The "Recombinant Danio rerio Abhydrolase domain-containing protein FAM108C1 (fam108c1)" refers to a protein found in zebrafish (Danio rerio) that contains an abhydrolase domain and belongs to the FAM108C1 protein family. Abhydrolases are a broad class of enzymes that catalyze the hydrolysis of various chemical bonds, and the FAM108C1 protein is one member of a larger family of proteins with diverse functions . Recombinant proteins are produced using genetic engineering techniques, where the gene encoding the protein is inserted into a host cell, which then produces the protein.
Abhydrolase Domain: The presence of an abhydrolase domain suggests that this protein functions as a hydrolase, catalyzing the breakdown of chemical bonds through the addition of water .
Danio rerio:* This protein is found in zebrafish, a common model organism in biological research .
Protein Family: FAM108C1 is part of a protein family, implying it shares structural and possibly functional similarities with other members of this family .
While the specific function of Danio rerio FAM108C1 may not be thoroughly documented, understanding its characteristics allows for informed assumptions about its potential roles:
Hydrolytic Activity: As an abhydrolase, it likely participates in metabolic processes by hydrolyzing esters, amides, or other functional groups.
Gene Regulation: Some proteins in related pathways can regulate gene expression. For example, Far Upstream Element Binding Protein 1 (FUBP1) affects the expression of genes like c-Myc and p21 .
Viral Processes: In some viruses, proteins with hydrolase activity are crucial for replication and assembly. For example, in African Swine Fever Virus (ASFV), several proteins with enzymatic activity play roles in viral DNA replication, transcription, and mRNA capping .
Studies of FAM108C1 and related proteins often employ a variety of biochemical and molecular techniques:
Structural Analysis: Techniques such as X-ray crystallography and cryo-EM are used to determine the protein's three-dimensional structure, as seen in studies of ASFV proteins .
Functional Assays: These assays determine the specific enzymatic activity of the abhydrolase domain, often involving substrate specificity and reaction kinetics.
Interaction Studies: Methods like co-immunoprecipitation and cross-linking mass spectrometry can identify proteins that interact with FAM108C1, giving clues about its biological role.
Gene Expression Analysis: Techniques such as qPCR and RNA-seq can measure the expression levels of FAM108C1 under different conditions.
Cellular Localization: Fluorescence microscopy can determine where the protein is located within the cell .
Research on proteins like FAM108C1 can have broader implications:
Drug Discovery: Understanding the structure and function of these proteins can aid in developing targeted therapies. For example, inhibitors of farnesyltransferase have been developed as anticancer agents .
Understanding Disease Mechanisms: Proteins like FAM81A, which regulates postsynaptic protein condensation, show how these molecules can be critical in neurological functions .
Comparative Genomics: Studying proteins across different species can provide insights into evolutionary relationships and conserved functions .
Hydrolyzes fatty acids from S-acylated cysteine residues in proteins. Exhibits depalmitoylating activity towards nras.
Abhydrolase domain-containing protein FAM108C1 (fam108c1) is a member of the abhydrolase superfamily expressed in Danio rerio (zebrafish). It contains a characteristic alpha/beta hydrolase fold and is believed to play roles in lipid metabolism and cellular signaling pathways. The recombinant form of this protein can be produced in expression systems such as E. coli for research purposes . While the precise physiological function remains under investigation, its homologs in other species have been implicated in membrane lipid remodeling and metabolic processes.
Recombinant Danio rerio FAM108C1 can be effectively produced in prokaryotic expression systems, with E. coli being the most common host organism . For standard research applications, bacterial expression offers advantages of high yield and cost-effectiveness. The protein is typically produced with affinity tags (such as His-tag or GST-tag) to facilitate purification. Alternative expression systems including yeast, insect cells, or mammalian cells may provide improved post-translational modifications when needed for specific functional studies, although these systems typically result in lower yields and higher production costs.
