This protein catalyzes the condensation of isopentenyl diphosphate (IPP) with allylic pyrophosphates, resulting in the production of various terpenoids.
KEGG: lmf:LMOf2365_1332
Undecaprenyl pyrophosphate synthase (UppS) is an essential cytoplasmic enzyme that catalyzes the formation of isoprenoid UPP (C55-PP) from farnesyl pyrophosphate (FPP) and isopentenyl pyrophosphate (IPP) in the presence of Mg2+. This enzyme plays a critical role in peptidoglycan biosynthesis as UPP is a constituent of lipid II, the last peptidoglycan precursor responsible for transporting the GlcNAc-MurNAc-pentapeptide moiety across the cytoplasmic membrane. UppS is particularly significant because it is specific to bacteria and absent in human cells, making it an important target for developing novel antibacterial agents without causing host toxicity . Despite numerous published crystal structures of the enzyme in various states (apo, with substrates, or with inhibitors), there are currently no registered drugs targeting UppS, highlighting both its research potential and the challenges involved in drug development against this target .
L. monocytogenes serotype 4b is of special research interest because this serotype is responsible for a substantial fraction of food-borne listeriosis cases and is involved in the vast majority of common-source, food-borne outbreaks of the disease . Epidemiological data demonstrate that serotype 4b strains are particularly virulent in humans, making them a priority for research aimed at controlling foodborne listeriosis. Understanding the structure and function of essential enzymes like UppS in this serotype may provide insights into its enhanced virulence and pathogenicity. Additionally, serotype 4b has unique genetic markers and serotype-specific surface antigens that may interact with or affect UppS function and activity, potentially contributing to its distinctive virulence profile .
Researchers studying recombinant UppS in L. monocytogenes serotype 4b now have access to several sophisticated genetic tools. A significant breakthrough is the pheS*-based counterselection system, which enables efficient allelic exchange without requiring background genomic alterations . This system utilizes a mutated phenylalanine-tRNA synthetase gene (pheS*) that renders bacteria sensitive to p-chloro-phenylalanine (p-Cl-phe) when expressed under a constitutive promoter. The pLR16-pheS* suicide vector allows for precise genome editing, including sequential mutagenesis and introduction of point mutations . Additionally, serotype-specific molecular markers such as gtcA (specific to serogroup 4) and gltA and gltB (unique to serotype 4b complex) can be utilized for precise identification and manipulation of serotype 4b strains . These genetic tools collectively enable researchers to generate clean genetic modifications in L. monocytogenes for UppS functional studies, including expression of recombinant variants, domain swapping, and site-directed mutagenesis.
When expressing UppS in L. monocytogenes, researchers should consider the following approach:
Design the recombinant uppS construct with appropriate tags (His-tag or FLAG) for purification and detection
Clone the construct into the pLR16-pheS* vector between PspOMI and SalI restriction sites
Transform the construct into E. coli SM-10 cells for conjugation
Transfer to L. monocytogenes by conjugation with selection on media containing appropriate antibiotics
Facilitate integration through growth at 41°C followed by counterselection with p-Cl-phe at 37°C
Verify recombinants by PCR and confirm antibiotic sensitivity patterns
This methodological approach ensures stable expression of recombinant UppS variants for subsequent functional studies.
Several robust enzymatic assays can be employed to measure UppS activity in L. monocytogenes. The most common approaches include:
Radiometric assay: This traditional method utilizes 14C-labeled IPP to measure the incorporation of radiolabeled substrate into UPP. The reaction products are separated by thin-layer chromatography (TLC) or extracted with butanol and quantified by scintillation counting.
Continuous spectrophotometric assay: This assay couples pyrophosphate release during UppS catalysis to the oxidation of NADH through a series of enzyme reactions (typically involving pyrophosphatase, pyruvate kinase, and lactate dehydrogenase). The decrease in NADH absorbance at 340 nm provides a real-time measurement of UppS activity.
HPLC-based assay: High-performance liquid chromatography can be used to separate and quantify reaction products. This method is particularly valuable for characterizing inhibitors or substrate specificity.
For optimal results when studying L. monocytogenes serotype 4b UppS, the reaction conditions should include:
50 mM HEPES buffer (pH 7.5)
50-100 mM NaCl
0.1% Triton X-100
5-10 mM MgCl2 (essential cofactor)
20-50 μM FPP (substrate)
50-100 μM IPP (substrate)
Purified UppS enzyme (0.1-1.0 μg)
The reaction is typically conducted at 30°C for 15-30 minutes before analysis .
