Recombinant Legionella pneumophila subsp. pneumophila N-succinylglutamate 5-semialdehyde dehydrogenase (astD)

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 consult your local distributor for precise delivery estimates.

Note: All proteins are shipped with standard blue ice packs unless dry ice shipping is requested in advance. Additional fees apply for dry ice shipping.

Notes
Avoid repeated freeze-thaw cycles. Store working aliquots at 4°C for up to one week.
Reconstitution
Centrifuge the vial briefly before opening to collect the contents. Reconstitute the protein in sterile, deionized water to a concentration of 0.1-1.0 mg/mL. For long-term storage, we recommend adding 5-50% glycerol (final concentration) and aliquoting at -20°C/-80°C. Our standard glycerol concentration is 50% and can serve as a guideline.
Shelf Life
Shelf life depends on various factors, including 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
Upon receipt, store at -20°C/-80°C. Aliquoting is recommended 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. To ensure a specific tag type, please inform us during your order placement; we will prioritize development accordingly.

Synonyms
astD; lpg1707N-succinylglutamate 5-semialdehyde dehydrogenase; EC 1.2.1.71; Succinylglutamic semialdehyde dehydrogenase; SGSD
Buffer Before Lyophilization
Tris/PBS-based buffer, 6% Trehalose.
Datasheet
Please contact us to get it.
Expression Region
1-496
Protein Length
full length protein
Purity
>85% (SDS-PAGE)
Species
Legionella pneumophila subsp. pneumophila (strain Philadelphia 1 / ATCC 33152 / DSM 7513)
Target Names
astD
Target Protein Sequence
MMSKLQIIQS KGQYINGEWI KGNGLILEST NPASGTLLWQ GNNATDEEIA NACYVAHRAL KSWANTSFEE RARYTKAFVE QVEKNREQLA RLISLETGKP LWESQTEVSS VIGKVNLSIQ AYQERTWPKQ TETAEANACL RFKPHGIVVV LGAFNFPAHL SNGHIVPALL AGNTVLYKPS EHTPAVAELI IQCWHDSGLP PGVINCLQGN ANCGNTLLSQ DIQGVYFTGS YATGLRIHQQ FCNRPEVILA LEMGGNNPLV IDEVKDIDAA VYHTILSTMI TAGQRCTCAR RIIIPDSQTG DLFLERFAKA CKLMRIGSFD SQPEPFIGPV ISHVQALKHL HAQKQLIEMG GEIILPMSLL LEYTGLVSPG IIDMTRAKNP PDEEIFAPFA QIYRYNHFDE AIQLANQTRY GLSAGLLSDN KDHYQQFYQN IRAGLINWNR PTTGAASSLP FGGVGCSGNH RPSAYFAADY CAYPVASMEQ PLLTTPVQRL PGLVLE
Uniprot No.

Target Background

Function

Catalyzes the NAD-dependent reduction of succinylglutamate semialdehyde to succinylglutamate.

Database Links

KEGG: lpn:lpg1707

STRING: 272624.lpg1707

Protein Families
Aldehyde dehydrogenase family, AstD subfamily

Q&A

What is N-succinylglutamate 5-semialdehyde dehydrogenase (astD) and what is its functional role in Legionella pneumophila metabolism?

N-succinylglutamate 5-semialdehyde dehydrogenase (astD) is an oxidoreductase enzyme (EC 1.2.1.71) that catalyzes the conversion of N-succinyl-L-glutamate 5-semialdehyde to N-succinyl-L-glutamate while reducing NAD+ to NADH. This reaction is represented by:

N-succinyl-L-glutamate 5-semialdehyde + NAD+ + H₂O → N-succinyl-L-glutamate + NADH + 2H+

The enzyme plays a critical role in the arginine and proline metabolism pathways in Legionella pneumophila. It is also known by several other names including succinylglutamic semialdehyde dehydrogenase, SGSD, AruD, and AstD . In the context of L. pneumophila pathogenicity, this enzyme contributes to bacterial survival within host cells through regulation of metabolic pathways essential for intracellular replication. L. pneumophila is an intracellular parasite that invades and proliferates within different eukaryotic cells, including human alveolar macrophages , and astD may contribute to these processes through its metabolic functions.

What expression systems are most effective for recombinant production of Legionella pneumophila astD?

The selection of an appropriate expression system is critical for successful production of recombinant L. pneumophila astD. Based on current research in recombinant protein production, two bacteriophage-derived promoters are extensively used for protein expression in E. coli: T5 and T7 promoters .

