Recombinant Legionella pneumophila subsp. pneumophila FMN-dependent NADH-azoreductase (azoR)

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 unless dry ice shipping is specifically requested and pre-arranged. 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 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 is influenced by 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 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 a specific tag type is required, please inform us, and we will prioritize its development.
Synonyms
azoR; lpg2287FMN-dependent NADH-azoreductase; EC 1.7.1.17; Azo-dye reductase; FMN-dependent NADH-azo compound oxidoreductase
Buffer Before Lyophilization
Tris/PBS-based buffer, 6% Trehalose.
Datasheet
Please contact us to get it.
Expression Region
1-206
Protein Length
full length protein
Purity
>85% (SDS-PAGE)
Species
Legionella pneumophila subsp. pneumophila (strain Philadelphia 1 / ATCC 33152 / DSM 7513)
Target Names
azoR
Target Protein Sequence
MKLLAIDSSI LTNTSVSRQL TRSFVSRWQK IYPETEVVYR DLHAQPINHL SQKILAANSV PSTQISAEIR EEMNLSMQLI SELLSASVLV IGAPMYNFSI PSQLKSWIDR IVIAGKTFKY VDGKVQGLAT GKRAYILSSR GGFYNAEPAL NLDHQERYLT SILNFIGISD ITFIRAEGVN VGEEIRTQSL HQAEAKIQQL LQFQMA
Uniprot No.

Target Background

Function
Quinone reductase providing resistance to thiol-specific stress induced by electrophilic quinones. It also displays azoreductase activity, catalyzing the reductive cleavage of the azo bond in aromatic azo compounds to their corresponding amines.
Database Links

KEGG: lpn:lpg2287

STRING: 272624.lpg2287

Protein Families
Azoreductase type 1 family

Q&A

What is the Legionella pneumophila FMN-dependent NADH-azoreductase (azoR)?

Legionella pneumophila FMN-dependent NADH-azoreductase (azoR) is a flavoenzyme belonging to the azoreductase type 1 family that catalyzes the reductive cleavage of azo bonds in aromatic azo compounds, converting them to corresponding amines. The enzyme specifically from Legionella pneumophila subsp. pneumophila (strain Philadelphia 1 / ATCC 33152 / DSM 7513) has a molecular mass of approximately 23.1 kDa and consists of 206 amino acid residues . This enzyme requires NADH as an electron donor for its catalytic activity and contains flavin mononucleotide (FMN) as a cofactor . Legionella pneumophila is primarily known as the causative agent of Legionnaires' disease, a severe form of pneumonia .

What is the amino acid sequence of L. pneumophila azoR?

The complete amino acid sequence of L. pneumophila azoR consists of 206 amino acids as follows:

MKLLAIDSSILTNTSVSRQLTRSFVSRWQKIYPETEVVYRDLHAQPINHLSQKILAANSVPSTQISAEIREEMNLSMQLISELLSASVLVIGAPMYNFSIPSQLKSWIDRIVIAGKTFKYVDGKVQGLATGKRAYILSSRGGFYNAEPALNLDHQERYLTSILNFIGISDITFIRAEGVNVGEEIRTQSLHQAEAKIQQLLQFQMA

Understanding this primary structure is fundamental for researchers investigating protein-ligand interactions, conducting site-directed mutagenesis, and performing structural analyses through techniques such as X-ray crystallography or computational modeling.

What are the general structural characteristics of azoreductases?

Azoreductases typically adopt a flavodoxin-like fold characteristic of FMN-binding proteins. Based on structural studies of homologous enzymes, we can infer that L. pneumophila azoR likely features:

  • A central β-sheet structure flanked by α-helices in a typical Rossmann fold arrangement

  • A binding pocket that accommodates both FMN and the substrate

  • Specific amino acid residues that facilitate electron transfer from NADH to FMN and subsequently to the azo substrate

  • Active sites that typically include aromatic residues (like tyrosine) and charged residues (like arginine or aspartate) that participate in substrate binding

Though the exact structure of L. pneumophila azoR has not been crystallographically resolved in the provided information, structural homology modeling based on related azoreductases would provide valuable insights for researchers.

How can recombinant L. pneumophila azoR be expressed and purified for research studies?

