Recombinant Bartonella henselae Dihydroorotate dehydrogenase (quinone) (pyrD)

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 in order notes for customized fulfillment.
Lead Time
Delivery times vary depending on the purchase method and location. Please contact your local distributor for precise delivery estimates.
Note: Standard shipping includes blue ice packs. Dry ice shipping requires advance notice 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 settle 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% and can serve as a guideline.
Shelf Life
Shelf life depends on storage conditions, buffer components, temperature, and protein stability. Generally, liquid forms 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. Avoid 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
pyrD; BH03870; Dihydroorotate dehydrogenase; quinone; EC 1.3.5.2; DHOdehase; DHOD; DHODase; Dihydroorotate oxidase
Buffer Before Lyophilization
Tris/PBS-based buffer, 6% Trehalose.
Datasheet
Please contact us to get it.
Expression Region
1-362
Protein Length
full length protein
Purity
>85% (SDS-PAGE)
Species
Bartonella henselae (strain ATCC 49882 / DSM 28221 / Houston 1) (Rochalimaea henselae)
Target Names
pyrD
Target Protein Sequence
MSFFRCIGRS ALFMLDPEHA HRLAIMGLKS GLNSYQKVVD NRLCVTIAGL KFENFIGLAA GFDKNAEVVN DVFHLGFGFT EIGTVTPRPQ VGNPKPRLFR LRKDEAIINR MGFNNDGRQI VYGRLHGYKR LGIVGINIGA NKDTVDKIDD YITSIAYFYD VADYFTVNIS SPNTPGLRDL QVRDSLHLLM NAISQARNEQ KKKHGFFVPI FLKIAPDLSE KELDDVAEEM KLSDFDGLIV SNTTLSRQGL RECTLRNEEG GLSGRPLFER STIVLAKMRQ KLGKKIAIIG VGGIRDAKTA LEKVKAGADL VQLYSGMVYE GPDLAITILK EILQFMQKDG VESIKAYRDQ RVEYWAKHML SS
Uniprot No.

Target Background

Function

Function: Catalyzes the conversion of dihydroorotate to orotate using quinone as an electron acceptor.

Database Links

KEGG: bhe:BH03870

STRING: 283166.BH03870

Protein Families
Dihydroorotate dehydrogenase family, Type 2 subfamily
Subcellular Location
Cell membrane; Peripheral membrane protein.

Q&A

What is Bartonella henselae and why is its pyrD enzyme significant for research?

Bartonella henselae is a fastidious, slow-growing gram-negative bacillus that serves as the etiologic agent of cat scratch disease, primarily affecting humans through transmission via cats and cat fleas (Ctenocephalides felis) . The organism demonstrates remarkable environmental resilience, with studies showing it can remain viable in various biological fluids and even after desiccation . Within bacterial metabolism, dihydroorotate dehydrogenase (pyrD) plays a critical role in the de novo pyrimidine biosynthesis pathway, catalyzing the oxidation of dihydroorotate to orotate. This reaction represents the sole redox step in pyrimidine biosynthesis and connects this pathway to the respiratory chain through quinone electron acceptors.

The significance of pyrD extends beyond its metabolic role, as several characteristics make it an attractive research target. First, the enzyme represents a potential vulnerability in bacterial survival since pyrimidine nucleotides are essential for DNA and RNA synthesis. Second, bacterial DHODHs differ structurally from their mammalian counterparts, offering opportunities for selective targeting in antimicrobial development. Third, given B. henselae's intracellular lifestyle and association with various inflammatory and proliferative conditions, understanding its metabolic enzymes provides insight into pathogenicity mechanisms.

Research on B. henselae pyrD contributes to our understanding of this pathogen's metabolic adaptations, potentially revealing how it survives in diverse environments ranging from the cat flea gut to human vasculature. The enzyme's central role in nucleotide metabolism makes it particularly relevant for investigating bacterial persistence mechanisms in chronic infections.

How does the structure and function of B. henselae pyrD compare with other bacterial dihydroorotate dehydrogenases?

