Recombinant Nocardia farcinica Malate synthase G (glcB), partial

Shipped with Ice Packs
In Stock

Description

Introduction to Recombinant Nocardia farcinica Malate Synthase G (glcB), Partial

Recombinant Nocardia farcinica Malate Synthase G (glcB), partial, refers to a genetically engineered version of the Malate Synthase G enzyme, which is involved in the glyoxylate cycle. This enzyme plays a crucial role in the metabolism of certain bacteria, including Nocardia farcinica, by facilitating the conversion of acetyl-CoA and glyoxylate into malate. The glyoxylate cycle is essential for the survival and growth of these bacteria, particularly under conditions where glucose is not readily available.

Function and Importance of Malate Synthase G

Malate Synthase G is a key enzyme in the glyoxylate cycle, which allows bacteria to utilize two-carbon compounds like acetate as a sole carbon source. This metabolic pathway is vital for the pathogenicity and survival of Nocardia farcinica, as it enables the bacterium to thrive in environments with limited nutrient availability.

EnzymeFunctionPathway
Malate Synthase G (glcB)Converts acetyl-CoA and glyoxylate into malateGlyoxylate cycle

Research Findings on Nocardia farcinica

While specific research on Recombinant Nocardia farcinica Malate Synthase G (glcB), partial, is limited, studies on Nocardia farcinica itself highlight its importance as a pathogen. Nocardia farcinica is known for causing severe infections, particularly in immunocompromised individuals . Research has focused on understanding its virulence factors and developing strategies to combat its infections .

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 preparation.
Lead Time
Delivery times vary depending on the purchase method and location. Please consult your local distributor for precise delivery estimates.
Note: Standard shipping includes blue ice packs. Dry ice shipping requires advance notification 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% and can serve as a reference.
Shelf Life
Shelf life depends on several factors: storage conditions, buffer components, temperature, and protein stability. Generally, liquid formulations have a 6-month shelf life at -20°C/-80°C, while lyophilized formulations have a 12-month shelf life at -20°C/-80°C.
Storage Condition
Store at -20°C/-80°C upon receipt. Aliquot for multiple uses. Avoid repeated freeze-thaw cycles.
Tag Info
Tag type is determined during the manufacturing process.
The specific tag will be determined during production. If you require a specific tag, please inform us, and we will prioritize its development.
Synonyms
glcB; NFA_25030Malate synthase G; EC 2.3.3.9
Buffer Before Lyophilization
Tris/PBS-based buffer, 6% Trehalose.
Datasheet
Please contact us to get it.
Protein Length
Partial
Purity
>85% (SDS-PAGE)
Species
Nocardia farcinica (strain IFM 10152)
Target Names
glcB
Uniprot No.

Target Background

Function

Involved in glycolate utilization. This enzyme catalyzes the condensation and subsequent hydrolysis of acetyl-coenzyme A (acetyl-CoA) and glyoxylate to form malate and CoA.

Database Links
Protein Families
Malate synthase family, GlcB subfamily
Subcellular Location
Cytoplasm.

Q&A

What is Malate synthase G (glcB) and what is its role in Nocardia farcinica metabolism?

Malate synthase G is a key enzyme in the glyoxylate cycle that catalyzes the condensation of acetyl-CoA with glyoxylate to form malate. In N. farcinica, this enzyme likely plays a critical role in alternative carbon source utilization, particularly during infection conditions where primary carbon sources may be limited.

To characterize glcB function in N. farcinica, researchers typically employ:

  • Gene cloning and recombinant expression

  • Enzyme activity assays measuring oxaloacetate formation

  • Knockout studies examining growth on acetate or fatty acids

  • Complementation experiments to confirm phenotype restoration

The function should be investigated in context with other metabolic pathways, as N. farcinica demonstrates sophisticated metabolic adaptation mechanisms during infection processes .

How can Nocardia farcinica be reliably identified in clinical and research settings?

N. farcinica identification requires specialized methods due to its similarity to other Nocardia species. The most reliable approach utilizes PCR amplification with species-specific primers:

The Nf1 (5′-CCGCAGACCACGCAAC) and Nf2 (5′-ACGAGGTGACGGCTGC) primer pair has been validated to specifically amplify a 314-bp fragment only present in N. farcinica strains . This PCR assay provides rapid (within 24 hours of obtaining DNA) and highly specific identification, distinguishing N. farcinica from other Nocardia species and related genera.

