Recombinant Listeria monocytogenes serotype 4b Cobyric acid synthase (cobQ), partial

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 during order placement for customized preparation.
Lead Time
Delivery times vary depending on the purchasing method and location. Please contact your local distributor for precise delivery estimates.
Note: All proteins are shipped with standard blue ice packs. Dry ice shipping requires prior arrangement and incurs additional charges.
Notes
Avoid repeated freeze-thaw cycles. Store working aliquots at 4°C for up to one week.
Reconstitution
Centrifuge the vial briefly before opening to consolidate 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%, provided as a guideline for your reference.
Shelf Life
Shelf life depends on various factors including storage conditions, buffer composition, temperature, and the protein's inherent 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. Aliquoting is essential 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 for preferential development.
Synonyms
cobQ; LMOf2365_1217; Cobyric acid synthase
Buffer Before Lyophilization
Tris/PBS-based buffer, 6% Trehalose.
Datasheet
Please contact us to get it.
Protein Length
Partial
Purity
>85% (SDS-PAGE)
Species
Listeria monocytogenes serotype 4b (strain F2365)
Target Names
cobQ
Uniprot No.

Target Background

Function
Catalyzes amidations at positions B, D, E, and G on adenosylcobyrinic acid C-diamide. Glutamine provides the NH₂ groups, and one ATP molecule is hydrolyzed per amidation.
Database Links
Protein Families
CobB/CobQ family, CobQ subfamily

Q&A

What is cobyric acid synthase and what role does it play in Listeria monocytogenes?

Cobyric acid synthase is an essential enzyme in the cobalamin (vitamin B12) biosynthesis pathway of Listeria monocytogenes. In particular, the enzyme CbiP catalyzes the synthesis of adenosyl-cobyric acid, a critical step that occurs near the end of the cobalamin biosynthesis pathway . This enzyme is encoded by the cbiP gene in L. monocytogenes and represents one of the twenty-two genes involved in the complete cobalamin biosynthesis pathway according to the KEGG pathway database . Cobalamin is a vital cofactor for several metabolic enzymes and plays crucial roles in DNA synthesis, fatty acid metabolism, and amino acid metabolism in bacterial cells. The functional importance of cobyric acid synthase extends beyond basic metabolism to stress response mechanisms, as evidenced by research showing its involvement in bacterial tolerance to environmental stressors like low temperature and copper exposure .

How does the cobalamin biosynthesis pathway contribute to Listeria survival under stress conditions?

The cobalamin biosynthesis pathway appears to be integral to Listeria monocytogenes' ability to withstand various environmental stresses, particularly the combination of low temperature and presence of copper. Research has demonstrated that when L. monocytogenes is exposed to low temperature (8°C) and copper stress (0.5 mM CuSO₄ × 5H₂O), the expression of several cobalamin biosynthesis-related genes, including cbiP, cbiB, and cysG, increases significantly after 6 hours of exposure . This upregulation suggests that the cobalamin biosynthesis pathway is activated as part of the bacterial stress response mechanism. Furthermore, experimental evidence indicates that deletion of the cbiP gene results in decreased tolerance to the combination of low temperature and copper, particularly at higher copper concentrations (3 mM) . The addition of cyanocobalamin (5 nM) to the growth medium restored the growth capacity of cbiP mutant strains, confirming that cobalamin is indeed necessary for L. monocytogenes growth under these stress conditions . These findings collectively indicate that cobalamin biosynthesis is a crucial component of L. monocytogenes' survival strategy under environmental stress.

Why is recombinant production of Listeria monocytogenes proteins important for research purposes?

