Recombinant Geobacter sulfurreducens Tetraacyldisaccharide 4'-kinase (lpxK)

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 in order notes for customized fulfillment.
Lead Time
Delivery times vary depending on the purchase method and location. Consult your local distributor for precise delivery estimates.
Note: All proteins are shipped with standard blue ice packs. Dry ice shipping requires prior arrangement and incurs additional charges.
Notes
Avoid repeated freeze-thaw cycles. Store working aliquots at 4°C for up to one week.
Reconstitution
Centrifuge the vial briefly before opening to consolidate the contents. Reconstitute the protein in sterile, deionized water to a concentration of 0.1-1.0 mg/mL. For long-term storage, we recommend adding 5-50% glycerol (final concentration) and aliquoting at -20°C/-80°C. Our standard glycerol concentration is 50% 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 forms 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
Tag type is determined during manufacturing.
The tag type is determined during production. Specify your preferred tag type for prioritized development.
Synonyms
lpxK; GSU2258; Tetraacyldisaccharide 4'-kinase; EC 2.7.1.130; Lipid A 4'-kinase
Buffer Before Lyophilization
Tris/PBS-based buffer, 6% Trehalose.
Datasheet
Please contact us to get it.
Expression Region
1-353
Protein Length
full length protein
Purity
>85% (SDS-PAGE)
Species
Geobacter sulfurreducens (strain ATCC 51573 / DSM 12127 / PCA)
Target Names
lpxK
Target Protein Sequence
MSCTGFWRKL ASGEECGAAA RLLGAVLAIP AALYGLVVRL RALAYARGLS TVRHLDRPVI SVGNLTVGGT GKTPMVAYLA RRLMARGKRV AVISRGYGGS LEGETRIVSD GRTIVLSAAE AGDEPVHLAT SVPGLMTVIG TDRYTAGLLA LEQLDPDVFI LDDGYQHLRL HRDLNILLMD CNRPLGNGRT LPAGLLREPQ TAVRRADLVV YTRCTGGKAP AVHGMIPSCR AGHALTGAAL LPDGEVQPLA ALRGLRGVAC AGIAEPEGFF DALRREGLDI VAAIPFADHA SYGEREVSTL REAAAGADYL ITTGKDGVKL SAHLARLLPV YATVLEMRPL DPAPLEAALD KVL
Uniprot No.

Target Background

Function
This protein catalyzes the transfer of the gamma-phosphate from ATP to the 4'-position of a tetraacyldisaccharide 1-phosphate intermediate (DS-1-P), forming tetraacyldisaccharide 1,4'-bis-phosphate (lipid IVA).
Database Links

KEGG: gsu:GSU2258

STRING: 243231.GSU2258

Protein Families
LpxK family

Q&A

What is Tetraacyldisaccharide 4'-kinase (lpxK) and what role does it play in Geobacter sulfurreducens?

Tetraacyldisaccharide 4'-kinase (lpxK) is a critical enzyme that catalyzes the sixth step in lipid A biosynthesis in Gram-negative bacteria including Geobacter sulfurreducens. LpxK belongs to the diverse P-loop-containing nucleoside triphosphate hydrolase superfamily and is the only known P-loop kinase that acts upon a lipid substrate at the membrane interface . The enzyme specifically phosphorylates the 4' position of the tetraacyldisaccharide precursor, which is essential for the subsequent steps in lipopolysaccharide (LPS) biosynthesis. This phosphorylation is crucial for maintaining the structural integrity of the bacterial outer membrane and directly influences cell surface properties that enable G. sulfurreducens to interact with extracellular electron acceptors .

Why is understanding lipopolysaccharide (LPS) structure important in Geobacter sulfurreducens research?

The lipopolysaccharide structure in G. sulfurreducens is particularly significant because this bacterium produces a rough LPS (lacking O-antigen), which plays multiple critical roles in its environmental adaptations and applications. The rough LPS facilitates surface interactions with minerals, which is essential for the bacterium's ability to reduce metals and participate in biogeochemical cycling . Additionally, this specialized LPS structure influences cell-cell aggregation for biofilm formation, functions as a permeability barrier against toxic metal cations, and optimizes electron transfer between outer membrane cytochromes and external electron acceptors . These properties make understanding LPS structure fundamental for applications in bioremediation of metal-contaminated environments and bioelectrochemical systems for energy generation .

How does Geobacter sulfurreducens respire metals and why is this significant?

