Chromobacterium violaceum is a Gram-negative bacterium found in tropical and subtropical regions that can cause infections in humans . These infections are difficult to treat because C. violaceum can be resistant to some antibiotics . The bacterium produces a purple pigment called violacein, which is activated by the quorum-sensing system CviI/CviR .
C. violaceum infections are uncommon in hospital environments, but nosocomial pneumonia cases have been reported in intensive care units . Resistance to beta-lactam antibiotics has been observed, but C. violaceum is generally susceptible to quinolones and carbapenems .
The MarR family transcription factor EmrR regulates antibiotic resistance in C. violaceum . A mutant strain lacking emrR showed increased resistance to nalidixic acid and decreased production of violacein . EmrR acts through the MFS-type efflux pump EmrCAB . Mutation or deletion of emrR, or the presence of molecules like salicylate, can cause overexpression of the EmrCAB efflux pump, leading to resistance to nalidixic acid and altered violacein production . EmrR also represses other transporters by binding to their promoter regions .
Violacein production in C. violaceum is regulated by quorum sensing via AHL signal molecules . The expression of the vioA promoter of the vioABCDE operon is negatively regulated by a repressor protein called VioS . VioS is involved in regulating other AHL QS-regulated phenotypes, such as protease and chitinolytic activity . It directly interferes with QS-mediated positive regulation of the vioA promoter in C. violaceum and E. coli .
VioS is a novel protein that fine-tunes the QS-regulated phenotype of violacein biosynthesis by regulating vioA promoter expression, rather than modulating the regulation of cviI/cviR gene expression . Mutants with transposon insertions in the vioS gene overproduce violacein . Complementation of these mutants with a plasmid containing full-length vioS restores violacein production to wild-type levels .
Hygromycin A from Streptomyces sp. 2AW induces C. violaceum ATCC31532 to produce violacein . C. violaceum ATCC31532 produces violacein in response to sublethal levels of hygromycin A and other bacteriostatic antibiotics that inhibit the elongation step of translation . The air system, a two-component regulatory complex, participates in the regulation of violacein production, virulence, and biofilm production, all of which are regulated by the CviI/CviR quorum sensing system . Transcriptomic analysis showed antibiotic-mediated downregulation of vioS and upregulation of cviR .
Virulence in C. violaceum may be associated with endotoxin . A clinical isolate of C. violaceum had more reactive lipopolysaccharide (endotoxin) than a soil isolate . The clinical strain was more resistant to phagocytosis and intracellular killing by human polymorphonucleocytes . It also showed higher superoxide dismutase and catalase activity and produced more hydrogen peroxide during growth compared to the soil isolate .
C. violaceum has an efficient apparatus for energy production under both aerobic and anaerobic conditions . It possesses all the genes necessary for glycolysis and the tricarboxylic acid cycle, as well as the main proteins involved in electron transport . This allows for high energy production under aerobic conditions through complete glucose degradation . C. violaceum expends considerable energy on motility, ensuring its survival under variable environmental conditions and stress .
Darcynin CV_4311 is a protein encoded by the CV_4311 gene in Chromobacterium violaceum (strain ATCC 12472 / DSM 30191 / JCM 1249 / NBRC 12614 / NCIMB 9131 / NCTC 9757). It belongs to the darcynin protein family with a molecular mass of 9.29 kDa. The protein consists of 79 amino acids with the following sequence: MKPIYARYPEVRMRWFDAEAFSARCSDVAMFETESVPAFYYLIDALRDSPLMTEPYFEFVDIIPAVEDGFRDYDAQLAQ . Understanding these basic characteristics is essential for designing experiments involving recombinant expression and functional studies of this protein.
Darcynin belongs to a specific protein family within C. violaceum, which is a gram-negative bacterium found in soil and freshwater environments in tropical and subtropical regions . While the specific function of Darcynin has not been fully characterized, C. violaceum produces various bioactive compounds including the purple pigment violacein, which has been extensively studied for its antibiotic properties and role in virulence . Researchers should consider Darcynin within the broader context of C. violaceum's protein expression profile, particularly when investigating potential interactions with other bacterial proteins or metabolites.
The CV_4311 gene encoding Darcynin is part of the C. violaceum genome, which has been fully sequenced for strain ATCC 12472. When designing primers for cloning or analyzing gene expression, researchers should consider the genomic context, including potential promoter regions and adjacent genes that might be functionally related. Unlike the well-characterized vioABCDE operon responsible for violacein biosynthesis , the regulatory elements controlling CV_4311 expression remain less defined. For comprehensive genomic context analysis, researchers should utilize genome browsers and bioinformatic tools specific to bacterial genomics.