Recombinant production of Listeria monocytogenes proteins, including enzymes like cobyric acid synthase, offers numerous advantages for research applications. The recombinant approach allows for controlled expression and purification of specific proteins, facilitating detailed studies of their structure, function, and biochemical properties. For instance, recombinant L. monocytogenes GAPDH proteins have been successfully used in cross-reactive vaccine development, demonstrating the versatility of recombinant bacterial proteins as research tools . Recombinant protein production enables researchers to work with purified enzymes without the need to handle large volumes of pathogenic bacteria, addressing significant safety concerns associated with cultivating virulent strains of L. monocytogenes. Additionally, recombinant proteins can be engineered with specific tags or modifications to facilitate detection, purification, or functional studies. In the context of cobyric acid synthase research, recombinant protein production allows for detailed characterization of enzyme kinetics, substrate specificity, and response to environmental factors that might influence enzyme activity.

What are the most effective methods for creating and validating cbiP/cobQ gene deletions in Listeria monocytogenes?

Creating and validating gene deletions in Listeria monocytogenes requires careful experimental design and robust validation strategies. For cbiP gene deletion, homologous recombination has been successfully employed, as demonstrated in research examining the role of cbiP in L. monocytogenes tolerance to environmental stressors . The process typically involves constructing a deletion vector containing homologous regions flanking the target gene, along with a selectable marker. This construct is then introduced into L. monocytogenes cells, where homologous recombination results in the replacement of the target gene with the selectable marker. Confirmation of successful gene deletion is critical and typically involves multiple approaches. PCR verification using primers that flank the deletion site can confirm the absence of the target gene and the presence of the selectable marker. Sequencing the deletion region provides additional validation of the genetic modification. Phenotypic validation is equally important, especially for genes like cbiP that have observable growth phenotypes under specific conditions. For cbiP deletions, comparing the growth of wild-type and mutant strains under stress conditions (e.g., low temperature and copper exposure) provides functional validation of the deletion . Complementation studies, in which the deleted gene is reintroduced, can further confirm that observed phenotypes are specifically due to the gene deletion.

How can researchers effectively measure cobalamin biosynthesis pathway activity in Listeria monocytogenes under various stress conditions?

Measuring cobalamin biosynthesis pathway activity in Listeria monocytogenes under stress conditions requires a multi-faceted approach combining molecular, biochemical, and physiological techniques. At the transcriptional level, quantitative PCR (qPCR) can be used to measure the expression levels of key genes in the cobalamin biosynthesis pathway, such as cbiP, cbiB, and cysG, as demonstrated in previous research . The expression patterns of these genes provide insights into the activation of the pathway under different stress conditions. Proteomic approaches, including western blotting or mass spectrometry, can be employed to quantify the protein levels of cobalamin biosynthesis enzymes, offering a more direct measure of potential enzymatic activity. Direct measurement of cobalamin and its intermediates using high-performance liquid chromatography (HPLC) or liquid chromatography-mass spectrometry (LC-MS) provides definitive evidence of pathway activity. Growth experiments comparing wild-type and pathway-deficient mutants (e.g., ΔcbiP) under stress conditions, with and without cobalamin supplementation, offer functional evidence of pathway activity . Additionally, enzymatic assays using purified recombinant proteins can directly measure the activity of specific enzymes in the pathway under controlled conditions, allowing for detailed characterization of how stress factors influence enzyme function.

What are the potential research applications of recombinant Listeria cobyric acid synthase beyond basic enzymatic studies?

Recombinant Listeria cobyric acid synthase has potential applications extending well beyond basic enzymatic characterization. One significant application is in the development of targeted antimicrobial strategies. Since cobalamin biosynthesis appears crucial for L. monocytogenes survival under stress conditions commonly used in food preservation (low temperature, presence of copper) , inhibitors of cobyric acid synthase could potentially serve as novel antimicrobial agents specific to Listeria. The recombinant enzyme could be used in high-throughput screening assays to identify such inhibitors. Structural biology studies using purified recombinant enzyme could reveal the three-dimensional structure of cobyric acid synthase, potentially identifying unique structural features that could be targeted for pathogen-specific inhibition. Immunological studies represent another promising application, as bacterial enzymes can serve as antigenic targets. Research has already demonstrated that certain L. monocytogenes proteins, like GAPDH, have potential as vaccine candidates due to their immunogenicity and sequence conservation across related bacterial genera . Metabolic engineering applications are also possible, where recombinant expression of cobyric acid synthase could be used to enhance cobalamin production in non-pathogenic organisms, potentially creating safer systems for studying cobalamin biosynthesis or even for biotechnological production of vitamin B12.

