RASL12 is functionally linked to diverse biological processes, though its precise role remains under-characterized compared to oncogenic Ras isoforms (e.g., KRAS, NRAS) .
GTPase Activity: Predicted to bind GDP/GTP and hydrolyze GTP .
Subfamily Context: Classified within the RAS subfamily, which includes both oncogenic (e.g., KRAS) and tumor-suppressive (e.g., DIRAS) members .
Disease Associations: Functional associations include links to molecular profiles, phenotypes, and diseases, though direct evidence for RASL12’s role in pathology is limited .
RASL12 exhibits tissue-specific expression, with notable activity in brain regions and other organs.
In brain tissues, RASL12 expression is detected in regions such as the amygdala, basal ganglia, and cerebellum .
Studies in model organisms highlight RASL12’s responsiveness to environmental and chemical stressors.
These findings suggest RASL12 may serve as a biomarker for xenobiotic exposure or toxicity .
Recombinant RASL12 is used in experimental settings for functional and structural studies.
RASL12 (RAS-like, family 12) is a human protein that belongs to the RAS small GTPases superfamily, which includes over 170 RAS-related proteins involved in signal transduction . The RAS superfamily is typically subclassified into RAS, RHO, RAB, and ARF families, along with the closely related Gα family, with each family arranged into evolutionarily conserved branches . These groupings reflect structural, biochemical, and functional conservation across family members. RASL12, also known as RIS (Ras family member Ris), represents one of the less characterized members of this extensive protein network.
Expression data for RASL12 can be accessed through resources like the Harmonizome database, which aggregates information from multiple expression datasets . According to available data, RASL12 expression has been cataloged in various tissue types including brain tissues from the Allen Brain Atlas datasets . Researchers investigating tissue-specific expression patterns should consider consulting these comprehensive databases, which provide standardized expression values across different tissues and developmental stages. For more specific expression analysis, quantitative PCR or RNA-seq approaches comparing RASL12
across tissue panels represent reliable methodological approaches.
While specific information about RASL12 gene regulation is limited in the search results, research approaches to study its regulation would include:
Promoter analysis: Identify transcription factor binding sites in the RASL12 promoter region using bioinformatic tools and validate with chromatin immunoprecipitation (ChIP)
Epigenetic regulation: Investigate DNA methylation patterns and histone modifications using bisulfite sequencing and ChIP-seq
Response elements: Test the promoter's responsiveness to various stimuli using reporter gene assays
Alternative splicing: Analyze potential splice variants through RNA-seq data and RT-PCR validation
Studies on other RAS family genes have shown regulation through alternative splicing, as observed with KRAS and HRAS, which generate isoforms with distinct subcellular localizations . Similar mechanisms might apply to RASL12, though this requires experimental verification.
To characterize the GTPase activity of RASL12, researchers should consider the following methodological approaches:
| Method | Application | Advantages | Limitations |
|---|---|---|---|
| Radioactive GTP hydrolysis assay | Measures intrinsic and GAP-stimulated GTPase activity | Gold standard for quantitative kinetic analysis | Requires radioactive materials and specialized facilities |
| Fluorescence-based assays (MANT-GTP) | Real-time monitoring of nucleotide binding/release | Non-radioactive, continuous measurement | Potential interference from protein fluorescence |
| HPLC analysis | Quantification of GDP/GTP ratio | Precise measurement of nucleotide states | Labor-intensive, requires specialized equipment |
| Bioluminescence-based assays | High-throughput screening applications | Amenable to drug screening, high sensitivity | Indirect measurement, potential artifacts |
For reliable results, researchers should produce recombinant RASL12 protein using bacterial or mammalian expression systems, with careful consideration of potential post-translational modifications that might affect activity . The protein should be purified to homogeneity using affinity chromatography followed by size exclusion chromatography to ensure sample quality for enzymatic assays.
The RAS subfamily in humans comprises approximately 35 proteins arranged in 12 structural or functional branches . While specific functional data on RASL12 is limited, comparison with other RAS proteins suggests potential roles in signal transduction. Unlike classic RAS proteins (HRAS, KRAS, NRAS) that typically undergo prenylation for membrane localization, some RAS family members show variations in their localization mechanisms .
