Recombinant Mouse FERM domain-containing protein 4A (Frmd4a), partial, refers to a genetically engineered version of the mouse Frmd4a protein, which is part of the FERM superfamily. This protein plays a crucial role in cell structure, transport, and signaling, particularly in regulating cell polarity in epithelial cells and neurons . The term "partial" indicates that this recombinant protein may not include the full-length sequence of the native Frmd4a protein.
Frmd4a is involved in connecting the Par3 complex with Arf6 signaling through cytohesin-1, which is essential for epithelial polarization and membrane trafficking . It acts as a scaffolding protein, facilitating interactions between various cellular components to maintain cell polarity and integrity .
Studies have shown that Frmd4a interacts with Par3 and cytohesin-1 to form a complex that ensures accurate activation of Arf6, a key player in actin cytoskeleton dynamics and membrane trafficking . This complex is crucial for junctional remodeling and epithelial polarization.
In cancer studies, particularly in tongue squamous cell carcinoma, Frmd4a has been identified as a potential therapeutic target. Overexpression of Frmd4a is associated with tumor progression, and silencing its expression can inhibit cell proliferation and reduce migration and invasion capabilities .
Mutations in the Frmd4a gene have been linked to neurological phenotypes, including microcephaly and intellectual disability. Additionally, Frmd4a mutations may influence the risk of schizophrenia and late-onset Alzheimer's disease by affecting tau secretion through cytohesin-Arf6 signaling .
Protein Name | Function | Score |
---|---|---|
CYTH1 | Promotes guanine-nucleotide exchange on ARF1, ARF5, and ARF6 | 0.919 |
CYTH4 | Promotes guanine-nucleotide exchange on ARF1 and ARF5 | 0.720 |
CYTH2 | Acts as a guanine-nucleotide exchange factor (GEF) for ARF1, ARF3, and ARF6 | 0.683 |
CYTH3 | Promotes guanine-nucleotide exchange on ARF1 and ARF6 | 0.681 |
RUFY2 | RUN and FYVE domain containing 2 | 0.586 |
Assay Type | Effect of FRMD4A Silencing |
---|---|
Cell Proliferation | Inhibited at 24 and 48 hours post-transfection |
Cell Cycle | Arrested in G1 phase |
Migration and Invasion | Reduced |
Frmd4a is a scaffolding protein that regulates epithelial cell polarity by linking ARF6 activation to the PAR3 complex. It functions redundantly with FRMD4B in epithelial polarization and may regulate MAPT secretion via ARF6 signaling activation.
FRMD4A is a scaffolding protein belonging to the FERM domain-containing protein family. It is localized in the cytoplasm and cytoskeleton, where it binds molecules in the undercoat of cell-to-cell adherens junctions . The protein plays critical roles in:
Regulation of cell polarity in epithelial cells and neurons
Cell structure maintenance
Transport processes
Signal transduction pathways
FRMD4A contains conserved domains that facilitate its interaction with other cellular components. The FERM domain (Four-point-one, Ezrin, Radixin, Moesin) is particularly important for mediating protein-protein interactions at the interface between the plasma membrane and the cytoskeleton.
FRMD4A is expressed in multiple tissues throughout the body, with notably higher expression levels in the brain . In normal epidermal tissue, FRMD4A is primarily expressed in the basal layer of human epidermis . The protein shows differential expression patterns during development and in various physiological states.
Research has shown:
Expression in neural tissues during development
Tissue-specific regulation that may be influenced by various transcription factors
When designing experiments to study FRMD4A expression, researchers should consider using tissue-specific controls and evaluating expression across multiple developmental timepoints for comprehensive understanding.
FRMD4A has been identified as significantly upregulated in head and neck squamous cell carcinoma (HNSCC), with high expression levels correlating with increased risks of relapse . Functional studies have revealed several critical aspects of FRMD4A's role in cancer:
FRMD4A silencing decreases growth and metastasis of human SCC xenografts in skin and tongue models
It reduces SCC proliferation and intercellular adhesion
Attenuation of FRMD4A stimulates caspase-3 activity and increases expression of terminal differentiation markers
FRMD4A appears to influence the Hippo signaling pathway, as its attenuation causes nuclear accumulation of YAP (Yes-associated protein)
Treatment with HSP90 inhibitor 17-DMAG or ligation of CD44 with hyaluronan causes nuclear depletion of FRMD4A and reduced SCC growth and metastasis
These findings suggest FRMD4A may represent a novel therapeutic target in HNSCC treatment strategies. For researchers investigating cancer pathways, examining the interaction between FRMD4A and the Hippo signaling pathway may provide valuable insights into mechanisms of cancer progression.
