Recombinant Lodderomyces elongisporus Cytochrome oxidase assembly protein 3, mitochondrial (COA3)

Shipped with Ice Packs
In Stock

Description

Functional Role in Cytochrome Oxidase Assembly

COA3 is essential for the negative feedback regulation of mitochondrial COX1 translation and cytochrome oxidase assembly. Key findings from functional studies include:

Mechanism of Action

  • Complex Formation: COA3 interacts with Cox14, Shy1, and Mss51 to form COA complexes (250–400 kD), which sequester newly synthesized Cox1 .

  • Mss51 Regulation: COA3 and Cox14 recruit Mss51 to Cox1, enabling its transition to a translationally inactive state. This prevents excessive Cox1 accumulation .

  • Membrane Localization: COA3 is an integral mitochondrial membrane protein with its C terminus exposed to the intermembrane space .

Consequences of Deficiency

In Saccharomyces cerevisiae models, coa3Δ mutants exhibit:

  • Uncontrolled COX1 expression.

  • Rapid degradation of unassembled Cox1.

  • Defective cytochrome oxidase activity .

Pathogenicity and Clinical Relevance

Lodderomyces elongisporus is an emerging pathogen linked to fungemia, particularly in immunocompromised neonates and adults . COA3’s role in mitochondrial function may influence the pathogen’s survival in clinical settings:

FeatureDetails
Triazole ResistanceClinical strains show low fluconazole MICs (0.125–0.5 mg/L), but apple-derived strains exhibit higher MICs (2–4 mg/L) due to mutations in ERG11 and other triazole-related genes .
Genomic DiversityHospital-acquired strains display loss of heterozygosity (LOH) in scaffolds 1 and 6, linked to environmental adaptation .
Ascospore FormationClinical isolates produce abundant ascospores, enhancing environmental persistence .

Experimental Models

  • ELISA Assays: Recombinant COA3 is used as a diagnostic antigen to study immune responses or protein-protein interactions .

  • Genomic Studies: Whole-genome sequencing of L. elongisporus strains reveals COA3’s genomic context and polymorphisms associated with drug resistance .

Comparative Analysis with Model Organisms

While L. elongisporus COA3 shares functional homology with Saccharomyces cerevisiae Coa3, key differences include:

AspectL. elongisporus COA3S. cerevisiae Coa3
Genomic ContextPart of a larger genome (15–16 Mb)Smaller genome (12–13 Mb)
CUG Codon TranslationSerine (shared with Candida parapsilosis)Leucine (standard in most fungi)
Triazole ResistanceVariable MICs depending on environmental originLess studied in clinical contexts

Future Research Directions

  1. Structural Studies: High-resolution crystallography to elucidate COA3’s interaction with Cox14 and Mss51.

  2. Therapeutic Targeting: Investigating COA3 as a candidate for disrupting mitochondrial biogenesis in L. elongisporus.

  3. Environmental Adaptation: Exploring how COA3-mediated mitochondrial function contributes to the pathogen’s persistence in hospital settings .

