RRAS regulates actin cytoskeleton organization and integrin-mediated cell adhesion . Its GTPase activity enables signal transduction through interactions with:
RASSF5: Modulates lymphocyte adhesion and integrin activation
RAF1: Activates MAPK/ERK cascades influencing proliferation/apoptosis
This protein also binds Bcl-2 and ARAF, linking it to apoptosis and kinase signaling .
STRING-db analysis identifies critical functional partners :
Interactor | Role in RRAS Pathway | Confidence Score |
---|---|---|
RALGDS | GDP/GTP exchange for Ral GTPases | 0.999 |
RAF1 | MAPK/ERK cascade activation | 0.991 |
RASSF5 | Integrin activation and tumor suppression | 0.971 |
RALBP1 | GTP hydrolysis for RAC1/CDC42 | 0.970 |
A genome-scale RNAi screen identified RRAS as a modifier of mutant huntingtin toxicity :
Cellular models: RRAS knockdown suppressed toxicity in human/mouse neurons
Drosophila studies: Reduced RRAS homolog levels rescued motor defects (p<0.01)
Pathogenic mechanism: Mutant huntingtin increased GTP-bound RRAS levels by 35% in striatal cells
Dysregulated RRAS contributes to:
Noonan syndrome: Mutations cause cardiovascular/neurological abnormalities
Metastasis: Mediates aberrant cell migration via integrin β1 activation
Therapeutic targeting: Farnesyltransferase inhibitors suppress RRAS-driven toxicity
Recent studies highlight RRAS as a biomarker and therapeutic target:
Recombinant RRAS Human serves as a critical reagent for:
GTPase activity assays
Protein interaction studies (e.g., co-IP with RASSF5 or RAF1)
Structural analyses of Ras family dynamics
This protein’s stability in glycerol-containing buffers (20mM Tris-HCl, pH 8) facilitates experimental reproducibility .
Ras-related protein R-Ras is a protein encoded by the RRAS gene located on human chromosome 19. It belongs to the RAS family of small GTPases and plays critical roles in various cellular functions including cell adhesion, migration, and proliferation . The significance of RRAS in human research stems from its involvement in multiple signaling pathways that regulate fundamental cellular processes. Unlike other RAS family members that have been extensively studied, RRAS presents unique research challenges and opportunities due to its distinct functional properties and interaction networks. Methodologically, researchers approach RRAS studies through molecular techniques including gene expression analysis, protein-protein interaction assays, and functional genomics, which collectively provide insights into RRAS's role in normal physiology and disease states.
Randomized experimental designs are essential for establishing causal relationships in RRAS functional studies. When investigating RRAS's effects on cellular processes, random assignment of experimental units (cells, tissues, or animal models) to treatment groups ensures that any observed differences can be attributed to the experimental manipulation rather than pre-existing variations . This approach minimizes selection bias and strengthens internal validity. Methodologically, researchers should implement simple randomization techniques when assigning experimental units to different RRAS manipulation conditions (e.g., overexpression, knockdown, or pharmacological intervention). For instance, cell cultures can be randomly assigned to RRAS overexpression versus control groups using computer-generated random number sequences, ensuring that each experimental unit has an equal probability of assignment to any condition . This randomization approach is particularly important when studying RRAS function across different cell types which may have varying baseline expression levels.
When designing experiments to study RRAS interactions with other proteins or cellular components, researchers should consider several methodological factors:
Interaction specificity verification: Multiple complementary techniques (co-immunoprecipitation, proximity ligation assays, FRET) should be employed to confirm direct interactions.
Physiological relevance: Experiments should maintain near-physiological expression levels of RRAS to avoid artifacts from overexpression systems.
Temporal dynamics: Designs should incorporate time-course analyses to capture transient interactions.
Subcellular localization: Experimental approaches should account for the spatial distribution of RRAS within cells.
Post-translational modifications: Methods should consider how modifications affect interaction profiles.
The experimental design should include appropriate controls for antibody specificity, expression levels, and cellular context. When studying RRAS interactions in human samples, randomized controlled trial designs can strengthen the validity of findings by minimizing systematic differences between experimental groups . For cell-based studies, quasi-experimental designs may be appropriate when randomization at the cellular level is challenging, though researchers should acknowledge the increased risk of confounding variables .
Designing randomized controlled trials (RCTs) for RRAS-targeted interventions requires careful methodological consideration. The gold standard approach involves:
Stratified randomization: Group participants based on relevant RRAS expression patterns or genetic variants before randomization to ensure balanced distribution across treatment arms . This is particularly important given the heterogeneity of RRAS expression across different tissues and disease states.
