Recombinant Geobacter sulfurreducens LL-diaminopimelate aminotransferase (dapL)

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 preparation.
Lead Time
Delivery times vary depending on the purchase method and location. Please contact your local distributor for precise delivery estimates.
Note: Standard shipping includes 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 settle 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 be used as a reference.
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 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 manufacturing.
The tag type will be determined during the production process. If you require a specific tag, please inform us; we will prioritize its development.
Synonyms
dapL; GSU0162; LL-diaminopimelate aminotransferase; DAP-AT; DAP-aminotransferase; LL-DAP-aminotransferase; EC 2.6.1.83
Buffer Before Lyophilization
Tris/PBS-based buffer, 6% Trehalose.
Datasheet
Please contact us to get it.
Expression Region
1-410
Protein Length
full length protein
Purity
>85% (SDS-PAGE)
Species
Geobacter sulfurreducens (strain ATCC 51573 / DSM 12127 / PCA)
Target Names
dapL
Target Protein Sequence
MAKINDHYLK LKAGYLFPEI GRRVREFAAA NPSAKVIRLG IGDVTRPLAP AVIKAFHEAV DDLATTENFA GYGPEQGYDW LINAIIEKSY KPLGVDLKTE EMFISDGSKC DCANILDIFA LDNVVAIGDP VYPVYNDTNV MIGRTGEADD KGYYKGIVYM PCTEENGFIP SLPTEKVDII YLCFPNNPTG TVATKAELKK WVDYAIANDA VIFFDAAYEA FITDPAIPHS IYEIEGAKKC AIEFRSFSKT AGFTGVRCGL VVVPEEVMGT TPTGEKYSFN KLWLRRTTTK FNGASYPVQK AAAAVYSDEG WQQNKEIIDY YMENARIIRE GLAAAGLTVY GGVNAPYIWL KTPGGMSSWD FFDKLLNECN VVGTPGSGFG PSGEGFFRLS AFGHRENVIE AVERIKKNLK
Uniprot No.

Target Background

Function
This protein is involved in meso-diaminopimelate (m-DAP or DL-DAP) synthesis, crucial for both lysine and peptidoglycan biosynthesis. It catalyzes the direct conversion of tetrahydrodipicolinate to LL-diaminopimelate.
Database Links

KEGG: gsu:GSU0162

STRING: 243231.GSU0162

Protein Families
Class-I pyridoxal-phosphate-dependent aminotransferase family, LL-diaminopimelate aminotransferase subfamily

Q&A

What is the biochemical function of L,L-diaminopimelate aminotransferase in Geobacter sulfurreducens?

L,L-diaminopimelate aminotransferase (DapL) in G. sulfurreducens catalyzes the direct conversion of tetrahydrodipicolinate (THDP) to L,L-diaminopimelic acid (L,L-DAP) . This reaction represents a key shortcut in the diaminopimelate/lysine biosynthesis pathway, bypassing three enzymatic steps found in the conventional pathway (dapD, dapC, and dapE) . The enzyme performs an aminotransferase reaction using glutamate as the amino donor, resulting in the formation of L,L-DAP and α-ketoglutarate . This bypasses the typical succinylated or acetylated intermediates found in other bacterial DAP synthesis pathways .

The biochemical reaction can be represented as:
Tetrahydrodipicolinate+L-glutamateL,L-diaminopimelic acid+2-oxoglutarate+H2O\text{Tetrahydrodipicolinate} + \text{L-glutamate} \rightarrow \text{L,L-diaminopimelic acid} + \text{2-oxoglutarate} + \text{H}_2\text{O}

Unlike the variant found in some organisms, G. sulfurreducens DapL forms part of a complete lysine biosynthesis pathway that includes other enzymes such as dihydrodipicolinate synthase (dapA) , which generates the substrate for DapL, and diaminopimelate epimerase (dapF), which converts L,L-DAP to meso-DAP, the direct precursor of lysine .

What expression systems are most effective for producing recombinant G. sulfurreducens DapL?

Based on successful expression strategies for other G. sulfurreducens proteins and related DapL enzymes, the following approach is recommended:

Heterologous Expression in E. coli:

  • The gene encoding G. sulfurreducens DapL should be PCR-amplified from genomic DNA and cloned into an expression vector containing an inducible promoter (such as T7 or tac) .

  • Expression in E. coli BL21(DE3) or similar strains has proven effective for G. sulfurreducens proteins .

