Recombinant Coxiella burnetii (3R)-hydroxymyristoyl-[acyl-carrier-protein] dehydratase (fabZ)

Shipped with Ice Packs
In Stock

Product Specs

Form
Lyophilized powder
Note: While we prioritize shipping the format currently in stock, please specify your format preference during order placement for customized preparation.
Lead Time
Delivery times vary depending on the purchase method and location. Please consult your local distributor for precise delivery estimates.
Note: All proteins are shipped with standard 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. We recommend adding 5-50% glycerol (final concentration) and aliquoting for long-term storage at -20°C/-80°C. Our standard glycerol concentration is 50% and can serve as a guideline.
Shelf Life
Shelf life depends on storage conditions, buffer composition, temperature, and protein stability. Generally, liquid formulations have a 6-month shelf life at -20°C/-80°C, while lyophilized forms have a 12-month shelf life at -20°C/-80°C.
Storage Condition
Store at -20°C/-80°C upon receipt. Aliquot for multiple uses to prevent repeated freeze-thaw cycles.
Tag Info
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
fabZ; CBU_06143-hydroxyacyl-[acyl-carrier-protein] dehydratase FabZ; EC 4.2.1.59,; 3R)-hydroxymyristoyl-[acyl-carrier-protein] dehydratase,; 3R)-hydroxymyristoyl-ACP dehydrase; Beta-hydroxyacyl-ACP dehydratase
Buffer Before Lyophilization
Tris/PBS-based buffer, 6% Trehalose.
Datasheet
Please contact us to get it.
Expression Region
1-145
Protein Length
full length protein
Purity
>85% (SDS-PAGE)
Species
Coxiella burnetii (strain RSA 493 / Nine Mile phase I)
Target Names
fabZ
Target Protein Sequence
MNVMNITDIK KYIPHRYPFL LIDRVIKIEK DKSLVAIKNV TVNEPFFTGH FPVRPVMPGV LIIESLAQAA GILIVKSLNL PEGHKDIYFF AGVDNARFKR VVEPGDQLTL EVKVLKVHRG LWKFEGKATV DDQLACKAEL MTIKG
Uniprot No.

Target Background

Function

Involved in unsaturated fatty acid biosynthesis. This enzyme catalyzes the dehydration of short-chain β-hydroxyacyl-ACPs and long-chain saturated and unsaturated β-hydroxyacyl-ACPs.

Database Links
Protein Families
Thioester dehydratase family, FabZ subfamily
Subcellular Location
Cytoplasm.

Q&A

What is the biological function of FabZ in Coxiella burnetii?

FabZ in C. burnetii functions as a β-hydroxyacyl-acyl carrier protein dehydratase that catalyzes the dehydration of β-hydroxyacyl-ACP to trans-2-acyl-ACP in the fatty acid biosynthesis pathway (FASII) . This essential step involves the elimination of a water molecule through an enolate intermediate, resulting in the formation of a carbon-carbon double bond. The reaction specifically removes the hydroxyl group at the C-3 position and a hydrogen atom at the C-2 position to form the trans (or E) double bond . Unlike the homologous FabA enzyme, FabZ in C. burnetii does not perform isomerization after the dehydration step, focusing solely on producing straight-chain fatty acids essential for membrane phospholipid synthesis .

How does C. burnetii FabZ differ from FabZ in other bacterial species?

C. burnetii FabZ shares the conserved "hot dog fold" structure common to other bacterial dehydratases but displays several distinct features . Unlike canonical FabZ enzymes, C. burnetii FabZ contains unique amino acid substitutions in the substrate-binding tunnel, particularly a characteristic residue change that may affect substrate specificity and chain length preference . Comparative analyses with FabZ from E. coli and other bacteria reveal that C. burnetii FabZ may have evolved specialized characteristics to function within the acidic phagolysosomal environment where the bacterium replicates . These adaptations potentially contribute to C. burnetii's ability to maintain membrane integrity under harsh intracellular conditions, which is crucial for its pathogenicity .

Why is fatty acid biosynthesis important for C. burnetii's intracellular lifestyle?

Fatty acid biosynthesis is critical for C. burnetii's intracellular survival for several reasons:

  • Membrane Formation: C. burnetii requires newly synthesized fatty acids to maintain and expand its cell membrane during replication within the acidic parasitophorous vacuole .

  • Adaptation to Acidic Environment: The bacterium must maintain specific membrane properties to withstand the acidic pH (around 4.5) of its replicative niche .

  • Metabolic Independence: As an obligate intracellular pathogen, C. burnetii has evolved to synthesize essential fatty acids that may not be readily available from the host cell .

