissue-specific expression patterns of closely related RABs
Use single-cell RNA sequencing to identify cell types with differential expression
Domain-specific studies
Create chimeric proteins swapping domains between RAB22A and related RABs
Conduct mutagenesis of RAB22A-specific residues
Use structural biology approaches to identify unique interaction interfaces
Rescue experiments
Knockdown RAB22A and attempt rescue with related RABs
Assess which functions are RAB22A-specific versus redundant
Implement double knockdown approaches to identify compensatory mechanisms
Research indicates RAB22A shows highest sequence homology to Rab5 and acts downstream of Rab14 in establishing epithelial polarity . These relationships should be considered when designing experiments to isolate RAB22A-specific functions.
When exploring RAB22A as a therapeutic target:
Target validation
Evaluate RAB22A expression across multiple cancer types
Confirm oncogenic functions in multiple models
Assess phenotypes of RAB22A inhibition in normal versus cancer cells
Targeting strategies
Consider direct inhibition of GTPase activity
Explore disruption of protein-protein interactions
Evaluate indirect targeting through upstream regulators like HIFs
Combination approaches
Test RAB22A targeting with established therapies
Combine with PI3K/Akt/mTOR pathway inhibitors
Assess synergy with immunotherapies given RAB22A's immune functions
Biomarker development
Develop assays for patient stratification based on RAB22A expression
Identify correlations with treatment response
Create companion diagnostics for RAB22A-targeted therapies
Research demonstrates that RAB22A promotes multiple cancer hallmarks including proliferation, migration, and invasion in lung adenocarcinoma cells through PI3K/Akt/mTOR signaling . Additionally, in breast cancer, RAB22A is linked to microvesicle formation and metastasis through HIF-dependent mechanisms .