Recombinant Pseudomonas syringae pv. tomato Integration host factor subunit alpha (ihfA)

Shipped with Ice Packs
In Stock

Product Specs

Form
Lyophilized powder
Note: While we prioritize shipping the format currently in stock, please specify your format preference during order placement for customized preparation.
Lead Time
Delivery times vary depending on the purchasing method and location. Please consult your local distributor for precise delivery estimates.
Note: Our proteins are shipped with standard blue ice packs. Dry ice shipping requires prior arrangement and incurs additional charges.
Notes
Avoid repeated freeze-thaw cycles. Store working aliquots at 4°C for up to one week.
Reconstitution
Centrifuge the vial briefly before opening to consolidate the contents. Reconstitute the protein in sterile deionized water to a concentration of 0.1-1.0 mg/mL. For long-term storage, we recommend adding 5-50% glycerol (final concentration) and aliquoting at -20°C/-80°C. Our standard glycerol concentration is 50% and can serve as a guideline.
Shelf Life
Shelf life depends on various factors including storage conditions, buffer composition, temperature, and protein stability. Generally, liquid formulations have a 6-month shelf life at -20°C/-80°C, while lyophilized forms have a 12-month shelf life at -20°C/-80°C.
Storage Condition
Store at -20°C/-80°C upon receipt. Aliquoting is crucial for multiple uses. Avoid repeated freeze-thaw cycles.
Tag Info
Tag type is determined during the manufacturing process.
The tag type is determined during production. If a specific tag type is required, please inform us, and we will prioritize its development.
Synonyms
ihfA; himA; PSPTO_2384; Integration host factor subunit alpha; IHF-alpha
Buffer Before Lyophilization
Tris/PBS-based buffer, 6% Trehalose.
Datasheet
Please contact us to get it.
Expression Region
1-100
Protein Length
full length protein
Purity
>85% (SDS-PAGE)
Species
Pseudomonas syringae pv. tomato (strain ATCC BAA-871 / DC3000)
Target Names
ihfA
Target Protein Sequence
MGALTKAEMA ERLYEELGLN KREAKELVEL FFEEIRHALE DNEQVKLSGF GNFDLRDKRQ RPGRNPKTGE EIPITARRVV TFRPGQKLKA RVEAYAGTKS
Uniprot No.

Target Background

Function
This protein constitutes one of the two subunits of integration host factor (IHF), a DNA-binding protein essential for genetic recombination and the regulation of transcription and translation.
Database Links
Protein Families
Bacterial histone-like protein family

Q&A

What is the integration host factor subunit alpha (ihfA) in Pseudomonas syringae pv. tomato?

Integration host factor subunit alpha (ihfA) is one of two subunits that form the heterodimeric DNA-binding protein known as integration host factor (IHF). In Pseudomonas syringae pv. tomato, ihfA works cooperatively with the beta subunit to bind and bend DNA, thereby regulating gene expression. The protein recognizes specific DNA sequences and induces sharp bends (often >160°) in the DNA molecule, facilitating the formation of nucleoprotein complexes that affect transcription, recombination, and other DNA transactions. IHF functions as a global regulator that influences multiple virulence pathways in Pst, including motility systems and secretion mechanisms that are critical for plant infection .

How is ihfA structurally organized in P. syringae compared to other bacterial species?

The ihfA subunit in P. syringae pv. tomato shares significant structural homology with ihfA proteins in other Pseudomonads and gram-negative bacteria. Typically, the protein contains DNA-binding motifs characterized by a core of three α-helices flanked by β-sheets that form an arm-like structure. These arms intercalate between DNA base pairs causing the sharp bend.

While the core structure remains conserved, comparative genomic analyses suggest that the promoter regions and regulatory elements of ihfA in P. syringae pv. tomato have evolved specific features that correlate with its plant pathogenic lifestyle. This contrasts with the ihfA regulation in non-pathogenic Pseudomonads or animal pathogens like P. aeruginosa. The structural conservation alongside regulatory divergence allows ihfA to participate in specialized virulence programs while maintaining its fundamental DNA-bending functions .

What physiological processes does ihfA influence in P. syringae pv. tomato?

The ihfA subunit influences numerous physiological processes in P. syringae pv. tomato:

  • Virulence gene regulation: IHF including the ihfA subunit regulates multiple virulence pathways by controlling the expression of key genetic loci associated with pathogenicity .

  • Motility control: IHF likely influences flagellar gene expression hierarchies similar to those observed in related Pseudomonads, where a four-tiered transcriptional regulation system controls flagellar production .

  • Chemotaxis system modulation: ihfA may play a role in regulating chemoreceptors (like PsPto-PscC) that are essential for sensing plant signals such as GABA and L-Pro, which are critical for bacterial entry into the plant apoplast .

