Recombinant Leptospira interrogans serogroup Icterohaemorrhagiae serovar copenhageni Uridylate kinase (pyrH)

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 purchasing method and location. Contact your local distributor for precise delivery estimates.
Note: Standard shipping includes 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. 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 serves 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
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 the production process. If a specific tag type is required, please inform us for preferential development.
Synonyms
pyrH; LIC_10852; Uridylate kinase; UK; EC 2.7.4.22; Uridine monophosphate kinase; UMP kinase; UMPK
Buffer Before Lyophilization
Tris/PBS-based buffer, 6% Trehalose.
Datasheet
Please contact us to get it.
Expression Region
1-248
Protein Length
full length protein
Purity
>85% (SDS-PAGE)
Species
Leptospira interrogans serogroup Icterohaemorrhagiae serovar copenhageni (strain Fiocruz L1-130)
Target Names
pyrH
Target Protein Sequence
MATEAKYNRI LIKLSGEALA GEGEFGIDTN KAHSLAEEIK EVHDLGVEIA LVVGGGNIIR GTNLAKAGID RATADYMGML ATIQNALALQ DACEKKGLYT RVQSAIEINS IAESYIRRRA VRHLEKRRIV IFAGGTGNPY FTTDTTASLR AVEVGCDVIL KATKVDGVYT ADPKKDNGAK RYSQISFMES INRRLKVMDS TALSLCMENN MSIIVFDIFK RGNLKDLVTG KNIGTLISNS EDIQIDGK
Uniprot No.

Target Background

Function
Catalyzes the reversible phosphorylation of UMP to UDP.
Database Links
Protein Families
UMP kinase family
Subcellular Location
Cytoplasm.

Q&A

What is Leptospira interrogans and why is it significant for research?

Leptospira interrogans is a species of obligate aerobic spirochaete bacteria with a distinctive corkscrew shape featuring hooked and spiral ends. It belongs to the spirochaete phylum and is a significant pathogen causing leptospirosis, a severe zoonotic disease affecting both humans and animals. L. interrogans is particularly prevalent in tropical and temperate regions, capable of surviving for weeks to months in soil or water .

The significance of L. interrogans for research stems from:

  • Its role as a causative agent of leptospirosis, which affects over a million people annually and causes approximately 60,000 deaths worldwide

  • Its complex pathogenesis involving two distinct phases of infection (anicteric and icteric)

  • The challenges in diagnosis due to symptoms resembling other conditions such as influenza, dengue, and viral hemorrhagic diseases

  • Its potential as a target for vaccine development, with over 200 pathogenic serovars complicating effective vaccine creation

How does Leptospira interrogans cause disease in humans and animals?

Leptospira interrogans causes leptospirosis through a complex infection process:

  • Transmission route: Humans typically acquire the bacteria through:

    • Direct contact with urine from infected animals

    • Exposure to urine-contaminated water, soil, or food

    • Entry through skin abrasions or mucous membranes (eyes, nose)

  • Biphasic disease progression:

    • Phase 1 (Anicteric/Septicemic): Characterized by fever, headache, myalgia, and nausea, lasting 4-9 days

    • Brief asymptomatic period

    • Phase 2 (Immune/Icteric): More severe symptoms including hemorrhages, jaundice, renal tubular failure, and potentially meningitis

  • Pathogenic mechanisms:

    • The bacteria utilizes two periplasmic flagella for movement and tissue penetration

    • Leptospira damages endothelial cell linings, allowing bacterial spread to various organs

    • The organism primarily targets the kidneys, where it multiplies optimally

    • The bacteria can evade innate immune responses; human TLR4 cannot recognize leptospiral LPS, unlike mouse TLR4, explaining species differences in susceptibility

    • In severe cases, patients experience a "cytokine storm" with elevated IL-6, TNF-alpha, and other inflammatory markers

The severity of disease depends on both the infecting serovar and host factors, with fatality rates increasing with age and reaching up to 20% in patients with jaundice or kidney damage .

What are the current diagnostic methods for leptospirosis?

Current diagnostic methods for leptospirosis include:

  • Microscopic Agglutination Test (MAT):

    • Considered the gold standard by the WHO

    • Detects antibodies against Leptospira

    • Limitations: Low sensitivity during early disease, technically demanding, requires live cultures and specialized expertise

  • PCR-based detection:

    • Effective during the first 8 days of fever before antibody formation

    • Can detect leptospiremia levels as high as 10^6/ml of blood

    • Provides more rapid results than culture methods

  • Culture methods:

    • Isolation of bacteria from blood, spinal fluid, or urine

    • Time-consuming process requiring specialized media

    • Most reliable during early infection stage

  • Serological tests:

    • Rising Leptospira antibody levels indicate infection

    • Include ELISA and other immunoassays

    • Most effective during convalescent phase

  • Emerging approaches:

    • Recombinant chimeric proteins comprising multiple leptospiral epitopes

    • The rChi2 protein (containing 10 conserved leptospiral surface antigens) shows promise for early detection, recognizing antibodies in 75% of early (MAT-negative) samples and 82% of convalescent (MAT-positive) samples

How is recombinant pyrH expressed and purified for research applications?