T5 Promoter System:

  • Utilizes the host RNA polymerase for transcription

  • Offers wider utility across E. coli strains without requiring additional genetic modifications

  • Provides more balanced expression, which can be advantageous for soluble protein production

T7 Promoter System:

  • Requires co-expression of T7 RNA polymerase in the host

  • Provides a dedicated RNA polymerase enabling exclusive expression of genes under T7 promoter control

  • Often yields higher expression levels but may lead to increased inclusion body formation

When expressing recombinant astD, the choice between these systems should consider:

  • Required protein yield

  • Need for soluble, active enzyme

  • Potential toxicity to the host cell

  • Post-translational modification requirements

For astD specifically, the T5 promoter system may offer advantages if the goal is to obtain active enzyme, as the more moderate expression level could reduce the formation of inclusion bodies and metabolic burden on the host cell.

How can researchers optimize purification protocols for recombinant Legionella pneumophila astD?

Optimizing purification of recombinant L. pneumophila astD requires careful consideration of the enzyme's biochemical properties. A methodological approach should include:

Initial Purification Strategy:

  • Affinity chromatography using His-tag or other fusion tags (GST, MBP)

  • Ion exchange chromatography based on the theoretical pI of astD

  • Size exclusion chromatography as a polishing step

Critical Parameters to Optimize:

  • Buffer composition: Test buffers containing components that stabilize oxidoreductases, such as reducing agents (DTT, β-mercaptoethanol)

  • pH optimization: Test range from pH 6.5-8.0 to identify optimal stability conditions

  • Salt concentration: Typically 100-300 mM NaCl works well for initial solubilization

  • Inclusion of glycerol (10-20%) to enhance protein stability

Activity Preservation:

  • Include cofactor (NAD+) at low concentrations (0.1-1 mM) in purification buffers

  • Avoid extended exposure to room temperature; maintain samples at 4°C

  • Consider addition of protease inhibitors to prevent degradation

The purification protocol should be validated by assessing enzyme activity at each step to ensure that the purification process preserves the catalytic function of astD.

What are the standard methods for assessing the enzymatic activity of recombinant astD?

Enzymatic activity of recombinant astD can be measured using spectrophotometric assays that track the reduction of NAD+ to NADH, which can be monitored by the increase in absorbance at 340 nm. A standard assay protocol includes:

Components of the Reaction Mixture:

  • Buffer: 50 mM Tris-HCl or phosphate buffer (pH 7.5-8.0)

  • Substrate: N-succinyl-L-glutamate 5-semialdehyde (typically 0.1-1 mM)

  • Cofactor: NAD+ (typically 1-2 mM)

  • Enzyme: Purified recombinant astD (concentration to be optimized)

  • Total volume: 200-1000 μL

Procedure:

  • Prepare reaction mixture without enzyme

  • Establish baseline at 340 nm

  • Add enzyme to initiate reaction

  • Monitor increase in absorbance at 340 nm over time

  • Calculate enzyme activity using extinction coefficient of NADH (6,220 M⁻¹cm⁻¹)

Activity Calculation:
Enzyme activity (U/mg) = (ΔA₃₄₀/min × reaction volume)/(6.22 × enzyme amount × path length)

Where one unit (U) is defined as the amount of enzyme that catalyzes the formation of 1 μmol of NADH per minute under the specified conditions.

How does the structure of astD relate to its function in Legionella pneumophila?

While the specific three-dimensional structure of L. pneumophila astD has not been fully elucidated, functional predictions can be made based on homology to other members of the aldehyde dehydrogenase family. Key structural features likely include:

Predicted Structural Elements:

  • A Rossmann fold for NAD+ binding, characterized by a βαβαβ motif

  • A catalytic domain containing conserved active site residues

  • An oligomerization domain that may facilitate dimer or tetramer formation

Functional Domains and Motifs:

  • NAD+-binding motif: Typically GXGXXG sequence

  • Catalytic cysteine residue: Essential for nucleophilic attack on the substrate

  • Conserved glutamate residue: Important for activating water molecule for hydrolysis

The enzyme likely functions through a mechanism involving:

  • Binding of the substrate (N-succinyl-L-glutamate 5-semialdehyde)

  • Nucleophilic attack by the catalytic cysteine

  • Hydride transfer to NAD+

  • Hydrolysis of the thiohemiacetal intermediate

  • Release of the product (N-succinyl-L-glutamate) and NADH

Understanding this structure-function relationship is critical for developing inhibitors or for engineering the enzyme for biotechnological applications.

How does astD contribute to Legionella pneumophila pathogenesis and intracellular survival?

