NAD-dependent lysine deacetylase and desuccinylase. This enzyme specifically removes acetyl and succinyl groups from target proteins, thereby modulating the activity of various proteins that are inactive in their acylated form.
KEGG: rba:RB8404
STRING: 243090.RB8404
CobB functions as a NAD⁺-dependent lysine deacetylase that plays critical roles in bacterial metabolism and cellular regulation. As demonstrated across multiple bacterial species, CobB removes acetyl groups from lysine residues of target proteins, thereby modifying their function, activity, or stability.
In Escherichia coli, CobB has been shown to control energy metabolism, chemotaxis, and DNA supercoiling . One well-established substrate is acetyl-CoA synthetase (Acs), which CobB activates through deacetylation of lysine-609 (K609) . This activation is essential for the synthesis of acetyl-CoA, which directly impacts cell growth and energy production .
While specific R. baltica CobB studies are more limited, the high conservation of this enzyme across prokaryotes suggests similar functional roles in metabolic regulation and cellular signaling pathways.
The structure of CobB contains distinct domains that each contribute to its deacetylase function. Based on research with E. coli CobB (which serves as a model for other bacterial CobB proteins), the enzyme contains a catalytic domain and an N-terminal regulatory region.
The N-terminal tail (residues 1-37) has been identified as essential for regulatory interactions, particularly for binding secondary messengers like c-di-GMP . Within this region, specific residues including R8, R17, and E21 have been identified as critical for c-di-GMP binding . Mutagenesis studies demonstrated that changing these residues to alanine (CobB R8A, CobB R17A, and CobB E21A) significantly reduced c-di-GMP binding affinity without affecting the intrinsic deacetylase activity of the enzyme .
The catalytic domain contains the NAD⁺-binding site that is essential for the deacetylation reaction. As a member of the sirtuin family, CobB utilizes NAD⁺ as a co-substrate in the deacetylation reaction, which distinguishes it from zinc-dependent histone deacetylases.
While the structure of R. baltica CobB has not been fully characterized in the available literature, the high conservation of CobB across bacterial species suggests similar structural features and mechanisms.
CobB deacetylases target numerous protein substrates across diverse cellular pathways. The most well-characterized substrate is acetyl-CoA synthetase (Acs), which is activated through deacetylation of a specific lysine residue (K609 in E. coli) . This activation is critical for cell growth, particularly in conditions where acetate is a primary carbon source.
Quantitative acetylome studies comparing wild-type and ΔcobB strains have revealed hundreds of potential CobB substrates. In enterohemorrhagic E. coli O157:H7, comparative acetylation proteomics identified:
Among the most dramatically affected substrates (>100-fold increase in acetylation in the ΔcobB strain) were:
| Protein | Site | Fold Increase in ΔcobB |
|---|---|---|
| Serine hydroxymethyltransferase | K251 | 998-fold |
| Enolase (ENO) | K195 | 331-fold |
| Glyceraldehyde-3-phosphate dehydrogenase A (GapA) | K138 | 300-fold |
| LEE chaperone CesA (T3SS) | K44 | 705-fold |
Other heavily acetylated proteins identified included RNA polymerase beta prime subunit (40 sites), enolase (20 sites), and ribosomal proteins at multiple sites . These findings suggest CobB plays a broad regulatory role in metabolism, biosynthesis, and transcription.
The interaction between c-di-GMP and CobB represents a significant regulatory mechanism. Research has established that c-di-GMP binds specifically to CobB with physiologically relevant affinity, and this binding inhibits CobB's deacetylase activity through the following mechanism:
The binding specificity is remarkable - while c-di-GMP binds CobB with a dissociation constant (Kd) of 4.7 μM and a binding stoichiometry of 0.95, other cyclic nucleotides like cGMP and c-di-AMP show no detectable binding to CobB in isothermal titration calorimetry (ITC) assays .
The c-di-GMP binding site has been mapped to the N-terminal tail of CobB (residues 1-37). Mutagenesis studies identified three critical residues for this interaction:
When these residues were mutated to alanine, the binding affinity for c-di-GMP decreased dramatically:
CobB R8A: Kd = 274.7 μM (58-fold reduction)
CobB R17A: Kd = 89.3 μM (19-fold reduction)
Importantly, these mutations did not affect the intrinsic deacetylase activity of CobB, confirming that c-di-GMP binding and catalytic activity involve different regions of the protein .