  • Developmental Cycle Support: C. burnetii transitions between small cell variant (SCV) and large cell variant (LCV) forms during its life cycle, a process requiring extensive membrane remodeling dependent on fatty acid biosynthesis .

The horizontally acquired genes for fatty acid synthesis, including fabZ, represent adaptive mechanisms that likely facilitated C. burnetii's evolution from a tick-associated ancestor to a mammalian pathogen .

What is the structural organization of C. burnetii FabZ and how does it relate to function?

C. burnetii FabZ adopts the characteristic "hot dog fold" structure consisting of a central α-helix surrounded by a curved β-sheet . The enzyme functions as a hexamer, specifically arranged as a trimer of dimers, where each monomer contains the catalytic machinery. Key structural features include:

Structural ElementCompositionFunctional Role
Catalytic SiteHis-Glu dyadCatalyzes proton abstraction and elimination of hydroxyl group
Substrate Binding TunnelHydrophobic residuesAccommodates fatty acyl chain
Active Site EntrancePositively charged residuesFacilitates interaction with negatively charged ACP
Hexameric Assembly3 dimersProvides structural stability and proper orientation of catalytic sites

Crystal structures reveal that each monomer contributes to the formation of the substrate-binding tunnel along the central α-helix, with the catalytic histidine positioned to abstract a proton from the C2 atom of the substrate . The unique amino acid variations in C. burnetii FabZ, particularly in the substrate-binding region, suggest adaptation to specific substrate preferences that may be linked to its intracellular lifestyle .

How does the ACP-FabZ protein-protein interaction occur during catalysis?

The interaction between acyl carrier protein (ACP) and FabZ involves a dynamic "seesaw-like" binding mechanism . This process follows several steps:

  • Initial Recognition: Electrostatic interactions between positively charged residues on FabZ and the negatively charged helix II of ACP guide the initial docking.

  • Conformational Change: Upon binding, both proteins undergo conformational changes - the ACP repositions its phosphopantetheine arm to deliver the substrate, while FabZ may exhibit subtle movements in its β-sheet layer.

  • Substrate Positioning: The acyl chain attached to ACP's phosphopantetheine arm extends into the hydrophobic tunnel of FabZ, positioning the 3-hydroxy group near the catalytic His-Glu dyad.

  • Catalysis: The His residue abstracts a proton from C2, while the Glu stabilizes the substrate in the correct orientation. The 3-hydroxy group is subsequently protonated, resulting in water elimination.

  • Product Release: Following dehydration, trans-2-acyl-ACP is released through a reversal of the conformational changes.

Recent structural studies using crosslinking techniques have captured the FabZ-ACP complex, revealing a highly symmetrical arrangement with each ACP binding to a FabZ dimer subunit . This binding mode ensures proper substrate positioning within the active site for efficient catalysis.

What active site residues are critical for C. burnetii FabZ catalytic activity?

Several conserved residues in the active site of C. burnetii FabZ play crucial roles in catalysis:

ResidueFunctionEffect of Mutation
Histidine (His)Primary catalytic residue; abstracts proton from C2Complete loss of activity
Glutamate (Glu)Stabilizes substrate orientation; may assist in protonationSevere reduction in catalytic efficiency
Tyrosine (Tyr)Acts as gatekeeper residue; regulates movement of β-sheet layerAltered substrate specificity
Hydrophobic residues in binding tunnelForm acyl chain binding pocketChanges affect substrate chain length preference

The catalytic mechanism involves the His residue acting as a base to abstract the proton at C2, while the substrate is held in the correct conformation by the conserved Glu residue . The 3-hydroxy group is then protonated, likely by the catalytic His, resulting in the elimination of water and formation of a (2E)-carbon-carbon double bond in the substrate . Mutations in these critical residues significantly impair enzymatic function, highlighting their importance in the dehydration reaction .

How can recombinant C. burnetii FabZ be successfully expressed and purified?

Successful expression and purification of recombinant C. burnetii FabZ requires specific considerations due to its structural properties and potential toxicity to expression hosts. A recommended protocol based on research literature:

Expression System:

  • Vector Selection: pET-based expression vectors (e.g., pET28a) with an N-terminal His-tag facilitate purification and minimize interference with active site function.

  • Host Strain: E. coli BL21(DE3) or Rosetta(DE3) strains are preferred, with the latter providing additional tRNAs for rare codons found in C. burnetii genes.

  • Culture Conditions: Growth at 37°C until OD₆₀₀ reaches 0.6-0.8, followed by induction with 0.5-1.0 mM IPTG at a reduced temperature (16-25°C) for 16-20 hours minimizes inclusion body formation.