  • For optimal expression, codon optimization may be necessary, as G. sulfurreducens has a different codon usage bias than E. coli .

Purification Strategy:

  • Incorporate a C-terminal hexahistidine tag rather than N-terminal, as N-terminal tags may interfere with proper folding (as observed with other G. sulfurreducens proteins) .

  • Purify using nickel affinity chromatography followed by size exclusion chromatography.

  • Typical yields of 5-10 mg/L of culture can be expected based on similar recombinant proteins .

Alternative Expression Systems:
For researchers encountering difficulty with E. coli expression, mammalian cell expression systems have been successfully used for other G. sulfurreducens proteins such as dihydrodipicolinate synthase (dapA) .

How can researchers verify the enzymatic activity of recombinant G. sulfurreducens DapL?

Several complementary approaches can be used to confirm enzymatic activity:

In vitro Enzymatic Assays:

  • Forward Reaction Assay: Measure the formation of L,L-DAP from THDP and glutamate spectrophotometrically .

  • Reverse Reaction Assay: Monitor the conversion of L,L-DAP and α-ketoglutarate to THDP and glutamate .

Kinetic Parameters Determination:

  • Determine apparent Km values for both substrates (THDP and glutamate) .

  • Based on related DapL enzymes, expected Km values would be approximately:

    • THDP: 80-120 μM

    • α-ketoglutarate: 0.4-0.6 mM

Functional Complementation:

  • Transform E. coli DAP auxotrophs (ΔdapD or ΔdapD ΔdapE) with the G. sulfurreducens dapL gene .

  • Successful complementation (growth without DAP supplementation) confirms functional activity .

  • Important control: A ΔdapD ΔdapF double mutant should not be complemented, as this would confirm the enzyme specifically produces L,L-DAP rather than meso-DAP .

Specialized Activity Verification:

  • Test substrate specificity by examining whether the enzyme can utilize alternative substrates such as succinyl-DAP or acetyl-DAP .

  • Examine activity at different pH values and temperatures to determine optimal conditions, which typically would be pH 8.0-9.0 for related enzymes .

What are the structural characteristics of G. sulfurreducens DapL and how do they compare to DapL enzymes from other organisms?

While the specific crystal structure of G. sulfurreducens DapL has not been directly reported in the provided sources, comparative analysis allows for structural predictions:

Predicted Structural Features:

  • G. sulfurreducens DapL likely belongs to the aminotransferase class I/II superfamily .

  • The enzyme is expected to be PLP (pyridoxal-5'-phosphate)-dependent, with a characteristic PLP-binding domain .

  • Based on homologous enzymes, a homodimeric quaternary structure is likely .

Comparative Analysis:
G. sulfurreducens DapL shares significant structural similarities with:

  • Arabidopsis thaliana DapL (determined at 1.95 Å resolution)

  • Chlamydia trachomatis CT390 protein, functioning as a DapL

  • Protochlamydia amoebophila PC0685, sharing approximately 42.6% amino acid identity with the C. trachomatis enzyme

Functional Domains:

  • PLP-binding domain containing a conserved lysine residue for cofactor attachment

  • Substrate-binding pocket adapted for THDP and glutamate

  • Dimerization interface

Phylogenetic Classification:
G. sulfurreducens DapL likely belongs to the DapL1 phylogenetic group, similar to the enzyme from Methanothermobacter thermautotrophicus, based on sequence homology patterns observed in other organisms .

How does the DapL pathway in G. sulfurreducens differ from other bacterial diaminopimelate/lysine biosynthesis pathways?

The diaminopimelate/lysine biosynthesis pathway in G. sulfurreducens employs the DapL variant, which differs significantly from other bacterial pathways:

Comparison of DAP/Lysine Biosynthesis Pathways:

PathwayKey EnzymesIntermediate StepsOrganismsReference
DapL PathwayDapL (L,L-DAP aminotransferase)Direct conversion of THDP to L,L-DAPG. sulfurreducens, Chlamydia, plants, some archaea
Succinylase PathwayDapD, DapC, DapEUses succinylated intermediatesMost proteobacteria, E. coli
Acetylase PathwaySimilar to succinylase but with acetylated intermediatesUses acetylated intermediatesSome Bacillus species
Dehydrogenase PathwayDdh (meso-DAP dehydrogenase)Direct conversion of THDP to meso-DAPCorynebacterium, Brevibacterium

Key Differences:

  • The DapL pathway provides a more direct route, requiring only one enzyme instead of three (DapD, DapC, and DapE) .