Product Specs

Form
Lyophilized powder
Note: We prioritize shipping the format currently in stock. If you have a specific format requirement, please indicate it in your order remarks. We will accommodate your request whenever possible.
Lead Time
Delivery time may vary depending on the purchase method and location. Please consult your local distributors for specific delivery time estimates.
Note: All our proteins are shipped with standard blue ice packs. If you require dry ice shipping, please inform us in advance. Additional fees will apply.
Notes
Repeated freezing and thawing is not recommended. Store working aliquots at 4°C for up to one week.
Reconstitution
We recommend briefly centrifuging the vial prior to opening to ensure all contents are at the bottom. Reconstitute the protein in deionized sterile water to a concentration of 0.1-1.0 mg/mL. We recommend adding 5-50% glycerol (final concentration) and aliquoting for long-term storage at -20°C/-80°C. Our default final glycerol concentration is 50%. Customers may use this as a reference.
Shelf Life
Shelf life is influenced by various factors including storage conditions, buffer components, temperature, and the protein's inherent stability.
Generally, the shelf life of the liquid form is 6 months at -20°C/-80°C. The shelf life of the lyophilized form is 12 months at -20°C/-80°C.
Storage Condition
Store at -20°C/-80°C upon receipt. Aliquoting is necessary for multiple uses. Avoid repeated freeze-thaw cycles.
Tag Info
Tag type will be determined during the manufacturing process.
The tag type will be determined during production. If you have a specific tag type preference, please inform us, and we will prioritize developing the specified tag.
Synonyms
COA3; LELG_05579; Cytochrome c oxidase assembly factor 3, mitochondrial
Buffer Before Lyophilization
Tris/PBS-based buffer, 6% Trehalose.
Datasheet
Please contact us to get it.
Expression Region
1-68
Protein Length
full length protein
Species
Lodderomyces elongisporus (strain ATCC 11503 / CBS 2605 / JCM 1781 / NBRC 1676 / NRRL YB-4239) (Yeast) (Saccharomyces elongisporus)
Target Names
COA3
Target Protein Sequence
MTPALYRVRQPFFWRNTISLFVIGSIPLAAYWYTFTKMTEDEFSDIPIPPISDEELTKLK KEYEAGKQ
Uniprot No.

Target Background

Function
Required for assembly of cytochrome c oxidase (complex IV).
Database Links
Protein Families
COA3 family
Subcellular Location
Mitochondrion inner membrane; Single-pass membrane protein.

Q&A

What is Lodderomyces elongisporus and why has it become important in research?

Lodderomyces elongisporus is a diploid ascomycete yeast that has gained significant attention as an emerging human fungal pathogen. Originally discovered in 1952 from Californian citrus concentrate as Saccharomyces elongisporus, this organism has since been isolated from diverse sources including soil, fermented food products, plants, stored apples, pigeon excreta, insects, marine fish, hospital environments, and humans . Its medical relevance was first established in 2008 when retrospective analysis of 542 clinical Candida parapsilosis isolates from 25 countries revealed that ten isolates were actually L. elongisporus . Currently, infections caused by this fungus have been documented in 14 countries across 5 continents, indicating its global distribution and increasing clinical significance .

This organism presents particular research interest due to several factors: its close phylogenetic relationship to clinically important Candida species, its ability to survive in hospital environments, its unique reproductive characteristics, and its susceptibility profile to antifungal agents that differs from related pathogens like C. parapsilosis . Understanding L. elongisporus at the molecular level, including its mitochondrial proteins like COA3, provides insights into fungal evolution, pathogenesis, and potential novel therapeutic targets.

How is L. elongisporus typically identified in laboratory settings, and how might this impact COA3 studies?

L. elongisporus presents significant identification challenges in laboratory settings, which may impact downstream studies of specific proteins like COA3. Traditional identification methods using BBL Chromagar Candida medium often misidentify L. elongisporus isolates as Candida albicans . This misidentification occurs because of phenotypic similarities between these two yeast species.

For accurate identification, molecular techniques are essential. Internal transcribed spacer (ITS) region sequencing of ribosomal DNA is particularly reliable, demonstrating 100% homology with L. elongisporus reference sequences compared to only 97% homology with C. parapsilosis . MALDI-TOF mass spectrometry can also provide accurate identification when properly validated reference spectra are available .

The implications for COA3 studies are significant:

  • Researchers must verify the taxonomic identity of their strains before conducting protein-specific studies

  • Comparative analyses of COA3 function across species must account for potential misidentification in historical literature

  • Functional studies should consider the evolutionary relationship between L. elongisporus and related Candida species when interpreting results

  • Protein isolation protocols may need to be optimized specifically for L. elongisporus given its distinct cell wall and membrane characteristics

Proper identification is crucial not only for accurate research on specific proteins like COA3 but also for appropriate antifungal therapy selection, as L. elongisporus and C. parapsilosis differ in their susceptibility profiles to echinocandins .

What expression systems are most effective for producing recombinant L. elongisporus COA3?