FRMD4A has been identified as a genetic risk factor for late-onset Alzheimer's disease through genome-wide haplotype association studies . Several key findings illuminate its potential role:
Specific haplotypes within FRMD4A on Chr.10p13 have been consistently associated with Alzheimer's disease risk (OR: 1.68; 95% CI: 1.43–1.96; P=1.1 × 10^-10)
FRMD4A polymorphisms are associated with plasma Aβ42/Aβ40 ratio, with the best signal at P=5.4 × 10^-7
The protein interacts with Arf6, which controls APP processing
FRMD4A mutations can disrupt tau secretion by activating cytohesin-Arf6 signaling
This suggests FRMD4A may influence amyloid precursor protein (APP) metabolism and contribute to the development of Alzheimer's disease pathology. Researchers studying Alzheimer's disease mechanisms should consider investigating FRMD4A's impact on both amyloid and tau pathways.
Mutations in the FRMD4A gene have been associated with several neurodevelopmental conditions, including:
Corpus callosum anomalies (abnormal genu and splenium of corpus callosum)
Global developmental delay and intellectual disability
Macrocephaly or microcephaly (reported cases show both phenotypes)
Ataxia
A case report identified compound heterozygous missense mutations in FRMD4A [c.1830G>A, p.(Met610Ile) and c.2973G>C, p.(Gln991His)] in a 3-year-old boy with these features . The differences in head circumference phenotypes (macrocephaly versus microcephaly) between reported cases suggest phenotypic variability, though intellectual disability/global developmental delay and ataxia appear to be consistent features.
When investigating FRMD4A function in vitro, several methodological approaches have proven effective:
Gene Silencing Techniques:
siRNA or shRNA-mediated knockdown to assess loss-of-function effects
CRISPR-Cas9 gene editing for complete knockout models
Recombinant Protein Expression:
Expression of tagged FRMD4A constructs (e.g., GFP-tagged) to visualize subcellular localization
Domain-specific mutants to identify functional regions
Cell-Based Assays:
Proliferation assays to assess impact on cell growth
Migration and invasion assays to evaluate metastatic potential
Intercellular adhesion assays
Caspase activity assays to measure apoptotic effects
Terminal differentiation marker expression analysis
Protein Interaction Studies:
Co-immunoprecipitation to identify binding partners
Proximity ligation assays to confirm protein-protein interactions in situ
Yeast two-hybrid screening for novel interactors
Research has specifically demonstrated successful application of these techniques in studying FRMD4A's role in cancer progression, showing that silencing FRMD4A reduced SCC proliferation and intercellular adhesion while stimulating caspase-3 activity .
Several animal models have proven valuable for in vivo FRMD4A research:
Mouse Models:
Xenograft models using human SCC cells with manipulated FRMD4A expression have been successfully used to study cancer progression and metastasis
Frmd4a knockout or conditional knockout mice can help understand developmental roles
Mouse models with specific mutations that mimic human FRMD4A variants found in patients
Zebrafish Models:
Experimental Considerations:
When designing xenograft experiments, consider both subcutaneous implantation and orthotopic models (e.g., tongue for HNSCC studies) as demonstrated in previous research
Include appropriate controls for genetic background effects
For neurodevelopmental studies, comprehensive behavioral testing alongside structural brain imaging may reveal important phenotypes
The choice of model should align with specific research questions, with consideration for tissue-specific expression patterns and phenotypic readouts relevant to the disease or developmental process being studied.