The functional consequence of c-di-GMP binding is inhibition of CobB's deacetylase activity. In vitro deacetylation assays with known CobB substrates showed reduced deacetylation in the presence of c-di-GMP . In vivo studies demonstrated that strains with elevated c-di-GMP levels (through DgcZ overexpression) exhibited increased protein acetylation and reduced growth on acetate medium, consistent with CobB inhibition .
CobB participates in sophisticated feedback regulatory circuits that coordinate metabolism and signaling in bacterial cells. One particularly well-characterized circuit involves the interplay between CobB and the c-di-GMP signaling pathway.
A negative feedback loop exists where:
CobB deacetylates and activates DgcZ (a diguanylate cyclase)
Activated DgcZ produces more c-di-GMP
Elevated c-di-GMP binds to CobB and inhibits its deacetylase activity
This inhibition prevents further activation of DgcZ, creating a homeostatic regulatory circuit
This feedback loop was demonstrated through several experimental approaches:
In vitro deacetylation assays showed that c-di-GMP inhibits CobB activity towards substrates like Acs and DgcZ
Strains overexpressing DgcZ showed elevated c-di-GMP levels and increased protein acetylation, consistent with CobB inhibition
Growth assays on acetate medium showed reduced growth in strains with elevated c-di-GMP, indicating impaired Acs activity due to CobB inhibition
The physiological significance of this feedback loop likely extends beyond c-di-GMP signaling. By linking protein acetylation status to second messenger levels, bacteria can coordinate multiple aspects of cellular physiology including metabolism, motility, and potentially virulence in response to environmental conditions.
In enterohemorrhagic E. coli, CobB has been implicated in virulence regulation through its deacetylation of the LEE chaperone CesA, potentially creating additional regulatory circuits in pathogenic strains .
Assessing CobB deacetylase activity in vitro requires specialized techniques that can detect the removal of acetyl groups from substrate proteins. Based on the research literature, several methodologies have proven effective:
Western Blotting with Anti-Acetyllysine Antibodies:
This approach allows for detection of acetylation changes in specific proteins. The method involves:
Incubating purified CobB with acetylated substrate proteins and NAD⁺
Separating proteins by SDS-PAGE and transferring to membranes
Probing with anti-acetyllysine antibodies
This method has been successfully used to monitor CobB activity toward substrates like Acs and to assess the impact of potential inhibitors like c-di-GMP.
Isothermal Titration Calorimetry (ITC):
While not directly measuring deacetylase activity, ITC provides valuable information about binding interactions that can affect activity. ITC has been used to:
Determine the binding affinity (Kd) between CobB and c-di-GMP (4.7 μM)
Compare binding affinities of wild-type and mutant CobB proteins
Assess binding specificity by testing different cyclic nucleotides
Activity Assays with Synthetic Acetylated Peptides:
Using synthetic peptides containing acetylated lysine residues derived from known CobB substrates allows for quantitative assessment of deacetylation rates. These assays can be coupled to spectrophotometric or fluorometric detection systems that monitor NAD⁺ consumption or product formation.
Mass Spectrometry-Based Approaches:
Mass spectrometry provides the most comprehensive analysis of acetylation changes. The approach typically involves:
In vitro deacetylation reactions with purified components
Protein digestion to generate peptides
Enrichment of acetylated peptides if necessary
LC-MS/MS analysis to identify and quantify acetylation sites
Comparison of acetylation levels in samples with and without active CobB
This approach was used to identify 2,128 acetylated proteins in E. coli O157:H7 and to quantify changes in acetylation status in the absence of CobB .
While the provided literature doesn't offer specific protocols for R. baltica CobB expression and purification, we can derive recommendations based on successful approaches with bacterial CobB proteins and general recombinant protein methodologies:
Expression System Selection:
E. coli BL21(DE3) or similar strains typically provide high expression levels for bacterial proteins
Cold-shock inducible systems may improve solubility if standard induction leads to inclusion body formation
For physiological studies, consider complementation of a cobB-knockout strain to verify functional activity
Vector Design Considerations:
Include an N-terminal or C-terminal affinity tag (His6, GST, or MBP) for purification
If studying c-di-GMP binding, avoid N-terminal tags that might interfere with the N-terminal tail (residues 1-37), which contains critical binding residues (R8, R17, E21)
Consider a cleavable tag system to remove affinity tags after purification
Expression Conditions:
Inducer concentration: Typically 0.1-1.0 mM IPTG for T7-based systems
Temperature: Lower temperatures (16-20°C) often improve solubility
Duration: Extended expression (overnight) at lower temperatures can increase yield of soluble protein
Purification Strategy:
Affinity chromatography (Ni-NTA for His-tagged proteins)
Ion exchange chromatography to remove nucleic acid contamination
Size exclusion chromatography for final polishing and buffer exchange
Buffer Optimization:
Include 10-20% glycerol to enhance stability
Add reducing agents (DTT or β-mercaptoethanol) to prevent oxidation of cysteine residues
Consider including zinc or other metal ions if required for structural integrity
For activity studies, ensure buffer compatibility with NAD⁺-dependent assays
Activity Verification:
Test deacetylase activity using known substrates like acetylated Acs
Confirm binding to c-di-GMP using ITC or other binding assays
Assess oligomeric state by size exclusion chromatography or native PAGE
These recommendations should provide a starting point for developing a specific protocol for R. baltica CobB expression and purification, which can then be optimized based on experimental results.