What molecular biology techniques are most suitable for studying the expression and regulation of cobalamin biosynthesis genes in Listeria?

Several molecular biology techniques have proven effective for investigating the expression and regulation of cobalamin biosynthesis genes in Listeria monocytogenes. Quantitative reverse transcription PCR (RT-qPCR) is particularly valuable for measuring the transcriptional response of cobalamin biosynthesis genes under different experimental conditions. This technique has been successfully employed to demonstrate increased expression of cbiP, cbiB, and cysG genes in response to low temperature and copper stress . RNA sequencing (RNA-seq) provides a more comprehensive approach, enabling genome-wide transcriptional profiling that can reveal coordinated regulation of multiple genes in the cobalamin biosynthesis pathway alongside other stress response systems. Promoter-reporter fusion constructs, in which the promoter regions of cobalamin biosynthesis genes are fused to reporter genes such as lacZ or fluorescent proteins, allow for visual or quantifiable monitoring of gene expression in living cells under various conditions. Chromatin immunoprecipitation followed by sequencing (ChIP-seq) can identify transcription factors that directly regulate cobalamin biosynthesis genes by binding to their promoter regions. Electrophoretic mobility shift assays (EMSAs) or DNA footprinting can further characterize these protein-DNA interactions in vitro. For post-transcriptional regulation studies, ribosome profiling can reveal translational efficiency of cobalamin biosynthesis mRNAs under different conditions, while protein stability assays can assess post-translational regulatory mechanisms affecting enzyme levels.

How can researchers design experiments to assess the impact of cobyric acid synthase on Listeria virulence and pathogenicity?

Designing experiments to assess the impact of cobyric acid synthase on Listeria monocytogenes virulence requires a strategic approach combining in vitro, ex vivo, and in vivo methodologies. In vitro invasion and intracellular replication assays using human cell lines (e.g., Caco-2, HeLa) can compare the invasive capacity and intracellular growth of wild-type L. monocytogenes versus cobyric acid synthase mutants (ΔcbiP). These assays should be conducted under conditions that mimic the host environment, including physiological temperature (37°C) and potentially microaerobic conditions. Ex vivo organ culture models using tissues like intestinal explants can provide more complex systems to assess bacterial adherence, invasion, and early host responses. In vivo infection models using appropriate animal models (typically mice) represent the gold standard for virulence assessment. These studies should compare the infection progression, bacterial burden in organs, histopathological changes, and survival rates between animals infected with wild-type and mutant strains. Complementation experiments, where the deleted cbiP gene is reintroduced into the mutant strain, are essential to confirm that any observed virulence differences are specifically due to the absence of cobyric acid synthase rather than unintended genetic changes. Additionally, experiments assessing the impact of cobyric acid synthase on specific virulence mechanisms, such as resistance to host antimicrobial defenses, biofilm formation, and stress tolerance under host-like conditions, can provide mechanistic insights into how this enzyme might contribute to pathogenicity.

What are the key considerations for optimizing recombinant expression of Listeria monocytogenes cobyric acid synthase?

