KEGG: ago:AGOS_AER248W
STRING: 33169.AAS52929
Ashbya gossypii is a filamentous fungus that has gained prominence as a model organism in biotechnology, primarily due to its ability to produce riboflavin (vitamin B2) at industrial scales. It has long been considered a paradigm of White Biotechnology for riboflavin production . Its industrial relevance has led to the development of significant molecular toolkits and in silico modeling capabilities that facilitate genetic manipulation and metabolic engineering . While initially valued for riboflavin production, A. gossypii has emerged as a versatile host for producing recombinant proteins, including enzymes like UBA4, due to several advantageous characteristics:
A fully sequenced and well-annotated genome
Genetic similarity to Saccharomyces cerevisiae, allowing application of yeast genetic tools
Efficient secretion machinery for protein expression
Established protocols for transformation and gene integration
Ability to grow on simple, cost-effective media
The genomic knowledge and molecular tools available for A. gossypii make it particularly suitable for expression of proteins like UBA4 with experimental manipulations possible through techniques such as homologous recombination in S. cerevisiae .
UBA4 in A. gossypii (UBA4_ASHGO) is an enzyme with dual catalytic functions of adenylyltransferase and sulfurtransferase activities . Based on ortholog analysis, UBA4 appears to be involved in essential cellular functions that are conserved across diverse species from fungi to humans . Specifically:
Adenylyltransferase activity: Catalyzes the transfer of an adenylyl group from ATP to target substrates, an essential step in various metabolic pathways.
Sulfurtransferase activity: Mediates the transfer of sulfur to various acceptor molecules, playing a crucial role in pathways such as tRNA thiolation and molybdenum cofactor biosynthesis.
The dual functionality of UBA4 positions it as a key player in multiple biochemical pathways that contribute to cellular homeostasis and survival in A. gossypii. Based on comparative genomics data, the protein appears to be highly conserved, suggesting its fundamental importance in fungal metabolism .
Ortholog analysis reveals that A. gossypii UBA4 is part of at least 522 full-length protein ortholog groups, indicating significant conservation across diverse species . This extensive conservation suggests that UBA4 serves essential biological functions. Notable orthologous relationships include:
| Ortholog Group | Species | Protein | Bitscore | Inparalog Score | Seed Score |
|---|---|---|---|---|---|
| 525 | Loa loa | A0A1S0UJ30 (MOCS3 homolog) | 240 | 1.0 | 0.996 |
| 92 | Micromonospora echinospora | A0A1C4X5N7 | 245 | 1.0 | 1.0 |
| 598 | Actinia tenebrosa | A0A6P8I810 (MOCS3 homolog) | 275 | 1.0 | 1.0 |
| 999 | Kazachstania africana | H2AVZ7 (KAFR0E03960) | 593 | 1.0 | 1.0 |
The high bitscore (593) between A. gossypii UBA4 and the Kazachstania africana ortholog indicates particularly strong sequence similarity between these fungal species . The conservation extends beyond fungi to diverse organisms including nematodes (Loa loa), bacteria (Micromonospora echinospora), and cnidarians (Actinia tenebrosa), highlighting the evolutionary importance of this enzyme .
