Recombinant Danio rerio N-alpha-acetyltransferase 35 (naa35) functions as an auxiliary subunit of the NatC complex, which is responsible for N-terminal acetylation of proteins. This post-translational modification alters the electrostatic properties of substrate N-termini, potentially affecting protein folding, stability, half-life, interactions, and subcellular targeting . The protein is also known as Embryonic growth-associated protein (zEGAP) or MAK10 homolog in zebrafish . As part of the heterotrimeric NatC complex, naa35 works alongside the catalytic Naa30 subunit and a second auxiliary subunit Naa38 to co-translationally acetylate the N-termini of numerous target proteins .
The NatC complex, which includes naa35 as an auxiliary subunit, exhibits a strikingly different architecture compared to previously described N-terminal acetyltransferase (NAT) complexes . While the catalytic mechanism of acetyl transfer is conserved across NAT complexes, the heterotrimeric NatC complex has evolved a unique structure-function relationship. Crystal structure analysis has revealed that the NatC complex recognizes the first four amino acids of cognate substrates at the Naa30–Naa35 interface through a sequence-specific recognition mechanism . This structural divergence reflects how NAT machineries have evolved distinct architectures to acetylate specific subsets of target proteins .
Zebrafish (Danio rerio) models benefit significantly from studies utilizing recombinant naa35, particularly in developmental biology and neuroscience research. Since naa35 is also known as Embryonic growth-associated protein (zEGAP) , it likely plays important roles in early development. The methodological approach to studying naa35 function typically involves:
Loss-of-function studies using morpholinos or CRISPR-Cas9
Rescue experiments with recombinant naa35 protein
Comparative analysis with other vertebrate models
The recombinant protein allows researchers to conduct in vitro acetylation assays to characterize substrate preferences and compare enzymatic activities across developmental stages or tissue types. When designing such experiments, researchers should consider the proper controls and validation strategies to confirm specificity of the recombinant protein's activity.
For optimal storage and reconstitution of Recombinant Danio rerio naa35, researchers should follow these methodological guidelines:
Storage Protocol:
Store lyophilized form at -20°C/-80°C for up to 12 months
Store liquid form at -20°C/-80°C for up to 6 months
Avoid repeated freeze-thaw cycles, as this can compromise protein activity
Reconstitution Protocol:
Briefly centrifuge the vial before opening to bring contents to the bottom
Reconstitute in deionized sterile water to a concentration of 0.1-1.0 mg/mL
Add 5-50% glycerol (with 50% as the recommended final concentration)
Prepare small working aliquots to minimize freeze-thaw cycles
These guidelines are critical for maintaining protein stability and activity. The shelf life depends on multiple factors including storage state, buffer ingredients, storage temperature, and the intrinsic stability of the protein itself .
Validating the enzymatic activity of Recombinant Danio rerio naa35 requires a multifaceted approach, as naa35 is an auxiliary subunit rather than the catalytic component of the NatC complex. A comprehensive validation methodology includes:
Complex Reconstitution: Express and purify all three components of the NatC complex (naa35, Naa30, and Naa38) and reconstitute the complex in vitro.
Acetylation Assay: Monitor the transfer of an acetyl group from acetyl-CoA to specific peptide substrates using:
Substrate Specificity Analysis: Test various peptide substrates with different N-terminal sequences to confirm the expected substrate specificity of the NatC complex.
Controls:
When analyzing enzymatic data, researchers should be aware that some NATs are prone to substrate or product inhibition, which may affect kinetic measurements .
Co-expressing the complete functional NatC complex requires careful consideration of multiple factors to ensure proper complex formation and enzymatic activity. The methodological approach should include:
Construct Design:
Expression System Selection:
Purification Strategy:
Quality Control:
Assess complex integrity by dynamic light scattering or native PAGE
Confirm enzymatic activity using acetylation assays
Verify proper subunit stoichiometry using quantitative mass spectrometry
These considerations ensure the production of a functionally active NatC complex suitable for subsequent biochemical and structural studies.
