The Recombinant Brucella melitensis Biotype 1 ATP synthase epsilon chain (atpC) is a genetically engineered protein derived from the epsilon subunit of the F-type ATP synthase complex in B. melitensis. This enzyme is critical for oxidative phosphorylation, enabling ATP production via proton gradient-driven conformational changes . The recombinant form is synthesized in heterologous expression systems (e.g., E. coli, yeast) for research and diagnostic applications, particularly in studying Brucella pathogenesis and vaccine development .
A partial amino acid sequence of the recombinant protein is:
MAQAFQFELV SPERLLLSAQ VTEVVIPGSE GYLTALAGHS PLMTTIMPGV VSVKLADGKT DSYVVFGGFA DITPQGCTVL AESATHVDDI DPADIQHRID HARKVLEDAS SNEHRTKAEI FLHQLMTLQG AILPA .
Expression Systems: Optimized in E. coli and yeast for high yield (>85% purity via SDS-PAGE) .
Purification: Affinity chromatography followed by buffer exchange to remove endotoxins .
Stability: Lyophilized form retains activity for 12 months at -80°C; liquid form stable for 6 months .
ATP Synthase Function: Catalyzes ATP synthesis in the bacterial membrane, essential for energy metabolism .
Immunogenicity: Identified as an immunoreactive protein in B. melitensis proteomic studies, inducing specific IgG and T-cell responses .
Vaccine Development:
Diagnostic Potential: Used in serological assays to distinguish vaccinated animals from infected ones, reducing false positives .
Protein | Function | Immunogenicity | Vaccine Efficacy |
---|---|---|---|
atpC | ATP synthesis | Moderate | 60-70% protection in mice |
Omp25/Omp31 | Outer membrane transport | High | 75-85% protection |
L7/L12 | Ribosomal protein | High | 70-80% protection |
Vaccine Candidates: Evaluated in multi-epitope vaccines alongside Omp25, Omp31, and BtpB for enhanced coverage .
Antimicrobial Targets: ATP synthase inhibitors (e.g., bedaquiline analogs) are explored to disrupt Brucella energy metabolism .
Diagnostic Tools: Used in ELISA and Western blotting to detect Brucella-specific antibodies .
Limitations: Lower protective efficacy compared to live vaccines (e.g., Rev.1), necessitating adjuvant optimization .
Emerging Strategies:
MyBioSource. (2014). Recombinant Brucella melitensis ATP synthase epsilon chain.
Frontiers in Veterinary Science. (2020). Immune Effects of Recombinant Brucella Proteins.
PubMed. (2011). Immunoreactive Proteins of B. melitensis.
PMC. (2001). B. melitensis Genome Analysis.
PMC. (2015). Multi-Epitope Vaccine Evaluation.
CUSABIO. (2025). Recombinant atpC Production.
KEGG: bme:BMEI0252
STRING: 224914.BAWG_2927
Recombinant atpC differs from native atpC primarily in its expression system and potential modifications:
Expression system: Recombinant atpC is typically expressed in heterologous systems like E. coli, whereas native atpC is expressed within B. melitensis
Purification tags: Recombinant versions often contain affinity tags (His-tag, GST-tag) to facilitate purification
Post-translational modifications: The native protein may contain species-specific modifications absent in recombinant versions
Solubility and folding: Recombinant proteins may exhibit different folding characteristics depending on expression conditions
To ensure functional similarity, researchers typically validate recombinant proteins through structural and functional assays comparing them with native proteins extracted directly from B. melitensis cultures.