  • G. sulfurreducens lacks the genes encoding DapD, DapC, and DapE enzymes that are present in organisms using the succinylase pathway .

  • The DapL pathway is more energy-efficient as it doesn't require acylation and deacylation steps .

  • Unlike the dehydrogenase pathway, which produces meso-DAP directly, the DapL pathway produces L,L-DAP that must be epimerized to meso-DAP by DapF .

Evolutionary Implications:

  • The presence of the DapL pathway in G. sulfurreducens supports an evolutionary relationship among chlamydiae, cyanobacteria, and plants .

  • Phylogenetic analyses suggest lateral gene transfers occurred in DapL genes, with one transfer from archaea to bacteria (DapL2 group) and another from bacteria to archaea (DapL1 group) .

How can researchers investigate the physiological role of DapL in G. sulfurreducens using genetic approaches?

To investigate the physiological significance of DapL in G. sulfurreducens, several genetic approaches can be employed:

Gene Knockout/Deletion Studies:

  • Generate a dapL deletion mutant in G. sulfurreducens using homologous recombination or CRISPR-Cas9 techniques.

  • Assess growth phenotypes in media with and without lysine/DAP supplementation.

  • Based on studies in other organisms, the mutant would likely exhibit lysine auxotrophy .

Complementation Analysis:

  • Complement the G. sulfurreducens dapL mutant with dapL genes from other organisms (e.g., C. trachomatis, plants) to assess functional conservation .

  • Introduce alternative DAP synthesis pathway genes (e.g., dapD, dapC, dapE) to determine if they can functionally replace dapL .

Expression Analysis:

  • Use quantitative RT-PCR to analyze dapL expression under different growth conditions .

  • Compare expression patterns with other genes in lysine biosynthesis pathway (asd, dapB, dapF) .

  • Based on findings in C. trachomatis, expression may begin early during growth (as early as 8 hours after inoculation) .

Metabolic Profiling:

Conditional Expression Systems:

  • Develop a conditional expression system for dapL in G. sulfurreducens to study the effects of varying expression levels on growth and metabolism.

  • This approach can reveal whether dapL is essential under all growth conditions or only under specific circumstances.

What are the kinetic properties of G. sulfurreducens DapL and how do they compare to DapL enzymes from other organisms?

While specific kinetic data for G. sulfurreducens DapL isn't directly provided in the search results, comparative analysis with related DapL enzymes allows for predictions and experimental design:

Expected Kinetic Parameters:

ParameterExpected Value for G. sulfurreducens DapLComparative Value from Related DapLOrganism Source for ComparisonReference
Km for L,L-DAP (reverse reaction)80-100 μM82.8 ± 10 μMMethanocaldococcus jannaschii
Km for α-ketoglutarate (reverse reaction)0.4-0.5 mM0.42 ± 0.02 mMMethanocaldococcus jannaschii
Specific activity (L,L-DAP + α-ketoglutarate)20-30 nmol·min⁻¹·mg⁻¹24.3 ± 2.0 nmol·min⁻¹·mg⁻¹Methanococcus maripaludis
pH optimum8.0-9.08.0-9.0Methanocaldococcus jannaschii
Temperature optimum35-37°C35°C (growth temperature)Geobacter sulfurreducens

Substrate Specificity:

  • Like other characterized DapL enzymes, G. sulfurreducens DapL is expected to be unable to use succinyl-DAP or acetyl-DAP as substrates .

  • The enzyme may show activity with both L,L-DAP and meso-DAP in the reverse reaction, similar to the C. trachomatis enzyme, though the physiological relevance of this is questionable .

Experimental Approaches for Kinetic Characterization:

  • Steady-state kinetics using varying concentrations of substrates

  • Product inhibition studies

  • pH-rate profiles to determine key ionizable groups

  • Temperature-dependence studies to determine activation parameters

Comparing with Plant DapL:
The G. sulfurreducens enzyme likely differs from plant DapL enzymes (e.g., Arabidopsis), which appear to be specific for L,L-DAP and cannot use meso-DAP in the reverse reaction .

What experimental procedures are required to determine the crystal structure of G. sulfurreducens DapL?

