Recombinant Helicobacter hepaticus (3R)-hydroxymyristoyl-[acyl-carrier-protein] dehydratase, commonly referred to as FabZ, is an enzyme involved in the biosynthesis of fatty acids. This enzyme catalyzes the dehydration of beta-hydroxyacyl-acyl carrier proteins (ACPs) to form trans-2-acyl-ACPs, a crucial step in the fatty acid synthesis pathway. While specific research on the recombinant form of FabZ from Helicobacter hepaticus is limited, understanding its function and significance can be inferred from studies on similar enzymes in other bacteria.
FabZ is a key enzyme in the type II fatty acid synthase (FASII) system, which is responsible for producing fatty acids in bacteria. The enzyme's primary function is to catalyze the dehydration of beta-hydroxyacyl-ACPs, converting them into trans-2-acyl-ACPs. This process is essential for the elongation and modification of fatty acid chains, which are vital components of bacterial membranes and other cellular structures.
Catalytic Activity: Dehydration of beta-hydroxyacyl-ACPs to trans-2-acyl-ACPs.
Structural Organization: Typically exists as a hexamer in its native state, which enhances stability and enzymatic activity.
Substrate Specificity: Can act on both short-chain and long-chain saturated and unsaturated beta-hydroxyacyl-ACPs.
While specific studies on recombinant Helicobacter hepaticus FabZ are not readily available, research on similar enzymes provides valuable insights into their potential roles and applications:
Helicobacter pylori FabZ: Studies on Helicobacter pylori have shown that FabZ is crucial for the bacterium's survival and pathogenicity. The enzyme's stability and activity make it a potential target for antibacterial therapies .
Helicobacter hepaticus Pathogenicity: Helicobacter hepaticus is known to cause hepatitis and liver preneoplasia in mice, suggesting that enzymes like FabZ could play roles in its pathogenic mechanisms by supporting bacterial growth and survival .
Understanding the function and structure of FabZ enzymes can lead to several applications:
Antibacterial Drug Development: Targeting FabZ could provide a novel approach to inhibiting bacterial growth, especially in pathogens like Helicobacter pylori and potentially Helicobacter hepaticus.
Biotechnological Applications: The ability to modify fatty acid synthesis pathways could be exploited in biotechnology for producing specific fatty acids or related compounds.
| Characteristics | Description |
|---|---|
| Enzyme Function | Catalyzes dehydration of beta-hydroxyacyl-ACPs to trans-2-acyl-ACPs. |
| Structural Form | Typically exists as a hexamer. |
| Substrate Specificity | Acts on short-chain and long-chain saturated and unsaturated beta-hydroxyacyl-ACPs. |
| Stability | High thermal stability, as observed in Helicobacter pylori FabZ. |
| Potential Applications | Target for antibacterial therapies; biotechnological applications in fatty acid synthesis. |
KEGG: hhe:HH_1181
STRING: 235279.HH1181
(3R)-hydroxymyristoyl-[acyl-carrier-protein] dehydratase (fabZ) in Helicobacter hepaticus is an essential enzyme involved in type II fatty acid synthesis (FAS II) pathway. Based on homology to related species like H. pylori, it catalyzes the dehydration of short-chain beta-hydroxyacyl-ACPs and long-chain saturated and unsaturated beta-hydroxyacyl-ACPs . The enzyme is crucial for bacterial membrane phospholipid biosynthesis and thus bacterial survival. The gene encoding this protein is designated as fabZ, and the enzyme functions in the cytoplasm where fatty acid biosynthesis occurs. While specific characterization data for H. hepaticus fabZ is limited, related enzymes in H. pylori have a molecular weight of approximately 18 kDa and function in both fatty acid biosynthesis and lipid A biosynthetic processes .