Distinguishing between substrates of different NAT complexes in proteomic studies requires a sophisticated methodological approach that leverages the unique substrate preferences of each complex. For effective differentiation:
N-terminal Peptide Enrichment Techniques:
Differential Knockdown/Knockout Analysis:
Substrate Sequence Analysis:
NatA typically acetylates N-termini with small amino acids (Ala, Ser, Thr, Gly, Val) after initiator methionine removal
NatC preferentially acetylates proteins with hydrophobic N-terminal residues (Met-Leu, Met-Ile, Met-Phe, Met-Trp)
Create position-specific scoring matrices for each NAT complex based on confirmed substrates
Quantitative Proteomics Workflow:
| Step | Methodology | Output |
|---|---|---|
| Sample preparation | siRNA knockdown of specific NAT complex components | Cells with reduced expression of target NAT |
| Protein extraction and digestion | Tryptic digestion with optimized protocols for N-terminal peptides | Peptide mixture |
| N-terminal enrichment | TAILS or COFRADIC | Enriched N-terminal peptides |
| LC-MS/MS analysis | High-resolution mass spectrometry | Raw spectral data |
| Data analysis | Database searching with variable N-terminal acetylation | Identified acetylated and non-acetylated N-termini |
| Statistical analysis | Comparison of acetylation levels between conditions | Statistically significant differences in acetylation |
By systematically applying these approaches, researchers can confidently assign proteins as substrates of specific NAT complexes and understand the complex interplay between different acetylation machineries .
The structural features of naa35 that are critical for substrate recognition in the NatC complex have been elucidated through crystallographic studies. These structural elements create a unique molecular architecture that enables specific substrate binding:
Interface Formation with Naa30:
naa35 forms a critical interface with the catalytic Naa30 subunit, creating a substrate binding pocket
This interface is responsible for recognizing the first four amino acids of cognate substrates
The binding pocket accommodates the hydrophobic N-terminal sequences typically acetylated by NatC (Met-Leu, Met-Ile, Met-Phe, Met-Trp)
Conformational Changes:
Ribosome Association:
Evolutionary Divergence:
Unlike catalytic subunits, the large auxiliary subunits like naa35 share low sequence identity with other NAT complex auxiliary subunits
This divergence reflects the specialized roles these subunits play in substrate selection
The unique structural features of naa35 contribute to the distinct substrate specificity of the NatC complex
Understanding these critical structural features provides insights into the molecular basis of substrate selectivity and catalytic efficiency of the NatC complex, which can inform the design of specific inhibitors or engineered variants with altered specificity.
The expression and function of naa35 in Danio rerio show both conserved and species-specific characteristics when compared to its orthologs in other model organisms. This comparative analysis reveals important evolutionary insights:
Cross-Species Comparison:
Developmental Expression Patterns:
In zebrafish, naa35 (as zEGAP) may have specific roles in embryonic development
Temporal and spatial expression analyses across developmental stages reveal tissue-specific patterns
Functional studies using morpholinos or CRISPR-based approaches can elucidate developmental roles
Functional Conservation:
Experimental Approaches for Comparative Studies:
Complementation studies in yeast to test functional conservation
Heterologous expression of orthologs to assess interchangeability
Cross-species rescue experiments to determine functional equivalence
Structural studies to compare binding interfaces and substrate recognition mechanisms
These comparative analyses are particularly valuable for translating findings from model organisms to human disease contexts and for understanding the evolutionary conservation of N-terminal acetylation machinery .
Achieving high purity and yield of recombinant naa35 presents several challenges that researchers commonly encounter. These challenges and their methodological solutions include:
Protein Solubility Issues:
Challenge: naa35 is a large protein that may form inclusion bodies when expressed alone.
Solution: Co-express with partner proteins (Naa30 and Naa38) to promote proper folding and solubility.
