Recombinant Cyclopropane mycolic acid synthase MmaA2 (cmaC)

Shipped with Ice Packs
In Stock

Description

Introduction to Recombinant Cyclopropane Mycolic Acid Synthase MmaA2 (CmaC)

Recombinant Cyclopropane Mycolic Acid Synthase MmaA2, also known as CmaC, is an enzyme involved in the biosynthesis of mycolic acids in Mycobacterium tuberculosis . Mycolic acids are long-chain fatty acids that are major components of the mycobacterial cell wall, contributing to its unique structure and properties . These acids play crucial roles in the bacterium's virulence, drug resistance, and interaction with the host immune system .

Functional Role of MmaA2 (CmaC)

MmaA2 is a cyclopropane synthase that catalyzes the formation of cyclopropane rings on mycolic acids . Specifically, MmaA2 is involved in the distal cyclopropane modification of alpha-mycolates, a function initially attributed to another enzyme, CmaA1 . Research has demonstrated that MmaA2 is essential for this distal cyclopropane modification, while CmaA1 is not required for alpha-mycolate modification .

In addition to its role in modifying alpha-mycolates, MmaA2 also influences the cis-cyclopropanation of methoxymycolates . A loss of MmaA2 function leads to an accumulation of unsaturated methoxymycolate derivatives, indicating its importance in the cyclopropanation process .

Genetic and Biochemical Studies

The mmaA2 gene encodes the MmaA2 enzyme . Studies involving the creation and analysis of M. tuberculosis mmaA2 null mutants have been instrumental in elucidating the enzyme's function . These studies revealed that MmaA2 is a distal cyclopropane synthase that modifies both alpha- and oxygenated mycolates .

Further research has explored the redundancy between MmaA2 and another mycolic acid cyclopropane synthase, CmaA2 . Deletion of mmaA1 abolishes trans-cyclopropanation without the accumulation of trans-unsaturated oxygenated mycolates, which places MmaA1 in the biosynthetic pathway before CmaA2 . It has been found that CmaA2 can function as both a cis and trans cyclopropane synthase for oxygenated mycolates, highlighting a functional redundancy between MmaA2 and CmaA2 .

Impact on Mycobacterium tuberculosis Pathogenesis

Mycolic acid modification, including cyclopropanation, is crucial for the pathogenesis of M. tuberculosis . Strains lacking functional MmaA2 exhibit altered mycolic acid profiles, which affects the bacterium's virulence and interaction with the host immune system .

Research has demonstrated that a complete lack of cyclopropanation confers severe attenuation during the early stages of infection in a mouse model . While deletion of cmaA2 alone does not affect bacterial loads, it causes hypervirulence, suggesting that mycolic acid modification influences the severity and nature of the host immune response .

MmaA2 (CmaC) as a Drug Target

Given the importance of MmaA2 in mycolic acid biosynthesis and M. tuberculosis pathogenesis, it has been identified as a potential target for antimycobacterial drug development . Inhibiting MmaA2 could disrupt the mycobacterial cell wall, leading to bacterial death or reduced virulence .

Chemical Synthesis and Anti-Inflammatory Activity

(−)-Majusculoic acid, a marine natural product, and its analogs have been synthesized to evaluate their anti-inflammatory activity . These compounds were tested for nitric oxide (NO) production in lipopolysaccharide (LPS)-induced mouse macrophages RAW264.7 . This research provides a broader context for understanding the biological activities of cyclopropane-containing compounds, though not directly related to MmaA2 .

Product Specs

Form
Lyophilized powder
Lead Time
Delivery times vary depending on the purchase method and location. Please contact your local distributor for specific delivery timelines.
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 pellet 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%.
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
Store at -20°C/-80°C upon receipt. Aliquoting is recommended for multiple uses. Avoid repeated freeze-thaw cycles.
Tag Info
The tag type is determined during the manufacturing process. If you require a specific tag, please inform us, and we will prioritize its development.
Synonyms
cmaC; mmaA2; mmas-2; BQ2027_MB0663C; Cyclopropane mycolic acid synthase MmaA2; CMAS; EC 2.1.1.79; Cyclopropane-fatty-acyl-phospholipid synthase; CFA synthase; Mycolic acid methyltransferase; MA-MT; S-adenosylmethionine-dependent methyltransferase; AdoMet-MT; SAM-MT
Buffer Before Lyophilization
Tris/PBS-based buffer, 6% Trehalose.
Datasheet
Please contact us to get it.
Expression Region
1-287
Protein Length
full length protein
Purity
>85% (SDS-PAGE)
Species
Mycobacterium bovis (strain ATCC BAA-935 / AF2122/97)
Target Names
cmaC
Target Protein Sequence
MVNDLTPHFE DVQAHYDLSD DFFRLFLDPT QTYSCAHFER EDMTLEEAQI AKIDLALGKL GLQPGMTLLD IGCGWGATMR RAIAQYDVNV VGLTLSKNQA AHVQKSFDEM DTPLDRRVLL AGWEQFNEPV DRIVSIGAFE HFGHDRHADF FARAHKILPP DGVLLLHTIT GLTRQQMVDH GLPLTLWLAR FLKFIATEIF PGGQPPTIEM VEEQSAKTGF TLTRRQSLQP HYARTLDLWA EALQEHKSEA IAIQSEEVYE RYMKYLTGCA KLFRVGYIDV NQFTLAK
Uniprot No.

Target Background

Function
Recombinant Cyclopropane mycolic acid synthase MmaA2 (cmaC) catalyzes the conversion of a double bond to a cis cyclopropane ring at the distal position of an alpha mycolic acid. This reaction involves the transfer of a methylene group from S-adenosyl-L-methionine. MmaA2 also catalyzes the biosynthesis of cis-cyclopropanated methoxymycolates. Cyclopropanated mycolic acids are crucial for cell envelope permeability, host immunomodulation, and bacterial persistence.
Protein Families
CFA/CMAS family

Q&A

What is the primary function of MmaA2 in mycobacterial cell wall synthesis?

MmaA2 functions as a distal cyclopropane synthase that specifically modifies the alpha-mycolate in Mycobacterium tuberculosis. It is an S-adenosylmethionine (SAM)-dependent methyltransferase that introduces cyclopropane rings into the distal position of alpha-mycolic acids . This enzyme is unique among mycolic acid methyltransferases in that it can modify both alpha-mycolates and oxygenated mycolates (specifically methoxymycolates), making it the first identified cyclopropane synthase with dual modification capabilities . The cyclopropanation of mycolic acids plays a significant role in the pathogenicity of M. tuberculosis, affecting the structural integrity of the cell envelope and modulating host immune responses .

How does MmaA2 differ from other mycolic acid methyltransferases (MAMTs) in M. tuberculosis?

Despite substantial sequence identity with other mycolic acid cyclopropane synthases, MmaA2 catalyzes highly specific cyclopropane modifications. While PcaA and CmaA2 are responsible for proximal modification of alpha-mycolate and trans-cyclopropane modification respectively, MmaA2 specifically performs distal cyclopropane modification of alpha-mycolates .

Unlike other MAMTs that typically modify either alpha-mycolates or oxygenated mycolates, MmaA2 has the unique ability to modify both classes. It is essential for distal cyclopropanation of alpha-mycolates and also participates in cis-cyclopropanation of methoxymycolates . This contrasts with the initially proposed function of CmaA1, which was incorrectly attributed to alpha-mycolate modification, as demonstrated by studies showing unaffected alpha-mycolates in cmaA1 null mutants .

Why is the study of MmaA2 important in the context of tuberculosis research?

The study of MmaA2 and other mycolic acid modifying enzymes has significant implications for tuberculosis research for several reasons:

  • Mycolic acid modifications directly impact M. tuberculosis pathogenesis through effects on the inflammatory activity of trehalose dimycolate (cord factor) .

  • MAMTs as a class have been suggested to be essential for M. tuberculosis viability, making them potential targets for antimycobacterial drug development .

  • Understanding the specific roles of each MAMT helps elucidate the relationship between mycolic acid fine structure and the pathogen's ability to establish infection and evade host defenses .

  • MmaA2 specifically contributes to acid fastness and resistance to detergent stress, key characteristics that allow M. tuberculosis to survive in hostile environments .

  • Research has shown that the net effect of mycolate cyclopropanation is to dampen host immunity, making these modifications important virulence factors .

What is the biochemical pathway for mycolic acid biosynthesis involving MmaA2?

The biosynthesis of mycolic acids in M. tuberculosis follows a complex pathway involving multiple enzyme systems. The pathway proceeds as follows:

  • Initial fatty acid synthesis: Fatty acid synthetase I (FAS-I) produces C20-S-coenzyme A, which serves as the substrate for the fatty acid synthetase II (FAS-II) system .

  • Introduction of unsaturation: The FAS-IIA module introduces cis unsaturation at specific positions on the growing meroacid chain. This involves β-hydroxyacyl-ACP dehydrase and 2-trans-enoyl-ACP isomerase, which together produce cis unsaturated intermediates .

  • Chain elongation: The unsaturated intermediates undergo multiple cycles of elongation by FAS-II modules, with KasA involved in early elongation steps and KasB in later steps .

  • Cyclopropanation: MmaA2 acts at this stage to introduce cyclopropane rings into the distal positions of alpha-mycolates. For alpha-mycolate synthesis, MmaA2 introduces a cyclopropane ring at the distal unsaturation in cis,cis-Δ19,31-C50:2-S-ACP to help yield the mature C52-α-meroacid .

  • Proximal cyclopropanation: Another enzyme, PcaA, introduces cyclopropane rings into the proximal positions .