  • Type III secretion system (T3SS) regulation: Given that IHF impacts virulence, it likely influences the expression of the T3SS which is essential for delivering effector proteins into plant cells to suppress immunity .

  • Population heterogeneity: IHF may contribute to the phenotypic heterogeneity observed in P. syringae populations during plant colonization, potentially modulating the balance between motile and T3SS-expressing subpopulations .

What are the most effective methods for cloning and expressing recombinant ihfA from P. syringae pv. tomato?

For effective cloning and expression of recombinant ihfA from P. syringae pv. tomato, researchers should consider the following methodology:

Cloning Strategy:

  • Gene amplification: Use PCR with high-fidelity polymerase and primers containing appropriate restriction sites flanking the ihfA coding sequence.

  • Vector selection: For structural studies, pET-series vectors work well for robust expression, while pBAD vectors offer more controlled expression for functional studies.

  • Fusion tags: An N-terminal 6×His-tag facilitates purification while minimizing interference with ihfA function. TEV or PreScission protease cleavage sites should be included for tag removal.

Expression Conditions:

  • Expression host: E. coli BL21(DE3) or derivatives like Rosetta for rare codon optimization.

  • Induction parameters: Conduct expression at 18-20°C after induction with 0.1-0.5 mM IPTG for 16-18 hours to maximize soluble protein yield.

  • Culture medium: Use enriched media like Terrific Broth supplemented with appropriate antibiotics.

Purification Protocol:

  • Cell lysis in buffer containing 50 mM Tris-HCl pH 8.0, 300 mM NaCl, 10% glycerol, and 5 mM β-mercaptoethanol.

  • Initial purification via Ni-NTA affinity chromatography.

  • Tag cleavage followed by reverse Ni-NTA step.

  • Final polishing via size exclusion chromatography.

This approach typically yields 10-15 mg of >95% pure recombinant ihfA protein per liter of bacterial culture, suitable for downstream functional and structural studies .

How can researchers effectively study ihfA-DNA interactions in vitro?

To effectively study ihfA-DNA interactions in vitro, researchers can employ multiple complementary approaches:

Electrophoretic Mobility Shift Assay (EMSA):

  • Use fluorescently labeled DNA fragments (25-40 bp) containing putative IHF binding sites.

  • Incubate with purified recombinant ihfA and ihfB subunits (as IHF functions as a heterodimer).

  • Analyze complex formation on 6-8% non-denaturing polyacrylamide gels.

  • Calculate binding affinities through titration experiments with varying protein concentrations.

DNase I Footprinting:

  • End-label DNA fragments (100-250 bp) with 32P.

  • Incubate with purified IHF complex (ihfA+ihfB).

  • Perform limited DNase I digestion.

  • Analyze protected regions by denaturing gel electrophoresis alongside sequencing ladders.

DNA Bending Assays:

  • Use circular permutation assays with ihfA binding sites positioned at different locations within DNA fragments.

  • Analyze mobility shifts to determine bend angles induced by IHF binding.

  • Incorporate FRET-based approaches with dual-labeled DNA to measure bend angles in solution.

Isothermal Titration Calorimetry (ITC):

  • Titrate IHF into solution containing target DNA.

  • Measure heat changes to determine thermodynamic parameters (ΔH, ΔS, Kd).

  • Compare different DNA sequences to establish binding specificity profiles.

For these experiments, buffer conditions should typically include 20-40 mM Tris-HCl (pH 7.5-8.0), 50-100 mM NaCl, 1-5 mM MgCl2, and 0.1-1 mM DTT. The inclusion of competitor DNA (poly[dI-dC]) at 50-100 μg/ml helps reduce non-specific interactions .

What approaches are most suitable for generating and validating ihfA knockout mutants in P. syringae pv. tomato?

For generating and validating ihfA knockout mutants in P. syringae pv. tomato, researchers should employ a systematic approach:

Mutant Generation Strategies:

  • Allelic exchange mutagenesis:

    • Create a suicide vector containing ihfA flanking regions with an antibiotic resistance cassette.

    • Introduce via conjugation or electroporation.

    • Select for double crossover events using positive (antibiotic resistance) and negative (e.g., sacB-mediated sucrose sensitivity) selection.

  • CRISPR-Cas9 approach:

    • Design sgRNAs targeting ihfA.

    • Introduce Cas9+sgRNA on a broad-host-range plasmid along with a repair template.

    • Select transformants and screen for successful editing.

Validation Methods:

  • Genetic verification:

    • PCR verification of gene deletion using primers flanking the targeted region.

    • Whole-genome sequencing to ensure no off-target mutations.

    • RT-PCR to confirm absence of ihfA transcript.

  • Complementation analysis:

    • Reintroduce wild-type ihfA in trans on a stable plasmid (e.g., pBBR1MCS series).

    • Verify restoration of wild-type phenotypes.

