Gloeobacter violaceus is a cyanobacterium notable for conducting photosynthesis in its plasma membrane, unlike most other cyanobacteria that use thylakoid membranes . UDP-N-acetylglucosamine--N-acetylmuramyl-(pentapeptide) pyrophosphoryl-undecaprenol N-acetylglucosamine transferase, commonly known as MurG, is an essential enzyme involved in peptidoglycan biosynthesis . Peptidoglycan is crucial for maintaining the integrity and shape of bacterial cell walls in both Gram-positive and Gram-negative bacteria .
MurG is a cytoplasmic enzyme that participates in the peptidoglycan synthesis pathway . It catalyzes the transfer of N-acetylglucosamine (GlcNAc) from UDP-N-acetylglucosamine to N-acetylmuramyl-(pentapeptide) pyrophosphoryl-undecaprenol (MurNAc-PP-Und), forming GlcNAc-MurNAc-PP-Und . This step is vital for constructing the peptidoglycan layer, which provides structural support to the bacterial cell wall .
Gloeobacter violaceus is phylogenetically distant from other cyanobacteria and possesses unique genomic features . Its genome is a single circular chromosome of 4,659,019 base pairs with a GC content of 62% . The absence of thylakoid membranes and the presence of photosynthetic complexes in the plasma membrane distinguish it from other cyanobacteria .
UDP-N-acetylglucosamine enolpyruvyl transferase (MurA) is another key enzyme in the peptidoglycan pathway . MurA catalyzes the transfer of enolpyruvate from phosphoenolpyruvate (PEP) to UDP-N-acetylglucosamine (UNAG), forming UDP-N-acetylglucosamine enolpyruvate (EP-UNAG) . Fosfomycin, an antibiotic, inhibits MurA by covalently binding to a cysteine residue in its active site, thus blocking peptidoglycan biosynthesis .
Gloeobacter violaceus lacks NADPH-dependent thioredoxin reductase (NTR) and ferredoxin-dependent thioredoxin reductase (FTR), but contains a thioredoxin reductase flavoenzyme (FFTR) . FFTR uses ferredoxin (Fdx) as an electron donor and functionally substitutes for FTR in light-linked enzyme regulation .
Gloeobacter violaceus retains ancestral properties of carotenoid biosynthesis . It utilizes a bacterial-type phytoene desaturase (CrtI) to produce lycopene and CrtW to catalyze echinenone synthesis .
Gloeobacter violaceus Ribulose-1,5-bisphosphate carboxylase/oxygenase (RuBisCO) has been expressed and analyzed for its kinetic properties . The RuBisCO large subunit (RbcL) and small subunit (RbcS) accumulate in the insoluble fraction . The activity of RbcL increases with RuBP concentration, and complementation with RbcS in the presence of RbcX enhances the activity of the protein .
| Protein | Occurrence in Membrane Fractions |
|---|---|
| NADH dehydrogenase | Orange fraction |
| PrtA protein | Orange fraction |
| Phycobilisome core membrane linker protein apcE | Both fractions |
| Chaperone GroEL | Both fractions |
KEGG: gvi:gll1671
STRING: 251221.gll1671
Gloeobacter violaceus represents the earliest diverging lineage of known cyanobacteria, having branched from other cyanobacteria approximately 2.7 billion years ago . This organism lacks thylakoid membranes, which are present in all other known cyanobacteria, indicating it has retained primitive or ancestral traits that provide unique insights into early photosynthetic machinery evolution . As a basal member of the cyanobacterial lineage (confirmed through analysis of small and large subunit rDNA and 137 protein sequences), G. violaceus serves as an invaluable window into early evolutionary adaptations preceding the Great Oxygenation Event . To study G. violaceus and its proteins like murG, researchers should employ phylogenetic analyses incorporating multiple genetic markers and protein sequences to establish evolutionary context. Metagenomic analyses of diverse environmental samples can help identify related lineages and provide broader evolutionary context for functional studies of G. violaceus proteins.
UDP-N-acetylglucosamine--N-acetylmuramyl-(pentapeptide) pyrophosphoryl-undecaprenol N-acetylglucosamine transferase (murG) catalyzes a critical step in peptidoglycan biosynthesis by transferring N-acetylglucosamine (GlcNAc) from UDP-GlcNAc to lipid-linked N-acetylmuramyl-(pentapeptide) (Lipid I), forming Lipid II. Unlike many glycosyltransferases that function within membrane compartments, murG operates at the cytoplasmic face of the plasma membrane in G. violaceus, as this organism lacks internal membrane systems like thylakoids found in other cyanobacteria . This location is significant because in other cyanobacteria, some components of cell wall synthesis may associate with thylakoid membranes. For experimental characterization, researchers should employ radiometric assays using 14C-labeled UDP-GlcNAc to monitor substrate conversion to Lipid II, accompanied by thin-layer chromatography for product verification. Kinetic parameters should be determined using varied substrate concentrations and analyzed via Michaelis-Menten or Lineweaver-Burk plots to establish Km and Vmax values.
