KEGG: lpl:lp_2361
STRING: 220668.lp_2361
UDP-N-acetylglucosamine 1-carboxyvinyltransferase 1 (murA1) is an essential enzyme that catalyzes the first committed step in bacterial peptidoglycan biosynthesis. In L. plantarum, murA1 transfers the enolpyruvate moiety of phosphoenolpyruvate (PEP) to UDP-N-acetylglucosamine (UNAG), forming UDP-N-acetylglucosamine-enolpyruvate, which is critical for cell wall formation . MurA1 is particularly significant in L. plantarum as it has been identified as one of the key stress-responsive proteins that helps maintain cell wall integrity during environmental challenges such as cold and acid stress conditions . The enzyme is part of the broader Mur family proteins (including MurB, MurC, MurD, MurE1, and MurF) that collectively regulate peptidoglycan synthesis, which is essential for bacterial survival, growth, and resistance to external stressors.
MurA1 functions at the initial stage of the peptidoglycan synthesis pathway in L. plantarum. The complete pathway involves:
MurA1 catalyzes the transfer of enolpyruvate from PEP to UNAG
MurB reduces the enolpyruvate moiety to form UDP-N-acetylmuramic acid (UNAM)
MurC adds L-alanine to UNAM
MurD incorporates D-glutamate
MurE1 adds meso-diaminopimelic acid or L-lysine
MurF attaches D-alanyl-D-alanine
This sequential process forms the complete peptidoglycan precursor that is then incorporated into the cell wall structure . In response to stress conditions, L. plantarum has been observed to upregulate proteins including DacA1, DacB, MurA1, MurB, MurC, MurD, MurE1, and MurF to increase total peptidoglycan production, which enhances cellular protection against environmental challenges .
MurA1 has several distinctive biochemical characteristics:
It utilizes a unique catalytic mechanism involving the formation of a tetrahedral intermediate between PEP and UNAG
Contains a critical cysteine residue (similar to Cys-115 in Enterobacter cloacae MurA) that is essential for catalytic activity and forms a covalent adduct with PEP
Is specifically targeted by the antibiotic fosfomycin, which covalently modifies the active site cysteine residue
Has structural and sequence similarity to 5-enolpyruvylshikimate 3-phosphate synthase, the only other known enzyme that catalyzes the transfer of enolpyruvate from PEP to a substrate
Forms part of a feedback regulation system, where the enzyme can exist in a "dormant" complex with UNAM and covalently bound PEP, enabling efficient regulation of murein biosynthesis
For recombinant expression of L. plantarum murA1, several expression systems have proven effective:
The selection of an appropriate expression system depends on the research objectives. For structural and biochemical characterization, E. coli systems typically provide sufficient yields. For immunological applications and vaccine development, expression in L. plantarum itself using vectors like pSIP409 offers advantages of proper protein folding and potential for surface display .
Purification of recombinant murA1 requires a systematic approach:
Cell Lysis: Optimized lysis buffers containing protease inhibitors to prevent degradation of the target protein
Initial Capture:
Affinity chromatography using His-tagged constructs with Ni-NTA resin
Alternative affinity tags (GST, MBP) if solubility issues are encountered
Intermediate Purification:
Ion exchange chromatography (typically DEAE or Q-Sepharose)
Optimization of pH and salt gradients based on murA1's theoretical pI
Polishing:
Size exclusion chromatography to separate monomeric from aggregated forms
Typical yields of >95% purity can be achieved through this three-step process
Quality Control:
SDS-PAGE and Western blotting to confirm identity
Activity assays to verify functional integrity
Mass spectrometry to confirm protein sequence and modifications
The purification strategy should be tailored to the intended application, with structural studies requiring higher purity than functional assays or immunological applications.