  • Phenotypic characterization:

    • Assess growth kinetics in various media including minimal medium and apoplast-mimicking HIM medium using methods similar to those described for other P. syringae mutants .

    • Evaluate swimming motility using soft agar assays (0.3% agar).

    • Measure biofilm formation capacity.

    • Test for altered expression of known IHF-regulated genes via RT-qPCR.

  • Virulence assessment:

    • Use spray-inoculation and infiltration methods to assess plant colonization and symptom development .

    • Perform competitive index (CI) assays mixing the ihfA mutant with wild-type tagged strains .

    • Measure bacterial population dynamics in planta over time.

Special Considerations:
Since ihfA may be essential for viability, conditional mutants might be necessary, using inducible promoter systems (e.g., arabinose-inducible pBAD system adapted for Pseudomonas) to control ihfA expression levels .

How does ihfA contribute to the regulation of the type III secretion system in P. syringae?

The integration host factor subunit alpha (ihfA) plays a multifaceted role in regulating the type III secretion system (T3SS) in P. syringae pv. tomato, which is essential for delivering effector proteins into plant cells to suppress host immunity:

Direct Transcriptional Regulation:
IHF (including the ihfA subunit) likely binds to promoter regions of key T3SS regulatory genes, particularly upstream of the master regulator HrpL, which controls the expression of most T3SS genes. By inducing DNA bending, IHF facilitates the formation of transcriptional complexes that modulate hrpL expression. This regulation may be similar to the observed interactions between flagellar expression and T3SS expression, where a regulatory link between these systems has been demonstrated .

Integration with Environmental Sensing:
ihfA contributes to the integration of environmental signals with T3SS expression. In an apoplast-mimicking medium (HIM), P. syringae displays phenotypic heterogeneity in T3SS expression, with bistable ON and OFF states . IHF likely participates in this regulatory network by influencing how bacteria respond to plant-derived signals that modulate virulence gene expression.

Coordination with Other Virulence Systems:
The ihfA subunit helps coordinate T3SS expression with flagellar motility. Research has shown that mutants lacking HrpL (T3SS master regulator) display increased swimming motility, while mutants lacking HrpV (a repressor of hrpL expression) show reduced motility . This suggests a regulatory crosstalk between these systems, with IHF potentially serving as a global regulator that influences both pathways.

Impact on Population Heterogeneity:
ihfA likely contributes to the observed phenotypic heterogeneity in T3SS expression during plant colonization. This heterogeneity is advantageous as it allows bacterial subpopulations to specialize, with some cells expressing T3SS to suppress plant immunity while others maintain motility for spreading through plant tissues . IHF may influence the frequency of switching between these phenotypic states.

The regulatory influence of ihfA on T3SS appears to be critical for balancing the energetic costs associated with simultaneous expression of multiple virulence systems. The significant growth advantage observed for double mutants lacking both flagella and T3SS supports the notion that expression of both systems carries a substantial fitness cost , with IHF potentially helping to optimize resource allocation.

What role does ihfA play in chemotaxis and bacterial entry into the plant apoplast?

The integration host factor subunit alpha (ihfA) contributes significantly to chemotaxis and plant apoplast entry through several interconnected mechanisms:

Regulation of Chemoreceptor Expression:
ihfA likely regulates the expression of crucial chemoreceptors like PsPto-PscC, which mediates the perception of plant-derived compounds such as GABA and L-Pro. These amino acids are abundant in the tomato apoplast and serve as important chemical cues for bacterial orientation and entry . Through its DNA-bending activity, IHF may modulate promoter accessibility of genes encoding these chemoreceptors.

Coordination of Chemotaxis Gene Clusters:
P. syringae possesses three chemotaxis gene clusters (che1, che2, and che3), but only che2 appears dedicated to controlling swimming motility . ihfA likely influences the differential expression of these clusters, ensuring appropriate deployment of chemotaxis machinery. Research has demonstrated that chemotaxis-deficient strains show reduced virulence in spray-inoculation assays due to impaired ability to locate and move toward plant entry points .

Integration of Environmental Signals:
ihfA helps integrate multiple environmental inputs to fine-tune bacterial behavior during the infection process. For instance, GABA levels significantly increase in tomato plants upon pathogen infection, and P. syringae has evolved to use this as a cue to access the plant apoplast . IHF likely participates in the regulatory networks that process these environmental signals.

Flagellar Gene Expression:
Entry into the plant apoplast requires directed bacterial movement toward stomata, which depends on functional flagella. ihfA participates in the hierarchical regulation of flagellar genes, influencing the expression of the flagellar master regulator FleQ . Experimental evidence shows that the perception of plant signals through chemoreceptors drives the entry of P. syringae into the tomato apoplast, with mutation of specific chemoreceptors impairing entry and reducing virulence .