The dihydroorotate dehydrogenase from Bartonella henselae belongs to the Class 2 family of DHODHs, which are membrane-associated enzymes that utilize quinones as electron acceptors. This contrasts with Class 1 DHODHs found in some anaerobic bacteria and certain eukaryotes, which are cytosolic and use NAD+ as electron acceptors. The Class 2 designation is significant because it indicates potential structural similarities with DHODHs from other alpha-proteobacteria while maintaining distinct characteristics that may reflect B. henselae's unique ecological niche.

Structurally, B. henselae pyrD is expected to possess the characteristic alpha/beta barrel fold with a FMN cofactor binding site, which is evolutionarily conserved across Class 2 enzymes. The quinone binding site, positioned within the membrane-associated domain, likely shows greater variation compared to other bacterial species, potentially reflecting adaptation to different electron transport chains. These structural differences may influence substrate specificity, catalytic efficiency, and susceptibility to inhibition.

From a functional perspective, pyrD catalyzes the oxidation of dihydroorotate to orotate while reducing a quinone to quinol, with this electron transfer eventually feeding into the respiratory chain. The enzyme's parameters such as optimal pH, temperature stability, and kinetic constants may show species-specific variations that reflect B. henselae's adaptation to its intracellular lifestyle. The enzyme's subcellular localization, potentially associated with the bacterial membrane, aligns with its role in connecting pyrimidine synthesis to respiration.

Comparative analysis with DHODHs from other bacterial pathogens like Helicobacter pylori and Mycobacterium tuberculosis may reveal evolutionary adaptations specific to Bartonella's lifecycle, which includes survival in both feline hosts and arthropod vectors. These adaptations could manifest as differences in catalytic efficiency, inhibitor susceptibility, or protein stability under various environmental conditions.

What are the most effective methods for expression and purification of recombinant B. henselae pyrD?

The expression and purification of recombinant B. henselae dihydroorotate dehydrogenase presents several technical challenges that require careful experimental design. The preferred expression system for functional bacterial membrane-associated proteins like Class 2 DHODHs is typically E. coli, with BL21(DE3) or its derivatives serving as common host strains. The pyrD gene should be PCR-amplified from B. henselae genomic DNA with primers designed to include appropriate restriction sites for subsequent cloning into a suitable expression vector such as pET-28a(+) or pET-22b(+), which provide N-terminal or C-terminal His-tags for affinity purification.

Optimization of expression conditions is critical for obtaining functional enzyme. Induction with IPTG concentrations between 0.1-0.5 mM at lower temperatures (16-25°C) often yields better results for membrane-associated proteins by slowing expression and allowing proper folding. The addition of 1% glucose to the growth medium may help reduce basal expression and prevent toxicity. Expression trials comparing different media formulations (such as LB, TB, or auto-induction media) and induction parameters should be conducted to maximize protein yield while maintaining enzyme activity.

For purification, a multi-step approach typically yields the best results. Initial lysis under native conditions using a buffer containing 50 mM Tris-HCl pH 8.0, 300 mM NaCl, 10% glycerol, and a mild detergent such as 0.1% Triton X-100 or 1% CHAPS helps solubilize the membrane-associated enzyme while preserving activity. The first purification step employs immobilized metal affinity chromatography (IMAC) using Ni-NTA resin, with carefully optimized imidazole concentrations in the wash and elution buffers to minimize non-specific binding while maximizing target protein recovery.

Secondary purification steps often include ion exchange chromatography on Q or SP Sepharose depending on the protein's theoretical pI, followed by size exclusion chromatography to remove aggregates and achieve high purity. Throughout the purification process, maintaining a detergent concentration above its critical micelle concentration is essential to prevent protein aggregation. The addition of stabilizing agents such as glycerol (10-20%) and reducing agents like DTT or β-mercaptoethanol may help preserve enzyme activity. Final preparation quality should be assessed through SDS-PAGE, Western blotting, and enzymatic activity assays to confirm identity, purity, and functionality.

What are the optimal conditions for measuring B. henselae pyrD enzymatic activity and inhibition?

