S-adenosylmethionine (AdoMet) is a crucial metabolite involved in various metabolic processes . Methionine adenosyltransferase (MAT), also known as AdoMet synthetase or MetK, is the enzyme responsible for producing AdoMet from L-methionine and ATP . Given its role, MetK is a target in the development of anti-cancer and antimicrobial agents .
Chromobacterium violaceum is a bacterium known for producing violacein, a purple pigment with antimicrobial, antiviral, and anticancer properties . C. violaceum is adaptable, but the mechanisms of stress resistance are not fully understood . The metK gene from C. violaceum can be expressed in Escherichia coli, allowing for detailed studies of its function .
The gene encoding phenylalanine hydroxylase from Chromobacterium violaceum has been cloned and expressed in Escherichia coli . Similarly, the metK gene can be isolated and expressed in E. coli to produce recombinant MetK . This involves subcloning the DNA fragment containing the metK gene into a plasmid vector, such as pBluescript II or pMAC 5-8, and inducing its expression using isopropyl-beta-D-thiogalactopyranoside (IPTG) .
Recombinant MetK exhibits similar physical and chemical characteristics to the wild-type enzyme . For phenylalanine hydroxylase, the copper content and Michaelis-Menten parameters of the recombinant protein are comparable to those of the wild-type enzyme .
Violacein biosynthesis involves multiple enzymes, including VioD and VioC, which are flavin-dependent oxygenases . VioD hydroxylates one indole ring to produce protoviolaceinic acid, while VioC hydroxylates the second indole ring, leading to violacein formation .
MetK can interact with various RNA species, including small non-coding RNAs (sRNAs) . Studies have shown that MetK from Sinorhizobium meliloti binds to sRNAs that regulate nutrient uptake and cell cycle mRNAs . In vitro assays confirmed that MetK binds to AbcR2, NfeR1, and EcpR1 sRNAs with varying affinities . This suggests that MetK may have a role in RNA metabolism beyond its function as a synthetase .
Chromobacterium violaceum has mechanisms to resist stress, including a cytochrome bd (CioAB) that protects against multiple stressors . A cioAB mutant strain was sensitive to iron, streptonigrin, zinc, hydrogen peroxide, nitric oxide, sulfide, and cyanide . Expression analysis showed that the cioRAB operon and the cioAB genes were increased in a ΔcioR mutant, indicating that CioR regulates the cio operon .
Sublethal levels of antibiotics like hygromycin A can induce violacein production in C. violaceum . A two-component regulatory complex, the air system, participates in this process . The air system modulates violacein production independently of VioS .
KEGG: cvi:CV_0963
STRING: 243365.CV_0963
S-adenosylmethionine synthase (metK) in Chromobacterium violaceum catalyzes the formation of S-adenosylmethionine (AdoMet or SAM) from methionine and ATP. This enzyme (EC 2.5.1.6) is also known as methionine adenosyltransferase (MAT) . The reaction produces S-adenosylmethionine, which serves as the principal biological methyl donor in numerous cellular processes. In C. violaceum specifically, SAM plays a crucial role in quorum sensing pathways where it contributes to the conversion of fatty acids to AHLs (acyl-homoserine lactones) via the CviI synthase enzyme . This process ultimately influences the expression of the vioABCDE operon that governs violacein production, the characteristic purple pigment of this bacterium.
The significance of metK extends beyond basic metabolism, as methyl donation reactions are essential for numerous cellular processes including DNA methylation, protein modification, and various biosynthetic pathways that contribute to bacterial adaptation and virulence.
The recombinant C. violaceum S-adenosylmethionine synthase can be successfully expressed in yeast expression systems, as indicated by the product information . When designing expression experiments, researchers should consider the following methodological approaches:
Expression System Selection:
Optimization Parameters:
Induction conditions: For IPTG-inducible systems, concentrations between 0.1-1.0 mM IPTG at OD600 of 0.6-0.8
Temperature: Lower post-induction temperatures (16-25°C) often improve solubility
Duration: 4-18 hours depending on temperature and expression system
Fusion Tags:
For optimal results, pilot expressions should be conducted to determine the best combination of these parameters for your specific experimental context. Expression levels should be monitored via SDS-PAGE and Western blotting using antibodies against the target protein or fusion tag.
Purification of recombinant C. violaceum S-adenosylmethionine synthase should follow a multi-step approach to achieve high purity (>85% as indicated in product specifications) :
| Step | Method | Buffer Composition | Expected Result |
|---|---|---|---|
| 1 | Cell lysis | 50 mM Tris-HCl pH 8.0, 300 mM NaCl, 10 mM imidazole, 1 mM PMSF, protease inhibitors | Crude extract containing soluble metK |
| 2 | Affinity chromatography | Same as lysis buffer with gradient to 250 mM imidazole | ~70-80% purity |
| 3 | Size exclusion chromatography | 20 mM Tris-HCl pH 7.5, 150 mM NaCl, 1 mM DTT | >85% purity |
| 4 | Ion exchange (optional) | 20 mM Tris-HCl pH 8.0, gradient 50-500 mM NaCl | >95% purity |
During purification, researchers should:
Monitor enzyme activity at each step using a spectrophotometric assay measuring SAM formation
Verify protein identity through mass spectrometry or Western blotting
Assess purity by SDS-PAGE and determine final concentration using Bradford or BCA assays
After purification, the protein should be stored properly to maintain activity. As indicated in the product information, avoiding repeated freeze-thaw cycles is critical, and working aliquots can be stored at 4°C for up to one week . For long-term storage, adding glycerol to a final concentration of 50% and storing at -20°C/-80°C is recommended, with an expected shelf life of 6 months for liquid form and 12 months for lyophilized form .