Geobacter sulfurreducens has the remarkable ability to respire metals by transferring electrons generated from internal metabolism to extracellular electron acceptors beyond the cell membranes. This process involves specialized electron transfer pathways including inner membrane cytochromes such as ImcH and CbcL . The bacterium can reduce a variety of electron acceptors including Fe(III) oxides and electrodes poised at different potentials, with different electron transfer proteins being utilized depending on the redox potential of the acceptor . This respiratory versatility is significant for several reasons: it enables G. sulfurreducens to thrive in anaerobic, metal-rich environments; it makes these bacteria valuable for bioremediation of metal contaminants like uranium; and it allows them to generate electricity in microbial fuel cells by transferring electrons directly to electrode surfaces .

What expression systems are optimal for recombinant production of G. sulfurreducens lpxK?

For optimal recombinant production of G. sulfurreducens lpxK, researchers should consider the following methodological approaches:

The expression construct should include:

  • An N-terminal His₆-tag for purification (C-terminal tags may interfere with membrane association)

  • A TEV protease cleavage site for tag removal

  • A low-copy number vector to reduce potential toxicity

Expression should be conducted at lower temperatures (16-20°C) after induction to promote proper folding of this membrane-associated enzyme . Given lpxK's association with the membrane, inclusion of mild detergents (0.05% DDM or 0.1% Triton X-100) in the lysis buffer is essential for efficient extraction and maintaining enzymatic activity throughout purification .

What analytical methods are most effective for assessing lpxK enzymatic activity?

Several complementary analytical methods can be employed to effectively assess lpxK enzymatic activity:

  • Radiometric phosphorylation assay:

    • Substrate: Tetraacyldisaccharide 1-phosphate

    • Co-substrate: [γ-³²P]ATP (specific activity ~3000 Ci/mmol)

    • Detection: Thin-layer chromatography followed by autoradiography

    • Sensitivity: Can detect pmol quantities of phosphorylated product

    • Quantification: Phosphorimager analysis with standard curves

  • Coupled spectrophotometric assay:

    • Reaction coupling: ATP → ADP conversion linked to NADH oxidation via pyruvate kinase and lactate dehydrogenase

    • Monitoring: Decrease in absorbance at 340 nm

    • Advantages: Real-time kinetic measurements, no radioactivity

    • Limitations: Potential interference from other ATPases

  • Mass spectrometry-based assay:

    • Method: LC-MS/MS analysis of lipid extracts

    • Measurement: Relative abundance of non-phosphorylated and phosphorylated lipid A precursors

    • Advantage: Provides structural confirmation of products and precise molecular identification

    • Applications: Suitable for complex samples and in vivo activity assessment

These methods should be calibrated using purified enzyme with known specific activity and standardized substrate preparations to ensure reproducibility across different research groups .

How can researchers effectively crystallize G. sulfurreducens lpxK for structural studies?

Based on structural studies of related lpxK enzymes, researchers can effectively crystallize G. sulfurreducens lpxK using the following optimized approach:

  • Protein preparation:

    • Purify to >95% homogeneity (assessed by SDS-PAGE)

    • Concentrate to 10-15 mg/ml in a buffer containing:

      • 20 mM HEPES pH 7.5

      • 150 mM NaCl

      • 5 mM MgCl₂

      • 0.05% n-dodecyl-β-D-maltoside (DDM)

    • Remove aggregates by centrifugation (100,000 × g, 30 min)

  • Crystallization screening:

    • Primary method: Sitting-drop vapor diffusion

    • Temperature: 18°C (optimal for slow crystal growth)

    • Drop composition: 1 μl protein + 1 μl reservoir solution

    • Initial screens: Sparse matrix commercially available screens focused on membrane proteins

  • Optimization conditions for diffraction-quality crystals:

    • Precipitant: 15-25% PEG 3350

    • Buffer: 100 mM MES or HEPES, pH 6.5-7.5

    • Salt: 100-200 mM sodium acetate or ammonium sulfate

    • Additives: 5-10% glycerol, 1-5 mM ADP or ATP analog, 5 mM MgCl₂

  • Co-crystallization with substrates:

    • For nucleotide-bound structures: Pre-incubate with 2-5 mM ADP/ATP and 5 mM MgCl₂

    • For substrate complex: Incorporate lipid substrate analogs solubilized in suitable detergent

  • Crystal harvesting and cryoprotection:

    • Cryoprotectant: Mother liquor supplemented with 20-25% glycerol or ethylene glycol

    • Flash-cooling: Directly in liquid nitrogen stream

These approaches have proven successful for crystallizing membrane-associated enzymes of the P-loop kinase family and can be adapted for G. sulfurreducens lpxK structural studies .