Researchers can generate site-specific mutations in the L. monocytogenes uppS gene using the pheS*-based allelic exchange system, which represents a significant advancement over previous methods. This approach allows for precise genetic modifications without leaving antibiotic resistance markers in the chromosome. The procedure involves:
Design of mutation strategy:
Create two ~500 bp homology arms flanking the mutation site
Introduce desired point mutations using overlap extension PCR with primers containing the mutations
Cloning into pLR16-pheS* vector:
Digest both the PCR product and pLR16-pheS* plasmid with appropriate restriction enzymes (e.g., PspOMI and SalI)
Ligate the fragments and transform into E. coli XL-1 Blue cells
Verify constructs by PCR and sequencing
Conjugation and first recombination:
Transform verified plasmids into E. coli SM-10 cells
Transfer to L. monocytogenes by conjugation on BHI agar plates
Select transconjugants on BHI agar containing streptomycin and chloramphenicol
Grow selected colonies at 41°C to facilitate plasmid integration
Counterselection and second recombination:
Culture integrated strains at 30°C to allow for plasmid excision
Plate diluted cultures on BHI agar containing 18 mM p-Cl-phe
Only cells that have lost the plasmid through a second recombination event will grow
Verify mutations by PCR and sequencing
Confirmation of mutants:
This method has been demonstrated to be highly efficient, with successful recombination rates significantly higher than previous approaches, making it ideal for studying structure-function relationships in UppS.
Inhibition of UppS in L. monocytogenes serotype 4b has profound effects on virulence and pathogenesis due to its essential role in peptidoglycan biosynthesis. When UppS activity is compromised, several key pathogenic processes are affected:
Cell wall integrity: Reduced UPP production leads to compromised peptidoglycan synthesis, resulting in weakened cell walls. This structural deficiency impairs bacterial survival under osmotic stress conditions encountered during infection.
Surface protein display: Many virulence factors in L. monocytogenes are anchored to the cell wall via processes dependent on proper peptidoglycan synthesis. Inhibition of UppS disrupts the proper display of these virulence-associated proteins, including internalin A and B, which are crucial for host cell invasion.
Stress response: L. monocytogenes serotype 4b strains with impaired UppS function show increased sensitivity to environmental stresses, including antimicrobial peptides, acidic conditions, and oxidative stress - all encountered during infection.
Intracellular survival: L. monocytogenes is an intracellular pathogen that must actively replicate within host cells. UppS inhibition prevents proper cell division and intracellular replication, severely limiting bacterial spread.
Cell-to-cell spread: The ability of L. monocytogenes to spread directly between adjacent host cells depends on actin-based motility and proper cell wall synthesis at the bacterial pole, both processes affected by UppS inhibition.
Research using anthranilic acid inhibitors of UppS has demonstrated significant reduction in virulence potential, making this enzyme a promising target for antimicrobial development against this important foodborne pathogen .
While comprehensive structural data specific to L. monocytogenes serotype 4b UppS is still emerging, comparative analyses with UppS from other bacterial species reveal several important structural differences that may impact enzyme function and inhibitor development:
UppS typically functions as a homodimer with each monomer containing a large central β-sheet surrounded by α-helices. The active site is located at the interface between the two domains of each monomer. Key structural differences include:
Substrate binding pocket composition: L. monocytogenes UppS contains unique amino acid residues in the hydrophobic binding pocket that interacts with the growing isoprenoid chain. These variations affect substrate specificity and inhibitor binding profiles.
Catalytic site architecture: While the catalytic mechanism involving Mg2+ coordination is conserved, subtle differences in the positioning of catalytic residues influence reaction kinetics and inhibitor sensitivity.
Conformational dynamics: L. monocytogenes UppS exhibits distinct conformational changes during catalysis compared to other bacterial UppS enzymes, particularly in the flexible loop regions that control substrate access to the active site.
Allosteric regulatory sites: Unique allosteric binding sites have been identified in L. monocytogenes UppS that are not present or are differently structured in other bacterial species.
Surface electrostatics: The distribution of charged residues on the enzyme surface differs between L. monocytogenes and other bacterial UppS enzymes, affecting protein-protein interactions and potentially membrane association.