Temporal Regulation:
ihfA likely contributes to the temporal control of motility during infection. Evidence shows that after successful entry into plant tissues, P. syringae reduces flagellar expression to avoid activating plant immunity . This transition from a motile to a less motile state is critical for successful colonization and likely involves IHF-mediated regulation.

The significance of ihfA in these processes is underscored by the observation that spray-inoculation (which requires natural entry) versus direct infiltration (bypassing entry) produces different outcomes for various bacterial mutants affecting chemotaxis and motility genes , highlighting the importance of properly regulated entry mechanisms.

How does ihfA influence flagellar motility regulation in P. syringae?

The integration host factor subunit alpha (ihfA) exerts significant influence on flagellar motility regulation in P. syringae pv. tomato through several key mechanisms:

Hierarchical Transcriptional Control:
ihfA participates in the four-tiered hierarchical transcriptional regulation system that controls flagellar production in P. syringae. Similar to its close relative P. aeruginosa, P. syringae possesses between one and five polar flagella with a complex regulatory network . IHF likely influences the expression of the master regulator FleQ, which sits at the top of this hierarchy . By binding to specific DNA sequences and inducing bending, IHF can alter the accessibility of promoter regions to transcription factors involved in flagellar gene expression.

Coordination with Virulence Systems:
ihfA helps coordinate flagellar expression with other virulence determinants, particularly the type III secretion system (T3SS). Research has demonstrated a regulatory link between these systems: deletion of the T3SS master regulator HrpL increases swimming motility, while deletion of the T3SS repressor HrpV reduces motility . This suggests that IHF participates in a regulatory network that balances the expression of these costly systems.

Response to Environmental Cues:
ihfA mediates the response of motility systems to environmental signals. For example, novel motility regulators like PSPTO_1042 (chrR) function as part of an extracytoplasmic sensing system that enables P. syringae to alter motility when encountering oxidative stressors . IHF likely integrates these signals into the flagellar regulatory network.

Impact on Cyclic di-GMP Signaling:
ihfA may influence motility through effects on cyclic di-GMP metabolism. Genome-wide studies have identified proteins like PSPTO_0406 (dipA), a cyclic di-GMP degrading enzyme, as novel motility regulators . Since cyclic di-GMP is a key second messenger controlling the transition between motile and sessile lifestyles, IHF might affect motility by regulating the expression of such enzymes.

Population Heterogeneity Regulation:
ihfA contributes to the phenotypic heterogeneity observed in flagellar expression both in vitro and in planta. Flow cytometry and microscopy studies have demonstrated that flagellar expression undergoes phenotypic heterogeneity in apoplast-mimicking medium and within apoplastic microcolonies throughout plant colonization . This heterogeneity appears advantageous as it balances the energetic costs of flagellar production with the benefits of motility during different phases of infection.

The complex role of ihfA in motility regulation is evidenced by data showing significant growth penalties associated with flagellar expression, particularly when coupled with T3SS expression . By influencing multiple regulatory pathways, ihfA helps optimize resource allocation to maximize bacterial fitness during plant colonization.

How can transcriptomics and proteomics approaches be optimized to study the ihfA regulon in P. syringae?

To comprehensively characterize the ihfA regulon in P. syringae pv. tomato, researchers should implement optimized transcriptomic and proteomic approaches that capture the global regulatory impact of this DNA-binding protein:

Transcriptomic Analysis Optimization:

  • Experimental design for RNA-seq:

    • Conditional expression systems: Utilize tetracycline-inducible or arabinose-inducible promoters to control ihfA expression levels rather than simple knockout/overexpression comparisons.

    • Time-course sampling: Collect samples at multiple timepoints (15, 30, 60, 120 minutes) post-induction to capture immediate and secondary regulatory effects.

    • Environmental conditions: Perform RNA-seq under multiple conditions relevant to infection (minimal media, apoplast-mimicking HIM, oxidative stress, low iron) to identify condition-specific regulation .

  • Integrated binding site analysis:

    • ChIP-seq protocol optimization: Use epitope-tagged ihfA (HA or FLAG) expressed at near-native levels to minimize artifacts.

    • Peak calling algorithms: Implement specialized algorithms that account for the unique binding properties of architectural proteins that cause DNA bending.

    • Motif enrichment analysis: Identify IHF binding motifs using advanced algorithms like MEME-ChIP with parameters optimized for AT-rich binding sites.

  • Single-cell transcriptomics:

    • Apply bacterial single-cell RNA-seq to investigate population heterogeneity in ihfA-regulated genes, similar to the heterogeneity observed in flagellar and T3SS expression .

    • Optimize cell sorting protocols using the methods described for sorting P. syringae populations with different expression profiles .

Proteomic Analysis Optimization:

  • Sample preparation:

    • Fractionate cells to enrich for different cellular compartments (membrane, cytoplasm, periplasm).

    • Use mild crosslinking to preserve protein complexes involving ihfA.

    • Implement SILAC or TMT labeling for accurate quantification across conditions.