Identifying novel CobB substrates requires comprehensive approaches that can detect changes in protein acetylation. Based on successful studies in the field, several strategies are recommended:
Comparative Acetylome Analysis:
This approach has proven highly effective for identifying CobB substrates on a proteome-wide scale:
Generate cobB knockout and wild-type strains
Extract proteins and digest to peptides
Enrich acetylated peptides using anti-acetyllysine antibodies
Analyze by LC-MS/MS with quantitative methods (e.g., SILAC, TMT, label-free quantification)
Identify peptides with significantly increased acetylation in the cobB knockout strain
This method successfully identified 581 acetylated peptides with significantly increased abundance in a ΔcobB strain compared to wild-type, representing 426 potential CobB substrate proteins .
Targeted Candidate Approach:
For testing specific proteins of interest:
Express and purify candidate proteins from cobB knockout strains to ensure acetylation
Incubate with purified recombinant CobB and NAD⁺
Analyze acetylation status before and after CobB treatment using:
Western blotting with anti-acetyllysine antibodies
Mass spectrometry to identify specific deacetylated residues
Functional assays to determine the impact of deacetylation on protein activity
In Vitro Biotinylated Protein Microarray:
For high-throughput screening:
Generate protein microarrays from your organism of interest
Treat with specific acetyltransferases and acetyl-CoA to acetylate proteins
Incubate with CobB and NAD⁺
Detect changes in acetylation using fluorescently labeled anti-acetyllysine antibodies
Identify proteins showing reduced acetylation after CobB treatment
Biochemical Affinity Purification:
To identify physical interaction partners:
Generate tagged CobB (consider catalytically inactive mutants to stabilize interactions)
Perform pull-down experiments coupled with mass spectrometry
Validate interactions by reciprocal co-immunoprecipitation
Confirm substrate status by demonstrating CobB-dependent deacetylation
Bioinformatic Prediction:
Enhance experimental approaches with computational methods:
Analyze known CobB substrates to identify consensus sequence motifs around acetylated lysines
Predict potential substrates based on sequence similarity and structural accessibility
Prioritize candidates for experimental validation based on prediction scores
These complementary approaches can be combined to develop a comprehensive understanding of the CobB substrate landscape in R. baltica or other bacterial species of interest.
Investigating CobB's role in metabolic regulation requires a multi-faceted approach that combines genetic, biochemical, and systems-level analyses. The following methodological framework is recommended:
Genetic Manipulation Strategies:
Generate precise cobB deletion mutants using CRISPR-Cas9 or homologous recombination
Create complementation strains expressing wild-type cobB
Engineer point mutants affecting:
Develop inducible expression systems for temporal control of CobB levels
Metabolic Phenotyping:
Growth curve analysis in different carbon sources, particularly:
Measure key metabolites using:
Targeted LC-MS for acetyl-CoA, CoA, and central carbon metabolites
NMR metabolomics for broader metabolic profiles
Assess oxygen consumption and carbon dioxide production rates
Integration with c-di-GMP Signaling:
Manipulate c-di-GMP levels by:
Measure c-di-GMP levels using:
HPLC-MS/MS quantification
Fluorescent biosensor systems
Correlate c-di-GMP levels with:
CobB activity (measured by substrate acetylation)
Growth phenotypes
Metabolic parameters
Systems-Level Analysis:
Combine acetylome profiling with:
Transcriptomics to identify regulatory networks
Proteomics to assess protein abundance changes
Fluxomics to measure metabolic pathway activities
Develop computational models integrating:
CobB-mediated protein deacetylation
c-di-GMP signaling dynamics
Metabolic flux distributions
Specific Experimental Approaches:
For acetate metabolism studies:
For central carbon metabolism:
Analyze acetylation status of key enzymes like enolase and glyceraldehyde-3-phosphate dehydrogenase
Measure enzyme activities in cell-free extracts
Perform 13C metabolic flux analysis to quantify pathway activities
These approaches will provide comprehensive insights into how CobB-mediated deacetylation regulates metabolic pathways in R. baltica and related bacteria.
