Recombinant Trichophyton rubrum Extracellular metalloproteinase 3 (MEP3)

Shipped with Ice Packs
In Stock

Description

Introduction to Recombinant Trichophyton rubrum Extracellular Metalloproteinase 3 (MEP3)

Recombinant Trichophyton rubrum Extracellular metalloproteinase 3 (MEP3) is a protein derived from the dermatophyte fungus Trichophyton rubrum. This enzyme belongs to the metalloproteinase family, which plays a crucial role in the pathogenicity of dermatophytes by facilitating invasion and degradation of host tissues. MEP3 is produced through recombinant DNA technology, allowing for its expression in various host systems such as E. coli, yeast, baculovirus, or mammalian cells .

Biological Function and Pathogenicity

Metalloproteinases, including MEP3, are essential for the virulence of dermatophytes. They contribute to the breakdown of keratin and other proteins in the host's skin, nails, and hair, enabling the fungus to invade and colonize these tissues . The expression of MEP3 and other proteases is often upregulated in environments that mimic host conditions, such as media containing nail chips, indicating their role in adapting to and infecting human tissues .

Research Findings and Applications

Research on MEP3 and similar metalloproteinases has focused on understanding their role in dermatophyte infections and exploring potential therapeutic targets. Studies have shown that these enzymes are critical for the adhesion and invasion processes of dermatophytes like T. rubrum and T. tonsurans . The recombinant form of MEP3 can be used in scientific research to study its enzymatic activity, its interaction with host tissues, and its potential as a target for antifungal therapies.

Production and Purity

Recombinant MEP3 is produced with a purity of greater than or equal to 85%, as determined by SDS-PAGE, ensuring its suitability for various biochemical and immunological assays . The availability of recombinant MEP3 facilitates detailed studies on its structure, function, and potential applications in biotechnology and medicine.

Table: Comparison of MEP3 with Other Metalloproteinases

EnzymeSourceFunctionPurity
MEP3T. rubrumPathogenicity, Tissue Invasion≥ 85%
MEP3T. tonsuransPathogenicity, Tissue Invasion≥ 85%
MMP3Human/MouseTissue Remodeling, Inflammation> 90%

Product Specs

Form
Lyophilized powder
Note: While we prioritize shipping the format currently in stock, please specify your preferred format in order notes for customized preparation.
Lead Time
Delivery times vary depending on the purchasing method and location. Please consult your local distributor for precise delivery estimates.
Note: Standard shipping includes blue ice packs. Dry ice shipping requires prior arrangement and incurs additional charges.
Notes
Avoid repeated freeze-thaw cycles. Store working aliquots at 4°C for up to one week.
Reconstitution
Centrifuge the vial briefly before opening to collect the contents. Reconstitute the protein in sterile, deionized water to a concentration of 0.1-1.0 mg/mL. For long-term storage, we recommend adding 5-50% glycerol (final concentration) and aliquoting at -20°C/-80°C. Our standard glycerol concentration is 50%, provided as a reference.
Shelf Life
Shelf life depends on various factors, including storage conditions, buffer composition, temperature, and protein stability. Generally, liquid formulations have a 6-month shelf life at -20°C/-80°C, while lyophilized formulations have a 12-month shelf life at -20°C/-80°C.
Storage Condition
Upon receipt, store at -20°C/-80°C. Aliquot for multiple uses. Avoid repeated freeze-thaw cycles.
Tag Info
The tag type is determined during the manufacturing process.
The tag type is determined during production. If you require a specific tag, please inform us, and we will prioritize its development.
Synonyms
MEP3Extracellular metalloproteinase 3; EC 3.4.24.-; Fungalysin MEP3
Buffer Before Lyophilization
Tris/PBS-based buffer, 6% Trehalose.
Datasheet
Please contact us to get it.
Expression Region
247-633
Protein Length
Extracellular domain
Purity
>85% (SDS-PAGE)
Species
Trichophyton rubrum (Athlete's foot fungus) (Epidermophyton rubrum)
Target Names
MEP3
Target Protein Sequence
AEYK VFAWGLNDPT EGNPTSIRDP WTDASPYTWN SDGMSKYPTT RGNNAIAQDN PTGGSTYINN YRPQSPNLIF SYPWSPTATP PSSYKDFSIT QLFYTTNRYH DLLYSFGFNE AAGNFQVNNG NKGGKGNDFA IVNAQDGSGT NNANFATPPD GSPGRMRMYN WTTARPNRDG CLEAGIVIHE YTHGLSNRLC GGPANSACLN ALESGGMGEG WGDFYATAIR LKPRDTKNTN YSMGAWAANN PKGIRAYLYS TNLQTNPYMY TSVNSLREVH QIGTVWASML YDLMWALIEA HGGTYSANPV FRNGVPQDGR HLSMKLVMDG MALQPCNPNF VQARDAILDA DRALTNSANK CTIWKAFAKR GLGYGAKYDA RNRTGSNKLP PGC
Uniprot No.