The expression and purification of recombinant pyrH from Leptospira interrogans involves several key methodological considerations:

Expression Systems:

  • Yeast expression system: The commercial recombinant pyrH (CSB-YP744676LEV) is expressed in yeast, which provides eukaryotic post-translational modifications that may be beneficial for certain applications .

  • E. coli expression: Many recombinant leptospiral proteins are expressed in E. coli as fusion proteins with tags such as:

    • Glutathione-S-transferase (GST) - Commonly used for leptospiral recombinant proteins like LigA

    • His-tag - Facilitates purification via metal affinity chromatography

Purification Protocol:

  • Initial clarification of lysate via centrifugation

  • Affinity chromatography using the appropriate resin (glutathione-agarose for GST-tagged proteins)

  • Ion exchange chromatography for further purification

  • Size exclusion chromatography for final polishing

  • Verification of purity via SDS-PAGE (>85% purity is typically desired)

Quality Control Metrics:

  • Protein concentration determination

  • SDS-PAGE to assess purity

  • Western blotting to confirm identity

  • Enzymatic activity assays to verify functionality

  • Endotoxin testing for applications requiring endotoxin-free preparations

Storage Considerations:

  • Lyophilized form maintains stability for up to 12 months at -20°C/-80°C

  • Liquid forms typically remain stable for 6 months at -20°C/-80°C

  • Addition of 5-50% glycerol as a cryoprotectant is recommended for liquid storage

What role does pyrH play in Leptospira pathogenesis during host infection?

Recent multi-omics studies reveal significant insights into pyrH's role during Leptospira pathogenesis:

  • Differential expression during infection:

    • Transcriptomic analysis shows pyrH (LA_3296) is significantly upregulated in Leptospira interrogans during interaction with human macrophages

    • Fold change values: 3.52 (transcript level) and 2.90 (protein level)

    • This upregulation suggests pyrH plays an important role during the host-pathogen interaction phase

  • Metabolic adaptation during infection:

    • As an essential enzyme in nucleotide metabolism, increased pyrH expression likely reflects the bacteria's adaptation to the intracellular environment

    • Enhanced nucleotide metabolism may support the increased replication and energetic demands during infection

  • Potential role in immune evasion:

    • Leptospira can survive within macrophages despite being phagocytosed

    • The upregulation of metabolic enzymes like pyrH may contribute to this survival mechanism

    • Altered metabolic states may help the bacteria evade host immune responses

  • Relationship to virulence factors:

    • While not directly identified as a virulence factor like the loa22 gene, pyrH functions as part of the metabolic network supporting virulence

    • Its significant expression changes during infection suggest it may be part of a coordinated response to host immunological pressures

The integrated multi-omics approach has helped identify pyrH as one of the critical molecular factors involved in Leptospira pathogenesis, highlighting its potential as a research target for understanding disease mechanisms .

What are the potential applications of recombinant pyrH in vaccine development against leptospirosis?

While recombinant pyrH itself has not been extensively studied as a vaccine candidate, its potential can be evaluated in context of other recombinant protein approaches for leptospirosis vaccines:

Current Recombinant Vaccine Approaches:

  • LigA and LigB proteins: Recombinant leptospiral immunoglobulin-like proteins have shown promising results:

    • rLigA provided complete protection in hamster challenge models

    • Vaccinated animals showed no significant histopathological changes after challenge

    • All vaccinated animals developed strong antibody responses

  • Multi-epitope chimeric proteins: The rChi2 approach demonstrates how multiple conserved leptospiral antigens can be combined:

    • Includes 10 conserved leptospiral surface antigens

    • Elicits strong humoral response in hamsters

    • Antibodies recognize multiple Leptospira species

Potential of pyrH as a vaccine component:

  • Conserved nature: As a metabolic enzyme, pyrH sequences are likely conserved across Leptospira serovars, potentially offering broader protection than serovar-specific antigens

  • Upregulation during infection: The significant increase in pyrH expression during host infection suggests the protein may be accessible to the immune system during infection

  • Combination approaches: Rather than a standalone vaccine, pyrH might be more valuable as part of a multi-component vaccine:

    • Could be incorporated into chimeric constructs with established immunoprotective proteins

    • Might complement outer membrane proteins with proven vaccine efficacy

  • Limitations to consider:

    • As an enzymatic protein rather than a surface-exposed antigen, immunogenicity may be limited

    • Metabolic enzymes might not elicit strong protective responses compared to outer membrane proteins

Research on other metabolic enzymes as vaccine components suggests that pyrH would likely need to be combined with adjuvants and/or other antigenic components to provide meaningful protection against leptospirosis.

How can recombinant pyrH be used to study Leptospira metabolism and potential drug targets?