The interplay between CobB and c-di-GMP signaling represents a sophisticated regulatory network with bidirectional control mechanisms. Based on current research, this interaction can be characterized as follows:
Molecular Basis of Interaction:
c-di-GMP directly binds to CobB with a physiologically relevant affinity (Kd = 4.7 μM)
This binding occurs specifically at the N-terminal region of CobB, with residues R8, R17, and E21 playing critical roles
The binding is highly specific for c-di-GMP; other cyclic nucleotides like cGMP and c-di-AMP show no detectable binding
Regulatory Mechanisms:
c-di-GMP inhibits CobB activity:
In vitro deacetylation assays demonstrate that c-di-GMP inhibits CobB-mediated deacetylation of substrates like Acs
This inhibition is dose-dependent and occurs at physiologically relevant concentrations
Point mutations in the c-di-GMP binding site of CobB (R8A, R17A, E21A) render the enzyme resistant to c-di-GMP inhibition
CobB regulates c-di-GMP production:
DgcZ, a diguanylate cyclase responsible for c-di-GMP synthesis, is a substrate of CobB
Deacetylation of DgcZ by CobB enhances its activity, increasing c-di-GMP production
This creates a negative feedback loop: CobB activates DgcZ → DgcZ produces c-di-GMP → c-di-GMP inhibits CobB → reduced DgcZ activation
Physiological Consequences:
The interplay affects multiple cellular processes:
Metabolic Regulation:
Potential Impact on Biofilm Formation:
c-di-GMP is a key regulator of biofilm formation in many bacteria
The feedback loop between CobB and c-di-GMP may help modulate the transition between planktonic and biofilm lifestyles
Possible Virulence Regulation:
In pathogenic strains, CobB regulates virulence factors
The CobB/c-di-GMP circuit could potentially coordinate virulence with metabolic state
Experimental Evidence Supporting This Interplay:
This complex regulatory network allows bacteria to coordinate metabolic activities with environmental sensing and cellular signaling, providing an integrated response system for adaptation to changing conditions.
Studying CobB function presents several technical and conceptual challenges that researchers should anticipate and address:
Challenge 1: Distinguishing Direct from Indirect Effects
CobB affects numerous proteins, making it difficult to determine which phenotypic changes result from direct deacetylation versus downstream effects.
Solutions:
Use catalytically inactive CobB mutants as controls to separate binding from enzymatic effects
Employ site-specific acetylation mimics (K→Q mutations) or non-deacetylatable variants (K→R mutations) in target proteins
Conduct in vitro deacetylation assays with purified components to confirm direct effects
Use time-course experiments to identify primary versus secondary effects
Challenge 2: Low Abundance of Acetylated Proteins
Many acetylated proteins exist at low stoichiometry, making detection challenging.
Solutions:
Enrich acetylated peptides using anti-acetyllysine antibodies before mass spectrometry
Use sensitive detection methods like selected reaction monitoring (SRM) or parallel reaction monitoring (PRM)
Consider chemical approaches to stabilize acetylation during sample preparation
Increase detection sensitivity by using ΔcobB strains where acetylation accumulates
Challenge 3: Preserving Acetylation Status During Sample Preparation
Acetylation can be lost during protein extraction and processing.
Solutions:
Include deacetylase inhibitors (e.g., nicotinamide) in lysis buffers
Add protease and deacetylase inhibitor cocktails immediately upon cell disruption
Use rapid sample preparation protocols to minimize processing time
Consider flash-freezing samples before processing
Challenge 4: Specificity of Anti-acetyllysine Antibodies
Antibodies may have varying affinities for different acetylated sequences.
Solutions:
Validate antibody specificity using synthetic acetylated peptides
Use multiple antibodies from different sources when possible
Complement antibody-based methods with mass spectrometry
Consider pan-specific and site-specific antibodies for comprehensive coverage
Challenge 5: Physiological Relevance of c-di-GMP Concentrations
Ensuring experiments use physiologically relevant c-di-GMP concentrations.