Establishing reliable assays for measuring B. henselae pyrD enzymatic activity requires consideration of the enzyme's unique characteristics as a Class 2 DHODH. The standard spectrophotometric assay monitors the conversion of dihydroorotate to orotate by following the reduction of an artificial electron acceptor such as 2,6-dichloroindophenol (DCIP) at 600 nm, or alternatively, using CoQ0 (ubiquinone-0) or CoQ1 with absorbance monitored at 275 nm. The reaction buffer typically contains 50 mM Tris-HCl (pH 8.0), 150 mM KCl, and 0.1% Triton X-100, with all measurements performed at 30°C to maintain enzyme stability while enabling sufficient reaction rates.

Kinetic parameters should be determined by varying substrate concentrations (typically 10-500 μM dihydroorotate) and electron acceptor concentrations (20-100 μM DCIP or 10-100 μM CoQ0/CoQ1). Michaelis-Menten kinetics analysis yields important parameters including Km values for both substrates and the maximal velocity (Vmax), providing insights into the enzyme's catalytic efficiency. The influence of pH and temperature on activity should be systematically investigated to establish optimal assay conditions, with pH profiles typically spanning 6.0-9.0 and temperature ranges from 20-45°C.

For inhibition studies, researchers should employ a standardized protocol where varying concentrations of potential inhibitors are pre-incubated with the enzyme before initiating the reaction by adding substrates. IC50 values are determined using a minimum of 8 inhibitor concentrations spanning at least three orders of magnitude. More detailed mechanistic insights can be obtained through inhibition pattern analysis, determining whether inhibitors compete with dihydroorotate (competitive inhibition), with the quinone (uncompetitive inhibition), or exhibit mixed inhibition patterns.

Alternative assay methods include coupling DHODH activity to cytochrome c reduction or monitoring oxygen consumption using polarographic methods. High-throughput screening adaptations using 384-well plate formats with reduced reaction volumes enable larger-scale inhibitor libraries to be tested. For all assay formats, appropriate controls must be included: no-enzyme controls to account for non-enzymatic background reactions, controls with known DHODH inhibitors such as brequinar or leflunomide derivatives as positive controls, and vehicle controls when testing compounds dissolved in DMSO or other organic solvents.

How can researchers effectively analyze the structure-function relationship of B. henselae pyrD?

Elucidating the structure-function relationship of B. henselae pyrD requires a multidisciplinary approach combining structural biology techniques with functional analysis. X-ray crystallography represents the gold standard for determining the three-dimensional structure of the enzyme. Researchers should optimize crystallization conditions by employing sparse matrix screens with the purified protein, typically at concentrations between 5-15 mg/mL, in the presence of appropriate detergents to maintain solubility. Co-crystallization with substrates, products, or inhibitors can provide additional insights into binding mechanisms and conformational changes associated with catalysis.

When crystallization proves challenging, alternative structural approaches include cryo-electron microscopy (cryo-EM), particularly suitable for membrane-associated proteins, or small-angle X-ray scattering (SAXS) to obtain low-resolution structural information in solution. Homology modeling based on structurally characterized DHODHs from related organisms can provide theoretical structural models that guide experimental design before experimental structures become available. These models should be validated through molecular dynamics simulations to assess stability and identify potentially important dynamic regions.

Site-directed mutagenesis serves as a powerful approach to test hypotheses about specific residues' roles in catalysis or substrate binding. Based on sequence alignments with other DHODHs and structural predictions, researchers should systematically mutate conserved active site residues, potential quinone-binding residues, and membrane-interaction domains. Each mutant should undergo thorough kinetic characterization to determine effects on Km, kcat, and inhibitor susceptibility. Thermal shift assays (differential scanning fluorimetry) can provide insights into how mutations affect protein stability.

Hydrogen-deuterium exchange mass spectrometry (HDX-MS) offers complementary information about protein dynamics and solvent accessibility, potentially revealing regions involved in conformational changes during catalysis. For investigating membrane interactions, techniques such as surface plasmon resonance with model membranes or microscale thermophoresis to study protein-ligand interactions provide valuable insights. Integration of these structural and functional data ultimately yields a comprehensive understanding of how the enzyme's architecture enables its catalytic function and how this might be exploited for therapeutic intervention.

How can B. henselae pyrD be exploited as a potential therapeutic target for Bartonella infections?

