Candida glabrata is a fungal species known for its increasing prevalence in mucosal and systemic infections, especially in immunocompromised individuals . C. glabrata exhibits innate resistance to azole antifungal drugs and can rapidly develop clinical drug resistance, making the study of its cellular mechanisms crucial . Recombinant Candida glabrata Enolase-phosphatase E1, also known as UTR4, is a bifunctional enzyme that has roles in metabolic pathways .
Recombinant Candida glabrata Enolase-phosphatase E1 (UTR4) is a protein expressed in E. coli and derived from the Candida glabrata species . It is also referred to as 2,3-diketo-5-methylthio-1-phosphopentane phosphatase and has the EC number 3.1.3.77 . The protein sequence includes 251 amino acids, and it is recommended to reconstitute the protein in deionized sterile water to a concentration of 0.1-1.0 mg/mL, with the addition of 5-50% glycerol for long-term storage .
| Property | Description |
|---|---|
| Product Code | CSB-EP738906CZI |
| Abbreviation | UTR4 |
| UniProt No. | Q6FLR5 |
| Immunogen Species | Candida glabrata (strain ATCC 2001 / CBS 138 / JCM 3761 / NBRC 0622 / NRRL Y-65) |
| Purity | >85% (SDS-PAGE) |
| Source | E. coli |
| Target Names | UTR4 |
| Protein Names | Enolase-phosphatase E1 |
| Expression Region | 1-251 |
| Storage | Liquid form: 6 months at -20°C/-80°C; Lyophilized form: 12 months at -20°C/-80°C |
| Reconstitution | Deionized sterile water to a concentration of 0.1-1.0 mg/mL, with 5-50% glycerol for long-term storage at -20°C/-80°C |
UTR4 is a bifunctional enzyme that catalyzes the enolization of 2,3-diketo-5-methylthiopentyl-1-phosphate (DK-MTP-1-P) into an intermediate in metabolic processes . Enolases, in general, are involved in the glycolysis pathway, and significant amounts are found on the cell surface of Candida species, where they contribute to adhesion to human tissues and activation of fibrinolysis .
Enolase is a cytosolic enzyme that is exposed on the cell surface of Candida yeast . This exposed form of enolase is hypothesized to contribute to infection-related phenomena, such as fungal adhesion to human tissues, activation of fibrinolysis, and extracellular matrix degradation .
Binding Affinity: Candida albicans and Candida tropicalis enolases bind tightly to human vitronectin, fibronectin, and plasminogen, with dissociation constants in the 10-7–10-8 M range . Saccharomyces cerevisiae enolase shows a much lower binding affinity for these human proteins .
Inhibition of Human Protein Binding: Soluble fungal enolases and anti-enolase antibodies can inhibit the adsorption of vitronectin (VTR), fibronectin (FN), and plasminogen (HPG) to C. albicans and C. tropicalis hyphae/pseudohyphae, reducing human protein binding to fungal cells by 20–40% .
Catalytic Activity: Candidal enolases retain their enzymatic activity even after forming complexes with VTR, FN, or HPG, suggesting that the interaction sites with human proteins are distant from the enolase active center .
Candida glabrata has innate resistance to azole antifungal drugs, which target Erg11, a key enzyme in ergosterol biosynthesis . This species can also rapidly develop clinical drug resistance, making the study of its resistance mechanisms critical . The mechanisms controlling azole-induced ERG gene expression and drug resistance in C. glabrata primarily involve Upc2 and/or Pdr1 .
KEGG: cgr:CAGL0L01287g
STRING: 284593.XP_448829.1
Enolase-phosphatase E1 (UTR4) in Candida glabrata is encoded by a specific open reading frame in the C. glabrata genome. Similar to other C. glabrata genes, expression of UTR4 can be analyzed using quantitative real-time PCR (RT-PCR) with specific primers designed against the gene sequence. When designing primers, researchers should ensure specificity by comparing with other sequences in the C. glabrata genome to avoid cross-reactivity. For normalization purposes, housekeeping genes like CgACT1 (actin) are commonly used as internal reference standards .
For recombinant expression of C. glabrata proteins, several expression systems can be employed based on research needs. Homologous expression within modified C. glabrata strains offers advantages for proper protein folding and post-translational modifications. This can be achieved using inducible promoters such as the copper-inducible MTI promoter, which has been successfully used for controlled expression of C. glabrata genes . Alternatively, heterologous expression in Saccharomyces cerevisiae often provides good yields while maintaining proper eukaryotic processing. For higher protein yields, bacterial systems like E. coli can be used, though they may lack proper post-translational modifications that could be essential for UTR4 activity.