Recombinant pyrH provides a valuable tool for investigating Leptospira metabolism and identifying potential therapeutic interventions:

Metabolic Studies:

  • Enzymatic characterization:

    • Determination of kinetic parameters (Km, Vmax, kcat) specific to Leptospira pyrH

    • Comparison with homologous enzymes from other bacteria to identify unique properties

    • Assessment of substrate specificities and cofactor requirements

  • Metabolic network analysis:

    • Using purified pyrH to determine its interactions with other metabolic enzymes

    • Mapping the pyrimidine biosynthesis pathway in Leptospira

    • Understanding how pyrH activity connects to other metabolic pathways

  • Growth condition adaptation:

    • Examining pyrH activity under various environmental conditions mimicking different infection stages

    • Determining how pyrH contributes to Leptospira's ability to survive in diverse environments

Drug Discovery Applications:

  • High-throughput screening platform:

    • Developing assays using recombinant pyrH to screen compound libraries

    • Identifying small molecule inhibitors that selectively target leptospiral pyrH

    • Biochemical assays measuring UDP production or ATP consumption as readouts

  • Structure-based drug design:

    • Using purified recombinant pyrH for crystallization and structural determination

    • Virtual screening against the active site

    • Fragment-based drug discovery approaches

  • Validation studies:

    • Confirmed pyrH inhibitors can be tested against live Leptospira cultures

    • Correlation between enzyme inhibition and bacterial growth inhibition

    • Assessment of selectivity by comparing effects on mammalian homologs

Methodological Approach Table:

ApplicationMethodologyKey MeasurementsChallenges
Enzyme KineticsSpectrophotometric assaysKm, Vmax, inhibition constantsAssay optimization for pyrH-specific conditions
Thermal StabilityDifferential scanning fluorimetryMelting temperature (Tm)Protein stability during purification
Inhibitor ScreeningNADH-coupled enzymatic assaysIC50 valuesDeveloping selective inhibitors
Structural StudiesX-ray crystallography, cryo-EM3D structure at atomic resolutionObtaining diffraction-quality crystals
In vivo ValidationGrowth inhibition assaysMIC valuesTranslation from enzyme to whole-cell activity

The significant upregulation of pyrH during macrophage infection (3.52-fold transcript increase, 2.90-fold protein increase) suggests that targeting this enzyme might be particularly effective against the intracellular stage of Leptospira infection.

What are the optimal conditions for measuring the enzymatic activity of recombinant Leptospira pyrH?

Establishing optimal conditions for measuring Leptospira pyrH activity requires careful consideration of several parameters aligned with the bacterium's physiological environment:

Buffer Composition and Reaction Conditions:

  • pH optimization:

    • Initial screening should center around pH 7.2-7.6, the optimal growth range for Leptospira interrogans

    • Phosphate, HEPES, or Tris buffers at 50-100 mM are commonly used

    • A systematic pH screen (pH 6.5-8.5) should be conducted to determine the enzyme's pH optimum

  • Temperature considerations:

    • Primary assays should be conducted at 28-30°C, matching Leptospira's optimal growth temperature

    • Temperature stability profiles should be established (20-40°C range)

    • For kinetic studies, temperature must be precisely controlled

  • Metal ion requirements:

    • Mg²⁺ (typically 5-10 mM) is essential as a cofactor for ATP binding

    • Other divalent cations (Mn²⁺, Ca²⁺) should be tested at 1-5 mM concentrations

    • EDTA controls should be included to confirm metal dependency

  • Salt concentration:

    • NaCl in the range of 50-150 mM, with attention to the fact that high salt (120 mM NaCl) affects protein expression in Leptospira

Substrate Considerations:

  • ATP concentration: 0.5-5 mM range, with 2 mM as a starting point

  • UMP concentration: 0.1-2 mM range for determining Km values

  • Substrate stock preparation: Fresh preparation recommended to avoid degradation

Assay Methodologies:

Assay TypePrincipleAdvantagesLimitations
Coupled Enzyme AssayLink UDP production to NADH oxidation via pyruvate kinase and lactate dehydrogenaseContinuous monitoring, high sensitivityPotential interference from coupling enzymes
Direct ADP MeasurementLuminescent detection of ADP productionSimple, direct measurementEndpoint assay, expensive reagents
Radiometric AssayUsing [γ-³²P]ATP to detect transfer to UMPHighly sensitive, direct product detectionRequires radioactive materials, specialized equipment
HPLC AnalysisDirect separation and quantification of substrates and productsDefinitive product identificationLower throughput, specialized equipment

Data Analysis Guidelines:

  • Initial velocity measurements should be determined from the linear portion of progress curves

  • Michaelis-Menten parameters should be calculated using non-linear regression

  • Controls must include no-enzyme, no-substrate, and heat-inactivated enzyme samples

Specific Considerations for Leptospira pyrH:

  • Considering Leptospira's metabolism relies on beta-oxidation of long-chain fatty acids , assess whether these metabolites have any regulatory effects on pyrH activity

  • The optimal activity should reflect the microaerophilic nature of the organism, so oxygen concentration may need to be controlled

What challenges exist in producing functional recombinant pyrH and how can they be overcome?

Producing functional recombinant Leptospira pyrH presents several challenges that require specific methodological solutions:

Challenge 1: Ensuring Proper Folding

  • Problem: Bacterial metabolic enzymes may fold improperly in heterologous expression systems.