Blinding procedures: Implement double-blinding where both participants and investigators are unaware of group assignments to minimize expectation bias .
Sample size determination: Calculate required sample sizes based on anticipated effect sizes from preliminary RRAS modulation studies, accounting for potential dropouts and subgroup analyses.
Outcome measurement standardization: Develop standardized protocols for measuring molecular and clinical endpoints related to RRAS pathway activation.
Biomarker integration: Include validated biomarkers of RRAS activity as secondary endpoints to establish mechanism-based evidence.
The following table outlines key considerations for different phases of RRAS-targeted intervention RCTs:
Trial Phase | Primary Objective | RRAS-Specific Design Considerations | Sample Size Considerations |
---|---|---|---|
Phase I | Safety and optimal dosing | Monitor RRAS pathway biomarkers for target engagement | 20-30 subjects, dose-escalation design |
Phase II | Preliminary efficacy | Stratify by baseline RRAS expression/activity | 100-200 subjects, powered for surrogate endpoints |
Phase III | Definitive efficacy | Include genetic analyses of RRAS variants | 300+ subjects, powered for clinical endpoints |
While RCTs provide the strongest evidence for causality, they present challenges including high costs and extended timeframes. Researchers should consider whether the ethical and practical constraints of their specific RRAS research question warrant this rigorous approach .
Measuring RRAS activation in heterogeneous human tissue samples presents significant methodological challenges requiring specialized approaches:
Single-cell analysis techniques: Implement single-cell RNA sequencing and mass cytometry to quantify RRAS expression and activation at the individual cell level, overcoming the limitations of bulk tissue analysis. This approach allows identification of cell-specific RRAS activation patterns within heterogeneous samples.
Spatial transcriptomics: Employ techniques like spatial transcriptomics or MERFISH to map RRAS expression and activation while preserving spatial context within tissue architecture.
Phospho-specific detection: Utilize phospho-specific antibodies against RRAS or downstream effectors in combination with laser capture microdissection to isolate specific cell populations before biochemical analysis.
Multiplexed imaging: Implement multiplexed immunofluorescence or imaging mass cytometry to simultaneously visualize RRAS activation alongside cell-type markers.
Computational deconvolution: Apply bioinformatic algorithms to deconvolute bulk tissue data into cell-type-specific signals when single-cell approaches are not feasible.
The experimental design should include appropriate tissue-matched controls and standardized processing protocols to minimize technical variation. When analyzing data from heterogeneous samples, researchers should employ statistical approaches that account for cellular composition differences . This comprehensive approach enables more accurate assessment of RRAS activation patterns within complex human tissues than conventional bulk analysis methods.
Designing effective genetic association studies for RRAS variants requires rigorous methodological approaches:
Study design selection: Choose between case-control, cohort, or family-based designs based on the specific research question. For rare RRAS variants, family-based designs may provide greater statistical power.
Population stratification control: Implement principal component analysis or genomic control methods to account for population substructure that might confound RRAS variant associations .
Sample size determination: Calculate required sample sizes based on anticipated effect sizes, variant frequencies, and desired statistical power. For common RRAS variants with modest effects, several thousand participants may be necessary.
Variant selection strategy: Consider both common and rare variants within the RRAS gene and its regulatory regions. Include tag SNPs that capture haplotype blocks and potentially functional variants identified through bioinformatic prediction.
Phenotype definition: Establish clear, reproducible definitions of phenotypes potentially influenced by RRAS function, using standardized assessment tools where possible.
The table below summarizes methodological approaches for different types of RRAS genetic association studies:
Study Type | Suitable Research Questions | Methodological Approach | Analytical Considerations |
---|---|---|---|
GWAS | Identifying common RRAS variants associated with disease risk | Genome-wide genotyping with imputation | Multiple testing correction; replication in independent cohorts |
Targeted Sequencing | Identifying rare RRAS variants with functional impact | Deep sequencing of RRAS locus and regulatory regions | Burden tests or variant collapsing methods for rare variant analysis |
Expression QTL | Identifying variants affecting RRAS expression | Integrated genomic and transcriptomic analysis | Cell/tissue-specific eQTL mapping; mediation analysis |
Researchers should address potential biases through appropriate control selection, blinding during phenotyping, and rigorous quality control of genetic data . Meta-analysis approaches can be employed to increase statistical power by combining data across multiple RRAS genetic studies.
Translating RRAS research findings from in vitro to in vivo human contexts requires systematic methodological approaches to bridge this critical gap:
Physiologically relevant model systems: Employ organoids, patient-derived xenografts, and humanized animal models that better recapitulate human RRAS signaling dynamics compared to traditional cell lines.