What are the key structural features of lpxK that enable its function in lipid A biosynthesis?

The crystal structure of lpxK reveals key structural features that are essential for its function in lipid A biosynthesis:

  • Domain organization:

    • N-terminal domain: Contains the catalytic P-loop motif responsible for ATP binding and phosphoryl transfer

    • C-terminal domain: A smaller substructure unique to lpxK that helps bind nucleotide substrate and Mg²⁺ cation

    • Two-stranded β-sheet linker: Connects the domains and enables a 25° hinge motion necessary for catalysis

  • Membrane association elements:

    • N-terminal amphipathic helix (residues 6-26): Contains hydrophobic residues (L2, L6, F9, L12, I16, F19, L23, F28) that anchor the enzyme to the inner membrane

    • The outward face of this helix shows significant hydrophobicity, facilitating membrane insertion

  • Catalytic machinery:

    • P-loop motif: Binds the phosphate groups of ATP

    • Conserved aspartate residues (D138 and D139): Critical for catalytic activity, likely involved in positioning the phosphoryl acceptor or direct catalysis

    • Magnesium coordination site: Essential for positioning ATP for phosphoryl transfer

  • Substrate binding regions:

    • Hydrophobic pocket: Accommodates the lipid chains of the tetraacyldisaccharide substrate

    • Positively charged region: Interacts with the phosphate group of the substrate

    • Nucleotide binding pocket: Specifically positions the ATP molecule for optimal reaction geometry

These structural elements work in concert to position the lipid substrate and ATP at the inner membrane interface, facilitating the phosphoryl transfer reaction that is critical for lipid A biosynthesis and subsequent LPS assembly .

How does the enzymatic mechanism of lpxK contribute to bacterial membrane development?

The enzymatic mechanism of lpxK plays a crucial role in bacterial membrane development through a carefully coordinated phosphoryl transfer reaction:

  • Reaction chemistry:

    • lpxK catalyzes the transfer of the γ-phosphate from ATP to the 4'-hydroxyl group of the tetraacyldisaccharide 1-phosphate substrate

    • This produces tetraacyldisaccharide 1,4'-bisphosphate, a key precursor in lipid A biosynthesis

  • Membrane interface catalysis:

    • lpxK functions at the cytoplasmic face of the inner membrane, where it can access both the membrane-embedded lipid substrate and cytoplasmic ATP

    • The enzyme's amphipathic helix anchors it to the membrane in the proper orientation for substrate access

  • Role in LPS assembly pathway:

    • The 4'-phosphorylation catalyzed by lpxK is essential for subsequent enzymatic steps in the LPS biosynthetic pathway

    • This modification creates the proper molecular recognition element for later enzymes

    • The absence of lpxK activity would interrupt LPS biosynthesis, compromising membrane integrity

  • Impact on membrane properties:

    • The proper phosphorylation pattern of lipid A directly influences:

      • The packing of LPS molecules in the outer membrane

      • Surface charge distribution of the bacterial cell

      • Permeability barriers against toxic compounds

      • Cell-cell and cell-surface interactions necessary for biofilm formation

  • Electron transfer optimization:

    • In G. sulfurreducens, the rough LPS structure dependent on lpxK activity reduces the distance between outer membrane electron carriers (like c-type cytochromes) and extracellular electron acceptors

    • This proximity is critical for efficient respiratory electron transfer to external metals or electrodes

Through these mechanisms, lpxK activity directly shapes the outer membrane architecture that enables G. sulfurreducens to interact with its environment and perform its unique electron transfer functions .

What is the relationship between lpxK activity and the adaptation of G. sulfurreducens to metal-rich environments?

The activity of lpxK is integrally connected to G. sulfurreducens' remarkable adaptation to metal-rich environments through several interconnected mechanisms:

  • LPS tailoring for metal interactions:

    • lpxK catalyzes a critical step in producing the rough LPS structure of G. sulfurreducens

    • This specialized LPS lacks the O-antigen typically found in many Gram-negative bacteria, which is an adaptive feature for metal-reducing bacteria

    • G. sulfurreducens modulates its LPS structure depending on electron acceptor availability, producing only the shortest LPS variant (lacking methyl-quinovosamine) when growing with Fe(III) oxides

  • Surface chemistry optimization:

    • The rough LPS structure increases the hydrophilicity of the cell surface, facilitating electrostatic interactions with positively charged metal particles

    • This enhanced surface chemistry promotes attachment to Fe(III) oxides and other mineral surfaces, which is dominated by electrostatic rather than hydrophobic interactions