The role of astD in L. pneumophila pathogenesis likely involves its function in arginine and proline metabolism, which may contribute to bacterial adaptation within host cells. While direct evidence linking astD to pathogenicity is limited, several aspects warrant investigation:

Potential Contributions to Pathogenesis:

  • Metabolic adaptation: astD may help L. pneumophila utilize specific nitrogen sources within the host cell environment

  • Stress response: The enzyme might participate in metabolic pathways that enable bacterial survival under stress conditions

  • Integration with virulence mechanisms: Metabolic processes involving astD could be coordinated with virulence factor expression

Legionella pneumophila has evolved sophisticated mechanisms to modulate host cell functions, including inhibition of phagosome acidification, prevention of phagosome-lysosome fusion, and suppression of oxidative burst . While these mechanisms are primarily associated with secreted effector proteins, metabolic enzymes like astD may indirectly support these processes by maintaining bacterial fitness during infection.

Research has shown that L. pneumophila produces virulence factors such as a glucosyltransferase that modifies host elongation factor 1A, inhibiting protein synthesis . Similarly, astD's role in arginine metabolism might influence bacterial-host interactions through modulation of amino acid pools or by affecting signaling pathways dependent on arginine availability.

What approaches can be used to investigate the impact of astD gene knockout on Legionella pneumophila virulence?

Investigating the impact of astD gene knockout on L. pneumophila virulence requires a multifaceted approach combining genetic manipulation, cellular assays, and animal models:

Genetic Manipulation Strategies:

  • Allelic exchange mutagenesis to create clean deletion mutants

  • CRISPR-Cas9 genome editing for precise modifications

  • Complementation studies to verify phenotypes are specifically due to astD deletion

In Vitro Virulence Assays:

  • Intracellular replication in macrophages and amoebae

    • Quantify bacterial numbers at different time points post-infection

    • Compare wild-type vs. ΔastD mutant growth curves

  • Phagosome maturation analysis

    • Fluorescence microscopy to track phagosome-lysosome fusion

    • Quantification of phagosomal markers (LAMP-1, Rab7)

Animal Model Studies:
The mouse model of pulmonary legionellosis can provide insights into the role of astD in vivo:

  • Administer wild-type and ΔastD strains via intranasal route

  • Monitor bacterial burden in lungs at different time points

  • Assess histopathological changes and inflammatory responses

  • Measure survival rates

This comprehensive approach would help determine whether astD is essential for L. pneumophila virulence or if it plays a more subtle role in bacterial fitness during infection.

How can proteomics and metabolomics approaches be integrated to understand the role of astD in Legionella pneumophila metabolism?

Integrating proteomics and metabolomics offers powerful insights into the metabolic networks involving astD in L. pneumophila:

Proteomic Approaches:

  • Comparative proteomics of wild-type vs. ΔastD strains

    • Identify proteins with altered abundance

    • Map changes to specific metabolic pathways

  • Protein-protein interaction studies

    • Co-immunoprecipitation with tagged astD

    • Proximity labeling approaches (BioID, APEX)

    • Yeast two-hybrid screening

Metabolomic Approaches:

  • Targeted analysis of arginine and proline pathway metabolites

    • Quantify N-succinyl-L-glutamate and related compounds

    • Track isotope-labeled substrates through the pathway

  • Untargeted metabolomics to identify global metabolic shifts

    • Compare metabolite profiles between wild-type and ΔastD strains

    • Identify unexpected metabolic alterations

Integration Strategy:

  • Correlate changes in protein abundance with metabolite levels

  • Construct metabolic flux models incorporating enzyme kinetics

  • Validate key findings through targeted genetic manipulations

This integrated approach would provide a systems-level understanding of how astD influences L. pneumophila metabolism and potentially its pathogenicity. For example, a study utilizing proteomics to investigate recombinant protein production demonstrated how such approaches can reveal the dynamics of metabolic burden on host cells , and similar techniques could be applied to understand astD's role in bacterial metabolism.

What are the optimal conditions for expressing and purifying catalytically active recombinant astD for structural studies?