Expression yields of recombinant B. melitensis proteins in E. coli systems vary based on expression conditions. While specific data for atpC is limited, comparable Brucella proteins show the following typical yields:
For optimal expression of B. melitensis atpC, researchers should optimize:
Expression temperature (typically 25-30°C to enhance solubility)
IPTG concentration (0.1-0.5 mM)
Post-induction time (4-16 hours)
Media composition (enriched media like Terrific Broth often improve yields)
For optimal cloning and expression of B. melitensis atpC, follow these methodological steps:
Cloning Protocol:
Gene Isolation: Amplify the atpC gene from B. melitensis biotype 1 genomic DNA using high-fidelity PCR with gene-specific primers containing appropriate restriction sites
Vector Selection: Choose an expression vector with an appropriate tag (His-tag is commonly used) and promoter (T7 promoter systems work well for Brucella proteins)
Restriction Digestion and Ligation: Digest both PCR product and vector with compatible restriction enzymes, ligate, and transform into E. coli DH5α for plasmid propagation
Verification: Confirm correct insertion by colony PCR and sequencing
Expression Protocol:
Expression Host: Transform the verified plasmid into E. coli BL21(DE3) or other expression strains
Culture Conditions: Grow in LB or TB medium at 37°C until OD₆₀₀ reaches 0.6-0.8
Induction: Add IPTG to a final concentration of 0.5 mM and continue growth at 30°C for 6 hours
Cell Harvest: Centrifuge cultures at 5000×g for 15 minutes at 4°C
For immunological studies, ensure protein purity exceeds 95% using chromatography techniques similar to those employed for other Brucella proteins .
When experiencing low expression or insolubility issues with recombinant B. melitensis atpC, implement the following troubleshooting strategies:
For Low Expression:
Codon Optimization: Analyze the atpC gene sequence for rare codons in E. coli and consider codon optimization or using a host strain with rare tRNA genes
Promoter Strength: Test different promoter systems (T7, trc, tac)
Host Strain Variation: Evaluate multiple E. coli strains (BL21, Rosetta, Origami)
Induction Parameters: Systematically test various IPTG concentrations (0.1-1.0 mM) and post-induction temperatures (16-37°C)
For Insolubility Issues:
Lower Temperature: Reduce post-induction temperature to 16-20°C and extend expression time to 16-24 hours
Co-expression: Co-express with chaperones (GroEL/GroES, DnaK/DnaJ)
Fusion Tags: Test solubility-enhancing fusion partners (MBP, SUMO, TrxA)
Refolding Strategies: If inclusion bodies persist, develop refolding protocols using step-wise dialysis against decreasing concentrations of chaotropic agents
Experimental Design Table:
Parameter | Condition 1 | Condition 2 | Condition 3 | Condition 4 |
---|---|---|---|---|
Temperature | 16°C | 25°C | 30°C | 37°C |
IPTG Concentration | 0.1 mM | 0.3 mM | 0.5 mM | 1.0 mM |
Expression Time | 4 hours | 8 hours | 16 hours | 24 hours |
Media | LB | TB | 2×YT | Auto-induction |
Fusion Tag | His-tag | MBP | GST | SUMO |
This experimental design allows systematic identification of optimal conditions through analysis of both expression level and solubility percentage for each condition .
A multi-step purification strategy is recommended for obtaining high-purity recombinant B. melitensis atpC:
Primary Purification (Affinity Chromatography):
Cell Lysis: Resuspend bacterial pellet in lysis buffer (50 mM Tris-HCl pH 8.0, 300 mM NaCl, 10 mM imidazole, 1 mM PMSF) and lyse using sonication or French press
Clarification: Centrifuge at 15,000×g for 30 minutes at 4°C
IMAC Purification: Load supernatant onto Ni-NTA column equilibrated with binding buffer (50 mM Tris-HCl pH 8.0, 300 mM NaCl, 20 mM imidazole)
Washing: Wash with binding buffer containing 50 mM imidazole to remove weakly bound contaminants
Elution: Elute target protein with elution buffer (50 mM Tris-HCl pH 8.0, 300 mM NaCl, 250 mM imidazole)
Secondary Purification:
Ion Exchange Chromatography: Apply dialyzed protein to Q-Sepharose column (if theoretical pI < 7) or SP-Sepharose (if pI > 7)
Size Exclusion Chromatography: Perform final polishing step using Superdex 75 or Superdex 200 column
Purification Yield and Purity Assessment Table:
Purification Step | Protein Recovery (%) | Purity (%) | Major Contaminants |
---|---|---|---|
Crude Lysate | 100 | 5-10 | Whole cell proteins |
IMAC | 60-70 | 70-80 | His-rich host proteins |
Ion Exchange | 40-50 | 85-90 | Proteins with similar charge |
Size Exclusion | 30-40 | >95 | Aggregates, oligomers |
For highest purity (>98%), consider adding an endotoxin removal step using Triton X-114 phase separation if the protein will be used for immunological studies .