The catalytic activity of purified C. violaceum S-adenosylmethionine synthase can be evaluated through several complementary approaches:
Spectrophotometric Coupled Assays:
The formation of SAM can be coupled to reactions that produce measurable spectroscopic changes
For example, coupling to 5'-methylthioadenosine nucleosidase and adenine deaminase reactions allows monitoring at 265 nm
Calculate enzyme activity in μmol of SAM produced per minute per mg protein
HPLC-based Product Quantification:
Separate reaction components (methionine, ATP, SAM, and Pi) using reverse-phase HPLC
Monitor SAM formation at 254 nm using appropriate standards
This method provides direct quantification without interference from coupled reactions
Isothermal Titration Calorimetry:
Measure heat released during the enzymatic reaction
Determine thermodynamic parameters (ΔH, ΔS, ΔG) and binding constants
Particularly useful for comparing wild-type and mutant variants
When designing activity assays, researchers should consider:
The optimal reaction conditions (buffer composition, pH 7.5-8.0, Mg²⁺ concentration)
Temperature dependency (typically assayed at 37°C)
Substrate concentrations (methionine and ATP)
The presence of potential inhibitors or activators
Control experiments should include heat-inactivated enzyme and reactions without key substrates to establish baseline activity levels.
While the search results don't provide specific structural information for C. violaceum metK, comparative analysis with homologous enzymes suggests several distinguishing features:
The full sequence information for C. violaceum metK provided in the product datasheet can be analyzed for unique structural elements through:
Sequence Alignment Analysis:
Align C. violaceum metK with homologs from other bacteria, particularly pathogenic species
Identify conserved catalytic residues and C. violaceum-specific substitutions
Secondary structure prediction tools can identify potential unique structural elements
Homology Modeling:
Generate 3D structural models based on crystal structures of homologous proteins
Evaluate the geometry of active sites and potential allosteric sites
Analyze surface electrostatics and potential protein-protein interaction interfaces
Experimental Structure Determination:
X-ray crystallography or cryo-EM studies to determine the actual structure
Conduct ligand binding studies using differential scanning fluorimetry or surface plasmon resonance
The sequence provided in the product information contains regions predicted to be involved in methionine binding (GFDFRGC) and ATP binding (GLDLNQGAG), which may have subtle variations from homologs that influence substrate specificity or catalytic efficiency. These variations could potentially be exploited for species-specific inhibitor design.
S-adenosylmethionine synthase (metK) contributes significantly to C. violaceum pathogenicity through several interconnected mechanisms:
Quorum Sensing Regulation:
SAM produced by metK serves as a precursor for acyl-homoserine lactone (AHL) synthesis
The CviI synthase enzyme utilizes SAM for converting fatty acids to AHLs
AHLs form complexes with CviR that stimulate the vioABCDE operon, regulating violacein production
This quorum sensing system coordinates population-dependent expression of virulence factors
Methylation of Virulence Factors:
SAM-dependent methyltransferases modify numerous cellular components
Methylation of cell surface components can affect host recognition and immune evasion
DNA methylation may regulate expression of virulence genes
Connection to Iron Acquisition Systems:
C. violaceum infections cause severe complications including bacterial hemophagocytic syndrome, brain abscess, chronic cellulitis, conjunctivitis, chronic granulomatosis, and other life-threatening conditions . The high lethality rate despite infrequent pathogenicity in humans may be partly attributed to the regulatory roles of metK in virulence factor production.
While the search results don't directly address the relationship between metK and siderophore production, we can infer potential connections based on known biochemical pathways:
Biosynthetic Connection:
Regulatory Interaction:
Experimental Investigation:
To investigate this relationship, researchers could:
Generate metK knockdown strains and measure siderophore production using the chrome azurol S (CAS) plate assay
Analyze siderophore halos that develop after incubation for 24 hours at 37°C using Image J software for quantification
Combine mutations in metK with mutations in cbaF or vbaF to understand the specific siderophore pathways affected
The regulation of siderophore production in C. violaceum appears to be complex, involving multiple systems including the ChuPRSTUV operon that encodes a heme uptake system . Further research is needed to clarify the specific role of metK in this regulatory network.
