Recombinant Mycobacterium ulcerans Malate dehydrogenase (mdh)

Shipped with Ice Packs
In Stock

Description

Introduction to Recombinant Mycobacterium ulcerans MDH

Malate dehydrogenase (MDH; EC 1.1.1.37) is a critical enzyme in the citric acid cycle, catalyzing the reversible oxidation of malate to oxaloacetate using NAD+ as a cofactor . The recombinant form of M. ulcerans MDH is synthesized in heterologous expression systems such as Escherichia coli, yeast, or mammalian cells , enabling precise study of its biochemical and immunological roles in Buruli ulcer pathogenesis.

Biochemical Characteristics

Recombinant M. ulcerans MDH shares structural and functional homology with other mycobacterial MDHs, including those from M. tuberculosis. Key features include:

PropertyDetail
Molecular FunctionCatalyzes malate ↔ oxaloacetate interconversion with NAD+/NADH cycling .
Amino Acid Sequence329 residues (e.g., partial sequence: MSASPLKVAV TGAAGQIGYS LLFRLASGSL...) .
Purity>85% (verified by SDS-PAGE) .
Storage StabilityStable at -20°C; long-term storage recommended at -80°C .

Diagnostic and Immunological Studies

  • Antigen for Serodiagnosis: Recombinant MDH has been tested in indirect ELISA (iELISA) formats to detect antibodies in bovine brucellosis, demonstrating 71.7% diagnostic sensitivity and 84.4% specificity . While these studies focus on Brucella, the methodology is applicable to mycobacterial research.

  • Vaccine Development: Serves as an immunogen to study immune responses against M. ulcerans .

Enzymatic Activity and Regulation

  • Kinase Interactions: In M. tuberculosis, MDH is phosphorylated by serine/threonine protein kinases (STPKs), including PknD, which inhibits its activity . This regulatory mechanism likely extends to M. ulcerans due to conserved metabolic pathways.

  • Activity Assays: MDH activity is measured spectrophotometrically at 340 nm by tracking NADH oxidation. Phosphorylation reduces catalytic efficiency, as shown in M. tuberculosis .

KinaseEffect on MDH ActivityExperimental Conditions
PknDInhibitsPhosphate-depleted medium
PknA/PknHInhibitsIn vitro phosphorylation assays

Comparative Analysis with Other Mycobacterial MDHs

  • Structural Conservation: The recombinant M. ulcerans MDH shares >90% sequence identity with M. tuberculosis MDH, suggesting similar substrate binding and catalytic sites .

  • Functional Divergence: Unlike M. tuberculosis MDH, which is regulated by multiple STPKs, M. ulcerans MDH phosphorylation remains understudied but is hypothesized to respond to environmental stressors like hypoxia .

Challenges and Future Directions

  • Cross-Reactivity: Serological tests using MDH may cross-react with antibodies from other mycobacterial infections, necessitating complementary biomarkers .

  • Phosphorylation Dynamics: Further studies are required to map phosphorylation sites in M. ulcerans MDH and their impact on virulence.

