KEGG: bja:blr7378
STRING: 224911.blr7378
Bradyrhizobium japonicum is a soil bacterium belonging to the α-Proteobacteria group. It has significant agricultural importance as it forms nitrogen-fixing symbiotic relationships with legumes, particularly soybeans. Research has demonstrated that B. japonicum strains, such as USDA6 and E109, exhibit distinct physiological characteristics including relatively fast growth rates compared to related species like B. diazoefficiens . Beyond nitrogen fixation, certain strains (e.g., FCBP-SB-406) have shown remarkable potential as biocontrol agents against soil-borne pathogens like Macrophomina phaseolina, reducing disease severity by up to 81.25% in controlled studies . This dual role in plant growth promotion and disease suppression makes B. japonicum a valuable subject for agricultural microbiology research.
Transcription elongation factor GreA belongs to a family of prokaryotic proteins that play crucial roles in transcriptional regulation. This factor facilitates RNA polymerase progression during transcription by helping resolve paused or arrested elongation complexes. In molecular terms, GreA promotes the hydrolytic cleavage of nascent RNA in backtracked transcription complexes, allowing transcription to resume efficiently. The protein contains two consensus prokaryotic transcription elongation factor signatures (Prosite PS00829 and PS00830) that are conserved across bacterial species . In rhizobia, GreA appears in genomic arrangements with LPS biosynthesis genes, suggesting potential co-regulation or functional relationships between transcription processes and cell envelope biosynthesis, which may be particularly relevant during host-microbe interactions.
While the specific organization in B. japonicum isn't directly detailed in the provided sources, comparative genomic analysis of related rhizobia provides insights. In Sinorhizobium meliloti, the greA gene is contiguous with and transcribed in the same direction as lpsB, which encodes an enzyme involved in lipopolysaccharide biosynthesis. This organization is similar to that observed in Rhizobium leguminosarum bv. viciae, where greA is adjacent to lpcC . The conservation of this genetic arrangement across multiple rhizobial species suggests functional significance, potentially indicating coordinated regulation of transcription elongation and cell envelope biosynthesis. The B. japonicum greA gene likely shares sequence homology with other rhizobial greA genes, particularly with R. leguminosarum (which shows 77% identity to S. meliloti greA) .
For recombinant production of B. japonicum GreA, Escherichia coli-based expression systems are typically most effective due to their high yield, ease of genetic manipulation, and well-established protocols. Based on research with similar proteins, the following approach is recommended:
Expression system selection:
pET vector systems (particularly pET28a with an N-terminal His-tag) offer tight regulation and high expression levels
BL21(DE3) or its derivatives are preferred host strains due to their deficiency in lon and ompT proteases
For difficult-to-express proteins, specialized strains like Rosetta(DE3) may overcome codon usage bias issues
Expression conditions:
Induction with 0.1-0.5 mM IPTG at OD₆₀₀ of 0.6-0.8
Post-induction growth at lower temperatures (16-25°C for 16-18 hours) often improves solubility
Supplementing growth media with glucose (0.5-1%) can reduce basal expression prior to induction
Alternative eukaryotic expression systems are generally unnecessary for bacterial transcription factors like GreA, as they typically fold correctly in prokaryotic hosts .
A multi-step purification strategy is recommended for obtaining high-purity, active recombinant GreA protein:
Initial capture:
Immobilized Metal Affinity Chromatography (IMAC) using Ni-NTA resin for His-tagged GreA
Lysis buffer: 50 mM Tris-HCl (pH 8.0), 300 mM NaCl, 10 mM imidazole, 10% glycerol, 1 mM DTT
Wash buffer: Same as lysis with 20-40 mM imidazole
Elution buffer: Same as lysis with 250-300 mM imidazole
Secondary purification:
2. Size Exclusion Chromatography (SEC) using Superdex 75 or similar
Buffer: 25 mM Tris-HCl (pH 7.5), 150 mM NaCl, 5% glycerol, 1 mM DTT
Optional polishing step:
3. Ion Exchange Chromatography (theoretical pI of GreA should be considered)
For basic proteins: Cation exchange (SP Sepharose)
For acidic proteins: Anion exchange (Q Sepharose)
Quality assessment:
SDS-PAGE: >95% purity
Western blot: His-tag detection and GreA-specific antibodies if available
Activity assay: Transcript cleavage assay using stalled transcription complexes
Final storage is optimal in 25 mM Tris-HCl (pH 7.5), 150 mM NaCl, 50% glycerol at -80°C, avoiding repeated freeze-thaw cycles .
