RAB31 governs membrane trafficking and signaling pathways critical for cellular homeostasis:
RAB31 overexpression is associated with aggressive tumor behavior and poor prognosis across multiple cancers.
RAB31 represents a promising target for cancer therapy:
RAB31 (also known as Rab22b) is a monomeric GTP-binding protein belonging to the Rab5 subfamily of small GTPases . It was first cloned from human melanocytes and subsequently from human platelets . RAB31 primarily functions in regulating intracellular membrane trafficking, particularly:
Transport of the cation-dependent mannose 6-phosphate receptor (CD-M6PR) from the Golgi to endosomes
Regulation of early endosome-late endosome transport, especially for the epidermal growth factor receptor (EGFR)
Involvement in tubulovesicular carrier structures originating from the trans-Golgi network
For studying RAB31's basic functions, researchers should employ multiple complementary techniques:
Subcellular fractionation to isolate membrane compartments
Fluorescence microscopy with RAB31-specific antibodies or fluorescent protein fusions
Live-cell imaging to track vesicular movement in real-time
Co-localization studies with established organelle markers
RAB31 expression is regulated through multiple mechanisms that differ between normal and cancer contexts:
Transcriptional regulation:
RAB31 is an Estrogen Receptor α (ERα)-responsive gene, with expression regulated through the estrogen response element (ERE)
The oncoprotein mucin1-C (MUC1-C) forms a transcriptional complex with ERα that activates RAB31 expression
Post-transcriptional regulation:
The RNA binding protein HuR stabilizes RAB31 transcripts, contributing to elevated expression in cancer cells
An auto-inductive loop exists where elevated RAB31 stabilizes MUC1-C levels, which further enhances RAB31 expression
For investigating RAB31 regulation, researchers should consider:
Chromatin immunoprecipitation (ChIP) to confirm transcription factor binding
Luciferase reporter assays with wild-type and mutated RAB31 promoter constructs
RNA stability assays following actinomycin D treatment
siRNA knockdown of regulatory factors to establish pathway relationships
When analyzing RAB31 expression in patient samples, multiple detection methods should be employed:
Technique | Application | Advantages | Limitations |
---|---|---|---|
qRT-PCR | mRNA quantification | High sensitivity, quantitative | Doesn't reflect protein levels or localization |
Western blot | Protein detection | Semi-quantitative, detects post-translational modifications | Requires tissue lysis, loses spatial information |
Immunohistochemistry | Tissue localization | Preserves tissue architecture, allows cellular localization | Semiquantitative, antibody-dependent |
RNA-seq/SAGE | Transcriptome profiling | Unbiased, genome-wide assessment | Expensive, complex data analysis |
Protein mass spectrometry | Protein detection and quantification | Unbiased, can detect modifications | Requires specialized equipment, complex sample preparation |
When designing such studies, researchers should:
Include appropriate reference genes/proteins for normalization
Use RAB31-specific antibodies validated for cross-reactivity
Include both tumor and matched normal tissue controls
Consider correlation with clinicopathological parameters
Based on current research, RAB31 plays a significant role in cancer metastasis . An effective experimental design should incorporate multiple approaches:
In vitro models:
Migration and invasion assays with RAB31 manipulation (overexpression/knockdown)
3D organoid cultures to better mimic the tissue microenvironment
Analysis of epithelial-mesenchymal transition (EMT) markers
Exosome isolation and characterization (following the approach in )
In vivo models:
Orthotopic xenograft models that reflect the primary tumor environment
Tail vein injection models to assess pulmonary metastasis as demonstrated in gastric cancer research
Patient-derived xenografts for higher clinical relevance
Analysis techniques:
Multi-omics approaches (transcriptomics, proteomics)
Tracking of exosome production and cargo composition
Intravital imaging to monitor cell behavior in living animals
Research has shown that cells overexpressing RAB31 demonstrate enhanced migratory ability both in vitro and in pulmonary metastatic models of gastric cancer . Additionally, exosomes derived from RAB31 overexpressing cells promoted pulmonary metastasis when injected in vivo .