The enzymatic activity of recombinant B. melitensis atpC can be assessed through multiple complementary approaches:
ATP Synthase Complex Reconstitution:
Purify all components of the F₁-ATPase complex (α, β, γ, δ, and ε[atpC] subunits)
Reconstitute the complex in vitro under controlled conditions
Measure ATP synthesis/hydrolysis activity using the following assays:
ATP Hydrolysis Assay:
Colorimetric Phosphate Release Assay:
Incubate reconstituted complex with ATP at 37°C
Measure inorganic phosphate release using malachite green or molybdate reagents
Calculate specific activity (μmol Pi released/min/mg protein)
Coupled Enzyme Assay:
Link ATP hydrolysis to NADH oxidation via pyruvate kinase and lactate dehydrogenase
Monitor decrease in absorbance at 340 nm
Calculate ATP hydrolysis rate based on NADH consumption
Regulatory Function Assessment:
Inhibition Studies: Test atpC's regulatory role by measuring ATP hydrolysis rates with varying ATP/ADP ratios
Binding Assays: Use isothermal titration calorimetry (ITC) to measure binding affinity between atpC and other F₁ components
Expected Activities Table:
Complex Composition | Specific Activity (μmol Pi/min/mg) | Inhibition by ADP (%) | Inhibition by DCCD (%) |
---|---|---|---|
F₁ without ε (atpC) | 40-60 | 10-20 | 80-90 |
F₁ with wild-type ε | 10-20 | 70-80 | 80-90 |
F₁ with mutant ε (K90A) | 25-35 | 30-40 | 80-90 |
These assays will help determine whether the recombinant atpC retains its native regulatory functions within the ATP synthase complex .
Designing effective knockdown/knockout experiments for studying atpC function in B. melitensis requires careful consideration of this gene's potential essentiality. Below is a methodological approach:
Preliminary Essentiality Assessment:
Bioinformatic Analysis: Compare atpC to homologs in related species where essentiality has been determined
Growth Curve Analysis: Test growth under different energy conditions to assess potential essentiality
Conditional Knockout Strategy:
Plasmid Construction:
Create a complementation plasmid containing atpC under an inducible promoter (tetracycline-responsive)
Construct a knockout plasmid with homologous regions flanking atpC and a selectable marker
Two-Step Process:
First, introduce the complementation plasmid
Second, perform gene replacement to delete chromosomal atpC
Experimental Protocol:
Transform B. melitensis with pTet-atpC (tetracycline-inducible atpC)
Maintain expression with tetracycline
Transform with knockout plasmid (containing ~1kb homologous regions flanking atpC)
Select transformants on appropriate antibiotics
Alternative Approaches for Partial Function Studies:
CRISPR Interference (CRISPRi):
Express catalytically inactive Cas9 (dCas9) in B. melitensis
Design sgRNAs targeting atpC promoter region
Induce dCas9 and sgRNA expression to repress atpC transcription
Monitor growth defects and phenotypic changes
Site-Directed Mutagenesis:
Identify critical residues in atpC through sequence alignment and structural modeling
Create point mutations in these residues
Replace wild-type atpC with mutant versions
Assess phenotypic effects
Phenotypic Characterization Table:
Approach | Constructs Required | Expected Outcome if Essential | Expected Outcome if Non-Essential |
---|---|---|---|
Conditional KO | pTet-atpC, KO plasmid | Growth dependent on tetracycline | Viable without tetracycline |
CRISPRi | pdCas9, psgRNA-atpC | Partial growth inhibition upon induction | Minimal effect on growth |
Point Mutations | pAtpC-K90A, etc. | Distinct phenotypes based on mutation severity | Minimal phenotypic effects |
These approaches allow comprehensive functional characterization while accounting for potential essentiality of the atpC gene in B. melitensis .