For recombinant production of A. gossypii UBA4, researchers should consider several expression systems, each with distinct advantages depending on research objectives:
Homologous Expression in A. gossypii:
Benefits: Native post-translational modifications, proper folding, and physiological relevance
Methodology: Integration of expression cassettes can be achieved through homologous recombination techniques similar to those used for other A. gossypii genes, as demonstrated in septin studies
Considerations: Growth conditions should be optimized based on established protocols for A. gossypii cultivation, potentially using media compositions similar to those used for riboflavin production experiments (RPM media)
Heterologous Expression in S. cerevisiae:
Benefits: Genetic similarity to A. gossypii, well-established tools, suitable for protein interaction studies
Methodology: Yeast homologous recombination can be employed to generate expression constructs, as shown for other A. gossypii proteins
Considerations: May require codon optimization based on S. cerevisiae preferences
E. coli Expression System:
Benefits: High yields, simple cultivation, rapid results
Methodology: Standard protocols for recombinant protein expression in E. coli with appropriate affinity tags
Considerations: May lack post-translational modifications; refolding might be necessary if inclusion bodies form
Comparative Expression Performance:
| Expression System | Yield Potential | Post-translational Modifications | Purification Complexity | Time to Results |
|---|---|---|---|---|
| A. gossypii | Moderate | Native | Moderate | 5-7 days |
| S. cerevisiae | Moderate-High | Similar to native | Moderate | 3-5 days |
| E. coli | High | Limited | Low-Moderate | 1-3 days |
The selection of an appropriate expression system should be guided by specific research requirements, particularly concerning protein authenticity, yield, and downstream applications.
To investigate UBA4's role in A. gossypii sulfur metabolism, researchers should employ a multifaceted approach:
1. Gene Deletion/Knockout Studies:
Generate UBA4 deletion strains using homologous recombination-based gene targeting, similar to methods used for CDC11 studies in A. gossypii
Assess phenotypic consequences on growth, morphology, and metabolite profiles
Compare with wild-type strains to identify metabolic pathways affected by UBA4 absence
2. Protein-Protein Interaction Analysis:
Employ tandem affinity purification (TAP) tagging of UBA4
Use mass spectrometry to identify interaction partners
Validate key interactions through co-immunoprecipitation or yeast two-hybrid assays
3. Metabolomic Profiling:
Compare metabolite profiles between wild-type and UBA4-modified strains
Focus particularly on sulfur-containing metabolites and molybdenum cofactor synthesis intermediates
Use liquid chromatography-mass spectrometry (LC-MS) to detect changes in metabolic pathways
4. Enzymatic Activity Assays:
Develop in vitro assays to measure both adenylyltransferase and sulfurtransferase activities
Determine kinetic parameters (Km, Vmax) for different substrates
Assess how mutations in key residues affect catalytic efficiency
5. Transcriptome Analysis:
Perform RNA sequencing to identify genes differentially expressed in UBA4 mutants
Look for compensatory changes in expression of other sulfur metabolism genes
Compare transcript patterns across different growth phases, similar to studies of CDC11a/b expression patterns in A. gossypii
Each of these approaches provides complementary insights into UBA4 function, collectively building a comprehensive understanding of its role in A. gossypii metabolism.
To design an efficient gene targeting strategy for UBA4 modification in A. gossypii, follow this methodological approach:
Construct targeting vectors in S. cerevisiae using homologous recombination, similar to techniques used for CDC11 modification
Design primers with 40-50 bp homology arms complementary to sequences flanking your target integration site in the UBA4 locus
Amplify selection markers (such as G418 or NAT resistance cassettes) with primers containing these homology arms
Transform S. cerevisiae with the PCR products and a linearized vector backbone
Extract and verify the resulting plasmids by restriction analysis and sequencing
Prepare A. gossypii spores or mycelial fragments for transformation
Use electroporation or PEG/LiAc-mediated transformation protocols
Select transformants on appropriate media containing antibiotics (G418 at 200 μg/ml or clonNAT at 50 μg/ml)
Verify integration by PCR and/or Southern blotting
Confirm proper integration by PCR with primers binding outside the integration site
Verify protein expression/modification by Western blotting or fluorescence microscopy (for tagged versions)
Assess strain stability by repeated cultivation and verification of the genetic modification
Specific Modifications for Different Research Goals:
| Modification Type | Selection Marker | Purpose | Additional Components |
|---|---|---|---|
| Gene deletion | G418 or NAT | Loss-of-function studies | None |
| C-terminal tagging | G418 | Localization/purification | GFP or affinity tag |
| Promoter replacement | NAT | Expression modulation | Constitutive or inducible promoter |
| Point mutations | G418 | Structure-function studies | Silent marker mutations |
This strategy builds on established A. gossypii genetic manipulation techniques, as demonstrated in the successful modification of CDC11 and other genes .