Accurate quantification of FRMD4A expression in tissue samples requires multiple complementary approaches:
Protein Detection Methods:
Immunohistochemistry (IHC) for localization in tissue sections, particularly useful for observing expression in specific cell types (e.g., basal layer of epidermis)
Western blotting for semi-quantitative protein level determination
ELISA for more precise quantification
Mass spectrometry for absolute quantification and post-translational modification analysis
mRNA Detection Methods:
RT-qPCR for relative quantification of transcript levels
RNA-seq for comprehensive transcriptome analysis and alternative splicing detection
In situ hybridization to visualize spatial expression patterns in tissues
Methodological Considerations:
Use multiple antibodies targeting different epitopes to confirm specificity
Include appropriate positive and negative control tissues
Consider analysis of different splice variants that may have distinct functions
Normalize expression data to appropriate reference genes/proteins that are stable in the tissue of interest
Data Analysis Approach:
Employ quantitative image analysis for IHC data
Use statistical methods appropriate for expression data distribution
Consider analyzing expression correlation with clinical outcomes or other molecular markers
When working with clinical samples, correlating FRMD4A expression with patient outcomes (as demonstrated in HNSCC studies showing association with relapse risk ) can provide valuable prognostic insights.
FRMD4A plays a critical role in neuronal development and polarization through several mechanisms:
Cell Polarity Regulation:
FRMD4A functions as a scaffolding protein that regulates cell polarity in neurons
Its interaction with cytoskeletal components is essential for establishing neuronal polarity during development
Mutations disrupt these interactions, potentially affecting neuronal migration and axon/dendrite specification
Molecular Pathway Involvement:
FRMD4A likely interfaces with conserved polarity complexes (Par, Crumbs, Scribble)
It may regulate membrane protein trafficking needed for polarized growth
The protein potentially influences small GTPase signaling that directs cytoskeletal rearrangements
Corpus Callosum Development:
Experimental Approaches to Study These Effects:
Primary neuronal cultures with FRMD4A knockdown/mutation to assess polarity establishment
Time-lapse microscopy of developing neurons to track morphological changes
Analysis of axon/dendrite specification markers in mutant conditions
In vivo brain imaging in animal models with FRMD4A mutations
The compound heterozygous mutations [c.1830G>A, p.(Met610Ile) and c.2973G>C, p.(Gln991His)] identified in patients alter protein side chains and likely disrupt FRMD4A function and interactions with other molecules , potentially explaining the observed neurodevelopmental phenotypes.
The relationship between FRMD4A and the Hippo signaling pathway represents an important area of investigation with implications for both normal development and disease:
Observed Molecular Interactions:
Functional Consequences:
Hippo pathway dysregulation affects cell proliferation, apoptosis, and differentiation
FRMD4A's influence on YAP may explain its role in cancer progression
This relationship could be relevant to developmental processes requiring coordinated growth control
Experimental Evidence:
Research Approaches:
Co-immunoprecipitation studies to identify direct vs. indirect interactions
Phosphorylation analysis of YAP in FRMD4A mutant conditions
Transcriptional reporter assays for YAP target genes with FRMD4A manipulation
Investigation of upstream Hippo kinases (MST1/2, LATS1/2) in relation to FRMD4A
Understanding this relationship more fully could provide insights into both normal tissue homeostasis mechanisms and potential therapeutic approaches for conditions like cancer where Hippo pathway dysregulation contributes to pathology.
The relationship between FRMD4A polymorphisms and amyloid-beta metabolism in Alzheimer's disease is complex and multifaceted:
Genetic Association Evidence:
Molecular Mechanisms:
Experimental Evidence:
Analytical Approaches:
To study these effects, researchers should consider:
Cell-based assays measuring APP processing with different FRMD4A variants
In vivo models expressing human FRMD4A risk variants to assess Aβ dynamics
Correlation studies between FRMD4A genotypes and CSF/plasma biomarkers in patients
Structural biology approaches to understand how polymorphisms affect protein function
This research area has significant potential for identifying new therapeutic targets in Alzheimer's disease, as modulating FRMD4A function might influence amyloid metabolism through novel mechanisms distinct from direct secretase targeting.