When designing expression systems for recombinant L. elongisporus COA3, researchers should consider several factors specific to this mitochondrial membrane protein. Based on experimental approaches used for similar proteins, the following expression systems have demonstrated effectiveness:

Expression SystemAdvantagesChallengesOptimization Strategies
E. coli (BL21 DE3)Rapid growth, high yield, cost-effectivePotential improper folding, lack of post-translational modificationsUse fusion tags (MBP, SUMO), lower induction temperature (16-18°C), specialized E. coli strains for membrane proteins
Pichia pastorisEukaryotic folding machinery, post-translational modificationsLonger production time, more complex protocolsOptimize methanol induction parameters, use strong constitutive promoters like GAP
Saccharomyces cerevisiaeCloser evolutionary relationship to L. elongisporus, appropriate folding environmentLower yield than bacterial systemsUse GAL1 promoter, optimize codon usage for high expression
Baculovirus-Insect CellExcellent for complex eukaryotic membrane proteinsTechnical complexity, higher costUse honeybee melittin signal sequence for secretion, optimize MOI

When expressing mitochondrial membrane proteins like COA3, inclusion of the appropriate targeting sequences may be necessary for proper folding studies, while these sequences should be removed for structural analyses. Codon optimization based on the expression host is essential for maximizing protein yield. For functional studies, maintaining the native structure is critical, making yeast expression systems potentially advantageous despite their lower yields.

The choice of expression system should ultimately be guided by the specific research questions being addressed and the downstream applications planned for the recombinant protein.

How can researchers design experiments to characterize the role of COA3 in L. elongisporus mitochondrial function?

Designing experiments to characterize COA3 function in L. elongisporus requires a multi-faceted approach that accounts for this organism's unique biological properties. The following experimental design framework is recommended:

  • Gene disruption and phenotypic analysis:

    • Generate COA3 knockout mutants using CRISPR-Cas9 or homologous recombination methods

    • Evaluate growth phenotypes under respiratory conditions (glycerol medium) versus fermentative conditions (glucose medium)

    • Assess mitochondrial morphology using fluorescent microscopy with mitochondrial-specific dyes

    • Measure oxygen consumption rates in wild-type versus knockout strains

  • Protein localization and interaction studies:

    • Generate tagged versions of COA3 (GFP, FLAG, or HA tags)

    • Confirm mitochondrial localization using subcellular fractionation and immunofluorescence

    • Identify interaction partners through co-immunoprecipitation followed by mass spectrometry

    • Validate key interactions using techniques like bimolecular fluorescence complementation (BiFC)

  • Functional complementation experiments:

    • Test whether L. elongisporus COA3 can rescue phenotypes of COA3 mutants in model yeasts like S. cerevisiae

    • Introduce site-specific mutations to identify critical functional domains

    • Compare complementation efficiency across different pathogenic and non-pathogenic fungal species

  • Stress response evaluation:

    • Assess COA3 mutant susceptibility to oxidative stress, antifungal drugs, and other environmental stressors

    • Determine if COA3 expression changes under conditions that mimic the host environment

    • Evaluate biofilm formation capacity in wild-type versus COA3 mutant strains

When designing these experiments, researchers should consider L. elongisporus's unique characteristics, including its diploid nature and potential for sexual reproduction through ascospore formation . This may necessitate different genetic manipulation strategies than those used for haploid yeast species.

What purification strategies yield the highest purity and activity for recombinant L. elongisporus COA3?

Purifying recombinant membrane proteins like COA3 from L. elongisporus presents significant challenges due to their hydrophobic nature and tendency to aggregate. The following purification strategy has been optimized based on experiences with similar mitochondrial membrane proteins:

  • Solubilization optimization:

    • Test a panel of detergents including DDM, LMNG, CHAPS, and digitonin

    • Determine the critical micelle concentration (CMC) for each detergent with L. elongisporus membranes

    • Evaluate protein stability in each detergent using thermal shift assays

  • Affinity chromatography:

    • Incorporate polyhistidine (His6) or Strep-II tags at either N- or C-terminus, avoiding disruption of targeting sequences

    • Use cobalt resins rather than nickel for His-tagged proteins to reduce non-specific binding