  • Solutions:

    • Expression temperature optimization: Lower temperatures (16-20°C) slow protein synthesis, allowing more time for proper folding

    • Co-expression with chaperones: GroEL/GroES or DnaK/DnaJ/GrpE systems can improve folding

    • Fusion partners: Solubility-enhancing tags like MBP (maltose-binding protein) often improve folding compared to smaller tags like His6

    • Testing multiple expression hosts: Besides E. coli, consider yeast-based systems as used in commercial production

Challenge 2: Protein Solubility

  • Problem: Recombinant proteins often form inclusion bodies, particularly at high expression levels.

  • Solutions:

    • Optimization of induction conditions: Lower IPTG concentrations (0.1-0.5 mM) and OD600 at induction (0.4-0.8)

    • Buffer optimization: Including osmolytes (glycerol 5-10%, sucrose), mild detergents, or arginine in lysis buffers

    • Refolding protocols: If inclusion bodies form, optimized refolding procedures using gradual dialysis

    • Cell-free expression systems: Alternative for difficult-to-express proteins

Challenge 3: Maintaining Enzymatic Activity

  • Problem: Purification procedures may compromise the native structure and activity.

  • Solutions:

    • Stabilizing additives: Including glycerol (10-20%), reducing agents (DTT or β-mercaptoethanol), and specific metal ions

    • Gentle purification methods: Avoiding harsh elution conditions and extreme pH

    • Activity assays at each purification step: Monitoring activity retention throughout purification

    • Minimizing freeze-thaw cycles: Aliquoting final product and limiting freeze-thaw to preserve activity

Challenge 4: Removing Contaminating Activities

  • Problem: Host-derived enzymes with similar functions may contaminate preparations.

  • Solutions:

    • Multi-step purification strategy: Combining affinity, ion exchange, and size exclusion chromatography

    • Specific activity measurements: Calculating units of activity per mg protein to track purification progress

    • Western blot analysis: Confirming identity and purity using specific antibodies

    • Mass spectrometry analysis: Verifying protein identity and detecting contaminants

Experimental Design Table: Optimization Matrix for Recombinant pyrH Production

ParameterVariables to TestEvaluation MethodSuccess Criteria
Expression HostE. coli BL21(DE3), Rosetta, Arctic Express; YeastSDS-PAGE, Western blot>30% soluble expression
Induction Temperature37°C, 30°C, 25°C, 16°CSolubility comparison>50% in soluble fraction
Induction Time3h, 6h, 16h, 24hActivity assay, yieldOptimal activity/yield ratio
Purification StrategyIMAC, Ion exchange, Size exclusion combinationsPurity, yield, activity>85% purity, >50% activity retention
Storage ConditionsVarious buffers with/without glycerol, -20°C vs -80°CActivity retention over time<20% activity loss after 3 months

The experience with other leptospiral recombinant proteins suggests that yeast expression systems (as used for the commercial product ) may offer advantages for pyrH production, particularly for applications requiring post-translational modifications or enhanced solubility.

How can recombinant pyrH be incorporated into high-throughput screening assays for antimicrobial discovery?

Developing high-throughput screening (HTS) assays using recombinant Leptospira pyrH creates opportunities for discovering novel antimicrobials targeting this essential pathway. A comprehensive methodological approach includes:

Assay Development and Validation:

  • Primary Assay Optimization:

    • Coupled enzyme system: Link pyrH activity (UDP production) to NADH oxidation via pyruvate kinase and lactate dehydrogenase

    • Fluorescence-based detection: ATP consumption can be monitored using ATP-sensitive fluorescent probes

    • Luminescence readout: ADP formation measured using commercially available luciferase-based kits

    • Optimal 384-well plate format to maximize throughput while maintaining signal reliability

  • Assay Quality Metrics:

    • Z'-factor optimization to >0.7 for robust screening

    • Signal-to-background ratio >5:1

    • Coefficient of variation <10% for high reproducibility

    • DMSO tolerance evaluation (typically up to 1-2%)

  • Controls Integration:

    • Positive controls: Known nucleotide kinase inhibitors

    • Negative controls: Reaction mixture with inactive enzyme or missing substrate

    • Vehicle controls: Buffer with appropriate DMSO concentration

Screening Implementation Protocol:

StageMethodologyCriteria for ProgressionTechnical Considerations
Primary ScreenSingle concentration (10-20 μM), single replicate>50% inhibition1-5% hit rate target
Dose-Response Confirmation8-10 point dose-response curvesIC50 <10 μM, Hill slope 0.8-1.2Eliminate compounds with irregular curves
Counter ScreenActivity against mammalian UMP/CMP kinaseSelectivity index >10Ensure target specificity
Orthogonal AssayAlternative detection method (e.g., HPLC)Confirmation of mechanismEliminate false positives from primary assay
Whole-Cell ActivityGrowth inhibition of Leptospira culturesMIC <50 μMVerify cellular penetration and efficacy