  • Permeability barrier function:

    • The LPS layer formed through the lpxK-dependent pathway functions as a critical permeability barrier against toxic metal cations

    • This barrier prevents intracellular accumulation of potentially toxic metals while still allowing their extracellular reduction

    • Mutants with defective LPS core oligosaccharides show increased penetration and intracellular mineralization of toxic metals like uranyl cation

  • Electron transfer efficiency:

    • The shorter LPS structure, dependent on proper lpxK function, reduces the distance between outer membrane electron carriers and extracellular metal acceptors

    • This proximity is crucial for efficient discharge of respiratory electrons to metals or electrodes

    • Experimental evidence shows that G. sulfurreducens adapts its LPS structure specifically when utilizing Fe(III) oxides as electron acceptors

Through these mechanisms, lpxK activity contributes significantly to G. sulfurreducens' ability to thrive in metal-rich environments, making it valuable for applications in bioremediation of metal contaminants and bioelectrochemical systems .

How can electrochemical techniques be used to study the impact of lpxK modifications on electron transfer in G. sulfurreducens?

Advanced electrochemical techniques provide powerful tools for investigating how lpxK modifications affect electron transfer capabilities in G. sulfurreducens:

  • Cyclic voltammetry (CV) analysis:

    • Experimental setup: Three-electrode configuration with working electrode (graphite or gold), reference electrode (Ag/AgCl), and counter electrode (platinum)

    • Non-turnover conditions: Perform CV in the absence of electron donor to identify redox-active components

      • Wild-type G. sulfurreducens typically shows distinct low-potential redox peaks associated with outer membrane cytochromes

      • lpxK mutants may show altered peak positions, intensities, or complete absence of certain peaks

    • Catalytic conditions: Perform CV in the presence of acetate as electron donor

      • lpxK modifications may cause shifts in onset potential for catalytic current

      • Similar to ΔcbcL mutants, which showed a 50 mV positive shift in driving force required for electron transfer

  • Chronoamperometry for biofilm growth assessment:

    • Method: Poised-potential experiments at -0.1 V vs. SHE (standard hydrogen electrode)

    • Measurements:

      • Current production over time as biofilm develops

      • Maximum current density achieved at steady state

      • Response to electron donor addition/depletion

    • Expected outcomes for lpxK mutants:

      • Delayed attachment phase

      • Reduced maximum current density

      • Altered biofilm structure and conductivity

  • Electrochemical impedance spectroscopy (EIS):

    • Application: Characterize charge transfer resistance at the bacteria-electrode interface

    • Parameters: Apply small-amplitude sinusoidal potential perturbation over range of frequencies

    • Analysis: Fit data to equivalent circuit models to extract:

      • Solution resistance

      • Charge transfer resistance

      • Double-layer capacitance

      • Biofilm conductivity

  • Scanning electrochemical microscopy (SECM):

    • Method: Use microelectrode probe to scan across bacterial biofilm

    • Measurements: Map local electron transfer rates with spatial resolution

    • Advantage: Directly correlates electron transfer activity with biofilm structure

    • Application: Compare wild-type and lpxK-modified strains to identify localized defects in electron transfer capability

These electrochemical approaches provide mechanistic insights into how lpxK-dependent LPS modifications alter the thermodynamics and kinetics of extracellular electron transfer, which is fundamental to G. sulfurreducens' metal reduction capabilities .

What experimental designs can elucidate the relationship between lpxK function and biofilm formation in microbial fuel cells?

A comprehensive experimental design to investigate the relationship between lpxK function and biofilm formation in microbial fuel cells (MFCs) would include:

  • Genetic manipulation strategies:

    • Controlled expression systems:

      • Construct strains with inducible lpxK expression using tetracycline-responsive promoters

      • Create titrable expression levels to correlate lpxK activity with biofilm properties

    • Site-directed mutagenesis:

      • Generate catalytically impaired lpxK variants (e.g., D138A and D139A mutations) that show reduced activity

      • Create chimeric lpxK proteins with domains from other bacteria to alter LPS structure

  • Multi-parameter biofilm characterization:

    • Real-time monitoring:

      • Current production as measure of biofilm development and activity

      • Confocal laser scanning microscopy with live/dead staining for biofilm structure

      • Fluorescent reporter strains to track gene expression during biofilm formation

    • Developmental stage analysis:

      • Initial attachment (0-12 hours): Crystal violet staining and microscopy

      • Monolayer formation (12-24 hours): Protein quantification and surface coverage

      • Microcolony development (24-48 hours): 3D structure analysis

      • Mature biofilm (48-72+ hours): Thickness, biomass, and activity measurements

  • Comparative experimental matrix:

    lpxK VariantElectrode MaterialMeasurement Parameters
    Wild-typeGraphiteCurrent density, biofilm thickness, protein content
    Inducible lpxKGraphiteExpression level vs. current correlation
    D138A mutantGraphiteAttachment efficiency, biofilm morphology
    D139A mutantGraphiteElectron transfer rates, cytochrome exposure
    Wild-typeGoldSurface chemistry effects vs. graphite
    lpxK variantsGoldMaterial-dependent effects of lpxK modification
  • Analytical techniques:

    • Biofilm composition analysis:

      • Extraction and characterization of extracellular polymeric substances (EPS)

      • Quantification of c-type cytochromes in biofilm vs. planktonic cells

      • LPS profiling using PAGE and silver staining

    • Advanced imaging:

      • Electron microscopy to visualize cell-electrode and cell-cell interfaces

      • Correlative microscopy combining structural and functional imaging

  • Electrochemical performance metrics:

    • Power density curves at different biofilm developmental stages

    • Internal resistance measurements to assess electron transfer limitations

    • Long-term stability under variable load conditions

This experimental design would systematically link lpxK function to specific aspects of biofilm development and electrochemical performance in MFCs, providing insights for optimizing bioelectrochemical systems .

How can research on lpxK in G. sulfurreducens contribute to advancements in bioremediation of metal-contaminated environments?

Research on lpxK in G. sulfurreducens offers significant potential for advancing bioremediation technologies for metal-contaminated environments through several innovative approaches:

These research directions could significantly improve the effectiveness, efficiency, and applicability of G. sulfurreducens-based bioremediation technologies for diverse metal-contaminated environments .

What are the major challenges in expressing and purifying active recombinant G. sulfurreducens lpxK, and how can they be addressed?

Expressing and purifying active recombinant G. sulfurreducens lpxK presents several significant challenges due to its membrane association and catalytic properties. The following table outlines these challenges and effective solutions:

ChallengeUnderlying CauseSolution StrategiesEffectiveness Metrics
Low expression levelsMembrane protein toxicity to host cellsUse C41/C43 E. coli strains designed for membrane proteins; Lower induction temperature (16-20°C); Use tightly controlled promoters≥5 mg purified protein per liter of culture
Protein aggregationHydrophobic surfaces exposed during expressionCo-express with chaperones (GroEL/ES); Add mild detergents during lysis (0.05-0.1% DDM or Triton X-100); Include 10% glycerol in all buffers<10% aggregation assessed by size exclusion chromatography
Loss of activity during purificationDetergent-mediated disruption of structureScreen detergent panel (DDM, LDAO, CHAPS); Incorporate lipid additives (E. coli polar lipids); Minimize time between lysis and assay>70% retention of specific activity
Inconsistent enzymatic activityVariability in lipid substrate preparationStandardize lipid substrate preparation; Use synthetic tetraacyldisaccharide analogues; Incorporate lipid substrate into nanodiscs<15% variation between batches
Poor solubility of lipid substrateHighly hydrophobic nature of substrateUse appropriate detergent:substrate ratios; Sonicate lipid stocks before use; Prepare fresh substrate solutions for each assayComplete solubilization verified by dynamic light scattering

Additional methodological considerations:

  • Protein stability enhancement:

    • Include metal chelators (EDTA) to prevent metal-catalyzed oxidation

    • Add reducing agents (DTT or TCEP) to maintain cysteine residues

    • Aliquot and flash-freeze purified enzyme to prevent freeze-thaw damage

  • Membrane mimetic systems:

    • Reconstitute purified lpxK into nanodiscs or liposomes composed of E. coli lipids

    • This approach maintains the native-like membrane environment crucial for activity

    • Increases stability during storage and provides a more relevant system for functional studies

  • Activity verification approaches:

    • Develop a multi-method validation protocol combining:

      • Radiometric assays for high sensitivity

      • Coupled spectrophotometric assays for kinetic studies

      • Mass spectrometry for product confirmation

    • Compare activity metrics across methods to ensure consistent results

These comprehensive strategies address the significant challenges in obtaining functional recombinant G. sulfurreducens lpxK and provide a robust framework for consistent experimental outcomes.

How can researchers accurately analyze and interpret contradictory data in studies of lpxK's role in electron transfer?

