FAM206A is a gene that encodes a protein known as F206A_HUMAN in humans. It is also referred to by synonyms such as CG-8, C9ORF6, and SIMIATE . The protein has been associated with numerous functional associations across different categories, including molecular profiles, organisms, chemicals, and diseases .
FAM206A has been shown to interact with other proteins, such as HMBOX1, in human protein-protein interaction studies. These interactions are typically identified through high-throughput methods like yeast two-hybrid screens . The interaction between FAM206A and HMBOX1 suggests potential roles in cellular processes, although the exact nature of these roles remains to be elucidated.
Given the lack of specific data on recombinant Xenopus laevis Protein FAM206A, we can look at general trends in protein research and interactions. For example, the human interactome map (HuRI) provides insights into how proteins like FAM206A might interact with others in complex networks .
Note: While we prioritize shipping the format currently in stock, please specify your format preference in order notes for customized preparation.
Note: Standard shipping includes blue ice packs. Dry ice shipping requires prior arrangement and incurs additional charges.
The tag type is determined during production. Please specify your desired tag type for preferential development.
KEGG: xla:443761
UniGene: Xl.47795
FAM206A (Family with sequence similarity 206 member A) in Xenopus laevis is a protein that shares structural characteristics with other FAM proteins found in the secretory pathway. While specific functions of FAM206A remain under investigation, research suggests its potential role in cytoskeletal organization during early embryonic development, similar to other characterized proteins in Xenopus such as actin-depolymerizing factor/cofilin. The significance lies in understanding conserved developmental pathways, as many FAM family proteins demonstrate important functions across vertebrate species.
The most effective expression system depends on experimental requirements and downstream applications. Based on related recombinant Xenopus protein production methods:
| Expression System | Advantages | Disadvantages | Tag Compatibility |
|---|---|---|---|
| E. coli | High yield, cost-effective, rapid production | May lack post-translational modifications, potential inclusion body formation | His, GST, MBP tags all compatible |
| Baculovirus/insect cells | Better post-translational modifications, proper folding | Higher cost, longer production time | His, FLAG, HA tags recommended |
| Mammalian cells | Most authentic post-translational modifications | Most expensive, lower yields | Multiple tag options, flexible positioning |
For basic structural studies and antibody production, E. coli systems similar to those used for other Xenopus proteins can be effective when optimized with appropriate solubility tags . For functional studies requiring proper folding and modifications, insect or mammalian expression systems may be preferable.
A multi-step purification strategy is recommended for achieving high purity FAM206A protein:
Initial capture: Immobilized metal affinity chromatography (IMAC) for His-tagged FAM206A, similar to protocols used for other Xenopus recombinant proteins
Intermediate purification: Ion exchange chromatography to separate based on charge properties
Polishing: Size exclusion chromatography to remove aggregates and achieve final purity
Typical purification conditions for Xenopus recombinant proteins involve buffering at pH 7.5-8.0 with appropriate protease inhibitors. When using His-tagged constructs, consideration should be given to tag position (N- or C-terminal) based on predicted protein structure to ensure accessibility during purification. Purification yields should be verified by SDS-PAGE with expected purity >90% for most research applications.
Structural integrity verification should employ multiple complementary techniques:
Circular dichroism (CD) spectroscopy to assess secondary structure elements
Thermal shift assays to determine protein stability
Dynamic light scattering (DLS) to evaluate homogeneity and detect aggregation
Limited proteolysis to identify stable domains
When analyzing CD data, comparison with predicted secondary structure based on sequence analysis provides valuable validation. For thermal stability, establishing a baseline denaturation profile is essential for evaluating the effects of buffer conditions or mutations in future experiments.
For studying FAM206A interactions in developmental contexts, a multi-method approach yields the most reliable results:
| Method | Application | Sensitivity | In vivo relevance |
|---|---|---|---|
| Co-immunoprecipitation | Endogenous interactions | Medium | High |
| Proximity labeling (BioID) | Transient interactions | High | Medium-High |
| Yeast two-hybrid | Binary interactions | High | Low |
| Pull-down assays | Direct binding | Medium | Low |
| FRET/BRET | Real-time interactions | High | Medium |
When designing experiments, consider that many Xenopus proteins involved in cytoskeletal organization exhibit stage-specific interactions during embryonic development . Particular attention should be paid to temporal expression patterns and potential developmental regulation of these interactions.
While specific data on FAM206A phosphorylation is limited, insights can be drawn from related proteins in Xenopus. Actin-depolymerizing factor/cofilin in Xenopus shows regulated phosphorylation crucial for its function in cytokinesis . Similarly, FAM206A may undergo phosphorylation regulating its activity.