Specialized Considerations for pyrH-Based Screening:

  • Fragment-Based Approaches:

    • Thermal shift assays to identify fragments that bind and stabilize pyrH

    • NMR-based fragment screening for detecting weak binders

    • Crystallographic screening for fragment binding sites

  • Computational Pre-Screening:

    • Homology modeling of Leptospira pyrH if crystal structure unavailable

    • Virtual screening against the ATP-binding and UMP-binding sites

    • Pharmacophore models based on known kinase inhibitors

  • Multiplexed Approaches:

    • Simultaneous screening against multiple leptospiral targets including pyrH

    • Phenotypic screening followed by target identification

    • Pathway-based screening looking at pyrimidine metabolism disruption

  • Advanced Data Analysis:

    • Machine learning algorithms to identify structure-activity relationships

    • Clustering of active compounds by chemical scaffold

    • Network analysis to identify synergistic target combinations

The significant upregulation of pyrH during macrophage infection (3.52-fold increase in transcription, 2.90-fold increase in protein expression) suggests that inhibitors identified through this screening cascade may be particularly effective during the intracellular phase of Leptospira infection, addressing a critical need in leptospirosis treatment.

What techniques are most effective for studying the interaction between recombinant pyrH and other components of the pyrimidine biosynthesis pathway?

Investigating the interactions between recombinant Leptospira pyrH and other components of the pyrimidine biosynthesis pathway requires a multi-faceted methodological approach:

Protein-Protein Interaction Analysis:

  • Pull-down Assays:

    • Immobilize tagged recombinant pyrH on appropriate resin

    • Incubate with Leptospira lysate or purified pathway components

    • Analyze interacting partners by mass spectrometry

    • Confirmation with reverse pull-down using identified partners

  • Surface Plasmon Resonance (SPR):

    • Quantitative measurement of binding kinetics (kon, koff, KD)

    • Direct observation of real-time interactions

    • No labeling required, minimizing interference

    • Protocol parameters:

      • Immobilization: 2000-5000 RU of pyrH on CM5 chip

      • Analyte concentration: 0.1-10x expected KD

      • Flow rate: 20-50 μL/min

      • Temperature: 25°C (standard) or 30°C (Leptospira optimal growth temperature)

  • Microscale Thermophoresis (MST):

    • Detects interactions in solution with minimal sample consumption

    • Suitable for membrane-associated components of the pathway

    • Can detect subtle conformational changes upon binding

Enzyme Complex Formation and Activity:

  • Enzyme Cascade Reconstitution:

    • Sequential addition of purified enzymes from the pathway

    • Monitoring reaction progression through multiple steps

    • Comparing kinetic parameters of isolated vs. complexed enzymes

  • Analytical Ultracentrifugation:

    • Detecting complex formation through sedimentation velocity experiments

    • Determining stoichiometry of multi-enzyme complexes

    • Assessing stability of complexes under varying conditions

  • Native PAGE and Blue Native PAGE:

    • Preserves non-covalent interactions during electrophoresis

    • Identifies stable complexes formed between pathway components

    • In-gel activity assays to confirm functional complex formation

Structural Analysis of Interactions:

TechniqueApplicationResolutionAdvantagesLimitations
X-ray CrystallographyHigh-resolution structures of pyrH in complex with pathway partners1-3 ÅAtomic detail of interaction interfacesRequires crystal formation
Cryo-Electron MicroscopyVisualization of larger assemblies2.5-4 ÅMinimal sample preparation, captures multiple statesEquipment accessibility
Hydrogen-Deuterium Exchange MSMapping interaction surfacesPeptide levelDetects conformational changes upon bindingIndirect structural information
SAXS/SANSLow-resolution envelope of complexes in solution10-20 ÅNative conditions, flexible systemsLimited resolution

Integration with Metabolic Pathway Analysis:

  • Metabolic Flux Analysis:

    • Isotope labeling to trace pyrimidine precursors through the pathway

    • Comparing flux with and without functional pyrH

    • Identifying rate-limiting steps and regulatory points

  • In silico Pathway Modeling:

    • Creating computational models of the pyrimidine pathway

    • Predicting effects of pyrH inhibition or enhancement

    • Simulating pathway behavior under different conditions

  • Spatial Organization Studies:

    • Fluorescence microscopy with tagged pathway components

    • Colocalization analysis in fixed or live Leptospira cells

    • Assessment of potential metabolon formation

Understanding these interactions is particularly relevant given Leptospira's unique metabolic adaptations compared to other bacteria, including its ability to synthesize protoheme and cobalamin de novo and its dependence on beta-oxidation of long-chain fatty acids for energy .

How can comparative genomics approaches help understand pyrH evolution and function across Leptospira species?