The exploitation of B. henselae pyrD as a therapeutic target stems from several advantageous characteristics that position this enzyme as a promising intervention point. Foremost among these is the essential nature of pyrimidine biosynthesis for bacterial replication, particularly for an intracellular pathogen like B. henselae that must compete with host cells for resources while evading immune responses. The structural and functional differences between bacterial Class 2 DHODHs and the human Class 2 DHODH create an opportunity window for selective inhibition, potentially minimizing off-target effects on host pyrimidine metabolism.

Rational drug design approaches should begin with fragment-based screening or virtual screening of compound libraries against structural models or experimentally determined structures of B. henselae pyrD. Particularly promising are compounds that target the quinone-binding site, which differs significantly from the ubiquinone-binding site in human DHODH. Initial hits from these screens should be validated through biochemical assays measuring inhibition constants (Ki) and determining inhibition mechanisms. Structure-activity relationship (SAR) studies with series of related compounds can guide medicinal chemistry optimization efforts to improve potency, selectivity, and pharmacokinetic properties.

The therapeutic potential must be evaluated through a progressive series of experimental models. Initial testing in cell culture systems using B. henselae-infected human endothelial cells or macrophages can establish whether compounds can penetrate host cells and inhibit intracellular bacterial replication. Pharmacokinetic studies should assess absorption, distribution, metabolism, and excretion parameters, with particular attention to whether compounds can reach sufficient concentrations in tissues where Bartonella typically persists, such as the vascular endothelium and lymph nodes . Animal models, such as experimentally infected cats or immunocompromised mice, provide the necessary in vivo validation before consideration for clinical development.

A particular advantage of targeting pyrD lies in the potential for combination therapy approaches. Inhibitors targeting different metabolic pathways might exhibit synergistic effects with reduced likelihood of resistance development. For instance, combining pyrD inhibitors with compounds targeting folate metabolism or protein synthesis could enhance efficacy against persistent infections. The development pipeline should also include resistance studies to understand potential escape mechanisms and proactively design inhibitors less susceptible to resistance development through rational modification of lead compounds.

What are the challenges in studying genetic manipulation of pyrD in B. henselae and potential metabolic engineering applications?

Genetic manipulation of pyrD in Bartonella henselae presents several significant technical challenges that have hindered rapid research progress in this area. B. henselae is a fastidious, slow-growing organism with limited genetic tools compared to model bacteria, requiring specialized media and extended incubation periods of up to 7 days for visible colony formation . The development of effective transformation protocols is complicated by the presence of restriction-modification systems that can degrade introduced DNA. Researchers must typically methylate plasmid DNA before transformation or use conjugation-based methods for DNA transfer, with electroporation parameters requiring extensive optimization for efficient transformation.

Creating targeted pyrD mutations requires specialized genetic tools adapted for Bartonella. The CRISPR-Cas9 system has been adapted for some Bartonella species, though efficiency varies and off-target effects must be carefully controlled. Alternative approaches include allelic exchange vectors containing pyrD variants flanked by homologous regions, combined with selection markers such as antibiotic resistance genes. Conditional expression systems, such as tetracycline-inducible promoters, are particularly valuable for studying essential genes like pyrD, allowing researchers to control expression levels and create depletion strains rather than complete knockouts if the gene proves essential.

Phenotypic analysis of pyrD mutants must address several conceptual challenges. First, as pyrimidine biosynthesis is likely essential, complete loss-of-function mutations may be lethal unless supplemented with pyrimidines or under conditional expression. Second, even partial reduction in pyrD activity may have pleiotropic effects beyond pyrimidine limitation, potentially affecting bacterial membrane potential if quinone pools are altered. Third, the slow growth of B. henselae makes traditional growth-based phenotypic assays time-consuming, necessitating the development of more sensitive readouts such as reporter gene systems or competitive index assays in mixed infections.

From a metabolic engineering perspective, modulating pyrD expression or activity could have applications beyond antimicrobial development. Controlled manipulation of pyrimidine metabolism might enable the development of attenuated strains for vaccine research, alter the production of bacterial factors involved in host cell interaction, or modify B. henselae's resistance to environmental stresses relevant to transmission cycles . As techniques for Bartonella genetic manipulation continue to improve, pyrD represents an important target for understanding fundamental aspects of this pathogen's physiology and host-microbe interactions.