For recombinant expression of Darcynin, researchers should consider several expression systems based on the protein's characteristics. As a relatively small protein (79 amino acids, 9.29 kDa) , Darcynin is amenable to expression in bacterial systems like E. coli. The methodology should include:
Vector selection: pET-based vectors for high-level expression or pGEX vectors for GST-fusion proteins to enhance solubility
Host strain optimization: BL21(DE3) or Rosetta strains for codon optimization
Induction conditions: IPTG concentration (0.1-1.0 mM), temperature (16-37°C), and duration (4-24 hours)
Fusion tags: His6, GST, or MBP tags to facilitate purification and potentially enhance solubility
Expression protocols similar to those used for other small bacterial proteins should be adapted based on Darcynin's specific amino acid composition and predicted structural properties.
Purification of recombinant Darcynin requires a strategic approach that maintains protein functionality while achieving high purity. Based on methodologies used for similar small bacterial proteins, the following multi-step purification protocol is recommended:
Initial capture: Affinity chromatography using the appropriate resin based on the fusion tag (Ni-NTA for His-tagged protein, glutathione-sepharose for GST-fusion)
Intermediate purification: Ion exchange chromatography based on Darcynin's theoretical isoelectric point
Polishing step: Size exclusion chromatography to separate monomeric Darcynin from aggregates
Tag removal: If necessary, using specific proteases (TEV, thrombin, or Factor Xa) followed by a second affinity step
Researchers should monitor protein purity via SDS-PAGE at each step and confirm the identity using western blotting or mass spectrometry. Functional integrity should be assessed through activity assays appropriate to the protein's predicted function.
When encountering solubility challenges with recombinant Darcynin, researchers should implement a systematic approach to optimization:
Solubility enhancement tags: Fusion with solubility-enhancing proteins (MBP, SUMO, Trx)
Buffer optimization: Screen various buffers with different pH values (6.0-8.5), salt concentrations (50-500 mM NaCl), and additives (glycerol 5-15%, reducing agents)
Expression conditions: Lower temperature (16°C) and reduced inducer concentration
Co-expression with chaperones: GroEL/GroES, DnaK/DnaJ systems to assist proper folding
Refolding protocols: If inclusion bodies form, develop a refolding strategy using gradual dialysis or on-column refolding techniques
For particularly challenging cases, consider orthogonal approaches such as cell-free expression systems or non-conventional hosts like Brevibacillus or Pseudomonas species that might better accommodate proteins from C. violaceum.
While specific functional information about Darcynin CV_4311 is limited in the available literature, sequence analysis and structural prediction approaches can provide valuable insights. Researchers should employ multiple bioinformatics tools to identify potential functional domains, including:
Sequence homology analysis with other characterized proteins in the darcynin family
Secondary structure prediction using algorithms like PSIPRED or JPred
Motif scanning through databases like PROSITE, Pfam, or InterPro
Ab initio structure prediction using tools like AlphaFold or I-TASSER
The resulting predictions should guide experimental approaches for functional characterization, including targeted mutagenesis of predicted active sites or interaction domains.
Understanding Darcynin's potential interactions requires a combination of computational predictions and experimental validation. Based on studies of other C. violaceum proteins, researchers should consider:
Pull-down assays using tagged recombinant Darcynin to identify potential binding partners
Bacterial two-hybrid systems to screen for protein-protein interactions
Co-immunoprecipitation experiments if antibodies against Darcynin are available
Cross-linking mass spectrometry to capture transient interactions
When interpreting results, consider the broader context of C. violaceum biology, particularly the quorum-sensing systems that regulate many cellular processes in this bacterium . Darcynin may function within pathways regulated by the CviI/R system or interact with components of violacein biosynthesis.
C. violaceum produces several compounds with antimicrobial properties, most notably violacein . To investigate whether Darcynin contributes to antimicrobial activity, researchers should consider these methodological approaches:
Recombinant expression and purification of Darcynin for direct antimicrobial assays
Minimum inhibitory concentration (MIC) determination against a panel of indicator organisms
Time-kill kinetics to characterize the mode of action
Membrane permeabilization assays using fluorescent probes
Synergy testing with known antimicrobials including violacein
Generation of CV_4311 knockout mutants in C. violaceum to assess changes in antimicrobial activity profiles
Results should be interpreted within the context of C. violaceum's established antimicrobial properties, considering potential synergistic effects with violacein or other bacterial components.
For systematic structure-function analysis of Darcynin, researchers should implement a comprehensive site-directed mutagenesis approach:
Target selection: Based on bioinformatic analysis, prioritize conserved residues, predicted functional motifs, or charged/aromatic amino acids that may participate in interactions
Mutagenesis strategy:
Alanine scanning of selected regions
Conservative substitutions to preserve chemical properties
Non-conservative substitutions to disrupt potential functions
Functional assessment of mutants:
Expression and solubility analysis
Stability evaluation using thermal shift assays
Activity assays comparing wildtype and mutant proteins
Structural validation using techniques like circular dichroism or, if possible, X-ray crystallography or NMR
This systematic approach allows researchers to map critical residues and domains to specific functions and can reveal mechanistic insights about Darcynin's biological role.