Comparative genomics provides powerful insights into the evolution and functional conservation of pyrH across Leptospira species and strains, with important implications for research and therapeutic development:

Methodological Approach to pyrH Comparative Analysis:

  • Sequence-based Analysis:

    • Multiple sequence alignment of pyrH genes from diverse Leptospira species and serovars

    • Calculation of nucleotide and amino acid conservation metrics

    • Identification of conserved functional domains versus variable regions

    • Phylogenetic tree construction to trace evolutionary relationships

  • Structural Prediction and Comparison:

    • Homology modeling based on known bacterial UMP kinase structures

    • Structural alignment to identify conservation of catalytic sites

    • Analysis of surface properties and electrostatic potential differences

    • Prediction of species-specific structural features

  • Genomic Context Analysis:

    • Examination of pyrH gene neighborhood across species

    • Identification of conserved operon structures

    • Detection of potential horizontal gene transfer events

    • Analysis of promoter regions and regulatory elements

Key Research Applications:

  • Species Identification and Typing:

    • Development of pyrH-based PCR assays for Leptospira species identification

    • Analysis of pyrH sequence variation as a potential typing marker

    • Correlation of pyrH variants with virulence or host specificity

  • Functional Evolution Understanding:

    • Identification of pyrH sequence signatures associated with:

      • Pathogenic vs. saprophytic Leptospira species

      • Host adaptation in different reservoir animals

      • Environmental persistence capabilities

    • Correlation of pyrH mutations with growth characteristics and metabolic capabilities

  • Drug Target Validation:

    • Identification of highly conserved regions as potential broad-spectrum targets

    • Prediction of potential resistance mechanisms based on natural variants

    • Design of inhibitors targeting conserved active sites

Analytical Framework Example:

Analysis LevelComparison MetricsExpected OutcomesResearch Applications
Sequence ConservationdN/dS ratios, Shannon entropyIdentification of selection pressure regionsTarget druggable conserved regions
Structural MappingRMSD of predicted structures, binding pocket conservationActive site comparison across speciesStructure-based drug design
Regulatory ElementsPromoter motif detection, transcription factor binding sitesDifferences in expression controlUnderstanding strain-specific expression
Horizontal Gene TransferAnomalous GC content, phylogenetic incongruenceDetermination if pyrH underwent lateral transferEvolutionary history reconstruction

Advanced Integration with Multi-omics Data:

  • Transcriptomic Integration:

    • Comparing pyrH expression patterns across species under similar conditions

    • Correlating sequence variations with expression differences

    • Identifying species-specific regulatory mechanisms

  • Proteomic Correlation:

    • Comparing post-translational modifications of pyrH across species

    • Relating protein abundance to genomic features

    • Identifying species-specific interaction partners

  • Metabolomic Context:

    • Assessing species differences in pyrimidine metabolism

    • Correlating pyrH sequence variations with metabolite profiles

    • Mapping pyrH evolutionary changes to metabolic network adaptations

The significant upregulation of pyrH during macrophage infection (3.52-fold transcript increase, 2.90-fold protein increase) makes understanding its evolutionary conservation particularly relevant for investigating pathogenesis mechanisms across the Leptospira genus.

What role might pyrH play in Leptospira's adaptation to different environmental conditions and host tissues?

The adaptation of Leptospira to diverse environmental niches and host tissues likely involves metabolic reprogramming, with pyrH potentially serving as a key regulatory node:

Environmental Adaptation Mechanisms:

Host Tissue Adaptation:

  • Kidney Colonization Mechanisms:

    • Leptospira optimally survives and multiplies in kidneys

    • Research approach: Compare pyrH expression in Leptospira isolated from different tissues in animal models

    • Expected pattern: Potentially higher pyrH expression in kidney-adapted Leptospira

  • Immune Evasion Correlation:

    • Leptospira can survive within macrophages despite phagocytosis

    • Experimental methodology: Monitor pyrH expression during different stages of macrophage infection

    • Key finding: Significant upregulation (3.52-fold transcript increase, 2.90-fold protein increase) during macrophage interaction

  • Persistence in Chronic Carriers:

    • Infected animals can excrete Leptospira for years

    • Research design: Compare genomic and expression profiles of pyrH between acute infection isolates and chronic carrier isolates

    • Hypothesis: Metabolic adaptations including pyrH regulation may contribute to long-term persistence

Integrated Analytical Framework:

Adaptation ContextExperimental ApproachMeasurementsExpected Insights
Temperature StressGrowth at 20-42°CpyrH expression, enzyme kinetics at different temperaturesTemperature-dependent regulation mechanisms
Oxygen LimitationAerobic vs. microaerobic cultureMetabolic flux analysis, pyrH activity correlationRole in adapting to oxygen-limited environments
Host Immune ResponseExposure to oxidative stressROS effects on pyrH expression and activityConnection to stress response pathways
Biofilm FormationComparison of planktonic vs. biofilm growthSpatial expression patternsRole in adaptation to surface-attached lifestyle

Methodological Considerations for Tissue-Specific Analysis:

  • Laser Capture Microdissection:

    • Isolation of Leptospira from specific infected tissue microenvironments

    • Single-cell or small population RNA analysis

    • Correlation of pyrH expression with tissue-specific markers

  • In vivo Expression Technology:

    • Construction of pyrH promoter reporter fusions

    • Monitoring expression during different infection stages

    • Identification of environmental cues triggering expression changes

  • Metabolic Labeling:

    • Isotope-labeled nucleotide precursors to track pyrH activity in different niches

    • Measurement of nucleotide synthesis rates in different environmental conditions

    • Correlation with bacterial replication rates

Understanding these adaptation mechanisms is crucial given Leptospira's remarkable ability to transition between environmental persistence (weeks to months in soil/water) and infection of mammalian hosts , with pyrH potentially serving as a metabolic switch during these transitions.