For experimental comparison of RASL12 with other RAS proteins, researchers should:
Perform phylogenetic analysis to identify the most closely related RAS proteins
Compare sequence conservation in functional domains, particularly switch regions
Analyze subcellular localization patterns using fluorescent protein fusions
Conduct effector binding studies to identify interaction partners
Perform rescue experiments in cells depleted of specific RAS proteins
Comparative functional studies would ideally include analysis of downstream signaling pathway activation (MAPK, PI3K, etc.) by techniques such as western blotting for phosphorylated effectors or transcriptional reporter assays.
While specific effector proteins for RASL12 are not directly described in the search results, research approaches to identify potential effectors would include:
Pull-down assays using purified RASL12-GTP (active) versus RASL12-GDP (inactive)
Yeast two-hybrid screening to identify protein-protein interactions
Proximity labeling approaches (BioID, APEX) in cellular contexts
Mass spectrometry-based interaction proteomics
In silico prediction based on conservation of effector binding regions
Other RAS subfamily members interact with effectors containing specific interaction domains such as RBD (RAS-binding domain), RA (RAS association) domain, or others that recognize the GTP-bound state . For example, classic RAS proteins interact with effectors like RAF kinases, PI3K, and RALGDS . The RRAS branch proteins (RRAS1, RRAS2, MRAS) show overlapping effector specificity with classical RAS proteins, including interactions with PI3K, RALGDS and RAF kinases . Given the sequence-based classification of RASL12 as a RAS family member, similar interactions might be expected, though with potentially unique specificities.
To investigate potential roles of RASL12 in cancer, researchers could employ the following approaches:
Analysis of RASL12 expression across cancer types using public databases (TCGA, CCLE)
Correlation of expression levels with patient outcomes and clinical parameters
Functional studies using:
Overexpression of wild-type or mutant RASL12
CRISPR-Cas9 knockout or knockdown approaches
Rescue experiments with specific effector pathway inhibitors
Single-cell RNA sequencing to assess RASL12 expression in tumor microenvironment contexts
The blueprint approach described in search result provides a framework for analyzing protein expression in different cancer types and cellular contexts. This study profiled multiple cancer types including lung cancer, colorectal cancer, and ovarian cancer at single-cell resolution . Such approaches could be valuable for understanding RASL12 expression patterns within the heterogeneous tumor microenvironment.
Generating functional RASL12 expression constructs requires careful consideration of several factors:
Codon optimization: Depending on the expression system, codon optimization may improve protein yield
Affinity tags: Consider tag position (N- or C-terminal) based on potential functional domains
Mutations for functional studies:
Subcellular localization signals: Consider including or excluding potential membrane localization motifs
For mammalian expression, vectors with CMV or EF1α promoters typically provide strong expression, while inducible systems (Tet-On/Off) allow for temporal control. For biochemical studies, bacterial expression in E. coli (typically BL21 strains) with T7-based vectors is recommended, potentially with chaperone co-expression to improve folding.
Post-translational modifications (PTMs) are crucial for RAS protein function and localization. While specific information about RASL12 PTMs is not provided in the search results, several approaches can be used to investigate potential modifications:
Mass spectrometry-based proteomics:
Enrichment strategies for specific PTMs (phosphorylation, ubiquitination, etc.)
Targeted and untargeted approaches to identify modification sites
Site-directed mutagenesis of predicted modification sites followed by functional assays
Western blotting with PTM-specific antibodies
In silico prediction tools for potential modification sites
Other RAS family proteins undergo various PTMs including prenylation, palmitoylation, phosphorylation, and nitrosylation . For example, HRAS undergoes serine phosphorylation and nitrosylation, while RRAS1 can be tyrosine phosphorylated . Analysis of the RASL12 sequence for conserved modification motifs could guide experimental design for PTM studies.
Single-cell RNA sequencing (scRNA-seq) has emerged as a powerful tool for understanding gene expression heterogeneity across cell populations. For RASL12 research, the following approaches are recommended:
Unbiased profiling: Process tissues to single-cell suspensions followed by droplet-based (10x Genomics) or plate-based (Smart-seq2) scRNA-seq
Targeted analysis: Use the blueprint approach described in to align new data with existing cellular profiles
Cellular indexing: CITE-seq or REAP-seq to correlate transcript levels with surface protein expression
Spatial transcriptomics: Techniques like Visium or MERFISH to preserve spatial context of expression
The cancer blueprint described in search result demonstrates how single-cell profiling across multiple cancer types can reveal shared and unique cellular phenotypes. This study generated approximately 1 billion unique transcripts from 183,373 cells across 50 tumor tissues and 17 normal tissues . Similar approaches could be valuable for understanding RASL12 expression patterns in normal and pathological contexts.