Multiple complementary approaches should be employed to assess proper folding and activity:
Structural integrity assessment:
Circular Dichroism (CD) spectroscopy to evaluate secondary structure composition
Thermal shift assays to determine protein stability and proper folding
Limited proteolysis to assess compact, folded domains resistant to proteolytic cleavage
Dynamic Light Scattering (DLS) to evaluate homogeneity and absence of aggregation
Functional activity assays:
In vitro transcript cleavage assay using:
Purified RNA polymerase from B. japonicum or E. coli
Template DNA containing a promoter and sequence prone to pausing
Visualization of cleaved RNA products by gel electrophoresis
Binding assays to measure GreA-RNA polymerase interactions:
Surface Plasmon Resonance (SPR)
Fluorescence Anisotropy with labeled GreA or polymerase components
Complementation assays in GreA-deficient bacterial strains to assess functionality in vivo
The most definitive evidence of proper folding is demonstration of expected enzymatic activity, which for GreA is the ability to stimulate the transcript cleavage activity of RNA polymerase .
Comparative analysis of GreA across bacterial species reveals both conserved features and species-specific differences:
Conserved features:
Differences in Bradyrhizobium and other rhizobia:
Sequence alignment with R. leguminosarum shows approximately 77% identity, indicating significant conservation but with potential functional adaptations
Genomic context differs among species, with rhizobia commonly having greA positioned near LPS biosynthesis genes, suggesting potential specialized roles in symbiosis
Functional implications:
B. japonicum GreA may have evolved specific interaction parameters with its cognate RNA polymerase
The protein might have acquired specialized roles related to symbiotic association with host plants
Differential regulation of greA expression could reflect adaptation to the slower growth and metabolism characteristic of Bradyrhizobium species (generation times of 9.4-15.7 hours compared to minutes or few hours for E. coli)
A comprehensive understanding requires direct experimental comparison of purified GreA proteins from multiple species in standardized activity assays.
GreA likely plays several critical roles in the complex symbiotic relationship between B. japonicum and legume hosts:
Regulation of symbiosis-specific gene expression:
Transcription of nod, nif, and fix genes requires precise regulation during establishment of symbiosis
GreA could help RNA polymerase navigate through GC-rich sequences or complex secondary structures in symbiosis-related genes
Connection to LPS biosynthesis:
The genetic linkage between greA and LPS biosynthesis genes (as observed in related rhizobia) suggests coordinated regulation
LPS is crucial for establishing successful plant-microbe interactions and avoiding host defense responses
GreA may ensure proper expression of LPS synthesis genes during critical phases of nodule development
Stress adaptation during nodule formation:
The microaerobic, acidic environment of developing nodules imposes transcriptional challenges
GreA-mediated resolution of paused transcription complexes might be essential for adaptation to these conditions
Metabolic adaptation:
Transition from free-living to symbiotic lifestyle requires metabolic reprogramming
GreA could facilitate the required transcriptional shifts during this transition
Research examining differential expression of greA during various stages of symbiosis would provide valuable insights into its symbiotic relevance.
Several complementary approaches can effectively characterize GreA-RNA polymerase interactions:
In vitro biochemical methods:
Pull-down assays using His-tagged GreA to capture interacting RNAP subunits
Surface Plasmon Resonance (SPR) to determine binding kinetics and affinity constants
Hydrogen-deuterium exchange mass spectrometry (HDX-MS) to map interaction interfaces
Single-molecule FRET to observe real-time dynamics of GreA-RNAP interactions during transcription
Structural biology approaches:
Cryo-electron microscopy of GreA-RNAP complexes at different functional states
X-ray crystallography of co-crystals containing GreA and RNAP components
NMR spectroscopy for mapping interaction sites using chemical shift perturbations
In vivo approaches:
Bacterial two-hybrid systems adapted for B. japonicum
ChIP-seq to identify genomic regions where GreA associates with transcribing RNAP
RNA-seq comparing wild-type and greA mutant strains to identify genes most affected by GreA activity
A combination of these methods would provide comprehensive insights into both the physical nature of the interactions and their functional consequences in B. japonicum.