Based on successful heterologous expression of other G. violaceus proteins like gloeobacter rhodopsin in E. coli , a similar approach is recommended for murG. The optimal expression system utilizes E. coli BL21(DE3) with pET-based vectors containing a 6x His-tag for purification purposes . The expression protocol should include: (1) Transformation of the construct into E. coli BL21(DE3); (2) Culture growth at 37°C until OD600 reaches 0.6-0.8; (3) Induction with 0.5-1 mM IPTG; (4) Temperature reduction to 18-20°C for overnight expression to enhance protein folding; (5) Cell harvesting by centrifugation at 5,000 × g for 15 minutes. For membrane-associated proteins like murG, inclusion of 1% glycerol in the growth medium and expression at lower temperatures (16-18°C) can significantly improve protein solubility. Alternative systems to consider include cell-free expression systems for proteins that may be toxic to host cells, or expression in Lactococcus lactis for membrane proteins that are difficult to express in E. coli.
Purification of recombinant G. violaceus murG requires a carefully designed strategy that preserves enzyme functionality while achieving high purity. The recommended protocol involves:
Cell lysis using a French press (15,000 psi) or sonication in buffer containing 50 mM Tris-HCl (pH 8.0), 300 mM NaCl, 10% glycerol, and protease inhibitors
Membrane fraction isolation by ultracentrifugation (100,000 × g, 1 hour)
Solubilization with mild detergents such as 0.02% n-dodecyl-β-D-maltopyranoside (DDM)
IMAC purification using Ni-NTA resin with a stepwise imidazole gradient (20-300 mM)
Size exclusion chromatography using Superdex 200 column for final polishing
This multi-step approach has proven effective for other G. violaceus membrane-associated proteins . Enzyme activity should be monitored throughout purification using a radiometric assay with UDP-[14C]GlcNAc. The purification table should record protein concentration, specific activity, total activity, purification fold, and yield for each step. Critical parameters include maintaining 10% glycerol and 0.02% DDM in all buffers to prevent protein aggregation and activity loss. For long-term storage, the purified enzyme should be flash-frozen in liquid nitrogen and stored at -80°C in the presence of 20% glycerol.
Crystallization of membrane-associated enzymes like murG presents significant challenges. The recommended approach involves:
Protein concentration optimization (typically 5-15 mg/ml) in buffer containing 20 mM Tris-HCl pH 7.5, 150 mM NaCl, and 0.02% DDM
Screening using commercial sparse matrix kits (Hampton Research, Molecular Dimensions)
Optimization of hit conditions by varying:
Precipitant concentration (PEG 400-8000, 10-30%)
pH range (6.0-8.5)
Salt concentration (50-500 mM)
Additives (glycerol, small amphiphiles)
Temperature (4°C and 18°C)
Co-crystallization with substrate analogs or product mimics can stabilize the enzyme and improve crystal quality. Lipidic cubic phase (LCP) crystallization represents an alternative approach particularly suitable for membrane-associated proteins. Diffraction quality assessment should be performed at synchrotron radiation sources, with cryo-protection using 20-30% glycerol or PEG 400. For proteins resistant to crystallization, cryo-electron microscopy provides an alternative structural determination method, requiring protein at 1-3 mg/ml in detergent micelles or reconstituted into nanodiscs.
Comprehensive substrate specificity analysis of G. violaceus murG requires multiple complementary approaches:
Radiometric assays using structurally diverse UDP-GlcNAc analogs and Lipid I variants
High-performance liquid chromatography (HPLC) for product identification and quantification
Mass spectrometry to confirm product identity and structural details
Isothermal titration calorimetry (ITC) to determine binding constants
The experimental design should include a systematic panel of substrate analogs with modifications at various positions. Kinetic parameters (Km, kcat, kcat/Km) should be determined for each substrate to construct a structure-activity relationship. Substrate docking studies using homology models can guide the design of analogs. Competition assays with the natural substrate can provide additional insights into binding modes. Results should be presented as a comprehensive table comparing kinetic parameters across all substrates tested, accompanied by structural analyses explaining observed specificity patterns.