Verification of amplification products can be performed through:

  • Restriction enzyme digestion using CfoI

  • Direct sequencing of the 314-bp fragment

  • Comparison with reference strain profiles

This molecular approach significantly outperforms traditional phenotypic methods, which are time-consuming and often lead to misidentification in clinical laboratories .

What expression systems are recommended for producing recombinant Nocardia proteins?

For successful expression of N. farcinica glcB, selection of an appropriate expression system is critical. When designing recombinant DNA experiments involving Nocardia proteins, researchers must consider:

  • E. coli expression systems: Typically using pET vectors with T7 promoters for high yield, though codon optimization may be necessary due to GC content differences

  • Induction parameters: Optimal conditions often include 0.2 mM IPTG at 28°C, similar to what has been effective for other N. farcinica proteins

  • Regulatory compliance: All recombinant DNA work must comply with NIH Guidelines, especially when conducted at institutions receiving NIH funding

  • Biosafety considerations: Appropriate containment levels must be maintained as N. farcinica is an opportunistic pathogen

Remember that expression systems should be selected based on downstream applications. For structural studies, yeast or insect cell systems may provide better protein folding, while E. coli systems are preferred for rapid, high-yield production.

What purification challenges are specific to recombinant proteins from Nocardia farcinica?

Purification of recombinant N. farcinica proteins presents several methodological challenges:

  • Solubility issues: GC-rich organisms like Nocardia often produce proteins with unique folding properties that may form inclusion bodies in heterologous expression systems

  • Contaminating enzymes: N. farcinica produces numerous hydrolytic enzymes that may co-purify or degrade target proteins

  • Endotoxin removal: For immunological studies, lipid components must be carefully removed

  • Protein authentication: Confirmation of protein identity through mass spectrometry is essential, particularly for partial protein constructs

A methodological approach to overcome these challenges involves:

  • Using fusion tags (His, GST, MBP) to improve solubility and facilitate purification

  • Implementing multiple chromatography steps (IMAC followed by size exclusion)

  • Including protease inhibitors throughout purification

  • Validating functional activity through enzyme-specific assays

How does antimicrobial resistance in Nocardia farcinica impact recombinant protein studies?

N. farcinica exhibits characteristic resistance to several extended-spectrum antimicrobials, which impacts experimental design in recombinant protein studies. This resistance is primarily mediated by β-lactamases with distinct biochemical properties:

β-Lactamase PropertiesCharacteristics in N. farcinica
Isoelectric pointsMajor bands at pI 4.56 and 4.49 (87% of strains)
Substrate preferencePrimarily penicillinases (hydrolyzing cephalosporins at ≤12% rate of penicillins)
Inhibitor susceptibilityHighly susceptible to clavulanic acid (IC50: 0.025-0.045 μg/ml)
Cellular locationPresent in both culture supernatants and cell extracts
InducibilityNon-inducible

When designing selection strategies for recombinant protein expression:

  • Avoid ampicillin as a selection marker for plasmid maintenance

  • Consider using alternative antibiotics like kanamycin or tetracycline

  • For expression in Nocardia species, utilize non-β-lactam antibiotics for selection

  • Be aware that antibiotic resistance genes may transfer horizontally in mixed cultures

The β-lactamases of N. farcinica are taxonomically distinct from those of other Nocardia species, supporting the species' unique identity and potentially affecting genetic manipulation strategies .

What immunological considerations are important when working with recombinant N. farcinica proteins?

Immunological aspects of N. farcinica proteins require careful consideration in recombinant protein studies:

  • Host immune response mechanisms: N. farcinica triggers production of GM-CSF and TNF-α in monocytes within 2-6 hours of exposure . This rapid response requires consideration in both in vitro and in vivo studies.

  • STAT5 phosphorylation pathway: N. farcinica induces STAT5 phosphorylation after approximately 1.5 hours, peaking at 2.5 hours in normal monocytes . This time course differs from direct GM-CSF stimulation, indicating a distinct signaling mechanism.

  • Autoantibody concerns: N. farcinica infection has been associated with anti-GM-CSF autoantibodies in some patients, which may complicate interpretation of immunological experiments .