How can systems biology approaches integrate pyrH function into the broader understanding of Leptospira pathogenesis?

Systems biology provides a framework to position pyrH within the complex network of interactions that drive Leptospira pathogenesis, offering a more comprehensive understanding of disease mechanisms:

Multi-omics Data Integration:

Key Research Methodologies:

ApproachTechniqueApplication to pyrHExpected Outcome
Network AnalysisWeighted Gene Co-expression Network Analysis (WGCNA)Identify genes with expression patterns correlated with pyrHGene modules functionally linked to pyrH
Computational ModelingConstraint-based flux balance analysisModel effects of pyrH perturbation on metabolic networkPrediction of system-wide effects of pyrH inhibition
Multi-factorial DesignCombinatorial environmental conditionsMeasure pyrH response across multiple variablesMap of pyrH regulation in complex environments
Host-Pathogen InteractomeDual RNA-seq of infected tissuesCorrelate pyrH expression with host response genesIdentification of host factors influencing pyrH

Systems-Level Experimental Design:

  • Genome-Scale Knockout/Knockdown Screening:

    • CRISPR interference targeting pyrH and related pathways

    • Phenotypic profiling under infection-relevant conditions

    • Identification of synthetic lethal interactions

  • Metabolic Control Analysis:

    • Measure control coefficients for pyrH in pyrimidine biosynthesis

    • Determine flux control distribution across pathway enzymes

    • Identify rate-limiting steps in nucleotide metabolism during infection

  • Signaling Pathway Integration:

    • Map connections between stress response pathways and pyrH regulation

    • Identify second messengers affecting pyrH expression/activity

    • Correlate with virulence factor expression patterns

Advanced Data Integration Framework:

  • Multi-layer Network Visualization:

    • Integrate proteomic, transcriptomic, and metabolomic data layers

    • Position pyrH within hierarchical regulation networks

    • Identify regulatory motifs and feedback loops involving pyrH

  • Machine Learning Applications:

    • Train predictive models using multi-omics data to predict pyrH behavior

    • Feature importance analysis to identify key determinants of pyrH function

    • Classification of infection states based on pyrH-associated network signatures

The significant upregulation of pyrH during macrophage interaction (3.52-fold transcript increase, 2.90-fold protein increase) places it among the important factors in host-pathogen interactions, with systems biology approaches helping to contextualize this within broader virulence mechanisms.

Specific Insights from Multi-omics Data:

Integrating the finding that pyrH is significantly upregulated during macrophage infection alongside other differentially expressed factors provides a systems-level view of how Leptospira adapts to intracellular environments. This includes coordinated changes in multiple metabolic pathways, stress responses, and virulence mechanisms that collectively contribute to pathogen survival and dissemination.

What emerging technologies might enhance our understanding of pyrH's role in Leptospira biology?

Several cutting-edge technologies show promise for advancing our understanding of pyrH's role in Leptospira biology and pathogenesis:

Advanced Molecular Technologies:

  • CRISPR-based Approaches:

    • Development of CRISPR interference (CRISPRi) systems for Leptospira

    • Tunable repression of pyrH expression to determine threshold effects

    • CRISPRa (activation) to study effects of pyrH overexpression

    • Precise genome editing to create point mutations in catalytic domains

  • Single-Cell Technologies:

    • Single-cell RNA sequencing of Leptospira populations during infection

    • Analysis of cell-to-cell variation in pyrH expression

    • Correlation with other metabolic genes to identify coordinated responses

    • Spatial transcriptomics to map expression in different microenvironments

  • Protein Engineering Approaches:

    • Development of pyrH activity biosensors

    • Real-time monitoring of nucleotide metabolism during infection

    • Structure-guided design of conditionally active pyrH variants

    • Optogenetic control of pyrH activity to study temporal effects

High-Resolution Imaging Techniques:

  • Super-resolution Microscopy:

    • Nanoscale visualization of pyrH localization within Leptospira cells

    • Co-localization with other pyrimidine pathway enzymes

    • Detection of potential metabolon formation under different conditions

    • Correlative light and electron microscopy to link ultrastructure to function

  • Live-Cell Imaging:

    • Fluorescent reporter fusions to monitor pyrH expression dynamics

    • Real-time tracking during host cell invasion and intracellular survival

    • FRET-based biosensors to detect pyrH-substrate interactions

    • Intravital microscopy to observe expression in animal models

Advanced Computational Approaches:

ApproachApplication to pyrH ResearchExpected InsightsImplementation Timeline
AI-powered Protein Structure PredictionGenerate high-confidence Leptospira pyrH structuresNovel structural features, species-specific binding sitesNear-term (1-2 years)
Molecular Dynamics SimulationsModel pyrH conformational changes during catalysisDynamic aspects of enzyme function, allosteric regulationMedium-term (2-3 years)
Deep Learning for Multi-omics IntegrationIdentify patterns in pyrH regulation across datasetsNovel regulatory relationships, condition-specific functionsMedium-term (2-3 years)
Quantum Computing for Drug DesignScreen vast compound libraries against pyrHNovel inhibitor scaffolds with higher specificityLong-term (5+ years)

Next-Generation Functional Genomics:

  • Transposon Sequencing (Tn-seq):

    • Genome-wide fitness profiling under conditions requiring pyrH function

    • Identification of genetic interactions and synthetic lethal partners

    • Mapping of compensatory pathways that become essential when pyrH is compromised

  • CRISPR Screening Approaches:

    • Pooled CRISPRi screens targeting metabolism genes

    • Identification of genes that modify pyrH-dependent phenotypes

    • Discovery of condition-specific genetic interactions

  • Spatial Multi-omics:

    • Integration of spatial transcriptomics, proteomics, and metabolomics

    • Mapping pyrH activity and expression within infected tissues

    • Correlation with host response patterns at the tissue level

The significant upregulation of pyrH during host cell interaction (3.52-fold transcript increase, 2.90-fold protein increase) makes it an excellent candidate for these emerging approaches, potentially revealing new aspects of Leptospira's adaptation to host environments and identifying novel intervention strategies.

What are the most promising research questions about Leptospira pyrH that remain unaddressed?

Several critical research questions about Leptospira pyrH remain unexplored, presenting opportunities for significant discoveries in understanding leptospirosis pathogenesis and developing novel interventions:

Fundamental Biology Questions:

  • Regulatory Mechanisms:

    • What transcriptional and post-transcriptional mechanisms control pyrH expression?

    • How do environmental signals modulate pyrH activity during host invasion?

    • Does pyrH expression correlate with virulence in different Leptospira strains?

    • Are there strain-specific regulatory elements affecting pyrH expression?

  • Protein-Protein Interactions:

    • Does Leptospira pyrH form complexes with other metabolic enzymes?

    • Are there host factors that directly interact with pyrH during infection?

    • How does the interactome of pyrH change during environmental transitions?

    • Could pyrH participate in moonlighting functions beyond its enzymatic role?

  • Structural Biology:

    • What unique structural features distinguish Leptospira pyrH from other bacterial homologs?

    • Are there allosteric regulatory sites specific to leptospiral pyrH?

    • How does the structure adapt to varying environmental conditions?

    • What structural changes occur during catalysis?

Pathogenesis-Related Questions:

  • Host Adaptation Role:

    • How does pyrH upregulation (3.52-fold transcript, 2.90-fold protein) contribute to intracellular survival?

    • Is pyrH expression different across host species (rodents vs. humans)?

    • Does pyrH activity correlate with bacterial persistence in reservoir hosts?

    • How does pyrH contribute to Leptospira's transition between environment and host?

  • Virulence Connection:

    • Is there a direct relationship between pyrH activity and virulence factor expression?

    • Does pyrH upregulation represent a specific adaptation to immune evasion?

    • Could targeting pyrH attenuate virulence without affecting environmental survival?

    • Is there differential expression in acute versus chronic infection stages?

Translational Research Opportunities:

Research QuestionExperimental ApproachPotential ImpactTechnical Challenges
Can pyrH inhibitors prevent Leptospira intracellular survival?Chemical genetics screeningNovel therapeutic approachDeveloping selective inhibitors
Is pyrH a viable component for multi-epitope vaccine formulations?Immunization studies with pyrH epitopesImproved vaccine strategiesIdentifying immunogenic epitopes
Can pyrH expression levels predict virulence potential?Comparative analysis across clinical isolatesDiagnostic/prognostic toolStandardizing measurement methods
Are there natural compounds that selectively inhibit leptospiral pyrH?Ethnobotanical screeningLow-cost intervention strategiesBioavailability and selectivity issues

Methodological Development Needs:

  • Genetic Manipulation Tools:

    • Development of inducible pyrH expression systems for Leptospira

    • Creation of reporter fusions to monitor expression in vivo

    • Establishment of pyrH conditional knockdown approaches

    • CRISPR-based fine-tuning of pyrH expression

  • Functional Assays:

    • High-throughput methods to measure pyrH activity in complex matrices

    • In vivo tracking of nucleotide metabolism in infected tissues

    • Single-cell assays for pyrH expression heterogeneity

    • Methods to correlate pyrH activity with bacterial fitness

  • Structural Characterization:

    • Crystallization protocols optimized for Leptospira pyrH

    • Cryo-EM approaches for visualizing macromolecular complexes

    • NMR methodologies for studying dynamics

    • Hydrogen-deuterium exchange approaches for conformational studies

Addressing these questions will provide a comprehensive understanding of how this metabolic enzyme contributes to Leptospira pathogenesis and potentially reveal new avenues for therapeutic intervention against leptospirosis.

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.