Target Background

Function

Secreted metalloproteinase; likely functions as a virulence factor.

Protein Families
Peptidase M36 family
Subcellular Location
Secreted.

Q&A

What is the evolutionary significance of the MEP gene family in dermatophytes?

The MEP (metalloprotease) family in dermatophytes represents an important group of virulence-related factors. Phylogenetic analysis of MEP genes from T. rubrum, T. mentagrophytes, and M. canis reveals that metalloproteases secreted by these three species are encoded by orthologous genes. This strongly suggests that multiplication of an ancestral metalloprotease gene occurred prior to dermatophyte species divergence . The conservation of these genes across multiple dermatophyte species indicates their fundamental importance in fungal biology and pathogenesis. MEP3 specifically shows high conservation, being present in approximately 81% of tested dermatophyte species, suggesting it may play a critical role in dermatophyte survival and pathogenicity .

How does MEP3 relate to other members of the metalloprotease family in T. rubrum?

MEP3 is one of five members of the secreted metalloprotease family in T. rubrum. While all MEP family members share structural similarities, they exhibit distinct expression patterns and potentially different substrate specificities. Among the MEP family members, MEP3 has one of the highest conservation rates across dermatophyte species (81%), compared to MEP1 and MEP2 (70%), MEP4 (54%), and MEP5 (36%) . This differential conservation suggests varying functional importance of each MEP in dermatophyte biology. Research indicates that these metalloproteases likely evolved from a common ancestral gene through duplication events that occurred before dermatophyte species diverged from one another .

What are the structural characteristics of the MEP3 gene and protein from T. rubrum?

The MEP3 gene in T. rubrum belongs to a family of genes encoding secreted metalloproteases. Based on analysis of related dermatophyte MEP genes, the gene likely contains multiple exons and introns with typical fungal consensus sequences at exon-intron boundaries and splice signals for lariat formation . The intron structure would follow patterns similar to other characterized fungal genes, containing the canonical GT at 5' splice sites and AG at 3' splice sites.

The encoded MEP3 protein belongs to the M36 fungalysin metalloprotease family. As a metalloprotease, it requires metal ions (typically zinc) for catalytic activity and contains a conserved HEXXH motif in its active site, where the two histidines coordinate with the metal ion and the glutamate serves as a catalytic base. The protein is secreted extracellularly, containing a signal peptide sequence at its N-terminus that directs it to the secretory pathway .

What is the functional role of MEP3 in T. rubrum pathogenesis?

MEP3, as a secreted metalloprotease, likely plays a crucial role in T. rubrum pathogenesis through multiple mechanisms:

  • Nutrient acquisition: MEP3 helps T. rubrum obtain nutrients by degrading host proteins, particularly keratin, the predominant protein in skin, hair, and nails .

  • Host tissue invasion: By breaking down structural proteins in the host's epidermis, MEP3 facilitates fungal penetration and colonization of skin tissues .

  • Immune modulation: MEP3 may degrade components of the host immune system, helping the fungus evade host defenses.

  • Virulence factor: The high conservation rate of MEP3 (81%) across dermatophyte species compared to other MEP family members suggests its importance as a virulence determinant .

Research comparing T. rubrum with other dermatophytes reveals that these organisms are enriched for several classes of proteases necessary for fungal growth and nutrient acquisition on keratinized tissues . The secretion of these proteases, including MEP3, represents an adaptation to the ecological niche of dermatophytes as specialized pathogens of keratinized structures.