MurA1 enzymatic activity can be measured through several complementary approaches:
Spectrophotometric Coupled Assay:
Measures the release of inorganic phosphate during the transfer of enolpyruvate
Uses enzymes like pyruvate kinase and lactate dehydrogenase to couple PEP utilization to NADH oxidation
Monitored at 340 nm for NADH consumption
Advantages: Continuous monitoring, readily available reagents
Limitations: Indirect measurement, potential for interference
Direct Product Formation Assay:
HPLC or LC-MS analysis of UDP-N-acetylglucosamine-enolpyruvate formation
Allows direct quantification of product formation
Advantages: Direct measurement, high specificity
Limitations: Equipment-intensive, discontinuous measurement
Radioactive Assay:
Uses 14C or 3H-labeled PEP to track transfer to UNAG
Advantages: High sensitivity, direct measurement of transfer
Limitations: Requires radioisotope handling facilities
Fluorescence-Based Assays:
Utilizes fluorescently-labeled substrate analogs
Advantages: High sensitivity, potential for high-throughput screening
Limitations: May alter enzyme kinetics, requires specialized substrates
For kinetic characterization, researchers should determine Km values for both PEP and UNAG substrates, as well as the catalytic efficiency (kcat/Km) under various pH and temperature conditions relevant to L. plantarum's natural environment.
Based on structural studies of MurA from related species, L. plantarum murA1 likely possesses the following key structural features:
Domain Organization:
Two globular domains connected by a flexible hinge region
Substrate binding occurs in the cleft between domains
Conformational change from "open" to "closed" state upon substrate binding
Active Site Architecture:
Loop Regions:
Dynamic loops that participate in substrate recognition and binding
Contribute to specificity for UNAG over other UDP-sugars
Regulatory Features:
Understanding these structural features is essential for rational design of inhibitors or engineering variant enzymes with modified properties for research applications.
L. plantarum has evolved sophisticated regulatory mechanisms for murA1 expression under various stress conditions:
During cold and acid stress, L. plantarum NMGL2 shows upregulation of murA1 along with other peptidoglycan synthesis genes (MurB, MurC, MurD, MurE1, and MurF) to increase total peptidoglycan production . This adaptation helps strengthen the cell wall, providing enhanced protection against environmental challenges. The upregulation of murA1 is part of a coordinated response involving multiple stress-response pathways, including proteins associated with cell repair systems and membrane modification.
The expression of murA1 in L. plantarum is regulated through multiple mechanisms:
Transcriptional Regulation:
Stress-responsive promoter elements that respond to environmental signals
Potential binding sites for global regulators that coordinate stress responses
Post-Translational Regulation:
Metabolic Regulation:
Availability of substrates (PEP and UNAG) influences activity
Energy status of the cell affects resource allocation to cell wall synthesis
Integration with Stress Response Pathways:
Understanding these regulatory elements provides potential targets for manipulating murA1 expression in recombinant systems for research applications.
Recombinant L. plantarum strains can elicit specific immune responses influenced by murA1 expression:
Mucosal Immunity:
Cellular Immunity:
Cytokine Modulation:
Innate Immunity Activation:
These immunomodulatory effects suggest recombinant L. plantarum expressing murA1 may have applications in vaccine development and as therapeutic agents for inflammatory conditions.
Recombinant L. plantarum expressing murA1, either alone or in combination with other antigens, shows promise for vaccine development through several mechanisms:
Expression System Design:
Immune Response Induction:
Advantages Over Conventional Vaccines:
Experimental Validation:
The design of such vaccines requires careful optimization of antigen expression levels, selection of appropriate promoters, and validation of stability during gastric transit.
Investigating the covalent reaction mechanism of murA1 requires sophisticated methodological approaches:
X-ray Crystallography:
Mass Spectrometry:
Identification of covalent adducts between the active site cysteine and PEP
Peptide mapping to confirm modification sites
Time-resolved analysis to track reaction intermediates
Techniques: MALDI-TOF, LC-MS/MS with electron transfer dissociation (ETD)
NMR Spectroscopy:
Real-time monitoring of reaction progression
Structural dynamics information during catalysis
Chemical shift changes indicating covalent bond formation
Requires isotopic labeling (13C, 15N) of the enzyme or substrates
Site-Directed Mutagenesis:
Systematic mutation of active site residues
Creation of catalytically inactive variants for trapping reaction intermediates
Development of cysteine-free variants to prevent covalent adduct formation
Comparative kinetic analysis of wild-type and mutant enzymes
These approaches have revealed that murA1 forms a covalent phospholactoyl adduct with PEP during catalysis, and this covalent reaction serves dual functions: tightening the complex with UNAM for feedback regulation and priming the PEP molecule for reaction with UNAG .