  • Cross-reactivity assessment: When developing antibodies against recombinant N. farcinica proteins, cross-reactivity testing is essential. For example, NFA47630 protein is recognized by anti-N. farcinica and anti-N. cyriacigeorgica sera, but not by anti-N. asteroids, anti-N. brasiliensis, anti-N. nova or anti-M. bovis sera .

Methodological approach for immunological characterization of recombinant N. farcinica proteins:

  • Evaluate activation of MAPK signaling pathways (ERK, JNK, P38 phosphorylation)

  • Measure cytokine production (TNF-α, IL-10, IL-12, IFN-γ) in response to protein exposure

  • Assess neutrophil and whole blood killing assays to determine functional immunological effects

  • Verify species-specificity using sera from various related bacterial infections

How can structural analysis of Malate synthase G inform drug development against Nocardia infections?

Structural analysis of N. farcinica Malate synthase G provides valuable insights for structure-based drug design approaches:

  • Active site mapping: Crystal structures of glcB, even partial constructs, can reveal the architecture of the active site, including catalytic residues and substrate binding pockets.

  • Comparative analysis: Structural differences between human and bacterial enzymes can be exploited for selective inhibitor design following this methodology:

    • Superimpose structures of bacterial and mammalian homologs

    • Identify unique pockets or conformations in the bacterial enzyme

    • Design inhibitors that target these unique features

  • Virtual screening pipeline:

    • Generate a pharmacophore model based on active site configuration

    • Screen compound libraries against this model

    • Dock top candidates for binding energy calculation

    • Prioritize compounds for in vitro testing

  • Fragment-based approach: For enzymes like glcB, identification of fragment hits that bind to different regions of the active site can lead to more effective inhibitors through fragment linking or growing strategies.

The glyoxylate shunt, including Malate synthase G, represents an attractive target for antimicrobial development as it is essential for bacterial persistence but absent in humans. This pathway enables bacteria to utilize acetate or fatty acids as carbon sources during infection, making it crucial for pathogen survival in host tissues.

What are the optimal conditions for expressing functional recombinant N. farcinica glcB?

Based on successful expression of other N. farcinica proteins, the following methodological approach is recommended for functional glcB expression:

Expression optimization protocol:

  • Vector selection: pET system vectors with T7 promoter and appropriate fusion tags (His6, GST, or MBP)

  • Host strain selection:

    • E. coli BL21(DE3) for standard expression

    • E. coli Rosetta for rare codon optimization

    • E. coli Arctic Express for improved folding at lower temperatures

  • Induction parameters:

    • IPTG concentration: 0.2 mM (optimal based on other N. farcinica proteins)

    • Induction temperature: 28°C (reduces inclusion body formation)

    • Duration: 4-6 hours (standard) or overnight at reduced temperature (16-18°C)

  • Buffer optimization:

    • Include 5-10% glycerol to improve protein stability

    • Test multiple pH conditions (pH 7.0-8.5)

    • Evaluate metal ion requirements (Mg2+, Mn2+) for functional activity

  • Functional validation:

    • Enzymatic activity assay measuring condensation of acetyl-CoA with glyoxylate

    • Circular dichroism to confirm proper secondary structure

    • Size exclusion chromatography to verify oligomeric state

Experimental data indicates that expression temperature significantly impacts the quality of recombinant Nocardia proteins, with lower temperatures generally yielding more soluble and functional protein.

What PCR-based methods can be used to clone the glcB gene from Nocardia farcinica?

Cloning the glcB gene from N. farcinica requires careful consideration of the organism's high GC content and specific amplification challenges. A methodological approach includes:

  • Genomic DNA extraction:

    • Mechanical disruption (bead-beating) combined with enzymatic lysis

    • Purification using specialized kits for high-GC content bacteria

    • Quality verification through spectrophotometry (A260/A280 ratio)

  • Primer design considerations:

    • Account for high GC content (typically >60%)

    • Use OLIGO 4.0 or similar software for optimal primer design

    • Include restriction sites for directional cloning

    • Consider codon optimization for expression host

  • PCR optimization protocol:

    • Use high-fidelity polymerases designed for GC-rich templates

    • Include DMSO (5-10%) or betaine (1-2M) to reduce secondary structure

    • Implement touchdown PCR or two-step PCR protocols

    • Optimize annealing temperature through gradient PCR

  • Cloning verification:

    • Restriction enzyme digestion

    • Colony PCR screening

    • Sequence verification of the entire gene

This approach has been successfully used for amplification of specific gene fragments from N. farcinica, such as the 314-bp diagnostic fragment using primers Nf1 and Nf2 .