  • Advanced MS techniques:

    • Apply data-independent acquisition (DIA) for deeper proteome coverage.

    • Implement targeted proteomics (PRM/MRM) for key virulence factors to achieve higher sensitivity.

    • Use non-canonical amino acid labeling (BONCAT) to focus on newly synthesized proteins after ihfA induction.

  • Post-translational modification analysis:

    • Profile phosphorylation changes in signaling proteins (particularly two-component systems) affected by ihfA.

    • Analyze changes in protein acetylation, which may respond to metabolic shifts caused by ihfA-mediated regulation.

Integrated Data Analysis Pipeline:

  • Multi-omics integration:

    • Implement Bayesian network modeling to integrate transcriptomic, ChIP-seq, and proteomic data.

    • Use dynamic regulatory event miner (DREM) algorithms to model temporal changes.

    • Develop custom network analysis approaches that account for the hierarchical nature of regulatory networks involving architectural proteins like IHF.

  • Comparative analysis:

    • Contrast the ihfA regulon with regulons of other global regulators (GacA, RpoN, AlgR) to identify regulatory overlaps.

    • Compare the ihfA regulon across different Pseudomonas species to identify core and variable components.

This comprehensive approach will provide a high-resolution map of the ihfA regulon, revealing both direct binding targets and downstream regulatory cascades that contribute to P. syringae virulence and adaptation .

What are the current challenges in understanding the interplay between ihfA and other global regulators in P. syringae pathogenicity?

Understanding the interplay between ihfA and other global regulators in P. syringae pathogenicity presents several significant challenges that require innovative experimental approaches:

Regulatory Network Complexity:
One major challenge is deciphering the highly interconnected nature of regulatory networks. IHF interacts with numerous other global regulators including alternative sigma factors (particularly RpoN/σ54), two-component systems, and other nucleoid-associated proteins. These interactions create complex feedback loops and feed-forward circuits that are difficult to dissect. For example, the relationship between IHF and the FleQ master regulator of flagellar genes likely involves multiple regulatory inputs similar to the four-tiered hierarchical control observed in related Pseudomonads . Resolution of these networks requires sophisticated genetic approaches such as epistasis analysis with double and triple mutants, and temporal control systems to determine the sequence of regulatory events.

Condition-Specific Regulation:
ihfA-mediated regulation varies dramatically across different environmental conditions. During plant infection, P. syringae encounters diverse microenvironments with varying nutrient availability, oxidative stress levels, and plant defense compounds. The regulatory role of ihfA likely differs in each context, as evidenced by the observation that expression of some virulence factors displays phenotypic heterogeneity in planta . Capturing these condition-specific regulatory events requires development of techniques for in situ analysis of bacterial gene expression during infection, which remains technically challenging.

Functional Redundancy:
Functional overlap between IHF and other nucleoid-associated proteins (NAPs) like HU, Fis, and H-NS complicates the identification of ihfA-specific effects. These proteins share the ability to bend DNA and may compensate for each other's absence. Additionally, the existence of multiple chemotaxis clusters (che1, che2, and che3) with seemingly redundant functions creates situations where phenotypes may only emerge when multiple systems are compromised. Overcoming this challenge requires sophisticated approaches like synthetic lethality screens and the generation of mutants with gradually reduced levels of multiple regulators.

Temporal Dynamics:
ihfA regulation involves complex temporal dynamics, with cascading effects that propagate through the regulatory network over time. For instance, the transition from motile to sessile states during infection likely involves temporal shifts in ihfA activity. Current analytical approaches often provide static snapshots that fail to capture these dynamics. Development of time-resolved methods for tracking regulatory changes in real-time during infection is needed.

Population Heterogeneity:
Perhaps the most significant challenge is understanding how ihfA contributes to the observed phenotypic heterogeneity in bacterial populations. Flow cytometry and microscopy studies have demonstrated that flagellar and T3SS expression display heterogeneity both in vitro and in planta . This heterogeneity appears to enable bacterial specialization and division of labor during colonization, but the mechanisms by which ihfA influences the frequency and stability of different phenotypic states remain poorly understood. Single-cell approaches combined with lineage tracking will be essential to resolve these questions.

Integration of Stress Responses:
ihfA likely participates in integrating various stress responses with virulence regulation. For example, the identification of PSPTO_1042 (chrR) as part of an extracytoplasmic sensing system responding to oxidative stress highlights the connection between stress sensing and motility regulation . Understanding how ihfA coordinates these responses requires methods to simultaneously monitor multiple cellular pathways under dynamically changing conditions.

Addressing these challenges will require development of new experimental paradigms that combine single-cell analysis, temporal resolution, spatial tracking during infection, and computational modeling approaches .

How might structural studies of ihfA contribute to developing novel antimicrobial strategies against P. syringae?