Potential kinases to investigate include:
Fam20 family kinases, known to phosphorylate secretory pathway proteins
Cytoskeletal regulatory kinases (LIMK, ROCK)
Cell cycle-regulated kinases (CDKs)
Phosphorylation site prediction tools suggest potential sites, but experimental verification through mass spectrometry following in vitro kinase assays is essential. Phosphomimetic mutants (S/T to D/E) and phosphodeficient mutants (S/T to A) can help evaluate the functional significance of identified sites.
To characterize FAM206A expression patterns during Xenopus development:
RT-qPCR analysis across developmental stages from oocyte to tadpole
Whole-mount in situ hybridization to visualize spatial distribution
Immunohistochemistry with specific antibodies for protein localization
Western blotting of stage-specific embryo lysates
Based on patterns observed with other developmentally regulated proteins in Xenopus, expression may vary significantly across developmental stages. For example, Xenopus ADF/cofilin shows differential localization during early cleavage stages and increased expression in neuroectoderm-derived tissues, notochord, somites, and epidermis during stages 18-34 . Similar stage-specific analyses should be conducted for FAM206A.
For FAM206A functional analysis in Xenopus embryos:
| Approach | Advantages | Limitations | Controls Required |
|---|---|---|---|
| Morpholino knockdown | Rapid, dose-adjustable | Potential off-target effects | Standard control MO, rescue experiments |
| CRISPR/Cas9 knockout | Specific, complete KO | Mosaic effects in F0 | Multiple sgRNAs, off-target analysis |
| mRNA overexpression | Quick gain-of-function | Non-physiological levels | Dose-response, inactivated construct |
| Transgenic lines | Tissue-specific manipulation | Time-consuming | Multiple founder lines |
When evaluating phenotypes, detailed analysis should include:
Survival rates and developmental timing
Morphological defects (measured quantitatively)
Tissue-specific impacts (histological analysis)
Molecular consequences (changes in marker gene expression)
Based on approaches used for other cytoskeletal regulators in Xenopus, injection of constitutively active or inhibitory constructs at specific blastomeres can reveal cell-autonomous functions .
Comparative analysis across species provides evolutionary context for FAM206A function:
Sequence conservation analysis using multiple sequence alignment
Expression pattern comparison across zebrafish, mouse, and human tissues
Functional complementation tests (can orthologs rescue Xenopus phenotypes?)
Xenopus laevis being pseudotetraploid may have two FAM206A allelic variants with slight sequence differences, similar to the situation observed with Xenopus ADF/cofilins which differ in 12 residues spread throughout the sequence . Analysis of both homeologs and their expression patterns can provide insights into potential subfunctionalization.
Domain-specific expression requires careful boundary determination:
Perform bioinformatic domain prediction analysis
Design constructs with 5-10 amino acid overlaps at domain boundaries
Test multiple constructs for each domain
Optimize buffer conditions specific to each domain
| Domain Type | Buffer Recommendations | Common Challenges | Solubility Enhancement |
|---|---|---|---|
| Hydrophobic domains | Detergent inclusion (0.05-0.1% Triton X-100) | Aggregation | Fusion to MBP tag |
| Charged domains | Higher salt concentration (300-500mM NaCl) | Nonspecific interactions | Optimize pH based on pI |
| Catalytic domains | Include specific cofactors | Activity loss | Express with substrate analogs |
Expression in E. coli at lower temperatures (16-18°C) after induction can improve folding of difficult domains. For domains requiring post-translational modifications, eukaryotic expression systems may be necessary.
Development of specific antibodies against Xenopus FAM206A requires:
Selection of immunogenic epitopes unique to FAM206A
Avoid conserved regions shared with other FAM family proteins
Target regions with predicted surface exposure
Consider species-specific regions for Xenopus-specific antibodies
Immunization strategy
Use of full-length protein versus synthetic peptides
Multiple host species for different application needs
Affinity purification against immobilized antigen
Validation methods
Western blotting against recombinant protein and endogenous protein
Immunoprecipitation efficiency testing
Immunohistochemistry with appropriate controls (pre-immune serum, blocking peptides)
Testing in FAM206A knockdown/knockout embryos
For monoclonal antibody development, consider epitopes that are accessible in native protein conformation for applications requiring detection of non-denatured protein.
Several complementary high-throughput approaches can identify developmental stage-specific interaction partners:
IP-Mass Spectrometry
Tag-based pulldown from embryo lysates at different developmental stages
Quantitative comparison using SILAC or TMT labeling
Stringent statistical analysis to identify specific interactors
Proximity labeling approaches
BioID or TurboID fusion expression in embryos
Stage-specific biotinylation followed by streptavidin pulldown
MS identification of labeled proteins
Yeast two-hybrid screening
Using domain-specific baits against Xenopus embryonic cDNA libraries
Verification of positive hits in embryo contexts
Protein arrays
Screening of recombinant FAM206A against arrays of developmental proteins
Validation of hits using orthogonal methods
Data analysis should include ontology enrichment, interaction network construction, and comparison with interactomes of related proteins to identify unique and shared functions.
| Issue | Possible Causes | Recommended Solutions |
|---|---|---|
| Low expression yield | Codon bias, protein toxicity | Optimize codon usage, use tight expression control systems |
| Inclusion body formation | Rapid expression, improper folding | Lower induction temperature, co-express chaperones |
| Proteolytic degradation | Protease sensitivity, unstructured regions | Include protease inhibitors, optimize purification speed |
| Loss of activity | Denaturation, cofactor loss | Include stabilizing agents, identify required cofactors |
| Aggregation post-purification | Improper buffer conditions, concentration issues | Screen buffer components, add stabilizing agents |
For protein-specific issues, consider characterizing the biochemical properties of FAM206A through limited proteolysis to identify stable domains, and thermal shift assays to optimize buffer conditions .