How can recombinant N. farcinica glcB be validated for functional activity?

Validating the functional activity of recombinant glcB requires multiple complementary approaches:

  • Enzymatic activity assays:

    • Spectrophotometric measurement of malate formation

    • Coupled enzyme assays tracking NADH oxidation

    • Isothermal titration calorimetry for binding studies

  • Structural validation:

    • Circular dichroism to confirm secondary structure

    • Thermal shift assays to assess protein stability

    • Limited proteolysis to verify proper folding

  • Complementation studies:

    • Expression in glcB-deficient bacterial strains

    • Restoration of growth on acetate or fatty acids as sole carbon sources

    • Metabolic flux analysis to confirm pathway functionality

  • Comparative analysis:

    • Side-by-side comparison with commercially available malate synthase enzymes

    • Determination of kinetic parameters (Km, Vmax, kcat)

    • Inhibitor sensitivity profiling

ParameterTypical Range for Functional glcBMethod of Determination
Km (glyoxylate)0.05-0.2 mMMichaelis-Menten kinetics
Km (acetyl-CoA)0.01-0.1 mMMichaelis-Menten kinetics
pH optimum7.5-8.5Activity vs. pH profile
Temperature optimum30-37°CActivity vs. temperature profile
Metal dependencyMg2+ (1-5 mM)Activity with/without metal ions

Each validation approach provides complementary evidence of proper folding and functional activity, essential for downstream applications in drug discovery or metabolic engineering.

What animal models are appropriate for studying recombinant N. farcinica proteins?

Selection of appropriate animal models for studying N. farcinica proteins should consider:

  • Mouse models: Most commonly used and well-validated

    • BALB/c mice: Suitable for immunological studies

    • C57BL/6: Useful for genetic manipulation studies

    • Immunocompromised models (e.g., SCID mice): Replicate opportunistic infection scenarios

  • Route of administration:

    • Intranasal inoculation (50 μl of bacterial suspension) mimics natural respiratory infection

    • Intravenous challenge for disseminated infection models

    • Subcutaneous inoculation for localized infection studies

  • Dosing considerations:

    • Typical challenge dose: 1 × 10^7 CFU for N. farcinica

    • Protein immunization: 2-3 doses at 10-day intervals

    • Adjuvant selection critical for immunogenicity

  • Outcome measurements:

    • Weight loss and body temperature monitoring

    • Bacterial burden in tissues (lung, spleen, liver)

    • Cytokine profiles in BALF and tissue homogenates

    • LDH assessment for tissue damage quantification

    • Survival rate analysis for vaccine efficacy studies

The mouse model has been validated for N. farcinica infection studies, with demonstrated utility in evaluating prophylactic vaccines. For example, mice immunized with recombinant N. farcinica proteins exhibited higher antibody titers, greater bacterial clearance, milder organ infection, and higher survival rates compared to control animals .

How can researchers assess the immunogenicity of recombinant N. farcinica proteins?

A comprehensive immunogenicity assessment of recombinant N. farcinica proteins involves multiple methodological approaches:

  • Antibody response characterization:

    • ELISA for antibody titer determination

    • Western blot for antibody specificity confirmation

    • Isotype analysis (IgG1, IgG2a, IgG2b, IgA) for response profiling

    • Avidity assays to determine antibody maturation

  • Cellular immune response evaluation:

    • Flow cytometry for T cell phenotyping

    • ELISpot for antigen-specific T cell quantification

    • Cytokine profiling (TNF-α, IL-10, IL-12, IFN-γ)

    • Proliferation assays to measure T cell activation

  • Functional immunological assays:

    • Whole blood killing assays to evaluate antibody-mediated clearance

    • Neutrophil killing assays to assess cellular immunity

    • Opsonophagocytic assays to measure antibody functionality

    • Complement activation studies

  • In vivo challenge studies:

    • Bacterial burden determination post-challenge

    • Histopathological examination of infected tissues

    • Survival rate analysis

    • Immune cell infiltration in target organs

This methodology has been successfully applied to other N. farcinica proteins, demonstrating that immunization can upregulate the phosphorylation status of ERK, JNK, P38 and elevate cytokine levels of TNF-α, IL-10, IL-12, and IFN-γ, correlating with protective immunity .