  • Processing into final products: The modified mycolic acids are then incorporated into cell wall components such as arabinogalactan-mycolate and trehalose dimycolate .

MmaA2 specifically catalyzes the transfer of a methyl group from S-adenosyl-L-methionine (SAM) to the double bond, forming the cyclopropane ring in the distal position of alpha-mycolates .

How do the structural features of cyclopropanated mycolic acids contribute to M. tuberculosis pathogenesis?

Cyclopropanated mycolic acids contribute to M. tuberculosis pathogenesis through several mechanisms:

  • Cell envelope integrity: Cyclopropanation affects the structural properties of the cell envelope, enhancing its impermeability and contributing to the characteristic acid-fast properties of M. tuberculosis .

  • Stress resistance: Mycolic acid modification confers resistance to various stresses, including detergent stress, which is important for bacterial survival in hostile environments such as within macrophages .

  • Immune modulation: The net effect of mycolate cyclopropanation is to dampen host immunity, allowing M. tuberculosis to establish infection. This immunomodulatory function is partly mediated through effects on the inflammatory activity of trehalose dimycolate (cord factor) .

  • Virulence timing: Studies with cyclopropane-deficient strains have shown that MAMTs are required at different stages of infection. Complete lack of cyclopropanation causes severe attenuation during the first week after aerosol infection in mouse models, while complete loss of MAMTs results in attenuation in the second week of infection .

  • Altered host recognition: Modifications to mycolic acids can change how pathogen-associated molecular patterns are recognized by host pattern recognition receptors, potentially allowing immune evasion .

What is the mechanism by which MmaA2 introduces cyclopropane rings into mycolic acids?

MmaA2 catalyzes the addition of cyclopropane rings through an S-adenosylmethionine (SAM)-dependent mechanism:

  • The enzyme binds both SAM as a methyl donor and the unsaturated mycolic acid substrate (typically attached to an acyl carrier protein) .

  • MmaA2 specifically recognizes the distal double bond in alpha-mycolates and certain oxygenated mycolates .

  • The enzyme facilitates the transfer of a methyl group from SAM to the carbon-carbon double bond in the mycolic acid .

  • This transfer is followed by the formation of a cyclopropane ring at the site of the former double bond .

  • The reaction produces S-adenosylhomocysteine (SAH) as a byproduct .

This mechanism requires precise positioning of the substrate within the enzyme active site to ensure regiospecific and stereospecific cyclopropanation. MmaA2's ability to modify both alpha-mycolates and methoxymycolates suggests it has a more flexible substrate binding site compared to other more specific MAMTs .

What methods have been used to create mmaA2 null mutants, and what are the key phenotypic changes observed?

The creation of mmaA2 null mutants has been accomplished through targeted gene disruption techniques:

  • Homologous recombination: Researchers have used allelic exchange methods where a disrupted copy of the mmaA2 gene (containing an antibiotic resistance marker) is introduced into M. tuberculosis. Through homologous recombination, the functional mmaA2 gene is replaced with the disrupted version .

  • Selection and verification: Transformants are selected on media containing appropriate antibiotics, and the gene disruption is verified using PCR and Southern blot analysis to confirm the absence of the functional mmaA2 gene .

Key phenotypic changes observed in mmaA2 null mutants include:

  • Altered mycolic acid profile: The most prominent change is the lack of distal cyclopropanation in alpha-mycolates. Analysis of the mycolic acid methyl esters (MAMEs) from the mmaA2 mutant shows the absence of the characteristic distal cyclopropane modification in alpha-mycolates .

  • Methoxymycolate changes: While fully cyclopropanated methoxymycolates are still produced in the mmaA2 mutant, cis-cyclopropanation is impaired, leading to the accumulation of unsaturated methoxymycolate derivatives .

  • Viability: Unlike what might be expected based on studies with MAMT inhibitors, M. tuberculosis remains viable without cyclopropanation, indicating that while important for virulence, cyclopropanation is not essential for bacterial survival under laboratory conditions .

  • Acid fastness reduction: Complete loss of cyclopropanation results in reduced acid fastness, a key diagnostic characteristic of mycobacteria .

  • Decreased detergent resistance: Mutants lacking MAMTs show increased susceptibility to detergent stress .

  • Attenuated virulence: Complete lack of cyclopropanation confers severe attenuation during the first week after aerosol infection in mouse models .

How can one distinguish between the functions of different mycolic acid methyltransferases when studying their roles in M. tuberculosis?

Distinguishing between the functions of different mycolic acid methyltransferases requires a multi-faceted approach:

  • Gene knockout studies: Creating individual null mutants for each methyltransferase gene allows researchers to directly observe the effects on mycolic acid composition. For example, studies with mmaA2 null mutants revealed its role in distal cyclopropanation of alpha-mycolates, while cmaA1 null mutants showed unaffected alpha-mycolates, disproving earlier attributions of alpha-mycolate modification to cmaA1 .

