Recombinant Helicobacter hepaticus (3R)-hydroxymyristoyl-[acyl-carrier-protein] dehydratase (fabZ)

Shipped with Ice Packs
In Stock

Description

Introduction to Recombinant Helicobacter hepaticus (3R)-hydroxymyristoyl-[acyl-carrier-protein] dehydratase (fabZ)

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.

Function and Role of FabZ

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.

Key Features of FabZ:

  • 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.

Research Findings and Implications

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 .

Potential Applications and Future Research Directions

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.

Data Table: Characteristics of FabZ Enzymes

CharacteristicsDescription
Enzyme FunctionCatalyzes dehydration of beta-hydroxyacyl-ACPs to trans-2-acyl-ACPs.
Structural FormTypically exists as a hexamer.
Substrate SpecificityActs on short-chain and long-chain saturated and unsaturated beta-hydroxyacyl-ACPs.
StabilityHigh thermal stability, as observed in Helicobacter pylori FabZ.
Potential ApplicationsTarget for antibacterial therapies; biotechnological applications in fatty acid synthesis.

Product Specs

Form
Lyophilized powder
Note: While we prioritize shipping the format currently in stock, please specify your format preference during order placement for fulfillment according to your requirements.
Lead Time
Delivery times vary depending on the purchasing method and location. Please contact your local distributor for precise delivery estimates.
Note: Our 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 default glycerol concentration is 50% and serves as a guideline.
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 forms have a 12-month shelf life at -20°C/-80°C.
Storage Condition
Upon receipt, store at -20°C/-80°C. Aliquot for multiple uses to prevent repeated freeze-thaw cycles.
Tag Info
Tag type is determined during manufacturing.
The tag type is determined during production. If you require a specific tag, please inform us; we will prioritize its development.
Synonyms
fabZ; HH_11813-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
1-169
Protein Length
full length protein
Purity
>85% (SDS-PAGE)
Species
Helicobacter hepaticus (strain ATCC 51449 / 3B1)
Target Names
fabZ
Target Protein Sequence
MDTNKIQGDK PIMNVEKIRQ ILPHRYPMLL VDRVMELVQN TSDSEPNGYI KAYKNVTINE EVFLGHFPNK PIYPGVMQIE GMAQAGGLLA FVSMFGDNVA EAKNKIVYFM TIDNVKFRIP VVPGDRLVYE LKVLKHKGSI WQLGANAFVE DKLVSEAELK AMITEAKES
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

KEGG: hhe:HH_1181

STRING: 235279.HH1181

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

Q&A

What is (3R)-hydroxymyristoyl-[acyl-carrier-protein] dehydratase (fabZ) in Helicobacter hepaticus?

(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 .

What expression systems are effective for producing recombinant H. hepaticus fabZ?

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)

How should researchers design experiments to investigate H. hepaticus fabZ inhibition?

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:

    • Enzyme concentration

    • Substrate concentration and purity

    • Buffer composition and pH

    • Temperature and incubation times

  • 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):

    • Design treatment groups with appropriate controls

    • Monitor bacterial colonization in different organs

    • Assess markers of inflammation and tissue damage

    • Evaluate HMGB1 and other inflammatory markers to connect with pathogenesis mechanisms

Table: Experimental design for fabZ inhibitor evaluation:

VariableControl GroupTreatment Group 1Treatment Group 2Treatment Group 3
Inhibitor concentration0 (vehicle)Low doseMedium doseHigh dose
Duration of treatmentBased on pilot studiesSame as controlSame as controlSame as control
Sample sizen≥5n≥5n≥5n≥5
Primary outcome measuresBacterial load, tissue histology, inflammatory markersSame as controlSame as controlSame as control

What approaches can be used to study the relationship between H. hepaticus fabZ activity and bacterial pathogenesis?

