Recombinant Legionella pneumophila subsp. pneumophila Probable glycine dehydrogenase [decarboxylating] subunit 1 (gcvPA)

Shipped with Ice Packs
In Stock

Product Specs

Form
Lyophilized powder
Lead Time
Delivery times vary depending on the purchasing method and location. Please contact your local distributor for specific delivery timelines.
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%.
Shelf Life
Shelf life depends on various factors including storage conditions, buffer composition, 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
Upon receipt, store at -20°C/-80°C. Aliquot for multiple uses. Avoid repeated freeze-thaw cycles.
Tag Info
The tag type is determined during manufacturing. If you require a specific tag, please inform us, and we will prioritize its development.
Synonyms
gcvPA; lpg0116Probable glycine dehydrogenase; decarboxylating) subunit 1; EC 1.4.4.2; Glycine cleavage system P-protein subunit 1; Glycine decarboxylase subunit 1; Glycine dehydrogenase; aminomethyl-transferring) subunit 1
Buffer Before Lyophilization
Tris/PBS-based buffer, 6% Trehalose.
Datasheet
Please contact us to get it.
Expression Region
1-456
Protein Length
full length protein
Purity
>85% (SDS-PAGE)
Species
Legionella pneumophila subsp. pneumophila (strain Philadelphia 1 / ATCC 33152 / DSM 7513)
Target Names
gcvPA
Target Protein Sequence
MPYIPHTPDD TKEMLEAIGA QDIQDLFDEI PASLQYAGFQ NIPAGINEME MLKEAQNQAQ KNRNGICFIG AGCYEHHIPA AVWDIASRGE FLTAYTPYQA EASQGTLQLL YEYQTMICEL TGMEVSNASM YDGATALAEA VLMAVRLNKH SKTNRVLIAG TVHPFYRETI ETIVRNQHIE VITLPFDEQQ GITNLGSLNQ YTGEDITALV IAQPNFFGCL EQVDKMTSWA HHNKTISVAC VNPTSLALLK PPGSWGEHGV DIVCGEGQPL GSPMASGGPY FGFLSTRMAH VRQMPGRIIG RTVDKDGKTG FSLTLQAREQ HIRRAKATSN ICTNQGLLVT AATIYMSLLG PEGLSQVATQ CHQNTHELIT ALTQIEGVEL AFKAPFFHEA LIKLNQPVQS VLQQLADAGI AGGYAPEQHY PQLANTLLVC ATEVRTAEDI AKYAKTLKTI MSKRGA
Uniprot No.

Target Background

Function
The glycine cleavage system facilitates glycine degradation. The P protein binds glycine's alpha-amino group via its pyridoxal phosphate cofactor; CO₂ is released, and the remaining methylamine moiety transfers to the lipoamide cofactor of the H protein.
Database Links

KEGG: lpn:lpg0116

STRING: 272624.lpg0116

Protein Families
GcvP family, N-terminal subunit subfamily

Q&A

What is the function of glycine dehydrogenase (gcvPA) in Legionella pneumophila?

Glycine dehydrogenase [decarboxylating] subunit 1 (gcvPA) is a critical component of the glycine cleavage system (GCS) in Legionella pneumophila. This multienzyme complex catalyzes the decarboxylation of glycine and transfers a one-carbon unit into folate one-carbon metabolism. The GCS consists of four proteins: P-protein (glycine decarboxylase), H-protein (a carrier protein), T-protein (aminomethyltransferase), and L-protein (dihydrolipoamide dehydrogenase) .

In L. pneumophila, this system plays several important roles:

  • Contributing to bacterial metabolism and energy production

  • Potentially supporting survival within host cells

  • Providing one-carbon units for nucleotide biosynthesis

  • Regulating glycine levels, which might impact bacterial growth and virulence

Unlike many other bacterial species, genome analysis suggests that L. pneumophila's glycine metabolism system may have evolved specific adaptations that support its intracellular lifestyle and pathogenicity .

How is gcvPA structured and organized in the Legionella pneumophila genome?

The gcvPA gene in L. pneumophila is part of the glycine cleavage system operon. Genomic analysis reveals that unlike some bacterial species that have a single glycine decarboxylase gene, L. pneumophila contains distinct genomic organization. This organization reflects the evolutionary adaptations that support its dual lifestyle as both an environmental organism and human pathogen .

The gene structure typically includes:

  • Promoter regions that may respond to metabolic signals

  • Open reading frame encoding the gcvPA protein

  • Regulatory elements that coordinate expression with other GCS components

  • Potential integration with pathways involved in virulence and stress response

Comparative genomic studies between L. pneumophila and other Legionella species have identified species-specific genetic differences, which may contribute to the unique intracellular lifestyle and pathogenic potential of L. pneumophila .

What experimental evidence confirms the role of gcvPA in L. pneumophila metabolism?

While direct experimental evidence specifically for gcvPA in L. pneumophila is limited in the provided references, similar glycine decarboxylase systems have been well-characterized in other organisms. Studies of glycine decarboxylase function in model organisms demonstrate its essential role in glycine metabolism and one-carbon transfer reactions .

Research approaches that have been used to confirm the function include:

  • Metabolic labeling experiments tracing the fate of glycine

  • Genetic disruption studies analyzing phenotypic changes

  • Enzyme activity assays measuring glycine cleavage

  • Protein interaction studies identifying binding partners

Understanding the function in L. pneumophila specifically would require targeted experimental approaches drawing on methodology established in model systems.

What are the optimal conditions for expressing recombinant L. pneumophila gcvPA in E. coli?

For successful expression of recombinant L. pneumophila gcvPA in E. coli, researchers should consider the following optimized protocol:

Expression System Design:

  • Vector selection: pET-based vectors with T7 promoter systems are recommended for high-level expression

  • Strain selection: BL21(DE3) or Rosetta strains accommodate potential codon bias in Legionella genes

  • Fusion tags: N-terminal 6×His tag facilitates purification while minimizing interference with protein folding

Culture Conditions:

  • Temperature: 18-20°C post-induction (reducing inclusion body formation)

  • Media: Enriched media (e.g., Terrific Broth) with appropriate antibiotics

  • Induction: 0.1-0.5 mM IPTG at OD600 ~0.6-0.8

  • Duration: 16-18 hours post-induction at reduced temperature

Extraction Parameters:

  • Lysis buffer: 50 mM Tris-HCl pH 8.0, 300 mM NaCl, 10% glycerol, 1 mM DTT

  • Protease inhibitors: Complete protease inhibitor cocktail

  • Solubilization: Gentle detergents (0.1% Triton X-100) may improve yield of soluble protein

This approach draws on principles of heterologous protein expression while addressing specific characteristics of Legionella proteins. Verification of expression should include SDS-PAGE, western blot, and activity assays.

How can I purify recombinant gcvPA while maintaining its enzymatic activity?

Purification of recombinant gcvPA requires careful consideration of protein stability and activity. The following methodology is recommended:

Purification Strategy:

  • Immobilized metal affinity chromatography (IMAC) using Ni-NTA resin for His-tagged protein

  • Size exclusion chromatography to remove aggregates and ensure homogeneity

  • Ion exchange chromatography as a polishing step

Buffer Optimization:

  • Purification buffer: 50 mM HEPES pH 7.5, 300 mM NaCl, 10% glycerol, 1 mM DTT

  • Stabilizing additives: Consider pyridoxal 5'-phosphate (PLP, 0.1 mM) as gcvPA is likely PLP-dependent

  • Storage buffer: 25 mM HEPES pH 7.5, 150 mM NaCl, 20% glycerol, 1 mM DTT at -80°C

Activity Preservation:

  • Avoid freeze-thaw cycles (aliquot before freezing)

  • Maintain reducing conditions throughout purification

  • Consider co-purification with other glycine cleavage system components for enhanced stability

Verification Methods:

  • Purity assessment: SDS-PAGE, mass spectrometry

  • Activity assay: Measure glycine-dependent CO2 release or NAD+ reduction

  • Structural integrity: Circular dichroism, thermal shift assays

This purification protocol balances yield with preservation of enzymatic activity, which is critical for downstream functional studies.

What assays can accurately measure gcvPA enzymatic activity in vitro?

Several complementary approaches can be used to measure gcvPA enzymatic activity:

Spectrophotometric Assays:

  • NAD+ reduction assay: Monitor NADH formation at 340 nm as the glycine cleavage reaction proceeds

  • Coupled enzyme assays: Link glycine decarboxylation to a secondary reaction with spectrophotometric readout

Direct Product Measurement:

  • CO2 evolution: Capture and quantify 14C-labeled CO2 released from [1-14C]glycine

  • H-protein lipoylation state: Monitor the redox state of the lipoamide group on the H-protein

Table 1: Comparison of Methods for Measuring gcvPA Activity

MethodSensitivityAdvantagesLimitationsEquipment Required
NAD+ ReductionModerateReal-time measurement, Simple setupRequires full GCS complexSpectrophotometer
14CO2 EvolutionHighDirect measurement of decarboxylationRequires radioactive materialsScintillation counter
Coupled EnzymeHighAmplified signalPotential interference from coupling enzymesSpectrophotometer
Mass SpectrometryVery HighCan track multiple metabolitesComplex sample preparation, ExpensiveLC-MS/MS

For complete characterization, the glycine cleavage reaction requires all four components of the glycine cleavage system (P, H, T, and L proteins). Reconstitution of the complete system may be necessary for accurate activity measurements .

How can I construct and validate a gcvPA knockout mutant in L. pneumophila?

Creating a gcvPA knockout mutant in L. pneumophila requires leveraging the organism's natural competence for DNA transformation. The following methodology is recommended:

Mutant Construction Strategy:

  • Allelic exchange: Design a construct with upstream and downstream flanking regions of gcvPA with an antibiotic resistance cassette

  • Transformation: Utilize L. pneumophila's natural competence system for uptake of the knockout construct

  • Selection: Identify transformants on selective media containing appropriate antibiotics

  • Verification: Confirm gene deletion by PCR, sequencing, and Southern blot analysis

Transformation Protocol:

  • Culture L. pneumophila to early stationary phase (OD600 ~1.8-2.0) in BYE medium

  • Mix 1-5 μg of knockout construct DNA with 1 ml of bacterial culture

  • Incubate at 30°C for 2-3 days on selective BCYE agar plates

  • Verify transformation using L. pneumophila's type IV pili-dependent DNA uptake system

Validation Approaches:

  • Molecular verification: PCR amplification across deletion junctions

  • Transcriptional analysis: RT-PCR or RNA-seq to confirm absence of gcvPA transcript

  • Protein analysis: Western blot using anti-gcvPA antibodies

  • Phenotypic characterization: Growth curves, intracellular replication assays, and metabolite profiling

This approach leverages L. pneumophila's natural competence mechanism, which is associated with the expression of type IV pili under specific growth conditions .

What phenotypes might be expected in a L. pneumophila gcvPA mutant?

Based on the functional role of gcvPA in glycine metabolism and one-carbon transfer, a gcvPA knockout mutant would likely exhibit several distinct phenotypes:

Metabolic Phenotypes:

  • Glycine accumulation due to impaired glycine catabolism

  • Altered one-carbon metabolism affecting nucleotide biosynthesis

  • Potential growth defects in glycine-rich environments

Intracellular Growth Phenotypes:

  • Reduced replication in macrophages and amoebae hosts

  • Altered vacuole formation or trafficking

  • Modified stress resistance profiles

Virulence Phenotypes:

  • Potential attenuation in infection models

  • Changes in expression of virulence factors

  • Altered ability to manipulate host cell processes

The phenotype severity may vary depending on environmental conditions and availability of alternative metabolic pathways. For example, while L. pneumophila RpoS mutants show defects in replication within Acanthamoeba castellanii , gcvPA mutants might exhibit distinct but related phenotypes based on their metabolic functions.

How does natural competence in L. pneumophila affect recombinant gcvPA studies?

L. pneumophila's natural competence for DNA transformation provides both opportunities and challenges for recombinant gcvPA studies:

Advantages for Research:

  • Direct transformation: Introducing modified gcvPA constructs without electroporation

  • Homologous recombination: Facilitating precise genomic modifications at the native locus

  • Complementation studies: Reintroducing wild-type gcvPA in mutant backgrounds

Key Considerations:

  • Transformation efficiency correlates with expression of type IV pili (CAP)

  • DNA uptake may not require specific uptake sequences, but transformation frequency is affected by competing DNA

  • The pilEL locus is required for competence, suggesting molecular connections between adhesion and DNA uptake mechanisms

Methodological Approach:

  • Optimize transformation by culturing L. pneumophila under conditions that promote type IV pili expression

  • Design constructs with sufficient homologous sequence (>500 bp) flanking the gcvPA region

  • Consider the timing of transformation relative to growth phase for maximal efficiency

Understanding these natural competence mechanisms provides valuable tools for genetic manipulation in L. pneumophila and informs experimental design for gcvPA studies .

How does gcvPA contribute to L. pneumophila pathogenesis and host-pathogen interactions?

The role of gcvPA in L. pneumophila pathogenesis likely involves several interconnected mechanisms:

Metabolic Support During Infection:

  • Providing one-carbon units essential for nucleotide synthesis during intracellular replication

  • Contributing to amino acid metabolism in nutrient-limited intracellular environments

  • Supporting bacterial adaptation to changing metabolic conditions within the host

Potential Immunomodulatory Effects:

  • Modulation of glycine levels may affect host cell signaling

  • Metabolic products from glycine cleavage might influence host responses

  • Interaction with host mitochondrial metabolism during infection

Integration with Virulence Systems:

  • L. pneumophila contains many unique genes, including the dot/icm effector lepB and various virulence determinants

  • Metabolism and virulence are often coordinated through regulatory networks

  • The specific genomic context of gcvPA in L. pneumophila suggests potential co-regulation with virulence factors

Research approaches to investigate these connections include:

  • Infection studies comparing wild-type and gcvPA mutants

  • Transcriptomic analysis during different stages of infection

  • Metabolomic profiling of host cells during infection

  • Identification of potential protein-protein interactions between gcvPA and host factors

Understanding these pathogenesis mechanisms requires integration of metabolic analysis with infection models to establish the complete picture of gcvPA's role.

How can structural analysis of gcvPA inform inhibitor design for antimicrobial development?

Structural analysis of gcvPA provides critical insights for rational inhibitor design:

Structural Characterization Approaches:

  • X-ray crystallography of purified recombinant gcvPA

  • Cryo-EM analysis of the complete glycine cleavage complex

  • Homology modeling based on related glycine decarboxylase structures

  • Molecular dynamics simulations to identify conformational changes during catalysis

Key Structural Features for Inhibitor Design:

  • Active site architecture containing the pyridoxal phosphate (PLP) cofactor binding pocket

  • Substrate binding regions specific to glycine

  • Protein-protein interaction interfaces with other GCS components

  • Unique structural elements distinguishing bacterial from human homologs

Inhibitor Development Strategy:

  • Structure-based virtual screening targeting active site or allosteric pockets

  • Fragment-based approaches identifying building blocks for inhibitor design

  • Rational modification of known glycine analogs or PLP-dependent enzyme inhibitors

  • Evaluation of selectivity against human glycine decarboxylase to minimize toxicity

This structural information guides the development of small molecules that can selectively inhibit gcvPA function, potentially leading to novel antimicrobials targeting L. pneumophila metabolism.

How can systems biology approaches integrate gcvPA function within the broader metabolic network of L. pneumophila?

Systems biology provides powerful frameworks to understand gcvPA within L. pneumophila's metabolic network:

Multi-omics Integration:

  • Transcriptomics: RNA-seq to identify co-regulated genes under various conditions

  • Proteomics: Quantitative analysis of protein abundance and post-translational modifications

  • Metabolomics: Profiling metabolite changes in wild-type versus gcvPA mutants

  • Fluxomics: Measuring metabolic flux distributions using isotope labeling

Network Analysis Approaches:

  • Reconstruction of genome-scale metabolic models incorporating gcvPA reactions

  • Flux balance analysis to predict growth phenotypes under different conditions

  • Identification of synthetic lethal interactions with gcvPA

  • Regulatory network analysis connecting metabolism with virulence programs

Table 2: Systems Biology Datasets Relevant to gcvPA Function

Data TypeExperimental ApproachInformation GainedIntegration Points
TranscriptomicsRNA-seq across growth conditionsCo-expression networksRegulatory mechanisms
ProteomicsMass spectrometryProtein abundance changesPost-transcriptional control
MetabolomicsLC-MS/MSMetabolite profilesPathway activities
InteractomicsCo-IP, PPI screensProtein interaction partnersFunctional complexes
PhenomicsGrowth/virulence assaysPhysiological outcomesPhenotype prediction

These systems approaches reveal how gcvPA function is coordinated with other metabolic pathways and virulence mechanisms, providing a comprehensive understanding of its role in L. pneumophila biology.

Why might recombinant gcvPA show limited activity compared to native enzyme, and how can this be addressed?

Several factors may contribute to limited activity of recombinant gcvPA compared to the native enzyme:

Common Challenges:

  • Incorrect folding or post-translational modifications in heterologous expression systems

  • Absence of other glycine cleavage system components required for full activity

  • Suboptimal assay conditions that don't reflect the native bacterial environment

  • Loss of cofactors (particularly pyridoxal phosphate) during purification

Optimization Strategies:

  • Co-expression with chaperones to improve folding (GroEL/GroES system)

  • Reconstitution with other purified GCS components (H, T, and L proteins)

  • Addition of cofactors (0.1 mM pyridoxal phosphate) to purification and assay buffers

  • Expression at lower temperatures (16-18°C) to promote proper folding

  • Testing different solubilization and stabilization additives (glycerol, low concentrations of detergents)

Expression System Alternatives:

  • Consider using closer relatives of Legionella as expression hosts

  • Explore cell-free expression systems that can be optimized for membrane proteins

  • Test expression of different protein constructs with varying N- and C-terminal boundaries

Systematic optimization of these parameters can significantly improve recombinant protein activity, bringing it closer to native enzyme functionality.

What are the critical controls needed when studying gcvPA function in L. pneumophila?

Genetic Controls:

  • Wild-type L. pneumophila strain (positive control)

  • Clean deletion mutant (ΔgcvPA)

  • Complemented strain (ΔgcvPA + gcvPA) to confirm phenotype reversibility

  • Point mutant with catalytically inactive gcvPA to distinguish enzymatic from structural roles

Biochemical Controls:

  • Enzyme assays with heat-inactivated enzyme (negative control)

  • Substrate specificity controls using non-glycine amino acids

  • Cofactor dependence controls (±pyridoxal phosphate)

  • Reconstitution controls with defined combinations of GCS components

Experimental Validation Controls:

  • Multiple biological and technical replicates

  • Alternative assay methods confirming the same result

  • Growth medium controls to rule out media composition effects

  • Time course analyses to capture dynamic changes

This comprehensive control strategy ensures that observed phenotypes can be confidently attributed to gcvPA function rather than experimental artifacts or secondary effects.

How can contradictory results in gcvPA research be reconciled?

Contradictory findings in gcvPA research may arise from several sources and can be reconciled through systematic investigation:

Sources of Contradiction:

  • Strain differences: Genetic background variations between laboratory strains

  • Growth conditions: Differences in media composition or culture conditions

  • Assay methodologies: Variations in sensitivity or specificity of detection methods

  • Regulatory context: Environmental factors affecting gcvPA expression or activity

Reconciliation Approach:

  • Direct side-by-side comparison of strains and methodologies

  • Controlled environmental variables across experiments

  • Examination of strain-specific genetic modifications

  • Integration of multiple experimental approaches to test hypotheses

Case Study Analysis:
When contradictory results emerge, construct a detailed comparison table documenting all experimental variables:

Table 3: Framework for Reconciling Contradictory Results

Experimental FactorStudy AStudy BPotential Impact on Results
L. pneumophila strainPhiladelphia-1ParisGenomic differences affecting regulation
Growth mediumBYEAYENutrient composition affecting metabolism
Growth phase at harvestExponentialStationaryRpoS-dependent regulation
Temperature37°C30°CExpression of type IV pili affecting transformation
Assay methodDirect enzyme activityGrowth phenotypeDifferent sensitivity and specificity

By systematically addressing these variables, researchers can identify the specific conditions under which seemingly contradictory results can be reconciled, advancing understanding of gcvPA function.

How might CRISPR-Cas9 technology advance genetic studies of gcvPA in L. pneumophila?

CRISPR-Cas9 technology offers transformative potential for gcvPA research in L. pneumophila:

Advanced Genetic Manipulation:

  • Precise editing: Creating clean deletions, point mutations, or tagged versions at the native locus

  • Multiplexed modifications: Targeting gcvPA alongside related genes to study pathway interactions

  • Conditional regulation: CRISPRi/CRISPRa systems for tunable repression or activation of gcvPA

Implementation Strategy:

  • Deliver Cas9 and sgRNA via vectors compatible with L. pneumophila's natural competence system

  • Design repair templates with homology arms optimized for L. pneumophila recombination

  • Incorporate selectable markers that can be subsequently removed

Potential Applications:

  • Domain-specific mutagenesis to identify functional regions of gcvPA

  • Scarless tagging for live-cell imaging of protein localization

  • Temporal control of gene expression during infection processes

  • High-throughput screening of gcvPA interaction partners

This technology would complement existing genetic approaches in L. pneumophila while providing unprecedented precision and efficiency for genetic manipulation.

What potential exists for utilizing gcvPA in synthetic biology applications?

The glycine cleavage system including gcvPA has several promising applications in synthetic biology:

Metabolic Engineering Applications:

  • One-carbon metabolism enhancement: Increasing flux through pathways dependent on one-carbon units

  • Glycine utilization: Engineering microbes to use glycine as a primary carbon or nitrogen source

  • Co-factor regeneration systems: Coupling glycine oxidation to NADH production for biocatalysis

Biosensor Development:

  • Designing gcvPA-based biosensors for glycine detection in environmental samples

  • Coupling glycine detection to reporter gene expression for diagnostic applications

  • Creating whole-cell biosensors for monitoring glycine levels in complex environments

Synthetic Pathway Construction:

  • Incorporation of modified gcvPA into artificial metabolic pathways

  • Development of minimal glycine metabolism modules for synthetic cells

  • Creation of orthogonal metabolic systems that function independently from host metabolism

The unique properties of the glycine cleavage system components make them valuable building blocks for synthetic biology applications ranging from bioproduction to biosensing.

How might cross-species comparative analysis of gcvPA advance understanding of its role in bacterial pathogenesis?

Comparative analysis across bacterial species provides valuable insights into gcvPA evolution and function:

Evolutionary Analysis Approaches:

  • Phylogenetic analysis of gcvPA sequences across pathogenic and non-pathogenic bacteria

  • Identification of conserved and variable regions correlating with pathogenicity

  • Analysis of horizontal gene transfer events that may have shaped gcvPA evolution

  • Examination of selection pressures acting on different functional domains

Functional Comparison Framework:

  • Complement L. pneumophila gcvPA mutants with orthologs from other species

  • Compare enzyme kinetics and substrate specificity across evolutionarily diverse gcvPA proteins

  • Analyze regulatory networks controlling gcvPA expression in different bacterial species

  • Correlate gcvPA sequence variations with host range and pathogenicity

Translational Relevance:

  • Identification of pathogen-specific features that could be targeted therapeutically

  • Understanding how metabolic adaptations contribute to host-specific virulence

  • Revealing convergent evolution of metabolic systems supporting intracellular lifestyles

This comparative approach provides context for understanding how gcvPA function has been adapted through evolution to support the specific lifestyle and pathogenic mechanisms of L. pneumophila.

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.