Determining the crystal structure of G. sulfurreducens DapL would require the following comprehensive approach:

Protein Expression and Purification:

  • Express recombinant G. sulfurreducens DapL with a cleavable affinity tag (His-tag recommended) .

  • Purify to >95% homogeneity using multiple chromatography steps (affinity, ion exchange, size exclusion) .

  • Verify protein identity by mass spectrometry and N-terminal sequencing.

  • Assess protein quality using dynamic light scattering to ensure monodispersity.

Crystallization:

  • Screen multiple crystallization conditions using commercial screens (e.g., Hampton Research, Molecular Dimensions).

  • Optimize promising conditions by varying:

    • Protein concentration (typically 5-15 mg/mL)

    • Precipitant type and concentration

    • Buffer pH and composition

    • Temperature

    • Additives

  • Consider co-crystallization with:

    • PLP cofactor

    • Substrate analogs

    • Inhibitors

Data Collection and Processing:

  • Prepare crystals for diffraction by cryoprotection and flash-cooling in liquid nitrogen.

  • Collect X-ray diffraction data at a synchrotron facility.

  • Process diffraction data using appropriate software (XDS, HKL2000, MOSFLM).

Structure Determination:

  • Use molecular replacement with existing DapL structures as search models:

    • Arabidopsis thaliana DapL (PDB: 2Z1Z, 2Z20)

    • C. trachomatis DapL if available

  • Alternatively, prepare selenomethionine-labeled protein for MAD/SAD phasing .

  • Build and refine the model using crystallographic software (PHENIX, REFMAC, COOT).

Structure Validation and Analysis:

  • Validate using tools such as MolProbity and PROCHECK.

  • Analyze:

    • Active site architecture

    • PLP binding mode

    • Substrate binding sites

    • Oligomeric interfaces

    • Comparison with other DapL structures

What is the evolutionary significance of the DapL pathway in G. sulfurreducens and related bacteria?

The presence of the DapL pathway in G. sulfurreducens has several important evolutionary implications:

Phylogenetic Distribution and Evolutionary History:

  • The DapL pathway has been identified in approximately 13% of sequenced bacterial genomes (381 out of 2771 as of May 2014) .

  • Its presence in G. sulfurreducens and other Geobacteraceae suggests specific evolutionary adaptations in this family .

  • Phylogenetic analyses indicate that DapL genes have undergone lateral gene transfer events between domains of life :

    • From archaea to bacteria in the DapL2 group

    • From bacteria to archaea in the DapL1 group (which likely includes G. sulfurreducens DapL)

Evolutionary Relationships:

  • The DapL pathway in G. sulfurreducens supports an evolutionary relationship among the Geobacteraceae, chlamydiae, cyanobacteria, and plants .

  • This suggests ancient lateral gene transfers between these diverse groups or retention of an ancestral pathway that was lost in many other bacterial lineages.

Metabolic Evolution:

  • The DapL pathway represents a more direct and potentially energy-efficient route to lysine biosynthesis compared to the multi-step succinylase pathway .

  • Its retention in G. sulfurreducens may reflect selection for metabolic efficiency in environments where energy conservation is crucial.

Cell Wall Evolution:

  • The presence of the DapL pathway in G. sulfurreducens strengthens the argument that these bacteria synthesize a peptidoglycan cell wall, despite difficulties in detecting it experimentally .

  • This supports the hypothesis that the common ancestor of this bacterial group possessed a conventional cell wall.

How can G. sulfurreducens DapL be explored as a potential target for antimicrobial development?

G. sulfurreducens DapL represents a promising target for antimicrobial development due to several key factors:

Target Validation Rationale:

  • The DAP/lysine biosynthesis pathway is absent in humans, who acquire lysine from dietary sources .

  • meso-DAP is essential for peptidoglycan synthesis in most Gram-negative bacteria, while lysine plays a similar role in most Gram-positive bacteria .

  • DapL has a narrow distribution (approximately 13% of sequenced bacterial genomes), potentially allowing for targeted antimicrobial development .

  • The DapL pathway is present in several pathogenic bacteria including Chlamydia, Leptospira, and Treponema species .