How does pyrD activity in B. henselae correlate with bacterial persistence and adaptation to different environmental conditions?

The correlation between pyrD activity and B. henselae's remarkable environmental persistence represents a complex and fascinating research question with implications for understanding pathogenesis. Bartonella henselae exhibits extraordinary environmental resilience, with research demonstrating its viability in various biological fluids including blood, serum, urine, and milk for extended periods, and even after complete desiccation and subsequent rehydration . This persistence capability suggests sophisticated metabolic adaptation mechanisms that may involve regulated pyrimidine biosynthesis through pyrD activity modulation.

During transitions between mammalian hosts and arthropod vectors, B. henselae experiences dramatic changes in temperature, pH, nutrient availability, and oxidative stress conditions. Experimental approaches to explore pyrD's role in these transitions should include controlled exposure studies measuring enzyme activity, expression levels, and bacterial viability under conditions mimicking these different environments. Quantitative RT-PCR and western blotting can track transcriptional and translational changes in pyrD expression across these conditions, while enzyme activity assays with cellular extracts can determine whether post-translational modifications affect catalytic efficiency in response to environmental stressors.

The connection between pyrD activity and persistent infection states requires investigation through both in vitro and in vivo models. In vitro persistence models using nutrient limitation, antibiotic exposure, or host cell co-culture systems can reveal whether pyrimidine metabolism shifts during transition to non-replicating persistent states. Particularly valuable would be transcriptomic and proteomic profiling comparing acute versus persistent infection models, potentially revealing whether pyrD and related pyrimidine biosynthesis enzymes are differentially regulated during these states. The cat reservoir host model provides an opportunity to study long-term persistence mechanisms, as B. henselae can establish asymptomatic bacteremia lasting months in cats .

The enzyme's dual role in both pyrimidine biosynthesis and electron transport chain interaction positions pyrD at a critical metabolic junction potentially involved in sensing and adapting to environmental changes. Modifications to the quinone pool composition under different oxygen tensions or nutrient conditions could directly affect pyrD activity, creating a regulatory feedback mechanism. Future research directions should explore whether B. henselae possesses pyrD isoforms or post-translational modifications that optimize enzyme function across the diverse environments encountered during its complex life cycle between mammalian hosts and arthropod vectors.

What are the common challenges in expressing and purifying active recombinant B. henselae pyrD and how can they be overcome?

Researchers frequently encounter several challenging issues when attempting to express and purify active recombinant B. henselae pyrD. The membrane association of Class 2 DHODHs often leads to inclusion body formation when the protein is overexpressed in E. coli, resulting in insoluble and non-functional enzyme. This problem can be addressed through multiple strategies, beginning with reducing the expression rate by lowering incubation temperature to 16-20°C and decreasing IPTG concentration to 0.1-0.2 mM. Alternative E. coli strains specifically designed for membrane protein expression, such as C41(DE3) or C43(DE3), often yield better results than standard BL21(DE3) hosts by providing a more accommodating membrane environment for foreign proteins.

Protein solubility issues frequently arise during purification attempts, manifesting as precipitation during extraction or chromatography steps. The selection of appropriate detergents is critical to overcome this challenge, with mild non-ionic detergents like n-dodecyl-β-D-maltoside (DDM) or CHAPS often proving more effective than harsher alternatives like Triton X-100. A systematic detergent screen examining different types and concentrations should be performed to identify optimal solubilization conditions. Additionally, incorporating stabilizing agents such as glycerol (10-15%), specific salts (KCl or NaCl at 100-300 mM), and redox stabilizers like reduced glutathione can significantly improve protein stability throughout the purification process.

Many researchers encounter low enzymatic activity in their purified preparations despite reasonable protein yields. This issue often stems from improper handling of the FMN cofactor essential for DHODH function. Supplementing purification buffers with 10-20 μM FMN helps maintain cofactor association with the enzyme. Flash-freezing in liquid nitrogen rather than slow freezing, and storage at -80°C in small aliquots to avoid freeze-thaw cycles, helps preserve activity. For particularly problematic preparations, reconstitution approaches involving controlled denaturation in mild denaturants followed by refolding in the presence of FMN and appropriate detergent micelles can sometimes rescue activity.