Nutrient availability significantly impacts B. japonicum metabolism and likely influences greA expression:
Response to varying nutrient conditions:
B. japonicum strains exhibit distinct growth responses to increasing yeast extract (YE) concentrations, with optimal growth at 1.5-2.0 g/L and growth inhibition at 5 g/L
These growth patterns suggest sophisticated transcriptional regulation mechanisms that may involve GreA
Potential regulatory mechanisms:
Nutrient-sensing systems likely modulate greA expression to adjust transcriptional elongation efficiency
Under nutrient limitation, increased GreA activity could help maintain expression of essential genes by resolving transcriptional pauses caused by low NTP concentrations
In nutrient-rich conditions, greA regulation might shift to optimize expression of metabolic pathways
Experimental approach to study nutrient effects:
qRT-PCR analysis of greA expression under varying YE concentrations (0.5-5.0 g/L)
Western blot quantification of GreA protein levels under different nutrient conditions
ChIP-seq analysis to map GreA occupancy across the genome under varying nutrient conditions
Transcriptome analysis of wild-type vs. greA mutant strains under different nutrient regimes
Research implications:
Understanding greA regulation in response to nutrients could provide insights into B. japonicum's adaptation to the changing nutrient environment during nodule development
This knowledge could inform strategies to optimize symbiotic performance in agricultural settings
Studying GreA's role in stress responses presents several methodological challenges:
Experimental design challenges:
Slow growth kinetics: With generation times of 9.4-18.8 hours , experiments require extended timeframes, complicating acute stress response studies
Genetic manipulation difficulties: Bradyrhizobium's large genome (~9 Mb) and slow growth make genetic modifications time-consuming
Multiple stress response pathways: Distinguishing GreA-specific effects from other stress response mechanisms requires careful controls
Environmental sensitivity: B. japonicum exhibits significant experimental variability , necessitating rigorous replication
Methodological considerations and solutions:
| Challenge | Recommended Approach | Limitations | Advantages |
|---|---|---|---|
| Creating greA mutants | CRISPR-Cas9 system optimized for Bradyrhizobium | Potential off-target effects | Faster than traditional homologous recombination |
| Measuring acute responses | Microfluidic systems with real-time microscopy | Technical complexity | Captures rapid responses despite slow growth |
| Distinguishing direct vs. indirect effects | GreA-ChIP-seq during stress conditions | Requires validated antibodies | Maps direct GreA involvement genome-wide |
| Experimental variability | Standardized media and growth conditions with ≥5 biological replicates | Resource intensive | Statistical robustness |
Stress conditions relevant to Bradyrhizobium ecology:
Acidic pH (simulating rhizosphere conditions)
Microaerobic environments (nodule-like conditions)
Oxidative stress (host defense response)
Osmotic stress (soil moisture fluctuations)
Thermal stress (soil temperature variations)
Advanced structural biology techniques can provide critical insights into GreA function at the molecular level:
Cryo-electron microscopy (Cryo-EM):
Enables visualization of GreA-RNA polymerase complexes in different functional states
Can capture conformational changes during transcript cleavage
Resolution now approaching 2-3Å allows visualization of catalytic residues and water molecules
Sample preparation challenges: Ensuring homogeneity of B. japonicum RNAP-GreA complexes
X-ray crystallography:
Provides atomic-resolution structures of GreA alone or in complex with RNAP fragments
Reveals precise details of interaction interfaces and catalytic residues
Crystallization challenges: Obtaining diffraction-quality crystals of flexible transcription complexes
Integrative structural biology approaches:
Combining lower-resolution Cryo-EM data with high-resolution crystal structures of components
Molecular dynamics simulations to model conformational changes during GreA function
Hydrogen-deuterium exchange mass spectrometry to map protein-protein interaction surfaces
Cross-linking mass spectrometry (XL-MS) to identify interacting regions
Structural insights likely to be revealed:
Conformational changes in RNA polymerase induced by GreA binding
Molecular basis for species-specific differences in GreA function
Structural consequences of mutations in conserved GreA motifs (PS00829 and PS00830)
Dynamic rearrangements during transcript cleavage and elongation restart
These structural studies would significantly advance understanding of transcription regulation in Bradyrhizobium and potentially reveal unique adaptations related to its symbiotic lifestyle.