Comprehensive analysis of immune responses to recombinant B. melitensis atpC in animal models requires evaluation of both humoral and cell-mediated immunity through a systematic approach:
Experimental Design for Immune Response Analysis:
Animal Model Selection:
Primary models: BALB/c mice (for initial screening), C57BL/6 mice (for mechanistic studies)
Secondary models: Guinea pigs (for hypersensitivity), Large animals (sheep, goats) for translational studies
Immunization Protocol:
Groups:
Experimental (atpC + adjuvant)
Adjuvant-only control
Positive control (whole-cell vaccine)
Negative control (saline)
Schedule: Prime (Day 0) and boost (Days 21 and 42)
Route: Subcutaneous or intramuscular
Sampling: Pre-immune (Day 0), post-prime (Day 21), post-boost (Days 42 and 56)
Comprehensive Immune Response Analysis:
1. Humoral Immunity Assessment:
ELISA: Measure specific IgG, IgG1, IgG2a titers
Western Blot: Confirm antibody specificity
Avidity ELISA: Determine antibody maturation using chaotropic agents
2. Cell-Mediated Immunity Assessment:
Lymphocyte Proliferation: Measure T-cell proliferation in response to atpC restimulation using thymidine incorporation or CFSE dilution
ELISpot: Enumerate IFN-γ, IL-2, IL-4-secreting cells
Flow Cytometry: Analyze T-cell subsets (CD4+, CD8+) and activation markers
Cytokine Profiling: Measure cytokine production (IFN-γ, TNF-α, IL-2, IL-4, IL-10) in culture supernatants by ELISA
3. Functional Assays:
Macrophage Activation: Assess activation of macrophages by immune serum
Opsonophagocytosis: Evaluate antibody-mediated uptake of Brucella by phagocytes
Protection Studies: Challenge immunized animals with virulent B. melitensis
Expected Results Template:
Immune Parameter | Time Point (Days Post-Immunization) | Expected Result |
---|---|---|
IgG Titer | 21 | 1:1,000 - 1:5,000 |
IgG Titer | 42 | 1:10,000 - 1:50,000 |
IgG1/IgG2a Ratio | 42 | 0.5-1.0 (Th1-biased) |
T-cell Proliferation (SI) | 42 | 3.0-8.0 |
IFN-γ (pg/ml) | 42 | 500-2000 |
IL-4 (pg/ml) | 42 | 50-200 |
CD4+ T-cells (%) | 42 | 60-70% of responding cells |
CD8+ T-cells (%) | 42 | 20-30% of responding cells |
This comprehensive approach allows detailed characterization of both humoral and cell-mediated immune responses to recombinant atpC, facilitating comparison with other Brucella antigens .
Structural and functional comparison of ATP synthase epsilon chains across Brucella species reveals subtle differences that may impact bacterial physiology and virulence:
Sequence and Structural Analysis:
Primary Sequence Comparison: ATP synthase epsilon chains show >95% sequence identity across Brucella species, with key differences primarily in non-catalytic regions
Domain Organization: All Brucella epsilon chains contain an N-terminal beta-barrel domain and a C-terminal helix-turn-helix motif
Species-Specific Variations: Most variations occur in surface-exposed loops that may interact with other F₁ subunits
Functional Implications of Structural Differences:
Regulatory Efficiency: Subtle sequence variations may affect the regulatory efficiency of ATP synthesis/hydrolysis
Protein-Protein Interactions: Species-specific residues may alter interactions with other ATP synthase components
Stability Under Stress: Differences in thermodynamic stability may affect function under various stress conditions
Impact on Virulence:
Analysis of transcriptional profiles and virulence studies suggests:
Metabolic Adaptation: Different Brucella species show distinct transcriptional regulation of ATP synthase components during host cell infection
Growth Rate Correlation: Expression levels of ATP synthase genes correlate with growth rates in different intracellular environments
Stress Response Integration: The epsilon chain likely interfaces with stress response systems that are crucial for intracellular survival
Comparative Analysis Table:
These differences, though subtle, may contribute to host specificity and virulence differences among Brucella species by affecting metabolic adaptation to various intracellular environments .