    • Include low concentrations of detergent in all buffers to prevent aggregation

    • Optimize imidazole concentration gradients for elution to maximize purity

  • Size exclusion chromatography:

    • Separate monomeric protein from aggregates and other contaminants

    • Assess protein oligomeric state in different detergent conditions

    • Collect fractions and confirm identity by Western blotting with COA3-specific antibodies

  • Activity assessment:

    • Develop reconstitution protocols in proteoliposomes or nanodiscs

    • Measure interaction with cytochrome c oxidase subunits using surface plasmon resonance

    • Assess copper binding capacity using isothermal titration calorimetry

Purification StepKey ParametersQuality ControlExpected Yield
Membrane extractionpH 7.5, 4°C, protease inhibitorsWestern blot100% (reference)
Detergent solubilization1% DDM or 0.5% LMNG, 1 hour, 4°CClear supernatant after ultracentrifugation60-70%
Affinity chromatography20 mM imidazole (wash), 250 mM imidazole (elution)SDS-PAGE, silver stain30-40%
Size exclusionSuperdex 200, flow rate 0.5 ml/minSingle symmetrical peak15-20%
Concentration100 kDa cutoff concentratorProtein concentration assay10-15%

Researchers should be aware that yield and activity can vary significantly based on the expression system chosen and the specific detergents used during purification. Maintaining protein stability throughout the purification process is critical for downstream functional studies.

How does COA3 function in L. elongisporus potentially contribute to its emerging pathogenicity?

The potential contribution of COA3 to L. elongisporus pathogenicity represents an intriguing area of investigation that connects mitochondrial function to virulence mechanisms. Several hypotheses can be proposed based on current knowledge of fungal pathogenesis and mitochondrial proteins:

  • Metabolic adaptation: COA3's role in cytochrome oxidase assembly may enable L. elongisporus to rapidly adapt its energy metabolism in response to the nutrient-limited host environment. The fungus has been isolated from diverse clinical specimens, including blood, suggesting it can survive in different host niches .

  • Stress response coordination: Proper mitochondrial function is essential for responding to host-induced stresses, including oxidative bursts from immune cells. COA3 disruption could potentially compromise the pathogen's ability to survive these challenges. This is particularly relevant given that L. elongisporus has been isolated from immunocompromised patients .

  • Biofilm formation support: Mitochondrial proteins like COA3 may indirectly contribute to biofilm formation through energy provision. L. elongisporus has been isolated from catheter tips, suggesting biofilm formation capacity in at least some clinical strains . The energy requirements for extracellular matrix production and maintenance could depend on optimal cytochrome oxidase function.

  • Hyphal morphogenesis: While L. elongisporus is not known for extensive hyphal formation like C. albicans, phenotypic switching involving elongated cell formation has been observed in related species . COA3-dependent mitochondrial activity might support the metabolic demands of such morphological transitions.

  • Antifungal resistance connection: Mitochondrial function has been linked to azole drug resistance in some fungi. While L. elongisporus generally remains susceptible to conventional antifungals, elevated fluconazole MICs have been observed in some environmental isolates , suggesting potential resistance mechanisms that might involve mitochondrial functions.

Future research investigating COA3 knockout mutants in infection models like Galleria mellonella, which has been used to study L. elongisporus virulence , could provide direct evidence for this protein's contribution to pathogenicity.

What approaches can be used to study the structural biology of L. elongisporus COA3?

Studying the structural biology of membrane proteins like L. elongisporus COA3 presents significant challenges but can provide valuable insights into function. The following methodological approaches are recommended:

  • Computational structural prediction:

    • Generate homology models based on COA3 homologs with known structures

    • Apply AlphaFold2 or RoseTTAFold algorithms for ab initio prediction

    • Validate models through molecular dynamics simulations in simulated membrane environments

    • Predict functional domains and transmembrane regions using TMHMM, TOPCONS, and similar tools

  • X-ray crystallography approach:

    • Screen detergent and lipid combinations systematically to identify conditions promoting crystal formation

    • Use lipidic cubic phase (LCP) crystallization methods optimized for small membrane proteins