These structural differences provide opportunities for developing serotype-specific inhibitors and understanding the evolutionary adaptations of this essential enzyme in L. monocytogenes serotype 4b .
Designing serotype-specific inhibitors targeting UppS in L. monocytogenes serotype 4b requires a multifaceted approach that leverages both structural and functional distinctions of this enzyme. Effective strategies include:
Structure-based design: Utilize crystal structures of UppS to identify unique binding pockets or conformational states specific to L. monocytogenes serotype 4b. Recent research on anthranilic acid inhibitors has shown promise in this regard, as these compounds can be modified to interact with serotype-specific residues in the enzyme active site .
Fragment-based screening: Begin with small molecular fragments that show binding affinity to unique regions of L. monocytogenes UppS, then systematically expand these molecules to enhance potency and specificity.
Allosteric inhibitor development: Target non-active site regions unique to L. monocytogenes serotype 4b UppS that can alter enzyme conformation or dynamics upon binding.
Exploiting serotype-specific cellular contexts: Design inhibitors that leverage interactions with serotype 4b-specific cell wall components or membrane characteristics to enhance local concentration or activity of UppS inhibitors.
Combination approaches: Develop dual-action inhibitors that simultaneously target UppS and serotype 4b-specific structures, such as the unique teichoic acid decorations controlled by gltA and gtcA genes .
Implementation of these strategies requires:
Detailed enzymological characterization of purified recombinant serotype 4b UppS
Comparative analysis of inhibitor binding kinetics across different L. monocytogenes serotypes
Validation of inhibitor specificity using genetic approaches such as the pheS*-based allelic exchange system to introduce resistance mutations
Evaluation of inhibitor efficacy in cellular and infection models
The most promising compounds from this research could serve as leads for developing narrow-spectrum antimicrobials that specifically target L. monocytogenes serotype 4b while minimizing disruption to beneficial microbiota.
Purifying active recombinant UppS from L. monocytogenes serotype 4b presents several technical challenges that researchers must address to obtain functional enzyme for biochemical and structural studies:
Membrane association: Although UppS is classified as a cytoplasmic enzyme, it often exhibits peripheral membrane association due to its interaction with hydrophobic substrates. This necessitates careful buffer optimization during purification.
Solution: Include low concentrations (0.1-0.5%) of non-ionic detergents such as Triton X-100 or CHAPS in purification buffers to maintain solubility without denaturing the enzyme.
Protein instability: L. monocytogenes UppS can show significant instability during purification, leading to activity loss.
Solution: Include stabilizing agents such as glycerol (10-20%), reducing agents (1-5 mM DTT or 2-mercaptoethanol), and protease inhibitors throughout the purification process. Perform all steps at 4°C and minimize the time between purification stages.
Cofactor requirements: Maintaining Mg2+ binding is essential for proper folding and activity.
Solution: Include 5-10 mM MgCl2 in all purification buffers to maintain the native conformation of the enzyme.
Aggregation during concentration: Recombinant UppS often aggregates during concentration steps required for crystallography or enzymatic assays.
Solution: Use step-wise concentration with gentle mixing between steps, and include 0.1-0.2% CHAPS or 150-300 mM NaCl to prevent protein-protein interactions leading to aggregation.
Expression level optimization: Achieving sufficient expression levels of recombinant UppS can be challenging, particularly when using the native L. monocytogenes expression system.
Solution: When using the pheS*-based system for homologous expression, optimize growth conditions (temperature, media composition) and consider using stronger constitutive promoters to enhance expression levels .
A detailed purification protocol that addresses these challenges typically achieves 80-90% purity with retention of >70% enzymatic activity, suitable for most biochemical and structural studies.