The expression of many C. glabrata genes is strongly influenced by environmental conditions encountered during host infection. Similar to CgDTR1, which shows differential expression under stress conditions, UTR4 expression likely responds to specific environmental cues. Researchers should examine UTR4 expression under various conditions including different carbon sources, pH variations, oxidative stress (e.g., hydrogen peroxide exposure), and during internalization within host cells. RT-PCR can be used to quantify these expression changes by extracting RNA from C. glabrata cells grown under these various conditions or when internalized within host cells .
CRISPR-Cas9 technology has revolutionized genome engineering in C. glabrata. To create UTR4 knockout strains, researchers should consider the following approach: First, design guide RNAs targeting the UTR4 gene using specialized algorithms to maximize efficiency and minimize off-target effects. Transform a recombinant C. glabrata strain constitutively expressing the CRISPR-Cas9 system with these guide RNAs . The efficiency of homologous recombination can be significantly increased (up to 8-fold higher) when using 200 bp of homology domains compared to shorter 20 bp domains . After transformation, mutants can be identified using the Surveyor technique followed by sequencing confirmation. This approach provides a robust method for generating clean deletions without marker genes remaining in the genome.
For site-directed mutagenesis of UTR4, first identify conserved residues through multiple sequence alignment with homologous proteins from related species. Design mutagenic primers containing the desired nucleotide changes with approximately 15-20 nucleotides of perfect matching sequence on either side of the mutation. Use a high-fidelity polymerase for PCR amplification to introduce mutations. After DpnI digestion to remove template DNA, transform the amplified product into E. coli. Screen colonies by sequencing and then introduce the validated mutant constructs into expression vectors with appropriate promoters, such as the copper-inducible MTI promoter system, which allows for controlled expression in C. glabrata .
For complementation studies, plasmid vectors that enable controlled expression are essential. The pGREG576 plasmid, modified to contain the copper-inducible MTI promoter rather than the GAL1 promoter, has proven effective for controlled gene expression in C. glabrata . This system allows for the verification of gene function through complementation of knockout phenotypes. When designing the complementation construct, include the full UTR4 coding sequence verified by sequencing. For more precise expression control, consider using the native UTR4 promoter region in place of the inducible promoter to maintain physiological expression patterns in response to environmental signals.
To assess the impact of UTR4 on virulence, researchers should compare wild-type C. glabrata strains with UTR4 deletion mutants in appropriate infection models. The Galleria mellonella larval model provides a useful system for initial virulence assessments, as demonstrated for other C. glabrata genes . When conducting such experiments, standardize the inoculum to approximately 5 × 10^7 CFU/larvae and monitor survival over 72 hours using Kaplan-Meier survival curves to document differences in killing ability. Complementary studies should evaluate the ability of UTR4 mutants to proliferate within the host by recovering hemolymph at different time points (e.g., 1, 24, and 48 hours post-infection) and quantifying viable C. glabrata cells .
To investigate UTR4's role in stress resistance, compare growth of wild-type and UTR4 deletion strains under various stress conditions relevant to host environments. These should include oxidative stress (hydrogen peroxide), acidic stress (varied pH), antifungal compounds, and nutritional limitation. Perform both spot dilution assays on solid media and growth curve analyses in liquid culture to quantify differences in stress tolerance. Additionally, examine gene expression changes of UTR4 under these stress conditions using RT-PCR with primers specific for UTR4 and normalizing with housekeeping genes like CgACT1 . This approach will help determine whether UTR4 is part of the stress response machinery that contributes to C. glabrata survival within host environments.
To assess UTR4's role in phagocyte survival, co-culture wild-type and UTR4 mutant C. glabrata strains with either isolated hemocytes from G. mellonella or mammalian macrophages . Measure the concentration of viable C. glabrata cells within phagocytes after various time points (1, 4, 24, and 48 hours) to determine if UTR4 affects intracellular survival and proliferation. Complement these studies with microscopy techniques to visualize phagosome maturation and potential escape mechanisms. Additionally, measure the expression levels of UTR4 during internalization within phagocytic cells using RT-PCR to determine if the gene is up- or down-regulated during this critical phase of host-pathogen interaction .
For mapping UTR4 interaction networks, several complementary approaches should be employed. Create a tagged version of UTR4 (e.g., with HA, FLAG, or His tags) and express it under its native promoter to maintain physiological expression levels. Perform co-immunoprecipitation followed by mass spectrometry to identify interacting proteins. Validate key interactions using techniques such as yeast two-hybrid or bimolecular fluorescence complementation. Analyze the resulting interaction network to identify biological processes where UTR4 participates. For more dynamic assessment, compare interaction networks under different environmental conditions that mimic the host environment, such as oxidative stress or acidic pH, which have been shown to affect the function of other C. glabrata virulence factors .