When faced with contradictory data regarding lpxK's role in electron transfer, researchers should employ a systematic analytical framework to reconcile discrepancies:

  • Source identification of contradictions:

    • Experimental design variations:

      • Growth conditions (medium composition, temperature, electron acceptor)

      • Genetic background differences between strains

      • Electrode materials and electrochemical setup variations

    • Methodological differences:

      • Protein expression and purification protocols

      • Activity assay conditions and detection methods

      • Electrochemical measurement parameters

  • Standardization approach:

    • Establish baseline conditions:

      • Define standard growth protocols (NBAFYE medium at 30°C for consistency with structural studies)

      • Standardize electron acceptor conditions (fumarate vs. Fe(III) oxides)

      • Create reference standards for electrochemical measurements

    • Cross-laboratory validation:

      • Exchange strains between laboratories

      • Perform identical experiments in different settings

      • Analyze data using standardized processing methods

  • Multi-level analysis framework:

    • Gene expression level:

      • Quantify lpxK transcript abundance under different conditions

      • Correlate expression with observed phenotypes

    • Protein activity level:

      • Measure enzyme kinetics under standardized conditions

      • Determine substrate specificity and cofactor requirements

    • LPS structure level:

      • Analyze LPS profiles using consistent extraction and visualization methods

      • Correlate structural changes with growth conditions

    • Electron transfer level:

      • Use multiple electrochemical techniques (CV, chronoamperometry, EIS)

      • Correlate electrochemical signatures with LPS characteristics

  • Statistical approaches for data integration:

    • Meta-analysis techniques:

      • Combine data from multiple studies using effect size calculations

      • Weight results based on methodological robustness

      • Identify consistent trends across diverse experimental conditions

    • Multivariate analysis:

      • Principal component analysis to identify key variables driving differences

      • Cluster analysis to group consistent results

      • Bayesian networks to establish causal relationships

  • Resolution strategies for specific contradictions:

    • Direct vs. indirect effects:

      • Time-course experiments to distinguish immediate vs. delayed responses

      • Complementation studies with wild-type and mutant lpxK variants

      • Chemical rescue experiments to bypass specific enzymatic steps

    • Strain-specific vs. general mechanisms:

      • Compare multiple G. sulfurreducens strains

      • Examine lpxK function in related Geobacter species

      • Create chimeric constructs to isolate strain-specific elements

By systematically applying these approaches, researchers can transform contradictory data into a more nuanced understanding of lpxK's role in electron transfer, distinguishing between context-dependent effects and fundamental mechanisms .

What quality control measures are essential when studying the effects of lpxK modifications on LPS structure and function?

Rigorous quality control measures are essential for obtaining reliable and reproducible results when studying lpxK modifications and their effects on LPS structure and function:

  • Genetic modification verification:

    • DNA sequence confirmation:

      • Complete sequencing of the modified lpxK gene and surrounding regions

      • Verification of promoter integrity for expression studies

      • Detection of potential second-site mutations using whole-genome sequencing

    • Expression level validation:

      • RT-qPCR to quantify transcript levels

      • Western blotting with specific antibodies to measure protein abundance

      • Activity assays to confirm functional consequences of modifications

  • LPS extraction and analysis quality controls:

    • Extraction method consistency:

      • Standardize cell growth phase (mid-log vs. stationary)

      • Document detailed extraction protocols with precise timing

      • Include wild-type controls in every extraction batch

    • Analysis technique validation:

      • Include molecular weight markers for PAGE analysis

      • Use multiple staining methods (silver stain plus specific carbohydrate stains)

      • Perform technical replicates across independent biological samples

    • Reference standards:

      • Maintain laboratory reference LPS preparations for comparison

      • Include purified LPS with known modifications as controls

      • Develop quantitative metrics for band intensity comparisons

  • Functional assay standardization:

    • Surface property measurements:

      • Conduct hydrophobicity assays using multiple hydrocarbons (xylene, toluene, hexane)

      • Measure zeta potential across a range of pH values

      • Compare results with established controls (wild-type, ΔrfaC, ΔrfaQ)

    • Metal interaction assays:

      • Standardize metal oxide preparations (particle size, crystallinity)

      • Control metal:cell ratios precisely

      • Include time-course measurements to capture kinetic differences

    • Biofilm formation assessments:

      • Maintain consistent surface materials

      • Control environmental parameters (temperature, medium flow rates)

      • Use multiple quantification methods (crystal violet, confocal microscopy, protein quantification)

  • Electron transfer measurement controls:

    • Electrode preparation:

      • Standardize electrode polishing and conditioning procedures

      • Perform abiotic controls with each experimental batch

      • Validate electrode surface area and roughness

    • Reference strain comparisons:

      • Include wild-type, cytochrome deletion mutants (ΔcbcL), and complemented strains

      • Maintain consistent inoculum preparation

      • Perform measurements at standardized time points during biofilm development

  • Data validation and reporting:

    • Statistical robustness:

      • Minimum of biological triplicates for each condition

      • Appropriate statistical tests with clear reporting of p-values

      • Effect size calculations to assess biological significance

    • Method cross-validation:

      • Verify key findings with complementary techniques

      • Compare results across different growth conditions

      • Validate with in vivo and in vitro approaches

These comprehensive quality control measures ensure that observed phenotypes can be confidently attributed to specific lpxK modifications rather than experimental artifacts or secondary effects .

What emerging technologies could transform our understanding of lpxK function in G. sulfurreducens?

Several cutting-edge technologies show promise for revolutionizing our understanding of lpxK function in G. sulfurreducens:

  • Cryo-electron microscopy for membrane protein structural biology:

    • Single-particle analysis:

      • Determine high-resolution structures of lpxK in different conformational states

      • Visualize substrate binding and catalytic intermediates

      • Map interactions with other membrane proteins

    • Cryo-electron tomography:

      • Visualize lpxK in its native membrane environment

      • Observe spatial organization relative to electron transfer complexes

      • Capture the enzyme in action within intact cells

  • Advanced genetic manipulation tools:

    • CRISPR-Cas9 genome editing:

      • Create precise point mutations in lpxK with minimal off-target effects

      • Generate conditional knockdowns using CRISPRi

      • Implement multiplexed modifications to study lpxK interactions with other pathways

    • Inducible degradation systems:

      • Control lpxK protein levels with temporal precision

      • Study acute vs. chronic effects of lpxK depletion

      • Investigate compensation mechanisms following lpxK loss

  • Synthetic biology approaches:

    • Minimal LPS systems:

      • Reconstitute simplified LPS biosynthetic pathways in vitro

      • Engineer artificial membranes with defined LPS compositions

      • Create orthogonal lpxK variants with altered substrate specificity

    • Bio-orthogonal chemistry:

      • Incorporate clickable lipid A precursors to track biosynthesis in real-time

      • Label newly synthesized LPS for visualization in living cells

      • Map the spatial distribution of lpxK activity across the cell

  • Advanced biophysical techniques:

    • Single-molecule FRET:

      • Monitor lpxK conformational changes during catalysis

      • Measure substrate binding kinetics in membrane environments

      • Observe interactions with other components of the LPS biosynthetic machinery

    • High-speed atomic force microscopy:

      • Visualize dynamic changes in membrane topography

      • Track lpxK movement within the membrane plane

      • Correlate enzyme activity with nanoscale membrane organization

  • Systems biology integration:

    • Multi-omics approaches:

      • Integrate transcriptomics, proteomics, lipidomics, and metabolomics data

      • Develop comprehensive models of LPS biosynthesis regulation

      • Identify unexpected connections between lpxK activity and other cellular processes

    • Machine learning applications:

      • Predict lpxK activity based on environmental variables

      • Identify patterns in electron transfer data not obvious through conventional analysis

      • Optimize experimental design through active learning algorithms

These emerging technologies will provide unprecedented insights into lpxK function at molecular, cellular, and systems levels, potentially transforming our understanding of bacterial membrane biogenesis and electron transfer mechanisms .

How might lpxK research contribute to the development of new antimicrobial strategies targeting Gram-negative pathogens?

Research on lpxK from G. sulfurreducens provides valuable insights that could inform novel antimicrobial strategies targeting Gram-negative pathogens:

  • Structure-guided inhibitor design:

    • The crystal structure of lpxK reveals unique features that can be exploited for selective inhibition:

      • The catalytic site containing the P-loop motif offers opportunities for ATP-competitive inhibitors

      • The membrane-binding interface presents target sites for amphipathic molecules

      • The lipid substrate binding pocket provides a unique target for substrate mimetics

    • The conserved aspartate residues (D138, D139) critical for lpxK function represent promising targets for rational drug design

  • Selective toxicity opportunities:

    • G. sulfurreducens lpxK research illuminates structural features that may differ between environmental bacteria and pathogens:

      • Differences in membrane association regions

      • Substrate binding pocket variations

      • Catalytic residue positioning

    • These differences could be exploited to develop inhibitors that selectively target pathogen lpxK while sparing beneficial environmental bacteria

  • Novel screening approaches:

    • Insights from G. sulfurreducens lpxK enable development of:

      • Cell-based assays monitoring LPS modifications

      • In vitro high-throughput screens using purified recombinant lpxK

      • Structure-based virtual screening against the ATP and lipid binding sites

    • These screens could identify inhibitor scaffolds with activity against multiple Gram-negative species

  • Biofilm disruption strategies:

    • G. sulfurreducens research demonstrates the critical role of LPS in biofilm formation:

      • Compounds modulating lpxK activity could disrupt biofilm development without killing bacteria

      • Sub-inhibitory concentrations might prevent biofilm formation while avoiding selective pressure for resistance

      • Targeting biofilm architecture could enhance the effectiveness of conventional antibiotics

  • Combination therapy approaches:

    • lpxK inhibition could synergize with other treatments by:

      • Increasing membrane permeability to conventional antibiotics

      • Disrupting outer membrane vesiculation mechanisms that contribute to resistance

      • Altering surface charge to enhance interaction with cationic antimicrobial peptides

  • Resistance mitigation strategies:

    • G. sulfurreducens adaptability insights suggest approaches to counter resistance:

      • Multi-target inhibitors affecting several LPS biosynthesis enzymes

      • Molecules that bind to highly conserved regions less prone to mutation

      • Cycling between different inhibitor classes targeting various steps in the pathway

These applications demonstrate how fundamental research on G. sulfurreducens lpxK provides a foundation for addressing the urgent need for new antimicrobial strategies against Gram-negative pathogens, which represent a significant global health challenge .

What potential biotechnological applications could emerge from a deeper understanding of lpxK's role in electron transfer and metal interactions?

A comprehensive understanding of lpxK's role in G. sulfurreducens could catalyze innovative biotechnological applications across multiple fields:

  • Advanced bioelectrochemical systems:

    • Next-generation microbial fuel cells:

      • Engineered strains with optimized lpxK expression for enhanced power output

      • Tailored LPS structures for improved electrode colonization and electron transfer

      • Multi-species electroactive biofilms with complementary electron transfer capabilities

    • Biosensors for environmental monitoring:

      • Living electrochemical sensors with lpxK-dependent response to specific metals

      • Real-time detection systems for environmental contaminants

      • Long-term deployable sensors with stable biofilm architecture

  • Biomaterials with programmable properties:

    • Conductive biofilms for electronic applications:

      • Controlled production of extracellular electron conduits

      • Living electrical components with self-healing properties

      • Biofilm-based computing elements with lpxK-modulated conductivity

    • Biofabricated materials with defined surface chemistry:

      • Tailored hydrophobicity/hydrophilicity through LPS engineering

      • Controlled cell-surface and cell-cell interactions for 3D biomaterial structures

      • Living materials that respond to environmental stimuli through lpxK-mediated adaptations

  • Environmental remediation technologies:

    • Enhanced bioremediation platforms:

      • Engineered strains with optimized metal reduction capabilities

      • Specialized biofilms for targeting specific contaminants

      • Systems with improved resistance to toxic environments

    Metal ContaminantlpxK Optimization GoalExpected Performance Improvement
    UraniumMaximize extracellular reduction>75% reduction of soluble U(VI) to insoluble U(IV)
    ChromiumEnhance Cr(VI) binding and reduction>65% conversion of toxic Cr(VI) to less toxic Cr(III)
    MercuryImprove volatilization or sequestration>50% removal from contaminated sediments
    IronOptimize Fe(III) oxide reduction kinetics>90% increase in reduction rates
  • Synthetic biology applications:

    • Designer cell-surface interfaces:

      • Programmed cell-cell communication through modified LPS structures

      • Control of cellular adhesion properties for tissue engineering

      • Artificial symbiotic relationships between engineered microorganisms

    • Biocomputing platforms:

      • Cellular logic gates based on electron transfer properties

      • Redox-based information processing systems

      • Environmental computing with distributed bacterial networks

  • Sustainable manufacturing processes:

    • Bioelectrosynthesis:

      • Direct electron input for microbial production of value-added chemicals

      • Carbon dioxide capture and conversion to fuels

      • lpxK-optimized strains for efficient electron uptake from cathodes

    • Green mining technologies:

      • Enhanced metal extraction from low-grade ores

      • Selective recovery of valuable metals from waste streams

      • Closed-loop metal cycling in industrial processes

These diverse applications leverage the fundamental understanding of lpxK's role in shaping bacterial surface properties and electron transfer capabilities, potentially addressing significant challenges in renewable energy, environmental remediation, and sustainable manufacturing .

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.