What are the optimal expression systems for producing recombinant T. rubrum MEP3?

For producing recombinant T. rubrum MEP3, several expression systems can be considered, each with distinct advantages and limitations:

  • E. coli expression system:

    • Advantages: Rapid growth, high yield, cost-effectiveness

    • Challenges: Lack of post-translational modifications, potential improper folding of eukaryotic proteins

    • Optimization: Use of specialized strains (BL21(DE3), Rosetta), codon optimization, fusion with solubility-enhancing tags (MBP, SUMO)

  • Yeast expression systems (S. cerevisiae, P. pastoris):

    • Advantages: Eukaryotic post-translational modifications, secretion capacity

    • Optimization: Use of strong inducible promoters (AOX1 for P. pastoris), optimization of culture conditions

    • Secretion: Incorporation of yeast α-factor secretion signal

  • Baculovirus-insect cell system:

    • Advantages: Superior folding, post-translational modifications closer to mammalian systems

    • Applications: When high biological activity and proper folding are critical

The optimal choice depends on the research requirements for protein yield, purity, activity, and downstream applications. For structural studies requiring high purity, the P. pastoris system often provides the best balance of yield and proper folding for fungal metalloproteases .

What purification strategies are most effective for isolating recombinant MEP3 while preserving enzymatic activity?

A multi-step purification strategy is recommended for isolating functional recombinant MEP3:

Stage 1: Initial Capture

  • Immobilized Metal Affinity Chromatography (IMAC): For His-tagged constructs

  • Affinity purification: Using specifically designed substrates or inhibitors

  • Ion exchange chromatography: Based on MEP3's predicted isoelectric point

Stage 2: Intermediate Purification

  • Size Exclusion Chromatography: To separate monomeric protein from aggregates

  • Hydrophobic Interaction Chromatography: For additional purity

Stage 3: Activity Preservation

  • Buffer optimization: Include 10μM ZnCl₂ to maintain metalloprotease activity

  • pH control: Maintain pH 7.0-7.5 for optimal stability

  • Storage conditions: Add 10% glycerol and store at -80°C in small aliquots

Critical considerations:

  • Avoid metal chelators (EDTA) at all stages as they will inactivate the metalloprotease

  • Include protease inhibitors specific for serine and cysteine proteases to prevent degradation while not affecting MEP3 activity

  • Monitor enzymatic activity throughout purification using fluorogenic peptide substrates

Typical yield from optimized P. pastoris expression can reach 5-10 mg of purified MEP3 per liter of culture, with specific activity of approximately 50-200 units/mg protein depending on substrate used .

How can researchers design assays to accurately measure MEP3 enzymatic activity?

Substrate-Based Activity Assays:

Substrate TypeAdvantagesLimitationsDetection Method
Synthetic peptides with FRET pairsHigh sensitivity, real-time monitoringNot physiologicalFluorescence spectroscopy
Keratin azureNatural substrateLow sensitivity, endpoint onlyAbsorbance (595nm)
Fluorescein-labeled keratinNatural substrate with high sensitivityComplex preparationFluorescence
AzocaseinEasy preparation, moderate sensitivityNot a natural substrateAbsorbance (440nm)

Recommended Standard Assay Protocol:

  • Buffer conditions: 50mM Tris-HCl (pH 7.5), 100mM NaCl, 5mM CaCl₂, 1μM ZnCl₂

  • Temperature: 28°C (physiological for dermatophytes)

  • Substrate concentration: 5-50μM for synthetic substrates

  • Controls:

    • Negative: Heat-inactivated enzyme

    • Positive: Commercial metalloproteases

    • Specificity: Include metalloprotease inhibitors (1,10-phenanthroline)

Data Analysis:

  • Calculate specific activity (μmol product/min/mg enzyme)

  • Determine kinetic parameters (Km, Vmax, kcat) using Michaelis-Menten analysis

  • For inhibition studies, calculate IC₅₀ and Ki values

The enzymatic activity of MEP3 can be influenced by pH, temperature, and metal ion concentration, so these parameters should be carefully controlled and reported in publications .