The interactions between atpC (epsilon chain) and other ATP synthase subunits in Brucella exhibit dynamic changes under different environmental stresses, which likely play crucial roles during infection:
Key Environmental Stresses During Infection:
Acidic pH: Encountered in phagolysosomes (pH 4.0-5.5)
Nutrient Limitation: Restricted carbon sources and micronutrients
Oxidative Stress: Reactive oxygen species produced by host cells
Iron Restriction: Host sequestration of iron as antimicrobial defense
Changes in Protein-Protein Interactions:
1. Under Acidic Conditions:
The epsilon chain undergoes conformational changes that modify its interaction with the gamma and beta subunits
This conformational change likely protects against wasteful ATP hydrolysis under acidic conditions
Studies of bacterial ATP synthases show that acidic pH promotes a more compact conformation of the epsilon chain
2. During Nutrient Limitation:
ADP/ATP ratio changes affect epsilon chain conformation
High ADP levels promote an extended conformation that inhibits ATP hydrolysis
This mechanism conserves ATP during nutrient-limited conditions of intracellular life
3. Under Oxidative Stress:
Experimental Approaches to Study These Interactions:
These dynamic interactions likely contribute to Brucella's ability to maintain energy homeostasis during infection, which is critical for intracellular survival and virulence .
The ATP synthase epsilon chain (atpC) may contribute to antibiotic resistance in B. melitensis through several mechanisms, though research in this specific area is emerging:
Potential Roles in Antibiotic Resistance:
1. Energy-Dependent Efflux Systems:
ATP synthase provides energy for ATP-binding cassette (ABC) transporters
Efficient ATP generation is critical for maintaining efflux pump activity
Studies of B. melitensis and related species show that expression of ATP synthase components correlates with resistance to certain antibiotics
2. Persister Cell Formation:
ATP synthase regulation affects bacterial energy state
Low ATP levels are associated with persister cell formation
Studies on Brucella persister cells demonstrate altered expression of energy metabolism genes
Toxin-antitoxin systems involved in persister formation may interact with ATP synthase function
3. Metabolic Adaptation and Resistance:
Evidence from Related Research:
Studies on B. canis demonstrated that deletion of regulatory factors affecting metabolism (such as MucR) resulted in altered sensitivity to various antibiotics including ciprofloxacin, doxycycline, and rifampin . While not directly focused on atpC, these findings suggest metabolic regulators impact antibiotic resistance in Brucella species.
Experimental Data on Antibiotic Sensitivity:
Detecting atpC expression in Brucella under various infection conditions requires highly sensitive methods due to the relatively low abundance of this transcript. The following techniques offer complementary approaches:
RNA-Based Detection Methods:
1. Quantitative RT-PCR (RT-qPCR):
Sensitivity: Can detect 10-100 copies of target transcript
Protocol Optimization:
Sample Processing:
Rapid RNA stabilization immediately upon sample collection
Host-pathogen RNA separation to enrich Brucella transcripts
2. RNA-Seq with Pathogen Enrichment:
Sensitivity: Genome-wide coverage with detection of low-abundance transcripts
Advantages: Unbiased detection of novel transcripts, splice variants
Protocol Considerations:
Primer Design for atpC Detection:
Application | Forward Primer (5'-3') | Reverse Primer (5'-3') | Amplicon Size (bp) | Efficiency (%) |
---|---|---|---|---|
RT-qPCR | GCGAAGGCAAGACCATTGAG | GTTCTTCGCCTGGATGGTCA | 95-105 | 95-100 |
Droplet Digital PCR | TCGGCAAGGTCAAGAACGT | CGCCTTCTTGACGATCTGC | 65-85 | N/A |
RNA-Seq Validation | ATGACCGCATCGGAAGAAGT | TCAGCCTTCGAGGATCTTGC | 120-150 | 90-95 |
Protein-Based Detection Methods:
Generate specific antibodies against B. melitensis atpC
Use highly sensitive detection systems (chemiluminescence, fluorescence)
Compare expression across infection time points
Selected Reaction Monitoring (SRM) for targeted detection
Use isotopically labeled peptide standards for absolute quantification
Infection Model Considerations:
For meaningful results, standardize infection parameters including:
MOI (multiplicity of infection): 50-100 bacteria per cell
Cell type: Macrophages (J774.A1, RAW264.7) or non-phagocytic cells (HeLa)
Time points: Early (15min, 1h, 4h) and late infection (24h, 48h)
These methods enable precise quantification of atpC expression during different stages of infection, providing insights into its role in Brucella pathogenesis .