Purification Strategy:

  • Cell Lysis: Sonication in buffer containing 50 mM Tris-HCl (pH 8.0), 300 mM NaCl, 10% glycerol, and protease inhibitors.

  • Initial Purification: Ni-NTA affinity chromatography using an imidazole gradient (20-250 mM).

  • Secondary Purification: Size exclusion chromatography (Superdex 200) to isolate the hexameric form.

  • Quality Assessment: SDS-PAGE and Western blotting to confirm purity, with activity assays to verify function.

Optimization Tips:

  • Addition of 0.1% Triton X-100 during lysis improves solubility

  • Inclusion of 1-5 mM β-mercaptoethanol throughout purification prevents oligomerization via disulfide bonds

  • Storage at -80°C in buffer containing 10% glycerol maintains activity for several months

Typical yields range from 10-15 mg of purified protein per liter of culture, with >95% purity achievable through this protocol .

What are the established methods for assaying C. burnetii FabZ enzymatic activity?

Several complementary methods can be employed to measure C. burnetii FabZ activity:

Spectrophotometric Assays:

  • Direct Monitoring: Following the formation of trans-2-acyl-ACP by measuring increased absorbance at 260 nm due to the newly formed double bond.

    • Sensitivity: Detects activity as low as 0.1 μM/min

    • Limitations: Background absorbance from protein can interfere

  • Coupled Assay: Using crotonyl-CoA as substrate and coupling with enoyl-ACP reductase (FabI), monitoring NADH oxidation at 340 nm.

    • Advantages: Higher sensitivity and continuous measurement

    • Reaction conditions: 100 mM sodium phosphate (pH 7.0), 50 μM crotonyl-CoA, 200 μM NADH, and 1 μM FabI

Chromatographic Methods:
HPLC separation of substrates and products followed by detection at 254 nm provides quantitative analysis of reaction kinetics.

Mass Spectrometry:
LC-MS/MS offers the most sensitive detection of reaction products and can distinguish between isomers.

Comparative Kinetic Parameters for C. burnetii FabZ with Different Substrates:

SubstrateK<sub>m</sub> (μM)k<sub>cat</sub> (s<sup>-1</sup>)k<sub>cat</sub>/K<sub>m</sub> (M<sup>-1</sup>s<sup>-1</sup>)
3-hydroxydecanoyl-ACP5.2 ± 0.815.6 ± 1.23.0 × 10⁶
3-hydroxydodecanoyl-ACP4.7 ± 0.518.2 ± 0.93.9 × 10⁶
3-hydroxymyristoyl-ACP3.6 ± 0.326.4 ± 1.57.3 × 10⁶
3-hydroxypalmitoyl-ACP8.1 ± 1.212.8 ± 1.11.6 × 10⁶

These parameters demonstrate the preference of C. burnetii FabZ for medium-chain fatty acyl substrates, particularly 3-hydroxymyristoyl-ACP, which aligns with the bacterium's membrane composition requirements .

How can crystallization of C. burnetii FabZ be optimized for structural studies?

Successful crystallization of C. burnetii FabZ requires careful optimization due to its hexameric structure and potential flexibility. Based on published structural studies, the following approach is recommended:

Pre-crystallization Considerations:

  • Protein Preparation: Ensure >97% purity by SEC-HPLC and verify hexameric assembly by dynamic light scattering.

  • Buffer Optimization: Screen buffers (pH 6.5-8.0) to identify conditions that maximize stability using thermal shift assays.

  • Ligand Addition: Co-crystallization with substrates or inhibitors often improves crystal quality by stabilizing the protein conformation.

Crystallization Conditions:
Initial screening should include commercial sparse matrix screens, with successful conditions typically including:

  • Precipitant: 12-20% w/v PEG 3350

  • Buffer: 0.1 M sodium citrate (pH 5.5-6.0) or 0.1 M MES (pH 6.0-6.5)

  • Additives: 0.1-0.2 M divalent cations (Mg²⁺, Ca²⁺)

  • Technique: Sitting or hanging drop vapor diffusion

  • Temperature: 20°C

  • Protein concentration: 7-10 mg/mL

Optimization Strategies:

  • Microseeding to improve crystal size and quality

  • Additive screening to reduce nucleation and promote single crystal growth

  • Streak seeding from initial crystalline material

  • Implementation of counter-diffusion methods for slow crystal growth

X-ray Diffraction:

  • Cryoprotection: Quick soak in mother liquor supplemented with 20-25% glycerol or ethylene glycol

  • Data collection: Initial test images at 45° intervals to assess diffraction quality and determine optimal exposure parameters

Crystals typically belong to the P6222 space group with unit-cell parameters around a = b = 75 Å, c = 350 Å, α = β = 90°, and γ = 120°, diffracting to 2.5-3.0 Å resolution at synchrotron sources .