Recombinant S-adenosylmethionine synthase (metK) from C. violaceum offers several strategic applications in drug discovery:
High-Throughput Inhibitor Screening:
Establish in vitro enzyme assays using purified recombinant metK
Screen chemical libraries for compounds that selectively inhibit C. violaceum metK
Prioritize compounds that show selectivity over human MAT enzymes
Structure-Based Drug Design:
Whole-Cell Testing Platform:
Target Validation Studies:
Create conditional knockdown strains of metK in C. violaceum
Confirm the essentiality of metK for virulence in infection models
Determine whether partial inhibition is sufficient to attenuate pathogenicity
Combination Therapy Approaches:
Test metK inhibitors in combination with:
Iron chelators (to enhance the effect of siderophore disruption)
Quorum sensing inhibitors (to synergistically target virulence pathways)
Conventional antibiotics (to potentially restore sensitivity)
The development of drugs targeting metK is particularly promising given C. violaceum's genome has a broad but incomplete array of ORFs coding for mammalian pathogenicity-associated proteins, which may explain its high lethality rate but infrequent pathogenicity in humans .
Investigating the metabolic interconnections of metK requires multi-faceted experimental approaches:
Systems Biology Analysis:
Transcriptomics: Compare RNA-seq profiles of wild-type and metK-modulated strains under various conditions
Proteomics: Use mass spectrometry to identify differentially expressed proteins and post-translational modifications
Metabolomics: Measure changes in SAM, methionine, and downstream metabolites
Genetic Interaction Studies:
Biochemical Pathway Analysis:
Track isotopically labeled metabolites (¹³C-methionine) to map flux through SAM-dependent pathways
Measure activities of key enzymes in methionine recycling pathways
Characterize the methylome (all methylated substrates) dependent on SAM availability
Computational Modeling:
Develop metabolic flux models incorporating metK-dependent reactions
Predict metabolic bottlenecks and potential compensatory pathways
Identify synthetic lethal interactions that could be exploited therapeutically
| Approach | Key Methods | Expected Outcomes | Limitations |
|---|---|---|---|
| Transcriptomics | RNA-seq, qRT-PCR | Gene regulatory networks | Indirect measurement of activity |
| Proteomics | LC-MS/MS, protein arrays | Protein abundance changes | May miss transient interactions |
| Metabolomics | HPLC-MS, NMR | Metabolite concentration profiles | Complex sample preparation |
| Genetic studies | CRISPR-Cas9, allelic exchange | Phenotypic interactions | Potential pleiotrophic effects |
The regulatory protein ChuP, which connects heme and siderophore utilization in C. violaceum , may serve as an excellent starting point for studying these metabolic interconnections, as it demonstrates how regulatory systems can integrate multiple iron acquisition pathways.
Researchers working with recombinant C. violaceum S-adenosylmethionine synthase frequently encounter several technical challenges:
Protein Solubility Issues:
Challenge: Formation of inclusion bodies during overexpression
Solution: Express at lower temperatures (16-20°C), use solubility-enhancing fusion tags (MBP, SUMO), or optimize buffer conditions with stabilizing additives (glycerol, arginine)
Enzyme Stability:
Substrate Availability:
Challenge: ATP degradation during activity assays
Solution: Prepare fresh ATP solutions, add phosphatase inhibitors, and include an ATP-regenerating system (phosphoenolpyruvate and pyruvate kinase)
Interference in Assays:
Protein Quantification:
Challenge: Inaccurate determination of enzyme concentration
Solution: Use multiple quantification methods (Bradford, BCA, and A280 measurements) and calibrate with appropriate protein standards
For reconstitution of lyophilized protein, researchers should follow the specific protocol: briefly centrifuge the vial prior to opening, reconstitute in deionized sterile water to a concentration of 0.1-1.0 mg/mL, and add glycerol (final concentration 50%) for long-term storage .
Differentiating direct metK effects from secondary metabolic perturbations requires careful experimental design:
Complementation Strategies:
Challenge: Determining if observed phenotypes are directly due to metK alterations
Solution: Create genetic complementation strains restoring wild-type metK function in mutant backgrounds
Approach: Similar to methods used for chuP complementation in research on heme utilization , where complementation fully restored growth in the presence of heme and hemoglobin
Targeted Metabolite Supplementation:
Challenge: Separating methionine cycle disruption from SAM depletion effects
Solution: Supplement growth media with key metabolites (methionine, SAM, spermidine) to bypass specific blocks
Analysis: Compare phenotypic rescue patterns to map the primary metabolic block
Time-Course Analysis:
Challenge: Distinguishing primary from secondary effects
Solution: Monitor transcriptomic, proteomic, and metabolomic changes at multiple time points following metK perturbation
Interpretation: Primary effects typically manifest earlier than downstream consequences
Enzyme Activity Variants:
Challenge: Generating targeted enzyme function alterations
Solution: Create point mutations that affect specific aspects of metK function (e.g., catalytic efficiency, substrate binding, protein interactions)
Analysis: Compare phenotypic profiles of different variants to dissect functional domains
Controlled Expression Systems:
When evaluating experimental results, researchers should implement appropriate statistical analyses and include multiple biological and technical replicates, as demonstrated in studies of the ChuP regulatory protein where experiments were performed in three biological replicates .