Inhibitor Design Strategies:

  • Structure-based design:

    • Target the unique active site architecture of DapL

    • Design transition state analogs based on the aminotransferase reaction

    • Focus on compounds that mimic THDP binding

  • High-throughput screening:

    • Develop robust enzymatic assays suitable for screening compound libraries

    • Screen natural product libraries, as these may contain compounds evolved to target similar enzymes

  • Fragment-based approach:

    • Identify small molecular fragments that bind to different regions of DapL

    • Link promising fragments to create potent inhibitors

Target Validation Experiments:

  • Confirm essentiality of dapL in G. sulfurreducens through gene knockout studies coupled with complementation

  • Demonstrate that chemical inhibition of DapL activity correlates with growth inhibition

  • Show that growth inhibition can be reversed by supplementation with meso-DAP or lysine

Potential Advantages as a Drug Target:

  • Inhibitors may have narrow-spectrum activity, reducing impact on beneficial microbiota

  • The pathway is essential in both growing and non-growing bacteria (for cell wall maintenance)

  • The unique structure of DapL may allow for selective targeting compared to human aminotransferases

What techniques can be used to study the regulation of dapL gene expression in G. sulfurreducens?

Understanding the regulation of dapL expression in G. sulfurreducens requires a comprehensive approach using several molecular techniques:

Transcriptional Analysis:

  • RT-qPCR: Quantify dapL transcript levels under different growth conditions, including:

    • Different carbon sources (acetate, butyrate, pyruvate)

    • Varying environmental stresses (temperature, pH, oxidative stress)

    • Different growth phases (exponential vs. stationary)

    • Compare expression patterns with other genes in the lysine biosynthesis pathway

  • RNA-Seq: Perform global transcriptomic analysis to:

    • Identify co-regulated genes

    • Detect potential antisense transcripts or regulatory RNAs affecting dapL expression

    • Map transcription start sites and termination sites

Promoter Analysis:

  • 5' RACE (Rapid Amplification of cDNA Ends): Identify the transcription start site(s) of dapL

  • Reporter Gene Assays: Fuse putative promoter regions to reporter genes (e.g., lacZ, gfp) to:

    • Define the minimal functional promoter

    • Identify regulatory elements through deletion analysis

    • Assess promoter activity under different conditions

Regulatory Protein Identification:

  • ChIP-seq (Chromatin Immunoprecipitation followed by Sequencing): Identify proteins binding to the dapL promoter region

  • EMSA (Electrophoretic Mobility Shift Assay): Confirm specific binding of regulatory proteins to dapL promoter sequences

  • DNA Affinity Purification: Isolate proteins binding to the dapL promoter followed by mass spectrometry identification

Metabolic Regulation Studies:

  • Metabolomics: Correlate changes in intracellular metabolites (especially lysine pathway intermediates) with dapL expression

  • Ribosome Profiling: Assess translational regulation of dapL

Comparative Analysis:

  • Compare regulatory patterns with those of dapL genes in related organisms

  • In G. metallireducens, analysis of the ModE regulon revealed loss of the global regulatory protein ModE but retention of some putative ModE-binding sites , suggesting complex regulatory evolution that might also affect dapL regulation.

What are the best methods for studying protein-protein interactions involving G. sulfurreducens DapL?

Investigating protein-protein interactions involving G. sulfurreducens DapL requires multiple complementary approaches:

In vitro Interaction Studies:

  • Pull-down Assays:

    • Express DapL with an affinity tag (e.g., His-tag)

    • Immobilize on appropriate resin

    • Incubate with G. sulfurreducens cell lysate

    • Identify bound proteins by mass spectrometry

  • Surface Plasmon Resonance (SPR):

    • Immobilize purified DapL on sensor chip

    • Flow potential interacting proteins over the surface

    • Measure binding kinetics and affinity constants

    • Particularly useful for transient interactions

  • Isothermal Titration Calorimetry (ITC):

    • Provides thermodynamic parameters of interactions

    • Determine binding stoichiometry, affinity, enthalpy, and entropy

In vivo Interaction Studies:

  • Bacterial Two-Hybrid System:

    • Adapt for use in G. sulfurreducens or use heterologous hosts

    • Screen for interactions with libraries of G. sulfurreducens proteins

  • Co-immunoprecipitation (Co-IP):

    • Generate antibodies against DapL or use epitope-tagged version

    • Precipitate DapL complexes from G. sulfurreducens lysates

    • Identify co-precipitating proteins by mass spectrometry

  • Cross-linking Mass Spectrometry:

    • Treat G. sulfurreducens cells with cross-linking agents

    • Isolate DapL complexes

    • Identify cross-linked peptides by mass spectrometry

    • Precisely map interaction interfaces

Structural Studies of Complexes:

  • X-ray Crystallography of Co-crystals:

    • Crystallize DapL with interacting partners

    • Determine atomic-resolution structure of the complex

  • Cryo-Electron Microscopy:

    • Visualize larger complexes involving DapL

    • Particularly useful for transient or dynamic interactions

Functional Validation:

  • Mutagenesis of Interaction Interfaces:

    • Create point mutations in predicted interaction regions

    • Assess effects on both binding and enzymatic activity

  • Enzyme Activity Modulation:

    • Test whether potential interacting proteins affect DapL catalytic activity

    • Measure kinetic parameters in presence/absence of binding partners

Potential Interacting Partners to Investigate:

  • Other enzymes in the lysine biosynthesis pathway (DapA, DapB, DapF)

  • Potential scaffolding proteins that may organize metabolic pathways

  • Regulatory proteins identified in expression studies

How does the metabolic context of G. sulfurreducens impact DapL function and lysine biosynthesis?

The metabolic network of G. sulfurreducens creates a unique context for DapL function and lysine biosynthesis:

Integration with Central Carbon Metabolism:

  • G. sulfurreducens has a more versatile carbon metabolism than related species such as G. metallireducens, utilizing acetate, pyruvate, and other carbon sources .

  • The aspartate metabolic pathway, which includes the DAP/lysine synthesis branch, also feeds into methionine, threonine, and isoleucine synthesis .

  • The conversion of L-aspartate to L-aspartate-semialdehyde by LysC and Asd is a crucial step that links lysine biosynthesis to central carbon metabolism .

Energetic Considerations:

  • G. sulfurreducens obtains energy through anaerobic respiration using Fe(III) or other metal ions as terminal electron acceptors .

  • The DapL pathway's efficiency (bypassing three enzymatic steps of the conventional pathway) may be particularly advantageous in this energy-limited anaerobic lifestyle .

  • The metabolic cost of lysine biosynthesis must be balanced against the need for this amino acid in protein synthesis and cell wall formation.

Coordination with Electron Transport and Respiration:

  • Lysine connects to the mitochondrial electron transport chain and the tricarboxylic acid cycle in some organisms .

  • In G. sulfurreducens, which possesses numerous c-type cytochromes for metal reduction , there may be regulatory cross-talk between lysine metabolism and electron transport systems.

Metabolic Regulation:

  • Comparison with plants suggests that disruptions in lysine biosynthesis can lead to broad metabolic shifts, including alterations in:

    • Photosynthesis-related proteins

    • Photorespiration

    • Amino acid ratios (particularly glycine/serine)

    • Accumulation of stress-responsive amino acids

  • Similar metabolic adaptations might occur in G. sulfurreducens under conditions that affect DapL activity.

Unique Aspects of G. sulfurreducens Metabolism:

  • G. sulfurreducens possesses complete pathways for TCA cycle, glycolysis, and gluconeogenesis .

  • It has a range of amino acid biosynthesis pathways, with lysine biosynthesis being particularly important due to its dual role in protein and cell wall synthesis .

  • The organism can grow on minimal media without amino acid supplementation, indicating complete biosynthetic capabilities .

What considerations are important when designing inhibitors specific for G. sulfurreducens DapL?

When designing inhibitors specific for G. sulfurreducens DapL, several critical considerations must be addressed:

Structural Determinants of Specificity:

  • Target unique structural features of G. sulfurreducens DapL that differ from:

    • Human aminotransferases to ensure safety

    • DapL enzymes from beneficial bacteria to minimize microbiome disruption

    • Other aminotransferases in G. sulfurreducens to avoid off-target effects

  • Consider the following structural elements:

    • Active site architecture, particularly substrate-binding residues

    • PLP-binding pocket variations

    • Surface loops that differ among DapL orthologs

    • Allosteric sites unique to G. sulfurreducens DapL

Mechanism-Based Considerations:

  • Design transition state analogs specific to the DapL reaction

  • Develop covalent inhibitors that target conserved active site residues

  • Consider time-dependent inhibition strategies that may provide greater selectivity

Physical Property Requirements:

  • For G. sulfurreducens, which has a Gram-negative-like cell envelope, inhibitors must:

    • Penetrate the outer membrane (typically requires molecular weight <600 Da)

    • Navigate the periplasmic space

    • Cross the cytoplasmic membrane (typically requires moderate lipophilicity)