Cross-validation strategy: Implement a systematic cross-validation framework where RRAS findings are sequentially verified across increasingly complex models before human studies:
Cell lines → Primary human cells → 3D cultures → Organoids → Animal models → Human subjects
Context-specific validation: Verify RRAS functions across multiple relevant tissue and cell types, acknowledging that RRAS may have context-dependent effects.
Multi-omics integration: Combine transcriptomic, proteomic, and metabolomic analyses to comprehensively assess RRAS pathway effects across different experimental systems.
Intermediate biomarker development: Establish translational biomarkers of RRAS activity that can be measured consistently across in vitro, animal, and human studies.
A particular methodological challenge involves controlling for differences in microenvironmental factors that influence RRAS signaling. Researchers should design experiments that systematically account for these differences, potentially through co-culture systems or engineered matrices that mimic human tissue conditions . Additionally, careful documentation of passage number, culture conditions, and cell authentication is essential for reproducibility of in vitro RRAS findings.
When confronting data inconsistencies in RRAS functional studies, researchers should employ robust statistical approaches:
When faced with contradictory findings regarding RRAS function, researchers should systematically evaluate potential sources of variation including cell type, experimental conditions, RRAS expression levels, and analysis methods . This comprehensive approach enables more nuanced interpretation of apparently inconsistent results and facilitates integration of findings across studies.
Measuring dynamic RRAS activation in living human tissues presents unique methodological challenges requiring specialized approaches:
Real-time biosensor development: Design and validate FRET or BRET-based biosensors specific to RRAS activation states that can be delivered via viral vectors or expressed in patient-derived cells.
Intraoperative measurement protocols: Develop standardized protocols for rapid tissue processing and real-time RRAS activity measurement during surgical procedures, preserving activation state fidelity.
Ex vivo tissue maintenance: Implement precision-cut tissue slice cultures or microfluidic tissue culture systems that maintain viable human tissue architecture while enabling real-time imaging of RRAS dynamics.
Multiplexed activation state profiling: Combine phospho-flow cytometry with single-cell sequencing to simultaneously assess RRAS activation alongside upstream regulators and downstream effectors.
Computational modeling: Develop mathematical models of RRAS activation dynamics calibrated with experimental data from accessible human tissues to predict behavior in less accessible tissues.
The experimental design should include appropriate time-course analyses to capture the potentially transient nature of RRAS activation events. When analyzing dynamic data, researchers should employ statistical approaches designed for time-series analysis rather than single-timepoint comparisons . This comprehensive approach enables more accurate assessment of RRAS activation dynamics than conventional endpoint analyses.
CRISPR-based methodologies offer powerful approaches for investigating RRAS function in human cellular contexts:
Domain-specific mutagenesis: Implement precise CRISPR-Cas9 editing to introduce specific mutations in functional domains of endogenous RRAS, enabling assessment of structure-function relationships without overexpression artifacts.
Activation/repression systems: Employ CRISPRa and CRISPRi technologies to modulate endogenous RRAS expression at physiologically relevant levels, providing advantages over traditional overexpression or knockdown approaches.
Temporal control strategies: Utilize inducible CRISPR systems (e.g., Tet-regulated or chemically-inducible Cas9) to study time-dependent aspects of RRAS function.
Base and prime editing: Apply these newer CRISPR technologies to introduce specific point mutations in RRAS without DNA double-strand breaks, reducing off-target effects.
Genetic interaction mapping: Implement CRISPR screens targeting RRAS pathway components to systematically map genetic dependencies and functional interactions.
The experimental design should include comprehensive off-target analysis through next-generation sequencing and appropriate controls for potential cellular responses to Cas9 expression. When analyzing CRISPR editing efficiency and phenotypic outcomes, researchers should employ quantitative approaches that account for heterogeneity in editing outcomes at the single-cell level. This methodological approach enables more precise manipulation of RRAS than conventional genetic techniques, facilitating detailed functional characterization.