  • Comprehensive lipid analysis: Detailed analysis of mycolic acid methyl esters (MAMEs) using techniques such as thin-layer chromatography (TLC), high-performance liquid chromatography (HPLC), and mass spectrometry allows precise characterization of modifications in different mycolic acid subclasses .

  • Multiple mutant creation: Generating strains lacking multiple methyltransferases can reveal functional redundancies or synergistic effects. For example, strains lacking all functional MAMTs have been created to study the cumulative impact of cyclopropane deficiency .

  • Complementation studies: Reintroducing individual methyltransferase genes into multiple knockout strains can confirm specific functions and rule out polar effects of gene disruption .

  • Temporal expression analysis: Examining when different methyltransferases are expressed during infection can provide insights into their stage-specific roles .

  • In vitro enzyme assays: Purified recombinant enzymes can be tested with various substrates to determine their specific activities and substrate preferences .

  • Structural studies: X-ray crystallography of methyltransferases with bound substrates or substrate analogs can reveal the molecular basis for their specificity .

What are the technical challenges in expressing and purifying recombinant MmaA2 for biochemical studies?

The expression and purification of recombinant MmaA2 presents several technical challenges:

  • Membrane association: As an enzyme involved in mycolic acid modification, MmaA2 is likely to have hydrophobic domains that associate with the membrane or membrane-bound substrates, making it difficult to express in soluble form .

  • Cofactor requirements: SAM-dependent methyltransferases often require the presence of their cofactor for proper folding and stability. Ensuring adequate SAM availability during expression and purification can be challenging .

  • Substrate complexity: The natural substrates of MmaA2 are long-chain fatty acids attached to acyl carrier proteins, which are difficult to synthesize or isolate for in vitro studies .

  • Expression host selection: E. coli, the most common expression host, lacks many of the auxiliary factors present in mycobacteria that might be necessary for proper folding and function of MmaA2 .

  • Protein solubility: Overexpression often leads to inclusion body formation, necessitating refolding procedures that may not yield fully active enzyme .

  • Detergent compatibility: Purification may require detergents to solubilize the enzyme, but these must be compatible with downstream activity assays .

  • Enzyme stability: Maintaining enzyme stability during purification and storage can be challenging, especially if the enzyme requires specific lipid environments for activity .

  • Active site integrity: The active site may contain critical cysteine residues that are susceptible to oxidation during purification, potentially affecting enzyme activity .

To address these challenges, researchers typically employ strategies such as:

  • Using mycobacterial expression systems rather than E. coli

  • Addition of stabilizing agents during purification

  • Expression as fusion proteins with solubility-enhancing tags

  • Co-expression with mycobacterial chaperones

  • Development of membrane-mimetic systems for activity assays

How do the structural features of MmaA2 determine its specificity for distal cyclopropanation of alpha-mycolates?

The structural features that determine MmaA2's specificity for distal cyclopropanation of alpha-mycolates involve several key elements:

  • Substrate binding pocket architecture: Though detailed structural information specific to MmaA2 is limited in the provided search results, it can be inferred that MmaA2 possesses a unique binding pocket that accommodates the distal portion of alpha-mycolates. This pocket likely has dimensions and chemical properties that allow precise positioning of the distal double bond for cyclopropanation .

  • Dual substrate recognition: MmaA2's unique ability to modify both alpha-mycolates and methoxymycolates suggests it has a more adaptable substrate binding site compared to other more specific MAMTs. This flexibility enables it to recognize and position different mycolic acid precursors for cyclopropanation .

  • SAM binding domain: As an S-adenosylmethionine-dependent methyltransferase, MmaA2 contains a conserved SAM binding domain. The orientation of this domain relative to the substrate binding pocket likely determines the regiospecificity of the methyl transfer reaction .

  • Recognition elements for distal positioning: MmaA2 must contain structural elements that specifically recognize features of the distal portion of the meroacid chain, distinguishing it from the proximal region. This allows MmaA2 to target the distal double bond specifically, while enzymes like PcaA target the proximal position .

  • Active site architecture: The arrangement of catalytic residues in the active site likely facilitates the specific mechanism of cyclopropane ring formation at the distal position .

The specific structural features of MmaA2 that enable its function are of particular interest for drug design efforts targeting this enzyme class. Understanding the molecular basis for the specificity of different MAMTs could lead to the development of selective inhibitors with potential as anti-tuberculosis agents .

What is the comparative analysis of mycolic acid profiles in wild-type, mmaA2 mutant, and complemented strains?

Comparative analysis of mycolic acid profiles in wild-type, mmaA2 mutant, and complemented strains reveals specific alterations in mycolic acid structure:

Wild-type M. tuberculosis:

  • Complete cyclopropanation of alpha-mycolates at both proximal and distal positions

  • Fully cyclopropanated methoxymycolates

  • Normal proportions of alpha-, methoxy-, and keto-mycolates

  • Characteristic acid-fast properties

  • Normal resistance to detergent stress

mmaA2 mutant strain:

Complemented strain (mmaA2 mutant with reintroduced functional mmaA2):

  • Restoration of distal cyclopropanation in alpha-mycolates

  • Restoration of normal methoxymycolate cyclopropanation

  • Return to wild-type mycolic acid profile

  • Recovery of acid-fast properties

  • Restored detergent resistance

  • Virulence phenotype similar to wild-type

These comparative analyses confirm the specific role of MmaA2 in distal cyclopropanation of alpha-mycolates and its contribution to cis-cyclopropanation of methoxymycolates. The fact that complementation restores the wild-type phenotype confirms that the observed changes in the mutant are directly attributable to the loss of MmaA2 function rather than polar effects on adjacent genes .

How does the substrate specificity of MmaA2 compare with other mycolic acid methyltransferases?

The substrate specificity of MmaA2, compared to other mycolic acid methyltransferases (MAMTs), demonstrates both shared characteristics and unique features:

MAMTPrimary SubstrateModification TypePositionMycolic Acid Type
MmaA2cis,cis-Δ19,31-C50:2-S-ACPCyclopropanationDistalAlpha-mycolates
MmaA2Unsaturated methoxymycolatescis-CyclopropanationVariableMethoxymycolates
PcaAcis,cis-Δ19,31-C50:2-S-ACPCyclopropanationProximalAlpha-mycolates
CmaA2Unsaturated precursorstrans-CyclopropanationVariableVarious mycolates
MmaA4Unsaturated precursorsMethoxy introductionVariableMethoxymycolates
CmaA1Unknown (not alpha-mycolates)UnknownUnknownUnknown
MmaA1Methoxymycolate precursorsMethylationVariableMethoxymycolates
MmaA3Methoxymycolate precursorsMethyl introductionVariableMethoxymycolates

Key distinctions in substrate specificity:

  • Dual substrate capability: MmaA2 is unique among MAMTs in its ability to modify both alpha-mycolates and methoxymycolates. This dual substrate capability distinguishes it from other more specialized enzymes in the family .

  • Positional specificity: While MmaA2 targets the distal position of alpha-mycolates, PcaA specifically modifies the proximal position. This positional specificity is maintained despite substantial sequence identity between these enzymes .

  • Stereochemical control: Different MAMTs create either cis- or trans-cyclopropane rings. MmaA2 is specifically involved in cis-cyclopropanation, whereas enzymes like CmaA2 introduce trans-cyclopropane rings .

  • Substrate chain length: The specificity for certain chain lengths of meroacid precursors differs among MAMTs. MmaA2 acts on longer-chain precursors that have already undergone substantial elongation by the FAS-II system .

  • Functional overlap: Despite their specificities, there appears to be some functional overlap among MAMTs, as suggested by the finding that methoxymycolates can still be fully cyclopropanated in mmaA2 mutants, albeit with some impairment in cis-cyclopropanation .

The remarkable substrate specificity of these enzymes, despite their sequence similarity, makes them interesting subjects for structural and biochemical studies aimed at understanding the molecular determinants of substrate recognition and catalytic specificity .

What are the experimental approaches to study the impact of MmaA2-mediated mycolic acid modifications on host immune responses?

Studying the impact of MmaA2-mediated mycolic acid modifications on host immune responses requires multiple experimental approaches:

  • Infection studies with gene knockout strains:

    • Mouse infection models using wild-type, mmaA2 mutant, and complemented strains

    • Monitoring bacterial burden in lungs and other organs over time

    • Histopathological assessment of infected tissues

    • Survival studies to assess virulence

  • Immune cell response characterization:

    • Ex vivo analysis of cells from infected animals (flow cytometry, cell sorting)

    • In vitro infection of macrophages, dendritic cells, and other immune cells with wild-type vs. mutant strains

    • Measurement of phagocytosis, phagosome maturation, and bacterial killing

    • Assessment of antigen presentation and T cell activation

  • Cytokine and chemokine profiling:

    • Multiplex analysis of cytokine/chemokine production in infected tissues

    • Real-time PCR for cytokine gene expression

    • ELISA for protein levels in tissue homogenates and cell culture supernatants

    • Intracellular cytokine staining to identify cellular sources

  • Isolation and testing of purified cell wall components:

    • Extraction and purification of trehalose dimycolate and other mycolic acid-containing molecules from wild-type and mutant strains

    • Testing the inflammatory activity of these components on cultured cells

    • In vivo administration to assess inflammatory potential

    • Structure-function studies correlating molecular structure with immunostimulatory activity

  • Transcriptomic and proteomic analyses:

    • RNA-seq of infected tissues or cells to assess global host response patterns

    • Proteomics to identify differentially expressed proteins

    • Pathway analysis to identify key immunological processes affected by mycolic acid modifications

  • Imaging approaches:

    • Confocal microscopy to visualize bacterial-host cell interactions

    • Electron microscopy to examine changes in bacterial cell envelope and host cell ultrastructure

    • Live cell imaging to track dynamic interactions in real-time

These approaches have revealed that the net effect of mycolate cyclopropanation is to dampen host immunity, suggesting that mycolic acid modifications serve as immunomodulatory virulence factors that help M. tuberculosis establish persistent infection .

What analytical methods are most effective for characterizing the structural changes in mycolic acids resulting from MmaA2 mutation?

Several analytical methods are particularly effective for characterizing structural changes in mycolic acids resulting from MmaA2 mutation:

  • Thin-Layer Chromatography (TLC):

    • Allows separation of different mycolic acid classes (alpha-, methoxy-, keto-mycolates)

    • Can detect changes in mobility resulting from altered cyclopropanation

    • Useful for rapid screening of mycolic acid profiles

    • Can be coupled with autoradiography when using radiolabeled precursors

  • High-Performance Liquid Chromatography (HPLC):

    • Provides superior resolution compared to TLC

    • Can separate closely related mycolic acid species

    • Enables quantitative analysis of different mycolic acid types

    • Can be coupled with various detection methods (UV, fluorescence, charged aerosol detection)

  • Gas Chromatography-Mass Spectrometry (GC-MS):

    • Particularly useful for analyzing the methyl esters of mycolic acids (MAMEs)

    • Provides structural information through fragmentation patterns

    • Can identify positional isomers and distinguish between cis and trans configurations

    • Enables accurate mass determination for formula confirmation

  • Liquid Chromatography-Mass Spectrometry (LC-MS):

    • Allows analysis of intact mycolic acids without derivatization

    • Provides detailed structural information through MS/MS fragmentation

    • Can detect subtle modifications in the mycolic acid structure

    • Enables quantification of different mycolic acid species

  • Nuclear Magnetic Resonance (NMR) Spectroscopy:

    • Provides detailed structural information, including the presence and position of cyclopropane rings

    • Can distinguish between cis and trans cyclopropane configurations

    • Requires larger sample amounts but gives unambiguous structural data

    • Particularly valuable for confirming novel structures

  • Matrix-Assisted Laser Desorption/Ionization-Time of Flight (MALDI-TOF) MS:

    • Useful for analyzing complex mixtures of mycolic acids

    • Provides accurate molecular weight information

    • Can be used for rapid profiling of mycolic acid patterns

    • Less structural detail than MS/MS methods but faster analysis

  • Pyrolysis Mass Spectrometry:

    • Involves thermal degradation of mycolic acids

    • Generates characteristic fragments (meroaldehydes) that can be analyzed

    • Useful for distinguishing between different cyclopropanated structures

    • Can reveal distal vs. proximal modifications

The most comprehensive structural characterization typically employs a combination of these methods. For example, HPLC might be used for initial separation, followed by mass spectrometry for structural analysis and NMR for confirmation of specific structural features like cyclopropane ring positions .

How can recombinant MmaA2 be used to develop new anti-tuberculosis compounds?

Recombinant MmaA2 can serve as a valuable tool in the development of new anti-tuberculosis compounds through several strategic approaches:

  • High-throughput screening platforms:

    • Development of enzymatic assays using purified recombinant MmaA2

    • Screening of chemical libraries for compounds that inhibit MmaA2 activity

    • Measurement of SAM utilization or cyclopropane formation as readouts

    • Counter-screening against human methyltransferases to identify selective inhibitors

  • Structure-based drug design:

    • X-ray crystallography or cryo-EM studies of recombinant MmaA2 alone and in complex with substrates

    • Computational modeling of the active site and substrate binding pocket

    • Virtual screening of compound libraries against the MmaA2 structure

    • Rational design of inhibitors based on transition state mimics or substrate analogs

  • Fragment-based drug discovery:

    • Screening of fragment libraries against recombinant MmaA2

    • NMR or X-ray crystallography to identify fragment binding sites

    • Fragment growing, linking, or merging to develop more potent inhibitors

    • Optimization of pharmacokinetic properties

  • Mechanism-based inhibitor development:

    • Design of mechanism-based inhibitors that target the SAM binding site

    • Development of substrate analogs that bind irreversibly to the enzyme

    • Creation of bisubstrate analogs that bridge the SAM and mycolic acid binding sites

    • Exploration of allosteric inhibitors that affect enzyme conformation

  • Whole-cell assay development:

    • Creation of reporter strains that indicate MmaA2 inhibition

    • Correlation of whole-cell activity with enzymatic inhibition

    • Medicinal chemistry optimization for cell penetration

    • Assessment of effects on mycolic acid profiles as a readout

  • Target validation studies:

    • Comparison of inhibitor effects with genetic knockouts

    • Resistance studies to confirm on-target activity

    • Assessment of synergy with existing TB drugs

    • Evaluation of effects on pathogenesis in infection models

The attractiveness of MmaA2 as a drug target is supported by several factors:

  • MAMTs have been suggested to be essential for M. tuberculosis viability

  • Cyclopropane-deficient strains show severe attenuation in infection models

  • There are no human homologs of mycolic acid cyclopropane synthases

  • The enzyme class represents a novel mechanism of action compared to existing TB drugs

  • Inhibition could potentially reduce both bacterial viability and virulence

These approaches could contribute to the development of new anti-tuberculosis compounds with novel mechanisms of action, addressing the critical need for alternatives to combat drug-resistant tuberculosis .

What are common challenges in analyzing mycolic acid profiles, and how can they be addressed?

Analyzing mycolic acid profiles presents several challenges that researchers must overcome for accurate results:

  • Sample preparation complexities:

    • Challenge: Mycolic acids are covalently attached to cell wall components, making extraction difficult.

    • Solution: Use standardized protocols involving alkaline hydrolysis (saponification) followed by acidification and organic extraction. Consistent application of these methods ensures reproducible extraction efficiency .

  • Structural diversity and complexity:

    • Challenge: M. tuberculosis produces multiple types of mycolic acids with subtle structural variations.

    • Solution: Employ complementary analytical techniques such as TLC, HPLC, and mass spectrometry to comprehensively characterize the mycolic acid profile. Two-dimensional TLC can help separate closely related species .

  • Low abundance of specific species:

    • Challenge: Some mycolic acid species may be present in low abundance, making detection difficult.

    • Solution: Increase sensitivity through derivatization (e.g., p-bromophenacyl esters for HPLC) or use more sensitive detection methods like mass spectrometry with selected ion monitoring .

  • Overlapping chromatographic profiles:

    • Challenge: Different mycolic acid species may co-elute or have similar migration patterns.

    • Solution: Optimize chromatographic conditions (solvent systems, temperature, column selection) and consider using multiple separation techniques in sequence to resolve overlapping peaks .

  • Quantification challenges:

    • Challenge: Accurate quantification is difficult due to differential extraction, derivatization efficiency, and detector response.

    • Solution: Use internal standards with similar chemical properties, develop standard curves with authentic standards, and apply correction factors for known differences in detector response .

  • Distinguishing cyclopropanated from unsaturated species:

    • Challenge: Cyclopropanated and unsaturated mycolic acids have similar molecular weights and chromatographic properties.

    • Solution: Use specialized techniques such as silver ion chromatography, which separates compounds based on unsaturation, or tandem mass spectrometry to identify diagnostic fragment ions .

  • Artifactual modifications during analysis:

    • Challenge: Harsh extraction conditions or derivatization may introduce artifacts or modify cyclopropane rings.

    • Solution: Use milder extraction conditions when possible, control reaction temperatures carefully, and include control samples to identify potential artifacts .

  • Reproducibility across different growth conditions:

    • Challenge: Mycolic acid profiles can vary depending on growth conditions, making comparisons difficult.

    • Solution: Standardize growth conditions (medium, growth phase, temperature) and include appropriate wild-type controls grown under identical conditions for direct comparison .

By addressing these challenges methodically, researchers can obtain reliable and reproducible mycolic acid profiles that accurately reflect the impact of genetic manipulations or drug treatments on mycolic acid biosynthesis .

What factors should be considered when designing gene knockout experiments for MmaA2 and related genes?

When designing gene knockout experiments for MmaA2 and related genes, several critical factors should be considered to ensure valid and interpretable results:

  • Genetic context and potential polar effects:

    • Consideration: Disruption of mmaA2 might affect the expression of neighboring genes.

    • Approach: Design knockout constructs that minimize disruption of adjacent genes. Use complementation studies with the wild-type gene to confirm that phenotypes are specifically due to the target gene disruption .

  • Selection of appropriate knockout strategy:

    • Consideration: Different knockout strategies (deletion, insertion, point mutation) may have different effects.

    • Approach: Consider unmarked, in-frame deletions to avoid polar effects. For essential genes, conditional knockouts or point mutations affecting function may be necessary .

  • Potential functional redundancy:

    • Consideration: Other methyltransferases might partially compensate for MmaA2 function.

    • Approach: Consider creating double or multiple knockouts to address redundancy. For example, studying an mmaA2 knockout in combination with knockouts of other methyltransferases might reveal synergistic effects .

  • Verification of gene disruption:

    • Consideration: Confirming complete disruption of gene function is essential.

    • Approach: Use multiple methods to verify knockout: PCR, Southern blotting, RT-PCR to confirm absence of transcript, and ultimately, biochemical analysis to confirm the expected changes in mycolic acid profile .

  • Choice of parental strain:

    • Consideration: Different M. tuberculosis strains may have subtle differences in mycolic acid profiles.

    • Approach: Use well-characterized laboratory strains with established mycolic acid profiles. Consider creating knockouts in clinical isolates to assess strain-specific effects .

  • Growth conditions for phenotypic analysis:

    • Consideration: Phenotypes may only be apparent under specific growth conditions.

    • Approach: Test mutants under various conditions, including standard laboratory media, stress conditions (low pH, nutrient limitation, oxidative stress), and in vivo infection models .

  • Comprehensive phenotypic characterization:

    • Consideration: Mycolic acid modifications affect multiple bacterial properties.

    • Approach: Assess various phenotypes: growth rates, cell envelope permeability, resistance to antibiotics and detergents, acid fastness, biofilm formation, and virulence in infection models .

  • Temporal considerations in infection models:

    • Consideration: Effects of mycolic acid modifications may be stage-specific during infection.

    • Approach: Examine mutant phenotypes at multiple timepoints during infection, as different MAMTs have been shown to be important at different stages (e.g., early vs. late infection) .

  • Biochemical confirmation of enzymatic function:

    • Consideration: Gene knockout should result in specific biochemical changes.

    • Approach: Perform detailed analysis of mycolic acid profiles using TLC, HPLC, and mass spectrometry to confirm the specific modifications attributed to the disrupted enzyme .

  • Reversibility through complementation:

    • Consideration: True confirmation of gene function requires reversal of knockout phenotype.

    • Approach: Complement the mutant with the wild-type gene under control of its native promoter or an inducible promoter. This should restore the wild-type mycolic acid profile and associated phenotypes .

Careful consideration of these factors ensures that gene knockout experiments provide clear and interpretable information about the specific roles of MmaA2 and related enzymes in mycolic acid biosynthesis and M. tuberculosis pathogenesis .

How can researchers distinguish between direct effects of MmaA2 inhibition and secondary metabolic consequences when evaluating new inhibitory compounds?

Distinguishing between direct effects of MmaA2 inhibition and secondary metabolic consequences when evaluating potential inhibitory compounds is a critical challenge that requires multiple experimental approaches:

  • Enzymatic assays with purified recombinant protein:

    • Direct effect assessment: Measure inhibition of purified recombinant MmaA2 in vitro using biochemical assays.

    • Controls: Include other mycolic acid methyltransferases to assess specificity and non-related methyltransferases to rule out general SAM-dependent enzyme inhibition .

  • Dose-response relationships:

    • Direct effect assessment: Compare dose-response curves for enzymatic inhibition versus whole-cell effects.

    • Interpretation: Direct target effects typically show correlation between enzymatic IC50 and cellular EC50 values, accounting for factors like cell penetration .

  • Time-course analysis:

    • Direct effect assessment: Monitor changes in mycolic acid profiles over time after compound addition.

    • Interpretation: Direct inhibition of MmaA2 should lead to rapid changes in cyclopropanation patterns before secondary metabolic effects become apparent .

  • Targeted metabolomic analysis:

    • Direct effect assessment: Analyze specific changes in mycolic acid profiles using TLC, HPLC, and mass spectrometry.

    • Interpretation: Direct MmaA2 inhibition should produce a profile similar to mmaA2 knockout mutants, specifically affecting distal cyclopropanation of alpha-mycolates .

  • Genetic validation approaches:

    • MmaA2 overexpression: Test if overexpression of MmaA2 increases the MIC of the compound.

    • Resistant mutant analysis: Select for resistant mutants and sequence mmaA2 and related genes to identify target-based resistance mechanisms .

  • Comparative analysis with knockout strains:

    • Direct effect assessment: Compare phenotypic and biochemical effects of the compound with characterized mmaA2 knockout strains.

    • Interpretation: Similar profiles suggest on-target activity, while significant differences may indicate off-target or secondary effects .

  • Transcriptomic and proteomic profiling:

    • Global response assessment: Analyze changes in gene expression and protein levels in response to the compound.

    • Interpretation: Specific inhibition should affect pathways directly linked to mycolic acid biosynthesis, while broad stress responses suggest non-specific effects .

  • Chemical genetic approaches:

    • Direct effect validation: Test compound effects in strains with reduced MmaA2 expression.

    • Interpretation: Hypersensitivity in strains with reduced target levels supports on-target activity .

  • Structure-activity relationship (SAR) studies:

    • Direct effect correlation: Correlate structural modifications of the compound with changes in both enzymatic inhibition and cellular effects.

    • Interpretation: Parallel SAR between biochemical and cellular assays supports on-target activity .

  • In situ target engagement assays:

    • Direct binding demonstration: Use techniques like cellular thermal shift assays (CETSA) to demonstrate direct binding to MmaA2 in cells.

    • Interpretation: Thermal stabilization of the target protein in the presence of the compound provides evidence for direct interaction .

By systematically applying these approaches, researchers can build a strong case for whether observed effects are due to direct MmaA2 inhibition or secondary consequences, guiding further development of potential anti-tuberculosis compounds targeting this enzyme .

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.