Structural studies of integration host factor subunit alpha (ihfA) could significantly advance the development of novel antimicrobial strategies against P. syringae pv. tomato through several promising avenues:

Targeting Protein-DNA Interactions:
High-resolution crystal or cryo-EM structures of P. syringae ihfA-DNA complexes would reveal the precise contact points between the protein and DNA, particularly the residues responsible for sequence-specific recognition. These structures could identify unique binding pockets suitable for small molecule inhibitors. Computational approaches like structure-based virtual screening could then be used to identify compounds that disrupt these interactions. Since IHF regulates multiple virulence pathways , such inhibitors would simultaneously compromise various aspects of pathogenicity rather than targeting a single virulence factor.

Disrupting Subunit Dimerization:
Structural characterization of the interface between ihfA and ihfB subunits could reveal opportunities for developing peptide mimetics or small molecules that prevent heterodimer formation. Since IHF functions as a heterodimer, preventing its assembly would effectively neutralize its regulatory function. NMR studies of the dimerization interface, coupled with alanine-scanning mutagenesis to identify key residues, would provide the foundation for rational design of dimerization inhibitors.

Allosteric Modulation:
Detailed structural studies might reveal allosteric sites in ihfA that, when occupied by small molecules, could alter its DNA-binding properties. Such allosteric modulators could either prevent binding altogether or lock IHF into conformations that disrupt proper regulation of target genes. This approach could be particularly valuable because allosteric sites often show greater structural divergence than active sites, potentially allowing for greater specificity against P. syringae compared to beneficial microbes.

Identifying Species-Specific Features:
Comparative structural analysis of ihfA across multiple bacterial species could identify unique structural features in P. syringae ihfA that could be selectively targeted. This approach would minimize off-target effects on beneficial soil and plant microbiota. Molecular dynamics simulations comparing ihfA from different species under various conditions could reveal dynamic properties and transient pockets unique to P. syringae.

Structure-Based Rational Design of Dominant-Negative Variants:
Structural insights could guide the engineering of dominant-negative ihfA variants that, when delivered into bacteria (perhaps via phage vectors), would incorporate into IHF complexes but render them non-functional. This approach could leverage the bacterial type VI secretion system or bacteriophage delivery systems as vectors.

Targeting Regulatory Interfaces:
Structural studies of ihfA in complex with other regulatory proteins (like RNA polymerase, HrpL, or FleQ) could reveal protein-protein interfaces critical for coordinating virulence gene expression . These interfaces could be targeted by peptide inhibitors or small molecules to disrupt regulatory networks rather than single interactions.

Practical Implementation Strategies:
To translate structural insights into practical antimicrobial strategies, several approaches could be pursued:

  • Plant-produced inhibitors: Engineer crop plants to produce peptides or small RNAs that target ihfA expression or function.

  • Biocontrol agents: Develop modified beneficial bacteria that secrete ihfA inhibitors within the plant environment.

  • Chemical treatments: Design structural analogs of natural compounds that plants produce during defense responses that could specifically target ihfA function.

  • Combination approaches: Design inhibitors that simultaneously target ihfA and other regulators like FleQ to achieve synergistic disruption of virulence.

The feasibility of these approaches is supported by the observation that virulence and motility systems in P. syringae carry significant fitness costs , suggesting that forcing their dysregulation through interference with ihfA could create substantial metabolic burdens that compromise bacterial survival .

How could engineered ihfA variants be used as tools for studying bacterial gene regulation?

Engineered ihfA variants offer powerful opportunities to dissect and manipulate bacterial gene regulatory networks in P. syringae and other bacteria. These variants can serve as sophisticated tools for fundamental research and applied biotechnology:

Inducible DNA Architecture Modifiers:
Engineered ihfA variants with altered DNA binding specificities could be developed as inducible tools to reshape the bacterial nucleoid structure on demand. By creating chimeric proteins that fuse the DNA-bending domain of ihfA with alternative DNA-binding domains (e.g., from LacI or TetR), researchers could direct DNA bending to specific genomic loci. These systems could be placed under tight control of chemical inducers, allowing temporal manipulation of chromosome architecture. Such tools would enable investigation of how DNA topology influences transcription of virulence genes, particularly in regulatory regions where multiple global regulators compete for binding .

Fluorescent Biosensors:
Fusion of ihfA to split fluorescent proteins (like split-GFP) can generate biosensors that report on the formation of functional IHF heterodimers in vivo. Such tools would enable real-time visualization of IHF activity during infection processes, revealing when and where IHF-mediated regulation occurs. This approach could be extended to create FRET-based sensors that detect conformational changes in IHF upon DNA binding, providing insights into the dynamics of IHF-mediated regulation during host-pathogen interactions .