CRISPR-Cas9 technology offers powerful approaches for precise manipulation of murA1 in L. plantarum:
Gene Knockout Studies:
Design of specific sgRNAs targeting murA1
Integration of CRISPR-Cas9 components via transformation of appropriate vectors
Selection of knockout mutants through appropriate markers
Phenotypic characterization of murA1 deficiency on cell wall integrity and stress response
Promoter Modification:
Targeted modification of native murA1 promoter to alter expression levels
Integration of inducible or constitutive promoters to control murA1 expression
Creation of reporter fusions to monitor expression under different conditions
Protocol adaptation: lower transformation temperature (30°C) and extended recovery times for L. plantarum
Site-Specific Mutations:
Introduction of specific mutations in the catalytic site
Creation of variants resistant to specific inhibitors
Engineering of MurA1 with altered substrate specificity
Homology-directed repair with carefully designed donor templates
Regulatory Element Engineering:
Modification of regulatory elements controlling murA1 expression
Creation of stress-responsive expression systems
Implementation of tunable expression systems for detailed functional studies
Utilization of base editing approaches for precise nucleotide modifications
Implementation of CRISPR-Cas9 in L. plantarum requires optimization of several parameters including transformation efficiency, guide RNA design specific to L. plantarum genomic context, and appropriate selection methods for identifying successful transformants.
Inhibition of murA1 has multifaceted implications for L. plantarum:
Cell Viability and Morphology:
Reduced peptidoglycan synthesis leading to compromised cell wall integrity
Potential cell elongation or abnormal morphology due to incomplete cell division
Increased susceptibility to osmotic stress and mechanical damage
Possible induction of cell wall stress response pathways
Stress Response Capabilities:
Probiotic Functions:
Applications in Research:
Controllable inhibition provides a tool for studying cell wall biosynthesis
Potential for creating attenuated strains for vaccine development
Model system for investigating bacterial adaptation to cell wall stress
Platform for screening novel peptidoglycan-targeting antimicrobials
The selective inhibition of murA1 through chemical or genetic approaches provides valuable insights into the relationship between cell wall integrity, stress response, and the probiotic properties of L. plantarum.
Analysis of proteomics data for murA1 expression requires comprehensive methodological approaches:
Data Acquisition Strategies:
Data-independent acquisition (DIA) proteomics for comprehensive protein profiling
Multiple reaction monitoring (MRM) for targeted quantification of murA1 and related proteins
Label-free quantification for broad coverage of the proteome
Tandem mass tag (TMT) labeling for multiplexed comparative analysis
Statistical Analysis Pipeline:
Normalization methods accounting for global protein changes
Statistical significance determination using appropriate tests (t-test, ANOVA)
Multiple testing correction (Benjamini-Hochberg, Bonferroni)
Fold change thresholds (typically >1.5-fold) for biological relevance
Functional Data Integration:
Visualization Approaches:
Heat maps of differentially expressed proteins across conditions
Volcano plots highlighting statistical and biological significance
Protein interaction networks with color-coded expression changes
Time-course visualizations for temporal dynamics of response
This systematic approach has been successfully applied to understand how L. plantarum NMGL2 responds to combinational cold and acid stresses, revealing that murA1 is part of a coordinated stress response involving multiple cellular systems .