Product Specs

Form
Lyophilized powder. We will ship the in-stock format by default. If you have specific format requirements, please note them when ordering.
Lead Time
Delivery times vary based on purchasing method and location. Consult your local distributor for specific delivery times. All proteins are shipped with standard blue ice packs. Contact us in advance for dry ice shipping (additional fees apply).
Notes
Avoid repeated freezing and thawing. Working aliquots can be stored at 4°C for up to one week.
Reconstitution
Briefly centrifuge the vial before opening. Reconstitute protein in sterile deionized water to 0.1-1.0 mg/mL. Add 5-50% glycerol (final concentration) and aliquot for long-term storage at -20°C/-80°C. Our default final glycerol concentration is 50%.
Shelf Life
Shelf life depends on storage conditions, buffer components, storage temperature, and protein stability. Liquid form is generally stable for 6 months at -20°C/-80°C. Lyophilized form is generally stable for 12 months at -20°C/-80°C.
Storage Condition
Store at -20°C/-80°C upon receipt. Aliquot for multiple uses. Avoid repeated freeze-thaw cycles.
Tag Info
Tag type is determined during manufacturing. If you have a specific tag type requirement, please inform us, and we will prioritize development of that tag.
Synonyms
mdh; MUL_4504Malate dehydrogenase; EC 1.1.1.37
Buffer Before Lyophilization
Tris/PBS-based buffer, 6% Trehalose.
Datasheet
Please contact us to get it.
Expression Region
1-329
Protein Length
full length protein
Purity
>85% (SDS-PAGE)
Species
Mycobacterium ulcerans (strain Agy99)
Target Names
mdh
Target Protein Sequence
MSASPLKVAV TGAAGQIGYS LLFRLASGSL LGPDRPIELR LLEIEPALKA LEGVVMELDD CAFPLLSGVE IGSDANKIFD GANLALLVGA RPRGPGMERS DLLEANGAIF TAQGKALNEV AADDIRVGVT GNPANTNALI AMTNAPDIPR ERFSALTRLD HNRAISQLAA KTGVAVTDIK KMTIWGNHSA TQYPDLFHAE VKGKNAAEVV NDQAWIEEYF IPTVAKRGAT IIDARGASSA ASAASASVDA ARSWLLGTPA DDWVSMAVLS DGSYGVPEGL ISSFPVTTKD GNWSIVKGLE IDEFSRGRID KTAAELADER KAVTELGLI
Uniprot No.

Target Background

Function
Catalyzes the reversible oxidation of malate to oxaloacetate.
Database Links

KEGG: mul:MUL_4504

Protein Families
LDH/MDH superfamily, MDH type 2 family

Q&A

What is Mycobacterium ulcerans Malate Dehydrogenase and what is its role in bacterial metabolism?

Mycobacterium ulcerans malate dehydrogenase (MDH) is a critical metabolic enzyme that catalyzes the reversible conversion of malate to oxaloacetate using NAD+ as a cofactor. This reaction is essential for several metabolic pathways, including the tricarboxylic acid (TCA) cycle, gluconeogenesis, and various anaplerotic reactions.

MDH plays multiple important roles in M. ulcerans metabolism:

  • Energy production through the TCA cycle, essential for ATP generation

  • Maintenance of redox balance by regenerating NAD+ from NADH

  • Carbon flux regulation between different metabolic pathways

  • Potential involvement in adaptation to environmental stresses encountered during infection

The enzyme likely contributes to M. ulcerans survival during transitions between environmental reservoirs and human hosts. Similar to other mycobacterial pathogens, M. ulcerans must adapt to changing environmental conditions, including oxygen limitation, nutrient fluctuations, and host defense mechanisms. MDH activity may be regulated in response to these environmental cues, allowing the bacterium to adjust its metabolism accordingly.

Biofilm formation, which has been identified as an important aspect of M. ulcerans pathogenesis, likely involves significant metabolic remodeling where MDH could play a key role . Understanding MDH function could provide insights into bacterial persistence mechanisms relevant to Buruli ulcer pathogenesis.

What expression systems are most effective for producing recombinant M. ulcerans MDH?

Several expression systems have been successfully employed for recombinant mycobacterial proteins, though specific optimization may be required for M. ulcerans MDH:

coli-based expression systems:

  • pET vector systems with BL21(DE3) hosts represent the most common approach

  • Cold-shock expression systems (pCold vectors) have proven effective for other M. ulcerans proteins, as demonstrated with M. ulcerans DNA gyrase subunits

  • Codon optimization may be necessary due to the GC-rich nature of mycobacterial genes

Mycobacterial expression hosts:

  • M. smegmatis expression systems provide a more native environment for mycobacterial protein folding

  • BCG expression systems have been used for M. ulcerans antigens, as seen with Ag85A expression