How should researchers interpret contradictory results in N. farcinica glcB activity assays?

When encountering contradictory results in enzyme activity assays, a methodical troubleshooting approach is essential:

  • Enzyme preparation variables:

    • Protein purity: Verify by SDS-PAGE and mass spectrometry

    • Storage conditions: Test fresh vs. stored preparations

    • Buffer composition: Systematically vary buffer components

    • Metal ion dependence: Screen different metal ions and concentrations

  • Assay condition variables:

    • Temperature effects: Perform temperature optimization studies

    • pH dependence: Create a detailed pH activity profile

    • Substrate quality: Use fresh reagents from multiple suppliers

    • Enzyme concentration: Verify linearity across enzyme concentrations

  • Instrument and detection variables:

    • Validate instrument calibration with standards

    • Compare different detection methods (direct vs. coupled assays)

    • Evaluate potential interference from assay components

    • Include appropriate internal controls

  • Biological sample variables:

    • Compare recombinant vs. native enzyme activity

    • Test different expression systems and purification methods

    • Evaluate the impact of fusion tags on activity

    • Consider post-translational modifications

For data reconciliation, a systematic matrix experiment varying multiple parameters simultaneously can identify interaction effects that might explain contradictory results. Statistical design of experiments (DoE) approaches are particularly valuable for identifying optimal conditions and resolving contradictions.

What statistical approaches are most appropriate for analyzing N. farcinica protein expression data?

Appropriate statistical analysis of protein expression data requires careful consideration of experimental design and data characteristics:

  • Experimental design considerations:

    • Include appropriate biological and technical replicates

    • Implement randomization to minimize batch effects

    • Include appropriate positive and negative controls

    • Consider factorial designs to identify interaction effects

  • Data preprocessing approaches:

    • Normalization methods (global normalization, LOESS, quantile)

    • Log-transformation for variance stabilization

    • Outlier detection and handling

    • Missing value imputation strategies

  • Statistical tests for hypothesis testing:

    • Student's t-test for pairwise comparisons

    • ANOVA for multiple condition comparison

    • Non-parametric alternatives (Mann-Whitney, Kruskal-Wallis)

    • Post-hoc tests with multiple testing correction (Bonferroni, FDR)

  • Advanced analytical methods:

    • Principal component analysis for dimension reduction

    • Hierarchical clustering for pattern identification

    • Partial least squares for correlation with biological outcomes

    • Time series analysis for temporal expression patterns

Statistical AnalysisApplicable ScenarioKey AssumptionsSoftware Tools
ANOVAMultiple expression conditionsNormal distribution, equal varianceR, GraphPad Prism
Repeated measures ANOVATime course experimentsSphericity, normalitySPSS, R
Linear mixed modelsNested experimental designsLinear relationships, normal residualsR (lme4), SAS
Survival analysisChallenge experimentsProportional hazardsR (survival), GraphPad Prism

Power analysis should be performed prior to experimentation to determine appropriate sample sizes, particularly for in vivo studies where variability may be high and ethical considerations limit animal numbers.

How can researchers determine if observed phenotypes are directly related to glcB function?

Establishing a causal relationship between N. farcinica glcB function and observed phenotypes requires a multi-faceted approach:

  • Genetic manipulation strategies:

    • Gene knockout: Complete deletion of glcB

    • Point mutations: Site-directed mutagenesis of catalytic residues

    • Conditional expression: Inducible or repressible systems

    • Complementation: Restoration of wild-type phenotype

  • Functional complementation:

    • Expression of wild-type glcB in knockout strains

    • Cross-species complementation with homologous enzymes

    • Domain swapping experiments to identify critical regions

    • Rescue with metabolic intermediates

  • Biochemical verification:

    • Metabolite profiling to track carbon flux

    • Isotope labeling to confirm pathway utilization

    • In vitro enzyme assays correlating with in vivo phenotypes

    • Protein-protein interaction studies to identify relevant complexes

  • Control experiments:

    • Exclude polar effects through transcriptional analysis

    • Rule out compensatory pathways through multi-gene analysis

    • Test growth on various carbon sources as metabolic controls

    • Include related enzymes as specificity controls

This systematic approach helps differentiate direct effects of glcB function from indirect consequences or experimental artifacts, establishing clear genotype-phenotype relationships essential for mechanistic understanding.

