Rat G4 (Uncharacterized protein C6orf47 homolog) remains largely uncharacterized in terms of its specific biological functions. Current research indicates it is homologous to the human C6orf47 protein, with counterparts in other species including Macaca mulatta (Rhesus monkey) and Mus musculus (house mouse) .
The protein's structure has not been fully elucidated through crystallography. Based on sequence analysis, it shares evolutionary conserved regions with other mammalian C6orf47 homologs, suggesting potential functional importance across species. Researchers should consider comparative genomic approaches when investigating this protein's function, particularly examining conserved domains across species.
Methodological approach: To study G4's function, implement a multi-faceted approach combining:
Differential expression analysis in various tissues
Knockdown/knockout studies followed by phenotypic characterization
Protein-protein interaction studies using techniques like co-immunoprecipitation
Expression profiling under various experimental conditions
G4 expression appears to be highly responsive to various chemical compounds. According to the Rat Genome Database annotations, multiple chemicals demonstrate significant effects on G4 expression :
| Chemical Compound | Effect on G4 Expression | Evidence | Reference |
|---|---|---|---|
| (-)-epigallocatechin 3-gallate | Decreased expression when co-treated with potassium chromate | ISO | PMID:22079256 |
| 2,2',4,4'-Tetrabromodiphenyl ether | Decreased expression | ISO | PMID:31675489 |
| 2,3,7,8-tetrachlorodibenzodioxine | Decreased expression | EXP | PMID:33387578 |
| 2,4,6-tribromophenol | Decreased expression | ISO | PMID:31675489 |
| 2-hydroxypropanoic acid (Lactic Acid) | Decreased expression | ISO | PMID:30851411 |
| 3,3',5,5'-tetrabromobisphenol A | Decreased expression | ISO | PMID:31675489 |
| all-trans-retinoic acid | Increased expression | ISO | N/A |
| aflatoxin B1 | Decreased methylation | ISO | PMID:30157460 |
Methodological approach: When studying chemical regulation of G4:
Design dose-response experiments with time-course measurements
Use qPCR to validate expression changes at the mRNA level
Employ ChIP-seq to investigate transcription factor binding and epigenetic modifications
Consider investigating multiple cell types, as regulation may be tissue-specific
The most validated method for G4 protein detection is enzyme-linked immunosorbent assay (ELISA). Commercial ELISA kits for rat G4 are available with the following specifications :
| Parameter | Specification |
|---|---|
| Test Range | 0.156 ng/ml - 10 ng/ml |
| Sample Types | Serum, plasma, tissue homogenates, cell culture supernatants and other biological fluids |
| Detection Method | Colorimetric |
| Assay Type | Quantitative |
| Storage | Shipped at 4°C, store according to kit instructions |
| Recommended Dilution | Optimal dilutions should be determined by end user |
| Availability | Typically shipped within 5-15 working days |
| Validity | 6 months |
For accurate results, sample concentrations must be diluted to mid-range of the kit detection limits
ELISA kits are optimized for detection of native samples rather than recombinant proteins
Western blotting can serve as a complementary method for verification, though specific antibodies may need to be validated
Mass spectrometry-based approaches can be used for unbiased detection and quantification
G4 has several identified homologs across mammalian species :
| Species | Gene Symbol | Protein Accession |
|---|---|---|
| Rattus norvegicus (Norway rat) | G4 | NP_001003975.1 |
| Homo sapiens (human) | C6orf47 | NP_067007.3 |
| Macaca mulatta (Rhesus monkey) | C4H6orf47 | XP_001112658.1 |
| Pan troglodytes (chimpanzee) | C6H6orf47 | NP_001231632.1 |
| Canis lupus familiaris (dog) | C12H6orf47 | XP_003639451.1 |
| Bos taurus (cattle) | C23H6orf47 | NP_001094737.1 |
| Mus musculus (house mouse) | D17H6S53E | NP_258438.1 |
While sequence conservation suggests functional importance, the precise biological roles remain to be fully elucidated across species. The differences in chromosomal location (e.g., chromosome 6 in humans versus chromosome 4 in macaques) reflect evolutionary genomic reorganization.
Perform comparative genomic analysis to identify conserved domains
Use phylogenetic analysis to reconstruct evolutionary relationships
Consider the utility of model organisms based on sequence conservation
Examine expression patterns in homologous tissues across species
While comprehensive expression profiling of G4 across all rat tissues is not fully documented in the provided literature, research approaches should focus on:
Quantitative assessment using qPCR across major tissues
Developmental time course studies from embryonic to adult stages
Comparison with known expression patterns of human C6orf47
Preliminary data suggests G4 gene expression may be altered during brain development and differentiation processes, though it wasn't specifically highlighted among the significantly regulated genes in sexually mature male rat brains .