Development of specific antibodies against RASL12 requires careful planning:
Antigen selection:
Full-length protein may provide better recognition but risks cross-reactivity with related RAS proteins
Unique peptide regions (typically 12-20 amino acids) offer higher specificity
Consider native protein conformation for applications requiring conformational epitopes
Validation approaches:
Western blotting against recombinant protein and endogenous RASL12
Immunoprecipitation followed by mass spectrometry
Immunofluorescence with appropriate controls (knockdown/knockout cells)
Testing against related RAS family proteins to confirm specificity
Applications:
Separate antibodies may be needed for different applications (WB, IP, IF, IHC)
Consider generating antibodies that distinguish GTP/GDP-bound states
Commercial antibodies should be thoroughly validated before use in research applications, as cross-reactivity with other RAS family members is a common issue due to sequence similarity.
Phylogenetic analysis provides valuable context for understanding RASL12's evolutionary relationships and potential functional conservation:
Sequence collection: Gather RASL12 sequences across species and related RAS proteins
Multiple sequence alignment: Use tools like MUSCLE, MAFFT, or Clustal Omega
Tree construction: Apply maximum likelihood or Bayesian methods
Functional inference: Identify conserved domains and species-specific variations
The RAS subfamily shows strong evolutionary conservation, with most branches containing representatives from humans, flies, and worms . Phylogenetic analysis of RAS subfamily sequences suggests 12 structural or functional branches with varying degrees of conservation across species . Positioning RASL12 within this evolutionary framework can provide insights into its potential functional specialization.
Researchers studying RASL12 should consult the following databases and resources:
The Harmonizome database indicates that RASL12 has 2,805 functional associations with biological entities spanning 8 categories extracted from 73 datasets , providing a rich source of information for hypothesis generation.
Based on the available information, several critical knowledge gaps and future research directions for RASL12 include:
Structural characterization: Crystal or cryo-EM structures of RASL12 in different nucleotide-bound states
Effector identification: Comprehensive mapping of RASL12 interaction partners
Regulatory mechanisms: Identification of GEFs and GAPs that regulate RASL12 activity
Physiological functions: Development of animal models to understand in vivo roles
Disease associations: Systematic analysis of RASL12 alterations in human diseases
Addressing these gaps will require interdisciplinary approaches combining structural biology, biochemistry, cell biology, and systems-level analyses.
The RAS-Like Family 12 (RASL12) is a member of the Ras superfamily of small GTPases. These proteins play crucial roles in various cellular processes, including signal transduction, cell growth, and differentiation. The Ras superfamily is known for its involvement in transmitting signals within cells, which ultimately control gene transcription and influence fundamental processes such as cell growth and differentiation .
RASL12, like other members of the Ras superfamily, has a conserved GTP-binding core. This core is essential for its function as a molecular switch, cycling between an active GTP-bound state and an inactive GDP-bound state . The protein’s ability to bind and hydrolyze GTP is critical for its role in signal transduction .
The RASL12 protein is predicted to enable GDP binding activity, GTP binding activity, and GTPase activity. It is involved in signal transduction and is active in the plasma membrane . The protein’s structure, as determined by crystallography, reveals a typical GTPase fold, which is crucial for its function .
RASL12 is involved in various cellular processes, including cell proliferation, differentiation, and survival. It plays a role in the regulation of intracellular signaling pathways that control these processes . The protein is expressed in multiple tissues, including lymphoid tissue, bone marrow, testis, and skeletal muscle .
Mutations in Ras genes, including those in the Ras superfamily, can lead to the production of permanently activated Ras proteins. These mutations can cause unintended and overactive signaling inside the cell, even in the absence of incoming signals. Overactive Ras signaling can ultimately lead to cancer . Therefore, Ras inhibitors are being studied as potential treatments for cancer and other diseases associated with Ras overexpression .
Recombinant RAS-Like Family 12 (Human) is produced using recombinant DNA technology. This involves inserting the gene encoding RASL12 into a suitable expression system, such as bacteria or yeast, to produce the protein in large quantities. Recombinant proteins are used in various research applications, including studying protein function, drug development, and therapeutic interventions .