The ancestral position of G. violaceus in cyanobacterial evolution (divergence ~2.7 billion years ago) provides a unique opportunity to study the evolution of cell wall biosynthesis. A comparative approach should:
Express and purify murG from G. violaceus and representative cyanobacteria from different evolutionary lineages
Compare enzyme kinetics, substrate specificity, and inhibitor sensitivity
Analyze structural differences using X-ray crystallography or homology modeling
Assess membrane association patterns given G. violaceus' lack of thylakoids
The absence of thylakoid membranes in G. violaceus likely influences murG localization and potentially its functional properties. Research indicates that in thylakoid-containing cyanobacteria, some membrane proteins utilize specialized trafficking pathways absent in G. violaceus . Experimental approaches should include fluorescence microscopy with GFP-tagged murG to visualize cellular localization, and membrane fractionation studies to quantify distribution patterns. Enzyme assays performed at different pH values, temperatures, and salt concentrations can reveal adaptations to primitive environmental conditions. Results should be presented as comparative tables of kinetic parameters, accompanied by phylogenetic trees correlating functional properties with evolutionary relationships.
Investigating membrane interactions of G. violaceus murG requires multiple biophysical techniques:
Tryptophan fluorescence spectroscopy to monitor conformational changes upon membrane binding
Surface plasmon resonance (SPR) with immobilized lipid bilayers to determine binding kinetics
Atomic force microscopy (AFM) to visualize protein-membrane interactions
Hydrogen-deuterium exchange mass spectrometry (HDX-MS) to identify membrane-interacting regions
The experimental design should compare murG interaction with various membrane models, including phospholipid vesicles, nanodiscs, and lipid monolayers. The unique membrane composition of G. violaceus, which lacks thylakoids , suggests potentially distinct lipid interactions compared to other cyanobacteria. Fluorescence resonance energy transfer (FRET) experiments between labeled murG and membrane probes can provide spatial information about protein orientation relative to the membrane. Results should be presented as binding constants, dissociation rates, and structural models depicting the membrane interaction interface, with particular attention to how the primitive evolutionary state of G. violaceus might influence these interactions.
G. violaceus murG represents an excellent model for studying peptidoglycan biosynthesis inhibitors due to its ancestral nature. A comprehensive inhibitor screening workflow should include:
High-throughput fluorescence-based activity assays using BODIPY-labeled UDP-GlcNAc
IC50 determination for hit compounds
Mechanism of inhibition studies (competitive, non-competitive, uncompetitive)
Structure-activity relationship (SAR) development through analog testing
Thermal shift assays (differential scanning fluorimetry) can rapidly identify compounds that bind to and stabilize murG. Isothermal titration calorimetry provides detailed thermodynamic parameters of inhibitor binding. Computer-aided drug design approaches, including virtual screening and molecular dynamics simulations, can identify novel inhibitor scaffolds. The ancestral nature of G. violaceus murG may reveal inhibitor sensitivities that evolved in later cyanobacterial lineages. Results should be presented as inhibition constant (Ki) values, accompanied by Lineweaver-Burk plots illustrating inhibition mechanisms and molecular docking models depicting predicted binding modes.
G. violaceus uniquely lacks thylakoid membranes , which fundamentally alters the cellular organization of membrane-associated processes. This distinctive trait impacts murG in several ways:
Subcellular localization is restricted to the plasma membrane, unlike other cyanobacteria where proteins may distribute between plasma and thylakoid membranes
Membrane protein biogenesis pathways differ from those in thylakoid-containing cyanobacteria
Lipid environment composition may have unique characteristics that influence enzyme activity
Research approaches should include immunogold electron microscopy to precisely localize murG within G. violaceus cells. Membrane fractionation studies comparing G. violaceus with thylakoid-containing cyanobacteria can reveal distribution differences. Lipidomic analysis using mass spectrometry can identify specific lipid components that may influence murG activity. Reconstitution experiments with defined lipid compositions can determine optimal membrane environments for enzyme function. The absence of specialized membrane protein trafficking systems found in thylakoid-containing organisms may result in distinctive post-translational modifications or interaction partners for murG that can be identified through proteomic approaches.
Directed evolution offers powerful tools to enhance properties of G. violaceus murG for research and biotechnological applications:
Error-prone PCR to generate mutation libraries with varying mutation frequencies
DNA shuffling with murG homologs from diverse bacterial species
Site-saturation mutagenesis targeting active site residues
Compartmentalized self-replication (CSR) for in vitro selection
The selection strategy should employ a high-throughput fluorescence-based assay or a growth complementation system in a murG-deficient E. coli strain. Iterative rounds of mutagenesis and selection can progressively improve desired properties such as expression level, stability, catalytic efficiency, or substrate scope. Beneficial mutations should be characterized by detailed kinetic analysis and structural studies to understand their mechanistic basis. This approach can generate variants with enhanced stability for crystallization studies or improved catalytic properties for chemoenzymatic synthesis applications. Results should be presented as comprehensive mutation tables correlating sequence changes with functional improvements, accompanied by structural analyses explaining the molecular basis of enhanced properties.
Recent advances in cryo-electron microscopy (cryo-EM) provide unprecedented opportunities to study membrane-associated enzymes like murG in near-native environments:
Single-particle cryo-EM of detergent-solubilized murG for high-resolution structure determination
Cryo-electron tomography (cryo-ET) of murG reconstituted into liposomes or nanodiscs
Subtomogram averaging to reveal organizational patterns in membrane contexts
Correlative light and electron microscopy (CLEM) to study murG localization in G. violaceus cells
Sample preparation should focus on reconstituting purified murG into membrane mimetics that closely resemble its native environment. For single-particle analysis, protein concentration should be optimized at 1-3 mg/ml, with particle distribution assessed by negative staining prior to cryo-grid preparation. Data collection parameters include 300 kV acceleration voltage, 0.5-1.5 μm defocus range, and ≤30 e−/Å2 total electron dose. Image processing workflow utilizing motion correction, CTF estimation, particle picking, 2D classification, 3D reconstruction, and refinement can achieve resolutions sufficient to resolve side chain details (3-4 Å). Time-resolved cryo-EM using microfluidic mixing devices can potentially capture distinct catalytic states, providing unprecedented insights into the reaction mechanism.
As a representative of the earliest branching cyanobacterial lineage , G. violaceus murG provides a window into early evolutionary adaptations. Future research should explore:
Ancestral sequence reconstruction to infer properties of proto-murG enzymes
Comparative genomics across diverse cyanobacterial lineages to track evolutionary changes
Physiological studies under conditions mimicking ancient Earth (high CO2, low O2, variable salinity)
Integration with geochemical data to correlate enzyme properties with evolutionary transitions
The branching of G. violaceus from other cyanobacteria occurred approximately 2.7 billion years ago , predating or coinciding with the Great Oxygenation Event. Experiments should examine murG function across temperature, pH, and salinity gradients to understand environmental adaptations. Molecular clock analyses can correlate functional changes with geological time periods. Synthetic biology approaches reconstructing ancient metabolic pathways incorporating ancestral murG variants can provide insights into evolutionary constraints. Research should particularly focus on how cell wall biosynthesis adapted during the transition from ancient low-oxygen to oxygen-rich environments, with results presented as evolutionary trajectory models correlating structural and functional changes with geological timelines.
Advanced computational methods offer powerful tools for understanding murG at atomic resolution:
AlphaFold2 or RoseTTAFold for accurate protein structure prediction
Molecular dynamics simulations (100+ ns) to study conformational dynamics
Quantum mechanics/molecular mechanics (QM/MM) calculations to elucidate reaction mechanisms
Machine learning models to predict substrate specificity or inhibitor sensitivity
Simulations should be performed in explicit membrane environments mimicking G. violaceus plasma membrane composition. Enhanced sampling techniques like metadynamics or umbrella sampling can characterize free energy landscapes governing substrate binding and product release. Coarse-grained simulations extending to microsecond timescales can capture large-scale conformational changes and membrane interactions. Computational mutagenesis studies can predict the impact of specific residue changes on enzyme function, guiding experimental site-directed mutagenesis efforts. Molecular docking combined with binding free energy calculations can screen virtual libraries for potential inhibitors. Results should be presented as energy landscapes, reaction coordinate diagrams, and conformational ensembles, with experimental validation of key computational predictions.
Achieving sufficient quantities of properly folded G. violaceus murG for comprehensive structural and functional studies presents several challenges:
| Challenge | Solution | Expected Improvement |
|---|---|---|
| Low expression yield | Codon optimization for E. coli | 2-5 fold increased expression |
| Auto-induction media | Consistent high-density cultures | |
| Fusion partners (MBP, SUMO) | Enhanced solubility | |
| Inclusion body formation | Low temperature expression (16°C) | Reduced aggregation |
| Co-expression with chaperones | Improved folding | |
| Refolding from inclusion bodies | Recovery of active protein | |
| Membrane association | Optimized detergent screening | Better extraction efficiency |
| Amphipol stabilization | Enhanced stability | |
| Nanodisc reconstitution | Native-like environment | |
| Purification challenges | Tandem affinity tags | Higher purity |
| On-column refolding | Improved recovery | |
| Automated chromatography | Consistent preparation |
Implementation of these strategies typically yields 1-5 mg of pure, active enzyme per liter of bacterial culture. For structural studies requiring 10+ mg of protein, fermenter-based production (10+ liter scale) with optimized parameters provides a scalable approach. Insect cell or mammalian expression systems offer alternatives for proteins that remain challenging in bacterial systems. Economic analyses indicate that optimized E. coli expression remains the most cost-effective approach, with production costs approximately 10-fold lower than eukaryotic expression systems.