Expression System Selection:

  • Prokaryotic expression using E. coli BL21(DE3) with pET vectors is commonly employed for recombinant azoreductases

  • Alternative systems include insect cell expression or cell-free systems for proteins that may form inclusion bodies

Optimized Expression Protocol:

  • Transform expression vector containing the L. pneumophila azoR gene into competent E. coli cells

  • Culture in LB medium supplemented with appropriate antibiotics at 37°C until OD600 reaches 0.6-0.8

  • Induce protein expression with IPTG (0.1-1.0 mM), preferably at lower temperatures (16-25°C) to enhance solubility

  • Harvest cells after 4-16 hours by centrifugation

Purification Strategy:

  • Resuspend cell pellet in lysis buffer containing protease inhibitors

  • Lyse cells by sonication or alternative methods

  • Clarify lysate by centrifugation at 15,000 × g for 30 minutes

  • For His-tagged constructs, perform immobilized metal affinity chromatography (IMAC)

  • Further purify by size exclusion chromatography or ion-exchange chromatography

  • Verify purity by SDS-PAGE and protein identity by mass spectrometry or Western blotting

Researchers should monitor protein folding by measuring enzymatic activity throughout the purification process, as proper folding is essential for incorporating the FMN cofactor.

What assays can be used to measure L. pneumophila azoR enzymatic activity?

Spectrophotometric Assays:
The primary method for measuring azoreductase activity involves monitoring the decrease in absorbance as colored azo dyes are reduced to colorless amine products:

  • Standard Assay Components:

    • Reaction buffer (typically phosphate buffer, pH 7.0-7.5)

    • Recombinant azoR enzyme (1-10 μg)

    • FMN (1-10 μM)

    • NADH (100-500 μM)

    • Azo substrate such as Methyl Red (10-100 μM)

  • Measurement Protocol:

    • Prepare reaction mixture excluding NADH

    • Establish baseline in spectrophotometer at wavelength appropriate for the substrate

    • Initiate reaction by adding NADH

    • Monitor continuous decrease in absorbance at 25-37°C

    • Calculate reaction rate using substrate-specific extinction coefficients

  • Data Analysis:

    • Determine initial velocities from the linear portion of absorbance vs. time plots

    • For kinetic studies, vary substrate concentrations and fit data to Michaelis-Menten equation

    • Calculate Km, Vmax, and kcat values to characterize enzyme efficiency

  • Controls and Validations:

    • Include no-enzyme controls to account for non-enzymatic reduction

    • Perform reactions without NADH to confirm cofactor dependency

    • Include known azoreductase inhibitors as positive controls

How can site-directed mutagenesis inform our understanding of L. pneumophila azoR?

Site-directed mutagenesis is a powerful approach for investigating structure-function relationships in azoR. Based on studies of homologous azoreductases, researchers should consider the following experimental design:

Target Selection Strategy:

  • Identify conserved residues across azoreductase family members

  • Focus on residues predicted to be involved in:

    • FMN binding (typically positively charged residues)

    • NADH binding (residues within the Rossmann fold)

    • Substrate specificity (residues lining the active site cavity)

    • Catalytic activity (e.g., tyrosine residues that may participate in electron transfer)

Key Residues to Consider:

  • By analogy with other azoreductases, residues like Tyr-129 and Asp-184 may be crucial for substrate binding and catalysis

  • Positively charged residues (Arg, Lys) that may interact with the negatively charged phosphate groups of FMN and NADH

  • Aromatic residues that may form π-π interactions with azo substrates

Experimental Approach:

  • Generate single point mutations using PCR-based techniques

  • Express and purify mutant proteins following the same protocol as wild-type

  • Compare biochemical properties:

    • Substrate affinity (Km)

    • Catalytic efficiency (kcat/Km)

    • Thermal stability (Tm)

    • Substrate specificity profiles

Data Interpretation:

  • Decreased activity with FMN binding site mutations may indicate impaired cofactor association

  • Changes in substrate specificity when mutating active site residues can reveal the molecular basis for recognition

  • Altered NADH affinity in Rossmann fold mutations confirms the electron donor binding site

This systematic approach allows researchers to construct a comprehensive model of the enzyme's functional architecture.

What is known about the binding mechanisms of substrates and cofactors in azoreductase enzymes?

While specific information for L. pneumophila azoR is limited in the provided sources, general principles of azoreductase binding mechanisms can be inferred from studies of homologous enzymes:

FMN Binding:

  • The isoalloxazine ring of FMN typically sits in a pocket near the active site

  • The phosphate group often interacts with positively charged residues

  • The ribityl chain usually forms hydrogen bonds with polar residues

NADH Binding:

  • NADH typically binds in a Rossmann fold configuration

  • The adenine moiety often interacts with hydrophobic residues

  • The nicotinamide portion positions near the FMN for efficient electron transfer

  • In homologous enzymes, the adenine ribose moiety of NADH is surrounded by specific structural elements (e.g., loop l2 on chain B and α3 on chain A)

Substrate Binding:

  • Azo compounds like Methyl Red bind in proximity to the N5 position of FMN (within 5Å) to facilitate electron transfer

  • Substrate binding may involve both aromatic stacking interactions and hydrogen bonding

  • Based on homologous enzymes, approximately 12 amino acid residues may participate in binding azo substrates

Electron Transfer Mechanism:

  • NADH transfers electrons to FMN, converting it to FMNH2

  • FMNH2 subsequently transfers electrons to the azo substrate

  • This results in the reduction of the N=N bond to produce two amine products

Research using techniques such as X-ray crystallography, molecular docking, and isothermal titration calorimetry would further elucidate these binding mechanisms for L. pneumophila azoR specifically.