Heterologous expression systems beyond E. coli merit consideration when conventional approaches fail. Insect cell expression systems using baculovirus vectors often provide superior results for membrane-associated proteins, offering a eukaryotic membrane environment that sometimes better accommodates proper folding. Additionally, the creation of fusion constructs with solubility-enhancing partners like maltose-binding protein (MBP) or SUMO, combined with careful design of linker regions and protease cleavage sites for tag removal, represents another strategy to improve soluble expression while maintaining enzyme activity.

How can researchers troubleshoot inconsistent results in B. henselae pyrD enzyme activity assays?

Inconsistent results in B. henselae pyrD enzyme activity assays frequently stem from multiple methodological variables that require systematic troubleshooting. Reagent quality represents a common source of variability, particularly regarding the stability of substrates and electron acceptors. Dihydroorotate should be stored in small aliquots at -80°C to prevent oxidation, while quinone electron acceptors like CoQ0 or CoQ1 require storage under nitrogen or argon to prevent auto-oxidation. Freshly prepared DCIP solutions should be standardized spectrophotometrically before each experiment to account for potential degradation. Establishing standardized protocols for reagent preparation, including defined storage conditions and maximum storage times, helps minimize this source of variability.

Assay component concentrations often contribute to inconsistency, particularly when working near detection limits. Optimizing enzyme concentration is critical—too low concentrations result in poor signal-to-noise ratios, while excessive enzyme can cause rapid substrate depletion and non-linear reaction kinetics. The reaction should be monitored in real-time rather than at single endpoints to ensure linearity throughout the measurement period. Temperature fluctuations during assays significantly impact enzymatic rates, necessitating pre-equilibration of all components and the use of temperature-controlled spectrophotometer cells. Even small variations of 1-2°C can produce measurable differences in activity, especially when working near the enzyme's temperature optimum.

The detergent environment fundamentally affects membrane enzyme activity and represents another common source of inconsistency. Detergent concentrations should always exceed the critical micelle concentration (CMC) but remain below levels that might denature the enzyme. Detergent stocks should be freshly prepared or properly stored to prevent degradation, particularly for easily oxidized detergents. The inclusion of internal standards and reference compounds with established inhibition profiles allows inter-assay normalization and helps identify problematic assay conditions. For particularly challenging compounds, orthogonal assay methods (such as coupling pyrD activity to cytochrome c reduction) can confirm results obtained from the primary assay system.

Statistical validation is essential when troubleshooting variable results. Each experiment should include at least three technical replicates, with key findings confirmed through independent biological replicates using different enzyme preparations. Systematic positive and negative controls should be incorporated into every assay plate or run to establish baseline performance parameters. For high-throughput screening applications, statistical parameters such as Z' factor should be calculated to ensure assay robustness. Careful documentation of all experimental parameters, including buffer composition details, reagent sources and lot numbers, and precise timing of each step enables more effective troubleshooting of variability sources.

What strategies can optimize structural and biophysical studies of B. henselae pyrD for drug discovery applications?

Optimizing structural and biophysical studies of B. henselae pyrD for drug discovery requires strategic approaches addressing the enzyme's membrane association and potential conformational dynamics. Construct design represents a critical first step, with systematic truncation and mutation of the membrane-interacting regions potentially yielding more stable protein variants amenable to crystallization while maintaining catalytic activity. Fusion partners such as T4 lysozyme or thermostabilized apocytochrome b562RIL (BRIL) can sometimes be successfully introduced into loop regions to provide additional crystal contacts without disrupting the active site architecture. High-throughput construct screening using fluorescence-based thermal stability assays can rapidly identify promising candidates from dozens of engineered variants.

Crystallization of membrane-associated proteins often benefits from specialized approaches beyond traditional vapor diffusion methods. Lipidic cubic phase (LCP) crystallization has proven successful for many challenging membrane proteins and may be applicable to pyrD with proper optimization. The addition of specific lipids or the use of bicelles as crystallization facilitators can sometimes induce crystal formation when traditional approaches fail. Fragment-based crystallography approaches, where crystals are soaked with small molecule fragments rather than larger inhibitors, may prove more successful for obtaining co-crystal structures when the protein exhibits poor tolerance for larger compounds.