Purification of recombinant A. gossypii UBA4 requires careful consideration of protein stability and activity. The following protocol is recommended:
Express UBA4 with an N-terminal or C-terminal affinity tag (His6, GST, or MBP)
For E. coli expression, use BL21(DE3) or Rosetta strains to address potential codon bias
Consider co-expression with chaperones if folding issues are encountered
Optimize expression conditions: temperature (16-30°C), IPTG concentration (0.1-1.0 mM), and induction time (4-24 hours)
Resuspend cells in buffer containing:
50 mM Tris-HCl or HEPES, pH 7.5-8.0
150-300 mM NaCl
10% glycerol (stabilizer)
1 mM DTT or 2 mM β-mercaptoethanol (to maintain reduced cysteines)
1 mM PMSF and protease inhibitor cocktail
Lyse cells by sonication or high-pressure homogenization
Clarify lysate by centrifugation at 20,000 × g for 30 minutes at 4°C
Load clarified lysate onto appropriate affinity resin
For His-tagged UBA4: Ni-NTA or TALON resin
Wash extensively to remove non-specifically bound proteins
Elute with imidazole gradient (for His-tag) or reduced glutathione (for GST-tag)
Perform ion exchange chromatography based on predicted pI of UBA4
Follow with size exclusion chromatography for highest purity
Monitor protein purity by SDS-PAGE after each step
Add stabilizing agents to final buffer:
10-20% glycerol
1-5 mM DTT
0.1-0.5 mM EDTA (if metal ions are not required for activity)
Determine optimal storage conditions (temperature, buffer composition)
Test enzyme activity after each purification step to identify potential activity loss
Enzymatic Activity Monitoring:
Develop adenylyltransferase activity assay measuring ATP consumption or AMP formation
Establish sulfurtransferase activity assay using appropriate sulfur acceptor substrates
Document specific activity throughout purification process to calculate recovery and purification fold
This protocol integrates approaches used for other recombinant enzymes expressed in A. gossypii and related organisms, with specific adaptations for preserving the dual catalytic activities of UBA4.
Analyzing orthologous relationships for A. gossypii UBA4 requires a systematic bioinformatic approach:
Obtain the amino acid sequence of A. gossypii UBA4 from databases like UniProt (Q756K6)
Identify conserved domains using tools such as NCBI CDD, Pfam, or InterPro
Generate a domain architecture diagram highlighting functional motifs
Use ortholog databases like InParanoid, OrthoMCL, or OrthoDB
As shown in the search results, A. gossypii UBA4 is part of 522 full-length protein ortholog groups
Perform reciprocal BLAST searches to confirm orthologous relationships
Focus on key model organisms and closely related fungi for detailed comparison
Align UBA4 sequences using MUSCLE, MAFFT, or T-Coffee
Refine alignments manually if necessary, focusing on catalytic domains
Identify conserved residues across different taxonomic groups
Pay special attention to adenylyltransferase and sulfurtransferase active sites
Construct phylogenetic trees using Maximum Likelihood or Bayesian methods
Evaluate branch support using bootstrap or posterior probability values
Analyze evolutionary rates across different lineages
Identify potential gene duplication or loss events
Researchers working with recombinant A. gossypii UBA4 often encounter several challenges. Here are common issues and methodological solutions:
Problem: Insufficient protein yields for downstream applications.
Solutions:
Optimize codon usage for the expression host (particularly important for E. coli)
Test different promoters with varying strength (constitutive vs. inducible)
Explore alternative expression hosts (A. gossypii itself, P. pastoris, or mammalian cells)
Adjust induction conditions (temperature, inducer concentration, timing)
For A. gossypii expression, consider the temporal regulation pattern observed in other genes like CDC11a/b
Problem: Recombinant UBA4 forms insoluble aggregates, particularly in bacterial systems.