What potential roles might azoR play in L. pneumophila pathogenesis and environmental survival?

The physiological role of azoR in L. pneumophila is not explicitly detailed in the provided sources, but several hypotheses can be proposed based on current understanding of azoreductases and L. pneumophila biology:

Potential Functions in Host-Pathogen Interactions:

  • Detoxification: The enzyme may help neutralize host-derived antimicrobial compounds containing azo bonds, contributing to bacterial survival within host cells

  • Adaptation to Oxidative Stress: Azoreductases may participate in managing oxidative stress encountered during infection, particularly within alveolar macrophages

  • Metabolic Versatility: The ability to reduce azo compounds may expand the metabolic capabilities of L. pneumophila when parasitizing amoebae or during human infection

Environmental Persistence Mechanisms:

  • Biofilm Formation: Azoreductases have been implicated in biofilm formation in other bacteria, potentially contributing to L. pneumophila persistence in water systems

  • Amoeba Interactions: Since L. pneumophila naturally parasitizes free-living amoeba , azoR may play a role in this ecological relationship

  • Biodegradation Capacity: The ability to metabolize diverse compounds containing azo bonds might provide adaptive advantages in various aquatic environments

Research Approaches to Test These Hypotheses:

  • Generate azoR knockout mutants and compare virulence in cell culture and animal models

  • Perform comparative proteomics to identify changes in azoR expression under different environmental conditions

  • Investigate potential azo-containing compounds in host systems that might serve as natural substrates

  • Examine azoR expression during different stages of the L. pneumophila life cycle

Understanding the physiological role of azoR could potentially reveal new targets for therapeutic intervention against Legionnaires' disease, which has a case fatality rate ranging from 5% to 30% .

How does the genetic context of azoR in the L. pneumophila genome inform our understanding of its function?

Understanding the genomic context of the azoR gene can provide valuable insights into its regulation and potential functional associations:

Genomic Organization Considerations:

  • Operon Structure: Determine whether azoR is part of an operon with functionally related genes

  • Regulatory Elements: Identify promoters, transcription factor binding sites, and potential regulatory RNAs that may control azoR expression

  • Horizontal Gene Transfer: Assess whether there is evidence of azoR acquisition through horizontal gene transfer, similar to the lag-1 gene distribution observed across L. pneumophila lineages

Comparative Genomic Analysis:

  • Compare the genomic location and neighborhood of azoR across different L. pneumophila strains

  • Identify co-occurring genes that may indicate functional relationships

  • Examine azoR presence/absence patterns in clinical versus environmental isolates to assess potential association with virulence

Experimental Approaches:

  • Perform transcriptomics to identify co-regulated genes under different conditions

  • Use techniques like ChIP-seq to identify transcription factors regulating azoR expression

  • Conduct reporter gene assays to characterize promoter activity under various physiological states

This genomic context analysis would complement biochemical studies and help position azoR within the broader framework of L. pneumophila biology and pathogenesis.

How does L. pneumophila azoR differ from azoreductases in other bacterial species?

Understanding the unique features of L. pneumophila azoR compared to homologous enzymes provides insights into its specialized functions:

Sequence and Structural Comparison:

FeatureL. pneumophila azoRE. coli AzoROther Bacterial Azoreductases
Length206 amino acids ~200 amino acidsVariable (150-250 aa)
Cofactor preferenceFMN-dependent, requires NADH FMN-dependentSome use NADPH or both
Key residuesNot specifically identified in sourcesArg-59, Tyr-129, Asp-184 Varies by species
Substrate specificityAzo bond cleavage Various azo dyes including Methyl Red Species-dependent specificity
Oligomeric stateNot specified in sourcesTypically dimericMonomeric to tetrameric

Functional Distinctions:

  • Substrate range may be adapted to the specific ecological niche of L. pneumophila

  • Electron donor preference (NADH vs. NADPH) reflects the cellular redox environment of L. pneumophila

  • Catalytic efficiency may be optimized for the temperature range encountered in water systems where L. pneumophila thrives

Evolutionary Considerations:

  • Azoreductases likely evolved independently multiple times across bacterial lineages

  • The specific properties of L. pneumophila azoR may reflect adaptation to its dual lifestyle as an environmental organism and human pathogen

  • Comparative phylogenetic analysis would reveal whether azoR acquisition patterns mirror those of virulence factors like lag-1

Understanding these differences would guide researchers in developing specific inhibitors or diagnostic tools targeting L. pneumophila azoR.