Designing robust experiments to evaluate murA1 modifications requires a comprehensive approach:
Strain Construction Strategy:
Generation of murA1 overexpression strains using controlled promoters
Creation of murA1 knockout or knockdown strains using CRISPR-Cas9 or antisense RNA
Development of strains expressing modified murA1 (site-directed mutants)
Inclusion of appropriate control strains (empty vector, wild-type, etc.)
Stress Challenge Parameters:
Acid stress: pH gradients (3.0-5.5) with varied exposure times
Cold stress: Temperature ranges (4-15°C) with monitoring over extended periods
Combined stresses: Factorial design incorporating multiple stressors
Recovery conditions: Standard and stress-relief media
Assessment Metrics:
Viability measurements: Colony forming units, fluorescent viability indicators
Growth kinetics: Lag phase duration, growth rate, maximum cell density
Cell morphology: Phase contrast microscopy, transmission electron microscopy
Membrane integrity: Fluorescent dye exclusion assays
Molecular Analysis:
Proteomics: Global protein expression changes
Transcriptomics: RNA-seq for genome-wide expression patterns
Metabolomics: Changes in key metabolic pathways
Cell wall composition: Peptidoglycan structure and modifications
Statistical Design Considerations:
Minimum of biological triplicates for all experiments
Power analysis to determine appropriate sample size
Blocking factors to account for batch effects
Appropriate statistical tests based on data distribution
This experimental framework allows for comprehensive evaluation of how murA1 modifications affect L. plantarum's ability to withstand environmental challenges, with implications for both basic research and applied probiotic development.
Several cutting-edge technologies show promise for advancing murA1 research:
Cryo-Electron Microscopy:
Near-atomic resolution structures of murA1 in different conformational states
Visualization of murA1 within the context of multiprotein peptidoglycan synthesis complexes
Time-resolved structural changes during catalysis
Advantages: No crystallization required, native-like conditions
Single-Cell Technologies:
Analysis of cell-to-cell variation in murA1 expression
Correlation between murA1 levels and individual cell stress resistance
Real-time monitoring of murA1 activity using fluorescent reporters
Applications: Understanding heterogeneous responses in bacterial populations
Synthetic Biology Approaches:
Creation of murA1 variants with non-canonical amino acids for mechanistic studies
Development of orthogonal translation systems in L. plantarum
Design of synthetic regulatory circuits controlling murA1 expression
Potential: Precise control over murA1 function and regulation
Advanced Computational Methods:
Molecular dynamics simulations of murA1-substrate interactions
Machine learning algorithms to predict murA1 expression under various conditions
Systems biology models integrating murA1 into whole-cell metabolic networks
Benefits: In silico prediction of experimental outcomes, hypothesis generation
These technologies promise to provide unprecedented insights into murA1 function and regulation, potentially leading to novel applications in biotechnology and medicine.
Despite significant advances, several critical questions remain unanswered:
Mechanistic Understanding:
How does murA1 expression level correlate with specific immunomodulatory effects?
What is the precise mechanism by which recombinant L. plantarum murA1 influences cytokine production?
Are there strain-specific differences in immunomodulatory effects related to murA1 variants?
How do post-translational modifications of murA1 affect immunogenic properties?
Host-Microbe Interactions:
Does murA1 directly interact with host pattern recognition receptors?
How does the host microbiome influence the immunomodulatory effects of recombinant L. plantarum murA1?
Are there host genetic factors that influence response to L. plantarum murA1?
What is the durability of immune responses elicited by recombinant L. plantarum murA1?
Clinical Applications:
Can recombinant L. plantarum murA1 be effective in treating specific inflammatory conditions?
What dosing regimens optimize immunomodulatory effects while minimizing potential adverse reactions?
How does recombinant L. plantarum murA1 interact with conventional immunomodulatory therapies?
What biomarkers can predict individual response to recombinant L. plantarum murA1 treatment?
Technical Challenges:
How can expression stability be maintained in vivo over extended periods?
What delivery systems optimize the presentation of recombinant murA1 to the immune system?
How can murA1 expression be targeted to specific gastrointestinal regions?
What quality control metrics ensure consistent immunomodulatory properties?