Several factors can contribute to low activity of recombinant GreA in functional assays:
Protein-related factors:
Improper folding: The expression conditions may have resulted in misfolded protein
Solution: Try lower induction temperatures (16°C) and slower expression
Missing post-translational modifications: B. japonicum may modify GreA in ways not replicated in E. coli
Solution: Consider expression in related α-proteobacteria hosts
N- or C-terminal tags interfering with function:
Solution: Test constructs with removable tags or different tag positions
Assay-related factors:
Incompatibility with RNA polymerase source: B. japonicum GreA may have evolved specificity for its cognate RNAP
Solution: Use B. japonicum RNAP instead of E. coli RNAP in assays
Suboptimal buffer conditions:
Solution: Systematically optimize salt concentration, pH, and divalent cation concentrations
Template sequence incompatibility: The DNA template may lack appropriate pause sites
Solution: Use templates derived from B. japonicum genes with known regulatory pauses
Experimental validation approaches:
Circular dichroism to confirm secondary structure integrity
Size exclusion chromatography to verify monodispersity
Limited proteolysis to assess folding quality
Activity comparison with GreA from E. coli or other well-characterized species as positive controls
Systematic troubleshooting of these factors should help identify and address the source of low activity.
Creating stable greA mutants in B. japonicum presents challenges due to its slow growth and potential essentiality of the gene. The following strategies can help overcome these difficulties:
Genetic manipulation approaches:
Conditional knockout systems:
Tetracycline-inducible expression systems to control greA levels
Temperature-sensitive plasmids for controlled gene disruption
Partial activity mutants:
Point mutations in catalytic residues rather than complete gene deletion
Domain truncations that preserve some functionality
CRISPR-Cas9 based approaches:
Direct editing of the chromosome without antibiotic markers
Multiplex editing to create compensatory mutations if needed
Cultivation considerations:
Optimized recovery media:
Extended recovery periods:
Screening and verification protocols:
PCR-based screening optimized for high GC content
RT-qPCR to confirm reduced/eliminated greA expression
Phenotypic analyses focusing on growth rate and stress responses
Whole genome sequencing to confirm mutation and detect any compensatory mutations
For essential genes, creating depletion strains rather than complete knockouts may be necessary, allowing for controlled study of GreA function while maintaining viability.
Differentiating direct from indirect effects of GreA on gene expression requires a multi-layered experimental approach:
Integrated experimental strategy:
Direct binding and occupancy studies:
ChIP-seq to map genome-wide GreA binding sites
NET-seq (nascent elongating transcript sequencing) to identify transcriptional pause sites affected by GreA
GRO-seq (global run-on sequencing) to measure active transcription in WT vs. greA mutants
Temporal resolution experiments:
Time-course RNA-seq following GreA depletion in conditional mutants
Metabolic labeling of newly synthesized RNA to distinguish primary from secondary effects
Ribosome profiling to determine translation effects downstream of transcriptional changes
Perturbation analysis:
Testing effects of specific inhibitors of GreA activity
Creating point mutations in GreA that affect specific functions
Complementation studies with heterologous GreA proteins with known functional differences
Data integration framework:
| Data Type | Direct Effect Evidence | Indirect Effect Evidence |
|---|---|---|
| ChIP-seq | GreA binding at affected gene | No GreA binding detected |
| NET-seq | Changed pause site profile | Normal pause site distribution |
| Time-course | Immediate expression change | Delayed expression change |
| Point mutations | Specific mutation affects specific genes | Global expression changes |
Statistical approaches:
Principal component analysis to identify patterns in gene expression changes
Network analysis to identify regulatory cascades downstream of direct GreA targets
Bayesian modeling to infer causal relationships between transcriptional events
This integrated approach allows confident classification of gene expression changes as direct consequences of GreA activity versus downstream regulatory effects.