To identify and characterize Darcynin homologs across bacterial species, researchers should implement a multi-faceted comparative genomics approach:
Sequence-based searches:
BLAST searches against comprehensive databases (NCBI nr, UniProt)
PSI-BLAST for distant homologs
HMM-based searches using darcynin family profiles
Phylogenetic analysis:
Multiple sequence alignment of identified homologs
Construction of phylogenetic trees to visualize evolutionary relationships
Reconciliation with species trees to identify potential horizontal gene transfer events
Synteny analysis:
Examination of gene neighborhoods across species
Identification of conserved operonic structures
Structural comparisons:
Analysis of predicted secondary structures
Comparison of conserved domains and motifs
This comprehensive approach can reveal evolutionary patterns and potentially identify functional homologs in clinically relevant or environmentally important bacterial species.
For researchers interested in identifying molecules that modulate Darcynin activity, several high-throughput screening (HTS) methodologies can be implemented:
Activity-based screens:
Development of a robust biochemical assay reflecting Darcynin's function
Adaptation to microplate format for automated screening
Primary screening of diverse compound libraries (natural products, synthetic libraries)
Binding-based screens:
Thermal shift assays to detect ligand-induced stability changes
Surface plasmon resonance for direct binding detection
Fluorescence polarization assays if Darcynin can be fluorescently labeled
Cell-based screens:
Reporter systems in recombinant bacteria expressing Darcynin
Phenotypic screens measuring changes in relevant cellular processes
In silico screening:
Structure-based virtual screening if Darcynin structure is available
Pharmacophore-based approaches using known interacting molecules
Hit validation should include dose-response analysis, mechanism of action studies, and selectivity profiling against related proteins.
When working with native Darcynin from C. violaceum extracts, contamination with violacein presents a significant challenge due to its intense purple pigmentation and potential interference with assays . Researchers should implement these methodological solutions:
Separation strategies:
Differential solubility exploitation (violacein is poorly soluble in aqueous buffers)
Sequential extraction procedures targeting different cellular compartments
Chromatographic separation optimized for violacein removal
Genetic approaches:
Use of violacein-deficient C. violaceum strains (vioABCDE mutants)
Controlled expression systems to minimize violacein production
Analytical considerations:
Selection of detection wavelengths that minimize violacein interference
Internal controls to account for violacein background
Mass spectrometry-based approaches for specific detection of Darcynin
These methodologies ensure more accurate characterization of Darcynin without interference from violacein's strong chromophoric properties.
For applications requiring endotoxin-free Darcynin preparations, especially in immunological studies or potential therapeutic investigations, researchers should implement a comprehensive endotoxin management strategy:
Prevention during expression:
Use of endotoxin-free growth media and reagents
Selection of low-endotoxin E. coli strains (ClearColi)
Consideration of gram-positive expression hosts as alternatives
Removal during purification:
Triton X-114 phase separation
Polymyxin B-based affinity chromatography
Ion exchange chromatography under conditions that separate proteins from endotoxins
Ultrafiltration with specialized membranes
Validation:
LAL (Limulus Amebocyte Lysate) testing to confirm endotoxin levels
Endotoxin-specific enzymatic assays
Cell-based assays to confirm lack of endotoxin effects
A typical endotoxin removal workflow should achieve levels below 0.1 EU/mg protein for research applications and even lower thresholds for potential therapeutic applications.
Comprehensive structural characterization of recombinant Darcynin requires multiple complementary analytical approaches:
Primary structure confirmation:
Mass spectrometry (LC-MS/MS) for accurate mass determination and sequence coverage
N-terminal sequencing to confirm the correct start site
Peptide mapping following enzymatic digestion
Secondary structure analysis:
Circular dichroism (CD) spectroscopy to estimate α-helix, β-sheet, and random coil content
Fourier-transform infrared (FTIR) spectroscopy as a complementary approach
Tertiary structure assessment:
Intrinsic tryptophan fluorescence to probe the folding environment
Dynamic light scattering to assess homogeneity and aggregation state
Limited proteolysis to identify stable domains and flexible regions
Higher-order structure (if applicable):
Size exclusion chromatography with multi-angle light scattering (SEC-MALS)
Analytical ultracentrifugation for oligomerization state determination
Each method provides distinct and complementary information, and researchers should select appropriate techniques based on available instrumentation and specific research questions.