Alternative: Optimize expression conditions (temperature, induction time, media composition) and consider fusion tags (SUMO, MBP) that enhance solubility .
Degradation During Purification:
Challenge: Proteolytic degradation during cell lysis and purification.
Solution: Include protease inhibitor cocktails, maintain cold temperatures throughout purification, and minimize processing time.
Monitoring: Track protein integrity via SDS-PAGE at each purification step.
Co-purification of Contaminants:
Low Expression Yield:
Complex Stability Issues:
Challenge: Dissociation of the NatC complex during purification.
Solution: Include stabilizing agents in buffers (glycerol, NaCl, reducing agents) and optimize pH and ionic strength.
Analysis: Use analytical size exclusion chromatography to monitor complex integrity.
Implementing these strategic approaches can significantly improve the yield and purity of recombinant naa35, facilitating downstream biochemical and structural studies.
Interpreting unexpected changes in enzymatic activity of the NatC complex containing recombinant naa35 requires systematic troubleshooting and analysis. Researchers should follow this methodological framework:
Establish Baseline Activity:
Before investigating changes, establish reproducible baseline activity measurements using standardized substrates and conditions.
Document key parameters (temperature, pH, buffer composition, substrate and cofactor concentrations).
Systematic Analysis of Activity Changes:
Investigate Structural Integrity:
Control Experiments:
Compare activity with fresh preparations of the complex.
Test activity of the catalytic subunit alone versus the complete complex.
Introduce known mutations that affect activity as reference points.
Advanced Troubleshooting:
This structured approach allows researchers to systematically identify and address factors affecting NatC complex activity, leading to more reliable and reproducible experimental results.
Emerging techniques for studying the in vivo role of naa35 in zebrafish development and disease models encompass several cutting-edge methodological approaches:
CRISPR-Cas9 Genome Editing:
Generate precise point mutations in naa35 to study structure-function relationships
Create conditional knockouts using inducible systems (e.g., CreERT2/loxP)
Implement base editing or prime editing for subtle modifications
Establish tissue-specific knockouts to distinguish local versus global effects
Live Imaging Techniques:
Use fluorescent protein fusions to track naa35 localization during development
Implement light-sheet microscopy for long-term, low-phototoxicity imaging
Apply FRET-based sensors to monitor protein-protein interactions in real-time
Utilize super-resolution microscopy to visualize subcellular localization
Single-Cell Approaches:
Perform single-cell RNA-seq to identify cell populations affected by naa35 dysfunction
Apply spatial transcriptomics to map expression patterns in tissue context
Implement CyTOF or CODEX for protein-level single-cell analysis
Use lineage tracing to follow cell fate decisions influenced by naa35
Chemical Biology Tools:
Develop small molecule inhibitors specific to the NatC complex
Implement photocrosslinking to capture transient interactions
Apply chemical genetics with engineered naa35 variants
Utilize targeted protein degradation (PROTACs, dTAGs) for rapid protein depletion
Disease Modeling Applications:
Engineer zebrafish models mimicking human disease mutations
Implement high-throughput behavioral assays to detect subtle phenotypes
Apply metabolomics and proteomics to characterize systemic effects
Utilize drug screening platforms for therapeutic development
These emerging techniques provide powerful tools for dissecting the complex roles of naa35 in zebrafish development and disease, potentially revealing novel insights into N-terminal acetylation biology with translational implications .