Solutions:
Lower expression temperature (16-20°C) to slow folding
Co-express with molecular chaperones (GroEL/ES, DnaK/J)
Use solubility-enhancing fusion partners (MBP, SUMO, or TrxA)
Develop refolding protocols if inclusion bodies persist:
Solubilize in 6-8 M urea or 6 M guanidine-HCl
Remove denaturant by dialysis or rapid dilution
Include redox pairs (GSH/GSSG) to assist disulfide formation if needed
Perform multiple sequence alignments of UBA4 orthologs across different species
Identify strictly conserved residues, particularly within predicted catalytic domains
Use homology modeling based on crystallized orthologous proteins to predict structure
Identify potential catalytic residues based on:
Conservation across species
Proximity to predicted active sites
Known catalytic motifs for adenylyltransferases and sulfurtransferases
Design primers for site-directed mutagenesis of predicted catalytic residues
Create a panel of mutations including:
Conservative substitutions (maintaining chemical properties)
Non-conservative substitutions (altering chemical properties)
Alanine scanning of potentially important regions
Generate mutant constructs using PCR-based methods similar to those used for other A. gossypii genes
Express and purify each mutant protein using identical conditions
Develop quantitative assays for both enzymatic activities:
Adenylyltransferase activity: measure ATP consumption or AMP formation
Sulfurtransferase activity: quantify sulfur transfer to acceptor substrates
Compare kinetic parameters (kcat, Km) between wild-type and mutant proteins
Document effects on protein stability and folding through thermal shift assays
Create a comprehensive data table correlating mutations with activity changes:
| Residue | Mutation | Adenylyltransferase Activity (% of WT) | Sulfurtransferase Activity (% of WT) | Structural Effect |
|---|---|---|---|---|
| X123 | X123A | 5 | 95 | Disrupts ATP binding |
| Y234 | Y234F | 90 | 85 | Minor effect on substrate positioning |
| C345 | C345S | 85 | <1 | Critical for sulfur transfer |
Map activity-altering mutations onto the structural model
Distinguish between residues affecting substrate binding versus catalysis
Identify residues specifically involved in each catalytic function
This systematic approach allows for precise identification of functionally critical residues and provides insights into the mechanistic basis of UBA4's dual catalytic activities.
To investigate UBA4's role in A. gossypii stress response pathways, implement this comprehensive experimental strategy:
Create UBA4 deletion mutants using homologous recombination techniques
Develop conditional expression strains (e.g., using regulatable promoters)
Generate point mutants that specifically affect adenylyltransferase or sulfurtransferase activities
Include appropriate control strains with similar genetic backgrounds
Expose wild-type and UBA4-modified strains to various stressors:
Oxidative stress (H₂O₂, paraquat)
Heavy metal stress (cadmium, arsenic)
Nutritional stress (sulfur limitation, nitrogen starvation)
Temperature stress (heat shock, cold shock)
Quantify growth parameters (lag phase, doubling time, final biomass)
Document morphological changes through microscopy
Measure survival rates under acute stress conditions
Perform RNA-seq on wild-type and UBA4-modified strains under normal and stress conditions
Identify differentially expressed genes and enriched pathways
Validate key findings with RT-qPCR
Compare expression patterns to those observed in studies of other A. gossypii genes like CDC11a/b that show developmental regulation
Use mass spectrometry to identify proteins with altered abundance or modification
Focus particularly on changes in thiolation status of target proteins
Analyze profiles of oxidatively damaged proteins
Quantify levels of stress-responsive metabolites
Construct a network model integrating transcriptomic and proteomic data
Identify key regulatory nodes linking UBA4 to stress response pathways
Validate predicted connections through targeted experiments
Compare findings with known stress response pathways in related fungi
This methodological framework provides a comprehensive approach to understanding UBA4's contribution to stress response mechanisms in A. gossypii, potentially revealing novel regulatory connections and pathway interactions.