Functional validation depends on the identified biological activities of FAM206A. Based on approaches used for related proteins:
Binding assays
Surface plasmon resonance (SPR) or bio-layer interferometry (BLI) for interaction kinetics
Fluorescence polarization for small molecule interactions
ELISA-based binding assays for antibody validation
Cell-based functional assays
Microinjection of recombinant protein into Xenopus embryos to assess activity
Rescue experiments in FAM206A-depleted embryos or cells
Analysis of cytoskeletal organization if FAM206A has cytoskeletal functions
Biochemical activity assays
If enzymatic activity is identified, develop specific substrate-based assays
Monitor potential post-translational modification status
Each validation method should include positive and negative controls to ensure specificity of the observed effects.
Establishing rigorous quality control metrics ensures reproducible research results:
Purity assessment
SDS-PAGE analysis (>95% purity recommended)
Size exclusion chromatography profile consistency
Mass spectrometry verification of intact mass
Identity confirmation
Peptide mass fingerprinting
Western blot with specific antibodies
N-terminal sequencing for first 5-10 amino acids
Functional metrics
Specific activity measurements (if enzymatic)
Binding affinity consistency (if receptor or ligand)
Secondary structure content by CD spectroscopy
Stability parameters
Thermal denaturation profile
Time-course activity retention
Freeze-thaw stability testing
Record and maintain detailed batch records of production conditions, purification steps, and quality metrics for each preparation to ensure experimental reproducibility .
Design considerations for chimeric FAM206A constructs include:
Domain boundary determination
Secondary structure prediction to avoid disrupting structural elements
Evolutionary conservation analysis to identify functional units
Experimental validation through limited proteolysis
Linker design
Flexible linkers (GGGGS)n for independent domain function
Rigid linkers (EAAAK)n when specific orientation is required
Cleavable linkers for post-expression separation
Tag positioning
Terminal tags versus internal tags based on predicted structure
Impact assessment of tags on domain function
Split-tag approaches for complex assemblies
Functional readouts
Domain-swapping with related proteins to assess conservation of function
Deletion series to map minimal functional regions
Point mutations of conserved residues to probe specific functions
When expressing chimeric constructs, additional optimization may be required due to altered folding properties compared to native protein .
CRISPR-Cas9 approaches for studying FAM206A in Xenopus should consider:
sgRNA design
Target early exons to ensure functional disruption
Design multiple sgRNAs targeting different regions
Check for potential off-targets throughout Xenopus genome
Consider both homeologs if targeting in Xenopus laevis
Delivery methods
Microinjection at one-cell stage for global knockout
Targeted injection at specific blastomeres for tissue-specific analysis
Inducible CRISPR systems for temporal control
Efficiency assessment
T7 endonuclease assay for mutation detection
High-resolution melt analysis for rapid screening
Deep sequencing to characterize indel spectrum
Western blotting to confirm protein loss
Advanced applications
Homology-directed repair for tagging endogenous FAM206A
Base editing for specific amino acid substitutions
Prime editing for precise modifications
Given potential functional redundancy, consider compound targeting of related family members or paralogs for complete functional analysis.
Integrative -omics approaches provide comprehensive understanding of FAM206A function:
Experimental design considerations
Stage-specific sampling across development
Tissue-specific analyses when appropriate
FAM206A loss-of-function and gain-of-function conditions
Biological replicates (minimum n=3) for statistical power
RNA-seq analysis workflow
Differential expression analysis between control and FAM206A-manipulated embryos
Temporal expression clustering to identify co-regulated genes
Pathway enrichment analysis for functional interpretation
Comparison with other developmental regulator datasets
Proteomics integration
Global proteome changes following FAM206A manipulation
Phosphoproteomics to identify signaling pathway alterations
Correlation between transcriptome and proteome changes
Protein-protein interaction network construction
Validation approaches
qRT-PCR for key differentially expressed genes
Western blotting for protein-level changes
Whole-mount in situ hybridization for spatial validation
Functional testing of identified pathways
Implementation of artificial transcriptional systems may enhance the production of recombinant FAM206A for these advanced studies, utilizing approaches similar to those developed for other recombinant proteins in plant expression systems .