    • Incorporate thermostabilizing mutations based on alanine scanning results to improve crystallization propensity

    • Consider fusion with crystallization chaperones like T4 lysozyme or BRIL to provide crystal contacts

  • Cryo-electron microscopy (cryo-EM):

    • Prepare samples in nanodiscs or amphipols to maintain native-like lipid environment

    • Use Volta phase plates to enhance contrast for this small (~10-15 kDa) protein

    • Consider studying COA3 in complex with interaction partners to increase molecular weight for better cryo-EM analysis

    • Apply 3D variability analysis to capture potential conformational states

  • NMR spectroscopy:

    • Express isotopically labeled protein (15N, 13C) in minimal media

    • Use selective methyl labeling strategies for improved spectral quality

    • Apply solid-state NMR approaches for membrane-embedded states

    • Measure residual dipolar couplings to establish domain orientations

Structural MethodResolution RangeSample RequirementsKey AdvantagesMajor Limitations
Homology ModelingN/ASequence onlyRapid, low costAccuracy depends on template quality
X-ray Crystallography1.5-3.5 Å5-10 mg purified proteinAtomic resolution possibleCrystallization challenging for membrane proteins
Cryo-EM2.5-4 Å100-500 μg proteinNative-like conditionsSize limitations for small proteins
Solution NMR3-5 Å0.5-1 mM (2-5 mg) labeled proteinDynamic informationSize limitations, extensive optimization

The combination of these complementary approaches provides the most comprehensive structural understanding, with computational methods informing experimental design and experimental data validating computational predictions.

How can researchers investigate COA3's role in the context of L. elongisporus antifungal susceptibility?

Investigating the relationship between COA3 function and antifungal susceptibility in L. elongisporus requires carefully designed experiments that connect mitochondrial function to drug response. The following methodological framework is recommended:

  • Expression analysis during drug exposure:

    • Quantify COA3 mRNA and protein levels in response to different classes of antifungals

    • Use RT-qPCR and Western blotting to track expression changes at various time points after drug exposure

    • Compare expression patterns between susceptible isolates and those with elevated MICs (particularly for azoles)

    • Correlate expression levels with respiratory chain activity measurements

  • Susceptibility testing of COA3 mutants:

    • Generate COA3 knockout and overexpression strains

    • Determine MICs using standardized methods like those in the E-test

    • Test susceptibility to multiple drug classes (azoles, polyenes, echinocandins)

    • Examine potential synergistic effects between antifungals and mitochondrial inhibitors

  • Mitochondrial function assessment:

    • Measure oxygen consumption rates and membrane potential in wild-type versus mutant strains during drug exposure

    • Assess ROS production using fluorescent probes in response to antifungal treatment

    • Examine mitochondrial morphology changes using confocal microscopy during drug exposure

    • Quantify ATP production capacity under antifungal stress

  • Drug combination studies:

    • Evaluate synergistic potential between traditional antifungals and compounds targeting mitochondrial function

    • Use checkerboard assays similar to those used for testing fluconazole and FK506 combinations in L. elongisporus

    • Calculate fractional inhibitory concentration (FIC) indices to quantify interactions

    • Identify combinations that specifically target COA3-dependent pathways

What are common challenges in COA3 expression studies and how can they be addressed?

Researchers working with recombinant L. elongisporus COA3 may encounter several technical challenges that can impact experimental success. The following troubleshooting guide addresses common issues and their solutions:

  • Low expression yields:

    • Problem: Hydrophobic membrane proteins like COA3 often express poorly in standard systems

    • Solution: Optimize codon usage for the expression host, reduce induction temperature to 16-18°C, use specialized strains like C41(DE3) for E. coli expression, or switch to eukaryotic expression systems

    • Validation: Confirm improvement through Western blot analysis of small-scale test expressions

  • Protein aggregation and inclusion body formation:

    • Problem: Improper folding leading to insoluble aggregates

    • Solution: Add solubilizing fusion partners (MBP, SUMO, or Thioredoxin), include mild detergents during lysis, optimize induction conditions, or consider in vitro refolding protocols

    • Validation: Analyze soluble versus insoluble fractions by SDS-PAGE and Western blotting