To comprehensively characterize UTR4 function, implement a multi-tiered phenotypic analysis approach. Begin with growth assays under various conditions (carbon sources, pH, temperature, stress agents) comparing wild-type and UTR4 mutant strains. Examine biofilm formation capacity using crystal violet staining and confocal microscopy. Assess adhesion to epithelial cells and abiotic surfaces. Evaluate metabolic profiles using techniques like Biolog phenotype microarrays to identify specific metabolic pathways affected by UTR4 deletion. For virulence assessment, use both the G. mellonella infection model and cell culture systems with phagocytes to measure survival rates as performed for other virulence factors . Create a comprehensive phenotypic profile table comparing wild-type, deletion mutant, and complemented strains under all tested conditions.
To gain structural insights into UTR4 function, purify recombinant UTR4 to homogeneity using affinity chromatography followed by size exclusion chromatography. Assess protein folding and stability using circular dichroism spectroscopy. For crystallization trials, screen multiple conditions varying pH, precipitants, and additives. If crystallization proves challenging, consider nuclear magnetic resonance (NMR) for solution structure determination or cryo-electron microscopy for larger complexes. In parallel, perform computational modeling using homology-based approaches with known structures of related enolase-phosphatases. Identify conserved catalytic residues through sequence alignment and verify their importance through site-directed mutagenesis followed by enzymatic activity assays. Structural information can guide the design of specific inhibitors that might serve as potential antifungal agents.
To compare UTR4 across Candida species, perform comprehensive sequence analyses using multiple sequence alignment tools to identify conserved domains and species-specific variations. Calculate evolutionary distances and construct phylogenetic trees to understand the evolutionary relationship of UTR4 among different species. Complement sequence analysis with functional comparisons by expressing orthologues from different species in a C. glabrata UTR4 deletion background to assess functional complementation. Pay particular attention to differences between pathogenic and non-pathogenic species to identify adaptations that might contribute to virulence. Present the sequence conservation data in a table format showing percent identity and similarity across key Candida species, along with highlighting functionally important residues that are either conserved or divergent.
| Species | Sequence Identity (%) | Sequence Similarity (%) | Key Functional Residues Conserved |
|---|---|---|---|
| C. glabrata | 100 | 100 | All |
| C. albicans | ~65-75* | ~80-85* | Catalytic residues conserved |
| C. parapsilosis | ~60-70* | ~75-80* | Most catalytic residues conserved |
| C. tropicalis | ~65-75* | ~75-85* | All catalytic residues conserved |
| S. cerevisiae | ~80-90* | ~85-95* | All functional domains conserved |
*Note: These are estimated ranges based on typical conservation patterns between these species; exact values should be determined experimentally.
The CRISPR-Cas9 system has transformed genetic manipulation in C. glabrata by significantly increasing the efficiency of gene editing. Prior to CRISPR, gene deletion in C. glabrata required long homology arms and had low efficiency. Now, researchers can achieve efficient gene disruption with just 20-200 bp of homology domains, with homologous recombination efficiency increasing up to 8-fold when using 200 bp homology domains compared to 20 bp . For UTR4 studies, CRISPR-Cas9 allows precise gene deletion, site-directed mutagenesis of specific domains, and seamless gene tagging for localization and interaction studies. Additionally, the system enables multiplexed gene editing to study genetic interactions between UTR4 and other genes. When designing CRISPR experiments, researchers should use optimized guide RNA selection tools specific for C. glabrata to maximize editing efficiency and minimize off-target effects .
Recombinant UTR4 purification faces several challenges that require methodical approaches to resolve. Solubility issues can be addressed by optimizing expression conditions (temperature, induction timing, media composition) or by using solubility-enhancing tags such as MBP or SUMO. If protein aggregation persists, consider refolding from inclusion bodies using a gradual dialysis approach. Proteolytic degradation can be minimized by including protease inhibitors throughout the purification process and working at reduced temperatures (4°C). For optimal yield, design a multi-step purification strategy starting with affinity chromatography (His-tag or GST-tag based), followed by ion-exchange chromatography and size-exclusion chromatography. Verify protein purity using SDS-PAGE and confirm identity with Western blotting and mass spectrometry. Assess protein activity using enzyme kinetics assays specific to enolase-phosphatase function, comparing parameters such as Km and Vmax with those of related enzymes.
When facing inconsistent phenotypes between different UTR4 mutant strains, a systematic troubleshooting approach is essential. First, confirm the genetic modifications by re-sequencing the UTR4 locus in all strains to ensure the expected alterations are present. Check for potential second-site mutations by whole-genome sequencing or backcrossing when possible. Verify that the expression levels of neighboring genes remain unaffected by the UTR4 deletion using RT-PCR. Standardize experimental conditions meticulously, as minor variations in media composition, pH, temperature, or cell density can significantly impact phenotypic outcomes. Create isogenic strains where the only difference is the UTR4 modification by performing genetic manipulations in the same parental background. For virulence studies, ensure consistent inoculum preparation and standardize infection protocols, as variation in these parameters can strongly influence virulence outcomes .