Validation of recombinant FAM108C1 activity can be performed through multiple complementary approaches:
Enzymatic activity assays using synthetic substrates that contain ester or amide bonds
Thermal shift assays to assess protein stability and ligand binding
Western blotting with specific antibodies to confirm protein identity and integrity
Mass spectrometry to verify protein sequence and post-translational modifications
Size exclusion chromatography to assess oligomeric state and proper folding
Activity should be compared against appropriate positive and negative controls, including heat-inactivated protein samples and known substrates for abhydrolase domain-containing proteins.
For optimal stability of Recombinant Danio rerio FAM108C1, the following storage conditions are recommended:
Storage Period | Recommended Conditions | Additives | Notes |
---|---|---|---|
Short-term (<1 week) | 4°C | 5-10% glycerol | Avoid repeated freeze-thaw cycles |
Medium-term (1-6 months) | -20°C | 20% glycerol | Aliquot to minimize freeze-thaw cycles |
Long-term (>6 months) | -80°C | 50% glycerol or lyophilized | Reconstitute in appropriate buffer |
The protein should be maintained in a pH-buffered solution (typically pH 7.4-8.0) with stabilizing agents such as glycerol and potentially reducing agents if the protein contains critical cysteine residues . Avoid shipping without dry ice as temperature fluctuations can significantly reduce activity.
FAM108C1 has potential roles in zebrafish kidney development and function, though specific mechanisms remain under investigation. The zebrafish pronephric kidney shares significant structural and functional homology with mammalian nephrons, making it a valuable model for studying kidney development and disease .
When investigating FAM108C1 in kidney research:
Expression patterns can be visualized in zebrafish larvae using fluorescently-tagged antibodies against FAM108C1 and confocal microscopy
Morpholino knockdown or CRISPR/Cas9 gene editing can be used to assess phenotypic effects of FAM108C1 depletion
Microinjection of modified mRNA can be employed for rescue experiments to confirm specificity
Functional assays can measure pronephric filtration rates using fluorescent dextran clearance assays
Significantly, zebrafish larvae offer advantages for nephrotoxicity studies as demonstrated with gentamicin , potentially providing insights into how FAM108C1 may modulate nephrotoxic responses or contribute to kidney development.
Systems biology approaches provide comprehensive frameworks for characterizing FAM108C1 function:
Multi-omics integration: Combine transcriptomics, proteomics, and metabolomics data from wild-type and FAM108C1-deficient zebrafish to construct functional networks
Pathway analysis: Employ bioinformatic tools to identify enriched pathways and biological processes associated with FAM108C1 expression or depletion
Protein-protein interaction mapping: Use techniques such as proximity labeling, co-immunoprecipitation combined with mass spectrometry, or yeast two-hybrid screening to identify direct interaction partners
Computational modeling: Develop predictive models for FAM108C1 function based on sequence homology with better-characterized abhydrolase family members
Cross-species validation: Compare findings in zebrafish with mammalian models to establish evolutionary conservation of function
Implementing these approaches requires careful experimental design with appropriate controls and statistical analyses to account for biological variability .
CRISPR/Cas9 gene editing provides powerful tools for studying FAM108C1 function through targeted genetic manipulation:
Strategy | Application | Advantages | Considerations |
---|---|---|---|
Complete knockout | Phenotypic analysis of loss-of-function | Straightforward interpretation | May have developmental lethality |
Conditional knockout | Temporal control of gene inactivation | Avoids developmental confounds | Requires more complex genetic tools |
Knock-in mutations | Study specific domains or variants | Models disease-relevant mutations | Lower efficiency than knockouts |
Reporter fusion | Visualize expression patterns | Tracks protein localization | May interfere with protein function |
For optimal CRISPR/Cas9 editing efficiency:
Design multiple guide RNAs targeting conserved functional domains of FAM108C1
Validate editing efficiency using T7 endonuclease assays or direct sequencing
Screen F0 mosaic founders for germline transmission
Establish stable lines through careful breeding and genotyping
Confirm knockout at protein level using specific antibodies
Remember that off-target effects must be controlled through careful guide RNA design and validation with rescue experiments using wild-type FAM108C1 mRNA microinjection.