  • Protein degradation:

    • Problem: Proteolytic cleavage during expression or purification

    • Solution: Add protease inhibitor cocktails, reduce purification time, use protease-deficient expression hosts, or identify and modify protease-sensitive sites

    • Validation: Western blot analysis with antibodies targeting different protein regions to identify degradation patterns

  • Poor protein activity:

    • Problem: Recombinant protein lacks expected functional characteristics

    • Solution: Preserve native N- and C-termini if possible, optimize detergent type and concentration, include stabilizing lipids during purification, or consider native purification approaches

    • Validation: Develop activity assays specific to COA3 function, such as cytochrome oxidase assembly complementation tests

IssuePossible CausesTroubleshooting ApproachesSuccess Indicators
No visible expressionToxicity, codon bias, improper inductionUse tightly regulated promoters, optimize codons, adjust induction parametersDetectable band on Western blot
Protein in inclusion bodiesRapid expression rate, improper foldingLower temperature, co-express chaperones, use solubility tagsIncreased protein in soluble fraction
Multiple bands/degradationProteolytic sensitivity, sample heatingAdd protease inhibitors, maintain cold chain, reduce purification timeSingle band of expected size
Poor functional activityImproper folding, missing cofactorsOptimize buffer conditions, add stabilizing agents, co-purify with partnersMeasurable biochemical activity

Researchers should also consider that L. elongisporus's optimal growth temperature and conditions may differ from standard laboratory yeasts, which could affect heterologous expression systems. Adapting protocols based on this organism's specific biology may improve results.

How can researchers resolve contradictory data when studying mitochondrial proteins in pathogenic yeasts?

When investigating mitochondrial proteins like COA3 in pathogenic yeasts such as L. elongisporus, researchers may encounter seemingly contradictory results. The following methodological framework helps resolve such discrepancies:

  • Organism-specific variations:

    • Contradiction: Functions established in model yeasts may not translate to L. elongisporus

    • Resolution approach: Perform comparative studies across multiple species, focusing on evolutionary conservation and divergence

    • Validation: Phylogenetic analysis combined with functional complementation experiments

  • Strain-to-strain variations:

    • Contradiction: Different L. elongisporus isolates may show varying phenotypes despite targeting the same gene

    • Resolution approach: Sequence the COA3 gene and its regulatory regions across multiple strains to identify polymorphisms

    • Validation: Whole genome sequencing and comparative genomics to identify compensatory mutations or strain-specific factors

  • Environmental condition dependencies:

    • Contradiction: COA3 function may appear different under varying growth or stress conditions

    • Resolution approach: Systematically test phenotypes across a matrix of conditions (temperature, pH, carbon source, oxygen levels)

    • Validation: Use transcriptomics and proteomics to identify condition-dependent expression patterns

  • Technical artifacts:

    • Contradiction: Different experimental methods yield conflicting results

    • Resolution approach: Apply multiple complementary techniques to address the same question

    • Validation: Independent verification in different laboratories or with different methodological approaches

When analyzing contradictory data specifically related to L. elongisporus, researchers should consider this organism's unique characteristics, including its ability to form ascospores despite lacking complete mating loci , its diploid nature, and evidence of recombination through secondary homothallism and/or frequent mitotic recombination . These biological features may explain phenotypic variations that initially appear contradictory.

The methodology for resolving contradictions should be systematic, with careful documentation of all experimental variables and thoughtful experimental design that includes appropriate controls.

What considerations are important when designing experiments to study COA3's interaction with the host immune system?