Design tissue-specific qPCR assays using validated primer sets
Consider RNA-seq for unbiased transcriptomic profiling
Implement in situ hybridization to determine cell-type specificity
Use reporter constructs to monitor expression in real-time in vivo
Production of recombinant G4 protein can be approached using several expression systems:
| Expression System | Advantages | Considerations |
|---|---|---|
| E. coli | Rapid, high yield, cost-effective | Potential issues with folding, lacks post-translational modifications |
| Mammalian (HEK293/CHO) | Proper folding, post-translational modifications | Lower yield, higher cost, longer production time |
| Insect (Baculovirus) | High yield, some post-translational modifications | Intermediate complexity and cost |
| Cell-free | Rapid, avoids toxicity issues | Limited scaling, may require optimization |
Clone the G4 coding sequence into appropriate expression vectors, considering fusion tags for purification (His, GST, MBP)
For E. coli expression:
Use BL21(DE3) or Rosetta strains to enhance expression
Optimize induction conditions (temperature, IPTG concentration, time)
Consider fusion partners to improve solubility
For mammalian expression:
HEK293T or CHO cells offer robust expression capabilities
Consider stable cell line development for repeated production
Purification strategy:
Implement multi-step chromatography (affinity, ion exchange, size exclusion)
Validate protein identity by mass spectrometry
Assess purity by SDS-PAGE and activity by functional assays
CRISPR-Cas9 gene editing presents a powerful approach to investigate G4 function in rat models:
gRNA design:
Design several guide RNAs targeting different exons of G4
Validate guides in vitro before in vivo application
Consider potential off-target effects using computational predictions
Delivery methods:
For cell lines: plasmid transfection, lentiviral vectors
For in vivo: adeno-associated virus (AAV), electroporation, or direct pronuclear injection for germline modifications
Verification of editing:
PCR-based genotyping
Sanger sequencing of targeted regions
Western blotting to confirm protein knockout
qPCR to assess transcript levels
Phenotypic characterization:
Systematic profiling of physiological parameters
Molecular analysis (transcriptomics, proteomics)
Context-specific functional assays based on expression patterns
Complementary approaches:
Consider conditional knockout systems (Cre-loxP)
Implement rescue experiments to confirm specificity
Use knockin strategies for reporter or tagged versions
While specific post-translational modifications (PTMs) of G4 have not been comprehensively characterized in the provided literature, this represents an important area for investigation.
PTM profiling by mass spectrometry:
Purify G4 protein from different tissues/conditions
Perform MS/MS analysis with PTM-specific enrichment strategies
Common modifications to investigate: phosphorylation, glycosylation, ubiquitination, acetylation
Site-directed mutagenesis:
Generate mutants at predicted modification sites
Assess functional consequences in appropriate assays
Modification-specific antibodies:
Develop or acquire antibodies recognizing specific PTMs
Use for western blotting and immunoprecipitation
Enzyme inhibitor studies:
Use inhibitors of kinases, phosphatases, acetyltransferases, etc.
Determine effects on G4 function and localization
Disease context:
Investigate if alterations in PTMs are associated with pathological conditions
Compare PTM patterns between normal and disease states
Researchers working with recombinant G4 protein should be aware of several technical challenges:
Solubility issues:
G4 may form aggregates or inclusion bodies in bacterial expression systems
Optimization strategies: lower induction temperature, reduced IPTG concentration, solubility tags
Stability concerns:
The half-life of purified G4 may be limited
Buffer optimization is critical (pH, salt concentration, additives)
Consider storage in aliquots with cryoprotectants
Detection limitations:
Functional assays:
Due to limited characterization, establishing functional assays is challenging
Consider comparative approaches with homologs from other species
Leverage interacting partners to develop indirect functional readouts
Crystallization challenges:
Uncharacterized proteins often present difficulties in crystallization
Consider alternative structural approaches (NMR, cryo-EM)
Experiment with various truncation constructs to identify stable domains
Current understanding of G4's involvement in disease mechanisms is limited, but several research approaches can help elucidate potential roles:
Expression analysis in disease models:
Compare G4 expression in normal versus disease tissues
Use quantitative techniques (qPCR, western blot, immunohistochemistry)
Genetic association studies:
Investigate if variations in G4 are associated with specific conditions
Examine SNPs or structural variants in patient cohorts
Pathway analysis:
Based on interaction partners, determine if G4 participates in known disease pathways
Consider systems biology approaches to position G4 in cellular networks
Knockdown/knockout consequences:
Evaluate if G4 depletion causes disease-relevant phenotypes
Analyze in context of specific tissue types or developmental stages
Therapeutic potential:
Assess if G4 could serve as a biomarker or therapeutic target
Investigate potential for antibody or small molecule targeting
The involvement of C6orf47 in human Lynch syndrome suggests potential roles for G4 in DNA mismatch repair pathways , though direct evidence for the rat homolog requires further investigation.
The search results indicate that G4 expression may be regulated by methylation changes, as demonstrated by the effects of aflatoxin B1 and B2, which decrease methylation of the G4 promoter .
Methylation analysis:
Bisulfite sequencing of the G4 promoter region
Compare methylation patterns across tissues and conditions
Investigate the effects of DNA methyltransferase inhibitors
Histone modification profiling:
ChIP-seq for various histone marks (H3K4me3, H3K27ac, H3K27me3)
Determine correlation between modifications and expression levels
Chromatin accessibility:
ATAC-seq or DNase-seq to assess open chromatin regions
Identify potential regulatory elements affecting G4 expression
Transcription factor binding:
ChIP-seq for relevant transcription factors
Perform reporter assays with mutated binding sites
Non-coding RNA interactions:
Investigate if miRNAs or lncRNAs regulate G4 expression
Perform RNA immunoprecipitation to identify RNA-protein interactions