Researchers face several significant challenges when attempting to distinguish the phenotypic effects of different FRMD4A mutations:
Phenotypic Variability:
Mutation Characterization Challenges:
Complex Genetic Architecture:
Compound heterozygosity (as seen in reported cases) makes it difficult to attribute specific phenotypic features to individual mutations
Possible interactions with other genetic modifiers may influence phenotypic expression
The large size of the FRMD4A gene (23 exons) increases the complexity of comprehensive genetic screening
Research Strategies to Address These Challenges:
Detailed clinical phenotyping across multiple cases with the same mutation
Development of isogenic cell lines with specific mutations for direct comparison
Animal models expressing human mutations to assess developmental trajectories
Structure-function studies to determine how different domains contribute to protein activity
Analysis of different mutations' effects on interaction partners (protein-protein interaction networks)
When confronted with contradictory findings about FRMD4A function across different experimental systems, researchers should consider several methodological and biological factors:
Context-Dependent Functions:
FRMD4A may have distinct roles in different cell types or tissues (e.g., epithelial cells versus neurons)
Developmental timing may influence protein function and pathway interactions
Disease states may alter normal protein functions or reveal conditional phenotypes
Technical Considerations:
Different antibodies may have varying specificities and recognize different FRMD4A epitopes or isoforms
The use of tagged constructs (e.g., GFP-FRMD4A) might affect protein localization or function
Knockdown efficiency and off-target effects vary between siRNA/shRNA approaches
Experimental System Limitations:
In vitro systems may not recapitulate the complex microenvironment of tissues
Overexpression systems may create artifactual interactions or phenotypes
Animal models may have species-specific differences in FRMD4A function or regulation
Recommended Analytical Approach:
Triangulate findings using multiple complementary techniques
Consider dose-dependent effects – partial vs. complete loss of function
Validate key findings across different cell types or model systems
Carefully document experimental conditions that may influence outcomes
Design experiments to directly test contradictory hypotheses
The multifunctional nature of FRMD4A as both a scaffolding protein in cell polarity and a potential regulator of signaling pathways (e.g., Hippo, Arf6) suggests its effects may be highly context-dependent, explaining some apparent contradictions in research findings.
When analyzing FRMD4A genetic association data in disease cohorts, researchers should consider these statistical approaches and considerations:
Association Testing Strategies:
Single-SNP analysis using logistic regression models adjusted for relevant covariates (age, sex, population structure)
Haplotype-based approaches have proven effective in identifying FRMD4A associations with Alzheimer's disease that might be missed in traditional GWAS
Gene-based tests that aggregate multiple variants (e.g., SKAT, burden tests) for rare variant analysis
Study Design Considerations:
Specific Statistical Methods:
For quantitative traits (e.g., Aβ plasma levels), general linear models under additive genetic models adjusted for relevant covariates
Inverse-variance weighting (fixed-effects meta-analysis) to investigate homogeneity of effects across studies
z-score transformations for biomarker data to normalize distributions across centers/studies
Multiple Testing Correction:
Data Visualization:
Forest plots for meta-analysis results
Manhattan plots for genome-wide significance
Linkage disequilibrium (LD) plots to understand haplotype structure
The FRMD4A Alzheimer's disease association study demonstrated the value of these approaches, identifying significant associations that were consistently replicated across multiple cohorts and validated through biomarker correlations .
Integrating FRMD4A functional data with clinical observations requires sophisticated approaches to bridge laboratory findings and patient outcomes:
Translational Research Frameworks:
Biomarker Development:
Genotype-Phenotype Correlation:
Create detailed phenotyping protocols for patients with FRMD4A variants
Compare clinical features across mutation types using standardized assessments
Develop databases integrating genetic, molecular, and clinical data
Methodological Integration Strategies:
Patient-derived cell models (iPSCs, organoids) to study mutation effects in relevant cell types
Humanized animal models expressing patient-specific mutations
Systems biology approaches to contextualize FRMD4A within broader pathway networks
Statistical and Computational Approaches:
Machine learning models to identify patterns in complex datasets
Bayesian networks to infer causal relationships between molecular findings and clinical outcomes
Pathway enrichment analysis to contextualize FRMD4A functions
This integration is particularly important for neurodevelopmental disorders associated with FRMD4A mutations, where understanding the connection between molecular dysfunction and clinical manifestations could guide development of targeted interventions or therapies.