Investigating interactions between L. elongisporus COA3 and host immune responses requires carefully designed experiments that bridge microbiology, immunology, and molecular biology. The following methodological considerations are critical:

  • Purification of immunologically active protein:

    • Challenge: Obtaining COA3 preparations that maintain native conformational epitopes

    • Approach: Use gentle extraction methods that preserve protein structure, consider membrane protein nanodiscs to maintain native conformation

    • Validation: Circular dichroism spectroscopy to confirm secondary structure integrity, binding assays with known interaction partners

  • Host cell interaction models:

    • Challenge: Selecting appropriate immune cell types that reflect in vivo interactions

    • Approach: Use primary human neutrophils, macrophages, and dendritic cells; compare responses to those against whole L. elongisporus cells

    • Controls: Include parallel experiments with other mitochondrial proteins to distinguish COA3-specific effects from general mitochondrial protein recognition

  • Immune recognition assessment:

    • Challenge: Determining if COA3 is recognized by pattern recognition receptors

    • Approach: Use reporter cell lines expressing individual TLRs, NLRs, or CLRs to identify specific recognition pathways

    • Measurements: Quantify NF-κB activation, cytokine production, and phagocytosis efficiency

  • In vivo relevance evaluation:

    • Challenge: Connecting in vitro findings to in vivo infection scenarios

    • Approach: Use L. elongisporus COA3 mutants in Galleria mellonella infection models (already established for this organism )

    • Assessment: Compare survival curves, fungal burden, and immune cell recruitment between wild-type and mutant infections

This research direction is particularly relevant given the increasing clinical significance of L. elongisporus infections, especially in immunocompromised patients . Understanding how mitochondrial proteins like COA3 interact with host immunity may reveal novel therapeutic approaches or diagnostic markers.

Researchers should be aware that L. elongisporus has been isolated from diverse clinical sources and can cause various manifestations including systemic mycoses, fungemia, endocarditis, meningitis, oropharyngeal infections, and dermatitis . This clinical diversity suggests that interactions with the host immune system may be tissue-specific and should be considered in experimental design.

How might COA3 be targeted for novel antifungal development?

Exploring COA3 as a potential antifungal target requires understanding its unique features and essential functions in L. elongisporus. The following methodological framework outlines approaches for drug discovery targeting this protein:

  • Target validation approaches:

    • Genetic approach: Generate conditional mutants to confirm essentiality under various conditions

    • Chemical approach: Use known mitochondrial inhibitors to establish proof-of-concept for targeting respiratory chain assembly

    • Comparative genomics: Identify structural or functional differences between fungal and human homologs that could be exploited

  • High-throughput screening strategies:

    • Assay development: Create yeast-based reporter systems where COA3 function is linked to a readily measurable output (fluorescence, growth)

    • Compound libraries: Screen diverse chemical libraries, focusing on compounds with known activity against mitochondrial targets

    • Fragment-based approaches: Identify small molecular binders that can be elaborated into more potent inhibitors

  • Structure-based drug design:

    • Virtual screening: Use computational models of COA3 to identify potential binding pockets

    • Molecular docking: Screen in silico libraries against identified pockets to prioritize compounds

    • Medicinal chemistry: Optimize lead compounds through iterative design and testing

  • Biological evaluation pipeline:

    • Primary testing: Measure direct binding to recombinant COA3 and effects on protein function

    • Secondary screening: Assess antifungal activity against L. elongisporus and related pathogens

    • Specificity assessment: Compare activity against human mitochondrial homologs to establish selectivity

    • Resistance development: Evaluate the potential for resistance emergence through extended exposure experiments

This approach is particularly promising given that L. elongisporus remains susceptible to conventional antifungal agents but has shown the potential to develop resistance, as evidenced by elevated MICs against sodium hypochlorite (a common disinfecting agent) in multiple clinical-related isolates and elevated fluconazole MICs in several environmental isolates .

The specific targeting of mitochondrial assembly factors like COA3 may provide advantages over current antifungals by exploiting unique aspects of fungal respiratory metabolism. This could be especially valuable given the rising incidence of L. elongisporus infections and its emerging significance as a global pathogen .

What experimental approaches can elucidate the role of COA3 in L. elongisporus stress response and adaptation?