  • These systems may better maintain native conformation and post-translational modifications

For optimal expression of soluble, active M. ulcerans MDH, key considerations include:

  • Induction conditions: Lower temperatures (16-25°C) often improve solubility of recombinant mycobacterial proteins

  • Media composition: Rich media supplemented with glycerol and appropriate antibiotics

  • Fusion tags: N-terminal hexahistidine tags have proven effective for M. ulcerans proteins

  • Extraction buffers: Inclusion of protease inhibitors, reducing agents, and potentially glycerol for stability

The purification strategy typically involves immobilized metal affinity chromatography (IMAC) followed by at least one additional chromatographic step. For M. ulcerans DNA gyrase, "expressed WT/mutant GyrA and GyrB subunits were purified to homogeneity using a two-step column chromatographic procedure" resulting in preparations of ">95% purity in milligram amounts" .

How should researchers optimize recombinant M. ulcerans MDH activity assays?

Developing robust activity assays for recombinant M. ulcerans MDH requires careful optimization of multiple parameters:

Spectrophotometric assay considerations:

  • MDH activity can be monitored by tracking NAD+/NADH conversion at 340 nm

  • Forward reaction: malate → oxaloacetate (with NAD+ reduction)

  • Reverse reaction: oxaloacetate → malate (with NADH oxidation)

  • The reverse reaction is often preferred due to favorable equilibrium

Buffer optimization:

  • pH range: Typically 7.0-8.5, with systematic testing to determine optimum

  • Common buffers: Tris-HCl, phosphate, or HEPES (50-100 mM)

  • Salt concentration: Usually 50-150 mM NaCl or KCl

  • Cofactor concentration: Typically 0.1-0.5 mM NAD+/NADH

  • Substrate concentration: Range spanning Km (often 0.1-10 mM)

Quality control parameters:

  • Linearity with respect to enzyme concentration and time

  • Reproducibility across technical and biological replicates

  • Controls for non-enzymatic background reactions

  • Stability monitoring during storage conditions

Advanced assay formats:

  • Coupled enzyme assays for signal amplification

  • Fluorescence-based detection for increased sensitivity

  • High-throughput adaptations in microplate format

The table below summarizes key assay parameters that should be systematically optimized:

ParameterTypical RangeOptimization ApproachQuality Control Metric
pH7.0-8.50.5 unit incrementsActivity vs. pH curve
Temperature25-37°C5°C incrementsTemperature stability profile
Substrate concentration0.1-10 mM2-fold serial dilutionsMichaelis-Menten kinetics
Cofactor concentration0.1-1.0 mM2-fold serial dilutionsSaturation curve
Enzyme concentration1-100 nMLinear dilution seriesLinearity of rate vs. [E]
Ionic strength50-300 mM50 mM incrementsActivity vs. salt curve

Similar to approaches used for M. ulcerans DNA gyrase characterization, researchers should determine key enzymatic parameters (Km, kcat, optimal temperature range) under standardized conditions .

What structural and functional differences exist between M. ulcerans MDH and human MDH?

Understanding the structural and functional differences between M. ulcerans MDH and human MDH isoforms is essential for developing selective inhibitors and understanding host-pathogen interactions:

Structural differences:

  • Bacterial MDHs typically form dimers, while eukaryotic MDHs exist as both dimeric (cytosolic) and tetrameric (mitochondrial) forms

  • M. ulcerans MDH likely features the characteristic Rossmann fold for NAD+ binding

  • Substrate binding pocket architecture shows subtle but significant differences from human counterparts

  • Surface charge distribution and loop regions often differ substantially between bacterial and human enzymes

Functional differences:

  • Cofactor specificity: Most bacterial MDHs are strictly NAD+-dependent, while some human MDHs can utilize NADP+

  • pH optimum: Bacterial MDHs typically function optimally at slightly more alkaline pH

  • Allosteric regulation: Human and bacterial MDHs respond to different metabolic signals

  • Inhibitor sensitivity profiles differ substantially between bacterial and human enzymes

Evolutionary considerations:

  • M. ulcerans has undergone significant genome reduction during evolution from M. marinum

  • Core metabolic enzymes like MDH are typically retained despite reductive evolution, suggesting essential functions

  • M. ulcerans has adapted to specific environmental niches, potentially reflected in MDH properties

Research approaches to characterize these differences include:

  • Homology modeling and molecular dynamics simulations

  • Comparative biochemical characterization

  • Differential scanning fluorimetry to compare stability profiles

  • Inhibitor screening against both enzymes to identify selective compounds

These structural and functional differences can be exploited for therapeutic development, targeting M. ulcerans MDH without affecting human MDH function.

How does recombinant M. ulcerans MDH activity change under different environmental conditions?

M. ulcerans encounters diverse environmental conditions during its lifecycle, from aquatic environments to human tissue. Understanding how MDH activity responds to these conditions provides insights into metabolic adaptation mechanisms:

Temperature effects:

M. ulcerans thrives at temperatures between 28-33°C in the environment but must adapt to human body temperature (37°C) during infection. Research on other M. ulcerans enzymes indicates significant temperature-dependent activity profiles, such as observed with M. ulcerans DNA gyrase, where "the optimum temperature was 30 to 37°C and its activity decreased at 40°C" .

Oxygen availability:

  • Aerobic conditions: MDH functions primarily in the TCA cycle

  • Microaerobic/anaerobic conditions: MDH may participate in alternative pathways

  • Biofilm environments: Oxygen gradients within biofilms may create heterogeneous MDH activity

pH sensitivity:

Environmental and infection-site pH variations affect MDH catalytic efficiency:

  • Environmental waters: pH typically ranges from 6.5-8.0

  • Macrophage phagosomes: Acidic environment (pH 4.5-6.0)

  • Necrotic lesions: Variable pH depending on stage of disease

Growth phase considerations:

MDH activity profiles differ between:

  • Early exponential phase: Rapid growth requires high TCA cycle activity

  • Stationary phase: Metabolic remodeling for persistence

  • Biofilm growth: "M. ulcerans biofilm on transmissibility to ecological niches and Buruli ulcer pathogenesis" represents a distinct physiological state

The table below presents hypothetical activity parameters under different conditions based on studies of related mycobacterial enzymes:

ConditionRelative Activity (%)Km for Malate (mM)Vmax (μmol/min/mg)Stability (t1/2, h)
28°C, pH 7.0701.2 ± 0.245 ± 524 ± 3
37°C, pH 7.01000.8 ± 0.165 ± 712 ± 2
28°C, pH 6.0451.8 ± 0.330 ± 418 ± 3
37°C, pH 6.0601.5 ± 0.240 ± 58 ± 1
Biofilm conditions502.2 ± 0.425 ± 336 ± 5

Understanding these environmental adaptations is crucial for developing inhibitors effective under relevant physiological conditions and for comprehending the role of MDH in M. ulcerans persistence.

What role does M. ulcerans MDH play in biofilm formation and pathogenesis?

M. ulcerans forms biofilms that significantly contribute to its pathogenesis and environmental persistence. While MDH has not been specifically studied in this context, its potential role in biofilm formation and disease progression can be inferred:

MDH in biofilm metabolism:

Biofilm formation requires metabolic adaptation, with central carbon metabolism enzymes like MDH playing key roles:

  • Energy provision for extracellular matrix production

  • Maintenance of redox balance in the biofilm microenvironment

  • Adaptation to nutrient gradients within biofilm structures

Research has shown that "biofilm changes confer selective advantages to M. ulcerans in colonizing various ecological niches successfully, with repercussions for Buruli ulcer pathogenesis" . The extracellular matrix (ECM) "confers to the mycobacterium increased resistance to antimicrobial agents, and enhances colonization of insect vectors and mammalian hosts" .

MDH in virulence and pathogenesis:

Several mechanisms link MDH to virulence:

  • Metabolic adaptation during host colonization

  • Potential interaction with mycolactone production pathways

  • Mycolactone is "the key virulence factor" of M. ulcerans

  • Contribution to stress resistance during infection

MDH in environmental persistence:

M. ulcerans survives in various ecological niches where MDH may contribute to:

  • Adaptation to changing carbon sources in aquatic environments

  • Survival under nutrient limitation in environmental reservoirs

  • Metabolic flexibility during transitions between vectors, environment, and hosts

Experimental approaches to study MDH in biofilms:

  • Comparative transcriptomics/proteomics of planktonic vs. biofilm bacteria

  • Metabolic flux analysis using stable isotope labeling

  • Genetic manipulation (knockdown/overexpression) to assess impact on biofilm formation

  • Chemical inhibition studies targeting MDH in biofilm models

Understanding the role of MDH in biofilm formation could reveal new targets for disrupting M. ulcerans environmental persistence and pathogenesis, potentially leading to novel therapeutic strategies for Buruli ulcer.

How can recombinant M. ulcerans MDH be used for drug discovery?

Recombinant M. ulcerans MDH represents a valuable target for drug discovery efforts aimed at developing new treatments for Buruli ulcer. A systematic approach to MDH-focused drug discovery includes:

Assay development for inhibitor screening:

  • Primary screening assays:

    • Spectrophotometric assays monitoring NAD+/NADH conversion

    • Fluorescence-based activity assays for higher sensitivity

    • Thermal shift assays to detect ligand binding

  • Secondary validation assays:

    • Enzyme kinetic studies to determine inhibition mechanisms

    • Surface plasmon resonance for direct binding measurements

    • Isothermal titration calorimetry for thermodynamic characterization

Compound library selection:

  • Focused libraries of known dehydrogenase inhibitors

  • Natural product collections, particularly from sources in Buruli ulcer-endemic regions

  • Fragment libraries for structure-based drug design

  • Repurposing screens of approved drugs and clinical candidates

Hit-to-lead optimization workflow:

  • Primary hit identification using the methods described above

  • Dose-response characterization to determine IC50 values, similar to the approach used for fluoroquinolones against M. ulcerans DNA gyrase

  • Structure-activity relationship studies

  • Medicinal chemistry optimization for potency, selectivity, and drug-like properties

The table below summarizes common metrics used in M. ulcerans MDH inhibitor evaluation:

ParameterDefinitionTypical MethodThreshold for Progression
IC50Concentration causing 50% inhibitionDose-response curves<10 μM
KiInhibition constantEnzyme kinetic studies<1 μM
Selectivity IndexRatio of human MDH IC50 to M. ulcerans MDH IC50Comparative inhibition studies>10
MIC against M. ulceransMinimum inhibitory concentrationBacterial growth inhibition<10 μg/mL
Cytotoxicity (CC50)Concentration causing 50% cytotoxicityMammalian cell viability assays>100 μM

Similar approaches have been successfully applied to other M. ulcerans enzyme targets, as described for DNA gyrase inhibition by fluoroquinolones, where researchers determined "IC50, half of maximal inhibitory concentrations" and "CC25, FQ concentrations required to induce 25% of DNA cleavage" .

What methods are available for crystallizing recombinant M. ulcerans MDH?

Determining the three-dimensional structure of M. ulcerans MDH through X-ray crystallography requires successful protein crystallization, which presents several challenges:

Protein preparation considerations:

  • Expression optimization:

    • High-level expression systems (typically E. coli with pET vectors)

    • Culture conditions favoring soluble protein production

    • Temperature reduction during expression (16-20°C)

  • Purification strategy:

    • Multi-step chromatography to achieve >95% purity

    • Tag removal considerations (TEV or thrombin cleavage)

    • Similar to M. ulcerans DNA gyrase purification: "All recombinant DNA gyrase subunits were obtained at high purity (>95%) in milligram amounts"

  • Quality control:

    • Dynamic light scattering to assess monodispersity

    • Thermal stability analysis

    • Activity verification of purified protein

Crystallization approaches:

  • Initial screening:

    • Commercial sparse matrix screens (typically 500-1000 conditions)

    • Vapor diffusion methods (hanging drop and sitting drop)

    • Batch crystallization alternatives

  • Optimization strategies:

    • Fine grid screens around promising conditions

    • Additive screens to improve crystal quality

    • Seeding techniques to control nucleation

  • Co-crystallization strategies:

    • With NAD+/NADH cofactors

    • With substrate/product analogs

    • With identified inhibitors

Protein engineering for crystallization:

  • Surface entropy reduction:

    • Mutation of surface-exposed flexible residues (typically Lys, Glu)

    • N- or C-terminal truncations to remove disordered regions

  • Fusion protein approaches:

    • T4 lysozyme fusions to provide crystal contacts

    • Nanobody co-crystallization

  • Crystallization chaperones:

    • Antibody fragment (Fab, scFv) co-crystallization

    • Designed ankyrin repeat proteins (DARPins)

The table below outlines a typical crystallization optimization workflow:

StageMethodsVariablesSuccess Criteria
Initial screeningSparse matrix screens, 18°C500-1000 conditionsAny crystal hits
Grid optimizationFine screens around hitspH (±0.5), precipitant (±5%)Larger, single crystals
Additive screening96 additive conditionsSmall molecules, ions, detergentsImproved morphology
SeedingMicroseed matrix screeningSeed stock dilutionsSingle, diffracting crystals
CryoprotectionGlycerol, PEG, sucrose screeningCryoprotectant type and concentrationNo ice rings, preserved diffraction

Successful crystallization enables structure determination, which provides insights into MDH function and facilitates structure-based drug design targeting M. ulcerans.

How can researchers differentiate between M. ulcerans MDH and host MDH activity in infection models?

Distinguishing bacterial MDH activity from host MDH activity presents significant challenges in infection models but is crucial for understanding M. ulcerans metabolism during pathogenesis:

Biochemical differentiation strategies:

  • Enzyme kinetic discrimination:

    • Substrate specificity differences (alternative substrates)

    • Differential inhibitor sensitivity profiles

    • Cofactor preference variations (if any)

  • Assay condition optimization:

    • pH optima differences (bacterial vs. host enzymes)

    • Temperature sensitivity variations

    • Selective inhibition of host MDH during assays

Molecular and immunological approaches:

  • Genetic tagging strategies:

    • Expression of epitope-tagged MDH in M. ulcerans

    • Fluorescent protein fusions for microscopy studies

    • Affinity tags for selective purification

  • Antibody-based methods:

    • Development of antibodies specific to M. ulcerans MDH epitopes

    • Immunoprecipitation to isolate bacterial enzyme

    • Immunohistochemistry for localization within infected tissues

Advanced analytical techniques:

  • Mass spectrometry approaches:

    • Selected reaction monitoring (SRM) of unique peptides

    • Activity-based protein profiling

    • Stable isotope labeling to track bacterial metabolism

  • Genetic approaches in model systems:

    • Gene knockout/knockdown studies

    • Complementation with modified MDH variants

    • Reporter systems linked to MDH expression

The table below summarizes key differences that can be exploited to differentiate M. ulcerans MDH from host MDH:

ParameterM. ulcerans MDHMammalian MDHExploitation Strategy
Molecular Weight~35 kDa~36 kDa (cytosolic), ~35 kDa (mitochondrial)Western blot with specific antibodies
Optimal pH7.5-8.0 (predicted)7.0-7.5 (cytosolic), 7.5-8.0 (mitochondrial)Activity assays at discriminating pH
Thermal StabilityModerately thermostableLess thermostableHeat treatment before assay
Inhibitor SensitivityUnique sensitivity profileDifferent inhibition patternSelective inhibition approach
Sequence-Specific PeptidesUnique tryptic peptidesDistinct tryptic peptidesMS/MS identification

These approaches enable researchers to study M. ulcerans MDH activity during infection, providing insights into bacterial metabolism in the host environment and facilitating the development of targeted interventions.

Experimental design considerations:

  • Replication requirements:

    • Minimum triplicate measurements for each condition

    • Independent protein preparations for biological replicates

    • Power analysis to determine appropriate sample sizes

  • Controls and standards:

    • Positive and negative controls for each assay

    • Standard enzyme preparations for inter-experimental normalization

    • Vehicle controls for inhibitor studies

  • Randomization and blinding:

    • Randomized plate layouts to minimize positional effects

    • Blinded analysis where possible

Basic kinetic parameter analysis:

  • Michaelis-Menten kinetics:

    • Non-linear regression for Km and Vmax determination

    • Reporting with 95% confidence intervals

    • Residual analysis to verify model fit

  • Inhibition studies:

    • IC50 determination through dose-response curves

    • Similar to approaches used for M. ulcerans DNA gyrase: "IC50, half of maximal inhibitory concentrations"

    • Inhibition mechanism determination (competitive, non-competitive, uncompetitive)

  • Environmental variable effects:

    • ANOVA for comparing activity across conditions

    • Post-hoc tests with appropriate multiple comparison corrections

    • Regression analysis for continuous variables (pH, temperature)

Advanced statistical approaches:

  • Global fitting methods:

    • Simultaneous analysis of multiple datasets

    • Shared parameter constraints across experiments

    • More robust parameter estimation

  • Model discrimination:

    • Akaike Information Criterion (AIC) for comparing competing models

    • F-test for nested models

    • Cross-validation approaches

  • Outlier handling:

    • Objective criteria for outlier identification

    • Grubbs' test or Dixon's Q-test

    • Transparent reporting of any data exclusion

The table below outlines appropriate statistical tests for common MDH research questions:

Research QuestionAppropriate Statistical ApproachSoftware ToolsKey Metrics to Report
Kinetic parameter determinationNon-linear regression (Michaelis-Menten)GraphPad Prism, RKm, Vmax with 95% CI
Inhibitor comparisonDose-response curves, IC50 determinationGraphPad Prism, SigmaPlotIC50 with 95% CI
pH/temperature optimaNon-linear regression with Gaussian modelsGraphPad Prism, ROptimal value with 95% CI
Effect of mutationst-test or ANOVA with post-hoc testsSPSS, Rp-values with multiple testing correction
Inhibition mechanismGlobal fitting of multiple substrate concentrationsDynaFit, COPASIKi values, model discrimination metrics

How does M. ulcerans MDH compare to MDH from other mycobacterial species?

Comparative analysis of MDH across mycobacterial species provides evolutionary insights and may reveal M. ulcerans-specific adaptations relevant to its unique ecological niche and pathogenesis:

Sequence and structural comparisons:

  • M. ulcerans has undergone significant genome reduction during evolution from M. marinum

  • Core metabolic enzymes like MDH are typically conserved despite this reduction

  • Subtle amino acid differences may exist in substrate binding sites and regulatory regions

  • Conservation of catalytic residues across mycobacterial MDHs

Functional comparisons:

  • Kinetic parameters may differ between species, reflecting metabolic adaptations

  • Regulation of expression and activity might vary according to lifestyle differences

  • M. tuberculosis MDH has been more extensively characterized and serves as a reference

  • Environmental mycobacteria may show different MDH properties compared to pathogenic species

Evolutionary considerations:

  • M. ulcerans evolved from M. marinum through horizontal gene transfer and reductive evolution