Several cutting-edge genomic approaches could significantly advance understanding of GreA function in Bradyrhizobium:
Comparative genomics approaches:
Pan-genome analysis across multiple Bradyrhizobium species to identify:
Conservation patterns in greA sequence and regulatory regions
Co-evolution with RNA polymerase components
Variation in genomic context of greA among species with different host ranges
Phylogenomic analysis to correlate GreA sequence variations with:
Host specificity
Geographical distribution
Symbiotic effectiveness
Functional genomics strategies:
Genome-wide CRISPR interference (CRISPRi) to identify genetic interactions with greA
Transposon sequencing (Tn-seq) under various stress conditions in wild-type vs. greA mutant backgrounds
RNA structurome analysis to identify RNA secondary structures affected by GreA activity
Global proteomics to assess post-transcriptional effects of GreA perturbation
Integrative multi-omics frameworks:
Correlation of transcriptomic, proteomic, and metabolomic data in response to greA manipulation
Network modeling to position GreA within the broader regulatory architecture of Bradyrhizobium
Machine learning approaches to identify subtle patterns in gene expression associated with GreA activity
These genomic approaches could reveal how GreA function has been tailored through evolution to support the unique ecological niche and symbiotic lifestyle of Bradyrhizobium species.
Understanding GreA function could lead to several applications for improving B. japonicum as a bioinoculant:
Enhanced stress tolerance engineering:
If GreA is confirmed to play a key role in stress response, overexpression could enhance survival of bioinoculants in challenging field conditions
Targeted modifications of GreA to optimize transcriptional response to specific agricultural stresses
Improved symbiotic performance:
Engineering GreA expression to enhance transcription of key symbiosis genes
Modifying GreA to optimize expression under the microaerobic conditions found in nodules
Creating GreA variants that improve coordination between nitrogen fixation and plant growth promotion traits
Biocontrol enhancement:
Given B. japonicum's demonstrated biocontrol potential (reducing disease severity by 81.25%) , optimizing GreA function could enhance expression of antimicrobial compounds
Coordinating expression of both plant growth-promoting and biocontrol traits through GreA-mediated transcriptional regulation
Field application considerations:
| Potential Improvement | Mechanism | Expected Benefit | Possible Risks |
|---|---|---|---|
| Stress-tolerant strains | GreA overexpression | Improved survival in field conditions | Metabolic burden, altered symbiotic properties |
| Enhanced nitrogen fixation | GreA variants optimized for nif gene expression | Increased crop yields | Energy diversion from other essential functions |
| Improved biocontrol | GreA-mediated upregulation of antimicrobial production | Reduced crop disease | Ecological impacts on soil microbiome |
Research combining laboratory optimization with field trials under diverse environmental conditions would be essential to realize these potential applications.
Several emerging technologies offer promising approaches to study the temporal dynamics of GreA function during symbiosis:
In situ visualization technologies:
Time-lapse microscopy with fluorescent reporters:
GreA-fluorescent protein fusions to track localization during nodule development
Dual-color imaging to simultaneously monitor GreA and RNA polymerase
Microfluidic devices to observe living bacteria during early infection events
Advanced tissue imaging techniques:
Expansion microscopy of nodule sections to visualize GreA distribution at subcellular resolution
Light sheet microscopy for 3D imaging of developing nodules with minimal photodamage
Super-resolution microscopy to resolve GreA-RNAP interactions within bacteroids
Temporal transcriptomics approaches:
Single-cell RNA-seq of bacteria at different stages of symbiosis
Spatial transcriptomics to map gene expression patterns across nodule zones
TIME-seq (transient induction measurement by RNA sequencing) to capture rapid transcriptional responses
SLAM-seq for metabolic labeling of newly synthesized RNA during symbiotic transitions
Biosensor technologies:
FRET-based sensors to detect GreA activity in real-time
Optogenetic control of GreA to precisely manipulate its activity during specific symbiotic stages
Nanobody-based probes for tracking GreA interactions in living cells
Integration with host plant systems:
Multi-organism transcriptomics to correlate GreA activity with host developmental transitions
Metabolic flux analysis to link transcriptional regulation to symbiotic metabolism
Signalome analysis to uncover how plant signals might modulate GreA function
These technologies would provide unprecedented insights into when, where, and how GreA functions during the complex developmental progression of Bradyrhizobium-legume symbiosis.