Genetic manipulation of L. monocytogenes serotype 4b uppS can encounter several obstacles due to the essential nature of this gene and technical challenges with the genetic tools. Here is a systematic troubleshooting guide for addressing common issues:
Possible causes:
Poor electroporation efficiency
Restriction barriers in recipient strain
Toxic effects of constructs
Solutions:
Use conjugation instead of electroporation as described in the pLR16-pheS* system protocol
Ensure SM-10 donor cells are in log phase before conjugation
Optimize donor:recipient ratio (typically 1:3 works well)
Use appropriate antibiotic concentrations for selection (chloramphenicol 7.5 μg/ml, streptomycin 200 μg/ml)
Possible causes:
Insufficient expression of pheS*
Suboptimal p-Cl-phe concentration
Second crossover events resulting in reversion to wild-type
Solutions:
Possible causes:
Secondary mutations
Polar effects
Altered monocin activity affecting bacterial growth
Solutions:
Troubleshooting decision tree for L. monocytogenes genetic manipulation:
No transconjugants obtained:
Check antibiotic sensitivity of donor and recipient strains
Verify plasmid integrity by restriction digestion
Reduce incubation temperature to 30°C for initial conjugation
First recombination successful but no counterselected colonies:
Increase counterselection incubation time to 48 hours
Verify pheS* expression by RT-PCR
Plate larger culture volumes on counterselection media
PCR verification shows mixed populations:
Understanding the interaction between UppS and other components of the cell wall biosynthesis machinery in L. monocytogenes serotype 4b requires sophisticated techniques that can capture these complex biological interactions. Several advanced methodologies have proven valuable:
Protein-Protein Interaction Analysis:
Bacterial two-hybrid systems: Adapted for Gram-positive organisms, this technique can identify direct protein partners of UppS.
Co-immunoprecipitation with crosslinking: Using antibodies against tagged UppS variants generated with the pheS* system, researchers can capture transient interactions with other cell wall synthesis enzymes.
Label-free protein interaction analysis using surface plasmon resonance (SPR) or microscale thermophoresis (MST) provides quantitative binding parameters for UppS interactions with putative partners.
Subcellular Localization Studies:
Fluorescence microscopy using GFP-tagged UppS (integrated using the pheS* system) can reveal the dynamic localization patterns during different growth phases.
Super-resolution microscopy techniques (STORM, PALM) can visualize co-localization of UppS with other cell wall synthesis enzymes at a resolution below the diffraction limit.
Immunogold electron microscopy offers nanometer-scale resolution of UppS localization relative to the membrane and cell wall structures.
Metabolic Labeling and Imaging:
Use of fluorescent or clickable isoprenoid analogs that can be incorporated by UppS to track the fate of its products through the peptidoglycan synthesis pathway.
Pulse-chase experiments with these labels can reveal the dynamics of UPP synthesis and utilization in serotype 4b.
Genetic Interaction Mapping:
Synthetic genetic arrays or targeted suppressor screens using the pheS* genetic manipulation system to identify genetic interactions between uppS and other cell wall biosynthesis genes.
Construction of conditional mutants in uppS combined with transcriptome analysis to identify compensatory mechanisms and regulatory networks.
In situ Activity Assays:
Development of FRET-based biosensors to monitor UppS activity in living L. monocytogenes cells.
Correlation of UppS activity with cell wall synthesis rates using dual-labeling approaches with orthogonal chemistries.
These methods, particularly when combined with knowledge of serotype 4b-specific genes like gtcA and gltA/gltB that affect cell wall structure , can provide unprecedented insights into how UppS integrates into the serotype-specific cell wall synthesis machinery. This understanding is crucial for developing targeted antimicrobial strategies and comprehending the heightened virulence of serotype 4b strains.
Several critical research questions are emerging regarding UppS as a drug target in L. monocytogenes serotype 4b, reflecting both recent advances and persistent knowledge gaps:
Inhibitor specificity and resistance mechanisms:
How do the structural differences in L. monocytogenes serotype 4b UppS influence inhibitor binding compared to other bacterial species?
What is the genetic barrier to resistance for novel UppS inhibitors, and do resistance mutations compromise bacterial fitness and virulence?
Can serotype-specific inhibitors be developed that preferentially target serotype 4b strains implicated in outbreaks?
Regulatory networks and adaptation:
How is uppS expression regulated in response to environmental stresses encountered during infection?
Does L. monocytogenes serotype 4b possess unique regulatory mechanisms for UppS activity that differ from other serotypes?
What compensatory mechanisms exist when UppS activity is compromised, and how do these affect pathogenesis?
Integration with serotype-specific pathways:
How does UppS functionally interact with serotype 4b-specific cell wall modification enzymes encoded by gtcA and gltA/gltB genes?
Does the product of UppS (undecaprenyl pyrophosphate) undergo serotype-specific modifications that influence virulence?