Structural studies of naa35 provide crucial insights that can directly inform the rational design of specific inhibitors or modulators of the NatC complex. This structure-guided drug discovery approach involves:
Target Site Identification:
Crystal structures reveal that naa35 forms a critical interface with Naa30 for substrate recognition
The substrate binding pocket at the Naa30-naa35 interface presents a druggable site
Sequence-specific, ligand-induced conformational changes in Naa30 enable efficient acetylation, suggesting potential allosteric sites
Structure-Based Drug Design Strategies:
Virtual screening against the substrate binding pocket or protein-protein interfaces
Fragment-based approaches to identify chemical scaffolds with binding potential
Structure-activity relationship (SAR) studies to optimize lead compounds
Molecular dynamics simulations to identify cryptic pockets or transient states
Targeting NatC-Specific Features:
The divergent architecture of NatC compared to other NAT complexes enables development of selective inhibitors
The ribosome-binding patch in the elongated tip region of NatC presents a unique target site
Species-specific structural differences can be exploited for selective targeting
Methodological Framework for Inhibitor Development:
| Phase | Methodology | Key Considerations |
|---|---|---|
| Initial screening | Structure-based virtual screening, biochemical assays | Target specific protein-protein interfaces or catalytic site |
| Hit validation | Biophysical binding assays (SPR, ITC, NMR) | Confirm direct binding to target site |
| Mechanism studies | Enzymatic assays, X-ray crystallography with bound inhibitors | Determine inhibition mechanism (competitive, non-competitive, etc.) |
| Selectivity profiling | Counter-screening against other NAT complexes | Ensure specificity for NatC over NatA, NatB, etc. |
| Cellular studies | Cell penetration, target engagement, phenotypic assays | Verify activity in cellular context |
| In vivo validation | Zebrafish models with naa35 mutations, rescue experiments | Confirm efficacy and selectivity in vivo |
Therapeutic Applications:
Alterations in N-terminal acetylation are implicated in several diseases, including cancers and developmental disorders
Selective NatC inhibitors could provide new therapeutic approaches for conditions where NatC activity is dysregulated
Zebrafish models provide an excellent system for initial in vivo validation of compound efficacy and toxicity
This structure-guided approach leverages the unique architectural features of the NatC complex to develop selective modulators with potential research and therapeutic applications .
Ensuring reproducibility in experiments using recombinant naa35 requires adherence to a comprehensive set of best practices that address multiple aspects of experimental design, execution, and reporting:
Protein Production and Quality Control:
Document complete expression and purification protocols, including vector constructs, expression systems, and purification steps
Implement consistent quality control measures (SDS-PAGE, mass spectrometry, activity assays)
Record and report protein concentration determination methods
Storage and Handling Protocols:
Store lyophilized protein at -20°C/-80°C (shelf life up to 12 months)
Store liquid formulations at -20°C/-80°C (shelf life up to 6 months)
Avoid repeated freeze-thaw cycles
Prepare working aliquots for short-term use (up to one week at 4°C)
Document buffer composition and additives (glycerol percentage, salt concentration)
Experimental Design Considerations:
Include positive and negative controls in all experiments
Perform technical and biological replicates (minimum triplicate measurements)
Blind analysis where applicable to reduce bias
Use statistical power calculations to determine appropriate sample sizes
Assay Standardization:
For kinetic studies, standardize substrate concentration ranges (consider potential substrate/product inhibition)
Maintain consistent reaction conditions (temperature, pH, buffer composition)
Calibrate instruments regularly and include calibration standards
Document detailed assay protocols including all reagents and their sources
Data Management and Reporting:
Maintain comprehensive laboratory records with raw data
Report all experimental conditions, including those that yielded negative results
Share complete datasets through appropriate repositories
Document all statistical analyses and data transformations
By implementing these best practices, researchers can significantly enhance the reproducibility of experiments using recombinant naa35, facilitating comparison of results across different studies and advancing our understanding of NatC complex function .