When crystallization proves particularly challenging, integrative structural biology approaches combining multiple techniques can yield valuable insights. Hydrogen-deuterium exchange mass spectrometry (HDX-MS) offers detailed information about protein dynamics and ligand-induced conformational changes without requiring crystals. This technique can identify stable regions suitable for construct optimization and reveal binding-induced protection patterns that map inhibitor binding sites. Cryo-electron microscopy (cryo-EM) continues to advance for smaller proteins and may soon become viable for proteins in the size range of pyrD, particularly if they can be incorporated into larger assemblies or nanodiscs to increase effective molecular weight.

Biophysical characterization techniques optimized for membrane proteins provide crucial binding data to support drug discovery. Microscale thermophoresis (MST) and surface plasmon resonance (SPR) with properly functionalized sensor chips can generate reliable binding constants for inhibitor series with minimal protein consumption. The thermal shift assay (TSA) approach can be adapted for pyrD by monitoring protein unfolding in the presence of various detergents, buffer conditions, and potential stabilizing ligands. For fragment screening, techniques like saturation transfer difference (STD) NMR or WaterLOGSY can detect weak binding events that might be missed by activity-based assays but represent valuable starting points for medicinal chemistry optimization.

How does B. henselae pyrD activity correlate with bacterial pathogenicity in various disease models?

The relationship between B. henselae pyrD activity and pathogenicity represents a complex interaction requiring multifaceted experimental approaches for thorough characterization. Establishing this correlation begins with the development of conditional expression systems or specific inhibitors that allow controlled modulation of pyrD activity in vivo. Genetic approaches might include the creation of attenuated strains with reduced pyrD expression or activity through mutations in catalytic residues or regulatory elements. These strains can then be evaluated in infection models to assess virulence parameters including bacterial load, dissemination patterns, and host inflammatory responses compared to wild-type bacteria.

Cell culture infection models provide valuable initial insights into pyrD's role in pathogenicity. Human endothelial cells, which represent a primary target for B. henselae during infection, can be infected with wild-type bacteria in the presence of pyrD inhibitors or with genetically modified strains having altered pyrD activity . Key readouts should include bacterial adhesion efficiency, intracellular replication rates, and the induction of angiogenic factors and inflammatory cytokines that characterize B. henselae infection. Macrophage infection models allow assessment of how pyrD activity affects bacterial survival within professional phagocytes, potentially revealing connections between pyrimidine metabolism and resistance to host cellular defenses.

Animal models, though challenging to establish for B. henselae, provide essential in vivo validation of pathogenicity correlations. While cats serve as the natural reservoir host with typically asymptomatic infection, experimental infection in immunocompromised mice can reproduce aspects of human disease . In these models, researchers should assess how pyrD manipulation affects bacterial persistence, tissue tropism, and the development of characteristic lesions such as bacillary angiomatosis in immunocompromised models. Particular attention should be given to bacterial loads in the vasculature, lymph nodes, and spleen, which represent primary sites of B. henselae colonization.

The mechanistic connection between pyrD activity and virulence likely extends beyond simple growth requirements to include potential moonlighting functions or metabolic interactions. Transcriptomic and proteomic profiling comparing wild-type bacteria to pyrD-modulated strains during infection can reveal whether altered pyrimidine metabolism triggers broader virulence program changes. For instance, limited nucleotide availability might serve as a stress signal inducing virulence factor expression or biofilm formation. Additionally, metabolomic analysis might reveal how pyrD activity influences the production of other bacterial metabolites involved in host-pathogen interactions or quorum sensing mechanisms.

How can computational approaches enhance our understanding of B. henselae pyrD function and inhibitor design?

Computational approaches offer powerful tools for enhancing our understanding of B. henselae pyrD and accelerating inhibitor discovery through multiple complementary methodologies. Homology modeling provides the initial foundation when experimental structures are unavailable, utilizing known DHODH structures from related organisms as templates. These models can be refined through molecular dynamics simulations incorporating explicit membrane environments to better capture the enzyme's native context. Advanced sampling techniques such as metadynamics or replica exchange methods enable exploration of conformational states that might not be captured in static crystal structures but are relevant for inhibitor binding and catalytic function.