How might glcB function in Nocardia farcinica relate to pathogenicity?

The relationship between Malate synthase G function and N. farcinica pathogenicity involves several mechanistic connections:

  • Metabolic adaptation in host environments:

    • Enables growth on alternative carbon sources during infection

    • Facilitates survival during nutrient limitation in host tissues

    • Contributes to persistence in macrophages where primary carbon sources are restricted

  • Immunomodulatory effects:

    • Metabolic byproducts may influence host immune responses

    • Carbon flux through the glyoxylate shunt affects inflammatory mediator production

    • Bacterial metabolic state impacts susceptibility to host defense mechanisms

  • Therapeutic target potential:

    • Essential for bacterial survival under certain infection conditions

    • Absence in mammals makes it an attractive selective target

    • Inhibition may synergize with conventional antibiotics

  • Bacterial stress response:

    • Upregulation during oxidative stress conditions

    • Role in biofilm formation and maintenance

    • Contribution to antimicrobial tolerance states

Research has demonstrated that N. farcinica infection triggers specific immune responses, including GM-CSF and TNF-α production in monocytes , which may be influenced by metabolic state. The bacterium's ability to persist in immunocompromised hosts suggests sophisticated metabolic adaptation mechanisms potentially involving glcB and the glyoxylate shunt.

What are the emerging technologies for studying metabolic enzymes in Nocardia species?

Cutting-edge technologies are revolutionizing the study of metabolic enzymes in Nocardia species:

  • CRISPR-Cas9 genome editing:

    • Precise genetic manipulation in high-GC content organisms

    • Multiplexed gene targeting for pathway analysis

    • CRISPRi for tunable gene repression

    • Base editing for point mutations without double-strand breaks

  • Single-cell approaches:

    • Single-cell RNA-seq for population heterogeneity assessment

    • Single-cell metabolomics for individual cell metabolic profiling

    • Microfluidic devices for real-time enzyme activity monitoring

    • Single-cell proteomics for protein expression analysis

  • Advanced structural biology:

    • Cryo-EM for high-resolution protein structures without crystallization

    • Hydrogen-deuterium exchange mass spectrometry for dynamic structural information

    • AlphaFold2 and related AI tools for structure prediction

    • Time-resolved X-ray crystallography for reaction mechanism elucidation

  • Systems biology integration:

    • Multi-omics data integration (genomics, transcriptomics, proteomics, metabolomics)

    • Genome-scale metabolic modeling of Nocardia metabolism

    • Flux balance analysis for predicting metabolic capabilities

    • Machine learning approaches for predicting enzyme function

These technologies enable unprecedented insights into enzyme function in the context of cellular metabolism and host-pathogen interactions, accelerating both fundamental understanding and application development.

How does research on N. farcinica glcB contribute to broader understanding of bacterial metabolism?

Research on N. farcinica Malate synthase G contributes to broader understanding of bacterial metabolism through several important dimensions:

  • Evolutionary perspectives:

    • Comparative analysis across bacterial phyla reveals evolutionary conservation and divergence

    • Horizontal gene transfer patterns illuminate metabolic adaptation

    • Enzyme structure-function relationships provide insights into evolutionary constraints

    • Paralog analysis informs understanding of functional specialization

  • Metabolic network understanding:

    • Integration of glcB function into global metabolic models

    • Elucidation of regulatory networks controlling carbon flux

    • Identification of metabolic bottlenecks and control points

    • Understanding of metabolic robustness and redundancy

  • Host-pathogen interaction insights:

    • Bacterial adaptation to host nutritional immunity

    • Metabolic requirements for intracellular survival

    • Competition for nutrients in polymicrobial infections

    • Metabolic triggering of virulence factor expression

  • Biotechnological applications:

    • Enzyme engineering for improved catalytic properties

    • Metabolic engineering for bioproduction of valuable compounds

    • Development of novel biosensors based on metabolic enzymes

    • Identification of new antimicrobial targets and screening approaches

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 2024 Thebiotek. All Rights Reserved.