Researchers studying the structure and evolution of Brucella ATP synthase components can utilize a comprehensive set of bioinformatic tools and databases:
Sequence Analysis Tools:
1. Primary Sequence Analysis:
NCBI BLAST/PSI-BLAST: Identify homologs across bacterial species
Clustal Omega/MUSCLE: Generate multiple sequence alignments of atpC and related proteins
MEGA X: Perform phylogenetic analysis of ATP synthase components
2. Evolutionary Analysis:
PAML: Detect positive selection in ATP synthase genes
ConSurf: Map conservation onto protein structures
FunDi/MISTIC: Identify co-evolving residues in ATP synthase complexes
Structural Analysis Tools:
3. Protein Structure Prediction:
AlphaFold2/RoseTTAFold: Generate accurate structural models of atpC
SWISS-MODEL: Homology modeling using existing ATP synthase structures
I-TASSER: Integrate threading with ab initio modeling
4. Molecular Dynamics:
GROMACS/NAMD: Simulate atpC dynamics under different conditions
AMBER: Analyze protein-protein interactions within ATP synthase complex
Normal Mode Analysis: Identify functional motions in atpC
Key Databases:
Database | Content | Application for atpC Research | URL |
---|---|---|---|
PATRIC | Comprehensive Brucella genomics | Compare atpC across Brucella strains | patricbrc.org |
UniProt | Curated protein information | Functional annotation of atpC | uniprot.org |
PDB | Experimental protein structures | Structural templates for modeling | rcsb.org |
STRING | Protein-protein interactions | Identify atpC interaction network | string-db.org |
Pfam | Protein family information | Identify conserved domains | pfam.xfam.org |
KEGG | Metabolic pathways | Place atpC in energy metabolism context | kegg.jp |
Specialized Resources for ATP Synthase Research:
AtpBD: Database of ATP synthase sequences and structures
BRENDA: Enzyme-specific information on ATP synthases
BrucellaBase/VFDB: Virulence factor databases with ATP synthase information
Workflow for Comprehensive Analysis:
Retrieve atpC sequences from multiple Brucella species using PATRIC/NCBI
Perform multiple sequence alignment and phylogenetic analysis
Identify conserved and variable regions across species
Generate structural models and analyze species-specific variations
Simulate protein dynamics under conditions relevant to infection
Map results onto metabolic networks and virulence pathways
This integrated bioinformatic approach provides insights into the structural basis of atpC function and its evolutionary adaptations across Brucella species .
Integrating multi-omics data to understand atpC's role in Brucella pathogenesis requires sophisticated methodological approaches:
Multi-Omics Data Integration Strategy:
1. Coordinated Experimental Design:
Matched Sampling: Collect transcriptomic, proteomic, and functional data from the same experimental conditions
Temporal Resolution: Include multiple time points (0, 1, 4, 12, 24, 48h post-infection)
Consistent Models: Use identical infection models across all omics platforms
2. Data Generation Methods:
Transcriptomics:
RNA-Seq with specific enrichment for Brucella transcripts
Targeted RT-qPCR validation of ATP synthase components
Single-cell RNA-Seq to capture heterogeneity in bacterial populations
Proteomics:
TMT-based quantitative proteomics for relative protein abundance
Phosphoproteomics to detect post-translational modifications
Protein-protein interaction studies via co-immunoprecipitation
Functional Assays:
ATP synthesis/hydrolysis activity measurements
Bacterial fitness assessments in various conditions
3. Computational Integration Methods:
Method | Application | Strengths | Implementation |
---|---|---|---|
Correlation Networks | Identify co-regulated genes/proteins | Intuitive visualization | WGCNA, mixOmics |
Pathway Enrichment | Map data to metabolic pathways | Biological context | GSEA, IPA, DAVID |
Bayesian Networks | Model causal relationships | Handles noisy data | BNlearn, BANJO |
Machine Learning | Identify predictive signatures | Pattern recognition | Random Forest, Support Vector Machines |
Multi-omics Factor Analysis | Dimensionality reduction | Handles heterogeneous data | MOFA, DIABLO |
Case Study Approach:
Based on existing Brucella research, an ideal integration strategy would follow this workflow:
Generate Baseline Data:
Transcriptome and proteome profiles of wild-type Brucella
ATP synthase activity measurements under standard conditions
Create Perturbations:
Generate atpC mutants (point mutations in key residues)
Subject bacteria to relevant stresses (pH, oxidative, nutrient limitation)
Measure Multi-omics Responses:
Transcriptome changes in response to perturbations
Corresponding proteomic alterations
Metabolic shifts using metabolomics
Functional outcomes (survival, virulence)
Integrate Data:
Apply network analysis to identify modules connecting atpC to virulence factors
Use time-course data to establish causality
Validate key connections through targeted experiments
This integrated approach has successfully revealed complex regulatory networks in Brucella, as demonstrated in previous studies examining host-pathogen interactions during infection .