  • Acquisition of the virulence plasmid pMUM, enabling mycolactone production

  • Mycolactone is "the key virulence factor" of M. ulcerans

  • Potential co-evolution of central metabolism (including MDH) with pathogenicity factors

Methodological approaches for comparative studies:

  • Recombinant expression of MDH from multiple species:

    • Same expression system and conditions

    • Identical purification protocols

    • Standardized activity assays

  • Comparative biochemical characterization:

    • Side-by-side kinetic analysis

    • Thermal stability comparisons

    • Inhibitor sensitivity profiling

  • Structural biology approaches:

    • Homology modeling based on available mycobacterial structures

    • Comparative crystallography if structures are available

    • Molecular dynamics simulations to identify functional differences

The table below compares predicted properties of MDH across selected mycobacterial species:

SpeciesNatural HabitatPathogenicityPredicted MDH CharacteristicsEvolutionary Relationship to M. ulcerans
M. ulceransAquatic, human hostBuruli ulcerAdaptation to environmental transitionsReference species
M. marinumAquatic, fish, humanFish TB, granulomasSimilar to M. ulcerans, potential differences in regulationDirect ancestor
M. tuberculosisHuman hostTuberculosisWell-characterized, adaptation to intracellular lifestyleDistant relation
M. lepraeHuman hostLeprosyReduced genome, potential gene decayDistant relation
M. smegmatisEnvironmentalNon-pathogenicAdaptation to diverse environmental conditionsDistant relation

Understanding these comparative aspects helps contextualize M. ulcerans MDH within mycobacterial evolution and may reveal adaptations specific to Buruli ulcer pathogenesis.

What role could M. ulcerans MDH play in vaccine development strategies?

While MDH itself has not been widely explored as a vaccine antigen for Buruli ulcer, its potential in this context warrants investigation:

MDH as a potential vaccine antigen:

  • Advantages:

    • Highly conserved among M. ulcerans strains

    • Essential metabolic enzyme (likely cannot be mutated without fitness cost)

    • Potentially expressed throughout infection stages

    • Could generate cross-protection against multiple mycobacterial pathogens

  • Challenges:

    • Homology with host MDH may limit immunogenicity or raise autoimmunity concerns

    • May not be sufficiently exposed to immune system

    • Immune responses may not effectively neutralize bacterial function

  • Precedents:

    • Other metabolic enzymes have shown promise as vaccine antigens in tuberculosis research

    • Ag85A, another M. ulcerans antigen, has shown protective effects in mouse models

Integration with current vaccine approaches:

M. ulcerans vaccine development has explored several strategies, including:

  • BCG vaccination provides limited protection

  • "Mice vaccinated with a single subcutaneous dose of BCG MU-Ag85A or prime-boost displayed significantly enhanced survival, reduced tissue pathology, and lower bacterial load compared to mice vaccinated with BCG"

  • MDH could potentially be incorporated into similar recombinant approaches

Research approaches to evaluate MDH vaccine potential:

  • Antigen discovery:

    • Epitope mapping to identify immunogenic regions

    • In silico prediction of MHC-binding peptides

    • Testing recombinant MDH for immunogenicity

  • Delivery strategies:

    • Recombinant protein formulations with adjuvants

    • DNA vaccines encoding MDH

    • Recombinant BCG or M. smegmatis expressing M. ulcerans MDH, similar to the approach used with Ag85A

  • Immune response characterization:

    • T-cell response profiling (CD4+, CD8+)

    • Cytokine production patterns

    • Antibody titer measurement

    • Similar to assessments where "rBCG MU-Ag85A followed by an M. smegmatis MU-Ag85A boost strongly induced murine antigen-specific CD4+ T cells and elicited functional IFNγ-producing splenocytes"

The integration of MDH into vaccine strategies could potentially enhance the efficacy of current candidates or provide a new avenue for exploration. Research in this area would benefit from the established M. ulcerans challenge models described in the literature, where "M. ulcerans JKD8049 as a suitable human challenge strain" has been identified .

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.