Solutions:
Measure endogenous c-di-GMP levels using HPLC-MS/MS
Use genetic approaches to modulate c-di-GMP levels within physiological ranges
Include appropriate controls (e.g., DgcZ G206A,G207A mutants) that maintain normal c-di-GMP levels
Perform dose-response experiments to determine concentration thresholds
Challenge 6: Functional Redundancy with Other Deacetylases
Some bacteria have multiple deacetylases with overlapping functions.
Solutions:
Generate single and multiple deacetylase knockouts to assess redundancy
Use phylogenetic and structural analysis to identify all potential deacetylases
Characterize substrate specificity profiles for each deacetylase
Implement conditional depletion strategies for essential deacetylases
By anticipating these challenges and implementing appropriate methodological solutions, researchers can generate more robust and physiologically relevant insights into CobB function in R. baltica and other bacterial systems.
Comparing CobB function across different bacterial species requires systematic approaches that account for evolutionary diversity while maintaining methodological consistency. The following framework enables meaningful cross-species comparisons:
Sequence and Structural Analysis:
Perform multiple sequence alignments of CobB homologs to identify:
Conserved catalytic residues
Species-specific insertions/deletions
Divergent regulatory domains
Construct phylogenetic trees to visualize evolutionary relationships
Apply homology modeling to predict structural differences, particularly in:
Recombinant Protein Expression and Characterization:
Express CobB from multiple species using identical expression systems and purification protocols
Perform side-by-side biochemical characterization:
Develop standardized deacetylation assays with identical substrate proteins or peptides
Cross-Species Complementation:
Generate cobB knockout strains in multiple bacterial species
Complement with cobB genes from diverse species under identical promoters
Assess functional complementation through:
Comparative Acetylome Analysis:
Apply identical acetylome profiling methods to wild-type and ΔcobB strains from different species
Compare:
Regulatory Network Mapping:
Characterize c-di-GMP/CobB interactions across species:
Compare expression regulation of cobB across species:
Promoter architecture
Transcriptional regulators
Growth phase-dependent expression
Based on available research, we can outline how this comparative approach might apply to E. coli and R. baltica CobB:
| Aspect | E. coli CobB | R. baltica CobB | Methodological Considerations |
|---|---|---|---|
| Key substrates | Acs, DgcZ | Likely conserved metabolic enzymes | Test deacetylation of Acs homologs from both species |
| c-di-GMP binding | Kd = 4.7 μM; requires R8, R17, E21 | Predicted based on sequence conservation | Express both proteins and compare binding affinities |
| Metabolic impact | Critical for acetate utilization | Likely important based on genome content | Compare growth on acetate in reciprocal complementation strains |
| Acetylome impact | Affects 426+ proteins | Not comprehensively characterized | Apply identical acetylome methods to both species |
By systematically comparing these and other aspects across species, researchers can gain insights into both the conserved core functions of CobB and the species-specific adaptations that have evolved to meet particular ecological niches.
Rigorous control experiments are essential to ensure the validity and reproducibility of findings related to CobB function. The following controls address key aspects of experimental design:
Controls for Genetic Studies:
Complementation Controls:
Catalytic Mutant Controls:
Express catalytically inactive CobB mutants to distinguish between enzyme activity and protein presence
These mutants should maintain normal protein folding and interactions
c-di-GMP Binding Mutant Controls:
Controls for c-di-GMP Experiments:
c-di-GMP Level Controls:
Cyclic Nucleotide Specificity Controls:
Controls for Biochemical Assays:
Deacetylation Assay Controls:
Binding Assay Controls:
Controls for Acetylome Studies:
Sample Preparation Controls:
Include deacetylase inhibitors in all samples
Process all samples identically and simultaneously
Include internal standards for normalization
Strain Controls:
Statistical Controls:
Multiple biological replicates (minimum three)
Technical replicates to assess method variation
Appropriate statistical tests with multiple testing correction
Validation Controls:
Orthogonal Method Controls:
Verify key findings using independent methodologies
For example, confirm mass spectrometry results with western blotting
Site-Specific Mutation Controls:
For key substrates, generate K→R (non-acetylatable) and K→Q (acetylation mimic) mutations
These distinguish effects of acetylation from other post-translational modifications
An experimental example from the literature demonstrates the implementation of these controls:
When investigating whether c-di-GMP inhibits CobB activity in vivo, researchers used multiple control strains:
Wild-type strain (baseline c-di-GMP)
dgcZ overexpression strain (elevated c-di-GMP)
ΔcobB strain (negative control)
ΔcobB::cobB strain (complementation control)
ΔcobB::cobB dgcZ+ strain (to test CobB inhibition by elevated c-di-GMP)
This comprehensive set of controls allowed researchers to establish that: