Domains: Contains an N-terminal Ras association (RA) domain and a C-terminal Salvador/RASSF/Hippo (SARAH) domain, facilitating interactions with Ras proteins and Hippo pathway components like MST1/2 .
Isoforms: Three splice variants (RASSF3A, RASSF3B, RASSF3C). Only RASSF3A retains functional RA and SARAH domains .
Property | Details |
---|---|
Amino Acids | 261 (Recombinant form includes a 23-amino acid His-tag) |
Molecular Weight | 30 kDa (Recombinant) / 28.6 kDa (Native) |
Expression System | E. coli (Recombinant production) |
Apoptosis Induction: Overexpression of RASSF3 activates pro-apoptotic proteins (Bax, caspase-3) and suppresses anti-apoptotic Bcl-2, dependent on p53 .
Cell Cycle Arrest: Triggers G1/S phase arrest by stabilizing p53 and modulating cyclin-dependent kinases .
DNA Repair: Depletion of RASSF3 impairs DNA damage response, leading to polyploidy .
RASSF3 binds MST1/2 kinases but induces apoptosis independently of the Hippo cascade, instead relying on MOAP1-Bax mitochondrial pathways .
Promoter Hypermethylation: Observed in pituitary somatotroph adenomas, correlating with reduced mRNA expression .
Downregulation: Reported in lung, uterus, colon, and liver cancers compared to normal tissues .
Tissue Type | Methylation Level (Tumor vs. Normal) | Expression Impact |
---|---|---|
Pituitary Adenomas | ↑ 2.5-fold | ↓ 60% mRNA |
Lung Tumors | N/A | ↓ 40–50% mRNA |
RASSF3 (Ras Association Domain Family Member 3) is a protein-coding gene that belongs to the RASSF family of tumor suppressors. It functions as an effector protein that interacts with specific GTPases and plays important roles in cell growth regulation, apoptosis signaling, and mitochondrial homeostasis. The RASSF family proteins have been linked to Hippo signaling pathways, cell cycle regulation, and maintenance of genomic stability . RASSF3 contains a characteristic Ras Association (RA) domain that enables it to interact with specific GTPases, though unlike some other family members, RASSF3 does not appear to bind directly to classical RAS proteins such as H/K/NRAS .
The RASSF3 gene contains multiple regions including a promoter region and intronic sequences that contain important regulatory elements. Notable polymorphic sites include rs6581580 T>G in the promoter region, and rs7313765 G>A, rs12311754 G>C, and rs1147098 T>C in the intron 1 region . The coding sequence of RASSF3 contains the information necessary to produce a functional protein that includes the characteristic Ras Association domain. In certain model organisms like Phaethon lepturus (White-tailed tropicbird), the RASSF3 ORF has been documented to be 543bp in length, though human RASSF3 may differ in specific nucleotide composition .
Researchers typically employ several experimental systems to investigate RASSF3 function:
Cell culture models with manipulated RASSF3 expression (overexpression or knockdown)
Recombinant protein expression systems for protein-protein interaction studies
Patient-derived samples for clinical correlations
Genotyping assays for SNP analysis
For protein interaction studies, GST-pull down assays using GST-RA domains from RASSF3 are frequently utilized to investigate binding with potential partner proteins like MIRO GTPases . TaqMan methodology is commonly employed for genotyping RASSF3 SNPs in patient populations .
While the search results don't provide comprehensive information about RASSF3 expression patterns across all human tissues, research indicates that RASSF family members generally show tissue-specific expression profiles. Like other RASSF family proteins, RASSF3 expression may be regulated by epigenetic mechanisms, particularly promoter methylation, which can lead to downregulation in certain cancers . Expression analysis typically requires techniques such as qRT-PCR, western blotting, or immunohistochemistry to detect RASSF3 at mRNA or protein levels in specific tissues.
RASSF3 has been identified as the first potential effector of mitochondrial GTPases MIRO1 and MIRO2, which play crucial roles in mitochondrial homeostasis, trafficking, and PINK1/PARKIN-mediated mitophagy . Unlike RASSF5 which binds to classical RAS proteins (H/K/NRAS) in a nucleotide-dependent manner, RASSF3 binds strongly to the nGTPase domains of MIRO1 and MIRO2 independent of the loaded nucleotide state .
Additional binding partners include:
RHOBTB1 and RHOBTB2 from the RHO subfamily
RHOF and RHOH GTPases
ARL4C, ARL11, and ARL5C from the ARF subfamily
This interaction network suggests RASSF3 may integrate signals from multiple small GTPase families to regulate diverse cellular processes including mitochondrial function and cytoskeletal organization. The table below summarizes the key RASSF3 binding partners:
GTPase Family | Specific Binding Partners | Binding Characteristics | Cellular Functions |
---|---|---|---|
MIRO GTPases | MIRO1, MIRO2 | Nucleotide-independent, direct binding | Mitochondrial homeostasis, trafficking, mitophagy |
RHO subfamily | RHOBTB1, RHOBTB2, RHOF, RHOH | Via RA domain | Cytoskeletal organization, cell morphology |
ARF subfamily | ARL4C, ARL11, ARL5C | Via RA domain | Membrane trafficking, cytoskeletal remodeling |
Several SNPs in the RASSF3 gene have been investigated for their potential association with cancer risk, particularly in squamous cell carcinoma of the head and neck (SCCHN). The most studied polymorphisms include rs6581580 T>G in the promoter region and rs7313765 G>A, rs12311754 G>C, and rs1147098 T>C in intron 1 .
Research methodologies to establish these associations typically involve:
Case-control studies comparing SNP frequencies between cancer patients and healthy controls
Logistic regression analysis to calculate odds ratios (ORs) with adjustment for known cancer risk factors
Functional studies such as Electrophoretic Mobility Shift Assay (EMSA) to determine how SNPs affect protein-DNA binding
Assessment of potential gene-gene interactions, such as between RASSF3 and MDM2 variants
The mechanisms by which these SNPs might influence cancer risk could involve altered gene expression, modified splicing patterns, or changes in protein function that affect RASSF3's tumor suppressor activities.
RASSF3's direct binding to mitochondrial GTPases MIRO1 and MIRO2 suggests a significant role in mitochondrial dynamics . MIRO proteins are known to regulate mitochondrial trafficking, distribution, and quality control processes including PINK1/PARKIN-mediated mitophagy.
To experimentally validate RASSF3's role in mitochondrial dynamics, researchers could employ:
Live-cell imaging techniques to visualize mitochondrial movement and morphology in cells with manipulated RASSF3 expression
Co-localization studies using fluorescently tagged RASSF3 and mitochondrial markers
Mitochondrial fractionation followed by western blotting to confirm RASSF3's presence in mitochondrial compartments
In vitro binding assays with purified proteins to characterize the RASSF3-MIRO interaction:
Functional assays measuring mitochondrial membrane potential, respiration, or mitophagy rates in response to RASSF3 manipulation
Understanding this relationship may provide insights into diseases with mitochondrial dysfunction components, including neurodegenerative disorders and cancer.
Several complementary approaches can be employed to study RASSF3-GTPase interactions:
GST pull-down assays: The RA domain of RASSF3 can be expressed as a GST-fusion protein and used to precipitate potential GTPase binding partners from cell lysates or to test binding with purified GTPases. This method has successfully demonstrated direct binding between RASSF3 and MIRO GTPases .
Co-immunoprecipitation (Co-IP): This technique can detect interactions in a more physiological context using antibodies against endogenous proteins or epitope tags.
Nucleotide loading assays: To test nucleotide dependency of interactions, GTPases can be loaded with different nucleotides (GDP, GTP, or non-hydrolyzable GTP analogs like GMPPNP or GTPγS) .
Systematic screening approaches: Comprehensive interaction mapping can be performed by testing binding between RASSF3 and multiple GTPases from different subfamilies (RAS, RHO, ARF) .
Protocol example for direct binding assay:
Express and purify GST-RASSF3-RA domain from E. coli
Express and purify target GTPases (e.g., MIRO1/2)
Exchange GTPases with different nucleotides (GDP, GTP, GTP analogs)
Mix purified proteins and precipitate with glutathione beads
Perform extensive washing to remove non-specific binding
Analyze bound proteins by SDS-PAGE and western blotting or mass spectrometry
For effective genotyping of RASSF3 SNPs in human populations, researchers commonly employ the following methods:
TaqMan methodology: This approach is particularly effective for analyzing known SNPs (e.g., rs6581580, rs7313765, rs12311754, rs1147098) and can be performed in 384-well plates using an ABI-Prism 7900 instrument . The typical protocol includes:
PCR-RFLP (Restriction Fragment Length Polymorphism): This method is useful for SNPs that create or abolish restriction enzyme recognition sites .
Next-generation sequencing approaches: For comprehensive analysis of multiple SNPs or discovery of novel variants.
Digital PCR: For highly accurate quantification of allele frequencies.
Statistical analysis of genotyping data typically includes:
Hardy-Weinberg equilibrium testing using goodness-of-fit χ² test
Case-control comparisons using χ² tests
Calculation of odds ratios (ORs) and 95% confidence intervals (CIs) using logistic regression
Adjustment for known confounding factors (age, sex, lifestyle factors)
Selecting appropriate cell models is critical for studying RASSF3 function. Based on the available research information, the following approaches are recommended:
Cell line selection:
Cancer cell lines from tissues where RASSF3 has pathological relevance
Primary cells to study physiological functions
Cell lines with easily manipulatable mitochondrial dynamics for studying RASSF3-MIRO interactions
Expression systems:
Transient transfection with tagged RASSF3 constructs
Stable cell lines with inducible RASSF3 expression
CRISPR/Cas9-mediated knockout or knockin models
siRNA or shRNA for RASSF3 knockdown studies
Functional challenges to reveal RASSF3's role:
Apoptotic stimuli to assess cell death regulation
Cell cycle synchronization to study cell cycle effects
Mitochondrial stressors (e.g., CCCP, rotenone) to investigate mitochondrial functions
RAS pathway activators or inhibitors to examine signaling integration
Co-expression studies with binding partners:
MIRO1/2 for mitochondrial function studies
RHO family GTPases for cytoskeletal regulation studies
Other RASSF family members for comparative functional analysis
Readout systems:
Live-cell imaging for dynamic processes
Flow cytometry for cell cycle and apoptosis quantification
Biochemical assays for signaling pathway activation
Mitochondrial function assays (membrane potential, respiration, calcium handling)
While the search results do not provide comprehensive information about RASSF3 in cancer, they do suggest potential links that warrant further investigation. Some key points include:
SNP associations: Functional single nucleotide polymorphisms of RASSF3 have been studied in relation to squamous cell carcinoma of the head and neck (SCCHN) risk. This research included 1087 SCCHN patients and 1090 cancer-free controls of non-Hispanic white population .
Tumor suppressor properties: RASSF3 belongs to the RASSF family, which includes known tumor suppressors like RASSF1A and RASSF5. RASSF1A is downregulated in >80% of lung cancers, and mice lacking RASSF1A are prone to tumorigenesis . By extension, RASSF3 may have similar tumor suppressor functions.
Epigenetic regulation: RASSF genes are frequently hypermethylated to inhibit their expression, suggesting a mechanism by which their tumor suppressor functions might be impaired in cancer .
GTPase interactions: RASSF3's interactions with various GTPases might influence cell growth and proliferation pathways relevant to cancer development. Unlike RASSF5, which interacts with growth-promoting RAS GTPases, RASSF3 binds to different GTPases, potentially mediating distinct effects on cellular growth .
Experimental approaches to investigate RASSF3's role in cancer typically include methylation analysis, expression profiling in tumor vs. normal tissues, functional studies in cancer cell lines, and animal models with altered RASSF3 expression.
RASSF3 has been identified as "the first potential effector of the MIRO GTPases," which are important regulators of mitochondrial homeostasis, trafficking, and PINK1/PARKIN-mediated mitophagy . This finding suggests RASSF3 may play significant roles in mitochondrial-related diseases:
Neurodegenerative disorders: Mitochondrial dysfunction is implicated in conditions like Parkinson's disease, where PINK1/PARKIN-mediated mitophagy is crucial. RASSF3's interaction with MIRO1/2 might influence these processes.
Metabolic diseases: Proper mitochondrial function is essential for energy metabolism, and RASSF3 might affect metabolic disorders through its mitochondrial interactions.
Cancer metabolism: Mitochondrial dynamics influence cancer cell metabolism, and RASSF3's role might extend to metabolic reprogramming in tumors.
Research approaches to investigate these connections could include:
Genetic association studies between RASSF3 variants and mitochondrial-related diseases
Functional studies examining how RASSF3 affects MIRO-dependent mitochondrial processes:
Mitochondrial trafficking and distribution
Mitochondrial quality control and mitophagy
Mitochondrial morphology and dynamics
Disease models (cell-based and animal) with manipulated RASSF3 expression to assess phenotypic outcomes relevant to mitochondrial diseases
Biochemical characterization of how RASSF3-MIRO interactions affect MIRO GTPase activity and downstream effectors
The direct binding between RASSF3 and MIRO GTPases provides a mechanistic foundation for exploring how this interaction might contribute to disease pathogenesis or potentially offer therapeutic targets.
Emerging technologies offer new opportunities to investigate RASSF3 functions:
CRISPR-based screening approaches: Genome-wide or focused CRISPR screens can identify genes that interact functionally with RASSF3, potentially revealing new pathways or mechanisms.
Proximity labeling techniques: BioID or APEX2-based approaches could identify proteins that interact with or are near RASSF3 in living cells, providing a more comprehensive interactome.
Live-cell imaging with optogenetic tools: These approaches allow temporal and spatial control of RASSF3 activity, enabling precise dissection of its functions in specific cellular compartments.
Single-cell omics techniques: These can reveal cell-to-cell variability in RASSF3 expression or function, potentially identifying specific cell populations where RASSF3 plays critical roles.
Patient-derived organoids or iPSC models: These systems provide more physiologically relevant contexts for studying RASSF3 function in human tissues.
When researchers encounter contradictory findings about RASSF3 function, several methodological approaches can help reconcile these discrepancies:
Context-dependent analysis: Systematically evaluate whether contradictions arise from differences in:
Cell or tissue types studied
Experimental conditions (stress, growth factors, etc.)
Expression levels of RASSF3 or interacting partners
Specific isoforms or variants analyzed
Technical validation across platforms:
Employ multiple independent techniques to verify key findings
Use both gain-of-function and loss-of-function approaches
Validate in multiple cell lines or model systems
Comprehensive literature analysis:
Perform systematic reviews or meta-analyses where appropriate
Contact authors of conflicting studies to discuss methodological differences
Consider establishing collaborative projects to directly address contradictions
Mechanistic dissection:
Identify specific domains or residues responsible for different functions
Examine post-translational modifications that might alter function
Investigate conditional interactions that may explain context-specific effects
By applying these methodological approaches, researchers can develop more nuanced models of RASSF3 function that account for apparent contradictions in the literature.
Ras Association Domain-Containing Protein 3 (RASSF3) is a member of the Ras association domain family (RASSF), which plays a crucial role in intracellular signal transduction pathways. This protein is encoded by the RASSF3 gene and is involved in various cellular processes, including tumor suppression and signal transduction.
The RASSF3 gene is located on chromosome 12 and encodes a protein that contains a Ras association (RA) domain. This domain is essential for binding to GTP-bound Ras, a member of the Ras superfamily of small GTPases . The RA domain facilitates the localization of RASSF3 to the plasma membrane, where it can interact with other signaling molecules to propagate downstream signaling events .
RASSF3 is primarily known for its role as a tumor suppressor. It is involved in the regulation of cell proliferation, apoptosis, and cell cycle progression. The protein exerts its tumor-suppressive effects through various mechanisms, including the modulation of the Hippo signaling pathway . The Hippo pathway is a critical regulator of organ size and tissue homeostasis, and its dysregulation is often associated with cancer development.
RASSF3, along with other members of the C-RASSF subclass (RASSF1-6), contains a C-terminal coiled-coil motif known as the Salvador/RASSF/Hippo (SARAH) domain . This domain is involved in interactions with mammalian Ste20-like kinases, which are core components of the Hippo pathway. By interacting with these kinases, RASSF3 can influence the activity of downstream effectors, such as the transcriptional co-activators YAP and TAZ, thereby regulating cell proliferation and apoptosis.
The expression of RASSF3 is frequently downregulated in various human cancers due to DNA hypermethylation . This epigenetic modification leads to the silencing of the RASSF3 gene, contributing to tumor progression and poor prognosis. Restoring the expression of RASSF3 in cancer cells has been shown to inhibit tumor growth and induce apoptosis, highlighting its potential as a therapeutic target.
Human recombinant RASSF3 is produced using recombinant DNA technology, which involves the insertion of the RASSF3 gene into an expression vector, followed by the expression of the protein in a suitable host system, such as E. coli or mammalian cells. The recombinant protein can be purified and used for various research applications, including studies on its function, mechanism of action, and potential therapeutic uses.