Obtaining catalytically active recombinant astD suitable for structural studies requires careful optimization of expression and purification conditions:

Expression Optimization:

ParameterOptions to TestConsiderations
Host strainBL21(DE3), Rosetta, SHuffleCodon usage, disulfide bond formation
Temperature16°C, 25°C, 37°CLower temperatures often increase solubility
InductionIPTG concentration (0.1-1 mM)Lower concentrations may improve folding
MediaLB, TB, auto-inductionNutrient-rich media can improve yields
AdditivesGlycylglycine, ethanol, sucroseMay improve protein folding

Purification Strategy for Structural Studies:

  • Initial capture using affinity chromatography (His-tag)

  • Tag removal using specific proteases (TEV, PreScission)

  • Ion exchange chromatography

  • Size exclusion chromatography in crystallization buffer

Protein Quality Assessment:

  • Thermal shift assay to identify stabilizing buffer conditions

  • Dynamic light scattering to confirm monodispersity

  • Activity assays to verify catalytic function

  • Mass spectrometry to confirm protein integrity

Crystallization Screening:

  • Commercial sparse matrix screens at multiple protein concentrations

  • Addition of substrate analogs or cofactors to stabilize protein conformation

  • Optimization of initial crystallization hits by varying pH, precipitant, and additives

The presence of NAD+ or NADH during purification and crystallization may stabilize the enzyme structure, as is common for many dehydrogenases. Additionally, including substrate analogs could lock the enzyme in a specific conformational state, potentially facilitating crystal formation.

How can computational approaches be used to predict substrate specificity and inhibitor design for Legionella pneumophila astD?

Computational approaches provide valuable tools for investigating astD substrate specificity and designing potential inhibitors:

Homology Modeling:

  • Identify structurally characterized homologs (other aldehyde dehydrogenases)

  • Generate multiple models using different templates

  • Validate models through energy minimization and Ramachandran plot analysis

  • Refine active site architecture based on conserved catalytic residues

Substrate Docking and Molecular Dynamics:

  • Dock N-succinyl-L-glutamate 5-semialdehyde into the predicted active site

  • Perform molecular dynamics simulations to assess stability of enzyme-substrate complex

  • Identify key residues involved in substrate recognition and catalysis

  • Test substrate analogs to predict enzyme promiscuity

Virtual Screening for Inhibitor Discovery:

  • Develop a pharmacophore model based on substrate binding mode

  • Screen virtual compound libraries against the active site

  • Rank compounds based on predicted binding affinity and interactions

  • Filter candidates for drug-like properties (Lipinski's rules)

Enzyme-Inhibitor Complex Optimization:

  • Perform molecular dynamics simulations of top inhibitor candidates

  • Calculate binding free energies using MM/GBSA or FEP methods

  • Optimize lead compounds through structural modifications

  • Predict ADMET properties to guide compound selection

These computational predictions would guide experimental validation through enzyme assays with predicted substrates or inhibitors, potentially leading to the development of specific probes for studying astD function or even therapeutic agents targeting this enzyme in L. pneumophila.

What controls should be included when characterizing recombinant Legionella pneumophila astD activity?

Rigorous experimental design requires appropriate controls to ensure reliable characterization of recombinant astD activity:

Positive Controls:

  • Well-characterized homologous enzyme from another organism

  • Commercial aldehyde dehydrogenase with known activity

  • Purified native astD from L. pneumophila (if available)

Negative Controls:

  • Heat-inactivated recombinant astD

  • Reaction mixture without enzyme

  • Reaction mixture without substrate

  • Catalytically inactive mutant (e.g., active site cysteine mutation)

Specificity Controls:

  • Testing structural analogs of the substrate

  • Testing alternative cofactors (NADP+ instead of NAD+)

  • Activity assays in the presence of known inhibitors of aldehyde dehydrogenases

Validation Controls:

  • Multiple independent protein preparations

  • Enzyme activity at different protein concentrations (linearity assessment)

  • Time-course studies to ensure initial velocity conditions

Including these controls helps identify potential artifacts, confirms enzyme specificity, and ensures that measured activity is genuinely attributable to the recombinant astD. This is particularly important when working with enzymes from pathogens like L. pneumophila, where experimental manipulation may alter native enzyme characteristics.

How can researchers investigate the relationship between astD and other virulence factors in Legionella pneumophila?

Investigating the relationship between astD and known virulence factors requires integrative approaches:

Transcriptional Analysis:

  • qRT-PCR to measure co-expression patterns

  • RNA-seq to identify global transcriptional networks

  • Promoter-reporter fusions to monitor gene expression under different conditions

Genetic Interaction Studies:

  • Construction of double mutants (ΔastD + Δvirulence factor)

  • Complementation studies with different alleles

  • Suppressor screens to identify compensatory mutations

Protein-Protein Interaction Analysis:

  • Bacterial two-hybrid assays

  • Co-immunoprecipitation studies

  • Cross-linking mass spectrometry

  • Fluorescence resonance energy transfer (FRET)

Functional Interdependence Assessment:

  • Measure virulence factor secretion/function in ΔastD background

  • Examine metabolite levels that might influence virulence gene expression

  • Test virulence in infection models using various genetic backgrounds

L. pneumophila has multiple virulence determinants, including the lag-1 gene, which confers resistance to complement-mediated killing , and a glucosyltransferase that modifies host elongation factor 1A . Understanding how astD interacts with these established virulence mechanisms could reveal new insights into the pathogenesis of Legionnaires' disease.