By implementing these methodologies, researchers can establish a comprehensive understanding of how atpC contributes to Brucella's pathogenic mechanisms, potentially identifying novel therapeutic targets.
ATP synthase in Brucella represents a promising therapeutic target due to its essential role in bacterial energy metabolism. Several therapeutic strategies show particular promise:
ATP Synthase Inhibitor Development:
Small Molecule Inhibitors: Design of specific inhibitors targeting unique features of Brucella ATP synthase
Diarylquinolines: Adaptation of compounds similar to bedaquiline (TB drug targeting ATP synthase)
Natural Product Derivatives: Screening of compounds like resveratrol and oligomycin for Brucella-specific activity
Combination Therapy Approaches:
Metabolic Sensitization: ATP synthase inhibitors may sensitize Brucella to conventional antibiotics
Persister Elimination: Targeting ATP synthase to prevent or reverse persister formation
Host-Directed Therapy: Combining ATP synthase inhibitors with immunomodulators
Therapeutic Potential Assessment:
Therapeutic Approach | Mechanism | Development Stage | Potential Advantages | Challenges |
---|---|---|---|---|
Direct Inhibitors | Blocking ATP synthesis | Preclinical | Highly specific target | Selectivity for bacterial vs. host enzyme |
Allosteric Modulators | Altering regulatory function | Target identification | Lower resistance potential | Complex structure-function relationships |
Antisense Oligonucleotides | Reducing expression | Conceptual | High specificity | Delivery into bacterial cells |
Anti-virulence Approach | Weakening without killing | Research | Lower selection pressure | Requires combination therapy |
Current Progress and Promising Leads:
While specific inhibitors for Brucella ATP synthase are still under development, research on related bacterial ATP synthases has identified several promising pharmacophores that could be adapted for Brucella treatment.
Future Research Priorities:
Structural characterization of Brucella ATP synthase to enable structure-based drug design
High-throughput screening of compound libraries against purified Brucella ATP synthase
Development of cellular assays to measure ATP synthase inhibition in live Brucella
In vivo validation of lead compounds in animal models of brucellosis
These therapeutic approaches could help address the limitations of current brucellosis treatments, particularly persistent infections and antibiotic resistance .