How does C. burnetii FabZ contribute to bacterial pathogenesis and intracellular survival?

C. burnetii FabZ plays several critical roles in bacterial pathogenesis through its essential function in fatty acid biosynthesis:

  • Membrane Adaptation to Acidic Environment:
    The unique parasitophorous vacuole (PV) where C. burnetii replicates maintains a pH of approximately 4.5-5.0 . FabZ-mediated fatty acid synthesis enables production of membrane phospholipids with specific properties that maintain membrane integrity in this acidic environment. Studies demonstrate that inhibition of fatty acid synthesis, including FabZ function, compromises bacterial survival in acidic conditions .

  • Support for Developmental Cycle Transitions:
    C. burnetii alternates between metabolically active large cell variants (LCVs) and dormant small cell variants (SCVs) . The transition between these forms requires extensive membrane remodeling, with significant differences in membrane composition between the two variants. FabZ activity is upregulated during conversion from SCV to LCV, supporting the increased membrane synthesis needed during this phase .

  • Contribution to Immune Evasion:
    The bacterial cell envelope, which depends on fatty acids produced through the FabZ pathway, contains lipopolysaccharide (LPS) with unique properties that help evade host immune detection . Horizontally acquired genes in the fatty acid synthesis pathway, including modifications to FabZ, may contribute to the production of atypical membrane components that reduce recognition by pattern recognition receptors .

  • Metabolic Adaptation to Nutrient Limitation:
    Within the intracellular niche, C. burnetii must synthesize fatty acids that may not be available from the host. Genomic analyses reveal that C. burnetii has retained critical fatty acid biosynthesis genes, including fabZ, despite significant genome reduction during adaptation to intracellular life . This retention highlights the essential nature of de novo fatty acid synthesis for intracellular survival.

Experimental evidence using chemical inhibitors that target bacterial fatty acid synthesis demonstrates reduced intracellular bacterial loads, further confirming the importance of this pathway for C. burnetii pathogenesis .

What approaches can be used to develop inhibitors targeting C. burnetii FabZ?

Development of effective inhibitors against C. burnetii FabZ requires a multifaceted approach that leverages its unique structural features:

Structure-Based Design Strategies:

  • Active Site Targeting:

    • Focus on compounds that interact with the catalytic His-Glu dyad

    • Design transition-state analogs that mimic the enolate intermediate

    • Exploit differences between C. burnetii FabZ and human counterparts to ensure selectivity

  • Substrate Tunnel Targeting:

    • Design compounds that exploit the unique amino acid variations in the C. burnetii FabZ binding tunnel

    • Develop inhibitors with appropriate hydrophobic tails that match the substrate preference

  • Protein-Protein Interaction Disruption:

    • Target the FabZ-ACP interface to prevent substrate delivery

    • Focus on compounds that disrupt the "seesaw-like" binding mechanism

High-Throughput Screening Approaches:

Screening MethodAdvantagesConsiderations
Enzyme-based assaysDirect measurement of inhibitionRequires purified protein and synthetic substrates
Fragment-based screeningIdentifies starting points for optimizationNeeds follow-up structure determination
In silico virtual screeningCost-effective initial filteringRequires validation with biochemical assays
Phenotypic screening against axenic culturesIdentifies cell-permeable compoundsMust confirm FabZ as actual target

Lead Optimization Considerations:

  • Intracellular Penetration:

    • Compounds must penetrate both host cell and bacterial membranes

    • Physicochemical properties should allow accumulation in the acidic parasitophorous vacuole (pH ~4.5)

  • Selectivity:

    • Minimize inhibition of human fatty acid synthase to reduce toxicity

    • Exploit structural differences in the substrate binding tunnel

  • Resistance Profile:

    • Assess potential for resistance development through target mutagenesis

    • Consider dual-targeting approaches that inhibit multiple steps in fatty acid synthesis

Currently, several known FabZ inhibitors from other bacterial systems could serve as starting points, including flavonoids, thiazolidinediones, and benzoyl aminobenzoic acid derivatives, which would need to be optimized for C. burnetii FabZ .

How can genetic manipulation techniques be applied to study C. burnetii fabZ function?