What are the common challenges in expressing functional recombinant MEP3 and how can they be overcome?

Challenge 1: Low expression levels

  • Solution: Optimize codon usage for the expression host, use strong promoters (AOX1 for P. pastoris)

  • Assessment: Verify mRNA levels via qRT-PCR to determine if the issue is transcriptional

Challenge 2: Protein misfolding and inclusion body formation

  • Solution: Lower induction temperature (16-20°C), co-express chaperones, use solubility-enhancing fusion tags

  • Assessment: Analyze soluble vs. insoluble fractions by SDS-PAGE

Challenge 3: Proteolytic degradation

  • Solution: Use protease-deficient strains, optimize culture harvest timing, include protease inhibitors

  • Assessment: Western blot analysis with anti-MEP3 antibodies to identify degradation products

Challenge 4: Inactive enzyme

  • Solution: Ensure proper metal ion (Zn²⁺) incorporation, optimize refolding protocols

  • Assessment: Compare activity with native enzyme using standardized assays

Challenge 5: Glycosylation differences

  • Solution: Identify and mutate non-essential N-glycosylation sites or use EndoH treatment

  • Assessment: Analyze glycosylation pattern by mass spectrometry or lectin binding assays

Statistical approach to optimization:
Implement a Design of Experiments (DoE) approach to systematically test multiple parameters (temperature, pH, media composition, induction time) simultaneously rather than the traditional one-factor-at-a-time method. This can reduce experimental time by up to 75% while identifying important parameter interactions .

How does T. rubrum MEP3 compare functionally to orthologous proteases in other dermatophyte species?

Comparative Functional Analysis of MEP3 Across Dermatophyte Species:

SpeciesMEP3 PrevalenceSubstrate PreferenceEnzymatic Efficiency (kcat/Km)Notable Differences
T. rubrumHighBroad spectrumHigh on keratinReference standard
T. mentagrophytesHighBroad spectrumSimilar to T. rubrumHigher thermostability
M. canisPresentHigher activity on hair keratinModerateDifferent pH optimum
T. simiiPresentNot fully characterizedNot fully characterizedContains all five MEP genes, higher antifungal resistance

Key Functional Comparisons:

  • Substrate specificity: While all dermatophyte MEP3 enzymes can degrade keratin, subtle differences in preference for specific keratin types (hair, nail, skin) exist between species, reflecting their natural infection sites.

  • Catalytic efficiency: Comparative studies suggest that MEP3 enzymes from anthropophilic species (T. rubrum) may have evolved different catalytic properties compared to zoophilic species (M. canis), potentially reflecting adaptation to human hosts.

  • Immunogenic properties: MEP3 proteins from different species exhibit varying degrees of immunogenicity, which may contribute to differences in inflammatory responses observed in infections by different dermatophytes.

  • Gene expression patterns: In T. rubrum, MEP3 expression is induced under specific nutrient conditions, particularly in the presence of keratin, while regulatory patterns may differ in other species .

The high conservation of MEP3 across dermatophyte species (81%) suggests its fundamental importance in dermatophyte biology, though species-specific adaptations in enzyme properties likely contribute to host preference and infection characteristics .

What is the relationship between MEP3 expression and antifungal drug resistance in T. rubrum?

Recent research has revealed intriguing correlations between MEP genes and antifungal resistance in dermatophytes. While MEP3 specifically is found in 81% of tested dermatophyte species, comprehensive analysis shows that:

  • Correlation with resistance profile: Dermatophyte species containing more MEP genes (particularly all five genes) demonstrate higher resistance to common antifungals. For example, T. simii, which contains all five MEP genes (MEP1-5), shows high resistance to multiple antifungals .

  • MEP5 as a resistance marker: The presence of MEP5 particularly correlates with increased antifungal resistance, suggesting either a direct role in resistance or co-selection of resistance factors .

  • Mechanistic hypotheses:

    • MEPs may degrade antifungal compounds directly

    • MEPs could modify cell wall components, affecting drug penetration

    • MEP expression might be co-regulated with efflux pumps or other resistance mechanisms

  • Clinical implications: The presence of specific MEP genes could potentially serve as molecular markers for predicting treatment outcomes.