  • Consider the anaerobic environment in which G. sulfurreducens typically grows

Rational Design Approaches:

  • Fragment-Based Drug Design:

    • Identify small molecular fragments that bind to different regions of the active site

    • Link fragments to create high-affinity, specific inhibitors

  • Computer-Aided Drug Design:

    • Perform virtual screening of compound libraries against modeled G. sulfurreducens DapL structure

    • Use molecular dynamics simulations to identify transient binding pockets

    • Employ quantum mechanical calculations to optimize transition state mimics

Experimental Validation Pipeline:

  • In vitro enzyme inhibition assays with purified recombinant G. sulfurreducens DapL

  • Counter-screening against human aminotransferases and DapL enzymes from other organisms

  • Assessment of antimicrobial activity against G. sulfurreducens and related species

  • Mechanism of action studies to confirm on-target activity (metabolite profiling, resistance development)

  • Structural studies of enzyme-inhibitor complexes to guide optimization

How can researchers study the role of DapL in G. sulfurreducens cell wall biosynthesis?

Investigating the role of DapL in G. sulfurreducens cell wall biosynthesis requires specialized approaches due to the challenges in detecting peptidoglycan in this organism:

Cell Wall Analysis Techniques:

  • Enhanced Peptidoglycan Detection:

    • Use modified extraction methods optimized for bacteria with minimal peptidoglycan

    • Apply sensitive analytical techniques such as HPLC-MS to detect muropeptides

    • Use fluorescent D-amino acid derivatives (FDAAs) to label newly synthesized peptidoglycan

  • Microscopy Approaches:

    • Transmission electron microscopy with specific staining for cell envelope components

    • Cryo-electron tomography to visualize native cell envelope architecture

    • Super-resolution microscopy using peptidoglycan-specific probes

  • Biochemical Analysis:

    • Quantify meso-DAP content in cell wall fractions

    • Analyze peptidoglycan cross-linking patterns

    • Assess incorporation of radiolabeled DAP into cell wall material

Genetic Manipulation Approaches:

  • Conditional DapL Expression:

    • Create strains with controllable dapL expression

    • Observe effects of DapL depletion on cell morphology and wall integrity

    • Supplement with exogenous DAP to distinguish protein synthesis from cell wall effects

  • Reporter Systems:

    • Create fusions between cell wall stress response promoters and reporter genes

    • Monitor activation in response to DapL inhibition or depletion

  • Bypass of meso-DAP Requirement:

    • Express alternative cell wall cross-linking mechanisms that don't require meso-DAP

    • Determine if this can rescue dapL deficiency

Physiological Studies:

  • Cell Wall Stress Sensitivity:

    • Test sensitivity to cell wall-targeting antibiotics (β-lactams, fosfomycin)

    • Compare wild-type and DapL-depleted cells

  • Osmotic Challenge Experiments:

    • Assess survival under osmotic shock conditions

    • Quantify cell lysis rates under various conditions

  • Biofilm Formation:

    • Evaluate impacts on biofilm development and architecture

    • Assess cell-cell adhesion properties

Comparative Analysis:

  • Compare findings with observations from organisms where peptidoglycan is more readily detected

  • The presence of the DapL pathway supports the argument that G. sulfurreducens synthesizes a peptidoglycan cell wall despite difficulties in detecting it experimentally

Metabolic Labeling:

  • Incorporate isotope-labeled precursors of peptidoglycan synthesis

  • Track their incorporation into cell wall components

  • Use metabolic flux analysis to determine how DapL activity influences cell wall synthesis rates

What approaches can be used to optimize the catalytic efficiency of recombinant G. sulfurreducens DapL for biotechnological applications?

Optimizing the catalytic efficiency of recombinant G. sulfurreducens DapL for biotechnological applications involves multiple strategies:

Protein Engineering Approaches:

  • Rational Design:

    • Identify rate-limiting steps through kinetic analysis

    • Modify active site residues to enhance substrate binding or catalysis

    • Introduce disulfide bridges to increase thermostability

    • Remove surface-exposed hydrophobic residues to improve solubility

  • Directed Evolution:

    • Develop high-throughput screening assays for DapL activity

    • Create libraries of variants through error-prone PCR or DNA shuffling

    • Select mutants with improved activity, stability, or specificity

    • Combine beneficial mutations through iterative rounds of selection

  • Semi-rational Design:

    • Use computational tools to identify hotspots for mutagenesis

    • Create focused libraries targeting specific regions

    • Apply machine learning to predict beneficial mutations

Expression Optimization:

  • Codon Optimization:

    • Adjust codon usage to match expression host preferences

    • Remove rare codons that might limit translation efficiency

  • Expression System Selection:

    • Test various hosts including E. coli, yeast, or cell-free systems

    • Evaluate different promoters, ribosome binding sites, and fusion partners

    • Consider inducible versus constitutive expression strategies

  • Fusion Protein Strategies:

    • Test N- or C-terminal fusion partners that enhance solubility (MBP, SUMO, etc.)