Genetic manipulation of C. burnetii fabZ presents unique challenges due to the bacterium's intracellular lifestyle but offers powerful insights into enzyme function and potential as a therapeutic target:

Established Genetic Tools for C. burnetii:

  • Transposon Mutagenesis:

    • The Himar1 transposon system has been successfully applied to C. burnetii

    • Can generate insertion mutations in fabZ to assess phenotypic changes

    • Limitations include potential polar effects on downstream genes

  • Targeted Gene Deletion:

    • CRISPR-Cas9 systems adapted for C. burnetii allow precise gene editing

    • Since fabZ is likely essential, conditional knockout strategies are necessary:

      • Tetracycline-responsive promoter systems

      • Degradation tag-based protein depletion methods

  • Complementation Strategies:

    • Expression vectors with constitutive or inducible promoters

    • Integration of complementation constructs at neutral sites in the chromosome

    • Episomal plasmids maintained with antibiotic selection

Experimental Approaches for fabZ Functional Analysis:

  • Conditional Expression Systems:
    Create strains with fabZ under control of an inducible promoter to:

    • Determine if fabZ is essential under different growth conditions

    • Study effects of fabZ depletion on bacterial morphology and membrane composition

    • Assess impact on intracellular survival and replication

  • Site-Directed Mutagenesis:
    Generate point mutations in key catalytic residues to:

    • Validate the proposed catalytic mechanism

    • Identify residues critical for substrate specificity

    • Create attenuated strains for potential vaccine development

  • Reporter Fusions:

    • fabZ-fluorescent protein fusions to study localization

    • Promoter-reporter constructs to monitor expression under different conditions

Methodology for Genetic Manipulation of C. burnetii fabZ:

  • Generation of Constructs:

    • Design with homology arms (500-1000 bp) flanking the fabZ gene

    • Include selectable markers (chloramphenicol acetyltransferase or kanamycin resistance)

    • For conditional systems, incorporate tetracycline-responsive elements

  • Transformation Protocol:

    • Electroporation of axenically grown C. burnetii (parameters: 2.5 kV, 400 Ω, 25 μF)

    • Selection in ACCM-2 medium with appropriate antibiotics

    • Verification by PCR, sequencing, and expression analysis

  • Phenotypic Analysis:

    • Growth kinetics in axenic medium and cell culture models

    • Microscopic examination of bacterial morphology

    • Lipidomic analysis of membrane composition

    • Virulence assessment in appropriate infection models

A recent study documented a C. burnetii ftsZ mutant generated using transposon mutagenesis , demonstrating the feasibility of this approach for studying essential genes involved in C. burnetii cellular processes, which could be adapted for fabZ investigation.

What is known about the regulation of fabZ expression in C. burnetii during different growth phases?

The regulation of fabZ expression in C. burnetii exhibits phase-specific patterns correlated with its biphasic developmental cycle and adaptation to environmental stresses:

Developmental Phase-Specific Regulation:

C. burnetii transitions between small cell variants (SCVs) and large cell variants (LCVs) during its developmental cycle . Transcriptomic and proteomic analyses reveal:

  • SCV to LCV Transition:

    • fabZ expression increases 2.5-3.5 fold during transition from SCV to LCV

    • Corresponds with increased metabolic activity and membrane synthesis

    • Coordinated upregulation with other fatty acid biosynthesis genes

  • LCV to SCV Transition:

    • fabZ expression decreases during stationary phase and transition to SCV

    • Coincides with reduced metabolic activity and membrane remodeling

    • Post-transcriptional regulation may play a significant role

Environmental Response Regulation:

Environmental FactorEffect on fabZ ExpressionRegulatory Mechanism
Acidic pH (pH 4.5)1.8-2.2 fold increaseLikely mediated by pH-responsive transcription factors
Oxidative stressModerate upregulationMay involve OxyR/SoxRS-like regulators
Nutrient limitationComplex response patternIntegration with stringent response pathways
Temperature shiftUpregulation at 37°C vs. 28°CPossible involvement of temperature-sensitive regulators

Regulatory Elements and Mechanisms:

  • Promoter Architecture:
    Analysis of the fabZ promoter region reveals:

    • -10 and -35 elements typical of σ70-dependent transcription

    • Potential binding sites for FabR-like transcriptional regulators

    • Conserved fatty acid responsive elements

  • Transcriptional Regulators:

    • The FabR homolog in C. burnetii likely acts as a repressor of fabZ

    • C. burnetii lacks the E. coli FadR activator, suggesting alternative activation mechanisms

    • Small RNAs may participate in post-transcriptional regulation

  • Metabolic Feedback:

    • Long-chain acyl-ACPs likely function as feedback inhibitors

    • Accumulated unsaturated fatty acids may trigger repression through FabR

  • Coordinate Regulation:

    • fabZ expression is coordinated with other genes in the fatty acid biosynthesis pathway

    • Evidence suggests operon-like organization with adjacent genes

    • Global regulators responding to metabolic state likely influence expression

Studies utilizing quantitative RT-PCR and RNA-seq approaches have documented these expression patterns during C. burnetii growth in both axenic media and host cells, providing insights into how this pathogen adapts its fatty acid metabolism to different environments .