Developing patient-derived models for RRAS research requires careful methodological consideration:
Patient selection strategy: Implement systematic selection criteria considering:
Disease heterogeneity and staging
Prior treatments that might affect RRAS pathway activity
Comorbidities that could confound RRAS-specific effects
Genetic background variations affecting RRAS signaling
Model validation framework: Establish comprehensive validation requirements including:
Genomic fidelity assessment through sequencing
Transcriptomic comparison to original patient tissue
RRAS pathway activation state verification
Functional recapitulation of disease phenotypes
Technical standardization: Develop standardized protocols for:
Tissue processing and cell isolation
Culture conditions optimized for maintaining RRAS signaling fidelity
Passage number limitations to prevent drift
Cryopreservation methods that preserve RRAS pathway function
Heterogeneity considerations: Address intratumoral or tissue heterogeneity through:
Multiple sampling regions from individual patients
Single-cell approaches to characterize subpopulations
Clonal isolation and comparison when appropriate
The following table outlines key methodological considerations for different patient-derived model types in RRAS research:
Model Type | Advantages for RRAS Research | Technical Considerations | Validation Requirements |
---|---|---|---|
Primary Cell Cultures | Direct assessment of patient RRAS pathway | Limited lifespan; potential for selection bias | Compare RRAS expression/activation to original tissue |
Patient-Derived Xenografts | Preserved tumor architecture and heterogeneity | Murine microenvironment effects on RRAS signaling | Verify stability of RRAS pathway across passages |
Organoids | 3D architecture with epithelial organization | Matrix composition effects on RRAS function | Compare morphological features dependent on RRAS signaling |
iPSC-Derived Models | Differentiation into multiple RRAS-expressing lineages | Maturation status effects on RRAS function | Verify developmental stage-appropriate RRAS expression |
The most promising methodological innovations for advancing RRAS research in human contexts integrate cutting-edge technologies with robust experimental design:
Spatial multi-omics approaches: Technologies that simultaneously map RRAS expression, activation states, and downstream effects while preserving spatial tissue context represent a significant methodological advance. These approaches overcome limitations of traditional bulk analyses by revealing cell type-specific and spatially restricted RRAS functions within complex human tissues.
Advanced live-cell imaging techniques: Super-resolution microscopy combined with optogenetic RRAS activation tools allows unprecedented visualization of RRAS dynamics in near-native contexts. These methods enable researchers to study RRAS activation with subcellular spatial resolution and millisecond temporal precision.
Artificial intelligence integration: Machine learning algorithms trained on large-scale RRAS pathway datasets can identify subtle patterns and relationships not apparent through conventional analyses. AI approaches may be particularly valuable for predicting RRAS network behaviors across different cellular contexts and disease states.
Organ-on-chip technologies: Microfluidic systems that recapitulate tissue-specific microenvironments while enabling precise manipulation of RRAS signaling represent promising platforms for studying RRAS function in physiologically relevant settings with experimental control not possible in vivo.
Multi-scale modeling approaches: Integrating molecular dynamics simulations of RRAS protein interactions with cellular and tissue-level models creates comprehensive frameworks for predicting how molecular perturbations affect higher-order biological processes.
These methodological innovations should be implemented within rigorous experimental designs that include appropriate controls, randomization where possible, and thoughtful statistical approaches . As these technologies continue to evolve, researchers should develop standardized protocols and quality control metrics to ensure reproducibility across different laboratories and experimental contexts.
The Related RAS Viral (r-ras) Oncogene Homolog, commonly referred to as RRAS, is a member of the Ras superfamily of small GTP-binding proteins. These proteins play a crucial role in various cellular processes, including cell growth, differentiation, and survival. The RRAS gene is located on chromosome 19q13.33 in humans .
The RRAS gene was first isolated by Lowe et al. in 1987 through low-stringency hybridization with a Harvey-ras probe . The predicted RRAS protein consists of 218 amino acids and has an amino-terminal extension of 26 residues compared to HRAS p21. The human RRAS protein shares 55% sequence identity with HRAS p21 .
RRAS is a plasma membrane-associated GTP-binding protein with intrinsic GTPase activity. It cycles between an active GTP-bound state and an inactive GDP-bound state at the cytoplasmic face of the plasma membrane . RRAS is involved in promoting cell adhesion and neurite outgrowth. It has been implicated in various cellular signaling pathways, including those mediated by the semaphorin-4D (SEMA4D) receptor plexin B1 .
RRAS is primarily expressed in vascular smooth muscle cells in small arterioles and major arteries, as well as in endothelial cells of lung capillaries . Lower levels of RRAS are found in smooth muscle cells of veins, renal glomeruli, and venous endothelium of the spleen. In smooth muscle cells, RRAS is distributed along the plasma membrane .
RRAS has been implicated in the pathogenesis of various human cancers. It is known to transduce growth inhibitory signals across the cell membrane, exerting its effect through an effector shared with other Ras proteins . Downregulation of RRAS activity by the plexin B1/RND1 complex is essential for SEMA4D-induced growth cone collapse in hippocampal neurons .
Studies on Rras-null mice have shown that these mice are viable and fertile with no obvious abnormalities. However, they exhibit exaggerated neointimal thickening in response to arterial injury and increased angiogenesis in implanted tumors . Overexpression of activated RRAS suppresses mitogenic and invasive activities of growth factor-stimulated vascular cells .