Optimizing recombinant expression of Listeria monocytogenes cobyric acid synthase requires careful consideration of several factors to ensure high yield of functional protein. Selection of an appropriate expression system is critical - while E. coli is commonly used for bacterial protein expression, specialized strains like those carrying additional tRNAs for rare codons may be necessary to accommodate Listeria's different codon usage patterns. Alternative expression hosts like Bacillus subtilis, which is more closely related to Listeria, could potentially provide better expression of properly folded protein. Vector design considerations include selecting an appropriate promoter strength, incorporating solubility-enhancing fusion tags (e.g., His-tag, MBP, SUMO), and optimizing the sequence for expression efficiency while maintaining the critical catalytic residues. Expression conditions must be carefully optimized, including temperature (often lowered to improve protein folding), inducer concentration, media composition, and duration of expression. The presence of cofactors or substrates in the expression media might also enhance proper folding of the enzyme. Purification strategies should be designed based on the incorporated tags and the biophysical properties of the protein, potentially including affinity chromatography, ion exchange, and size exclusion steps. Functional validation through enzymatic activity assays is essential to confirm that the recombinant protein retains its native catalytic properties. Stability assessment under various buffer conditions, pH values, and temperatures can identify optimal storage conditions. Finally, structural characterization using techniques like circular dichroism, thermal shift assays, or more advanced methods like X-ray crystallography or cryo-electron microscopy can provide valuable insights into the protein's structural integrity and potential for crystallization studies.

What insights can comparative studies between Listeria and other bacterial genera provide about cobalamin biosynthesis evolution?

Comparative studies between Listeria and other bacterial genera offer valuable insights into the evolution and functional diversification of cobalamin biosynthesis pathways. Research has shown interesting parallels between cobalamin biosynthesis genes in Listeria, Mycobacterium, and other bacterial genera. For instance, mutation of the cobK gene in Mycobacterium smegmatis affects cobalamin synthesis, suggesting that alterations in one gene within the pathway can have broader impacts on cobalamin production across diverse bacteria . Similarly, studies in Halobacterium have demonstrated that cbiP deletion reduces growth in corrinoid-deficient media, with growth capacity restored through cobyric acid supplementation – a pattern that mirrors observations in Listeria monocytogenes cbiP mutants . These parallels suggest conservation of certain functional aspects of the cobalamin biosynthesis pathway across phylogenetically diverse bacteria. Interestingly, some bacterial proteins show remarkable sequence conservation across genera – for example, the N-terminal GAPDH 1-22 peptide shows more than 95% homology across Listeria, Mycobacterium, and Streptococcus . This high degree of conservation in certain metabolic proteins raises the possibility that similar patterns might exist in cobalamin biosynthesis enzymes, potentially reflecting fundamental functional constraints. Evolutionary analysis of cobalamin biosynthesis genes across bacterial phyla can reveal ancient gene duplication events, horizontal gene transfer, and lineage-specific adaptations that have shaped the diverse strategies bacteria employ for cobalamin acquisition and synthesis. Such comparative approaches not only enhance our understanding of evolutionary processes but may also identify conserved features that could serve as broad-spectrum antimicrobial targets.

How do environmental isolates of Listeria monocytogenes differ from clinical isolates in their cobalamin biosynthesis pathways?

Environmental and clinical isolates of Listeria monocytogenes likely exhibit differences in their cobalamin biosynthesis pathways that reflect adaptation to their respective ecological niches. Clinical isolates, which must survive and replicate within host tissues, may show adaptations in their cobalamin biosynthesis pathways that enhance virulence and stress tolerance under host conditions. These adaptations could include modifications to enzyme structure that improve function at host body temperature (37°C) or regulatory changes that coordinate cobalamin synthesis with other virulence mechanisms. In contrast, environmental isolates, particularly those from food processing environments, might show adaptations that enhance survival under food preservation conditions. Research has already identified strains like List2-2 that demonstrate enhanced tolerance to combined stressors of low temperature (8°C) and copper, with evidence linking this tolerance to cobalamin biosynthesis . The upregulation of cobalamin biosynthesis genes, including cbiP, in response to these stressors suggests potential regulatory differences between environmental and clinical isolates. Comparative genomic and transcriptomic analyses of environmental versus clinical isolates could reveal differences in gene sequence, copy number, or expression patterns within the cobalamin biosynthesis pathway. Analysis of cbiP/cobQ genetic diversity across a large collection of isolates from different sources might identify lineage-specific alleles associated with particular ecological niches or clinical outcomes. The adaptation of Listeria to diverse environments likely involves optimization of metabolic pathways, including cobalamin biosynthesis, to the specific challenges and resources available in each niche, potentially leading to functionally significant differences between environmental persisters and clinical isolates.