Synthetic Gene Circuits:
Precisely engineered ihfA variants with orthogonal binding specificities could serve as core components in synthetic gene circuits. These circuits could be designed to respond to specific environmental conditions relevant to plant infection, such as apoplastic pH or plant defense compounds. By incorporating ihfA binding sites into synthetic promoters controlling reporter genes or effector proteins, researchers could create customizable switches to track bacterial behavior during infection or to trigger specific bacterial responses .

Optogenetic Control Systems:
Light-responsive ihfA variants could be engineered by fusion with photoreceptor domains (e.g., LOV or PhyB domains). These constructs would allow precise spatiotemporal control of gene expression through light exposure, enabling experimental manipulation of gene regulatory networks with unprecedented resolution. Such tools would be particularly valuable for studying the rapid adaptive responses of P. syringae to changing conditions during infection .

Programmable Epigenetic Regulators:
Fusion of enzymatic domains (e.g., methyltransferases or acetylases) to DNA-binding fragments of ihfA could create targeted epigenetic modifiers. These engineered proteins could introduce specific modifications at selected genomic loci, allowing researchers to investigate how DNA and protein modifications influence gene expression patterns in bacterial pathogens. This approach could reveal new targets for antimicrobial development by identifying critical epigenetic switches in virulence pathways .

Single-Cell Analysis Tools:
Engineering ihfA-based transcriptional reporters could enable high-resolution tracking of regulatory events in single bacterial cells. By incorporating ihfA binding sites into promoters driving fluorescent protein expression, researchers could visualize the heterogeneity in gene regulation across bacterial populations. This approach would be particularly valuable for understanding the phenotypic heterogeneity observed in P. syringae populations during plant colonization, where subpopulations specialize in different functions .

Methodological Considerations for Implementation:
When designing these tools, researchers should consider:

  • Protein stability and folding in the target organism

  • Potential interference with native IHF functions

  • Dose-dependent effects that may require tight control of expression levels

  • Validation of specificity using methods like ChIP-seq or ATAC-seq

By developing these engineered ihfA variants, researchers can create a sophisticated toolkit for dissecting the complex regulatory networks that govern P. syringae pathogenicity and potentially extend these approaches to other bacterial systems .

What potential exists for exploiting ihfA-mediated regulation to develop biocontrol strategies for agricultural applications?

The integration host factor subunit alpha (ihfA) presents several promising avenues for developing novel biocontrol strategies against P. syringae pv. tomato in agricultural settings:

Engineered Competitive Inhibitors:
Recombinant proteins based on modified ihfA structures could be developed to competitively inhibit native IHF function. These proteins would retain DNA-binding capability but lack regulatory functionality, effectively disrupting virulence gene expression networks. Such engineered proteins could be applied as foliar sprays with appropriate formulations to enhance stability and cellular uptake. The advantage of this approach is that it would simultaneously disrupt multiple virulence pathways regulated by IHF, including chemotaxis, motility, and type III secretion systems .

Anti-virulence Compounds Targeting IHF Regulation:
Small molecules identified through structure-based screening that disrupt ihfA-DNA interactions could serve as anti-virulence agents. Unlike traditional antibiotics, these compounds would not kill bacteria but rather prevent them from expressing virulence traits. This approach reduces selection pressure for resistance development. Initial screening could focus on natural plant compounds that might have evolved to interfere with bacterial regulatory systems, as plants have co-evolved with pathogens like P. syringae for millions of years .

Probiotic Bacterial Consortia:
Beneficial bacteria engineered to secrete peptides that interfere with ihfA function could be applied as protective biofilms on plant surfaces. These bacteria would compete with P. syringae for space and nutrients while simultaneously suppressing its virulence through interference with IHF-mediated regulation. Potential chassis organisms include plant growth-promoting Pseudomonads that are already adapted to the phyllosphere environment but lack pathogenicity factors .

CRISPR-Cas Delivery Systems:
Phage-based delivery systems could be engineered to introduce CRISPR-Cas constructs targeting ihfA into P. syringae populations. This approach would specifically reduce ihfA expression in the pathogen without affecting beneficial microbiota. The specificity could be enhanced by designing guide RNAs that target sequences unique to P. syringae pv. tomato ihfA .

Plant-Expressed Inhibitory Molecules:
Transgenic crops could be developed to express RNA molecules (RNAi) or peptides that interfere with ihfA expression or function. These molecules would be present in the apoplast, directly targeting the pathogen at its site of infection. The advantage of this approach is that it provides continuous protection integrated into the plant's biology .

Exploiting Population Heterogeneity:
Biocontrol strategies could be designed to disrupt the phenotypic heterogeneity observed in P. syringae populations during infection. By interfering with ihfA-mediated regulation, these approaches could force all bacteria into a single phenotypic state, preventing the division of labor that enables successful colonization. For example, compounds that lock bacteria in a flagellar-expressing state would increase visibility to plant immune systems and energy expenditure .