RAB31 has been identified as a key factor in cisplatin resistance in stomach adenocarcinoma (STAD) . To investigate this mechanism:
Recommended experimental approaches:
Cell viability assays following cisplatin treatment in RAB31-manipulated cells
Analysis of apoptotic markers and DNA damage response
Assessment of cisplatin uptake, retention, and efflux
Investigation of downstream molecular pathways
Key molecular pathway components:
RAB31 mediates cisplatin resistance via the epithelial-mesenchymal transition (EMT) pathway
Both RAB31 overexpression and cisplatin treatment increase Twist1 expression
RAB31 activates Twist1 through:
Depletion of Twist1 enhances sensitivity to cisplatin in STAD cells, which cannot be fully reversed by RAB31 overexpression
This suggests a RAB31/Stat3/MUC-1/Twist1/EMT pathway in drug resistance. A comprehensive experimental design would include:
Sequential manipulation of pathway components
Phospho-specific detection of Stat3 activation
Chromatin immunoprecipitation to analyze Twist1 promoter regulation
Rescue experiments to confirm the proposed pathway
RAB31 plays a significant role in regulating exosomes in gastric cancer, affecting both metastasis and cell-cell communication . To investigate this mechanism:
Exosome isolation and characterization:
Differential ultracentrifugation or commercial precipitation kits
Nanoparticle tracking analysis (NTA) for size and concentration measurement
Electron microscopy for morphological assessment
Western blotting for exosomal markers (CD63, CD9, TSG101)
Functional analyses:
Track exosome release using fluorescently labeled markers
Assess exosome uptake by recipient cells
Investigate exosomal cargo composition by proteomics/RNA-seq
Perform in vivo tracking of labeled exosomes
Key experimental findings to build upon:
Both number and size of exosomes secreted by gastric cancer cells were reduced when RAB31 expression was depleted
Injection of exosomes derived from RAB31-overexpressing cells promoted pulmonary metastasis in vivo
PSMA1 was identified as an exosomal protein overexpressed in gastric cancer tissue in accordance with RAB31 expression
PSMA1 overexpression was highly associated with poor prognosis in gastric cancer patients
A comprehensive experimental approach would include controls such as:
Comparison with other Rab proteins known to regulate exosome secretion
Assessment of general secretory pathway function
Validation in multiple cell lines to ensure reproducibility
The literature presents an interesting contradiction: RAB31 enhances EGFR degradation through early endosome-late endosome transport , which would suggest a tumor-suppressive role, yet it promotes cancer progression in multiple studies . Experimental designs to address this paradox should include:
Comparative analysis across cell systems:
Parallel studies in normal versus transformed cells
Investigation across different cancer subtypes
Assessment of RAB31 function in paired drug-sensitive and resistant cell lines
Mechanistic investigations:
EGFR trafficking assays using fluorescently labeled EGFR
Quantification of surface versus internalized EGFR
Assessment of EGFR degradation rates with varying RAB31 levels
Analysis of downstream EGFR signaling pathways (MAPK, PI3K/AKT)
Pathway interaction studies:
Investigation of how the RAB31-MUC1-C auto-inductive loop interfaces with EGFR trafficking
Assessment of whether RAB31 differentially affects trafficking of distinct receptor tyrosine kinases
Examination of possible compensatory mechanisms in cancer cells
Experimental design considerations:
Use of inducible expression systems to study acute versus chronic effects
Employment of domain-specific RAB31 mutants to dissect different functions
Development of computational models to predict the net outcome of multiple RAB31 functions
When designing experiments to manipulate RAB31 expression or function, researchers should consider these approaches:
Approach | Application | Advantages | Considerations |
---|---|---|---|
siRNA knockdown | Transient reduction (3-5 days) | Simple delivery, minimal off-target effects | Temporary effect, variable efficiency |
shRNA knockdown | Stable reduction | Long-term studies, selection possible | Potential off-target effects, adaptation |
CRISPR-Cas9 knockout | Complete elimination | Complete loss of function, specificity | May be lethal, compensation by other proteins |
CRISPR-Cas9 knock-in | Specific mutations | Study specific domains/functions | Technical complexity, efficiency |
Dominant-negative constructs | Functional inhibition | Can target specific functions | Overexpression artifacts, incomplete inhibition |
Constitutively active constructs | Functional activation | Study gain-of-function effects | Overexpression artifacts, non-physiological |
Experimental design recommendations:
Use multiple targeting sequences/approaches in parallel
Include appropriate controls (non-targeting, wild-type overexpression)
Validate manipulation at both mRNA and protein levels
Perform rescue experiments with RNAi-resistant constructs
Consider temporal control using inducible systems
For maximum rigor, experimental designs should incorporate both loss-of-function and gain-of-function approaches to fully characterize RAB31's role in the biological process under investigation.
Proper experimental controls are crucial for RAB31 research validity:
For expression studies:
Positive control: Tissues/cells known to express high RAB31 levels (brain oligodendrocytes)
Negative control: Tissues/cells with minimal RAB31 expression
Technical controls: Loading controls, reference genes for normalization
For functional studies:
Wild-type RAB31 expression alongside mutant constructs
Empty vector controls for overexpression studies
Non-targeting siRNA/shRNA for knockdown studies
Other Rab protein manipulations (especially Rab5 subfamily members) to test specificity
For clinical correlation studies:
Matched normal-tumor tissue pairs
Stratification by relevant clinical parameters
Multiple cohorts for validation
A rigorous experimental design should include time-course analyses and dose-response relationships where applicable, with appropriate statistical analyses determined during experimental planning.
When facing contradictory results about RAB31 function, consider these methodological approaches:
Systematic comparison:
Document key differences in experimental systems (cell types, culture conditions, etc.)