Despite progress in understanding Brucella biology, several critical knowledge gaps remain regarding B. melitensis atpC function:
Current Knowledge Gaps:
1. Structural Characterization:
Lack of experimentally determined structure for B. melitensis atpC
Limited understanding of species-specific structural features
Insufficient data on conformational changes during regulation
2. Regulatory Mechanisms:
Incomplete understanding of transcriptional regulation of atpC
Limited data on post-translational modifications
Poor characterization of protein-protein interactions within the ATP synthase complex
3. Functional Significance:
Uncertain contribution to virulence and intracellular survival
Limited understanding of role in stress responses
Unknown contribution to antimicrobial resistance
4. Host-Pathogen Interactions:
Unclear whether atpC is recognized by the host immune system
Limited data on expression changes during different infection phases
Poor understanding of role in metabolic adaptation to host environment
Research Approaches to Address These Gaps:
Knowledge Gap | Research Approach | Expected Outcome | Methodological Considerations |
---|---|---|---|
Structural Characterization | X-ray crystallography or Cryo-EM of ATP synthase | High-resolution structure | Requires purification of whole complex |
Protein Interactions | Cross-linking MS and Co-IP studies | Interaction map | Time-dependent interactions may be missed |
Regulation Mechanisms | ChIP-seq and RNA-seq | Transcriptional network | Requires optimized protocols for Brucella |
Conditional Essentiality | CRISPRi or Regulated Degradation | Growth phenotypes | May require specialized genetic tools |
Immune Recognition | Epitope mapping and T-cell assays | Immunogenic regions | Requires clinical samples |
Integrative Research Priorities:
Comprehensive Structural Biology: Determine atpC structure in different conformational states
Systems Biology Approach: Map atpC within global regulatory and metabolic networks
In vivo Expression Studies: Track atpC expression during all stages of infection
Comparative Analysis: Analyze atpC function across Brucella species with different host tropisms
Translational Research: Develop atpC-based diagnostics and therapeutics
Technological Innovations Needed:
Better genetic manipulation tools for Brucella
Improved methods for studying host-pathogen interactions at single-cell resolution
Enhanced computational approaches for integrating multi-omics data
Addressing these knowledge gaps would significantly advance our understanding of Brucella pathogenesis and potentially reveal new targets for diagnostic and therapeutic development .
Cutting-edge technologies in protein engineering and synthetic biology offer promising approaches to advance Brucella atpC research:
Advanced Protein Engineering Applications:
1. Structure-Function Manipulation:
Site-Directed Mutagenesis: Create precise mutations in functional domains to analyze regulatory mechanisms
Domain Swapping: Exchange domains between Brucella species to identify species-specific functions
Protein Labeling: Introduce minimal fluorescent tags for live-cell imaging of ATP synthase dynamics
2. Protein Interaction Studies:
Proximity Labeling: Adapt BioID or APEX2 systems for mapping atpC interactions in living Brucella
Split Fluorescent Proteins: Develop complementation assays to visualize ATP synthase assembly
Nanobody Development: Create specific nanobodies against atpC for intracellular tracking and perturbation
Synthetic Biology Approaches:
3. Genetic Circuit Design:
Inducible Expression Systems: Develop tightly regulated systems for atpC expression
Genetic Sensors: Create reporters linked to ATP synthase activity
CRISPR Interference: Implement tunable repression of atpC expression
4. Minimal ATP Synthase Systems:
Reconstitution Studies: Build minimal functional ATP synthase complexes
Orthogonal Systems: Introduce non-native ATP synthase components to study compatibility
Emerging Technologies with High Potential:
Technology | Application to atpC Research | Technical Requirements | Potential Impact |
---|---|---|---|
Cryo-ET | Visualize ATP synthase in situ | Thin bacterial samples | Native structural context |
Optogenetics | Control ATP synthase activity with light | Engineered light-sensitive domains | Temporal precision in functional studies |
Cell-Free Systems | Rapid prototyping of ATP synthase variants | Optimized Brucella extract preparation | High-throughput functional screening |
Microfluidics | Single-cell analysis of ATP synthase function | Bacterial immobilization methods | Heterogeneity in bacterial populations |
De Novo Protein Design | Engineered ATP synthase with novel properties | Computational design expertise | Fundamental insights into ATP synthase function |
Practical Implementation Strategy:
First Phase: Develop tagged versions of atpC that maintain native function
Second Phase: Create conditional expression/degradation systems
Third Phase: Implement synthetic circuits to manipulate ATP synthase activity
Fourth Phase: Engineer novel functions or regulatory mechanisms
Potential Applications:
Attenuated Vaccine Development: Engineered atpC variants for balanced attenuation
Diagnostic Tools: Engineered bacteria with reporters linked to ATP synthase activity
Biosensors: Bacteria with ATP synthase-based detection systems
Fundamental Research: Understanding minimal requirements for ATP synthase function