What approaches can resolve contradictory data regarding astD function in different experimental systems?

When faced with contradictory data about astD function, systematic troubleshooting and validation approaches are essential:

Source of Variability Assessment:

  • Strain differences: Compare astD sequences and genomic context across L. pneumophila strains

  • Experimental conditions: Systematically vary temperature, pH, buffer composition

  • Host cell types: Test activity in different cell types (macrophages vs. amoebae)

Methodological Validation:

  • Cross-validate using independent techniques

    • Enzymatic assays using different detection methods

    • Genetic approaches alongside biochemical studies

    • In vitro versus in vivo models

  • Blind testing by independent researchers

  • Standardization of protocols across laboratories

Reconciliation Strategies:

  • Identify conditional factors that explain different outcomes

  • Develop unified models that incorporate context-dependent functions

  • Design critical experiments specifically addressing contradictions

Meta-analysis Approach:

  • Collate all available data on astD function

  • Weight evidence based on methodological rigor

  • Identify patterns explaining apparent contradictions

This systematic approach helps distinguish genuine biological complexity from technical artifacts, leading to a more nuanced understanding of astD function across different experimental contexts.

How might astD be exploited as a potential drug target for treating Legionella infections?

Exploring astD as a potential drug target requires assessment of its essentiality, druggability, and validation through multiple approaches:

Target Validation Strategy:

  • Determine essentiality through:

    • Conditional knockout studies

    • Chemical genetic approaches

    • Transposon mutagenesis screens

  • Evaluate impact on virulence:

    • Infection studies with astD mutants

    • Assessment of fitness cost in relevant infection models

Druggability Assessment:

  • Structural analysis of active site

    • Pocket size and shape

    • Hydrophobic vs. polar characteristics

    • Flexibility of binding site

  • Chemical starting points:

    • Natural product inhibitors of similar enzymes

    • Known aldehyde dehydrogenase inhibitors

    • Fragment-based discovery approaches

Drug Development Pipeline:

  • Primary screening using:

    • Enzymatic assays with recombinant astD

    • Cell-based assays measuring bacterial survival

  • Hit validation and optimization:

    • Structure-activity relationship studies

    • Pharmacokinetic improvement

    • Selectivity enhancement over human homologs

  • Preclinical testing:

    • Efficacy in cell culture and animal models

    • Toxicity assessment

    • Resistance development monitoring

Given that L. pneumophila causes approximately 8,000-18,000 hospitalizations annually in the United States with fatality rates of 5-30% , novel therapeutic approaches targeting metabolic enzymes like astD could provide alternatives to conventional antibiotics, potentially addressing issues of antimicrobial resistance.

What are the emerging technologies that could advance our understanding of astD function in Legionella pneumophila?

Several cutting-edge technologies show promise for deepening our understanding of astD function:

Single-Cell Technologies:

  • Single-cell RNA-seq to capture heterogeneity in astD expression

  • Single-cell proteomics to correlate protein levels with bacterial phenotypes

  • Microfluidic systems to track individual bacterial cells during infection

Advanced Imaging Approaches:

  • Super-resolution microscopy to visualize enzyme localization

  • Correlative light and electron microscopy (CLEM) to link function with ultrastructure

  • Live-cell imaging with fluorescent activity-based probes

CRISPR-Based Technologies:

  • CRISPRi for tunable gene repression

  • CRISPR-Cas13 for RNA targeting and modulation

  • Base editing for precise genetic modifications

Structural Biology Innovations:

  • Cryo-electron microscopy for structure determination without crystallization

  • Hydrogen-deuterium exchange mass spectrometry for dynamics analysis

  • Integrative structural biology combining multiple data sources

Systems Biology Approaches:

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

  • Machine learning for pathway analysis and prediction

  • Genome-scale metabolic modeling to contextualize astD function

These emerging technologies could help resolve current knowledge gaps regarding astD function and potentially reveal unexpected roles in bacterial metabolism and pathogenesis. Integration of these approaches would provide a more holistic understanding of how astD contributes to L. pneumophila biology.

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.