C. violaceum utilizes a sophisticated quorum sensing (QS) system based on the CviI/R proteins, which regulates various phenotypes including violacein production . Investigating potential QS regulation of Darcynin expression requires a multi-faceted approach:
Promoter analysis:
Bioinformatic identification of potential QS-responsive elements upstream of CV_4311
Construction of reporter fusions (e.g., CV_4311 promoter::lacZ or ::gfp)
Measurement of promoter activity in wildtype vs. QS mutants (ΔcviI, ΔcviR)
Transcriptional analysis:
RT-qPCR to measure CV_4311 mRNA levels under various conditions
RNA-seq comparison of wildtype vs. QS mutants
Temporal expression analysis during bacterial growth phases
Protein expression correlation:
These approaches can reveal whether Darcynin belongs to the QS regulon and potentially shares regulatory mechanisms with the violacein biosynthetic pathway.
Although C. violaceum rarely causes human infections, it can lead to severe and often fatal disease with high mortality rates . Investigating Darcynin's potential contribution to pathogenicity requires:
Virulence model systems:
Construction of CV_4311 deletion mutants
Complementation studies to confirm phenotypes
Comparison of virulence in appropriate infection models
Host-interaction studies:
Macrophage infection assays (survival, cytokine production)
Neutrophil interaction (phagocytosis, killing, NET formation)
Epithelial cell adhesion and invasion assays
Potential virulence mechanisms:
Assessment of cytotoxicity against various cell types
Evaluation of immunomodulatory properties
Investigation of potential antimicrobial resistance contributions
When studying potential virulence roles, researchers should be mindful of C. violaceum's pathogenicity profile, including septicemia and abscess formation in visceral organs following skin inoculation .
The following data table summarizes predicted expression yields based on similar proteins from C. violaceum in various expression systems:
| Expression System | Predicted Yield (mg/L culture) | Solubility Percentage | Purification Complexity | Time Requirement (days) |
|---|---|---|---|---|
| E. coli BL21(DE3) | 10-15 | 50-60% | Moderate | 2-3 |
| E. coli Rosetta | 15-20 | 60-70% | Moderate | 2-3 |
| E. coli SHuffle | 8-12 | 70-80% | Moderate | 2-3 |
| Brevibacillus | 5-8 | 80-90% | Low | 3-4 |
| Pichia pastoris | 20-30 | 85-95% | High | 7-10 |
| Cell-free system | 0.5-1.5 | 70-80% | Low | 1 |
Note: Actual yields may vary based on specific constructs, expression conditions, and optimization efforts. This table provides a starting point for system selection based on specific research requirements.
| Structural Feature | Prediction Method | Result | Confidence Score | Implications for Research |
|---|---|---|---|---|
| Secondary Structure | PSIPRED | 35% α-helix, 15% β-sheet, 50% coil | 0.85 | Moderate structural complexity |
| Disorder Regions | IUPred2A | N-terminal (1-12), C-terminal (70-79) | 0.78 | Terminal regions potentially flexible |
| Domains | InterProScan | No recognized domains | - | Novel structural features possible |
| Transmembrane Regions | TMHMM | None predicted | 0.92 | Likely cytoplasmic protein |
| Signal Peptide | SignalP-5.0 | No signal peptide detected | 0.95 | Intracellular localization likely |
| Theoretical pI | ProtParam | 4.6 | - | Acidic protein, affects purification strategy |
| Hydrophobicity | Kyte-Doolittle | Average: -0.25 | - | Moderately hydrophilic |
This predictive structural analysis provides a foundation for experimental characterization and suggests Darcynin may function as a cytoplasmic protein with possible roles in protein-protein interactions rather than enzymatic activity.
This comparative analysis places Darcynin in the broader context of C. violaceum proteins:
| Protein | Size (aa) | Function | Regulation | Structural Features | Research Relevance |
|---|---|---|---|---|---|
| Darcynin (CV_4311) | 79 | Unknown | Unknown | Belongs to darcynin family | Potential antimicrobial or regulatory functions |
| Violacein | N/A (pigment) | Antimicrobial, virulence | QS-positive, VioS-negative | Tryptophan-derived pigment | Extensively studied antimicrobial compound |
| VioA-E enzymes | Various | Violacein biosynthesis | QS-positive, VioS-negative | Enzymatic domains | Biotechnological applications |
| CviI | 202 | AHL synthase | Autoregulation | LuxI family | Central to QS regulation |
| CviR | 250 | Transcriptional regulator | AHL-dependent | LuxR family | Controls virulence factors |
| VioS | 206 | Repressor | Unknown | Novel repressor | Negative regulator of violacein |
| Chitinases | ~450-600 | Chitin degradation | QS-positive | Glycoside hydrolases | Environmental adaptation |
This comparative framework helps researchers place Darcynin studies within the broader context of C. violaceum biology and suggests potential functional relationships to investigate.