Understanding how COA3 contributes to stress response and adaptation in L. elongisporus requires integrative experimental approaches that connect mitochondrial function to broader cellular processes. The following methodology framework is recommended:

  • Transcriptional and translational response analysis:

    • Approach: Perform RNA-seq and ribosome profiling under various stress conditions (oxidative, osmotic, pH, temperature) in wild-type versus COA3 mutant strains

    • Controls: Include other mitochondrial protein mutants to distinguish COA3-specific responses

    • Data analysis: Focus on differential expression patterns and identification of co-regulated gene networks

  • Metabolic flux analysis:

    • Approach: Use 13C-labeled substrates to track metabolic changes in response to stress

    • Measurements: Quantify TCA cycle intermediates, amino acids, and lipids using LC-MS/MS

    • Comparison: Analyze wild-type versus COA3 mutant metabolic adaptations to identify COA3-dependent pathways

  • Mitochondrial dynamics assessment:

    • Approach: Use live-cell imaging with fluorescently tagged mitochondria to monitor morphological changes

    • Parameters: Quantify fission/fusion events, motility, and distribution patterns under stress

    • Analysis: Correlate dynamics with respiratory capacity and stress resistance

  • Stress granule and protein aggregation studies:

    • Approach: Examine formation of stress granules and protein aggregates in response to various stressors

    • Techniques: Use fluorescent protein tagging and confocal microscopy

    • Hypothesis: Test if COA3 dysfunction alters cellular protein quality control mechanisms

This research direction is particularly relevant given L. elongisporus's environmental adaptability, evidenced by its isolation from diverse sources and its ability to survive in challenging environments including hospital settings . Understanding COA3's role in stress response may provide insights into how this organism persists in clinical environments and transitions to a pathogenic lifestyle.

The ability of L. elongisporus to form ascospores likely contributes to its environmental persistence, as ascospores are generally more resistant to environmental pH fluctuation and extreme temperatures than vegetative cells . Investigating how COA3 and mitochondrial function contribute to ascospore formation and stress resistance could reveal important adaptation mechanisms.

How can comparative studies of COA3 across fungal species advance our understanding of mitochondrial evolution in pathogenic yeasts?

Comparative studies of COA3 across different fungal species offer valuable insights into mitochondrial evolution and potential connections to pathogenicity. The following methodological framework outlines approaches for such comparative analyses:

  • Phylogenomic analysis:

    • Approach: Perform comprehensive sequence analysis of COA3 homologs across diverse fungal lineages

    • Tools: Maximum likelihood and Bayesian phylogenetic methods, selection pressure analysis (dN/dS ratios)

    • Focus: Identify lineage-specific adaptations, particularly in pathogenic versus non-pathogenic species

  • Structural comparison:

    • Approach: Generate structural models for COA3 from multiple species using AlphaFold2 or similar methods

    • Analysis: Compare binding pockets, surface electrostatics, and interaction interfaces

    • Hypothesis: Test whether structural features correlate with pathogenicity or ecological niche

  • Functional complementation experiments:

    • Approach: Express COA3 from different species in a standard S. cerevisiae COA3 knockout model

    • Measurements: Assess respiratory growth, cytochrome c oxidase assembly, and stress response

    • Analysis: Correlate functional conservation with phylogenetic distance and ecological niche

  • Protein interaction network comparison:

    • Approach: Identify COA3 interaction partners across species using immunoprecipitation followed by mass spectrometry

    • Analysis: Construct and compare protein interaction networks to identify conserved versus species-specific interactions

    • Hypothesis: Test whether interaction network complexity correlates with pathogenic potential

This comparative approach is particularly relevant for L. elongisporus given its phylogenetic positioning. L. elongisporus shares close evolutionary relationships with significant pathogenic Candida species but has some distinct characteristics, including its ability to form ascospores . Understanding the evolution of mitochondrial proteins like COA3 may provide insights into the emergence of pathogenicity in this and related species.

The comparison should include both closely related pathogens (C. parapsilosis, C. orthopsilosis, C. metapsilosis) and more distant relatives, as well as environmental versus clinical isolates. This approach may reveal how mitochondrial proteins have evolved in response to different selective pressures, including host adaptation and antifungal exposure.

Quick Inquiry

Personal Email Detected
Please use an institutional or corporate email address for inquiries. Personal email accounts ( such as Gmail, Yahoo, and Outlook) are not accepted. *
© Copyright 2025 TheBiotek. All Rights Reserved.