Several key challenges exist when attempting to correlate in vitro enzymatic characterization with in vivo function:
Physiological substrate identification: The natural substrates of FAM108C1 in zebrafish may differ from synthetic substrates used in vitro, requiring unbiased metabolomic approaches to identify endogenous targets
Cellular context: The protein may require specific cellular cofactors, membrane environments, or post-translational modifications not present in recombinant systems
Developmental and tissue-specific regulation: Expression and activity of FAM108C1 likely varies across developmental stages and tissues, necessitating temporal and spatial resolution in analysis
Redundant enzymatic functions: Other hydrolases may compensate for FAM108C1 deficiency in vivo, masking phenotypes observed in knockout models
Technical limitations: Quantifying enzymatic activity in living zebrafish remains challenging and may require development of specific biomarkers or reporter systems
These challenges highlight the importance of integrating multiple experimental approaches, from biochemical characterization to in vivo functional studies, to develop a comprehensive understanding of FAM108C1 biology.
When designing zebrafish models to study FAM108C1 function, consider these methodological approaches:
Selection of developmental stages: Embryonic and larval stages (24-120 hours post-fertilization) offer advantages of transparency and rapid development for high-throughput screening
Genetic background considerations: Use established wild-type strains (AB, TU, or WIK) with known genetic backgrounds to minimize variability
Control groups: Include appropriate controls such as:
Wild-type siblings from the same clutch
Sham-injected controls for microinjection studies
Non-targeting guide RNA controls for CRISPR studies
Vehicle controls for drug treatment studies
Sample size determination: Power analysis should be performed to determine appropriate sample sizes based on expected effect sizes
Housing conditions standardization: Maintain consistent temperature (28.5°C), photoperiod (14h light/10h dark), and water quality parameters
Ethical considerations: Design experiments following the 3Rs principles (Replacement, Reduction, Refinement) with appropriate ethical approvals
For imaging studies of zebrafish larvae, confocal microscopy with specific staining protocols allows visualization of pronephric structures to assess FAM108C1 localization and function within kidney tissues .
Multiple complementary approaches can be used to assess FAM108C1 expression and localization:
Quantitative PCR (qPCR): Measures mRNA expression levels across different tissues and developmental stages
Requires careful primer design specific to zebrafish FAM108C1
Normalized against stable reference genes (e.g., ef1α, rpl13a)
In situ hybridization: Visualizes spatial expression patterns of FAM108C1 mRNA
Whole-mount techniques are effective for embryos and larvae
Section-based approaches for adult tissues
Immunohistochemistry/Immunofluorescence: Determines protein localization at cellular and subcellular levels
May require generation of zebrafish-specific antibodies
Co-staining with organelle markers clarifies subcellular localization
Transgenic reporter lines: Generation of FAM108C1-fluorescent protein fusions
Enables real-time visualization in living zebrafish
Can be combined with tissue-specific promoters for targeted expression
Western blotting: Quantifies protein expression across different tissues
Requires careful sample preparation and loading controls
Can detect post-translational modifications with specific antibodies
For confocal imaging of kidney structures, techniques using fluorescent dextran conjugates can be employed to assess functional aspects alongside localization studies .
RNA-seq analysis for understanding FAM108C1 regulatory networks should follow these methodological steps:
Experimental design:
Compare wild-type vs. FAM108C1 knockout/knockdown models
Include biological replicates (minimum n=3 per condition)
Consider developmental time points and tissue specificity
Quality control and preprocessing:
Assess read quality with tools like FastQC
Trim adapters and low-quality bases
Align to zebrafish reference genome (GRCz11/danRer11)
Differential expression analysis:
Use established tools (DESeq2, edgeR) with appropriate statistical thresholds
Apply false discovery rate correction for multiple testing
Validate key findings with qPCR
Network analysis:
Functional enrichment:
Analyze enriched Gene Ontology terms and pathways
Compare with known abhydrolase domain protein functions
Cross-reference with human ortholog data when available
Integration with other omics data:
Combine with proteomics and metabolomics when available
Develop predictive models for FAM108C1 regulatory networks
The analysis should distinguish between direct and indirect effects of FAM108C1 perturbation by incorporating time-course data when possible .