How do modifications in peptidoglycan precursor metabolism affect the expression of virulence determinants specific to serotype 4b?
Therapeutic potential:
Can UppS inhibitors be combined with other antimicrobials to create synergistic effects specific for L. monocytogenes?
What is the in vivo efficacy of UppS inhibitors in animal models of listeriosis?
Could UppS inhibitors be used prophylactically in high-risk foods to prevent L. monocytogenes growth without affecting beneficial microbiota?
Structural biology frontiers:
What are the conformational dynamics of L. monocytogenes UppS during catalysis and how do these differ from other bacterial UppS enzymes?
Can allosteric sites unique to L. monocytogenes serotype 4b UppS be identified and exploited for inhibitor design?
These research questions represent fertile ground for future investigations that could lead to novel therapeutic strategies against this important foodborne pathogen .
The pheS*-based counterselection system represents a powerful tool that can be adapted for high-throughput studies of UppS function in L. monocytogenes through several innovative approaches:
Creation of uppS mutation libraries:
The pheS* system can be modified to incorporate randomized mutagenesis of specific UppS domains
Design degenerate primers targeting functional domains of UppS for PCR mutagenesis
Clone mutagenized PCR products into the pLR16-pheS* vector and perform pooled conjugations
Use next-generation sequencing to identify successful recombinants and correlate mutations with phenotypes
Adapting for CRISPR-Cas9 integration:
Combine the pheS* counterselection with CRISPR-Cas9 for precise genome editing
Express Cas9 and guide RNAs targeting uppS from the pLR16-pheS* vector
Use homology-directed repair templates with specific mutations
This combined approach increases editing efficiency and enables high-throughput mutant generation
Development of arrayed mutant libraries:
Create a systematic collection of defined UppS variants using the pheS* system
Design mutations based on structural data and evolutionary conservation
Organize mutants in 96-well format for parallel phenotypic screening
This approach allows systematic structure-function analysis
Reporter system integration:
Use the pheS* system to integrate fluorescent or luminescent reporters downstream of uppS
Create transcriptional or translational fusions to monitor expression and activity
Design reporters responsive to cell wall stress for indirect measurement of UppS function
This enables real-time monitoring of UppS activity in live cells
Conditional expression systems:
Integrate inducible promoters upstream of uppS using the pheS* system
Create a suite of strains with varying expression levels by modifying ribosome binding sites
This approach allows titration of UppS levels to determine minimal functional thresholds
The advantages of the pheS* system for these applications include:
No antibiotic marker remains in the chromosome, preventing interference with phenotypic assays
High efficiency of recombination compared to traditional methods
Ability to make sequential modifications without introducing multiple selection markers
Compatibility with various L. monocytogenes strains without requiring background mutations
By leveraging these approaches, researchers can rapidly generate comprehensive data on UppS structure-function relationships, potentially identifying novel vulnerabilities for antimicrobial development.
Advancing our understanding of UppS in L. monocytogenes serotype 4b pathogenesis requires innovative interdisciplinary approaches that integrate multiple scientific fields. These collaborative strategies can provide comprehensive insights beyond what traditional microbiology approaches alone can achieve:
Systems Biology and Network Analysis:
Integration of transcriptomics, proteomics, and metabolomics data to create comprehensive models of cell wall biosynthesis networks in serotype 4b
Flux analysis of isoprenoid pathways to understand how UppS activity influences downstream processes
Computational modeling of cell wall dynamics during different phases of L. monocytogenes infection
Structural Biology and Biophysics:
Cryo-electron microscopy of UppS in complex with serotype 4b-specific cell wall synthesis machinery
Neutron scattering and hydrogen-deuterium exchange mass spectrometry to map protein dynamics during catalysis
Molecular dynamics simulations to understand how serotype-specific variations influence enzyme function
Chemical Biology and Probe Development:
Design of activity-based probes that report on UppS activity in live bacteria during infection
Development of photoaffinity labels to capture transient protein-protein interactions in the cell wall synthesis pathway
Creation of "clickable" UppS substrates to track the fate of enzyme products in the cell
Advanced Infection Models:
Organoid systems to study the interaction of UppS-dependent cell wall structures with human tissues
Humanized mouse models expressing human receptors targeted by L. monocytogenes
Ex vivo infection models using human placental tissue to understand serotype 4b's tropism for this tissue
Synthetic Biology and Bioengineering:
Creation of L. monocytogenes strains with orthogonal UppS enzymes to study function without perturbing essential processes
Development of genetic circuits to place UppS under precise spatiotemporal control during infection
Engineering L. monocytogenes with modified cell walls to probe the role of UppS products in host-pathogen interactions
Evolutionary Microbiology:
These interdisciplinary approaches, particularly when combined with the genetic manipulation capabilities of the pheS* system and knowledge of serotype 4b-specific genetic markers , can provide unprecedented insights into how this essential enzyme contributes to the unique virulence properties of L. monocytogenes serotype 4b. Such understanding is crucial for developing targeted interventions against this important foodborne pathogen.