While direct mechanistic links between MEP3 and antifungal resistance remain to be fully elucidated, these correlations suggest important avenues for future research. Understanding this relationship could lead to improved therapeutic approaches and potentially novel combination therapies targeting both the fungal proteases and traditional antifungal targets .

How can structural analysis of recombinant MEP3 inform the design of specific inhibitors?

Structural Approaches to MEP3 Inhibitor Design:

Comprehensive structural analysis of MEP3 can accelerate the development of specific inhibitors through several approaches:

  • X-ray crystallography and cryo-EM studies:

    • Resolution of 3D structure at atomic level (typically 1.5-2.5Å)

    • Co-crystallization with substrate analogs to identify binding interactions

    • Analysis of the catalytic site geometry and metal coordination

  • Computational approaches:

    • Molecular dynamics simulations to understand conformational flexibility

    • Virtual screening of compound libraries against the MEP3 active site

    • Structure-based drug design targeting unique features of the catalytic domain

  • Structure-activity relationship analysis:

    • Identification of crucial active site residues through site-directed mutagenesis

    • Mapping of substrate specificity determinants

    • Analysis of conformational changes upon substrate binding

Key Structural Features for Inhibitor Design:

  • The HEXXH motif in the active site and zinc coordination geometry

  • Unique substrate-binding pockets that differ from human metalloproteases

  • Allosteric sites that could be targeted for non-competitive inhibition

  • Species-specific structural elements that could enable selective targeting

Using these approaches, researchers can design inhibitors with high specificity for MEP3 while minimizing cross-reactivity with human metalloproteases, potentially leading to novel antifungal therapies with reduced side effects .

What role does MEP3 play in the complex pathogenesis of dermatophyte infections compared to other virulence factors?

MEP3 in the Context of Dermatophyte Pathogenesis:

Dermatophyte infections involve a complex interplay of multiple virulence factors, with MEP3 serving as an important component in this pathogenic machinery. Analysis of its role relative to other factors reveals:

  • Initial Infection Phase:

    • Adhesins and cell wall components mediate initial attachment

    • Non-specific proteases create initial penetration

    • MEP3 likely plays a secondary role during this phase

  • Established Infection:

    • MEP3 becomes crucial for nutrient acquisition from keratin

    • Works synergistically with other proteases (subtilisins, dipeptidyl-peptidases)

    • Contributes to tissue degradation and fungal spread

  • Host Response Modulation:

    • MEP3 may degrade host antimicrobial peptides

    • Other factors (mannans, cell wall components) primarily drive inflammatory responses

    • Secondary metabolites and toxins modulate local tissue environment

Comparative Contribution Analysis:

Virulence FactorPrimary FunctionRelative ContributionInteraction with MEP3
Keratinases (Subtilisins)Keratin degradationHighSynergistic, may process substrates for MEP3
LipasesLipid degradationModerateIndependent pathway
MannansImmune modulationHigh in inflammatory responseNo direct interaction
LysM domain proteinsChitin binding, immune evasionHigh in persistenceMay protect MEP3 from host proteases
Secondary metabolitesVarious, including toxicityVariableMay create optimal pH for MEP3 activity

The genomic analysis of T. rubrum and related dermatophytes has revealed enrichment for several protease families, LysM domain-containing proteins, and secondary metabolite biosynthesis genes, suggesting an integrated virulence strategy where MEP3 functions as part of a coordinated system rather than in isolation .

What are the most reliable methods for analyzing MEP3 gene expression in T. rubrum under different experimental conditions?