When designing zebrafish-based drug screening assays targeting FAM108C1 function:
Assay development:
Establish clear, quantifiable phenotypic readouts related to FAM108C1 function
Develop high-content imaging protocols for automated analysis
Optimize compound delivery methods (water exposure vs. microinjection)
Screening logistics:
Determine appropriate developmental stages for treatment (typically 24-120 hpf)
Standardize embryo collection and dechorionation procedures
Implement appropriate positive and negative controls
Dosing considerations:
Establish concentration ranges that balance efficacy and toxicity
Account for absorption, distribution, and metabolism differences between zebrafish and mammals
Consider exposure duration and timing relative to developmental events
Validation strategies:
Confirm target engagement through direct binding assays
Perform dose-response studies to establish potency
Validate hits in secondary assays with different readouts
Translational aspects:
Consider pharmacokinetic differences between zebrafish and mammals
Validate findings in mammalian cell culture or other model organisms
Assess structural similarities between zebrafish and human FAM108C1 orthologs
Zebrafish larvae provide an excellent model for nephrotoxicity studies, making them particularly valuable for screening compounds that might affect FAM108C1 function in kidney tissues .
Emerging research trends for FAM108C1 in zebrafish models include:
Integration of CRISPR/Cas9 genome editing with high-throughput phenotypic screening
Application of single-cell RNA sequencing to characterize cell type-specific roles
Development of zebrafish reporter lines for real-time visualization of FAM108C1 activity
Comparative studies between zebrafish and mammalian FAM108C1 orthologs
Investigation of potential roles in disease models, particularly kidney-related disorders
Systems biology approaches that integrate multi-omics data to build comprehensive functional networks
These developments are enabling more sophisticated understanding of FAM108C1 biology while leveraging the unique advantages of zebrafish as a vertebrate model system, including optical transparency, external development, and genetic tractability.
Translating zebrafish FAM108C1 findings to human applications requires:
Comparative genomics analysis:
Establish orthology relationships between zebrafish FAM108C1 and human counterparts
Compare protein domain structures and key functional residues
Assess conservation of regulatory elements and expression patterns
Validation in mammalian models:
Confirm key findings in mammalian cell cultures
Validate in mouse models when appropriate
Consider human tissue samples or organoids for direct relevance
Disease relevance assessment:
Explore associations with human conditions through database mining
Investigate potential biomarker applications
Consider therapeutic targeting strategies if disease associations are established
Methodological considerations:
Account for species-specific differences in experimental design
Apply appropriate statistical methods for cross-species comparisons
Implement rigorous controls to validate translational relevance
The zebrafish model offers particularly strong translational potential for kidney-related studies due to the structural and functional similarities between zebrafish pronephros and human nephrons , potentially making FAM108C1 findings especially relevant to human nephrology research.
Current limitations in FAM108C1 research include:
Limited functional characterization:
The physiological substrates remain largely unknown
Specific signaling pathways and interaction partners are not fully characterized
Solution: Apply unbiased substrate screening and interactome analysis
Technical challenges:
Lack of highly specific antibodies for zebrafish FAM108C1
Challenges in measuring enzymatic activity in vivo
Solution: Develop improved tools including CRISPR knock-in tags and activity-based probes
Knowledge gaps in regulatory mechanisms:
Translational barriers:
Species differences may limit direct application to human health
Compensatory mechanisms in model systems can mask phenotypes
Solution: Validate findings across multiple model systems and human samples