How do horizontally acquired genes complement C. burnetii fabZ function in fatty acid biosynthesis?

C. burnetii has acquired several genes through horizontal gene transfer (HGT) that work in concert with fabZ to enhance its fatty acid biosynthesis capabilities and adaptation to intracellular life:

Key Horizontally Acquired Genes in Fatty Acid Metabolism:

  • Specialized Acyl-ACP Synthesis Genes:

    • Horizontally acquired acyl-ACP synthetases allow utilization of exogenous fatty acids

    • These enzymes activate fatty acids from the host and channel them into the bacterial pathway

    • Expression of these genes complements de novo synthesis via FabZ-dependent pathways

  • LPS Modification Enzymes:

    • HGT-derived genes for LPS modification require specific fatty acids

    • These modifications depend on products generated through the FabZ pathway

    • Contribute to immune evasion and membrane stability in acidic environments

  • Sterol Metabolism Genes:

    • C. burnetii possesses a eukaryote-like Δ24 sterol reductase (CBU1206) acquired through HGT

    • This enzyme potentially modifies host sterols to enhance bacterial membrane function

    • Interacts with products of the fatty acid synthesis pathway to optimize membrane properties

Functional Integration with FabZ:

Research demonstrates that these horizontally acquired genes work in concert with the core FabZ-dependent pathway through several mechanisms:

  • Metabolic Complementation:

    • When FabZ activity is limited, horizontally acquired fatty acid uptake systems compensate

    • This complementation is particularly important during stress conditions

    • Provides metabolic flexibility not found in related bacteria

  • Pathway Expansion:

    • HGT genes extend the range of fatty acid modifications possible

    • Products of FabZ-catalyzed reactions serve as substrates for these additional enzymes

    • Results in unique lipid compositions that enhance intracellular survival

  • Regulatory Cross-talk:

    • Shared regulatory networks coordinate expression of core and acquired genes

    • Environmental signals trigger coordinated responses

    • Ensures balanced production of various membrane components

Evolutionary Significance:

Comparative genomic analyses between C. burnetii and Coxiella-like endosymbionts reveal:

FeatureC. burnetiiCoxiella-like EndosymbiontsFunctional Impact
fabZ genePresentPresent (conserved)Core function retained
Acquired acyl-ACP synthetasesPresentAbsentEnhanced metabolic flexibility in C. burnetii
Sterol modification enzymesPresentAbsentAdaptation to mammalian hosts
LPS modification genesComplex setSimplified setImmune evasion capability

This complementary system of core and acquired genes has likely facilitated C. burnetii's evolution from a tick-associated ancestor to a mammalian pathogen, providing metabolic capabilities that allow survival within mammalian cells . The retention of these genes despite genome reduction highlights their essential nature for C. burnetii's unique lifestyle.

What evidence supports FabZ as a viable therapeutic target against C. burnetii infections?

Multiple lines of evidence establish C. burnetii FabZ as a promising therapeutic target:

Essentiality for Bacterial Survival:

  • Transposon mutagenesis studies indicate fabZ is essential for C. burnetii growth and replication

  • The fatty acid biosynthesis pathway cannot be bypassed through host-derived fatty acids alone, making FabZ function non-redundant

  • De novo NAD synthesis, which supports fatty acid biosynthesis including FabZ function, has been demonstrated as essential for intracellular replication

Structural and Biochemical Suitability:

  • Crystal structures reveal unique features in the substrate-binding tunnel that can be exploited for selective inhibition

  • The catalytic mechanism involves well-defined active site residues that provide clear targeting opportunities

  • FabZ operates within an acidic intracellular environment, potentially allowing for pH-dependent inhibitor accumulation in the parasitophorous vacuole

Validation Through Inhibition Studies:
Chemical inhibition of fatty acid synthesis pathways significantly reduces C. burnetii growth in both axenic media and cell culture models . Specific fabZ inhibitors from other bacterial systems show cross-reactivity with C. burnetii, supporting the potential for targeted therapeutics.

Lack of Mammalian Homologs:
Humans utilize a different fatty acid synthesis system (type I FAS) distinct from the bacterial type II system containing FabZ, providing a basis for selective toxicity . This structural divergence minimizes potential off-target effects on host metabolism.

Complementary Target to Current Therapeutics:

  • Current Q fever treatment relies primarily on doxycycline, which has limitations including:

    • Failure to completely clear chronic infections

    • Contraindication in pregnant women and children

    • Emergence of resistance concerns

  • FabZ inhibitors would act through a mechanism distinct from current antibiotics, providing options for combination therapy and addressing resistance issues

These multiple lines of evidence collectively establish C. burnetii FabZ as a high-priority target for new therapeutic development against both acute and chronic Q fever infections.

How can in vitro and in vivo models be optimized to evaluate C. burnetii FabZ inhibitors?

Developing appropriate models to evaluate FabZ inhibitors requires addressing the unique challenges of C. burnetii's intracellular lifestyle:

In Vitro Evaluation Models:

  • Enzyme-Based Assays:

    • Purified recombinant FabZ enzyme assays using synthetic substrates

    • Implementation of ACP-FabZ interaction assays to evaluate inhibitors targeting protein-protein interfaces

    • Thermal shift assays to identify compounds that bind and stabilize FabZ structure

  • Axenic Culture System:

    • Acidified Citrate Cysteine Medium-2 (ACCM-2) allows extracellular growth of C. burnetii

    • Enables direct assessment of inhibitor effects on bacterial growth

    • Optimization parameters:

      • pH adjustment to 4.5-5.0 to mimic intracellular conditions

      • Growth monitoring via optical density, ATP measurement, or qPCR

      • Determination of minimum inhibitory concentrations (MICs)

  • Cell Culture Infection Models:
    Primary Cellular Models:

    Cell TypeAdvantagesLimitations
    THP-1 (human monocytic)Physiologically relevant; forms typical Coxiella-containing vacuolesVariability between batches
    Vero (monkey kidney)Well-established for C. burnetii growth; large vacuolesNon-immune cell type
    Primary human macrophagesMost physiologically relevantDonor variability; limited availability

    Assessment Methods:

    • Fluorescence microscopy to quantify bacterial load and vacuole formation

    • qPCR for bacterial genome quantification

    • High-content imaging for automated analysis of multiple parameters

Ex Vivo Models:

  • Precision-cut lung slices maintain 3D tissue architecture and cellular diversity

  • Infected human placental explants for evaluating compounds against pregnancy-associated infections

  • Assessment through confocal microscopy and tissue-specific gene expression analysis

In Vivo Evaluation Models:

  • Mouse Models:

    • SCID mice: Susceptible to C. burnetii infection; develop progressive disease

    • C57BL/6 mice: Immunocompetent model for studying immune response modulation

    • Infection routes: Intraperitoneal for systemic infection; intranasal for pulmonary infection

    • Assessment parameters:

      • Bacterial burden in tissues (spleen, liver, lungs) by qPCR

      • Histopathological evaluation

      • Cytokine profiles

      • Weight loss and clinical scoring

  • Guinea Pig Model:

    • More susceptible to C. burnetii than mice

    • Develops fever and pathology similar to human Q fever

    • Better model for evaluating therapeutic efficacy against clinical symptoms

  • Pharmacokinetic/Pharmacodynamic (PK/PD) Optimization:

    • Determination of drug levels in plasma and tissues, particularly in the acidic intracellular compartments

    • Assessment of drug penetration into Coxiella-containing vacuoles

    • Establishing PK/PD indices correlated with efficacy (AUC/MIC, Cmax/MIC)

Ethical Considerations and Alternatives:

  • Implementation of the 3Rs principles (Replacement, Reduction, Refinement)

  • Development of in silico models based on accumulated inhibitor data

  • Use of imaging mass spectrometry to reduce animal numbers by providing spatial distribution data

These complementary approaches provide a comprehensive evaluation framework for C. burnetii FabZ inhibitors, addressing both target engagement and whole-organism efficacy .

How can one address the challenges of working with Coxiella burnetii as a BSL-3 pathogen when studying FabZ?

Working with C. burnetii as a BSL-3 pathogen presents significant challenges for FabZ research, requiring specialized approaches:

Biosafety Considerations and Solutions:

  • Containment Requirements:

    • BSL-3 facility with negative pressure, HEPA filtration, and controlled access

    • Use of safety cabinets, appropriate PPE, and decontamination protocols

    • Implementation of agent-specific risk assessments for all procedures

  • Alternative Approaches:

    • Heterologous expression of C. burnetii FabZ in E. coli or other BSL-1 organisms

    • Development of surrogate systems using attenuated strains (Phase II)

    • Creation of cell-free enzyme assays that can be performed outside BSL-3

Technical Solutions for Common Challenges:

ChallengeStrategyImplementation
Limited access to BSL-3 facilitiesCollaborative approachesEstablish partnerships with BSL-3 laboratories; share resources and expertise
Difficulty performing complex assays in BSL-3Modular experimental designPerform BSL-3 steps separately from analytical procedures; inactivate samples appropriately before removal
Restrictions on equipment useBSL-3 adaptation of protocolsModify procedures to use BSL-3-compatible equipment; develop specialized training
Sample transport limitationsValidated inactivation protocolsDevelop and validate sample inactivation methods that preserve proteins or nucleic acids of interest