The field of L. monocytogenes serotype 4b UppS research has witnessed several significant advancements that have collectively enhanced our understanding of this essential enzyme and its potential as an antimicrobial target:
Development of efficient genetic manipulation tools: The creation of the pheS*-based counterselection system represents a major breakthrough, enabling precise genetic modifications in L. monocytogenes without leaving antibiotic resistance markers in the chromosome. This tool has accelerated functional studies of UppS and other essential genes by allowing clean deletions, point mutations, and sequential genetic manipulations .
Identification of serotype-specific genetic markers: The characterization of genes unique to serotype 4b (such as gltA and gltB) and serogroup 4 (such as gtcA) has improved our understanding of the genetic basis of serotype differences. These markers provide important context for studying UppS function within the specific genetic background of serotype 4b strains associated with listeriosis outbreaks .
Structural and biochemical characterization of UppS: Significant progress has been made in understanding the structure-function relationships of UppS enzymes, including the identification of key catalytic residues, substrate binding sites, and conformational changes during catalysis. This knowledge forms the foundation for rational drug design approaches targeting this enzyme .
Discovery of novel UppS inhibitor classes: The identification of anthranilic acid derivatives and other chemical scaffolds as UppS inhibitors has opened new avenues for antimicrobial development. These compounds provide valuable chemical probes for studying UppS function and represent promising leads for drug development .
Elucidation of the monocin locus function: The characterization of the monocin genomic region in L. monocytogenes has revealed its role in bacterial lysis upon DNA damage, providing important context for interpreting phenotypes of genetic manipulations, including those targeting uppS .
These advancements collectively provide a robust foundation for future research aimed at exploiting UppS as a target for controlling L. monocytogenes serotype 4b infections and understanding the molecular basis of its enhanced virulence.
Understanding UppS function in L. monocytogenes serotype 4b has several practical applications with significant implications for both food safety and clinical treatment:
Food Safety Applications:
Development of specific biocontrol agents: Knowledge of UppS structure and function enables the design of targeted inhibitors that could be used as food preservatives or surface sanitizers specific for L. monocytogenes without affecting beneficial food microbiota.
Enhanced detection methods: Understanding serotype-specific aspects of UppS and related cell wall structures permits the development of more specific detection methods for serotype 4b strains in food processing environments, potentially utilizing antibodies or aptamers targeting unique cell surface features.
Risk assessment tools: Characterization of how UppS function influences stress resistance can improve predictive microbiology models for assessing the behavior of serotype 4b strains under various food processing and storage conditions.
Natural antimicrobial discovery: Insights from UppS structure can guide screening of natural compounds from food sources that may specifically inhibit this enzyme, leading to new natural preservatives.
Clinical Applications:
Targeted antimicrobial development: UppS represents a promising target for developing narrow-spectrum antibiotics specifically effective against L. monocytogenes, potentially addressing the challenge of listeriosis treatment without disrupting the patient's microbiome.
Combination therapy strategies: Understanding how UppS inhibition affects bacterial physiology can inform rational combination therapies with existing antibiotics, potentially enhancing their efficacy against resistant strains.
Virulence attenuation approaches: Modulating UppS activity could provide strategies to attenuate virulence without killing bacteria, potentially reducing the selection pressure for resistance while still controlling infection.
Diagnostic improvements: Knowledge of serotype 4b-specific aspects of UppS and related cell wall components can lead to improved diagnostic tests that rapidly identify this clinically significant serotype in patient samples.
Vaccine development: Understanding UppS-dependent cell wall structures unique to serotype 4b could inform the development of more effective vaccines targeting high-risk populations, including pregnant women and immunocompromised individuals .