What are the common difficulties in working with recombinant azoreductases and how can they be overcome?

Researchers working with recombinant azoreductases often encounter several technical challenges that require specific solutions:

Challenge 1: Maintaining Cofactor Association

  • Problem: Loss of FMN during purification resulting in reduced activity

  • Solutions:

    • Supplement buffers with 5-10 μM FMN throughout purification

    • Perform reconstitution by incubating purified protein with excess FMN followed by dialysis

    • Measure FMN:protein ratio spectrophotometrically to ensure full occupancy

Challenge 2: Solubility and Stability Issues

  • Problem: Formation of inclusion bodies or protein aggregation

  • Solutions:

    • Express at lower temperatures (16-20°C) with reduced inducer concentration

    • Add solubility-enhancing tags (MBP, SUMO) that can be later removed

    • Include stabilizing agents such as glycerol (10-20%) in storage buffers

    • Optimize buffer conditions through thermal shift assays to identify stabilizing additives

Challenge 3: Accurate Activity Measurements

  • Problem: Background reduction of azo dyes by reducing agents

  • Solutions:

    • Include appropriate controls without enzyme

    • Use anaerobic conditions to prevent oxygen interference

    • Consider alternative assay methods such as HPLC to detect product formation rather than substrate disappearance

Challenge 4: Substrate Solubility Limitations

  • Problem: Many azo compounds have limited water solubility

  • Solutions:

    • Prepare stock solutions in DMSO or ethanol (keeping final concentration below 5%)

    • Use cyclodextrins as solubilizing agents

    • Develop microemulsion systems for highly hydrophobic substrates

    • Ensure that control reactions contain identical solvent concentrations

Challenge 5: Kinetic Characterization Complexities

  • Problem: Multi-substrate enzyme mechanisms complicate kinetic analysis

  • Solutions:

    • Employ steady-state approximations by keeping one substrate in excess

    • Utilize global fitting approaches for simultaneous analysis of multiple datasets

    • Consider stopped-flow spectroscopy for analyzing rapid kinetic phases

Addressing these technical challenges systematically will enhance the quality and reproducibility of research on L. pneumophila azoR.

How can advanced analytical techniques enhance our understanding of L. pneumophila azoR reaction mechanisms?

Modern analytical techniques offer powerful approaches to elucidate the detailed reaction mechanisms of L. pneumophila azoR:

Transient Kinetics Analysis:

  • Stopped-flow Spectroscopy: Monitor rapid changes in absorbance during catalysis on millisecond timescales

  • Rapid-Quench Flow: Capture reaction intermediates by chemical quenching at defined time points

  • Applications: Identify rate-limiting steps and detect transient intermediates in the electron transfer process

Structural Analysis Techniques:

  • X-ray Crystallography: Determine high-resolution structures of azoR in complex with substrates or inhibitors

  • Cryo-EM: Visualize conformational changes during catalysis

  • HDX-MS: Identify regions undergoing conformational changes upon substrate or cofactor binding

  • SAXS: Analyze solution structure and conformational flexibility

Spectroscopic Methods:

  • EPR Spectroscopy: Monitor the redox state of the flavin during catalysis

  • Resonance Raman Spectroscopy: Probe the electronic structure of the flavin and its interaction with substrates

  • Fluorescence Spectroscopy: Measure binding affinities and conformational changes

  • Applications: Characterize the electronic and structural changes occurring during catalysis

Computational Approaches:

  • QM/MM Simulations: Model electron transfer pathways at the quantum level

  • Molecular Dynamics: Simulate protein dynamics during substrate binding and catalysis

  • Virtual Screening: Identify potential inhibitors or alternative substrates

  • Applications: Predict the impact of mutations and design rational protein engineering strategies

Mass Spectrometry-Based Techniques:

  • LC-MS/MS: Identify reaction products and unexpected side reactions

  • Protein Footprinting: Map solvent accessibility changes during catalysis

  • Cross-linking MS: Identify residues in proximity during different catalytic states

  • Applications: Verify proposed reaction mechanisms and identify novel reaction products

Integration of these techniques would provide a comprehensive understanding of the L. pneumophila azoR catalytic mechanism, enabling rational enzyme engineering and inhibitor design.

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.