How might CRISPR-Cas9 technologies advance research on Listeria monocytogenes cobalamin biosynthesis?

CRISPR-Cas9 technology offers revolutionary approaches for investigating cobalamin biosynthesis in Listeria monocytogenes, enabling precise genetic manipulations that were previously challenging or impossible. The precision and efficiency of CRISPR-Cas9 make it ideal for creating clean deletion mutants of cobalamin biosynthesis genes without introducing polar effects on neighboring genes, addressing a common limitation of traditional recombination-based methods. Beyond simple gene deletions, CRISPR-based methods can generate point mutations that specifically alter catalytic residues or regulatory elements within genes like cbiP, allowing researchers to distinguish between the structural and enzymatic functions of cobyric acid synthase. Multiplexed CRISPR systems enable simultaneous targeting of multiple genes in the cobalamin biosynthesis pathway, facilitating studies of pathway redundancy, gene interaction effects, and the relative importance of different pathway branches. CRISPR interference (CRISPRi) and CRISPR activation (CRISPRa) systems provide reversible and tunable control over gene expression, allowing temporal studies of cobalamin biosynthesis regulation without permanent genetic modifications. These approaches are particularly valuable for studying essential genes where complete deletion would be lethal. Genome-wide CRISPR screens in Listeria could identify novel genes that interact with or regulate the cobalamin biosynthesis pathway, potentially revealing unexpected connections to stress response systems or virulence mechanisms. Additionally, CRISPR-mediated homology-directed repair can introduce reporter tags directly into the native genetic loci of cobalamin biosynthesis genes, enabling real-time visualization of protein localization and expression dynamics in living cells under various conditions.

What potential exists for structure-based drug design targeting Listeria monocytogenes cobyric acid synthase?

Structure-based drug design targeting Listeria monocytogenes cobyric acid synthase represents a promising approach for developing novel antimicrobials with specificity for this important foodborne pathogen. The critical role of cobyric acid synthase in bacterial survival under stress conditions relevant to food preservation methods makes it an attractive target for antimicrobial development. The first step in structure-based drug design would be obtaining high-resolution structural data of L. monocytogenes cobyric acid synthase through X-ray crystallography, cryo-electron microscopy, or computational modeling based on homologous proteins with known structures. Once structural data is available, computational approaches including molecular docking, virtual screening, and molecular dynamics simulations could identify potential binding pockets and predict small molecules that might bind to and inhibit the enzyme. The catalytic site is an obvious target, but allosteric sites that affect enzyme conformation or substrate binding could also offer opportunities for inhibition. Comparative structural analysis between L. monocytogenes cobyric acid synthase and human enzymes could identify structural differences that might be exploited to develop inhibitors with high selectivity for the bacterial enzyme, reducing the risk of off-target effects. Rational drug design approaches could focus on developing transition state analogs that mimic the structure of reaction intermediates in the cobyric acid synthesis reaction, potentially creating highly specific competitive inhibitors. Additionally, fragment-based drug discovery, where small molecular fragments are screened for binding to the target protein and then optimized and combined to create potent inhibitors, could be particularly effective for this enzyme target.

How can understanding cobalamin biosynthesis in Listeria contribute to improved food safety measures?