Recombinant H. hepaticus fabZ can be expressed using several prokaryotic and eukaryotic expression systems, each with advantages and limitations. The methodological approach should be selected based on research objectives:
E. coli expression systems: Commonly using BL21(DE3) or Rosetta strains with pET or pGEX vectors. Optimal conditions typically include:
Induction with 0.5-1.0 mM IPTG
Expression temperature of 18-25°C (to enhance solubility)
Growth in LB or TB media supplemented with appropriate antibiotics
Purification strategies:
IMAC (Immobilized Metal Affinity Chromatography) for His-tagged constructs
GST affinity chromatography for GST fusion proteins
Size exclusion chromatography as a polishing step
Experimental design should include optimization of:
Induction parameters (temperature, duration, inducer concentration)
Cell lysis methods (sonication, French press, detergent-based)
Buffer composition (pH, salt concentration, reducing agents)
Protein stabilizing additives (glycerol, detergents for membrane-associated forms)
When investigating H. hepaticus fabZ inhibition, researchers should implement a systematic experimental design approach that progresses from in vitro enzymatic assays to cellular and potentially in vivo studies. A comprehensive inhibition study would include:
Enzyme-level inhibition assessment:
Purify recombinant H. hepaticus fabZ to >95% homogeneity
Establish a reliable activity assay (spectrophotometric or HPLC-based)
Screen potential inhibitors using concentration gradients
Determine inhibition kinetics (Ki values) and mechanisms (competitive, non-competitive, uncompetitive)
Variables to control:
Cellular-level assessment:
Minimum Inhibitory Concentration (MIC) determination for H. hepaticus growth
Membrane lipid composition analysis following inhibitor treatment
Assessment of morphological changes via electron microscopy
In vivo evaluation (using mouse models):
Table: Experimental design for fabZ inhibitor evaluation:
| Variable | Control Group | Treatment Group 1 | Treatment Group 2 | Treatment Group 3 |
|---|---|---|---|---|
| Inhibitor concentration | 0 (vehicle) | Low dose | Medium dose | High dose |
| Duration of treatment | Based on pilot studies | Same as control | Same as control | Same as control |
| Sample size | n≥5 | n≥5 | n≥5 | n≥5 |
| Primary outcome measures | Bacterial load, tissue histology, inflammatory markers | Same as control | Same as control | Same as control |
To investigate the relationship between H. hepaticus fabZ activity and bacterial pathogenesis, researchers should employ a multi-tiered approach:
Generate and characterize fabZ mutants:
In vitro characterization:
Enzymatic activity assays comparing wild-type and mutant enzymes
Lipid profiling to assess changes in fatty acid composition
Growth curve analysis under various conditions (temperature, pH, nutrient limitation)
Infection models:
Mouse colonization studies with wild-type vs. fabZ-attenuated strains
Immunohistochemical detection of:
Integration of mechanistic data:
This approach enables researchers to establish causal relationships between fabZ activity and pathogenesis while controlling for confounding variables.
Structural biology approaches provide crucial insights into H. hepaticus fabZ function, inhibition mechanisms, and evolutionary relationships. Researchers should consider the following methodological framework:
Protein structure determination:
X-ray crystallography of purified H. hepaticus fabZ (apo form)
Co-crystallization with substrates and inhibitors
NMR spectroscopy for dynamic regions and ligand interactions
Cryo-EM for larger complexes (e.g., fabZ with ACP)
Structure-function relationship analysis:
Applied outcomes:
Structure-based design of specific inhibitors
Rational engineering of fabZ with altered substrate specificity
Identification of species-specific structural features for targeted therapeutics
Integration with experimental validation:
Site-directed mutagenesis of predicted key residues
Enzymatic assays of mutants to confirm structural hypotheses
Thermal shift assays to evaluate protein stability changes
A comparative structural analysis might reveal subtle differences in the active site architecture between H. hepaticus and H. pylori fabZ that could be exploited for species-specific inhibitor design.
The potential role of H. hepaticus fabZ in hepatic preneoplasia development represents an advanced research question that bridges bacterial metabolism and host pathology. Based on available evidence, researchers should investigate:
Fatty acid metabolism and membrane composition:
How fabZ-dependent changes in bacterial membrane composition affect:
Bacterial persistence in the liver
Interaction with host cell receptors
Resistance to host defense mechanisms
Host-pathogen interaction mechanisms:
Experimental approaches:
Potential mechanisms linking fabZ to preneoplasia:
This research direction could establish whether fabZ is a potential therapeutic target for preventing H. hepaticus-induced hepatic disease progression.