Replicate published protocols precisely before introducing variations
Perform side-by-side comparisons using standardized assays
Context-dependent analysis:
Evaluate RAB31 function across a panel of cell lines representing different tissues/cancer types
Assess the impact of the microenvironment on RAB31 function
Consider the influence of co-expressed proteins and signaling pathways
Validation strategies:
Employ multiple technical approaches to measure the same endpoint
Use both in vitro and in vivo models when possible
Validate with patient-derived samples or public datasets
Resolution framework:
Identify specific variables that might explain discrepancies
Design targeted experiments to test each variable systematically
Consider multifactorial designs to assess interaction effects
Develop integrated models that account for context-dependent functions
To establish RAB31 as a clinically relevant biomarker, researchers should implement:
Study design requirements:
Prospective collection with standardized protocols
Adequate sample size based on power calculations
Inclusion of diverse patient populations
Longitudinal sample collection where possible
Technical validation:
Multiple detection methods (IHC, qRT-PCR, proteomics)
Blinded assessment by multiple observers
Standardized scoring systems
Analytical validation (reproducibility, accuracy, precision)
Clinical validation:
Correlation with established clinicopathological parameters
Multivariate analysis controlling for confounding factors
Assessment of sensitivity, specificity, positive/negative predictive values
Independent validation cohorts
Current evidence supporting RAB31 as a biomarker includes:
Identification as a marker of ERα-positive breast carcinomas
Association with cisplatin resistance in stomach adenocarcinoma
To bridge the gap between RAB31 basic research and clinical applications:
Target validation strategies:
Genetic manipulation in patient-derived models
Correlation between RAB31 inhibition and clinical endpoints
Demonstration of synthetic lethality with existing therapies
Identification of patient subgroups most likely to benefit
Therapeutic approaches to explore:
Small molecule inhibitors targeting RAB31 GTPase activity
Disruption of the RAB31-MUC1-C auto-inductive loop
Targeting downstream effectors (Twist1, Stat3)
Exosome-targeted therapies in RAB31-high tumors
Experimental design for therapeutic development:
High-throughput screening for RAB31 modulators
Structure-based drug design targeting specific RAB31 domains
Combination studies with established chemotherapeutics
Assessment in patient-derived xenograft models
Translational considerations:
Development of companion diagnostics for patient selection
Biomarkers of response to RAB31-targeted therapy
Potential resistance mechanisms
Assessment of on-target versus off-target effects
Researchers should employ rigorous statistical methods when analyzing RAB31 expression data:
For expression level analysis:
Normality testing before selecting parametric/non-parametric tests
Appropriate paired/unpaired tests for tumor-normal comparisons
ANOVA with post-hoc tests for multi-group comparisons
Correction for multiple testing when performing genome-wide analyses
For survival analysis:
Kaplan-Meier analysis with log-rank test for initial assessment
Cox proportional hazards models for multivariate analysis
Consideration of competing risks when appropriate
Testing for proportional hazards assumption
For biomarker evaluation:
ROC curve analysis for diagnostic potential
Determination of optimal cutoff values
Net reclassification improvement assessment
Decision curve analysis for clinical utility
Advanced approaches:
Machine learning algorithms for complex pattern recognition
Nomogram development incorporating RAB31 with other factors
Propensity score matching to reduce bias in observational studies
For comprehensive understanding of RAB31 biology:
Data integration strategies:
Pathway enrichment analysis incorporating RAB31 expression data
Correlation networks linking RAB31 to other molecular features
Multi-omics factor analysis to identify latent factors
Causal inference methods to establish directionality
Computational approaches:
Gene set enrichment analysis (GSEA) with RAB31-correlated genes
Protein-protein interaction network analysis
Systems biology modeling of RAB31-related pathways
Inference of master regulators controlling RAB31 expression
Visualization methods:
Heatmaps for correlation patterns
Volcano plots for differential expression
Network graphs for interaction mapping
Sankey diagrams for pathway relationships
Validation requirements:
In silico validation using independent datasets
Experimental validation of key predictions
Cross-platform validation using different omics technologies
RAB31, also known as Ras-related protein Rab-31, is a member of the RAS oncogene family. This protein is encoded by the RAB31 gene and plays a crucial role in intracellular membrane trafficking. It is involved in various cellular processes, including protein transport, receptor internalization, and cellular response to insulin stimulus .
The RAB31 gene is located on chromosome 18 and is a protein-coding gene. The protein itself is a small GTPase, which means it can bind and hydrolyze GTP (guanosine triphosphate). RAB31 cycles between an inactive GDP-bound form and an active GTP-bound form. This cycling is essential for its role in membrane trafficking, as the active form recruits downstream effectors responsible for vesicle formation, movement, tethering, and fusion .
RAB31 is required for the integrity and normal function of the Golgi apparatus and the trans-Golgi network. It plays a significant role in the insulin-stimulated translocation of GLUT4 to the cell membrane, which is crucial for glucose uptake in cells. Additionally, RAB31 is involved in the transport of mannose-6-phosphate receptors (M6PR) from the trans-Golgi network to endosomes .
The protein also plays a role in the internalization of the epidermal growth factor receptor (EGFR) from the cell membrane into endosomes. This process is vital for regulating the availability of EGFR on the cell surface and, consequently, the cell’s response to growth signals .
Recombinant RAB31 is produced in E. coli as a single, non-glycosylated polypeptide chain containing 232 amino acids. It has a molecular mass of approximately 25.9 kDa and is often fused to a His-tag at the N-terminus for purification purposes. This recombinant protein is used in various research applications to study its function and role in different cellular processes .