Structure-based virtual screening represents an efficient approach to identify potential inhibitors from large compound libraries. Molecular docking against the active site and quinone-binding pocket can be performed using software such as Glide, AutoDock Vina, or FRED, with scoring functions optimized for protein-ligand interactions. Ensemble docking against multiple protein conformations derived from molecular dynamics simulations improves the chances of identifying compounds that might bind to transiently exposed pockets. Machine learning approaches increasingly enhance virtual screening by developing customized scoring functions based on known DHODH inhibitors or by directly predicting binding affinities through deep learning architectures trained on protein-ligand interaction data.

Pharmacophore modeling offers a complementary ligand-based approach, particularly valuable when multiple inhibitors with diverse chemical scaffolds have been identified. By extracting essential features required for activity—such as hydrogen bond donors/acceptors, hydrophobic centers, and aromatic rings—researchers can develop pharmacophore hypotheses that guide the design of new compounds with improved properties. Fragment-based approaches in silico, where libraries of small chemical fragments are screened and promising hits are gradually grown or linked to create more potent inhibitors, parallel experimental fragment-based drug discovery efforts while requiring significantly fewer resources.

Quantitative structure-activity relationship (QSAR) models become increasingly valuable as experimental data accumulates on inhibitor series. These statistical models correlate molecular descriptors with biological activity, enabling the prediction of potency for novel compounds before synthesis. Modern QSAR approaches incorporating 3D information and molecular interaction fields provide particularly useful insights for optimizing inhibitor properties. Network pharmacology approaches can place pyrD inhibition in broader context by modeling potential impacts on interconnected metabolic pathways, helping predict synergistic drug combinations or potential resistance mechanisms.

How can integrated multi-omics approaches provide insights into the role of pyrD in B. henselae metabolism and host interaction?

Integrated multi-omics approaches offer comprehensive insights into pyrD's role in Bartonella henselae metabolism and host interactions by capturing system-wide responses across multiple biological levels. Transcriptomic analysis using RNA-seq can reveal how pyrD expression changes under various environmental conditions relevant to the B. henselae lifecycle, including temperature shifts mimicking transition between mammalian hosts (37°C) and arthropod vectors (28°C), pH variations, nutrient limitation, and oxidative stress. Comparing these expression patterns with those of other genes involved in nucleotide metabolism, energy production, and virulence can uncover regulatory networks connecting pyrimidine biosynthesis to broader adaptive responses.

Proteomic approaches complement transcriptomics by addressing post-transcriptional regulation and protein-protein interactions. Mass spectrometry-based proteomics can quantify pyrD protein levels and identify post-translational modifications potentially regulating enzyme activity in response to environmental cues. Protein interaction studies using approaches such as affinity purification coupled with mass spectrometry (AP-MS) or bacterial two-hybrid systems may reveal previously uncharacterized interactions between pyrD and other proteins, potentially uncovering moonlighting functions beyond its established catalytic role. Targeted analysis of the bacterial membrane proteome can provide context for understanding how pyrD integrates with the electron transport chain components.

Metabolomic profiling provides direct evidence of pyrD's impact on cellular metabolism by measuring changes in pyrimidine intermediates, nucleotides, and connected metabolic pathways. Untargeted metabolomics using liquid chromatography-mass spectrometry can characterize global metabolic shifts when pyrD activity is modulated, while targeted approaches can precisely quantify specific compounds like dihydroorotate, orotate, and downstream pyrimidines. Stable isotope labeling experiments using 13C or 15N tracers can map carbon and nitrogen flux through the pyrimidine pathway under different conditions, revealing how B. henselae prioritizes resources during host infection versus environmental persistence.

Integration of these multi-omics datasets requires sophisticated computational approaches. Network analysis can identify co-regulated genes, proteins, and metabolites, revealing functional modules connecting pyrD to other cellular processes. Machine learning approaches can identify patterns across datasets that might not be apparent through conventional analysis. Most importantly, multi-omics integration during host-pathogen interaction studies can reveal how pyrD activity affects both bacterial and host metabolism simultaneously. This dual perspective is particularly relevant for intracellular pathogens like B. henselae, where bacterial metabolism directly interfaces with and potentially manipulates host metabolic pathways during infection .

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.