Immunomodulatory Approaches:
Treatments that alter plant apoplastic conditions could be developed to modify ihfA-regulated behaviors. For example, altering the levels of GABA and L-Pro in the apoplast could disrupt the chemotactic responses that P. syringae relies on for locating stomata and entering the plant . This approach would work by manipulating the signals that the pathogen uses rather than directly targeting the pathogen itself.

Implementation Strategies:
For practical agricultural application, these approaches could be deployed through:

  • Seed treatments that establish protective microbial communities

  • Precision timing of applications based on weather conditions favoring infection

  • Integration with existing integrated pest management practices

  • Combination treatments targeting multiple aspects of bacterial pathogenicity

This multi-faceted approach targeting ihfA-mediated regulation offers sustainable alternatives to traditional chemical control methods, potentially reducing environmental impact while providing effective disease management for tomato bacterial speck and related diseases .

What computational approaches would be most effective for predicting ihfA binding sites and regulatory networks in P. syringae?

To effectively predict ihfA binding sites and regulatory networks in P. syringae pv. tomato, researchers should implement an integrated computational framework combining multiple approaches:

Advanced Binding Site Prediction Algorithms:

  • Position Weight Matrix (PWM) Models with Architectural Considerations:

    • Develop specialized PWMs that incorporate not only sequence preferences but also DNA shape features.

    • Implement nucleotide interdependency models that account for the cooperative binding effects observed with IHF.

    • Train models using both experimentally validated binding sites and high-throughput data sets.

  • Deep Learning Approaches:

    • Implement convolutional neural networks (CNNs) trained on sequences flanking experimentally determined ihfA binding sites.

    • Incorporate attention mechanisms to capture long-range dependencies in DNA sequence patterns.

    • Use transfer learning from well-characterized bacterial systems to improve prediction accuracy for P. syringae.

  • Biophysical Models:

    • Develop algorithms that predict DNA bendability and minor groove width, key features for IHF binding.

    • Implement energy-based models that calculate the thermodynamic favorability of IHF-DNA interactions at specific sites.

    • Incorporate molecular dynamics simulations to predict conformational changes in DNA upon IHF binding.

Network Inference and Integration:

  • Bayesian Network Reconstruction:

    • Implement dynamic Bayesian networks to capture temporal aspects of ihfA regulation during infection stages.

    • Incorporate prior knowledge of regulatory hierarchies (e.g., the four-tiered flagellar regulation system) .

    • Use structure learning algorithms that can handle cyclic dependencies common in bacterial regulatory networks.

  • Multi-omics Data Integration:

    • Develop computational pipelines that integrate RNA-seq, ChIP-seq, and proteomics data.

    • Implement matrix factorization methods to identify regulatory modules from heterogeneous data types.

    • Apply network alignment algorithms to compare ihfA regulatory networks across different Pseudomonas species.

  • Condition-Specific Network Modeling:

    • Develop context-specific regulatory network models for different environments (e.g., epiphytic vs. apoplastic).

    • Implement differential network analysis to identify condition-specific regulatory interactions.

    • Use hidden Markov models to predict state transitions in regulatory networks during infection progression.

Advanced Analytical Techniques:

  • Stochastic Modeling for Population Heterogeneity:

    • Implement stochastic differential equation models to capture the phenotypic heterogeneity observed in P. syringae populations .

    • Develop agent-based models that simulate individual bacterial cells within a population to predict emergent behaviors.

    • Use bifurcation analysis to identify parameters controlling bistable expression patterns.

  • Comparative Genomics Approaches:

    • Develop phylogenetic footprinting methods optimized for identifying conserved IHF binding sites across Pseudomonas species.

    • Implement network-based orthology detection to identify functionally equivalent regulatory circuits across species.

    • Use synteny analysis to identify conserved gene neighborhoods potentially co-regulated by ihfA.

  • Structural Bioinformatics Methods:

    • Implement molecular docking simulations to predict IHF-DNA complex structures specific to P. syringae.

    • Develop algorithms that predict allosteric communication pathways within the IHF protein.

    • Use normal mode analysis to identify dynamic properties of IHF that influence its regulatory function.

Implementation and Validation Strategy:

To maximize prediction accuracy, these computational approaches should be implemented within an iterative framework:

  • Initial predictions based on sequence and comparative genomics

  • Experimental validation of high-confidence predictions using methods like ChIP-seq

  • Model refinement using new experimental data

  • Prediction of second-tier regulatory targets

  • Functional validation through phenotypic analysis of mutants

This comprehensive computational strategy would provide unprecedented insights into the global regulatory impact of ihfA in P. syringae, facilitating the development of targeted interventions for agricultural applications .

Quick Inquiry

Personal Email Detected
Please use an institutional or corporate email address for inquiries. Personal email accounts ( such as Gmail, Yahoo, and Outlook) are not accepted. *
© Copyright 2025 TheBiotek. All Rights Reserved.