Understanding cobalamin biosynthesis in Listeria monocytogenes offers several avenues for enhancing food safety strategies and control measures. The discovery that cobalamin biosynthesis plays a role in Listeria's tolerance to low temperature and copper stress has direct implications for food preservation methods. This knowledge could inform the development of more effective preservation strategies that specifically target or circumvent this protective mechanism. For instance, combining cold storage with compounds that inhibit cobalamin biosynthesis might synergistically reduce Listeria survival in food products. The identification of specific genes like cbiP whose deletion reduces stress tolerance provides potential targets for biocontrol approaches. Bacteriophages or other biocontrol agents could potentially be engineered to specifically target and disrupt these genes or their products in Listeria contaminating food processing environments. Insights into cobalamin biosynthesis could also inform the development of novel detection methods for Listeria in food samples. For example, molecular assays targeting cobalamin biosynthesis genes might offer advantages in specificity or sensitivity compared to current detection methods. The finding that specific bacterial species often co-occur with Listeria in food processing environments raises the possibility that these microbial associations might influence cobalamin biosynthesis or availability. Understanding these ecological interactions could potentially lead to biocontrol strategies based on competitive exclusion or manipulation of the microbiome in food processing environments. Additionally, cobalamin-related metabolites might serve as biomarkers for Listeria presence or stress response, potentially enabling the development of rapid, metabolite-based detection methods for monitoring food safety.

What considerations are important when designing experiments to screen for inhibitors of Listeria cobyric acid synthase?

Designing effective screening assays for inhibitors of Listeria monocytogenes cobyric acid synthase requires careful consideration of several factors to ensure identification of compounds with genuine therapeutic potential. The primary screening assay should directly measure cobyric acid synthase enzymatic activity using purified recombinant enzyme. This could involve spectrophotometric detection of reaction products or consumption of substrates, radioactive tracer methods to monitor substrate conversion, or coupled enzyme assays that link cobyric acid synthase activity to a more easily measured output. Counter-screening against human enzymes with similar functions is essential to identify compounds with selectivity for the bacterial target, reducing the risk of toxicity. Cell-based secondary assays using both wild-type Listeria and cbiP deletion mutants can confirm that potential inhibitors identified in enzymatic screens actually penetrate bacterial cells and exert their effects through the intended target rather than through off-target effects. Testing candidate inhibitors under various relevant conditions, including different temperatures, pH values, and in the presence of food components, is crucial given that cobyric acid synthase is particularly important under stress conditions like low temperature . Combination testing with existing food preservatives or antimicrobials could identify synergistic effects that might be leveraged for more effective Listeria control. Screening assays should include controls to identify false positives resulting from non-specific mechanisms like protein denaturation or aggregation. Finally, considering the potential for resistance development, screening approaches that identify compounds targeting highly conserved regions of cobyric acid synthase might yield inhibitors with higher barriers to resistance.

How might recombinant Listeria enzymes be utilized in cross-reactive vaccine development?

Recombinant Listeria enzymes, including potentially cobyric acid synthase, offer promising opportunities for developing cross-reactive vaccines that could provide protection against multiple bacterial pathogens. Research has already demonstrated that recombinant Listeria monocytogenes GAPDH proteins can serve as effective components in dendritic cell (DC)-based vaccines, conferring protection not only against Listeria but also against Mycobacterium and Streptococcus infections . This cross-protection is facilitated by high sequence conservation in certain protein domains across these bacterial genera, such as the N-terminal GAPDH 1-22 peptide that shows more than 95% homology . Similar approaches could potentially be applied using recombinant cobyric acid synthase or other conserved metabolic enzymes. The success of DC vaccines loaded with recombinant L. monocytogenes GAPDH suggests a general strategy wherein DCs are loaded with purified recombinant bacterial proteins to create safe and immunogenic vaccine vectors . This approach avoids the safety concerns associated with live attenuated bacterial vaccines while still effectively activating both CD4+ and CD8+ T cell responses. The strong adjuvant properties observed with recombinant protein-loaded DCs, attributed to their ability to induce non-specific DC activation, represent an additional advantage of this approach . Epitope mapping of cobyric acid synthase could identify highly conserved regions that might serve as the basis for peptide vaccines eliciting cross-protective immunity. Recombinant enzymes could also be incorporated into novel vaccine delivery systems, such as nanoparticles or liposomes, potentially enhancing immunogenicity while allowing for precise control over antigen presentation. Finally, combining multiple recombinant antigens from different metabolic pathways could potentially enhance the breadth and effectiveness of cross-protection.

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.