Comprehensive MEP3 Expression Analysis Methodology:

For accurate quantification of MEP3 expression under various conditions, researchers should employ a multi-method approach:

  • Quantitative RT-PCR (RT-qPCR):

    • Primers design: Target unique regions of MEP3 to avoid cross-amplification of other MEP family members

    • Reference genes: Use at least three stable reference genes (e.g., actin, 18S rRNA, GAPDH) validated for stability under your experimental conditions

    • Normalization: Apply the 2^-ΔΔCt method with proper validation of primer efficiency

  • RNA-Seq analysis:

    • Sample preparation: Ensure high RNA integrity (RIN > 8)

    • Sequencing depth: Minimum 20 million reads per sample for adequate coverage

    • Bioinformatic analysis: Use specialized pipelines for fungal transcriptomes with proper normalization for GC content

  • Protein-level validation:

    • Western blotting: Using specific anti-MEP3 antibodies

    • Enzymatic activity assays: Correlate transcript levels with functional activity

    • Proteomics: Quantitative mass spectrometry for broader protein expression context

Experimental Conditions to Test:

ConditionRationaleKey ControlsExpected Outcome
Keratin mediumNatural substrateGlucose mediumUpregulation
Various pH values (4.0-7.0)Environmental adaptationStandard pH 5.5pH-dependent expression
Antifungal exposureStress responseVehicle controlPossible upregulation
Temperature variationHost fever response28°C (optimal growth)Temperature-dependent regulation
Co-culture with keratinocytesHost-pathogen interactionFungus-only cultureComplex regulation patterns

Statistical Analysis:

  • Use appropriate statistical tests (ANOVA with post-hoc tests for multiple conditions)

  • Apply false discovery rate correction for RNA-Seq data

  • Perform at least three biological replicates for reliable results .

How can researchers address the challenge of MEP3 sequence variability when designing experiments across different T. rubrum strains?

Managing MEP3 Sequence Variability in Experimental Design:

MEP3 sequence variations across T. rubrum strains present significant challenges for experimental design and data interpretation. A systematic approach includes:

  • Sequence Comparison and Variant Identification:

    • Perform multiple sequence alignment of MEP3 from different strains

    • Identify conserved regions, variable domains, and critical functional motifs

    • Classify variations as synonymous or non-synonymous substitutions

  • Primer and Probe Design Strategies:

    • Universal detection: Design primers/probes targeting highly conserved regions

    • Strain-specific detection: Design primers spanning unique variant regions

    • Degenerate primers: Include mixed bases at variable positions

    • Verification: Always verify amplicon identity by sequencing

  • Expression Construct Considerations:

    • Use strain-specific promoters when studying native expression

    • For recombinant expression, test multiple strain variants to assess functional differences

    • Consider creating chimeric constructs to isolate effects of specific variations

  • Functional Impact Assessment:

    • Comparative enzymatic assays: Test activity of variants on standardized substrates

    • Structural modeling: Predict effects of amino acid substitutions on protein folding and activity

    • Mutagenesis studies: Create site-directed mutants to directly test the impact of specific variations

Statistical Approaches for Handling Variability:

Analytical ChallengeRecommended ApproachBenefits
Multiple strain comparisonsHierarchical clustering analysisIdentifies related strain groups
Correlating sequence to functionMultivariate regression modelsReveals key determinants of functional differences
Evolutionary analysisSelection pressure calculation (dN/dS)Identifies functionally important residues
Structure-function relationshipsPrincipal component analysis of sequence-activity dataReduces dimensionality of complex datasets

By systematically addressing sequence variability, researchers can design more robust experiments, avoid misinterpretation of data, and potentially uncover strain-specific adaptations that contribute to virulence or drug resistance profiles .

What are the most promising applications of recombinant MEP3 in dermatophyte vaccine development?

Recombinant MEP3 as a Vaccine Candidate:

The development of vaccines against dermatophytosis remains challenging, but recombinant MEP3 offers several promising approaches:

  • Inactivated Protease Vaccines:

    • Recombinant MEP3 could be chemically or genetically inactivated while preserving immunogenic epitopes

    • Advantages: Maintains protein structure, potentially elicits neutralizing antibodies

    • Challenges: Ensuring complete inactivation, potential for allergic reactions

  • Epitope-Based Vaccines:

    • Identification of immunodominant B-cell and T-cell epitopes from MEP3

    • Design of multi-epitope constructs combining epitopes from MEP3 and other virulence factors

    • Advantages: Reduced allergenicity, focused immune response

    • Applications: Particularly promising for preventing recurrent infections

  • DNA Vaccine Approaches:

    • Plasmids encoding modified MEP3 sequences

    • Advantages: Induces both humoral and cell-mediated immunity

    • Challenges: Optimizing delivery and expression in host cells

Immunological Considerations:

  • MEP3's high conservation (81%) across dermatophyte species suggests potential for cross-protection

  • Need to balance protective immunity vs. hypersensitivity reactions

  • Requirement for appropriate adjuvants to direct non-allergenic immune responses

Preliminary Research Data Required:

  • Animal model validation of protection efficacy

  • Determination of correlates of protection

  • Assessment of cross-protection against multiple dermatophyte species

The high conservation of MEP3 across dermatophyte species makes it particularly attractive as a broadly protective antigen, though careful immunological studies are needed to ensure safety and efficacy .

How might genomic and proteomic approaches advance our understanding of MEP3 regulation networks in T. rubrum?

Integrated Omics Approaches for MEP3 Regulation Networks:

Advanced genomic and proteomic techniques offer unprecedented opportunities to unravel the complex regulatory networks controlling MEP3 expression and function:

  • Chromatin Immunoprecipitation Sequencing (ChIP-seq):

    • Identification of transcription factors binding to MEP3 promoter regions

    • Mapping of chromatin modifications associated with MEP3 activation/repression

    • Integration with transcriptomic data to build regulatory models

  • Proteome-Wide Interaction Studies:

    • Yeast two-hybrid or proximity labeling approaches to identify MEP3 protein interaction partners

    • Mass spectrometry-based interactome analysis under various infection conditions

    • Correlation of protein complexes with enzymatic activity and localization

  • Systems Biology Integration:

    • Network analysis combining transcriptomic, proteomic, and metabolomic data

    • Identification of key regulatory hubs controlling virulence factor expression

    • Mathematical modeling of MEP regulation in response to environmental signals

Emerging Technologies with High Potential:

TechnologyApplication to MEP3 ResearchExpected Insights
Single-cell RNA-seqAnalysis of expression heterogeneityIdentification of fungal subpopulations with distinct virulence profiles
CRISPR-Cas9 gene editingSystematic disruption of putative regulatorsDirect validation of regulatory relationships
Ribosome profilingAnalysis of translational regulationUnderstanding post-transcriptional control mechanisms
Spatial transcriptomicsMapping gene expression in infection modelsContextual understanding of MEP3 expression during host interaction

Translational Impact:
These approaches could identify master regulators controlling multiple virulence factors simultaneously, potentially revealing novel drug targets that could inhibit virulence expression rather than fungal growth, representing a promising strategy to overcome antifungal resistance .

What are the remaining knowledge gaps in our understanding of T. rubrum MEP3 and how might they be addressed?

Despite significant advances in our understanding of T. rubrum MEP3, several critical knowledge gaps remain that warrant further investigation:

  • Structure-Function Relationships:

    • High-resolution crystal structures of T. rubrum MEP3 are lacking

    • The precise mechanisms of substrate recognition remain undefined

    • The role of potential post-translational modifications is poorly understood

  • Regulatory Networks:

    • The complete transcriptional and post-transcriptional regulatory mechanisms controlling MEP3 expression are not fully mapped

    • Environmental sensing pathways linking host conditions to MEP3 expression remain obscure

    • Potential cross-talk between MEP3 and other virulence factors needs clarification

  • Host-Pathogen Interactions:

    • The exact role of MEP3 in modulating host immune responses requires further study

    • Potential interactions between MEP3 and host proteases or inhibitors are understudied

    • The contribution of MEP3 to chronic or recurrent infections remains to be determined

Recommended Research Approaches:

To address these knowledge gaps, researchers should consider:

  • Structural biology approaches: X-ray crystallography and cryo-EM studies of MEP3 alone and in complex with substrates or inhibitors

  • Systems biology: Integration of transcriptomic, proteomic, and metabolomic data to build comprehensive regulatory networks

  • Advanced imaging techniques: In situ visualization of MEP3 during infection using fluorescently tagged proteins or specific antibodies

  • Humanized animal models: Development of better infection models that more accurately recapitulate human dermatophytosis

  • Clinical correlations: Studies linking MEP3 variants or expression levels with disease severity and treatment outcomes

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.