When faced with discrepancies between in vitro fabZ enzymatic data and in vivo phenotypic observations, researchers should implement a systematic approach to reconcile these differences:
Methodological considerations:
In vitro limitations: Recombinant enzyme may lack post-translational modifications or proper folding
Assay conditions: Buffer composition, temperature, and pH may not reflect in vivo environment
Substrate availability: Natural substrates in complex with ACP may behave differently than simplified in vitro substrates
Biological complexity factors:
Metabolic compensation: Alternative pathways may compensate for reduced fabZ activity in vivo
Regulatory networks: Feedback mechanisms may modulate enzyme activity differently in living cells
Host interactions: Host factors may influence bacterial enzyme function in infection models
Statistical approach to reconciliation:
Experimental validation strategies:
Design intermediate complexity experiments (e.g., cell-free extracts, liposome reconstitution)
Perform time-course studies to capture dynamic changes
Utilize genetic approaches (point mutations vs. knockdowns) to create activity gradients
Measure multiple endpoints simultaneously to capture system-level responses
When analyzing such discrepancies, researchers should consider that in vivo phenotypes like H. hepaticus-induced liver inflammation involve complex interactions between bacterial factors and host responses, including HMGB1 activation and downstream signaling pathways .
When analyzing data from H. hepaticus fabZ inhibition studies, researchers should employ appropriate statistical methods based on experimental design, data characteristics, and research questions:
For enzyme kinetic studies:
Non-linear regression for determining inhibition constants (Ki)
Lineweaver-Burk or Hanes-Woolf plots to distinguish inhibition mechanisms
Global fitting approaches for complex inhibition patterns
Bootstrap analysis for robust confidence interval estimation
For microbial growth inhibition:
For in vivo infection studies:
For multi-parameter analyses:
Correlation analyses between bacterial load, enzymatic activity, and disease parameters
Principal component analysis to identify patterns across multiple variables
Hierarchical clustering to identify treatment response patterns
Path analysis to test causal relationships between fabZ inhibition and disease outcomes
Importantly, researchers should establish clear statistical significance thresholds (p < 0.05 is typically considered statistically significant) and report effect sizes alongside p-values to enable proper interpretation of biological significance.
Comparative analysis of fabZ across bacterial species provides valuable insights into evolutionary conservation, functional divergence, and potential species-specific targeting approaches:
Sequence and structural comparison:
Alignment of H. hepaticus fabZ with H. pylori fabZ shows high conservation of catalytic residues
Phylogenetic analysis places Helicobacter fabZ enzymes in a distinct clade among epsilon-proteobacteria
Structural homology modeling reveals conservation of the characteristic hot dog fold common to dehydratases
Functional conservation and divergence:
Core dehydratase activity is preserved across species
Substrate chain-length preference may vary between organisms
Regulatory mechanisms and protein-protein interactions may be species-specific
Inhibitor sensitivity profiles often differ between orthologous enzymes
Methodological approach for comparative studies:
Heterologous expression of multiple fabZ orthologs
Standardized enzymatic assays under identical conditions
Thermal stability comparison across species
Cross-complementation studies in fabZ-deficient strains
Comparative data presentation:
This comparative approach enables researchers to identify both universally conserved features essential for function and species-specific characteristics that could be exploited for targeted inhibitor development.
Evolutionary analysis of fabZ provides a powerful framework for developing inhibitors with specificity toward H. hepaticus while minimizing off-target effects:
Evolutionary conservation mapping:
Identify residues under purifying selection (highly conserved) across all bacterial fabZ enzymes
Map residues under positive selection that may confer species-specific functions
Locate H. hepaticus-specific residues in or near the active site that differ from human gut microbiome species
Structure-guided approach:
Generate homology models based on H. pylori fabZ crystal structure
Use molecular dynamics simulations to identify conformational differences
Perform virtual screening against H. hepaticus-specific binding pockets
Design inhibitors targeting species-specific residues while avoiding conserved catalytic machinery
Experimental validation workflow:
Enzymatic assays comparing inhibition against multiple bacterial fabZ orthologs
Bacterial growth inhibition panels including H. hepaticus and microbiome representatives
Mouse model studies assessing H. hepaticus clearance vs. microbiome disruption
Pharmacokinetic/pharmacodynamic studies to optimize in vivo efficacy
Potential benefits of this approach:
By integrating evolutionary insights with structural biology and medicinal chemistry, researchers can develop inhibitors that exploit the unique features of H. hepaticus fabZ while minimizing disruption to the broader microbiome.