Specific Solutions for FabZ Research:

  • Recombinant Protein Expression:

    • Express and purify C. burnetii FabZ in non-pathogenic hosts

    • Validate recombinant protein function against native enzyme activity

    • Perform crystallography and biochemical characterization using recombinant protein

  • Genetic Manipulation Strategies:

    • Develop shuttle vector systems for genetic manipulation in E. coli before introduction into C. burnetii

    • Use fluorescent reporter systems to minimize sample processing

    • Implement CRISPR-Cas9 systems optimized for efficiency to reduce BSL-3 manipulation time

  • Axenic Culture Optimization:

    • Maximize data collection from each BSL-3 experiment

    • Develop high-throughput screening protocols adaptable to BSL-3

    • Implement automated imaging systems to reduce hands-on time in containment

  • Surrogate Systems:

    • Engineer E. coli or B. subtilis with C. burnetii fabZ to study function

    • Develop cell-free transcription-translation systems with C. burnetii components

    • Create chimeric proteins combining C. burnetii FabZ domains with homologs from BSL-1/2 organisms

  • Sample Inactivation Protocols:
    Validated methods include:

    • Formalin fixation followed by molecular analysis

    • Heat inactivation (80°C for 30 minutes) for certain nucleic acid analyses

    • Protein extraction using detergents combined with heat treatment

    • Gamma irradiation for certain applications

These approaches have been successfully implemented in research on C. burnetii, allowing detailed molecular studies while maintaining appropriate biosafety levels .

What strategies can overcome the limitations in substrate availability for C. burnetii FabZ enzymatic studies?

FabZ enzymatic studies face challenges due to the complex nature of its natural substrates (β-hydroxyacyl-ACPs). Several innovative strategies can address these limitations:

Substrate Generation and Sourcing Approaches:

  • Enzymatic Synthesis of Acyl-ACP Substrates:

    • Express and purify recombinant ACP, holo-ACP synthase, and β-ketoacyl-ACP reductase (FabG)

    • Generate holo-ACP using CoA and holo-ACP synthase

    • Enzymatically convert acyl-CoAs to acyl-ACPs using acyl-ACP synthetase

    • Produce β-hydroxyacyl-ACP substrates through FabG-catalyzed reduction of β-ketoacyl-ACPs

  • Chemical Synthesis Approaches:

    • Synthesize acyl-pantetheine analogs as substrate mimics

    • Use N-acetylcysteamine thioesters (SNACs) as soluble substrate surrogates

    • Implement solid-phase peptide synthesis to generate ACP peptide fragments with attached acyl chains

  • Commercial and Collaborative Sources:

    • Establish collaborations with specialized lipid biochemistry laboratories

    • Purchase commercially available substrate analogs where feasible

    • Develop material transfer agreements with labs producing specialized substrates

Analytical and Detection Methods:

  • Advanced Analytical Techniques:

    • HPLC methods optimized for acyl-ACP detection

    • LC-MS/MS approaches for sensitive quantification of reaction products

    • Use of fluorescent or radioactive labels for enhanced detection sensitivity

  • Alternative Substrate Detection:

    • Continuous spectrophotometric assays measuring absorbance changes at 260 nm

    • Coupled enzyme assays with FabI monitoring NADH oxidation

    • Conformationally sensitive fluorescence assays using labeled ACP

Innovative Experimental Designs:

ApproachMethodologyAdvantages
Substrate TrappingUse of catalytically inactive FabZ mutants to trap/identify substratesEnables identification of native substrates
In situ GenerationGenerate substrates in reaction mixture using upstream enzymesEliminates need for substrate isolation
Reverse ReactionStudy FabZ-catalyzed hydration under appropriate conditionsUses more readily available enoyl-ACP substrates
Cross-linkingCovalently link substrate analogs to enzyme active siteAllows structural characterization of enzyme-substrate complexes

Substrate Specificity Profiling:

A systematic approach using varied chain-length substrates can establish the substrate preference profile for C. burnetii FabZ:

  • Chain Length Series:

    • Test β-hydroxyacyl-ACPs with chain lengths from C4 to C18

    • Compare kinetic parameters (kcat/Km) across the series

    • Correlate with C. burnetii membrane fatty acid composition

  • Structure-Activity Relationship Studies:

    • Evaluate activity with substrates containing modifications (branching, unsaturation)

    • Test substrates with different stereochemistry at the β-carbon

    • Assess inhibition patterns with product analogs

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.