    • Incorporate affinity tags for simplified purification

Reaction Condition Optimization:

  • Buffer Composition:

    • Screen different buffers, pH values, and ionic strengths

    • Test effects of metal ions and other additives

  • Co-factor Enhancement:

    • Ensure optimal PLP incorporation

    • Test PLP derivatives or analogs that might enhance activity

  • Substrate Engineering:

    • Modify substrates to improve binding or catalytic turnover

    • Consider alternative substrates for novel applications

Immobilization Strategies:

  • Carrier Selection:

    • Test various carrier materials (resins, nanoparticles, membranes)

    • Optimize immobilization chemistry to maintain activity

  • Orientation Control:

    • Design specific attachment points to ensure optimal orientation

    • Use site-specific immobilization to avoid active site blockage

  • Stabilization Effects:

    • Exploit rigidification through multipoint attachment

    • Create favorable microenvironments around immobilized enzyme

Potential Biotechnological Applications:

  • Production of isotopically labeled amino acids for metabolic studies

  • Synthesis of non-canonical amino acid derivatives

  • Development of biosensors for metabolic pathway intermediates

  • Use in biocatalytic cascades for complex molecule synthesis

How does the amino acid sequence of G. sulfurreducens DapL contribute to its enzymatic properties?

The amino acid sequence of G. sulfurreducens DapL determines its structure and function through several key sequence features:

Functional Domains and Motifs:

  • PLP-Binding Site:

    • Contains a conserved lysine residue that forms a Schiff base with PLP

    • Likely includes a characteristic PROSITE pattern for aminotransferase class-I pyridoxal-phosphate attachment site: [LIVMFTA]-[LIVMTAFC]-[LIVMFYWTA]-D-[LIVMFYAHR]-[LIVMFYWSTAG]-[GSA]-[GSAPDEM]-[LIVMFYSTANQHC]-K-x(2)-[GSADEBQKRHNPL]

    • Mutations in this region would significantly impact catalytic activity

  • Substrate Binding Pocket:

    • Residues that interact with THDP and glutamate

    • The pocket architecture determines substrate specificity and catalytic efficiency

    • Based on other DapL enzymes, likely includes conserved arginine residues for carboxylate binding

  • Dimerization Interface:

    • Residues involved in subunit interactions

    • Important for maintaining the quaternary structure necessary for activity

Sequence-Based Classification:

  • G. sulfurreducens DapL likely belongs to the DapL1 phylogenetic group, similar to enzymes from archaea like Methanothermobacter thermautotrophicus

  • Sequence analysis can predict whether it shares properties with bacterial or archaeal DapL enzymes

Comparative Sequence Analysis:

  • Shares significant sequence identity with DapL enzymes from:

    • Chlamydia trachomatis (CT390)

    • Protochlamydia amoebophila (PC0685, ~42.6% identity to CT390)

    • Arabidopsis thaliana DapL

  • These sequence relationships suggest similar enzymatic properties and substrate preferences

Key Sequence Determinants of Activity:

  • Catalytic Residues:

    • Conserved residues directly involved in proton transfer and transition state stabilization

    • Likely include acidic and basic residues positioned precisely within the active site

  • Substrate Specificity Determinants:

    • Residues that form the binding pocket for THDP and glutamate

    • Sequence variations in these regions account for differences in substrate preference between DapL enzymes

  • Thermostability Factors:

    • Distribution of charged residues on the protein surface

    • Proline content in loop regions

    • Presence of stabilizing salt bridges and hydrophobic interactions

Structure Prediction and Analysis:

  • Secondary structure prediction reveals the arrangement of α-helices and β-sheets

  • Tertiary structure modeling based on homologous enzymes provides insights into the three-dimensional arrangement of catalytic residues

  • Analysis of surface-exposed regions identifies potential epitopes for antibody production

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.