To investigate the relationship between H. hepaticus fabZ activity and bacterial pathogenesis, researchers should employ a multi-tiered approach:

  • Generate and characterize fabZ mutants:

    • Site-directed mutagenesis of catalytic residues (based on homology to H. pylori FabZ)

    • Conditional knockdown systems to modulate expression levels

    • Complementation studies with wild-type versus mutant fabZ

  • 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

    • Liver histopathology assessments for preneoplastic changes

    • Immunohistochemical detection of:

      • Inflammatory markers (IL-33, TNF-α, TGF-β)

      • HMGB1 translocation from nucleus to cytoplasm in hepatocytes

      • Activation of downstream signaling pathways (MAPK, STAT3, ERK1/2)

  • Integration of mechanistic data:

    • Correlate fabZ activity levels with:

      • Bacterial colonization persistence

      • Host inflammatory response intensity

      • Development of hepatic preneoplasia timeline

      • HMGB1-dependent pathways activation

This approach enables researchers to establish causal relationships between fabZ activity and pathogenesis while controlling for confounding variables.

How can structural biology approaches enhance our understanding of H. hepaticus fabZ?

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:

    • Identification of catalytic residues through structural comparison with H. pylori fabZ

    • Molecular docking studies to predict substrate binding modes

    • Molecular dynamics simulations to understand conformational changes during catalysis

    • Computational alanine scanning to identify critical residues

  • 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.

What role might H. hepaticus fabZ play in the development of hepatic preneoplasia?

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:

    • Whether fabZ activity influences release of bacterial products that trigger:

      • HMGB1 translocation from nucleus to cytoplasm in hepatocytes

      • Activation of inflammatory pathways (MAPK, STAT3, ERK1/2)

      • Cellular proliferation markers like Ki67

  • Experimental approaches:

    • Compare wild-type H. hepaticus with fabZ-attenuated strains in BALB/c mice

    • Conduct long-term infection studies (12-18 months) to observe progression to preneoplasia

    • Perform temporal analysis of:

      • Bacterial colonization levels in liver and colon

      • Hepatic gene expression patterns

      • Histopathological changes and fibrosis development

  • Potential mechanisms linking fabZ to preneoplasia:

    • FabZ-dependent bacterial products may trigger initial hepatocyte damage

    • Chronic inflammation mediated by HMGB1 and IL-33 release

    • Sustained activation of oncogenic signaling pathways (STAT3, MAPK)

    • Increased cellular proliferation (Ki67) without appropriate apoptosis

This research direction could establish whether fabZ is a potential therapeutic target for preventing H. hepaticus-induced hepatic disease progression.

How should researchers interpret discrepancies between in vitro fabZ enzyme activity and in vivo phenotypes?

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:

    • Use multivariate analysis to identify correlational patterns

    • Employ principal component analysis to identify key variables explaining phenotypic variance

    • Develop mathematical models incorporating both enzyme kinetics and host response parameters

  • 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 .

What statistical approaches are most appropriate for analyzing H. hepaticus fabZ inhibition studies?

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:

    • Two-tailed Student's t-tests for comparing growth rates between treatment groups

    • ANOVA with post-hoc tests (Tukey or Bonferroni) for multiple treatment comparisons

    • Regression analysis for dose-response relationships

    • Time-series analysis for growth curve comparison

  • For in vivo infection studies:

    • Power analysis to determine appropriate sample sizes (typically n≥5 per group)

    • Mixed-effects models to account for repeated measures and individual variation

    • Survival analysis for time-to-event data

    • Non-parametric tests when data violate normality assumptions

  • 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.

How does H. hepaticus fabZ compare functionally to orthologous enzymes in other bacterial pathogens?

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.

How can researchers leverage evolutionary analysis of fabZ to develop species-specific inhibitors?

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:

    • Reduced disruption of beneficial microbiome bacteria

    • Lower likelihood of resistance development through horizontal gene transfer

    • Possibility of targeting multiple Helicobacter species with a single compound class

    • Potential therapeutic application for preventing H. hepaticus-induced liver pathology

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.

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.