When interpreting results from studies using recombinant naa35, researchers should consider several key factors that may influence experimental outcomes and their biological relevance:
Protein Context Dependencies:
naa35 functions as part of the heterotrimeric NatC complex, and its activity depends on proper complex formation with Naa30 and Naa38
Results from studies using isolated naa35 may not reflect its native biological function
Consider whether experiments with the complete NatC complex would provide more physiologically relevant insights
Species-Specific Variations:
Technical Limitations and Artifacts:
Recombinant protein may lack post-translational modifications present in vivo
Expression tags (His, GST, etc.) may influence protein function or interactions
E. coli-expressed proteins lack eukaryotic-specific co-factors that might be important for function
The purity of the preparation (target >85%) can affect experimental outcomes
Physiological Relevance Assessment:
Consider whether experimental conditions (substrate concentrations, buffer composition, etc.) reflect the cellular environment
Evaluate whether observed in vitro effects would occur at physiological concentrations
Validate key findings in cellular or in vivo models when possible
Data Integration Framework:
Integrate results from multiple experimental approaches (biochemical, structural, cellular, in vivo)
Consider alternative explanations for unexpected results
Place findings in the context of the broader literature on N-terminal acetylation
Recognize the limitations of current models and methodologies
By carefully considering these factors, researchers can avoid misinterpretation of experimental results and develop more accurate models of naa35 function in the context of the NatC complex and N-terminal acetylation biology .
Researchers working with recombinant naa35 can access several essential resources and tools that facilitate experimental design, data analysis, and interpretation:
Protein Information Resources:
UniProt entry Q7T322 for Danio rerio naa35 - provides curated protein information, sequence data, and functional annotations
Protein Data Bank (PDB) - contains structural information for NAT complexes
The Zebrafish Information Network (ZFIN) - offers genetic and expression data for zebrafish naa35
Commercial Sources for Recombinant Protein:
Sequence Analysis Tools:
Expression and Purification Resources:
Functional Assay Methodologies:
Zebrafish Research Tools:
Established zebrafish lines for developmental studies
CRISPR-Cas9 protocols optimized for zebrafish
Phenotypic analysis pipelines for developmental studies
Data Analysis Software:
Kinetics software for enzyme activity analysis
Proteomics data analysis tools for N-terminal acetylation studies
Structural visualization software for analyzing protein-protein interfaces
These resources provide researchers with the necessary tools to conduct comprehensive studies on recombinant naa35 and its role in the NatC complex, facilitating advances in our understanding of N-terminal acetylation biology .
Effective interdisciplinary collaboration to advance understanding of naa35 function requires strategic approaches that bridge different research domains and methodologies. A comprehensive framework includes:
Establishing Cross-Disciplinary Teams:
Integrate structural biologists, biochemists, developmental biologists, and computational scientists
Create collaborative networks that span basic and translational research
Develop shared terminology and conceptual frameworks to facilitate communication
Complementary Methodological Approaches:
| Discipline | Contribution | Methodologies | Integration Points |
|---|---|---|---|
| Structural Biology | Elucidation of protein structure and interactions | X-ray crystallography, cryo-EM, NMR | Inform biochemical assay design and molecular modeling |
| Biochemistry | Characterization of enzymatic activity and specificity | Enzyme kinetics, substrate profiling | Connect structure to function, identify key residues |
| Cell Biology | Cellular context and regulation | Microscopy, protein interaction studies | Link biochemical findings to cellular functions |
| Developmental Biology | In vivo function in zebrafish | Genetic manipulations, phenotypic analysis | Provide physiological relevance to molecular findings |
| Computational Biology | Data integration and prediction | Molecular dynamics, systems biology | Generate testable hypotheses, integrate diverse datasets |
Data Sharing and Integration Strategies:
Implement FAIR principles (Findable, Accessible, Interoperable, Reusable) for data management
Utilize common data repositories and standardized formats
Develop integrated analytical pipelines that combine diverse data types
Create visualization tools that communicate complex findings across disciplines
Collaborative Research Models:
Establish regular interdisciplinary meetings or workshops focused on N-terminal acetylation
Create shared resource centers for specialized techniques or equipment
Develop joint funding proposals that explicitly value interdisciplinary approaches
Implement team science training to enhance collaborative effectiveness
Translational Research Connections:
Partner with clinical researchers to explore disease relevance
Connect basic findings to potential therapeutic applications
Collaborate with pharmaceutical researchers on drug development
Engage with patient advocacy groups when disease connections are established